1
|
Liu J, Qu B, Wang S, Qian L, Liu F, Zhang X, Zhao Q, Chen Y, Chen W, Wang L, Zhang S. Fengshi gutong capsules attenuates CIA-induced RA bone destruction in rats by targeting TNF-α inhibition: Integration and experimental validation of network pharmacology and proteomics. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119535. [PMID: 39987998 DOI: 10.1016/j.jep.2025.119535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fengshi Gutong Capsule (FSGT) is a proprietary Chinese medicine with established clinical efficacy in Rheumatoid arthritis (RA); however, its underlying mechanisms remain unclear. AIM This study aims to elucidate the mechanisms by which FSGT alleviates RA. MATERIALS AND METHODS A collagen-induced arthritis (CIA) rat model was employed to assess the therapeutic effects of FSGT in RA. Network pharmacology and proteomics were integrated to identify potential mechanism and molecular targets, which were further validated via Western blot analysis. Molecular docking and microscale thermophoresis (MST) were utilized to assess the binding affinities of FSGT's active components to key proteins. RESULTS FSGT (280 and 840 mg/kg) alleviated CIA-induced RA in rats without significant side effects. Network pharmacology and label-free proteomic analysis displayed that FSGT exerted its therapeutic effects by modulating inflammation and bone destruction. FSGT significantly reduced serum levels of inflammatory cytokines and their protein expression in the ankle joints and synovial tissues. Additionally, FSGT attenuated bone destruction and significantly reversed the expression of bone destruction-related proteins. Molecular docking revealed that 18 active compounds in FSGT exhibited strong binding affinity for TNF-α, with hypaconitine, 18α-glycyrrhizic acid, and naringenin further validated by MST assays. CONCLUSION FSGT improved CIA-induced RA in rats by targeting TNF-α to reduce inflammation and inhibit bone destruction, offering insights into its therapeutic mechanisms in RA.
Collapse
Affiliation(s)
- Jiahui Liu
- Anhui University of Chinese Medicine, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Biao Qu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Sheng Wang
- Center for Scientific Research, Anhui Medical University, Hefei, 230601, China
| | - Linkai Qian
- Anhui University of Chinese Medicine, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Feifei Liu
- Anhui University of Chinese Medicine, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Xueting Zhang
- Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Quan Zhao
- Bozhou China Sciences Testing Co. , Ltd., Bozhou, 236000, China
| | - Yunna Chen
- Anhui University of Chinese Medicine, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Weidong Chen
- Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Lei Wang
- Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Sheng Zhang
- Anhui University of Chinese Medicine, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China; "The Classic and Innovation" Stroke Research Team, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
2
|
He X, Wu T, He H, Chen L, Han K, Zheng J, Zhang Z, Yuan S, Wang Y, Zhang Y, Zhang X, Xie C. Study of kaempferol in the treatment of rheumatoid arthritis through modulation of the NLRP3/CASP1/GSDMD axis and T-cell activation: Based on network pharmacology, single-cell analysis, and experimental validation. Int Immunopharmacol 2024; 143:113357. [PMID: 39388897 DOI: 10.1016/j.intimp.2024.113357] [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: 07/29/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Kaempferol (Kae) is a natural flavonol compound with excellent anti-inflammatory and immunomodulatory effects, which is of great importance in the treatment of inflammatory diseases. The efficacy of Kae in the treatment of rheumatoid arthritis (RA) has been demonstrated. However, its relevant pharmacodynamic mechanism requires further investigation. PURPOSES This study aimed to further explore the potential mechanism of action of Kae in the treatment of RA using network pharmacology, single-cell analysis, and animal experiments. METHODS Drug target genes were downloaded and screened from the Comparative Toxicogenomics Database (CTD), SwissTargetPrediction database, BindingDB database, and TargetNet database. Transcriptome data from GEO databases (GSE55235, GSE89408, and GSE200815) were selected for disease transcriptome analysis and single-cell matrix data. Network pharmacology and molecular docking were used to investigate the potential mechanism of action of Kae in treating RA. Single-cell analysis, immune infiltration co-expression analyses, and Mendelian-Randomization (MR) studies were conducted to explore the relationship between Kae's target genes and immune cells. Collagen-induced arthritis (CIA) was induced in DBA/1 mouse models through enhanced immunization. Therapeutic efficacy of Kae was assessed using arthritis score, paw swelling index, body weight monitoring, microCT, hematoxylin and eosin (HE) staining, Safranin O-Fast green staining, and Tartrate-resistant acid phosphatase (TRAP) staining. Tissue immunofluorescence and flow cytometry were used to detect expression levels of key genes and immune cell activation status. RESULTS In vivo experiments demonstrated the efficacy of Kae in treating CIA mice. Network pharmacology indicated that Kae might exert anti-inflammatory effects through the NLRP3/CASP1/GSDMD axis. Immune infiltration, single-cell, and MR analyses revealed close associations between Kae's target genes and CD4+, CD8+, and regulatory T cells. Kae inhibited cellular pyroptosis in joint tissues and down-regulated NLRP3, CASP1, and GSDMD expression. Flow cytometry results showed decreased CD4/CD8 ratio, reduced proportion of CD4+ effector memory T cells (Tem), and increased naïve and regulatory T cells (Treg). CONCLUSION Kae might exert anti-inflammatory effects by modulating the NLRP3/CASP1/GSDMD axis to inhibit pyroptosis and suppress overactive immune responses by regulating T-cell proliferation. In summary, Kae demonstrated significant therapeutic efficacy in treating RA.
Collapse
Affiliation(s)
- Xiaoyu He
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Tianyu Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, 21009, China.
| | - Haohua He
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu 233003, China.
| | - Lili Chen
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Ke Han
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Jiaqian Zheng
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Zheng Zhang
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Shuqi Yuan
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Yanxin Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu 233003, China.
| | - Yan Zhang
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Xiaonan Zhang
- Bengbu Medical University Key Laboratory of Cardiovascular and Cerebrovascular Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| | - Changhao Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, Anhui, 233004, China; Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu 233003, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, 287 Changhuai Road, Bengbu, Anhui, 233004, China.
| |
Collapse
|
3
|
Wang Y, Zhao X, Wang J, Zhu X. Norisoboldine Reduces Arthritis Severity by Attenuating Inflammation, Oxidative Stress, and Extracellular Matrix Degradation in a Rat Model of Rheumatoid Arthritis. J Inflamm Res 2024; 17:8839-8852. [PMID: 39564548 PMCID: PMC11575444 DOI: 10.2147/jir.s476824] [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: 05/05/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic autoimmune disorder marked by persistent joint inflammation, pain, and tissue degradation. This study evaluates the therapeutic potential of Norisoboldine (NOR), an isoquinoline alkaloid from Lindera aggregata, in a rat model of RA. Methods Rats were divided into five groups: normal control (G1), RA model (G2), NOR-treated groups at 15 mg/kg (G3) and 30 mg/kg (G4), and methotrexate-treated group (G5). NOR's anti-arthritic effects were assessed by measuring clinical arthritis scores and inflammatory markers (RF, CRP, TNF-α, IL-6, IL-10). Oxidative stress markers (MDA, SOD, catalase, GPx) and pathways (NF-κB/IKKβ and Nrf2/Keap1) were also evaluated. Histopathology assessed synovial inflammation and tissue degradation. Results NOR treatment significantly reduced arthritis severity, as evidenced by decreased clinical arthritis scores and inflammatory markers in RA rats. NOR also exhibited strong antioxidant effects, demonstrated by decreased MDA levels and enhanced SOD, catalase, and GPx activities. NOR further downregulated matrix metalloproteinases (Mmp-2, Mmp-3), aggrecanases (Adamts-4, Adamts-5), and PCNA expression. Histopathology confirmed marked reductions in synovial inflammation and tissue damage in NOR-treated groups. Discussion These findings suggest that NOR's anti-inflammatory and antioxidant properties contribute to reducing both inflammation and the overall severity of RA. NOR's multifaceted actions support its potential as a novel therapeutic agent for RA. Conclusion NOR demonstrates protective effects in RA rats by reducing inflammation, oxidative stress, and extracellular matrix degradation, offering promise as a therapeutic option to manage RA pathology comprehensively.
