Next Article in Journal
Sex and APOE ε2 Interactive Effects on the Longitudinal Change in Cognition in a Population-Based Cohort of Older Adults with Vascular Risk Factors
Previous Article in Journal
Topical Ruxolitinib and Delgocitinib Versus Systemic JAK Inhibitors: A Comparative Safety Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Rheumatoid Arthritis: Biomarkers and the Latest Breakthroughs

Sutton Arthritis Research Laboratory, The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(21), 10594; https://doi.org/10.3390/ijms262110594
Submission received: 28 September 2025 / Revised: 22 October 2025 / Accepted: 25 October 2025 / Published: 30 October 2025
(This article belongs to the Section Molecular Biology)

Abstract

Rheumatoid arthritis (RA) is a heterogeneous autoimmune disease characterized by variable clinical manifestations and a complex, often unpredictable disease trajectory, which hinders early diagnosis and personalized treatment. This review highlights recent breakthroughs in biomarker discovery, emphasizing the transformative impact of multi-omics technologies and deep profiling of the synovial microenvironment. Advances in genomics and transcriptomics have identified key genetic variants and expression signatures associated with disease susceptibility, progression, and therapeutic response. Complementary insights from proteomics and metabolomics have elucidated dynamic molecular patterns linked to inflammation and joint destruction. Concurrently, microbiome research has positioned gut microbiota as a compelling source of non-invasive biomarkers with both diagnostic and immunomodulatory relevance. The integration of these diverse data modalities through advanced bioinformatics platforms enables the construction of comprehensive biomarker panels, offering a multidimensional molecular portrait of RA. When coupled with synovial tissue profiling, these approaches facilitate the identification of spatially resolved biomarkers essential for localized disease assessment and precision therapeutics. These innovations are transforming RA care by enabling earlier detection, improved disease monitoring, and personalized treatment strategies that aim to optimize patient outcomes.

1. Introduction

Rheumatoid arthritis (RA) is an autoimmune disease characterized by persistent inflammation, ongoing joint damage, and various systemic manifestations. Worldwide, RA affects 0.5% to 1% of the population, with a noticeably higher prevalence in women [1]. Although the precise cause of RA remains only partly understood, current evidence suggests a multifactorial interplay of genetic predisposition, environmental factors, and hormonal influences in the onset and progression of the disease [2].
RA is broadly divided into seropositive and seronegative categories, based on the presence or absence of autoantibodies, primarily rheumatoid factor (RF) and anti-citrullinated protein antibodies (ACPA). These subtypes have different genetic backgrounds, clinical features, immunopathological profiles, radiographic progressions, and prognoses [3]. Seropositive RA, identified by detectable RF and/or ACPA, benefits from well-established diagnostic and prognostic biomarkers. Conversely, seronegative RA, which accounts for about 15% to 25% of cases, presents a significant diagnostic and treatment challenge due to the lack of these serological markers.
RA remains incurable but is clinically manageable with conventional synthetic and biologic disease-modifying antirheumatic drugs (cs/bDMARDs). Despite therapeutic advances, up to 40% of patients exhibit suboptimal responses or develop pharmacologic resistance, leading to refractory RA [4]. Long-term DMARD use, especially methotrexate, is linked to significant adverse effects, including gastrointestinal, pulmonary, and hematologic toxicity [5].
Early therapeutic intervention, ideally within 3 to 6 months of symptom onset, has been shown to improve remission rates and long-term outcomes [6]. The preclinical phase of RA, characterized by silent autoimmunity, can precede overt symptoms by several years. Its heterogeneity and lack of predictive tools for early diagnosis and treatment selection hinder timely intervention, increasing the risk of irreversible joint damage. There is a critical need for robust biomarkers to enable early diagnosis and inform personalized DMARD selection, especially in refractory RA. This review explores both established and novel biomarkers, highlighting the role of advanced technologies in identifying potential biomarkers in precision medicine for managing RA.

2. Current Biomarkers in RA

A biomarker is a measurable biological characteristic that indicates a normal or abnormal process, condition, or disease. In RA, current biomarkers predominantly include protein-based, genetic, and epigenetic signatures, most of which are detected in peripheral blood. Furthermore, imaging biomarkers obtained from modalities like ultrasound and magnetic resonance imaging (MRI) provide crucial structural and functional insights. These biomarkers support diagnostic and prognostic assessments, enable prediction of disease course, and inform therapeutic response and drug toxicity monitoring [7]. Their integration into clinical practice has significantly improved diagnostic accuracy and facilitated more personalized, outcome-focused treatment approaches.

2.1. Protein Biomarkers

2.1.1. Autoantibodies

Autoantibodies are crucial for the development and clinical management of RA and are important in its diagnosis and progression. RF and ACPA often appear before symptom onset and are associated with more severe disease, forming part of the ACR/EULAR 2010 diagnostic criteria [8]. Additional autoantibodies, including anti-carbamylated protein (anti-CarP) [9] and anti-peptidyl arginine deiminase 4 (anti-PAD4) [10], further improve diagnostic and prognostic accuracy. Table 1 outlines the key autoantibodies associated with RA, including their sensitivity, specificity, prevalence in other conditions, and clinical applications.
Emerging data indicate that multiplex autoantibody profiling improves the prediction of treatment response. Co-expression of RF and ACPA is associated with better outcomes to tumor necrosis factor inhibitors (TNFi), while anti-CarP and anti-PAD4 may indicate a poorer therapeutic response [11]. A broader autoantibody repertoire may indicate increased immune activation [12,13], potentially boosting responsiveness to anti-inflammatory treatments.

2.1.2. Inflammatory Biomarkers

Inflammatory biomarkers, or acute-phase reactants, constitute a critical component in the diagnostic stratification, longitudinal monitoring, and therapeutic evaluation of RA. Principal analytes include C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) [14], serum amyloid A (SAA) [15], matrix metalloproteinase (MMP)-3 [16], calprotectin (S100A8/S100A9) [17], and 14-3-3η protein [18]. Additionally, key pro-inflammatory cytokines, such as interleukin (IL)-6 and TNF-α, play pivotal roles in RA pathophysiology and are increasingly leveraged as both biomarkers and therapeutic targets. Table 2 delineates their respective diagnostic accuracy, specificity profiles, and clinical applicability.
Despite their utility, these inflammatory biomarkers exhibit elevated levels across a spectrum of inflammatory and autoimmune disorders, thereby limiting their disease-specificity. Accordingly, their optimal use lies in integrative diagnostic frameworks that combine inflammatory biomarkers with autoantibody panels and clinical phenotyping.

2.2. Genetic Biomarkers

RA pathogenesis is largely driven by genetic predisposition, with heritability estimates approaching 60% [19]. The genetic contribution is particularly pronounced in seropositive RA [20], where strong associations have been established with the HLA-DRB104 and DRB110 alleles [21]. In contrast, HLA associations in seronegative RA are comparatively attenuated. However, haplotypes such as HLA-B08/DRB103 [22], as well as non-HLA variants including single-nucleotide polymorphisms (SNPs) in CLYBL and ANKRD55 [23,24] have emerged as potential contributors to disease susceptibility. Table 3 lists some key genetic markers in RA, including their sensitivity, specificity, presence in other conditions, and clinical applications.
Variants within the JAK/STAT signaling pathway have been implicated in seropositive RA [25]. Notably, therapeutic response to JAK inhibitors appears to be independent of autoantibody status [26,27], suggesting a broader applicability across RA subtypes. Additional loci with established roles in RA pathogenesis include CTLA4, TRAF1/C5, PTPN22, and PADI2/4, each exhibiting differential allele frequencies and functional relevance across serotypes and populations.
The polygenic nature of RA, compounded by complex gene-environment interactions, such as tobacco exposure, gut microbiota dysbiosis, and epigenetic modifications, poses substantial challenges in pinpointing causal genetic variants. Individual genetic markers frequently exhibit limited diagnostic sensitivity and specificity, emphasizing the need for integrative approaches that account for the multifactorial nature of disease susceptibility.

2.3. Epigenetic Biomarkers

Epigenetic regulation, including DNA methylation, post-translational histone modifications, and non-coding (nc)RNAs, serves as a critical interface between genetic predisposition and environmental exposures in the pathogenesis of RA. These mechanisms orchestrate immune cell differentiation, modulate pro-inflammatory signaling cascades, and influence synovial tissue remodeling, positioning them as promising avenues for biomarker discovery [28,29,30].
Aberrant methylation patterns have been identified in peripheral T and B lymphocytes as well as synovial fibroblasts, even in early-stage RA [28], and are predictive of differential responsiveness to DMARDs [29]. Circulating methylation levels of genes such as CXCR5 and HTR2A correlate with disease activity and may serve as minimally invasive biomarkers [31,32]. Histone acetylation dysregulation within synovial fibroblasts promotes transcriptional upregulation of IL-6 and MMPs, exacerbating local inflammation and joint destruction [33,34,35]. Pharmacologic inhibition of histone deacetylases (HDACs) has demonstrated therapeutic potential in preclinical models, underscoring HDACs as viable drug targets [36]. Beyond acetylation, additional histone modifications, including phosphorylation, ubiquitylation, and sumoylation, contribute to the dynamic epigenetic landscape of RA [37].
Integrating epigenomic profiling into clinical workflows may enhance early diagnostic accuracy, facilitate molecular stratification, and support the development of personalized treatment regimens. As such, epigenetic mechanisms represent a frontier in the advancement of precision medicine for RA.

2.4. Imaging Biomarkers

Imaging biomarkers are essential tools in RA management. They provide objective measures that significantly enhance early diagnosis, prognosis, and monitoring of treatment response. These biomarkers offer insights that go beyond conventional clinical indices, such as the DAS28 and ESR/CRP [38,39]. The most sensitive modalities for visualizing active joint inflammation are Musculoskeletal Ultrasound (MSUS) and MRI [40].
MSUS is a high-resolution, accessible, and dynamic imaging technique that is vital for initial diagnosis, especially in cases of undifferentiated or seronegative arthritis [41]. It is particularly useful for identifying subclinical synovitis and bone erosions that are often overlooked during physical examinations and conventional X-rays [39]. MSUS provides clear images of soft tissues, cartilage, and bone, allowing for the effective identification of early RA signs, including synovial hypertrophy and bone erosions [41]. Its role in guiding clinical decision-making is vital, especially in evaluating synovial inflammation and identifying patients at increased risk of relapse [41]. Notably, studies have demonstrated that patients in clinical remission with residual synovitis visible on MSUS are significantly more likely to experience flare-ups if their medication is tapered [42].
MRI is considered the most sensitive technique for detecting early inflammatory and destructive changes in RA [42,43,44]. It provides superior visualization of both soft tissue and bone structures, making it a valuable tool for early diagnosis and monitoring disease progression [42,43,44]. Key MRI findings include synovitis, bone marrow edema (BME), and erosions [38]. The presence of BME on MRI is especially important, as it signifies a more aggressive disease course and an increased risk of joint damage progression [42]. This insight frequently encourages rheumatologists to initiate early and assertive Treat-to-Target strategies [42]. Moreover, in cases of suspected RA, MRI-detected BME can predict the future development of RA, and in established early RA, it can predict later structural damage progression [45].
Other modalities also have specialized utility despite the dominance of MRI and MSUS in clinical practice. Recent advancements in positron emission tomography/computed tomography (PET/CT) scans are emerging as useful tools for monitoring disease activity and treatment response, providing a functional assessment of inflammation [46]. CT scans offer superior detail of bone structures compared to MRI, making them especially effective for visualizing bony erosions [38]. Interestingly, ultra-low-dose CT has been investigated for its potential to detect synovitis and may identify more differential diagnoses than MRI in patients with suspected RA [47].
Incorporating advanced imaging techniques into clinical practice offers significant diagnostic and prognostic benefits. However, challenges such as high MRI costs, limited accessibility, and the necessity for standardized training have restricted their use in practice [48]. Overcoming these barriers is crucial for maximizing the potential of imaging biomarkers in the personalized management of RA.

2.5. Limitations of Classical Biomarkers

Conventional biomarkers in RA, such as RF and ACPA, often lack clinical utility due to limited specificity and sensitivity. These markers can be present in other autoimmune or inflammatory conditions and are absent in a significant subset of RA patients, particularly in early or seronegative cases. Moreover, they offer minimal insight into disease heterogeneity, failing to capture the dynamic and localized nature of joint pathology [49].
This lack of precision hampers accurate diagnosis, stratification, and prediction of treatment response. As a result, reliance on single biomarkers leads to oversimplified assessments that do not reflect the complex immunopathology of RA [50]. Overcoming these limitations requires a shift toward multi-dimensional biomarker frameworks that integrate molecular, cellular, and tissue-level data for a more robust and individualized approach to disease management.

