1
|
Gu H, Zhang Y, Sun J, Liu L, Liu Z. Exploring the effect and mechanism of action of Jinlida granules (JLD) in the treatment of diabetes-associated cognitive impairment based on network pharmacology with experimental validation. Ann Med 2025; 57:2445181. [PMID: 39723533 DOI: 10.1080/07853890.2024.2445181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/19/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES To explore the effect and the probable mechanisms of JLD in the treatment of type 2 diabetes mellitus (T2DM) - associated cognitive impairment (TDACI). METHODS The effect of JLD in combating TDACI was assessed in T2DM model mice by conducting Morris water maze (MWM) behaviour testing. Active components and their putative targets, as well as TDACI-related targets, were collected from public databases. Protein-protein interactions (PPIs), Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses and molecular docking were then utilized to explore potential molecular network mechanisms. Finally, the main targets were verified in animal model experiments. RESULTS MWM test showed that JLD improved aspects of behaviour in T2DM model mice. JLD improved glucose intolerance, tissue insulin sensitivity, lipid metabolism and enhanced synapse-associated protein expression in hippocampus tissue. Network pharmacology revealed 185 active components, 337 targets of JLD, and 7998 TDACI related targets were obtained . PPI network analyses revealed 39 core targets. GO and KEGG analyses suggested that JLD might improve TDACI by regulating gene expression, apoptotic processes and inflammatory responses mainly via PI3K-AKT and AGE-RAGE signaling pathways. Molecular docking revealed strong binding of the main components to core targets. JLD reduced hippocampus tissue expression of the inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL6), core targets of treatment of TDACI. CONCLUSIONS The findings suggested that JLD has the potential to improve TDACI through multiple components, multiple targets and multiple pathways. JLD may be a promising treatment for diabetic cognitive impairment.
Collapse
Affiliation(s)
- Haiyan Gu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Yuxin Zhang
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Jinghua Sun
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Lipeng Liu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Zanchao Liu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| |
Collapse
|
2
|
Prajapati MK, Mittal A, Panda P. Phytoflavonoids as alternative therapeutic effect for melanoma: Integrative Network pharmacology, molecular dynamics and drug-likeness profiling for lead discovery. Comput Biol Chem 2025; 117:108390. [PMID: 40056707 DOI: 10.1016/j.compbiolchem.2025.108390] [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: 12/23/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 03/10/2025]
Abstract
Melanoma, an aggressive skin cancer, poses significant therapeutic challenges due to its resistance to conventional therapies and high metastatic potential. From this perspective, phytoflavonoids from different medicinal and aromatic plants gained attention due to their diverse multimodal anticancer effects with higher antioxidant and anti-inflammatory properties. This study explores phytoflavonoid potency against melanoma via a computer-aided drug design (CADD) platform. Using the core moiety of flavonoids (flavan), four most putative targets, such as cyclin-dependent kinases 1 and 5 (CDK1, CDK5), cell division cycles 25B and 225 C (CDC25B, and CDC225C), have been identified through a network pharmacology approach using TNMplot datasets (GenChip and RNA sequence). Further, 44 phytoflavonoids were selected from extensive literature, and molecular docking studies were carried out against four targets along with standard drugs using AutoDock 4.2 software. Subsequently, physicochemical, toxicity, pharmacokinetics, and drug-ability profiles of phytoflavonoids were predicted. Based on potency and drug-ability, we have selected 'CDK1-naringenin' with the standard drug complex, 'CDK1-dinaciclib,' for molecular dynamic simulation at 100 nanoseconds using GROMACS 2020 software. Based on potency (average docking score: 8.35 kcal/mol.), physicochemical properties (obeyed Lipinski rule of five), toxicity (class-IV), fifty percent lethal dose (2000 mg/kg), bioavailability (0.55), drug-likeness score (0.82), along with ideal pharmacokinetics profiles and higher protein-ligand stability, naringenin is considered as a potential and non-toxic anticancer candidate to be used for melanoma as alternative or complementary agent. The integrative and systematic analyses not only highlight the potential of phytoflavonoids but also select the potential lead from the library within limited resources to accelerate the current anticancer drug discovery process.
Collapse
Affiliation(s)
- Manoj Kumar Prajapati
- NIMS Institute of Pharmacy, NIMS University, Jaipur, Rajasthan 303121, India; Kashi Institute of Pharmacy, Mirzamurad, Varanasi, Uttar Pradesh 221307, India.
| | - Abhilasha Mittal
- NIMS Institute of Pharmacy, NIMS University, Jaipur, Rajasthan 303121, India
| | - Pritipadma Panda
- School of Pharmacy, Kalinga Institute of Industrial Technology Deemed to be University, Patia, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
3
|
Zhang B, Chen L, Kang M, Ai L, Tao Y. Gegen Qinlian Decoction improves Alzheimer's disease through TLR4/NF-κB/NLRP3 pathway. Tissue Cell 2025; 95:102818. [PMID: 40056656 DOI: 10.1016/j.tice.2025.102818] [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: 07/30/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
OBJECTIVE Alzheimer's disease (AD) is a neurodegenerative disease that leads to dementia, but effective treatments are lacking. This study aims to evaluate the therapeutic effects of Gegen Qinlian Decoction (GGQLD) on AD and investigate the underlying mechanisms. METHODS Using network pharmacology and bioinformatics, we identified 376 active ingredients of GGQLD and 427 drug targets. Among these, 7 potential targets (CASP1, MKI67, NFKB1, TLR4, NLRP3, IL1B, and AKT1) were identified as intersecting targets of both GGQLD and AD. Functional enrichment analysis revealed that GGQLD regulates pyroptosis-related pathways. In vivo, GGQLD was administered to AD rat models to assess its effects on spatial learning, memory, and brain tissue injury. RESULTS GGQLD significantly reduced latency time by 40 % and increased platform crossings by 60 % in AD rats, demonstrating improved spatial learning and memory abilities. It also reduced hippocampal tissue damage and abnormal Aβ deposition. Mechanistically, GGQLD downregulated pyroptosis-related targets (TLR4, NF-κB, NLRP3, IL-1β, and Caspase-1), which were significantly upregulated in AD. ROC analysis demonstrated strong diagnostic significance for these genes, with AUC values exceeding 0.70. Functional enrichment and KEGG analysis further indicated that GGQLD exerts its therapeutic effects through multiple pathways, particularly the NOD-like receptor pathway, Necroptosis, and NF-kappa B pathway. CONCLUSIONS This study demonstrates that GGQLD improves spatial learning, reduces brain tissue damage, and alleviates inflammation in AD through the regulation of pyroptosis-related pathways, providing evidence for its potential as a therapeutic agent for AD.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Liudan Chen
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Mengru Kang
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Liang Ai
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Yangu Tao
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China.
| |
Collapse
|
4
|
Wang P, Ma Y, Rao X, Luo Q, Xiao X, Wang T, Long F. Kaempferol targets Src to exert its chemopreventive effects on mammary tumorigenesis via regulation of the PI3K/AKT pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156701. [PMID: 40220416 DOI: 10.1016/j.phymed.2025.156701] [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: 09/20/2024] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Breast cancer (BC) is a prevalent malignancy that poses significant risks to the health of women worldwide. The incidence and mortality rates of BC continue to be high, despite improvements in diagnosis and treatment, indicating a need for novel prevention strategies. Kaempferol (KAM) is a common dietary flavonoid with known antitumour properties, but its role in the chemoprevention of BC and the underlying mechanisms largely unexplored. PURPOSE This study aimed to evaluate the chemopreventive effects of KAM on carcinogen-induced BC in vivo and in vitro and to elucidate the underlying molecular mechanisms. METHODS In this study, we used an N-methyl-N-nitrosourea (NMU)-induced rat model of BC and 17β-oestradiol (E2)-treated MCF-10A cells to evaluate the chemopreventive effects of KAM on mammary tumorigenesis. The antioxidant capacity of KAM was assessed by measuring oxidative damage marker levels and antioxidant enzyme expression. Flow cytometry and Hoechst 33258 staining were utilized to analyse cell cycle distribution and apoptosis. The core target of KAM was identified by network pharmacology and validated by molecular docking, MD simulation, CESTA, and BLI. KEGG enrichment analysis, molecular biology tests and the application of specific protein inhibitors were conducted to elucidate the molecular mechanisms modulated by KAM. RESULTS In vivo, KAM inhibited the progression of mammary tumours and delayed pathological changes in the morphological structure of mammary gland cells to varying degrees. In vitro, KAM reduced cell viability, migration, and anchorage-independent growth while triggering cell cycle arrest and apoptosis in E2-treated MCF-10A cells. Furthermore, KAM increased cellular antioxidant capacity and attenuated E2-induced oxidative stress. Mechanistically, KAM directly interacted with Src and inhibited its phosphorylation, thus leading to PI3K/AKT pathway inhibition. Notably, the inhibition of E2-induced cell migration and anchorage-independent growth in vitro by Src- or PI3K/AKT pathway-specific inhibitors was not further enhanced when the cells were cultured with KAM. CONCLUSION In summary, KAM targets the Src-mediated PI3K/AKT pathway to reduce oxidative stress and facilitate apoptosis and cell cycle arrest, thereby inhibiting mammary tumorigenesis. Our study is the first to identify Src kinase as a direct target of KAM in mammary tumorigenesis. These findings give significant perspectives on the potential application of KAM in BC chemoprevention.
Collapse
Affiliation(s)
- Pinghan Wang
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Yu Ma
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaohui Rao
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Qianwen Luo
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Xiao Xiao
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Ting Wang
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China.
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China.
| |
Collapse
|
5
|
Yao Y, Guo M, Wu Y, Chen M, Ni K, María De Los Angeles Mibuy MM. Imaging pharmacological and edible behavior of Sinomenii Caulis volatile organic compounds: chain model insight. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:4280-4296. [PMID: 39925206 DOI: 10.1002/jsfa.14169] [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: 06/06/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND As a traditional Chinese medicine, Sinomenii Caulis (SC) has not only attracted much attention for its medicinal value, but also its volatile organic compounds have shown potential application in the agriculture and food fields. This study combined chain modeling (fingerprinting, machine learning, network pharmacology, metabolic analysis, spectroscopic experiments, and physical simulations) with the aim of interdisciplinarity exploring the potential applications of SC, at the interface of agriculture and food, especially in the area of food additives and active ingredient pharmacology. RESULTS Palmitic acid (PA), linoleic acid (LA), and isomenthone (IM), which are closely related to depression, were shown to have good biocompatibility and stability, allowing them to cross the blood-brain barrier and exert their therapeutic effects on depression. Through metabolic analysis, these ingredients can be effectively metabolized in the human body, thus ensuring their safety in food. Through spectral analysis and physical simulation, it was verified that PA/LA/IM can interact with transporter proteins for in vivo transport. CONCLUSION The volatile organic compounds of SC can be used for the treatment of depression and as food additives, which can be fully transported in vivo to perform their roles, which is beneficial for the development of medicines and flavored foods. This study provides a feasible guide for the development of food products with pharmacological activities. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yecen Yao
- College of Chemistry and Materials Engineering, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Ming Guo
- College of Chemistry and Materials Engineering, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Yanan Wu
- College of Chemistry and Materials Engineering, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Mengdie Chen
- College of Chemistry and Materials Engineering, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Kaijie Ni
- College of Chemistry and Materials Engineering, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | | |
Collapse
|
6
|
Chen J, Qu B, Yang D, Wang Y, Zhu H, Wang Z, Zhang X, Ma H, Zhao N, Zhao L, Zhou L, He X, Li P. Combined metabolomics and network pharmacology to elucidate the mechanisms of Huiyang Shengji decoction in treating diabetic skin ulcer mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156569. [PMID: 40120541 DOI: 10.1016/j.phymed.2025.156569] [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: 01/30/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Diabetic skin ulcer is a clinical disorder of glucose metabolism that has a long treatment period and is prone to recurrent episodes. Huiyang Shengji decoction (HYSJD) is an effective traditional Chinese medicine for its clinical treatment, but its metabolic effects in patients with diabetic skin ulcers have not been well studied. PURPOSE Our study aimed to investigate the mechanism of pharmacological treatment of HYSJD in treating diabetic skin ulcers. METHODS The potential mechanism underlying diabetic wound treatment by HYSJD was screened using network pharmacology. Ultra-high performance liquid chromatography-MS/MS metabolomics analysis and correlation analysis were performed to investigate potential target pathways and genes. Furthermore, the db/db diabetic wound tissues and RAW264.7 macrophage inflammation model verified the mechanism using molecular biology experiments. RESULTS In network pharmacology, HYSJD played a mainly therapeutic effect by regulating PI3K/AKT signaling pathway, EGFR tyrosine kinase inhibitor resistance, metabolic pathway, and other related metabolic-related pathways. Metabolomics analysis disclosed that L-lysine content increased, while those of linoleic and deoxycholic acids decreased in plasma between the HYSJD-treated group and the control group, participating in biotin metabolism. Among them, PPARγ played an important role. The experiments conducted in db/db mice indicated that HYSJD facilitates VEGF secretion and PPARγ expression. In vitro experiments have revealed that HYSJD inhibits macrophage ROS production, augments mitochondrial ATP production, elevates mitochondrial membrane potential, and diminishes the mitochondrial ECAR rate. Furthermore, these effects culminate in promoting M2 macrophage polarization through PPARγ activation. The molecular docking results revealed that the active compounds from HYSJD were capable of binding to PPARγ protein primarily through hydrogen bonding interactions. Notably, all binding energies were found to be lower than -3 kcal/mol, indicating strong and favorable interactions between the active compounds and the target receptor. CONCLUSIONS The findings suggested that HYSJD regulates biotin metabolism by reducing excess levels of linoleic and deoxycholic acids and increasing levels of L-lysine, which in turn promotes diabetic wound healing by promoting M2 macrophage polarization through PPARγ up-regulation. These findings indicated that HYSJD is a decoction that can effectively treat diabetic skin ulcers.
Collapse
Affiliation(s)
- Jia Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China; Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, 430022, China
| | - Baoquan Qu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Danyang Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Yazhuo Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Haoyue Zhu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Zhengchun Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Xiawei Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Huike Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Ning Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Li Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China; Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, 430022, China
| | - Lijiaming Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Xiujuan He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China.
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China.
| |
Collapse
|
7
|
Wang JY, Tang R, Chen SM, Su WR, Wang J, Qin ZF, Yin JH, Xuan ZH, Zhao HS, Zhang M, Zhang W, Jia XY. The key active ingredients identification and pharmacological mechanism investigation of extract of ethyl acetate from Er Miao San aganist rheumatoid arthritis. Fitoterapia 2025; 183:106534. [PMID: 40204052 DOI: 10.1016/j.fitote.2025.106534] [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: 10/08/2024] [Revised: 03/21/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Er Miao San (EMS) is a basic formula for clearing heat and drying dampness in traditional Chinese medicine (TCM), which is mainly used in the treatment of rheumatoid arthritis (RA). Previous studies have found that the ethyl acetate extract of EMS (EMS-EA) has the best anti-inflammatory effect, but its specific pharmacological material basis is still unclear. PURPOSE The aim of the study was to investigate the active components of the EMS-EA against RA and its mechanism of action using a combination of serum pharmacochemistry and network pharmacology. METHODS The anti-RA efficacy of EMS-EA was evaluated by establishing a rat model of adjuvant arthritis (AA). The chemical constituents of the EMS-EA and the blood components in the serum of rats after the administration of EMS-EA were detected by the ultra-high liquid chromatography-quadrupole Extractive Orbitrap Mass spectrometry (UPLC-QE-Orbitrap-MS). Network pharmacological analysis was utilized to predict the potential mechanism of action of key blood-entry components against RA, molecular docking, molecular dynamics simulations and in vitro experiments were performed to preliminarily validate the results of network drug prediction. The anti-proliferative activity and pro-apoptotic ability of the key blood-entry components against TNF-α (10 ng/mL)-induced inflammatory injury model of MH7A were detected by MTT assay and TUNEL staining, the levels of IL-6 and IL-1β in the supernatant of the cells were detected by ELISA, and pathway proteins by WB assay. RESULTS Compared with the model group, EMS-EA treatment significantly attenuated the ankle joint injury condition in AA rats, reduced foot volume, arthritis index, organ index and serum levels of TNF-α, IL-6 and IL-1β in rats, and alleviated the pathologies such as formation of vascular opacities and synovial hyperplasia of knee joints to different degrees. In positive and negative ion mode, 51 compounds including 19 alkaloids, 8 terpenoids, Subsequently, berberine (BER) and atractylenolide I (AT-I) were detected in the serum collected from rats after EMS-EA administration. Phellodendrine (PHE) found in rat abdominal aorta serum. Network pharmacology,molecular docking and molecular dynamics simulations results revealed that BER、AT-I and PHE may exert anti-RA effects by modulating the MAPK signaling pathway, whose core targets are mainly CASP3, MAPK1 and MAPK8. Finally, we performed in vitro experiments to investigate the anti-RA activity of the three blood entry components mentioned above. The results showed that all three compounds were able to significantly reduce the TNF-α-induced proliferation level of MH7A cells and increase their apoptotic ability, while inhibiting the release of IL-1β and IL-6. WB experiments showed that all three compounds significantly elevated the level of Cleaved-caspase 3 in TNF-α-induced MH7A cells and down-regulated the phosphorylation levels of JNK and ERK. CONCLUSION EMS-EA has excellent therapeutic effects on AA rats, and its chemical components are mainly alkaloids, organic acids and terpenoids. Among them, BER、AT-I and PHE may be its direct acting substances in vivo, and the mechanism of action may be related to the inhibition of MAPK signaling pathway.
Collapse
Affiliation(s)
- Jia-Yu Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China
| | - Ran Tang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China
| | - Si-Meng Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China
| | - Wen-Rui Su
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China
| | - Jin Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China
| | - Zhi-Fang Qin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China
| | - Jia-Hua Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China
| | - Zi-Hua Xuan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China
| | - Hong-Su Zhao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Min Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Wei Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Xiao-Yi Jia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei 230012, China.
| |
Collapse
|
8
|
Fei L, Zhang D, Mao Y, Mkunga JJ, Chen P, He C, Shan C, Yang X, Cai W. Metabolomics combined with network pharmacology reveals the regional and variety heterogeneity of grape metabolites and their potential antioxidant mechanisms. Food Res Int 2025; 211:116443. [PMID: 40356120 DOI: 10.1016/j.foodres.2025.116443] [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: 11/21/2024] [Revised: 03/23/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
China is one of the world's three major grape-producing regions. However, limited research has focused on the differential metabolites of cross-regional and cross-varietal grapes, and the specific metabolites responsible for their pharmacological effects. Thus, this study comparatively analyzed the antioxidant activities and metabolite compositions of grapes from different regions and varieties to explore the potential antioxidant mechanisms of flavonoid metabolites. The results revealed that the production region primarily influenced the 2,2-Diphenyl-1-picrylhydrazyl (DPPH) of grapes, whereas variety significantly affected the ferric ion reducing antioxidant power (FRAP). Both region and variety had highly significant effects on the total phenolic content (TPC) and total flavonoid content (TFC) of grapes (P < 0.001) and showed significant effects on the 2,2'-Azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS+) and grape metabolites (P < 0.05). However, variety exerted a stronger influence on metabolite composition than region (P < 0.001). Flavonoid metabolites have emerged as key antioxidants, with compounds such as kaempferol, fisetin, and 6-hydroxyluteolin playing critical roles. These metabolites primarily exert their antioxidant effects through signaling pathways, notably the PI3K-Akt pathway. Among all samples, Xinjiang's 'Summer Black' grapes showed the best antioxidant capacity. These findings provide insights into the biochemical basis underlying the differences grapes in China, offering a theoretical foundation for further research on the pharmacological efficacy and antioxidant mechanisms of secondary metabolites in Chinese grapes.
Collapse
Affiliation(s)
- Liyue Fei
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Food Nutrition and Safety Control of Xinjiang Production and Construction Corps, School of Food Science, Shihezi University, Shihezi, Xinjiang, China
| | - Dongsheng Zhang
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Office of the Party Committee of Xinjiang Production and Construction Corps, Urumqi, Xinjiang, China
| | - Yiwen Mao
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Food Nutrition and Safety Control of Xinjiang Production and Construction Corps, School of Food Science, Shihezi University, Shihezi, Xinjiang, China
| | - Johane Johari Mkunga
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Dar-es-salaam Institute of Technology, Dar-es-salaam, Tanzania
| | - Panpan Chen
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Food Nutrition and Safety Control of Xinjiang Production and Construction Corps, School of Food Science, Shihezi University, Shihezi, Xinjiang, China
| | - Chenglong He
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; School of Agriculture, Shihezi University, Shihezi, Xinjiang, China
| | - Chunhui Shan
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Food Nutrition and Safety Control of Xinjiang Production and Construction Corps, School of Food Science, Shihezi University, Shihezi, Xinjiang, China
| | - Xinquan Yang
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China
| | - Wenchao Cai
- Engineering Research Center of Storage and Processing of Xinjiang Characteristic Fruits and Vegetables, Ministry of Education, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Processing and Quality and Safety Control of Specialty Agricultural Products (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Food Science, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Food Nutrition and Safety Control of Xinjiang Production and Construction Corps, School of Food Science, Shihezi University, Shihezi, Xinjiang, China.
| |
Collapse
|
9
|
Mudondo J, Happy K, Okello D, Kang Y. Trichosanthis Radix: A comprehensive review on botany, ethnomedicine, phytochemistry, pharmacology, quality control and toxicology. Fitoterapia 2025; 183:106597. [PMID: 40334818 DOI: 10.1016/j.fitote.2025.106597] [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: 12/30/2024] [Revised: 04/16/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
Trichosanthis Radix, derived from the roots of Trichosanthes kirilowii Maximowicz and Trichosanthes rosthornii Harms, is used widely in traditional Asian medicine. It has been used for centuries across China, Japan, South Korea, and other Asian countries to treat several ailments, including diabetes, cancer, inflammation, cardiovascular and respiratory conditions. The pharmacopoeias in several countries recognize its ability to clear heat, reduce swelling, expel pus, generate fluids, and regulate menstruation. This review provides a comprehensive synopsis of botanical, and ethnomedicinal uses of Trichosanthis Radix. In addition, the phytochemical constituents, including proteins (trichosanthin), terpenoids (cucurbitacins), alkaloids, lignans, coumarins, and flavonoids, which contribute to its diverse pharmacological effects including antimicrobial, antiinflammatory, anticancer, antidiabetic, abortifacient, neuroprotective, immunoregulatory, and antiviral activities are examined. Furthermore, the clinical, pharmacokinetic, quality control measures, processing methods, and toxicity associated with Trichosanthis Radix are discussed. Finally, future research opportunities and potential applications of Trichosanthis Radix in modern medicine are explored with a focus on expanding its therapeutic use and ensuring safe and effective medicinal applications.
Collapse
Affiliation(s)
- Joyce Mudondo
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine (KIOM), 111, Geonjae-ro, Naju-si, Jeollanam-do 58245, Republic of Korea; Korean Convergence Medicine Science Major, University of Science and Technology (UST), Daejeon, 34113, South Korea
| | - Kenneth Happy
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine (KIOM), 111, Geonjae-ro, Naju-si, Jeollanam-do 58245, Republic of Korea; Korean Convergence Medicine Science Major, University of Science and Technology (UST), Daejeon, 34113, South Korea
| | - Dennis Okello
- Department of Biological Sciences, Kabale University, P.O. Box 317, Kabale, Uganda
| | - Youngmin Kang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine (KIOM), 111, Geonjae-ro, Naju-si, Jeollanam-do 58245, Republic of Korea; Korean Convergence Medicine Science Major, University of Science and Technology (UST), Daejeon, 34113, South Korea.
| |
Collapse
|
10
|
Zou QF, Chen DJ, Liu CJ, Chen ZH, Yang X, Xu RH, Zhou ZH, Chen JX, Shi W, Zhang FX. Combination of Metabolite Analysis and Network Pharmacology to Explore the Potential Anticough Mechanism of Protopine-A Marker in Zhi-Ke-Bao Tablets. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e10012. [PMID: 39967298 DOI: 10.1002/rcm.10012] [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: 11/05/2024] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
RATIONALE Protopine, an active alkaloid in Papaver somniferum L., was abundant in a well-known anticough traditional Chinese medicine preparation-Zhi-Ke-Bao tablets. Till now, the metabolism feature and anticough mechanism of protopine have not been fully elucidated, restricting its further development. METHODS The metabolites of protopine in rats were profiled by using ultra-high performance liquid chromatography coupled with time-of-flight mass spectrometry, and its anticough targets and mechanism were predicted by network pharmacology. RESULTS In rats, a total of 19 metabolites were identified following ingestion of protopine (21 mg/kg/day, i.g.), including 4 in plasma, 6 in urine, 5 in feces, 10 in liver, 2 in spleen, 4 in lung, 3 in kidney, 3 in heart, and 3 in brain. The main metabolic features were ring-opening, methylation, demethylation, glucuronidation, sulfation, and hydroxylation. Among them, methylation, sulfation, and hydroxylation of protopine in vivo were revealed for the first time. The network pharmacology results show that protopine and its metabolites regulate physiological activities by acting on STAT3, SRC, CASP3, MTOR, MMP9, ESR1, and other targets, involving PI3K-Akt signaling pathway, FoxO signaling pathway, and TNF signaling pathway, etc. CONCLUSIONS: The metabolic features of protopine and its potential mechanisms for anticough effects were outlined, providing data for further anticough pharmacological validation of protopine.
Collapse
Affiliation(s)
- Qi-Feng Zou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, People's Republic of China
| | - De-Jian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
| | - Cheng-Jun Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, People's Republic of China
| | - Zi-Hao Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, People's Republic of China
| | - Xia Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, People's Republic of China
| | - Rong-Huang Xu
- Teyi Pharmaceutical Group Co., Ltd., Jiangmen, People's Republic of China
| | - Zhen-Hui Zhou
- Teyi Pharmaceutical Group Co., Ltd., Jiangmen, People's Republic of China
| | - Jian-Xin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
| | - Wei Shi
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, People's Republic of China
| | - Feng-Xiang Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, People's Republic of China
| |
Collapse
|
11
|
Luo JX, Zhan Y, Liu YL, Wu ZP, Liu YQ, Cheng XY, Sun DL, Liang XL, Li YM, Xiong YX, Li Q, Liu WJ. Integrating network pharmacology and multi-omics to explore the potentiating effect and mechanism of Shenlingcao oral liquid on cisplatin chemotherapy in Lewis lung cancer mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119843. [PMID: 40258526 DOI: 10.1016/j.jep.2025.119843] [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: 01/18/2025] [Revised: 03/21/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Shenlingcao oral liquid (SLC), derived from traditional clinical formulations, can enhance physical strength and vitality during chemotherapy for malignant tumors. Previous studies have indicated that SLC can enhance the quality of life and daily functioning of patients with non-small cell lung cancer (NSCLC) within 6 months after adjuvant chemotherapy following radical surgery. Nevertheless, its precise mechanism of action is unclear. AIM OF THE STUDY To elucidate the function and underlying mechanism of SLC in enhancing the effect of cisplatin in treating NSCLC. MATERIALS AND METHODS UPLC-Q-Exactive Plus-MS/MS was used to characterize the chemical constituents of SLC. The main gene targets and the pathways of SLC that impact NSCLC were predicted using network pharmacology. A mouse model of Lewis lung cancer and the combination of transcriptomics, metabolomics, and 16S rRNA analysis were used to investigate the underlying mechanisms of the SLC-cisplatin combination in influencing the pathology of NSCLC. RESULTS The SLC-cisplatin combination significantly reduced tumor volume and weight (P < 0.01), boosted T lymphocytes (P < 0.05), increased cleaved-caspase-3/caspase-3 expression (P < 0.05), and decreased p-PI3K/PI3K, p-AKT/AKT, and Bcl-2/Bax protein levels (P < 0.05) compared with cisplatin monotherapy. It also improved the gut flora by enriching the abundance of Bacteroidaceae, S24-7, and Porphyromonadaceae, while modulating metabolic pathways, including caffeine metabolism and fatty acid degradation. CONCLUSIONS SLC enhanced the effect of cisplatin in inhibiting NSCLC by inducing cell apoptosis, augmenting immune responses, balancing the gut microbiota, and regulating key metabolic pathways. Our findings highlight the potential of SLC as an effective adjuvant therapy for NSCLC.
Collapse
MESH Headings
- Animals
- Cisplatin/pharmacology
- Cisplatin/administration & dosage
- Cisplatin/therapeutic use
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/genetics
- Mice, Inbred C57BL
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/administration & dosage
- Network Pharmacology
- Mice
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Male
- Metabolomics
- Antineoplastic Agents/pharmacology
- Drug Synergism
- Administration, Oral
- Cell Line, Tumor
- Multiomics
Collapse
Affiliation(s)
- Jia-Xin Luo
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Yang Zhan
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, PR China.
| | - Yong-Lin Liu
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China.
| | - Zhen-Peng Wu
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Yu-Qing Liu
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Xiao-Ying Cheng
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China.
| | - Deng-Long Sun
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China.
| | - Xin-Li Liang
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Ying-Meng Li
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China.
| | - Yan-Xia Xiong
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China.
| | - Qiong Li
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Wen-Jun Liu
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| |
Collapse
|
12
|
Sheng Y, Li X, Ye X, Fan Q, Li J, Qiao C, Chen X, Yang Q, Wang Z, Li J, Dai S, Chen Y, Tang Y, Zhao C. Integrated transcriptomic and proteomic analysis of hepatotoxic effects of Venenum Bufonis in zebrafish. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119865. [PMID: 40274029 DOI: 10.1016/j.jep.2025.119865] [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: 12/30/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Venenum Bufonis (VB), a traditional Chinese medicine (TCM), is renowned for its therapeutic detoxification, pain relief, and cognitive enhancement effects. VB has been classified as a toxic TCM in medical literature, and its clinical usage is currently subject to several limitations. However, the toxicological characteristics of VB and underlying mechanisms remain unclear. METHODS We conducted a comprehensive assessment to confirm the target organs affected by VB using the zebrafish model. Subsequently, network pharmacology, transcriptomic and proteomic analyses were performed to explore the associated mechanisms, with the aim of providing a basis for its clinical application. RESULTS VB exhibited dose-dependent toxic effects on zebrafish, particularly causing gross morphological abnormalities in the liver along with aggravated hepatocyte apoptosis. Pericardial edema and an enlarged atrioventricular septum were also observed. The combined analyses revealed significant alterations induced by VB in gene expression enriched in multiple pathways and biological processes. Importantly, TLR4/RIPK2/NF-κB and Wnt signaling-mediated inflammation, fibrosis, and apoptosis were identified as the key functional signaling pathways underlying VB-mediated liver toxicity. CONCLUSION Our results present robust and direct evidence of the hepatotoxic effects induced by VB in zebrafish, while also providing novel insights into the molecular pathways involved. These results establish a solid theoretical foundation for the appropriate clinical application of VB.
