Проблемы иммунопатологии ревматоидного артрита: эволюция болезни

Бесплатный доступ

В обзоре рассмотрены новые данные, касающиеся иммунопатологии ревматоидного артрита (РА), с акцентом на раннюю стадию заболевания. Эволюция РА включает несколько последовательно (или дискретно) развивающихся стадий, завершающихся развитием симптомокомплекса, характерного для РА. Однако характер взаимодействия факторов внешней среды, генетической предрасположенности и иммунных механизмов, определяющих переход от стадии к стадии, варианты прогрессирования, характер и выраженность внесуставных (системных) проявлений и риск коморбидных заболеваний, до конца не ясен и в настоящее время является предметом интенсивных исследований. Среди патогенетических механизмов развития РА важное место занимает гиперпродукция аутоантител - ревматоидные факторы (РФ) и антитела к белкам, подвергнутым посттрансляционной модификации (ПТМ) - цитруллинированию, карбамилированию, ацетилированию и т. д. Развитие иммунного ответа против посттрансляционно модифицированных (в первую очередь цитруллинированных) белков является ключевым патогенетическим механизмом развития РА на всех стадиях заболевания. Новые данные, касающиеся роли АЦБ в развитии боли и костной резорбции в отсутствие воспаления, патогенетически обосновывают существование «преклинической» фазы заболевания, характеризующейся артралгиями и гиперпродукцией аутоантител. В заключение рассматриваются новые возможности профилактики РА в группах риска (АЦБ-позитивная клинически подозрительная артралгия) с использованием метотрексата, анти-В-клеточного препарата ритуксимаба, блокатора костимуляции Т-лимфоцитов абатацепта и др.

Еще

Болезнь стилла взрослых, генно-инженерные биологические препараты, глюкокортикоиды

Короткий адрес: https://sciup.org/14946007

IDR: 14946007   |   DOI: 10.14412/1995-4484-2017-277-294

Список литературы Проблемы иммунопатологии ревматоидного артрита: эволюция болезни

  • Catrina AI, Svensson CI, Malmström V, et al. Mechanisms leading from systemic autoimmunity to joint-specific disease in rheumatoid arthritis. Nat Rev Immunol. 2017;13(2):79-86 DOI: 10.1038/nrrheum.2016.200
  • Smolen JS, Aletaha D, McInnes IB. Rheumatoid arthritis. Lancet. 2016;388(10055):2023-38 DOI: 10.1016/S0140-6736(16)30173-8
  • Gerlag DM, Raza K, van Baarsen LGM, et al. EULAR recommendations for terminology and research in individuals at risk of rheumatoid arthritis: report from the Study Group for Risk Factors for Rheumatoid Arthritis. Ann Rheum Dis. 2012;71:638- DOI: 10.1136/annrheumdis-2011-200990
  • Mankia K, Emery P. Preclinical rheumatoid arthritis. Progress toward prevention. Arthritis Rheum. 2016;68:779-88 DOI: 10.1002/art.39603
  • Leandro M. B cells and rheumatoid factors in autoimmunity. Int J Rheum Dis. 2015;18:379-81 DOI: 10.1111/1756-185X.12690
  • Dö rner T, Jacobi AM, Lipsky PE. B cells in autoimmunity. Arthritis Res Ther. 2009;11:247 DOI: 10.1186/ar2780
  • Cantagrel A, Degboe Y. New autoantibodies associated with rheumatoid arthritis recognize posttranslationally modified self-protein. Joint Bone Spain. 2016;83:11-7 DOI: 10.1016/j.jbspin.2015.10.003
  • Aletaha D, Blüml S. Therapeutic implications of autoantibodies in rheumatoid arthritis. RMD Open. 2016 May 17;2(1):e000009 DOI: 10.1136/rmdopen-2014-000
  • Mastrangelo A, Colasanti T, Barbati C, et al. The role of post-translational protein modifications in rheumatological diseases: focus on rheumatoid arthritis. J Immunol Res. 2015;2015:Article ID 712490, 10 p DOI: 10.1155/2015/712490
  • Darrah E, Andrade F. Citrullination, and carbamylation, and malondialdehyde-acetaldehyde! Oh My! Entering the forest of autoantigen modifications in rheumatoid arthritis. Arthritis Rheum. 2015;67:604-8 DOI: 10.1002/art.38970
  • Trouw LA, Rispens T, Toes REM. Beyond citrullination: other post-translation protein modifications in rheumatoid arthritis. Nat Rev Rheumatol. 2017 Published online: 09 March 2017 DOI: 10.1038/nrrheum.2017.15
  • Anzilotti C, Pratesi F, Tommasi C, Migliorini P. Peptidylarginine deiminase 4 and citrullination in health and disease. Autoimmun Rev. 2010;9:158-60 DOI: 10.1016/j.autrev.2009.06.002
  • Willemze A, Trouw LA, Toes RE, Huizinga TWJ. The influence of ACPA status and characteristics on the course of RA. Nat Rev Rheumatol. 2012;8:114-52 DOI: 10.1038/nrrheum.2011;204
  • Klareskof L, Amara K, Malmstrom V. Adaptive immunity in rheumatoid arthritis: anticitrulline and other antibodies in the pathogenesis of rheumatoid arthritis. Curr Opin Rheumatol. 2014;26:72-9 DOI: 10.1097/B0R.0000000000000016
  • Nishimura K, Sugiyama D, Kogata Y, et al. Meta-analysis: diagnostic accuracy of anti-cyclic citrullinated peptide antibody and rheumatoid factor for rheumatoid arthritis. Ann Intern Med. 2007;146:797-808 DOI: 10.7326/0003-4819-146-11-20070605000008
  • Taylor P, Gartemann J, Hsieh J, Greeden J. A systemic review of serum biomarkers anti-cyclic citrullinated peptide and rheumatoid factor as test for rheumatoid arthritis. Autoimmune Dis. 2011;article ID 815038 DOI: 10.4061/2011/815038
  • Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 2010;62:2569-81 DOI: 10.1002/art.27584
  • Jilani AA, Mackworth-Young CG. The role of citrullinated protein antibodies in predicting erosive disease in rheumatoid arthritis: a systemic literature review and meta-analysis. Int J Rheumatol. 2015;Article ID 728610 DOI: 10.1155/2015/728610
  • Kuller LH, Mackey RH, Walitt BT, et al. Determinants of mortality among postmenopausal women in the Women's health initiative who report rheumatoid arthritis. Arthritis Rheum. 2014;66:497-507 DOI: 10.1002/art.38268
  • Humphreys J, van Nies JAB, Chupping J, et al. Rheumatoid factor and anti-citrullinated protein antibody positivity, but not level, are associated with increased mortality in patients with rheumatoid arthritis: results from two large independent cohort. Arthritis Res Ther. 2014;16:483 DOI: 10.1186/s13075-014-0483-3
  • Sakkas LI, Bogdanos DP, Katsiari C, Platsoucas CD. Anti-citrul-linated peptide as autoantigen in rheumatoid arthritis -relevance to treatment. Autoimmune Rev. 2014;13:1114-20 DOI: 10.1016/j.autrev.2014.08.012
  • Malmstrom V, Cartina AI, Klareskog L. The immunopathogenesis of seropositive rheumatoid arthritis: from triggering to targeting. Nat Rev Rheumatol. 2017;13(2):79-86 DOI: 10.1038/nrrheum.2016.200
  • Yarwood A, Huizinga TWJ, Worthington J. The genetics of rheumatoid arthritis: risk and protection in different stages of the evolution of RA. Rheumatology (Oxford). 2016;55(2):199-209 DOI: 10.1093/rheumatology/keu323
  • Ioan-Facsinay A, El-Bannoudi H, Scherer H.U, et al. Anti-cyclic citrullinated peptide antibodies are a collection of anti-citrullinated protein antibodies and contain overlapping and non-overlapping reactivities. Ann Rheum Dis. 2011;70:188-93 DOI: 10.1136/ard.2010.131102
  • Uysal H, Bockermann R, Nandakumar KS, et al. Structure and pathogenicity of antibodies specific for citrullinated collagen type II in experimental arthritis. J Exp Med. 2009;206:449-62 DOI: 10.1084/jem.20081862
  • Amara K, Steen J, Murray F, et al. Monoclonal IgG antibodies generated from joint-derived B cells of RA patients have a strong bias toward citrullinated autoantigen recognition. J Exp Med. 2013;210:445-55 DOI: 10.1084/jem.20121486
  • Aho K, Heliovaara M, Maatela J, et al. Rheumatoid factors antedating clinical rheumatoid arthritis. J Rheumatol. 1991;18:1282-4.
  • Aho K, Palosuo T, Heliovaara M, et al. Antifilaggrin antibodies within «normal» range predict rheumatoid arthritis in a linear fashion. J Rheumatol. 2000;27:2743-6.
  • Aho K, von Essen R, Kurki P, et al. Antikeratin antibody and antiperinuclear factor as markers for subclinical rheumatoid disease process. J Rheumatol. 1993;20:1278-81.
  • Del Puente A, Knowler WC, Pettitt DJ, Bennett PH. The incidence of rheumatoid arthritis is predicted by rheumatoid factor titer in a longitudinal population study. Arthritis Rheum. 1988;31:1239-44 DOI: 10.1002/art.1780311004
  • Silman AJ, Hennessy E, Ollier B. Incidence of rheumatoid arthritis in a genetically predisposed population. Br J Rheumatol. 1992;31:365-8 DOI: 10.1093/rheumatology/31.6.365
  • Majka DS, Deane KD, Parrish LA, et al. Duration of preclinical rheumatoid arthritis-related autoantibody positivity increases in subjects with older age at time of disease diagnosis. Ann Rheum Dis. 2008;67:801-7 DOI: 10.1136/ard.2007.076679
  • Chibnik LB, Mandl LA, Costenbader KH, et al. Comparison of threshold cutpoints and continuous measures of anti-cyclic citrul-linated peptide antibodies in predicting future rheumatoid arthritis. J Rheumatol. 2009;36:706-11 DOI: 10.3899/jrheum.080895
  • Jorgensen KT, Wiik A, Pedersen M, et al. 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;67:860-6 DOI: 10.1136/ard.2007.073825
  • Rantapaa-Dahlqvist S, Boman K, Tarkowski A, Hallmans G. Up regulation of monocyte chemoattractant protein-1 expression in anti-citrulline antibody and immunoglobulin M rheumatoid factor positive subjects precedes onset of inflammatory response and development of overt rheumatoid arthritis. Ann Rheum Dis. 2007;66:121-3 DOI: 10.1136/ard.2006.057331
  • Nielen MM, van Schaardenburg D, Reesink HW, et al. Simultaneous development of acute phase response and autoantibodies in preclinical rheumatoid arthritis. Ann Rheum Dis. 2006;65:535-7 DOI: 10.1136/ard.2005.040659
  • Rantapaa-Dahlqvist S, de Jong BAW, Berglin E, et al. Antibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritis. Arthritis Rheum. 2003;48:2741-9 DOI: 10.1002/art.11223
  • Nielen MMJ, van Schaardenburg D, Reesink HW, et al. Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors. Arthritis Rheum. 2004;50:380-6. 9 DOI: 10.1002/art.20018
  • Koppejan H, Trouw LA, Sokolove J, et al. Role of anti-carbamy-lated protein antibodies compared to anti-citrullinated protein antibodies in indigenous North Americans with rheumatoid arthritis, their first-degree relatives, and healthy controls. Arthritis Rheumatol (Hoboken, NJ). 2016;68:2090-8 DOI: 10.1002/art.39664
  • Brink M, Hansson M, Mathsson L, et al. Multiplex analyses of antibodies against citrullinated peptides in individuals prior to development of rheumatoid arthritis. Arthritis Rheum. 2013;65:899-910 DOI: 10.1002/art.37835
  • Nam JL, Hunt L, Hensor EM, Emery P. Enriching case selection for imminent RA: the use of anti-CCP antibodies in individuals with new non-specific musculoskeletal symptoms - a cohort study. Ann Rheum Dis. 2016 Aug;75(8):1452-6 DOI: 10.1136/annrheumdis-2015-207871
  • Rakieh C, Nam JL, Hunt L, et al. Predicting the development of clinical arthritis in anti-CCP positive individuals with non-specific musculoskeletal symptoms: a prospective observational cohort study. Ann Rheum Dis. 2015 Sep;74(9):1659-66 DOI: 10.1136/annrheumdis-2014-205227
  • Steenbergen HW, Mangnus L, Reijnierse M, et al. Clinical factors, anticitrullinated peptide antibodies and MRI-detected sub-clinical inflammation in relation to progression from clinically suspect arthralgia to arthritis.van Ann Rheum Dis. 2016 0ct;75(10):1824-30 DOI: 10.1136/annrheumdis-2015-208138
  • Van Zanten A, Arends S, Roozendaal C, et al. Presence of anticitrullinated protein antibodies in a large population-based cohort from the Netherlands. Ann Rheum Dis. 2017 DOI: 10.1136/annrheumdid-2016-209991
  • Hensvold AH, Frisell T, Magnusson PKE, et al. How well do ACPA discrimination and predict RA in the general population: a study based on 125090 population-representative Swedish twins. Ann Rherum Dis. 2017;76:119-25. dis-2015-208980 DOI: 10.1036/annrgeum
  • Ten Brinck RM, van Steenbergen HW, Verheul MK, et al. The prognostic value of different auto-antibodies for arthritis development in patients with clinically suspect arthralgia. Presented at: ACR/ARHP Annual Meeting; November 11-16, 2016; Washington D.C. Abstract #1035.
  • Terao C, Ohmura K, Ikari K, et al. Effects of smoking and shared epitope on the production of anti-citrullinated peptide antibody in a Japanese adult population. Arthritis Care Res. 2014;66:1818-27 DOI: 10.1002/acr.22385
  • Hensvold AH, Magnusson PK, Joshua V, et al. Environmental and genetic factors in the development of anticitrullinated protein antibodies (ACPAs) and ACPA-positive rheumatoid arthritis: an epidemiological investigation in twins. Ann Rheum Dis. 2015;74:375-80 DOI: 10.1136/annrheumdis-2013-203947
  • Rönnelid J, Wick MC, Lampa J, et al. Longitudinal analysis of citrullinated protein/peptide antibodies (anti-CP) during 5 year follow up in early rheumatoid arthritis: anti-CP status predicts worse disease activity and greater radiological progression. Ann Rheum Dis. 2005;64:1744-9 DOI: 10.1136/ard.2004.033571
  • Cornaby C, Gibbons L, Mayhew V, et al. B cell epitope spreading: mechanism and contribution to autoimmune diseases. Immunol Let. 2015;163:56-68 DOI: 10.1016/j.imlet.2014.11.001
  • Van de Stadt LA, de Koning MHMT, van de Stadt RJ, et al. Development of the anti-citrullinated protein antibody repertoire prior to the onset of rheumatoid arthritis. Arthritis Rheum. 2011;63:3226-33 DOI: 10.1002/art.30537
  • Van de Stadt LA, van der Horst AR, de Koning MHMT, et al. The extent of the anti-citrullinated protein antibody repertoire is associated with arthritis development in patients with seropositive arthralgia. Ann Rheum Dis. 2011;70:128-33 DOI: 10.1136/ard.2010.132662
  • Suwannalai P, van de Stadt LA, Radner H, et al. Avidity maturation of anti-citrullinated protein antibodies in rheumatoid arthritis. Arthritis Rheum. 2012;64:1323-8 DOI: 10.1002/art.33489
  • Van der Woude D, Rantapaa-Dahlqvist S, Ioan-Facsinay A, et al. Epitope spreading of the anti-citrullinated protein antibody response occurs before disease onset and is associated with the disease course of early arthritis. Ann Rheum Dis. 2010;69:1554-61 DOI: 10.1136/ard.2009.124537
  • Willemze A, Shi J, Mulder M, et al. The concentration of anti-citrullinated protein antibodies in serum and synovial fluid in relation to total immunoglobulin concentrations. Ann Rheum Dis. 2013;72:1059-63 DOI: 10.1136/annrheumdis-2012-202747
  • Ioan-Facsinay A, Willemze A, Robinson DB, et al. Marked differences in fine specificity and isotype usage of the anti-citrullinated protein antibody in health and disease. Arthritis Rheum. 2008;58:3000-8 DOI: 10.1002/art.23763
  • Kokkonen H, Mullazehi M, Berglin E, et al. Antibodies of IgG, IgA and IgM isotypes against cyclic citrullinated peptide precede the development of rheumatoid arthritis. Arthritis Res Ther. 2011;13:R13 DOI: 10.1186/ar3237
  • Van der Woude D, Syversen SW, van der Voort EI, et al. The ACPA isotype profile reflects long-term radiographic progression in rheumatoid arthritis. Ann Rheum Dis. 2010;69:1110-6 DOI: 10.1136/ard.2009.116384
  • Goulabchand R, Vincent T, Batteux F, et al. Impact of autoantibody glycosylation in autoimmune disease. Autoimmune Rev. 2014;13:742-50 DOI: 10.1016/j.autrev.2014.02.005
  • Scherer HU, Wang J, Toes RE, et al. Immunoglobulin 1 (IgG1) Fc-glycosylation profiling of anti-citrullinated peptide antibodies from human serum. Proteomics Clin Appl. 2009;3:106-15 DOI: 10.1002/prca.200800098
  • Scherer HU, van der Woude D, Ioan-Facsinay A, et al. Glycan profiling of anti-citrullinated protein antibodies isolated from human serum and synovial fluid. Arthritis Rheum. 2010;62:1620-9 DOI: 10.1002/art.27414
  • Rombouts Y, Ewing E, van de Stadt LA, et al. Anti-citrullinated protein antibodies acquire a pro-inflammatory Fc glycosylation phenotype prior to the onset of rheumatoid arthritis. Ann Rheum Dis. 2015;74:234-41 DOI: 10.1136/annrheumdis-2013-203565
  • Arnold JN, Wormald MR, Sim RB, et al. The impact of glycosylation on the biological function and structure of human immunoglobulins. Ann Rev Immunol. 2007;25:21-50. doi: 10.1146/annurev.immunol.25.022106.141702
  • Stadlmann J, Pabst M, Altmann F. Analytical and functional aspects of antibody sialylation. J Clin Immunol. 2010;30:15-9 DOI: 10.1007/s10875-010-9409-2
  • Koning F, Thomas R, Rossjohn J, Toes RE. Coeliac disease and rheumatoid arthritis: simmilar mechanisms, different antigens. Nat Rev Rheumatol. 2015;11:450-61 DOI: 10.1038/nrrheum.2015.59
  • Clavel C, Nogueira L, Laurent L, et al. Induction of macrophage secretion of tumor necrosis factor alpha through Fcgamma receptor IIa engagement by rheumatoid arthritis-specific autoantibodies to citrullinated proteins complexed with fibrinogen. Arthritis Rheum. 2008;58:678-88 DOI: 10.1002/art.23284
  • Sokolove J, Zhao X, Chandra PE, Robinson WH. Immune complexes containing citrullinated fibrinogen costimulate macrophages via Toll-like receptor 4 and Fcgamma receptor. Arthritis Rheum. 2011;63:53-62 DOI: 10.1002/art.30081
  • Sohn DH, Rhodes C, Onuma K, et al. Local joint inflammation and histone citrullination in a murine model of the transition from preclinical autoimmunity to inflammatory arthritis. Arthritis Rheum. 2015;67:2877-87 DOI: 10.1002/art.39283
  • Zhu W, Li X, Fang S, et al. Anti-citrullinated protein antibodies induce macrophage subset disequilibrium in RA patients. Inflammation. 2015;38:2067-75 DOI: 10.1007/s10753-015-0188-z
  • Sokolove J, Johnson DS, Lahey LJ, et al. Rheumatoid factor as a potentiator of anti-citrullinated protein antibody-mediated inflammation in rheumatoid arthritis. Arthritis Rheum. 2014;66:813-21 DOI: 10.1002/art.38307
  • Laurent L, Anquetil F, Clavel C, et al. IgM rheumatoid factor amplifies the inflammatory response of macrophages induced by the rheumatoid arthritis-specific immune complexes containing anticitrullinated protein antibodies. Ann Rheum Dis. 2015;74:1425-31 DOI: 10.1136/annrheumdis-2013-204543
  • Anquetil F, Clavel C, Offer G, et al. IgM and IgA rheumatoid factors purified from rheumatoid arthritis sera boost the Fc receptor-and complement-dependent effector functions of the disease-specific anticitrullinated protein autoantibodies. J Immunol. 2015;194:3664-74 DOI: 10.4049/jimmunol.1402334
  • Khandpur R, Carmona-Rivera C, Vivekanandan-Giri A, et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med. 2013;5:178ra40 DOI: 10.1126/scitranslmed.3005580
  • Sur Chowdhury C, Giaglis S, Walker UA, et al. Enhanced neutrophil extracellular trap generation in rheumatoid arthritis: analysis of underlying signal transduction pathways and potential diagnostic utility. Arthritis Res Ther. 2014;16:R122 DOI: 10.1186/ar4579
  • Trouw LA, Haisma EM, Levarht EW, et al. Anti-cyclic citrullinated peptide antibodies from rheumatoid arthritis patients activate complement via both the classical and alternative pathways. Arthritis Rheum. 2009;60:1923-31 DOI: 10.1002/art.24622
  • Harre U, Georgess D, Bang H, et al. Induction of osteoclastoge-nesis and bone loss by human autoantibodies against citrullinated vimentin. J Clin Invest. 2012;122:1791-802 DOI: 10.1172/JCI60975
  • Krishnamurthy A, Joshua V, Haj Hensvold A, et al. Identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss. Ann Rheum Dis. 2016;75:721-9 DOI: 10.1136/annrheumdis-2015-208093
  • Wigerblad G, Bas DB, Fernades-Cerqueira C, et al. Autoantibodies to citrullinated proteins induce joint pain independent of inflammation via a chemokine-dependent mechanism. Ann Rheum Dis. 2016;75:730-8 DOI: 10.1136/annrheumdis-2015-208094
  • Suurmond J, Rivellese F, Dorjee AL, et al. Toll-like receptor triggering augments activation of human mast cells by anticitrullinated protein antibodies. Ann Rheum Dis. 2015;74:1915-23 DOI: 10.1136/annrheumdis-2014-205562
  • Habets KL, Trouw LA, Levarht EW, et al. Anti-citrullinated protein antibodies contribute to platelet activation in rheumatoid arthritis. Arthritis Res Ther. 2015;17:209. 015-0665-7 DOI: 10.1186/s13075-
  • Kuhn KA, Kulik L, Tomooka B, et al. Antibodies against citrullinated proteins enhance tissue injury in experimental autoimmune arthritis. J Clin Invest. 2006;116:961-73 DOI: 10.1172/JCI25422
  • Ho PP, Lee LY, Zhao X, et al. Autoimmunity against fibrinogen mediates inflammatory arthritis in mice. J Immunol. 2010;184:379-90 DOI: 10.4049/jimmunol.0901639
  • Dwivedi N, Radic M. Citrullination of autoantigens implicated NETosis in the induction of autoimmunity. Ann Rheum Dis. 2014;73:483-91 DOI: 10.1136/annrheumdis-2013-203844
  • Harre U, Lang SC, Pfeifle R, et al. Glycosylation of immunoglobulin G determines osteoclast differentiation and bone lose. Nat Communocation. 2015 DOI: 10.1038/ncomms7651
  • Kleyer A, Finsel S, Rech J, et al. Bone loss before the clinical onset of rheumatoid arthritis in subjects with anticitrullinated protein antibodies. Ann Rheum Dis. 2014;73:854-60 DOI: 10.1136/annrhgeumdis-2012-202958
  • Bugatt S, Bogliolo L,Vitolo B, et al. Anti-citrullinated protein antibodies and high levels of rheumatoid factor are associated with systemic bone loss in patients with early untreated rheumatoid arthritis Arthritis Res Ther. 201618:226. 016-1116-9 DOI: 10.1186/s13075-
  • Titcombe PJ, Amara K, Barsness LO, et al. Citrullinated self anti-gen-specific blood B cells carry cross reactive immunoglobulins with effector potential. Ann Rheum Dis. 2016;75 Suppl 1:A28-A29 DOI: 10.1136/annrheumdis-2016-209124.68
  • Zhang ZJ, Cao DL, Zhang X, et al. Chemokine contribution to neuropathic Pain: respective induction of CXCL1 and CXCR2 in spinal cord astrocytes and neurons. Pain. 2013;154:2185-97 DOI: 10.1016/j.pain.2013.07.002
  • Lubberts E. The IL-23-IL-17 axis in inflammatory arthritis. Nat Rev Rheumatol. 2015;11(7):415-29 DOI: 10.1038/nrrheum.2015.53
  • Pfeifle R, Rothe T, Ipseiz N, et al. Regulation of autoantibody activity by the IL-23-TH17 axis determines the onset of autoimmune disease. Nat Immunol. 2017;18:104-13 DOI: 10.1038/ni.3579
  • Насонов ЕЛ. Новые возможности фармакотерапии иммуновоспалительных ревматических заболеваний: фокус на ингибиторы интерлейкина 17. Научно-практическая ревматология. 2017;55(1):68-86 DOI: 10.14412/1995-44842017-68-86
  • Fasching P, Stradner M, Graninger W, et al. Therapeutic potential of targeting the Th17/Treg axis in autoimmune disorders. Molecules. 2017;22(1):134 DOI: 10.3390/molecules22010134
  • Ajeganova S, van Steenbergen HW, Verheul MK, et al. The association between anti-carbamylated protein (anti-CarP) antibodies and radiographic progression in early rheumatoid arthritis: a study exploring replication and the added value to ACPA and rheumatoid factor. Ann Rheum Dis. 2017;76(1):112-8 DOI: 10.1136/annrheumdis-2015-208870
  • Shi J, van de Stadt LA, Levarth EWN, et al. Anti-carbamylated protein antibodies are present in arthralgia patients and predict the development of rheumatoid arthritis. Arthritis Rheum. 2013;65:911-5 DOI: 10.1002/art.37830
  • Harris ML, Darrah E, Lam GK, et al. Association of autoimmunity to peptidyl arginine deiminase type 4 with genotype and disease severity in rheumatoid arthritis. Arthritis Rheum. 2008;58(7):1958-67 DOI: 10.1002/art.23596
  • Kolfenbach JR, Deane KD, Derber LA, et al. Autoimmunity to peptidyl arginine deiminase type 4 precedes clinical onset of rheumatoid arthritis. Arthritis Rheum. 2010;62(9):2633-9 DOI: 10.1002/art.27570
  • Gan RW, Trouw LA, Shi J, et al. Anti-carbamylated protein anti-bodies are present prior to rheumatoid arthritis and are associated with its future diagnosis. J Rheumatol. 2015;42:572-9 DOI: 10.3899/jrheum.140767
  • Kokkonen H, Soderstrom I, Rocklov J, et al. Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis. Arthritis Rheum. 2010;62:383-91 DOI: 10.1002/art.27186
  • Deane KD, O'Donnell CI, Hueber W, et al. The number of elevated cytokines and chemokines in preclinical seropositive rheumatoid arthritis predicts time to diagnosis in an age-dependent manner. Arthritis Rheum. 2010;62(11):3161-72 DOI: 10.1002/art.27638
  • Hughes-Austin JM, Deane KD, Derber LA, et al. Multiple cytokines and chemokines are associated with rheumatoid arthritis-related autoimmunity in first-degree relatives without rheumatoid arthritis: Studies of the Aetiology of Rheumatoid Arthritis (SERA). Ann Rheum Dis. 2013;72(6):901-7 DOI: 10.1136/annrheumdis-2012-201505
  • Barra L, Summers K, Bell D, Cairns E. Serum Cytokine Profile of Unaffected First-degree Relatives of Patients with Rheumatoid Arthritis. J Rheumatol. 2014;4 (2):280-5 DOI: 10.3899/jrheum.130539
  • Chalan P, Bijzet J, van den Berg A, et al. Analysis of serum immune markers in seropositive and seronegative rheumatoid arthritis and in high-risk seropositive arthralgia patients. Sci Peport. 2016;6:26021 DOI: 10.1038/srep26021
  • Masi AT, Rehman AA, Elmore KB, Aldag JC. Serum acute phase protein and inflammatory cytokine network correlations: comparison of a pre-rheumatoid arthritis and non-rheumatoid arthritis community cohort. J Innate Immun. 2013;5:100-13 DOI: 10.1159/000345700
  • Maksymowych WP, Naides SJ, Bykerk V, et al. Serum 14-3-3eta is a novel marker that complements current serological measurements to enhance detection of patients with rheumatoid arthritis. J Rheumatol. 2014;41(11):2104-13 DOI: 10.3899/jrheum.131446
  • Van Heusden GP. 14-3-3 proteins: regulators of numerous eukaryotic proteins. IUBMBLife. 2005;57(9):623-9 DOI: 10.1080/15216540500252666
  • Hitchon CA, Smolik I, Meng X, et al. Serum 14-3-3eta are elevated in indigenous North Americans with rheumatoid arthritis and may predict imminent synovitis in their in at-risk first degree relatives. Arthritis Rheum. 2015;67(10).
  • Van Beers-Tas MH, Marotta A, Boers M, et al. A prospective cohort study of 14-3-3n in ACPA and/or RF-positive patients with arthralgia. Arthritis Res Ther. 2016;18:76 DOI: 10.1186/s13075-016-0975-4
  • Issa SF, Duer A, Ostergaard M, et al. Increased galectin-3 may serve as a serologic signature of pre-rheumatoid arthritis while markers of synovitis and cartilage do not differ between early undifferentiated arthritis subsets. Arthritis Res Ther. 2017;26;19(1):80 DOI: 10.1186/s13075-017-1282-4
  • Tan YC, Kongpachith S, Blum LK, et al. Barcode-enabled sequencing of plasmablast antibody repertoires in rheumatoid arthritis. Arthritis Rheum. 2014;66:2706-15 DOI: 10.1002/art.38754
  • Li S, Yu Y, Yue Y, et al. Autoantibodies from single circulating plasmablasts react with citrullinated antigens and Porphyromonas gingivalis in rheumatoid arthritis. Arthritis Rheum. 2016;68:614-26 DOI: 10.1002/art.39455
  • Corsiero E, Bombardieri M, Carlotti E, et al. Single cell cloning and recombinant monoclonal antibodies generation from RA synovial B cells reveal frequent targeting of citrullinated histones of NETs. Ann Rheum Dis. 2015;75:1866-75 DOI: 10.1136/annrheumdis-2015-208356
  • Kerkman PF, Kempers AC, van der Voort EI, et al. Synovial fluid mononuclear cells provide an environment for long-term survival of antibody-secreting cells and promote the spontaneous production of anticitrullinated protein antibodies. Ann Rheum Dis. 2016;75(12):2201-7. 2016 DOI: 10.1136/annrheumdis-2015-208554
  • De Hair MJH, van de Sande MGH, Ramwadhdoebe TH, et al. Features of the synovium of individuals at risk of developing rheumatoid arthritis: implications for understanding preclinical rheumatoid arthritis. Arthritis Rheum (Hoboken, NJ). 2014;66:513-22 DOI: 10.1002/art.38273
  • Van Baarsen LGM, de Hair MJH, Ramwadhdoebe TH, et al. The cellular composition of lymph nodes in the earliest phase of inflammatory arthritis. Ann Rheum Dis. 2013;72:1420-4 DOI: 10.1136/annrheumdis-2012-202990
  • Ramwadhdoebe TH, Hahnlein J, Maijer KI, et al. Lymph node biopsy analysis reveals an altered immunoregulatory balance already during the at-risk phase of autoantibody positive rheumatoid arthritis. Eur J Immunol. 2016;46:2812-21 DOI: 10.1002/eji.201646393
  • Ramwadhdoebe TH, Hahnlein J, van Kuijk BJ, et al. Human lymph-node CD8(+) T cells display an altered phenotype during systemic autoimmunity. Clin TranslImmunol. 2016;5:e67 DOI: 10.1038/cti.2016.8
  • Lübbers J, van Beers-Tas MH, Vosslamber S, et al. Changes in peripheral blood lymphocyte subsets during arthritis development in arthralgia patients. Arthritis Res Ther. 2016 Sep 14;18(1):205 DOI: 10.1186/s13075-016-1102-2
  • Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev Immunol. 2014;14:36-49 DOI: 10.1038/nri3581
  • Van Baarsen LG, Bos WH, Rustenburg F, et al. Gene expression profiling in autoantibody-positive patients with arthralgia predicts development of arthritis. Arthritis Rheum. 2010;62:694-704 DOI: 10.1002/art.27294
  • Lubbers J, Vosslamber S, van de Stadt LA, et al. B cell signature contributes to the prediction of RA development in patients with arthralgia. Ann Rheum Dis. 2015;74:1786-8 DOI: 10.1136/annrheumdis-2015-207324
  • Lubbers J, Brink M, van de Stadt LA, et al. The type I IFN signature as a biomarker of preclinical rheumatoid arthritis. Ann Rheum Dis. 2013;72:776-80 DOI: 10.1136/annrheumdis-2012-202753
  • Castaneda-Delgado JE, Bastian-Hernandez Y, Macias-Segura N, et al. Type I interferon gene response is increased in early and established rheumatoid arthritis and correlates with autoantibody production. Front Immunol. 2017;8:285 DOI: 10.3389/fimmu.2017.00285
  • Hunt L, Hensor EM, Nam J, et al. T cell subsets: an immunological biomarker to predict progression to clinical arthritis in ACPA-positive individuals. Ann Rheum Dis. 2016;75:1884-9 DOI: 10.1136/annrheumdis-2015-207991
  • Chalan P, Bijzet J, Kroesen B-J, et al. Altered natural killer cell subsets in seropositive arthralgia and early rheumatoid arthritis are associated with autoantibody status. J Rheumatol. 2016;43(6):1008-16 DOI: 10.3899/jrheum.150644
  • Rodriguez-Carrio J, Hahnlein JS, Ramwadhdoebe TH, et al. Brief report: altered innate lymphoid cell subsets in human lymph node biopsy specimens obtained during the at-risk and earliest phases of rheumatoid arthritis. Arthritis Rheum (Hoboken, NJ). 2017;69:70-6 DOI: 10.1002/art.39811
  • Shikhagaie MM, Germar K, Bal SM, et al. Innate lymphoid cells in autoimmunity: emerging regulators in rheumatic diseases. Nat Rev Rheumatol. 2017;13(3):164-73 DOI: 10.1038/nrrheum.2016.218
  • Makrygiannakis D, Hermansson M, Ulfgren A-K, et al. Smoking increases peptidylarginine deiminase 2 enzyme expression in human lungs and increases citrullination in BAL cells. Ann Rheum Dis. 2008;67:1488-92 DOI: 10.1136/ard.2007.075192
  • Stolt P, Bengtsson C, Nordmark B, et al. Quantification of the influence of cigarette smoking on rheumatoid arthritis: results from a population based case-control study, using incident cases. Ann Rheum Dis. 2003;62:835-41 DOI: 10.1136/ard.62.9.835
  • Lugli EB, Correia RESM, Fischer R, et al. Expression of citrulline and homocitrulline residues in the lungs of non-smokers and smokers: implications for autoimmunity in rheumatoid arthritis. Arthritis Res Ther. 2015;17:9 DOI: 10.1186/s13075-015-0520-x
  • Demoruelle MK, Weisman MH, Simonian PL, et al. Brief report: airways abnormalities and rheumatoid arthritis-related autoantibodies in subjects without arthritis: early injury or initiating site of autoimmunity? Arthritis Rheum. 2012;64:1756-61 DOI: 10.1002/art.34344
  • Willis VC, Demoruelle MK, Derber LA, et al. Sputum autoantibodies in patients with established rheumatoid arthritis and subjects at risk of future clinically apparent disease. Arthritis Rheum. 2013;65:2545-54 DOI: 10.1002/art.38066
  • Janssen KMJ, de Smit MJ, Brouwer E, et al. Rheumatoid arthritis-associated autoantibodies in non-rheumatoid arthritis patients with mucosal inflammation: a case-control study. Arthritis Res Ther. 2015;17:174 DOI: 10.1186/s13075-015-0690-6
  • Reynisdottir G, Olsen H, Joshua V, et al. Signs of immune activation and local inflammation are present in the bronchial tissue of patients with untreated early rheumatoid arthritis. Ann Rheum Dis. 2016;75:1722-7 DOI: 10.1136/annrheumdis-2015-208216
  • Ytterberg AJ, Joshua V, Reynisdottir G, et al. Shared immunological targets in the lungs and joints of patients with rheumatoid arthritis: identification and validation. Ann Rheum Dis. 2015;74:1772-7 DOI: 10.1136/annrheumdis-2013-204912
  • Kinslow JD, Blum LK, Deane KD, et al. IgA plasmablasts are elevated in subjects at risk for future rheumatoid arthritis. Arthritis Rheum. 2016;68:2372-83 DOI: 10.1002/art.39771
  • Roos K, Martinsson K, Ziegelasch M, et al. Circulating secretory IgA antibodies against cyclic citrullinated peptides in early rheumatoid arthritis associate with inflammatory activity and smoking. Arthritis Res Ther. 2016;23;18(1):119 DOI: 10.1186/s13075-016-1014-1
  • Bas S, Genevay S, Meyer O, Gabay C. Anti-cyclic citrullinated peptide antibodies, IgM and IgA rheumatoid factors in the diagnosis and prognosis of rheumatoid arthritis. Rheumatology. 2003;42:677-80 DOI: 10.1093/rheumatology/keg184
  • Svärd A, Skogh T, Alfredsson L, et al. Associations with smoking and shared epitope differ between IgA-and IgG-class antibodies to cyclic citrullinated peptides in early rheumatoid arthritis. Arthritis Rheum. 2015;67:2032-7 DOI: 10.1002/art.39170
  • Watkin LB, Jessen B, Wiszniewski W, et al. COPA mutations impair ER-Golgi transport and cause hereditary autoimmune-mediated lung disease and arthritis. Nat Genet. 2015;47:654-60 DOI: 10.1038/ng.3279
  • Nesse W, Dijkstra PU, Abbas F, et al. Increased prevalence of cardiovascular and autoimmune diseases in periodontitis patients: a cross-sectional study. J Periodontol. 2010;81:1622-8 DOI: 10.1902/jop.2010.100058
  • Dissick A, Redman RS, Jones M, et al. Association of periodontitis with rheumatoid arthritis: a pilot study. J Periodontol. 2010;81:223-30 DOI: 10.1902/jop.2009.090309
  • De Smit M, Westra J, Vissink A, et al. Periodontitis in established rheumatoid arthritis patients: a cross-sectional clinical, microbiological and serological study. Arthritis Res Ther. 2012;14:R222 DOI: 10.1186/ar4061
  • Nesse W, Westra J, van der Wal JE, et al. The periodontium of periodontitis patients contains citrullinated proteins which may play a role in ACPA (anti-citrullinated protein antibody) formation. J Clin Periodontol. 2012;39:599-607 DOI: 10.1111/j.1600-051X.2012.01885.x
  • Harvey GP, Fitzsimmons TR, Dhamarpatni AASSK, et al. Expression of peptidylarginine deiminase-2 and -4, citrullinated proteins and anti-citrullinated protein antibodies in human gingiva. J Periodontal Res. 2013;48:252-61 DOI: 10.1111/jre.12002
  • Wegner N, Wait R, Sroka A, et al. Peptidylarginine deiminase from Porphyromonas gingivalis citrullinates human fibrinogen and alpha-enolase: implications for autoimmunity in rheumatoid arthritis. Arthritis Rheum. 2010;62:2662-72 DOI: 10.1002/art.27552
  • Wegner N, Lundberg K, Kinloch A, et al. Autoimmunity to specific citrullinated proteins gives the first clues to the etiology of rheumatoid arthritis. Immunol Rev. 2010; 233:34-54 DOI: 10.1111/j.0105-2896.2009.00850.x
  • Mikuls TR, Thiele GM, Deane KD, et al. Porphyromonas gingivalis and disease-related autoantibodies in individuals at increased risk of rheumatoid arthritis. Arthritis Rheum. 2012;64:3522-30 DOI: 10.1002/art.34595
  • Bello-Gualtero JM, Lafaurie GI, Hoyos LX, et al. Periodontal disease in individuals with a genetic risk of developing arthritis and early rheumatoid arthritis: a cross-sectional study. J Periodontol. 2016;87:346-56 DOI: 10.1902/jop.2015.150455
  • Quirke A-M, Lugli EB, Wegner N, et al. Heightened immune response to autocitrullinated Porphyromonas gingivalis peptidy-larginine deiminase: a potential mechanism for breaching immunologic tolerance in rheumatoid arthritis. Ann Rheum Dis. 2014;73:263-9 DOI: 10.1136/annrheumdis-2012-202726
  • Fisher BA, Cartwright AJ, Quirke A-M, et al. Smoking, Porphyromonas gingivalis and the immune response to citrullinated autoantigens before the clinical onset of rheumatoid arthritis in a Southern European nested case-control study. BMC Musculoskelet Disord. 2015;16:331 DOI: 10.1186/s12891-015-0792-y
  • Eriksson K, Nise L, Kats A, et al. Prevalence of periodontitis in patients with established rheumatoid arthritis: a Swedish population based case-control study. PLoSONE. 2016;11:e0155956 DOI: 10.1371/journal.pone.0155956
  • Kharlamova N, Jiang X, Sherina N, et al. Antibodies to Porphyromonas gingivalis indicate interaction between oral infection, smoking, and risk genes in rheumatoid arthritis etiology. Arthritis Rheum. 2016;68:604-13 DOI: 10.1002/art.39491
  • Konig MF, Abusleme L, Reinholdt J, et al. Aggregatibacter actinomycetemcomitans-induced hypercitrullination links periodontal infection to autoimmunity in rheumatoid arthritis. Sci Transl Med. 2016;8(369):369ra176 DOI: 10.1126/scitranslmed.aaj1921
  • Ling S, Cline EN, Haug TS, et al. Citrullinated calreticulin potentiates rheumatoid arthritis shared epitope signaling. Arthritis Rheum. 2013;65(3):618-26 DOI: 10.1002/art.37814
  • Chen B, Sun L, Zhang X. Integration of microbiome and epigenome to decipher the pathogenesis of autoimmune diseases. J Autoimmun. 2017. pii: S0896-8411(17)30178-6 DOI: 10.1016/j.jaut.2017.03.009
  • Diamanti AP, Manuela Rosado M, Lagana B, D'Amelio R. Microbiota and chronic inflammatory arthritis: an interwoven link. J Transl Med. 2016;14(1):233 DOI: 10.1186/s12967-016-0989-3
  • Wu X, He B, Liu J, et al. Molecular insight into gut microbiota and rheumatoid arthritis. Int J Mol Sci. 2016;17(3):431 DOI: 10.3390/ijms17030431
  • Scher JU, Littman DR, Abramson SB. Microbiome in inflammatory arthritis and human rheumatic diseases. Arthritis Rheum. 2016;68(1):35-45 DOI: 10.1002/art.39259
  • Scher JU, Sczesnak A, Longman RS, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013;2:e01202 DOI: 10.7554/eLife.01202
  • Pianta A, Arvikar S, Strle K, et al. Evidence for immune relevance of Prevotella copri, a gut microbe, in patients with rheumatoid arthritis. Arthritis Rheum (Hoboken, NJ). 2016 DOI: 10.1002/art.40003
  • Zhang X, Zhang D, Jia H, et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med. 2015;21:895-905 DOI: 10.1038/nm.3914
  • MacGregor AJ, Snieder H, Rigby AS, et al. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum. 2000;43(1):30-7. doi: 10.1002/1529-0131(200001)43:13.0.co;2-b
  • Gregersen PK, Silver J, Winchester RJ. The shared epitope hypothesis. An approach to understanding the molecular genetics of susceptibility to rheumatoid arthritis. Arthritis Rheum. 1987;30:1205-13 DOI: 10.1002/art.1780301102
  • Huizinga TWJ, Amos CI, van der Helm-van Mil AHM, et al. Refining the complex rheumatoid arthritis phenotype based on specificity of the HLA-DRB1 shared epitope for antibodies to citrullinated proteins. Arthritis Rheum. 2005;52:3433-8 DOI: 10.1002/art.21385
  • Raychaudhuri S, Sandor C, Stahl EA, et al. Five amino acids in three HLA proteins explain most of the association between MHC and seropositive rheumatoid arthritis. Nat Genet. 2012;44:291-6 DOI: 10.1038/ng.1076
  • Scally SW, Petersen J, Law SC, et al. A molecular basis for the association of the HLA-DRB1 locus, citrullination, and rheumatoid arthritis. J Exp Med. 2013;210:2569-82 DOI: 10.1084/jem.20131241
  • De Almeida DE, Ling S, Holoshitz J. New insights into the functional role of the rheumatoid arthritis shared epitope. FEBS Lett. 2011;585(23):3619-26 DOI: 10.1016/j.febslet.2011.03.035
  • Bos WH, Ursum J, de Vries N, et al. The role of the shared epitope in arthralgia with anti-cyclic citrullinated peptide antibodies (anti-CCP), and its effect on anti-CCP levels. Ann Rheum Dis. 2008;67:1347-50 DOI: 10.1136/ard.2008.089953
  • Tracy A, Buckley CD, Raza K. Pre-symptomatic autoimmunity in rheumatoid arthritis: when does the disease start? Semin Immunopathol. 2017 Mar 23 DOI: 10.1007/s00281-017-0620-6
  • Snir 0, Rieck M, Gebe JA, et al. Identification and functional characterization of T cells reactive to citrullinated vimentin in HLA-DRB1*0401-positive humanized mice and rheumatoid arthritis patients. Arthritis Rheum. 2011;63:2873-83 DOI: 10.1002/art.30445
  • James EA, Rieck M, Pieper J, et al. Citrulline-specific Th1 cells are increased in rheumatoid arthritis and their frequency is influenced by disease duration and therapy. Arthritis Rheum. 2014;66:1712-22 DOI: 10.1002/art.38637
  • Padyukov L, Silva C, Stolt P, et al. A gene-environment interaction between smoking and shared epitope genes in HLA-DR provides a high risk of seropositive rheumatoid arthritis. Arthritis Rheum. 2004;50:3085-92 DOI: 10.1002/art.20553
