Skip to main content

REVIEW article

Front. Immunol., 06 September 2013
Sec. Molecular Innate Immunity
This article is part of the Research Topic Type I Interferon in Human Autoimmunity View all 10 articles

The Role of Type 1 Interferon in Systemic Sclerosis

  • Division of Rheumatology and Clinical Immunogenetics, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA

Systemic Sclerosis (Scleroderma, SSc) is an autoimmune disease characterized by vasculopathy, inflammation, and fibrosis that can lead to loss of organ function. Type I interferons (IFNs) are family of cytokines that mitigate the deleterious effects of viral and bacterial infections in the innate immunity system. Past several years, research efforts have been focused on the role of type I IFN and IFN-inducible genes in the pathogenesis of SSc. Polymorphisms in the Interferon regulatory factor (IRF)-5, IRF7, and IRF8 are associated with SSc, Similarly, polymorphism of Signal Transducer and Activator of Transcription (STAT)-4, has been established as a genetic risk factor of SSc. IRFs and STAT4 proteins are key activators of type I IFN signaling pathways. An IFN signature (increased expression and activation of IFN-regulated genes) has been observed in the peripheral blood and skin biopsy samples of patients with SSc. Furthermore, a plasma IFN-inducible chemokine score correlated with markers of disease severity and autoantibody subtypes in SSc. In this review, we summarize our current knowledge of the role of type I IFNs and IFN-inducible genes in the pathogenesis of SSc and their potential role as biomarkers and therapeutic targets.

Introduction

Systemic sclerosis (Scleroderma, SSc) is characterized by immune dysregulation, fibrosis, and vasculopathy although its pathogenesis is not completely understood (1). Disease morbidity and mortality remain high (2, 3). There is no definite cure for SSc and the available treatments have limited efficacy. The major hurdle in developing effective therapies for SSc is an incomplete understanding of disease pathogenesis. A better understanding of SSc pathogenesis is important for identifying more targeted and effective therapeutic approaches.

Recently, there has been an increasing interest in the role of type I interferons (IFNs) in pathogenesis and severity of SSc. IFNs are a heterogeneous family of multifunctional cytokines. They were originally identified as proteins responsible for induction of cellular resistance to viral infections. Type I IFNs include IFN-α, -β, and -ω, and alleviate the effects of viral and bacterial infections in the innate immunity system (4, 5). Type I IFN subtypes-α and -β share common multicomponent, cell surface receptors, and elicit a similar range of biological responses, including antiviral, anti-proliferative (6), and immune modulatory activities.

In this review, we summarize the current knowledge about the role of type I IFN and its inducible genes in the SSc pathogenesis and biomarker development.

Innate Immunity and SSc

The innate immune system is the first line of host defense against pathogens. It plays an important part in triggering inflammation and promoting development of fibrosis in many organ systems. The dominant cellular components of innate immunity are mainly neutrophils, macrophages, and dendritic cells. These cells sense pathogens and destroy them, followed by secretion of pro-inflammatory chemokines and cytokines to activate T cells and other components of adaptive immune system. There is an increasing evidence for activation of the innate immune system in SSc. Cells involved in the innate immune system are detected at the end organ damage site of SSc (7, 8). Being the first cells in line in the defense against pathogens of any sort, the antigen presenting cells (APCs) are often considered the most influential cell of the innate immune system. The specific nature of these APC and how they contribute to the development of fibrosis is still unclear. The perivascular infiltrates in the non-lesional skin of SSc patients mainly consists of macrophages/monocytes and CD4+ T cells suggesting that the aberrant or dysregulated immune system precedes fibrosis (911). Alternatively activated macrophages are present in SSc skin biopsies (8, 12, 13), this type of macrophages are potentially important source of profibrotic cytokines including transforming growth factor β (TGF-β) which contribute to resolving inflammation and promoting wound healing (14). The sub-classification of macrophages into classically activated M1 macrophages and alternatively activated M2 macrophages is also of special interest in SSc because the M1 type is clearly more inflammatory and the M2 type is thought to be more involved in tissue remodeling and profibrotic phenotypes. M2 macrophages highly express several receptors such as hemoglobin scavenger receptor (CD163), class A scavenger receptor (CD204), and mannose receptor (CD206) (15, 16). SSc patients show significantly higher serum soluble CD163 levels, and the number of CD163+ and CD204+ activated M2 macrophages is significantly greater in SSc skin (17, 18). The role of M2 macrophages for the development of fibrosis in SSc is still speculative, further studies are needed to clarify the potential mechanism of M2 macrophages in this disorder.

In this non-specific immune system, mast cells, basophils, and natural killer (NK) T cells play more specialized immune functions. For instance, dermal mast cell number density was significantly higher in diffuse SSc patients than in unaffected controls (19, 20). Electron microscopy (EM) with immunogold labeling in skin biopsy samples revealed that patients with progressive SSc (worsening skin thickening and/or organ function in the year preceding biopsy) had higher number of mast cells. Furthermore, mast cell vesicles containing active TGF-β in patients with SSc showed higher level of degranulation than those from unaffected controls (21). The number of basophils, a circulating counterpart of mast cells was increased in SSc patients. Spontaneous histamine releasability, its reactivity to IgE and response to IL-3 were increased in basophiles from patients with SSc (22). On the other hand, the absolute number and proportion of NK T cells were decreased in patients with SSc which possibly can lead to down-regulation of the normal immune response (23). Altogether, these observations implicate a dysregulated immune system in the pathogenesis of SSc.

In line with those observations, large efforts have been made to find the genetic risk factors for abnormal immune system in SSc. These studies independently replicated genetic risk factors such as STAT4 (2426), BLK (2730), BANK1 (31, 32), Interferon regulatory factor (IRF)-5 (33, 34), IRF-7, and -8 (3538), and the T cell receptor zeta-chain (CD247) (26) which are involved in innate and adaptive immune system.

Type I IFNs and SSc

Type I IFNs are important key regulators of the innate immune system. They modulate immune cell differentiation and proliferation, as well as inflammatory cytokine production. Recent studies have provided considerable evidence that implicates a dysregulation in type I IFN and IFN-inducible genes in the pathophysiology of autoimmune diseases including SSc (3943). SSc shares this common characteristic with systemic lupus erythematosus (SLE) (44). Anti-IFNα mAb, sifalimumab, was evaluated in a phase Ia study with an open-label extension of 67 SLE patients with moderately active disease. Sifalimumab caused dose-dependent inhibition of type I IFN-induced mRNAs in whole blood and corresponding changes in related proteins in affected skin. Exploratory analyses showed consistent trends toward improvement in disease activity (44). In a follow-up phase Ib randomized, controlled trial with 161 SLE patients, no statistically significant differences in clinical activity between sifalimumab and placebo were observed. However, when adjusted for excess burst steroids, change in disease activity, and complement levels from baseline showed a positive trend over time (45).

Approximately, half of SSc patients have an increased expression of IFN-regulated genes (termed the “IFN signature”) in their peripheral whole blood cells (46). Recent studies demonstrated activation of type I IFN system was present in SSc sera and plasmacytoid dendritic cells (pDCs) were the main source of IFN-α production (47, 48). Tan et al. first reported a distinct transcript pattern of dysregulated type I IFN-inducible genes in peripheral blood cells (whole blood) from patients with SSc (40). This finding was subsequently confirmed in peripheral blood mononuclear cells (41). The development of SSc has been reported in patients undergoing IFN treatment (49). Furthermore, a randomized, placebo-controlled trial of subcutaneous IFN-α injection in patients with early SSc showed that treatment with IFN-α resulted in worsening lung function and a smaller degree of improvement in skin thickening scores compared to placebo (50). Although the presence of an activated IFN system could be demonstrated, the exact mechanism by which the dysregulated type I IFN signaling contributes to the pathophysiology of fibrosis in SSc is still unknown.

The innate immune system responds rapidly to the presence of certain motifs or patterns that microbes possess commonly such as unmethylated DNA rich in CpG dinucleotides, dsRNA, and bacterial cell wall components via pattern recognition receptors (PRRs) (51). These PRRs are widely expressed on cells of the immune system, as well as endothelial, epithelial, and mesenchymal cells including fibroblasts. Some of the most prominent PRRs are the Toll-like receptors (TLRs). TLRs located on various cellular membranes to sense exogenous and endogenous danger-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), and play a critical role in innate immune responses. TLR activation triggers production and secretion of several inflammatory cytokines including type I IFNs (52, 53). The TLR family includes both extracellular and endosomal receptors. The first is based on the cell surface like TLR-2, -4, -5, and -6 and recognize patterns found primarily on bacteria, mycobacteria, fungal, and parasitic organisms (54), the latter is located in endosome like TLR-7 and -8. TLR-9 is localized in the endoplasmic reticulum and translocated to the endosome upon response to bacteria DNA. They recognize a wide variety of pathogen components, and all the TLRs except TLR3 signal through the adaptor molecule MyD88, activate and stimulate type I IFN production. Bhattacharyya et al. reported that TLR4 was overexpressed in the skin and lung tissues, as well as explanted skin fibroblasts from patients with SSc (55). Our recent findings revealed that TLR3 expression was upregulated in patients with SSc and IFN-α2 induced an up-regulation of TLR3 in human dermal fibroblasts which is more prominent in SSc patients than in unaffected control subjects (56). These findings are suggesting an important role for TLR activation via type I IFNs in fibroblast biology.

Interferon Regulatory Factors and SSc

Interferon regulatory factors are best characterized as transcriptional regulators of type I IFNs and IFN-inducible genes and play a pivotal role in regulation of many facets of innate and adaptive immune response (57). This family is composed of nine members: IRF1, IRF2, IRF3, IRF4 (also known as LSIRF, PIP, or ICSAT), IRF5, IRF6, IRF7, IRF8 (also known as ICSBP), and IRF9 (also known as ISGF3γ) (58, 59). As transcriptional factors, each IRF contains a well-conserved DNA-binding domain which is located at the amino terminus and forms a helix-turn-helix-motif. This region recognizes a consensus DNA sequence known as the IFN-stimulated response element (ISRE) in the promoters of targeted genes (5961). These IRFs coordinate the expression of type I IFNs and type I IFN-inducible genes (57). Several genetic polymorphisms have been associated with SSc in multiple case–control studies and a few family studies (Table 1). Some of these genetic variants are associated with susceptibility for development of SSc, while others act as disease modifiers. Recent genome-wide association studies (GWASs) also confirmed IRFs as genetic susceptibility loci in autoimmune diseases.

TABLE 1
www.frontiersin.org

Table 1. Polymorphisms in the interferon regulatory factors associated with systemic sclerosis.

IRF5 is a transcription factor which induces the transcription of IFN-α and other early pro-inflammatory cytokines (62, 63). In vitro experiments have shown that in virus-infected cells, IRF5 is activated by phosphorylation, resulting in nuclear translocation and stimulation of IFN-α (64). Initial analysis of the role of IRF5 in the innate antiviral response utilizing IRF5 mutant mice showed impairment of interleukin-6 (IL-6) and TNF-α production in splenic dendritic cells. IRF5 mutant mice are highly sensitive to viral infection and show lower levels of type I IFN in the serum. IFN production was also impaired in the infected macrophages from IRF5 mutant mice (65, 66). Genetic variants of IRF5 are associated with SSc susceptibility (6769).

The minor allele of the IRF5 single-nucleotide polymorphism (SNP) rs4728142 was shown to be predictive of longer survival in the two independent SSc cohorts. The association of this SNP with survival was independent of age at disease onset, disease type, and autoantibody profiles (33). This minor allele was also associated with lower IRF5 transcript expression in monocytes of patients and controls suggesting functional relevance of rs4728142 or it associated SNPs for IRF5 expression.

IRF7 is one of transcription factors involved in IFN signaling pathways which is activated by TLRs TLR3/7/9 or retinoic acid-inducible gene 1 (RIG-1) in response to nucleic acid (both DNA and RNA) immune complexes. Activated IRF7 leads to secretion of a large amount of type I IFN (70). Its expression can potentially be enhanced via a positive feedback loop through IFN receptor and ISGF3 activation, leading to increased IRF7 over-expression and subsequently additional IFNα, transcription (71). IRF7 is essential for the induction of IFN-α/β genes via the virus-activated, MyD88-independent pathway and the TLR-activated, MyD88-dependent pathway (72). Inactive IRF7 resides in the cytoplasm. With pathogenic stimulation, IRF7 is phosphorylated, activated, and translocated into the nucleus, where it forms a transcriptional complex with other co-activators and binds to promoter regions of target genes including IFN-α/-β (73, 74). IRF7 also regulates the pro-inflammatory cytokine IL-6 in pDCs and monocytes (75, 76). The viral induction of MyD88-independent IFN-α/β genes is severely impaired in IRF7 null fibroblasts. Consistently, markedly decreased serum IFN-α level were also observed in IRF7 null mice (72). These studies demonstrated the importance of IRF7 dependent systemic IFN response for the innate immunity. Furthermore, recent genetic studies have established IRF7 as a susceptibility locus in SLE (7780). Similarly, our group recently reported that a functional variant in the IRF7 exonic region, rs1131665 was associated with SSc (35). These findings support that IRF7 may represent a common risk factor for systemic autoimmune disease processes, including SSc. Microarray studies revealed up-regulation of IRF7 mRNA level in whole peripheral blood cells from SSc patients with early diseases (40). Another independent study showed no statistically significant difference in IRF7 transcript levels in PBMCs of SSc patients compared to controls by quantitative PCR analysis (81). However, patients with late stage disease and a smaller sample size were investigated in this study. Further investigations are needed to determine the contributory role of IRF7 in pathogenesis of SSc.

IRF8 is another immune cell specific IRF family member. It participates in the MyD88-dependent signaling pathway through interaction with TRAF6 (82). IRF8 is required for the induction of Type I IFN genes by viruses and TLR ligands in DCs (83). IRF8 is known to be involved in the development of dendritic cells (84). IRF8 also promotes B cell differentiation (85). Recently, the IRF8 SNP, rs11642873 was identified as a risk factor for limited and anti-centromere positive SSc patients in a large GWAS follow-up study conducted in European and North-American cohorts (36). Another independent study identified rs2280381 polymorphism in IRF8 as a susceptibility locus of SSc in the Japanese population (37). The association of IRF8 genetic variants with SSc supports possible involvement of B cells and dendritic cells in the development of SSc. However, the role and importance of B cells or dendritic cells in the fibrotic component of SSc has not been well established (8688).

Further fine-mapping and functional studies are crucial for elucidating the role of genetic variants in the IRFs in the pathogenesis of SSc.

Interferon Inducible Cytokines and Chemokines in Scleroderma

Interleukin-6 is one of the most prominent cytokines activated by IFN pathway. It is involved in the pathogenesis of many immune-mediated diseases including SSc (8991). IL-6 is a classic inflammatory cytokine produced by various cells and involved in B cell differentiation, induction of acute phase proteins in liver cells, proliferation, and differentiation of T cells (92, 93). By binding to the IL-6 receptor (IL-6R)-α chain and the signal transducing component gp130 (CD130), pleiotropic IL-6 activates downstream signaling mediated by STAT1 or STAT3 through tyrosine phosphorylation. Previous studies have shown that IL-6 plays an important role in the initiation and promotion of fibrosis (94, 95). Production of IL-6 and soluble IL-6R by cultured peripheral blood mononuclear cells were significantly higher in patients with SSc and soluble IL-6R levels significantly correlated with the severity of pulmonary fibrosis in patients with SSc (96). Serum IL-6 levels might be predictive of disease progression in Interstitial lung disease associated with SSc (97). IL-6 shifts T cells from regulatory response to pathogenic Th17 response (98), and promotes the differentiation of CD4+ cells to a profibrotic Th2 type while suppressing Th1 differentiation (99). IL-6 stimulation induces increased collagen production in dermal fibroblasts (100, 101). These studies demonstrate that IL-6 is involved in the pathogenesis of SSc and may contribute to progression of fibrosis and disease severity in SSc.

A combined score of the plasma IFN-inducible chemokines, IFNγ-inducible protein 10 (IP-10/CXCL10), and IFN-inducible T cell α chemoattractant (I-TAC/CXCL11) highly correlated with the IFN gene expression signature in SSc patients in the Genetics versus Environment in Scleroderma Outcome Study (GENISOS) cohort study (102). As expected, SSc patients had higher IFN-inducible chemokine scores than age-, gender-, and ethnicity-matched controls. Among 266 SSc patients, the IFN-inducible chemokine score was associated with presence of anti-U1 RNP antibodies while patients with anti-RNA polymerase III antibodies had lower levels of this chemokine score. The lower IFN chemokine levels in patients with anti-RNA polymerase III antibodies might be of important biological significance because these antibodies are associated with presence of diffuse cutaneous involvement and absence of severe interstitial lung disease. The IFN-inducible chemokine score was not associated with disease duration, disease type, or other auto-antibodies. The chemokine score correlated positively with the concomitantly obtained scores on the Medsger Severity Index for muscle, skin, and lung involvement, as well as creatine kinase levels in SSc. There was also a negative correlation with forced vital capacity and diffusing capacity for carbon monoxide. These results support the aforementioned findings that the IFN activation is associated with the more severe form of SSc. There was no significant change observed in the IFN-inducible chemokine score over time in SSc patients. The fact that the IFN chemokine score did not show a consistent trend of change and that it was not associated with disease duration at the baseline visit indicates that the IFN signature is a stable marker for the more severe subtype of disease rather than a time-dependent immune dysregulation that improves after the initial phase of SSc (102).

Conclusion

There are many distinct immunological and molecular mechanisms that can contribute to pathogenesis and progression of SSc. Dysregulated innate and adaptive immune responses are major contributors to fibrosis and disease severity of SSc. This review summarized a possible role of type I IFN and IFN-inducible genes in pathogenesis of SSc, and provides support for a link between type I IFN and fibrosis in SSc. Potential role of type I IFN or IFN-inducible genes as treatment targets or biomarkers in SSc need to be further explored. A better understanding of the relationship between type I IFN and fibrosis could bring us closer to the ultimate goal of reversing or slowing the fibrotic process and regenerating the normal end organ tissue in SSc.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

Grant support: NIH/NIAMS-K23AR061436 – DoD PR1206877.

References

1. Sapadin AN, Esser AC, Fleischmajer R. Immunopathogenesis of scleroderma – evolving concepts. Mt Sinai J Med (2001) 68(4–5):233–42.

Pubmed Abstract | Pubmed Full Text

2. Mayes MD, Lacey JV Jr, Beebe-Dimmer J, Gillespie BW, Cooper B, Laing TJ, et al. Prevalence, incidence, survival, and disease characteristics of systemic sclerosis in a large US population. Arthritis Rheum (2003) 48(8):2246–55. doi:10.1002/art.11073

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

3. Elhai M, Meune C, Avouac J, Kahan A, Allanore Y. Trends in mortality in patients with systemic sclerosis over 40 years: a systematic review and meta-analysis of cohort studies. Rheumatology (Oxford) (2012) 51(6):1017–26. doi:10.1093/rheumatology/ker269

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

4. Platanias LC. Interferons: laboratory to clinic investigations. Curr Opin Oncol (1995) 7(6):560–5. doi:10.1097/00001622-199511000-00015

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

5. Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells respond to interferons. Annu Rev Biochem (1998) 67:227–64. doi:10.1146/annurev.biochem.67.1.227

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

6. Paucker K, Cantell K, Henle W. Quantitative studies on viral interference in suspended L cells. III. Effect of interfering viruses and interferon on the growth rate of cells. Virology (1962) 17:324–34. doi:10.1016/0042-6822(62)90123-X

CrossRef Full Text

7. Kraling BM, Maul GG, Jimenez SA. Mononuclear cellular infiltrates in clinically involved skin from patients with systemic sclerosis of recent onset predominantly consist of monocytes/macrophages. Pathobiology (1995) 63(1):48–56. doi:10.1159/000163933

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

8. Ishikawa O, Ishikawa H. Macrophage infiltration in the skin of patients with systemic sclerosis. J Rheumatol (1992) 19(8):1202–6.

Pubmed Abstract | Pubmed Full Text

9. Prescott RJ, Freemont AJ, Jones CJ, Hoyland J, Fielding P. Sequential dermal microvascular and perivascular changes in the development of scleroderma. J Pathol (1992) 166(3):255–63. doi:10.1002/path.1711660307

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

10. Roumm AD, Whiteside TL, Medsger TA Jr., Rodnan GP. Lymphocytes in the skin of patients with progressive systemic sclerosis. Quantification, subtyping, and clinical correlations. Arthritis Rheum (1984) 27(6):645–53. doi:10.1002/art.1780270607

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

11. Fiocco U, Rosada M, Cozzi L, Ortolani C, De Silvestro G, Ruffatti A, et al. Early phenotypic activation of circulating helper memory T cells in scleroderma: correlation with disease activity. Ann Rheum Dis (1993) 52(4):272–7. doi:10.1136/ard.52.4.272

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

12. Higashi-Kuwata N, Makino T, Inoue Y, Takeya M, Ihn H. Alternatively activated macrophages (M2 macrophages) in the skin of patient with localized scleroderma. Exp Dermatol (2009) 18(8):727–9. doi:10.1111/j.1600-0625.2008.00828.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

13. Andrews BS, Friou GJ, Berman MA, Sandborg CI, Mirick GR, Cesario TC. Changes in circulating monocytes in patients with progressive systemic sclerosis. J Rheumatol (1987) 14(5):930–5.

Pubmed Abstract | Pubmed Full Text

14. Anders HJ, Ryu M. Renal microenvironments and macrophage phenotypes determine progression or resolution of renal inflammation and fibrosis. Kidney Int (2011) 80(9):915–25. doi:10.1038/ki.2011.217

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

15. Gordon S. Alternative activation of macrophages. Nat Rev Immunol (2003) 3(1):23–35. doi:10.1038/nri978

CrossRef Full Text

16. Martinez FO, Gordon S, Locati M, Mantovani A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol (2006) 177(10):7303–11.

Pubmed Abstract | Pubmed Full Text

17. Higashi-Kuwata N, Jinnin M, Makino T, Fukushima S, Inoue Y, Muchemwa FC, et al. Characterization of monocyte/macrophage subsets in the skin and peripheral blood derived from patients with systemic sclerosis. Arthritis Res Ther (2010) 12(4):R128. doi:10.1186/ar3066

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

18. Nakayama W, Jinnin M, Makino K, Kajihara I, Makino T, Fukushima S, et al. Serum levels of soluble CD163 in patients with systemic sclerosis. Rheumatol Int (2012) 32(2):403–7. doi:10.1007/s00296-010-1691-z

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

19. Yukawa S, Yamaoka K, Sawamukai N, Shimajiri S, Kubo S, Miyagawa I, et al. Dermal mast cell density in fingers reflects severity of skin sclerosis in systemic sclerosis. Mod Rheumatol (2012):doi:10.1007/s10165-012-0813-8

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

20. Mason GI, Hamburger J, Matthews JB. Mast cells, extracellular matrix components, TGFbeta isoforms and TGFbeta receptor expression in labial salivary glands in systemic sclerosis. Ann Rheum Dis (2000) 59(3):183–9. doi:10.1136/ard.59.3.183

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

21. Hugle T, Hogan V, White KE, van Laar JM. Mast cells are a source of transforming growth factor beta in systemic sclerosis. Arthritis Rheum (2011) 63(3):795–9. doi:10.1002/art.30190

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

22. de Paulis PA, Valentini G, Spadaro G, Lupoli S, Tirri G, Marone G. Human basophil releasability. VIII. Increased basophil releasability in patients with scleroderma. Arthritis Rheum (1991) 34(10):1289–96. doi:10.1002/art.1780341013

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

23. Riccieri V, Parisi G, Spadaro A, Scrivo R, Barone F, Moretti T, et al. Reduced circulating natural killer T cells and gamma/delta T cells in patients with systemic sclerosis. J Rheumatol (2005) 32(2):283–6.

Pubmed Abstract | Pubmed Full Text

24. Rueda B, Broen J, Simeon C, Hesselstrand R, Diaz B, Suarez H, et al. The STAT4 gene influences the genetic predisposition to systemic sclerosis phenotype. Hum Mol Genet (2009) 18(11):2071–7. doi:10.1093/hmg/ddp119

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

25. Gourh P, Agarwal SK, Divecha D, Assassi S, Paz G, Arora-Singh RK, et al. Polymorphisms in TBX21 and STAT4 increase the risk of systemic sclerosis: evidence of possible gene-gene interaction and alterations in Th1/Th2 cytokines. Arthritis Rheum (2009) 60(12):3794–806. doi:10.1002/art.24958

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

26. Radstake TR, Gorlova O, Rueda B, Martin JE, Alizadeh BZ, Palomino-Morales R, et al. Genome-wide association study of systemic sclerosis identifies CD247 as a new susceptibility locus. Nat Genet (2010) 42(5):426–9. doi:10.1038/ng.565

CrossRef Full Text

27. Dawidowicz K, Dieude P, Avouac J, Wipff J, Hachulla E, Diot E, et al. Association study of B-cell marker gene polymorphisms in European caucasian patients with systemic sclerosis. Clin Exp Rheumatol (2011) 29(5):839–42.

Pubmed Abstract | Pubmed Full Text

28. Coustet B, Dieude P, Guedj M, Bouaziz M, Avouac J, Ruiz B, et al. C8orf13-BLK is a genetic risk locus for systemic sclerosis and has additive effects with BANK1: results from a large French cohort and meta-analysis. Arthritis Rheum (2011) 63(7):2091–6. doi:10.1002/art.30379

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

29. Gourh P, Agarwal SK, Martin E, Divecha D, Rueda B, Bunting H, et al. Association of the C8orf13-BLK region with systemic sclerosis in North-American and European populations. J Autoimmun (2010) 34(2):155–62. doi:10.1016/j.jaut.2009.08.014

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

30. Ito I, Kawaguchi Y, Kawasaki A, Hasegawa M, Ohashi J, Kawamoto M, et al. Association of the FAM167A-BLK region with systemic sclerosis. Arthritis Rheum (2010) 62(3):890–5. doi:10.1002/art.27303

CrossRef Full Text

31. Dieude P, Wipff J, Guedj M, Ruiz B, Melchers I, Hachulla E, et al. BANK1 is a genetic risk factor for diffuse cutaneous systemic sclerosis and has additive effects with IRF5 and STAT4. Arthritis Rheum (2009) 60(11):3447–54. doi:10.1002/art.24885

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

32. Rueda B, Gourh P, Broen J, Agarwal SK, Simeon C, Ortego-Centeno N, et al. BANK1 functional variants are associated with susceptibility to diffuse systemic sclerosis in caucasians. Ann Rheum Dis (2010) 69(4):700–5. doi:10.1136/ard.2009.118174

CrossRef Full Text

33. Sharif R, Mayes MD, Tan FK, Gorlova OY, Hummers LK, Shah AA, et al. IRF5 polymorphism predicts prognosis in patients with systemic sclerosis. Ann Rheum Dis (2012) 71(7):1197–202. doi:10.1136/annrheumdis-2011-200901

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

34. Dieude P, Guedj M, Wipff J, Avouac J, Fajardy I, Diot E, et al. Association between the IRF5 rs2004640 functional polymorphism and systemic sclerosis: a new perspective for pulmonary fibrosis. Arthritis Rheum (2009) 60(1):225–33. doi:10.1002/art.24183

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

35. Carmona FD, Gutala R, Simeon CP, Carreira P, Ortego-Centeno N, Vicente-Rabaneda E, et al. Novel identification of the IRF7 region as an anticentromere autoantibody propensity locus in systemic sclerosis. Ann Rheum Dis (2012) 71(1):114–9. doi:10.1136/annrheumdis-2011-200275

CrossRef Full Text

36. Gorlova O, Martin JE, Rueda B, Koeleman BP, Ying J, Teruel M, et al. Identification of novel genetic markers associated with clinical phenotypes of systemic sclerosis through a genome-wide association strategy. PLoS Genet (2011) 7(7):e1002178. doi:10.1371/journal.pgen.1002178

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

37. Terao C, Ohmura K, Kawaguchi Y, Nishimoto T, Kawasaki A, Takehara K, et al. PLD4 as a novel susceptibility gene for systemic sclerosis in a Japanese population. Arthritis Rheum (2013) 65(2):472–80. doi:10.1002/art.37777

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

38. Martin JE, Broen JC, Carmona FD, Teruel M, Simeon CP, Vonk MC, et al. Identification of CSK as a systemic sclerosis genetic risk factor through genome wide association study follow-up. Hum Mol Genet (2012) 21(12):2825–35. doi:10.1093/hmg/dds099

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

39. Farina GA, York MR, Di MM, Collins CA, Meller S, Homey B, et al. Poly(I:C) drives type I IFN- and TGFbeta-mediated inflammation and dermal fibrosis simulating altered gene expression in systemic sclerosis. J Invest Dermatol (2010) 130(11):2583–93. doi:10.1038/jid.2010.200

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

40. Tan FK, Zhou X, Mayes MD, Gourh P, Guo X, Marcum C, et al. Signatures of differentially regulated interferon gene expression and vasculotrophism in the peripheral blood cells of systemic sclerosis patients. Rheumatology (Oxford) (2006) 45(6):694–702. doi:10.1093/rheumatology/kei244

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

41. York MR, Nagai T, Mangini AJ, Lemaire R, van Seventer JM, Lafyatis R. A macrophage marker, Siglec-1, is increased on circulating monocytes in patients with systemic sclerosis and induced by type I interferons and toll-like receptor agonists. Arthritis Rheum (2007) 56(3):1010–20. doi:10.1002/art.22382

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

42. Duan H, Fleming J, Pritchard DK, Amon LM, Xue J, Arnett HA, et al. Combined analysis of monocyte and lymphocyte messenger RNA expression with serum protein profiles in patients with scleroderma. Arthritis Rheum (2008) 58(5):1465–74. doi:10.1002/art.23451

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

43. Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, et al. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis (2011) 70(11):2029–36. doi:10.1136/ard.2011.150326

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

44. Merrill JT, Wallace DJ, Petri M, Kirou KA, Yao Y, White WI, et al. Safety profile and clinical activity of sifalimumab, a fully human anti-interferon alpha monoclonal antibody, in systemic lupus erythematosus: a phase I, multicentre, double-blind randomised study. Ann Rheum Dis (2011) 70(11):1905–13. doi:10.1136/ard.2010.144485

CrossRef Full Text

45. Petri M, Wallace DJ, Spindler A, Chindalore V, Kalunian K, Mysler E, et al. Sifalimumab, a human anti-interferon-alpha monoclonal antibody, in systemic lupus erythematosus: a phase I randomized, controlled, dose-escalation study. Arthritis Rheum (2013) 65(4):1011–21. doi:10.1002/art.37824

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

46. Assassi S, Mayes MD, Arnett FC, Gourh P, Agarwal SK, McNearney TA, et al. Systemic sclerosis and lupus: points in an interferon-mediated continuum. Arthritis Rheum (2010) 62(2):589–98. doi:10.1002/art.27224

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

47. Eloranta ML, Franck-Larsson K, Lovgren T, Kalamajski S, Ronnblom A, Rubin K, et al. Type I interferon system activation and association with disease manifestations in systemic sclerosis. Ann Rheum Dis (2010) 69(7):1396–402. doi:10.1136/ard.2009.121400

CrossRef Full Text

48. Kim D, Peck A, Santer D, Patole P, Schwartz SM, Molitor JA, et al. Induction of interferon-alpha by scleroderma sera containing autoantibodies to topoisomerase I: association of higher interferon-alpha activity with lung fibrosis. Arthritis Rheum (2008) 58(7):2163–73. doi:10.1002/art.23486

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

49. Solans R, Bosch JA, Esteban I, Vilardell M. Systemic sclerosis developing in association with the use of interferon alpha therapy for chronic viral hepatitis. Clin Exp Rheumatol (2004) 22(5):625–8.

Pubmed Abstract | Pubmed Full Text

50. Black CM, Silman AJ, Herrick AI, Denton CP, Wilson H, Newman J, et al. Interferon-alpha does not improve outcome at one year in patients with diffuse cutaneous scleroderma: results of a randomized, double-blind, placebo-controlled trial. Arthritis Rheum (1999) 42(2):299–305. doi:10.1002/1529-0131(199902)42:2<299::AID-ANR12>3.0.CO;2-R

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

51. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell (2006) 124(4):783–801. doi:10.1016/j.cell.2006.02.015

CrossRef Full Text

52. Hua J, Kirou K, Lee C, Crow MK. Functional assay of type I interferon in systemic lupus erythematosus plasma and association with anti-RNA binding protein autoantibodies. Arthritis Rheum (2006) 54(6):1906–16. doi:10.1002/art.21890

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

53. Van der Pouw Kraan TC, Wijbrandts CA, van Baarsen LG, Voskuyl AE, Rustenburg F, Baggen JM, et al. Rheumatoid arthritis subtypes identified by genomic profiling of peripheral blood cells: assignment of a type I interferon signature in a subpopulation of patients. Ann Rheum Dis (2007) 66(8):1008–14. doi:10.1136/ard.2006.063412

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

54. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on toll-like receptors. Nat Immunol (2010) 11(5):373–84. doi:10.1038/ni.1863

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

55. Bhattacharyya S, Kelley K, Melichian DS, Tamaki Z, Fang F, Su Y, et al. Toll-like receptor 4 signaling augments transforming growth factor-beta responses: a novel mechanism for maintaining and amplifying fibrosis in scleroderma. Am J Pathol (2013) 182(1):192–205. doi:10.1016/j.ajpath.2012.09.007

CrossRef Full Text

56. Agarwal SK, Wu M, Livingston CK, Parks DH, Mayes MD, Arnett FC, et al. Toll-like receptor 3 upregulation by type I interferon in healthy and scleroderma dermal fibroblasts. Arthritis Res Ther (2011) 13(1):R3. doi:10.1186/ar3221

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

57. Tamura T, Yanai H, Savitsky D, Taniguchi T. The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol (2008) 26:535–84. doi:10.1146/annurev.immunol.26.021607.090400

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

58. Mamane Y, Heylbroeck C, Genin P, Algarte M, Servant MJ, LePage C, et al. Interferon regulatory factors: the next generation. Gene (1999) 237(1):1–14. doi:10.1016/S0378-1119(99)00262-0

CrossRef Full Text

59. Taniguchi T, Ogasawara K, Takaoka A, Tanaka N. IRF family of transcription factors as regulators of host defense. Annu Rev Immunol (2001) 19:623–55. doi:10.1146/annurev.immunol.19.1.623

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

60. Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol (2005) 5(5):375–86. doi:10.1038/nri1604

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

61. Decker T, Muller M, Stockinger S. The yin and yang of type I interferon activity in bacterial infection. Nat Rev Immunol (2005) 5(9):675–87. doi:10.1038/nri1684

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

62. Barnes BJ, Richards J, Mancl M, Hanash S, Beretta L, Pitha PM. Global and distinct targets of IRF-5 and IRF-7 during innate response to viral infection. J Biol Chem (2004) 279(43):45194–207. doi:10.1074/jbc.M400726200

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

63. Barnes BJ, Kellum MJ, Pinder KE, Frisancho JA, Pitha PM. Interferon regulatory factor 5, a novel mediator of cell cycle arrest and cell death. Cancer Res (2003) 63(19):6424–31.

Pubmed Abstract | Pubmed Full Text

64. Barnes BJ, Moore PA, Pitha PM. Virus-specific activation of a novel interferon regulatory factor, IRF-5, results in the induction of distinct interferon alpha genes. J Biol Chem (2001) 276(26):23382–90. doi:10.1074/jbc.M101216200

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

65. Honda K, Taniguchi T. IRFs: master regulators of signalling by toll-like receptors and cytosolic pattern-recognition receptors. Nat Rev Immunol (2006) 6(9):644–58. doi:10.1038/nri1900

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

66. Takaoka A, Yanai H, Kondo S, Duncan G, Negishi H, Mizutani T, et al. Integral role of IRF-5 in the gene induction programme activated by toll-like receptors. Nature (2005) 434(7030):243–9. doi:10.1038/nature03308

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

67. Dieude P, Guedj M, Wipff J, Ruiz B, Hachulla E, Diot E, et al. STAT4 is a genetic risk factor for systemic sclerosis having additive effects with IRF5 on disease susceptibility and related pulmonary fibrosis. Arthritis Rheum (2009) 60(8):2472–9. doi:10.1002/art.24688

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

68. Ito I, Kawaguchi Y, Kawasaki A, Hasegawa M, Ohashi J, Hikami K, et al. Association of a functional polymorphism in the IRF5 region with systemic sclerosis in a Japanese population. Arthritis Rheum (2009) 60(6):1845–50. doi:10.1002/art.24600

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

69. Dieude P, Dawidowicz K, Guedj M, Legrain Y, Wipff J, Hachulla E, et al. Phenotype-haplotype correlation of IRF5 in systemic sclerosis: role of 2 haplotypes in disease severity. J Rheumatol (2010) 37(5):987–92. doi:10.3899/jrheum.091163

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

70. Dietrich N, Lienenklaus S, Weiss S, Gekara NO. Murine toll-like receptor 2 activation induces type I interferon responses from endolysosomal compartments. PLoS One (2010) 5(4):e10250. doi:10.1371/journal.pone.0010250

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

71. Sato M, Taniguchi T, Tanaka N. The interferon system and interferon regulatory factor transcription factors – studies from gene knockout mice. Cytokine Growth Factor Rev (2001) 12(2–3):133–42. doi:10.1016/S1359-6101(00)00032-0

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

72. Honda K, Yanai H, Negishi H, Asagiri M, Sato M, Mizutani T, et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature (2005) 434(7034):772–7. doi:10.1038/nature03464

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

73. Wathelet MG, Lin CH, Parekh BS, Ronco LV, Howley PM, Maniatis T. Virus infection induces the assembly of coordinately activated transcription factors on the IFN-beta enhancer in vivo. Mol Cell (1998) 1(4):507–18. doi:10.1016/S1097-2765(00)80051-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

74. Yang H, Lin CH, Ma G, Baffi MO, Wathelet MG. Interferon regulatory factor-7 synergizes with other transcription factors through multiple interactions with p300/CBP coactivators. J Biol Chem (2003) 278(18):15495–504. doi:10.1074/jbc.M212940200

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

75. Yasuda K, Richez C, Maciaszek JW, Agrawal N, Akira S, Marshak-Rothstein A, 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 (2007) 178(11):6876–85.

Pubmed Abstract | Pubmed Full Text

76. Sweeney SE. Targeting interferon regulatory factors to inhibit activation of the type I IFN response: implications for treatment of autoimmune disorders. Cell Immunol (2011) 271(2):342–9. doi:10.1016/j.cellimm.2011.07.014

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

77. Harley JB, Alarcon-Riquelme ME, Criswell LA, Jacob CO, Kimberly RP, Moser KL, et al. Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nat Genet (2008) 40(2):204–10. doi:10.1038/ng.81

CrossRef Full Text

78. Kawasaki A, Furukawa H, Kondo Y, Ito S, Hayashi T, Kusaoi M, et al. Association of PHRF1-IRF7 region polymorphism with clinical manifestations of systemic lupus erythematosus in a Japanese population. Lupus (2012) 21(8):890–5. doi:10.1177/0961203312439333

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

79. Lin LH, Ling P, Liu MF. The potential role of interferon-regulatory factor 7 among Taiwanese patients with systemic lupus erythematosus. J Rheumatol (2011) 38(9):1914–9. doi:10.3899/jrheum.101004

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

80. Fu Q, Zhao J, Qian X, Wong JL, Kaufman KM, Yu CY, et al. Association of a functional IRF7 variant with systemic lupus erythematosus. Arthritis Rheum (2011) 63(3):749–54. doi:10.1002/art.30193

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

81. De Oliveira DB, Almeida GM, Guedes AC, Santos FP, Bonjardim CA, Ferreira PC, et al. Basal activation of type I interferons (alpha2 and beta) and 2’5’OAS genes: insights into differential expression profiles of interferon system components in systemic sclerosis. Int J Rheumatol (2011) 2011:275617. doi:10.1155/2011/275617

CrossRef Full Text

82. Zhao J, Kong HJ, Li H, Huang B, Yang M, Zhu C, et al. IRF-8/interferon (IFN) consensus sequence-binding protein is involved in toll-like receptor (TLR) signaling and contributes to the cross-talk between TLR and IFN-gamma signaling pathways. J Biol Chem (2006) 281(15):10073–80. doi:10.1074/jbc.M507788200

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

83. Tailor P, Tamura T, Kong HJ, Kubota T, Kubota M, Borghi P, et al. The feedback phase of type I interferon induction in dendritic cells requires interferon regulatory factor 8. Immunity (2007) 27(2):228–39. doi:10.1016/j.immuni.2007.06.009

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

84. Hambleton S, Salem S, Bustamante J, Bigley V, Boisson-Dupuis S, Azevedo J, et al. IRF8 mutations and human dendritic-cell immunodeficiency. N Engl J Med (2011) 365(2):127–38. doi:10.1056/NEJMoa1100066

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

85. Lu R, Medina KL, Lancki DW, Singh H. IRF-4,8 orchestrate the pre-B-to-B transition in lymphocyte development. Genes Dev (2003) 17(14):1703–8. doi:10.1101/gad.1104803

CrossRef Full Text

86. Whitfield ML, Finlay DR, Murray JI, Troyanskaya OG, Chi JT, Pergamenschikov A, et al. Systemic and cell type-specific gene expression patterns in scleroderma skin. Proc Natl Acad Sci U S A (2003) 100(21):12319–24. doi:10.1073/pnas.1635114100

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

87. Lafyatis R, Kissin E, York M, Farina G, Viger K, Fritzler MJ, et al. B cell depletion with rituximab in patients with diffuse cutaneous systemic sclerosis. Arthritis Rheum (2009) 60(2):578–83. doi:10.1002/art.24249

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

88. Smith V, Van Praet JT, Vandooren B, Van der Cruyssen B, Naeyaert JM, Decuman S, et al. Rituximab in diffuse cutaneous systemic sclerosis: an open-label clinical and histopathological study. Ann Rheum Dis (2010) 69(1):193–7. doi:10.1136/ard.2008.095463

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

89. Sfrent-Cornateanu R, Mihai C, Balan S, Ionescu R, Moldoveanu E. The IL-6 promoter polymorphism is associated with disease activity and disability in systemic sclerosis. J Cell Mol Med (2006) 10(4):955–9. doi:10.2755/jcmm010.004.08

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

90. Lee YH, Lee HS, Choi SJ, Ji JD, Song GG. The association between interleukin-6 polymorphisms and systemic lupus erythematosus: a meta-analysis. Lupus (2012) 21(1):60–7. doi:10.1177/0961203311422711

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

91. Madhok R, Crilly A, Watson J, Capell HA. Serum interleukin 6 levels in rheumatoid arthritis: correlations with clinical and laboratory indices of disease activity. Ann Rheum Dis (1993) 52(3):232–4. doi:10.1136/ard.52.3.232

CrossRef Full Text

92. Kishimoto T, Hirano T. Molecular regulation of B lymphocyte response. Annu Rev Immunol (1988) 6:485–512. doi:10.1146/annurev.iy.06.040188.002413

CrossRef Full Text

93. Hirano T. Interleukin-6 and its relation to inflammation and disease. Clin Immunol Immunopathol (1992) 62(1 Pt 2):S60–5. doi:10.1016/0090-1229(92)90042-M

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

94. Qiu Z, Fujimura M, Kurashima K, Nakao S, Mukaida N. Enhanced airway inflammation and decreased subepithelial fibrosis in interleukin 6-deficient mice following chronic exposure to aerosolized antigen. Clin Exp Allergy (2004) 34(8):1321–8. doi:10.1111/j.1365-2222.2004.02013.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

95. Saito F, Tasaka S, Inoue K, Miyamoto K, Nakano Y, Ogawa Y, et al. Role of interleukin-6 in bleomycin-induced lung inflammatory changes in mice. Am J Respir Cell Mol Biol (2008) 38(5):566–71. doi:10.1165/rcmb.2007-0299OC

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

96. Hasegawa M, Sato S, Ihn H, Takehara K. Enhanced production of interleukin-6 (IL-6), oncostatin M and soluble IL-6 receptor by cultured peripheral blood mononuclear cells from patients with systemic sclerosis. Rheumatology (Oxford) (1999) 38(7):612–7. doi:10.1093/rheumatology/38.7.612

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

97. De Lauretis A, Sestini P, Pantelidis P, Hoyles R, Hansell DM, Goh NS, et al. Serum interleukin 6 is predictive of early functional decline and mortality in interstitial lung disease associated with systemic sclerosis. J Rheumatol (2013) 40(4):435–46. doi:10.3899/jrheum.120725

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

98. Laurence A, O’Shea JJ. T(H)-17 differentiation: of mice and men. Nat Immunol (2007) 8(9):903–5. doi:10.1038/ni0907-903

CrossRef Full Text

99. Glimcher LH, Murphy KM. Lineage commitment in the immune system: the T helper lymphocyte grows up. Genes Dev (2000) 14(14):1693–711.

100. Kondo K, Okada T, Matsui T, Kato S, Date K, Yoshihara M, et al. Establishment and characterization of a human B cell line from the lung tissue of a patient with scleroderma; extraordinary high level of IL-6 secretion by stimulated fibroblasts. Cytokine (2001) 13(4):220–6. doi:10.1006/cyto.2000.0822

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

101. Duncan MR, Berman B. Stimulation of collagen and glycosaminoglycan production in cultured human adult dermal fibroblasts by recombinant human interleukin 6. J Invest Dermatol (1991) 97(4):686–92. doi:10.1111/1523-1747.ep12483971

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

102. Liu X, Mayes MD, Tan FK, Wu M, Reveille JD, Harper BE, et al. Correlation of interferon-inducible chemokine plasma levels with disease severity in systemic sclerosis. Arthritis Rheum (2013) 65(1):226–35. doi:10.1002/art.37742

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Keywords: systemic sclerosis, innate immunity, type 1 IFN, interferon regulatory factor, IFN-inducible cytokines and chemokines

Citation: Wu M and Assassi S (2013) The role of type 1 interferon in systemic sclerosis. Front. Immunol. 4:266. doi: 10.3389/fimmu.2013.00266

Received: 15 June 2013; Paper pending published: 10 July 2013;
Accepted: 19 August 2013; Published online: 06 September 2013.

Edited by:

Timothy B. Niewold, Mayo Clinic, USA

Reviewed by:

Janos G. Filep, University of Montreal, Canada
Annapurna Nayak, Brunel University, UK

Copyright: © 2013 Wu and Assassi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Minghua Wu, Division of Rheumatology and Clinical Immunogenetics, Department of Internal Medicine, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA e-mail: minghua.wu@uth.tmc.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.