ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Eosinophilic esophagitis (EoE): Genetics and immunopathogenesis

Eosinophilic esophagitis (EoE): Genetics and immunopathogenesis
Literature review current through: Jan 2024.
This topic last updated: Jan 10, 2023.

INTRODUCTION — Eosinophilic esophagitis (EoE) is a chronic, immune/antigen-mediated esophageal disease characterized clinically by symptoms related to esophageal dysfunction and histologically by eosinophil-predominant inflammation [1]. The pathogenesis of EoE is the result of an interplay among genetic, environmental, and host immune system factors.

This topic reviews the genetics and immunopathogenesis underlying EoE. The clinical features, diagnosis, and management of EoE are discussed in greater detail separately. (See "Clinical manifestations and diagnosis of eosinophilic esophagitis (EoE)" and "Allergy testing in eosinophilic esophagitis" and "Treatment of eosinophilic esophagitis (EoE)" and "Dietary management of eosinophilic esophagitis".)

OVERVIEW OF PATHOGENESIS — The pathogenesis of EoE is incompletely understood but involves genetic, environmental, and host immune system factors. Molecular analysis has elucidated that EoE is caused by a primary defect in esophageal epithelial function rather than an eosinophil defect. This is best illustrated by the finding that genetic susceptibility is mediated by genes expressed by the esophageal epithelia rather than eosinophils. The esophagus of EoE patients has an impairment of epithelial cell differentiation and barrier function.

The esophagus is normally devoid of eosinophils, although they are permanent residents in the rest of the gastrointestinal tract beginning during early embryonic development. Thus, the finding of esophageal eosinophils denotes pathology, typically EoE or gastroesophageal reflux disease (GERD) [2,3]. The diagnosis of EoE originally required the presence of esophageal eosinophilia that was resistant to proton pump inhibitor (PPI) therapy. It is now appreciated that a substantial number of patients with esophageal eosinophilia can respond to PPI therapy, an entity that was referred to as PPI-responsive esophageal eosinophilia (PPI-REE) [4]. It is increasingly appreciated that PPI-REE may be nearly indistinguishable from EoE based upon clinical, endoscopic, histologic, and molecular features [4]. As such, the PPI requirement to diagnosis EoE is no longer a requirement, and PPIs are now considered a therapy for EoE. As such, the diagnosis of EoE is solely based upon the requisite level of eosinophils (at least 15 eosinophils per high-powered field) in patients with clinical manifestations [5]. Diagnosing EoE and differentiating it from GERD are reviewed in greater detail separately. (See "Clinical manifestations and diagnosis of eosinophilic esophagitis (EoE)", section on 'Distinction from GERD'.)

Antigenic proteins, typically derived from food and less commonly from inhaled proteins, trigger an adaptive T helper type 2 (Th2) cell-mediated response that produces cytokines, such as interleukin (IL) 5 and IL-13 [6]. IL-13 subsequently triggers resident cells, such as esophageal epithelial cells, to produce a large set of proteins. The most strongly induced gene in this process is eotaxin 3, which in turn recruits eosinophils from the peripheral blood into the tissue [7]. Antigen-driven Th2 cells also produce IL-5 and IL-13. IL-5 is a chief eosinophil growth and activation factor that primes eosinophils to have enhanced responsiveness to eotaxin 3 and prolongs their cellular survival. IL-13 induces other key mediators of EoE including eotaxin 3 and calpain 14 (CAPN14; the gene product of the primary EoE susceptibility locus) [8]. These type 2 cytokines are largely produced by activated effector memory pathogenic CD4+ T cells that accumulate in the esophagus [9]. IL-13 also decreases the expression of genes that encode proteins involved in barrier function such as filaggrin and components of esophageal desmosomes, such as desmoglein 1 [10]. Rare genetic loss-of-function variants in other desmosome genes, including those encoding periplakin and desmoplakin, are also associated with EoE [11].

ROLE OF THE IMMUNE SYSTEM AND ENVIRONMENTAL FACTORS — Host immune system mechanisms in EoE appear to fall somewhere in between pure immunoglobulin E (IgE) mediated and delayed T helper type 2 (Th2) responses [3,12-14]. Eosinophils, T cells, and mast cells are elevated in esophageal mucosal biopsies [15], and mast cell degranulation and eosinophil cytolysis and degranulation are common findings in tissue specimens from patients with eosinophilic gastrointestinal disorders (EGIDs) [16-18]. Mast cell levels are more closely linked with clinical symptoms than eosinophil levels [19]. Studies have identified contributory roles for allergens, cytokines, micro-ribonucleic acid (microRNA or miRNA), chemokines, and polarization of Th2 immunity in the disease pathophysiology. In addition, EoE is uniquely characterized by high levels of systemic and esophageal immunoglobulin G4 (IgG4), which include food-specific responses [20-22]. The high levels of IgG4 may play a role in blocking IgE responses and may explain the dissociation between the absence of positive skin tests and effective elimination diets.

Allergens — The association of EoE with allergies suggests that eosinophil recruitment to the esophagus may be an immune response to environmental antigens in genetically predisposed individuals. Several lines of evidence support an allergic etiology. The role of allergies in EoE is briefly reviewed here and is discussed in greater detail separately. (See "Allergy testing in eosinophilic esophagitis", section on 'Evidence for role of allergies in EoE' and "Dietary management of eosinophilic esophagitis".)

Approximately 75 percent of patients with EoE or other EGIDs are atopic [23-32]. The majority of patients have evidence of food allergen and aeroallergen sensitization, as defined by skin prick and/or allergen-specific IgE tests, although only a minority has a history of food anaphylaxis (an IgE-mediated reaction) [2]. EoE can typically be reversed by institution of an allergen-free diet [23,24,33,34], and relapse is common upon food reintroduction [1]. In addition, priming with epicutaneous exposure to antigens can result in esophageal eosinophil recruitment in a mouse model [35]. Patients with EoE also frequently report seasonal variations in their symptoms, with fewer cases being diagnosed in the winter months (when outdoor aeroallergens are relatively low) [36-38]. In addition, increased eosinophil accumulation is seen in the esophagus of patients with EoE and seasonal allergic rhinitis with hypersensitivity to grass pollen [39]. EoE has also been reported in a few patients treated with sublingual pollen immunotherapy [40,41] and can be triggered by oral immunotherapy [42].

Eosinophils and chemokines — EoE typically occurs in the absence of substantial peripheral blood eosinophilia, indicating the potential significance of esophageal-specific mechanisms for regulating eosinophil levels. When blood eosinophilia occurs in EoE, it is typically mild in magnitude (<500 cells/microL of blood). Eotaxin plays a central role in antigen-mediated eosinophil recruitment [6,7,43,44], and the importance of a local eotaxin pathway in this process has been demonstrated in several studies [43-48]. Collectively, these results strongly implicate eotaxin 3 in the pathoetiology of EoE and offer a molecular connection between Th2 inflammation and the development of EoE.

In murine models of EoE, eosinophil recruitment is attenuated in the absence of eotaxin [47], and mice with a genetic ablation of the eotaxin receptor (C-C motif chemokine receptor 3 [CCR3]) are protected from the development of experimental EoE [45]. The later study also reported on a genome-wide microarray expression profile analysis of esophageal tissue that was used to compare gene transcript expression in the esophageal tissue of patients with EoE or chronic esophagitis (typical of gastroesophageal reflux disease [GERD]) and normal individuals. Eotaxin 3 (also called chemokine [C-C motif] ligand 26, CCL26) was the most overexpressed gene in patients with EoE.

Levels of eotaxin 3 correlated with disease severity in a study in which overexpression of eotaxin 3 alone had a predictive value of 89 percent in diagnosing EoE from a single esophageal biopsy [46]. Another study demonstrated that treatment of EoE with topical glucocorticoids downregulated esophageal eotaxin 3 levels (as well as other cytokines implicated in EoE) [48]. A number of studies in experimental systems in rodents have shown that eosinophils have an effector role in mediating the pathologic features of EoE [49].

Th2 immunity and cytokines — Adaptive T cell immunity, driven by T helper type 2 (Th2) cells and involving interleukin (IL) 13, IL-5, and IL-20 family member expression, appears to play a major role in the pathogenesis of EoE [9,15,50-54].

Studies of cytokines in EoE have shown the following:

IL-5 is expressed by Th2 cells and eosinophils. It is a key mediator in eosinophil activation and appears to mediate eosinophil-induced esophageal remodeling and collagen deposition [49]. IL-5 expression is increased in tissue samples from children with EoE [55]. In a murine model of EoE, eosinophil recruitment was ablated in the absence of IL-5 [47]. Overexpression of IL-5 in mice leads to increased esophageal eosinophils, and development of EoE is blocked in IL-5-deficient mice [56].

IL-13 is secreted by Th2 cells in particular. One of its effects is to increase eotaxin 3 expression. Elevated levels of IL-13 messenger RNA (mRNA) are seen in esophageal biopsy specimens from patients with EoE [57]. These findings are ablated with glucocorticoid treatment. In addition, IL-13 stimulation induces an EoE-specific esophageal transcriptome, as well as the gene product from the main EoE susceptibility locus, CAPN14 [58,59]. Repeated delivery of specific allergens or IL-13 to the lung of mice, as well transgenic overexpression of IL-13 in the lung of mice, induces experimental EoE [52,60,61], suggesting that esophageal eosinophilic inflammation is mechanistically linked with pulmonary inflammation. Blockade of IL-13 in vivo with human anti-IL-13 antibodies attenuates esophageal eosinophilia and clinical features and improves endoscopic and molecular markers of EoE [62,63]. The key role of IL-13 and the related cytokine IL-4 is best exemplified by the ability of dupilumab to improve symptomatic, endoscopic, histologic, and molecular aspects of EoE [63], resulting in its US Food and Drug Administration (FDA) approval as the first drug for EoE. (See 'Mast cells' below and "Treatment of eosinophilic esophagitis (EoE)", section on 'Dupilumab'.)

