Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 May 1.
Published in final edited form as: Nat Genet. 2013 Sep 29;45(11):1353–1360. doi: 10.1038/ng.2770

Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis

International Multiple Sclerosis Genetics Consortium (IMSGC), Ashley H Beecham 1,126, Nikolaos A Patsopoulos 2,3,4,5,6,126, Dionysia K Xifara 7, Mary F Davis 8, Anu Kemppinen 9, Chris Cotsapas 10,11,12, Tejas S Shahi 13, Chris Spencer 7, David Booth 14, An Goris 15, Annette Oturai 16, Janna Saarela 17, Bertrand Fontaine 18, Bernhard Hemmer 19,20,21, Claes Martin 22, Frauke Zipp 23, Sandra D’alfonso 24, Filippo Martinelli-Boneschi 25,26, Bruce Taylor 27, Hanne F Harbo 28,29, Ingrid Kockum 30, Jan Hillert 30, Tomas Olsson 30, Maria Ban 9, Jorge R Oksenberg 31, Rogier Hintzen 32, Lisa F Barcellos 33,34,35; Wellcome Trust Case Control Consortium 2 (WTCCC2)36; International IBD Genetics Consortium (IIBDGC)36, Cristina Agliardi 37, Lars Alfredsson 38, Mehdi Alizadeh 39, Carl Anderson 13, Robert Andrews 13, Helle Bach Søndergaard 16, Amie Baker 9, Gavin Band 7, Sergio E Baranzini 31, Nadia Barizzone 24, Jeffrey Barrett 13, Céline Bellenguez 7, Laura Bergamaschi 24, Luisa Bernardinelli 40, Achim Berthele 19, Viola Biberacher 19, Thomas M C Binder 41, Hannah Blackburn 13, Izaura L Bomfim 30, Paola Brambilla 25, Simon Broadley 42, Bruno Brochet 43, Lou Brundin 30, Dorothea Buck 19, Helmut Butzkueven 44,45, Stacy J Caillier 31, William Camu 46, Wassila Carpentier 47, Paola Cavalla 48,49, Elisabeth G Celius 28, Irène Coman 50, Giancarlo Comi 25,26, Lucia Corrado 24, Leentje Cosemans 15, Isabelle Cournu-Rebeix 18, Bruce A C Cree 31, Daniele Cusi 51, Vincent Damotte 18, Gilles Defer 52, Silvia R Delgado 53, Panos Deloukas 13, Alessia di Sapio 54, Alexander T Dilthey 7, Peter Donnelly 7, Bénédicte Dubois 15, Martin Duddy 55, Sarah Edkins 13, Irina Elovaara 56, Federica Esposito 25,26, Nikos Evangelou 57, Barnaby Fiddes 9, Judith Field 58, Andre Franke 59, Colin Freeman 7, Irene Y Frohlich 2, Daniela Galimberti 60,61, Christian Gieger 62, Pierre-Antoine Gourraud 31, Christiane Graetz 23, Andrew Graham 63, Verena Grummel 19, Clara Guaschino 25,26, Athena Hadjixenofontos 1, Hakon Hakonarson 64,65, Christopher Halfpenny 66, Gillian Hall 67, Per Hall 68, Anders Hamsten 69, James Harley 70, Timothy Harrower 71, Clive Hawkins 72, Garrett Hellenthal 73, Charles Hillier 74, Jeremy Hobart 75, Muni Hoshi 19, Sarah E Hunt 13, Maja Jagodic 30, Ilijas Jelčić 76,77, Angela Jochim 19, Brian Kendall 78, Allan Kermode 79,80, Trevor Kilpatrick 81, Keijo Koivisto 82, Ioanna Konidari 1, Thomas Korn 19, Helena Kronsbein 19, Cordelia Langford 13, Malin Larsson 83, Mark Lathrop 84,85,86, Christine Lebrun-Frenay 87, Jeannette Lechner-Scott 88, Michelle H Lee 2, Maurizio A Leone 89, Virpi Leppä 17, Giuseppe Liberatore 25,26, Benedicte A Lie 29,90, Christina M Lill 23,91, Magdalena Lindén 30, Jenny Link 30, Felix Luessi 23, Jan Lycke 92, Fabio Macciardi 93,94, Satu Männistö 95, Clara P Manrique 1, Roland Martin 76,77, Vittorio Martinelli 26, Deborah Mason 96, Gordon Mazibrada 97, Cristin McCabe 10, Inger-Lise Mero 28,29,90, Julia Mescheriakova 32, Loukas Moutsianas 7, Kjell-Morten Myhr 98, Guy Nagels 99, Richard Nicholas 100, Petra Nilsson 101, Fredrik Piehl 30, Matti Pirinen 7, Siân E Price 102, Hong Quach 33, Mauri Reunanen 103,104, Wim Robberecht 105,106,107, Neil P Robertson 108, Mariaemma Rodegher 26, David Rog 109, Marco Salvetti 110, Nathalie C Schnetz-Boutaud 8, Finn Sellebjerg 16, Rebecca C Selter 19, Catherine Schaefer 35, Sandip Shaunak 111, Ling Shen 35, Simon Shields 112, Volker Siffrin 23, Mark Slee 113, Per Soelberg Sorensen 16, Melissa Sorosina 25, Mireia Sospedra 76,77, Anne Spurkland 114, Amy Strange 7, Emilie Sundqvist 30, Vincent Thijs 105,106,107, John Thorpe 115, Anna Ticca 116, Pentti Tienari 117, Cornelia van Duijn 118, Elizabeth M Visser 119, Steve Vucic 14, Helga Westerlind 30, James S Wiley 58, Alastair Wilkins 120, James F Wilson 121, Juliane Winkelmann 19,20,122,123, John Zajicek 75, Eva Zindler 23, Jonathan L Haines 8, Margaret A Pericak-Vance 1, Adrian J Ivinson 124, Graeme Stewart 14, David Hafler 10,11,125, Stephen L Hauser 31, Alastair Compston 9, Gil McVean 7, Philip De Jager 2,5,10,126, Stephen Sawcer 9,126, Jacob L McCauley 1,126
PMCID: PMC3832895  NIHMSID: NIHMS521200  PMID: 24076602

Abstract

Using the ImmunoChip custom genotyping array, we analysed 14,498 multiple sclerosis subjects and 24,091 healthy controls for 161,311 autosomal variants and identified 135 potentially associated regions (p-value < 1.0 × 10-4). In a replication phase, we combined these data with previous genome-wide association study (GWAS) data from an independent 14,802 multiple sclerosis subjects and 26,703 healthy controls. In these 80,094 individuals of European ancestry we identified 48 new susceptibility variants (p-value < 5.0 × 10-8); three found after conditioning on previously identified variants. Thus, there are now 110 established multiple sclerosis risk variants in 103 discrete loci outside of the Major Histocompatibility Complex. With high resolution Bayesian fine-mapping, we identified five regions where one variant accounted for more than 50% of the posterior probability of association. This study enhances the catalogue of multiple sclerosis risk variants and illustrates the value of fine-mapping in the resolution of GWAS signals.


Multiple sclerosis (OMIM 126200) is an inflammatory demyelinating disorder of the central nervous system that is a common cause of chronic neurological disability.1,2 It has its greatest prevalence amongst individuals of Northern European ancestry3 and is moderately heritable,4 with a sibling relative recurrence risk (λs) of ~ 6.3.5 Aside from the early success in demonstrating the important effects exerted by variants in the Human Leukocyte Antigen (HLA) genes from the Major Histocompatibility Complex (MHC),6 there was little progress in unravelling the genetic architecture underlying multiple sclerosis susceptibility prior to the advent of genome-wide association studies (GWAS). Over the last decade, our Consortium has performed several GWAS and meta-analyses in large cohorts, 7-10 cumulatively identifying more than 50 non-MHC susceptibility alleles. As in other complex diseases, available data suggest that many additional susceptibility alleles remain to be identified.11

The striking overlap in the genetic architecture underlying susceptibility to autoimmune diseases9,10,12,13 prompted the collaborative construction of the “ImmunoChip” (see Supplementary Note and Supplementary Figs. 1 and 2 for details of IMSGC nominated content), an efficient genotyping platform designed to deeply interrogate 184 non-MHC loci with genome-wide significant associations to at least one autoimmune disease and provide lighter coverage of other genomic regions with suggestive evidence of association.14 Here, we report a large-scale effort that leverages the ImmunoChip to detect association with multiple sclerosis susceptibility and refine these associations via Bayesian fine-mapping.

After stringent quality control (QC), we report genotypes on 28,487 individuals of European ancestry (14,498 multiple sclerosis subjects, 13,989 healthy controls) that are independent of previous GWAS efforts. We supplemented these data with 10,102 independent control subjects provided by the International Inflammatory Bowel Disease Genetics Consortium (IIBDGC)15 bringing the total to 38,589 individuals (14,498 multiple sclerosis subjects and 24,091 healthy controls). We performed variant level QC, population outlier identification, and subsequent case-control analysis in 11 country-organized strata. To account for within-stratum population stratification we used the first five principal components as covariates in the association analysis. Per stratum odds ratios (OR) and respective standard errors (SE) were then combined with an inverse variance meta-analysis under a fixed effects model. In total we tested 161,311 autosomal variants that passed QC in at least two of the 11 strata (Online Methods). A circos plot16 summarising the results from this discovery phase analysis is shown in Figure 1.

Figure 1. Discovery phase results.

Figure 1

Primary association analysis of 161,311 autosomal variants in the discovery phase (based on 14,498 cases and 24,091 healthy controls). The outer most track shows the numbered autosomal chromosomes. The second track indicates the gene closest to the most associated SNP meeting all replication criteria. Previously identified associations are indicated in grey. The third track indicates the physical position of the 184 fine-mapping intervals (green). The inner most track indicates −log(p) (two-sided) for each SNP (scaled from 0-12 which truncates the signal in several regions, see Supplementary Table 1). Additionally, contour lines are given at the a priori discovery(−log(p) = 4) and genome-wide significance (-log(p) = 7.3) thresholds. Orange indicates -log(p) ≥ 4 and < 7.3, while red indicates −log(p) ≥ 7.3. Details of the full discovery phase results can be found in ImmunoBase.

We defined an a priori discovery threshold of p-value <1 × 10-4 and identified 135 primary statistically independent association signals; 67 in the designated fine-mapping regions and 68 in less densely covered regions selected for deep replication of earlier GWAS. Another 13 secondary and 2 tertiary statistically independent signals were identified by forward stepwise logistic regression. A total of 48 of the 150 statistically independent association signals (Supplementary Table 1) reached a genome-wide significance p-value <5 × 10-8 at the discovery phase alone. Next, we replicated our findings in 14,802 multiple sclerosis subjects and 26,703 healthy controls with available GWAS data imputed to the 1000 Genomes European phase I (a) panel (Online Methods). Finally, we performed a joint analysis of the discovery and replication phases.

We identified 97 statistically independent SNPs meeting replication criteria (preplication < 0.05, pjoint < 5 × 10-8, and pjoint < pdiscovery); 93 primary signals (Supplementary Figs. 3-95) and four secondary signals. Of these, 48 are novel to multiple sclerosis (Table 1) and 49 correspond to previously identified multiple sclerosis effects (Table 2). An additional 11 independent SNPs showed suggestive evidence of association (pjoint < 1 × 10-6) (Supplementary Table 2).

Table 1.

48 Novel non-MHC susceptibility loci associated with multiple sclerosis at a genome-wide significance level

Discovery Replication Joint

SNP Chr Positiona RA RAF P-value OR RAF P-value OR P-value OR Geneb Function
rs3007421 1 6530189 A 0.12 9.6 × 10-7 1.12 0.13 8.8 × 10-5 1.10 4.7 × 10-10 1.11 PLEKHG5 intronic
rs12087340 1 85746993 A 0.09 5.1 × 10-12 1.22 0.09 2.9 × 10-10 1.20 1.1 × 10-20 1.21 BCL10 intergenic
rs11587876 1 85915183 A 0.79 8.4 × 10-8 1.12 0.81 2.9 × 10-3 1.06 4.4 × 10-9 1.09 DDAH1 intronic
rs666930 1 120258970 G 0.53 7.5 × 10-8 1.09 0.53 1.3 × 10-5 1.07 6.0 × 10-12 1.08 PHGDH intronic
rs2050568 1 157770241 G 0.53 1.3 × 10-6 1.08 0.54 2.3 × 10-5 1.07 1.5 × 10-10 1.08 FCRL1 intronic
rs35967351 1 160711804 A 0.67 1.7 × 10-6 1.09 0.68 5.9 × 10-6 1.09 4.4 × 10-11 1.09 SLAMF7 intronic
rs4665719 2 25017860 G 0.25 6.8 × 10-6 1.09 0.25 1.1 × 10-4 1.08 3.1 × 10-9 1.08 CENPO intronic
rs842639 2 61095245 A 0.65 1.7 × 10-9 1.11 0.67 1.4 × 10-6 1.09 2.0 × 10-14 1.10 FLJ16341 ncRNA
rs9967792 2 191974435 G 0.62 1.8 × 10-9 1.11 0.64 1.2 × 10-4 1.07 3.5 × 10-12 1.09 STAT4 intronic
rs11719975 3 18785585 C 0.27 5.4 × 10-6 1.09 0.28 4.1 × 10-4 1.07 1.1 × 10-8 1.08 intergenic
rs4679081 3 33013483 G 0.52 1.2 × 10-5 1.08 0.55 3.7 × 10-4 1.07 2.2 × 10-9 1.07 CCR4 intergenic
rs9828629 3 71530346 G 0.62 5.5 × 10-6 1.08 0.64 8.5 × 10-6 1.08 1.9 × 10-10 1.08 FOXP1 intronic
rs2726518 4 106173199 C 0.55 1.2 × 10-5 1.09 0.58 4.7 × 10-4 1.06 3.9 × 10-8 1.07 TET2 intronic
rs756699 5 133446575 A 0.87 3.0 × 10-6 1.12 0.88 6.5 × 10-6 1.11 8.8 × 10-11 1.12 TCF7 intergenic
nonec 5 141506564 C 0.61 6.0 × 10-5 1.07 0.62 1.5 × 10-5 1.08 3.6 × 10-9 1.07 NDFIP1 intronic
rs4976646 5 176788570 G 0.34 1.0 × 10-12 1.13 0.36 5.0 × 10-7 1.10 4.4 × 10-18 1.12 RGS14 intronic
rs17119 6 14719496 A 0.81 1.9 × 10-6 1.11 0.80 1.2 × 10-5 1.10 1.0 × 10-10 1.10 intergenic
rs941816 6 36375304 G 0.18 4.5 × 10-9 1.13 0.20 8.3 × 10-5 1.08 3.9 × 10-12 1.11 PXT1 intronic
rs1843938 7 3113034 A 0.44 2.2 × 10-6 1.08 0.44 1.1 × 10-5 1.08 1.2 × 10-10 1.08 CARD11 intergenic
rs706015 7 27014988 C 0.18 1.3 × 10-9 1.14 0.18 9.9 × 10-3 1.06 1.1 × 10-9 1.10 intergenic
rs917116 7 28172739 C 0.20 2.1 × 10-8 1.12 0.21 5.8 × 10-3 1.06 3.3 × 10-9 1.09 JAZF1 intronic
rs60600003 7 37382465 C 0.10 2.5 × 10-8 1.16 0.10 4.2 × 10-7 1.14 6.0 × 10-14 1.15 ELMO1 intronic
rs201847125d 7 50325567 G 0.70 2.9 × 10-8 1.11 0.70 6.7 × 10-5 1.09 1.2 × 10-11 1.10 IKZF1 intergenic
rs2456449 8 128192981 G 0.36 2.2 × 10-8 1.10 0.37 3.8 × 10-3 1.05 1.8 × 10-9 1.08 intergenic
rs793108 10 31415106 A 0.50 5.6 × 10-8 1.09 0.51 1.8 × 10-5 1.07 6.1 × 10-12 1.08 intergenic
rs2688608 10 75658349 A 0.55 6.4 × 10-5 1.07 0.56 2.0 × 10-4 1.06 4.6 × 10-8 1.07 C10orf55 intergenic
rs7120737 11 47702395 G 0.15 7.6 × 10-8 1.13 0.15 1.0 × 10-3 1.08 1.0 × 10-9 1.10 AGBL2 intronic
rs694739 11 64097233 A 0.62 1.3 × 10-5 1.08 0.62 3.8 × 10-5 1.07 2.0 × 10-9 1.07 PRDX5 intergenic
rs9736016 11 118724894 T 0.63 2.2 × 10-8 1.10 0.63 2.6 × 10-8 1.10 3.0 × 10-15 1.10 CXCR5 intergenic
rs12296430 12 6503500 C 0.19 3.6 × 10-10 1.14 0.21 1.7 × 10-5 1.09 7.2 × 10-14 1.12 LTBR intergenic
rs4772201 13 100086259 A 0.82 1.7 × 10-7 1.12 0.83 1.1 × 10-4 1.09 1.3 × 10-10 1.10 MIR548AN intergenic
rs12148050 14 103263788 A 0.35 1.5 × 10-5 1.08 0.36 4.3 × 10-9 1.10 5.1 × 10-13 1.09 TRAF3 intronic
rs59772922 15 79207466 A 0.83 4.0 × 10-6 1.11 0.83 5.4 × 10-4 1.08 1.2 × 10-8 1.09 CTSH intergenic
rs8042861 15 90977333 A 0.44 9.8 × 10-7 1.08 0.45 3.4 × 10-4 1.06 2.2 × 10-9 1.07 IQGAP1 intronic
rs6498184 16 11435990 G 0.81 2.1 × 10-10 1.15 0.82 6.5 × 10-9 1.14 7.4 × 10-18 1.15 RMI2 intergenic
rs7204270* 16 30156963 G 0.50 9.3 × 10-8 1.09 0.49 3.7 × 10-5 1.08 1.6 × 10-11 1.09 MAPK3 intergenic
rs1886700 16 68685905 A 0.14 8.8 × 10-6 1.11 0.14 3.2 × 10-4 1.08 1.3 × 10-8 1.10 CDH3 intronic
rs12149527 16 79110596 A 0.47 1.7 × 10-6 1.08 0.47 4.3 × 10-6 1.08 3.3 × 10-11 1.08 WWOX intronic
rs7196953 16 79649394 A 0.29 2.6 × 10-5 1.08 0.30 7.9 × 10-7 1.09 1.0 × 10-10 1.09 MAF intergenic
rs12946510 17 37912377 A 0.47 8.5 × 10-6 1.08 0.48 8.0 × 10-5 1.07 2.9 × 10-9 1.07 IKZF3 intergenic
rs4794058 17 45597098 A 0.50 1.6 × 10-5 1.07 0.52 3.5 × 10-10 1.11 1.0 × 10-13 1.09 NPEPPS intergenic
rs2288904 19 10742170 G 0.77 9.6 × 10-10 1.14 0.78 5.4 × 10-4 1.07 1.6 × 10-11 1.10 SLC44A2 exonic
rs1870071 19 16505106 G 0.29 5.7 × 10-10 1.12 0.30 4.6 × 10-7 1.09 2.0 × 10-15 1.10 EPS15L1 intronic
rs17785991 20 48438761 A 0.35 6.4 × 10-7 1.09 0.34 5.9 × 10-3 1.05 4.2 × 10-8 1.07 SLC9A8 intronic
rs2256814 20 62373983 A 0.19 8.3 × 10-7 1.11 0.21 6.4 × 10-4 1.08 3.5 × 10-9 1.09 SLC2A4RG intronic
Secondary
rs7769192e 6 137962655 G 0.55 1.3 × 10-5 1.08 0.54 5.1 × 10-5 1.07 3.3 × 10-9 1.08 intergenic
rs533646f 11 118566746 G 0.68 3.6 × 10-7 1.10 0.68 3.9 × 10-5 1.08 7.6 × 10-11 1.09 TREH intergenic
rs4780346g 16 11288806 A 0.23 6.8 × 10-6 1.09 0.25 1.5 × 10-5 1.09 4.4 × 10-10 1.09 CLEC16A intergenic

All listed signals had a discovery P-value ≤ 1.0 × 10-4, a replication P-value ≤ 5.0 × 10-2, and a joint P-value ≤ 5.0 × 10-8

All P-values are two-sided

RA= Risk Allele, RAF = Risk Allele Frequency

a

Position is based on human genome 19 and dbSNP 137.

b

Nearest gene listed if within 50Kb. Bold indicates Gene Ontology Immune System Process.

c

A proxy SNP (rs1036207, r2 = 0.99) and

d

(rs716719, r2=1.00) was used in replication.

e

The P-value and OR values provided are after conditioning on rs67297943 (Known – see Table 2),

f

rs9736016, and

g

rs12927355 (Known – see Table 2).

*

Note primary was rs11865086 (P-value = 1.77 × 10-8) in Discovery but not available in Replication so the best proxy was used.

Table 2.

49 Known non-MHC susceptibility loci associated with multiple sclerosis at a genome-wide significance level

Discovery Replication Joint

SNP Chr Positiona RA RAF P-value OR RAF P-value OR P-value OR Geneb Function
rs3748817 1 2525665 A 0.64 1.3 × 10-12 1.14 0.65 1.2 × 10-15 1.15 1.3 × 10-26 1.14 MMEL1 intronic
rs41286801 1 92975464 A 0.14 7.9 × 10-16 1.20 0.16 2.1 × 10-12 1.17 1.4 × 10-26 1.19 EVI5 UTR3
rs7552544* 1 101240893 A 0.56 3.7 × 10-6 1.08 0.43 3.3 × 10-12 1.12 1.9 × 10-16 1.10 VCAM1 intergenic
rs6677309 1 117080166 A 0.88 1.5 × 10-28 1.34 0.88 4.1 × 10-16 1.24 5.4 × 10-42 1.29 CD58 intronic
rs1359062 1 192541472 C 0.82 1.8 × 10-13 1.18 0.83 2.1 × 10-8 1.13 4.8 × 10-20 1.15 RGS1 intergenic
rs55838263 1 200874728 A 0.71 1.4 × 10-9 1.12 0.71 3.9 × 10-11 1.13 4.0 × 10-19 1.13 C1orf106 intronic
rs2163226 2 43361256 A 0.71 7.0 × 10-8 1.10 0.73 3.8 × 10-10 1.14 2.1 × 10-16 1.12 intergenic
rs7595717 2 68587477 A 0.26 3.3 × 10-7 1.10 0.27 6.8 × 10-8 1.10 1.2 × 10-13 1.10 PLEK intergenic
rs9989735 2 231115454 C 0.18 7.8 × 10-14 1.17 0.19 6.8 × 10-11 1.14 4.2 × 10-23 1.16 SP140 intronic
rs2371108 3 27757018 A 0.38 2.1 × 10-6 1.08 0.39 5.8 × 10-11 1.12 1.5 × 10-15 1.10 EOMES downstream
rs1813375 3 28078571 A 0.47 5.7 × 10-18 1.15 0.49 4.4 × 10-16 1.15 1.9 × 10-32 1.15 intergenic
rs1131265 3 119222456 C 0.80 2.0 × 10-15 1.19 0.81 4.8 × 10-10 1.14 1.4 × 10-23 1.17 TIMMDC1 exonic
rs1920296* 3 121543577 C 0.64 6.8 × 10-15 1.14 0.64 5.5 × 10-9 1.10 6.5 × 10-22 1.12 IQCB1 intronic
rs2255214* 3 121770539 C 0.52 5.3 × 10-13 1.13 0.52 3.3 × 10-13 1.13 1.2 × 10-24 1.13 CD86 intergenic
rs1014486 3 159691112 G 0.43 1.2 × 10-9 1.11 0.44 1.4 × 10-10 1.11 1.1 × 10-18 1.11 IL12A intergenic
rs7665090 4 103551603 G 0.52 2.4 × 10-6 1.08 0.53 5.0 × 10-4 1.13 1.0 × 10-8 1.09 MANBA intergenic
rs6881706 5 35879156 C 0.72 4.9 × 10-9 1.12 0.73 1.7 × 10-9 1.12 4.3 × 10-17 1.12 IL7R intergenic
rs6880778 5 40399096 G 0.60 1.7 × 10-8 1.10 0.61 3.9 × 10-13 1.13 8.1 × 10-20 1.12 intergenic
rs71624119 5 55440730 G 0.76 2.7 × 10-9 1.12 0.76 1.9 × 10-5 1.09 3.4 × 10-13 1.11 ANKRD55 intronic
rs72928038 6 90976768 A 0.17 7.6 × 10-7 1.11 0.19 9.0 × 10-11 1.17 1.5 × 10-15 1.14 BACH2 intronic
rs11154801 6 135739355 A 0.37 2.3 × 10-9 1.11 0.37 1.0 × 10-12 1.13 1.8 × 10-20 1.12 AHI1 intronic
rs17066096 6 137452908 G 0.23 5.9 × 10-12 1.14 0.25 4.1 × 10-13 1.15 1.6 × 10-23 1.14 IL22RA2 intergenic
rs67297943 6 138244816 A 0.78 4.8 × 10-8 1.12 0.80 2.5 × 10-6 1.11 5.5 × 10-13 1.11 TNFAIP3 intergenic
rs212405 6 159470559 T 0.62 1.4 × 10-15 1.15 0.64 1.8 × 10-7 1.10 8.0 × 10-21 1.12 TAGAP intergenic
rs1021156 8 79575804 A 0.24 5.6 × 10-10 1.12 0.26 2.1 × 10-8 1.11 8.5 × 10-17 1.11 ZC2HC1A intergenic
rs4410871 8 128815029 G 0.72 2.0 × 10-9 1.12 0.72 3.4 × 10-8 1.11 4.3 × 10-16 1.11 MIR1204 intergenic
rs759648 8 129158945 C 0.31 2.8 × 10-6 1.09 0.31 3.7 × 10-5 1.08 5.0 × 10-10 1.08 MIR1208 intergenic
rs2104286 10 6099045 A 0.72 7.6 × 10-23 1.21 0.73 3.6 × 10-26 1.23 2.3 × 10-47 1.22 IL2RA intronic
rs1782645 10 81048611 A 0.43 4.3 × 10-7 1.09 0.41 6.2 × 10-10 1.11 2.5 × 10-15 1.10 ZMIZ1 intronic
rs7923837 10 94481917 G 0.61 4.6 × 10-9 1.11 0.62 2.0 × 10-9 1.11 4.3 × 10-17 1.11 HHEX intergenic
rs34383631 11 60793330 A 0.40 5.7 × 10-10 1.11 0.39 4.5 × 10-15 1.15 3.7 × 10-23 1.13 CD6 intergenic
rs1800693 12 6440009 G 0.40 6.9 × 10-16 1.14 0.41 1.0 × 10-13 1.14 6.7 × 10-28 1.14 TNFRSF1A intronic
rs11052877 12 9905690 G 0.36 5.4 × 10-9 1.10 0.38 1.2 × 10-5 1.08 5.6 × 10-13 1.09 CD69 UTR3
rs201202118c 12 58182062 A 0.67 7.4 × 10-13 1.14 0.67 1.6 × 10-10 1.12 9.0 × 10-22 1.13 TSFM intronic
rs7132277 12 123593382 A 0.19 1.9 × 10-6 1.10 0.19 1.4 × 10-8 1.13 1.9 × 10-13 1.12 PITPNM2 intronic
rs2236262 14 69261472 A 0.50 1.2 × 10-5 1.08 0.50 3.8 × 10-8 1.09 2.5 × 10-12 1.08 ZFP36L1 intronic
rs74796499 14 88432328 C 0.95 8.5 × 10-11 1.31 0.95 4.5 × 10-11 1.33 2.4 × 10-20 1.32 GALC intronic
rs12927355 16 11194771 G 0.68 8.2 × 10-27 1.21 0.69 4.3 × 10-21 1.18 6.4 × 10-46 1.20 CLEC16A intronic
rs35929052 16 85994484 G 0.89 3.3 × 10-7 1.14 0.88 3.6 × 10-6 1.15 5.9 × 10-12 1.15 IRF8 intergenic
rs4796791 17 40530763 A 0.36 1.8 × 10-8 1.10 0.36 1.2 × 10-13 1.14 3.7 × 10-20 1.12 STAT3 intronic
rs8070345 17 57816757 A 0.45 5.4 × 10-16 1.14 0.46 1.9 × 10-9 1.10 2.2 × 10-23 1.12 VMP1 intronic
rs1077667 19 6668972 G 0.79 3.5 × 10-13 1.16 0.79 8.4 × 10-13 1.16 1.7 × 10-24 1.16 TNFSF14 intronic
rs34536443 19 10463118 C 0.95 1.2 × 10-8 1.28 0.96 2.9 × 10-7 1.30 1.8 × 10-14 1.29 TYK2 exonic
rs11554159 19 18285944 G 0.73 2.6 × 10-13 1.15 0.74 1.4 × 10-12 1.15 1.9 × 10-24 1.15 IFI30 exonic
rs8107548 19 49870643 G 0.25 2.0 × 10-6 1.09 0.26 2.5 × 10-10 1.13 5.7 × 10-15 1.11 DKKL1 intronic
rs4810485 20 44747947 A 0.25 1.8 × 10-5 1.08 0.25 1.4 × 10-12 1.14 7.7 × 10-16 1.11 CD40 intronic
rs2248359 20 52791518 G 0.60 9.8 × 10-5 1.07 0.62 8.2 × 10-11 1.12 2.0 × 10-13 1.09 CYP24A1 intergenic
rs2283792 22 22131125 C 0.51 1.1 × 10-6 1.08 0.53 5.4 × 10-11 1.11 5.5 × 10-16 1.10 MAPK1 intronic
Secondary
rs523604d 11 118755738 A 0.53 2.5 × 10-7 1.09 0.54 4.0 × 10-9 1.11 6.2 × 10-15 1.10 CXCR5 intronic

All listed signals had a discovery P-value ≤ 1.0 × 10-4, a replication P-value ≤ 5.0 × 10-2, and a joint P-value ≤ 5.0 × 10-8

All P-values are two-sided

RA = Risk Allele, RAF = Risk Allele Frequency

a

Position is based on human genome 19 and dbSNP 137.

b

Nearest gene listed if within 50Kb. Bold indicates Gene Ontology Immune System Process.

c

A proxy SNP (rs10431552, r2 = 0.99) was used in replication.

d

The P-value and OR values provided are after conditioning on rs9736016 and rs533646 (both Novel – see Table 1).

*

These three SNPs were not primary in the 2011 GWAS, two were secondary and the third was tertiary in that study.

The strongest novel association, rs12087340 (pjoint = 1.1 × 10-20, OR = 1.21), lies between BCL10 (B-cell CLL / lymphoma 10) and DDAH1 (dimethylarginine dimethylaminohylaminohydrolase 1). The protein encoded by BCL10 contains a caspase recruitment domain (CARD) and has been shown to activate NF-kappaB.17 The latter is a signalling molecule that plays an important role in controlling gene expression in inflammation, immunity, cell proliferation, and apoptosis. It has been pursued as a potential therapeutic target for multiple sclerosis.18 BCL10 is also reported to interact with other CARD domain containing proteins including CARD11.19 We have also identified a novel association of rs1843938 (pjoint = 1.2 × 10-10, OR = 1.08), which is only 30 kb from CARD11.

One novel SNP was found within an exon, rs2288904 (pjoint = 1.6 × 10-11, OR= 1.10); a missense variant in SLC44A2 (solute carrier family 44, member 2). Notably, this variant is also reported as a monocyte-specifccis-acting eQTL for the antisense transcript of the nearby ILF3 (interleukin enhancer binding factor 3).20 This protein was first discovered to be a subunit of a nuclear factor found in activated T-cells, which is required for T-cell expression of IL2, an important molecule regulating many aspects of inflammation.

Of the 49 previously identified effects,9,10,21 37 are in designated fine-mapping regions, and 23 of these 37 signals were localized to a single gene based on genomic position (Supplementary Table 3). Recognizing that proximity does not necessarily indicate functional importance, this emphasizes the utility of dense mapping in localizing signals from a genome-wide screen. The ImmunoChip analysis furthered the understanding of previously proposed secondary signals at three loci (Supplementary Note and Supplementary Tables 4-6); in particular we showed that the effects of two previously proposed independent associations at the IL2RA locus are driven by a single variant, rs2104286.7,22.

In an effort to define the functionally relevant variants underlying these associations, we further studied the regions surrounding the 97 associated SNPs using both a Bayesian and frequentist approach in 6,356 multiple sclerosis subjects and 9,617 healthy controls from the UK (Online Methods).23 Based on imputation quality, fine-mapping was possible in 68 regions (Supplementary Table 7): 66 of 93 primary (Fig. 2A) and two of four secondary. Eight of the 68 regions were fine-mapped to high resolution (Table 3, Fig. 2B and Supplementary Fig. 96). One third of the variants identified in these eight regions were imputed, indicating reliance on imputation even with dense genotyping coverage.

Figure 2. Bayesian fine-mapping within primary regions of association.

Figure 2

a) Summary of the extent of fine-mapping across 66 regions in 9,617 healthy controls from the UK, showing the the physical extent of, the number of variants, and the number of genes spanned by the posterior 90% and 50% credible sets. b) Detail of fine-mapping in region of TNFSF14. Above the x-axis indicates the Bayes Factor summarizing evidence for association for the SNPs prior to conditioning (blue markers) while below the x-axis indicates the Bayes Factor after conditioning on the lead SNP (rs1077667). Mb=Megabases.

Table 3.

The 18 variants from the 8 regions with consistent high resolution fine-mapping

Gene SNP Chr Positiona Posterior GERP Functional Annotationb
TNFSF14 rs1077667 19 6668972 0.74 -3.89 intronic, TFBS / DNase1 peak, correlates with serum levels of TNFSF14
IL2RA rs2104286 10 6099045 0.93 -0.47 intronic, correlates with soluble IL-2RA levels
TNFRSF1A rs1800693 12 6440009 0.69 2.53 intronic, causes splicing defect and truncated soluble TNFRSF1A
rs4149580c 12 6446990 0.10 2.06 intronic
IL12A rs1014486 3 159691112 0.67 0.24 -
CD6 rs34383631 11 60793330 0.20 1.66 -
rs4939490c 11 60793651 0.14 -0.53 -
rs4939491c 11 60793722 0.14 -0.37 -
rs4939489 11 60793648 0.10 3.25 -
TNFAIP3 rs632574 6 137959118 0.27 -1.15 -
rs498549c 6 137984935 0.20 0.52 -
rs651973 6 137996134 0.17 2.41 downstream of RP11-95M15.1 lincRNA gene
rs536331 6 137993049 0.15 0.19 upstream of RP11-95M15.1 lincRNA gene
CD58 rs6677309 1 117080166 0.21 -1.18 intronic, TFBS / DNase1 peak
rs35275493c 1 117095502 0.24 0.75 intronic (insertion)
rs10754324c 1 117093035 0.22 0.32 intronic
rs1335532 1 117100957 0.17 -1.32 intronic
STAT4 rs78712823 2 191958581 0.59 -3.98 intronic

All listed variants have posterior ≥ 0.1 in regions where ≤ 5 variants explain the top 50% of the posterior and the top SNP from the frequentist analysis lives in the 90% confidence interval, ordered by maximum posterior.

Posterior denotes the posterior probability of any variant driving association. GERP denotes Genomic Evolutionary Rate Profiling.

a

Position is based on human genome 19 and dbSNP 137.

b

Functional data from VEP, eQTL browser, Fairfax et al. (2012), pubmed searches, 1000G. Dash indicates intergenic with no additional annotation. Variants without annotation are intergenic and have no reported regulatory consequence.

c

Imputed variant.

To assess whether functional annotation24 provides clues about the molecular mechanisms associated with genetic risk, we considered the relationship of variants to described coding and regulatory features in these eight regions. Although we found no variants with missense or nonsense effects, there was a notable enrichment for variants with functional effects: one known to affect splicing,25 three known to correlate with RNA or serum protein levels22,26,27 and several in transcription-factor binding and DNase I hypersensitive sites.28, 29 Four of the 18 variants in the fine-mapped regions are within conserved regions (GERP > 2).30 This lack of functional annotation likely reflects the limited repertoire of reference expression and epigenomic profiles and suggests that the function of the variants may be cell-type or cell-state specific, as has been reported for many eQTLs in immune cell types.20

To determine the Gene Ontology (GO) processes of the 97 associated variants, we used MetaCore from Thomson Reuters (Online Methods). We found the majority of the 97 variants lie within 50 kb of genes having immunological function. Of the 86 unique genes represented, 35 are linked to the GO immune system process (Table 1 and Table 2). We do not see a substantial over- or under- representation of certain GO processes when comparing the novel and previously identified loci, but this may be a limitation of ImmunoChip targeting genomic loci enriched for immunologically active genes, with more subtle distinctions between them not adequately captured by broad annotations such as GO.

Finally, we explored the overlap between our findings and those in autoimmune diseases with reported ImmunoChip analyses. We calculated the percentage of multiple sclerosis signals (110 non-MHC, Supplementary Table 8) overlapping those of other autoimmune diseases by requiring an r2 ≥ 0.8 between the best variants reported in each study using SNAP.31 In total we find that ~22% of our signals overlap at least one other autoimmune disease. More specificially, ~9.1% overlap with inflammatory bowel disease (IBD) - ~7.3% with ulcerative colitis (UC), ~9.1% with Crohn’s disease (CD) -15, ~9.1% with primary biliary cirrhosis (PBC),32, 33 ~4.5% with celiac disease (CeD),34 ~4.5% with rheumatoid arthritis (RhA),35 ~0.9% with psoriasis (PS),36 and ~2.7% with autoimmune thyroid disease (AITD).37 We report the same top variant seen in PBC for 7 loci. We also note that our best TYK2 variant (rs34536443)38 is also the most associated variant for PBC, PS and RhA. Lastly, AITD, CeD, PBC, and RhA report variants with pairwise r2 ≥ 0.8 to the multiple sclerosis variant near MMEL139 (Supplementary Table 8).

In summary, we have identified 48 new multiple sclerosis susceptibility variants. These novel loci expand our understanding of the immune system processes implicated in multiple sclerosis. We estimate that the 110 non-MHC established risk variants explain 20% of the sibling recurrence risk; 28% including the already identified MHC effects9 (Supplementary Note). Additionally, we have identified five regions where consistent high resolution fine-mapping implicated one variant which accounted for more than 50% of the posterior in previously identified regions of TNFSF14, IL2RA, TNFRSF1A, IL12A, and STAT4. Our study further implicates NF-kappaB in multiple sclerosis pathobiology18, emphasizes the value of dense fine-mapping in large follow-up data sets, and exposes the urgent need for functional annotation in relevant tissues. Understanding the implicated networks and their relation to environmental risk factors will promote the development of rational therapies and may enable the development of preventive strategies.

Online Methods

ImmunoChip data (discovery set)

Details of case ascertainment, processing and genotyping for the discovery phase are provided in the Supplementary Note (Supplementary Table 9). Genotype calling for all samples was performed using Opticall.40 Samples that performed poorly or were determined to be related were removed (Supplementary Table 10). The data were organized in 11 country level strata: ANZ (Australia + New Zealand), Belgium, Denmark, Finland, France, Germany, Italy, Norway, Sweden, United Kingdom (UK), and the United States of America (USA). SNP level quality control (Supplementary Table 11) and population outlier identification using principal components analysis (Supplementary Fig. 97) were done in each stratum separately.

Discovery set analysis

We applied logistic regression, assuming a per-allelic genetic model per data set, including the first five principal components as covariates to correct for population stratification (Supplementary Table 12 lists the per data set genomic inflation factors, λ). We then performed an inverse-variance meta-analysis of the 11 strata, under a fixed effects model, as implemented in PLINK.41 To be more conservative and account for any residual inflation in the test statistic, we applied the genomic control equivalent to the per-SNP standard error in each stratum. Specifically, we corrected the SNP standard errors by multiplying them with the square root of the raw genomic inflation factor λ, per data set, if the λ was >1.

Within the designated fine-mapping intervals, we applied a forward stepwise logistic regression to identify statistically independent effects. The primary SNP in each interval was included as a covariate, and the association analysis was repeated for the remaining SNPs. This process was repeated until no SNPs reached the minimum level of significance (p-value <1 × 10-4). Outside of the designated fine-mapping intervals, all SNPs having a p-value <1 × 10-4 were identified and grouped into sets based on a physical distance of less than 2Mb and a similar stepwise regression model was applied. Any SNPs to enter the model with p-value <1 × 10-4 after conditioning were considered statistically independent primary signals.

In addition, because of the close physical proximity between some fine-mapping intervals and SNP sets, independence was tested for all identified signals within 2Mb of one another. The and cluster plots (Supplementary Fig. 98) of all independent SNPs were examined, and the SNP was excluded if unsatisfactory. If any SNP was excluded, the forward stepwise logistic regression within that fine-mapping interval or SNP set was repeated after removal of the SNP. During this process, 17 additional SNPs were excluded based on cluster or forest plot review.

Replication Set

The replication phase included GWAS data organized into 15 strata. Within each stratum, poorly performing samples (call rate < 95%, gender discordance, excess heterozygosity) and poorly performing SNPs (Hardy-Weinberg equilibrium (HWE) p-value <1 × 10-6, minor allele frequency (MAF) < 1%, call rate < 95%) were removed. Principal components analysis was performed to identify population outliers per stratum, and the genomic control inflation factor was < 1.1 for each. The data included in the final discovery and replication analyses are summarized in Supplementary Table 13 and Supplementary Table 14. All the samples used in the replication set were unrelated to those in the discovery set; verified by identity-by-descent analysis.

We attempted replication of all non-MHC independent signals that reached a discovery p-value of <1 × 10-4 in a meta-analysis set of GWAS. Each data set was imputed to the 1000 Genomes European phase I (a) panel using BEAGLE42 to maximize the overlap between the Immunochip SNP content and the GWAS data. Post-imputation genotypic probabilities were used in a logistic regression model, per stratum, to estimate SNP effect sizes and p-values. By using the post-imputation genotypic probabilities, we penalized SNPs that didn’t have good imputation quality, thus ensuring a conservative analysis. Furthermore, we accounted for population stratification in each data set by including the first five principal components in the logistic model. We then meta-analysed the effect size and respective standard errors of the 15 strata using a fixed effects model inverse-variance method. We applied the genomic control equivalent to the per-SNP standard error in each stratum, controlling for the respective genomic inflation factor λ (Supplementary Table 14).

To replicate the primary SNPs per identified signal in the discovery phase, we used the replication effect size and respective standard error. For the secondary and tertiary SNPs, we fitted the same exact models as in the discovery phase, per data set. We then performed fixed effects meta-analysis to estimate an effect size that corresponds to the same logistic model. In the case that a SNP was not present in the replication set, we replaced it with a perfectly tagging SNP, i.e. a SNP that had r2 and D’ equal to 1. If a perfectly tagging SNP was not available, we selected a SNP that had equivalent MAF and the highest possible r2 and D’. Estimation of r2 and D’ for this objective were based on the ImmunoChip control samples.

Joint analysis (discovery and replication sets)

The discovery and replication phase effect sizes and respective standards errors were meta-analysed under a fixed effects model. A SNP was considered replicated when all three of the following criteria were met: 1) replication p-value <5.0 × 10-2, 2) joint p-value <5 × 10-8, and 3) the joint p-value was more statistically significant than the discovery p-value. SNPs that reached a p-value of <1 × 10-6 but did not pass the genome-wide threshold, were coined suggested if the above criteria 1) and 3) were met.

Fine-mapping of association signals

To fine-map signals of association we used a combination of imputation and Bayesian methodology.23 Around each of the 97 associated SNPs, 2Mb were isolated in the discovery and replication phase UK data as well as the European samples from the Phase 1 1000G.28 Forming the single largest cohort, only UK samples were considered to minimize the effects of differential imputation quality between populations of different ancestry. In addition to the previous quality control, SNPs with failed alignment or a difference in MAF > 10% between the typed cohorts and the 1000G samples, MAF < 1%, or HWE p-value <1.0 × 10-4 were removed.

Imputation was performed separately for the UK discovery and replication cohorts on each 2Mb region using the default settings of IMPUTEv2.43,44 Missing genotypes in the genotyped SNPs were not imputed, and any imputed SNP that failed the HWE and MAF threshold was subsequently removed. We carried out frequentist and Bayesian association tests on all SNPs in each cohort separately, assuming additivity, using the default settings of SNPTESTv2.45 Frequentist fixed-effect meta-analysis was carried out using the software META.46 Bayesian meta-analysis was carried out using an independence prior (near-identical results were obtained using a fixed-effect Bayesian meta-analysis).

To identify regions where reliable fine-mapping could be achieved, we used the information score (INFO, obtained from IMPUTEv2) as identified from the 1000G samples. Specifically, we measured the fraction of variants with both r2 > 0.5 and r2 > 0.8 to the primary associated variant, having greater than 50% and 80% INFO scores respectively. Regions where any SNP with r2 > 0.5 had INFO < 50% were excluded. We also excluded regions where the top hit from imputation had an INFO score less than 80%. Regions were considered to be fine-mapped with high quality when all variants with r2 > 0.8 had at least 80% INFO. Within these regions, we excluded variants where the inferred direction of association was opposite in the UK discovery and replication cohorts.

To measure the posterior probability that any single variant drives association, we calculated the Bayes Factor. Under the assumption that there is a single causal variant in the region, this is proportional to the probability that the variant drives the association.23 We identified the smallest set of variants that contained 90% and 50% of the posterior probability. We called a region successfully and consistently fine-mapped if there were at most five variants in the 50% confidence interval and the top SNP from the frequentist analysis lived in the 90% confidence interval. For these regions, we annotated variants with information about evolutionary conservation, predicted coding consequence, regulation, published associations to expression or DNase I hypersensitive sites using ANNOVAR,47 VEP,24 and the eQTL browser, a recent immune cell expression study20, and other literature.

Gene Ontology

To determine the GO processes for which our associated variants were involved, we used MetaCore from Thomson Reuters. We annotated the processes for the unique genes within 50Kb of the variants.

Cross disease comparison

In order to explore the potential overlap with variants identified across other autoimmune diseases, we calculated the percentage overlap of reported variants found in other ImmunoChip reports to our ImmunoChip results. The top variants reported as either novel or previously known in other ImmunoChip reports were compared with the 110 variants representing both our novel and previous discoveries in multiple sclerosis. In order for a signal to be considered as overlapping, we required an r2 ≥ 0.8 using the Pairwise LD function of the SNAP tool in European samples.31

Secondary analyses

We performed a severity based analysis of MSSS in cases only from the discovery phase (Supplementary Fig. 99). In addition, a transmission disequilibrium test was done in 633 trios to test for transmission of the 97 identified risk alleles (Supplementary Fig. 100). Details are given in the Supplementary Note.

Supplementary Material

1
2

Acknowledgments

We thank participants, referring nurses, physicians, and funders - National Institutes of Health, Wellcome Trust, UK MS Society, UK Medical Research Council, US National MS Society, Cambridge NIHR BRC, DeNDRon, Bibbi and Niels Jensens Foundation, Swedish Brain Foundation, Swedish Research Council, Knut and Alice Wallenberg Foundation, Swedish Heart-Lung Foundation, Foundation for Strategic Research, Stockholm County Council, Karolinska Institutet, Institut National de la Santé et de la Recherche Médicale, Fondation d’Aide pour la Recherche sur la Sclérose En Plaques, Association Française contre les Myopathies, GIS-IBISA, German Ministry for Education and Research, German Competence Network MS, Deutsche Forschungsgemeinschaft, Munich Biotec Cluster M4, Fidelity Biosciences Research Initiative, Research Foundation Flanders, Research Fund KU Leuven, Belgian Charcot Foundation, Gemeinnützige Hertie Stiftung, University Zurich, Danish MS Society, Danish Council for Strategic Research, Academy of Finland, Sigrid Juselius Foundation, Helsinki University, Italian MS Foundation, Fondazione Cariplo, Italian Ministry of University and Research, CRT Foundation of Turin, Italian Ministry of Health, Italian Institute of Experimental Neurology, MS association of Oslo, Norwegian Research Council, South Eastern Norwegian Health Authorities, Australian National Health and Medical Research Council, Dutch MS Foundation, Kaiser Permanente. We acknowledge British 1958 Birth Cohort, UK National Blood Service, Vanderbilt University Medical Center’s BioVU DNA Resources Core, Centre de Ressources Biologiques du Réseau Français d’Etude Génétique de la Sclérose en Plaques, Norwegian Bone Marrow Registry, Norwegian MS Registry and Biobank, North American Research Committee on MS Registry, Brigham and Womens Hospital PhenoGenetic Project and DILGOM funded by the Academy of Finland. See Supplementary Note for details.

Footnotes

Author Contributions

M.F.D., D. Booth, A.O., J.S., B. Fontaine, B.H., C. Martin, F.Z., S.D.’A., F.M.-B., B.T., H.F.H., I. Kockum, J. Hillert, T.O., J.R.O., R.H., L.F.B., C. Agliardi, L.A., L. Bernardinelli, V.B., S.B., B.B., L. Brundin, D. Buck, H. Butzkeuven, W. Camu, P.C., E.G.C., I.C., G.C., I.C.-R., B.A.C.C., G.D., S.R.D., A.D.S., B.D., M.D., I.E., F.E., N.E., J.F., A.F., I.Y.F., D.G., C. Graetz, A. Graham, C. Guaschino, C. Halfpenny, G. Hall, J. Harley, T.H., C. Hawkins, C. Hillier, J. Hobart, M.H., I.J., A.J., B.K., A. Kermode, T. Kilpatrick, K.K., T. Korn, H.K., C.L.-F., J.L.-S, M.H.L., M.A.L., G.L., B.A.L., C.M.L., F.L., J. Lycke, S.M., C.P.M., R.M., V.M., D.M., G. Mazibrada, J.M., K.M., G.N., R.N., P.N., F.P., S.E.P., H.Q., M. Reunanen, W.R., N.P.R., M. Rodegher, D.R., M. Salvetti, F.S., R.C.S., C. Schaefer, S. Shaunak, L.S., S. Shields, V.S., M. Slee, P.S.S., M. Sospedra, A. Spurkland, V.T., J.T., A.T., P.T., C.V.D., E.M.V., S.V., J.S.W., A.W., J.F.W., J.Z., E.Z., J.L.H., M.A.P.-V., G.S., D.H., S.L.H., A.C., P.D.J., S.J.S. and J.L.M. were involved with case ascertainment and phenotyping. A. Kemppinen, D. Booth, A. Goris, A.O., B. Fontaine, S.D.’A., F.M.-B., H.F.H., I. Kockum, M.B., J.R.O., L.F.B., IIBDGC, H.B.S., A. Baker, N.B., L. Bergamaschi, I.L.B., P.B., D. Buck, S.J.C., L. Corrado, L. Cosemans, I.C.-R., V.D., J.F., A.F., V.G., I.J., I. Konidari, V.L., C.M.L., M. Lindén, J. Link, C. McCabe, I.M., H.Q., M. Sorosina, E.S., H.W., P.D.J., S.J.S. and J.L.M. processed the DNA. A. Kemppinen, A.O., B. Fontaine, M.B., R.H., L.F.B., WTCCC2, IIBDGC, R.A., H.B.S., N.B., T.M.C.B., H. Blackburn, P.B., W. Carpentier, L. Corrado, I.C.-R., D.C., V.D., P. Deloukas, S.E., A.F., H.H., P.H., A. Hamsten, S.E.H., I.J., I. Konidari, C.L., M. Larsson, M. Lathrop, F.M., I.M., J.M., H.Q., F.S., M. Sorosina, C.V.D., J.W., D.H., P.D.J., S.J.S. and J.L.M. conducted and supervised the genotyping of samples. A.H.B., N.A.P., D.K.X., M.F.D., A. Kemppinen, C.C., T.S.S., C. Spencer, M.B., IIBDGC, C. Anderson, S.E.B., A.T.D., P. Donnelly, B. Fiddes, P.G., G. Hellenthal, S.E.H., L.M., M.P., N.C.S.-B., J.L.H., M.A.P.-V., G. McVean, P.D.J., S.J.S. and J.L.M. performed the statistical analysis. A.H.B., N.A.P., D.K.X., M.F.D., A. Kemppinen, C.C., T.S.S., C. Spencer, D. Booth, A. Goris, A.O., J.S., B. Fontaine, B.H., F.Z., S.D.’A., F.M.-B., H.F.H., I. Kockum, M.B., R.H., L.F.B., C. Agliardi, M.A., C. Anderson, R.A., H.B.S., A. Baker, G.B., N.B., J.B., C.B., L. Bernardinelli, A. Berthele, V.B., T.M.C.B., H. Blackburn, I.L.B., B.B., D. Buck, S.J.C., W. Camu, P.C., E.G.C., I.C., G.C., L. Corrado, L. Cosemans, I.C.-R., B.A.C.C., D.C., G.D., S.R.D., P. Deloukas, A.D.S., A.T.D., P. Donnelly, B.D., M.D., S.E., F.E., N.E., B. Fiddes, J.F., A.F., C.F., D.G., C. Gieger, C. Graetz, A. Graham, V.G., C. Guaschino, A. Hadjixenofontos, H.H., C. Halfpenny, P.H., G. Hall, A. Hamsten, J. Harley, T.H., C. Hawkins, G. Hellenthal, C. Hillier, J. Hobart, M.H., S.E.H., I.J., A.J., B.K., I. Konidari, H.K., C.L., M. Larsson, M. Lathrop, C.L.-F., M.A.L., V.L., G.L., B.A.L., C.M.L., F.M., C.P.M., R.M., V.M., G. Mazibrada, C. McCabe, I.M., L.M., K.M., R.N., M.P., S.E.P., H.Q., N.P.R., M. Rodegher, D.R., M. Salvetti, N.C.S.-B., R.C.S., C. Schaefer, S. Shaunak, L.S., S. Shields, M. Sospedra, A. Strange, J.T., A.T., E.M.V., A.W., J.F.W., J.W., J.Z., J.L.H., A.J.I., G. McVean, P.D.J., S.J.S. and J.L.M. collected and managed the project data. A.H.B., N.A.P., M.F.D., A. Kemppinen, C.C., T.S.S., C. Spencer, J.S., B.H., F.Z., S.D.’A., F.M.-B., H.F.H., J. Hillert, T.O., M.B., J.R.O., R.H., L.F.B., L.A., C. Anderson, G.B., J.B., C.B., A. Berthele, E.G.C., G.C., P. Donnelly, F.E., C.F., C. Gieger, C. Graetz, G. Hellenthal, M.J., T. Korn, M.A.L., R.M., M.P., M. Sospedra, A. Spurkland, A. Strange, J.W., J.L.H., M.A.P.-V., A.J.I., G.S., D.H., S.L.H., A.C., G. McVean, P.D.J., S.J.S. and J.L.M. contributed to the study concept and design. A.H.B., N.A.P., D.K.X., G. McVean, P.D.J., S.J.S. and J.L.M. prepared the manuscript. All authors reviewed the final manuscript.

The authors have no competing financial interests

References

  • 1.Gourraud PA, Harbo HF, Hauser SL, Baranzini SE. The genetics of multiple sclerosis: an up-to-date review. Immunol Rev. 2012;248:87–103. doi: 10.1111/j.1600-065X.2012.01134.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Nylander A, Hafler DA. Multiple sclerosis. J Clin Invest. 2012;122:1180–8. doi: 10.1172/JCI58649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Compston A, et al. McAlpine’s Multiple Sclerosis. Churchill Livingstone; London: 2006. [Google Scholar]
  • 4.Dyment DA, Yee IM, Ebers GC, Sadovnick AD. Multiple sclerosis in stepsiblings: recurrence risk and ascertainment. J Neurol Neurosurg Psychiatry. 2006;77:258–9. doi: 10.1136/jnnp.2005.063008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hemminki K, Li X, Sundquist J, Hillert J, Sundquist K. Risk for multiple sclerosis in relatives and spouses of patients diagnosed with autoimmune and related conditions. Neurogenetics. 2009;10:5–11. doi: 10.1007/s10048-008-0156-y. [DOI] [PubMed] [Google Scholar]
  • 6.Jersild C, Svejgaard A, Fog T. HL-A antigens and multiple sclerosis. Lancet. 1972;1:1240–1. doi: 10.1016/s0140-6736(72)90962-2. [DOI] [PubMed] [Google Scholar]
  • 7.IMSGC. Risk Alleles for Multiple Sclerosis Identified by a Genomewide Study. N Engl J Med. 2007;357:851–62. doi: 10.1056/NEJMoa073493. [DOI] [PubMed] [Google Scholar]
  • 8.De Jager PL, et al. Meta-analysis of genome scans and replication identify CD6, IRF8 and TNFRSF1A as new multiple sclerosis susceptibility loci. Nat Genet. 2009;41:776–82. doi: 10.1038/ng.401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.IMSGC & WTCCC2. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476:214–9. doi: 10.1038/nature10251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Patsopoulos NA, et al. Genome-wide meta-analysis identifies novel multiple sclerosis susceptibility loci. Ann Neurol. 2011;70:897–912. doi: 10.1002/ana.22609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.IMSGC. Evidence for polygenic susceptibility to multiple sclerosis--the shape of things to come. Am J Hum Genet. 2010;86:621–5. doi: 10.1016/j.ajhg.2010.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Baranzini SE. The genetics of autoimmune diseases: a networked perspective. Curr Opin Immunol. 2009;21:596–605. doi: 10.1016/j.coi.2009.09.014. [DOI] [PubMed] [Google Scholar]
  • 13.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]
  • 14.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]
  • 15.Jostins L, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119–24. doi: 10.1038/nature11582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Krzywinski M, et al. Circos: an information aesthetic for comparative genomics. Genome Res. 2009;19:1639–45. doi: 10.1101/gr.092759.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Willis TG, et al. Bcl10 is involved in t(1;14)(p22;q32) of MALT B cell lymphoma and mutated in multiple tumor types. Cell. 1999;96:35–45. doi: 10.1016/s0092-8674(00)80957-5. [DOI] [PubMed] [Google Scholar]
  • 18.Yan J, Greer JM. NF-kappa B, a potential therapeutic target for the treatment of multiple sclerosis. CNS Neurol Disord Drug Targets. 2008;7:536–57. doi: 10.2174/187152708787122941. [DOI] [PubMed] [Google Scholar]
  • 19.Wegener E, Krappmann D. CARD-Bcl10-Malt1 signalosomes: missing link to NF-kappaB. Sci STKE. 2007;2007:pe21. doi: 10.1126/stke.3842007pe21. [DOI] [PubMed] [Google Scholar]
  • 20.Fairfax BP, et al. Genetics of gene expression in primary immune cells identifies cell type-specific master regulators and roles of HLA alleles. Nat Genet. 2012;44:502–10. doi: 10.1038/ng.2205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lill CM, et al. Genome-wide significant association of ANKRD55 rs6859219 and multiple sclerosis risk. J Med Genet. 2013;50:140–3. doi: 10.1136/jmedgenet-2012-101411. [DOI] [PubMed] [Google Scholar]
  • 22.Maier LM, et al. IL2RA genetic heterogeneity in multiple sclerosis and type 1 diabetes susceptibility and soluble interleukin-2 receptor production. PLoS Genet. 2009;5:e1000322. doi: 10.1371/journal.pgen.1000322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Maller JB, et al. Bayesian refinement of association signals for 14 loci in 3 common diseases. Nat Genet. 2012;44:1294–301. doi: 10.1038/ng.2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.McLaren W, et al. Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics. 2010;26:2069–70. doi: 10.1093/bioinformatics/btq330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gregory AP, et al. TNF receptor 1 genetic risk mirrors outcome of anti-TNF therapy in multiple sclerosis. Nature. 2012;488:508–11. doi: 10.1038/nature11307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.De Jager PL, et al. The role of the CD58 locus in multiple sclerosis. Proc Natl Acad Sci U S A. 2009;106:5264–9. doi: 10.1073/pnas.0813310106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Malmestrom C, et al. Serum levels of LIGHT in MS. Mult Scler. 2012 doi: 10.1177/1352458512463766. [DOI] [PubMed] [Google Scholar]
  • 28.Dunham I, et al. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74. doi: 10.1038/nature11247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Schaub MA, Boyle AP, Kundaje A, Batzoglou S, Snyder M. Linking disease associations with regulatory information in the human genome. Genome Res. 2012;22:1748–59. doi: 10.1101/gr.136127.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Davydov EV, et al. Identifying a high fraction of the human genome to be under selective constraint using GERP++ PLoS Comput Biol. 2010;6:e1001025. doi: 10.1371/journal.pcbi.1001025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Johnson AD, et al. SNAP: a web-based tool for identification and annotation of proxy SNPs using HapMap. Bioinformatics. 2008;24:2938–9. doi: 10.1093/bioinformatics/btn564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Juran BD, et al. Immunochip analyses identify a novel risk locus for primary biliary cirrhosis at 13q14, multiple independent associations at four established risk loci and epistasis between 1p31 and 7q32 risk variants. Hum Mol Genet. 2012;21:5209–21. doi: 10.1093/hmg/dds359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Liu JZ, et al. Dense fine-mapping study identifies new susceptibility loci for primary biliary cirrhosis. Nat Genet. 2012;44:1137–41. doi: 10.1038/ng.2395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.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]
  • 35.Eyre S, et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat Genet. 2012;44:1336–40. doi: 10.1038/ng.2462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tsoi LC, et al. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nat Genet. 2012;44:1341–8. doi: 10.1038/ng.2467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cooper JD, et al. Seven newly identified loci for autoimmune thyroid disease. Hum Mol Genet. 2012;21:5202–8. doi: 10.1093/hmg/dds357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ban M, et al. Replication analysis identifies TYK2 as a multiple sclerosis susceptibility factor. Eur J Hum Genet. 2009;17:1309–13. doi: 10.1038/ejhg.2009.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ban M, et al. A non-synonymous SNP within membrane metalloendopeptidase-like 1 (MMEL1) is associated with multiple sclerosis. Genes Immun. 2010;11:660–4. doi: 10.1038/gene.2010.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Shah TS, et al. optiCall: a robust genotype-calling algorithm for rare, low-frequency and common variants. Bioinformatics. 2012;28:1598–603. doi: 10.1093/bioinformatics/bts180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.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]
  • 42.Browning BL, Yu Z. Simultaneous genotype calling and haplotype phasing improves genotype accuracy and reduces false-positive associations for genome-wide association studies. Am J Hum Genet. 2009;85:847–61. doi: 10.1016/j.ajhg.2009.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.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]
  • 44.Howie B, Marchini J, Stephens M. Genotype imputation with thousands of genomes. G3 (Bethesda) 2011;1:457–70. doi: 10.1534/g3.111.001198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Marchini J, Howie B. Genotype imputation for genome-wide association studies. Nat Rev Genet. 2010;11:499–511. doi: 10.1038/nrg2796. [DOI] [PubMed] [Google Scholar]
  • 46.Liu JZ, et al. Meta-analysis and imputation refines the association of 15q25 with smoking quantity. Nat Genet. 2010;42:436–40. doi: 10.1038/ng.572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164. doi: 10.1093/nar/gkq603. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

1
2

RESOURCES