1
|
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
|
2
|
Wei S, Li X, Li X, Wang R, Wang Y, Li Y. An integrated approach using molecular docking, network pharmacology, and UPLC-Q-TOF-MS analysis to investigate the chemical makeup and mechanism of Xiaoqinglong decoction against asthma. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1254:124490. [PMID: 39923611 DOI: 10.1016/j.jchromb.2025.124490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVE This study aims to investigate the potential mechanisms by which Xiaoqinglong decoction (XQLD) exerts its therapeutic effects on asthma. This will be achieved through the application of the UPLC-Q-TOF-MS coupling technique, integrated with network pharmacology and molecular docking methodologies. METHODS The UPLC-Q-TOF-MS technique was employed to perform a qualitative analysis of both the aqueous extract of XQLD and the drug-containing serum. The Swiss TargetPrediction, OMIM, and GeneCards databases were utilized to identify blood-derived components and disease-associated targets. Subsequently, a protein-protein interaction (PPI) network was constructed by intersecting these datasets to identify key targets, which were then subjected to Gene Ontology (GO) functional analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Cytoscape software facilitated the construction of a 'drug-component-disease-target' network to enable visualization and analysis, thereby aiding in the prediction of targets and signaling pathways of XQLD in the treatment of asthma. Finally, molecular docking of the pertinent incoming components to the central target was conducted utilizing AutoDock Vina and PyMol software. RESULTS A comprehensive analysis identified 102 components within the aqueous extract of XQLD, alongside 93 components in the drug-containing serum. Additionally, 90 compound-disease shared targets and 45 key targets were identified through PPI network analysis. Notably, compounds such as apigenin, l-asarinin, 6-shogaol, ellagic acid, kaempferol, and naringenin are pivotal in mediating the therapeutic effects of XQLD in asthma treatment. The primary molecular targets of XQLD for asthma include SRC, AKT1, EGFR, ESR1, HIF1A, and PIK3CA. The results of the molecular docking analysis indicated that the binding energies between the core target and the active ingredient were ≤ -5.5 kcal/mol, demonstrating a strong affinity. CONCLUSION This study elucidated the chemical composition, potential targets, and action pathways of the aqueous extract of XQLD and its drug-containing serum. It preliminarily identified the material basis and mechanism of action, thereby providing a foundation for further in-depth research into the mechanisms underlying XQLD and its clinical applications.
Collapse
Affiliation(s)
- Shuang Wei
- College of traditional Chinese medicine, Tianjin University of traditional Chinese medicine, Tianjin 301617, China
| | - Xueting Li
- College of traditional Chinese medicine, Tianjin University of traditional Chinese medicine, Tianjin 301617, China
| | - Xinyu Li
- College of traditional Chinese medicine, Tianjin University of traditional Chinese medicine, Tianjin 301617, China
| | - Rui Wang
- College of traditional Chinese medicine, Tianjin University of traditional Chinese medicine, Tianjin 301617, China
| | - Yuming Wang
- College of traditional Chinese medicine, Tianjin University of traditional Chinese medicine, Tianjin 301617, China.
| | - Yubo Li
- College of traditional Chinese medicine, Tianjin University of traditional Chinese medicine, Tianjin 301617, China.
| |
Collapse
|
3
|
Zhu Z, Zhong Y, He R, Zhong C, Chen J, Peng H. Mechanistic Insights into Salvigenin for Glucocorticoid-Induced Femoral Head Osteonecrosis: A Network Pharmacology and Experimental Study. Biomedicines 2025; 13:614. [PMID: 40149590 PMCID: PMC11940827 DOI: 10.3390/biomedicines13030614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Glucocorticoid-induced osteonecrosis of the femoral head (GIOFH) is a debilitating condition resulting from impaired bone metabolism and vascular disruption due to prolonged glucocorticoid use. This study aimed to explore the therapeutic potential of salvigenin, a flavonoid with antioxidative and estrogen-like properties, in alleviating GIOFH by modulating estrogen receptor alpha (ESR1) pathways. Methods: A network pharmacology approach was utilized to identify salvigenin's potential targets and their association with GIOFH. Protein-protein interaction networks, along with Gene Ontology and KEGG pathway analyses, were conducted to clarify salvigenin's multi-target mechanisms. Molecular docking and dynamics simulations assessed the interaction between salvigenin and ESR1. Experimental validation included in vitro assays on MG63 cells treated with dexamethasone (Dex) to mimic GIOFH, evaluating oxidative stress, apoptosis, osteogenic differentiation, and ESR1 expression. Results: Network analysis identified ESR1, NOS3, and MMP9 as key hub targets of salvigenin. Molecular docking and dynamics simulations confirmed stable binding of salvigenin to ESR1. Salvigenin significantly reduced Dex-induced oxidative stress and apoptosis in osteoblasts while restoring osteogenic differentiation and ESR1 expression. Functional assays showed improved mineralized nodule formation, ALP activity, and mitochondrial integrity in salvigenin-treated cells. Conclusions: Salvigenin exhibits significant therapeutic potential in addressing GIOFH through ESR1-mediated pathways. These results offer a strong foundation for future translational studies and the development of salvigenin-based therapies for glucocorticoid-induced bone disorders.
Collapse
Affiliation(s)
- Zhengjie Zhu
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Z.Z.); (Y.Z.); (C.Z.)
| | - Yujian Zhong
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Z.Z.); (Y.Z.); (C.Z.)
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Changheng Zhong
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Z.Z.); (Y.Z.); (C.Z.)
| | - Junwen Chen
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Z.Z.); (Y.Z.); (C.Z.)
| | - Hao Peng
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Z.Z.); (Y.Z.); (C.Z.)
| |
Collapse
|
4
|
Cao S, Lv B, Tai Y, Zuo HX, Xing Y, Surh YJ, Li MY, Ma J, Jin X. Formononetin ameliorates DSS-induced colitis by inhibiting the MAPK/PPAR-γ/NF-κB/ROS signaling pathways. Toxicol Appl Pharmacol 2025; 496:117239. [PMID: 39855309 DOI: 10.1016/j.taap.2025.117239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND AND AIM Formononetin (FMN) is a compound isolated from Astragalus membranaceus, that exhibits a range of pharmacological activities, including antitumor, anti-inflammatory, hypolipidemic, and antioxidant effects. Although preliminary study suggests that FMN have a therapeutic role in Inflammatory Bowel Disease (IBD), its specific mechanism of action requires further investigation. This study aimed to investigate the mechanism by which FMN treats DSS-induced colitis in mice. METHODS RAW264.7 and Bone marrow-derived macrophages (BMDMs) were treated with LPS to establish an inflammatory cell model. Biochemical parameters and morphological characteristics were assessed in the present or absent of FMN. 4 % solution of DSS was administered to C57BL/6 mice to induce IBD, which served as an animal model for investigating the pharmacodynamics of FMN. RESULTS FMN significantly reduced colitis-associated injury, as evidenced by a decrease in the disease activity index (DAI), weight gain, and restoration of colon length. Furthermore, FMN inhibits protein expression of NLRP3 inflammasome, suppressed the nuclear translocation of NF-κB/p65, and prevented mitochondrial damage, this process results in a reduction in the accumulation of reactive oxygen species (ROS). Additionally, FMN inhibited the mitogen-activated protein kinase (MAPK) signaling pathway, upregulated peroxisome proliferator-activated receptor gamma (PPAR-γ) in the nucleus, and decreased the release of inflammatory factors, thereby exerting anti-inflammatory effects. CONCLUSION By inhibiting mitochondrial damage, activating the MAPK/PPAR-γ/ROS signaling pathway, reducing the nuclear translocation of NF-κB, and suppressing the expression of NLRP3 inflammasome-associated proteins, FMN exerts anti-inflammatory effects.
