Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Pediatr Gastroenterol Nutr ; 76(3): 347-354, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36525669

ABSTRACT

OBJECTIVE: The aim of the study was to determine the mucosal microbiota associated with eosinophilic esophagitis (EoE) and eosinophilic gastritis (EoG) in a geographically diverse cohort of patients compared to controls. METHODS: We conducted a prospective study of individuals with eosinophilic gastrointestinal disease (EGID) in the Consortium of Eosinophilic Gastrointestinal Disease Researchers, including pediatric and adult tertiary care centers. Eligible individuals had clinical data, mucosal biopsies, and stool collected. Total bacterial load was determined from mucosal biopsy samples by quantitative polymerase chain reaction (PCR). Community composition was determined by small subunit rRNA gene amplicons. RESULTS: One hundred thirty-nine mucosal biopsies were evaluated corresponding to 93 EoE, 17 EoG, and 29 control specimens (18 esophageal) from 10 sites across the United States. Dominant community members across disease activity differed significantly. When comparing EoE and EoG with controls, the dominant taxa in individuals with EGIDs was increased ( Streptococcus in esophagus; Prevotella in stomach). Specific taxa were associated with active disease for both EoE ( Streptococcus , Gemella ) and EoG ( Leptotrichia ), although highly individualized communities likely impacted statistical testing. Alpha diversity metrics were similar across groups, but with high variability among individuals. Stool analyses did not correlate with bacterial communities found in mucosal biopsy samples and was similar in patients and controls. CONCLUSIONS: Dominant community members ( Streptococcus for EoE, Prevotella for EoG) were different in the mucosal biopsies but not stool of individuals with EGIDs compared to controls; taxa associated with EGIDs were highly variable across individuals. Further study is needed to determine if therapeutic interventions contribute to the observed community differences.


Subject(s)
Eosinophilic Esophagitis , Microbiota , Adult , Humans , Child , Eosinophilic Esophagitis/pathology , Prospective Studies
2.
Front Cell Dev Biol ; 9: 656867, 2021.
Article in English | MEDLINE | ID: mdl-33937256

ABSTRACT

Macrophages are pivotal effectors of host immunity and regulators of tissue homeostasis. Understanding of human macrophage biology has been hampered by the lack of reliable and scalable models for cellular and genetic studies. Human induced pluripotent stem cell (hiPSC)-derived monocytes and macrophages, as an unlimited source of subject genotype-specific cells, will undoubtedly play an important role in advancing our understanding of macrophage biology and implication in human diseases. In this study, we present a fully optimized differentiation protocol of hiPSC-derived monocytes and granulocyte-macrophage colony-stimulating factor (GM-CSF) or macrophage colony-stimulating factor (M-CSF). We present characterization of iPSC-derived myeloid lineage cells at phenotypic, functional, and transcriptomic levels, in comparison with corresponding subsets of peripheral blood-derived cells. We also highlight the application of hiPSC-derived monocytes and macrophages as a gene-editing platform for functional validation in research and drug screening, and the study also provides a reference for cell therapies.

3.
J Pediatr Gastroenterol Nutr ; 72(4): 520-527, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33394582

ABSTRACT

BACKGROUND: Acid blockade is commonly prescribed in patients with cystic fibrosis (CF). Growing concerns, however, exist about its possible role in the pathophysiology of pulmonary infections. We aimed to investigate if acid blockade alters esophageal and respiratory microbiota leading to dysbiosis and inflammation. METHODS: We performed a cross sectional study of children with CF who were either prescribed acid blockade or not. Samples from the gastrointestinal and respiratory tracts were obtained and microbiome analyzed. Mixed effect models were used to compare outcomes between cohorts and across sampling sites. A random subject intercept was included to account for the multiple sampling sites per individual. RESULTS: A cohort of 25 individuals, 44% girls with median age of 13.8 years [IQR 11.2--14.8] were enrolled. Alpha diversity, total bacterial load, and beta diversity were similar across anatomic compartments, across the upper gastrointestinal tract, and in respiratory samples. Similar alpha diversity, total bacterial load, and beta diversity results were also observed when comparing individuals on versus those off acid blockade. IL-8 was elevated in the distal versus proximal esophagus in the whole cohort (P < 0.01). IL-8 concentrations were similar in the distal esophagus in patients on and off acid blockade, but significantly greater in the proximal esophagus of subjects on treatment (P < 0.01). CONCLUSIONS: On the basis of these data, acid blockade use does not appear to influence the microbiome of the aerodigestive tract in children with cystic fibrosis suggesting a complex interplay between these medications and the bacterial composition of the esophagus and lung.


Subject(s)
Cystic Fibrosis , Microbiota , Adolescent , Bacteria , Child , Cross-Sectional Studies , Cystic Fibrosis/drug therapy , Dysbiosis , Female , Humans , Male
4.
Nat Commun ; 9(1): 1393, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29643332

ABSTRACT

In infants intolerant of enteral feeding because of intestinal disease, parenteral nutrition may be associated with cholestasis, which can progress to end-stage liver disease. Here we show the function of hepatic macrophages and phytosterols in parenteral nutrition-associated cholestasis (PNAC) pathogenesis using a mouse model that recapitulates the human pathophysiology and combines intestinal injury with parenteral nutrition. We combine genetic, molecular, and pharmacological approaches to identify an essential function of hepatic macrophages and IL-1ß in PNAC. Pharmacological antagonism of  IL-1 signaling or genetic deficiency in CCR2, caspase-1 and caspase-11, or IL-1 receptor (which binds both IL-1α and IL-1ß) prevents PNAC in mice. IL-1ß increases hepatocyte NF-κB signaling, which interferes with farnesoid X receptor and liver X receptor bonding to respective promoters of canalicular bile and sterol transporter genes (Abcc2, Abcb11, and Abcg5/8), resulting in transcriptional suppression and subsequent cholestasis. Thus, hepatic macrophages, IL-1ß, or NF-κB may be targets for restoring bile and sterol transport to treat PNAC.


Subject(s)
Cholestasis/genetics , Interleukin-1beta/genetics , Liver/immunology , Macrophages/immunology , NF-kappa B/genetics , Receptors, CCR2/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11/immunology , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/immunology , Animals , Caspase 1/genetics , Caspase 1/immunology , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Cholestasis/etiology , Cholestasis/immunology , Cholestasis/pathology , Disease Models, Animal , Gene Expression Regulation , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Infant, Newborn , Interleukin-1beta/immunology , Lipoproteins/genetics , Lipoproteins/immunology , Liver/pathology , Liver X Receptors/genetics , Liver X Receptors/immunology , Macrophages/pathology , Male , Mice , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/immunology , NF-kappa B/immunology , Parenteral Nutrition/adverse effects , Receptors, CCR2/deficiency , Receptors, CCR2/immunology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Signal Transduction
6.
Cell Host Microbe ; 19(3): 388-99, 2016 03 09.
Article in English | MEDLINE | ID: mdl-26962947

ABSTRACT

Maternal infection during pregnancy is associated with adverse outcomes for the fetus, including postnatal cognitive disorders. However, the underlying mechanisms are obscure. We find that bacterial cell wall peptidoglycan (CW), a universal PAMP for TLR2, traverses the murine placenta into the developing fetal brain. In contrast to adults, CW-exposed fetal brains did not show any signs of inflammation or neuronal death. Instead, the neuronal transcription factor FoxG1 was induced, and neuroproliferation leading to a 50% greater density of neurons in the cortical plate was observed. Bacterial infection of pregnant dams, followed by antibiotic treatment, which releases CW, yielded the same result. Neuroproliferation required TLR2 and was recapitulated in vitro with fetal neuronal precursor cells and TLR2/6, but not TLR2/1, ligands. The fetal neuroproliferative response correlated with abnormal cognitive behavior in CW-exposed pups following birth. Thus, the bacterial CW-TLR2 signaling axis affects fetal neurodevelopment and may underlie postnatal cognitive disorders.


Subject(s)
Bacterial Infections/complications , Brain/pathology , Cell Proliferation/drug effects , Cognition Disorders/physiopathology , Maternal-Fetal Exchange , Neurons/drug effects , Peptidoglycan/metabolism , Animals , Behavior, Animal , Brain/drug effects , Cognition Disorders/chemically induced , Female , Mice , Neurons/physiology , Pregnancy , Toll-Like Receptor 2/metabolism
7.
PLoS One ; 10(5): e0128346, 2015.
Article in English | MEDLINE | ID: mdl-26020633

