Reviews and feature article
Genetic dissection of eosinophilic esophagitis provides insight into disease pathogenesis and treatment strategies

https://doi.org/10.1016/j.jaci.2011.03.046Get rights and content

Eosinophilic esophagitis (EoE) is a chronic inflammatory disorder of the esophagus that is compounded by both genetic predisposition and aberrant responses to environmental antigens, particularly those that are food derived. Data have indicated a unique transcriptional response in vivo that defines EoE and that appears to be partially attributable to the TH2 cytokine IL-13. Moreover, a number of genetic risk variants in proinflammatory and epithelial cell genes associate with EoE susceptibility, demonstrating novel heritable mechanisms that contribute to disease risk. Here we discuss recent advances in our understanding of the intrinsic (genetic) and extrinsic (environmental) components that illustrate the complex nature of EoE.

Section snippets

Epidemiology

With the expansion of EoE cases reported worldwide, multiple studies have aimed to establish a baseline prevalence of EoE and determine whether disease incidence has increased. From 2000-2003, the estimated prevalence of EoE in a pediatric population was approximately 4 in 10,000, with an incidence rate of 0.9 to 1.3 in 10,000 new cases per year.5 A similar prevalence (approximately 2 cases per 10,000) and incidence rate (1.4 per 100,000) during a 16-year period were observed in an adult Swiss

Genetic heritability

An underlying genetic predisposition to EoE has been proposed by multiple groups that show a disproportionate prevalence of disease in white subjects and male subjects and within families of affected subjects.15, 23 For instance, data over a 14-year period demonstrated that 90% of the patients with EoE were white and 75% were male.15 Reports of a familial occurrence of EoE and esophageal dilatation in 6.8% and 9.7% of patients with EoE, respectively, suggest that the prevalence of EoE and

Transcriptome analysis

A major step toward the molecular mapping of EoE was achieved when gene expression profiling of patients’ esophageal biopsy specimens showed a remarkable transcript signature that distinguishes patients with EoE from healthy control subjects and patients with chronic esophagitis.27 Altered expression of approximately 574 genes comprises this EoE “transcriptome,” which exhibits a high level of conservation among patients’ sex, age, and atopic history and strongly correlates with esophageal

Genetic variants and disease susceptibility

The number of studies investigating genetic variants associated with EoE are few compared with those for other more common and more widely recognized atopic diseases, such as AD and asthma. Regardless, there have been significant strides in uncovering EoE risk variants in a relatively short period of time14, 27, 52, 53 due in part to the technological advances in genotyping single nucleotide polymorphisms (SNPs) in large case-control cohorts. In all, there have been 4 candidate gene studies

Conclusions and future directions

In just over 10 years since the recognition of EoE as a distinct inflammatory disorder, the rapid progress toward characterizing the disease on multiple fronts has underscored its complexity. We now have insight into the natural history of EoE, its strong association with specific ethnicities and sexes, the genetic and environmental factors involved, and the molecular pathogenesis of the disease (Fig 1). Moreover, the burst of data illustrating EoE risk variants in CCL26, TGFB1, TSLP and CRLF2,

References (81)

  • C. Blanchard et al.

    Eosinophilic esophagitis: pathogenesis, genetics, and therapy

    J Allergy Clin Immunol

    (2006)
  • M.H. Collins et al.

    Clinical, pathologic, and molecular characterization of familial eosinophilic esophagitis compared with sporadic cases

    Clin Gastroenterol Hepatol

    (2008)
  • C. Blanchard et al.

    IL-13 involvement in eosinophilic esophagitis: transcriptome analysis and reversibility with glucocorticoids

    J Allergy Clin Immunol

    (2007)
  • A.P. South et al.

    Human epidermal differentiation complex in a single 2.5 Mbp long continuum of overlapping DNA cloned in bacteria integrating physical and transcript maps

    J Invest Dermatol

    (1999)
  • S. Weidinger et al.

    Loss-of-function variations within the filaggrin gene predispose for atopic dermatitis with allergic sensitizations

    J Allergy Clin Immunol

    (2006)
  • G.M. O’Regan et al.

    Filaggrin in atopic dermatitis

    J Allergy Clin Immunol

    (2009)
  • C. Blanchard et al.

    Periostin facilitates eosinophil tissue infiltration in allergic lung and esophageal responses

    Mucosal Immunol

    (2008)
  • S.S. Aceves et al.

    Esophageal remodeling in pediatric eosinophilic esophagitis

    J Allergy Clin Immunol

    (2007)
  • T. Maruhashi et al.

    Interaction between periostin and BMP-1 promotes proteolytic activation of lysyl oxidase

    J Biol Chem

    (2010)
  • S.C. Chae et al.

    Analysis of the polymorphisms in eotaxin gene family and their association with asthma, IgE, and eosinophil

    Biochem Biophys Res Commun

    (2004)
  • T. Ueda et al.

    TGFB1 promoter polymorphism C-509T and pathophysiology of asthma

    J Allergy Clin Immunol

    (2008)
  • S.F. Ziegler

    The role of thymic stromal lymphopoietin (TSLP) in allergic disorders

    Curr Opin Immunol

    (2010)
  • M. Mao et al.

    T lymphocyte activation gene identification by coregulated expression on DNA microarrays

    Genomics

    (2004)
  • S.M. Ho

    Environmental epigenetics of asthma: an update

    J Allergy Clin Immunol

    (2010)
  • E.J. Lim et al.

    Epigenetic regulation of the IL-13-induced human eotaxin-3 gene by CBP-mediated histone 3 acetylation

    J Biol Chem

    (2011)
  • C.A. Liacouras et al.

    Eosinophilic esophagitis: updated consensus recommendations for children and adults

    J Allergy Clin Immunol

    (2011)
  • R.J. Noel et al.

    Eosinophilic esophagitis

    N Engl J Med

    (2004)
  • R.J. Noel et al.

    Eosinophilic esophagitis

    Curr Opin Pediatr

    (2005)
  • D. Atkins et al.

    Eosinophilic esophagitis: the newest esophageal inflammatory disease

    Nat Rev Gastroenterol Hepatol

    (2009)
  • M. Vicario et al.

    Local B cells and IgE production in the oesophageal mucosa in eosinophilic oesophagitis

    Gut

    (2010)
  • A.J. Lucendo et al.

    Treatment with topical steroids downregulates IL-5, eotaxin-1/CCL11, and eotaxin-3/CCL26 gene expression in eosinophilic esophagitis

    Am J Gastroenterol

    (2008)
  • J. Fuentebella et al.

    Increased number of regulatory T cells in children with eosinophilic esophagitis

    J Pediatr Gastroenterol Nutr

    (2010)
  • S.S. Aceves et al.

    Mast cells infiltrate the esophageal smooth muscle in patients with eosinophilic esophagitis, express TGF-beta1, and increase esophageal smooth muscle contraction

    J Allergy Clin Immunol

    (2010)
  • S.S. Aceves et al.

    Resolution of remodeling in eosinophilic esophagitis correlates with epithelial response to topical corticosteroids

    Allergy

    (2010)
  • J.M. Spergel et al.

    14 years of eosinophilic esophagitis: clinical features and prognosis

    J Pediatr Gastroenterol Nutr

    (2009)
  • M. Chehade et al.

    Food allergy and eosinophilic esophagitis

    Curr Opin Allergy Clin Immunol

    (2010)
  • T.F. Brown-Whitehorn et al.

    The link between allergies and eosinophilic esophagitis: implications for management strategies

    Expert Rev Clin Immunol

    (2010)
  • J. Ronkainen et al.

    Prevalence of oesophageal eosinophils and eosinophilic oesophagitis in adults: the population-based Kalixanda study

    Gut

    (2007)
  • J.M. Spergel et al.

    Variation in prevalence, diagnostic criteria, and initial management options for eosinophilic gastrointestinal diseases in the United States

    J Pediatr Gastroenterol Nutr

    (2011)
  • C. Blanchard et al.

    Eotaxin-3 and a uniquely conserved gene-expression profile in eosinophilic esophagitis

    J Clin Invest

    (2006)
  • Cited by (111)

    • Epidemiologic and Clinical Clues to the Etiology of Eosinophilic Esophagitis

      2024, Immunology and Allergy Clinics of North America
    • Eosinophilic esophagitis

      2024, Dysphagia: A Clinical Guide
    • The Role of the Environment in Eosinophilic Esophagitis

      2021, Journal of Allergy and Clinical Immunology: In Practice
    View all citing articles on Scopus

    Series editors: Joshua A. Boyce, MD, Fred Finkelman, MD, William T. Shearer, MD, PhD, and Donata Vercelli, MD

    Supported in part by National Institutes of Health (NIH) grants 2U19 AI066738, U19 AI070235, R01 DK076893, and R37 AI1045898; PHS Grant P30 DK078392; the Department of Defense; the Food Allergy Project; the Buckeye Foundation; and the Campaign Urging Research for Eosinophilic Disease (CURED) Foundation. J.D.S. is supported by a T32 NIH training grant (HL091805).

    Terms in boldface and italics are defined in the glossary on page 24.

    View full text