1
|
Wang XR, Chen AJ, Hou CC, Wang YY, Guo J, Li MY. Unveiling the immunopharmacological mechanisms of Danggui Yinzi (DGYZ) in treating chronic urticaria: insights from network pharmacology and experimental validation. Chin Med 2025; 20:81. [PMID: 40490797 DOI: 10.1186/s13020-025-01137-7] [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: 03/23/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Chronic urticaria (CU), a prevalent and often debilitating allergic skin disorder, is primarily triggered by mast cell degranulation. Danggui Yinzi (DGYZ), a traditional Chinese medicine formula, has been employed to treat pruritic conditions. However, the molecular mechanisms underlying its effects in CU remain unclear. This study aimed to investigate the immunopharmacological mechanisms of DGYZ in CU, hypothesizing that it modulates immune responses through its bioactive components, which is critical for the development of novel therapeutic agents. METHODS Ultra-performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS) was used to identify the active compounds in DGYZ. In vivo, BALB/c mouse models of DNP-IgE/DNFB-induced CU were established and grouped into Normal Control (NC), Model, various-dose DGYZ, and Loratadine groups. Post-treatment, immunopharmacological parameters were assessed, and skin tissue was collected for histopathological analysis, mast cell quantification, and immunohistochemistry to evaluate the impact on immune cells and molecules. Serum levels of inflammatory cytokines (TNF-α, IL-6) were quantified using ELISA kits. In vitro, the human mast cell line LAD2 was pretreated with key active components of DGYZ (Quercetin and Paeoniflorin) at different concentrations before mast cell degranulation was induced. Degranulation markers (β-HEX, HIS) and the expression of proteins in immune-related signaling pathways (PI3K-Akt, TLR4) were then measured. RESULTS A total of 38 active components were identified in DGYZ. In vivo, DGYZ inhibited mast cell degranulation, blue spot reactions, and skin damage in mice. It also decreased the levels of inflammatory cytokines (TNF-α, IL-6) and suppressed the activation of associated signaling pathways. In vitro, both Quercetin and Paeoniflorin reduced mast cell degranulation and the activation of TLR4 and PI3K-Akt pathways. CONCLUSION This study, employing UPLC-Q-TOF-MS and both in vivo and in vitro experiments, provides a comprehensive analysis of the mechanism of DGYZ in CU. The findings indicate that DGYZ exerts therapeutic effects in CU by modulating immune responses. This research lays the foundation for a deeper understanding of its immunopharmacological mechanisms, potentially aiding the development of novel drugs and therapeutic strategies for CU management.
Collapse
Affiliation(s)
- Xu-Rui Wang
- Department of Chinese Medicine Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - An-Jing Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chang-Cheng Hou
- Jiangsu Province Hospital of Traditional Chinese Medicine Chongqing Hospital, Chongqing, China
| | - Yue-Yue Wang
- Jiangsu Province Hospital of Traditional Chinese Medicine Chongqing Hospital, Chongqing, China
| | - Jing Guo
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, No.39, 12 Qiao Road, Jinniu District, Chengdu, Sichuan, China.
| | - Ming-Yue Li
- Special Needs Outpatient Department, Hospital of Chengdu University of Traditional Chinese Medicine, No.39, 12 Qiao Road, Jinniu District, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Chen Y, Wang Q, Bian S, Dong J, Xiong J, Le J. Exploration of the mechanism of Polyphyllin I against hepatocellular carcinoma based on network pharmacology, molecular docking and experimental validation. Discov Oncol 2025; 16:941. [PMID: 40434621 PMCID: PMC12120097 DOI: 10.1007/s12672-025-02341-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/08/2025] [Indexed: 05/29/2025] Open
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide. Targeted therapies hold promise for HCC treatment, and understanding the molecular mechanisms of action is crucial for developing novel therapeutic strategies. Polyphyllin I, a natural compound with known antitumor activity, represents a potential therapeutic candidate. METHODS This study employed a network pharmacology approach to investigate the anti-HCC effects of Polyphyllin I and its underlying mechanisms. Drug and disease related targets were identified and intersected to construct Components-Gene Symbols-Disease and Protein-Protein Interaction networks. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. Molecular docking simulations were conducted to explore the interactions between Polyphyllin I and key pathway proteins (VEGF-C and β-catenin). Finally, in vitro and in vivo experiments validated the anti-HCC effects and underlying mechanisms of Polyphyllin I. RESULTS Network pharmacology analysis revealed that Polyphyllin I targets multiple genes and pathways implicated in HCC development and progression. GO and KEGG analyses identified significant enrichment of pathways related to cell proliferation, apoptosis and angiogenesis, including VEGF and the Wnt/β-catenin signaling pathways. Molecular docking simulations demonstrated strong binding affinities between Polyphyllin I and VEGF-C and β-catenin. In vitro and in vivo experiments confirmed that Polyphyllin I effectively inhibits HCC cell proliferation, induces apoptosis, and suppresses angiogenesis, potentially by modulating the VEGF-C and Wnt/β-catenin signaling pathways. CONCLUSIONS The study provides compelling evidence for the antitumor activity of Polyphyllin I in HCC and elucidates its possible molecular mechanisms, suggesting that Polyphyllin I holds great potential as a therapeutic agent for HCC.
Collapse
Affiliation(s)
- Yilong Chen
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Qiuying Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Shuixiu Bian
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jing Dong
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jie Xiong
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Jiamei Le
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| |
Collapse
|
3
|
Sun Y, Jia Y, Yan X, Zhang Q, Li X, Yang Z, Wei D, Wu X, Mao Z, Cao X, Tong X, Huang F. Huatanhuoxue decoction alleviates airway inflammation by regulating IL-17A signaling pathway in obese asthmatic mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119814. [PMID: 40245963 DOI: 10.1016/j.jep.2025.119814] [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: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huatanhuoxue Decoction (HTHX) is a traditional Chinese formula consisting of nine herbs. It is used to improve obesity-related asthma symptoms and reduce airway inflammation. AIM OF THE STUDY To study HTHX effects on airway inflammation in obese asthmatic mice via the IL-17 A signaling pathway. MATERIALS AND METHODS Network pharmacology was used to predict the bioactive ingredients in HTHX. Subsequently, an obese asthma model was established by high-fat diet feeding and exposure to house dust mite. The effects of HTHX on obesity-related asthma progression were investigated using histopathological examinations, airway hyperresponsiveness determinations, and enzyme-linked immunosorbent assays. The mechanism of action of HTHX was confirmed by Western blots, flow cytometry, immunohistochemistry, and immunofluorescence analyses. RESULTS HTHX alleviated the development in obese asthma mice by improving the pathological condition of lung tissue, airway hyperresponsiveness, and inflammatory factors. Network pharmacology identified the involvement of the IL-17 signaling pathway. HTHX decreased the production of neutrophils and the expression of NETs in lung tissue. HTHX also reduced group 3 innate lymphoid cells and Th17 cells, which are responsible for producing IL-17 A. The production of IL-17 A-related protein was also suppressed. The results indicate that HTHX inhibited the excessive activation of the IL-17 A signaling pathway. CONCLUSIONS HTHX alleviated airway inflammation by regulating the IL-17 A signaling pathway in obese asthmatic mice.
Collapse
Affiliation(s)
- Yun Sun
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Yongrui Jia
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Xiaodong Yan
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Qiushi Zhang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Xiaohong Li
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Zhuya Yang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Danxia Wei
- The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming 650599, China
| | - Xiangnong Wu
- The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming 650021, China
| | - Zewei Mao
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Xue Cao
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, 650500, China
| | - Xiaoyun Tong
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China; The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming 650021, China.
| | - Feng Huang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| |
Collapse
|
4
|
Dakpa G, Chiang YT, Lin LY, Tsao NW, Wang CH, Pérez-Sánchez H, Fernández JRA, Wang SY. Essential oil-derived compounds target core fatigue-related genes: A network pharmacology and molecular Docking approach. PLoS One 2025; 20:e0314125. [PMID: 40435272 PMCID: PMC12118864 DOI: 10.1371/journal.pone.0314125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/20/2025] [Indexed: 06/01/2025] Open
Abstract
Fatigue is a widespread condition associated with various health issues, yet identifying specific bioactive compounds for its management remains challenging. This study integrates network pharmacology and molecular docking to uncover essential oil-derived compounds with potential antifatigue properties by targeting key genes and molecular pathways. A comprehensive analysis of 872 essential oil compounds was conducted using PubChem, with target prediction via SwissTargetPrediction. The protein-protein interaction (PPI) network and KEGG pathway analysis identified core fatigue-related targets, including ALB, BCL2, EGFR, IL-6, and STAT3, in metabolic dysregulation and inflammatory responses linked to fatigue. Molecular docking exhibits strong binding affinity between key compounds such as Calamenene, T-cadinol, and Bornyl acetate and core targets, suggesting their potential antifatigue effects. However, ADMET analysis confirmed T-cadinol's drug-likeness, suggesting good bioavailability and minimal toxicity risks. Thus, molecular docking revealed high binding affinity, which was further validated through a 100 ns MD simulation and demonstrated stable interactions with low root mean square deviation (RMSD). Additionally, hydrogen bond analysis confirmed that T-cadinol maintained consistent interactions with key residues such as Thr-790 in EGFR, Arg-222 in ALB, and Arg-104 in IL-6, indicating strong binding stability. While this study provides valuable computational insights, further in vitro and in vivo validation is necessary to confirm these findings and explore potential therapeutic applications.
Collapse
Affiliation(s)
- Gyaltsen Dakpa
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, Taiwan
| | | | - Li-Yin Lin
- Liyu International Co., Ltd, Taichung, Taiwan
| | - Nai-Wen Tsao
- Special Crop and Metabolome Discipline Cluster, Academy of Circle Economy, National Chung Hsing University, Taichung, Taiwan.
| | - Chung-Hsuan Wang
- Special Crop and Metabolome Discipline Cluster, Academy of Circle Economy, National Chung Hsing University, Taichung, Taiwan.
| | | | - Jorge Ricardo Alonso Fernández
- Structural Bioinformatics and High-Performance Computing (BIO-HPC), Campus de los Jerónimos, Universidad Católica de Murcia (UCAM), Guadalupe, Murcia, España (Spain),
| | - Sheng-Yang Wang
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Special Crop and Metabolome Discipline Cluster, Academy of Circle Economy, National Chung Hsing University, Taichung, Taiwan.
- Department of Forestry, National Chung-Hsing University, Taichung, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
5
|
Leng L, Wang X, Wang H, Hu Y, Deng Y, Wang C. Molecular mechanisms of arecoline-induced oral cancer: a network toxicology and molecular docking techniques integrated analysis. Discov Oncol 2025; 16:842. [PMID: 40397259 PMCID: PMC12095829 DOI: 10.1007/s12672-025-02659-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/10/2025] [Indexed: 05/22/2025] Open
Abstract
The IARC classified betel nut as Group 1 carcinogen (2004) and arecoline as Group 2B carcinogen (2020), with approximately one-third of global oral cancer cases attributed to smokeless tobacco or betel nut consumption. While current evidence establishes an association between arecoline and oral cancer, the underlying molecular mechanisms remain complex and poorly elucidated. This study employs network toxicology integrated with molecular docking techniques to systematically investigate the potential molecular pathogenesis of arecoline-induced oral cancer, aiming to provide novel insights for targeted therapeutic strategies. The SMILES structure of arecoline was retrieved from PubChem for foundational data preparation. Toxicity profiling was conducted using ProTox-3.0 and ADMETlab databases. Potential targets of arecoline were identified via STITCH and SwissTargetPrediction. Oral cancer-related targets were collated from GeneCards, OMIM, and TTD. Intersection analysis between arecoline targets and oral cancer-associated targets was performed to identify shared targets, which were further utilized to construct compound-target regulatory network and subjected to PPI, GO, and KEGG analyses. Core targets driving oral cancer were screened using the cytoHubba plugin. Then, the correlation between core targets and immune cell infiltration in oral cancer was explored, and molecular docking validated the binding affinity of arecoline to core targets. Finally, Gromacs 2022.3 software was used to simulate the molecular dynamics of the complexes obtained by molecular docking for 100 ns. Using the STITCH and SwissTargetPrediction databases, a total of 46 potential targets of arecoline were identified. Concurrently, 2,375 oral cancer-related targets were retrieved from GeneCards, OMIM, and TTD. Intersection analysis of these two target sets yielded 26 overlapping targets. PPI analysis revealed that TP53, IL6, SNAI1, and CASP3 occupied central positions in the network, exhibiting extensive interactions with other target proteins. Enrichment analysis comprehensively elucidated the molecular functions, biological processes, cellular components, and associated pathways of these overlapping targets. Further screening using Cytoscape software identified four core targets: TP53, TNF, IL6, and CASP3. Immune infiltration analysis indicated that the expression levels of TP53, TNF, IL6, and CASP3 in oral cancer tissues were positively correlated with the infiltration levels of immune cells, including CD8 + T cells, Th1 cells, NK cells, and macrophages. Molecular docking experiments demonstrated strong binding activities between arecoline and TP53, IL6, and CASP3, while TNF also exhibited moderate binding affinity. Dynamic simulation further verified the stable binding of arecoline to TP53, TNF, IL6 and CASP3. Arecoline may induce oral cancer by acting on core targets including TP53, TNF, IL6, and CASP3, which interfere with normal cellular growth regulation, inflammatory responses, and apoptotic mechanisms. Therapeutic strategies targeting TP53, TNF, IL6, and CASP3 may represent novel research directions for clinical diagnosis and treatment of oral cancer.
Collapse
Affiliation(s)
- Linghan Leng
- Department of Intensive Care Unit, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan, China
| | - Xin Wang
- School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Wang
- Clinical Medical College, Southwest Medical University, Luzhou, Sichuan, China
| | - Yingchun Hu
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yaxing Deng
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chenglin Wang
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
6
|
Xin J, Zhou C, Wang Y, Chen H, Yin K, Gao L, Shao S. Unraveling DINCH - Induced hepatotoxicity mechanisms via network toxicology and molecular docking with experimental validation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118305. [PMID: 40393321 DOI: 10.1016/j.ecoenv.2025.118305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025]
Abstract
Phthalates, as a class of known endocrine disruptors, have been controversial because of their potential carcinogenicity and toxicity. Diisononyl cyclohexane-1,2-dicarboxylate (DINCH) is considered to be less toxic and more prone to environmental degradation, and is widely used as a substitute for phthalate. With the increasing use of DINCH in consumer products and industrial materials, the frequency of its detection in the air and human urine has also increased, which has aroused concern about its potential toxicity in food safety. Despite the increasing popularity of DINCH, toxicological studies on this topic are still limited. This study first predicted the hepatotoxicity and carcinogenicity of DINCH via the ADMETlab 3.0 platform. Next, the potential hepatotoxic genes associated with DINCH were collected through multiple databases, and a gene network was constructed. Through proteinprotein interaction, GO enrichment and KEGG pathway analyses, we elucidated the primary mechanism by which DINCH may induce hepatotoxicity. The expression of the selected key genes in related diseases was subsequently validated via the liver cancer database of TCGA and the NASH dataset of GEO. In addition, molecular docking technology and dynamics simulation were used to simulate the interaction and binding ability between DINCH and the core target. Cell experiments verified that DINCH increases hepatotoxicity primarily by upregulating TNF, TP53, and PPARG. In summary, this study elucidates the potential biological mechanisms of DINCH-induced hepatotoxicity, providing new scientific insights for the prevention and management of related toxicities.
Collapse
Affiliation(s)
- Jingxin Xin
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Changxu Zhou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Ying Wang
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Huiqi Chen
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Keling Yin
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong 250021, China; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Shanshan Shao
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong 250021, China; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China.
| |
Collapse
|
7
|
Tang N, Sun W, Zhang J, Ma X, Wang Y. Assessing the toxicological effects and mechanism of plasticizer exposure on inflammatory bowel disease based on network toxicology and molecular docking. Food Chem Toxicol 2025; 202:115543. [PMID: 40354873 DOI: 10.1016/j.fct.2025.115543] [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: 02/17/2025] [Revised: 04/13/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Phthalates (PAEs) are one of the most commonly used plasticizers. Due to their good performance, they are widely used in daily production, such as food packaging, paints, adhesives, children's toys, lubricants and building materials. However, PAEs usually have weak interactions with polymers, which can easily cause environmental pollution in use. These plasticizers have been linked to various health conditions, including inflammatory disorders. They are less intensively studied in the occurrence of inflammation, especially inflammatory bowel disease (IBD), and the necessity to evaluate their pathogenic molecular toxicity is particularly urgent. In this study, network toxicology and molecular docking methods were used to study the toxicological mechanism of IBD induced by four common plasticizers (DBP, DEHP, DEP, DNOP). Potential related targets were predicted using the PharmMapper, SwissStargetPrediction, GeneCards, DisGeNET, OMIM and TTD databases, and 286 related targets were identified. Using Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, binding protein-Protein Interaction (PPI) networks and cytoHubba plug-ins, ten relevant signaling pathways (PI3K-Akt signaling pathway, lipid and atherosclerosis, AGE-RAGE signaling pathway in diabetic complications, Proteoglycans in cancer, and so on.) and ten hub genes were identified. Four plasticizers (DBP, DEHP, DEP, DNOP) and the top 10 selected Hub gene targets (SRC, KRAS, PIK3CA, PIK3R1, JAK2, PTPN11, PIK3CD, HRAS, PIK3CG, EGFR) were analyzed by molecular docking. This study provides valuable insights into the molecular mechanisms of plasticizer-induced IBD and highlights the practicality of network toxicology in assessing the toxicity of emerging environmental pollutants. It enhances our understanding of the health risks posed by plasticizers and offers new strategies for mitigating their impact on inflammatory diseases.
Collapse
Affiliation(s)
- Ning Tang
- Department of Integrative Medicine, Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, 110167, PR China
| | - Wentao Sun
- Department of Acupuncture and Massage, Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, 110167, PR China
| | - Jingke Zhang
- Department of Integrative Medicine, Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, 110167, PR China
| | - Xin Ma
- The Fourth People's Hospital of Shenyang, Shenyang, 110000, PR China
| | - Yan Wang
- Department of Integrative Medicine, Dalian Medical University, Dalian, 116044, PR China.
| |
Collapse
|
8
|
He P, Wang Z, Yang J, Pan P, Shi T, Xu S, Lan J, Hao Z, Yang A, Chen L, Xi Y, Wang J. Mechanism of Ligusticum wallichii-Borneol in the Treatment of Cerebral Ischemic Stroke in Rats Based On Network Pharmacology, Molecular Docking, and Experimental Verification. Chem Biodivers 2025; 22:e202401893. [PMID: 39714965 DOI: 10.1002/cbdv.202401893] [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: 08/09/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
The pharmacodynamics, molecular biology, network pharmacology, and molecular docking mechanisms of the Ligusticum wallichii and borneol medication pair (CXBP) were investigated for ischemic stroke treatment. Effective chemical components and targets of CXBP were identified using TCMSP, ETCM, and SymMap databases, whereas ischemic stroke targets were sourced from OMIM, GeneCards, TTD, PubMed, Web of Science, CNKI, Wanfang Data, and VIP databases. Protein-protein interaction (PPI) networks were generated using the STRING database, and GO and KEGG enrichment analyses were conducted using Metascape. A "disease-pathway-target-component-drug" network was created in Cytoscape, and molecular docking was confirmed with PyMOL and AutoDock tools. Rat models of MCAO were established to evaluate neurological scores, triphenyltetrazolium chloride (TTC) staining, and Nissl staining. Key components were validated through enzyme-linked immunosorbent assay (ELISA), real-time polymerase chain reaction (PCR), and immunohistochemistry. Thirty-three active ingredients and 419 potential targets for CXBP, with key compounds including Z-6,8',7,3'-diligustilide, cedrene, (+)-alpha-funebrene, POL, dipterocarpol, oleanolic acid, 1-acetyl-beta-carboline, and erythrodiol. Critical targets included ESR1, PRKCA, and PTPN6. KEGG pathway analysis revealed 179 signaling pathways, primarily neuroactive ligand-receptor interactions, whereas GO enrichment analysis identified 2911 biological processes, 398 molecular activities, and 203 cellular components. The neurological function scores and TTC staining of the infarcted brain regions were significantly improved following CXBP intervention compared to the MCAO group. These findings were supported by Nissl staining, which demonstrated better preserved cellular morphology and a significantly higher number of Nissl vesicles in the CXBP group. ELISA analysis revealed substantial modulation in gene expression: levels of PRKCA, PTPN6, ESR1, and TNF-α changed significantly, whereas IL-1β, IL-6, and TNF-α were notably downregulated compared to the MCAO group. PCR results corroborated these findings, showing a significant decrease in PRKCA expression and marked downregulation of IL-1β, IL-6, and TNF-α, whereas ESR1 and PTPN6 levels increased significantly. Immunohistochemical analysis further confirmed these results, with the CXBP and nimodipine groups exhibiting higher ESR1 and PTPN6 expression and lower PRKCA expression compared to the MCAO group. To improve cerebral ischemia and reperfusion injury, CXBP improves ischemic stroke outcomes through key active ingredients (e.g., Z-6,8',7,3'-diligustilide, cedrene, and oleanolic acid) and targets ESR1, PRKCA, and PTPN6, modulating multiple signaling pathways to alleviate cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Pengfen He
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhifeng Wang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jiao Yang
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Pan Pan
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Ting Shi
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Shuangfeng Xu
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Junfeng Lan
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhihui Hao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Aiming Yang
- Department of Neurology, Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
| | - Liang Chen
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yujiang Xi
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jian Wang
- Department of Neurology, First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
9
|
Gong B, Zhang X, Yue D, Ma C, Kou Y, Li Y. Bawei Chenxiang Powder Protects Cardiomyocytes From Myocardial Ischemia/Reperfusion Injury via the PI3K-AKT Pathway. Chem Biodivers 2025; 22:e202401424. [PMID: 39676557 DOI: 10.1002/cbdv.202401424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 11/20/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
Ischemia-reperfusion damage to cardiomyocytes is one of the main directions of cardiovascular disease research, and Bawei Chenxiang powder (BWCX) is a traditional ethnomedicinal compound preparation mainly used in the treatment of cardiovascular diseases. On the basis of serum pharmacology, the present study aimed to explore the potential mechanism of BWCX against myocardial ischemia-reperfusion damage to cardiomyocytes. We prepared BWCX-serum containing. Using serum pharmacology and bioinformatics approaches, we explored its protective effects on H9C2 cells in a hypoxia/reoxygenation (H/R) model. Additionally, we investigated the underlying mechanisms. BWCX-containing serum can increase the survival rate of H9C2 cells and reduce oxidative stress levels in an H/R model. Specifically, it decreases the release of malondialdehyde (MDA), lactate dehydrogenase (LDH), creatine kinase (CK), and reactive oxygen species (ROS), while increasing the levels of catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GSH-PX), and Complex I. Additionally, it downregulates the expression of NADH dehydrogenase (ubiquinone) 1 alpha sub-Complex 10 (NDUFA-10), thioredoxin (Trx), heme oxygenase 1 (HO-1), and kelch-like ECH-associated protein 1 (Keap1), and it upregulates the expression of nuclear factor erythroid 2-related factor 2 (Nrf2). These effects are potentially mediated through the PI3K-AKT pathway. In the present study, we found that BWCX powder exhibited significant ameliorative and reparative effects on H/R-induced cardiomyocyte injury by alleviating the level of oxidative stress during H/R and acting on the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Baihui Gong
- Department of Pharmacy, Faculty of Medicine, Qinghai University, Xining, China
| | - Xingfang Zhang
- Department of Pharmacy, Faculty of Medicine, Qinghai University, Xining, China
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining, China
| | - Dongfang Yue
- Department of Pharmacy, Faculty of Medicine, Qinghai University, Xining, China
| | - Chunxiu Ma
- Department of Pharmacy, Faculty of Medicine, Qinghai University, Xining, China
| | - Yiying Kou
- Department of Pharmacy, Faculty of Medicine, Qinghai University, Xining, China
| | - Yongfang Li
- Department of Pharmacy, Faculty of Medicine, Qinghai University, Xining, China
| |
Collapse
|
10
|
Tuo Y, Peng S, Li Y, Dang J, Feng Z, Ding L, Du S, Liu X, Wang L. Quinoa protein and its hydrolysate improve the fatigue resistance of mice: a potential mechanism to relieve oxidative stress and inflammation and improve energy metabolism. J Nutr Biochem 2025; 139:109863. [PMID: 39952621 DOI: 10.1016/j.jnutbio.2025.109863] [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: 09/21/2024] [Revised: 01/05/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Fatigue is commonly marked by reduced endurance and impaired function, often linked to overexertion and chronic conditions. Quinoa (Chenopodium quinoa Willd.), with its rich amino acids and resilience to harsh conditions, offers a novel strategy for combating fatigue. This study explored the antifatigue effects of quinoa protein (QPro) and its hydrolysate (QPH) in weight-loaded swimming mice. After 4 weeks of oral administration, QPro and QPH significantly prolonged swimming duration, reduced serum fatigue biomarkers (lactic acid, urea nitrogen, lactate dehydrogenase, creatine kinase), and elevated glycogen reserves in the liver and muscle. RT-qPCR analysis indicated that QPH activated hepatic gluconeogenesis via G6Pase and PEPCK signaling and enhanced mitochondrial function through PGC-1α/NRF1/TFAM signaling in muscle. Additionally, QPro and QPH boosted antioxidant defenses by improving antioxidant enzyme activity, reducing malondialdehyde through the Nrf2/HO-1 pathway, and suppressing inflammation by reducing TNF-α and IL-6 levels. Network pharmacology identified 31 key targets involved in energy metabolism and inflammation, providing novel insights into the molecular mechanisms underlying the antifatigue properties of quinoa peptides. These findings highlight the potential of QPro and QPH as natural and bioactive ingredients in functional foods for enhancing endurance and mitigating fatigue.
Collapse
Affiliation(s)
- Yuanrong Tuo
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Siwang Peng
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Yiju Li
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Jiamin Dang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Zhi Feng
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Long Ding
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China.
| | - Shuangkui Du
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China; Engineering Research Center of Grain and Oil Functionalized Processing, Universities of Shaanxi Province, Yangling, Shaanxi, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Liying Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China; Engineering Research Center of Grain and Oil Functionalized Processing, Universities of Shaanxi Province, Yangling, Shaanxi, China.
| |
Collapse
|
11
|
Jing S, Zhenhai C, Xiao H, Chunhan L, Wanlin W, Xuting S, Han Y, Xiaoyu Z, Meiying J. Jiedu Tongluo Tiaogan Formula Modulates Glycolipid Metabolism in Type 2 Diabetes via Pyroptosis: Network Pharmacology and In Vivo Analysis. Biomed Chromatogr 2025; 39:e70077. [PMID: 40219662 DOI: 10.1002/bmc.70077] [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: 12/11/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by pancreatic β-cell dysfunction and insulin resistance, with pyroptosis emerging as a key contributor to β-cell loss. Jiedu Tongluo Tiaogan Formula (JTTF), a traditional Chinese medicine (TCM), has shown clinical efficacy in T2DM management, but its mechanism linking pyroptosis remains unexplored. This study integrates UPLC-MS/MS, network pharmacology, and in vivo experiments to elucidate JTTF's anti-diabetic mechanisms. UPLC-MS/MS identified 441 compounds in JTTF, predominantly alkaloids, flavonoids, phenols, and terpenoids. Network pharmacology revealed JTTF's multi-target effects on T2DM-associated pyroptosis, particularly via the NLRP3/Caspase-1/GSDMD pathway. In diabetic mice, JTTF dose-dependently reduced fasting blood glucose, insulin resistance, and dyslipidemia, while restoring pancreatic β-cell morphology. Mechanistically, JTTF suppressed NLRP3 inflammasome activation, downregulated Caspase-1 and GSDMD expression, and attenuated IL-1β/IL-18 release. Notably, this study provides the first evidence of JTTF's anti-pyroptotic effects in T2DM, highlighting its unique ability to modulate glycolipid metabolism and inflammatory cell death concurrently. These findings underscore JTTF's translational promise for preserving β-cell function and suggest future exploration of non-classical pyroptosis pathways. Our work bridges traditional medicine and molecular pharmacology, paving the way for clinical trials and integrative T2DM therapies.
Collapse
Affiliation(s)
- Su Jing
- Changchun University of Chinese Medicine, Changchun, China
| | - Cui Zhenhai
- Changchun University of Chinese Medicine, Changchun, China
| | - Han Xiao
- Changchun University of Chinese Medicine, Changchun, China
| | - Li Chunhan
- Jilin Hospital of Integrated Traditional Chinese and Western Medicine, Jilin, China
| | - Wang Wanlin
- Changchun University of Chinese Medicine, Changchun, China
| | - Song Xuting
- Changchun University of Chinese Medicine, Changchun, China
| | - Yu Han
- Changchun University of Chinese Medicine, Changchun, China
| | - Zhuang Xiaoyu
- Changchun University of Chinese Medicine, Changchun, China
| | - Jin Meiying
- Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
12
|
Dai JM, Zhang JD, Liu X, Zhang LF, Wang J, Xu Y, Yang GY, Li J, Chen ML, Hu QF. Gene editing, metabolomics, network pharmacology strategies to explore terpenoid content and anti-TMV activity in NtSPS1 knockout Nicotiana tabacum. Sci Rep 2025; 15:14581. [PMID: 40280998 PMCID: PMC12032281 DOI: 10.1038/s41598-025-98745-y] [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: 10/27/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
The content of terpenoids in tobacco can alter its resistance to TMV. NtSPS1, a pivotal structural gene in tobacco, is capable to regulate the terpenoid content. In this study, we investigated the effect of NtSPS1 knockout in HD on the content of terpenoids and the anti-TMV activity of this mutant using gene editing, widely targeted metabolomics, network pharmacology, and molecular docking. 48 terpenoids (six up-regulated and five down-regulated) in NtSPS1 knockout tobacco compared with WT leaves. Notably, solanesol was remarkable downregulation which was lowered by fourfold and compounds 1 (log2FC = 18.2), 8 (log2FC = 16.7) were significant upregulation between the mutants and wild-type line leaves. The 46 terpenoid's target network encompassed 150 nodes, 509 edges and their underlying mechanisms in the therapeutic management of TMV are discussed. Furthermore, the network pharmacology and molecular docking revealed that compounds 16, 18, 23, 27, and 36 exhibited significant affinity in their respective interactions. Ultimately, five compounds were assayed for their anti-TMV effects, noteworthily, compounds 36 showed potential anti-TMV activity. Above all, we adopted a multifaceted approach to gain a comprehensive understanding of the terpenoid content and anti-TMV properties in NtSPS1 knockout HD. It enlightens the therapeutic potential of NtSPS1 knockout tobacco and it is helpful to find undescribed anti-TMV activity inhibitors, as well as searching for new anti-TMV candidates from the mutants.
Collapse
Affiliation(s)
- Jia-Meng Dai
- Yunnan Key Laboratory of Tobacco Chemistry, China Tobacco Yunnan Industrial Co., Ltd, Kunming, 650231, P.R. China
| | - Jian-Duo Zhang
- Yunnan Key Laboratory of Tobacco Chemistry, China Tobacco Yunnan Industrial Co., Ltd, Kunming, 650231, P.R. China
| | - Xin Liu
- Yunnan Key Laboratory of Tobacco Chemistry, China Tobacco Yunnan Industrial Co., Ltd, Kunming, 650231, P.R. China
| | - Ling-Fang Zhang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650031, P. R. China
| | - Jin Wang
- Yunnan Key Laboratory of Tobacco Chemistry, China Tobacco Yunnan Industrial Co., Ltd, Kunming, 650231, P.R. China
| | - Yong Xu
- Yunnan Key Laboratory of Tobacco Chemistry, China Tobacco Yunnan Industrial Co., Ltd, Kunming, 650231, P.R. China
| | - Guang-Yu Yang
- Yunnan Key Laboratory of Tobacco Chemistry, China Tobacco Yunnan Industrial Co., Ltd, Kunming, 650231, P.R. China
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650031, P. R. China
| | - Jing Li
- Yunnan Key Laboratory of Tobacco Chemistry, China Tobacco Yunnan Industrial Co., Ltd, Kunming, 650231, P.R. China.
| | - Ming-Li Chen
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China.
| | - Qiu-Fen Hu
- Yunnan Key Laboratory of Tobacco Chemistry, China Tobacco Yunnan Industrial Co., Ltd, Kunming, 650231, P.R. China.
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650031, P. R. China.
| |
Collapse
|
13
|
Zhou K, Yu Y, Li W, Zhu M. Clostridium butyricum Regulates the Inflammatory and Immunoregulatory Pathway Through NFKB1 in Colorectal Cancer Treatment. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10547-w. [PMID: 40279041 DOI: 10.1007/s12602-025-10547-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Colorectal cancer (CRC) ranks among the top three most prevalent malignancies globally and is a leading cause of cancer-related mortality. Traditional therapeutic approaches usually cause significant adverse effects, highlighting the urgent demand for alternative, more effective treatments. Probiotics have gained attentions as potential cancer therapy due to their beneficial impacts on host health. Clostridium butyricum (Cl. butyricum) has shown anticancer properties in recent studies, though the underlying mechanisms remain inadequately understood. This study presents an integrative analysis of network pharmacology and proteomics to elucidate the key targets of Cl. butyricum in CRC treatment. The network pharmacology analysis identified 72 overlapping genes, and functional analysis of these genes indicated that most pathways were related to pathways in cancer and inflammation, and butyrate emerging as the pivotal product of Cl. butyricum due to its strong associations with the identified hub genes. In parallel, proteomics analysis revealed 168 differential expressed proteins (DEPs) in Cl. butyricum-treated HCT-116 cells, comprising 78 upregulated and 90 downregulated proteins. These DEPs were primarily enriched in apoptosis and inflammatory pathways. PPI analysis further highlighted NFKB1 as key contributors to the anticancer effects of Cl. butyricum. The integrative analysis revealed a significant convergence of pathways enrichment patterns, particularly in inflammatory and immune-related pathways. Computational and experimental validation identified NFKB1 as a pivotal molecular target in CRC intervention. These collective findings elucidate the mechanistic basis of the antitumor properties of Cl. butyricum, highlighting its regulatory effects on NFKB1 through both inflammatory and, to a lesser extent, immunoregulatory pathways.
Collapse
Affiliation(s)
- Kun Zhou
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China
| | - Yue Yu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China
| | - Wei Li
- College of Medicine, Translational Medicine Research Institute, Yangzhou University, Yangzhou, 225001, China
| | - Mingchang Zhu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China.
- College of Environmental and Safety Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China.
| |
Collapse
|
14
|
Liu X, Li Z, Xie H, Cui S, Li X, Lang M, Liu M, Shi L. Immunomodulatory effects of Sanghuangporus flavonoids: Key insights into enhancing immunity and restoring immune function. Int Immunopharmacol 2025; 153:114474. [PMID: 40117809 DOI: 10.1016/j.intimp.2025.114474] [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: 09/02/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
The immune system is vital for maintaining homeostasis, defending against external threats, and regulating inflammation, forming the cornerstone of human health. Sanghuangporus, a natural medicinal mushroom, contains flavonoids that may increase immune cell activity, potentially improving human health. This study investigated the immunomodulatory effects of Sanghuangporus flavonoids (PBF) via network pharmacology and in vitro and in vivo experiments. Network pharmacology identified PBF compounds targeting 46 immunosuppression-related targets, with rutin emerging as a key component. Molecular docking confirmed the strong binding affinity of rutin for the core targets IL6, TNF, and IL1B. In vitro, PBF activated mouse macrophages, promoting their proliferation, phagocytic activity, NO production, and cytokine regulation. In vivo, PBF enhanced immune function in normal mice by promoting thymus and spleen growth, increasing cellular and humoral immunity, and restoring immune function in immunosuppressed mice. These findings highlight the potential of PBFs in increasing immunity and treating immunosuppressive diseases.
Collapse
Affiliation(s)
- Xue Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhinan Li
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China.
| | - Hongqing Xie
- Institute of Industrial Crops, Shandong Academy of Agricultral Sciences, Jinan 250100, China.
| | - Shiyao Cui
- College of Life Sciences, Westlake University, Hangzhou 310058, China.
| | - Xiaotong Li
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Mingzi Lang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Mingming Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Liangen Shi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
15
|
Yao K, Yang L, Zhang Q, Li C, Tian H, Zhuo C. Aripiprazole alleviates the high prolactin levels induced by amisulpride via distinct molecular mechanisms: a network pharmacology and molecular docking study. BMC Psychiatry 2025; 25:373. [PMID: 40229786 PMCID: PMC11995546 DOI: 10.1186/s12888-025-06818-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Amisulpride, a unique atypical antipsychotic, significantly increases prolactin secretion during schizophrenia treatment, resulting in adverse effects that reduce patient quality of life and treatment adherence. Aripiprazole, a partial dopamine D2 receptor agonist, reduces prolactin elevation induced by antipsychotic drugs used for schizophrenia treatment. The molecular targets and mechanisms underlying the contrasting effects of these two drugs on prolactin regulation are unclear. The objective of this study was to systematically explore the molecular mechanisms of prolactin regulation by aripiprazole and amisulpride using network pharmacology and molecular docking techniques. METHODS Relevant targets of amisulpride and aripiprazole and for schizophrenia and elevated prolactin treatment were obtained from online databases and screened for significance. A protein-protein interaction network was constructed. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses of the core targets were performed to identify key biological processes and signaling pathways, and a target-pathway-drug integrated network was established. The binding affinities of amisulpride and aripiprazole with core targets were predicted using molecular docking analyses. RESULTS Screening and matching drug and disease targets combined with GO and KEGG pathway enrichment analyses revealed several key signaling pathways involved in prolactin regulation, including MAPK, PI3K/AKT, and dopamine receptor pathways. The core targets of aripiprazole include MAPK3, PPARG, DRD2, and ESR1, and amisulpride primarily targets MMP9, CDC42, mTOR, and AKT1. Molecular docking analysis demonstrated that aripiprazole and amisulpride have high binding affinities for their respective targets, supporting the hypothesis that these drugs regulate prolactin levels through target-ligand interactions. CONCLUSION These findings highlight the distinct signaling pathways and molecular networks involved in prolactin regulation by aripiprazole and amisulpride and provide new insights into the mechanisms of these drugs in schizophrenia treatment. Further pharmacological and clinical research is needed to validate the complex regulatory networks and in vivo effects.
Collapse
Affiliation(s)
- Kaifang Yao
- Computational Biology and Animal Imaging Centre (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
- Key Laboratory of Psychiatric-Neuroimaging-Genetics Laboratory, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China
| | - Lei Yang
- Computational Biology and Animal Imaging Centre (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
- Key Laboratory of Psychiatric-Neuroimaging-Genetics Laboratory, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China
| | - Qiuyu Zhang
- Computational Biology and Animal Imaging Centre (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
- Key Laboratory of Psychiatric-Neuroimaging-Genetics Laboratory, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China
| | - Chao Li
- Computational Biology and Animal Imaging Centre (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
- Key Laboratory of Psychiatric-Neuroimaging-Genetics Laboratory, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China
| | - Hongjun Tian
- Department of Psychiatry and Psychology, Tianjin Fourth Center Hospital, Tianjin, 300041, China
| | - Chuanjun Zhuo
- Computational Biology and Animal Imaging Centre (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China.
- Key Laboratory of Psychiatric-Neuroimaging-Genetics Laboratory, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China.
- Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, People's Republic of China.
| |
Collapse
|
16
|
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
|
17
|
Li Y, Zhou L, Sun K, Guo R, Li Z, Wen Q, Fu G, Yang S. Integrated network pharmacology, proteomics, molecular docking, and experiments in vivo and in vitro to explore the efficacy and potential mechanism of bufalin against hepatocellular carcinoma angiogenesis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119589. [PMID: 40057142 DOI: 10.1016/j.jep.2025.119589] [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: 10/27/2024] [Revised: 02/16/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bufalin is a potent bioactive compound extracted from the venom of toads such as Bufo gargarizans. It has rich pharmacological effects, and its traditional applications mainly include anti-cancer, anti-inflammatory and analgesic, especially in cancer treatment, which has been a hot topic of research. Prior research has suggested that bufalin may have anti-tumor angiogenic effects. However, the efficacy and mechanism of bufalin inhibiting hepatocellular carcinoma (HCC) angiogenesis have yet to be further investigated. AIM OF THE STUDY An extensive detailed strategy via network pharmacology, proteomics, histopathological analysis, molecular docking, in vitro experiments, and in vivo magnetic resonance imaging (MRI) examinations were adopted to investigate the efficacy and mechanisms of bufalin against HCC angiogenesis. MATERIALS AND METHODS Micro-vessel density (MVD) and intravoxel incoherent motion (IVIM) perfusion-related parameters based on magnetic resonance diffusion-weighted imaging were used to identify the effect of bufalin against HCC angiogenesis. Potential bufalin and HCC targets were gathered from appropriate databases. The STRING database was used to construct the target protein interaction networks. The "clusterprofiler" package (version 4.2.2) in R was applied to conduct the target-related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment and Gene Ontology (GO) analysis. Network pharmacology and proteomics were integrated to identify key targets and pathways related to bufalin against HCC angiogenesis. Molecular docking and Western Blot were utilized to validate the findings. RESULTS Analysis through IVIM and MVD showed that bufalin could inhibit HCC angiogenesis in nude mice models. A total of 159 common targets of bufalin and HCC were identified by network pharmacology. GO analysis revealed that these targets focused on multiple angiogenesis-related biological processes, including endothelial cell proliferation and migration, sprouting angiogenesis, and regulation of angiogenesis. The KEGG enrichment results suggested that bufalin could regulate multiple signaling pathways to inhibit HCC angiogenesis, including VEGF, MAPK, PI3K-Akt, mTOR, and HIF-1 signaling pathways. MAPK1, MAPK14, PRKCA, EIF4E, and APEX1 might be critical targets in regulating the above pathways. The molecular docking and Western blot analysis verified the effects of bufalin on target proteins. CONCLUSION This study demonstrated that bufalin might inhibit HCC angiogenesis by regulating multiple targets and pathways. These findings offer theoretical insights and experimental foundations for the clinical application and commercial development of bufalin in the treatment of HCC.
Collapse
Affiliation(s)
- Yuanchao Li
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Lingwei Zhou
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Kang Sun
- Department of Laboratory, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Ran Guo
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Zehua Li
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Qingqing Wen
- GE Healthcare, MR Research, Beijing, PK, People's Republic of China
| | - Guifeng Fu
- GE Healthcare, MR Research, Beijing, PK, People's Republic of China
| | - Shuohui Yang
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China.
| |
Collapse
|
18
|
Chen YP, Li M, Liu Z, Wu J, Chen F, Zhang S. Inhibition of Tyrosinase and Melanogenesis by Carboxylic Acids: Mechanistic Insights and Safety Evaluation. Molecules 2025; 30:1642. [PMID: 40286213 PMCID: PMC11990924 DOI: 10.3390/molecules30071642] [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: 03/18/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025] Open
Abstract
It is well established that certain carboxylic acid compounds can effectively inhibit tyrosinase activity. This study investigated the mechanisms by which four carboxylic acid compounds-3-phenyllactic acid, lactic acid, L-pyroglutamic acid, and malic acid-inhibit tyrosinase and melanogenesis. IC50 values for mushroom tyrosinase inhibition ranged from 3.38 to 5.42 mM, with 3-phenyllactic acid (3.50 mM), lactic acid (5.42 mM), and malic acid (3.91 mM) exhibiting mixed-type inhibition, while L-pyroglutamic acid (3.38 mM) showed competitive inhibition, as determined by enzymatic kinetic analysis. Additionally, the acidification effects of lactic acid, L-pyroglutamic acid, and malic acid contributed to the reduction in tyrosinase activity. Furthermore, all four carboxylic acid compounds effectively inhibited DOPA auto-oxidation (IC50 = 0.38-0.66 mM), ranking in potency as follows: malic acid (0.38 mM) > lactic acid (0.57 mM) > 3-phenyllactic acid (0.63 mM) > L-pyroglutamic acid (0.66 mM). These compounds also demonstrated a dose-dependent reduction in melanin production in B16-F10 cells. Proteomic analysis further revealed that these compounds not only inhibit key proteins involved in melanin synthesis, such as tyrosinase, tyrosinase-related protein 1, and tyrosinase-related protein 2, but also potentially modulate other genes associated with melanogenesis and metabolism, including Pmel, Slc45a2, Ctns, Oca2, and Bace2. Network toxicology analysis indicated that these four compounds exhibit a low risk of inducing dermatitis. These findings suggest that these compounds may indirectly regulate melanin-related pathways through multiple mechanisms, highlighting their potential for further applications in cosmetics and pharmaceuticals.
Collapse
Affiliation(s)
- Yu-Pei Chen
- The School of Public Health, Fujian Medical University, Fuzhou 350122, China;
- The School of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, China; (Z.L.); (J.W.); (F.C.); (S.Z.)
- Engineering Research Center of Natural Cosmeceuticals College of Fujian Province, Xiamen Medical College, Xiamen 361023, China
| | - Mingyu Li
- The School of Public Health, Fujian Medical University, Fuzhou 350122, China;
| | - Zirong Liu
- The School of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, China; (Z.L.); (J.W.); (F.C.); (S.Z.)
| | - Jinxiong Wu
- The School of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, China; (Z.L.); (J.W.); (F.C.); (S.Z.)
| | - Fangfang Chen
- The School of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, China; (Z.L.); (J.W.); (F.C.); (S.Z.)
- Engineering Research Center of Natural Cosmeceuticals College of Fujian Province, Xiamen Medical College, Xiamen 361023, China
| | - Shudi Zhang
- The School of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, China; (Z.L.); (J.W.); (F.C.); (S.Z.)
- Engineering Research Center of Natural Cosmeceuticals College of Fujian Province, Xiamen Medical College, Xiamen 361023, China
| |
Collapse
|
19
|
Deng Y, Cui J, Jiang Y, Zhang J, Jiang J, Zhang Q, Hu Y. Exploring the Nutraceutical Potential of a Food-Medicine Compound for Metabolic-Associated Fatty Liver Disease via Lipidomics and Network Pharmacology. Foods 2025; 14:1257. [PMID: 40238509 PMCID: PMC11988326 DOI: 10.3390/foods14071257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a prevalent global health issue closely tied to dietary habits, impacting a significant portion of the adult population. MAFLD is linked to various metabolic disorders, elevating risks of cirrhosis and hepatocellular carcinoma and severely impacting patients' quality of life. While therapeutic research has progressed, effective food-based interventions remain scarce. Natural products, rich in bioactive compounds and offering health benefits, have gained attention for their potential in managing MAFLD. This study employed network pharmacology and lipidomics to investigate the therapeutic effects of Food and Medicine Homology (FMH) on MAFLD using a high-fat-diet-induced HepG2 cell model. We identified 169 potential bioactive components from Radix Puerariae, Hericium erinaceus, Rhizoma Curcumae longae, Camellia oleifera, and Hoveniae Dulcis Semen, constructing a drug-component-target network that highlighted 34 key targets. The characteristic components of this FMH compound solution (HSD) were identified using UPLC-QTOF-MS/MS. In vitro, HSD significantly reduced intracellular lipid accumulation, decreased inflammatory markers, and mitigated hepatocyte damage. Lipidomics analysis revealed significant alterations in lipid metabolites, suggesting HSD's potential to modulate sphingolipid and glycerophospholipid metabolism, thus improving MAFLD outcomes. This research underscores the critical role of the FMH complex in modulating lipid metabolism and inflammatory pathways, offering valuable insights for developing FMH-based dietary supplements and functional foods to alleviate MAFLD. By leveraging the synergistic effects of natural compounds, our findings hold significant implications for innovative nutritional strategies in managing this prevalent metabolic disorder.
Collapse
Affiliation(s)
- Yuru Deng
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; dengyuru-@njtech.edu.cn (Y.D.); (J.Z.); (J.J.); (Y.H.)
| | - Jie Cui
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; dengyuru-@njtech.edu.cn (Y.D.); (J.Z.); (J.J.); (Y.H.)
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China;
| | - Yuxuan Jiang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China;
| | - Jian Zhang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; dengyuru-@njtech.edu.cn (Y.D.); (J.Z.); (J.J.); (Y.H.)
| | - Jinchi Jiang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; dengyuru-@njtech.edu.cn (Y.D.); (J.Z.); (J.J.); (Y.H.)
| | - Quanbin Zhang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China;
| | - Yonghong Hu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; dengyuru-@njtech.edu.cn (Y.D.); (J.Z.); (J.J.); (Y.H.)
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China;
| |
Collapse
|
20
|
Wang D, Scalici A, Wang Y, Lin H, Pitsillides A, Heard-Costa N, Cruchaga C, Ziegemeier E, Bis JC, Fornage M, Boerwinkle E, De Jager PL, Wijsman E, Dupuis J, Renton AE, Seshadri S, Goate AM, DeStefano AL, Peloso GM. Frequency of variants in Mendelian Alzheimer's disease genes within the Alzheimer's Disease Sequencing Project. J Alzheimers Dis 2025; 104:841-851. [PMID: 40084664 DOI: 10.1177/13872877251320375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
BackgroundPrior studies examined variants within presenilin-2 (PSEN2), presenilin-1 (PSEN1), and amyloid precursor protein (APP) genes. However, previously-reported clinically-relevant variants and other predicted damaging missense (DM) variants have not been characterized in a newer release of the Alzheimer's Disease Sequencing Project (ADSP).ObjectiveTo characterize previously-reported clinically-relevant variants and DM variants in PSEN2, PSEN1, APP within the participants from the ADSP.MethodsWe identified rare variants (MAF < 1%) in PSEN2, PSEN1, and APP in 14,641 individuals with whole genome sequencing and 16,849 individuals with whole exome sequencing available (Ntotal = 31,490). We additionally curated variants from ClinVar, OMIM, and Alzforum and report carriers of variants in clinical databases as well as predicted DM variants in these genes.ResultsWe detected 31 previously-reported clinically-relevant variants with alternate alleles observed within the ADSP: 4 variants in PSEN2, 25 in PSEN1, and 2 in APP. The overall variant carrier rate for the 31 clinically-relevant variants in the ADSP was 0.3%. We observed that 79.5% of the variant carriers were cases compared to 3.9% were controls. In those with AD, the mean age of onset of AD among carriers of these clinically-relevant variants was 19.6 ± 1.4 years earlier compared with noncarriers (p = 7.8 × 10-57). Additionally, we identified 197 rare variants (MAF < 1%) within ADSP participants not reported in known clinical databases.ConclusionsA small proportion of individuals in the ADSP are carriers of a previously-reported clinically-relevant variant allele for AD and these participants have significantly earlier age of AD onset compared to noncarriers.
Collapse
Affiliation(s)
- Dongyu Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alexandra Scalici
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yanbing Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Achilleas Pitsillides
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Nancy Heard-Costa
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- NHLBI Framingham Heart Study, Framingham, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Ellen Ziegemeier
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Ellen Wijsman
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alan E Renton
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sudha Seshadri
- NHLBI Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's Disease and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- NHLBI Framingham Heart Study, Framingham, MA, USA
| | - Gina M Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
21
|
Wang J, Wang H, Kang X, Wang X, Li X, Guo J, Jing X, Chu X, Han X. Integrated network pharmacology, molecular docking, and animal experiments to reveal the potential mechanism of hesperetin on COPD. Sci Rep 2025; 15:11024. [PMID: 40164657 PMCID: PMC11958725 DOI: 10.1038/s41598-025-95810-4] [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: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Hesperetin (HE), a natural flavonoid exhibiting anti-inflammatory and antioxidant properties, holds significant potential in treating chronic obstructive pulmonary disease (COPD). Nonetheless, the precise mechanisms underlying its effects are yet to be fully elucidated. In this study, we aim to explore the role and potential mechanism of HE in treating COPD using network pharmacology, molecular docking and experimental validation. We screened for HE and COPD-related targets from public databases, and then imported potential targets into a STRING database to establish a protein-protein interaction network. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes enrichment analysis were performed to obtain key signaling pathways. We then predicted the binding interactions between HE and core targets using molecular docking. The animal model of COPD was established through lipopolysaccharide and cigarette smoke induction in mice to observe lung function, inflammatory factors, pathology, and the expression of related proteins. Network pharmacology findings unveiled that HE and COPD shared 105 common targets. MAPKs and NF-κB signaling pathways were selected for further validation. In animal experiment, HE enhanced lung function and histopathological morphology, while reducing inflammation levels. The results of Western blot tests indicated that HE treatment considerably inhibited the expression of MAPKs and NF-κB. HE effectively reduced lung inflammation and improved lung function in mice. This mechanism may be achieved by inhibition of MAPKs and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Jingxi Wang
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Industrial Technology Institute for Traditional Chinese Medicine Preparation, Shijiazhuang, China
| | - Hongyang Wang
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xin Kang
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiaotian Wang
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xi Li
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jie Guo
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xuan Jing
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China.
- Hebei Industrial Technology Institute for Traditional Chinese Medicine Preparation, Shijiazhuang, China.
| | - Xi Chu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China.
| |
Collapse
|
22
|
Kwiatkowska KM, Garagnani P, Bonafé M, Bacalini MG, Sala C, Castellani G, Gentilini D, Calzari L, Ziegler D, Gerrits MM, Faber CG, Malik RA, Marchi M, Salvi E, Lauria G, Pirazzini C. High-Resolution Whole-Genome DNA Methylation Revealed Unique Signatures of Painful Diabetic Neuropathy. Diabetes 2025; 74:640-650. [PMID: 39774670 PMCID: PMC11926268 DOI: 10.2337/db24-0930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
ARTICLE HIGHLIGHTS Approximately one out of two patients with diabetes develops diabetic neuropathy; of these, 20% experience neuropathic pain. Risk factors for neuropathic pain are largely unknown; however, DNA methylation was recently associated with neuropathies and degeneration of nerve fibers. The aim of this work was to describe the DNA methylation signature and identify genes associated with neuropathic pain in type 2 diabetes mellitus (T2DM). We discovered distinct DNA methylation signatures that differentiate painful and painless neuropathy phenotypes associated with T2DM and identified genes with potential as therapeutic targets for neuropathic pain, such as GCH1, MYT1L, and MED16. This work can serve as reference hallmark for future studies on painful diabetic neuropathy and other chronic pain conditions.
Collapse
Affiliation(s)
| | - Paolo Garagnani
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Massimiliano Bonafé
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maria G. Bacalini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Claudia Sala
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Gastone Castellani
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Luciano Calzari
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Monique M. Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Catharina G. Faber
- Department of Neurology, Institute of Mental Health and Neuroscience, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Rayaz A. Malik
- Institute of Cardiovascular Sciences, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, U.K
- Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Margherita Marchi
- Neuroalgology Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico “Carlo Besta”, Milan, Italy
| | - Erika Salvi
- Neuroalgology Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico “Carlo Besta”, Milan, Italy
| | - Giuseppe Lauria
- Neuroalgology Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico “Carlo Besta”, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Chiara Pirazzini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
23
|
Ma J, Chen W, Vaishnani DK, Wang C, Xue S, Yang Q, Tong Y, Lei N, Zhao Z, Ying F. Curcumin Analog J7 Attenuates Liver Fibrosis and Metabolic Dysregulation in a Rat Model of Type 2 Diabetes via Modulation of TGF-β/Smad and NF-κB/BCL-2/BAX Pathways. Drug Des Devel Ther 2025; 19:2411-2432. [PMID: 40190815 PMCID: PMC11971964 DOI: 10.2147/dddt.s511372] [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: 12/20/2024] [Accepted: 03/23/2025] [Indexed: 04/09/2025] Open
Abstract
Objective To evaluate the therapeutic potential of the curcumin analog J7 in protecting the liver and regulating glucose and lipid metabolism in rats with type 2 diabetes. Methods Bioinformatics methods were used to identify signaling pathways linked to diabetic liver disease. Diabetic rats were treated with curcumin, low-dose J7, or high-dose J7, and liver function and fibrosis were assessed through biochemical analyses, histopathology, immunohistochemistry, and ELISA. Results J7 administration significantly improved lisver function, reduced fibrosis, and regulated metabolic profiles in diabetic rats. J7 downregulated TGF-β1, NF-κB p65, and BAX, while upregulating BCL-2, showing superior effects to traditional curcumin in reducing TGF-β1 and inhibiting α-SMA expression. Conclusion J7 demonstrates potential as a therapeutic agent for managing liver complications in type 2 diabetes, effectively attenuating liver fibrosis and regulating metabolism through the modulation of key signaling pathways and proteins.
Collapse
Affiliation(s)
- Jun Ma
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Wei Chen
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Deep K Vaishnani
- School of International Studies, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Congying Wang
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Shuman Xue
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Qiuqin Yang
- School of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Yuheng Tong
- School of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Ningjia Lei
- Pharmacy College, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People’s Republic of China
| | - Zhichao Zhao
- Department of Critical Care Medicine, Yuyao People’s Hospital, Yuyao, Zhejiang, 315400, People’s Republic of China
| | - Furong Ying
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| |
Collapse
|
24
|
Shen R, Wang J, Zhao Y, Dang Z, Zhang K, Li M, Yang Q, Gao LN. Polysaccharides from Scrophularia ningpoensis Hemsl. improve reserpine-induced depression-like behavior by inhibiting HTR2A/HTR2C mediated AKT/GSK3β/β-catenin/CBP/BDNF signalling. Int J Biol Macromol 2025; 301:140445. [PMID: 39884598 DOI: 10.1016/j.ijbiomac.2025.140445] [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: 07/15/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Scrophularia ningpoensis Hemsl. is a traditional Chinese medicine used to regulate blood sugar levels, immunity, etc. We previously isolated polysaccharides from S. ningpoensis Hemsl. (SNPS) and innovatively observed that SNPS exhibit antidepressant properties; however, the underlying mechanism is still unclear. Here, we employed network pharmacology to predict the potential targets and antidepressant mechanism of SNPS. Accordingly, we detected the effects of SNPS on monoamine neurotransmitter synthesis, metabolism, receptor expression and signal transduction in reserpine (RES)-treated mice using ELISA, HPLC-electrochemistry, metabonomics, Golgi-Cox staining and Western blotting. Finally, the mechanism of SNPS on key targets (HTR2A and HTR2C) was verified in vivo and in vitro. Results showed that SNPS ameliorated depression by restoring monoamine neurotransmitter homeostasis and hippocampal neurogenesis. SNPS reversed the depletion of 5-HT, NE and DA by activating the tryptophan (Trp)/5-HT and tyrosine (Tyr)/DA/NE metabolic pathways. SNPS decreased HTR2A and HTR2C contents, leading to the phosphorylation of AKT and GSK3β, followed by increases in β-catenin, CBP and BDNF levels. Mechanistically, SNPS reduced the levels of HTR2A and HTR2C proteins by inhibiting their mRNA transcription, rather than inducing protein degradation. In conclusion, by inhibiting the transcription of HTR2A and HTR2C, SNPS activated the AKT/GSK3β/β-catenin/CBP/BDNF pathway, thereby exerting dose-dependent antidepressant effects.
Collapse
Affiliation(s)
- Ruhui Shen
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China; College of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, PR China; Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai 20080, PR China
| | - Jian'an Wang
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Yijin Zhao
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Zhaojin Dang
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Ke Zhang
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Ming Li
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Qian Yang
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Li-Na Gao
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China; College of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, PR China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong 272067, PR China; Jining Key Laboratory of Depression Prevention and Treatment, Jining Medical University, Jining, Shandong 272067, PR China.
| |
Collapse
|
25
|
Bao ZC, Zhang Y, Liu ZD, Dai HJ, Ren F, Li N, Lv SY, Zhang Y. Tetrahydrocurcumin-induced apoptosis of hepatocellular carcinoma cells involves the TP53 signaling pathway, as determined by network pharmacology. World J Gastrointest Oncol 2025; 17:101174. [PMID: 40092919 PMCID: PMC11866214 DOI: 10.4251/wjgo.v17.i3.101174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/04/2024] [Accepted: 01/16/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant disease with high incidence and mortality worldwide. This study focuses on the TP53 target protein to investigate the potential therapeutic effect of tetrahydrocurcumin (THC) on HCC and its mechanism of action. The research hypothesis is that THC can inhibit the proliferation, migration, and invasion of HCC cells, and promote their apoptosis by regulating the TP53 target protein. AIM To explore the mechanism by which THC inhibits HCC cell proliferation via the TP53 signaling pathway. METHODS Potential targets of THC and HCC were identified from multiple databases. The core targets were subjected to analyses using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases, and visualization processing, using the online platform Metascape to identify the key molecules and signaling pathways involved in the action of THC against HCC. The molecular mechanisms of action of THC against TP53 in the inhibition of HCC cells were verified using cell counting kit-8, Transwell, apoptosis, and western blotting assays. RESULTS Molecular docking results showed that THC had a high score for the TP53 target protein. In vitro experiments indicated that THC effectively inhibited the proliferation and migration of HCC cells, and affected the expression levels of TP53, MDM2, cyclin B, Bax, Bcl-2, caspase-9, and caspase-3. CONCLUSION THC induces the apoptosis of HCC cells through the TP53 signaling pathway, thereby inhibiting their proliferation and migration.
Collapse
Affiliation(s)
- Zhuo-Cong Bao
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ye Zhang
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Zhao-Dong Liu
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Hui-Jun Dai
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Fu Ren
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ning Li
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Shang-Yu Lv
- Department of Clinical Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Yan Zhang
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- International Education School, International Exchange and Cooperation Office, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| |
Collapse
|
26
|
Bao ZC, Liu ZD, Zhang Y, Dai HJ, Jia H, Ren F, Li N, Zhao L, Wang YW, Lv SY, Zhang Y. To investigate the effect and mechanism of tetrahydrocurcumin on hepatocellular carcinoma based on phosphoinositide 3-kinases/AKT signaling pathway. World J Gastrointest Oncol 2025; 17:102187. [PMID: 40092949 PMCID: PMC11866248 DOI: 10.4251/wjgo.v17.i3.102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/20/2024] [Accepted: 01/02/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Liver cancer has a high incidence and mortality worldwide, especially in China. Herein, we investigated the therapeutic effect and mechanism of tetrahydrocurcumin against hepatocellular carcinoma (HCC), with a focus on the of phosphoinositide 3-kinases (PI3K)/AKT signaling pathway. AIM To investigate the effects and mechanism of tetrahydrocurcumin in HCC cell lines HepG2 and Huh7. METHODS Using Metascape, we analyzed the potential targets of tetrahydrocurcumin in HCC. Molecular docking validation was performed using SYBYL2.0. Cell Counting Kit-8, wound healing, and transwell assays were performed to evaluate the effects of tetrahydrocurcumin on HepG2 and Huh7 cell migration, invasion, and apoptosis. The expression of PI3K/AKT signaling pathway-related proteins was detected by western blotting. RESULTS Network pharmacology and molecular docking showed that tetrahydrocurcumin has high binding affinity for phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha. In vitro experiments demonstrated that tetrahydrocurcumin suppressed the migration and invasion of liver cancer cells, promoted their apoptosis, and downregulated the expression of p-PI3K, p-AKT, and B cell leukemia/lymphoma 2, while upregulating caspase-3, p53, and B cell leukemia/lymphoma 2 associated X. CONCLUSION In summary, tetrahydrocurcumin suppresses PI3K/AKT signaling, promotes apoptosis, and prevents the migration and invasion of liver cancer cells.
Collapse
Affiliation(s)
- Zhuo-Cong Bao
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Zhao-Dong Liu
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ye Zhang
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Hui-Jun Dai
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Hui Jia
- School of Traditional Chinese Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Fu Ren
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ning Li
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Lu Zhao
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Yi-Wei Wang
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Molecular Morphology Laboratory, College of Basic Medical Sciences, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Shang-Yu Lv
- Batch 2022, Clinical Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Yan Zhang
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- International Education School, International Exchange and Cooperation Office, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| |
Collapse
|
27
|
He M, Shi Y, Han F, Cai Y. Prediction of adverse drug reactions based on pharmacogenomics combination features: a preliminary study. Front Pharmacol 2025; 16:1448106. [PMID: 40129949 PMCID: PMC11931068 DOI: 10.3389/fphar.2025.1448106] [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: 06/12/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Introduction Adverse Drug Reactions (ADRs), a widespread phenomenon in clinical drug treatment, are often associated with a high risk of morbidity and even death. Drugs and changes in gene expression are the two important factors that affect whether and how adverse reactions occur. Notably, pharmacogenomics data have recently become more available and could be used to predict ADR occurrence. However, there is a challenge in effectively analyzing the massive data lacking guidance on mutual relationship for ADRs prediction. Methods We constructed separate similarity features for drugs and ADRs using pharmacogenomics data from the Comparative Toxicogenomics Database [CTD, including Chemical-Gene Interactions (CGIs) and Gene-Disease Associations (GDAs)]. We proposed a novel deep learning architecture, DGANet, based on the constructed features for ADR prediction. The algorithm uses Convolutional Neural Networks (CNN) and cross-features to learn the latent drug-gene-ADR associations for ADRs prediction. Results and Discussion The performance of DGANet was compared to three state-of-the-art algorithms with different genomic features. According to the results, GDANet outperformed the benchmark algorithms (AUROC = 92.76%, AUPRC = 92.49%), demonstrating a 3.36% AUROC and 4.05% accuracy improvement over the cutting-edge algorithms. We further proposed new genomic features that improved DGANet's predictive capability. Moreover, case studies on top-ranked candidates confirmed DGANet's ability to predict new ADRs.
Collapse
Affiliation(s)
- Mingxiu He
- College of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Information, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yiyang Shi
- College of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Fangfang Han
- College of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangzhou, China
- Guangdong Provincial Traditional Chinese Medicine Precision Medicine Big Data Engineering Technology Research Center, Guangzhou, China
| | - Yongming Cai
- College of Medical Information and Engineering, Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangzhou, China
- Guangdong Provincial Traditional Chinese Medicine Precision Medicine Big Data Engineering Technology Research Center, Guangzhou, China
| |
Collapse
|
28
|
Islam KMT, Mahmud S. In-silico exploring pathway and mechanism-based therapeutics for allergic rhinitis: Network pharmacology, molecular docking, ADMET, quantum chemistry and machine learning based QSAR approaches. Comput Biol Med 2025; 187:109754. [PMID: 39908918 DOI: 10.1016/j.compbiomed.2025.109754] [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: 08/08/2024] [Revised: 01/07/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
Allergic rhinitis is a devastating health complication that interrupts the quality of daily life and significantly affects around 40 % of the population worldwide. Despite the availability of various treatment options, many patients continue to struggle with persistent symptoms and side effects, highlighting the need for innovative therapeutic approaches. Therefore, identifying pathway and mechanism-based targeted therapies with more effective and fewer side effects could aid current therapeutics and provide novel therapeutic advantages. This study aimed to identify potential drug candidates for allergic rhinitis treatment by employing in-silico approaches, including network pharmacology, molecular docking, ADMET, similarity, pharmacophore modeling, quantum chemistry, and machine learning-based QSAR modeling. From three traditionally used medicinal plants known as allergic rhinitis curing, Xanthium strumarium, Magnolia liliiflora, and Tylophora indica, 241 compounds were obtained, and their favorable ADMET properties were analyzed. Network pharmacology revealed 203 potential therapeutic targets, with 15 hub targets identified through protein-protein interaction network analysis and most of them play key roles in inflammatory and immune pathways confirmed by KEGG pathway analysis. Molecular docking, similarity testing, and pharmacophore modeling studies identified promising compounds Quercetin, Yinyanghuo E, Uralenin, CID:90643991, CID:42607537, CID:76329670, Heracetin, and Fisetin exhibiting strong binding affinities with key regulatory targets, NFKB1, TRAF6, and key cytokines IL5, and IL6 that directly and indirectly involved in allergic reactions. Quantum chemistry calculations revealed favorable electronic properties and reactivities of these compounds. The machine learning-based QSAR model predicted IC50 < 50 nM for almost all compounds, indicating highly potent inhibitors. Hence, this in-silico study identified some novel promising drug candidates for treating allergic rhinitis by targeting crucial inflammatory and immune pathways, offering improved treatment outcomes and reduced side effects, subject to further experimental validation.
Collapse
Affiliation(s)
- K M Tanjida Islam
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
| | - Shahin Mahmud
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh.
| |
Collapse
|
29
|
Vaghasiya MD, Sain A, Mendapara JV, Khamrai D, Naskar D, Kumari P. Thiazepine-Based Hybrids as Promising Anti-Colon Cancer Agents: Design, Synthesis, Computational and In Vitro Screening. Chem Biodivers 2025; 22:e202401550. [PMID: 39413279 DOI: 10.1002/cbdv.202401550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/20/2024] [Accepted: 10/15/2024] [Indexed: 10/18/2024]
Abstract
Novel thiazepine-based hybrids (9 a-d) were designed and synthesized to create lead molecules with exceptional anti-colon cancer efficacy. Analytical methods, including IR, NMR, and HR-MS, characterized the synthesized compounds. The in vitro colorectal study was carried out to compare the biological activity of newly developed compounds with the computational data. The tested compounds induced cytotoxicity in HT-29 cells for both 24 h and 48 h in a dose-dependent manner. However, compound 9 a induced cytotoxicity at much higher concentrations compared to the rest of the compounds. 9 b and 9 c caused 50 % cell death (compared to the untreated cells) at a dose of ~50 μM and 40 μM in case of 24-hour exposure, respectively. On the contrary, for 48 h exposure, both 9 b and 9 c induced 50 % cell death concerning untreated cells at a dose of around ~20 μM, whereas 9 d exhibited 50 % cell death at 5 μM in the case of 48 h exposure. In silico ADMET was also carried out to understand the pharmacokinetics and safety profiles of the drug candidates. We found some of the critical targets of these compounds, which eventually will be integral to exploring the mechanistic actions of these compounds in colon cancer.
Collapse
Affiliation(s)
- Mahesh D Vaghasiya
- Department of Chemistry, S.V. National Institute of Technology, Surat, Gujarat, 395007, India
| | - Arindam Sain
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, Nadia, West Bengal, 741249, India
| | - Jigarkumar V Mendapara
- Department of Chemistry, S.V. National Institute of Technology, Surat, Gujarat, 395007, India
| | - Dipshikha Khamrai
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, Nadia, West Bengal, 741249, India
| | - Debdut Naskar
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, Nadia, West Bengal, 741249, India
| | - Premlata Kumari
- Department of Chemistry, S.V. National Institute of Technology, Surat, Gujarat, 395007, India
| |
Collapse
|
30
|
Zhou M, Jiang Z, Zhang M, Feng S, Ma B, Kan S, Fu X, Zhu R. Exploring the molecular mechanism of icariin improving spinal cord injury through network pharmacology combined with experimental verification. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03904-7. [PMID: 40014127 DOI: 10.1007/s00210-025-03904-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/08/2025] [Indexed: 02/28/2025]
Abstract
This study aimed to investigate the potential pharmacological effects of icariin (ICA) in the treatment of spinal cord injury (SCI). Network pharmacology was used to focus on the potential targets and biological processes of ICA in SCI. Molecular docking was used to verify the ability of ICA to bind to its core targets. Finally, valuate the efficacy and potential mechanisms of ICA in treating spinal cord injury through in vitro and in vivo experiments. A total of 37 targets were screened out, and core genes were screened out from the protein‒protein interaction network. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that these targets are enriched mainly in response to hypoxia, regulation of the cellular response to stress, and the TGF-beta signaling pathway. Molecular docking analysis showed that ICA has good docking ability with core targets. In animal experiments, Basso, Beattie and Bresnahan scores, catwalk gait analysis, hematoxylin and eosin staining, and RT-qPCR showed that ICA can inhibit spinal cord inflammation and effectively improve the behavioral and histological recovery after SCI rats. Western blot and immunofluorescence showed that ICA can reduce astrocyte activation and downregulate the TGF-beta signaling pathway after SCI. In addition, ICA can promote axonal nerve elongation and promotes angiogenesis after spinal cord injury in rats. In vitro experiments revealed that ICA can inhibit TGFβ1-induced activation of the TGF-beta signaling pathway and astrocyte activation. ICA treats SCI through multiple targets and pathways. ICA plays a major role in protecting nerves, promoting angiogenesis, and inhibiting reactive astrocyte activation in the treatment of SCI.
Collapse
Grants
- 2022JZXK05, 2022JZXK02, 2023YJZD002, 2022JZXK06 Tianjin Union Medical Center
- 2022JZXK05, 2022JZXK02, 2023YJZD002, 2022JZXK06 Tianjin Union Medical Center
- 2022JZXK05, 2022JZXK02, 2023YJZD002, 2022JZXK06 Tianjin Union Medical Center
- 2022JZXK05, 2022JZXK02, 2023YJZD002, 2022JZXK06 Tianjin Union Medical Center
- TJYXZDXK-064B Tianjin Key Medical Discipline (Specialty) Construction Project
- 2021125 Tianjin Municipal Health Commission's Integrated Traditional Chinese Medicine and Western Medicine Project
- 202305 Tianjin Health Commission Science and Technology Project
- TJWJ2024QN045 Tianjin Health Science and Technology Project
- 2023RC006 Yunnan Province talented Xing border plan
Collapse
Affiliation(s)
- Mengmeng Zhou
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China
- Tianjin Institute of Rehabilitation, Tianjin, China
| | - Zehua Jiang
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China
- Tianjin Institute of Rehabilitation, Tianjin, China
| | - Maosen Zhang
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
| | - Sa Feng
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
| | - Boyuan Ma
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Shunli Kan
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China
- Tianjin Institute of Rehabilitation, Tianjin, China
| | - Xuanhao Fu
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China
- Tianjin Institute of Rehabilitation, Tianjin, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China.
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China.
- Tianjin Institute of Rehabilitation, Tianjin, China.
| |
Collapse
|
31
|
Xiao F, Huang C, Chen A, Xiao W, Li Z. Identification of metabolite-disease associations based on knowledge graph. Metabolomics 2025; 21:32. [PMID: 39987424 DOI: 10.1007/s11306-025-02227-1] [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: 09/15/2024] [Accepted: 01/25/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Despite the insights that metabolite analysis can provide into the onset, development, and progression of diseases-thus offering new concepts and methodologies for prevention, diagnosis, and treatment-traditional wet lab experiments are often time-consuming and labor-intensive. Consequently, this study aimed to develop a machine learning model named COM-RAN, which is based on a knowledge graph and random forest algorithm, to identify potential associations between metabolites and diseases. METHODS Firstly, we integrated the known associations between diseases and metabolites. Secondly, we provided a synthesis of the extant data regarding diseases and metabolites, accompanied by supplementary information pertinent to these entities. Thirdly, knowledge graph-based embedded features were used to characterize disease-metabolite associations. Finally, a random forest algorithm was employed to construct a model for identifying potential disease-metabolite associations. RESULTS The experimental results demonstrated that the proposed model achieved an Area Under the Receiver Operating Characteristic Curve (AUC) of 0.968 in 5-fold cross-validations, while the Area Under the Precision-Recall Curve (AUPR) was 0.901, outperforming the vast majority of existing prediction methods. The case studies corroborated the majority of the novel associations identified by COM-RAN, thereby further demonstrating the reliability of the current method in predicting the potential relationship between metabolites and diseases. CONCLUSION The COM-RAN model demonstrated promise in predicting associations between diseases and metabolites, suggesting that integrating knowledge graphs with machine learning methodologies can significantly improve the accuracy and reliability of predictions related to disease-associated metabolites.
Collapse
Affiliation(s)
- Fuheng Xiao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, P.R. China
| | - Canling Huang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, P.R. China
| | - Ali Chen
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, P.R. China
| | - Wei Xiao
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, P.R. China.
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, P.R. China.
| | - Zhanchao Li
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, P.R. China.
| |
Collapse
|
32
|
Pan J, Chai X, Li C, Wu Y, Ma Y, Wang S, Xue Y, Zhao Y, Chen S, Zhu X, Zhao S. Eucommia ulmoides Oliv. Bark Extracts Alleviate MCAO/Reperfusion-Induced Neurological Dysfunction by Suppressing Microglial Inflammation in the Gray Matter. Int J Mol Sci 2025; 26:1572. [PMID: 40004043 PMCID: PMC11855810 DOI: 10.3390/ijms26041572] [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: 12/24/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Ischemic stroke ranks as the second leading cause of global mortality. The limited time for effective thrombolytic treatment has prompted the exploration of alternative prevention approaches. Eucommia ulmoides (E. ulmoides) Oliv. bark has shown multiple pharmacological effects, including neuroprotection, anti-inflammation and autophagy modulation. This study aims to elucidate the neuroprotective effects of water extract of E. ulmoides (WEU) supplementation in a middle cerebral artery occlusion (MCAO) mouse model and to further explore the underlying molecular mechanisms. Seven bioactive compounds in WEU-aucubin, chlorogenic acid, geniposidic acid, quercetin, protocatechuic acid, betulin and pinoresinol diglucoside-were identified using HPLC-MS. Our results showed that WEU supplementation significantly decreased infarct volume and ameliorated neurological dysfunction in mice following MCAO/reperfusion (MCAO/R) injury. Furthermore, the administration of WEU significantly attenuated microglia activation induced by cortical ischemia in mice and inhibited the production of pro-inflammatory mediators, including interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Importantly, in contrast with the vehicle group, the protein expression levels of Toll-like receptor 4 (TLR4), phospho-p38 (p-p38) and nuclear factor kappa B (NF-κB) were reduced in the WEU group. Therefore, this present study provides evidence that E. ulmoides improves neurological behaviors by suppressing neuroinflammation and inhibiting the activation of the TLR4/ p38 MAPK and NF-κB pathways in mice after ischemia, which indicates that E.ulmoides is a promising candidate for alleviating gray matter ischemic change.
Collapse
Affiliation(s)
- Jiarong Pan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Xuejun Chai
- College of Basic Medicine, Xi’an Medical University, Xi’an 710021, China;
| | - Cixia Li
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China;
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Songlin Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Yuhuan Xue
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Yongkang Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| |
Collapse
|
33
|
Xia Q, Hu J, Jiao Z, Wang G, Sun J, Pang X, Ma Y, Huang Y, Liang X, Guo J, Peng C, Jin C, Jia X, Gui S. Exploring the mechanisms of Yang Wei Shu granule for the treatment of chronic atrophic gastritis using UPLC-QTOF-MS/MS, network pharmacology, and cell experimentation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119326. [PMID: 39798675 DOI: 10.1016/j.jep.2025.119326] [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: 10/31/2024] [Revised: 12/25/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic atrophic gastritis (CAG) is a global disease of the digestive system and is an important precancerous lesion in the development of gastric cancer. Yang Wei Shu granule (YWSG), which evolved from the formula "Warm Stomach Soup" of the Jin and Yuan Dynasties in China, is frequently used as a classic herbal compound in the treatment of CAG. However, the active ingredients and mechanisms by which it works are not clear. AIM OF THE STUDY To elucidate the chemical composition of YWSG and investigate the potential mechanisms of YWSG on CAG by composition analysis, network pharmacology and cellular experimental studies. MATERIALS AND METHODS The chemical and blood-entry constituents of YWSG were analyzed by ultra-high performance liquid chromatography-Quadrupole tandem time-of-flight mass spectrometry (UPLC-QTOF-MS/MS). Subsequently, potential targets of YWSG for CAG treatment were identified through utilization of publicly available online resources. The YWSG-component-target-pathway network and protein-protein interaction (PPI) network were constructed using Cytoscape software. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of potential targets was performed using the DAVID database. Finally, a cellular model of lipopolysaccharide (LPS)-activated RAW 264.7 macrophages was established and validated by in vitro experiments. RESULTS A total of 150 compounds in YWSG and 47 blood-entry constituents were identified by using UPLC-QTOF-MS/MS. Based on network pharmacology, a total of 132 target genes were identified as being involved in the therapeutic effect of YWSG on CAG. Network pharmacology and molecular docking results suggest that AKT1, PIK3CA, PTPN11, SRC and STAT3 may be potential targets of YWSG for the treatment of CAG. Cellular experiments showed that the YWSG-containing serum had no cytotoxic effect on RAW264.7 cells and could inhibit nitric oxide (NO) production and the expression of pro-inflammatory factors TNF-α, IL-6, and IL-1β. Additionally, it was observed to promote the expression of the anti-inflammatory factor IL-10 in LPS-stimulated RAW264.7 cells. The immunofluorescence results showed that YWSG treated CAG by inhibiting the PI3K-Akt pathway. CONCLUSIONS The application of UPLC-Q-TOF-MS/MS, network pharmacology and cellular experiments provided elucidation to understand the components and mechanisms of the therapeutic effects of YWSG on CAG, providing useful directions for further research.
Collapse
Affiliation(s)
- Qijun Xia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Jingjing Hu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Zhiyong Jiao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Guichun Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Jianwen Sun
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Xingyuan Pang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Yuhan Ma
- Hefei China Resources Shenlu Pharmaceutical Co. Ltd, Hefei, 230012, Anhui, China
| | - Yuzhe Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China
| | - Xiao Liang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Jian Guo
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China
| | - Chengjun Peng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China
| | - Cheng Jin
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China.
| | - Xiaoyi Jia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Shuangying Gui
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China; Anhui Engineering Research Center for Quality Improvement and Utilization of Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| |
Collapse
|
34
|
Li N, Wang B, Yang M, Feng M, Xu X, Xian CJ, Li T, Zhai Y. The Multi-Target Action Mechanism for the Anti-Periodontitis Effect of Astragali radix Based on Bioinformatics Analysis and In Vitro Verification. Nutrients 2025; 17:627. [PMID: 40004956 PMCID: PMC11858088 DOI: 10.3390/nu17040627] [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: 01/15/2025] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background:Astragali radix is a traditional Chinese medicine with potential therapeutic effects on periodontitis; however, its underlying mechanisms require further investigation. Methods: We employed network pharmacology, molecular docking, molecular dynamics simulations, and in vitro experiments to explore the potential actions and mechanisms of Astragali radix in treating periodontitis. Results: A total of 17 compounds (including the most prevalent one, Kaempferol) from Astragali radix and 464 corresponding targets were identified, from which five major active ingredients were selected based on the drug-active ingredient and periodontitis gene network. Protein-protein interaction (PPI) network analysis identified the top ten core potential targets, seven of which possess suitable crystal structures for molecular docking. These include interleukin-6 (IL6), tumor necrosis factor (TNF), AKT serine/threonine kinase 1 (AKT1), interleukin-1β (IL1β), prostaglandin G/H synthase-2 (PTGS2), matrix metalloproteinase-9 (MMP9), and caspase-3 (CASP3). Additionally, 58 Gene Ontology (GO) terms and 146 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were identified. The five major active ingredients and seven core targets mentioned above were subjected to molecular docking analysis using Discovery Studio 2019 software. Molecular dynamic simulations confirmed a stable interaction between the CASP3 and the Kaempferol ligand system. In vitro experiments indicated that Kaempferol significantly inhibited lipopolysaccharide (LPS)-induced apoptosis in human periodontal ligament stem cells and reduced the expression levels of IL6, CASP3 and MMP9. Conclusions: This study systematically elucidates that the primary active ingredients derived from Astragali radix exert their pharmacological effects (including anti-inflammation and anti-apoptosis) primarily by interacting with multiple targets. These findings establish a promising foundation for the targeted application of Astragali radix in the treatment of periodontitis.
Collapse
Affiliation(s)
- Ningli Li
- School of Stomatology, Henan University, Kaifeng 475004, China; (N.L.); (M.Y.); (M.F.); (X.X.); (T.L.)
| | - Bowen Wang
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475004, China;
| | - Mingzhen Yang
- School of Stomatology, Henan University, Kaifeng 475004, China; (N.L.); (M.Y.); (M.F.); (X.X.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475004, China;
| | - Miaomiao Feng
- School of Stomatology, Henan University, Kaifeng 475004, China; (N.L.); (M.Y.); (M.F.); (X.X.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475004, China;
| | - Xiaoran Xu
- School of Stomatology, Henan University, Kaifeng 475004, China; (N.L.); (M.Y.); (M.F.); (X.X.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475004, China;
| | - Cory J. Xian
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia;
| | - Tiejun Li
- School of Stomatology, Henan University, Kaifeng 475004, China; (N.L.); (M.Y.); (M.F.); (X.X.); (T.L.)
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yuankun Zhai
- School of Stomatology, Henan University, Kaifeng 475004, China; (N.L.); (M.Y.); (M.F.); (X.X.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475004, China;
| |
Collapse
|
35
|
Zhu G, Jiang Z, Zhu N, Wang D, Guo T, Meng Y, Zhu Y, Tan K, Hu M, Tang H, Wang X. Exploring the multi-targeted mechanism of Saikosaponin A in prostate cancer treatment: a network pharmacology and molecular docking approach. Front Pharmacol 2025; 16:1530715. [PMID: 39995416 PMCID: PMC11847854 DOI: 10.3389/fphar.2025.1530715] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
Background Prostate cancer (PCa) is one of the prevalent malignant tumors among men. It can progress to castration-resistant prostate cancer (CRPC), which is significantly more challenging to treat. Saikosaponin A (SSA), a triterpenoid saponin extracted from the genus Bupleurum, exerts numerous pharmacological effects, including anti-inflammatory and anti-tumorigenic effects. However, the mechanism underlying the effects of SSA in prostate cancer treatment remains elusive. Methods In this study, a network pharmacology approach was applied to identify relevant targets from drug- and disease-related databases, and intersections were analyzed using Venny2.1 to construct a Protein-Protein interaction (PPI) interaction network. Next, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to elucidate associated biological functions and signaling pathways. Meanwhile, molecular docking between core targets and SSA was performed using Autodock software. Lastly, in vitro experiments were performed for validation. Results A least of four key targets, namely BCL2 apoptosis regulator (BCL2), estrogen receptor 1 (ESR1), hypoxia-inducible factor 1 subunit alpha (HIF1A), and signal transducer and activator of transcription 3 (STAT3) were identified in this study, and molecular docking analyses revealed that SSA stably binds to these targets. Moreover, the results of in vitro experiments revealed that SSA significantly inhibited the proliferative and migratory abilities of PC3 cells in a dose-dependent manner. Finally, SSA also induced G1-phase blockade and apoptosis in PC3 cells, further highlighting its potential role in prostate cancer treatment. Conclusion The present study revealed that SSA exerts anti-tumorigenic effects in prostate cancer by targeting multiple pathways, laying a theoretical reference for its use as a therapeutic candidate for prostate cancer.
Collapse
Affiliation(s)
- Genbao Zhu
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Zhiming Jiang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Niuping Zhu
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
| | - Donghui Wang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
| | - Tianpeng Guo
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
| | - Yiqing Meng
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
| | - Yi Zhu
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
| | - Kemeng Tan
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Mengxue Hu
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Heng Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Xuannian Wang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
| |
Collapse
|
36
|
Stroescu R, Chiriţă-Emandi A, Puiu M, Chisavu F, Steflea R, Doroş G, Gafencu M. Nephrogenic Diabetes Insipidus Affecting Three Males in Two Generations-Case Report and Review of the Literature. CHILDREN (BASEL, SWITZERLAND) 2025; 12:195. [PMID: 40003297 PMCID: PMC11854844 DOI: 10.3390/children12020195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025]
Abstract
Background: Nephrogenic diabetes insipidus (NDI) is defined as the inability of the kidney to concentrate urine owing to the insensitivity of the distal nephron to the antidiuretic hormone, arginine vasopressin. NDI is a heterogeneous rare autosomal dominant or X-linked disease. Objective: We present a family with nephrogenic diabetes affecting three males in two generations. Methods: We report two boys with NDI: a 4-month-old infant who was treated for fever, vomiting, and failure to thrive, and his 10-year-old uncle (the mother's brother), who was admitted concurrently for consuming 11 L of fluid per day. According to family history, the mother's sibling passed away at the age of two from severe hypernatremic dehydration. Results: The infant's clinical and laboratory evaluation revealed a 7.8 mL/kg/h urine output, hypernatremic hyperchloremic alkalosis, extremely low urine density (1002), and elevated copeptin level. In contrast, the uncle's clinical and laboratory evaluation revealed marked polyuria, low urine density, and elevated copeptin, all of which were suggestive of diabetes insipidus. After starting hydrochlorothiazide treatment (2 mg/kg/body), the infant's urine production reduced (2.85 mL/kg/h); however, severe hypokalemia and alkalosis followed. Spironolactone, an aldosterone antagonist, were added, with good therapeutic response. Hydrochlorothiazide was administered to the uncle, and his daily fluid intake decreased to 3-4 L. Given the family history, Sanger sequencing for the AVPR2 variant was performed on the boys and the infant's mother. Analysis showed hemizygous likely pathogenic variant c.335G>A p. (Cys112Tyr) in the 2 boys and heterozygous (carrier) status of the mother. Within the same family, we observed phenotypic heterogeneity: one child died at the age of two, another lived well into ten years without therapy, and a four month-old baby could have had a poor outcome without specific treatment. Conclusions: NDI is a rare and possibly fatal genetic disorder with heterogeneous manifestations. In families with a history of NDI, molecular genetic testing is crucial for family planning.
Collapse
Affiliation(s)
- Ramona Stroescu
- “Louis Turcanu” Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timisoara, Romania; (R.S.); (A.C.-E.); (M.P.); (R.S.); (G.D.); (M.G.)
- Department XI of Pediatrics, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Adela Chiriţă-Emandi
- “Louis Turcanu” Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timisoara, Romania; (R.S.); (A.C.-E.); (M.P.); (R.S.); (G.D.); (M.G.)
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Maria Puiu
- “Louis Turcanu” Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timisoara, Romania; (R.S.); (A.C.-E.); (M.P.); (R.S.); (G.D.); (M.G.)
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Flavia Chisavu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine “Victor Babes”, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Ruxandra Steflea
- “Louis Turcanu” Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timisoara, Romania; (R.S.); (A.C.-E.); (M.P.); (R.S.); (G.D.); (M.G.)
- Department XI of Pediatrics, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Gabriela Doroş
- “Louis Turcanu” Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timisoara, Romania; (R.S.); (A.C.-E.); (M.P.); (R.S.); (G.D.); (M.G.)
- Department XI of Pediatrics, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Mihai Gafencu
- “Louis Turcanu” Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timisoara, Romania; (R.S.); (A.C.-E.); (M.P.); (R.S.); (G.D.); (M.G.)
- Department XI of Pediatrics, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| |
Collapse
|
37
|
Yang Z, Liang Y, Wu C, Xie H, Liu S, Sun P, Zhang Y. Kemin capsule ameliorates post-infectious cough by modulating the PI3K/AKT signaling pathway and TRPA1/TRPV1 channels. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118837. [PMID: 39306207 DOI: 10.1016/j.jep.2024.118837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/18/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kemin capsule (KMC), as an innovative traditional Chinese medicine (TCM), has shown excellent efficacy in treating PIC in China. The post-infectious cough (PIC) is a common condition in pediatrics, and the inflammatory responses to PIC are intricately linked to the immune mechanisms of the host. However, the precise mechanisms involved remain uncertain. AIM OF STUDY The objective of this research is to investigate the mechanisms by which KMC treats PIC using a combination of UPLC-MS, bioinformatics, network pharmacology, and molecular docking. The study's findings will be corroborated through in vitro and in vivo experiments. MATERIALS AND METHODS This study identified the main components of KMC using UPLC-MS. The mechanism by which these capsules treat PIC was explored through transcriptomics, network pharmacology, and molecular docking. PIC model in Balb/c mice was induced with respiratory syncytial virus (RSV) at a titer of 10^5.5 TCID50/mL. From day 14 post-infection, the mice were orally administered the capsules at doses of 0.3, 0.6, and 1.2 g/kg for two weeks. Cough was stimulated with capsaicin at 10^-4 mol/mL, and the effects on PIC mice were measured by cough frequency, latency, ELISA, and H&E staining. Expression levels of transient receptor potential (TRP) channel proteins and the PI3K/AKT signaling pathway were analyzed using RT-qPCR, immunohistochemistry (IHC), and western blot (WB). The effect of KMC on A549 cells proliferation in vitro was also assessed. RESULTS The therapeutic efficacy of KMC is potentially exerted through its inherent bioactive constituents, including deoxyandrographolide, quercetin, and chryseriol. These compounds are hypothesized to modulate the PI3K/AKT signaling pathway and influence the function of TRP channel proteins, consequently mitigating the pathological state associated with PIC. In vivo experiments have demonstrated that KMC significantly reduces the frequency of coughs and extends the cough latency period in mice with PIC. KMC mitigates airway inflammation by suppressing the production of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6. The expression or phosphorylation levels of key regulators in the PI3K/AKT/TRP axis in mouse lung tissue, including PI3K, AKT, NF-κB p65, TLR4, STAT3, TRPV1, TRPA1 were significantly reduced. CONCLUSION KMC exerts its therapeutic effect on PIC by dampening the activation of the PI3K/AKT signaling pathway and the activity of TRPA1 and TRPV1 ion channels.
Collapse
Affiliation(s)
- Zhicong Yang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, China.
| | - Yuxue Liang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, China.
| | - Chenxi Wu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, China.
| | - Huiguo Xie
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, China.
| | - Shengmei Liu
- Shandong Kangzhonghong Pharmaceutical Technology Development Co., Ltd, 250014, China.
| | - Peng Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, China.
| | - Yingying Zhang
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 250355, China.
| |
Collapse
|
38
|
Mester B, Lipták Z, Farkas-Sütő KA, Grebur K, Gyulánczi FK, Fábián A, Fekete BA, György TA, Bödör C, Kovács A, Merkely B, Szűcs A. Inherited Hypertrabeculation? Genetic and Clinical Insights in Blood Relatives of Genetically Affected Left Ventricular Excessive Trabeculation Patients. Life (Basel) 2025; 15:150. [PMID: 40003559 PMCID: PMC11856360 DOI: 10.3390/life15020150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Genetically determined left ventricular excessive trabeculation (LVET) has a wide clinical spectrum ranging from asymptomatic subjects to severe heart failure with arrhythmias and thromboembolic events. Unlike other cardiomyopathies, the relatives of LVET patients never reach the spotlight of guidelines and clinical practice, although these family members can be often affected by these conditions. Thus, we aimed to investigate the relatives of LVET by multidimensional analysis, such as genetic testing, ECG and cardiac ultrasound (ECHO). We included 55 blood relatives from the family of 18 LVET patients (male = 27, age = 44 ± 20.8y), who underwent anamnesis registration. With Sanger sequencing, the relatives were classified into genetically positive (GEN-pos) and unaffected (GEN-neg) subgroups. In addition to regular ECG parameters, Sokolow-Lyon Index (SLI) values were calculated. 2D ECHO images were analysed with TomTec Arena, evaluating LV volumetric, functional (EF) and strain parameters. Individuals were categorized into JENNI-pos and JENNI-neg morphological subgroups according to the Jenni LVET ECHO criteria. Family history showed frequent involvement (arrhythmia 61%, stroke 56%, syncope 39%, sudden cardiac death 28%, implanted device 28%), as well as personal anamnesis (subjective symptoms 75%, arrhythmias 44%). ECG and ECHO parameters were within the normal range. In terms of genetics, 78% of families and 38% of relatives carried the index mutation. LV_SLI and QT duration were lower in the GEN-pos group; ECHO parameters were comparable in the subgroups. Morphologically, 33% of the relatives met Jenni-LVET criteria were genetically affected and showed lower LV_EF values. The frequently found genetic, morphological and clinical involvement may indicate the importance of screening and, if necessary, regular follow-up of relatives in the genetically affected LVET population.
Collapse
Affiliation(s)
- Balázs Mester
- Heart and Vascular Centre, Semmelweis University, 1085 Budapest, Hungary; (B.M.)
| | - Zoltán Lipták
- Heart and Vascular Centre, Semmelweis University, 1085 Budapest, Hungary; (B.M.)
| | | | - Kinga Grebur
- Heart and Vascular Centre, Semmelweis University, 1085 Budapest, Hungary; (B.M.)
| | | | - Alexandra Fábián
- Heart and Vascular Centre, Semmelweis University, 1085 Budapest, Hungary; (B.M.)
| | - Bálint András Fekete
- Heart and Vascular Centre, Semmelweis University, 1085 Budapest, Hungary; (B.M.)
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Tamás Attila György
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Csaba Bödör
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Attila Kovács
- Heart and Vascular Centre, Semmelweis University, 1085 Budapest, Hungary; (B.M.)
| | - Béla Merkely
- Heart and Vascular Centre, Semmelweis University, 1085 Budapest, Hungary; (B.M.)
| | - Andrea Szűcs
- Heart and Vascular Centre, Semmelweis University, 1085 Budapest, Hungary; (B.M.)
| |
Collapse
|
39
|
Wang J, Li S, Chen Y, Chen J, Wang C, Kang Z, Huang M, Cai Z, Fan Y, Lan Y, Yu Y, Bai R, Chen F, Jiang J, Li W. ScRNA-seq unveils the functional characteristics of glioma-associated macrophages and the regulatory effects of chlorogenic acid on the immune microenvironment-a study based on mouse models and clinical practice. Front Immunol 2025; 15:1494806. [PMID: 39867913 PMCID: PMC11757274 DOI: 10.3389/fimmu.2024.1494806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Glioma is the most common primary malignant brain tumor. Despite advances in surgical techniques and treatment regimens, the therapeutic effects of glioma remain unsatisfactory. Immunotherapy has brought new hope to glioma patients, but its therapeutic outcomes are limited by the immunosuppressive nature of the tumor microenvironment (TME). This study aimed to reveal the subpopulations and functional characteristics of tumor-associated macrophages (TAMs) and explore the regulatory effects of chlorogenic acid (CHA) on the immune microenvironment, as well as its potential for clinical application. Methods In this study, CHA was used in model mice. ScRNA - seq analysis was conducted to elucidate the differentiation trajectories and functional characteristics of bone marrow - derived monomacrophages (BMDMs) and microglia. A PPI and molecular docking model were constructed using the target prediction database. A case of a patient treated with CHA was reviewed. Results CHA slowed tumor growth in model mice and extended the survival time of mice. It enhanced the antigen - presenting function of macrophages and T - cell immune activation - related gene expression, activated microglia through the JAK - STAT pathway, and improved the antitumor functions. The good affinity of CHA with STAT1 was confirmed. The patient treated with CHA survived for 5 years and 6 months, achieved partial remission (PR) after 9 months of treatment, and remained alive without any new symptoms or toxic side effects. Our study revealed the subtypes and differentiation trajectories of TAMs. CHA significantly improved the immune microenvironment of glioma by modulating the function of BMDMs and microglia. Discussion This study may provide new insights into targeting the regulation of TME and offer theoretical and practical support for the clinical application of CHA. The results demonstrated the potential of CHA in improving the immune microenvironment and antitumor effects, which could have implications for future glioma treatment strategies.
Collapse
Affiliation(s)
- Jiachen Wang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shenglan Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuxiao Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinyi Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Capital Medical University, Beijing, China
| | - Can Wang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuang Kang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mengqian Huang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zehao Cai
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuxiang Fan
- Department of Neurosurgery, Capital Medical University Xuanwu Hospital, Beijing, China
| | - Yanjie Lan
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yumeng Yu
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruijing Bai
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Feng Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiandong Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenbin Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Yalcouyé A, Schrauwen I, Traoré O, Bamba S, Aboagye ET, Acharya A, Bharadwaj T, Latanich R, Esoh K, Fortes-Lima CA, de Kock C, Jonas M, Maiga ADB, Cissé CAK, Sangaré MA, Guinto CO, Landouré G, Leal SM, Wonkam A. Whole-exome sequencing reveals known and candidate genes for hearing impairment in Mali. HGG ADVANCES 2025; 6:100391. [PMID: 39663698 PMCID: PMC11730241 DOI: 10.1016/j.xhgg.2024.100391] [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: 08/09/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
Hearing impairment (HI) is the most common neurosensory disorder globally and is reported to be more prevalent in low-income countries. In high-income countries, up to 50% of congenital childhood HI is of genetic origin. However, there are limited genetic data on HI from sub-Saharan African populations. In this study, we investigated the genetic causes of HI in the Malian populations, using whole-exome sequencing. Furthermore, cDNA was transfected into HEK293T cells for localization and expression analysis in a candidate gene. Twenty-four multiplex families were enrolled, 50% (12/24) of which are consanguineous. Clustering methods showed patterns of admixture from non-African sources in some Malian populations. Variants were found in six known nonsyndromic HI (NSHI) genes, four genes that can underlie either syndromic HI (SHI) or NSHI, one SHI gene, and one novel candidate HI gene. Overall, 75% of families (18/24) were solved, and 94.4% (17/18) had variants in known HI genes including MYO15A, CDH23, MYO7A, GJB2, SLC26A4, PJVK, OTOGL, TMC1, CIB2, GAS2, PDCH15, and EYA1. A digenic inheritance (CDH23 and PDCH15) was found in one family. Most variants (59.1%, 13/22) in known HI genes were not previously reported or associated with HI. The UBFD1 candidate HI gene, which was identified in one consanguineous family, is expressed in human inner ear organoids. Cell-based experiments in HEK293T showed that mutants UBFD1 had a lower expression, compared to wild type. We report the profile of known genes and the UBFD1 candidate gene for HI in Mali and emphasize the potential of gene discovery in African populations.
Collapse
Affiliation(s)
- Abdoulaye Yalcouyé
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali; Division of Human Genetics, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; McKusick-Nathans Institute, and Department of Genetic Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Isabelle Schrauwen
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Oumou Traoré
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali
| | - Salia Bamba
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali
| | - Elvis Twumasi Aboagye
- Division of Human Genetics, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Anushree Acharya
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Thashi Bharadwaj
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Rachel Latanich
- McKusick-Nathans Institute, and Department of Genetic Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Kevin Esoh
- Division of Human Genetics, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; McKusick-Nathans Institute, and Department of Genetic Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Cesar A Fortes-Lima
- McKusick-Nathans Institute, and Department of Genetic Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Carmen de Kock
- Division of Human Genetics, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mario Jonas
- Division of Human Genetics, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Cheick A K Cissé
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali
| | - Moussa A Sangaré
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali
| | - Cheick O Guinto
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali; Service de Neurologie, Centre Hospitalier Universitaire du Point G, Bamako, Mali
| | - Guida Landouré
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali; Service de Neurologie, Centre Hospitalier Universitaire du Point G, Bamako, Mali
| | - Suzanne M Leal
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Ambroise Wonkam
- Division of Human Genetics, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; McKusick-Nathans Institute, and Department of Genetic Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
41
|
Xu L, Zhou S, Li J, Yu W, Gao W, Luo H, Fang X. The Anti-Inflammatory Effects of Formononetin, an Active Constituent of Pueraria montana Var. Lobata, via Modulation of Macrophage Autophagy and Polarization. Molecules 2025; 30:196. [PMID: 39795251 PMCID: PMC11721999 DOI: 10.3390/molecules30010196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Pueraria montana var. lobata (Willd.) Maesen & S.M.Almeida ex Sanjappa & Predeep (P. lobata) is a medicinal herb widely used in the food and pharmaceutical industries, and studies have shown that P. lobata possesses significant anti-inflammatory pharmacological activities. In this paper, a total of 16 compounds were isolated and identified from P. lobata, among which compounds 1-3, 7, 14, and 16 were isolated from P. lobata for the first time. The results of an in vitro anti-inflammatory activity screening assay showed that compounds 1, 4, 6, 8, and 15 were able to significantly reduce the levels of pro-inflammatory cytokines IL-6 and IL-1β in LPS-induced RAW264.7 macrophages, with the most obvious effect produced by compound 6 (formononetin), while formononetin was able to significantly reduce the number of macrophages at the site of inflammation in transgenic zebrafish. In addition, network pharmacological analysis revealed that the anti-inflammatory activity of formononetin is closely related to autophagy and polarization targets such as TNF, EGFR, PTGS2, and ESR1. In vitro validation experiments showed that formononetin could enhance the expression of LCII/LCI and reduce the expression of P62 protein, reduce the expression of CD86, and enhance the expression of CD206, which further indicated that formononetin could reduce inflammation by regulating macrophage autophagy and polarization processes.
Collapse
Affiliation(s)
- Linyi Xu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (L.X.); (S.Z.); (J.L.); (W.Y.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Shuo Zhou
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (L.X.); (S.Z.); (J.L.); (W.Y.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jing Li
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (L.X.); (S.Z.); (J.L.); (W.Y.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Wenbo Yu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (L.X.); (S.Z.); (J.L.); (W.Y.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Wenyi Gao
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (L.X.); (S.Z.); (J.L.); (W.Y.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Haoming Luo
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (L.X.); (S.Z.); (J.L.); (W.Y.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xiaoxue Fang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (L.X.); (S.Z.); (J.L.); (W.Y.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
42
|
Jiang B, Wang Y, Zhi X, Liu A, Wang L, Wang X, Wang Z, Duan Y, Li Y, Zhang Z. Elucidating the mechanism of action of astragalus polysaccharide on ionizing radiation-induced myocardial damage based on network pharmacology and experimental research. Int Immunopharmacol 2025; 145:113758. [PMID: 39657540 DOI: 10.1016/j.intimp.2024.113758] [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: 09/20/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
Due to the unavoidable impact of ionizing radiation on the heart located near the mediastinum, varying degrees of myocardial damage may occur. As a result, the clinical application of radiotherapy in cancer treatment is significantly limited. However, the molecular mechanisms underlying radiation-induced heart disease (RIHD) are not yet fully understood, and there is a lack of disease-specific treatment strategies. Astragalus polysaccharide (APS), is an active compound abundant in the traditional Chinese herb Astragalus membranaceus (Fisch.) Bunge (AS), has been shown to have cardioprotective effects against various cardiovascular diseases. Thus, this study aims to investigate the potential cardioprotective effect of APS on RIHD and its underlying molecular mechanisms. The network pharmacology results indicated that 9 core genes were identified from the biological network of the effective components of AS acting on RIHD. The results of GO enrichment analysis showed that these hub genes were mainly involved in biological processes such as cell apoptosis, cell proliferation, inflammatory response, and response to external stimuli. The results of KEGG enrichment analysis showed that these hub genes mainly regulated the occurrence of RIHD through pathways such as the EGFR signaling pathway, PI3K/Akt signaling pathway, IL-17 signaling pathway, and so on. In molecular docking analysis, we found that AKT1 and mTOR had good and stable binding abilities with the three types of glucosides rich in AS. The results of in vitro and in vivo experiments all showed that APS could not only improve cardiac dysfunction, myocardial injury, inflammatory response, and myocardial fibrosis in RIHD rats, but also alleviated apoptosis and atrophy of H9C2 cells under ionizing radiation stimulation. In addition, we also found that APS improved the accumulation of autophagic flux induced by ionizing radiation, which could be confirmed by the reversal of Beclin1, p62, LC3B proteins and accelerated degradation of accumulated autophagic vesicles. Rapamycin (Rap) was a classic autophagy flux inducer that could attenuate the improvement effect of APS on H9C2 cell apoptosis under ionizing radiation stimulation. Finally, we found that APS could reverse the inhibition of PI3K/Akt/mTOR signaling pathway activity by ionizing radiation in vitro, thereby improving ionizing radiation-induced autophagy flux accumulation, cardiomyocyte apoptosis, and atrophy. All in all, this study provides important evidence for understanding the molecular mechanisms of the cross-talk between autophagy and apoptosis, and provides new directions and insights for APS combined with autophagy regulators as a therapeutic strategy for RIHD.
Collapse
Affiliation(s)
- Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yan Wang
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Xiaodong Zhi
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou, Gansu 730000, China
| | - Ai Liu
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Lingyun Wang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Xuehan Wang
- Department of First Clinical Medicine, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zheng Wang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Ying Duan
- Department of Ultrasound, Gansu Provincial Cancer Hospital, Lanzhou, Gansu 730050, China
| | - Yingdong Li
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou, Gansu 730000, China
| | - Zheng Zhang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Center for Heart, Lanzhou University of the First Hospital, Lanzhou, Gansu 730030, China.
| |
Collapse
|
43
|
Ding W, Huang C, Chen J, Zhang W, Wang M, Ji X, Nie S, Sun Z. Exploring the Molecular Mechanism by which Kaempferol Attenuates Sepsis-related Acute Respiratory Distress Syndrome Based on Network Pharmacology and Experimental Verification. Curr Comput Aided Drug Des 2025; 21:166-178. [PMID: 38321908 DOI: 10.2174/0115734099295805240126043059] [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: 12/11/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Sepsis-related acute respiratory distress syndrome (ARDS) is a fatal disease without effective therapy. Kaempferol is a flavonoid compound extracted from natural plant products; it exerts numerous pharmacological effects. Kaempferol attenuates sepsis-related ARDS; however, the underlying protective mechanism has not been elucidated completely. OBJECTIVES This study aimed to use network pharmacology and experimental verification to investigate the mechanisms by which kaempferol attenuates sepsis-related ARDS. METHODS We screened the targets of kaempferol by PharMapper, Swiss Target Prediction, and CTD database. We identified the targets of sepsis-related ARDS by GeneCards, DisGeNet, OMIM, and TTD. The Weishengxin platform was used to map the targets of both kaempferol and sepsis-related ARDS. We created a Venn diagram to identify the intersection targets. We constructed the "component-intersection targets-disease" network diagram using Cytoscape 3.9.1 software. The intersection targets were imported into the STRING database for developing the protein-protein interaction network. Metascape was used for the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. We selected the leading 20 KEGG pathways to establish the KEGG relationship network. Finally, we performed experimental verification to confirm our prediction results. RESULTS Through database screening, we obtained 502, 360, and 78 kaempferol targets, disease targets of sepsis-related ARDS, and intersection targets, respectively. The core targets consisted of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, albumin (ALB), IL-1β, and AKT serine/ threonine kinase (AKT)1. GO enrichment analysis identified 426 items, which were principally involved in response to lipopolysaccharide, regulation of inflammatory response, inflammatory response, positive regulation of cell migration, positive regulation of cell adhesion, positive regulation of protein phosphorylation, response to hormone, regulation of reactive oxygen species (ROS) metabolic process, negative regulation of apoptotic signaling pathway, and response to decreased oxygen levels. KEGG enrichment analysis identified 151 pathways. After eliminating the disease and generalized pathways, we obtained the hypoxia-inducible factor 1 (HIF-1), nuclear factor κB (NF-κB), and phosphoinositide 3-kinase (PI3K)-Akt signaling pathways. Our experimental verification confirmed that kaempferol blocked the HIF-1, NFκκB, and PI3K-Akt signaling pathways, diminished TNF-α, IL-1β, and IL-6 expressions, suppressed ROS production, and inhibited apoptosis in lipopolysaccharide (LPS)-induced murine alveolar macrophage (MH-S) cells. CONCLUSION Kaempferol can reduce inflammatory response, ROS production, and cell apoptosis by acting on the HIF-1, NF-κB, and PI3K-Akt signaling pathways, thereby alleviating sepsis- related ARDS.
Collapse
Affiliation(s)
- Weichao Ding
- Department of Emergency Medicine, Jinling Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210000, China
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Changbao Huang
- Department of Emergency Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, China
| | - Juan Chen
- Department of Emergency Medicine, Jinling Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210000, China
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
- Department of Emergency Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221000, China
| | - Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Mengmeng Wang
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Xiaohang Ji
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Shinan Nie
- Department of Emergency Medicine, Jinling Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210000, China
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Zhaorui Sun
- Department of Emergency Medicine, Jinling Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210000, China
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| |
Collapse
|
44
|
Chen J, Zhang X. An Integrated Approach of Network Pharmacology, Bioinformatics, Molecular Docking, and Experimental Verification Uncovers Prunellae Spica as the Potential Medicine of Prognosis Improvement for Oral Squamous Cell Carcinoma. Curr Pharm Des 2025; 31:391-412. [PMID: 39289945 DOI: 10.2174/0113816128328547240827045955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Prunellae Spica (PS), the spike from Prunella vulgaris L., is a traditional Chinese medicine that can treat Oral Squamous Cell Carcinoma (OSCC), whereas its molecular mechanisms and effects on the prognosis of patients remain unclear. METHODS Our study aimed to identify the potential anti-OSCC targets of PS and explore its mechanisms and effects on prognosis through network pharmacology, bioinformatics analysis, molecular docking, and in-vitro cell assays. RESULTS Sixty-two potential targets of 11 active anti-OSCC ingredients of PS were identified, with Quercetin, the core ingredient of PS, exhibiting the most significant number of OSCC-related targets. GO analysis indicated that the primary biological processes involved in OSCC treatment by PS were the cellular response to nitrogen compound, response to xenobiotic stimulus, and cellular response to organonitrogen compound. KEGG analysis revealed that pathways in cancer were the top highly enriched signaling pathway in the treatment of OSCC by PS. DisGeNET analysis is mainly about Lip and Oral Cavity Carcinoma. More importantly, 6 of the 62 targets were markedly related to prognosis. Molecular docking revealed high affinities between the key component and the prognosis-related target proteins. Treatment of OSCC cell line SCC-25 with Quercetin could inhibit malignant biological behaviors, such as cell proliferation, colony formation, invasion, and migration, as well as affect the targets related to prognosis and promote autophagy. CONCLUSION Overall, these results suggest that PS plays a significant role in treating and improving the prognosis of OSCC by directly influencing various processes in OSCC.
Collapse
MESH Headings
- Humans
- Molecular Docking Simulation
- Network Pharmacology
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/pathology
- Mouth Neoplasms/diagnosis
- Mouth Neoplasms/metabolism
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/diagnosis
- Carcinoma, Squamous Cell/metabolism
- Computational Biology
- Prunella/chemistry
- Cell Proliferation/drug effects
- Prognosis
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/isolation & purification
- Drug Screening Assays, Antitumor
Collapse
Affiliation(s)
- Jiahui Chen
- Beijing Stomatological Hospital and School of Stomatology, Beijing Institute of Dental Research, Capital Medical University, No. 4 Tiantanxili, Dongcheng District, Beijing 100050, China
| | - Xinyan Zhang
- Beijing Stomatological Hospital and School of Stomatology, Beijing Institute of Dental Research, Capital Medical University, No. 4 Tiantanxili, Dongcheng District, Beijing 100050, China
| |
Collapse
|
45
|
Xiao C, Wang Y, Liu J, Li X, Wang P, Zhou J, Xiu H, Lu S, Zhu H, Wang R. Mechanism of Fangji Huangqi decoction against acute kidney injury based on network pharmacology and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156345. [PMID: 39742571 DOI: 10.1016/j.phymed.2024.156345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/21/2024] [Accepted: 10/07/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND Fangji Huangqi Decoction (FJHQD), a famous Traditional Chinese Medicine (TCM) formula, has been widely applied in improving renal function. However, the interaction of bioactives from FJHQD with the targets involved in acute renal injury (AKI) has not been elucidated yet. PURPOSE A network pharmacology-based approach combined with molecular docking and in vitro and in vivo validation was performed to determine the bioactives, key targets, and potential pharmacological mechanism of FJHQD against AKI. MATERIALS AND METHODS The model of mouse renal ischemic reperfusion was adopted to verify the curative effect of FJHQD against renal injury. FJHQD was analyzed and separated by Ultra-High performance liquid chromatography (UHPLC). Bioactives and potential targets of FJHQD, as well as AKI-related targets, were retrieved from public databases. Crucial bioactive ingredients, potential targets, and signaling pathways were acquired through bioinformatics analysis, including protein-protein interaction (PPI), as well as the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Subsequently, molecular docking was carried out to predict the combination of active compounds with core targets. Besides, in vivo and vitro experiments were conducted to verify the findings. RESULTS A total of 20 bioactive ingredients of FJHQD (top 10 positive ion and negative ion compounds) and 274 FJHQD-AKI overlaped targets were screened. Bioinformatics analysis revealed that apoptosis mediated by PI3K-AKT signaling pathway might play an important role in FJHQD against AKI. Further experiments showed that FJHQD alleviated I/R-induced renal injury and OGD/R induced TEC apoptosis by activating PI3K-AKT signaling pathway. Moreover, molecular docking suggested (9Z,12Z,14E)-16-Hydroxy-9,12,14-octadecatrienoic acid, 2-Hydroxyacetophenone, Liquiritigenin, (S)-[10]-Gingerol and Isookanin-7-O-glucoside may be potential candidate agents, among which, PIK3CA interacted with Liquiritigenin, (S)-[10]-Gingerol, Isookanin-7-O-glucoside and 2-Hydroxyacetophenone respectively. AKT1 interacted with (9Z,12Z,14E)-16-Hydroxy-9,12,14-octadecatrienoic acid and 2-Hydroxyacetophenone. Cell experiments showed that the most important ingredient of FJHQD, Liquiritigenin, could inhibit the TEC apoptosis and up-regulate PI3K-Akt signaling pathway, which further confirmed the prediction by network pharmacology strategy and molecular docking. CONCLUSION Our results comprehensively illustrated the bioactives, potential targets, and molecular mechanism of FJHQD against AKI. It also provided a promising strategy to uncover the scientific basis and therapeutic mechanism of TCM formulae in treating diseases.
Collapse
Affiliation(s)
- Chengcheng Xiao
- Department of Urology, Qingdao Municipal Hospital, Qingdao, PR China
| | - Yayun Wang
- Department of Hematology, Qingdao Municipal Hospital, Qingdao, PR China
| | - Jingwei Liu
- Department of Urology, Qingdao Chengyang People's Hospital, Qingdao, PR China
| | - Xin Li
- Department of Anorectal, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Peng Wang
- Department of Urology, Qingdao Municipal Hospital, Qingdao, PR China
| | - Junran Zhou
- Department of Thoracic Surgery, Qingdao Municipal Hospital, Qingdao, PR China
| | - Hao Xiu
- Department of Traditional Chinese Medicine, Qingdao Municipal Hospital, Qingdao, PR China
| | - Shun Lu
- Department of Traditional Chinese Medicine, Qingdao Municipal Hospital, Qingdao, PR China
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital, Qingdao, PR China
| | - Renhe Wang
- Department of Traditional Chinese Medicine, Qingdao Municipal Hospital, Qingdao, PR China.
| |
Collapse
|
46
|
Li B, Liu J, He C, Deng Z, Zhou X, Peng R. Unveiling the Therapeutic Potential of Berberine in Rheumatoid Arthritis: A Comprehensive Study of Network Pharmacology, Metabolomics, and Intestinal Flora. J Inflamm Res 2024; 17:10849-10869. [PMID: 39677295 PMCID: PMC11645930 DOI: 10.2147/jir.s493892] [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: 08/30/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease influenced by environmental triggers, including the commensal microbiota. Recent research has highlighted distinctive features of the gut microbiota in RA patients. This study investigates the therapeutic potential of berberine (BBR), a gut microbiota modulator known for its significant anti-RA effects, and elucidates the underlying mechanisms. Methods Utilizing the collagen-induced arthritis (CIA) rat model, we comprehensively evaluated the anti-rheumatoid arthritis effects of BBR in vivo through various indices, such as paw edema, arthritis index, ankle diameter, inflammatory cytokine levels, pathological conditions, and micro-CT analysis. Employing network pharmacology, we identified potential targets involved in RA alleviation by BBR. To analyze comprehensive metabolic profiles and identify underlying metabolic pathways, we conducted a serum-based widely targeted metabolomics analysis utilizing LC-MS technology. An integrated network encompassing metabolomics and network pharmacology data was constructed using Cytoscape. The potential therapeutic targets and signaling pathways of BBR in the management of RA were predicted using network pharmacology. Key targets and pathways were further validated by molecular docking and immunofluorescent staining, which integrated findings from serum metabolomics and network pharmacology analysis. Additionally, we analyzed the gut microbiota composition in rats employing 16S rDNA sequencing and investigated the effects of BBR on the microbiota of CIA rats through bioinformatics and statistical methods. Results Our results showed that BBR demonstrated significant efficacy in alleviating RA symptoms in CIA rats, as evidenced by improvements in paw redness and swelling, attenuation of bone and cartilage damage, reduction in synovial hyperplasia, inflammatory cell infiltration, and suppression of proinflammatory cytokines IL-1β, IL-6, IL-17A, and TNF-α. KEGG analysis highlighted the PI3K/AKT signaling pathway as a key mediator of BBR's anti-RA effects. Metabolomics profiling via LC-MS revealed 22 potential biomarkers. Arginine and proline metabolism, cutin, suberine and wax biosynthesis, glycine, serine and threonine metabolism and taurine and hypotaurine metabolism are the most related pathways of BBR anti-RA. Molecular docking studies corroborated high affinities between BBR and key targets. Furthermore, 16S analysis demonstrated BBR's capacity to modulate gut bacteria composition, including an increase in the abundance of Lachnoclostridium, Akkermansia, Blautia, Romboutsia, and Faecalibacterium genera, alongside a decrease in Prevotella_9 abundance in genus level. Integrated analysis underscored a strong correlation between serum microbiota and fecal metabolites. Conclusion Our findings elucidate the multifaceted mechanisms underlying BBR's therapeutic efficacy in RA, involving inhibition of the PI3K/AKT pathway, modulation of intestinal flora, and regulation of host metabolites. These insights provide novel perspectives on BBR's role in RA management.
Collapse
Affiliation(s)
- Bocun Li
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Jing Liu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Chuan He
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Zhou Deng
- Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of Acupuncture, Wuhan, Hubei, People’s Republic of China
| | - Xiaohong Zhou
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Rui Peng
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| |
Collapse
|
47
|
Sillence DO. A Dyadic Nosology for Osteogenesis Imperfecta and Bone Fragility Syndromes 2024. Calcif Tissue Int 2024; 115:873-890. [PMID: 38942908 PMCID: PMC11607092 DOI: 10.1007/s00223-024-01248-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024]
Abstract
In 2023 following extensive consultation with key stakeholders, the expert Nosology Working Group of the International Skeletal Dysplasia Society (ISDS) published the new Dyadic Nosology for Genetic Disorders of the Skeleton. Some 770 entities were delineated associated with 552 genes. From these entities, over 40 genes resulting in distinct forms of Osteogenesis Imperfecta (OI) and Bone Fragility and/or Familial Osteoporosis were identified. To assist clinicians and lay stake holders and bring the considerable body of knowledge of the matrix biology and genomics to people with OI as well as to clinicians and scientists, a dyadic nosology has been recommended. This combines a genomic co-descriptor with a phenotypic naming based on the widely used Sillence nosology for the OI syndromes and the many other syndromes characterized in part by bone fragility.This review recapitulates and explains the evolution from the simple Congenita and Tarda subclassification of OI in the 1970 nosology, which was replaced by the Sillence types I-IV nosology which was again replaced in 2009 with 5 clinical groups, type 1 to 5. Qualitative and quantitative defects in type I collagen polypeptides were postulated to account for the genetic heterogeneity in OI for nearly 30 years, when OI type 5, a non-collagen disorder was recognized. Advances in matrix biology and genomics since that time have confirmed a surprising complexity both in transcriptional as well as post-translational mechanisms of collagens as well as in the many mechanisms of calcified tissue homeostasis and integrity.
Collapse
Affiliation(s)
- David Owen Sillence
- Specialities of Genomic Medicine and Paediatrics and Adolescent Health, Children's Hospital Westmead, Sydney University Clinical School, Westmead, NSW, 2145, Australia.
- Department of Genetic Medicine, Honorary Emeritus Consultant, Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
48
|
Jiang L, Yang D, Zhang Z, Xu L, Jiang Q, Tong Y, Zheng L. Elucidating the role of Rhodiola rosea L. in sepsis-induced acute lung injury via network pharmacology: emphasis on inflammatory response, oxidative stress, and the PI3K-AKT pathway. PHARMACEUTICAL BIOLOGY 2024; 62:272-284. [PMID: 38445620 PMCID: PMC10919309 DOI: 10.1080/13880209.2024.2319117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/07/2024] [Indexed: 03/07/2024]
Abstract
CONTEXT Sepsis-induced acute lung injury (ALI) is associated with high morbidity and mortality. Rhodiola rosea L. (Crassulaceae) (RR) and its extracts have shown anti-inflammatory, antioxidant, immunomodulatory, and lung-protective effects. OBJECTIVE This study elucidates the molecular mechanisms of RR against sepsis-induced ALI. MATERIALS AND METHODS The pivotal targets of RR against sepsis-induced ALI and underlying mechanisms were revealed by network pharmacology and molecular docking. Human umbilical vein endothelial cells (HUVECs) were stimulated by 1 μg/mL lipopolysaccharide for 0.5 h and treated with 6.3, 12.5, 25, 50, 100, and 200 μg/mL RR for 24 h. Then, the lipopolysaccharide-stimulated HUVECs were subjected to cell counting kit-8 (CCK-8), enzyme-linked immunosorbent, apoptosis, and Western blot analyses. C57BL/6 mice were divided into sham, model, low-dose (40 mg/kg), mid-dose (80 mg/kg), and high-dose (160 mg/kg) RR groups. The mouse model was constructed through caecal ligation and puncture, and histological, apoptosis, and Western blot analyses were performed for further validation. RESULTS We identified six hub targets (MPO, HRAS, PPARG, FGF2, JUN, and IL6), and the PI3K-AKT pathway was the core pathway. CCK-8 assays showed that RR promoted the viability of the lipopolysaccharide-stimulated HUVECs [median effective dose (ED50) = 18.98 μg/mL]. Furthermore, RR inhibited inflammation, oxidative stress, cell apoptosis, and PI3K-AKT activation in lipopolysaccharide-stimulated HUVECs and ALI mice, which was consistent with the network pharmacology results. DISCUSSION AND CONCLUSION This study provides foundational knowledge of the effective components, potential targets, and molecular mechanisms of RR against ALI, which could be critical for developing targeted therapeutic strategies for sepsis-induced ALI.
Collapse
Affiliation(s)
- Lu Jiang
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Dongdong Yang
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Zhuoyi Zhang
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Liying Xu
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Qingyu Jiang
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yixin Tong
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Lanzhi Zheng
- Department of Medical Administration, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
49
|
Manaithiya A, Bhowmik R, Acharjee S, Sharma S, Kumar S, Imran M, Mathew B, Parkkila S, Aspatwar A. Elucidating molecular mechanism and chemical space of chalcones through biological networks and machine learning approaches. Comput Struct Biotechnol J 2024; 23:2811-2836. [PMID: 39045026 PMCID: PMC11263914 DOI: 10.1016/j.csbj.2024.07.006] [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: 05/09/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/25/2024] Open
Abstract
We developed a bio-cheminformatics method, exploring disease inhibition mechanisms using machine learning-enhanced quantitative structure-activity relationship (ML-QSAR) models and knowledge-driven neural networks. ML-QSAR models were developed using molecular fingerprint descriptors and the Random Forest algorithm to explore the chemical spaces of Chalcones inhibitors against diverse disease properties, including antifungal, anti-inflammatory, anticancer, antimicrobial, and antiviral effects. We generated and validated robust machine learning-based bioactivity prediction models (https://github.com/RatulChemoinformatics/QSAR) for the top genes. These models underwent ROC and applicability domain analysis, followed by molecular docking studies to elucidate the molecular mechanisms of the molecules. Through comprehensive neural network analysis, crucial genes such as AKT1, HSP90AA1, SRC, and STAT3 were identified. The PubChem fingerprint-based model revealed key descriptors: PubchemFP521 for AKT1, PubchemFP180 for SRC, PubchemFP633 for HSP90AA1, and PubchemFP145 and PubchemFP338 for STAT3, consistently contributing to bioactivity across targets. Notably, chalcone derivatives demonstrated significant bioactivity against target genes, with compound RA1 displaying a predictive pIC50 value of 5.76 against HSP90AA1 and strong binding affinities across other targets. Compounds RA5 to RA7 also exhibited high binding affinity scores comparable to or exceeding existing drugs. These findings emphasize the importance of knowledge-based neural network-based research for developing effective drugs against diverse disease properties. These interactions warrant further in vitro and in vivo investigations to elucidate their potential in rational drug design. The presented models provide valuable insights for inhibitor design and hold promise for drug development. Future research will prioritize investigating these molecules for mycobacterium tuberculosis, enhancing the comprehension of effectiveness in addressing infectious diseases.
Collapse
Affiliation(s)
- Ajay Manaithiya
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ratul Bhowmik
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Satarupa Acharjee
- Department of Pharmacy, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bangalore 560043, India
| | - Sunil Kumar
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS, Health Sciences Campus, Kochi, India
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS, Health Sciences Campus, Kochi, India
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Ltd., Tampere University Hospital, Tampere, Finland
| | - Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
50
|
Wang N, Cui J, Sun Z, Chen F, He X. Exploring the protective effect and molecular mechanism of betulin in Alzheimer's disease based on network pharmacology, molecular docking and experimental validation. Mol Med Rep 2024; 30:232. [PMID: 39392030 PMCID: PMC11529172 DOI: 10.3892/mmr.2024.13356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that impairs learning and memory, with high rates of mortality. Birch bark has been traditionally used in the treatment of various skin ailments. Betulin (BT) is a key compound of birch bark that exhibits diverse pharmacological benefits and therapeutic potential in AD. However, the therapeutic effects and molecular mechanisms of BT in AD remain unclear. The present study aimed to predict the potential therapeutic targets of BT in the treatment of AD, and to determine the specific underlying molecular mechanisms through network pharmacology analysis and experimental validation. PharmMapper was used to predict the target genes of BT, and four disease databases were searched to screen for AD targets. The intersection targets were identified using the jveen website. Drug‑disease target protein‑protein interaction networks and hub genes were obtained and visualized using the Search Tool for the Retrieval of Interacting Genes/Proteins database and Cytoscape. The Database for Annotation, Visualization and Integrated Discovery was used for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and AutoDock was used for molecular docking analysis of BT and hub genes. Subsequently, the network‑predicted mechanisms of BT in AD were verified in vitro. A total of 495 BT and 1,386 AD targets were identified, and 120 were identified as potential targets of BT in the treatment of AD. The results of the molecular docking analysis revealed a strong binding affinity between BT and the hub genes. In addition, enrichment analyses of GO and KEGG pathways indicated that the neuroprotective effects of BT mainly involved the 'PI3K‑Akt signaling pathway'. The results of in vitro experiments demonstrated that pretreatment with BT for 2 h may ameliorate formaldehyde (FA)‑induced cytotoxicity and morphological changes in HT22 cells, and decrease FA‑induced Tau hyperphosphorylation and reactive oxygen species levels. Furthermore, the PI3K/AKT signaling pathway was activated and the expression levels of downstream proteins, namely GSK3β, Bcl‑2 and Bax, were modified following pre‑treatment with BT. Overall, the results of network pharmacology and in vitro analyses revealed that BT may reduce FA‑induced AD‑like pathology by modulating the PI3K/AKT signaling pathway, highlighting it as a potential multi‑target drug for the treatment of AD.
Collapse
Affiliation(s)
- Na Wang
- Laboratory of Brain and Cognitive Science, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671003, P.R. China
| | - Jiali Cui
- Yunnan Institute of Materia Medica, Yunnan Province Company Key Laboratory for TCM and Ethnic Drug of New Drug Creation, Kunming, Yunnan 650111, P.R. China
| | - Ziteng Sun
- Laboratory of Brain and Cognitive Science, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671003, P.R. China
| | - Fan Chen
- Department of Psychiatry, The Affiliated Mental Health Center of Jiangnan University, Wuxi, Jiangsu 214151, P.R. China
- Laboratory of Heart Disease Mechanism and Translational Research, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Xiaping He
- Laboratory of Brain and Cognitive Science, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671003, P.R. China
| |
Collapse
|