Publicado jul 26, 2018



PLUMX
Almetrics
 
Dimensions
 

Google Scholar
 
Search GoogleScholar


Julián Esteban Barahona Correa

Manuel Antonio Franco Cortés

Juana Ángel Uribe

Luz Stella Rodríguez Camacho

##plugins.themes.bootstrap3.article.details##

Resumen

La coexistencia de más de una enfermedad autoinmune (EAI) en un paciente se conoce como poliautoinmunidad (PAI) y se observa en el 35% de los pacientes con EAI. La eliminación de linfocitos B usando rituximab (RTX) controla la actividad de diferentes EAI. En el lupus eritematoso sistémico (LES) y en PAI no es clara la producción de citocinas por los linfocitos B. Métodos: Estudio exploratorio. Se obtuvo plasma de 11 pacientes con artritis reumatoide (AR) y poliautoinmunidad asociada a LES (PAILES) antes y después de rituximab (i. e., 6 meses). Como controles se utilizaron ocho individuos sanos. Las citocinas se midieron por ELISA (IFN-α, TGF-β1) o Cytometric Bead Array (TNF-α, IL-1β, IL-6, IL-8, IL-10, IL-12p70). Resultados: Previo a RTX, IL-6 se encontró elevada únicamente en AR, mientras que IL-8 lo estuvo en AR y en PAILES, comparados con controles. Después de RTX se encontró una disminución significativa de IL-6 en AR y de IL-8 en PAILES. Las concentraciones de otras citocinas medidas fueron similares (IFN-α, TGF-β1) o se encontraron por debajo de límite de detección (TNF-α, IL-1β, IL-10, IL-12p70), tanto en pacientes como en controles. Conclusión: Los datos resaltan la importancia de la secreción de citocinas por los linfocitos B y sugieren un rol diferencial en cada patología. El incremento de IL-8 previo a RTX en ambos grupos y la reducción después de la terapia en PAILES respaldan el potencial de la IL-8 como objetivo terapéutico. La heterogeneidad de la población de pacientes con PAI reafirma la importancia de la selección de subgrupos específicos en estudios futuros.

Keywords

citocinas; interleucina-8; artritis reumatoide; lupus eritematoso sistémico; rituximabcytokines; interleukin-8; rheumatoid arthritis; systemic lupus erythematosus; rituximab

References
1. Hayter SM, Cook MC. Updated assessment of the prevalence, spectrum and case definition of autoimmune disease. Autoimmun Rev. 2012;11(10):754–65.

2. Rojas-Villarraga A, Amaya-Amaya J, Rodriguez-Rodriguez A, Mantilla RD, Anaya J-M. Introducing polyautoimmunity: Secondary autoimmune diseases no longer exist. Autoimmune Dis. 2012;(1):254319.

3. Lino AC, Dörner T, Bar-Or A, Fillatreau S. Cytokine-producing B cells: a translational view on their roles in human and mouse autoimmune diseases. Immunol Rev. 2016;269(1):130–44.

4. Shen P, Fillatreau S. Antibody-independent functions of B cells: A focus on cytokines. Nat Rev Immunol. 2015;15(7):441–51.

5. Bao Y, Cao X. The immune potential and immunopathology of cytokine-producing B cell subsets: A comprehensive review. J Autoimmun. 2014;55(1):10–23.

6. Ospina FE, Betancur JF, Suso JP, Muñoz-Buitron E, Cañas CA, Tobón GJ. Papel de la citocina BAFF en las enfermedades autoinmunes: Rol fisiopatológico y estrategias terapeúticas. Rev Colomb Reumatol. 2016;23(3):177–94.

7. Barr TA, Shen P, Brown S, Lampropoulou V, Roch T, Lawrie S, et al. B cell depletion therapy ameliorates autoimmune disease through ablation of IL-6–producing B cells. J Exp Med. 2012;209(5):1001–10.

8. Ali MF, Driscoll CB, Walters PR, Limper AH, Carmona EM. Glucan-Activated Human B Lymphocytes Participate in Innate Immune Responses by Releasing Proinflammatory Cytokines and Stimulating Neutrophil Chemotaxis. J Immunol. 2015;195(11):5318–26.

