Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1532 2023-11-14 12:09:57 |
2 update references and layout Meta information modification 1532 2023-11-15 02:53:27 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Paroli, M.; Sirinian, M.I. Seronegative Rheumatoid Arthritis. Encyclopedia. Available online: https://encyclopedia.pub/entry/51536 (accessed on 30 July 2024).
Paroli M, Sirinian MI. Seronegative Rheumatoid Arthritis. Encyclopedia. Available at: https://encyclopedia.pub/entry/51536. Accessed July 30, 2024.
Paroli, Marino, Maria Isabella Sirinian. "Seronegative Rheumatoid Arthritis" Encyclopedia, https://encyclopedia.pub/entry/51536 (accessed July 30, 2024).
Paroli, M., & Sirinian, M.I. (2023, November 14). Seronegative Rheumatoid Arthritis. In Encyclopedia. https://encyclopedia.pub/entry/51536
Paroli, Marino and Maria Isabella Sirinian. "Seronegative Rheumatoid Arthritis." Encyclopedia. Web. 14 November, 2023.
Seronegative Rheumatoid Arthritis
Edit

Seronegative rheumatoid arthritis (SNRA) is characterized by the absence of both rheumatoid factor (RF) and antibodies against the cyclic citrullinated protein (ACPA) in serum.

seronegative rheumatoid arthritis rheumatoid factor anti-citrullinated protein antibodies

1. Introduction

Rheumatoid arthritis (RA) is an inflammatory disease that primarily affects synovial joints through an autoimmune mechanism. If not treated properly, the disease can lead to bone erosion, joint deformities, and disability. Arthritis can also cause serious extra-articular disorders, including interstitial lung disease, vasculitis, and lymphoma [1][2]. According to the latest 2010 ACR/EULAR criteria [3], the diagnosis is based on a scoring system calculated using symptom duration, the number and type of joints affected, altered acute-phase reactants, and the presence of autoantibodies, such as rheumatoid factor (RF) and/or anti-citrullinated protein antibodies (ACPAs), in serum [4]. Because the presence of RF and ACPA in serum is not necessary for the diagnosis of RA, a substantial number of patients presenting with the typical clinical features of RA in the absence of these autoantibodies can be diagnosed as having RA. The form of RA without RF and/or ACPA is termed seronegative RA (SNRA) [5][6]. Numerous observations have reported that the clinical presentation, course severity, and response to therapy appear to be significantly different between SNRA and seropositive RA (SPRA) [7][8][9]. In recent years, the focus on seronegative forms of RA has increased due to clinicians’ sensitivity to the different clinical presentations of RA, as well as the advent of increasingly sophisticated means of both molecular and imaging investigations. In addition, the availability of therapeutic means that can act on different effector functions of immunity has indirectly clarified further distinctive aspects between SNRA and SPRA.

2. Epidemiology

Available epidemiological data have traditionally reported a lower prevalence of SNRA than SPRA, ranging from 20 to 30 percent of total cases of RA [10][11]. However, the incidence of SNRA has been reported to be increased in recent decades [12][13]. Many hypotheses have been advanced to explain this finding. One possible cause is the increasing age of the general population. In fact, late-onset RA occurring in elderly patients is commonly seronegative, suggesting that the dysregulation of inflammation, typical in people of old age, may underlie SNRA [14][15][16]. Another cause is an overall reduction in smoking habits, with cigarette smoking being a strong risk factor for protein citrullination [17]. It is believed that the process of citrullination, by changing the self-nature of joint antigens to non-self-antigens, induces an autoimmune process that leads to the generation of ACPA and causes the humoral and cellular immune systems to attack the altered joint antigens, resulting in synovitis with tissue damage. Therefore, it is likely that the reduction in smoking increased the incidence ratio between SNRA and the seropositive forms through this immunological mechanism [18]. Other factors reported to explain the increased incidence/prevalence of SNRA are changes in the microbiome, possibly from chronic triggers by gut flora via microbial DNA and pepdidoglycans [19], and some environmental factors, including increased occupational exposure to crystalline free silica [20]. Of course, further studies are needed to clarify whether the upward trend in the incidence/prevalence of SNRA compared with that of SNPA is a finding that can proceed over time and whether there are additional genetic and/or environmental factors causing it, which have yet to be elucidated.