Collapse
Affiliation(s)
- Yuhan Sheng
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Xinlin Li
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Xinmeng Ye
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - QiQi Fan
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jiaqi Li
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Chuanqi Qiao
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Xiaolu Chen
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Qianwen Yang
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Zetong Wang
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jian Li
- Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Shengyun Dai
- National Institutes for Food and Drug Control, Beijing, 102629, PR China
| | - Yijun Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, PR China.
| | - Yang Tang
- Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| | - Chongjun Zhao
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing, 100102, PR China; China and Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
13
|
Xing Q, Li W, Chen J, Liu Z, Hu Y, Li W, Liu X, Xiao C. Screening the absorbed active components of Danggui Sini Decoction in the treatment of diabetic peripheral neuropathy by network pharmacology combined with molecular docking and dynamics simulation. J Biomol Struct Dyn 2025:1-14. [PMID: 40382779 DOI: 10.1080/07391102.2025.2505251] [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: 02/04/2024] [Accepted: 05/21/2024] [Indexed: 05/20/2025]
Abstract
Diabetic peripheral neuropathy (DPN) is a prevalent and detrimental condition that can be debilitating and even fatal if not treated; however, there remains a dearth of efficacious pharmaceutical interventions for DPN. The Danggui Sini Decoction (DSD), a renowned traditional Chinese medicine prescription, has been utilized in the clinic for the treatment of DPN because of its efficacy in addressing yang deficiency and cold coagulation. However, the active components and underlying mechanisms of DSD remain unclear. In this study, we devised a conventional approach to screen the absorbed active ingredients in DSD, employing LC-MS to identify the principal active compounds of DSD in the blood of rats and then validating these components using network pharmacology for target prediction and molecular dynamics simulations for further validation. We identified 17 potentially active components in the serum and 47 main targets that might be relevant for treating DPN with DSD. These targets were associated with pathways including neuroactive ligand-receptor interaction, HIF-1, and AGE-RAGE signaling pathways, all of which are implicated in diabetic complications. Through molecular docking, we found that glycyrrhetinic acid and betulonic acid-two active components identified by LC-MS in the DSD-containing serum of rats-exhibited strong binding activities with AKT1 and STAT3. Furthermore, molecular dynamics simulations of the docking results indicated that the AKT1-glycyrrhetinic acid and AKT1-betulonic acid complexes were highly stable throughout the kinetic simulations. These findings suggest that the molecular mechanism underlying DSD treatment of DPN may involve the activation of AKT1 by glycyrrhetinic acid and betulonic acid.
Collapse
Affiliation(s)
- Qichang Xing
- Clinical pharmacy, Xiangtan Central Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Wei Li
- Clinical pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | | | - Zheng Liu
- Clinical pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Yixiang Hu
- Clinical pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Wencan Li
- Clinical pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Xiang Liu
- Clinical pharmacy, Xiangtan Central Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Can Xiao
- Clinical pharmacy, Xiangtan Central Hospital, Xiangtan, China
| |
Collapse
|
14
|
Liu M, Liao H, Peng Q, Huang J, Liu W, Dai M, Li Z, Xie Y, Liu J, Ying Y, Zeng X. Comprehensive network pharmacology and experimentation to unveil the therapeutic efficacy and mechanisms of gypenoside LI in anaplastic thyroid cancer. BMC Cancer 2025; 25:870. [PMID: 40369482 PMCID: PMC12076972 DOI: 10.1186/s12885-025-14231-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Anaplastic thyroid cancer (ATC) is a markedly invasive subtype of thyroid cancer with a poor prognosis. The Gynostemma pentaphyllum-derived Gypenoside LI (Gyp LI) can inhibit the growth and metastasis of various tumors. This study was designed to evaluate the pharmacological mechanisms of Gyp LI against ATC via network pharmacology analysis combined with experimental verification. METHODS Core targets and signaling pathways were obtained by using the network pharmacological analysis method. Utilizing a combination of in vitro and in vivo methodologies, we conducted a rigorous examination to ascertain the suppressive impact of Gyp LI on the ATC cell lines, specifically 8305 C and C643. Then used western blotting and immunohistochemistry to analyze the inhibitory effects of Gyp LI on SRC kinase and its downstream signaling pathways. RESULTS Through integrative analysis of Gyp LI and ATC-target interactions, 78 candidate targets were identified. Network-based protein-protein interaction (PPI) analysis, combined with molecular docking, pinpointed HSP90AA1, SRC, and CASP3 as pivotal hub genes modulated by Gyp LI. KEGG enrichment analysis further emphasized the PI3K/AKT pathway, highlighting its critical involvement in ATC therapy. Gyp LI significantly inhibits ATC cell proliferation, migration, and invasion while inducing apoptosis, likely via modulation of the SRC/PI3K/AKT axis. Moreover, it enhances iodine uptake in ATC cells by regulating the sodium-iodide symporter pathway. CONCLUSIONS Gyp LI effectively inhibits ATC progression by modulating SRC/PI3K/AKT signaling, enhancing apoptosis, and promoting iodine uptake, offering potential therapeutic benefits for ATC treatment.
Collapse
Affiliation(s)
- Meiyu Liu
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Ganzhou Key Laboratory of Thyroid Cancer, Ganzhou, Jiangxi, 341000, China
| | - Haidong Liao
- Department of General Surgery, First People's Hospital of Longnan, Longnan, Jiangxi, 341706, China
| | - Qin Peng
- Department of Pathology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Junwei Huang
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Weixiang Liu
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Mengqiao Dai
- Institute of Thyroid Diseases of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Zanbing Li
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Institute of Thyroid Diseases of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Yang Xie
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Institute of Thyroid Diseases of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Jiafeng Liu
- Institute of Thyroid Diseases of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Yong Ying
- Ganzhou Key Laboratory of Thyroid Cancer, Ganzhou, Jiangxi, 341000, China.
| | - Xiangtai Zeng
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Department of General Surgery, First People's Hospital of Longnan, Longnan, Jiangxi, 341706, China.
| |
Collapse
|
15
|
Yang Z, Sun T, Zhang M, Li X, Wang Y, Li W, Wang Q, Wang X, Liu S, Wang W, Wang P. Sodium houttuyfonate prevents NiO-NPs induced lung injury in rats through the TNF-α/JAK-STAT signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118306. [PMID: 40367609 DOI: 10.1016/j.ecoenv.2025.118306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Nickel oxide nanomaterials (NiO-NPs), as engineered nanomaterials, are frequently implicated in the onset of various inflammatory disorders. However, no specific therapeutic agent is currently available for pneumonia induced by NiO-NPs exposure. In the present study, a cytotoxicity model was established using rat type Ⅱ alveolar epithelial cells (ACE-Ⅱ) to investigate the cellular effects of NiO-NPs, while a rat model of lung injury was developed in Wistar rats via a non-exposure tracheal instillation technique. To address this pathology, we formulated a sodium houttuyfonate (SH) nanoemulsion characterized by favorable fluidity and uniform dispersion, and evaluated its therapeutic efficacy and underlying mechanisms in NiO-NPs-induced acute lung injury through three distinct administration routes: Houttuynia cordata decoction, SH tablets and SH nanoemulsion. Concurrently, network pharmacology and transcriptomic analyses were conducted to identify key molecular targets and signaling pathways. The findings demonstrated that both Houttuynia cordata and SH significantly reduced TNF-α levels and inhibited the activation of the JAK-STAT signaling pathway both in vitro and in vivo, thereby mitigating NiO-NPs-induced pulmonary injury. Among the various formulations tested, the SH nanoemulsion exhibited the most pronounced therapeutic efficacy. This study offers a promising therapeutic strategy for NiO-NPs-induced lung injury and contributes to the advancement of pharmacological interventions for nanomaterial-related inflammatory diseases.
Collapse
Affiliation(s)
- Zongtong Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; Institute of Pharmacology of Traditional Chinese Medicine, Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Tiefeng Sun
- Institute of Pharmacology of Traditional Chinese Medicine, Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Mengru Zhang
- Institute of Pharmacology of Traditional Chinese Medicine, Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Xiaojing Li
- Institute of Pharmacology of Traditional Chinese Medicine, Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Yi Wang
- Institute of Pharmacology of Traditional Chinese Medicine, Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Wei Li
- Shandong University, Biomedical Engineering Institute, School of Control Science and Engineering, Jinan 250014, China
| | - Qiwei Wang
- Qilu Institute of Technology, Jinan 250200, China
| | - Xinru Wang
- Qilu Institute of Technology, Jinan 250200, China
| | - Shizeng Liu
- Jining Medical University, Jining 276826, China
| | - Wenhui Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China.
| | - Ping Wang
- Institute of Pharmacology of Traditional Chinese Medicine, Shandong Academy of Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
16
|
Sharma A, Singh R, Virmani T, Chouhan NK, Kapoor G, Kumar G, Bhutani R, Rana N, Sharma S. Cannabichromene, a key non-psychotropic phytocannabinoid in treatment of major depressive disorder: in silico and in vivo explorations. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04236-2. [PMID: 40358684 DOI: 10.1007/s00210-025-04236-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025]
Abstract
Cannabichromene, a non-psychotropic cannabinoid with antioxidant and neuroprotective properties, is hypothesized to possess antidepressant potential. This study aimed to evaluate cannabichromene's depression-alleviating effects in mice exposed to chronic unpredictable mild stress and unstressed mice using a combination of in silico and in vivo approaches. Initially, gene targets associated with major depressive disorder were identified through GeneCards, while cannabichromene's target genes were predicted using SwissTargetPrediction. Overlapping targets were visualized using Venny software, and protein-protein interaction networks were constructed with the STRING database. The cannabinoid receptor two genes, encoding the cannabinoid 2 receptor, emerged as a key shared target. Molecular docking studies revealed that cannabichromene exhibited a strong binding affinity to cannabinoid 2 receptors (docking score: - 9.4) compared to cannabidiol (CBD) (- 8.8) and Δ9-tetrahydrocannabinol (- 9.1). For in vivo analysis, male Swiss albino mice were subjected to chronic unpredictable mild stress for 3 weeks to induce depression-like behavior. Cannabichromene (10 and 20 mg/kg) and imipramine (15 mg/kg) were administered for 21 days. Cannabichromene at 20 mg/kg significantly reduced immobility in stressed mice, like imipramine, without affecting locomotor activity. Additionally, both cannabichromene and imipramine reduced elevated plasma nitrite and corticosterone levels and inhibited monoamine oxidase-A activity in the brain. Cannabichromene also reversed stress-induced catalase suppression. In conclusion, cannabichromene revealed a relatively substantial antidepressant character with chronic unpredictable mild stress model of depression in Swiss albino male mice, likely through interaction with cannabinoid 2 receptors encoded by the cannabinoid 2 gene, as ratified via in silico modeling and in vivo findings. This highlights cannabichromene's potential as a novel therapeutic agent for depression after further in vitro and clinical assessments in other models.
Collapse
Affiliation(s)
- Abhishek Sharma
- Department of Pharmacy, School of Healthcare and Allied Sciences, G D Goenka University, Gurugram, 122103, Haryana, India.
- Amity Institute of Pharmacy, Amity University, Greater Noida Campus, Knowledge Park III, Greater Noida, Uttar Pradesh, 201308, India.
| | - Rahul Singh
- Department of Pharmacy, School of Healthcare and Allied Sciences, G D Goenka University, Gurugram, 122103, Haryana, India
| | - Tarun Virmani
- Amity Institute of Pharmacy, Amity University, Greater Noida Campus, Knowledge Park III, Greater Noida, Uttar Pradesh, 201308, India
| | - Neeraj Kumar Chouhan
- Amity Institute of Pharmacy, Amity University, Greater Noida Campus, Knowledge Park III, Greater Noida, Uttar Pradesh, 201308, India
| | - Garima Kapoor
- School of Pharmacy, KIET Group of Institutions, Uttar Pradesh, Ghaziabad, 201206, India
| | - Girish Kumar
- Amity Institute of Pharmacy, Amity University, Greater Noida Campus, Knowledge Park III, Greater Noida, Uttar Pradesh, 201308, India
| | - Rubina Bhutani
- Department of Pharmacy, School of Healthcare and Allied Sciences, G D Goenka University, Gurugram, 122103, Haryana, India
| | - Neha Rana
- School of Pharmacy, Noida International University, Yamuna Expressway, Gautam Budh Nagar, 203201, Uttar Pradesh, India
| | - Shivkant Sharma
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122003, Haryana, India.
| |
Collapse
|
17
|
Li X, Liu X, Li B, Liu Z, Guo S, Xiong S, Xiao T, Jiao S, Chen Y, Xu Y. Identifying the active components and mechanisms of Persicae Semen in treating androgenetic alopecia: Insights from network pharmacology and experimental evaluations. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119755. [PMID: 40220939 DOI: 10.1016/j.jep.2025.119755] [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: 01/27/2025] [Revised: 03/22/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Androgenic alopecia (AGA) is the most prevalent form of hair loss, which affects self-perception and life satisfaction. Current treatments for AGA are limited. In Traditional Chinese Medicine, Persicae Semen (Taoren, TR) is used in formula to mitigate alopecia. However, the principal active constituents and their mechanisms of action in anti-alopecia effects remain fully undefined. AIM OF THE STUDY This study aimed to elucidate the active constituents and multifaceted mechanisms of TR in AGA treatment through network pharmacology analysis, molecular docking, and experimental validation. MATERIALS AND METHODS The chemical constituents of TR were systematically characterized using ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS). In addition, a comprehensive compound-disease-target interaction network was constructed to elucidate the molecular mechanism underlying its therapeutic effects on AGA. Molecular docking was performed to validate the interactions between the key bioactive components and core targets. Furthermore, a multi-level pharmacological investigation comprising in vitro cellular assays, ex vivo organ studies, and in vivo animal experiments was conducted to preliminarily explore the therapeutic mechanisms by which high-content active compound treated AGA. RESULTS UPLC-Q-TOF-MS analysis identified 32 chemical constituents in TR. Through integrated network pharmacology analysis, six bioactive components and 10 core targets were systematically screened for molecular docking, which revealed therapeutic pathways primarily involved in anti-inflammatory responses, angiogenesis, and hair follicle microenvironment modulation. Given its high abundance and superior bioactivity, amygdalin (Am) was selected as the primary research focus. Combined with the pathological mechanism of the disease, which was confirmed through in vitro and in vivo experiments, Am promoted hair regeneration by regulating genes, proteins and inflammatory factors related to the androgen, Wnt/β-catenin, and vascular endothelial growth factor pathways. CONCLUSIONS In this study the multi-component, multi-target, and multi-pathway mechanisms underlying the therapeutic effects of TR on AGA as well as the molecular mechanism by which Am treats AGA was elucidated. These findings provide substantial theoretical foundations and experimental evidence for the development of novel AGA therapeutics based on TR and its active constituents.
Collapse
Affiliation(s)
- Xiaojuan Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Pharmacy School of Shihezi University, Xinjiang, 832000, China.
| | - Xiaoli Liu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China.
| | - Bin Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Ziyi Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Shiqi Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Sha Xiong
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Ting Xiao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Siwen Jiao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Yu Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Yuehong Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
18
|
Fang Y, Min S, Wu Y, Xu F, Chen H, Li Y, Lu Y, Hu J, Zhu L, Shen H. Integration of Multi-Omics and Network Pharmacology Analysis Reveals the Mechanism of Qingchang Huashi Jianpi Bushen Formula in Repairing the Epithelial Barrier of Ulcerative Colitis. J Inflamm Res 2025; 18:6167-6189. [PMID: 40386180 PMCID: PMC12083493 DOI: 10.2147/jir.s510966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
Purpose Derivation of Qingchang Huashi formula, named Qingchang Huashi Jianpi Bushen (QCHS_JPBS) formula, has shown significant therapeutic effect on patients with ulcerative colitis (UC). In this study, the potential mechanism of QCHS_JPBS formula in repairing mucosal damage was explored from the perspective of intestinal stem cell (ISCs) differentiation, and potential targets of the QCHS_JPBS formula to improve UC were predicted using network pharmacology analysis. Methods The therapeutic efficacy of QCHS_JPBS formula was evaluated in a mouse model of 2.5% dextran sulfate sodium (DSS) induced colitis. The effect of this formula on the ISC differentiation was evaluated using tissue transmission electron microscopy, immunofluorescence, and RT-qPCR. The cecal contents were subjected to 16s RNA sequencing analysis and non-target metabolomics analysis using LC-MS/MS. The fecal microbiota transplantation method verified the essential role of gut microbiota in promoting ISC differentiation and repairing mucosal damage. Results The results indicated that QCHS_JPBS formula suppressed the inflammatory response and repaired the damaged intestinal epithelial barrier in DSS-induced colitis mice. QCHS_JPBS formula promoted ISC differentiation, particularly in the direction of goblet cells. QCHS_JPBS formula restored gut dysbiosis and regulated metabolic disorders in DSS-induced colitis mice. And then, the results of fecal microbiota transplantation indicated that QCHS_JPBS formula promoted differentiation of intestinal stem cells to repair mucosal damage through gut microbiota. Finally, a total of 79 active ingredients of QCHS_JPBS formula were identified based on LC-MS analysis and EGFR, STAT3, SRC, AKT1, and HSP90AA1 were considered as potential therapeutic UC targets of QCHS_JPBS formula based on network pharmacology analysis. Conclusion The present study demonstrated that QCHS_JPBS formula promoted the differentiation of ISCs through gut microbiota to repair the damaged intestinal epithelial barrier in UC mice.