3. Breakthroughs in the Discovery of RA Biomarkers

Advances in multi-omics technologies are accelerating biomarker discovery in RA by integrating genomics, transcriptomics, proteomics, epigenomics, metabolomics, and microbiomics data with clinical parameters and machine learning (ML). High-throughput platforms such as next-generation sequencing (NGS) and mass spectrometry enable comprehensive molecular profiling, supporting earlier diagnosis, refined prognostication, and personalized therapy [51].
Synovial tissue analysis complements systemic profiling by offering direct insight into local inflammatory mechanisms. Multi-omics interrogation of synovial biopsies has identified molecular subtypes, predicted therapeutic responses [52], and improved diagnostic clarity in undifferentiated arthritis [46]. Though not yet standard practice, synovial profiling holds promise for advancing precision medicine in RA [53,54,55].

3.1. Multi-Omics Approaches

3.1.1. Genomics

Genomics research has substantially advanced our understanding of RA, identifying both HLA and non-HLA loci that influence disease susceptibility, progression, and serological heterogeneity [56]. High-throughput platforms, including genome-wide association studies (GWAS), whole-genome sequencing, and SNP arrays, have identified over 100 loci, notably within HLA-DRB1 and genes regulating immunity, inflammation, and signal transduction. These discoveries have elucidated the genetic architecture of ACPA-positive RA and its divergence from seronegative forms [22].
Beyond the major histocompatibility complex, non-HLA genes such as ATXN2L, MMP-14, GALNT12, and KCNN2 are implicated in joint destruction and comorbidities [57,58,59], with Mendelian randomization and transcriptomic profiling supporting their translational relevance. Notably, IL-17 signaling [60] and angiopoietin-2 polymorphisms [61] have emerged as pivotal in RA pathogenesis.
Population-specific GWAS underscore the necessity of inclusive genomics; studies in Chinese cohorts revealed novel loci (e.g., IL12RB2, CCR2) [62], while multi-ancestry meta-analyses uncovered 124 markers, 34 previously unreported [63]. These findings highlight ethnic variability in genetic risk.
Genomic insights also inform RA subphenotyping. A large GWAS (>31,000 cases) identified JAK/STAT pathway genes as central to seropositive RA, though seronegative RA remains genetically undercharacterized [25]. Collectively, these advances are propelling precision medicine in RA, enabling earlier diagnosis, individualized treatment, and preventive strategies.

3.1.2. Epigenomics

Epigenomics, the study of heritable, reversible modifications to the genome that do not alter DNA sequence, is increasingly recognized as a critical dimension in RA research. Key epigenetic mechanisms, including DNA methylation, histone modifications, and non-coding(nc)RNAs, offer promising avenues for biomarker discovery, patient stratification, and personalized therapeutic strategies [64,65,66].
DNA methylation profiles are particularly valuable due to their stability and accessibility in easily obtainable samples like peripheral blood mononuclear cells and whole blood [28]. This stability makes them robust indicators for disease stratification and prognosis. Research has shown that specific CpG methylation signatures in peripheral blood can differentiate responders from non-responders to TNFi therapy [67] and predict progression from undifferentiated arthritis to RA [68]. A seven differentially methylated positions signature has shown utility in forecasting Leflunomide response [69], while methylation changes in TRIM15, SORC2, and STAT3 correlate with methotrexate efficacy [70].
To address the challenges posed by large-scale analysis, targeted studies focusing on the methylation status of specific individual genes have provided significant and actionable insights for RA. Notably, altered methylation patterns in the Absent in melanoma 2 (AIM2) gene, an essential component of the inflammasome pathway, have been observed in RA patients compared to healthy controls [71]. This finding suggests a connection between epigenetic regulation and inflammation mediated by the AIM2 gene. Additionally, hypomethylation of Homeodomain Interacting Protein Kinase 3 (HIPK3) found in peripheral blood is associated with RA and shows a negative correlation with inflammatory markers, such as CRP [72,73]. This highlights the potential of HIPK3 as a diagnostic biomarker. Overall, these results underscore the promise of DNA methylation markers as non-invasive tools for early diagnosis, prognosis, and therapeutic stratification in RA.
Histone modifications, such as acetylation, methylation, and citrullination, play a crucial role in modulating chromatin architecture and gene expression in RA [74]. Specifically, the enrichment of trimethylation on histone H3 at lysine 4 in synovial fibroblasts serves as a marker for active promoters associated with pro-inflammatory gene expression [75]. Additionally, ncRNAs, including micro (mi)RNAs and long nc (lnc)RNAs, are increasingly recognized for their role in fine-tuning immune signaling pathways and are emerging as valuable diagnostic and prognostic tools for RA.
Environmental exposures such as smoking and pollutants influence epigenetic regulation. Methylation at cg21325723 mediates the interaction between rs6933349 and smoking, modulating ACPA-positive RA risk [76]. Gut microbiota-derived short-chain fatty acids (SCFAs) also impact on the histone deacetylase activity, promoting Treg differentiation and immune tolerance [77,78]. Longitudinal epigenomic studies are mapping methylation trajectories in at-risk individuals, revealing early predictive loci and dynamic changes in immune-related pathways during treatment [66,79]. Early-life exposures, e.g., maternal diet, stress, and neonatal factors, may induce lasting epigenetic changes that elevate RA susceptibility [80].
Combining DNA methylation and histone data with ML models is being developed to predict drug responses [81]. The integration of epigenetic markers with conventional biomarkers, such as RF and ACPA, enhances diagnostic accuracy, particularly for seronegative RA [82]. These advances underscore the potential of epigenomic profiling to refine RA diagnosis, forecast treatment outcomes, and guide precision medicine approaches.

3.1.3. Transcriptomics

Transcriptomics, the comprehensive analysis of RNA transcripts, is a critical tool for biomarker discovery and mechanistic insight in RA. By capturing dynamic gene expression profiles, transcriptomic approaches facilitate disease stratification and support precision medicine. Advances in bulk RNA sequencing (RNA-Seq), single-cell (sc)RNA-Seq, and spatial transcriptomics have enabled high-resolution mapping of transcriptomic activity across distinct cell types and tissue microenvironments [29].
Transcriptomic biomarkers encompass both protein-coding genes and diverse classes of ncRNAs. Differential expression of genes such as ADAMDEC1 distinguishes RA from osteoarthritis [83], while SAA4 correlates with disease activity [84]. Alternative splicing events, including those affecting CD44 and survivin genes, generate isoforms linked to joint destruction and disease progression [85] offering novel diagnostic and therapeutic targets.
NcRNAs play a pivotal role in RA pathogenesis by modulating inflammatory and matrix-degrading pathways [86,87,88]. MiRNAs, such as miR-146a and miR-155, are consistently dysregulated in RA and correlate with disease activity [82]; miR-499 polymorphisms are associated with susceptibility [89]. lncRNAs, including HOTAIR and NEAT1, influence fibroblast-like synoviocytes and immune cell function [90], contributing to chronic inflammation [91]. Circular (circ)RNAs, such as circPTPN22 [92], act as miRNA sponges [93,94] and are emerging as promising biomarkers for diagnosis and patient stratification.
Transcriptomic profiling has also identified early-stage RA biomarkers, such as CXCL10, MMP-3, IL-6, TNF, and S100A8/S100A9, that support early diagnosis and disease monitoring. Moreover, blood-based transcriptomic signatures have demonstrated predictive value for therapeutic response to TNFi, enabling more informed treatment decisions [95].
Collectively, transcriptomics offers a powerful framework for elucidating RA pathogenesis, identifying clinically actionable biomarkers, and guiding individualized therapeutic strategies.

3.1.4. Proteomics

Proteomics provides a comprehensive analysis of protein composition, interactions, and post-translational modifications [96], offering critical insights into molecular and cellular mechanisms underlying various autoimmune diseases. Advanced technologies such as Liquid chromatography-tandem mass spectrometry (LC-MS/MS), matrix-assisted laser desorption/ionization mass spectrometry (MALDI-TOF MS), Two-dimensional gel electrophoresis, and isobaric tagging (e.g., iTRAQ) [97] enable high-throughput quantification and characterization of protein profiles. Mass spectrometry remains central to proteomic workflows and is often integrated with immunoassays like enzyme-linked immunosorbent assay and cytometric bead arrays for validation [98].
Proteomics has emerged as a powerful platform for identifying diagnostic and prognostic biomarkers in RA. Recent studies have demonstrated its utility in detecting early-stage RA before clinical diagnosis by profiling serum analytes such as collagen triple helix repeat containing 1 [99,100], pregnancy zone protein, vitamin D binding protein (VDBP), SAA [84], and cytokines. Sirtuin 1 [101] and SAA4 have also shown promise in distinguishing RA from healthy controls and other inflammatory conditions.
Mun et al. [84] employed LC-MS/MS to identify serum proteins (Angiotensinogen, SAA4, VDBP, Retinol-binding protein 4) that differentiate RA patients, including seronegative cases, from controls. Additionally, integrative approaches combining proteomics with ML and protein–protein interaction networks have revealed 24 candidate biomarkers [102] with potential clinical relevance.
Methodological innovations continue to expand proteomic capabilities. Optimization of the Sequential window acquisition of all theoretical mass spectra (SWATH-MS) workflow has improved data reproducibility and proteome coverage, enabling more detailed plasma profiling. Targeted proteomics of synovial fluid has further elucidated post-translational modifications in autoantigens, offering new avenues for biomarker development and disease activity monitoring.

3.1.5. Metabolomics

Metabolomics, which involves the comprehensive profiling of small-molecule metabolites, provides a high-resolution overview of systemic physiology and is gaining prominence in RA research. Notably, metabolic signatures, especially those originating from the gut microbiome, are being recognized as significant contributors to the pathogenesis of RA [103]. This emerging understanding holds promise for applications in early diagnosis, disease monitoring, and therapeutic stratification [104]. Table 4 presents a summary of various metabolites, highlighting their diagnostic performance, specificity, and potential clinical applicability as biomarkers for RA.
Microbial metabolites have demonstrated strong associations with RA-related inflammatory pathways [103,105]. SCFAs, such as butyrate and propionate, produced via microbial fermentation of dietary fibers, exert anti-inflammatory effects. Their reduced levels in RA patients suggest a role in promoting systemic inflammation and disease progression. Indole-3-propionate (IPA), a tryptophan-derived microbial metabolite, has been inversely correlated with RA risk [106], indicating a protective immunomodulatory function.
Sphingolipids, including sphingomyelin species like SM 16:1, are integral to membrane structure and signaling. These lipids have been linked to reduced RA susceptibility [106]. Heptadecasphin-4-enine, another sphingolipid, has shown diagnostic potential [107], reinforcing the relevance of lipid metabolism in RA. Similarly, Glycine, an anti-inflammatory amino acid, has been negatively associated with RA incidence [108]. Conversely, elevated argininosuccinate, an intermediate in the urea cycle and nitric oxide synthesis, is associated with increased RA risk [106], likely due to its pro-inflammatory properties.
Although current findings are largely correlational, they provide mechanistic insights into metabolic dysregulation in RA and highlight novel therapeutic targets. Strategies such as modulating microbial-derived metabolites or supplementing protective compounds may offer new avenues for intervention.

3.1.6. Microbiomics

The human microbiota is a diverse ecosystem of microorganisms, including bacteria, archaea, fungi, and viruses, that plays a critical role in immune regulation, metabolism, and host homeostasis. Recent studies increasingly suggest that gut dysbiosis, which refers to an imbalance in these microbial communities, may be a key factor in the development of RA [109]. This disruption of microbial equilibrium often occurs before the appearance of clinical symptoms and can cause inflammation in the intestines, thereby impairing the integrity of the epithelial barrier [110]. Dysbiosis can produce microbial metabolites and directly influence immune cell activity, resulting in abnormal systemic immune responses that may initiate disease in individuals with a genetic predisposition.
Comparative studies reveal significant differences in microbial taxa and metabolite profiles between RA patients and healthy controls [111]. One notable finding is the increased relative abundance of Prevotella copri in the gut microbiota in newly diagnosed RA patients [112,113]. This dysbiosis creates a pro-inflammatory environment by promoting the differentiation and expansion of Th17 cells within the intestinal lamina propria [113]. Additionally, an immunogenic protein derived from Prevotella copri has been linked to the breakdown of immune tolerance through a mechanism called molecular mimicry. This process can trigger Th1 cell responses and antibody production in certain RA patients [114]. In contrast, Akkermansia muciniphila, a bacterium that degrades mucin and is typically associated with enhanced mucosal barrier function and metabolic health, shows variable abundance in individuals with RA [115,116]. While its mucolytic activity can have both beneficial and adverse effects, its positive contributions are often related to strengthening tight junction integrity and producing SCFAs like acetate and propionate [115]. These metabolites are crucial for modulating T-cell responses and maintaining immune tolerance, potentially offering protection against inflammation [112,117]. Other important microbial metabolites, such as IPA, sphingomyelin species, argininosuccinate, glycine, and heptadecasphin-4-enine, have also been associated with either protective or pro-inflammatory roles in RA pathogenesis [118].
Microbiome-derived biomarkers are emerging as tools for diagnosis, disease monitoring, and therapeutic stratification (Table 5). Interventions targeting the microbiota, including dietary modulation and the use of specific probiotics or fecal microbiota transplantation, show promise as adjunctive strategies for RA management [119].

3.1.7. Bioinformatics

Bioinformatics is a foundational pillar of contemporary RA research, providing the computational infrastructure to integrate and interrogate complex biological and clinical datasets. Through a systems biology framework, bioinformatics enables the fusion of multi-omics data, including genomics, transcriptomics, proteomics, epigenomics, and metabolomics, and microbiomics with clinical phenotypes. This integrative approach offers a multidimensional view of RA pathogenesis and facilitates the identification of robust biomarkers and actionable therapeutic targets [120,121,122,123,124,125].
Multi-omics integration and predictive modeling: Advanced bioinformatics pipelines harmonize heterogeneous omics datasets, allowing for the discovery of composite biomarkers for diagnosis, disease activity monitoring, and treatment response prediction [56,126,127]. Data fusion techniques are particularly valuable for constructing unified disease models that reflect RA’s molecular heterogeneity. ML and deep learning algorithms further enhance predictive modeling by extracting clinically relevant patterns from high-dimensional data. For instance, the Random Forest algorithm effectively integrates transcriptomic (RNA-Seq) and DNA methylation signatures from CD4+ T cells and monocytes [128]. This approach successfully predicts responses to biologics such as adalimumab (ADA) with approximately 86% accuracy and etanercept (ETN) with about 79% accuracy. It also identifies pathways, such as TNF signaling, that are enriched in ADA and ETN responders [128,129]. In the analysis of clinical data from thousands of RA patients, ML models like Lasso regression have been shown to outperform traditional methods in predicting the one-year persistence of methotrexate therapy [130]. Additionally, deep learning approaches, including Autoencoders, are being employed to combine high-dimensional multi-omics data to extract and define more homogeneous molecular subtypes of RA [131,132]. This work is paving the way for precise patient stratification.
Pattern recognition and computational validation: Advanced bioinformatics tools employ sophisticated pattern recognition and data mining algorithms to detect subtle regulatory perturbations in gene expression, miRNA profiles, and DNA methylation landscapes. These insights illuminate key pathogenic pathways [120,121,122,123]. Computational validation, through cross-referencing candidate biomarkers with curated databases and literature, is indispensable for ensuring reproducibility, robustness, and translational relevance before experimental validation.
Data processing and visualization: Initial data preprocessing and quality control are typically conducted using established platforms such as Bioconductor, FastQC, and DeepVariant. However, the increasing scale and complexity of RA datasets necessitate the adoption of ML- and artificial intelligence (AI)-driven analytical frameworks capable of extracting clinically actionable insights [14]. Visualization tools, such as heatmaps, network graphs, and dimensionality-reduction plots, play a vital role in translating complex data into interpretable formats for interdisciplinary collaboration.
These computational approaches are vital for advancing precision medicine in RA, offering scalable solutions for early diagnosis, patient stratification, and individualized therapeutic interventions.

3.1.8. Summary

The integration of multi-omics datasets with advanced bioinformatics frameworks is reshaping RA research. This system-level approach enables a deeper understanding of RA pathogenesis by linking molecular data, including genomics, transcriptomics, proteomics, metabolomics, and epigenomics, with clinical phenotypes. For example, the integration of plasma proteomics and metabolomics with clinical data allows for the identification of distinct molecular signatures in ACPA-negative RA patients, which are often poorly defined by traditional methods. This process reveals unique immune and metabolic pathways that differentiate them from ACPA-positive patients and can be used to refine diagnostic and stratification strategies [133]. Analyzing baseline transcriptomic and proteomic profiles (e.g., specific genes and protein levels in peripheral blood cells) can predict which patients will be non-responders to an anti-TNF drug [134]. This allows clinicians to switch patients immediately to a more effective therapy, preventing joint damage and unnecessary drug exposure [134]. During this integration, bioinformatics plays a pivotal role in harmonizing these complex datasets, facilitating the identification of high-confidence biomarkers (Figure 1).
Despite its potential, the widespread adoption of multi-omics technologies faces significant challenges. One major barrier is the high cost of comprehensive multi-omics testing, which particularly affects its implementation in routine clinical settings. To overcome this issue, innovations are needed in assay miniaturization, cost-effective sequencing platforms, and the strategic selection of omics layers based on their clinical utility. Moreover, the complexity of data analysis requires robust computational infrastructure and interdisciplinary expertise, highlighting the importance of developing scalable, user-friendly bioinformatics tools and standardized workflows.
Currently, multi-omics approaches are mainly used in research environments to support hypothesis generation and biomarker discovery. However, commercial panels, especially in the fields of genomics and transcriptomics, are starting to emerge, providing targeted assays for clinical decision-making [135]. As analytical frameworks advance and costs decrease, the shift from research to clinical applications is expected to accelerate, opening the door to personalized medicine in RA.

3.1.9. Molecular Signatures and Synovial Biopsy-Based Biomarkers

Persistent synovial inflammation is a defining feature of RA, and its direct interrogation is essential for elucidating disease mechanisms, refining patient stratification, and informing therapeutic decisions. The advent of minimally invasive ultrasound-guided biopsy has enabled consistent acquisition of high-fidelity synovial tissue [136], catalyzing synovial biomarker discovery.
Advanced molecular profiling techniques, including bulk and single-cell transcriptomics, proteomics, and spatial cellular analyses, have delineated distinct synovial pathotypes. The lympho-myeloid subtype, characterized by dense immune cell infiltration, correlates with aggressive joint damage [52] and increased reliance on bDMARDs [137]. Conversely, the fibroid/pauci-immune pathotype exhibits poor responsiveness to TNFi such as certolizumab-pegol [138], underscoring the need for tailored therapeutic strategies.
Biopsy-driven molecular stratification has demonstrated clinical utility. In TNFi-refractory RA, patients with low B-cell molecular signatures responded preferentially to tocilizumab over rituximab [139]. Transcriptomic analyses further identified a fibroblast-enriched signature predictive of rituximab resistance [140], highlighting the role of stromal cell phenotypes in treatment failure.
Integrative studies, notably by Zhang et al., have mapped the synovial cellular landscape using multi-modal approaches, identifying 18 immune and stromal populations [141], and constructing a comprehensive single-cell atlas from over 314,000 cells [142]. Reduced PD-1hiCXCR5- peripheral helper T-cell frequencies in the synovium were associated with favorable TNFi responses [143], suggesting novel predictive biomarkers.
These insights affirm synovial tissue as a critical reservoir for precision medicine in RA. Integrating synovial and peripheral immune profiling holds the potential to refine therapeutic strategies and accelerate the development of biomarker-driven treatments. However, the successful integration of synovial biopsy in both clinical and research settings relies on overcoming several critical procedural challenges. One major issue is accessibility; the technique requires specialized equipment and skilled personnel, which limits its availability to a small number of medical centers. Therefore, it is essential to standardize biopsy protocols, including site selection, tissue handling, and analytical methods, to ensure reproducibility and comparability across different studies and institutions. Furthermore, improving the consistency of procedures is crucial for effective long-term monitoring and comparative analyses, especially in clinical trials assessing therapeutic responses. Advancements in these areas are vital for optimizing the use of synovial tissue in research and clinical applications for RA.

4. Challenges and Future Directions of Biomarkers in RA

The incorporation of biomarkers into the care of RA represents a major step forward in precision medicine. However, the molecular and clinical diversity associated with RA complicates patient classification and reduces the predictive power of individual biomarkers. To address this challenge, multi-omics approaches and synovial biopsy generate high-dimensional datasets that require advanced computational tools, particularly ML and AI, to identify actionable biomarkers and clarify disease mechanisms. The integration of multi-omics data and AI-driven analysis is not merely a trend; it represents a critical evolution in the development of personalized care for RA. Looking forward, several innovative directions show promise for transforming RA management. Liquid biopsy methods, such as blood-based assays [144], may provide non-invasive alternatives to synovial tissue profiling, enhancing accessibility and facilitating dynamic disease monitoring. Furthermore, the integration of multi-omics data into digital twin models [145] could allow for the simulation of individual treatment responses, enabling clinicians to evaluate therapeutic strategies in a virtual environment before actual implementation. These advancements, coupled with improvements in wearable biosensors, real-time data analytics, and cloud-based diagnostic platforms, are poised to reshape the landscape of RA care and accelerate the realization of precision medicine.
Despite promising discoveries, translation into clinical practice remains challenging. Assay validation, cross-platform standardization, and reproducibility across diverse populations are essential for regulatory approval and clinical adoption. Interdisciplinary collaboration among clinicians, data scientists, and regulatory bodies is vital to overcoming translational barriers.
To bridge the gap between research and routine care, it is essential to focus on technology translation. This includes conducting thorough cost-effectiveness analyses to demonstrate the clinical value of biomarker-driven approaches, particularly in resource-constrained settings. Establishing standardized protocols for sample collection, data processing, and interpretation will enhance reproducibility and facilitate collaboration across multiple centers. Additionally, developing automated analysis platforms that can integrate multi-omics data with clinical parameters will streamline workflows and reduce the burden on healthcare providers. Equally important is the careful integration of these technologies into existing clinical pathways, ensuring that diagnostic innovations enhance rather than disrupt current practices. Future priorities include developing personalized biomarker panels, embedding AI-driven analytics into clinical workflows, and establishing interoperable diagnostic platforms. Addressing economic and logistical hurdles, such as assay cost, regulatory complexity, and clinician education, is critical for widespread implementation. Advancing these strategies will enable biomarker-guided care that supports earlier diagnosis, individualized treatment selection, and dynamic disease monitoring, ultimately improving outcomes for patients with RA.

