1
|
Yang X, Li J, Xu C, Zhang G, Che X, Yang J. Potential mechanisms of rheumatoid arthritis therapy: Focus on macrophage polarization. Int Immunopharmacol 2024; 142:113058. [PMID: 39236455 DOI: 10.1016/j.intimp.2024.113058] [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: 07/18/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that affects multiple organs and systems in the human body, often leading to disability. Its pathogenesis is complex, and the long-term use of traditional anti-rheumatic drugs frequently results in severe toxic side effects. Therefore, the search for a safer and more effective antirheumatic drug is extremely important for the treatment of RA. As important immune cells in the body, macrophages are polarized. Under pathological conditions, macrophages undergo proliferation and are recruited to diseased tissues upon stimulation. In the local microenvironment, they polarize into different types of macrophages in response to specific factors and perform unique functions and roles. Previous studies have shown that there is a link between macrophage polarization and RA, indicating that certain active ingredients can ameliorate RA symptoms through macrophage polarization. Notably, Traditional Chinese medicine (TCM) monomer component and compounds demonstrate a particular advantage in this process. Building upon this insight, we reviewed and analyzed recent studies to offer valuable and meaningful insights and directions for the development and application of anti-rheumatic drugs.
Collapse
Affiliation(s)
- Xinyu Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinling Li
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengchao Xu
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinzhen Che
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
2
|
Ahmed N, Tabassum N, Rashid PT, Deea BJ, Richi FT, Chandra A, Agarwal S, Mollick S, Dipto KZ, Mim SA, Alam S. Clitoria ternatea L. (Butterfly Pea) Flower Against Endometrial Pain: Integrating Preliminary In Vivo and In Vitro Experimentations Supported by Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulation Studies. Life (Basel) 2024; 14:1473. [PMID: 39598271 PMCID: PMC11595475 DOI: 10.3390/life14111473] [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: 08/28/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024] Open
Abstract
Clitoria ternatea L. (CT) is a perennial herbaceous plant with deep blue flowers native to tropical Asia. This work explores the endometrial pain (EP) regulation of CT flower through a multifaceted approach. Phytochemical screening unveiled the presence of alkaloids, steroids, flavonoids, glycosides, and tannins in CT flower methanolic extract (ME). In the in vitro membrane stabilizing experiment, the ME demonstrated 91.47% suppression of heat-induced hemolysis. Upon carrageenan-induced paw edema assay conducted on male Swiss albino mice at doses of 200 mg/kg and 400 mg/kg, 65.28% and 81.89% inhibition rates, respectively, of paw edema were reported. For the same doses, upon acetic acid-induced-writhing assay, 75.6% and 76.78% inhibition rates, respectively, were observed. For network pharmacology analyses, a protein-protein interaction network was constructed for 92 overlapping gene targets of CT and EP, followed by GO and KEGG pathway enrichment analyses. Network pharmacology-based investigation identified the anti-EP activity of CT to be mostly regulated by the proteins SRC homology, ESR1, and PI3KR1. Physicochemical, pharmacokinetic, and toxicity property predictions for the compounds with stable ligand-target interactions and a molecular dynamics simulation for the highest interacting complex further validated these findings. This work affirmed the anti-EP role of CT flower against EP, suggesting a probable molecular mechanism involved.
Collapse
Affiliation(s)
- Najneen Ahmed
- Department of Pharmacy, East West University, Dhaka 1212, Bangladesh; (N.T.); (P.T.R.); (B.J.D.); (K.Z.D.); (S.A.M.)
| | - Nazifa Tabassum
- Department of Pharmacy, East West University, Dhaka 1212, Bangladesh; (N.T.); (P.T.R.); (B.J.D.); (K.Z.D.); (S.A.M.)
| | - Parisa Tamannur Rashid
- Department of Pharmacy, East West University, Dhaka 1212, Bangladesh; (N.T.); (P.T.R.); (B.J.D.); (K.Z.D.); (S.A.M.)
| | - Basrat Jahan Deea
- Department of Pharmacy, East West University, Dhaka 1212, Bangladesh; (N.T.); (P.T.R.); (B.J.D.); (K.Z.D.); (S.A.M.)
| | - Fahmida Tasnim Richi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh;
| | - Anshuman Chandra
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (A.C.); (S.A.)
| | - Shilpi Agarwal
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (A.C.); (S.A.)
| | - Saima Mollick
- Pharmaceutical Research Division, BCSIR Dhaka Laboratories, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh;
| | - Kaushik Zaman Dipto
- Department of Pharmacy, East West University, Dhaka 1212, Bangladesh; (N.T.); (P.T.R.); (B.J.D.); (K.Z.D.); (S.A.M.)
| | - Sadia Afrin Mim
- Department of Pharmacy, East West University, Dhaka 1212, Bangladesh; (N.T.); (P.T.R.); (B.J.D.); (K.Z.D.); (S.A.M.)
| | - Safaet Alam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh;
- Chemical Research Division, BCSIR Dhaka Laboratories, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh
| |
Collapse
|
3
|
Li C, Xue Q, Li H, Peng Y, Wu Y, Yuan M, Duan Q, Hong X, Chen G, Liao F, Wu P, Miao C. Huangqin Qingre Chubi Capsule improves rheumatoid arthritis accompanied depression through the Wnt1/β-catenin signaling pathway. Int Immunopharmacol 2024; 138:112474. [PMID: 38917529 DOI: 10.1016/j.intimp.2024.112474] [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: 03/30/2024] [Revised: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024]
Abstract
AIM OF THE STUDY Research on the mechanism of Huangqin Qingre Chubi Capsules (HQC) in improving rheumatoid arthritis accompanied depression (RA-dep) model rats. METHODS We employed real-time qPCR (RT-qPCR), western blotting (WB), confocal microscopy, bioinformatics, and other methods to investigate the anti-RA-dep effects of HQC and its underlying mechanisms. RESULTS HQC alleviated the pathological indexes of inflammation and depression in RA-dep model rats, decreased the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6, increased the levels of norepinephrine(NE) and serotonin(5-HT), and improved the injury of hippocampus. The analysis of network pharmacology suggests that HQC may target the Wnt/β-catenin pathway in the treatment of RA-dep. Furthermore, molecular dynamics simulations revealed a strong affinity between HQC and the Wnt1 molecule. RT-qPCR and Western Blot (WB) experiments confirmed the critical role of the Wnt1/β-catenin signaling pathway in the treatment of RA-dep model rats with HQC. In vitro, the HQC drug-containing serum (HQC-serum) activates the Wnt1/β-catenin signaling pathway in hippocampal cells and, in conjunction with Wnt1, ameliorates RA-dep. In summary, HQC exerts its anti-inflammatory and antidepressant effects in the treatment of RA-dep by binding to Wnt1 and regulating the Wnt1/β-catenin signaling pathway. CONCLUSIONS HQC improved the inflammatory reaction and depression-like behavior of RA-dep model rats by activating Wnt1/β-catenin signal pathway. This study revealed a new pathogenesis of RA-dep and contributes to the clinical promotion of HQC in the treatment of RA-dep.
Collapse
Affiliation(s)
- Chen Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Qiuyun Xue
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Hui Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yanhui Peng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yajie Wu
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Meiling Yuan
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Qiangjun Duan
- Department of Experimental Teaching Center, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xinghui Hong
- Department of Experimental Teaching Center, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Guangliang Chen
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Faxue Liao
- The First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Public Health Clinical Center, Hefei, China.