Collapse
Affiliation(s)
- Yulai Fang
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Shichen Min
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yuguang Wu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Feng Xu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Hongxin Chen
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yanan Li
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yizhou Lu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Jingyi Hu
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Lei Zhu
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Hong Shen
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
19
|
Yang G, Tan W, Yan L, Lao Q, Zheng W, Ding H, Yu J, Liu Y, Zou L, Guo M, Yu L, Zhou X, Li W, Yang L. Phillyrin for sepsis-related acute lung injury: A potential strategy suppressing GSK-3β. Mol Immunol 2025; 183:115-136. [PMID: 40359720 DOI: 10.1016/j.molimm.2025.04.017] [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: 12/21/2024] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
The efficacy of clinical drugs for acute lung injury/acute respiratory distress syndrome (ALI/ARDS) remains suboptimal. Phillyrin (PHN), a compound derived from Forsythia, is believed to alleviate sepsis-related ALI/ARDS; however, its mechanisms are not fully elucidated. In this study, we screened 8331 target genes associated with ALI/ARDS from public databases and identified six hub genes relevant to PHN treatment: AKT1, GSK-3β, PPP2CA, PPP2CB, PPP2R1A, and AR. Receiver operating characteristic analysis and single-cell sequencing analysis revealed the expression of AKT1, GSK-3β, PPP2CA, PPP2CB, and PPP2R1A were markedly elevated. Molecular docking and dynamics simulations indicated that PHN forms a structurally stable complex with glycogen synthase kinase-3β (GSK-3β). Mendelian randomization analyses suggested that PHN, as a potent GSK-3β inhibitor, may promote M2 macrophage polarization and reduce neutrophil recruitment. We validated these findings through in vivo and in vitro experiments, demonstrating that PHN lowers iNOS levels and raises MMR levels by downregulating GSK-3β mRNA expression and protein activity during lipopolysaccharide (LPS)-induced macrophage inflammation. Additionally, PHN inhibited GSK-3β mRNA expression and protein activity, reducing NF-κB-p65 nuclear translocation in LPS-induced zebrafish inflammation and mice ALI. This inhibition decreased levels of TNF-α and IL-6, increased IL-10 levels, promoted M2 macrophage polarization, suppressed neutrophil recruitment, and ultimately ameliorated ALI/ARDS. In conclusion, our results indicate that PHN effectively alleviates LPS-induced ALI/ARDS by suppressing GSK-3β signaling.
Collapse
Affiliation(s)
- Guangli Yang
- Department of Central Laboratory, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Weifu Tan
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Lijun Yan
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qiaocong Lao
- Central Laboratory, The Tenth Affiliated Hospital, Southern Medical University, Dongguan People's Hospital, Dongguan 523059, China
| | - Wujuan Zheng
- Department of Pharmacy, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Hongyan Ding
- Omega-3 Research and Conversion Center, Dongguan Innovation Research Institute, Guangdong Medical University, Dongguan 523900, China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yong Liu
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Liyi Zou
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Maorun Guo
- Pingyi Health Center of Pingyi County, Linyi 273300, China
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Wei Li
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China.
| | - Liling Yang
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China; Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Pharmacy, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China.
| |
Collapse
|
20
|
Hua C, Wu M, Xiao Y, Zhang R, Yuan Y, Zhang L, Guo F, Liu J, Yang Z, Liu G. Dendrobium nobile lindl extract modulates integrin αIIbβ3-mediated signaling pathways to inhibit platelet activation and thrombosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119728. [PMID: 40180000 DOI: 10.1016/j.jep.2025.119728] [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: 12/30/2024] [Revised: 03/11/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dendrobium nobile Lindl. (DNL) is a promising medicinal plant. It has the traditional medicinal effects of promoting blood circulation and resolving stasis, as well as regulating the meridians and collaterals. AIM OF THE STUDY We studyed how DNL extract was involved in platelet activation and thrombosis and used network pharmacology and molecular docking analysis to help clarify the underlying mechanisms. MATERIALS AND METHODS The effect of DNL extract on platelet aggregation and ATP release function was examined by aggregometer; The effect of DNL extract on the binding of PAC-1 and fibrinogen to integrin was determined by flow cytometry; The effect of DNL extract on "outside-in" platelet signals was detected by platelet adhesion, spreading and clot retraction; Key compounds and major targets of platelet interactions with DNL extract were analyzed by network pharmacology and molecular docking and verified against related pathway proteins by western blotting; The effect of DNL extract on thrombosis was tested by mesenteric artery embolism model. RESULTS DNL extract exhibited inhibition of platelet function and PAC-1 and fibrinogen binding to integrin αIIbβ3. In addition, it delayed FeCl3-induced mesenteric artery thrombosis without affecting the clotting time and the hemostatic time of tail in mice. The detection of platelet "inside-out" and "outside-in" signaling by Western blot further confirmed the inhibitory effect of DNL extract on platelet activation. CONCLUSIONS DNL extract may affect the thrombosis process by inhibiting platelet activation via inhibiting integrin αⅡbβ3-mediated bidirectional signaling pathway proteins.
Collapse
Affiliation(s)
- Chaoying Hua
- Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 561113, China
| | - Meng Wu
- Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 561113, China
| | - Yi Xiao
- Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 561113, China
| | - Rui Zhang
- School of Pharmacy, Guizhou Medical University, Guiyang, 561113, China
| | - Yujing Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 561113, China
| | - Li Zhang
- School of Pharmacy, Guizhou Medical University, Guiyang, 561113, China
| | - Fang Guo
- Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 561113, China
| | - Jian Liu
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Zhanzhan Yang
- School of Pharmacy, Guizhou Medical University, Guiyang, 561113, China.
| | - Gang Liu
- Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 561113, China; Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, 561113, China.
| |
Collapse
|
21
|
Xu W, Zhu Q, Chen J, He J, Yuan A, Cao P, Zhang L. Exploring the mechanisms of artemisinin and its derivatives in the treatment of atopic dermatitis based on network pharmacology and molecular docking: A review. Medicine (Baltimore) 2025; 104:e42287. [PMID: 40355181 PMCID: PMC12074105 DOI: 10.1097/md.0000000000042287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 04/10/2025] [Indexed: 05/14/2025] Open
Abstract
This study investigates the therapeutic mechanisms of artemisinin (ARS) and its derivatives in atopic dermatitis (AD) using network pharmacology and molecular docking. Molecules and disease targets were screened using public databases, including SwissTargetPrediction, PharmMapper, and Genecards. Core targets were identified, and a protein-protein interaction (PPI) network was constructed using STRING and Cytoscape for topological analysis. Relevant data were obtained from the DAVID database for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Molecular docking of ARS and its derivatives with target genes was performed using AutoDock, with results visualized in Pymol. A functional PPI network was established, and molecular docking demonstrated strong binding activity between ARS derivatives and target protein. Mitogen-Activated Protein Kinase14 (MAPK14) and Mitogen-Activated Protein Kinase10 (MAPK10) was found to be a common target for their treatment of AD. ARS and its derivatives may treat AD by modulating pathways such as Prolactin signaling, cancer pathways, neuroactive ligand-receptor interaction, and IL-17 signaling. ARS and its derivatives have the potential to treat AD. Artemisinin, artesunate, dihydroartemisinin, artemether, artemisinin and artemisinone could potentially treat AD by targeting MAPK14 and MAPK10.
Collapse
Affiliation(s)
- Wenjing Xu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianyu Zhu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Jiaxing Chen
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junchen He
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
- Tianjin Institute of Integrative Dermatology, Tianjin, China
| | - Aijie Yuan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peng Cao
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
- Tianjin Institute of Integrative Dermatology, Tianjin, China
| | - Litao Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
- Tianjin Institute of Integrative Dermatology, Tianjin, China
| |
Collapse
|
22
|
Xu F, Liu Q, Cai S, Yu Q, Zhang Y, Liu Z, Zhao H, Zhang L. Effects and Mechanisms of Paeoniflorin in Relieving Neuropathic Pain: Network Pharmacological Analysis and Experimental Validation. Neurochem Res 2025; 50:160. [PMID: 40343651 PMCID: PMC12064598 DOI: 10.1007/s11064-025-04411-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/27/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Neuropathic pain (NP) is a prevalent condition that can lead to a variety of complications, significantly impacting patients' quality of life. Previous studies have confirmed that paeoniflorin (PF) demonstrates both therapeutic pain relief and neuroprotective effects. However, its therapeutic efficacy in managing NP remains to be thoroughly investigated. We conducted a systematic study to explore the underlying mechanisms of PF in the treatment of NP through combining network pharmacological analysis with experimental validation. Our studies revealed that PF alleviates NP through a multifaceted approach, mainly involving protein kinase C (PKC), serotonin receptors, calcium signaling pathways, inflammatory mediator regulation of transient receptor potential (TRP) channels, and G-protein coupled receptor signaling pathways. Additionally, our animal experiments indicated that PF reduces pain-related behavior on spinal nerve ligation-induced NP in rats by modulating the PKCε-TRPV1 pathway. PF was found to inhibit the expression of inflammatory factors such as interleukin 6 and tumor necrosis factor α, as well as the activation of microglia, thereby alleviating NP. These findings suggest a potential therapeutic role for PF in the treatment of NP, providing a valuable reference for clinical applications.
Collapse
Affiliation(s)
- Fangning Xu
- Department of Anesthesiology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210008, P. R. China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, P. R. China.
| | - Shenquan Cai
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, P. R. China
| | - Qiuyan Yu
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, P. R. China
| | - Yue Zhang
- Department of Anesthesiology, Jinling Hospital, Medical College of Nanjing Medical University, Nanjing, Jiangsu Province, 210008, P. R. China
| | - Zhi Liu
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, P. R. China
| | - Haishu Zhao
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, P. R. China
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210008, P. R. China.
| |
Collapse
|
23
|
Heidari-Japelaghi R, Valizadeh M, Haddad R. Interferon gamma-induced hub genes and key pathways: A study based on biological network analysis and experimental validation. J Biotechnol 2025; 405:72-87. [PMID: 40348089 DOI: 10.1016/j.jbiotec.2025.04.022] [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: 03/16/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
By performing a biological network analysis, we identified some hub genes, which were up- or down-regulated in the breast cancer (BC) cell line after treatment with IFN-γ. Moreover, several pathways including cytokine-cytokine receptor interaction, TNF signaling pathway, NOD-like receptor signaling pathway, and NF-κB signaling pathway were detected that their activation leads to the antiproliferation, proapoptosis, and antiviral activities. To validate in silico results, the bioactivity of recombinant human IFN-γ (hIFN-γ) produced in different hosts was analyzed by antiviral and anticancer assays. The antiviral role of the hIFN-γ preparations was evaluated by inhibition of Vesicular Stomatitis Virus (VSV)-mediated cytopathic effects on Vero cells. A dose-dependent increase in cell viability was observed at different concentrations of recombinant proteins. The maximum amount of the cell viability detected for the hIFN-γ preparations was determined at a concentration of 32.00 pg/mL. To analyze the cytotoxic efficacy of the hIFN-γ preparations on the growth and development of tumor cells, a BC cell line (MCF-7) was treated with both recombinant protein forms in a time- and dose-dependent way. The highest level of inhibiting cell proliferation was detected at a concentration of 32.00 pg/mL hIFN-γ after 72 h incubation. Anticancer and antiviral functions of IFN-γ were confirmed via the expression analysis of hub genes cd74, cxcl10, il6, and stat1 using RT-PCR. Furthermore, the hIFN-γ preparations were significantly able to up-regulate the expression of proapoptotic Bax and p53 and to down-regulate Bcl-2 as an antiapoptotic gene, showing the cytotoxic effect of hIFN-γ toward MCF-7 cells via apoptosis induction.
Collapse
Affiliation(s)
- Reza Heidari-Japelaghi
- Department of Biotechnology Engineering, Faculty of Agriculture and Natural Resources, Imam Khomeini International University, Qazvin, Iran.
| | - Mostafa Valizadeh
- Department of Plant Breeding and Biotechnology, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Raheem Haddad
- Department of Biotechnology Engineering, Faculty of Agriculture and Natural Resources, Imam Khomeini International University, Qazvin, Iran
| |
Collapse
|
24
|
Gu S, Chen C, Wang J, Wang Y, Zhao L, Xiong Z, Zhang H, Deng T, Pan Q, Zheng Y, Li Y. Camellia Japonica Radix modulates gut microbiota and 9(S)-HpODE-mediated ferroptosis to alleviate oxidative stress against MASLD. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156806. [PMID: 40334428 DOI: 10.1016/j.phymed.2025.156806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/04/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Camellia japonica radix (CJR), derived from the root of Camellia japonica L., has the potential to function as an herbal tea substitute for the prevention and intervention of metabolic dysfunction-associated steatotic liver disease (MASLD). It can provide systemic therapeutic benefits, boast a favorable safety profile, facilitate convenient consumption, and support long-term applicability. Despite its potential, research on CJR remains limited. PURPOSE The aim of this study aims is to elucidate the therapeutic mechanisms of CJR in MASLD, thereby providing evidence to support its clinical application. METHODS The therapeutic effects of CJR were evaluated using a water-supplementation model in MASLD mice. Integrated microbiome, transcriptome, proteome, and metabolome analyses were employed to comprehensively explore the mechanisms involved. A drug-target pull-down assay was performed to identify specific protein targets of small molecule metabolites in vitro. Fecal microbiota transplantation in antibiotic-treated ABX mice was conducted to confirm the critical role of gut microbiota and its metabolites. Furthermore, customized medicated feed supplemented with linoleic acid was used to explore the intervention effect of its metabolite, 9(S)-HpODE, as well as to evaluate its dietary intervention potential. RESULTS This present study explicitly elucidates the efficacy of CJR extract in alleviating hepatic inflammation and steatosis in a MASLD model mice, with its pharmacological mechanism associated with gut microbiota, linoleic acid metabolism, and GPX4-mediated ferroptosis. Notably, 9(S)-HpODE was discovered to be a key metabolite of linoleic acid, which could target both KEAP1 and SLC7A11, bidirectionally regulating GPX4-mediated ferroptosis, while acting as a signaling molecule at low doses to induce redox adaptation via oxidative preconditioning, thus ameliorating oxidative stress in MASLD. CONCLUSION Our findings indicate that both CJR and linoleic acid exhibit significant potential as dietary interventions for the management of MASLD, offering promising avenues for future research and clinical application.
Collapse
Affiliation(s)
- Simin Gu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chong Chen
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junmin Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanping Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lina Zhao
- Department of Hepatobiliary Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Zhekun Xiong
- Department of Spleen, Stomach and Hepatobiliary, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Hui Zhang
- Department of Spleen, Stomach and Hepatobiliary, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Taoying Deng
- Department of Spleen, Stomach and Hepatobiliary, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Qihui Pan
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyuan Zheng
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yong Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
25
|
Sangande F, Ningsih S, Agustini K, Andrina Kusumastuti S, Nuralih N, Arditya Fajriawan A, Chandra M, Rosmalawati S, Asnawi A, Budipramana K. Effects and molecular mechanisms of the combination of Andrographis paniculata and Anredera cordifolia as an insulin sensitizer: in vitro, network pharmacology, molecular docking, and dynamics studies. J Biomol Struct Dyn 2025:1-12. [PMID: 40326956 DOI: 10.1080/07391102.2025.2499224] [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: 11/30/2023] [Accepted: 08/27/2024] [Indexed: 05/07/2025]
Abstract
Due to the complex mechanism of insulin resistance (IR), multi-component herbal medicines might be an alternative approach in the treatment of IR-related diseases, such as type 2 diabetes mellitus (T2DM). Andrographis paniculata (AP) and Anredera cordifolia (AC) have been reported to have anti-diabetic effects. However, their effect and mechanism of action in a mixed formula (FAPAC), especially as an insulin sensitizer have not been reported. Therefore, in vitro studies were performed to investigate the effect of FAPAC, and the molecular mechanisms were predicted by in silico studies through network pharmacology, molecular docking, and dynamics simulations. In vitro studies demonstrated that FAPAC at 2 µg/mL was comparable to metformin in increasing glucose uptake in IR-HepG2 cells. KEGG analysis revealed that IR was the top pathway and predicted that FAPAC acts as an insulin-sensitizing agent by inhibiting three main targets: IKBKB, PRKCD, and PTPN1. Consensus docking suggested 7-O-methyl wogonin, ninandrographolide, and 3-O-β-D-glucopyranosyl andrographolide as the potent inhibitors for IKBKB, PRKCD, and PTPN1, respectively. Furthermore, molecular dynamics confirmed that the potential compounds remained in the binding pocket throughout the simulation and had a good affinity toward their respective targets, comparable to native ligands.
