Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters










Publication year range
1.
Allergy ; 73(9): 1892-1901, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29790577

ABSTRACT

BACKGROUND: Recent data associate eosinophilic esophagitis (EoE) with IgG4 rather than IgE, but its significance and function have not been determined. Our aims were to measure esophageal IgG4 levels and to determine functional correlations as assessed by histologic and transcriptome analyses. METHODS: This case-control study included pediatric subjects with EoE (≥15 eosinophils/HPF) and non-EoE controls. Protein lysates were analyzed for IgA, IgM, and IgG1-IgG4 using the Luminex 100 system; IgE was quantified by ELISA. Esophageal biopsies were scored using the EoE histology scoring system. Transcripts were probed by the EoE diagnostic panel, designed to examine the expression of 96 esophageal transcripts. RESULTS: Esophageal IgG subclasses, IgA, and IgM, but not IgE, were increased in subjects with EoE relative to controls. The greatest change between groups was seen in IgG4 (4.2 mg/g protein [interquartile range: 1.0-13.1 mg/g protein] vs 0.2 mg/g protein [0.1-0.9]; P < .0001). Tissue IgG4 levels correlated with esophageal eosinophil counts (P = .0006); histologic grade (P = .0011) and stage (P = .0112) scores; and IL4, IL10, IL13, but not TGFB1, expression and had strong associations with a subset of the EoE transcriptome. Esophageal IgG4 transcript expression was increased and correlated with IgG4 protein levels and IL10 expression. CONCLUSION: These findings extend prior studies on IgG4 in adult EoE to the pediatric population and provide deeper understanding of the potential significance and regulation of IgG4, demonstrating that IgG4 is a relevant feature of the disease; is closely related to esophageal eosinophil levels, type 2 immunity and T regulatory cytokines; and is likely produced locally.


Subject(s)
Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/etiology , Immunoglobulin G/immunology , Transcriptome , Biopsy , Case-Control Studies , Child , Child, Preschool , Esophageal Mucosa/immunology , Esophageal Mucosa/metabolism , Esophageal Mucosa/pathology , Esophagus/immunology , Esophagus/metabolism , Esophagus/pathology , Female , Gene Expression , Histocytochemistry , Humans , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Isotypes/immunology , Male
2.
Sci Rep ; 7(1): 17563, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29242581

ABSTRACT

The molecular and cellular etiology of eosinophilic esophagitis (EoE), an emerging tissue-specific allergic disease, involves dysregulated gene expression in esophageal epithelial cells. Herein, we assessed the esophageal expression of IL-33, an epithelium-derived alarmin cytokine, in patients with EoE. IL-33 protein was markedly overexpressed within the nuclei of a subpopulation of basal layer esophageal epithelial cells in patients with active EoE compared to control individuals. IL-33 exhibited dynamic expression as levels normalized upon EoE remission. IL-33-positive basal epithelial cells expressed E-cadherin and the undifferentiated epithelial cell markers keratin 5 and 14 but not the differentiation marker keratin 4. Moreover, the IL-33-positive epithelial cells expressed the epithelial progenitor markers p75 and p63 and lacked the proliferation markers Ki67 and phospho-histone H3. Additionally, the IL-33-positive cells had low expression of PCNA. IL-33 expression was detected in ex vivo-cultured primary esophageal epithelial cells in a subpopulation of cells lacking expression of proliferation markers. Collectively, we report that IL-33 expression is induced in an undifferentiated, non-dividing esophageal epithelial cell population in patients with active EoE.


Subject(s)
Eosinophilic Esophagitis/metabolism , Eosinophilic Esophagitis/pathology , Epithelial Cells/metabolism , Esophagus/pathology , Interleukin-33/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Humans
5.
Mucosal Immunol ; 10(3): 580-588, 2017 05.
Article in English | MEDLINE | ID: mdl-28224995

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic, allergic disease associated with marked mucosal eosinophil accumulation. EoE disease risk is multifactorial and includes environmental and genetic factors. This review will focus on the contribution of genetic variation to EoE risk, as well as the experimental tools and statistical methodology used to identify EoE risk loci. Specific disease-risk loci that are shared between EoE and other allergic diseases (TSLP, LRRC32) or unique to EoE (CAPN14), as well as Mendellian Disorders associated with EoE, will be reviewed in the context of the insight that they provide into the molecular pathoetiology of EoE. We will also discuss the clinical opportunities that genetic analyses provide in the form of decision support tools, molecular diagnostics, and novel therapeutic approaches.


Subject(s)
Calpain/genetics , Cytokines/genetics , Eosinophilic Esophagitis/genetics , Membrane Proteins/genetics , Mucous Membrane/immunology , Animals , Clinical Decision-Making , Eosinophils , Gene-Environment Interaction , Genetic Predisposition to Disease , Genetic Testing , Genotype , Humans , Polymorphism, Genetic , Thymic Stromal Lymphopoietin
6.
J Exp Med ; 214(3): 669-680, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28126831

ABSTRACT

Nonimmunological connective tissue phenotypes in humans are common among some congenital and acquired allergic diseases. Several of these congenital disorders have been associated with either increased TGF-ß activity or impaired STAT3 activation, suggesting that these pathways might intersect and that their disruption may contribute to atopy. In this study, we show that STAT3 negatively regulates TGF-ß signaling via ERBB2-interacting protein (ERBIN), a SMAD anchor for receptor activation and SMAD2/3 binding protein. Individuals with dominant-negative STAT3 mutations (STAT3mut ) or a loss-of-function mutation in ERBB2IP (ERBB2IPmut ) have evidence of deregulated TGF-ß signaling with increased regulatory T cells and total FOXP3 expression. These naturally occurring mutations, recapitulated in vitro, impair STAT3-ERBIN-SMAD2/3 complex formation and fail to constrain nuclear pSMAD2/3 in response to TGF-ß. In turn, cell-intrinsic deregulation of TGF-ß signaling is associated with increased functional IL-4Rα expression on naive lymphocytes and can induce expression and activation of the IL-4/IL-4Rα/GATA3 axis in vitro. These findings link increased TGF-ß pathway activation in ERBB2IPmut and STAT3mut patient lymphocytes with increased T helper type 2 cytokine expression and elevated IgE.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Hypersensitivity/immunology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Adaptor Proteins, Signal Transducing/deficiency , Humans , Interleukin-4/physiology , Receptors, Interleukin-4/physiology , Smad2 Protein/analysis , Smad2 Protein/physiology , Smad3 Protein/analysis , Smad3 Protein/physiology
7.
Mucosal Immunol ; 10(5): 1190-1201, 2017 09.
Article in English | MEDLINE | ID: mdl-28051089

ABSTRACT

Cadherins (CDH) mediate diverse processes critical in inflammation, including cell adhesion, migration, and differentiation. Herein, we report that the uncharacterized cadherin 26 (CDH26) is highly expressed by epithelial cells in human allergic gastrointestinal tissue. In vitro, CDH26 promotes calcium-dependent cellular adhesion of cells lacking endogenous CDHs by a mechanism involving homotypic binding and interaction with catenin family members (alpha, beta, and p120), as assessed by biochemical assays. Additionally, CDH26 enhances cellular adhesion to recombinant integrin α4ß7 in vitro; conversely, recombinant CDH26 binds αE and α4 integrins in biochemical and cellular functional assays, respectively. Interestingly, CDH26-Fc inhibits activation of human CD4+ T cells in vitro including secretion of IL-2. Taken together, we have identified a novel functional CDH regulated during allergic responses with unique immunomodulatory properties, as it binds α4 and αE integrins and regulates leukocyte adhesion and activation, and may thus represent a novel checkpoint for immune regulation and therapy via CDH26-Fc.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cadherins/metabolism , Epithelial Cells/physiology , Hypersensitivity/immunology , Inflammation/immunology , Intestinal Mucosa/metabolism , Adolescent , Adult , Cadherins/genetics , Cell Adhesion , Child , Child, Preschool , Female , HEK293 Cells , Humans , Infant , Integrin alpha Chains/metabolism , Integrin alpha4/metabolism , Integrin beta Chains/metabolism , Intestines/pathology , Lymphocyte Activation , Male , Protein Binding , Young Adult
8.
Dis Esophagus ; 30(3): 1-8, 2017 02 01.
Article in English | MEDLINE | ID: mdl-26857345

ABSTRACT

Eosinophilic esophagitis (EoE) is diagnosed by symptoms, and at least 15 intraepithelial eosinophils per high power field in an esophageal biopsy. Other pathologic features have not been emphasized. We developed a histology scoring system for esophageal biopsies that evaluates eight features: eosinophil density, basal zone hyperplasia, eosinophil abscesses, eosinophil surface layering, dilated intercellular spaces (DIS), surface epithelial alteration, dyskeratotic epithelial cells, and lamina propria fibrosis. Severity (grade) and extent (stage) of abnormalities were scored using a 4-point scale (0 normal; 3 maximum change). Reliability was demonstrated by strong to moderate agreement among three pathologists who scored biopsies independently (P ≤ 0.008). Several features were often abnormal in 201 biopsies (101 distal, 100 proximal) from 104 subjects (34 untreated, 167 treated). Median grade and stage scores were significantly higher in untreated compared with treated subjects (P ≤ 0.0062). Grade scores for features independent of eosinophil counts were significantly higher in biopsies from untreated compared with treated subjects (basal zone hyperplasia P ≤ 0.024 and DIS P ≤ 0.005), and were strongly correlated (R-square >0.67). Principal components analysis identified three principal components that explained 78.2% of the variation in the features. In logistic regression models, two principal components more closely associated with treatment status than log distal peak eosinophil count (PEC) (R-square 17, area under the curve (AUC) 77.8 vs. R-square 9, AUC 69.8). In summary, the EoE histology scoring system provides a method to objectively assess histologic changes in the esophagus beyond eosinophil number. Importantly, it discriminates treated from untreated patients, uses features commonly found in such biopsies, and is utilizable by pathologists after minimal training. These data provide rationales and a method to evaluate esophageal biopsies for features in addition to PEC.


Subject(s)
Biopsy/statistics & numerical data , Eosinophilic Esophagitis/diagnosis , Eosinophils , Leukocyte Count/methods , Severity of Illness Index , Area Under Curve , Biopsy/methods , Child , Esophagus/pathology , Female , Humans , Logistic Models , Male , Prospective Studies , Reproducibility of Results
9.
Mucosal Immunol ; 10(1): 194-204, 2017 01.
Article in English | MEDLINE | ID: mdl-27049063

ABSTRACT

Eosinophils contribute to type II immune responses in helminth infections and allergic diseases; however, their influence on intracellular pathogens is less clear. We previously reported that CCR2-/- mice exposed to the intracellular fungal pathogen Histoplasma capsulatum exhibit dampened immunity caused by an early exaggerated interleukin (IL)-4 response. We sought to identify the cellular source promulgating IL-4 in infected mutant animals. Eosinophils were the principal instigators of non-protective IL-4 and depleting this granulocyte population improved fungal clearance in CCR2-/- animals. The deleterious impact of eosinophilia on mycosis was also recapitulated in transgenic animals overexpressing eosinophils. Mechanistic examination of IL-4 induction revealed that phagocytosis of H. capsulatum via the pattern recognition receptor complement receptor (CR) 3 triggered the heightened IL-4 response in murine eosinophils. This phenomenon was conserved in human eosinophils; exposure of cells to the fungal pathogen elicited a robust IL-4 response. Thus, our findings elucidate a detrimental attribute of eosinophil biology in fungal infections that could potentially trigger a collapse in host defenses by instigating type II immunity.


Subject(s)
Eosinophils/immunology , Histoplasma/immunology , Histoplasmosis/immunology , Interleukin-4/metabolism , Receptors, CCR2/metabolism , Animals , Antigens, Fungal/immunology , Cells, Cultured , Eosinophils/microbiology , Humans , Immune Evasion , Immunity, Innate , Macrophage-1 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Receptors, CCR2/genetics
10.
Allergy ; 71(5): 611-20, 2016 05.
Article in English | MEDLINE | ID: mdl-26799684

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic disease characterized clinically by symptoms of esophageal dysfunction and histologically by eosinophil-predominant inflammation. EoE is frequently associated with concomitant atopic diseases and immunoglobulin E (IgE) sensitization to food allergens in children as well as to aeroallergens and cross-reactive plant allergen components in adults. Patients with EoE respond well to elemental and empirical food elimination diets. Recent research has, however, indicated that the pathogenesis of EoE is distinct from IgE-mediated food allergy. In this review, we discuss the individual roles of epithelial barrier defects, dysregulated innate and adaptive immune responses, and of microbiota in the pathogenesis of EoE. Although food has been recognized as a trigger factor of EoE, the mechanism by which it initiates or facilitates eosinophilic inflammation appears to be largely independent of IgE and needs to be further investigated. Understanding the pathogenic role of food in EoE is a prerequisite for the development of specific diagnostic tools and targeted therapeutic procedures.


Subject(s)
Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/etiology , Food Hypersensitivity/diagnosis , Food Hypersensitivity/etiology , Allergens/immunology , Anti-Asthmatic Agents/therapeutic use , Eosinophilic Esophagitis/drug therapy , Eosinophilic Esophagitis/metabolism , Epithelium/immunology , Epithelium/metabolism , Epithelium/pathology , Food/adverse effects , Food Hypersensitivity/metabolism , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Immunity, Innate , Immunoglobulin E/immunology , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Omalizumab/therapeutic use , Skin/immunology , Skin/metabolism , Skin/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Treatment Outcome
11.
Mucosal Immunol ; 9(3): 744-56, 2016 05.
Article in English | MEDLINE | ID: mdl-26462420

ABSTRACT

Eosinophilic esophagitis (EoE) is an allergic inflammatory disease of the esophagus featuring increased esophageal interleukin-13 (IL-13) levels and impaired barrier function. Herein, we investigated leucine-rich repeat-containing protein 31 (LRRC31) in human EoE esophageal tissue and IL-13-treated esophageal epithelial cells. LRRC31 had basal mRNA expression in colonic and airway mucosal epithelium. Esophageal LRRC31 mRNA and protein increased in active EoE and strongly correlated with esophageal eosinophilia and IL13 and CCL26 (chemokine (C-C motif) ligand 26) mRNA expression. IL-13 treatment increased LRRC31 mRNA and protein in air-liquid interface-differentiated esophageal epithelial cells (EPC2s). At baseline, differentiated LRRC31-overexpressing EPC2s had increased barrier function (1.9-fold increase in transepithelial electrical resistance (P<0.05) and 2.8-fold decrease in paracellular flux (P<0.05)). RNA sequencing analysis of differentiated LRRC31-overexpressing EPC2s identified 38 dysregulated genes (P<0.05), including five kallikrein (KLK) serine proteases. Notably, differentiated LRRC31-overexpressing EPC2s had decreased KLK expression and activity, whereas IL-13-treated, differentiated LRRC31 gene-silenced EPC2s had increased KLK expression and suprabasal epithelial detachment. We identified similarly dysregulated KLK expression in the esophagus of patients with active EoE and in IL-13-treated esophageal epithelial cells. We propose that LRRC31 is induced by IL-13 and modulates epithelial barrier function, potentially through KLK regulation.


Subject(s)
Eosinophilic Esophagitis/immunology , Epithelium/metabolism , Esophagus/immunology , Interleukin-13/metabolism , Kallikreins/metabolism , Nuclear Proteins/metabolism , Proteins/metabolism , Adult , Biological Transport , Cell Differentiation , Cell Line , Dextrans/pharmacokinetics , Electric Impedance , Epithelium/pathology , Esophagus/pathology , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Gene Expression Regulation , Humans , Interleukin-13/genetics , Kallikreins/genetics , Leucine-Rich Repeat Proteins , Nuclear Proteins/genetics , Proteins/genetics , RNA, Small Interfering/genetics
12.
Mucosal Immunol ; 8(3): 464-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25807184

ABSTRACT

Eosinophils, multifunctional cells that contribute to both innate and adaptive immunity, are involved in the initiation, propagation, and resolution of immune responses, including tissue repair. They achieve this multifunctionality by expression of a diverse set of activation receptors, including those that directly recognize pathogens and opsonized targets, and by their ability to store and release preformed cytotoxic mediators that participate in host defense, to produce a variety of de novo pleotropic mediators and cytokines, and to interact directly and indirectly with diverse cell types, including adaptive and innate immunocytes and structural cells. Herein, we review the basic biology of eosinophils and then focus on new emerging concepts about their role in mucosal immune homeostasis, particularly maintenance of intestinal IgA. We review emerging data about their development and regulation and describe new concepts concerning mucosal eosinophilic diseases. We describe recently developed therapeutic strategies to modify eosinophil levels and function and provide collective insight about the beneficial and detrimental functions of these enigmatic cells.


Subject(s)
Asthma/immunology , Enteritis/immunology , Eosinophilia/immunology , Eosinophilic Esophagitis/immunology , Eosinophils/immunology , Gastritis/immunology , Immunity, Mucosal , Inflammatory Bowel Diseases/immunology , Adaptive Immunity , Animals , Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Asthma/pathology , Cytokines/genetics , Cytokines/immunology , Enteritis/drug therapy , Enteritis/pathology , Eosinophilia/drug therapy , Eosinophilia/pathology , Eosinophilic Esophagitis/drug therapy , Eosinophilic Esophagitis/pathology , Eosinophils/drug effects , Eosinophils/pathology , Gastritis/drug therapy , Gastritis/pathology , Gene Expression , Homeostasis/drug effects , Homeostasis/immunology , Humans , Immunity, Innate , Immunologic Factors/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology
13.
Mucosal Immunol ; 8(4): 930-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25563499

ABSTRACT

Eosinophils are multifunctional leukocytes that reside in the gastrointestinal (GI) lamina propria, where their basal function remains largely unexplored. In this study, by examining mice with a selective deficiency of systemic eosinophils (by lineage ablation) or GI eosinophils (eotaxin-1/2 double deficient or CC chemokine receptor 3 deficient), we show that eosinophils support immunoglobulin A (IgA) class switching, maintain intestinal mucus secretions, affect intestinal microbial composition, and promote the development of Peyer's patches. Eosinophil-deficient mice showed reduced expression of mediators of secretory IgA production, including intestinal interleukin 1ß (IL-1ß), inducible nitric oxide synthase, lymphotoxin (LT) α, and LT-ß, and reduced levels of retinoic acid-related orphan receptor gamma t-positive (ROR-γt(+)) innate lymphoid cells (ILCs), while maintaining normal levels of APRIL (a proliferation-inducing ligand), BAFF (B cell-activating factor of the tumor necrosis factor family), and TGF-ß (transforming growth factor ß). GI eosinophils expressed a relatively high level of IL-1ß, and IL-1ß-deficient mice manifested the altered gene expression profiles observed in eosinophil-deficient mice and decreased levels of IgA(+) cells and ROR-γt(+) ILCs. On the basis of these collective data, we propose that eosinophils are required for homeostatic intestinal immune responses including IgA production and that their affect is mediated via IL-1ß in the small intestine.


Subject(s)
Eosinophils/immunology , Eosinophils/metabolism , Homeostasis , Immunoglobulin A/biosynthesis , Interleukin-1beta/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Adoptive Transfer , Animals , Cell Count , Gastrointestinal Microbiome , Gene Expression , Immune Tolerance , Immunoglobulin A, Secretory/biosynthesis , Interleukin-1beta/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Small/microbiology , Lymphotoxin-alpha/genetics , Lymphotoxin-beta/genetics , Mice , Mice, Knockout , Mucus/metabolism , Peyer's Patches/immunology , Peyer's Patches/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism
14.
Mucosal Immunol ; 8(4): 785-98, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25389033

ABSTRACT

Although interleukin (IL)-13 and neurotrophins are functionally important for the pathogenesis of immune responses, the interaction of these pathways has not been explored. Herein, by interrogating IL-13-induced responses in human epithelial cells we show that neurotrophic tyrosine kinase receptor, type 1 (NTRK1), a cognate, high-affinity receptor for nerve growth factor (NGF), is an early transcriptional IL-13 target. Induction of NTRK1 was accompanied by accumulation of activating epigenetic marks in the promoter; transcriptional and epigenetic changes were signal transducer and activator of transcription 6 dependent. Using eosinophilic esophagitis as a model for human allergic inflammation, we found that NTRK1 was increased in inflamed tissue and dynamically expressed as a function of disease activity and that the downstream mediator of NTRK1 signaling early growth response 1 protein was elevated in allergic inflammatory tissue compared with control tissue. Unlike NTRK1, its ligand NGF was constitutively expressed in control and disease states, indicating that IL-13-stimulated NTRK1 induction is a limiting factor in pathway activation. In epithelial cells, NGF and IL-13 synergistically induced several target genes, including chemokine (C-C motif) ligand 26 (eotaxin-3). In summary, we have demonstrated that IL-13 confers epithelial cell responsiveness to NGF by regulating NTRK1 levels by a transcriptional and epigenetic mechanism and that this process likely contributes to allergic inflammation.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Hypersensitivity/genetics , Hypersensitivity/metabolism , Interleukin-13/metabolism , Receptor, trkA/genetics , Transcription, Genetic , Cluster Analysis , Early Growth Response Protein 1/metabolism , Eosinophilic Esophagitis/genetics , Eosinophilic Esophagitis/metabolism , Eosinophilic Esophagitis/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Silencing , Humans , Interleukin-13/pharmacology , Nerve Growth Factor/pharmacology , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism
15.
Genes Immun ; 15(6): 361-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24920534

ABSTRACT

Eosinophilic esophagitis (EoE) is an allergic inflammatory disorder of the esophagus that is compounded by genetic predisposition and hypersensitivity to environmental antigens. Using high-density oligonucleotide expression chips, a disease-specific esophageal transcript signature was identified and was shown to be largely reversible with therapy. In an effort to expand the molecular signature of EoE, we performed RNA sequencing on esophageal biopsies from healthy controls and patients with active EoE and identified a total of 1607 significantly dysregulated transcripts (1096 upregulated, 511 downregulated). When clustered by raw expression levels, an abundance of immune cell-specific transcripts are highly induced in EoE but expressed at low (or undetectable) levels in healthy controls. Moreover, 66% of the gene signature identified by RNA sequencing was previously unrecognized in the EoE transcript signature by microarray-based expression profiling and included several long non-coding RNAs (lncRNA), an emerging class of transcriptional regulators. The lncRNA BRAF-activated non-protein coding RNA (BANCR) was upregulated in EoE and induced in interleukin-13 (IL-13)-treated primary esophageal epithelial cells. Repression of BANCR significantly altered the expression of IL-13-induced proinflammatory genes. Together, these data comprise new potential biomarkers of EoE and demonstrate a novel role for lncRNAs in EoE and IL-13-associated responses.


Subject(s)
Eosinophilic Esophagitis/genetics , Oligonucleotide Array Sequence Analysis/methods , Sequence Analysis, RNA/methods , Transcriptome , Cell Line , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Interleukin-13/pharmacology , RNA Interference , RNA, Untranslated/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
16.
Mucosal Immunol ; 7(3): 718-29, 2014 May.
Article in English | MEDLINE | ID: mdl-24220297

ABSTRACT

The desmosomal cadherin desmoglein-1 (DSG1) is an essential intercellular adhesion molecule that is altered in various human cutaneous disorders; however, its regulation and function in allergic disease remains unexplored. Herein, we demonstrate a specific reduction in DSG1 in esophageal biopsies from patients with eosinophilic esophagitis (EoE), an emerging allergic disorder characterized by chronic inflammation within the esophageal mucosa. Further, we show that DSG1 gene silencing weakens esophageal epithelial integrity, and induces cell separation and impaired barrier function (IBF) despite high levels of desmoglein-3. Moreover, DSG1 deficiency induces transcriptional changes that partially overlap with the transcriptome of inflamed esophageal mucosa; notably, periostin (POSTN), a multipotent pro-inflammatory extracellular matrix molecule, is the top induced overlapping gene. We further demonstrate that IBF is a pathological feature in EoE, which can be partially induced through the downregulation of DSG1 by interleukin-13 (IL-13). Taken together, these data identify a functional role for DSG1 and its dysregulation by IL-13 in the pathophysiology of EoE and suggest that the loss of DSG1 may potentiate allergic inflammation through the induction of pro-inflammatory mediators such as POSTN.


Subject(s)
Desmoglein 1/metabolism , Eosinophilic Esophagitis/immunology , Eosinophilic Esophagitis/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Cell Differentiation/genetics , Cluster Analysis , Desmoglein 1/deficiency , Desmoglein 1/genetics , Eosinophilic Esophagitis/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Immunity, Innate/genetics , Immunohistochemistry , Interleukin-13/metabolism , Models, Biological , Mucous Membrane/pathology , Transcription, Genetic
17.
Mucosal Immunol ; 5(4): 388-96, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22453679

ABSTRACT

Interleukin 13 (IL-13)-induced epithelial gene and protein expression changes are central to the pathogenesis of multiple allergic diseases. Herein, using human esophageal squamous and bronchial columnar epithelial cells, we identified microRNAs (miRNAs) that were differentially regulated after IL-13 stimulation. Among the IL-13-regulated miRNAs, miR-375 showed a conserved pattern of downregulation. Furthermore, miR-375 was downregulated in the lung of IL-13 lung transgenic mice. We subsequently analyzed miR-375 levels in a human disease characterized by IL-13 overproduction--the allergic disorder eosinophilic esophagitis (EE)--and observed downregulation of miR-375 in EE patient samples compared with control patients. MiR-375 expression levels reflected disease activity, normalized with remission, and inversely correlated with the degree of allergic inflammation. Using a lentiviral strategy and whole-transcriptome analysis in epithelial cells, miR-375 overexpression was sufficient to markedly modify IL-13-associated immunoinflammatory pathways in epithelial cells in vitro, further substantiating interactions between miR-375 and IL-13. Taken together, our results support a key role of miRNAs, particularly miR-375, in regulating and fine-tuning IL-13-mediated responses.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Immunologic Factors/pharmacology , Interleukin-13/pharmacology , MicroRNAs/genetics , Transcriptome , Animals , Cell Line , Cluster Analysis , Eosinophilic Esophagitis/genetics , Esophagus/metabolism , Gene Expression Profiling , Humans , Mice , Mice, Transgenic , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
18.
Child Care Health Dev ; 38(4): 477-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21671982

ABSTRACT

BACKGROUND AND AIMS: Current research outcomes in paediatric eosinophilic oesophagitis (EoE) are directed towards histological improvement with no attention to health-related quality of life (HRQOL). The primary objective of this study was to identify key patient-reported and parent proxy outcome elements of EoE disease-specific HRQOL. METHODS: The research team comprised clinical allergists and gastroenterologists with expertise in paediatric EoE as well as two PhD psychologists with extensive experience in qualitative research. Focused interview techniques were adapted from the Pediatric Quality of Life Inventory 4.0™ methodology and the consolidated criteria for reporting qualitative research. A semi-structured interview guide of open-ended questions was developed, and extensive review of audio-taped transcripts was performed. RESULTS: A total of 42 focus interviews were conducted. Child self-reports were obtained for patients in the 5-7, 8-12 and 13-18 years of age groups, and parent proxy reports were obtained in the 2-4, 5-7, 8-12 and 13-18 years of age groups. We discovered that patients and parents often had different concerns, illustrating unique aspects of EoE-specific HRQOL that were not captured in generic HRQOL instruments. Specific themes that emerged from these interviews included, but are not limited to: feelings of being different than family and peers, diet and medication adherence, difficulties with eating food and worry about symptoms and illness. CONCLUSION: Paediatric EoE patient and parent proxy interviews revealed many EoE-specific aspects of HRQOL that are not captured in generic HRQOL instruments. Outcome measures that reflect patient- and parent proxy-reported HRQOL are a critical need in paediatric EoE.


Subject(s)
Attitude to Health , Eosinophilic Esophagitis/rehabilitation , Quality of Life , Activities of Daily Living , Adolescent , Child , Child, Preschool , Communication , Eosinophilic Esophagitis/physiopathology , Eosinophilic Esophagitis/psychology , Eosinophilic Esophagitis/therapy , Feeding Behavior , Female , Humans , Interpersonal Relations , Male , Ohio , Psychometrics , Schools , Treatment Outcome
19.
Gut ; 59(1): 12-20, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19528036

ABSTRACT

BACKGROUND: Eosinophilic oesophagitis (EO) is an emerging yet increasingly prevalent disorder characterised by a dense and selective eosinophilic infiltration of the oesophageal wall. While EO is considered an atopic disease primarily triggered by food antigens, disparities between standard allergen testing and clinical responses to exclusion diets suggest the participation of distinct antigen-specific immunoglobulin E (IgE) in the pathophysiology of EO. AIM: To find evidence for a local IgE response. METHODS: Endoscopic biopsies of the distal oesophagus of atopic and non-atopic EO and control individuals (CTL) were processed for immunohistochemistry and immunofluorescence to assess the presence of B cells, mast cells, and IgE-bearing cells. Oesophageal RNA was analysed for the expression of genes involved in B cell activation, class switch recombination to IgE and IgE production, including germline transcripts (GLTs), activation-induced cytidine deaminase (AID), IgE heavy chain (Cepsilon) and mature IgE mRNA using polymerase chain reaction and microarray analysis. RESULTS: Regardless of atopy, EO showed increased density of B cells (p<0.05) and of IgE-bounded mast cells compared to CTL. Both EO and CTL expressed muGLT, epsilonGLT, gamma4GLT, AID, Cepsilon and IgE mRNA. However, the frequency of expression of total GLTs (p = 0.002), epsilonGLT (p = 0.024), and Cepsilon (p = 0.0003) was significantly higher in EO than in CTL, independent of the atopic status. CONCLUSION: These results support the heretofore unproven occurrence of both local immunoglobulin class switching to IgE and IgE production in the oesophageal mucosa of EO patients. Sensitisation and activation of mast cells involving local IgE may therefore critically contribute to disease pathogenesis.


Subject(s)
B-Lymphocytes/immunology , Eosinophilia/immunology , Esophagitis/immunology , Hypersensitivity, Immediate/immunology , Immunoglobulin E/biosynthesis , Adolescent , Cell Count , Child , Child, Preschool , Esophagus/immunology , Female , Humans , Immunoglobulin E/genetics , Interleukin-13/biosynthesis , Interleukin-4/biosynthesis , Lymphocyte Activation/immunology , Male , Mast Cells/immunology , Mucous Membrane/immunology , Oligonucleotide Array Sequence Analysis/methods , Polymerase Chain Reaction/methods , RNA, Messenger/genetics , Retrospective Studies , Transcription, Genetic
20.
Mucosal Immunol ; 1(4): 289-96, 2008 Jul.
Article in English | MEDLINE | ID: mdl-19079190

ABSTRACT

Periostin is an extracellular matrix protein that has been primarily studied in the context of the heart, where it has been shown to promote cardiac repair and remodeling. In this study, we focused on the role of periostin in an allergic eosinophilic inflammatory disease (eosinophilic esophagitis (EE)) known to involve extensive tissue remodeling. Periostin was indeed markedly overexpressed (35-fold) in the esophagus of EE patients, particularly in the papillae, compared with control individuals. Periostin expression was downstream from transforming growth factor-beta and interleukin-13, as these cytokines were elevated in EE esophageal samples and markedly induced periostin production by primary esophageal fibroblasts (107- and 295-fold, respectively, at 10 ng ml(-1)). A functional role for periostin in eliciting esophageal eosinophilia was demonstrated, as periostin-null mice had a specific defect in allergen-induced eosinophil recruitment to the lungs and esophagus (66 and 72% decrease, respectively). Mechanistic analyses revealed that periostin increased (5.8-fold) eosinophil adhesion to fibronectin. As such, these findings extend the involvement of periostin to esophagitis and uncover a novel role for periostin in directly regulating leukocyte (eosinophil) accumulation in T helper type 2-associated mucosal inflammation in both mice and humans.


Subject(s)
Cell Adhesion Molecules/physiology , Eosinophils/physiology , Esophagitis/immunology , Hypersensitivity/immunology , Pulmonary Eosinophilia/immunology , Animals , Asthma/immunology , Asthma/pathology , Cell Adhesion/physiology , Cell Adhesion Molecules/genetics , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Eosinophils/immunology , Esophagitis/pathology , Esophagus/metabolism , Esophagus/pathology , Fibroblasts/physiology , Humans , Hypersensitivity/pathology , Interleukin-13/immunology , Lung/immunology , Lung/pathology , Mice , Mice, Knockout , Pulmonary Eosinophilia/pathology , Rhinitis/immunology , Rhinitis/pathology , Transforming Growth Factor beta/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...