Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nature ; 595(7868): 560-564, 2021 07.
Article in English | MEDLINE | ID: mdl-34262176

ABSTRACT

The balance between bacterial colonization and its containment in the intestine is indispensable for the symbiotic relationship between humans and their bacteria. One component to maintain homeostasis at the mucosal surfaces is immunoglobulin A (IgA), the most abundant immunoglobulin in mammals1,2. Several studies have revealed important characteristics of poly-reactive IgA3,4, which is produced naturally without commensal bacteria. Considering the dynamic changes within the gut environment, however, it remains uncertain how the commensal-reactive IgA pool is shaped and how such IgA affects the microbial community. Here we show that acetate-one of the major gut microbial metabolites-not only increases the production of IgA in the colon, but also alters the capacity of the IgA pool to bind to specific microorganisms including Enterobacterales. Induction of commensal-reactive IgA and changes in the IgA repertoire by acetate were observed in mice monocolonized with Escherichia coli, which belongs to Enterobacterales, but not with the major commensal Bacteroides thetaiotaomicron, which suggests that acetate directs selective IgA binding to certain microorganisms. Mechanistically, acetate orchestrated the interactions between epithelial and immune cells, induced microbially stimulated CD4 T cells to support T-cell-dependent IgA production and, as a consequence, altered the localization of these bacteria within the colon. Collectively, we identified a role for gut microbial metabolites in the regulation of differential IgA production to maintain mucosal homeostasis.


Subject(s)
Acetates/pharmacology , Bacteria/immunology , Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Colon/immunology , Diet , Fatty Acids, Volatile/metabolism , Homeostasis/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Symbiosis
2.
Immunity ; 46(5): 863-874.e4, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514691

ABSTRACT

Mast cells are important for eradication of intestinal nematodes; however, their precise mechanisms of action have remained elusive, especially in the early phase of infection. We found that Spi-B-deficient mice had increased numbers of mast cells and rapidly expelled the Heligmosomoides polygyrus (Hp) nematode. This was accompanied by induction of interleukin-13 (IL-13)-producing group 2 innate lymphoid cells (ILC2) and goblet cell hyperplasia. Immediately after Hp infection, mast cells were rapidly activated to produce IL-33 in response to ATP released from apoptotic intestinal epithelial cells. In vivo inhibition of the P2X7 ATP receptor rendered the Spi-B-deficient mice susceptible to Hp, concomitant with elimination of mast cell activation and IL-13-producing ILC2 induction. These results uncover a previously unknown role for mast cells in innate immunity in that activation of mast cells by ATP orchestrates the development of a protective type 2 immune response, in part by producing IL-33, which contributes to ILC2 activation.


Subject(s)
Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Immunity, Innate , Lymphocyte Subsets/immunology , Mast Cells/immunology , Adenosine Triphosphate/metabolism , Animals , Cell Communication , Cell Differentiation , Disease Models, Animal , Disease Resistance/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression , Helminthiasis/genetics , Immunophenotyping , Interleukin-33/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/metabolism , Male , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Knockout , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Purinergic P2X7/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Nat Immunol ; 13(8): 729-36, 2012 Jun 17.
Article in English | MEDLINE | ID: mdl-22706340

ABSTRACT

Intestinal microfold cells (M cells) are an enigmatic lineage of intestinal epithelial cells that initiate mucosal immune responses through the uptake and transcytosis of luminal antigens. The mechanisms of M-cell differentiation are poorly understood, as the rarity of these cells has hampered analysis. Exogenous administration of the cytokine RANKL can synchronously activate M-cell differentiation in mice. Here we show the Ets transcription factor Spi-B was induced early during M-cell differentiation. Absence of Spi-B silenced the expression of various M-cell markers and prevented the differentiation of M cells in mice. The activation of T cells via an oral route was substantially impaired in the intestine of Spi-B-deficient (Spib(-/-)) mice. Our study demonstrates that commitment to the intestinal M-cell lineage requires Spi-B as a candidate master regulator.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Intestinal Mucosa/cytology , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Animals , Cell Lineage , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Immunity, Mucosal/genetics , Intestinal Mucosa/embryology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , RANK Ligand/pharmacology , T-Lymphocytes/immunology
4.
Traffic ; 21(1): 34-44, 2020 01.
Article in English | MEDLINE | ID: mdl-31647148

