A meta-analysis of genome-wide association studies of childhood wheezing phenotypes identifies ANXA1 as a susceptibility locus for persistent wheezing ====================================================================================================================================================== * R Granell * JA Curtin * S Haider * N Kitaba * S Mathie * L Gregory * LL Yates * M Tutino * J Hankinson * M Perretti * JM Vonk * SH Arshad * P Cullinan * S Fontanella * G Roberts * GH Koppelman * A Simpson * S Turner * CS Murray * CM Lloyd * JW Holloway * A Custovic * UNICORN Investigators * Breathing Together Investigators ## ABSTRACT **Background** Many genes associated with asthma explain only a fraction of its heritability. Most genome-wide association studies (GWASs) used a broad definition of “doctor-diagnosed asthma”, thereby diluting genetic signals by not considering asthma heterogeneity. The objective of our study was to identify genetic associates of childhood wheezing phenotypes. **Methods** We conducted a novel multivariate GWAS meta-analysis of wheezing phenotypes jointly derived using unbiased analysis of data collected from birth to 18 years in 9,568 individuals from five UK birth-cohorts. **Results** 44 independent SNPs were associated with early-onset persistent, 25 with preschool remitting, 33 with mid-childhood remitting and 32 with late onset wheeze. We identified a novel locus on chr9q21.13 (close to annexin 1 (*ANXA1*), p<6.7×10−9), associated exclusively with early-onset persistent wheeze. We identified rs75260654 as the most likely causative single nucleotide polymorphism (SNP) using Promoter Capture Hi-C loops, and then showed that the risk allele (T) confers a reduction in *ANXA1* expression. Finally, in a murine model of house dust mite (HDM)-induced allergic airway disease, we demonstrated that anxa1 protein expression increased and anxa1 mRNA was significantly induced in lung tissue following HDM exposure. Using anxa1−/− deficient mice, we showed that loss of anxa1 results in heightened airway hyperreactivity and Th2 inflammation upon allergen challenge. **Conclusions** We discovered a novel locus uniquely associated with early-onset persistent wheeze, identified the most likely causative variant, and showed that *ANXA1* may play a role in regulating the pulmonary immune response to allergens. Targeting this pathway in persistent disease may represent an exciting therapeutic prospect. Keywords * GWAS * asthma * wheeze phenotypes * Annexin 1 ## INTRODUCTION Asthma is a complex disorder caused by a variety of mechanisms which result in multiple clinical phenotypes(1). It has a strong genetic component, and twin studies estimate its heritability to be ∼ 60-70%(2). “Asthma genes” have been identified through a range of approaches, from candidate gene association studies(3) and family-based genome-wide linkage analyses(4) to genome-wide association studies (GWASs)(5-7). The first asthma GWAS (2007) identified multiple markers on chromosome 17q21 associated with childhood-onset asthma(5). A comprehensive review summarising the results of 42 GWASs of asthma and asthma-related traits has been published recently(8). The most widely replicated locus is 17q12-21, followed by 6p21 (*HLA* region), 2q12 (*IL1RL1/IL18R1*), 5q22 (*TSLP*) and 9p24 (*IL33*)(9). Overall, the evidence suggests that multiple genes are underlying the association peaks(9). However, despite undeniable successes, genetic studies of asthma have produced relatively heterogeneous results, and only a small proportion of the heritability is accounted for(10). One part of the explanation for the paucity of precise replication are numerous gene-environment interactions(11). Another important consideration is asthma heterogeneity, in that asthma diagnosis comprises several conditions with distinct pathophysiology(12, 13), each potentially underpinned by different genetic associations(14). However, in order to maximise sample size, most GWASs used a definition of “doctor-diagnosed asthma”(15). Such aggregated outcome definitions are imprecise(16) and phenotypically and mechanistically heterogeneous(17), and this heterogeneity may dilute important genetic signals(14). One way of disaggregating asthma diagnosis is to use data-driven methods to derive subtypes in a hypothesis-neutral way(18). For example, we jointly modelled data on wheezing from birth to adolescence in five UK population-based birth cohorts and identified five distinct phenotypes(19). However, although latent modelling approaches have been instrumental in elucidating the heterogenous nature of childhood asthma diagnosis(13), there has been little research into the genetic associations of phenotypes derived using data-driven methods. This is the first study to investigate the genetic architecture of wheezing phenotypes from infancy to adolescence, to identify genes specific to each phenotype and better understand the genetic heterogeneity between the disease class profiles. ## MATERIALS & METHODS ### Study design, setting, participants and data sources/measurement The Study Team for Early Life Asthma Research (STELAR) consortium(20) brings together five UK population-based birth cohorts: Avon Longitudinal Study of Parents and Children (ALSPAC)(21), Ashford(22) and Isle of Wight (IOW)(23) cohorts, Manchester Asthma and Allergy Study (MAAS)(24) and the Aberdeen Study of Eczema and Asthma to Observe the Effects of Nutrition (SEATON)(25). The cohorts are described in detail in the **Online Supplement** (OLS). All studies were approved by research ethics committees. Informed consent was obtained from parents, and study subjects gave their assent/consent when applicable. Validated questionnaires were completed on multiple occasions from infancy to adolescence(19). A list of variables, per cohort, is shown in **Table E1**, and the cohort-specific time points and sample sizes in **Table E2**. Data were harmonised and imported into Asthma eLab web-based knowledge management platform to facilitate joint analyses(20). ### Definition of primary outcome (wheeze phenotypes from infancy to adolescence) In the pooled analysis among 15,941 subjects with at least two observations on current wheeze, we used latent class analysis (LCA) to derive wheeze phenotypes from birth to age 18 years(19). A detailed description of the analysis is presented in(19) and in the OLS. A five-class solution was selected as the optimal model(19), and the classes (wheeze phenotypes) were labeled as: (1) *never/infrequent wheeze* (52.4%); (2) *early-onset pre-school remitting wheeze* (18.6%); (3) *early-onset middle-childhood remitting wheeze* (9.8%); (4) *early-onset persistent wheeze* (10.4%); and (5) *late-onset wheeze* (8.8%). These latent classes were used in the subsequent GWAS. ### Genotyping, imputation and GWAS Meta-Analysis Genotyping, quality control, imputation and exclusions are described in the OLS. Analyses were performed independently in ALSPAC, MAAS and the combined IOW-SEATON-ASHFORD (genotyped on the same platform, at the same time, and imputed together). We used SNPTEST v2.5.2(26) with a multinomial logistic regression model (-method newml), using the never/infrequent wheeze as the reference. A meta-analysis of the three GWASs was performed using METAL(27) with a total of 8,057,852 single nucleotide polymorphism (SNPs). LD pruning, pre-selection and gene annotation is described in the OLS. ### Post-GWAS studies Our GWAS identified a novel locus in chr9q21 nearby *Annexin A1* (*ANXA1*), exclusively associated with early-onset persistent wheeze (see results section). We therefore proceeded with studies to identify causal variants and explore the biological mechanisms underlying this locus (detail in OLS). To this end, we firstly identified the most likely causative SNP using Promoter Capture Hi-C loops. We then ascertained genotype effect on gene expression and assessed the potential biological function of *ANXA1* in asthma. Finally, we used a murine model of house dust mite (HDM)-induced allergic airway disease to investigate whether *ANXA1* was important in regulating immune responses to a clinically relevant aeroallergen and used knock-out mice to derive further *in vivo* functional data to support our GWAS-finding. ## RESULTS ### Participants and descriptive data We included a total of 9,568 subjects with European ancestry: ALSPAC, n=6833; MAAS, n=887; SEATON, n=548; ASHFORD, n=348; and IOW, n=952. Demographic characteristics of the study population(s) are shown in **Table E3**. Comparison of included vs. excluded participants across cohorts (per cohort and time point) is shown in **Table E4**. ### GWAS Meta-Analysis We conducted three GWASs (ALSPAC, MAAS, IOW-SEATON-Ashford) in parallel and results were meta-analyzed. QQ-plots are shown in **Figure E1**. A circular Manhattan plot showing an overview of the GWAS results by wheeze phenotype is shown in **Figure 1**. A total of 589 SNPs were associated with at least one phenotype with p<10−5. After clumping, we identified 134 independent SNPs uniquely associated with different phenotypes (p<10−5): of these, 44 were associated with early-onset persistent, 25 with early-onset preschool remitting, 33 with early-onset mid-childhood remitting and 32 with late-onset wheeze (**Table E5**). For some SNPs there were nominal associations with more than one phenotype. For example, chr17q21 was identified as a top locus for early-onset persistent wheeze (p=5.42×10−9), but some of the SNPs in this region were also associated with the early-onset mid-childhood remitting phenotype (p<0.0001). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F1) Figure 1. Circular Manhattan plot showing an overview of the GWAS results by wheeze phenotype (from outside to inside: early-onset persistent, early-onset pre-school remitting, early-onset mid-childhood remitting and late-onset wheeze). The red line indicates the genome-wide significance threshold (P < 5 × 10−8), while the blue line indicates the threshold for genetic variants that showed a suggestive significant association (P < 10−5). To help identify functional elements located near the GWAS-associated variants (potential causal variants), we used locus zoom plots (LZP) for the 134 independent SNPs (p<10−5). Following close inspection of all plots, we short-listed 85 independent SNPs for which the LZPs potentially indicated more than one causal variant (**Figures E2-E5**) and followed them up for further annotation. The results of GWAS meta-analysis for these 85 SNPs with main associations across the four wheeze phenotypes are presented in **Table 1**. Previously associated traits for each region/gene associated with the different wheeze phenotypes are shown in **Table E6**. Briefly, one region (6q27) among the top hits for early-onset preschool remitting wheeze was previously associated with asthma, but in the context of obesity with a nominal association with asthma and BMI(28). Another region/gene (3q26.31/*NAALADL2*) identified as top hit for early-onset preschool remitting wheeze, was reported as an associate of severe asthma exacerbations, but only at nominal level (29). No regions/genes identified as top hits for early-onset mid-childhood remitting wheeze were found to have previous associations with asthma. Several genes/loci identified as top hits for late-onset wheeze were previously associated with asthma: *ACOXL* chr2q13 (later onset asthma and obesity(30)), *PRKAA2* chr1p32.2 (lymphocyte count and asthma susceptibility(31)), *CD200* 3q13.2 (adult onset non-allergic asthma(32)), *GIMAP* family 7q36.1 (autoimmune diabetes, asthma, allergy(33)), 9p22.3 (asthma in <16 year-old (34)) and *16p12.1* (asthma and rhino-conjunctivitis at 10-15 years(35)). View this table: [Table 1.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T1) Table 1. GWAS Meta-analysis: Short-listed 85 top independent SNPs across the four wheezing phenotypes We identified two GWAS-significant loci for early-onset persistent wheeze: 17q21, p<5.5×10−9, and a novel locus on 9q21.13 (*ANXA1*), p<6.7×10−9. The *ANXA1* locus was the only GWAS-significant locus that had not previously been associated with asthma or atopic traits, with one previous study showing an association with FEV1/FVC and bronchodilator response in smokers(36). *ANXA1* is strongly expressed in bronchial mast cells and has anti-inflammatory properties(37), and may be involved in epithelial airway repair(38) **(Table E6**). We therefore followed up top SNPs from this locus. ### *ANXA1* locus and persistent wheeze Two SNPs (rs75260654, the lead SNP, and rs116849664 located downstream of *ANXA1*) were associated with early-onset persistent wheeze at genome-wide significance (GWS), with an additional SNP rs78320984 almost reaching GWS (**Table E7**). These SNPs are in LD with each other (**Figure E6**), but not with any other SNPs. #### Promoter Capture identifies rs75260654 as the most likely causative variant To identify the most likely causative variant, we investigated the overlap of the SNPs with Promoter Capture Hi-C interactions involving the *ANXA1* promoter in CD4+ cells in MAAS cohort subjects. Of the three SNPs, only rs75260654 overlapped a region interacting with the *ANXA1* promoter (**Figure 2**). Moreover, rs75260654 overlapped a *POLR2A* ChIP-seq peaks and an ATAC-seq peak and active enhancer in the type II pneumocyte derived A549 cell line. This shows that rs75260654 is located in a region directly interacting with the *ANXA1* promoter and is transcriptionally active in relevant cell types. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F2) Figure 2. Chromatin interactions between rs75260654 and the ANXA1 promoter in CD4+ cells in MAAS *rs75260654* physically interacts with *ANXA1* promoter in CD4+ T-cells and overlaps a region of active (POLR2AphosphoS2 ChIP-seq) open (ATAC-seq) chromatin in A549 cell line (lung epithelial carcinoma). The region is also predicted to be an active enhancer (ChromHMM 18-state model) in the A549 cell type. Only ChromHMM enhancer chromatin are displayed. Yellow shaded area indicates the PCHi-C fragment overlapping rs75260654 (red bar) and interacting with the *ANXA1* promoter. Allele Frequencies of rs75260654 (MAF=0.02) across wheeze phenotypes are shown in **Table E8**. Two individuals (one in MAAS and one in ALSPAC) were homozygote for the minor allele (T), and both were in the early-onset persistent wheeze class. One subject reported current wheeze and asthma through childhood, with hospitalizations for lower respiratory tract infection in the 1st year of life confirmed in healthcare records. The second individual reported current wheezing at 1.5, 2.5 and 8-9 years and doctor-diagnosed asthma and the use of asthma medication at 8-9 years. #### rs75260654: Effect on Genomic Features VEP prediction shows the SNP rs75260654 (C changed to T) to be located downstream of three protein-coding transcripts of *AXNA1* and overlapping the known regulatory region ID ENSR00000882742 on Chromosome 9: 73,173,001-73,173,200. This region is active in the GI tract, M2 macrophages, neural progenitor cells, and trophoblasts, but is repressed in T lymphocytes including CD4+ CD25+, Treg, and CD8+ cells. #### rs75260654: Effect on gene expression The effect of rs75260654 on the expression of nearby genes was investigated by browsing the eQTL GTEX data available in Ensembl. Compared to C, the T allele was found to reduce the expression of *ANXA1* in naive B-cells (effect size=-2.36795, p=0.01) and to increase expression in Lymphoblasoid Cell Lines (LCL) (effect size=0.848856, pe=0.001) (**Figure 3)**. This SNP affects expression of the neighboring gene *ALDH1A1* (aldehyde dehydrogenase-1 family member A1) (effect size=-2.40446, p=0.0039 in macrophages infected with Salmonella). In the eQTL catalogue, rs75260654 is identified as an eQTL of *ANXA1* in various immune cells (at nominal significance) including T cells, monocytes, fibroblasts, whole blood, Th2 memory cells, naive B cells. rs75260654 is also an eQTL of *ANXA1* in monocytes that were stimulated with R848 (agonist of TLRs 7 and 8) and a human seasonal influenza A virus (39) (at nominal significance) (**Table E9**). In the lung rs116849664 and rs78320984 (both in LD with rs75260654) were eQTLs of *ANXA1* (**Table E10**) as well as LINC01474 at nominal significance levels. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F3) Figure 3. eQTL ANXA1 and rs75260654 across different tissue types. Point size is proportional to -log10 p-value. Additional supporting evidence regarding the significance of the T-allele on the expression of these genes was provided using eQTLGene Consortium meta-analysis of 24 Cohorts and 24331 samples(40). This method reproduced the previous modest results showing a cis-eQTL effect of rs75260654 on both the *ANXA1* (p=6.02×10−23) and *ALDH1A1* (p=1.11×10−19) at FDR=0. No significant trans-eQTLs were observed. #### Potential biological function of ANXA1 in asthma Protein-protein network analysis demonstrated that *ANXA1* interacts directly with genes enriched for asthma (including *IL4* and *IL13*) and inflammatory regulation (*NR3C1*, Glucocorticoid receptor) showing its significance in dysregulation of the immune response (see **Figure E7** and **Table E11**). ### Functional studies of *anxa1* in a murine model #### Pulmonary expression of anxa1 is modulated by aeroallergen exposure We first analysed expression of *anxa1* using a model of HDM-induced allergic airway disease (**Figure 4A**)(41). Consistently, immunohistochemistry analysis revealed anxa1 protein expression increased following HDM challenge (**Figure 4B,C**). Anxa1 mRNA was significantly induced in lung tissue following HDM exposure (**Figure 4D**). This increase suggests that the pro-resolving *anxa1* may play a role in regulating the pulmonary immune response to allergen. ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F4) Figure 4. Annexin A1 is induced following HDM challenge and mice deficient in ANXA1 have exacerbated airway hyperreactivity. (A) Schematic of house dust mite allergen dosing protocol, N=4-6 per group, data representative of two animal experiments. (B, C) Immunofluorescent staining of paraffin embedded lung tissue sections incubated with anti-Annexin A1, counterstained with DAPI (N=4 per group). (D) mRNA expression of annexin A1 in lung tissue following HDM exposure, expression normalized to housekeeping gene hprt. Mice receiving HDM were analysed for changes in airway hyper-reactivity following methacholine (MCh) challenge in tracheotomized restrained mice. (E) Airway resistance at top MCh dose 100mg/ml (F) Eosinophils quantified in BAL, (F) T1/ST2+ lymphocytes quantified in the BAL. (H) IL-4 (I) IL-5 and (J) IL-13 quantified in lung tissue by ELISA. * p<0.05 and ** p<0.01 relative to PBS control group by Mann Whitney test. + p<0.05 and ++ p<0.01 comparing HDM AnnexinA1 KO mice relative to HDM WT group by Mann Whitney test. #### Anxa1 suppresses allergen-induced airway hyperresponsiveness (AHR) and type-2 inflammation To confirm a functional role for *anxa1* in allergic airway disease, we exposed *anxa1−/−* mice to intranasal HDM. Wildtype mice given HDM over 3 weeks developed significant AHR compared to PBS control mice. Mice deficient in *anxa1* had significantly worse allergen-induced lung function (greater airway resistance) compared to WT treated mice (**Figure 4E**). *Anxa1−/−* mice exhibited significantly increased airway eosinophilia and elevated numbers of Th2 lymphocytes (**Figure 4F,G**). Lung tissue cytokine levels reflected the exacerbated airway Th2 inflammation, with elevation in IL-4, and significant induction of IL-5 and IL-13 (**Figure 4H,J**). Thus, *anxa1* deficiency results in an alteration of the pulmonary immune response, with uncontrolled eosinophilia and an exacerbation of type-2 inflammation and AHR in response to allergen. ## DISCUSSION Herein, we present a comprehensive description of the genetic architecture of childhood wheezing disorders. Using a novel approach applied to a unique dataset from five UK birth cohorts, we identified subsets of SNPs differentially associated across four wheezing phenotypes: early-onset persistent (44 SNPs, 19 loci), early-onset preschool remitting (25 SNPs, 10 loci), early-onset mid-childhood remitting (33 SNPs, 9 loci) and late-onset (32 SNPs, 20 loci). We found little evidence of genetic associations spanning across different phenotypes. This suggests that genetic architecture of different wheeze phenotypes comprises a limited number of variants likely underpinning mechanisms which are shared across phenotypes, but that each phenotype is also characterized by unique phenotype-specific genetic associations. Importantly, we identified a novel locus in chr9q21 nearby *ANXA1* exclusively associated with early-onset persistent wheeze (p<6.7×10−9). To identify the most likely causative variant, we investigated the overlap of the associated SNPs with Promoter Capture Hi-C interactions to demonstrate that SNP rs75260654 overlapped a region interacting with the *ANXA1* promoter. Using eQTL data, we identified that the risk allele (T) o rs75260654 associated with early-onset persistent wheeze is also associated with *ANXA1* expression. Further investigation of the biological function of *ANXA1* revealed that it interacts with genes enriched for asthma (including *IL4* and *IL13*) and inflammatory regulation (*NR3C1*, Glucocorticoid receptor). In functional mouse experiments, anxa1 protein expression increased and anxa1 mRNA was significantly induced in lung tissue following HDM exposure, suggesting that the pro-resolving anxa1 may play a role in regulating the pulmonary immune response to allergen. Concurrently, by utilizing *anxa1−/−* deficient mice we demonstrated that loss of anxa1 results in heightened AHR and Th2 inflammation upon allergen challenge, providing important *in vivo* functional data to support our GWAS-finding. *ANXA1* is a 37-kDa glycoprotein with potent anti-inflammatory and pro-resolving properties that are mediated by interaction with a specific G-protein coupled receptor FPR2(42). This axis represents an important resolution pathway in chronic inflammatory settings such as those of rheumatoid arthritis(43) and ulcerative colitis(44). *ANXA1* belongs to the annexin family of Ca2+- dependent phospholipid-binding proteins, and through inhibition of phospholipase A2, it reduces eicosanoid production, which also contributes to its anti-inflammatory activities. Modulation of M2 macrophage phenotype is also promoted by *ANXA1* to attenuate tissue inflammation(45). Corticosteroids (a mainstay of asthma treatment) increase the synthesis of *ANXA1*(46). Plasma *ANXA1* levels are significantly lower in asthmatic patients with frequent exacerbations compared to those with stable disease, suggesting a link between this mediator and disease state(47). Furthermore, children with wheeze have reduced airway levels of *ANXA1*(48). Previous functional studies using *anxa1−/−* deficient mice challenged with ovalbumin showed *anxa1-*deficient mice to have elevated AHR compared to WT mice(49). Ng *et al.* demonstrated that untreated *anxa1*-deficient mice have spontaneous AHR that predisposes them to exacerbated response to allergen(49). In the current study, we demonstrated in the murine lung the induction of Anxa1 in response to HDM exposure. In addition, genetic deletion of *anxa1* potentiated the development of AHR and enhanced eosinophilia and markers of Th2 inflammation in mice treated with HDM, which is consistent with and extends previous reports. Of interest, in mice, *anxa1* expression was recently found to be characteristic of a novel cell-type called the Hillock cell, which may be involved in squamous barrier function and immunomodulation(50). These data identify the ANXA1/FPR2 signaling axis as an important regulator of allergic disease, that could be manipulated for therapeutic benefit. Our study has several limitations. By GWAS standards, our study is comparatively small and may be considered to be underpowered. The sample size may be an issue when using an aggregated definition (such as “doctor-diagnosed asthma”) but is less likely to be an issue when primary outcome is determined by deep phenotyping. This is indirectly confirmed in our analyses. Our primary outcome was derived through careful phenotyping over a period of more than two decades in five independent birth cohorts, and although comparatively smaller than some asthma GWASs, our study proved to be powered enough to detect previously identified key associations (e.g. chr17q21 locus). Precise phenotyping has the potential to identify new risk loci. For example, a comparatively small GWAS (1,173 cases and 2,522 controls) which used a specific subtype of early-onset childhood asthma with recurrent severe exacerbations as an outcome, identified a functional variant in a novel susceptibility gene *CDHR3* (SNP rs6967330) as an associate of this disease subtype, but not of doctor-diagnosed asthma(51). This important discovery was made with a considerably smaller sample size but using a more precise asthma subtype. In contrast, the largest asthma GWAS to date had a ∼40-fold higher sample size(7), but reported no significant association between *CDHR3* and aggregated asthma diagnosis. Therefore, with careful phenotyping, smaller sample sizes may be adequately powered to identify larger effect sizes than those in large GWASs with broader outcome definitions(52). The importance of the precise outcome definition was highlighted in our previous studies in ALSPAC which explored genetic associates of wheeze phenotypes derived by LCA(53, 54). Our current findings are consistent with our earlier report suggesting that 17q21 SNPs are associated with early-onset persistent, but not with early transient or late-onset wheeze (53). Further analysis using genetic prediction scores based on 10-200,000 SNPs ranked according to their associations with physician-diagnosed asthma found that the 46 highest ranked SNPs predicted persistent wheeze more strongly than doctor-diagnosed asthma(54). Finally, a candidate gene study combining data from ALSPAC and PIAMA found different associations of IL33-IL1RL1 pathway polymorphisms with different phenotypes(55). We are cognisant that there may be a perception of the lack of replication of our GWAS findings. We would argue that direct replication is almost certainly not possible in other cohorts, as phenotypes for replication studies should be homogenous(56). However, there is a considerable heterogeneity in LCA-derived wheeze phenotypes between studies, and although phenotypes in different studies are usually designated with the same names, they differ between studies in temporal trajectories, distributions within a population, and associated risk factors(57). This heterogeneity is in part consequent on the number and the non-uniformity of the timepoints used, and is likely one of the factors responsible for the lack of consistent associations of discovered phenotypes with risk factors reported in previous studies(58). This will also adversely impact the ability to identify phenotype-specific genetic associates. For example, we have previously shown that less distinct wheeze phenotypes in PIAMA were identified compared to those derived in ALSPAC(59). Thus, phenotypes that are homogeneous to those in our study almost certainly cannot readily be derived in available populations. This is exemplified in our attempted replication of *ANXA1* findings in PIAMA cohort (see OLS, **Table E12**). In this analysis, the number of individuals assigned to persistent wheezing in PIAMA was small (40), associates of this phenotype differed to those in STELAR cohorts, and the SNPs’ imputation scores were low (<0.60), which meant the conditions for replication were not met. Our study population is of European descent, and we cannot generalize the results to different ethnicities or environments. It is important to highlight the under-representation of ethnically diverse populations in most GWASs(9). To mitigate against this, large consortia have been formed, which combine the results of multiple ethnically diverse GWASs to increase the overall power to identify asthma-susceptibility loci. Examples include the GABRIEL(6), EVE(60) and TAGC(7) consortia, and the value of diverse, multi-ethnic participants in large-scale genomic studies has recently been shown(61). However, such consortia do not have the depth of longitudinal data to allow the type of analyses which we carried out to derive a multivariable primary outcome. One strength of our approach is that we used data from five birth cohorts with detailed and lifelong phenotyping, which were harmonised in a common knowledge management platform(20), allowing joint analyses. We performed three parallel GWASs that produced estimates with remarkably consistent directions of effects. In conclusion, using unique data from five UK birth cohorts, we identified subsets of SNPs differentially associated across four wheezing phenotypes from infancy to adolescence. We found little evidence of genetic associations spanning across different phenotypes. We discovered a novel locus in chr9q21 uniquely associated with early-onset persistent wheeze (p<6.7×10−9), identified SNP rs75260654 as the most likely causative variant, and demonstrated that the risk allele (T) confers a reduction in *ANXA1* expression. In mouse experiments, *ANXA1* expression increased in lung tissue following allergen exposure, suggesting that the pro-resolving ANXA1 may play a role in regulating the pulmonary immune response to allergen. Using *ANXA1*-deficient mice, we demonstrated that loss of *ANXA1* results in heightened AHR and Th2 inflammation upon allergen challenge, providing important *in vivo* functional data to support our GWAS finding. Targeting these pathways to promote the clearance of chronic inflammation in persistent disease may represents an exciting therapeutic prospect. ## Data Availability The informed consent obtained from all included participants does not allow the data to be made freely available through any third party maintained public repository. However, data used for this submission can be made available on request to the corresponding cohort Executive. The ALSPAC website provides information on how to request and access its data (http://www.bristol.ac.uk/alspac/researchers/access/). For queries regarding access of data from MAAS, IoW, SEATON or Ashford please contact Philip Couch philip.couch@manchester.ac.uk). All summary data used to plot the figures in our manuscript has been deposited in Dryad. [https://datadryad.org/stash/share/NtN5Q2cplp016xWs3QAbYkR8LamBxRVIGOQ6JmzWopQ](https://datadryad.org/stash/share/NtN5Q2cplp016xWs3QAbYkR8LamBxRVIGOQ6JmzWopQ) ## Data availability The informed consent obtained from all included participants does not allow the data to be made freely available through any third party maintained public repository. However, data used for this submission can be made available on request to the corresponding cohort Executive. The ALSPAC website provides information on how to request and access its data ([http://www.bristol.ac.uk/alspac/researchers/access/](http://www.bristol.ac.uk/alspac/researchers/access/)). For queries regarding access of data from MAAS, IoW, SEATON or Ashford please contact Philip Couch philip.couch{at}manchester.ac.uk). All summary data used to plot the figures in our manuscript has been deposited in Dryad. ## Code availability Code used for this submission has been deposited in Dryad. ## Author Contributions R.G., J.C., S.H. and A.C. conceived and planned the study. R.G., A.C., J.W.H., L.L.Y. and C.M.L. wrote the manuscript. R.G. and J.C. analysed the data. J.C. and N.K. provided eQTL analysis for *ANXA1*, J.W.H. supervised eQTL analyses. J.H. and M.T. performed the PCHi-C experiments and analysis. J.C. and M.T. interpreted the genetic architecture results for *ANXA1*. G.H.K and J.M.V. performed a replication study in PIAMA. C.M.L, S.A.M. and L.G.G. conceived and designed murine experiments. S.A.M and L.G.G. conducted experiments in mouse models. M.P. provided annexin*−/−* mice and advice on annexin. All authors provided critical feedback and helped shape the research, analysis, and manuscript. This publication is the work of the authors and Raquel Granell and Adnan Custovic will serve as guarantors for the contents of this paper. ## Competing Interests statement Graham Roberts: MRC grant to my institution President of the British Society of Allergy and Clinical Immunology. Gerard Koppelman: Dutch Lung Foundation, Ubbo Emmius Foundation (Money to insititution) Dutch Lung Foundation, Vertex, TEVA the Netherlands, GSK, ZON-MW (VICI grant), European Union (Money to institution) Astra Zeneca, Pure IMS, GSK (Money to institution) Sanofi, Boehringer Ingelheim (Money to institution). Angela Simpson: Medical research council Research grant JP Moulton Charitable Foundation Research grant Asthma UK Research grant. Clare Murray: Asthma Uk National Institute for Health Research Moulton Charitable Foundation North West Lung Centre Charity GSK (Lecture fees) Novartis (Lecture fees). Clare Lloyd: Wellcome Trust 107059/Z/15/Z. John Holloway: Medial Research Council grant MR/S025340/1 (to institution) American Academy of Allergy Asthma and Immunology (AAAI) (Support for speaker travel to AAAAI annual congress). Adnan Custovic: MRC (research grants) EPSRC (research grant) Wellcome Trust (research grant) Worg Pharmaceoticals (Personal payment 3%), insufficient sample replication (IBD < 0.8) or evidence of cryptic relatedness (IBD > 0.1). Following imputation, single nucleotide polymorphisms (SNPs) with a minor allele frequency of <1%, a call rate of <95%, evidence for violations of Hardy-Weinberg equilibrium (P<5E-7) or imputation quality measure (MaCH-Rsq or IMPUTE-info score) <0.40 were excluded. All individuals with non-European ancestry and siblings were removed. #### GWAS Meta-analysis GWAS of the joint wheezing phenotypes were performed independently in ALSPAC, MAAS and the combined IOW-SEATON-ASHFORD (combined as they were genotyped on the same platform, at the same time, and quality-controlled and imputed together). All genetic data were imputed to a new Haplotype Reference Consortium panel. This comprises around 31,000 sequenced individuals (mostly European), so the coverage of European haplotypes is much greater than in other panels. As a consequence, we expect to improve imputation accuracy, particularly at lower frequencies. We used SNPTEST v2.5.214 with a multinomial regression model (-method newml, never/infrequent wheeze as the reference) to investigate the association between SNPs and wheezing phenotypes. A meta-analysis of the three GWASs including 5,887 controls and 943 cases for early-onset persistent, 1482 cases for early-onset remitting, 603 cases for mid-childhood-onset remitting and 652 cases for late-onset wheeze, was performed using METAL15 with a total of 8,057,852 SNPs present. We used the option SCHEME STDERR in METAL to implement an effect-size based method weighted by each study- specific standard error in a fixed-effects model. We performed clumping to keep only one representative SNP per Linkage disequilibrium (LD) block and used locus zoom plots to short-list independent SNPs for further annotation. #### LD clumping, pre-Selection and Gene Annotation LD clumping was performed for all SNPs with p-value<10−5 for at least one wheezing phenotype. In order to avoid redundancy between SNPs and to ensure associations are independent, we used significance thresholds of 0.05 for index and clumped SNPs (--clump-p1 0.05,--clump-p2 0.05), LD threshold of 0.80 (--clump-r2 0.80) and physical distance threshold of 250kb (--clump-kb 250). European 1000 Genome data were used to infer LD structure. Locus Zoom plots ([http://locuszoom.org/](http://locuszoom.org/))16 were used for close inspection of all independent signals. Loci showing a peak with different colour dots (possibly indicating more than one causal variant) were short-listed for further annotation. SNPnexus database ([https://www.snp-nexus.org/v4/](https://www.snp-nexus.org/v4/))17 was used to annotate the overlapping, upstream and downstream genes; the GWAS catalogue (by SNP and then gene) ([https://www.ebi.ac.uk/gwas/search](https://www.ebi.ac.uk/gwas/search)), GeneCards ([https://www.genecards.org/](https://www.genecards.org/))18 database and phenoscanner ([http://www.phenoscanner.medschl.cam.ac.uk/](http://www.phenoscanner.medschl.cam.ac.uk/)) were used to further explore previously associated relevant phenotypes and gene function. Lead SNPs were looked in [https://www.regulomedb.org](https://www.regulomedb.org)/ to assess potential functionality. #### Gene expression in whole blood and lung tissues The top independent SNPs associated with each of the wheeze phenotypes were assessed for their association with cis- and trans-acting gene expression (mRNA) in whole blood and lung tissues. We identified potential eQTL signals using Genotype-Tissue Expression database ([https://gtexportal.org](https://gtexportal.org)) using the European reference panel. #### Post-GWAS: rs75260654 (*ANXA1*) ##### Annotation & distribution Information including chromosome, strand, clinical significance was retrieved from ENSEMBL using the R package biomaRt19, 20. The effects of rs75260654 on genomic features were predicted by querying the using Ensembl Variant Effect Predictor (VEP) 21 web tool. rs75260654 distribution in the GRCh38.p13 build of the human genome across African, Asian and European populations of the 1000 Genomes Project Phase 3 were accessed by querying the Ensembl ([www.ensembl.org](http://www.ensembl.org)) web browser on 24 May 2021. ##### Promoter Capture The Hi-C libraries were prepared from CD4+ T-cells isolated from 7 healthy individuals (2 libraries per individual) from the MAAS cohort using the Arima-HiC kit (Arima Genomics). Promoter Capture Hi-C (PCHi-C) libraries were generated by capturing the restriction fragments (RF) overlapping the TSS of 18775 protein coding genes using the Agilent SureSelectXT HS Target Enrichment System according to the manufacturers’ protocols. The final design included 305419 probes covering 13.476Mb and 18630 protein-coding genes. The restriction fragments (RF) overlapping the TSS (+/-1 RF, 3 RF per promoter) were captured with custom-designed biotinylated RNA baits. Libraries were sequenced to ∼300M 2×150bp reads each (∼600M reads/individual). The 3’-end of the reads was quality trimmed with Sickle. The sequencing data were processed with the HiCUP pipeline to map the sequencing reads and eliminate experimental artefacts and PCR duplicates22. The BAM files from technical replicates were merged. Promoter interactions were called using the CHiCAGO pipeline23, which calls statistically significant interactions in PCHi-C data while accounting for noise and PCHi-C specific bias. A CHiCAGO score > 5 (soft-thresholded -log weighted p-value) was considered significant. To gain information from all the available data, the BAM files from all 7 individuals were supplied as biological replicates in the analysis with CHiCAGO. Moreover, to increase power, restriction fragments were binned as follows: 10 consecutive RF that were not covered by the baits were binned together; the 3 baited fragments for each promoter were binned with 1 RF upstream and 1 downstream, totaling 5 fragments per promoter. If the bins for two consecutive promoters overlapped, these were binned together into a single larger bin. Publicly available ENCODE ATAC-seq (A549 cell line) and ChIP-seq (A549 cell line ENCFF900GVO) and POLR2A ChIP-seq (A549 cell line, ENCFF737ZKN) data and 18-state ChromHMM from the EpiMap Project (BSS00007) 24 for A549 cell line were downloaded. The PCHi-C interactions of interest and their overlap with ATAC-seq and ChIP-seq peaks, and putative enhancers from the 18-state ChromHMM model were visualised using the Sushi R package. ##### eQTL catalogue lookup We queried the eQTL catalogue ([https://www.ebi.ac.uk/eqtl/](https://www.ebi.ac.uk/eqtl/); accessed 6 May 2021) using tabix-0.2.6 to assess if *rs75260654*, *rs116849664* or *rs78320984* are eQTLs in studies that utilised the following cell types: lung, T cells, blood, monocytes, neutrophils, NK cells, fibroblasts, B cells, CD4+ T cells, CD8+ T cells, Th17 cells, Th1 cells, Th2 cells, Treg naive, Treg memory, CD16+ monocytes, Cultured fibroblasts, EBV-transformed lymphocytes. We defined nominal significance as p<=0.05. ##### Variant effect Variant effect on tissue-specific gene expression, which is based on GTEx eQTL, was retrieved on May 24 from eQTL Ensembl database ([https://www.ensembl.org/](https://www.ensembl.org/)) and eQTLGene Consortium ([https://www.eqtlgen.org/cis-eqtls.html](https://www.eqtlgen.org/cis-eqtls.html)).Using downloaded correlation of variant on tissue specific gene expression from Ensembl, the relative effect of T allele on the *ANXA1* expression across 86 tissue types was presented in scatter plot using R version 3.6.1.25 To get information on the functional role of *ANXA1*, the top 30 interacting proteins and enrichment were retrieved from STRING database26 into cystoscape for visualization.27 #### Functional mouse experiments ##### Mice In accordance with the Animals (scientific procedures) act 1986, all animal experiments were conducted under the approved UK Home Office Project License No: PPL 70/7643, reviewed by Imperial College’s Animal Welfare and Ethical Review body. Female WT BALB/c and annexin A1 knockout (KO) mice were purchased from Charles River (Bicester, UK). Animals aged 6-8 weeks of age received 25ug intranasal instillation of either HDM (Greer Laboratories, Lenoir, NC, USA; Cat: XPB70D3A25), or PBS 3x a week for 3 weeks. Mice were sacrificed 4 hours post-final HDM challenge. Mice were housed under specific pathogen-free conditions and a 12:12 light:dark cycle. Food and water were supplied ad libitum. All animal experiments were completed twice, with N=4-6 per group. ##### Airway hyperresponsiveness Airway hyperreactivity was measured using Flexivent™. Lung resistance was measured in response to increasing doses of methacholine (3-100mg/ml, Sigma, Poole, UK, Cat: A2251) in tracheotomised anaesthetised mice using an EMMS system (Electro-Medical Measurement Systems, UK). ##### Flow Cytometry Analysis Bronchoalveolar lavage (BAL) was collected. BAL cells were restimulated with ionomycin and phorbol 12-myristate 13-acetate in the presence of brefeldin (Sigma), as previously described28. Specific antibodies for T1/ST2 staining were purchased from Morwell Diagnostics (Zurich, Switzerland). Cells were also stained for lineage negative cocktail, Ly6G, CD45, CD11b, CD11c, SiglecF. Labelled cells were acquired on a BD Fortessa (BD Biosciences, Oxford, UK) and analysed using FlowJo software (Treestar, Ashland, Oregan, USA). Details of antibodies used can be found in the table below. ![Table2](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/T2.medium.gif) [Table2](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T2) ##### Analysis of cytokines and chemokines Murine lung tissue homogenate supernatants were processed as previously described28. Cytokine levels were analysed by ELISA: IL-4, IL-5 (PharMingen, Oxford, UK), IL-13 Ready-Set-Go kits (eBioscience). ##### Real time-PCR Total RNA was extracted from murine lung tissue using an RNeasy Mini Kit (Qiagen). Total RNA (1μg) was reverse transcribed into cDNA using a High-Capacity cDNA Reverse Transcription Kit (Life Technologies, UK). Real-time PCR reactions were performed using TaqMan Gene Expression Master Mix and TaqMan Gene Expression probes, annexin A1, and HPRT (Applied Biosystems). Values were normalised to HPRT and gene expression was analysed using the change-in-threshold 2-ΔCT method. ##### Annexin A1 Immunohistochemistry Paraffin-embedded mouse lung sections were stained with annexin A1 (R&D Systems, MAB3770). Annexin A1 primary antibody was followed by a secondary detection antibody (donkey anti-goat 488, Thermofisher, A11055). Annexin A1+ cells were quantified by manual counting under microscope and numbers averaged over four fields, from five biological replicates per group. ##### Statistical analysis Data are expressed as median ± IQR. Statistical differences between groups were calculated using Mann Whitney U test, unless otherwise specified. p -values are indicated in figures. #### Replication of ANXA1 top hits in PIAMA cohort ##### PIAMA cohort description PIAMA (Prevention and Incidence of Asthma and Mite Allergy) is an ongoing birth cohort study. Details of the study design have been published previously29, 30. In brief, pregnant women were recruited from the general population through antenatal clinics in the north, west and centre of the Netherlands in 1996-1997. The baseline study population consisted of 3963 new-borns. Questionnaires were completed by the parents during pregnancy when the child was 3 months old, and then annually from 1 up to 8 years; at ages 11, 14 and 17 years, questionnaires were completed by the parents as well as the participants themselves. ##### LCA wheezing phenotypes A 6 class LCA model was identified including 3,832 individuals with at least 2 observations of wheeze between 1 and 11-12 years of age. The identified classes were labelled: never/Infrequent (2909,75.91%), pre-school onset remitting (571, 14.90%), mid-childhood school remitting (108, 2.82%), intermediate onset remitting (106, 2.77%), school-age onset persisting (74, 1.93%) and continuous wheeze (64, 1.67%). ##### Replication analyses We analyzed aassociations between SNPs downstream of *ANXA1* (**Table E7**, **Figure E6**) and continuous wheezing in PIAMA, using the never/infrequent wheezing as the baseline category. Analyses were carried out in SPSS using a logistic regression model. ### ONLINE RESULTS #### Participants and descriptive data Demographic characteristics of the participants in STELAR cohorts included in this analysis are shown in **Table E3**. Cohorts contain similar proportions of males (range 48%-54%), maternal history of asthma (11%-14%), maternal smoking (14%-23%), (doctor-diagnosed) asthma ever during mid-childhood (16%-24%) and adolescence (20%-30%), current wheeze (12%-20% mid-childhood, 9%-25% adolescence) and current use of asthma medication (12%-17% mid-childhood, 11%-17% adolescence). ##### Comparison of participants included and excluded from this analysis In ALSPAC, 11,176 individuals had data on wheezing phenotypes, of these 6,833 were white unrelated and had genetic data. We found more children from mothers who smoked during pregnancy in the excluded sample compared to the included sample; no difference in gender, maternal history of asthma, current wheezing at 8 or 15 years, and small evidence for more asthma ever and current medication at 8 years in the excluded sample (**Table E4**). In MAAS, 1150 individuals had data on wheezing phenotypes, of these 887 were white unrelated and had genetic data. We found no difference in children from mothers who smoked during pregnancy in the excluded sample compared to the included sample; no difference in gender, maternal history of asthma or current wheezing at both 8 and 16 years. There was small evidence for more asthma ever and current medication at 8 years in the excluded sample (**Table E4**). In SEATON, 1535 individuals had data on joint wheezing phenotypes, of these 548 were white unrelated and had genetic data. We found evidence for more children from mothers who smoked during pregnancy in the excluded sample compared to the included sample; and more males in the excluded sample. There was no difference in maternal history of asthma or current wheezing, asthma ever or current medication at both 10 and 15 years in the excluded sample compared to the included sample (**Table E4**). In ASHFORD, 620 individuals had data on joint wheezing phenotypes, of these 348 were white unrelated and had genetic data. We found evidence for more children from mothers who smoked during pregnancy in the excluded sample compared to the included sample; no difference in gender, maternal history of asthma or asthma ever. There was small evidence for less current wheezing at 8 years, or current medication at 8 years in the excluded sample compared to the included sample (**Table E4**). In IOW, 1460 individuals had data on joint wheezing phenotypes, of these 952 were white unrelated and had genetic data. We found evidence for more children from mothers who smoked during pregnancy in the excluded sample compared to the included sample; no difference in gender, maternal history of asthma, asthma ever at 10 and 18 years in the excluded sample compared to the included sample. There was small evidence for more children with current wheeze and medication at 8 years in the included sample compared to the included sample (**Table E4**). #### GWAS Meta-Analysis: Main Results The distribution of the minor allele frequencies was consistent across genotyped datasets (mean SD 0.01). We assessed the deviation of the observed p-values from the null hypothesis by plotting QQ- plots for each wheezing phenotype (**Figure E1**). Some observed p-values were clearly more significant than expected under the null hypothesis, particularly for early-onset persistent wheeze, without an early separation of the expected from the observed (low evidence of population stratification; genomic control lambda≥0.93 for all four phenotypes). ##### Results of GWAS meta-analysis for 85 SNPs with main associations across wheeze phenotypes Previously associated traits for each region/gene associated with different wheezing phenotypes are presented in **Table E6.** ##### Persistent Wheeze We identified two GWAS-significant loci: 17q21, p<5.5×10−9, and a novel locus on 9q21.13 (*ANXA1*), p<6.7×10−9. The remaining 17 loci (4.0×10−7≤p-values≤9.8×10−6) included regions previously associated with childhood asthma (1q43, 4p16.3, 4q31.21, 5p15.31, 7q22.3, 17q12), asthma and rhinitis (2p25.1), eosinophil count (3q21.3, 10q24.2, 11q23.3, 22q11.1), bronchial hyper-responsiveness (2q12.2), lung function (1q43, 3q21.3, 5q13.3, 15q25.2, 19p13.2), triglycerides measurement and/or glucose metabolism (11q23.3, 19p13.2 and 22q11.1), severe asthma (14q22.1) and severe asthma and insulin resistance (11p15.4). ##### Early-onset Preschool Remitting Wheeze Among the regions associated with early-onset preschool remitting wheeze, we identified loci previously associated with smoking (3q26.31, 7q21.11 and 15q21.1), waist circumference and obesity (1q32.3), asthma and/or BMI (5q13.3, 6q27, 7q21.11), allergic disease and atopy (7q21.11) and airway repair (2p24.2 and 9p13.3). ##### Early-onset Mid-childhood Remitting Wheeze Loci associated with this phenotype were previously associated with neutrophil counts (1q23.2, 3q29, 20p12.3-p12.2), eosinophil counts and allergic rhinitis (4q24), pollution and DNA methylation (2p22.3), atopy (3p25.3), food allergy (13q31.1) and BMI (3q29, 5q14.1). ##### Late-onset Wheeze Regions associated with late-onset wheeze were previously associated with adult-onset non-allergic asthma (3q13.2), asthma/allergic disease and allergy/atopic sensitization (3q23, 7q36.1), asthma and/or allergy in adolescence (9p22.3, 16p12.1), late-onset asthma and obesity (2q13), lung function or body height (2q14.3, 3p24.2, 3q13.2, 15q25.2), lymphocyte count and asthma susceptibility (1p32.2), obesity and/or metabolic syndrome/dysfunction (1p36.13 and 22q13.32), eczema (7q36.3), insulin resistance (10q23.31), type 1 diabetes (8q21.3), alcohol drinking (15q15.3-q21.1) and sex hormone-binding globulin levels (11q23.3). ### ONLINE TABLES View this table: [Table E1.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T3) Table E1. Definition of variables in each of the five STELAR birth cohorts View this table: [Table E2.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T4) Table E2. The cohort-specific time points and sample size used to ascertain wheeze phenotypes View this table: [Table E3.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T5) Table E3. Characteristics of the participants in STELAR cohorts included in this analysis (restricted to individuals with genetic data). Numbers are N (%) except for age, where we report Mean (SD). View this table: [Table E4.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T6) Table E4. Comparison of included vs. excluded participants in the five cohorts at different ages View this table: [Table E5.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T7) Table E5. List of 134 independent SNPs identified after clumping and associated with at least one wheezing phenotype (p<10×−5) View this table: [Table E6.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T8) Table E6. References to previous relevant associated traits for each region/gene identified by wheezing phenotype View this table: [Table E7.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T9) Table E7. SNPs near ANXA1 associated with persistent wheeze View this table: [Table E8.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T10) Table E8. Allele Frequencies of *rs75260654* across different wheeze phenotypes View this table: [Table E9.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T11) Table E9. Selected immune eQTLs of rs75260654 View this table: [Table E10.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T12) Table E10. Lung eQTLs of rs75260654 View this table: [Table E11:](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T13) Table E11: Functional enrichment for ANXA1: Top 10 GO terms View this table: [Table E12.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/T14) Table E12. Replication of associations between SNPs downstream of *ANXA1* and early-onset persistent wheezing in PIAMA ### SUPPLEMENTARY FIGURES ![Figure E1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F5.medium.gif) [Figure E1.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F5) Figure E1. QQ-plots for each wheezing phenotype ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F6/graphic-36.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F6/graphic-36) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F6/graphic-37.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F6/graphic-37) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F6/graphic-38.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F6/graphic-38) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F6/graphic-39.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F6/graphic-39) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F6/graphic-40.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F6/graphic-40) Figure E2. Zoom locus plots for short-listed independent top hits for Persistent Wheezing ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F7/graphic-41.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F7/graphic-41) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F7/graphic-42.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F7/graphic-42) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F7/graphic-43.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F7/graphic-43) Figure E3. Zoom locus plots for short-listed independent top hits for Early-onset Pre-school Remitting Wheezing ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F8/graphic-44.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F8/graphic-44) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F8/graphic-45.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F8/graphic-45) Figure E4. Zoom locus plots for short-listed independent top hits for Early-onset Mid-childhood Remitting Wheezing ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F9/graphic-46.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F9/graphic-46) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F9/graphic-47.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F9/graphic-47) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F9/graphic-48.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F9/graphic-48) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F9/graphic-49.medium.gif) [](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F9/graphic-49) Figure E5. Zoom locus plots for short-listed independent top hits for Late-onset Wheezing ![Figure E6.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F10.medium.gif) [Figure E6.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F10) Figure E6. Linkage disequilibrium between SNPs downstream of *ANXA1* that were associated with persistent wheeze. ![Figure E7.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/03/05/2023.03.03.23284449/F11.medium.gif) [Figure E7.](http://medrxiv.org/content/early/2023/03/05/2023.03.03.23284449/F11) Figure E7. *ANXA1* interactors. Protein-protein interaction of *ANXA1* including *IL-4*, *IL-13* and *NR3C1* ## Footnotes * 13 **STELAR/UNICORN investigators:** Professor Graham Devereux, Dr Dimitrios Charalampopoulos * 14 **Breathing Together investigators:** Prof Andrew Bush, Prof Sejal Saglani, Prof Benjamin Marsland, Prof Jonathan Grigg, Prof Jurgen Schwarze, Prof Mike Shields, Prof Peter Ghazal, Prof Ultan Power, Dr Ceyda Oksel * **This article has an online appendix.** This article is dedicated to the memory of our wonderful colleague and friend Professor John Henderson (1958-2019), whose contribution to the understanding of the heterogeneity of childhood asthma cannot be overstated. Rainbow-chasers and UNICORN riders forever. * Received March 3, 2023. * Revision received March 3, 2023. * Accepted March 5, 2023. * © 2023, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution 4.0 International), CC BY 4.0, as described at [http://creativecommons.org/licenses/by/4.0/](http://creativecommons.org/licenses/by/4.0/) ## REFERENCES 1. 1.Pavord ID, Beasley R, Agusti A, Anderson GP, Bel E, Brusselle G, Cullinan P, Custovic A, Ducharme FM, Fahy JV, Frey U, Gibson P, Heaney LG, Holt PG, Humbert M, Lloyd CM, Marks G, Martinez FD, Sly PD, von Mutius E, Wenzel S, Zar HJ, Bush A. After asthma: redefining airways diseases. Lancet 2018; 391: 350–400. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(17)30879-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28911920&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 2. 2.Duffy DL, Martin NG, Battistutta D, Hopper JL, Mathews JD. Genetics of asthma and hay fever in Australian twins. Am Rev Respir Dis 1990; 142: 1351–1358. [CrossRef](http://medrxiv.org/lookup/external-ref?access\_num=10.1164/ajrccm/142.6_Pt_1.1351&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2252253&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1990EM38500020&link_type=ISI) 3. 3.Simpson A, Custovic A, Tepper R, Graves P, Stern DA, Jones M, Hankinson J, Curtin JA, Wu J, Blekic M, Bukvic BK, Aberle N, Marinho S, Belgrave D, Morgan WJ, Martinez FD. Genetic variation in vascular endothelial growth factor-a and lung function. Am J Respir Crit Care Med 2012; 185: 1197–1204. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1164/rccm.201112-2191OC&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22461367&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 4. 4.Daniels SE, Bhattacharrya S, James A, Leaves NI, Young A, Hill MR, Faux JA, Ryan GF, le Souef PN, Lathrop GM, Musk AW, Cookson WO. A genome-wide search for quantitative trait loci underlying asthma. Nature 1996; 383: 247–250. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/383247a0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8805698&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 5. 5.Moffatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S, Depner M, von Berg A, Bufe A, Rietschel E, Heinzmann A, Simma B, Frischer T, Willis-Owen SA, Wong KC, Illig T, Vogelberg C, Weiland SK, von Mutius E, Abecasis GR, Farrall M, Gut IG, Lathrop GM, Cookson WO. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature 2007; 448: 470–473. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature06014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17611496&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000248302700046&link_type=ISI) 6. 6.Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, von Mutius E, Farrall M, Lathrop M, Cookson W, Consortium G. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med 2010; 363: 1211–1221. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa0906312&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20860503&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000282050000005&link_type=ISI) 7. 7.Demenais F, Margaritte-Jeannin P, Barnes KC, Cookson WOC, Altmuller J, Ang W, Barr RG, Beaty TH, Becker AB, Beilby J, Bisgaard H, Bjornsdottir US, Bleecker E, Bonnelykke K, Boomsma DI, Bouzigon E, Brightling CE, Brossard M, Brusselle GG, Burchard E, Burkart KM, Bush A, Chan-Yeung M, Chung KF, Couto Alves A, Curtin JA, Custovic A, Daley D, de Jongste JC, Del-Rio-Navarro BE, Donohue KM, Duijts L, Eng C, Eriksson JG, Farrall M, Fedorova Y, Feenstra B, Ferreira MA, Australian Asthma Genetics Consortium c, Freidin MB, Gajdos Z, Gauderman J, Gehring U, Geller F, Genuneit J, Gharib SA, Gilliland F, Granell R, Graves PE, Gudbjartsson DF, Haahtela T, Heckbert SR, Heederik D, Heinrich J, Heliovaara M, Henderson J, Himes BE, Hirose H, Hirschhorn JN, Hofman A, Holt P, Hottenga J, Hudson TJ, Hui J, Imboden M, Ivanov V, Jaddoe VWV, James A, Janson C, Jarvelin MR, Jarvis D, Jones G, Jonsdottir I, Jousilahti P, Kabesch M, Kahonen M, Kantor DB, Karunas AS, Khusnutdinova E, Koppelman GH, Kozyrskyj AL, Kreiner E, Kubo M, Kumar R, Kumar A, Kuokkanen M, Lahousse L, Laitinen T, Laprise C, Lathrop M, Lau S, Lee YA, Lehtimaki T, Letort S, Levin AM, Li G, Liang L, Loehr LR, London SJ, Loth DW, Manichaikul A, Marenholz I, Martinez FJ, Matheson MC, Mathias RA, Matsumoto K, Mbarek H, McArdle WL, Melbye M, Melen E, Meyers D, Michel S, Mohamdi H, Musk AW, Myers RA, Nieuwenhuis MAE, Noguchi E, O’Connor GT, Ogorodova LM, Palmer CD, Palotie A, Park JE, Pennell CE, Pershagen G, Polonikov A, Postma DS, Probst-Hensch N, Puzyrev VP, Raby BA, Raitakari OT, Ramasamy A, Rich SS, Robertson CF, Romieu I, Salam MT, Salomaa V, Schlunssen V, Scott R, Selivanova PA, Sigsgaard T, Simpson A, Siroux V, Smith LJ, Solodilova M, Standl M, Stefansson K, Strachan DP, Stricker BH, Takahashi A, Thompson PJ, Thorleifsson G, Thorsteinsdottir U, Tiesler CMT, Torgerson DG, Tsunoda T, Uitterlinden AG, van der Valk RJP, Vaysse A, Vedantam S, von Berg A, von Mutius E, Vonk JM, Waage J, Wareham NJ, Weiss ST, White WB, Wickman M, Widen E, Willemsen G, Williams LK, Wouters IM, Yang JJ, Zhao JH, Moffatt MF, Ober C, Nicolae DL. Multiancestry association study identifies new asthma risk loci that colocalize with immune-cell enhancer marks. Nat Genet 2018; 50: 42–53. 8. 8.El-Husseini ZW, Gosens R, Dekker F, Koppelman GH. The genetics of asthma and the promise of genomics-guided drug target discovery. Lancet Respir Med 2020; 8: 1045–1056. 9. 9.Kim KW, Ober C. Lessons Learned From GWAS of Asthma. Allergy Asthma Immun 2019; 11: 170–187. 10. 10.Ober C, Yao TC. The genetics of asthma and allergic disease: a 21st century perspective. Immunol Rev 2011; 242: 10–30. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1600-065X.2011.01029.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21682736&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 11. 11.Custovic A, Marinho S, Simpson A. Gene-environment interactions in the development of asthma and atopy. Expert Rev Respir Med 2012; 6: 301–308. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22788944&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 12. 12.Custovic A. “Asthma” or “Asthma Spectrum Disorder”? J Allergy Clin Immunol Pract 2020; 8: 2628–2629. 13. 13.Haider S, Granell R, Curtin J, Fontanella S, Cucco A, Turner S, Simpson A, Roberts G, Murray CS, Holloway JW, Devereux G, Cullinan P, Arshad SH, Custovic A. Modeling Wheezing Spells Identifies Phenotypes with Different Outcomes and Genetic Associates. Am J Respir Crit Care Med 2022; 205: 883–893. 14. 14.Custovic A, Henderson J, Simpson A. Does understanding endotypes translate to better asthma management options for all? J Allergy Clin Immunol 2019; 144: 25–33. 15. 15.Aaron SD, Vandemheen KL, FitzGerald JM, Ainslie M, Gupta S, Lemiere C, Field SK, McIvor RA, Hernandez P, Mayers I, Mulpuru S, Alvarez GG, Pakhale S, Mallick R, Boulet LP, Canadian Respiratory Research N. Reevaluation of Diagnosis in Adults With Physician-Diagnosed Asthma. JAMA 2017; 317: 269–279. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2016.19627&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28114551&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 16. 16.Looijmans-van den Akker I, van Luijn K, Verheij T. Overdiagnosis of asthma in children in primary care: a retrospective analysis. Br J Gen Pract 2016; 66: e152–157. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiYmpncCI7czo1OiJyZXNpZCI7czoxMToiNjYvNjQ0L2UxNTIiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wMy8wNS8yMDIzLjAzLjAzLjIzMjg0NDQ5LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 17. 17.Robinson PFM, Fontanella S, Ananth S, Martin Alonso A, Cook J, Kaya-de Vries D, Polo Silveira L, Gregory L, Lloyd C, Fleming L, Bush A, Custovic A, Saglani S. Recurrent Severe Preschool Wheeze: From Prespecified Diagnostic Labels to Underlying Endotypes. Am J Respir Crit Care Med 2021; 204: 523–535. 18. 18.Howard R, Rattray M, Prosperi M, Custovic A. Distinguishing asthma phenotypes using machine learning approaches. Current allergy and asthma reports 2015; 15: 1–10. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s11882-014-0501-1&link_type=DOI) 19. 19.Oksel C, Granell R, Haider S, Fontanella S, Simpson A, Turner S, Devereux G, Arshad SH, Murray CS, Roberts G, Holloway JW, Cullinan P, Henderson J, Custovic A, Stelar investigators bTi. Distinguishing Wheezing Phenotypes from Infancy to Adolescence. A Pooled Analysis of Five Birth Cohorts. Ann Am Thorac Soc 2019; 16: 868–876. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 20. 20.Custovic A, Ainsworth J, Arshad H, Bishop C, Buchan I, Cullinan P, Devereux G, Henderson J, Holloway J, Roberts G, Turner S, Woodcock A, Simpson A. The Study Team for Early Life Asthma Research (STELAR) consortium ‘Asthma e-lab’: team science bringing data, methods and investigators together. Thorax 2015; 70: 799–801. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjg6IjcwLzgvNzk5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDMvMDUvMjAyMy4wMy4wMy4yMzI4NDQ0OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 21. 21.Golding J, Pembrey M, Jones R. ALSPAC-the Avon longitudinal study of parents and children. I. Study methodology. Paediatric and perinatal epidemiology 2001; 15: 74–87. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1046/j.1365-3016.2001.00325.x&link_type=DOI) 22. 22.Cullinan P, MacNeill SJ, Harris JM, Moffat S, White C, Mills P, Newman Taylor AJ. Early allergen exposure, skin prick responses, and atopic wheeze at age 5 in English children: a cohort study. Thorax 2004; 59: 855–861. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjk6IjU5LzEwLzg1NSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzAzLzA1LzIwMjMuMDMuMDMuMjMyODQ0NDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 23. 23.Arshad SH, Holloway JW, Karmaus W, Zhang H, Ewart S, Mansfield L, Matthews S, Hodgekiss C, Roberts G, Kurukulaaratchy R. Cohort Profile: The Isle Of Wight Whole Population Birth Cohort (IOWBC). Int J Epidemiol 2018; 47: 1043–1044i. 24. 24.Custovic A, Simpson BM, Murray CS, Lowe L, Woodcock A, Asthma NACM, Allergy Study G. The National Asthma Campaign Manchester Asthma and Allergy Study. Pediatr Allergy Immunol 2002; 13: 32–37. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1034/j.1399-3038.13.s.15.3.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12688622&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 25. 25.Martindale S, McNeill G, Devereux G, Campbell D, Russell G, Seaton A. Antioxidant intake in pregnancy in relation to wheeze and eczema in the first two years of life. American journal of respiratory and critical care medicine 2005; 171: 121–128. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1164/rccm.200402-220OC&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15531754&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000226258400007&link_type=ISI) 26. 26.Marchini J, Howie B. Genotype imputation for genome-wide association studies. Nat Rev Genet 2010; 11: 499–511. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrg2796&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20517342&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000278998500012&link_type=ISI) 27. 27.Willer CJ, Li Y, Abecasis GR. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 2010; 26: 2190–2191. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btq340&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20616382&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000281738900017&link_type=ISI) 28. 28.Melen E, Himes BE, Brehm JM, Boutaoui N, Klanderman BJ, Sylvia JS, Lasky-Su J. Analyses of shared genetic factors between asthma and obesity in children. J Allergy Clin Immunol 2010; 126: 631–637 e631-638. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2010.06.030&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20816195&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000281512500033&link_type=ISI) 29. 29.Herrera-Luis E, Espuela-Ortiz A, Lorenzo-Diaz F, Keys KL, Mak ACY, Eng C, Huntsman S, Villar J, Rodriguez-Santana JR, Burchard EG, Pino-Yanes M. Genome-wide association study reveals a novel locus for asthma with severe exacerbations in diverse populations. Pediatr Allergy Immunol 2021; 32: 106–115. 30. 30.Zhu Z, Guo Y, Shi H, Liu CL, Panganiban RA, Chung W, O’Connor LJ, Himes BE, Gazal S, Hasegawa K, Camargo CA, Jr., Qi L, Moffatt MF, Hu FB, Lu Q, Cookson WOC, Liang L. Shared genetic and experimental links between obesity-related traits and asthma subtypes in UK Biobank. J Allergy Clin Immunol 2020; 145: 537–549. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2019.09.035&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 31. 31.Cusanovich DA, Billstrand C, Zhou X, Chavarria C, De Leon S, Michelini K, Pai AA, Ober C, Gilad Y. The combination of a genome-wide association study of lymphocyte count and analysis of gene expression data reveals novel asthma candidate genes. Hum Mol Genet 2012; 21: 2111–2123. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/hmg/dds021&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22286170&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000302302800016&link_type=ISI) 32. 32.Siroux V, Gonzalez JR, Bouzigon E, Curjuric I, Boudier A, Imboden M, Anto JM, Gut I, Jarvis D, Lathrop M, Omenaas ER, Pin I, Wjst M, Demenais F, Probst-Hensch N, Kogevinas M, Kauffmann F. Genetic heterogeneity of asthma phenotypes identified by a clustering approach. Eur Respir J 2014; 43: 439–452. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiZXJqIjtzOjU6InJlc2lkIjtzOjg6IjQzLzIvNDM5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDMvMDUvMjAyMy4wMy4wMy4yMzI4NDQ0OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 33. 33.Heinonen MT, Laine AP, Soderhall C, Gruzieva O, Rautio S, Melen E, Pershagen G, Lahdesmaki HJ, Knip M, Ilonen J, Henttinen TA, Kere J, Lahesmaa R, Finnish Pediatric Diabetes R. GIMAP GTPase family genes: potential modifiers in autoimmune diabetes, asthma, and allergy. J Immunol 2015; 194: 5885–5894. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTE6IjE5NC8xMi81ODg1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDMvMDUvMjAyMy4wMy4wMy4yMzI4NDQ0OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 34. 34.Denham S, Koppelman GH, Blakey J, Wjst M, Ferreira MA, Hall IP, Sayers I. Meta-analysis of genome-wide linkage studies of asthma and related traits. Respir Res 2008; 9: 38. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1465-9921-9-38&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18442398&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 35. 35.Sottile G, Ferrante G, Torregrossa M, Cibella F, Cilluffo G, Fasola S, Alessandro R, Seidita G, Viegi G, La Grutta S. An association analysis to identify genetic variants linked to asthma and rhino-conjunctivitis in a cohort of Sicilian children. Ital J Pediatr 2019; 45: 16. 36. 36.Lutz SM, Cho MH, Young K, Hersh CP, Castaldi PJ, McDonald ML, Regan E, Mattheisen M, DeMeo DL, Parker M, Foreman M, Make BJ, Jensen RL, Casaburi R, Lomas DA, Bhatt SP, Bakke P, Gulsvik A, Crapo JD, Beaty TH, Laird NM, Lange C, Hokanson JE, Silverman EK, Investigators E, Investigators CO. A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry. BMC Genet 2015; 16: 138. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12863-015-0299-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26634245&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 37. 37. Vieira Braga FA, Kar G, Berg M, Carpaij OA, Polanski K, Simon LM, Brouwer S, Gomes T, Hesse L, Jiang J, Fasouli ES, Efremova M, Vento-Tormo R, Talavera-Lopez C, Jonker MR, Affleck K, Palit S, Strzelecka PM, Firth HV, Mahbubani KT, Cvejic A, Meyer KB, Saeb-Parsy K, Luinge M, Brandsma CA, Timens W, Angelidis I, Strunz M, Koppelman GH, van Oosterhout AJ, Schiller HB, Theis FJ, van den Berge M, Nawijn MC, Teichmann SA. A cellular census of human lungs identifies novel cell states in health and in asthma. Nat Med 2019; 25: 1153–1163. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/S41591-019-0468-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31209336&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 38. 38.Leoni G, Neumann PA, Kamaly N, Quiros M, Nishio H, Jones HR, Sumagin R, Hilgarth RS, Alam A, Fredman G, Argyris I, Rijcken E, Kusters D, Reutelingsperger C, Perretti M, Parkos CA, Farokhzad OC, Neish AS, Nusrat A. Annexin A1-containing extracellular vesicles and polymeric nanoparticles promote epithelial wound repair. J Clin Invest 2015; 125: 1215–1227. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI76693&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25664854&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 39. 39.Quach H, Rotival M, Pothlichet J, Loh YE, Dannemann M, Zidane N, Laval G, Patin E, Harmant C, Lopez M, Deschamps M, Naffakh N, Duffy D, Coen A, Leroux-Roels G, Clement F, Boland A, Deleuze JF, Kelso J, Albert ML, Quintana-Murci L. Genetic Adaptation and Neandertal Admixture Shaped the Immune System of Human Populations. Cell 2016; 167: 643–656 e617. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2016.09.024&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27768888&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 40. 40.Võsa U, Claringbould A, Westra H-J, Bonder MJ, Deelen P, Zeng B, Kirsten H, Saha A, Kreuzhuber R, Kasela S, Pervjakova N, Alvaes I, Fave M-J, Agbessi M, Christiansen M, Jansen R, Seppälä I, Tong L, Teumer A, Schramm K, Hemani G, Verlouw J, Yaghootkar H, Sönmez R, Brown A, Kukushkina V, Kalnapenkis A, Rüeger S, Porcu E, Kronberg-Guzman J, Kettunen J, Powell J, Lee B, Zhang F, Arindrarto W, Beutner F, Brugge H, Dmitreva J, Elansary M, Fairfax BP, Georges M, Heijmans BT, Kähönen M, Kim Y, Knight JC, Kovacs P, Krohn K, Li S, Loeffler M, Marigorta UM, Mei H, Momozawa Y, Müller-Nurasyid M, Nauck M, Nivard M, Penninx B, Pritchard J, Raitakari O, Rotzchke O, Slagboom EP, Stehouwer CDA, Stumvoll M, Sullivan P, Hoen PACt, Thiery J, Tönjes A, van Dongen J, van Iterson M, Veldink J, Völker U, Wijmenga C, Swertz M, Andiappan A, Montgomery GW, Ripatti S, Perola M, Kutalik Z, Dermitzakis E, Bergmann S, Frayling T, van Meurs J, Prokisch H, Ahsan H, Pierce B, Lehtimäki T, Boomsma D, Psaty BM, Gharib SA, Awadalla P, Milani L, Ouwehand W, Downes K, Stegle O, Battle A, Yang J, Visscher PM, Scholz M, Gibson G, Esko T, Franke L. Unraveling the polygenic architecture of complex traits using blood eQTL metaanalysis. 2018: 447367. 41. 41.Gregory LG, Causton B, Murdoch JR, Mathie SA, O’Donnell V, Thomas CP, Priest FM, Quint DJ, Lloyd CM. Inhaled house dust mite induces pulmonary T helper 2 cytokine production. Clin Exp Allergy 2009; 39: 1597–1610. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1365-2222.2009.03302.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19545261&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000269742000019&link_type=ISI) 42. 42.Perretti M, Chiang N, La M, Fierro IM, Marullo S, Getting SJ, Solito E, Serhan CN. Endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoid and aspirin treatment activate the lipoxin A4 receptor. Nat Med 2002; 8: 1296–1302. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm786&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12368905&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000179221200037&link_type=ISI) 43. 43.D’Acquisto F, Paschalidis N, Raza K, Buckley CD, Flower RJ, Perretti M. Glucocorticoid treatment inhibits annexin-1 expression in rheumatoid arthritis CD4+ T cells. Rheumatology (Oxford) 2008; 47: 636–639. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/rheumatology/ken062&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18390587&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000255315700016&link_type=ISI) 44. 44.Vong L, Ferraz JG, Dufton N, Panaccione R, Beck PL, Sherman PM, Perretti M, Wallace JL. Up-regulation of Annexin-A1 and lipoxin A(4) in individuals with ulcerative colitis may promote mucosal homeostasis. PLoS One 2012; 7: e39244. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0039244&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22723974&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 45. 45.McArthur S, Juban G, Gobbetti T, Desgeorges T, Theret M, Gondin J, Toller-Kawahisa JE, Reutelingsperger CP, Chazaud B, Perretti M, Mounier R. Annexin A1 drives macrophage skewing to accelerate muscle regeneration through AMPK activation. J Clin Invest 2020; 130: 1156–1167. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI124635&link_type=DOI) 46. 46.Rhen T, Cidlowski JA. Antiinflammatory action of glucocorticoids--new mechanisms for old drugs. N Engl J Med 2005; 353: 1711–1723. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMra050541&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16236742&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000232653200011&link_type=ISI) 47. 47.Lee SH, Lee PH, Kim BG, Seo HJ, Baek AR, Park JS, Lee JH, Park SW, Kim DJ, Park CS, Jang AS. Annexin A1 in plasma from patients with bronchial asthma: its association with lung function. BMC Pulm Med 2018; 18: 1. 48. 48. Eke Gungor H, Tahan F, Gokahmetoglu S, Saraymen B. Decreased levels of lipoxin A4 and annexin A1 in wheezy infants. Int Arch Allergy Immunol 2014; 163: 193–197. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1159/000358490&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24525704&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 49. 49.Ng FS, Wong KY, Guan SP, Mustafa FB, Kajiji TS, Bist P, Biswas SK, Wong WS, Lim LH. Annexin-1-deficient mice exhibit spontaneous airway hyperresponsiveness and exacerbated allergen-specific antibody responses in a mouse model of asthma. Clin Exp Allergy 2011; 41: 1793–1803. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1365-2222.2011.03855.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22092555&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 50. 50.Montoro DT, Haber AL, Biton M, Vinarsky V, Lin B, Birket SE, Yuan F, Chen S, Leung HM, Villoria J, Rogel N, Burgin G, Tsankov AM, Waghray A, Slyper M, Waldman J, Nguyen L, Dionne D, Rozenblatt-Rosen O, Tata PR, Mou H, Shivaraju M, Bihler H, Mense M, Tearney GJ, Rowe SM, Engelhardt JF, Regev A, Rajagopal J. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 2018; 560: 319–324. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-018-0393-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30069044&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 51. 51.Bonnelykke K, Sleiman P, Nielsen K, Kreiner-Moller E, Mercader JM, Belgrave D, den Dekker HT, Husby A, Sevelsted A, Faura-Tellez G, Mortensen LJ, Paternoster L, Flaaten R, Molgaard A, Smart DE, Thomsen PF, Rasmussen MA, Bonas-Guarch S, Holst C, Nohr EA, Yadav R, March ME, Blicher T, Lackie PM, Jaddoe VW, Simpson A, Holloway JW, Duijts L, Custovic A, Davies DE, Torrents D, Gupta R, Hollegaard MV, Hougaard DM, Hakonarson H, Bisgaard H. A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat Genet 2014; 46: 51–55. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.2830&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24241537&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 52. 52.Schoettler N, Rodriguez E, Weidinger S, Ober C. Advances in asthma and allergic disease genetics: Is bigger always better? J Allergy Clin Immunol 2019; 144: 1495–1506. 53. 53.Granell R, Henderson AJ, Timpson N, St Pourcain B, Kemp JP, Ring SM, Ho K, Montgomery SB, Dermitzakis ET, Evans DM, Sterne JA. Examination of the relationship between variation at 17q21 and childhood wheeze phenotypes. J Allergy Clin Immunol 2013; 131: 685–694. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2012.09.021&link_type=DOI) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000315587800008&link_type=ISI) 54. 54.Spycher B, Henderson J, Granell R, Evans DM, Smith GD, Timpson NJ, Sterne JAC. Genome-wide prediction of childhood asthma and related phenotypes in a longitudinal birth cohort. Eur Respir J 2012; 40. 55. 55.Savenije OE, Mahachie John JM, Granell R, Kerkhof M, Dijk FN, de Jongste JC, Smit HA, Brunekreef B, Postma DS, Van Steen K, Henderson J, Koppelman GH. Association of IL33-IL-1 receptor-like 1 (IL1RL1) pathway polymorphisms with wheezing phenotypes and asthma in childhood. J Allergy Clin Immunol 2014; 134: 170–177. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2013.12.1080&link_type=DOI) 56. 56.. Crawford F CG, Chappell FM, et al. A systematic review and individual patient data meta-analysis of prognostic factors for foot ulceration in people with diabetes: the international research collaboration for the prediction of diabetic foot ulcerations (PODUS). Southampton (UK): NIHR Journals Library; 2015 Jul. (Health Technology Assessment, No. 19.57.) Chapter 5, Validation of the model. Available from: [https://www.ncbi.nlm.nih.gov/books/NBK305598/](https://www.ncbi.nlm.nih.gov/books/NBK305598/). 57. 57.Oksel C, Haider S, Fontanella S, Frainay C, Custovic A. Classification of Pediatric Asthma: From Phenotype Discovery to Clinical Practice. Front Pediatr 2018; 6: 258. 58. 58.Oksel C, Granell R, Mahmoud O, Custovic A, Henderson AJ, Stelar, Breathing Together i. Causes of variability in latent phenotypes of childhood wheeze. J Allergy Clin Immunol 2019; 143: 1783–1790 e1711. 59. 59.Savenije OE, Granell R, Caudri D, Koppelman GH, Smit HA, Wijga A, de Jongste JC, Brunekreef B, Sterne JA, Postma DS, Henderson J, Kerkhof M. Comparison of childhood wheezing phenotypes in 2 birth cohorts: ALSPAC and PIAMA. J Allergy Clin Immunol 2011; 127: 1505–1512 e1514. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2011.02.002&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21411131&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000291048500029&link_type=ISI) 60. 60.Torgerson DG, Ampleford EJ, Chiu GY, Gauderman WJ, Gignoux CR, Graves PE, Himes BE, Levin AM, Mathias RA, Hancock DB, Baurley JW, Eng C, Stern DA, Celedon JC, Rafaels N, Capurso D, Conti DV, Roth LA, Soto-Quiros M, Togias A, Li X, Myers RA, Romieu I, Van Den Berg DJ, Hu D, Hansel NN, Hernandez RD, Israel E, Salam MT, Galanter J, Avila PC, Avila L, Rodriquez-Santana JR, Chapela R, Rodriguez-Cintron W, Diette GB, Adkinson NF, Abel RA, Ross KD, Shi M, Faruque MU, Dunston GM, Watson HR, Mantese VJ, Ezurum SC, Liang L, Ruczinski I, Ford JG, Huntsman S, Chung KF, Vora H, Li X, Calhoun WJ, Castro M, Sienra-Monge JJ, del Rio-Navarro B, Deichmann KA, Heinzmann A, Wenzel SE, Busse WW, Gern JE, Lemanske RF, Jr., Beaty TH, Bleecker ER, Raby BA, Meyers DA, London SJ, Mexico City Childhood Asthma S, Gilliland FD, Children’s Health S, study H, Burchard EG, Genetics of Asthma in Latino Americans Study SoG-E, Admixture in Latino A, Study of African Americans AG, Environments, Martinez FD, Childhood Asthma R, Education N, Weiss ST, Childhood Asthma Management P, Williams LK, Study of Asthma P, Pharmacogenomic Interactions by R-E, Barnes KC, Genetic Research on Asthma in African Diaspora S, Ober C, Nicolae DL. Meta-analysis of genome-wide association studies of asthma in ethnically diverse North American populations. Nat Genet 2011; 43: 887–892. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.888&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21804549&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 61. 61.Wojcik GL, Graff M, Nishimura KK, Tao R, Haessler J, Gignoux CR, Highland HM, Patel YM, Sorokin EP, Avery CL, Belbin GM, Bien SA, Cheng I, Cullina S, Hodonsky CJ, Hu Y, Huckins LM, Jeff J, Justice AE, Kocarnik JM, Lim U, Lin BM, Lu Y, Nelson SC, Park SL, Poisner H, Preuss MH, Richard MA, Schurmann C, Setiawan VW, Sockell A, Vahi K, Verbanck M, Vishnu A, Walker RW, Young KL, Zubair N, Acuna-Alonso V, Ambite JL, Barnes KC, Boerwinkle E, Bottinger EP, Bustamante CD, Caberto C, Canizales-Quinteros S, Conomos MP, Deelman E, Do R, Doheny K, Fernandez-Rhodes L, Fornage M, Hailu B, Heiss G, Henn BM, Hindorff LA, Jackson RD, Laurie CA, Laurie CC, Li Y, Lin DY, Moreno-Estrada A, Nadkarni G, Norman PJ, Pooler LC, Reiner AP, Romm J, Sabatti C, Sandoval K, Sheng X, Stahl EA, Stram DO, Thornton TA, Wassel CL, Wilkens LR, Winkler CA, Yoneyama S, Buyske S, Haiman CA, Kooperberg C, Le Marchand L, Loos RJF, Matise TC, North KE, Peters U, Kenny EE, Carlson CS. Genetic analyses of diverse populations improves discovery for complex traits. Nature 2019; 570: 514–518. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-019-1310-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) ## REFERENCES 1. 1.Custovic, A. et al. The Study Team for Early Life Asthma Research (STELAR) consortium ‘Asthma e- lab’: team science bringing data, methods and investigators together. Thorax 70, 799–801 (2015). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjg6IjcwLzgvNzk5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDMvMDUvMjAyMy4wMy4wMy4yMzI4NDQ0OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 2. 2.Boyd A, Golding J, Macleod J, Lawlor DA, Fraser A, Henderson J, Molloy L, Ness A, Ring S, Davey Smith G. Cohort Profile: The ‘Children of the 90s’--the index offspring of the Avon Longitudinal Study of Parents and Children. Int J Epidemiol. 42, 111–27 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dys064&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22507743&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000316699300012&link_type=ISI) 3. 3.Fraser, A. et al. Cohort Profile: the Avon Longitudinal Study of Parents and Children: ALSPAC mothers cohort. Int J Epidemiol 42, 97–110 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dys066&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22507742&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000316699300011&link_type=ISI) 4. 4.Custovic, A. et al. The National Asthma Campaign Manchester Asthma and Allergy Study. Pediatr Allergy Immunol 13, 32–7 (2002). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1034/j.1399-3038.13.s.15.3.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12688622&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 5. 5.Martindale, S. et al. Antioxidant intake in pregnancy in relation to wheeze and eczema in the first two years of life. Am J Respir Crit Care Med 171, 121–8 (2005). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1164/rccm.200402-220OC&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15531754&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000226258400007&link_type=ISI) 6. 6.Atkinson, W. et al. Domestic aeroallergen exposures among infants in an English town. Eur Respir J 13, 583–9 (1999). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiZXJqIjtzOjU6InJlc2lkIjtzOjg6IjEzLzMvNTgzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDMvMDUvMjAyMy4wMy4wMy4yMzI4NDQ0OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 7. 7.Kurukulaaratchy, R.J., Fenn, M., Twiselton, R., Matthews, S. & Arshad, S.H. The prevalence of asthma and wheezing illnesses amongst 10-year-old schoolchildren. Respir Med 96, 163–9 (2002). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1053/rmed.2001.1236&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11905550&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000174203800005&link_type=ISI) 8. 8.Kurukulaaratchy, R.J. et al. Characterization of wheezing phenotypes in the first 10 years of life. Clin Exp Allergy 33, 573–8 (2003). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1046/j.1365-2222.2003.01657.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12752584&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000182776000007&link_type=ISI) 9. 9.Arshad, S.H. et al. Cohort Profile: The Isle Of Wight Whole Population Birth Cohort (IOWBC). Int J Epidemiol 47, 1043–1044i (2018). 10. 10.Oksel, C. et al. Distinguishing Wheezing Phenotypes from Infancy to Adolescence. A Pooled Analysis of Five Birth Cohorts. Ann Am Thorac Soc 16, 868–876 (2019). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 11. 11.Loh, P.R. et al. Reference-based phasing using the Haplotype Reference Consortium panel. Nat Genet 48, 1443–1448 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3679&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27694958&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 12. 12.McCarthy, S. et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet 48, 1279–83 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3643&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27548312&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 13. 13.Durbin, R. Efficient haplotype matching and storage using the positional Burrows-Wheeler transform (PBWT). Bioinformatics 30, 1266–72 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btu014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24413527&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000336095100011&link_type=ISI) 14. 14.Marchini, J. & Howie, B. Genotype imputation for genome-wide association studies. Nat Rev Genet 11, 499–511 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrg2796&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20517342&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000278998500012&link_type=ISI) 15. 15.Willer, C.J., Li, Y. & Abecasis, G.R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–1 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btq340&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20616382&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000281738900017&link_type=ISI) 16. 16.Pruim, R.J. et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26, 2336–7 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btq419&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20634204&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000281714100054&link_type=ISI) 17. 17. Dayem Ullah, A.Z., et al. SNPnexus: assessing the functional relevance of genetic variation to facilitate the promise of precision medicine. Nucleic Acids Res 46, W109–W113 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gky399&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29757393&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 18. 18.Stelzer, G. et al. The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Curr Protoc Bioinformatics 54, 1 30 1–1 30 33 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/cpbi.5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27322403&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 19. 19.Durinck, S. et al. BioMart and Bioconductor: a powerful link between biological databases and microarray data analysis. Bioinformatics 21, 3439–3440 (2005). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/bti525&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16082012&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000231360600020&link_type=ISI) 20. 20.Durinck, S., Spellman, P.T., Birney, E. & Huber, W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat Protoc 4, 1184–91 (2009). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nprot.2009.97&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19617889&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000268858800008&link_type=ISI) 21. 21.McLaren, W. et al. The Ensembl Variant Effect Predictor. Genome Biol 17, 122 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13059-016-0974-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27268795&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 22. 22.Wingett, S. et al. HiCUP: pipeline for mapping and processing Hi-C data. F1000Res 4, 1310 (2015). 23. 23.Cairns, J. et al. CHiCAGO: robust detection of DNA looping interactions in Capture Hi-C data. Genome Biol 17, 127 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13059-016-0992-2&link_type=DOI) 24. 24.Adsera, C.B., Park, Y.P., Meuleman, W. & Kellis, M. Integrative analysis of 10,000 epigenomic maps across 800 samples for regulatory genomics and disease dissection. 810291 (2019). 25. 25.Ihaka, R. & Gentleman, R. R: A Language for Data Analysis and Graphics. Journal of Computational and Graphical Statistics 5, 299–314 (1996). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=https://doi.org/10.1080/10618600.1996.10474713&link_type=DOI) 26. 26.Szklarczyk, D. et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47, D607–D613 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gky1131&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30476243&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) 27. 27.Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13, 2498–504 (2003). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjEwOiIxMy8xMS8yNDk4IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDMvMDUvMjAyMy4wMy4wMy4yMzI4NDQ0OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 28. 28.Branchett, W.J., Walker, S.A. & Lloyd, C.M. Experimental Mouse Models of Asthma and Analysis of CD4 T Cells. Methods Mol Biol 2285, 329–348 (2021). 29. 29.Brunekreef, B. et al. The prevention and incidence of asthma and mite allergy (PIAMA) birth cohort study: design and first results. Pediatr Allergy Immunol 13, 55–60 (2002). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1034/j.1399-3038.13.s.15.1.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12688626&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000180571000012&link_type=ISI) 30. 30.Wijga, A.H. et al. Cohort profile: the prevention and incidence of asthma and mite allergy (PIAMA) birth cohort. Int J Epidemiol 43, 527–35 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dys231&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23315435&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F03%2F05%2F2023.03.03.23284449.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000335919500022&link_type=ISI)