Skip to main content
Erschienen in: Arthritis Research & Therapy 2/2014

Open Access 01.04.2014 | Research article

T-helper 17 cell cytokines and interferon type I: partners in crime in systemic lupus erythematosus?

verfasst von: Zana Brkic, Odilia BJ Corneth, Cornelia G van Helden-Meeuwsen, Radboud JEM Dolhain, Naomi I Maria, Sandra MJ Paulissen, Nadine Davelaar, Jan Piet van Hamburg, Paul L van Daele, Virgil A Dalm, P Martin van Hagen, Johanna MW Hazes, Marjan A Versnel, Erik Lubberts

Erschienen in: Arthritis Research & Therapy | Ausgabe 2/2014

Abstract

Introduction

A hallmark of systemic autoimmune diseases like systemic lupus erythematosus (SLE) is the increased expression of interferon (IFN) type I inducible genes, so-called IFN type I signature. Recently, T-helper 17 subset (Th17 cells), which produces IL-17A, IL-17F, IL-21, and IL-22, has been implicated in SLE. As CCR6 enriches for Th17 cells, we used this approach to investigate whether CCR6+ memory T-helper cells producing IL-17A, IL-17F, IL-21, and/or IL-22 are increased in SLE patients and whether this increase is related to the presence of IFN type I signature.

Methods

In total, 25 SLE patients and 15 healthy controls (HCs) were included. SLE patients were divided into IFN type I signature-positive (IFN+) (n = 16) and negative (IFN-) (n = 9) patients, as assessed by mRNA expression of IFN-inducible genes (IFIGs) in monocytes. Expression of IL-17A, IL-17F, IL-21, and IL-22 by CD4+CD45RO+CCR6+ T cells (CCR6+ cells) was measured with flow cytometry and compared between IFN+, IFN- patients and HCs.

Results

Increased percentages of IL-17A and IL-17A/IL-17F double-producing CCR6+ cells were observed in IFN+ patients compared with IFN- patients and HCs. IL-17A and IL-17F expression within CCR6+ cells correlated significantly with IFIG expression. In addition, we found significant correlation between B-cell activating factor of the tumor necrosis family (BAFF)–a factor strongly correlating with IFN type I - and IL-21 producing CCR6+ cells.

Conclusions

We show for the first time higher percentages of IL-17A and IL-17A/IL-17F double-producing CCR6+ memory T-helper cells in IFN+ SLE patients, supporting the hypothesis that IFN type I co-acts with Th17 cytokines in SLE pathogenesis.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​ar4499) contains supplementary material, which is available to authorized users.
Zana Brkic, Odilia BJ Corneth, Marjan A Versnel contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

ZB and OBJC were involved in the study design and clinical and laboratory data collection, analyzed the data, and drafted and revised the manuscript. CGvH-M, NIM, SMJP, and ND were involved in laboratory data collection and revising the draft article. RJEMD, PLvD,VAD, PMvH, JMWH were involved in collection of clinical data and revising the draft. JPvH was involved in the design of the study and helped to draft the manuscript. MAV and EL were involved in study design, monitoring of data collection, and final draft and approval of the manuscript. All authors read and approved the final manuscript.
Abkürzungen
BAFF
B cell-activating factor of the tumor necrosis family
CCR6
chemokine (C-C motif) receptor 6
EAE
experimental autoimmune encephalomyelitis
HC
healthy control
IFIG
IFN-inducible gene
IFN
interferon
IL
interleukin
IRF
IFN-regulatory factor
MS
multiple sclerosis, pSS, primary Sjögren syndrome
NETs
neutrophil extracellular traps
pDC
plasmacytoid dendritic cell
SLE
systemic lupus erythematosus
SLEDAI
Systemic Lupus Erythematosus Disease Activity Index
STAT
signal transducer and activator of transcription
Tfh
T-follicular helper
Th
T helper
TLR
Toll-like receptor.

Introduction

Systemic lupus erythematosus (SLE) is a debilitating systemic autoimmune disease characterized by the production of autoreactive antibodies and multiorgan inflammation [1]. A hallmark of systemic autoimmune diseases is the increased expression of interferon (IFN) type I in both blood and disease-affected tissues [2]. About half of the SLE patients exhibit an IFN type I signature or upregulation of IFN type I-induced genes (IFIGs), which have been found to correlate with disease activity and severity [35].
Another key factor in the pathogenesis of SLE, apart from IFN type I, is interleukin-17A (IL-17A) [69]. IL-17A is produced by several immune cell types, including CD4+ T cells (Th17 cells), CD8+ T cells, CD4-CD8-CD3+ (double-negative, DN) T cells, natural killer cells, γδ-T cells, and mast cells [10, 11]. Naïve CD4+ T cells differentiate to Th17 cells under the influence of IL-6 and TGF-β [12]. The expansion and stability of the Th17 population is regulated by IL-21 and IL-23, respectively [13, 14]. C57BL/6-lpr/lpr mice that lack IL-23 receptor signaling are protected for SLE development [15]. In SLE patients, increased plasma levels of IL-17A correlate with disease activity (SLEDAI) [6]. In addition, in peripheral blood of SLE patients, an increased number of IL-17-producing cells is observed. These cells correlate with disease activity and decrease with treatment [7, 8]. IL-17-producing cells have also been found in several affected organs of SLE patients [7, 9].
Co-activity between IFN type I and IL-17/Th17 cells has been suggested in autoimmune diseases [16, 17]. In experimental autoimmune encephalomyelitis (EAE), a mouse model for multiple sclerosis (MS), IFN type I treatment caused exacerbation if the disease was Th17 driven but was effective if the disease was Th1 driven [17]. In the same study, MS patients that did not respond to IFN type I therapy had higher serum levels of IL-17A before therapy onset [17]. These two observations suggest additional effects of the IFN type I and Th17 system co-acting in the pathogenesis of autoimmune diseases.
Co-activity of IFN type I and Th17 pathways has also been suggested for SLE by the Ro52/TRIM21-/- mouse model. Ro52/TRIM21 is involved in the ubiquitination of interferon regulatory factors (IRFs), a process that limits the IFN type I response [18]. After ear tagging, Ro52/TRIM21-/- mice develop an SLE-like phenotype [19]. Interestingly, when these mice are crossed on an IL-23p19-/- mouse line, they do not develop SLE, indicating that the development of an SLE phenotype through enhanced IFN type I production in these mice is dependent on the IL-17/Th17 pathway.
Yet another important factor involved in the pathogenesis of SLE is B cell-activating factor of the tumor necrosis factor family (BAFF). BAFF transgenic mice develop lupus-like disease [20], and increased expression of BAFF protein has been found in SLE patients, correlating with increased disease activity [2123]. We previously described a strong correlation between BAFF mRNA expression in monocytes and the IFN type I signature in primary Sjögren syndrome (pSS) patients [24]. Interestingly, IL-21, a cytokine produced by Th17 cells, in combination with BAFF, has been reported to induce synergistically the differentiation of human memory B cells into antibody-producing plasma cells in the absence of further co-stimulation [25]. BAFF is known to be involved in germinal center formation [26], a process in which IL-17 is also involved [27].
The previously mentioned literature suggests an association between the pathogenic IFN type I and Th17 pathways. So far, no studies have been performed on the co-occurrence of these pathogenic pathways in SLE patients. In this study, we report for the first time a higher percentage of IL-17A and IL-17A/F producing CCR6+ T-memory cells in IFN type I-positive SLE patients. Moreover, BAFF gene expression in monocytes correlates significantly with IL-21 expression in these CCR6+ cells, supporting the concept of co-activity of IFN type I, Th17, and BAFF in the pathogenesis of SLE.

Methods

Patients

The 25 patients fulfilling the American College of Rheumatology revised criteria for SLE [28] were recruited at the outpatient clinic of the Immunology Department and the Rheumatology Department of the Erasmus Medical Center Rotterdam. The level of disease activity was assessed by using the SLEDAI [29]. Fifteen healthy controls (HCs), neither with autoimmune diseases nor using corticosteroids, were included. Characteristics of patients and controls are summarized in Table 1. The Medical Ethical Review Committee of the Erasmus MC approved the study, and written informed consent was obtained.
Table 1
Demographics and clinical characteristics of participants
Variable
SLE patients
Healthy controls
 
(n = 25)
(n = 15)
 
IFN type I negative
IFN type I positive
 
 
(n = 9)
(n = 16)
 
Demographics
   
   Number of females, n (%)
9/9 (100%)
14/16 (88%)
15/15 (100%)
   Age (years)
41.3 ± 17.5
39.8 ± 15.7
41.0 ± 14.0
   Race, n (%)
   
   Caucasian
8/9 (89%)
7/16 (44%)
15/15 (100%)
   Black
0/9 (0)
8/16 (50%)
0/15 (0)
   Asian
0/9 (0)
0/16 (0)
0/15 (0)
   Mixed race
1/9 (11%)
1/16 (6%)
0/15 (0)
Clinical
   
   Disease duration (years)
12.1 ± 8.0
14.4 ± 11.3
-
   Mucocutaneous, n (%)
3/9 (33%)
10/16 (63%)
-
   Arthritis, n (%)
7/9 (78%)
6/16 (38%)
-
   Serositis, n (%)
1/9 (11%)
3/16 (19%)
-
   Nephritis, n (%)
3/9 (33%)
9/16 (56%)
-
   Neuropsychiatric, n (%)
1/9 (11%)
3/16 (19%)
-
   Hematologic, n (%)
6/9 (67%)
12/16 (75%)
-
Laboratory
   
   ANA positivity, n (%)
9/9 (100%)
16/16 (100%)
-
   Anti-ds-DNA positivity, n (%)
5/9 (56%)
10/16 (63%)
-
Treatment
   
   Hydroxychloroquine, n (%)
7/9 (78%)
11/16 (69%)
-
   Corticosteroids, n (%)
5/9 (56%)
10/16 (63%)
-
   Mycophenolate mofetil, n (%)
0/9 (0)
4/16 (25%)
-
   Azathioprine, n (%)
2/9 (22%)
3/16 (19%)
-
   Cyclophosphamide, n (%)
0/9 (0)
1/16 (6%)
-
Values are the mean ± SD, median (25% quartile,75% quartile) or number (%) of patients, depending on whether the data are continuous or dichotomous, and whether the data are normally distributed or not.

Blood collection and isolation of monocytes

Blood was collected in clotting tubes for serum preparation (stored at -80°C) and in sodium-heparin tubes for peripheral blood mononuclear cell (PBMC) preparation, as described previously [30]. CD14-positive monocytes were isolated as described [30].

RQ-PCR

Total RNA was isolated from purified monocytes followed by cDNA preparation and real-time quantitative polymerase chain reaction (RQ-PCR) analysis by using predesigned primer/probe sets (Applied Biosystems) [30]. For calculation of relative expression, all samples were normalized against expression of the household gene Abl [31]. Fold-change values were determined from normalized CT values by using the 2-ΔΔCT method (User Bulletin; Applied Biosystems, Foster City, CA, USA).

Flow cytometry

PBMCs were restimulated, stained, and measured with flow cytometry, as previously described [32]. For extracellular staining, CD4, CD45RO, and CCR6 monoclonal antibodies were obtained from BD Biosciences (San Diego, CA, USA), and CD25 antibodies from Biolegend Inc. (San Diego, CA, USA). For intracellular staining, IL-17A, IL-17F, IL-21, and IL-22 monoclonal antibodies were obtained from eBioscience, and IL-17A monoclonal antibodies from Biolegend Inc. Samples were measured on a FACScantoII flow cytometer (BD Biosciences). Analysis was performed by using FlowJo v7.6 research software (Tree Star Inc. Ashland, OR, USA).

Factor analysis

The expression levels of 11 IFN type I-inducible genes (previously found to be increased in SLE) were submitted to a principal component analysis to identify correlated groups of genes to reduce data complexity. Kaiser-Meyer-Olkin measure of sampling adequacy was 0.839 with a significant Bartlett test of sphericity (P < 0.001). Eigenvalues were derived to assess the amount of variance explained by each component factor.

Statistical analyses

Statistical analyses were performed by using the SPSS 20.0 package. When data were not normally distributed, values were expressed as medians with interquartile ranges (IQRs), and comparisons were made by using the nonparametric Mann–Whitney U test. In case of more than two samples, the nonparametric Kruskal-Wallis test was performed. Correlations were assessed by using either the Pearson correlation test for normally distributed data or Spearman rho when data were not normally distributed. Differences were considered statistically significant if P < 0.05.

Results

Prevalence of the IFN type I signature in SLE patients

In monocytes of 25 SLE patients and 15 HCs, we assessed the expression levels of 11 IFIGs previously assessed in monocytes from patients with primary Sjögren syndrome (pSS) (IFI27, IFI44L, IFIT3, IFITM1, SERPING1, IFIT1, IFIT2, LY6E, IFI44, XAF1, and MXA) [24], and the expression of which was also found to be increased in SLE patients [2, 3, 33, 34]. To reduce data complexity, expression levels of the 11 genes were submitted to a principal component analysis to identify correlated groups of genes. The results of the principal component analysis identified a subset of four genes (IFI44L, IFITM1, SERPING1, and LY6E) to explain 95% of the total variance of the 11 IFN type I-inducible genes within the SLE cohort. Given that the expression of these four IFN type I-inducible genes was not normally distributed, log transformations of expression values were performed, and IFN scores were calculated as described for pSS [24]. MeanHC and SDHC of each gene in the HC-group were used to standardize expression levels. IFN scores per subject represent the sum of these standardized scores. When we set the threshold for a positive IFN type I signature at IFN score of 8 [24], 64% of SLE patients displayed an IFN type I signature, and one of the 15 HC subjects (7%) (Figure 1A,B).

SLE patients with IFN type I signature show higher percentages of IL-17A, IL-17F, and IL-21 producing CCR6+ cells

Because CCR6 enriches for Th17 cells [3537], CCR6+ cells were selected after gating on lymphocytes and memory Th cells (CD4+CD45RO+ cells) within PBMCs and after CD25+ cells were excluded. To investigate whether the IFN type I signature is associated with an increase in Th17 cytokines expressed by memory CCR6+ T cells, we measured the percentages of IL-17A, IL-17F, IL-22 and IL-21 producing CCR6+ T memory cells in SLE patients positive for the IFN type I signature (IFN+) and patients negative for the signature (IFN-) and HC. The percentages of CCR6+ cells within the CD4+CD45RO+ T-cell population and within total lymphocytes were comparable between the three studied groups (data not shown). Interestingly, the percentages of CCR6+IL-17A+ cells were significantly increased in IFN+ patients, as compared with IFN- patients (P = 0.03), and a higher trend was observed compared with HCs (P = 0.07) (Figure 2A). The percentages of CCR6+IL-17F+ and, in particular, the IL-17A/IL-17F double producers were significantly increased in the IFN+ group compared with HC (P = 0.009) (Figure 2B, and see Additional file 1: Figure S1). The percentages of CCR6+IL-22+ cells showed a higher trend for IFN+ versus IFN- patients (P = 0.06) and IFN+ patients versus HC (P = 0.09) (Figure 2C). The percentages of IL-21 expressing CCR6+ cells were also significantly increased in IFN+ patients compared with IFN- patients (Figure 2D). These data suggest an association between the presence of the IFN type I signature and the expression of Th17 cytokines in SLE.
In addition, we investigated whether the Th17 cytokine production is associated with disease activity as assessed by SLEDAI scores. No significant correlations were observed between the SLEDAI scores and IL-17A and/or IL-17F expression (data not shown). Except for race, no differences in demographic, clinical, or laboratory data were found between IFN+, IFN- SLE patients and/or HCs (Table 1).

BAFF mRNA expression is correlated with IL-21 expression within CCR6+ memory T cells

Correlating the expression of IFIGs (as reflected by the total IFN score) with other parameters assessed in this study, we observed a significant positive correlation between the expression of IL-17A and IL-17F within CCR6+ cells and IFIG expression (Figure 3A,B). Also in this SLE cohort, IFIG expression correlated strongly with the BAFF mRNA expression in monocytes (r = 0.527; P < 0.0001) (Figure 3C). No correlation was observed between BAFF and IL-17A and/or IL-17F expression. However, we did find a significant correlation between BAFF mRNA and the percentages of IL-21 producing CCR6+ cells (r = 0.406; P = 0.010) (Figure 3D). Both BAFF and IL-21 are involved in the selection and activation of B cells, which is crucial in the pathogenesis of SLE, indicating that downstream factors of the IFN type I and Th17 pathways might also be associated.

Discussion

Here we show for the first time a co-occurrence of increased IFN type I activity and increased IL-17/Th17 system in SLE patients. We found increased percentages of IL-17A, IL-17A/IL-17F, and IL-21 producing CCR6+ T memory cells in IFN type I-positive SLE patients. This finding further strengthens the hypothesis that IFN type I and Th17 cells, by co-acting, contribute to the pathogenesis of SLE. Further research to understand the link between these two pathways is warranted.
A possible mechanism explaining the co-occurrence of IFN type I and IL-17/Th17 immune pathway in SLE could be that both IFN type I and production of IL-6 and IL-23 by DCs are regulated through IRF-5 [38, 39]. Activation of TLR signaling on DCs will then lead to simultaneous enhancement of both pathways. Evidence suggests that TLR7 activation of plasmacytoid DCs, the main producers of IFN type I, promotes and modifies Th17 cell differentiation and function [40]. IFN type I itself is also able to promote Th17 differentiation and IL-17 production through induction of STAT-3 in T cells and IL-6 in DCs [41, 42]. In addition, IFN type I-conditioned monocytes differentiate into DCs, driving the development of Th17 cells from autologous naive CD4+ T cells [43].
In addition to the direct effect of IFN type I on Th17 cells, IFN type I may also act indirectly through the production of BAFF. BAFF is reported to be involved in DC maturation and DC-driven Th17 cell differentiation in vitro[44]. BAFF gene silencing ameliorated joint pathology and inhibited the generation of Th17 cells in the joints of a collagen-induced arthritis (CIA) mouse model [44]. In turn, IL-17A can induce the formation of neutrophil extracellular traps (NETs) [45], which could potentially provide new autoantigens to active TLRs on DCs, thereby forming a proinflammatory loop.
We find a correlation between BAFF, an IFN type I inducible factor, and the Th17 produced cytokine IL-21. Ettinger et al.[25] showed that IL-21 together with BAFF promotes B-cell responses by bypassing the need for T-cell help or TLR signaling. As these downstream factors are both involved in activation and selection of B cells, these findings again support the concept that IFN type I and the Th17 pathway act together in driving the disease process of SLE.
IL-21 is also produced by T follicular helper (Tfh) cells, and production by Tfh is crucial for B-cell immunity. By gating for CCR6 expression, we exclude the Tfh cells from our analysis. We therefore measured the production of IL-21 by total memory T cells (CD4+CD45RO+), which include the Tfh cells. The expression of IL-21 by these cells is significantly increased in IFN+ patients compared with IFN- patients and HCs, and IL-21 expression by memory cells also strongly correlated with IFN score (data not shown). These data suggest that Tfh effector function may also be increased in IFN+ patients; however, further studies are required.
By gating on CCR6+ cells, we may miss certain IL-17A-producing cells, including Tfh cells. However, the percentages of IL-17A, IL-17F, and IL-22-producing cells within the CCR6- population were 10– to 20-fold lower than in the CCR6+ population, as described previously by Acosta-Rodriguez et al.[35]. In addition, we did not find a difference in cytokine production between the groups when gating on total CD4+CD45RO+ memory T cells, possibly because the percentages are very small.
In contrast to others, we did not find a correlation between SLEDAI and Th17 cytokines [6]. This might be due to the relatively low patient number, which is a limitation of our study.
In addition to flow-cytometry analysis of cytokine expression by PBMCs, we also measured cytokine levels in the serum of the participants in this study (data not shown). Unfortunately, we were unable to detect IL-17A and F in most of the samples. We did find IL-22 in serum samples of all subjects, but they were not different between the groups. We also found higher levels of IL-21 in IFN+ patients compared with IFN- patients and HCs, but we could detect IL-21 in only one third of the samples.
Although we do not show a functional link between IFN type I and the Th17 pathway, our findings provide the first support for co-occurrence of increased IFN type I activity and increased IL-17/Th17 system in SLE. The Th17-IFN type I interaction found in this study might have implications for future treatment of SLE and other systemic autoimmune diseases in which IFN type I plays a role. Preliminary results from a phase IIa trial with human IgG1κ anti-IFNα antibody, in 87 SLE patients, showed so far a 40% reduction in IFN type I-induced gene expression but no clinical effect compared with placebo (abstract; Merrill J et al. a ). Our data indicate that IFN type I might act in concert with Th17 cytokines, paving the way for combination therapies, possibly resulting in more significant clinical effects in the future.

Conclusion

The aim of this study was to investigate whether CCR6+ memory T-helper cells and their cytokine expression was increased in SLE patients. In addition, we examined whether this increase is related to the presence of IFN type I signature. In the present study, we showed that IFN+ patients had an increased percentage of IL-17A and IL-17A/IL-17F double-producing CCR6+ memory T helper cells in the blood compared to IFN negative patients and HCs. Interestingly, the IL-17A and IL-17F expression within the CCR6+ cells correlated significantly with IFIG expression. Moreover, monocyte BAFF expression in these patients correlated significantly with IL-21 producing CCR6+ memory T-helper cells.
Thus, this study adds new insight into the co-occurrence of the two pathogenic pathways in SLE, the IFN type I and the Th17 pathway, and showed for the first time a higher percentage of IL-17A and IL-17A/IL-17F double-producing CCR6+ memory T-helper cells in IFN+ SLE patients. These findings indicate that IFN type I co-acts with Th17 cytokines in SLE pathogenesis, and further functional studies, including understanding the mechanism, are warranted.

Endnote

aJ. Merrill, V. Chindalore, J. Box, N. Rothfield, J. Fiechtner, J. Sun, D. Ethgen. Results of a randomized placebo-controlled, phase 2a study of sifalimumab, an anti-interferon-alpha monoclonal antibody, administered subcutaneously in subjects with systemic lupus erythematosus [abstract]. [2011] [THU0411].

Acknowledgements

This project was financially supported by grants from The Netherlands Organization for Scientific Research (NWO) and The Dutch Arthritis Association (Reumafonds).

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

ZB and OBJC were involved in the study design and clinical and laboratory data collection, analyzed the data, and drafted and revised the manuscript. CGvH-M, NIM, SMJP, and ND were involved in laboratory data collection and revising the draft article. RJEMD, PLvD,VAD, PMvH, JMWH were involved in collection of clinical data and revising the draft. JPvH was involved in the design of the study and helped to draft the manuscript. MAV and EL were involved in study design, monitoring of data collection, and final draft and approval of the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Choi J, Kim ST, Craft J: The pathogenesis of systemic lupus erythematosus: an update. Curr Opin Immunol. 2012, 24: 651-657. 10.1016/j.coi.2012.10.004.PubMedCentralCrossRefPubMed Choi J, Kim ST, Craft J: The pathogenesis of systemic lupus erythematosus: an update. Curr Opin Immunol. 2012, 24: 651-657. 10.1016/j.coi.2012.10.004.PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, Brohawn P, Kiener PA, Richman L, Fiorentino D, Greenberg SA, Jallal B, Yao Y: Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis. 2011, 70: 2029-2036. 10.1136/ard.2011.150326.CrossRefPubMed Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, Brohawn P, Kiener PA, Richman L, Fiorentino D, Greenberg SA, Jallal B, Yao Y: Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis. 2011, 70: 2029-2036. 10.1136/ard.2011.150326.CrossRefPubMed
3.
Zurück zum Zitat Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, Behrens TW: Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A. 2003, 100: 2610-2615. 10.1073/pnas.0337679100.PubMedCentralCrossRefPubMed Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, Behrens TW: Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A. 2003, 100: 2610-2615. 10.1073/pnas.0337679100.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, Pascual V: Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003, 197: 711-723. 10.1084/jem.20021553.PubMedCentralCrossRefPubMed Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, Pascual V: Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003, 197: 711-723. 10.1084/jem.20021553.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK: Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum. 2005, 52: 1491-1503. 10.1002/art.21031.CrossRefPubMed Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK: Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum. 2005, 52: 1491-1503. 10.1002/art.21031.CrossRefPubMed
6.
Zurück zum Zitat Chen XQ, Yu YC, Deng HH, Sun JZ, Dai Z, Wu YW, Yang M: Plasma IL-17A is increased in new-onset SLE patients and associated with disease activity. J Clin Immunol. 2010, 30: 221-225. 10.1007/s10875-009-9365-x.CrossRefPubMed Chen XQ, Yu YC, Deng HH, Sun JZ, Dai Z, Wu YW, Yang M: Plasma IL-17A is increased in new-onset SLE patients and associated with disease activity. J Clin Immunol. 2010, 30: 221-225. 10.1007/s10875-009-9365-x.CrossRefPubMed
7.
Zurück zum Zitat Yang J, Chu Y, Yang X, Gao D, Zhu L, Wan L, Li M: Th17 and natural Treg cell population dynamics in systemic lupus erythematosus. Arthritis Rheum. 2009, 60: 1472-1483. 10.1002/art.24499.CrossRefPubMed Yang J, Chu Y, Yang X, Gao D, Zhu L, Wan L, Li M: Th17 and natural Treg cell population dynamics in systemic lupus erythematosus. Arthritis Rheum. 2009, 60: 1472-1483. 10.1002/art.24499.CrossRefPubMed
8.
Zurück zum Zitat Shah K, Lee WW, Lee SH, Kim SH, Kang SW, Craft J, Kang I: Dysregulated balance of Th17 and Th1 cells in systemic lupus erythematosus. Arthritis Res Ther. 2010, 12: R53-10.1186/ar2964.PubMedCentralCrossRefPubMed Shah K, Lee WW, Lee SH, Kim SH, Kang SW, Craft J, Kang I: Dysregulated balance of Th17 and Th1 cells in systemic lupus erythematosus. Arthritis Res Ther. 2010, 12: R53-10.1186/ar2964.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Wang Y, Ito S, Chino Y, Goto D, Matsumoto I, Murata H, Tsutsumi A, Hayashi T, Uchida K, Usui J, Yamagata K, Sumida T: Laser microdissection-based analysis of cytokine balance in the kidneys of patients with lupus nephritis. Clin Exp Immunol. 2010, 159: 1-10. 10.1111/j.1365-2249.2009.04031.x.PubMedCentralCrossRefPubMed Wang Y, Ito S, Chino Y, Goto D, Matsumoto I, Murata H, Tsutsumi A, Hayashi T, Uchida K, Usui J, Yamagata K, Sumida T: Laser microdissection-based analysis of cytokine balance in the kidneys of patients with lupus nephritis. Clin Exp Immunol. 2010, 159: 1-10. 10.1111/j.1365-2249.2009.04031.x.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Hueber AJ, Asquith DL, Miller AM, Reilly J, Kerr S, Leipe J, Melendez AJ, McInnes IB: Mast cells express IL-17A in rheumatoid arthritis synovium. J Immunol. 2010, 184: 3336-3340. 10.4049/jimmunol.0903566.CrossRefPubMed Hueber AJ, Asquith DL, Miller AM, Reilly J, Kerr S, Leipe J, Melendez AJ, McInnes IB: Mast cells express IL-17A in rheumatoid arthritis synovium. J Immunol. 2010, 184: 3336-3340. 10.4049/jimmunol.0903566.CrossRefPubMed
11.
Zurück zum Zitat Cua DJ, Tato CM: Innate IL-17-producing cells: the sentinels of the immune system. Nat Rev Immunol. 2010, 10: 479-489. 10.1038/nri2800.CrossRefPubMed Cua DJ, Tato CM: Innate IL-17-producing cells: the sentinels of the immune system. Nat Rev Immunol. 2010, 10: 479-489. 10.1038/nri2800.CrossRefPubMed
12.
Zurück zum Zitat McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, Cua DJ: TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol. 2007, 8: 1390-1397. 10.1038/ni1539.CrossRefPubMed McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, Cua DJ: TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol. 2007, 8: 1390-1397. 10.1038/ni1539.CrossRefPubMed
13.
Zurück zum Zitat Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, McClanahan T, Kastelein RA, Cua DJ: IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005, 201: 233-240. 10.1084/jem.20041257.PubMedCentralCrossRefPubMed Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, McClanahan T, Kastelein RA, Cua DJ: IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005, 201: 233-240. 10.1084/jem.20041257.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, Levy DE, Leonard WJ, Littman DR: IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 2007, 8: 967-974. 10.1038/ni1488.CrossRefPubMed Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, Levy DE, Leonard WJ, Littman DR: IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 2007, 8: 967-974. 10.1038/ni1488.CrossRefPubMed
15.
Zurück zum Zitat Kyttaris VC, Zhang Z, Kuchroo VK, Oukka M, Tsokos GC: Cutting edge: IL-23 receptor deficiency prevents the development of lupus nephritis in C57BL/6-lpr/lpr mice. J Immunol. 2010, 184: 4605-4609. 10.4049/jimmunol.0903595.PubMedCentralCrossRefPubMed Kyttaris VC, Zhang Z, Kuchroo VK, Oukka M, Tsokos GC: Cutting edge: IL-23 receptor deficiency prevents the development of lupus nephritis in C57BL/6-lpr/lpr mice. J Immunol. 2010, 184: 4605-4609. 10.4049/jimmunol.0903595.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Ambrosi A, Espinosa A, Wahren-Herlenius M: IL-17: a new actor in IFN-driven systemic autoimmune diseases. Eur J Immunol. 2012, 42: 2274-2284. 10.1002/eji.201242653.CrossRefPubMed Ambrosi A, Espinosa A, Wahren-Herlenius M: IL-17: a new actor in IFN-driven systemic autoimmune diseases. Eur J Immunol. 2012, 42: 2274-2284. 10.1002/eji.201242653.CrossRefPubMed
17.
Zurück zum Zitat Axtell RC, de Jong BA, Boniface K, van der Voort LF, Bhat R, De Sarno P, Naves R, Han M, Zhong F, Castellanos JG, Mair R, Christakos A, Kolkowitz I, Katz L, Killestein J, Polman CH, de Waal Malefyt R, Steinman L, Raman C: T helper type 1 and 17 cells determine efficacy of interferon-beta in multiple sclerosis and experimental encephalomyelitis. Nat Med. 2010, 16: 406-412. 10.1038/nm.2110.PubMedCentralCrossRefPubMed Axtell RC, de Jong BA, Boniface K, van der Voort LF, Bhat R, De Sarno P, Naves R, Han M, Zhong F, Castellanos JG, Mair R, Christakos A, Kolkowitz I, Katz L, Killestein J, Polman CH, de Waal Malefyt R, Steinman L, Raman C: T helper type 1 and 17 cells determine efficacy of interferon-beta in multiple sclerosis and experimental encephalomyelitis. Nat Med. 2010, 16: 406-412. 10.1038/nm.2110.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Higgs R, Ni Gabhann J, Ben Larbi N, Breen EP, Fitzgerald KA, Jefferies CA: The E3 ubiquitin ligase Ro52 negatively regulates IFN-beta production post-pathogen recognition by polyubiquitin-mediated degradation of IRF3. J Immunol. 2008, 181: 1780-1786.PubMedCentralCrossRefPubMed Higgs R, Ni Gabhann J, Ben Larbi N, Breen EP, Fitzgerald KA, Jefferies CA: The E3 ubiquitin ligase Ro52 negatively regulates IFN-beta production post-pathogen recognition by polyubiquitin-mediated degradation of IRF3. J Immunol. 2008, 181: 1780-1786.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Espinosa A, Dardalhon V, Brauner S, Ambrosi A, Higgs R, Quintana FJ, Sjostrand M, Eloranta ML, Ni Gabhann J, Winqvist O, Sundelin B, Jefferies CA, Rozell B, Kuchroo VK, Wahren-Herlenius M: Loss of the lupus autoantigen Ro52/Trim21 induces tissue inflammation and systemic autoimmunity by disregulating the IL-23-Th17 pathway. J Exp Med. 2009, 206: 1661-1671. 10.1084/jem.20090585.PubMedCentralCrossRefPubMed Espinosa A, Dardalhon V, Brauner S, Ambrosi A, Higgs R, Quintana FJ, Sjostrand M, Eloranta ML, Ni Gabhann J, Winqvist O, Sundelin B, Jefferies CA, Rozell B, Kuchroo VK, Wahren-Herlenius M: Loss of the lupus autoantigen Ro52/Trim21 induces tissue inflammation and systemic autoimmunity by disregulating the IL-23-Th17 pathway. J Exp Med. 2009, 206: 1661-1671. 10.1084/jem.20090585.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Mackay F, Woodcock SA, Lawton P, Ambrose C, Baetscher M, Schneider P, Tschopp J, Browning JL: Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med. 1999, 190: 1697-1710. 10.1084/jem.190.11.1697.PubMedCentralCrossRefPubMed Mackay F, Woodcock SA, Lawton P, Ambrose C, Baetscher M, Schneider P, Tschopp J, Browning JL: Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med. 1999, 190: 1697-1710. 10.1084/jem.190.11.1697.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Becker-Merok A, Nikolaisen C, Nossent HC: B-lymphocyte activating factor in systemic lupus erythematosus and rheumatoid arthritis in relation to autoantibody levels, disease measures and time. Lupus. 2006, 15: 570-576. 10.1177/0961203306071871.CrossRefPubMed Becker-Merok A, Nikolaisen C, Nossent HC: B-lymphocyte activating factor in systemic lupus erythematosus and rheumatoid arthritis in relation to autoantibody levels, disease measures and time. Lupus. 2006, 15: 570-576. 10.1177/0961203306071871.CrossRefPubMed
22.
Zurück zum Zitat Petri M, Stohl W, Chatham W, McCune WJ, Chevrier M, Ryel J, Recta V, Zhong J, Freimuth W: Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus. Arthritis Rheum. 2008, 58: 2453-2459. 10.1002/art.23678.CrossRefPubMed Petri M, Stohl W, Chatham W, McCune WJ, Chevrier M, Ryel J, Recta V, Zhong J, Freimuth W: Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus. Arthritis Rheum. 2008, 58: 2453-2459. 10.1002/art.23678.CrossRefPubMed
23.
Zurück zum Zitat Stohl W, Metyas S, Tan SM, Cheema GS, Oamar B, Xu D, Roschke V, Wu Y, Baker KP, Hilbert DM: B lymphocyte stimulator overexpression in patients with systemic lupus erythematosus: longitudinal observations. Arthritis Rheum. 2003, 48: 3475-3486. 10.1002/art.11354.CrossRefPubMed Stohl W, Metyas S, Tan SM, Cheema GS, Oamar B, Xu D, Roschke V, Wu Y, Baker KP, Hilbert DM: B lymphocyte stimulator overexpression in patients with systemic lupus erythematosus: longitudinal observations. Arthritis Rheum. 2003, 48: 3475-3486. 10.1002/art.11354.CrossRefPubMed
24.
Zurück zum Zitat Brkic Z, Maria NI, van Helden-Meeuwsen CG, van de Merwe JP, van Daele PL, Dalm VA, Wildenberg ME, Beumer W, Drexhage HA, Versnel MA: Prevalence of interferon type I signature in CD14 monocytes of patients with Sjogren’s syndrome and association with disease activity and BAFF gene expression. Ann Rheum Dis. 2013, 72: 728-735. 10.1136/annrheumdis-2012-201381.PubMedCentralCrossRefPubMed Brkic Z, Maria NI, van Helden-Meeuwsen CG, van de Merwe JP, van Daele PL, Dalm VA, Wildenberg ME, Beumer W, Drexhage HA, Versnel MA: Prevalence of interferon type I signature in CD14 monocytes of patients with Sjogren’s syndrome and association with disease activity and BAFF gene expression. Ann Rheum Dis. 2013, 72: 728-735. 10.1136/annrheumdis-2012-201381.PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Ettinger R, Sims GP, Robbins R, Withers D, Fischer RT, Grammer AC, Kuchen S, Lipsky PE: IL-21 and BAFF/BLyS synergize in stimulating plasma cell differentiation from a unique population of human splenic memory B cells. J Immunol. 2007, 178: 2872-2882.CrossRefPubMed Ettinger R, Sims GP, Robbins R, Withers D, Fischer RT, Grammer AC, Kuchen S, Lipsky PE: IL-21 and BAFF/BLyS synergize in stimulating plasma cell differentiation from a unique population of human splenic memory B cells. J Immunol. 2007, 178: 2872-2882.CrossRefPubMed
26.
Zurück zum Zitat Zotos D, Coquet JM, Zhang Y, Light A, D’Costa K, Kallies A, Corcoran LM, Godfrey DI, Toellner KM, Smyth MJ, Nutt SL, Tarlinton DM: IL-21 regulates germinal center B cell differentiation and proliferation through a B cell-intrinsic mechanism. J Exp Med. 2010, 207: 365-378. 10.1084/jem.20091777.PubMedCentralCrossRefPubMed Zotos D, Coquet JM, Zhang Y, Light A, D’Costa K, Kallies A, Corcoran LM, Godfrey DI, Toellner KM, Smyth MJ, Nutt SL, Tarlinton DM: IL-21 regulates germinal center B cell differentiation and proliferation through a B cell-intrinsic mechanism. J Exp Med. 2010, 207: 365-378. 10.1084/jem.20091777.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Hsu HC, Yang P, Wang J, Wu Q, Myers R, Chen J, Yi J, Guentert T, Tousson A, Stanus AL, Le TV, Lorenz RG, Xu H, Kolls JK, Carter RH, Chaplin DD, Williams RW, Mountz JD: Interleukin 17-producing T helper cells and interleukin 17 orchestrate autoreactive germinal center development in autoimmune BXD2 mice. Nat Immunol. 2008, 9: 166-175. 10.1038/ni1552.CrossRefPubMed Hsu HC, Yang P, Wang J, Wu Q, Myers R, Chen J, Yi J, Guentert T, Tousson A, Stanus AL, Le TV, Lorenz RG, Xu H, Kolls JK, Carter RH, Chaplin DD, Williams RW, Mountz JD: Interleukin 17-producing T helper cells and interleukin 17 orchestrate autoreactive germinal center development in autoimmune BXD2 mice. Nat Immunol. 2008, 9: 166-175. 10.1038/ni1552.CrossRefPubMed
28.
Zurück zum Zitat Hochberg MC: Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997, 40: 1725-CrossRefPubMed Hochberg MC: Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997, 40: 1725-CrossRefPubMed
29.
Zurück zum Zitat Bombardier C, Gladman DD, Urowitz MB, Caron D, Chang CH: Derivation of the SLEDAI: a disease activity index for lupus patients: The Committee on Prognosis Studies in SLE. Arthritis Rheum. 1992, 35: 630-640. 10.1002/art.1780350606.CrossRefPubMed Bombardier C, Gladman DD, Urowitz MB, Caron D, Chang CH: Derivation of the SLEDAI: a disease activity index for lupus patients: The Committee on Prognosis Studies in SLE. Arthritis Rheum. 1992, 35: 630-640. 10.1002/art.1780350606.CrossRefPubMed
30.
Zurück zum Zitat Drexhage RC, van der Heul-Nieuwenhuijsen L, Padmos RC, van Beveren N, Cohen D, Versnel MA, Nolen WA, Drexhage HA: Inflammatory gene expression in monocytes of patients with schizophrenia: overlap and difference with bipolar disorder: a study in naturalistically treated patients. Int J Neuropsychopharmacol. 2010, 13: 1369-1381. 10.1017/S1461145710000799.CrossRefPubMed Drexhage RC, van der Heul-Nieuwenhuijsen L, Padmos RC, van Beveren N, Cohen D, Versnel MA, Nolen WA, Drexhage HA: Inflammatory gene expression in monocytes of patients with schizophrenia: overlap and difference with bipolar disorder: a study in naturalistically treated patients. Int J Neuropsychopharmacol. 2010, 13: 1369-1381. 10.1017/S1461145710000799.CrossRefPubMed
31.
Zurück zum Zitat Beillard E, Pallisgaard N, van der Velden VH, Bi W, Dee R, van der Schoot E, Delabesse E, Macintyre E, Gottardi E, Saglio G, Watzinger F, Lion T, van Dongen JJ, Hokland P, Gabert J: Evaluation of candidate control genes for diagnosis and residual disease detection in leukemic patients using ‘real-time’ quantitative reverse-transcriptase polymerase chain reaction (RQ-PCR): a Europe against cancer program. Leukemia. 2003, 17: 2474-2486. 10.1038/sj.leu.2403136.CrossRefPubMed Beillard E, Pallisgaard N, van der Velden VH, Bi W, Dee R, van der Schoot E, Delabesse E, Macintyre E, Gottardi E, Saglio G, Watzinger F, Lion T, van Dongen JJ, Hokland P, Gabert J: Evaluation of candidate control genes for diagnosis and residual disease detection in leukemic patients using ‘real-time’ quantitative reverse-transcriptase polymerase chain reaction (RQ-PCR): a Europe against cancer program. Leukemia. 2003, 17: 2474-2486. 10.1038/sj.leu.2403136.CrossRefPubMed
32.
Zurück zum Zitat van Hamburg JP, Asmawidjaja PS, Davelaar N, Mus AM, Colin EM, Hazes JM, Dolhain RJ, Lubberts E: Th17 cells, but not Th1 cells, from patients with early rheumatoid arthritis are potent inducers of matrix metalloproteinases and proinflammatory cytokines upon synovial fibroblast interaction, including autocrine interleukin-17A production. Arthritis Rheum. 2011, 63: 73-83. 10.1002/art.30093.CrossRefPubMed van Hamburg JP, Asmawidjaja PS, Davelaar N, Mus AM, Colin EM, Hazes JM, Dolhain RJ, Lubberts E: Th17 cells, but not Th1 cells, from patients with early rheumatoid arthritis are potent inducers of matrix metalloproteinases and proinflammatory cytokines upon synovial fibroblast interaction, including autocrine interleukin-17A production. Arthritis Rheum. 2011, 63: 73-83. 10.1002/art.30093.CrossRefPubMed
33.
Zurück zum Zitat Nikpour M, Dempsey AA, Urowitz MB, Gladman DD, Barnes DA: Association of a gene expression profile from whole blood with disease activity in systemic lupus erythaematosus. Ann Rheum Dis. 2008, 67: 1069-1075. 10.1136/ard.2007.074765.CrossRefPubMed Nikpour M, Dempsey AA, Urowitz MB, Gladman DD, Barnes DA: Association of a gene expression profile from whole blood with disease activity in systemic lupus erythaematosus. Ann Rheum Dis. 2008, 67: 1069-1075. 10.1136/ard.2007.074765.CrossRefPubMed
34.
Zurück zum Zitat Lood C, Amisten S, Gullstrand B, Jonsen A, Allhorn M, Truedsson L, Sturfelt G, Erlinge D, Bengtsson AA: Platelet transcriptional profile and protein expression in patients with systemic lupus erythematosus: up-regulation of the type I interferon system is strongly associated with vascular disease. Blood. 2010, 116: 1951-1957. 10.1182/blood-2010-03-274605.CrossRefPubMed Lood C, Amisten S, Gullstrand B, Jonsen A, Allhorn M, Truedsson L, Sturfelt G, Erlinge D, Bengtsson AA: Platelet transcriptional profile and protein expression in patients with systemic lupus erythematosus: up-regulation of the type I interferon system is strongly associated with vascular disease. Blood. 2010, 116: 1951-1957. 10.1182/blood-2010-03-274605.CrossRefPubMed
35.
Zurück zum Zitat Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, Lanzavecchia A, Sallusto F, Napolitani G: Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol. 2007, 8: 639-646. 10.1038/ni1467.CrossRefPubMed Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, Lanzavecchia A, Sallusto F, Napolitani G: Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol. 2007, 8: 639-646. 10.1038/ni1467.CrossRefPubMed
36.
Zurück zum Zitat Annunziato F, Cosmi L, Liotta F, Maggi E, Romagnani S: The phenotype of human Th17 cells and their precursors, the cytokines that mediate their differentiation and the role of Th17 cells in inflammation. Int Immunol. 2008, 20: 1361-1368. 10.1093/intimm/dxn106.CrossRefPubMed Annunziato F, Cosmi L, Liotta F, Maggi E, Romagnani S: The phenotype of human Th17 cells and their precursors, the cytokines that mediate their differentiation and the role of Th17 cells in inflammation. Int Immunol. 2008, 20: 1361-1368. 10.1093/intimm/dxn106.CrossRefPubMed
37.
Zurück zum Zitat Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR: The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006, 126: 1121-1133. 10.1016/j.cell.2006.07.035.CrossRefPubMed Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR: The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006, 126: 1121-1133. 10.1016/j.cell.2006.07.035.CrossRefPubMed
38.
Zurück zum Zitat Takaoka A, Yanai H, Kondo S, Duncan G, Negishi H, Mizutani T, Kano S, Honda K, Ohba Y, Mak TW, Taniguchi T: Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors. Nature. 2005, 434: 243-249. 10.1038/nature03308.CrossRefPubMed Takaoka A, Yanai H, Kondo S, Duncan G, Negishi H, Mizutani T, Kano S, Honda K, Ohba Y, Mak TW, Taniguchi T: Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors. Nature. 2005, 434: 243-249. 10.1038/nature03308.CrossRefPubMed
39.
Zurück zum Zitat Lazear HM, Lancaster A, Wilkins C, Suthar MS, Huang A, Vick SC, Clepper L, Thackray L, Brassil MM, Virgin HW, Nikolich-Zugich J, Moses AV, Gale M, Früh K, Diamond MS: IRF-3, IRF-5, and IRF-7 coordinately regulate the type I IFN response in myeloid dendritic cells downstream of MAVS signaling. PLoS Pathog. 2013, 9: e1003118-10.1371/journal.ppat.1003118.PubMedCentralCrossRefPubMed Lazear HM, Lancaster A, Wilkins C, Suthar MS, Huang A, Vick SC, Clepper L, Thackray L, Brassil MM, Virgin HW, Nikolich-Zugich J, Moses AV, Gale M, Früh K, Diamond MS: IRF-3, IRF-5, and IRF-7 coordinately regulate the type I IFN response in myeloid dendritic cells downstream of MAVS signaling. PLoS Pathog. 2013, 9: e1003118-10.1371/journal.ppat.1003118.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Yu CF, Peng WM, Oldenburg J, Hoch J, Bieber T, Limmer A, Hartmann G, Barchet W, Eis-Hubinger AM, Novak N: Human plasmacytoid dendritic cells support Th17 cell effector function in response to TLR7 ligation. J Immunol. 2010, 184: 1159-1167. 10.4049/jimmunol.0901706.CrossRefPubMed Yu CF, Peng WM, Oldenburg J, Hoch J, Bieber T, Limmer A, Hartmann G, Barchet W, Eis-Hubinger AM, Novak N: Human plasmacytoid dendritic cells support Th17 cell effector function in response to TLR7 ligation. J Immunol. 2010, 184: 1159-1167. 10.4049/jimmunol.0901706.CrossRefPubMed
41.
Zurück zum Zitat Matikainen S, Sareneva T, Ronni T, Lehtonen A, Koskinen PJ, Julkunen I: Interferon-alpha activates multiple STAT proteins and upregulates proliferation-associated IL-2Ralpha, c-myc, and pim-1 genes in human T cells. Blood. 1999, 93: 1980-1991.PubMed Matikainen S, Sareneva T, Ronni T, Lehtonen A, Koskinen PJ, Julkunen I: Interferon-alpha activates multiple STAT proteins and upregulates proliferation-associated IL-2Ralpha, c-myc, and pim-1 genes in human T cells. Blood. 1999, 93: 1980-1991.PubMed
42.
Zurück zum Zitat Yasuda K, Richez C, Maciaszek JW, Agrawal N, Akira S, Marshak-Rothstein A, Rifkin IR: 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: 6876-6885.CrossRefPubMed Yasuda K, Richez C, Maciaszek JW, Agrawal N, Akira S, Marshak-Rothstein A, Rifkin IR: 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: 6876-6885.CrossRefPubMed
43.
Zurück zum Zitat Santini SM, Lapenta C, Donati S, Spadaro F, Belardelli F, Ferrantini M: Interferon-alpha-conditioned human monocytes combine a Th1-orienting attitude with the induction of autologous Th17 responses: role of IL-23 and IL-12. PLoS One. 2011, 6: e17364-10.1371/journal.pone.0017364.PubMedCentralCrossRefPubMed Santini SM, Lapenta C, Donati S, Spadaro F, Belardelli F, Ferrantini M: Interferon-alpha-conditioned human monocytes combine a Th1-orienting attitude with the induction of autologous Th17 responses: role of IL-23 and IL-12. PLoS One. 2011, 6: e17364-10.1371/journal.pone.0017364.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Lai Kwan Lam Q, King Hung Ko O, Zheng BJ, Lu L: Local BAFF gene silencing suppresses Th17-cell generation and ameliorates autoimmune arthritis. Proc Natl Acad Sci U S A. 2008, 105: 14993-4998. 10.1073/pnas.0806044105.PubMedCentralCrossRefPubMed Lai Kwan Lam Q, King Hung Ko O, Zheng BJ, Lu L: Local BAFF gene silencing suppresses Th17-cell generation and ameliorates autoimmune arthritis. Proc Natl Acad Sci U S A. 2008, 105: 14993-4998. 10.1073/pnas.0806044105.PubMedCentralCrossRefPubMed
45.
Zurück zum Zitat Khandpur R, Carmona-Rivera C, Vivekanandan-Giri A, Gizinski A, Yalavarthi S, Knight JS, Friday S, Li S, Patel RM, Subramanian V, Thompson P, Chen P, Fox DA, Pennathur S, Kaplan MJ: NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med. 2013, 5: 178ra140-CrossRef Khandpur R, Carmona-Rivera C, Vivekanandan-Giri A, Gizinski A, Yalavarthi S, Knight JS, Friday S, Li S, Patel RM, Subramanian V, Thompson P, Chen P, Fox DA, Pennathur S, Kaplan MJ: NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med. 2013, 5: 178ra140-CrossRef
Metadaten
Titel
T-helper 17 cell cytokines and interferon type I: partners in crime in systemic lupus erythematosus?
verfasst von
Zana Brkic
Odilia BJ Corneth
Cornelia G van Helden-Meeuwsen
Radboud JEM Dolhain
Naomi I Maria
Sandra MJ Paulissen
Nadine Davelaar
Jan Piet van Hamburg
Paul L van Daele
Virgil A Dalm
P Martin van Hagen
Johanna MW Hazes
Marjan A Versnel
Erik Lubberts
Publikationsdatum
01.04.2014
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 2/2014
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/ar4499

Weitere Artikel der Ausgabe 2/2014

Arthritis Research & Therapy 2/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.