Author Contributions

Conceptualization: M.X. and L.M. Data curation: M.X., H.W., and F.C. Formal analysis: M.X., F.C., H.W., C.J.J., and L.M. Funding acquisition: M.X. and L.M. Visualization: M.X. and H.W. Writing—original draft: M.X., F.C., and H.W. Writing—review and editing: M.X., C.J.J., and L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work is generously supported by the CLEARbridge Foundation, the Lincoln Centre, and the Woodend Foundation.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The authors thanked Yi Jie for her insightful comments.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
RARheumatoid arthritis
RFRheumatoid factor
ACPAAnti-citrullinated protein antibodies
cs/bDMARDsConventional synthetic/biologic disease-modifying antirheumatic drugs
anti-CarPAnti-carbamylated protein
anti-PAD4Anti-peptidyl arginine deiminase 4
CRPC-reactive protein
ESRErythrocyte sedimentation rate
SAASerum amyloid A
MMPMatrix metalloproteinase
ILInterleukin
TNFTumor necrosis factor
SNPSingle-nucleotide polymorphism
HDACInhibition of histone deacetylase
MLMachine learning
NGSNext-generation sequencing
GWASGenome-wide association studies
SCFAShort-chain fatty acid
VDBPVitamin D binding protein
IPAIndole-3-propionate

References

  1. van Vollenhoven, R.F. Sex differences in rheumatoid arthritis: More than meets the eye. BMC Med. 2009, 7, 12. [Google Scholar] [CrossRef]
  2. Almutairi, K.; Nossent, J.; Preen, D.; Keen, H.; Inderjeeth, C. The global prevalence of rheumatoid arthritis: A meta-analysis based on a systematic review. Rheumatol. Int. 2021, 41, 863–877. [Google Scholar] [CrossRef] [PubMed]
  3. Pongratz, G.; Frieser, R.; Brinks, R.; Schneider, M.; Hartung, W.; Fleck, M.; Ehrenstein, B. Association between autoantibody level and disease activity in rheumatoid arthritis is dependent on baseline inflammation. Clin. Exp. Rheumatol. 2020, 38, 691–698. [Google Scholar]
  4. Khader, Y.; Beran, A.; Ghazaleh, S.; Lee-Smith, W.; Altorok, N. Predictors of remission in rheumatoid arthritis patients treated with biologics: A systematic review and meta-analysis. Clin. Rheumatol. 2022, 41, 3615–3627. [Google Scholar] [CrossRef]
  5. Wang, W.; Zhou, H.; Liu, L. Side effects of methotrexate therapy for rheumatoid arthritis: A systematic review. Eur. J. Med. Chem. 2018, 158, 502–516. [Google Scholar] [CrossRef]
  6. Burmester, G.R.; Pope, J.E. Novel treatment strategies in rheumatoid arthritis. Lancet 2017, 389, 2338–2348. [Google Scholar] [CrossRef]
  7. Takeuchi, T. Biomarkers as a treatment guide in rheumatoid arthritis. Clin. Immunol. 2018, 186, 59–62. [Google Scholar] [CrossRef]
  8. Aletaha, D.; Neogi, T.; Silman, A.J.; Funovits, J.; Felson, D.T.; Bingham, C.O., 3rd; Birnbaum, N.S.; Burmester, G.R.; Bykerk, V.P.; Cohen, M.D.; et al. 2010 rheumatoid arthritis classification criteria: An American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann. Rheum. Dis. 2010, 69, 1580–1588. [Google Scholar] [CrossRef]
  9. Shi, J.; Knevel, R.; Suwannalai, P.; van der Linden, M.P.; Janssen, G.M.; van Veelen, P.A.; Levarht, N.E.; van der Helm-van Mil, A.H.; Cerami, A.; Huizinga, T.W. Autoantibodies recognizing carbamylated proteins are present in sera of patients with rheumatoid arthritis and predict joint damage. Proc. Natl. Acad. Sci. USA 2011, 108, 17372–17377. [Google Scholar] [CrossRef]
  10. Darrah, E.; Giles, J.T.; Davis, R.L.; Naik, P.; Wang, H.; Konig, M.F.; Cappelli, L.C.; Bingham, C.O., 3rd; Danoff, S.K.; Andrade, F. Autoantibodies to Peptidylarginine Deiminase 2 Are Associated With Less Severe Disease in Rheumatoid Arthritis. Front. Immunol. 2018, 9, 2696. [Google Scholar] [CrossRef]
  11. Julià, A.; López-Lasanta, M.; Blanco, F.; Gómez, A.; Haro, I.; Mas, A.J.; Erra, A.; Vivar, M.L.G.; Monfort, J.; Sánchez-Fernández, S.; et al. Interactions between rheumatoid arthritis antibodies are associated with the response to anti-tumor necrosis factor therapy. BMC Musculoskelet. Disord. 2021, 22, 372. [Google Scholar] [CrossRef] [PubMed]
  12. Petro, A.D.; Dougherty, J.; England, B.R.; Sayles, H.; Duryee, M.J.; Hunter, C.D.; Kremer, J.M.; Pappas, D.A.; Robinson, W.H.; Curtis, J.R.; et al. Associations between an expanded autoantibody profile and treatment responses to biologic therapies in patients with rheumatoid arthritis. Int. Immunopharmacol. 2021, 91, 107260. [Google Scholar] [CrossRef]
  13. de Moel, E.C.; Derksen, V.; Stoeken, G.; Trouw, L.A.; Bang, H.; Goekoop, R.J.; Speyer, I.; Huizinga, T.W.J.; Allaart, C.F.; Toes, R.E.M.; et al. Baseline autoantibody profile in rheumatoid arthritis is associated with early treatment response but not long-term outcomes. Arthritis Res. Ther. 2018, 20, 33. [Google Scholar] [CrossRef] [PubMed]
  14. Abdelhafiz, D.; Baker, T.; Glascow, D.A.; Abdelhafiz, A. Biomarkers for the diagnosis and treatment of rheumatoid arthritis—A systematic review. Postgrad. Med. 2023, 135, 214–223. [Google Scholar] [CrossRef]
  15. Sorić Hosman, I.; Kos, I.; Lamot, L. Serum Amyloid A in Inflammatory Rheumatic Diseases: A Compendious Review of a Renowned Biomarker. Front. Immunol. 2020, 11, 631299. [Google Scholar] [CrossRef] [PubMed]
  16. Sun, S.; Bay-Jensen, A.C.; Karsdal, M.A.; Siebuhr, A.S.; Zheng, Q.; Maksymowych, W.P.; Christiansen, T.G.; Henriksen, K. The active form of MMP-3 is a marker of synovial inflammation and cartilage turnover in inflammatory joint diseases. BMC Musculoskelet. Disord. 2014, 15, 93. [Google Scholar] [CrossRef]
  17. Inciarte-Mundo, J.; Frade-Sosa, B.; Sanmartí, R. From bench to bedside: Calprotectin (S100A8/S100A9) as a biomarker in rheumatoid arthritis. Front. Immunol. 2022, 13, 1001025. [Google Scholar] [CrossRef]
  18. Li, P.; Wang, X.; Guo, X.; Wen, Y.; Liu, L.; Liang, X.; Du, Y.; Wu, C.; Wang, S.; Zhang, F. Integrative analysis of genome-wide association study and expression quantitative trait loci datasets identified various immune cell-related pathways for rheumatoid arthritis. Ann. Hum. Genet. 2020, 84, 72–79. [Google Scholar] [CrossRef]
  19. Dedmon, L.E. The genetics of rheumatoid arthritis. Rheumatology 2020, 59, 2661–2670. [Google Scholar] [CrossRef]
  20. Frisell, T.; Holmqvist, M.; Källberg, H.; Klareskog, L.; Alfredsson, L.; Askling, J. Familial risks and heritability of rheumatoid arthritis: Role of rheumatoid factor/anti-citrullinated protein antibody status, number and type of affected relatives, sex, and age. Arthritis Rheum. 2013, 65, 2773–2782. [Google Scholar] [CrossRef]
  21. Gregersen, P.K.; Silver, J.; Winchester, R.J. The shared epitope hypothesis. An approach to understanding the molecular genetics of susceptibility to rheumatoid arthritis. Arthritis Rheum. 1987, 30, 1205–1213. [Google Scholar] [CrossRef]
  22. Padyukov, L. Genetics of rheumatoid arthritis. Semin. Immunopathol. 2022, 44, 47–62. [Google Scholar] [CrossRef] [PubMed]
  23. Bossini-Castillo, L.; de Kovel, C.; Kallberg, H.; van’t Slot, R.; Italiaander, A.; Coenen, M.; Tak, P.P.; Posthumus, M.D.; Wijmenga, C.; Huizinga, T.; et al. A genome-wide association study of rheumatoid arthritis without antibodies against citrullinated peptides. Ann. Rheum. Dis. 2015, 74, e15. [Google Scholar] [CrossRef] [PubMed]
  24. Eyre, S.; Bowes, J.; Diogo, D.; Lee, A.; Barton, A.; Martin, P.; Zhernakova, A.; Stahl, E.; Viatte, S.; McAllister, K.; et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat. Genet. 2012, 44, 1336–1340. [Google Scholar] [CrossRef] [PubMed]
  25. Saevarsdottir, S.; Stefansdottir, L.; Sulem, P.; Thorleifsson, G.; Ferkingstad, E.; Rutsdottir, G.; Glintborg, B.; Westerlind, H.; Grondal, G.; Loft, I.C.; et al. Multiomics analysis of rheumatoid arthritis yields sequence variants that have large effects on risk of the seropositive subset. Ann. Rheum. Dis. 2022, 81, 1085–1095. [Google Scholar] [CrossRef]
  26. Martinez-Molina, C.; Gich, I.; Diaz-Torné, C.; Park, H.S.; Feliu, A.; Vidal, S.; Corominas, H. Patient-related factors influencing the effectiveness and safety of Janus Kinase inhibitors in rheumatoid arthritis: A real-world study. Sci. Rep. 2024, 14, 172. [Google Scholar] [CrossRef]
  27. Pombo-Suarez, M.; Sanchez-Piedra, C.; Gómez-Reino, J.; Lauper, K.; Mongin, D.; Iannone, F.; Pavelka, K.; Nordström, D.C.; Inanc, N.; Codreanu, C.; et al. After JAK inhibitor failure: To cycle or to switch, that is the question—Data from the JAK-pot collaboration of registries. Ann. Rheum. Dis. 2023, 82, 175–181. [Google Scholar] [CrossRef]
  28. Ciechomska, M.; Roszkowski, L.; Maslinski, W. DNA Methylation as a Future Therapeutic and Diagnostic Target in Rheumatoid Arthritis. Cells 2019, 8, 953. [Google Scholar] [CrossRef]
  29. Wang, Y.; Liu, B.; Zhao, G.; Lee, Y.; Buzdin, A.; Mu, X.; Zhao, J.; Chen, H.; Li, X. Spatial transcriptomics: Technologies, applications and experimental considerations. Genomics 2023, 115, 110671. [Google Scholar] [CrossRef]
  30. Glant, T.T.; Mikecz, K.; Rauch, T.A. Epigenetics in the pathogenesis of rheumatoid arthritis. BMC Med. 2014, 12, 35. [Google Scholar] [CrossRef]
  31. Shi, Y.; Chang, C.; Xu, L.; Jiang, P.; Wei, K.; Zhao, J.; Xu, L.; Jin, Y.; Zhang, R.; Wang, H.; et al. Circulating DNA methylation level of CXCR5 correlates with inflammation in patients with rheumatoid arthritis. Immun. Inflamm. Dis. 2023, 11, e902. [Google Scholar] [CrossRef] [PubMed]
  32. Zhao, J.; Xu, L.; Chang, C.; Jiang, P.; Wei, K.; Shi, Y.; Xu, L.; Zheng, Y.; Shan, Y.; Bian, Y.; et al. Circulating methylation level of HTR2A is associated with inflammation and disease activity in rheumatoid arthritis. Front. Immunol. 2022, 13, 1054451. [Google Scholar] [CrossRef] [PubMed]
  33. Burchill, M.A.; Yang, J.; Vogtenhuber, C.; Blazar, B.R.; Farrar, M.A. IL-2 receptor [beta]-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J. Immunol. 2007, 178, 280–290. [Google Scholar] [CrossRef] [PubMed]
  34. Seto, E.; Yoshida, M. Erasers of histone acetylation: The histone deacetylase enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef]
  35. Araki, Y.; Tsuzuki Wada, T.; Aizaki, Y.; Sato, K.; Yokota, K.; Fujimoto, K.; Kim, Y.T.; Oda, H.; Kurokawa, R.; Mimura, T. Histone Methylation and STAT-3 Differentially Regulate Interleukin-6-Induced Matrix Metalloproteinase Gene Activation in Rheumatoid Arthritis Synovial Fibroblasts. Arthritis Rheumatol. 2016, 68, 1111–1123. [Google Scholar] [CrossRef]
  36. Pai, P.; Vijeev, A.; Phadke, S.; Shetty, M.G.; Sundara, B.K. Epi-revolution in rheumatology: The potential of histone deacetylase inhibitors for targeted rheumatoid arthritis intervention. Inflammopharmacology 2024, 32, 2109–2123. [Google Scholar] [CrossRef]
  37. Araki, Y.; Mimura, T. The Histone Modification Code in the Pathogenesis of Autoimmune Diseases. Mediat. Inflamm. 2017, 2017, 2608605. [Google Scholar] [CrossRef]
  38. Šenolt, L.; Grassi, W.; Szodoray, P. Laboratory biomarkers or imaging in the diagnostics of rheumatoid arthritis? BMC Med. 2014, 12, 49. [Google Scholar] [CrossRef]
  39. Walter, W.R.; Samim, M. Imaging Updates in Rheumatoid Arthritis. Semin. Musculoskelet. Radiol. 2025, 29, 156–166. [Google Scholar] [CrossRef]
  40. Baker, J.F.; Conaghan, P.G.; Gandjbakhch, F. Update on magnetic resonance imaging and ultrasound in rheumatoid arthritis. Clin. Exp. Rheumatol. 2018, 36 (Suppl. 114), 16–23. [Google Scholar]
  41. Silvagni, E.; Zandonella Callegher, S.; Mauric, E.; Chiricolo, S.; Schreiber, N.; Tullio, A.; Zabotti, A.; Scirè, C.A.; Dejaco, C.; Sakellariou, G. Musculoskeletal ultrasound for treating rheumatoid arthritis to target-a systematic literature review. Rheumatology 2022, 61, 4590–4602. [Google Scholar] [CrossRef] [PubMed]
  42. D’Ignazio, E.; Corradini, D.; Cazenave, T.; Bixio, R.; Baldi, C.; Ubhi, H.K.; Smith, K.; Wakefield, R.J.; Emery, P.; Di Matteo, A. Ultrasound Beyond Joints: A Review of Extra-Articular Applications in Rheumatology. Curr. Rheumatol. Rep. 2025, 27, 20. [Google Scholar] [CrossRef] [PubMed]
  43. Hunter, D.J.; Conaghan, P.G. Imaging outcomes and their role in determining outcomes in osteoarthritis and rheumatoid arthritis. Curr. Opin. Rheumatol. 2006, 18, 157–162. [Google Scholar] [CrossRef] [PubMed]
  44. Emery, P. Magnetic resonance imaging: Opportunities for rheumatoid arthritis disease assessment and monitoring long-term treatment outcomes. Arthritis Res. 2002, 4 (Suppl. 2), S6–S10. [Google Scholar] [CrossRef]
  45. Østergaard, M.; Boesen, M. Imaging in rheumatoid arthritis: The role of magnetic resonance imaging and computed tomography. Radiol. Med. 2019, 124, 1128–1141. [Google Scholar] [CrossRef]
  46. Singh, S.B.; Bhandari, S.; Bhandari, S.; Bhandari, S.; Singh, R.; Raynor, W.Y.; Hess, S.; Werner, T.J.; Alavi, A.; Revheim, M.E. Role of PET/CT in diagnosing and monitoring disease activity in rheumatoid arthritis: A review. Ann. Nucl. Med. 2024, 38, 165–175. [Google Scholar] [CrossRef]
  47. Diekhoff, T.; Ulas, S.T.; Poddubnyy, D.; Schneider, U.; Hermann, S.; Biesen, R.; Burmester, G.R.; Hamm, B.; Hermann, K.G. Ultra-low-dose CT detects synovitis in patients with suspected rheumatoid arthritis. Ann. Rheum. Dis. 2019, 78, 31–35. [Google Scholar] [CrossRef]
  48. Minopoulou, I.; Kleyer, A.; Yalcin-Mutlu, M.; Fagni, F.; Kemenes, S.; Schmidkonz, C.; Atzinger, A.; Pachowsky, M.; Engel, K.; Folle, L.; et al. Imaging in inflammatory arthritis: Progress towards precision medicine. Nat. Rev. Rheumatol. 2023, 19, 650–665. [Google Scholar] [CrossRef]
  49. Lord, M.S.; Farrugia, B.L.; Rnjak-Kovacina, J.; Whitelock, J.M. Current serological possibilities for the diagnosis of arthritis with special focus on proteins and proteoglycans from the extracellular matrix. Expert Rev. Mol. Diagn. 2015, 15, 77–95. [Google Scholar] [CrossRef]
  50. Curtis, J.R.; Weinblatt, M.E.; Shadick, N.A.; Brahe, C.H.; Østergaard, M.; Hetland, M.L.; Saevarsdottir, S.; Horton, M.; Mabey, B.; Flake, D.D., 2nd; et al. Validation of the adjusted multi-biomarker disease activity score as a prognostic test for radiographic progression in rheumatoid arthritis: A combined analysis of multiple studies. Arthritis Res. Ther. 2021, 23, 1. [Google Scholar] [CrossRef]
  51. Curtis, J.R.; Strand, V.; Golombek, S.; Zhang, L.; Wong, A.; Zielinski, M.C.; Akmaev, V.R.; Saleh, A.; Asgarian, S.; Withers, J.B. Patient outcomes improve when a molecular signature test guides treatment decision-making in rheumatoid arthritis. Expert Rev. Mol. Diagn. 2022, 3, 1–10. [Google Scholar] [CrossRef] [PubMed]
  52. Humby, F.; Lewis, M.; Ramamoorthi, N.; Hackney, J.A.; Barnes, M.R.; Bombardieri, M.; Setiadi, A.F.; Kelly, S.; Bene, F.; DiCicco, M.; et al. Synovial cellular and molecular signatures stratify clinical response to csDMARD therapy and predict radiographic progression in early rheumatoid arthritis patients. Ann. Rheum. Dis. 2019, 78, 761–772. [Google Scholar] [CrossRef] [PubMed]
  53. Orr, C.; Vieira-Sousa, E.; Boyle, D.L.; Buch, M.H.; Buckley, C.D.; Canete, J.D.; Catrina, A.I.; Choy, E.H.S.; Emery, P.; Fearon, U.; et al. Synovial tissue research: A state-of-the-art review. Nat. Rev. Rheumatol. 2017, 13, 463–475, Erratum in Nat. Rev. Rheumatol. 2017, 13, 630; Erratum in Nat. Rev. Rheumatol. 2017, 14, 60. [Google Scholar] [CrossRef] [PubMed]
  54. Small, A.; Wechalekar, M.D. Synovial biopsies in inflammatory arthritis: Precision medicine in rheumatoid arthritis. Expert Rev. Mol. Diagn. 2020, 20, 315–325. [Google Scholar] [CrossRef]
  55. Triaille, C.; Lauwerys, B.R. Synovial Tissue: Turning the Page to Precision Medicine in Arthritis. Front. Med. 2019, 6, 46. [Google Scholar] [CrossRef]
  56. Gong, X.; Su, L.; Huang, J.; Liu, J.; Wang, Q.; Luo, X.; Yang, G.; Chi, H. An overview of multi-omics technologies in rheumatoid arthritis: Applications in biomarker and pathway discovery. Front. Immunol. 2024, 15, 1381272. [Google Scholar] [CrossRef]
  57. Zhao, R.; Zhang, Y.W.; Guo, J.C.; Qiao, J.; Song, S.; Zhang, T.T.; Zhang, H.Y.; Zhang, S.X. Genetic evidence reveals a causal relationship between rheumatoid arthritis and interstitial lung disease. Front. Genet. 2024, 15, 1395315. [Google Scholar] [CrossRef]
  58. Lo, H.J.; Tsai, C.H.; Huang, T.W. Apoptosis-associated genetic mechanisms in the transition from rheumatoid arthritis to osteoporosis: A bioinformatics and functional analysis approach. APL Bioeng. 2024, 8, 046107. [Google Scholar] [CrossRef]
  59. Hayashi, S.; Matsubara, T.; Fukuda, K.; Maeda, T.; Funahashi, K.; Hashimoto, M.; Kamenaga, T.; Takashima, Y.; Kuroda, R. A genome-wide association study identifying the SNPs predictive of rapid joint destruction in patients with rheumatoid arthritis. Biomed. Rep. 2021, 14, 31. [Google Scholar] [CrossRef]
  60. Zhao, R.; Zhang, Y.W.; Yao, J.Y.; Qiao, J.; Song, S.; Zhang, S.X.; Wang, C.H.; Li, X.F. Genetic association between interleukin-17 and susceptibility to rheumatoid arthritis. BMC Med. Genom. 2023, 16, 277. [Google Scholar] [CrossRef]
  61. Nasef, S.I.; Ellawindy, A.; Askar, A.M.; Hashem, A.A.; Omar, H.H. Assessment of Angiopoietin-2 Single Nucleotide Polymorphism in Patients with Rheumatoid Arthritis. Inflammation 2023, 46, 853–860. [Google Scholar] [CrossRef] [PubMed]
  62. Leng, R.X.; Di, D.S.; Ni, J.; Wu, X.X.; Zhang, L.L.; Wang, X.F.; Liu, R.S.; Huang, Q.; Fan, Y.G.; Pan, H.F.; et al. Identification of new susceptibility loci associated with rheumatoid arthritis. Ann. Rheum. Dis. 2020, 79, 1565–1571. [Google Scholar] [CrossRef] [PubMed]
  63. Ishigaki, K.; Sakaue, S.; Terao, C.; Luo, Y.; Sonehara, K.; Yamaguchi, K.; Amariuta, T.; Too, C.L.; Laufer, V.A.; Scott, I.C.; et al. Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis. Nat. Genet. 2022, 54, 1640–1651. [Google Scholar] [CrossRef]
  64. Hammaker, D.; Firestein, G.S. Epigenetics of inflammatory arthritis. Curr. Opin. Rheumatol. 2018, 30, 188–196. [Google Scholar] [CrossRef]
  65. Yang, C.; Li, D.; Teng, D.; Zhou, Y.; Zhang, L.; Zhong, Z.; Yang, G.J. Epigenetic Regulation in the Pathogenesis of Rheumatoid Arthritis. Front. Immunol. 2022, 13, 859400. [Google Scholar] [CrossRef]
  66. Prideaux, E.B.; Boyle, D.L.; Choi, E.; Buckner, J.H.; Robinson, W.H.; Holers, V.M.; Deane, K.D.; Firestein, G.S.; Wang, W. Epigenetic trajectory predicts development of clinical rheumatoid arthritis in ACPA+ individuals: Targeting Immune Responses for Prevention of Rheumatoid Arthritis (TIP-RA). bioRxiv 2025. [Google Scholar] [CrossRef]
  67. Hageman, I.; Mol, F.; Atiqi, S.; Joustra, V.; Sengul, H.; Henneman, P.; Visman, I.; Hakvoort, T.; Nurmohamed, M.; Wolbink, G.; et al. Novel DNA methylome biomarkers associated with adalimumab response in rheumatoid arthritis patients. Front. Immunol. 2023, 14, 1303231. [Google Scholar] [CrossRef]
  68. de la Calle-Fabregat, C.; Niemantsverdriet, E.; Cañete, J.D.; Li, T.; van der Helm-van Mil, A.H.M.; Rodríguez-Ubreva, J.; Ballestar, E. Prediction of the Progression of Undifferentiated Arthritis to Rheumatoid Arthritis Using DNA Methylation Profiling. Arthritis Rheumatol. 2021, 73, 2229–2239. [Google Scholar] [CrossRef]
  69. Chen, Y.; Wang, Q.; Liu, H.; Jin, L.; Feng, X.; Dai, B.; Chen, M.; Xin, F.; Wei, T.; Bai, B.; et al. The prognostic value of whole-genome DNA methylation in response to Leflunomide in patients with Rheumatoid Arthritis. Front. Immunol. 2023, 14, 1173187. [Google Scholar] [CrossRef]
  70. Feng, X.; Hao, X.; Shi, R.; Xia, Z.; Huang, L.; Yu, Q.; Zhou, F. Detection and Comparative Analysis of Methylomic Biomarkers of Rheumatoid Arthritis. Front. Genet. 2020, 11, 238. [Google Scholar] [CrossRef]
  71. Zhao, J.; He, B.; Shan, Y.; Wei, K.; Jiang, P.; Shi, Y.; Chang, C.; Zheng, Y.; Zhao, F.; Li, Y.; et al. Identifying AIM2 Circulating Methylation Levels as a Novel Diagnostic Biomarker for Rheumatoid Arthritis Using Targeted DNA Methylation Sequencing. Endocr. Metab. Immune Disord. Drug Targets 2025. [Google Scholar] [CrossRef] [PubMed]
  72. Jiang, P.; Wei, K.; Xu, L.; Chang, C.; Zhang, R.; Zhao, J.; Jin, Y.; Xu, L.; Shi, Y.; Qian, Y.; et al. DNA methylation change of HIPK3 in Chinese rheumatoid arthritis and its effect on inflammation. Front. Immunol. 2022, 13, 1087279. [Google Scholar] [CrossRef] [PubMed]
  73. Jiang, P.; Shen, Y.; Chang, C.; Shi, Y.; Wei, K.; Zhao, J.; Shan, Y.; Zheng, Y.; Zhao, F.; Guo, S.; et al. HIPK3 hypomethylation as a potential epigenetic biomarker in rheumatic immune diseases with emphasis on rheumatoid arthritis. Clin. Rheumatol. 2025, 44, 115–123. [Google Scholar] [CrossRef] [PubMed]
  74. Tessarz, P.; Kouzarides, T. Histone core modifications regulating nucleosome structure and dynamics. Nat. Rev. Mol. Cell Biol. 2014, 15, 703–708. [Google Scholar] [CrossRef]
  75. Ntougkos, E.; Chouvardas, P.; Roumelioti, F.; Ospelt, C.; Frank-Bertoncelj, M.; Filer, A.; Buckley, C.D.; Gay, S.; Nikolaou, C.; Kollias, G. Genomic Responses of Mouse Synovial Fibroblasts During Tumor Necrosis Factor-Driven Arthritogenesis Greatly Mimic Those in Human Rheumatoid Arthritis. Arthritis Rheumatol. 2017, 69, 1588–1600. [Google Scholar] [CrossRef]
  76. Meng, W.; Zhu, Z.; Jiang, X.; Too, C.L.; Uebe, S.; Jagodic, M.; Kockum, I.; Murad, S.; Ferrucci, L.; Alfredsson, L.; et al. DNA methylation mediates genotype and smoking interaction in the development of anti-citrullinated peptide antibody-positive rheumatoid arthritis. Arthritis Res. Ther. 2017, 19, 71. [Google Scholar] [CrossRef]
  77. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
  78. Campbell, C.; McKenney, P.T.; Konstantinovsky, D.; Isaeva, O.I.; Schizas, M.; Verter, J.; Mai, C.; Jin, W.B.; Guo, C.J.; Violante, S.; et al. Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 2020, 581, 475–479. [Google Scholar] [CrossRef]
  79. Julià, A.; Gómez, A.; López-Lasanta, M.; Blanco, F.; Erra, A.; Fernández-Nebro, A.; Mas, A.J.; Pérez-García, C.; Vivar, M.L.G.; Sánchez-Fernández, S.; et al. Longitudinal analysis of blood DNA methylation identifies mechanisms of response to tumor necrosis factor inhibitor therapy in rheumatoid arthritis. eBioMedicine 2022, 80, 104053. [Google Scholar] [CrossRef]
  80. Simard, J.F.; Costenbader, K.H.; Hernán, M.A.; Liang, M.H.; Mittleman, M.A.; Karlson, E.W. Early life factors and adult-onset rheumatoid arthritis. J. Rheumatol. 2010, 37, 32–37. [Google Scholar] [CrossRef]
  81. Barik, R.R.; Bhatt, L.K. Emerging epigenetic targets in rheumatoid arthritis. Rheumatol. Int. 2021, 41, 2047–2067. [Google Scholar] [CrossRef]
  82. Chang, C.; Xu, L.; Zhang, R.; Jin, Y.; Jiang, P.; Wei, K.; Xu, L.; Shi, Y.; Zhao, J.; Xiong, M.; et al. MicroRNA-Mediated Epigenetic Regulation of Rheumatoid Arthritis Susceptibility and Pathogenesis. Front. Immunol. 2022, 13, 838884. [Google Scholar] [CrossRef]
  83. Kang, L.; Dai, C.; Wang, L.; Pan, X. Potential biomarkers that discriminate rheumatoid arthritis and osteoarthritis based on the analysis and validation of datasets. BMC Musculoskelet. Disord. 2022, 23, 319. [Google Scholar] [CrossRef] [PubMed]
  84. Mun, S.; Lee, J.; Park, A.; Kim, H.J.; Lee, Y.J.; Son, H.; Shin, M.; Lim, M.K.; Kang, H.G. Proteomics Approach for the Discovery of Rheumatoid Arthritis Biomarkers Using Mass Spectrometry. Int. J. Mol. Sci. 2019, 20, 4368. [Google Scholar] [CrossRef] [PubMed]
  85. Turkkila, M.; Andersson, K.M.; Amu, S.; Brisslert, M.; Erlandsson, M.C.; Silfverswärd, S.; Bokarewa, M.I. Suppressed diversity of survivin splicing in active rheumatoid arthritis. Arthritis Res. Ther. 2015, 17, 175. [Google Scholar] [CrossRef]
  86. Pathania, A.S. Crosstalk between Noncoding RNAs and the Epigenetics Machinery in Pediatric Tumors and Their Microenvironment. Cancers 2023, 15, 2833. [Google Scholar] [CrossRef]
  87. Tofigh, R.; Hosseinpourfeizi, M.; Baradaran, B.; Teimourian, S.; Safaralizadeh, R. Rheumatoid arthritis and non-coding RNAs; how to trigger inflammation. Life Sci. 2023, 315, 121367. [Google Scholar] [CrossRef]
  88. Wang, J.; Yan, S.; Yang, J.; Lu, H.; Xu, D.; Wang, Z. Non-coding RNAs in Rheumatoid Arthritis: From Bench to Bedside. Front. Immunol. 2019, 10, 3129. [Google Scholar] [CrossRef]
  89. Zhang, Y.; Yang, M.; Xie, H.; Hong, F.; Yang, S. Role of miRNAs in Rheumatoid Arthritis Therapy. Cells 2023, 12, 1749. [Google Scholar] [CrossRef]
  90. Zhao, X.; Lin, W.; Zhou, W. Clinical significance of long non-coding RNA NORAD in rheumatoid arthritis. Adv. Rheumatol. 2024, 64, 9. [Google Scholar] [CrossRef]
  91. Mattick, J.S.; Amaral, P.P.; Carninci, P.; Carpenter, S.; Chang, H.Y.; Chen, L.-L.; Chen, R.; Dean, C.; Dinger, M.E.; Fitzgerald, K.A. Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat. Rev. Mol. Cell Biol. 2023, 24, 430–447. [Google Scholar] [CrossRef] [PubMed]
  92. Jiang, Z.; Zhong, Z.; Miao, Q.; Zhang, Y.; Ni, B.; Zhang, M.; Tang, J. circPTPN22 as a novel biomarker and ceRNA in peripheral blood mononuclear cells of rheumatoid arthritis. Mol. Med. Rep. 2021, 24, 617. [Google Scholar] [CrossRef] [PubMed]
  93. Zhao, R.-J.; Zhang, W.-Y.; Fan, X.-X. Circular RNAs: Potential biomarkers and therapeutic targets for autoimmune diseases. Heliyon 2024, 10, e23694. [Google Scholar] [CrossRef] [PubMed]
  94. Barrett, S.P.; Salzman, J. Circular RNAs: Analysis, expression and potential functions. Development 2016, 143, 1838–1847. [Google Scholar] [CrossRef]
  95. Oliver, J.; Nair, N.; Orozco, G.; Smith, S.; Hyrich, K.L.; Morgan, A.; Isaacs, J.; Wilson, A.G.; Barton, A.; Plant, D. Transcriptome-wide study of TNF-inhibitor therapy in rheumatoid arthritis reveals early signature of successful treatment. Arthritis Res. Ther. 2021, 23, 80, Erratum in Arthritis Res. Ther. 2021, 23, 139. [Google Scholar] [CrossRef]
  96. Al-Amrani, S.; Al-Jabri, Z.; Al-Zaabi, A.; Alshekaili, J.; Al-Khabori, M. Proteomics: Concepts and applications in human medicine. World J. Biol. Chem. 2021, 12, 57–69. [Google Scholar] [CrossRef]
  97. Song, X.; Lin, Q. Genomics, transcriptomics and proteomics to elucidate the pathogenesis of rheumatoid arthritis. Rheumatol. Int. 2017, 37, 1257–1265. [Google Scholar] [CrossRef]
  98. Burska, A.; Boissinot, M.; Ponchel, F. Cytokines as biomarkers in rheumatoid arthritis. Mediat. Inflamm. 2014, 2014, 545493. [Google Scholar] [CrossRef]
