Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Genome-wide association analysis of eosinophilic esophagitis provides insight into the tissue specificity of this allergic disease

Abstract

Eosinophilic esophagitis (EoE) is a chronic inflammatory disorder associated with allergic hypersensitivity to food. We interrogated >1.5 million genetic variants in EoE cases of European ancestry and subsequently in a multi-site cohort with local and out-of-study control subjects. In addition to replicating association of the 5q22 locus (meta-analysis P = 1.9 × 10−16), we identified an association at 2p23 spanning CAPN14 (P = 2.5 × 10−10). CAPN14 was specifically expressed in the esophagus, was dynamically upregulated as a function of disease activity and genetic haplotype and after exposure of epithelial cells to interleukin (IL)-13, and was located in an epigenetic hotspot modified by IL-13. Genes neighboring the top 208 EoE-associated sequence variants were enriched for esophageal expression, and multiple loci for allergic sensitization were associated with EoE susceptibility (4.8 × 10−2 < P < 5.1 × 10−11). We propose a model to explain the tissue-specific nature of EoE that involves the interplay of allergic sensitization with an EoE-specific, IL-13–inducible esophageal response involving CAPN14.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Manhattan plot of the P values obtained from the genome-wide association analysis.
Figure 2: CAPN14 is specifically expressed in esophageal epithelium and is dynamically upregulated as a function of disease activity and genetic haplotype and after exposure of epithelial cells to IL-13.
Figure 3: IL-13 induces CAPN14 expression and calpain activity.
Figure 4: Genes at EoE risk loci with differential expression in biopsies from EoE cases relative to controls.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Liacouras, C.A. et al. Eosinophilic esophagitis: updated consensus recommendations for children and adults. J. Allergy Clin. Immunol. 128, 3–20 (2011).

    PubMed  Google Scholar 

  2. Rothenberg, M.E. Biology and treatment of eosinophilic esophagitis. Gastroenterology 137, 1238–1249 (2009).

    CAS  PubMed  Google Scholar 

  3. Collins, M.H. et al. Clinical, pathologic, and molecular characterization of familial eosinophilic esophagitis compared with sporadic cases. Clin. Gastroenterol. Hepatol. 6, 621–629 (2008).

    PubMed  PubMed Central  Google Scholar 

  4. Putnam, P.E. & Rothenberg, M.E. Eosinophilic esophagitis: concepts, controversies, and evidence. Curr. Gastroenterol. Rep. 11, 220–225 (2009).

    PubMed  Google Scholar 

  5. Abonia, J.P. & Rothenberg, M.E. Eosinophilic esophagitis: rapidly advancing insights. Annu. Rev. Med. 63, 421–434 (2012).

    CAS  PubMed  Google Scholar 

  6. Davis, B.P. & Rothenberg, M.E. Emerging concepts of dietary therapy for pediatric and adult eosinophilic esophagitis. Expert Rev. Clin. Immunol. 9, 285–287 (2013).

    CAS  PubMed  Google Scholar 

  7. Henderson, C.J. et al. Comparative dietary therapy effectiveness in remission of pediatric eosinophilic esophagitis. J. Allergy Clin. Immunol. 129, 1570–1578 (2012).

    PubMed  PubMed Central  Google Scholar 

  8. Rothenberg, M.E. et al. Working with the US Food and Drug Administration: progress and timelines in understanding and treating patients with eosinophilic esophagitis. J. Allergy Clin. Immunol. 130, 617–619 (2012).

    PubMed  Google Scholar 

  9. Rothenberg, M.E. et al. Common variants at 5q22 associate with pediatric eosinophilic esophagitis. Nat. Genet. 42, 289–291 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Sherrill, J.D. et al. Variants of thymic stromal lymphopoietin and its receptor associate with eosinophilic esophagitis. J. Allergy Clin. Immunol. 126, 160–165 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Zuo, L. et al. IL-13 induces esophageal remodeling and gene expression by an eosinophil-independent, IL-13Rα2–inhibited pathway. J. Immunol. 185, 660–669 (2010).

    CAS  PubMed  Google Scholar 

  12. Blanchard, C. et al. Coordinate interaction between IL-13 and epithelial differentiation cluster genes in eosinophilic esophagitis. J. Immunol. 184, 4033–4041 (2010).

    CAS  PubMed  Google Scholar 

  13. Blanchard, C. et al. Eotaxin-3 and a uniquely conserved gene-expression profile in eosinophilic esophagitis. J. Clin. Invest. 116, 536–547 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. McAleer, M.A. & Irvine, A.D. The multifunctional role of filaggrin in allergic skin disease. J. Allergy Clin. Immunol. 131, 280–291 (2013).

    CAS  PubMed  Google Scholar 

  15. Fallon, P.G. et al. A homozygous frameshift mutation in the mouse Flg gene facilitates enhanced percutaneous allergen priming. Nat. Genet. 41, 602–608 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Shin, H.D. et al. Association of Eotaxin gene family with asthma and serum total IgE. Hum. Mol. Genet. 12, 1279–1285 (2003).

    CAS  PubMed  Google Scholar 

  17. Marchini, J., Howie, B., Myers, S., McVean, G. & Donnelly, P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat. Genet. 39, 906–913 (2007).

    CAS  PubMed  Google Scholar 

  18. Altshuler, D.M. et al. Integrating common and rare genetic variation in diverse human populations. Nature 467, 52–58 (2010).

    CAS  PubMed  Google Scholar 

  19. Ferreira, M.A. et al. Identification of IL6R and chromosome 11q13.5 as risk loci for asthma. Lancet 378, 1006–1014 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Tang, H.Y. et al. Association analysis of single nucleotide polymorphisms at five loci: comparison between atopic dermatitis and asthma in the Chinese Han population. PLoS ONE 7, e35334 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Esparza-Gordillo, J. et al. A common variant on chromosome 11q13 is associated with atopic dermatitis. Nat. Genet. 41, 596–601 (2009).

    CAS  PubMed  Google Scholar 

  22. Greisenegger, E.K., Zimprich, F., Zimprich, A., Gleiss, A. & Kopp, T. Association of the chromosome 11q13.5 variant with atopic dermatitis in Austrian patients. Eur. J. Dermatol. 23, 142–145 (2013).

    CAS  PubMed  Google Scholar 

  23. Hirota, T. et al. Genome-wide association study identifies eight new susceptibility loci for atopic dermatitis in the Japanese population. Nat. Genet. 44, 1222–1226 (2012).

    CAS  PubMed  Google Scholar 

  24. Paternoster, L. et al. Meta-analysis of genome-wide association studies identifies three new risk loci for atopic dermatitis. Nat. Genet. 44, 187–192 (2012).

    CAS  Google Scholar 

  25. Barrett, J.C. et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat. Genet. 40, 955–962 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Ramasamy, A. et al. A genome-wide meta-analysis of genetic variants associated with allergic rhinitis and grass sensitization and their interaction with birth order. J. Allergy Clin. Immunol. 128, 996–1005 (2011).

    CAS  PubMed  Google Scholar 

  27. Tran, D.Q. et al. GARP (LRRC32) is essential for the surface expression of latent TGF-β on platelets and activated FOXP3+ regulatory T cells. Proc. Natl. Acad. Sci. USA 106, 13445–13450 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Fuentebella, J. et al. Increased number of regulatory T cells in children with eosinophilic esophagitis. J. Pediatr. Gastroenterol. Nutr. 51, 283–289 (2010).

    PubMed  Google Scholar 

  29. Stuck, M.C., Straumann, A. & Simon, H.U. Relative lack of T regulatory cells in adult eosinophilic esophagitis—no normalization after corticosteroid therapy. Allergy 66, 705–707 (2011).

    CAS  PubMed  Google Scholar 

  30. Tantibhaedhyangkul, U., Tatevian, N., Gilger, M.A., Major, A.M. & Davis, C.M. Increased esophageal regulatory T cells and eosinophil characteristics in children with eosinophilic esophagitis and gastroesophageal reflux disease. Ann. Clin. Lab. Sci. 39, 99–107 (2009).

    CAS  PubMed  Google Scholar 

  31. Zhu, X., Wang, M., Crump, C.H. & Mishra, A. An imbalance of esophageal effector and regulatory T cell subsets in experimental eosinophilic esophagitis in mice. Am. J. Physiol. Gastrointest. Liver Physiol. 297, G550–G558 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Frischmeyer-Guerrerio, P.A. et al. TGFβ receptor mutations impose a strong predisposition for human allergic disease. Sci. Transl. Med. 5, 195ra94 (2013).

    PubMed  PubMed Central  Google Scholar 

  33. Maher, B. ENCODE: the human encyclopaedia. Nature 489, 46–48 (2012).

    PubMed  Google Scholar 

  34. Ecker, J.R. et al. Genomics: ENCODE explained. Nature 489, 52–55 (2012).

    CAS  PubMed  Google Scholar 

  35. Skipper, M., Dhand, R. & Campbell, P. Presenting ENCODE. Nature 489, 45 (2012).

    CAS  PubMed  Google Scholar 

  36. Ueta, M., Mizushima, K., Yokoi, N., Naito, Y. & Kinoshita, S. Expression of the interleukin-4 receptor α in human conjunctival epithelial cells. Br. J. Ophthalmol. 94, 1239–1243 (2010).

    PubMed  Google Scholar 

  37. Ueta, M., Sotozono, C. & Kinoshita, S. Expression of interleukin-4 receptor α in human corneal epithelial cells. Jpn. J. Ophthalmol. 55, 405–410 (2011).

    CAS  PubMed  Google Scholar 

  38. Sherrill, J.D. et al. Analysis and expansion of the eosinophilic esophagitis transcriptome by RNA sequencing. Genes Immun. doi: 10.1038/gene.2014.27 (12 June 2014).

    CAS  Google Scholar 

  39. Bønnelykke, K. et al. Meta-analysis of genome-wide association studies identifies ten loci influencing allergic sensitization. Nat. Genet. 45, 902–906 (2013).

    PubMed  PubMed Central  Google Scholar 

  40. Hinds, D.A. et al. A genome-wide association meta-analysis of self-reported allergy identifies shared and allergy-specific susceptibility loci. Nat. Genet. 45, 907–911 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Krahn, M. et al. Eosinophilic infiltration related to CAPN3 mutations: a pathophysiological component of primary calpainopathy? Clin. Genet. 80, 398–402 (2011).

    CAS  PubMed  Google Scholar 

  42. Krahn, M. et al. CAPN3 mutations in patients with idiopathic eosinophilic myositis. Ann. Neurol. 59, 905–911 (2006).

    CAS  PubMed  Google Scholar 

  43. Bartoli, M. et al. Validation of comparative genomic hybridization arrays for the detection of genomic rearrangements of the calpain-3 and dysferlin genes. Clin. Genet. 81, 99–101 (2012).

    CAS  PubMed  Google Scholar 

  44. Brown, R.H. Jr. & Amato, A. Calpainopathy and eosinophilic myositis. Ann. Neurol. 59, 875–877 (2006).

    PubMed  Google Scholar 

  45. Amato, A.A. Adults with eosinophilic myositis and calpain-3 mutations. Neurology 70, 730–731 (2008).

    PubMed  Google Scholar 

  46. Oflazer, P.S., Gundesli, H., Zorludemir, S., Sabuncu, T. & Dincer, P. Eosinophilic myositis in calpainopathy: could immunosuppression of the eosinophilic myositis alter the early natural course of the dystrophic disease? Neuromuscul. Disord. 19, 261–263 (2009).

    PubMed  Google Scholar 

  47. Sorimachi, H., Hata, S. & Ono, Y. Calpain chronicle—an enzyme family under multidisciplinary characterization. Proc. Jpn. Acad., Ser. B, Phys. Biol. Sci. 87, 287–327 (2011).

    CAS  Google Scholar 

  48. Arnandis, T. et al. Differential functions of calpain 1 during epithelial cell death and adipocyte differentiation in mammary gland involution. Biochem. J. 459, 355–368 (2014).

    CAS  PubMed  Google Scholar 

  49. Wang, Y. & Zhang, Y. Regulation of TET protein stability by calpains. Cell Rep. 6, 278–284 (2014).

    PubMed  PubMed Central  Google Scholar 

  50. Meephansan, J., Tsuda, H., Komine, M., Tominaga, S. & Ohtsuki, M. Regulation of IL-33 expression by IFN-γ and tumor necrosis factor-α in normal human epidermal keratinocytes. J. Invest. Dermatol. 132, 2593–2600 (2012).

    CAS  PubMed  Google Scholar 

  51. Hayakawa, M. et al. Mature interleukin-33 is produced by calpain-mediated cleavage in vivo. Biochem. Biophys. Res. Commun. 387, 218–222 (2009).

    CAS  PubMed  Google Scholar 

  52. Wu, C. et al. BioGPS: an extensible and customizable portal for querying and organizing gene annotation resources. Genome Biol. 10, R130 (2009).

    PubMed  PubMed Central  Google Scholar 

  53. McCall, M.N., Uppal, K., Jaffee, H.A., Zilliox, M.J. & Irizarry, R.A. The Gene Expression Barcode: leveraging public data repositories to begin cataloging the human and murine transcriptomes. Nucleic Acids Res. 39, D1011–D1015 (2011).

    CAS  PubMed  Google Scholar 

  54. Prahalad, S. et al. Juvenile rheumatoid arthritis: linkage to HLA demonstrated by allele sharing in affected sibpairs. Arthritis Rheum. 43, 2335–2338 (2000).

    CAS  PubMed  Google Scholar 

  55. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Pruim, R.J. et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26, 2336–2337 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Zimmermann, N. et al. Dissection of experimental asthma with DNA microarray analysis identifies arginase in asthma pathogenesis. J. Clin. Invest. 111, 1863–1874 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Flicek, P. et al. Ensembl 2013. Nucleic Acids Res. 41, D48–D55 (2013).

    CAS  PubMed  Google Scholar 

  59. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    PubMed  PubMed Central  Google Scholar 

  62. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We kindly thank the participants and their families. This study would not be possible without the CoFAR site investigators: S.H. Sicherer, S.M. Jones, T.T. Perry, A. Scurlock, E. Kim, E. Dellon, A.W. Burks, G.T. Furuta, D.Y.M. Leung and D. Stablein. Other people who contributed to this study include J. Gau, J. Fronda, D. Ananos, K. Mudd, T. Fitzgerald, P. Chandler, E. O'Brien, S. Carlisle, A. Hiegel, P. Steele, J. Gebhart, D. Hamilton, W. Moore, J. Newton, S. Leung, D. Brown, M. Plaut and J. Poyser.

Funding for this project was provided by the US NIH, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Heart, Lung, and Blood Institute (NHLBI) and National Human Genome Research Institute (NHGRI) (U19 AI066738, U01 HG006828, U01 HG006828-S1, U01 HG006828-S2, U01 AI066560, R37 AI024717, P01 AI083194, T32 HL7752-19, K23 AI099083 and P01 AR049084). The CoFAR arm of the study was supported by US NIH grant U19 AI066738 from NIAID and NIDDK. The project was also supported by several grants from the National Center for Research Resources (NCRR), a component of the US NIH: UL1 TR001082 (National Jewish), UL1 TR-000067 (Mount Sinai), UL1 TR-000039 (Arkansas), UL1 TR-000083 (University of North Carolina) and UL1 TR-000424 (Johns Hopkins). Support was also received from the US Department of Veteran Affairs (IMMA 9) and the US Department of Defense (PR094002). This research was supported in part by the Cincinnati Children's Research Foundation and its Cincinnati Genomic Control Cohort. Other support was derived from the Campaign Urging Research for Eosinophilic Diseases (CURED), the Buckeye Foundation, the Food Allergy Research Education (FARE) Foundation and the Foundation of the American College of Allergy, Asthma and Immunology.

The paper's contents are solely the responsibility of the authors and do not necessarily represent the official view of the NCRR or NIH. We also thank S. Hottinger for editorial assistance.

Author information

Authors and Affiliations

Authors

Contributions

M.E.R. and J.B.H. conceived the project and supervised the study. L.C.K. and M.E.R. wrote the manuscript. L.C.K. led and performed the genetic analysis. H.H. and L.J.M. contributed statistical consultation and support. B.P.D., J.D.S. and M.R. performed the experiments with assistance from E.M.S. and K.A.K., K.L., K.K., M.T.W., A.M.R., M.K., P.D. and S.V. provided analytical and bioinformatics support. S.L. genotyped samples with assistance from A.M., M.C., R.A.W., R.D.P., B.P.V., D.M.F., R.L. and H.A.S. provided samples through the CoFAR consortium. V.A.M., P.E.P., J.P.A. and M.E.R. provided samples for the CCHMC cohort. All of the authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Marc E Rothenberg.

Ethics declarations

Competing interests

M.E.R., J.B.H. and L.C.K. are co-inventors on a Cincinnati Children's Hospital-owned patent application concerning some of the content in this manuscript.

Integrated supplementary information

Supplementary Figure 1 Manhattan plot of the P values obtained from the genome-wide association analysis.

Data are from 1,468,075 genetic variants with minor allele frequencies greater than 1% in the subjects with EoE in 514 EoE cases and 6,758 controls (a) and 222 cases and 2,488 controls (b). The –log value of the probability is shown as a function of genomic position on the autosomes. Genome-wide significance (red dotted line, P ≤ 5 × 10–8) and suggestive significance (blue line, P ≤ 10–7) are indicated.

Supplementary Figure 2 Expression of XKR6.

(Data collected from http://www.biogps.org/). The colors of the bars correspond to the tissue source.

Supplementary Figure 3 Residual association at 5q22 after adjusting for rs1438672.

After adjusting for any of the most highly associated variants in the region (rs1438672), a group of 12 variants between TSLP and WDR36 demonstrate residual association (0.001 < P < 0.05). After accounting for multiple testing, we cannot reject the null hypothesis that there is one independent genetic effect in the region. P values (–log10) for the genetic association analysis are plotted as a function of the genomic positions of each imputed variant (minor allele frequency (MAF) > 0.01) on chromosome 5 (Chr5) using a logistic regression model adjusting for the genotype of rs1438672. Genes in the region are shown below. The blue lines indicate the recombination rates in cM per Mb using HapMap controls.

Supplementary Figure 4 Expression of CAPN14.

(Data collected from http://www.biogps.org/). The colors of the bars correspond to the tissue source.

Supplementary Figure 5 Fine mapping of loci associated (replicated and 5 × 10–8 < P < 10–6) with EoE risk.

P values (–log10) for the genetic association analysis of imputed variants on the y axis are plotted as a function of the genomic position of each genotyped and imputed SNP (MAF > 0.05). Genes in the region are shown below. The LD values (r2) between the lead SNP and the other SNPs as assessed in the March 2012 release of the 1000 Genomes Project are indicated in different colors. The blue lines indicate the recombination rates in cM per Mb using HapMap controls.

Supplementary Figure 6 Expression of genes near EoE-associated genetic variants (P < 10–4) in the esophageal biopsies of EoE cases versus controls.

In this data set, 9 of the 215 transcripts (representing 208 genes) have an average difference of ≥2 FPKM between esophagi from active EoE cases and controls. The differences in expression were sufficient to segregate EoE cases from controls. The eight transcripts with differences in expression of greater than twofold can be found in Figure 4. Subjects with EoE are noted with blue bars, and subjects without EoE are indicated with red bars. All expression is normalized to the average expression in esophageal biopsies without EoE.

Supplementary Figure 7 Two genes including CAPN14 are replicated in EoE genetic, expression and epigenetic analyses.

We assessed overlap from analyses of genes within 25 kb of genetic variants with P < 10–4, genes with increased H3K27ac levels after 6-h exposure to IL-13 (P < 0.01, M < –1 at 5 kb from TSS) and genes with increased esophageal expression in EoE (twofold increase in expression in esophageal biopsy in EoE compared to control, corrected P < 0.05).

Supplementary Figure 8 Proposed model of increased EoE risk at the CAPN14 locus.

CAPN14 is expressed specifically in the esophagus (Fig. 2a). Allergic inflammatory mediators including IL-13 and IL-4 induce CAPN14 expression and activity (Figs. 2h and 3a and refs. 36, 37), and regulation of increased expression is mediated in part through the acetylation of histones (Fig. 2i). The abundance of IL-13 and IL-4 in the esophagus of patients with EoE5–8results in increased CAPN14 expression (Fig. 2b–d) and activity (Fig. 3b), and the calpain activity of CAPN14 potentially attenuates further inflammation by digesting endogenous proteins. The genetic variants associated with EoE risk at the CAPN14 locus lead to decreased CAPN14 expression (Fig. 2e) feasibly through the binding of a protein (Fig. 2j), a transcription factor that potentially acts as a transcriptional repressor. Together, our data are consistent with a model (a) in which CAPN14 is induced along with other anti-inflammatory agents such as microRNAs, cell surface receptors and T regulatory cells. (b) When exposed to IL-13 and IL-4, chromosomes in epithelial cells with the CAPN14 EoE risk allele are unable to induce CAPN14 expression to the extent of chromosomes with the non-risk allele. We propose a model in which the allelic change in CAPN14 expression dysregulates a critical negative feedback loop in the esophagus resulting in increased risk of pathology and EoE.

Supplementary Figure 9 Association of variants in loci previously reported to be associated with allergic sensitization with EoE risk.

The variant that was most highly associated in the allergic sensitization study is indicated by a purple diamond and is given in each panel. The LD values (r2) between the lead SNP and the other SNPs as assessed in the March 2012 release of the 1000 Genomes Project are indicated in different colors. The blue lines behind the genetic variants indicate the recombination rates in cM per Mb using HapMap controls.

Supplementary Figure 10 Flow chart of the EoE GWAS analytical pipeline.

External controls from the University of Michigan (acquired through dbGaP) were randomly assigned to the CCHMC and CoFAR cohorts. CoFAR, NIH Consortium of Food Allergy Research; EoE, eosinophilic esophagitis; GWAS, genome-wide association study; MAF, minor allele frequency; SNP, single-nucleotide polymorphism.

Supplementary Figure 11 The Genome Studio cluster plot for rs8041227 (also known as kgp344467) at 15q13 from the OMNI5 analysis (all EoE cases and local controls).

The normalized theta (Norm Theta) value is the ratio of the signal intensities assayed for the A and B forms of the rs8041227 allele against normalized R (signal intensity; Norm R). Subjects in this graph are homozygous for the risk allele (red), heterozygous (purple) or homozygous for the non-risk allele (blue).

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–6 and Supplementary Figures 1–11 (PDF 2812 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kottyan, L., Davis, B., Sherrill, J. et al. Genome-wide association analysis of eosinophilic esophagitis provides insight into the tissue specificity of this allergic disease. Nat Genet 46, 895–900 (2014). https://doi.org/10.1038/ng.3033

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3033

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing