Abstract

Background: Wutou Decoction (WTD), as a classic prescription, has been generally used to treat rheumatoid arthritis (RA) for two thousand years in China. However, the potential protective effects of WTD on rheumatoid arthritis and its possible mechanism have rarely been reported. Purpose: The aim of this study was to explore the possible mechanism of WTD against RA and a promising alternative candidate for RA therapy. Methods: A model of collagen-induced arthritis (CIA) was constructed in rats to assess the therapeutic effects of WTD. Histopathological staining, immunofluorescence, and western blotting of synovial sections were conducted to detect the antiangiogenic effects of WTD. Then, cell viability assays, flow cytometry, scratch healing assays, and invasion assays were conducted to explore the effects of WTD on MH7A human fibroblast-like synoviocyte (FLS) cell proliferation, apoptosis, migration, and invasion in vitro. The ability of WTD to induce blood vessel formation after MH7A cell and human umbilical vein endothelial cell line (HUVEC) coculture with WTD intervention was detected by a tube formation assay. The mechanisms of WTD were screened by network pharmacology and confirmed by in vivo and in vitro experiments. Results: WTD ameliorated the symptoms and synovial pannus hyperplasia of CIA rats. Treatment with WTD inhibited MH7A cell proliferation, migration, and invasion and promoted MH7A apoptosis. WTD could inhibit MH7A cell expression of proangiogenic factors, including VEGF and ANGI, to induce HUVEC tube formation. Furthermore, the PI3K-AKT-mTOR-HIF-1α pathway was enriched as a potential target of WTD for the treatment of RA through network pharmacology enrichment analysis. Finally, it was confirmed in vitro and in vivo that WTD inhibits angiogenesis in RA by interrupting the PI3K-AKT-mTOR-HIF-1α pathway. Conclusion: WTD can inhibit synovial hyperplasia and angiogenesis, presumably by inhibiting the migration and invasion of MH7A cells and blocking the production of proangiogenic effectors in MH7A cells. The possible underlying mechanism by which WTD ameliorates angiogenesis in RA is the PI3K-AKT-mTOR-HIF-1α pathway.

Highlights

  • Rheumatoid arthritis (RA) is a chronic autoimmune disease with the characteristic pathological changes of persistent synovitis, hyperplastic synovial pannus tissue formation, the destruction of cartilage and bone, and the presence of autoantibodies (Mcinnes and Schett, 2011)

  • The PI3K-AKT-mTOR-HIF-1α pathway was enriched as a potential target of Wutou Decoction (WTD) for the treatment of RA through network pharmacology enrichment analysis

  • The relative content of each substance in WTD was calculated and the detection of nine compounds in different wavelength ranges were better shown by 3D High-Performance Liquid Chromatography (HPLC) fingerprint (Figure 1C)

Read more

Summary

Introduction

Rheumatoid arthritis (RA) is a chronic autoimmune disease with the characteristic pathological changes of persistent synovitis, hyperplastic synovial pannus tissue formation, the destruction of cartilage and bone, and the presence of autoantibodies (especially rheumatoid factor and citrullinated peptide) (Mcinnes and Schett, 2011). In recent decades, diseasemodifying antirheumatic drugs (DMARDs) have been the predominant therapeutic agents for RA. Biological agents and small molecule targeted drugs are used when RA cannot be controlled or toxic side effects arise with DMARD treatment (Smolen et al, 2017; Sparks et al, 2021). Biological targeted DMARDs are expensive and may increase the risk of infection (Burmester and Pope, 2017). More than 30% of patients still cannot satisfactorily control this disease after using the drugs mentioned above (Singh et al, 2015; Favalli et al, 2017; Aletaha and Smolen, 2018). The potential protective effects of WTD on rheumatoid arthritis and its possible mechanism have rarely been reported. Purpose: The aim of this study was to explore the possible mechanism of WTD against RA and a promising alternative candidate for RA therapy

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call