  99. Myngbay, A.; Manarbek, L.; Ludbrook, S.; Kunz, J. The Role of Collagen Triple Helix Repeat-Containing 1 Protein (CTHRC1) in Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 2426. [Google Scholar] [CrossRef]
  100. Nassef, E.M.; Elabd, H.A.; Elzomor, H.M.; El Nagger, B.; Ibrahim, A.S.; Ibrahim, A.H.; Kotb, H.G.; Hassan, D.A.; Abd ElAziz, R.E.M.; Mohamed, E.E.S. Serum Collagen Triple Helix Repeat Containing-1 Levels are Related to Radiological Affection and Disease Activity in Rheumatoid Arthritis. Open Access Rheumatol. 2022, 14, 291–299. [Google Scholar] [CrossRef]
  101. Tan, L.; Ouyang, T.; Li, X.; Chen, Y.; Ke, S.; Chen, J.; Liu, Y.; Zeng, F.; Chen, Y.; Long, J.; et al. Serum sirtuin-1 a potential marker in the diagnosis of rheumatoid arthritis. Autoimmunity 2023, 56, 2181234. [Google Scholar] [CrossRef] [PubMed]
  102. Hu, C.; Dai, Z.; Xu, J.; Zhao, L.; Xu, Y.; Li, M.; Yu, J.; Zhang, L.; Deng, H.; Liu, L.; et al. Proteome Profiling Identifies Serum Biomarkers in Rheumatoid Arthritis. Front. Immunol. 2022, 13, 865425. [Google Scholar] [CrossRef] [PubMed]
  103. Ermencheva, P.; Kotov, G.; Shumnalieva, R.; Velikova, T.; Monov, S. Exploring the Role of the Microbiome in Rheumatoid Arthritis—A Critical Review. Microorganisms 2024, 12, 1387. [Google Scholar] [CrossRef] [PubMed]
  104. Hur, B.; Gupta, V.K.; Huang, H.; Wright, K.A.; Warrington, K.J.; Taneja, V.; Davis, J.M., III; Sung, J. Plasma metabolomic profiling in patients with rheumatoid arthritis identifies biochemical features predictive of quantitative disease activity. Arthritis Res. Ther. 2021, 23, 164. [Google Scholar] [CrossRef]
  105. Tsetseri, M.-N.; Silman, A.J.; Keene, D.J.; Dakin, S.G. The role of the microbiome in rheumatoid arthritis: A review. Rheumatol. Adv. Pract. 2023, 7, rkad034. [Google Scholar] [CrossRef]
  106. Zhou, D.; Jiao, W.; Shi, W.; Wang, Q.; Chen, M. Mendelian randomization identifies causal associations between GWAS-associated bacteria and their metabolites and rheumatoid arthritis. Front. Microbiol. 2024, 15, 1431367. [Google Scholar] [CrossRef]
  107. Yun, H.; Wang, X.; Wei, C.; Liu, Q.; Li, X.; Li, N.; Zhang, G.; Cui, D.; Liu, R. Alterations of the intestinal microbiome and metabolome in women with rheumatoid arthritis. Clin. Exp. Med. 2023, 23, 4695–4706. [Google Scholar] [CrossRef]
  108. Yang, X.Y.; Zheng, K.D.; Lin, K.; Zheng, G.; Zou, H.; Wang, J.M.; Lin, Y.Y.; Chuka, C.M.; Ge, R.S.; Zhai, W.; et al. Energy Metabolism Disorder as a Contributing Factor of Rheumatoid Arthritis: A Comparative Proteomic and Metabolomic Study. PLoS ONE 2015, 10, e0132695. [Google Scholar] [CrossRef]
  109. Chu, C.Q. The bacterial link with rheumatoid arthritis. Int. J. Rheum. Dis. 2023, 26, 603–604. [Google Scholar] [CrossRef]
  110. Dong, Y.; Yao, J.; Deng, Q.; Li, X.; He, Y.; Ren, X.; Zheng, Y.; Song, R.; Zhong, X.; Ma, J. Relationship between gut microbiota and rheumatoid arthritis: A bibliometric analysis. Front. Immunol. 2023, 14, 1131933. [Google Scholar] [CrossRef]
  111. Luo, Y.; Tong, Y.; Wu, L.; Niu, H.; Li, Y.; Su, L.C.; Wu, Y.; Bozec, A.; Zaiss, M.M.; Qing, P. Alteration of gut microbiota in individuals at high-risk for rheumatoid arthritis associated with disturbed metabolome and the initiation of arthritis through the triggering of mucosal immunity imbalance. Arthritis Rheumatol. 2023, 75, 1736–1748. [Google Scholar] [CrossRef]
  112. Scher, J.U.; Sczesnak, A.; Longman, R.S.; Segata, N.; Ubeda, C.; Bielski, C.; Rostron, T.; Cerundolo, V.; Pamer, E.G.; Abramson, S.B.; et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. eLife 2013, 2, e01202. [Google Scholar] [CrossRef]
  113. Maeda, Y.; Kurakawa, T.; Umemoto, E.; Motooka, D.; Ito, Y.; Gotoh, K.; Hirota, K.; Matsushita, M.; Furuta, Y.; Narazaki, M.; et al. Dysbiosis Contributes to Arthritis Development via Activation of Autoreactive T Cells in the Intestine. Arthritis Rheumatol. 2016, 68, 2646–2661. [Google Scholar] [CrossRef]
  114. Pianta, A.; Arvikar, S.; Strle, K.; Drouin, E.E.; Wang, Q.; Costello, C.E.; Steere, A.C. Evidence of the Immune Relevance of Prevotella copri, a Gut Microbe, in Patients With Rheumatoid Arthritis. Arthritis Rheumatol. 2017, 69, 964–975. [Google Scholar] [CrossRef] [PubMed]
  115. Ottman, N.; Reunanen, J.; Meijerink, M.; Pietilä, T.E.; Kainulainen, V.; Klievink, J.; Huuskonen, L.; Aalvink, S.; Skurnik, M.; Boeren, S.; et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS ONE 2017, 12, e0173004. [Google Scholar] [CrossRef] [PubMed]
  116. Stoll, M.L.; Pierce, M.K.; Watkins, J.A.; Zhang, M.; Weiss, P.F.; Weiss, J.E.; Elson, C.O.; Cron, R.Q.; Kumar, R.; Morrow, C.D.; et al. Akkermansia muciniphila is permissive to arthritis in the K/BxN mouse model of arthritis. Genes Immun. 2019, 20, 158–166. [Google Scholar] [CrossRef]
  117. Romero-Figueroa, M.D.S.; Ramírez-Durán, N.; Montiel-Jarquín, A.J.; Horta-Baas, G. Gut-joint axis: Gut dysbiosis can contribute to the onset of rheumatoid arthritis via multiple pathways. Front. Cell. Infect. Microbiol. 2023, 13, 1092118. [Google Scholar] [CrossRef]
  118. Lin, L.; Zhang, K.; Xiong, Q.; Zhang, J.; Cai, B.; Huang, Z.; Yang, B.; Wei, B.; Chen, J.; Niu, Q. Gut microbiota in pre-clinical rheumatoid arthritis: From pathogenesis to preventing progression. J. Autoimmun. 2023, 141, 103001. [Google Scholar] [CrossRef]
  119. Yang, Y.; Hong, Q.; Zhang, X.; Liu, Z. Rheumatoid arthritis and the intestinal microbiome: Probiotics as a potential therapy. Front. Immunol. 2024, 15, 1331486. [Google Scholar] [CrossRef]
  120. Ge, Y.; Zhou, L.; Chen, Z.; Mao, Y.; Li, T.; Tong, P.; Shan, L. Identification of differentially expressed genes, signaling pathways and immune infiltration in rheumatoid arthritis by integrated bioinformatics analysis. Hereditas 2021, 158, 5. [Google Scholar] [CrossRef]
  121. Zhang, R.; Zhou, X.; Jin, Y.; Chang, C.; Wang, R.; Liu, J.; Fan, J.; He, D. Identification of differential key biomarkers in the synovial tissue between rheumatoid arthritis and osteoarthritis using bioinformatics analysis. Clin. Rheumatol. 2021, 40, 5103–5110. [Google Scholar] [CrossRef]
  122. Chen, H.; Xu, J.; Wei, S.; Jia, Z.; Sun, C.; Kang, J.; Guo, X.; Zhang, N.; Tao, J.; Dong, Y.; et al. RABC: Rheumatoid Arthritis Bioinformatics Center. Nucleic Acids Res. 2023, 51, D1381–D1387. [Google Scholar] [CrossRef]
  123. Sun, F.; Zhou, J.L.; Peng, P.J.; Qiu, C.; Cao, J.R.; Peng, H. Identification of Disease-Specific Hub Biomarkers and Immune Infiltration in Osteoarthritis and Rheumatoid Arthritis Synovial Tissues by Bioinformatics Analysis. Dis. Markers 2021, 2021, 9911184. [Google Scholar] [CrossRef] [PubMed]
  124. Luo, S.F.; Fang, R.Y.; Hsieh, H.L.; Chi, P.L.; Lin, C.C.; Hsiao, L.D.; Wu, C.C.; Wang, J.S.; Yang, C.M. Involvement of MAPKs and NF-kappaB in tumor necrosis factor alpha-induced vascular cell adhesion molecule 1 expression in human rheumatoid arthritis synovial fibroblasts. Arthritis Rheum. 2010, 62, 105–116. [Google Scholar] [CrossRef] [PubMed]
  125. Aterido, A.; Cañete, J.D.; Tornero, J.; Blanco, F.; Fernández-Gutierrez, B.; Pérez, C.; Alperi-López, M.; Olivè, A.; Corominas, H.; Martínez-Taboada, V.; et al. A Combined Transcriptomic and Genomic Analysis Identifies a Gene Signature Associated With the Response to Anti-TNF Therapy in Rheumatoid Arthritis. Front. Immunol. 2019, 10, 1459. [Google Scholar] [CrossRef] [PubMed]
  126. Kang, M.; Ko, E.; Mersha, T.B. A roadmap for multi-omics data integration using deep learning. Brief. Bioinform. 2022, 23, bbab454. [Google Scholar] [CrossRef]
  127. Subramanian, I.; Verma, S.; Kumar, S.; Jere, A.; Anamika, K. Multi-omics Data Integration, Interpretation, and Its Application. Bioinform. Biol. Insights 2020, 14, 1177932219899051. [Google Scholar] [CrossRef]
  128. Tao, W.; Concepcion, A.N.; Vianen, M.; Marijnissen, A.C.A.; Lafeber, F.; Radstake, T.; Pandit, A. Multiomics and Machine Learning Accurately Predict Clinical Response to Adalimumab and Etanercept Therapy in Patients with Rheumatoid Arthritis. Arthritis Rheumatol. 2021, 73, 212–222. [Google Scholar] [CrossRef]
  129. Sussulini, A.; Xia, J.; Orešič, M. Editorial: Multi-omics: Trends and applications in clinical research. Front. Mol. Biosci. 2022, 9, 994239. [Google Scholar] [CrossRef]
  130. Westerlind, H.; Maciejewski, M.; Frisell, T.; Jelinsky, S.A.; Ziemek, D.; Askling, J. What Is the Persistence to Methotrexate in Rheumatoid Arthritis, and Does Machine Learning Outperform Hypothesis-Based Approaches to Its Prediction? ACR Open Rheumatol. 2021, 3, 457–463. [Google Scholar] [CrossRef]
  131. Chang, M.J.; Feng, Q.F.; Hao, J.W.; Zhang, Y.J.; Zhao, R.; Li, N.; Zhao, Y.H.; Han, Z.Y.; He, P.F.; Wang, C.H. Deciphering the molecular landscape of rheumatoid arthritis offers new insights into the stratified treatment for the condition. Front. Immunol. 2024, 15, 1391848. [Google Scholar] [CrossRef]
  132. Jung, S.M.; Park, K.S.; Kim, K.J. Deep phenotyping of synovial molecular signatures by integrative systems analysis in rheumatoid arthritis. Rheumatology 2021, 60, 3420–3431. [Google Scholar] [CrossRef]
  133. Hur, B.; Cunningham, K.; Davis, J.; Sung, J. Integrative multi-omic profiling in blood reveals distinct immune and metabolic signatures between ACPA-negative and ACPA-positive rheumatoid arthritis. Front. Immunol. 2025, 16. [Google Scholar] [CrossRef]
  134. Yoosuf, N.; Maciejewski, M.; Ziemek, D.; Jelinsky, S.A.; Folkersen, L.; Müller, M.; Sahlström, P.; Vivar, N.; Catrina, A.; Berg, L.; et al. Early prediction of clinical response to anti-TNF treatment using multi-omics and machine learning in rheumatoid arthritis. Rheumatology 2022, 61, 1680–1689. [Google Scholar] [CrossRef] [PubMed]
  135. Kerle, I.A.; Gross, T.; Kögler, A.; Arnold, J.S.; Werner, M.; Eckardt, J.N.; Möhrmann, E.E.; Arlt, M.; Hutter, B.; Hüllein, J.; et al. Translational and clinical comparison of whole genome and transcriptome to panel sequencing in precision oncology. NPJ Precis. Oncol. 2025, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  136. Kelly, S.; Humby, F.; Filer, A.; Ng, N.; Di Cicco, M.; Hands, R.E.; Rocher, V.; Bombardieri, M.; D’Agostino, M.A.; McInnes, I.B.; et al. Ultrasound-guided synovial biopsy: A safe, well-tolerated and reliable technique for obtaining high-quality synovial tissue from both large and small joints in early arthritis patients. Ann. Rheum. Dis. 2015, 74, 611–617. [Google Scholar] [CrossRef]
  137. Lliso-Ribera, G.; Humby, F.; Lewis, M.; Nerviani, A.; Mauro, D.; Rivellese, F.; Kelly, S.; Hands, R.; Bene, F.; Ramamoorthi, N.; et al. Synovial tissue signatures enhance clinical classification and prognostic/treatment response algorithms in early inflammatory arthritis and predict requirement for subsequent biological therapy: Results from the pathobiology of early arthritis cohort (PEAC). Ann. Rheum. Dis. 2019, 78, 1642–1652. [Google Scholar] [CrossRef]
  138. Nerviani, A.; Di Cicco, M.; Mahto, A.; Lliso-Ribera, G.; Rivellese, F.; Thorborn, G.; Hands, R.; Bellan, M.; Mauro, D.; Boutet, M.A.; et al. A Pauci-Immune Synovial Pathotype Predicts Inadequate Response to TNFα-Blockade in Rheumatoid Arthritis Patients. Front. Immunol. 2020, 11, 845. [Google Scholar] [CrossRef]
  139. Humby, F.; Durez, P.; Buch, M.H.; Lewis, M.J.; Rizvi, H.; Rivellese, F.; Nerviani, A.; Giorli, G.; Mahto, A.; Montecucco, C.; et al. Rituximab versus tocilizumab in anti-TNF inadequate responder patients with rheumatoid arthritis (R4RA): 16-week outcomes of a stratified, biopsy-driven, multicentre, open-label, phase 4 randomised controlled trial. Lancet 2021, 397, 305–317. [Google Scholar] [CrossRef]
  140. Rivellese, F.; Surace, A.E.A.; Goldmann, K.; Sciacca, E.; Çubuk, C.; Giorli, G.; John, C.R.; Nerviani, A.; Fossati-Jimack, L.; Thorborn, G.; et al. Rituximab versus tocilizumab in rheumatoid arthritis: Synovial biopsy-based biomarker analysis of the phase 4 R4RA randomized trial. Nat. Med. 2022, 28, 1256–1268. [Google Scholar] [CrossRef]
  141. Zhang, F.; Wei, K.; Slowikowski, K.; Fonseka, C.Y.; Rao, D.A.; Kelly, S.; Goodman, S.M.; Tabechian, D.; Hughes, L.B.; Salomon-Escoto, K.; et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol. 2019, 20, 928–942. [Google Scholar] [CrossRef]
  142. Zhang, F.; Jonsson, A.H.; Nathan, A.; Millard, N.; Curtis, M.; Xiao, Q.; Gutierrez-Arcelus, M.; Apruzzese, W.; Watts, G.F.M.; Weisenfeld, D.; et al. Deconstruction of rheumatoid arthritis synovium defines inflammatory subtypes. Nature 2023, 623, 616–624. [Google Scholar] [CrossRef]