| | - Peng Wu
- Department of Anatomy, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; Institute of Prevention and Treatment of Rheumatoid Arthritis, Anhui University of Chinese Medicine, Hefei, Anhui, China; School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
4
|
Wang S, Chen B, Du R, Zhong M, Zhang C, Jin X, Cui X, Zhou Y, Kang Q, Xu H, Li Y, Wu Q, Tong G, Luo L. An herbal formulation "Shugan Xiaozhi decoction" ameliorates methionine/choline deficiency-induced nonalcoholic steatohepatitis through regulating inflammation and apoptosis-related pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118127. [PMID: 38583728 DOI: 10.1016/j.jep.2024.118127] [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: 12/23/2023] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shugan Xiaozhi (SGXZ) decoction is a traditional Chinese medicine used for treating nonalcoholic steatohepatitis (NASH). It has been used clinically for over 20 years and proved to be effective; however, the molecular mechanism underlying the effects of SGXZ decoction remains unclear. AIM OF THE STUDY We analyzed the chemical components, core targets, and molecular mechanisms of SGXZ decoction to improve NASH through network pharmacology and in vivo experiments. MATERIALS AND METHODS The chemical components, core targets, and related signaling pathways of SGXZ decoction intervention in NASH were predicted using network pharmacology. Molecular docking was performed to verify chemical components and their core targets. The results were validated in the NASH model treated with SGXZ decoction. Mouse liver function was assessed by measuring ALT and AST levels. TC and TG levels were determined to evaluate lipid metabolism, and lipid deposition was assessed via oil red O staining. Mouse liver damage was determined via microscopy following hematoxylin and eosin staining. Liver fibrosis was assessed via Masson staining. Western blot (WB) and immunohistochemical (IHC) analyses were performed to detect inflammation and the expression of apoptosis-related proteins, including IL-1β, IL-6, IL-18, TNF-α, MCP1, p53, FAS, Caspase-8, Caspase-3, Caspase-9, Bax, Bid, Cytochrome c, Bcl-2, and Bcl-XL. In addition, WB and IHC were used to assess protein expression associated with the TLR4/MyD88/NF-κB pathway. RESULTS Quercetin, luteolin, kaempferol, naringenin, and nobiletin in SGXZ decoction were effective chemical components in improving NASH, and TNF-α, IL-6, and IL-1β were the major core targets. Molecular docking indicated that these chemical components and major core targets might interact. KEGG pathway analysis showed that the pathways affected by SGXZ decoction, primarily including apoptosis and TLR4/NF-κB signaling pathways, interfere with NASH. In vivo experiments indicated that SGXZ decoction considerably ameliorated liver damage, fibrosis, and lipid metabolism disorder in MCD-induced NASH mouse models. In addition, WB and IHC verified the underlying molecular mechanisms of SGXZ decoction as predicted via network pharmacology. SGXZ decoction inhibited the activation of apoptosis-related pathways in MCD-induced NASH mice. Moreover, SGXZ decoction suppressed the activation of TLR4/MyD88/NF-κB pathway in MCD-induced NASH mice. CONCLUSION SGXZ decoction can treat NASH through multiple targets and pathways. These findings provide new insights into the effective treatment of NASH using SGXZ decoction.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Bohao Chen
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Ruili Du
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Mei Zhong
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Chunmei Zhang
- School of Basic Medical Science of Luoyang Polytechnic, No. 6 Keji Avenue, Yibin District, Henan, 471099, China
| | - Xiaoming Jin
- Department of Nephrology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Xiang Cui
- Ankang Traditional Chinese Medicine Hospital, Ankang, 725000, Shaanxi, China
| | - Yuhang Zhou
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Qinyang Kang
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Hang Xu
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Yuting Li
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China.
| | - Guangdong Tong
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China.
| | - Lidan Luo
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China.
| |
Collapse
|
5
|
Alejandra SUM, Salud P, Elisa VA, Enrique JF, Julissa RM, Jaime T, Alejandro Z, David Osvaldo SS, Maribel HR. Triterpenes G-A and G-E from Galphimia glauca with anti-inflammatory and anti-arthritic activity in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118104. [PMID: 38531431 DOI: 10.1016/j.jep.2024.118104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/17/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Galphimia glauca is a medicinal plant that treats inflammatory and anti-rheumatic problems. Its anti-inflammatory capacity has been reported pharmacologically, attributed to the triterpenes G-A and G-E. AIM The objective of the present work was to measure the anti-inflammatory and immunomodulatory effect of the methanolic extract (GgMeOH) of Galphimia glauca and the isolated galphimines G-A and G-E, first in an acute test of plantar edema with carrageenan, and later in the model of experimental-induced arthritis with CFA. The effect was measured by quantifying joint inflammation, the concentration of pro- (TNF-α, IL-6, IL-17) and anti-inflammatory (IL-10, and IL-4) cytokines, and the ADA enzyme in joints, kidneys, and spleen from mice with experimental arthritis. METHOD The extract and the active triterpenes were obtained according to established methods using different chromatographic techniques. Female ICR strain mice were subjected to intraplantar administration with carrageenan and treated with different doses of GgMeOH, G-A, and G-E; edema was monitored at different times. Subsequently, the concentration of TNF-a and IL-10 in the spleen and swollen paw was quantified. Meloxicam (MEL) was used as an anti-inflammatory control drug. The most effective doses of each treatment were analyzed using a complete Freunds adjuvant (CFA)-induced experimental arthritis model. Joint inflammation was followed throughout the experiment. Ultimately, the concentration of inflammation markers, oxidant stress, and ADA activity was quantified. In this experimental stage, methotrexate (MTX) was used as an antiarthritic drug. RESULTS Treatments derived from G. glauca, GgMeOH (DE50 = 158 mg/kg), G-A (DE50 = 2 mg/kg), and G-E (DE50 = 1.5 mg/kg) caused an anti-inflammatory effect in the plantar edema test with carrageenan. In the CFA model, joint inflammation decreased with all natural treatments; GgMeOH and G-A inhibited the ADA enzyme in all organs analyzed (joints, serum, spleen, left and right kidneys), while G-E inhibited the enzyme in joints, serum, and left kidney. CFA caused an increase in the weight index of the organs, an effect that was counteracted by the administration of G. glauca treatments, which also modulate the response to the cytokines analyzed in the different organs (IL-4, IL-10, IL-17, IL-6, and TNF- α). CONCLUSION It is shown, for the first time, that the GgMeOH extract and the triterpenes G-A and G-E of Galphimia glauca have an anti-arthritic effect (anti-inflammatory, immunomodulatory, antioxidant, and ADA inhibitor), using an experimental arthritis model with CFA. Therefore, knowledge of the plant as a possible therapeutic agent for this rheumatic condition is expanding.
Collapse
Affiliation(s)
- Santillan-Urquiza Mayra Alejandra
- Centro de Investigación Biomédica Del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec, Morelos, C.P, 62790, Mexico; Departamento de Ciencias de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco No. 186, Col. Leyes de Reforma 1a Sección, Ciudad de México, C.P., 09310, Mexico.
| | - Pérez Salud
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, A, Calz. Del Hueso 1100, Coapa, Villa Quietud, Coyoacán, Ciudad de México, C.P., 04960, Mexico.
| | - Vega-Avila Elisa
- Departamento de Ciencias de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco No. 186, Col. Leyes de Reforma 1a Sección, Ciudad de México, C.P., 09310, Mexico.
| | - Jiménez-Ferrer Enrique
- Centro de Investigación Biomédica Del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec, Morelos, C.P, 62790, Mexico.
| | - Rendón-Martínez Julissa
- Centro de Investigación Biomédica Del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec, Morelos, C.P, 62790, Mexico.
| | - Tortoriello Jaime
- Centro de Investigación Biomédica Del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec, Morelos, C.P, 62790, Mexico.
| | - Zamilpa Alejandro
- Centro de Investigación Biomédica Del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec, Morelos, C.P, 62790, Mexico.
| | - Salinas-Sánchez David Osvaldo
- Centro de Investigación en Biodiversidad y Conservación, Universidad Autónoma Del Estado de Morelos (UAEM), Av. Universidad 1001, Col. Chamilpa, Cuernavaca, 62209, Morelos, Mexico.
| | - Herrera-Ruiz Maribel
- Centro de Investigación Biomédica Del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec, Morelos, C.P, 62790, Mexico.
| |
Collapse
|
6
|
Gao C, Song XD, Chen FH, Wei GL, Guo CY. The protective effect of natural medicines in rheumatoid arthritis via inhibit angiogenesis. Front Pharmacol 2024; 15:1380098. [PMID: 38881875 PMCID: PMC11176484 DOI: 10.3389/fphar.2024.1380098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/10/2024] [Indexed: 06/18/2024] Open
Abstract
Rheumatoid arthritis is a chronic immunological disease leading to the progressive bone and joint destruction. Angiogenesis, accompanied by synovial hyperplasia and inflammation underlies joint destruction. Delaying or even blocking synovial angiogenesis has emerged as an important target of RA treatment. Natural medicines has a long history of treating RA, and numerous reports have suggested that natural medicines have a strong inhibitory activity on synovial angiogenesis, thereby improving the progression of RA. Natural medicines could regulate the following signaling pathways: HIF/VEGF/ANG, PI3K/Akt pathway, MAPKs pathway, NF-κB pathway, PPARγ pathway, JAK2/STAT3 pathway, etc., thereby inhibiting angiogenesis. Tripterygium wilfordii Hook. f. (TwHF), sinomenine, and total glucoside of Paeonia lactiflora Pall. Are currently the most representative of all natural products worthy of development and utilization. In this paper, the main factors affecting angiogenesis were discussed and different types of natural medicines that inhibit angiogenesis were systematically summarized. Their specific anti-angiogenesis mechanisms are also reviewed which aiming to provide new perspective and options for the management of RA by targeting angiogenesis.
Collapse
Affiliation(s)
- Chang Gao
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Xiao-Di Song
- Gannan Medical University, Jiangxi, Ganzhou, China
| | - Fang-Hui Chen
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Gui-Lin Wei
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Chun-Yu Guo
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| |
Collapse
|
7
|
Li Y, Yang J, Zhang Y, Zhang C, Wei Y, Wang Y, Wu N, Sun J, Wu Z. [The Miao medicine Sidaxue alleviates rheumatoid arthritis in rats possibly by downregulating matrix metalloproteinases]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:739-747. [PMID: 38708508 DOI: 10.12122/j.issn.1673-4254.2024.04.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
OBJECTIVE To explore the inhibitory effect of Sidaxue, a traditional Miao herbal medicine formula, on articular bone and cartilage destruction and synovial neovascularization in rats with collagen-induced arthritis (CIA). METHODS In a SD rat model of CIA, we tested the effects of daily gavage of Sidaxue at low, moderate and high doses (10, 20, and 40 g/kg, respectively) for 21 days, with Tripterygium glycosides (GTW) as the positive control, on swelling in the hind limb plantar regions by arthritis index scoring. Pathologies in joint synovial membrane of the rats were observed with HE staining, and serum TNF-α and IL-1β levels were detected with ELISA. The expressions of NF-κB p65, matrix metalloproteinase 1 (MMP1), MMP2 and MMP9 at the mRNA and protein levels in the synovial tissues were detected using real-time PCR and Western blotting. Network pharmacology analysis was conducted to identify the important target proteins in the pathways correlated with the therapeutic effects of topical Sidaxue treatment for RA, and the core target proteins were screened by topological analysis. RESULTS Treatment with GTW and Sidaxue at the 3 doses all significantly alleviated plantar swelling, lowered arthritis index scores, improved cartilage and bone damage and reduced neovascularization in CIA rats (P<0.05), and the effects of Sidaxue showed a dose dependence. Both GTW and Sidaxue treatments significantly lowered TNF-α, IL-1β, NF-κB p65, MMP1, MMP2, and MMP9 mRNA and protein expressions in the synovial tissues of CIA rats (P<0.05). Network pharmacological analysis identified MMPs as the core proteins associated with topical Sidaxue treatment of RA. CONCLUSION Sidaxue alleviates articular bone and cartilage damages and reduces synovial neovascularization in CIA rats possibly by downregulating MMPs via the TNF-α/IL-1β/NF-κB-MMP1, 2, 9 signaling pathway, and MMPs probably plays a key role in mediating the effect of Sidaxue though the therapeutic pathways other than oral administration.
Collapse
Affiliation(s)
- Y Li
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - J Yang
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Y Zhang
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - C Zhang
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Y Wei
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Y Wang
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - N Wu
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - J Sun
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Z Wu
- Laboratory of Chemistry and Biochemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| |
Collapse
|
8
|
Xu Y, Yang J, Han X, Gan C, Wei X. Active substance and mechanisms of Actinidia chinensis Planch for the treatment of breast cancer was explored based on network pharmacology and in silico study. Medicine (Baltimore) 2024; 103:e37829. [PMID: 38608062 PMCID: PMC11018190 DOI: 10.1097/md.0000000000037829] [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: 11/07/2023] [Accepted: 03/15/2024] [Indexed: 04/14/2024] Open
Abstract
In this paper, our objective was to investigate the potential mechanisms of Actinidia chinensis Planch (ACP) for breast cancer treatment with the application of network pharmacology, molecular docking, and molecular dynamics. "Mihoutaogen" was used as a key word to query the Traditional Chinese Medicine Systems Pharmacology database for putative ingredients of ACP and its related targets. DrugBank, GeneCards, Online Mendelian Inheritance in Man, and therapeutic target databases were used to search for genes associated with "breast cancer." Using Cytoscape 3.9.0 we then constructed the protein-protein interaction and drug-ingredient-target-disease networks. An enrichment analysis of Kyoto encyclopedia of genes and genomes pathway and gene ontology were performed to exploration of the signaling pathways associated with ACP for breast cancer treatment. Discovery Studio software was applied to molecular docking. Finally, the ligand-receptor complex was subjected to a 50-ns molecular dynamics simulation using the Desmond_2020.4 tools. Six main active ingredients and 176 targets of ACP and 2243 targets of breast cancer were screened. There were 118 intersections of targets for both active ingredients and diseases. Tumor protein P53 (TP53), AKT serine/threonine kinase 1 (AKT1), estrogen receptor 1 (ESR1), Erb-B2 receptor tyrosine kinase 2 (ERBB2), epidermal growth factor receptor (EGFR), Jun Proto-Oncogene (JUN), and Heat Shock Protein 90 Alpha Family Class A Member 1 (HSP90AA1) selected as the most important genes were used for verification by molecular docking and molecular dynamics simulation. The primary active compounds of ACP against breast cancer were predicted preliminarily, and its mechanism was studied, thereby providing a theoretical basis for future clinical studies.
Collapse
Affiliation(s)
- Yujing Xu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, P. R. China
| | - Jinrong Yang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, P. R. China
| | - Xiaoyu Han
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, P. R. China
| | - Chunchun Gan
- School of Medicine, Quzhou College of Technology, Quzhou 324000, P. R. China
| | - Xiaopeng Wei
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, P. R. China
| |
Collapse
|
9
|
Ren C, Carrillo ND, Cryns VL, Anderson RA, Chen M. Environmental pollutants and phosphoinositide signaling in autoimmunity. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133080. [PMID: 38091799 PMCID: PMC10923067 DOI: 10.1016/j.jhazmat.2023.133080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 02/08/2024]
Abstract
Environmental pollution stands as one of the most critical challenges affecting human health, with an estimated mortality rate linked to pollution-induced non-communicable diseases projected to range from 20% to 25%. These pollutants not only disrupt immune responses but can also trigger immunotoxicity. Phosphoinositide signaling, a pivotal regulator of immune responses, plays a central role in the development of autoimmune diseases and exhibits high sensitivity to environmental stressors. Among these stressors, environmental pollutants have become increasingly prevalent in our society, contributing to the initiation and exacerbation of autoimmune conditions. In this review, we summarize the intricate interplay between phosphoinositide signaling and autoimmune diseases within the context of environmental pollutants and contaminants. We provide an up-to-date overview of stress-induced phosphoinositide signaling, discuss 14 selected examples categorized into three groups of environmental pollutants and their connections to immune diseases, and shed light on the associated phosphoinositide signaling pathways. Through these discussions, this review advances our understanding of how phosphoinositide signaling influences the coordinated immune response to environmental stressors at a biological level. Furthermore, it offers valuable insights into potential research directions and therapeutic targets aimed at mitigating the impact of environmental pollutants on the pathogenesis of autoimmune diseases. SYNOPSIS: Phosphoinositide signaling at the intersection of environmental pollutants and autoimmunity provides novel insights for managing autoimmune diseases aggravated by pollutants.
Collapse
Affiliation(s)
- Chang Ren
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Noah D Carrillo
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Vincent L Cryns
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Richard A Anderson
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mo Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
10
|
Wen Y, Zhang S, Meng X, Zhao C, Hou B, Zhu X, Cai W, Zhou Y, Qiu L, Sun H. Water extracts of Tibetan medicine Wuweiganlu attenuates experimental arthritis via inducing macrophage polarization towards the M2 type. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116934. [PMID: 37480967 DOI: 10.1016/j.jep.2023.116934] [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: 06/03/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wuweiganlu (WGL) is a well-known formulation described in the "Four Medical Scriptures of Tibetan medicine", which is mainly used for the treatment of Rheumatoid Arthritis (RA) and other chronic ailments prescribed by Tibetan medicine. Nonetheless, the active constituents present in the water extracts of Wuweiganlu (WGLWE) specifically targeting arthritis treatment are largely unknown. AIM OF THE STUDY The aim of this study is to explore the effects and underlying mechanisms of the active components in WGLWE on RA. MATERIALS AND METHODS We utilized ultra-performance liquid chromatography coupled with Q-TOF mass spectrometry (UPLC-Q-TOF-MS) to identify the main chemical compositions of WGLWE. The polarization effect of WGLWE on bone marrow-derived macrophages (BMDM) was determined. A rat model of collagen-induced arthritis (CIA) was established by injecting an emulsion of bovine type II collagen mixed with an equal volume of incomplete Freund's adjuvant into the tail, paw and back of rats. A WGLWE-based ointment was topically applied to the legs and paws of the rats for 30 days. The rats' ankles were photographed to measure the degree of swelling. Micro-CT was used to image the knee joint and paw of rats, and the bone mineral density (BMD) and bone volume fraction (BV/TV) of knee joint in rats were analyzed. High-frequency ultrasound imaging of the rat knee joint was performed to observe knee joint effusion. Further, the serum levels of tumor necrosis factor (TNF-α), interleukin-6 (IL-6), IL-10, and arginine (Arg-1) in CIA rats were detected by enzyme-linked immunosorbent assay (ELISA). Immunohistochemistry (IHC) and immunofluorescence (IF) co-staining were employed to detect the expression levels of inflammatory factors in synovium. RESULTS A total of 28 main components were identified in WGLWE, and these compounds can directly bind to the inflammatory pathway proteins such as JAK2, NFκB and STAT3. In vitro experiments demonstrated that WGLWE promoted the transformation of M1 macrophages into M2 macrophages and suppressed the release of proinflammatory cytokines TNF-α and IL-6. In vivo studies showed that WGLWE effectively reduced ankle swelling, alleviated knee joint effusion, and improved BV/TV while also reducing synovial inflammation levels. Furthermore, WGLWE compounds induced the transition of M1-type macrophages to M2-type macrophages in synovial tissue, resulting in decreased secretion of inflammatory factors TNF-α, WGLWE improved the synovial inflammatory state. CONCLUSION Our results indicated that WGLWE alleviated joint inflammation in CIA rats and the underlying mechanism may be related to inducing the transformation of bone marrow-derived M1 macrophages to M2 macrophages, leading to an increase in the secretion of anti-inflammatory factors and a decrease in pro-inflammatory factors. Therefore, WGLWE may be used as a potential herbal preparation for the treatment of RA.
Collapse
Affiliation(s)
- Yuanyuan Wen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China
| | - Shijie Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China
| | - Xinyu Meng
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China
| | - Chenyang Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China
| | - Bao Hou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China
| | - Xuexue Zhu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China
| | - Weiwei Cai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China
| | - Yuetao Zhou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China
| | - Liying Qiu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China.
| | - Haijian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| |
Collapse
|
11
|
Wang J, Li J, Hu M. Mechanism analysis of Buyang Huanwu decoction in treating atherosclerosis based on network pharmacology and in vitro experiments. Chem Biol Drug Des 2024; 103:e14447. [PMID: 38230788 DOI: 10.1111/cbdd.14447] [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: 09/28/2023] [Revised: 12/04/2023] [Accepted: 12/27/2023] [Indexed: 01/18/2024]
Abstract
Atherosclerosis (AS) is one of the main risk factors of ischemic cardiovascular and cerebrovascular diseases. Buyang Huanwu decoction (BYHWT) is a classic Chinese medicine prescription that is used for treating AS. However, the underlying pharmacological mechanism remains unclear. This study aims to clarify the molecular mechanism of BYHWT in treatment of AS through network pharmacology and in vitro experiments. Molecular structure information and targets of core components of BYHWT were obtained from PubChem and UniProtKB databases. Genes involved in AS were obtained from DisGeNet, GeneCards and OMIM databases. The core targets of BYHWT in AS treatment were identified by protein-protein interaction (PPI) network analysis with STRING platform, and analyzed by gene ontology (GO) analysis and the Kyoto Encyclopedia of Genes and Genomics (KEGG) pathway enrichment analysis. Molecular docking was used to verify the binding affinity between the core targets and the bioactive ingredients. HUVEC viability, inflammatory response and mRNA expression levels of core target genes were evaluated by cell counting kit 8 assay, enzyme-linked immunosorbent assay (ELISA) and qRT-PCR. A total of 60 candidate compounds and 325 predicted target genes were screened. PPI network analysis suggested that TP53, SRC, STAT3, and AKT1 may be the core targets. BYHWT in AS treatment was associated with 46 signaling pathways. GA120, baicalein, and 3,9-di-o-methylnissolin had good binding affinity with core target proteins. Baicalein treatment could significantly promoted the viability and repress the inflammatory response of HUVEC cells stimulated by ox-LDL. In addition, Baicalein can regulate the expression of core targets including AKT1, MAPK1, PIK3CA, JUN, TP53, SRC, EGFR, and ESR1. In conclusion, BYHWT and its main bioactive component baicalein, inhibit inflammatory response and modulate multiple downstream genes of endothelial cells, and show good potential to block the progression of AS and cardiovascular/cerebrovascular diseases.
Collapse
Affiliation(s)
- Jing Wang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiajun Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Hu
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Chen T, Li S, Lian D, Hu Q, Hou H, Niu D, Li H, Song L, Gao Y, Chen Y, Hu X, Li J, Ye Z, Peng B, Zhang G. Integrated Network Pharmacology and Experimental Approach to Investigate the Protective Effect of Jin Gu Lian Capsule on Rheumatoid Arthritis by Inhibiting Inflammation via IL-17/NF-κB Pathway. Drug Des Devel Ther 2023; 17:3723-3748. [PMID: 38107658 PMCID: PMC10725692 DOI: 10.2147/dddt.s423022] [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: 06/22/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Purpose This study aimed to investigate the main pharmacological action and underlying mechanisms of Jin Gu Lian Capsule (JGL) against rheumatoid arthritis (RA) based on network pharmacology and experimental verification. Methods Network pharmacology approaches were performed to explore the core active compounds of JGL, key therapeutic targets, and signaling pathways. Molecular docking was used to predict the binding affinity of compounds with targets. In vivo experiments were undertaken to validate the findings from network analysis. Results A total of 52 targets were identified as candidate JGL targets for RA. Sixteen ingredients were identified as the core active compounds, including, quercetin, myricetin, salidroside, etc. Interleukin-1 beta (IL1B), transcription factor AP-1 (JUN), growth-regulated alpha protein (CXCL1), C-X-C motif chemokine (CXCL)3, CXCL2, signal transducer and activator of transcription 1 (STAT1), prostaglandin G/H synthase 2 (PTGS2), matrix metalloproteinase (MMP)1, inhibitor of nuclear factor kappa-B kinase subunit beta (IKBKB) and transcription factor p65 (RELA) were obtained as the key therapeutic targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed that the efficacy of JGL was functionally involved in regulating immune-mediated inflammation, in which IL-17/NF-κB signaling was recommended as one of the main pathways. Molecular docking suggested that the core active compounds bound strongly to their respective targets. Experimentally, JGL treatment mitigated inflammation, showed analgesic activity, and ameliorated collagen-induced arthritis. Enzyme-linked immunosorbent assay showed that JGL effectively reduced the serum levels of cytokines, chemokines, and MMPs. Immunohistochemistry staining showed that JGL markedly reduced the expression of the targets in IL-17/NF-κB pathway including IL-17A, IL-17RA, NF-κB p65, C-X-C motif ligand 2, MMP1 and MMP13. Conclusion This investigation provided evidence that JGL may alleviate RA symptoms by partially inhibiting the immune-mediated inflammation via IL-17/NF-κB pathway.
Collapse
Affiliation(s)
- Tengfei Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Sihan Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Dongyin Lian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qin Hu
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, People's Republic of China
| | - Hongping Hou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Delian Niu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Han Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Ling Song
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Yunhang Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Xiaoru Hu
- National Institute for Food and Drug Control, Beijing, People's Republic of China
| | - Jianrong Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Zuguang Ye
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Bo Peng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Guangping Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
13
|
Meng F, Tao X, Li L, Jia W, Yang X, Yang Y. Network Pharmacology and Molecular Docking Explore the Mechanism of Mubiezi-Yinyanghuo Herb Pair in the Treatment of Rheumatoid Arthritis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:4502994. [PMID: 38106514 PMCID: PMC10723923 DOI: 10.1155/2023/4502994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/06/2023] [Accepted: 09/08/2023] [Indexed: 12/19/2023]
Abstract
Objective Our previous studies have shown that the Mubiezi-Yinyanghuo (MBZ-YYH) herb pair inhibits rheumatoid arthritis (RA) cell proliferation and glycolysis, promising results with an obscure mechanism of action. Methods Therefore, it is necessary to explore the main components of MBZ-YYH and unravel the potential mechanism in RA based on network pharmacology and molecular docking methods. Components and targets of MBZ-YYH were retrieved from the TCMSP. Relevant targets of RA were searched in GeneCards, therapeutic target database (TTD), and DisGeNET databases; the common targets of the MBZ-YYH compounds and RA were obtained by comparison; and a component-target interaction network was established by Cytoscape 3.9.1. Gene ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analysis were performed through the David database. Molecular docking was performed by PyMoL2.3.0 and AutoDock Vina1.1.2 software. Results 7 active ingredients and 58 putatively identified target genes were screened from MBZ, and 16 effective components of YYH and 230 potential targets were identified. There were 29 mutual targets between the two herbs and RA. Through the PPI network, 9 hub targets which contain JUN, CASP3, PPARG, PTGS2, GSK3B, CASP8, HMOX1, ICAM1, and HK2 were screened out. GO term enrichment analysis indicated that positive regulation of the apoptotic process, response to drugs, and response to hypoxia were significantly enriched. Based on KEGG analysis, it was mainly associated with the IL-17 signaling pathway, the TNF signaling pathway, and the p53 signaling pathway. The docking analysis revealed that the effective components showed strong binding activity with the receptors. Conclusion The effects of the MBZ-YYH herb pair on RA were coordinated by the interaction of diverse components, which may be through the IL-17 signaling pathway and the TNF signaling pathway, which target GSK3B, HK2, caspase 3, and caspase 8, inhibiting the proliferation and glycolysis of rheumatoid arthritis fibroblast-like synovial cells (RA-FLS) and tending towards an increasing efficacy and decreasing toxicity effect on RA.
