The genetics of type I interferon in systemic lupus erythematosus

https://doi.org/10.1016/j.coi.2012.07.008Get rights and content

The discovery that type I interferon (IFN)-inducible genes were strongly upregulated in peripheral blood in SLE over a decade ago sparked interest in understanding the relationship between type I IFN and SLE. Genome-wide association studies provide strong genetic evidence that type I IFNs are important for SLE risk. Of 47 genetic variants associated with SLE, over half (27/47, 57%) can be linked to type I IFN production or signaling. The recent identification of single gene mutations for disorders that share features with SLE – Aicardi–Goutières syndrome, chilblain lupus, and spondyloenchondrodysplasia – provide additional support for the hypothesis that type I IFNs are central drivers of SLE pathogenesis. These insights provide significant focus for efforts to tackle SLE therapeutically.

Highlights

► Type I IFN appears to be a central pathway dysregulated in SLE. ► Most SLE patients have a prominent IFN gene signature in blood. ► Over half of the common alleles associated with SLE map into the type I IFN pathway. ► Rare, monogenic SLE-like diseases also show activation of type I IFN.

Introduction

SLE is characterized by IgG autoantibodies to highly conserved nuclear antigens such as double-stranded DNA (dsDNA), histones and ribonuclear proteins. Immune complexes comprised of IgG and self-antigens deposit in tissues and blood vessels, including skin, kidneys, and pleura/pericardium, leading to end-organ disease. Additional autoantibodies directly target cell surface antigens, causing cytopenias and other pathologies. Similar to most autoimmune diseases, human leukocyte antigen (HLA) genes are associated with increased SLE risk, with strongest evidence for the class II HLA alleles DRB1*15:01 and DRB1*03:01 [1]. Although the survival rate for SLE has improved greatly over the years, patients are 2.5 times more likely to die than the general population [2], and there is a pressing need to develop new therapies.

Here, we review evidence from genomic, candidate gene, and genomewide association studies (GWAS), together with recent findings from monogenic SLE and SLE-related diseases, that support the hypothesis that type I interferons (IFNs) are key factors in SLE pathogenesis.

Section snippets

Pleiotropic effects of type I IFN and related pathways

Type I IFNs (13 IFN-α isotypes, IFN-β, IFN-ɛ, IFN-κ, and IFN-ω) are produced primarily by plasmacytoid dendritic cells (pDCs), and are regulated by engagement of cell membrane or endosomal receptors, such as certain toll-like receptors (TLRs) (TLR 3, 7/8, 9), that recognize nucleic acids in viruses, bacteria and protozoa. Intracellular viral RNA and DNA are recognized by a specialized set of receptors (e.g. RIG-I, IFIH1) that also trigger type I IFN production. Type I IFN binds to the

The IFN gene expression signature and IFN-α in SLE

Using microarray gene expression analysis, we generated initial data in 1999 showing that about two-thirds of adult SLE patients exhibited elevated expression of type I IFN-inducible genes in peripheral blood cells, and coined the term ‘IFN signature’ [9••]. Additional groups reported similar findings in SLE [10••, 11, 12], and subsequently the IFN signature was found to be prominent in Sjögren's syndrome, dermatomyositis, subacute cutaneous lupus and discoid lupus erythematosus [13, 14, 15, 16

Genetic risk for SLE and the type I IFN pathway

An important development over the past 5 years has been the identification of genes that associate with SLE and that map into the type I IFN pathway. An initial candidate gene study by Sigurdsson et al. [36] found an association of the transcription factor interferon regulatory factor 5 (IRF5) with SLE. Further genetic and functional studies identified a single risk haplotype for IRF5 characterized by three functional alleles [37, 38•]. The non-risk haplotypes for IRF5 contain a splice site

Single gene lupus-related disorders of the type I IFN pathway

In addition to studying sporadic SLE, it is worthwhile to examine highly penetrant phenotypes with a clinical presentation that is similar to or shared with SLE. These single-gene, or Mendelian, disorders can offer insight into therapeutic targets for SLE. However, they may not be representative of sporadic SLE.

Recent data on rare single gene variants that lead to either SLE or SLE-like disease further support the idea that inappropriate activation of type I IFN is on the causal pathway for SLE

Type I IFN as a therapeutic target in SLE

Developing novel therapeutics for SLE is particularly important because there are so few effective therapies for SLE available today. The FDA recently approved Belimumab, a human monoclonal antibody that inhibits the B-lymphocyte stimulator (BLyS), for use in non-renal SLE, and it is the first SLE therapeutic approved in over five decades. The relatively modest clinical benefits reported in the pivotal trials for Belimumab suggest the optimal patient subset for this drug has not yet been

Conclusions

In aggregate, the evidence supporting the direct involvement of type I IFNs in SLE pathogenesis is quite compelling: (a) IFN-α immunotherapy can induce lupus [98]; (b) circulating immune complexes from SLE blood can initiate type I IFN production; (c) there are prominent IFN regulated gene and cytokine signatures in SLE; (d) polymorphisms in IFN pathway genes are associated with SLE in GWAS studies; and (e) several single gene disorders cause increased type I IFN and SLE or SLE-like symptoms.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We would like to thank A. Manoharan and H. Sun for their invaluable assistance in curating the published SLE literature for genetic associations.

References (98)

  • R.R. Graham et al.

    Specific combinations of HLA-DR2 and DR3 class II haplotypes contribute graded risk for disease susceptibility and autoantibodies in human SLE

    Eur J Hum Genet

    (2007)
  • S. Bernatsky et al.

    Mortality in systemic lupus erythematosus

    Arthritis Rheum

    (2006)
  • L.C. Platanias

    Mechanisms of type-I-and type-II-interferon-mediated signalling

    Nat Rev Immunol

    (2005)
  • P. Blanco et al.

    Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus

    Science

    (2001)
  • S. Hervas-Stubbs et al.

    Direct effects of type I interferons on cells of the immune system

    Clin Cancer Res

    (2011)
  • E.C. Baechler et al.

    Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus

    Proc Natl Acad Sci USA

    (2003)
  • L. Bennett et al.

    Interferon and granulopoiesis signatures in systemic lupus erythematosus blood

    J Exp Med

    (2003)
  • M.K. Crow et al.

    Microarray analysis of interferon-regulated genes in SLE

    Autoimmunity

    (2003)
  • G.M. Han et al.

    Analysis of gene expression profiles in human systemic lupus erythematosus using oligonucleotide microarray

    Genes Immun

    (2003)
  • E.S. Emamian et al.

    Peripheral blood gene expression profiling in Sjögren's syndrome

    Genes Immun

    (2009)
  • S.A. Greenberg et al.

    Interferon-alpha/beta-mediated innate immune mechanisms in dermatomyositis

    Ann Neurol

    (2005)
  • E.C. Baechler et al.

    An interferon signature in the peripheral blood of dermatomyositis patients is associated with disease activity

    Mol Med

    (2007)
  • I. Braunstein et al.

    The IFN-regulated gene signature is elevated in SCLE and DLE and correlates with CLASI score

    Br J Dermatol

    (2012)
  • E.C. Baechler et al.

    Gene expression profiling in human autoimmunity

    Immunol Rev

    (2006)
  • K.M. Irvine et al.

    Peripheral blood monocyte gene expression profile clinically stratifies patients with recent-onset type 1 diabetes

    Diabetes

    (2012)
  • S.R. Ytterberg et al.

    Serum interferon levels in patients with systemic lupus erythematosus

    Arthritis Rheum

    (1982)
  • H. Vallin et al.

    Patients with systemic lupus erythematosus (SLE) have a circulating inducer of interferon-alpha production acting on leucocytes resembling immature dendritic cells

    Clin Exp Immunol

    (1999)
  • L. Rönnblom et al.

    A pivotal role for the natural interferon-alpha producing cells (plasmacytoid dendritic cells) in the pathogenesis of lupus

    J Exp Med

    (2001)
  • S.R. Christensen et al.

    Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus

    Immunity

    (2006)
  • E.A. Leadbetter et al.

    Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors

    Nature

    (2002)
  • M.W. Boule et al.

    Toll-like receptor 9-dependent and -independent dendritic cell activation by chromatin-immunoglobulin G complexes

    J Exp Med

    (2004)
  • C. Landolt-Marticorena et al.

    Lack of association between the interferon-alpha signature and longitudinal changes in disease activity in systemic lupus erythematosus

    Ann Rheum Dis

    (2009)
  • M. Petri et al.

    Longitudinal expression of type I interferon responsive genes in systemic lupus erythematosus

    Lupus

    (2009)
  • K.A. Kirou et al.

    Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease

    Arthritis Rheum

    (2005)
  • J.W. Bauer et al.

    Interferon-alpha regulated chemokines as biomarkers of systemic lupus erythematosus disease activity: a validation study

    Arthritis Rheum

    (2009)
  • J.W. Bauer et al.

    Elevated serum levels of interferon-regulated chemokines are biomarkers for active human systemic lupus erythematosus

    PLoS Med

    (2006)
  • A.M. Morimoto et al.

    Association of endogenous anti-interferon-alpha autoantibodies with decreased interferon-pathway and disease activity in patients with systemic lupus erythematosus

    Arthritis Rheum

    (2011)
  • S. Panem et al.

    Antibodies to alpha-interferon in a patient with systemic lupus erythematosus

    J Immunol

    (1982)
  • W.L. Sibbitt et al.

    Relationship between circulating interferon and anti-interferon antibodies and impaired natural killer cell activity in systemic lupus erythematosus

    Arthritis Rheum

    (1985)
  • P. von Wussow et al.

    Presence of interferon and anti-interferon in patients with systemic lupus erythematosus

    Rheumatol Int

    (1988)
  • M. Slavikova et al.

    Incidence of autoantibodies against type I and type II interferons in a cohort of systemic lupus erythematosus patients in Slovakia

    J Interferon Cytokine Res

    (2003)
  • S. Sigurdsson et al.

    Polymorphisms in the tyrosine kinase 2 and interferon regulatory factor 5 genes are associated with systemic lupus erythematosus

    Am J Hum Genet

    (2005)
  • R.R. Graham et al.

    A common haplotype of interferon regulatory factor 5 (IRF5) regulates splicing and expression and is associated with increased risk of systemic lupus erythematosus

    Nat Genet

    (2006)
  • R.R. Graham et al.

    Three functional variants of IFN regulatory factor 5 (IRF5) define risk and protective haplotypes for human lupus

    Proc Natl Acad Sci USA

    (2007)
  • C. Richez et al.

    IFN regulatory factor 5 is required for disease development in the Fc-gamma-RIIB−/− Yaa and Fc-gamma-RIIB−/− mouse models of systemic lupus erythematosus

    J Immunol

    (2010)
  • T.B. Niewold et al.

    IRF5 haplotypes demonstrate diverse serological associations which predict serum interferon alpha activity and explain the majority of the genetic association with systemic lupus erythematosus

    Ann Rheum Dis

    (2012)
  • C. Wang et al.

    Genome-wide profiling of target genes for the systemic lupus erythematosus-associated transcription factors IRF5 and STAT4

    Ann Rheum Dis

    (2012)
  • G. Hom et al.

    Association of systemic lupus erythematosus with C8orf13-BLK and ITGAM-ITGAX

    N Engl J Med

    (2008)
  • S.V. Kozyrev et al.

    Functional variants in the B-cell gene BANK1 are associated with systemic lupus erythematosus

    Nat Genet

    (2008)
  • Cited by (99)

    • Interferon Signature Analysis

      2022, Encyclopedia of Infection and Immunity
    • Systemic Lupus Erythematosus

      2022, Comprehensive Pharmacology
    • What can we learn from DNA methylation studies in lupus?

      2022, Clinical Immunology
      Citation Excerpt :

      This complement component is responsible of inhibiting IFNα production by innate immune cells [49]. Not only monogenic forms, but also susceptibility alleles have also been described in Type I interferon pathway (reviewed in [47]), linking once again this pathway with the development of the disease. In the case of DNA methylation, the hypomethylated profile on interferon-signaling pathway is also relevant in neutrophils and low-density granulocytes [50], CD19+ B cells and CD14+ monocytes [51], CD8+ T cells [52] and memory, regulatory and naïve CD4+ T cells [51,53,54].

    • Genes and genetics in human SLE

      2020, Systemic Lupus Erythematosus: Basic, Applied and Clinical Aspects
    • Lupus, DNA Methylation, and Air Pollution: A Malicious Triad

      2022, International Journal of Environmental Research and Public Health
    View all citing articles on Scopus
    View full text