Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Mol Gastroenterol Hepatol ; : 101378, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38992465

ABSTRACT

BACKGROUND & AIMS: Addition of sialic acids (sialylation) to glycoconjugates is a common capping step of glycosylation. Our study aims to determine the roles of the overall sialylation in intestinal mucosal homeostasis. METHODS: Mice with constitutive deletion of intestinal epithelial sialylation (IEC Slc35a1-/- mice) and mice with inducible deletion of sialylation in intestinal epithelium (TM-IEC Slc35a1-/- mice) were generated, which were used to determine the roles of overall sialylation in intestinal mucosal homeostasis by ex vivo and muti-omics studies. RESULTS: IEC Slc35a1-/- mice developed mild spontaneous microbiota-dependent colitis. Additionally, 30% of IEC Slc35a1-/- mice had spontaneous tumors in the rectum over the age of 12 months. TM-IEC Slc35a1-/- mice were highly susceptible to acute inflammation induced by 1% DSS vs controls. Loss of total sialylation was associated with reduced mucus thickness on fecal sections and within colon tissues. TM-IEC Slc35a1-/- mice showed altered microbiota with an increase in Clostridia disporicum, which is associated a global reduction in the abundance of at least 20 unique taxa; however, metabolomic analysis did not show any significant differences in short-chain fatty acid levels. Treatment with 5-fluorouracil (5-FU) led to more severe small intestine mucositis in the IEC Slc35a1-/- mice vs. WT littermates, which was associated with reduced Lgr5+ cell representation in small intestinal crypts in IEC Slc35a1-/-;Lgr5-GFP mice. CONCLUSIONS: Loss of overall sialylation impairs mucus stability and the stem cell niche leading to microbiota-dependent spontaneous colitis and tumorigenesis.

2.
J Biol Chem ; 300(3): 105675, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38272223

ABSTRACT

The O-glycoprotein Mucin-2 (MUC2) forms the protective colon mucus layer. While animal models have demonstrated the importance of Muc2, few studies have explored human MUC2 in similar depth. Recent studies have revealed that secreted MUC2 is bound to human feces. We hypothesized human fecal MUC2 (HF-MUC2) was accessible for purification and downstream structural and functional characterization. We tested this via histologic and quantitative imaging on human fecal sections; extraction from feces for proteomic and O-glycomic characterization; and functional studies via growth and metabolic assays in vitro. Quantitative imaging of solid fecal sections showed a continuous mucus layer of varying thickness along human fecal sections with barrier functions intact. Lectin profiling showed HF-MUC2 bound several lectins but was weak to absent for Ulex europaeus 1 (α1,2 fucose-binding) and Sambucus nigra agglutinin (α2,6 sialic acid-binding), and did not have obvious b1/b2 barrier layers. HF-MUC2 separated by electrophoresis showed high molecular weight glycoprotein bands (∼1-2 MDa). Proteomics and Western analysis confirmed the enrichment of MUC2 and potential MUC2-associated proteins in HF-MUC2 extracts. MUC2 O-glycomics revealed diverse fucosylation, moderate sialylation, and little sulfation versus porcine colonic MUC2 and murine fecal Muc2. O-glycans were functional and supported the growth of Bacteroides thetaiotaomicron (B. theta) and short-chain fatty acid (SCFA) production in vitro. MUC2 could be similarly analyzed from inflammatory bowel disease stools, which displayed an altered glycomic profile and differential growth and SCFA production by B. theta versus healthy samples. These studies describe a new non-invasive platform for human MUC2 characterization in health and disease.


Subject(s)
Colon , Feces , Proteomics , Animals , Humans , Mice , Colon/metabolism , Glycoproteins/metabolism , Intestinal Mucosa/metabolism , Mucin-2/genetics , Mucin-2/metabolism , Mucus/metabolism , Swine , Male , Mice, Inbred C57BL , Gastrointestinal Microbiome
3.
PLoS Pathog ; 11(8): e1005108, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26285214

ABSTRACT

Enterohemorrhagic Escherichia coli and related food and waterborne pathogens pose significant threats to human health. These attaching/effacing microbes infect the apical surface of intestinal epithelial cells (IEC), causing severe diarrheal disease. Colonizing the intestinal luminal surface helps segregate these microbes from most host inflammatory responses. Based on studies using Citrobacter rodentium, a related mouse pathogen, we speculate that hosts rely on immune-mediated changes in IEC, including goblet cells to defend against these pathogens. These changes include a CD4+ T cell-dependent increase in IEC proliferation to replace infected IEC, as well as altered production of the goblet cell-derived mucin Muc2. Another goblet cell mediator, REsistin-Like Molecule (RELM)-ß is strongly induced within goblet cells during C. rodentium infection, and was detected in the stool as well as serum. Despite its dramatic induction, RELM-ß's role in host defense is unclear. Thus, wildtype and RELM-ß gene deficient mice (Retnlb-/-) were orally infected with C. rodentium. While their C. rodentium burdens were only modestly elevated, infected Retnlb-/- mice suffered increased mortality and mucosal ulceration due to deep pathogen penetration of colonic crypts. Immunostaining for Ki67 and BrDU revealed Retnlb-/- mice were significantly impaired in infection-induced IEC hyper-proliferation. Interestingly, exposure to RELM-ß did not directly increase IEC proliferation, rather RELM-ß acted as a CD4+ T cell chemoattractant. Correspondingly, Retnlb-/- mice showed impaired CD4+ T cell recruitment to their infected colons, along with reduced production of interleukin (IL)-22, a multifunctional cytokine that directly increased IEC proliferation. Enema delivery of RELM-ß to Retnlb-/- mice restored CD4+ T cell recruitment, concurrently increasing IL-22 levels and IEC proliferation, while reducing mucosal pathology. These findings demonstrate that RELM-ß and goblet cells play an unexpected, yet critical role in recruiting CD4+ T cells to the colon to protect against an enteric pathogen, in part via the induction of increased IEC proliferation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Colitis/immunology , Goblet Cells/immunology , Hormones, Ectopic/immunology , Intestinal Mucosa/immunology , Animals , Cell Separation , Citrobacter rodentium , Colitis/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Goblet Cells/metabolism , Hormones, Ectopic/metabolism , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction
4.
PLoS One ; 10(5): e0125225, 2015.
Article in English | MEDLINE | ID: mdl-25932952

ABSTRACT

Inflammatory bowel disease is a chronic gastrointestinal inflammatory disorder associated with changes in neuropeptide expression and function, including vasoactive intestinal peptide (VIP). VIP regulates intestinal vasomotor and secretomotor function and motility; however, VIP's role in development and maintenance of colonic epithelial barrier homeostasis is unclear. Using VIP deficient (VIPKO) mice, we investigated VIP's role in epithelial barrier homeostasis, and susceptibility to colitis. Colonic crypt morphology and epithelial barrier homeostasis were assessed in wildtype (WT) and VIPKO mice, at baseline. Colitic responses were evaluated following dinitrobenzene sulfonic acid (DNBS) or dextran-sodium sulfate (DSS) exposure. Mice were also treated with exogenous VIP. At baseline, VIPKO mice exhibited distorted colonic crypts, defects in epithelial cell proliferation and migration, increased apoptosis, and altered permeability. VIPKO mice also displayed reduced goblet cell numbers, and reduced expression of secreted goblet cell factors mucin 2 and trefoil factor 3. These changes were associated with reduced expression of caudal type homeobox 2 (Cdx2), a master regulator of intestinal function and homeostasis. DNBS and DSS-induced colitis were more severe in VIPKO than WT mice. VIP treatment rescued the phenotype, protecting VIPKO mice against DSS colitis, with results comparable to WT mice. In conclusion, VIP plays a crucial role in the development and maintenance of colonic epithelial barrier integrity under physiological conditions and promotes epithelial repair and homeostasis during colitis.


Subject(s)
Colitis/prevention & control , Homeostasis/drug effects , Intestines/pathology , Protective Agents/pharmacology , Vasoactive Intestinal Peptide/metabolism , Animals , CDX2 Transcription Factor , Cell Count , Colitis/pathology , Dinitrofluorobenzene/analogs & derivatives , Disease Susceptibility , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Goblet Cells/pathology , Homeodomain Proteins/metabolism , Intestines/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Transcription Factors/metabolism , Vasoactive Intestinal Peptide/deficiency
5.
Infect Immun ; 81(12): 4649-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24101690

