Next Article in Journal
Correlation of the FIB-4 Liver Biomarker Score with the Severity of Heart Failure
Previous Article in Journal
Lifetime Practice and Intention to Use Contraception After Induced Abortion Among Serbian Women in Belgrade
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Synovial Fluid Markers and Extracellular Vesicles in Rheumatoid Arthritis

1
Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia
2
National Institute of Rheumatic Diseases, Nábrežie Ivana Krasku 4, 921 12 Piestany, Slovakia
*
Author to whom correspondence should be addressed.
Medicina 2024, 60(12), 1945; https://doi.org/10.3390/medicina60121945
Submission received: 23 October 2024 / Revised: 19 November 2024 / Accepted: 20 November 2024 / Published: 26 November 2024
(This article belongs to the Section Hematology and Immunology)

Abstract

:
In recent years, numerous potential prognostic biomarkers for rheumatoid arthritis (RA) have been investigated. Despite these advancements, clinical practice primarily relies on autoantibody tests—for rheumatoid factor (RF) and anti-citrullinated protein antibody (anti-CCP)—alongside inflammatory markers, such as the erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP). Expanding the repertoire of diagnostic and therapeutic biomarkers is critical for improving clinical outcomes in RA. Emerging evidence highlights the significance of synovial fluid biomarkers, including aggrecan, matrix metalloproteinases, glucosyl-galactosyl-pyridinoline, hyaluronic acid, S100 proteins, calprotectin, and various cytokines, as well as immunological markers. Additionally, specific components of extracellular vesicles, such as non-coding RNAs, heat shock proteins, and lipids, are gaining attention. This review focuses on molecular markers found in synovial fluid and extracellular vesicles, excluding clinical and imaging biomarkers, and explores their potential applications in the diagnosis and management of RA.

1. Introduction

Rheumatoid arthritis (RA) is a complex, chronic autoimmune disorder that primarily targets the synovial joints but can also impact multiple organs. It progresses in distinct phases, beginning with an asymptomatic genetic risk phase, moving to a preclinical stage with detectable biomarkers such as rheumatoid factor (RF) and anti-citrullinated protein antibodies (ACPAs)—mostly anti-cyclic citrullinated peptides (anti-CCPs)—and culminating in a clinical phase where joint inflammation and symptoms become apparent. The disease manifests with pain, stiffness, swelling, and loss of joint function, which can lead to deformity and disability if untreated. Systemic symptoms, including fatigue, fever, and weight loss, may also occur, and some individuals may develop complications involving the heart, lungs, and blood vessels [1]. Early and effective intervention is essential, as it can significantly improve patient outcomes by reducing irreversible joint damage and enhancing quality of life [2]. The identification of biomarkers that can differentiate aggressive, fast-progressing RA from milder forms is becoming more and more important in clinical practice. Such biomarkers enable clinicians to initiate targeted, intensive treatment plans promptly, optimizing patient prognosis by preventing long-term complications [3]. A systematic review reported the highest mean point prevalence in North America at 1.46%, followed by Europe at 0.53% [4].
Research in the field of biological and targeted synthetic disease-modifying anti-rheumatic drugs (DMARDs) gradually increases the number of available treatments. Methotrexate, a conventional synthetic DMARD, remains the first-line treatment for most patients due to its effectiveness and familiarity in clinical settings. However, biological and targeted synthetic DMARDs, such as TNF and Janus kinase (JAK) inhibitors, have expanded therapeutic options and are increasingly used for patients with inadequate responses to traditional therapies [5]. Although these newer agents offer potent anti-inflammatory effects and can halt disease progression, they also vary widely in cost, toxicity, and patient-specific efficacy. This variability underscores the importance of robust prognostic biomarkers to guide treatment decisions, helping clinicians to balance the intensity of therapy with potential risks, particularly in early-stage RA [6].
Biomarkers play a crucial role in clinical practice by providing objective measurements that reflect biological conditions. They can be utilized to evaluate risk, confirm the presence of a disease (diagnostic), monitor the progression of a disease (prognostic), and assess both the response (predictive) and the intensity of response (pharmacodynamic) to treatment. A significant challenge in rheumatology is the difficulty in forecasting long-term outcomes for patients exhibiting early symptoms indicative of arthritis [7]. Increasing evidence suggests that implementing aggressive treatments during the early stages of arthritis is vital for reducing radiographic damage and preventing disability. The early detection of easily measurable and predictive biomarkers is essential for categorizing arthritis patients according to their disease trajectory and mitigating the risks associated with over- or undertreatment. The current biochemical assays that measure RF, an autoantibody commonly used in RA diagnosis, demonstrate a sensitivity of only approximately 63%. Research is urgently needed to discover reliable biomarkers during the pre-symptomatic and initial clinical phases of arthritis to facilitate timely and effective disease management [8,9].
This review examines the latest advancements concerning RA biomarkers sourced from synovial fluid, emphasizing their potential for diagnostic applications.

2. Synovial Fluid Markers

Understanding key joint components—synovial fluid (SF), synovial membrane, and articular cartilage—is essential to study joint diseases effectively. The synovial membrane, made of macrophages and synovial fibroblasts in a collagen-rich matrix, regulates exchanges with nearby blood capillaries. Articular cartilage, composed of chondrocytes within a collagen-proteoglycan matrix, is critical for joint function. SF, a hyaluronic acid-rich fluid secreted by the synovial membrane, lubricates joints, nourishes avascular cartilage, and carries proteins into circulation. Changes in SF composition can indicate joint disease, such as RA [10,11,12,13]. Inflammatory cells and mediators in SF signal pathology, making it a valuable source of biomarkers. Extracellular vesicles (EVs) in SF contain exosomes and microvesicles, which may mark cartilage degradation or inflammation. Studying SF’s molecular and cellular makeup can reveal metabolic and inflammatory processes in joint diseases. A biomarker discovery pipeline includes four stages: discovery, qualification, verification, and validation (Figure 1). Proteomics identifies proteins in high-quality samples, and selected candidates undergo testing in larger sample groups. Verified biomarkers are then validated and commercialized through clinical assays approved by agencies like the European Medicines Agency (EMA) and the Food and Drug Administration (FDA) in the USA [14,15].
Proteomic analysis for arthritis biomarkers often targets biological fluids, primarily blood (serum or plasma) and SF. While the serum proteome offers a comprehensive view of internal biological activities by circulating through tissues and organs, SF, as a plasma filtrate enriched with specialized proteins, provides a more direct snapshot of joint health. Due to its proximity to critical joint structures, SF is an ideal medium for identifying arthritis biomarkers, which may also help develop serum-based tests for joint disease indicators [16]. Proteomic methods, especially mass spectrometry, enable detailed SF analysis by identifying protein profiles, expression levels, and post-translational modifications (PTMs), which can reveal disease-specific patterns [17,18]. Mass spectrometry combines non-targeted shotgun proteomics with quantitative approaches to link protein identities, concentrations, and disease states [19,20].
In RA, biomarkers can be free in SF or encapsulated within EVs. The composition of EVs depends on their origin and environment, containing elements such as citrullinated epitopes, pro-inflammatory cytokines, immune complexes, and MHC molecules. Research has shown that EVs derived from fibroblast-like synoviocytes (FLS) in RA can promote macrophage migration via PTX3 and PSMB5, rather than directly inducing cytokine release. EVs from RA SF may also contain IgG and citrullinated peptides, suggesting they could act as autoantigens in RA [21]. Exosomes from RA patients’ SF interact with citrullinated proteins—identified as autoantigens—including fibrin α-chain fragments, fibrin β-chains, fibrinogen β-chain precursors, fibrinogen D fragments, and Spα receptor, which can stimulate pro-inflammatory cytokines and immune cell activation, notably Th1 and Th17 cells [22,23].
Although many potential biomarkers for RA are under investigation (see Table 1), clinical tests remain limited to autoantibody tests (RF and anti-CCP) and inflammatory markers (ESR and CRP). These tests are constrained because they primarily detect immune responses or general inflammation, which may not fully capture early or subclinical disease states. The anti-CCP2 antibody test, though highly predictive of erosions, has limited sensitivity in the initial stages, often missing early RA cases when intervention could be the most beneficial. Expanding the biomarker pool for diagnostics and treatment is essential to improve RA patient outcomes [24]. Mass spectrometry, especially at disease sites, is particularly promising for discovering biomarkers that more accurately reflect RA’s underlying biological processes [25].

3. Newly Established Markers

3.1. Aggrecan

Aggrecan, a primary structural protein in cartilage, is broken down by metalloproteinases (MMPs) and aggrecanases, resulting in fragments that reveal neoepitopes, which indicate cartilage turnover [51]. Elevated levels of aggrecan fragments in the SF of RA patients have been linked to disease severity, making aggrecan a promising marker [52]. For instance, studies using ELISA detected significantly higher aggrecan levels in SF from patients with rapid joint destruction compared to those with slower progression [53]. Additionally, increased aggrecan fragments have been observed in patients with RA and osteoarthritis (OA), suggesting that these patients have altered aggrecan profile [54,55]. Interestingly, a unique aggrecan fragment, 374ARGSVI, may help distinguish RA from other types of arthritis, if validated, offering potential diagnostic specificity [56].
The CS846-epitope, a specific chondroitin sulfate marker detected by monoclonal antibody 846, reflects new aggrecan synthesis and is found in minimal amounts in normal cartilage. Initial studies indicated that this epitope is directly linked to aggrecan production and mirrors the breakdown of newly formed aggrecan. In early RA, the serum levels of the CS846-epitope differed among patients with varying disease progressions; those with slower joint deterioration had higher CS846 levels, while patients with rapid joint damage showed lower CS846 levels but increased C-propeptide of type II procollagen (CPII), suggesting a selective rise in collagen synthesis associated with severe cartilage turnover [57,58]. These findings underscore aggrecan’s potential as a biomarker for assessing joint damage progression and tailoring treatments in RA. Further clinical studies are needed to confirm its diagnostic and prognostic utility, paving the way for targeted therapies that could improve disease outcomes by monitoring cartilage health at a molecular level.

3.2. Matrix Metalloproteinases

MMPs serve as crucial biomarkers in RA due to their role in cartilage degradation [59]. In RA, inflamed synovium and activated chondrocytes produce MMPs, notably MMP-1, MMP-3, MMP-9, and MT1-MMP, which are secreted into the SF and actively degrade cartilage [59,60]. Elevated MMP-3 levels strongly correlate with joint damage and predict radiographic progression in RA patients. Rooney et al. highlighted the potential of biomarkers like MMPs in tracking therapeutic responses and predicting joint damage progression [61]. Chang et al. demonstrated significantly elevated MMP-2 and MMP-9 levels in RA, underscoring their role in disease pathogenesis [62]. Active MMPs degrade cartilage components, positioning them as both biomarkers and therapeutic targets [62,63]. However, the clinical use of MMP inhibitors remains limited due to side effects like joint stiffness and fibrosis.
Through an EV-mediated route, endothelial cells are linked to angiogenesis by activating c-Jun N-terminal kinase (JNK) signaling through the encapsulated inhibitor of DNA binding 1 (Id1) [64]. Other in vitro research has demonstrated that granulocyte- and monocyte-derived EVs induce the generation of thrombin, which has a pro-coagulatory impact. This finding raises the possibility of an EV-mediated link between coagulation and inflammation. Furthermore, it has been demonstrated that exosomes generated from FLSs include enzymes that break down the cartilage matrix and that they increase the expression of MMP-13, MMP-3, IL-6, and VEGF in chondrocytes, both of which worsen cartilage injury [65]. Together, these insights into MMP expression and activity reinforce their significant potential as biomarkers and therapeutic targets in the management of RA.

3.3. Glucosyl–Galactosyl–Pyridinoline

Glucosyl–galactosyl–pyridinoline (Glc-Gal-PYD) shows strong potential as a biomarker for joint destruction, particularly in RA [35]. Elevated urinary levels of Glc-Gal-PYD are linked to synovial tissue degradation and the progression of joint damage. In a study of 66 RA patients treated with infliximab and methotrexate, baseline Glc-Gal-PYD levels were significantly higher compared to the healthy controls, suggesting its utility in identifying patients at risk for rapid joint damage [66]. Glc-Gal-PYD, a glycosylated form of pyridinoline (PYD), is primarily found in synovial tissue and is released during its degradation, distinguishing it from other collagen fragments released in joint degradation. Its presence in urine correlates with joint damage severity in RA and OA, making it a promising tool for both the diagnosis and monitoring of disease progression. As a specific marker of synovial tissue breakdown, Glc-Gal-PYD may provide a more accurate measurement of disease activity, improving early diagnosis and enabling targeted treatment adjustments in joint-destructive diseases [35,37].

3.4. Hyaluronic Acid

Hyaluronic acid (HA) levels have attracted attention as biomarkers in RA due to their link to disease activity and joint damage. In RA patients, serum HA levels can be elevated 10- to 20-fold compared to healthy controls, with higher baseline levels correlating with long-term radiographic scores and joint damage progression [67]. However, HA levels show significant diurnal variations, peaking during physical activity, which complicates their use as consistent indicators. In early RA, elevated serum HA correlates with traditional inflammatory markers like CRP and ESR, though the associations are modest (0.436 for ESR and 0.422 for CRP) and may diminish during treatment [67]. These findings suggest the limited utility of HA as an early diagnostic tool, particularly in seronegative RA patients where elevated levels do not clearly correlate with clinical outcomes.
A study by Cylwik et al. highlighted that HA levels can vary not only among individuals with different rheumatic diseases—such as systemic sclerosis and systemic lupus erythematosus—but also within RA subgroups, emphasizing the need to account for baseline inflammation levels and disease activity in interpreting HA data [68]. Also, synovial fluid HA concentrations in RA and OA patients may be influenced by other molecular markers, such as antigenic keratan sulfate, suggesting a more complex relationship between HA levels and disease pathology [69].

3.5. S100 Proteins

The S100 protein family, consisting of over 20 members (e.g., S100A1–S100A18, S100B, and S100P), includes low-molecular-weight calcium-binding proteins (9000–14,000 Da) involved in calcium-dependent cellular processes. S100 proteins typically function as dimers, acting as calcium sensors to regulate gene expression, enzyme activation, cell cycle, and inflammation [70]. Their expression is notably altered in various pathological conditions, including cancer, neurodegenerative disorders, and autoimmune diseases, highlighting their potential as biomarkers [71,72]. Supplementing these findings is research on members of the S100 family in atherosclerosis or Alzheimer’s disease [73,74].
Neutrophils are a major source of S100A8, S100A9, and S100A12, with S100A8/A9 (calprotectin) comprising up to 40% of neutrophil cytoplasmic proteins, while S100A12 is present at lower levels (approximately 5–15%). Elevated levels of S100A8, S100A9, and S100A12 have been identified in the synovial fluid and serum of RA patients, highlighting their role as biomarkers for inflammation, and their potential as diagnostic and prognostic indicators of disease activity and progression [75,76].
Calprotectin, a member of the S100 family, is a valuable biomarker for diagnosing inflammatory conditions, including RA [77]. It is a heterodimer of two calcium-binding proteins, MRP-8 (S100A8) and MRP-14 (S100A9), found in granulocytes and monocytes [78]. At inflammation sites, calprotectin is released as an acute-phase reactant, with levels increasing significantly, often over 100-fold during active inflammation [77]. Unlike CRP and ESR, which are produced in the liver, calprotectin is produced directly at the inflamed synovial tissue, providing a more localized measure of inflammation [79,80]. This unique characteristic makes calprotectin particularly useful for assessing RA activity and progression, especially alongside other S100 family proteins, like S100A12 and S100A4, which are also linked to chronic inflammatory states.

3.6. Cytokines/Inhibitors/Adipocytokines

In RA, an imbalance between pro- and anti-inflammatory cytokines drives the chronic inflammatory processes that characterize the disease [81]. Key pro-inflammatory cytokines, including IL-1, IL-6, IL-15, IL-17, TNF-α, and granulocyte macrophage–colony stimulating factor (GM-CSF), are elevated in the synovial fluid and serum of RA patients, contributing to synovial inflammation, cartilage destruction, and bone erosion [45]. IL-6, one of the most abundant cytokines in RA, plays a central role in systemic inflammation and correlates with disease activity. Therapeutic targeting of the IL-6 receptor has demonstrated significant clinical benefits, though the direct link between IL-6 levels and joint damage remains an area of ongoing study [82].
In addition, markers like soluble TNF receptor II (sTNFR-II) have shown promise as early indicators of RA, with elevated levels detectable years before clinical onset, highlighting their potential role in preclinical diagnosis [83]. In early synovitis cases that progress to RA, unique cytokine profiles have been identified. These include elevated levels of IL-2, IL-4, IL-13, IL-15, and IL-17, which promote inflammatory responses and osteoclastogenesis. Among these, IL-17 is particularly potent in driving bone resorption and joint destruction, making it a key target for therapeutic intervention [46,47].
Chemokines such as CXCL8 (IL-8) and CCL2 (MCP-1) are significantly elevated in RA synovial fluid and correlate with disease activity [84]. Additionally, CXCL13, a chemokine associated with B-cell trafficking, and CCL23, which recruits monocytes and other immune cells, have been identified as potential biomarkers of disease activity in RA patients [85]. Despite these findings, traditional markers like ESR and CRP remain the most reliable for clinical RA assessment.

3.7. Immunological Markers

T-cell infiltrates are a key indicator of a high concentration of CD4+ T cells, particularly regulatory T cells (Tregs) like CD4 + CD25+ Tregs, which are vital for maintaining self-tolerance and preventing autoimmunity [86]. However, Tregs may function inadequately or be defective in RA [87,88]. Studies have shown that while CD4 + CD25+ Tregs are abundant in inflammatory joints and have strong suppressive effects, RA progression continues, possibly due to the reduced sensitivity of effector cells to Treg suppression [49,89]. Van Amelsfort et al. demonstrated elevated Treg levels in synovial fluid compared to peripheral blood, but their suppressive function was significantly diminished in the inflammatory environment of RA [89]. Additionally, proinflammatory cytokines, such as TNF-α and IL-6, have been shown to interfere with Treg-mediated suppression, further contributing to chronic inflammation and joint damage [50]. While Tregs hold potential as biomarkers for immune regulation in RA, findings regarding their diagnostic utility remain inconsistent [49,88].
The current RA biomarker research includes markers such as CTX-I, CTX-II, COMP, Glc-Gal-PYD, MMP3, and C2C in the urine and serum. Although correlations between these markers and patient outcomes have been noted, their interactions and added value over traditional markers remain unclear. A combined biomarker panel could improve prognosis and aid in identifying patients likely to experience aggressive disease, enabling more personalized treatment decisions and potentially sparing others from intensive therapies.

4. Novel Markers

4.1. ADAMEDC1

RA and OA share similar symptoms but differ fundamentally in their underlying mechanisms. Analyzing differentially expressed genes (DEGs) between RA and OA could significantly enhance diagnostic precision and inform targeted treatments. In the study by Kang et al., RA samples showed the upregulation of 14 genes and the downregulation of 3 compared to OA, with gene ontology analysis indicating these DEGs largely regulate inflammation in synovial tissue. Notably, ADAMDEC1 in SF emerged as a promising RA biomarker, potentially aiding in early molecular diagnosis and targeted therapies [90]. The current diagnostic methods for RA and OA, such as imaging and symptom evaluation, are limited in detecting early disease stages, as they primarily assess structural changes and physical function [91]. DEGs like CXCL13, CD247, GZMB, CCL18, IL7R, and ADAMDEC1, identified by Kang et al. and validated by Li et al., reveal molecular distinctions between RA and OA, offering more precise diagnostic and therapeutic targets [92].
The validation of ADAMDEC1 in clinical SF samples demonstrated its robustness in identifying RA lesions, even in patients’ post-joint replacement, underscoring its potential as a diagnostic and prognostic biomarker. The limitations of Kang’s study included a small cohort size and advanced disease stages among the participants. Future research should explore ADAMDEC1’s efficacy in detecting early-stage RA, aiming to establish its role in guiding early intervention and personalized treatment [90].

4.2. MAGE-I-mRNA

A study by Aiman et al. evaluated melanoma-associated antigen gene 1 (MAGE-1) mRNA expression in SF cells, alongside the serum levels of anti-CCP and rheumatoid factor (RF) as early diagnostic biomarkers for RA [93]. The study included 213 participants, with 135 RA patients and 78 patients with traumatic knee injuries as controls. Using ELISA for RF and anti-CCP and RT-PCR for MAGE-1 mRNA, the researchers found significantly elevated serum RF IgM and anti-CCP levels in the RA patients compared to the controls, showing a strong correlation between the two. Remarkably, MAGE-1 mRNA expression was present in 100% of the RA patients but absent in the controls, yielding 100% sensitivity and specificity for MAGE-1, RF, and anti-CCP as diagnostic biomarkers.
The study concluded that MAGE-1 expression in SF cells holds promise as a specific and sensitive diagnostic biomarker for RA. The researchers advocate combining MAGE-1 transcript analysis with RF and anti-CCP to enhance early RA detection, which could lead to improved patient management and outcomes. Further studies with larger cohorts are recommended to confirm MAGE-1’s predictive value and role in chronic RA inflammation. The research underscored the established values of RF and anti-CCP while introducing MAGE-1 as a potential addition to the diagnostic toolkit for early RA intervention [94].

4.3. Non-Coding RNAs

Murata et al. explored microRNAs (miRNAs) in SF and plasma as potential biomarkers for distinguishing RA and OA. Their study assessed specific miRNAs (miR-16, miR-132, miR-146a, miR-155, and miR-223) in the SF of RA and OA patients and in the plasma of RA, OA, and healthy individuals. Using quantitative PCR, they found that miRNA expression differed markedly between SF and plasma, indicating that SF miRNAs likely originate from synovial tissue. Plasma miR-132 successfully differentiated RA and OA patients from the healthy controls, while the SF miRNAs were particularly useful in distinguishing RA from OA. The plasma miRNAs were also correlated positively with RA disease activity, making them valuable for clinical assessments. Notably, miR-16, miR-146a, miR-155, and miR-223 were elevated in RA, underscoring their diagnostic potential [95].
Recent studies have aimed to investigate exosomal miRNAs and other arthritis-related illnesses. Exosomes in synovial fluid are closely linked to arthritis pathophysiology, contributing to joint deterioration, inflammation, and cartilage degradation [22]. Through the exchange of lipids, proteins, various signaling molecules, and nucleic acids—including miRNAs and long non-coding RNAs (lncRNAs)—exosomes are involved in various pathological and physiological processes [96]. miRNAs are the most extensively researched compounds in exosomes and have been employed as potential biomarkers [97,98]. Xie et al. isolated exosomes from osteoarthritis patients’ serum and synovial fluid, using miRNA sequencing to examine the expression. They identified 31 upregulated and 33 downregulated miRNAs in synovial fluid compared to serum, suggesting that serum-derived exosomes may not accurately represent synovial fluid exosomes. These differentially expressed miRNAs were mostly linked to metabolic and cancer-related pathways, providing insights into joint conditions and early indicators for arthritis diagnosis [99]. Notably, miRNAs such as miR-155, miR-150, miR-146a, and miR-21 are often elevated in exosomes from RA patients and are related to IL-17-producing T cell differentiation in RA patients [100,101]. miR-146a, a miRNA that inhibits osteoclasts from destroying bone, and miR-221-3p, an FLS-derived miRNA involved in bone signaling pathways, have been suggested as RA diagnostic biomarkers [102,103]. Yu et al. studied the function of two exosomal miRNAs, miR-335p, and miR-483-5p. They discovered that the SRSF4 gene, which is in charge of cartilage repair, is a typical target for these sequences. This gene’s expression was downregulated, which resulted in the inhibition of cartilage regeneration [104].
Additionally, the homeostasis and imbalance of T-regulatory cells (Tregs)/Th cells have also been connected to exosomes and their miRNA. In particular, the downregulation of TGFBR II (transforming growth factor beta receptor II) expression was linked to the overexpression of miR-17, preventing Treg development in vitro [105].
Additionally, lncRNAs in SF exosomes, particularly a specific transcript—ENST00000433825.1—strongly correlated with CRP levels, which are traditional inflammatory markers. This unique lncRNA expression profile in RA SF may aid in differentiating RA from other joint diseases, such as OA and gout, and offers diagnostic and therapeutic insights [106].

4.4. GFAP and A1BG

Biswas et al. used an immune-proteomic approach to identify novel autoantigens involved in RA pathogenesis, analyzing SF samples from RA patients with non-RA rheumatic samples (OA and trauma) as controls. Through two-dimensional gel electrophoresis (2-DE) and Western blotting, they identified 18 distinct autoantigens in RA, including five key proteins: vimentin, gelsolin, alpha 2 HS glycoprotein (AHSG), glial fibrillary acidic protein (GFAP), and a1B-glycoprotein (A1BG), which showed significantly higher expression in RA SF than in the controls [107]. GFAP has been previously discovered as a novel biomarker of an autoimmune disorder [108]. A1BG is a human plasma protein with unknown functions [109]. ELISA tests further confirmed elevated autoantibodies against GFAP and A1BG in RA plasma compared to OA patients, suggesting their potential as diagnostic biomarkers [107].
The aforementioned study highlights GFAP and A1BG as promising new biomarkers for RA, addressing the limitations in the current RA diagnostics that often rely on non-specific antibody assays. Identifying these autoantigens could improve early diagnosis and prognosis, enabling more personalized treatment approaches. These findings demonstrate the utility of immunoproteomic methods in autoimmune research and suggest similar strategies could be applied to other autoimmune conditions.

4.5. ORM2

Kim et al. investigated orosomucoid-2 (ORM2), an acute-phase protein, in RA, revealing its substantial role in chronic inflammation. Their proteome profiling showed that ORM2 was significantly upregulated in the serum of RA patients, displaying the highest fold change among acute-phase reactants [110]. ORM2 was also elevated in RA synovial fluid and membranes, primarily produced by synovial macrophages and FLSs. The study demonstrated that recombinant ORM2 stimulates RA macrophages and FLSs to produce proinflammatory cytokines (IL-6 and TNF-α) and chemokines (CCL2 and CXCL8) via the NF-κB and p38 MAPK signaling pathways. Glycophorin C was identified as a receptor mediating ORM2’s proinflammatory effects [111].
In mouse models, intra-articular ORM2 injections increased arthritis severity and promoted macrophage infiltration. Serum ORM2 levels were correlated with RA activity and radiographic progression, highlighting its potential as a biomarker for assessing disease severity. The study suggested that ORM2 triggers reciprocal activation between macrophages and FLSs, creating a feedback loop that intensifies inflammation. While limited by short experimental durations and a lack of ORM2-neutralizing antibodies, the research established ORM2 as a promising biomarker and therapeutic target in RA, with liver-derived ORM2 potentially contributing to RA pathogenesis [111].

4.6. 14-3-3η Protein

Hammam et al. studied the 14-3-3η protein as a potential biomarker for assessing joint damage and predicting radiographic progression in established RA. While traditional markers like RF and anti-CCP are commonly used, their effectiveness is limited by issues such as seronegativity, impacting their predictive value for joint damage [112,113]. The study highlighted the need for biomarkers associated with joint damage mechanisms to improve radiographic predictions and inform management strategies [114].
The researchers found that elevated serum and SF levels of 14-3-3η were significantly correlated with radiographic damage and erosion scores. Notably, the SF levels of 14-3-3η were significantly higher than the serum levels, underscoring its value as a direct indicator of joint damage processes [115]. The protein was shown to stimulate pro-inflammatory cytokines, including TNF-α and IL-6, as well as matrix metalloproteinases (MMPs), which contribute to cartilage and bone destruction [115,116]. Additionally, the correlation between SF 14-3-3η levels and patient age suggests that older RA patients, who are at greater risk of severe joint deterioration, may benefit from therapies tailored to their 14-3-3η profile [117].
See Table 2 for the categorization of biomarkers.

5. Specific Applications of Extracellular Vesicles

Exosomes, a specific type of EVs between 40 and 100 nm in size, play a vital role in cellular communication by transporting molecular cargo among cells. In RA, exosomes are abundant in synovial fluid and primarily derived from platelets, as well as monocytes, neutrophils, and granulocytes. Both B and T cells also produce exosomes in the synovial fluid, with the quantity of T-cell-derived exosomes correlating strongly with blood rheumatoid factor levels [118,119]. See Figure 2 for the exosomal contents in an RA joint.
By mediating joint destruction and promoting pathological processes such as cartilage degradation and bone erosion, exosomes contribute significantly to disease progression in RA. Specifically, exosomes derived from RA FLS can stimulate CD4+ T cells, augment IFNγ and IL-2 production, amplify NF-κB and Akt activation, and prevent caspase 3 and caspase 8 cleavages [120]. These exosomes can break down bone and cartilage through direct metalloproteinase activity or the autocrine activation of MMP-1 released by FLS. Additionally, FLS can be the target of exosomes produced by monocytes and activated T cells, leading to an increased release of MMPs, cytokines, and chemokines, and potentially resulting in angiogenesis and the chemokine-induced migration of human microvascular endothelial cells [121]. Exosomes also promote osteoclast activity and bone resorption by upregulating RANK and RANKL expression, critical factors in bone metabolism and RA-associated bone erosion [122]. Given their impact on inflammation, cartilage degradation, and bone resorption, exosomes represent promising targets for diagnostic and therapeutic strategies in RA. The contents of exosomes are summarized in Table 3.

5.1. Heat Shock Proteins

In RA, transmembrane exosomal markers, such as chaperones Hsp70 and Hsp90 (internal components) and tetraspanins CD9 and CD81 (surface markers), are significantly elevated, with increases of 3.2-fold for CD9 and 2.2-fold for Hsp70. These markers serve as potential indicators of exosomal involvement in RA, contributing to inflammation and immune response regulation. Specifically, CD9 and Hsp70 have shown promise as diagnostic markers due to their strong expression in RA-related exosomes. Exosomal proteins also exhibit bidirectional changes in enzymatic activity in RA. Acidic endonucleases, responsible for DNA and RNA breakdown, show a dramatic increase in activity (up to 55–30-fold), while alkaline endonucleases decrease by 2–3 times. This altered enzymatic activity reflects the acidic shift in SF pH seen in RA, from a normal 7.75 to 7.21, which influences the exosomal secretion of proteins and nucleic acids. This pH-dependent change affects intercellular immunomodulatory communication, suggesting that exosomes not only serve as biomarkers but could also be targeted to modulate inflammation [123].

5.2. Lysophosphatidylcholine, Sphingolipids, and Cholesterol

Lipid molecules, including lysophosphatidylcholine, sphingolipids, and cholesterol, play emerging roles in RA-associated inflammation. During acute inflammation, phosphatidylethanolamines and hexosylceramides increase significantly, suggesting a potential link between lipid changes and synovial inflammation. Specific lipids, such as fatty acids, metabolites, and lipoproteins, exhibit immunomodulatory and anti-inflammatory effects, which could impact RA progression. Additionally, lipid classes like hexosylceramides and sphingomyelin are involved in EV biogenesis, potentially influencing EV-mediated communication in RA. However, understanding the role of lipid content within EVs in RA is still in the early stages, and further research is needed to determine their precise impact on disease mechanisms [124].

5.3. Proteomic Analysis of EVs

Proteomic analysis of EVs in the synovial fluid of RA patients has revealed a range of pro-inflammatory proteins that may drive disease progression. Key factors include signal transducers like STAT1 and STAT3, tyrosine kinases JAK1, JAK2, and TYK2, TLR2, and apoptosis-associated PYCARD. Additionally, MMP9, PRKCB, CCR1, leukotriene A–4 hydrolase (LTA4H), and several S100 calcium-binding proteins were identified, indicating their roles in joint inflammation and cartilage breakdown. Autoantigen-associated proteins, such as calreticulin-3, vimentin, HSP90, and histones H1, H2, and H3 were also present, suggesting EVs’ role in autoantigen presentation in RA [125]. Moreover, elevated levels of GTPases (KRas, NRas, Ral-A, and RhoA) and components of the NADPH oxidase complex (RAC1, RAC2, CYBA, and CYBB) indicate involvement in immune response regulation [65].
Further research by Yoo et al. explored serum exosomes from 60 RA patients, half in clinical remission and half with active disease. The results show that arachidonic acid (AA) levels were higher in patients with active disease, while LYVE-1 levels were lower, suggesting these markers could aid in assessing RA disease activity and progression [126].

6. Conclusions

The study of biomarkers in RA, especially those derived from synovial fluid and EVs, opens new avenues for understanding disease mechanisms and enhancing clinical care. Synovial fluid biomarkers provide direct insight into joint-specific inflammatory and degenerative processes, offering a more precise assessment of the disease state than systemic markers like serum proteins. EVs, which carry proteins, lipids, and genetic material, such as microRNAs and heat shock proteins, are particularly promising due to their roles in modulating immune responses and inflammation in RA. These vesicles act as biomarkers and carriers of molecular signatures that reflect disease activity and progression.
As research advances, these biomarkers hold significant promise for improving RA diagnosis, tracking disease activity, predicting therapeutic responses, and identifying patients at higher risk for joint damage. Enhanced the understanding and application of these markers could lead to more personalized, effective RA management strategies and improved patient outcomes.

Author Contributions

V.S., D.K. and S.H.: investigation, writing—original draft, and writing—review and editing: L.D.: writing—review and editing, L.D.: funding acquisition; S.H.: conceptualization. All authors have read and agreed to the published version of the manuscript.

Funding

This study is a result of the project implementation of the Center for Advanced Therapies of Chronic Inflammatory Diseases of the Musculoskeletal System (CPT-ZOPA), ITMS2014+: 313011W410, supported by the Operational Programme Integrated infrastructure funded by the European Regional Development Fund.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Majka, D.S.; Holers, V.M. Can We Accurately Predict the Development of Rheumatoid Arthritis in the Preclinical Phase? Arthritis Rheum. 2003, 48, 2701–2705. [Google Scholar] [CrossRef] [PubMed]
  2. Landewé, R.B.M. The Benefits of Early Treatment in Rheumatoid Arthritis: Confounding by Indication, and the Issue of Timing. Arthritis Rheum. 2003, 48, 1–5. [Google Scholar] [CrossRef] [PubMed]
  3. Lard, L.R.; Visser, H.; Speyer, I.; Vander Horst-Bruinsma, I.E.; Zwinderman, A.H.; Breedveld, F.C.; Hazes, J.M.W. Early versus Delayed Treatment in Patients with Recent-Onset Rheumatoid Arthritis: Comparison of Two Cohorts Who Received Different Treatment Strategies. Am. J. Med. 2001, 111, 446–451. [Google Scholar] [CrossRef] [PubMed]
  4. Almutairi, K.B.; Nossent, J.C.; Preen, D.B.; Keen, H.I.; Inderjeeth, C.A. The Prevalence of Rheumatoid Arthritis: A Systematic Review of Population-Based Studies. J. Rheumatol. 2021, 48, 669–676. [Google Scholar] [CrossRef] [PubMed]
  5. Benjamin, O.; Goyal, A.; Lappin, S.L. Disease-Modifying Antirheumatic Drugs (DMARD). In StatPearls; StatPearls Publishing: St. Petersburg, FL, USA, 2023. [Google Scholar]
  6. Krishnan, Y.; Grodzinsky, A.J. Cartilage Diseases. Matrix Biol. 2018, 71–72, 51–69. [Google Scholar] [CrossRef]
  7. Goekoop-Ruiterman, Y.P.M.; De Vries-Bouwstra, J.K.; Allaart, C.F.; Van Zeben, D.; Kerstens, P.J.S.M.; Hazes, J.M.W.; Zwinderman, A.H.; Ronday, H.K.; Han, K.H.; Westedt, M.L.; et al. Clinical and Radiographic Outcomes of Four Different Treatment Strategies in Patients with Early Rheumatoid Arthritis (the Best Study): A Randomized, Controlled Trial. Arthritis Rheum. 2005, 52, 3381–3390. [Google Scholar] [CrossRef]
  8. Ahmed, U.; Anwar, A.; Savage, R.S.; Costa, M.L.; MacKay, N.; Filer, A.; Raza, K.; Watts, R.A.; Winyard, P.G.; Tarr, J.; et al. Biomarkers of Early Stage Osteoarthritis, Rheumatoid Arthritis and Musculoskeletal Health. Sci. Rep. 2015, 5, 9259. [Google Scholar] [CrossRef]
  9. Nielsen, S.F.; Bojesen, S.E.; Schnohr, P.; Nordestgaard, B.G. Elevated Rheumatoid Factor and Long Term Risk of Rheumatoid Arthritis: A Prospective Cohort Study. BMJ 2012, 345, e5244. [Google Scholar] [CrossRef]
  10. Jones, E.A.; Crawford, A.; English, A.; Henshaw, K.; Mundy, J.; Corscadden, D.; Chapman, T.; Emery, P.; Hatton, P.; McGonagle, D. Synovial Fluid Mesenchymal Stem Cells in Health and Early Osteoarthritis: Detection and Functional Evaluation at the Single-Cell Level. Arthritis Rheum. 2008, 58, 1731–1740. [Google Scholar] [CrossRef]
  11. Gibson, D.S.; Rooney, M.E. The Human Synovial Fluid Proteome: A Key Factor in the Pathology of Joint Disease. Proteom.—Clin. Appl. 2007, 1, 889–899. [Google Scholar] [CrossRef]
  12. Hui, A.Y.; McCarty, W.J.; Masuda, K.; Firestein, G.S.; Sah, R.L. A Systems Biology Approach to Synovial Joint Lubrication in Health, Injury, and Disease. Wiley Interdiscip. Rev. Syst. Biol. Med. 2012, 4, 15–37. [Google Scholar] [CrossRef] [PubMed]
  13. Choy, E. Understanding the Dynamics: Pathways Involved in the Pathogenesis of Rheumatoid Arthritis. Rheumatology 2012, 51, 3–11. [Google Scholar] [CrossRef] [PubMed]
  14. Cook, A.D.; Turner, A.L.; Braine, E.L.; Pobjoy, J.; Lenzo, J.C.; Hamilton, J.A. Regulation of Systemic and Local Myeloid Cell Subpopulations by Bone Marrow Cell-Derived Granulocyte-Macrophage Colony-Stimulating Factor in Experimental Inflammatory Arthritis. Arthritis Rheum. 2011, 63, 2340–2351. [Google Scholar] [CrossRef]
  15. Yonezawa, I.; Okuda, T.; Won, J.; Sakoda, J.; Nakahara, D.; Nojiri, H.; Muto, O.; Momomura, R.; Kaneko, K. Retrodental Mass in Rheumatoid Arthritis. J. Spinal Disord. Tech. 2013, 26, 65–69. [Google Scholar] [CrossRef]
  16. Bhattacharjee, M.; Raju, R.; Radhakrishnan, A.; Nanjappa, V.; Muthusamy, B.; Singh, K.; Kuppusamy, D.; Lingala, B.T.; Pan, A.; Mathur, P.P.; et al. A Bioinformatics Resource for TWEAK-Fn14 Signaling Pathway. J. Signal Transduct. 2012, 2012, 376470. [Google Scholar] [CrossRef]
  17. Parker, C.E.; Borchers, C.H. Mass Spectrometry Based Biomarker Discovery, Verification, and Validation—Quality Assurance and Control of Protein Biomarker Assays. Mol. Oncol. 2014, 8, 840–858. [Google Scholar] [CrossRef]
  18. Balakrishnan, L.; Bhattacharjee, M.; Ahmad, S.; Nirujogi, R.S.; Renuse, S.; Subbannayya, Y.; Marimuthu, A.; Srikanth, S.M.; Raju, R.; Dhillon, M.; et al. Differential Proteomic Analysis of Synovial Fluid from Rheumatoid Arthritis and Osteoarthritis Patients. Clin. Proteom. 2014, 11, 1. [Google Scholar] [CrossRef] [PubMed]
  19. Cretu, D.; Prassas, I.; Saraon, P.; Batruch, I.; Gandhi, R.; Diamandis, E.P.; Chandran, V. Identification of Psoriatic Arthritis Mediators in Synovial Fluid by Quantitative Mass Spectrometry. Clin. Proteom. 2014, 11, 27. [Google Scholar] [CrossRef]
  20. Matsuo, K.; Xiang, Y.; Nakamura, H.; Masuko, K.; Yudoh, K.; Noyori, K.; Nishioka, K.; Saito, T.; Kato, T. Identification of Novel Citrullinated Autoantigens of Synovium in Rheumatoid Arthritis Using a Proteomic Approach. Arthritis Res. Ther. 2006, 8, R175. [Google Scholar] [CrossRef]
  21. Nakamachi, Y.; Uto, K.; Hayashi, S.; Okano, T.; Morinobu, A.; Kuroda, R.; Kawano, S.; Saegusa, J. Exosomes Derived from Synovial Fibroblasts from Patients with Rheumatoid Arthritis Promote Macrophage Migration That Can Be Suppressed by miR-124-3p. Heliyon 2023, 9, e14986. [Google Scholar] [CrossRef]
  22. Li, Z.; Wang, Y.; Xiao, K.; Xiang, S.; Li, Z.; Weng, X. Emerging Role of Exosomes in the Joint Diseases. Cell. Physiol. Biochem. 2018, 47, 2008–2017. [Google Scholar] [CrossRef] [PubMed]
  23. Skriner, K.; Adolph, K.; Jungblut, P.R.; Burmester, G.R. Association of Citrullinated Proteins with Synovial Exosomes. Arthritis Rheum. 2006, 54, 3809–3814. [Google Scholar] [CrossRef] [PubMed]
  24. Bugatti, S.; Manzo, A.; Caporali, R.; Montecucco, C. Assessment of Synovitis to Predict Bone Erosions in Rheumatoid Arthritis. Ther. Adv. Musculoskelet. Dis. 2012, 4, 235–244. [Google Scholar] [CrossRef] [PubMed]
  25. Ali, M.; Manolios, N. Proteomics in Rheumatology: A New Direction for Old Diseases. Semin. Arthritis Rheum. 2005, 35, 67–76. [Google Scholar] [CrossRef]
  26. Burton, P.R.; Clayton, D.G.; Cardon, L.R.; Craddock, N.; Deloukas, P.; Duncanson, A.; Kwiatkowski, D.P.; McCarthy, M.I.; Ouwehand, W.H.; Samani, N.J.; et al. Genome-Wide Association Study of 14,000 Cases of Seven Common Diseases and 3000 Shared Controls. Nature 2007, 447, 661–678. [Google Scholar] [CrossRef]
  27. Kurreeman, F.A.; Padyukov, L.; Marques, R.B.; Schrodi, S.J.; Seddighzadeh, M.; Stoeken-Rijsbergen, G.; van der Helm-van Mil, A.H.; Allaart, C.F.; Verduyn, W.; Houwing-Duistermaat, J.; et al. A Candidate Gene Approach Identifies the TRAF1/C5 Region as a Risk Factor for Rheumatoid Arthritis. PLoS Med. 2007, 4, e278. [Google Scholar] [CrossRef]
  28. Hoppe, B.; Häupl, T.; Egerer, K.; Gruber, R.; Kiesewetter, H.; Salama, A.; Burmester, G.R.; Dörner, T. Influence of Peptidylarginine Deiminase Type 4 Genotype and Shared Epitope on Clinical Characteristics and Autoantibody Profile of Rheumatoid Arthritis. Ann. Rheum. Dis. 2009, 68, 898–903. [Google Scholar] [CrossRef]
  29. Van Tuyl, L.H.D.; Voskuyl, A.E.; Boers, M.; Geusens, P.; Landewe, R.B.M.; Dijkmans, B.A.C.; Lems, W.F. Baseline RANKL:OPG Ratio and Markers of Bone and Cartilage Degradation Predict Annual Radiological Progression over 11 Years in Rheumatoid Arthritis. Ann. Rheum. Dis. 2010, 69, 1623–1628. [Google Scholar] [CrossRef]
  30. Syversen, S.W.; Goll, G.L.; Van Der Heijde, D.; Landewé, R.; Gaarder, P.I.; Ødegård, S.; Haavardsholm, E.A.; Kvien, T.K. Cartilage and Bone Biomarkers in Rheumatoid Arthritis: Prediction of 10-Year Radiographic Progression. J. Rheumatol. 2009, 36, 266–272. [Google Scholar] [CrossRef]
  31. Mansson, B. Release of Cartilage and Bone Macromolecules into Synovial Fluid: Differences between Psoriatic Arthritis and Rheumatoid Arthritis. Ann. Rheum. Dis. 2001, 60, 27–31. [Google Scholar] [CrossRef]
  32. Nakamura, R.M. Progress in the Use of Biochemical and Biological Markers for Evaluation of Rheumatoid Arthritis. J. Clin. Lab. Anal. 2000, 14, 305–313. [Google Scholar] [CrossRef] [PubMed]
  33. Van Den Broek, T.; Tesser, J.R.; Albani, S. The Evolution of Biomarkers in Rheumatoid Arthritis: From Clinical Research to Clinical Care. Expert Opin. Biol. Ther. 2008, 8, 1773–1785. [Google Scholar] [CrossRef] [PubMed]
  34. Skoumal, M.; Haberhauer, G.; Feyertag, J.; Kittl, E.M.; Bauer, K.; Dunky, A. Serum Levels of Cartilage Oligomeric Matrix Protein Are Elevated in Rheumatoid Arthritis, but Not in Inflammatory Rheumatic Diseases Such as Psoriatic Arthritis, Reactive Arthritis, Raynaud’s Syndrome, Scleroderma, Systemic Lupus Erythematosus, Vasculitis and Sjögren’s Syndrome. Arthritis Res. Ther. 2004, 6, 73. [Google Scholar] [CrossRef] [PubMed]
  35. Garnero, P.; Gineyts, E.; Christgau, S.; Finck, B.; Delmas, P.D. Association of Baseline Levels of Urinary Glucosyl-Galactosyl-Pyridinoline and Type II Collagen C-Telopeptide with Progression of Joint Destruction in Patients with Early Rheumatoid Arthritis. Arthritis Rheum. 2002, 46, 21–30. [Google Scholar] [CrossRef] [PubMed]
  36. Fraser, A.; Fearon, U.; Billinghurst, R.C.; Ionescu, M.; Reece, R.; Barwick, T.; Emery, P.; Poole, A.R.; Veale, D.J. Turnover of Type II Collagen and Aggrecan in Cartilage Matrix at the Onset of Inflammatory Arthritis in Humans: Relationship to Mediators of Systemic and Local Inflammation. Arthritis Rheum. 2003, 48, 3085–3095. [Google Scholar] [CrossRef]
  37. Gineyts, E.; Garnero, P.; Delmas, P.D. Urinary Excretion of Glucosyl-galactosyl Pyridinoline: A Specific Biochemical Marker of Synovium Degradation. Rheumatology 2001, 40, 315–323. [Google Scholar] [CrossRef]
  38. Jonsson, T. Predictive Value of Rheumatoid Factor Isotypes for Radiological Progression in Patients with Rheumatoid Arthritis. Scand. J. Rheumatol. 1996, 25, 189–190. [Google Scholar] [CrossRef]
  39. Tedesco, A.; D’Agostino, D.; Soriente, I.; Amato, P.; Piccoli, R.; Sabatini, P. A New Strategy for the Early Diagnosis of Rheumatoid Arthritis: A Combined Approach. Autoimmun. Rev. 2009, 8, 233–237. [Google Scholar] [CrossRef]
  40. Vossenaar, E.R.; Després, N.; Lapointe, E.; Van Der Heijden, A.; Lora, M.; Senshu, T.; Van Venrooij, W.J.; Ménard, H.A. Rheumatoid Arthritis Specific Anti-Sa Antibodies Target Citrullinated Vimentin. Arthritis Res. Ther. 2004, 6, R142. [Google Scholar] [CrossRef]
  41. Vincent, C.; Serre, G.; Lapeyre, F.; Fournie, B.; Ayrolles, C.; Fournie, A.; Soleilhavoup, J.P. High Diagnostic Value in Rheumatoid Arthritis of Antibodies to the Stratum Corneum of Rat Oesophagus Epithelium, so-Called “Antikeratin Antibodies”. Ann. Rheum. Dis. 1989, 48, 712–722. [Google Scholar] [CrossRef]
  42. van Leeuwen, M.A.; van Rijswijk, M.H.; van der Heijde, D.M.F.M.; Meerman, G.J.T.; van Riel, P.L.C.M.; Houtman, P.M.; van de Putte, L.B.A.; Limburg, P.C. The Acute-Phase Response in Relation to Radiographic Progression in Early Rheumatoid Arthritis: A Prospective Study During the First Three Years of the Disease. Rheumatology 1993, 32, 9–13. [Google Scholar] [CrossRef] [PubMed]
  43. Hammer, H.B.; Odegard, S.; Fagerhol, M.K.; Landewe, R.; Heijde, D.V.D.; Uhlig, T.; Mowinckel, P.; Kvien, T.K. Calprotectin (a Major Leucocyte Protein) Is Strongly and Independently Correlated with Joint Inflammation and Damage in Rheumatoid Arthritis. Ann. Rheum. Dis. 2007, 66, 1093–1097. [Google Scholar] [CrossRef]
  44. Kokubun, M.; Imafuku, Y.; Okada, M.; Ohguchi, Y.; Ashikawa, T.; Yamada, T.; Yoshida, H. Serum Amyloid A (SAA) Concentration Varies among Rheumatoid Arthritis Patients Estimated by SAA/CRP Ratio. Clin. Chim. Acta 2005, 360, 97–102. [Google Scholar] [CrossRef] [PubMed]
  45. Feldmann, M.; Brennan, F.M.; Maini, R.N. Role of Cytokines in Rheumatoid Arthritis. Annu. Rev. Immunol. 1996, 14, 397–440. [Google Scholar] [CrossRef] [PubMed]
  46. Kotake, S.; Udagawa, N.; Takahashi, N.; Matsuzaki, K.; Itoh, K.; Ishiyama, S.; Saito, S.; Inoue, K.; Kamatani, N.; Gillespie, M.T.; et al. IL-17 in Synovial Fluids from Patients with Rheumatoid Arthritis Is a Potent Stimulator of Osteoclastogenesis. J. Clin. Investig. 1999, 103, 1345–1352. [Google Scholar] [CrossRef] [PubMed]
  47. Karlson, E.W.; Chibnik, L.B.; Tworoger, S.S.; Lee, I.; Buring, J.E.; Shadick, N.A.; Manson, J.E.; Costenbader, K.H. Biomarkers of Inflammation and Development of Rheumatoid Arthritis in Women from Two Prospective Cohort Studies. Arthritis Rheum. 2009, 60, 641–652. [Google Scholar] [CrossRef]
  48. Rho, Y.H.; Solus, J.; Sokka, T.; Oeser, A.; Chung, C.P.; Gebretsadik, T.; Shintani, A.; Pincus, T.; Stein, C.M. Adipocytokines Are Associated with Radiographic Joint Damage in Rheumatoid Arthritis. Arthritis Rheum. 2009, 60, 1906–1914. [Google Scholar] [CrossRef]
  49. Cao, D.; Vollenhoven, R.V.; Klareskog, L.; Trollmo, C.; Malmström, V. CD25brightCD4+regulatory T Cells Are Enriched in Inflamed Joints of Patients with Chronic Rheumatic Disease. Arthritis Res. Ther. 2004, 6, R335. [Google Scholar] [CrossRef]
  50. Van Amelsfort, J.M.R.; Van Roon, J.A.G.; Noordegraaf, M.; Jacobs, K.M.G.; Bijlsma, J.W.J.; Lafeber, F.P.J.G.; Taams, L.S. Proinflammatory Mediator–Induced Reversal of CD4+,CD25+ Regulatory T Cell–Mediated Suppression in Rheumatoid Arthritis. Arthritis Rheum. 2007, 56, 732–742. [Google Scholar] [CrossRef]
  51. Nagase, H.; Kashiwagi, M. Aggrecanases and Cartilage Matrix Degradation. Arthritis Res. Ther. 2003, 5, 94. [Google Scholar] [CrossRef]
  52. Poole, A.R.; Ionescu, M.; Swan, A.; Dieppe, P.A. Changes in Cartilage Metabolism in Arthritis Are Reflected by Altered Serum and Synovial Fluid Levels of the Cartilage Proteoglycan Aggrecan. Implications for Pathogenesis. J. Clin. Investig. 1994, 94, 25–33. [Google Scholar] [CrossRef] [PubMed]
  53. Larsson, S.; Lohmander, L.S.; Struglics, A. Synovial Fluid Level of Aggrecan ARGS Fragments Is a More Sensitive Marker of Joint Disease than Glycosaminoglycan or Aggrecan Levels: A Cross-Sectional Study. Arthritis Res. Ther. 2009, 11, R92. [Google Scholar] [CrossRef]
  54. Rims, C.; Uchtenhagen, H.; Kaplan, M.J.; Carmona-Rivera, C.; Carlucci, P.; Mikecz, K.; Markovics, A.; Carlin, J.; Buckner, J.H.; James, E.A. Citrullinated Aggrecan Epitopes as Targets of Autoreactive CD 4+ T Cells in Patients With Rheumatoid Arthritis. Arthritis Rheumatol. 2019, 71, 518–528. [Google Scholar] [CrossRef] [PubMed]
  55. Rousseau, J.C.; Sumer, E.U.; Hein, G.; Sondergaard, B.C.; Madsen, S.H.; Pedersen, C.; Neumann, T.; Mueller, A.; Qvist, P.; Delmas, P.; et al. Patients with Rheumatoid Arthritis Have an Altered Circulatory Aggrecan Profile. BMC Musculoskelet. Disord. 2008, 9, 74. [Google Scholar] [CrossRef]
  56. Kalai, E.; Bahlous, A.; Charni, N.; Bouzid, K.; Sahli, H.; Laadhar, L.; Chelly, M.; Rajhi, H.; Zouari, B.; Makni, S.; et al. Association of Serum Levels of Aggrecan ARGS, NITEGE Fragments and Radiologic Knee Osteoarthritis in Tunisian Patients. Jt. Bone Spine 2012, 79, 610–615. [Google Scholar] [CrossRef]
  57. Månsson, B.; Carey, D.; Alini, M.; Ionescu, M.; Rosenberg, L.C.; Poole, A.R.; Heinegård, D.; Saxne, T. Cartilage and Bone Metabolism in Rheumatoid Arthritis: Differences between Rapid and Slow Progression of Disease Identified by Serum Markers of Cartilage Metabolism. J. Clin. Investig. 1995, 95, 1071–1077. [Google Scholar] [CrossRef]
  58. de Grauw, J.C.; van de Lest, C.H.A.; van Weeren, P.R. Inflammatory Mediators and Cartilage Biomarkers in Synovial Fluid after a Single Inflammatory Insult: A Longitudinal Experimental Study. Arthritis Res. Ther. 2009, 11, R35. [Google Scholar] [CrossRef]
  59. Close, D.R. Matrix Metalloproteinase Inhibitors in Rheumatic Diseases. Ann. Rheum. Dis. 2001, 60 (Suppl. 3), iii62–iii67. [Google Scholar] [CrossRef] [PubMed]
  60. Miller, M.; Manning, H.B.; Jain, A.; Troeberg, L.; Dudhia, J.; Essex, D.; Sandison, A.; Seiki, M.; Nanchahal, J.; Nagase, H.; et al. Membrane Type 1 Matrix Metalloproteinase Is a Crucial Promoter of Synovial Invasion in Human Rheumatoid Arthritis. Arthritis Rheum. 2009, 60, 686–697. [Google Scholar] [CrossRef]
  61. Rooney, T.; Roux-Lombard, P.; Veale, D.J.; FitzGerald, O.; Dayer, J.-M.; Bresnihan, B. Synovial Tissue and Serum Biomarkers of Disease Activity, Therapeutic Response and Radiographic Progression: Analysis of a Proof-of-Concept Randomised Clinical Trial of Cytokine Blockade. Ann. Rheum. Dis. 2010, 69, 706–714. [Google Scholar] [CrossRef]
  62. Chang, Y.-H.; Lin, I.-L.; Tsay, G.J.; Yang, S.-C.; Yang, T.-P.; Ho, K.-T.; Hsu, T.-C.; Shiau, M.-Y. Elevated Circulatory MMP-2 and MMP-9 Levels and Activities in Patients with Rheumatoid Arthritis and Systemic Lupus Erythematosus. Clin. Biochem. 2008, 41, 955–959. [Google Scholar] [CrossRef] [PubMed]
  63. Young-Min, S.; Cawston, T.; Marshall, N.; Coady, D.; Christgau, S.; Saxne, T.; Robins, S.; Griffiths, I. Biomarkers Predict Radiographic Progression in Early Rheumatoid Arthritis and Perform Well Compared with Traditional Markers. Arthritis Rheum. 2007, 56, 3236–3247. [Google Scholar] [CrossRef] [PubMed]
  64. Edhayan, G.; Ohara, R.A.; Stinson, W.A.; Amin, M.A.; Isozaki, T.; Ha, C.M.; Haines, G.K.; Morgan, R.; Campbell, P.L.; Arbab, A.S.; et al. Inflammatory Properties of Inhibitor of DNA Binding 1 Secreted by Synovial Fibroblasts in Rheumatoid Arthritis. Arthritis Res. Ther. 2016, 18, 87. [Google Scholar] [CrossRef] [PubMed]
  65. Foers, A.D.; Dagley, L.F.; Chatfield, S.; Webb, A.I.; Cheng, L.; Hill, A.F.; Wicks, I.P.; Pang, K.C. Proteomic Analysis of Extracellular Vesicles Reveals an Immunogenic Cargo in Rheumatoid Arthritis Synovial Fluid. Clin. Trans. Immunol. 2020, 9, e1185. [Google Scholar] [CrossRef] [PubMed]
  66. Marotte, H.; Gineyts, E.; Miossec, P.; Delmas, P.D. Effects of Infliximab Therapy on Biological Markers of Synovium Activity and Cartilage Breakdown in Patients with Rheumatoid Arthritis. Ann. Rheum. Dis. 2009, 68, 1197–1200. [Google Scholar] [CrossRef]
  67. Majeed, M. Relationship between Serum Hyaluronic Acid Level and Disease Activity in Early Rheumatoid Arthritis. Ann. Rheum. Dis. 2004, 63, 1166–1168. [Google Scholar] [CrossRef]
  68. Cylwik, B.; Gruszewska, E.; Gindzienska-Sieskiewicz, E.; Kowal-Bielecka, O.; Chrostek, L. Comparison of Hyaluronic Acid in Patients with Rheumatoid Arthritis, Systemic Sclerosis and Systemic Lupus Erythematosus. Biochem. Med. (Online) 2021, 31, 240–249. [Google Scholar] [CrossRef]
  69. Chubinskaya, S.; Frank, B.S.; Michalska, M.; Kumar, B.; Merrihew, C.A.; Thonar, E.; Lenz, M.; Otten, L.; Rueger, D.C.; Block, J.A. Osteogenic Protein 1 in Synovial Fluid from Patients with Rheumatoid Arthritis or Osteoarthritis: Relationship with Disease and Levels of Hyaluronan and Antigenic Keratan Sulfate. Arthritis Res. Ther. 2006, 8, R73. [Google Scholar] [CrossRef]
  70. Gonzalez, L.L.; Garrie, K.; Turner, M.D. Role of S100 Proteins in Health and Disease. Biochim. Et Biophys. Acta (BBA)—Mol. Cell Res. 2020, 1867, 118677. [Google Scholar] [CrossRef]
  71. Brenner, A.K.; Bruserud, Ø. S100 Proteins in Acute Myeloid Leukemia. Neoplasia 2018, 20, 1175–1186. [Google Scholar] [CrossRef]
  72. Austermann, J.; Spiekermann, C.; Roth, J. S100 Proteins in Rheumatic Diseases. Nat. Rev. Rheumatol. 2018, 14, 528–541. [Google Scholar] [CrossRef] [PubMed]
  73. Cristóvão, J.S.; Gomes, C.M. S100 Proteins in Alzheimer’s Disease. Front. Neurosci. 2019, 13, 463. [Google Scholar] [CrossRef] [PubMed]
  74. Xiao, X.; Yang, C.; Qu, S.-L.; Shao, Y.-D.; Zhou, C.-Y.; Chao, R.; Huang, L.; Zhang, C. S100 Proteins in Atherosclerosis. Clin. Chim. Acta 2020, 502, 293–304. [Google Scholar] [CrossRef] [PubMed]
  75. Baillet, A.; Trocme, C.; Berthier, S.; Arlotto, M.; Grange, L.; Chenau, J.; Quetant, S.; Seve, M.; Berger, F.; Juvin, R.; et al. Synovial Fluid Proteomic Fingerprint: S100A8, S100A9 and S100A12 Proteins Discriminate Rheumatoid Arthritis from Other Inflammatory Joint Diseases. Rheumatology 2010, 49, 671–682. [Google Scholar] [CrossRef]
  76. Yao, Z.; Zhang, B.; Niu, G.; Yan, Z.; Tong, X.; Zou, Y.; Li, Y.; Yang, M. Neutrophil Infiltration Characterized by Upregulation of S100A8, S100A9, S100A12 and CXCR2 Is Associated With the Co-Occurrence of Crohn’s Disease and Peripheral Artery Disease. Front. Immunol. 2022, 13, 896645. [Google Scholar] [CrossRef]
  77. 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]
  78. Odink, K.; Cerletti, N.; Brüggen, J.; Clerc, R.G.; Tarcsay, L.; Zwadlo, G.; Gerhards, G.; Schlegel, R.; Sorg, C. Two Calcium-Binding Proteins in Infiltrate Macrophages of Rheumatoid Arthritis. Nature 1987, 330, 80–82. [Google Scholar] [CrossRef]
  79. Menees, S.B.; Powell, C.; Kurlander, J.; Goel, A.; Chey, W.D. A Meta-Analysis of the Utility of C-Reactive Protein, Erythrocyte Sedimentation Rate, Fecal Calprotectin, and Fecal Lactoferrin to Exclude Inflammatory Bowel Disease in Adults With IBS. Am. J. Gastroenterol. 2015, 110, 444–454. [Google Scholar] [CrossRef]
  80. Yang, Z.; Clark, N.; Park, K.T. Effectiveness and Cost-Effectiveness of Measuring Fecal Calprotectin in Diagnosis of Inflammatory Bowel Disease in Adults and Children. Clin. Gastroenterol. Hepatol. 2014, 12, 253–262.e2. [Google Scholar] [CrossRef]
  81. Firestein, G.S. Evolving Concepts of Rheumatoid Arthritis. Nature 2003, 423, 356–361. [Google Scholar] [CrossRef]
  82. Dayer, J.-M.; Choy, E. Therapeutic Targets in Rheumatoid Arthritis: The Interleukin-6 Receptor. Rheumatology 2010, 49, 15–24. [Google Scholar] [CrossRef] [PubMed]
  83. Jorgensen, K.T.; Wiik, A.; Pedersen, M.; Hedegaard, C.J.; Vestergaard, B.F.; Gislefoss, R.E.; Kvien, T.K.; Wohlfahrt, J.; Bendtzen, K.; Frisch, M. Cytokines, Autoantibodies and Viral Antibodies in Premorbid and Postdiagnostic Sera from Patients with Rheumatoid Arthritis: Case-Control Study Nested in a Cohort of Norwegian Blood Donors. Ann. Rheum. Dis. 2008, 67, 860–866. [Google Scholar] [CrossRef] [PubMed]
  84. Iwamoto, T.; Okamoto, H.; Toyama, Y.; Momohara, S. Molecular Aspects of Rheumatoid Arthritis: Chemokines in the Joints of Patients. FEBS J. 2008, 275, 4448–4455. [Google Scholar] [CrossRef]
  85. Rioja, I.; Hughes, F.J.; Sharp, C.H.; Warnock, L.C.; Montgomery, D.S.; Akil, M.; Wilson, A.G.; Binks, M.H.; Dickson, M.C. Potential Novel Biomarkers of Disease Activity in Rheumatoid Arthritis Patients: CXCL13, CCL23, Transforming Growth Factor α, Tumor Necrosis Factor Receptor Superfamily Member 9, and Macrophage Colony-stimulating Factor. Arthritis Rheum. 2008, 58, 2257–2267. [Google Scholar] [CrossRef]
  86. Cope, A.P. T Cells in Rheumatoid Arthritis. Arthritis Res. Ther. 2008, 10, S1. [Google Scholar] [CrossRef]
  87. Ehrenstein, M.R.; Evans, J.G.; Singh, A.; Moore, S.; Warnes, G.; Isenberg, D.A.; Mauri, C. Compromised Function of Regulatory T Cells in Rheumatoid Arthritis and Reversal by Anti-TNFα Therapy. J. Exp. Med. 2004, 200, 277–285. [Google Scholar] [CrossRef]
  88. Londei, M. Role of Regulatory T Cells in Experimental Arthritis and Implications for Clinical Use. Arthritis Res. Ther. 2005, 7, 118. [Google Scholar] [CrossRef]
  89. Van Amelsfort, J.M.R.; Jacobs, K.M.G.; Bijlsma, J.W.J.; Lafeber, F.P.J.G.; Taams, L.S. CD4+CD25+ Regulatory T Cells in Rheumatoid Arthritis: Differences in the Presence, Phenotype, and Function between Peripheral Blood and Synovial Fluid. Arthritis Rheum. 2004, 50, 2775–2785. [Google Scholar] [CrossRef] [PubMed]
  90. 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]
  91. Xue, F.; Zhang, C.; He, Z.; Ding, L.; Xiao, H. Analysis of Critical Molecules and Signaling Pathways in Osteoarthritis and Rheumatoid Arthritis. Mol. Med. Rep. 2013, 7, 603–607. [Google Scholar] [CrossRef]
  92. Li, W.C.; Bai, D.L.; Xu, Y.; Chen, H.; Ma, R.; Hou, W.B.; Xu, R.J. Identification of Differentially Expressed Genes in Synovial Tissue of Rheumatoid Arthritis and Osteoarthritis in Patients. J. Cell. Biochem. 2019, 120, 4533–4544. [Google Scholar] [CrossRef] [PubMed]
  93. Abd Elsalam, E.; El-Maghawry, I.; Emerah, A. Qualitative Assay of Synovial MAGE-1 m-RNA as a New Trend in Diagnosis of Rheumatoid Arthritis. BESPS 2011, 31, 25–36. [Google Scholar] [CrossRef]
  94. Al-Qtaitat, A.; Mwafi, N.; Albtoosh, A.; Al-Dalaien, N. A New Tool for Early Diagnosis of Rheumatoid Arthritis Using Combined Biomarkers; Synovial MAGE-1 mRNA and Serum Anti-CCP and RF. Pan Afr. Med. J. 2020, 36, 270. [Google Scholar] [CrossRef]
  95. Murata, K.; Yoshitomi, H.; Tanida, S.; Ishikawa, M.; Nishitani, K.; Ito, H.; Nakamura, T. Plasma and Synovial Fluid microRNAs as Potential Biomarkers of Rheumatoid Arthritis and Osteoarthritis. Arthritis Res. Ther. 2010, 12, R86. [Google Scholar] [CrossRef] [PubMed]
  96. Mahendran, S.M.; Oikonomopoulou, K.; Diamandis, E.P.; Chandran, V. Synovial Fluid Proteomics in the Pursuit of Arthritis Mediators: An Evolving Field of Novel Biomarker Discovery. Crit. Rev. Clin. Lab. Sci. 2017, 54, 495–505. [Google Scholar] [CrossRef] [PubMed]
  97. Mori, M.A.; Ludwig, R.G.; Garcia-Martin, R.; Brandão, B.B.; Kahn, C.R. Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab. 2019, 30, 656–673. [Google Scholar] [CrossRef]
  98. Liu, T.; Zhang, Q.; Zhang, J.; Li, C.; Miao, Y.-R.; Lei, Q.; Li, Q.; Guo, A.-Y. EVmiRNA: A Database of miRNA Profiling in Extracellular Vesicles. Nucleic Acids Res. 2019, 47, D89–D93. [Google Scholar] [CrossRef]
  99. Xie, Y.; Chen, W.; Zhao, M.; Xu, Y.; Yu, H.; Qin, J.; Li, H. Exploration of Exosomal miRNAs from Serum and Synovial Fluid in Arthritis Patients. Diagnostics 2022, 12, 239. [Google Scholar] [CrossRef]
  100. Murata, K.; Yoshitomi, H.; Furu, M.; Ishikawa, M.; Shibuya, H.; Ito, H.; Matsuda, S. MicroRNA-451 Down-Regulates Neutrophil Chemotaxis via P38 MAPK. Arthritis Rheumatol. 2014, 66, 549–559. [Google Scholar] [CrossRef]
  101. Niimoto, T.; Nakasa, T.; Ishikawa, M.; Okuhara, A.; Izumi, B.; Deie, M.; Suzuki, O.; Adachi, N.; Ochi, M. MicroRNA-146a Expresses in Interleukin-17 Producing T Cells in Rheumatoid Arthritis Patients. BMC Musculoskelet. Disord. 2010, 11, 209. [Google Scholar] [CrossRef]
  102. Maeda, Y.; Farina, N.H.; Matzelle, M.M.; Fanning, P.J.; Lian, J.B.; Gravallese, E.M. Synovium-Derived MicroRNAs Regulate Bone Pathways in Rheumatoid Arthritis. J. Bone Miner. Res. 2017, 32, 461–472. [Google Scholar] [CrossRef] [PubMed]
  103. Bogunia-Kubik, K.; Wysoczańska, B.; Piątek, D.; Iwaszko, M.; Ciechomska, M.; Świerkot, J. Significance of Polymorphism and Expression of miR-146a and NFkB1 Genetic Variants in Patients with Rheumatoid Arthritis. Arch. Immunol. Ther. Exp. 2016, 64, 131–136. [Google Scholar] [CrossRef] [PubMed]
  104. Yu, Y.; Park, S.; Lee, H.; Kwon, E.J.; Park, H.R.; Kim, Y.H.; Lee, S.-G. Exosomal Hsa-miR-335-5p and Hsa-miR-483-5p Are Novel Biomarkers for Rheumatoid Arthritis: A Development and Validation Study. Int. Immunopharmacol. 2023, 120, 110286. [Google Scholar] [CrossRef]
  105. Wang, L.; Wang, C.; Jia, X.; Yu, J. Circulating Exosomal miR-17 Inhibits the Induction of Regulatory T Cells via Suppressing TGFBR II Expression in Rheumatoid Arthritis. Cell. Physiol. Biochem. 2018, 50, 1754–1763. [Google Scholar] [CrossRef]
  106. Sun, S.; Liang, L.; Tian, R.; Huang, Q.; Ji, Z.; Li, X.; Lin, P.; Zheng, S.; Peng, Y.; Yuan, Q.; et al. LncRNA Expression Profiling in Exosomes Derived from Synovial Fluid of Patients with Rheumatoid Arthritis. Int. Immunopharmacol. 2024, 130, 111735. [Google Scholar] [CrossRef] [PubMed]
  107. Biswas, S.; Sharma, S.; Saroha, A.; Bhakuni, D.S.; Malhotra, R.; Zahur, M.; Oellerich, M.; Das, H.R.; Asif, A.R. Identification of Novel Autoantigen in the Synovial Fluid of Rheumatoid Arthritis Patients Using an Immunoproteomics Approach. PLoS ONE 2013, 8, e56246. [Google Scholar] [CrossRef]
  108. Smith, K.M.; Amin, S.; Jones, L.K.; Lachance, D.H.; Flanagan, E.P.; Jentoft, M.E.; Kantarci, O.H. Glial Fibrillary Acidic Protein (GFAP) Autoimmunity in the Setting of Seropositive Rheumatoid Arthritis Treated With Etanercept. Neurologist 2019, 24, 152–154. [Google Scholar] [CrossRef]
  109. El Atab, O.; Gupta, B.; Han, Z.; Stribny, J.; Asojo, O.A.; Schneiter, R. Alpha-1-B Glycoprotein (A1BG) Inhibits Sterol-Binding and Export by CRISP2. J. Biol. Chem. 2024, 300, 107910. [Google Scholar] [CrossRef]
  110. Brink, M.; Lundquist, A.; Alexeyenko, A.; Lejon, K.; Rantapää-Dahlqvist, S. Protein Profiling and Network Enrichment Analysis in Individuals before and after the Onset of Rheumatoid Arthritis. Arthritis Res. Ther. 2019, 21, 288. [Google Scholar] [CrossRef]
  111. Kim, K.M.; Lee, K.G.; Lee, S.; Hong, B.K.; Yun, H.; Park, Y.J.; Yoo, S.A.; Kim, W.U. The Acute Phase Reactant Orosomucoid-2 Directly Promotes Rheumatoid Inflammation. Exp. Mol. Med. 2024, 56, 890–903. [Google Scholar] [CrossRef]
  112. Dénarié, D.; Constant, E.; Thomas, T.; Marotte, H. Could Biomarkers of Bone, Cartilage or Synovium Turnover Be Used for Relapse Prediction in Rheumatoid Arthritis Patients? Mediat. Inflamm. 2014, 2014, 537324. [Google Scholar] [CrossRef] [PubMed]
  113. Vermeer, M.; Kuper, H.H.; Hoekstra, M.; Haagsma, C.J.; Posthumus, M.D.; Brus, H.L.M.; Van Riel, P.L.C.M.; Van De Laar, M.A.F.J. Implementation of a Treat-to-Target Strategy in Very Early Rheumatoid Arthritis: Results of the Dutch Rheumatoid Arthritis Monitoring Remission Induction Cohort Study. Arthritis Rheum. 2011, 63, 2865–2872. [Google Scholar] [CrossRef] [PubMed]
  114. Hammam, N.; Salah, S.; Kholef, E.F.; Moussa, E.M.; Marotta, A. 14-3-3η Protein in Serum and Synovial Fluid Correlates with Radiographic Damage and Progression in a Longitudinal Evaluation of Patients with Established Rheumatoid Arthritis. Mod. Rheumatol. 2020, 30, 664–670. [Google Scholar] [CrossRef] [PubMed]
  115. Maksymowych, W.P.; Van Der Heijde, D.; Allaart, C.F.; Landewé, R.; Boire, G.; Tak, P.P.; Gui, Y.; Ghahary, A.; Kilani, R.; Marotta, A. 14-3-3η Is a Novel Mediator Associated with the Pathogenesis of Rheumatoid Arthritis and Joint Damage. Arthritis Res. Ther. 2014, 16, R99. [Google Scholar] [CrossRef] [PubMed]
  116. McInnes, I.B.; Schett, G. Cytokines in the Pathogenesis of Rheumatoid Arthritis. Nat. Rev. Immunol. 2007, 7, 429–442. [Google Scholar] [CrossRef]
  117. Mohamed, A.H.M.; Sameh, A.; Essam, H.E.-N. Serum Level of 14-3-3η (Eta) Protein as a Diagnostic Marker for Rheumatoid Arthritis and Potential Correlation with Disease Activity. MOJ Orthop. Rheumatol. 2017, 7, 280. [Google Scholar] [CrossRef]
  118. Tavasolian, F.; Moghaddam, A.S.; Rohani, F.; Abdollahi, E.; Janzamin, E.; Momtazi-Borojeni, A.A.; Moallem, S.A.; Jamialahmadi, T.; Sahebkar, A. Exosomes: Effectual Players in Rheumatoid Arthritis. Autoimmun. Rev. 2020, 19, 102511. [Google Scholar] [CrossRef]
  119. Wu, X.; Liu, Y.; Wei, W.; Liu, M.-L. Extracellular Vesicles in Autoimmune Vasculitis—Little Dirts Light the Fire in Blood Vessels. Autoimmun. Rev. 2019, 18, 593–606. [Google Scholar] [CrossRef]
  120. Stanczyk, J.; Ospelt, C.; Karouzakis, E.; Filer, A.; Raza, K.; Kolling, C.; Gay, R.; Buckley, C.D.; Tak, P.P.; Gay, S.; et al. Altered Expression of microRNA-203 in Rheumatoid Arthritis Synovial Fibroblasts and Its Role in Fibroblast Activation. Arthritis Rheum. 2011, 63, 373–381. [Google Scholar] [CrossRef]
  121. Fu, H.; Hu, D.; Zhang, L.; Tang, P. Role of Extracellular Vesicles in Rheumatoid Arthritis. Mol. Immunol. 2018, 93, 125–132. [Google Scholar] [CrossRef]
  122. Xing, L.; Schwarz, E.M.; Boyce, B.F. Osteoclast Precursors, RANKL/RANK, and Immunology. Immunol. Rev. 2005, 208, 19–29. [Google Scholar] [CrossRef] [PubMed]
  123. De La Poza Plaza, E.; Barykin, S.; Shishkin, V.; Shishkin, V.V.; Kudryavtseva, G.V. AB0234 prognostic potential of Exosomes (EX) as immunomodulators in Synovial Fluid (SF) in Rheumatoid Arthritis (RA). In Proceedings of the Scientific Abstracts; BMJ Publishing Group Ltd. and European League Against Rheumatism: Zürich, Switzerland, 2023; pp. 1301.1–1302. [Google Scholar]
  124. Varela, L.; Van De Lest, C.H.A.; Boere, J.; Libregts, S.F.W.M.; Lozano-Andrés, E.; Van Weeren, P.R.; Wauben, M.H.M. Acute Joint Inflammation Induces a Sharp Increase in the Number of Synovial Fluid EVs and Modifies Their Phospholipid Profile. Biochim. Et Biophys. Acta (BBA)—Mol. Cell Biol. Lipids 2023, 1868, 159367. [Google Scholar] [CrossRef] [PubMed]
  125. Muller, S.; Radic, M. Citrullinated Autoantigens: From Diagnostic Markers to Pathogenetic Mechanisms. Clin. Rev. Allerg. Immunol. 2015, 49, 232–239. [Google Scholar] [CrossRef] [PubMed]
  126. Yoo, J.; Lee, S.K.; Lim, M.; Sheen, D.; Choi, E.-H.; Kim, S.A. Exosomal Amyloid A and Lymphatic Vessel Endothelial Hyaluronic Acid Receptor-1 Proteins Are Associated with Disease Activity in Rheumatoid Arthritis. Arthritis Res. Ther. 2017, 19, 119. [Google Scholar] [CrossRef]
Figure 1. Synovial fluid collection, laboratory analysis, and biomarker detection.
Figure 1. Synovial fluid collection, laboratory analysis, and biomarker detection.
Medicina 60 01945 g001
Figure 2. Healthy joint vs. joint affected by RA.
Figure 2. Healthy joint vs. joint affected by RA.
Medicina 60 01945 g002
Table 1. The categorization of RA markers and their locations and potential associations with the disease.
Table 1. The categorization of RA markers and their locations and potential associations with the disease.
Marker TypeMarker Name/DescriptionLocationAssociation with RAReference
Genetic MarkersHLA-DRB1Genetic–6p21.32The strongest association with RA risk and severity, also with anti-CCP antibodies.[26]
TRAF1/C5Genetic–9q33-34Associated with erosive changes in RA, though of modest clinical utility.[27]
PADI4Genetic–1p36.13Controversial association with RA susceptibility across different populations.[28]
Bone MarkersRANKL/OPG ratioSerum/synoviumPredicts radiographic progression of joint damage.[29]
Collagen (type I) cross-linked C-telopeptide (CTX-I)Serum/urineHigh levels predict the risk of radiological progression and joint destruction.[30]
Bone sialoprotein (BSP)Serum/synovial fluidElevated levels may indicate joint damage and help in the early identification of destructive RA.[31]
Cathepsin KSerum/synovial fluidPotential marker of bone resorption but requires further validation.[32]
OsteocalcinSerumReflects bone metabolism but is less effective at predicting disease activity.[33]
Cartilage MarkersCartilage oligomeric matrix protein (COMP)SerumChanges in COMP linked to cartilage destruction and joint damage in RA.[34]
C-terminal cross-linking telopeptide of type II collagen (CTX-II)UrineHigh levels predict joint destruction progression in RA.[30,35]
Col2-3/4Clong mono and Col2-3/4Cshort (Type II)SerumMay indicate cartilage degradation and predict joint space narrowing.[36]
Hyaluronic acid (HA)Serum/synovial fluidElevated levels correlate with disease activity but are variable throughout the day.[35,37]
AutoantibodiesRheumatoid factor (RF)SerumCommonly detected but not specific to RA; correlates with disease activity and prognosis.[38]
Anti-cyclic citrullinated peptide (anti-CCP)SerumHighly specific for RA and predictive of joint destruction; may be detected before RA onset.[39]
Anti-mutated citrullinated vimentin (MCV)SerumHigh specificity for RA; predicts severe joint involvement.[40]
Antiperinuclear factor (APF)SerumPresent in many RA patients; assists in early intervention.[41]
Inflammatory MarkersErythrocyte sedimentation rate (ESR)BloodReflects disease activity and can predict long-term radiological progression useful in clinical settings.[42]
C-reactive protein (CRP)BloodReflects short-term disease activity and is associated with joint destruction; widely used.[32]
CalprotectinSynovial fluidHigh levels correlate with joint damage and predict erosive disease.[43]
Serum amyloid-associated protein (SAA)BloodCorrelates with clinical parameters; may not provide additional information over CRP.[44]
Cytokines, Inhibitors
Adipokines
Various cytokines (e.g., IL-1, IL-6, TNF-α)Synovial fluidElevated levels contribute to joint destruction; dynamic balance is necessary for predictive value.[45,46]
Soluble TNF receptor II (sTNFR-II)Blood/synovial fluidMay indicate future RA development.[47]
Adipocytokines (e.g., leptin, visfatin)BloodElevated in RA; associated with radiographic joint damage.[48]
Immunological MarkersCD4 + CD25+ T-regulatory cellsSynovial fluidIncreased in inflamed joints; controversial role in RA pathology and potential biomarkers.[49,50]
Table 2. Categorization of discussed biomarkers.
Table 2. Categorization of discussed biomarkers.
MarkerPrognosticDiagnosticTherapeutic
Aggrecan++
MMPs++
Glc-Gal-PYD+
Hyaluronic acid+
S100 proteins+
Calprotectin+
Cytokines++
Immunological markers
ADAMEDC1++
MAGE-I-mRNA+
Non-coding RNAs+++
GFAP and A1BG++
ORM2++
14-3-3η protein++
Table 3. The contents of exosomes and their roles in RA.
Table 3. The contents of exosomes and their roles in RA.
Exosomal Content Source/Type Role in RA Key Markers/Findings Ref.
miRNAs Synovial fluid exosomesModulate inflammation, serve as biomarkers, contribute to joint damage and cartilage degradationmiR-155, miR-150, miR-146a, miR-21, miR-221-3p, miR-335p, miR-483-5p; associated with T cell differentiation and inhibition of bone repair[97,98]
Autoantigens EVs from fibroblast-like synoviocytes (FLS)Participate in the immune response by interacting with citrullinated proteins and presenting autoantigensCitrullinated peptides, MHC-I/II molecules, immunological complexes, IgG, fibrin α-chain, β-chain, fibrinogen[20]
Heat Shock Proteins (HSPs) Exosomes from synovial cellsContribute to inflammatory pathways and exosome secretionElevated Hsp70 and CD9 tetraspanins; increased acidic endonuclease activity and decreased alkaline activity[123]
MMPs Exosomes from FLSPromote cartilage breakdown and angiogenesisMMP-13, MMP-3, IL-6, VEGF; granulocyte-derived vesicles linked to pro-coagulation effects[65]
Lipids Exosomal lipid contentInvolved in synovial inflammation and EV biogenesisHexosylceramides, sphingomyelin; increased phosphatidylethanolamines during acute inflammation[124]
Proteins EVs in synovial fluidAssociated with inflammation, bone destruction, autoantigen presentation, and signaling pathwaysSTAT1, STAT3, JAK1, JAK2, TLR2, MMP9, PYCARD, CCR1, HSP90AB1, F-actin, Histones H1, H2, H3; GTPases, NADPH oxidase, MPO, Rac1, Rac2[125,126]
lncRNAs Exosomal lncRNAsServe as RA biomarkers; modulate inflammation via miRNA interactionENST00000433825.1 (correlates with CRP levels in RA); HIX003209 linked to inflammatory processes[106]
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

Smolinska, V.; Klimova, D.; Danisovic, L.; Harsanyi, S. Synovial Fluid Markers and Extracellular Vesicles in Rheumatoid Arthritis. Medicina 2024, 60, 1945. https://doi.org/10.3390/medicina60121945

AMA Style

Smolinska V, Klimova D, Danisovic L, Harsanyi S. Synovial Fluid Markers and Extracellular Vesicles in Rheumatoid Arthritis. Medicina. 2024; 60(12):1945. https://doi.org/10.3390/medicina60121945

Chicago/Turabian Style

Smolinska, Veronika, Daniela Klimova, Lubos Danisovic, and Stefan Harsanyi. 2024. "Synovial Fluid Markers and Extracellular Vesicles in Rheumatoid Arthritis" Medicina 60, no. 12: 1945. https://doi.org/10.3390/medicina60121945

APA Style

Smolinska, V., Klimova, D., Danisovic, L., & Harsanyi, S. (2024). Synovial Fluid Markers and Extracellular Vesicles in Rheumatoid Arthritis. Medicina, 60(12), 1945. https://doi.org/10.3390/medicina60121945

Article Metrics

Back to TopTop