Abstract
Eosinophilic esophagitis (EoE) is a chronic and progressive immune-mediated disease of the esophagus associated with antigen-driven type 2 inflammation and symptoms of esophageal dysfunction. Our understanding of EoE pathophysiology has evolved since its initial recognition more than 20 years ago and has translated into diagnostic and novel therapeutic approaches that are affecting patient care. The mechanisms underlying disease development and progression are influenced by diverse factors, such as genetics, age, allergic comorbidities, and allergen exposures. Central to EoE pathophysiology is a dysregulated feed-forward cycle that develops between the esophageal epithelium and the immune system. Allergen-induced, type 2-biased immune activation by the esophageal epithelium propagates a cycle of impaired mucosal barrier integrity and allergic inflammation, eventually leading to tissue remodeling and progressive organ dysfunction. Herein, we review the current understanding of fundamental pathophysiological mechanisms contributing to EoE pathogenesis.
Introduction
Eosinophilic esophagitis (EoE) is a chronic, progressive immunemediated disease of the esophagus associated with antigen-driven type 2 inflammation and symptoms of esophageal dysfunction.1 EoE is a common cause of feeding dysfunction in the pediatric population2,3 and manifests clinically with gastrointestinal symptoms that often vary based on a patient’s age. Younger patients with EoE often manifest with vomiting, heartburn, abdominal pain, feeding intolerance, and/or failure to thrive, whereas adolescents and adults most often present with progressive esophageal dysfunction, including dysphagia and food impactions.4–8 Individuals with EoE frequently report adverse symptoms related to food ingestion and have multiple immunoglobulin (Ig)E food sensitizations,9 which can result in numerous dietary restrictions. The chronic gastrointestinal symptoms, feeding dysfunction, and restricted diets reduce patient quality of life, with EoE ranking among the lowest quality of life scores in pediatric patients with chronic disease.10
Characteristic EoE endoscopic features include linear furrows, esophageal rings, white plaques, exudates, and esophageal narrowing. However, endoscopic abnormalities can be subtle, and up to 30% of children with active disease can have an endoscopically normal-appearing esophagus.11 Furthermore, the symptoms and active eosinophilic inflammation can be discordant.12,13 Consequently, the reference standard for EoE diagnosis and disease surveillance remains histopathologic evaluation of esophageal mucosal biopsy specimens for increased intraepithelial eosinophils, with active disease defined by a tissue eosinophil density greater than or equal to 15 eosinophils/high-power field (HPF).1,14 Additional histopathologic features in the esophageal mucosa associated with active disease are being increasingly incorporated into histologic evaluations of esophageal tissue biopsy specimens for EoE, including epithelial cell morphology changes, basal zone hyperplasia (BZH), dilated intercellular spaces (DIS), and lamina propria (LP) fibrosis.15,16
Since the recognition of EoE as a distinct clinical disease in the early 1990s,17,18 our understanding of the disease pathophysiology has advanced significantly. Mechanisms underlying disease development and progression are influenced by diverse factors, such as genetics, age, allergic comorbidities, and allergen exposures. Central to EoE pathophysiology is a dysregulated feed-forward cycle that develops between an abnormal esophageal epithelium and the immune system (Fig 1). The esophageal epithelium with impaired barrier function stimulates allergen-induced, type 2-biased immune activation. This allergic inflammation promotes further impairment of mucosal integrity and propagates chronic immune activation, tissue remodeling, and progressive organ dysfunction. Herein, we breakdown the complex pathophysiological mechanisms contributing to EoE pathogenesis.
Figure 1.
EoE is an allergen-driven, chronic inflammatory disease of the esophagus. Impaired epithelial barrier function and dysregulated immune cell responses influenced by genetic polymorphisms are central to EoE pathogenesis, generating a feed-forward cycle leading to loss of immunologic tolerance to exogenous allergens and chronic eosinophilic inflammation. Created with BioRender.com. EoE, eosinophilic esophagitis; HPF, high-power field; IL, interleukin; TH2, T helper 2 cells; TSLP, thymic stromal lymphopoietin.
Genetic Contributions to Eosinophilic Esophagitis Pathogenesis
EoE pathophysiology is driven by an interplay between genetic, environmental, and immunologic components. A recent comprehensive review focused on the contributions of genetics to the etiology of EoE19; we refer readers to this article for more detailed discussions. A relatively strong genetic contribution to EoE is supported given the increased EoE frequency among first-degree family members.20,21 The familial inheritance pattern of EoE is inconsistent with a Mendelian/monogenic disorder, with the exception of rare families having single-gene mutations that drive disease transmission patterns. Most frequently, EoE has a complex inheritance pattern that is influenced by the effects of multiple genetic risk loci and the host environment. Notably, disease concordance among siblings is nearly 10-fold higher among dizygotic twins than in nontwin siblings (22% vs 2.4%, respectively),21 suggesting that shared early life environmental factors substantially influence risk of disease development. Indeed, Jensen et al22 evaluated the role of early life factors and determined that prenatal (maternal fever, preterm labor), intrapartum (cesarean section), and early life medical treatments (antibiotics, acid suppressants) are associated with an increased risk of developing EoE.
EoE is a rare disease (prevalence of approximately 0.5–1 case per 1000 individuals23), resulting in genetic studies that have been relatively limited to small samples sizes. However, 5 independent genome-wide association studies (GWAS) completed with subjects with EoE have identified numerous susceptibility loci with genome-wide significance (Table 1).24–28 Four genetic loci (5q22 [TSLP/WDR36], 2p23 [CAPN14], 11q13 [LRRC32/EMSY], and 16p13 [CLEC16A/DEX1]) have been reproduced across multiple studies, with a number of other risk loci identified in single studies. Candidate gene–phenotype association studies have identified additional genetic loci that are also associated with the disease (Table 2).24,29–37 Collectively, many of the genes identified in these different analyses seem to influence epithelial barrier function or TH2-mediated immune responses, consistent with the underlying pathoetiology of EoE. Furthermore, most EoE genetic risk variants identified are located outside of gene-coding regions, suggesting a key role for genotype-dependent gene regulation in EoE.19
Table 1.
Statistically Significant EoE Genetic Risk Loci Identified by GWAS
EoE Risk Locus (chromosome location) | Candidate genes at or near risk variants | Known function/putative mechanism in EoE pathogenesis | References |
---|---|---|---|
| |||
Established risk loci using independent cohorts | |||
2p23.1 | CAPN14 | CAPN14 is an intracellular calcium-activated protease specifically expressed in the esophagus. CAPN14 expression is increased during active EoE and is upregulated by IL-13. Overexpression and silencing of CAPN14 in cultured esophageal cells have a disruptive effect on epithelial barrier function. | Kottyan et al,25 2021 Chang et al,26 2022 Kottyan et al,27 2014 Sleiman et al,28 2014 |
5q22.1 |
TSLP
WDR36 |
TSLP is an epithelium-derived alarmin cytokine that induces allergic Th2 immune responses and has direct effects on multiple allergic effector cells, including eosinophils, mast cells, and basophils. | Rothenberg et al,24 2010 Kottyan et al,25 2021 Chang et al,26 2022 Kottyan et al,27 2014 Sleiman et al,28 2014 |
11q13.5 |
LRRC32
c11orf30/EMSY |
LRRC32 encodes a TGF-β-binding protein that is involved in regulatory T-cell-mediated suppression of colitis. LRRC32 is upregulated by IL-13. EMSY is involved in epithelial cell differentiation. Both LRRC32 and EMSY are expressed in esophageal epithelial cells. | Kottyan et al,25 2021 Chang et al,26 2022 Sleiman et al,28 2014 |
16pl3.13 |
CLEC16A
DEX1 |
CLEC16A and DEX1 are expressed in esophageal epithelial cells. Many cells of the immune system also express CLEC16A. Expression of CLEC16A is upregulated by IL-13. The function of CLEC16A and DEX1 proteins in EoE remains unclear. | Kottyan et al,25 2021 Chang et al,26 2022 Kottyan et al,176 2019 |
Suggestive risk loci identified by single GWAS | |||
8p23.1 | XKR6 | Unknown | Kottyan et al,27 2014 |
15ql3.3 | between LOC283710 and KLF13 | Unknown | |
12q13.3 | STAT6 | STAT6 is a transcription factor activated by IL-4 and IL-13 and has a role in the development of TH2 cells and promotes type 2 immune responses. STAT6 also induces CAPNI4 expression. | Sleiman et al,28 2014 |
19q13.11 | ANKRD27 | ANKRD27 inhibits the activity of the SNARE complex, which could impact vesical trafficking and wound healing. May have a role in mucosal barrier integrity. | |
10p14 | ITIH5 | ITIH5 is a serine protease inhibitor from a class of protease inhibitors that have been implicated in mucosal protease dysregulation and decreased epithelial barrier function in the esophagus. | Kottyan et al,25 2021 |
2q12.1 | TMEM182 | Unknown | Chang et al,26 2022 |
5q31.1 | RAD50 | Unknown | |
6p22.3 | SOX4 | Unknown; involved in immune-associated pathways. | |
8q22.1 | MATN2 | Involved in inflammatory responses, macrophage M2 polarization, and T cell differentiation. | |
10q21.1 | PRKG1 | PRKG1 is involved in inflammatory signaling pathways. | |
11p15.4 | RHOG | Ras homolog gene family, member G (Rho G) is a member of the Rac subfamily of Rho GTPases that are highly expressed in lymphocytes. | |
11q13.4 | SHANK2 | Unknown | |
13q12.13 | GPR12 | Unknown | |
15q22.2 | RORA | Unknown; involved in immune-associated signaling pathways. | |
15q23 | SMAD3 | Unknown; involved in immune-associated signaling pathways. | |
18q12.2 | GALNTI | Unknown |
Abbreviations: CAPN14, calpain 14; EoE, eosinophilic esophagitis; GWAS, genome-wide association study; ITIH5, inter-alpha-trypsin inhibitor heavy chain 5; PRKG1, protein kinase CGMP-dependent 1; TH2, T helper 2 cell; TGF-β, transforming growth factor beta; TSLP, thymic stromal lymphopoietin.
Table 2:
Additional EoE Genetic Risk Loci Identified by Phenotype Association
Gene associated with EoE risk | Known function/putative mechanism in EoE pathogenesis | Reference |
---|---|---|
| ||
CCL26 | CCL26 encodes eotaxin-3, which is a chemokine that promotes eosinophil chemotaxis and recruitment into the tissue and is induced by IL-13. | Blanchard et al,30 2006 |
CRLF2 | CRLF2 encodes the TSLP receptor | Sherill et al,177 2010 |
FLG | FLG is important for maintenance of esophageal barrier function. | Blanchard et al,31 2010 |
DSG1 | DSG1 is a major constituent of desmosomes; important for maintenance of esophageal barrier function. | Samuelov et al,32 2013 Sherill et al,33 2014 |
IL5 | IL-5 is a cytokine important for eosinophil development and function, promoting eosinophil maturation, proliferation, activation, and survival. | Namjou et al,29 2014 |
IL13 | IL-13 is a cytokine with central role in EoE pathogenesis, directing eosinophil-predominant inflammatory responses and characteristic histologic changes to the esophageal epithelium associated with barrier dysfunction. | Namjou et al,29 2014 |
STAT3 | STAT3 is a transcription factor involved in signaling pathways for multiple cytokines. Abnormalities in STAT3 signaling can lead to dysregulated responses to IL-6 and possibly IL-5, leading to enhanced type 2 immune responses. | Arora et al,37 2017 |
SPINK5 | SPINK5 is a serine peptidase inhibitor downregulated in EoE. SPINK5 targets serine proteases and inhibits their proteolytic function. Loss leads to unrestricted protease activity, impaired epithelial barrier function and proinflammatory immune responses. | Paluel et al,36 2017 |
DSP | DSP is a member of the plakin protein family and a critical component of desmosome structures in the epithelium; DSP is highly expressed in the esophagus and is important for maintenance of esophageal barrier function. | Shoda et al,35 2021 |
PPL | PPL is a member of the plakin protein family and a critical component of desmosome structures in the epithelium; PPL is highly expressed in the esophagus and is important for maintenance of esophageal barrier function. | Shoda et al,35 2021 |
Abbreviations: DSG1, desmoglein 1; DSP, desmoplakin; EoE, eosinophilic esophagitis; FLG, filaggrin; IL, interleukin; PPL, periplakin; SPINK5, serine peptidase inhibitor kazal type 5; TSLP, thymic stromal lymphopoietin.
Disease Triggers
In the past 2 decades, the incidence and prevalence of EoE have been increasing at rates that outpace increased recognition.23 This suggests that environmental factors, rather than genetic changes, have a critical role in disease pathogenesis. EoE is an antigen-driven disease associated with exposures to food antigens and possibly environmental aeroallergens.38,39
Food allergens (eg, milk, egg, wheat, soy) are well-established disease triggers,40–42 with dietary elimination of specific food allergen triggers or elemental diet therapy resulting in disease remission in most subjects.43 Individuals with IgE-mediated food allergy (IgE-FA) have an increased risk for the development of EoE. Using a large pediatric cohort from a single academic referral center, Hill et al44 reported that children with IgE-FA had an EoE prevalence of approximately 1 case per 20 individuals, nearly 100-fold higher than population-based estimates of 0.5 to 1 case per 1000. Furthermore, 68% of children with EoE had self- and/or parent-reported IgE-FA.44 Pelz et al45 also revealed that physician-diagnosed IgE-FA was common in patients with EoE, with patients with EoE and concurrent IgE-FA having distinct clinical characteristics compared with patients with EoE without IgE-FA. Interestingly, some individuals with IgE-FA may have a propensity to develop esophageal eosinophilia. Endoscopy results of patients with a history of anaphylaxis to cow’s milk revealed that 38% of these individuals had esophageal eosinophilia (>15 eosinophils/HPF) at baseline, many of whom did not report chronic gastrointestinal or esophageal symptoms.46 Similarly, endoscopy results on adults with peanut allergy found that 14% of the adult subjects had subclinical esophageal eosinophilia (>15 eosinophils/HPF).47 Notably, individuals undergoing food oral immunotherapy (OIT) for treatment of their IgE-FA can develop EoE, with an incidence of biopsy-proven EoE ranging between 3.2% and 5.4%.48,49 However, this incidence is likely an underrepresentation given that many patients experience gastrointestinal adverse events during OIT, but few actually undergo endoscopic evaluation.50 A small study of adults undergoing peanut OIT revealed that most subjects developed asymptomatic esophageal eosinophilia (6 of 7 subjects [87%] with peak eosinophil counts [PECs] > 5 eosinophils/HPF; 4 of 7 subjects [57%] with PECs > 15 eosinophils/HPF) accompanied by mild endoscopic and other histologic abnormalities during their first year of therapy.51 One subject met clinicopathologic criteria for EoE with symptoms of dysphagia and food impaction, prompting withdrawal from the study early for safety concerns. Notably, the tissue eosinophilia and other histologic abnormalities resolved in most of the remaining subjects (4 of 6 subjects) during the second year of OIT. Larger studies are needed to determine whether individuals with IgE-FA and baseline eosinophilia represent a subpopulation of patients with food allergy with chronic mucosal inflammation and whether OIT-induced EoE is an exacerbation of this chronic disease or a new pathologic development. Together, these data establish food antigens as critical pathologic drivers of EoE and suggest that links between the immunopathogenesis of IgE-FA and EoE are likely.
Aeroallergens have also been implicated as potential EoE triggers and/or disease exacerbators, supported by the use of aeroallergens in EoE animal models,52,53 development of denovo EoE in human subjects during sublingual immunotherapy to aeroallergens for allergic rhinitis,54–56 and seasonal variation of EoE symptoms, food impactions, and esophageal eosinophilia.57–62 Furthermore, retrospective review and case reports have suggested that subcutaneous immunotherapy (SCIT) may be useful for patients with EoE as an adjunctive therapy in patients with comorbid allergic rhinitis and/or asthma,63,64 with another case report revealing efficacy of SCIT as a monotherapy to induce and maintain clinicohistologic remission of EoE.65 Additional studies are necessary to evaluate the safety and efficacy of SCIT as an adjunctive therapy, and possibly even monotherapy, for patients with EoE and comorbid allergic rhinitis and/or allergic asthma and determine how immunomodulation to environmental aeroallergens affects disease pathogenesis.
Esophageal Epithelial Cells and Mucosal Barrier Dysfunction
Epithelium-derived alarmin cytokines, thymic stromal lymphopoietin (TSLP), and interleukin (IL)-33 function as danger signals that are rapidly released into the extracellular milieu in response to tissue damage. These cytokines facilitate TH2 immune responses by skewing developing adaptive responses and have direct effects on multiple allergic effector cells, including eosinophils, mast cells, and basophils.66 GWAS implicated genetic variants in TSLP in EoE susceptibility.24,25,27,28 Indeed, individuals carrying the risk allele for the TSLP variant most associated with EoE had elevated esophageal TSLP RNA expression.24 Moreover, both TSLP and IL33 gene expression were increased in esophageal biopsy specimens from children with EoE,24,30,67–69 and mice genetically deficient in TSLPR or IL-33R/ST2 had attenuated inflammation in experimental EoE-like disease.69,70 Finally, IL-33 protein was markedly increased within the nuclei of basal layer esophageal epithelial cells in patients with active EoE vs controls, with levels normalizing on EoE remission.68 One mechanism by which allergenic proteins may trigger alarmin cytokine production is through a novel RipIL-33 allergen-sensing pathway in esophageal epithelial cells.71 The RIPK1–caspase 8 ripoptosome complex in the epithelial cells functions as an allergen-sensing platform that detects diverse allergic stimuli, resulting in caspase 8-directed activation and release of IL-33 (Fig 2). This pathway may be the first molecular evidence for how food and environmental allergens trigger/promote esophageal mucosal inflammation and a novel cellular pathway for therapeutic inhibition.
Figure 2.
EoE pathophysiology model. Exogenous allergens trigger epithelial-derived cytokine TSLP and IL-33 production, the latter through activating the intracellular allergen sensor RIPK1-caspase-8 ripoptosome. An impaired mucosal barrier from dysregulated endogenous proteases and an abnormal epithelium allow translocation of food antigens to the dendritic cells, which process and present them to the CD4+ T cells. TSLP and IL-33 influence the dendritic cells to mature TH2-biased effector T cells and stimulate ILC2s; both populations secrete cytokines IL-4, IL-5, and IL-13, which recruit and activate mast cells, eosinophils, and basophils. Mast cells and eosinophils propagate allergic inflammation through cytokine and inflammatory mediator production (eg, PGD2, leukotrienes, granule enzymes), leading to immune cell activation and epithelial changes that further impair barrier function. A feed-forward cycle develops, causing chronic inflammation that stimulates tissue remodeling/fibrosis through the cytokine TGF-β, epithelial-mesenchymal transition, and pro- and anti-fibrotic mediator (TSPAN-12, TSP1) modulation. Created with BioRender.com. EoE, eosinophilic esophagitis; IL, interleukin; ILC2, type 2 innate lymphoid cell; PGD2, prostaglandin D2; TGF-β, transforming growth factor beta; TH2, T helper 2 cells; TSLP, thymic stromal lymphopoietin.
The lower layers of human esophageal squamous epithelium comprise actively proliferating cells. As the epithelial cells migrate toward the luminal surface, the cells differentiate, flatten, and establish cell-cell connections to form a barrier that protects the underlying tissue from repeated exposures to exogenous antigens. In a disrupted epithelial barrier, antigens pass through and activate the immune system. In EoE, the epithelial barrier is abnormal. The epithelium has characteristic proliferative responses, with BZH being a well-recognized histologic change associated with active EoE.16 In-depth transcriptional and functional analyses of the esophageal epithelium have revealed profound loss of cell differentiation, with genes involved in differentiation and keratinization being the most down-regulated biological processes in the esophagus-specific transcripts altered during EoE.72,73 Furthermore, junctional proteins (eg, E-cadherin, claudin-1, desmoglein-1) necessary to maintain barrier integrity are significantly down-regulated during EoE.33,35,74,75
Dysregulated epithelial protease activity has also been implicated in EoE mucosal barrier abnormalities. Several GWAS identified calpain 14 (CAPN14), encoding an intracellular calcium-activated protease, as the gene most highly associated with EoE.25–28 CAPN14 is specifically expressed in the esophagus, and overexpression and silencing experiments in cultured esophageal cells had a significant disruptive effect on epithelial barrier function.76,77 Serine protease inhibitors (SERPINs) and serine protease inhibitors, Kazal type (SPINKs) were also among the most dysregulated peptidase families in EoE.72 SPINK7 expression is reduced in EoE, leading to increased proteolytic activity in esophageal epithelial cells that was associated with loss of epithelial differentiation, decreased barrier integrity, and enhanced proinflammatory responses, specifically through TSLP production.34,78
Collectively, these data suggest that allergen-triggered, epithelial-derived alarmin cytokines and impaired epithelial cell barrier function associated with loss of cell differentiation and dysregulated endogenous proteases promote allergic inflammatory responses in the esophageal mucosa. These immune responses, as reviewed subsequently, further impair mucosal integrity to propagate chronic immune activation that leads to progressive tissue dysfunction (Fig 1).
Eosinophilic Esophagitis Immunopathogenesis
Interleukin-13 and Interleukin-4
IL-13 has a central role in EoE pathogenesis, directing eosinophil-predominant inflammatory responses and characteristic histologic changes to the epithelium associated with barrier dysfunction (Fig 2).31,33,79–82 IL-13 is produced in the esophagus by infiltrating immune cells, including eosinophils, T helper 2 (TH2) lymphocytes, and mast cells.83–85 IL-13 is highly up-regulated in human esophageal tissue during EoE, and treating cultured primary esophageal epithelial cells with IL-13 alone induces a gene program that largely overlaps with the esophageal transcriptome from EoE biopsy specimens.80 IL-13 stimulation of cultured human epithelial cells also promotes barrier dysfunction and reduces epithelial cell differentiation in the absence of eosinophils.31,33 Furthermore, IL-13 induces expression of CCL26 (eotaxin-3), which promotes eosinophil chemotaxis and recruitment into the tissue.80 Murine models support a prominent role for IL-13 in disease, as esophageal eosinophilic inflammation is induced directly by intranasal or intratracheal IL-13 administration,79 and IL-13 overexpression drives esophageal eosinophilia, epithelial hyperplasia, and esophageal remodeling (fibrosis, angiogenesis).81 Conversely, IL-13–deficient mice fail to develop esophageal eosinophilia in an aeroallergen-induced EoE model.53 These studies provided the rationale for monoclonal antibodies (mAbs) targeting IL-13 in human clinical trials for EoE, which revealed therapeutic efficacy in reducing EoE histologic and endoscopic signs, but limited efficacy in reducing clinical symptoms.86,87 Collectively, these studies implicate IL-13 as a central immune mediator in the observed immunologic and histologic changes characteristic of active EoE.
IL-4 promotes differentiation of TH2 cells and regulates eosinophil trafficking by inducing eosinophil chemotaxins and eosinophil adhesion molecules.88,89 IL4 expression is also increased in EoE by disease activity.82 Notably, dupilumab, a fully human mAb targeting the IL-4Rα chain, which antagonizes both IL-4 and IL-13 signaling, was found to have efficacy in improving histologic, endoscopic, and symptomatic measures in phase 3 clinical trials. Consequently, the US Food and Drug Administration approved dupilumab in May 2022 as the first therapeutic for treating EoE in adolescents aged 12 years and above and adults.90,91
CCL26 and Interleukin-5
Mechanistic studies have implicated CCL26 (eotaxin-3) and IL-5 as additional immune mediators that affect eosinophil function during EoE. CCL26 is a chemokine that causes eosinophil chemotaxis by CCR3, the CCL26 receptor. Notably, CCL26 is the most highly up-regulated gene (53 fold) in the EoE transcriptome and CCL26 expression significantly correlates with disease activity.30 Furthermore, CCR3-deficient mice are protected from developing experimental EoE. CCL26 production is produced mainly by the esophageal epithelium and induced by IL-13 stimulation.80
IL-5 is an important cytokine in eosinophil development and function, promoting eosinophil maturation, proliferation, activation, and survival. In allergen-induced EoE, disease can be induced in wildtype, but not IL-5–deficient, mice and treating wild-type mice with IL-5–neutralizing antibody blocks both allergen– and IL-13–induced EoE.52,53,92,93 These data supported human trials targeting eosinophils through IL-5 inhibition in individuals with EoE.94–97 Mepolizumab and reslizumab are humanized mAbs that neutralize IL-5 by binding and preventing cytokine-receptor interaction. These biologic medications reduce both blood (>90%) and tissue (55%) eosinophil levels in human EoE.94,96,97 However, neither medication induced complete histologic remission (PECs < 15 eosinophils/HPF) in most individuals, and there was no significant clinical improvement in the symptoms. It remains unclear whether the incomplete histologic response and poor clinical improvement reflect eosinophil persistence in the tissue or evidence that symptoms are driven by other cell populations. Ongoing trials with biologics targeting and more completely depleting tissue eosinophils (eg, benralizumab, lirentelimab) through antibody-dependent cell cytotoxicity may further clarify our understanding of IL-5 and eosinophils’ role in disease pathogenesis and the effectiveness of therapeutic strategies specifically targeting this immune axis.
Eosinophils
Eosinophils are pleotropic cells often residing within the mucosal tissues with known effector functions in allergic inflammation, immunoregulation, and tissue remodeling and repair. Eosinophil infiltration into the esophageal tissue was recognized early in the study of EoE as a fundamental pathologic and diagnostic feature of the disease.17,18 Although not pathognomonic for EoE, intraepithelial eosinophils are absent within the esophageal mucosa of healthy individuals,98 and their migration into the epithelium implies underlying tissue pathology. In human EoE, esophageal eosinophil levels correlate with disease severity by endoscopy,99,100 histopathologic severity (eg, epithelial hyperplasia, DIS),12,30,99,101 and tissue remodeling and fibrosis.102,103
Although esophageal mucosal eosinophils are a disease hallmark, the mechanisms by which eosinophils contribute to pathophysiology are not fully understood. Eosinophils generate and release reactive oxygen species and toxic granule proteins (eg, major basic protein, eosinophil peroxidase, eosinophil cationic protein) that damage the esophageal epithelium and potentially decrease epithelial barrier function. Indeed, degranulating eosinophils and extracellular granule proteins are observed in human EoE.30,94,104 Major basic protein can also increase smooth muscle reactivity through the muscarinic M2 receptors105 and activate mast cells and basophils106 also present in the tissue during EoE.107,108 Esophageal eosinophils from human subjects with EoE also express high levels of TH2 cytokines that sustain and augment allergic inflammatory responses (eg, IL-4, IL-5, IL-13, IL-9) and produce profibrotic factors (eg, transforming growth factor beta [TGF-β]) that promote tissue remodeling and fibrosis.84,109 Eosinophils also generate leukotrienes that contribute to smooth muscle proliferation and hyperresponsiveness, which could promote esophageal dysfunction.
Not all EoE features associate with eosinophilic inflammation, however. In experimental EoE, eosinophil-deficient mice have reduced stricture formation and decreased epithelial BZH and LP thickness but still develop esophageal motility dysfunction.93,110 In humans with EoE, esophageal eosinophilia dissociates from symptoms of esophageal dysfunction and endoscopic abnormalities,12,99,100,111 some of which correlate with tissue mast cell levels.111 Furthermore, individuals with an EoE-like disease have similar clinical features, response to steroid therapy, and strong familial association with EoE but have no tissue eosinophilia by histopathology.112 Interestingly, these individuals had reduced CCL26 and increased esophageal lymphocytic infiltrates compared with individuals with EoE. Finally, EoE endotypes (see subsequent discussion) had variable degrees of histologic, endoscopic, and clinical abnormalities, but their classification was independent of PECs.113
A review by Doyle et al114 proposed a model whereby eosinophils have divergent roles in EoE pathogenesis by disease stage (ie, early vs established disease). Given that eosinophils have homeostatic roles in the gastrointestinal tract, their appearance at early stages of EoE may reflect functions to protect/restore a damaged epithelial barrier. In later stages of disease, these cells may become activated and promote tissue damage, thereby perpetuating inflammation and fibrosis.
Mast Cells
Mast cells are tissue-resident immune cells that have central roles in allergic inflammatory responses. Mast cells are normally present within the esophageal mucosa during homeostasis and predominantly reside in the LP. During active EoE, mast cells infiltrate and expand within the esophageal epithelium, where they become activated and degranulate.83,108,115–117 Clinically, mast cell numbers correlate with patient-reported pain symptoms.111,118 A clinical trial of anti–IL-5 therapy in children with EoE revealed that the severity of reported pain during EoE correlated with esophageal mast cell, not eosinophil, levels.109 A recent study corroborated these findings and suggested that a relationship between mast cells and the molecular expression of esophageal TRPV1, an ion channel serving as a detector of painful stimuli on sensory neurons, may contribute to the mast cell–pain association in EoE.118 Notably, mast cell infiltration into the epithelium persists despite treatment-induced resolution of tissue eosinophilia, and mast cell levels correlate with persistent symptoms and endoscopic (furrowing, rings) and histologic abnormalities (BZH, DIS).83,111,117
Mast cells are equipped with effector mechanisms that contribute to disease pathogenesis. They prominently produce esophageal IL-13,83 cytokines that activate eosinophils (IL-3, IL-5, granulocyte-macrophage colony-stimulating factor), and inflammatory mediators, including histamine, prostaglandins, leukotrienes, and thromboxanes, which increase vascular permeability and smooth muscle contraction.119 In experimental EoE, mast cell infiltration into the esophageal muscle layers associates with smooth muscle hypertrophy, increased contractility, and decreased relaxation responses partially mediated by TGF-β1 expression.110,115,120 Mast cells also promote fibroblast activation and collagen secretion.120,121
Single-cell RNA sequencing (scRNA-seq) analysis of the esophageal mast cell population in human EoE reveals a heterogeneous population with cell subsets associated with distinct spatial compartments that fluctuate by EoE disease status.83 During homeostasis, a resident mast cell population with a quiescent phenotype is present in the LP. During active disease, 2 additional mast cell populations emerge in the intraepithelial compartment; these populations assumed a proinflammatory state and expressed proliferation-associated genes. Notably, one of these populations persists during disease remission, poised to reinitiate inflammation.
Collectively, these data emphasize the importance of mast cells in EoE disease pathogenesis. Future diagnostic testing methods for clinical and outcomes–based research will need to consider this effector cell population when assessing disease activity and remission.
T Lymphocytes
Accumulating evidence suggests that effector TH2 cells producing cytokines IL-4, IL-5, and IL-13 have central roles in EoE pathogenesis. T cells infiltrate the esophageal mucosa during active EoE, and their levels are elevated in individuals with active disease vs inactive disease or normal controls.30,122–124 Although levels of both esophageal CD4+ and CD8+ T cells increase,85,123–125 allergen-induced EoE models in mice revealed that CD4+ T cells were pathogenic but that CD8+ T-cells were dispensable.125
Wen et al85 analyzed human tissue-residing CD3+ T cells using scRNA-seq and revealed a prominent polyclonal memory CD4+ T cell population expressing IL-4, IL-5, and IL-13 that correlated strongly with esophageal tissue eosinophilia. Subsequently, Morgan et al126 used scRNA-seq and TCR sequencing to further characterize the esophageal T cell population during active EoE vs remission and confirmed that a pathogenic memory effector TH2 cell population was enriched in the esophagus of individuals with active EoE. This pathogenic TH2 population expressed distinct gene signatures associated with gene up-regulation of TH2 cytokines and eicosanoid prostaglandin D2 (PGD2) synthesis. Notably, PGD2 signals through the chemoattractant receptor-homologous molecule expressed on TH2 cells (CRTH2) expressed on TH2 CD4+ cells, eosinophils, and basophils and stimulates these cells’ recruitment into the tissue.127,128 Signaling through CRTH2 also stimulates eosinophil activation,128–130 proinflammatory cytokine production in TH2 cells,131 and basophil activation.132
Altogether, these data reveal that pathogenic effector TH2 cells infiltrate the esophagus during active disease and release inflammatory mediators capable of (1) promoting inflammation through the recruitment and activation of eosinophils, basophils, and additional pathogenic TH2 cells and (2) inducing epithelial cell changes that contribute to loss of barrier integrity.
Additional Allergic Effector Cells
Additional allergic effector cells with potential roles in EoE include basophils, type 2 innate lymphoid cells (ILC2s), and dendritic cells (DCs). Basophil levels are increased in the blood and esophageal biopsy specimens of human active EoE.69,107,133 Although the role of basophils in disease pathogenesis remains unclear, a mouse model of EoE-like disease revealed that allergic inflammation depended on TSLP and basophils,69 suggesting a possible TSLP-basophil axis in EoE pathogenesis. ILC2s are tissue-resident immune cells that are activated by the epithelial alarmins IL-33 and TSLP and capable of robust IL-4, IL-5, IL-9, and IL-13 production.134 Doherty et al135 revealed that levels of esophageal ILC2s were increased in individuals with active EoE vs remission or controls and that tissue ILC2 levels correlated strongly with tissue eosinophilia. DCs normally reside in the esophageal epithelium,123,124 but DC levels increase during EoE.123 TSLP and IL-33 promote DC-mediated TH2 cell polarization,136 suggesting that DCs may be involved in promoting TH2 cell accumulation and polarization in the esophageal mucosa. Additional research will be necessary to better understand the contributions of these allergic effector cells to the pathogenesis of EoE.
Immunoglobulins (Immunoglobulin E, Immunoglobulin G4)
EoE is associated with other atopic diseases with known IgE-mediated pathophysiology, including food allergy, allergic rhinitis, and asthma.39 Elevated total serum IgE level and specific IgE sensitization to food or environmental allergens are common in individuals with EoE.5,9,137,138 Moreover, IgE production in the esophageal mucosa has been observed in children with EoE.139 However, human studies treating EoE with omalizumab, a humanized anti-IgE mAb, have not supported a significant role for IgE in disease pathogenesis. In one non–placebo-controlled study, anti-IgE therapy significantly reduced esophageal tissue IgE levels but was only marginally effective at inducing remission (33% of subjects).140 In a double-blind, placebo-controlled study, omalizumab did not differ from placebo in inducing histologic remission or symptom improvement.141 Collectively, these data suggest that IgE sensitization associates with EoE but is not directly involved in pathogenesis.
Interestingly, several lines of evidence implicate the antibody subclass IgG4 in EoE pathogenesis. Adults with EoE had a 45-fold increase in esophageal IgG4 levels vs controls, with no significant increase in other IgG subclasses.141 IgG4 deposits were noted in the esophageal tissue in 76% of adults with EoE vs 0% of controls.142 Furthermore, adults with EoE had elevated levels of serum total IgG4 and food-specific IgG4 vs controls.143 Children with active EoE had increased esophageal levels of IgG4-positive plasma cells,144 and elevated esophageal IgG4 levels correlated with disease activity as assessed by histopathology and transcriptomic features.145 Finally, individuals with EoE who were treated with topical steroids or responsive to 6-food elimination diet therapy had reduced serum and esophageal IgG4 levels.143,146 Additional studies are necessary to clarify the specific role of IgG4 in disease pathogenesis.
Interferons (Interferon Alfa, Interferon Gamma)
As noted previously, studies of EoE immunopathology strongly support a critical role for type 2 inflammatory networks. However, levels of type 1 inflammatory signals, including IL-1 and IL-6, and type I and II interferons (IFNs) are elevated in the esophagus of individuals with EoE.82,147,148 Ruffner et al147 found a strong IFN-responsive gene signature (IFN-α– and IFN-γ–responsive genes) in transcriptomic analyses of esophageal biopsy tissue from both adult and pediatric patients with EoE and revealed that blood CD4+ T cells from children with EoE produce IFN-γ on activation with EoE-causal allergens. Previous studies have also detected IFN-γ expression in CD8+ T cells that are enriched in EoE biopsy tissues,85,148 suggesting that they could also contribute to the increased IFN-responsive gene signature in EoE esophageal tissue. Together, these data highlight a potential role for non–type 2 inflammatory networks in EoE immunopathology, but additional studies using animal models and human tissue are needed to clarify the cellular participants in the IFN response and the role that these cytokines may play in pathogenesis.
Tissue Fibrosis and Remodeling and Transforming Growth Factor Beta
Natural history studies suggest that EoE progresses from an early eosinophil-rich, inflammatory disease to a fibrostenotic disease associated with subepithelial collagen deposition, smooth muscle hypertrophy, and angiogenesis.4,23 Indeed, endoscopic features of fibrostenotic disease and histopathologic fibrosis increase with age, which is associated clinically with worsening dysphagia, stricture formation, and food impactions.4,6,7,149 However, tissue remodeling is variable; not all adults with longstanding inflammation develop fibrostenotic disease, whereas some children with EoE develop tissue fibrosis at a young age.5,7,150 Consequently, which individuals will develop fibrostenosis remains unclear, although a fibrostenotic endoscopic phenotype has been associated with a distinct EoE endotype by esophageal gene expression.4,113
TGF-β promotes esophageal remodeling by inducing fibroblast activation and secretion of extracellular matrix (ECM) proteins (eg, collagen, fibronectin),151–153 including smooth muscle proliferation, hyperplasia, and contractility.154 TGF-β level is increased in EoE esophageal biopsy specimens and produced by infiltrating eosinophils and mast cells.103,115 TGF-β also promotes epithelial-mesenchymal transition (EMT), during which epithelial cells acquire myofibroblast characteristics and lose certain epithelial cell features,155,156 allowing these cells to participate in ECM synthesis and deposition. Treatment of adult patients with EoE using an anti–IL-13 antibody significantly reduces EMT markers in the esophageal tissue, suggesting that targeting this immune pathway may reduce the fibrotic response in patients with active EoE.157 In a retrospective study in children with EoE, EMT markers correlated strongly with eosinophil counts and were reversible after treatment.156
Recent studies have begun to shed light on some of the molecular components involved in EoE fibrostenosis. Shoda et al158 used a set of 94 esophageal mRNAs dysregulated in EoE to compare children and adults with fibrostenotic vs nonfibrostenotic EoE phenotypes and associated loss of esophageal tetraspanin 12 (TSPAN12) expression in endothelial cells with EoE tissue fibrostenosis. Notably, IL-13 reduces TSPAN12 expression, which promotes endothelial production of profibrotic mediators (eg, endothelin-1) capable of increasing ECM production by fibroblasts. A proteomic study revealed that fibroblasts from human EoE secrete a unique ECM proteome, identifying thrombospondin-1 (TSP-1) expression specifically in the EoE ECM.159 Notably, TSP-1 was capable of inducing fibroblast collagen I production,159 suggesting a profibrotic role in EoE.
Phenotypic Variability and Eosinophilic Esophagitis Endotypes
EoE is increasingly recognized as a heterogeneous disease with phenotypic variability in clinical presentation by age, race, sex, symptoms, endoscopic and histologic abnormalities, comorbidities, disease triggers, and treatment response.160–162 Males have a higher risk for active disease, food impactions, and strictures, and African Americans exhibit classic EoE endoscopic findings but are less likely to present with dysphagia.163–167 Although most individuals with EoE have atopy,39,168 a subset of patients have no allergic comorbidities, and several non-atopic diseases associate with EoE, including inflammatory bowel disease and connective tissue disorders.169,170 Furthermore, differential responses to therapy have been recognized (eg, response to proton pump inhibitors, swallowed steroids, or dupilumab).90,171
Greater understanding of the disease pathogenesis and clinical heterogeneity has driven efforts to begin characterizing EoE cases by the distinct molecular mechanisms driving disease development and progression (ie, endotypes). Using a machine-learning approach to evaluate histologic, endoscopic, and molecular disease features, Shoda et al113 identified 3 discrete EoE endotypes. EoE endotype 1 (EoEe1) is associated with a normal-appearing esophagus, relatively mild histologic and molecular changes, and steroid responsiveness. EoE endotype 2 (EoEe2) is associated with pediatric onset, the highest degree of endoscopic and histologic severity for inflammation, highest expression of inflammatory cytokines (eg, IL-4, TSLP), and steroid-refractory disease. EoE endotype 3 (EoEe3) is associated with adult onset, the highest degree of endoscopic and histologic severity for fibrostenotic components, and lowest expression of epithelial differentiation genes. Using unsupervised clustering of transcriptional responses from tissue biopsy specimens, Dunn et al172 found heterogeneity in genes associated with type 2 immunity among EoEe1-e3, suggesting that these endotypes could be further subdivided. Notably, none of the endotypes were differentiated by tissue eosinophil levels, reaffirming that the underlying differences in pathophysiology extend beyond eosinophil-directed immunopathogenesis. Longitudinal studies are necessary to determine whether these endotypes associate with distinct disease mechanisms or represent a disease continuum. Aligning specific molecular signatures to individuals with distinct clinical phenotypes and longitudinal outcomes may eventually enable physicians to begin targeting specific effector pathways to individualize therapeutic approaches for EoE.
Conclusion
EoE is a chronic inflammatory disease of the esophagus associated with clinical and molecular heterogeneity and characterized by epithelial barrier defects, eosinophilic TH2-predominant inflammation, and tissue remodeling leading to progressive esophageal dysfunction (Fig 2). Dysregulated epithelial and immune cell responses are central to disease pathogenesis and generate a feed-forward cycle leading to chronic inflammation. Although research efforts in the past 2 decades have drastically improved understanding of EoE, many knowledge gaps remain, including the precise steps underlying development of EoE and how the different elements of pathogenesis influence one another and the natural history of disease. Additional basic and clinical research is necessary to develop methods that efficiently identify allergen exposures that are clinically relevant in individual patients, further understand the underlying mechanisms by which food and possibly aeroallergens are driving disease pathogenesis, and determine the role of the gastrointestinal microbiome in establishing, furthering, and/or mitigating EoE pathophysiology, as Benitez et al173 revealed that the microbiome was altered by the presence and activity of EoE (microbiota in EoE further reviewed in Busing et al174 and Mennini et al175). Prospective studies will be needed to further define how specific therapeutic interventions modulate disease pathogenesis over time and determine which molecular components drive the disease in specific individuals. Ultimately, an improved understanding of EoE pathogenesis will lead to identification of molecular pathways important for the disease that will improve biomarker development, assignment of disease endotypes, and novel treatments for the disease.
Key Messages.
Eosinophilic esophagitis (EoE) is a chronic inflammatory disease of the esophagus associated with clinical and molecular heterogeneity.
EoE pathophysiology is characterized by epithelial barrier defects, eosinophilic TH2-predominant inflammation, and tissue remodeling leading to progressive esophageal dysfunction.
Dysregulated epithelial and immune cell responses are central to disease pathogenesis and generate a feed-forward cycle leading to chronic inflammation.
Improved understanding of EoE pathogenesis has led to identification of disease pathways that have identified novel pathways for disease treatment.
Acknowledgment
The authors thank Shawna Hottinger for editorial assistance.
Funding:
This work was supported by the Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR). The CEGIR (U54 AI117804) is part of the Rare Disease Clinical Research Network, an initiative of the Office of Rare Diseases Research and the National Center for Advancing Translational Sciences (NCATS), and is funded through collaboration between the National Institute of Allergy and Infectious Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, and NCATS. The CEGIR is also supported by patient advocacy groups including the American Partnership for Eosinophilic Disorders, Campaign Urging Research for Eosinophilic Diseases, and Eosinophilic Family Coalition. As a member of the Rare Disease Clinical Research Network, the CEGIR is also supported by its Data Management and Coordinating Center (U2CTR002818). Funding support for the Data Management and Coordinating Center is provided by NCATS and the National Institute of Neurological Disorders and Stroke. Dr Troutman was supported in part by PHS Grant P30 DK078392.
Footnotes
Disclosures: Dr Schwartz has consulted for Shire/Takeda and received research funding from Knopp Biosciences.
References
- 1.Dellon ES, Liacouras CA, Molina-Infante J, Furuta GT, Spergel JM, Zevit N, et al. Updated international consensus diagnostic criteria for eosinophilic esophagitis: proceedings of the AGREE conference. Gastroenterology. 2018;155(4):1022–1033. e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mukkada VA, Haas A, Maune NC, Capocelli KE, Henry M, Gilman N, et al. Feeding dysfunction in children with eosinophilic gastrointestinal diseases. Pediatrics. 2010;126(3):e672–e677. [DOI] [PubMed] [Google Scholar]
- 3.Muir AB, Brown-Whitehorn T, Godwin B, Cianferoni A. Eosinophilic esophagitis: early diagnosis is the key. Clin Exp Gastroenterol. 2019;12:391–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Dellon ES, Kim HP, Sperry SLW, Rybnicek DA, Woosley JT, Shaheen NJ. A phenotypic analysis shows that eosinophilic esophagitis is a progressive fibrostenotic disease. Gastrointest Endosc. 2014;79(4):577–585. e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Straumann A, Aceves SS, Blanchard C, Collins MH, Furuta GT, Hirano I, et al. Pediatric and adult eosinophilic esophagitis: similarities and differences. Allergy. 2012;67(4):477–490. [DOI] [PubMed] [Google Scholar]
- 6.Warners MJ, Oude Nijhuis RAB, de Wijkerslooth LRH, Smout AJPM, Bredenoord AJ. The natural course of eosinophilic esophagitis and long-term consequences of undiagnosed disease in a large cohort. Am J Gastroenterol. 2018;113(6):836–844. [DOI] [PubMed] [Google Scholar]
- 7.Schoepfer AM, Safroneeva E, Bussmann C, Kuchen T, Portmann S, Simon H-U, et al. Delay in diagnosis of eosinophilic esophagitis increases risk for stricture formation in a time-dependent manner. Gastroenterology. 2013;145(6):1230–1236. e1-e2. [DOI] [PubMed] [Google Scholar]
- 8.Lucendo AJ, Sánchez-Cazalilla M. Adult versus pediatric eosinophilic esophagitis: important differences and similarities for the clinician to understand. Expert Rev Clin Immunol. 2012;8(8):733–745. [DOI] [PubMed] [Google Scholar]
- 9.Erwin EA, James HR, Gutekunst HM, Russo JM, Kelleher KJ, Platts-Mills TAE. Serum IgE measurement and detection of food allergy in pediatric patients with eosinophilic esophagitis. Ann Allergy Asthma Immunol. 2010;104(6):496–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ingerski LM, Modi AC, Hood KK, Pai AL, Zeller M, Piazza-Waggoner C, et al. Health-related quality of life across pediatric chronic conditions. J Pediatr. 2010;156(4):639–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Liacouras CA, Spergel JM, Ruchelli E, Verma R, Mascarenhas M, Semeao E, et al. Eosinophilic esophagitis: a 10-year experience in 381 children. Clin Gastroenterol Hepatol. 2005;3(12):1198–1206. [DOI] [PubMed] [Google Scholar]
- 12.Pentiuk S, Putnam PE, Collins MH, Rothenberg ME. Dissociation between symptoms and histological severity in pediatric eosinophilic esophagitis. J Pediatr Gastroenterol Nutr. 2009;48(2):152–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Prasad GA, Talley NJ, Romero Y, Arora AS, Kryzer LA, Smyrk TC, et al. Prevalence and predictive factors of eosinophilic esophagitis in patients presenting with dysphagia: a prospective study. Am J Gastroenterol. 2007;102(12):2627–2632. [DOI] [PubMed] [Google Scholar]
- 14.Furuta GT, Liacouras CA, Collins MH, Gupta SK, Justinich C, Putnam PE, et al. Eosinophilic esophagitis in children and adults: a systematic review and consensus recommendations for diagnosis and treatment. Gastroenterology. 2007;133 (4):1342–1363. [DOI] [PubMed] [Google Scholar]
- 15.Ma C, Jairath V, Feagan BG, Guizzetti L, Zou G, McFarlane SC, et al. Responsiveness of a histologic scoring system compared with peak eosinophil count in eosinophilic esophagitis. Am J Gastroenterol. 2022;117(2):264–271. [DOI] [PubMed] [Google Scholar]
- 16.Collins MH, Martin LJ, Alexander ES, Todd Boyd J, Sheridan R, He H, et al. Newly developed and validated eosinophilic esophagitis histology scoring system and evidence that it outperforms peak eosinophil count for disease diagnosis and monitoring. Dis Esophagus. 2017;30(3):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kelly KJ, Lazenby AJ, Rowe PC, Yardley JH, Perman JA, Sampson HA. Eosinophilic esophagitis attributed to gastroesophageal reflux: improvement with an amino acid-based formula. Gastroenterology. 1995;109(5):1503–1512. [DOI] [PubMed] [Google Scholar]
- 18.Attwood SE, Smyrk TC, Demeester TR, Jones JB. Esophageal eosinophilia with dysphagia. A distinct clinicopathologic syndrome. Dig Dis Sci. 1993;38(1):109–116. [DOI] [PubMed] [Google Scholar]
- 19.Kottyan LC, Parameswaran S, Weirauch MT, Rothenberg ME, Martin LJ. The genetic etiology of eosinophilic esophagitis. J Allergy Clin Immunol. 2020;145 (1):9–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Allen-Brady K, Firszt R, Fang JC, Wong J, Smith KR, Peterson KA. Population-based familial aggregation of eosinophilic esophagitis suggests a genetic contribution. J Allergy Clin Immunol. 2017;140(4):1138–1143. [DOI] [PubMed] [Google Scholar]
- 21.Alexander ES, Martin LJ, Collins MH, Kottyan LC, Sucharew H, He H, et al. Twin and family studies reveal strong environmental and weaker genetic cues explaining heritability of eosinophilic esophagitis. J Allergy Clin Immunol. 2014;134 (5):1084–1092. e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Jensen ET, Kuhl JT, Martin LJ, Langefeld CD, Dellon ES, Rothenberg ME. Early-life environmental exposures interact with genetic susceptibility variants in pediatric patients with eosinophilic esophagitis. J Allergy Clin Immunol. 2018;141 (2):632–637. e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Dellon ES, Hirano I. Epidemiology and natural history of eosinophilic esophagitis. Gastroenterology. 2018;154(2):319–332. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Rothenberg ME, Spergel JM, Sherrill JD, Annaiah K, Martin LJ, Cianferoni A, et al. Common variants at 5q22 associate with pediatric eosinophilic esophagitis. Nat Genet. 2010;42(4):289–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kottyan LC, Trimarchi MP, Lu X, Caldwell JM, Maddox A, Parameswaran S, et al. Replication and meta-analyses nominate numerous eosinophilic esophagitis risk genes. J Allergy Clin Immunol. 2021;147(1):255–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chang X, March M, Mentch F, Nguyen K, Glessner J, Qu H, et al. A genome-wide association meta-analysis identifies new eosinophilic esophagitis loci. J Allergy Clin Immunol. 2022;149(3):988–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kottyan LC, Davis BP, Sherrill JD, Liu K, Rochman M, Kaufman K, et al. Genome-wide association analysis of eosinophilic esophagitis provides insight into the tissue specificity of this allergic disease. Nat Genet. 2014;46(8):895–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sleiman PMA, Wang M-L, Cianferoni A, Aceves S, Gonsalves N, Nadeau K, et al. GWAS identifies four novel eosinophilic esophagitis loci. Nat Commun. 2014;5:5593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Namjou B, Marsolo K, Caroll RJ, Denny JC, Ritchie MD, Verma SS, et al. Phenomewide association study (PheWAS) in EMR-linked pediatric cohorts, genetically links PLCL1 to speech language development and IL5-IL13 to eosinophilic esophagitis. Front Genet. 2014;5:401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Blanchard C, Wang N, Stringer KF, Mishra A, Fulkerson PC, Pablo Abonia J, et al. Eotaxin-3 and a uniquely conserved gene-expression profile in eosinophilic esophagitis. J Clin Invest. 2006;116(2):536–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Blanchard C, Stucke EM, Burwinkel K, Caldwell JM, Collins MH, Ahrens A, et al. Coordinate interaction between IL-13 and epithelial differentiation cluster genes in eosinophilic esophagitis. J Immunol. 2010;184(7):4033–4041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Samuelov L, Sarig O, Harmon RM, Rapaport D, Ishida-Yamamoto A, Isakov O, et al. Desmoglein 1 deficiency results in severe dermatitis, multiple allergies and metabolic wasting. Nat Genet. 2013;45(10):1244–1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sherrill JD, Kc K, Wu D, Djukic Z, Caldwell JM, Stucke EM, et al. Desmoglein-1 regulates esophageal epithelial barrier function and immune responses in eosinophilic esophagitis. Mucosal Immunol. 2014;7(3):718–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Azouz NP, Ynga-Durand MA, Caldwell JM, Jain A, Rochman M, Fischesser DM, et al. The antiprotease SPINK7 serves as an inhibitory checkpoint for esophageal epithelial inflammatory responses. Sci Transl Med. 2018;10(444):eaap9736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Shoda T, Kaufman KM, Wen T, Caldwell JM, Osswald GA, Purnima P, et al. Desmoplakin and periplakin genetically and functionally contribute to eosinophilic esophagitis. Nat Commun. 2021;12(1):6795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Paluel-Marmont C, Bellon N, Barbet P, Leclerc-Mercier S, Hadj-Rabia S, Dupont C, et al. Eosinophilic esophagitis and colonic mucosal eosinophilia in Netherton syndrome. J Allergy Clin Immunol. 2017;139(6):2003–2005. e1. [DOI] [PubMed] [Google Scholar]
- 37.Arora M, Bagi P, Strongin A, Heimall J, Zhao X, Lawrence MG, et al. Gastrointestinal manifestations of STAT3-deficient hyper-IgE syndrome. J Clin Immunol. 2017;37(7):695–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cianferoni A, Jensen E, Davis CM. The role of the environment in eosinophilic esophagitis. J Allergy Clin Immunol Pract. 2021;9(9):3268–3274. [DOI] [PubMed] [Google Scholar]
- 39.Capucilli P, Hill DA. Allergic comorbidity in eosinophilic esophagitis: mechanistic relevance and clinical implications. Clin Rev Allergy Immunol. 2019;57(1):111–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kagalwalla AF, Shah A, Li BUK, Sentongo TA, Ritz S, Manuel-Rubio M, et al. Identification of specific foods responsible for inflammation in children with eosinophilic esophagitis successfully treated with empiric elimination diet. J Pediatr Gastroenterol Nutr. 2011;53(2):145–149. [DOI] [PubMed] [Google Scholar]
- 41.Spergel JM, Brown-Whitehorn TF, Cianferoni A, Shuker M, Wang M-L, Verma R, et al. Identification of causative foods in children with eosinophilic esophagitis treated with an elimination diet. J Allergy Clin Immunol. 2012;130(2):461–467. [DOI] [PubMed] [Google Scholar]
- 42.Simon D, Cianferoni A, Spergel JM, Aceves S, Holbreich M, Venter C, et al. Eosinophilic esophagitis is characterized by a non-IgE-mediated food hypersensitivity. Allergy. 2016;71(5):611–620. [DOI] [PubMed] [Google Scholar]
- 43.Davis BP, Rothenberg ME. Emerging concepts of dietary therapy for pediatric and adult eosinophilic esophagitis. Expert Rev Clin Immunol. 2013;9(4):285–287. [DOI] [PubMed] [Google Scholar]
- 44.Hill DA, Dudley JW, Spergel JM. The prevalence of eosinophilic esophagitis in pediatric patients with IgE-mediated food allergy. J Allergy Clin Immunol Pract. 2017;5(2):369–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pelz BJ, Wechsler JB, Amsden K, Johnson K, Singh AM, Wershil BK, et al. IgE-associated food allergy alters the presentation of paediatric eosinophilic esophagitis. Clin Exp Allergy. 2016;46(11):1431–1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Barbosa AC, Castro FM, Meireles PR, Arruda LK, Cardoso SR, Kalil J, et al. Eosinophilic esophagitis: latent disease in patients with anaphylactic reaction to cow’s milk. J Allergy Clin Immunol Pract. 2018;6(2):451–456. e1. [DOI] [PubMed] [Google Scholar]
- 47.Wright BL, Fernandez-Becker NQ, Kambham N, Purington N, Tupa D, Zhang W, et al. Baseline gastrointestinal eosinophilia is common in oral immunotherapy subjects with IgE-mediated peanut allergy. Front Immunol. 2018;9:2624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Cafone J, Capucilli P, Hill DA, Spergel JM. Eosinophilic esophagitis during sublingual and oral allergen immunotherapy. Curr Opin Allergy Clin Immunol. 2019;19 (4):350–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jin H, Trogen B, Nowak-Wegrzyn A. Eosinophilic esophagitis as a complication of food oral immunotherapy. Curr Opin Allergy Clin Immunol. 2020;20(6):616–623. [DOI] [PubMed] [Google Scholar]
- 50.Chu DK, Spergel JM, Vickery BP. Management of eosinophilic esophagitis during oral immunotherapy. J Allergy Clin Immunol Pract. 2021;9(9):3282–3287. [DOI] [PubMed] [Google Scholar]
- 51.Wright BL, Fernandez-Becker NQ, Kambham N, Purington N, Cao S, Tupa D, et al. Gastrointestinal eosinophil responses in a longitudinal, randomized trial of peanut oral immunotherapy. Clin Gastroenterol Hepatol. 2021;19(6):1151–1159. e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mishra A, Hogan SP, Brandt EB, Rothenberg ME. An etiological role for aeroallergens and eosinophils in experimental esophagitis. J Clin Invest. 2001;107(1):83–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Akei HS, Mishra A, Blanchard C, Rothenberg ME. Epicutaneous antigen exposure primes for experimental eosinophilic esophagitis in mice. Gastroenterology. 2005;129(3):985–994. [DOI] [PubMed] [Google Scholar]
- 54.Miehlke S, Alpan O, Schröder S, Straumann A. Induction of eosinophilic esophagitis by sublingual pollen immunotherapy. Case Rep Gastroenterol. 2013;7(3):363–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Antico A, Fante R. Esophageal hypereosinophilia induced by grass sublingual immunotherapy. J Allergy Clin Immunol. 2014;133(5):1482–1484. [DOI] [PubMed] [Google Scholar]
- 56.Béné J, Ley D, Roboubi R, Gottrand F, Gautier S. Eosinophilic esophagitis after desensitization to dust mites with sublingual immunotherapy. Ann Allergy Asthma Immunol. 2016;116(6):583–584. [DOI] [PubMed] [Google Scholar]
- 57.Moawad FJ, Veerappan GR, Lake JM, Maydonovitch CL, Haymore BR, Kosisky SE, et al. Correlation between eosinophilic oesophagitis and aeroallergens. Aliment Pharmacol Ther. 2010;31(4):509–515. [DOI] [PubMed] [Google Scholar]
- 58.Almansa C, Krishna M, Buchner AM, Ghabril MS, Talley N, DeVault KR, et al. Seasonal distribution in newly diagnosed cases of eosinophilic esophagitis in adults. Am J Gastroenterol. 2009;104(4):828–833. [DOI] [PubMed] [Google Scholar]
- 59.Iwanczak B, Janczyk W, Ryzko J, Banaszkiewicz A, Radzikowski A, Jarocka-Cyrta E, et al. Eosinophilic esophagitis in children: frequency, clinical manifestations, endoscopic findings, and seasonal distribution. Adv Med Sci. 2011;56(2):151–157. [DOI] [PubMed] [Google Scholar]
- 60.Prasad GA, Alexander JA, Schleck CD, Zinsmeister AR, Smyrk TC, Elias RM, et al. Epidemiology of eosinophilic esophagitis over three decades in Olmsted County, Minnesota. Clin Gastroenterol Hepatol. 2009;7(10):1055–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Larsson H, Bergquist H, Bove M. The incidence of esophageal bolus impaction: is there a seasonal variation? Otolaryngol Head Neck Surg. 2011;144(2):186–190. [DOI] [PubMed] [Google Scholar]
- 62.Ram G, Lee J, Ott M, Brown-Whitehorn TF, Cianferoni A, Shuker M, et al. Seasonal exacerbation of esophageal eosinophilia in children with eosinophilic esophagitis and allergic rhinitis. Ann Allergy Asthma Immunol. 2015;115(3):224–228. e1. [DOI] [PubMed] [Google Scholar]
- 63.Ramirez RM, Jacobs RL. Eosinophilic esophagitis treated with immunotherapy to dust mites. J Allergy Clin Immunol. 2013;132(2):503–504. [DOI] [PubMed] [Google Scholar]
- 64.Robey BS, Eluri S, Reed CC, Jerath MR, Hernandez ML, Commins SP, et al. Subcutaneous immunotherapy in patients with eosinophilic esophagitis. Ann Allergy Asthma Immunol. 2019;122(5):532–533. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Iglesia EGA, Commins SP, Dellon ES. Complete remission of eosinophilic esophagitis with multi-aeroallergen subcutaneous immunotherapy: a case report. J Allergy Clin Immunol Pract. 2021;9(6):2517–2519. e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hasegawa T, Oka T, Demehri S. Alarmin cytokines as central regulators of cutaneous immunity. Front Immunol. 2022;13: 876515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Judd LM, Heine RG, Menheniott TR, Buzzelli J, O’Brien-Simpson N, Pavlic D, et al. Elevated IL-33 expression is associated with pediatric eosinophilic esophagitis, and exogenous IL-33 promotes eosinophilic esophagitis development in mice. Am J Physiol Gastrointest Liver Physiol. 2016;310(1):G13–G25. [DOI] [PubMed] [Google Scholar]
- 68.Travers J, Rochman M, Caldwell JM, Besse JA, Miracle CE, Rothenberg ME. IL-33 is induced in undifferentiated, non-dividing esophageal epithelial cells in eosinophilic esophagitis. Sci Rep. 2017;7(1):17563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Noti M, Wojno EDT, Kim BS, Siracusa MC, Giacomin PR, Nair MG, et al. Thymic stromal lymphopoietin-elicited basophil responses promote eosinophilic esophagitis. Nat Med. 2013;19(8):1005–1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Venturelli N, Lexmond WS, Ohsaki A, Nurko S, Karasuyama H, Fiebiger E, et al. Allergic skin sensitization promotes eosinophilic esophagitis through the IL-33-basophil axis in mice. J Allergy Clin Immunol. 2016;138(5):1367–1380. e5. [DOI] [PubMed] [Google Scholar]
- 71.Brusilovsky M, Rochman M, Rochman Y, Caldwell JM, Mack LE, Felton JM, et al. Environmental allergens trigger type 2 inflammation through ripoptosome activation. Nat Immunol. 2021;22(10):1316–1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Rochman M, Travers J, Miracle CE, Bedard MC, Wen T, Azouz NP, et al. Profound loss of esophageal tissue differentiation in patients with eosinophilic esophagitis. J Allergy Clin Immunol. 2017;140(3):738–749. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rochman M, Wen T, Kotliar M, Dexheimer PJ, Ben-Baruch Morgenstern N, Caldwell JM, et al. Single-cell RNA sequencing of human esophageal epithelium in homeostasis and allergic inflammation. JCI Insight. 2022;7(11): e159093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Doshi A, Khamishon R, Rawson R, Duong L, Dohil L, Myers SJ, et al. Interleukin 9 alters epithelial barrier and E-cadherin in eosinophilic esophagitis. J Pediatr Gastroenterol Nutr. 2019;68(2):225–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Abdulnour-Nakhoul SM, Al-Tawil Y, Gyftopoulos AA, Brown KL, Hansen M, Butcher KF, et al. Alterations in junctional proteins, inflammatory mediators and extracellular matrix molecules in eosinophilic esophagitis. Clin Immunol. 2013;148(2):265–278. [DOI] [PubMed] [Google Scholar]
- 76.Litosh VA, Rochman M, Rymer JK, Porollo A, Kottyan LC, Rothenberg ME. Calpain-14 and its association with eosinophilic esophagitis. J Allergy Clin Immunol. 2017;139(6):1762–1771. e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Davis BP, Stucke EM, Khorki ME, Litosh VA, Rymer JK, Rochman M, et al. Eosinophilic esophagitis-linked calpain 14 is an IL-13-induced protease that mediates esophageal epithelial barrier impairment. JCI Insight. 2016;1(4):e86355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Azouz NP, Klingler AM, Pathre P, Besse JA, Ben-Baruch-Morgenstern N, Ballaban AY, et al. Functional role of kallikrein 5 and proteinase-activated receptor 2 in eosinophilic esophagitis. Sci Transl Med. 2020;12(545):eaaz7773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Mishra A, Rothenberg ME. Intratracheal IL-13 induces eosinophilic esophagitis by an IL-5, eotaxin-1, and STAT6-dependent mechanism. Gastroenterology. 2003;125(5):1419–1427. [DOI] [PubMed] [Google Scholar]
- 80.Blanchard C, Mingler MK, Vicario M, Abonia JP, Wu YY, Lu TX, et al. IL-13 involvement in eosinophilic esophagitis: transcriptome analysis and reversibility with glucocorticoids. J Allergy Clin Immunol. 2007;120(6):1292–1300. [DOI] [PubMed] [Google Scholar]
- 81.Zuo L, Fulkerson PC, Finkelman FD, Mingler M, Fischetti CA, Blanchard C, et al. IL-13 induces esophageal remodeling and gene expression by an eosinophil-independent, IL-13R alpha 2-inhibited pathway. J Immunol. 2010;185(1):660–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Blanchard C, Stucke EM, Rodriguez-Jimenez B, Burwinkel K, Collins MH, Ahrens A, et al. A striking local esophageal cytokine expression profile in eosinophilic esophagitis. J Allergy Clin Immunol. 2011;127(1):208–217. 217.e1-e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ben-Baruch Morgenstern N, Ballaban AY, Wen T, Shoda T, Caldwell JM, Kliewer K, et al. Single-cell RNA sequencing of mast cells in eosinophilic esophagitis reveals heterogeneity, local proliferation, and activation that persists in remission. J Allergy Clin Immunol. 2022;149(6):2062–2077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Straumann A, Kristl J, Conus S, Vassina E, Spichtin H-P, Beglinger C, et al. Cytokine expression in healthy and inflamed mucosa: probing the role of eosinophils in the digestive tract. Inflamm Bowel Dis. 2005;11(8):720–726. [DOI] [PubMed] [Google Scholar]
- 85.Wen T, Aronow BJ, Rochman Y, Rochman M, Kc K, Dexheimer PJ, et al. Single-cell RNA sequencing identifies inflammatory tissue T cells in eosinophilic esophagitis. J Clin Invest. 2019;129(5):2014–2028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hirano I, Collins MH, Assouline-Dayan Y, Evans L, Gupta S, Schoepfer AM, et al. RPC4046, a monoclonal antibody against IL13, reduces histologic and endoscopic activity in patients with eosinophilic esophagitis. Gastroenterology. 2019;156 (3):592–603. e10. [DOI] [PubMed] [Google Scholar]
- 87.Rothenberg ME, Wen T, Greenberg A, Alpan O, Enav B, Hirano I, et al. Intravenous anti-IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. J Allergy Clin Immunol. 2015;135(2):500–507. [DOI] [PubMed] [Google Scholar]
- 88.Heller NM, Gwinn WM, Donnelly RP, Constant SL, Keegan AD. IL-4 engagement of the type I IL-4 receptor complex enhances mouse eosinophil migration to eotaxin-1 in vitro. PLoS One. 2012;7(6):e39673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Schleimer RP, Sterbinsky SA, Kaiser J, Bickel CA, Klunk DA, Tomioka K, et al. IL-4 induces adherence of human eosinophils and basophils but not neutrophils to endothelium. Association with expression of VCAM-1. J Immunol. 1992;148 (4):1086–1092. [PubMed] [Google Scholar]
- 90.Hirano I, Dellon ES, Hamilton JD, Collins MH, Peterson K, Chehade M, et al. Efficacy of dupilumab in a phase 2 randomized trial of adults with active eosinophilic esophagitis. Gastroenterology. 2020;158(1):111–122. e10. [DOI] [PubMed] [Google Scholar]
- 91.US Food and Drug Administration. FDA approves first treatment for eosinophilic esophagitis, a chronic immune disorder. Available at: https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-eosinophilicesophagitis-chronic-immune-disorder. Accessed August 31, 2022.
- 92.Mishra A, Hogan SP, Brandt EB, Rothenberg ME. IL-5 promotes eosinophil trafficking to the esophagus. J Immunol. 2002;168(5):2464–2469. [DOI] [PubMed] [Google Scholar]
- 93.Mishra A, Wang M, Pemmaraju VR, Collins MH, Fulkerson PC, Abonia JP, et al. Esophageal remodeling develops as a consequence of tissue specific IL-5-induced eosinophilia. Gastroenterology. 2008;134(1):204–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Straumann A, Conus S, Grzonka P, Kita H, Kephart G, Bussmann C, et al. Anti-interleukin-5 antibody treatment (mepolizumab) in active eosinophilic oesophagitis: a randomised, placebo-controlled, double-blind trial. Gut. 2010;59(1):21–30. [DOI] [PubMed] [Google Scholar]
- 95.Stein ML, Collins MH, Villanueva JM, Kushner JP, Putnam PE, Buckmeier BK, et al. Anti-IL-5 (mepolizumab) therapy for eosinophilic esophagitis. J Allergy Clin Immunol. 2006;118(6):1312–1319. [DOI] [PubMed] [Google Scholar]
- 96.Assa’ad AH, Gupta SK, Collins MH, Thomson M, Heath AT, Smith DA, et al. An antibody against IL-5 reduces numbers of esophageal intraepithelial eosinophils in children with eosinophilic esophagitis. Gastroenterology. 2011;141(5):1593–1604. [DOI] [PubMed] [Google Scholar]
- 97.Spergel JM, Rothenberg ME, Collins MH, Furuta GT, Markowitz JE, Fuchs G 3rd, et al. Reslizumab in children and adolescents with eosinophilic esophagitis: results of a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2012;129(2):456–463. 463.e1-e3. [DOI] [PubMed] [Google Scholar]
- 98.DeBrosse CW, Case JW, Putnam PE, Collins MH, Rothenberg ME. Quantity and distribution of eosinophils in the gastrointestinal tract of children. Pediatr Dev Pathol. 2006;9(3):210–218. [DOI] [PubMed] [Google Scholar]
- 99.O’Shea KM, Rochman M, Shoda T, Zimmermann N, Caldwell J, Rothenberg ME. Eosinophilic esophagitis with extremely high esophageal eosinophil counts. J Allergy Clin Immunol. 2021;147(1):409–412. e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wechsler JB, Bolton SM, Amsden K, Wershil BK, Hirano I, Kagalwalla AF. Eosinophilic esophagitis reference score accurately identifies disease activity and treatment effects in children. Clin Gastroenterol Hepatol. 2018;16(7):1056–1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Noel RJ, Putnam PE, Collins MH, Assa’ad AH, Guajardo JR, Jameson SC, et al. Clinical and immunopathologic effects of swallowed fluticasone for eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2004;2(7):568–575. [DOI] [PubMed] [Google Scholar]
- 102.Rajan J, Newbury RO, Anilkumar A, Dohil R, Broide DH, Aceves SS. Long-term assessment of esophageal remodeling in patients with pediatric eosinophilic esophagitis treated with topical corticosteroids. J Allergy Clin Immunol. 2016;137 (1):147–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Aceves SS, Newbury RO, Dohil R, Bastian JF, Broide DH. Esophageal remodeling in pediatric eosinophilic esophagitis. J Allergy Clin Immunol. 2007;119(1):206–212. [DOI] [PubMed] [Google Scholar]
- 104.Mueller S, Aigner T, Neureiter D, Stolte M. Eosinophil infiltration and degranulation in oesophageal mucosa from adult patients with eosinophilic oesophagitis: a retrospective and comparative study on pathological biopsy. J Clin Pathol. 2006;59(11):1175–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Jacoby DB, Gleich GJ, Fryer AD. Human eosinophil major basic protein is an endogenous allosteric antagonist at the inhibitory muscarinic M2 receptor. J Clin Invest. 1993;91(4):1314–1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.O’Donnell MC, Ackerman SJ, Gleich GJ, Thomas LL. Activation of basophil and mast cell histamine release by eosinophil granule major basic protein. J Exp Med. 1983;157(6):1981–1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wen T, Kuhl J, Putnam P, Mukkada V, Farrell M, Kaul A, et al. A flow cytometry-based diagnosis of eosinophilic esophagitis. J Allergy Clin Immunol. 2017;140 (6):1736–1739. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Abonia JP, Blanchard C, Butz BB, Rainey HF, Collins MH, Stringer K, et al. Involvement of mast cells in eosinophilic esophagitis. J Allergy Clin Immunol. 2010;126 (1):140–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Otani IM, Anilkumar AA, Newbury RO, Bhagat M, Beppu LY, Dohil R, et al. Anti-IL-5 therapy reduces mast cell and IL-9 cell numbers in pediatric patients with eosinophilic esophagitis. J Allergy Clin Immunol. 2013;131(6):1576–1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Mavi P, Rajavelu P, Rayapudi M, Paul RJ, Mishra A. Esophageal functional impairments in experimental eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol. 2012;302(11):G1347–G1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Bolton SM, Kagalwalla AF, Arva NC, Wang M-Y, Amsden K, Melin-Aldana H, et al. Mast cell infiltration is associated with persistent symptoms and endoscopic abnormalities despite resolution of eosinophilia in pediatric eosinophilic esophagitis. Am J Gastroenterol. 2020;115(2):224–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Straumann A, Blanchard C, Radonjic-Hoesli S, Bussmann C, Hruz P, Safroneeva E, et al. A new eosinophilic esophagitis (EoE)-like disease without tissue eosinophilia found in EoE families. Allergy. 2016;71(6):889–900. [DOI] [PubMed] [Google Scholar]
- 113.Shoda T, Wen T, Aceves SS, Abonia JP, Atkins D, Bonis PA, et al. Eosinophilic oesophagitis endotype classification by molecular, clinical, and histopathological analyses: a cross-sectional study. Lancet Gastroenterol Hepatol. 2018;3(7):477–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Doyle AD, Masuda MY, Kita H, Wright BL. Eosinophils in eosinophilic esophagitis: the road to fibrostenosis is paved with good intentions. Front Immunol. 2020;11:603295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Aceves SS, Chen D, Newbury RO, Dohil R, Bastian JF, Broide DH. Mast cells infiltrate the esophageal smooth muscle in patients with eosinophilic esophagitis, express TGF-β1, and increase esophageal smooth muscle contraction. J Allergy Clin Immunol. 2010;126(6):1198–1204. e4. [DOI] [PubMed] [Google Scholar]
- 116.Dellon ES, Chen X, Miller CR, Fritchie KJ, Rubinas TC, Woosley JT, et al. Tryptase staining of mast cells may differentiate eosinophilic esophagitis from gastroesophageal reflux disease. Am J Gastroenterol. 2011;106(2):264–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Strasser DS, Seger S, Bussmann C, Pierlot GM, Groenen PMA, Stalder AK, et al. Eosinophilic oesophagitis: relevance of mast cell infiltration. Histopathology. 2018;73(3):454–463. [DOI] [PubMed] [Google Scholar]
- 118.Zhang S, Shoda T, Aceves SS, Arva NC, Chehade M, Collins MH, et al. Mast cell-pain connection in eosinophilic esophagitis. Allergy. 2022;77(6):1895–1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ali Komi D, Wöhrl S, Bielory L. Mast cell biology at molecular level: a comprehensive review. Clin Rev Allergy Immunol. 2020;58(3):342–365. [DOI] [PubMed] [Google Scholar]
- 120.Niranjan R, Mavi P, Rayapudi M, Dynda S, Mishra A. Pathogenic role of mast cells in experimental eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol. 2013;304(12):G1087–G1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Berton A, Levi-Schaffer F, Emonard H, Garbuzenko E, Gillery P, Maquart FX. Activation of fibroblasts in collagen lattices by mast cell extract: a model of fibrosis. Clin Exp Allergy. 2000;30(4):485–492. [DOI] [PubMed] [Google Scholar]
- 122.Straumann A, Bauer M, Fischer B, Blaser K, Simon HU. Idiopathic eosinophilic esophagitis is associated with a T(H)2-type allergic inflammatory response. J Allergy Clin Immunol. 2001;108(6):954–961. [DOI] [PubMed] [Google Scholar]
- 123.Teitelbaum JE, Fox VL, Twarog FJ, Nurko S, Antonioli D, Gleich G, et al. Eosinophilic esophagitis in children: immunopathological analysis and response to fluticasone propionate. Gastroenterology. 2002;122(5):1216–1225. [DOI] [PubMed] [Google Scholar]
- 124.Lucendo AJ, Navarro M, Comas C, Pascual JM, Burgos E, Santamaría L, et al. Immunophenotypic characterization and quantification of the epithelial inflammatory infiltrate in eosinophilic esophagitis through stereology: an analysis of the cellular mechanisms of the disease and the immunologic capacity of the esophagus. Am J Surg Pathol. 2007;31(4):598–606. [DOI] [PubMed] [Google Scholar]
- 125.Mishra A, Schlotman J, Wang M, Rothenberg ME. Critical role for adaptive T cell immunity in experimental eosinophilic esophagitis in mice. J Leukoc Biol. 2007;81(4):916–924. [DOI] [PubMed] [Google Scholar]
- 126.Morgan DM, Ruiter B, Smith NP, Tu AA, Monian B, Stone BE, et al. Clonally expanded, GPR15-expressing pathogenic effector TH2 cells are associated with eosinophilic esophagitis. Sci Immunol. 2021;6(62):eabi5586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Nagata K, Hirai H, Tanaka K, Ogawa K, Aso T, Sugamura K, et al. CRTH2, an orphan receptor of T-helper-2-cells, is expressed on basophils and eosinophils and responds to mast cell-derived factor(s). FEBS Lett. 1999;459(2):195–199. [DOI] [PubMed] [Google Scholar]
- 128.Zhang S, Wu X, Yu S. Prostaglandin D2 receptor D-type prostanoid receptor 2 mediates eosinophil trafficking into the esophagus. Dis Esophagus. 2014;27 (6):601–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Heinemann A, Schuligoi R, Sabroe I, Hartnell A, Peskar BA. Delta 12-prostaglandin J2, a plasma metabolite of prostaglandin D2, causes eosinophil mobilization from the bone marrow and primes eosinophils for chemotaxis. J Immunol. 2003;170 (9):4752–4758. [DOI] [PubMed] [Google Scholar]
- 130.Böhm E, Sturm GJ, Weiglhofer I, Sandig H, Shichijo M, McNamee A, et al. 11-Dehydro-thromboxane B2, a stable thromboxane metabolite, is a full agonist of chemoattractant receptor-homologous molecule expressed on TH2 cells (CRTH2) in human eosinophils and basophils. J Biol Chem. 2004;279(9):7663–7670. [DOI] [PubMed] [Google Scholar]
- 131.Xue L, Gyles SL, Wettey FR, Gazi L, Townsend E, Hunter MG, et al. Prostaglandin D2 causes preferential induction of proinflammatory Th2 cytokine production through an action on chemoattractant receptor-like molecule expressed on Th2 cells. J Immunol. 2005;175(10):6531–6536. [DOI] [PubMed] [Google Scholar]
- 132.Monneret G, Boumiza R, Gravel S, Cossette C, Bienvenu J, Rokach J, et al. Effects of prostaglandin D(2) and 5-lipoxygenase products on the expression of CD203c and CD11b by basophils. J Pharmacol Exp Ther. 2005;312(2):627–634. [DOI] [PubMed] [Google Scholar]
- 133.Siracusa MC, Saenz SA, Hill DA, Kim BS, Headley MB, Doering TA, et al. TSLP promotes interleukin-3-independent basophil haematopoiesis and type 2 inflammation. Nature. 2011;477(7363):229–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Doherty TA. At the bench: understanding group 2 innate lymphoid cells in disease. J Leukoc Biol. 2015;97(3):455–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Doherty TA, Baum R, Newbury RO, Yang T, Dohil R, Aquino M, et al. Group 2 innate lymphocytes (ILC2) are enriched in active eosinophilic esophagitis. J Allergy Clin Immunol. 2015;136(3):792–794. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Paul WE, Zhu J. How are T(H)2-type immune responses initiated and amplified? Nat Rev Immunol. 2010;10(4):225–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Erwin EA, Tripathi A, Ogbogu PU, Commins SP, Slack MA, Cho CB, et al. IgE antibody detection and component analysis in patients with eosinophilic esophagitis. J Allergy Clin Immunol Pract. 2015;3(6):896–904. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Assa’ad AH, Putnam PE, Collins MH, Akers RM, Jameson SC, Kirby CL, et al. Pediatric patients with eosinophilic esophagitis: an 8-year follow-up. J Allergy Clin Immunol. 2007;119(3):731–738. [DOI] [PubMed] [Google Scholar]
- 139.Vicario M, Blanchard C, Stringer KF, Collins MH, Mingler MK, Ahrens A, et al. Local B cells and IgE production in the oesophageal mucosa in eosinophilic oesophagitis. Gut. 2010;59(1):12–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Loizou D, Enav B, Komlodi-Pasztor E, Hider P, Kim-Chang J, Noonan L, et al. A pilot study of omalizumab in eosinophilic esophagitis. PLoS One. 2015;10(3):e0113483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Clayton F, Fang JC, Gleich GJ, Lucendo AJ, Olalla JM, Vinson LA, et al. Eosinophilic esophagitis in adults is associated with IgG4 and not mediated by IgE. Gastroenterology. 2014;147(3):602–609. [DOI] [PubMed] [Google Scholar]
- 142.Zukerberg L, Mahadevan K, Selig M, Deshpande V. Oesophageal intrasquamous IgG4 deposits: an adjunctive marker to distinguish eosinophilic oesophagitis from reflux oesophagitis. Histopathology. 2016;68(7):968–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Wright BL, Kulis M, Guo R, Orgel KA, Wolf WA, Burks AW, et al. Food-specific IgG4 is associated with eosinophilic esophagitis. J Allergy Clin Immunol. 2016;138 (4):1190–1192. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Mohammad N, Avinashi V, Chan E, Vallance BA, Portales-Casamar E, Bush JW. Pediatric eosinophilic esophagitis is associated with increased lamina propria immunoglobulin G4-positive plasma cells. J Pediatr Gastroenterol Nutr. 2018;67 (2):204–209. [DOI] [PubMed] [Google Scholar]
- 145.Rosenberg CE, Mingler MK, Caldwell JM, Collins MH, Fulkerson PC, Morris DW, et al. Esophageal IgG4 levels correlate with histopathologic and transcriptomic features in eosinophilic esophagitis. Allergy. 2018;73(9):1892–1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Weidlich S, Nennstiel S, Jesinghaus M, Brockow K, Slotta-Huspenina J, Bajbouj M, et al. IgG4 is elevated in eosinophilic esophagitis but not in gastroesophageal reflux disease patients. J Clin Gastroenterol. 2020;54(1):43–49. [DOI] [PubMed] [Google Scholar]
- 147.Ruffner MA, Hu A, Dilollo J, Benocek K, Shows D, Gluck M, et al. Conserved IFN signature between adult and pediatric eosinophilic esophagitis. J Immunol. 2021;206(6):1361–1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Sayej WN, Ménoret A, Maharjan AS, Fernandez M, Wang Z, Balarezo F, et al. Characterizing the inflammatory response in esophageal mucosal biopsies in children with eosinophilic esophagitis. Clin Transl Immunol. 2016;5(7):e88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Lucendo AJ, Arias A, De Rezende LC, Yagüe-Compadre JL, Mota-Huertas T, González-Castillo S, et al. Subepithelial collagen deposition, profibrogenic cytokine gene expression, and changes after prolonged fluticasone propionate treatment in adult eosinophilic esophagitis: a prospective study. J Allergy Clin Immunol. 2011;128(5):1037–1046. [DOI] [PubMed] [Google Scholar]
- 150.Bolton SM, Kagalwalla AF, Wechsler JB. Eosinophilic esophagitis in children: endoscopic findings at diagnosis and post-intervention. Curr Gastroenterol Rep. 2018;20(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Rieder F, Nonevski I, Ma J, Ouyang Z, West G, Protheroe C, et al. T-helper 2 cytokines, transforming growth factor β1, and eosinophil products induce fibrogenesis and alter muscle motility in patients with eosinophilic esophagitis. Gastroenterology. 2014;146(5):1266–1277. e1-e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Frangogiannis N Transforming growth factor-β in tissue fibrosis. J Exp Med. 2020;217(3): e20190103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Gomes I, Mathur SK, Espenshade BM, Mori Y, Varga J, Ackerman SJ. Eosinophilfibroblast interactions induce fibroblast IL-6 secretion and extracellular matrix gene expression: implications in fibrogenesis. J Allergy Clin Immunol. 2005;116 (4):796–804. [DOI] [PubMed] [Google Scholar]
- 154.Beppu LY, Anilkumar AA, Newbury RO, Dohil R, Broide DH, Aceves SS. TGF-β1-induced phospholamban expression alters esophageal smooth muscle cell contraction in patients with eosinophilic esophagitis. J Allergy Clin Immunol. 2014;134(5):1100–1107. e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Muir AB, Lim DM, Benitez AJ, Chandramouleeswaran PM, Lee AJ, Ruchelli ED, et al. Esophageal epithelial and mesenchymal cross-talk leads to features of epithelial to mesenchymal transition in vitro. Exp Cell Res. 2013;319(6):850–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Kagalwalla AF, Akhtar N, Woodruff SA, Rea BA, Masterson JC, Mukkada V, et al. Eosinophilic esophagitis: epithelial mesenchymal transition contributes to esophageal remodeling and reverses with treatment. J Allergy Clin Immunol. 2012;129(5):1387–1396. e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Gann PH, Deaton RJ, McMahon N, Collins MH, Dellon ES, Hirano I, et al. An anti-IL-13 antibody reverses epithelial-mesenchymal transition biomarkers in eosinophilic esophagitis: phase 2 trial results. J Allergy Clin Immunol. 2020;146(2):367–376. e3. [DOI] [PubMed] [Google Scholar]
- 158.Shoda T, Wen T, Caldwell JM, Ben-Baruch Morgenstern N, Osswald GA, Rochman M, et al. Loss of endothelial TSPAN12 promotes fibrostenotic eosinophilic esophagitis via endothelial cell-fibroblast crosstalk. Gastroenterology. 2022;162(2):439–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Hsieh LY, Chiang AWT, Duong LD, Kuo C-C, Dong SX, Dohil R, et al. A unique esophageal extracellular matrix proteome alters normal fibroblast function in severe eosinophilic esophagitis. J Allergy Clin Immunol. 2021;148(2):486–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Ruffner MA, Cianferoni A. Phenotypes and endotypes in eosinophilic esophagitis. Ann Allergy Asthma Immunol. 2020;124(3):233–239. [DOI] [PubMed] [Google Scholar]
- 161.Atkins D, Furuta GT, Liacouras CA, Spergel JM. Eosinophilic esophagitis phenotypes: ready for prime time? Pediatr Allergy Immunol. 2017;28(4):312–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Ferguson AE, Mukkada VA, Fulkerson PC. Pediatric eosinophilic esophagitis endotypes: are we closer to predicting treatment response? Clin Rev Allergy Immunol. 2018;55(1):43–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Moawad FJ, Dellon ES, Achem SR, Ljuldjuraj T, Green DJ, Maydonovitch CL, et al. Effects of race and sex on features of eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2016;14(1):23–30. [DOI] [PubMed] [Google Scholar]
- 164.Collins CA, Palmquist J, Proudfoot JA, Qian A, Wangberg H, Khosh-Hemmat E, et al. Evaluation of long-term course in children with eosinophilic esophagitis reveals distinct histologic patterns and clinical characteristics. J Allergy Clin Immunol. 2019;144(4):1050–1057. e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Erwin EA, Jaramillo LM, Smith B, Kruszewski PG, Kahwash B, Grayson MH, et al. Sex differences in blood transcriptional profiles and clinical phenotypes in pediatric patients with eosinophilic esophagitis. J Allergy Clin Immunol Pract. 2021;9 (9):3350–3358. e8. [DOI] [PubMed] [Google Scholar]
- 166.Schreiner P, Safroneeva E, Rossel J-B, Limacher A, Saner C, Greuter T, et al. Sex impacts disease activity but not symptoms or quality of life in adults with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2022;20(8):1729–1738. e1. [DOI] [PubMed] [Google Scholar]
- 167.Sperry SL, Woosley JT, Shaheen NJ, Dellon ES. Influence of race and gender on the presentation of eosinophilic esophagitis. Am J Gastroenterol. 2012;107(2):215–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.González-Cervera J, Arias Á, Redondo-González O, Cano-Mollinedo MM, Terreehorst I, Lucendo AJ. Association between atopic manifestations and eosinophilic esophagitis: a systematic review and meta-analysis. Ann Allergy Asthma Immunol. 2017;118(5):582–590. e2. [DOI] [PubMed] [Google Scholar]
- 169.Capucilli P, Cianferoni A, Grundmeier RW, Spergel JM. Comparison of comorbid diagnoses in children with and without eosinophilic esophagitis in a large population. Ann Allergy Asthma Immunol. 2018;121(6):711–716. [DOI] [PubMed] [Google Scholar]
- 170.Abonia JP, Wen T, Stucke EM, Grotjan T, Griffith MS, Kemme KA, et al. High prevalence of eosinophilic esophagitis in patients with inherited connective tissue disorders. J Allergy Clin Immunol. 2013;132(2):378–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Lucendo AJ, Arias Á, Molina-Infante J. Efficacy of proton pump inhibitor drugs for inducing clinical and histologic remission in patients with symptomatic esophageal eosinophilia: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2016;14(1):13–22. e1. [DOI] [PubMed] [Google Scholar]
- 172.Dunn JLM, Shoda T, Caldwell JM, Wen T, Aceves SS, Collins MH, et al. Esophageal type 2 cytokine expression heterogeneity in eosinophilic esophagitis in a multisite cohort. J Allergy Clin Immunol. 2020;145(6):1629–1640. e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Benitez AJ, Hoffmann C, Muir AB, Dods KK, Spergel JM, Bushman FD, et al. Inflammation-associated microbiota in pediatric eosinophilic esophagitis. Microbiome. 2015;3:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Busing JD, Buendia M, Choksi Y, Hiremath G, Das SR. Microbiome in eosinophilic esophagitis-metagenomic, metatranscriptomic, and metabolomic changes: a systematic review. Front Physiol. 2021;12: 731034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Mennini M, Tambucci R, Riccardi C, Rea F, De Angelis P, Fiocchi A, et al. Eosinophilic esophagitis and microbiota: state of the art. Front Immunol. 2021;12:595762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Kottyan LC, Maddox A, Braxton JR, Stucke EM, Mukkada V, Putnam PE, et al. Genetic variants at the 16p13 locus confer risk for eosinophilic esophagitis. Genes Immun. 2019;20(4):281–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Sherrill JD, Gao PS, Stucke EM, Blanchard C, Collins MH, Putnam PE, et al. Variants of thymic stromal lymphopoietin and its receptor associate with eosinophilic esophagitis. J Allergy Clin Immunol. 2010;126(1):160–165. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]