Skip to content

Acpa-Negative Rheumatoid Arthritis

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by inflammation of the joints, leading to pain, swelling, stiffness, and progressive joint damage. It is a heterogeneous condition, meaning it can manifest differently among individuals. A key differentiator in classifying RA is the presence or absence of specific autoantibodies, particularly anti-citrullinated protein antibodies (ACPAs). ACPA-negative rheumatoid arthritis refers to the subtype of RA where these specific autoantibodies are not detected in a patient’s blood. This distinction is crucial because ACPA-positive and ACPA-negative RA are increasingly recognized as potentially distinct disease entities with differing genetic underpinnings, clinical presentations, and responses to treatment.

Rheumatoid arthritis is broadly categorized into two main groups based on serological status: ACPA-positive RA and ACPA-negative RA. While ACPA-positive RA is often associated with a more aggressive disease course and a stronger association with certain genetic factors, ACPA-negative RA presents unique challenges in diagnosis and research due to the absence of these definitive biomarkers. The importance of distinguishing between these subtypes stems from observations that they may have different genetic predispositions and clinical trajectories.[1] Understanding ACPA-negative RA is essential for developing more precise diagnostic tools and targeted therapies for this specific patient population.

The biological basis of ACPA-negative RA is less clearly defined compared to ACPA-positive RA. Genetic studies, particularly Genome-Wide Association Studies (GWAS), have explored the genetic landscape of both subtypes. While numerous susceptibility loci have been identified for ACPA-positive RA [2] research indicates “little or no overlap” in genetic associations for some tentative SNPs between ACPA-negative and ACPA-positive RA. [1] For instance, a GWAS specifically on ACPA-negative RA found no single SNP reaching genome-wide significance, and the five tentative SNPs identified showed only nominal or no association with ACPA-positive RA. [1] This suggests that ACPA-negative RA may involve different immune pathways or a more complex, perhaps multifactorial, etiology that is not as strongly driven by the genetic factors currently linked to autoantibody production. The challenge in replicating findings for ACPA-negative RA due to the inability to identify additional appropriately-sized case-control studies further highlights the need for more focused research in this area. [1]

The distinction between ACPA-positive and ACPA-negative RA holds significant clinical relevance. ACPA-positive RA is often associated with more severe disease, including rapid joint erosion and a poorer prognosis. While ACPA-negative RA can also be severe, some studies suggest a potentially milder disease course for a subset of these patients. However, the absence of ACPA can make diagnosis more challenging, often delaying the definitive identification of the disease and the initiation of appropriate treatment, as clinicians must rely more heavily on clinical symptoms and other, less specific, inflammatory markers. Furthermore, treatment responses, particularly to certain biologic therapies, may differ between the two subtypes, making accurate classification vital for personalized medicine and guiding therapeutic decisions.

Rheumatoid arthritis, regardless of its subtype, imposes a considerable burden on individuals, healthcare systems, and society at large. For patients with ACPA-negative RA, the lack of specific biomarkers and distinct genetic insights can contribute to diagnostic delays, prolonged uncertainty, and potentially less targeted or effective treatment strategies. Continued research into ACPA-negative RA is critical to elucidate its unique pathophysiology, identify specific diagnostic biomarkers, and develop effective, tailored therapies. Improved understanding and management of ACPA-negative RA will not only enhance the quality of life for affected individuals by reducing pain, disability, and disease progression but also alleviate the broader societal costs associated with chronic illness and healthcare utilization.

Challenges in Study Design and Statistical Power

Section titled “Challenges in Study Design and Statistical Power”

The genetic investigation of ACPA-negative rheumatoid arthritis (RA) faces significant limitations stemming from study design and statistical constraints. Initial genome-wide association studies (GWAS) for ACPA-negative RA have been conducted with relatively small cohorts, such as one with 774 cases and 1079 controls, which limits statistical power to detect associations, resulting in no single single nucleotide polymorphism (SNP) reaching genome-wide significance.[1]This restricted sample size also contributes to the inability to independently replicate tentative findings, a critical step for validating genetic associations, as appropriately-sized replication cohorts for ACPA-negative RA have been difficult to identify.[1]The historical focus of larger GWAS on ACPA-positive RA or combined RA cohorts further exacerbates this issue, leading to a persistent disparity in the available data for ACPA-negative disease, where even in large meta-analyses, ACPA-negative cases represent a substantially smaller proportion.[3]

The limited power in these studies means that potentially significant polymorphisms with smaller effect sizes may be missed in the discovery phase. [4] Furthermore, the lack of strong deviation from the expected distribution in quantile-quantile plots for ACPA-negative RA suggests that the current studies may not be sufficiently powered to uncover many true genetic signals, or that the genetic architecture is characterized by numerous variants with very modest effects. [1] When ACPA-positive and ACPA-negative cases are combined, some loci only reach genome-wide significance, implying distinct genetic underpinnings for the two subtypes and highlighting the challenges in identifying specific genetic drivers for ACPA-negative RA when analyzed separately. [5]

Phenotypic Heterogeneity and Generalizability Across Populations

Section titled “Phenotypic Heterogeneity and Generalizability Across Populations”

The interpretation of genetic findings for ACPA-negative RA is impacted by phenotypic heterogeneity and limitations in generalizability across diverse populations. Current studies often rely on broad diagnostic criteria for RA, such as the 1987 American College of Rheumatology criteria, which may group together individuals with differing underlying pathologies within the ACPA-negative subset, thereby obscuring distinct genetic signals. [3] Furthermore, directly linking identified genetic polymorphisms to specific clinical phenotypes within the ACPA-negative patient population remains challenging, often requiring much larger cohorts to elucidate these genotype-phenotype correlations. [4]

A significant limitation is the predominant focus on populations of European ancestry in many large-scale GWAS and replication cohorts for RA. [6] While some trans-ethnic meta-analyses have begun to include individuals of Asian ancestry, the representation of ACPA-negative cases within these non-European cohorts remains disproportionately small. [3] This ancestry bias limits the generalizability of findings to other ethnic groups and suggests that unique genetic risk factors or differing effect sizes for known variants in ACPA-negative RA across diverse populations may be overlooked, preventing a comprehensive understanding of its global genetic architecture.

Unexplored Genetic and Environmental Factors

Section titled “Unexplored Genetic and Environmental Factors”

A comprehensive understanding of ACPA-negative RA is further constrained by unexplored genetic and environmental factors, contributing to remaining knowledge gaps. Notably, many studies have not thoroughly examined the contribution of the HLA-DRB1 shared epitope, which, while more strongly associated with ACPA-positive RA, still warrants detailed genotyping and linkage analysis in larger ACPA-negative cohorts to fully ascertain its role, if any, in this subtype. [4] The observed genetic associations likely represent only a fraction of the heritability of ACPA-negative RA, indicating that a significant portion of its genetic risk, or “missing heritability,” remains undiscovered, possibly due to rare variants, structural variations, or complex epistatic interactions not fully captured by current GWAS methodologies.

Beyond genetic predispositions, the interplay between genetic variants and environmental exposures is largely unexplored for ACPA-negative RA. While environmental factors are known to influence RA risk, their specific interactions with genetic susceptibility in the context of ACPA-negative disease are not well-defined in the provided research. Future investigations incorporating detailed environmental exposure data alongside comprehensive genetic profiling are essential to unravel these complex gene-environment confounders and provide a more complete etiological picture for ACPA-negative RA.

Genetic variations play a crucial role in determining an individual’s susceptibility to rheumatoid arthritis (RA), including the ACPA-negative subtype. These variants often affect genes involved in immune regulation, T-cell activation, and inflammatory pathways, contributing to the complex etiology of the disease. Understanding these genetic influences provides insight into the underlying mechanisms of RA pathogenesis.

The PTPN22gene, encoding protein tyrosine phosphatase, non-receptor type 22, is a prominent genetic risk factor for rheumatoid arthritis. This gene produces a protein called lymphoid-specific phosphatase (LYP), which acts as a negative regulator of T-cell receptor signaling, essentially putting a “brake” on immune cell activation. Thers2476601 variant, often referred to as R620W, results in a change in the LYP protein that reduces its inhibitory function, leading to hyperactive T cells and an increased risk of autoimmunity. [7] This variant has been reproducibly associated with RA, conferring a genetic relative risk of approximately 1.8. [7] While rs2476601 is strongly linked to PTPN22, the AP4B1-AS1 gene, a long non-coding RNA, is located in the vicinity and may also be influenced by regulatory elements affected by this variant, potentially modulating immune responses relevant to ACPA-negative RA.

Another critical gene is STAT4 (Signal Transducer and Activator of Transcription 4), a transcription factor that transmits signals from various cytokines, such as IL-12 and IL-23, to the cell nucleus, influencing the differentiation of T helper cells (Th1 and Th17) that drive inflammatory responses. Variants within STAT4, including rs4853458 and rs11889341 , are associated with an increased risk of rheumatoid arthritis and systemic lupus erythematosus.[2]These single nucleotide polymorphisms can affectSTAT4expression or its signaling efficiency, thereby promoting a pro-inflammatory state that contributes to autoimmune disease development, including ACPA-negative RA.[7] The enhanced signaling through STAT4 can lead to an imbalance in immune cell populations, fostering chronic inflammation.

Several other variants also contribute to RA susceptibility. The IL2RA gene (Interleukin 2 Receptor Alpha), also known as CD25, is essential for the function of regulatory T cells (Tregs), which maintain immune tolerance. The rs3134883 variant in IL2RA can influence the expression of this receptor, potentially impairing Treg function and contributing to autoimmune conditions like ACPA-negative RA. [5] ANKRD55 (Ankyrin Repeat Domain 55) is a gene whose rs7731626 variant has been linked to autoimmune diseases, potentially by affecting immune cell activation or differentiation pathways relevant to chronic inflammation. [1] Finally, TYK2 (Tyrosine Kinase 2) is a key enzyme in the JAK-STAT signaling pathway, mediating responses to cytokines that are critical for immune cell development and function. The rs34536443 variant in TYK2can alter its enzymatic activity, leading to dysregulated cytokine signaling and an elevated risk of autoimmune diseases, including rheumatoid arthritis, by promoting sustained inflammatory responses.[7] These genetic variations collectively highlight the complex interplay of immune pathways in RA pathogenesis.

RS IDGeneRelated Traits
rs2476601 PTPN22, AP4B1-AS1rheumatoid arthritis
autoimmune thyroid disease, type 1 diabetes mellitus
leukocyte quantity
ankylosing spondylitis, psoriasis, ulcerative colitis, Crohn’s disease, sclerosing cholangitis
late-onset myasthenia gravis
rs7731626 ANKRD55rheumatoid arthritis
autoimmune thyroid disease, systemic lupus erythematosus, type 1 diabetes mellitus, ankylosing spondylitis, psoriasis, common variable immunodeficiency, celiac disease, ulcerative colitis, Crohn’s disease, autoimmune disease, juvenile idiopathic arthritis
multiple sclerosis
red blood cell density
rheumatoid arthritis, ACPA-positive rheumatoid arthritis, rheumatoid factor seropositivity measurement
rs3134883 IL2RAplatelet-to-lymphocyte ratio
lymphocyte count
rheumatoid arthritis
rheumatoid arthritis, ACPA-negative rheumatoid arthritis
rheumatoid arthritis, anti-citrullinated protein antibody seropositivity, rheumatoid factor seropositivity measurement
rs4853458
rs11889341
STAT4rheumatoid arthritis, ACPA-positive rheumatoid arthritis, rheumatoid factor seropositivity measurement
rheumatoid arthritis
rheumatoid arthritis, ACPA-negative rheumatoid arthritis
systemic scleroderma
thyroid disease
rs34536443 TYK2psoriasis vulgaris
platelet count
rheumatoid arthritis
psoriasis
multiple sclerosis

Classification, Definition, and Terminology

Section titled “Classification, Definition, and Terminology”

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease primarily characterized by inflammation of the joints, leading to pain, swelling, stiffness, and potential joint damage. The diagnosis and classification of RA traditionally rely on established criteria, such as the American Rheumatism Association (ACR) 1987 revised classification criteria.[8] While these criteria provide a standardized framework, a diagnosis can also be based on a comprehensive clinical evaluation by a board-certified Rheumatologist. [9] These foundational criteria are essential for consistently identifying RA patients, which is crucial for both clinical practice and research studies.

The ACPA-Negative Subtype and its Genetic Significance

Section titled “The ACPA-Negative Subtype and its Genetic Significance”

ACPA-negative rheumatoid arthritis represents a specific subtype of RA defined by the absence of antibodies to citrullinated peptide antigens (ACPA).[1] This distinguishes it from ACPA-positive RA, where these autoantibodies are detectable. [1]Related autoantibodies, such as anti-citrullinated protein antibodies (CCP) and Rheumatoid Factor (RF), also play a role in classifying RA and other rheumatic conditions like Juvenile Idiopathic Arthritis (JIA), which includes subtypes such as “Polyarthritis, RF negative” and “Polyarthritis, RF positive”.[6] The distinction between ACPA-positive and ACPA-negative RA is critical in scientific research, particularly in genome-wide association studies (GWAS), as these subgroups often exhibit contrasting genetic associations with little or no overlap for certain genetic variants, indicating potentially distinct pathophysiological pathways. [1]

The clinical management and research of rheumatoid arthritis, including its ACPA-negative subtype, depend on robust methods for assessing disease activity and treatment response. The Disease Activity Score 28 (DAS28), specifically the DAS28CRP3 variant, is a widely used operational definition that incorporates the count of tender and swollen joints (out of 28) and C-reactive protein (CRP) levels to provide a quantitative measure of disease activity.[10]Modified disease activity scores that include twenty-eight-joint counts have been developed and validated to enhance precision in evaluating disease status.[11]Furthermore, the European League Against Rheumatism (EULAR) response criteria are employed to evaluate the effectiveness of therapeutic interventions, providing standardized thresholds for defining clinical improvement or remission.[12]These tools are indispensable for monitoring disease progression, guiding therapeutic strategies, and serving as outcome measures in both clinical trials and observational studies.

Clinical Presentation and Diagnostic Classification

Section titled “Clinical Presentation and Diagnostic Classification”

Patients diagnosed with ACPA-negative rheumatoid arthritis (RA) are defined by their fulfillment of the 1987 criteria of the American College of Rheumatology (ACR) for RA, while testing negative for anti-citrullinated peptide antibodies (ACPA).[13]These classification criteria provide the clinical framework for diagnosis, typically involving objective signs of inflammatory arthritis identified through clinical evaluation by a board-certified rheumatologist.[8] The absence of ACPA is the distinguishing serological feature, setting this subtype apart from ACPA-positive RA, even as the fundamental presentation aligns with general RA classifications emphasizing inflammatory joint involvement. [1]

The defining serological characteristic of ACPA-negative RA is the absence of anti-citrullinated peptide antibodies, which serves as a critical diagnostic biomarker.[13]Beyond this seronegativity, the activity of the disease is quantitatively assessed using standardized tools such as the Disease Activity Score in 28 joints (DAS28). [10]This comprehensive score incorporates various objective and subjective measures, including 28-joint counts and C-reactive protein (CRP) levels, providing a valuable indicator of inflammation and overall disease severity in affected individuals.[10]

Phenotypic Heterogeneity and Diagnostic Considerations

Section titled “Phenotypic Heterogeneity and Diagnostic Considerations”

The distinct serological profile of ACPA-negative RA suggests a unique clinical phenotype, differentiating it from ACPA-positive RA. [6]Genetic studies further support this distinction, revealing contrasting genetic associations between ACPA-negative and ACPA-positive RA, which implies different underlying biological pathways and potential variations in disease progression.[1] Consequently, the diagnosis of ACPA-negative RA relies heavily on the careful application of established clinical classification criteria, given the absence of this specific autoantibody biomarker. [13]

The genetic underpinnings of ACPA-negative rheumatoid arthritis (RA) appear to differ significantly from those of ACPA-positive RA, presenting a distinct genetic landscape. Genome-wide association studies (GWAS) specifically analyzing ACPA-negative RA have generally not identified single nucleotide polymorphisms (SNPs) that reach genome-wide significance.[1]This suggests a more complex, potentially polygenic architecture where multiple common variants of smaller effect collectively contribute to disease risk, rather than a few strong genetic drivers. Furthermore, initial findings indicate little to no overlap in the specific genetic variants tentatively associated with ACPA-negative RA compared to those linked with ACPA-positive RA.[1]

While some studies have identified five SNPs from three genetic loci with suggestive p-values (<10^-5) in ACPA-negative RA, these have not been consistently replicated due to the challenges in assembling sufficiently large, independent case-control cohorts for this specific subgroup. [1] This contrasts with ACPA-positive RA, where numerous susceptibility loci have been robustly identified. The lack of strong, consistent genetic signals in ACPA-negative RA underscores the possibility that its etiology involves a different set of genetic risk factors, or that the genetic contribution is more diffuse and harder to pinpoint with current methodologies.

Environmental factors and demographic characteristics are considered in the design of studies investigating ACPA-negative RA, though specific causal mechanisms are not detailed within the provided context. For instance, controls in genetic studies are often selected by matching for age, sex, and residential area. [1]This methodological approach indirectly highlights the potential influence of geographic or local environmental exposures, as well as age and sex, on disease development or presentation, by ensuring that differences observed between cases and controls are less likely to be attributable to these broad factors. However, the exact nature of how specific environmental elements, such as lifestyle, diet, or unique exposures, contribute to the onset or progression of ACPA-negative RA is not elaborated upon in the available research.

Biological Background of ACPA-Negative Rheumatoid Arthritis

Section titled “Biological Background of ACPA-Negative Rheumatoid Arthritis”

Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation, primarily affecting the joints.[6]The disease exhibits considerable variability in its outcomes, and the presence or absence of antibodies to citrullinated peptide antigens (ACPA) serves as a significant clinical predictor of disease severity.[1] ACPA-negative RA represents a distinct clinical subset, differing from ACPA-positive RA in its genetic underpinnings and potentially in its pathophysiological mechanisms. [1] Understanding the unique biological characteristics of ACPA-negative RA is crucial for developing targeted diagnostic and therapeutic strategies.

Genetic Architecture and Subtype Specificity

Section titled “Genetic Architecture and Subtype Specificity”

The genetic landscape of RA is complex, involving an interplay of various genetic variants and environmental exposures. [1] Notably, ACPA-positive and ACPA-negative RA display significant differences in risk allele frequencies, primarily within the human leukocyte antigen (HLA) region. [1] While variants of HLA-DRB1 and PTPN22 are strongly associated with ACPA-positive RA, the HLA-DR3 allele shows an association with ACPA-negative RA. [2] Furthermore, specific HLA-DRB1*13 alleles exhibit opposing effects on the risk of developing ACPA-positive versus ACPA-negative RA, underscoring the distinct genetic etiologies of these subsets. [14]

Beyond the HLAregion, a single nucleotide polymorphism (SNP) near theRPS12P4 locus on chromosome 2 has been identified as a tentative candidate locus for ACPA-negative RA. [1] In contrast, genes like PADI4, which are involved in the citrullination of peptides leading to the formation of ACPA autoantibodies, are more strongly implicated in ACPA-positive disease.[5] Other genetic loci, such as STAT4, TRAF1-C5, CTLA4, IRAK1 (on the X chromosome, relevant given RA’s female predominance), ARID5B, IKZF3, APOM and REL have been associated with RA overall, but their specific roles or differential associations in ACPA-negative RA require further elucidation. [2]

Immune Cell Dysregulation and Molecular Signaling

Section titled “Immune Cell Dysregulation and Molecular Signaling”

Rheumatoid arthritis pathogenesis involves complex immune cell dysregulation and intricate signaling pathways. T-cells are central to this process, with genes likeRBPJ, a transcription factor in the Notch signaling pathway critical for T-cell development, and CD247, encoding the T-cell receptor zeta chain involved in signal transduction, highlighting their importance. [15] The PTPN22 gene, while primarily linked to ACPA-positive RA, encodes a protein tyrosine phosphatase that alters T-cell activation, influencing immune responses. [15] Additionally, CD84 functions as a homophilic adhesion molecule and enhances interferon-gamma secretion, contributing to T-cell activation and inflammatory responses. [16]

The NF-kappaB family of transcription factors, including REL, also plays a crucial role in regulating immune and inflammatory responses, with REL identified as a risk locus for RA. [9] Moreover, the IRF5 gene, encoding an interferon regulatory factor, has a promoter region haplotype associated with RA, influencing immune regulation. [17] These molecular and cellular pathways, involving key proteins, enzymes, and transcription factors, contribute to the complex regulatory networks that underpin immune dysregulation in RA, with distinctions likely present between ACPA-positive and ACPA-negative subsets.

Inflammatory Mediators and Complement Pathways

Section titled “Inflammatory Mediators and Complement Pathways”

The inflammatory processes in RA are driven by a cascade of molecular mediators, including cytokines and components of the complement system. The IL6R locus, with the non-synonymous SNP rs8192284 , is associated with circulating IL6R levels and is linked to RA risk, making IL6a critical cytokine in inflammation.[5] Another key molecule, CCR6, a cell surface protein that distinguishes Th17 cells from other helper T-cells, plays a role in inflammation, as synoviocytes from arthritic joints produce its ligand, CCL20. [15]The innate immune system is critically involved in arthritis, with several players contributing to disease progression.[18]

The complement system, particularly the C5 component, also contributes significantly to antibody-mediated inflammation in RA. [2]Studies have shown that C5-deficient mice are resistant to collagen-induced arthritis, and anti-C5 monoclonal antibody therapy can prevent and ameliorate established disease.[19] The TRAF1-C5locus, linked to complement activation, is a confirmed risk locus for RA, further implicating these molecular pathways in disease pathophysiology.[2] These biomolecules and their associated pathways represent crucial targets for understanding and modulating the inflammatory response in RA, including the ACPA-negative subset.

Rheumatoid arthritis is characterized by chronic inflammation predominantly affecting the joints, leading to progressive joint damage.[6] The severity of joint destruction in RA is influenced by genetic predisposition. [20]While the specific mechanisms that differentiate joint destruction in ACPA-negative versus ACPA-positive RA are still being elucidated, the distinct genetic profiles suggest variations in how the disease manifests at the tissue level. For instance, the involvement ofCCR6 in Th17 cell activity and its ligand CCL20 produced by synoviocytes points to specific cellular interactions within the joint microenvironment. [15]

Systemically, RA is an autoimmune condition with broader consequences beyond the joints. The female predominance in RA (a 3:1 ratio of females to males) highlights potential sex-linked biological factors, such as the IRAK1locus on the X chromosome, which may occasionally escape X inactivation in female cells and contribute to disease susceptibility.[5] Environmental factors, such as smoking, interact with genetic predispositions like the HLA-DR shared epitope genes, significantly increasing the risk of seropositive RA. [21] This gene-environment interaction underscores the complex etiology of RA and suggests that different environmental triggers or their interactions with genetic factors might also contribute to the distinct clinical presentation of ACPA-negative RA.