3. Pathogenesis

The first distinction between SNRA and SPRA is their different genetic backgrounds. Although RA is a polygenic disease, some genetic risk factors have been identified for SNRA. Among them, the HLA-B*08/DRB1*03 haplotype is one of the genetic markers most frequently associated with SNRA, while the classical HLA-DRB1*04 and *10 alleles have been shown to be risk factors exclusively for SPRA [21][22]. Non-HLA genes also play a relevant role in determining susceptibility to RA, including mutations in genes coding for Janus kinase (JAK)/signal transducer and activator of transcription (STAT) proteins, which are risk factors for SPRA but not for SNRA [23][24]. A genome-wide association study revealed an association with single-nucleotide polymorphism of non-HLA genes ANKRD55 [25] and CLYBL [26] in SNRA but not in SPRA. It is possible to speculate that mutations in non-HLA genes represent changes in the innate-type immune response through the modulation of the synthesis of cytokines and other soluble factors. Indeed, innate immunity seems to be more prominent in SNRA than in SPRA, while antigen/autoantigen presentation to T lymphocytes by HLA molecules represents a key element of adaptive immunity that is more typical of SPRA pathogenesis.
The study of the expression of miRNA is a new exciting field of research aiming to identify biomarkers for differentiating SNRA from SPRA. In this regard, it was recently reported that the miRNAs has-miR-362-5p and has-miR-708-3p were upregulated in SNRA but not in SPRA. Other miRNAs were found to be downregulated differently in the two forms of RA, including the mRNAs expressed exclusively in SPRA and others common to both forms [27][28]. Table 1 summarizes the different genetic backgrounds and miRNA expressions between SNRA and SPRA.
Table 1. Susceptibility to SNRA and SPRA is favored by the presence of HLA and non-HLA genetic factors and miRNA expression.
Some studies were conducted to determine the differences between SPRA and SNRA at the cellular level. The synovial histological score for CD4+ T cells, CD68+ cells in the lining layer, and sublining CD3+ and vessel CD31+ positive cells was less abundant in undifferentiated seronegative arthritis than in differentiated SPRA [28]. It has also been reported that synovium-infiltrating monocytes and macrophages predominate in SNRA [21]. In an attempt to identify biomarkers that can differentiate SNRA from psoriatic arthritis (PsA) because they share some clinical features, a study was conducted that analyzed the synovial histopathology of the two diseases. It was reported that plasma cells predominate in the synovium of SNRA, while mast cells predominate in PsA [31].
An immunohistochemical analysis of the synovium also revealed a higher percentage of tissue-resident dendritic cells and a reduced expression of the PD-1 inhibitory receptor on T cells in SNRA compared with its seropositive counterpart [32]. Therefore, the finding that the immune checkpoint inhibitor PD-1 can induce SNRA in the course of cancer therapy is of particular interest [33]. Table 2 shows the inflammatory cells detected in the synovial membrane during SNRA and SPRA.
Table 2. Immunological features of SNRA and SPRA.
Interestingly, SNRA occurrence has been reported during asthma therapy with anti-IL4/IL-13 biologics with the activation of the IL-23-IL-17 axis, suggesting a protective role of T helper-2 (TH-2) cells in the disease [34]. This evidence further supports the idea that SNRA is a form of RA that diverges substantially from SPRA and suggests a similarity of SNRA with SpA, which depends primarily on IL-17 [5][35][36]. Several observations point out that SNRA has a more variable outcome, generally associated with a better prognosis than SPRA [37][38]. It is interesting to note the reported association between SNRA with NLRP3 inflammasome activation. In this regard, studies have demonstrated a role for interleukin-beta (IL1β), a key component of the inflammasome, in the pathogenesis of SNRA [39]. The pathogenetic relationship between SNRA and IL-1β may explain the favorable response to the interleukin-1 receptor antagonist (IL-1ra), as observed in some patients with SNRA, and the minor response to JAK inhibitor (JAKi) therapy of SNRA compared with SPRA, as reported in some studies [40][41][42]. This can be related to the fact that IL-1 does not depend on the JAK/STAT transduction pathway. As is well known, the activation of the NLRP3 inflammasome by monosodium urate crystals with the release of IL-1β plays a major role in the initiation of gout flare [43]. Interestingly, elevated uric acid levels and crystal deposition are occasionally observed in SNRA but not in SPRA, indirectly suggesting an at least partially autoinflammatory nature of SNRA [43]. Although it is not easy to give an explanation for these observations, they suggest a possible pathogenetic link between SNRA and crystal deposition arthritis. Similarly, an autoinflammatory nature has also been proposed for spondyloarthritis (SpA) [44]. To elucidate the possible autoinflammatory component of SNRA, further studies using methods to study the inflammasome and the genetic substrate of this form of RA are needed. In addition, it should be noted that the study of synovial histology is providing very promising results due to the precise characterization of the cells that infiltrate this tissue. Using the methods of histochemistry and flow cytometry, many research groups are trying to identify new biomarkers that can differentiate SNRA from SPRA. Although synovial biopsy is an invasive procedure, it cannot be ruled out that, in the near future, the results obtained may allow for the development of serologic tests that allow for differential diagnosis through simpler diagnostic tests. Pathogenetic characterization, of course, not only has a scientific or diagnostic purpose but also appears essential for the setting of targeted therapies and the possible realization of the so-called personalized therapy tailored to the individual.

References

  1. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016, 388, 2023–2038.
  2. Figus, F.A.; Piga, M.; Azzolin, I.; McConnell, R.; Iagnocco, A. Rheumatoid arthritis: Extra-articular manifestations and comorbidities. Autoimmun. Rev. 2021, 20, 102776.
  3. Aletaha, D.; Neogi, T.; Silman, A.J.; Funovits, J.; Felson, D.T.; Bingham, C.O., 3rd; Birnbaum, N.S.; Burmester, G.R.; Bykerk, V.P.; Cohen, M.D.; et al. 2010 Rheumatoid arthritis classification criteria: An American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 2010, 62, 2569–2581.
  4. Kay, J.; Upchurch, K.S. ACR/EULAR 2010 rheumatoid arthritis classification criteria. Rheumatology (Oxford) 2012, 51 (Suppl. S6), vi5–vi9.
  5. Pratt, A.G.; Isaacs, J.D. Seronegative rheumatoid arthritis: Pathogenetic and therapeutic aspects. Best Pract Res. Clin. Rheumatol. 2014, 28, 651–659.
  6. Lenti, M.V.; Rossi, C.M.; Melazzini, F.; Gastaldi, M.; Bugatti, S.; Rotondi, M.; Bianchi, P.I.; Gentile, A.; Chiovato, L.; Montecucco, C.; et al. Seronegative autoimmune diseases: A challenging diagnosis. Autoimmun. Rev. 2022, 21, 103143.
  7. Sokolova, M.V.; Schett, G.; Steffen, U. Autoantibodies in Rheumatoid Arthritis: Historical Background and Novel Findings. Clin. Rev. Allergy Immunol. 2022, 63, 138–151.
  8. Kolarz, B.; Podgorska, D.; Podgorski, R. Insights of rheumatoid arthritis biomarkers. Biomarkers 2021, 26, 185–195.
  9. Derksen, V.; Huizinga, T.W.J.; van der Woude, D. The role of autoantibodies in the pathophysiology of rheumatoid arthritis. Semin. Immunopathol. 2017, 39, 437–446.
  10. Curtis, J.R.; Jain, A.; Askling, J.; Bridges, S.L., Jr.; Carmona, L.; Dixon, W.; Finckh, A.; Hyrich, K.; Greenberg, J.D.; Kremer, J.; et al. A comparison of patient characteristics and outcomes in selected European and U.S. rheumatoid arthritis registries. Semin. Arthritis Rheum. 2010, 40, 2–14 e11.
  11. Courvoisier, D.S.; Chatzidionysiou, K.; Mongin, D.; Lauper, K.; Mariette, X.; Morel, J.; Gottenberg, J.E.; Bergstra, S.A.; Suarez, M.P.; Codreanu, C.; et al. The impact of seropositivity on the effectiveness of biologic anti-rheumatic agents: Results from a collaboration of 16 registries. Rheumatology 2021, 60, 820–828.
  12. Myasoedova, E.; Davis, J.; Matteson, E.L.; Crowson, C.S. Is the epidemiology of rheumatoid arthritis changing? Results from a population-based incidence study, 1985-2014. Ann. Rheum. Dis. 2020, 79, 440–444.
  13. Matthijssen, X.M.E.; Huizinga, T.W.J.; van der Helm-van Mil, A.H.M. Increasing incidence of autoantibody-negative RA is replicated and is partly explained by an aging population. Ann. Rheum. Dis. 2022, 81, e69.
  14. Matthijssen, X.M.E.; Niemantsverdriet, E.; Huizinga, T.W.J.; van der Helm-van Mil, A.H.M. Enhanced treatment strategies and distinct disease outcomes among autoantibody-positive and -negative rheumatoid arthritis patients over 25 years: A longitudinal cohort study in the Netherlands. PLoS Med. 2020, 17, e1003296.
  15. Takanashi, S.; Takeuchi, T.; Kaneko, Y. Effects of Aging on Rheumatoid Factor and Anticyclic Citrullinated Peptide Antibody Positivity in Patients With Rheumatoid Arthritis. J. Rheumatol. 2023, 50, 330–334.
  16. Turkcapar, N.; Demir, O.; Atli, T.; Kopuk, M.; Turgay, M.; Kinikli, G.; Duman, M. Late onset rheumatoid arthritis: Clinical and laboratory comparisons with younger onset patients. Arch. Gerontol. Geriatr. 2006, 42, 225–231.
  17. van Delft, M.A.M.; Huizinga, T.W.J. An overview of autoantibodies in rheumatoid arthritis. J. Autoimmun. 2020, 110, 102392.
  18. Regueiro, C.; Rodriguez-Rodriguez, L.; Lopez-Mejias, R.; Nuno, L.; Triguero-Martinez, A.; Perez-Pampin, E.; Corrales, A.; Villalba, A.; Lopez-Golan, Y.; Abasolo, L.; et al. A predominant involvement of the triple seropositive patients and others with rheumatoid factor in the association of smoking with rheumatoid arthritis. Sci. Rep. 2020, 10, 3355.
  19. Bergot, A.S.; Giri, R.; Thomas, R. The microbiome and rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2019, 33, 101497.
  20. Morotti, A.; Sollaku, I.; Franceschini, F.; Cavazzana, I.; Fredi, M.; Sala, E.; De Palma, G. Systematic Review and Meta-analysis on the Association of Occupational Exposure to Free Crystalline Silica and Rheumatoid Arthritis. Clin. Allergy Immunol. 2022, 62, 333–345.
  21. De Stefano, L.; D’Onofrio, B.; Manzo, A.; Montecucco, C.; Bugatti, S. The Genetic, Environmental, and Immunopathological Complexity of Autoantibody-Negative Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 12386.