  • Pedersen M, Jacobsen S, Garred P, et al. Strong combined geneenvironment effects in anti-cyclic citrullinated peptide-positive rheumatoid arthritis: a nationwide case-control study in Denmark. Arthritis Rheum. 2007;56:1446-53 DOI: 10.1002/art.22597
  • Fisher BA, Bang S-Y, Chowdhury M, et al. Smoking, the HLA-DRB1 shared epitope and ACPA fine-specificity in Koreans with rheumatoid arthritis: evidence for more than one pathogenic pathway linking smoking to disease. Ann Rheum Dis. 2014;73:741-7 DOI: 10.1136/annrheumdis-2012-202535
  • Lee H-S, Irigoyen P, Kern M, et al. Interaction between smoking, the shared epitope, and anti-cyclic citrullinated peptide: a mixed picture in three large North American rheumatoid arthritis cohorts. Arthritis Rheum. 2007;56:1745-53 DOI: 10.1002/art.22703
  • Van Heemst J, Hensvold AH, Jiang X, et al. Protective effect of HLA-DRB1*13 alleles during specific phases in the development of ACPA-positive RA. Ann Rheum Dis. 2016;75:1891-8 DOI: 10.1136/annrheumdis-2015-207802
  • Van Heemst J, Jansen DTSL, Polydorides S, et al. Crossreactivity to vinculin and microbes provides a molecular basis for HLA-based protection against rheumatoid arthritis. Nat Commun. 2015;6:6681 DOI: 10.1038/ncomms7681
  • Diogo D, Okada Y, Plenge RM. Genome-wide association studies to advance our understanding of critical cell types and pathways in rheumatoid arthritis: recent findings and challenges. Curr Opin Rheumatol. 2014;26(1):85-92 DOI: 10.1097/bor.0000000000000012
  • Salmond RJ, Brownlie RJ, Morrison VL, Zamoyska R. The tyrosine phosphatase PTPN22 discriminates weak self peptides from strong agonist TCR signals. Nat Immunol. 2014;15:875-83 DOI: 10.1038/ni.2958
  • Menard L, Saadoun D, Isnardi I, et al. The PTPN22 allele encoding an R620W variant interferes with the removal of developing autoreactive B cells in humans. J Clin Invest. 2011;121:3635-44 DOI: 10.1172/JCI45790
  • Chang HH, Liu GY, Dwivedi N, et al. A molecular signature of preclinical rheumatoid arthritis triggered by dysregulated PTPN22. JCIInsight. 2016;1(17):e90045 DOI: 10.1172/jci.insight.90045
  • Kallberg H, Padyukov L, Plenge RM, et al. Epidemiological Investigation of Rheumatoid Arthritis study group. Gene-gene and gene-environment interactions involving HLA-DRB1, PTPN22, and smoking in two subsets of rheumatoid arthritis. Am J Hum Genet. 2007;80:867-75 DOI: 10.1086/516736
  • Yang Z, Shen Y, Oishi H, et al. Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci Transl Med. 2016;8:331ra38 DOI: 10.1126/sci-translmed.aad7151
  • Di Giuseppe D, Crippa A, Orsini N, Wolk A. Fish consumption and risk of rheumatoid arthritis: a dose-response meta-analysis. Arthritis Res Ther. 2014;16:446 DOI: 10.1186/s13075-014-0446-8
  • Gan RW, Demoruelle MK, Deane KD, et al. Omega-3 fatty acids are associated with a lower prevalence of autoantibodies in shared epitope-positive subjects at risk for rheumatoid arthritis. Ann Rheum Dis. 2017;76:147-52 DOI: 10.1136/annrheumdis-2016-209154
  • Goldberg RJ, Katz J. A meta-analysis of the analgesic effects of omega-3 polyunsaturated fatty acid supplementation for inflammatory joint Pain. Pain. 2007;129:210-23 DOI: 10.1016/j.pain.2007.01.020
  • Mas E, Croft KD, Zahra P, et al. Resolvins D1, D2, and other mediators of self-limited resolution of inflammation in human blood following n-3 fatty acid supplementation. Clin Chem. 2012;58:1476-84 DOI: 10.1373/clinchem.2012.190199
  • Arnardottir HH, Dalli J, Norling LV, et al. Resolvin D3 is dysregulated in arthritis and reduces arthritic inflammation. J Immunol. 2016;197:2362-8 DOI: 10.4049/jimmunol.1502268
  • McAllen RM, Cook AD, Khiew HW, et al. The interface between cholinergic pathways and the immune system and its relevance to arthritis. Arthritis Res Ther. 2015;17:87 DOI: 10.1186/s13075-015-0597-2
  • Koopman FA, Tang MW, Vermeij J, et al. Autonomic dysfunction precedes development of rheumatoid arthritis: a prospective cohort study. EBioMedicine. 2016;6:231-7 DOI: 10.1016/j.ebiom.2016.02.029
  • Liu Z, Han B, Li P, et al. Activation of alpha7nAChR by nicotine reduced the Th17 response in CD4(+)T lymphocytes. Immunol Investig. 2014;43:667-74 DOI: 10.3109/08820139.2014.914532
  • Baez-Pagan CA, Delgado-Velez M, Lasalde-Dominicci JA. Activation of the macrophage alpha7 nicotinic acetylcholine receptor and control of inflammation. J NeuroImmune Pharmacol. 2015;10:468-76 DOI: 10.1007/s11481-015-9601-5
  • Koopman FA, Chavan SS, Miljko S, et al. Vagus nerve stimulation inhibits cytokine production and attenuates disease severity in rheumatoid arthritis. Proc Natl Acad Sci U S A. 2016;113:8284-9 DOI: 10.1073/pnas.1605635113
  • Van Steenbergen HW, Huizinga TW, van der Helm-van Mil AH. Review: the preclinical phase of rheumatoid arthritis: what is acknowledged and what needs to be assessed? Arthritis Rheum. 2013;65:2219-32 DOI: 10.1002/art.38013
  • Raza K, Gerlag DM. Preclinical inflammatory rheumatic diseases: an overview and relevant nomenclature. Rheum Dis Clin N Am. 2014;40:569-80 DOI: 10.1016/j.rdc.2014.07.001
  • Stack RJ, van Tuyl LH, Sloots M, et al. Symptom complexes in patients with seropositive arthralgia and in patients newly diagnosed with rheumatoid arthritis: a qualitative exploration of symptom development. Rheumatology (Oxford). 2014;53(9):1646- DOI: 10.1093/rheumatology/keu159
  • Bos WH, Wolbink GJ, Boers M, et al. Arthritis development in patients with arthralgia is strongly associated with anti-citrullinat-ed protein antibody status: a prospective cohort study. Ann Rheum Dis. 2010;69:490-4 DOI: 10.1136/ard.2008.105759
Еще
Статья научная