International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Copyright: © Cui et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is a serious extra-articular manifestation of RA that significantly increases morbidity and mortality. Epidemiological studies indicate that 10-40% of patients with RA develop ILD, with mortality rates up to 3-fold higher than those with RA without pulmonary involvement (1,2). RA-ILD is marked by progressive pulmonary fibrosis and inflammation, ultimately leading to respiratory failure and reduced survival. It accounts for 10-20% of RA-related deaths (3,4). The high prevalence and poor prognosis highlight the urgent need for improved understanding of its pathogenesis and more effective management strategies.
The pathogenesis of RA-ILD remains poorly understood. Diagnostic delays are common due to nonspecific early symptoms and the absence of validated screening protocols, which often result in treatment initiation only after irreversible lung damage has occurred (5-7). Current evidence points to a multifactorial etiology involving genetic susceptibility (for example, HLA-DRB1 alleles), environmental exposures (for example, smoking), and autoimmune dysregulation (8,9). Molecular mechanisms driving RA-ILD include abnormal immune activation (for example, TNF-α and IL-6), fibroblast proliferation and dysregulated extracellular matrix deposition-pathways that parallel those in idiopathic pulmonary fibrosis (IPF) (10-12). Emerging evidence implicates neutrophil extracellular traps (NETs), YKL-40 and KL-6 in lung injury, while signaling pathways such as AKT/TMEM175 and JAK-STAT may contribute to fibrosis progression (13-16). However, the exact molecular drivers linking RA to ILD remain unclear, and currently available biomarkers (for example, KL-6 and YKL-40) lack sufficient specificity for broad clinical application (13). Management primarily relies on immunosuppressive agents (for example, methotrexate and rituximab) and antifibrotic therapies (for example, nintedanib and pirfenidone), but their effectiveness is limited by heterogeneous patient responses and the lack of RA-ILD-specific targeted treatments (12,17). For instance, antifibrotic agents have demonstrated benefit in IPF, their effectiveness in RA-ILD has been inconsistent, likely due to distinct underlying molecular pathways (18). These challenges highlight the urgent need for precision medicine strategies tailored to the unique pathogenesis of RA-ILD (19,20).
Elucidating the molecular mechanisms underlying RA-ILD is crucial for the discovery of novel therapeutic targets. Emerging evidence implicates that dysregulated immune pathways (such as IL-36 and NETs), aberrant fibroblast activation and interactions between the gut and lungs play a role in disease progression (11,21). For example, targeting NETs may help mitigate lung injury, while modulation of the gut microbiota could attenuate systemic inflammation (9,16). Despite these advances, our understanding of RA-ILD pathogenesis remains incomplete, limiting the development of effective targeted therapies. The present study aimed to identify novel molecular mechanisms that could inform the design of disease-modifying strategies for RA-ILD.
The present study was conducted as part of a multi-center cohort that includes participants from Beijing Haidian Hospital and Beijing Shunyi Hospital. The study population consisted of three groups: (i) Patients diagnosed with RA-ILD, (ii) patients with RA but without ILD and (iii) healthy individuals. All patients with RA met the 2010 American College of Rheumatology/European League Against Rheumatism classification criteria for RA (22), while patients with RA-ILD also met the diagnostic criteria for ILD established by the American Thoracic Society/European Respiratory Society (23). Health controls were selected from individuals without inflammatory or rheumatic diseases. All participants underwent pulmonary function tests and chest high-resolution computed tomography (HRCT). Patients with RA but without ILD and healthy controls showing pulmonary symptoms or abnormal HRCT findings were excluded to ensure specificity. The study protocol was approved by the Research Ethics Committees of Beijing Haidian Hospital (approval no. 2024-006; Beijing, China) and Beijing Shunyi Hospital (approval no. 2023k-021; Beijing, China). Written informed consent was obtained from all participants prior to enrollment. Blood samples were collected for further analysis.