Collapse
Affiliation(s)
- Shen Cao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Baojiang Lv
- Medical Supplies Center of PLA General Hospital, Beijing 100853, China
| | - Yi Tai
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Hong Xiang Zuo
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Yue Xing
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Cancer Research Institute, Seoul National University, Seoul 03080, South Korea.
| | - Ming Yue Li
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| | - Xuejun Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| |
Collapse
|
5
|
Joshi A, Kathuria D, Paul M, Singh N. An overview on the potential application of nanotechnology in enhancing the therapeutic efficacy of phytoestrogens. Food Chem 2025; 464:141779. [PMID: 39481307 DOI: 10.1016/j.foodchem.2024.141779] [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: 05/23/2024] [Revised: 09/20/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Phytoestrogens, derived from plants possesses structural similarity with 17 β-estradiol found in mammals. It is abundantly present in soybean along with red clove, alfalfa as well as other legumes, nuts, vegetables and seeds. It is used as hormone replacement therapy and exhibits both anti-estrogenic and estrogenic properties that linked to therapeutic benefits as well as plays active role in sports nutrition. Despite the potential benefits of phytoestrogens, their low solubility, bioavailability, and stability make it challenging to target them effectively. Recent advancements in nanotechnology have paved in facilitating target delivery. Scaling at nano level offered greater surface area, improved solubility, and bioavailability of phytoestrogens which has ultimately reduced the required medication dosage, and enhanced cost-effectiveness, particularly for expensive bioactive substances where precise dosages are recommended. The present article discussed about the potential application of nanotechnology in enhancing therapeutic benefits of phytoestrogens while minimizing their potential side effects.
Collapse
Affiliation(s)
- Aroma Joshi
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India
| | - Deepika Kathuria
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India
| | - Maman Paul
- Department of Physiotherapy, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Narpinder Singh
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India.
| |
Collapse
|
6
|
Yin L, Huang X, Zhang B, Zhu Q, Zhao H. Narciclasine attenuates sepsis-associated acute kidney injury through the ESR1/S100A11 axis. Funct Integr Genomics 2025; 25:13. [PMID: 39808340 DOI: 10.1007/s10142-024-01513-w] [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: 09/19/2024] [Revised: 11/15/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025]
Abstract
Narciclasine (Ncs) was effective in sepsis management due to its antioxidant properties. The present study dissected the protective effects of Ncs against sepsis-associated acute kidney injury (SA-AKI) and the molecular mechanisms. The SA-AKI mice were developed using cecum ligation and puncture and pretreated with Ncs and adenoviruses. Human renal microvascular endothelial cells (RMECs) were induced with LPS and treated with Ncs. Ncs alleviated proximal tubular dilatation, interstitial widening, and necrosis in renal tissues and reduced the renal injury marker and pro-inflammatory cytokine levels in the serum of SA-AKI mice. Ncs promoted the expression of ZO-1, VE-cadherin, and CD31 and the activities of SOD, GSH-Px, and CAT, and inhibited the levels of pro-inflammatory cytokines, and apoptosis rate in LPS-treated RMECs. Estrogen receptor 1 (ESR1) was a target protein of Ncs, and S100 calcium-binding protein A11 (S100A11) was a target of the transcription factor ESR1. Ncs blocked transcription of S100A11 by inhibiting ESR1. Silencing of S100A11 overturned the deteriorating effects of ESR1 overexpression on SA-AKI progression in vivo and RMEC injury in vitro. These findings suggest that Ncs may ameliorate SA-AKI by repressing the ESR1/S100A11 signaling, providing a novel perspective for research on SA-AKI.
Collapse
Affiliation(s)
- Liping Yin
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
- Department of Emergency and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
| | - Xiaofei Huang
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
- Department of Emergency and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
| | - Beibei Zhang
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
- Department of Emergency and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
| | - Qiyong Zhu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
| | - Hui Zhao
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China.
- Department of Emergency and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China.