Collapse
Affiliation(s)
- Frangky Sangande
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center, Bogor, Indonesia
| | - Sri Ningsih
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center, Bogor, Indonesia
| | - Kurnia Agustini
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center, Bogor, Indonesia
| | - Siska Andrina Kusumastuti
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center, Bogor, Indonesia
| | - Nuralih Nuralih
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center, Bogor, Indonesia
| | - Adam Arditya Fajriawan
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center, Bogor, Indonesia
| | | | - Syofi Rosmalawati
- Research Center for Applied Microbiology, National Research and Innovation Agency (BRIN), Cibinong Science Center, Bogor, Indonesia
| | - Aiyi Asnawi
- Faculty of Pharmacy, Universitas Bhakti Kencana, Bandung, Indonesia
| | - Krisyanti Budipramana
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Surabaya, Surabaya, Indonesia
| |
Collapse
|
26
|
Duan Q, Wang M, Cui Z, Ma J. Saikosaponin D suppresses esophageal squamous cell carcinoma via the PI3K-AKT signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6059-6070. [PMID: 39638887 DOI: 10.1007/s00210-024-03676-6] [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: 10/21/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
Saikosaponin D is the saikosaponin with the highest biological activity in Bupleurum chinense DC, which has anti-tumor effects on a variety of human tumors. In this study, we aimed to explore the SSD-induced apoptosis mechanism in ESCC cells. We predicted the targets of SSD and ESCC through several databases and analyzed the intersecting targets to identify the connections and possible pathways between proteins. We evaluated the binding activity between proteins and SSD through molecular docking. Based on the network pharmacology results, different concentrations of SSD were used to treat Eca-109 alongside Te-10 cells. The CCK-8, colony formation, wound healing, transwell, apoptosis, and western blot assays were performed to verify the inhibitory SSD impact on Eca-109 and Te-10 cells. Network pharmacology predicted 186 potential targets of SSD, and 500 targets of ESCC, along with 31 common targets, 5 core protein targets, and 94 potential pathways. Depending on molecular docking findings, SSD was closely bound to five core targets. Cellular experiments showed that SSD suppressed the Eca-109 and Te-10 cell proliferation and metastasis and enhanced apoptosis via the PI3K-AKT signaling. This study suggests SSD inhibited Eca-109 and Te-10 cell proliferation and migration by inhibiting the PI3K-AKT pathway and promoting apoptosis.
Collapse
Affiliation(s)
- Qiong Duan
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China
| | - Mingxiao Wang
- Sichuan Integrative Medicine Hospital, Chengdu, 610000, China
| | - Zhenting Cui
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China
| | - Jianxin Ma
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China.
| |
Collapse
|
27
|
Li M, Liu B, Xian M, Wang S, Liu P. Bioinformatics combined with network pharmacology and experimental validation to identify key biomarkers of hepatocellular carcinoma and corresponding compounds in Radix Astragali and Pueraria Mirifica. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5351-5371. [PMID: 39549064 DOI: 10.1007/s00210-024-03597-4] [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: 09/30/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024]
Abstract
The occurrence and death rates of primary hepatocellular carcinoma (HCC) are increasing, and there remains a shortage of effective oral medications with minimal side effects. We aim to identify potential biomarkers and compounds from Radix Astragali (RA) and Pueraria Mirifica (PM) to treat liver cancer and improve prognosis. Differentially expressed genes (DEGs) associated with HCC were identified by bioinformatics analysis of three datasets, GSE112791, GSE101685, and GSE45114. Using public databases to predict the bioactive components and possible targets of RA and PM. Target crossover from Gene Expression Omnibus (GEO) and public databases were used to identify potential biomarkers for HCC. Subsequently, validation and prognostic value analyses were performed using the Gene Expression Profile Interaction Analysis (GEPIA) platform. The Cytoscape software created a network of "compound targets" to pinpoint compounds linked to the biomarkers. Molecular docking techniques were utilized to validate the connection between these compounds and the identified biomarkers. Ultimately, the HepG2 liver cancer cell line was chosen to assess the inhibitory effect of Hederagenin (HDG) and to confirm the expression of ADH1B through Western blot analysis. In this study, four key biomarkers (NR1I2, ADH1B, NQO1, GHR) were identified. Molecular docking showed that these four core targets could form stable conformations with the corresponding compounds. As the drug concentration decreases, the inhibitory effect on HepG2 diminishes, and the survival rate of HepG2 cells significantly declines following the administration of 100 µmol/L HDG. Compared to the control, the expression of ADH1B protein is significantly increased in HepG2 cells treated with 100 µmol/L HDG. The study identified four key biomarkers (ADH1B, GHR, NQO1, NR1I2) that have prognostic ability for HCC. This study provides biomarkers and potential targeted monomeric medicines for treating HCC.
Collapse
Affiliation(s)
- Mohan Li
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Traditional Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Engineering and Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bang Liu
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Traditional Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Engineering and Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Minghua Xian
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Traditional Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Engineering and Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shumei Wang
- Guangdong Pharmaceutical University, Guangzhou, 510006, China.
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Traditional Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.
- Engineering and Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China.
- Traditional Chinese Medicine Resource Germplasm Bank Management Center, Yunfu, 527300, China.
| | - Peiyi Liu
- Huangpu People's Hospital of Zhongshan, Zhongshan, 528429, China
| |
Collapse
|
28
|
Wang Y, Dai X, Zhu X, Wang W, Wang C, Li S, Sun S, Gao X, Qin K. Integrating Serum Pharmacochemistry With Network Pharmacology to Elucidate the Mechanism of Wushen Decoction in the Prevention and Treatment of Lower Extremity Erysipelas. Biomed Chromatogr 2025; 39:e70076. [PMID: 40207497 DOI: 10.1002/bmc.70076] [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: 12/10/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
Lower extremity erysipelas (LEE), a frequently seen skin and soft tissue infection caused predominantly by streptococci, usually presents with fever, erythema and pain. Wushen Decoction (WSD), a Compound traditional Chinese medicine, has been used historically to treat LEE, though its exact mechanism of action remains unclear. In this study, we explored the therapeutic mechanisms of WSD in treating LEE by employing a combination of serum pharmacochemistry, network pharmacology, and molecular docking techniques. Initially, using UPLC-Q-Exactive Orbitrap-MS/MS, 39 candidate active compounds in the serum of rats treated with WSD were identified. Subsequently, network pharmacology analysis identified 35 overlapping targets between LEE and the active components, and 23 related signaling pathways. Further analysis and molecular docking studies have confirmed that the key active components (rutin, hyperoside and luteoloside) possess potential for effective therapeutic effects with the core targets (PTGS 2 and TNF). Furthermore, in vitro experiments demonstrated that WSD significantly downregulated the expression of PTGS 2 and TNF, thereby validating the network pharmacology findings and providing insights into the potential mechanisms. Results suggested that WSD may exert its therapeutic effects on LEE by modulating the TNF and NF-kappa B signaling pathway, offering a promising approach for the prevention and treatment of LEE.
Collapse
Affiliation(s)
- Ying Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| | - Xiao Dai
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| | - Xianhong Zhu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| | - Weiping Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| | - Chen Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| | - Shiyu Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| | - Shuai Sun
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| | - Xun Gao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| | - Kunming Qin
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Jiangsu Key Laboratory on Innovation for Marine Medicine and Modern Chinese Medicine, Lianyungang, China
| |
Collapse
|
29
|
Ling D, Xiang C, Guolin H, Huisheng S, Xiaohua N. Ellipticine targets FGFR3 to mediate the RAS/MAPK-P38 signalling pathway to induce apoptosis in hepatocellular carcinoma cells. 3 Biotech 2025; 15:111. [PMID: 40191451 PMCID: PMC11968639 DOI: 10.1007/s13205-025-04269-7] [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: 01/07/2025] [Accepted: 03/12/2025] [Indexed: 04/09/2025] Open
Abstract
This study aimed to investigate the toxic effects of ellipticine on liver cancer cells and predict its anti-liver cancer mechanism through network pharmacology, especially by targeting FGFR3 to regulate the RAS/MAPK-P38 signaling pathway, thereby inducing apoptosis of liver cancer cells. The inhibitory effect of ellipticine on the proliferation of HepG2, Huh-7, SMMC7721, BEL-7402, SK-HEP-1, LX-2, and MHCC97H cells was detected by CCK-8 assay, and the IC50 value was calculated. The potential targets of ellipticine were predicted by the database, and the intersection analysis with liver cancer-related targets was performed to construct a protein interaction network (PPI), (KEGG) pathway enrichment analysis, and molecular docking verification. FGFR3 in HepG2 cells was knocked down by siRNA, and the effects on cell proliferation, apoptosis, and ROS levels were observed. The expression changes of FGFR3, RAS, P38, and their phosphorylated forms after ellipticine treatment, as well as the effects of RAS agonist ML-908 and P38 inhibitor PD169316 on cell proliferation, apoptosis, and migration, were detected by Western blotting. Ellipticine has an inhibitory effect on all tested liver cancer cell lines, among which HepG2 has the strongest inhibitory effect, with an IC50 of 5.15 ± 0.25 μM. Ellipticine is predicted to have 32 potential targets, and 5 common targets among the 225 targets related to liver cancer, including PDGFRA, KIT, FGFR3, ERBB2, and STAT3. KEGG analysis showed that these targets are mainly involved in cancer pathways. Molecular docking showed that Ellipticine can bind strongly to FGFR3. FGFR3 expression is highest in HepG2 cells. After knocking down FGFR3, the proliferation ability of HepG2 cells is further weakened, and the addition of apoptosis inhibitor ZVAD can partially restore the proliferation ability. ROS levels increase after Ellipticine treatment, and ROS levels further increase after knocking down FGFR3, and ZVAD treatment can reduce ROS levels. After Ellipticine treatment, the expression levels of FGFR3, RAS, and p-P38 decrease. Ellipticine-induced cell proliferation inhibition and apoptosis were reversed by RAS agonist ML-908, whereas P38 inhibitor PD169316 exacerbated cell apoptosis and migration inhibition. Ellipticine induces apoptosis of liver cancer cells by targeting FGFR3 and inhibiting the RAS/MAPK-P38 signaling pathway. This discovery provides new mechanistic insights into Ellipticine as a liver cancer treatment and may lay the foundation for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Deng Ling
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| | - Chen Xiang
- Department of General Surgery, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| | - Hu Guolin
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| | - Song Huisheng
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| | - Niu Xiaohua
- Department of General Surgery, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
30
|
Zhang W, Wu P, Song Y, Liang D, Meng G, Zeng H. Immunomodulatory mechanism of Huangqi-Guizhi-Wuwu Decoction in alleviating autoimmune arthritis based on network pharmacology and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156644. [PMID: 40339552 DOI: 10.1016/j.phymed.2025.156644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 09/20/2024] [Accepted: 03/11/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) is the most common type of childhood autoimmune arthritis. Huangqi Guizhi Wuwu decoction (HGWD), a traditional Chinese herbal formula, is widely used in China to treat patients with autoimmune arthritis. However, the bioactive ingredients and their complex regulatory mechanisms remain unclear. PURPOSE To investigate the active components of HGWD using a novel comprehensive strategy and clarify the mechanism underlying immunomodulation. METHODS The main active components of HGWD were determined using ultra-high-performance liquid chromatography-high resolution mass spectrometry (UPLCHRMS). The core target and biological immune regulation mechanism of HGWD in alleviating JIA were predicted using combined network pharmacology and molecular docking analyses, followed by in vitro and in vivo experiments. RESULTS A total of 1387 active components were identified by UPLC-MS, of which eight were the main active ingredients. Network pharmacology showed that HGWD acted on core targets, such as STAT3. Further combined analysis revealed that regulation of the Th17 differentiation pathway may be an important mechanism by which HGWD relieves JIA. Molecular docking verification showed that the key component of HGWD can stably bind JAK/STAT-related proteins. The induced differentiation of Th17 and Treg in vitro experiment confirmed the immunoregulatory effects of HGWD. in vivo experiments, HGWD significantly alleviated symptoms of arthritis in a mouse model of collagen-induced arthritis (CIA) and was closely associated with restoring the Th17/Treg balance. CONCLUSION Taken together, serum components/UPLC-MS, network pharmacology, and molecular biology analyses are feasible strategies for exploring the active ingredients in HGWD. This study highlights the clinical potential of HGWD in alleviating JIA and provides evidence of its therapeutic potential through immune regulation.
Collapse
Affiliation(s)
- Wenbo Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Joint Center for Infection and Immunity, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; The Joint Center for Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ping Wu
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yue Song
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dandan Liang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Guangxun Meng
- The Joint Center for Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development and Health, Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Huasong Zeng
- The Joint Center for Infection and Immunity, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; Department of Allergy, Immunology and Rheumatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
31
|
Zhang M, Zhang D, Ren X, Yue S, Sun J, Li N, Bai S, Wang C, Liu C. Study on the pharmacodynamic substances and mechanism of hepatoprotection of Acanthus ilicifolius Linn. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156526. [PMID: 40073778 DOI: 10.1016/j.phymed.2025.156526] [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: 10/17/2024] [Revised: 02/12/2025] [Accepted: 02/15/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The coastal wetland mangrove plant Acanthus ilicifolius l. (AI) is used as traditional medicine for liver protection and liver fibrosis treatment, but the pharmacodynamics of the hepatoprotective substance and the mechanisms of liver protection are not clear. PURPOSE This work aimed to assess the liver-protective ability of AI and elucidate the pharmacodynamics of the hepatoprotective substance of AI responsible for its liver activity. STUDY DESIGN AND METHODS This study first appraised the hepatoprotective activity of the alcohol extract of AI. To identify the hepatoprotective substance in AI, network topology and the contribution index were comprehensively analyzed and screened. The screened medicinal substances, acteoside (ACT) and isoacteoside (IACT), were tested for hepatoprotective activity using mouse liver damage model and l-02 hepatocyte injury model, and metabolomics was employed to explore the mechanism of liver protection. RESULTS AI could restore the biochemical indicators of liver damage induced by CCl4 to normal conditions. The phenylethanoid glycoside compounds ACT and IACT, are the hepatoprotective substances of AI. ACT protects the liver tissue by regulating α-linolenic acid metabolism, glycerophospholipid metabolism, and amino acid-related pathway. CONCLUSION This research provides basic information of the research and development of liver-protective effects of AI and ACT.
Collapse
Affiliation(s)
- Mengqi Zhang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan 250100, PR China
| | - Dahu Zhang
- Shandong bigtree Life Health Technology Co., LTD, 5509 Huanghe West Road, Heze 274000, PR China
| | - Xia Ren
- Marine traditional Chinese medicine research center, Qingdao Academy Shandong University of Traditional Chinese Medicine, Qingdao 266114, PR China
| | - Shijun Yue
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, PR China
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan 250100, PR China
| | - Ningyang Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, PR China
| | - Shujin Bai
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, PR China.
| | - Changyun Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, PR China.
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan 250100, PR China.
| |
Collapse
|
32
|
Liu X, Zuo H, Wu C, Li N, Zhou Q, Cao F, Chu B, Zeng S, Feng H, Wang Y, Lei F, Hu K, Hou S. Muscone Attenuates Uveitis Through the PI3K/AKT Signaling Pathway. Invest Ophthalmol Vis Sci 2025; 66:21. [PMID: 40341311 PMCID: PMC12068523 DOI: 10.1167/iovs.66.5.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/15/2025] [Indexed: 05/10/2025] Open
Abstract
Purpose Uveitis is an immune-mediated ocular disorder that poses a significant threat to vision, particularly among young and middle-aged adults. The treatment of uveitis is complicated by the presence of the blood-retinal barrier (BRB), which restricts the passage of large molecular drugs into the eye, thus limiting effective therapeutic options. The primary objective of this study is to identify a novel therapeutic agent capable of treating uveitis and explore its underlying mechanism. Methods In this study, we used a mouse model of experimental autoimmune uveitis (EAU) induced by interphotoreceptor retinoid-binding protein (IRBP) and lipopolysaccharide (LPS) and interferon-gamma (IFN-γ)-induced inflammatory BV2 cells. Evans blue and fundus fluorescein angiography (FFA) experiments were performed to evaluate the destruction of BRB. Silt lamp and hematoxylin and eosin (H&E) staining were conducted to evaluate the inflammatory response. In vivo proteomics and Western blot were carried to investigate the underlying mechanisms. Results Our study reveals that Muscone significantly alleviates EAU and restores the integrity of BRB. Moreover, Muscone treatment markedly downregulated inflammatory factors within the retinas and BV2 cells. In vivo proteomic combined with liquid chromatography-mass spectrometry (LC-MS) has elucidated that Muscone exerts its anti-inflammatory effects by modulating the PI3K-AKT signaling pathway. Moreover, by using LY294002 to specifically inhibit PI3K, we observed a marked decrease in inflammatory phenotype and BRB destruction of EAU. Conclusions In summary, this study establishes the protective efficacy of Muscone against the progression of EAU and provides insights into the molecular mechanisms responsible for its therapeutic action.
Collapse
Affiliation(s)
- Xianyang Liu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hangjia Zuo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| | - Na Li
- Department of Laboratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qian Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Cao
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| | - Baorui Chu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| | - Shuhao Zeng
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Feng
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| | - Yakun Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fengyang Lei
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengping Hou
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| |
Collapse
|
33
|
Du T, Yang J, Qin Y, Huang X, Li J, Xiong S, Xu X, Zhang L, Zhao M, Li H, Huang T, Xiong T, Xie M. Transport and action of sesame protein-derived ACE inhibitory peptides ITAPHW and IRPNGL. Food Chem 2025; 472:142965. [PMID: 39842202 DOI: 10.1016/j.foodchem.2025.142965] [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: 08/20/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
Vascular endothelial dysfunction is an important pathogenic factor in hypertension, in which angiotensin-converting enzyme (ACE) plays an important role. Peptides that bind to ACE may attenuate vascular endothelial dysfunction by altering the structure of ACE. This study demonstrated that ITAPHW and IRPNGL were resistant to simulated gastrointestinal fluid and were transported across the Caco-2 monolayer via the intercellular space, with ITAPHW showing a high apparent permeability coefficient of (1.44 ± 0.01) × 10-5 cm/s. Subsequently, multispectral analysis and molecular dynamic simulation revealed the stability, conformation changes, and potential binding sites of ITAPHW- and IRPNGL-ACE complex. Furthermore, ITAPHW and IRPNGL alleviated endothelial dysfunction in the angiotensin I-induced human umbilical vein endothelial cells (HUVECs) by reducing ACE activity and the concentrations of angiotensin II and endothelin-1 (ET-1), while promoting the level of nitric oxide (NO), endothelial nitric oxide synthase (eNOS), cyclic guanosine 3', 5'-monophosphate (cGMP), and ACE2.
Collapse
Affiliation(s)
- Tonghao Du
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Jiahui Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Yuan Qin
- Party Committee Office for Faculty Affairs, Jiangxi Vocational Technical College of Industry & Trade, No. 699 Jiayan Road, Nanchang, Jiangxi, 330038, PR China
| | - Xizhuo Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Jiahui Li
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Shijin Xiong
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Xiaoyan Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Linli Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Mingwei Zhao
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Huiyu Li
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Tao Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Tao Xiong
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; Jiangxi Academy of Nutrition and Health Management Medicine, The First Affiliated Hospital of Nanchang University, No. 1519 Dongyue Avenue, Nanchang, Jiangxi 330209, PR China.
| | - Mingyong Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| |
Collapse
|
34
|
Zhai Y, Fu J, Yang J, Zhou Y. Quercetin in Shengxian Decoction exhibits anti-ferroptosis protective roles in a myocardial infarction model via targeting DPP4/ HMOX1, based on network pharmacology and molecular docking. Front Pharmacol 2025; 16:1583509. [PMID: 40365322 PMCID: PMC12069271 DOI: 10.3389/fphar.2025.1583509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Background Myocardial infarction (MI) is characterized by high morbidity. In this study, we aimed to elucidate potential targets of Shengxian Decoction (SXD) against MI. Methods Pairing of SXD active ingredients and MI targets was conducted using the Chinese Medicine System Pharmacological Database, Gene Expression Omnibus (GEO), and STRING databases. The effects of SXD on MI were validated in vitro. Molecular docking was verified using cellular thermal shift assay (CETSA). Results A total of 40 active ingredients and 28 MI-related targets were obtained. Cross-analysis on 28 targets and cell death-related genes identified two crucial ferroptosis-related targets, namely, dipeptidyl peptidase 4 (DPP4) and heme oxygenase 1 (HMOX1). In cobalt chloride (CoCl2)-induced hypoxic H9c2 cells, SXD could remarkably improve cell viability and inhibit cell death. Meanwhile, SXD treatment significantly affected the ferroptosis-related markers in hypoxic H9c2 cells. Molecular docking and CETSA results showed that quercetin had good binding activity with DPP4 and HMOX1. Conclusion Important active ingredient quercetin in SXD could exert anti-ferroptosis protective roles on MI through targeting ferroptosis-related genes (DPP4/HMOX1), thereby contributing to the protective role of SXD on MI.
Collapse
Affiliation(s)
- Yuming Zhai
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jiamei Fu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jianfei Yang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yabin Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
35
|
Singh P, Borkar M, Doshi G. Network pharmacology approach to unravel the neuroprotective potential of natural products: a narrative review. Mol Divers 2025:10.1007/s11030-025-11198-3. [PMID: 40279084 DOI: 10.1007/s11030-025-11198-3] [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: 12/22/2024] [Accepted: 04/13/2025] [Indexed: 04/26/2025]
Abstract
Aging is a slow and irreversible biological process leading to decreased cell and tissue functions with higher risks of multiple age-related diseases, including neurodegenerative diseases. It is widely accepted that aging represents the leading risk factor for neurodegeneration. The pathogenesis of these diseases involves complex interactions of genetic mutations, environmental factors, oxidative stress, neuroinflammation, and mitochondrial dysfunction, which complicate treatment with traditional mono-targeted therapies. Network pharmacology can help identify potential gene or protein targets related to neurodegenerative diseases. Integrating advanced molecular profiling technologies and computer-aided drug design further enhances the potential of network pharmacology, enabling the identification of biomarkers and therapeutic targets, thus paving the way for precision medicine in neurodegenerative diseases. This review article delves into the application of network pharmacology in understanding and treating neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and spinal muscular atrophy. Overall, this article emphasizes the importance of addressing aging as a central factor in developing effective disease-modifying therapies, highlighting how network pharmacology can unravel the complex biological networks associated with aging and pave the way for personalized medical strategies.
Collapse
Affiliation(s)
- Pankaj Singh
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai Campus, V. M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Maheshkumar Borkar
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V. M. Road, Vile Parle (W), Mumbai, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai Campus, V. M. Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
36
|
Zhai Y, Pang X, Mei X, Pang Y, Shu J, Xiao Y, Ma W, Zou M, Yang P, Yue G, Lan D. Shuanglu tongnao formula alleviates cerebral ischemia/reperfusion injury by rebuilding inflammatory microenvironment after cerebral ischemia. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119640. [PMID: 40107474 DOI: 10.1016/j.jep.2025.119640] [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: 12/06/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
ETHNOPHARMACOLOGICAL SIGNIFICANCE Shuanglu tongnao formula (SLTNF) has been clinically proven to have significant efficacy in the treatment of Ischemic stroke (IS), and is a promising formula for IS treatment. Still, the underlying mechanism is not clear. Whether SLTNF ameliorates ischemic brain injury by reversing the pro-inflammatory microenvironment after IS is an interesting field of investigation. AIM OF THE STUDY Based on the result of network pharmacology and single-cell RNA sequencing (scRNA-seq), whether SLTNF mitigates cerebral ischemia/reperfusion (I/R) injury by reversing the pro-inflammatory microenvironment was investigated in vivo for the first time. MATERIALS AND METHODS The mice middle cerebral artery occlusion (MCAO) model was established to induce focal cerebral I/R. Subsequently, the remission effects of SLTNF treatment for cerebral I/R injury were evaluated in the MCAO model. scRNA-seq data was used to analyze the immune microenvironment after IS in mice. scRNA-seq and Network pharmacology were applied to predict the mechanism of the treatment of IS by SLTNF. Western blot (WB) and immunofluorescence techniques were employed to validate the potential mechanism. RESULTS The experimental results demonstrated that SLTNF dosage-dependently attenuated the infarct volume, neurobehavioral, cell morphology and Nissl bodies damage, and inhibited the apoptosis in cerebral I/R mice. Moreover, scRNA-seq results revealed that the number of NK cells, neutrophils, monocytes, astrocytes and microglia significantly increased after IS. The cell-cell interactions dominated by microglia after IS, the cell-cell interactions between microglia and other immune cells significantly heightened. Furthermore, SLTNF promoted the transition of M1 microglia to M2 type, eventually reversing the pro-inflammatory microenvironment. Combined analysis of scRNA-seq and Network pharmacology results predicted that AGE-RAGE signaling pathway could involve in the regulation of microglia polarization by SLTNF. WB results revealed that SLTNF significantly inhibited the protein expression of CCND1, IL-1β and p-STAT3, which belong to crucial targets of SLTNF and AGE-RAGE signaling pathway. CONCLUSION SLTNF attenuated cerebral I/R injury by reversing the pro-inflammatory microenvironment via the AGE-RAGE signaling pathway in mice.
Collapse
Affiliation(s)
- Yang Zhai
- Department of Traditional Chinese Medicine, Nanning Seventh People's Hospital, Nanning, 530000, China
| | - Xingwang Pang
- Department of Traditional Chinese Medicine, Nanning Seventh People's Hospital, Nanning, 530000, China
| | - Xiaoping Mei
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China
| | - Yan Pang
- Department of Emergency, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, 530000, China
| | - Jianlong Shu
- Department of Traditional Chinese Medicine, Nanning Seventh People's Hospital, Nanning, 530000, China
| | - Yuhan Xiao
- Department of Traditional Chinese Medicine, Nanning Seventh People's Hospital, Nanning, 530000, China
| | - Wei Ma
- Neurology Department, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Min Zou
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China
| | - Peng Yang
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China
| | - Guihua Yue
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China.
| | - Dazhi Lan
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China.
| |
Collapse
|
37
|
Huang X, Pan Z, Shen L, Chen H, Chen C, Lv T, Mei Y. The mechanism of Weiqi decoction treating gastric cancer: a work based on network pharmacology and experimental verification. Hereditas 2025; 162:67. [PMID: 40259338 PMCID: PMC12012975 DOI: 10.1186/s41065-025-00434-3] [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: 12/23/2024] [Accepted: 04/08/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Weiqi Decoction (WQD) is an empirical prescription traditionally used in China for the treatment of precancerous gastric cancer (GC) lesions. This study aimed to elucidate the potential pharmacological mechanisms of WQD in GC therapy. METHODS Active ingredients, corresponding targets, and GC-related genes were identified using public databases. A protein-protein interaction (PPI) network was constructed via the STRING database, and functional enrichment analyses were conducted using the DAVID platform. Gene expression and survival analyses were performed using the GEPIA database. Molecular docking was conducted with AutoDock Vina and visualized using PyMOL. The effects of WQD on GC cell viability, proliferation, migration, and invasion were evaluated through CCK-8, colony formation, and Transwell assays. RESULTS WQD contained 43 active ingredients targeting 751 potential genes, including 458 GC-related targets. Quercetin, luteolin, and kaempferol were identified as key active compounds. PPI network analysis revealed nine core targets, including TP53 and SRC, which may mediate the anti-GC effects of WQD. GO enrichment analysis indicated involvement in 726 biological processes, 91 cellular components, and 177 molecular functions, while KEGG pathway analysis suggested modulation of the AGE-RAGE, PI3K-Akt, and HIF-1 signaling pathways. GEPIA database analysis confirmed that EP300, HSP90AA1, HSP90AB1, SRC, and TP53 were highly expressed in GC. Molecular docking demonstrated strong binding affinities between the key active compounds and core targets. In vitro experiments further validated that WQD extract inhibited GC cell viability, proliferation, migration, and invasion. CONCLUSION WQD exhibits therapeutic potential against GC by regulating multiple targets and signaling pathways. These findings provide mechanistic insights into the pharmacological actions of WQD in GC treatment.
Collapse
Affiliation(s)
- Xu Huang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China
| | - Zhihong Pan
- Department of Gastroenterology, The First College of Clinical Medical Science, China Three Gorges University, Jiefang Road No. 2, Xiling District, Yichang, Hubei, 443000, P.R. China
| | - Lei Shen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P.R. China.
| | - Huan Chen
- Department of Gastroenterology, The First College of Clinical Medical Science, China Three Gorges University, Jiefang Road No. 2, Xiling District, Yichang, Hubei, 443000, P.R. China
| | - Chang Chen
- Department of Gastroenterology, The First College of Clinical Medical Science, China Three Gorges University, Jiefang Road No. 2, Xiling District, Yichang, Hubei, 443000, P.R. China
| | - Tingting Lv
- Department of Gastroenterology, The First College of Clinical Medical Science, China Three Gorges University, Jiefang Road No. 2, Xiling District, Yichang, Hubei, 443000, P.R. China
| | - Yuzhou Mei
- Department of Gastroenterology, The First College of Clinical Medical Science, China Three Gorges University, Jiefang Road No. 2, Xiling District, Yichang, Hubei, 443000, P.R. China
| |
Collapse
|
38
|
Shi Y, Zhang Y, Cai Y, Yan Z, Jing Y, Zhu Y, Li H, Zheng G, Chen J, Yang X, Huang Y, Gan Y, Han Y, Yong H, Xiong Q. Key proteins identification of Buyang Huanwu Decoction in regulating macrophage polarization to alleviate atherosclerosis via an integrated strategy combining network pharmacology, batch molecular docking and experimental verification. Int J Biol Macromol 2025; 310:143380. [PMID: 40258538 DOI: 10.1016/j.ijbiomac.2025.143380] [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: 01/08/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025]
Abstract
Buyang Huanwu Decoction (BYHWD) effectively treats atherosclerosis (AS), but its unclear mechanism limits clinical use. Traditional mechanism research strategies overlook BYHWD's multi-target synergy, while conventional network pharmacology neglects its organelle-level Supplement Qi function. The purpose of this paper was to decipher the mechanism of BYHWD in regulating macrophage polarization to mitigate AS via a novel integrated strategies combining network pharmacology, batch molecular docking, and experimental verification. Firstly, BYHWD was confirmed to significantly improve macrophage polarization to reduce AS. Then, using network pharmacological analysis, TP53, AKT1, and BCL2 were identified as core targets of BYHWD in regulation of macrophage polarization to alleviate AS and its key biological processes was found be mitochondrial function and metabolism. Meanwhile, main material basis of BYHWD was clarified by batch molecular docking as 3-O-p-coumaroylquinic acid, D-mandelonitrile, Ellagic acid, Ferulic acid, 5-hydroxy-L-tryptophan zwitterion, Isoliquiritigenin, Senkyunolide-F, Anofinic acid, Trimethylhydroquinone and Senkyunolide-E. On this basis, in vitro experiments confirmed that TP53, AKT1, and BCL2 were critical targets in BYHWD that restore mitochondrial function and regulate macrophage polarization to alleviate AS. This study clarified BYHWD's mechanism and offered a novel approach for studying traditional Chinese medicine. Future work should advance clinical applications and develop BYHWD-based therapies targeting macrophage polarization in AS.
Collapse
Affiliation(s)
- Yingying Shi
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Yuhan Zhang
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Yisa Cai
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Zhimin Yan
- Department of pharmacy, Huai'an Hospital of Traditional Chinese Medicine (Affiliated Hospital of Nanjing University of Traditional Chinese Medicine), Huai'an 223002, Jiangsu, PR China
| | - Yi Jing
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Yong Zhu
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Heng Li
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Guangzhen Zheng
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Jie Chen
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Xiaoyao Yang
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Yi Huang
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China
| | - Yimin Gan
- Department of Rehabilitation, Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223002, Jiangsu, PR China.
| | - Yun Han
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, Shandong, PR China.
| | - Hui Yong
- Department of Cardiology, Huai'an Hospital Affiliated to Yangzhou University(The Fifth People's Hospital of Huai'an), Huai'an 223000, Jiangsu, PR China.
| | - Qingping Xiong
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, PR China.
| |
Collapse
|
39
|
Bi A, Liu R, Xie M, He B, Yan T, Du Y, Jia Y. Semen Ziziphi Spinosae alleviates cardiomyocyte apoptosis in rats with coronary heart disease via the AMPK/SIRT1/PGC-1α signaling pathway activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156743. [PMID: 40250033 DOI: 10.1016/j.phymed.2025.156743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/23/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Coronary heart disease (CHD) represents a significant cardiovascular condition, with its occurrence increasing as a result of alterations in lifestyle and dietary habits. Semen Ziziphi Spinosae (SZS) is commonly utilized for the management of disorders associated with the nervous system, including conditions like depression and insomnia. Recent research has revealed its potential therapeutic properties for cardiovascular issues. Nevertheless, there exists a limited amount of research addressing the mechanisms involved. PURPOSE This research seeks to explore the protective effects that SZS has on cardiac tissue, specifically within the framework of CHD. By conducting this investigation, the study aims to uncover the various mechanisms that play a role in these protective effects. This understanding could yield significant insights into how SZS may result in the preservation and enhancement of cardiac health in patients affected by CHD. STUDY DESIGN The study innovatively combines multiple advanced techniques. It first integrates UPLC-Q-TOF/MS analysis and network pharmacology to identify SZS components. In vitro experiments were conducted using H9c2 rat cardiomyocytes, and in vivo experiments used a CHD model in SD rats. Multiple assays were performed for multi - level and multi - dimensional validation. METHODS In the initial stage, the primary components of SZS and their possible mechanisms for combating CHD were examined through UPLC-Q-TOF/MS analysis in conjunction with network pharmacology approaches. For the in vitro investigation, an ischemia-hypoxia model was established utilizing H9c2 rat cardiomyocytes. The CCK-8 assay was used to assess myocardial injury markers. TUNEL staining and Western blot techniques were employed to confirm the impact of SZS treatment on apoptosis in H9c2 cells. The expression levels of proteins associated with the AMPK/SIRT1/PGC-1α signaling pathway were measured using RT-qPCR and Western blotting, and the results were validated with the AMPK inhibitor, compound C. In the in vivo segment, a model of coronary heart disease (CHD) in SD rats was established through the administration of a high-fat emulsion diet combined with pituitrin injections. Cardiac function in the rats was evaluated through electrocardiograms and echocardiograms. Pathological changes in the heart were observed utilizing TTC and H&E staining. Kits were implemented to measure the serum biochemical indicators in the rats.RT - qPCR and Western blotting were employed to measure the expression levels of proteins related to the AMPK/SIRT1/PGC - 1α signaling pathway. RESULTS The study identified 67 in vitro components, 27 blood - absorbed components, and 12 metabolic components of SZS. Network pharmacology analysis suggested the AMPK/SIRT1/PGC - 1α signaling pathway as a key mechanism. In vitro and in vivo experiments showed that SZS increased cell viability, reduced apoptosis, and activated the AMPK/SIRT1/PGC - 1α signaling pathway. Inhibiting AMPK abolished SZS's effects. SZS also improved cardiac function and reduced myocardial damage in rats with CHD. CONCLUSION This study for the first time highlights that Semen Ziziphi Spinosae plays a beneficial role in cardiovascular health by activating the AMPK/SIRT1/PGC-1α signaling pathway and reducing apoptosis in cardiomyocytes. These findings support its potential application in the treatment of CHD and other cardiac conditions.
Collapse
Affiliation(s)
- Anqi Bi
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Rihong Liu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Min Xie
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Bosai He
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Yiyang Du
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China.
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China.
| |
Collapse
|
40
|
Pan D, Xia J, Qu Y, Shi C, Du H, Zhang J, Peng F, Wang X, Liu R, Wu L, Hong Q, Chen X. Oleanolic acid inhibits mesangial cell proliferation and inflammatory response in mesangial proliferative glomerulonephritis through IL-17/ERK/AKT pathway. Int Immunopharmacol 2025; 152:114459. [PMID: 40088868 DOI: 10.1016/j.intimp.2025.114459] [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: 02/09/2024] [Revised: 02/13/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Mesangial proliferative glomerulonephritis (MsPGN) is a common form of glomerulonephritis characterized by mesangial cell proliferation and inflammatory responses. However, current clinical treatment options for MsPGN are rather limited. Oleanolic acid (OA), a natural pentacyclic triterpenoid compound, exhibits anti-tumor and anti-inflammatory properties and has been proven to have renal protective effects. We speculate that OA could potentially serve as an alternative therapy for MsPGN. OBJECTIVE This study aimed to investigate the therapeutic efficacy and mechanism of OA against MsPGN. METHODS Tail vein injection of anti-Thy1 antibody was used to establish the MsPGN model, followed by a comprehensive assessment of the effects of OA on renal function, histopathological changes, and inflammatory responses in anti-Thy1 nephritis rats. Subsequently, network pharmacology was employed to predict the key targets and pathways of OA in treating MsPGN. Finally, in vivo and in vitro experiments were conducted to validate the results of network pharmacology. RESULTS OA significantly improved renal function, and attenuated mesangial cell proliferation and inflammatory reactions in anti-Thy1 nephritis rats. Network pharmacology analysis identified TNF-α, IL-6, IL-1β, MAPK3, and AKT1 as key targets of OA in the treatment of MsPGN, and involved the IL-17 signaling pathway. Additionally, we observed increased phosphorylation levels of ERK and AKT, as well as activation of downstream inflammatory responses, in both anti-Thy1 nephritis rats and mesangial cells stimulated with IL-17. In contrast, treatment with OA and the ERK inhibitor PD98059 reversed these effects. Furthermore, we identified IL17RA within this pathway as a potential target of OA. CONCLUSIONS Our study demonstrates that OA can modulate the IL-17/ERK/AKT signaling pathway, thereby improving anti-Thy1 antibody-induced MsPGN. This establishes a theoretical basis for OA to potentially serve as a therapeutic agent for treating MsPGN.
Collapse
Affiliation(s)
- Dan Pan
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; The College Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jikai Xia
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Yilun Qu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Chunru Shi
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; The College Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hongjian Du
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Jie Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; School of Medicine, Nankai University, Tianjin, 300191, China
| | - Fei Peng
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; School of Medicine, Nankai University, Tianjin, 300191, China.
| | - Xu Wang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Ran Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Lingling Wu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China.
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China.
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; The College Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
41
|
Zhang A, Kong L, Li T, Shi L, Jiang J, Li P, Li H, Han X, Li J, Feng S, Li F, Meng Z. Studying the efficacy of JBOL volatile components in idiopathic pulmonary fibrosis (IPF) using GC-MS and network pharmacology. Sci Rep 2025; 15:13188. [PMID: 40240792 PMCID: PMC12003768 DOI: 10.1038/s41598-025-97374-9] [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: 12/04/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Jin Bei oral liquid (JBOL) is a Chinese medicinal preparation for the treatment of idiopathic pulmonary fibrosis (IPF), Clinical trials have shown that IPF patients using JBOL have improved their lung function indicators FVC% and DLCO% by approximately 2.10% and 7.74%, suggesting that the agent has a positive effect in slowing disease progression. In this study, the active volatile components of JBOL were systematically identified and analyzed using gas chromatography-mass spectrometry (GC-MS), network pharmacology and molecular docking techniques. It was found that JBOL contains a variety of compounds with antifibrotic potential, which act through multi-target and multi-pathway mechanisms. Network pharmacological analyses revealed multiple targets of JBOL associated with key pathological processes in IPF, and key active ingredients were screened based on degree values (including Sedanolide, Ligustilide, Senkyunolide H, Senkyunolide I, α-Terpineol, and 4-Terpineol). Molecular docking results showed that these compounds have high affinity for target proteins. Finally, suitable quantitative methods were established and methodologically validated for these six compounds, and these methods were used to determine the content of 8 batches of JBOL and analyze the differences in content between batches.The present study provides a scientific basis for the quality control and standardization of its JBOL by identifying and analyzing its active volatile components.
Collapse
Affiliation(s)
- Aijun Zhang
- College of pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Shandong Hongji-tang Pharmaceutical Group Co., Ltd., Jinan, 250301, China
| | - Linghui Kong
- College of pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tong Li
- Shandong Hongji-tang Pharmaceutical Group Co., Ltd., Jinan, 250301, China
| | - Lei Shi
- College of pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Juanjuan Jiang
- Shandong Hongji-tang Pharmaceutical Group Co., Ltd., Jinan, 250301, China
| | - Ping Li
- Shandong Hongji-tang Pharmaceutical Group Co., Ltd., Jinan, 250301, China
| | - Honglin Li
- Shandong Hongji-tang Pharmaceutical Group Co., Ltd., Jinan, 250301, China
| | - Xinru Han
- College of pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jian Li
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Shuai Feng
- College of pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Feng Li
- College of pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Zhaoqing Meng
- Shandong Hongji-tang Pharmaceutical Group Co., Ltd., Jinan, 250301, China.
| |
Collapse
|
42
|
Yuan Y, Chu G, Ma Q, Liang Z, Liang Y, Niu H. Multiscale screening and identifying specific targets for artesunate in suppressing bladder cancer. Front Pharmacol 2025; 16:1584502. [PMID: 40303931 PMCID: PMC12037512 DOI: 10.3389/fphar.2025.1584502] [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: 02/27/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Background Bladder cancer (BLCA) is a highly aggressive urinary malignancy with high mortality in advanced stages, posing a significant health risk. Artesunate (ART), a derivative of artemisinin, has been demonstrated with potent anti-tumor activity in some studies, yet its specific targets for BLCA and the molecular mechanisms have not been fully elucidated. Purpose This study screened potential targets of ART against BLCA through network pharmacology, followed by molecular docking simulations and experimental validation in vitro and in vivo to elucidate the underlying mechanisms. Methods This study identified the critical targets of BLCA and ART by employing multiscale screening from public databases, and a protein-protein interaction (PPI) network was constructed. Molecular docking simulations confirmed the stable binding of ART to the identified tumor-related targets promoting BLCA progression. These computational findings were further validated through experiments in vivo and in vitro, ensuring robust and reliable results. Results Based on network pharmacology analysis, the effects of ART on BLCA were multifaceted. Molecular docking simulations confirmed the binding stability of ART with core targets. The experiments in vitro proved that ART could inhibit BLCA cell proliferation and migration by downregulating the expression of BCL-2, inducing Caspase 3-mediated apoptosis, resulting in cell cycle arrest and suppressing the PI3K/Akt/mTOR classical pathway involved in BLCA growth and metabolism. Studies in vivo also confirmed that ART had significant anti-tumor effects with minimal side effects. Conclusion This study identified the mechanism by which ART inhibited BLCA through multiple specific targets, revealing its potential anti-cancer pathways and laying the foundation for the clinical application of traditional Chinese medicine in BLCA therapy.
Collapse
Affiliation(s)
- Yi Yuan
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guangdi Chu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qingyue Ma
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhijuan Liang
- Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ye Liang
- Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haitao Niu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Wu Y, Guo F, Liu Y, Li J, Shi W, Song L, Wang G, Liu J. Curcumin mitigates heatstroke-induced myocardial injury by modulating the Akt/Bad/Caspase-3 pathway. Biochem Biophys Res Commun 2025; 758:151653. [PMID: 40112539 DOI: 10.1016/j.bbrc.2025.151653] [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: 12/09/2024] [Revised: 03/01/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Heatstroke (HS) presents a major health threat, especially during summer, and is linked to myocardial injury and persistent cardiovascular complications.Curcumin has shown promise in treating myocardial damage, but its mechanisms in HS-induced myocardial damage remain unclear. We integrated curcumin targets from BATMAN-TCM, DGIdb, and PharmMapper, and identified HS-related targets from GeneCards and OMIM. The intersection of these targets was identified using Venn diagrams, and subsequently analyzed through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis.Protein interactions were analyzed using STRING and visualized in Cytoscape to screen core proteins. Molecular docking was performed with these proteins and curcumin. HS mouse model was constructed for pathological assessments and WB validation of core protein expression. We identified 132 potential therapeutic targets and selected AKT1, Bad, and CASP3 as our targets for validation. Molecular docking indicated that these proteins all have good affinity with curcumin. In HS mouse model, we observed that HS led to significant myocardial cell edema, disordered arrangement, and pronounced mitochondrial swelling accompanied by the destruction of cristae. The application of curcumin effectively alleviated myocardial cell edema and the degree of mitochondrial swelling. WB revealed that HS decreased p-Akt and p-Bad while increasing cleaved-caspase-3. Curcumin treatment reversed these effects, inhibiting HS-induced myocardial cell apoptosis. Our research demonstrates that curcumin effectively safeguards against HS-induced myocardial injury in mice, potentially through the modulation of the Akt/Bad/caspase-3 pathway.
Collapse
Affiliation(s)
- Yizhan Wu
- Department of Graduate School, Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China; Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Fei Guo
- Department of Emergency Trauma Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang Uygur Autonomous Region, China
| | - Ya Liu
- Department of Rehabilitation, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jiajia Li
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Wenhui Shi
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Laiyang Song
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Guangjun Wang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
44
|
Zhang X, Cai Y, Chen M, Chen L, Mao Y, He R, Yang P, Xu M, Yan H, Zhao Q. Danshen-Chuanxiong-Honghua ameliorates neurological function and inflammation in traumatic brain injury in rats via modulating Ghrelin/GHSR. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119625. [PMID: 40074098 DOI: 10.1016/j.jep.2025.119625] [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: 11/09/2024] [Revised: 12/19/2024] [Accepted: 03/10/2025] [Indexed: 03/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Guanxin II, proposed by Chen Keji (National master of traditional Chinese medicine), possesses neuroprotective effect. Interestingly, its simplified prescription Danshen-Chuanxiong-Honghua (DCH) can also clinically ameliorate cerebral impairment and improve spatial cognitive deficits, similar to the function of original formula. AIM OF THE STUDY We aimed to elucidate the rationality of DCH's natural existence, qualitatively identify DCH-derived phytochemicals, thereby to validate cerebral protective effect, and expose the potential mechanism of DCH and its main absorbed compound ferulic acid (FA). MATERIALS AND METHODS The natural rationality of DCH's existence for treating TBI was verified using data mining. The qualitative analysis of DCH extract-derived phytochemicals was conducted through liquid chromatography with mass spectrometry (LC-MS). Controlled cortical impact (CCI) was chosen to establish TBI model. Neurological behavior tests, blood-brain barrier (BBB) permeability test, brain water content measurement, and proinflammatory factors consisting of IL-6, IL-1β, and TNF-α of plasma, and HPA axis-related hormone levels of DA, NA, 5-HT, ghrelin, and BDNF in hippocampus were analyzed by enzyme-linked immunosorbent assay. Network pharmacology was employed to predict potential targets and pathways of DCH intervening TBI. Growth hormone secretagogue receptor (GHSR) antagonist [D-Lys3]-GHRP-6 (D-Lys3) was injected intraperitoneally in TBI rats after waking up. Molecular docking and pharmacological experiment with D-Lys3 were used to verify the pathway. RESULTS Twenty-six phytochemicals were identified based on LC-MS. FA, as the primary contributor of DCH, alleviated disruption of BBB and reduced brain edema, suppressed the secretion of proinflammatory factors, such as IL-6, IL-1β, TNF-α, as well as HPA axis-related hormones such as DA, NA, and 5-HT, and ghrelin, and BDNF by regulating the Ghrelin/GHSR pathway. These results were validated by GHSR receptor antagonist, as well as molecule docking. CONCLUSIONS Taken together, DCH, when prescribed for the treatment of TBI, has a certain degree of reasonableness. FA, as the main absorbed component, demonstrated a similar function to DCH in improving the blood-brain barrier, promoting neural recovery, and anti-inflammatory effects in TBI rats, primarily via modulating Ghrelin/GHSR.
Collapse
Affiliation(s)
- Xiaohang Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yawen Cai
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Meng Chen
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Li Chen
- Hukou County Chinese Medicine Hospital, Jiujiang, 332500, China
| | - Yaqing Mao
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Runtian He
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Peishan Yang
- Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Min Xu
- Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Hui Yan
- Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Qiulong Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518104, China.
| |
Collapse
|
45
|
Zheng J, Jiao Z, Yang X, Ruan Q, Huang Y, Jin C, Gui S, Xuan Z, Jia X. Network pharmacology-based exploration of the mechanism of Wenweishu granule in treating chronic atrophic gastritis with spleen-stomach cold deficiency syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119591. [PMID: 40054637 DOI: 10.1016/j.jep.2025.119591] [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: 12/12/2024] [Revised: 02/14/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wenweishu (WWS) is a traditional Chinese medicine compound formulated for chronic atrophic gastritis (CAG) treatment by warming the stomach and alleviating pain. However, its pharmacological mechanisms remain underexplored. AIM OF THE STUDY This study investigated the therapeutic effects and potential mechanisms of WWS on CAG with spleen-stomach cold deficiency syndrome (SSCDS). METHODS To achieve this, an SSCDS-CAG rat model and a human gastric mucosal epithelial cells (GES-1) cell model were established using multi-factor modeling and N-Methyl-N'-nitro-N-nitrosoguanidine (MNNG) induction, respectively. WWS's effects on gastric injury were evaluated through pathology, inflammation, serum biomarkers, and apoptosis. Additionally, MNNG's effects on GES-1 cells were analyzed. Network pharmacology, involving protein-protein interaction networks, GO/KEGG enrichment, and molecular docking, was employed to predict WWS's potential targets and mechanisms in SSCDS-CAG. Mechanistic insights were further validated using immunohistochemistry, quantitative reverse transcription polymerase chain reaction, and western blotting. RESULTS In vivo results showed that WWS alleviated symptoms in SSCDS-CAG rats, lowering symptom scores and improving gastric histopathology. It modulated serum biomarkers and reduced inflammation and apoptosis in both in vivo and in vitro studies. Network pharmacology results revealed 263 overlapping targets between WWS and SSCDS-CAG, associated with apoptosis, inflammation, and the PI3K/AKT pathway. Molecular docking revealed strong binding affinity between the core target and active WWS components. In SSCDS-CAG rats and GES-1 cells, WWS inhibited PI3K/AKT phosphorylation, increased PTEN expression, and regulated Bcl-2, Bax, and cleaved caspase-3 levels. CONCLUSION WWS reduces inflammation and apoptosis in multi-factor CAG rats and MNNG-induced GES-1 cells by modulating the PTEN/PI3K/AKT signaling pathway and apoptosis-related proteins.
Collapse
Affiliation(s)
- Jia Zheng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Zhiyong Jiao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Xinyu Yang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Qing Ruan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Yuzhe Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Cheng Jin
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Shuangying Gui
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Zihua Xuan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Xiaoyi Jia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China.
| |
Collapse
|
46
|
Du H, Yang K, Yang J, Wan J, Pan Y, Song W, Xu S, Chen C, Li J. Euphorbia humifusa Willd. ex Schltdl. Mitigates Liver Injury via KEAP1-NFE2L2-Mediated Ferroptosis Regulation: Network Pharmacology and Experimental Validation. Vet Sci 2025; 12:350. [PMID: 40284852 PMCID: PMC12030869 DOI: 10.3390/vetsci12040350] [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: 03/07/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Liver injury poses major health risks in livestock, necessitating effective therapeutic interventions. This study elucidates the hepatoprotective mechanisms of Euphorbia humifusa Willd. ex Schltdl. (EHW) by integrating network pharmacology, molecular docking, and experimental validation. Using a CCl4-induced liver injury model mimicking veterinary clinical scenarios, EHW markedly alleviated hepatic damage, demonstrated by reduced liver index, serum ALT and AST levels, histopathological lesions, iron accumulation, inflammatory cytokines, and ferroptosis-associated gene expression. Network pharmacology identified EHW's core bioactive components (quercetin, kaempferol, and β-sitosterol) and critical targets (IL-6, STAT3, HIF-1α, PTGS2, NFE2L2, and KEAP1) which were linked to ferroptosis and oxidative stress. Molecular docking revealed robust binding affinities between these compounds and ferroptosis-related proteins. In vivo validation confirmed that EHW inhibited KEAP1, activated NFE2L2-mediated antioxidant defenses (upregulating SOD1 and NQO1), restored iron homeostasis (lowering TFR1, elevating FTH1), and attenuated phospholipid peroxidation by suppressing ACSL4 and ALOX12. These results indicate that EHW mitigates ferroptosis-driven liver injury via KEAP1-NFE2L2 signaling to restore iron homeostasis and reduce oxidative stress, offering a mechanistic foundation for its clinical application in veterinary hepatoprotection.
Collapse
Affiliation(s)
- Hongxu Du
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- Institute of Traditional Chinese Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Kunzhao Yang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Jingyi Yang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Junjie Wan
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Yu Pan
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Weijie Song
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Shuang Xu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Cheng Chen
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Jiahui Li
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| |
Collapse
|
47
|
Ding Z, Lu Y, Zhao J, Zhang D, Gao B. Network Pharmacology and Molecular Dynamics Identified Potential Androgen Receptor-Targeted Metabolites in Crocus alatavicus. Int J Mol Sci 2025; 26:3533. [PMID: 40331986 PMCID: PMC12027412 DOI: 10.3390/ijms26083533] [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: 02/16/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
The objective of this study is to identify the active components of Crocus alatavicus and potential targets through a combination of network pharmacology, molecular docking technology combined with molecular dynamics simulation, and binding free energy analyses. A total of 253 active ingredients from C. alatavicus were screened, and 1360 associated targets were predicted through systematic searches conducted using TCMSP, SwissDrugDesign, and SymMap, which were integrated to construct a pharmacological network to dissect the relationships among active components, targets, diseases, and pathways; we found prostate cancer-related genes were significantly enriched among the targets. Subsequently, the core prostate cancer-related targets were identified in the network, and the binding interactions between protein targets and active components were evaluated using molecular docking technology. Furthermore, molecular dynamics simulation and binding free energy analyses were performed to verify the binding stability of the most promising complex. Then, protein-protein interaction network analysis was conducted to evaluate the core target sites, leading to the identification of nine target proteins with significant correlations, providing potential targets for cancer treatment. Furthermore, these targets were found to be associated with 20 signaling pathways, including neuroactive ligand-receptor interactions, prostate cancer, lipid metabolism and atherosclerosis, as well as calcium signaling pathways. The active component-target-disease-pathway network diagram suggests that Capillarisin, Eugenol, 1-(4-Methoxyphenyl)-1-propanol, 2,4,2',4'-tetrahydroxy-3'-prenylchalcone, and 4-Hydroxymandelonitrile may serve as key components targeting prostate cancer. Molecular docking analyses demonstrated that Capillarisin has a high affinity for the androgen receptor (AR), and molecular dynamics simulation was performed to further verify the binding stability, indicating that Capillarisin may exert its pharmacological effects in prostate cancer. Based on the integrated strategies of network pharmacology, molecular docking, molecular dynamics simulation, and binding free energy analysis, this study generated novel insights into the active components of C. alatavicus and potential targets related to prostate cancer, thus providing valuable biological resources for future drug research and development.
Collapse
Affiliation(s)
- Zhen Ding
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100019, China
| | - Yuanfeng Lu
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
- College of Life Sciences, Nanjing Forestry University, Nanjing 210008, China
| | - Jichen Zhao
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100019, China
| | - Daoyuan Zhang
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
| | - Bei Gao
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
| |
Collapse
|
48
|
Xue H, Feng Z, Jin C, Zhang Y, Ai Y, Wang J, Zheng M, Shi D. Soy Isoflavones Protects Against Stroke by Inhibiting Keap1/NQO1/Nrf2/HO-1 Signaling Pathway: Network Pharmacology Analysis Combined with the Experimental Validation. Pharmaceuticals (Basel) 2025; 18:548. [PMID: 40283984 PMCID: PMC12030689 DOI: 10.3390/ph18040548] [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: 03/03/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Objectives: Ischemic stroke is a severe neurological disorder with high morbidity, mortality, and disability rates, posing a substantial burden on patients, families, and healthcare systems. Soy isoflavone (SI), a naturally occurring phytoestrogen, has demonstrated promising neuroprotective effects. This study aimed to evaluate the anti-stroke efficacy of SI and elucidate its underlying mechanisms through integrated phytochemical profiling, network pharmacology, and both in vitro and in vivo experimental validation. Methods: Active constituents of SI were extracted via reflux and identified using liquid chromatography-mass spectrometry (LC-MS). Network pharmacology was employed to predict therapeutic targets and signaling pathways. The neuroprotective effects of SI were first assessed in PC12 cells subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) injury in vitro. For in vivo evaluation, transient cerebral ischemia-reperfusion injury was induced using the bilateral common carotid artery occlusion (BCCAO) model in adult male ICR rats (27.3 ± 1.8 g; 6-8 weeks old), obtained from the Shanghai Experimental Animal Center, Chinese Academy of Sciences. Forty-eight rats were randomly assigned into four groups (n = 12): sham, model (BCCAO), SI-treated (100 mg/kg, oral gavage for 5 days), and edaravone (EDA)-treated (10 mg/kg, i.p., positive control). All procedures were approved by the Institutional Animal Care and Use Committee of Changchun Normal University (Approval No. 2024003, 13 March 2024) and conducted in accordance with the NIH guidelines and ARRIVE 2.0 reporting standards. Results: In vitro, SI significantly enhanced PC12 cell viability from 57.23 ± 2.88% to 80.76 ± 4.43% following OGD/R. It also reduced intracellular Ca2+ by 58.42%, lactate dehydrogenase (LDH) release by 37.67%, caspase-3 activity by 55.05%, and reactive oxygen species (ROS) levels by 74.13% (p < 0.05). A flow cytometry analysis revealed that OGD/R increased the apoptosis rate from 5.34% (control) to 30.85% (model group), which was significantly attenuated by SI treatment, especially in the 560 µg/mL group (20.00%), followed by the 140 and 280 µg/mL groups. In vivo, SI improved neurological scores from 8.3 ± 1.09 to 6.8 ± 1.68, reduced cerebral infarction volume by 18.49%, and alleviated brain edema by 10.42% (p < 0.05). SI also decreased malondialdehyde (MDA) and LDH levels by 31.15% and 39.46%, respectively, while increasing the activity of antioxidant enzymes: superoxide dismutase (SOD) by 11.70%, catalase (CAT) by 26.09%, and glutathione peroxidase (GSH-px) by 27.55% (p < 0.01). Scratch assay results showed that SI restored the impaired migratory ability of the OGD/R-treated PC12 cells, further supporting its role in cellular repair. A Western blot analysis demonstrated the upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and NAD(P)H:quinone oxidoreductase 1 (NQO1) and the downregulation of Kelch-like, ECH-associated protein 1 (Keap1) in the cerebral ischemia-reperfusion model. Conclusions: These findings indicate that soy isoflavone confers significant neuroprotective effects against cerebral ischemia-reperfusion injury by enhancing endogenous antioxidant defense mechanisms, reducing oxidative stress, inhibiting apoptosis, and promoting cell migration. The protective effects are likely mediated through the activation of the Nrf2/Keap1 signaling pathway, supporting the therapeutic potential of SI in ischemic stroke treatment.
Collapse
Affiliation(s)
- Huiming Xue
- College of Life Sciences, Changchun Normal University, Changchun 130032, China; (H.X.); (Z.F.); (C.J.); (Y.Z.)
| | - Zhen Feng
- College of Life Sciences, Changchun Normal University, Changchun 130032, China; (H.X.); (Z.F.); (C.J.); (Y.Z.)
| | - Chang Jin
- College of Life Sciences, Changchun Normal University, Changchun 130032, China; (H.X.); (Z.F.); (C.J.); (Y.Z.)
| | - Yue Zhang
- College of Life Sciences, Changchun Normal University, Changchun 130032, China; (H.X.); (Z.F.); (C.J.); (Y.Z.)
| | - Yongxing Ai
- College of Animal Science, Jilin University, Changchun 130062, China;
| | - Jing Wang
- Central Laboratory, Changchun Normal University, Changchun 130032, China;
| | - Meizhu Zheng
- Central Laboratory, Changchun Normal University, Changchun 130032, China;
| | - Dongfang Shi
- Central Laboratory, Changchun Normal University, Changchun 130032, China;
| |
Collapse
|
49
|
Panossian A. Trends and Pitfalls in the Progress of Network Pharmacology Research on Natural Products. Pharmaceuticals (Basel) 2025; 18:538. [PMID: 40283973 PMCID: PMC12030339 DOI: 10.3390/ph18040538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Herbs, used as food and a source of medicine for centuries, have been extensively studied over time for their chemical and pharmacological properties, with two main aims [...].
Collapse
|
50
|
Sun H, Wang D, Zheng Y, Ye Y. Elucidating the Molecular Mechanisms of Hederagenin-Regulated Mitophagy in Cervical Cancer SiHa Cells through an Integrative Approach Combining Proteomics and Advanced Network Association Algorithm. J Proteome Res 2025; 24:2081-2095. [PMID: 40135937 PMCID: PMC11976847 DOI: 10.1021/acs.jproteome.5c00022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Hederagenin (Hed), a natural triterpenoid, exhibits antitumor potential in cervical cancer. The present study was designed to explore Hed's regulatory mechanisms on mitophagy in SiHa cervical cancer cells, employing tandem mass tag (TMT) proteomics and an advanced network association algorithm (NAA). Our findings revealed that Hed decreased SiHa cell viability, induced apoptosis, and altered mitochondrial membrane potential. Notably, Hed inhibited mitophagic flux under both normoxic and hypoxic conditions. Through TMT proteomics analysis and innovative NAA, we identified a close association between the HIF-1 signaling pathway and mitophagy. Network analysis further suggested that Hed acts on a target network centered on SRC, STAT3, AKT1, and HIF1A. Western blot analysis confirmed the expression and phosphorylation status of these targets in response to Hed. This study elucidates the molecular mechanisms underlying Hed's regulation of mitophagy in SiHa cells, offering novel insights and potential therapeutic targets for cervical cancer treatment.
Collapse
Affiliation(s)
- Hao Sun
- Pharmacy
Department, Women’s Hospital, Zhejiang
University School of Medicine, Hangzhou 310006, China
| | - Dan Wang
- Pharmacy
Department, Zhejiang Hospital, Hangzhou 310030, China
| | - Yongquan Zheng
- Pharmacy
Department, Women’s Hospital, Zhejiang
University School of Medicine, Hangzhou 310006, China
| | - Yiqing Ye
- Pharmacy
Department, Women’s Hospital, Zhejiang
University School of Medicine, Hangzhou 310006, China
| |
Collapse
|