Collapse
Affiliation(s)
- Fuxue Meng
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Xiaomai Tao
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Longkuan Li
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Wei Jia
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Xin Yang
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Yuchen Yang
- The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| |
Collapse
|
14
|
Liu X, Huang M, Wang L, Li J, Wu W, Wang Q. Network pharmacology and experimental validation methods to reveal the active compounds and hub targets of Curculigo orchioides Gaertn in rheumatoid arthritis. J Orthop Surg Res 2023; 18:861. [PMID: 37957674 PMCID: PMC10644664 DOI: 10.1186/s13018-023-04352-w] [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: 09/25/2023] [Accepted: 11/07/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease that can lead to joint destruction and deformity. Curculigo orchioides Gaertn (CO) was previously revealed to play a significant role in RA treatment. However, the main active ingredients and molecular mechanisms of CO in regulating RA are still unclear. METHODS The active ingredients of CO were obtained from the Traditional Chinese Medicine Systems Pharmacology database and published literature. The targets corresponding to these compounds and the targets linked to RA were collected from public databases. The "ingredient-target" and "protein-protein interaction" networks were constructed to screen the main active ingredients and hub targets of CO in the treatment of RA. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment assays were used to elucidate the potential pharmacological mechanism of CO in RA. Molecular docking was performed to detect the binding between the main active ingredients and hub targets. Collagen-induced arthritis rats were used to validate the hub targets of CO against RA. RESULTS Network pharmacological topology analysis showed that caffeine, 2,4-dichloro-5-methoxy-3-methylphenol, curculigoside, orcinol glucoside, and orcin were the main active ingredients of CO, and matrix metalloproteinase 9 (MMP9), transcription factor AP-1 (JUN), prostaglandin-endoperoxide synthase 2 (PTGS2), brain-derived neurotrophic factor, and receptor-type tyrosine-protein phosphatase C were the hub targets of CO for RA treatment. Molecular docking revealed that curculigoside and orcinol glucoside had effective binding potential with MMP9, JUN, and PTGS2, respectively. In vivo experiments demonstrated that CO alleviated RA symptoms and inhibited the expression of MMP9, JUN, and PTGS2 proteins. CONCLUSIONS Our study demonstrates the main active ingredients and potential targets of CO against RA, laying an experimental foundation for the development and application of CO as an anti-RA drug.
Collapse
Affiliation(s)
- Xia Liu
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi Qizhi Road, Chongqing, 400021, China
| | - Mingchun Huang
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi Qizhi Road, Chongqing, 400021, China
| | - Lijuan Wang
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi Qizhi Road, Chongqing, 400021, China
| | - Jie Li
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi Qizhi Road, Chongqing, 400021, China
| | - Weihui Wu
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi Qizhi Road, Chongqing, 400021, China.
| | - Qin Wang
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi Qizhi Road, Chongqing, 400021, China.
| |
Collapse
|
15
|
Huo CL, Wang B, Zhang X, Sun ZG. Skimmianine attenuates liver ischemia/reperfusion injury by regulating PI3K-AKT signaling pathway-mediated inflammation, apoptosis and oxidative stress. Sci Rep 2023; 13:18232. [PMID: 37880319 PMCID: PMC10600244 DOI: 10.1038/s41598-023-45354-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 10/18/2023] [Indexed: 10/27/2023] Open
Abstract
Liver ischemia/reperfusion (I/R) injury is a common injury after liver transplantation and hepatectomy. Skimmianine (Ski) has antibacterial, antiviral pharmacological effects. However, it is not clear whether Ski has a protective effect against liver I/R injury. In the present study, we established a mouse liver I/R model and an AML12 cell hypoxia-reoxygenation (H/R) model, both pretreated with different concentrations of Ski. Serum transaminase levels, necrotic liver area, cell viability, inflammatory factors, oxidative stress and apoptosis-related levels were measured to assess the protective effect of Ski against liver I/R injury. Western blotting was used to detect apoptosis-related proteins and PI3K-AKT pathway-related proteins. Mice and cells were also treated with PI3K inhibitor LY294002 to assess changes in indicators of liver injury. The results showed that Ski significantly reduced transaminase levels, liver necrosis area, oxidative stress, and apoptosis levels in mice with I/R. Ski also inhibited cell injury and apoptosis after H/R. Moreover, Ski activated phosphorylation of PI3K-AKT pathway-related proteins after liver I/R and cell H/R. Importantly, the PI3K inhibitor LY294002 effectively reversed the alleviation of I/R injury caused by Ski. These results confirm that Ski exerts a protective effect against liver I/R injury through activation of the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Cheng-Long Huo
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26, Chuyuan Avenue, Jingzhou District, Jingzhou, Hubei, China
| | - Bing Wang
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26, Chuyuan Avenue, Jingzhou District, Jingzhou, Hubei, China
| | - Xuewen Zhang
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26, Chuyuan Avenue, Jingzhou District, Jingzhou, Hubei, China
| | - Zhen-Gang Sun
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26, Chuyuan Avenue, Jingzhou District, Jingzhou, Hubei, China.
| |
Collapse
|
16
|
Hu Y, Liu T, Zheng G, Zhou L, Ma K, Xiong X, Zheng C, Li J, Zhu Y, Bian W, Zheng X, Xiong Q, Lin J. Mechanism exploration of 6-Gingerol in the treatment of atherosclerosis based on network pharmacology, molecular docking and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154835. [PMID: 37121058 DOI: 10.1016/j.phymed.2023.154835] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND The 6-Gingerol has significant anti-inflammatory, anti-oxidative and hypolipidemic activities and is widely used for treating cardiac-cerebral vascular diseases. However, the multi-target mechanism of 6-Gingerol in the treatment of atherosclerosis remains to be elucidated. METHODS Firstly, the therapeutic actions of 6-Gingerol anti-atherosclerosis were researched based on an atherosclerotic ApoE-deficient mice model induced by high-fat feed. Then, network pharmacology and molecular docking were employed to reveal the anti-atherogenic mechanism of 6-Gingerol. Finally, the target for these predictions was validated by target protein expression assay in vitro and in vivo experiments and further correlation analysis. RESULTS Firstly, 6-Gingerol possessed obvious anti-atherogenic activity, which was manifested by a significant reduction in the plaque area, decrease in the atherosclerosis index and vulnerability index. Secondly, based on network pharmacology, 14 predicted intersection target genes between the targets of 6-Gingerol and atherogenic-related targets were identified. The key core targets of 6-Gingerol anti-atherosclerosis were found to be TP53, RELA, BAX, BCL2, and CASP3. Lipid and atherosclerosis pathways might play a critical role in 6-Gingerol anti-atherosclerosis. Molecular docking results also further revealed that the 6-Gingerol bound well and stable to key core targets from network pharmacological predictions. Then, the experimental results in vivo and in vitro verified that the up-regulation of TP53, RELA, BAX, CASP3, and down-regulation of BCL2 from atherosclerotic ApoE-deficient mice model can be improved by 6-Gingerol intervention. Meanwhile, the correlation analysis further confirmed that 6-Gingerol anti-atherosclerosis was closely related to these targets. CONCLUSION The 6-Gingerol can markedly improve atherosclerosis by modulating key multi-targets TP53, RELA, BAX, CASP3, and BCL2 in lipid and atherosclerosis pathways. These novel findings shed light on the anti-atherosclerosis mechanism of 6-Gingerol from the perspective of multiple targets and pathways.
Collapse
Affiliation(s)
- Youdong Hu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Tingting Liu
- Department of Gynecology, Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, 223002, China
| | - Guangzhen Zheng
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Li Zhou
- Department of Intensive Care Unit, Dazhou Central Hospital, Dazhou, 635000, Sichuan, China
| | - Ke Ma
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Xiaolian Xiong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Cheng Zheng
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jin Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yong Zhu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Wenhui Bian
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Xiangde Zheng
- Department of Intensive Care Unit, Dazhou Central Hospital, Dazhou, 635000, Sichuan, China.
| | - Qingping Xiong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China.
| | - Jiafeng Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
17
|
Zhang L, Li H, Zhang L, Zu Z, Xu D, Zhang J. Network Pharmacology Analysis of the Mechanisms Underlying the Therapeutic Effects of Yangjing Zhongyu Tang on Thin Endometrium. Drug Des Devel Ther 2023; 17:1805-1818. [PMID: 37350984 PMCID: PMC10284302 DOI: 10.2147/dddt.s409659] [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] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/12/2023] [Indexed: 06/24/2023] Open
Abstract
Purpose Yangjing Zhongyu Tang (YJZYT) is a classic Chinese prescription for infertility treatment and exerts therapeutic effects via activity on the thin endometrium (TE). However, the major components and underlying mechanisms of YJZYT actions remain to be established. The main objectives of this study were to clarify the effects of YJZYT on the TE and provide insights into the related mechanisms based on network pharmacology and molecular docking analyses. Methods Network pharmacology was employed to explore the main bioactive components and targets of YJZYT. TE-related genes were obtained from the Genecards database and screened for intersections with YJZYT. The Cytoscape 3.8.2 was used to build a "compounds-disease-targets" network and molecular docking analysis performed on key targets. The mechanism of action of YJZYT was further validated in vivo using a rat model. Results A total of 98 YJZYT active ingredients, 2409 thin endometrium-associated genes, and 186 common targets were obtained. Through topological analysis, 10 core objectives were screened. Data from the PPI network suggest that AKT1, TNF, VEGFA, IL-6, TP53, INS, ESR1, MMP9, ALB, and ACTB serve as key targets in the action of YJZYT on TE. PI3K-Akt, TNF, apoptosis, IL-17 and MAPK were established as the main functional pathways. Molecular docking analysis revealed high affinity of the active ingredients of YJZYT, specifically, ursolic acid, palbinone, stigmasterol, and beta-sitosterol, for TNF, VEGFA, IL-6, AKT, and MMP9. YJZYT improved endometrial recovery, promoted endometrial angiogenesis, and upregulated protein expression of VEGF, PI3K, AKT, and p-AKT in the TE rat model. Conclusion Network pharmacological and animal studies facilitated the prediction and validation of the active components and key targets of YJZYT potentially contributing to TE. Preliminary evidence from in vivo experiments showed that YJZYT promotes angiogenesis and thin endometrial repair via regulation of the PI3K/AKT pathway, providing a reference for further research.
Collapse
Affiliation(s)
- Lei Zhang
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Honglin Li
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Liang Zhang
- Gynecology, Obstetrics and Reproductive Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Zhihui Zu
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Dinglin Xu
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Jianwei Zhang
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
- Gynecology, Obstetrics and Reproductive Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| |
Collapse
|
18
|
Wang L, Li P, Zhou Y, Gu R, Lu G, Zhang C. Magnoflorine Ameliorates Collagen-Induced Arthritis by Suppressing the Inflammation Response via the NF-κB/MAPK Signaling Pathways. J Inflamm Res 2023; 16:2271-2296. [PMID: 37265745 PMCID: PMC10231344 DOI: 10.2147/jir.s406298] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/18/2023] [Indexed: 06/03/2023] Open
Abstract
Objective Magnoflorine (Mag) has been reported to have anxiolytics, anti-cancer, and anti-inflammatory properties. In this study, we aim to investigate the effects of Mag on the rheumatoid arthritis (RA) and explore the underlying mechanism using a collagen-induced arthritis (CIA) mouse model and a lipopolysaccharide (LPS)-stimulated macrophage inflammation model. Methods The in vivo effects of Mag on CIA were studied by inducing CIA in a mouse model using DBA/1J mice followed by treatment with vehicle, methotrexate (MTX, 1 mg/kg/d), and Mag (5 mg/kg/d, 10 mg/kg/d, and 20 mg/kg/d), and the in vitro effects of Mag on macrophages were examined by stimulation of RAW264.7 cells line and peritoneal macrophages (PMs) by LPS in the presence of different concentrations of Mag. Network pharmacology and molecular docking was then performed to predict the the binding ability between Mag and its targets. Inflammatory mediators were assayed by quantitative real-time PCR and enzyme linked immunosorbent assay (ELISA). Signaling pathway changes were subsequently determined by Western blotting and immunohistochemistry (IHC). Results In vivo experiments demonstrated that Mag decreased arthritis severity scores, joints destruction, and macrophages infiltration into the synovial tissues of the CIA mice. Network pharmacology analysis revealed that Mag interacted with TNF-α, IL-6, IL-1β, and MCP-1. Consistent with this, analysis of the serum, synovial tissue of the CIA mice, and the supernatant of the cultured RAW264.7 cells and PMs showed that Mag suppressed the expression of TNF-α, IL-6, IL-1β, MCP-1, iNOS, and IFN-β. Furthermore, Mag attenuated the phosphorylation of p65, IκBα, ERK, JNK, and p38 MAPKs in the synovial tissues of the CIA mice and LPS-stimulated RAW 264.7 cells. Conclusion Mag may exert anti-arthritic and anti-inflammatory effects by inhibiting the activation of NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Lei Wang
- College of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Pengfei Li
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Yu Zhou
- College of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Renjun Gu
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Ge Lu
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Chunbing Zhang
- College of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
19
|
Yu YX, Wang S, Liu ZN, Zhang X, Hu ZX, Dong HJ, Lu XY, Zheng JB, Cui HJ. Traditional Chinese medicine in the era of immune checkpoint inhibitor: theory, development, and future directions. Chin Med 2023; 18:59. [PMID: 37210537 DOI: 10.1186/s13020-023-00751-7] [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: 12/06/2022] [Accepted: 04/13/2023] [Indexed: 05/22/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer management and have been widely applied; however, they still have some limitations in terms of efficacy and toxicity. There are multiple treatment regimens in Traditional Chinese Medicine (TCM) that play active roles in combination with Western medicine in the field of oncology treatment. TCM with ICIs works by regulating the tumor microenvironment and modulating gut microbiota. Through multiple targets and multiple means, TCM enhances the efficacy of ICIs, reverses resistance, and effectively prevents and treats ICI-related adverse events based on basic and clinical studies. However, there have been few conclusions on this topic. This review summarizes the development of TCM in cancer treatment, the mechanisms underlying the combination of TCM and ICIs, existing studies, ongoing trials, and prospects for future development.
Collapse
Affiliation(s)
- Yi-Xuan Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Shuo Wang
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Zhe-Ning Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Xu Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Zi-Xin Hu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Hui-Jing Dong
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Xing-Yu Lu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Jia-Bin Zheng
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China.
| | - Hui-Juan Cui
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
20
|
Duan Z, Jin C, Deng Y, Liu J, Gu C, Wang J, Cai X, Li S, Zhou Y. Exploring the chondroprotective effect of Chaenomeles speciosa on Glucose-6-Phosphate Isomerase model mice using an integrated approach of network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116553. [PMID: 37178981 DOI: 10.1016/j.jep.2023.116553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) has been used in China for a long time and is gradually gaining more and more recognition worldwide. Chaenomeles speciosa (CSP) (Chinese Pinyin: mugua) is a medicinal and food herb that has long been used as a folk medicine for rheumatic diseases, yet its bioactive components and therapeutic mechanisms are not clear. AIM OF THE STUDY Exploring anti-inflammatory and chondroprotective effects of CSP on rheumatoid arthritis (RA) and its possible targets of action. MATERIALS AND METHODS In this study, we performed an integrated approach of network pharmacology, molecular docking and experimental studies to explore the potential mechanism of action of CSP in the treatment of cartilage damage in RA. RESULTS Studies have shown that Quercetin, ent-Epicatechin and Mairin may be the main active compounds of CSP in the treatment of RA, while AKT1, VEGFA, IL-1β, IL-6, MMP9 etc. are considered as core target proteins to which the main active compounds in CSP bind, as further confirmed by molecular docking. In addition, the potential molecular mechanism of CSP for the treatment of cartilage damage in RA predicted by network pharmacology analysis was validated by in vivo experiments. CSP was found to downregulate the expression of AKT1, VEGFA, IL-1β, IL-6, MMP9, ICAM1, VCAM1, MMP3, MMP13 and TNF-α and increase the expression of COL-2 in the joint tissue of Glucose-6-Phosphate Isomerase (G6PI) model mice. Thus CSP contributes to the treatment of rheumatoid arthritis cartilage destruction. CONCLUSION This study showed that CSP has multi-component, multi-target and multi-pathway characteristics in treating cartilage damage in RA, which can achieve the effect of treating RA by inhibiting the expression of inflammatory factors, reducing neovascularization and alleviating the damage to cartilage caused by the diffusion of synovial vascular opacities, and reducing the degradation of cartilage by MMPs to play a protective role in RA cartilage damage. In conclusion, this study indicates that CSP is a candidate Chinese medicine for further research in treating cartilage damage in RA.
Collapse
Affiliation(s)
- Zhihao Duan
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Can Jin
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Ying Deng
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Jinlang Liu
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Chengyi Gu
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Jie Wang
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Xiangquan Cai
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Shigang Li
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China.
| | - You Zhou
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China.
| |
Collapse
|
21
|
Zong K, Xu K, Zhang X, Wang P, Wang Z, Yang S, Li H, Ke H, He S, Hu Y, Go Y, Chan XHF, Wu J, Huang Q. Explorating the mechanism of Huangqin Tang against skin lipid accumulation through network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116581. [PMID: 37142143 DOI: 10.1016/j.jep.2023.116581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/26/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqin Tang (HQT), a famous prescription with the effect of clearing pathogenic heat and detoxifying, was first recorded in "Treatise on Typhoid and Miscellaneous Diseases". It has proved that HQT has good anti-inflammatory and antioxidant effects and can improve acne symptoms clinically. However, the study on the regulation of HQT on sebum secretion which is one of the inducements of acne is not enough. AIM OF THE STUDY This paper aimed to investigate the mechanisms of HQT in the treatment of skin lipid accumulation by network pharmacology and validating the results via in vitro experiments. MATERIALS AND METHODS Network pharmacology was employed to predict the potential targets of HQT against sebum accumulation. Then, the palmitic acid (PA)-induced SZ95 cell model was established to evaluate the effect of HQT on lipid accumulation and anti-inflammation, and the core pathways predicted by network pharmacology were verified in cell studies. RESULTS 336 chemical compounds and 368 targets in HQT were obtained by network pharmacology, of which 65 targets were related to sebum synthesis. 12 core genes were revealed by protein-protein interaction (PPI) network analysis. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment results suggested that AMP-activated protein kinase (AMPK) signaling pathway might play a crucial role in regulating lipogenesis. In vitro experiments, HQT suppressed lipid accumulation, downregulated the expressions of sterol-regulatory element binding protein-1 (SREBP-1) and fatty acid synthase (FAS), and upregulated AMPK phosphorylation. Furthermore, AMPK inhibitor reversed HQT-mediated sebosuppressive effect. CONCLUSION The results disclosed that HQT ameliorates lipogenesis in PA-induced SZ95 sebocytes partially through the AMPK signaling pathway.
Collapse
Affiliation(s)
- Kaile Zong
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Kewei Xu
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Xingjiang Zhang
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Pan Wang
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Zhekun Wang
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Shan Yang
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Huijuan Li
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Hui Ke
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Shengsheng He
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Yunwei Hu
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China
| | - Yuyo Go
- Royal Victoria Hospital, 274 Grosvenor Rd, Belfast, BT12 6BA, United Kingdom
| | | | - Jianxin Wu
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China.
| | - Qing Huang
- Skin Health and Cosmetic Development & Evaluation Laboratory, China Pharmaceutical University, No.639 Longmian Lane, Nanjing, 211198, China.
| |
Collapse
|
22
|
Chen J, Liu H, Chen Y, Hu H, Huang C, Wang Y, Liang L, Liu Y. Iridium(III) complexes inhibit the proliferation and migration of BEL-7402 cells through the PI3K/AKT/mTOR signaling pathway. J Inorg Biochem 2023; 241:112145. [PMID: 36709684 DOI: 10.1016/j.jinorgbio.2023.112145] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023]
Abstract
Iridium(III) complexes are largely studied as anti-cancer complexes due to their excellent anti-cancer activity. In this article, two new iridium(III) complexes [Ir(piq)2(THPIP)]PF6 (THPIP = 2,4-di-tert-butyl-6-(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)phenol, piq = deprotonated 1-phenylisoquinoline) (Ir1) and [Ir(bzq)2(THPIP)]PF6 (bzq = deprotonated benzo[h]quinolone) (Ir2) were synthesized. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays showed that complex Ir1 exhibits moderate activity (IC50 = 29.9 ± 4.6 μM) and Ir2 shows high cytotoxicity (IC50 = 9.8 ± 1.8 μM) against BEL-7402 cells. Further studies on the mechanism showed that Ir1 and Ir2 induced apoptosis by changing the mitochondrial membrane potential, Ca2+ release, ROS accumulation, and cell cycle arrest at the S phase. The complexes can effectively inhibit cell colony formation and migration. The expression of B-cell lymphoma-2 (Bcl-2) family proteins, PI3K (phosphatidylinositol 3-kinase), AKT (protein kinase B), mTOR (mammalian target of rapamycin), and p-mTOR was studied by immunoblotting. Complexes Ir1 and Ir2 downregulated the expression of anti-apoptotic protein Bcl-2 and increased the expression of autophagy-related proteins of Beclin-1 and LC3-II. Further experiments showed that the complexes inhibited the production of glutathione (GSH) and increased the amounts of malondialdehyde (MDA). Fluorescence of HMGB1 was significantly increased. We also investigated the effect of the complexes on the expression of genes using RNA-sequence analysis, we further calculated the lowest binding energies between the complexes and proteins using molecular docking. Taken together, the above results indicated that complexes Ir1 and Ir2 induce apoptosis in BEL-7402 cells through a ROS-mediated mitochondrial dysfunction and inhibition of the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jing Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Haimei Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yichuan Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huiyan Hu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Chunxia Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yi Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
23
|
Sun L, Wang Z, Yang Z, Liu X, Dong H. Virtual screening and structure-activity relationship study of novel BTK inhibitors in Traditional Chinese Medicine for the treatment of rheumatoid arthritis. J Biomol Struct Dyn 2023; 41:15219-15233. [PMID: 36914235 DOI: 10.1080/07391102.2023.2188418] [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: 10/31/2022] [Accepted: 02/26/2023] [Indexed: 03/14/2023]
Abstract
Bruton tyrosine kinase (BTK) is a known drug target for the treatment of autoimmune diseases, including rheumatoid arthritis (RA). In this study, a series of 1-amino-1H-imidazole-5-carboxamide derivatives with good inhibitory activity against BTK were selected to explore the structure-activity relationships of these BTK inhibitors (BTKIs). Furthermore, we concentrated on 182 prescriptions of Traditional Chinese Medicine with therapeutic effects on RA. 54 herbs with a frequency of ≥10 were counted to establish a database containing 4027 ingredients for virtual screening. Five compounds with relatively higher docking scores and better absorption, distribution, metabolism, elimination and toxicity (ADMET) parameters were then selected for higher precision docking. The results demonstrated that the potentially active molecules form hydrogen bond interactions with the hinge region residues Met477, Glu475, glycine-rich P-loop residue Val416, Lys430 and DFG motif Asp539. In particular, they also interact with the key residues Thr474 and Cys481 of BTK. The molecular dynamics (MD) results demonstrated that all five compounds above could bind with BTK stably as its cognate ligand in dynamic conditions. This work identified several potential BTKIs using a computer-aided drug design approach and may provide crucial information for developing novel BTKIs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Lili Sun
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zixiao Wang
- Department of Pharmacy, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhigang Yang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - XiuJuan Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Haiyan Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
24
|
Li X, Hou R, Ding H, Gao X, Wei Z, Qi T, Fang L. Mollugin ameliorates murine allergic airway inflammation by inhibiting Th2 response and M2 macrophage activation. Eur J Pharmacol 2023; 946:175630. [PMID: 36871665 DOI: 10.1016/j.ejphar.2023.175630] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/19/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Mollugin, isolated from Rubia cordifolia L, is a pharmacological compound with anti-inflammatory activity. This study aimed to investigate whether mollugin protects mice against shrimp tropomyosin (ST)-induced allergic airway inflammation. Mice were sensitized with ST combined with Al(OH)3 administered intraperitoneally (i.p.) once weekly for 3 wk followed by ST challenge for 5 d. Mice were i.p.-administered daily with mollugin for 7 d. Results showed that mollugin attenuated ST-induced infiltration of eosinophils and epithelial mucus secretion in the lung tissues and suppressed lung eosinophil peroxidase activity. Additionally, mollugin lowered the Th2 cytokine, IL-4 and IL-5, production and downregulated the mRNA levels of Il-4, Il-5, Il-13, eotaxin, Ccl-17, Muc5ac, arginase-1, Ym-1, and Fizz-1 in the lung tissues. Network pharmacology was employed to predict core targets, and the molecular docking approach was used to verify the compound targets. The results of the molecular docking study of mollugin into p38 MAPK or poly(ADP-ribose) polymerase 1 (PARP1) binding sites revealed that its mechanism was possibly similar to that of SB203580 (a p38 MAPK inhibitor) or olaparib (a PARP1 inhibitor). Immunohistochemistry analysis revealed that mollugin mitigated ST-induced elevation of arginase-1 expression and macrophage levels in the lungs and bronchoalveolar lavage fluid, respectively. Furthermore, arginase-1 mRNA level and phosphorylation of p38 MAPK were inhibited in IL-4-stimulated peritoneal macrophages. In ST-stimulated mouse primary splenocytes, mollugin notably inhibited IL-4 and IL-5 production and downregulated PARP1 and PAR protein expression. According to our findings, mollugin ameliorated allergic airway inflammation by inhibiting Th2 response and macrophage polarization.
Collapse
Affiliation(s)
- Xiuru Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Ruitao Hou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Hao Ding
- Baoying People's Hospital, Yangzhou, 225800, China; Baoying Maternal and Child Health Hospital, Yangzhou, 225800, China
| | - Xiang Gao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Zichen Wei
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Ting Qi
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Lei Fang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, Yangzhou, 225009, China.
| |
Collapse
|
25
|
Zeng W, Fang Y, Mo S, Shen C, Yang H, Luo G, Xiao L, Zhan R, Yan P. The Underling Mechanisms Exploration of Rubia cordifolia L. Extract Against Rheumatoid Arthritis by Integrating Network Pharmacology and Metabolomics. Drug Des Devel Ther 2023; 17:439-457. [PMID: 36818604 PMCID: PMC9930591 DOI: 10.2147/dddt.s388932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/02/2023] [Indexed: 02/13/2023] Open
Abstract
Purpose Rubia cordifolia L. (RC) is a classic herbal medicine for the treatment of rheumatoid arthritis (RA) and has been used since ancient times. The ethanol extract of Rubia cordifolia L. (RCE) showed obvious anti-RA effects in our previous study. However, further potential mechanisms require more exploration. We aimed to investigate the mechanism of RCE for the treatment of RA by integrating metabolomics and network pharmacology in this study. Methods An adjuvant-induced arthritis (AIA) rat model was established, and we evaluated the therapeutic effects of RCE. Metabolomics of serum and urine was used to identify the differential metabolites. Network pharmacology was applied to determine the key metabolites and potential targets. Finally, the potential targets and compounds of RCE were verified by molecular docking. Results The results indicated that RCE suppressed foot swelling and alleviated joint damage and also had anti-inflammatory properties by inhibiting the expressions of tumor necrosis factor (TNF)-α, Interleukin (IL)-1β, prostaglandin E2 (PGE2), and P65. Ten and seven differential metabolites were found in the serum and urine, respectively, of rats. Six key targets, ie, phospholipase A2 group IIA (PLA2G2A), phospholipase A2 group X (PLA2G10), cytidine deaminase (CDA), uridine-cytidine kinase 2 (UCK2), charcot-leyden crystal galectin (CLC), and 5',3'-nucleotidase, mitochondrial (NT5M), were discovered by network pharmacology and metabolite analysis and were found to be related to glycerophospholipid metabolism and pyrimidine metabolism. Molecular docking confirmed that the favorable compounds showed affinities with the key targets, including alizarin, 6-hydroxyrubiadin, ruberythric acid, and munjistin. Conclusion This study revealed the underlying mechanisms of RCE and provided evidence that will allow researchers to further investigate the functions and components of RCE against RA.
Collapse
Affiliation(s)
- Weiya Zeng
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Yuan Fang
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Suifen Mo
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Caihong Shen
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Huiling Yang
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Guihua Luo
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Luhua Xiao
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Ruoting Zhan
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China,Correspondence: Ruoting Zhan; Ping Yan, Guangzhou University of Chinese Medicine, No. 232, Outer Ring East Road, Guangzhou, Guangdong, People’s Republic of China, Tel/Fax +86 20-39358045, Email ;
| | - Ping Yan
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| |
Collapse
|
26
|
Li J, Liu X, Li J, Han D, Li Y, Ge P. Mechanism of andrographis paniculata on lung cancer by network pharmacology and molecular docking. Technol Health Care 2023:THC220698. [PMID: 36641698 DOI: 10.3233/thc-220698] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Traditional Chinese medicine (TCM) has been widely recognized and accepted worldwide to provide favorable therapeutic effects for cancer patients. As Andrographis paniculata has an anti-tumor effect, it might inhibit lung cancer. OBJECTIVE The drug targets and related pathways involved in the action of Andrographis paniculata against lung cancer were predicted using network pharmacology, and its mechanism was further explored at the molecular level. METHODS This work selected the effective components and targets of Andrographis paniculata against the Traditional Chinese Medicine System Pharmacology (TCMSP) database. Targets related to lung cancer were searched for in the GEO database (accession number GSE136043). The volcanic and thermal maps of differential expression genes were produced using the software R. Then, the target genes were analyzed by GO and KEGG analysis using the software R. This also utilized the AutoDock tool to study the molecular docking of the active component structures downloaded from the PubChem database and the key target structures downloaded from the PDB database, and the docking results were visualized using the software PyMol. RESULTS The results of molecular docking show that wogonin, Mono-O-methylwightin, Deoxycamptothecine, andrographidine F_qt, Quercetin tetramethyl (3',4',5,7) ether, 14-deoxyandrographolide, andrographolide-19-β-D-glucoside_qt and 14-deoxy-11-oxo-andrographolide were potential active components, while AKT1, MAPK14, RELA and NCOA1 were key targets. CONCLUSION This study showed the main candidate components, targets, and pathways involved in the action of Andrographis paniculata against lung cancer.
Collapse
Affiliation(s)
- Jiaxin Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xiaonan Liu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiaxin Li
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Dongwei Han
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yu Li
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Pengling Ge
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
27
|
王 远, 戴 兴, 陈 春, 程 培, 陈 松, 黎 欣, 毕 炫, 程 瑶, 吴 昌, 吴 宁. [ Sidaxue, a traditional Guizhou Miao herbal medicine formula, reduces necroptosis and synovial vascular proliferation in rats with collagen-induced arthritis by inhibiting the RIPK1/RIPK3/MLKL pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1774-1782. [PMID: 36651244 PMCID: PMC9878417 DOI: 10.12122/j.issn.1673-4254.2022.12.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To explore the inhibitory effect of Sidaxue (SX), a traditional Guizhou Miao herbal medicine formula, on necrotic apoptosis and synovial angiogenesis in rats with collagen-induced arthritis (CIA) and the role of the RIPK1/RIPK3/MLKL pathway in mediating this effect. METHODS Forty-two SD rats were randomized into 6 groups (n=7), including a normal control group, a CIA model group, 3 SX treatment groups at low (10 g/kg), moderate (20 g/kg) and high (40 g/kg) doses, and a GTW treatment group. CIA rat models were established by subcutaneous injections of bovine type II collagen, and the treatments were administered daily by gavage for 21 days. The rats were observed for swelling of the hind limb joints, which was rated using the arthritis index (AI) score on a weekly basis. Serum levels of TNF-α, IL-1β and IL-17 in the rats were detected using ELISA, and the pathological changes in the synovium were observed with HE staining. Real-time PCR was performed to detect the mRNA expression levels of VEGF, MMP-9, Ang-1, RIPK1, RIPK3, and caspase-8 in the synovial tissues, and the protein expressions of VEGF, MMP9, Ang-1, Stat-3, RIPK1, RIPK3, MLKLl, p-MLKL and caspase-8 were detected using Western blotting. RESULTS Compared with those in CIA model group, the rats receiving treatment with GTW and SX showed milder swelling of the hind limb joints with significantly lower AI scores (P < 0.05). In CIA model group, a large number of inflammatory cells were observed in the synovium with obvious damages of the tissue structure. In the drug treatment groups, inflammatory cell infiltration, synovial angiogenesis and synovial hyperplasia were alleviated, and the therapeutic effects were obviously enhanced as SX dose increased. Compared with those in the model group, the rats treated with GTW and high-dose SX showed significantly decreased serum levels of IL-1β, IL-17 and TNF-α (P < 0.05), lower mRNA and protein expressions of RIPK1, RIPK3, VEGF, Ang-1, and MMP9 (P < 0.05), higher expressions of caspase-8 (P < 0.01), and obviously lowered expression of Stat-3 protein and phosphorylation level of MLKL (P < 0.05). CONCLUSION SX can improve synovial angiogenesis in CIA rats possibly by inhibiting the activation of RIP1/RIP3/MLKL signaling pathway and down-regulating the expression of the vascular growth factors VEGF, Ang-1, MMP9, and Stat-3.
Collapse
Affiliation(s)
- 远迎 王
- 贵州医科大学基础医学院,贵州 贵阳 550004College of Basic Medical Science, Guizhou Medical University, Guiyang 55004, China
| | - 兴月 戴
- 贵州医科大学麻醉医学院,贵州 贵阳 550004College of Anesthesia Medicine, Guizhou Medical University, Guiyang 55004, China
| | - 春彦 陈
- 贵州医科大学基础医学院,贵州 贵阳 550004College of Basic Medical Science, Guizhou Medical University, Guiyang 55004, China
| | - 培钧 程
- 贵州医科大学基础医学院,贵州 贵阳 550004College of Basic Medical Science, Guizhou Medical University, Guiyang 55004, China
| | - 松江 陈
- 贵州医科大学临床医学院,贵州 贵阳 550004College of Clinical Medicine, Guizhou Medical University, Guiyang 55004, China
| | - 欣悦 黎
- 贵州医科大学基础医学院,贵州 贵阳 550004College of Basic Medical Science, Guizhou Medical University, Guiyang 55004, China
| | - 炫 毕
- 贵州医科大学基础医学院,贵州 贵阳 550004College of Basic Medical Science, Guizhou Medical University, Guiyang 55004, China
| | - 瑶 程
- 贵州医科大学基础医学院,贵州 贵阳 550004College of Basic Medical Science, Guizhou Medical University, Guiyang 55004, China
| | - 昌学 吴
- 贵州医科大学医学分子生物学重点实验室,贵州 贵阳 550004Key Laboratory of Molecular Biology, Guizhou Medical University, Guiyang 55004, China
| | - 宁 吴
- 贵州医科大学基础医学院,贵州 贵阳 550004College of Basic Medical Science, Guizhou Medical University, Guiyang 55004, China
| |
Collapse
|