| |
Collapse
|
7
|
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
|
8
|
Intharuksa A, Kuljarusnont S, Sasaki Y, Tungmunnithum D. Flavonoids and Other Polyphenols: Bioactive Molecules from Traditional Medicine Recipes/Medicinal Plants and Their Potential for Phytopharmaceutical and Medical Application. Molecules 2024; 29:5760. [PMID: 39683916 DOI: 10.3390/molecules29235760] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Currently, natural bioactive ingredients and/or raw materials are of significant interest to scientists around the world. Flavonoids and other polyphenols are a major group of phytochemicals that have been researched and noted as bioactive molecules. They offer several pharmacological and medical benefits. This current review aims to (1) illustrate their benefits for human health, such as antioxidant, anti-aging, anti-cancer, anti-inflammatory, anti-microbial, cardioprotective, neuroprotective, and UV-protective effects, and also (2) to perform a quality evaluation of traditional medicines for future application. Consequently, keywords were searched on Scopus, Google Scholar, and PubMed so as to search for related publications. Then, those publications were carefully checked in order to find current and non-redundant studies that matched the objective of this review. According to this review, researchers worldwide are very interested in discovering the potential of flavonoids and other polyphenols, used in traditional medicines and taken from medicinal plants, in relation to medical and pharmaceutical applications. Many studies focus on the health benefits of flavonoids and other polyphenols have been tested using in silico, in vitro, and in vivo models. However, few studies have been carried out using clinical trials that have trustworthy subject sizes and are in accordance with clinical practice guidelines. Additionally, interesting research directions and perspectives for future studies are highlighted in this work.
Collapse
Affiliation(s)
- Aekkhaluck Intharuksa
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sompop Kuljarusnont
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Yohei Sasaki
- Division of Pharmaceutical Sciences, Graduate School of Medical Plant Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | - Duangjai Tungmunnithum
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Le Studium Institute for Advanced Studies, 1 Rue Dupanloup, 45000 Orléans, France
| |
Collapse
|
9
|
Liu Y, Zheng Y, Wei C, Cai X. DMRT3-mediated lncRNA OIP5-AS1 promotes the pyroptosis of bronchial epithelial cells by binding with EIF4A3 to enhance YAP mRNA stability. Immunol Res 2024; 72:1365-1383. [PMID: 39287912 DOI: 10.1007/s12026-024-09534-0] [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: 03/07/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
Asthma is featured by persistent airway inflammation. Long noncoding RNAs (lncRNAs) are reported to play critical roles in asthma. However, the function of Opa interacting protein 5-antisense 1 (OIP5-AS1) in pyroptosis during the development of asthma remains unexplored. The blood samples of asthma patients (n = 32) as well as the baseline characteristics of asthma patients or healthy people were collected. An in vivo model of asthma was established using house dust mites (HDM). To mimic asthma in vitro, BEAS-2B cells were treated with HDM. Cell pyroptosis and apoptosis were examined by flow cytometry. The levels of interleukin-1 beta (IL-1β) and interleukin-18 (IL-18) were detected by enzyme-linked immunosorbent assay (ELISA). The binding among messenger RNAs (mRNAs) was assessed by chromatin immunoprecipitation (ChIP), dual luciferase report assay, RNA immunoprecipitation (RIP), co-immunoprecipitation (Co-IP), and RNA pull-down assay, respectively. The cellular localization was observed by fluorescence in situ hybridization (FISH) staining. The level of OIP5-AS1 was upregulated in asthma patients. HDM induced pyroptosis and increased the levels of IL-18, IL-1β, and lactate dehydrogenase (LDH) in BEAS-2B cells, which was obviously reversed by OIP5-AS1 knockdown. Consistently, the expressions of NOD-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), c-caspase 1, and pyroptosis-related gasdermin D-1 (GSDMD-1) in BEAS-2B cells were upregulated by HDM treatment, while these phenomena were partially abolished by silencing of OIP5-AS1. Moreover, HDM promoted the progression of asthma in vivo, which was rescued by the downregulation of OIP5-AS1. OIP5-AS1 silencing decreased HDM-induced cell pyroptosis by inactivation of NLRP3. More importantly, OIP5-AS1 promoted the mRNA stability of yes-associated protein (YAP) via binding with eukaryotic translation initiation factor 4A3 (EIF4A3), and OIP5-AS1 was transcriptionally upregulated by doublesex and mab-3 related transcription factor 3 (DMRT3). DMRT3-mediated OIP5-AS1 aggravated the progression of asthma by mediation of the EIF4A3/YAP axis, which might provide a new therapeutic strategy against asthma.
