Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jun 1.
Published in final edited form as: Nat Genet. 2012 Nov 11;44(12):1341–1348. doi: 10.1038/ng.2467

Identification of fifteen new psoriasis susceptibility loci highlights the role of innate immunity

Lam C Tsoi 1,*, Sarah L Spain 2,*, Jo Knight 3,4,*, Eva Ellinghaus 5,*, Philip E Stuart 6, Francesca Capon 2, Jun Ding 1, Yanming Li 1, Trilokraj Tejasvi 6, Johann E Gudjonsson 6, Hyun M Kang 1, Michael H Allen 2, Ross McManus 7,8, Giuseppe Novelli 9,10, Lena Samuelsson 11, Joost Schalkwijk 12, Mona Ståhle 13, A David Burden 14, Catherine H Smith 15, Michael J Cork 16, Xavier Estivill 17, Anne M Bowcock 18, Gerald G Krueger 19, Wolfgang Weger 20, Jane Worthington 21, Rachid Tazi-Ahnini 16, Frank O Nestle 2, Adrian Hayday 22, Per Hoffmann 23,24, Juliane Winkelmann 25,26,27, Cisca Wijmenga 28, Cordelia Langford 29, Sarah Edkins 29, Robert Andrews 29, Hannah Blackburn 29, Amy Strange 30, Gavin Band 30, Richard D Pearson 30, Damjan Vukcevic 30, Chris CA Spencer 30, Panos Deloukas 29, Ulrich Mrowietz 31, Stefan Schreiber 5,32,33, Stephan Weidinger 31, Sulev Koks 34, Külli Kingo 35, Tonu Esko 36, Andres Metspalu 36, Henry W Lim 37, John J Voorhees 6, Michael Weichenthal 31, H Erich Wichmann 38,39,40, Vinod Chandran 41, Cheryl F Rosen 42, Proton Rahman 43, Dafna D Gladman 41, Christopher EM Griffiths 44, Andre Reis 45, Juha Kere 46,47,48; Collaborative Association Study of Psoriasis49; Genetic Analysis of Psoriasis Consortium49; Psoriasis Association Genetics Extension49; Wellcome Trust Case Control Consortium 249, Rajan P Nair 6, Andre Franke 5, Jonathan NWN Barker 2,15, Goncalo R Abecasis 1,, James T Elder 6,50,, Richard C Trembath 2,51,
PMCID: PMC3510312  NIHMSID: NIHMS416128  PMID: 23143594

Summary

To gain further insight into the genetic architecture of psoriasis, we conducted a meta-analysis of three genome-wide association studies (GWAS) and two independent datasets genotyped on the Immunochip, involving 10,588 cases and 22,806 controls in total. We identified 15 new disease susceptibility regions, increasing the number of psoriasis-associated loci to 36 for Caucasians. Conditional analyses identified five independent signals within previously known loci. The newly identified shared disease regions encompassed a number of genes whose products regulate T-cell function (e.g. RUNX3, TAGAP and STAT3). The new psoriasis-specific regions were notable for candidate genes whose products are involved in innate host defense, encoding proteins with roles in interferon-mediated antiviral responses (DDX58), macrophage activation (ZC3H12C), and NF-κB signaling (CARD14 and CARM1). These results portend a better understanding of shared and distinctive genetic determinants of immune-mediated inflammatory disorders and emphasize the importance of the skin in innate and acquired host defense.


Psoriasis is a chronic, potentially disfiguring, immune-mediated inflammatory disease of the skin with a prevalence of 0.2 to 2%, depending on the population of origin. About one-quarter of psoriatics develop a painful and debilitating arthritis, and there is increasing awareness of co-morbidities, including metabolic syndrome and cardiovascular disease1,2. Current evidence suggests that a dysregulated cutaneous immune response characterized by tumor necrosis factor-α (TNF) dependence and exaggerated Th1 and Th17 activation occurs in genetically susceptible individuals1,2. Recent large-scale association studies have identified 26 loci that are associated with psoriasis310, 21 of which show association in Caucasians36,10. Several of these signals overlap with other autoimmune diseases (e.g. Crohn’s disease, ankylosing spondylitis, and celiac disease), particularly those near genes involved in Th17 differentiation and IL-17 responsiveness (e.g. IL23R, IL12B, IL23A, TRAF3IP2)11. To accelerate our understanding of the genetic architecture of this disease, we helped design a custom single-nucleotide polymorphism (SNP) array (the “Immunochip”). The aims of the Immunochip are to fine-map genome-wide significant (i.e. P<5×10−8) susceptibility loci and to explore replication of thousands of SNPs representing additional promising signals12,13. In this study, we use Immunochip data to identify new genetic determinants of psoriasis, and to relate them to other autoimmune disorders.

We combined three existing GWAS datasets (hereafter referred to as Kiel3, CASP4 and WTCCC25) with two independent European-descent case-control datasets genotyped on the Immunochip: the Psoriasis Association Genetics Extension (PAGE: 3,580 cases and 5,902 controls) and the Genetic Analysis of Psoriasis Consortium (GAPC: 2,997 cases and 9,183 controls) (datasets are described in Supplementary Tables 1 and 2). After quality control, the combined dataset consisted of 10,588 patients with psoriasis and 22,806 healthy controls. For each GWAS, we increased the SNP density through imputation by using European haplotype sequences generated by the 1000 Genomes Project as templates (20100804 release). Overall, our analysis includes 111,236 SNPs that were genotyped in both Immunochip datasets and also had good imputation quality in at least two of the three GWAS (see Online Methods).

Meta-analysis of all five datasets yielded genome-wide significance for 19 of the 21 known psoriasis loci (Supplementary Fig 1, Table 1, Supplementary Table 3). We found nominal evidence for the remaining two loci in the combined analysis (ZMIZ1 and PRDX5, each with P < 3×10−6) as well as nominal evidence for all loci in separate analyses including only GWAS (all with P < 5×10−3) or Immunochip data (all with P < 4×10−4). In addition, we identified 15 new risk loci at P < 5×10−8 (Supplementary Fig 1, Table 1, and Supplementary Table 3). Nine of the new signals were submitted, during design of the Immunochip, as genome-wide significant Immunochip loci by at least one other disease consortium (see “Disease Overlap” column in Supplementary Table 4), though we also submitted three of these (rs11121129, rs10865331, and rs9504361) based on a preliminary meta-analysis of our GWAS datasets. Notably, of the remaining six signals, four were submitted as genome-wide significant loci for psoriasis (SNPs rs11795343, rs4561177, rs11652075, and rs545979). The strongest new association was observed for rs892085 at 19p13.2, near the ILF3 and CARM1 genes (combined Pvalue (Pcomb) = 3.0 × 10−17; OR = 1.17). Despite its proximity (< 500kb) to TYK2, conditional analysis demonstrated that this is an independent signal (Supplementary Table 5). Other associated loci included 1p36.11 near RUNX3; 6p25.3 near EXOC2 and IRF4; 9p21.1 near DDX58; 11q22.3 near ZC3H12C, 11q24.3 in the ETS1 gene and 17q21.2 near STAT3, STAT5A and STAT5B. Box 1 summarizes the functional characteristics of notable genes from the newly identified loci, and the regional association plots are shown in Supplementary Fig 2.

Table 1.

Meta-analysis results for psoriasis loci. For known loci, the most significant SNP within 500kb (3Mb for MHC region) of the previously published SNP is shown. rs34536443 was the most strongly associated SNP in the TYK2 region, but found to be independent of the previously published SNP (rs12720356). ‘GWAS P value’: P value from the meta-analysis of the 3 GWAS datasets. ‘Immunochip P value’: the result of the meta-analysis of the two Immunochip datasets. ‘Combined P-value’: the P-value from the meta-analysis including all 5 datasets, RAF: Risk allele frequency, ‘Notable genes’: genes most likely to have an effect on the development of psoriasis.

SNP Chr. Position (bp) GWAS P-value (meta) Immunochip p-value (meta) Combined P-value Risk/ Non-risk allele RAF (Case) RAF (Ctrls) ORa (meta) Notable genes No. of genes +/− 500kb
Known Loci
rs7552167 1 24,518,643 2.3×10−5 8.4×10−8 8.5×10−12 G/A 0.878 0.858 1.21 IL28RA 26
rs9988642 1 67,726,104 2.5×10−13 3.5×10−15 1.1×10−26 T/C 0.952 0.929 1.52 IL23R 17
rs6677595 1 152,590,187 8.1×10−15 2.7×10−20 2.1×10−33 T/C 0.689 0.640 1.26 LCE3B, LCE3D 43
rs62149416 2 61,083,506 3.4×10−10 3.2×10−9 1.8×10−17 T/C 0.671 0.635 1.17 FLJ16341, REL 9
rs17716942 2 163,260,691 4.1×10−9 1.0×10−10 3.3×10−18 T/C 0.891 0.863 1.27 KCNH7, IFIH1 7
rs27432 5 96,119,273 4.4×10−8 7.5×10−14 1.9×10−20 A/G 0.309 0.274 1.20 ERAP1 7
rs1295685 5 131,996,445 8.5×10−6 6.7×10−6 3.4×10−10 G/A 0.807 0.798 1.18 IL13, IL4 21
rs2233278 5 150,467,189 4.9×10−17 5.2×10−27 2.2×10−42 C/G 0.090 0.058 1.59 TNIP1 17
rs12188300 5 158,829,527 7.5×10−23 3.3×10−32 3.2×10−53 T/A 0.132 0.095 1.58 IL12B 5
rs4406273 6 31,266,090 5.3×10−300 3.6×10−427 4.5×10−723 A/G 0.259 0.092 4.32 HLA-B, HLA-C 56
rs33980500 6 111,913,262 4.3×10−20 7.6×10−27 4.2×10−45 T/C 0.108 0.074 1.52 TRAF3IP2 8
rs582757 6 138,197,824 2.0×10−14 3.7×10−13 2.2×10−25 C/T 0.315 0.273 1.23 TNFAIP3 5
rs1250546 10 81,032,532 5.1×10−4 3.2×10−4 6.8×10−7 A/G 0.605 0.579 1.10 ZMIZ1 9
rs645078 11 64,135,298 4.7×10−3 1.5×10−4 2.2×10−6 A/C 0.626 0.609 1.09 RPS6KA4, PRDX5 36
rs2066819 12 56,750,204 7.5×10−12 8.9×10−8 5.4×10−17 C/T 0.948 0.934 1.39 STAT2, IL23A 40
rs8016947 14 35,832,666 1.4×10−9 1.6×10−9 2.5×10−17 G/T 0.600 0.564 1.16 NFKBIA 11
rs12445568 16 31,004,812 1.2×10−6 1.8×10−11 1.2×10−16 C/T 0.403 0.368 1.16 PRSS53, FBXL19 46
rs28998802 17 26,124,908 3.6×10−6 1.7×10−11 3.3×10−16 A/G 0.170 0.145 1.22 NOS2 9
rs34536443 19 10,463,118 5.1×10−10 2.6×10−22 9.1×10−31 G/C 0.974 0.953 1.88 TYK2 42
rs1056198 20 48,556,229 6.2×10−9 1.6×10−7 1.5×10−14 C/T 0.600 0.573 1.16 RNF114 11
rs4821124 22 21,979,289 5.4×10−5 1.2×10−4 3.8×10−8 C/T 0.208 0.189 1.13 UBE2L3 16
Newly Identified Loci
rs11121129 1 8,268,095 7.3×10−5 4.6×10−5 1.7×10−8 A/G 0.308 0.287 1.13 SLC45A1, TNFRSF9 15
rs7536201 1 25,293,084 7.8×10−5 6.4×10−9 2.3×10−12 C/T 0.528 0.494 1.13 RUNX3 18
rs10865331 2 62,551,472 4.5×10−4 2.6×10−7 4.7×10−10 A/G 0.404 0.374 1.12 B3GNT2 6
rs9504361 6 577,820 5.1×10−7 4.2×10−6 2.1×10−11 A/G 0.574 0.546 1.12 EXOC2, IRF4 5
rs2451258 6 159,506,600 4.4×10−4 2.0×10−5 3.4×10−8 C/T 0.362 0.348 1.12 TAGAP 8
rs2700987 7 37,386,237 3.3×10−7 4.6×10−4 4.3×10−9 A/C 0.591 0.564 1.11 ELMO1 3
rs11795343 9 32,523,737 2.8×10−7 2.1×10−5 8.4×10−11 T/C 0.628 0.597 1.11 DDX58 7
rs10979182 9 110,817,020 2.8×10−5 1.2×10−4 2.3×10−8 A/G 0.617 0.591 1.12 KLF4 0
rs4561177 11 109,962,432 1.1×10−4 1.4×10−9 7.7×10−13 A/G 0.617 0.581 1.14 ZC3H12C 4
rs3802826 11 128,406,438 1.1×10−3 2.0×10−7 9.5×10−10 A/G 0.505 0.484 1.12 ETS1 7
rs367569 16 11,365,500 2.6×10−4 4.6×10−5 4.9×10−8 C/T 0.729 0.709 1.13 PRM3, SOCS1 14
rs963986 17 40,561,579 9.9×10−5 1.2×10−5 5.3×10−9 C/G 0.169 0.154 1.15 PTRF, STAT3, STAT5A/B 42
rs11652075 17 78,178,893 1.3×10−3 7.0×10−6 3.4×10−8 C/T 0.530 0.502 1.11 CARD14 16
rs545979 18 51,819,750 1.4×10−6 2.4×10−5 3.5×10−10 T/C 0.317 0.291 1.12 POL1, STARD6, MBD2 6
rs892085 19 10,818,092 1.2×10−7 4.5×10−11 3.0×10−17 A/G 0.593 0.558 1.17 ILF3,CARM1 37
a

The overall OR was calculated using the effective sample size-weighted approach.

Box 1. The function of notable genes in the regions of newly identified associations.

RERE, SLC45A1, ERRFI1, TNFRSF9 (1p36.23)

This signal falls between the RERE, SLC45A1, ERRFI1, and TNFRSF9 genes. RERE encodes an arginine-glutamic acid dipeptide repeat-containing protein that controls retinoic acid signalling38. ERRFI1 encodes a feedback inhibitor of the EGF receptor39. SLC45A1 encodes a solute carrier protein that mediates the uptake of glucose40. The TNFRSF9 gene encodes a co-stimulatory molecule that has a role in generation of memory CD8+ T-cells.

RUNX3 (1p36.11)

RUNX3 is a member of the Runt domain-containing family of transcription factors and has an essential role in T-cell biology, particularly in the generation of CD8+ cells. RUNX3 also has a role in promoting Th1 differentiation through binding with T-bet41.

B3GNT2 (2p15)

B3GNT2 is a member of the beta-1,3-N-acetylglucosaminyl transferase family. It catalyzes the initiation and elongation of poly-N-acetyllactosamine chains42. Deficiency has shown to results in hyperactivation of lymphocytes43.

EXOC2, IRF4 (6p25.3)

EXOC2 encodes a component of the multi-protein complex which mediates the docking of exocytic vesicles to the plasma membrane44. IRF4 encodes a transcription factor that regulates IL17A promoter activity and controls RORyt-dependent Th17 colitis in vivo45,46. IRF4 also plays a role in stabilization of the Th17 phenotype through IL-2147 and may regulate CD4/CD8 differentiation through regulation of RUNX3 expression48.

TAGAP (6q25.3)

This gene is a Rho-GTPase activating protein that is involved in T-cell activation49.

ELMO1 (7p14.2-7p14.1)

ELMO1 is a member of the engulfment and cell motility protein family, which binds to DOCK2, and is essential for TLR7- and TLR9-mediated IFNinduction by plasmacytoid dendritic cells50 and plasmacytoid dendritic cell migration.51 DOCK2 also has a role in antigen-uptake and presentation, and lymphocyte trafficking51.

DDX58 (9p21.1)

DDX58 encodes the RIG-I innate antiviral receptor, which recognizes cytosolic double-stranded RNA.52 It is induced by IFN-γ53 and regulates type I and type II IFN production54.

KLF4 (9p31.2)

KLF4 is a Kruppel-like transcription factor, which is required for the establishment of skin barrier function55 and regulates key signaling pathways related to macrophage activation56. KLF4 also binds to the promoter of IL17A and positively regulates its expression.

ZC3H12C (11q22.3)

Zinc-finger protein regulating macrophage activation57.

ETS1 (11q24.3)

Transcription factor activated downstream of the Ras-MAPK pathway, involved in homeostasis of squamous epithelia58. Involved in CD8 lineage differentiation and acts in part by promoting RUNX3 expression59. Negative regulator of Th17 differentiation60

SOCS1 (16p13.13)

SOCS1 is a member of the suppressor of cytokine signalling family of proteins and inhibits signalling events downstream of IFN-γ61. It regulates Th17 differentiation by maintaining STAT3 transcriptional activity62 and interacts with TYK2 in cytokine signalling63.

STAT3 , STAT5A/5B (17q21.2)

STAT3 and STAT5A/5B are members of the STAT family of transcription activators. STAT3 participates in signalling downstream of multiple cytokines implicated in psoriasis such as IL-6, IL-10, IL-20, IL-22 and IL-23 and may have a role in mediating the innate immune response in psoriatic epidermis64. STAT3 is required for the differentiation of Th17 cells65. STAT5A/5B participate in signalling downstream of the IL-2 family of cytokines, including IL-2, IL-7, IL-15 and IL-21. Both proteins contribute to the development of Treg cells and inhibit the differentiation of Th17 cells66.

CARD14 (17q25.3)

Member of a family of Caspase Recruitment Domain containing scaffold proteins, known as CARD- and membrane-associated guanylate kinase-like domain-containing protein (CARMA). CARD14/CARMA2 is primarily expressed in epithelial tissues and mediates recruitment and activation of the NF-κB pathway67.

MBD2,POLI ,STARD6 (18q21.2)

MBD2 is a transcriptional repressor that binds to methylated DNA and has a role in the generation of memory CD8+ T-cells68. POLI is an error-prone DNA polymerase, which contributes to the hyper-mutation of immunoglobulin genes69. Sterol transport is mediated by vesicles or by soluble protein carriers, such as steroidogenic acute regulatory protein (STAR; MIM 600617). STAR is homologous to a family of proteins containing a 200- to 210-amino acid STAR-related lipid transfer (START) domain, including STARD6.

ILF3, CARM1 (19p13.2)

ILF3 encodes a double-stranded RNA (dsRNA) binding protein that complexes with other proteins, dsRNAs, small noncoding RNAs, and mRNAs to regulate gene expression and stabilize mRNAs. It is a subunit of the nuclear factor of activated T-cells (NFAT); a transcription factor required for T-cell expression of IL-2. CARM1 is a transcriptional coactivator of NF-κB and functions as a promoter-specific regulatory of NF-κB recruitment to chromatin.

To identify independent secondary signals, we performed conditional analysis using as covariates the strongest signals from the 34 loci achieving genome-wide significance in this study. We identified secondary signals in five loci: 2q24.2, 5q15, 5q33.3, 6p21.33, and 19q13.2 (Supplementary Figs. 3 and 4, Supplementary Tables 6 and 7). The strongest signal from the conditional analysis maps to the MHC region near the MICA gene (rs13437088: P=3.1 × 10−40; OR = 1.32), in agreement with a previous conditional analysis14. The 5q15 conditional signal is in the ERAP2 gene (rs2910686: P = 2.0 × 10−8), which did not show any evidence of association in the unconditional analysis (P = 0.46). Further investigation revealed that the risk-increasing alleles at ERAP1 and the risk-decreasing alleles at ERAP2 preferentially appear on the same haplotype, and the signal near ERAP2 is thus masked by ERAP1 prior to conditional analysis (Supplementary Note). The strongest conditional signal in the 19q13.2 region was rs12720356 in the TYK2 gene (OR=1.25, MAFcontrols=0.09, P = 3.2 × 10−10). The association of this SNP with psoriasis has been previously reported5 and is independent of the strongest TYK2 signal identified by our meta-analysis (rs34536443, OR=1.88, MAFcases=0.03, P = 1.5 × 10−39). As rs34536443 was a low-frequency imputed SNP and manifested the highest effect size outside of the MHC, we directly genotyped this SNP in 3,390 independent Michigan samples (1,844 cases and 1,546 controls), robustly replicating the association (OR = 2.80, MAFcases= 0.02, P = 7.8 × 10−14) and experimentally confirming the validity of our imputation procedures.

We next tested for statistical interaction among the top SNPs in the 34 significant loci (Supplementary Note; Supplementary Table 8). We identified two significant pairwise interactions after correction for multiple testing (P < 5 × 10−5): HLA-C (rs4406273)-LCE (rs6677595) and HLA-C (rs4406273)-ERAP1 (rs27432). These interactions confirm results of previous studies5,15,16.

In order to identify potential causal alleles in coding sequence, we looked for missense variants in tight LD (r2>0.9 in 1000 Genomes Project European samples) with the lead SNPs from each of the 34 identified loci (Table 1 and Supplementary Table 6). We found 10 potentially causal SNPs (Table 2), nine of which were included in our meta-analysis. For the known loci near TRAF3IP2 and TYK2, damaging non-synonymous substitutions were themselves the index SNPs in our initial and conditional analyses. Among the newly identified loci, the index SNP from CARD14, a gene that harbors Mendelian variants predisposing to psoriasis17, was also a common and damaging variant as has been described elsewhere18. For the remaining loci, we could account for essentially all index SNP signals by conditioning on nearby missense SNPs, consistent with the possibility that they are causal. Notable non-synonymous variants include the protective c.R381Q polymorphism in IL23R19; a SNP in the PRSS53 gene20, which is also the most highly over-expressed gene in psoriatic skin in this locus6; and a variant in YDJC that also increases risk for celiac disease21, rheumatoid arthritis22 and Crohn’s disease23.

Table 2.

SNPs that are missense mutations from the 1000 Genome Project and that are in LD (r2>=0.9) with primary signals from the known and newly identified loci that achieve genomewide significance in the meta-analysis, or with secondary signals from the conditional analysis (“Index SNP”). The “Index SNP” columns show the information of SNPs with the most significant P-value in our analysis, and the “Potential causal SNP” columns show the information for the SNPs that have high LD with our strongest signal. The “Combined p-value” column shows the meta-analysis P-value for the index SNP, potential causal SNP, and the P-values for the index SNPs while conditioning on the potential causal SNPs, respectively. Note the potential causal SNP rs7199949 is not present in our meta-analysis study therefore its P-value is not shown.

Index SNP
Potential Causal SNP
Combined P-value
Markera RAF Annotation Markerc RAF Gene with variant Amino acid substitution (Damaging effectd) r2 Index SNP Potential causal SNP Index SNP (conditioning on causal SNP)
rs9988642 0.93 454bp downstream IL23R rs11209026 0.94 IL23R R381Q (P) 0.91 1.1×10−26 1.5×10−26 0.13
rs27432 0.29 Intron ERAP1 rs27044 0.29 ERAP1 Q730E 1 1.9×10−20 2.3×10−20 0.14
rs1295685 0.77 3′ UTR IL13 rs20541 0.77 IL13 R144Q 0.97 3.4×10−10 3.5×10−10 0.78
rs33980500 0.09 Missense Self 0.09 TRAF3IP2 D19N (S/P) 1 4.2×10−45 4.2×10−45 NA
rs2066819 0.93 Intron STAT2 rs2066807 0.93 STAT2 M594I 0.9 5.4×10−17 5.1×10−16 0.036
rs12445568 0.36 Intron STX1B rs7199949 0.37 PRSS53 P406A 0.9 1.2×10−16 NA NA
rs11652075 0.51 Missense Self 0.51 CARD14 R820W (S) 1 3.4×10−8 3.4×10−8 NA
rs34536443 0.97 Missense Self 0.97 TYK2 P1104A (S/P) 1 1.5×10−39 1.5×10−39 NA
rs12720356b 0.9 Missense Self 0.9 TYK2 I684S (S/P) 1 3.2×10−10 3.2×10−10 NA
rs4821124 0.19 966bp downstream UBE2L3 rs2298428 0.18 YDJC A263T 0.96 3.8×10−8 6.2×10−8 0.48
a

SNPs with the most significant p-value in our analysis.

b

The meta-analysis p-value from the conditional analysis is shown.

c

SNPs that are missense mutations and have high LD with our strongest signal.

d

High confidence damaging effect predicted by SIFT (S) or Polyphen (P). RAF: Risk Allele Frequency. For the potential causal SNP rs7199949, the P value is ‘NA’ as the SNP was not included on the Immunochip.

Utilizing the results of a large-scale study of gene expression in psoriatic vs. normal skin24 , we found 14 up-regulated genes (IL12RB2, LCE3D, REL, PUS10, CDSN, PRSS53, PRSS8, NOS2, DDX58, ZC3H12C, SOCS1, STAT3, CARD14, IFIH1) and 4 down-regulated genes (MICA, RNF114, PTRF, POLI) in the 34 associated regions (FDR<0.05 and fold-change>1.5 or <0.67; Supplementary Table 9). The number of differentially expressed genes in psoriasis susceptibility loci was not greater than expected by chance (P=0.39). None of the 34 top SNPs met the Bonferroni corrected (P < 1×10−7) threshold as expression quantitative trait loci (eQTL) in skin tissue, as assessed by microarray analysis of mRNA levels25. However, rs2910686, one of the five SNPs identified by conditional analysis, was a cis-eQTL for ERAP2 in both normal and psoriatic skin (see Supplementary Note for details). Genetic control of ERAP2 expression has been noted previously26,27 and has been suggested as a determinant of balancing selection at this locus28.

This study increases the number of psoriasis-associated regions in European ancestry samples to 36, with conditional analysis increasing the number of independent signals to 41. The 39 independent signals with P < 5×10−8 in the current study collectively account for 14.3% of the total variance in psoriasis risk, or approximately 22% of its estimated heritability29 (see Supplementary Table 10 for details), indicating that further genetic studies, including fine mapping studies and searches for uncommon susceptibility variants are in order.

Sharing of susceptibility loci between autoimmune diseases has been demonstrated previously11 and we find similar patterns in this study. Notably, ten of the psoriasis susceptibility loci reported here overlap with Crohn’s disease and ten others with celiac disease, two diseases that are enriched in individuals with psoriasis30,31 (Supplementary Table 4; illustrated in Supplementary Fig. 5). We caution that the statistical significance of these overlaps is hard to assess, given the ongoing process of gene discovery for many autoimmune disorders and biases in the list of SNPs evaluated for association in this experiment.

As the primary interface with the external environment, the skin provides a critical first line of host defense to microbial pathogens. Consistent with this function, it possesses a diverse and well-conserved set of innate immune mechanisms32,33, which emerged long before the development of adaptive immunity34. In this context, we found it interesting that five of the six newly identified loci that are thus far uniquely associated with psoriasis are involved in innate immune responses (DDX58, KLF4, ZC3H12C, CARD14 and CARM1, Supplementary Table 4 and Box 1). Among all confirmed psoriasis susceptibility loci, 11 out of 14 psoriasis specific loci (the five listed above along with IL28RA, LCE3D, NOS2, FBXL19, NFKBIA and RNF114) encode plausible regulators of innate host defense1,2,35. Conversely, only 6 out of 20 loci shared with other autoimmune diseases contain genes (REL, IFIH1, TNIP1, TNFAIP3, IRF4 and ELMO1) that contribute to innate immunity. These provisional comparisons further illustrate the insights that can be gained by developing and comparing complete and well-annotated sets of risk loci for autoimmune disorders.

The known and newly identified psoriasis susceptibility loci implicated by this study encode several proteins engaged in the TNF, IL-23, and IL17 signaling pathways targeted by highly effective biologic therapies36. Interestingly, our strongest non-MHC signal directly implicates TYK2, a druggable target that contributes to several autoimmune diseases. Agents targeting the closely related JAK kinases are showing encouraging results in clinical trials37. Our findings will help prioritize and interpret the results of sequencing and gene expression studies. Further genomic studies will allow us to identify the underlying causal variants within psoriasis susceptibility loci and lead to increased understanding of pathogenetic mechanisms and new therapeutic targets.

Online Methods

Sample Collections

The samples used in the 3 GWAS data sets (Kiel, CASP and WTCCC2) were previously described35. Samples of the Psoriasis/Arthritis Genetics Extension (PAGE) and the Genetic Analysis of Psoriasis Consortium (GAPC) datasets (Supplementary Table 1 and 2) were collected from subjects of European Caucasian descent at the participating institutions after obtaining informed consent in adherence with the Declaration of Helsinki Principles. DNA was isolated from blood or EBV-immortalized lymphoblastoid cell lines using standard methods.

The collections used in the GAPC and PAGE ImmunoChip studies are described in Supplementary Table 2.

The samples from GAPC substantially overlapped with those described as replication datasets in Strange et al. 20105. All cases had been diagnosed as having psoriasis vulgaris. The GAPC cases and the Irish and Spanish controls were genotyped at the Wellcome Trust Sanger Institute (WTSI) and all samples were provided by the relevant groups given in Supplementary Table 2 and listed in the GAP consortium members list (Supplementary Note 2). The UK controls were the WTCCC common controls that did not overlap with samples included in the original GWA studies (the dataset consisted of 6,740 1958 British Birth cohort and 2,900 UK Blood Service samples genotyped at the WTSI and the University of Virginia). The German controls were obtained from the PopGen biobank and genotyped at the Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel. The Finland control data were from the DILGOM (Dietary, Lifestyle, and Genetic determinants of Obesity and Metabolic syndrome) collection70. The Irish controls were provided by the Irish Blood Transfusion Service / TCD Biobank and the Irish cases collected with the aid of the Dublin Centre for Clinical Research. We did not include specific controls from Austria or Sweden, but PCA analysis suggested that the cases from these cohorts were well matched to the controls from the Netherlands and Germany.

For the PAGE Immunochip study, samples also substantially overlapped with previously published replication datasets. The German cases (described as a replication dataset in Ellinghaus et al. 20103), all samples from the United States and Canada, as well as 439 Estonian cases from the University of Tartu were genotyped at the Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel. The respective samples were provided by the groups given in Supplementary Table 2 and listed in the PAGE members list (Supplementary Note). The German controls were obtained from a population-based sample from the general population living in the region of Augsburg, Southern Germany (Collaborative Health Research in the Region of Augsburg; KORA S4/F471), which was genotyped at the Helmholtz Center in Munich, and from the population-based epidemiological Heinz-Nixdorf Recall study (HNR), which was genotyped at the Life and Brain Center at the University Clinic in Bonn. The remaining Estonian samples were obtained from and genotyped at the Estonian Genome Center University of Tartu (EGCUT).

Genotyping panel and SNPs

The Immunochip is a custom Illumina Infinium high-density array consisting of 196,524 variants (after Illumina quality control) compiled largely from variants identified in previous GWAS of 12 different immune-mediated inflammatory diseases, including psoriasis13. The main aims of the Immunochip were deeper replication and fine-mapping of genome-wide significant loci, as well as increasing power to promote promising but less significant SNPs to genome-wide significance. For fine mapping, SNPs within 0.2 cM on either side of the GWAS top SNPs for 186 loci were selected from 1000 Genomes Project72 low coverage pilot CEU sequencing data as well as additional variants identified by resequencing from groups involved in the chip design. For promotion of promising signals and those not quite reaching genome-wide significance, each disease-focused group was allowed to submit approximately 3,000 additional SNPs. We submitted 17 of the 19 confirmed genome-wide significant psoriasis regions (Table 1) for fine mapping based on a preliminary meta-analysis of our data, while one of the confirmed signals (IL28RA) and nine of the new psoriasis signals (indicated in the “disease overlap” column of Supplementary Table 5) were submitted for fine mapping by other disease groups (though we also submitted three of them as part of our additional SNP allocation SNPs: rs11121129, rs10865331, and rs9504361). Six additional signals were detected based on individual groups additional SNP allocation; four of these (rs11795343, rs4561177, rs11652075, and rs545979) were submitted by our group. All Immunochip samples were genotyped as described in Illumina’s protocols.

Genotype calling

For the PAGE dataset genotype calling was performed using Illumina’s GenomeStudio Data Analysis software and the custom-generated cluster file of Trynka et al. (based on an initial clustering of 2000 UK samples with the GenTrain2.0 algorithm and subsequent manual readjustment and quality control)13. The genotype calling for the GAPC dataset was performed using GenoSNP73 from allele intensities, except for the German, Italian, Dutch and Finnish controls, which were called using the same method described for the PAGE dataset.

Imputation

To increase the number of overlapping SNPs between datasets, we performed imputation on the 3 GWAS datasets using minimac74 (Kiel and CASP) and IMPUTE275,76 (WTCCC2) based on CEU reference haplotypes from the 1000 Genomes Project72; December 2010 version of the 10/08/04 sequence and alignment release containing 629 individuals of European descent). SNPs with low imputation quality (r2 ≤0.3 for minimac, info score < 0.5 for IMPUTE2) were removed. For all 3 datasets, cases and controls were imputed together.

Sample and genotype quality control

For the Immunochip datasets, we first excluded SNPs with a call rate below 95% or with a Hardy-Weinberg p-value < 1 × 10−6. Samples with less than 98% SNP call rates were then excluded. Because the Immunochip includes a large proportion of fine-mapping SNPs that are associated with autoimmune disease, we used a set of independent SNPs which have p-values > 0.5 from the meta-analysis of the 3 GWAS datasets as a quality control tool for each individual Immunochip dataset. Using the HapMap 3 samples as reference77, we performed principal component (PC) analysis to identify and remove samples with non-European ancestry. We also removed samples with extreme inbreeding coefficients or heterozygosity values computed by PLINK78.

To assess possible stratification in the datasets, principal components analysis was also performed in each of the Immunochip datasets separately (excluding HapMap). There was no evidence of stratification between the cases and controls of each sample group. However, as expected, the top principal components (PCs) do separate the samples well by country of origin. The use of the top 10 eigenvectors as covariates in the analysis did not completely correct for this stratification and so a linear mixed model method (Efficient Mixed-Model Association eXpedited (EMMAX)) was used for the association analysis instead. These methods have been shown to outperform PCs at correcting for this type of population stratification and cryptic relatedness79, which is becoming more common as sample sizes get larger and studies comprise of more collaborative efforts.

To identify duplicate pairs or highly related individuals among datasets, we used a panel of 873 independent SNPs that were genotyped in both the GWAS and Immunochip samples, and performed pairwise comparisons using the ‘genome’ function in PLINK78 , with the criterion Pi-HAT≥ 0.5. We identified 1,142 (885 from GAPC and 257 from PAGE) related sample pairs (mostly duplicates) and removed one sample from each pair. We also removed 4,828 controls from the UK common ImmunoChip controls owing to duplication in the WTCCC2 GWAS sample. For GWAS samples that were duplicated in the Immunochip datasets (the majority of duplicates), we removed the samples from the Immunochip datasets to keep the previously published datasets intact.

The GWAS datasets underwent quality control as previously described and were analysed for association using the top PCs from the previous analyses, as covariates35.

We visually checked the signal intensity cluster plots for all SNPs meeting genome-wide significance to confirm high quality genotype calling.

Genomic Control

Genomic control inflation factors for the five datasets were 1.09 (Kiel), 1.06 (CASP), 1.04 (WTCCC2), 0.99 (PAGE), and 0.96 (GAPC), indicating that population structure and cryptic relatedness were adequately controlled for in these datasets. Because the Immunochip was designed for deep replication and fine mapping of loci associated with autoimmune diseases12, using all independent SNPs from the chip would not give an accurate estimate of the genomic control80GC) value. Therefore, we selected common (MAF > 0.05) SNPs from the Immunochip that had p-values > 0.5 based on a meta-analysis combining the CASP, Kiel, and the WTCCC2 GWAS, and then performed LD-pruning to identify an independent SNP set to compute λGC for the association results from the Immunochip datasets. Due to the SNP selection bias, the genomic control of the final meta-analysis was computed using a set of independent SNPs associated with “reading and writing ability” (personal communication, J.C. Barrett). We further removed SNPs that were within ±500 kb of previously detected psoriasis loci (±3 Mb was used for the MHC region), and the remaining 1,426 SNPs yielded a λGC value of 1.11 for the meta-analysis overall. By using the λ100081, the genomic control inflation factor for an equivalent study of 1000 cases and 1000 controls, the rescaled λ equals 1.01.

Supplementary Material

1

Acknowledgments

Major support for this study was provided by the National Institutes of Health, the Wellcome Trust, and the German Research Foundation. We acknowledge use of the British 1958 Birth Cohort DNA collection, funded by the Medical Research Council (G0000934) and the Wellcome Trust (068545/Z/02), and of the UK National Blood Service controls funded by the Wellcome Trust. We acknowledge the Collaborative Association Study of Psoriasis (CASP) for the contribution of GWAS data, as well as the provision of control DNA samples by the Cooperative Research in the Region of Augsburg (KORA) and Heinz Nixdorf Recall (Risk Factors, Evaluation of Coronary Calcification, and Lifestyle) study (HNR), and genotyping data generated by the Dietary, Lifestyle, and Genetic determinants of Obesity and Metabolic syndrome (DILGOM) consortium. We thank the Barbara and Neal Henschel Charitable Foundation for their support of the National Psoriasis Victor Henschel BioBank. We thank Jeffrey C Barrett for contribution to the design of the Immunochip and helpful analytical discussion, as well as Emma Gray, Suzannah Bumpstead, Douglas Simpkin and staff of the Wellcome Trust Sanger Institute Sample Management and Genotyping teams for their genotyping and analytical contributions. We acknowledge the Genetic repository in Ireland for Psoriasis and Psoriatic Arthritis (GRIPPsA), the Irish blood transfusion service / TCD Biobank and the Dublin Centre for Clinical Research (funded by HRB and the Wellcome Trust). Detailed consortium contributorship lists and relevant funding support are detailed in the Supplementary Note.

Consortia

Wellcome Trust Case Control Consortium 2

Peter Donnelly30,52, Leena Peltonen29, Jenefer M Blackwell53,54, Elvira Bramon55,56, Matthew A Brown57, Juan P Casas58, Aiden Corvin59, Nicholas Craddock60, Audrey Duncanson61, Janusz Jankowski62, Hugh S Markus63, Christopher G Mathew2, Mark I McCarthy64, Colin NA Palmer65, Robert Plomin66, Anna Rautanen30, Stephen J Sawcer67, Nilesh Samani68, Ananth C Viswanathan69,70, Nicholas W Wood71, Céline Bellenguez30, Colin Freeman30, Garrett Hellenthal30, Eleni Giannoulatou30, Matti Pirinen30, Zhan Su30, Sarah E Hunt29, Rhian Gwilliam29, Suzannah J Bumpstead29, Serge Dronov29, Matthew Gillman29, Emma Gray29, Naomi Hammond29, Alagurevathi Jayakumar29, Owen T McCann29, Jennifer Liddle29, Marc L Perez29, Simon C Potter29, Radhi Ravindrarajah29, Michelle Ricketts29, Matthew Waller29, Paul Weston29, Sara Widaa29, Pamela Whittaker29.

Genetic Analysis of Psoriasis Consortium (GAPC)

Alexandros Onoufriadis2, Michael E Weale2, Angelika Hofer20, Wolfgang Salmhofer20, Peter Wolf20, Kati Kainu72, Ulpu Saarialho-Kere72, Sari Suomela72, Petra Badorf45, Ulrike Hüffmeier45, Werner Kurrat73, Wolfgang Küster74, Jesus Lascorz75, Rotraut Mössner76, Funda Schürmeier-Horst77, Markward Ständer78, Heiko Traupe77, Judith G M Bergboer12, Martin den Heijer79,80, Peter C. van de Kerkhof12, Patrick L J M Zeeuwen12, Louise Barnes7,8, Linda E Campbell81, Catriona Cusack82, Ciara Coleman7,8, Judith Conroy7, 8, Sean Ennis7, 8, Oliver Fitzgerald83, Phil Gallagher83, Alan D Irvine84, Brian Kirby83, Trevor Markham82, WH Irwin McLean81, Joe McPartlin7, 8, Sarah F Rogers83, Anthony W Ryan7, 8, Agnieszka Zawirska83, Emiliano Giardina9, Tiziana Lepre9, Carlo Perricone9, Gemma Martín-Ezquerra85, Ramon M Pujol85, Eva Riveira-Munoz17, Annica Inerot86, Åsa T Naluai11, Lotus Mallbris13, Katarina Wolk13, Joyce Leman14, Anne Barton21, Richard B Warren44, Helen S Young44, Isis Ricano-Ponce28, Gosia Trynka28

Collaborative Association Study of Psoriasis (CASP)

Kristina Callis Duffin19, Cindy Helms18, David Goldgar19, Yun Li1, Justin Paschall87, M. J. Malloy88, C. R. Pullinger88, J. P. Kane88, J. Gardner18, A. Perlmutter89, A. Miner89, Bing Jian Feng19, Ravi Hiremagalore6, Robert W. Ike90, Enno Christophers31, Tilo Henseler31, Andreas Ruether5, Steven J. Schrodi91, Sampath Prahalad92, Stephen L Guthery92, Judith Fischer93, Wilson Liao94, Pui Kwok94, Alan Menter95, G. Mark Lathrop93, C. Wise96, Ann B. Begovich91

Psoriasis Association Genetics Extension (PAGE)

Fawnda J Pellett41, Andrew Henschel97, Marin Aurand97, Bruce Bebo97

Cooperative Research in the Region of Augsburg (KORA)

Christian Gieger98, Thomas Illig99

Heinz Nixdorf Recall (Risk Factors, Evaluation of Coronary Calcification, and Lifestyle) study (HNR)

Susanne Moebus100, Karl-Heinz Jöckel100, Raimund Erbel101

Footnotes

AUTHOR CONTRIBUTIONS

J.T.E., R.C.T. and G.R.A. designed and directed the study. R.P.N., M.W., J.D., J.V., J.T.E., F.C., J.N.B., M.A., C.S., A.D.B., C.G., A.R., J.Ke., X.E., W.W., J.Wo., R.T-A., M.S., G.N., L.S., R.M., M.C., J.S., A.F., S.W., S.K., K.K., T.E., A.M., A.B., G.K., D.G., P.R., U.M., F.N., A.H., J.W., S.S., C.W., C.L., S.E., R.A., V.C., and C.F.R., and H.B. contributed to sample collection and phenotyping. J.K. coordinated the GAP consortium’s samples and datasets. J.T.E. coordinated the PAGE samples and datasets. P.De., A.S., G.B., R.D.P., D.V., and C.C.A.S. contributed to the design of the Immunochip. J.K., P.E.S, G.R.A., and H.M.K. advised on the statistical analysis. C.L., S.E., R.A., H.B., E.E., P.H., and R.P.N. performed genotyping. E.E., S.L.S., L.C.T., and H.M.K. performed the genotype calling. S.L.S., L.C.T., Y.L., and D.J. performed genotype imputation and statistical analysis. F.C., J.N.B., J.E.G., T.T., J.T.E., and A.F. prepared Box 1. L.C.T., S.L.S., F.C. and J.T.E. drafted the manuscript, and prepared the figures and tables. E.E., J.E.G., J.K., P.E.S., R.P.N., R.C.T., T.T., G.R.A., J.N.K., and A.F. edited and revised the manuscript. All authors approved the final draft.

COMPETING FINANCIAL INTERESTS

The authors declare no competing financial interests.

Note: Supplementary information is available on the Nature Genetics website.

URLs

WTCCC common controls: http://www.wtccc.org.uk 1000 Genomes Project data are available at: ftp://ftp.1000genomes.ebi.ac.uk/vol1/ftp/release/20100804/ National Human Genome Research Institute (NHGRI) GWAS catalog: http://www.genome.gov/gwastudies eQTL database: http://www.sph.umich.edu/csg/junding/eQTL/TableDownload/

METHODS

Methods and any associated references are available in the online version of the paper at http://www.nature.com/naturegenetics/.

52

Dept Statistics, University of Oxford, Oxford OX1 3TG, UK;

53

Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, 100 Roberts Road, Suciaco, Western Australia 6008;

54

Genetics and Infection Laboratory, Cambridge Institute of Medical Research, Addenbrooke’s Hospital, Cambridge CB2 0XY, UK;

55

Division of Psychological Medicine and Psychiatry, Biomedical Research Centre for Mental Health at the Institute of Psychiatry, King’s College London

56

The South London and Maudsley NHS Foundation Trust, Denmark Hill, London SE5 8AF, UK;

57

Diamantina Institute of Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia;

58

Dept Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK;

59

Neuropsychiatric Genetics Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 2, Eire;

60

Dept Psychological Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK;

61

Molecular and Physiological Sciences, The Wellcome Trust, London NW1 2BE;

62

Centre for Gastroenterology, Bart’s and the London School of Medicine and Dentistry, London E1 2AT, UK;

63

Clinical Neurosciences, St George’s University of London, London SW17 0RE;

64

Oxford Centre for Diabetes, Endocrinology and Metabolism (ICDEM), Churchill Hospital, Oxford OX3 7LJ, UK;

65

Biomedical Research Centre, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK;

66

Social, Genetic and Developmental Psychiatry Centre, King’s College London Institute of Psychiatry, Denmark Hill, London SE5 8AF, UK;

67

University of Cambridge Dept Clinical Neurosciences, Addenbrooke’s Hospital, Cambridge CB2 2QQ, UK;

68

Dept Cardiovascular Science, University of Leicester, Glenfield Hospital, Leicester LE3 9QP;

69

Glaucoma Research Unit, Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD,UK;

70

Dept Genetics, University College London Institute of Ophthalmology, London EC1V 9EL, UK;

71

Dept Molecular Neuroscience, Institute of Neurology, Queen Square, London WC1N 3BG, UK;

72

Department of Dermatology and Venerology, University of Helsinki, Helsinki, Finland;

73

Asklepios Nordseeklinik, Westerland/Sylt, Germany;

74

TOMESA Clinics, Bad Salschlirf, Germany;

75

Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany;

76

Department of Dermatology, University of Göttingen, Göttingen, Germany;

77

Department of Dermatology, University of Münster, Münster, Germany;

78

Psoriasis Rehabilitation Hospital, Bad Bentheim, Germany;

79

Department of Endocrinology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands;

80

Department of Epidemiology and Biostatistics, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands;

81

University of Dundee, Dundee, UK;

82

University College Hospital Galway, Galway, Ireland;

83

St Vincent’s University Hospital, Dublin, Ireland;

84

Department of Clinical Medicine, Trinity College Dublin, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland;

85

Dermatology Service, Hospital del Mar-IMAS, Barcelona, Spain;

86

Department of Dermatology and Venereology, Sahlgrenska University Hospital, Gothenburg, Sweden;

87

National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA;

88

Cardiovascular Research Institute and Center for Human Genetics, University of California-San Francisco, CA;

89

Department of Psychiatry, Washington University School of Medicine, St. Louis, MO;

90

Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI;

91

Celera, 1401 Harbor Bay Parkway, Alameda, CA;

92

Departments of Pediatrics, Rheumatology and Gastroenterology, University of Utah, Salt Lake City, UT;

93

Centre National de Génotypage, Institut Génomique, Commissariat á l’Énergie Atomique, Evry, France;

94

Department of Dermatology, University of California, San Francisco;

95

Department of Dermatology, Baylor University Medical Center, Dallas, TX;

96

Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX;

97

National Psoriasis Foundation, Portland, OR 97223 USA;

98

Institute of Genetic Epidemiology, Helmholtz Centre Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany,

99

Research Unit Molecular Epidemiology, Helmholtz Centre Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany;

100

Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany;

101

Clinic of Cardiology, West German Heart Centre, University Hospital of Essen, University Duisburg-Essen, Essen, Germany.

References

  • 1.Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med. 2009;361:496–509. doi: 10.1056/NEJMra0804595. [DOI] [PubMed] [Google Scholar]
  • 2.Elder JT, et al. Molecular dissection of psoriasis: integrating genetics and biology. J Invest Dermatol. 2010;130:1213–26. doi: 10.1038/jid.2009.319. [DOI] [PubMed] [Google Scholar]
  • 3.Ellinghaus E, et al. Genome-wide association study identifies a psoriasis susceptibility locus at TRAF3IP2. Nat Genet. 2010;42:991–5. doi: 10.1038/ng.689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Nair RP, et al. Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways. Nat Genet. 2009;41:199–204. doi: 10.1038/ng.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Strange A, et al. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010;42:985–90. doi: 10.1038/ng.694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Stuart PE, et al. Genome-wide association analysis identifies three psoriasis susceptibility loci. Nat Genet. 2010;42:1000–4. doi: 10.1038/ng.693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sun LD, et al. Association analyses identify six new psoriasis susceptibility loci in the Chinese population. Nat Genet. 2010;42:1005–9. doi: 10.1038/ng.690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zhang XJ, et al. Psoriasis genome-wide association study identifies susceptibility variants within LCE gene cluster at 1q21. Nat Genet. 2009;41:205–10. doi: 10.1038/ng.310. [DOI] [PubMed] [Google Scholar]
  • 9.de Cid R, et al. Deletion of the late cornified envelope LCE3B and LCE3C genes as a susceptibility factor for psoriasis. Nat Genet. 2009;41:211–5. doi: 10.1038/ng.313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ellinghaus D, et al. Combined Analysis of Genome-wide Association Studies for Crohn Disease and Psoriasis Identifies Seven Shared Susceptibility Loci. Am J Hum Genet. 2012;90:636–47. doi: 10.1016/j.ajhg.2012.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cotsapas C, et al. Pervasive sharing of genetic effects in autoimmune disease. PLoS Genet. 2011;7:e1002254. doi: 10.1371/journal.pgen.1002254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Cortes A, Brown MA. Promise and pitfalls of the Immunochip. Arthritis Res Ther. 2011;13:101. doi: 10.1186/ar3204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Trynka G, et al. Dense genotyping identifies and localizes multiple common and rare variant association signals in celiac disease. Nat Genet. 2011;43:1193–201. doi: 10.1038/ng.998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Feng BJ, et al. Multiple Loci within the major histocompatibility complex confer risk of psoriasis. PLoS Genet. 2009;5:e1000606. doi: 10.1371/journal.pgen.1000606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zheng HF, et al. Variants in MHC, LCE and IL12B have epistatic effects on psoriasis risk in Chinese population. J Dermatol Sci. 2011;61:124–8. doi: 10.1016/j.jdermsci.2010.12.001. [DOI] [PubMed] [Google Scholar]
  • 16.Riveira-Munoz E, et al. Meta-analysis confirms the LCE3C_LCE3B deletion as a risk factor for psoriasis in several ethnic groups and finds interaction with HLA-Cw6. J Invest Dermatol. 2011;131:1105–9. doi: 10.1038/jid.2010.350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jordan CT, et al. PSORS2 is due to mutations in CARD14. Am J Hum Genet. 2012;90:784–95. doi: 10.1016/j.ajhg.2012.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Jordan CT, et al. Rare and common variants in CARD14, encoding an epidermal regulator of NF-kappaB, in psoriasis. Am J Hum Genet. 2012;90:796–808. doi: 10.1016/j.ajhg.2012.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Di Meglio P, et al. The IL23R R381Q gene variant protects against immune-mediated diseases by impairing IL-23-induced Th17 effector response in humans. PLoS One. 2011;6:e17160. doi: 10.1371/journal.pone.0017160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cal S, et al. Identification and characterization of human polyserase-3, a novel protein with tandem serine-protease domains in the same polypeptide chain. BMC Biochem. 2006;7:9. doi: 10.1186/1471-2091-7-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dubois PC, et al. Multiple common variants for celiac disease influencing immune gene expression. Nat Genet. 2010;42:295–302. doi: 10.1038/ng.543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhernakova A, et al. Meta-analysis of genome-wide association studies in celiac disease and rheumatoid arthritis identifies fourteen non-HLA shared loci. PLoS Genet. 2011;7:e1002004. doi: 10.1371/journal.pgen.1002004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Franke A, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet. 2010;42:1118–25. doi: 10.1038/ng.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gudjonsson JE, et al. Assessment of the psoriatic transcriptome in a large sample: additional regulated genes and comparisons with in vitro models. J Invest Dermatol. 2010;130:1829–40. doi: 10.1038/jid.2010.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ding J, et al. Gene expression in skin and lymphoblastoid cells: Refined statistical method reveals extensive overlap in cis-eQTL signals. Am J Hum Genet. 2010;87:779–89. doi: 10.1016/j.ajhg.2010.10.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mason CC, et al. Bimodal distribution of RNA expression levels in human skeletal muscle tissue. BMC Genomics. 2011;12:98. doi: 10.1186/1471-2164-12-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Song MY, Kim HE, Kim S, Choi IH, Lee JK. SNP-based large-scale identification of allele-specific gene expression in human B cells. Gene. 2011;493:211–8. doi: 10.1016/j.gene.2011.11.058. [DOI] [PubMed] [Google Scholar]
  • 28.Andres AM, et al. Balancing selection maintains a form of ERAP2 that undergoes nonsense-mediated decay and affects antigen presentation. PLoS Genet. 2010;6:e1001157. doi: 10.1371/journal.pgen.1001157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Grjibovski AM, Olsen AO, Magnus P, Harris JR. Psoriasis in Norwegian twins: contribution of genetic and environmental effects. J Eur Acad Dermatol Venereol. 2007;21:1337–43. doi: 10.1111/j.1468-3083.2007.02268.x. [DOI] [PubMed] [Google Scholar]
  • 30.Najarian DJ, Gottlieb AB. Connections between psoriasis and Crohn’s disease. J Am Acad Dermatol. 2003;48:805–21. doi: 10.1067/mjd.2003.540. quiz 822–4. [DOI] [PubMed] [Google Scholar]
  • 31.Ludvigsson JF, Lindelof B, Zingone F, Ciacci C. Psoriasis in a nationwide cohort study of patients with celiac disease. J Invest Dermatol. 2011;131:2010–6. doi: 10.1038/jid.2011.162. [DOI] [PubMed] [Google Scholar]
  • 32.Modlin RL. Innate immunity: ignored for decades, but not forgotten. J Invest Dermatol. 2012;132:882–6. doi: 10.1038/jid.2011.373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nakatsuji T, Gallo RL. Antimicrobial peptides: old molecules with new ideas. J Invest Dermatol. 2011;132:887–95. doi: 10.1038/jid.2011.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wolfle U, Martin S, Emde M, Schempp C. Dermatology in the Darwin anniversary. Part 2: Evolution of the skin-associated immune system. J DtschDermatol Ges. 2009;7:862–9. doi: 10.1111/j.1610-0387.2009.07202.x. [DOI] [PubMed] [Google Scholar]
  • 35.Capon F, Burden AD, Trembath RC, Barker JN. Psoriasis and other complex trait dermatoses: from Loci to functional pathways. J Invest Dermatol. 2012;132:915–22. doi: 10.1038/jid.2011.395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gudjonsson JE, Elder JT. Psoriasis. In: Goldsmith L, et al., editors. Dermatology in General Medicine. Vol. 1. McGraw-Hill; New York: 2012. pp. 197–231. [Google Scholar]
  • 37.Garber K. Psoriasis: from bed to bench and back. Nat Biotechnol. 2011;29:563–6. doi: 10.1038/nbt.1906. [DOI] [PubMed] [Google Scholar]
  • 38.Vilhais-Neto GC, et al. Rere controls retinoic acid signalling and somite bilateral symmetry. Nature. 2010;463:953–7. doi: 10.1038/nature08763. [DOI] [PubMed] [Google Scholar]
  • 39.Ferby I, et al. Mig6 is a negative regulator of EGF receptor-mediated skin morphogenesis and tumor formation. Nat Med. 2006;12:568–73. doi: 10.1038/nm1401. [DOI] [PubMed] [Google Scholar]
  • 40.Amler LC, et al. Identification and characterization of novel genes located at the t(1;15)(p36.2;q24) translocation breakpoint in the neuroblastoma cell line NGP. Genomics. 2000;64:195–202. doi: 10.1006/geno.1999.6097. [DOI] [PubMed] [Google Scholar]
  • 41.Djuretic IM, et al. Transcription factors T-bet and Runx3 cooperate to activate Ifng and silence Il4 in T helper type 1 cells. Nat Immunol. 2007;8:145–53. doi: 10.1038/ni1424. [DOI] [PubMed] [Google Scholar]
  • 42.Shiraishi N, et al. Identification and characterization of three novel beta 1,3-N-acetylglucosaminyltransferases structurally related to the beta 1,3-galactosyltransferase family. J Biol Chem. 2001;276:3498–507. doi: 10.1074/jbc.M004800200. [DOI] [PubMed] [Google Scholar]
  • 43.Togayachi A, et al. Beta3GnT2 (B3GNT2), a major polylactosamine synthase: analysis of B3GNT2-deficient mice. Methods Enzymol. 2010;479:185–204. doi: 10.1016/S0076-6879(10)79011-X. [DOI] [PubMed] [Google Scholar]
  • 44.Grindstaff KK, et al. Sec6/8 complex is recruited to cell-cell contacts and specifies transport vesicle delivery to the basal-lateral membrane in epithelial cells. Cell. 1998;93:731–40. doi: 10.1016/s0092-8674(00)81435-x. [DOI] [PubMed] [Google Scholar]
  • 45.Biswas PS, et al. Phosphorylation of IRF4 by ROCK2 regulates IL-17 and IL-21 production and the development of autoimmunity in mice. J Clin Invest. 2010;120:3280–95. doi: 10.1172/JCI42856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Mudter J, et al. IRF4 regulates IL-17A promoter activity and controls RORgammat-dependent Th17 colitis in vivo. Inflamm Bowel Dis. 2011;17:1343–58. doi: 10.1002/ibd.21476. [DOI] [PubMed] [Google Scholar]
  • 47.Huber M, et al. IRF4 is essential for IL-21-mediated induction, amplification, and stabilization of the Th17 phenotype. Proc Natl Acad Sci U S A. 2008;105:20846–51. doi: 10.1073/pnas.0809077106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bowcock AM, et al. Insights into psoriasis and other inflammatory diseases from large-scale gene expression studies. Hum Mol Genet. 2001;10:1793–805. doi: 10.1093/hmg/10.17.1793. [DOI] [PubMed] [Google Scholar]
  • 49.Chang IF, Hsiao HY. Induction of RhoGAP and pathological changes characteristic of Alzheimer’s disease by UAHFEMF discharge in rat brain. Curr Alzheimer Res. 2005;2:559–69. doi: 10.2174/156720505774932269. [DOI] [PubMed] [Google Scholar]
  • 50.Gotoh K, et al. Selective control of type I IFN induction by the Rac activator DOCK2 during TLR-mediated plasmacytoid dendritic cell activation. J Exp Med. 2010;207:721–30. doi: 10.1084/jem.20091776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ippagunta SK, et al. The inflammasome adaptor ASC regulates the function of adaptive immune cells by controlling Dock2-mediated Rac activation and actin polymerization. Nat Immunol. 2011;12:1010–6. doi: 10.1038/ni.2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Loo YM, Gale M., Jr Immune signaling by RIG-I-like receptors. Immunity. 2011;34:680–92. doi: 10.1016/j.immuni.2011.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Cui XF, Imaizumi T, Yoshida H, Borden EC, Satoh K. Retinoic acid-inducible gene-I is induced by interferon-gamma and regulates the expression of interferon-gamma stimulated gene 15 in MCF-7 cells. Biochem Cell Biol. 2004;82:401–5. doi: 10.1139/o04-041. [DOI] [PubMed] [Google Scholar]
  • 54.Negishi H, et al. A critical link between Toll-like receptor 3 and type II interferon signaling pathways in antiviral innate immunity. Proc Natl Acad Sci U S A. 2008;105:20446–51. doi: 10.1073/pnas.0810372105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Patel S, Xi ZF, Seo EY, McGaughey D, Segre JA. Klf4 and corticosteroids activate an overlapping set of transcriptional targets to accelerate in utero epidermal barrier acquisition. Proc Natl Acad Sci U S A. 2006;103:18668–73. doi: 10.1073/pnas.0608658103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Feinberg MW, et al. Kruppel-like factor 4 is a mediator of proinflammatory signaling in macrophages. J Biol Chem. 2005;280:38247–58. doi: 10.1074/jbc.M509378200. [DOI] [PubMed] [Google Scholar]
  • 57.Liang J, et al. A novel CCCH-zinc finger protein family regulates proinflammatory activation of macrophages. J Biol Chem. 2008;283:6337–46. doi: 10.1074/jbc.M707861200. [DOI] [PubMed] [Google Scholar]
  • 58.Nagarajan P, et al. Ets1 blocks terminal differentiation of keratinocytes and induces expressionof matrix metalloproteases and innate immune mediators. J Cell Sci. 2010;123:3566–75. doi: 10.1242/jcs.062240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zamisch M, et al. The transcription factor Ets1 is important for CD4 repression and Runx3 up-regulation during CD8 T cell differentiation in the thymus. J Exp Med. 2009;206:2685–99. doi: 10.1084/jem.20092024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Moisan J, Grenningloh R, Bettelli E, Oukka M, Ho IC. Ets-1 is a negative regulator of Th17 differentiation. J Exp Med. 2007;204:2825–35. doi: 10.1084/jem.20070994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Sakamoto H, et al. A Janus kinase inhibitor, JAB, is an interferon-gamma-inducible gene and confers resistance to interferons. Blood. 1998;92:1668–76. [PubMed] [Google Scholar]
  • 62.Tanaka K, et al. Loss of suppressor of cytokine signaling 1 in helper T cells leads to defective Th17 differentiation by enhancing antagonistic effects of IFN-gamma on STAT3and Smads. J Immunol. 2008;180:3746–56. doi: 10.4049/jimmunol.180.6.3746. [DOI] [PubMed] [Google Scholar]
  • 63.Piganis RA, et al. Suppressor of cytokine signaling (SOCS) 1 inhibits type I interferon (IFN) signaling via the interferon alpha receptor (IFNAR1)-associated tyrosine kinase Tyk2. J Biol Chem. 2011;286:33811–8. doi: 10.1074/jbc.M111.270207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Sano S, et al. Stat3 links activated keratinocytes and immunocytes required for development of psoriasis in a novel transgenic mouse model. Nat Med. 2005;11:43–9. doi: 10.1038/nm1162. [DOI] [PubMed] [Google Scholar]
  • 65.Harris TJ, et al. Cutting edge: An in vivo requirement for STAT3 signaling in TH17 development and TH17-dependent autoimmunity. J Immunol. 2007;179:4313–7. doi: 10.4049/jimmunol.179.7.4313. [DOI] [PubMed] [Google Scholar]
  • 66.Wei L, Laurence A, O’Shea JJ. New insights into the roles of Stat5a/b and Stat3 in T cell development and differentiation. Semin Cell Dev Biol. 2008;19:394–400. doi: 10.1016/j.semcdb.2008.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Blonska M, Lin X. NF-kappaB signaling pathways regulated by CARMA family of scaffold proteins. Cell Res. 2011;21:55–70. doi: 10.1038/cr.2010.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kersh EN. Impaired memory CD8 T cell development in the absence of methyl-CpG-binding domain protein 2. J Immunol. 2006;177:3821–6. doi: 10.4049/jimmunol.177.6.3821. [DOI] [PubMed] [Google Scholar]
  • 69.Faili A, et al. Induction of somatic hypermutation in immunoglobulin genes is dependent on DNA polymerase iota. Nature. 2002;419:944–7. doi: 10.1038/nature01117. [DOI] [PubMed] [Google Scholar]
  • 70.Inouye M, et al. An immune responsenetwork associated with blood lipid levels. PLoS Genet. 2010;6 doi: 10.1371/journal.pgen.1001113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Wichmann HE, Gieger C, Illig T. KORA-gen--resource for population genetics, controls and a broad spectrum of disease phenotypes. Gesundheitswesen. 2005;67 (Suppl 1):S26–30. doi: 10.1055/s-2005-858226. [DOI] [PubMed] [Google Scholar]
  • 72.1000 Genomes Project Consortium. A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061–73. doi: 10.1038/nature09534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Giannoulatou E, Yau C, Colella S, Ragoussis J, Holmes CC. GenoSNP: a variational Bayes within-sample SNP genotyping algorithm that does not require a reference population. Bioinformatics. 2008;24:2209–14. doi: 10.1093/bioinformatics/btn386. [DOI] [PubMed] [Google Scholar]
  • 74.Howie B, Fuchsberger C, Stephens M, Marchini J, Abecasis G. Fast and accurate genotype imputation in genome-wide association studies through pre-phasing. Nature Genetics. 2012;44:955–959. doi: 10.1038/ng.2354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Marchini J, Howie B, Myers S, McVean G, Donnelly P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat Genet. 2007;39:906–13. doi: 10.1038/ng2088. [DOI] [PubMed] [Google Scholar]
  • 76.Howie BN, Donnelly P, Marchini J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet. 2009;5:e1000529. doi: 10.1371/journal.pgen.1000529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Altshuler DM, et al. Integrating common and rare genetic variation in diverse human populations. Nature. 2010;467:52–8. doi: 10.1038/nature09298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Purcell S, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–75. doi: 10.1086/519795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kang HM, et al. Variance component model to account for sample structure in genome-wide association studies. Nat Genet. 2010;42:348–54. doi: 10.1038/ng.548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Devlin B, Roeder K. Genomic control for association studies. Biometrics. 1999;55:997–1004. doi: 10.1111/j.0006-341x.1999.00997.x. [DOI] [PubMed] [Google Scholar]
  • 81.Freedman ML, et al. Assessing the impact of population stratification on genetic association studies. Nat Genet. 2004;36:388–93. doi: 10.1038/ng1333. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES