Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Polymorphisms in the Inflammatory Pathway Genes TLR2, TLR4, TLR9, LY96, NFKBIA, NFKB1, TNFA, TNFRSF1A, IL6R, IL10, IL23R, PTPN22, and PPARG Are Associated with Susceptibility of Inflammatory Bowel Disease in a Danish Cohort

  • Steffen Bank ,

    stb@mb.au.dk

    Affiliations Medical Department, Viborg Regional Hospital, Viborg, Denmark, Biomedicine, University of Aarhus, Aarhus, Denmark

  • Paal Skytt Andersen,

    Affiliation Microbiology and Infection Control, Statens Serum Institut, Copenhagen, Denmark

  • Johan Burisch,

    Affiliation Department of Gastroenterology, Herlev Hospital, Herlev, Denmark

  • Natalia Pedersen,

    Affiliation Department of Gastroenterology, Herlev Hospital, Herlev, Denmark

  • Stine Roug,

    Affiliation Department of Gastroenterology, Hvidovre Hospital, Hvidovre, Denmark

  • Julie Galsgaard,

    Affiliation Medical Department, Køge Hospital, Køge, Denmark

  • Stine Ydegaard Turino,

    Affiliation Medical Department, Hillerød Hospital, Hillerød, Denmark

  • Jacob Broder Brodersen,

    Affiliation Medical Department, Sydvestjysk Hospital, Esbjerg, Denmark

  • Shaista Rashid,

    Affiliation Medical Department, Bispebjerg Hospital, Bispebjerg, Denmark

  • Britt Kaiser Rasmussen,

    Affiliation Medical Department, Nykøbing Falster Hospital, Nykøbing Falster, Denmark

  • Sara Avlund,

    Affiliation Medical Department V, Aarhus University Hospital, Aarhus, Denmark

  • Thomas Bastholm Olesen,

    Affiliation Medical Department, Slagelse Hospital, Slagelse, Denmark

  • Hans Jürgen Hoffmann,

    Affiliation Department of Respiratory Diseases B, Institute for Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark

  • Marianne Kragh Thomsen,

    Affiliation Department of Clinical Microbiology, Aarhus University Hospital, Aarhus, Denmark

  • Vibeke Østergaard Thomsen,

    Affiliation International Reference Laboratory of Mycobacteriology, Statens Serum Institut, Copenhagen, Denmark

  • Morten Frydenberg,

    Affiliation Section of Biostatistics, Department of Public health, Aarhus University, Aarhus, Denmark

  • Bjørn Andersen Nexø,

    Affiliation Biomedicine, University of Aarhus, Aarhus, Denmark

  • Jacob Sode,

    Affiliations Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark, Clinical Biochemistry, Immunology & Genetics, Statens Serum Institut, Copenhagen, Denmark, Department of Rheumatology, Frederiksberg Hospital, Frederiksberg, Denmark

  • Ulla Vogel,

    Affiliation National Research Centre for the Working Environment, Copenhagen, Denmark

  •  [ ... ],
  • Vibeke Andersen

    Affiliations Medical Department, Viborg Regional Hospital, Viborg, Denmark, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark, Organ Centre, Hospital of Southern Jutland Aabenraa, Aabenraa, Denmark, OPEN Odense Patient data Explorative Network, Odense University Hospital, Odense, Denmark

  • [ view all ]
  • [ view less ]

Abstract

Background

The inflammatory bowel diseases (IBD), Crohn's disease (CD) and ulcerative colitis (UC), result from the combined effects of susceptibility genes and environmental factors. Polymorphisms in genes regulating inflammation may explain part of the genetic heritage.

Methods

Using a candidate gene approach, 39 mainly functional single nucleotide polymorphisms (SNPs) in 26 genes regulating inflammation were assessed in a clinical homogeneous group of severely diseased patients consisting of 624 patients with CD, 411 patients with UC and 795 controls. The results were analysed using logistic regression.

Results

Sixteen polymorphisms in 13 genes involved in regulation of inflammation were associated with risk of CD and/or UC (p≤0.05). The polymorphisms TLR2 (rs1816702), NFKB1 (rs28362491), TNFRSF1A (rs4149570), IL6R (rs4537545), IL23R (rs11209026) and PTPN22 (rs2476601) were associated with risk of CD and the polymorphisms TLR2 (rs1816702), TLR4 (rs1554973 and rs12377632), TLR9 (rs352139), LY96 (rs11465996), NFKBIA (rs696), TNFA (rs1800629), TNFRSF1A (rs4149570), IL10 (rs3024505), IL23R (rs11209026), PTPN22 (rs2476601) and PPARG (rs1801282) were associated with risk of UC. When including all patients (IBD) the polymorphisms TLR2 (rs4696480 and rs1816702), TLR4 (rs1554973 and rs12377632), TLR9 (rs187084), TNFRSF1A (rs4149570), IL6R (rs4537545), IL10 (rs3024505), IL23R (rs11209026) and PTPN22 (rs2476601) were associated with risk. After Bonferroni correction for multiple testing, both the homozygous and the heterozygous variant genotypes of IL23R G>A(rs11209026) (ORCD,adj: 0.38, 95% CI: 0.21–0.67, p = 0.03; ORIBD,adj 0.43, 95% CI: 0.28–0.67, p = 0.007) and PTPN22 1858 G>A(rs2476601) (ORCD,unadj 0.54, 95% CI: 0.41–0.72, p = 7*10−4; ORIBD,unadj: 0.61, 95% CI: 0.48–0.77, p = 0.001) were associated with reduced risk of CD.

Conclusion

The biological effects of the studied polymorphisms suggest that genetically determined high inflammatory response was associated with increased risk of CD. The many SNPs found in TLRs suggest that the host microbial composition or environmental factors in the gut are involved in risk of IBD in genetically susceptible individuals.

Introduction

Chronic inflammatory bowel diseases (IBDs), Crohn's disease (CD) and ulcerative colitis (UC), are complex diseases that result from the interaction of numerous genetic and environmental factors [1].

Genetic association studies have identified innate immunity as a critical component in the development of IBD. Until now, more than 163 IBD susceptibility polymorphisms have been confirmed, most of which are associated with both CD and UC, by candidate and genome wide association studies (GWAS) [2][11]. However, these polymorphisms have been estimated to only account for 20% of the genetic heritage involved in IBD [12].

Functional polymorphisms in genes in the inflammatory pathways may explain some of the genetic heritage involved in IBD. The transcription factor NFκB is a central regulator of inflammation. NFκB can be activated by Toll like receptors (TLRs). TLRs recognize pathogen-associated molecular patterns (PAMPs) that are broadly shared by pathogens but distinguishable from host molecules such as bacterial or viral DNA, flagellin or lipopolysaccharide (LPS). The TLRs initiates a kinase cascade that ultimately activates the IKK-complex, which phosphorylates and degrades the NFκB inhibitor IκBα. NFκB is shuttled from the cytosol to the nucleus where it initiates expression of pro- and anti-inflammatory cytokines including TNF-α, IL-6 and IL-10 [13].

To identify susceptibility loci we assessed 39 mainly functional polymorphisms in genes involved in inflammation, particular in the NFκB pathway, in a homogeneous Danish cohort of 624 patients with severe CD, 411 patients with severe UC and 795 healthy controls. The candidate gene approach using functional polymorphisms allows interpretation of the underlying biological mechanisms based on increased or decreased gene expression or protein activity.

Functional polymorphisms in genes in the inflammatory pathways involved in regulation of the NFκB pathway (TLR2, TLR4, TLR5, TLR9, LY96, CD14, MAP3K14, SUMO4, NFKBIA and NFKB1), TNF-α signaling (TNFA, TNFRSF1A and TNFAIP3), cytokines regulated by NFκB (IL1B, IL1RN, IL6, IL10, IL17A and IFNG) and other genes involved in regulation of inflammation (IL4R, IL6R, IL23R, TGFB1, PTPN22, PPARG and NLRP3) were studied, as genetically determined variation in the inflammatory pathways may be associated with severe disease among patients with CD and UC.

Materials and Methods

Cohort

A prior anti-TNF naïve Danish cohort of patients with IBD was established. In short, blood samples retrieved as part of the routine screening for latent Mycobacterium tuberculosis at Statens Serum Institut (SSI, Copenhagen, Denmark) and the Department of Respiratory Diseases B or the Department Clinical Microbiology, Aarhus University Hospital (Aarhus, Denmark) were collected from 01.09.2009 to 30.03.2011 (9217 patients). Patients with intestinal diseases (ICD-10 code K50–K63) were identified by linking the unique personal identification number of Danish citizens (CPR-number) from each blood sample with the National Patient Registry (2659 patients). Patient records from 18 medical departments were examined (1378 patients) and identified 1035 ethnic Danish patients with IBD where blood and clinical data were available. The patients either received or were considered candidates to anti-tumor necrosis factor-α (TNF-α) therapy (infliximab or adalimumab). The control group consisted of 795 healthy blood donors recruited from Viborg, Denmark [4].

Selection of polymorphisms

A candidate gene approach was used with focus on polymorphisms in the TNF-α and NFκB pathways. In addition, polymorphisms in genes which have been shown to be associated with CD and/or UC, polymorphisms in inflammatory cytokines and polymorphisms in TLR2 and TLR4 were included [14].

Functional polymorphisms in relevant genes were found by searching pubmed with “polymorphism AND gene-name AND (reporter gene OR luciferase OR ELISA OR enzyme-linked immunosorbent assay OR RT-PCR OR reverse transcriptase PCR OR EMSA OR electrophoretic mobility shift assay OR flow cytometry)”.

Genotyping

For patients with IBD the DNA was extracted from cryopreserved blood clots by using the Maxwell 16 Blood purification kit (Promega) according to the manufacturers' instructions with a median yield of 4.90 µg (range 0.8–25 µg) pr 300 µl total blood [15]. For the healthy controls, DNA was extracted from EDTA-stabilized peripheral blood by either PureGene (Qiagen, Hilden, Germany) or Wizard Genomic (Promega, Madison, Wisconsin, USA) DNA purification kit according to the manufacturers' instructions [4]. Competitive Allele-Specific Polymerase chain reaction (KASP), an end-point PCR technology, was used by LGC Genomics for genotyping (LGC Genomics , Hoddesdon, United Kingdom) (http://www.lgcgenomics.com/). The SNPs studied were TLR2 (rs4696480, rs1816702, rs11938228, rs3804099), TLR4 (rs12377632, rs5030728, rs1554973), TLR5 (rs5744168), TLR9 (rs187084, rs352139), LY96 (MD-2) (rs11465996), CD14 (rs2569190), MAP3K14 (NIK) (rs7222094), SUMO4 (rs237025), NFKBIA (IκBα) (rs696, rs17103265), NFKB1 (NFκB1) (rs28362491), TNFA (TNF-α) (rs1800629, rs1800630, rs1799724, rs361525), TNFRSF1A (TNFR1) (rs4149570), TNFAIP3 (A20) (rs6927172), IL1B (IL-1β) (rs1143623, rs4848306, rs1143627), IL-1RN (IL-1RA) (rs4251961), IL4R (rs1805010), IL6 (rs10499563), IL6R (rs4537545), IL10 (rs1800872, rs3024505), IL17A (rs2275913), IL23R (rs11209026), IFNG (IFN-γ) (rs2430561), TGFB1 (TGF-β1) (rs1800469), PTPN22 (rs2476601), PPARG (PPAR-γ) (rs1801282) and NLRP3 (rs4612666).

Genotyping of TNFA (TNF-α) −857 C>T (rs1799724) and −863 C>A (rs1800630) failed due to their close proximity to each other. All genotyping of −857 C>T (rs1799724) either failed or were erroneously genotyped as homozygous wild type when the patients were carriers of the AA genotype of −863 C>A (rs1800630) due to genotyping bias.

The 39 genotypes were replicated in 94 randomly selected samples and yielded >99% identical genotypes.

Statistical analysis

Logistic regression was used to compare genotype distributions among patients with CD, UC and IBD versus healthy controls (Table S1 and S2). Crude odds ratio and odds ratio adjusted for age, gender and smoking status were assessed. A chi-square test was used to test for deviation from Hardy-Weinberg equilibrium in the healthy controls and for haplotype analysis (Table S3, S4, S5).

Statistical analyses were performed using STATA version 11 (STATA Corp., Texas, USA).

Ethics statement

The study was conducted in accordance with the Declaration of Helsinki and was approved by the Regional Ethics Committees of Central (M20100153) and Southern (S-20120113) Denmark and the Danish Data Protection Agency of Central (RM: J. 2010-41-4719) and Southern (RSD: 2008-58-035) Denmark. The Ethics Committees gave suspension for obtaining written informed consent.

Results

Study population

Characteristics of the Danish patients with CD, UC and healthy controls are shown in Table 1.

The genotype distributions among the healthy controls deviated from Hardy-Weinberg equilibrium for TLR2 (−16934 A>T (rs4696480)) (p = 0.02), TLR4 (rs1554973 T>C) (p = 0.03), TLR9 (1174 G>A (rs352139)) (p = 0.02), and TGFB1 (−509 C>T (rs1800469)) (p = 0.02). None of the deviations remained statistically significant after correction for multiple testing.

Polymorphisms associated with risk of CD

The homozygous variant genotype of TLR2 C>T (rs1816702) (ORadj: 2.80, 95% CI: 1.03–7.62, p = 0.04), TNFRSF1A −609 G>T (rs4149570) (ORadj: 1.84, 95% CI: 1.19–2.84, p = 0.01) and IL6R C>T (rs4537545) (ORadj: 1.73, 95% CI: 1.12–2.66, p = 0.01) were associated with increased risk of CD. Both the homozygous and the heterozygous variant genotypes of NFKB1 −94ins/del (rs28362491) (ORunadj: 0.80, 95% CI: 0.65–1.00, p = 0.05), IL23R G>A (rs11209026) (ORadj: 0.38, 95% CI: 0.21–0.67, p = 9*10−4) and PTPN22 1858 G>A (rs2476601) (ORadj: 0.57, 95% CI: 0.39–0.83, p = 4*10−3) were associated with reduced risk of CD (Table S1 and S2).

After Bonferroni correction for multiple testing both the homozygous and the heterozygous variant genotypes of IL23R G>A (rs11209026) (ORadj: 0.38, 95% CI: 0.21–0.67, p = 0.03) and PTPN22 1858 G>A (rs2476601) (ORunadj: 0.54, 95% CI: 0.41–0.72, p = 7*10−4) were associated with reduced risk of CD.

The variant allele of the polymorphisms have been shown to increase TLR2 levels (TLR2 C>T (rs1816702)), increase TNFRSF1A expression (TNFRSF1A −609 G>T (rs4149570)), increase IL-6r and IL-6 levels (IL6R C>T (rs4537545)), decrease NF-κB p50 subunit expression (NFKB1 −94ins/del (rs28362491)), decrease IL-17 serum levels (IL23R G>A (rs11209026)) and decrease TNF-α serum levels (PTPN22 1858 G>A (rs2476601)), respectively (Table 2).

thumbnail
Table 2. The biologic effect of the studied single nucleotide polymorphism (SNP) and odds ratios (OR) for polymorphisms which have been shown to be associated with risk of Crohn's disease (CD), ulcerative colitis (UC) or inflammatory bowel disease (IBD) in previous studies and in this study.

https://doi.org/10.1371/journal.pone.0098815.t002

Thus, polymorphisms associated with higher TLR2 levels (TLR2 C>T (rs1816702)), increased TNFRSF1A expression (TNFRSF1A −609 G>T (rs4149570)) and higher IL-6r and IL-6 levels (IL6R C>T (rs4537545)) were associated with increased risk of CD. In addition, decreased IL-17 serum levels (IL23R G>A (rs11209026)) and decreased TNF-α serum levels (PTPN22 1858 G>A (rs2476601)) were associated with reduced risk of CD. The NF-κB p50 subunit can act both pro-inflammatory as part of the p50/p65 complex or anti-inflammatory as p50 homodimer (Table 2). A lowered NF-κB p50 subunit expression (NFKB1 −94ins/del (rs28362491)) could be a differential lowering of the anti-inflammatory response which was associated with reduced risk of CD.

Polymorphisms associated with risk of UC

The homozygous variant genotype of TNFRSF1A −609 G>T (rs4149570) (ORadj: 1.85, 95% CI: 1.16–2.97, p = 0.01) and PPARG C>G (rs1801282) (ORunadj: 2.12, 95% CI: 1.01–4.45, p = 0.05) and both the homozygous and the heterozygous variant genotypes of TLR2 C>T (rs1816702) (ORadj: 1.63, 95% CI: 1.13–2.36, p = 0.01), TLR4 T>C (rs12377632) (ORadj: 1.42, 95% CI: 1.01–2.00, p = 0.04), NFKBIA 2758 A>G (rs696) (ORadj: 1.45, 95% CI: 1.03–2.05, p = 0.03) and IL10 C>T (rs3024505) (ORunadj: 1.42, 95% CI: 1.10–1.82, p = 0.01) were associated with increased risk of UC.

The homozygous variant genotype of TLR9 1174 G>A (rs352139) (ORunadj: 0.67, 95% CI: 0.47–0.95, p = 0.03) and the heterozygous genotype of LY96 −1625 C>G (rs11465996) (ORunadj: 0.74, 95% CI: 0.57–0.96, p = 0.02) were associated with reduced risk of UC. In addition, both the homozygous and the heterozygous variant genotypes of TLR4 T>C (rs1554973) (ORadj: 0.67, 95% CI: 0.48–0.94, p = 0.02), TNFA −308 G>A (rs1800629) (ORunadj: 0.75, 95% CI: 0.58–0.98, p = 0.04), IL23R G>A (rs11209026) (ORadj: 0.52, 95% CI: 0.29–0.94, p = 0.03) and PTPN22 1858 G>A (rs2476601) (ORunadj: 0.71, 95% CI: 0.52–0.96, p = 0.03) were associated with reduced risk of UC (Table S1 and S2). No associations were found after Bonferroni correction for multiple testing.

The variant allele of the polymorphisms have been shown to increase TNFRSF1A expression (TNFRSF1A −609 G>T (rs4149570)), increase TLRs and TNF-α mRNA levels (PPARG C>G (rs1801282)), increase TLR2 levels (TLR2 C>T (rs1816702)), increase NFKBIA expression (NFKBIA 2758 A>G (rs696)), increase MD-2 (LY96) and TNF-α levels (LY96 −1625 C>G (rs11465996)), increases TNFA expression (TNFA −308 G>A (rs1800629)), decrease IL-17 serum levels (IL23R G>A (rs11209026)) and decrease TNF-α serum levels (PTPN22 1858 G>A (rs2476601)), respectively (Table 2). The biological function of the polymorphisms TLR4 T>C (rs12377632), TLR4 T>C (rs1554973), TLR9 1174 G>A (rs352139) and IL10 C>T (rs3024505) are unknown.

Thus, polymorphisms associated with increased TNFRSF1A expression (TNFRSF1A −609 G>T (rs4149570)), increased TLRs and TNF-α mRNA levels (PPARG C>G (rs1801282)) and increased TLR2 levels (TLR2 C>T (rs1816702)) were associated with increased risk of UC. Furthermore, polymorphisms associated with decreased IL-17 serum levels (IL23R G>A (rs11209026)) and decreased TNF-α serum levels (PTPN22 1858 G>A (rs2476601)) were associated with reduced risk of UC. In contrary, lower activity of the NFκB pathway through increased NFKBIA (IκBα) expression (NFKBIA 2758 A>G (rs696)), an inhibitor of the NFκB pathway, was associated with increased risk of UC. In addition, polymorphisms associated with increased MD-2 (LY96 −1625 C>G (rs11465996)) and TNF-α levels (TNFA −308 G>A (rs1800629)) were associated with reduced risk of UC.

Polymorphisms associated with risk of IBD

The studied polymorphisms all showed the same direction of effect for both diseases except for the polymorphism in PPARG (rs1801282) (Table S1 and S2).

When including all patients (IBD), the homozygous variant genotype of TLR2 A>T (rs4696480) (ORunadj: 1.33, 95% CI: 1.01–1.74, p = 0.04) and IL6R C>T (rs4537545) (ORadj: 1.46, 95% CI: 1.02–2.08, p = 0.04) and both the homozygous and the heterozygous variant genotypes of TLR2 C>T (rs1816702) (ORadj: 1.42, 95% CI: 1.08–1.88, p = 0.01), TLR4 T>C (rs12377632) (ORadj: 1.28, 95% CI: 1.01–1.64, p = 0.05), TLR9 −1486 T>C (rs187084) (ORadj: 1.29, 95% CI: 1.00–1.66, p = 0.05), TNFRSF1A −609 G>T (rs4149570) (ORadj: 1.32, 95% CI: 1.03–1.68, p = 0.03) and IL10 C>T (rs3024505) (ORunadj: 1.25, 95% CI: 1.02–1.52, p = 0.03) were associated with increased risk of IBD. The homozygous variant genotype of TLR4 T>C (rs1554973) (ORunadj: 0.67, 95% CI: 0.46–0.98, p = 0.04) and both the homozygous and the heterozygous variant genotypes of IL23R G>A (rs11209026) (ORadj: 0.43, 95% CI: 0.28–0.67, p = 2*10−4) and PTPN22 1858 G>A (rs2476601) (ORadj: 0.62, 95% CI: 0.46–0.85, p = 3*10−3) were associated with reduced risk of IBD (Table S1 and S2).

After Bonferroni correction for multiple testing both the homozygous and the heterozygous variant genotypes of IL23R G>A (rs11209026) (ORadj: 0.43, 95% CI: 0.28–0.67, p = 0.007) and PTPN22 1858 G>A (rs2476601) (ORunadj: 0.61, 95% CI: 0.48–0.77, p = 0.001) were associated with reduced risk of IBD.

The variant allele of the polymorphisms have been shown to increase IL-6r and IL-6 levels (IL6R C>T (rs4537545)), increase TLR2 levels (TLR2 C>T (rs1816702)), increase TNFRSF1A expression (TNFRSF1A −609 G>T (rs4149570)), decrease IL-17 serum levels (IL23R G>A (rs11209026)) and decrease TNF-α serum levels (PTPN22 1858 G>A (rs2476601)), respectively (Table 2). The biological function of the polymorphisms TLR2 A>T (rs4696480), TLR4 T>C (rs12377632), TLR9 −1486 T>C (rs187084), TLR4 T>C (rs1554973) and IL10 C>T (rs3024505) are unknown.

Thus, polymorphisms associated with higher IL-6r and IL-6 levels (IL6R C>T (rs4537545)), higher TLR2 levels (TLR2 C>T (rs1816702)) and increased TNFRSF1A expression (TNFRSF1A −609 G>T (rs4149570)) were associated with increased risk of IBD. In addition, polymorphisms associated with decreased IL-17 serum levels (IL23R G>A (rs11209026)) and decreased TNF-α serum levels (PTPN22 1858 G>A (rs2476601)) were associated with reduced risk of IBD.

Haplotype analysis

Haplotype analyses of TLR2, TLR4 and TLR9 among patients with CD, UC and IBD versus healthy controls are shown in Table S3, S4, S5, respectively. Four haplotypes in TLR2, three in TLR4 and two in TLR9 described 88%, 94% and 97% of the observed genotypes, respectively.

The TLR2 haplotype combination 33 encompassing all the wildtype alleles (rs4696480AA, rs11938228CC, rs1816702CC and rs3804099TT) was associated with reduced risk of CD (OR: 0.19, 95% CI: 0.08–0.45, p = 0.00007) and combined CD and UC (OR: 0.32, 95% CI: 0.17–0.60, p = 0.0005).

No associations were found for TLR4.

The TLR9 haplotype combination 11 (rs187084CC and rs352139GG) was associated with increased risk of UC (OR: 1.53, 95% CI: 1.03–2.28, p = 0.04). The TLR9 haplotype combination 12 was associated with increased risk of IBD (OR: 1.35, 95% CI: 1.04–1.76, p = 0.03). Indirectly, these results support the analysis of the individual SNPs, where the variant allele of rs352139 (included in haplotype combination 22 which was used as reference) was fond to be associated with lowered risk of UC.

Discussion

In this study of severely ill patients, 16 functional polymorphisms in 13 genes involved in regulation of inflammation were found to be associated with CD, UC or CD and UC combined (IBD) (Figure 1). As shown in Table 2, four known susceptibility loci for CD (PTPN22 (1858 G>A) [16]), UC (IL10 (rs3024505 C>T) [3], [17] and PPARG (rs1801282 C>G) [18]) or CD and UC (IL23R (rs11209026 G>A) [16], [17]) were replicated. Eleven new polymorphisms associated with risk of CD (IL6R (rs4537545 C>T)), UC (TLR4 (rs1554973 T>C and rs12377632 T>C), TLR9 (1174 G>A), LY96 (−1625 C>G), NFKBIA (2758 A>G) and PTPN22 (1858 G>A)) or CD and UC (TLR2 (−16934 A>T and rs1816702 C>T), TLR9 (−1486 T>C) and TNFRSF1A (−609 G>T)) were identified. Other cohort studies of patients with CD and UC have found that other polymorphisms in the TLRs (toll like receptors) [19] and TNFRSFs (TNF receptors) [16], [17], [20], [21] were associated with CD or UC.

thumbnail
Figure 1. Sixteen functional single nucleotide polymorphisms (SNPs) in 13 genes involved in regulation of inflammation were found to be associated with susceptability of severe Crohn's disease (CD), ulcerative colitis (UC) or inflammatory bowel diseases (IBD).

Eleven of the SNPs have not previously been reported as susceptability polymorphisms of CD, UC or IBD (TLR2 (rs4696480 and rs1816702), TLR4 (rs1554973 and rs12377632), TLR9 (rs187084 and rs352139), LY96 (rs11465996), NFKBIA (rs696), TNFRSF1A (rs4149570), IL6R (rs4537545) and PTPN22 (rs2476601)).

https://doi.org/10.1371/journal.pone.0098815.g001

The biological interpretation indicates that a genetically determined higher activity of the inflammatory genes TLR2 (rs1816702 C>T), TNFRSF1A (−609 G>T) and IL6R (rs4537545 C>T) was associated with increased risk of CD and lower activity of the inflammatory genes NFKB1 (NFκB) (−94ins/del ATTG), IL23R (rs11209026 G>A) and PTPN22 (1858 G>A) was associated with reduced risk of CD (Table 2). The picture was less clear for UC. A genetically determined higher activity of the inflammatory genes TNFRSF1A (−609 G>T), PPARG (rs1801282 C>G) and TLR2 (rs1816702 C>T) was associated with increased risk of UC and lower activity of the inflammatory genes IL23R (rs11209026 G>A) and PTPN22 (1858 G>A) was associated with reduced risk of UC. In contrast, a genetically determined higher activity of the NFκB inhibitor IκBα (NFKBIA 2758 A>G) and lower activity of TLR9 (haplotype 11 (−1486CC and 1174GG)) were associated with increased risk of UC. Furthermore, a genetically determined higher activity of LY96 (−1625 C>G) and TNFA (−308 G>A) was associated with reduced risk of UC. However, the risk of CD and UC seem to have shared mechanisms through the TLR2 (rs1816702 C>T), TNFRSF1A (−609 G>T), IL23R (rs11209026 G>A) and PTPN22 (1858 G>A) polymorphisms confirming that the inflammatory pathways are involved in risk of both CD and UC [16], [17].

This study was unable to confirm the associations between the variant allele of IL10 (rs3024505 C>T) and increased risk of CD [3], [16] or the associations between the variant allele of CD14 (−159 G>A) and increased risk of CD, UC and IBD [22]. This study may be underpowered to detect an association in IL10 (rs3024505 C>T), as a GWA study with more than six thousands CD cases and fifteen thousands control found that the varaint allele was associated with increased risk of CD but with an odds ratio of 1.12 [16]. However, IL10 has previously been found to be associated with risk of CD and UC in the Danish population [3]. The difference of association in CD14 (−159 G>A) may be due to genetic differences between the Korean and Danish population.

The variant allele in NFKBIA (IκBα) (2758 A>G) [23][25] and TNFA (−308 G>A) were associated with higher and lower risk of UC, respectively, and the deletion polymorphism in NFKB1 (NFκB) (−94ins/del ATTG) was associated with lower risk of CD in our cohort study. However, there seems to be no consensus regarding these polymorphisms in other cohort studies [24][27]. TLR5stop (1174 C>T) has been found to be associated with reduced risk of CD in a Jewish cohort [28] but not in a non-Jewish and German cohort [28], [29]. No association with TLR5stop was found in this cohort study.

No associations were found for IL1B (−31 T>C) [3], IL-10 (−592 C>A) [3] or PPARG (rs1801282 C>G) [18] in accordance with our previous Danish IBD cohort study using independent cases but the same control group [3]. The other polymorphisms studied in MAP3K14 (rs7222094 T>C), SUMO4 (163 T>C), TNFAIP3 (rs6927172 C>G), IL1RN (rs4251961 T>C), IL4R (rs1805010 A>G), IL6 (−6331 T>C), IL17A (197G>A), IFNG (874 T>A), TGFB1 (−509 C>T) and NLRP3 (rs4612666 C>T) were not statistically associated with CD, UC or IBD in our cohort. None of the polymorphisms studied in these genes have been reported to be associated with CD or UC. We can not exclude that we did not find associations between these polymorphisms and risk of disease due to lack of power in this cohort.

The results in this study should be interpreted with care. TLR2 (rs4696480 A>T), TLR4 (rs1554973 T>C), TLR9 (1174 G>A) and TGFB1 (−509 C>T) were not in Hardy-Weinberg equilibrium among the healthy controls which is probable due to chance because of the number of polymorphisms analyzed. When corrected adequately for multiple testing they did not deviate from Hardy-Weinberg equilibrium. In the light of the obtained P-values and the number of statistical tests performed, we cannot exclude that some of our positive findings may be due to chance. If the results were corrected for multiple testing only the well known susceptibility polymorphisms in IL23R (rs11209026 G>A) and PTPN22 (1858 G>A) were associated with reduced risk of both CD and IBD. We successfully tested 37 polymorphisms and, assuming a 5% acceptance level, two would be expected to be associated with susceptibility by pure chance. In this study 16 polymorphisms were found to be associated with susceptibility and the found associations were biologically plausible. A major strength was that this clinically homogeneous and well-characterised cohort was rather large including 1035 patients with IBD and 795 healthy controls. All the patients were considered for anti-TNF treatment and were therefore considered to have a severe disease course. Genetic determinants may be expected to be strong among severely ill cases [30].

In conclusion, 16 functional SNPs in 13 genes involved in regulation of inflammation were found to be associated with susceptibility of severe CD, UC or IBD. Eleven of the SNPs have not previously been reported as susceptibility polymorphisms of CD, UC or IBD (Figure 1) although other polymorphisms in most of these genes have previously been associated with susceptibility of CD, UC or IBD. Our results suggest that genetically determined high inflammatory response was associated with increased risk of CD and the large number of polymorphisms in the TLRs associated with risk of CD or UC support that the host microbial composition, diet or environmental molecules in the gut are important factors driving the inflammatory response in genetically susceptible individuals.

Supporting Information

Table S1.

Odds ratios (OR) (unadjusted) for genotypes studied among healthy controls and patients with Crohn's disease (CD), ulcerative colitis (UC) and combined inflammatory bowel disease (IBD).

https://doi.org/10.1371/journal.pone.0098815.s001

(DOC)

Table S2.

Odds ratios (OR) (adjusted for age, sex and smoking status) for genotypes studied among healthy controls and patients with Crohn's disease (CD), ulcerative colitis (UC) and combined inflammatory bowel disease (IBD).

https://doi.org/10.1371/journal.pone.0098815.s002

(DOC)

Table S3.

Association of the TLR2 haplotype combinations and risk of Crohn's disease (CD), ulcerative colitis (UC) and all inflammatory bowel disease (IBD).

https://doi.org/10.1371/journal.pone.0098815.s003

(DOC)

Table S4.

Association between TLR4 haplotype combinations and risk of Crohn's disease (CD), ulcerative colitis (UC) and all inflammatory bowel disease (IBD).

https://doi.org/10.1371/journal.pone.0098815.s004

(DOC)

Table S5.

Association between TLR9 haplotype combinations and risk of Crohn's disease (CD), ulcerative colitis (UC) and all inflammatory bowel disease (IBD).

https://doi.org/10.1371/journal.pone.0098815.s005

(DOC)

Author Contributions

Conceived and designed the experiments: SB PSA MF BAN JS UV VA. Performed the experiments: SB JB NP S. Roug JG SYT JBB S. Rashid BKR SA TBO HJH MKT VØT. Analyzed the data: SB PSA MF BAN JS UV VA. Contributed reagents/materials/analysis tools: SB JB NP S. Roug JG SYT JBB S. Rashid BKR SA TBO HJH MKT VØT MF. Wrote the paper: SB PSA UV VA. Drafting the article or revising it critically: SB PSA JB NP S. Roug JG SYT JBB S. Rashid BKR SA TBO HJH MKT VØT MF BAN JS UV VA. Final approval of the version to be published: SB PSA JB NP S. Roug JG SYT JBB S. Rashid BKR SA TBO HJH MKT VØT MF BAN JS UV VA.

References

  1. 1. Podolsky DK (2002) Inflammatory bowel disease. N Engl J Med 347: 417–429.
  2. 2. Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, et al. (2012) Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491: 119–124.
  3. 3. Andersen V, Ernst A, Christensen J, Ostergaard M, Jacobsen BA, et al. (2010) The polymorphism rs3024505 proximal to IL-10 is associated with risk of ulcerative colitis and Crohns disease in a Danish case-control study. BMC Med Genet 11: 82.
  4. 4. Ernst A, Jacobsen B, Ostergaard M, Okkels H, Andersen V, et al. (2007) Mutations in CARD15 and smoking confer susceptibility to Crohn's disease in the Danish population. Scand J Gastroenterol 1445–1451.
  5. 5. Ostergaard M, Ernst A, Labouriau R, Dagiliené E, Krarup HB, et al. (2009) Cyclooxygenase-2, multidrug resistance 1, and breast cancer resistance protein gene polymorphisms and inflammatory bowel disease in the Danish population. Scand J Gastroenterol 44: 65–73.
  6. 6. Andersen V, Nimmo E, Krarup HB, Drummond H, Christensen J, et al. (2011) Cyclooxygenase-2 (COX-2) polymorphisms and risk of inflammatory bowel disease in a Scottish and Danish case-control study. Inflamm Bowel Dis 937–946.
  7. 7. Ernst A, Andersen V, Ostergaard M, Jacobsen BA, Pedersen IS, et al. (2011) Common polymorphisms in the microsomal epoxide hydrolase and N-acetyltransferase 2 genes in association with inflammatory bowel disease in the Danish population. Eur J Gastroenterol Hepatol 269–274.
  8. 8. Andersen V, Christensen J, Ernst A, Jacobsen BA, Tjønneland A, et al. (2011) Polymorphisms in NF-kappaB, PXR, LXR, PPARgamma and risk of inflammatory bowel disease. World J Gastroenterol 197–206.
  9. 9. Ellinghaus D, Zhang H, Zeissig S, Lipinski S, Till A, et al. (2013) Association Between Variants of PRDM1 and NDP52 and Crohn's Disease, Based on Exome Sequencing and Functional Studies. Gastroenterology 339–347.
  10. 10. Skieceviciene J, Kiudelis G, Ellinghaus E, Balschun T, Jonaitis LV, et al. (2013) Replication Study of Ulcerative Colitis Risk Loci in a Lithuanian-Latvian Case-Control Sample. Inflamm Bowel Dis 11: 2349–2355.
  11. 11. Andersen V, Ernst A, Sventoraityte J, Kupcinskas L, Jacobsen BA, et al. (2011) Assessment of heterogeneity between European Populations: a Baltic and Danish replication case-control study of SNPs from a recent European ulcerative colitis genome wide association study. BMC Med Genet 12: 139.
  12. 12. Muise AM, Walters T, Xu W, Shen-Tu G, Guo CH, et al. (2011) Single nucleotide polymorphisms that increase expression of the guanosine triphosphatase RAC1 are associated with ulcerative colitis. Gastroenterology 141: 633–641.
  13. 13. Verstrepen L, Bekaert T, Chau TL, Tavernier J, Chariot A, et al. (2008) TLR-4, IL-1R and TNF-R signaling to NF-kappaB: variations on a common theme. Cell Mol Life Sci 65: 2964–2978.
  14. 14. Gast A, Bermejo JL, Claus R, Brandt A, Weires M, et al. (2011) Association of inherited variation in Toll-like receptor genes with malignant melanoma susceptibility and survival. PLoS One 6: e24370.
  15. 15. Bank S, Nexo BA, Andersen V, Vogel U, Andersen PS (2013) High-Quality and -Quantity DNA Extraction from Frozen Archival Blood Clots for Genotyping of Single-Nucleotide Polymorphisms. Genet Test Mol Biomarkers 6: 501–503.
  16. 16. Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, et al. (2010) Genome-wide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci. Nat Genet 42: 1118–1125.
  17. 17. Anderson CA, Boucher G, Lees CW, Franke A, D'Amato M, et al. (2011) Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat Genet 43: 246–252.
  18. 18. Andersen V, Christensen J, Ernst A, Jacobsen BA, Tjønneland A, et al. (2011) Polymorphisms in NF-kappaB, PXR, LXR, PPARgamma and risk of inflammatory bowel disease. World J Gastroenterol 17: 197–206.
  19. 19. Cario E (2010) Toll-like receptors in inflammatory bowel diseases: a decade later. Inflamm Bowel Dis 16: 1583–1597.
  20. 20. Sashio H, Tamura K, Ito R, Yamamoto Y, Bamba H, et al. (2002) Polymorphisms of the TNF gene and the TNF receptor superfamily member 1B gene are associated with susceptibility to ulcerative colitis and Crohn's disease, respectively. Immunogenetics 53: 1020–1027.
  21. 21. Ferguson LR, Han DY, Huebner C, Petermann I, Barclay ML, et al. (2009) Tumor necrosis factor receptor superfamily, member 1B haplotypes increase or decrease the risk of inflammatory bowel diseases in a New Zealand caucasian population. Gastroenterol Res Pract 591704
  22. 22. Kim EJ, Chung WC, Lee KM, Paik CN, Jung SH, et al. (2012) Association between toll-like receptors/CD14 gene polymorphisms and inflammatory bowel disease in Korean population. J Korean Med Sci 27: 72–77.
  23. 23. Szamosi T, Lakatos PL, Szilvasi A, Lakatos L, Kovacs A, et al. (2009) The 3′UTR NFKBIA variant is associated with extensive colitis in Hungarian IBD patients. Dig Dis Sci 54: 351–359.
  24. 24. Klein W, Tromm A, Folwaczny C, Hagedorn M, Duerig N, et al. (2004) A polymorphism of the NFKBIA gene is associated with Crohn's disease patients lacking a predisposing allele of the CARD15 gene. Int J Colorectal Dis 19: 153–156.
  25. 25. Hong J, Leung E, Fraser AG, Merriman TR, Vishnu P, et al. (2007) Polymorphisms in NFKBIA and ICAM-1 genes in New Zealand Caucasian Crohn's disease patients. J Gastroenterol Hepatol 22: 1666–1670.
  26. 26. Zou YF, Wang F, Feng XL, Tao JH, Zhu JM, et al. (2011) Association of NFKB1 -94ins/delATTG promoter polymorphism with susceptibility to autoimmune and inflammatory diseases: a meta-analysis. Tissue Antigens 77: 9–17.
  27. 27. Ferguson LR, Huebner C, Petermann I, Gearry RB, Barclay ML, et al. (2008) Single nucleotide polymorphism in the tumor necrosis factor-alpha gene affects inflammatory bowel diseases risk. World J Gastroenterol 14: 4652–4661.
  28. 28. Gewirtz AT, Vijay-Kumar M, Brant SR, Duerr RH, Nicolae DL, et al. (2006) Dominant-negative TLR5 polymorphism reduces adaptive immune response to flagellin and negatively associates with Crohn's disease. Am J Physiol Gastrointest Liver Physiol 290: G1157–G1163.
  29. 29. Glas J (2008) Functional Toll-like receptor 5 gene variants are not associated with susceptibility to inflammatory bowel disease in the German population. 46: 41.
  30. 30. Fowler EV, Doecke J, Simms LA, Zhao ZZ, Webb PM, et al. (2008) ATG16L1 T300A shows strong associations with disease subgroups in a large Australian IBD population: further support for significant disease heterogeneity. Am J Gastroenterol 2519–2526.
  31. 31. Bielinski SJ, Hall JL, Pankow JS, Boerwinkle E, Matijevic-Aleksic N, et al. (2011) Genetic variants in TLR2 and TLR4 are associated with markers of monocyte activation: the Atherosclerosis Risk in Communities MRI Study. Hum Genet 129: 655–662.
  32. 32. Zhang F, Gao XD, Wu WW, Gao Y, Zhang YW, et al. (2013) Polymorphisms in toll-like receptors 2, 4 and 5 are associated with Legionella pneumophila infection. Infection 41: 941–948.
  33. 33. Hawn TR, Verbon A, Lettinga KD, Zhao LP, Li SS, et al. (2003) A common dominant TLR5 stop codon polymorphism abolishes flagellin signaling and is associated with susceptibility to legionnaires' disease. J Exp Med 198: 1563–1572.
  34. 34. Tao K, Fujii M, Tsukumo S, Maekawa Y, Kishihara K, et al. (2007) Genetic variations of Toll-like receptor 9 predispose to systemic lupus erythematosus in Japanese population. Ann Rheum Dis 66: 905–909.
  35. 35. Gu W, Shan YA, Zhou J, Jiang DP, Zhang L, et al. (2007) Functional significance of gene polymorphisms in the promoter of myeloid differentiation-2. Ann Surg 246: 151–158.
  36. 36. Baldini M, Lohman IC, Halonen M, Erickson RP, Holt PG, et al. (1999) A Polymorphism* in the 5′ flanking region of the CD14 gene is associated with circulating soluble CD14 levels and with total serum immunoglobulin E. Am J Respir Cell Mol Biol 20: 976–983.
  37. 37. Mertens J, Bregadze R, Mansur A, Askar E, Bickeböller H, et al. (2009) Functional impact of endotoxin receptor CD14 polymorphisms on transcriptional activity. J Mol Med (Berl) 87: 815–824.
  38. 38. Thair SA, Walley KR, Nakada TA, McConechy MK, Boyd JH, et al. (2011) A single nucleotide polymorphism in NF-kappaB inducing kinase is associated with mortality in septic shock. J Immunol 186: 2321–2328.
  39. 39. Guo D, Li M, Zhang Y, Yang P, Eckenrode S, et al. (2004) A functional variant of SUMO4, a new I kappa B alpha modifier, is associated with type 1 diabetes. Nat Genet 36: 837–841.
  40. 40. Song S, Chen D, Lu J, Liao J, Luo Y, et al. (2011) NFkappaB1 and NFkappaBIA polymorphisms are associated with increased risk for sporadic colorectal cancer in a southern Chinese population. PLoS One 6: e21726.
  41. 41. Wang S, Zhang M, Zeng Z, Tian L, Wu K, et al. (2011) IkappaBalpha polymorphisms were associated with increased risk of gastric cancer in a southern Chinese population: a case-control study. Life Sci 88: 792–797.
  42. 42. Vogel U, Jensen MK, Due KM, Rimm EB, Wallin H, et al. (2011) The NFKB1 ATTG ins/del polymorphism and risk of coronary heart disease in three independent populations. Atherosclerosis 219: 200–204.
  43. 43. Park JY, Farrance IK, Fenty NM, Hagberg JM, Roth SM, et al. (2007) NFKB1 promoter variation implicates shear-induced NOS3 gene expression and endothelial function in prehypertensives and stage I hypertensives. Am J Physiol Heart Circ Physiol 293: H2320–H2327.
  44. 44. Udalova IA, Richardson A, Denys A, Smith C, Ackerman H, et al. (2000) Functional consequences of a polymorphism affecting NF-kappaB p50-p50 binding to the TNF promoter region. Mol Cell Biol 20: 9113–9119.
  45. 45. Ahirwar DK, Kesarwani P, Singh R, Ghoshal UC, Mittal RD (2012) Role of tumor necrosis factor-alpha (C-863A) polymorphism in pathogenesis of inflammatory bowel disease in Northern India. J Gastrointest Cancer 43: 196–204.
  46. 46. Lv K, Chen R, Cai Q, Fang M, Sun S (2006) Effects of a single nucleotide polymorphism on the expression of human tumor necrosis factor-alpha. Scand J Immunol 64: 164–169.
  47. 47. Karimi M, Goldie LC, Cruickshank MN, Moses EK, Abraham LJ (2009) A critical assessment of the factors affecting reporter gene assays for promoter SNP function: a reassessment of −308 TNF polymorphism function using a novel integrated reporter system. Eur J Hum Genet 17: 1454–1462.
  48. 48. Kaluza W, Reuss E, Grossmann S, Hug R, Schopf RE, et al. (2000) Different transcriptional activity and in vitro TNF-alpha production in psoriasis patients carrying the TNF-alpha 238A promoter polymorphism. J Invest Dermatol 114: 1180–1183.
  49. 49. Wang GB, Li CR, Yang J, Wen PQ, Jia SL (2011) A regulatory polymorphism in promoter region of TNFR1 gene is associated with Kawasaki disease in Chinese individuals. Hum Immunol 72: 451–457.
  50. 50. Elsby LM, Orozco G, Denton J, Worthington J, Ray DW, et al. (2011) Functional evaluation of TNFAIP3 (A20) in rheumatoid arthritis. Clin Exp Rheumatol 28: 708–714.
  51. 51. Yoshida M, Shiroiwa K, Mouri K, Ishiguro H, Supriyanto I, et al. (2012) Haplotypes in the expression quantitative trait locus of interleukin-1beta gene are associated with schizophrenia. Schizophr Res 140: 185–191.
  52. 52. Chen H, Wilkins LM, Aziz N, Cannings C, Wyllie DH, et al. (2006) Single nucleotide polymorphisms in the human interleukin-1B gene affect transcription according to haplotype context. Hum Mol Genet 15: 519–529.
  53. 53. Wen AQ, Gu W, Wang J, Feng K, Qin L, et al. (2010) Clinical relevance of IL-1beta promoter polymorphisms (−1470, −511, and −31) in patients with major trauma. Shock 33: 576–582.
  54. 54. Lind H, Haugen A, Zienolddiny S (2007) Differential binding of proteins to the IL1B −31 T/C polymorphism in lung epithelial cells. Cytokine 38: 43–48.
  55. 55. Rafiq S, Stevens K, Hurst AJ, Murray A, Henley W, et al. (2007) Common genetic variation in the gene encoding interleukin-1-receptor antagonist (IL-1RA) is associated with altered circulating IL-1RA levels. Genes Immun 8: 344–351.
  56. 56. Carrol ED, Payton A, Payne D, Miyajima F, Chaponda M, et al. (2011) The IL1RN promoter rs4251961 correlates with IL-1 receptor antagonist concentrations in human infection and is differentially regulated by GATA-1. J Immunol 186: 2329–2335.
  57. 57. Wallis SK, Cooney LA, Endres JL, Lee MJ, Ryu J, et al. (2011) A polymorphism in the interleukin-4 receptor affects the ability of interleukin-4 to regulate Th17 cells: a possible immunoregulatory mechanism for genetic control of the severity of rheumatoid arthritis. Arthritis Res Ther 13: R15.
  58. 58. Smith AJ, D'Aiuto F, Palmen J, Cooper JA, Samuel J, et al. (2008) Association of serum interleukin-6 concentration with a functional IL6 −6331T>C polymorphism. Clin Chem 54: 841–850.
  59. 59. Rafiq S, Frayling TM, Murray A, Hurst A, Stevens K, et al. (2007) A common variant of the interleukin 6 receptor (IL-6r) gene increases IL-6r and IL-6 levels, without other inflammatory effects. Genes Immun 8: 552–559.
  60. 60. Rees LE, Wood NA, Gillespie KM, Lai KN, Gaston K, et al. (2002) The interleukin-10-1082 G/A polymorphism: allele frequency in different populations and functional significance. Cell Mol Life Sci 59: 560–569.
  61. 61. Franke A, Balschun T, Karlsen TH, Sventoraityte J, Nikolaus S, et al. (2008) Sequence variants in IL10, ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility. Nat Genet 40: 1319–1323.
  62. 62. Espinoza JL, Takami A, Nakata K, Onizuka M, Kawase T, et al. (2011) A genetic variant in the IL-17 promoter is functionally associated with acute graft-versus-host disease after unrelated bone marrow transplantation. PLoS One 6: e26229.
  63. 63. Oosting M, ter Hofstede H, van de Veerdonk FL, Sturm P, Kullberg BJ, et al. (2011) Role of interleukin-23 (IL-23) receptor signaling for IL-17 responses in human Lyme disease. Infect Immun 79: 4681–4687.
  64. 64. Pravica V, Perrey C, Stevens A, Lee JH, Hutchinson IV (2000) A single nucleotide polymorphism in the first intron of the human IFN-gamma gene: absolute correlation with a polymorphic CA microsatellite marker of high IFN-gamma production. Hum Immunol 61: 863–866.
  65. 65. Shah R, Hurley CK, Posch PE (2006) A molecular mechanism for the differential regulation of TGF-beta1 expression due to the common SNP -509C-T (c. -1347C > T). Hum Genet 120: 461–469.
  66. 66. Kariuki SN, Crow MK, Niewold TB (2008) The PTPN22 C1858T polymorphism is associated with skewing of cytokine profiles toward high interferon-alpha activity and low tumor necrosis factor alpha levels in patients with lupus. Arthritis Rheum 58: 2818–2823.
  67. 67. Aoyagi Y, Nagata S, Kudo T, Fujii T, Wada M, et al. (2010) Peroxisome proliferator-activated receptor gamma 2 mutation may cause a subset of ulcerative colitis. Pediatr Int 52: 729–734.
  68. 68. Poliska S, Penyige A, Lakatos PL, Papp M, Palatka K, et al. (2012) Association of peroxisome proliferator-activated receptor gamma polymorphisms with inflammatory bowel disease in a Hungarian cohort. Inflamm Bowel Dis 18: 472–479.
  69. 69. Hitomi Y, Ebisawa M, Tomikawa M, Imai T, Komata T, et al. (2009) Associations of functional NLRP3 polymorphisms with susceptibility to food-induced anaphylaxis and aspirin-induced asthma. J Allergy Clin Immunol 124: 779–785.