Collapse
Affiliation(s)
- Yunchan Liu
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19 Xiuhua Road, Xiuying District, Haikou, 570311, Hainan Province, P.R. China
| | - Yamei Zheng
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19 Xiuhua Road, Xiuying District, Haikou, 570311, Hainan Province, P.R. China
| | - Chaochao Wei
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19 Xiuhua Road, Xiuying District, Haikou, 570311, Hainan Province, P.R. China
| | - Xingjun Cai
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19 Xiuhua Road, Xiuying District, Haikou, 570311, Hainan Province, P.R. China.
| |
Collapse
|
10
|
Zhao S, Huang S, Wu Y, Yao X, Cai X. GATA1-activated lncRNA OIP5-AS1 and GAS5 promote pyroptosis to exacerbate asthma through regulating miR-136-5p/LIFR axis. FASEB J 2024; 38:e70159. [PMID: 39535503 DOI: 10.1096/fj.202401186rr] [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: 05/28/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Pyroptosis plays a pivotal role in airway epithelial inflammation during the progression of asthma. This study aimed to explore the influence and mechanisms of opa-interacting protein 5 antisense RNA1 (OIP5-AS1) and growth arrest-specific transcript 5 (GAS5) on pyroptosis in asthmatic models. Pyroptosis was induced in Dermatophagoides pteronyssinus 1 (Der p1)-exposed 16HBE cells and ovalbumin (OVA)-challenged rats. Subsequently, pyroptosis and its related molecular mechanisms were investigated. Our results indicated that GATA1, OIP5-AS1, GAS5, and LIFR were upregulated, while miR-136-5p was downregulated in the patients and experimental models of asthma. OIP5-AS1/GAS5 knockdown repressed NLRP3 inflammasome-mediated pyroptosis in 16HBE cells. Mechanistically, OIP5-AS1/GAS5 sponged miR-136-5p to enhance LIFR expression and subsequently activated NF-κB pathway. OIP5-AS1, GAS5, or LIFR-mediated induction of pyroptosis was abrogated by miR-136-5p mimics or NF-κB inhibitors (BAY11-7082). Finally, GATA1 transcriptionally activated OIP5-AS1/GAS5 to trigger pyroptosis, thereby driving asthma progression in vivo and in vitro. In conclusion, OIP5-AS1/GAS5 transcriptionally activated by GATA1 promoted NLRP3 inflammasome-mediated pyroptosis via the modulation of miR-136-5p/LIFR/NF-κB axis and consequently resulted in airway inflammation in asthma. Our results may provide novel therapeutic strategies for asthma.
Collapse
Affiliation(s)
- Suzhi Zhao
- Department of Pulmonary and Critical Care Medicine, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People's Republic of China
| | - Sini Huang
- Department of Pulmonary and Critical Care Medicine, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People's Republic of China
| | - Yawei Wu
- Department of Pulmonary and Critical Care Medicine, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People's Republic of China
| | - Xiaozhou Yao
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People's Republic of China
| | - Xingjun Cai
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People's Republic of China
| |
Collapse
|
11
|
Jia D, Tian X, Chen Y, Liu J, Wang M, Hao Z, Wang C, Zhao D. Preparation of enzymatic hydrolysates of mulberry leaf flavonoids and investigation into its treatment and mechanism for zebrafish inflammatory bowel disease. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109960. [PMID: 39393613 DOI: 10.1016/j.fsi.2024.109960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/24/2024] [Accepted: 10/09/2024] [Indexed: 10/13/2024]
Abstract
INTRODUCTION The incidence of inflammatory bowel disease (IBD) has been increasing year by year in recent years. Flavonoids are the main components of mulberry leaves exerting anti-inflammatory effects and have potential applications in drug screening for IBD treatment. Enzymatic hydrolysis can enhance the bioavailability and activity of flavonoid glycosides. No relevant reports on the potentiation of mulberry leaf flavonoids (MLF) after deglycosylation have been retrieved. METHODS An enzymatic method was used to prepare enzymatic hydrolysates of MLF (EMLF). The protective effect of EMLF on zebrafish with IBD was evaluated by observing zebrafish intestinal length and width, intestinal histopathological morphology, as well as neutrophil and goblet expression. Network pharmacology and metabolomics were performed to explore the mechanism of action of EMLF against IBD. Finally, the mechanism of EMLF against IBD was validated using Western Blot and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS The EMLF was successfully prepared by hydrolyzing MLF with snailase for the first time, and the anti-inflammatory effect of EMLF was clarified to be superior to that of MLF in IBD zebrafish. The network pharmacology and metabolomics studies showed that the mechanism of EMLF for IBD may be related to regulation of purine metabolic pathway. The expression levels of adenosine deaminase (ADA), critical targets in purine metabolism, were significantly down-regulated, while the expression of adenine phosphoribosyltransferase (APRT) and xanthine dehydrogenase (XDH) was significantly increased when compared with the TNBS group. CONCLUSION The results suggested that enzymatic deglycosylation is an effective measure to enhance the anti-inflammatory activity of MLF, which provides referable ideas and methods for the deep processing and utilization of natural drug resources.
Collapse
Affiliation(s)
- Dongsheng Jia
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China; Institute of Cash Crop, Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang, Hebei, 050051, PR China
| | - Xi Tian
- Institute of Cash Crop, Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang, Hebei, 050051, PR China
| | - Yuting Chen
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Jie Liu
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Man Wang
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Zhangsen Hao
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Changshun Wang
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Ding Zhao
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China.
| |
Collapse
|
12
|
Zou Z, Xue Y, Adu-Frimpong M, Wang C, Jin Z, Xu Y, Yu J, Xu X, Zhu Y. Formononetin-Loaded Self-Microemulsion Drug Delivery Systems for Improved Solubility and Oral Bioavailability: Fabrication, Characterization, In Vitro and In Vivo Evaluation. AAPS PharmSciTech 2024; 25:261. [PMID: 39487315 DOI: 10.1208/s12249-024-02975-8] [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: 06/24/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
This study aimed to construct a self-microemulsion drug delivery system (SMEDDS) for Formononetin (FMN) to improve its solubility and bioavailability while combining the nanocrystals (NCs) technology. The SMEDDS prescription composition was optimized with a pseudo-three-phase diagram, followed by a series of in vitro and in vivo evaluations of the selected optimal prescriptions. FMN-NCs loaded SMEDDS showed a homogeneous spherical shape in the Transmission electron microscope and the particle size was measured as (20.65 ± 1.42) nm. The in vitro cumulative release rate in each dissolution medium within 30 min was higher than 80%, much higher than that of FMN (6%) and FMN-NCs (40%); Cellular experiments confirm that the formulation has a high safety profile and significantly promotes cellular uptake. The results of pharmacokinetics and intestinal absorption in rats showed that the relative bioavailability of FMN-NCs and FMN-NCs loaded SMEDDS were (154.80 ± 3.76)% and (557.73 ± 32.88)%, respectively, and both of them significantly increased the rate and extent of absorption of the drug in intestinal segments. FMN-NCs loaded SMEDDS significantly enhanced the solubility and bioavailability of FMN.
