1
|
Liu H, Wei X, He Y, Pan S, Wang C, Cheng J, Zhao Q, Shi K, Si H. Elucidating the antiviral effects of a novel compound throat anti-viral through metabolomics and network pharmacology: A study on infectious bronchitis virus in poultry. Poult Sci 2025; 104:104956. [PMID: 40127563 PMCID: PMC11984592 DOI: 10.1016/j.psj.2025.104956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/09/2025] [Accepted: 02/26/2025] [Indexed: 03/26/2025] Open
Abstract
Infectious bronchitis virus (IBV) is a major pathogen that causes significant economic losses in the global poultry industry. Current vaccination strategies provide only partial protection, highlighting the need for more effective prevention and treatment methods. This study aimed to develop a novel compound throat anti-viral (CTA) from natural plants using data from the Traditional Chinese Medicine Inheritance System and identification through liquid chromatography-mass spectrometry. CTA demonstrated substantial anti-IBV effects both in vitro and in vivo studies. In vitro, CTA significantly inhibited IBV multiplication and alleviated the pathological lesions in chicken embryonic kidney cells, tracheal rings, and chicken embryos. In vivo, a seven-day treatment with CTA obtained much milder clinical signs, enhanced growth performance, and better immune organ indices in infected chickens. Additionally, CTA treatment reduced IBV levels in the trachea and lungs and increased specific antibody titers. CTA also maintained body homeostasis, exhibiting strong antioxidant and anti-inflammatory properties that mitigated respiratory tract damage. Metabolomics and network pharmacology analyses, revealed that CTA's antiviral effects are mediated through the FoxO signaling pathway. This study successfully developed an effective prescription database based on the Traditional Chinese Medicine Inheritance System and validated the antiviral efficacy of CTA through comprehensive in vitro and in vivo experiments. The findings elucidated the mechanisms of CTA's action, particularly through the FoxO signaling pathway, and highlighted its potential for clinical application as a novel antiviral treatment for IBV in the poultry industry.
Collapse
Affiliation(s)
- Huixin Liu
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Xiaofang Wei
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Yang He
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Sijia Pan
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Chenchen Wang
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Junze Cheng
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Qiyuan Zhao
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Kaichuang Shi
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China; Guangxi Center for Animal Disease Control and Prevention, Nanning 530001, China
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China.
| |
Collapse
|
2
|
Mu J, Li Y, Chen Q, Xiao Y, Hu M, He Z, Zeng J, Ding Y, Song P, He X, Yang X, Zhang X. Revealing the molecular mechanism of baohuoside I for the treatment of breast cancer based on network pharmacology and molecular docking. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118918. [PMID: 39396715 DOI: 10.1016/j.jep.2024.118918] [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: 05/07/2024] [Revised: 09/13/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Traditional Chinese Medicine (TCM), there are many prescriptions for treating breast cancer (BC) that utilize the herb Epimedium brevicornum Maxim, which warms and replenishes kidney yang. Baohuoside I (BI) is a flavonoid compound found in Epimedium brevicornum Maxim. As a single glycoside, it is not easily hydrolyzed in the intestine and is typically absorbed as a precursor. As a natural product with potential anti-cancer properties, studies have shown that BI possesses anti-cancer activity and can inhibit the invasion and migration of BC cells. However, its underlying mechanisms remain unclear, thus further research is needed to validate its modern mechanisms for traditional uses. AIM OF THE STUDY This study aimed to explore the regulatory mechanism of BI in the signaling pathways of BC cells through network pharmacology (NP), molecular docking (MD) techniques and cellular experiments. METHODS Potential targets were predicted using public databases, and a protein-protein interaction (PPI) network was constructed. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed. Key signaling pathways were validated through MD techniques, cellular experiments, RNA interference and Western blot (WB) analysis. RESULTS Treatment-associated targets included SRC, MAPK1, HSP90AA1, PIK3CA, TP53, AKT1, and EGFR. GO enrichment, KEGG enrichment analyses, and MD results indicated that BI exerts its anti-breast cancer effects by inhibiting the tyrosine kinase activity of EGFR, as well as through downstream MAPK signaling pathway and PI3K-Akt signaling pathway pathways. In vitro experiments confirmed that BI primarily induce cell apoptosis through the EGFR-mediated MAPK signaling pathway and PI3K-Akt signaling pathway. CONCLUSION BI can inhibit EGFR activation and promote BC cell apoptosis through the MAPK signaling pathway and PI3K-Akt signaling pathway, thereby exerting therapeutic effects on BC. This study not only provides experimental evidence for the accuracy of NP but also offers an effective approach for rational utilization of Baohuoside I-like flavonoid compounds as anti-breast cancer drugs.
Collapse
Affiliation(s)
- Junjie Mu
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Ying Li
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Qiuxiong Chen
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Yujie Xiao
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Min Hu
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Ziyue He
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Jun Zeng
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Yiling Ding
- Pengshui County Forestry Bureau, Chongqing, 409600, PR China
| | - Pengyang Song
- Wansheng Economic and Technological Development Zone Planning and Natural Resources Bureau, Chongqing, 400800, PR China
| | - Xiao He
- Chongqing Three Gorges Medical College, Chongqing, 404120, PR China
| | - Xian Yang
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China.
| | - Xue Zhang
- Chongqing Medical and Pharmaceutical College, Chongqing, 401331, PR China.
| |
Collapse
|
3
|
Jiang S, Zhang H, Song Y, Xiao M, Hu H, Yu S, Xie F. Metabolic profiles and potential antioxidant mechanisms of hawk tea. Sci Rep 2025; 15:3600. [PMID: 39875806 PMCID: PMC11775316 DOI: 10.1038/s41598-025-88160-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/24/2025] [Indexed: 01/30/2025] Open
Abstract
Hawk tea has received increasing attention for its unique flavor and potential health benefits, with antioxidant function being one of its significant bioactivities. However, the metabolic profiles, potential antioxidant components, and action mechanisms of different types of hawk tea are still unclear. In this study, the chemical components of five hawk teas were determined using untargeted metabolomics. Then, the potential antioxidant metabolites and their possible action mechanisms were revealed by integrating network pharmacology and molecular docking. The results showed that the metabolic profiles of various hawk teas differed significantly, but the content of flavonoids was the highest in each group. Network pharmacology analyses suggested that 11 potential antioxidant metabolites-four of which were the same metabolites with high levels in the five types, and seven were differential metabolites-could be involved in several metabolic pathways in vivo. These pathways included the MAPK and PI3K/AKT signaling pathways, which may be closely related to antioxidant activity. Finally, molecular docking revealed potential antioxidant metabolites bound to 25 core antioxidant targets through hydrogen bonding and hydrophobic interactions. Among them, artemisinin, astragalin, isoquercetrin, isoquercitrin, kaempferol-3-glucuronide, and UDP-L-rhamnose exhibited low binding energies to core antioxidant targets such as AKT1, RELA, and MTOR, forming stable conformation. These insights lay the basis for further elucidating the antioxidant mechanism of hawk tea.
Collapse
Affiliation(s)
- Sixia Jiang
- Department of Food Science and Engineering, Moutai Institute, Renhuai, 564502, People's Republic of China
| | - Hong Zhang
- Department of Food Science and Engineering, Moutai Institute, Renhuai, 564502, People's Republic of China
| | - Ya Song
- Department of Food Science and Engineering, Moutai Institute, Renhuai, 564502, People's Republic of China
| | - Mingji Xiao
- Institute of Biology, Guizhou Academy of Sciences, Guiyang, 50009, Guizhou, People's Republic of China
| | - Hao Hu
- Department of Food Science and Engineering, Moutai Institute, Renhuai, 564502, People's Republic of China
| | - Shirui Yu
- Department of Food Science and Engineering, Moutai Institute, Renhuai, 564502, People's Republic of China.
| | - Feng Xie
- Department of Food Science and Engineering, Moutai Institute, Renhuai, 564502, People's Republic of China.
| |
Collapse
|
4
|
Zhou J, Hu J, Liu J, Zhang W. Elucidating the gastroprotective mechanisms of Imperata cylindrica Beauv.var. major (Nees) C.E.Hubb through UHPLC-MS/MS and systems network pharmacology. Sci Rep 2024; 14:27815. [PMID: 39537788 PMCID: PMC11560922 DOI: 10.1038/s41598-024-79483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Imperata cylindrica Beauv.var. major (Nees) C.E.Hubb., commonly known as BaiMaoGen (BMG), a medicinal and edible traditional Chinese medicinal (TCM) herb commonly used in health supplements, has been observed to offer protective effects against gastrointestinal disorders. However, the specific bioactive compounds and their molecular mechanisms have not been fully elucidated. This study employed ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) and systematic network pharmacology to analyze and identify the key active components and their interactions with biological targets. Thirty-six main active compounds, including 3,4-dihydroxybenzoic acid and p-hydroxybenzoic acid, were identified and analyzed for their interaction with key protein targets using molecular docking and dynamic simulations. This combined approach highlighted the therapeutic pathways involved, particularly the PI3K/AKT signaling pathways, providing new insights into the molecular basis of BMG's gastroprotective effects. Our findings suggested that BMG's complex multi-target action can potentially be harnessed to develop effective treatments for gastrointestinal toxicity.
Collapse
Affiliation(s)
- Jiaxin Zhou
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Jianping Hu
- Academician Workstation, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Jiancheng Liu
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Wenchun Zhang
- School of life science, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China.
| |
Collapse
|
5
|
Li W, Zhang Y, Wang Q, Wang Y, Fan Y, Shang E, Jiang S, Duan J. 6-Gingerol ameliorates ulcerative colitis by inhibiting ferroptosis based on the integrative analysis of plasma metabolomics and network pharmacology. Food Funct 2024; 15:6054-6067. [PMID: 38753306 DOI: 10.1039/d4fo00952e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
6-Gingerol (6-G), an active ingredient of ginger with anti-inflammation and anti-oxidation properties, can treat ulcerative colitis (UC). However, its underlying mechanism is still unclear. In this study, the pharmacodynamic evaluation of 6-G for treating UC was performed, and the mechanism of 6-G in ameliorating UC was excavated by plasma metabolomics and network pharmacology analysis, which was further validated by experimental and molecular docking. The results showed that 6-G could notably reduce diarrhea, weight loss, colonic pathological damage, and inflammation in UC mice. Plasma metabolomic results indicated that 6-G could regulate 19 differential metabolites, and its metabolic pathways mainly involved linoleic acid metabolism and arachidonic acid metabolism, which were closely associated with ferroptosis. Moreover, 60 potential targets for 6-G intervention on ferroptosis in UC were identified by network pharmacology, and enrichment analysis revealed that 6-G suppressed ferroptosis by modulating lipid peroxidation. Besides, the integration of metabolomics and network pharmacology showed that the regulation of 6-G on ferroptosis focused on 3 key targets, including ALOX5, ALOX15, and PTGS2. Further investigation indicated that 6-G significantly inhibited ferroptosis by decreasing iron load and malondialdehyde (MDA), and enhanced antioxidant capacity by reducing the content of glutathione disulfide (GSSG) and increasing the levels of superoxide dismutase (SOD) and glutathione (GSH) in UC mice and RSL3-induced Caco-2 cells. Furthermore, molecular docking showed the high affinity of 6-G with the identified 3 key targets. Collectively, this study elucidated the potential of 6-G in ameliorating UC by inhibiting ferroptosis. The integrated strategy also provided a theoretical basis for 6-G in treating UC.
Collapse
Affiliation(s)
- Wenwen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Quyi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Yu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Yuwen Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Erxin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| |
Collapse
|
6
|
Xie X, Fu J, Gou W, Qin Y, Wang D, Huang Z, Wang L, Li X. Potential mechanism of tea for treating osteoporosis, osteoarthritis, and rheumatoid arthritis. Front Med (Lausanne) 2024; 11:1289777. [PMID: 38420363 PMCID: PMC10899483 DOI: 10.3389/fmed.2024.1289777] [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: 09/06/2023] [Accepted: 01/08/2024] [Indexed: 03/02/2024] Open
Abstract
Osteoporosis (OP), osteoarthritis (OA), and rheumatoid arthritis (RA) are common bone and joint diseases with a high incidence and long duration. Thus, these conditions can affect the lives of middle-aged and elderly people. Tea drinking is a traditional lifestyle in China, and the long-term intake of tea and its active ingredients is beneficial to human health. However, the mechanisms of action of tea and its active ingredients against OP, OA, and RA are not completely elucidated. This study aimed to assess the therapeutic role and related mechanisms of tea and its active ingredients in OP, OA, and RA. Moreover, it expanded the potential mechanisms of tea efficacy based on network pharmacology and molecular docking. Results showed that tea has potential anti-COX properties and hormone-like effects. Compared with a single component, different tea components synergize or antagonize each other, thereby resulting in a more evident dual effect. In conclusion, tea has great potential in the medical and healthcare fields. Nevertheless, further research on the composition, proportion, and synergistic mechanism of several tea components should be performed.
Collapse
Affiliation(s)
- Xinyu Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiehui Fu
- Department of Sports Medicine (Orthopedics), Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou, China
| | - Weiying Gou
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yifei Qin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Dingzhen Wang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zuer Huang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lili Wang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xihai Li
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|