Differential Immune Recognition and Antigen Presentation

Section titled “Differential Immune Recognition and Antigen Presentation”

The pathogenesis of ACPA-negative rheumatoid arthritis (RA) is characterized by distinct genetic etiologies, primarily differing from ACPA-positive RA within the human leukocyte antigen (HLA) region.[1] This region, critical for presenting antigens to T-cells, harbors alleles that influence susceptibility to ACPA-negative RA. For instance, HLA-DR3 has been specifically associated with ACPA-negative RA, suggesting a unique profile of antigen processing and presentation that drives the immune response in this subset. [22] Furthermore, specific HLA-DRB1*13alleles exhibit opposing effects on the risk of developing ACPA-positive versus ACPA-negative RA, highlighting a fundamental divergence in the immune recognition pathways underlying these disease phenotypes.[14]These distinct HLA associations imply that different sets of self-antigens, or a different manner of presenting them, initiate and perpetuate the inflammatory cascade in ACPA-negative disease.

Modulation of Lymphocyte Signaling and Activation

Section titled “Modulation of Lymphocyte Signaling and Activation”

Beyond antigen presentation, various intracellular signaling cascades contribute to the dysregulated immune cell activation seen in ACPA-negative RA. The CD247gene, encoding the T-cell receptor zeta chain, is a crucial component of the T-cell receptor-CD3 complex that couples antigen recognition to downstream signal transduction pathways, and its mutation can lead to inflammatory arthritis.[15] Another relevant locus is IRAK1, an X chromosome gene previously linked to systemic lupus erythematosus, which plays a role in innate immune signaling and has been identified as a novel association for RA. [5] Transcription factors such as RBPJ, a component of the Notch signaling pathway essential for T-cell development, and REL, a member of the NF-kappaB family involved in immune responses, regulate gene expression critical for lymphocyte proliferation, differentiation, and survival, with their dysregulation potentially contributing to the autoimmune state. [15]

The perpetuation of inflammation in ACPA-negative RA involves complex cytokine networks and signaling pathways. TheIL6Rlocus, encoding the receptor for interleukin-6 (IL-6), is associated with RA, and its ligand IL-6 is a key inflammatory cytokine targeted by biologic therapies like tocilizumab.[5] The TRAF1-C5 locus represents another risk factor for RA, implicating the tumor necrosis factor receptor-associated factor 1 (TRAF1) and complement component 5 (C5) pathways in inflammatory responses, where C5has been shown to be critical for antibody-mediated inflammation and arthritis in animal models.[2] Additionally, CCR6, a cell surface protein that distinguishes Th17 cells, is involved in recruiting these pro-inflammatory T-cells to affected joints, as synoviocytes produce its ligand CCL20, making anti-CCR6antibodies a potential therapeutic strategy for mitigating arthritis.[15]

Genetic Predisposition and Regulatory Loci

Section titled “Genetic Predisposition and Regulatory Loci”

The overall genetic architecture of ACPA-negative RA demonstrates significant differences compared to its ACPA-positive counterpart, underscoring the need for separate genetic and functional studies. [1]While the major differences are concentrated in the HLA region, other tentative candidate loci contribute to susceptibility. For instance, a single nucleotide polymorphism (SNP) near theRPS12P4locus on chromosome 2 has been identified as a tentative candidate locus for ACPA-negative RA, suggesting its potential role in ribosomal protein synthesis or other cellular processes that could influence disease development.[1]These specific genetic variations likely influence gene regulation, protein modification, and other molecular controls, leading to altered cellular functions and ultimately contributing to the unique clinical presentation and disease course of ACPA-negative RA.

Genetic Distinction and Diagnostic Implications

Section titled “Genetic Distinction and Diagnostic Implications”

ACPA-negative rheumatoid arthritis is recognized as genetically distinct from ACPA-positive rheumatoid arthritis, with studies indicating minimal or no overlap in specific genetic associations identified to date.[1] This genetic heterogeneity suggests that ACPA-negative RA likely arises from different underlying molecular pathways compared to its ACPA-positive counterpart. [1]Such distinctions are crucial for refining diagnostic utility, moving beyond broad clinical criteria to potentially more precise, genetically informed classifications of rheumatoid arthritis subsets. The unique genetic landscape of ACPA-negative RA highlights the need for dedicated research to uncover its specific biomarkers, which could lead to more accurate diagnoses and a better understanding of its pathogenesis.[1]

The distinct genetic underpinnings of ACPA-negative rheumatoid arthritis imply potentially different disease courses and prognoses compared to ACPA-positive disease.[1] This genetic divergence also suggests that responses to various therapies may differ between the two subsets, necessitating a nuanced approach to treatment selection. For instance, while ACPA seropositivity was not a statistically significant predictor of anti-tumor necrosis factor (TNF) response in one Japanese cohort, the very inclusion of ACPA status in pharmacogenomic analyses underscores its potential role in guiding therapeutic decisions. [10] Understanding these unique genetic profiles could pave the way for personalized medicine, where treatments are tailored to the specific molecular pathways driving ACPA-negative RA, optimizing patient outcomes and minimizing ineffective therapies. [1]

Overlapping Phenotypes and Risk Stratification

Section titled “Overlapping Phenotypes and Risk Stratification”

Genetic research indicates that ACPA-negative rheumatoid arthritis may share genetic linkages and pathogenic pathways with autoimmune entities distinct from those associated with ACPA-positive RA.[1] For example, ACPA-positive RA has established genetic associations, such as with PTPN22, which are also observed in type I diabetes. [1]This suggests that ACPA-negative RA might have a unique spectrum of associated conditions or overlapping phenotypes. Further genetic insights are expected to refine the classification of criterion-based diseases like RA, providing indications of the specific molecular pathways involved in different disease subsets.[1] This improved understanding is vital for more precise risk stratification, enabling the identification of high-risk individuals and the development of prevention strategies tailored to the distinct mechanisms underlying ACPA-negative RA.

Frequently Asked Questions About Acpa Negative Rheumatoid Arthritis

Section titled “Frequently Asked Questions About Acpa Negative Rheumatoid Arthritis”

These questions address the most important and specific aspects of acpa negative rheumatoid arthritis based on current genetic research.


1. Why did it take so long for my doctor to figure out I have RA?

Section titled “1. Why did it take so long for my doctor to figure out I have RA?”

Because you have ACPA-negative RA, which lacks the specific antibody biomarkers doctors usually look for. This means diagnosis relies more heavily on your symptoms and less specific inflammatory markers, often leading to delays in definitive identification.

2. My friend has RA, but her symptoms seem different than mine. Why?

Section titled “2. My friend has RA, but her symptoms seem different than mine. Why?”

Rheumatoid arthritis is a very diverse condition. Your ACPA-negative RA and your friend’s ACPA-positive RA are increasingly recognized as potentially distinct disease entities. They can have different genetic underpinnings, clinical presentations, and even responses to treatment.

3. Is my ACPA-negative RA less severe than other types of RA?

Section titled “3. Is my ACPA-negative RA less severe than other types of RA?”

It depends. While ACPA-positive RA is often associated with a more aggressive disease course, some studies suggest a potentially milder disease course for a subset of ACPA-negative patients. However, ACPA-negative RA can still be severe and cause significant joint damage.

4. Why don’t doctors have a simple blood test just for my type of RA?

Section titled “4. Why don’t doctors have a simple blood test just for my type of RA?”

It’s a significant challenge because ACPA-negative RA lacks the specific autoantibody biomarkers that define ACPA-positive RA. Researchers are actively working to identify unique diagnostic biomarkers for ACPA-negative RA to improve and speed up diagnosis.

5. Should I expect my treatment to be different from someone with ACPA-positive RA?

Section titled “5. Should I expect my treatment to be different from someone with ACPA-positive RA?”

Potentially, yes. There’s evidence that responses to certain biologic therapies may differ between the two subtypes. Your doctor will consider your specific ACPA-negative diagnosis when guiding therapeutic decisions to personalize your treatment plan.

6. Will my children definitely inherit my ACPA-negative RA?

Section titled “6. Will my children definitely inherit my ACPA-negative RA?”

Not necessarily. While there’s a genetic component to RA, the genetic links for ACPA-negative RA are less clearly defined and often don’t overlap with those for ACPA-positive RA. It’s considered a complex, multifactorial condition, not simply passed down through one gene.

7. Why do scientists seem to know less about my kind of RA?

Section titled “7. Why do scientists seem to know less about my kind of RA?”

Historically, much of the research, especially large genetic studies, has focused on ACPA-positive RA. This has resulted in fewer specific genetic insights and challenges in studying ACPA-negative RA, making its biological basis less understood compared to the ACPA-positive form.

8. Does my family history of RA mean anything for my specific ACPA-negative diagnosis?

Section titled “8. Does my family history of RA mean anything for my specific ACPA-negative diagnosis?”

Family history generally indicates a higher risk for RA. However, for ACPA-negative RA, the specific genetic predispositions are still being uncovered and appear to be different from those linked to ACPA-positive RA. It suggests a general susceptibility but doesn’t pinpoint specific genetic pathways for your subtype.

9. Can a genetic test tell me how my ACPA-negative RA might progress?

Section titled “9. Can a genetic test tell me how my ACPA-negative RA might progress?”

Currently, genetic tests are not routinely used to predict the progression of ACPA-negative RA. Due to the less defined genetic landscape and the absence of strong single genetic associations, researchers are still working to identify specific markers that could offer such predictive insights.

10. Why is it so hard for researchers to find clear answers about ACPA-negative RA?

Section titled “10. Why is it so hard for researchers to find clear answers about ACPA-negative RA?”

A major hurdle is the small size of study cohorts for ACPA-negative RA, which limits the statistical power needed to detect significant genetic associations. This also makes it difficult to replicate findings, which is crucial for validating research results and advancing understanding.


This FAQ was automatically generated based on current genetic research and may be updated as new information becomes available.

Disclaimer: This information is for educational purposes only and should not be used as a substitute for professional medical advice. Always consult with a healthcare provider for personalized medical guidance.

[1] Padyukov, L, et al. “A genome-wide association study suggests contrasting associations in ACPA-positive versus ACPA-negative rheumatoid arthritis.”Ann Rheum Dis, 2011.

[2] Plenge, R. M. et al. “STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus.”N Engl J Med, vol. 357, 2007, pp. 977–986.

[3] Okada, Yuta, et al. “Genetics of rheumatoid arthritis contributes to biology and drug discovery.”Nature, vol. 506, no. 7486, 2014, pp. 37-43.

[4] Hu, H. J., et al. “Common variants at the promoter region of the APOM confer a risk of rheumatoid arthritis.”Exp Mol Med, vol. 43, no. 11, 2011, pp. 770-776.

[5] Eyre, S, et al. “High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis.”Nature Genetics, vol. 45, no. 5, 2013, pp. 511–16. PMID: 23143596.

[6] Raychaudhuri, S, et al. “Common variants at CD40and other loci confer risk of rheumatoid arthritis.”Nature Genetics, vol. 40, no. 11, 2008, pp. 1313–18. PMID: 18794853.

[7] Wellcome Trust Case Control Consortium. “Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls.” Nature, vol. 447, no. 7145, 2007, pp. 661-78. PMID: 17554300.

[8] Arnett, FC, et al. “The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis.”Arthritis & Rheumatism, vol. 31, no. 3, 1988, pp. 315–24.

[9] Gregersen, P. K., et al. “REL, encoding a member of the NF-kappaB family of transcription factors, is a newly defined risk locus for rheumatoid arthritis.”Nat Genet, vol. 41, no. 7, 2009, pp. 820-32. PMID: 19503088.

[10] Honne, K, et al. “A longitudinal genome-wide association study of anti-tumor necrosis factor response among Japanese patients with rheumatoid arthritis.”Arthritis Res Ther, 2016.

[11] Prevoo, ML, et al. “Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis.”Arthritis & Rheumatism, vol. 38, no. 1, 1995, pp. 44–48.

[12] Cui, J, et al. “Genome-wide association study and gene expression analysis identifies CD84as a predictor of response to etanercept therapy in rheumatoid arthritis.”PLoS Genetics, vol. 9, no. 4, 2013, e1003444. PMID: 23555300.

[13] Wei, W. H., et al. “Genotypic variability based association identifies novel non-additive loci DHCR7 and IRF4in sero-negative rheumatoid arthritis.”Scientific Reports, vol. 7, no. 1, 2017, p. 5371.

[14] Lundström, E., et al. “Opposing effects of HLA-DRB1*13 alleles on the risk of developing anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis.”Arthritis Rheum, vol. 60, no. 4, 2009, pp. 924-930.

[15] Stahl, E. A., et al. “Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci.”Nat Genet, vol. 42, no. 6, 2010, pp. 508-514.

[16] Martin, M., et al. “CD84 functions as a homophilic adhesion molecule and enhances IFN-gamma secretion: adhesion is mediated by Ig-like domain 1.” Journal of Immunology, vol. 167, no. 7, 2001, pp. 3668-3676.

[17] Sigurdsson, S., et al. “Association of a haplotype in the promoter region of the interferon regulatory factor 5 gene with rheumatoid arthritis.”Arthritis & Rheumatism, vol. 56, no. 7, 2007, pp. 2202-2210.

[18] Ji, Hong, et al. “Arthritis critically dependent on innate immune system players.”Immunity, vol. 16, no. 1, 2002, pp. 157-168.

[19] Wang, Yang, et al. “A role for complement in antibody-mediated inflammation: C5-deficient DBA/1 mice are resistant to collagen-induced arthritis.”Journal of Immunology, vol. 164, no. 8, 2000, pp. 4340-4347.

[20] Knevel, Rachel, et al. “Genetic predisposition of the severity of joint destruction in rheumatoid arthritis: a population-based study.”Annals of the Rheumatic Diseases, vol. 71, no. 5, 2012, pp. 707-709.

[21] Padyukov, Leonid, et al. “A gene-environment interaction between smoking and shared epitope genes in HLA-DR provides a high risk of seropositive rheumatoid arthritis.”Arthritis & Rheumatism, vol. 50, no. 10, 2004, pp. 3085-3092.

[22] Verpoort, K. N., et al. “Association of HLA-DR3 with anti-cyclic citrullinated peptide antibody-negative rheumatoid arthritis.”Arthritis Rheum, vol. 52, no. 10, 2005, pp. 3058-3062.