Collapse
Affiliation(s)
- Zhihui Zou
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yuanyuan Xue
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), 0215-5321, Navrongo, UK, Ghana
| | - ChengWei Wang
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Zhou Jin
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Ying Xu
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yuan Zhu
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
13
|
Wang J, Wang L, Han L, Han Y, Gu J, Chen Z. Formononetin attenuates hepatic injury in diabetic mice by regulating macrophage polarization through the PTP1B/STAT6 axis. Int Immunopharmacol 2024; 140:112802. [PMID: 39088924 DOI: 10.1016/j.intimp.2024.112802] [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: 04/15/2024] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND Formononetin (FNT) is an isoflavone known for its anti-inflammatory properties and has been shown to reduce insulin resistance in Type 2 Diabetes Mellitus (T2DM). However, its effects and the underlying mechanisms in diabetic liver injury remain largely unexplored. METHODS We established a T2DM-induced liver injury mouse model by feeding high-fat diet, followed by injecting streptozotocin. The mice were then treated with FNT and the liver function in these mice was assessed. Macrophage markers in FNT-treated T2DM mice or human THP-1 cells were evaluated using flow cytometry, RT-qPCR, and Western blotting. The expression of PTP1B and STAT6 in mouse liver tissues and THP-1 cells was analyzed. Molecular docking predicted the interaction between PTP1B and STAT6, which was validated via co-immunoprecipitation (Co-IP) and phos-tag analysis. Microscale thermophoresis (MST) assessed the binding affinity of FNT to PTP1B. RESULTS FNT treatment significantly ameliorated blood glucose levels, hepatocyte apoptosis, inflammatory response, and liver dysfunction in T2DM mice. Moreover, FNT facilitated M2 macrophage polarization in both T2DM mice and high glucose (HG)-induced THP-1-derived macrophages. The PTP1B/STAT6 axis, deregulated in T2DM mice, was normalized by FNT treatment, which counteracted the T2DM-induced upregulation of PTP1B and downregulation of phosphorylated STAT6. Molecular docking and subsequent analyses revealed that PTP1B binds to and dephosphorylates STAT6 at the S325A site. In contrast, FNT strongly binds to PTP1B and influences its expression at the K116A site, promoting M2 polarization of THP-1 cells via downregulation of PTP1B. CONCLUSION Formononetin mitigates diabetic hepatic injury by fostering M2 macrophage polarization via the PTP1B/STAT6 axis.
Collapse
Affiliation(s)
- Jinchun Wang
- Department of Pharmacy, Jiangsu Health Vocational College, 150 Fenghuang W St, Gulou, Nanjing, Jiangsu 211800, China
| | - Lei Wang
- Department of Pharmacy, Jiangsu Health Vocational College, 150 Fenghuang W St, Gulou, Nanjing, Jiangsu 211800, China
| | - Lei Han
- Department of Pharmacy, Jiangsu Health Vocational College, 150 Fenghuang W St, Gulou, Nanjing, Jiangsu 211800, China
| | - Yiwen Han
- Department of Clinical Medicine, Jiangsu Health Vocational College, 150 Fenghuang W St, Gulou, Nanjing, Jiangsu 211800, China
| | - Jun Gu
- Department of Public Health, Nanjing Medical University, 140 Hanzhong Rd, Gulou, Nanjing, Jiangsu 211166, China
| | - Zhujing Chen
- Department of Outpatient, Jurong People's Hospital, Jurong, No 66. Two holy road, Jurong, Zhenjiang, Jiangsu 212400, China.
| |
Collapse
|
14
|
Musa I, Wang ZZ, Yang N, Li XM. Formononetin inhibits IgE by huPlasma/PBMCs and mast cells/basophil activation via JAK/STAT/PI3-Akt pathways. Front Immunol 2024; 15:1427563. [PMID: 39221239 PMCID: PMC11363073 DOI: 10.3389/fimmu.2024.1427563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
Rationale Food allergy is a prevalent disease in the U.S., affecting nearly 30 million people. The primary management strategy for this condition is food avoidance, as limited treatment options are available. The elevation of pathologic IgE and over-reactive mast cells/basophils is a central factor in food allergy anaphylaxis. This study aims to comprehensively evaluate the potential therapeutic mechanisms of a small molecule compound called formononetin in regulating IgE and mast cell activation. Methods In this study, we determined the inhibitory effect of formononetin on the production of human IgE from peripheral blood mononuclear cells of food-allergic patients using ELISA. We also measured formononetin's effect on preventing mast cell degranulation in RBL-2H3 and KU812 cells using beta-hexosaminidase assay. To identify potential targets of formononetin in IgE-mediated diseases, mast cell disorders, and food allergies, we utilized computational modeling to analyze mechanistic targets of formononetin from various