  143. Julià, A.; Ávila, G.; Celis, R.; Sanmartí, R.; Ramírez, J.; Marsal, S.; Cañete, J.D. Lower peripheral helper T cell levels in the synovium are associated with a better response to anti-TNF therapy in rheumatoid arthritis. Arthritis Res. Ther. 2020, 22, 196. [Google Scholar] [CrossRef]
  144. Bo, L.; Jin, X.; Hu, Y.; Yang, R. Role of Liquid Biopsies in Rheumatoid Arthritis. Methods Mol. Biol. 2023, 2695, 237–246. [Google Scholar]
  145. Zerrouk, N.; Augé, F.; Niarakis, A. Building a modular and multi-cellular virtual twin of the synovial joint in Rheumatoid Arthritis. NPJ Digit. Med. 2024, 7, 379. [Google Scholar] [CrossRef]
Figure 1. The diagram of biomarker discovery and biomarker application in rheumatoid arthritis.
Figure 1. The diagram of biomarker discovery and biomarker application in rheumatoid arthritis.
Ijms 26 10594 g001
Table 1. The key autoantibodies in rheumatoid arthritis (RA), along with their sensitivity, specificity, prevalence in other conditions, clinical applications, and typical algorithms used.
Table 1. The key autoantibodies in rheumatoid arthritis (RA), along with their sensitivity, specificity, prevalence in other conditions, clinical applications, and typical algorithms used.
AutoantibodiesSensitivitySpecificityPresence in Other ConditionsClinical Application in RATypical Algorithms Used
Rheumatoid Factor 60–80%70–80%Healthy individuals,
Sjögren’s syndrome, lupus,
chronic infections
Diagnostic and prognostic marker, prediction of treatment responses to rituximab, sarilumab, tofacitinibEnsemble Tree-Based Models, Multivariate Logistic Regression, Feature Importance Algorithms, ACR/EULAR Classification Criteria (2010)
Anti-Citrullinated Protein Antibodies 60–70%>95%Rarely in other conditionsDiagnostic and prognostic marker, prediction of treatment responses to rituximab, sarilumab, tofacitinibClassification Algorithms, Receiver Operating Characteristic (ROC) Analysis, Random Forest, Feedforward Neural Networks, ACR/EULAR Classification Criteria (2010)
Anti-Carbamylated Protein Antibodies35–50%~90%Rarely in other conditionsUseful for the diagnosis of seronegative RA, associated with a more aggressive diseaseMultivariate Logistic Regression, Meta-Analysis.
Anti-Peptidyl Arginine Deiminase 4 20–30%>90%Rarely in other conditionsAssociated with a more severe and erosive disease; may predict the need for biologic treatmentsROC Analysis, Correlation Analysis, Simple Classification Algorithms
Anti-Sa Antibodies20–30%>95%Rarely in other conditionsPrediction of disease severity and joint erosionsMultivariate Regression Models, Correlation Analysis
Anti-Mutated Citrullinated Vimentin antibodies70–80%85–95%Rarely in other conditionsHighly useful for the diagnosis of seronegative RA, associated with disease activity and severityROC Analysis, Classification Algorithms
Anti-Nuclear Antibodies20–30%30–50%Lupus, Sjögren’s syndrome, SclerodermaThe presence may indicate an overlap with another autoimmune diseaseArtificial Intelligence /Machine Learning models
Anti-Ro/SSA Antibodies3–15%30–50%Sjögren’s syndrome, Lupus, Systemic sclerosisThe presence may indicate an overlap with Sjögren’s syndrome Clustering Algorithms
Anti-La/SSB Antibodies3–10%30–50%Sjögren’s syndrome, Lupus, Systemic sclerosisThe presence usually indicates an overlap with Sjögren’s syndromeClustering Algorithms
Table 2. Key inflammatory biomarkers in rheumatoid arthritis (RA), including their sensitivity, specificity, presence in other conditions, clinical applications, and typical algorithms used.
Table 2. Key inflammatory biomarkers in rheumatoid arthritis (RA), including their sensitivity, specificity, presence in other conditions, clinical applications, and typical algorithms used.
Inflammatory BiomarkersSensitivitySpecificityPresence in Other ConditionsClinical Application in RATypical Algorithm
CRP40–60%~40%Elevated in a wide range of inflammatory, infectious, and tissue-damaging conditionsA key component of disease activity scores, monitoring disease activity and treatment response DAS28-CRP, ACR/EULAR Classification Criteria (2010)
ESR40–60%~40%Elevated in many inflammatory and infectious conditions, and certain cancersA key component of disease activity scores, monitoring disease activity and treatment responseDAS28-ESR, ACR/EULAR Classification Criteria (2010)
SAA40–60%40–60%Elevated in various inflammatory conditionsMonitoring disease activity, predicting treatment responseMulti-Biomarker Disease Activity (MBDA) Score
MMP-340–80%50–70%Osteoarthritis, other joint diseases, and lupusA marker of joint destruction and cartilage breakdownMBDA Score
Calprotectin60–80%~90%Inflammatory bowel disease, psoriatic arthritis, and infectionsCorrelated with disease activity, predicting a poor radiographic outcomeReceiver Operating Characteristic (ROC) Analysis, MBDA Scores
14-3-3η60–70%80–90%Osteoporosis, other autoimmune diseasesEarly diagnostic and prognostic marker, predicting a more erosive diseaseROC Analysis
TNF-α40–70%40–60%Various autoimmune /inflammatory diseasesA key therapeutic target, monitoring disease activityMBDA Score
IL-640–70%40–60%Various autoimmune/inflammatory diseasesA key therapeutic target, monitoring disease activityMBDA Score
CRP: C-reactive protein. ESR: Erythrocyte sedimentation rate. IL: Interleukin. MMP: Matrix Metalloproteinase. SAA: Serum amyloid A. TNF: Tumor necrosis factor.
Table 3. Key genetic biomarkers in rheumatoid arthritis (RA), including their sensitivity, specificity, presence in other conditions, clinical applications, and typical algorithms used.
Table 3. Key genetic biomarkers in rheumatoid arthritis (RA), including their sensitivity, specificity, presence in other conditions, clinical applications, and typical algorithms used.
Genetic BiomarkersSensitivitySpecificityPresence in Other ConditionsClinical Application in RATypical Algorithm
HLA-DRB180–90%60–70%A key shared risk factor for multiple autoimmune diseases May be used for risk assessment and early diagnosisPolygenic Risk Scores (PRS) Models
PTPN2250–60%60–80%A key shared risk factor for multiple autoimmune diseases Potential use in predicting disease risk and tailoring treatmentMachine learning (ML) Models (e.g., Logistic Regression, Decision Trees, XGBoost)
STAT450–70%40–60%Associated with a variety of autoimmune and inflammatory conditionsLimited clinical application, could be a potential target for new therapiesPRS / ML Models
TRAF1/C540–60%40–60%Associated with a variety of autoimmune and inflammatory conditionsNot used in clinical practice for diagnosis or prognosisPRS Models
PADI4~70%40–60%Rarely in other conditionsPotential use for predicting a more aggressive and erosive diseaseML Models
TNFAIP340–60%40–60%Associated with numerous other autoimmune conditionsNot used in clinical practice for diagnosis or prognosisML Models
IL-2RA40–50%40–50%Associated with numerous other autoimmune conditionsNot used in clinical practice for diagnosis or prognosisML Models
CD4040–60%40–60%Associated with numerous other autoimmune conditionsNot used in clinical practice for diagnosis or prognosisPRS Models
CTLA440–70%40–60%A well-known risk factor for numerous autoimmune diseasesNot used in clinical practice for diagnosis or prognosisML Models
Table 4. Key metabolite biomarkers in rheumatoid arthritis (RA), along with their sensitivity, specificity, presence in other conditions, potential clinical applications, and typical algorithms.
Table 4. Key metabolite biomarkers in rheumatoid arthritis (RA), along with their sensitivity, specificity, presence in other conditions, potential clinical applications, and typical algorithms.
Metabolite BiomarkersSensitivitySpecificityPresence in Other Conditions Clinical Application in RATypical Algorithm Used
Glyceric AcidLimited data availableLimited data availableGlyceric Aciduria, some cardiovascular diseasesA potential marker for disease activityMachine Learning (ML) Classifiers (e.g., Random Forest, Logistic Regression)
Lactic Acid30–40% 30–50% Elevated in a wide range of conditionsA general marker for increased tissue inflammationStatistical Analysis (Correlation/Regression), ML
3-Hydroxyisovaleric AcidLimited data availableLimited data availableLeucine deficiency and other metabolic disordersNot used clinically for diagnosis or monitoring.ML Classifiers
Angiotensinogen40–60% 40–60% Elevated in hypertension and metabolic syndromeA potential diagnostic marker for seronegative RAStatistical Analysis, ML
Serum Amyloid A-4 ProteinLimited data availableLimited data availableElevated in various inflammatory conditionsA potential prescreening marker when used in combination with other markers Statistical Analysis, ML
Vitamin D-Binding ProteinLimited data availableLimited data availableLiver disease, kidney disease, and sepsisA component of a multi-biomarker panel for the diagnosis of seronegative RAStatistical Analysis, ML
Retinol-Binding Protein-440–60%40–60%Metabolic syndrome and cardiovascular diseasesA potential component of a diagnostic panel for seronegative RAStatistical Analysis, ML
Table 5. Key microbiota biomarkers in rheumatoid arthritis (RA), along with their sensitivity, specificity, observed changes in RA and other conditions, potential clinical applications, and typical algorithms used.
Table 5. Key microbiota biomarkers in rheumatoid arthritis (RA), along with their sensitivity, specificity, observed changes in RA and other conditions, potential clinical applications, and typical algorithms used.
Microbiota BiomarkersSensitivitySpecificityChanges in RAChanges in Other Conditions Clinical Application for RATypical Algorithm Used
Prevotella copri70%~70% Inflammatory bowel disease, psoriatic arthritis, and other autoimmune diseasesA diagnostic marker for
new-onset RA, predict response to MTX therapy
Statistical Analysis, Linear Discriminant Analysis, Effect Size, Machine Learning (ML) Classifiers (Random Forest)
Collinsella30–50%30–50%Psoriasis, ankylosing spondylitis, and other spondyloarthropathiesAssociated with high ACPA levels, used to understand pathogenesis Differential Abundance Analysis (DAA), Correlation Analysis
LactobacillusVariesVariesInflammatory bowel disease, metabolic disorders, allergies, and cardiovascular diseaseCan be used for potential probiotic interventionsDAA, Aitchison Distance (Beta-diversity)
Bacteroides20–50%20–50%Obesity, diabetes, and Inflammatory bowel diseaseA component of a predictive model, associated with a poorer response to MTXStatistical Analysis, Regression Models
Faecalibacterium20–50%20–50%Inflammatory bowel diseases and chronic fatigue syndromeA general marker of dysbiosis, a potential probiotic treatment targetStatistical Analysis, Functional Prediction Tools (e.g., PICRUSt or Tax4Fun)
Eggerthellales20–50%20–50%Some species are associated with gut infections and inflammationA potential marker for disease severity and a potential probiotic treatment targetDAA, Correlation Analysis
Enterococcus20–50%20–50%A wide range of infections, including urinary tract infections The general decrease is a marker of dysbiosisDAA, Principal Coordinate Analysis/Non-metric Multidimensional Scaling
Bifidobacterium species20–50%20–50%Depleted in various inflammatory and metabolic diseasesMonitoring gut health and potential probiotic treatmentsStatistical Analysis, Correlation Analysis
MTX: methotrexate; ACPA: anti-citrullinated protein antibodies. indicates decrease and increase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xue, M.; Wang, H.; Campos, F.; Jackson, C.J.; March, L. Rheumatoid Arthritis: Biomarkers and the Latest Breakthroughs. Int. J. Mol. Sci. 2025, 26, 10594. https://doi.org/10.3390/ijms262110594

AMA Style

Xue M, Wang H, Campos F, Jackson CJ, March L. Rheumatoid Arthritis: Biomarkers and the Latest Breakthroughs. International Journal of Molecular Sciences. 2025; 26(21):10594. https://doi.org/10.3390/ijms262110594

Chicago/Turabian Style

Xue, Meilang, Hui Wang, Frida Campos, Christopher J. Jackson, and Lyn March. 2025. "Rheumatoid Arthritis: Biomarkers and the Latest Breakthroughs" International Journal of Molecular Sciences 26, no. 21: 10594. https://doi.org/10.3390/ijms262110594

APA Style

Xue, M., Wang, H., Campos, F., Jackson, C. J., & March, L. (2025). Rheumatoid Arthritis: Biomarkers and the Latest Breakthroughs. International Journal of Molecular Sciences, 26(21), 10594. https://doi.org/10.3390/ijms262110594

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop