Journal List > Allergy Asthma Immunol Res > v.11(2) > 1111452

Kim and Ober: Lessons Learned From GWAS of Asthma

Abstract

Asthma is a common complex disease of the airways. Genome-wide association studies (GWASs) of asthma have identified many risk alleles and loci that have been replicated in worldwide populations. Although the risk alleles identified by GWAS have small effects and explain only a small portion of prevalence, the discovery of asthma loci can provide an understanding of its genetic architecture and the molecular pathways involved in disease pathogenesis. These discoveries can translate into advances in clinical care by identifying therapeutic targets, preventive strategies and ultimately approaches for personalized medicine. In this review, we summarize results from GWAS of asthma from the past 10 years and the insights gleaned from these discoveries.

INTRODUCTION

Asthma is a heterogeneous and genetically complex respiratory disease.1 Approaches for gene discovery in asthma were initially candidate gene association studies, followed by family-based genome-wide linkage analyses and, most recently, genome-wide association studies (GWASs).23 For the last decade, GWASs of asthma have dominated, providing bias-free discovery of novel risk loci.4
The first GWAS of asthma was reported in 2007.5 As of July 10, 2018 there were 72 papers written in English on asthma or asthma-related traits reported in the GWAS catalog (https://www.ebi.ac.uk/gwas/). Among these 72 papers, 24 are GWASs of asthmatic subjects and controls, including 7 meta-analyses of asthma GWASs (Table 1); 5 are GWASs of asthma sub-phenotypes such as severe asthma or asthma exacerbations; 13 are GWASs of asthma-related traits such as bronchodilator response (BDR), airway hyperresponsiveness (AHR) and total serum immunoglobulin E (IgE) levels; 15 are GWASs of asthma combined with other diseases, such as allergic rhinitis, or factors such as smoking interaction or age of onset; 2 are GWASs of occupational asthma; 2 are GWASs of aspirin-exacerbated respiratory disease (AERD); and 11 are GWASs of asthma pharmacologic responses.
Table 1

Characteristics of GWASs of asthma

aair-11-170-i001
Year Author Discovery stage Replication stage Combined analysis Reference
Ethnicity Sample size Childhood onset asthma only No. of genome-wide significant loci* Ethnicity Sample size Childhood onset asthma only No. of replicated loci in genome-wide significant loci No. of genome-wide significant loci in combined analysis
2007 Moffatt MF European 994 cases and 1,243 controls Yes 1 European 5,621 subjects Yes 1 NA 5
2009 Hancock DB Latino 492 trios Yes 0 Hispanic 177 trios Yes NA NA 76
2009 Himes BE European 359 cases and 846 controls Yes 0 Multi-ethnic 24,155 subjects Yes NA NA 39
2010 Sleiman PM European 793 cases and 1,988 controls Yes 2 European, African American 6,175 subjects Yes 1 2 77
2010 Himes BE European 359 cases, 846 controls, and 403 trios Yes 0 Multi-ethnic 8,550 subjects and 583 trios No NA NA 78
2010 Mathias RA African American 498 cases and 500 controls No 0 African Caribbean, African American 6,134 subjects No NA 0 79
2010 DeWan AT Multi-ethnic 66 cases and 42 controls Yes 0 European, Hispanic 12,337 subjects No NA 0 80
2011 Ferreira MA European 986 cases and 1,846 controls No 0 European 604 subjects No NA NA 81
2011 Ferreira MA European 12,475 cases and 19,967 controls No 8§ European 25,358 subjects No NA 2 82
2011 Noguchi E Asian 938 cases and 2,376 controls Yes 2 Asian 3,106 subjects Yes 2 2 83
2011 Hirota T Asian 1,532 cases and 3,304 controls No 1 Asian 30,247 subjects No 0 5 84
2012 Lasky-Su J European 1,238 cases and 2,617 controls No 2 European 11,199 subjects No NA 1** 85
2012 Li X European 813 cases and 1,564 controls No 0 Multi-ethnic 41,400 subjects No NA NA 86
2014 Galanter JM Latino 1,893 cases and 1,881 controls Yes 1 Multi-ethnic 12,560 subjects No NA NA 87
2016 White MJ African American 812 cases and 415 controls Yes 1 NA NA NA NA NA 88
2016 Nieuwenhuis MA European 920 cases and 980 controls No 0 Multi-ethnic 11,656 subjects No NA 1 89
2016 Barreto-Luis A European 380 cases and 552 controls No 0 European 2,352 subjects No NA 0 90
2010 Moffatt MF†† European 10,365 cases and 16,110 controls No 7‡‡ NA NA NA NA NA 16
2011 Torgerson DG§§ Multi-ethnic 5,416 cases and 7,144 controls No 4∥∥ Multi-ethnic 12,649 subjects No 3¶¶ 3¶¶ 19
2012 Ramasamy A*** European 1,716 cases and 16,888 controls No 0 European 15,286 subjects No NA 2 91
2016 Pickrell JK European 28,399 cases and 128,843 controls No 27 NA NA NA NA NA 21
2017 Yan Q Latino 2,144 cases and 2,893 controls No 1 NA NA NA NA NA 92
2017 Almoguera B European, African 5,309 cases and 16,335 controls No 2 NA NA NA NA NA 34
2018 Demenais F††† Multi-ethnic 23,948 cases and 118,538 controls No 18 NA NA NA NA NA 14
References are sorted by year. “Mixed” in childhood onset asthma denotes the unknown proportion of childhood onset asthma.
NA, not applicable; GWAS, genome-wide association study; GABRIEL, Multidisciplinary Study to Identify the Genetic and Environmental Causes of Asthma in the European Community; SNP, single nucleotide polymorphism.
*Specifications of the discovery stage genome-wide significant P value definitions are in Supplementary Table S1; Replication data were shown in only the non-17q12-21 region; Both loci are also genome-wide significant in the discovery GWAS; §One loci from the results of the Australian GWAS only and seven loci from the results of the Australian GWAS and GABRIEL; Genome-wide significant P value of the replication stage was less than 5.0 × 10−8; From the adult asthma GWAS results only; **From the adult asthma combined analysis; ††Meta-Analysis includes GWAS from reference 5; ‡‡Loci including SNPs showing genome-wide significant association with asthma in at least one group using fixed models; §§Meta-Analysis includes GWAS from references 1939767779; ∥∥Loci including SNPs showing genome-wide significant association with asthma in at least one ethnic group; ¶¶Replication and combined analysis were done in selected 15 loci; ***Meta-Analysis includes GWAS from reference s19,39,83,85; †††Meta-Analysis includes GWAS from references 5197982838590.
In this review, we summarize the results of the 42 GWASs of asthma, asthma sub-phenotypes (e.g., severe asthma, asthma exacerbation) and asthma-related traits (e.g., BDR, AHR, total serum IgE) that are registered in the GWAS catalog. We discuss the challenges posed by GWASs of complex diseases and strategies to overcome these challenges. Other aspects of asthma genetics, such as gene-environment interactions,678 occupational asthma,9 AERD1011 or pharmacogenetics1213 are reviewed elsewhere.

GWAS OF ASTHMA

Table 1 summarizes the study populations, sample sizes, and results of the 17 GWASs and 7 meta-analyses of asthma. Additional information on characteristics of the study populations is included in Supplementary Table S1.
Eight GWASs and 6 meta-analyses reported one or more association with genome-wide significance in the discovery population. Two additional GWASs reported genome-wide significance in a combined — discovery and replication — sample. These 16 studies together described 35 loci that were significant in at least 1 study (Tables 2 and 3, Supplementary Tables S2 and S3). Sixteen of the 35 loci showed nominal significance when replicated in other GWASs, and 14 of those 16 loci showed genome-wide significant associations in at least 2 papers. Taken together, 5 GWASs and 5 meta-analyses of asthma identified genome-wide significant single nucleotide polymorphisms (SNPs) (P < 5 × 10−8) at the 17q12-21 (ORMDL3, GSDMB), making this the most widely replicated asthma loci. The 6p21 (HLA region), 2q12 (IL1RL1/IL18R1), 5q22 (TSLP) and 9p24 (IL33) loci showed the next 4 most genome-wide significant associations (Figure, Table 3).
Table 2