databases, including SEA, Swiss Target Prediction, PubChem, Gene Cards, and Mala Cards. We generated a KEGG pathway, Gene Ontology, and Compound Target Pathway Disease Network using these targets. Finally, we used qRT-PCR to measure the gene expression of selected targets in KU812 and U266 cell lines. Results Formononetin significantly decreased IgE production in IgE-producing human myeloma cells and PBMCs from food-allergic patients in a dose-dependent manner without cytotoxicity. Formononetin decreased beta-hexosaminidase release in RBL-2H3 cells and KU812 cells. Formononetin regulates 25 targets in food allergy, 51 in IgE diseases, and 19 in mast cell diseases. KEGG pathway and gene ontology analysis of targets showed that formononetin regulated disease pathways, primary immunodeficiency, Epstein-Barr Virus, and pathways in cancer. The biological processes regulated by formononetin include B cell proliferation, differentiation, immune response, and activation processes. Compound target pathway disease network identified NFKB1, NFKBIA, STAT1, STAT3, CCND1, TP53, TYK2, and CASP8 as the top targets regulated at a high degree by formononetin. TP53, STAT3, PTPRC, IL2, and CD19 were identified as the proteins mostly targeted by formononetin. qPCR validated genes of Formononetin molecular targets of IgE regulation in U266 cells and KU812 cells. In U266 cells, formononetin was found to significantly increase the gene expression of NFKBIA, TP53, and BCL-2 while decreasing the gene expression of BTK TYK, CASP8, STAT3, CCND1, STAT1, NFKB1, IL7R. In basophils KU812 cells, formononetin significantly increased the gene expression of NFKBIA, TP53, and BCL-2 while decreasing the gene expression of BTK, TYK, CASP8, STAT3, CCND1, STAT1, NFKB1, IL7R. Conclusion These findings comprehensively present formononetin's mechanisms in regulating IgE production in plasma cells and degranulation in mast cells.
Collapse
Affiliation(s)
- Ibrahim Musa
- Department of Pathology Microbiology & Immunology, New York Medical College, New York, NY, United States
| | - Zhen-Zhen Wang
- Department of Pathology Microbiology & Immunology, New York Medical College, New York, NY, United States
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Nan Yang
- R&D Division, General Nutraceutical Technology LLC, Elmsford, NY, United States
| | - Xiu-Min Li
- Department of Pathology Microbiology & Immunology, New York Medical College, New York, NY, United States
- Department of Otolaryngology, School of Medicine, New York Medical College, New York, NY, United States
| |
Collapse
|
15
|
Feng Z, Gu L, Lin J, Wang Q, Yu B, Yao X, Feng Z, Zhao G, Li C. Formononetin protects against Aspergillus fumigatus Keratitis: Targeting inflammation and fungal load. Int Immunopharmacol 2024; 132:112046. [PMID: 38593508 DOI: 10.1016/j.intimp.2024.112046] [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: 01/08/2024] [Revised: 03/29/2024] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE To investigate the potential treatment of formononetin (FMN) on Aspergillus fumigatus (A. fumigatus) keratitis with anti-inflammatory and antifungal activity. METHODS The effects of FMN on mice with A. fumigatus keratitis were evaluated through keratitis clinical scores, hematoxylin-eosin (HE) staining, and plate counts. The expression of pro-inflammatory factors was measured using RT-PCR, ELISA, or Western blot. The distribution of macrophages and neutrophils was explored by immunofluorescence staining. The antifungal properties of FMN were assessed through minimum inhibitory concentration (MIC), propidium iodide (PI) staining, fungal spore adhesion, and biofilm formation assay. RESULTS In A. fumigatus keratitis mice, FMN decreased the keratitis clinical scores, macrophages and neutrophils migration, and the expression of TNF-α, IL-6, and IL-1β. In A. fumigatus-stimulated human corneal epithelial cells (HCECs), FMN reduced the expression of IL-6, TNF-α, IL-1β, and NLRP3. FMN also decreased the expression of thymic stromal lymphopoietin (TSLP) and thymic stromal lymphopoietin receptor (TSLPR). Moreover, FMN reduced the levels of reactive oxygen species (ROS) induced by A. fumigatus in HCECs. Furthermore, FMN inhibited A. fumigatus growth, prevented spore adhesion and disrupted fungal biofilm formation in vitro. In vivo, FMN treatment reduced the fungal load in mice cornea at 3 days post infection (p.i.). CONCLUSION FMN demonstrated anti-inflammatory and antifungal properties, and exhibited a protective effect on mouse A. fumigatus keratitis.
Collapse
Affiliation(s)
- Zhuhui Feng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Bing Yu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaofeng Yao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Zheng Feng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|