Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Front Immunol ; 14: 1277637, 2023.
Article En | MEDLINE | ID: mdl-38022673

Peyer's patches (PPs) are specialized gut-associated lymphoid tissues that initiate follicular helper T (Tfh)-mediated immunoglobulin A (IgA) response to luminal antigens derived from commensal symbionts, pathobionts, and dietary sources. IgA-producing B cells migrate from PPs to the small intestinal lamina propria and secrete IgA across the epithelium, modulating the ecological balance of the commensal microbiota and neutralizing pathogenic microorganisms. α-glucosidase inhibitors (α-GIs) are antidiabetic drugs that inhibit carbohydrate digestion in the small intestinal epithelium, leading to alterations in the commensal microbiota composition and metabolic activity. The commensal microbiota and IgA responses exhibit bidirectional interactions that modulate intestinal homeostasis and immunity. However, the effect of α-GIs on the intestinal IgA response remains unclear. We investigated whether α-GIs affect IgA responses by administering voglibose and acarbose to mice via drinking water. We analyzed Tfh cells, germinal center (GC) B cells, and IgA-producing B cells in PPs by flow cytometry. We also assessed pathogen-specific IgA responses. We discovered that voglibose and acarbose induced Tfh cells, GCB cells, and IgA-producing B cells in the PPs of the proximal small intestine in mice. This effect was attributed to the modification of the microbiota rather than a shortage of monosaccharides. Furthermore, voglibose enhanced secretory IgA (S-IgA) production against attenuated Salmonella Typhimurium. Our findings reveal a novel mechanism by which α-GIs augment antigen-specific IgA responses by stimulating Tfh-GCB responses in PPs, and suggest a potential therapeutic application as an adjuvant for augmenting mucosal vaccines.


Glycoside Hydrolase Inhibitors , Immunoglobulin A , Animals , Mice , Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolase Inhibitors/metabolism , Peyer's Patches , Acarbose/metabolism , Antigens/metabolism
2.
Front Allergy ; 3: 996657, 2022.
Article En | MEDLINE | ID: mdl-36277615

Food allergy is a type I allergic reaction induced by mast cells and is mainly activated by allergen-specific immunoglobulin (Ig)E. Spi-B is an E26-transformation-specific (Ets) family transcription factor essential for the differentiation and functional maturation of several immune cell subsets, including mast cells. However, the possible involvement of Spi-B in food allergy remains unclear. In this study, we found that Spi-B-deficient mice were highly susceptible to food allergy to ovalbumin (OVA), as indicated by the exacerbation of diarrhea and elevation of serum IgE levels. These pathological changes were associated with enhanced mast cell infiltration into the intestinal lamina propria. Activation of mast cells in the intestinal mucosa was observed in Spib -/- mice, even under physiological conditions. Accordingly, Spi-B deficiency increased the translocation of fluorescently labeled dextran from the lumen to the serum, suggesting increased intestinal permeability in Spib -/- mice. Moreover, Spib -/- mice showed defects in oral tolerance induction to OVA. These data illustrate that Spi-B suppresses the development of food allergies by controlling the activation of intestinal mast cells and by inducing immune tolerance to food allergens.

3.
Cell Rep ; 40(3): 111087, 2022 07 19.
Article En | MEDLINE | ID: mdl-35858544

Microbiota-accessible carbohydrates (MACs) exert health-promoting effects, but how each MAC impacts gut microbiota and regulates host physiology remains unclear. Here, we show that l-arabinose and sucrose cooperatively act on gut microbiota and exert anti-obesogenic effects. Specifically, l-arabinose, a monosaccharide that is poorly absorbed in the gut and inhibits intestinal sucrase, suppresses diet-induced obesity in mice in the presence of sucrose. Additionally, the suppressive effect of l-arabinose on adiposity is abrogated in mice lacking the short-chain fatty acid (SCFA) receptors GPR43 and GPR41. Mechanistically, l-arabinose increases the relative abundance of acetate and propionate producers (e.g., Bacteroides), while sucrose enhances SCFA production. Furthermore, l-arabinose and sucrose activate the glycolytic and pentose phosphate pathways of Bacteroides, respectively, indicating that they synergistically promote acetate production through distinct pathways. These findings suggest that each MAC has a unique property and thus may serve as a precision gut-microbiota modulator to promote host homeostasis.


Gastrointestinal Microbiome , Microbiota , Animals , Arabinose/pharmacology , Bacteroides/metabolism , Carbohydrates , Fatty Acids, Volatile/metabolism , Mice , Obesity/metabolism , Sucrose
4.
Nutrients ; 13(8)2021 Aug 16.
Article En | MEDLINE | ID: mdl-34444972

Metabolic syndrome (MetS) is a multifactorial chronic metabolic disorder that affects approximately one billion people worldwide. Recent studies have evaluated whether targeting the gut microbiota can prevent MetS. This study aimed to assess the ability of dietary fiber to control MetS by modulating gut microbiota composition. Sodium alginate (SA) is a seaweed-derived dietary fiber that suppresses high-fat diet (HFD)-induced MetS via an effect on the gut microbiota. We observed that SA supplementation significantly decreased body weight gain, cholesterol levels, and fat weight, while improving glucose tolerance in HFD-fed mice. SA changed the gut microbiota composition and significantly increased the abundance of Bacteroides. Antibiotic treatment completely abolished the suppressive effects of SA on MetS. Mechanistically, SA decreased the number of colonic inflammatory monocytes, which promote MetS development, in a gut microbiota-dependent manner. The abundance of Bacteroides was negatively correlated with that of inflammatory monocytes and positively correlated with the levels of several gut metabolites. The present study revealed a novel food function of SA in preventing HFD-induced MetS through its action on gut microbiota.


Alginates/pharmacology , Gastrointestinal Microbiome/drug effects , Metabolic Syndrome/metabolism , Monocytes/drug effects , Seaweed , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Dietary Fiber/pharmacology , Inflammation/metabolism , Mice , Mice, Inbred C57BL
5.
EBioMedicine ; 58: 102913, 2020 Aug.
Article En | MEDLINE | ID: mdl-32711255

BACKGROUND: Rheumatoid arthritis (RA) is a chronic debilitating autoimmune disorder with a high prevalence, especially in industrialized countries. Dysbiosis of the intestinal microbiota has been observed in RA patients. For instance, new-onset untreated RA (NORA) is associated with the underrepresentation of the Clostridium cluster XIVa, including Lachnospiraceae, which are major butyrate producers, although the pathological relevance has remained obscure. Follicular regulatory T (TFR) cells play critical regulatory roles in the pathogenesis of autoimmune diseases, including RA. Reduced number of circulating TFR cells has been associated with the elevation of autoantibodies and disease severity in RA. However, the contribution of commensal microbe-derived butyrate in controlling TFR cell differentiation remains unknown. METHODS: We examined the contribution of microbe-derived butyrate in controlling autoimmune arthritis using collagen-induced arthritis (CIA) and SKG arthritis models. We phenotyped autoimmune responses in the gut-associated lymphoid tissues (GALT) in the colon and joint-draining lymph nodes in the CIA model. We developed an in vitro CXCR5+Bcl-6+Foxp3+ TFR (iTFR) cell culture system and examined whether butyrate promotes the differentiation of iTFR cells. FINDINGS: Microbe-derived butyrate suppressed the development of autoimmune arthritis. The immunization of type II collagen (CII) caused hypertrophy of the GALT in the colon by amplifying the GC reaction prior to the onset of the CIA. Butyrate mitigated these pathological events by promoting TFR cell differentiation. Butyrate directly induced the differentiation of functional TFR cells in vitro by enhancing histone acetylation in TFR cell marker genes. This effect was attributed to histone deacetylase (HDAC) inhibition by butyrate, leading to histone hyperacetylation in the promoter region of the TFR-cell marker genes. The adoptive transfer of the butyrate-treated iTFR cells reduced CII-specific autoantibody production and thus ameliorated the symptoms of arthritis. INTERPRETATION: Accordingly, microbiota-derived butyrate serves as an environmental cue to enhance TFR cells, which suppress autoantibody production in the systemic lymphoid tissue, eventually ameliorating RA. Our findings provide mechanistic insights into the link between the gut environment and RA risk. FUNDING: This work was supported by AMED-Crest (16gm1010004h0101, 17gm1010004h0102, 18gm1010004h0103, and 19gm1010004s0104 to KH), the Japan Society for the Promotion of Science (JP17KT0055, JP16H01369, and JP18H04680 to KH; JP17K15734 to DT), Keio University Special Grant-in-Aid for Innovative Collaborative Research Projects (KH), Keio Gijuku Fukuzawa Memorial Fund for the Advancement of Education and Research (DT), the SECOM Science and Technology Foundation (KH), the Cell Science Research Foundation (KH), the Mochida Memorial Foundation for Medical and Pharmaceutical Research (DT), the Suzuken Memorial Foundation (KH and DT), the Takeda Science Foundation (KH and DT), The Science Research Promotion Fund, and The Promotion and Mutual Aid Corporation for Private Schools of Japan (KH).


Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , Bacteria/metabolism , Butyrates/pharmacology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/transplantation , Acetylation , Adoptive Transfer , Aged , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/immunology , Autoimmunity , Cell Differentiation/drug effects , Cells, Cultured , Gastrointestinal Microbiome , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Lymphoid Tissue/cytology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Middle Aged , T-Lymphocytes, Regulatory/drug effects
6.
Mucosal Immunol ; 13(4): 679-690, 2020 07.
Article En | MEDLINE | ID: mdl-32042052

Infectious colitis is one of the most common health issues worldwide. Microfold (M) cells actively transport luminal antigens to gut-associated lymphoid tissue to induce IgA responses; however, it remains unknown whether M cells contribute to the induction of cellular immune responses. Here we report that M cell-dependent antigen transport plays a critical role in the induction of Th1, Th17, and Th22 responses against gut commensals in the steady state. The establishment of commensal-specific cellular immunity was a prerequisite for preventing bacterial dissemination during enteropathogenic Citrobacter rodentium infection. Therefore, M cell-null mice developed severe colitis with increased bacterial dissemination. This abnormality was associated with mucosal barrier dysfunction. These observations suggest that antigen transport by M cells may help maintain gut immune homeostasis by eliciting antigen-specific cellular immune responses.


Antigens/immunology , Antigens/metabolism , Colitis/etiology , Colitis/metabolism , Immunity, Cellular , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Colitis/pathology , Disease Models, Animal , Disease Susceptibility , Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions/immunology , Immunoglobulin A/immunology , Intestinal Mucosa/cytology , Mice , Mice, Knockout , Organ Specificity/immunology , Peyer's Patches/immunology , Peyer's Patches/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
7.
Int Immunol ; 32(4): 243-258, 2020 04 12.
Article En | MEDLINE | ID: mdl-31858119

Secretory immunoglobulin A (SIgA), the most abundant antibody isotype in the body, maintains a mutual relationship with commensal bacteria and acts as a primary barrier at the mucosal surface. Colonization by commensal bacteria induces an IgA response, at least partly through a T-cell-independent process. However, the mechanism underlying the commensal-bacteria-induced T-cell-independent IgA response has yet to be fully clarified. Here, we show that commensal-bacteria-derived butyrate promotes T-cell-independent IgA class switching recombination (CSR) in the mouse colon. Notably, the butyrate concentration in human stools correlated positively with the amount of IgA. Butyrate up-regulated the production of transforming growth factor ß1 and all-trans retinoic acid by CD103+CD11b+ dendritic cells, both of which are critical for T-cell-independent IgA CSR. This effect was mediated by G-protein-coupled receptor 41 (GPR41/FFA3) and GPR109a/HCA2, and the inhibition of histone deacetylase. The butyrate-induced IgA response reinforced the colonic barrier function, preventing systemic bacterial dissemination under inflammatory conditions. These observations demonstrate that commensal-bacteria-derived butyrate contributes to the maintenance of the gut immune homeostasis by facilitating the T-cell-independent IgA response in the colon.


Butyrates/pharmacology , Colon/drug effects , Immunoglobulin A/immunology , T-Lymphocytes/drug effects , Animals , Cells, Cultured , Coculture Techniques , Colon/immunology , Humans , Immunoglobulin Class Switching/drug effects , Immunoglobulin Class Switching/immunology , Male , Mice , Mice, Inbred Strains , Mice, Knockout , T-Lymphocytes/immunology
8.
EBioMedicine ; 48: 513-525, 2019 Oct.
Article En | MEDLINE | ID: mdl-31521614

BACKGROUND: The dysbiosis of gut microbiota has been implicated in the pathogenesis of inflammatory bowel diseases; however, the underlying mechanisms have not yet been elucidated. Heavily glycosylated mucin establishes a first-line barrier against pathogens and serves as a niche for microbial growth. METHODS: To elucidate relationships among dysbiosis, abnormal mucin utilisation, and microbial metabolic dysfunction, we analysed short-chain fatty acids (SCFAs) and mucin components in stool samples of 40 healthy subjects, 49 ulcerative colitis (UC) patients, and 44 Crohn's disease (CD) patients from Japan. FINDINGS: Levels of n-butyrate were significantly lower in stools of both CD and UC patients than in stools of healthy subjects. Correlation analysis identified seven bacterial species positively correlated with n-butyrate levels; the major n-butyrate producer, Faecalibacterium prausnitzii, was particularly underrepresented in CD patients, but not in UC patients. In UC patients, there were inverse correlations between mucin O-glycan levels and the production of SCFAs, such as n-butyrate, suggesting that mucin O-glycans serve as an endogenous fermentation substrate for n-butyrate production. Indeed, mucin-fed rodents exhibited enhanced n-butyrate production, leading to the expansion of RORgt+Treg cells and IgA-producing cells in colonic lamina propria. Microbial utilisation of mucin-associated O-glycans was significantly reduced in n-butyrate-deficient UC patients. INTERPRETATION: Mucin O-glycans facilitate symbiosynthesis of n-butyrate by gut microbiota. Abnormal mucin utilisation may lead to reduced n-butyrate production in UC patients. FUND: Japan Society for the Promotion of Science, Health Labour Sciences Research Grant, AMED-Crest, AMED, Yakult Foundation, Keio Gijuku Academic Development Funds, The Aashi Grass Foundation, and The Canon Foundation.


Homeostasis , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mucins/metabolism , Polysaccharides/metabolism , Adult , Animals , Biomarkers , Butyrates/metabolism , Case-Control Studies , Female , Gastrointestinal Microbiome , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Male , Metagenome , Metagenomics , Mice , Middle Aged , Symbiosis
9.
Cell ; 178(5): 1072-1087.e14, 2019 08 22.
Article En | MEDLINE | ID: mdl-31442401

Nutritional status potentially influences immune responses; however, how nutritional signals regulate cellular dynamics and functionality remains obscure. Herein, we report that temporary fasting drastically reduces the number of lymphocytes by ∼50% in Peyer's patches (PPs), the inductive site of the gut immune response. Subsequent refeeding seemingly restored the number of lymphocytes, but whose cellular composition was conspicuously altered. A large portion of germinal center and IgA+ B cells were lost via apoptosis during fasting. Meanwhile, naive B cells migrated from PPs to the bone marrow during fasting and then back to PPs during refeeding when stromal cells sensed nutritional signals and upregulated CXCL13 expression to recruit naive B cells. Furthermore, temporal fasting before oral immunization with ovalbumin abolished the induction of antigen-specific IgA, failed to induce oral tolerance, and eventually exacerbated food antigen-induced diarrhea. Thus, nutritional signals are critical in maintaining gut immune homeostasis.


B-Lymphocytes/physiology , Immunity, Mucosal , Animals , Antigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Fasting , Gene Expression Regulation , Glycolysis , Immunoglobulin A/metabolism , Male , Mice , Mice, Inbred BALB C , Nutritional Status , Ovalbumin/immunology , Peyer's Patches/immunology , Peyer's Patches/metabolism , Peyer's Patches/pathology , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , TOR Serine-Threonine Kinases/metabolism
10.
EMBO Rep ; 15(12): 1297-304, 2014 Dec.
Article En | MEDLINE | ID: mdl-25378482

Intrinsic Notch signaling in intestinal epithelial cells restricts secretory cell differentiation. In gut-associated lymphoid tissue (GALT), stromal cells located beneath the follicle-associated epithelium (FAE) abundantly express the Notch ligand delta-like 1 (Dll1). Here, we show that mice lacking Rbpj-a gene encoding a transcription factor implicated in Notch signaling-in intestinal epithelial cells have defective GALT maturation. This defect can be attributed to the expansion of goblet cells, which leads to the down-regulation of CCL20 in FAE. These data demonstrate that epithelial Notch signaling maintained by stromal cells contributes to the full maturation of GALT by restricting secretory cell differentiation in FAE.


Epithelial Cells/cytology , Epithelial Cells/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Receptors, Notch/metabolism , Stromal Cells/cytology , Animals , Cell Differentiation/physiology , In Vitro Techniques , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Signal Transduction/physiology , Stromal Cells/metabolism
11.
Nat Immunol ; 15(6): 571-9, 2014 Jun.
Article En | MEDLINE | ID: mdl-24777532

Intestinal regulatory T cells (Treg cells) are necessary for the suppression of excessive immune responses to commensal bacteria. However, the molecular machinery that controls the homeostasis of intestinal Treg cells has remained largely unknown. Here we report that colonization of germ-free mice with gut microbiota upregulated expression of the DNA-methylation adaptor Uhrf1 in Treg cells. Mice with T cell-specific deficiency in Uhrf1 (Uhrf1(fl/fl)Cd4-Cre mice) showed defective proliferation and functional maturation of colonic Treg cells. Uhrf1 deficiency resulted in derepression of the gene (Cdkn1a) that encodes the cyclin-dependent kinase inhibitor p21 due to hypomethylation of its promoter region, which resulted in cell-cycle arrest of Treg cells. As a consequence, Uhrf1(fl/fl)Cd4-Cre mice spontaneously developed severe colitis. Thus, Uhrf1-dependent epigenetic silencing of Cdkn1a was required for the maintenance of gut immunological homeostasis. This mechanism enforces symbiotic host-microbe interactions without an inflammatory response.


Colitis/immunology , Colon/immunology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Epigenesis, Genetic , Nuclear Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CCAAT-Enhancer-Binding Proteins , Cell Cycle Checkpoints , Cell Proliferation , Cells, Cultured , Clostridium/immunology , Colitis/genetics , Colon/microbiology , DNA Methylation , Gene Expression Profiling , Interleukin-2 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbiota/immunology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering , Symbiosis/immunology , Ubiquitin-Protein Ligases , Up-Regulation
12.
Nature ; 504(7480): 446-50, 2013 Dec 19.
Article En | MEDLINE | ID: mdl-24226770

Gut commensal microbes shape the mucosal immune system by regulating the differentiation and expansion of several types of T cell. Clostridia, a dominant class of commensal microbe, can induce colonic regulatory T (Treg) cells, which have a central role in the suppression of inflammatory and allergic responses. However, the molecular mechanisms by which commensal microbes induce colonic Treg cells have been unclear. Here we show that a large bowel microbial fermentation product, butyrate, induces the differentiation of colonic Treg cells in mice. A comparative NMR-based metabolome analysis suggests that the luminal concentrations of short-chain fatty acids positively correlates with the number of Treg cells in the colon. Among short-chain fatty acids, butyrate induced the differentiation of Treg cells in vitro and in vivo, and ameliorated the development of colitis induced by adoptive transfer of CD4(+) CD45RB(hi) T cells in Rag1(-/-) mice. Treatment of naive T cells under the Treg-cell-polarizing conditions with butyrate enhanced histone H3 acetylation in the promoter and conserved non-coding sequence regions of the Foxp3 locus, suggesting a possible mechanism for how microbial-derived butyrate regulates the differentiation of Treg cells. Our findings provide new insight into the mechanisms by which host-microbe interactions establish immunological homeostasis in the gut.


Butyrates/metabolism , Cell Differentiation , Colon/immunology , Colon/microbiology , Fermentation , Symbiosis , T-Lymphocytes, Regulatory/cytology , Acetylation/drug effects , Adoptive Transfer , Animals , Butyrates/analysis , Butyrates/pharmacology , Cell Differentiation/drug effects , Colitis/drug therapy , Colitis/pathology , Colon/cytology , Colon/metabolism , Conserved Sequence , Female , Forkhead Transcription Factors/genetics , Germ-Free Life , Histones/metabolism , Homeostasis/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocyte Count , Magnetic Resonance Spectroscopy , Male , Metabolome , Mice , Promoter Regions, Genetic/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
13.
Gastroenterology ; 145(3): 625-35, 2013 Sep.
Article En | MEDLINE | ID: mdl-23684748

BACKGROUND & AIMS: In epithelial cells, protein sorting mechanisms regulate localization of plasma membrane proteins that generate and maintain cell polarity. The clathrin-adaptor protein (AP) complex AP-1B is expressed specifically in polarized epithelial cells, where it regulates basolateral sorting of membrane proteins. However, little is known about its physiological significance. METHODS: We analyzed the intestinal epithelia of mice deficient in Ap1m2 (Ap1m2(-/-) mice), which encodes the AP-1B µ1B subunit, and compared it with 129/B6/CD1 littermates (controls). Notch signaling was inhibited by intraperitoneal injection of dibenzazepine, and ß-catenin signaling was inhibited by injection of IWR1. Intestinal tissue samples were collected and analyzed by immunofluorescence analysis. RESULTS: Ap1m2(-/-) mice developed intestinal epithelial cell hyperplasia. The polarity of intestinal epithelial cells was disrupted, as indicated by the appearance of ectopic microvilli-like structures on the lateral plasma membrane and mislocalization of basolateral membrane proteins, including the low-density lipoprotein receptor and E-cadherin. The E-cadherin-ß-catenin complex therefore was disrupted at the adherens junction, resulting in nuclear translocation of ß-catenin. This resulted in up-regulation of genes regulated by ß-catenin/transcription factor 4 (Tcf4) complex, and increased the proliferation of intestinal epithelial cells. CONCLUSIONS: AP-1B is required for protein sorting and polarization of intestinal cells in mice. Loss of AP-1B in the intestinal epithelia results in mislocalization of E-cadherin, activation of ß-catenin/Tcf4 complex, proliferation, and hyperplasia.


Adaptor Protein Complex 1/deficiency , Adaptor Protein Complex mu Subunits/deficiency , Cell Polarity , Cell Proliferation , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Adaptor Protein Complex 1/physiology , Adaptor Protein Complex mu Subunits/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Biomarkers/metabolism , Cadherins/metabolism , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , Fluorescent Antibody Technique , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Intestine, Small/pathology , Intestine, Small/physiopathology , Male , Mice , Mice, 129 Strain , Mice, Knockout , Transcription Factor 4 , beta Catenin/metabolism
14.
J Immunol ; 188(5): 2427-36, 2012 Mar 01.
Article En | MEDLINE | ID: mdl-22279105

Intestinal epithelial cells (IECs) have important functions as the first line of defense against diverse microorganisms on the luminal surface. Impaired integrity of IEC has been implicated in increasing the risk for inflammatory disorders in the gut. Notch signaling plays a critical role in the maintenance of epithelial integrity by regulating the balance of secretory and absorptive cell lineages, and also by facilitating epithelial cell proliferation. We show in this article that mice harboring IEC-specific deletion of Rbpj (RBP-J(ΔIEC)), a transcription factor that mediates signaling through Notch receptors, spontaneously develop chronic colitis characterized by the accumulation of Th17 cells in colonic lamina propria. Intestinal bacteria are responsible for the development of colitis, because their depletion with antibiotics prevented the development of colitis in RBP-J(ΔIEC) mice. Furthermore, bacterial translocation was evident in the colonic mucosa of RBP-J(ΔIEC) mice before the onset of colitis, suggesting attenuated epithelial barrier functions in these mice. Indeed, RBP-J(ΔIEC) mice displayed increase in intestinal permeability after rectal administration of FITC-dextran. In addition to the defect in physical barrier, loss of Notch signaling led to arrest of epithelial cell turnover caused by downregulation of Hes1, a transcriptional repressor of p27(Kip1) and p57(Kip2). Thus, epithelial cell-intrinsic Notch signaling ensures integrity and homeostasis of IEC, and this mechanism is required for containment of intestinal inflammation.


Homeostasis/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Receptors, Notch/physiology , Signal Transduction/immunology , Animals , Bacterial Translocation/genetics , Bacterial Translocation/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Chronic Disease , Colitis/genetics , Colitis/immunology , Colitis/pathology , Gene Deletion , Homeostasis/genetics , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Notch/genetics , Signal Transduction/genetics , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/pathology
15.
Nature ; 462(7270): 226-30, 2009 Nov 12.
Article En | MEDLINE | ID: mdl-19907495

The mucosal immune system forms the largest part of the entire immune system, containing about three-quarters of all lymphocytes and producing grams of secretory IgA daily to protect the mucosal surface from pathogens. To evoke the mucosal immune response, antigens on the mucosal surface must be transported across the epithelial barrier into organized lymphoid structures such as Peyer's patches. This function, called antigen transcytosis, is mediated by specialized epithelial M cells. The molecular mechanisms promoting this antigen uptake, however, are largely unknown. Here we report that glycoprotein 2 (GP2), specifically expressed on the apical plasma membrane of M cells among enterocytes, serves as a transcytotic receptor for mucosal antigens. Recombinant GP2 protein selectively bound a subset of commensal and pathogenic enterobacteria, including Escherichia coli and Salmonella enterica serovar Typhimurium (S. Typhimurium), by recognizing FimH, a component of type I pili on the bacterial outer membrane. Consistently, these bacteria were colocalized with endogenous GP2 on the apical plasma membrane as well as in cytoplasmic vesicles in M cells. Moreover, deficiency of bacterial FimH or host GP2 led to defects in transcytosis of type-I-piliated bacteria through M cells, resulting in an attenuation of antigen-specific immune responses in Peyer's patches. GP2 is therefore a previously unrecognized transcytotic receptor on M cells for type-I-piliated bacteria and is a prerequisite for the mucosal immune response to these bacteria. Given that M cells are considered a promising target for oral vaccination against various infectious diseases, the GP2-dependent transcytotic pathway could provide a new target for the development of M-cell-targeted mucosal vaccines.


Adhesins, Escherichia coli/metabolism , Antigens, Bacterial/metabolism , Epithelial Cells/immunology , Fimbriae Proteins/metabolism , Immunity, Mucosal/immunology , Membrane Glycoproteins/metabolism , Peyer's Patches/cytology , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/immunology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Cell Line , Epithelial Cells/metabolism , Escherichia coli/immunology , Escherichia coli/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , GPI-Linked Proteins , Glycoproteins , HeLa Cells , Humans , Intestines/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peyer's Patches/immunology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism , Substrate Specificity
...