ORIGINAL RESEARCH article
Front. Immunol.
Sec. Autoimmune and Autoinflammatory Disorders : Autoimmune Disorders
Volume 16 - 2025 | doi: 10.3389/fimmu.2025.1608262
This article is part of the Research TopicIntestinal microenvironment and autoimmune diseasesView all 6 articles
Integrative multi-omics analysis reveals the interaction mechanisms between gut microbiota metabolites and ferroptosis in rheumatoid arthritis
Provisionally accepted- 1The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- 2Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- 3The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- 4State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guanghzou, Guangdong, China
- 5Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- 6Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, Guangdong Province, China
Select one of your emails
You have multiple emails registered with Frontiers:
Notify me on publication
Please enter your email address:
If you already have an account, please login
You don't have a Frontiers account ? You can register here
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic synovitis and joint destruction. To systematically investigate the regulatory relationship between key ferroptosis genes and gut metabolites in RA, this study employed an integrative multi-omics approach combined with machine learning algorithms and single-cell transcriptomic data, identifying and validating GPX3 and MYC as potential critical ferroptosis regulators in RA. First, 16 candidate genes were obtained by intersecting WGCNA, differential expression analysis results, and targets related to ferroptosis and gut microbiota. Following cross-validation with machine learning approaches including LASSO, SVM, and RFE-RF, GPX3 and MYC were ultimately identified as crucial genes. GSVA and GSEA analyses revealed that high expression of GPX3 and MYC was enriched in interferon response and TNFA signaling pathways, while their low expression was associated with fatty acid metabolism and oxidative phosphorylation pathways. Further single-cell RNA sequencing analysis demonstrated that MYC was expressed in multiple immune cell types, particularly in CD4+ T cells and NK cells. Ferroptosis scoring for CD8+ T cells and subsequent cell communication analysis revealed stronger interactions between CD8+ T cells with higher ferroptosis scores and other immune cells through IFN-II and CCL signaling, further intensifying the activation of the inflammatory microenvironment. Additionally, molecular docking analysis of GPX3 and MYC with the gut metabolites Diosgenin and Differentiation-inducing factor 3 (DIF-3) respectively showed that the GPX3-Diosgenin complex had the lowest binding energy, and a 100 ns molecular dynamics simulation was performed on this complex. Results showed good stability of the complex across indicators such as RMSD, RMSF, SASA, and radius of gyration, suggesting that Diosgenin may intervene in ferroptosis and inflammatory injury in RA by binding to and modulating GPX3 function. In summary, this study elucidated the multifaceted mechanisms of GPX3 and MYC in RA pathogenesis and preliminarily validated the potential role of gut metabolites in mediating ferroptosis regulation, offering novel theoretical foundations and potential strategies for diagnostic biomarker screening and targeted therapy in RA.
Keywords: gutMGene, Rheumatoid arthritis, Gut Microbiota, ferroptosis, GPX3, MYC
Received: 08 Apr 2025; Accepted: 23 Jun 2025.
Copyright: © 2025 Liang, Liang, He, Zhang and Ke. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
* Correspondence:
Lifang Liang, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
Peifeng Ke, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.