Collapse
Affiliation(s)
- Ying Wang
- Department of Endocrine Nephrology, The First Affiliated Hospital of Xingtai Medical College (Xingtai First Hospital), Xingtai, Hebei Province, 054001, People's Republic of China
| | - Xiangzhuo Zhao
- Department of Rheumatic Immunology, Xingtai People's Hospital, Xingtai, Hebei Province, 054000, People's Republic of China
| | - Jingxu Wang
- Department of Rheumatic Immunology, Xingtai People's Hospital, Xingtai, Hebei Province, 054000, People's Republic of China
| | - Xiaoli Zhu
- Department of Critical Care Medicine, Xingtai People's Hospital, Xingtai, Hebei Province, 054000, People's Republic of China
| |
Collapse
|
4
|
Guo W, Li H, Wei X, Luo N, Shi M. Molecular and Morphological Characteristics of a Novel Cyst Nematode in the Rhizosphere of Artemisia lavandulaefolia DC. in Gansu Province, Northwest China. Pathogens 2024; 13:881. [PMID: 39452752 PMCID: PMC11510491 DOI: 10.3390/pathogens13100881] [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: 09/09/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
Cyst nematodes are obligate parasitic nematodes found in the fields of many cultivated crops. These nematodes, which have great economic importance, pose a threat to food security, though they are frequently ignored or misdiagnosed as pests because of covert parasitism. A cyst nematode population parasitizing on Artemisia lavandulaefolia DC., one of the traditional Chinese medicines was collected in Gansu Province. The species was diagnosed using integrative taxonomy and molecular approaches. The cyst population is spherical or lemon-shaped, light brown or dark brown in color, with a long neck and a protruding vulval cone. The stylet of the second-stage juvenile is strong, and the front end of the ball at the base of the stylet is concave; the median bulb and excretory pore are prominent; the tail is blunt and circular, and the transparent tail is usually shorter than the stylet. A phylogenetic analysis was carried out using the internal transcribed spacer (ITS) and 28S genes of ribosomal DNA, which further confirmed the presence of Cactodera chenopodiae. According to our literature review, this is the first report on C. chenopodiae in Compositae. By following this research, we can better understand the challenges posed by A. lavandulaefolia DC. and develop effective strategies for managing its spread and impacts. This will help to protect vulnerable ecosystems and ensure the sustainability of agricultural and forestry activities in affected areas.
Collapse
Affiliation(s)
| | - Huixia Li
- Biocontrol Engineering Laboratory of Crop Disease and Pests of Gansu Province, College of Plant Protection, Gansu Agricultural University, Lanzhou 730070, China; (W.G.)
| | | | | | | |
Collapse
|
5
|
Huang C, Zhang Y, Xu Y, Wei S, Yang T, Wang S, Li C, Lin H, Li X, Zhao S, Zhu L, Pan W. Prepared Radix Polygoni Multiflori and emodin alleviate lipid droplet accumulation in nonalcoholic fatty liver disease through MAPK signaling pathway inhibition. Aging (Albany NY) 2024; 16:2362-2384. [PMID: 38284886 PMCID: PMC10911387 DOI: 10.18632/aging.205485] [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: 04/17/2023] [Accepted: 12/06/2023] [Indexed: 01/30/2024]
Abstract
As one of the most common liver diseases, nonalcoholic fatty liver disease (NAFLD) affects almost one-quarter of the world's population. Although the prevalence of NAFLD is continuously rising, effective medical treatments are still inadequate. Radix Polygoni Multiflori (RPM) is a traditional Chinese herbal medicine. As a processed product of RPM, prepared Radix Polygoni Multiflori (PRPM) has been reported to have antioxidant and anti-inflammatory effects. This study investigated whether PRPM treatment could significantly improve NAFLD. We used recent literature, the Herb database and the SwissADME database to isolate the active compounds of PRPM. The OMIM, DisGeNET and GeneCards databases were used to isolate NAFLD-related target genes, and GO functional enrichment and KEGG pathway enrichment analyses were conducted. Moreover, PRPM treatment in NAFLD model mice was evaluated. The results indicate that the target genes are mainly enriched in the AMPK and de novo lipogenesis signaling pathways and that PRPM treatment improves NAFLD disease in model mice. Here, we found the potential benefits of PRPM against NAFLD and demonstrated in vivo and in vitro that PRPM and its ingredient emodin downregulate phosphorylated P38/P38, phosphorylated ERK1/2 and genes related to de novo adipogenesis signaling pathways and reduce lipid droplet accumulation. In conclusion, our findings revealed a novel therapeutic role for PRPM in the treatment of NAFLD and metabolic inflammation.
Collapse
Affiliation(s)
- Changyudong Huang
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Yiqiong Zhang
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Yongjie Xu
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Sijia Wei
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Tingting Yang
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Shuang Wang
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Chengcheng Li
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Hairong Lin
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Xing Li
- School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang 550004, Guizhou, P.R. China
| | - Shuyun Zhao
- Reproductive Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Liying Zhu
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| | - Wei Pan
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, P.R. China
| |
Collapse
|
6
|
Rarison RHG, Truong VL, Yoon BH, Park JW, Jeong WS. Antioxidant and Anti-Inflammatory Mechanisms of Lipophilic Fractions from Polyscias fruticosa Leaves Based on Network Pharmacology, In Silico, and In Vitro Approaches. Foods 2023; 12:3643. [PMID: 37835296 PMCID: PMC10573055 DOI: 10.3390/foods12193643] [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: 09/04/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Polyscias fruticosa leaf (PFL) has been used in food and traditional medicine for the treatment of rheumatism, ischemia, and neuralgia. However, the lipophilic components of PFL and their biological properties remain unknown. This study, integrating network pharmacology analysis with in silico and in vitro approaches, aimed to elucidate the antioxidant and anti-inflammatory capacities of lipophilic extracts from PFL. A total of 71 lipophilic compounds were identified in PFL using gas chromatography-mass spectrometry. Network pharmacology and molecular docking analyses showed that key active compounds, mainly phytosterols and sesquiterpenes, were responsible for regulating core target genes, such as PTGS2, TLR4, NFE2L2, PRKCD, KEAP1, NFKB1, NR1l2, PTGS1, AR, and CYP3A4, which were mostly enriched in oxidative stress and inflammation-related pathways. Furthermore, lipophilic extracts from PFL offered powerful antioxidant capacities, as evident in our cell-free antioxidant assays. These extracts also provided a protection against oxidative stress by inducing the expression of catalase and heme oxygenase-1 in lipopolysaccharide (LPS)-treated RAW 264.7 cells. Additionally, lipophilic fractions from PFL showed anti-inflammatory potential in downregulating the level of pro-inflammatory factors in LPS-treated macrophages. Overall, these findings provide valuable insights into the antioxidant and anti-inflammatory properties of lipophilic extracts from PFL, which can be used as a fundamental basis for developing nutraceuticals and functional foods.
Collapse
Affiliation(s)
- Razanamanana H. G. Rarison
- School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Van-Long Truong
- School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
- Food and Bio-industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea (V.-L.T.)
| | - Byoung-Hoon Yoon
- School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ji-Won Park
- School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Woo-Sik Jeong
- School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
- Food and Bio-industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea (V.-L.T.)
| |
Collapse
|