ABSTRACT

Both idiopathic and infectious forms of colitis disrupt normal intestinal epithelial cell (IEC) proliferation and differentiation, although the mechanisms involved remain unclear. Recently, we demonstrated that infection by the attaching and effacing murine pathogen Citrobacter rodentium leads to a significant reduction in colonic goblet cell numbers (goblet cell depletion). This pathology depends on T and/or B cells, as Rag1(-/-) mice do not suffer this depletion during infection, instead suffering high mortality rates. To address the immune mechanisms involved, we reconstituted Rag(-/-) mice with either CD4(+) or CD8(+) T cells. Both T cell subsets increased Rag1(-/-) mouse survival during infection, with mice that received CD8(+) T cells developing colonic ulcers but not goblet cell depletion. In contrast, mice that received CD4(+) T cells showed goblet cell depletion in concert with exaggerated IEC proliferation. To define the possible involvement of T cell-derived cytokines, we infected gamma interferon receptor gene knockout (IFN-γR(-/-)) mice and wild-type mice given interleukin 17A (IL-17A) neutralizing antibodies and found that IFN-γ signaling was required for both goblet cell depletion and increased IEC proliferation. Immunostaining revealed that C. rodentium cells preferentially localized to nonhyperplastic crypts containing numerous goblet cells, whereas hyperplastic, goblet cell-depleted crypts appeared protected from infection. To address whether goblet cell depletion benefits the C. rodentium-infected host, we increased goblet cell numbers using the γ-secretase inhibitor dibenzazepine (DBZ), which resulted in greatly increased pathogen burdens and mortality rates. These results demonstrate that goblet cell depletion reflects host immunomodulation of IEC homeostasis and reflects a novel host defense mechanism against mucosal-adherent pathogens.


Subject(s)
Antibodies, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Enterobacteriaceae Infections/immunology , Goblet Cells/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , Bacterial Load/drug effects , Cell Differentiation , Cell Proliferation , Citrobacter rodentium/immunology , Colitis/immunology , Colitis/microbiology , Colitis/mortality , Dibenzazepines , Enterobacteriaceae Infections/mortality , Epithelial Cells/immunology , Epithelial Cells/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-17/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology
6.
Glycobiology ; 23(9): 1026-37, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23752712

ABSTRACT

Mucin-type O-glycans are the primary constituents of mucins that are expressed on various mucosal sites of the body, especially the bacteria-laden intestinal tract. Mucins are the main components of mucus, which is secreted by goblet cells and forms a protective homeostatic barrier between the resident microbiota and the underlying immune cells in the colon. However, the specific role of mucin-type O-glycans in mucus barrier function has been uncertain. Recent studies utilizing mice deficient in key glycosyltransferases involved in O-glycan biosynthesis on intestinal mucins have underscored the importance of mucin-type O-glycosylation in mucus barrier function. This review will highlight recent advances in our understanding of mucin-type O-glycan function in the mucus barrier and how they promote mutualism with our resident microbiota.


Subject(s)
Homeostasis , Intestinal Mucosa/metabolism , Mucins/chemistry , Mucins/metabolism , Polysaccharides/metabolism , Humans , Intestines/chemistry , Polysaccharides/chemistry
7.
J Immunol ; 190(5): 2292-300, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355735

ABSTRACT

Resistin-like molecule (RELM)α belongs to a family of secreted mammalian proteins that have putative immunomodulatory functions. Recent studies have identified a pathogenic role for RELMα in chemically induced colitis through effects on innate cell populations. However, whether RELMα regulates intestinal adaptive immunity to enteric pathogens is unknown. In this study, we employed Citrobacter rodentium as a physiologic model of pathogenic Escherichia coli-induced diarrheal disease, colitis, and Th17 cell responses. In response to Citrobacter, RELMα expression was induced in intestinal epithelial cells, infiltrating macrophages, and eosinophils of the infected colons. Citrobacter-infected RELMα(-/-) mice exhibited reduced infection-induced intestinal inflammation, characterized by decreased leukocyte recruitment to the colons and reduced immune cell activation compared with wild-type (WT) mice. Interestingly, Citrobacter colonization and clearance were unaffected in RELMα(-/-) mice, suggesting that the immune stimulatory effects of RELMα following Citrobacter infection were pathologic rather than host-protective. Furthermore, infected RELMα(-/-) mice exhibited decreased CD4(+) T cell expression of the proinflammatory cytokine IL-17A. To directly test whether RELMα promoted Citrobacter-induced intestinal inflammation via IL-17A, infected WT and IL-17A(-/-) mice were treated with rRELMα. RELMα treatment of Citrobacter-infected WT mice exacerbated intestinal inflammation and IL-17A expression whereas IL-17A(-/-) mice were protected from RELMα-induced intestinal inflammation. Finally, infected RELMα(-/-) mice exhibited reduced levels of serum IL-23p19 compared with WT mice, and RELMα(-/-) peritoneal macrophages showed deficient IL-23p19 induction. Taken together, these data identify a proinflammatory role for RELMα in bacterial-induced colitis and suggest that the IL-23/Th17 axis is a critical mediator of RELMα-induced inflammation.


Subject(s)
Citrobacter rodentium/immunology , Inflammation/immunology , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-17/immunology , Intestines/drug effects , Macrophages, Peritoneal/drug effects , Th17 Cells/drug effects , Adaptive Immunity/drug effects , Animals , Citrobacter rodentium/pathogenicity , Dextran Sulfate , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/pathology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Gene Expression/drug effects , Inflammation/chemically induced , Inflammation/microbiology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-23 Subunit p19/blood , Interleukin-23 Subunit p19/immunology , Intestines/immunology , Intestines/microbiology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Th17 Cells/immunology , Th17 Cells/pathology
8.
Am J Physiol Gastrointest Liver Physiol ; 303(3): G311-23, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22679002

ABSTRACT

Salmonella enterica serovar Typhimurium is a clinically important gram-negative, enteric bacterial pathogen that activates several Toll-like receptors (TLRs). While TLR signaling through the adaptor protein MyD88 has been shown to promote inflammation and host defense against the systemic spread of S. Typhimurium, curiously, its role in the host response against S. Typhimurium within the mammalian gastrointestinal (GI) tract is less clear. We therefore used the recently described Salmonella-induced enterocolitis and fibrosis model: wild-type (WT) and MyD88-deficient (MyD88(-/-)) mice pretreated with streptomycin and then orally infected with the ΔaroA vaccine strain of S. Typhimurium. Tissues were analyzed for bacterial colonization, inflammation, and epithelial damage, while fibrosis was assessed by collagen quantification and Masson's trichrome staining. WT and MyD88(-/-) mice carried similar intestinal pathogen burdens to postinfection day 21. Infection of WT mice led to acute mucosal and submucosal inflammation and edema, as well as significant intestinal epithelial damage and proliferation, leading to widespread goblet cell depletion. Impressive collagen deposition in the WT intestine was also evident in the submucosa at postinfection days 7 and 21, with fibrotic regions rich in fibroblasts and collagen. While infected MyD88(-/-) mice showed levels of submucosal inflammation and edema similar to WT mice, they were impaired in the development of mucosal inflammation, along with infection-induced epithelial damage, proliferation, and goblet cell depletion. MyD88(-/-) mouse tissues also had fewer submucosal fibroblasts and 60% less collagen. We noted that cyclooxygenase (Cox)-2 expression was MyD88-dependent, with numerous Cox-2-positive cells identified in fibrotic regions of WT mice at postinfection day 7, but not in MyD88(-/-) mice. Treatment of WT mice with the Cox-2 inhibitor rofecoxib (20 mg/kg) significantly reduced fibroblast numbers and collagen levels without altering colitis severity. In conclusion, MyD88 and Cox-2 signaling play roles in intestinal fibrosis during Salmonella-induced enterocolitis.


Subject(s)
Cyclooxygenase 2/physiology , Enterocolitis/physiopathology , Myeloid Differentiation Factor 88/physiology , Salmonella Infections, Animal/microbiology , Animals , Cyclooxygenase 2 Inhibitors , Enterocolitis/microbiology , Enterocolitis/pathology , Fibrosis/pathology , Intestinal Mucosa/pathology , Lactones/pharmacology , Mice , Salmonella typhimurium , Signal Transduction , Sulfones/pharmacology
9.
Cell Microbiol ; 14(4): 475-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22233392

ABSTRACT

Infection by enteric bacterial pathogens activates pathogen recognition receptors, leading to innate responses that promote host defence. While responses that promote host 'resistance' to infection, through the release of antimicrobial mediators, or the recruitment of inflammatory cells aimed at clearing the infection are best known, recent studies have begun to identify additional innate driven responses that instead promote intestinal tissue repair and host survival. Described as infection 'tolerance' responses, we and others have primarily studied these responses in the Citrobacter rodentium infection model. In this review we discuss the impact of innate resistance mechanisms on host defence, and describe how 'tolerance' responses act primarily on the intestinal epithelium, triggering epithelial cell proliferation, repair or promoting barrier function. Resistance and tolerance responses appear to work together, with tolerance repairing the tissue injury caused by resistance driven inflammation. Tolerance responses fit a pattern where innate immunity and inflammation are tightly regulated in the gastrointestinal tract. Moreover, tolerance may have developed due to the successful subversion and avoidance of host resistance by enteric bacterial pathogens. Further studies are needed to clarify the contribution of different pathogen recognition receptors to tolerance and resistance responses against bacterial pathogens, in the gut or in other host tissues.


Subject(s)
Disease Resistance , Enterobacteriaceae/immunology , Host-Pathogen Interactions , Immunity, Innate , Animals , Cell Proliferation , Enterobacteriaceae/pathogenicity , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mammals , Signal Transduction , Symbiosis , Toll-Like Receptors/immunology
10.
Gastroenterology ; 139(4): 1277-88, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20600022

ABSTRACT

BACKGROUND & AIMS: The roles of intestinal Toll-like receptors (TLR) in the pathogenesis of colitis are not known. TLR2 and TLR4 appear to protect against dextran sodium sulfate-induced colitis by promoting mucosal integrity, but it is not clear whether this method of protection occurs in other models of colitis. We investigated the roles of TLR2 and TLR4 and the cell types that express these receptors during infectious colitis. METHODS: We generated chimeric mice with TLR2(-/-) or TLR4(-/-) bone marrow and infected them with the bacterial pathogen Citrobacter rodentium. We assessed their susceptibility to colitis and the mechanisms of TLR-mediated mucosal integrity. RESULTS: TLR2-expressing tissue resident cells prevented lethal colitis, whereas TLR4-dependent inflammatory responses of hematopoietic cells mediated intestinal damage. TLR2 expression protected against intestinal damage by maintaining epithelial barrier function and inducing expression of interleukin (IL)-11 from tissue resident cells in the muscularis mucosae, concurrent with epithelial activation of the transcription factor STAT3. Addition of exogenous IL-11 protected against the lethal colitis in TLR2-deficient mice via STAT3 activation in intestinal epithelial cells. CONCLUSIONS: TLR2-dependent cytoprotective responses from tissue resident cells maintain mucosal integrity against the ultimately lethal TLR4-dependent inflammatory responses of hematopoietic cells. Whereas TLR2 protects against various noxious agents, the role of TLR4 during colitis can be either protective or damaging, depending on the stimulus. Therefore, therapeutics that reduce innate immunity (TLR2 signaling in particular) may not be beneficial to patients with colitis; they could worsen symptoms. Therapies that stimulate cytoprotective responses, like IL-11, could have benefits for patients with colitis.


Subject(s)
Colitis/etiology , Interleukin-11/physiology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/physiology , Animals , Colitis/immunology , Colitis/metabolism , Inflammatory Bowel Diseases/drug therapy , Interleukin-11/therapeutic use , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency
11.
PLoS Pathog ; 6(5): e1000902, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20485566

ABSTRACT

Despite recent advances in our understanding of the pathogenesis of attaching and effacing (A/E) Escherichia coli infections, the mechanisms by which the host defends against these microbes are unclear. The goal of this study was to determine the role of goblet cell-derived Muc2, the major intestinal secretory mucin and primary component of the mucus layer, in host protection against A/E pathogens. To assess the role of Muc2 during A/E bacterial infections, we inoculated Muc2 deficient (Muc2(-/-)) mice with Citrobacter rodentium, a murine A/E pathogen related to diarrheagenic A/E E. coli. Unlike wildtype (WT) mice, infected Muc2(-/-) mice exhibited rapid weight loss and suffered up to 90% mortality. Stool plating demonstrated 10-100 fold greater C. rodentium burdens in Muc2(-/-) vs. WT mice, most of which were found to be loosely adherent to the colonic mucosa. Histology of Muc2(-/-) mice revealed ulceration in the colon amid focal bacterial microcolonies. Metabolic labeling of secreted mucins in the large intestine demonstrated that mucin secretion was markedly increased in WT mice during infection compared to uninfected controls, suggesting that the host uses increased mucin release to flush pathogens from the mucosal surface. Muc2 also impacted host-commensal interactions during infection, as FISH analysis revealed C. rodentium microcolonies contained numerous commensal microbes, which was not observed in WT mice. Orally administered FITC-Dextran and FISH staining showed significantly worsened intestinal barrier disruption in Muc2(-/-) vs. WT mice, with overt pathogen and commensal translocation into the Muc2(-/-) colonic mucosa. Interestingly, commensal depletion enhanced C. rodentium colonization of Muc2(-/-) mice, although colonic pathology was not significantly altered. In conclusion, Muc2 production is critical for host protection during A/E bacterial infections, by limiting overall pathogen and commensal numbers associated with the colonic mucosal surface. Such actions limit tissue damage and translocation of pathogenic and commensal bacteria across the epithelium.


Subject(s)
Citrobacter rodentium , Colitis/immunology , Enterobacteriaceae Infections/immunology , Intestinal Mucosa/immunology , Mucin-2/metabolism , Animals , Bacterial Adhesion/immunology , Bacterial Translocation/immunology , Colitis/metabolism , Colitis/microbiology , Enterobacteriaceae Infections/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mucin-2/genetics , Mucin-2/immunology
12.
Immunity ; 32(3): 367-78, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20226691

ABSTRACT

Inflammatory caspases are essential effectors of inflammation and cell death. Here, we investigated their roles in colitis and colorectal cancer and report a bimodal regulation of intestinal homeostasis, inflammation and tumorigenesis by caspases-1 and -12. Casp1(-/-) mice exhibited defects in mucosal tissue repair and succumbed rapidly after dextran sulfate sodium administration. This phenotype was rescued by administration of exogenous interleukin-18 and was partially reproduced in mice deficient in the inflammasome adaptor ASC. Casp12(-/-) mice, in which the inflammasome is derepressed, were resistant to acute colitis and showed signs of enhanced repair. Together with their increased inflammatory response, the enhanced repair response of Casp12(-/-) mice rendered them more susceptible to colorectal cancer induced by azoxymethane (AOM)+DSS. Taken together, our results indicate that the inflammatory caspases are critical in the induction of inflammation in the gut after injury, which is necessary for tissue repair and maintenance of immune tolerance.


Subject(s)
Caspase 12/metabolism , Caspase 1/metabolism , Colitis/enzymology , Colitis/immunology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/immunology , Homeostasis , Animals , Caspase 1/deficiency , Caspase 1/immunology , Caspase 12/deficiency , Caspase 12/immunology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Colitis/complications , Colitis/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Immune Tolerance , Interleukin-18/biosynthesis , Interleukin-18/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism
13.
J Immunol ; 184(5): 2305-13, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20130217

ABSTRACT

Intestinal epithelial cells (IECs) are constantly exposed to enteric microbes. Although IECs express TLRs that recognize bacterial products, the activation of these TLRs is strictly controlled through poorly understood mechanisms, producing a state of hyporesponsiveness and preventing unwanted inflammation. The single IgG IL-1-related receptor (Sigirr) is a negative regulator of TLRs that is expressed by IECs and was recently shown to inhibit experimental colitis. However, the importance of Sigirr in IEC hyporesponsiveness and its distribution within the human colon is unknown. In this study, we investigated the role of Sigirr in regulating epithelial-specific TLR responses and characterized its expression in colonic biopsy specimens. Transformed and nontransformed human IECs were cultured as monolayers. Transient gene silencing and stable overexpression of Sigirr was performed to assess innate IEC responses. Sigirr expression in human colonic biopsy specimens was examined by immunohistochemistry. Bacterial infection of IECs and exposure to flagellin transiently decreased Sigirr protein expression, concurrent with secretion of the neutrophil chemokine IL-8. Sigirr gene silencing augmented chemokine responses to bacterial flagellin, Pam3Cys, and the cytokine IL-1beta. Conversely, stable overexpression of Sigirr diminished NF-kappaB-mediated IL-8 responses to TLR ligands. We also found that Sigirr expression increased as IECs differentiated in culture. This observation was confirmed in biopsy sections, in which Sigirr expression within colonic crypts was prominent in IECs at the apex and diminished at the base. Our findings show that Sigirr broadly regulates innate responses in differentiated human IECs; therefore, it may modulate epithelial involvement in infectious and inflammatory bowel diseases.


Subject(s)
Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Receptors, Interleukin-1/metabolism , Toll-Like Receptors/metabolism , Animals , Blotting, Western , CHO Cells , Caco-2 Cells , Cell Differentiation , Cell Line , Colon/metabolism , Cricetinae , Cricetulus , Epithelial Cells/microbiology , Flagellin/genetics , Flagellin/metabolism , Fluorescent Antibody Technique , HT29 Cells , Humans , Immunohistochemistry , Interleukin-8/metabolism , Intestinal Mucosa/cytology , NF-kappa B/metabolism , RNA Interference , Receptors, Interleukin-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/genetics
14.
Gastroenterology ; 134(3): 768-80, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18325390

ABSTRACT

BACKGROUND & AIMS: Intestinal fibrosis and stricture formation are serious complications of Crohn's disease, often requiring surgical intervention. Unfortunately, the mechanisms underlying intestinal fibrosis development are poorly understood, in part because of the lack of relevant animal models. Here, we present a novel murine model of severe and persistent intestinal fibrosis caused by chronic bacterial-induced colitis. METHODS: Mice were treated with streptomycin 24 hours prior to oral infection with Salmonella enterica serovar Typhimurium. Tissues were analyzed for bacterial colonization and inflammation, and fibrosis was assessed by Masson's trichrome staining and collagen quantification. Expression of the profibrotic cytokines transforming growth factor-beta1, connective tissue growth factor and insulin-like growth factor-I was determined, and the cell types present in fibrotic tissues were assessed by immunohistochemistry. RESULTS: Infection led to chronic Salmonella colonization of the cecum and colon followed by edema, mucosal ulcerations, and severe transmural inflammation. This pathology was accompanied by significantly elevated expression of transforming growth factor-beta1, connective tissue growth factor, and insulin-like growth factor-I along with extensive type I collagen deposition in the cecal mucosa, submucosa, and muscularis mucosa of infected mice. Fibrosis was evident by 7 days postinfection, peaking at day 21 and still present at day 70. The fibrotic regions were found to be rich in fibroblasts and myofibroblasts. CONCLUSIONS: These data demonstrate that chronic Salmonella infection of the murine gastrointestinal tract leads to severe tissue fibrosis. Because this model is highly reproducible and easy to perform, it provides great potential for investigating both host and bacterial contributions to intestinal fibrosis.


Subject(s)
Cecum/pathology , Colon/pathology , Enterocolitis/complications , Intestinal Diseases/pathology , Salmonella Infections/complications , Salmonella typhimurium , Animals , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cecum/metabolism , Cecum/microbiology , Chronic Disease , Collagen Type I/metabolism , Colon/metabolism , Colon/microbiology , Connective Tissue Growth Factor , Cytokines/metabolism , Disease Models, Animal , Enterocolitis/genetics , Enterocolitis/metabolism , Enterocolitis/microbiology , Enterocolitis/pathology , Fibroblasts/pathology , Fibrosis , Immediate-Early Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Diseases/genetics , Intestinal Diseases/metabolism , Intestinal Diseases/microbiology , Mice , Mice, Inbred C3H , Mice, Inbred DBA , Mutation , Salmonella Infections/genetics , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Severity of Illness Index , Species Specificity , Streptomycin/administration & dosage , Time Factors , Transforming Growth Factor beta1/metabolism , Up-Regulation , Virulence Factors/metabolism
15.
Infect Immun ; 76(4): 1410-22, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18227166

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) and the murine pathogen Citrobacter rodentium belong to the attaching and effacing (A/E) family of bacterial pathogens. These noninvasive bacteria infect intestinal enterocytes using a type 3 secretion system (T3SS), leading to diarrheal disease and intestinal inflammation. While flagellin, the secreted product of the EPEC fliC gene, causes the release of interleukin 8 (IL-8) from epithelial cells, it is unclear whether A/E bacteria also trigger epithelial inflammatory responses that are FliC independent. The aims of this study were to characterize the FliC dependence or independence of epithelial inflammatory responses to direct infection by EPEC or C. rodentium. Following infection of Caco-2 intestinal epithelial cells by wild-type and DeltafliC EPEC, a rapid activation of several proinflammatory genes, including those encoding IL-8, monocyte chemoattractant protein 1, macrophage inflammatory protein 3alpha (MIP3alpha), and beta-defensin 2, occurred in a FliC-dependent manner. These responses were accompanied by mitogen-activated protein kinase activation, as well as the Toll-like receptor 5 (TLR5)-dependent activation of NF-kappaB. At later infection time points, a subset of these proinflammatory genes (IL-8 and MIP3alpha) was also induced in cells infected with DeltafliC EPEC. The nonmotile A/E pathogen C. rodentium also triggered similar innate responses through a TLR5-independent but partially NF-kappaB-dependent mechanism. Moreover, the EPEC FliC-independent responses were increased in the absence of the locus of enterocyte effacement-encoded T3SS, suggesting that translocated bacterial effectors suppress rather than cause the FliC-independent inflammatory response. Thus, we demonstrate that infection of intestinal epithelial cells by A/E pathogens can trigger an array of proinflammatory responses from epithelial cells through both FliC-dependent and -independent pathways, expanding our understanding of the innate epithelial response to infection by these pathogens.


Subject(s)
Citrobacter rodentium/physiology , Enterobacteriaceae Infections/metabolism , Enteropathogenic Escherichia coli/physiology , Epithelial Cells/metabolism , Flagellin/metabolism , Inflammation/metabolism , Animals , Caco-2 Cells , Chemokine CCL20/metabolism , Dendritic Cells/cytology , Dendritic Cells/physiology , Enterobacteriaceae Infections/microbiology , Epithelial Cells/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation , Humans , Interleukin-8/metabolism , Mice , Mice, Inbred C3H , Mutation , NF-kappa B/metabolism , Time Factors , Toll-Like Receptor 5/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Cell Microbiol ; 10(2): 388-403, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17910742

ABSTRACT

Inflammatory bowel diseases and infectious gastroenteritis likely occur when the integrity of intestinal barriers is disrupted allowing luminal bacterial products to cross into the intestinal mucosa, stimulating immune cells and triggering inflammation. While specific Toll-like receptors (TLR) are involved in the generation of inflammatory responses against enteric bacteria, their contributions to the maintenance of intestinal mucosal integrity are less clear. These studies investigated the role of TLR2 in a model of murine colitis induced by the bacterial pathogen Citrobacter rodentium. C. rodentium supernatants specifically activated TLR2 in vitro while infected TLR2-/- mice suffered a lethal colitis coincident with colonic mucosal ulcerations, bleeding and increased cell death but not increased pathogen burden. TLR2-/- mice suffered impaired epithelial barrier function mediated via zonula occludens (ZO)-1 in naïve mice and claudin-3 in infected mice, suggesting this could underlie their susceptibility. TLR2 deficiency was also associated with impaired production of IL-6 by bone marrow-derived macrophages and infected colons cultured ex vivo. As IL-6 has antiapoptotic and epithelial repair capabilities, its reduced expression could contribute to the impaired mucosal integrity. These studies report for the first time that TLR2 plays a critical role in maintaining intestinal mucosal integrity during infection by a bacterial pathogen.


Subject(s)
Citrobacter rodentium/pathogenicity , Colitis/metabolism , Intestinal Mucosa/metabolism , Toll-Like Receptor 2/physiology , Animals , Apoptosis , Citrobacter rodentium/growth & development , Colitis/microbiology , Colitis/pathology , Colon/metabolism , Colon/microbiology , Colon/pathology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Interleukin-6/biosynthesis , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Toll-Like Receptor 2/genetics , Weight Loss
17.
Infect Immun ; 76(2): 796-811, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17984203

ABSTRACT

The attaching and effacing (A/E) bacterial pathogens enteropathogenic Escherichia coli and enterohemorrhagic E. coli and the related mouse pathogen Citrobacter rodentium colonize their hosts' intestines by infecting the apical surfaces of enterocytes, subverting their function, and they ultimately cause diarrhea. Surprisingly, little is known about the interactions of these organisms with goblet cells, which are specialized epithelial cells that secrete the protective molecules Muc2 and trefoil factor 3 (Tff3) into the intestinal lumen. C. rodentium infection leads to dramatic goblet cell depletion within the infected colon, yet it is not clear whether C. rodentium infects goblet cells or if this pathology is pathogen or host mediated. As determined by immunostaining and PCR, both the number of goblet cells and the expression of genes encoding Muc2 and Tff3 were significantly reduced by day 10 postinfection. While electron microscopy and immunostaining revealed that C. rodentium directly infected a fraction of colonic goblet cells, C. rodentium localization did not correlate with goblet cell depletion. To assess the role of the host immune system in these changes, Rag1 knockout (KO) (T- and B-cell-deficient) mice were infected with C. rodentium. Rag1 KO mice did not exhibit the reduction in the number of goblet cells or in mediator (Muc2 and Tff3) expression observed in infected immunocompetent mice. However, reconstitution of Rag1 KO mice with T and B lymphocytes from C57BL/6 mice restored the goblet cell depletion phenotype during C. rodentium infection. In conclusion, these studies demonstrated that while colonic goblet cells can be subject to direct infection and potential subversion by A/E pathogens in vivo, it is the host immune system that primarily modulates the function of these cells during infection.


Subject(s)
Citrobacter rodentium/physiology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Goblet Cells/immunology , Goblet Cells/microbiology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Colon/pathology , Female , Gene Expression , Homeodomain Proteins/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mucin-2 , Mucins/biosynthesis , Mucins/genetics , T-Lymphocytes/immunology , Trefoil Factor-3
SELECTION OF CITATIONS
SEARCH DETAIL
...