1
|
Zhai J, Yan H, Liu M, Jiang C, Jin M, Xie B, Ma C, Cong B, Wen D. Decoding gelsenicine-induced neurotoxicity in mice via metabolomics and network toxicology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156753. [PMID: 40250031 DOI: 10.1016/j.phymed.2025.156753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Gelsenicine, the most toxic constituent of Gelsemium elegans Benth., is known for its diverse pharmacological activities alongside potent neurotoxicity, frequently leading to poisoning incidents following mistaken ingestion. However, its molecular mechanisms remain largely unexplored. PURPOSE This study aimed to elucidate the key mechanistic network underlying gelsenicine-induced neurotoxicity by employing a comprehensive strategy that integrated metabolomics, network toxicology, molecular docking, and experimental validation. METHODS Acute oral toxicity tests were conducted in C57BL/6J mice to assess toxic symptoms, determine the median lethal dose (LD50), and evaluate histopathological changes. Untargeted metabolomics was performed to identify differential metabolites and associated pathways in serum, hippocampus (HIP), and medulla oblongata (MO). Integration of network toxicology pinpointed core targets and pathways, which were further validated through molecular docking and RT-qPCR. A core "compound-target-metabolite-pathway" network involved in gelsenicine-induced neurotoxicity was established. RESULTS Gelsenicine exhibited an oral LD50 of approximately 1.82 mg/kg and induced neurotoxic damage in the HIP and MO. Two untargeted metabolomic approaches detected a broad range of metabolites, revealing that gelsenicine markedly altered the metabolic profiles of serum, HIP, and MO. Network toxicology analysis identified 187 key targets associated with gelsenicine neurotoxicity. Integrated analyses with the predicted targets of differential metabolites indicated that gelsenicine primarily interferes with the energy metabolism network centered on the malate-aspartate shuttle (MAS), affecting pathways such as carbon metabolism, amino acid metabolism, TCA cycle, and PPAR signaling pathway. Malate, glutamate, and aspartate were identified as core metabolites and potential biomarkers of gelsenicine poisoning. RT-qPCR validation revealed that gelsenicine interfered with the expression of core targets, including GLUD1, MDH, GOT and ME, all of which exhibited good binding energy with gelsenicine. CONCLUSION This study unveiled a novel mechanistic insight into gelsenicine-induced neurotoxicity, demonstrating its capacity to perturb multiple energy metabolism pathways associated with MAS. These findings could enhance the theoretical understanding of gelsenicine's neurotoxic effects and highlight potential applications in clinical diagnosis and forensic identification.
Collapse
Affiliation(s)
- Jinxiao Zhai
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China; College of Forensic Medicine, Jining Medical University, Jining 272067, China
| | - Hui Yan
- Department of Forensic Toxicology, Shanghai Key Laboratory of Forensic Medicine, Academy of Forensic Science, 1347 West Guangfu Road, Shanghai 200063, China
| | - Minghao Liu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Chen Jiang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Mingyang Jin
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China.
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China.
| |
Collapse
|
2
|
Yang X, Peng S, Wang P, Tao Y. Multi-omics analysis reveals classic decoction alleviates ulcerative colitis by modulating tryptophan metabolism, gut microbiota homeostasis, and mitochondrial Bax/Bcl-2 pathways. J Pharm Biomed Anal 2025; 264:116972. [PMID: 40398245 DOI: 10.1016/j.jpba.2025.116972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
Gegen Qinlian Decoction (GQD), a classic decoction documented in the Treatise on Febrile Diseases, is clinically used to treat ulcerative colitis (UC). However, the active components and underlying pharmacological mechanisms of GQD in UC remain inadequately explored. Herein, we identify the active components and elucidate the underlying pharmacological mechanisms of GQD in the treatment of UC. A mouse model of UC was induced using dextran sulfate sodium (DSS) and treated with low, medium, and high doses of GQD. The therapeutic effects were assessed by monitoring body weight, disease activity index (DAI), colon length, spleen index, pathological changes, and cytokine levels. Plasma biomarkers were analyzed using Ultra-Performance Liquid Chromatography Quadrupole-Orbitrap mass spectrometer (UPLC-Q-Orbitrap MS)-based metabolomics. Changes in gut microbiota were also examined to identify key microbes involved in the intestinal pathological shifts. Additionally, network pharmacology analysis was conducted to predict the potential pathways of GQD in UC, which were subsequently validated by western blotting and ELISA assays. GQD significantly improved UC symptoms.A total of 98 chemical constituents in GQD were identified. Of these, 25 differential metabolites were found between normal and UC mice, with GQD modulating 22 of these metabolites. Metabolic pathways related to tryptophan, arginine, and proline were normalized. Network pharmacology predicted the apoptosis pathway as a potential target for GQD in UC intervention. Further experiments confirmed that GQD reduced pro-inflammatory cytokines (TNF-α, IL-6, IL-1β) and downregulated the Bax/Bcl-2 apoptosis pathway. Moreover, gut microbiota analysis showed that GQD enhanced the diversity and abundance of beneficial bacteria. GQD alleviates UC by modulating tryptophan, arginine, and proline metabolism, maintaining gut microbiota balance, and reducing apoptosis in intestinal cells.
Collapse
Affiliation(s)
- Xujin Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, China.
| | - Shen Peng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, China.
| | - Ping Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, China.
| | - Yi Tao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, China.
| |
Collapse
|
3
|
Shao J, Zheng Y, Wang Y, Li G, Wei J, Cheng W, Li Y. Rapid classification and identification of chemical compositions of Pu-zhi-hui-ling decoction by UHPLC-Q-Orbitrap HRMS. Nat Prod Res 2025; 39:2453-2462. [PMID: 38166578 DOI: 10.1080/14786419.2023.2299302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/04/2024]
Abstract
Pu-zhi-hui-ling decoction (PZHLD) is a traditional Chinese medicine (TCM) formula for the treatment of Alzheimer's disease (AD), but its chemical composition has not been reported. In this study, we aimed to establish a mass spectrometry (MS) analysis method for rapid classification and identification of the chemical constituents in PZHLD. The sample was analysed by ultrahigh-performance liquid chromatography coupled to quadrupole Orbitrap high-resolution mass spectrometry (UHPLC-Q-Orbitrap HRMS). The chemical constituents of PZHLD were identified based on accurate MS data, fragmentation characteristics of MS/MS, and reference information described in the literature. A total of 123 chemical constituents were identified. In addition, we summarised the fragmentation pathways of the chemical constituents in PZHLD. Our finding might lay the foundation for the further pharmacodynamic study and clinical application of PZHLD.
Collapse
Affiliation(s)
- Jia Shao
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin, China
| | - Yanxue Zheng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuanyuan Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guohui Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinxia Wei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenbo Cheng
- Mass Spectrometry Application Center, Tianjin Key Laboratory of Medical Mass Spectrometry for Accurate Diagnosis, Tianjin, China
| | - Yubo Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
4
|
Ye Z, Cheng L, Xuan Y, Yu K, Li J, Gu H. Chlorogenic acid alleviates the development of severe acute pancreatitis by inhibiting NLPR3 Inflammasome activation via Nrf2/HO-1 signaling. Int Immunopharmacol 2025; 151:114335. [PMID: 39987635 DOI: 10.1016/j.intimp.2025.114335] [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: 12/28/2024] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Severe acute pancreatitis (SAP), marked by profound tissue inflammation within the pancreatic tissue, is an abrupt and intense inflammation of the pancreas. Chlorogenic acid (CGA) is one of the effective pharmacological ingredients components in JinHong Tablet (JHT). The role of CGA in protecting pancreas from severe injury in pancreatitis needs to be studied. The intervention with CGA led to a significant decline in serum amylase and lipase levels in rats with SAP, concurrently mitigating the pathological impairment within the pancreatic tissue. CGA effectively diminishes the levels of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) in SAP rats by inhibiting the activation of NF-κB and the NLRP3 inflammasome. Additionally, in AR42J cells, the application of CGA was found to reduce the inflammatory response induced by caerulein. Mechanically, CGA alleviates the inflammatory response in SAP models by activating the Nrf2/HO-1 pathway. Together, CGA reduces the inflammatory response of SAP by activating the Nrf2/HO-1 pathway, thus alleviating the development of SAP. Our results provide a basis for the treatment of SAP.
Collapse
Affiliation(s)
- Zhen Ye
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China
| | - Lin Cheng
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China
| | - Yujun Xuan
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China
| | - Kui Yu
- Department of General Surgery, Pudong Branch, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiong Li
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China.
| | - Honggang Gu
- Department of General Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725, Wanping South Road, Xuhui District, Shanghai 200032, China.
| |
Collapse
|
5
|
Chen G, Yuan H, Zhang L, Zhang J, Li K, Wang X. Pancreatic lipase immobilization on cellulose filter paper for inhibitors screening and network pharmacology study of anti-obesity mechanism. Talanta 2024; 280:126750. [PMID: 39213890 DOI: 10.1016/j.talanta.2024.126750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/08/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The discovery of pancreatic lipase (PL) inhibitors is an essential route to develop new anti-obesity drugs. In this experiment, chitosan was used to add amino groups to cellulose filter paper (CFP) and then glutaraldehyde was used to covalently combine PL with amino-modified CFP through the Schiff base reaction. Under optimal immobilization conditions, CFP immobilized PL has a wide range of pH and temperature tolerance, as well as excellent reproducibility, reusability and storage stability. Subsequently, 26 natural products (NPs) were screened by immobilized PL with black tea extract having the highest inhibition rate. Three compounds with binding effects on PL (epigallocatechin gallate, theaflavin-3-gallate and theaflavin-3,3'-digallate) were captured. Molecular docking proved that these three compounds have a strong binding affinity for PL. Fluorescence spectra further revealed that theaflavin-3,3'-digallate could statically quench the intrinsic fluorescence of pancreatic lipase. The molecular docking and thermodynamic parameters indicated that electrostatic interaction was considered as the main interaction force between PL and theaflavin-3,3'-digallate. Finally, the potential anti-obesity targets and pathways of the three compounds were discussed through network pharmacology. This study not only proposes a simple and efficient method for screening PL inhibitors, but also sheds light on the anti-obesity mechanism of active compounds in black tea.
Collapse
Affiliation(s)
- Guangxuan Chen
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Huicong Yuan
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Lumei Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Jingran Zhang
- SCIEX, Analytical Instrument Trading Co., Ltd, Beijing, 100015, China
| | - Kefeng Li
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR, China.
| | - Xu Wang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China.
| |
Collapse
|
6
|
Yang B, Wang Q, Li Y, Zhang S, Sun Y, Wei Y, Jiang Q, Huang Y. Resveratrol inhibits white adipose deposition by the ESR1-mediated PI3K/AKT signaling pathway. Cell Signal 2024; 124:111448. [PMID: 39369759 DOI: 10.1016/j.cellsig.2024.111448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Excessive adipose accumulation is the primary cause of obesity. Resveratrol (RES), a natural polyphenolic compound, has garnered significant attention for its anti-obesity properties. However, the precise mechanisms by which RES influences fat deposition have not yet been explored. In this study, the aim was to identify the target proteins and associated pathways of RES in order to elucidate the mechanisms by which RES reduces fat deposition. In this study, mice were administered 400 mg/kg of RES via gavage for 12 weeks. We found that while 400 mg/kg RES had no impact on the growth of the mice, it significantly reduced the weight of various white adipose tissues, as well as the serum and liver concentrations of total cholesterol and triglycerides. Network pharmacology identified 15 potential targets of RES and highlighted the PI3K/AKT signaling pathway as a key pathway. Molecular docking and dynamic simulations suggested that ESR1 might be the target protein through which RES exerts its anti-fat deposition effects. In vitro experiments revealed that ESR1 promotes the proliferation and inhibits the differentiation of 3 T3-L1 adipocytes, and suppresses the PI3K/AKT signaling pathway. Silencing the ESR1 gene altered the ability of RES to inhibit cell differentiation via the PI3K/AKT pathway. Gene expression results in subcutaneous adipose tissue, epididymal fat tissue, and liver tissue of mice were consistent with observations in cells. In summary, RES reduces white fat deposition by directly targeting the ESR1 protein and inhibiting the PI3K/AKT signaling pathway. Our findings provide new insights into the potential use of RES in the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Bao Yang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Qian Wang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yin Li
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Sanbao Zhang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Yanjie Sun
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yangyang Wei
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Qinyang Jiang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China.
| | - Yanna Huang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China.
| |
Collapse
|
7
|
Han X, Wu X, Liu F, Chen H, Hou H. Inhibition of LPS-induced inflammatory response in RAW264.7 cells by natural Chlorogenic acid isomers involved with AKR1B1 inhibition. Bioorg Med Chem 2024; 114:117942. [PMID: 39396466 DOI: 10.1016/j.bmc.2024.117942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
Inflammation is the physiological response of the immune system to injury or infection, typically manifested by local tissue congestion, swelling, heat, and pain. Prolonged or excessive inflammation can lead to tissue damage and the development of many diseases. The anti-inflammatory effects of natural ingredients have been extensively researched and confirmed. This study investigated the effects of Chlorogenic acid (CGA) isomers -- 3-Caffeolyquninic acid (3-CQA), 4-Caffeolyquninic acid (4-CQA), and 5-Caffeolyquninic acid (5-CQA) -- on the inflammatory response and oxidative stress reaction induced by LPS in RAW264.7 cells. Overall, 3-CQA exhibited the most significant reduction in levels of TNF-α, IL-6, NO, and ROS. 4-CQA showed superior inhibition of TNF-α compared to 5-CQA (p < 0.05), while no significant difference in other parameters. We further used DARTS and CETSA to demonstrate that CGA isomers have stable affinity with AKR1B1. As a positive control, the AKR1B1 antagonist epalrestat exhibited similar effects to the CGA isomers. 3-CQA having the smallest half-inhibitory concentration (IC50) for AKR1B1, while 4-CQA and 5-CQA have similar values. AutoDock simulations of the docking conformations revealed minimal differences in the average binding energies of the CGA isomers. The main differences were that VAL47 formed a hydrogen bond with 3-CQA, whereas GLN49 formed hydrogen bonds with 4-CQA and 5-CQA. Additionally, the number of hydrophobic bonds involving PHE122 and LEU300 varies. Our conclusion is that differences in non-covalent interactions result in the varying inhibitory abilities of CGA isomers on AKR1B1, which further affect the anti-inflammatory and antioxidant effects of CGA isomers.
Collapse
Affiliation(s)
- Xu Han
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China; China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, PR China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, PR China
| | - Xiaqing Wu
- China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, PR China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, PR China
| | - Fanglin Liu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China.
| | - Huan Chen
- China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, PR China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, PR China.
| | - Hongwei Hou
- China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, PR China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, PR China.
| |
Collapse
|
8
|
Cai J, Zhan Y, Huang K, Han S, Lin Z, Chen R, Luo Q, Li Z, Chen B, Li S. Integration of network pharmacology and proteomics analysis to identify key target pathways of Ginsenoside Re for myocardial ischemia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155728. [PMID: 38853122 DOI: 10.1016/j.phymed.2024.155728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/17/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Clinically, various diseases cause myocardial ischemia (MI), which further induces severe cardiac injury and leads to high mortality in patients. Ginsenoside Re, one of the major ginsenosides in ginseng, can regulate the level of oxidative stress in the injured myocardium. Thus, it may attenuate MI injury, but the related mechanism has not been comprehensively studied. PURPOSE This study aimed to investigate the anti-MI effect and comprehensively mechanisms of Ginsenoside Re. STUDY DESIGN/METHODS Oxygen-glucose deprivation (OGD), oxidative-induced cardiomyocyte injury, and isoproterenol-induced MI mice were used to explore their protective effect of Ginsenoside Re. An integrated approach of network pharmacology, molecular docking, and tandem mass tag proteomics was applied to determine the corresponding common potential targets of Ginsenoside Re against MI, such as target proteins and related pathways. The major anti-MI target proteins and related pathways were validated by immunofluorescence (IF) assay and Western blotting (WB). RESULTS Ginsenoside Re (1.32-168.93 µM) had low toxicity to normal cardiomyocytes, and increased the survival of oxidative stress-injured (OGD-induced injury or H2O2-induced injury) cardiomyocytes in this concentration range. It regulated the reactive oxygen species (ROS) level in OGD-injured cardiomyocytes; stabilized the nuclear morphology, mitochondrial membrane potential (MMP), and mitochondrial function; and reduced apoptosis. Meanwhile, Ginsenoside Re (5-20 mg/kg) alleviated cardiac injury in MI mice and maintained cardiac function. Through network pharmacology and proteomics, the relevant mechanisms revealed several key pathways of Ginsenoside Re anti-MI, including inhibition of MAPK pathway protein phosphorylation, downregulation of phosphorylated PDPK1, AKT, and STAT3, and upregulation of TGF-β3, ferroptosis pathway (upregulation of GPX4 and downregulation of phosphorylation level of MDM2) and AMPK pathway (regulating the synthesis of cholesterol in the myocardium by downregulation of HMGCR). The key proteins of these target pathways were validated by IF and/or WB. CONCLUSION Ginsenoside Re may target MAPK, AKT, ferroptosis pathways and AMPK pathway to prevent and/or treat MI injury and protect cardiomyocytes from oxidative damage.
Collapse
Affiliation(s)
- Jiasong Cai
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Yuying Zhan
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Kunlong Huang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Shengnan Han
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Zhan Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Ruichan Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Qiu Luo
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Zhijun Li
- Center of Chemistry Experiment, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Bing Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Shaoguang Li
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
9
|
Jiang Y, Guo N, Zhang Q, Xu X, Qiang M, Lv Y. MrgX2-targeted ligand screening from Artemisia capillaris Thunb. extract and receptor-ligand interaction analysis based on MrgX2-HALO-tag/CMC. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1244:124252. [PMID: 39067315 DOI: 10.1016/j.jchromb.2024.124252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Abstract
Artemisia capillaris Thunb. (A. capillaris) is a well-known traditional Chinese herbal medicine with a wide range of pharmacological effects, such as soothing the liver and gallbladder, heat clearance, and detoxifying. Hence, its extract is commonly added to various traditional Chinese medicine formulas. Traditional Chinese medicine injection (TCMI) is a mature pharmaceutical dosage form developed using TCM theory combined with modern science and technology. Notably, allergic reactions, especially pseudo‑allergic reactions (PARs), greatly limited the use of these injections. Therefore, screening pseudo‑allergic components in A. capillaris extract is clinically significant. In the present study, we proposed a two-dimensional screening and identification system based on mas-related G protein-coupled receptor X2-HALO-tag/cell membrane chromatography (MrgX2-HALO-tag/CMC) high performance liquid chromatography mass spectrometry (HPLC-MS); seven potential active components were screened from 75 % ethanol extract of A. capillaris: NCA, CA, CCA, 1,3-diCQA, ICA-B, ICA-A, and ICA-C. The receptor-ligand interactions between these seven compounds and MrgX2 protein were analyzed using frontal analysis and molecular docking technology. Furthermore, a mast cell degranulation-related assay was used to assess the pseudo‑allergic activity of these compounds. The screened compounds can serve as ligands of MrgX2, and this study provides a research basis for pseudo‑allergic reactions caused by TCMIs containing A. capillaris.
Collapse
Affiliation(s)
- Yuhan Jiang
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, China; Institute of Pharmaceutical Science and Technology, Western China Science & Technology Innovation Harbour, Xi'an 710115, China
| | - Na Guo
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, China; Institute of Pharmaceutical Science and Technology, Western China Science & Technology Innovation Harbour, Xi'an 710115, China
| | - Quan Zhang
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, China; Institute of Pharmaceutical Science and Technology, Western China Science & Technology Innovation Harbour, Xi'an 710115, China
| | - Xiaochan Xu
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, China
| | - Mengyang Qiang
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, China
| | - Yanni Lv
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, China; Institute of Pharmaceutical Science and Technology, Western China Science & Technology Innovation Harbour, Xi'an 710115, China.
| |
Collapse
|
10
|
Lv Y, Li H, Zhai BT, Sun J, Cheng JX, Zhang XF, Guo DY. Evidence of synergistic mechanisms of hepatoprotective botanical herbal preparation of Pueraria montana var. lobata and Schisandra sphenanthera. Front Pharmacol 2024; 15:1412816. [PMID: 38978983 PMCID: PMC11228302 DOI: 10.3389/fphar.2024.1412816] [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: 04/05/2024] [Accepted: 06/03/2024] [Indexed: 07/10/2024] Open
Abstract
Background Pueraria montana var. lobata (Willd.) Maesen & S.M.Almeida ex Sanjappa & Predeep (syn. Pueraria lobata (Willd.) Ohwi) and Schisandra sphenanthera Rehder & E.H. Wilson are traditional edible and medicinal hepatoprotective botanical drugs. Studies have shown that the combination of two botanical drugs enhanced the effects of treating acute liver injury (ALI), but the synergistic effect and its action mechanisms remain unclear. This study aimed to investigate the synergistic effect and its mechanism of the combination of Pueraria montana var. lobata (Willd.) Maesen & S.M.Almeida ex Sanjappa & Predeep (syn. Pueraria lobata (Willd.) Ohwi) (PM) and Schisandra sphenanthera Rehder & E.H. Wilson (SS) in the treatment of ALI. Methods High performance liquid chromatography (HPLC) were utilized to conduct the chemical interaction analysis. Then the synergistic effects of botanical hybrid preparation of PM-SS (BHP PM-SS) against ALI were comprehensively evaluated by the CCl4 induced ALI mice model. Afterwards, symptom-oriented network pharmacology, transcriptomics and metabolomics were applied to reveal the underlying mechanism of action. Finally, the key target genes were experimentally by RT-qPCR. Results Chemical analysis and pharmacodynamic experiments revealed that BHP PM-SS was superior to the single botanical drug, especially at 2:3 ratio, with a better dissolution rate of active ingredients and synergistic anti-ALI effect. Integrated symptom-oriented network pharmacology combined with transcriptomics and metabolomics analyses showed that the active ingredients of BHP PM-SS could regulate Glutathione metabolism, Pyrimidine metabolism, Arginine biosynthesis and Amino acid sugar and nucleotide sugar metabolism, by acting on the targets of AKT1, TNF, EGFR, JUN, HSP90AA1 and STAT3, which could be responsible for the PI3K-AKT signaling pathway, MAPK signaling pathway and Pathway in cancer to against ALI. Conclusion Our study has provided compelling evidence for the synergistic effect and its mechanism of the combination of BHP PM-SS, and has contributed to the development and utilization of BHP PM-SS dietary supplements.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dong-Yan Guo
- State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, China
| |
Collapse
|
11
|
Lu S, Huang J, Zhang J, Wu C, Huang Z, Tao X, You L, Stalin A, Chen M, Li J, Tan Y, Wu Z, Geng L, Li Z, Fan Q, Liu P, Lin Y, Zhao C, Wu J. The anti-hepatocellular carcinoma effect of Aidi injection was related to the synergistic action of cantharidin, formononetin, and isofraxidin through BIRC5, FEN1, and EGFR. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117209. [PMID: 37757991 DOI: 10.1016/j.jep.2023.117209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aidi injection (ADI) is a popular anti-tumor Chinese patent medicine, widely used in clinics for the treatment of hepatocellular carcinoma (HCC) with remarkable therapeutic effects through multiple targets and pathways. However, the scientific evidence of the synergistic role of the complex chemical component system and the potential mechanism for treating diseases are ignored and remain to be elucidated. AIM OF THE STUDY This study aimed to elucidate and verify the cooperative association between the potential active ingredient of ADI, which is of significance to enlarge our understanding of its anti-HCC molecular mechanisms. MATERIALS AND METHODS Firstly, the anti-HCC effect of ADI was evaluated in various HCC cells and the zebrafish xenograft model. Subsequently, a variety of bioinformatic technologies, including network pharmacology, weighted gene co-expression network analysis (WGCNA), meta-analysis of gene expression profiles, and pathway enrichment analysis were performed to construct the competitive endogenous RNA (ceRNA) network of ADI intervention in HCC and to establish the relationship between the critical targets/pathways and the key corresponding components, which were involved in ADI against HCC in a synergistic way and were validated by molecular biology experiments. RESULTS ADI exerted remarkable anti-HCC in vitro cells and in vivo zebrafish model, especially that the Hep 3B2.1-7 cell showed substantial sensibility to ADI. The ceRNA network revealed that the EGFR/PI3K/AKT signaling pathway was identified as the promising pathway. Furthermore, the meta-analysis also demonstrated the critical role of BIRC5 and FEN1 as key targets. Finally, the synergistic effect of ADI was revealed by discovering the inhibitory effect of cantharidin on BIRC5, formononetin on FEN1 and EGFR, as well as isofraxidin on EGFR. CONCLUSION Our study unveiled that the incredible protective effect of ADI on HCC resulted from the synergistic inhibition effect of cantharidin, formononetin, and isofraxidin on multiple targets/pathways, including BIRC5, FEN1, and EGFR/PI3K/AKT, respectively, providing a scientific interpretation of ADI against HCC and a typical example of pharmacodynamic evaluation of other proprietary Chinese patent medicine.
Collapse
Affiliation(s)
- Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jiaqi Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Xiaoyu Tao
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Leiming You
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Meilin Chen
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jiaqi Li
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Libo Geng
- Guizhou Yibai Pharmaceutical Co. Ltd, Guiyang, 550008, Guizhou, China.
| | - Zhiqi Li
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Qiqi Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Pengyun Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Yifan Lin
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Chongjun Zhao
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
12
|
Liu Y, Xiang R, Lu W, Qin X. Symptom-oriented network pharmacology revealed the mechanism of HuangQi-DanShen herb pair against cerebral ischemia coupled with comprehensive chemical characterization. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116845. [PMID: 37437791 DOI: 10.1016/j.jep.2023.116845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In the clinical practice of traditional Chinese medicine, HuangQi-DanShen (HD) is an important drug pair for the treatment of cerebral ischemia (CI). AIM OF THE STUDY Elucidate the mechanism of HD against CI based on symptom-oriented network pharmacology coupled with comprehensive chemical characterization. MATERIALS AND METHODS UHPLC-Q-Exactive Orbitrap-MS technology was firstly used to obtain the chemical profile of HD constituents. A comprehensive strategy combining in-house library, diagnostic ions, Compound Discover software and network databases was then established to identify its chemical constitutes. Symptomatic treatment is a treatment aimed at relieving or eliminating symptoms which is often characterized as a stop-gap measure due to its inability to cure the disease fundamentally. Nevertheless, symptomatic treatment is an indispensable part of clinical practice and has an important place in medical therapeutics. Therefore, network pharmacology technique were used to elucidate molecular mechanisms from the symptoms of CI. Finally, some literatures were further mined to support our conclusions. RESULTS A total of 190 ingredients were identified in HD. Symptom-oriented network pharmacology analysis indicated that compounds of HD relieved "blood" through the regulation of ADORA2A, ADORA1, PTPN11, MMP9 and EGFR, relieved "qi" via the regulation of ADORA2A, EGFR, MMP9 and CA2. The therapeutic effect of HD on "faint" was linked to PTPN11 and MMP9, while the regulation of "dyskinesia" was related to ADORA2A and EGFR, and ADORA1, PTPN11 and MMP9 were associated withe its effect on "speech disorder". ADORA1, ADORA2A and MMP9 were key to the HD component in treating "visual disturbance". CONCLUSION The approach of symptom-oriented network pharmacology coupled with comprehensive chemical characterization proposed a further orientation for exploring the mechanisms of HD against CI.
Collapse
Affiliation(s)
- Yuetao Liu
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| | - Ruoxin Xiang
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| | - Wentian Lu
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| |
Collapse
|
13
|
Wang Y, Li X, Qi M, Li X, Zhang F, Wang Y, Wu J, Shu L, Fan S, Li Y, Li Y. Pharmacological effects and mechanisms of YiYiFuZi powder in chronic heart disease revealed by metabolomics and network pharmacology. Front Mol Biosci 2023; 10:1203208. [PMID: 37426419 PMCID: PMC10327484 DOI: 10.3389/fmolb.2023.1203208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction: YiYiFuZi powder (YYFZ) is a classical formula in Chinese medicine, which is commonly used clinically for the treatment of Chronic Heart Disease (CHD), but it's pharmacological effects and mechanism of action are currently unclear. Methods: An adriamycin-induced CHD model rat was established to evaluate the pharmacological effects of YYFZ on CHD by the results of inflammatory factor level, histopathology and echocardiography. Metabolomic studies were performed on rat plasma using UPLC-Q-TOF/MS to screen biomarkers and enrich metabolic pathways; network pharmacology analysis was also performed to obtain the potential targets and pathways of YYFZ for the treatment of CHD. Results: The results showed that YYFZ significantly reduced the levels of TNF-α and BNP in the serum of rats, alleviated the disorder of cardiomyocyte arrangement and inflammatory cell infiltration, and improved the cardiac function of rats with CHD. The metabolomic analysis identified a total of 19 metabolites, related to amino acid metabolism, fatty acid metabolism, and other metabolic pathways. Network pharmacology showed that YYFZ acts through PI3K/Akt signaling pathway, MAPK signaling pathway and Ras signaling pathway. Discussion: YYFZ treatment of CHD modulates blood metabolic pattern and several protein phosphorylation cascades but importance specific changes for therapeutic effect require further studies.
Collapse
Affiliation(s)
- Yuming Wang
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xue Li
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Min Qi
- TIPRHUYA Advancing Innovative Medicines Ltd., Tianjin, China
| | - Xiaokai Li
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fangfang Zhang
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuyu Wang
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junke Wu
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lexin Shu
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Simiao Fan
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunfei Li
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yubo Li
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
14
|
Yang L, Liu X, Zhu J, Zhang X, Li Y, Chen J, Liu H. Progress in traditional Chinese medicine against chronic gastritis: From chronic non-atrophic gastritis to gastric precancerous lesions. Heliyon 2023; 9:e16764. [PMID: 37313135 PMCID: PMC10258419 DOI: 10.1016/j.heliyon.2023.e16764] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/13/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023] Open
Abstract
Chronic gastritis (CG) is a persistent inflammation of the gastric mucosa that can cause uncomfortable symptoms in patients. Traditional Chinese medicine (TCM) has been widely used to treat CG due to its precise efficacy, minimal side effects, and holistic approach. Clinical studies have confirmed the effectiveness of TCM in treating CG, although the mechanisms underlying this treatment have not yet been fully elucidated. In this review, we summarized the clinical research and mechanisms of TCM used to treat CG. Studies have shown that TCM mechanisms for CG treatment include H. pylori eradication, anti-inflammatory effects, immune modulation, regulation of gastric mucosal cell proliferation, apoptosis, and autophagy levels.
Collapse
Affiliation(s)
- Liangjun Yang
- Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Xinying Liu
- Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Jiajie Zhu
- Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Xi Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ya Li
- Lin ‘an Hospital of Traditional Chinese Medicine, Hangzhou 311300, China
| | - Jiabing Chen
- Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Haiyan Liu
- Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| |
Collapse
|
15
|
Comprehensive characterization of the chemical composition of Lurong dabu decoction and its absorbed prototypes and metabolites in rat plasma using UHPLC–Q Exactive Orbitrap–HRMS. Food Res Int 2022; 161:111852. [DOI: 10.1016/j.foodres.2022.111852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/12/2022] [Accepted: 08/21/2022] [Indexed: 11/23/2022]
|
16
|
Yan P, Wei Y, Wang M, Tao J, Ouyang H, Du Z, Li S, Jiang H. Network pharmacology combined with metabolomics and lipidomics to reveal the hypolipidemic mechanism of Alismatis rhizoma in hyperlipidemic mice. Food Funct 2022; 13:4714-4733. [PMID: 35383784 DOI: 10.1039/d1fo04386b] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Alismatis rhizoma (AR), the dried rhizome of Alisma orientale (Sam) Juzep, is effective in treating hyperlipidemia, but the mechanisms involved require further exploration. This study evaluated the hypolipidemic properties of AR using an integrated strategy combining network pharmacology with metabolomics and lipidomics. Firstly, a hyperlipidemia mouse model induced by a high-fat diet was established to evaluate the therapeutic effects of AR. Secondly, plasma metabolomics and lipidomics were used to identify differential metabolites and lipids, and metabolic pathway analysis was performed using MetaboAnalyst. Thirdly, network pharmacology, based on the metabolic profile of AR in vivo, was used to discover potential therapeutic targets. Finally, key targets were obtained through a compound-target-metabolite network, which was verified by molecular docking and quantitative real-time PCR (qPCR). Biochemistry analysis and histological examinations showed that AR exerted hypolipidemic effects on hyperlipidemic mice. Seventy potential biomarkers for the AR treatment of hyperlipidemia were identified by metabolomics and lipidomics, which were mainly involved in lipid metabolism, energy metabolism and amino acid metabolism. Eighteen potentially active compounds were identified in the plasma of mice after oral administration of AR, which were associated with 83 potential therapeutic targets. The PPAR signaling pathway was considered a crucial signaling pathway of AR against hyperlipidemia by KEGG analysis. The joint analysis showed that 6 upstream key targets were regulated by AR, including ALB, TNF, IL1B, MMP9, PPARA and PPARG. Molecular docking showed that active compounds of AR had high binding affinity with these key targets. qPCR further demonstrated that AR could reverse the mRNA expression of these key targets in hyperlipidemic mice. This study integrates network pharmacology with metabolomics and lipidomics to reveal the regulatory effects of AR on endogenous metabolites and validates key therapeutic targets, and represents the most systematic and in-depth study on the hypolipidemic activity of AR.
Collapse
Affiliation(s)
- Pan Yan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yinyu Wei
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Meiqin Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jianmei Tao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hui Ouyang
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330000, China
| | - Zhifeng Du
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Sen Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hongliang Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|