Skip to main content
Erschienen in: Clinical Rheumatology 10/2021

Open Access 08.06.2021 | Original Article

Association analysis of juvenile idiopathic arthritis genetic susceptibility factors in Estonian patients

verfasst von: Tiit Nikopensius, Priit Niibo, Toomas Haller, Triin Jagomägi, Ülle Voog-Oras, Neeme Tõnisson, Andres Metspalu, Mare Saag, Chris Pruunsild

Erschienen in: Clinical Rheumatology | Ausgabe 10/2021

Abstract

Background

Juvenile idiopathic arthritis (JIA) is the most common chronic rheumatic condition of childhood. Genetic association studies have revealed several JIA susceptibility loci with the strongest effect size observed in the human leukocyte antigen (HLA) region. Genome-wide association studies have augmented the number of JIA-associated loci, particularly for non-HLA genes. The aim of this study was to identify new associations at non-HLA loci predisposing to the risk of JIA development in Estonian patients.

Methods

We performed genome-wide association analyses in an entire JIA case–control sample (All-JIA) and in a case–control sample for oligoarticular JIA, the most prevalent JIA subtype. The entire cohort was genotyped using the Illumina HumanOmniExpress BeadChip arrays. After imputation, 16,583,468 variants were analyzed in 263 cases and 6956 controls.

Results

We demonstrated nominal evidence of association for 12 novel non-HLA loci not previously implicated in JIA predisposition. We replicated known JIA associations in CLEC16A and VCTN1 regions in the oligoarticular JIA sample. The strongest associations in the All-JIA analysis were identified at PRKG1 (P = 2,54 × 10−6), LTBP1 (P = 9,45 × 10−6), and ELMO1 (P = 1,05 × 10−5). In the oligoarticular JIA analysis, the strongest associations were identified at NFIA (P = 5,05 × 10−6), LTBP1 (P = 9,95 × 10−6), MX1 (P = 1,65 × 10−5), and CD200R1 (P = 2,59 × 10−5).

Conclusion

This study increases the number of known JIA risk loci and provides additional evidence for the existence of overlapping genetic risk loci between JIA and other autoimmune diseases, particularly rheumatoid arthritis. The reported loci are involved in molecular pathways of immunological relevance and likely represent genomic regions that confer susceptibility to JIA in Estonian patients.
Key Points
Juvenile idiopathic arthritis (JIA) is the most common childhood rheumatic disease with heterogeneous presentation and genetic predisposition.
Present genome-wide association study for Estonian JIA patients is first of its kind in Northern and Northeastern Europe.
The results of the present study increase the knowledge about JIA risk loci replicating some previously described associations, so adding weight to their relevance and describing novel loci.
The study provides additional evidence for the existence of overlapping genetic risk loci between JIA and other autoimmune diseases, particularly rheumatoid arthritis.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10067-021-05756-x.
Tiit Nikopensius and Priit Niibo contributed equally to this work.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Juvenile idiopathic arthritis (JIA) is the most common chronic rheumatic disease of childhood, encompassing all forms of chronic inflammatory arthritis of unknown cause and having an onset before the age of 16 years. JIA can be subdivided into seven distinctive categories according to the International League of Associations for Rheumatology (ILAR) classification criteria [1]. Prevalence and incidence studies have outlined a great extent of ethnic diversity within JIA; however, JIA is more common in populations of European ancestry, particularly in Northern European populations with the highest prevalence varying around 1/1,000 [2, 3]. As JIA is much less common than adult-onset rheumatoid arthritis (RA), the assembly of large samples required for genetic studies is challenging.
There is a strong genetic contribution to the risk of JIA, and multiple susceptibility loci have been established through genome-wide association studies [GWASs) of the most common JIA subtypes oligoarticular and rheumatoid factor (RF)-negative polyarticular JIA, with 17 genome-wide and 20 suggestively significant associations described [4, 5]. The human leukocyte antigen (HLA) region and the top 27 non-HLA loci were estimated to explain 13% and 6%, respectively, of the risk of JIA [4]. Many identified JIA loci have not been characterized functionally, as the majority of associated variants is in intronic and intergenic regions, with their presumable functions accented on target gene expression regulation and not directly affecting the sequence of any protein product [4].
The prevalence of other autoimmune diseases is increased in the relatives of patients with JIA, and particularly for RA – sharing the most similar clinical and pathological features with JIA, and considerable overlap in genetic susceptibility loci for these two traits has been outlined [5, 6]. Many additional RA loci have been identified in recent years, and they likewise may represent acceptable candidate loci for JIA. As a significant proportion of JIA heritability risk remains unclear, it is important to conduct comparative studies in different populations, considering that the replication of previous findings is paramount to determine true risk loci, thus unraveling key disease-related pathways warranting further in-depth studies.
In this study, we adopted a GWAS approach to detect novel loci associated with JIA and replicate several previously reported signals of association, particularly in non-HLA regions in an Estonian JIA case–control sample. We also sought to ascertain additional associations of common variants with lower odds ratios (ORs), as well as uncommon risk alleles with higher odds ratios (ORs).

Methods

Study population

The case group consisted of 263 consecutive Estonian patients with JIA (146 girls and 117 boys) who fulfilled the ILAR diagnostic criteria and had JIA onset at < 16 years of age. Patients were subsequently included in the study during regular visits to the pediatric rheumatologist. The mean age at onset of JIA for the whole sample was 10 years and 5 months (IQR 5 − 13 years); 8 years and 4 months (IQR 4 − 12 years) for boys; and 11 years (IQR 6 − 13 years 5 months) for girls, respectively.
The distribution of JIA subtypes reflected typical proportions, comprising mainly oligoarthritis (86 persistent and 109 extended) and polyarthritis (56 rheumatoid factor (RF)-negative and 2 RF-positive); four patients had psoriatic arthritis, three had enthesitis-related arthritis, and one patient of each had systemic and other arthritis. Additional descriptive data are presented in Supplementary Table S1.
Six thousand ninety hundred and fifty-six healthy controls were selected from the Estonian Biobank cohort. All controls lacked medical records of RA (neither seropositive (M05) nor seronegative (M06)), juvenile arthritides (and with other diseases, M08 and M09), seronegative spondyloarthropathies (M45), and non-specified arthritides (M13). Individuals with family history of JIA or RA were excluded.

Ethics statement

Ethical approval for this study was obtained from the Research Ethics Review Committee of the University of Tartu. All patients and controls provided informed consent for participation in the study.

Genotyping

For the GWAS, all discovery samples were genotyped on the Illumina HumanOmniExpress BeadChip microarrays according to the manufacturer’s protocol. After pruning of single-nucleotide polymorphisms (SNPs) in linkage disequilibrium (r2 > 0.2), EIGENSTRAT was used to compute principal components for use as covariates in the regression analyses. Principal-component analysis (PCA) was performed using EIGENSTRAT to identify ancestry outliers.
Quality control procedures included the following exclusion criteria: marker call rate < 0.95, minor allele frequency (MAF) < 0.01, sample call rate (missingness) < 0.95, and significant departure from Hardy–Weinberg equilibrium (P < 1 × 10−6). We excluded population outliers and all individuals who had unidentifiable sex discrepancies.

Imputation

For imputation, we obtained high-coverage (~ × 40) whole-genome sequencing data from 2240 population-based controls from Estonian Biobank. After quality control, we constructed a reference panel including 16,583,468 single-nucleotide variants (SNVs). The imputation of this nationwide reference panel substantially increases the imputation accuracy for low-frequency variants (allele frequency 0.5 − 10%) in comparison to commonly used reference panels such as 1000 Genomes Project Phase 3. Imputation was performed using IMPUTE version 2.2.2.

Statistical analysis

Variants were tested for an association with JIA case–control status by logistic regression. Primary inference was based on an additive genetic model. Case–control association tests were performed with SNPTEST version 2.5.2, applying gender and first four PCA axes as covariates. Considering our available sample size, we selected P < 5 × 10−8 as the threshold for genome-wide significance; P < 1 × 10−4 level was considered to represent nominal significance for associations. The genomic control inflation factor value was calculated for the whole dataset, and showed only very modest inflation (λGC = 1.04), indicating adequate control of population stratification. Only genotyped SNPs of high quality were used to inform imputation. Imputed SNP quality was assessed using the information score (> 0.6) and the confidence score (> 0.9).

Results

In total, 16,5 million SNVs were genotyped in the Estonian sample. Table 1 presents the strongest associations (P < 1 × 10−4) in the entire case–control sample (All-JIA) and Table 2 in an oligoarticular JIA endophenotype (OA-JIA) sample.
Table 1
Nominally significant case–control association test results: All-JIA sample
Chr
Position
dbSNP ID
Alleles
Gene (region)
MAF
P
OR
95% CI
(Ref/Alt)
Cases
Controls
1
18,013,430
rs12742364
T/A
PADI4
0,096
0,154
5,06 × 10−5
0,583
0,434–0,782
2
33,218,037
rs219158
A/G
LTBP1
0,513
0,413
9,45 × 10−6
1,498
1,258–1,783
6
106,629,698
rs182399829
A/T
PRDM1
0,400
0,327
7,23 × 10−5
1,371
1,148–1,638
6
106,651,006
rs73776556
C/T
ATG5
0,389
0,315
3,20 × 10−5
1,389
1,161–1,661
7
37,157,641
rs2111035
C/A
ELMO1
0,245
0,160
1,05 × 10−5
1,703
1,389–2,087
8
122,539,123
rs1564569
A/G
HAS2
0,337
0,251
2,47 × 10−5
1,510
1,255–1,817
10
53,762,921
rs2454539
C/T
PRKG1
0,513
0,413
2,54 × 10−6
1,501
1,261–1,787
22
24,420,128
rs9624387
C/T
CABIN1
0,062
0,031
3,74 × 10−5
2,066
1,429–2,985
Chr chromosome; MAF minor allele frequency; OR odds ratio; CI confidence intervals
Table 2
Nominally significant case–control association test results: OA-JIA sample
Chr
Position
dbSNP ID
Alleles
Gene (region)
MAF
P
OR
95% CI
(Ref/Alt)
Cases
Controls
1
62,090,537
rs1436238830
T/A
NFIA
0,478
0,377
5,05 × 10−6
0,661
0,540–0,809
1
117,884,022
rs11586858
G/A
VTCN1
0,178
0,256
5,80 × 10−5
1,590
1,222–2,067
2
33,218,037
rs219158
A/G
LTBP1
0,528
0,413
9,95 × 10−6
1,591
1,300–1,947
3
112,484,400
rs62263666
T/C
CD200R1
0,265
0,347
2,59 × 10−5
0,677
0,539–0,850
7
37,157,641
rs2111035
C/A
ELMO1
0,247
0,160
9,37 × 10−5
1,723
1,362–2,179
10
61,620,478
rs141412077
A/G
UBE2D1
0,076
0,034
1,45 × 10−5
2,357
1,600–3,473
13
81,150,170
rs2154130
C/T
SPRY2
0,322
0,433
6,18 × 10−5
1,605
1,294–1,991
16
11,563,226
rs11074991
C/A
CLEC16A
0,204
0,292
6,03 × 10−5
0,620
0,483–0,796
19
6,785,961
rs808843
G/A
VAV1
0,159
0,103
2,75 × 10−5
1,637
1,240–2,160
21
42,816,368
rs28449257
C/G
MX1
0,250
0,324
1,65 × 10−5
0,696
0,551–0,878
Chr chromosome; MAF minor allele frequency; OR odds ratio; CI confidence intervals
In total, 1131 SNPs showed nominal evidence for association in the All-JIA analysis. Seven biologically plausible new JIA loci had nominally significance associations with variants in PADI4, LTBP1, PRDM1/ATG5, ELMO1, HAS2, PRKG1, and CABIN1 defined as genomic regions not previously associated with the trait. None of the markers reached the genome-wide level of significance. The most strongly associated variants were identified at PRKG1 rs2454539 (P = 2,54 × 10−6), LTBP1 rs219158 (P = 9,45 × 10−6), and ELMO1 rs2111035 (P = 1,05 × 10−5). All top variants, except for PADI4, were associated with a higher risk of JIA development.
In total, 1067 SNPs showed nominal evidence for association in the OA-JIA analysis. We replicated previously described association signals [7] for CLEC16A region with the top variant rs11074991 (P = 6,03 × 10−5) and VCTN1 [8] with the top variant rs11586858 (P = 5,80 × 10−5). Seven biologically plausible new loci showed nominally significant associations with variants in regions neighboring NFIA, LTBP1, CD200R1, ELMO1, SPRY2, VAV1, and MX1. The most strongly associated variants were identified at NFIA rs1436238830 (P = 5,05 × 10−6), LTBP1 rs219158 (P = 9,95 × 10−6), MX1 rs28449257 (P = 1,65 × 10−5), and CD200R1 rs62263666 (P = 2,59 × 10−5). Top variants in LTBP1, NFIA, ELMO1, UBE2D1, and VAV1 were associated with a higher risk of oligoarticular JIA development.
The above-mentioned genes have roles in immune system regulation and/or major histocompatibility complex (MHC) antigen processing and presentation, apoptosis, and T cell receptor repertoire. Some genes are associated with other autoimmune diseases.

Discussion

This paper describes a genome-wide association (GWA) study to identify novel JIA susceptibility loci outside HLA region, performed in an Estonian JIA case–control sample (263 cases and 6956 controls). Variants were tested for association with JIA risk by logistic regression. The majority of reported associations appeared in genes encoding proteins that have roles in immune system regulation and/or MHC antigen processing and presentation. As expected, most associated regions fall within non-coding regulatory regions. The majority of reported nominally significant associations overlap with previously reported susceptibility loci for RA; overlaps with other autoimmune diseases were observed for UBE2D1 associated with Crohn’s disease [9], PRDM1 with systemic sclerosis [10], and VAV1 with multiple sclerosis [11]. Altogether, we identified 12 novel loci not previously implicated in genetics of JIA and/or JIA subtypes.
We have replicated previously described association of the CLEC16A gene region with the risk for JIA and also anti-cyclic citrullinated peptide antibody [anti-CCP] negative RA in Norwegian Caucasians [7]. We report association in oligoarticular JIA sample. GWASs have also shown that CLEC16A variants confer susceptibility to autoimmune diseases such as type 1 diabetes and multiple sclerosis; with evidence of association with RA [12]. CLEC16A – C-type lectin domain containing 16A – is expressed almost exclusively in immune cells, such as CD4 and CD8 T cells, dendritic cells, B lymphocytes, and natural killer (NK) cells, demonstrating its importance in immune system regulation. Our findings provide further support for CLEC16A as an autoimmune risk locus, and particularly for oligoarticular JIA. Further efforts are necessary to elucidate the function of CLEC16A, and to identify actual causal variants, which appear to be distinct for different autoimmune diseases.
The second replicated JIA locus, for which we report an association in OA-JIA sample, is VTCN1 – V-set domain-containing T cell activation inhibitor 1 – showing strongest association with JIA among non-HLA genes in previous studies [13, 14], and has been implicated as a potentially predictive disease-course marker for selected JIA subtypes [8].
The ATG5/PRDM1 region has been linked to susceptibility of several autoimmune diseases, including RA [15, 16] and systemic lupus erythematosus (SLE) [17]. ATG5 (autophagy-related 5)-encoded protein is involved in MHC class II antigen presentation, apoptosis, and lymphocyte development and is essential for B and T lymphocyte survival and proliferation. PRDM1 – PR/SET domain 1 – encodes a transcription factor that mediates a transcriptional program in various innate and adaptive immune tissue-resident lymphocyte T cell types. Increased mRNA and protein expression levels of autophagy-related proteins, including Atg5, were found in the synovial tissue of RA patients [18]. The inhibition of autophagy and increased apoptotic activation correlates with a favorable clinical outcome in RA patients treated with anti-tumor necrosis factor (TNF) drugs [19]. This finding suggests that reducing the expression levels of autophagy-related genes might become a new therapeutic target for active RA; the ATG5 suppressor oridonin was recently introduced as a potential therapeutic target for RA [20].
Silencing of transcription factor Prdm1 (B lymphocyte-induced maturation protein-1; Blimp1) in osteoclast precursor cells attenuated the stimulatory effect of TNF-α on osteoclastogenesis; however, TNF-α-induced Prdm1 expression was rescued discernibly by blocking the PI3K/AKT signaling pathway. Upregulation of Prdm1 expression by TNF-α through the activation of PI3K/AKT signaling during osteoclastogenesis seems a plausible molecular mechanism regulating TNF-α-induced osteoclasts differentiation that has profound effects in RA [21]. Interleukin-23 (IL-23) is a pro-inflammatory cytokine required for the pathogenicity of T helper 17 (Th17) cells, and Prdm1 was identified as a key IL-23-induced factor that activates the Th17 inflammatory program [22]. Interactions between Th17 cells and their effector molecules interferon (IFN)-γ and TNF-α are implicated in the pathology of RA, and PRDM1 – among other established genetic risk factors for RA – is directly involved in Th17 cell differentiation and/or function. Interactions between Th17 cells and other immune cells in the synovial tissue during the early phases of RA lead to chronic inflammation, irreversible cartilage degradation, and bone erosion [23]. Downregulation of the expression of Prdm1, a repressor for negative regulators of osteoclast differentiation Irf8 and Bcl6, was proposed as another mechanism of action for compounds possessing effective therapeutic potential for the treatment and/or prevention of bone loss [24].
LTBP1 – latent-transforming growth factor beta-binding protein 1 – encodes a component of the large latent TGF-β complex binding TGF-β to the extracellular matrix and acts as key regulator of latent-state TGF-β activation; overexpression of TGF-β-related genes supports the importance of this pathway in synovial pathology. Global gene expression profiling of JIA fibroblast-like synoviocytes (FLS) exhibited a hypertrophic chondrocyte phenotype and demonstrated that dysregulation of TGF-β signaling pathway along with contributions from the upregulated β-catenin signaling pathway, leading to inhibition of chondrocyte differentiation, and partially downregulated canonical Wnt signaling pathway may have implications for endochondral bone formation and contribute to bony overgrowth in oligoarticular JIA. Moreover, overexpression of bone morphogenetic protein 4 (BMP4) in FLSs from patients with oligoarticular JIA was inferred to have a direct effect on functional outcome in terms of disease pathogenesis and was implicated as a target for future treatment [25]. Recently, involvement of the TGF-β1/Smad signaling pathway in epithelial-mesenchymal transition and contribution to migration and invasion in RA FLSs were shown [26], warranting further studies of JIA FLSs.
ELMO1 (engulfment and cell motility 1)-encoded protein is involved in cytoskeletal rearrangements required for phagocytosis of apoptotic cells and cell migration. Integrative omics analysis demonstrated enhanced expression of ELMO1 protein and hypomethylation of ELMO1 locus, ELMO1-promoted cell migration and invasion, and Rac1 activity regulation in RA FLSs, implicating a link between ELMO1 and RA pathogenicity and the potential of ELMO1 as a RA therapeutic target [27]. Additionally, loss of the apoptotic cell-engulfment signaling protein ELMO1 alleviated disease severity in mouse models of arthritis through regulation of neutrophil chemotaxis to inflamed joints, and ELMO1 knockdown reduces human neutrophil migration to chemokines linked to inflammatory arthritis [28].
AKT and ERK pathways are known to be activated in human RA FLSs, which play crucial roles in RA pathogenesis and joint destruction. SPRY2 – sprouty RTK signaling antagonist 2 – has been known as a tumor suppressor by preventing ERK and AKT signaling activation. By downregulation of Ras/Raf/ERK and PTEN/PI3K/AKT signaling, Spry2 overexpression suppressed AKT and ERK pathways and production of proinflammatory cytokines IL-1β, IL-6, matrix metalloproteinase (MMP)-1, and MMP-3 and effectively inhibited the hyperproliferation induced by TNF-α in RA FLSs [29]. Thus, SPRY2 has a significantly suppressive effect on inflammatory responses in RA, and the enhancement of SPRY2 activity in FLSs contributes to the amelioration or prevention-orientated treatment of RA.
NFIA encodes a member of the NF1 (nuclear factor 1) family of transcription factors and is expressed in CD8 T cells. NFIA has been shown to regulate the production, differentiation, and/or function of immune cell subsets of the innate immune system, including monocytes/macrophages [30] and CD314 − CD158a + NK cells [31]. NFIA represents an example of genetic susceptibility factors specific for anti-CCP-negative RA, with the presumption that production of anti-CCP antibodies additionally requires the engagement of the adaptive immune system [32].
VAV1 – Vav guanine nucleotide exchange factor 1 – encoded protein is important in hematopoiesis, participating in T and B cell development and activation. VAV1 variants were also associated with anti-CCP negative RA, with an involvement in modulation of T cell signaling [33].
Experimental studies have revealed that CD200/CD200R1 signaling pathway has an immunosuppressive effect on the inflammatory cellular immune response and maintains immune homeostasis in the context of autoimmune diseases. Significant negative correlation between CD200R1 – CD200 receptor 1 – expression in monocytes-derived macrophages and disease activity in RA patients was shown to support potential involvement of the CD200/CD200R1 pathway in the pathogenesis of RA and implicate CD200R1 as a valuable biomarker of disease activity [34]. Aberrant CD200/CD200R1 expression in RA was shown to contribute to abnormal Th17 cell differentiation, chemotaxis, and osteoclastogenesis; this abnormal expression was corrected after treatment with a TNF-α antagonist plus methotrexate, demonstrating that CD200/CD200R1 exerts anti-inflammatory functions via multiple mechanisms and delineating a potential immunotherapeutic role for targeting CD200/CD200R1 signaling in RA [35].
Numerous association studies focusing on PADI4 – peptidyl arginine deiminase 4 – variants and RA risk in different populations have given controversial results in different populations. Recent meta-analyses have revealed diversity among different PADI4 variants contributing to RA susceptibility in Asian, but not European populations, as well as in both populations [3638]. The involvement of PADI4 rs2240340 variant in the risk of RA development in patients positive for autoantibodies to citrullinated antigens was shown only for HLA-DRB1*04 non-carriers [39].
In contrast to a previous report where no associations were found between JIA clinical subtypes and PADI4 variants [40], we demonstrated an association with overall JIA in Estonian population. An anti-apoptotic role of PADI4 in RA development was proposed as a molecular mechanism contributing to RA pathogenesis as the knockdown of PADI4 promoted the apoptosis of FLSs and upregulated the expression of p53 and p21 [41].
PRKG1 – protein kinase CGMP-dependent 1 – has demonstrated the strongest association with high levels of circulating IFN-α in SLE patients [42]. The biological function for PRKG1 in type I IFN production or signaling was supported in dendritic cells and NK cells, which conjointly generate IFN-α in SLE [43]; however, PRKG1 function was less strongly supported in T and B lymphocytes, which are not considered to be major IFN-α producing cells [42].
MX1 – MX dynamin-like GTPase 1 – is known as an important contributor in the type 1 interferon pathway, and the type I IFN signature in RA has shown clinical relevance in relation to disease onset and therapeutic response. Increased MX1 expression levels were shown to be correlated with disease activity in fibroblast cells of synovial tissue in RA patients with high C-reactive protein levels [44]. Gene expression signatures in RA synovial samples have shown downregulation of the protein–protein interaction network comprising MX1 and significant enrichment of this network in the immune response pathway [45]. A recent study demonstrated that RA-associated DNA methylation sites have regulatory effects on mRNA expressions and create an IFN-inducible gene interaction network (comprising MX1) associated with RA [46].
An imbalance of synthesis and degradation causes overproduction of hyaluronan (HA) – a hallmark of joint swelling associated with RA caused by the undesired activation of three hyaluronan synthase genes: HAS1, HAS2, and HAS3, which seem to have distinct functions. IL1-β, TNF-α, and TNF-β strongly induce HA synthesis via NF-kappaβ signaling pathway – this pathway mediates hyaluronan synthase 2 (HAS2) mRNA expression, which modulates monocyte adhesion by CD44, initializing an inflammatory process wherein HA and adhesion molecules participate in the recruitment of immune cells [47]. TGF-β1 contributes most efficiently to accumulation of high-molecular-weight HA by upregulating HAS1/HAS2 expression and downregulating KIAA1199 expression in RA FLSs [48].
Calcineurin plays an important role in the T cell receptor-mediated signal transduction pathway. CABIN1 encodes calcineurin-binding protein 1 – a negative regulator for calcineurin-mediated signal transduction in T lymphocytes. The abnormal activation of calcineurin by stimulation with IL-1β and TNF-α in RA synoviocytes may contribute to the pathogenesis of inflammatory arthritis; however, overexpression of CABIN1, a natural calcineurin antagonist, suppressed calcineurin activity by inhibiting IL-6 and MMP-2 production by rheumatoid synoviocytes [49]. Investigation of transgenic mice overexpressing hCABIN-1 demonstrated that CABIN1 has an important role for in promoting FLS apoptosis, in attenuating cartilage and bone destruction and inflammation in RA [50].
GWAS approach has been successful in searching for susceptibility genes for multifactorial autoimmune diseases. Cumulative data from genome-wide JIA-focused screens indicate that the identified risk alleles are relatively common in general population, have only modest effects on risk, are located in intergenic regions and introns, and together explain only a small proportion of the variance in disease risk. The majority of identified JIA risk loci and variants represents genetic risk factors shared with other autoimmune diseases, pointing to a common genetic background with discrete biological pathways underlying autoinflammatory processes. Notably, several risk-contributing loci are not necessarily shared among different ethnic groups; thus, genetic risk factors for JIA likely differ among worldwide populations. The low prevalence and clinical heterogeneity of JIA have rendered the assembly of large cohorts difficult. We acknowledge that with the current sample size, this study was insufficiently powered to identify common variants having only modest effect sizes; therefore, we were unable to replicate genome-wide-significant associations found for several previously known JIA-predisposing genes, such as PTPN22, STAT4, PTPN2, ANKRD55, and IL2/IL21 (4). Clearly, previous studies attempting to replicate RA-specific loci likely had limited power to detect associations with JIA. Large-scale GWASs of RA and JIA, respectively, render us prospects to identify potential common mechanisms in disease pathogenesis, which may lead to the identification of potential novel targets for therapeutic intervention and extending treatment strategies from commonly used molecular therapy with antibodies against TNF-α or IL-1 inhibitors to include recent advances in efforts to unravel effective targets influencing autoinflammatory and/or autoimmune processes.
Our findings markedly increase the estimated number of JIA susceptibility loci to be about 50. Their overlap with other autoimmune disease-related phenotypes infers shared etiologies for JIA and RA, giving an opportunity to identify therapies that more broadly influence JIA in the context of inflammatory arthritis risk. The implicated genes highlight the importance of autophagy in apoptosis resistance in inflammatory arthritis; DNA methylation complemented with the type I IFN signature network; and additional autoimmune and inflammatory pathways such as NF-κB, TGF-induced MAPK, CD200/CD200R1, Wnt, AKT, and ERK. Further functional characterization of essential pathways and pinpointing of genes through which their effects are mediated could provide additional insights into biological processes underlying the identified associations.

Conclusions

The results of this study further emphasize the role of common genetic variation and add to the understanding of the genomic architecture influencing the risk of oligoarticular and RF-negative polyarticular JIA in Estonian patients. Certainly, more JIA-specific genetic risk factors remain to be identified, and well-powered GWASs are required for such identification. The future clinical relevance of our findings include contributions toward better understanding of JIA pathogenesis, identification of targets for drug development and/or repositioning of treatment, and presumably to improved prediction of JIA and its subtypes.

Declarations

Ethics approval and consent to participate are approved by IRB.
Not applicable.

Disclosures

None.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Petty RE, Southwood TR, Manners P, Baum J, Glass DN, Goldenberg J et al (2004) International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: second revision, Edmonton, 2001. J Rheumatol. 31(2):390–392PubMed Petty RE, Southwood TR, Manners P, Baum J, Glass DN, Goldenberg J et al (2004) International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: second revision, Edmonton, 2001. J Rheumatol. 31(2):390–392PubMed
2.
Zurück zum Zitat Thierry S, Fautrel B, Lemelle I, Guillemin F (2014) Prevalence and incidence of juvenile idiopathic arthritis: a systematic review. J Joint Bone Spine 81(2):112–117CrossRef Thierry S, Fautrel B, Lemelle I, Guillemin F (2014) Prevalence and incidence of juvenile idiopathic arthritis: a systematic review. J Joint Bone Spine 81(2):112–117CrossRef
3.
Zurück zum Zitat Pruunsild C, Uibo K, Liivamägi H, Tarraste S, Talvik T, Pelkonen P (2007) Prevalence and short-term outcome of juvenile idiopathic arthritis: a population-based study in Estonia. Clin Exp Rheumatol 25(4):649–653PubMed Pruunsild C, Uibo K, Liivamägi H, Tarraste S, Talvik T, Pelkonen P (2007) Prevalence and short-term outcome of juvenile idiopathic arthritis: a population-based study in Estonia. Clin Exp Rheumatol 25(4):649–653PubMed
4.
Zurück zum Zitat Hinks A, Cobb J, Marion MC, Prahalad S, Sudman M, Bowes J et al (2013) Dense genotyping of immune-related disease regions identifies 14 new susceptibility loci for juvenile idiopathic arthritis. Nat Genet 45(6):664–669PubMedPubMedCentralCrossRef Hinks A, Cobb J, Marion MC, Prahalad S, Sudman M, Bowes J et al (2013) Dense genotyping of immune-related disease regions identifies 14 new susceptibility loci for juvenile idiopathic arthritis. Nat Genet 45(6):664–669PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat McIntosh LA, Marion MC, Sudman M, Comeau ME, Becker ML, Bohnsack JF et al (2017) Genome-wide association meta-analysis reveals novel juvenile idiopathic arthritis susceptibility loci. Arthritis Rheumatol 69(11):2222–2232PubMedPubMedCentralCrossRef McIntosh LA, Marion MC, Sudman M, Comeau ME, Becker ML, Bohnsack JF et al (2017) Genome-wide association meta-analysis reveals novel juvenile idiopathic arthritis susceptibility loci. Arthritis Rheumatol 69(11):2222–2232PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Hinks A, Cobb J, Sudman M, Eyre S, Martin P, Flynn E et al (2012) Investigation of rheumatoid arthritis susceptibility loci in juvenile idiopathic arthritis confirms high degree of overlap. Ann Rheum Dis 71(7):1117–1121PubMedCrossRef Hinks A, Cobb J, Sudman M, Eyre S, Martin P, Flynn E et al (2012) Investigation of rheumatoid arthritis susceptibility loci in juvenile idiopathic arthritis confirms high degree of overlap. Ann Rheum Dis 71(7):1117–1121PubMedCrossRef
7.
Zurück zum Zitat Skinningsrud B, Lie BA, Husebye ES, Kvien TK, Førre Ø, Flatø B et al (2010) A CLEC16A variant confers risk for juvenile idiopathic arthritis and anti-cyclic citrullinated peptide antibody negative rheumatoid arthritis. Ann Rheum Dis 69(8):1471–1474PubMedCrossRef Skinningsrud B, Lie BA, Husebye ES, Kvien TK, Førre Ø, Flatø B et al (2010) A CLEC16A variant confers risk for juvenile idiopathic arthritis and anti-cyclic citrullinated peptide antibody negative rheumatoid arthritis. Ann Rheum Dis 69(8):1471–1474PubMedCrossRef
8.
Zurück zum Zitat Albers HM, Reinards TH, Brinkman DM, Kamphuis SS, van Rossum MA, Hoppenreijs EP et al (2014) Genetic variation in VTCN1 (B7–H4) is associated with course of disease in juvenile idiopathic arthritis. Ann Rheum Dis. 73(6):1198–1201PubMedCrossRef Albers HM, Reinards TH, Brinkman DM, Kamphuis SS, van Rossum MA, Hoppenreijs EP et al (2014) Genetic variation in VTCN1 (B7–H4) is associated with course of disease in juvenile idiopathic arthritis. Ann Rheum Dis. 73(6):1198–1201PubMedCrossRef
9.
Zurück zum Zitat Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, Ahmad T et al (2010) Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet 42(12):1118–1125PubMedPubMedCentralCrossRef Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, Ahmad T et al (2010) Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet 42(12):1118–1125PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Terao C, Kawaguchi T, Dieude P, Varga J, Kuwana M, Hudson M et al (2017) Transethnic meta-analysis identifies GSDMA and PRDM1 as susceptibility genes to systemic sclerosis. Ann Rheum Dis 76:1150–1158PubMedCrossRef Terao C, Kawaguchi T, Dieude P, Varga J, Kuwana M, Hudson M et al (2017) Transethnic meta-analysis identifies GSDMA and PRDM1 as susceptibility genes to systemic sclerosis. Ann Rheum Dis 76:1150–1158PubMedCrossRef
11.
Zurück zum Zitat Jagodic M, Colacios C, Nohra R, Dejean AS, Beyeen AD, Khademi M et al (2009) A role for VAV1 in experimental autoimmune encephalomyelitis and multiple sclerosis. Sci Transl Med 1(10):10ra21PubMedCrossRef Jagodic M, Colacios C, Nohra R, Dejean AS, Beyeen AD, Khademi M et al (2009) A role for VAV1 in experimental autoimmune encephalomyelitis and multiple sclerosis. Sci Transl Med 1(10):10ra21PubMedCrossRef
12.
Zurück zum Zitat Martínez A, Perdigones N, Cénit MC, Espino L, Varadé J, Lamas JR et al (2010) Chromosomal region 16p13: further evidence of increased predisposition to immune diseases. Ann Rheum Dis 69(1):309–311PubMedCrossRef Martínez A, Perdigones N, Cénit MC, Espino L, Varadé J, Lamas JR et al (2010) Chromosomal region 16p13: further evidence of increased predisposition to immune diseases. Ann Rheum Dis 69(1):309–311PubMedCrossRef
13.
Zurück zum Zitat Hinks A, Barton A, Shephard N, Eyre S, Bowes J, Cargill M et al (2009) Identification of a novel susceptibility locus for juvenile idiopathic arthritis by genome-wide association analysis. Arthritis Rheum 60(1):258–263PubMedPubMedCentralCrossRef Hinks A, Barton A, Shephard N, Eyre S, Bowes J, Cargill M et al (2009) Identification of a novel susceptibility locus for juvenile idiopathic arthritis by genome-wide association analysis. Arthritis Rheum 60(1):258–263PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Reinards TH, Albers HM, Brinkman DM, Kamphuis SS, van Rossum MAJ, Girschick HJ et al (2015) CD226 (DNAM-1) is associated with susceptibility to juvenile idiopathic arthritis. Ann Rheum Dis 74(12):2193–2198PubMedCrossRef Reinards TH, Albers HM, Brinkman DM, Kamphuis SS, van Rossum MAJ, Girschick HJ et al (2015) CD226 (DNAM-1) is associated with susceptibility to juvenile idiopathic arthritis. Ann Rheum Dis 74(12):2193–2198PubMedCrossRef
15.
Zurück zum Zitat Raychaudhuri S, Thomson BP, Remmers EF, Eyre S, Hinks A, Guiducci C et al (2009) Genetic variants at CD28, PRDM1 and CD2/CD58 are associated with rheumatoid arthritis risk. Nat Genet 41(12):1313–1318PubMedPubMedCentralCrossRef Raychaudhuri S, Thomson BP, Remmers EF, Eyre S, Hinks A, Guiducci C et al (2009) Genetic variants at CD28, PRDM1 and CD2/CD58 are associated with rheumatoid arthritis risk. Nat Genet 41(12):1313–1318PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Okada Y, Wu D, Trynka G, Raj T, Terao C, Ikari K et al (2014) Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506(7488):376–381PubMedCrossRef Okada Y, Wu D, Trynka G, Raj T, Terao C, Ikari K et al (2014) Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506(7488):376–381PubMedCrossRef
17.
Zurück zum Zitat Gateva V, Sandling JK, Hom G, Taylor KE, Chung SA, Sun X et al (2009) A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nat Genet 41(11):1228–1233PubMedPubMedCentralCrossRef Gateva V, Sandling JK, Hom G, Taylor KE, Chung SA, Sun X et al (2009) A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nat Genet 41(11):1228–1233PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Zhu L, Wang H, Wu Y, He Z, Qin Y, Shen Q (2017) The autophagy level is increased in the synovial tissues of patients with active rheumatoid arthritis and is correlated with disease severity. Mediators Inflamm 2017:7623145PubMedPubMedCentralCrossRef Zhu L, Wang H, Wu Y, He Z, Qin Y, Shen Q (2017) The autophagy level is increased in the synovial tissues of patients with active rheumatoid arthritis and is correlated with disease severity. Mediators Inflamm 2017:7623145PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Vomero M, Manganelli V, Barbati C, Colasanti T, Capozzi A, Finucci A et al (2019) Reduction of autophagy and increase in apoptosis correlates with a favorable clinical outcome in patients with rheumatoid arthritis treated with anti-TNF drugs. Arthritis Res Ther 21(1):39PubMedPubMedCentralCrossRef Vomero M, Manganelli V, Barbati C, Colasanti T, Capozzi A, Finucci A et al (2019) Reduction of autophagy and increase in apoptosis correlates with a favorable clinical outcome in patients with rheumatoid arthritis treated with anti-TNF drugs. Arthritis Res Ther 21(1):39PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat He SD, Huang SG, Zhu HJ, Luo XG, Liao KH, Zhang JY et al (2020) Oridonin suppresses autophagy and survival in rheumatoid arthritis fibroblast-like synoviocytes. Pharm Biol 58(1):146–151PubMedPubMedCentralCrossRef He SD, Huang SG, Zhu HJ, Luo XG, Liao KH, Zhang JY et al (2020) Oridonin suppresses autophagy and survival in rheumatoid arthritis fibroblast-like synoviocytes. Pharm Biol 58(1):146–151PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Wu L, Guo Q, Yang J, Ni B (2017) Tumor necrosis factor alpha promotes osteoclast formation via PI3K/Akt pathway-mediated blimp1 expression upregulation. J Cell Biochem 118(6):1308–1315PubMedCrossRef Wu L, Guo Q, Yang J, Ni B (2017) Tumor necrosis factor alpha promotes osteoclast formation via PI3K/Akt pathway-mediated blimp1 expression upregulation. J Cell Biochem 118(6):1308–1315PubMedCrossRef
22.
Zurück zum Zitat Jain R, Chen Y, Kanno Y, Joyce-Shaikh B, Vahedi G, Hirahara K et al (2016) Interleukin-23-induced transcription factor Blimp-1 promotes pathogenicity of t helper 17 cells. Immunity 44(1):131–142PubMedCrossRef Jain R, Chen Y, Kanno Y, Joyce-Shaikh B, Vahedi G, Hirahara K et al (2016) Interleukin-23-induced transcription factor Blimp-1 promotes pathogenicity of t helper 17 cells. Immunity 44(1):131–142PubMedCrossRef
23.
Zurück zum Zitat van Hamburg JP, Tas SW (2018) Molecular mechanisms underpinning T helper 17 cell heterogeneity and functions in rheumatoid arthritis. J Autoimmun 87:69–81PubMedCrossRef van Hamburg JP, Tas SW (2018) Molecular mechanisms underpinning T helper 17 cell heterogeneity and functions in rheumatoid arthritis. J Autoimmun 87:69–81PubMedCrossRef
24.
Zurück zum Zitat Ihn HJ, Lee T, Lee D, Bae JS, Kim SH, Jang IH et al (2019) Inhibitory effect of KP-A038 on osteoclastogenesis and inflammatory bone loss is associated with downregulation of Blimp1. Front Pharmacol 10:367PubMedPubMedCentralCrossRef Ihn HJ, Lee T, Lee D, Bae JS, Kim SH, Jang IH et al (2019) Inhibitory effect of KP-A038 on osteoclastogenesis and inflammatory bone loss is associated with downregulation of Blimp1. Front Pharmacol 10:367PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Brescia AC, Simonds MM, McCahan SM, Fawcett PT, Rose CD (2014) The role of transforming growth factor β signaling in fibroblast-like synoviocytes from patients with oligoarticular juvenile idiopathic arthritis: dysregulation of transforming growth factor β signaling, including overexpression of bone morphogenetic protein 4, may lead to a chondrocyte phenotype and may contribute to bony hypertrophy. Arthritis Rheumatol. 66(5):1352–1362PubMedPubMedCentralCrossRef Brescia AC, Simonds MM, McCahan SM, Fawcett PT, Rose CD (2014) The role of transforming growth factor β signaling in fibroblast-like synoviocytes from patients with oligoarticular juvenile idiopathic arthritis: dysregulation of transforming growth factor β signaling, including overexpression of bone morphogenetic protein 4, may lead to a chondrocyte phenotype and may contribute to bony hypertrophy. Arthritis Rheumatol. 66(5):1352–1362PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Zhu D, Zhao J, Lou A, Huang Q, OuYang Q, Zhu J et al (2019) Transforming growth factor β1 promotes fibroblast-like synoviocytes migration and invasion via TGF-β1/Smad signaling in rheumatoid arthritis. Mol Cell Biochem 459(1–2):141–150PubMedCrossRef Zhu D, Zhao J, Lou A, Huang Q, OuYang Q, Zhu J et al (2019) Transforming growth factor β1 promotes fibroblast-like synoviocytes migration and invasion via TGF-β1/Smad signaling in rheumatoid arthritis. Mol Cell Biochem 459(1–2):141–150PubMedCrossRef
27.
Zurück zum Zitat Whitaker JW, Boyle DL, Bartok B, Ball ST, Gay S, Wang W et al (2015) Integrative omics analysis of rheumatoid arthritis identifies non-obvious therapeutic targets. PLoS One 10(4):e0124254PubMedPubMedCentralCrossRef Whitaker JW, Boyle DL, Bartok B, Ball ST, Gay S, Wang W et al (2015) Integrative omics analysis of rheumatoid arthritis identifies non-obvious therapeutic targets. PLoS One 10(4):e0124254PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Arandjelovic S, Perry JSA, Lucas CD, Penberthy KK, Kim T-H, Zhou M et al (2019) A noncanonical role for the engulfment gene ELMO1 in neutrophils that promotes inflammatory arthritis. Nat Immunol 20(2):141–151PubMedPubMedCentralCrossRef Arandjelovic S, Perry JSA, Lucas CD, Penberthy KK, Kim T-H, Zhou M et al (2019) A noncanonical role for the engulfment gene ELMO1 in neutrophils that promotes inflammatory arthritis. Nat Immunol 20(2):141–151PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Zhang W, Du Z, Zhu J, Yu J, Xu Y (2015) Sprouty2 suppresses the inflammatory responses in rheumatoid arthritis fibroblast-like synoviocytes through regulating the Raf/ERK and PTEN/AKT signals. Mol Immunol 67(2 Pt B):532–539PubMedCrossRef Zhang W, Du Z, Zhu J, Yu J, Xu Y (2015) Sprouty2 suppresses the inflammatory responses in rheumatoid arthritis fibroblast-like synoviocytes through regulating the Raf/ERK and PTEN/AKT signals. Mol Immunol 67(2 Pt B):532–539PubMedCrossRef
30.
Zurück zum Zitat Rosa A, Ballarino M, Sorrentino A, Sthandier O, De Angelis FG, Marchioni M et al (2007) The interplay between the master transcription factor PU.1 and miR-424 regulates human monocyte/macrophage differentiation. Proc Natl Acad Sci U S A 104(50):19849–19854PubMedPubMedCentralCrossRef Rosa A, Ballarino M, Sorrentino A, Sthandier O, De Angelis FG, Marchioni M et al (2007) The interplay between the master transcription factor PU.1 and miR-424 regulates human monocyte/macrophage differentiation. Proc Natl Acad Sci U S A 104(50):19849–19854PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Roederer M, Quaye L, Mangino M, Beddall MH, Mahnke Y, Chattopadhyay P et al (2015) The genetic architecture of the human immune system: a bioresource for autoimmunity and disease pathogenesis. Cell 161(2):387–403PubMedPubMedCentralCrossRef Roederer M, Quaye L, Mangino M, Beddall MH, Mahnke Y, Chattopadhyay P et al (2015) The genetic architecture of the human immune system: a bioresource for autoimmunity and disease pathogenesis. Cell 161(2):387–403PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Viatte S, Massey J, Bowes J, Duffus K, arcOGEN Consortium, Eyre S et al (2016) Replication of associations of genetic loci outside the HLA region with susceptibility to anti-cyclic citrullinated peptide-negative rheumatoid arthritis. Arthritis Rheumatol 68(7):1603–1613PubMedPubMedCentralCrossRef Viatte S, Massey J, Bowes J, Duffus K, arcOGEN Consortium, Eyre S et al (2016) Replication of associations of genetic loci outside the HLA region with susceptibility to anti-cyclic citrullinated peptide-negative rheumatoid arthritis. Arthritis Rheumatol 68(7):1603–1613PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Guerreiro-Cacais AO, Norin U, Gyllenberg A, Berglund R, Beyeen AD, Rheumatoid Arthritis Consortium International (RACI) et al (2017) VAV1 regulates experimental autoimmune arthritis and is associated with anti-CCP negative rheumatoid arthritis. Genes Immun 18(1):48–56PubMedCrossRef Guerreiro-Cacais AO, Norin U, Gyllenberg A, Berglund R, Beyeen AD, Rheumatoid Arthritis Consortium International (RACI) et al (2017) VAV1 regulates experimental autoimmune arthritis and is associated with anti-CCP negative rheumatoid arthritis. Genes Immun 18(1):48–56PubMedCrossRef
34.
Zurück zum Zitat Gao S, Hao B, Yang XF, Chen WQ (2014) Decreased CD200R expression on monocyte-derived macrophages correlates with Th17/Treg imbalance and disease activity in rheumatoid arthritis patients. Inflamm Res 63(6):441–450PubMedCrossRef Gao S, Hao B, Yang XF, Chen WQ (2014) Decreased CD200R expression on monocyte-derived macrophages correlates with Th17/Treg imbalance and disease activity in rheumatoid arthritis patients. Inflamm Res 63(6):441–450PubMedCrossRef
35.
Zurück zum Zitat Ren Y, Yang B, Yin Y, Leng X, Jiang Y, Zhang L et al (2015) Aberrant CD200/CD200R1 expression and its potential role in Th17 cell differentiation, chemotaxis and osteoclastogenesis in rheumatoid arthritis. Rheumatology (Oxford) 54(4):712–721CrossRef Ren Y, Yang B, Yin Y, Leng X, Jiang Y, Zhang L et al (2015) Aberrant CD200/CD200R1 expression and its potential role in Th17 cell differentiation, chemotaxis and osteoclastogenesis in rheumatoid arthritis. Rheumatology (Oxford) 54(4):712–721CrossRef
36.
Zurück zum Zitat Burr ML, Naseem H, Hinks A, Eyre S, Gibbons LJ, Bowes J et al (2010) PADI4 genotype is not associated with rheumatoid arthritis in a large UK Caucasian population. Ann Rheum Dis 69(4):666–670PubMedCrossRef Burr ML, Naseem H, Hinks A, Eyre S, Gibbons LJ, Bowes J et al (2010) PADI4 genotype is not associated with rheumatoid arthritis in a large UK Caucasian population. Ann Rheum Dis 69(4):666–670PubMedCrossRef
37.
Zurück zum Zitat Lee YH, Bae SC (2016) Association between susceptibility to rheumatoid arthritis and PADI4 polymorphisms: a meta-analysis. Clin Rheumatol 35(4):961–971PubMedCrossRef Lee YH, Bae SC (2016) Association between susceptibility to rheumatoid arthritis and PADI4 polymorphisms: a meta-analysis. Clin Rheumatol 35(4):961–971PubMedCrossRef
38.
Zurück zum Zitat Lu C, Xu K, Guo H, Peng K, Yang Z, Hao YQ et al (2018) The relationship of PADI4_94 polymorphisms with the morbidity of rheumatoid arthritis in Caucasian and Asian populations: a meta-analysis and system review. Clin Rheumatol 37(2):289–296PubMedCrossRef Lu C, Xu K, Guo H, Peng K, Yang Z, Hao YQ et al (2018) The relationship of PADI4_94 polymorphisms with the morbidity of rheumatoid arthritis in Caucasian and Asian populations: a meta-analysis and system review. Clin Rheumatol 37(2):289–296PubMedCrossRef
39.
Zurück zum Zitat Snir O, Gomez-Cabrero D, Montes A, Perez-Pampin E, Gómez-Reino JJ, Seddighzadeh M et al (2014) Non-HLA genes PTPN22, CDK6 and PADI4 are associated with specific autoantibodies in HLA-defined subgroups of rheumatoid arthritis. Arthritis Res Ther 16(4):414PubMedPubMedCentralCrossRef Snir O, Gomez-Cabrero D, Montes A, Perez-Pampin E, Gómez-Reino JJ, Seddighzadeh M et al (2014) Non-HLA genes PTPN22, CDK6 and PADI4 are associated with specific autoantibodies in HLA-defined subgroups of rheumatoid arthritis. Arthritis Res Ther 16(4):414PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Hisa K, Yanagimachi MD, Naruto T, Miyamae T, Kikuchi M, Hara R et al (2017) PADI4 and the HLA-DRB1 shared epitope in juvenile idiopathic arthritis. PLoS One 12(2):e0171961PubMedPubMedCentralCrossRef Hisa K, Yanagimachi MD, Naruto T, Miyamae T, Kikuchi M, Hara R et al (2017) PADI4 and the HLA-DRB1 shared epitope in juvenile idiopathic arthritis. PLoS One 12(2):e0171961PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Fan L, Zong M, Gong R, He D, Li N, Sun LS et al (2017) PADI4 epigenetically suppresses p21 transcription and inhibits cell apoptosis in fibroblast-like synoviocytes from rheumatoid arthritis patients. Int J Biol Sci 13(3):358–366PubMedPubMedCentralCrossRef Fan L, Zong M, Gong R, He D, Li N, Sun LS et al (2017) PADI4 epigenetically suppresses p21 transcription and inhibits cell apoptosis in fibroblast-like synoviocytes from rheumatoid arthritis patients. Int J Biol Sci 13(3):358–366PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Kariuki SN, Ghodke-Puranik Y, Dorschner JM, Chrabot BS, Kelly JA, Tsao BP et al (2015) Genetic analysis of the pathogenic molecular sub-phenotype interferon-alpha identifies multiple novel loci involved in systemic lupus erythematosus. Genes Immun 16(1):15–23PubMedCrossRef Kariuki SN, Ghodke-Puranik Y, Dorschner JM, Chrabot BS, Kelly JA, Tsao BP et al (2015) Genetic analysis of the pathogenic molecular sub-phenotype interferon-alpha identifies multiple novel loci involved in systemic lupus erythematosus. Genes Immun 16(1):15–23PubMedCrossRef
43.
Zurück zum Zitat Hagberg N, Berggren O, Leonard D, Weber G, Bryceson YT, Alm GV et al (2011) IFN-α production by plasmacytoid dendritic cells stimulated with RNA-containing immune complexes is promoted by NK cells via MIP-1β and LFA-1. J Immunol 186(9):5085–5094PubMedCrossRef Hagberg N, Berggren O, Leonard D, Weber G, Bryceson YT, Alm GV et al (2011) IFN-α production by plasmacytoid dendritic cells stimulated with RNA-containing immune complexes is promoted by NK cells via MIP-1β and LFA-1. J Immunol 186(9):5085–5094PubMedCrossRef
44.
Zurück zum Zitat Galligan CL, Baig E, Bykerk V, Keystone EC, Fish EN (2007) Distinctive gene expression signatures in rheumatoid arthritis synovial tissue fibroblast cells: correlates with disease activity. Genes Immun 8(6):480–491PubMedCrossRef Galligan CL, Baig E, Bykerk V, Keystone EC, Fish EN (2007) Distinctive gene expression signatures in rheumatoid arthritis synovial tissue fibroblast cells: correlates with disease activity. Genes Immun 8(6):480–491PubMedCrossRef
45.
Zurück zum Zitat He P, Zhang Z, Liao W, Xu D, Fu M, Kang Y (2016) Screening of gene signatures for rheumatoid arthritis and osteoarthritis based on bioinformatics analysis. Mol Med Rep 14(2):1587–1593PubMedPubMedCentralCrossRef He P, Zhang Z, Liao W, Xu D, Fu M, Kang Y (2016) Screening of gene signatures for rheumatoid arthritis and osteoarthritis based on bioinformatics analysis. Mol Med Rep 14(2):1587–1593PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Zhu H, Wu LF, Mo XB, Lu X, Tang H, Zhu XW et al (2019) Rheumatoid arthritis-associated DNA methylation sites in peripheral blood mononuclear cells. Ann Rheum Dis 78(1):36–42PubMedCrossRef Zhu H, Wu LF, Mo XB, Lu X, Tang H, Zhu XW et al (2019) Rheumatoid arthritis-associated DNA methylation sites in peripheral blood mononuclear cells. Ann Rheum Dis 78(1):36–42PubMedCrossRef
47.
Zurück zum Zitat Vigetti D, Genasetti A, Karousou E, Viola M, Moretto P, Clerici M et al (2010) Proinflammatory cytokines induce hyaluronan synthesis and monocyte adhesion in human endothelial cells through hyaluronan synthase 2 (HAS2) and the nuclear factor-kappaB (NF-kappaB) pathway. J Biol Chem 285(32):24639–24645PubMedPubMedCentralCrossRef Vigetti D, Genasetti A, Karousou E, Viola M, Moretto P, Clerici M et al (2010) Proinflammatory cytokines induce hyaluronan synthesis and monocyte adhesion in human endothelial cells through hyaluronan synthase 2 (HAS2) and the nuclear factor-kappaB (NF-kappaB) pathway. J Biol Chem 285(32):24639–24645PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Nagaoka A, Yoshida H, Nakamura S, Morikawa T, Kawabata K, Kobayashi M et al (2015) Regulation of Hyaluronan (HA) Metabolism Mediated by HYBID (Hyaluronan-binding Protein Involved in HA Depolymerization, KIAA1199) and HA Synthases in Growth Factor-stimulated Fibroblasts. J Biol Chem 290(52):30910–30923PubMedPubMedCentralCrossRef Nagaoka A, Yoshida H, Nakamura S, Morikawa T, Kawabata K, Kobayashi M et al (2015) Regulation of Hyaluronan (HA) Metabolism Mediated by HYBID (Hyaluronan-binding Protein Involved in HA Depolymerization, KIAA1199) and HA Synthases in Growth Factor-stimulated Fibroblasts. J Biol Chem 290(52):30910–30923PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Yoo SA, Park BH, Park GS, Koh HS, Lee MS, Ryu SH et al (2006) Calcineurin is expressed and plays a critical role in inflammatory arthritis. J Immunol 177(4):2681–2690PubMedCrossRef Yoo SA, Park BH, Park GS, Koh HS, Lee MS, Ryu SH et al (2006) Calcineurin is expressed and plays a critical role in inflammatory arthritis. J Immunol 177(4):2681–2690PubMedCrossRef
50.
Zurück zum Zitat Yi JK, Kim HJ, Yu DH, Park SJ, Shin MJ, Yuh HS et al (2012) Regulation of inflammatory responses and fibroblast-like synoviocyte apoptosis by calcineurin-binding protein 1 in mice with collagen-induced arthritis. Arthritis Rheum 64(7):2191–2200PubMedCrossRef Yi JK, Kim HJ, Yu DH, Park SJ, Shin MJ, Yuh HS et al (2012) Regulation of inflammatory responses and fibroblast-like synoviocyte apoptosis by calcineurin-binding protein 1 in mice with collagen-induced arthritis. Arthritis Rheum 64(7):2191–2200PubMedCrossRef
Metadaten
Titel
Association analysis of juvenile idiopathic arthritis genetic susceptibility factors in Estonian patients
verfasst von
Tiit Nikopensius
Priit Niibo
Toomas Haller
Triin Jagomägi
Ülle Voog-Oras
Neeme Tõnisson
Andres Metspalu
Mare Saag
Chris Pruunsild
Publikationsdatum
08.06.2021
Verlag
Springer International Publishing
Erschienen in
Clinical Rheumatology / Ausgabe 10/2021
Print ISSN: 0770-3198
Elektronische ISSN: 1434-9949
DOI
https://doi.org/10.1007/s10067-021-05756-x

Weitere Artikel der Ausgabe 10/2021

Clinical Rheumatology 10/2021 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.