1
|
Chen M, Fu B, Zhou H, Wu Q. Therapeutic potential and mechanistic insights of astragaloside IV in the treatment of arrhythmia: a comprehensive review. Front Pharmacol 2025; 16:1528208. [PMID: 40276608 PMCID: PMC12018449 DOI: 10.3389/fphar.2025.1528208] [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: 11/14/2024] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
Arrhythmia, a common cardiovascular disorder, results from disturbances in cardiac impulse generation and conduction, leading to decreased cardiac output and myocardial oxygenation, with potentially life-threatening consequences. Despite advancements in therapeutic approaches, the incidence and mortality associated with arrhythmia remain high, and drug-related adverse effects continue to pose significant challenges. Traditional Chinese Medicine (TCM) has attracted considerable attention for its potential as a complementary and alternative approach in treating cardiovascular diseases, including arrhythmia. Astragalus, a prominent herb in TCM, is commonly used in clinical practice for its multi-faceted therapeutic properties, encompassing anti-arrhythmic, cardiotonic, anti-inflammatory, and immunomodulatory effects. Astragaloside IV, a primary active compound in Astragalus membranaceus, has demonstrated cardioprotective effects through mechanisms such as antioxidant, anti-inflammatory, and anti-apoptotic activities. Although evidence suggests that astragaloside IV holds promise in arrhythmia treatment, comprehensive reviews of its specific mechanisms and clinical applications in arrhythmia are scarce. This review systematically explores the pharmacological properties and underlying mechanisms of astragaloside IV in arrhythmia treatment. Utilizing a targeted search of databases including PubMed, Web of Science, Cochrane Library, Embase, CNKI, and Wanfang Data, we summarize recent findings and examine astragaloside IV's potential applications in arrhythmia prevention and treatment. Our analysis aims to provide a theoretical foundation for the development of novel arrhythmia treatment strategies, while offering insights into future research directions for clinical application.
Collapse
Affiliation(s)
- Meilian Chen
- Cardiac and Pulmonary Department, Quanzhou Hospital of Traditional Chinese Medicine, Fujian, China
| | - Binlan Fu
- Department of Internal Medicine, Chen Dai Central Health Center, Jinjiang, China
| | - Hao Zhou
- Department of Cardiology, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, China
| | - Qiaomin Wu
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
2
|
El-Shoura EAM, Hassanein EHM, Taha HH, Shalkami AGS, Hassanein MMH, Ali FEM, Bakr AG. Edaravone and obeticholic acid protect against cisplatin-induced heart toxicity by suppressing oxidative stress and inflammation and modulating Nrf2, TLR4/p38MAPK, and JAK1/STAT3/NF-κB signals. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5649-5662. [PMID: 38285279 PMCID: PMC11329704 DOI: 10.1007/s00210-024-02956-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/15/2024] [Indexed: 01/30/2024]
Abstract
Cardiotoxicity is a significant adverse effect of cisplatin (CIS) that necessitates extensive medical care. The current study examines the cardioprotective effects of edaravone (EDV), obeticholic acid (OCA), and their combinations on CIS-induced cardiac damage. Rats were allocated into five groups: the normal control group, the remaining four groups received CIS (7.5 mg/kg, i.p.) as a single dose on the fifth day and were assigned to CIS, OCA (10 mg/kg/day) + CIS, EDV (20 mg/kg/day) + CIS, and the (EDV + OCA) + CIS group. Compared to the CIS-treated group, co-treating rats with EDV, OCA, or their combinations significantly decreased ALP, AST, LDH, CK-MB, and troponin-I serum levels and alleviated histopathological heart abnormalities. Biochemically, EDV, OCA, and EDV plus OCA administration mitigated cardiac oxidative stress as indicated by a marked decrease in heart MDA content with a rise in cardiac antioxidants SOD and GSH associated with upregulating Nrf2, PPARγ, and SIRT1 expression. Besides, it dampened inflammation by decreasing cardiac levels of TNF-α, IL-1β, and IL-6, mediated by suppressing NF-κB, JAK1/STAT3, and TLR4/p38MAPK signal activation. Notably, rats co-administered with EDV plus OCA showed noticeable protection that exceeded that of EDV and OCA alone. In conclusion, our study provided that EDV, OCA, and their combinations effectively attenuated CIS-induced cardiac intoxication by activating Nrf2, PPARγ, and SIRT1 signals and downregulating NF-κB, JAK1/STAT3, and TLR4/p38MAPK signals.
Collapse
Affiliation(s)
- Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hesham H Taha
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Abdel-Gawad S Shalkami
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
- Clinical Pharmacy Program, Faculty of Health Science and Nursing, Al-Rayan Colleges, Medina, Kingdom of Saudi Arabia
| | | | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Adel G Bakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| |
Collapse
|
3
|
Fagone P, Mangano K, Basile MS, Munoz-Valle JF, Perciavalle V, Nicoletti F, Bendtzen K. Evaluation of Toll-like Receptor 4 (TLR4) Involvement in Human Atrial Fibrillation: A Computational Study. Genes (Basel) 2024; 15:634. [PMID: 38790263 PMCID: PMC11121426 DOI: 10.3390/genes15050634] [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: 03/27/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
In the present study, we have explored the involvement of Toll-like Receptor 4 (TLR4) in atrial fibrillation (AF), by using a meta-analysis of publicly available human transcriptomic data. The meta-analysis revealed 565 upregulated and 267 downregulated differentially expressed genes associated with AF. Pathway enrichment analysis highlighted a significant overrepresentation in immune-related pathways for the upregulated genes. A significant overlap between AF differentially expressed genes and TLR4-modulated genes was also identified, suggesting the potential role of TLR4 in AF-related transcriptional changes. Additionally, the analysis of other Toll-like receptors (TLRs) revealed a significant association with TLR2 and TLR3 in AF-related gene expression patterns. The examination of MYD88 and TICAM1, genes associated with TLR4 signalling pathways, indicated a significant yet nonspecific enrichment of AF differentially expressed genes. In summary, this study offers novel insights into the molecular aspects of AF, suggesting a pathophysiological role of TLR4 and other TLRs. By targeting these specific receptors, new treatments might be designed to better manage AF, offering hope for improved outcomes in affected patients.
Collapse
Affiliation(s)
- Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | | | - José Francisco Munoz-Valle
- Institute for Research in Biomedical Sciences, University Center for Health Sciences, University of Guadalajara, Guadalajara 44100, Jalisco, Mexico
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Klaus Bendtzen
- Institute for Inflammation Research, Rigshospitalet University Hospital, 2100 Copenhagen, Denmark
| |
Collapse
|
4
|
He J, Huang L, Sun K, Li J, Han S, Gao X, Wang QQ, Yang S, Sun W, Gao H. Oleuropein alleviates myocardial ischemia-reperfusion injury by suppressing oxidative stress and excessive autophagy via TLR4/MAPK signaling pathway. Chin Med 2024; 19:59. [PMID: 38589925 PMCID: PMC11003011 DOI: 10.1186/s13020-024-00925-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Myocardial ischemia/reperfusion injury (MIRI) is an important complication of reperfusion therapy, and has a lack of effective prevention and treatment methods. Oleuropein (OP) is a natural strong antioxidant with many protective effects on cardiovascular diseases, but its protective effect on MIRI has not yet been studied in depth. METHODS Tert-Butyl hydroperoxide (tBHP) was used to establish an in vitro oxidative stress model. Cell viability was detected by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) and lactate dehydrogenase (LDH). Flow cytometry and fluorescence assays were performed for evaluating the ROS levels and mitochondrial membrane potential (MMP). Immunofluorescence analysis detected the NRF2 nuclear translocation and autophagy indicators. Further, Western blotting and quantitative real-time PCR were performed to evaluate the expression levels of proteins and mRNAs. Molecular docking, CETSA, and molecular interaction analysis explored the binding between OP and TLR4. The protective effects of OP in vivo were determined using a preclinical MIRI rat model. RESULTS OP protected against tBHP-treated injury, reduced ROS levels and reversed the damaged MMP. Mechanistically, OP activated NRF2-related antioxidant pathways, inhibited autophagy and attenuated the TLR4/MAPK signaling pathway in tBHP-treated H9C2 cells with a high binding affinity to TLR4 (KD = 37.5 µM). The TLR4 inhibitor TAK242 showed a similar effect as OP. In vivo, OP could alleviate cardiac ischemia/reperfusion injury and it ameliorated adverse cardiac remodeling. Consistent with in vitro studies, OP inhibited TLR4/MAPK and autophagy pathway and activated NRF2-dependent antioxidant pathways in vivo. CONCLUSION This study shows that OP binds to TLR4 to regulate oxidative stress and autophagy for protecting damaged cardiomyocytes, supporting that OP can be a potential therapeutic agent for MIRI.
Collapse
Affiliation(s)
- Jia He
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Liting Huang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Kaili Sun
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Jilang Li
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Shan Han
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Research Center for Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Xiang Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Qin-Qin Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Shilin Yang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China.
| | - Wen Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China.
| |
Collapse
|
5
|
Ibrahim HA, Abd El-Alim AEAF, El-Hafeez MA, Metwally MMM, Khamis T, Galal AAA. Baicalein prevents capecitabine-induced heart damage in female Wistar rats and enhances its anticancer potential in MCF-7 breast cancer cells. Life Sci 2023; 319:121523. [PMID: 36842762 DOI: 10.1016/j.lfs.2023.121523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
AIM We investigated the ability of baicalein (BAI) to enhance the anticancer potential of capecitabine (CAP) in the MCF-7 cell line and its protective effect on CAP-induced cardiotoxicity in female Wistar rats. METHODS AND KEY FINDINGS In vitro study involved evaluating the effect of BAI and/or CAP on cell viability, cell cycle progression, and BAX and Bcl2 gene expression in MCF-7 cells. Co-treatment of BAI with CAP significantly reduced the viability of MCF-7 cells, improved their cytotoxic effect, markedly elevated the percentage of the sub-G1 population, drastically reduced the G2/M population, and significantly altered the mRNA expression of BAX and Bcl2 genes compared with each treatment alone. In vivo study revealed that the oral administration of CAP (140 mg/kg BW) to adult female rats significantly elevated the levels of serum creatine kinase-myocardial band (CK-MB), lactate dehydrogenase (LDH), tumor necrosis factor (TNF)-α, and interleukin (IL)-1β and cardiac TNF-α, IL-1β malondialdehyde (MDA) concentration, whereas it reduced the serum and cardiac total antioxidant capacity (TAC), level of cardiac glutathione (GSH) and activity of glutathione peroxidase (GPx) with a vast array of circulatory, inflammatory, degenerative, and necrotic alterations in the cardiac tissue. Furthermore, CAP administration significantly upregulated the mRNA expression of NF-κB, TLR4, MyD88, ATF6, CHOP, and JNK genes. Concurrent administration of BAI (200 mg/kg BW) and CAP significantly improved the biochemical alterations and cardiac oxidant/antioxidant status and architecture. In addition, it modulated the TLR4/MyD88/NF-κB pathway and endoplasmic reticulum stress. SIGNIFICANCE Altogether, BAI can augment the anticancer potential of CAP and alleviate its cardiotoxic effects during cancer treatment.
Collapse
Affiliation(s)
- Hosny A Ibrahim
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | | | - Mai Abd El-Hafeez
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Mohamed M M Metwally
- Pathology department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Tarek Khamis
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Azza A A Galal
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| |
Collapse
|
6
|
Hassanein EHM, Bakr AG, El-Shoura EAM, Ahmed LK, Ali FEM. Acetovanillone augmented the cardioprotective effect of carvedilol against cadmium-induced heart injury via suppression of oxidative stress and inflammation signaling pathways. Sci Rep 2023; 13:5278. [PMID: 37002251 PMCID: PMC10066216 DOI: 10.1038/s41598-023-31231-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 03/08/2023] [Indexed: 04/03/2023] Open
Abstract
Cardiac toxicity is a public health issue that can be caused by both environmental and occupational exposures. The current study aimed to investigate the effectiveness of carvedilol (CV), Acetovanillone (ACET), and their combination for ameliorating cadmium (Cd)-induced oxidative stress, inflammation, and necroptosis. Rats were assigned to; the normal group, Cd group (2 mg/kg; i.p., single dose), and the other three groups received orally CV (10 mg/kg), ACET (25 mg/kg), and CV plus ACET, respectively and a single dose of Cd. Oral administration of CV, ACET, and their combination significantly dampens cardiac oxidative injury by increasing antioxidants GSH and SOD levels, while it decreases MDA and NADPH oxidase levels mediated by decreasing cardiac abundance of Nrf2, HO-1, and SIRT1 and downregulating KEAP-1 and FOXO-3 levels. Also, they significantly attenuated inflammatory response as indicated by reducing MPO and NOx as well as proinflammatory cytokines TNF-α and IL-6 mediated by downregulating TLR4, iNOS, and NF-κB proteins expression as well as IκB upregulation. Moreover, they potently counteracted cardiac necroptosis by downregulating RIPK1, RIPK3, MLKL, and caspase-8 proteins expression. Of note, the combination of CV and ACET have marked protection that exceeded each drug alone. Conclusively, CV ad ACET potently mitigated Cd-induced cardiac intoxication by regulating NADPH oxidase, KEAP-1/Nrf2/HO-1, SIRT1/FOXO-3, TLR4/NF-κB/iNOS, and RIPK1/RIPK3/MLKL signals.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Adel G Bakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Ehab A M El-Shoura
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Lamiaa Khalaf Ahmed
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Assiut, Egypt
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| |
Collapse
|
7
|
Luo CJ, Li T, Li HL, Zhou Y, Li L. Resveratrol pretreatment alleviates NLRP3 inflammasome-mediated cardiomyocyte pyroptosis by targeting TLR4/MyD88/NF-κB signaling cascade in coronary microembolization-induced myocardial damage. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:143-155. [PMID: 36815254 PMCID: PMC9968948 DOI: 10.4196/kjpp.2023.27.2.143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 02/24/2023]
Abstract
Percutaneous coronary intervention and acute coronary syndrome are both closely tied to the frequently occurring complication of coronary microembolization (CME). Resveratrol (RES) has been shown to have a substantial cardioprotective influence in a variety of cardiac diseases, though its function and potential mechanistic involvement in CME are still unclear. The forty Sprague-Dawley rats were divided into four groups randomly: CME, CME + RES (25 mg/kg), CME + RES (50 mg/kg), and sham (10 rats per group). The CME model was developed. Echocardiography, levels of myocardial injury markers in the serum, and histopathology of the myocardium were used to assess the function of the cardiac muscle. For the detection of the signaling of TLR4/MyD88/NF-κB along with the expression of pyroptosis-related molecules, ELISA, qRT-PCR, immunofluorescence, and Western blotting were used, among other techniques. The findings revealed that myocardial injury and pyroptosis occurred in the myocardium following CME, with a decreased function of cardiac, increased levels of serum myocardial injury markers, increased area of microinfarct, as well as a rise in the expression levels of pyroptosis-related molecules. In addition to this, pretreatment with resveratrol reduced the severity of myocardial injury after CME by improving cardiac dysfunction, decreasing serum myocardial injury markers, decreasing microinfarct area, and decreasing cardiomyocyte pyroptosis, primarily by blocking the signaling of TLR4/MyD88/NF-κB and also reducing the NLRP3 inflammasome activation. Resveratrol may be able to alleviate CME-induced myocardial pyroptosis and cardiac dysfunction by impeding the activation of NLRP3 inflammasome and the signaling pathway of TLR4/MyD88/NF-κB.
Collapse
Affiliation(s)
- Chang-Jun Luo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning 530021, China,Department of Cardiology, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou 545007, China
| | - Tao Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning 530021, China
| | - Hao-Liang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning 530021, China
| | - You Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning 530021, China
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning 530021, China,Correspondence Lang Li, E-mail:
| |
Collapse
|
8
|
Zhang J, Hu Y, Wang H, Hou J, Xiao W, Wen X, Wang T, Long P, Jiang H, Wang Z, Liu H, Chen X. Advances in research on the protective mechanisms of traditional Chinese medicine (TCM) in myocardial ischaemia-reperfusion injury. PHARMACEUTICAL BIOLOGY 2022; 60:931-948. [PMID: 35587352 PMCID: PMC9132412 DOI: 10.1080/13880209.2022.2063342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/31/2022] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Developing effective drugs to treat myocardial ischaemia-reperfusion (MI/R) injury is imperative. Traditional Chinese medicines (TCMs) have had considerable success in the treatment of cardiovascular diseases. Elucidating the mechanisms by which TCMs improve MI/R injury can supplement the literature on MI/R prevention and treatment. OBJECTIVE To summarise TCMs and their main protective mechanisms against MI/R injury reported over the past 40 years. METHODS Relevant literature published between 1980 and 2020 in Chinese and English was retrieved from the Web of Science, PubMed, SpringerLink, PubMed Central, Scopus, and Chinese National Knowledge Infrastructure (CNKI) databases. Search terms included 'medicinal plants', 'myocardial ischaemia reperfusion injury', 'Chinese medicine prescriptions', 'mechanisms', 'prevention', 'treatment' and 'protection'. For inclusion in the analysis, medicinal plants had to be searchable in the China Medical Information Platform and Plant Database. RESULTS We found 71 medicinal species (from 40 families) that have been used to prevent MI/R injury, of which Compositae species (8 species) and Leguminosae species (7 species) made up the majority. Most of the effects associated with these plants are described as antioxidant and anti-inflammatory. Furthermore, we summarised 18 kinds of Chinese compound prescriptions, including the compound Danshen tablet and Baoxin pill, which mainly reduce oxidative stress and regulate mitochondrial energy metabolism. DISCUSSION AND CONCLUSIONS We summarised TCMs that protect against MI/R injury and their pharmacological mechanisms. This in-depth explanation of the roles of TCMs in MI/R injury protection provides a theoretical basis for the research and development of TCM-based treatment drugs.
Collapse
Affiliation(s)
- Jiexin Zhang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yonghe Hu
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Han Wang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Jun Hou
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Wenjing Xiao
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xudong Wen
- Department of Gastroenterology, The First People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Tingting Wang
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Pan Long
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hezhong Jiang
- Faculty of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Zhanhao Wang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Huawei Liu
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Xin Chen
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Bai LQ, Wang BZ, Liu QW, Li WQ, Zhou H, Yang XY. Effects of penehyclidine hydrochloride on myocardial ischaemia-reperfusion injury in rats by inhibiting TLR4/MyD88/NF-κB pathway via miR-199a-3p. Growth Factors 2022; 40:186-199. [PMID: 35984706 DOI: 10.1080/08977194.2022.2109469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
This study was to probe the role of penehyclidine hydrochloride (PHC) mediating the impact of toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)/nuclear factor-kappa B (NF-κB) signalling pathway on myocardial ischaemia-reperfusion injury (MI/RI) in rats through miR-199a-3p. The rat MI/RI model was established through ligating left anterior descending (LAD) coronary artery. PHC was injected preoperatively into the model rats, and injected with miR-199a-3p lentiviral vector or TLR4 antagonist (TAK-242). Next, cardiac function of rats was examined by echocardiography, and rat serum indicators, oxidative stress levels and inflammatory factors were detected. HE staining was applied to detect pathological tissue structure, TUNEL staining to detect apoptosis rate, qRCR and western blot to detect miR-199a-3p and TLR4/MyD88/NF-κB expressions in rat myocardial tissues. Dual luciferase reporter experiment was conducted to confirm the relationship between miR-199a-3p and TLR4. In conclusion, PHC suppresses TLR4/MyD88/NF-κB signalling pathway through miR-199a-3p, thereby improving MI/RI in rats.
Collapse
Affiliation(s)
- Ling Qiang Bai
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| | - Bin Zhe Wang
- Department of The First Outpatients, The 940th Hospital of Joint Logistics Support force of Chinese People's Liberation Army, Lanzhou City, China
| | - Qi Wei Liu
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| | - Wen Qiang Li
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| | - Hang Zhou
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| | - Xiao Yan Yang
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| |
Collapse
|
10
|
Sirasanagandla SR, Al-Huseini I, Sakr H, Moqadass M, Das S, Juliana N, Abu IF. Natural Products in Mitigation of Bisphenol A Toxicity: Future Therapeutic Use. Molecules 2022; 27:molecules27175384. [PMID: 36080155 PMCID: PMC9457803 DOI: 10.3390/molecules27175384] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/13/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Bisphenol A (BPA) is a ubiquitous environmental toxin with deleterious endocrine-disrupting effects. It is widely used in producing epoxy resins, polycarbonate plastics, and polyvinyl chloride plastics. Human beings are regularly exposed to BPA through inhalation, ingestion, and topical absorption routes. The prevalence of BPA exposure has considerably increased over the past decades. Previous research studies have found a plethora of evidence of BPA’s harmful effects. Interestingly, even at a lower concentration, this industrial product was found to be harmful at cellular and tissue levels, affecting various body functions. A noble and possible treatment could be made plausible by using natural products (NPs). In this review, we highlight existing experimental evidence of NPs against BPA exposure-induced adverse effects, which involve the body’s reproductive, neurological, hepatic, renal, cardiovascular, and endocrine systems. The review also focuses on the targeted signaling pathways of NPs involved in BPA-induced toxicity. Although potential molecular mechanisms underlying BPA-induced toxicity have been investigated, there is currently no specific targeted treatment for BPA-induced toxicity. Hence, natural products could be considered for future therapeutic use against adverse and harmful effects of BPA exposure.
Collapse
Affiliation(s)
- Srinivasa Rao Sirasanagandla
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Isehaq Al-Huseini
- College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Hussein Sakr
- College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Marzie Moqadass
- College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Srijit Das
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
- Correspondence: or
| | - Norsham Juliana
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Nilai 71800, Malaysia
| | - Izuddin Fahmy Abu
- Institute of Medical Science Technology, Universiti Kuala Lumpur, Kuala Lumpur 50250, Malaysia
| |
Collapse
|
11
|
Zhuang C, Chen R, Zheng Z, Lu J, Hong C. Toll-Like Receptor 3 in Cardiovascular Diseases. Heart Lung Circ 2022; 31:e93-e109. [PMID: 35367134 DOI: 10.1016/j.hlc.2022.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
Toll-like receptor 3 (TLR3) is an important member of the innate immune response receptor toll-like receptors (TLRs) family, which plays a vital role in regulating immune response, promoting the maturation and differentiation of immune cells, and participating in the response of pro-inflammatory factors. TLR3 is activated by pathogen-associated molecular patterns and damage-associated molecular patterns, which support the pathophysiology of many diseases related to inflammation. An increasing number of studies have confirmed that TLR3, as a crucial medium of innate immunity, participates in the occurrence and development of cardiovascular diseases (CVDs) by regulating the transcription and translation of various cytokines, thus affecting the structure and physiological function of resident cells in the cardiovascular system, including vascular endothelial cells, vascular smooth muscle cells, cardiomyocytes, fibroblasts and macrophages. The dysfunction and structural damage of vascular endothelial cells and proliferation of vascular smooth muscle cells are the key factors in the occurrence of vascular diseases such as pulmonary arterial hypertension, atherosclerosis, myocardial hypertrophy, myocardial infarction, ischaemia/reperfusion injury, and heart failure. Meanwhile, cardiomyocytes, fibroblasts, and macrophages are involved in the development of CVDs. Therefore, the purpose of this review was to explore the latest research published on TLR3 in CVDs and discuss current understanding of potential mechanisms by which TLR3 contributes to CVDs. Even though TLR3 is a developing area, it has strong treatment potential as an immunomodulator and deserves further study for clinical translation.
Collapse
Affiliation(s)
- Chunying Zhuang
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; First Clinical School, Guangzhou Medical University, Guangzhou, China
| | - Riken Chen
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenzhen Zheng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Guangzhou, China
| | - Jianmin Lu
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cheng Hong
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Li M, Han B, Zhao H, Xu C, Xu D, Sieniawska E, Lin X, Kai G. Biological active ingredients of Astragali Radix and its mechanisms in treating cardiovascular and cerebrovascular diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153918. [PMID: 35104756 DOI: 10.1016/j.phymed.2021.153918] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/12/2021] [Accepted: 12/30/2021] [Indexed: 05/13/2023]
Abstract
BACKGROUND With the rising age of the global population, the incidence rate of cardiovascular and cerebrovascular diseases (CCVDs) is increasing, which causes serious public health burden. The efforts for new therapeutic approaches are still being sought since the treatment effects of existing therapies are not quite satisfactory. Chinese traditional medicine proved to be very efficient in the treatment of CCVDs. Well described and established in Chinese medicine, Astragali Radix, has been commonly administered in the prophylaxis and cure of CCVDs for thousands of years. PURPOSE This review summarized the action mode and mechanisms of Astragali Radix phytochemicals on CCVDs, hoping to provide valuable information for the future application, development and improvement of Astragali Radix as well as CCVDs treatment. METHODS A plenty of literature on biological active ingredients of Astragali Radix used for CCVDs treatment were retrieved from online electronic PubMed and Web of Science databases. RESULTS This review highlighted the effects of five main active components in Astragali Radix including astragaloside Ⅳ, cycloastragenol, astragalus polysaccharide, calycosin-7-O-β-d-glucoside, and calycosin on CCVDs. The mechanisms mainly involved anti-oxidative damage, anti-inflammatory, and antiapoptotic through signaling pathways such as PI3K/Akt, Nrf2/HO-1, and TLR4/NF-κB pathway. In addition, the majority active constituents in AR have no obvious toxic side effects. CONCLUSION The main active components of Astragali Radix, especially AS-IV, have been extensively summarized. It has been proved that Astragali Radix has obvious therapeutic effects on various CCVDs, including myocardial and cerebral ischemia, hypertension, atherosclerosis, cardiac hypertrophy, chronic heart failure. CAG possesses anti-ischemia activity without toxicity, indicating a worthy of further development. However, high-quality clinical and pharmacokinetic studies are required to validate the current studies.
Collapse
Affiliation(s)
- Man Li
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Bing Han
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Huan Zhao
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Chongyi Xu
- Wenling Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Taizhou, Zhejiang, 317500, China
| | - Daokun Xu
- Wenling Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Taizhou, Zhejiang, 317500, China
| | - Elwira Sieniawska
- Department of Natural Products Chemistry, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| | - Xianming Lin
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Guoyin Kai
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
13
|
Wang Q, Chen W, Yang X, Song Y, Sun X, Tao G, Wang H, Zhao N, Huang Y, Chai E, Tang F. Inhibition of miRNA-1-Mediated Inflammation and Autophagy by Astragaloside IV Improves Lipopolysaccharide-Induced Cardiac Dysfunction in Rats. J Inflamm Res 2022; 15:2617-2629. [PMID: 35494314 PMCID: PMC9045596 DOI: 10.2147/jir.s362368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/05/2022] [Indexed: 12/22/2022] Open
Abstract
Introduction Astragaloside IV (AS-IV) is one of the main active components isolated from the traditional Chinese medicinal herb, Astragalus membranaceus. The present study was designed to investigate whether the regulation of microRNA-1 (miR-1)-mediated inflammation and autophagy contributes to the protective effect of AS-IV against cardiac dysfunction in rats treated with lipopolysaccharides (LPS). Methods Animal model of cardiac dysfunction in rats or cellular model of injured H9c2 heart cell line was established by using LPS. Echocardiography, electron microscopy, enzyme-linked immunosorbent assay, immunofluorescence, quantitative RT-PCR, and Western blotting were used to determine the cardiac function and expression of inflammation- and autophagy-related proteins at both the mRNA and protein levels. Results LPS caused cardiac dysfunction in rats or injury in H9c2 cells and induced inflammation and autophagy. Compared with LPS treatment, AS-IV treatment attenuated cardiac dysfunction or cell injury, accompanied by inhibition of inflammation and autophagy. However, the miR-1 mimics partly abolished the effects of AS-IV. In addition, the effect of the miR-1 inhibitor was similar to that of AS-IV in the LPS model. Further analyses showed that AS-IV treatment decreased the mRNA expression of miR-1 in the heart tissue of rats and H9c2 cells treated with LPS. Conclusion These results suggest that AS-IV attenuated cardiac dysfunction caused by LPS by inhibiting miR-1-mediated inflammation and autophagy, thereby providing a novel mechanism for the protection against cardiac diseases.
Collapse
Affiliation(s)
- Qiuning Wang
- Department of Pharmacology, Jinzhou Medical University, Jinzhou, Liaoning Province, People’s Republic of China
| | - Weiying Chen
- Department of Drug Quality Analysis, Jiuquan Drug Inspection and Testing Center, Jiuquan, Gansu Province, People’s Republic of China
| | - Xuefeng Yang
- Department of Physiology, Jinzhou Medical University, Jinzhou, Liaoning Province, People’s Republic of China
| | - Ying Song
- Cardiovascular Laboratory, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, People’s Republic of China
| | - Xiaowei Sun
- Department of Neurosurgery, China Resources Liaojian Group, General Hospital of Fuxin Mining Group (10th Clinical College of China Medical University), Fuxin, Liaoning Province, People’s Republic of China
| | - Guizhou Tao
- Internal Medicine-Cardiovascular Department, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, People’s Republic of China
| | - Hong Wang
- Allergy and Clinical Immunology Center, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, People’s Republic of China
| | - Nan Zhao
- Allergy and Clinical Immunology Center, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, People’s Republic of China
| | - Yue Huang
- Department of Pharmacology, Jinzhou Medical University, Jinzhou, Liaoning Province, People’s Republic of China
| | - Erqing Chai
- Neurointerventional Department, Emergency General Hospital, Beijing, People’s Republic of China
- Erqing Chai, Neurointerventional Department, Emergency General Hospital, Beijing, People’s Republic of China, Email
| | - Futian Tang
- Department of Cardiovascular Disease and Key Laboratory of Digestive System Tumor of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu Province, People’s Republic of China
- Correspondence: Futian Tang, Department of Cardiovascular Disease and Key Laboratory of Digestive System Tumor of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu Province, People’s Republic of China, Email
| |
Collapse
|
14
|
Zhang W, Zhang L, Zhou H, Li C, Shao C, He Y, Yang J, Wan H. Astragaloside IV Alleviates Infarction Induced Cardiomyocyte Injury by Improving Mitochondrial Morphology and Function. Front Cardiovasc Med 2022; 9:810541. [PMID: 35265681 PMCID: PMC8899080 DOI: 10.3389/fcvm.2022.810541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
The protective effect of astragaloside IV (AS-IV) on myocardial injury after myocardial infarction has been reported. However, the underlying mechanism is still largely unknown. We established a myocardial infarction model in C57BL/6 mice and injected intraperitoneally with 10 mg/kg/d AS-IV for 4 weeks. The cardiac function, myocardial fibrosis, and angiogenesis were investigated by echocardiography, Masson's trichrome staining, and CD31 and smooth muscle actin staining, respectively. Cardiac mitochondrial morphology was visualized by transmission electron microscopy. Cardiac function, infarct size, vascular distribution, and mitochondrial morphology were significantly better in AS-IV-treated mice than in the myocardial infarction model mice. In vitro, a hypoxia-induced H9c2 cell model was established to observe cellular apoptosis and mitochondrial function. H9c2 cells transfected with silent information regulator 3 (Sirt3) targeting siRNA were assayed for Sirt3 expression and activity. Sirt3 silencing eliminated the beneficial effects of AS-IV and abrogated the inhibitory effect of AS-IV on mitochondrial division. These results suggest that AS-IV protects cardiomyocytes from hypoxic injury by maintaining mitochondrial homeostasis in a Sirt3-dependent manner.
Collapse
Affiliation(s)
- Wen Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ling Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huifen Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chang Li
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chongyu Shao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu He
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
- Yu He
| | - Jiehong Yang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Jiehong Yang
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Haitong Wan
| |
Collapse
|
15
|
Chen C, Yu LT, Cheng BR, Xu JL, Cai Y, Jin JL, Feng RL, Xie L, Qu XY, Li D, Liu J, Li Y, Cui XY, Lu JJ, Zhou K, Lin Q, Wan J. Promising Therapeutic Candidate for Myocardial Ischemia/Reperfusion Injury: What Are the Possible Mechanisms and Roles of Phytochemicals? Front Cardiovasc Med 2022; 8:792592. [PMID: 35252368 PMCID: PMC8893235 DOI: 10.3389/fcvm.2021.792592] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Percutaneous coronary intervention (PCI) is one of the most effective reperfusion strategies for acute myocardial infarction (AMI) despite myocardial ischemia/reperfusion (I/R) injury, causing one of the causes of most cardiomyocyte injuries and deaths. The pathological processes of myocardial I/R injury include apoptosis, autophagy, and irreversible cell death caused by calcium overload, oxidative stress, and inflammation. Eventually, myocardial I/R injury causes a spike of further cardiomyocyte injury that contributes to final infarct size (IS) and bound with hospitalization of heart failure as well as all-cause mortality within the following 12 months. Therefore, the addition of adjuvant intervention to improve myocardial salvage and cardiac function calls for further investigation. Phytochemicals are non-nutritive bioactive secondary compounds abundantly found in Chinese herbal medicine. Great effort has been put into phytochemicals because they are often in line with the expectations to improve myocardial I/R injury without compromising the clinical efficacy or to even produce synergy. We summarized the previous efforts, briefly outlined the mechanism of myocardial I/R injury, and focused on exploring the cardioprotective effects and potential mechanisms of all phytochemical types that have been investigated under myocardial I/R injury. Phytochemicals deserve to be utilized as promising therapeutic candidates for further development and research on combating myocardial I/R injury. Nevertheless, more studies are needed to provide a better understanding of the mechanism of myocardial I/R injury treatment using phytochemicals and possible side effects associated with this approach.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Lin-Tong Yu
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bai-Ru Cheng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jiang-Lin Xu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yun Cai
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jia-Lin Jin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Ru-Li Feng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Long Xie
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Yan Qu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Dong Li
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Jing Liu
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yan Li
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Yun Cui
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Jin-Jin Lu
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Kun Zhou
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Qian Lin
| | - Jie Wan
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
- Jie Wan
| |
Collapse
|
16
|
Yan X, Wu L, Gao M, Yang P, Yang J, Deng Y. Omentin inhibits the resistin‑induced hypertrophy of H9c2 cardiomyoblasts by inhibiting the TLR4/MyD88/NF‑κB signaling pathway. Exp Ther Med 2022; 23:292. [PMID: 35340867 DOI: 10.3892/etm.2022.11222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/24/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Xiaoliang Yan
- Department of Cardiothoracic Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Lin Wu
- Department of Cardiology, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, Zhejiang 317500, P.R. China
| | - Min Gao
- Department of Cardiovascular Surgery, The Affiliated Cardiovascular Hospital of Shanxi Medical University and Shanxi Cardiovascular Hospital (Institute), Taiyuan, Shanxi 030024, P.R. China
| | - Pengjie Yang
- Department of Cardiovascular Surgery, The Affiliated Cardiovascular Hospital of Shanxi Medical University and Shanxi Cardiovascular Hospital (Institute), Taiyuan, Shanxi 030024, P.R. China
| | - Jinjing Yang
- Department of Cardiology and Central Laboratory, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi 030024, P.R. China
| | - Yongzhi Deng
- Department of Cardiovascular Surgery, The Affiliated Cardiovascular Hospital of Shanxi Medical University and Shanxi Cardiovascular Hospital (Institute), Taiyuan, Shanxi 030024, P.R. China
| |
Collapse
|
17
|
Bian Z, Liu H, Xu F, Du Y. Ursolic acid protects against anoxic injury in cardiac microvascular endothelial cells by regulating intercellular adhesion molecule-1 and toll-like receptor 4/MyD88/NF-κB pathway. Hum Exp Toxicol 2022; 41:9603271221093626. [PMID: 35438581 DOI: 10.1177/09603271221093626] [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] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiac microvascular endothelial cells (CMECs) are rapidly damaged after myocardial ischemia or hypoxia. In this study, we intend to explore whether ursolic acid (UA) can protect CMECs against hypoxia/reoxygenation (H/R) injury and to detect related molecular mechanism. METHODS CMECs were subjected to H/R condition in the absence or presence of UA. Cell behaviors were measured by Cell Counting Kit-8, transwell, ELISA and western blot assays. siRNA was applied to reduce ICAM1 expression, then the effect of co-treatment of UA and si-ICAM1 on CMECs has been detected by biological experiments. RESULTS Under H/R stimulation, the proliferation and migration of CMECs were inhibited, as well as the inflammation and oxidative stress were enhanced. UA treatment obviously reversed these H/R-induced injuries and reduced ICAM1 expression. Moreover, knockdown of ICAM1 could alleviate the H/R-induced injuries and strengthen the protective effect of UA on CMECs under H/R condition. Additionally, the protein levels of TLR4, MyD88 and p-P65 NF-κB were obviously increased after H/R stimulation, whereas the addition of UA could alter the phenomena by reducing TLR4, MyD88, and p-P65 NF-κB expression. CONCLUSIONS Our results insinuated that UA could alleviate H/R-induced injuries in CMECs by regulating ICAM1 and TLR4/MyD88/NF-κB pathway.
Collapse
Affiliation(s)
- Zhongrui Bian
- Department of Cardiology, 531675the Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Hui Liu
- Department of Cardiology, 531675the Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Fei Xu
- Department of Cardiology, 531675the Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yimeng Du
- Department of Cardiology, 531675the Second Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
18
|
Wu T, He S, Jiao Z, Liang X, Chen Y, Liu H, Zhang Y, He G. Low Molecular Weight Heparin Improves the Inflammatory State of Acute Sinusitis Rats Through Inhibiting the TLR4-MyD88-NF-κB Signaling Pathway. Front Pharmacol 2021; 12:726630. [PMID: 34867331 PMCID: PMC8635784 DOI: 10.3389/fphar.2021.726630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/02/2021] [Indexed: 01/20/2023] Open
Abstract
Introduction: Low molecular weight heparin (LMWH), a natural sulfated glycosaminoglycan with an affinity for proangiogenic factors, is produced by chemical or enzymatic depolymerization of unfractionated heparin (UFH). Known for its anticoagulant effects, LMWH has recently been reported to have a strong anti-inflammatory effect on colitis, myocarditis, and airway inflammation. However, as a newly-developed drug, its anti-inflammatory mechanism in upper respiratory tract inflammation has not been well-studied. Methods: SD rats were randomly divided into control and experimental groups. The experimental group was established by building an acute nasal sinusitis model with expansion sponges mixed with Streptococcus pneumoniae. Then the experimental group rats were subcutaneously injected with different concentrations of LMWH. After seven consecutive days of injection, some rats were sacrificed, and blood and nasal mucosa samples were taken to determine their inflammation status. The remaining acute sinusitis rats were randomly selected for a week of nasal irrigation with normal saline or saline mixed with different concentrations of LMWH. One week later, rats were sacrificed, and samples of blood and nasal mucosa were taken to determine the inflammation status. Results: Rat nasal mucosa in the model group had obvious inflammation. The degree of nasal mucosa inflammation damage in the experimental group was lower than in the experimental control group, proving that LMWH has a protective effect on the nasal mucosa and that the effect correlates with dosage. Irrigation of the nose with saline mixed with LMWH can improve the anti-inflammatory effect. Protein related to the TLR4-MyD88-NF-κB signaling pathway was activated in the acute sinusitis rat model, and LMWH can significantly inhibit its expression. Conclusion: This is the first report of the anti-inflammatory effect of LMWH in acute upper respiratory tract inflammation, together with an explanation of its anti-inflammatory mechanism. The findings contribute a theoretical basis for its potential anti-tumor effect.
Collapse
Affiliation(s)
- Tong Wu
- Department of Otolaryngology-Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China.,Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sihan He
- Institute of Molecular Precision Medicine, Xiangya Hospital and Center for Medical Genetics, Central South University, Changsha, China
| | - Zan Jiao
- Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiang Liang
- Department of Otolaryngology-Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yu Chen
- Department of Otolaryngology-Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Huow Liu
- Department of Otolaryngology-Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yongq Zhang
- Department of Otolaryngology-Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - GuangX He
- Department of Otolaryngology-Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
19
|
Liu Y, Liu Y, Zhang HL, Yu FF, Yin XR, Zhao YF, Ye F, Wu XQ. Amelioratory effect of neoandrographolide on myocardial ischemic-reperfusion injury by its anti-inflammatory and anti-apoptotic activities. ENVIRONMENTAL TOXICOLOGY 2021; 36:2367-2379. [PMID: 34397165 DOI: 10.1002/tox.23350] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
In the present study, we aimed to evaluate the cardioprotective effect of neoandrographolide (Neo) on myocardial ischemia/reperfusion injury (I/R) models and explore its possible mechanism. We randomly and equally divided male mice into sham-operation, I/R, and I/R + Neo groups. H9C2 cell line and primary neonatal rat cardiomyocytes were induced into the simulated I/R's status and used to further validate the Neo's role in vitro. Heart systolic function, indexes of myocardial injury (IMI), infarct size, pathological change, cell apoptosis, inflammatory cytokines, and indexes related to apoptotic and NF-κB signaling pathways were analyzed in vivo or in vitro after the Neo treatment. Compared to the I/R group, Neo significantly suppressed IMI, infarct size, inflammatory cell infiltration, cell apoptosis, inflammatory cytokines, bax, cleaved caspase-3, P-IKBa, and P-NF-κB protein expressions, and the translocation of NF-kB subunit p65 from the cytoplasm to the nucleus in vivo or in vitro. Still, ejected fraction, fractional shortening, and the bcl-2 protein expression were notably increased after the Neo treatment. Neo could be developed into a new drug for treating myocardial I/R by inhibiting myocardial inflammation and apoptosis, which was closely related to suppressing the activation of bax/bcl-2 and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Ying Liu
- Department of Cardiology, the Eighty-first Hospital of PLA Affiliated with Anhui Medical University, Nanjing, China
| | - Yun Liu
- Department of Cardiology, the Eighty-first Hospital of PLA Affiliated with Anhui Medical University, Nanjing, China
| | - Hong-Li Zhang
- Department of Cardiology, the Eighty-first Hospital of PLA Affiliated with Anhui Medical University, Nanjing, China
| | - Fen-Fang Yu
- Department of Respiratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Rui Yin
- Department of Obstetrics and Gynecology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yan-Fang Zhao
- Department of Cardiology, the Eighty-first Hospital of PLA Affiliated with Anhui Medical University, Nanjing, China
| | - Fei Ye
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiang-Qi Wu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
Essawy AE, Abd Elkader HTAE, Khamiss OA, Eweda SM, Abdou HM. Therapeutic effects of astragaloside IV and Astragalus spinosus saponins against bisphenol A-induced neurotoxicity and DNA damage in rats. PeerJ 2021; 9:e11930. [PMID: 34434659 PMCID: PMC8359804 DOI: 10.7717/peerj.11930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/18/2021] [Indexed: 12/28/2022] Open
Abstract
Background Bisphenol A (BPA) is an endocrine disruptor to which humans are often subjected during daily life. This study aimed to investigate the ameliorative effect of astragaloside IV (ASIV) or saponins extracted from Astragalus spinosus (A. spinosus) against DNA damage and neurotoxic effects induced by BPA in prefrontal cortex (PFC), hippocampal and striatal brain regions of developing male rats. Materials and Methods Juvenile PND20 (pre-weaning; age of 20 days) male Sprague Dawley rats were randomly and equally divided into four groups: control, BPA, BPA+ASIV and BPA+A. spinosus saponins groups. Bisphenol A (125 mg/kg/day) was administrated orally to male rats from day 20 (BPA group) and along with ASIV (80 mg/kg/day) (BPA+ASIV group) or A. spinosus saponin (100 mg/kg/day) (BPA+ A. spinosus saponins group) from day 50 to adult age day 117. Results Increased level of nitric oxide (NO) and decreased level of glutamate (Glu), glutamine (Gln), glutaminase (GA) and glutamine synthetase (GS) were observed in the brain regions of BPA treated rats compared with the control. On the other hand, co-administration of ASIV or A. spinosus saponin with BPA considerably improved levels of these neurochemicals. The current study also revealed restoration of the level of brain derived neurotrophic factor (BDNF) and N-methyl-D-aspartate receptors (NR2A and NR2B) gene expression in BPA+ ASIV and BPA+A. spinosus saponins groups. The co-treatment of BPA group with ASIV or A. spinosus saponin significantly reduced the values of comet parameters as well as the intensity of estrogen receptors (ERs) immunoreactive cells and improved the histological alterations induced by BPA in different brain regions. Conclusion It could be concluded that ASIV or A. spinosus saponins has a promising role in modulating the neurotoxicity and DNA damage elicited by BPA.
Collapse
Affiliation(s)
- Amina E Essawy
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | | | - Omaima A Khamiss
- Animal Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI-USC), University of Sadat City, Sadat City, Egypt
| | - Saber Mohamed Eweda
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt.,Department of Medical Laboratories Technology, College of Applied Medical Sciences, Taibah University, Madinah, KSA, Saudi Arabia
| | - Heba Mohamed Abdou
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
21
|
Huang X, Zhang MZ, Liu B, Ma SY, Yin X, Guo LH. Astragaloside IV Attenuates Polymicrobial Sepsis-Induced Cardiac Dysfunction in Rats via IKK/NF-κB Pathway. Chin J Integr Med 2021; 27:825-831. [PMID: 34432200 DOI: 10.1007/s11655-021-2869-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To evaluate the protective effects of Astragaloside IV (AST) in a rat model of myocardial injury induced by cecal ligation and puncture (CLP). METHODS The model of sepsis-induced cardiac dysfunction was induced by CLP. Using a random number table, 50 specific pathogen free grade of Sprague Dawley rats were randomized into 5 groups: the sham group (sham), the model group (CLP, 18 h/72 h) and AST group (18 h/72 h). Except the sham group, the rats in other groups received CLP surgery to induce sepsis. CLP groups received intragastric administration with normal saline after CLP. AST groups received intragastric administration with AST solution (40 mg/kg) once a day. The levels of inflammatory mediators and oxidative stress markers in the serum of the septic rats were determined via enzyme-linked immunosorbent assay (ELISA) at different time point, such as interleukin 6 (IL-6), IL-10, high mobility group box-1 protein B1 (HMGB-1), superoxide dismutase (SOD), and malondialdehyde (MDA). Cardiac function was determined by echocardiography. Moreover, changes in myocardial pathology were evaluated using hematoxylin and eosin staining. The levels of lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) were analysed to determine the status of CLP-induced myocardium. In addition, the apotosis of myocardial cells was analysed by terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL). The protein levels of B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X (Bax), IκB kinase α (IKKα), nuclear factor kappa B p65 (NF-κB p65) were detected by Western blot analysis. Moreover, survival rate was investigated. RESULTS AST improved the survival rate of CLP-induced rats by up to 33.3% (P<0.05). The cardioprotective effect of AST was observed by increased ejection fraction, fractional shortening and left ventricular internal diameter in diastole respectively (P<0.01 or P<0.05). Subsequently, AST attenuated CLP-induced myocardial apoptosis and the ratio of Bcl-2/Bax in the myocardium, as well as the histological alterations of myocardium (P<0.01 or P<0.05); the generation of inflammatory cytokines (IL-6, IL-10, HMGB-1) and oxidative stress markers (SOD, MDA) in the serum was significantly alleviated (P<0.01 or P<0.05). On the other hand, AST markedly suppressed CLP-induced accumulation of IKK-α and NF-κB p65 subunit phosphorylation (P<0.01 or P<0.05). CONCLUSIONS AST plays a significant protective role in sepsis-induced cardiac dysfunction and survival outcome. The possible mechanism of cardioprotection is dependent on the activation of the IKK/NF-κB pathway in cardiomyocytes.
Collapse
Affiliation(s)
- Xin Huang
- Intensive Care Research Team of Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Min-Zhou Zhang
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Bo Liu
- Department of Radiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Shi-Yu Ma
- Intensive Care Research Team of Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Xin Yin
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Li-Heng Guo
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
22
|
Li A, Zhang X, Luo Q. Neohesperidin alleviated pathological damage and immunological imbalance in rat myocardial ischemia-reperfusion injury via inactivation of JNK and NF-κB p65. Biosci Biotechnol Biochem 2021; 85:251-261. [PMID: 33604646 DOI: 10.1093/bbb/zbaa064] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022]
Abstract
Neohesperidin (NEO) exerts antiviral, antioxidant, anti-inflammation, and antitumor effects in some diseases. The purpose of this study was to investigate the effect and mechanism of NEO on myocardial ischemia-reperfusion (I/R) injury. Results indicated that NEO suppressed the levels of serum inflammatory cytokines, myocardial damage markers, and oxidative stress markers, and increased the levels of antioxidant in myocardial I/R rats. NEO also inhibited cell apoptosis. Besides, NEO also inhibited the phosphorylation of c-Jun N-terminal kinases (JNK) and nuclear factor kappa B (NF-κB) p65. Furthermore, the protective effects of NEO on myocardial tissue damage, inflammatory cytokines, myocardial injury markers, oxidative stress markers, cell apoptosis, spleen, thymus and liver indices, and phagocytic indices were reversed by JNK activator and NF-κB activator, respectively. In conclusion, NEO alleviates myocardial damage, oxidative stress, cell apoptosis, and immunological imbalance in I/R injury via the inactivation of JNK and NF-κB, making NEO a potential agent for myocardial I/R therapy.
Collapse
Affiliation(s)
- Aihua Li
- Internal Medicine-Cardiovascular Department, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Xin Zhang
- Internal Medicine-Cardiovascular Department, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Qiuping Luo
- Internal Medicine-Cardiovascular Department, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| |
Collapse
|
23
|
Abd Elkader HTAE, Abdou HM, Khamiss OA, Essawy AE. Anti-anxiety and antidepressant-like effects of astragaloside IV and saponins extracted from Astragalus spinosus against the bisphenol A-induced motor and cognitive impairments in a postnatal rat model of schizophrenia. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:35171-35187. [PMID: 33666843 DOI: 10.1007/s11356-021-12927-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/08/2021] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) is a chemical endocrine disruptor to which humans are often exposed in daily life. Postnatal administration of BPA results in schizophrenia (SCZ)-like behaviours in rats. The present study was designed to elucidate whether treatment with astragaloside IV (ASIV) or saponins extracted from Astragalus spinosus improves the neurobehavioural and neurochemical disturbances induced by BPA. Fifty-two juvenile (PND20) male Sprague Dawley rats were divided into four groups. The rats in Group I were considered the control rats, while the rats in Group II were orally administered BPA (125 mg/kg) daily from PND20 to adult age (PND117). The rats in the third and fourth groups were administered BPA (125 mg/kg/day) supplemented with astragaloside IV (80 mg/kg/d) on PND20 or A. spinosus saponins (100 mg/kg/d) from PND50 to PND117, respectively. Administration of ASIV and saponins extracted from Astragalus spinosus reversed the anxiogenic and depressive-like behaviours and the social defects that were observed in the rats treated with BPA alone. Additionally, these compounds improved memory impairments, restored dopamine (DA), serotonin (5-HT), and monoamine oxidase (MAO-A) levels and normalized Tph2 mRNA expression towards the control values. Taken together, it can be concluded that orally administered ASIV and A. spinosus saponins exhibit neuroprotective effects and that these compounds can be used as therapeutic strategies against BPA-induced neuropsychiatric symptoms in a rat model of SCZ.
Collapse
Affiliation(s)
| | - Heba Mohamed Abdou
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Omaima Ahmed Khamiss
- Department of Genetic Engineering and Biotechnology, Institute of Genetic Engineering and Biotechnology, Sadat City University, Sadat City, Egypt
| | - Amina Essawy Essawy
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
24
|
Salehi B, Carneiro JNP, Rocha JE, Coutinho HDM, Morais Braga MFB, Sharifi-Rad J, Semwal P, Painuli S, Moujir LM, de Zarate Machado V, Janakiram S, Anil Kumar NV, Martorell M, Cruz-Martins N, El Beyrouthy M, Sadaka C. Astragalus species: Insights on its chemical composition toward pharmacological applications. Phytother Res 2021; 35:2445-2476. [PMID: 33325585 DOI: 10.1002/ptr.6974] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/02/2020] [Accepted: 11/24/2020] [Indexed: 01/30/2023]
Abstract
Astragalus L. is widely distributed throughout the temperate regions of Europe, Asia, and North America. The genus is widely used in folk medicine and in dietary supplements, as well as in cosmetics, teas, coffee, vegetable gums, and as forage for animals. The major phytoconstituents of Astragalus species with beneficial properties are saponins, flavonoids, and polysaccharides. Astragalus extracts and their isolated components exhibited promising in vitro and in vivo biological activities, including antiaging, antiinfective, cytoprotective, antiinflammatory, antioxidant, antitumor, antidiabesity, and immune-enhancing properties. Considering their proven therapeutic potential, the aim of this work is to give a comprehensive summary of the Astragalus spp. and their active components, in an attempt to provide new insight for further clinical development of these xenobiotics. This is the first review that briefly describes their ethnopharmacology, composition, biological, and toxicological properties.
Collapse
Affiliation(s)
- Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | | | | | | | | | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Prabhakar Semwal
- Department of Biotechnology, Graphic Era University, Dehradun, India
- Uttarakhand State Council for Science and Technology, Dehradun, India
| | - Sakshi Painuli
- Department of Biotechnology, Graphic Era University, Dehradun, India
| | - Laila Moujir Moujir
- Department of Biochemistry, Microbiology, Molecular Biology and Genetics, University of La Laguna, Tenerife, Spain
| | - Victoria de Zarate Machado
- Department of Biochemistry, Microbiology, Molecular Biology and Genetics, University of La Laguna, Tenerife, Spain
| | - Shriyaa Janakiram
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| | | | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, Centre for Healthy Living, University of Concepción, Concepción, Chile
- Universidad de Concepción, Unidad de Desarrollo Tecnológico, UDT, Concepción, Chile
| | - Natalia Cruz-Martins
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Porto, Portugal
| | | | - Carmen Sadaka
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
25
|
Protective effects of Astragaloside IV against oxidative injury and apoptosis in cultured astrocytes by regulating Nrf2/JNK signaling. Exp Brain Res 2021; 239:1827-1840. [PMID: 33830313 DOI: 10.1007/s00221-021-06096-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/19/2021] [Indexed: 12/24/2022]
Abstract
Ischemic stroke is a worldwide complex brain disease that results in numerous disabilities and deaths. It leads to the deprivation of oxygen and glucose, which causes energy failure and neuronal death. The activation of astrocytes contributes to neuronal damage or repair after brain ischemia/reperfusion, although astrocytes get little attention as potential drug targets. This study investigated the protective effects of Astragaloside IV (AS-IV) on oxygen glucose deprivation/reoxygenation (OGD/R)-induced damage in rat primary cultured astrocytes and the underlying molecular mechanism. The results showed that compared with the control group, astrocytes under OGD/R exposure significantly decreased cell viability and increased the number of apoptotic cells, whereas AS-IV evidently protected the astrocytes against OGD/R-induced cell damage. In addition, low and medium concentrations of AS-IV can promote the increase of intracellular superoxide dismutase (SOD) level, as well as restored the morphological changes caused by OGD/R exposure. Supplementation with AS-IV after OGD/R exposure promoted the expression of oxidation and apoptosis indexes and further study demonstrated that AS-IV inhibited CXCR4 receptor and downregulated the activation of p-JNK/JNK pathway, which suppressed the expression of Bax/Bcl-2, and finally uprising Nrf2/Keap1 signaling. In conclusion, these findings revealed that AS-IV protected against OGD/R-induced astrocytes through inhibiting oxidative stress and apoptotic pathways.
Collapse
|
26
|
Zhang P, Yu Y, Wang P, Shen H, Ling X, Xue X, Yang Q, Zhang Y, Xiao J, Wang Z. Role of Hydrogen Sulfide in Myocardial Ischemia-Reperfusion Injury. J Cardiovasc Pharmacol 2021; 77:130-141. [PMID: 33165141 DOI: 10.1097/fjc.0000000000000943] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022]
Abstract
ABSTRACT Hydrogen sulfide (H2S), generally known as a new gas signal molecule after nitric oxide and carbon monoxide, has been found as an important endogenous gasotransmitter in the last few decades, and it plays a significant role in the cardiovascular system both pathologically and physiologically. In recent years, there is growing evidence that H2S provides myocardial protection against myocardial ischemia-reperfusion injury (MIRI), which resulted in an ongoing focus on the possible mechanisms of action accounting for the H2S cardioprotective effect. At present, lots of mechanisms of action have been verified through in vitro and in vivo models of I/R injury, such as S-sulfhydrated modification, antiapoptosis, effects on microRNA, bidirectional effect on autophagy, antioxidant stress, or interaction with NO and CO. With advances in understanding of the molecular pathogenesis of MIRI and pharmacology studies, the design, the development, and the pharmacological characterization of H2S donor drugs have made great important progress. This review summarizes the latest research progress on the role of H2S in MIRI, systematically explains the molecular mechanism of H2S affecting MIRI, and provides a new idea for the formulation of a myocardial protection strategy in the future.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Yue Yu
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Pei Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Hua Shen
- Department of Cardiovascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Ling
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Xiaofei Xue
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Qian Yang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Yufeng Zhang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Zhinong Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| |
Collapse
|
27
|
Liu YL, Zhang QZ, Wang YR, Fu LN, Han JS, Zhang J, Wang BM. Astragaloside IV Improves High-Fat Diet-Induced Hepatic Steatosis in Nonalcoholic Fatty Liver Disease Rats by Regulating Inflammatory Factors Level via TLR4/NF-κB Signaling Pathway. Front Pharmacol 2021; 11:605064. [PMID: 33708118 PMCID: PMC7941269 DOI: 10.3389/fphar.2020.605064] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/22/2020] [Indexed: 01/18/2023] Open
Abstract
Objective: Astragaloside IV (AS-IV) is the primary bioactive component purified from Astragalus membranaceus which is one of the traditional Chinese medicines. Research studies found that AS-IV has significant pharmacological effects on focal cerebral ischemia/reperfusion, cardiovascular disease, pulmonary disease, liver cirrhosis, and diabetic nephropathy, but little is known about the effects of AS-IV on nonalcoholic fatty liver disease (NAFLD). In this study, we investigated whether AS-IV has beneficial effects on NAFLD in rats and its potential mechanisms. Methods: Male SD rats were fed with high-fat diet (HFD) for 12 weeks to establish NAFLD rat model, and then, the rats were divided into five groups. The control group rats were fed with normal diet for 12 weeks and then were given normal saline (1.0 ml kg−1 day−1) by intragastric administration for 4 weeks. The model group rats were fed with HFD for 12 weeks and then were given normal saline (1.0 ml kg−1 day−1) by intragastric administration for 4 weeks. The AS-IV-L, AS-IV-M, and AS-IV-H groups were treated with 20, 40, and 80 mg kg−1 day−1 of AS-IV by intragastric administration for 4 weeks and given HFD diet. Then, we detected serum transaminase (ALT, AST), blood lipid (TG, TC), inflammatory cytokines (IL-6, IL-8 and TNF-α), liver histology(NAFLD activity score), TLR4/MyD88 signaling pathway in liver tissue. Results: We found AS-IV significantly reduced serum levels of AST, ALT, TG, TNF-α, IL-6, and IL-8 in NAFLD rats and downregulate the expression of TLR4 mRNA, MyD88 mRNA, NF-κB mRNA, and proteins in liver tissue. Moreover, AS-IV could significantly reduce the NAFLD activity score of NAFLD rat liver. Conclusion: In this study, we demonstrated that AS-IV have a protective effect on NAFLD by inhibiting TNF-α, IL-6 and IL-8 levels and down-regulating TLR4, MyD88 and NF-κB expression in rat liver tissues.
Collapse
Affiliation(s)
- Ying-Li Liu
- Gastroenterology, the Fourth Central Clinical College, Tianjin Medical University, Tianjin, China.,Gastroenterology, Tianjin Fourth Central Hospital, Tianjin, China
| | - Qiu-Zan Zhang
- Gastroenterology, the Fourth Central Clinical College, Tianjin Medical University, Tianjin, China.,Gastroenterology, Tianjin Fourth Central Hospital, Tianjin, China
| | - Yan-Rong Wang
- Gastroenterology, the Fourth Central Clinical College, Tianjin Medical University, Tianjin, China.,Gastroenterology, Tianjin Fourth Central Hospital, Tianjin, China
| | - Li-Na Fu
- Gastroenterology, the Fourth Central Clinical College, Tianjin Medical University, Tianjin, China.,Gastroenterology, Tianjin Fourth Central Hospital, Tianjin, China
| | - Jing-Shu Han
- Gastroenterology, the Fourth Central Clinical College, Tianjin Medical University, Tianjin, China.,Gastroenterology, Tianjin Fourth Central Hospital, Tianjin, China
| | - Jing Zhang
- Gastroenterology, the Fourth Central Clinical College, Tianjin Medical University, Tianjin, China.,Gastroenterology, Tianjin Fourth Central Hospital, Tianjin, China
| | - Bang-Mao Wang
- Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
28
|
Su HF, Shaker S, Kuang Y, Zhang M, Ye M, Qiao X. Phytochemistry and cardiovascular protective effects of Huang-Qi (Astragali Radix). Med Res Rev 2021; 41:1999-2038. [PMID: 33464616 DOI: 10.1002/med.21785] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/27/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
Huang-Qi (Astragali Radix) is an herbal tonic widely used in China and many other countries. It is derived from the roots of Astragalus membranaceus and A. membranaceus var. mongholicus and shows potent cardiovascular protective effects. In this article, we comprehensively reviewed 189 small molecules isolated from the two Astragalus species and discussed the interspecies chemical differences. Moreover, we summarized the pharmacological activities and mechanisms of action of Huang-Qi and its major bioactive compounds for the treatment of cardiovascular diseases. This review covers 171 references published between February 1983 and March 2020.
Collapse
Affiliation(s)
- Hui-Fei Su
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Sharpkate Shaker
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yi Kuang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Meng Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Min Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University, Beijing, China
| | - Xue Qiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
29
|
Astragaloside and/or Hydroxysafflor Yellow A Attenuates Oxygen-Glucose Deprivation-Induced Cultured Brain Microvessel Endothelial Cell Death through Downregulation of PHLPP-1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3597527. [PMID: 33381198 PMCID: PMC7755473 DOI: 10.1155/2020/3597527] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/19/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
The incidence of ischemic stroke, a life-threatening condition in humans, amongst Asians is high and the prognosis is poor. In the absence of effective therapeutics, traditional Chinese medicines have been used that have shown promising results. It is crucial to identify traditional Chinese medicine formulas that protect the blood-brain barrier, which is damaged by an ischemic stroke. In this study, we aimed to elucidate such formulas. Brain microvascular endothelial cells (BMECs) were used to establish an in vitro ischemia-reperfusion model for oxygen-glucose deprivation (OGD) experiments to evaluate the function of two traditional Chinese medicines, namely, astragaloside (AS-IV) and hydroxysafflor yellow A (HSYA), in protecting against BMEC. Our results revealed that AS-IV and HSYA attenuated the cell loss caused by OGD by increasing cell proliferation and inhibiting cell apoptosis. In addition, these compounds promoted the migration and invasion of BMECs in vitro. Furthermore, we found that BMECs rescued by AS-IV and HSYA could be functionally activated in vitro, with AS-IV and HSYA showing synergetic effects in rescuing BMECs survival in vitro by reducing the expression of PHLPP-1 and activating Akt signaling. Our results elucidated the potential of AS-IV and HSYA in the prevention and treatment of stroke by protecting against cerebral ischemia-reperfusion injury.
Collapse
|
30
|
Li D, Li G, Chen Y, Li Y, Zhang J, Gao D, Sun L, Liu B. Astragaloside IV protects ATDC5 cells from lipopolysaccharide-caused damage through regulating miR-203/MyD88. PHARMACEUTICAL BIOLOGY 2020; 58:89-97. [PMID: 31906765 PMCID: PMC6968705 DOI: 10.1080/13880209.2019.1705355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/21/2019] [Accepted: 12/10/2019] [Indexed: 06/10/2023]
Abstract
Context: Osteoarthritis (OA) is a degenerative arthrosis sickness. Astragaloside IV (AS-IV) functions by relieving inflammatory damage.Objective: We aimed to investigate the mechanism by which AS-IV protects ATD cells from lipopolysaccharide (LPS)-induced damage.Materials and methods: ATDC5 cells were transfected with miR-203 inhibitor and NC inhibitor (150 nM) or pEX-MyD88 and sh-MyD88 (50 nM) for 48 h, pre-treated by 15 μg/mL AS-IV for 24 h, then treated by 5 μg/mL LPS for 12 h. Dual-luciferase activity testing was used to determine whether miR-203 could bind to MyD88. CCK-8 and flow cytometry were used to detect cell activity and apoptosis, respectively, and qRT-PCR, western blots, and ELISA were performed to detect expression levels of miR-203 and inflammatory cytokines.Results: Based on the 50% inhibiting concentration (IC50), there was no significant difference of AS-IV (0 to 15 μg/mL) on cell viability. Fifteen μg/mL was the optimal concentration of AS-IV in treating LPS-induced inflammatory damage in subsequent experiments since this was a semi-lethal concentration. AS-IV significantly reduces LPS-induced viability, apoptosis and the release of TNF-α, IL-6 and iNOS mainly through up-regulating miR-203. Further, MyD88 was a target gene of miR-203 and negatively regulated by miR-203. Knockdown of MyD88 inhibited LPS-induced inflammatory damage by inhibiting the NF-κB signal pathway.Discussion and conclusions: AS-IV protects ATDC5 cells against LPS-induced damage mainly via regulating miR-203/MyD88. Our results support a theoretical basis for in-depth study of the function of AS-IV and the clinical cure of OA.
Collapse
Affiliation(s)
- Dexin Li
- Department of Sports Medicine, Jilin City Central Hospital, Jilin, China
| | - Guangcheng Li
- Department of Joint Surgery, Jilin City Central Hospital, Jilin, China
| | - Yang Chen
- Department of Dermatology, Jilin City Central Hospital, Jilin, China
| | - Yifei Li
- Department of Joint Surgery, Jilin City Central Hospital, Jilin, China
| | - Junfeng Zhang
- Department of Joint Surgery, Jilin City Central Hospital, Jilin, China
| | - Dexuan Gao
- Department of Joint Surgery, Jilin City Central Hospital, Jilin, China
| | - Linglong Sun
- Department of Sports Medicine, Jilin City Central Hospital, Jilin, China
| | - Bo Liu
- Department of Joint Surgery, Jilin City Central Hospital, Jilin, China
| |
Collapse
|
31
|
Chen L, Song M, Zhang L, Li C, Fang Z, Coffie JW, Zhang L, Ma L, Fang L, Wang Q, Yang W, Li F, Gao X, Wang H. The protective effects of different compatibility proportions of the couplet medicines for Astragali Radix and Angelica sinensis Radix on myocardial infarction injury. PHARMACEUTICAL BIOLOGY 2020; 58:165-175. [PMID: 32608342 PMCID: PMC8641686 DOI: 10.1080/13880209.2020.1725581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 11/27/2019] [Accepted: 01/30/2020] [Indexed: 05/15/2023]
Abstract
Context: Astragali Radix (AR) and Angelica sinensis Radix (ASR) combinations are used to treat cardiovascular disorders.Objectives: This study investigates the protective effects of different compatibility proportions of AR and ASR on cardiac dysfunction in a C57BL/6 mouse model of myocardial infarction (MI).Materials and methods: MI mice were induced by ligation of the left coronary artery and divided into six groups: sham, vehicle, 10 mg/kg/d simvastatin and combinations of AR and ASR at different ratios, including 1:1 (AR 2.5 g/kg + ASR 2.5 g/kg), 3:1 (AR 3.75 g/kg + ASR 1.25 g/kg) and 5:1 (AR 4.17 g/kg + ASR 0.83 g/kg). Both AR-ASR combinations and simvastatin were dissolved in saline solution and given daily by gavage. The left ventricle function, infarct size, heart tissue injury, apoptosis of cardiomyocytes, leukocyte infiltrates, capillary density and expression of cleaved caspase-3, cleaved caspase-9, Bcl-2, Bax, Bad, IL-1β, IL-6, VEGF, p-Akt and p-eNOS were analysed.Results: Different combinations of AR and ASR improve cardiac function and reduce infarct size (61.15% vs. 39.3%, 42.65% and 45.5%) and tissue injury through different mechanisms. When AR was combined with ASR at ratio of 1:1, the inflammation and cardiomyocyte apoptosis were suppressed (p < 0.05, p < 0.01). The combination ratio of 3:1 exerted effect in promoting angiogensis (p < 0.05). In the combination of AR and ASR at 5:1 ratio, angiogenesis was significantly improved (p < 0.01) and the apoptosis was inhibited (p < 0.05).Conclusions: Our results reflect the regulation of multiple targets and links in herb pairs and provide an important basis for the use of AR and ASR combinations in the treatment of MI.
Collapse
Affiliation(s)
- Lu Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Min Song
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lusha Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chunxiao Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhirui Fang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Joel Wake Coffie
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Liyuan Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lulu Ma
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Leyu Fang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianyi Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenjie Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fanggang Li
- Shandong Danhong Pharmaceutical Co., Ltd., Heze, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
32
|
Zhang Y, Zhao Q. AEG-1 deletion promotes cartilage repair and modulates bone remodeling-related cytokines via TLR4/MyD88/NF-κB inhibition in ovariectomized rats with osteoporosis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1298. [PMID: 33209878 PMCID: PMC7661885 DOI: 10.21037/atm-20-5842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background Osteoporosis is a systemic skeletal disorder that can impact a variety of bones throughout the body. Astrocyte-elevated gene-1 (AEG-1) is involved in multiple pro-tumorigenic functions and participates in various inflammatory reactions. However, whether it has an impact on osteoporosis-related cartilage repair and bone remodeling remains unknown. Methods We utilized an ovariectomy mouse model with AEG-1 deletion to investigate the role of AEG-1 in osteoporosis. The mRNA level of AEG-1 was detected by RT-PCR, bone markers, bone volume/total volume (BV/TV), trabecular bone surface/bone volume (BSA/BV) and trabecular bone thickness (Tb. Th) were detected by micro computed tomography (µCT), bone injury was observed by HE and alcian blue staining. The contents of IL-6, IL-17, iNOS and IL-10 in peripheral blood of the three groups were detected by ELISA. The expression of OSX, coi1a1, OC, TLR4, MyD88 and NF-κB were detected by Western Blot. Results µCT revealed increased bone volume in the AEG-1 knockout (KO) ovariectomy (OVX) group compared to the wildtype (WT) OVX group 4 weeks after surgery, indicating restored bone formation after AEG-1 deletion. Flow sorting revealed that AEG-1 deletion inhibited the production of inflammatory factors. Western blot demonstrated activation of the TLR4/MyD88/NF-κB pathway after LPS exposure, which was reduced by AEG-1 deletion. AEG-1 deletion also improved lipopolysaccharide (LPS) induced adverse reactions. Conclusions Taken together, these findings indicate that AEG-1 deletion improves cartilage repair and bone remodeling during osteoporosis, which may partly occur through the inhibition of the TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Rheumatology, Henan University Huai He Hospital, Kaifeng City, China
| | - Qing Zhao
- Department of Rheumatology, Henan University Huai He Hospital, Kaifeng City, China
| |
Collapse
|
33
|
Yuan H, Yang G, Li S, Li L, Wei T, Song G, Luan H, Meng J, Wang Q, Yu Y, Sun J. Calcium sensing receptor involving in therapy of embryonic stem cell transplantation alleviates acute myocardial infarction by inhibiting apoptosis and oxidative stress in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:1353-1359. [PMID: 33149870 PMCID: PMC7585542 DOI: 10.22038/ijbms.2020.47436.10916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Objective(s): The aims of the present study were to investigate the expression of calcium sensing receptor (CaSR) at different times in acute myocardial infarction (AMI) rat myocardial tissue after mouse embryonic stem cells (mESCs) transplantation treatment and to assess its effects on apoptosis and oxidative stress of cardiomyocytes. Materials and Methods: The AMI rats were treated with mESCs, Calindol (a CaSR agonist) and Calhex231 (a CaSR inhibitor). Serum measurements, Echocardiographic analysis and TUNEL assay were performed. Myocardial ultrastructure changes were viewed by electron microscopy. Additionally, western blotting was used to detect the protein expressions. Results: Compared to the sham group, it was found that the expression levels of CaSR, caspase-3, cytoplasmic cytochrome C (cyt-C) and Bcl2-associated x (Bax), and the levels of Malondialdehyde (MDA) were significantly increased in both AMI and AMI + mESCs + Calindol groups with the development of myocardial infarction. Furthermore, the ultra-microstructure of cardiomyocyte was highly damaged, the expression levels of mitochondrial cyt-C and B-cell lymphoma 2 (Bcl-2) were significantly decreased, and there was decreased activity of superoxide dismutase (SOD). However, the combination of Calhex231 and mESCs transplantation could inhibit these changes. Conclusion: Our results suggested that CaSR expression in myocardial tissue of AMI rats was increased over time, and that Calhex231 could enhance the efficacy of ESCs transplantation for the treatment of AMI, which would be a new therapeutic strategy for the treatment of AMI.
Collapse
Affiliation(s)
- Hui Yuan
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Guohong Yang
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Shu Li
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Li Li
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Tao Wei
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Gaochen Song
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Hairong Luan
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Jin Meng
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Qi Wang
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yaquan Yu
- Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Jian Sun
- Mudanjiang Medical University, Mudanjiang, 157011, China
| |
Collapse
|
34
|
Tan YQ, Chen HW, Li J. Astragaloside IV: An Effective Drug for the Treatment of Cardiovascular Diseases. Drug Des Devel Ther 2020; 14:3731-3746. [PMID: 32982178 PMCID: PMC7507407 DOI: 10.2147/dddt.s272355] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD), the number one cause of death worldwide, has always been the focus of clinical and scientific research. Due to the high number of deaths each year, it is essential to find alternative therapies that are safe and effective with minimal side effects. Traditional Chinese medicine (TCM) has a long history of significant impact on the treatment of CVDs. The mode of action of natural active ingredients of drugs and the development of new drugs are currently hot topics in research on TCM. Astragalus membranaceus is a commonly used Chinese medicinal herb. Previous studies have shown that Astragalus membranaceus has anti-tumor properties and can regulate metabolism, enhance immunity, and strengthen the heart. Astragaloside IV (AS-IV) is the active ingredient of Astragalus membranaceus, which has a prominent role in cardiovascular diseases. AS-IV can protect against ischemic and hypoxic myocardial cell injury, inhibit myocardial hypertrophy and myocardial fibrosis, enhance myocardial contractility, improve diastolic dysfunction, alleviate vascular endothelial dysfunction, and promote angiogenesis. It can also regulate blood glucose and blood lipid levels and reduce the risk of cardiovascular diseases. In this paper, the mechanism of AS-IV intervention in cardiovascular diseases in recent years is reviewed in order to provide a reference for future research and new drug development.
Collapse
Affiliation(s)
- Yu-Qing Tan
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing100053, People’s Republic of China
- Graduate School of Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing100029, People’s Republic of China
| | - Heng-Wen Chen
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing100053, People’s Republic of China
| | - Jun Li
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing100053, People’s Republic of China
| |
Collapse
|
35
|
An updated role of astragaloside IV in heart failure. Biomed Pharmacother 2020; 126:110012. [DOI: 10.1016/j.biopha.2020.110012] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/03/2020] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
|
36
|
Astragaloside IV protects human cardiomyocytes from hypoxia/reoxygenation injury by regulating miR-101a. Mol Cell Biochem 2020; 470:41-51. [PMID: 32394311 PMCID: PMC7272390 DOI: 10.1007/s11010-020-03743-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/02/2020] [Indexed: 12/27/2022]
Abstract
Astragaloside IV (AS/IV) is one of the extracted components from the traditional Chinese medicine Astragalus which has been demonstrated to have potential capacity for anti-inflammation activity and for treating cardiovascular disease. Our purpose was to determine the function and underlying molecular mechanism of AS/IV in hypoxia/reoxygenation (H/R) injured in cardiomyocytes. Differentially expressed genes (DEGs) were screened using bioinformatic analysis, and the molecular targeting relationship was verified by the dual-luciferase report system. H/R injured cardiomyocytes were employed to explore the effect of AS/IV. QRT-PCR and Western blot analysis were applied to detect the expression of mRNA and proteins, respectively. Additionally, superoxide dismutase (SOD), lactic dehydrogenase (LDH) and MDA (malondialdehyde) levels were detected to determine the oxidative damage. Cell viability was assessed by CCK-8, and flow cytometry was used to evaluate cell apoptosis ratio. TGFBR1 and TLR2 were selected as DEGs. Additionally, AS/IV could enhance cell proliferation and upregulated miR-101a expression, which suppressed TGFBR1 and TLR2 expression in H/R injured cardiomyocytes. Moreover, the results of Western blot exhibited that the downstream genes (p-ERK and p-p38) in the MAPK signaling pathway were suppressed, which meant AS/IV could inhibit this pathway in H/R injured cardiomyocytes. Overall, this study demonstrated AS/IV could attenuate H/R injury in human cardiomyocytes via the miR-101a/TGFBR1/TLR2/MAPK signaling pathway axis, which means that it could serve as a possible alternate for H/R treatment.
Collapse
|
37
|
Jiang S, Jiao G, Chen Y, Han M, Wang X, Liu W. Astragaloside IV attenuates chronic intermittent hypoxia-induced myocardial injury by modulating Ca 2+ homeostasis. Cell Biochem Funct 2020; 38:710-720. [PMID: 32306464 DOI: 10.1002/cbf.3538] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/12/2020] [Accepted: 03/29/2020] [Indexed: 12/16/2022]
Abstract
Obstructive sleep apnea syndrome (OSAS) is an important consequence of chronic intermittent hypoxia (CIH). Astragaloside IV (AS-IV) exerts multiple protective effects in diverse diseases. However, whether AS-IV can attenuate CIH-induced myocardial injury is unclear. In this study, rats exposed to CIH were established and treated with AS-IV for 4 weeks. In vitro, H9C2 cardiomyocytes subjected to CIH exposure were treated with AS-IV for 48 hours. Then the cardiac function, morphology, fibrosis, apoptosis and Ca2+ homeostasis were determined to assess cardiac damage. Results showed that AS-IV attenuated cardiac dysfunction and histological lesions in CIH rats. The increased TUNEL-positive cells and activated apoptotic proteins in CIH rats were reduced by AS-IV. We also noticed that AS-IV reversed the accumulation of Ca2+ and altered expressions of Ca2+ handling proteins (decreases of SERCA2a and RYR2, and increases of p-CaMKII and NCX1) under CIH exposure. Furthermore, CIH-induced reduction of SERCA2a activity was increased by AS-IV in rats. Similar results were also observed in H9C2 cells. Altogether, these findings indicate that AS-IV modulates Ca2+ homeostasis to inhibit apoptosis, protecting against CIH-induced myocardial injury eventually, suggesting it may be a potential agent for cardiac damage of OSAS patients. SIGNIFICANCE OF THE STUDY: Chronic intermittent hypoxia (CIH) is a great contributor of OSAS, which is closely associated with cardiovascular diseases. It is necessary for developing a promising drug to attenuate CIH-induced myocardial injury. This work suggests that AS-IV can attenuate myocardial apoptosis and calcium disruption, thus protecting against CIH-induced myocardial injury. It may represent a novel therapeutic for cardiac damage of OSAS.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Guangyu Jiao
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yunqiu Chen
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Mingxin Han
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xinzhuo Wang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Wenjuan Liu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
38
|
Yang M, Hua T, Yang Z, Chen L, Zou Y, Huang X, Li J. The Protective Effect of rhBNP on Postresuscitation Myocardial Dysfunction in a Rat Cardiac Arrest Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6969053. [PMID: 32149124 PMCID: PMC7049428 DOI: 10.1155/2020/6969053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE We investigated the protective effects and the underlying mechanisms through which recombinant human brain natriuretic peptide (rhBNP) acts on postresuscitation myocardial dysfunction (PRMD) in the cardiac arrest (CA) model. METHODS Ventricular fibrillation was induced and untreated for 6 min. And the time of cardiopulmonary resuscitation was 8 min, after which defibrillation was attempted in this rat model. 24 Sprague Dawley rats (450-550g) were randomized into cardiopulmonary resuscitation (CPR) + rhBNP and CPR + placebo groups after restoration of spontaneous circulation (ROSC). rhBNP was infused at PR 30 min (loading dose: 1.5 µg/kg, 3 min; maintenance dose: 0.01 µg/kg, 3 min; maintenance dose: 0.01 α (TNF-α (TNF-α (TNF-κB (NF-κB (NF. RESULTS The administration of rhBNP attenuated the severity of PRMD and myocardial tissue injuries, with improvement of MAP (mean arterial blood pressure), ETCO2 (end-tidal CO2), serum level of NT-proBNP, EF, CO, and MPI values. The serum levels and protein expression levels in myocardial tissue of IL-6 and TNF-α (TNF-κB (NF. CONCLUSION Our research demonstrated that the administration of rhBNP attenuated the severity of PRMD and myocardial tissue injuries and increased the 24 h survival rate in this CA model. rhBNP administration also reduced the serum and myocardial tissue levels of IL-6 and TNF-α after ROSC, likely due to the suppression of the TLR4/NF-κB signaling pathway and the regulation of inflammatory mediator secretion.α (TNF-κB (NF.
Collapse
Affiliation(s)
- Min Yang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Tianfeng Hua
- Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Zhengfei Yang
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Limin Chen
- Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yangyang Zou
- Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaohui Huang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun Li
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
39
|
Effects and Mechanisms of Traditional Chinese Herbal Medicine in the Treatment of Ischemic Cardiomyopathy. Pharmacol Res 2020; 151:104488. [DOI: 10.1016/j.phrs.2019.104488] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/28/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022]
|
40
|
Zhang Z, Wang J, Zhu Y, Zhang H, Wang H. Astragaloside IV alleviates myocardial damage induced by type 2 diabetes via improving energy metabolism. Mol Med Rep 2019; 20:4612-4622. [PMID: 31702040 PMCID: PMC6797977 DOI: 10.3892/mmr.2019.10716] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to evaluate the protective effect and mechanism of Astragaloside IV (ASIV) on myocardial injury induced by type 2 diabetes, with a focus on energy metabolism. Blood glucose, the hemodynamic index, left ventricular weight/heart weight (LVW/HW), the left ventricular systolic pressure (LVSP), the left ventricular end diastolic pressure (LVEDP) and cell survival rate were measured in streptozotocin‑induced diabetes model rats. Western blot analysis, PCR, hematoxylin‑eosin and TUNEL staining, flow cytometry and ELISA were used to detect: i) Cardiomyocyte damage indicators such as atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), cytochrome c (Cyt C), caspase‑3, cleaved caspase‑3 and the apoptotic rate; ii) energy metabolism indicators such as ATP/AMP and ADP/AMP; and iii) energy metabolism associated pathway proteins such as peroxisome proliferator‑activated receptor γ coactivator 1‑α (PGC‑1α) and nuclear respiratory factor 1 (NRF1). The present demonstrated increased blood glucose, LVW/HW, LVSP, LVEDP and the cardiomyocyte damage indicators (ANP, BNP, Cyt C and caspase‑3), in the diabetic and high glucose‑treated groups, which were decreased by ASIV. The expression of NRF‑1 and PGC‑1α significantly changed in the model group and was markedly improved following ASIV treatment. Furthermore, the abnormal energy metabolism in the model group was reversed by ASIV. According to the results, ASIV can regulate energy metabolism by regulating the release of PGC‑1α and NRF1 to rescue the abnormal energy metabolism caused by diabetes mellitus, thus decreasing the myocardial damage caused by diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Zhen Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Jing Wang
- The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Yingwei Zhu
- Institute of Physical Education, Bohai University, Jinzhou, Liaoning 121013, P.R. China
| | - Hui Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
41
|
Zhang X, Li M, Wang H. Astragaloside IV Alleviates the Myocardial Damage Induced by Lipopolysaccharide via the Toll-Like Receptor 4 (TLR4)/Nuclear Factor kappa B (NF-κB)/Proliferator-Activated Receptor α (PPARα) Signaling Pathway. Med Sci Monit 2019; 25:7158-7168. [PMID: 31545785 PMCID: PMC6775796 DOI: 10.12659/msm.916030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We previously reported that astragaloside IV (As-IV) can alleviate myocardial damage induced by lipopolysaccharide (LPS). However, the anti-inflammatory effects of As-IV following LPS stimulation in mice and H9C2 cardiomyocytes remain unclear. The present study was designed to explore the mechanism of action of As-IV. MATERIAL AND METHODS In vivo, C57BL/6J mice were randomly divided into 5 groups: the control group, the LPS group (10 mg/kg), and 3 LPS groups receiving different doses of As-IV (20, 40, and 80 mg/kg). The protective effect of As-IV on LPS-stimulated H9C2 cardiomyocytes was evaluated in vitro. Cardiac function was detected by echocardiography, and H&E staining was used to evaluate morphologic changes. Cardiomyocyte viability was detected by MTT assay. ELISA was used to detect free fatty acid (FFA), interleukin-6 (IL-6), interleukin-1ß (IL-1ß), and tumor necrosis factor alpha (TNF-alpha) levels in mouse serum and in cell supernatant. Adenosine triphosphate (ATP) and adenosine monophosphate (AMP) contents in myocardial tissues and cells were detected by high-performance liquid chromatography. ATP5D and TLR4/NF-kappaB/PPARalpha signaling pathway proteins (TLR4, NF-kappaB, p65, and PPARalpha) were detected by Western blotting. RESULTS As-IV significantly improved cardiac function, myocardial cell viability, and pathological changes and reduced FFA, IL-1ß, IL-6, and TNF-alpha levels. The ATP/AMP ratio in the cardiac tissues of mice and in H9C2 cardiomyocytes was increased compared to that in the LPS group. In addition, As-IV enhanced ATP synthase and PPARalpha protein expression. In H9C2 cardiomyocytes, the p65-specific inhibitor BAY11-7082 exerted similar effects as As-IV. CONCLUSIONS As-IV alleviates LPS-induced myocardial damage by modulating TLR4/NF-kappaB/PPARalpha signaling-mediated energy biosynthesis.
Collapse
Affiliation(s)
- Xiaoyao Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| | - Mengfei Li
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| |
Collapse
|
42
|
Han JY, Li Q, Pan CS, Sun K, Fan JY. Effects and mechanisms of QiShenYiQi pills and major ingredients on myocardial microcirculatory disturbance, cardiac injury and fibrosis induced by ischemia-reperfusion. Pharmacol Res 2019; 147:104386. [DOI: 10.1016/j.phrs.2019.104386] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
|
43
|
Zhang KJ, Zheng Q, Zhu PC, Tong Q, Zhuang Z, Zhu JZ, Bao XY, Huang YY, Zheng GQ, Wang Y. Traditional Chinese Medicine for Coronary Heart Disease: Clinical Evidence and Possible Mechanisms. Front Pharmacol 2019; 10:844. [PMID: 31427964 PMCID: PMC6688122 DOI: 10.3389/fphar.2019.00844] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 07/01/2019] [Indexed: 12/23/2022] Open
Abstract
Coronary heart disease (CHD) remains a major cause of mortality with a huge economic burden on healthcare worldwide. Here, we conducted a systematic review to investigate the efficacy and safety of Chinese herbal medicine (CHM) for CHD based on high-quality randomized controlled trials (RCTs) and summarized its possible mechanisms according to animal-based researches. 27 eligible studies were identified in eight database searches from inception to June 2018. The methodological quality was assessed using seven-item checklist recommended by Cochrane Collaboration. All the data were analyzed using Rev-Man 5.3 software. As a result, the score of study quality ranged from 4 to 7 points. Meta-analyses showed CHM can significantly reduce the incidence of myocardial infarction and percutaneous coronary intervention, and cardiovascular mortality (P < 0.05), and increase systolic function of heart, the ST-segment depression, and clinical efficacy (P < 0.05). Adverse events were reported in 11 studies, and CHMs were well tolerated in patients with CHD. In addition, CHM exerted cardioprotection for CHD, possibly altering multiple signal pathways through anti-inflammatory, anti-oxidation, anti-apoptosis, improving the circulation, and regulating energy metabolism. In conclusion, the evidence available from present study revealed that CHMs are beneficial for CHD and are generally safe.
Collapse
Affiliation(s)
- Ke-Jian Zhang
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Zheng
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peng-Chong Zhu
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiang Tong
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhuang Zhuang
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia-Zhen Zhu
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Yi Bao
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue-Yue Huang
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Wang
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
44
|
Du J, Liu J, Zhen J, Yang ST, Zheng EL, Leng JY. Astragaloside IV protects cardiomyocytes from hypoxia-induced injury by down-regulation of lncRNA GAS5. Biomed Pharmacother 2019; 116:109028. [DOI: 10.1016/j.biopha.2019.109028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 01/05/2023] Open
|
45
|
Nutritional preconditioning induced by astragaloside Ⅳ on isolated hearts and cardiomyocytes against myocardial ischemia injury via improving Bcl-2-mediated mitochondrial function. Chem Biol Interact 2019; 309:108723. [PMID: 31228469 DOI: 10.1016/j.cbi.2019.06.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/25/2019] [Accepted: 06/17/2019] [Indexed: 12/30/2022]
Abstract
Ischemic preconditioning and pharmacological preconditioning are common strategies to prevent lethal myocardial injury, especially nutritional preconditioning (NPC). In this study, we investigated the effects of astragaloside IV (Ast), as an NPC agent, on myocardium suffered anoxia/reoxygenation (A/R) injury. Rats received 5 mg/kg Ast daily for 3 weeks by intragastric administration. Then, hearts were harvested and underwent A/R treatment using a Langendorff apparatus. Ast- pretreatment significantly promoted functional recovery of the myocardium, reduced infarct size, and oxidative stress, and decreased the apoptotic index. Similar findings were demonstrated in H9c2 cardiomyocytes that were pretreated with Ast for 24 h. Moreover, Ast-pretreatment significantly upregulated Bcl-2 expression, especially in mitochondria. The effects of Ast treatment against A/R injury were also reflected by increased antioxidant potential, inhibited reactive oxygen species (ROS) burst, increased oxygen consumption rate, maintained mitochondrial membrane potential (MMP), inhibited mitochondrial permeability transition pore (mPTP) opening, and prevented apoptosis. Selective inhibition of Bcl-2 by ABT-737 decreased myocardial injury protection of Ast. Ast-pretreatment resulted in NPC- related effects against A/R, and mitochondria may be the target of a cascade of events elicited by upregulating Bcl-2 expression, promoting translocation of Bcl-2 into mitochondria, maintaining MMP, inhibiting ROS bursts, thereby leading to recovery of mitochondrial respiration, preventing mPTP opening, decreasing cytochrome C release, preventing apoptosis, and ultimately alleviating myocardial injury.
Collapse
|
46
|
Yin B, Hou XW, Lu ML. Astragaloside IV attenuates myocardial ischemia/reperfusion injury in rats via inhibition of calcium-sensing receptor-mediated apoptotic signaling pathways. Acta Pharmacol Sin 2019; 40:599-607. [PMID: 30030530 DOI: 10.1038/s41401-018-0082-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/20/2018] [Indexed: 02/07/2023]
Abstract
Astragaloside IV (AsIV) is an active saponin extracted from Astragalus membranaceus, which has shown cardioprotective effects in a number of experimental animals. In this study we investigated the molecular mechanisms by which AsIV attenuated the myocardial ischemia reperfusion (MI/R)-induced injury in vitro and in vivo by focusing on calcium-sensing receptor (CaSR) and extracellular signal-regulated kinase 1/2 (ERK1/2). Rat neonatal cardiac myocytes were subjected to a hypoxia/reoxygenation (H/R) procedure in vitro, which significantly decreased the cell viability, increased lactate dehydrogenase (LDH) release, induced cardiomyocyte apoptosis, and increased [Ca2+]i. H/R also increased the expression of CaSR and decreased ERK1/2 phosphorylation levels in H/R-exposed myocytes. Pretreatment with AsIV (60 μmol/L) significantly improved the cell viability and decreased LDH release, attenuated myocyte apoptosis, decreased [Ca2+]i and CaSR expression, and increased the ERK1/2 phosphorylation levels. The protective effects of AsIV against H/R injury were partially inhibited by co-treatment with a CaSR agonist, gadolinium chloride (GdCl3) or with a specific ERK1/2 inhibitor U0126. For in vivo studies, a rat MI/R model was established. Pre-administration of AsIV (80 mg/kg every day, ig) significantly decreased the myocardium infarct size, creatine kinase-MB (CK-MB) production, serum cardiac troponin (cTnI) levels, and cardiomyocyte apoptosis in the rats with MI/R injury. The therapeutic effects of AsIV were associated with the downregulation of CaSR expression and upregulation of ERK1/2 phosphorylation in myocardial tissues. In summary, astragaloside IV attenuates myocardial I/R injury via inhibition of CaSR/ERK1/2 and the related apoptotic signaling pathways.
Collapse
|
47
|
Yang P, Zhou Y, Xia Q, Yao L, Chang X. Astragaloside IV Regulates the PI3K/Akt/HO-1 Signaling Pathway and Inhibits H9c2 Cardiomyocyte Injury Induced by Hypoxia-Reoxygenation. Biol Pharm Bull 2019; 42:721-727. [PMID: 30867343 DOI: 10.1248/bpb.b18-00854] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Astragaloside IV (AS-IV) is one of the main pharmacologically active compounds found in Astragalus membranaceus. AS-IV has protective effects against ischemia-reperfusion injury (IRI), but its mechanism of action has not yet been determined. This study aims to investigate the effect of AS-IV on IRI and its effect on the phosphadylinositol 3-kinase (PI3K)/Akt/heme oxygenase (HO-1) signaling pathway through in vitro experiments. Firstly, a cell culture model of myocardiocyte hypoxia-reoxygenation (H/R) injury was replicated. After AS-IV treatment, cell viability, reactive oxygen species (ROS) levels, as well as the content or activity of the cellular factors lactate dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA), interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), were measured to evaluate the effect of treatment with AS-IV. The effect of AS-IV on HO-1 protein expression and nuclear factor E2-related factor 2 (Nrf2) and Bach1 protein expression was determined by Western blotting. Finally, a reversal of the effect of AS-IV treatment was observed following co-incubation with a PI3K inhibitor. Our results show that AS-IV has good protective effect on H/R injury and has anti-oxidative stress and anti-inflammatory effects. It can regulate the expression of Nrf2 and Bach1 proteins in the nucleus and promote the expression of HO-1 protein, while a PI3K inhibitor can partially reverse the above effects. This study suggests that the PI3K/Akt/HO-1 signaling pathway may be a key signaling pathway for the anti-IRI effect of AS-IV.
Collapse
Affiliation(s)
| | - Yuping Zhou
- The Affiliated Hospital of Medical School of Ningbo University
| | - Qing Xia
- Ningbo College of Health Sciences
| | | | | |
Collapse
|
48
|
Xing J, Xie T, Tan W, Li R, Yu C, Han X. microRNA‐183 improve myocardial damager via NF‐kb pathway: In vitro and in vivo study. J Cell Biochem 2018; 120:10145-10154. [PMID: 30548682 DOI: 10.1002/jcb.28298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 11/20/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Jie Xing
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Ting Xie
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Wei Tan
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Ruzheng Li
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Cheng Yu
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Xiaohu Han
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| |
Collapse
|
49
|
Boccellino M, Di Domenico M, Donniacuo M, Bitti G, Gritti G, Ambrosio P, Quagliuolo L, Rinaldi B. AT1-receptor blockade: Protective effects of irbesartan in cardiomyocytes under hypoxic stress. PLoS One 2018; 13:e0202297. [PMID: 30356256 PMCID: PMC6200178 DOI: 10.1371/journal.pone.0202297] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/31/2018] [Indexed: 12/21/2022] Open
Abstract
Hypoxia induces myocardial injury through the activation of inflammatory and oxidative processes. The pivotal role of the renin angiotensin system (RAS) in the pathogenesis of cardiovascular diseases has been firmly established in clinical trials and practice; in fact many experimental and clinical data have highlighted that its inhibition has a cardioprotective role. Activated RAS also stimulates inflammation directly inducing proinflammatory and oxidative gene expression. This study aimed to investigate the protective role of a pre-treatment (10 and 100 μM) with irbesartan on injury induced by 24 h of hypoxia in HL-1 cardiomyocytes; in particular, we have analyzed the natriuretic peptide (BNP) expression, a biomarker able to modulate inflammatory reaction to cardiac injury and some markers involved in oxidative stress and inflammation. Our results demonstrated that a pre-treatment with 100 μM irbesartan significantly increased SOD activity and catalase expression of 15 and 25%, respectively, compared to hypoxic cells (P<0.05). On the other hand, it was able to reduce the release of peroxynitrite and iNOS protein expression of 20 and 50% respectively (P<0.05). In addition irbesartan exerts an anti-inflammatory activity reducing Toll-like receptors (TLRs)-2 and -4 mRNA expression, TNF-alpha expression and activity (20%) and increasing the expression of the cytokine IL-17 (40%) (P<0.05 vs hypoxia). Our findings also showed that BNP induced by ischemia was significantly and in a concentration-dependent manner reduced by irbesartan. The findings of our study demonstrated that the AT1 receptor antagonist irbesartan exerts a protective role in an in vitro hypoxic condition reducing oxidative stress and inflammation.
Collapse
Affiliation(s)
- Mariarosaria Boccellino
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Marina Di Domenico
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States of America
- * E-mail:
| | - Maria Donniacuo
- Department of Experimental Medicine, Section of Pharmacology, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giuseppe Bitti
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giulia Gritti
- Department of Experimental Medicine, Section of Pharmacology, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Pasqualina Ambrosio
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Lucio Quagliuolo
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Barbara Rinaldi
- Department of Experimental Medicine, Section of Pharmacology, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
50
|
Yu W, Lv Z, Zhang L, Gao Z, Chen X, Yang X, Zhong M. Astragaloside IV reduces the hypoxia-induced injury in PC-12 cells by inhibiting expression of miR-124. Biomed Pharmacother 2018; 106:419-425. [PMID: 29990829 DOI: 10.1016/j.biopha.2018.06.127] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Astragalus membranaceus has been clinically used in cerebral ischemia treatment in China and its main component, Astragaloside IV (Ast IV) shows anti-hypoxia activity, but the underlying mechanism has not been clearly clarified. This study was aimed to investigate the effects of Ast IV on hypoxia-induced injury in PC-12 cells as well as the underlying mechanism. METHODS Relative miR-124 expression was detected by qRT-PCR. Hic-5 expression was analyzed by qRT-PCR and Western blot. To alter miR-124 and Hic-5 expressions, cells were respectively transfected with miR-124 mimic and pEX-Hic-5. Cell proliferation and apoptosis were measured by BrdU assay and Annexin V-fluorescein isothiocynate (FITC)/propidium iodide (PI) double staining method, respectively. Besides, apoptotic proteins and cell proliferation-associated factors were analyzed by Western blot. RESULTS Ast IV alleviated hypoxia-induced injury in PC-12 cells by decreasing apoptosis (P < 0.01). Ast IV inhibited up-regulation of miR-124 induced by hypoxia (P < 0.01). miR-124 mimic impaired the anti-apoptotic effect of Ast IV on PC-12 cells (P < 0.01). Hic-5 expression was significantly down-regulated in miR-124 overexpressed cells (P < 0.001) and Hic-5 overexpression activated Sp1/Survivin signaling pathway (P < 0.001). CONCLUSION Ast IV could ameliorate hypoxia-induced injury in PC-12 cells by decreasing miR-124 expression and then up-regulating Hic-5 expression.
Collapse
Affiliation(s)
- Wei Yu
- Department of Geriatrics, Shengli Oilfield Central Hospital, Dongying 257034, Shandong, China
| | - Zaigang Lv
- Department of Neurology, Shengli Oilfield Central Hospital, Dongying 257034, Shandong, China
| | - Ligong Zhang
- Department of Neurology, Shengli Oilfield Central Hospital, Dongying 257034, Shandong, China
| | - Zongen Gao
- Department of Neurology, Shengli Oilfield Central Hospital, Dongying 257034, Shandong, China
| | - Xiaohui Chen
- Department of Neurology, Shengli Oilfield Central Hospital, Dongying 257034, Shandong, China
| | - Xirui Yang
- Department of Rheumatology and Immunology, Shengli Oilfield Central Hospital, Dongying 257034, Shandong, China
| | - Mengfei Zhong
- Department of Neurology, Shengli Oilfield Central Hospital, Dongying 257034, Shandong, China.
| |
Collapse
|