ABSTRACT

OBJECTIVE: The microbiome has been implicated in the pathogenesis of a number of allergic and inflammatory diseases. The mucosa affected by eosinophilic esophagitis (EoE) is composed of a stratified squamous epithelia and contains intraepithelial eosinophils. To date, no studies have identified the esophageal microbiome in patients with EoE or the impact of treatment on these organisms. The aim of this study was to identify the esophageal microbiome in EoE and determine whether treatments change this profile. We hypothesized that clinically relevant alterations in bacterial populations are present in different forms of esophagitis. DESIGN: In this prospective study, secretions from the esophageal mucosa were collected from children and adults with EoE, Gastroesophageal Reflux Disease (GERD) and normal mucosa using the Esophageal String Test (EST). Bacterial load was determined using quantitative PCR. Bacterial communities, determined by 16S rRNA gene amplification and 454 pyrosequencing, were compared between health and disease. RESULTS: Samples from a total of 70 children and adult subjects were examined. Bacterial load was increased in both EoE and GERD relative to normal subjects. In subjects with EoE, load was increased regardless of treatment status or degree of mucosal eosinophilia compared with normal. Haemophilus was significantly increased in untreated EoE subjects as compared with normal subjects. Streptococcus was decreased in GERD subjects on proton pump inhibition as compared with normal subjects. CONCLUSIONS: Diseases associated with mucosal eosinophilia are characterized by a different microbiome from that found in the normal mucosa. Microbiota may contribute to esophageal inflammation in EoE and GERD.


Subject(s)
Eosinophilic Esophagitis/microbiology , Gastroesophageal Reflux/microbiology , Microbiota , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Adolescent , Adult , Child , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged
8.
Gut ; 64(8): 1236-47, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25209655

ABSTRACT

OBJECTIVE: Eosinophils reside in the colonic mucosa and increase significantly during disease. Although a number of studies have suggested that eosinophils contribute to the pathogenesis of GI inflammation, the expanding scope of eosinophil-mediated activities indicate that they also regulate local immune responses and modulate tissue inflammation. We sought to define the impact of eosinophils that respond to acute phases of colitis in mice. DESIGN: Acute colitis was induced in mice by administration of dextran sulfate sodium, 2,4,6-trinitrobenzenesulfonic acid or oxazolone to C57BL/6J (control) or eosinophil deficient (PHIL) mice. Eosinophils were also depleted from mice using antibodies against interleukin (IL)-5 or by grafting bone marrow from PHIL mice into control mice. Colon tissues were collected and analysed by immunohistochemistry, flow cytometry and reverse transcription PCR; lipids were analysed by mass spectroscopy. RESULTS: Eosinophil-deficient mice developed significantly more severe colitis, and their colon tissues contained a greater number of neutrophils, than controls. This compensatory increase in neutrophils was accompanied by increased levels of the chemokines CXCL1 and CXCL2, which attract neutrophils. Lipidomic analyses of colonic tissue from eosinophil-deficient mice identified a deficiency in the docosahexaenoic acid-derived anti-inflammatory mediator 10, 17- dihydroxydocosahexaenoic acid (diHDoHE), namely protectin D1 (PD1). Administration of an exogenous PD1-isomer (10S, 17S-DiHDoHE) reduced the severity of colitis in eosinophil-deficient mice. The PD1-isomer also attenuated neutrophil infiltration and reduced levels of tumour necrosis factor-α, IL-1ß, IL-6 and inducible NO-synthase in colons of mice. Finally, in vitro assays identified a direct inhibitory effect of PD1-isomer on neutrophil transepithelial migration. CONCLUSIONS: Eosinophils exert a protective effect in acute mouse colitis, via production of anti-inflammatory lipid mediators.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis/pathology , Eosinophils/pathology , Inflammation/pathology , Animals , Colitis/drug therapy , Colitis/immunology , Disease Models, Animal , Eosinophils/immunology , Inflammation/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL
9.
PLoS One ; 9(10): e110396, 2014.
Article in English | MEDLINE | ID: mdl-25329595

ABSTRACT

BACKGROUND: Parenteral nutrition (PN) has been a life-saving treatment in infants intolerant of enteral feedings. However, PN is associated with liver injury (PN Associated Liver Injury: PNALI) in a significant number of PN-dependent infants. We have previously reported a novel PNALI mouse model in which PN infusion combined with intestinal injury results in liver injury. In this model, lipopolysaccharide activation of toll-like receptor 4 signaling, soy oil-derived plant sterols, and pro-inflammatory activation of Kupffer cells (KCs) played key roles. The objective of this study was to explore changes in the intestinal microbiome associated with PNALI. METHODOLOGY AND PRINCIPAL FINDINGS: Microbiome analysis in the PNALI mouse identified specific alterations within colonic microbiota associated with PNALI and further association of these communities with the lipid composition of the PN solution. Intestinal inflammation or soy oil-based PN infusion alone (in the absence of enteral feeds) caused shifts within the gut microbiota. However, the combination resulted in accumulation of a specific taxon, Erysipelotrichaceae (23.8% vs. 1.7% in saline infused controls), in PNALI mice. Moreover, PNALI was markedly attenuated by enteral antibiotic treatment, which also was associated with significant reduction of Erysipelotrichaceae (0.6%) and a Gram-negative constituent, the S24-7 lineage of Bacteroidetes (53.5% in PNALI vs. 0.8%). Importantly, removal of soy oil based-lipid emulsion from the PN solution resulted in significant reduction of Erysipelotrichaceae as well as attenuation of PNALI. Finally, addition of soy-derived plant sterol (stigmasterol) to fish oil-based PN restored Erysipelotrichaceae abundance and PNALI. CONCLUSIONS: Soy oil-derived plant sterols and the associated specific bacterial groups in the colonic microbiota are associated with PNALI. Products from these bacteria may directly trigger activation of KCs and promote PNALI. Furthermore, the results indicate that lipid modification of PN solutions may alter specific intestinal bacterial species associated with PNALI, and thus suggest strategies for management of PNALI.


Subject(s)
Intestines/microbiology , Liver/injuries , Liver/microbiology , Microbiota , Parenteral Nutrition/adverse effects , Animals , Disease Models, Animal , Inflammation/etiology , Inflammation/immunology , Inflammation/microbiology , Intestines/drug effects , Intestines/immunology , Kupffer Cells/drug effects , Liver/drug effects , Liver/immunology , Male , Mice , Plant Oils/pharmacology , Glycine max/chemistry
10.
Gut ; 63(1): 43-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23161496

ABSTRACT

OBJECTIVE: Eosinophilic oesophagitis (EoE) is a chronic inflammatory condition of the oesophagus with limited treatment options. No previous transgenic model has specifically targeted the oesophageal mucosa to induce oesophageal eosinophilia. DESIGN: We developed a mouse model that closely resembles EoE by utilising oxazolone haptenation in mice with transgenic overexpression of an eosinophil poietic and survival factor (interleukin (IL)-5) in resident squamous oesophageal epithelia. RESULTS: Overexpression of IL-5 in the healthy oesophagus was achieved in transgenic mice (L2-IL5) using the squamous epithelial promoter Epstein-Barr virus ED-L2. Oxazolone-challenged L2-IL5 mice developed dose-dependent pan-oesophageal eosinophilia, including eosinophil microabscess formation and degranulation as well as basal cell hyperplasia. Moreover, oesophagi expressed increased IL-13 and the eosinophil agonist chemokine eotaxin-1. Treatment of these mice with corticosteroids significantly reduced eosinophilia and epithelial inflammation. CONCLUSIONS: L2-IL5 mice provide a novel experimental model that can potentially be used in preclinical testing of EoE-related therapeutics and mechanistic studies identifying pathogenetic features associated with mucosal eosinophilia.


Subject(s)
Disease Models, Animal , Eosinophilic Esophagitis/etiology , Interleukin-5/metabolism , Mice, Transgenic , Animals , Anti-Inflammatory Agents/therapeutic use , Biomarkers/metabolism , Dexamethasone/therapeutic use , Eosinophilic Esophagitis/drug therapy , Eosinophilic Esophagitis/metabolism , Epithelium , Herpesvirus 4, Human , Interleukin-5/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Mice, Transgenic/immunology , Mice, Transgenic/metabolism , Oxazolone , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Viral Regulatory and Accessory Proteins
11.
Pediatr Res ; 73(5): 647-54, 2013 May.
Article in English | MEDLINE | ID: mdl-23385963

ABSTRACT

BACKGROUND: Defensins are antimicrobial peptides expressed on mucosal surfaces that contribute to maintaining intestinal homeostasis by providing innate defense mechanisms for the epithelia. Defensin expression is altered in a number of diseases that affect mucosal surfaces, such as atopic dermatitis, allergic rhinitis, and inflammatory bowel disease. Similar to atopic dermatitis, eosinophilic esophagitis (EoE) is a chronic disease in which the squamous epithelial surface is affected by a similar TH2 microenvironment and eosinophil-predominant inflammation. Therefore, we hypothesized that defensin expression would be decreased in EoE. METHODS: To address this, we measured defensin expression in vitro in cell lines derived from patients with EoE (EoE1-T) or gastroesophageal reflux disease (GERD) (NES-G4T cells) and ex vivo in esophageal mucosal biopsy samples from children with EoE or GERD and control children without esophageal disease. RESULTS: Interleukin-5 induced a decrease in human ß-defensin (hBD) -1 and hBD3 expression in EoE1-T but not in NES-G4T cells. Compared with esophageal biopsy specimens from GERD and control children, specimens from EoE pediatric patients revealed a significant decrease in mRNA and protein expression for hBD1 and hBD3. CONCLUSION: Diminished expression of hBD1 and hBD3 may make the esophageal epithelium more susceptible to the development and/or perpetuation of EoE.


Subject(s)
Eosinophilic Esophagitis/metabolism , Esophagus/metabolism , beta-Defensins/metabolism , Adolescent , Adult , Child , Child, Preschool , Humans , Mucous Membrane/metabolism , Young Adult
12.
Gut ; 62(10): 1395-405, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22895393

ABSTRACT

OBJECTIVE: Eosinophil predominant inflammation characterises histological features of eosinophilic oesophagitis (EoE). Endoscopy with biopsy is currently the only method to assess oesophageal mucosal inflammation in EoE. We hypothesised that measurements of luminal eosinophil-derived proteins would correlate with oesophageal mucosal inflammation in children with EoE. DESIGN: The Enterotest diagnostic device was used to develop an oesophageal string test (EST) as a minimally invasive clinical device. EST samples and oesophageal mucosal biopsies were obtained from children undergoing upper endoscopy for clinically defined indications. Eosinophil-derived proteins including eosinophil secondary granule proteins (major basic protein-1, eosinophil-derived neurotoxin, eosinophil cationic protein, eosinophil peroxidase) and Charcot-Leyden crystal protein/galectin-10 were measured by ELISA in luminal effluents eluted from ESTs and extracts of mucosal biopsies. RESULTS: ESTs were performed in 41 children with active EoE (n=14), EoE in remission (n=8), gastro-oesophageal reflux disease (n=4) and controls with normal oesophagus (n=15). EST measurement of eosinophil-derived protein biomarkers significantly distinguished between children with active EoE, treated EoE in remission, gastro-oesophageal reflux disease and normal oesophagus. Levels of luminal eosinophil-derived proteins in EST samples significantly correlated with peak and mean oesophageal eosinophils/high power field (HPF), eosinophil peroxidase indices and levels of the same eosinophil-derived proteins in extracts of oesophageal biopsies. CONCLUSIONS: The presence of eosinophil-derived proteins in luminal secretions is reflective of mucosal inflammation in children with EoE. The EST is a novel, minimally invasive device for measuring oesophageal eosinophilic inflammation in children with EoE.


Subject(s)
Eosinophilic Esophagitis/diagnosis , Esophagus/metabolism , Mucositis/diagnosis , Adolescent , Biomarkers/metabolism , Biopsy , Child , Diagnosis, Differential , Eosinophil Granule Proteins/metabolism , Eosinophilic Esophagitis/metabolism , Eosinophilic Esophagitis/therapy , Esophagus/pathology , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/metabolism , Glycoproteins/metabolism , Humans , Lysophospholipase/metabolism , Mucositis/metabolism , Mucositis/therapy , Mucous Membrane/pathology , Sensitivity and Specificity , Specimen Handling/instrumentation , Specimen Handling/methods , Young Adult
13.
PLoS One ; 7(9): e42938, 2012.
Article in English | MEDLINE | ID: mdl-22957025

ABSTRACT

A growing number of studies implicate the microbiome in the pathogenesis of intestinal inflammation. Previous work has shown that adults with esophagitis related to gastroesophageal reflux disease have altered esophageal microbiota compared to those who do not have esophagitis. In these studies, sampling of the esophageal microbiome was accomplished by isolating DNA from esophageal biopsies obtained at the time of upper endoscopy. The aim of the current study was to identify the esophageal microbiome in pediatric individuals with normal esophageal mucosa using a minimally invasive, capsule-based string technology, the Enterotest™. We used the proximal segment of the Enterotest string to sample the esophagus, and term this the "Esophageal String Test" (EST). We hypothesized that the less invasive EST would capture mucosal adherent bacteria present in the esophagus in a similar fashion as mucosal biopsy. EST samples and mucosal biopsies were collected from children with no esophageal inflammation (n = 15) and their microbiome composition determined by 16S rRNA gene sequencing. Microbiota from esophageal biopsies and ESTs produced nearly identical profiles of bacterial genera and were different from the bacterial contents of samples collected from the nasal and oral cavity. We conclude that the minimally invasive EST can serve as a useful device for study of the esophageal microbiome.


Subject(s)
Esophagus/microbiology , Adolescent , Adult , Biopsy/methods , Child , Endoscopy/methods , Equipment Design , Expressed Sequence Tags , Female , Genes, Bacterial , Genome, Bacterial , Humans , Inflammation , Male , Metagenome , Models, Genetic , RNA, Ribosomal, 16S/metabolism , Sequence Analysis, DNA
14.
J Allergy Clin Immunol ; 129(5): 1387-1396.e7, 2012 May.
Article in English | MEDLINE | ID: mdl-22465212

ABSTRACT

BACKGROUND: Mechanisms underlying esophageal remodeling with subepithelial fibrosis in subjects with eosinophilic esophagitis (EoE) have not been delineated. OBJECTIVES: We sought to explore a role for epithelial mesenchymal transition (EMT) in subjects with EoE and determine whether EMT resolves with treatment. METHODS: Esophageal biopsy specimens from 60 children were immunostained for epithelial (cytokeratin) and mesenchymal (vimentin) EMT biomarkers, and EMT was quantified. Subjects studied had EoE (n = 17), indeterminate EoE (n = 15), gastroesophageal reflux disease (n = 7), or normal esophagus (n = 21). EMT was analyzed for relationships to diagnosis, eosinophil counts, and indices of subepithelial fibrosis, eosinophil peroxidase, and TGF-ß immunostaining. EMT was assessed in pretreatment and posttreatment biopsy specimens from 18 subjects with EoE treated with an elemental diet, 6-food elimination diet, or topical corticosteroids (n = 6 per group). RESULTS: TGF-ß1 treatment of esophageal epithelial cells in vitro for 24 hours induced upregulation of mesenchymal genes characteristic of EMT, including N-cadherin (3.3-fold), vimentin (2.1-fold), and fibronectin (7.5-fold). EMT in esophageal biopsy specimens was associated with EoE (or indeterminate EoE) but not gastroesophageal reflux disease or normal esophagus and was correlated to eosinophil counts (r = 0.691), eosinophil peroxidase (r = 0.738), and TGF-ß (r = 0.520) immunostaining and fibrosis (r = 0.644) indices. EMT resolved with EoE treatments that induced clinicopathologic remission with reduced eosinophil counts. EMT decreased significantly after treatment by 74.1% overall in the 18 treated subjects with EoE; pretreatment versus posttreatment EMT scores were 3.17 ± 0.82 versus 0.82 ± 0.39 (P < .001), with similar decreases within treatment groups. Pretreatment/posttreatment EMT was strongly correlated with eosinophil counts for combined (r = 0.804, P < .001) and individual treatment groups. CONCLUSIONS: EMT likely contributes to subepithelial fibrosis in subjects with EoE and resolves with treatments that decrease esophageal inflammation, and its resolution correlates with decreased numbers of esophageal eosinophils.


Subject(s)
Eosinophilic Esophagitis/pathology , Eosinophils/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Esophagus/pathology , Adolescent , Adrenal Cortex Hormones/therapeutic use , Child , Child, Preschool , Diet Therapy , Disease Progression , Eosinophilic Esophagitis/physiopathology , Eosinophilic Esophagitis/therapy , Eosinophils/pathology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Esophagus/drug effects , Female , Humans , Immunohistochemistry , Infant , Keratins/metabolism , Male , Remission Induction , Transforming Growth Factor beta/metabolism , Vimentin/metabolism
15.
Hepatology ; 55(5): 1518-28, 2012 May.
Article in English | MEDLINE | ID: mdl-22120983

ABSTRACT

UNLABELLED: Infants with intestinal failure who are parenteral nutrition (PN)-dependent may develop cholestatic liver injury and cirrhosis (PN-associated liver injury: PNALI). The pathogenesis of PNALI remains incompletely understood. We hypothesized that intestinal injury with increased intestinal permeability combined with administration of PN promotes lipopolysaccharide (LPS)-Toll-like receptor 4 (TLR4) signaling dependent Kupffer cell (KC) activation as an early event in the pathogenesis of PNALI. We developed a mouse model in which intestinal injury and increased permeability were induced by oral treatment for 4 days with dextran sulphate sodium (DSS) followed by continuous infusion of soy lipid-based PN solution through a central venous catheter for 7 (PN7d/DSS) and 28 (PN28d/DSS) days. Purified KCs were probed for transcription of proinflammatory cytokines. PN7d/DSS mice showed increased intestinal permeability and elevated portal vein LPS levels, evidence of hepatocyte injury and cholestasis (serum aspartate aminotransferase, alanine aminotransferase, bile acids, total bilirubin), and increased KC expression of interleukin-6 (Il6), tumor necrosis factor α (Tnfα), and transforming growth factor ß (Tgfß). Markers of liver injury remained elevated in PN28d/DSS mice associated with lobular inflammation, hepatocyte apoptosis, peliosis, and KC hypertrophy and hyperplasia. PN infusion without DSS pretreatment or DSS pretreatment alone did not result in liver injury or KC activation, even though portal vein LPS levels were elevated. Suppression of the intestinal microbiota with broad spectrum antibiotics or ablation of TLR4 signaling in Tlr4 mutant mice resulted in significantly reduced KC activation and markedly attenuated liver injury in PN7d/DSS mice. CONCLUSION: These data suggest that intestinal-derived LPS activates KC through TLR4 signaling in early stages of PNALI.


Subject(s)
Intestines/injuries , Kupffer Cells/metabolism , Liver/injuries , Parenteral Nutrition/adverse effects , Toll-Like Receptor 4/metabolism , Animals , Biopsy, Needle , Cell Membrane Permeability/physiology , Disease Models, Animal , Immunohistochemistry , Intestines/pathology , Lipopolysaccharides/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parenteral Nutrition/methods , Random Allocation , Reference Values , Signal Transduction
16.
Am J Pathol ; 179(5): 2302-14, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21945903

ABSTRACT

Intestinal remodeling and stricture formation is a complication of inflammatory bowel disease (IBD) that often requires surgical intervention. Although eosinophils are associated with mucosal remodeling in other organs and are increased in IBD tissues, their role in IBD-associated remodeling is unclear. Histological and molecular features of ileitis and remodeling were assessed using immunohistochemical, histomorphometric, flow cytometric, and molecular analysis (real-time RT-PCR) techniques in a murine model of chronic eosinophilic ileitis. Collagen protein was assessed by Sircol assay. Using a spontaneous eosinophilic Crohn's-like mouse model SAMP1/SkuSlc, we demonstrate an association between ileitis progression and remodeling over the course of 40 weeks. Mucosal and submucosal eosinophilia increased over the time course and correlated with increased histological inflammatory indices. Ileitis and remodeling increased over the 40 weeks, as did expression of fibronectin. CCR3-specific antibody-mediated reduction of eosinophils resulted in significant decrease in goblet cell hyperplasia, muscularis propria hypertrophy, villus blunting, and expression of inflammatory and remodeling genes, including fibronectin. Cellularity of local mesenteric lymph nodes, including T- and B-lymphocytes, was also significantly reduced. Thus, eosinophils participate in intestinal remodeling, supporting eosinophils as a novel therapeutic target.


Subject(s)
Eosinophils/physiology , Ileitis/physiopathology , Receptors, CCR3/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/physiology , Chemokine CCL11/metabolism , Chemokine CCL24/metabolism , Chronic Disease , Cytokines/metabolism , Dexamethasone/pharmacology , Female , Fibrosis , Ileitis/drug therapy , Ileitis/pathology , Immunoglobulin G/pharmacology , Intestinal Mucosa/pathology , Mice , Mice, Inbred Strains , Mucous Membrane/pathology , Permeability , Receptors, CCR3/immunology , Receptors, CCR3/metabolism
17.
J Pediatr Gastroenterol Nutr ; 52(6): 650-61, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21593640

ABSTRACT

Inflammatory bowel diseases (IBD) are characterized by the invasion of leukocytes into the intestinal mucosa. However, a mixed inflammatory picture is observed that includes neutrophils, lymphocytes, monocytes, and eosinophils. To this day, the role of eosinophils in health and in disease remains unclear. Investigations into their function stem primarily from allergic diseases, asthma, and parasitic infections. This makes it even more difficult to discern a role for the fascinating eosinophil in IBDs because, unlike the lung or the skin, eosinophils reside in normal intestinal mucosa and increase in disease states; consequently, an intricate system must regulate their migration and numbers. These granulocytes are equipped with the machinery to participate in gastrointestinal (GI) inflammation and in the susceptible microenvironment, they may initiate or perpetuate an inflammatory response. A significant body of literature characterizes eosinophils present in the GI microenvironment where they have the potential to interact with other resident cells, thus promoting intestinal remodeling, mucus production, epithelial barrier, cytokine production, angiogenesis, and neuropeptide release. A number of lines of evidence support both potential beneficial and deleterious roles of eosinophils in the gut. Although studies from the gut and other mucosal organs suggest eosinophils affect mucosal GI inflammation, definitive roles for eosinophils in IBDs await discovery.


Subject(s)
Eosinophils/physiology , Gastrointestinal Diseases/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Animals , Gastric Mucosa/immunology , Humans , Inflammation/immunology
18.
Inflamm Bowel Dis ; 16(5): 743-52, 2010 May.
Article in English | MEDLINE | ID: mdl-19856411

ABSTRACT

BACKGROUND: SAMP1/Yit mice develop spontaneous, segmental, transmural ileitis recapitulating many features of Crohn's disease (CD). The ileitic phenotype may have arisen during crosses of SAMP1 mice selected for the presence of skin lesions. We hereby describe that the original SAMP1 strain similarly develops ileitis. Our aim was to characterize the histopathological and immunological features of this model and assess its responsiveness to standard inflammatory bowel disease (IBD) therapy. METHODS: The time course of histopathological features of ileitis was assessed. Immune compartments were characterized by flow cytometry. Ileal cytokine profiles and transcription factors were determined by real-time reverse-transcription polymerase chain reaction (RT-PCR). Finally, response to corticosteroid therapy and its effect on immune compartments and cellularity was evaluated. RESULTS: Histological features and time course of disease were conserved, compared to those reported in SAMP1/Yit strains, with similar expansion of CD19+, CD4+, and CD8+ effector (CD44(high) CD62L(low)), and central memory lymphocytes (CD44(high)CD62L(high)). However, different from SAMP1/YitFc mice, analysis of ileal cytokine profiles revealed initial T(H)1 polarization followed by T(H)2-polarized profile accompanied by prominent eosinophilia during late disease. Lastly, corticosteroids attenuated ileitis, resulting in decreased lymphocyte subsets and cellularity of compartments. CONCLUSIONS: Here we report that the ileitic phenotype of SAMP1-related strains was already present in the original SAMP1 strain. By contrast, the cytokine profile within the terminal ilea of SAMP1 is distinct from the mixed T(H)1/T(H)2 profile of SAMP1/YitFc mice during late disease, as it shows predominant T(H)2 polarization. Dissemination of these strains may advance our understanding of CD pathogenesis, which in 60% of patients involves the terminal ileum.


Subject(s)
Disease Models, Animal , Ileitis/immunology , Ileitis/pathology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cells, Cultured , Crohn Disease/drug therapy , Crohn Disease/immunology , Crohn Disease/pathology , Cytokines/genetics , Cytokines/metabolism , Dexamethasone/pharmacology , Eosinophils/immunology , Eosinophils/pathology , Female , Flow Cytometry , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Ileitis/drug therapy , Immunologic Memory/drug effects , Mice , Mice, Mutant Strains , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
Mol Biol Cell ; 21(4): 538-46, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20032301

ABSTRACT

Mucosal surfaces, such as the lung and intestine, are lined by a monolayer of epithelia that provides tissue barrier and transport function. It is recently appreciated that a common feature of inflammatory processes within the mucosa is hypoxia (so-called inflammatory hypoxia). Given the strong association between bacterial translocation and mucosal inflammatory disease, we hypothesized that intestinal epithelial hypoxia influences bacterial translocation. Initial studies revealed that exposure of cultured intestinal epithelia to hypoxia (pO(2), 20 torr; 24-48 h) resulted in a increase of up to 40-fold in the translocation of some strains of Gram-positive bacteria, independently of epithelial barrier function. A screen of relevant pathway inhibitors identified a prominent role for the platelet-activating factor receptor (PAFr) in hypoxia-associated bacterial translocation, wherein pharmacologic antagonists of PAFr blocked bacterial translocation by as much as 80 +/- 6%. Extensions of these studies revealed that hypoxia prominently induces PAFr through a hypoxia-inducible factor (HIF)-dependent mechanism. Indeed, HIF and PAFr loss of function studies (short hairpin RNA) revealed that apically expressed PAFr is central to the induction of translocation for the Gram-positive bacteria Enterococcus faecalis. Together, these findings reveal that some strains of Gram-positive bacteria exploit HIF-regulated PAFr as a means for translocation through intestinal epithelial cells.


Subject(s)
Gram-Positive Bacteria/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intestinal Mucosa , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Biological Transport/physiology , Caco-2 Cells , Cell Membrane/metabolism , Gene Knockdown Techniques , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Platelet Membrane Glycoproteins/genetics , RNA Interference , Receptors, G-Protein-Coupled/genetics , Transcriptional Activation
20.
Immunol Allergy Clin North Am ; 29(1): 171-8, xii-xiii, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19141352

ABSTRACT

Eosinophilic gastrointestinal diseases (EGIDs) are characterized by a wide variety of gastrointestinal symptoms that occur in conjunction with increased numbers of eosinophils in intestinal tissues. With the precise role or roles of eosinophils in gastrointestinal dysfunction incompletely understood, this subject remains an area of intense investigation. Most studies suggest that the intimate anatomic association of eosinophils with the intestinal epithelium implicates participation in the pathophysiology of EGIDs. This article reviews the limited evidence suggesting that the epithelium and eosinophils interact in the gastrointestinal tract and in other organ systems and describes how the epithelium and eosinophils might participate in gastrointestinal inflammatory diseases.


Subject(s)
Cell Communication , Eosinophilia/pathology , Eosinophils/metabolism , Epithelial Cells/metabolism , Gastrointestinal Diseases/pathology , Animals , Cell Movement , Cytotoxicity, Immunologic , Eosinophil Cationic Protein/immunology , Eosinophilia/immunology , Eosinophils/immunology , Eosinophils/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Gastrointestinal Diseases/immunology , Humans , Interleukin-13/immunology , Signal Transduction , Transforming Growth Factor beta/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...