Interferon alpha in systemic lupus erythematosus

Timothy B Niewold, Daniel N Clark, Rafah Salloum, Brian D Poole, Timothy B Niewold, Daniel N Clark, Rafah Salloum, Brian D Poole

Abstract

The pleiotropic cytokine interferon alpha is involved in multiple aspects of lupus etiology and pathogenesis. Interferon alpha is important under normal circumstances for antiviral responses and immune activation. However, heightened levels of serum interferon alpha and expression of interferon response genes are common in lupus patients. Lupus-associated autoantibodies can drive the production of interferon alpha and heightened levels of interferon interfere with immune regulation. Several genes in the pathways leading to interferon production or signaling are associated with risk for lupus. Clinical and cellular manifestations of excess interferon alpha in lupus combined with the genetic risk factors associated with interferon make this cytokine a rare bridge between genetic risk and phenotypic effects. Interferon alpha influences the clinical picture of lupus and may represent a therapeutic target. This paper provides an overview of the cellular, genetic, and clinical aspects of interferon alpha in lupus.

Figures

Figure 1
Figure 1
Putative source and effects of interferon alpha in lupus. RNA-containing complexes from apoptotic cells are bound by autoantibodies. These immune complexes are internalized after binding to FC receptors on plasmacytoid dendritic cells and stimulate toll-like receptors in the endosomes. Toll-like receptor ligation drives production of interferon alpha, leading to alteration of T-cell profiles, disruption of regulatory T-cell networks, and alteration of B-cell development.
Figure 2
Figure 2
Multiple genes involved in interferon production and regulation are associated with risk for lupus. Shown are components of the signal transduction pathway from TLR stimulation by nucleic acids to IFN production. Genes that have been associated with risk for lupus are marked (*). IFN: interferon, IRAK: interleukin-1 receptor-associated kinase, IRF: interferon regulatory factor, MyD88: myeloid differentiation primary response gene 88, OPN: osteopontin, pDC: plasmacytoid dendritic cell, TLR: toll-like receptor, TNFAIP3: tumor necrosis factor alpha induced protein 3, TNIP1: TNFAIP3 interacting protein 1, and TRAF6: tumor necrosis factor receptor-associated factor 6.

References

    1. Fitzgerald-Bocarsly P, Dai J, Singh S. Plasmacytoid dendritic cells and type I IFN: 50 years of convergent history. Cytokine and Growth Factor Reviews. 2008;19(1):3–19.
    1. Ito T, Wang Y-H, Liu Y-J. Plasmacytoid dendritic cell precursors/type I interferon-producing cells sense viral infection by Toll-like receptor (TLR) 7 and TLR9. Springer Seminars in Immunopathology. 2005;26(3):221–229.
    1. Lövgren T, Eloranta M-L, Kastner B, Wahren-Herlenius M, Alm GV, Rönnblom L. Induction of interferon-alpha by immune complexes or liposomes containing systemic lupus erythematosus autoantigen- and Sjogren’s syndrome autoantigen-associated RNA. Arthritis and Rheumatism. 2006;54(6):1917–1927.
    1. Schoenemeyer A, Barnes BJ, Mancl ME, et al. The interferon regulatory factor, IRF5, is a central mediator of toll-like receptor 7 signaling. Journal of Biological Chemistry. 2005;280(17):17005–17012.
    1. Hoshino K, Sugiyama T, Matsumoto M, et al. IκB kinase-α is critical for interferon-α production induced by Toll-like receptors 7 and 9. Nature. 2006;440(7086):949–953.
    1. Kawai T, Sato S, Ishii KJ, et al. Interferon-α induction through Toll-like receptors involves a direct interaction of IRF7 with MyD88 and TRAF6. Nature Immunology. 2004;5(10):1061–1068.
    1. Löseke S, Grage-Griebenow E, Heine H, et al. In vitro-generated viral double-stranded RNA in contrast to polyinosinic: polycytidylic acid induces interferon-α in human plasmacytoid dendritic cells. Scandinavian Journal of Immunology. 2006;63(4):264–274.
    1. Fairhurst A-M, Hwang S-H, Wang A, et al. Yaa autoimmune phenotypes are conferred by overexpression of TLR7. European Journal of Immunology. 2008;38(7):1971–1978.
    1. Pisitkun P, Deane JA, Difilippantonio MJ, Tarasenko T, Satterthwaite AB, Bolland S. Autoreactive B cell responses to RNA-related antigens due to TLR7 gene duplication. Science. 2006;312(5780):1669–1672.
    1. Subramanian S, Tus K, Li Q-Z, et al. A Tlr7 translocation accelerates systemic autoimmunity in murine lupus. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(26):9970–9975.
    1. Ramanujam M, Kahn P, Huang W, et al. Interferon-α treatment of Female (NZW × BXSB)F1 mice mimics some but not all features associated with the Yaa mutation. Arthritis and Rheumatism. 2009;60(4):1096–1101.
    1. Barrat FJ, Meeker T, Chan JH, Guiducci C, Coffmann RL. Treatment of lupus-prone mice with a dual inhibitor of TLR7 and TLR9 leads to reduction of autoantibody production and amelioration of disease symptoms. European Journal of Immunology. 2007;37(12):3582–3586.
    1. Barrat FJ, Meeker T, Gregorio J, et al. Nucleic acids of mammalian origin can act as endogenous ligands for Toll-like receptors and may promote systemic lupus erythematosus. Journal of Experimental Medicine. 2005;202(8):1131–1139.
    1. Batteux F, Palmer P, Daëron M, Weill B, Lebon P. FCγRII (CD32)-dependent induction of interferon-alpha by serum from patients with lupus erythematosus. European Cytokine Network. 1999;10(4):509–514.
    1. Båve U, Magnusson M, Eloranta M-L, Perers A, Alm GV, Rönnblom L. FcγRIIa is expressed on natural IFN-α-producing cells (plasmacytoid dendritic cells) and is required for the IFN-α production induced by apoptotic cells combined with Lupus IgG. Journal of Immunology. 2003;171(6):3296–3302.
    1. Savarese E, Chae O-W, Trowitzsch S, et al. U1 small nuclear ribonucleoprotein immune complexes induce type I interferon in plasmacytoid dendritic cells through TLR7. Blood. 2006;107(8):3229–3234.
    1. Rajagopal D, Paturel C, Morel Y, Uematsu S, Akira S, Diebold SS. Plasmacytoid dendritic cell-derived type I interferon is crucial for the adjuvant activity of Toll-like receptor 7 agonists. Blood. 2010;115(10):1949–1957.
    1. Asselin-Paturel C, Brizard G, Chemin K, et al. Type I interferon dependence of plasmacytoid dendritic cell activation and migration. Journal of Experimental Medicine. 2005;201(7):1157–1167.
    1. Niewold TB, Rivera TL, Buyon JP, Crow MK. Serum type I interferon activity is dependent on maternal diagnosis in anti-SSA/Ro-positive mothers of children with neonatal lupus. Arthritis and Rheumatism. 2008;58(2):541–546.
    1. Farkas A, Tonel G, Nestle FO. Interferon-α and viral triggers promote functional maturation of human monocyte-derived dendritic cells. British Journal of Dermatology. 2008;158(5):921–929.
    1. Sakakibara M, Kanto T, Inoue M, et al. Quick generation of fully mature dendritic cells from monocytes with OK432, low-dose prostanoid, and interferon-α as potent immune enhancers. Journal of Immunotherapy. 2006;29(1):67–77.
    1. Blanco P, Palucka AK, Gill M, Pascual V, Banchereau J. Induction of dendritic cell differentiation by IFN-α in systemic lupus erythematosus. Science. 2001;294(5546):1540–1543.
    1. Ito T, Amakawa R, Inaba M, et al. Plasmacytoid dendritic cells regulate Th cell responses through OX40 ligand and type I IFNs. Journal of Immunology. 2004;172(7):4253–4259.
    1. Mohty M, Vialle-Castellano A, Nunes JA, Isnardon D, Olive D, Gaugler B. IFN-α skews monocyte differentiation into toll-like receptor 7-expressing dendritic cells with potent functional activities. Journal of Immunology. 2003;171(7):3385–3393.
    1. Cucak H, Yrlid U, Reizis B, Kalinke U, Johansson-Lindbom B. Type I interferon signaling in dendritic cells stimulates the development of lymph-node-resident T follicular helper cells. Immunity. 2009;31(3):491–501.
    1. Baráth S, Soltész P, Kiss E, et al. The severity of systemic lupus erythematosus negatively correlates with the increasing number of CD4+CD25highFoxP3+ regulatory T cells during repeated plasmapheresis treatments of patients. Autoimmunity. 2007;40(7):521–528.
    1. Bonelli M, Savitskaya A, Von Dalwigk K, et al. Quantitative and qualitative deficiencies of regulatory T cells in patients with systemic lupus erythematosus (SLE) International Immunology. 2008;20(7):861–868.
    1. Hu S, Xiao W, Kong F, Ke D, Qin R, Su M. Regulatory T cells and their molecular markers in peripheral blood of the patients with systemic lupus erythematosus. Journal of Huazhong University of Science and Technology—Medical Science. 2008;28(5):549–552.
    1. Suen J-L, Li H-T, Jong Y-J, Chiang B-L, Yen J-H. Altered homeostasis of CD4+FoxP3+ regulatory T-cell subpopulations in systemic lupus erythematosus. Immunology. 2009;127(2):196–205.
    1. Venigalla RKC, Tretter T, Krienke S, et al. Reduced CD4+,CD25-T cell sensitivity to the suppressive function of CD4+, CD25high, CD127−/low regulatory T cells in patients with active systemic lupus erythematosus. Arthritis and Rheumatism. 2008;58(7):2120–2130.
    1. Yates J, Whittington A, Mitchell P, Lechler RI, Lightstone L, Lombardi G. Natural regulatory T cells: number and function are normal in the majority of patients with lupus nephritis. Clinical and Experimental Immunology. 2008;153(1):44–55.
    1. Gómez J, Prado C, López P, Suárez A, Gutiérrez C. Conserved anti-proliferative effect and poor inhibition of TNFα secretion by regulatory CD4+CD25+ T cells in patients with systemic lupus erythematosus. Clinical Immunology. 2009;132(3):385–392.
    1. Gigante M, Mandic M, Wesa AK, et al. Interferon-alpha (IFN-α)-conditioned DC preferentially stimulate type-1 and limit treg-type in vitro T-cell responses from RCC patients. Journal of Immunotherapy. 2008;31(3):254–262.
    1. Yan B, Ye S, Chen G, Kuang M, Shen N, Chen S. Dysfunctional CD4+,CD25+ regulatory T cells in untreated active systemic lupus erythematosus secondary to interferon-α-producing antigen-presenting cells. Arthritis and Rheumatism. 2008;58(3):801–812.
    1. Ruuth K, Carlsson L, Hallberg B, Lundgren E. Interferon-α promotes survival of human primary B-lymphocytes via phosphatidylinositol 3-kinase. Biochemical and Biophysical Research Communications. 2001;284(3):583–586.
    1. Oka H, Hirohata S, Inoue T, Ito K. Effects of interferon-α on human B cell responsiveness: biphasic effects in cultures stimulated with Staphylococcus aureus. Cellular Immunology. 1992;139(2):478–492.
    1. Oka H, Hirohata S. Regulation of human B cell responsiveness by interferon-α: interferon-α-mediated suppression of B cell function is reversed through direct interactions between monocytes and B cells. Cellular Immunology. 1993;146(2):238–248.
    1. Alarcón-Segovia D, Alarcón-Riquelme ME, Cardiel MH, et al. Familial aggregation of systemic lupus erythematosus, rheumatoid arthritis, and other autoimmune diseases in 1,177 lupus patients from the GLADEL cohort. Arthritis and Rheumatism. 2005;52(4):1138–1147.
    1. Deapen D, Escalante A, Weinrib L, et al. A revised estimate of twin concordance in systemic lupus erythematosus. Arthritis and Rheumatism. 1992;35(3):311–318.
    1. Wandstrat A, Wakeland E. The genetics of complex autoimmune diseases: non-MHC susceptibility genes. Nature Immunology. 2001;2(9):802–809.
    1. Sebastiani GD, Galeazzi M. Immunogenetic studies on systemic lupus erythematosus. Lupus. 2009;18(10):878–883.
    1. Moser KL, Kelly JA, Lessard CJ, Harley JB. Recent insights into the genetic basis of systemic lupus erythematosus. Genes and Immunity. 2009;10(5):373–379.
    1. Harley ITW, Kaufman KM, Langefeld CD, Harley JB, Kelly JA. Genetic susceptibility to SLE: new insights from fine mapping and genome-wide association studies. Nature Reviews Genetics. 2009;10(5):285–290.
    1. Graham RR, Hom G, Ortmann W, Behrens TW. Review of recent genome-wide association scans in lupus. Journal of Internal Medicine. 2009;265(6):680–688.
    1. Rhodes B, Vyse TJ. The genetics of SLE: an update in the light of genome-wide association studies. Rheumatology. 2008;47(11):1603–1611.
    1. Lee YH, Song GG. Association between the rs2004640 functional polymorphism of interferon regulatory factor 5 and systemic lupus erythematosus: a meta-analysis. Rheumatology International. 2009;29(10):1137–1142.
    1. Kawasaki A, Kyogoku C, Ohashi J, et al. Association of IRF5 polymorphisms with systemic lupus erythematosus in a Japanese population: support for a crucial role of intron 1 polymorphisms. Arthritis and Rheumatism. 2008;58(3):826–834.
    1. Kelly JA, Kelley JM, Kaufman KM, et al. Interferon regulatory factor-5 is genetically associated with systemic lupus erythematosus in African Americans. Genes and Immunity. 2008;9(3):187–194.
    1. Reddy MVPL, Velázquez-Cruz R, Baca V, et al. Genetic association of IRF5 with SLE in Mexicans: higher frequency of the risk haplotype and its homozygozity than Europeans. Human Genetics. 2007;121(6):721–727.
    1. Shimane K, Kochi Y, Yamada R, et al. A single nucleotide polymorphism in the IRF5 promoter region is associated with susceptibility to rheumatoid arthritis in the Japanese population. Annals of the Rheumatic Diseases. 2009;68(3):377–383.
    1. Graham RR, Kyogoku C, Sigurdsson S, et al. Three functional variants of IFN regulatory factor 5 (IRF5) define risk and protective haplotypes for human lupus. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(16):6758–6763.
    1. Graham RR, Kyogoku C, Sigurdsson S, et al. Three functional variants of IFN regulatory factor 5 (IRF5) define risk and protective haplotypes for human lupus. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(16):6758–6763.
    1. Niewold TB, Kelly JA, Flesch MH, Espinoza LR, Harley JB, Crow MK. Association of the IRF5 risk haplotype with high serum interferon-α activity in systemic lupus erythematosus patients. Arthritis and Rheumatism. 2008;58(8):2481–2487.
    1. Harley JB, Alarcón-Riquelme ME, Criswell LA, et al. Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nature Genetics. 2008;40(2):204–210.
    1. Suarez-Gestal M, Calaza M, Endreffy E, et al. Replication of recently identified systemic lupus erythematosus genetic associations: a case-control study. Arthritis Research and Therapy. 2009;11(3, article 69)
    1. Salloum R, Franek B, Kariuki S, Utset T, Niewold TT. Genetic variation at the IRF7/KIAA1542 locus is associated with autoantibody profile and serum interferon alpha levels in lupus patients. Clinical Immunology. 2009;131(supplement 1):p. S54.
    1. Armstrong DL, Reiff A, Myones BL, et al. Identification of new SLE-associated genes with a two-step Bayesian study design. Genes and Immunity. 2009;10(5):446–456.
    1. Xu C-J, Zhang W-H, Pan H-F, Li X-P, Xu J-H, Ye D-Q. Association study of a single nucleotide polymorphism in the exon 2 region of toll-like receptor 9 (TLR9) gene with susceptibility to systemic lupus erythematosus among Chinese. Molecular Biology Reports. 2009;36(8):2245–2248.
    1. Jacob CO, Zhu J, Armstrong DL, et al. Identification of IRAK1 as a risk gene with critical role in the pathogenesis of systemic lupus erythematosus. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(15):6256–6261.
    1. Bates JS, Lessard CJ, Leon JM, et al. Meta-analysis and imputation identifies a 109kb risk haplotype spanning TNFAIP3 associated with lupus nephritis and hematologic manifestations. Genes and Immunity. 2009;10(5):470–477.
    1. Gateva V, Sandling JK, Hom G, et al. A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nature Genetics. 2009;41(11):1228–1233.
    1. Graham RR, Cotsapas C, Davies L, et al. Genetic variants near TNFAIP3 on 6q23 are associated with systemic lupus erythematosus. Nature Genetics. 2008;40(9):1059–1061.
    1. Han J-W, Zheng H-F, Cui Y, et al. Genome-wide association study in a Chinese Han population identifies nine new susceptibility loci for systemic lupus erythematosus. Nature Genetics. 2009;41(11):1234–1237.
    1. Musone SL, Taylor KE, Lu TT, et al. Multiple polymorphisms in the TNFAIP3 region are independently associated with systemic lupus erythematosus. Nature Genetics. 2008;40(9):1062–1064.
    1. Honma K, Tsuzuki S, Nakagawa M, et al. TNFAIP3/A20 functions as a novel tumor suppressor gene in several subtypes of non-Hodgkin lymphomas. Blood. 2009;114(12):2467–2475.
    1. Kato M, Sanada M, Kato I, et al. Frequent inactivation of A20 in B-cell lymphomas. Nature. 2009;459(7247):712–716.
    1. Kariuki SN, Kirou KA, MacDermott EJ, Barillas-Arias L, Crow MK, Niewold TB. Cutting edge: autoimmune disease risk variant of STAT4 confers increased sensitivity to IFN-alpha in lupus patients in vivo. Journal of Immunology. 2009;182(1):34–38.
    1. Abelson AK, Delgado-Vega AM, Kozyrev SV, et al. STAT4 associates with systemic lupus erythematosus through two independent effects that correlate with gene expression and act additively with IRF5 to increase risk. Annals of the Rheumatic Diseases. 2009;68(11):1746–1753.
    1. Sigurdsson S, Nordmark G, Garnier S, et al. A risk haplotype of STAT4 for systemic lupus erythematosus is over-expressed, correlates with anti-dsDNA and shows additive effects with two risk alleles of IRF5. Human Molecular Genetics. 2008;17(18):2868–2876.
    1. Cao W, Liu Y-J. Opn: key regulator of pDC interferon production. Nature Immunology. 2006;7(5):441–443.
    1. Kariuki SN, Moore JG, Kirou KA, Crow MK, Utset TO, Niewold TB. Age- and gender-specific modulation of serum osteopontin and interferon-α by osteopontin genotype in systemic lupus erythematosus. Genes and Immunity. 2009;10(5):487–494.
    1. Ronnblom LE, Alm GV, Oberg KE. Possible induction of systemic lupus erythematosus by interferon α-treatment in a patient with a malignant carcinoid tumour. Journal of Internal Medicine. 1990;227(3):207–210.
    1. Niewold TB, Swedler WI. Systemic lupus erythematosus arising during interferon-alpha therapy for cryoglobulinemic vasculitis associated with hepatitis C. Clinical Rheumatology. 2005;24(2):178–181.
    1. Ioannou Y, Isenberg DA. Current evidence for the induction of autoimmune rheumatic manifestations by cytokine therapy. Arthritis and Rheumatism. 2000;43(7):1431–1442.
    1. Gota C, Calabrese L. Induction of clinical autoimmune disease by therapeutic interferon-α. Autoimmunity. 2003;36(8):511–518.
    1. Tan EM, Cohen AS, Fries JF. The 1982 revised criteria for the classification of systemic lupus erythrematosus. Arthritis and Rheumatism. 1982;25(11):1271–1277.
    1. Ho V, McLean A, Terry S. Severe systemic lupus erythematosus induced by antiviral treatment for hepatitis C. Journal of Clinical Rheumatology. 2008;14(3):166–168.
    1. Niewold TB. Interferon alpha-induced lupus: proof of principle. Journal of Clinical Rheumatology. 2008;14(3):131–132.
    1. Niewold TB, Hua J, Lehman TJA, Harley JB, Crow MK. High serum IFN-α activity is a heritable risk factor for systemic lupus erythematosus. Genes and Immunity. 2007;8(6):492–502.
    1. Niewold TB, Adler JE, Glenn SB, Lehman TJA, Harley JB, Crow MK. Age- and sex-related patterns of serum interferon-α activity in lupus families. Arthritis and Rheumatism. 2008;58(7):2113–2119.
    1. Weckerle CE, Niewold TB. The unexplained female predominance of systemic lupus erythematosus: clues from genetic and cytokine studies. Clinical Reviews in Allergy and Immunology, 2010. In press.
    1. Kirou KA, Lee C, George S, Louca K, Peterson MGE, Crow MK. Activation of the interferon-α pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis and Rheumatism. 2005;52(5):1491–1503.
    1. Dall’Era MC, Cardarelli PM, Preston BT, Witte A, Davis JC., Jr. Type I interferon correlates with serological and clinical manifestations of SLE. Annals of the Rheumatic Diseases. 2005;64(12):1692–1697.
    1. Feng X, Wu H, Grossman JM, et al. Association of increased interferon-inducible gene expression with disease activity and lupus nephritis in patients with systemic lupus erythematosus. Arthritis and Rheumatism. 2006;54(9):2951–2962.
    1. Zhuang H, Narain S, Sobel E, et al. Association of anti-nucleoprotein autoantibodies with upregulation of type I interferon-inducible gene transcripts and dendritic cell maturation in systemic lupus erythematosus. Clinical Immunology. 2005;117(3):238–250.
    1. Landolt-Marticorena C, Bonventi G, Lubovich A, et al. Lack of association between the interferon-α signature and longitudinal changes in disease activity in systemic lupus erythematosus. Annals of the Rheumatic Diseases. 2009;68(9):1440–1446.
    1. Petri M, Singh S, Tesfasyone H, et al. Longitudinal expression of type I interferon responsive genes in systemic lupus erythematosus. Lupus. 2009;18(11):980–989.
    1. Bauer JW, Petri M, Batliwalla FM, et al. Interferon-regulated chemokines as biomarkers of systemic lupus erythematosus disease activity: a validation study. Arthritis and Rheumatism. 2009;60(10):3098–3107.
    1. Yao Y, Richman L, Higgs BW, et al. Neutralization of interferon-α/β-inducible genes and downstream effect in a phase I trial of an anti-interferon-α monoclonal antibody in systemic lupus erythematosus. Arthritis and Rheumatism. 2009;60(6):1785–1796.

Source: PubMed

3
구독하다