Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Sep 1.
Published in final edited form as: J Allergy Clin Immunol. 2016 Jun 11;138(3):676–699. doi: 10.1016/j.jaci.2016.02.045

Resolving the Etiology of Atopic Disorders by Genetic Analysis of Racial Ancestry

Jayanta Gupta a, Elisabet Johansson a, Jonathan A Bernstein b, Ranajit Chakraborty c, Gurjit K Khurana Hershey a, Marc E Rothenberg d, Tesfaye B Mersha a,*
PMCID: PMC5014679  NIHMSID: NIHMS797380  PMID: 27297995

Abstract

Atopic dermatitis (AD), food allergy (FA), allergic rhinitis (AR) and asthma are common atopic disorders of complex etiology. The frequently observed “atopic march” from early AD to asthma and/or AR later in life as well as the extensive comorbidity of atopic disorders, suggests common causal mechanisms in addition to distinct ones. Indeed, both disease-specific and shared genomic regions exist for atopic disorders. Their prevalence also varies among races; for example, AD and asthma have a higher prevalence in African-Americans when compared to European-Americans. Whether this disparity stems from true genetic or race-specific environmental risk factors or both is unknown. Thus far, the majority of the genetic studies on atopic diseases have utilized populations of European ancestry, limiting their generalizability. Large cohort initiatives and new analytic methods such as admixture mapping are currently being employed to address this knowledge gap. Here we discuss the unique and shared genetic risk factors for atopic disorders in the context of ancestry variations, and the promise of high-throughput “-omics” based systems biology approach in providing greater insight to deconstruct into their genetic and non-genetic etiologies. Future research will also focus on deep phenotyping and genotyping of diverse racial ancestry, gene-environment, and gene-gene interactions.

Keywords: Atopic march, food allergy, atopic dermatitis, allergic rhinitis, asthma, racial ancestry, admixture mapping, phenotyping, gene-environment interaction, omics

Introduction

The development of atopic disorders often follows an age-dependent pattern that starts early in life and progresses (marches) from one tissue to another in the same individual, a phenomenon referred to as the “atopic march”13 (Figure 1). The International Study of Asthma and Allergies in Children (ISAAC), which examined the global prevalence of atopic dermatitis (AD), food allergy (FA), allergic rhinitis (AR), and asthma, found that the prevalence of these disorders differed among races.4,5 Delineating genetic and environmental risk factors that relate more specifically to certain atopic disorders and ancestry than to other factors provides valuable information to address the following questions: Are atopic disorders genetically inherited and/or inter-related? Do all atopic disorders share the same loci or are there unique genetic loci associated with each atopic disorder? Are there ancestry differences in the risk of atopic disorders and what are their potential sources? Is an individual ancestry or race a good predictor of atopic disorders? In this review, we discuss the genetics of atopic disorders, racial ancestry risk differences, role of gene-environment interactions, and recent evidence of shared and unique etiologies of atopic disorders. As the world is becoming highly multiethnic and racially admixed, and, thus, genetically diverse, individuals who self-identified as a particular race may have widely varying genetic ancestry.6,7,8 Thus, we discuss admixture mapping approach from the vantage point of disentangling ancestry-specific risk loci. Enormous progress has been made on developing high-throughput genotyping technology but focus on devising high-throughput methods to standardize phenotyping is still at its infancy. Our review emphasizes the need for high-throughput methodologies for constructing causative models that will help resolve the underlying clinical and ancestral complex architecture of atopic disorders to yield greater insights. These approaches have the potential to target atopic phenotypes with interventions relevant to specific disorders and ancestry group. In addition, we provide an overview of the complex components of atopic disorders, as well as future research strategies.

Figure 1. Schematic diagram illustrating the age-dependent progression “atopic march” of atopic disorders.

Figure 1

Food allergy and atopic dermatitis peak in the first years of life and decline after that time. Asthma and allergic rhinitis increase over time as sensitization develops further. Food sensitization can be used as an early indicator for identifying children at risk for subsequent allergic disease who may benefit from early intervention.

Atopic Disorders and Race

It has long been recognized that there are differences among racial and ethnic groups in the prevalence and severity of atopic disorders,912 and these differences are believed to have both environmental and genetic components.1315 It is notable, however, that there are racial and ethnic minority populations in the U.S. that are under-represented in cohort studies and clinical trials and that the vast majority of genome-wide association studies (GWAS) of atopic disorders conducted thus far have used populations of European ancestry. Genomic studies often do not translate well from one race to another due to ancestral variation. Different populations have different linkage disequilibrium (LD) structures (otherwise known as haploblocks structures), allele frequencies and effect sizes16 and, thus, single-nucleotide polymorphism (SNP) associations may be population-specific.17 As a consequence, our current knowledge might be skewed toward the European ancestry–based etiology of atopic disorders, and important genetic and environmental clues to population differences may have been missed. This is because most of the risk alleles and index variants are derived using commercial genotyping arrays ascertained from populations of European ancestry. This drawback biases the generalization of results of these studies to other ancestral populations since not all atopic disorder loci found in Europeans are transferable to all populations. Identification of causal variants through trans-ancestry fine mapping and sequencing in different populations will help to understand the genetic contribution to the disparity in atopic disorder prevalence. In particular, the differential and lower level of LD in African ancestry will likely greatly improve the fine mapping of causal variants using trans-ancestry meta-analysis efforts. In addition, many of the diagnostic tools using in atopic diseases such as pulmonary function tests are developed mainly for Europe ancestry. However, marked differences in lung function and allergic sensitization between children of different racial backgrounds exist even when known confounders including socioeconomic status are taken into account.1820 For example, spirometric assessment of forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC) are known to be reduced by 14% in individuals of African ancestry compared with those of European ancestry.2124 The use of race-specific reference equations can help to minimize such differences and improve accuracy.20,25

Racial ancestry differences can have implications for clinical practice; an example is the use of long-acting β2-agonists (LABAs) in patients with asthma. Though combining corticosteroid treatment with a LABA can lead to better asthma control for most patients, a small proportion of patients, including African Americans, appear to be at increased risk for harmful effects and even fatal outcomes from the use of LABAs.26 The reason for this observed difference is still controversial. Some studies have suggested that genetic differences in the β2-adrenergic receptors and their interaction with race may be responsible.27,28 Several studies have shown that African American asthmatics have an increased likelihood for treatment failures and overall differential response to treatment that may be caused by genetic variants specific to their ancestry.29,30 However, race is a crude proxy to genetic ancestry and falls short of explaining the variation in response to medication.6 Investigators trying to address the knowledge gaps in our understanding of the racial aspects of atopic disease are faced with several challenges. Assembling minority cohorts large enough to power the detection of genetic associations can be difficult and requires very active outreach, especially to racial/ethnic groups that may be underserved by the health care system. Another approach is to increase power by using newer statistical methods such as admixture mapping. In conventional association studies, the high degree of recent genetic admixture in many racial minorities in the U.S. can lead to population stratification—ancestry differences between cases and controls. It is well recognized that racial minorities in the U.S., including African-Americans and Latino groups, have a lower socio-economic status, which is often associated with environmental factors such as diet, presence of allergens, and pollution exposure.15 These factors can have a direct effect on the development of allergic disorders and need to be carefully adjusted for in statistical analyses.31

Different ancestral populations have different proportions of rare genetic variants, and it has been shown that populations of African ancestry have up to three times as many rare variants as populations of European and Asian origin.32 Although rare variants are believed to be enriched for functionality,33,34 they present a challenge in association studies because of the large sample sizes needed for their detection.35,36 Trans-ancestry studies have been invaluable in deconstructing the genetic architecture of complex diseases, such as asthma. They provide an opportunity to replicate signals in independent populations and for meta-analyses to boost statistical power. For example, African genome analysis enables prioritizing candidate variants in follow-up and fine-mapping functional variants due to the unique short-range LD in African ancestry. The use of a population with short-range LD will result in the greatest localization success rate in distinguishing the causal SNP from its neighbors.3739 The recent completion of whole genome sequencing data from the 1000 Genomes Project (http://www.1000genomes.org), encompassing over 88 million variants from 26 world-wide populations (2,504 individuals), will make it easier to optimize LD pattern, imputation and SNP selection for specific racial populations, thereby increasing power in GWAS.40

Genetics of Atopic Disorders

The strongest evidence for the importance of genetic factors in atopic disease stems from twin studies. The approximately 80% concordance rate for AD among monozygotic (MZ) twins far exceeds the concordance rate of 20% observed among dizygotic (DZ) twins.41,42 Heritability estimates for atopic diseases are as high as 81% for FA, 84% for AD, 91% for AR, and 95% for asthma.43 Although FA, AD, AR, and asthma are clinically diagnosed as discrete diseases, current evidence suggests that they are genetically related conditions, with correlation estimates being 0.55 for asthma and AD, 0.47 for asthma and AR, and 0.62 for AD and AR.44 The phenotypic similarity among atopic disorders suggests that elements of biological etiology may also be shared among these disorders. Risk genetic variants that are shared or influence multiple atopic disorders, also known as “pleiotropic” effects, can help explain common pathogenic features and provide suggestions for development of novel therapies.

Atopic disorders are considered a type I hypersensitivity immune response associated and/or mediated in part by immunoglobulin E (IgE) antibodies to environmental antigens (Table 1). Pathways that have been associated with atopic disorders fall into several broad categories, such as those involved in the epidermal barrier function and those involved in regulating the innate and adaptive immune response, including IgE sensitization45,46 (Table 2). Lists of disorder-specific (Fig. 2A for FA; Fig. 2B for AD; Fig. 2C for AR; and Fig. 2D for asthma) enriched genes based on over 15.74 million PubMed abstracts are shown using Acumenta Biotech Literature Lab.47 The genes are ranked by the relative intensity of their co-occurrence with each atopic disorder. The ranking is shown with the most intensely related genes starting at the 12 o’clock position in the inner ring and descending in a clockwise direction. Table E1 shows list of genes ranked in the order of co-occurrence with each atopic disorder. To understand the relationship among atopic disorders in terms of their genetic etiology, we investigated genetic overlap using ranked gene lists from each disorder. Among the top ranked genes listed in Figure 2, there were 43 gene overlaps between AR and asthma, 29 between AD and asthma, 28 between FA and asthma, 27 between AR and AD, 22 between FA and AD, 22 between FA and AR, and 16 among all disorders (Figure 3). AR and asthma commonly coexist and are regarded as “unified airways disease.”48 Thus, there appeared to be more genetic commonalities between AR and asthma than between AR and FA or between AR and AD. To characterize the main functional networks/pathways underlying the 16 shared genes (IL5, IL4, TSLP, IL10, RNASE3, IGHG4, IL13, CCL11, IFNG, RNASE2, FCER2, CD4, FOXP3, IL4R, KCNE4, and CCL26) among atopic disorders, we carried out network analysis using Ingenuity Pathway Analysis (IPA). Figure 4 and Table E2 show the most overrepresented network and functional ontologies among these shared genes, respectively. There is 1 significant network which lies in regions of high network connectivity (“hubs”) centered at IL4, a key mediator of allergic airway diseases.49

Table 1.

Common atopic disorders

Atopic disorder Clinical symptoms Allergens
Food allergy Rash, especially around the mouth; sometimes diarrhea Peanuts, tree nuts, milk, egg, fish
Atopic dermatitis Chronic, relapsing, intense itching, particularly around skin flexures at the wrist, elbow, ankle, and knees Egg, cow’s milk, wool
Allergic rhinitis Blocked and runny nose, sneezing, and itching and streaming eyes Intermittent Pollen, house dust mite, animal dander
Asthma Episodic or recurrent breathlessness with cough, wheezing, and chest tightness Pollen, house dust mite, animal dander, molds, food allergens, viral infections, drugs, smoke

Table 2.

Select pathways involved in atopic disorders

Disorder Pathway Functions Reference
Atopic disorders Innate immunity and immunoregulation Triggering immune response
Immunoregulatory
Antigen presentation
306,307
Th2-cell differentiation and effector functions Th2-mediated responses 308,309
Epithelial biology and mucosal immunity Chemoattractants
Epithelial barrier integrity
Protection against proteases
310
Lung function, airway remodeling, and disease severity Formation of epithelial-mesenchymal trophic unit
Negative regulation of the TLR and IL-1R pathway
Bronchial smooth muscle relaxation
Allergic airway inflammation and airway remodeling
311,312

Figure 2. Genes associated in the literature with each atopic disorder.

Figure 2

Figure 2

Figure 2

Figure 2

Based on mining of 15.73 million PubMed abstracts ((01/01/90 to the present) by Literature Lab™ from Acumenta Biotech,57 genes enriched in FA, AD, AR, and asthma are shown. The rankings of relative strengths of the gene/disease associations in the literature are as follows. The genes with the strongest relationship to each atopic disorder revolve around the center as measured by LPF (log of the product of frequency) starting at the 12 o’clock position in the inner ring descending in a clockwise direction, as indicated by arrows, and outward. Thus IGHG4, FLG, RMASE3, and IL5 have the most co-occurrence and genes MIR604, HRH1, GBP5, and ALOX5AP have the least co-occurrence with FA, AD, AR, and asthma, respectively. The LPF measures relative intensity or strength of co-occurrences for pairs of items in a large corpus, in this case genes and diseases.47 The LPFs are computed by Literature Lab™ for co-occurrences of all human genes in the NCBI Gene Database and each disease identified in the NCBI MeSH Diseases ontology (about 4,200 terms) in 15.73 million PubMed abstracts. The LPF is calculated as: LPF= Lg(x/G * x/T), where G is number of genes, T is the number of terms and x is co-occurrence between G and T. The greater the absolute magnitude of the LPF the weaker the association ; the closer the LPF value to zero, the stronger the association.

Figure 3. Venn diagram of unique and shared genes among atopic disorders (FA, AD, AR and asthma).

Figure 3

There are more genes shared between AR and asthma than between any other pair of atopic disorders. To understand the relationship among atopic disorders in terms of their genetic etiology, we investigated genetic overlap using ranked gene lists from each disorder. Among the top ranked genes listed in Figure 2, there were 43 gene overlaps between AR and asthma, 29 between AD and asthma, 28 between FA and asthma, 27 between AR and AD, 22 between FA and AD, 22 between FA and AR, and 16 among all disorders. AR and asthma commonly coexist and are regarded as “unified airways disease.”48 Thus, there appeared to be more genetic commonalities between AR and asthma than between AR and FA or between AR and AD. However, it is to be noted that our overlap measure uses gene lists in each disorder based on studies in the literature so far, and these diseases are not equality studied (for example, asthma has 110,428 PubMed abstract compared with 6,348 for AR). As we generate more data for all diseases, we will be able to determine disease-specific or overlapping genes with more confidence.

Figure 4. IPA network for 16 genes shared among FA, AD, AR and asthma.

Figure 4

Genes with red nodes are focus genes in our analysis, the others are generated through the network analysis from the Ingenuity Pathways Knowledge Base (http://www.ingenuity.com). Edges are displayed with labels that describe the nature of the relationship between the nodes. The lines between genes represent known interactions, with solid lines representing direct interactions and dashed lines representing indirect interactions. Nodes are displayed using various shapes that represent the functional class of the gene product. Examination of the networks would move us toward a holistic understanding of atopic disorders.

As expected, the majority of the sixteen genes associated with all four atopic diseases are involved in fundamental inflammatory processes, and many of them, IL4, IL4R, IL13, IL5, IGHG4, FCER2 and TSLP, specifically function in the Th2 cell immune response characteristic of allergy. The eotaxins CCL11 (eotaxi-1) and CCL26 (eotaxin-3), and the RNases RNASE2 and RNASE3, as well as IL5, are important for eosinophil recruitment and function, also a hallmark of allergic disease. IL10 and FOXP3 are important for the activity of regulatory T cells. IL10 plays an essential role in inducing an immunoregulatory phenotype in B cells that exerts substantial anti-inflammatory and immunosuppressive functions.50 IL5 is the most important interleukin responsible for eosinophilic airway inflammation in asthmatics, and has become a definite target for treatment.51 IL5 is produced by CD4+ Th2 cells, mast cells, eosinophils, basophils and innate lymphoid cells type 2 (ILC2) and is involved in several steps of eosinophil development and function.52 Others, such as IFNGγ and IL17A are involved in the functioning of Th1 cells and Th17 cells, respectively. Several of the sixteen genes, CCL11, IFNγ, IL17A, RNASE2, and RNASE3, also have antimicrobial activity, but it is unclear what role, if any, this activity has in the development of allergic responses. TSLP is produced by epithelial cells and links the innate immune response to Th2 cell-driven inflammation. Its association with all four allergic diseases suggests the important role of the epithelial barrier in allergic response. Despite these findings, overlapping genes are based on studies in the literature so far, and these diseases are not equality studied (for example, asthma has 110,428 PubMed abstracts compared with 6,348 for AR). As we generate more and more data to all diseases, we will be able to determine disease-specific or overlapping genes with more confidence.

Recent studies suggest that the most important factor that predicts atopic march is an impaired epidermal barrier mechanism of the epithelium with multiple organ-specific inflammation involvement (i.e., skin, nose, airway, gut/gastrointestinal tract)53 (Figure 5). A better understanding of the role of epithelial dysfunction along with crosstalk with innate immunity in the lung, skin, and gut in ancestry-specific manner could provide a framework on which new therapeutic strategies can be planned to prevent exacerbations and to alter the natural course of these disorders. Figure 6 represents a conceptual model that deconstructs the molecular and genetic factors in the pathogenesis of atopic disorders. Major genes related to epidermal protease–inhibitor function, epidermal development, keratinocyte morphology/function, lipid metabolism, anti-oxidant function, cytokine/chemokine/growth factors, and innate immunity are shown. Genes related to protease function, epidermal development, and keratinocyte morphology can directly contribute to epidermal barrier dysfunction, whereas lipid genes can modulate barrier function, inflammation, and anti-oxidant function. Genes related to cytokine/chemokine function modulate lymphocyte activity, as well as the expression of innate immune genes. Therefore, atopic disorders can be viewed as a collection of related diseases, characterized by clinical manifestations in several different organs that are linked through similar pathophysiological processes including epidermal barrier dysfunction and cutaneous inflammation, environmental insults such as allergen challenge and microbial infections and “allergic” Th2 and Th17/Th22 cell responses.54

Figure 5. The genetic and environmental interplay of atopic disorders.

Figure 5

The relationship between genetic and environmental factors determine the overall outcome of atopic disorders. Skin barrier dysfunctions are part of the spectrum of allergic disorders with immunoglobulin E (IgE)-mediated sensitization and T helper type 2 (Th2) immune dysregulation. The interactions between epidermal barrier dysfunction and dysregulation of innate and adaptive immunity along with environmental risk factors contribute to the pathogenesis of atopic disorders.

Figure 6. Deconstructing atopic disorders using genetic and molecular information.

Figure 6

Complex interplay between functional gene clusters and interpretation based on a model that integrates each atopic disorder in a central theme are presented. A mechanism by which epidermal barrier dysfunction may lead to inflammation and allergic sensitization in atopic disorders is shown. In parallel, genetic and molecular analysis of atopic disorders have evidenced the key role played in the disease mechanisms by epithelial skin cells, especially epidermal keratinocytes, with an abnormal pattern of production of cytokines and chemokines that could trigger sustained, chronic inflammation. Current data favor the paradigm shift in which the downstream systemic effect of allergen penetration through the impaired skin barrier causes immune cells to mount an exaggerated inflammatory response at any allergen-exposed epithelial surface.304,305 A decreased skin barrier response allows for increased susceptibility of the skin to allergens. In addition, decreased skin barrier leads to an increase in skin pH, altered keratinocyte adhesion properties, and both increased serine protease activity and inflammation. Among multiple reported genes relevant to the cellular location of gene products, only the well-established ones are shown for clarity.

Systematic reviews have revealed that several candidate genes are clustered in chromosomal regions that have been linked to all atopic disorders, such as chromosome 5q31-32, 6p21.3, 11q13, and 12q14-24.55 Their close proximity in linked regions suggest increased potential for inheritance of all of these disorders simultaneously.56 Multiple studies including our 15.73 million PubMed abstracts search by Literature Lab™ from Acumenta Biotech have shown that several genes, including IL13, IL4 and IL5, belong to pathways that are common to atopic diseases.57 However, there are also disorder-specific genes. Examples of primary disorder-specific genes include ADAM33 and ORMDL3 for asthma,58 FLG for AD, and IL6 for AR59 and recently CAPN14 for the FA-related disorder eosinophilic esophagitis.60 Using the results of recently published SNPs from the GWAS catalog (www.genome.gov/gwastudies) and PhenoGram visualization software,61 we showed shared and unique genetic etiologies among atopic disorders grouped by ancestry (Figure 7). Chromosome 6 shows much of the genomic overlap both at the SNP and ancestry level for atopic disorders. Genetic loci that are shared among atopic disorders, as well as ancestries, would indicate shared molecular pathways in the pathogenesis of atopic march. On the other hand, the lack of shared etiology among atopic disorders and ancestries would indicate distinct pathogenic mechanisms or pathways underlying each atopic disorder and ancestry. A partial overlap would suggest that the combination of disease-specific and disease-overlapping genetic risk factors drive the progression of atopic disorders. Thus, studying the genetic/molecular determinants of these disorders will eventually enable us to deconstruct atopic disorders into distinct genetic/molecular phenotypes, leading to more customized disease management options. The challenges in disentangling atopic disorders include the very large sample sizes required to delineate associations that are specific to each atopic disorder, which results from the over 80% overlap among atopic disorders.43 Omics approaches such as DNA sequence variation (genomics), genetic variants in regulatory regions (regulomics), gene expression (transcriptomics), regulation of gene expression (epigenomics), protein modification (proteomics), microbes (microbiomics), metabolites (metabolomics), and environmental exposure (exposomics) could help to identify molecular signatures and unravel the interrelationships between these disorders and diseases pathogenesis. In addition, the application of a systems biology approach could help to examine shared and unique genetic risk factors and functional categories among atopic disorders to identify at-risk individuals in order to prioritize them for early preventive interventions.

Figure 7. Atopic disorder NHGRI GWAS catalog variants grouped by racial ancestry.

Figure 7

Atopic disorder–related variants identified through genome-wide association studies (GWAS) [NHGRI catalogue of published GWAS was searched using the following terms: food allergy, atopic dermatitis, asthma, and allergic rhinitis]. PhenoGram was used to plot and visualize NHGRI GWAS catalog association results for potentially pleiotropic single-nucleotide polymorphisms (SNPs) in atopic disorders.61 An Ideogram of all 22 chromosomes is shown, along with the X and Y chromosomes. Horizontal lines on the chromosomes correspond to the base-pair location of each SNP, while lines which project from the chromosomes connect the SNP to colored circles that represent the phenotype(s) associated with the SNP. Overlapping and allergy-specific susceptibility loci among ancestries from GWAS might help to understand the putative pathogenetic relationship between allergy-related phenotypes and racial ancestry. However, loci from GWAS should be interpreted with caution, due to potential issues such as limited sample size in minority populations. Longitudinal and deep phenotyping (measurements at different molecular levels), of individuals along with information on genetic and disease specific environmental risk factors are relevant to study atopic disorders across racial ancestry.

Gene-Environment Interactions in Atopic Disorders

The pathogenesis of atopic disorders usually involves the complex interplay of genetic and environmental factors, which can be assessed statistically as gene-environment interactions. For example, several studies implicate that in utero and early-life environmental tobacco smoke (ETS) exposure leads to impaired lung function and increased risk of asthma;62 the ETS exposure modifies the strength of the association between genetic variants and asthma.6369 Studies of gene-environment interactions can involve one or more specific candidate genes or comprehensive whole-genome scans. Since much larger sample sizes are needed to detect interactions than main effects, the cohort size required to power genome-wide environmental interaction (GWEI) studies can be prohibitive, and thus far most studies of gene-environment interactions have employed a candidate-gene approach.67,7072 Another challenge in conducting GWEI studies is the absence of well-characterized cohorts with regard to environmental variables. The best characterized, allergy-related gene-environment interaction to date involves SNPs in the microbial sensor CD14, which interacts with several toll-like receptors and plays a critical role in the innate immune response to microbial insult. Although most studies have focused on the impact of postnatal environmental exposures, there is increasing evidence that prenatal exposures may also be important.7375 Epidemiologic studies examining the effect of prenatal exposures on the development of allergic disease have shown that maternal exposure to farming environments during pregnancy is associated with decreased rates of AD, AR and asthma in their children.76 Early exposure to endotoxin, a bacterial cell wall component, appears to offer protection against allergic sensitization and asthma.77 Several reports have demonstrated a modifying effect of a SNP in the promoter region of the microbial sensor CD14 on the relationship between endotoxin exposure and atopy.7880 Polymorphisms in CD14 have also been shown to interact with microbial exposures81 and pet exposure,82 as well as environmental tobacco smoke,83 on the outcome of allergic disease.

Environmental factors associated with racial and ethnic groups are major confounders in genetic association studies.84 However, there are very few published reports on race-specific gene-environment interactions to date. An example of a gene-environment interaction modified by race/ethnicity is the impact of endotoxin exposure on the innate immune system. Using a birth cohort that had 53% African American and 38% European American ancestry children, Williams et al.85 showed that interaction between the CD14 genotype and endotoxin exposure on the number of regulatory T cells was limited to the African American subjects. Furthermore, in a study of CD14 gene-environment interaction in the U.S. Latino population, Shoudhry et al.68 found that a modifying effect of the CD14 genotype on the association between IgE levels and exposure to ETS was specific to the U.S. Puerto Rican subjects and not observed in the U.S. Mexican subjects. Importantly, Puerto Ricans have a higher admixture of African ancestry than do Mexicans.86 ETS has also been shown to interact with the IL4 genotype on the outcome of wheezing in infants.87 This effect was specific for the African American population, and neither ETS exposure nor IL4 genotype alone had a significant effect on wheezing. These studies show that race-specific gene-environment interactions can be demonstrated in relatively smaller cohorts using a hypothesis-driven, candidate-gene approach.

The major challenges in uncovering gene-environment interactions in population-based GWAS include inadequate sample size/power, extensive computational demand and the burden of multiple testing for the total numbers of SNPs.88 A potential approach to reduce the burden of multiple testing in relatively small sample sizes is to use admixture mapping (AM) and limit association testing to regions that exhibit different risk allele frequencies among ancestral populations. AM performs many fewer independent tests (i.e., smaller penalty due to multiple tests) across the genome than current GWAS.8993 Modeling interaction between ancestry and environmental exposures could potentially reveal effect modification on asthma risk.84 Large cohort initiatives such as the Genetics of Asthma in Latino Americans (GALA) study,68,94 as well as new analytic strategies,91 are expected to facilitate more systematic genome-wide searches for gene-environment interactions in the future.95 Furthermore, assessing high-throughput phenotype and exposure definitions with careful adjustment for potential confounders increases the chances of finding significant interactions. However, standardizing environmental exposure assessment methods for “environmental phenotyping” is needed. For example, cotinine measurement in biofluids may be a better and more objective way to assess exposure to environmental tobacco smoke than the use of questionnaires because of underreporting and/or exposure outside the home.96,97 The progressive accumulation of phenotypic differences between genetically identical MZ twins living in different environments illustrates how pollution, smoking, mold, and diet can shape human susceptibility to diseases. MZ twins were epigenetically indistinguishable early in life but as they adopted different lifestyles, substantial differences in disease susceptibility arose over time.98 Therefore, MZ twin discordance for atopic disorders could be interpreted as the result of environmental factors that modulate susceptibility through a change in the profile of epigenetic modifications that ultimately determine gene function. In addition to gene-environment interactions, gene-gene interactions, also known as epistasis, likely play an important role in the genetics of atopic disorders.99,100

Genetics of Food Allergy

FA is frequently associated with AD, and together they are associated with the development of AR and asthma later in life.101 Many epidemiological studies have suggested that early life food sensitization is associated with an increased risk of AD, AR and asthma later in life.102106 FA has a strong familial aggregation.107 A child with a parent or sibling with peanut allergy, one of the most common forms of FA, has a 7 times higher risk of developing the condition than do children without familial risk factors.108 Twin studies have demonstrated that the concordance of FA in MZ twins is much higher than in DZ twins,109,110 with concordance rates of 64.3% and 6.8% in MZ and DZ twins, respectively. Though the high concordance in MZ twins suggests a strong genetic component, environmental factors also contribute to the disease since the MZ twin concordance is not 100%. The same study estimated the heritability of FA to be as high as 81%, which also does not rule out roles of common family environmental factors. Thus far, multiple genes and their variants have been interrogated for their association with FA. Since FA, like AD, was primarily thought to be a disease of the immune system, all of the genes investigated and found to be associated with FA, aside from FLG, were immune-related and included the major histocompatibility complex cluster, CD14, FOXP3, STAT6, SPINK5, TNF, GSTP1, IL13, IL10,111113 and IL4RA.114 In general, these were small-scale, candidate-gene studies that lacked replication, and the findings were frequently inconsistent among studies.

In light of the very strong association of null mutations in the FLG gene with AD that was first discovered in 2006 and subsequently replicated in more than 30 studies, Brown et al. hypothesized that FLG would be a candidate gene for FA. Indeed, they found that FLG was associated with peanut allergy regardless of AD status.115 This was the first genetic evidence that FA, like AD, was at least in part due to a defect in the skin barrier. In a follow-up study, the same group demonstrated the association of FLG with peanut allergy regardless of asthma status.116 Though still a matter of debate, it has been hypothesized that in children with FA, sensitization to peanuts primarily occurs through a defective skin barrier rather than through ingestion.117 The first GWAS of well-defined FA in 2,197 participants of European ancestry from the Chicago Food Allergy Study, including specific subtypes (peanut, milk, and egg), identified peanut allergy–specific loci in the HLA-DR and HLA-DQ gene region at 6p21.32.118 This gene region accounted for approximately 20% of PA in the study population. These associations were replicated, and the variants were associated with differential DNA methylation levels at multiple CpG sites.118

Available evidence suggests that FA, like AD and asthma, is the result of a complex interaction between genetic and environmental factors that could include dietary habits (timing of food introduction and route of exposure, type of food (e.g., reduced consumption of specific fatty foods), strict hygiene, ETS exposure, prematurity, low birth-weight, and vitamin D deficiency among other factors.119,120 Available data indicate that there has been a rise in the prevalence of FA in recent times.119 Indeed, data from the National Health Interview Survey showed that in the age group of 0–17 years, the proportion of children with FA increased from 3.4% in 1997–1999 to 5.1% in 2009–2011.121 Since widespread fundamental and heritable sequence changes in the human genome are very unlikely during the span of a few decades, it is assumed that environmental factors are responsible for the rise in prevalence of FA.

Epigenetics, which includes changes in gene expression due to environmental influence without alteration in the DNA sequence, has been implicated. It has been suggested that heritable epigenetic changes resulting from the varying rate of development of tolerance to inhaled and ingested food allergens could be the reason for the current epidemic of FA.122 Indeed, food can be a strong inducer of epigenetic modification, and maternal diet has been proposed as a risk factor behind the subsequent development of FA in the offspring.112 A recent epigenome-wide association study that investigated the DNA methylation profile of CD4+ T cells, which are instrumental in regulating the allergic response, found differences in methylation in children with FA compared to normal controls, providing fresh evidence of epigenetic regulation of FA.123

Food Allergy and Race

Despite FA affecting between 1%–10% of all American children,124 many investigators have noted that the prevalence of FA varies among races. Specifically, studies have generally found African ancestry to be a risk factor for food sensitization and FA compared with European ancestry. In the multiethnic Boston Birth Cohort consisting chiefly of Black and Hispanic children, Kumar et al. noticed food sensitization to be associated with both self-reported Black race and African ancestry.125 In a large, nationwide cross-sectional survey, Gupta et al. found both Black and Asian children to be at a higher risk and Hispanic children to be at a lower risk of developing FA compared with White children.126 In a clinic serving low-income, minority populations in New York, Taylor-Black and Wang identified a higher prevalence of FA allergy among African American children compared with children who were of Hispanic or multi-racial origin.127 Preliminary data from a longitudinal, general birth cohort study in which skin testing for food allergens was performed indicated higher rates of sensitization in African American children than in their White counterparts.128 Data from the National Health Interview Survey, 1997–2011, demonstrated that the prevalence of FA was lower in Hispanic children than in non-Hispanic, White and non-Hispanic, Black children.121 A meta-analysis by Keet et al. found that despite the prevalence of self-reported FA among American children rising over the last 20 years, Black children as a group have experienced the greatest increase.129 Though the data generally tend to support a higher prevalence of FA in African Americans, there have also been reports that contradict this claim. In a study that included 2 public schools and 2 charter schools from New York, Taylor-Black et al. did not find a higher rate of FA among Black children compared to White children.130 A recent meta-analysis by Greenhawt et al. concluded that though there was some evidence of increased rates of food sensitization and FA among the Black population, the available data were still insufficient to label Black race as a risk factor for FA.131

Even if differences truly exist among racial groups in their susceptibility to FA, the reasons behind such differences are not readily apparent. Though it is tempting to point to genes, it is to be noted that self-reporting as “African American”, “Hispanic”, or “Latino” could also be a surrogate for environmental variables including diet, socioeconomic status, and other lifestyle factors.125 For example, African American children of families with significantly lower income were found to be at the greatest risk for FA and food sensitivities, pointing to the probability that discrete environmental and genetic factors are involved.110 Though many of the studies did adjust for some of these variables, the possibility of residual confounding factors cannot be excluded. Thus, methodological variation precludes firm inter-racial comparison of FA. Another possibility is gene-environment interaction. In a recent study, Koplin et al. found increased prevalence of peanut allergy among infants with parents born in East Asia when compared with infants who had parents born in Australia or Europe.132 Notably, Asian-born parents did not exhibit higher prevalence of FA compared with Australian-born parents. The authors speculated that although genetically susceptible, Asians born in Asia do not develop allergies due to protective effects of the environment in their native countries and that since Asian infants born in Australia lack this putative protective umbrella, the deleterious effects of allergy genes would be less restrained. Thus, the relationship between specific IgE level and skin prick test sensitization to FA needs to be considered within the context of race.133

Genetics of Atopic Dermatitis

AD is one of the most common skin disorders and affects 5–20% of infants worldwide with lesser prevalence in adulthood.134 The pathogenesis of AD is complex and requires the interplay of genetic and environmental risk factors. It has long been known that AD clusters in families. About 56% of the children who had a parent affected with AD developed the disease.135 Twin studies of AD have demonstrated concordance rates of up to 86% in MZ twins and up to 50% in DZ twins, respectively.136 Candidate-gene studies in AD have reported associations with numerous genes, the majority of which belong to one of two broad biological pathways, the epidermal barrier maintenance pathway (FLG) or the immune response pathway (e.g., CD14, IL4, IL4RA, IL13, TLR2, SPINK5, and RANTES).136 A total of 6 GWAS and GWAS meta-analyses have been performed on AD to date. These studies have identified 19 loci that reached the genome-wide significance level.137 These loci include 1p21.3, which contains the FLG gene, and 5q11.1, which is adjacent to the TSLP gene. The FLG gene, discussed in detail below, was first identified as a strong candidate for AD in 2006138 and has since been associated with AD in numerous studies conducted in people of European and Asian ancestry.139 The TSLP gene, which acts as a promoter of T helper type 2 (Th2) cell differentiation and an activator of Th2 cytokine–mediated inflammation140 has also been associated with AD in recent reports.141,142

Multiple studies have shown a link between AD and filaggrin, a protein that maintains skin and mucosal barrier function. The gene encoding filaggrin, FLG, is located within the epidermal differentiation complex (EDC), a cluster of genes in chromosome 1q21 that are involved in maintaining an intact skin barrier. The association of loss-of-function mutations in FLG with AD is one of the strongest associations to have been uncovered in complex disease genetics to date.143 Carriers of specific loss-of-function mutations in FLG can have an approximately 3-fold greater chance of developing AD than do non-carriers.139 This discovery has led to a paradigm shift in the understanding of AD, strengthening the “outside-inside” hypothesis, which postulates that AD is primarily a disease of the skin barrier. This is in contrast to the long-prevailing “inside-outside” hypothesis, which maintains that AD is primarily a disease of immune dysregulation. However, a skin barrier defect alone may not be sufficient for the development of AD. Though allergen entry and sensitization occur through a defective epidermal barrier, a favorable Th2 milieu already present in genetically predisposed individuals is also required for typical lesions of AD to develop. Indeed, as discussed earlier, candidate-gene studies and GWAS results of AD have implicated several immune-response genes that have the capability to alter the Th2 milieu. However, there is still considerable debate about whether initial skin barrier defects lead to secondary sensitization through the impaired skin or whether immune dysregulation is the first pathogenic mechanism.144 Thus, AD is considered as a disease of both immune dysfunction and skin barrier disruption.145

Atopic Dermatitis and Race

Various studies have examined the effect of racial group on the prevalence of AD. Studies of different racial groups in the same environment are particularly helpful to assess the contribution of genetic factors. Results from a 2003 U.S. national survey suggest that the prevalence of AD among individuals of African ancestry is 6 times higher than individuals of European ancestry,9,146148 affirming that AD places a high burden on the African American population.148 Similarly, a study in the United Kingdom showed that Black children with AD have approximately 6 times higher risk of having severe AD than do White children.146 In Australia, the 12-month cumulative incidence of AD is much higher in Chinese babies (44%) than in Caucasian babies living under similar environmental conditions (21%).149 Compared with infants born to White mothers, infants born to Black mothers have an adjusted odds ratio (OR) for risk of AD of 2.4 (95% confidence interval [CI]: 1.47 vs. 3.94).150 Though differences in socioeconomic status and environmental exposures between racial groups may contribute to prevalence differences, population genetic differences in skin barrier functions between the ancestral African, European, and Asian populations may be partly responsible for the loss of epithelial barrier functions in AD.

It is to be noted that the FLG mutations associated with AD in Europeans are different from those associated with AD in Chinese people, which again are different from those associated with AD in Japanese people. These findings suggest that different FLG mutations that essentially have the same effect are population specific and arose after the individual populations had separated from common ancestors. It has been speculated that the FLG mutations conferred survival advantage to these populations. In a recent report, Thyssen et al. hypothesized that FLG mutations are more prevalent in Northern Europeans, who receive relatively little amount of sunlight, in order to allow better penetration of ultraviolet-B rays through a defective skin barrier and facilitate the production of Vitamin D3 within the skin.151 Meta-analyses of studies involving thousands of European ancestry patients have confirmed FLG associations with an overall odds ratio for AD ranging from 3.12 to 4.78.152 Though FLG has been identified as a major locus causing skin barrier deficiency, not all patients with AD have mutations in this gene, so AD cannot be explained by FLG mutation alone. For example, though several common FLG mutations have been found to be associated with AD in Europeans and Asians in candidate-gene studies and GWAS, the search for common FLG mutations in native Africans has not been fruitful. The observation that filaggrin mutations play an important role in European American patients with AD but are absent in African patients, who also have reduced filaggrin protein levels in their skin lesions, suggests that both genetic and immune mechanisms can lead to skin barrier dysfunction.156 Despite a prevalence of 3.2% for the FLG mutations common in Europeans with AD having also been demonstrated in African Americans with AD, this finding could be explained by European admixture, which can be as high as 20% in the African American population.138,153,154 Interestingly, in a recent study, Margolis et al. have demonstrated the association of null mutations in FLG2, a gene belonging to the EDC and closely related to FLG, with persistent AD in African American subjects.155 These mutations were not present (or were infrequently present) in persons of European origin. This finding suggests that in African Americans, FLG2 could be more influential than FLG in predisposing individuals to AD. Studies exploring the role of FLG2 in patients with AD need to be conducted in native African populations to confirm this finding.

Genetics of Allergic Rhinitis

Similar to asthma and other allergic disorders, AR is a disease with a complex etiology. Typical AR-related allergens include tree, grass, and weed pollens; animal dander; dust mites; mold; and cockroach.157 The relationship between AR and asthma is very close, with AR often preceding the development of asthma. Although AR affects the upper airways and asthma the lower airways, the pathogenesis of both conditions involve airway epithelial cells, and the pattern of inflammatory response is very similar, giving rise to the concept of “one airway, one disease”.158 Preschool children and young adults with untreated AR are at threefold increased risk of asthma later in life.159,160 Evidence regarding the heritability of AR came from twin studies.161 Though MZ twins were 45–60% concordant, the concordance rate in DZ twins was not more than 25%, with the calculated heritability ranging between 33% and 91%.44,161 Several genome-wide linkage scans have been performed for the AR phenotype in European and Asian populations, in which loci in chromosomes 2,3, 4, and 9 were implicated.161

Candidate-gene studies performed in Asian and European populations have also implicated numerous genes, many of them belonging to the inflammatory/immune response pathways. These include chemokines/chemokine receptors (SDAD1, CXCL9, CXCL10, CXCL11, RANTES), interleukins/interleukin receptors (IL-13, IL-18, IL-23R, IL-12RB1, IL-12RB2, IL-27), and CD14, among others.161164 Three GWAS have been performed specifically for the AR phenotype. The first one by Andiappan et al. identified suggestive associations with two SNPs on the MRPL4 and BCAP genes in a Chinese cohort; both genes have putative roles in inflammatory/immune response pathways.165 However, none of these SNPs reached genome-wide significance. In the second GWAS meta-analysis of AR by Ramasamy et al., a SNP in a locus situated between chromosome 11 open reading frame 30 (C11orf30) and leucine-rich repeat containing 32 (LRRC32) attained genome-wide significance.166 Interestingly, this locus was previously identified in both asthma and AD/eczema GWAS.167,168 In a more recent study, Bunyavanich et al. integrated findings from GWAS, co-expression network, and expression SNP analysis in an attempt to test the biological context of GWAS findings and thus discover novel AR pathways.169 Using the GWAS approach, they were able to identify 4 loci in Latinos and 1 locus in a combined meta-analysis involving European Americans, Latinos, and African Americans/African Caribbeans that reached genome-wide significance, and the integrative approach allowed them to demonstrate the importance of mitochondrial pathways in AR.

Allergic Rhinitis and Race

AR occurs in persons of all races. In the U.S., Asian, but not Black, subjects are more likely to report hay fever than are White subjects.170 In addition, White children are more likely to have AR than are Black children, and white pregnant women are more likely to have AR than are black pregnant women.5 However, migrant studies suggest that environmental factors, rather than genetic ones, may be more important in explaining the apparent discrepancy in the prevalence of hay fever between races.171 In fact, studies that reported any difference in the prevalence of hay fever between races frequently acknowledged the possibility of bias due to unmeasured socio-economic factors, selective underreporting, or underdiagnoses as the driving factor behind the findings.5,170 The major problem is defining AR consistently, as there are no consensus clinical/phenotypic/endotypic definitions to accurately apply for AR studies.172,173

Genetics of Asthma

Although familial aggregation of asthma was demonstrated as early as the first half of the last century, twin studies performed in the later half showed a wide range in asthma heritability estimates, between 36% and 95%.174 Starting from the 1990s, more than 25 genetic linkage studies of asthma have been undertaken to date, and the combined approach of linkage and positional cloning has been able to identify 9 asthma susceptibility genes (ADAM33, DPP10, PHF11, NPSR1, HLA-G, CYFIP2, IRAK3, COL6A5, and OPN3/CHML).43 More than 100 loci for asthma and related phenotypes have also been identified through candidate-gene studies, with the majority of these genes being associated with the immune system (e.g., IL4RA, CD14, TLR2, and HLA class II genes).174

Close to 40 GWAS in asthma have also been performed to date,175 leading to the validation of several genes previously identified through positional cloning or candidate-gene studies (IL13, IL1RL1, DPP10, and HLA-DQ)176 and to the discovery of several novel loci, the most replicated of which is the 17q21 locus. In 2007, Moffatt et al. were the first to identify the 17q21 region as the site of an asthma susceptibility locus.177 The asthma-associated SNPs are part of a haplotype that extends across four genes—ORMDL3, GSDMB, ZPBP2, and IKZF3. Subsequent expression quantitative trait loci (eQTL) studies have shown that the haplotype has the ability to influence the expression of ORMDL3, GSDMB, and ZPBP2,178 providing strong, functional evidence in favor of these genes in asthma pathogenesis. Most of these variants are in non-genic regions, and additional studies are required to determine whether any of the variants are directly causal through effects on regulatory elements or are in LD with causal variants.179 Hence, it is still unclear which of these genes are responsible for the observed association with asthma and what role they play in asthma pathogenesis.

A major step in asthma gene discovery has been the formation of large consortia, which have brought together very large numbers of subjects in a meta-analysis. The Europe-based GABRIEL consortium comprised more than 26,000 subjects of European ancestry,180 whereas the U.S.-based EVE consortium181 included more than 24,000 subjects of White, African American, and Hispanic ancestries. The most strongly associated genes in both studies and across different ethnicities were IL1RL1/IL18R1, TSLP, and IL33.43 However, there are also inconsistent results between these studies, primarily due to clinical heterogeneity (different phenotype definitions), population stratification, and results without multiple-testing corrections. Over the last few years, there has been a strong focus on powering genetic studies with very large sample sizes; however, large cohorts might not help improve our understanding of the genetic underpinnings of atopic phenotypes as much as precise phenotyping.182 Thus, compiling large groups of patients with asthma without precise, careful phenotyping will not yield useful results, as demonstrated by the fact that asthma is a highly heterogeneous disease and the exact phenotype may be “lost in diagnosis”. Consequently, a shift has occurred from defining clinical asthma phenotypes to identifying mechanistically relevant endotypes.183185 For future studies, more efforts are needed to split these large resources into subgroups of patients sharing the same asthma phenotype. This strategy may impair statistical power due to decreased sample size but is likely to be enriched by patients sharing the same underlying molecular mechanisms. Thus, a small, homogeneous sample size is sufficient to detect enriched genetic effect in a more precisely selected group of patients.

Although the GWAS has identified several common risk variants for asthma, these variants are collectively responsible for only a small fraction of asthma heritability.186 One source of this “missing heritability” (discrepancy between the predicted and observed genetic risk) could be rare or structural variants that contribute to disease risk but were not covered in early GWAS (newer GWAS platforms have a greater representation of these variants). Another reason for missing heritability could be gene-gene and gene-environment interactions. Though interactions between candidate genes have been demonstrated in asthma (e.g., interaction between IL13 and IL4RA among African American187 and Dutch populations188) gene-gene interaction studies of asthma, or any complex disease, at a genome-wide level will require very large sample sizes and highly sophisticated statistical methods that are still being developed. Environmental factors, such as the gut microbiome, diet, or exposure to traffic, cigarette smoke, and mold, might play a role in triggering allergic disorders by modulating gene expression via epigenetic pathways.

There have been several studies that have investigated gene-environment interactions in asthma, mostly in the candidate-gene context. The interaction between CD14 and levels of endotoxin exposure is an important example.189 Ege et al. performed a genome-wide environment interaction study of asthma on a central European population for farm-related exposures. For example, the CD14(−260)C>T variant was associated with low total IgE in school children living in urban/suburban Tucson, Arizona, but the opposite association was reported in a farming community.190,191 Although this study failed to show interaction of common SNPs or SNPs previously reported to have interaction effects, rare SNPs in potentially relevant pathways were identified.192 Other factors that may contribute to poor replication of GWAS results include poorly matched control groups, LD, genetic heterogeneity between populations, differences in phenotype/endotype definitions, inadequate evaluation of environmental factors, chance due to multiple testing, and publication bias of positive results.193,194 The success of post-GWAS era studies in asthma will greatly depend on the ability to adequately quantify environmental exposures by standardizing sample collection, as well as generating more precise phenotype/endotype definitions. Novel genes of susceptibility to asthma were revealed only when relevant environmental exposures were considered. Therefore, it could be envisaged that detecting gene-environment interactions may help to target the preventive strategies for susceptible individuals. Finally, epigenetic mechanisms have been hypothesized to play an important role in the development and heritability of asthma and allergy.195 The advent of next-generation sequencing holds great promise for asthma genetics, including identifying rare variants with large effect sizes that could account for some of the missing heritability.

Asthma and Race

Significant racial differences exist in asthma prevalence rates in the U.S. The National Center for Health Statistics (NCHS) reported average annual prevalence rates of asthma to be 19% in Puerto Ricans, 14% in Blacks, 9% in American Indians, 8% in Whites, 5% in Mexican Americans, and 5% in Asian Americans for the period of 2008–2010.196 Although both Puerto Ricans and Mexican Americans are considered to be of Hispanic ethnicity, the former group has 12–15% Native American ancestry and 18–25% African ancestry, whereas the latter group has 35–64% Native American ancestry and only 3–5% African ancestry.197 When taken in conjunction with the fact that African Americans have a high prevalence rate for asthma, it would appear that African ancestry is an important risk factor for the development of asthma.

Studies have also revealed that African Americans a higher rate of allergic sensitization than do Whites.11 However, this association of African ancestry with asthma and allergic sensitization does not necessarily involve genetics factors since ancestry could also be a surrogate for unique environmental exposures. An analysis of NCHS data from the period of 2001–2010 by Akinbami et al.198 showed that asthma prevalence substantially increased in Blacks (1.2 million in 2001 and 1.7 million in 2010), whereas it remained essentially the same in Whites (4.4 million in 2001 and 4.5 million in 2010). Since a change in genetic sequence at the population level is highly improbable in this short time-span, it is likely that environmental exposures, either acting alone or in combination with genes, are responsible for this selective rise in asthma prevalence among African Americans.

There is a dearth of well-designed genetic studies of asthma in ethnic minorities. A genome-wide linkage scan and candidate-gene studies performed by The Collaborative Study on the Genetics of Asthma (CSGA) included African American and Hispanic families, but the regions with the highest candidate genes and linkage signals in African Americans did not overlap with the regions of highest linkage in Whites and Hispanics.199 Although the recent GWAS are robust compared to the many drawbacks of candidate-gene studies, the majority of the GWAS performed to date have focused on Whites, with relatively few including members of ethnic minority groups. Such studies have been limited by sample size, phenotypic and biologic heterogeneity, self-reported race, and inadequate data on environmental exposures and/or behavioral factors.

Results from the two largest meta-analyses of GWAS on asthma done to date, the GABRIEL and EVE consortia (discussed in the “Genetics of Asthma” section),180,181 indicated that some loci are ancestry specific, including ORMDL3/GSDML in White and Hispanic ancestry and PYHIN1 in African American ancestry.43 Recent, ethnic-specific associations of rare and low-frequency variants with asthma showed association of GRASP and GSDMB variants in the Latino ancestry and MTHFR variants in the African ancestry samples.200 Although GWAS have identified many candidate regions and genes, the underlying causes and genetic basis of asthma remains elusive. It is evident, however, that asthma is polygenetic and single-gene effects can account for only a small percent of the asthma phenotype.

Since GWAS chips typically provide good coverage for common variants and poor coverage for rare ones, a significant portion of the missing heritability in asthma could be due to rare and structural variants that are not included in a typical GWAS. It is also possible that rare and structural variants associated with asthma are more common in African Americans,34 both qualitatively and quantitatively, leading to a higher disease rate. Gene-environment interaction is another potential explanation for the higher disease prevalence in African Americans. It is possible that disease-causing genes are expressed differentially depending on environmental exposures, which could in turn depend on specific ethnic backgrounds (i.e., ethnicity-specific epigenetic factors). Indeed, in a genome-wide transcriptional profiling study, it was demonstrated that children with asthma who belonged to lower socioeconomic strata overexpressed genes regulating inflammatory pathways compared with their counterparts who came from higher socioeconomic backgrounds.201 Therefore, ethnicity can be a surrogate for environmental exposures that interact with the genome and lead to disease.

Patterns of ancestry differentiation and selection at loci associated with atopic disorders

Several population genetic studies have attempted to understand if population diferences of allergy and asthma risk alleles is present and identify the respective factors that may contribute to asthma disparity across racial ancestry. Most asthma risk alleles tend to show decreasing frequency along with human migration out of Africa, such that the African and East Asian populations carry the highest and lowest asthma genetic risk scores, respectively.202 This frequency variation across present-day populations was likely shaped by random genetic drift, migration history, and natural selection, particularly influenced by pathogens and differing environments.202206 While in random genetic drift allele frequencies change over time purely due to chance, in natural selection, a deleterious allele is selected in preference to the normal allele since it confers resistance to another disease with a high fatality rate. This confers the heterozygotes with one copy of the harmful allele a survival advantage over the homozygotes with two copies of the normal allele. Several genes related to atopic disorders are thought to have been subject to this balanced selection process; here we discuss a few that show the strongest evidence of evolutionary footprints. The observed differences in allele frequency and population differentiation for disease- associated SNPs may partly explain the disparities in AD prevalence observed in AAs.6

Malaria is known to have been a dominant selective force in shaping evolution in the African people. Interleukin (IL)-4 is an anti-inflammatory cytokine produced by activated T cells (Th2) that regulates humoral as well as adaptive immunity. IL-4 589(C>T) (rs2243250), a SNP located in the promoter region of the IL-4 gene, regulates its activity and has been reported to be associated with elevated levels of total serum IgE (a marker for atopy),207,208 and with asthma.209,210 The Fulani people of West Africa have fewer attacks of malaria and a lower prevalence of P. falciparum infection when compared to their geographical neighbors.211 They also have higher levels of anti-malarial IgG and IgE levels.212 Luoni et al.201 showed that the variant T allele of IL-4 589 (C>T) occurs at a much higher frequency in the Fulani when compared to the neighboring Mossi and Rimaibe people, and is also positively associated with anti-malarial antibodies in the Fulani population. In this case, positive selection of the variant T allele that is associated with allergy and asthma presumably gave the Fulani a survival advantage over malaria.211214

The variants Gln551Arg (rs1801275)215 and Ile50Val (rs1805010)216 of the IL-4 receptor (IL-4R) gene have been associated with atopy and asthma respectively, and the haplotype Ile50Arg551, which has been associated with “enhanced signaling” and increased IgE production, was more common in African-Americans than in European-Americans.217 IL-13, another important Th2 cytokine gene has multiple polymorphisms, including C-1112T ((rs1800925, also known as C-1111T), associated with allergy. The frequency of this allele was 12% in Caucasians, 13% in Chinese and 48% in Africans.218

It may be noted that the Th2 response—in which IL-4 plays a key role203—is likely to be advantageous in tropical regions, where the likelihood of helminthic infection is higher.219 This exaggerated Th2 response, which evolved for parasite expulsion early in the development of mankind, predisposes individuals to asthma and allergy in modern societies of developed countries, where exposure to parasites is rare. This could be one of the reasons why people of African ancestry in western countries have higher prevalence and greater severity of allergic symptoms than natives of these host countries, indicating that African ancestry might be a risk factor for allergic disease.219221 Indeed, several candidate genes that encode for Th2-related molecules, such as IL-4 and IL-13, show a higher frequency in people with African ancestry of alleles that have a strong Th2- promoting activity,219 suggesting that these alleles, which have been conserved to combat parasitic infections in Africa are leading to increased prevalence of atopic disorders in African origin people settled in western countries.

Two common and independent mutations in the FLG gene, namely R501Xand 2282del4, which have similar population frequencies, are present in about 10% of people of Northern European ancestry.151,222 These mutations lead to an absence of filaggrin protein in the skin, which causes a barrier defect allowing entry of external allergens and subsequent sensitization, believed to be the starting point of AD and the atopic march. As Irvine and McLean212 point out, such a high prevalence of two very similar mutations in the European population is unlikely to be a result of random genetic drift but could rather be a consequence of balanced selection. They speculate that heterozygote carriers of these mutations had a survival advantage due to greater immunity to infectious diseases such as tuberculosis, influenza and plague that occurred as pandemics throughout known history, due to “natural vaccination” resulting from low level exposure to microbial antigens through a disrupted skin barrier.222 Asian populations carry their own population specific FLG mutations at a prevalence rate of approximately 5%,151 and similar evolutionary advantage could well have been the cause. While FLG mutations have an extremely low prevalence in people of African origin, skin barrier defects in this population might be the result of mutations in a related gene, FLG2, as proposed by Margolis et al.155 Another hypothesis attributes the higher prevalence of FLG mutations in Northern Europeans when compared to Southern Europeans, Asians and Africans to better penetration of sunlight through a defective skin barrier in sunlight deprived northern latitudes, which helps in the synthesis of Vitamin D3.151 This hypothesis better explains the observed North-South gradient in prevalence of FLG mutations, with northern regions in Europe, Africa and Asia having a higher relative prevalence compared to the southern regions.

Mapping Atopic Disorder Genes Contributing to Racial Differences

Gene discovery started in the late 1970s with a functional approach, in which the gene product and its function were used to identify a gene.223 Since then, several analytic approaches, such as linkage analysis, candidate-gene, genome-wide association (GWA), and admixture mapping (AM) studies, have been used to map genes related to atopic disorders (Figure 8). One approach to localize genes in which risk alleles are distributed differentially between racial ancestry groups within an admixed population is using admixture mapping. Admixed individuals are those who inherit chromosomal segments of distinct continental ancestry, in which ancestral genomes have diverged over time due to genetic drift and/or natural selection.224 African Americans are an example of admixed populations, with ~80% African ancestry and ~20% European ancestry.225 Mapping atopic disorder susceptibility genes in an admixed population using AM involves screening the genome of individuals of mixed chromosomal regions that have a higher frequency of alleles from the parental population with higher disease risk.226 African ancestry individuals exhibit an increased prevalence of AD, asthma, and FA compared to European Americans and AM could be an idea approach to map variants associated with atopic disorders.227 AM takes advantage of the high level of admixture created in populations of mixed ancestry by searching for associations between the ancestry score of a marker and disease, as opposed to GWAS which is between the genotype score of the marker and disease.228 The theory behind AM is that chromosomal segments of affected individuals contain a significantly higher than average proportion of alleles from the high-risk parental population and thus are more likely to harbor a disease-gene allele(s).228 Criteria to evaluate the applicability of AM in atopic disorder-related gene mapping include:229 (1) the prevalence of the disease differences in ancestral populations from which the admixed population was formed; (2) a measurable difference in disease-causing alleles between the parental populations; (3) reduced linkage disequilibrium between unlinked loci across chromosomes and strong linkage disequilibrium between neighboring loci; (4) a set of markers with noticeable allele-frequency differences between parental populations that contributes to the admixed population. Among atopic disorders, asthma clinical variables are relatively well studied for ancestry-related variations. A lower responsiveness to bronchodilators has been observed among individuals of African descent with asthma compared to Whites with asthma.230,231 In African Americans, a decrease in lung function is associated with an increase in African ancestry proportion; adding genetically measured ancestry to the standard lung function prediction equations, rather than relying on self-identified race, reduced misclassification and resulted in reclassifying asthma severity by 5%.7 It is important to note that although ancestry is associated with asthma clinical phenotypes, socioeconomic status and related environmental exposure risk factors were not considered in most previous studies; thus, it is not clear whether ancestry is a surrogate for existing socio-environmental differences (i.e., may not be directly causal) or is an independent risk factor (serving as surrogate for genetic differences) for asthma risk. For example, if there is greater global ancestry noted across the genome in patients with asthma relative to controls, but no significant rise in local ancestry at a particular locus, this may point to a stronger role for environmental factors (e.g., exposures to traffic, home, cigarettes) independent of genetic ancestry.232234 Thus, a more careful assessment of the degree of ancestry, which is not captured by self-reported race/ethnicity alone, in larger cohorts while controlling for environmental exposure and other social determinants of health is needed to better understand the role of genetics, if any, in the observed racial differences in clinical asthma/allergy phenotypes. These insights are critical to develop intervention policies to reduce the burden of asthma due to non-genetic factors.235 Next-generation sequencing technologies now make it possible to genotype and measure hundreds of thousands of ancestry-specific markers across the genome, which have facilitated AM studies for discovery of asthma-associated genomic regions in admixed populations.228 AM has successfully been used to identify genomic regions harboring disease susceptibility loci for cardiovascular disease,236 multiple sclerosis,237 prostate cancer,238 serum IL-6 levels,239 and asthma in Mexican, Puerto Rican, and African American ancestry populations.86,240247 These results indicate that the admixed population provides an excellent opportunity to harness the power of LD represented by ancestral markers transmitted together, thereby making use of disease prevalence variation in the ancestral (founder) populations to map risk loci.

Figure 8. Atopic disorder variant discovery and follow-up strategies.

Figure 8

Methods ranging from genome-wide linkage, candidate gene, genome-wide association and admixture studies are presented. These gene mapping approaches yielded promising association results in the field of allergic diseases. However, association does not necessarily imply biological functionality, and follow-up studies are needed to translate initial findings into the biological insights that ultimately will advance prognostics, diagnostics and therapeutics.

Omics-based system biology approach to atopic disorders

Recent progress in high-throughput technology, including next-generation sequencing, has made it possible to quantitatively examine the genome, epigenome, transcriptome, proteome, metabolome and microbiome in different phenotypes, with the resultant generation of massive amounts of data, including in the allergy field.248 Dissection of atopic disorders using these -omics approaches has been attempted at least to some extent. For example, transcriptome analyses have identified genes differentially expressed between asthmatic and control subjects249, between lesional and nonlesional skin in AD,250 and between airway smooth muscles derived from patients with fatal asthma and healthy controls at baseline and after treatment with Vitamin D.251 A recent study of transcriptome-wide responses to glucocorticoid treatment provides an interesting example of the use of transcriptomics to investigate the causes of population differences in allergic disease.252 Proteomics has been used in several studies to identify biomarkers and peptide patterns associated with specific asthma endotypes.253 These include a proteomic study that measured cytokine levels in bronchoalveolar lavage to categorize asthma patients254 and a protein-protein interaction analysis of asthma to confirm the role of established asthma pathways and uncovered new candidate genes.255 More recent approaches include shotgun proteomics, which has been used to characterize protein patterns in induced sputum from asthmatic patients, including patients with exercise-induced asthma.256 While we are not aware of any studies exploring the use of proteomics to characterize the relationship between race and allergic disease, peptide profiles and protein signatures that are specifically associated with several different cancer forms in patients of African ancestry have been identified.257,258 Similarly, we can expect proteomics to play a future role in explaining racial disparities and identifying race-specific biomarkers associated with allergic disease. MicroRNAs (miRNAs) are small noncoding RNAs that act mainly as suppressors of gene expression at the posttranscriptional level,259261 which may act as key regulators of the development and activity of innate and adaptive immune systems and control inflammatory processes. Several miRNAs have recently been associated with allergic airway diseases.262265 The epigenome has been interrogated to reveal micro-RNAs specifically targeting HLA-G alleles in patients at risk for asthma266 and reducing the intensity of chronic skin inflammation in patients with AD.267 Environmental triggers including tobacco smoke268 and traffic-related pollutants have been shown to induce epigenetic and microRNA expression changes in patients with allergic disease.269271 The impact of race on the role of epigenetics in allergic disease is still largely unexplored. A recent study showed that DNA methylation profiles of European American, African American, and Chinese American populations can be partially traced back to genetic variation.272 A few studies have explored the relationship between smoking and DNA methylation in African-American cohorts. Dogan et al. performed genome-wide DNA methylation analysis in a cohort of African-American women, and found 910 loci that were significantly associated with smoking status.272 Because epigenetic marks are biologically related to both environmental exposure and genes, they may provide a new explanation of gene-environment interactions.273,274 A recent study showed that DNA methylation profiles of European American, African American, and Chinese American populations can be partially traced back to genetic variation.275 Genetic variants that influence the gene expression or methylation state include expression quantitative trait loci (eQTL) or methylation quantitative trait loci (meQTLs), are efficient approaches to identify functional SNPs regulating expression and methylation of allergy-associated genes.177,178,276278 Information obtained from metabolomic studies of asthma have proved useful in distinguishing asthmatic from non-asthmatic subjects as well as in differentiating between different asthma subphenotypes.279 Several studies have examined associations between changes in microbiota and the development of AD, AR, and asthma, but far less have evaluated the impact of the microbiome on these outcomes. The skin or gut is colonized by numerous species of bacteria, fungi, and viruses that together are known as the microbiome.280,281 It has been estimated that the human gut is populated with up to 100 trillion microbes.282 This microbiota roughly contain on the order of 150-fold more genes than are encoded in the human genome.283 The human microbiome (which might interact with human genome) is a source of genetic diversity, a modifier of disease and an essential component of immunity.284 These microbes might interact with human genome. The potential impact of the microbiome on human health including respiratory and allergic diseases was examined in children.285 Children living in farming environments had a significantly decreased frequency of AD, AR and asthma compared with children living in urban areas.286,287 This relationship was further explored in the GABRIELA and PARSIFAL cohorts, which confirmed previous observations that children living on farms had decreased rates of allergic disease compared with urban children.288,289 Changes in lifestyle such as clean environment have led to a decrease in the infectious burden (e.g., reduce H. pylori) and are associated with a rise in allergic diseases also referred as the hygiene hypothesis.290293 As we advance our understanding of the diversity of microbiomes across geographical regions, time, individuals, and tissues within individuals, we become better positioned to take advantage of this emerging technology to inform the practice of precision medicine in atopic disorders.

Application of the systems biology approach, which is the integration of high-throughput data for the comprehensive profiling of disease phenotypes, to allergic diseases will provide greater insight into disease pathology and pathogenesis, provide a better understanding of cell to cell signaling networks for more efficient treatment, and also help to better predict disease outcome.294 To avoid getting lost in the sea of “big data”, advanced computational and mathematical based-information theory models such as machine learning, dynamic Bayesian network models, and differential equation–based models are required to provide a deep insight and understanding of the complex and non-linear relationships within and between biological systems. The growing abundance of high volume genetic/molecular data in the field of allergy has prepared the ground for information theory to be used for a better understanding of the flow of biological information from the genotype to the eventual allergic phenotype through mathematical models of information exchange.295,296 For example, the machine learning approach uses computer algorithms to identify patterns from the systematically collected large molecular profile data, and along with clinical metadata, can assist personalized treatments for effective management of atopic disorders with similar molecular subtypes.297 Thus, research toward a systems-level understanding of allergic diseases will provide clinically valuable insight into atopic disorders pathogenesis.298

In critically evaluating existing literature, compared to other specialties, notably the field of cancer research, the application of systems biology to asthma and allergy has been limited, and there are even fewer reports describing the systems biology approach to characterize racial differences and elucidate their role in allergic disease. The increasing focus on endotyping of allergic diseases, however, can be expected to prompt efforts to harness the power of systems biology as an approach to improve endotypic stratification. Systems biology can also be expected to play an important role in the development of personalized medicine approaches that takes racial ancestry into account. It needs to be stressed, however, that any racial differences identified using proteomics, transciptomics, or epigenetics can be expected to be modified or confounded by environmental factors associated with ancestry, given the profound influence of the environment on epigenetics and gene regulation. For an in-depth coverage of the systems biology approach in asthma and other allergic conditions, we refer the reader to a few excellent review articles published in the recent past.279,294,297,299,300

Conclusion

Atopic disorders form a cluster of phenotypes that include AD, FA, AR, and asthma. The global prevalence of these disorders, which have strong genetic and environmental components, has increased steadily in recent decades. Increased awareness of atopic diseases, changes in environmental exposures, epigenetic/genetic factors, and possible immune imbalance could contribute to this substantial increase in prevalence. Indeed, environmental exposures such as smoking, air pollution, and stress have been shown to cause changes in epigenetic modifications of genes, as well as altered microRNA expression.301 Although racial disparities have been noted in the prevalence of atopic conditions, determining the relative contribution of ancestry-specific genetic risk factors from environmental factors has proved to be challenging because of the limited number of studies performed on minority populations.14 Shared and unique genes/pathways involved in the “atopic march” also need to be better defined. The delineation and deconstruction of shared and unique biologic and genetic pathways among atopic disorders and ancestry-specific gene-environment interactions can help resolve the clinical complexity and better inform the development of novel therapies.

Although the concept of the atopic march has led to the study of overlapping genetic susceptibility loci and pathways as possible causal links among atopic disorders, the co-existence and excess comorbidity of several atopic disorders in a single individual also make it harder to distinguish between shared and disorder-specific variants.302 As a result, many questions remain regarding the underlying mechanisms and causative models that attempt to connect atopic disorders and ancestry. As we move forward, multi-ethnic birth cohorts with deep phenotyping and longitudinal phenotypes are needed to investigate the course of atopic march progression (i.e., whether early life food sensitization leads to other allergic disease later in life) as well as to identify individuals (or ancestries) at high risk of these comorbidities. In this comprehensive review, we have provided up-to-date information on atopic disorders with particular emphasis on genetic components and racial variations. Future research should include deep phenotyping of diverse ancestral populations, better characterization of environmental determinants, and the application of new technologies utilizing “-omics” tools. These include next-generation sequencing, epigenetics, and eQTL approaches in appropriate tissues/cells (Figure 5) along with publicly available bioinformatics and ancestry tools (Table 3). For example, recent findings investigating eosinophilic esophagitis revealed that tissue specificity was important in the expression of genes harboring the eosinophilic esophagitis–associated variants.60 This provides a paradigm shift for focusing on tissue–specific predisposition that may apply to other disorders, which may help to explain gaps in our understanding of the “atopic march” theory.60,303 Systematic integration of “big data” coming from providers (e.g., EMR), from omics (e.g., genomic, proteomic, epigenomic, metabolomic), and from multi-ethnic patients and non-providers (e.g., smart phone, monitoring tools for environmental triggers) can thus provide valuable insights to resolve the clinical complexity and ancestry-specific (or shared) etiology of atopic disorders.

Table 3.

Web-based, publicly available and easily accessible genomic/epigenomic resources relevant to atopic disorders

Database/Tool URL address
Population databases
HapMap http://hapmap.ncbi.nlm.nih.gov/
1000 Genomes Project http://www.1000genomes.org
ENCODE project http://genome.ucsc.edu/ENCODE
NHGRI GWAS catalogue www.genome.gov/gwastudies
dbGaP http://www.ncbi.nlm.nih.gov/gap
Exome Variant Server http://evs.gs.washington.edu/EVS/
Genevar http://www.sanger.ac.uk/resources/software/genevar/
ClinVar http://www.ncbi.nlm.nih.gov/clinvar/
dbSNP http://www.ncbi.nlm.nih.gov/snp
dbVar http://www.ncbi.nlm.nih.gov/dbvar
OMIM http://www.omim.org
RefSeqGene http://www.ncbi.nlm.nih.gov/refseq/rsg
MitoMap http://www.mitomap.org/MITOMAP/HumanMitoSeq
SNPedia http://www.snpedia.com
IGV for iPad http://www.broadinstitute.org/igv/iPadLaunch
PhenX https://www.phenxtoolkit.org
Genome/Epigenome browsers
UCSC Genome Browser http://genome.ucsc.edu/
WashU Epigenome Browser http://epigenomegateway.wustl.edu/
The Ensembl Genome Browser http://www.ensembl.org/index.html
VEGA Genome Browser http://vega.sanger.ac.uk/index.html
Blood eQTL browser http://genenetwork.nl/bloodeqtlbrowser/
Functional genomics/Gene expression
HaploReg http://www.broadinstitute.org/haploreg
RegulomeDB http://regulome.stanford.edu
FunSeq2 http://funseq2.gersteinlab.org/
GWAS3D http://jjwanglab.org/gwas3d
Gene Ontology www.geneontology.org
GATK https://www.broadinstitute.org/gatk/
miRWalk http://zmf.umm.uni-heidelberg.de
Sherlock http://sherlock.ucsf.edu/submit.html
UniProt www.uniprot.org
GEO http://www.ncbi.nlm.nih.gov/geo
ArrayExpress http://www.ebi.ac.uk/microarray-as/ae/
Microbiome http://www.hmpdacc.org/
Metabolome http://www.hmdb.ca/
Human Gene Mutation Database http://www.hgmd.org
In silico functional predictive tools
PFAM http://pfam.sanger.ac.uk
PolyPhen-2 http://genetics.bwh.harvard.edu/pph2/
SIFT http://sift.jcvi.org
SIFT BLink http://sift.jcvi.org/www/SIFT_BLink_submit.html
SMART http://smart.embl.de
SNPeffect http://snpeffect.switchlab.org
Single-gene/variant browser
AncestrySNPminer www.cchmc.org/mershalab/AncestrySNPminer
SPSmart http://spsmart.cesga.es/hapmap.php?dataSet=hapmap
PupaSuite http://pupasuite.bioinfo.cipf.es
SNPper http://snpper.chip.org/bio/snpper-enter
GVS http://gvs.gs.washington.edu/GVS
SNAP https://www.broadinstitute.org/mpg/snap/ldsearchpw.php

Supplementary Material

1

Table E1: Genes ranked by level of association with each atopic disorder based on mining of 15.73 million PubMed abstracts using Literature Lab™ from Acumenta Biotech Software.

Table E2: IPA summary of associated networks, molecular and cellular functions, diseases and disorders and canonical pathways for the 16 genes shared among FA, AD, AR and asthma.

2

Acknowledgments

This work was supported by the National Institutes of Health grant K01 HL103165, U19 AI066738, U19 AI070235, R01 DK076893, R37 A1045898, and Diversity and Health Disparities Award of the Cincinnati Children’s Research Foundation. We thank Shawna Hottinger for editorial assistance.

Abbreviations

AD

atopic dermatitis

AM

admixture mapping

AIMs

ancestry informative markers

AR

allergic rhinitis

DZ

dizygotic

EDC

epidermal differentiation complex

EMR

Electronic medical record

ETS

environnemental tobacco smoke

FA

food allergy

FLG

Filaggrin

GABRIELA

Multidisciplinary study to identify the genetic and environmental causes of asthma in the european community

GALA

genetics of asthma in latin Americans

GWEI

genome-wide environmental interaction

GWAS

genome wide association study

HapMap

haplotype map

IgE

Immunoglobulin E

ISAAC

international study of asthma and allergies in children

LABAs

long-acting β2-agonists

LD

linkage disequilibrium

MAF

minor allele frequency

MZ

monozygotic

NCHS

national center for health statistics

PARSIFAL

Prevention of allergy – risk factors for sensitization related to farming and anthroposophic lifestyle

PUBMED

search engine for accessing the MEDLINE database of citations

SNPs

single nucleotide polymorphisms

tagSNPs

haplotype-tagging SNPs

Footnotes

Conflicts of Interest Statement

M.E.R. is a consultant for Genentech, Novartis, Receptos, and NKT Therapeutics and has an equity interest in NKT Therapeutics, Immune Pharmaceuticals, and Celsus Therapeutics, as well as royalty interest from Teva Pharmaceuticals and Cincinnati Children’s Medical Center–owned patents concerning eosinophilic esophagitis. The rest of the authors declare that they have no competing interests.

References

  • 1.Zheng T, Yu J, Oh MH, Zhu Z. The atopic march: progression from atopic dermatitis to allergic rhinitis and asthma. Allergy, asthma & immunology research. 2011 Apr;3(2):67–73. doi: 10.4168/aair.2011.3.2.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Spergel JM, Paller AS. Atopic dermatitis and the atopic march. The Journal of allergy and clinical immunology. 2003 Dec;112(6 Suppl):S118–127. doi: 10.1016/j.jaci.2003.09.033. [DOI] [PubMed] [Google Scholar]
  • 3.Spergel JM. Atopic march: link to upper airways. Current opinion in allergy and clinical immunology. 2005 Feb;5(1):17–21. doi: 10.1097/00130832-200502000-00005. [DOI] [PubMed] [Google Scholar]
  • 4.Akdis CA. Allergy and hypersensitivity: mechanisms of allergic disease. Current opinion in immunology. 2006 Dec;18(6):718–726. doi: 10.1016/j.coi.2006.09.016. [DOI] [PubMed] [Google Scholar]
  • 5.Litonjua AA, Celedon JC, Hausmann J, et al. Variation in total and specific IgE: effects of ethnicity and socioeconomic status. The Journal of allergy and clinical immunology. 2005 Apr;115(4):751–757. doi: 10.1016/j.jaci.2004.12.1138. [DOI] [PubMed] [Google Scholar]
  • 6.Mersha TB, Abebe T. Self-reported race/ethnicity in the age of genomic research: its potential impact on understanding health disparities. Human genomics. 2015 Jan 7;9(1):1. doi: 10.1186/s40246-014-0023-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kumar R, Seibold MA, Aldrich MC, et al. Genetic ancestry in lung-function predictions. The New England journal of medicine. 2010 Jul 22;363(4):321–330. doi: 10.1056/NEJMoa0907897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Moreno-Estrada A, Gignoux CR, Fernandez-Lopez JC, et al. Human genetics. The genetics of Mexico recapitulates Native American substructure and affects biomedical traits. Science. 2014 Jun 13;344(6189):1280–1285. doi: 10.1126/science.1251688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Shaw TE, Currie GP, Koudelka CW, Simpson EL. Eczema prevalence in the United States: data from the 2003 National Survey of Children’s Health. The Journal of investigative dermatology. 2011 Jan;131(1):67–73. doi: 10.1038/jid.2010.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Moorman JE, Akinbami LJ, Bailey CM, et al. National surveillance of asthma: United States, 2001–2010. Vital & health statistics Series 3, Analytical and epidemiological studies/[US Dept of Health and Human Services, Public Health Service, National Center for Health Statistics] 2012 Nov;(35):1–67. [PubMed] [Google Scholar]
  • 11.Wegienka G, Johnson CC, Zoratti E, Havstad S. Racial differences in allergic sensitization: recent findings and future directions. Current allergy and asthma reports. 2013 Jun;13(3):255–261. doi: 10.1007/s11882-013-0343-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Levin AM, Wang Y, Wells KE, et al. Nocturnal asthma and the importance of race/ethnicity and genetic ancestry. American journal of respiratory and critical care medicine. 2014 Aug 1;190(3):266–273. doi: 10.1164/rccm.201402-0204OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Forno E, Celedon JC. Asthma and ethnic minorities: socioeconomic status and beyond. Current opinion in allergy and clinical immunology. 2009 Apr;9(2):154–160. doi: 10.1097/aci.0b013e3283292207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Barnes KC. Genomewide association studies in allergy and the influence of ethnicity. Current opinion in allergy and clinical immunology. 2010 Oct;10(5):427–433. doi: 10.1097/ACI.0b013e32833de6ee. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Leong AB, Ramsey CD, Celedon JC. The challenge of asthma in minority populations. Clinical reviews in allergy & immunology. 2012 Aug;43(1–2):156–183. doi: 10.1007/s12016-011-8263-1. [DOI] [PubMed] [Google Scholar]
  • 16.Carlson CS, Matise TC, North KE, et al. Generalization and dilution of association results from European GWAS in populations of non-European ancestry: the PAGE study. PLoS biology. 2013 Sep;11(9):e1001661. doi: 10.1371/journal.pbio.1001661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nguyen EA, Burchard EG. Asthma Research for All of the United States. Pediatric allergy, immunology, and pulmonology. 2012 Sep;25(3):128–131. doi: 10.1089/ped.2012.0173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wegienka G, Havstad S, Joseph CL, Zoratti E, Ownby D, Johnson CC. Allergic sensitization frequency and wheezing differences in early life between black and white children. Allergy and asthma proceedings: the official journal of regional and state allergy societies. 2012 Nov-Dec;33(6):493–499. doi: 10.2500/aap.2012.33.3600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gamble C, Talbott E, Youk A, et al. Racial differences in biologic predictors of severe asthma: Data from the Severe Asthma Research Program. The Journal of allergy and clinical immunology. 2010 Dec;126(6):1149–1156 e1141. doi: 10.1016/j.jaci.2010.08.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lum S, Bountziouka V, Sonnappa S, et al. Lung function in children in relation to ethnicity, physique and socioeconomic factors. The European respiratory journal. 2015 Oct 22; doi: 10.1183/13993003.00415-2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Quanjer PH, Stanojevic S, Cole TJ, et al. Multi-ethnic reference values for spirometry for the 3–95-yr age range: the global lung function 2012 equations. The European respiratory journal. 2012 Dec;40(6):1324–1343. doi: 10.1183/09031936.00080312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kelso JM. Applicability of the global lung function spirometry equations in contemporary multiethnic children. Pediatrics. 2014 Nov;134(Suppl 3):S172. doi: 10.1542/peds.2014-1817OOO. [DOI] [PubMed] [Google Scholar]
  • 23.Bonner R, Lum S, Stocks J, Kirkby J, Wade A, Sonnappa S. Applicability of the global lung function spirometry equations in contemporary multiethnic children. American journal of respiratory and critical care medicine. 2013 Aug 15;188(4):515–516. doi: 10.1164/rccm.201212-2208LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bernstein IL, Li JT, Bernstein DI, et al. Allergy diagnostic testing: an updated practice parameter. Annals of allergy, asthma & immunology: official publication of the American College of Allergy, Asthma, & Immunology. 2008 Mar;100(3 Suppl 3):S1–148. doi: 10.1016/s1081-1206(10)60305-5. [DOI] [PubMed] [Google Scholar]
  • 25.Stocks J, Sonnappa S, Lum S. Lung function testing in children: importance of race and ethnic-specific reference equations. Expert review of respiratory medicine. 2014 Oct;8(5):527–531. doi: 10.1586/17476348.2014.927317. [DOI] [PubMed] [Google Scholar]
  • 26.Khianey R, Oppenheimer J. Controversies regarding long-acting beta2-agonists. Current opinion in allergy and clinical immunology. 2011 Aug;11(4):345–354. doi: 10.1097/ACI.0b013e328348a82e. [DOI] [PubMed] [Google Scholar]
  • 27.Elbahlawan L, Binaei S, Christensen ML, Zhang Q, Quasney MW, Dahmer MK. Beta2-adrenergic receptor polymorphisms in African American children with status asthmaticus. Pediatric critical care medicine: a journal of the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies. 2006 Jan;7(1):15–18. doi: 10.1097/01.pcc.0000194010.63115.a2. [DOI] [PubMed] [Google Scholar]
  • 28.Blake K, Cury JD, Hossain J, et al. Methacholine PC20 in African Americans and whites with asthma with homozygous genotypes at ADRB2 codon 16. Pulmonary pharmacology & therapeutics. 2013 Jun;26(3):342–347. doi: 10.1016/j.pupt.2013.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lemanske RF, Jr, Mauger DT, Sorkness CA, et al. Step-up therapy for children with uncontrolled asthma receiving inhaled corticosteroids. The New England journal of medicine. 2010 Mar 18;362(11):975–985. doi: 10.1056/NEJMoa1001278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wechsler ME, Castro M, Lehman E, et al. Impact of race on asthma treatment failures in the asthma clinical research network. American journal of respiratory and critical care medicine. 2011 Dec 1;184(11):1247–1253. doi: 10.1164/rccm.201103-0514OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Patterson N, Price AL, Reich D. Population structure and eigenanalysis. PLoS genetics. 2006 Dec;2(12):e190. doi: 10.1371/journal.pgen.0020190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Genomes Project C. Abecasis GR, Auton A, et al. An integrated map of genetic variation from 1,092 human genomes. Nature. 2012 Nov 1;491(7422):56–65. doi: 10.1038/nature11632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Marth GT, Yu F, Indap AR, et al. The functional spectrum of low-frequency coding variation. Genome biology. 2011;12(9):R84. doi: 10.1186/gb-2011-12-9-r84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tennessen JA, Bigham AW, O’Connor TD, et al. Evolution and functional impact of rare coding variation from deep sequencing of human exomes. Science. 2012 Jul 6;337(6090):64–69. doi: 10.1126/science.1219240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Auer PL, Lettre G. Rare variant association studies: considerations, challenges and opportunities. Genome medicine. 2015;7(1):16. doi: 10.1186/s13073-015-0138-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lee S, Abecasis GR, Boehnke M, Lin X. Rare-variant association analysis: study designs and statistical tests. American journal of human genetics. 2014 Jul 3;95(1):5–23. doi: 10.1016/j.ajhg.2014.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Baye TM, Butsch Kovacic M, Biagini Myers JM, et al. Differences in candidate gene association between European ancestry and African American asthmatic children. PloS one. 2011;6(2):e16522. doi: 10.1371/journal.pone.0016522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Li YR, Keating BJ. Trans-ethnic genome-wide association studies: advantages and challenges of mapping in diverse populations. Genome medicine. 2014;6(10):91. doi: 10.1186/s13073-014-0091-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Liu CT, Buchkovich ML, Winkler TW, et al. Multi-ethnic fine-mapping of 14 central adiposity loci. Human molecular genetics. 2014 Sep 1;23(17):4738–4744. doi: 10.1093/hmg/ddu183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Genomes Project C, Corresponding a, Steering c et al. A global reference for human genetic variation. Nature. 2015 Sep 30;526(7571):68–74. doi: 10.1038/nature15393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Schultz Larsen F. Atopic dermatitis: a genetic-epidemiologic study in a population-based twin sample. Journal of the American Academy of Dermatology. 1993 May;28(5 Pt 1):719–723. doi: 10.1016/0190-9622(93)70099-f. [DOI] [PubMed] [Google Scholar]
  • 42.Larsen FS, Holm NV, Henningsen K. Atopic dermatitis. A genetic-epidemiologic study in a population-based twin sample. Journal of the American Academy of Dermatology. 1986 Sep;15(3):487–494. [PubMed] [Google Scholar]
  • 43.Ober C, Yao TC. The genetics of asthma and allergic disease: a 21st century perspective. Immunological reviews. 2011 Jul;242(1):10–30. doi: 10.1111/j.1600-065X.2011.01029.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.van Beijsterveldt CE, Boomsma DI. Genetics of parentally reported asthma, eczema and rhinitis in 5-yr-old twins. The European respiratory journal. 2007 Mar;29(3):516–521. doi: 10.1183/09031936.00065706. [DOI] [PubMed] [Google Scholar]
  • 45.Bieber T. Atopic dermatitis 2.0: from the clinical phenotype to the molecular taxonomy and stratified medicine. Allergy. 2012 Dec;67(12):1475–1482. doi: 10.1111/all.12049. [DOI] [PubMed] [Google Scholar]
  • 46.Halapi E, Hakonarson H. Recent development in genomic and proteomic research for asthma. Current opinion in pulmonary medicine. 2004 Jan;10(1):22–30. doi: 10.1097/00063198-200401000-00005. [DOI] [PubMed] [Google Scholar]
  • 47.Hu Y, Hines LM, Weng H, et al. Analysis of genomic and proteomic data using advanced literature mining. Journal of proteome research. 2003 Jul-Aug;2(4):405–412. doi: 10.1021/pr0340227. [DOI] [PubMed] [Google Scholar]
  • 48.Bousquet J, Van Cauwenberge P, Khaltaev N, Aria Workshop G, World Health O Allergic rhinitis and its impact on asthma. The Journal of allergy and clinical immunology. 2001 Nov;108(5 Suppl):S147–334. doi: 10.1067/mai.2001.118891. [DOI] [PubMed] [Google Scholar]
  • 49.Minton K. Allergy and Asthma: What ‘drives’ IL-4 versus IL-13 signalling? Nature Reviews Immunology. 2008;8:166–167. [Google Scholar]
  • 50.Stanic B, van de Veen W, Wirz OF, et al. IL-10-overexpressing B cells regulate innate and adaptive immune responses. The Journal of allergy and clinical immunology. 2015 Mar;135(3):771–780 e778. doi: 10.1016/j.jaci.2014.07.041. [DOI] [PubMed] [Google Scholar]
  • 51.Uhm TG, Kim BS, Chung IY. Eosinophil development, regulation of eosinophil-specific genes, and role of eosinophils in the pathogenesis of asthma. Allergy, asthma & immunology research. 2012 Mar;4(2):68–79. doi: 10.4168/aair.2012.4.2.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Takatsu K, Nakajima H. IL-5 and eosinophilia. Current opinion in immunology. 2008 Jun;20(3):288–294. doi: 10.1016/j.coi.2008.04.001. [DOI] [PubMed] [Google Scholar]
  • 53.Kay AB. Allergy and allergic diseases. First of two parts. The New England journal of medicine. 2001 Jan 4;344(1):30–37. doi: 10.1056/NEJM200101043440106. [DOI] [PubMed] [Google Scholar]
  • 54.Coca A, Cooke R. On the Classification of the Phenomena of Hypersensitiveness. Journal of immunology. 1923;8:163–182. [Google Scholar]
  • 55.Kabesch M, Depner M, Dahmen I, et al. Polymorphisms in eosinophil pathway genes, asthma and atopy. Allergy. 2007 Apr;62(4):423–428. doi: 10.1111/j.1398-9995.2006.01300.x. [DOI] [PubMed] [Google Scholar]
  • 56.Willis-Owen SA, Morar N, Willis-Owen CA. Atopic dermatitis: insights from linkage overlap and disease co-morbidity. Expert reviews in molecular medicine. 2007;9(9):1–13. doi: 10.1017/S1462399407000282. [DOI] [PubMed] [Google Scholar]
  • 57.Febbo PG, Mulligan MG, Slonina DA, et al. Literature Lab: a method of automated literature interrogation to infer biology from microarray analysis. BMC genomics. 2007;8:461. doi: 10.1186/1471-2164-8-461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ono JG, Worgall TS, Worgall S. 17q21 locus and ORMDL3: an increased risk for childhood asthma. Pediatric research. 2014 Jan;75(1–2):165–170. doi: 10.1038/pr.2013.186. [DOI] [PubMed] [Google Scholar]
  • 59.Bussmann C, Weidinger S, Novak N. Genetics of atopic dermatitis. Journal der Deutschen Dermatologischen Gesellschaft = Journal of the German Society of Dermatology: JDDG. 2011 Sep;9(9):670–676. doi: 10.1111/j.1610-0387.2011.07656.x. [DOI] [PubMed] [Google Scholar]
  • 60.Kottyan LC, Davis BP, Sherrill JD, et al. Genome-wide association analysis of eosinophilic esophagitis provides insight into the tissue specificity of this allergic disease. Nature genetics. 2014 Aug;46(8):895–900. doi: 10.1038/ng.3033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wolfe D, Dudek S, Ritchie MD, Pendergrass SA. Visualizing genomic information across chromosomes with PhenoGram. BioData mining. 2013;6(1):18. doi: 10.1186/1756-0381-6-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.McLeish S, Turner SW. Gene-environment interactions in asthma. Archives of disease in childhood. 2007 Nov;92(11):1032–1035. doi: 10.1136/adc.2006.112185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Magnusson LL, Olesen AB, Wennborg H, Olsen J. Wheezing, asthma, hayfever, and atopic eczema in childhood following exposure to tobacco smoke in fetal life. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2005 Dec;35(12):1550–1556. doi: 10.1111/j.1365-2222.2005.02374.x. [DOI] [PubMed] [Google Scholar]
  • 64.Li YF, Gilliland FD, Berhane K, et al. Effects of in utero and environmental tobacco smoke exposure on lung function in boys and girls with and without asthma. American journal of respiratory and critical care medicine. 2000 Dec;162(6):2097–2104. doi: 10.1164/ajrccm.162.6.2004178. [DOI] [PubMed] [Google Scholar]
  • 65.Moshammer H, Hoek G, Luttmann-Gibson H, et al. Parental smoking and lung function in children: an international study. American journal of respiratory and critical care medicine. 2006 Jun 1;173(11):1255–1263. doi: 10.1164/rccm.200510-1552OC. [DOI] [PubMed] [Google Scholar]
  • 66.Raherison C, Penard-Morand C, Moreau D, et al. In utero and childhood exposure to parental tobacco smoke, and allergies in schoolchildren. Respiratory medicine. 2007 Jan;101(1):107–117. doi: 10.1016/j.rmed.2006.04.010. [DOI] [PubMed] [Google Scholar]
  • 67.Colilla S, Nicolae D, Pluzhnikov A, et al. Evidence for gene-environment interactions in a linkage study of asthma and smoking exposure. The Journal of allergy and clinical immunology. 2003 Apr;111(4):840–846. doi: 10.1067/mai.2003.170. [DOI] [PubMed] [Google Scholar]
  • 68.Choudhry S, Avila PC, Nazario S, et al. CD14 tobacco gene-environment interaction modifies asthma severity and immunoglobulin E levels in Latinos with asthma. American journal of respiratory and critical care medicine. 2005 Jul 15;172(2):173–182. doi: 10.1164/rccm.200409-1232OC. [DOI] [PubMed] [Google Scholar]
  • 69.Meyers DA, Postma DS, Stine OC, et al. Genome screen for asthma and bronchial hyperresponsiveness: interactions with passive smoke exposure. The Journal of allergy and clinical immunology. 2005 Jun;115(6):1169–1175. doi: 10.1016/j.jaci.2005.01.070. [DOI] [PubMed] [Google Scholar]
  • 70.Aschard H, Lutz S, Maus B, et al. Challenges and opportunities in genome-wide environmental interaction (GWEI) studies. Human genetics. 2012 Oct;131(10):1591–1613. doi: 10.1007/s00439-012-1192-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rava M, Smit LA, Nadif R. Gene-environment interactions in the study of asthma in the postgenomewide association studies era. Current opinion in allergy and clinical immunology. 2015 Feb;15(1):70–78. doi: 10.1097/ACI.0000000000000131. [DOI] [PubMed] [Google Scholar]
  • 72.Turner SW, Ayres JG, Macfarlane TV, et al. A methodology to establish a database to study gene environment interactions for childhood asthma. BMC medical research methodology. 2010;10:107. doi: 10.1186/1471-2288-10-107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jimenez E, Marin ML, Martin R, et al. Is meconium from healthy newborns actually sterile? Research in microbiology. 2008 Apr;159(3):187–193. doi: 10.1016/j.resmic.2007.12.007. [DOI] [PubMed] [Google Scholar]
  • 74.DiGiulio DB, Romero R, Amogan HP, et al. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PloS one. 2008;3(8):e3056. doi: 10.1371/journal.pone.0003056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Gosalbes MJ, Llop S, Valles Y, Moya A, Ballester F, Francino MP. Meconium microbiota types dominated by lactic acid or enteric bacteria are differentially associated with maternal eczema and respiratory problems in infants. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2013 Feb;43(2):198–211. doi: 10.1111/cea.12063. [DOI] [PubMed] [Google Scholar]
  • 76.Prince BT, Mandel MJ, Nadeau K, Singh AM. Gut Microbiome and the Development of Food Allergy and Allergic Disease. Pediatric clinics of North America. 2015 Dec;62(6):1479–1492. doi: 10.1016/j.pcl.2015.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Heederik D, von Mutius E. Does diversity of environmental microbial exposure matter for the occurrence of allergy and asthma? The Journal of allergy and clinical immunology. 2012 Jul;130(1):44–50. doi: 10.1016/j.jaci.2012.01.067. [DOI] [PubMed] [Google Scholar]
  • 78.Eder W, Klimecki W, Yu L, et al. Opposite effects of CD 14/-260 on serum IgE levels in children raised in different environments. The Journal of allergy and clinical immunology. 2005 Sep;116(3):601–607. doi: 10.1016/j.jaci.2005.05.003. [DOI] [PubMed] [Google Scholar]
  • 79.Simpson A, John SL, Jury F, et al. Endotoxin exposure, CD14, and allergic disease: an interaction between genes and the environment. American journal of respiratory and critical care medicine. 2006 Aug 15;174(4):386–392. doi: 10.1164/rccm.200509-1380OC. [DOI] [PubMed] [Google Scholar]
  • 80.Williams LK, McPhee RA, Ownby DR, et al. Gene-environment interactions with CD14 C-260T and their relationship to total serum IgE levels in adults. The Journal of allergy and clinical immunology. 2006 Oct;118(4):851–857. doi: 10.1016/j.jaci.2006.07.007. [DOI] [PubMed] [Google Scholar]
  • 81.Smit LA, Siroux V, Bouzigon E, et al. CD14 and toll-like receptor gene polymorphisms, country living, and asthma in adults. American journal of respiratory and critical care medicine. 2009 Mar 1;179(5):363–368. doi: 10.1164/rccm.200810-1533OC. [DOI] [PubMed] [Google Scholar]
  • 82.Biagini Myers JM, Wang N, LeMasters GK, et al. Genetic and environmental risk factors for childhood eczema development and allergic sensitization in the CCAAPS cohort. The Journal of investigative dermatology. 2010 Feb;130(2):430–437. doi: 10.1038/jid.2009.300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hussein YM, Shalaby SM, Zidan HE, Sabbah NA, Karam NA, Alzahrani SS. CD14 tobacco gene-environment interaction in atopic children. Cellular immunology. 2013 Sep-Oct;285(1–2):31–37. doi: 10.1016/j.cellimm.2013.08.001. [DOI] [PubMed] [Google Scholar]
  • 84.Kauffmann F, Demenais F. Gene-environment interactions in asthma and allergic diseases: challenges and perspectives. The Journal of allergy and clinical immunology. 2012 Dec;130(6):1229–1240. doi: 10.1016/j.jaci.2012.10.038. quiz 1241–1222. [DOI] [PubMed] [Google Scholar]
  • 85.Williams LK, Oliver J, Peterson EL, et al. Gene-environment interactions between CD14 C-260T and endotoxin exposure on Foxp3+ and Foxp3- CD4+ lymphocyte numbers and total serum IgE levels in early childhood. Annals of allergy, asthma & immunology: official publication of the American College of Allergy, Asthma, & Immunology. 2008 Feb;100(2):128–136. doi: 10.1016/S1081-1206(10)60421-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Salari K, Choudhry S, Tang H, et al. Genetic admixture and asthma-related phenotypes in Mexican American and Puerto Rican asthmatics. Genetic epidemiology. 2005 Jul;29(1):76–86. doi: 10.1002/gepi.20079. [DOI] [PubMed] [Google Scholar]
  • 87.Smith AM, Bernstein DI, LeMasters GK, et al. Environmental tobacco smoke and interleukin 4 polymorphism (C-589T) gene: environment interaction increases risk of wheezing in African-American infants. The Journal of pediatrics. 2008 May;152(5):709–715. doi: 10.1016/j.jpeds.2007.10.011. 715 e701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Duncan LE, Keller MC. A critical review of the first 10 years of candidate gene-by-environment interaction research in psychiatry. The American journal of psychiatry. 2011 Oct;168(10):1041–1049. doi: 10.1176/appi.ajp.2011.11020191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Bourgon R, Gentleman R, Huber W. Independent filtering increases detection power for high-throughput experiments. Proceedings of the National Academy of Sciences. 2010;107(21):9546–9551. doi: 10.1073/pnas.0914005107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Dai JY, Kooperberg C, Leblanc M, Prentice RL. Two-stage testing procedures with independent filtering for genome-wide gene-environment interaction. Biometrika. 2012 Dec;99(4):929–944. doi: 10.1093/biomet/ass044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Murcray CE, Lewinger JP, Conti DV, Thomas DC, Gauderman WJ. Sample size requirements to detect gene-environment interactions in genome-wide association studies. Genetic epidemiology. 2011 Apr;35(3):201–210. doi: 10.1002/gepi.20569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Murcray CE, Lewinger JP, Gauderman WJ. Gene-environment interaction in genome-wide association studies. American journal of epidemiology. 2009 Jan 15;169(2):219–226. doi: 10.1093/aje/kwn353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Gauderman WJ, Thomas DC, Murcray CE, Conti D, Li D, Lewinger JP. Efficient genome-wide association testing of gene-environment interaction in case-parent trios. American journal of epidemiology. 2010 Jul 1;172(1):116–122. doi: 10.1093/aje/kwq097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Burchard EG, Avila PC, Nazario S, et al. Lower bronchodilator responsiveness in Puerto Rican than in Mexican subjects with asthma. American journal of respiratory and critical care medicine. 2004 Feb 1;169(3):386–392. doi: 10.1164/rccm.200309-1293OC. [DOI] [PubMed] [Google Scholar]
  • 95.Thomas DC, Lewinger JP, Murcray CE, Gauderman WJ. Invited commentary: GE-Whiz! Ratcheting gene-environment studies up to the whole genome and the whole exposome. American journal of epidemiology. 2012 Feb 1;175(3):203–207. doi: 10.1093/aje/kwr365. discussion 208–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Puig C, Garcia-Algar O, Monleon T, et al. A longitudinal study of environmental tobacco smoke exposure in children: parental self reports versus age dependent biomarkers. BMC public health. 2008;8:47. doi: 10.1186/1471-2458-8-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Benowitz NL. Cotinine as a biomarker of environmental tobacco smoke exposure. Epidemiologic reviews. 1996;18(2):188–204. doi: 10.1093/oxfordjournals.epirev.a017925. [DOI] [PubMed] [Google Scholar]
  • 98.Fraga MF, Ballestar E, Paz MF, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proceedings of the National Academy of Sciences of the United States of America. 2005 Jul 26;102(30):10604–10609. doi: 10.1073/pnas.0500398102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Moore JH, Williams SM. Epistasis and its implications for personal genetics. American journal of human genetics. 2009 Sep;85(3):309–320. doi: 10.1016/j.ajhg.2009.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Biagini Myers JM, Martin LJ, Kovacic MB, et al. Epistasis between serine protease inhibitor Kazal-type 5 (SPINK5) and thymic stromal lymphopoietin (TSLP) genes contributes to childhood asthma. The Journal of allergy and clinical immunology. 2014 Oct;134(4):891–899 e893. doi: 10.1016/j.jaci.2014.03.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kijima A, Murota H, Takahashi A, et al. Prevalence and impact of past history of food allergy in atopic dermatitis. Allergology international: official journal of the Japanese Society of Allergology. 2013 Mar;62(1):105–112. doi: 10.2332/allergolint.12-OA-0468. [DOI] [PubMed] [Google Scholar]
  • 102.Alduraywish SA, Lodge CJ, Campbell B, et al. The march from early life food sensitisation to allergic disease: A systematic review and meta-analyses of birth cohort studies. Allergy. 2015 Oct 14; doi: 10.1111/all.12784. [DOI] [PubMed] [Google Scholar]
  • 103.Ostblom E, Lilja G, Pershagen G, van Hage M, Wickman M. Phenotypes of food hypersensitivity and development of allergic diseases during the first 8 years of life. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2008 Aug;38(8):1325–1332. doi: 10.1111/j.1365-2222.2008.03010.x. [DOI] [PubMed] [Google Scholar]
  • 104.Punekar YS, Sheikh A. Establishing the sequential progression of multiple allergic diagnoses in a UK birth cohort using the General Practice Research Database. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2009 Dec;39(12):1889–1895. doi: 10.1111/j.1365-2222.2009.03366.x. [DOI] [PubMed] [Google Scholar]
  • 105.Al-Hammadi S, Zoubeidi T, Al-Maskari F. Predictors of childhood food allergy: significance and implications. Asian Pacific journal of allergy and immunology/launched by the Allergy and Immunology Society of Thailand. 2011 Dec;29(4):313–317. [PubMed] [Google Scholar]
  • 106.Hill DJ, Hosking CS, de Benedictis FM, et al. Confirmation of the association between high levels of immunoglobulin E food sensitization and eczema in infancy: an international study. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2008 Jan;38(1):161–168. doi: 10.1111/j.1365-2222.2007.02861.x. [DOI] [PubMed] [Google Scholar]
  • 107.Tsai HJ, Kumar R, Pongracic J, et al. Familial aggregation of food allergy and sensitization to food allergens: a family-based study. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2009 Jan;39(1):101–109. doi: 10.1111/j.1365-2222.2008.03111.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Hourihane JO, Dean TP, Warner JO. Peanut allergy in relation to heredity, maternal diet, and other atopic diseases: results of a questionnaire survey, skin prick testing, and food challenges. BMJ. 1996 Aug 31;313(7056):518–521. doi: 10.1136/bmj.313.7056.518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Sicherer SH, Furlong TJ, Maes HH, Desnick RJ, Sampson HA, Gelb BD. Genetics of peanut allergy: a twin study. The Journal of allergy and clinical immunology. 2000 Jul;106(1 Pt 1):53–56. doi: 10.1067/mai.2000.108105. [DOI] [PubMed] [Google Scholar]
  • 110.Liu X, Zhang S, Tsai HJ, et al. Genetic and environmental contributions to allergen sensitization in a Chinese twin study. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2009 Jul;39(7):991–998. doi: 10.1111/j.1365-2222.2009.03228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hong X, Tsai HJ, Wang X. Genetics of food allergy. Current opinion in pediatrics. 2009 Dec;21(6):770–776. doi: 10.1097/MOP.0b013e32833252dc. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Tan TH, Ellis JA, Saffery R, Allen KJ. The role of genetics and environment in the rise of childhood food allergy. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2012 Jan;42(1):20–29. doi: 10.1111/j.1365-2222.2011.03823.x. [DOI] [PubMed] [Google Scholar]
  • 113.Chen TK, Lee JH, Yu HH, et al. Association between human IL-10 gene polymorphisms and serum IL-10 level in patients with food allergy. J Formos Med Assoc. 2012 Dec;111(12):686–692. doi: 10.1016/j.jfma.2011.11.027. [DOI] [PubMed] [Google Scholar]
  • 114.Brown P, Nair B, Mahajan SD, et al. Single nucleotide polymorphisms (SNPs) in key cytokines may modulate food allergy phenotypes. Eur Food Res Technol. 2012 Nov;235(5):971–980. doi: 10.1007/s00217-012-1827-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Brown SJ, Asai Y, Cordell HJ, et al. Loss-of-function variants in the filaggrin gene are a significant risk factor for peanut allergy. The Journal of allergy and clinical immunology. 2011 Mar;127(3):661–667. doi: 10.1016/j.jaci.2011.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Asai Y, Greenwood C, Hull PR, et al. Filaggrin gene mutation associations with peanut allergy persist despite variations in peanut allergy diagnostic criteria or asthma status. The Journal of allergy and clinical immunology. 2013 Jul;132(1):239–242. doi: 10.1016/j.jaci.2013.03.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Tordesillas L, Goswami R, Benede S, et al. Skin exposure promotes a Th2-dependent sensitization to peanut allergens. The Journal of clinical investigation. 2014 Nov;124(11):4965–4975. doi: 10.1172/JCI75660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Hong X, Hao K, Ladd-Acosta C, et al. Genome-wide association study identifies peanut allergy-specific loci and evidence of epigenetic mediation in US children. Nature communications. 2015;6:6304. doi: 10.1038/ncomms7304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sicherer SH, Sampson HA. Food allergy: Epidemiology, pathogenesis, diagnosis, and treatment. The Journal of allergy and clinical immunology. 2014 Feb;133(2):291–307. doi: 10.1016/j.jaci.2013.11.020. quiz 308. [DOI] [PubMed] [Google Scholar]
  • 120.Hong X, Wang X. Early life precursors, epigenetics, and the development of food allergy. Semin Immunopathol. 2012 Sep;34(5):655–669. doi: 10.1007/s00281-012-0323-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Jackson KD, Howie LD, Akinbami LJ. Trends in allergic conditions among children: United States, 1997–2011. NCHS Data Brief. 2013 May;(121):1–8. [PubMed] [Google Scholar]
  • 122.Prescott S, Allen KJ. Food allergy: riding the second wave of the allergy epidemic. Pediatric allergy and immunology: official publication of the European Society of Pediatric Allergy and Immunology. 2011 Mar;22(2):155–160. doi: 10.1111/j.1399-3038.2011.01145.x. [DOI] [PubMed] [Google Scholar]
  • 123.Martino D, Joo JE, Sexton-Oates A, et al. Epigenome-wide association study reveals longitudinally stable DNA methylation differences in CD4+ T cells from children with IgE-mediated food allergy. Epigenetics. 2014 Jul;9(7):998–1006. doi: 10.4161/epi.28945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Chafen JJ, Newberry SJ, Riedl MA, et al. Diagnosing and managing common food allergies: a systematic review. Jama. 2010 May 12;303(18):1848–1856. doi: 10.1001/jama.2010.582. [DOI] [PubMed] [Google Scholar]
  • 125.Kumar R, Tsai HJ, Hong X, et al. Race, ancestry, and development of food-allergen sensitization in early childhood. Pediatrics. 2011 Oct;128(4):e821–829. doi: 10.1542/peds.2011-0691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Gupta RS, Springston EE, Warrier MR, et al. The prevalence, severity, and distribution of childhood food allergy in the United States. Pediatrics. 2011 Jul;128(1):e9–17. doi: 10.1542/peds.2011-0204. [DOI] [PubMed] [Google Scholar]
  • 127.Taylor-Black S, Wang J. The prevalence and characteristics of food allergy in urban minority children. Annals of allergy, asthma & immunology: official publication of the American College of Allergy, Asthma, & Immunology. 2012 Dec;109(6):431–437. doi: 10.1016/j.anai.2012.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kim H, Havstad S, Ezell JM, Wegienka G, Zoratti EM, Johnson CC. African American race is a robust risk factor for food, but not aeroallergen sensitization. The Journal of allergy and clinical immunology. 2013;131(2):1A–4A. AB1–AB332. [Google Scholar]
  • 129.Keet CA, Savage JH, Seopaul S, Peng RD, Wood RA, Matsui EC. Temporal trends and racial/ethnic disparity in self-reported pediatric food allergy in the United States. Annals of allergy, asthma & immunology: official publication of the American College of Allergy, Asthma, & Immunology. 2014 Mar;112(3):222–229 e223. doi: 10.1016/j.anai.2013.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Taylor-Black SA, Mehta H, Weiderpass E, Boffetta P, Sicherer SH, Wang J. Prevalence of food allergy in New York City school children. Annals of allergy, asthma & immunology: official publication of the American College of Allergy, Asthma, & Immunology. 2014 Jun;112(6):554–556 e551. doi: 10.1016/j.anai.2014.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Greenhawt M, Weiss C, Conte ML, Doucet M, Engler A, Camargo CA., Jr Racial and ethnic disparity in food allergy in the United States: a systematic review. The journal of allergy and clinical immunology. In practice. 2013 Jul-Aug;1(4):378–386. doi: 10.1016/j.jaip.2013.04.009. [DOI] [PubMed] [Google Scholar]
  • 132.Koplin JJ, Peters RL, Ponsonby AL, et al. Increased risk of peanut allergy in infants of Asian-born parents compared to those of Australian-born parents. Allergy. 2014 Dec;69(12):1639–1647. doi: 10.1111/all.12487. [DOI] [PubMed] [Google Scholar]
  • 133.Du Toit G, Roberts G, Sayre PH, et al. Identifying infants at high risk of peanut allergy: the Learning Early About Peanut Allergy (LEAP) screening study. The Journal of allergy and clinical immunology. 2013 Jan;131(1):135–143. e131–112. doi: 10.1016/j.jaci.2012.09.015. [DOI] [PubMed] [Google Scholar]
  • 134.Carroll CL, Balkrishnan R, Feldman SR, Fleischer AB, Jr, Manuel JC. The burden of atopic dermatitis: impact on the patient, family, and society. Pediatric dermatology. 2005 May-Jun;22(3):192–199. doi: 10.1111/j.1525-1470.2005.22303.x. [DOI] [PubMed] [Google Scholar]
  • 135.Luoma R, Koivikko A, Viander M. Development of asthma, allergic rhinitis and atopic dermatitis by the age of five years. A prospective study of 543 newborns. Allergy. 1983 Jul;38(5):339–346. doi: 10.1111/j.1398-9995.1983.tb04128.x. [DOI] [PubMed] [Google Scholar]
  • 136.Barnes KC. An update on the genetics of atopic dermatitis: scratching the surface in 2009. The Journal of allergy and clinical immunology. 2010 Jan;125(1):16–29. e11–11. doi: 10.1016/j.jaci.2009.11.008. quiz 30–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Tamari M, Hirota T. Genome-wide association studies of atopic dermatitis. J Dermatol. 2014 Mar;41(3):213–220. doi: 10.1111/1346-8138.12321. [DOI] [PubMed] [Google Scholar]
  • 138.Palmer CN, Irvine AD, Terron-Kwiatkowski A, et al. Common loss-of-function variants of the epidermal barrier protein filaggrin are a major predisposing factor for atopic dermatitis. Nature genetics. 2006 Apr;38(4):441–446. doi: 10.1038/ng1767. [DOI] [PubMed] [Google Scholar]
  • 139.Brown SJ, McLean WH. One remarkable molecule: filaggrin. The Journal of investigative dermatology. 2012 Mar;132(3 Pt 2):751–762. doi: 10.1038/jid.2011.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Indra AK. Epidermal TSLP: a trigger factor for pathogenesis of atopic dermatitis. Expert review of proteomics. 2013 Aug;10(4):309–311. doi: 10.1586/14789450.2013.814881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Gao PS, Rafaels NM, Mu D, et al. Genetic variants in thymic stromal lymphopoietin are associated with atopic dermatitis and eczema herpeticum. The Journal of allergy and clinical immunology. 2010 Jun;125(6):1403–1407 e1404. doi: 10.1016/j.jaci.2010.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Margolis DJ, Kim B, Apter AJ, et al. Thymic stromal lymphopoietin variation, filaggrin loss of function, and the persistence of atopic dermatitis. JAMA Dermatol. 2014 Mar;150(3):254–259. doi: 10.1001/jamadermatol.2013.7954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Liang Y, Chang C, Lu Q. The Genetics and Epigenetics of Atopic Dermatitis-Filaggrin and Other Polymorphisms. Clinical reviews in allergy & immunology. 2015 Sep 18; doi: 10.1007/s12016-015-8508-5. [DOI] [PubMed] [Google Scholar]
  • 144.Wolf R, Wolf D. Abnormal epidermal barrier in the pathogenesis of atopic dermatitis. Clinics in dermatology. 2012 May-Jun;30(3):329–334. doi: 10.1016/j.clindermatol.2011.08.023. [DOI] [PubMed] [Google Scholar]
  • 145.Darlenski R, Kazandjieva J, Hristakieva E, Fluhr JW. Atopic dermatitis as a systemic disease. Clinics in dermatology. 2014 May-Jun;32(3):409–413. doi: 10.1016/j.clindermatol.2013.11.007. [DOI] [PubMed] [Google Scholar]
  • 146.Ben-Gashir MA, Hay RJ. Reliance on erythema scores may mask severe atopic dermatitis in black children compared with their white counterparts. The British journal of dermatology. 2002 Nov;147(5):920–925. doi: 10.1046/j.1365-2133.2002.04965.x. [DOI] [PubMed] [Google Scholar]
  • 147.Hanifin JM, Reed ML, Eczema P, Impact Working G. A population-based survey of eczema prevalence in the United States. Dermatitis. 2007 Jun;18(2):82–91. doi: 10.2310/6620.2007.06034. [DOI] [PubMed] [Google Scholar]
  • 148.Vachiramon V, Tey HL, Thompson AE, Yosipovitch G. Atopic dermatitis in African American children: addressing unmet needs of a common disease. Pediatric dermatology. 2012 Jul-Aug;29(4):395–402. doi: 10.1111/j.1525-1470.2012.01740.x. [DOI] [PubMed] [Google Scholar]
  • 149.Mar A, Tam M, Jolley D, Marks R. The cumulative incidence of atopic dermatitis in the first 12 months among Chinese, Vietnamese, and Caucasian infants born in Melbourne, Australia. Journal of the American Academy of Dermatology. 1999 Apr;40(4):597–602. doi: 10.1016/s0190-9622(99)70443-3. [DOI] [PubMed] [Google Scholar]
  • 150.Moore MM, Rifas-Shiman SL, Rich-Edwards JW, et al. Perinatal predictors of atopic dermatitis occurring in the first six months of life. Pediatrics. 2004 Mar;113(3 Pt 1):468–474. doi: 10.1542/peds.113.3.468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Thyssen JP, Bikle DD, Elias PM. Evidence That Loss-of-Function Gene Mutations Evolved in Northern Europeans to Favor Intracutaneous Vitamin D3 Production. Evolutionary biology. 2014 Sep 1;41(3):388–396. doi: 10.1007/s11692-014-9282-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Rodriguez E, Baurecht H, Herberich E, et al. Meta-analysis of filaggrin polymorphisms in eczema and asthma: robust risk factors in atopic disease. The Journal of allergy and clinical immunology. 2009 Jun;123(6):1361–1370 e1367. doi: 10.1016/j.jaci.2009.03.036. [DOI] [PubMed] [Google Scholar]
  • 153.Winge MC, Bilcha KD, Lieden A, et al. Novel filaggrin mutation but no other loss-of-function variants found in Ethiopian patients with atopic dermatitis. The British journal of dermatology. 2011 Nov;165(5):1074–1080. doi: 10.1111/j.1365-2133.2011.10475.x. [DOI] [PubMed] [Google Scholar]
  • 154.Gao PS, Rafaels NM, Hand T, et al. Filaggrin mutations that confer risk of atopic dermatitis confer greater risk for eczema herpeticum. The Journal of allergy and clinical immunology. 2009 Sep;124(3):507–513. 513 e501–507. doi: 10.1016/j.jaci.2009.07.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Margolis DJ, Gupta J, Apter AJ, et al. Filaggrin-2 variation is associated with more persistent atopic dermatitis in African American subjects. The Journal of allergy and clinical immunology. 2014 Mar;133(3):784–789. doi: 10.1016/j.jaci.2013.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Thawer-Esmail F, Jakasa I, Todd G, et al. South African amaXhosa patients with atopic dermatitis have decreased levels of filaggrin breakdown products but no loss-of-function mutations in filaggrin. The Journal of allergy and clinical immunology. 2014 Jan;133(1):280–282. e281–282. doi: 10.1016/j.jaci.2013.09.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Bjorksten B, Clayton T, Ellwood P, Stewart A, Strachan D, Group IPIS Worldwide time trends for symptoms of rhinitis and conjunctivitis: Phase III of the International Study of Asthma and Allergies in Childhood. Pediatric allergy and immunology: official publication of the European Society of Pediatric Allergy and Immunology. 2008 Mar;19(2):110–124. doi: 10.1111/j.1399-3038.2007.00601.x. [DOI] [PubMed] [Google Scholar]
  • 158.Grossman J. One airway, one disease. Chest. 1997 Feb;111(2 Suppl):11S–16S. doi: 10.1378/chest.111.2_supplement.11s. [DOI] [PubMed] [Google Scholar]
  • 159.Rochat MK, Illi S, Ege MJ, et al. Allergic rhinitis as a predictor for wheezing onset in school-aged children. The Journal of allergy and clinical immunology. 2010 Dec;126(6):1170–1175 e1172. doi: 10.1016/j.jaci.2010.09.008. [DOI] [PubMed] [Google Scholar]
  • 160.Scadding GK. Optimal management of allergic rhinitis. Archives of disease in childhood. 2015 Apr 2; doi: 10.1136/archdischild-2014-306300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Davila I, Mullol J, Ferrer M, et al. Genetic aspects of allergic rhinitis. Journal of investigational allergology & clinical immunology. 2009;19(Suppl 1):25–31. [PubMed] [Google Scholar]
  • 162.Hu D, Hu G, Zhu J, Shen Y, Kang H, Hong S. Association between polymorphisms of the IL-23R gene and allergic rhinitis in a Chinese Han population. PloS one. 2013;8(5):e63858. doi: 10.1371/journal.pone.0063858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Shen Y, Yuan XD, Hu D, et al. Association between interleukin-27 gene polymorphisms and susceptibility to allergic rhinitis. Human immunology. 2014 Sep;75(9):991–995. doi: 10.1016/j.humimm.2014.07.004. [DOI] [PubMed] [Google Scholar]
  • 164.Wei P, Kou W, Sun R, et al. Association study between interleukin-12 receptor beta1/beta2 genes and allergic rhinitis in the Chinese Han population. European archives of oto-rhino-laryngology: official journal of the European Federation of Oto-Rhino-Laryngological Societies. 2015 Apr;272(4):889–893. doi: 10.1007/s00405-014-3145-9. [DOI] [PubMed] [Google Scholar]
  • 165.Andiappan AK, Wang de Y, Anantharaman R, et al. Genome-wide association study for atopy and allergic rhinitis in a Singapore Chinese population. PloS one. 2011;6(5):e19719. doi: 10.1371/journal.pone.0019719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ramasamy A, Curjuric I, Coin LJ, et al. A genome-wide meta-analysis of genetic variants associated with allergic rhinitis and grass sensitization and their interaction with birth order. The Journal of allergy and clinical immunology. 2011 Nov;128(5):996–1005. doi: 10.1016/j.jaci.2011.08.030. [DOI] [PubMed] [Google Scholar]
  • 167.Ferreira MA, Matheson MC, Tang CS, et al. Genome-wide association analysis identifies 11 risk variants associated with the asthma with hay fever phenotype. The Journal of allergy and clinical immunology. 2014 Jun;133(6):1564–1571. doi: 10.1016/j.jaci.2013.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ferreira MA, Matheson MC, Duffy DL, et al. Identification of IL6R and chromosome 11q13.5 as risk loci for asthma. Lancet. 2011 Sep 10;378(9795):1006–1014. doi: 10.1016/S0140-6736(11)60874-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Bunyavanich S, Schadt EE, Himes BE, et al. Integrated genome-wide association, coexpression network, and expression single nucleotide polymorphism analysis identifies novel pathway in allergic rhinitis. BMC medical genomics. 2014;7:48. doi: 10.1186/1755-8794-7-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Chen JT, Krieger N, Van Den Eeden SK, Quesenberry CP. Different slopes for different folks: socioeconomic and racial/ethnic disparities in asthma and hay fever among 173,859 U.S. men and women. Environmental health perspectives. 2002 Apr;110(Suppl 2):211–216. doi: 10.1289/ehp.02110s2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Turkeltaub PC, Gergen PJ. Prevalence of upper and lower respiratory conditions in the US population by social and environmental factors: data from the second National Health and Nutrition Examination Survey, 1976 to 1980 (NHANES II) Annals of allergy. 1991 Aug;67(2 Pt 1):147–154. [PubMed] [Google Scholar]
  • 172.Bernstein JA, Levin LS, Al-Shuik E, Martin VT. Clinical characteristics of chronic rhinitis patients with high vs low irritant trigger burdens. Annals of allergy, asthma & immunology: official publication of the American College of Allergy, Asthma, & Immunology. 2012 Sep;109(3):173–178. doi: 10.1016/j.anai.2012.06.013. [DOI] [PubMed] [Google Scholar]
  • 173.Bernstein JA. Characterizing rhinitis subtypes. American journal of rhinology & allergy. 2013 Nov-Dec;27(6):457–460. doi: 10.2500/ajra.2013.27.3983. [DOI] [PubMed] [Google Scholar]
  • 174.Mathias RA. Introduction to genetics and genomics in asthma: genetics of asthma. Adv Exp Med Biol. 2014;795:125–155. doi: 10.1007/978-1-4614-8603-9_9. [DOI] [PubMed] [Google Scholar]
  • 175.Ortiz RA, Barnes KC. Genetics of Allergic Diseases. Immunol Allergy Clin North Am. 2015 Feb;35(1):19–44. doi: 10.1016/j.iac.2014.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Barnes KC. Genetic studies of the etiology of asthma. Proceedings of the American Thoracic Society. 2011 May;8(2):143–148. doi: 10.1513/pats.201103-030MS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Moffatt MF, Kabesch M, Liang L, et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature. 2007 Jul 26;448(7152):470–473. doi: 10.1038/nature06014. [DOI] [PubMed] [Google Scholar]
  • 178.Verlaan DJ, Berlivet S, Hunninghake GM, et al. Allele-specific chromatin remodeling in the ZPBP2/GSDMB/ORMDL3 locus associated with the risk of asthma and autoimmune disease. American journal of human genetics. 2009 Sep;85(3):377–393. doi: 10.1016/j.ajhg.2009.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Kruglyak L. Prospects for whole-genome linkage disequilibrium mapping of common disease genes. Nature genetics. 1999 Jun;22(2):139–144. doi: 10.1038/9642. [DOI] [PubMed] [Google Scholar]
  • 180.Moffatt MF, Gut IG, Demenais F, et al. A large-scale, consortium-based genomewide association study of asthma. The New England journal of medicine. 2010 Sep 23;363(13):1211–1221. doi: 10.1056/NEJMoa0906312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Torgerson DG, Ampleford EJ, Chiu GY, et al. Meta-analysis of genome-wide association studies of asthma in ethnically diverse North American populations. Nature genetics. 2011 Sep;43(9):887–892. doi: 10.1038/ng.888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Baye TM, Martin LJ, Khurana Hershey GK. Application of genetic/genomic approaches to allergic disorders. The Journal of allergy and clinical immunology. 2010 Sep;126(3):425–436. doi: 10.1016/j.jaci.2010.05.025. quiz 437–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Agache I, Akdis C, Jutel M, Virchow JC. Untangling asthma phenotypes and endotypes. Allergy. 2012 Jul;67(7):835–846. doi: 10.1111/j.1398-9995.2012.02832.x. [DOI] [PubMed] [Google Scholar]
  • 184.Chung KF, Adcock IM. Clinical phenotypes of asthma should link up with disease mechanisms. Current opinion in allergy and clinical immunology. 2015 Feb;15(1):56–62. doi: 10.1097/ACI.0000000000000134. [DOI] [PubMed] [Google Scholar]
  • 185.Chung KF, Adcock IM. How variability in clinical phenotypes should guide research into disease mechanisms in asthma. Annals of the American Thoracic Society. 2013 Dec;10(Suppl):S109–117. doi: 10.1513/AnnalsATS.201304-087AW. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Sadee W, Hartmann K, Seweryn M, Pietrzak M, Handelman SK, Rempala GA. Missing heritability of common diseases and treatments outside the protein-coding exome. Human genetics. 2014 Oct;133(10):1199–1215. doi: 10.1007/s00439-014-1476-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Battle NC, Choudhry S, Tsai HJ, et al. Ethnicity-specific gene-gene interaction between IL-13 and IL-4Ralpha among African Americans with asthma. American journal of respiratory and critical care medicine. 2007 May 1;175(9):881–887. doi: 10.1164/rccm.200607-992OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Howard TD, Koppelman GH, Xu J, et al. Gene-gene interaction in asthma: IL4RA and IL13 in a Dutch population with asthma. American journal of human genetics. 2002 Jan;70(1):230–236. doi: 10.1086/338242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Martinez FD. CD14, endotoxin, and asthma risk: actions and interactions. Proceedings of the American Thoracic Society. 2007 Jul;4(3):221–225. doi: 10.1513/pats.200702-035AW. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Baldini M, Lohman IC, Halonen M, Erickson RP, Holt PG, Martinez FD. A Polymorphism* in the 5′ flanking region of the CD14 gene is associated with circulating soluble CD14 levels and with total serum immunoglobulin E. American journal of respiratory cell and molecular biology. 1999 May;20(5):976–983. doi: 10.1165/ajrcmb.20.5.3494. [DOI] [PubMed] [Google Scholar]
  • 191.Ober C, Tselenko A, Cox N. Searching for asthma and atopy genes in the Hutterites: genome-wide studies using linkage and association. American journal of respiratory and critical care medicine. 2000;161(3):A600. [Google Scholar]
  • 192.Ege MJ, Strachan DP, Cookson WO, et al. Gene-environment interaction for childhood asthma and exposure to farming in Central Europe. The Journal of allergy and clinical immunology. 2011 Jan;127(1):138–144. 144 e131–134. doi: 10.1016/j.jaci.2010.09.041. [DOI] [PubMed] [Google Scholar]
  • 193.Ioannidis JP, Ntzani EE, Trikalinos TA, Contopoulos-Ioannidis DG. Replication validity of genetic association studies. Nature genetics. 2001 Nov;29(3):306–309. doi: 10.1038/ng749. [DOI] [PubMed] [Google Scholar]
  • 194.Colhoun HM, McKeigue PM, Davey Smith G. Problems of reporting genetic associations with complex outcomes. Lancet. 2003 Mar 8;361(9360):865–872. doi: 10.1016/s0140-6736(03)12715-8. [DOI] [PubMed] [Google Scholar]
  • 195.Kabesch M. Epigenetics in asthma and allergy. Current opinion in allergy and clinical immunology. 2014 Feb;14(1):62–68. doi: 10.1097/ACI.0000000000000025. [DOI] [PubMed] [Google Scholar]
  • 196.Akinbami LJ, Moorman JE, Bailey C, et al. Trends in asthma prevalence, health care use, and mortality in the United States, 2001–2010. NCHS Data Brief. 2012 May;(94):1–8. [PubMed] [Google Scholar]
  • 197.Rosser FJ, Forno E, Cooper PJ, Celedon JC. Asthma in Hispanics. An 8-year update. American journal of respiratory and critical care medicine. 2014 Jun 1;189(11):1316–1327. doi: 10.1164/rccm.201401-0186PP. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Akinbami LJ, Moorman JE, Simon AE, Schoendorf KC. Trends in racial disparities for asthma outcomes among children 0 to 17 years, 2001–2010. The Journal of allergy and clinical immunology. 2014 Sep;134(3):547–553 e545. doi: 10.1016/j.jaci.2014.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Scirica CV, Celedon JC. Genetics of asthma: potential implications for reducing asthma disparities. Chest. 2007 Nov;132(5 Suppl):770S–781S. doi: 10.1378/chest.07-1905. [DOI] [PubMed] [Google Scholar]
  • 200.Igartua C, Myers RA, Mathias RA, et al. Ethnic-specific associations of rare and low-frequency DNA sequence variants with asthma. Nature communications. 2015;6:5965. doi: 10.1038/ncomms6965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Chen E, Miller GE, Walker HA, Arevalo JM, Sung CY, Cole SW. Genome-wide transcriptional profiling linked to social class in asthma. Thorax. 2009 Jan;64(1):38–43. doi: 10.1136/thx.2007.095091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Kovacic MB, Myers JM, Wang N, et al. Identification of KIF3A as a novel candidate gene for childhood asthma using RNA expression and population allelic frequencies differences. PloS one. 2011;6(8):e23714. doi: 10.1371/journal.pone.0023714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Fumagalli M, Pozzoli U, Cagliani R, et al. Parasites represent a major selective force for interleukin genes and shape the genetic predisposition to autoimmune conditions. The Journal of experimental medicine. 2009 Jun 8;206(6):1395–1408. doi: 10.1084/jem.20082779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Casto AM, Feldman MW. Genome-wide association study SNPs in the human genome diversity project populations: does selection affect unlinked SNPs with shared trait associations? PLoS genetics. 2011;7(1):e1001266. doi: 10.1371/journal.pgen.1001266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Corona E, Chen R, Sikora M, et al. Analysis of the genetic basis of disease in the context of worldwide human relationships and migration. PLoS genetics. 2013 May;9(5):e1003447. doi: 10.1371/journal.pgen.1003447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Amato R, Pinelli M, Monticelli A, Marino D, Miele G, Cocozza S. Genome-wide scan for signatures of human population differentiation and their relationship with natural selection, functional pathways and diseases. PloS one. 2009;4(11):e7927. doi: 10.1371/journal.pone.0007927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Rosenwasser LJ, Klemm DJ, Dresback JK, et al. Promoter polymorphisms in the chromosome 5 gene cluster in asthma and atopy. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 1995 Nov;25(Suppl 2):74–78. doi: 10.1111/j.1365-2222.1995.tb00428.x. discussion 95–76. [DOI] [PubMed] [Google Scholar]
  • 208.Basehore MJ, Howard TD, Lange LA, et al. A comprehensive evaluation of IL4 variants in ethnically diverse populations: association of total serum IgE levels and asthma in white subjects. The Journal of allergy and clinical immunology. 2004 Jul;114(1):80–87. doi: 10.1016/j.jaci.2004.05.035. [DOI] [PubMed] [Google Scholar]
  • 209.Li Y, Guo B, Zhang L, Han J, Wu B, Xiong H. Association between C-589T polymorphisms of interleukin-4 gene promoter and asthma: a meta-analysis. Respiratory medicine. 2008 Jul;102(7):984–992. doi: 10.1016/j.rmed.2008.02.008. [DOI] [PubMed] [Google Scholar]
  • 210.Tang L, Lin HG, Chen BF. Association of IL-4 promoter polymorphisms with asthma: a meta-analysis. Genetics and molecular research: GMR. 2014;13(1):1383–1394. doi: 10.4238/2014.February.28.11. [DOI] [PubMed] [Google Scholar]
  • 211.Luoni G, Verra F, Arca B, et al. Antimalarial antibody levels and IL4 polymorphism in the Fulani of West Africa. Genes and immunity. 2001 Nov;2(7):411–414. doi: 10.1038/sj.gene.6363797. [DOI] [PubMed] [Google Scholar]
  • 212.Vafa M, Maiga B, Berzins K, et al. Associations between the IL-4 -590 T allele and Plasmodium falciparum infection prevalence in asymptomatic Fulani of Mali. Microbes and infection/Institut Pasteur. 2007 Jul;9(9):1043–1048. doi: 10.1016/j.micinf.2007.04.011. [DOI] [PubMed] [Google Scholar]
  • 213.Burchard EG, Silverman EK, Rosenwasser LJ, et al. Association between a sequence variant in the IL-4 gene promoter and FEV(1) in asthma. American journal of respiratory and critical care medicine. 1999 Sep;160(3):919–922. doi: 10.1164/ajrccm.160.3.9812024. [DOI] [PubMed] [Google Scholar]
  • 214.Rockman MV, Hahn MW, Soranzo N, Goldstein DB, Wray GA. Positive selection on a human-specific transcription factor binding site regulating IL4 expression. Current biology: CB. 2003 Dec 2;13(23):2118–2123. doi: 10.1016/j.cub.2003.11.025. [DOI] [PubMed] [Google Scholar]
  • 215.Hershey GK, Friedrich MF, Esswein LA, Thomas ML, Chatila TA. The association of atopy with a gain-of-function mutation in the alpha subunit of the interleukin-4 receptor. The New England journal of medicine. 1997 Dec 11;337(24):1720–1725. doi: 10.1056/NEJM199712113372403. [DOI] [PubMed] [Google Scholar]
  • 216.Mitsuyasu H, Izuhara K, Mao XQ, et al. Ile50Val variant of IL4R alpha upregulates IgE synthesis and associates with atopic asthma. Nature genetics. 1998 Jun;19(2):119–120. doi: 10.1038/472. [DOI] [PubMed] [Google Scholar]
  • 217.Caggana M, Walker K, Reilly AA, Conroy JM, Duva S, Walsh AC. Population-based studies reveal differences in the allelic frequencies of two functionally significant human interleukin-4 receptor polymorphisms in several ethnic groups. Genetics in medicine: official journal of the American College of Medical Genetics. 1999 Sep-Oct;1(6):267–271. doi: 10.1097/00125817-199909000-00005. [DOI] [PubMed] [Google Scholar]
  • 218.Zhou G, Zhai Y, Dong X, et al. Haplotype structure and evidence for positive selection at the human IL13 locus. Molecular biology and evolution. 2004 Jan;21(1):29–35. doi: 10.1093/molbev/msg231. [DOI] [PubMed] [Google Scholar]
  • 219.Le Souef PN, Candelaria P, Goldblatt J. Evolution and respiratory genetics. The European respiratory journal. 2006 Dec;28(6):1258–1263. doi: 10.1183/09031936.06.00088006. [DOI] [PubMed] [Google Scholar]
  • 220.Obeng BB, Hartgers F, Boakye D, Yazdanbakhsh M. Out of Africa: what can be learned from the studies of allergic disorders in Africa and Africans? Current opinion in allergy and clinical immunology. 2008 Oct;8(5):391–397. doi: 10.1097/ACI.0b013e32830ebb70. [DOI] [PubMed] [Google Scholar]
  • 221.Stevenson MD, Sellins S, Grube E, et al. Aeroallergen sensitization in healthy children: racial and socioeconomic correlates. The Journal of pediatrics. 2007 Aug;151(2):187–191. doi: 10.1016/j.jpeds.2007.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Irvine AD, McLean WH. Breaking the (un)sound barrier: filaggrin is a major gene for atopic dermatitis. The Journal of investigative dermatology. 2006 Jun;126(6):1200–1202. doi: 10.1038/sj.jid.5700365. [DOI] [PubMed] [Google Scholar]
  • 223.Thomson G. Significance levels in genome scans. Advances in genetics. 2001;42:475–486. doi: 10.1016/s0065-2660(01)42037-2. [DOI] [PubMed] [Google Scholar]
  • 224.Dries DL. Genetic ancestry, population admixture, and the genetic epidemiology of complex disease. Circulation. Cardiovascular genetics. 2009 Dec;2(6):540–543. doi: 10.1161/CIRCGENETICS.109.922898. [DOI] [PubMed] [Google Scholar]
  • 225.Patterson N, Hattangadi N, Lane B, et al. Methods for high-density admixture mapping of disease genes. Am J Hum Genet. 2004 May;74(5):979–1000. doi: 10.1086/420871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Mersha T. Mapping asthma-associated variants in admixed populations. Front Genet. 2015;6(170) doi: 10.3389/fgene.2015.00292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Okada H, Kuhn C, Feillet H, Bach JF. The ‘hygiene hypothesis’ for autoimmune and allergic diseases: an update. Clinical and experimental immunology. 2010 Apr;160(1):1–9. doi: 10.1111/j.1365-2249.2010.04139.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Baye TM, Wilke RA. Mapping genes that predict treatment outcome in admixed populations. Pharmacogenomics J. 2010 Dec;10(6):465–477. doi: 10.1038/tpj.2010.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Mersha TB. Mapping asthma-associated variants in admixed populations. Frontiers in genetics. 2015;6:292. doi: 10.3389/fgene.2015.00292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Joseph CL, Ownby DR, Peterson EL, Johnson CC. Racial differences in physiologic parameters related to asthma among middle-class children. Chest. 2000 May;117(5):1336–1344. doi: 10.1378/chest.117.5.1336. [DOI] [PubMed] [Google Scholar]
  • 231.Ortega VE, Meyers DA. Pharmacogenetics: implications of race and ethnicity on defining genetic profiles for personalized medicine. The Journal of allergy and clinical immunology. 2014 Jan;133(1):16–26. doi: 10.1016/j.jaci.2013.10.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Deo RC, Patterson N, Tandon A, et al. A high-density admixture scan in 1,670 African Americans with hypertension. PLoS genetics. 2007 Nov;3(11):e196. doi: 10.1371/journal.pgen.0030196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Gravlee CC, Non AL, Mulligan CJ. Genetic ancestry, social classification, and racial inequalities in blood pressure in Southeastern Puerto Rico. PloS one. 2009;4(9):e6821. doi: 10.1371/journal.pone.0006821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Gunther T, Coop G. Robust identification of local adaptation from allele frequencies. Genetics. 2013 Sep;195(1):205–220. doi: 10.1534/genetics.113.152462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Aldrich MC, Selvin S, Wrensch MR, et al. Socioeconomic Status and Lung Cancer: Unraveling the Contribution of Genetic Admixture. American journal of public health. 2013 Oct;103(10):e73–e80. doi: 10.2105/AJPH.2013.301370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Zhu X, Luke A, Cooper RS, et al. Admixture mapping for hypertension loci with genome-scan markers. Nature genetics. 2005 Feb;37(2):177–181. doi: 10.1038/ng1510. [DOI] [PubMed] [Google Scholar]
  • 237.Reich D, Patterson N, De Jager PL, et al. A whole-genome admixture scan finds a candidate locus for multiple sclerosis susceptibility. Nature genetics. 2005 Oct;37(10):1113–1118. doi: 10.1038/ng1646. [DOI] [PubMed] [Google Scholar]
  • 238.Freedman ML, Haiman CA, Patterson N, et al. Admixture mapping identifies 8q24 as a prostate cancer risk locus in African-American men. Proceedings of the National Academy of Sciences of the United States of America. 2006 Sep 19;103(38):14068–14073. doi: 10.1073/pnas.0605832103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Reich D, Patterson N, Ramesh V, et al. Admixture mapping of an allele affecting interleukin 6 soluble receptor and interleukin 6 levels. American journal of human genetics. 2007 Apr;80(4):716–726. doi: 10.1086/513206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Choudhry S, Taub M, Mei R, et al. Genome-wide screen for asthma in Puerto Ricans: evidence for association with 5q23 region. Human genetics. 2008 Jun;123(5):455–468. doi: 10.1007/s00439-008-0495-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Galanter JM, Gignoux CR, Torgerson DG, et al. Genome-wide association study and admixture mapping identify different asthma-associated loci in Latinos: the Genes-environments & Admixture in Latino Americans study. The Journal of allergy and clinical immunology. 2014 Aug;134(2):295–305. doi: 10.1016/j.jaci.2013.08.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Torgerson DG, Gignoux CR, Galanter JM, et al. Case-control admixture mapping in Latino populations enriches for known asthma-associated genes. The Journal of allergy and clinical immunology. 2012 Jul;130(1):76–82 e12. doi: 10.1016/j.jaci.2012.02.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Torgerson DG, Capurso D, Ampleford EJ, et al. Genome-wide ancestry association testing identifies a common European variant on 6q14.1 as a risk factor for asthma in African American subjects. The Journal of allergy and clinical immunology. 2012 Sep;130(3):622–629 e629. doi: 10.1016/j.jaci.2012.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Vergara C, Murray T, Rafaels N, et al. African ancestry is a risk factor for asthma and high total IgE levels in African admixed populations. Genetic epidemiology. 2013 May;37(4):393–401. doi: 10.1002/gepi.21702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Rumpel JA, Ahmedani BK, Peterson EL, et al. Genetic ancestry and its association with asthma exacerbations among African American subjects with asthma. The Journal of allergy and clinical immunology. 2012 Dec;130(6):1302–1306. doi: 10.1016/j.jaci.2012.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Flores C, Ma SF, Pino-Yanes M, et al. African ancestry is associated with asthma risk in African Americans. PloS one. 2012;7(1):e26807. doi: 10.1371/journal.pone.0026807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Vergara C, Caraballo L, Mercado D, et al. African ancestry is associated with risk of asthma and high total serum IgE in a population from the Caribbean Coast of Colombia. Human genetics. 2009 Jun;125(5–6):565–579. doi: 10.1007/s00439-009-0649-2. [DOI] [PubMed] [Google Scholar]
  • 248.Kottyan LC, Weirauch MT, Rothenberg ME. Making it big in allergy. The Journal of allergy and clinical immunology. 2015 Jan;135(1):43–45. doi: 10.1016/j.jaci.2014.10.041. [DOI] [PubMed] [Google Scholar]
  • 249.Yick CY, Zwinderman AH, Kunst PW, et al. Transcriptome sequencing (RNA-Seq) of human endobronchial biopsies: asthma versus controls. The European respiratory journal. 2013 Sep;42(3):662–670. doi: 10.1183/09031936.00115412. [DOI] [PubMed] [Google Scholar]
  • 250.Suarez-Farinas M, Ungar B, Correa da Rosa J, et al. RNA sequencing atopic dermatitis transcriptome profiling provides insights into novel disease mechanisms with potential therapeutic implications. The Journal of allergy and clinical immunology. 2015 May;135(5):1218–1227. doi: 10.1016/j.jaci.2015.03.003. [DOI] [PubMed] [Google Scholar]
  • 251.Himes BE, Koziol-White C, Johnson M, et al. Vitamin D Modulates Expression of the Airway Smooth Muscle Transcriptome in Fatal Asthma. PloS one. 2015;10(7):e0134057. doi: 10.1371/journal.pone.0134057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Maranville JC, Baxter SS, Torres JM, Di Rienzo A. Inter-ethnic differences in lymphocyte sensitivity to glucocorticoids reflect variation in transcriptional response. The pharmacogenomics journal. 2013 Apr;13(2):121–129. doi: 10.1038/tpj.2011.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Terracciano R, Pelaia G, Preiano M, Savino R. Asthma and COPD proteomics: current approaches and future directions. Proteomics. Clinical applications. 2015 Feb;9(1–2):203–220. doi: 10.1002/prca.201400099. [DOI] [PubMed] [Google Scholar]
  • 254.Brasier AR, Victor S, Boetticher G, et al. Molecular phenotyping of severe asthma using pattern recognition of bronchoalveolar lavage-derived cytokines. The Journal of allergy and clinical immunology. 2008 Jan;121(1):30–37 e36. doi: 10.1016/j.jaci.2007.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Hwang S, Son SW, Kim SC, Kim YJ, Jeong H, Lee D. A protein interaction network associated with asthma. Journal of theoretical biology. 2008 Jun 21;252(4):722–731. doi: 10.1016/j.jtbi.2008.02.011. [DOI] [PubMed] [Google Scholar]
  • 256.Gharib SA, Nguyen EV, Lai Y, Plampin JD, Goodlett DR, Hallstrand TS. Induced sputum proteome in healthy subjects and asthmatic patients. The Journal of allergy and clinical immunology. 2011 Dec;128(6):1176–1184 e1176. doi: 10.1016/j.jaci.2011.07.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Dillon ST, Bhasin MK, Feng X, Koh DW, Daoud SS. Quantitative proteomic analysis in HCV-induced HCC reveals sets of proteins with potential significance for racial disparity. Journal of translational medicine. 2013;11:239. doi: 10.1186/1479-5876-11-239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Libes JM, Seeley EH, Li M, et al. Race disparities in peptide profiles of North American and Kenyan Wilms tumor specimens. Journal of the American College of Surgeons. 2014 Apr;218(4):707–720. doi: 10.1016/j.jamcollsurg.2013.12.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Rodriguez E, Baurecht H, Wahn AF, et al. An integrated epigenetic and transcriptomic analysis reveals distinct tissue-specific patterns of DNA methylation associated with atopic dermatitis. The Journal of investigative dermatology. 2014 Jul;134(7):1873–1883. doi: 10.1038/jid.2014.87. [DOI] [PubMed] [Google Scholar]
  • 260.Rebane A, Akdis CA. MicroRNAs in allergy and asthma. Current allergy and asthma reports. 2014 Apr;14(4):424. doi: 10.1007/s11882-014-0424-x. [DOI] [PubMed] [Google Scholar]
  • 261.Rebane A, Akdis CA. MicroRNAs: Essential players in the regulation of inflammation. The Journal of allergy and clinical immunology. 2013 Jul;132(1):15–26. doi: 10.1016/j.jaci.2013.04.011. [DOI] [PubMed] [Google Scholar]
  • 262.Jiang X. The emerging role of microRNAs in asthma. Molecular and cellular biochemistry. 2011 Jul;353(1–2):35–40. doi: 10.1007/s11010-011-0771-z. [DOI] [PubMed] [Google Scholar]
  • 263.Shaoqing Y, Ruxin Z, Guojun L, et al. Microarray analysis of differentially expressed microRNAs in allergic rhinitis. American journal of rhinology & allergy. 2011 Nov-Dec;25(6):e242–246. doi: 10.2500/ajra.2011.25.3682. [DOI] [PubMed] [Google Scholar]
  • 264.Janssen HL, Reesink HW, Lawitz EJ, et al. Treatment of HCV infection by targeting microRNA. The New England journal of medicine. 2013 May 2;368(18):1685–1694. doi: 10.1056/NEJMoa1209026. [DOI] [PubMed] [Google Scholar]
  • 265.Lu TX, Rothenberg ME. Diagnostic, functional, and therapeutic roles of microRNA in allergic diseases. The Journal of allergy and clinical immunology. 2013 Jul;132(1):3–13. doi: 10.1016/j.jaci.2013.04.039. quiz 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Tan Z, Randall G, Fan J, et al. Allele-specific targeting of microRNAs to HLA-G and risk of asthma. American journal of human genetics. 2007 Oct;81(4):829–834. doi: 10.1086/521200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Rebane A, Runnel T, Aab A, et al. MicroRNA-146a alleviates chronic skin inflammation in atopic dermatitis through suppression of innate immune responses in keratinocytes. The Journal of allergy and clinical immunology. 2014 Oct;134(4):836–847 e811. doi: 10.1016/j.jaci.2014.05.022. [DOI] [PubMed] [Google Scholar]
  • 268.Wang IJ, Chen SL, Lu TP, Chuang EY, Chen PC. Prenatal smoke exposure, DNA methylation, and childhood atopic dermatitis. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2013 May;43(5):535–543. doi: 10.1111/cea.12108. [DOI] [PubMed] [Google Scholar]
  • 269.Baccarelli A, Wright RO, Bollati V, et al. Rapid DNA methylation changes after exposure to traffic particles. American journal of respiratory and critical care medicine. 2009 Apr 1;179(7):572–578. doi: 10.1164/rccm.200807-1097OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Perera F, Tang WY, Herbstman J, et al. Relation of DNA methylation of 5′-CpG island of ACSL3 to transplacental exposure to airborne polycyclic aromatic hydrocarbons and childhood asthma. PloS one. 2009;4(2):e4488. doi: 10.1371/journal.pone.0004488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Lovinsky-Desir S, Miller RL. Epigenetics, asthma, and allergic diseases: a review of the latest advancements. Current allergy and asthma reports. 2012 Jun;12(3):211–220. doi: 10.1007/s11882-012-0257-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Dogan MV, Shields B, Cutrona C, et al. The effect of smoking on DNA methylation of peripheral blood mononuclear cells from African American women. BMC genomics. 2014;15:151. doi: 10.1186/1471-2164-15-151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and implications. Nature reviews. Genetics. 2011 Feb;13(2):97–109. doi: 10.1038/nrg3142. [DOI] [PubMed] [Google Scholar]
  • 274.Feinberg AP, Fallin MD. Epigenetics at the Crossroads of Genes and the Environment. Jama. 2015 Sep 15;314(11):1129–1130. doi: 10.1001/jama.2015.10414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Heyn H, Moran S, Hernando-Herraez I, et al. DNA methylation contributes to natural human variation. Genome research. 2013 Sep;23(9):1363–1372. doi: 10.1101/gr.154187.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Caliskan M, Bochkov YA, Kreiner-Moller E, et al. Rhinovirus wheezing illness and genetic risk of childhood-onset asthma. The New England journal of medicine. 2013 Apr 11;368(15):1398–1407. doi: 10.1056/NEJMoa1211592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Gutierrez-Arcelus M, Ongen H, Lappalainen T, et al. Tissue-specific effects of genetic and epigenetic variation on gene regulation and splicing. PLoS genetics. 2015 Jan;11(1):e1004958. doi: 10.1371/journal.pgen.1004958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Zhang D, Cheng L, Badner JA, et al. Genetic control of individual differences in gene-specific methylation in human brain. American journal of human genetics. 2010 Mar 12;86(3):411–419. doi: 10.1016/j.ajhg.2010.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Bunyavanich S, Schadt EE. Systems biology of asthma and allergic diseases: a multiscale approach. The Journal of allergy and clinical immunology. 2015 Jan;135(1):31–42. doi: 10.1016/j.jaci.2014.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Grice EA, Kong HH, Conlan S, et al. Topographical and temporal diversity of the human skin microbiome. Science. 2009 May 29;324(5931):1190–1192. doi: 10.1126/science.1171700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Williams MR, Gallo RL. The Role of the Skin Microbiome in Atopic Dermatitis. Current allergy and asthma reports. 2015 Nov;15(11):65. doi: 10.1007/s11882-015-0567-4. [DOI] [PubMed] [Google Scholar]
  • 282.Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006 Feb 24;124(4):837–848. doi: 10.1016/j.cell.2006.02.017. [DOI] [PubMed] [Google Scholar]
  • 283.Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010 Mar 4;464(7285):59–65. doi: 10.1038/nature08821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Grice EA, Segre JA. The human microbiome: our second genome. Annual review of genomics and human genetics. 2012;13:151–170. doi: 10.1146/annurev-genom-090711-163814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Huang YJ, Charlson ES, Collman RG, Colombini-Hatch S, Martinez FD, Senior RM. The role of the lung microbiome in health and disease. A National Heart, Lung, and Blood Institute workshop report. American journal of respiratory and critical care medicine. 2013 Jun 15;187(12):1382–1387. doi: 10.1164/rccm.201303-0488WS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Von Ehrenstein OS, Von Mutius E, Illi S, Baumann L, Bohm O, von Kries R. Reduced risk of hay fever and asthma among children of farmers. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2000 Feb;30(2):187–193. doi: 10.1046/j.1365-2222.2000.00801.x. [DOI] [PubMed] [Google Scholar]
  • 287.Riedler J, Braun-Fahrlander C, Eder W, et al. Exposure to farming in early life and development of asthma and allergy: a cross-sectional survey. Lancet. 2001 Oct 6;358(9288):1129–1133. doi: 10.1016/S0140-6736(01)06252-3. [DOI] [PubMed] [Google Scholar]
  • 288.Alfven T, Braun-Fahrlander C, Brunekreef B, et al. Allergic diseases and atopic sensitization in children related to farming and anthroposophic lifestyle–the PARSIFAL study. Allergy. 2006 Apr;61(4):414–421. doi: 10.1111/j.1398-9995.2005.00939.x. [DOI] [PubMed] [Google Scholar]
  • 289.Genuneit J, Strachan DP, Buchele G, et al. The combined effects of family size and farm exposure on childhood hay fever and atopy. Pediatric allergy and immunology: official publication of the European Society of Pediatric Allergy and Immunology. 2013 May;24(3):293–298. doi: 10.1111/pai.12053. [DOI] [PubMed] [Google Scholar]
  • 290.Liu AH. Revisiting the hygiene hypothesis for allergy and asthma. The Journal of allergy and clinical immunology. 2015 Oct;136(4):860–865. doi: 10.1016/j.jaci.2015.08.012. [DOI] [PubMed] [Google Scholar]
  • 291.Brooks C, Pearce N, Douwes J. The hygiene hypothesis in allergy and asthma: an update. Current opinion in allergy and clinical immunology. 2013 Feb;13(1):70–77. doi: 10.1097/ACI.0b013e32835ad0d2. [DOI] [PubMed] [Google Scholar]
  • 292.Correa P, Piazuelo MB. Evolutionary History of the Helicobacter pylori Genome: Implications for Gastric Carcinogenesis. Gut and liver. 2012 Jan;6(1):21–28. doi: 10.5009/gnl.2012.6.1.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Sitaraman R. Allergies, Helicobacter pylori and the continental enigmas. Frontiers in microbiology. 2015;6:578. doi: 10.3389/fmicb.2015.00578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Kaminsky DA. Systems biology approach for subtyping asthma; where do we stand now? Current opinion in pulmonary medicine. 2014 Jan;20(1):17–22. doi: 10.1097/MCP.0000000000000004. [DOI] [PubMed] [Google Scholar]
  • 295.Mc Mahon SS, Sim A, Filippi S, et al. Information theory and signal transduction systems: from molecular information processing to network inference. Seminars in cell & developmental biology. 2014 Nov;35:98–108. doi: 10.1016/j.semcdb.2014.06.011. [DOI] [PubMed] [Google Scholar]
  • 296.Milenkovic O, Alterovitz G, Battail G, et al. Introduction to the Special Issue on Information Theory in Molecular Biology and Neuroscience. Information Theory, IEEE Transactions. 2010;56(2):649–652. [Google Scholar]
  • 297.Greene CS, Tan J, Ung M, Moore JH, Cheng C. Big data bioinformatics. Journal of cellular physiology. 2014 Dec;229(12):1896–1900. doi: 10.1002/jcp.24662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Clarke R, Ressom HW, Wang A, et al. The properties of high-dimensional data spaces: implications for exploring gene and protein expression data. Nature reviews. Cancer. 2008 Jan;8(1):37–49. doi: 10.1038/nrc2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Sittka A, Vera J, Lai X, Schmeck BT. Asthma phenotyping, therapy, and prevention: what can we learn from systems biology? Pediatric research. 2013 Apr;73(4 Pt 2):543–552. doi: 10.1038/pr.2013.8. [DOI] [PubMed] [Google Scholar]
  • 300.Wheelock CE, Goss VM, Balgoma D, et al. Application of ‘omics technologies to biomarker discovery in inflammatory lung diseases. The European respiratory journal. 2013 Sep;42(3):802–825. doi: 10.1183/09031936.00078812. [DOI] [PubMed] [Google Scholar]
  • 301.Mikhaylova L, Zhang Y, Kobzik L, Fedulov AV. Link between epigenomic alterations and genome-wide aberrant transcriptional response to allergen in dendritic cells conveying maternal asthma risk. PloS one. 2013;8(8):e70387. doi: 10.1371/journal.pone.0070387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Pinart M, Benet M, Annesi-Maesano I, et al. Comorbidity of eczema, rhinitis, and asthma in IgE-sensitised and non-IgE-sensitised children in MeDALL: a population-based cohort study. The Lancet. Respiratory medicine. 2014 Feb;2(2):131–140. doi: 10.1016/S2213-2600(13)70277-7. [DOI] [PubMed] [Google Scholar]
  • 303.Curtiss FR. Atopic march to a dead end or does the theory really have legs? Journal of managed care pharmacy: JMCP. 2007 Nov-Dec;13(9):810–811. doi: 10.18553/jmcp.2007.13.9.810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Hudson TJ. Skin barrier function and allergic risk. Nature genetics. 2006 Apr;38(4):399–400. doi: 10.1038/ng0406-399. [DOI] [PubMed] [Google Scholar]
  • 305.Cork MJ, Robinson DA, Vasilopoulos Y, et al. New perspectives on epidermal barrier dysfunction in atopic dermatitis: gene-environment interactions. The Journal of allergy and clinical immunology. 2006 Jul;118(1):3–21. doi: 10.1016/j.jaci.2006.04.042. quiz 22–23. [DOI] [PubMed] [Google Scholar]
  • 306.Vercelli D. Discovering susceptibility genes for asthma and allergy. Nature reviews. Immunology. 2008 Mar;8(3):169–182. doi: 10.1038/nri2257. [DOI] [PubMed] [Google Scholar]
  • 307.Litonjua AA, Tantisira KG, Lake S, et al. Polymorphisms in signal transducer and activator of transcription 3 and lung function in asthma. Respiratory research. 2005;6:52. doi: 10.1186/1465-9921-6-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Finkelman FD, Vercelli D. Advances in asthma, allergy mechanisms, and genetics in 2006. The Journal of allergy and clinical immunology. 2007 Sep;120(3):544–550. doi: 10.1016/j.jaci.2007.05.025. [DOI] [PubMed] [Google Scholar]
  • 309.Schedel M, Carr D, Klopp N, et al. A signal transducer and activator of transcription 6 haplotype influences the regulation of serum IgE levels. The Journal of allergy and clinical immunology. 2004 Nov;114(5):1100–1105. doi: 10.1016/j.jaci.2004.07.048. [DOI] [PubMed] [Google Scholar]
  • 310.Soderhall C, Marenholz I, Kerscher T, et al. Variants in a novel epidermal collagen gene (COL29A1) are associated with atopic dermatitis. PLoS biology. 2007 Sep;5(9):e242. doi: 10.1371/journal.pbio.0050242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Vercelli D. Genetics and biology of asthma 2010: La’ ci darem la mano. The Journal of allergy and clinical immunology. 2010 Feb;125(2):347–348. doi: 10.1016/j.jaci.2009.12.976. [DOI] [PubMed] [Google Scholar]
  • 312.Van Eerdewegh P, Little RD, Dupuis J, et al. Association of the ADAM33 gene with asthma and bronchial hyperresponsiveness. Nature. 2002 Jul 25;418(6896):426–430. doi: 10.1038/nature00878. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

1

Table E1: Genes ranked by level of association with each atopic disorder based on mining of 15.73 million PubMed abstracts using Literature Lab™ from Acumenta Biotech Software.

Table E2: IPA summary of associated networks, molecular and cellular functions, diseases and disorders and canonical pathways for the 16 genes shared among FA, AD, AR and asthma.

2

RESOURCES