Asthma susceptibility loci meeting criteria for genome-wide significance in either discovery or combined stage in each GWAS

aair-11-170-i002
Year Author Region Reported genes Lead SNP Location (Bp) RAF in controls P value OR 95% CI Stage Replication P value Reference
2007 Moffatt MF* 17q21 ORMDL3 rs7216389 39913696 NA 1.00.E-10 NA NA Discovery 7.94.E-04 5
2010 Sleiman PM 1q31 DENND1B rs2786098 197356778 0.78 8.55.E-09 1.59 1.28–1.61 Discovery 6.47.E-04 77
17q21 ORMDL3/GSDMB rs4795400 39910767 NA 2.08.E-08 1.28 NA Discovery NA
2011 Ferreira MA†,‡ 1q21 IL6R rs4129267 154453788 0.40 2.30.E-08 1.09 1.06–1.12 Combined 3.30.E-03 82
2q12 IL1RL1 rs3771166 102369762 0.61 7.90.E-15 1.16 1.11–0.20 Discovery NA
5q22 WDR36 rs1043828 111128310 0.35 1.10.E-08 1.11 1.07–1.15 Discovery NA
5q31 RAD50 rs6871536 132634182 0.19 2.40.E-09 1.14 1.09–1.19 Discovery NA
9p24 IL33 rs1342326 6190076 0.16 3.50.E-14 1.20 1.14–1.26 Discovery NA
11q13 C11orf30/LRRC32 rs7130588 76559639 0.36 1.80.E-08 1.09 1.06–1.13 Combined 3.28.E-02
15q22 RORA rs11071559 60777789 0.86 3.80.E-09 1.18 1.11–1.23 Discovery NA
15q22 SMAD3 rs744910 67154447 0.49 2.70.E-09 1.11 1.07–1.15 Discovery NA
17q21 ORMDL3 rs8079416 39936460 0.44 2.40.E-22 1.19 1.15–1.23 Discovery NA
22q12 IL2RB rs2284033 37137994 0.57 5.00.E-10 1.12 1.09–1.16 Discovery NA
2011 Noguchi E§ 6p21 HLA-DPB1 rs987870 33075103 0.14 7.50.E-09 1.51 1.31–1.74 Discovery 1.20.E-02 83
8q24 SLC30A8 rs3019885 117013406 0.31 1.30.E-14 1.55 1.39–1.73 Discovery 8.70.E-03
2011 Hirota T 4q31 USP38 rs7686660 143082006 0.27 1.87.E-12 1.16 1.11–1.21 Combined 3.33.E-09 84
5q22 TSLP rs1837253 111066174 0.35 1.24.E-16 1.17 1.13–1.22 Combined 1.02.E-12
6p21 PBX2/NOTCH4/C6orf10/BTNL2/HLA-DRA/HLA-DQB1/HLA-DQA2/HLA-DOA rs404860 32216568 0.50 4.07.E-23 1.21 1.16–1.25 Combined 6.42.E-18
10p14 - rs10508372 8930055 0.43 1.79.E-15 1.16 1.12–1.21 Combined 1.31.E-11
12q13 CDK2/IKZF4 rs1701704 56018703 0.18 2.33.E-13 1.19 1.14–1.25 Combined 7.22.E-09
2012 Lasky-Su J 5p15 FLJ25076 rs272474 6462225 NA 3.78.E-08 NA NA Discovery NA 85
6p21 HLA-DQA1 rs9272346 32636595 NA 2.20.E-08 NA NA Combined 6.70.E-03
14q13 AKAP6 rs17441370 32775658 NA 1.37.E-11 NA NA Discovery NA
2014 Galanter JM 17q12 IKZF3 rs907092 39766006 0.70 5.70.E-13 1.49 1.33–1.64 Discovery NA 87
2016 White MJ 10p12 PTCHD3 rs660498 27452030 0.46 2.20.E-07 1.62 1.35–1.95 Discovery NA 88
2016 Nieuwenhuis MA 17q21 IKZF3/ZPBP2/GSDMB/ORMDL3 rs2290400 39909987 NA 2.55.E-20 1.31 NA Combined 6.78.E-17 89
Meta-analysis
2010 Moffatt MF 2q12 IL1RL2/IL1RL1/IL18R1/IL18RAP rs3771166 102369762 0.62 3.40.E-09 1.15 1.10–1.20 Discovery NA 16
6p21 CCHCR1/HLA-DQB1 rs9273349 32658092 0.58 7.00.E-14 1.18 1.13–1.24 Discovery NA
9p24 RANBP6/IL33 rs1342326 6190076 0.16 9.20.E-10 1.20 1.13–1.28 Discovery NA
15q22 SMAD3 rs744910 67154447 0.49 3.90.E-09 1.12 1.09–1.16 Discovery NA
17q12 STARD3/TCAP/PGAP3/ERBB2/IKZF3/ZPBP2 rs9303277 39820216 0.51 1.62.E-16 0.82 0.79–0.86 Discovery NA
17q21 GSDMB/ORMDL3 rs2305480 39905943 0.55 9.60.E-08 1.18 1.11–1.23 Discovery NA
17q21 GSDMA/PSMD3/MED24 rs3894194 39965740 0.45 4.60.E-09 1.17 1.11–1.23 Discovery NA
22q12 IL2RB rs2284033 37137994 0.56 1.20.E-08 1.12 1.08–1.16 Discovery NA
2011 Torgerson DG 2q12 IL1RL1 rs3771180 102337157 0.86 1.50.E-15 1.20 1.11–1.29 Combined 5.30.E-07 19
3q27 RTP2 rs2017908 187699930 0.13 4.42.E-09 1.63 1.43–1.82 Discovery 8.80.E-01
5q22 TSLP rs1837253 111066174 0.74 1.00.E-14 1.19 1.12–1.27 Combined 1.60.E-06
9p24 IL33 rs2381416 6193455 0.70 1.70.E-12 1.18 1.08–1.28 Combined 1.30.E-06
17q21 GSDMB rs11078927 39908152 0.55 2.20.E-16 1.27 1.20–1.34 Combined 1.50.E-08
2012 Ramasamy A 2q12 IL1RL1/IL18R1 rs13408661 102338622 0.84 1.00.E-09 1.23 1.15–1.31 Combined 3.20.E-05 91
6p21 BTNL2/HLA-DRA rs9268516 32411712 0.24 1.00.E-08 1.15 1.10–1.21 Combined 1.00.E-03
2016 Pickrell JK 1q23 ADAMTS4 rs4233366 161189357 NA 4.80.E-15 1.09 1.07–1.11 Discovery NA 21
1q24 CD247 rs1723018 167464183 NA 1.40.E-08 0.95 0.93–0.96 Discovery NA
1q25 TNFSF4 rs6691738 173182897 NA 2.90.E-08 0.94 0.92–0.96 Discovery NA
1q32 ADORA1 rs6683383 203131376 NA 1.10.E-08 1.06 1.04–1.08 Discovery NA
1p36 PEX14 rs662064 10497194 NA 3.20.E-08 0.94 0.92–0.96 Discovery NA
2q12 IL1RL1 rs202011557 102297183 NA 5.10.E-31 0.84 0.82–0.87 Discovery NA
2p25 - rs13412757 8317950 NA 1.30.E-08 1.06 1.04–1.08 Discovery NA
2q37 D2HGDH rs34290285 241759225 NA 1.80.E-15 1.11 1.08–1.14 Discovery NA
3q28 LPP rs73196739 188684683 NA 6.50.E-09 0.92 0.90–0.95 Discovery NA
4p14 TLR1 rs5743618 38797027 NA 3.90.E-11 1.08 1.06–1.11 Discovery NA
5q22 TSLP rs1837253 111066174 NA 3.30.E-31 0.88 0.86–0.90 Discovery NA
5q31 RAD50 rs2244012 132565533 NA 2.10.E-16 1.10 1.08–1.13 Discovery NA
5q31 NDFIP1 rs200634877 142150197 NA 2.50.E-08 0.94 0.92–0.96 Discovery NA
6q15 BACH2 rs58521088 90275479 NA 7.10.E-11 0.93 0.92–0.95 Discovery NA
6p21 HLA-DQA1 rs3104367 32635710 NA 1.00.E-40 0.87 0.86–0.89 Discovery NA
6p21 HLA-C/MICA rs2428494 31354420 NA 1.40.E-16 0.92 0.90–0.94 Discovery NA
7q22 CDHR3 rs6959584 106035809 NA 2.00.E-08 1.09 1.06–1.12 Discovery NA
8q21 - rs10957978 80372904 NA 1.10.E-11 0.93 0.92–0.95 Discovery NA
9p24 IL33 rs144829310 3208030 NA 1.30.E-31 1.17 1.14–1.20 Discovery NA
10p14 - rs12413578 9007290 NA 8.10.E-12 0.89 0.86–0.92 Discovery NA
11q13 C11orf30/LRRC32 rs7936323 76582714 NA 1.40.E-16 0.92 0.91–0.94 Discovery NA
12q13 STAT6 rs3001426 57115272 NA 1.40.E-10 0.94 0.92–0.96 Discovery NA
14q24 RAD51B rs3784099 68283210 NA 1.60.E-08 0.94 0.92–0.96 Discovery NA
15q22 - rs10519068 60776505 NA 3.80.E-11 1.10 1.07–1.13 Discovery NA
15q22 SMAD3 rs56375023 67156025 NA 2.40.E-21 0.90 0.88–0.92 Discovery NA
16p13 CLEC16A rs7203459 11136846 NA 3.50.E-15 1.09 1.07–1.12 Discovery NA
17q12 ZPBP2 rs11655198 39869916 NA 1.00.E-63 0.85 0.83–0.86 Discovery NA
2017 Yan Q 17q12 IKZF3 rs907092 39766006 0.68 1.16.E-12 1.41 NA Discovery NA 92
2017 Almoguera B 6p21 GRM4 rs1776883 34188667 0.47 5.29.E-09 1.25 1.19–1.31 Discovery NA 34
9p21 EQTN rs72721168 27308290 0.96 7.02.E-10 1.83 1.28–2.37 Discovery NA
2018 Demenais F** 2q12 IL1RL1 rs1420101 102341256 0.37 3.9.E-21 1.12 1.09–1.15 Discovery NA 14
5q22 SLC25A46 rs10455025 111069301 0.34 9.4.E-26 1.15 1.12–1.18 Discovery NA
5q31 IL13 rs20541 111069301 0.79 5.0.E-16 0.89 0.87–0.92 Discovery NA
5q31 NDFIP1 rs7705042 142112854 0.63 7.9.E-9 1.09 1.06–1.12 Discovery NA
6p21 HLA-DRB1 rs9272346 32636595 0.56 5.7.E-24 1.16 1.12–1.19 Discovery NA
6p21 MICB rs2855812 31504943 0.23 8.9.E-12 1.1 1.07–1.13 Discovery NA
6p22 GPX5 rs1233578 28744470 0.13 5.9.E-7 1.09 1.05–1.12 Discovery NA
6q15 BACH2 rs2325291 90276967 0.33 2.2.E-12 0.91 0.89–0.94 Discovery NA
8q21 TPD52 rs12543811 80366650 0.66 1.1.E-10 0.92 0.90–0.95 Discovery NA
9p24 RANBP6 rs992969 6209697 0.75 7.2.E-20 0.86 0.83–0.88 Discovery NA
10p14 GATA3 rs2589561 9004682 0.82 3.5.E-9 0.91 0.88–0.94 Discovery NA
11q13 EMSY rs7927894 76590272 0.37 2.2.E-14 1.1 1.08–1.13 Discovery NA
12q13 STAT6 rs167769 57109992 0.4 3.9.E-9 1.08 1.05–1.11 Discovery NA
15q22 RORA rs11071558 60777222 0.14 1.3.E-9 0.89 0.86–0.92 Discovery NA
15q22 SMAD3 rs2033784 67157322 0.3 7.4.E-15 1.1 1.08–1.13 Discovery NA
16p13 CLEC16A rs17806299 11106123 0.2 2.7.E-10 0.91 0.88–0.94 Discovery NA
17q12 ERBB2 rs2952156 39720582 0.7 2.2.E-30 0.87 0.84–0.89 Discovery NA
17q21 ZNF652 rs17637472 49384071 0.39 6.6.E-9 1.08 1.05–1.11 Discovery NA
The most significant SNPs at each locus are shown and ordered by genomic location in each reference. Base pair positions (bp) correspond to GRCh38/hg38 genome assembly.
SNP, single nucleotide polymorphism; RAF, risk allele frequency; OR, odds ratio; CI, confidence interval; FDR, false discovery rate; GWAS, genome-wide association study; GABRIEL, Multidisciplinary Study to Identify the Genetic and Environmental Causes of Asthma in the European Community.
*With the exception of the 17q12-21 locus, none of the markers below 5% FDR, after controlling for stratification, were within 1 Mb of each other; Discovery GWAS was the meta-analysis of results from the Australian GWAS and GABRIEL; RAF was from the Australian GWAS only; §RAF was from the discovery GWAS only; P value of random effects; P vaule from the Latino GWAS only; **RAF was allele effect frequency from the European GWAS only.
Table 3

Locus-level replications in subsequent GWAS

aair-11-170-i003
Reported genes Region The initial report Genome-wide significant replication, reference Nominal replication, reference
Strongest SNP P value Reference
STARD3/TCAP/PGAP3/ERBB2/IKZF3/ZPBP2/GSDMB/ORMDL3/GSDMA/ZNF652/PSMD3/MED24 17q12-21 rs7216389 1.00.E-10 5 141619217782878992 343978818385869091
CCHCR1/PBX2/NOTCH4/C6orf10/BTNL2/GRM4/HLA region/MICB/MICA 6p21 rs9273349 7.00.E-14 16 14213483848591 19828687888992
IL1RL2/IL1RL1/IL18R1/IL18RAP 2q12 rs3771166 3.40.E-09 16 1419218291 3481848586879092
TSLP/WDR36/SLC25A46 5q22 rs1043828 1.10.E-08 82 14192184 348586879092
IL33/RANBP6 9p24 rs1342326 9.20.E-10 16 14192182 193484858687899091
SMAD3/RORA 15q22 rs744910 3.90.E-09 16 142182 1983849192
RAD50/IL13/NDFIP1 5q31 rs6871536 2.40.E-09 82 1421 1934838490
C11orf30/LRRC32/EMSY 11q13 rs7130588 1.80.E-08 82 1421 90
IKZF4/CDK2/STAT6 12q13 rs1701704 2.33.E-13 84 1421 90
IL2RB 22q12 rs2284033 1.20.E-08 16 82 8384878992
BACH2 6q15 rs58521088 7.10.E-11 21 14 NA
TPD52 8q21 rs12543811 1.10.E-10 21 14 NA
GATA3 10p14 rs2589561 3.50.E-09 84 14 NA
CLEC16A 16p13 rs17806299 3.50.E-15 21 14 NA
DENND1B 1q31 rs2786098 8.55.E-09 77 NA 84
SLC30A8 8q24 rs3019885 5.00.E-13 83 NA 88
PEX14 1p36 rs662064 3.20.E-08 21 NA NA
IL6R 1q21 rs4129267 2.30.E-08 82 NA NA
ADAMTS4 1q23 rs4233366 4.80.E-15 21 NA NA
CD247 1q24 rs1723018 1.40.E-08 21 NA NA
TNFSF4 1q25 rs6691738 2.90.E-08 21 NA NA
ADORA1 1q32 rs6683383 1.10.E-08 21 NA NA
- 2p25 rs13412757 1.30.E-08 21 NA NA
D2HGDH 2q37 rs34290285 1.80.E-15 21 NA NA
RTP2 3q27 rs2017908 4.42.E-09 19 NA NA
LPP 3q28 rs73196739 6.50.E-09 21 NA NA
TLR1 4p14 rs5743618 3.90.E-11 21 NA NA
USP38 4q31 rs7686660 1.87.E-12 84 NA NA
FLJ25076 5p15 rs272474 3.78.E-08 85 NA NA
GPX5 6p22 rs1233578 5.90.E-07 14 NA NA
CDHR3 7q22 rs6959584 2.00.E-08 21 NA NA
EQTN 9p21 rs72721168 7.02.E-10 34 NA NA
PTCHD3 10p12 rs660498 2.20.E-07 88 NA NA
AKAP6 14q13 rs17441370 1.37.E-11 85 NA NA
RAD51B 14q24 rs3784099 1.60.E-08 21 NA NA
The table is sorted by the most number of repeatedly replicated loci. There were no replication data of previously reported GWAS in references 5767980. Nominal replication signifies the SNPs at each locus with replication P value less than 0.05 when there were replication data of previously reported GWASs.
GWAS, genome-wide association study; SNP, single nucleotide polymorphism.
Figure

Word cloud consisting of asthma risk genes from asthma GWASs (see Table 2 for references). Genes at genome-wide significant loci were selected based on the nearest gene. Word weight was assigned based on the number of times these genes were at loci that met the criteria for genome-wide significance. Word cloud was made using R package ‘wordcloud’ version 2.5. Figure drawn by H. Jang.

GWAS, genome-wide association study.
aair-11-170-g001
A recent meta-analysis of 23,948 asthma cases and 118,538 controls from the Trans-National Asthma Genetic Consortium (TAGC) revealed 18 loci that met the criteria of genome-wide significance,14 including nine previously known asthma loci, 2 loci previously reported for asthma plus hay fever, 2 previously associated with asthma in ancestry-specific populations and 5 new asthma susceptible loci. The latter included loci at 5q31.3, 6p22.1, 6q15, 12q13.3 and 17q21.33. Nearly all of the lead SNPs at the new loci were located in noncoding regions, and some were expression quantitative trait loci (eQTL) for genes such as NDFIP1 (chromosome 5q31.3), ZSCAN12 and ZSCAN31 (6p22.1), BACH2 (6q15), STAT6 (12q13.3) and GNGT2 (17q21.33). An enrichment in enhancer marks, especially in immune cells, was found at the associated loci, suggesting that the associated SNPs, or SNPs in linkage disequilibrium (LD) with the associated SNPs, play a role in the regulation of the immune processes.
Since the first GWAS of asthma that identified variants at the 17q21 locus and the correlation of those variants with expression of ORMDL3,5 this region has been the most frequently studied and replicated locus. This region harbors a dense haploblock of SNPs that overlap at least 4 genes: IKZF3, ZPBP2, GSDMB and ORMDL3. The locus has since been extended to include regions flanking this core region, implicating PGAP3 and ERBB2 at the proximal end and GSDMA at the distal end as potentially representing independent asthma loci.15 Nineteen asthma GWASs overall reported associations with SNPs at the extended 17q12-21 locus (Table 3). Moffatt et al.16 carried out a subgroup analysis of childhood-onset asthma and reported the association of this region specific to childhood-onset asthma, but had few later-onset asthma individuals to separately analyze that subgroup in their consortium-based meta-analysis of asthma GWASs. The TAGC meta-analysis of asthma GWAS also showed that the 17q12-21 locus centered on ORMDL3/GSDMB was specific to early-onset asthma, while that SNPs at the PGAP3/ERBB2 loci were not.14 They also suggested that the asthma-associated signals near the PGAP3/ERBB2 and ORMDL3/GSDMB blocks may affect asthma risk through the expression of different genes in different tissues.1415 Of note, the effects of genotype at this locus on asthma risk and protection have been reported to be modified by early-life exposures including environmental tobacco smoking17 and rhinovirus (RV)-associated wheezing in the first 3 years of life.18 Despite its strong and consistent association with asthma, there has been little evidence of association at this locus in African ancestry populations,1419 possibly owing to the breakdown of LD on African-derived chromosome.15 Taken together, SNPs in this locus are robustly associated with childhood-onset asthma in European, Asian and Latino individuals. Stein et al.15 recently reviewed studies of the 17q12-21 locus that showed the asthma-associated 17q12-21 SNPs are eQTLs for the GSDMA, ORMDL3, GSDMB and PGAP3 in immune cells and/or lung cells. However, the role of 17q12-21 genes in asthma pathogenesis is still unknown. An overview of functional studies of genes at the 17q12-21 locus was reviewed recently by Das et al.20
Among the approximately half of the published GWAS of asthma that did not identify any genome-wide significant associations in their discovery stage, most had sample sizes < 2,000 subjects (Table 1) suggesting that larger sample sizes (≥10,000) are needed to identify asthma associated loci. For example, the TAGC meta-analysis showed that pooling data from ethnically diverse populations including 23,948 asthma cases and 118,538 controls,14 and a 23andMe GWAS in 28,399 European ancestry cases and 128,843 controls21 each detected new asthma loci. Although very large studies increase clinical heterogeneity, many true asthma loci can be detected in very large samples.

GWAS OF ASTHMA SUB-PHENOTYPES AND INTERACTIONS

GWASs of asthma sub-phenotypes reduce heterogeneity and can lead to the identification of new asthma risk loci, even in smaller samples, due to increased power in studies of extreme or more homogeneous phenotypes. These studies may unveil genetic factors that are ‘masked’ in very large GWAS of more heterogeneous cases. For example, this is best illustrated by a GWAS of early childhood asthma with acute exacerbations leading to hospitalization and emergency department visit by Bønnelykke et al.22 The CDHR3 at 7q22.3 was identified in this study as a new susceptibility gene; this locus was later shown to be genome-wide significant in the 23andMe GWAS in European ancestry individuals,21 but not in the TAGC meta-analysis of ethnically diverse individuals.14 Importantly though, subsequent studies showed that CDHR3 functions as a receptor for Rhinovirus C (RV-C),23 and that the CDHR3 asthma risk allele was associated specifically with RV-C-related respiratory illnesses in the first 3 years of life.24 This “exacerbation GWAS” also confirmed previously reported asthma loci at genome-wide significance — GSDMB at 17q21, IL33 at 9p24, RAD50 at 5q31 and IL1RL1 at 2q12 loci, but with larger effect sizes despite the smaller sample size (Table 2), demonstrating that careful phenotyping and reduced clinical heterogeneity can reveal both novel asthma loci and larger effects of associated loci in smaller sample sizes than typically required for GWAS.
Another GWAS of exacerbations in 2 pediatric cohorts reported a novel asthma locus at the 10q21.3 (CTNNA3) that was genome-wide significant.25 A meta-analysis of GWASs that included both physician-diagnosed asthma and hay fever compared to controls with neither asthma nor hay fever revealed 2 novel susceptible loci: ZBTB10 at 8q21.13 and CLEC16A at 16p13.13.26 A GWAS of asthma with reduced exposure to tobacco smoke identified a locus that included the gene, HAS2 at 8q24.13, as a susceptibility locus,27 and another GWAS of active adult-onset nonallergic asthma added novel loci to asthma susceptible genes, CD200 at 3q13.2 and GRIK2 at 6q16.3, compared to inactive and mild nonallergic asthma.28 A GWAS that investigated the age of onset of childhood asthma, revealed loci on 3p26 and 11q24 that were associated with early-onset asthma and potentially to more severe disease.29 These GWASs of asthma defined by the presence or absence of other conditions identify novel loci, but most still require replication and functional characterizations.
Another approach to disentangle the complexity of asthma phenotypes and account for potential heterogeneity of risk factors have been genome-wide interaction studies (GWISs). A GWIS of genotype-by-sex interactions revealed a male-specific asthma risk locus, which includes IRF1 at 5q31.1, in European ancestry individuals, and a female-specific asthma risk locus, which included RAP1GAP1 at 1p36.12, in Latino individuals.30 The SNPs at these 2 loci showed only nominally significant associations with asthma in an independent GWAS, but emerged as sex-specific asthma risk loci when the effects of both genotype and sex as an interaction were taken into account. Another GWIS of farm-related exposures on asthma and atopy risk did not show any significant associations with either novel or previously reported asthma loci, likely due to low statistical power.31 Although this is a promising approach to identify loci that may confer risk only in the presence of specific exposures (i.e., gene-environment interactions), it is challenging to conduct these studies in the very large samples because exposures histories are rarely available in those samples.8
Finally, gene discovery in smaller samples may be possible using validated phenotyping algorithms that mine electronic medical records (EMRs). This approach has recently been developed as a tool for genomic research by the Electronic Medical Records and Genomics (eMERGE) network.3233 A GWAS of asthma in 5,309 cases and 16,335 controls recruited from eMERGE network identified novel loci of 6p21.31 (GRM4) and 9p21.2 (EQTN),34 although these associations need further replication and functional characterization. Within EMRs, longitudinal phenotype data and immense amounts of secondary phenotype data, such as laboratory findings and drug responses, can be collected. These data can be analyzed along with genetic data to determine whether loci are specific to asthma or shared with other allergic phenotypes, or how these relationships change over time. Rapid adoption of EMRs and EMR data standardization across hospitals will make available extensive phenotype data on many diseases and, combined with patient genotyping, expedite the identification of shared and unique genetic signatures for asthma endotypes as well as all common diseases.

GWAS OF ASTHMA-RELATED TRAITS

GWASs have been reported for asthma-related traits such as BDR, AHR, blood eosinophils, total serum IgE levels and allergic sensitization. The general assumptions of these studies are that it may be easier to find genes influencing components of asthma because they are less heterogeneous than asthma per se, and those same genes may also contribute to asthma risk and potentially provide more direct pharmacologic targets.
A GWAS of BDR — defined as the percentage change in FEV1 after administration of a short-acting β2-adrenergic receptor agonist — identified rare variants (frequency, <5%) near the solute carrier (SLC) genes with genome-wide significance in 1,782 Latino asthmatic children.35 Another GWAS of BDR revealed genome-wide significant variants near the ASB3 gene at 2p16 in a combined analysis of 1,164 multi-ethnic individuals with asthma.36 A GWAS of AHR severity — defined as the natural log of the dosage of methacholine causing a 20% drop in FEV1 — in 994 non-Hispanic white asthmatic subjects did not identify any genome-wide significant genes,37 while another GWAS of AHR severity in 650 European adult asthmatics revealed SNPs at the PDE4D gene at 5q11 at genome-wide significance,38 which is a previously reported asthma gene.39 Overall however, the BDR and AHR genes identified in GWAS with relatively small sample sizes lack replication. In contrast, a large GWAS of blood eosinophils,40 pleotropic multifunctional leukocytes that are involved in the pathogenesis of inflammatory diseases including asthma, in 21,510 European subjects (comprised of a discovery, n = 9,392, and replication, n = 12,118, sample) reported SNPs near the IL1RL1 at 2q12, IKZF2 at 2q34, GATA2 at 3q21.3, IL5 at 5q31.1 and SH2B3 at 12q24.12 genes with genome-wide significance. Among them, a variant at IL1RL1 was also associated with asthma in 10 different populations included in this study. IL1RL1 has been reported as an asthma gene through multiple GWAS of asthma (Tables 2 and 3). This finding requires further functional characterization if its relationship to eosinophils, asthma, and especially eosinophilic asthma, and its potential as a therapeutic target.
The first GWAS of total serum IgE levels, which is a strongly heritable trait,4142 did not show any genome-wide significant associations in the discovery population of 1,530 individuals of European ancestry. However, by combining the GWAS results with 4 independent replication cohorts, the investigators showed that functional variants near the gene encoding FCER1A at 1q23.2 and at the RAD50-IL13 locus at 5q31 were associated with total serum IgE levels at genome-wide significant thresholds in a combined analysis in of 11,299 individuals of European ancestry.43 The Multidisciplinary Study to Identify the Genetic and Environmental Causes of Asthma in the European Community (GABRIEL) consortium identified SNPs near HLA-DRB1 at 6p21 as an IgE-associated locus that was independent of associations of this locus with asthma, and confirmed the previously reported associations between total serum IgE levels and SNPs near the FCER1A, RAD50-IL13 and STAT6 loci, at genome-wide significant level.16 Three more GWAS of total serum IgE levels revealed loci near the HLA region reaching genome-wide significance;444546 the EVE consortium confirmed that these associations were shared among diverse ethnic groups.47 A GWAS of total serum IgE levels in 3,334 Latinos and a following admixture mapping in 454 Latinos, 1,564 European Americans and 3,187 African Americans revealed a locus near the ZNF365 gene at 10q21, but this finding still lacks replication.45 Furthermore, a meta-analysis of GWASs of allergic sensitization in 15,845 individuals of European ancestry and replication in 16,034 individuals of European ancestry identified 10 genome-wide significant loci in or near TLR6 at 4p14, C11orf30 at 11q13, STAT6 at 12q13, SLC25A46 at 5q22, HLA-DQB1 at 6p21, IL1RL1 at 2q12, LPP at 3q28, MYC at 8q24, IL2 at 4q27 and HLA-B at 6p21.48 A recent GWAS of allergic disease in 360,838 individuals considered individuals with asthma, hay fever and/or eczema.49 They identified 136 genome-wide significant risk variants at 99 independent loci, most of which had similar effects on the individual diseases, reflecting etiologic pathways that are common to all 3 diseases. However, this did not explicitly test for independent effects of the associated loci among individuals with only one of the three diseases. The shared loci were predicted to influence the function of immune cells and their target genes suggested opportunities for genomics-guided drug repositioning.

FUNCTIONAL STUDIES OF ASSOCIATED SNPs FROM EXISTING GWAS

A limitation of GWAS is that it identifies SNPs but does not provide information on the genes that the associated SNPs influence or on the causal SNP(s) among all SNPs in strong LD. As a result, nearly all GWASs report the nearest gene(s) as potential asthma candidate genes. However, not all SNPs impact the function or expression of the nearest gene, even when the SNP is within the gene itself. For example, among disease-associated variants that are eQTLs, the target gene will differ from the nearest gene 34% of the time.50 On the other hand, SNPs that are eQTLs are more likely to be among significant GWAS SNPs compared to SNPs that are not eQTLs,51 and combining eQTL mapping with GWAS can link GWAS-associated variants with the gene(s) they regulate, particularly if studies are performed in disease-relevant tissues.15 For example, Li et al.52 performed cis-eQTL studies in human bronchial epithelial cells (BECs) and cells from bronchial alveolar lavage (BAL) using SNPs near 34 putative asthma genes at 23 loci from previous GWASs. SNPs at 9 of the 23 loci were associated with the expression of nine genes in either BEC or BAL: IL1RL1 (but not IL18R1) at 2q12, TSLP (but not WDR36) at 5q22, HLA-DQB1 at 6p21, CDHR3 at 7q22, ZBTB10 at 8q21, IL33 at 9p24, C11orf30 (but not LRRC32) at 11q13, DEXI (but not CLEC16A) at 16p13, and GSDMB (but not ORMDL3) at 17q21. There are likely to be additional cis-eQTLs at asthma-associated SNPs at some of these loci in other tissues or by considering more SNPs or genes at each locus.
Ferreira et al.53 used a gene-based association test that integrated published asthma GWAS and eQTL mapping studies to identify SNPs that are eQTLs and the genes they are associating with. They used 16 published eQTL studies in 12 cell types or tissues potentially relevant to asthma: whole blood, lymphoblastoid cell lines, peripheral blood mononuclear cells, monocytes, B cells, T cells, neutrophils, spleen, lung, small airways, fibroblasts, skin. They suggested that asthma risk was associated with the expression of genes related to nucleotide synthesis (B4GALT3 at 1q23.3 and USMG5 at 10q24.33) and nucleotide-dependent cell activation (P2RY13 and P2RY14 at 3q25.1), and referred to these genes as putative novel asthma risk genes. They applied this method to their recent large GWAS of allergic disease,49 and identified additional significant and reproducible gene-based associations with 19 genes at 11 loci that were missed by single-variant analyses reported in the previous GWASs.54 Among these were nine genes with known functions relevant to allergic disease: FOSL2 at 2p23, VPRBP at 3p21, IPCEF1 at 6q25, PRR5L at 11p13, NCF4 at 22q12, and APOBR, IL27, ATXN2L and LAT at 16p11. These putative novel associations still need further replication. Luo et al.55 combined asthma GWAS results and publicly available eQTL data from human epithelial cells from small and large airways. They demonstrated that asthma GWAS hits were enriched as airway epithelial eQTLs and genes regulated by asthma GWAS loci in epithelium were enriched in immune response pathways. Li et al.,52 Ferreria et al.,53 and Luo et al.55 linked asthma-associated SNPs to genes they regulate, potentially elucidating molecular mechanisms for their associations with asthma.5355 A great boon to this type of approaches is the Genotype-Tissue Expression (GTEx) consortium, which has made available eQTL data for 44 human tissues that can be used to identify genes and pathways affected by human disease-associated variation.56

GWAS OF ASTHMA OR ASTHMA-RELATED TRAITS IN THE KOREAN POPULATION

In 2008, the first GWAS of an asthma phenotype in 347 Korean subjects (84 cases and 263 controls) was published for toluene diisocyanate (TDI)-induced asthma, an occupation-associated form of asthma.57 Since then, GWASs of asthma in Korea focused on 80,58 100,59 11760 and 17961 subjects with AERD, which is characterized by the development of bronchoconstriction in asthmatic patients after ingestion of non-steroidal anti-inflammatory drugs including aspirin. However, no genome-wide significant loci were reported in these GWASs, likely due to small sample sizes.
A GWAS of total serum IgE levels was reported in 877 Korean asthmatic patients without any genome-wide significant hits,62 but a GWAS of asthma in the Korean population has not yet been published. Performing GWASs of asthma in Korean children and adults is called for in the near future in order to identify the major genetic susceptibilities that maybe unique to this population.

ISSUES AND CHALLENGES IN GWAS OF ASTHMA

Despite their power for identifying asthma risk loci, there are many limitations of GWASs. In particular, GWASs identify mostly common variants which tend to have small effect sizes. As a result, GWAS-discovered variants are largely common (MAF > 10%) and account for a small proportion of both the population prevalence and the genetic component of asthma (i.e., the heritability).636465 These results in limited predictive power of these variants.6667 Although rare and low-frequency variants have potentially larger phenotypic effects, they have not explained a significant fraction of the ‘missingness’ of asthma heritability thus far.68 Recently, in a whole-genome sequencing study, Smith et al.69 found a rare loss of function mutation in IL33 that was associated with both lower blood eosinophils in 103,104 European subjects and reduced risk of asthma in 6,465 European asthmatic subjects and 302,977 controls. This study suggests that rare variants with large effect sizes are segregating in the population. While it is unlikely that such rare variants will account for significant proportions of the population risk for asthma, they can identify new pharmacologic targets and therefore serve a very important function.
Another limitation of GWAS is the statistical approach that tests for association with each of potentially tens of millions of SNPs. As a result, adjustments for multiple testing, typically using a Bonferroni corrected P value of <5 × 10−8 to control the false positive rate, require very large sample sizes (potentially >100,000) to identify loci with modest effect sizes. This stringent significance threshold will miss many true associations, particularly with SNPs involved in gene-gene and gene-environment interactions or those that are associated with specific asthma endotypes or sub-phenotypes. These variants have been referred to as ‘mid-hanging fruit’ in GWAS,7 and differentiating true from false associations among variants with small P values (e.g., <10−4) that do not meet genome-wide significance thresholds in GWASs remains a major challenge.
Another limitation has been that most GWAS and large meta-analyses of asthma and related traits are in subjects of European ancestry. Thus, most inferences about the genetic architecture of asthma is based on observations in this one continental population, whereas much less is known about Asian, African and admixed populations. Because populations vary with respect to allele frequencies, patterns of LD, and effect sizes of variants that underlie disease risk,707172 inferences based on Europeans may have limited utility in other groups. For example, next-generation sequencing studies revealed differences in allele frequencies and haplotype structures at the 17q12-21 asthma-locus between Chinese and other ethnic groups.73 However, half of the 24 asthma GWAS are only in Europeans (Table 1), and those studies are in general the largest GWAS to date. Moreover, until recently, commercial genotyping arrays were based on European allele frequencies and LD patterns. As a result, GWAS in non-European populations likely missed variants specific to those populations. This also impacts the selection of tag SNPs in replication studies in non-European populations. These issues have recently been addressed by the development of ethnic-specific and pan-ethnic genotyping arrays and publicly available genome sequences that allow for ethnic-specific imputation of genome-wide SNPs.74 For the first time, GWAS in Asian, Latino and African populations can be performed with excellent SNP coverage. It is crucial to study populations of diverse ethnic backgrounds for identifying shared and unique genetic predictors of asthma and for capturing more global patterns of genetic risk and gene-environment interaction effects on asthma risk.

CONCLUSIONS

Asthma pathogenesis is complex, resulting from heterogeneous genetic and environmental factors that jointly give rise to extensive phenotypic heterogeneity among asthmatics. Age at time of exposure to environmental risk factors and the persistence of these exposures during the lifespan may be critical modifiers of genotype-specific risk. These considerations are rarely, if ever, accounted for in GWAS. Nonetheless, the identification of susceptibility variants has already provided mechanistic insights into asthma pathogenesis, suggesting that asthma risk variants play a role in the regulation of immune cell functions.14 GWAS findings, considered together with deep learning approaches that can incorporate longitudinal EMR data,75 have the potential to more fully elucidate the genetic architecture of asthma. Such insights can be translated into advances in clinical care through identifying therapeutic targets and preventive approaches and ultimately personalized medicine.67

ACKNOWLEDGMENTS

The authors thanks H. Jang for helping with tables and figure. This work was supported by the Korea Research Foundation Grant funded by the Korean Government (NRF-2015R1D1A1A01061217) and by Institute for Information & communications Technology Promotion (IITP) grant funded by the Korea government (MSIT) (No. 2017-0-00599, Development of Big Data Analytics Platform for Military Health Information). C.O. is supported in part by NIH grants R01 HL129735, R01 HL122712, P01 HL070831, U19 AI106683, and R01 HL085197.

Notes

Disclosure There are no financial or other issues that might lead to conflict of interest.

References

1. Anderson GP. Endotyping asthma: new insights into key pathogenic mechanisms in a complex, heterogeneous disease. Lancet. 2008; 372:1107–1119.
crossref pmid
2. Los H, Koppelman GH, Postma DS. The importance of genetic influences in asthma. Eur Respir J. 1999; 14:1210–1227.
crossref pmid
3. Vercelli D. Discovering susceptibility genes for asthma and allergy. Nat Rev Immunol. 2008; 8:169–182.
crossref pmid
4. Hirschhorn JN, Daly MJ. Genome-wide association studies for common diseases and complex traits. Nat Rev Genet. 2005; 6:95–108.
crossref pmid
5. Moffatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S, et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature. 2007; 448:470–473.
pmid
6. Ober C, Vercelli D. Gene-environment interactions in human disease: nuisance or opportunity? Trends Genet. 2011; 27:107–115.
crossref pmid pmc
7. Ober C. Asthma genetics in the post-GWAS era. Ann Am Thorac Soc. 2016; 13:Suppl 1. S85–S90.
8. Bønnelykke K, Ober C. Leveraging gene-environment interactions and endotypes for asthma gene discovery. J Allergy Clin Immunol. 2016; 137:667–679.
crossref pmid pmc
9. Bernstein DI. Genetics of occupational asthma. Curr Opin Allergy Clin Immunol. 2011; 11:86–89.
crossref pmid
10. Park SM, Park JS, Park HS, Park CS. Unraveling the genetic basis of aspirin hypersensitivity in asthma beyond arachidonate pathways. Allergy Asthma Immunol Res. 2013; 5:258–276.
crossref pmid pmc
11. Dahlin A, Weiss ST. Genetic and epigenetic components of aspirin-exacerbated respiratory disease. Immunol Allergy Clin North Am. 2016; 36:765–789.
crossref pmid pmc
12. Davis JS, Weiss ST, Tantisira KG. Asthma pharmacogenomics: 2015 update. Curr Allergy Asthma Rep. 2015; 15:42.
crossref pmid
13. Park HW, Tantisira KG. Genetic signatures of asthma exacerbation. Allergy Asthma Immunol Res. 2017; 9:191–199.
crossref pmid pmc
14. Demenais F, Margaritte-Jeannin P, Barnes KC, Cookson WO, Altmüller J, Ang W, et al. Multiancestry association study identifies new asthma risk loci that colocalize with immune-cell enhancer marks. Nat Genet. 2018; 50:42–53.
pmid
15. Stein MM, Thompson EE, Schoettler N, Helling BA, Magnaye KM, Stanhope C, et al. A decade of research on the 17q12-21 asthma locus: piecing together the puzzle. J Allergy Clin Immunol. 2018; 142:749–764.e3.
crossref pmid pmc
16. Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, et al. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010; 363:1211–1221.
crossref pmid pmc
17. Bouzigon E, Corda E, Aschard H, Dizier MH, Boland A, Bousquet J, et al. Effect of 17q21 variants and smoking exposure in early-onset asthma. N Engl J Med. 2008; 359:1985–1994.
crossref pmid
18. Çalışkan M, Bochkov YA, Kreiner-Møller E, Bønnelykke K, Stein MM, Du G, et al. Rhinovirus wheezing illness and genetic risk of childhood-onset asthma. N Engl J Med. 2013; 368:1398–1407.
crossref pmid pmc
19. Torgerson DG, Ampleford EJ, Chiu GY, Gauderman WJ, Gignoux CR, Graves PE, et al. Meta-analysis of genome-wide association studies of asthma in ethnically diverse North American populations. Nat Genet. 2011; 43:887–892.
pmid pmc
20. Das S, Miller M, Broide DH. Chromosome 17q21 genes ORMDL3 and GSDMB in asthma and immune diseases. Adv Immunol. 2017; 135:1–52.
pmid
21. Pickrell JK, Berisa T, Liu JZ, Ségurel L, Tung JY, Hinds DA. Detection and interpretation of shared genetic influences on 42 human traits. Nat Genet. 2016; 48:709–717.
pmid pmc
22. Bønnelykke K, Sleiman P, Nielsen K, Kreiner-Møller E, Mercader JM, Belgrave D, et al. A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat Genet. 2014; 46:51–55.
pmid
23. Bochkov YA, Watters K, Ashraf S, Griggs TF, Devries MK, Jackson DJ, et al. Cadherin-related family member 3, a childhood asthma susceptibility gene product, mediates rhinovirus C binding and replication. Proc Natl Acad Sci U S A. 2015; 112:5485–5490.
crossref pmid pmc
24. Bønnelykke K, Coleman AT, Evans MD, Thorsen J, Waage J, Vissing NH, et al. Cadherin-related family member 3 genetics and rhinovirus C respiratory illnesses. Am J Respir Crit Care Med. 2018; 197:589–594.
crossref pmid pmc
25. McGeachie MJ, Wu AC, Tse SM, Clemmer GL, Sordillo J, Himes BE, et al. CTNNA3 and SEMA3D: promising loci for asthma exacerbation identified through multiple genome-wide association studies. J Allergy Clin Immunol. 2015; 136:1503–1510.
pmid pmc
26. Ferreira MA, Matheson MC, Tang CS, Granell R, Ang W, Hui J, et al. Genome-wide association analysis identifies 11 risk variants associated with the asthma with hay fever phenotype. J Allergy Clin Immunol. 2014; 133:1564–1571.
crossref pmid
27. Yatagai Y, Sakamoto T, Yamada H, Masuko H, Kaneko Y, Iijima H, et al. Genomewide association study identifies HAS2 as a novel susceptibility gene for adult asthma in a Japanese population. Clin Exp Allergy. 2014; 44:1327–1334.
28. Siroux V, González JR, Bouzigon E, Curjuric I, Boudier A, Imboden M, et al. Genetic heterogeneity of asthma phenotypes identified by a clustering approach. Eur Respir J. 2014; 43:439–452.
crossref pmid
29. Forno E, Lasky-Su J, Himes B, Howrylak J, Ramsey C, Brehm J, et al. Genome-wide association study of the age of onset of childhood asthma. J Allergy Clin Immunol. 2012; 130:83–90.e4.
crossref pmid pmc
30. Myers RA, Scott NM, Gauderman WJ, Qiu W, Mathias RA, Romieu I, et al. Genome-wide interaction studies reveal sex-specific asthma risk alleles. Hum Mol Genet. 2014; 23:5251–5259.
crossref pmid pmc
31. Ege MJ, Strachan DP, Cookson WO, Moffatt MF, Gut I, Lathrop M, et al. Gene-environment interaction for childhood asthma and exposure to farming in Central Europe. J Allergy Clin Immunol. 2011; 127:138–144. 144.e1–134.
crossref pmid
32. Newton KM, Peissig PL, Kho AN, Bielinski SJ, Berg RL, Choudhary V, et al. Validation of electronic medical record-based phenotyping algorithms: results and lessons learned from the eMERGE network. J Am Med Inform Assoc. 2013; 20:e147–54.
crossref
33. Gottesman O, Kuivaniemi H, Tromp G, Faucett WA, Li R, Manolio TA, et al. The Electronic Medical Records and Genomics (eMERGE) Network: past, present, and future. Genet Med. 2013; 15:761–771.
crossref pmid pmc
34. Almoguera B, Vazquez L, Mentch F, Connolly J, Pacheco JA, Sundaresan AS, et al. Identification of four novel loci in asthma in European American and African American populations. Am J Respir Crit Care Med. 2017; 195:456–463.
crossref pmid pmc
35. Drake KA, Torgerson DG, Gignoux CR, Galanter JM, Roth LA, Huntsman S, et al. A genome-wide association study of bronchodilator response in Latinos implicates rare variants. J Allergy Clin Immunol. 2014; 133:370–378.
crossref pmid
36. Israel E, Lasky-Su J, Markezich A, Damask A, Szefler SJ, Schuemann B, et al. Genome-wide association study of short-acting β2-agonists. A novel genome-wide significant locus on chromosome 2 near ASB3 . Am J Respir Crit Care Med. 2015; 191:530–537.
pmid pmc
37. Himes BE, Qiu W, Klanderman B, Ziniti J, Senter-Sylvia J, Szefler SJ, et al. ITGB5 and AGFG1 variants are associated with severity of airway responsiveness. BMC Med Genet. 2013; 14:86.
crossref
38. Nieuwenhuis MA, Vonk JM, Himes BE, Sarnowski C, Minelli C, Jarvis D, et al. PTTG1IP and MAML3, novel genomewide association study genes for severity of hyperresponsiveness in adult asthma. Allergy. 2017; 72:792–801.
pmid
39. Himes BE, Hunninghake GM, Baurley JW, Rafaels NM, Sleiman P, Strachan DP, et al. Genome-wide association analysis identifies PDE4D as an asthma-susceptibility gene. Am J Hum Genet. 2009; 84:581–593.
pmid pmc
40. Gudbjartsson DF, Bjornsdottir US, Halapi E, Helgadottir A, Sulem P, Jonsdottir GM, et al. Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Genet. 2009; 41:342–347.
pmid
41. Jacobsen HP, Herskind AM, Nielsen BW, Husby S. IgE in unselected like-sexed monozygotic and dizygotic twins at birth and at 6 to 9 years of age: high but dissimilar genetic influence on IgE levels. J Allergy Clin Immunol. 2001; 107:659–663.
crossref pmid
42. Strachan DP, Wong HJ, Spector TD. Concordance and interrelationship of atopic diseases and markers of allergic sensitization among adult female twins. J Allergy Clin Immunol. 2001; 108:901–907.
crossref pmid
43. Weidinger S, Gieger C, Rodriguez E, Baurecht H, Mempel M, Klopp N, et al. Genome-wide scan on total serum IgE levels identifies FCER1A as novel susceptibility locus. PLoS Genet. 2008; 4:e1000166.
44. Granada M, Wilk JB, Tuzova M, Strachan DP, Weidinger S, Albrecht E, et al. A genome-wide association study of plasma total IgE concentrations in the Framingham Heart Study. J Allergy Clin Immunol. 2012; 129:840–845.e21.
crossref pmid
45. Pino-Yanes M, Gignoux CR, Galanter JM, Levin AM, Campbell CD, Eng C, et al. Genome-wide association study and admixture mapping reveal new loci associated with total IgE levels in Latinos. J Allergy Clin Immunol. 2015; 135:1502–1510.
crossref pmid
46. Yatagai Y, Sakamoto T, Masuko H, Kaneko Y, Yamada H, Iijima H, et al. Genome-wide association study for levels of total serum IgE identifies HLA-C in a Japanese population. PLoS One. 2013; 8:e80941.
47. Levin AM, Mathias RA, Huang L, Roth LA, Daley D, Myers RA, et al. A meta-analysis of genome-wide association studies for serum total IgE in diverse study populations. J Allergy Clin Immunol. 2013; 131:1176–1184.
crossref pmid
48. Bønnelykke K, Matheson MC, Pers TH, Granell R, Strachan DP, Alves AC, et al. Meta-analysis of genome-wide association studies identifies ten loci influencing allergic sensitization. Nat Genet. 2013; 45:902–906.
pmid pmc
49. Ferreira MA, Vonk JM, Baurecht H, Marenholz I, Tian C, Hoffman JD, et al. Shared genetic origin of asthma, hay fever and eczema elucidates allergic disease biology. Nat Genet. 2017; 49:1752–1757.
pmid pmc
50. GTEx Consortium. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015; 348:648–660.
pmid pmc
51. Nicolae DL, Gamazon E, Zhang W, Duan S, Dolan ME, Cox NJ. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 2010; 6:e1000888.
crossref
52. Li X, Hastie AT, Hawkins GA, Moore WC, Ampleford EJ, Milosevic J, et al. eQTL of bronchial epithelial cells and bronchial alveolar lavage deciphers GWAS-identified asthma genes. Allergy. 2015; 70:1309–1318.
crossref pmid pmc
53. Ferreira MA, Jansen R, Willemsen G, Penninx B, Bain LM, Vicente CT, et al. Gene-based analysis of regulatory variants identifies 4 putative novel asthma risk genes related to nucleotide synthesis and signaling. J Allergy Clin Immunol. 2017; 139:1148–1157.
crossref pmid
54. Ferreira MA, Vonk JM, Baurecht H, Marenholz I, Tian C, Hoffman JD, et al. Eleven loci with new reproducible genetic associations with allergic disease risk. J Allergy Clin Immunol. 2018.
crossref
55. Luo W, Obeidat M, Di Narzo AF, Chen R, Sin DD, Paré PD, et al. Airway epithelial expression quantitative trait loci reveal genes underlying asthma and other airway diseases. Am J Respir Cell Mol Biol. 2016; 54:177–187.
crossref pmid pmc
56. GTEx Consortium. Genetic effects on gene expression across human tissues. Nature. 2017; 550:204–213.
57. Kim SH, Cho BY, Park CS, Shin ES, Cho EY, Yang EM, et al. Alpha-T-catenin (CTNNA3) gene was identified as a risk variant for toluene diisocyanate-induced asthma by genome-wide association analysis. Clin Exp Allergy. 2009; 39:203–212.
crossref pmid
58. Kim JH, Park BL, Cheong HS, Bae JS, Park JS, Jang AS, et al. Genome-wide and follow-up studies identify CEP68 gene variants associated with risk of aspirin-intolerant asthma. PLoS One. 2010; 5:e13818.
59. Shin SW, Park J, Kim YJ, Uh ST, Choi BW, Kim MK, et al. A highly sensitive and specific genetic marker to diagnose aspirin-exacerbated respiratory disease using a genome-wide association study. DNA Cell Biol. 2012; 31:1604–1609.
crossref pmid
60. Park BL, Kim TH, Kim JH, Bae JS, Pasaje CF, Cheong HS, et al. Genome-wide association study of aspirin-exacerbated respiratory disease in a Korean population. Hum Genet. 2013; 132:313–321.
crossref pmid
61. Kim SH, Cho BY, Choi H, Shin ES, Ye YM, Lee JE, et al. The SNP rs3128965 of HLA-DPB1 as a genetic marker of the AERD phenotype. PLoS One. 2014; 9:e111220.
62. Kim JH, Cheong HS, Park JS, Jang AS, Uh ST, Kim YH, et al. A genome-wide association study of total serum and mite-specific IgEs in asthma patients. PLoS One. 2013; 8:e71958.
crossref
63. Maher B. Personal genomes: the case of the missing heritability. Nature. 2008; 456:18–21.
crossref pmid
64. Thomsen SF, van der Sluis S, Kyvik KO, Skytthe A, Backer V. Estimates of asthma heritability in a large twin sample. Clin Exp Allergy. 2010; 40:1054–1061.
crossref pmid
65. Gibson G. Hints of hidden heritability in GWAS. Nat Genet. 2010; 42:558–560.
crossref pmid
66. Janssens AC, Gwinn M, Subramonia-Iyer S, Khoury MJ. Does genetic testing really improve the prediction of future type 2 diabetes? PLoS Med. 2006; 3:e114.
crossref
67. McCarthy MI, Abecasis GR, Cardon LR, Goldstein DB, Little J, Ioannidis JP, et al. Genome-wide association studies for complex traits: consensus, uncertainty and challenges. Nat Rev Genet. 2008; 9:356–369.
crossref pmid
68. Igartua C, Myers RA, Mathias RA, Pino-Yanes M, Eng C, Graves PE, et al. Ethnic-specific associations of rare and low-frequency DNA sequence variants with asthma. Nat Commun. 2015; 6:5965.
crossref pmid pmc
69. Smith D, Helgason H, Sulem P, Bjornsdottir US, Lim AC, Sveinbjornsson G, et al. A rare IL33 loss-of-function mutation reduces blood eosinophil counts and protects from asthma. PLoS Genet. 2017; 13:e1006659.
70. Li JZ, Absher DM, Tang H, Southwick AM, Casto AM, Ramachandran S, et al. Worldwide human relationships inferred from genome-wide patterns of variation. Science. 2008; 319:1100–1104.
crossref pmid
71. Shriner D, Adeyemo A, Gerry NP, Herbert A, Chen G, Doumatey A, et al. Transferability and fine-mapping of genome-wide associated loci for adult height across human populations. PLoS One. 2009; 4:e8398.
crossref
72. Baye TM, Butsch Kovacic M, Biagini Myers JM, Martin LJ, Lindsey M, Patterson TL, et al. Differences in candidate gene association between European ancestry and African American asthmatic children. PLoS One. 2011; 6:e16522.
crossref
73. Leung TF, Ko FW, Sy HY, Tsui SK, Wong GW. Differences in asthma genetics between Chinese and other populations. J Allergy Clin Immunol. 2014; 133:42–48.
crossref pmid
74. Spencer CC, Su Z, Donnelly P, Marchini J. Designing genome-wide association studies: sample size, power, imputation, and the choice of genotyping chip. PLoS Genet. 2009; 5:e1000477.
crossref
75. LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 2015; 521:436–444.
crossref pmid
76. Hancock DB, Romieu I, Shi M, Sienra-Monge JJ, Wu H, Chiu GY, et al. Genome-wide association study implicates chromosome 9q21.31 as a susceptibility locus for asthma in mexican children. PLoS Genet. 2009; 5:e1000623.
crossref
77. Sleiman PM, Flory J, Imielinski M, Bradfield JP, Annaiah K, Willis-Owen SA, et al. Variants of DENND1B associated with asthma in children. N Engl J Med. 2010; 362:36–44.
pmid
78. Himes BE, Lasky-Su J, Wu AC, Wilk JB, Hunninghake GM, Klanderman B, et al. Asthma-susceptibility variants identified using probands in case-control and family-based analyses. BMC Med Genet. 2010; 11:122.
crossref pmid pmc
79. Mathias RA, Grant AV, Rafaels N, Hand T, Gao L, Vergara C, et al. A genome-wide association study on African-ancestry populations for asthma. J Allergy Clin Immunol. 2010; 125:336–346.e4.
pmid
80. DeWan AT, Triche EW, Xu X, Hsu LI, Zhao C, Belanger K, et al. PDE11A associations with asthma: results of a genome-wide association scan. J Allergy Clin Immunol. 2010; 126:871–873.e9.
pmid pmc
81. Ferreira MA, McRae AF, Medland SE, Nyholt DR, Gordon SD, Wright MJ, et al. Association between ORMDL3, IL1RL1 and a deletion on chromosome 17q21 with asthma risk in Australia. Eur J Hum Genet. 2011; 19:458–464.
pmid
82. Ferreira MA, Matheson MC, Duffy DL, Marks GB, Hui J, Le Souëf P, et al. Identification of IL6R and chromosome 11q13.5 as risk loci for asthma. Lancet. 2011; 378:1006–1014.
pmid pmc
83. Noguchi E, Sakamoto H, Hirota T, Ochiai K, Imoto Y, Sakashita M, et al. Genome-wide association study identifies HLA-DP as a susceptibility gene for pediatric asthma in Asian populations. PLoS Genet. 2011; 7:e1002170.
84. Hirota T, Takahashi A, Kubo M, Tsunoda T, Tomita K, Doi S, et al. Genome-wide association study identifies three new susceptibility loci for adult asthma in the Japanese population. Nat Genet. 2011; 43:893–896.
crossref pmid pmc
85. Lasky-Su J, Himes BE, Raby BA, Klanderman BJ, Sylvia JS, Lange C, et al. HLA-DQ strikes again: genome-wide association study further confirms HLA-DQ in the diagnosis of asthma among adults. Clin Exp Allergy. 2012; 42:1724–1733.
pmid pmc
86. Li X, Ampleford EJ, Howard TD, Moore WC, Torgerson DG, Li H, et al. Genome-wide association studies of asthma indicate opposite immunopathogenesis direction from autoimmune diseases. J Allergy Clin Immunol. 2012; 130:861–868.e7.
crossref
87. Galanter JM, Gignoux CR, Torgerson DG, Roth LA, Eng C, Oh SS, et al. Genome-wide association study and admixture mapping identify different asthma-associated loci in Latinos: the Genes-environments & Admixture in Latino Americans study. J Allergy Clin Immunol. 2014; 134:295–305.
pmid pmc
88. White MJ, Risse-Adams O, Goddard P, Contreras MG, Adams J, Hu D, et al. Novel genetic risk factors for asthma in African American children: Precision Medicine and the SAGE II Study. Immunogenetics. 2016; 68:391–400.
crossref pmid pmc
89. Nieuwenhuis MA, Siedlinski M, van den Berge M, Granell R, Li X, Niens M, et al. Combining genomewide association study and lung eQTL analysis provides evidence for novel genes associated with asthma. Allergy. 2016; 71:1712–1720.
crossref pmid pmc
90. Barreto-Luis A, Pino-Yanes M, Corrales A, Campo P, Callero A, Acosta-Herrera M, et al. Genome-wide association study in Spanish identifies ADAM metallopeptidase with thrombospondin type 1 motif, 9 (ADAMTS9), as a novel asthma susceptibility gene. J Allergy Clin Immunol. 2016; 137:964–966.
pmid
91. Ramasamy A, Kuokkanen M, Vedantam S, Gajdos ZK, Couto Alves A, Lyon HN, et al. Genome-wide association studies of asthma in population-based cohorts confirm known and suggested loci and identify an additional association near HLA. PLoS One. 2012; 7:e44008.
crossref
92. Yan Q, Brehm J, Pino-Yanes M, Forno E, Lin J, Oh SS, et al. A meta-analysis of genome-wide association studies of asthma in Puerto Ricans. Eur Respir J. 2017; 49:1601505.

SUPPLEMENTARY MATERIALS

Supplementary Table S1

Characteristics of the study populations of GWAS of asthma

Supplementary Table S2

Asthma susceptibility SNPs that met criteria for genome-wide significance in either discovery or combined stage in each GWAS

Supplementary Table S3

Asthma susceptibility SNPs that met criteria for genome-wide significance in meta-analyses
TOOLS
ORCID iDs

Kyung Won Kim
https://orcid.org/0000-0003-4529-6135

Similar articles