  22. Padyukov, L. Genetics of rheumatoid arthritis. Semin. Immunopathol. 2022, 44, 47–62.
  23. Saevarsdottir, S.; Stefansdottir, L.; Sulem, P.; Thorleifsson, G.; Ferkingstad, E.; Rutsdottir, G.; Glintborg, B.; Westerlind, H.; Grondal, G.; Loft, I.C.; et al. Multiomics analysis of rheumatoid arthritis yields sequence variants that have large effects on risk of the seropositive subset. Ann. Rheum. Dis. 2022, 81, 1085–1095.
  24. Yuan, S.; Li, X.; Lin, A.; Larsson, S.C. Interleukins and rheumatoid arthritis: Bi-directional Mendelian randomization investigation. Semin. Arthritis Rheum. 2022, 53, 151958.
  25. Eyre, S.; Bowes, J.; Diogo, D.; Lee, A.; Barton, A.; Martin, P.; Zhernakova, A.; Stahl, E.; Viatte, S.; McAllister, K.; et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat. Genet. 2012, 44, 1336–1340.
  26. Bossini-Castillo, L.; de Kovel, C.; Kallberg, H.; van’t Slot, R.; Italiaander, A.; Coenen, M.; Tak, P.P.; Posthumus, M.D.; Wijmenga, C.; Huizinga, T.; et al. A genome-wide association study of rheumatoid arthritis without antibodies against citrullinated peptides. Ann. Rheum. Dis. 2015, 74, e15.
  27. He, X.H.; Xiao, Y.T.; Chen, W.Y.; Wang, M.J.; Wu, X.D.; Mei, L.Y.; Gao, K.X.; Huang, Q.C.; Huang, R.Y.; Chen, X.M. In silico analysis of serum miRNA profiles in seronegative and seropositive rheumatoid arthritis patients by small RNA sequencing. PeerJ 2023, 11, e15690.
  28. Alivernini, S.; Tolusso, B.; Petricca, L.; Bui, L.; Di Mario, C.; Gigante, M.R.; Di Sante, G.; Benvenuto, R.; Fedele, A.L.; Federico, F.; et al. Synovial Predictors of Differentiation to Definite Arthritis in Patients With Seronegative Undifferentiated Peripheral Inflammatory Arthritis: microRNA Signature, Histological, and Ultrasound Features. Front. Med. 2018, 5, 186.
  29. Ding, B.; Padyukov, L.; Lundstrom, E.; Seielstad, M.; Plenge, R.M.; Oksenberg, J.R.; Gregersen, P.K.; Alfredsson, L.; Klareskog, L. Different patterns of associations with anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis in the extended major histocompatibility complex region. Arthritis Rheum. 2009, 60, 30–38.
  30. Chang, C.; Xu, L.; Zhang, R.; Jin, Y.; Jiang, P.; Wei, K.; Xu, L.; Shi, Y.; Zhao, J.; Xiong, M.; et al. MicroRNA-Mediated Epigenetic Regulation of Rheumatoid Arthritis Susceptibility and Pathogenesis. Front. Immunol. 2022, 13, 838884.
  31. Alivernini, S.; Bruno, D.; Tolusso, B.; Bui, L.; Petricca, L.; Gigante, M.R.; Birra, D.; Fedele, A.L.; Peluso, G.; Federico, F.; et al. Differential synovial tissue biomarkers among psoriatic arthritis and rheumatoid factor/anti-citrulline antibody-negative rheumatoid arthritis. Arthritis Res. Ther. 2019, 21, 116.
  32. Argyriou, A.; Wadsworth, M.H., 2nd; Lendvai, A.; Christensen, S.M.; Hensvold, A.H.; Gerstner, C.; van Vollenhoven, A.; Kravarik, K.; Winkler, A.; Malmstrom, V.; et al. Single cell sequencing identifies clonally expanded synovial CD4(+) T(PH) cells expressing GPR56 in rheumatoid arthritis. Nat. Commun. 2022, 13, 4046.
  33. Angelopoulou, F.; Bogdanos, D.; Dimitroulas, T.; Sakkas, L.; Daoussis, D. Immune checkpoint inhibitor-induced musculoskeletal manifestations. Rheumatol. Int. 2021, 41, 33–42.
  34. Bridgewood, C.; Wittmann, M.; Macleod, T.; Watad, A.; Newton, D.; Bhan, K.; Amital, H.; Damiani, G.; Giryes, S.; Bragazzi, N.L.; et al. T Helper 2 IL-4/IL-13 Dual Blockade with Dupilumab Is Linked to Some Emergent T Helper 17—Type Diseases, Including Seronegative Arthritis and Enthesitis/Enthesopathy, but Not to Humoral Autoimmune Diseases. J. Investig. Dermatol. 2022, 142, 2660–2667.
  35. Yarwood, A.; Huizinga, T.W.; Worthington, J. The genetics of rheumatoid arthritis: Risk and protection in different stages of the evolution of RA. Rheumatology 2016, 55, 199–209.
  36. Bardin, T.; Legrand, L.; Naveau, B.; Marcelli-Barge, A.; Debeyre, N.; Lathrop, G.M.; Poirier, J.C.; Schmid, M.; Ryckewaert, A.; Dryll, A. HLA antigens and seronegative rheumatoid arthritis. Ann. Rheum. Dis. 1985, 44, 50–53.
  37. Kaarela, K.; Kautiainen, H. Continuous progression of radiological destruction in seropositive rheumatoid arthritis. J. Rheumatol. 1997, 24, 1285–1287.
  38. Choi, S.; Lee, K.H. Clinical management of seronegative and seropositive rheumatoid arthritis: A comparative study. PLoS ONE 2018, 13, e0195550.
  39. Martin-Sanchez, F.; Diamond, C.; Zeitler, M.; Gomez, A.I.; Baroja-Mazo, A.; Bagnall, J.; Spiller, D.; White, M.; Daniels, M.J.; Mortellaro, A.; et al. Inflammasome-dependent IL-1beta release depends upon membrane permeabilisation. Cell Death Differ. 2016, 23, 1219–1231.
  40. Bird, P.; Littlejohn, G.; Butcher, B.; Smith, T.; O’Sullivan, C.; Witcombe, D.; Griffiths, H. Real-world evaluation of effectiveness, persistence, and usage patterns of monotherapy and combination therapy tofacitinib in treatment of rheumatoid arthritis in Australia. Clin. Rheumatol. 2022, 41, 53–62.
  41. Sugawara, M.; Fujieda, Y.; Noguchi, A.; Tanimura, S.; Shimizu, Y.; Nakagawa, I.; Yoshimura, M.; Abe, N.; Kono, M.; Kato, M.; et al. Prediction of the intolerance or non-responder to Janus kinase inhibitors in patients with rheumatoid arthritis: A preliminary retrospective study with integrative cluster analysis. Clin. Exp. Rheumatol. 2022, 40, 1674–1680.
  42. Jung, J.Y.; Lee, E.; Kim, J.W.; Suh, C.H.; Kim, H.A. Efficacy and drug retention of tofacitinib in rheumatoid arthritis: From the nationwide Korean College of Rheumatology Biologics registry. Clin. Exp. Rheumatol. 2023, 41, 1034–1041.
  43. Dalbeth, N.; Gosling, A.L.; Gaffo, A.; Abhishek, A. Gout. Lancet 2021, 397, 1843–1855.
  44. Mauro, D.; Thomas, R.; Guggino, G.; Lories, R.; Brown, M.A.; Ciccia, F. Ankylosing spondylitis: An autoimmune or autoinflammatory disease? Nat. Rev. Rheumatol. 2021, 17, 387–404.
More
Information
Subjects: Rheumatology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 199
Revisions: 2 times (View History)
Update Date: 15 Nov 2023
1000/1000
Video Production Service