Serum proteins were analyzed in three cohorts: Healthy controls (n=7), RA (n=40, with 10 subjects pooled), and RA-ILD (n=40, with 10 subjects pooled). The clinical characteristics of the participants are summarized in Table I. Serum cytokine profiling was performed using the Human Cytokine Antibody Array (cat. no. GSH-CAA-440; RayBiotech, Inc.), a high-throughput platform with 11 non-overlapping arrays for simultaneous detection of 440 cytokines. Peripheral blood serum samples were diluted 1:2 with blocking buffer and incubated overnight in array chambers coated with cytokine-specific capture antibodies. After washing away unbound proteins, a biotin-conjugated anti-cytokine antibody cocktail was added to form antibody-cytokine-antibody sandwich complexes. Cy3-conjugated streptavidin was then applied to amplify fluorescent signals via biotin-streptavidin binding. All incubation steps were conducted with 100 µl of reagents per well. Fluorescence intensity was measured with an InnoScan 300 Microarray Scanner (Innopsys) at optimized 532 nm excitation/emission wavelengths for Cy3. Raw signal values were normalized against internal positive and negative controls for assay reproducibility.
Insulin and IL-31 were validated by ELISAs using commercially available kits (Human Insulin ELISA Kit; cat. no. ELH-Insulin; Human IL-31 ELISA Kit; cat. no. ELH-IL31; RayBiotech, Inc.) with an expanded cohort consisting of 64 patients with RA-ILD, 64 patients with RA and 40 healthy controls (demographics provided in Table II). Briefly, serum samples were incubated in antibody-precoated wells overnight at 4˚C. Following washing steps, biotin-conjugated detection antibodies were added and incubated for 2 h. Horseradish peroxidase-conjugated streptavidin was then applied to bind the biotinylated complexes for 45 min. The enzymatic reaction was developed using tetramethylbenzidine substrate form 30 min. After the reaction was stopped, absorbance was measured at 450 nm using an ELx800NB microplate reader (BioTek; Agilent Technologies, Inc.).
Statistical comparisons between experimental groups were conducted using a one-way ANOVA followed by Bonferroni's post hoc test in SPSS version 20.0 (IBM Corp.). Differences were considered statistically significant when meeting two criteria: (i) P<0.05; (ii) a fold change threshold of <0.83 (downregulation) or >1.2 (upregulation). Continuous data were expressed as the mean ± standard deviation (SD). Correlation analyses between serum biomarkers (IL-31 and insulin) and clinical parameters of RA-ILD severity were performed using GraphPad Prism 9.0 (GraphPad Software; Dotmatics). Pearson's correlation coefficient (r) was calculated to assess linear relationships between biomarkers and clinical parameters. P<0.05 was considered to indicate a statistically significant difference.
A panel of 20 proteins demonstrated sequential upregulation, significantly elevated in patients with RA compared with healthy controls (P<0.05), and showing even greater increases in patients with RA-ILD compared with patients with RA (P<0.05) (Fig. 1). These proteins included carbonic anhydrase IX (CA9), IL-1F9, ectodysplasin A2 (EDA-A2), growth arrest-specific gene 1 (Gas1), IL-2Rb, IL-7, amphiregulin (AR), sonic hedgehog N-terminal (Shh-N), IGF-1R, MMP-7, insulin, Eotaxin, EpCAM, tissue factor pathway inhibitor (TFPI), MIP-3b, CRTAM, IL-31, tissue factor (TF), syndecan-1 and cathepsin L. The information of these 20 proteins is summarized in Table III.
To assess the discriminatory value of the 20 proteins, hierarchical clustering and principal component analysis (PCA) were performed. Using normalized expression data from the pooled analysis, hierarchical clustering of these proteins unveiled unique expression patterns and demonstrated 100% accuracy in classifying RA-ILD from RA and the healthy groups (Fig. 2). PCA effectively segregated RA, RA-ILD, and healthy controls into separate clusters within the reduced-dimensional space (Fig. 3).
Gene Ontology (GO) enrichment analysis revealed critical biological processes associated with the 20 proteins, including T cell differentiation, T cell activation, lymphocyte differentiation, mononuclear cell proliferation, leukocyte proliferation and positive regulation of cytokine production. Further analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway implicated two key pathways cytokine-cytokine receptor interaction and the PI3K-Akt signaling pathway (Fig. 4).
PPI network analysis identified insulin as the highest-degree hub protein (node degree=6), showing direct interactions with multiple profibrotic and immunomodulatory mediators: IGF-1R, Shh-N, AR, TF, IL-7 and EpCAM (Fig. 5). This indicates its central role in coordinating molecular crosstalk within the RA-ILD proteomic landscape.
The independent validation of insulin and IL-31 with an expanded cohort by ELISA demonstrated strong concordance with the initial antibody array data (Fig. 6). The diagnostic potential of these two biomarkers showed excellent discriminatory capacity with area under the curve values of >75% (Fig. 7). Notably, the correlation analysis revealed a strong positive correlation between these two biomarkers and the Scleroderma Lung Study I score of HRCT images (Fig. 8, r>0.6). This finding suggests that insulin and IL-31 may play a role in the disease activity of RA-ILD.
RA-ILD is a severe extra-articular manifestation of RA, characterized by progressive pulmonary fibrosis and high morbidity. Its etiology is multifactorial and complex, involving genetic predisposition (for example, MUC5B promoter variants), autoimmune-driven inflammation, aberrant fibroblast activation, and environmental exposures such as smoking. The interplay of these factors leads to dysregulated immune responses and irreversible lung remodeling (10). However, the precise molecular mechanisms triggering these immune responses remain elusive, limiting the development of effective targeted therapies for RA-ILD. In the present study, 20 proteins including CA9, IL-1F9, EDA-A2, Gas1, IL-2Rb, IL-7, AR, Shh-N, IGF-1R, MMP-7, insulin, Eotaxin, EpCAM, TFPI, MIP-3b, CRTAM, IL-31, TF, Syndecan-1 and Cathepsin L were identified, which were significantly elevated in patients with RA compared with healthy controls, with further upregulation in RA-ILD relative to RA alone. The stepwise increase in these proteins suggests their contribution to RA pathogenesis and progression to ILD, the most common and serious pulmonary complication of RA.
GO analysis revealed that these 20 proteins were enriched in immune-related processes, including T cell differentiation, T cell activation and cytokine production. These findings align with prior studies implicating aberrant T cell responses and cytokine dysregulation in RA-ILD pathogenesis (24-26). KEGG pathway analysis highlighted the cytokine-cytokine receptor interaction pathway and PI3K-Akt signaling pathway, both of which play key roles in immune cell proliferation and differentiation in ILD (27). The PI3K-Akt pathway is known to drive fibroblast activation and fibrosis in ILD (28). Moreover, cytokine-receptor interactions -such as IL-7/IL-7R, IGF-1R/insulin-may perpetuate chronic inflammation and fibrogenesis (29,30). Notably, Wu et al (31) reported that the AhR/IGF1R axis contributes to the development of IPF through activation of the TGF-β/Smad/STAT signaling cascade. In the present study, KEGG pathway analysis revealed that IGF-1R and insulin were involved in the PI3K-Akt signaling pathway, suggesting a potential role for the insulin/IGF-1R axis in the progression of RA to ILD through this pathway. It was hypothesized that the insulin/IGF1R axis could be a promising therapeutic target for the treatment of RA-ILD. PPI analysis revealed that insulin is a central hub protein that interacts with IGF-1R, Shh-N, AR, TF, IL-7 and EpCAM. These interactions suggest that insulin and its network partners may collectively regulate immune and metabolic pathways in RA-ILD, potentially exacerbating disease progression. While no previous studies have directly linked insulin to RA-ILD, its network prominence and functional interactions generate a strong interest in investigating the modulation of the insulin pathway as a potential therapeutic strategy. Further validation is needed to confirm the potential involvement of insulin in RA-ILD.
Furthermore, among these 20 proteins, several have well-established roles in RA or pulmonary fibrosis. For example, IL-7 promotes T cell survival and Th17 differentiation (29), and its elevation has been linked to the exacerbation of IPF (32). MMP-7 serves as a predictive biomarker of disease progression and mediates extracellular matrix remodeling in IPF (33). IL-1F9 acts as a proinflammatory cytokine in lung disease by enhancing chemokine production and inflammatory cell recruitment (34). Eotaxin is associated with increased pulmonary infiltration of eosinophils and neutrophils, as well as the production of profibrotic cytokines contributing to pulmonary fibrosis (35). Both cathepsin L and tissue factor have been implicated in the pathogenesis of IPF and ILD (36,37). AR is elevated in inflammatory lung disease associated with RA (38), and EpCAM, TFPI and MIP-3b are upregulated in IPF (39-41). Shh-N promotes pulmonary fibrosis through the hedgehog signaling pathway (42), and Syndecan-1 shedding exacerbates the transition from inflammation to fibrosis by releasing heparan sulfate-bound growth factors (43).
Notably, CA9, EDA-A2, Gas1, CRTAM, IL-2Rb and IL-31 have emerged as novel candidates, with no prior studies linking them to RA-ILD or fibrotic diseases. However, chronic inflammation is a well-established precursor to fibrotic tissue remodeling (44), suggesting that these proteins may contribute indirectly to fibrosis through sustained inflammatory signaling. Among these novel targets, EDA-A2 activates the inflammatory responses through NF-κB signaling by binding to the EDA receptor (45). CRTAM promotes STAT signaling via STAT1 phosphorylation (46). IL-2Rb contributes to immune microenvironment disorder by disrupting the Th1/Th2 cell differentiation balance (47), and IL-31 drives inflammation primarily through JAK-STAT pathway activation (48). Gas1 may play a critical role in fibrotic diseases, including RA-ILD through the Gas1/Axl axis, analogous to the pro-fibrotic Gas6/Axl signaling pathway (49,50). In addition, CA9, a hypoxia-inducible protein, may promote hypoxic-associated fibrosis (51). These findings indicate the potential of these novel candidates to drive RA-ILD progression through their roles in inflammatory and hypoxic signaling, warranting further investigation.
However, the present study has several limitations. First, the cohort included a relatively small number of healthy controls (n=7), which limited the statistical power to detect biologically relevant differences between healthy individuals and patient groups. Second, the use of pooled samples for the antibody array analysis-implemented due to cost constraints associated with high-throughput proteomic screening-represents a significant methodological limitation. Third, the present study only validated insulin (the highest-degree hub protein) and IL-31 (a novel candidate) using independent methods due to budget limitations. Fourth, the functional roles of the identified protein signatures were not experimentally validated. Future studies should prioritize individual sample analysis, larger cohort sizes, and rigorous validation of all these candidate proteins as potential biomarkers. In addition, in vitro and in vivo studies are warranted to elucidate the functional relevance of these proteins, particularly those with novel associations, and to explore their potential as therapeutic targets for RA-ILD.
In conclusion, our study highlights the complex molecular interplay underlying RA-ILD pathogenesis. A total of 20 dysregulated proteins that collectively drive immune dysregulation and fibrotic progression were identified. These proteins are functionally enriched in critical pathways, including T cell differentiation, cytokine-cytokine receptor interactions (such as IL-7/IL-7R and IGF-1R/Insulin), and the PI3K-Akt signaling pathway. The PPI network identifies insulin as a central hub, interacting with multiple profibrotic mediators (IGF-1R, Shh-N and AR) and immune modulators (IL-7, EpCAM), suggesting its pivotal role in orchestrating profibrotic and inflammatory responses in RA-ILD. While several proteins (MMP-7, IL-1F9 and Cathepsin L) have established roles in pulmonary fibrosis, our identification of novel candidates points to additional mechanisms involving sustained inflammatory signaling and hypoxia-responsive pathways. These findings provide new insight into RA-ILD pathogenesis and suggest that targeting the insulin/IGF1R-PI3K-Akt axis may represent a promising therapeutic strategy to disrupt the immune-fibrotic cascade in this disease. Further validation and mechanistic studies are warranted to explore these potential targets.
Not applicable.
Funding: The present study was supported by the 2024 Haidian Health Development Research, Cultivation Plan Project (grant no. HP2024-30-101004) and the Beijing Shunyi District Hospital Research and Development Special Fund (grant no. 2025Y01).
The data generated in the present study may be found in zenodo database under accession number 15852072 or at the following URL: (https://zenodo.org/records/15852072).
WC conducted all experiments and wrote the first draft of the manuscript. YZ and QY contributed to sample collection and processing. BY and QY conducted statistical analyses. GZ contributed to conception and design of the present study, and revised the manuscript. WC and GZ confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
Approval was obtained from the Research Ethics Committees of Beijing Haidian Hospital (approval no. 2024-006; Beijing, China) and Beijing Shunyi Hospital (approval no. 2023-k-021; Beijing, China). All participants provided informed consent to participate in the study.
Not applicable.
The authors declare that they have no competing interests.
|
Zhang M, Yin J and Zhang X: Factors associated with interstitial lung disease in patients with rheumatoid arthritis: A systematic review and meta-analysis. PLoS One. 18(e0286191)2023.PubMed/NCBI View Article : Google Scholar | |
|
Farquhar HJ, Beckert L, Edwards AL, Matteson EL, Frampton CMA, Ganly E, Yetton R, Thiessen R, Haslett J, Bucknall D and Stamp LK: Rheumatoid interstitial lung disease in Canterbury, Aotearoa New Zealand-A retrospective cohort study. Semin Arthritis Rheum. 64(152359)2024.PubMed/NCBI View Article : Google Scholar | |
|
Wang HF, Wang YY, Li ZY, He PJ, Liu S and Li QS: The prevalence and risk factors of rheumatoid arthritis-associated interstitial lung disease: A systematic review and meta-analysis. Ann Med. 56(2332406)2024.PubMed/NCBI View Article : Google Scholar | |
|
Juge PA, Wemeau L, Ottaviani S, Desjeux G, Zhuo J, Vannier-Moreau V, Flipo RM, Crestani B and Dieudé P: Increased mortality in patients with RA-associated interstitial lung disease: Data from a French administrative healthcare database. RMD Open. 9(e003491)2023.PubMed/NCBI View Article : Google Scholar | |
|
Sullivan DI and Ascherman DP: Rheumatoid arthritis-associated interstitial lung disease (RA-ILD): Update on prevalence, risk factors, pathogenesis, and therapy. Curr Rheumatol Rep. 26:431–449. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Narváez J: Moving forward in Rheumatoid arthritis-associated interstitial lung disease screening. J Clin Med. 13(5385)2024.PubMed/NCBI View Article : Google Scholar | |
|
Koduri G and Solomon JJ: Identification, monitoring, and management of rheumatoid arthritis-associated interstitial lung disease. Arthritis Rheumatol. 75:2067–2077. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Liu M, Jiang Z, Liu M, Ni H, Li Y, Fang J, Du Q and Dong Y: SLAMF1 as a novel molecule mediating the causal association between rheumatoid arthritis and interstitial lung disease: A Mendelian randomization study combined with transcriptomics and in vivo validation. Int Immunopharmacol. 142 (Pt A)(113082)2024.PubMed/NCBI View Article : Google Scholar | |
|
Zhong X, Wang X, Xu L, Zhang J, Yu W, Ji L, Huang J, Zhong X, Zhang J and Long L: Alterations in gut microbiota in Rheumatoid arthritis patients with interstitial lung Disease: A Comparative study. Hum Immunol. 86(111239)2025.PubMed/NCBI View Article : Google Scholar | |
|
Kim Y, Yang HI and Kim KS: Etiology and pathogenesis of rheumatoid arthritis-interstitial lung disease. Int J Mol Sci. 24(14509)2023.PubMed/NCBI View Article : Google Scholar | |
|
Zheng W, Hu X, Zou M, Hu N, Song W, Wang R, Liu Y, Hou Q, Liu Y, Chen X and Cheng Z: Plasma IL-36α and IL-36γ as potential biomarkers in interstitial lung disease associated with rheumatoid arthritis: A pilot study in the Chinese population. Inflammation. 46:285–296. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Bes C, Köybaşı G, İçaçan OC, Yalçın Mutlu M and Yıldırım F: Antifibrotic therapies in rheumatoid arthritis associated interstitial lung disease. Eur J Rheumatol. 9:176–179. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Liang B, Zhang Y, Ke D, Yan R, Jiang MN, Li L, Zhang LX, Zhao XG, Yuan GP, Xu B and Liu XM: Serum YKL-40 and Serum Krebs von den lungen-6 as potential predictive biomarkers for rheumatoid arthritis-associated interstitial lung disease. Immunol Invest. 53:989–1000. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Liu N, Fan X, Shao Y, Chen S, Wang T, Yao T and Chen X: Resveratrol attenuates inflammation and fibrosis in rheumatoid arthritis-associated interstitial lung disease via the AKT/TMEM175 pathway. J Transl Med. 22(457)2024.PubMed/NCBI View Article : Google Scholar | |
|
Liu H, Yang Y, Zhang J and Li X: Baricitinib improves pulmonary fibrosis in mice with rheumatoid arthritis-associated interstitial lung disease by inhibiting the Jak2/Stat3 signaling pathway. Adv Rheumatol. 63(45)2023.PubMed/NCBI View Article : Google Scholar | |
|
Venetsanopoulou AI, Ntinopoulou M, Papagianni E, Koletsos N, Voulgari PV and Chrysanthopoulou A: Neutrophil extracellular traps as immunofibrotic mediators in RA-ILD; pilot evaluation of the nintedanib therapy. Front Immunol. 15(1480594)2024.PubMed/NCBI View Article : Google Scholar | |
|
Shoda T, Kotani T, Mitsuhiro K, Yoshikawa A, Wada Y, Makino H, Osuga K and Takeuchi T: The therapeutic efficacy of abatacept for rheumatoid arthritis-associated interstitial lung disease: Insights from a 12-month trial using semi-quantitative chest high-resolution computed tomography imaging. J Clin Med. 13(5871)2024.PubMed/NCBI View Article : Google Scholar | |
|
Wang S, Liu M, Li X, Zhang J, Wang F, Zhang C, Roden A, Ryu JH, Warrington KJ, Sun J, et al: Canonical and noncanonical regulatory roles for JAK2 in the pathogenesis of rheumatoid arthritis-associated interstitial lung disease and idiopathic pulmonary fibrosis. FASEB J. 36(e22336)2022.PubMed/NCBI View Article : Google Scholar | |
|
Rodríguez Portal JA, Brito García N, Díaz Del Campo Fontecha P, Valenzuela C, Ortiz AM, Nieto MA, Mena-Vázquez N, Cano-Jiménez E, Castellví I, Aburto M, et al: SER-SEPAR recommendations for the management of rheumatoid arthritis-related interstitial lung disease. Part 1: Epidemiology, risk factors and prognosis. Reumatol Clin (Engl Ed). 18:443–452. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Albrecht K, Strangfeld A, Marschall U and Callhoff J: Interstitial lung disease in rheumatoid arthritis: Incidence, prevalence and related drug prescriptions between 2007 and 2020. RMD Open. 9(e002777)2023.PubMed/NCBI View Article : Google Scholar | |
|
Huang Z, Wu T, Lu R, Zhou H, Zhang Y, Huang L, Gan Y and He H: Prevalence and clinical significance of anti-neutrophil cytoplasmic antibodies in rheumatoid arthritis-associated interstitial lung disease. BMC Pulm Med. 25(177)2025.PubMed/NCBI View Article : Google Scholar | |
|
Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO III, Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD, et al: 2010 Rheumatoid arthritis classification criteria: An American college of rheumatology/European league against rheumatism collaborative initiative. Arthritis Rheum. 62:2569–2581. 2010.PubMed/NCBI View Article : Google Scholar | |
|
Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, Behr J, Cottin V, Danoff SK, Morell F, et al: Diagnosis of idiopathic pulmonary fibrosis. An Official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med. 198:e44–e68. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Kono M: New insights into the metabolism of Th17 cells. Immunol Med. 46:15–24. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Zhang Y, Zhu J, Xiao K, Liu H, Du K, Wu D and Zou Q: Single-cell sequencing of PBMC characterizes the transformation of T cell subsets in the inflammatory microenvironment of RA-ILD. Research Square: https://doi.org/10.21203/rs.3.rs-3990097/v1. | |
|
Jeong E, Hong H, Lee YA and Kim KS: Potential rheumatoid arthritis-associated interstitial lung disease treatment and computational approach for future drug development. Int J Mol Sci. 25(2682)2024.PubMed/NCBI View Article : Google Scholar | |
|
Liu C and Ge Y: Immune-related genes associated with interstitial lung disease in dermatomyositis. Int J Gen Med. 17:5261–5271. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Yang Z, Han R, Yin H, Li J, Cao Y, Guo R, Sheng Y, Song L and Zhang Y: Mechanism of Lycopodii herba for RA-ILD using integrated metabolomics and network pharmacology. Anal Biochem. 648(114679)2022.PubMed/NCBI View Article : Google Scholar | |
|
Wang C, Kong L, Kim S, Lee S, Oh S, Jo S, Jang I and Kim TD: The role of IL-7 and IL-7R in cancer pathophysiology and immunotherapy. Int J Mol Sci. 23(10412)2022.PubMed/NCBI View Article : Google Scholar | |
|
Knuever J, Willenborg S, Ding X, Akyüz MD, Partridge L, Niessen CM, Brüning JC and Eming SA: Myeloid cell-restricted insulin/IGF-1 receptor deficiency protects against skin inflammation. J Immunol. 195:5296–5308. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Wu SM, Tsai JJ, Pan HC, Arbiser JL, Elia L and Sheu ML: Aggravation of pulmonary fibrosis after knocking down the aryl hydrocarbon receptor in the insulin-like growth factor 1 receptor pathway. Br J Pharmacol. 179:3430–3451. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Liu Z, Peng Z, Lin H, Zhou K, Liang L, Cao J, Huang Z and Mei J: Identifying potential drug targets for idiopathic pulmonary fibrosis: A mendelian randomization study based on the druggable genes. Respir Res. 25(217)2024.PubMed/NCBI View Article : Google Scholar | |
|
Bauer Y, White ES, de Bernard S, Cornelisse P, Leconte I, Morganti A, Roux S and Nayler O: MMP-7 is a predictive biomarker of disease progression in patients with idiopathic pulmonary fibrosis. ERJ Open Res. 3:00074–2016. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Ramadas RA, Ewart SL, Medoff BD and LeVine AM: Interleukin-1 family member 9 stimulates chemokine production and neutrophil influx in mouse lungs. Am J Respir Cell Mol Biol. 44:134–145. 2010.PubMed/NCBI View Article : Google Scholar | |
|
Huaux F, Gharaee-Kermani M, Liu T, Morel V, McGarry B, Ullenbruch M, Kunkel SL, Wang J, Xing Z and Phan SH: Role of Eotaxin-1 (CCL11) and CC chemokine receptor 3 (CCR3) in bleomycin-induced lung injury and fibrosis. Am J Pathol. 167:1485–1496. 2005.PubMed/NCBI View Article : Google Scholar | |
|
Yuan L, Zou C, Ge W, Liu Y, Hu B, Wang J, Lin B, Li Y and Ma E: A novel cathepsin L inhibitor prevents the progression of idiopathic pulmonary fibrosis. Bioorg Chem. 94(103417)2020.PubMed/NCBI View Article : Google Scholar | |
|
Novelli F, Neri T, Tavanti L, Armani C, Noce C, Falaschi F, Bartoli ML, Martino F, Palla A, Celi A and Paggiaro P: Procoagulant, tissue factor-bearing microparticles in bronchoalveolar lavage of interstitial lung disease patients: An observational study. PLoS One. 9(e95013)2014.PubMed/NCBI View Article : Google Scholar | |
|
Poole JA, Thiele GM, Janike K, Nelson AJ, Duryee MJ, Rentfro K, England BR, Romberger DJ, Carrington JM, Wang D, et al: Combined collagen-induced arthritis and organic dust-induced airway inflammation to model inflammatory lung disease in rheumatoid arthritis. J Bone Miner Res. 34:1733–1743. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Schuliga M, Kanwal A, Read J, Blokland KEC, Burgess JK, Prêle CM, Mutsaers SE, Grainge C, Thomson C, James A, et al: A cGAS-dependent response links DNA damage and senescence in alveolar epithelial cells: A potential drug target in IPF. Am J Physiol Lung Cell Mol Physiol. 321:L859–L871. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Cella G, Cipriani A, Tommasini A, Rampin E, Sbarai A, Rocconi R, Mazzaro G and Luzzatto G: Tissue factor pathway inhibitor (TFPI) antigen plasma level in patients with interstitial lung disease before and after heparin administration. Semin Thromb Hemost. 23:45–49. 1997.PubMed/NCBI View Article : Google Scholar | |
|
Russo RC and Ryffel B: The chemokine system as a key regulator of pulmonary fibrosis: Converging pathways in human idiopathic pulmonary fibrosis (IPF) and the bleomycin-induced lung fibrosis model in mice. Cells. 13(2058)2024.PubMed/NCBI View Article : Google Scholar | |
|
He CH, Lv JM, Khan GJ, Duan H, Wang W, Zhai KF, Zou GA and Aisa HA: Total flavonoid extract from Dracocephalum moldavica L. improves pulmonary fibrosis by reducing inflammation and inhibiting the hedgehog signaling pathway. Phytother Res. 37:2745–2758. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Feng F, Wang LJ, Li JC, Chen TT and Liu L: Role of heparanase in ARDS through autophagy and exosome pathway (review). Front Pharmacol. 14(1200782)2023.PubMed/NCBI View Article : Google Scholar | |
|
Li Y, Zhao J, Yin Y, Li K, Zhang C and Zheng Y: The role of IL-6 in fibrotic diseases: Molecular and cellular mechanisms. Int J Biol Sci. 18:5405–5414. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Cai Z, Deng X, Jia J, Wang D and Yuan G: Ectodysplasin A/Ectodysplasin A receptor system and their roles in multiple diseases. Front Physiol. 12(788411)2021.PubMed/NCBI View Article : Google Scholar | |
|
Zheng S, Yang B, Li L, Chen M, Zhang L, Chi W, Shao ZM, Xiu B, Chi Y and Wu J: RTAM promotes antitumor immune response in triple negative breast cancer by enhancing CD8+ T cell infiltration. Int Immunopharmacol. 129(111625)2024.PubMed/NCBI View Article : Google Scholar | |
|
Liao Y, Ke B, Long X, Xu J and Wu Y: Upregulated expression of IL2RB causes disorder of immune microenvironment in patients with Kawasaki disease. Biomed Res Int. 2022(2114699)2022.PubMed/NCBI View Article : Google Scholar | |
|
Pastor Bandeira I, de Almeida Franzoi AE, Murillo Wollmann G, de Medeiros Junior WLG, Nogueira Brandão W, Schatzmann Peron JP, Becker J, Nascimento OJM and Magno Gonçalves MV: Interleukin-31 and soluble CD40L: New candidate serum biomarkers that predict therapeutic response in multiple sclerosis. Neurol Sci. 43:6271–6278. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Chen CC, Chen CY, Yeh CT, Liu YT, Leu YL, Chuang WY, Shih YH, Chou LF, Shieh TM and Wang TH: Corylin attenuates CCl4-induced liver fibrosis in mice by regulating the GAS6/AXL signaling pathway in hepatic stellate cells. Int J Mol Sci. 24(16936)2023.PubMed/NCBI View Article : Google Scholar | |
|
Fiebeler A, Park JK, Muller DN, Lindschau C, Mengel M, Merkel S, Banas B, Luft FC and Haller H: Growth arrest specific protein 6/Axl signaling in human inflammatory renal diseases. Am J Kidney Dis. 43:286–295. 2004.PubMed/NCBI View Article : Google Scholar | |
|
Cao Z, Li J, Hu W, Xu J, Zhao F, Wang Y, Qin S, Liu M, Wang P, Duan J, et al: Near-infrared imaging agent ABSi-148 alleviates CA IX-mediated hypoxic fibrosis in inflammation-cancer transition. Adv Healthc Mater. 18(e2404935)2025.PubMed/NCBI View Article : Google Scholar |