Skip to main content
Top
Published in: Clinical and Translational Medicine 1/2019

Open Access 01-12-2019 | Systemic Lupus Erythematosus | Research

Clinical features of Sjögren’s syndrome patients with autoantibodies against interferons

Authors: Peter D. Burbelo, Sarah Browne, Steve M. Holland, Michael J. Iadarola, Ilias Alevizos

Published in: Clinical and Translational Medicine | Issue 1/2019

Login to get access

Abstract

Background

Sjögren’s syndrome (SS) is an autoimmune disease characterized by immune attack on the salivary and lacrimal glands. Given the known cytokine activation and type I interferon gene expression signature found in SS, we hypothesized that anticytokine autoantibodies might be detectable by Luciferase immunoprecipitation systems in some SS patients and correlate with clinical symptoms.

Results

Luciferase immunoprecipitation systems was used to screen for serum anti-cytokine autoantibodies in 57 primary SS patients and 25 healthy volunteers. Autoantibodies were detected against GMCSF, interferon-γ, -α and, -ω in one, two, two and six patients with SS, respectively. None of the healthy volunteers showed anticytokine autoantibodies and none of the SS or control subjects showed autoantibodies against interferon-λ. One 51-year old female SS subject with the highest anti-interferon-α and -ω autoantibody levels had stable autoantibody levels over the course of a year. In vitro functional testing of serum autoantibodies from this subject demonstrated partially neutralizing activity for interferon-α signaling. Clinical information on this individual revealed a low focus score and high levels of unstimulated salivary flow, suggesting the possibility that interferon-α autoantibody neutralizing activity may have contributed to the milder sicca symptoms.

Conclusion

Overall, these findings demonstrate that a subset of SS patients (16%) harbor autoantibodies against GMCSF, interferon-γ, interferon-ω, and interferon-α. These data support the observation that high levels of interferon-α autoantibodies may attenuate disease symptoms in SS.
Literature
1.
go back to reference Goules AV, Tzioufas AG (2016) Primary Sjgren’s syndrome: clinical phenotypes, outcome and the development of biomarkers. Autoimmun Rev 15(7):695–703CrossRef Goules AV, Tzioufas AG (2016) Primary Sjgren’s syndrome: clinical phenotypes, outcome and the development of biomarkers. Autoimmun Rev 15(7):695–703CrossRef
2.
go back to reference Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T et al (2006) Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjogren’s syndrome. Proc Natl Acad Sci USA 103(8):2770–2775CrossRef Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T et al (2006) Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjogren’s syndrome. Proc Natl Acad Sci USA 103(8):2770–2775CrossRef
3.
go back to reference Hjelmervik TO, Petersen K, Jonassen I, Jonsson R, Bolstad AI (2005) Gene expression profiling of minor salivary glands clearly distinguishes primary Sjogren’s syndrome patients from healthy control subjects. Arthritis Rheum 52(5):1534–1544CrossRef Hjelmervik TO, Petersen K, Jonassen I, Jonsson R, Bolstad AI (2005) Gene expression profiling of minor salivary glands clearly distinguishes primary Sjogren’s syndrome patients from healthy control subjects. Arthritis Rheum 52(5):1534–1544CrossRef
4.
go back to reference Emamian ES, Leon JM, Lessard CJ, Grandits M, Baechler EC, Gaffney PM et al (2009) Peripheral blood gene expression profiling in Sjogren’s syndrome. Genes Immun 10(4):285–296CrossRef Emamian ES, Leon JM, Lessard CJ, Grandits M, Baechler EC, Gaffney PM et al (2009) Peripheral blood gene expression profiling in Sjogren’s syndrome. Genes Immun 10(4):285–296CrossRef
5.
go back to reference Wildenberg ME, van Helden-Meeuwsen CG, van de Merwe JP, Drexhage HA, Versnel MA (2008) Systemic increase in type I interferon activity in Sjogren’s syndrome: a putative role for plasmacytoid dendritic cells. Eur J Immunol 38(7):2024–2033CrossRef Wildenberg ME, van Helden-Meeuwsen CG, van de Merwe JP, Drexhage HA, Versnel MA (2008) Systemic increase in type I interferon activity in Sjogren’s syndrome: a putative role for plasmacytoid dendritic cells. Eur J Immunol 38(7):2024–2033CrossRef
6.
go back to reference Bave U, Nordmark G, Lovgren T, Ronnelid J, Cajander S, Eloranta ML et al (2005) Activation of the type I interferon system in primary Sjogren’s syndrome: a possible etiopathogenic mechanism. Arthritis Rheum 52(4):1185–1195CrossRef Bave U, Nordmark G, Lovgren T, Ronnelid J, Cajander S, Eloranta ML et al (2005) Activation of the type I interferon system in primary Sjogren’s syndrome: a possible etiopathogenic mechanism. Arthritis Rheum 52(4):1185–1195CrossRef
7.
go back to reference Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ et al (2003) Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci USA 100(5):2610–2615CrossRef Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ et al (2003) Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci USA 100(5):2610–2615CrossRef
8.
go back to reference Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J et al (2003) Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med 197(6):711–723CrossRef Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J et al (2003) Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med 197(6):711–723CrossRef
9.
go back to reference Greenberg SA, Pinkus JL, Pinkus GS, Burleson T, Sanoudou D, Tawil R et al (2005) Interferon-alpha/beta-mediated innate immune mechanisms in dermatomyositis. Ann Neurol 57(5):664–678CrossRef Greenberg SA, Pinkus JL, Pinkus GS, Burleson T, Sanoudou D, Tawil R et al (2005) Interferon-alpha/beta-mediated innate immune mechanisms in dermatomyositis. Ann Neurol 57(5):664–678CrossRef
10.
go back to reference Nestle FO, Conrad C, Tun-Kyi A, Homey B, Gombert M, Boyman O et al (2005) Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med 202(1):135–143CrossRef Nestle FO, Conrad C, Tun-Kyi A, Homey B, Gombert M, Boyman O et al (2005) Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med 202(1):135–143CrossRef
11.
go back to reference Bengtsson AA, Sturfelt G, Truedsson L, Blomberg J, Alm G, Vallin H et al (2000) Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies. Lupus. 9(9):664–671CrossRef Bengtsson AA, Sturfelt G, Truedsson L, Blomberg J, Alm G, Vallin H et al (2000) Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies. Lupus. 9(9):664–671CrossRef
12.
go back to reference Dall’era MC, Cardarelli PM, Preston BT, Witte A, Davis JC Jr (2005) Type I interferon correlates with serological and clinical manifestations of SLE. Ann Rheum Dis 64(12):1692–1697CrossRef Dall’era MC, Cardarelli PM, Preston BT, Witte A, Davis JC Jr (2005) Type I interferon correlates with serological and clinical manifestations of SLE. Ann Rheum Dis 64(12):1692–1697CrossRef
13.
go back to reference Hooks JJ, Moutsopoulos HM, Geis SA, Stahl NI, Decker JL, Notkins AL (1979) Immune interferon in the circulation of patients with autoimmune disease. New Engl J Med 301(1):5–8CrossRef Hooks JJ, Moutsopoulos HM, Geis SA, Stahl NI, Decker JL, Notkins AL (1979) Immune interferon in the circulation of patients with autoimmune disease. New Engl J Med 301(1):5–8CrossRef
14.
go back to reference Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK (2005) Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum 52(5):1491–1503CrossRef Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK (2005) Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum 52(5):1491–1503CrossRef
15.
go back to reference Ytterberg SR, Schnitzer TJ (1982) Serum interferon levels in patients with systemic lupus erythematosus. Arthritis Rheum 25(4):401–406CrossRef Ytterberg SR, Schnitzer TJ (1982) Serum interferon levels in patients with systemic lupus erythematosus. Arthritis Rheum 25(4):401–406CrossRef
16.
go back to reference Browne SK (2014) Anticytokine autoantibody-associated immunodeficiency. Annu Rev Immunol 32:635–657CrossRef Browne SK (2014) Anticytokine autoantibody-associated immunodeficiency. Annu Rev Immunol 32:635–657CrossRef
17.
go back to reference Barcenas-Morales G, Jandus P, Doffinger R (2016) Anticytokine autoantibodies in infection and inflammation: an update. Curr Opin Allergy Clin Immunol 16(6):523–529CrossRef Barcenas-Morales G, Jandus P, Doffinger R (2016) Anticytokine autoantibodies in infection and inflammation: an update. Curr Opin Allergy Clin Immunol 16(6):523–529CrossRef
18.
go back to reference Vincent T, Plawecki M, Goulabchand R, Guilpain P, Eliaou JF (2015) Emerging clinical phenotypes associated with anti-cytokine autoantibodies. Autoimmun Rev 14(6):528–535CrossRef Vincent T, Plawecki M, Goulabchand R, Guilpain P, Eliaou JF (2015) Emerging clinical phenotypes associated with anti-cytokine autoantibodies. Autoimmun Rev 14(6):528–535CrossRef
19.
go back to reference Uchida K, Beck DC, Yamamoto T, Berclaz PY, Abe S, Staudt MK et al (2007) GM-CSF autoantibodies and neutrophil dysfunction in pulmonary alveolar proteinosis. N Engl J Med 356(6):567–579CrossRef Uchida K, Beck DC, Yamamoto T, Berclaz PY, Abe S, Staudt MK et al (2007) GM-CSF autoantibodies and neutrophil dysfunction in pulmonary alveolar proteinosis. N Engl J Med 356(6):567–579CrossRef
20.
go back to reference Browne SK, Burbelo PD, Chetchotisakd P, Suputtamongkol Y, Kiertiburanakul S, Shaw PA et al (2012) Adult-onset immunodeficiency in Thailand and Taiwan. N Engl J Med 367(8):725–734CrossRef Browne SK, Burbelo PD, Chetchotisakd P, Suputtamongkol Y, Kiertiburanakul S, Shaw PA et al (2012) Adult-onset immunodeficiency in Thailand and Taiwan. N Engl J Med 367(8):725–734CrossRef
21.
go back to reference Hoflich C, Sabat R, Rosseau S, Temmesfeld B, Slevogt H, Docke WD et al (2004) Naturally occurring anti-IFN-gamma autoantibody and severe infections with Mycobacterium cheloneae and Burkholderia cocovenenans. Blood 103(2):673–675CrossRef Hoflich C, Sabat R, Rosseau S, Temmesfeld B, Slevogt H, Docke WD et al (2004) Naturally occurring anti-IFN-gamma autoantibody and severe infections with Mycobacterium cheloneae and Burkholderia cocovenenans. Blood 103(2):673–675CrossRef
22.
go back to reference Kampmann B, Hemingway C, Stephens A, Davidson R, Goodsall A, Anderson S et al (2005) Acquired predisposition to mycobacterial disease due to autoantibodies to IFN-gamma. J Clin Invest. 115(9):2480–2488CrossRef Kampmann B, Hemingway C, Stephens A, Davidson R, Goodsall A, Anderson S et al (2005) Acquired predisposition to mycobacterial disease due to autoantibodies to IFN-gamma. J Clin Invest. 115(9):2480–2488CrossRef
23.
go back to reference Lin CH, Chi CY, Shih HP, Ding JY, Lo CC, Wang SY et al (2016) Identification of a major epitope by anti-interferon-gamma autoantibodies in patients with mycobacterial disease. Nat Med 22(9):994–1001CrossRef Lin CH, Chi CY, Shih HP, Ding JY, Lo CC, Wang SY et al (2016) Identification of a major epitope by anti-interferon-gamma autoantibodies in patients with mycobacterial disease. Nat Med 22(9):994–1001CrossRef
24.
go back to reference Meager A, Vincent A, Newsom-Davis J, Willcox N (1997) Spontaneous neutralising antibodies to interferon–alpha and interleukin-12 in thymoma-associated autoimmune disease. Lancet 350(9091):1596–1597CrossRef Meager A, Vincent A, Newsom-Davis J, Willcox N (1997) Spontaneous neutralising antibodies to interferon–alpha and interleukin-12 in thymoma-associated autoimmune disease. Lancet 350(9091):1596–1597CrossRef
25.
go back to reference Burbelo PD, Browne SK, Sampaio EP, Giaccone G, Zaman R, Kristosturyan E et al (2010) Anti-cytokine autoantibodies are associated with opportunistic infection in patients with thymic neoplasia. Blood 116(23):4848–4858CrossRef Burbelo PD, Browne SK, Sampaio EP, Giaccone G, Zaman R, Kristosturyan E et al (2010) Anti-cytokine autoantibodies are associated with opportunistic infection in patients with thymic neoplasia. Blood 116(23):4848–4858CrossRef
26.
go back to reference Morimoto AM, Flesher DT, Yang J, Wolslegel K, Wang X, Brady A et al (2011) Association of endogenous anti-interferon-alpha autoantibodies with decreased interferon-pathway and disease activity in patients with systemic lupus erythematosus. Arthritis Rheum 63(8):2407–2415CrossRef Morimoto AM, Flesher DT, Yang J, Wolslegel K, Wang X, Brady A et al (2011) Association of endogenous anti-interferon-alpha autoantibodies with decreased interferon-pathway and disease activity in patients with systemic lupus erythematosus. Arthritis Rheum 63(8):2407–2415CrossRef
27.
go back to reference von Wussow P, Jakschies D, Hartung K, Deicher H (1988) Presence of interferon and anti-interferon in patients with systemic lupus erythematosus. Rheumatol Int 8(5):225–230CrossRef von Wussow P, Jakschies D, Hartung K, Deicher H (1988) Presence of interferon and anti-interferon in patients with systemic lupus erythematosus. Rheumatol Int 8(5):225–230CrossRef
28.
go back to reference Ching KH, Burbelo PD, Tipton C, Wei C, Petri M, Sanz I et al (2012) Two major autoantibody clusters in systemic lupus erythematosus. PLoS ONE 7(2):e32001CrossRef Ching KH, Burbelo PD, Tipton C, Wei C, Petri M, Sanz I et al (2012) Two major autoantibody clusters in systemic lupus erythematosus. PLoS ONE 7(2):e32001CrossRef
29.
go back to reference Gupta S, Tatouli IP, Rosen LB, Hasni S, Alevizos I, Manna ZG et al (2016) Distinct functions of autoantibodies against interferon in systemic lupus erythematosus: a comprehensive analysis of anticytokine autoantibodies in common rheumatic diseases. Arthritis Rheumatol. 68(7):1677–1687CrossRef Gupta S, Tatouli IP, Rosen LB, Hasni S, Alevizos I, Manna ZG et al (2016) Distinct functions of autoantibodies against interferon in systemic lupus erythematosus: a comprehensive analysis of anticytokine autoantibodies in common rheumatic diseases. Arthritis Rheumatol. 68(7):1677–1687CrossRef
30.
go back to reference Meyer S, Woodward M, Hertel C, Vlaicu P, Haque Y, Karner J et al (2016) AIRE-deficient patients harbor unique high-affinity disease-ameliorating autoantibodies. Cell 166(3):582–595CrossRef Meyer S, Woodward M, Hertel C, Vlaicu P, Haque Y, Karner J et al (2016) AIRE-deficient patients harbor unique high-affinity disease-ameliorating autoantibodies. Cell 166(3):582–595CrossRef
31.
go back to reference Burbelo PD, Lebovitz EE, Notkins AL (2015) Luciferase immunoprecipitation systems for measuring antibodies in autoimmune and infectious diseases. Transl Res. 165(2):325–335CrossRef Burbelo PD, Lebovitz EE, Notkins AL (2015) Luciferase immunoprecipitation systems for measuring antibodies in autoimmune and infectious diseases. Transl Res. 165(2):325–335CrossRef
32.
go back to reference Burbelo PD, Leahy HP, Issa AT, Groot S, Baraniuk JN, Nikolov NP et al (2009) Sensitive and robust luminescent profiling of anti-La and other autoantibodies in Sjogren’s syndrome. Autoimmunity. 42(6):515–524CrossRef Burbelo PD, Leahy HP, Issa AT, Groot S, Baraniuk JN, Nikolov NP et al (2009) Sensitive and robust luminescent profiling of anti-La and other autoantibodies in Sjogren’s syndrome. Autoimmunity. 42(6):515–524CrossRef
33.
go back to reference Burbelo PD, Ching KH, Issa AT, Loftus CM, Li Y, Satoh M et al (2009) Rapid serological detection of autoantibodies associated with Sjogren’s syndrome. J Transl Med. 7:83CrossRef Burbelo PD, Ching KH, Issa AT, Loftus CM, Li Y, Satoh M et al (2009) Rapid serological detection of autoantibodies associated with Sjogren’s syndrome. J Transl Med. 7:83CrossRef
34.
go back to reference Burbelo PD, Ching KH, Han BL, Bush ER, Reeves WH, Iadarola MJ (2010) Extraordinary antigenicity of the human Ro52 autoantigen. Am J Transl Res. 2(2):145–155PubMedPubMedCentral Burbelo PD, Ching KH, Han BL, Bush ER, Reeves WH, Iadarola MJ (2010) Extraordinary antigenicity of the human Ro52 autoantigen. Am J Transl Res. 2(2):145–155PubMedPubMedCentral
35.
go back to reference Volchenkov R, Jonsson R, Appel S (2012) Anti-Ro and anti-La autoantibody profiling in Norwegian patients with primary Sjogren’s syndrome using luciferase immunoprecipitation systems (LIPS). Scand J Rheumatol 41(4):314–315CrossRef Volchenkov R, Jonsson R, Appel S (2012) Anti-Ro and anti-La autoantibody profiling in Norwegian patients with primary Sjogren’s syndrome using luciferase immunoprecipitation systems (LIPS). Scand J Rheumatol 41(4):314–315CrossRef
36.
go back to reference Burbelo PD, Teos LY, Herche JL, Iadarola MJ, Alevizos I (2018) Autoantibodies against the immunoglobulin-binding region of Ro52 link its autoantigenicity with pathogen neutralization. Sci Rep. 8(1):3345CrossRef Burbelo PD, Teos LY, Herche JL, Iadarola MJ, Alevizos I (2018) Autoantibodies against the immunoglobulin-binding region of Ro52 link its autoantigenicity with pathogen neutralization. Sci Rep. 8(1):3345CrossRef
37.
go back to reference Bayat A, Burbelo PD, Browne SK, Quinlivan M, Martinez B, Holland SM et al (2015) Anti-cytokine autoantibodies in postherpetic neuralgia. J Transl Med. 13:333CrossRef Bayat A, Burbelo PD, Browne SK, Quinlivan M, Martinez B, Holland SM et al (2015) Anti-cytokine autoantibodies in postherpetic neuralgia. J Transl Med. 13:333CrossRef
38.
go back to reference Bichele R, Karner J, Truusalu K, Smidt I, Mandar R, Conti HR et al (2018) IL-22 neutralizing autoantibodies impair fungal clearance in murine oropharyngeal candidiasis model. Eur J Immunol 48(3):464–470CrossRef Bichele R, Karner J, Truusalu K, Smidt I, Mandar R, Conti HR et al (2018) IL-22 neutralizing autoantibodies impair fungal clearance in murine oropharyngeal candidiasis model. Eur J Immunol 48(3):464–470CrossRef
39.
go back to reference Burbelo PD, Seam N, Groot S, Ching KH, Han BL, Meduri GU et al (2010) Rapid induction of autoantibodies during ARDS and septic shock. J Transl Med. 8:97CrossRef Burbelo PD, Seam N, Groot S, Ching KH, Han BL, Meduri GU et al (2010) Rapid induction of autoantibodies during ARDS and septic shock. J Transl Med. 8:97CrossRef
40.
go back to reference Karner J, Pihlap M, Ranki A, Krohn K, Trebusak Podkrajsek K, Bratanic N et al (2016) IL-6-specific autoantibodies among APECED and thymoma patients. Immun Inflamm Dis. 4(2):235–243CrossRef Karner J, Pihlap M, Ranki A, Krohn K, Trebusak Podkrajsek K, Bratanic N et al (2016) IL-6-specific autoantibodies among APECED and thymoma patients. Immun Inflamm Dis. 4(2):235–243CrossRef
41.
go back to reference Vitali C, Bombardieri S, Jonsson R, Moutsopoulos HM, Alexander EL, Carsons SE et al (2002) Classification criteria for Sjogren’s syndrome: a revised version of the European criteria proposed by the American-European Consensus Group. Ann Rheum Dis 61(6):554–558CrossRef Vitali C, Bombardieri S, Jonsson R, Moutsopoulos HM, Alexander EL, Carsons SE et al (2002) Classification criteria for Sjogren’s syndrome: a revised version of the European criteria proposed by the American-European Consensus Group. Ann Rheum Dis 61(6):554–558CrossRef
42.
go back to reference Rosen LB, Freeman AF, Yang LM, Jutivorakool K, Olivier KN, Angkasekwinai N et al (2013) Anti-GM-CSF autoantibodies in patients with cryptococcal meningitis. J Immunol. 190(8):3959–3966CrossRef Rosen LB, Freeman AF, Yang LM, Jutivorakool K, Olivier KN, Angkasekwinai N et al (2013) Anti-GM-CSF autoantibodies in patients with cryptococcal meningitis. J Immunol. 190(8):3959–3966CrossRef
44.
go back to reference Saijo T, Chen J, Chen SC, Rosen LB, Yi J, Sorrell TC et al (2014) Anti-granulocyte-macrophage colony-stimulating factor autoantibodies are a risk factor for central nervous system infection by Cryptococcus gattii in otherwise immunocompetent patients. MBio. 5(2):e00912–e00914CrossRef Saijo T, Chen J, Chen SC, Rosen LB, Yi J, Sorrell TC et al (2014) Anti-granulocyte-macrophage colony-stimulating factor autoantibodies are a risk factor for central nervous system infection by Cryptococcus gattii in otherwise immunocompetent patients. MBio. 5(2):e00912–e00914CrossRef
45.
go back to reference Walter JE, Rosen LB, Csomos K, Rosenberg JM, Mathew D, Keszei M et al (2015) Broad-spectrum antibodies against self-antigens and cytokines in RAG deficiency. J Clin Invest. 125(11):4135–4148CrossRef Walter JE, Rosen LB, Csomos K, Rosenberg JM, Mathew D, Keszei M et al (2015) Broad-spectrum antibodies against self-antigens and cytokines in RAG deficiency. J Clin Invest. 125(11):4135–4148CrossRef
46.
go back to reference Bekisz J, Schmeisser H, Hernandez J, Goldman ND, Zoon KC (2004) Human interferons alpha, beta and omega. Growth Factors 22(4):243–251CrossRef Bekisz J, Schmeisser H, Hernandez J, Goldman ND, Zoon KC (2004) Human interferons alpha, beta and omega. Growth Factors 22(4):243–251CrossRef
47.
go back to reference Hagberg N, Ronnblom L (2015) Systemic lupus erythematosus—a disease with a dysregulated type i interferon system. Scand J Immunol 82(3):199–207CrossRef Hagberg N, Ronnblom L (2015) Systemic lupus erythematosus—a disease with a dysregulated type i interferon system. Scand J Immunol 82(3):199–207CrossRef
48.
go back to reference Theander E, Jonsson R, Sjostrom B, Brokstad K, Olsson P, Henriksson G (2015) Prediction of Sjogren’s syndrome years before diagnosis and identification of patients with early onset and severe disease course by autoantibody profiling. Arthritis Rheumatol. 67(9):2427–2436CrossRef Theander E, Jonsson R, Sjostrom B, Brokstad K, Olsson P, Henriksson G (2015) Prediction of Sjogren’s syndrome years before diagnosis and identification of patients with early onset and severe disease course by autoantibody profiling. Arthritis Rheumatol. 67(9):2427–2436CrossRef
49.
go back to reference de Lemos Rieper C, Galle P, Hansen MB (2009) Characterization and potential clinical applications of autoantibodies against cytokines. Cytokine Growth Factor Rev 20(1):61–75CrossRef de Lemos Rieper C, Galle P, Hansen MB (2009) Characterization and potential clinical applications of autoantibodies against cytokines. Cytokine Growth Factor Rev 20(1):61–75CrossRef
50.
go back to reference Wildbaum G, Nahir MA, Karin N (2003) Beneficial autoimmunity to proinflammatory mediators restrains the consequences of self-destructive immunity. Immunity 19(5):679–688CrossRef Wildbaum G, Nahir MA, Karin N (2003) Beneficial autoimmunity to proinflammatory mediators restrains the consequences of self-destructive immunity. Immunity 19(5):679–688CrossRef
51.
go back to reference Khamashta M, Merrill JT, Werth VP, Furie R, Kalunian K, Illei GG et al (2016) Sifalimumab, an anti-interferon-alpha monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis 75(11):1909–1916CrossRef Khamashta M, Merrill JT, Werth VP, Furie R, Kalunian K, Illei GG et al (2016) Sifalimumab, an anti-interferon-alpha monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis 75(11):1909–1916CrossRef
52.
go back to reference Lauwerys BR, Hachulla E, Spertini F, Lazaro E, Jorgensen C, Mariette X et al (2013) Down-regulation of interferon signature in systemic lupus erythematosus patients by active immunization with interferon alpha-kinoid. Arthritis Rheum 65(2):447–456CrossRef Lauwerys BR, Hachulla E, Spertini F, Lazaro E, Jorgensen C, Mariette X et al (2013) Down-regulation of interferon signature in systemic lupus erythematosus patients by active immunization with interferon alpha-kinoid. Arthritis Rheum 65(2):447–456CrossRef
Metadata
Title
Clinical features of Sjögren’s syndrome patients with autoantibodies against interferons
Authors
Peter D. Burbelo
Sarah Browne
Steve M. Holland
Michael J. Iadarola
Ilias Alevizos
Publication date
01-12-2019
Publisher
Springer Berlin Heidelberg
Published in
Clinical and Translational Medicine / Issue 1/2019
Electronic ISSN: 2001-1326
DOI
https://doi.org/10.1186/s40169-018-0218-1

Other articles of this Issue 1/2019

Clinical and Translational Medicine 1/2019 Go to the issue