Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Inflamm Bowel Dis ; 25(5): 875-885, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30576451

ABSTRACT

BACKGROUND: Adherent and invasive Escherichia coli (AIEC) is preferentially associated with ileal Crohn's disease (CD). The role of AIEC in the development of inflammation and its regional tropism is unresolved. The presence of long polar fimbriae (LPF) in 71% of ileal CD AIEC suggests a role for LPF in the tropism and virulence of AIEC. The aim of our study is to determine if AIEC, with or without LpfA, induces intestinal inflammation in monoassociated IL-10-/- mice. METHODS: We compared murine AIEC strains NC101 (phylogroup B2, LpfA-) and CUMT8 (phylogroup B1, LpfA+), and isogenic mutant CUMT8 lacking lpfA154, with a non-AIEC (E. coli K12), evaluating histologic inflammation, bacterial colonization, mucosal adherence and invasion, and immune activation. RESULTS: IL-10-/- mice monoassociated with AIEC (either CUMT8, CUMT8:ΔlpfA, or NC101) but not K12 developed diffuse small intestinal and colonic inflammation. There was no difference in the magnitude and distribution of inflammation in mice colonized with CUMT8:ΔlpfA compared with wild-type CUMT8. Bacterial colonization was similar for all E. coli strains. Fluorescence in situ hybridization revealed mucosal adherence and tissue invasion by AIEC but not K12. Production of the cytokines IL-12/23 p40 by the intestinal tissue and IFN-γ and IL-17 by CD4 T cells correlated with inflammation. CONCLUSIONS: IL-10-/- mice monoassociated with murine AIEC irrespective of LpfA expression developed chronic inflammation accompanied by IL-12/23 p40 production in the small and large intestines and IFN-γ/IL-17 production by CD4 T cells that model the interplay between enteric pathosymbionts, host susceptibility, and enhanced immune responses in people with IBD.


Subject(s)
Bacterial Adhesion , Escherichia coli Infections/immunology , Escherichia coli Proteins/metabolism , Fimbriae Proteins/metabolism , Inflammation/etiology , Interleukin-10/physiology , Intestine, Large/immunology , Intestine, Small/immunology , Animals , Escherichia coli/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Fimbriae, Bacterial/immunology , Fimbriae, Bacterial/pathology , Inflammation/metabolism , Inflammation/pathology , Intestine, Large/metabolism , Intestine, Large/microbiology , Intestine, Large/pathology , Intestine, Small/metabolism , Intestine, Small/microbiology , Intestine, Small/pathology , Mice , Mice, Knockout
2.
J Histochem Cytochem ; 59(9): 826-41, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21852692

ABSTRACT

Human Helicobacter pylori infection leads to multiple pathological consequences, including gastritis and adenocarcinoma. Although this association has led to the classification of H. pylori as a type 1 carcinogen, it is not clear if additional nonhelicobacter gastric microbiota play a role in these diseases. In this study, we utilized either specific pathogen-free C57BL/6 mice (B6.SPF) or mice colonized with altered Schaedler flora (B6.ASF) to evaluate the role of nonhelicobacter gastric microbiota in disease development after Helicobacter felis infection. Despite similar histological changes, H. felis persisted in B6.ASF stomachs, while H. felis could no longer be detected in the majority of B6.SPF mice. The B6.SPF mice also acquired multiple Lactobacillus spp. in their stomachs after H. felis infection. Our data indicate that potential mechanisms responsible for the ineffective H. felis clearance in the B6.ASF model include the absence of new gastric microbiota to compete for the gastric niche, the lack of expression of new gastric mucins, and a reduced ratio of H. felis-specific IgG2c:IgG1 serum antibodies. These data suggest that although H. felis is sufficient to initiate gastric inflammation and atrophy, bacterial eradication and the systemic immune response to infection are significantly influenced by pre-existing and acquired gastric microbiota.


Subject(s)
Helicobacter Infections/immunology , Helicobacter Infections/pathology , Helicobacter felis/immunology , Helicobacter felis/pathogenicity , Metagenome/physiology , Stomach Diseases/microbiology , Animals , Disease Progression , Female , Helicobacter felis/isolation & purification , Metagenome/immunology , Mice , Mice, Inbred C57BL , Stomach Diseases/immunology , Stomach Diseases/pathology
3.
J Histochem Cytochem ; 57(5): 457-67, 2009 May.
Article in English | MEDLINE | ID: mdl-19153195

ABSTRACT

The C57BL/6 mouse has been shown to develop gastric adenocarcinoma after Helicobacter felis infection. This model was used to determine whether mucin and trefoil factor (TFF) expression after infection was altered in a similar fashion to the changes seen in the protective gastric mucus layer of the human stomach after H. pylori infection. Our results indicate that this mouse model mimics many of the changes seen after human H. pylori infection, including increased expression of muc4 and muc5b and loss of muc5ac. These alterations in mucin expression occurred as early as 4 weeks postinfection, before the development of significant mucous metaplasia or gastric dysplasia. The decrease in muc5ac expression occurred only in the body of the stomach and was not secondary to the adaptive immune response to infection, because a similar decrease in expression was seen after infection of B6.Rag-1(-/-) mice, which lack B and T cells. Intriguingly, the increased expression of Muc4 and Muc5b in infected C57BL/6 mice was not seen in the infected B6.Rag-1(-/-) mice. Because B6.Rag-1(-/-) mice do not develop gastric pathology after H. felis infection, these findings point to the potential role of Muc4 and Muc5b in disease progression. This manuscript contains online supplemental material at http://www.jhc.org. Please visit this article online to view these materials.


Subject(s)
Gastric Mucosa/metabolism , Helicobacter Infections/metabolism , Mucus/metabolism , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Chronic Disease , Disease Progression , Female , Gastric Mucins/genetics , Gastric Mucins/metabolism , Gastric Mucosa/pathology , Gastritis/immunology , Gastritis/metabolism , Gastritis/pathology , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Helicobacter felis , Metaplasia , Mice , Mice, Inbred C57BL , Peptides/genetics , Peptides/metabolism , RNA, Messenger/metabolism , Trefoil Factor-2
4.
Gastroenterology ; 134(2): 491-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18242215

ABSTRACT

BACKGROUND & AIMS: Helicobacter pylori infection in children infrequently causes gastroduodenal mucosal ulceration. Because H pylori induces T-cell dependent gastric inflammation in adults and T regulatory (Treg) cells suppress T-cell-dependent pathology, we evaluated gastric histopathology and Treg cell responses in H pylori-infected children and adults. METHODS: Gastric tissue from 36 children and 79 adults with abdominal symptoms in Santiago, Chile, was evaluated prospectively for H pylori bacteria and histopathology using the Sydney classification and Treg responses using immunoassay, immunohistochemistry, and real-time polymerase chain reaction. RESULTS: Eighteen (50%) of the children and 51 (65%) of the adults were infected with H pylori. Children and adults were colonized with similar levels of H pylori. However, the level of gastritis in the children was reduced substantially compared with that of the adults (P < .05). Coincident with reduced gastric inflammation, the number of Treg cells and levels of Treg cytokines (transforming growth factor [TGF]-beta1 and interleukin-10) were increased markedly in the gastric mucosa of H pylori-infected children compared with that of infected adults (P < .03 and < .05, respectively). Also, H pylori infection in the children was associated with markedly increased levels of gastric TGF-beta1 and interleukin-10 messenger RNA. Importantly, gastric TGF-beta1 in H pylori-infected children localized predominantly to mucosal CD25(+) and Foxp3(+) cells, indicating a Treg source for the TGF-beta1. CONCLUSIONS: Gastric pathology is reduced and local Treg cell responses are increased in H pylori-infected children compared with infected adults, suggesting that gastric Treg cell responses down-regulate the inflammation and ulceration induced by H pylori in children.


Subject(s)
Gastritis/immunology , Gastritis/virology , Helicobacter Infections/immunology , Helicobacter pylori/pathogenicity , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/physiology , Adolescent , Adult , Aging/pathology , Cell Proliferation , Child , Chile , Down-Regulation , Female , Forkhead Transcription Factors/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastric Mucosa/virology , Gastritis/etiology , Helicobacter Infections/complications , Helicobacter Infections/pathology , Humans , Interleukin-10/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Prospective Studies , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/virology , Transforming Growth Factor beta1/metabolism
5.
J Histochem Cytochem ; 55(5): 515-24, 2007 May.
Article in English | MEDLINE | ID: mdl-17242461

ABSTRACT

The ELR(+) chemokine CXCL15, which recruits neutrophils during pulmonary inflammation, is also known as lungkine due to its reported exclusive expression in the lung. We now report that CXCL15 mRNA and protein are also expressed in other mucosal and endocrine organs including the gastrointestinal and urogenital tracts and the adrenal gland. Our results indicate that CXCL15 is expressed throughout the gastrointestinal tract, with the exception of the cecum. Gastric CXCL15 protein expression is approximately 10-fold lower than pulmonary expression and primarily occurs in a specific lineage of gastric epithelial cells, the prezymogenic and zymogenic cell. Similar to the increased expression of CXCL15 during pulmonary inflammation, gastric inflammation induced by infection with Helicobacter felis caused an increase in gastric CXCL15 expression. However, colonic CXCL15 expression was not altered in two different models of colonic inflammation, the Helicobacter hepaticus T-cell transfer model and the mdr1a(-/-) model of colitis. These findings clearly demonstrate that CXCL15, previously reported to be the only lung-specific chemokine, is also highly expressed in other murine mucosal and endocrine organs. The functional role of CXCL15 in mucosal disease remains to be elucidated. This manuscript contains online supplemental material at (http://www.jhc.org). Please visit this article online to view these materials.


Subject(s)
Chemokines, CXC/biosynthesis , Endocrine System/metabolism , Gastrointestinal Tract/metabolism , Uterus/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/genetics , Animals , Chemokines, CXC/genetics , Colitis/etiology , Colitis/metabolism , Female , Fluorescent Antibody Technique , Gastritis/metabolism , Helicobacter Infections/metabolism , Helicobacter felis , Helicobacter hepaticus , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/metabolism , Organ Specificity , RNA, Messenger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...