9. Tay C, Liu Y-H, Hosseini H, Kanellakis P, Cao A, Peter K, et al. B-cell-specific depletion of tumour necrosis factor alpha inhibits atherosclerosis development and plaque vulnerability to rupture by reducing cell death and inflammation. Cardiovasc Res. 2016;111(4):385–97.

10. Opata MM, Ye Z, Hollifield M, Garvy BA. B cell production of tumor necrosis factor in response to Pneumocystis murina infection in mice. Infect Immun. 2013;81(11):4252–60.

11. Airoldi I, Guglielmino R, Carra G, Corcione A, Gerosa F, Taborelli G, et al. The interleukin-12 and interleukin-12 receptor system in normal and transformed human B lymphocytes. Haematologica. 2002;87(4):434–42.

12. Rincón-Arévalo H, Sánchez-Parra CC, Castaño D, Yassin L, Vásquez G. Regulatory B cells and mechanisms. Int Rev Immunol. 2016;35(2):156–76.

13. Benjamin D, Dower SK. Human B cells express two types of interleukin-1 receptors. Blood. 1990;75(10):2017–23.

14. Pike BL, Nossal GJ. Interleukin 1 can act as a B-cell growth and differentiation factor. Proc Natl Acad Sci U S A. 1985;82(23):8153–7.

15. Rieckmann P, Tuscano JM, Kehrl JH. Tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) in B-lymphocyte function. Methods. 1997;11(1):128–32.

16. Rosser EC, Oleinika K, Tonon S, Doyle R, Bosma A, Carter NA, et al. Regulatory B cells are induced by gut microbiota-driven interleukin-1β and interleukin-6 production. Nat Med. 2014;20(11):1334–9.

17. Kiefer K, Oropallo MA, Cancro MP, Marshak-Rothstein A. Role of type I interferons in the activation of autoreactive B cells. Immunol Cell Biol. 2012;90(5):498–504.

18. Singh N, Kumar B, Aluri V, Lenert P. Interfering with baffled B cells at the lupus tollway: Promises, successes, and failed expectations. J Allergy Clin Immunol. 2016;137(5):1325–33.

19. Berggren O, Hagberg N, Weber G, Alm GV, Rönnblom L, Eloranta M-L. B lymphocytes enhance interferon-α production by plasmacytoid dendritic cells. Arthritis Rheum. 2012;64(10):3409–19.

20. Tak PP, Thurlings RM, Rossier C, Nestorov I, Dimic A, Mircetic V, et al. Atacicept in patients with rheumatoid arthritis: Results of a multicenter, phase Ib, double-blind, placebo-controlled, dose-escalating, single- and repeated-dose study. Arthritis Rheum. 2008;58(1):61–72.

21. Van Vollenhoven RF, Kinnman N, Vincent E, Wax S, Bathon J. Atacicept in patients with rheumatoid arthritis and an inadequate response to methotrexate: Results of a phase II, randomized, placebo-controlled trial. Arthritis Rheum. 2011;63(7):1782–92.

22. Lal P, Su Z, Holweg CTJ, Silverman GJ, Schwartzman S, Kelman A, et al. Inflammation and autoantibody markers identify rheumatoid arthritis patients with enhanced clinical benefit following rituximab treatment. Arthritis Rheum. 2011;63(12):3681–91.

23. Emery P, Fleischmann R, Filipowicz-Sosnowska A, Schechtman J, Szczepanski L, Kavanaugh A, et al. The efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: Results of a phase IIB randomized, double-blind, placebo-controlled, dose-ranging trial. Arthritis Rheum. 2006;54(5):1390–400.

24. Cohen SB, Emery P, Greenwald MW, Dougados M, Furie RA, Genovese MC, et al. Rituximab for rheumatoid arthritis refractory to anti–tumor necrosis factor therapy: Results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks. Arthritis Rheum. 2006;54(9):2793–806.

25. Emery P, Deodhar A, Rigby WF, Isaacs JD, Combe B, Racewicz AJ, et al. Efficacy and safety of different doses and retreatment of rituximab: a randomised, placebo-controlled trial in patients who are biological naive with active rheumatoid arthritis and an inadequate response to methotrexate. Ann Rheum Dis. 2010;69(9):1629–35.

26. Roll P, Dörner T, Tony H-P. Anti-CD20 therapy in patients with rheumatoid arthritis: Predictors of response and B cell subset regeneration after repeated treatment. Arthritis Rheum. 2008;58(6):1566–75.

27. Cambridge G, Perry HC, Nogueira L, Serre G, Parsons HM, De La Torre I, et al. The effect of B-cell depletion therapy on serological evidence of B-cell and plasmablast activation in patients with rheumatoid arthritis over multiple cycles of rituximab treatment. J Autoimmun. mayo de 2014;50:67–76.

28. Burmester GR, Feist E, Dörner T. Emerging cell and cytokine targets in rheumatoid arthritis. Nat Rev Rheumatol. 2014;10(2):77–88.

29. Rojas-Villarraga A, Toro C-E, Espinosa G, Rodríguez-Velosa Y, Duarte-Rey C, Mantilla RD, et al. Factors influencing polyautoimmunity in systemic lupus erythematosus. Autoimmun Rev. 2010;9(4):229–32.

30. Amador-Patarroyo MJ, Arbeláez JG, Mantilla RD, Rodríguez-Rodríguez A, Cárdenas-Roldán J, Pineda-Tamayo R, et al. Sjögren’s syndrome at the crossroad of polyautoimmunity. J Autoimmun. 2012;39(3):199–205.

31. Pacheco Y, Barahona-Correa J, Monsalve DM, Acosta-Ampudia Y, Rojas M, Rodríguez Y, et al. Cytokine and autoantibody clusters interaction in systemic lupus erythematosus. J Transl Med. 2017;15(1):239.

32. Chan OTM, Hannum LG, Haberman AM, Madaio MP, Shlomchik MJ. A Novel Mouse with B Cells but Lacking Serum Antibody Reveals an Antibody-independent Role for B Cells in Murine Lupus. J Exp Med. 1999;189(10):1639–48.

33. Navarra S V, Guzmán RM, Gallacher AE, Hall S, Levy RA, Jiménez RE, et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet.
2011;377(9767):721–31.

34. Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzová D, et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63(12):3918–30.

35. Merrill JT, Neuwelt CM, Wallace DJ, Shanahan JC, Latinis KM, Oates JC, et al. Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. 2010;62(1):222–33.

36. Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sánchez-Guerrero J, et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. 2012;64(4):1215–26.

37. Kroese FG, Abdulahad WH, Haacke E, Bos NA, Vissink A, Bootsma H. B-cell hyperactivity in primary Sjogren’s syndrome. Expert Rev Clin Immunol. 2014;10(4):483–99.

38. Meijer JM, Meiners PM, Vissink A, Spijkervet FKL, Abdulahad W, Kamminga N, et al. Effectiveness of rituximab treatment in primary sjögren’s syndrome: A randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2010;62(4):960–8.

39. Pollard RPE, Abdulahad WH, Bootsma H, Meiners PM, Spijkervet FKL, Huitema MG, et al. Predominantly proinflammatory cytokines decrease after B cell depletion therapy in patients with primary Sjogren’s syndrome. Ann Rheum Dis. 2013;72(12):2048–50.

40. Tani C, D’Aniello D, Delle Sedie A, Carli L, Cagnoni M, Possemato N, et al. Rhupus syndrome: assessment of its prevalence and its clinical and instrumental characteristics in a prospective cohort of 103 SLE patients. Autoimmun Rev. 2013;12(4):537–41.

41. Amaya-Amaya J, Molano-González N, Franco J-S, Rodríguez-Jiménez M, Rojas-Villarraga A, Anaya J-M. Anti-CCP antibodies as a marker of rhupus. Lupus. 2015;24(8):892–4.

42. Andrade-Ortega L, Irazoque-Palazuelos F, Muñoz-López S, Rosales-Don Pablo VM. Efficacy and tolerability of rituximab in patients with rhupus. Reumatol Clin. 2013;9(4):201–5.

43. Fabris M, Quartuccio L, Lombardi S, Saracco M, Atzeni F, Carletto A, et al. The CC homozygosis of the -174G>C IL-6 polymorphism predicts a lower efficacy of rituximab therapy in rheumatoid arthritis. Autoimmun Rev. marzo de 2012;11(5):315–20.

44. Das S, Vital EM, Horton S, Bryer D, El-Sherbiny Y, Rawstron AC, et al. Abatacept or tocilizumab after rituximab in rheumatoid arthritis?: An exploratory study suggests non-response to rituximab is associated with persistently high IL-6 and better clinical response to IL-6 blocking therapy. Ann Rheum Dis. mayo de 2014;73(5):909–12.

45. Herrera D, Rojas OL, Duarte-Rey C, Mantilla RD, Angel J, Franco MA. Simultaneous assessment of rotavirus-specific memory B cells and serological memory after B cell depletion therapy with rituximab. PLoS One. 2014;9(5):e97087.

46. Almandoz JP, Gharib H. Hypothyroidism: Etiology, Diagnosis, and Management. Med Clin North Am. 2012;96(2):203–21.

47. Singh JA, Furst DE, Bharat A, Curtis JR, Kavanaugh AF, Kremer JM, et al. 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken). 2012;64(5):625–39.

48. Bertsias G, Ioannidis JP a, Boletis J, Bombardieri S, Cervera R, Dostal C, et al. EULAR recommendations for the management of systemic lupus erythematosus. Report of a Task Force of the EULAR Standing Committee for International Clinical Studies Including Therapeutics. Ann Rheum Dis. 2008;67(2):195–205.

49. Parra M, Herrera D, Jácome MF, Mesa MC, Rodríguez L-S, Guzmán C, et al. Circulating rotavirus-specific T cells have a poor functional profile. Virology. 2014;468–470:340–50.

50. Krishnamurthy A, Joshua V, Haj Hensvold A, Jin T, Sun M, Vivar N, et al. Identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss. Ann Rheum Dis. 2016;75(4):721–9.

51. Kosek E, Altawil R, Kadetoff D, Finn A, Westman M, Le Maître E, et al. Evidence of different mediators of central inflammation in dysfunctional and inflammatory pain - Interleukin-8 in fibromyalgia and interleukin-1 β in rheumatoid arthritis. J Neuroimmunol. 2015;280:49–55.

52. Klimiuk PA, Sierakowski S, Domyslawska I, Chwiecko J. Serum chemokines in patients with rheumatoid arthritis treated with etanercept. Rheumatol Int. 2011;31(4):457–61.

53. Talaat RM, Mohamed SF, Bassyouni IH, Raouf A a. Th1/Th2/Th17/Treg cytokine imbalance in systemic lupus erythematosus (SLE) patients: Correlation with disease activity. Cytokine. 2015;72(2):146–53.

54. Schuh E, Berer K, Mulazzani M, Feil K, Meinl I, Lahm H, et al. Features of Human CD3+CD20+ T Cells. J Immunol. 2016;197(4):1111–7.

55. Wilk E, Witte T, Marquardt N, Horvath T, Kalippke K, Scholz K, et al. Depletion of functionally active CD20+ T cells by rituximab treatment. Arthritis Rheum. diciembre de 2009;60(12):3563–71.

56. Marino M, Bartoccioni E, Alboini PE, Evoli A. Rituximab in myasthenia gravis: A “to be or not to be” inhibitor of T cell function. Ann N Y Acad Sci. 2018;1413(1):41–8.

57. Kaneko S, Satoh T, Chiba J, Ju C, Inoue K, Kagawa J. Interleukin-6 and interleukin-8 levels in serum and synovial fluid of patients with osteoarthritis. Cytokines Cell Mol Ther. 2000;6(2):71–9.

58. Eilertsen G, Nikolaisen C, Becker-Merok A, Nossent J. Interleukin-6 promotes arthritis and joint deformation in patients with systemic lupus erythematosus. Lupus. 2011;20(6):607–13.

59. Willis R, Seif AM, McGwin G, Martinez-Martinez LA, González EB, Dang N, et al. Effect of hydroxychloroquine treatment on pro-inflammatory cytokines and disease activity in SLE patients: data from LUMINA (LXXV), a multiethnic US cohort. Lupus. 2012;21(8):830–5.

60. Hrycek E, Franek A, Błaszczak E, Dworak J, Hrycek A. Serum levels of selected chemokines in systemic lupus erythematosus patients. Rheumatol Int. 2013;33(9):2423–7.

61. Keren Z, Braun-Moscovici Y, Markovits D, Rozin A, Nahir M, Balbir-Gurman A, et al. Depletion of B lymphocytes in rheumatoid arthritis patients modifies IL-8-anti-IL-8 autoantibody network. Clin Immunol. octubre de 2009;133(1):108–16.

62. Fabre S, Guisset C, Tatem L, Dossat N, Dupuy AM, Cohen JD, et al. Protein biochip array technology to monitor rituximab in rheumatoid arthritis. Clin Exp Immunol. 2009;155(3):395–402.

63. Crow MK. Type I interferon in organ-targeted autoimmune and inflammatory diseases. Arthritis Res Ther. 2010;12 Suppl 1(Suppl 1):S5.

64. van Baarsen LGM, Bos WH, Rustenburg F, van der Pouw Kraan TCTM, Wolbink GJJ, Dijkmans BAC, et al. Gene expression profiling in autoantibody-positive patients with arthralgia predicts development of arthritis. Arthritis Rheum. 2010;62(3):694–704.

65. Emamian ES, Leon JM, Lessard CJ, Grandits M, Baechler EC, Gaffney PM, et al. Peripheral blood gene expression profiling in Sjögren’s syndrome. Genes Immun. 2009;10(4):285–96.

66. Niewold TB. Connective tissue diseases: Targeting type I interferon in systemic lupus erythematosus. Nat Rev Rheumatol. 2016;12(7):377–8.

67. Yamanishi Y, Boyle DL, Clark M, Maki RA, Tortorella MD, Arner EC, et al. Expression and Regulation of Aggrecanase in Arthritis: The Role of TGF-. J Immunol. 2002;168(3):1405–12.

68. Sheng J, Chen W, Zhu H-J. The immune suppressive function of transforming growth factor- β (TGF- β ) in human diseases. Growth Factors. 2015;33(2):92–101.

69. Manolova I, Gerenova J, Ivanova M. Serum levels of transforming growth factor-β1 (TGF-β1) in patients with systemic lupus erythematosus and Hashimoto’s thyroiditis. Eur Cytokine Netw. 2013;24(1):69–74.

70. Mieliauskaite D, Venalis P, Dumalakiene I, Venalis A, Distler J. Relationship between serum levels of TGF-beta1 and clinical parameters in patients with rheumatoid arthritis and Sjögren’s syndrome secondary to rheumatoid arthritis. Autoimmunity. 2009;42(4):356–8.

71. de Jager W, Bourcier K, Rijkers GT, Prakken BJ, Seyfert-Margolis V. Prerequisites for cytokine measurements in clinical trials with multiplex immunoassays. BMC Immunol. 2009;10:52.

72. Zhou X, Fragala MS, McElhaney JE, Kuchel GA. Conceptual and methodological issues relevant to cytokine and inflammatory marker measurements in clinical research. Curr Opin Clin Nutr Metab Care. septiembre de 2010;13(5):541–7.

73. Anaya J-M. The diagnosis and clinical significance of polyautoimmunity. Autoimmun Rev. 2014;13(4–5):423–6.

74. Johar A, Sarmiento-Monroy JC, Rojas-Villarraga A, Silva-Lara MF, Patel HR, Mantilla RD, et al. Definition of mutations in polyautoimmunity. J Autoimmun. 2016;72:65–72.

75. Anaya J-M, Duarte-Rey C, Sarmiento-Monroy JC, Bardey D, Castiblanco J, Rojas-Villarraga A. Personalized medicine. Closing the gap between knowledge and clinical practice. Autoimmun Rev. 2016;8:833–42.

76. Ortega-Hernández OD, López-Guzmán S, Rojas-Villarraga A, Anaya J-M. Predicción de las enfermedades autoinmunes: Mito, realidad y riesgo. Rev Med. 2008;16(1):56–73.
Cómo citar
Barahona Correa, J. E., Franco Cortés, M. A., Ángel Uribe, J., & Rodríguez Camacho, L. S. (2018). Comparación de citocinas plasmáticas antes y después del tratamiento con rituximab en pacientes con artritis reumatoide y lupus eritematoso sistémico asociado a poliautoinmunidad. Universitas Medica, 59(3), 1–16. https://doi.org/10.11144/Javeriana.umed59-3.cyto
Sección
Artículos originales

Artículos más leídos del mismo autor/a

1 2 > >>