ABSTRACT

Mucosal immune responses in the inductive lymphoid tissues of the intestine begin with uptake of particulate antigens, including components of the gut microbiota by specialized antigen sampling M cells. M cells represent a distinct lineage of enterocytes that arise from crypt stem cells in response to the cytokine receptor of NF-κB ligand (RANKL). Full differentiation of M cells requires the transcription factor Spi-B to yield mature M cells that express multiple receptors for bacteria including glycoprotein 2. M cell differentiation can be recapitulated in vitro using three-dimensional enteroid cultures of primary intestinal stem cells supplemented with RANKL. This article summarizes the current knowledge about the genesis of intestinal M cells and highlights some of the remaining unanswered questions about this enigmatic cell type.


Subject(s)
Microbiota , RANK Ligand , Cell Differentiation , Humans , Immunity, Mucosal , Intestinal Mucosa
5.
Int Immunol ; 29(8): 357-363, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28992252

ABSTRACT

Bacterial access to the gut immune system is a crucial process to promote host immune responses. The probiotic L-92 strain of Lactobacillus acidophilus exerts anti-allergic immunomodulatory effects upon oral administration in mice. Here, we show that microfold cells (M cells) are responsible for L-92 internalization for evoking L-92-mediated immune responses. L-92 specifically bound to uromodulin, a glycosylphosphatidylinositol-anchored protein expressed exclusively on M cells among intestinal epithelial cells. Internalization of L-92 into M cells was significantly reduced in uromodulin-deficient (Umod-/-) mice compared to Umod+/+ mice. Furthermore, the binding of L-92 to uromodulin was significantly decreased after removal of surface layer protein A (SlpA) from the bacteria. Our study thus revealed a crucial role of uromodulin on the M-cell surface for the uptake of SlpA-positive lactic acid bacteria into M cells, possibly leading to subsequent delivery of the bacteria to dendritic cells closely associated with M cells for immunomodulation. Our study also shed light on the possibility that SlpA and uromodulin could be used as vehicle and target, respectively, for efficient mucosal vaccine delivery.


Subject(s)
Bacterial Proteins/metabolism , Dendritic Cells/immunology , Intestinal Mucosa/metabolism , Lactobacillus acidophilus/physiology , Uromodulin/metabolism , Animals , Cells, Cultured , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Probiotics , Protein Binding , Transcriptome , Uromodulin/genetics
7.
J Immunol ; 188(5): 2427-36, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22279105

ABSTRACT

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.


Subject(s)
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
8.
Cell Metab ; 35(2): 361-375.e9, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36652945

ABSTRACT

Although recent studies have highlighted the impact of gut microbes on the progression of obesity and its comorbidities, it is not fully understood how these microbes promote these disorders, especially in terms of the role of microbial metabolites. Here, we report that Fusimonas intestini, a commensal species of the family Lachnospiraceae, is highly colonized in both humans and mice with obesity and hyperglycemia, produces long-chain fatty acids such as elaidate, and consequently facilitates diet-induced obesity. High fat intake altered the expression of microbial genes involved in lipid production, such as the fatty acid metabolism regulator fadR. Monocolonization with a FadR-overexpressing Escherichia coli exacerbated the metabolic phenotypes, suggesting that the change in bacterial lipid metabolism is causally involved in disease progression. Mechanistically, the microbe-derived fatty acids impaired intestinal epithelial integrity to promote metabolic endotoxemia. Our study thus provides a mechanistic linkage between gut commensals and obesity through the overproduction of microbe-derived lipids.


Subject(s)
Fatty Acids , Gastrointestinal Microbiome , Humans , Animals , Mice , Diet, High-Fat , Obesity/metabolism , Bacteria/genetics , Mice, Inbred C57BL
9.
Am J Pathol ; 179(3): 1301-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21763679

ABSTRACT

Transmissible spongiform encephalopathies are fatal neurodegenerative diseases. Infection by the oral route is assumed to be important, although its pathogenesis is not understood. Using prion protein (PrP) knockout mice, we investigated the sequence of events during the invasion of orally administered PrPs through the intestinal mucosa and the spread into lymphoid tissues and the peripheral nervous system. Orally administered PrPs were incorporated by intestinal epitheliocytes in the follicle-associated epithelium and villi within 1 hour. PrP-positive cells accumulated in the subfollicle region of Peyer's patches a few hours thereafter. PrP-positive cells spread toward the mesenteric lymph nodes and spleen after the accumulation of PrPs in the Peyer's patches. The number of PrP molecules in the mesenteric lymph nodes and spleen peaked at 2 days and 6 days after inoculation, respectively. The epitheliocytes in the follicle-associated epithelium incorporating PrPs were annexin V-positive microfold cells and PrP-positive cells in Peyer's patches and spleen were CD11b-positive and CD14-positive macrophages. Additionally, PrP-positive cells in Peyer's patches and spleen were detected in the vicinity of peripheral nerve fibers in the early stages of infection. These results indicate that orally delivered PrPs were incorporated by microfold cells promptly after challenge and that macrophages might act as a transporter of incorporated PrPs from the Peyer's patches to other lymphoid tissues and the peripheral nervous system.


Subject(s)
Brain/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , Peyer's Patches/metabolism , Prions/pharmacokinetics , Administration, Oral , Animals , Immunohistochemistry , Intestine, Small/metabolism , Lymph Nodes/metabolism , Male , Mesentery/metabolism , Mice , Mice, Knockout , Peyer's Patches/pathology , Prion Diseases/etiology , Prions/administration & dosage , Spleen/metabolism
10.
Sci Immunol ; 7(76): eabj8760, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36269840

ABSTRACT

Invariant natural killer T (iNKT) cells are a group of innate-like T lymphocytes that recognize lipid antigens. They are supposed to be tissue resident and important for systemic and local immune regulation. To investigate the heterogeneity of iNKT cells, we recharacterized iNKT cells in the thymus and peripheral tissues. iNKT cells in the thymus were divided into three subpopulations by the expression of the natural killer cell receptor CD244 and the chemokine receptor CXCR6 and designated as C0 (CD244-CXCR6-), C1 (CD244-CXCR6+), or C2 (CD244+CXCR6+) iNKT cells. The development and maturation of C2 iNKT cells from C0 iNKT cells strictly depended on IL-15 produced by thymic epithelial cells. C2 iNKT cells expressed high levels of IFN-γ and granzymes and exhibited more NK cell-like features, whereas C1 iNKT cells showed more T cell-like characteristics. C2 iNKT cells were influenced by the microbiome and aging and suppressed the expression of the autoimmune regulator AIRE in the thymus. In peripheral tissues, C2 iNKT cells were circulating that were distinct from conventional tissue-resident C1 iNKT cells. Functionally, C2 iNKT cells protected mice from the tumor metastasis of melanoma cells by enhancing antitumor immunity and promoted antiviral immune responses against influenza virus infection. Furthermore, we identified human CD244+CXCR6+ iNKT cells with high cytotoxic properties as a counterpart of mouse C2 iNKT cells. Thus, this study reveals a circulating subset of iNKT cells with NK cell-like properties distinct from conventional tissue-resident iNKT cells.


Subject(s)
Natural Killer T-Cells , Mice , Humans , Animals , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Interleukin-15 , Antiviral Agents , Granzymes , Receptors, Natural Killer Cell , Receptors, Chemokine/metabolism , Lipids
11.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G442-53, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193527

ABSTRACT

Microfold (M) cells in the follicle-associated epithelium (FAE) of Peyer's patches have an important role in mucosal immune responses. A primary difficulty for investigations of bovine M cells is the lack of a specific molecular marker. To identify such a marker, we investigated the expression of several kinds of intermediate filament proteins using calf Peyer's patches. The expression patterns of cytokeratin (CK) 18 in jejunal and ileal FAE were very similar to the localization pattern of M cells recognized by scanning electron microscopy. Mirror sections revealed that jejunal CK18-positive cells had irregular and sparse microvilli, as well as pocket-like structures containing lymphocytes, typical morphological characteristic of M cells. However, CK18-negative cells had regular and dense microvilli on their surface, typical of the morphology of enterocytes. In contrast, CK20 immunoreactivity was detected in almost all villous epithelial cells and CK18-negative cells in the FAE. CK18-positive proliferating transit-amplifying cells in the crypt exchanged CK18 for CK20 above the mouth of the crypt and after moving to the villi; however, CK18-positive M cells in the crypt continued their expression of CK18 during movement to the FAE region. Terminal deoxynucleotidyl-transferase-mediated deoxyuridine-triphosphate-biotin nick-end labeling-positive apoptotic cells were specifically detected at the apical region of villi and FAE in the jejunum and ileum, and all were also stained for CK20. These data indicate that CK18 may be a molecular marker for bovine M cells in FAE and that M cells may transdifferentiate to CK20-positive enterocytes and die by apoptosis in the apex of the FAE.


Subject(s)
Enterocytes/metabolism , Ileum/metabolism , Intestinal Mucosa/metabolism , Jejunum/metabolism , Keratin-18/metabolism , Peyer's Patches/metabolism , Age Factors , Animals , Apoptosis , Biomarkers/metabolism , Cattle , Cell Proliferation , Cell Transdifferentiation , Colon/metabolism , Duodenum/metabolism , Enterocytes/ultrastructure , Ileum/cytology , Immunohistochemistry , In Situ Nick-End Labeling , Intestinal Mucosa/cytology , Jejunum/cytology , Keratin-20/metabolism , Male , Microscopy, Electron, Scanning , Microvilli/metabolism , Peyer's Patches/cytology
12.
J Virol ; 84(23): 12285-91, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861256

ABSTRACT

Transmissible spongiform encephalopathies (TSE), including bovine spongiform encephalopathy (BSE), are fatal neurodegenerative disorders in humans and animals. BSE appears to have spread to cattle through the consumption of feed contaminated with BSE/scrapie agents. In the case of an oral infection, the agents have to cross the gut-epithelial barrier. We recently established a bovine intestinal epithelial cell line (BIE cells) that can differentiate into the M cell type in vitro after lymphocytic stimulation (K. Miyazawa, T. Hondo, T. Kanaya, S. Tanaka, I. Takakura, W. Itani, M. T. Rose, H. Kitazawa, T. Yamaguchi, and H. Aso, Histochem. Cell Biol. 133:125-134, 2010). In this study, we evaluated the role of M cells in the intestinal invasion of the murine-adapted BSE (mBSE) agent using our in vitro bovine intestinal epithelial model. We demonstrate here that M cell-differentiated BIE cells are able to transport the mBSE agent without inactivation at least 30-fold more efficiently than undifferentiated BIE cells in our in vitro model. As M cells in the follicle-associated epithelium are known to have a high ability to transport a variety of macromolecules, viruses, and bacteria from gut lumen to mucosal immune cells, our results indicate the possibility that bovine M cells are able to deliver agents of TSE, not just the mBSE agent.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Encephalopathy, Bovine Spongiform/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/virology , Prions/immunology , Transcytosis/immunology , Animals , Cattle , Cell Line , Immunoblotting , Interleukin-2 , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Microspheres
13.
JCI Insight ; 6(23)2021 10 28.
Article in English | MEDLINE | ID: mdl-34710062

ABSTRACT

Bacterial cancer therapy (BCT) shows great promise for treatment of solid tumors, yet basic mechanisms of bacterial-induced tumor suppression remain undefined. Attenuated strains of Salmonella enterica serovar Typhimurium (STm) have commonly been used in mouse models of BCT in xenograft and orthotopic transplant cancer models. We aimed to better understand the tumor epithelium-targeted mechanisms of BCT by using autochthonous mouse models of intestinal cancer and tumor organoid cultures to assess the effectiveness and consequences of oral treatment with aromatase A-deficient STm (STmΔaroA). STmΔaroA delivered by oral gavage significantly reduced tumor burden and tumor load in both a colitis-associated colorectal cancer (CAC) model and in a spontaneous Apcmin/+ intestinal cancer model. STmΔaroA colonization of tumors caused alterations in transcription of mRNAs associated with tumor stemness, epithelial-mesenchymal transition, and cell cycle. Metabolomic analysis of tumors demonstrated alteration in the metabolic environment of STmΔaroA-treated tumors, suggesting that STmΔaroA imposes metabolic competition on the tumor. Use of tumor organoid cultures in vitro recapitulated effects seen on tumor stemness, mesenchymal markers, and altered metabolome. Furthermore, live STmΔaroA was required, demonstrating active mechanisms including metabolite usage. We have demonstrated that oral BCT is efficacious in autochthonous intestinal cancer models, that BCT imposes metabolic competition, and that BCT has direct effects on the tumor epithelium affecting tumor stem cells.


Subject(s)
Biological Therapy , Colorectal Neoplasms/therapy , Salmonella typhimurium/physiology , Administration, Oral , Animals , Aromatase/metabolism , Disease Models, Animal , Epithelium , Mice , Organoids , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics
14.
Histochem Cell Biol ; 133(1): 125-34, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19830445

ABSTRACT

Membranous epithelial cells (M cells) of the follicle-associated epithelium in Peyer's patches have a high capacity for transcytosis of several viruses and microorganisms. Here, we report that we have successfully established a bovine intestinal epithelial cell line (BIE cells) and developed an in vitro M cell model. BIE cells have a cobblestone morphology and microvilli-like structures, and strongly express cell-to-cell junctional proteins and cytokeratin, which is a specific intermediate filament protein of epithelial cells. After co-culture with murine intestinal lymphocytes or treatment with supernatant from bovine PBMC cultured with IL-2, BIE cells acquired the ability of transcytosis. Therefore, BIE cells have typical characteristics of bovine intestinal epithelial cells and also have the ability to differentiate into an M cell like linage. In addition, our results indicate that contact between immune cells and epithelial cells may not be absolutely required for the differentiation of M cells. We think that BIE cells will be useful for studying the transport mechanisms of various pathogens and also the evaluation of drug delivery via M cells.


Subject(s)
Cell Line , Epithelial Cells/cytology , Intestine, Small/cytology , Animals , Cattle , Cell Differentiation , Cell Proliferation , Epithelial Cells/metabolism , Mice
15.
Nat Microbiol ; 5(1): 181-191, 2020 01.
Article in English | MEDLINE | ID: mdl-31686029

ABSTRACT

Intestinal epithelial cells (IECs) act as a physical barrier separating the commensal-containing intestinal tract from the sterile interior. These cells have found a complex balance allowing them to be prepared for pathogen attacks while still tolerating the presence of bacterial or viral stimuli present in the lumen of the gut. Using primary human IECs, we probed the mechanisms that allow for such a tolerance. We discovered that viral infections emanating from the basolateral side of IECs elicit a stronger intrinsic immune response in comparison to lumenal apical infections. We determined that this asymmetric immune response is driven by the clathrin-sorting adaptor AP-1B, which mediates the polarized sorting of Toll-like receptor 3 (TLR3) towards the basolateral side of IECs. Mice and human IECs lacking AP-1B showed an exacerbated immune response following apical stimulation. Together, these results suggest a model where the cellular polarity program plays an integral role in the ability of IECs to partially tolerate apical commensals while remaining fully responsive to invasive basolateral pathogens.


Subject(s)
Cell Polarity/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Toll-Like Receptor 3/metabolism , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 1/metabolism , Animals , Cells, Cultured , Gene Knockdown Techniques , Humans , Interferons/metabolism , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/virology , Mice , Toll-Like Receptor 3/agonists , Viruses/immunology
16.
Front Immunol ; 10: 2432, 2019.
Article in English | MEDLINE | ID: mdl-31681315

ABSTRACT

The primary induction sites for intestinal IgA are the gut-associated lymphoid tissues (GALT), such as Peyer's patches (PPs) and isolated lymphoid follicles (ILFs). The commensal microbiota is known to contribute to IgA production in the gut; however, the role of dietary antigens in IgA production is poorly understood. To understand the effect of dietary antigens on IgA production, post-weaning mice were maintained on an elemental diet without any large immunogenic molecules. We found that dietary antigens contribute to IgA production in PPs through induction of follicular helper T cells and germinal center B cells. The role of dietary antigens in the PP responses was further confirmed by adding bovine serum albumin (BSA) into the elemental diet. Although dietary antigens are important for PP responses, they have fewer effects than the microbiota on the development and maturation of ILFs. Furthermore, we demonstrated that dietary antigens are essential for a normal antigen-specific IgA response to Salmonella typhi serovar Typhimurium infection. These results provide new insights into the role of dietary antigens in the regulation of mucosal immune responses.


Subject(s)
Antigens , Diet , Germinal Center/immunology , Peyer's Patches , Animals , Disease Susceptibility , Gastrointestinal Microbiome , Germinal Center/metabolism , Immunoglobulin A/immunology , Immunoglobulin A, Secretory/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Salmonella/immunology , Salmonella Infections/immunology , Salmonella Infections/microbiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
17.
J Exp Med ; 216(4): 831-846, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30877171

ABSTRACT

Microfold (M) cells residing in the follicle-associated epithelium (FAE) of the gut-associated lymphoid tissue are specialized for antigen uptake to initiate mucosal immune responses. The molecular machinery and biological significance of M cell differentiation, however, remain to be fully elucidated. Here, we demonstrate that Sox8, a member of the SRY-related HMG box transcription factor family, is specifically expressed by M cells in the intestinal epithelium. The expression of Sox8 requires activation of RANKL-RelB signaling. Chromatin immunoprecipitation and luciferase assays revealed that Sox8 directly binds the promoter region of Gp2 to increase Gp2 expression, which is the hallmark of functionally mature M cells. Furthermore, genetic deletion of Sox8 causes a marked decrease in the number of mature M cells, resulting in reduced antigen uptake in Peyer's patches. Consequently, juvenile Sox8-deficient mice showed attenuated germinal center reactions and antigen-specific IgA responses. These findings indicate that Sox8 plays an essential role in the development of M cells to establish mucosal immune responses.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/metabolism , Immunity, Mucosal/immunology , Immunoglobulin A/metabolism , Intestinal Mucosa/immunology , SOXE Transcription Factors/metabolism , Weaning , Animals , Antigens/immunology , HEK293 Cells , Humans , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peyer's Patches/cytology , Peyer's Patches/immunology , SOXE Transcription Factors/genetics
18.
Vet Immunol Immunopathol ; 124(3-4): 201-8, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18440649

ABSTRACT

Recent studies have shown that undifferentiated stem cells act as immunomodulators. To investigate the immunomodulatory function of the progenitor cells of the anterior pituitary gland, we attempted to establish a stem/progenitor cell line from the porcine anterior pituitary gland, and to detail its inflammatory cytokine expression. A cloned cell line from the porcine anterior pituitary gland was established and was designated as the porcine anterior pituitary-derived cell line (PAPC). PAPC expressed the mRNA of Nanog and Oct-4, and showed positive immunoreactivity for beta-catenin and Hes1 in its nucleus. PAPC grew stably by repeated passage and rapidly in the EGF and bFGF containing medium. RT-PCR showed that PAPC expressed mRNA of IL-1alpha, IL-6, IL-12, IL-15, IL-18 and TLR4. PAPC expressed S100alpha and IL-18 protein, which was localized in the marginal epithelial cells of Rathke's pouch. These results suggest that PAPC is a stem/progenitor cell and may regulate anterior pituitary cell function through an immuno-endocrine pathway.


Subject(s)
Cytokines/biosynthesis , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/immunology , Stem Cells/cytology , Stem Cells/immunology , Swine/metabolism , Animals , Cytokines/genetics , Female , Immunohistochemistry/veterinary , Inflammation/immunology , Inflammation/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary
19.
J Exp Med ; 215(2): 501-519, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29339448

ABSTRACT

M cells are located in the follicle-associated epithelium (FAE) that covers Peyer's patches (PPs) and are responsible for the uptake of intestinal antigens. The differentiation of M cells is initiated by receptor activator of NF-κB. However, the intracellular pathways involved in M cell differentiation are still elusive. In this study, we demonstrate that the NF-κB pathway activated by RANK is essential for M cell differentiation using in vitro organoid culture. Overexpression of NF-κB transcription factors enhances the expression of M cell-associated molecules but is not sufficient to complete M cell differentiation. Furthermore, we evaluated the requirement for tumor necrosis factor receptor-associated factor 6 (TRAF6). Conditional deletion of TRAF6 in the intestinal epithelium causes a complete loss of M cells in PPs, resulting in impaired antigen uptake into PPs. In addition, the expression of FAE-associated genes is almost silenced in TRAF6-deficient mice. This study thus demonstrates the crucial role of TRAF6-mediated NF-κB signaling in the development of M cells and FAE.


Subject(s)
Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , NF-kappa B/metabolism , TNF Receptor-Associated Factor 6/metabolism , Animals , Cell Differentiation , Cell Lineage , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Organoids/cytology , Organoids/immunology , Organoids/metabolism , Peyer's Patches/cytology , Peyer's Patches/immunology , Peyer's Patches/metabolism , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/deficiency , TNF Receptor-Associated Factor 6/genetics
20.
Res Vet Sci ; 82(2): 141-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16949627

ABSTRACT

M cells play a central role in the initiation of mucosal immune responses. However, a primary source of difficulty for investigations of this is the lack of an available specific marker for bovine M cells. As M cells possess irregular and short microvilli, we investigated the distribution and localization of the microvillar proteins actin and villin by immunohistochemistry of the gut of calves. In ileum of the calf, actin and villin were clearly and continuously immunostained in the brush border of the villous epithelia, however, discontinuous immunostaining with patches of no staining were observed in follicle-associated epithelium (FAE). Electron microscopy revealed that M cells had irregular microvilli and lacked the typical brush border, and it was inferred that these patches of no staining might be the intercellular crevices of M cells. As the microvilli of M cells were very sparse, there were several areas of weak immunostaining in calf jejunal FAE. These results suggest that M cells in calf FAE are detectable by the absence of staining for actin and villin.


Subject(s)
Actins/metabolism , Cattle/anatomy & histology , Ileum/cytology , Jejunum/cytology , Microfilament Proteins/metabolism , Animals , Ileum/metabolism , Ileum/ultrastructure , Immunohistochemistry/veterinary , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Jejunum/metabolism , Jejunum/ultrastructure , Microscopy, Electron, Scanning/veterinary , Microscopy, Electron, Transmission/veterinary , Microvilli/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL