ArticlePDF Available

Seven newly identified loci for autoimmune thyroid disease

Authors:

Abstract and Figures

Autoimmune thyroid disease (AITD), including Graves' disease (GD) and Hashimoto's thyroiditis (HT), is one of the most common of the immune-mediated diseases. To further investigate the genetic determinants of AITD, we conducted an association study using a custom-made single-nucleotide polymorphism (SNP) array, the ImmunoChip. The SNP array contains all known and genotype-able SNPs across 186 distinct susceptibility loci associated with one or more immune-mediated diseases. After stringent quality control, we analysed 103 875 common SNPs (minor allele frequency >0.05) in 2285 GD and 462 HT patients and 9364 controls. We found evidence for seven new AITD risk loci (P < 1.12 × 10(-6); a permutation test derived significance threshold), five at locations previously associated and two at locations awaiting confirmation, with other immune-mediated diseases.
Content may be subject to copyright.
Seven newly identified loci for autoimmune thyroid
disease
Jason D. Cooper1,,
{
, Matthew J. Simmonds2,
{
, Neil M. Walker1, Oliver Burren1, Oliver J. Brand2,
Hui Guo1, Chris Wallace1, Helen Stevens1, Gillian Coleman1, Wellcome Trust Case Control
Consortium
{
, Jayne A. Franklyn3, John A. Todd1,
}
and Stephen C.L. Gough2,4,
}
1
Department of Medical Genetics, Cambridge Institute for Medical Research, Juvenile Diabetes Research Foundation/
Wellcome Trust Diabetes and Inflammation Laboratory, NIHR Cambridge Biomedical Research Centre, University of
Cambridge, Addenbrooke’s Hospital, Cambridge,
2
Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford,
3
School of Clinical and Experimental Medicine, Institute of Biomedical
Research, University of Birmingham, Birmingham, and
4
NIHR Oxford Biomedical Research Centre, Churchill Hospital,
Oxford, UK
Received May 31, 2012; Revised August 6, 2012; Accepted August 20, 2012
Autoimmune thyroid disease (AITD), including Graves’ disease (GD) and Hashimoto’s thyroiditis (HT), is one
of the most common of the immune-mediated diseases. To further investigate the genetic determinants of
AITD, we conducted an association study using a custom-made single-nucleotide polymorphism (SNP)
array, the ImmunoChip. The SNP array contains all known and genotype-able SNPs across 186 distinct sus-
ceptibility loci associated with one or more immune-mediated diseases. After stringent quality control, we
analysed 103 875 common SNPs (minor allele frequency >0.05) in 2285 GD and 462 HT patients and 9364 con-
trols. We found evidence for seven new AITD risk loci (P<1.12 310
26
; a permutation test derived signifi-
cance threshold), five at locations previously associated and two at locations awaiting confirmation, with
other immune-mediated diseases.
INTRODUCTION
Genome-wide association (GWA) studies have had a dramatic
impact on susceptibility locus discovery and in addition, high-
lighted and extended the previously observed (1,2) commonal-
ity of loci between immune-mediated diseases, which has
ultimately resulted in the ImmunoChip project. As described
previously (3), the ImmunoChip project is a collaboration
between 12 immune-mediated disease groups [autoimmune
thyroid disease (AITD), ankylosing spondylitis (AS), celiac
disease (CeD), Crohn’s disease (CD), IgA deficiency, multiple
sclerosis (MS), primary biliary cirrhosis (PBC), psoriasis (PS),
rheumatoid arthritis (RA), systemic lupus erythematosus
(SLE), type 1 diabetes (T1D) and ulcerative colitis (UC)]
and the Wellcome Trust Case Control Consortium
(WTCCC). The main objective of the project was to provide
cost-effective genotyping of all known and genotype-able
single-nucleotide polymorphisms (SNPs) across confirmed
and distinct susceptibility loci from the 12 disease groups
(www.ImmunoBase.org). The result was the ImmunoChip, a
custom Illumina 200 K Infinium high-density array containing
186 distinct susceptibility loci (3), mainly derived from GWA
studies, which have been convincingly associated (required
P,5×10
28
) with one or more immune-mediated diseases.
In addition, the ImmunoChip contained wildcard SNPs provided
by the 12 disease groups and SNPs for replication found in non-
immunological disease GWA studies conducted by the WTCCC
(www.wtccc.org.uk/ccc2/wtccc2_studies.shtml).
The AITDs are caused by an autoimmune attack in which
antibodies directed against components of the thyroid gland
To whom correspondence should be addressed. Tel: +44 1223762107; Fax: +44 1223762102; Email jason.cooper@cimr.cam.ac.uk
Joint first authors.
A full list of the Wellcome Trust Case Control Consortium investigators involved in this study are included in the Supplementary Information.
}
Joint senior authors.
#The Author 2012. Published by Oxford University Press.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/
licenses/by-nc/2.5), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is prop-
erly cited.
Human Molecular Genetics, 2012, Vol. 21, No. 23 5202–5208
doi:10.1093/hmg/dds357
Advance Access published on August 24, 2012
lead to either thyroid hormone excess and clinical features of
hyperthyroidism as seen in people with Graves’ disease (GD) or
thyroid gland damage, reduced thyroid hormone production and
clinical features of hypothyroidism as seen in people with
Hashimoto’s thyroiditis (HT). AITD shows a strong female pre-
ponderance with estimated incidence rates of GD in females as
80 per 100 000 person-years and in males as 8 per 100 000
person-years, and of HT in females as 350 per 100 000
person-years and in males as 8 per 100 000 person-years (4).
Although many regions of the genome have been reported
as AITD susceptibility loci, convincing evidence for associ-
ation has been limited to four regions: PTPN22/1q13.2,
CTLA4/2q33.2, HLA/6p21, and TSHR/14q31.1 (5). These sus-
ceptibility loci all fall within the antigen presentation/T-cell
receptor signalling pathway. In addition, there is mounting
evidence for a susceptibility locus in FCRL3/1q23.1 (57),
SCGB3A2/5q32 (8,9) and IL2RA/10p15.1 (10,11), and a
recent GWA and replication study in subjects recruited from
the Chinese Han population identified two novel loci at 4p14
and RNASET2-FGFR1OP-CCR6/6q27 (5).
Here, we used the ImmunoChip to further investigate the
genetic architecture of AITD in 2374 GD and 474 HT patients,
and 9953 controls.
RESULTS
After consideration of the number of AITD patients available,
we focused on susceptibility locus discovery using common
SNPs [minor allele frequency (MAF) 0.05] outside of the
HLA, which reduced the number of tests performed and con-
sequently, allowed us to use a less stringent significance
threshold. We adopted a permutation approach to correct the
0.05 level of significance for multiple testing and for correl-
ation between SNP genotypes, and obtained a threshold P¼
1.12 ×10
26
(see Materials and Methods). After stringent
quality control (see Materials and Methods), we analysed
103 875 common SNP genotypes in 2285 GD and 462 HT
patients, and 9364 controls. As the vast majority of our
AITD patients have been previously analysed in either discov-
ery or confirmation studies of the reported AITD susceptibility
loci, we initially used the AITD susceptibility loci on the
ImmunoChip as positive controls and as expected, found evi-
dence of association (P,1.12 ×10
26
and good genotype
signal intensity clusters; Supplementary Material, Table S1)
for PTPN22 (P¼9.7 ×10
223
), CTLA4 (P¼2.1 ×10
223
),
IL2RA (P¼2.7 ×10
27
) and TSHR (P¼1.3 ×10
238
in GD
patients/controls). We note that the AITD risk locus
SCGB3A2 was not on the ImmunoChip, but had previously
been associated in our AITD patients (P¼0.007; (8)). In add-
ition, we found some evidence of association for FCRL3 (P¼
1.1 ×10
25
in GD patients/controls; Supplementary Material,
Table S1) and found further evidence of association for the
recently reported RNASET2FGFR1OP CCR6/6q27
locus (5) (imm_6_167 338101 P¼1.6 ×10
27
;r
2
between
imm_6_167338101 and rs9355610 (5) in controls ¼0.72;
Table 1; Supplementary Material, Fig. S1) in subjects with
European ancestry. The recently reported 4p14 locus (5) was
not on the ImmunoChip.
We found evidence of association between AITD and seven
genomic locations (P,1.12 ×10
26
; Table 1). Five of these
new AITD risk loci were at locations previously associated
with at least one other immune-mediated disease (Table 1; Supple-
mentary Material, Figs S2S6); three of which included the most
AITD-associated SNPs being located within MMEL1/1p36.32,
LPP/3q28 and BACH2/6q15 genes. However, only BACH2 has
a known autoimmune function as a regulator of nucleic acid-
triggered antiviral responses in human cells (12). The remaining
two new AITD risk loci were unconfirmed risk loci: rs1534422
at 2p25.1 is an unconfirmed T1D risk locus (T1D P¼2.1 ×
10
26
;(13); Supplementary Material, Fig. S7); and rs4409785 at
11q21 is an unconfirmed MS risk locus (MS P¼6.3 ×10
27
;
(14); Supplementary Material, Fig. S8).
We tested for heterogeneity in disease association before
combining the GD and HT patients (Table 1and Supplemen-
tary Material, Table S1). After allowing for multiple testing,
only the disease association of the TSHR/14q31.1 locus was
significantly different between the two patient groups. This
difference between GD and HT patients was expected as
the TSHR represents the primary autoantigen in hyperthy-
roidism of GD (15). As we focused on susceptibility loci
discovery using common SNPs (MAF .0.05) outside of
the HLA to reduce the number of tests performed, which
was necessary given the number of GD and HT patients
available in this study, we did not proceed to test whether
the most disease-associated SNP alone was sufficient to
model the association signal in AITD risk loci with dense
SNP coverage.
We examined the overlap between AITD and the other 11
immune-mediated diseases that provided susceptibility loci
to the ImmunoChip in turn, by testing whether the pattern of
AITD association across immune-mediated disease regions
differed according to whether those regions had previously
been associated with each specific immune-mediated disease
or not (Table 2;(16)). We restricted the analysis to
non-HLA SNPs from densely mapped regions associated
with at least one immune-mediated disease. We found that
SNPs within RA-associated regions were significantly (P¼
6.56 ×10
25
) more likely to be associated with AITD than
SNPs within regions not associated with RA. In addition,
there was marginal evidence of an overlap between AITD
and T1D-associated regions (P¼0.0327). In contrast, we
found that SNPs within regions associated with AS, UC and
PS showed significantly less evidence of association with
AITD than SNPs within regions known to be associated
with each disease (P¼3.27 ×10
24
, 1.69 ×10
23
and
4.08 ×10
23
, respectively). There was also marginal evidence
of an overlap between AITD and regions not associated with
CD (P¼0.0250). We note that an important caveat about
interpreting these test results is that the susceptibility loci
for each disease are incomplete and the extent of the incom-
pleteness varies between diseases. This prevents us, for
example, from drawing any conclusions that ‘AITD is less
like AS than UC’. However, for the majority of the immune-
mediated diseases listed in Table 2, we can assume that the
common loci with the largest effects are known and that sig-
nificant results here are true representations of AITD-disease
overlap.
Human Molecular Genetics, 2012, Vol. 21, No. 23 5203
Table 1. Single locus test evidence for the seven newly identified AITD non-HLA susceptibility loci and 6q27 on the ImmunoChip.
Karyotype
band
Most
disease-associated
SNP
Located within
the gene
MAF in
controls
Immune-mediated diseases previously
associated with regions (P,5×10
28
)
(reported SNP; r
2
with reported SNP in
controls)
2285 GD patients and 9364
controls
462 HT patients and 9364
controls
Heterogeneity in
disease association
2282 GD, 451 HT patients and
9364 controls
a
OR
(95% CI)
P-value OR (95% CI) P-value P-value OR (95% CI) P-value
1p36.32 rs2843403
C.T
MMEL1 (32 kb
upstream of
TNFRSF1)
0.362 CeD (26) (rs3748816/TNFRSF14-MMEL1;
r
2
¼0.99)
PBC (27) (rs3748816; r
2
¼0.99)
RA (28) (rs3890745/TNFRSF14-MMEL1;
r
2
¼0.90)
0.84 (0.79– 0.90) 7.94 310
27
0.97 (0.85–1.12) 0.696 0.0593 0.86 (0.81– 0.92) 5.63 ×10
26
2p25.1 rs1534422
A.G
216 kb upstream
of TRIB2
0.455 Unconfirmed T1D locus
b
1.16 (1.09– 1.24) 4.69 ×10
26
1.24 (1.08– 1.41) 1.64 ×10
23
0.394 1.17 (1.11– 1.25) 1.76 310
27
3q27.3/
3q28
rs13093110
C.T
LPP 0.452 CeD (26) (rs1464510; r
2
¼0.98) 1.18 (1.10– 1.26) 8.17 ×10
27
1.20 (1.05– 1.37) 7.09 ×10
23
0.797 1.19 (1.12– 1.26) 3.69 310
28
6q15 rs72928038
G.A
BACH2 0.177 CD (29) (rs1847472; r
2
¼0.39)
CeD (26) (rs10806425/BACH2-MAP3K7;
r
2
¼0.30)
T1D (21) (rs11755527; r
2
¼0.19)
c
1.21 (1.12– 1.32) 3.63 ×10
26
1.30 (1.11– 1.53) 1.36 ×10
23
0.417 1.23 (1.14– 1.32) 1.23 310
27
6q27 imm_6_167338101
A.C
FGFR1OP
(118 kb
upstream of
CCR6)
0.408 AITD (5) (rs9355610; r
2
¼0.72)
CD (30) (rs2301436/CCR6;r
2
¼0.61)
RA (31) (rs3093023/CCR6;r
2
¼0.25
Vitiligo (32) (rs2236313; r
2
¼0.22)
0.84 (0.79– 0.90) 3.30 ×10
27
0.88 (0.76–1.00) 0.0564 0.575 0.85 (0.80– 0.90) 1.64 310
27
11q21 rs4409785
T.C
0.173 Unconfirmed MS locus
d
1.21 (1.11– 1.31) 5.37 ×10
26
1.34 (1.14– 1.57) 3.54 ×10
24
0.234 1.23 (1.14– 1.33) 7.69 310
28
12q12 rs4768412
C.T
PRICKLE1 0.363 CD (30) (rs11175593/LRRK2-MUC19)
d
1.19 (1.11– 1.27) 3.30 310
27
1.00 (0.88– 1.15) 0.949 0.0234
e
1.16 (1.09– 1.23) 3.56 ×10
26
16p11.2 rs57348955
G.A
83 kb upstream
of ITGAM
0.396 SLE (33) (rs1143679/ITGAM;r
2
¼0.056)
T1D (13) (rs4988084/IL27)
f
0.83 (0.77– 0.89) 3.76 310
28
0.91 (0.80–1.05) 0.188 0.190 0.84 (0.79– 0.89) 5.13 ×10
28
The reported SNPs have a P-value of ,1.12 ×10
26
(see Materials and Methods) and were the most disease-associated SNP in the region with good genotype signal intensity plots (www.ImmunoB ase.org). For each SNP, the most
significant P-value is shown in bold. Note that the same controls were analysed in each analysis. AITD, autoimmune thyroid disease; MAF, minor allele frequency; HT, Hashimoto’s thyroiditis; GD, Graves’ disease; OR, Odds ratio for
the minor allele; CI, confidence interval; CD, Crohn’s disease; CeD, celiac disease; PBC, primary biliary cirrhosis; and SLE, systematic lupus erythematosus.
a
We excluded 14 second degree relatives or closer between disease groups (see Materials and Methods).
b
rs1534422 at 2p25.1 is an unconfirmed T1D risk locus (T1D P¼2.1 ×10
26
;(13)).
c
We used CEU data from the 1000 Genomes Project to estimate r
2
as the T1D SNP, rs11755527, was not available in the ImmunoChip data.
d
rs4409785 at 11q21 is an unconfirmed MS risk locus [MS P¼6.3 ×10
27
;(14)].
e
Assuming 13 [8 (Table 1) and 5 (Supplementary Material, Table S1)] independent tests, the adjusted P-value was 3.85 ×10
23
for the 0.05 level of significance based on the Bonferroni correction for multiple testing.
f
No available genotype data which included both SNPs.
5204 Human Molecular Genetics, 2012, Vol. 21, No. 23
DISCUSSION
We have found evidence for seven risk loci not previously
associated with AITD and expect that even more AITD
risk loci will be discovered as more AITD patients are gen-
otyped on the ImmunoChip. In this study, as the Immuno-
Chip contains densely mapped immune-mediated disease
loci representing only a small proportion, 1%, of the
genome, we used a permutation approach to derive a signifi-
cance level rather than adopting a genome-wide significance
level. Importantly, regardless of which discovery significance
level was adopted, confirmation of the new AITD risk loci
will require independent replication evidence. We found
some statistical evidence that AITD, outside of the HLA,
has a shared genetic architecture with both RA and T1D.
AITD often co-exists with other immune-mediated diseases
and a recent study estimating the prevalence of these
co-existing disorders found that RA was the most common,
occurring in 3.15% of GD patients and 4.24% of HT patients
studied; T1D occurred in 1.11% of GD patients and 1.01% of
HT patients (17). We note that the prevalence study (17)
included vast majority of our AITD patients, and that
AITD patients were recruited from endocrine clinics and
not rheumatology clinics.
The implicated candidate genes for the established and
seven new AITD risk loci indicate the importance of T
lymphocyte signalling (PTPN22), T-regulatory cell function
(CTLA4 and IL2RA), lymphocyte trafficking (CCR6) and a
newly identified possible link with the anti-viral immune re-
sponse (BACH2). Nevertheless, other AITD-associated
regions suggest candidate genes of uncertain or unknown
functions or roles in autoimmunity of AITD, such as LPP or
TRIB2. Two regions, 11q21 and 12q12, have no annotated
protein-coding candidate genes, indicating how much of the
aetiology of this and other immune-mediated diseases
remain to be explained.
MATERIALS AND METHODS
Subjects
The British AITD cases consisted of 2374 (390 males and 1984
females) GD and 474 (69 males and 405 females) HT patients
(18). The British control population consisted of 6894 subjects
drawn from the British 1958 Birth Cohort (1958BC; http://
wwww.cls.ioe.ac.uk/studies.asp?section=000100020003) and
3059 subjects drawn from the UK Blood Services Common
Control Collection (UKBS-CC) (6,19). All subjects were of
white European ancestry with written informed consent and
Ethics Committee/Institutional Review Board approval.
SNP selection
The dense SNP map for 186 confirmed and distinct loci,
mainly derived from GWA studies, which have been asso-
ciated (required P,5×10
28
) with one or more autoimmune
diseases, consisted of all known SNPs, and small insertion
deletions, in the dbSNP database, in the 1000 Genomes
project (February 2010 release) and in additional sequencing
data provided by collaborators. The ImmunoChip contains
196 524 polymorphisms (718 insertion/deletions and 195 806
SNPs) (3,20).
Genotyping
Samples were genotyped using a custom Illumina 200K Infi-
nium high-density array according to the manufacturer’s
protocol at the Wellcome Trust Sanger Institute (WTSI) in
Hinxton, UK, and University of Virginia (UVA) in Charlottes-
ville, USA. In WTSI, 2374 GD, 474 HT, 1478 1958BC and
3059 UKBS-CC samples were processed, and in UVA, 5416
1958BC samples were processed.
Sample quality control
Samples were excluded based on per-sample call rate (Supple-
mentary Material, Figs S9 S11), outlying autosomal heterozy-
gosity (Supplementary Material, Figs S9 S11), inconsistent
recorded and genotype-inferred sex, non-European ancestry
(21) (Supplementary Material, Figs S12 S14), duplication
and being closely related to another sample in the study. Re-
latedness between the study participants was estimated by a
identity by state (IBS) statistic, we defined duplicates, or mono-
zygotic twins, as having an IBS .0.98 and second degree rela-
tives or closer, as having an IBS .0.1875 (22); the sample with
the lower call rate was excluded from further analyses. Finally,
we checked control sample’s identity using the 169 intentional
duplicates between the WTSI and the UVA, and using in-house
genotype data. We identified 256 samples, which had incorrect
identifiers as a result of manifest misreading, plate rotation and
chip-related issues, and excluded 95 samples from unknown
subjects (i.e. could not be assigned to identifiers). In addition,
we found ten additional duplicates between the genotyping
centers. All potential sample identity errors were discussed
with the relevant genotyping centers by Neil M. Walker. After
sample quality control, 2285 GD, 462 HT, 6541 1958BC and
Table 2. The overlap between AITD and the 11 other immune-mediated dis-
eases that provided susceptibility loci to the ImmunoChip. A negative
Z-value indicates that SNPs within disease-associated regions were more likely
to be associated with AITD than SNPs within regions not known to be asso-
ciated with the disease. A positive Z-value indicates that SNPs within regions
not known to be associated with the disease were more likely to be associated
with AITD than SNPs within associated regions.
Immune-mediated
disease
Number of
non-HLA
loci
Number of
SNPs within
loci
Wilcoxon rank test
ZP
AS 8 2079 3.593 3.27 ×10
24
CeD 30 9866 0.147 0.883
CD 66 22 585 2.242 0.0250
IgA deficiency 1 122 21.208 0.227
MS 26 7147 20.277 0.782
PBC 1 209 20.840 0.401
PS 25 6767 2.872 4.08 ×10
23
RA 25 7742 23.992 6.56 ×10
25
SLE 35 9099 20.726 0.468
T1D 41 12 658 22.146 0.0327
UC 28 8901 3.140 1.69 ×10
23
Human Molecular Genetics, 2012, Vol. 21, No. 23 5205
2823 UKBS-CC samples were available for analysis. When we
analysed GD and HT patients together, we excluded 14 second
degree relatives or closer between the disease groups, which
resulted in 2282 GD and 451 HT samples.
SNP quality control
SNP genotypes were called using Illumina GenomeStudio
GenTrain 2.0 algorithm and genotypes with a gencall score
(a quality metric for the distance a sample is from the center
of the nearest genotype cluster) ,0.15 were not assigned gen-
otypes. SNPs were excluded if the MAF fell ,5% in controls,
if they deviated from Hardy Weinberg equilibrium (HWE;
z-score .5) in controls or if the per-SNP call rate fell
,95% in cases or controls. We note that SNP quality
control was performed in WTSI and UVA-processed controls
separately. In addition, for SNPs from the pseudoautosomal
regions on chromosome X, we tested for deviation from
HWE in male and female controls separately after discovering
SNP genotype frequency differences between sexes in patients
and controls (e.g. rs306891). We excluded SNPs from the
HLA (NCBI build 36 coordinates chr6:29690000-33498585).
Previously, despite differences in subject’s age and in DNA
processing between 1958BC and UKBS-CC control subjects,
few differences in SNP allele frequencies between 1958BC
and UKBS-CC controls were observed (19), which justified
the combination of these two control groups into a single
control group. To assess the possible differential genotyping
errors between the WTSI and UVA genotyping centers,
we tested for differences in SNP allele frequencies in controls
between genotyping centers; the test was stratified by
geographical region of Great Britain (23). The quantile quan-
tile plot for the WTSI control versus UVA control 1
degree-of-freedom (1-df) association tests (Supplementary
Material, Fig. S15) showed a slight inflation of the test statistic
[inflation factor (
l
) was 1.017; as the inflation factor scales
with sample size, a more informative and comparable
measure of inflation is the inflation factor for an equivalent
study of 1000 cases and 1000 controls,
l
1000
(24), which
was 1.004 for the WTSI control/UVA control analysis]. As
the ImmunoChip was designed to include all known and
genotype-able SNPs across 186 distinct immune-mediated
disease susceptibility loci, there was inflation in the test statis-
tic for the AITD patient versus control association tests: HT
patient/control
l
1000
¼1.06 (l¼1.05); GD patient/control
l
1000
¼1.08 (
l
¼1.28); and HT and GD patient/control
l
1000
¼1.08 (l¼1.33). We note that
l
was based on
non-HLA common SNPs from regions not previously asso-
ciated with AITD (excluded regions reported in Supplemen-
tary Material, Table S1) that passed quality control. We also
estimated the inflation factor based on the WTCCC replication
SNPs for reading and maths ability, and psychosis endopheno-
types: HT patient/control
l
1000
¼1.02 (
l
¼1.02); GD patient/
control
l
1000
¼1.03 (
l
¼1.11); and HT and GD patient/
control
l
1000
¼1.04 (
l
¼1.17). We note that 2605 WTCCC
replication SNPs passed quality control and had a MAF
.0.05, and that the observed inflation of the test statistic
was a result of the replication SNPs overlapping some of the
immune-mediated disease loci, for example, the T1D loci
COBL/7p12.1 and GLIS3/9p24.2. As the inflation factor
l
1000
of the test statistics from the WTSI control/UVA
control analysis was just over 1 and we found a similar infla-
tion of the test statistics for GD patient/controls and for HT
patient/controls at 6 8% in the main analysis and at 2 3%
in the subset of WTCCC replication SNPs, population struc-
ture is unlikely to a major source of inflation. After SNP
quality control, 103 875 common SNPs were available for
analysis.
Statistical analysis
All statistical analyses were performed in R (http://www.r-p
roject.org) using the snpStats package available from the Bio-
conductor project (http://www.bioconductor.org). After apply-
ing sample and SNP quality control exclusions, we analysed
the case/control data using the 1-df Cochran-Armitage trend
test. SNPs were modelled using a numerical indicator variable
coded 0, 1 and 2, representing the occurrences of the minor
allele and assuming multiplicative allelic effects. We also
tested the disease-associated SNPs reported in Table 1for dif-
ferences between the multiplicative allelic effects model and
the full genotype model, and no significant differences were
found. We analysed SNPs on the X-chromosome using the
method proposed by Clayton (25).
We initially analysed the GD and HT patients separately,
but against the same controls. Before combining the GD and
HT patients into a single patient group, as we had analysed
GD and HT patients against the same controls, we tested for
disease association heterogeneity between the two AITDs by
testing for differences in SNP allele frequencies between GD
and HT patient groups. In the absence of disease association
heterogeneity, we proceeded to combine the GD and HT
patients and re-analysed.
As GD and HT patients, unlike the controls, were not
recruited from all 12 geographical regions of Great Britain
(seven and six regions, respectively), we did not stratify the ana-
lysis by geographical region to allow for population structure.
We note that previously, little difference was reported
between analyses with or without regional stratification (19).
However, to check that we were gaining power with the add-
itional controls rather than false-positive findings through ig-
noring the population structure, we tested for population
heterogeneity in SNP genotype frequencies across the regions
to identify SNPs that differentiate between the regions (Supple-
mentary Material, Table S2). None of the seven new or previ-
ously reported AITD risk loci showed large allele frequency
differences between controls from the 12 regions.
To adjust P-values for the multiple tests and to allow for the
correlation between SNP genotypes, we adopted a permutation
test approach. We performed 100 000 permutation tests of the
common SNPs on the ImmunoChip, permuting the disease
status label for each test, to provide the distribution of the
test statistic under the null hypothesis of no association
between SNP genotypes and disease status. The adjusted
P-value for 0.05 was 1.12 ×10
26
.
To examine the overlap between AITD and the other 11
immune-mediated diseases that provided susceptibility loci
to the ImmunoChip, we tested whether the distribution of
AITD test statistics varied between SNPs within regions asso-
ciated with another immune-mediated disease and within
5206 Human Molecular Genetics, 2012, Vol. 21, No. 23
regions not associated with that disease. We adopted a gene set
enrichment approach based on the Wilcoxon rank test as
implemented in the R package wgsea (Chris Wallace,
www-gene.cimr.cam.ac.uk/staff/wallace) (16). SNPs within
the non-HLA immune-mediated disease loci were divided
into two groups for each immune-mediated disease: (i) SNPs
within regions associated with disease at the time the Immuno-
Chip was designed; and (ii) SNPs within regions not asso-
ciated with that disease. We then tested the null hypothesis
that the distribution of P-values describing the evidence for as-
sociation of each SNP with AITD was the same in the two
groups using a Wilcoxon rank test with a permutation-based
variance estimate to allow for the considerable between SNP
correlation (16). We used 10 000 permutations to estimate
the variance of the Wilcoxon rank test.
SUPPLEMENTARY MATERIAL
Supplementary Material is available at HMG online.
ACKNOWLEDGEMENTS
We would like to thank all AITD patients and control subjects
for participating in this study. We thank nurses and doctors
for recruiting AITD patients into the AITD National Collection.
We thank the members of each disease consortium who initiated
and sustained the cross-disease ImmunoChip project. We thank
Jeffrey Barrett for assistance with ImmunoChip SNP selection
and for ImmunoChip-related correspondence. We thank Jenni-
fer Stone for co-ordinating the ImmunoChip design and produc-
tion at Illumina. The authors wish to acknowledge Sarah Edkins,
Emma Gray, Doug Simpkin, Sarah Hunt and staff of the Sanger
Institute’s Sample Logistics, Genotyping and Variation Inform-
atics facilities, respectively. The Diabetes and Inflammation La-
boratory is funded by the Juvenile Diabetes Research
Foundation International, the Wellcome Trust and the National
Institute for Health Research Cambridge Biomedical Centre.
The Cambridge Institute for Medical Research (CIMR) is in
receipt of a Wellcome Trust Strategic Award (079895). Core in-
frastructure support to the Wellcome Trust Centre for Human
Genetics, Oxford was provided by grant 090532/Z/09/Z. The
WTCCC is funded by Wellcome Trust awards 076113 and
083948. CW is supported by the Wellcome Trust (089989).
We acknowledge use of DNA from The UK Blood Services col-
lection of Common Controls (UKBS-CC collection), which is
funded by the Wellcome Trust grant 076113/C/04/Z and by
the USA National Institute for Health Research program grant
to the National Health Service Blood and Transplant
(RP-PG-0310-1002). We acknowledge the use of DNA from
the British 1958 Birth Cohort collection, which is funded by
the UK Medical Research Council grant G0000934 and the
Wellcome Trust grant 068545/Z/02. This research utilized
resources provided by the Type 1 Diabetes Genetics Consor-
tium, a collaborative clinical study sponsored by the National
Institute of Diabetes and Digestive and Kidney Diseases, the
National Institute of Allergy and Infectious Diseases, the Na-
tional Human Genome Research Institute, the National Institute
of Child Health and Human Development and the Juvenile
Diabetes Research Foundation International and is supported
by the USA National Institutes of Health grant U01-DK062418.
Conflict of Interest statement. None declared.
FUNDING
The research leading to these results has received funding from
the European Union’s 7th Framework Programme (FP7/2007
2013) under grant agreement No. 241447 [Natural immunomo-
dulators as novel immunotherapies for type 1 diabetes
(NAIMIT)]. Funding to pay the Open Access publication
charges for this article was provided by the Wellcome Trust.
REFERENCES
1. Smyth, D., Cooper, J.D., Collins, J.E., Heward, J.M., Franklyn, J.A.,
Howson, J.M., Vella, A., Nutland, S., Rance, H.E., Maier, L. et al. (2004)
Replication of an association between the lymphoid tyrosine phosphatase
locus (LYP/PTPN22) with type 1 diabetes, and evidence for its role as a
general autoimmunity locus. Diabetes,53, 3020– 3023.
2. Ueda, H., Howson, J.M., Esposito, L., Heward, J., Snook, H.,
Chamberlain, G., Rainbow, D.B., Hunter, K.M., Smith, A.N., Di Genova,
G. et al. (2003) Association of the T-cell regulatory gene CTLA4 with
susceptibility to autoimmune disease. Nature,423, 506– 511.
3. Trynka, G., Hunt, K.A., Bockett, N.A., Romanos, J., Mistry, V., Szperl,
A., Bakker, S.F., Bardella, M.T., Bhaw-Rosun, L., Castillejo, G. et al.
(2011) Dense genotyping identifies and localizes multiple common
and rare variant association signals in celiac disease. Nat. Genet.,43,
11931201.
4. McGrogan, A., Seaman, H.E., Wright, J.W. and de Vries, C.S. (2008) The
incidence of autoimmune thyroid disease: a systematic review of the
literature. Clin. Endocrinol. (Oxf.),69, 687– 696.
5. Chu, X., Pan, C.M., Zhao, S.X., Liang, J., Gao, G.Q., Zhang, X.M., Yuan,
G.Y., Li, C.G., Xue, L.Q., Shen, M. et al. (2011) A genome-wide
association study identifies two new risk loci for Graves’ disease. Nat.
Genet.,43, 897– 901.
6. Burton, P.R., Clayton, D.G., Cardon, L.R., Craddock, N., Deloukas, P.,
Duncanson, A., Kwiatkowski, D.P., McCarthy, M.I., Ouwehand, W.H.,
Samani, N.J. et al. (2007) Association scan of 14,500 nonsynonymous
SNPs in four diseases identifies autoimmunity variants. Nat. Genet.,39,
13291337.
7. Kochi, Y., Yamada, R., Suzuki, A., Harley, J.B., Shirasawa, S., Sawada,
T., Bae, S.C., Tokuhiro, S., Chang, X., Sekine, A. et al. (2005) A
functional variant in FCRL3, encoding Fc receptor-like 3, is associated
with rheumatoid arthritis and several autoimmunities. Nat. Genet.,37,
478485.
8. Simmonds, M.J., Yesmin, K., Newby, P.R., Brand, O.J., Franklyn, J.A.
and Gough, S.C. (2010) Confirmation of association of chromosome
5q3133 with United Kingdom Caucasian Graves’ disease. Thyroid,20,
413417.
9. Song, H.D., Liang, J., Shi, J.Y., Zhao, S.X., Liu, Z., Zhao, J.J., Peng, Y.D.,
Gao, G.Q., Tao, J., Pan, C.M. et al. (2009) Functional SNPs in the
SCGB3A2 promoter are associated with susceptibility to Graves’ disease.
Hum. Mol. Genet.,18, 1156– 1170.
10. Brand, O.J., Lowe, C.E., Heward, J.M., Franklyn, J.A., Cooper, J.D.,
Todd, J.A. and Gough, S.C. (2007) Association of the interleukin-2
receptor alpha (IL-2Ralpha)/CD25 gene region with Graves’ disease using
a multilocus test and tag SNPs. Clin. Endocrinol. (Oxf.),66, 508512.
11. Chistiakov, D.A., Chistiakova, E.I., Voronova, N.V., Turakulov, R.I. and
Savost’anov, K.V. (2011) A variant of the Il2ra/Cd25 gene predisposing
to graves’ disease is associated with increased levels of soluble
interleukin-2 receptor. Scand. J. Immunol.,74, 496– 501.
12. Hong, S.W., Kim, S. and Lee, D.K. (2008) The role of Bach2 in nucleic
acid-triggered antiviral innate immune responses. Biochem. Biophys. Res.
Commun.,365, 426– 432.
13. Barrett, J.C., Clayton, D.G., Concannon, P., Akolkar, B., Cooper, J.D.,
Erlich, H.A., Julier, C., Morahan, G., Nerup, J., Nierras, C. et al. (2009)
Human Molecular Genetics, 2012, Vol. 21, No. 23 5207
Genome-wide association study and meta-analysis find that over 40 loci
affect risk of type 1 diabetes. Nat. Genet.,41, 703707.
14. Sawcer, S., Hellenthal, G., Pirinen, M., Spencer, C.C., Patsopoulos, N.A.,
Moutsianas, L., Dilthey, A., Su, Z., Freeman, C., Hunt, S.E. et al. (2011)
Genetic risk and a primary role for cell-mediated immune mechanisms in
multiple sclerosis. Nature,476, 214– 219.
15. Brand, O.J. and Gough, S.C. (2010) Genetics of thyroid autoimmunity and
the role of the TSHR. Mol. Cell Endocrinol.,322, 135– 143.
16. Heinig, M., Petretto, E., Wallace, C., Bottolo, L., Rotival, M., Lu, H., Li,
Y., Sarwar, R., Langley, S.R., Bauerfeind, A. et al. (2010) A trans-acting
locus regulates an anti-viral expression network and type 1 diabetes risk.
Nature,467, 460– 464.
17. Boelaert, K., Newby, P.R., Simmonds, M.J., Holder, R.L., Carr-Smith,
J.D., Heward, J.M., Manji, N., Allahabadia, A., Armitage, M., Chatterjee,
K.V. et al. (2010) Prevalence and relative risk of other autoimmune
diseases in subjects with autoimmune thyroid disease. Am. J. Med.,
123, 183, e181189.
18. Manji, N., Carr-Smith, J.D., Boelaert, K., Allahabadia, A., Armitage, M.,
Chatterjee, V.K., Lazarus, J.H., Pearce, S.H., Vaidya, B., Gough, S.C.
et al. (2006) Influences of age, gender, smoking, and family history
on autoimmune thyroid disease phenotype. J. Clin. Endocrinol. Metab.,
91, 4873– 4880.
19. 2007) Genome-wide association study of 14,000 cases of seven common
diseases and 3,000 shared controls. Nature,447, 661 678.
20. Cortes, A. and Brown, M.A. (2011) Promise and pitfalls of the
Immunochip. Arthritis Res. Ther.,13, 101.
21. Cooper, J.D., Smyth, D.J., Smiles, A.M., Plagnol, V., Walker, N.M.,
Allen, J.E., Downes, K., Barrett, J.C., Healy, B.C., Mychaleckyj, J.C.
et al. (2008) Meta-analysis of genome-wide association study data
identifies additional type 1 diabetes risk loci. Nat. Genet.,40, 1399 1401.
22. Anderson, C.A., Pettersson, F.H., Clarke, G.M., Cardon, L.R., Morris,
A.P. and Zondervan, K.T. (2010) Data quality control in genetic
case-control association studies. Nat. Protoc.,5, 1564– 1573.
23. Clayton, D.G., Walker, N.M., Smyth, D.J., Pask, R., Cooper, J.D., Maier,
L.M., Smink, L.J., Lam, A.C., Ovington, N.R., Stevens, H.E. et al. (2005)
Population structure, differential bias and genomic control in a
large-scale, casecontrol association study. Nat. Genet.,37, 12431246.
24. Freedman, M.L., Reich, D., Penney, K.L., McDonald, G.J., Mignault,
A.A., Patterson, N., Gabriel, S.B., Topol, E.J., Smoller, J.W., Pato, C.N.
et al. (2004) Assessing the impact of population stratification on genetic
association studies. Nat. Genet.,36, 388– 393.
25. Clayton, D.G. (2009) Sex chromosomes and genetic association studies.
Genome Med.,1, 110.
26. Dubois, P.C., Trynka, G., Franke, L., Hunt, K.A., Romanos, J., Curtotti,
A., Zhernakova, A., Heap, G.A., Adany, R., Aromaa, A. et al. (2010)
Multiple common variants for celiac disease influencing immune gene
expression. Nat. Genet.,42, 295– 302.
27. Hirschfield, G.M., Liu, X., Han, Y., Gorlov, I.P., Lu, Y., Xu, C., Chen, W.,
Juran, B.D., Coltescu, C., Mason, A.L. et al. (2010) Variants at
IRF5-TNPO3, 17q12– 21 and MMEL1 are associated with primary biliary
cirrhosis. Nat. Genet.,42, 655– 657.
28. Kurreeman, F.A., Stahl, E.A., Okada, Y., Liao, K., Diogo, D.,
Raychaudhuri, S., Freudenberg, J., Kochi, Y., Patsopoulos, N.A., Gupta,
N. et al. (2012) Use of a multiethnic approach to identify rheumatoid-
arthritis-susceptibility loci, 1p36 and 17q12. Am. J. Hum. Genet.,90,
524532.
29. Franke, A., McGovern, D.P., Barrett, J.C., Wang, K., Radford-Smith,
G.L., Ahmad, T., Lees, C.W., Balschun, T., Lee, J., Roberts, R. et al.
(2010) Genome-wide meta-analysis increases to 71 the number
of confirmed Crohn’s disease susceptibility loci. Nat. Genet.,42,
1118– 1125.
30. Barrett, J.C., Hansoul, S., Nicolae, D.L., Cho, J.H., Duerr, R.H., Rioux,
J.D., Brant, S.R., Silverberg, M.S., Taylor, K.D., Barmada, M.M. et al.
(2008) Genome-wide association defines more than 30 distinct
susceptibility loci for Crohn’s disease. Nat. Genet.,40, 955– 962.
31. Stahl, E.A., Raychaudhuri, S., Remmers, E.F., Xie, G., Eyre, S., Thomson,
B.P., Li, Y., Kurreeman, F.A., Zhernakova, A., Hinks, A. et al. (2010)
Genome-wide association study meta-analysis identifies seven new
rheumatoid arthritis risk loci. Nat. Genet.,42, 508– 514.
32. Quan, C., Ren, Y.Q., Xiang, L.H., Sun, L.D., Xu, A.E., Gao, X.H., Chen,
H.D., Pu, X.M., Wu, R.N., Liang, C.Z. et al. (2010) Genome-wide
association study for vitiligo identifies susceptibility loci at 6q27 and the
MHC. Nat. Genet.,42, 614– 618.
33. Nath, S.K., Han, S., Kim-Howard, X., Kelly, J.A., Viswanathan, P.,
Gilkeson, G.S., Chen, W., Zhu, C., McEver, R.P., Kimberly, R.P. et al.
(2008) A nonsynonymous functional variant in integrin-alpha(M)
(encoded by ITGAM) is associated with systemic lupus erythematosus.
Nat. Genet.,40, 152– 154.
5208 Human Molecular Genetics, 2012, Vol. 21, No. 23

Supplementary resource (1)

... 22 For Graves' disease and Hashimoto's thyroiditis, GWAS had been performed using participants with European ancestry but included only 2,285 and 462 cases, respectively. 23 As these GWAS had limited power to detect meaningful associations for these diseases, we used GWAS summary statistics from meta-analyses of two European and one East Asian populations. 24 For Graves' disease, 1,678 cases from UKBB and FinnGen (release 3) were meta-analysed with 2,809 cases from Biobank Japan; for Hashimoto's thyroiditis, 15,654 cases from UK Biobank and FinnGen were meta-analysed with 537 cases from Biobank Japan. ...
Preprint
Full-text available
Background: Thyroid dysfunction, hypothyroidism in particular, has been associated with endometrial cancer, but it remains unclear whether hypothyroidism itself or other aspects of thyroid dysfunction have a causal effect on endometrial cancer risk. Methods: To clarify the effects of thyroid dysfunction phenotypes on endometrial cancer risk, we performed Mendelian randomisation analyses data from the largest available genome-wide association studies (GWAS). The robustness of associations was assessed through sensitivity analyses. To disentangle the potential influence of obesity on causal associations, we carried out multivariable Mendelian randomisation analysis. Results: Mendelian randomisation analysis demonstrated a significant causal association between hypothyroidism and decreased risk of endometrial cancer (OR = 0.93; 95% CI 0.89-0.97; p = 3.96 x 10-4). Hashimotos thyroiditis, a common cause of hypothyroidism, showed a similar, albeit nominal, association with endometrial cancer risk (OR = 0.92; 95% CI 0.86-0.99; p = 0.03). Hypothyroidism was also significantly associated with decreased risk of endometrioid endometrial cancer (OR = 0.93; 95% CI 0.88-0.98; p = 4.02 x 10-3), the most common histological subtype. Sensitivity analyses confirmed the robustness of the significant associations. Multivariable Mendelian randomisation analysis revealed that BMI and hypothyroidism had independent effects on endometrial cancer risk. Interpretation: This study provides evidence for a causal relationship between hypothyroidism and decreased risk of endometrial cancer. The protective effect of hypothyroidism is independent of BMI and may be related to the autoimmune effects of Hashimotos disease. Funding: National Health and Research Council of Australia (APP1173170). Worldwide Cancer Research and Cancer Australia (22-0253).
... These results contrast with other discordant findings, where no associations between IL-2Rα SNPs and the presence of AITD were found [147][148][149][150][151]. ...
Article
Full-text available
The most common cause of acquired thyroid dysfunction is autoimmune thyroid disease, which is an organ-specific autoimmune disease with two presentation phenotypes: hyperthyroidism (Graves-Basedow disease) and hypothyroidism (Hashimoto’s thyroiditis). Hashimoto’s thyroiditis is distinguished by the presence of autoantibodies against thyroid peroxidase and thyroglobulin. Meanwhile, autoantibodies against the TSH receptor have been found in Graves-Basedow disease. Numerous susceptibility genes, as well as epigenetic and environmental factors, contribute to the pathogenesis of both diseases. This review summarizes the most common genetic, epigenetic, and environmental mechanisms involved in autoimmune thyroid disease.
... Polymorphism of cytotoxic T-lymphocyte-associated protein 4 (CTLA4) was related to an increased risk of 1.3-1.5 of developing HT. GWAS revealed several susceptibility loci shared with other autoimmune disorders, such as the LIM domain containing preferred translocation partner in lipoma (LPP), BTB domain and CNC homolog 2 (BACH2), and MAGI3, which is associated with the progression to hypothyroidism [64,65]. Finally, HT severity was associated with TGF-β polymorphisms. ...
Article
Full-text available
Vitamin D is a secosteroid hormone that is highly involved in bone health. Mounting evidence revealed that, in addition to the regulation of mineral metabolism, vitamin D is implicated in cell proliferation and differentiation, vascular and muscular functions, and metabolic health. Since the discovery of vitamin D receptors in T cells, local production of active vitamin D was demonstrated in most immune cells, addressing the interest in the clinical implications of vitamin D status in immune surveillance against infections and autoimmune/inflammatory diseases. T cells, together with B cells, are seen as the main immune cells involved in autoimmune diseases; however, growing interest is currently focused on immune cells of the innate compartment, such as monocytes, macrophages, dendritic cells, and natural killer cells in the initiation phases of autoimmunity. Here we reviewed recent advances in the onset and regulation of Graves’ and Hashimoto’s thyroiditis, vitiligo, and multiple sclerosis in relation to the role of innate immune cells and their crosstalk with vitamin D and acquired immune cells.
... Patients with autoimmune thyroid disease can have other autoimmune diseases at the same time or successively, such as myasthenia gravis, type 1 diabetes, pernicious anemia, atrophic gastritis, etc. BACH2 is a protein coding gene, BACH2 affects the diseases of immunodeficiency, chronic myeloid leukemia and etc. Research indicated that the polymorphism of the BACH2 gene has close connection with the susceptibility to autoimmune thyroid disease. Individuals carrying the A allele at the key locus of this gene have a higher risk of autoimmune thyroid disease [17]. • Risk of common infectious diseases, contains the DNA testing items of: epidemic parotitis, streptococcal pharyngitis, and Herpes labialis. ...
Article
Full-text available
Immunity refers to the ability of the human immune system to resist pathogen infection. Immune system has the basic functions of immune defense, immune self stabilization and immune surveillance. Balanced nutrition is the cornerstone of the immune system to play its immune function, and nutritional intervention is also an important means to maintain and improve immunity. Previous studies have confirmed that T cells have individual differences in recognizing viral antigens of virus infected cells, and the body’s response to antigens is controlled by a variety of genetic genes, such as human leukocyte antigen (HLA) genes, immune response (Ir) genes, etc. In this paper, through immunity genetic testing, we screen out genetically susceptible people with low immunity and people with the risk of nutrient metabolism disorders; through using lifestyle questionnaire and physical examination results, we analyze people’s physical condition, dietary habits, and exercise habits to evaluate people’s nutrient deficiency degree. Then, combining multi-dimensional health data, we evaluate users’ immune status and nutritional deficiency risk comprehensively, further, we implemented personalized nutrition intervention on the types and doses of nutritional supplements to improve immunity. We also validated the effectiveness of our personalized nutrition solution through a population-based cohort study.
... 17,18 LPP is expressed in a variety of tissues including the adrenals and variants have been associated with a diverse range of conditions, including the autoimmune conditions coeliac disease and Graves' disease. [19][20][21] The SNP rs4686484 has been reported as a functional variant in coeliac disease by Almeida et al., who also found reduced LPP expression in duodenal biopsies from patients with severe coeliac disease. 15 In our study, the risk allele for rs4686484 was significantly overrepresented in among AAD cases relative to controls (OR of 1.46, P = 5.4 × 10 −5 ). ...
Article
Full-text available
Autoimmune Addison's disease (AAD) arises from a complex interplay between multiple genetic susceptibility polymorphisms and environmental factors. The first genome wide association study (GWAS) with patients from Scandinavian Addison's registries has identified association signals at four novel loci in the genes LPP, SH2B3, SIGLEC5, and UBASH3A. To verify these novel risk loci, we performed a case–control association study in our independent cohort of 420 patients with AAD from the across the UK. We report significant association of alleles of the LPP and UBASH3A genes [odds ratio (95% confidence intervals), 1.46 (1.21-1.75)and 1.40 (1.16-1.68), respectively] with AAD in our UK cohort. In addition, we report nominal association of AAD with SH2B3 [OR 1.18 (1.02-1.35)]. We confirm that variants at the LPP and UBASH3A loci confer susceptibility to AAD in a UK population. Further studies with larger patient cohorts are required to robustly confirm the association of SH2B3 and SIGLEC5/SPACA6 alleles.
... The genetics of AD is complicated by genetic pleiotropy, which describes a single gene or variant affecting multiple traits 31 . The A allele of rs2476601, mapping to PTPN22 has been identified as a susceptibility allele in genome-wide studies of multiple ADs 43,44 . Other variants have been associated with multiple ADs at genome significance but with opposite effects 45 . ...
Article
Full-text available
Immune checkpoint inhibitor (ICI) therapy has revolutionised the treatment of various cancer types. ICIs reinstate T-cell function to elicit an anti-cancer immune response. The resulting immune response can however have off-target effects which manifest as autoimmune type serious immune-related adverse events (irAE) in ~10–55% of patients treated. It is currently challenging to predict both who will experience irAEs and to what severity. Identification of patients at high risk of serious irAE would revolutionise patient care. While the pathogenesis driving irAE development is still unclear, host genetic factors are proposed to be key determinants of these events. This review presents current evidence supporting the role of the host genome in determining risk of irAE. We summarise the spectrum and timing of irAEs following treatment with ICIs and describe currently reported germline genetic variation associated with expression of immuno-modulatory factors within the cancer immunity cycle, development of autoimmune disease and irAE occurrence. We propose that germline genetic determinants of host immune function and autoimmune diseases could also explain risk of irAE development. We also endorse genome-wide association studies of patients being treated with ICIs to identify genetic variants that can be used in polygenic risk scores to predict risk of irAE.
Preprint
Full-text available
Autoimmune and inflammatory diseases are a group of > 80 complex diseases caused by loss of immune tolerance for self-antigens. The biological mechanisms of autoimmune diseases are largely unknown, preventing the development of effective treatment options. Integrative analysis of genome-wide association studies and chromatin accessibility data has shown that the risk variants of autoimmune diseases are enriched in open chromatin regions of immune cells, supporting their role in gene regulation. However, we still lack a systematic and unbiased identification of transcription factors involved in disease gene regulation. We hypothesized that for some of the disease-relevant transcription factors, their binding to DNA is affected at multiple genomic sites rather than a single site, and these effects are cell-type specific. We developed a statistical approach to assess enrichment of transcription factors in being affected by disease risk variants at multiple genomic sites. We used genetic association data of nine autoimmune diseases and identified 99% credible set SNPs for each trait. We then integrated the credible SNPs and chromatin accessibility data of 376 samples comprising 35 unique cell types, and employed a probabilistic model to identify the SNPs that are likely to change binding probability of certain transcription factors at specific cell types. Finally, for each transcription factor, we used a statistical test to assess whether the credible SNPs show enrichments in terms of changing the binding probability of that transcription factor at multiple sites. Our analysis resulted in identification of significantly enriched transcription factors and their relevant cell types for each trait. The prioritized immune cell types varied across the diseases. Our analysis proved that our predicted regulatory sites are active enhancers or promoters in the relevant cell types. Additionally, our pathway analysis showed that the majority of the significant biological pathways are immune-related. In summary, our study has identified disease-relevant transcription factors and their relevant cell types, and will facilitate discovering specific gene regulatory mechanisms of autoimmune diseases.
Article
Full-text available
Graves’ disease (GD) is the commonest cause of hyperthyroidism and has a strong female preponderance. Everyday clinical practice suggests strong aggregation within families and twin studies demonstrate that genetic factors account for 60-80% of risk of developing GD. In this review, we collate numerous genetic studies and outline the discoveries over the years, starting with historic candidate gene studies and then exploring more recent genome-wide linkage and association studies, which have involved substantial cohorts of East Asian patients as well as those of European descent. Variants in genes including HLA , CTLA4 , and PTPN22 have been shown to have substantial individual effects on disease susceptibility. In addition, we examine emerging evidence concerning the possibility that genetic variants may correlate with relevant clinical phenotypes including age of onset of GD, severity of thyrotoxicosis, goitre size and relapse of hyperthyroidism following antithyroid drug therapy, as well as thyroid eye disease. This review supports the inheritance of GD as a complex genetic trait, with a growing number of more than 80 susceptibility loci identified so far. Future implementation of more targeted clinical therapies requires larger studies investigating the influence of these genetic variants on the various phenotypes and different outcomes of conventional treatments.
Article
Autoimmune diseases (ADs) are common conditions in which an individual's immune system reacts against its healthy cells. This condition is a common cause of morbidity and mortality, with an estimated prevalence ranging from 5 per 100,000 to more than 500 per 100,000. According to the National Stem Cell Foundation (NSCF), ADs are prevalent in about 4% of the world's population, which creates a burden on society due to the high treatment cost. ADs show a clear gender bias with a higher prevalence among women, occurring at a rate of 2:1 female-to-male ratio. The etiology of ADs includes genetic and environmental factors. ADs are more likely to develop in genetically susceptible individuals. The higher concordance ratio between monozygotic twins compared to dizygotic twins or other siblings validates the role of genetic factors in the pathogenesis of many ADs. ADs diagnosis includes conventional immunoassay such as indirect immunofluorescence, complement fixation, passive agglutination, autoantibodies detection, and most recent advances, including multiplex platforms such as microspots, line-blot, addressable microbeads and barcoded nanoparticles that allow multiplex parallel testing of autoantibodies. ADs treatment includes biological and synthetic drugs that block many pathways and components of the immune system, including Janus kinase (JAK) inhibitors, non-receptor tyrosine-protein kinase (TYK2), and other cytokines. Generally, recent immune-modulatory drugs used in ADs treatment are non-disease specific with broad action and are associated with many side effects like infection and malignant diseases. Furthermore, gene therapy seeks to control the levels of proinflammatory cytokine molecules and lymphocyte infiltration through the delivery and expression of therapeutic genes. Recent genomic-wide association studies (GWAS) have allowed the identification of various genetic loci associated with disease susceptibility and have revealed candidate genes that can be used in targeted therapeutics. This review summarizes recent literature on the genetic factors associated with susceptibility to the 11 most common ADs, namely: Type 1 diabetes mellitus (T1DM), Multiple sclerosis (MS), Grave's disease, Sjögren's syndrome (SS), Celiac disease, Hashimoto's thyroiditis (HT), Anti-phospholipid syndrome (APS), Autoimmune hemolytic anemia, Rheumatoid arthritis (RA), Systemic lupus erythematosus (SLE), and Scleroderma (systemic sclerosis).
Article
Full-text available
We undertook a meta-analysis of six Crohn's disease genome-wide association studies (GWAS) comprising 6,333 affected individuals (cases) and 15,056 controls and followed up the top association signals in 15,694 cases, 14,026 controls and 414 parent-offspring trios. We identified 30 new susceptibility loci meeting genome-wide significance (P < 5 × 10⁻⁸). A series of in silico analyses highlighted particular genes within these loci and, together with manual curation, implicated functionally interesting candidate genes including SMAD3, ERAP2, IL10, IL2RA, TYK2, FUT2, DNMT3A, DENND1B, BACH2 and TAGAP. Combined with previously confirmed loci, these results identify 71 distinct loci with genome-wide significant evidence for association with Crohn's disease.
Article
Full-text available
There is increasing evidence that genome-wide association (GWA) studies represent a powerful approach to the identification of genes involved in common human diseases. We describe a joint GWA study (using the Affymetrix GeneChip 500K Mapping Array Set) undertaken in the British population, which has examined 2,000 individuals for each of 7 major diseases and a shared set of 3,000 controls. Case-control comparisons identified 24 independent association signals at P < 5 10-7: 1 in bipolar disorder, 1 in coronary artery disease, 9 in Crohn's disease, 3 in rheumatoid arthritis, 7 in type 1 diabetes and 3 in type 2 diabetes. On the basis of prior findings and replication studies thus-far completed, almost all of these signals reflect genuine susceptibility effects. We observed association at many previously identified loci, and found compelling evidence that some loci confer risk for more than one of the diseases studied. Across all diseases, we identified a large number of further signals (including 58 loci with single-point P values between 10-5 and 5 10-7) likely to yield additional susceptibility loci. The importance of appropriately large samples was confirmed by the modest effect sizes observed at most loci identified. This study thus represents a thorough validation of the GWA approach. It has also demonstrated that careful use of a shared control group represents a safe and effective approach to GWA analyses of multiple disease phenotypes; has generated a genome-wide genotype database for future studies of common diseases in the British population; and shown that, provided individuals with non-European ancestry are excluded, the extent of population stratification in the British population is generally modest. Our findings offer new avenues for exploring the pathophysiology of these important disorders. We anticipate that our data, results and software, which will be widely available to other investigators, will provide a powerful resource for human genetics research.
Article
Full-text available
Using variants from the 1000 Genomes Project pilot European CEU dataset and data from additional resequencing studies, we densely genotyped 183 non-HLA risk loci previously associated with immune-mediated diseases in 12,041 individuals with celiac disease (cases) and 12,228 controls. We identified 13 new celiac disease risk loci reaching genome-wide significance, bringing the number of known loci (including the HLA locus) to 40. We found multiple independent association signals at over one-third of these loci, a finding that is attributable to a combination of common, low-frequency and rare genetic variants. Compared to previously available data such as those from HapMap3, our dense genotyping in a large sample collection provided a higher resolution of the pattern of linkage disequilibrium and suggested localization of many signals to finer scale regions. In particular, 29 of the 54 fine-mapped signals seemed to be localized to single genes and, in some instances, to gene regulatory elements. Altogether, we define the complex genetic architecture of the risk regions of and refine the risk signals for celiac disease, providing the next step toward uncovering the causal mechanisms of the disease.
Article
Full-text available
Graves' disease is a common autoimmune disorder characterized by thyroid stimulating hormone receptor autoantibodies (TRAb) and hyperthyroidism. To investigate the genetic architecture of Graves' disease, we conducted a genome-wide association study in 1,536 individuals with Graves' disease (cases) and 1,516 controls. We further evaluated a group of associated SNPs in a second set of 3,994 cases and 3,510 controls. We confirmed four previously reported loci (in the major histocompatibility complex, TSHR, CTLA4 and FCRL3) and identified two new susceptibility loci (the RNASET2-FGFR1OP-CCR6 region at 6q27 (P(combined) = 6.85 × 10(-10) for rs9355610) and an intergenic region at 4p14 (P(combined) = 1.08 × 10(-13) for rs6832151)). These newly associated SNPs were correlated with the expression levels of RNASET2 at 6q27, of CHRNA9 and of a previously uncharacterized gene at 4p14, respectively. Moreover, we identified strong associations of TSHR and major histocompatibility complex class II variants with persistently TRAb-positive Graves' disease.
Article
Full-text available
Multiple sclerosis is a common disease of the central nervous system in which the interplay between inflammatory and neurodegenerative processes typically results in intermittent neurological disturbance followed by progressive accumulation of disability. Epidemiological studies have shown that genetic factors are primarily responsible for the substantially increased frequency of the disease seen in the relatives of affected individuals, and systematic attempts to identify linkage in multiplex families have confirmed that variation within the major histocompatibility complex (MHC) exerts the greatest individual effect on risk. Modestly powered genome-wide association studies (GWAS) have enabled more than 20 additional risk loci to be identified and have shown that multiple variants exerting modest individual effects have a key role in disease susceptibility. Most of the genetic architecture underlying susceptibility to the disease remains to be defined and is anticipated to require the analysis of sample sizes that are beyond the numbers currently available to individual research groups. In a collaborative GWAS involving 9,772 cases of European descent collected by 23 research groups working in 15 different countries, we have replicated almost all of the previously suggested associations and identified at least a further 29 novel susceptibility loci. Within the MHC we have refined the identity of the HLA-DRB1 risk alleles and confirmed that variation in the HLA-A gene underlies the independent protective effect attributable to the class I region. Immunologically relevant genes are significantly overrepresented among those mapping close to the identified loci and particularly implicate T-helper-cell differentiation in the pathogenesis of multiple sclerosis.
Article
Full-text available
In this study, we aimed to determine the relationship between flow-mediated endothelium-dependent vasodilatation (FMD) and carotid artery intima-media wall thickness (IMT), two surrogate markers of atherosclerosis, in a series of Spanish patients with rheumatoid arthritis (RA) without clinically evident cardiovascular (CV) disease. One hundred eighteen patients who fulfilled the 1987 American College of Rheumatology classification criteria for RA, had no history of CV disease and had at least one year of follow-up after disease diagnosis were randomly selected. Brachial and carotid ultrasonography were performed to determine FMD and carotid IMT, respectively. Carotid IMT values were higher and FMD percentages derived by performing ultrasonography were lower in individuals with a long duration from the time of disease diagnosis. Patients with a disease duration ≤ 7 years had significantly lower carotid IMT (mean ± SD) 0.69 ± 0.17 mm than those with long disease duration (0.81 ± 0.12 mm in patients with ≥ 20 years of follow-up). Also, patients with a long disease duration had severe endothelial dysfunction (FMD 4.0 ± 4.0% in patients with disease duration from 14.5 to 19.7 years) compared with those with shorter disease duration (FMD 7.4 ± 3.8% in patients with disease duration ≤ 7 years). Linear regression analysis revealed that carotid IMT was unrelated to FMD in the whole sample of 118 patients. However, carotid IMT was negatively associated with FMD when the time from disease diagnosis ranged from 7.5 to 19.7 years (P = 0.02). In patients with RA without CV disease, endothelial dysfunction and carotid IMT increased with the duration of RA. The association between FMD and carotid IMT values was observed only in patients with long disease duration.
Article
Multiple sclerosis is a common disease of the central nervous system in which the interplay between inflammatory and neurodegenerative processes typically results in intermittent neurological disturbance followed by progressive accumulation of disability. Epidemiological studies have shown that genetic factors are primarily responsible for the substantially increased frequency of the disease seen in the relatives of affected individuals, and systematic attempts to identify linkage in multiplex families have confirmed that variation within the major histocompatibility complex (MHC) exerts the greatest individual effect on risk. Modestly powered genome-wide association studies (GWAS) have enabled more than 20 additional risk loci to be identified and have shown that multiple variants exerting modest individual effects have a key role in disease susceptibility. Most of the genetic architecture underlying susceptibility to the disease remains to be defined and is anticipated to require the analysis of sample sizes that are beyond the numbers currently available to individual research groups. In a collaborative GWAS involving 9,772 cases of European descent collected by 23 research groups working in 15 different countries, we have replicated almost all of the previously suggested associations and identified at least a further 29 novel susceptibility loci. Within the MHC we have refined the identity of the HLA-DRB1 risk alleles and confirmed that variation in the HLA-A gene underlies the independent protective effect attributable to the class I region. Immunologically relevant genes are significantly overrepresented among those mapping close to the identified loci and particularly implicate T-helper-cell differentiation in the pathogenesis of multiple sclerosis.
Article
We have previously shown that rheumatoid arthritis (RA) risk alleles overlap between different ethnic groups. Here, we utilize a multiethnic approach to show that we can effectively discover RA risk alleles. Thirteen putatively associated SNPs that had not yet exceeded genome-wide significance (p < 5 × 10(-8)) in our previous RA genome-wide association study (GWAS) were analyzed in independent sample sets consisting of 4,366 cases and 17,765 controls of European, African American, and East Asian ancestry. Additionally, we conducted an overall association test across all 65,833 samples (a GWAS meta-analysis plus the replication samples). Of the 13 SNPs investigated, four were significantly below the study-wide Bonferroni corrected p value threshold (p < 0.0038) in the replication samples. Two SNPs (rs3890745 at the 1p36 locus [p = 2.3 × 10(-12)] and rs2872507 at the 17q12 locus [p = 1.7 × 10(-9)]) surpassed genome-wide significance in all 16,659 RA cases and 49,174 controls combined. We used available GWAS data to fine map these two loci in Europeans and East Asians, and we found that the same allele conferred risk in both ethnic groups. A series of bioinformatic analyses identified TNFRSF14-MMEL1 at the 1p36 locus and IKZF3-ORMDL3-GSDMB at the 17q12 locus as the genes most likely associated with RA. These findings demonstrate empirically that a multiethnic approach is an effective strategy for discovering RA risk loci, and they suggest that combining GWASs across ethnic groups represents an efficient strategy for gaining statistical power.
Article
Alpha-subunit of the IL-2 receptor (IL-2Rα) encoded by the IL2RA/CD25 gene binds IL-2 that plays a pivotal role in the regulation of T cell function. Levels of a soluble form of IL-2Rα (sIL-2Rα) lacking the transmembrane and cytoplasmic domains were shown to be increased in several autoimmune diseases including Graves' disease (GD). Recent studies showed association between the IL2RA/CD25 gene variants and several autoimmune diseases including GD. In this study, we analyzed whether polymorphic markers rs2104286, rs41295061, and rs11594656 located at the IL2RA/CD25 locus confer susceptibility to GD and are related to increased concentrations of sIL-2Rα. A total of 1474 Russian GD patients and 1609 control subjects were genotyped for rs2104286, rs41295061, and rs11594656 using a Taqman assay. Concentrations of sIL-2Rα in sera of affected and non-affected individuals were measured using an ELISA test. A minor allele A of rs41295061 showed significant association with increased risk of GD [odds ratio (OR) = 1.43, P(c)  = 0.00102]. The allele A of rs41295061 and allele A of rs11594656 constitute a higher risk haplotype AA (OR = 1.47, P(c)  = 0.0477). Compared to carriers of the protective haplogenotype GT/GT, the carriage of two copies of the haplogenotype AA/AA was associated with elevated levels of sIL-2Rα in both GD patients (AA/AA versus GT/GT: 1.35 ± 0.47 ng/ml versus 1.12 ± 0.45 ng/ml, P = 0.0065) and healthy controls (AA/AA versus GT/GT: 0.67 ± 0.28 ng/ml versus 0.51 ± 0.33 ng/ml, P = 0.0098). This is the first report presenting correlation between the carriage of disease-associated variants of IL2RA/CD25 with increased levels of sIL-2Rα in GD.