Thymic stromal lymphopoietin (TSLP) promotes Th2 responses, and elevated levels of TSLP are found in the esophagus of patients with EoE [64]. TSLP and its receptor, in concert with transforming growth factor beta 1 (TGF-beta-1), affect tolerance mechanisms in dendritic cells in vitro [65]. This suggests mechanistically that food tolerance mechanisms are disrupted in patients with EoE. A nonsynonymous polymorphism in the gene for the TSLP receptor was associated with EoE in male patients only [66]. TSLP is overproduced in the absence of the anti-protease serine peptidase inhibitor, kazal type 7 (SPINK7), which is deficient in the esophagus of patients with EoE [67]. (See 'Genetic defects' below.)

Increased expression of fibroblast growth factor 9 (FGF9) and other profibrogenic cytokine genes, such as IL-5 in the subepithelial layer of the esophagus, is seen in patients with EoE, suggesting that these cytokines may have a role in the fibrogenic response [68,69]. Prolonged treatment with swallowed glucocorticoids is associated with a downregulation of profibrogenic cytokine gene expression [69].

IL-20 subfamily members, IL-19, IL-20, and IL-24, are increased in the esophagus of EoE patients. Their overexpression induces epithelial barrier defects. Experimental knockout of the IL-20 receptor attenuated experimental EoE associated with esophageal eosinophilia, loss of filaggrin, and type 2 cytokine production [70].

Mast cells — The exact role of mast cells in EoE is unclear. Increased numbers of mast cells are seen in esophageal tissue samples from patients with EoE, and degranulation is common [71]. Results from a murine model of EoE suggest that mast cells may have an important role in esophageal remodeling in EoE by promoting muscle cell hyperplasia and hypertrophy [72]. Elevated TGF-beta, produced by eosinophils and mast cells, contributes to esophageal tissue remodeling and smooth muscle dysfunction in patients with EoE, similar to that seen in the airways of patients with asthma [73], further supporting the link between esophageal and pulmonary inflammation. However, cromolyn sodium, a mast cell stabilizer, has shown little benefit in the treatment of EoE [74]. (See 'Th2 immunity and cytokines' above.)

MicroRNA — MicroRNAs (miRNAs) act as regulators of mRNA expression and translation. In one study, esophageal microRNA expression correlated with eosinophil levels [75]. miR-21 and miR-223 showed the greatest upregulation in patients with EoE compared with normal controls. These two microRNAs may play a role in regulation of eosinophilia and polarization of adaptive immunity. miR-375 was the most downregulated. Levels of these microRNAs mostly reverted to normal with glucocorticoid treatment. Patients in remission showed increased expression of miR-675. In the plasma, the most differentially expressed microRNAs were miR-146a, miR-146b, and miR-223.

GENETICS AND FAMILY HISTORY — A genetic predisposition to EoE is supported by evidence of familial clustering [76] and twin studies, which have revealed a 58 percent concordance in monozygotic twins and a 36 percent concordance in dizygotic twins compared with regular fraternal siblings [77]. The risk of EoE for other siblings is 2 to 3 percent. Collectively, twin studies have indicated a strong heritability of EoE, largely accounted for by shared environment in early life.

Early-life exposures modify the risk of EoE, with maternal fever, pre- and postnatal antibiotics, proton pump inhibitor (PPI) therapy, and neonatal intensive care unit admission associated with increased risk of EoE and pet exposure in the first year of life associated with a decreased risk of EoE [78]. Indeed, the esophagus harbors a unique microbiome, and dysbiosis increases IgE and EoE susceptibility [79].

In addition, several genetic variants that may predispose to EoE have been identified, especially at 5q22 (TSLP gene) and 2p23 (CAPN14 gene) [80]. The identified genetic markers for EoE are primarily common genetic variants, although one appears to be specific for EoE. CAPN14 is induced by interleukin (IL) 13 in esophageal epithelial cells and is involved in regulating barrier function, an impaired process germane to the development of EoE [58,59,81]. Very early onset of EoE (first 18 months of life) is particularly enriched in genetic variants in the CAPN14 gene, as well as associated with cesarean birth compared with late-onset EoE [82].

Family history — Over 30 families with multiple affected individuals were reported by 2008 [83]. In a study of 103 children with EoE, a positive family history was observed in 7 percent [76]. This included three sibling pairs and the mother of one of the pairs of siblings. A report from an eosinophilic gastrointestinal disorder (EGID) registry database showed that 16 percent of patients had an immediate family member with a similar disorder [84]. Almost 10 percent of patients with EoE and evidence of esophageal strictures have parents diagnosed with EoE [3]. There is also a large sibling risk ratio, estimated at approximately 50-fold compared with the general population [3]. This translates into 3 percent of patients with EoE having siblings who develop EoE. A number of rare genetic variants are responsible for familial presentations of EoE including predicted damaging variants in the mitochondrial genes encoding oxidoreductases dehydrogenase E1 and transketolase domain containing 1 (DHTKD1) and oxoglutarate dehydrogenase L (OGDHL) [85,86].

Genetic defects — The earliest work demonstrating that genetic factors have a role revealed dysregulated expression of approximately 1 percent of the entire human genome, constituting an EoE genetic signature [45]. Additional dysregulated transcripts, including several long-noncoding RNAs (lcnRNAs) that act as transcriptional regulators, were identified when RNA sequencing on esophageal biopsies was performed [87].

An EoE susceptibility locus was also identified at 5q22, involving the gene encoding TSLP [64]. The genes for TSLP and its receptor are both associated with EoE [65]. In addition, genetic susceptibility has been found at 5p23, encoding for the CAPN14 gene [58]. Calpain 14 is an esophageal-specific enzyme that regulates IL-13 responses in esophageal epithelial cells and probably explains, at least in part, the tissue-specific nature of the disease [59]. Multiple genetic events collectively contribute to EoE, including an interplay of atopy genes with EoE-specific elements [88]. Meta-analyses of genome-wide association studies have identified at least six other susceptibility loci including EMSY transcriptional repressor, BRCA2 interacting (EMSY)/leucine rich repeat containing 32 (LRRC32; 11q13) and BTB domain and CNC homolog 2 (BACH2; 6q15) [89,90].

EoE involves loss of esophageal barrier function, mediated by depressed expression of desmoglein 1 and SPINK7 [18,67], associated with a loss of epithelial cell differentiation [91]. In addition, genetic variants in the filaggrin gene are linked with EoE, highlighting the potential key role of alterations in barrier function in EoE. Finally, genetic variants in transforming growth factor (TGF) receptors and/or genes associated with inherited connective tissue disorders involving hypermobility syndrome are associated with EoE [65]. These genetic findings have provided insight into molecular etiology and disease susceptibility, but they have not elucidated why patients develop the tissue-specific response characteristic of EoE, since genes such as TSLP have been linked with other atopic disorders.

Relatively large genome-wide association studies and meta-analyses have determined that a major susceptibility linkage exists at 2p23 [58]. This region of the chromosome contains the CAPN14 gene, which encodes for calpain 14. This protein is specifically expressed in the esophagus, is markedly upregulated in patients with EoE compared with controls in a manner dependent upon the CAPN14 genotype, and is dramatically induced by IL-13 in esophageal epithelial cells by an epigenetic mechanism. (See 'Th2 immunity and cytokines' above.)

EoE molecular transcriptome — A 96-gene EoE diagnostic panel (EDP) has been developed based upon analysis of esophageal biopsies and is available commercially. The EDP provides deep information concerning the contribution of individual genes to the pathogenesis of EoE, especially on a patient-to-patient basis [92]. This diagnostic panel, which differentiates EoE from control individuals, including those with gastroesophageal reflux disease (GERD), can also differentiate patients with active and inactive disease and identify glucocorticoid exposure, providing substantial clinical value. Combining analysis of EDP with endoscopic, histologic, and clinical features has led to the identification of three different EoE endotypes that have different disease features and possibly unique course and response to therapy [93]. Molecular profiling has also revealed variable disease groups based upon differential expression of type 2 cytokine-related genes [94]. Genetic testing for the EoE molecular transcriptome may be useful in both diagnosis and management of EoE. (See "Clinical manifestations and diagnosis of eosinophilic esophagitis (EoE)", section on 'Subtypes' and "Clinical manifestations and diagnosis of eosinophilic esophagitis (EoE)", section on 'Laboratory tests'.)

SUMMARY

Overview – Eosinophilic esophagitis (EoE) is a chronic, immune/antigen-mediated esophageal disease characterized clinically by symptoms related to esophageal dysfunction and histologically by eosinophil-predominant inflammation. (See 'Introduction' above.)

Pathogenesis – The pathogenesis of EoE is incompletely understood but involves an interplay between genetic, environmental, and host immune system factors. Molecular analysis has elucidated that EoE is caused by a primary defect in esophageal epithelial function rather than an eosinophil defect. The esophagus of EoE patients has an impairment of epithelial cell differentiation and barrier function. (See 'Overview of pathogenesis' above.)

Role of host immune system – Host immune system mechanisms in EoE appear to fall somewhere in between pure immunoglobulin E (IgE) mediated and delayed T helper type 2 (Th2) responses. Studies have identified contributory roles for allergens, cytokines, microRNAs (miRNAs), chemokines, and polarization of Th2 immunity in the disease pathophysiology. The role of type 2 immunity is underscored by the US Food and Drug Administration (FDA) approval of dupilumab (anti-type 2 biological agent) for the treatment of EoE. (See 'Role of the immune system and environmental factors' above.)

Genetics – A genetic predisposition to EoE is supported by evidence of familial clustering and twin studies. In addition, several genetic defects that may predispose to EoE have been identified, especially at 2p23, encoding for the esophagus specific gene product, calpain 14. Genetic testing for the EoE molecular transcriptome is available and may be useful in diagnosis, disease subgroup identification (endotyping), and management of EoE. (See 'Genetics and family history' above.)

  1. Liacouras CA, Furuta GT, Hirano I, et al. Eosinophilic esophagitis: updated consensus recommendations for children and adults. J Allergy Clin Immunol 2011; 128:3.
  2. Fox VL, Nurko S, Furuta GT. Eosinophilic esophagitis: it's not just kid's stuff. Gastrointest Endosc 2002; 56:260.
  3. Rothenberg ME. Eosinophilic gastrointestinal disorders (EGID). J Allergy Clin Immunol 2004; 113:11.
  4. Molina-Infante J, Bredenoord AJ, Cheng E, et al. Proton pump inhibitor-responsive oesophageal eosinophilia: an entity challenging current diagnostic criteria for eosinophilic oesophagitis. Gut 2016; 65:524.
  5. Dellon ES, Liacouras CA, Molina-Infante J, et al. Updated International Consensus Diagnostic Criteria for Eosinophilic Esophagitis: Proceedings of the AGREE Conference. Gastroenterology 2018; 155:1022.
  6. Hogan SP, Mishra A, Brandt EB, et al. A critical role for eotaxin in experimental oral antigen-induced eosinophilic gastrointestinal allergy. Proc Natl Acad Sci U S A 2000; 97:6681.
  7. Mishra A, Hogan SP, Lee JJ, et al. Fundamental signals that regulate eosinophil homing to the gastrointestinal tract. J Clin Invest 1999; 103:1719.
  8. Litosh VA, Rochman M, Rymer JK, et al. Calpain-14 and its association with eosinophilic esophagitis. J Allergy Clin Immunol 2017; 139:1762.
  9. Wen T, Aronow BJ, Rochman Y, et al. Single-cell RNA sequencing identifies inflammatory tissue T cells in eosinophilic esophagitis. J Clin Invest 2019; 129:2014.
  10. Sherrill JD, Kc K, Wu D, et al. Desmoglein-1 regulates esophageal epithelial barrier function and immune responses in eosinophilic esophagitis. Mucosal Immunol 2014; 7:718.
  11. Shoda T, Kaufman KM, Wen T, et al. Desmoplakin and periplakin genetically and functionally contribute to eosinophilic esophagitis. Nat Commun 2021; 12:6795.
  12. Sampson HA. Food allergy. Part 2: diagnosis and management. J Allergy Clin Immunol 1999; 103:981.
  13. Sampson HA. Food allergy. Part 1: immunopathogenesis and clinical disorders. J Allergy Clin Immunol 1999; 103:717.
  14. Bischoff S, Crowe SE. Gastrointestinal food allergy: new insights into pathophysiology and clinical perspectives. Gastroenterology 2005; 128:1089.
  15. Straumann A, Bauer M, Fischer B, et al. Idiopathic eosinophilic esophagitis is associated with a T(H)2-type allergic inflammatory response. J Allergy Clin Immunol 2001; 108:954.
  16. Oyaizu N, Uemura Y, Izumi H, et al. Eosinophilic gastroenteritis. Immunohistochemical evidence for IgE mast cell-mediated allergy. Acta Pathol Jpn 1985; 35:759.
  17. Saffari H, Hoffman LH, Peterson KA, et al. Electron microscopy elucidates eosinophil degranulation patterns in patients with eosinophilic esophagitis. J Allergy Clin Immunol 2014; 133:1728.
  18. Sherrill JD, Rothenberg ME. Genetic and epigenetic underpinnings of eosinophilic esophagitis. Gastroenterol Clin North Am 2014; 43:269.
  19. Zhang S, Shoda T, Aceves SS, et al. Mast cell-pain connection in eosinophilic esophagitis. Allergy 2022; 77:1895.
  20. Wright BL, Kulis M, Guo R, et al. Food-specific IgG4 is associated with eosinophilic esophagitis. J Allergy Clin Immunol 2016; 138:1190.
  21. Clayton F, Fang JC, Gleich GJ, et al. Eosinophilic esophagitis in adults is associated with IgG4 and not mediated by IgE. Gastroenterology 2014; 147:602.
  22. Zukerberg L, Mahadevan K, Selig M, Deshpande V. Oesophageal intrasquamous IgG4 deposits: an adjunctive marker to distinguish eosinophilic oesophagitis from reflux oesophagitis. Histopathology 2016; 68:968.
  23. Spergel JM, Beausoleil JL, Mascarenhas M, Liacouras CA. The use of skin prick tests and patch tests to identify causative foods in eosinophilic esophagitis. J Allergy Clin Immunol 2002; 109:363.
  24. Walsh SV, Antonioli DA, Goldman H, et al. Allergic esophagitis in children: a clinicopathological entity. Am J Surg Pathol 1999; 23:390.
  25. Caldwell JH, Sharma HM, Hurtubise PE, Colwell DL. Eosinophilic gastroenteritis in extreme allergy. Immunopathological comparison with nonallergic gastrointestinal disease. Gastroenterology 1979; 77:560.
  26. Caldwell JH, Tennenbaum JI, Bronstein HA. Serum IgE in eosinophilic gastroenteritis. Response to intestinal challenge in two cases. N Engl J Med 1975; 292:1388.
  27. Cello JP. Eosinophilic gastroenteritis--a complex disease entity. Am J Med 1979; 67:1097.
  28. Scudamore HH, Phillips SF, Swedlund HA, Gleich GJ. Food allergy manifested by eosinophilia, elevated immunoglobulin E level, and protein-losing enteropathy: the syndrome of allergic gastroenteropathy. J Allergy Clin Immunol 1982; 70:129.
  29. Furuta GT, Ackerman SJ, Wershil BK. The role of the eosinophil in gastrointestinal diseases. Curr Opin Gastroenterol 1995; 11:541.
  30. Iacono G, Carroccio A, Cavataio F, et al. Gastroesophageal reflux and cow's milk allergy in infants: a prospective study. J Allergy Clin Immunol 1996; 97:822.
  31. Sampson HA. Food allergy. JAMA 1997; 278:1888.
  32. Woodruff SA, Masterson JC, Fillon S, et al. Role of eosinophils in inflammatory bowel and gastrointestinal diseases. J Pediatr Gastroenterol Nutr 2011; 52:650.
  33. Kelly KJ, Lazenby AJ, Rowe PC, et al. Eosinophilic esophagitis attributed to gastroesophageal reflux: improvement with an amino acid-based formula. Gastroenterology 1995; 109:1503.
  34. Henderson CJ, Abonia JP, King EC, et al. Comparative dietary therapy effectiveness in remission of pediatric eosinophilic esophagitis. J Allergy Clin Immunol 2012; 129:1570.
  35. Akei HS, Mishra A, Blanchard C, Rothenberg ME. Epicutaneous antigen exposure primes for experimental eosinophilic esophagitis in mice. Gastroenterology 2005; 129:985.
  36. Prasad GA, Alexander JA, Schleck CD, et al. Epidemiology of eosinophilic esophagitis over three decades in Olmsted County, Minnesota. Clin Gastroenterol Hepatol 2009; 7:1055.
  37. Wang FY, Gupta SK, Fitzgerald JF. Is there a seasonal variation in the incidence or intensity of allergic eosinophilic esophagitis in newly diagnosed children? J Clin Gastroenterol 2007; 41:451.
  38. Almansa C, Krishna M, Buchner AM, et al. Seasonal distribution in newly diagnosed cases of eosinophilic esophagitis in adults. Am J Gastroenterol 2009; 104:828.
  39. Onbasi K, Sin AZ, Doganavsargil B, et al. Eosinophil infiltration of the oesophageal mucosa in patients with pollen allergy during the season. Clin Exp Allergy 2005; 35:1423.
  40. Antico A, Fante R. Esophageal hypereosinophilia induced by grass sublingual immunotherapy. J Allergy Clin Immunol 2014; 133:1482.
  41. Miehlke S, Alpan O, Schröder S, Straumann A. Induction of eosinophilic esophagitis by sublingual pollen immunotherapy. Case Rep Gastroenterol 2013; 7:363.
  42. Lucendo AJ, Arias A, Tenias JM. Relation between eosinophilic esophagitis and oral immunotherapy for food allergy: a systematic review with meta-analysis. Ann Allergy Asthma Immunol 2014; 113:624.
  43. Garcia-Zepeda EA, Rothenberg ME, Ownbey RT, et al. Human eotaxin is a specific chemoattractant for eosinophil cells and provides a new mechanism to explain tissue eosinophilia. Nat Med 1996; 2:449.
  44. Furuta GT, Sherman P. Eotaxin and eosinophilic homing to the gut. J Pediatr Gastroenterol Nutr 2000; 30:229.
  45. Blanchard C, Wang N, Stringer KF, et al. Eotaxin-3 and a uniquely conserved gene-expression profile in eosinophilic esophagitis. J Clin Invest 2006; 116:536.
  46. Blanchard C, Stucke EM, Rodriguez-Jimenez B, et al. A striking local esophageal cytokine expression profile in eosinophilic esophagitis. J Allergy Clin Immunol 2011; 127:208.
  47. Mishra A, Hogan SP, Brandt EB, Rothenberg ME. An etiological role for aeroallergens and eosinophils in experimental esophagitis. J Clin Invest 2001; 107:83.
  48. Lucendo AJ, De Rezende L, Comas C, et al. Treatment with topical steroids downregulates IL-5, eotaxin-1/CCL11, and eotaxin-3/CCL26 gene expression in eosinophilic esophagitis. Am J Gastroenterol 2008; 103:2184.
  49. Mishra A, Wang M, Pemmaraju VR, et al. Esophageal remodeling develops as a consequence of tissue specific IL-5-induced eosinophilia. Gastroenterology 2008; 134:204.
  50. Rothenberg ME. Biology and treatment of eosinophilic esophagitis. Gastroenterology 2009; 137:1238.
  51. Blanchard C, Stucke EM, Burwinkel K, et al. Coordinate interaction between IL-13 and epithelial differentiation cluster genes in eosinophilic esophagitis. J Immunol 2010; 184:4033.
  52. Zuo L, Fulkerson PC, Finkelman FD, et al. IL-13 induces esophageal remodeling and gene expression by an eosinophil-independent, IL-13R alpha 2-inhibited pathway. J Immunol 2010; 185:660.
  53. Zhu X, Wang M, Mavi P, et al. Interleukin-15 expression is increased in human eosinophilic esophagitis and mediates pathogenesis in mice. Gastroenterology 2010; 139:182.
  54. Furuta GT, Straumann A. Review article: the pathogenesis and management of eosinophilic oesophagitis. Aliment Pharmacol Ther 2006; 24:173.
  55. Gupta SK, Fitzgerald JF, Kondratyuk T, HogenEsch H. Cytokine expression in normal and inflamed esophageal mucosa: a study into the pathogenesis of allergic eosinophilic esophagitis. J Pediatr Gastroenterol Nutr 2006; 42:22.
  56. Mishra A, Hogan SP, Brandt EB, Rothenberg ME. IL-5 promotes eosinophil trafficking to the esophagus. J Immunol 2002; 168:2464.
  57. Blanchard C, Mingler MK, Vicario M, et al. IL-13 involvement in eosinophilic esophagitis: transcriptome analysis and reversibility with glucocorticoids. J Allergy Clin Immunol 2007; 120:1292.
  58. Kottyan LC, Davis BP, Sherrill JD, et al. Genome-wide association analysis of eosinophilic esophagitis provides insight into the tissue specificity of this allergic disease. Nat Genet 2014; 46:895.
  59. Davis BP, Stucke EM, Khorki ME, et al. Eosinophilic esophagitis-linked calpain 14 is an IL-13-induced protease that mediates esophageal epithelial barrier impairment. JCI Insight 2016; 1:e86355.
  60. Mishra A, Weaver TE, Beck DC, Rothenberg ME. Interleukin-5-mediated allergic airway inflammation inhibits the human surfactant protein C promoter in transgenic mice. J Biol Chem 2001; 276:8453.
  61. Mishra A, Rothenberg ME. Intratracheal IL-13 induces eosinophilic esophagitis by an IL-5, eotaxin-1, and STAT6-dependent mechanism. Gastroenterology 2003; 125:1419.
  62. Rothenberg ME, Wen T, Greenberg A, et al. Intravenous anti-IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. J Allergy Clin Immunol 2015; 135:500.
  63. Hirano I, Dellon ES, Hamilton JD, et al. Efficacy of Dupilumab in a Phase 2 Randomized Trial of Adults With Active Eosinophilic Esophagitis. Gastroenterology 2020; 158:111.
  64. Rothenberg ME, Spergel JM, Sherrill JD, et al. Common variants at 5q22 associate with pediatric eosinophilic esophagitis. Nat Genet 2010; 42:289.
  65. Abonia JP, Wen T, Stucke EM, et al. High prevalence of eosinophilic esophagitis in patients with inherited connective tissue disorders. J Allergy Clin Immunol 2013; 132:378.
  66. Sherrill JD, Gao PS, Stucke EM, et al. Variants of thymic stromal lymphopoietin and its receptor associate with eosinophilic esophagitis. J Allergy Clin Immunol 2010; 126:160.
  67. Azouz NP, Ynga-Durand MA, Caldwell JM, et al. The antiprotease SPINK7 serves as an inhibitory checkpoint for esophageal epithelial inflammatory responses. Sci Transl Med 2018; 10.
  68. Mulder DJ, Pacheco I, Hurlbut DJ, et al. FGF9-induced proliferative response to eosinophilic inflammation in oesophagitis. Gut 2009; 58:166.
  69. Lucendo AJ, Arias A, De Rezende LC, 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; 128:1037.
  70. Kaymak T, Kaya B, Wuggenig P, et al. IL-20 subfamily cytokines impair the oesophageal epithelial barrier by diminishing filaggrin in eosinophilic oesophagitis. Gut 2023; 72:821.
  71. Abonia JP, Blanchard C, Butz BB, et al. Involvement of mast cells in eosinophilic esophagitis. J Allergy Clin Immunol 2010; 126:140.
  72. Niranjan R, Mavi P, Rayapudi M, et al. Pathogenic role of mast cells in experimental eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol 2013; 304:G1087.
  73. Aceves SS, Newbury RO, Dohil R, et al. Esophageal remodeling in pediatric eosinophilic esophagitis. J Allergy Clin Immunol 2007; 119:206.
  74. Lieberman JA, Zhang J, Whitworth J, Cavender C. A randomized, double-blinded, placebo-controlled study of the use of viscous oral cromolyn sodium for the treatment of eosinophilic esophagitis. Ann Allergy Asthma Immunol 2018; 120:527.
  75. Lu TX, Sherrill JD, Wen T, et al. MicroRNA signature in patients with eosinophilic esophagitis, reversibility with glucocorticoids, and assessment as disease biomarkers. J Allergy Clin Immunol 2012; 129:1064.
  76. Noel RJ, Putnam PE, Rothenberg ME. Eosinophilic esophagitis. N Engl J Med 2004; 351:940.
  77. Alexander ES, Martin LJ, Collins MH, et al. Twin and family studies reveal strong environmental and weaker genetic cues explaining heritability of eosinophilic esophagitis. J Allergy Clin Immunol 2014; 134:1084.
  78. Jensen ET, Kuhl JT, Martin LJ, et al. Early-life environmental exposures interact with genetic susceptibility variants in pediatric patients with eosinophilic esophagitis. J Allergy Clin Immunol 2018; 141:632.
  79. Brusilovsky M, Bao R, Rochman M, et al. Host-Microbiota Interactions in the Esophagus During Homeostasis and Allergic Inflammation. Gastroenterology 2022; 162:521.
  80. Kottyan LC, Rothenberg ME. Genetics of eosinophilic esophagitis. Mucosal Immunol 2017; 10:580.
  81. Miller DE, Forney C, Rochman M, et al. Genetic, Inflammatory, and Epithelial Cell Differentiation Factors Control Expression of Human Calpain-14. G3 (Bethesda) 2019; 9:729.
  82. Lyles JL, Martin LJ, Shoda T, et al. Very early onset eosinophilic esophagitis is common, responds to standard therapy, and demonstrates enrichment for CAPN14 genetic variants. J Allergy Clin Immunol 2021; 147:244.
  83. Collins MH, Blanchard C, Abonia JP, et al. Clinical, pathologic, and molecular characterization of familial eosinophilic esophagitis compared with sporadic cases. Clin Gastroenterol Hepatol 2008; 6:621.
  84. Guajardo JR, Plotnick LM, Fende JM, et al. Eosinophil-associated gastrointestinal disorders: a world-wide-web based registry. J Pediatr 2002; 141:576.
  85. Sherrill JD, Kc K, Wang X, et al. Whole-exome sequencing uncovers oxidoreductases DHTKD1 and OGDHL as linkers between mitochondrial dysfunction and eosinophilic esophagitis. JCI Insight 2018; 3.
  86. Kottyan LC, Parameswaran S, Weirauch MT, et al. The genetic etiology of eosinophilic esophagitis. J Allergy Clin Immunol 2020; 145:9.
  87. Sherrill JD, Kiran KC, Blanchard C, et al. Analysis and expansion of the eosinophilic esophagitis transcriptome by RNA sequencing. Genes Immun 2014; 15:361.
  88. Martin LJ, He H, Collins MH, et al. Eosinophilic esophagitis (EoE) genetic susceptibility is mediated by synergistic interactions between EoE-specific and general atopic disease loci. J Allergy Clin Immunol 2018; 141:1690.
  89. Kottyan LC, Trimarchi MP, Lu X, et al. Replication and meta-analyses nominate numerous eosinophilic esophagitis risk genes. J Allergy Clin Immunol 2021; 147:255.
  90. Chang X, March M, Mentch F, et al. A genome-wide association meta-analysis identifies new eosinophilic esophagitis loci. J Allergy Clin Immunol 2022; 149:988.
  91. Rochman M, Azouz NP, Rothenberg ME. Epithelial origin of eosinophilic esophagitis. J Allergy Clin Immunol 2018; 142:10.
  92. Wen T, Stucke EM, Grotjan TM, et al. Molecular diagnosis of eosinophilic esophagitis by gene expression profiling. Gastroenterology 2013; 145:1289.
  93. Keely S, Talley NJ. Endophenotyping eosinophilic oesophagitis: a new era for management? Lancet Gastroenterol Hepatol 2018; 3:449.
  94. Dunn JLM, Shoda T, Caldwell JM, et al. Esophageal type 2 cytokine expression heterogeneity in eosinophilic esophagitis in a multisite cohort. J Allergy Clin Immunol 2020; 145:1629.
Topic 90146 Version 9.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