Cytokine xxx (2017) xxx–xxx
Contents lists available at ScienceDirect
Cytokine journal homepage: www.journals.elsevier.com/cytokine
Review article
Interferon regulatory factor signaling in autoimmune disease Bharati Matta 1, Su Song 1, Dan Li 1, Betsy J. Barnes ⇑ Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Manhasset, NY 11030, United States
a r t i c l e
i n f o
Article history: Received 3 February 2017 Accepted 6 February 2017 Available online xxxx Keywords: Interferon regulatory factors Autoimmune disease Cell signaling Pathogenesis
a b s t r a c t Interferon regulatory factors (IRFs) play critical roles in pathogen-induced innate immune responses and the subsequent induction of adaptive immune response. Dysregulation of IRF signaling is therefore thought to contribute to autoimmune disease pathogenesis. Indeed, numerous murine in vivo studies have documented protection from or enhanced susceptibility to particular autoimmune diseases in Irfdeficient mice. What has been lacking, however, is replication of these in vivo observations in primary immune cells from patients with autoimmune disease. These types of studies are essential as the majority of in vivo data support a protective role for IRFs in Irf-deficient mice, yet IRFs are often found to be overexpressed in patient immune cells. A significant body of work is beginning to emerge from both of these areas of study – mouse and human. Ó 2017 Elsevier Ltd. All rights reserved.
Contents 1. 2.
3.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . IRF genetic association with human autoimmune diseases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. IRF1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. IRF2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. IRF3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4. IRF4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.5. IRF5 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.6. IRF7 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.7. IRF8 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Dysregulation of IRF expression and/or signaling in human autoimmune diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. Multiple sclerosis (MS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. Sjögren ’s syndrome (SS). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.3. Rheumatoid arthritis (RA). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.4. Behcet’s disease (BD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.5. Inflammatory bowel disease (IBD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.6. Type 1 diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.7. Systemic lupus erythematosus (SLE) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.8. Irf loss-of-function studies in murine models of autoimmune disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.9. Experimental autoimmune encephalomyelitis (EAE) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.9.1. Arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.9.2. IBD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.9.3. Autoimmune diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.9.4. Lupus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00
⇑ Corresponding author at: The Feinstein Institute for Medical Research, 350 Community Drive, Rm. 3238, Manhasset, NY 11030, United States. 1
E-mail address:
[email protected] (B.J. Barnes). These authors contributed equally to the work.
http://dx.doi.org/10.1016/j.cyto.2017.02.006 1043-4666/Ó 2017 Elsevier Ltd. All rights reserved.
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
2
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
4.
Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1. Introduction Interferon regulatory factors (IRFs) are a family of transcription factors that were first identified as regulators of virus-induced type I interferon (IFNA and B) gene expression [1]. With the later discovery of other IFN families (type II and III), most of the IRFs, with the exception of IRF6, have been implicated in their regulation [2]. Subsequent findings revealed that IRFs play important roles in the regulation of both innate and adaptive immune responses. In particular, IRFs have been shown to be involved in the activation and differentiation of distinct immune cell populations [3,4]. To date, ten IRFs have been discovered (IRF1-10) in vertebrates; however, some are rendered inactive or eliminated in mice and humans, such as IRF10 [5]. A number of structural features within IRFs are well conserved. All IRF family members share a highly conserved N-terminal DNA-binding domain of approximately 120 amino acids, which binds to the core recognition sequence, GAAANNGAAAG/CT/C, termed the IFN stimulated response element (ISRE) [6]. With the exception of IRF1 and IRF2 that contain a PEST (proline-, glutamic acid-, serine-, and threonine-rich) domain for protein-protein interactions, all other family members contain a non-homologous C-terminal IRF association domain (IAD) for interaction with other family members and/or protein interacting partners [7,8]. In order to initiate signaling through the IRFs, many of them need to be ‘‘activated” by a signal that leads to post-translational modification, followed by altered cellular localization. The types of modifications shown to alter IRF activation/function include phosphorylation, acetylation, ubiquitination, and sumoylation [9]. The signaling cascades that lead to IRF activation are generally downstream of pathogen recognition receptors (PRRs). Identification of PRRs that lead to IFN gene expression has been the subject of intense research in the past decade. Notably, many of the cellular sensors shown to be responsible for pathogen recognition require IRF activation and downstream signaling. Cellular sensors shown to be involved in IRF signaling are: membrane bound Toll-like receptors (TLR), cytoplasmic RIG-like receptors (RLR), nucleotide binding oligomerization domain (NOD)-like receptors, and a disparate family of cytoplasmic and possibly nuclear DNA sensors [1,10–12]. Activation of IRFs through many of these pathways lead to the expression of IFNs, as well as other inflammation-associated cytokines [13]. The most studied pathway(s) that leads to IRF activation is through the TLRs. TLR signaling can be divided into two pathways: one that is dependent on the adapter protein MyD88 (myeloid differentiation primary response), and the other that is MYD88independent and requires the adapter protein TRIF (Toll/IL-1 receptor domain-containing adaptor inducing IFN-B). Depending on the IRF, one or both pathways may lead to activation, resulting in its binding to the promoters of target genes and modulation of immune responses [1,3,10]. The steps leading to IRF activation and function can be broadly summarized into five major stages: (1) signal, (2) post-translational modification, (3) dimer formation, (4) nuclear translocation, (5) regulation of target gene expression. With ongoing advances in the field of genome-wide association studies (GWAS), genetic variations in IRFs have been identified and associated with numerous autoimmune diseases. These genetic associations have opened up an expansive field of research focused on determining the role of IRFs in autoimmune disease pathogenesis. Autoimmune diseases are characterized by an attack on one’s
‘‘self”. The attack can be either specific to a particular organ, as is the case in inflammatory bowel disease (IBD), or systemic, as seen in systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and Sjögren’s Syndrome (SS). A number of genetic variants or single nucleotide polymorphisms (SNPs) within IRF genes have been detected at higher rates in patients with autoimmune diseases than matched healthy controls that enable an association with risk or protection from disease. Although this review will not cover the in-depth details from GWAS since a number of excellent reviews already exist [14–18], findings from GWAS are what initiated this new field of study - IRFs in autoimmunity. We instead provide a concise summary of in vivo and in vitro data that implicate dysregulated IRF signaling in autoimmune disease pathogenesis with a specific focus on the common and/or distinct pathways leading to and from the IRFs. Given the critical role of IRFs in IFN gene regulation, it was first postulated that genetic variants would contribute to elevated type I IFN expression that is now detected in multiple autoimmune diseases [19]. Based on emerging data, however, we propose that IRF function (or dysfunction) in autoimmune diseases is more robust than just as regulators of the IFNs. 2. IRF genetic association with human autoimmune diseases Below we summarize findings from GWAS that identified genetic variation within the IRFs that associate with either susceptibility to or protection from a particular autoimmune disease. Only IRFs in which genetic variation has been associated with autoimmune diseases are detailed here. It is this body of work that initiated a new field of research focused on understanding the pathogenic role of IRFs in autoimmunity. 2.1. IRF1 IRF1 regulates the transcription of genes that play essential roles in viral infection, tumor immune surveillance, proinflammatory injury, and immunity system development [20]. With regard to GWAS, little is currently known of IRF1 polymorphisms and risk of autoimmune diseases. Initial data found differences in IRF1 polymorphisms between healthy controls and patients with juvenile idiopathic arthritis (JIA) [21] and Behcet’s disease (BD) [22]. In BD, IRF1 polymorphisms associated with risk in women and patients with thrombosis [22]. In JIA, however, a subsequent study examining a larger cohort of patients with different control subjects contradicted the early associations [23]. IRF1 polymorphisms have been identified in multiple sclerosis (MS), yet more replicative studies are needed [24]. In RA, SNPs within the introns and untranslated regions (UTRs) of IRF1 were found to associate with risk in black South African patients [25]. Copy number variations were also identified in IRF1 that associated with RA [26]. Continued replication of GWAS in multiple ethnicities and cohorts will be required to confirm associations of IRF1 genetic variants in JIA, BD, MS and RA. 2.2. IRF2 IRF2 negatively regulates type I IFN responses and plays a role in the induction of Th1 differentiation [8,27]. While not well replicated, association of IRF2 genetic variants with susceptibility to SLE has been shown, and the risk haplotype was suggested to be associated with transcriptional activation of IRF2 [28,29].
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
2.3. IRF3 IRF3 is constitutively expressed in all cell types and is primarily known for its regulation of type I IFNs in response to pathogens (reviewed in [2]). Similar to IRF1, few replicative association studies have been performed to link IRF3 genetic variants to autoimmune diseases. The most studied has been IRF3 polymorphisms in SLE, yet little agreement has been reached [8,9,30]. In a Mexican cohort, the IRF3 SNP rs2304206 associated with increased susceptibility to SLE and elevated serum type I IFN levels in patients positive for dsDNA autoantibodies [31]; however, this finding has yet to be replicated. Surprisingly, the IRF3 rs2304204 GG and AG genotypes were found to confer decreased risk of SLE [31]. SNPs 925A/ G and 776C/T in the promoter region of IRF3 showed significant association with SLE risk while the GG genotype of 925A/G was protective [30]. Additional replicative studies will be required to confirm associations. 2.4. IRF4 Distinct from other IRF family members, IRF4 expression is restricted to immune cells, and can either function as a transcriptional activator or repressor depending on its interacting partner (s) [32]. IRF4 is induced upon the activation of T and B cells. In CD4+ T helper cells (TH), IRF4 plays an essential role in the regulation of IL21 production, whereas in B cells it controls class switch recombination and plasma cell differentiation [33]. The IRF4 SNP rs9378815 has been reported to associate with risk of RA in European and Asian ancestries [34,35]. IRF4 was also identified as a common susceptibility locus for systemic sclerosis (SSc) [35]. No association was found between IRF4 SNP rs872071 and SLE susceptibility a Han Chinese [36]. IRF4 continues to be examined for genetic variation associated with autoimmune diseases. 2.5. IRF5 IRF5 has been demonstrated to play a key role in modulating innate and adaptive immune responses in numerous cell types, including dendritic cells, monocytes/macrophages, and B cells. IRF5 is regarded as the ‘‘master regulator of proinflammatory cytokines” [11]. In addition to type I IFN production, IRF5 is responsible for upregulating expression of IL6, IL12b, IL17, IL23, IFNb, IP10, MCP1, and RANTES [37,38]. Findings from GWAS have shown that IRF5 genetic variants impact a broad range of autoimmune diseases. In the case of SLE, numerous GWAS have supported associations between IRF5 SNPs and functional risk variants in the IRF5 gene across multiple ancestral backgrounds [39–45]. IRF5 risk variants have also been identified in other autoimmune diseases such as RA [46,47], SSc [40,48], MS [49,50], and IBD [51] suggesting a common pathogenic role for IRF5 in autoimmune diseases. Of note, two SNPs - rs10954213 in the 30 UTR and rs2004640 in the 50 -UTR are shared amongst the most commonly associated with risk of any autoimmune diseases. Importantly, however, not all IRF5 SNPs are associated with risk. Some SNPs have been shown to be protective. In RA, the T allele of SNP rs2280714, C allele of rs729302 and G allele of rs2004640 associated with protection [52]. 2.6. IRF7 IRF7 expression is not restricted to immune cells but nor is it ubiquitously expressed. It is best known as a master regulator of type I IFN gene expression and IFN-dependent innate immune responses [53]. Recent findings from GWAS identified the IRF7 risk SNP rs1131665 in SLE patients from Asian, European American, and African American populations [54,55]. Similar to IRF5, genetic variants in IRF7 are associated with increased levels of IFNs in SLE
3
patients [1]. The IRF7 variant rs1131665 is associated with SLE [54]. Genetic variants of IRF7 may act as risk factors for SLE due SNPs in the IRF7/PHRF1 locus [56]. Additionally, IRF7 has been implicated in the pathogenesis of metabolic autoimmune disorders such as diabetes [57]. 2.7. IRF8 IRF8 is a key transcription factor for myeloid cell differentiation [58]. With regard to autoimmune diseases, two genetic variants in the IRF8 gene have been associated with BD in Han Chinese [59]. Increased IRF8 mRNA expression and IFNc production, together with a concomitant decrease in IL10 production, is described in individuals carrying the rs11642873/AA and rs17445836/GG genotype [59]. Two independent studies identified additional SNPs in IRF8 that associated with risk of SLE [60,61]. SNP rs17445836 was also found to associate with susceptibility to MS and SLE. In SLE, the risk SNP associated with anti-dsDNA antibodies and serum IFNa levels in patients of both African-American and European ancestry [62]. 3. Dysregulation of IRF expression and/or signaling in human autoimmune diseases In this section, we report on predominant findings of IRF function and signaling in some of the more common human autoimmune diseases. IRFs that have not yet been functionally characterized in these diseases will be excluded from the discussion. Figs. 1 and 2 depict a summary of findings from this section. 3.1. Multiple sclerosis (MS) MS is an inflammatory demyelinating disease of the central nervous system (CNS), which is characterized by an autoimmune inflammatory reaction against CNS myelin. IRF1 signaling in microglial cells has been shown to be involved in the pathogenesis of MS [24,63]. IRF1 SNPs have been associated with progressive MS [24,63] and IRF1-regulated genes, such as MHC class I (MHCI), TNF receptor (TNFR) and caspase 1, have been shown to be elevated in MS patients. In glial cells, IRF1 was found to cooperate with NFkB for the regulation of MHCI and inducible nitric oxide synthase (iNOS) expression leading to cytokine synergism [24,63–65]. Glial cells were found to be the primary CNS cell with IRF1 expression in demyelinating lesions of MS [66]. IRF1 has been also shown to control the expression of two pro-apoptotic genes, caspase 1 and double-stranded RNA-dependent protein kinase (PKR) that play important roles in the process of inflammatory demyelination. Later studies implicated IRF1 and signal transducer and transcription activator 1 (STAT1) in IFNc-induced oligodendrocyte progenitor cell death [66]. IRF8 is the only other family member that has been examined in the context of MS. Similar to IRF1, IRF8 was shown to mediate IFNc signaling leading to oligodendroglial progenitor cell apoptosis [67]. 3.2. Sjögren ’s syndrome (SS) SS is a systemic autoimmune disease involving multiple organs. It is characterized by extensive dryness, extreme fatigue, chronic pain, neuropathies and lymphomas. It is called primary SS (pSS) when it occurs alone or secondary SS when it is in association with other autoimmune diseases like SLE and RA. SS is further characterized by infiltration of TH cells and a TH1/TH2 imbalance has been shown to play a role in SS pathogenesis [67–74]. Like SLE, an IFN gene signature has been shown to be associated with SS and the strength of the IFN signature positively correlated with disease
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
4
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
severity. IRF1 plays an important role in TH differentiation and is known to regulate gene expression of factors involved in TH1 differentiation. IRF1, along with IFNc, is shown to induce and expand TH1 responses [8,75]. A higher expression of IRF1 was detected in peripheral blood of patients with primary SS along with more IRF1 positive cells in epithelial islands, lymphocytes, and ductal epithelial cells of the parotid glands compared to healthy control subjects [74]. Viral infections such as Epstein Barr Virus and chronic hepatitis infections have been known to be the initial inducers of pSS and initiate the production of IFNs. Plasmacytoid dendritic cells (PDCs) are considered to be a primary source of IFNa in pSS. IRF1 may also enhance and prolong the immune reaction leading to autoimmunity. A recent report associated B cell activating factor (BAFF) with pSS and suggested that the expression of BAFF is regulated by IRFs. BAFF is a cytokine required for the proper development and selection of B cells. In humans, BAFF is induced by type I IFNs [76]. In patients with pSS, BAFF expression in neutrophils positively correlated with type I IFN activity. BAFF is induced by type I IFN signaling via the transcription factors IRF1 and IRF2 whereas IRF4 and IRF8 were shown to negatively regulate BAFF expression [76]. Interestingly, SS patients carrying 4 repeats of the IRF5 CGGGG promoter insertion/deletion (indel) were found to have a high level of IRF5 mRNA in PBMCs and salivary gland epithelial cells after in vitro viral infection. SS patients with high levels of IRF5 mRNA also had high levels of mRNA for ISGs in PBMCs [77]. The mechanism(s) or pathway leading to upregulated IRF5 and ISG expression in SS is not known. 3.3. Rheumatoid arthritis (RA) RA is a systemic autoimmune disease that is characterized by expansion of synovial tissue, chronic inflammation and progressive destruction of a joint. Several immune cell subsets, including macrophages, monocytes, neutrophils, dendritic cells, natural killer cells, T cells, and B cells have been shown to play a role in RA pathogenesis [78]. TNFa, IL1b and IL6 are the major proinflammatory cytokines that play a prominent role in RA progression. Fibroblast-like synoviocytes (FLS) are the major constituent of synovial hyperplasia and the FLS-dependent effector molecules are implicated as important mediators of RA [78]. FLS play a critical role in invading cartilage and bone tissue [78]. IRF3 has been shown to play a major role in the regulation of ISGs in RA [79]. High levels of phosphorylated IRF3 were detected in RA synovial tissue. When FLS were transfected with siRNAs targeting IRF3 and then stimulated with poly (I-C) in culture, IRF3 was shown to regulate ISRE promoter activity as well as IFNb, IRF5, IRF7, RANTES, IFN-inducible protein-10, MCP-1, and MIP1a gene expression [79]. Moreover, IRF3 knockdown blocked expression of metalloproteinase 3 (MMP3), MMP9, IL6, and IL8. A significant decrease in AP-1 binding of activated c-Jun in IRF3 deficient cells was found. This study further demonstrated that phosphorylation of IRF3 was regulated by IKK-related kinase IKKe [79]. IL18 is another cytokine that plays an important role in the pathogenesis of RA [80]. IL18 promotes T cells to produce TH1 type cytokines and helps in the differentiation of TH1 cells. IL18 also regulates the cytotoxic activity of natural killer cells by upregulating Fas ligand [81–87] and promotes angiogenesis [88–91], all of which contribute to RA pathogenesis. The biologic activity of IL18 is controlled by IL18 binding protein (IL-18BP). There is an IRF1 binding site in the IL18BP promoter [92]. Using fibroblasts isolated from synovium of RA patients who had undergone total joint replacement or synovectomy, an in vitro knockdown system was utilized to analyze the effect of TNFa-induced IL18BP expression. TNFa was shown to induce IRF1 nuclear translocation and regulate IL18BP expression; however, when NFjB or JNK2 signaling pathways were blocked, IRF1 nuclear translocation was reduced.
Further, it was shown that IRF1 forms a complex with NFjB and cJun in the nucleus [93]. With regard to other family members, IRF4 and IRF5 are expressed in synovial fluid and synovium from RA patients. Anticitrullinated protein antibodies (ACPAs) were found to induce IRF4 and IRF5 protein expression. IRF5 siRNA knockdown impaired ACPA activity significantly while ACPAs induced IRF5 activity and led to M1 macrophage polarization [94]. Additional studies will be required to determine the pathological contribution of elevated IRF4 or IRF5 expression in RA pathogenesis. 3.4. Behcet’s disease (BD) BD is a systemic vasculitis disease with unknown etiology. It is characterized by recurrent attacks of oral aphthous ulcers, genital ulcers, and ocular uveitis. Altered T cell responses are associated with BD [22]. While IRF1 and IRF8 genetic variants are associated with BD, no functional studies have been performed yet. 3.5. Inflammatory bowel disease (IBD) There are two main forms of IBD: Crohn’s disease and ulcerative colitis. Both serum and mucosal autoantibodies against intestinal cells and perinuclear anti-neutrophil cytoplasmic antibodies (pANCA) have been detected and found to associate with disease activity [95]. IRF1 expression was initially detected as increased in the nuclei of lamina propria mononuclear cells in colonic specimens from pediatric patients with active Crohn’s disease [96]. Later studies, however, detected lower levels of IRF1 in patients who were undergoing standard medical treatment compared to untreated patients [96]. A study examining intestinal epithelial cells isolated from freshly obtained colonic mucosal biopsies revealed another role for IRF1. They found that intestinal epithelial cells expressed CARD4/NOD1 at both mRNA and protein levels [97]. CARD4/NOD1 transcription was induced by IFNc through IRF1. An IRF1 binding motif was identified within CARD4/NOD1 and found to be necessary for the IFNc effect [97]. IL7 plays a critical role in the regulation of mucosal immunity and has pleiotropic functions in the intestinal immune system. Intestinal epithelial cells produce IL7, which induces the proliferation of lamina propria lymphocytes and intraepithelial lymphocytes [98,99]. IL7 also increases the production of cytokines by lamina propria lymphocytes in humans [100]. IRF1 and IRF2 are expressed in normal human colonic epithelial cells and upregulate IFNc-induced IL7 [101]. Elevated TBK1, NAP1, IRF3 and IRF7 mRNA levels were found in biopsied mucosal specimens collected during colonoscopy and human colonic epithelial lines from inflamed colonic mucosa as compared to normal mucosa [102]. Rebamipide is used to treat gastric ulcers and has been used in a case study to treat IBD. Rebamipide reduced LPS (TLR4)/dsRNA (TLR3)-induced expression of IRF3, IRF7 and TBK1 in human colonic epithelial cells [102]. IRF4, IL17A and IL22 expression levels are increased in colonic biopsy specimens of IBD patients as compared to control patients. Moreover, their expression was upregulated in sections with inflamed mucosa compared to sections without inflammation. This is the first evidence to suggest that there might be a correlation between IRF4 levels and expression of Th17 cytokines [103]. 3.6. Type 1 diabetes Type 1 diabetes is characterized by the autoimmune destruction of insulin producing b cells of the islets of Langerhans in the pancreas. Cytotoxic T lymphocytes are the main effector cells involved in the death of b cells [104]. A substantial amount of work
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
5
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
IFNy
IRF1
IFNy, BAFF
IRF8
Glial cells
IRF1
IRF2
Poly(I-C)
IRF3
TH cells
IRF5
IRF1
Synoviocytes
Synovium
IRF1
IRF2
Intestinal Epithelial Cells
Neutrophils
MHC I TNFαR Caspase 1 INOS PKR STAT1
IFNy
ACPAs
TNF
Type I IFN
Inflammatory Demyelination and cell death
Regulation of TH responses
MS
SS
IFBβ IRF5 IRF7 RANTES IP10 MCP1 MIP1a Caspase 1 MMP3 MMP9 IL-18BP IL6 IL8
M1
Macrophage Polarization
CARD4 NOD1 IL7
Inflammation of GI tract
Synovial Hyperplasia
RA
IBD
Fig. 1. Implications of IRF signaling in human autoimmune diseases. Schematic summarizing the effect(s) of IRF signaling in specific cell types, in response to specific stimuli, and the specific autoimmune disease it has been implicated in. Gene expression affected by individual IRFs is shown by matched colored font for each IRF. For example, the IRF1 molecule is shown in blue and gene expression affected by IRF1 is shown in blue. Overlapping functions for the IRFs are written in black font. Diseases affected by the IRFs are shown at the bottom surrounded by green. MS, multiple sclerosis; SS, Sjögren’s syndrome; RA, rheumatoid arthritis; IBD, inflammatory bowel disease. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
has been done studying the importance of IRF signaling in mouse models of diabetes but no work has been published in humans. 3.7. Systemic lupus erythematosus (SLE) SLE is the prototype autoimmune disease where genes regulated by type I IFNs are overexpressed and shown to contribute to disease pathogenesis. SLE is characterized in part by elevated antinuclear antibodies (ANA). 95% of patients possess ANA that recognize ‘‘self” chromatin components including dsDNA, histones, and nucleosome. Each of these components contributes to a break in self-tolerance, resulting in autoimmune responses [105]. Circulating ANAs bind self-nucleic acids and associated proteins exposed on the surface of apoptotic cells, generating self-antigencontaining immune complexes (ICs). These ICs are internalized by PDCs in an FcgRIIB-dependent manner, resulting in TLR activation and type I IFN expression [106–108]. The most studied IRF to date in SLE is IRF5. IRF5, like IRF7, is an IFN-induced gene, thus its expression can be enhanced by IFN production via a positive feedback loop, and subsequently enhance type I IFN gene transcription [109]. We and others showed that IRF5 expression was significantly elevated in PBMCs from SLE patients as compared to healthy donors and in lymphoblastoid cell lines from SLE patients carrying the risk haplotype [39,45,110,111]. Subsequent studies found that elevated IRF5 expression associated with risk SNPs and enhanced type I IFN activity in SLE patients with high anti-RBP or antidsDNA autoantibodies [112]. Using imaging flow cytometry, we later showed that IRF5 was constitutively activated (i.e. nuclear localized) in monocytes from SLE patients and that activation could be replicated in healthy donor monocytes stimulated with SLE serum. Activated IRF5 resulted in elevated IL6, TNFa and IFNa
secretion [113]. Results from this study suggested that SLEinduced IRF5 activation may be TLR-, FccRIIB- and IFNindependent in monocytes [113]. Later studies suggested that IRF5 risk haplotypes may predict specific B cell functioning pathways [114]. Others described a correlation between the IRF5 rs2004640 risk SNP and elevated cytokine expression when monocyte-derived cells were treated with different Nod2 and TLR ligands [115]. Of significant interest, the rs2004640/rs2280714 TT/TT IRF5 disease-risk-carrier cells were shown to have increased IRF5 expression and increased PRR-induced Akt2 activation, glycolysis, pro-inflammatory cytokines, and M1 polarization relative to GG/CC carrier macrophages, supporting additional functions for IRF5 in autoimmune disease pathogenesis [116]. Much of the work done on IRF5 signaling in human SLE has been focused on TLRmediated IRF5 activation. While relevant given its critical role downstream of MyD88-dependent TLR signaling [117], this may be too simplistic of a view since SLE serum contains a number of complex triggers [113]. IRF3 and IRF7 signaling has also been shown to be involved in SLE pathogenesis (Fig. 2). Enhanced IRF3 expression was found in PDCs from SLE patients, which associated with higher IFNa serum levels [31]. Studies on IRF7 in human SLE have expanded in the last few years due to its role in IFN expression. Indeed, genetic variants of IRF7 were shown to be associated with increased serum IFNa levels in SLE patients with anti-Sm and anti-dsDNA autoantibodies [118]. Independent studies found that induction of IRF7 protein expression correlated with recurrent lupus disease activity. It was found that phosphorylation of IRF3 and activation of 4 E-BP1, a translational repressor of IRF7, preceded disease flare, while recurrent disease activity and induction of IRF7 protein expression correlated with induction of the IFN signature [119]. Using an
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
6
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
PBMCs T IRF7 IRF5
B
M
B cells
Mo/Mac
IRF8 IRF5
pDC
IRF5
IRF3 IL6 TNFα IFNy
IRF2, and IRF8 are more limited but many of the studies relate these IRFs back to type I IFN signaling or B cell function. Interestingly, 63% of genes with H4 hyperacetylation in SLE had potential IRF1 binding sites [120,121]. Since there exists a higher preponderance of SLE in women than men, IRF1 becomes a molecule of interest as it is one of the major downstream mediators of prolactin signaling. Prolactin is immune stimulatory and can break B cell tolerance. IRF1 provides a potential nexus of female hormones, inflammation and type I IFN signals [120]. IRF1 not only activates gene expression as a transcription factor but may perpetuate SLE disease by leading to a dysregulated epigenome [122]. Although there is limited data on IRF2 in SLE, elevated transcriptional activity was described using an IRF2 promoter reporter carrying risk polymorphisms [123]. Additionally, SNP rs17445836G was found to be associated with increased IRF8 expression in SLE patient B cells, decreased serum IFN activity in patients with anti-dsDNA antibodies, and decreased ISG expression in PBMCs from antidsDNA-negative patients [124]. 3.8. Irf loss-of-function studies in murine models of autoimmune disease Given the difficulties in obtaining appropriate samples sizes for functional studies in human primary immune cells, knockout mice offer one of the most powerful means for studying gene function in a living animal. Indeed, knockout mice have been widely utilized in the study of autoimmune disease pathogenesis. Here we summarize data from murine models of autoimmune diseases that lack one or more Irf(s) (Table 1).
ISG expression serum IFNα
3.9. Experimental autoimmune encephalomyelitis (EAE)
SLE
Fig. 2. Cell type-specific IRF signaling in human SLE. Schematic summarizing the role of individual IRFs in SLE pathogenesis. The primary downstream effect of all indicated IRFs is on type I IFN expression.
ISRE-Luc reporter assay, SNP rs1131665 in IRF7 was shown to confer elevated IRF7 transactivation ability [54]. Data on IRF1,
EAE is the most commonly used experimental model for MS and mice deficient of Irf1, 3, 4 or 8 are protected from EAE. Levels of protection were distinct and may be due in part to differences in background strains [125–128]. For instance, Irf1/ L/J mice challenged with myelin basic protein (MBP) were found to have a significantly lower incidence of EAE as compared to wild-type (WT) littermates but disease severity was not altered [126]. Decreased iNOS induction and subsequent NO production in macrophages was indicated as a possible mechanism. Two other EAE models in Irf1/ mice of different backgrounds, 129/Sv and C57Bl/6J, were almost completely resistant to myelin oligodendrocyte glycoprotein (MOG)-induced EAE [129,130]. Further mechanistic studies
Table 1 Autoimmune disease outcome in Irf-deficient models. Lupus
Arthritis
EAE
IBD
Autoimmune Diabetes
Irf1/
;auto-Ab ;kidneydisease
;incidence ;severity Delayed onset
"incidence "severity
Completely protected
Irf2/
Naturally develops CLE-like inflammation Association of disease protective ;auto-Ab ;kidney disease ;auto-Ab ;kidney disease ;auto-Ab No effect on kidney disease ;auto-Ab ;kidney disease ;auto-Ab ;kidney disease
NT
MBP-induced:;incidence No effect on severity MOG-induced: Completely protected NT
NT
NT
;incidence ;severity Completely protected
"severity
NT
NT
Completely protected No effect
Completely protected NT
"susceptibility
"severity
NT
NT
NT
Completely protected
NT
NT
NT
NT
NT
NT
/
Irf3
Irf4/ Irf5/ /
Irf7
/
Irf8
Irf9/
No effect Association of disease exacerbation No effect
Irf, interferon regulatory factor; EAE, experimental autoimmune encephalomyelitis; IBD, inflammatory bowel disease; CLE, cutaneous lupus erythematosus; NT, not tested.
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
showed that autoreactive T cells isolated from Irf1/ 129/Sv mice could not induce EAE in WT mice [129,130]. Bone marrow (BM) chimeric C57Bl/6J mice were generated to differentially express Irf1 in the CNS and the immune system to dissect the role of Irf1 in either compartment. In this model, it was found that the susceptibility to EAE was associated with Irf1-mediated immune reconstruction in the periphery, whereas differences in disease severity (clinical score and disease duration) was regulated by Irf1 expression in the CNS [130]. The cause of disparity in phenotypes between Irf1/ strains is likely due to differences in genetic backgrounds, as well as the mechanism of EAE induction. Irf3/ C57Bl/6 mice have significantly reduced incidence and severity of MOG-induced EAE [127], while Irf4/ C57Bl/6 mice [128] and Irf8/ C57Bl/6 mice [125] are completely resistant. The underlying mechanism(s) of protection in Irf3/ and Irf4/ mice seem to be similar and T cell-related. In both models, in vivo TH17 differentiation was impaired and in vitro IL17 production was not induced. Notably, transfer of myelin-reactive Irf3/ CD4+ cells to WT mice failed to induce EAE in TH17-polarised models as did WT cells transferred into Irf3/ recipients [127]. However, transfer of WT CD4+ TH cells into Irf4/ mice resulted in the development of EAE in these previously resistant Irf4/ mice, and contrary to expectations, injection of Irf4/ CD4+ TH cells into Irf4/ and WT mice had no substantial effect on disease course in either recipient mice. A decrease in IL6-induced RORct expression and lack of IL6-mediated downregulation of Foxp3 were likely the reasons for failure of TH17 differentiation in Irf4/ TH cells [128]. IRF8 in T cells is also capable of regulating TH17 differentiation and Irf8/ mice fail to generate TH1, TH17 and Treg cells after MOG injection. However, unlike Irf3 and Irf4, Irf8/ T cells do not have a consequential role in EAE. Mice with monocyte- and macrophage-specific Irf8 deletion, but not T cell-specific Irf8 deletion, remained resistant to EAE, indicating that Irf8 in the monocyte and macrophage is primarily responsible for causing EAE [125]. Last, Irf7 expression was elevated in the CNS of MOGinduced EAE; yet, knocking out Irf7 in C57Bl/6 mice resulted in more severe EAE. Irf7/ C57Bl/6 mice had significantly higher numbers of T cells in the CNS, and both IL17 and IFNc were expressed at higher levels in the CNS [131]. 3.9.1. Arthritis A commonly used model of arthritis in mice is the collageninduced arthritis (CIA) model. In CIA, IRF1 appears to be involved in both the induction and effector phases of disease. Irf1/ DBA/1 mice challenged with collagen type II (dCII) had reduced incidence, severity and delayed onset of disease [126]. IFNc production by lymph nodes cells and proinflammatory cytokine production by peritoneal macrophages in response to dCII were defective in Irf1/ mice. The role of IRF1 in the effector phase was studied by adoptive transfer. Injection of a CII-specific T cell line together with sera from arthritic mice into naïve DBA/1 mice showed that disease was suppressed in mice lacking Irf1. The suppression of CIA in Irf1/ mice was even more dramatic than that reported in CD4- or CD8-deficient mice or in TNFreceptor p55-deficient mice [126,132]. Chikungunya virus (CHIKV) infection is another model of murine arthritis that exhibits similar disease manifestations as those seen in humans [133]. While deficiency of Irf3 alone had no significant effect on disease progression, Irf7/ or double-deficient Irf3/ Irf7/ mice significantly increased susceptibility to CHIKV that resembled human disease [133,134]. Thus, in the CHICKV infection model, Irf3 and Irf7 appear functionally redundant. Irf7 was studied in another model of murine arthritis, passive K/BxN serum transfer arthritis [135,136]. Similar to findings from the CHIKV model, Irf7/ C57Bl/6 mice with K/BxN serum transfer had increased clinical severity of arthritis, with augmented sys-
7
temic and local proinflammatory cytokine profiles, indicating an overall counter-regulatory role of Irf7 in the later stages of disease [137]. IFNb and poly (I-C) treatment have been also shown to inhibit inflammatory arthritis in mice [138–140]. Both treatments resulted in decreased K/BxN arthritis in Irf7/ mice suggesting that Irf7 plays a counter-regulatory role in this model, possibly through its regulation of IFN production and cytokine gene expression. In the B6-mev/mev(viable mothaeten) model of murine arthritis, Irf5 + IL23 + macrophages were significantly increased, suggesting a possible link between Irf5 and arthritis [141]. However, Irf5/ C57Bl6 mice with CIA displayed similar severity of paw swelling, as well as serum IgM, total IgG, IgG2a and anti-CII autoantibody levels as WT mice [27]. Thus, at least under these experimental conditions, Irf5 is not required for clinical manifestation of CIA-induced arthritis. While the effects of Irf4 deficiency in mouse models of inflammatory arthritis have not yet been investigated, mice deficient in Irf4-binding protein (IBP/) rapidly develop rheumatoid arthritis-like joint disease and large-vessel vasculitis [142]. IBP deficiency leads to the aberrant production of IL17 and IL21, which are regulated by Irf4. Absence of IBP leads to the enhanced targeting of the IL17 and IL21 promoters by Irf4, and the presence of IBP prevents Irf4 from binding to and transactivating the IL21 promoter. Irf4 is likely a pro-disease factor. 3.9.2. IBD To date, only dextran sulfate sodium (DSS)- and oxazoloneinduced colitis has been studied in Irf-deficient mice. Contrary to findings in other autoimmune disease models, lack of Irf1 is not protective in DSS colitis [143]. Instead, Irf1/ C57Bl/6J mice had increased distortion of crypt architecture, higher incidence of colonic dysplasia, and decreased expression of caspases and genes involved in antigen presentation. Similar to Irf1/ mice, loss of Irf3 exacerbated DSS-induced colitis [144]. The weight of Irf3/ C57Bl/6 mice continued to decrease even after DSS was replaced with water, indicating a defect in recovery from colitis. Colonic expression of Tslp and Il33 genes, which are critical for recovery from colitis, was significantly decreased in Irf3/ mice before and after DSS treatment, suggesting that deficiency in their expression might account for disease pathogenesis [143]. Even though Irf5 is activated by TLR and RLR pathways, which contribute to the suppression of colitis [145,146], Irf5 was not essential for DSS-induced colitis. Knocking out Irf5 neither exacerbated nor improved DSS-induced colitis [144]. IL6 has an important role in intestinal inflammation as neutralizing anti-IL6R antibodies lead to suppression of established intestinal inflammation in murine models of IBD [147–149]. Irf4/ C57Bl/6J mice were completely protected from oxazolone-induced colitis, and only developed mild colitis after TNBS (oxazolone, trinitrobenzene sulfonic acid) treatment, whereas WT mice had severe colitis [150]. The striking phenotype seen in Irf4/ mice with either model of induced colitis was decreased mucosal IL6 production and suppression of anti-apoptotic IL6 signaling in Irf4/ T cells. Administration of recombinant IL6 or hyper–IL6 abrogated the protective effect of Irf4 deficiency in both TNBS and oxazolone colitis. Furthermore, mucosal IL6 production is a key regulator of T cell resistance against apoptosis in Crohn’s disease and experimental colitis [151]. Lastly, immunodeficient RAG mice reconstituted with CD45RBhiCD4+ T cells from Irf4/ mice, but not from WT mice, had less IL6 production and were resistant to colitis. This suggests that the protective effect of Irf4 deficiency in experimental colitis is mediated via T cell-derived IL6 [150]. 3.9.3. Autoimmune diabetes Non-obese diabetic (NOD) mice are a widely used animal model of autoimmune diabetes. Nakazawa et al. bred Irf1/ C57BL/6J
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
8
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
IRF3
IRF1 IRF2
IRF9
IRF4
IRF7 IRF8
IRF5
Mo/Mac T cells
DC Neutrophil B cells
Fig. 3. Cell-type specific roles for IRFs in murine lupus. A schematic showing which cell types IRF functions have been implicated in from Irf-deficient models of murine lupus.
mice onto the NOD background to examine its role in autoimmune diabetes development [152]. Loss of Irf1 completely blocked the development of insulitis and diabetes. It was found that the percentage of CD4+ and Mac1+ spleen cells were significantly increased, whereas CD3+, CD8+ and B220+ cells decreased in Irf1/ NOD mice. Furthermore, spleen cell proliferation in response to Con A or murine GAD65 peptide, a major autoantigen of the pancreatic b cell, significantly increased, and the IFNc/IL10 ratio in culture supernatants significantly decreased from Irf1/ mice. IFNc/TNFa synergism has been suggested to be responsible for autoimmune diabetes in vivo as well as b cell apoptosis in vitro [153]. IRF1, activated by STAT1 in pancreatic b cells, plays a pivotal role in this synergism. Thus, it is plausible that lack of Irf1 in NOD mice blocks the Stat1/Irf1 pathway, hence pancreatic b cell apoptosis. Irf4/ NOD mice were completely protected from the development of diabetes. Interestingly, Irf4+/ NOD mice were also protected. Insulin autoantibody (IAA) expression was absent in both Irf4+/ and Irf4/ NOD mice [154]. Disease resistance in Irf4-deficient mice may be due to the attenuation of effector T cell function in NOD mice. In an adoptive transfer study, WT T cells, but not pre-diabetic Irf4/ T cells, were able to induce diabetes in NOD/severe combined immunodeficiency (SCID) mice, while Irf4+/ T cells were only able to induce mild diabetes. Further stratification of CD4+ and CD8+ T cells revealed that diabetes is suppressed in both Irf4/ CD4/Irf4+/+ CD8 recipient mice and Irf4+/+ CD4/Irf4/ CD8 recipient mice [154]. These results indicate that Irf4 plays a crucial role in both CD4+ T cells and CD8+ T cells in autoimmune diabetes. 3.9.4. Lupus One of the first murine datasets that implicated Irf5 and Irf7 in lupus disease pathogenesis was stimulation of PDCs from Irf5/ and Irf7/ mice with purified human SLE ICs and measurement of type I IFN secretion; cells from both knockouts showed significantly reduced IFNa and b [155]. From then on, multiple murine models of lupus have been utilized to study the in vivo role of Irfs in disease pathogenesis. The majority of these models are genetic models of lupus onset, however, pristane-induced lupus is a commonly used model to study IFN-associated pathologies [156]. A number of Irf-deficient mice have been bred onto lupus prone backgrounds. Even though different Irf knockouts have been bred onto mice with different genetic backgrounds, including B6lpr, NZB, Faslpr/lpr, MRL/lpr, FccRIIB-/-Yaa (RII.Yaa) and FccRIIB-/-, most show protection against lupus [157–162]. Among the genetically modified models, decreased autoantibody production and ameliorated kidney disease was seen in
Irf1-, Irf4-, Irf5- and Irf8-deficient lupus models. One common mechanism among these models lies in their ability to regulate T cells. Multiple subpopulations of splenic T cells, such as CD4CD8-CD44+ T cells (decreased) and CD4 + CD25+ T cells (increased), are skewed in Irf1/ mice [157]. Lack of Irf4 reduced TH1 and TH17 effector T cells [159]; age-associated splenomegaly and expansion of T cells typical of NZB mice were largely abrogated in Irf8/ NZB mice, accompanied by a significant decrease in CD4+ T-cell activation [162]. In Irf5/ MRL/lpr mice, CD4+ T cells were significantly decreased [160] and CD69 + CD44+ expressing splenic T cells were markedly reduced in Irf5/ RII.Yaa mice [161]. Importantly, the observed T cell regulatory properties of these Irfs, alone, do not account for all mechanisms of protection. Reduced plasma cell and increased serum cytokine levels by activating DCs and macrophages were also seen in Irf4/ (Fas)lpr mice [159]. Irf8/ NZB mice were almost completely absent of PDCs (CD11clowCD11b–B220+Siglec-H+PDCA1+) in the spleen; yet had intact B cell function [162]. Deficiency of Irf5 led to increasedCD19+ B cells in the MRL/lpr background, and reduced CD69+ B cells in the RII.Yaa background [161]. Together, these observations indicate that the mechanism(s) of protection in Irf-deficient mice was often distinct (Fig. 3). In the pristane model of induced lupus, Irf5-, Irf7- and Irf9deficient mice were all protected from disease to varying levels [163–165]. IgG2a/c is the primary pathogenic autoantibody induced in this model and it was suppressed in Irf5/ C57Bl/6 and Irf9/ Balb/c mice [165,166]. The mechanism(s) of selective autoantibody reduction differed between Irf-deficient mice. For instance, the protection seen in Irf5/ mice was at least partially due to a failure in early T cell activation and a skew towards Th2 polarization [166]. Irf5 has also been shown to directly regulate the IgG2a locus [27] and regulate monocyte responses, including cytokine/chemokine expression and trafficking [163]. In Irf7/ mice, the level of glomerular IgG deposits was similar to WT mice treated with pristane, yet autoantibodies to DNA- and RNAcontaining antigens was impaired [164]. In Irf9/ mice, B cells were found to express significantly lower levels of TLR7 mRNA and this was suggested as an explanation for the observed protection [165]. Importantly, neither skewed TH polarization nor impaired B cell function was found in Irf7/ mice [164]. Instead, Irf7/ mice were found to have decreased neutrophils and PDCs, suggesting that protection may come from loss of these cell types [164]. Interestingly, while Irf5/ C57Bl/6 and Irf9/ Balb/c mice were protected from proteinuria and kidney disease, Irf7/ C57Bl/6 mice developed glomerulonephritis as their WT littermates, but additional inhibition of the NFjB pathway provided
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
protection from nephritis [164]. These data implicate NFjB in Irf7mediated kidney disease. It is important to point out that in pristane-induced lupus, nephritis starts before detection of antidsDNA IgG [164,167,168]. This suggests that unlike human SLE, anti-dsDNA IgG does not play a role in pristane-induced nephritis and therefore caution should be taken when translating data from this model into human disease. Although Irf3/ mice have not yet been studied in the context of murine lupus, the role of Irf3 in lupus pathogenesis has been implicated in an IL23R-deficient mouse model [158]. IL23R deficiency protects from the development of lupus in C57BL/6-lpr/lpr mice [169]. Irf3 physically interacts with the promoter of IL23p19 and bone marrow-derived macrophages from Irf3/ C57BL/6 mice failed to produce IL23p19 following TLR3 stimulation [158]. These findings suggest that Irf3 may be a ‘lupus prone factor’. Unlike other IRFs, Irf2 deficiency made mice susceptible to spontaneous cutaneous lupus erythematosus (CLE)-like inflammation as early as 9 weeks-old [169]. Even though depletion of CD8+ cells prevented disease development, transfer of Irf2/ CD8+ failed to induce disease in irradiated mice. Therefore, it is likely that the disease develops as a result of the interaction of CD8+ T cells with another cell type(s) [169]. 4. Conclusions Although we have only focused on the more common autoimmune diseases in this review, it is important to point out that additional work is being done in humans and Irf-deficient mice that are not limited to the above discussed diseases. For instance, IRFs have been implicated in Hashimoto’s thyroiditis [170] and myasthenia gravis [171]. Irf1/ mice have been examined in the context of autoimmune heart disease and lymphocytic thyroiditis [172,173]. We can expect that this area of research will continue to grow and provide clarification towards IRF-mediated pathways that should be targeted for therapeutic intervention. Thus far, data indicate that the signaling pathways leading to and from different IRFs in autoimmune diseases are not independent - they can be synergistic, complementary, overlapping, or contrary. Examples of this are highlighted in Figs. 1, 2 and 3 revealing the complexities of IRF signaling dependent on cell type. To further elucidate distinct and overlapping signaling between the IRFs, it will be necessary to generate double and triple knockouts. An astonishing fact that becomes apparent in this review is the significant lack in translation from in vivo murine studies to human disease. The majority of data on IRFs in human disease is from GWAS. Few studies have begun to interrogate IRF signaling in the pathogenic immune cells from patients with autoimmune disease. These types of studies will be essential to our understanding of dysregulated IRF signaling in human autoimmune diseases since in the majority of diseases studied thus far, IRF expression and signaling are upregulated rather than absent, which is what we have been studying in Irf-deficient mice. Last, data presented herein support that not all roads from the IRFs lead to IFNs. Acknowledgements We thank Miriam Fein for carefully reading of the manuscript. B.J.B. is supported by grants from the Alliance for Lupus Research and The National Institutes of Health (AR065959-01). References [1] S. Ning, Interferon regulatory factors and autoimmune diseases, HSOA J. Med. Genom. Biomarkers 1 (2014). [2] K. Honda, A. Takaoka, T. Taniguchi, Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors, Immunity 25 (3) (2006) 349–360.
9
[3] T. Tamura et al., The IRF family transcription factors in immunity and oncogenesis, Annu. Rev. Immunol. 26 (2008) 535–584. [4] A. Battistini, Interferon regulatory factors in hematopoietic cell differentiation and immune regulation, J. Interferon Cytokine Res. 29 (12) (2009) 765–780. [5] J. Nehyba, R. Hrdlickova, H.R. Bose, Dynamic evolution of immune system regulators: the history of the interferon regulatory factor family, Mol. Biol. Evol. 26 (11) (2009) 2539–2550. [6] C.R. Escalante et al., Structure of IRF-1 with bound DNA reveals determinants of interferon regulation, Nature 391 (6662) (1998) 103–106. [7] M.A. Jensen, T.B. Niewold, Interferon regulatory factors: critical mediators of human lupus, Transl. Res. 165 (2) (2015) 283–295. [8] M. Lohoff, T.W. Mak, Roles of interferon-regulatory factors in T-helper-cell differentiation, Nat. Rev. Immunol. 5 (2) (2005) 125–135. [9] I. Rogatsky et al., Epigenetics and the IRFs: a complex interplay in the control of immunity and autoimmunity, Autoimmunity 47 (4) (2014) 242–255. [10] T. Cavlar, A. Ablasser, V. Hornung, Induction of type I IFNs by intracellular DNA-sensing pathways, Immunol. Cell Biol. 90 (5) (2012) 474–482. [11] K. Santana-de Anda et al., Interferon regulatory factors: beyond the antiviral response and their link to the development of autoimmune pathology, Autoimmun. Rev. 11 (2) (2011) 98–103. [12] L.A. O’Neill, A.G. Bowie, Sensing and signaling in antiviral innate immunity, Curr. Biol. 20 (7) (2010) R328–R333. [13] D.E. Levy, I.J. Marie, J.E. Durbin, Induction and function of type I and III interferon in response to viral infection, Curr. Opin. Virol. 1 (6) (2011) 476– 486. [14] E.C. Baechler et al., Gene expression profiling in human autoimmunity, Immunol. Rev. 210 (2006) 120–137. [15] T.M. Aune et al., Profiles of gene expression in human autoimmune disease, Cell Biochem. Biophys. 40 (2) (2004) 81–96. [16] S.M. Mariani, Genes and autoimmune diseases - a complex inheritance, MedGenMed 6 (4) (2004) 18. [17] I. Ricano-Ponce et al., Refined mapping of autoimmune disease associated genetic variants with gene expression suggests an important role for noncoding RNAs, J. Autoimmun. 68 (2016) 62–74. [18] Y. Kochi, Genetics of autoimmune diseases: perspectives from genome-wide association studies, Int. Immunol. 28 (4) (2016) 155–161. [19] J.C. Hall, A. Rosen, Type I interferons: crucial participants in disease amplification in autoimmunity, Nat. Rev. Rheumatol. 6 (1) (2010) 40–49. [20] L. Dou et al., The regulation role of interferon regulatory factor-1 gene and clinical relevance, Hum. Immunol. 75 (11) (2014) 1110–1114. [21] R.P. Donn et al., Cytokine gene polymorphisms and susceptibility to juvenile idiopathic arthritis. British Paediatric Rheumatology Study Group, Arthritis Rheum. 44 (4) (2001) 802–810. [22] Y.J. Lee et al., Associations between interferon regulatory factor-1 polymorphisms and Behcet’s disease, Hum. Immunol. 68 (9) (2007) 770–778. [23] M.S. Fife et al., No evidence for genetic association of interferon regulatory factor 1 in juvenile idiopathic arthritis, Arthritis Rheum. 56 (3) (2007) 972– 976. [24] G. Fortunato et al., Multiple sclerosis and hepatitis C virus infection are associated with single nucleotide polymorphisms in interferon pathway genes, J. Interferon Cytokine Res. 28 (3) (2008) 141–152. [25] N. Govind et al., Immunochip identifies novel, and replicates known, genetic risk loci for rheumatoid arthritis in black South Africans, Mol. Med. 20 (2014) 341–349. [26] M. Uddin et al., Autosome-wide copy number variation association analysis for rheumatoid arthritis using the WTCCC high-density SNP genotype data, J. Rheumatol. 38 (5) (2011) 797–801. [27] D.A. Savitsky et al., Contribution of IRF5 in B cells to the development of murine SLE-like disease through its transcriptional control of the IgG2a locus, Proc. Natl. Acad. Sci. USA 107 (22) (2010) 10154–10159. [28] J. Kawasaki et al., RASA1 functions in EPHB4 signaling pathway to suppress endothelial mTORC1 activity, J. Clin. Invest. 124 (6) (2014) 2774–2784. [29] P.S. Ramos et al., Familial aggregation and linkage analysis of autoantibody traits in pedigrees multiplex for systemic lupus erythematosus, Genes Immun. 7 (5) (2006) 417–432. [30] M. Akahoshi et al., Promoter polymorphisms in the IRF3 gene confer protection against systemic lupus erythematosus, Lupus 17 (6) (2008) 568– 574. [31] K. Santana-de Anda et al., Interferon regulatory factor 3 as key element of the interferon signature in plasmacytoid dendritic cells from systemic lupus erythematosus patients: novel genetic associations in the Mexican mestizo population, Clin. Exp. Immunol. 178 (3) (2014) 428–437. [32] S. Marecki, M.J. Fenton, The role of IRF-4 in transcriptional regulation, J. Interferon Cytokine Res. 22 (1) (2002) 121–133. [33] P.S. Biswas et al., Dual regulation of IRF4 function in T and B cells is required for the coordination of T-B cell interactions and the prevention of autoimmunity, J. Exp. Med. 209 (3) (2012) 581–596. [34] Y. Okada et al., Genetics of rheumatoid arthritis contributes to biology and drug discovery, Nature 506 (7488) (2014) 376–381. [35] E. Lopez-Isac et al., Brief report: IRF4 newly identified as a common susceptibility locus for systemic sclerosis and rheumatoid arthritis in a cross-disease meta-analysis of genome-wide association studies, Arthritis Rheum. 68 (9) (2016) 2338–2344. [36] S.S. Liu et al., No evidence for a genetic association of IRF4 with systemic lupus erythematosus in a Chinese population, Z. Rheumatol. 73 (6) (2014) 565–570.
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
10
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
[37] T. Krausgruber et al., IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses, Nat. Immunol. 12 (3) (2011) 231–238. [38] S.E. Sweeney, Targeting interferon regulatory factors to inhibit activation of the type I IFN response: implications for treatment of autoimmune disorders, Cell Immunol. 271 (2) (2011) 342–349. [39] 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. 76 (3) (2005) 528–537. [40] L. Tang et al., Association between IRF5 polymorphisms and autoimmune diseases: a meta-analysis, Genet. Mol. Res. 13 (2) (2014) 4473–4485. [41] Y. Li et al., Association of the IRF5 rs2070197 polymorphism with systemic lupus erythematosus: a meta-analysis, Clin. Rheumatol. 34 (9) (2015) 1495– 1501. [42] M.E. Alarcon-Riquelme et al., Genome-wide association study in an amerindian ancestry population reveals novel systemic lupus erythematosus risk loci and the role of European admixture, Arthritis Rheum. 68 (4) (2016) 932–943. [43] 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 104 (16) (2007) 6758–6763. [44] G.B. Nordang et al., Interferon regulatory factor 5 gene polymorphism confers risk to several rheumatic diseases and correlates with expression of alternative thymic transcripts, Rheumatology (Oxford) 51 (4) (2012) 619– 626. [45] A. Hellquist et al., Evidence for genetic association and interaction between the TYK2 and IRF5 genes in systemic lupus erythematosus, J. Rheumatol. 36 (8) (2009) 1631–1638. [46] E.A. Stahl et al., Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci, Nat. Genet. 42 (6) (2010) 508–514. [47] X. Jia et al., Association of the IRF5 rs2004640 polymorphism with rheumatoid arthritis: a meta-analysis, Rheumatol. Int. 33 (11) (2013) 2757– 2761. [48] F.D. Carmona et al., The systemic lupus erythematosus IRF5 risk haplotype is associated with systemic sclerosis, PLoS ONE 8 (1) (2013) e54419. [49] G. Kristjansdottir et al., Interferon regulatory factor 5 (IRF5) gene variants are associated with multiple sclerosis in three distinct populations, J. Med. Genet. 45 (6) (2008) 362–369. [50] K. Vandenbroeck et al., Lack of association between the interferon regulatory factor-1 (IRF1) locus at 5q31.1 and multiple sclerosis in Germany, northern Italy, Sardinia and Sweden, Genes Immun. 1 (4) (2000) 290–292. [51] V. Dideberg et al., An insertion-deletion polymorphism in the interferon regulatory Factor 5 (IRF5) gene confers risk of inflammatory bowel diseases, Hum. Mol. Genet. 16 (24) (2007) 3008–3016. [52] S.W. Han et al., Association of polymorphisms in interferon regulatory factor 5 gene with rheumatoid arthritis: a metaanalysis, J. Rheumatol. 36 (4) (2009) 693–697. [53] K. Honda et al., Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction, Nature 434 (7036) (2005) 1035–1040. [54] Q. Fu et al., Association of a functional IRF7 variant with systemic lupus erythematosus, Arthritis Rheum. 63 (3) (2011) 749–754. [55] W.D. Xu et al., Targeting IRF4 in autoimmune diseases, Autoimmun. Rev. 11 (12) (2012) 918–924. [56] R. Salloum, T.B. Niewold, Interferon regulatory factors in human lupus pathogenesis, Transl. Res. 157 (6) (2011) 326–331. [57] M. Heinig et al., A trans-acting locus regulates an anti-viral expression network and type 1 diabetes risk, Nature 467 (7314) (2010) 460–464. [58] F. Rosenbauer, D.G. Tenen, Transcription factors in myeloid development: balancing differentiation with transformation, Nat. Rev. Immunol. 7 (2) (2007) 105–117. [59] Y. Jiang et al., Two Genetic Variations in the IRF8 region are associated with Behcet’s disease in Han Chinese, Sci. Rep. 6 (2016) 19651. [60] D.S. Cunninghame Graham et al., Association of NCF2, IKZF1, IRF8, IFIH1, and TYK2 with systemic lupus erythematosus, PLoS Genet. 7 (10) (2011) e1002341. [61] S.W. Li et al., Single-nucleotide polymorphisms of IRF8 gene are associated with systemic lupus erythematosus in Chinese Han population, Int. J. Immunogenet. 41 (2) (2014) 112–118. [62] C.E. Weckerle et al., Network analysis of associations between serum interferon-alpha activity, autoantibodies, and clinical features in systemic lupus erythematosus, Arthritis Rheum. 63 (4) (2011) 1044–1053. [63] Z. Ren et al., IRF-1 signaling in central nervous system glial cells regulates inflammatory demyelination, J. Neuroimmunol. 233 (1–2) (2011) 147–159. [64] C. Agresti et al., Synergistic stimulation of MHC class I and IRF-1 gene expression by IFN-gamma and TNF-alpha in oligodendrocytes, Eur. J. Neurosci. 10 (9) (1998) 2975–2983. [65] K.K. Dev et al., Brain sphingosine-1-phosphate receptors: implication for FTY720 in the treatment of multiple sclerosis, Pharmacol. Ther. 117 (1) (2008) 77–93. [66] Y. Wang et al., STAT1/IRF-1 signaling pathway mediates the injurious effect of interferon-gamma on oligodendrocyte progenitor cells, Glia 58 (2) (2010) 195–208. [67] M. Horiuchi et al., Cooperative contributions of interferon regulatory factor 1 (IRF1) and IRF8 to interferon-gamma-mediated cytotoxic effects on oligodendroglial progenitor cells, J. Neuroinflammat. 8 (2011) 8.
[68] D.I. Mitsias et al., The Th1/Th2 cytokine balance changes with the progress of the immunopathological lesion of Sjogren’s syndrome, Clin. Exp. Immunol. 128 (3) (2002) 562–568. [69] J.M. van Woerkom et al., Salivary gland and peripheral blood T helper 1 and 2 cell activity in Sjogren’s syndrome compared with non-Sjogren’s sicca syndrome, Ann. Rheum. Dis. 64 (10) (2005) 1474–1479. [70] Y. Ohyama et al., Cytokine messenger RNA expression in the labial salivary glands of patients with Sjogren’s syndrome, Arthritis Rheum. 39 (8) (1996) 1376–1384. [71] E. Hagiwara et al., Altered frequency of type 1 cytokine secreting cells in the peripheral blood of patients with primary Sjogren’s syndrome, J. Rheumatol. 25 (1) (1998) 89–93. [72] K. Kohriyama, Y. Katayama, Disproportion of helper T cell subsets in peripheral blood of patients with primary Sjogren’s syndrome, Autoimmunity 32 (1) (2000) 67–72. [73] R.I. Fox et al., Cytokine mRNA expression in salivary gland biopsies of Sjogren’s syndrome, J. Immunol. 152 (11) (1994) 5532–5539. [74] L. Zheng et al., Expression of interferon regulatory factor 1, 3, and 7 in primary Sjogren syndrome, Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 107 (5) (2009) 661–668. [75] Y. Mamane et al., Interferon regulatory factors: the next generation, Gene 237 (1) (1999) 1–14. [76] M. Sjostrand et al., The expression of BAFF is controlled by IRF transcription factors, J. Immunol. 196 (1) (2016) 91–96. [77] C. Miceli-Richard et al., The CGGGG insertion/deletion polymorphism of the IRF5 promoter is a strong risk factor for primary Sjogren’s syndrome, Arthritis Rheum. 60 (7) (2009) 1991–1997. [78] R. Ganesan, M. Rasool, Fibroblast-like synoviocytes-dependent effector molecules as a critical mediator for rheumatoid arthritis: Current status and future directions. Int. Rev. Immunol. (2017) 1–11. [79] S.E. Sweeney, T.B. Kimbler, G.S. Firestein, Synoviocyte innate immune responses: II. Pivotal role of IFN regulatory factor 3, J. Immunol. 184 (12) (2010) 7162–7168. [80] J.A. Gracie et al., A proinflammatory role for IL-18 in rheumatoid arthritis, J. Clin. Invest. 104 (10) (1999) 1393–1401. [81] H. Okamura et al., Cloning of a new cytokine that induces IFN-gamma production by T cells, Nature 378 (6552) (1995) 88–91. [82] S. Ushio et al., Cloning of the cDNA for human IFN-gamma-inducing factor, expression in Escherichia coli, and studies on the biologic activities of the protein, J. Immunol. 156 (11) (1996) 4274–4279. [83] H. Okamura et al., A novel costimulatory factor for gamma interferon induction found in the livers of mice causes endotoxic shock, Infect. Immun. 63 (10) (1995) 3966–3972. [84] K. Kohno et al., IFN-gamma-inducing factor (IGIF) is a costimulatory factor on the activation of Th1 but not Th2 cells and exerts its effect independently of IL-12, J. Immunol. 158 (4) (1997) 1541–1550. [85] M.J. Micallef et al., Interferon-gamma-inducing factor enhances T helper 1 cytokine production by stimulated human T cells: synergism with interleukin-12 for interferon-gamma production, Eur. J. Immunol. 26 (7) (1996) 1647–1651. [86] H. Tsutsui et al., IFN-gamma-inducing factor up-regulates Fas ligandmediated cytotoxic activity of murine natural killer cell clones, J. Immunol. 157 (9) (1996) 3967–3973. [87] H. Tsutsui et al., IL-18 accounts for both TNF-alpha- and Fas ligand-mediated hepatotoxic pathways in endotoxin-induced liver injury in mice, J. Immunol. 159 (8) (1997) 3961–3967. [88] J.C. Morel et al., A novel role for interleukin-18 in adhesion molecule induction through NF kappa B and phosphatidylinositol (PI) 3-kinasedependent signal transduction pathways, J. Biol. Chem. 276 (40) (2001) 37069–37075. [89] M.A. Amin et al., Interleukin-18 induces angiogenic factors in rheumatoid arthritis synovial tissue fibroblasts via distinct signaling pathways, Arthritis Rheum. 56 (6) (2007) 1787–1797. [90] C.C. Park et al., Evidence of IL-18 as a novel angiogenic mediator, J. Immunol. 167 (3) (2001) 1644–1653. [91] J.C. Morel et al., Interleukin-18 induces rheumatoid arthritis synovial fibroblast CXC chemokine production through NFkappaB activation, Lab. Invest. 81 (10) (2001) 1371–1383. [92] V. Hurgin, D. Novick, M. Rubinstein, The promoter of IL-18 binding protein: activation by an IFN-gamma -induced complex of IFN regulatory factor 1 and CCAAT/enhancer binding protein beta, Proc. Natl. Acad. Sci. USA 99 (26) (2002) 16957–16962. [93] H. Marotte et al., Blocking of interferon regulatory factor 1 reduces tumor necrosis factor alpha-induced interleukin-18 bioactivity in rheumatoid arthritis synovial fibroblasts by induction of interleukin-18 binding protein a: role of the nuclear interferon regulatory factor 1-NF-kappaB-c-jun complex, Arthritis Rheum. 63 (11) (2011) 3253–3262. [94] W. Zhu et al., Anti-citrullinated protein antibodies induce macrophage subset disequilibrium in RA patients, Inflammation 38 (6) (2015) 2067–2075. [95] Z. Wen, C. Fiocchi, Inflammatory bowel disease: autoimmune or immunemediated pathogenesis?, Clin Dev. Immunol. 11 (3–4) (2004) 195–204. [96] M. Clavell et al., Detection of interferon regulatory factor-1 in lamina propria mononuclear cells in Crohn’s disease, J. Pediatr. Gastroenterol. Nutr. 30 (1) (2000) 43–47.
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
B. Matta et al. / Cytokine xxx (2017) xxx–xxx [97] T. Hisamatsu, M. Suzuki, D.K. Podolsky, Interferon-gamma augments CARD4/ NOD1 gene and protein expression through interferon regulatory factor-1 in intestinal epithelial cells, J. Biol. Chem. 278 (35) (2003) 32962–32968. [98] M. Bilenker et al., Interleukin-7 activates intestinal lymphocytes, Dig. Dis. Sci. 40 (8) (1995) 1744–1749. [99] M. Watanabe et al., Interleukin 7 is produced by human intestinal epithelial cells and regulates the proliferation of intestinal mucosal lymphocytes, J. Clin. Invest. 95 (6) (1995) 2945–2953. [100] G. Monteleone et al., Response of human intestinal lamina propria T lymphocytes to interleukin 12: additive effects of interleukin 15 and 7, Gut 43 (5) (1998) 620–628. [101] S. Oshima et al., Interferon regulatory factor 1 (IRF-1) and IRF-2 distinctively up-regulate gene expression and production of interleukin-7 in human intestinal epithelial cells, Mol. Cell Biol. 24 (14) (2004) 6298–6310. [102] N. Ogasawara et al., Rebamipide suppresses TLR-TBK1 signaling pathway resulting in regulating IRF3/7 and IFN-alpha/beta reduction, J. Clin. Biochem. Nutr. 48 (2) (2011) 154–160. [103] J. Mudter et al., IRF4 regulates IL-17A promoter activity and controls RORgammat-dependent Th17 colitis in vivo, Inflamm. Bowel Dis. 17 (6) (2011) 1343–1358. [104] V.R. Sutton et al., A critical role for granzyme B, in addition to perforin and TNFalpha, in alloreactive CTL-induced mouse pancreatic beta cell death, Transplantation 81 (2) (2006) 146–154. [105] A.A. Bengtsson et al., Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies, Lupus 9 (9) (2000) 664–671. [106] U. Bave et al., Fc gamma RIIa is expressed on natural IFN-alpha-producing cells (plasmacytoid dendritic cells) and is required for the IFN-alpha production induced by apoptotic cells combined with lupus IgG, J. Immunol. 171 (6) (2003) 3296–3302. [107] U. Bave et al., Activation of natural interferon-alpha producing cells by apoptotic U937 cells combined with lupus IgG and its regulation by cytokines, J. Autoimmun. 17 (1) (2001) 71–80. [108] M.W. Boule et al., Toll-like receptor 9-dependent and -independent dendritic cell activation by chromatin-immunoglobulin G complexes, J. Exp. Med. 199 (12) (2004) 1631–1640. [109] B.J. Barnes et al., Multiple regulatory domains of IRF-5 control activation, cellular localization, and induction of chemokines that mediate recruitment of T lymphocytes, Mol. Cell Biol. 22 (16) (2002) 5721–5740. [110] D. Feng et al., Genetic variants and disease[HYPHEN]associated factors contribute to enhanced interferon regulatory factor 5 expression in blood cells of patients with systemic lupus erythematosus, Arthritis Rheum 62 (2) (2010) 562–573. [111] R.C. Stone et al., Interferon regulatory factor 5 activation in monocytes of systemic lupus erythematosus patients is triggered by circulating autoantigens independent of type I interferons, Arthritis Rheum 64 (3) (2012) 788–798. [112] T.B. Niewold et al., Association of the IRF5 risk haplotype with high serum interferon-alpha activity in systemic lupus erythematosus patients, Arthritis Rheum. 58 (8) (2008) 2481–2487. [113] R.C. Stone et al., Interferon regulatory factor 5 activation in monocytes of systemic lupus erythematosus patients is triggered by circulating autoantigens independent of type I interferons, Arthritis Rheum. 64 (3) (2012) 788–798. [114] J.M. Guthridge et al., Effects of IRF5 lupus risk haplotype on pathways predicted to influence B cell functions, J. Biomed. Biotechnol. 2012 (2012) 594056. [115] M. Hedl, C. Abraham, IRF5 risk polymorphisms contribute to interindividual variance in pattern recognition receptor-mediated cytokine secretion in human monocyte-derived cells, J. Immunol. 188 (11) (2012) 5348–5356. [116] M. Hedl, J. Yan, C. Abraham, IRF5 and IRF5 disease-risk variants increase glycolysis and human M1 macrophage polarization by regulating proximal signaling and Akt2 activation, Cell Rep. 16 (9) (2016) 2442–2455. [117] A. Takaoka et al., Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors, Nature 434 (7030) (2005) 243–249. [118] R. Salloum et al., Genetic variation at the IRF7/PHRF1 locus is associated with autoantibody profile and serum interferon-alpha activity in lupus patients, Arthritis Rheum. 62 (2) (2010) 553–561. [119] S.E. Sweeney, Hematopoietic stem cell transplant for systemic lupus erythematosus: Interferon regulatory factor 7 activation correlates with the IFN signature and recurrent disease, Lupus 20 (9) (2011) 975–980. [120] Z. Zhang et al., Global H4 acetylation analysis by ChIP-chip in systemic lupus erythematosus monocytes, Genes Immun. 11 (2) (2010) 124–133. [121] Y.T. Leung et al., Interferon regulatory factor 1 and histone H4 acetylation in systemic lupus erythematosus, Epigenetics 10 (3) (2015) 191–199. [122] Z. Zhang et al., Interferon regulatory factor 1 marks activated genes and can induce target gene expression in systemic lupus erythematosus, Arthritis Rheum. 67 (3) (2015) 785–796. [123] A. Kawasaki et al., Association of functional polymorphisms in interferon regulatory factor 2 (IRF2) with susceptibility to systemic lupus erythematosus: a case-control association study, PLoS ONE 9 (10) (2014) e109764. [124] B.S. Chrabot et al., Genetic variation near IRF8 is associated with serologic and cytokine profiles in systemic lupus erythematosus and multiple sclerosis, Genes Immun. 14 (8) (2013) 471–478.
11
[125] Y. Yoshida et al., The transcription factor IRF8 activates integrin-mediated TGF-beta signaling and promotes neuroinflammation, Immunity 40 (2) (2014) 187–198. [126] Y. Tada et al., Reduced incidence and severity of antigen-induced autoimmune diseases in mice lacking interferon regulatory factor-1, J. Exp. Med. 185 (2) (1997) 231–238. [127] D.C. Fitzgerald et al., Interferon regulatory factor (IRF) 3 is critical for the development of experimental autoimmune encephalomyelitis, J. Neuroinflammat. 11 (2014) 130. [128] A. Brustle et al., The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4, Nat. Immunol. 8 (9) (2007) 958–966. [129] T. Buch, C. Uthoff-Hachenberg, A. Waisman, Protection from autoimmune brain inflammation in mice lacking IFN-regulatory factor-1 is associated with Th2-type cytokines, Int. Immunol. 15 (7) (2003) 855–859. [130] Z. Ren et al., Central nervous system expression of interferon regulatory factor 1 regulates experimental autoimmune encephalomyelitis, J. Neuroimmune Pharmacol. 5 (2) (2010) 260–265. [131] M. Salem et al., Interferon regulatory factor-7 modulates experimental autoimmune encephalomyelitis in mice, J. Neuroinflammat. 8 (2011) 181. [132] Y. Tada et al., Collagen-induced arthritis in CD4- or CD8-deficient mice: CD8+ T cells play a role in initiation and regulate recovery phase of collageninduced arthritis, J. Immunol. 156 (11) (1996) 4520–4526. [133] B.A. Goupil et al., Novel lesions of bones and joints associated with chikungunya virus infection in two mouse models of disease: new insights into disease pathogenesis, PLoS ONE 11 (5) (2016) e0155243. [134] C. Schilte et al., Cutting edge: independent roles for IRF-3 and IRF-7 in hematopoietic and nonhematopoietic cells during host response to Chikungunya infection, J. Immunol. 188 (7) (2012) 2967–2971. [135] P.A. Monach, D. Mathis, C. Benoist, The K/BxN arthritis model, Curr. Protoc. Immunol. (2008) Chapter 15: p. Unit 15 22. [136] A.D. Christensen et al., K/BxN serum-transfer arthritis as a model for human inflammatory arthritis, Front. Immunol. 7 (2016) 213. [137] S.E. Sweeney, M. Corr, T.B. Kimbler, Role of interferon regulatory factor 7 in serum-transfer arthritis: regulation of interferon-beta production, Arthritis Rheum. 64 (4) (2012) 1046–1056. [138] J. van Holten et al., Treatment with recombinant interferon-beta reduces inflammation and slows cartilage destruction in the collagen-induced arthritis model of rheumatoid arthritis, Arthritis Res. Ther. 6 (3) (2004) R239–R249. [139] A. Yarilina, E. DiCarlo, L.B. Ivashkiv, Suppression of the effector phase of inflammatory arthritis by double-stranded RNA is mediated by type I IFNs, J. Immunol. 178 (4) (2007) 2204–2211. [140] M. Corr et al., Synergistic benefit in inflammatory arthritis by targeting I kappaB kinase epsilon and interferon beta, Ann. Rheum. Dis. 68 (2) (2009) 257–263. [141] J. Li et al., Death receptor 5-targeted depletion of interleukin-23-producing macrophages, Th17, and Th1/17 associated with defective tyrosine phosphatase in mice and patients with rheumatoid arthritis, Arthritis Rheum. 65 (10) (2013) 2594–2605. [142] Q. Chen et al., IRF-4-binding protein inhibits interleukin-17 and interleukin21 production by controlling the activity of IRF-4 transcription factor, Immunity 29 (6) (2008) 899–911. [143] E.E. Mannick et al., Altered phenotype of dextran sulfate sodium colitis in interferon regulatory factor-1 knock-out mice, J. Gastroenterol. Hepatol. 20 (3) (2005) 371–380. [144] H. Negishi et al., Essential contribution of IRF3 to intestinal homeostasis and microbiota-mediated Tslp gene induction, Proc. Natl. Acad. Sci. USA 109 (51) (2012) 21016–21021. [145] X.D. Li et al., Mitochondrial antiviral signaling protein (MAVS) monitors commensal bacteria and induces an immune response that prevents experimental colitis, Proc. Natl. Acad. Sci. USA 108 (42) (2011) 17390–17395. [146] P. Fuenzalida et al., Toll-like receptor 3 pre-conditioning increases the therapeutic efficacy of umbilical cord mesenchymal stromal cells in a dextran sulfate sodium-induced colitis model, Cytotherapy 18 (5) (2016) 630–641. [147] Z. Saadatdoust et al., Dietary cocoa inhibits colitis associated cancer: a crucial involvement of the IL-6/STAT3 pathway, J. Nutr. Biochem. 26 (12) (2015) 1547–1558. [148] R. Atreya, M.F. Neurath, Involvement of IL-6 in the pathogenesis of inflammatory bowel disease and colon cancer, Clin. Rev. Allergy Immunol. 28 (3) (2005) 187–196. [149] Y. Kai et al., Colitis in mice lacking the common cytokine receptor gamma chain is mediated by IL-6-producing CD4+ T cells, Gastroenterology 128 (4) (2005) 922–934. [150] J. Mudter et al., The transcription factor IFN regulatory factor-4 controls experimental colitis in mice via T cell-derived IL-6, J. Clin. Invest. 118 (7) (2008) 2415–2426. [151] R. Atreya et al., Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in crohn disease and experimental colitis in vivo, Nat. Med. 6 (5) (2000) 583– 588. [152] T. Nakazawa et al., Complete suppression of insulitis and diabetes in NOD mice lacking interferon regulatory factor-1, J. Autoimmun. 17 (2) (2001) 119– 125. [153] K. Suk et al., IFN-gamma/TNF-alpha synergism as the final effector in autoimmune diabetes: a key role for STAT1/IFN regulatory factor-1 pathway in pancreatic beta cell death, J. Immunol. 166 (7) (2001) 4481–4489.
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006
12
B. Matta et al. / Cytokine xxx (2017) xxx–xxx
[154] S. Akazawa et al., Haploinsufficiency of interferon regulatory factor 4 strongly protects against autoimmune diabetes in NOD mice, Diabetologia 58 (11) (2015) 2606–2614. [155] K. Yasuda et al., Murine dendritic cell type I IFN production induced by human IgG-RNA immune complexes is IFN regulatory factor (IRF)5 and IRF7 dependent and is required for IL-6 production, J. Immunol. 178 (11) (2007) 6876–6885. [156] D. Perry et al., Murine models of systemic lupus erythematosus, J. Biomed. Biotechnol. 2011 (2011) 271694. [157] C.M. Reilly et al., Interferon regulatory factor-1 gene deletion decreases glomerulonephritis in MRL/lpr mice, Eur. J. Immunol. 36 (5) (2006) 1296– 1308. [158] S. Smith et al., Enhanced interferon regulatory factor 3 binding to the interleukin-23p19 promoter correlates with enhanced interleukin-23 expression in systemic lupus erythematosus, Arthritis Rheum. 64 (5) (2012) 1601–1609. [159] M. Lech et al., IRF4 deficiency abrogates lupus nephritis despite enhancing systemic cytokine production, J. Am. Soc. Nephrol. 22 (8) (2011) 1443–1452. [160] Y. Tada et al., Interferon regulatory factor 5 is critical for the development of lupus in MRL/lpr mice, Arthritis Rheum. 63 (3) (2011) 738–748. [161] C. Richez et al., IFN regulatory factor 5 is required for disease development in the FcgammaRIIB-/-Yaa and FcgammaRIIB-/- mouse models of systemic lupus erythematosus, J. Immunol. 184 (2) (2010) 796–806. [162] R. Baccala et al., Essential requirement for IRF8 and SLC15A4 implicates plasmacytoid dendritic cells in the pathogenesis of lupus, Proc. Natl. Acad. Sci. USA 110 (8) (2013) 2940–2945. [163] L. Yang et al., Monocytes from Irf5-/- mice have an intrinsic defect in their response to pristane-induced lupus, J. Immunol. 189 (7) (2012) 3741–3750.
[164] F. Miyagawa et al., Essential requirement for IFN regulatory factor 7 in autoantibody production but not development of nephritis in murine lupus, J. Immunol. 197 (6) (2016) 2167–2176. [165] D.L. Thibault et al., IRF9 and STAT1 are required for IgG autoantibody production and B cell expression of TLR7 in mice, J. Clin. Invest. 118 (4) (2008) 1417–1426. [166] D. Feng et al., Irf5-deficient mice are protected from pristane-induced lupus via increased Th2 cytokines and altered IgG class switching, Eur. J. Immunol. 42 (6) (2012) 1477–1487. [167] H.B. Richards et al., Interleukin 6 dependence of anti-DNA antibody production: evidence for two pathways of autoantibody formation in pristane-induced lupus, J. Exp. Med. 188 (5) (1998) 985–990. [168] M. Satoh et al., Anti-nuclear antibody production and immune-complex glomerulonephritis in BALB/c mice treated with pristane, Proc. Natl. Acad. Sci. USA 92 (24) (1995) 10934–10938. [169] S. Hida et al., CD8(+) T cell-mediated skin disease in mice lacking IRF-2, the transcriptional attenuator of interferon-alpha/beta signaling, Immunity 13 (5) (2000) 643–655. [170] G. Ceresini et al., Association between idiopathic retroperitoneal fibrosis and autoimmune thyroiditis: a case-control study, Autoimmun Rev. 14 (1) (2015) 16–22. [171] Z. Zagoriti et al., Genetics of myasthenia gravis: a case-control association study in the Hellenic population, Clin. Dev. Immunol. 2012 (2012) 484919. [172] J. Tani et al., Prevention of lymphocytic thyroiditis in iodide-treated nonobese diabetic mice lacking interferon regulatory factor-1, Eur. J. Endocrinol. 147 (6) (2002) 809–814. [173] K. Bachmaier et al., INOS expression and nitrotyrosine formation in the myocardium in response to inflammation is controlled by the interferon regulatory transcription factor 1, Circulation 96 (2) (1997) 585–591.
Please cite this article in press as: B. Matta et al., Interferon regulatory factor signaling in autoimmune disease, Cytokine (2017), http://dx.doi.org/10.1016/ j.cyto.2017.02.006