Reviews and Feature Articles
Eosinophilic gastrointestinal disorders (EGID)

https://doi.org/10.1016/j.jaci.2003.10.047Get rights and content

Abstract

Primary eosinophilic gastrointestinal disorders are defined as disorders that selectively affect the gastrointestinal tract with eosinophil-rich inflammation in the absence of known causes for eosinophilia (eg, drug reactions, parasitic infections, and malignancy). These disorders include eosinophilic esophagitis, eosinophilic gastritis, eosinophilic gastroenteritis, eosinophilic enteritis, and eosinophilic colitis and are occurring with increasing frequency. Significant progress has been made in elucidating that eosinophils are integral members of the gastrointestinal mucosal immune system and that eosinophilic gastrointestinal disorders are primarily polygenic allergic disorders that involve mechanisms that fall between pure IgE-mediated and delayed TH2-type responses. Preclinical studies have identified a contributory role for the cytokine IL-5 and the eotaxin chemokines, providing a rationale for specific disease therapy. An essential question is to determine the cellular and molecular basis for each of these clinical problems and the best treatment regimen, which is the main subject of this review.

Section snippets

Overview of eosinophilic gastrointestinal disorders

Eosinophil accumulation in the gastrointestinal tract is a common feature of numerous gastrointestinal disorders, including classic IgE-mediated food allergy,1, 2 eosinophilic gastroenteritis,3, 4 allergic colitis,5, 6, 7 eosinophilic esophagitis (EE),8, 9, 10 inflammatory bowel disease (IBD),11, 12, 13 and gastroesophageal reflux disease (GERD).14, 15, 16, 17, 18 In IBD eosinophils usually represent only a small percentage of the infiltrating leukocytes,11, 19 but their level has been proposed

Eosinophil growth and development

The eosinophil is formed in the bone marrow, where it spends about 8 days maturing under the regulation of the transcription factors GATA-1, GATA-2, and c/EBP (Fig 2).40, 41, 42 Notably, GATA-1 and GATA-2 overexpression are sufficient signals for promoting eosinophil development in avian, murine, and human systems. Additionally, mice with a targeted genetic deletion in the high-affinity GATA site present in the GATA-1 promoter are selectively deficient in the eosinophil lineage.41 These

Regulation of eosinophil tissue accumulation

Numerous inflammatory mediators have been implicated in regulating eosinophil accumulation, including IL-1, IL-3, IL-4, IL-5, IL-13, and GM-CSF and the chemokines RANTES, monocyte chemoattractant protein (MCP) 3, MCP-4, macrophage inflammatory protein 1α, and eotaxin 1, eotaxin 2, and eotaxin 3.44, 50, 51 IL-3 and GM-CSF, in association with IL-5, enhance eosinophil development, migration, and effector function, whereas IL-1, IL-4, IL-13, and TNF-α regulate eosinophil trafficking by promoting

Eotaxin subfamily of chemokines

Eotaxin was initially discovered by using a biologic assay in guinea pigs designed to identify the molecules responsible for allergen-induced eosinophil accumulation in the lungs.53 A partial amino acid sequence facilitated the genetic cloning of the genes and cDNA for guinea pig, murine, and human eotaxin, which led to the identification of eotaxin as a member of the CC chemokine family most homologous to the MCP subfamily,72, 73, 74, 75, 76 all being present in the same genetic locus (human

Constitutive gastrointestinal eosinophils

Eosinophils have been noted to be present at low levels in numerous tissues. When a large series of biopsy and autopsy specimens were analyzed, the only organs that demonstrated tissue eosinophils (at substantial levels) were the gastrointestinal tract, spleen, lymph nodes, and thymus (Fig 2).110 Interestingly, eosinophil infiltrations were only associated with eosinophil degranulation in the gastrointestinal tract. Examination of eosinophils throughout the gastrointestinal tracts of

Role of baseline eosinophils

The beneficial function of eosinophils has been primarily attributed to their ability to defend the host against parasitic helminths. This is based on several lines of evidence, including (1) the ability of eosinophils to mediate antibody-dependent (or complement-dependent) cellular toxicity against helminths in vitro,111, 112 (2) the observation that eosinophil levels increase during helminth infections and that eosinophils aggregate and degranulate in the local vicinity of damaged parasites

Pro-inflammatory role of eosinophils in EGIDs

Eosinophils are pleiotropic cells stimulated by a variety of triggers (Fig 3). In vitro studies have shown that eosinophil granule constituents are toxic to a variety of tissues, including intestinal epithelium.125 Eosinophil granules contain a crystalloid core composed of major basic protein (MBP) 1 (and MBP-2) and a matrix composed of eosinophil cationic protein (ECP), eosinophil-derived neurotoxin (EDN), and eosinophil peroxidase (EPO; Fig 3).126 These cationic proteins share certain

Clinical evaluation for EGIDs

Patients with EGIDs present with a variety of clinical problems, most commonly failure to thrive, abdominal pain, irritability, gastric dysmotility, vomiting, diarrhea, dysphagia, microcytic anemia, and hypoproteinemia.21 A diagnostic evaluation for EGIDs should be performed on all patients with these refractory problems, especially in individuals with a strong history of allergic diseases, peripheral blood eosinophilia, and/or a family history of EGIDs (Table I). Depending on the intestinal

Evaluation for HES in patients with apparent EGIDs

The term HES was introduced by Anderson and Hardy143 in 1968 to designate patients with marked eosinophilia. They reported 3 patients, all men between the ages of 34 and 47, who had cardiopulmonary symptoms, fever, sweats, weight loss, and marked eosinophilia. Two of the patients died, and at autopsy, their hearts were enlarged and showed mural thrombi. Chusid and associates144 formulated the diagnostic criteria for HES to include (1) persistent eosinophilia of at least 1500 cells/mm2 for a

Eosinophilic esophagitis

The esophagus is normally devoid of eosinophils, and therefore the finding of esophageal eosinophils denotes pathology.9, 10 It is now appreciated that many disorders are accompanied by eosinophil infiltration in the esophagus, such as EE, eosinophilic gastroenteritis, GERD, recurrent vomiting, parasitic and fungal infections, IBD, HES, esophageal leiomyomatosis, myeloproliferative disorders, carcinomatosis, periarteritis, allergic vasculitis, scleroderma, and drug injury.10, 156

Eosinophilic gastritis and gastroenteritis

In contrast to the esophagus, the stomach and intestine have readily detectable baseline eosinophils under healthy conditions. As such, the diagnosis of eosinophilic gastritis, enteritis, and gastroenteritis is even more complex than EE. In this review, eosinophilic gastritis, enteritis, and gastroenteritis are grouped together because they are clinically similar and because there is a paucity of information available concerning their pathogenesis; however, it is likely that they are indeed

Eosinophilic colitis

Eosinophils accumulate in the colons of patients with a variety of disorders, including eosinophilic gastroenteritis, allergic colitis of infancy, infections (including pinworms and dog hookworms), drug reactions, vasculitis (eg, Churg-Strauss syndrome), and IBD.43, 44, 45, 46, 48, 110, 111, 181, 182 Allergic colitis in infancy (also known as dietary protein–induced proctocolitis of infancy syndrome) is the most common cause of bloody stools in the first year of life.183, 184 Similar to other

Summary and concluding remarks

In this article we have reviewed the basic properties of eosinophils, their regulation and role in healthy states, and the pathogenesis and treatment of EGIDs (Fig 4). In brief, under baseline healthy conditions, eosinophils normally account for a small subset of circulating blood cells and primarily reside in the gastrointestinal tract and hematopoietic organs. Their production is tightly regulated by the transcription factors GATA-1, GATA-2, and c/EBP, which instruct the hematopoietic

Acknowledgments

I thank the numerous colleagues who have contributed to the body of information presented in this review, including Drs Simon Hogan, Anil Mishra, Phil Putnam, Amal Assa'ad, Margaret Collins, Eric Brandt, Nives Zimmermann, Paul Foster, Jesus Guajardo, Richard Noel, Mitch Cohen, Glenn Furuta, and Ha Shem. I also thank Dr Troy Scribner for the initial draft of the cover illustration and Andrea Lippelman and Joy Rothenberg.

References (189)

  • KJ Kelly et al.

    Eosinophilic esophagitis attributed to gastroesophageal reflux: improvement with an amino acid-based formula

    Gastroenterology

    (1995)
  • SC Bischoff

    Mucosa allergy: role of mast cells and eosinophil granulocytes in the gut

    Baillieres Clin Gastroenterol

    (1996)
  • HA Sampson

    Food allergy. Part 1: immunopathogenesis and clinical disorders

    J Allergy Clin Immunol

    (1999)
  • PF Weller

    The idiopathic hypereosinophilic syndrome

    Blood

    (1994)
  • AJ Wardlaw

    Molecular basis for selective eosinophil trafficking in asthma: a multistep paradigm

    J Allergy Clin Immunol

    (1999)
  • DS Silberstein

    Eosinophil function in health and disease

    Crit Rev Oncol Hematol

    (1995)
  • BS Bochner

    Road signs guiding leukocytes along the inflammation superhighway

    J Allergy Clin Immunol

    (2000)
  • SK Shaw et al.

    The beta 7 integrins in mucosal homing and retention

    Semin Immunol

    (1995)
  • EC Butcher et al.

    Lymphocyte trafficking and regional immunity

    Adv Immunol

    (1999)
  • WJ Sandborn et al.

    Biologic therapy of inflammatory bowel disease

    Gastroenterology

    (2002)
  • A Mishra et al.

    Enterocyte expression of the eotaxin and interleukin-5 transgenes induces compartmentalized dysregulation of eosinophil trafficking

    J Biol Chem

    (2002)
  • H Floyd et al.

    Siglec-8. A novel eosinophil-specific member of the immunoglobulin superfamily

    J Biol Chem

    (2000)
  • KK Kikly et al.

    Identification of SAF-2, a novel siglec expressed on eosinophils, mast cells, and basophils

    J Allergy Clin Immunol

    (2000)
  • M Kitaura et al.

    Molecular cloning of a novel human CC chemokine (Eotaxin-3) that is a functional ligand of CC chemokine receptor 3

    J Biol Chem

    (1999)
  • H Nomiyama et al.

    Assignment of the human CC chemokine MPIF-2/eotaxin-2 (SCYA24) to chromosome 7q11.23

    Genomics

    (1998)
  • C Combadiere et al.

    Cloning and functional expression of a human eosinophil CC chemokine receptor

    J Biol Chem

    (1995)
  • BO Gerber et al.

    Functional expression of the eotaxin receptor CCR3 in T lymphocytes co-localizing with eosinophils

    Curr Biol

    (1997)
  • P Romagnani et al.

    Tryptase-chymase double-positive human mast cells express the eotaxin receptor CCR3 and are attracted by CCR3-binding chemokines

    Am J Pathol

    (1999)
  • A Moon et al.

    Allergic gastroenteropathy in children

    Ann Allergy Asthma Immunol

    (1995)
  • AM Saavedra-Delgado et al.

    Interactions between food antigens and the immune system in the pathogenesis of gastrointestinal diseases

    Ann Allergy

    (1985)
  • G Torpier et al.

    Eosinophilic gastroenteritis: ultrastructural evidence for a selective release of eosinophil major basic protein

    Clin Exp Immunol

    (1988)
  • SM Hill et al.

    Colitis caused by food allergy in infants

    Arch Dis Child

    (1990)
  • GT Furuta

    Eosinophils in the esophagus: acid is not the only cause

    J Pediatr Gastroenterol Nutr

    (1998)
  • RE Walsh et al.

    The eosinophil in inflammatory bowel disease

    Scand J Gastronenterol

    (1991)
  • SK Sarin et al.

    Significance of eosinophil and mast cell counts in rectal mucosa in ulcerative colitis. A prospective controlled study

    Dig Dis Sci

    (1978)
  • AM Dvorak

    Ultrastructural evidence for release of major basic protein-containing crystalline cores of eosinophil granules in vivo: cytotoxic potential in Crohn's disease

    J Immunol

    (1980)
  • CA Liacouras et al.

    Primary eosinophilic esophagitis in children: successful treatment with oral corticosteroids

    J Pediatr Gastroenterol Nutr

    (1998)
  • LF Brown et al.

    Intraepithelial eosinophils in endoscopic biopsies of adults with reflux esophagitis

    Am J Surg Pathol

    (1984)
  • ME Rothenberg et al.

    Gastrointestinal eosinophils

    Immunol Rev

    (2001)
  • H Nishitani et al.

    Infiltration of peroxidase-producing eosinophils into the lamina propria of patients with ulcerative colitis

    J Gastroenterol

    (1998)
  • S Khan et al.

    Eosinophilic gastroenteritis: epidemiology, diagnosis and management

    Paediatr Drugs

    (2002)
  • JH Caldwell et al.

    Serum IgE to eosinophilic gastroenteritis

    N Engl J Med

    (1975)
  • JP Cello

    Eosinophilic gastroenteritis: a complex disease entity

    Am J Med

    (1979)
  • HH Scudamore et al.

    Food allergy manifested by eosinophilia, elevated immunoglobulin E level, and protein-losing enteropathy: The syndrome of allergic gastroenteropathy

    J Allergy Clin Immunol

    (1982)
  • GT Furuta et al.

    The role of the eosinophil in gastrointestinal diseases

    Curr Opin Gastroenterol

    (1995)
  • G Iacono et al.

    Gastroesophageal reflux and cows milk allergy in infants: a prospective study

    J Allergy Clin Immunol

    (1996)
  • HA Sampson

    Food Allergy

    JAMA

    (1997)
  • SV Walsh et al.

    Allergic esophagitis in children: a clinicopathological entity

    Am J Surg Pathol

    (1999)
  • N Oyaizu et al.

    Eosinophilic gastroenteritis: Immunohistochemical evidence for IgE mast cell-mediated allergy

    Acta Pathol Jpn

    (1985)
  • B Bates

    ‘Explosion’ of eosinophilic esophagitis in children

    Pediatr News

    (2000)
  • Cited by (751)

    • Local type 2 immunity in eosinophilic gastritis

      2023, Journal of Allergy and Clinical Immunology
    View all citing articles on Scopus

    Series editors: William T. Shearer, MD, PhD, Lanny J. Rosenwasser, MD, and Bruce S. Bochner, MD

    This activity is available for CME credit. See page 41A for important information.

    Supported in part by the Burroughs Wellcome Fund, the Human Frontier Science Program RG 264/99, the International Life Science Institute, National Institutes of Health/National Institutes of Allergy and Infectious Disease R01 AI42242 and AI45898, and the kind support of Martin Schlaff.

    Disclosure of potential conflict of interest: M. E. Rothenberg has a consultant arrangement with Cambridge Antibody Technology and receives grants/ research support from Burroughs Wellcome.

    View full text