Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nature ; 595(7868): 560-564, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34262176

RESUMEN

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.


Asunto(s)
Acetatos/farmacología , Bacterias/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Colon/inmunología , Dieta , Ácidos Grasos Volátiles/metabolismo , Homeostasis/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simbiosis
2.
Immunity ; 46(5): 863-874.e4, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28514691

RESUMEN

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.


Asunto(s)
Helmintiasis/inmunología , Helmintiasis/parasitología , Helmintos/inmunología , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Mastocitos/inmunología , Adenosina Trifosfato/metabolismo , Animales , Comunicación Celular , Diferenciación Celular , Modelos Animales de Enfermedad , Resistencia a la Enfermedad/genética , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Expresión Génica , Helmintiasis/genética , Inmunofenotipificación , Interleucina-33/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Mucosa Intestinal/patología , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/metabolismo , Masculino , Mastocitos/citología , Mastocitos/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Nat Immunol ; 13(8): 729-36, 2012 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-22706340

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Células Epiteliales/citología , Mucosa Intestinal/citología , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Linaje de la Célula , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Inmunidad Mucosa/genética , Mucosa Intestinal/embriología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ligando RANK/farmacología , Linfocitos T/inmunología
4.
Traffic ; 21(1): 34-44, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31647148

RESUMEN

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.


Asunto(s)
Microbiota , Ligando RANK , Diferenciación Celular , Humanos , Inmunidad Mucosa , Mucosa Intestinal
5.
Int Immunol ; 29(8): 357-363, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28992252

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Dendríticas/inmunología , Mucosa Intestinal/metabolismo , Lactobacillus acidophilus/fisiología , Uromodulina/metabolismo , Animales , Células Cultivadas , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Probióticos , Unión Proteica , Transcriptoma , Uromodulina/genética
7.
J Immunol ; 188(5): 2427-36, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22279105

RESUMEN

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.


Asunto(s)
Homeostasis/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Receptores Notch/fisiología , Transducción de Señal/inmunología , Animales , Traslocación Bacteriana/genética , Traslocación Bacteriana/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Enfermedad Crónica , Colitis/genética , Colitis/inmunología , Colitis/patología , Eliminación de Gen , Homeostasis/genética , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores Notch/genética , Transducción de Señal/genética , Células Th17/inmunología , Células Th17/metabolismo , Células Th17/patología
8.
Cell Metab ; 35(2): 361-375.e9, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36652945

RESUMEN

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.


Asunto(s)
Ácidos Grasos , Microbioma Gastrointestinal , Humanos , Animales , Ratones , Dieta Alta en Grasa , Obesidad/metabolismo , Bacterias/genética , Ratones Endogámicos C57BL
9.
Am J Pathol ; 179(3): 1301-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21763679

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Ganglios Linfáticos Agregados/metabolismo , Priones/farmacocinética , Administración Oral , Animales , Inmunohistoquímica , Intestino Delgado/metabolismo , Ganglios Linfáticos/metabolismo , Masculino , Mesenterio/metabolismo , Ratones , Ratones Noqueados , Ganglios Linfáticos Agregados/patología , Enfermedades por Prión/etiología , Priones/administración & dosificación , Bazo/metabolismo
10.
Sci Immunol ; 7(76): eabj8760, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36269840

RESUMEN

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.


Asunto(s)
Células T Asesinas Naturales , Ratones , Humanos , Animales , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/patología , Interleucina-15 , Antivirales , Granzimas , Receptores de Células Asesinas Naturales , Receptores de Quimiocina/metabolismo , Lípidos
11.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G442-53, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21193527

RESUMEN

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.


Asunto(s)
Enterocitos/metabolismo , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Yeyuno/metabolismo , Queratina-18/metabolismo , Ganglios Linfáticos Agregados/metabolismo , Factores de Edad , Animales , Apoptosis , Biomarcadores/metabolismo , Bovinos , Proliferación Celular , Transdiferenciación Celular , Colon/metabolismo , Duodeno/metabolismo , Enterocitos/ultraestructura , Íleon/citología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Mucosa Intestinal/citología , Yeyuno/citología , Queratina-20/metabolismo , Masculino , Microscopía Electrónica de Rastreo , Microvellosidades/metabolismo , Ganglios Linfáticos Agregados/citología
12.
J Virol ; 84(23): 12285-91, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20861256

RESUMEN

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.


Asunto(s)
Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Encefalopatía Espongiforme Bovina/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/virología , Priones/inmunología , Transcitosis/inmunología , Animales , Bovinos , Línea Celular , Immunoblotting , Interleucina-2 , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL , Microesferas
13.
JCI Insight ; 6(23)2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34710062

RESUMEN

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.


Asunto(s)
Terapia Biológica , Neoplasias Colorrectales/terapia , Salmonella typhimurium/fisiología , Administración Oral , Animales , Aromatasa/metabolismo , Modelos Animales de Enfermedad , Epitelio , Ratones , Organoides , Salmonella typhimurium/enzimología , Salmonella typhimurium/genética
14.
Histochem Cell Biol ; 133(1): 125-34, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19830445

RESUMEN

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.


Asunto(s)
Línea Celular , Células Epiteliales/citología , Intestino Delgado/citología , Animales , Bovinos , Diferenciación Celular , Proliferación Celular , Células Epiteliales/metabolismo , Ratones
15.
Nat Microbiol ; 5(1): 181-191, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31686029

RESUMEN

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.


Asunto(s)
Polaridad Celular/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Receptor Toll-Like 3/metabolismo , Complejo 1 de Proteína Adaptadora/genética , Complejo 1 de Proteína Adaptadora/metabolismo , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Interferones/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virología , Ratones , Receptor Toll-Like 3/agonistas , Virus/inmunología
16.
Front Immunol ; 10: 2432, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31681315

RESUMEN

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.


Asunto(s)
Antígenos , Dieta , Centro Germinal/inmunología , Ganglios Linfáticos Agregados , Animales , Susceptibilidad a Enfermedades , Microbioma Gastrointestinal , Centro Germinal/metabolismo , Inmunoglobulina A/inmunología , Inmunoglobulina A Secretora/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones , Salmonella/inmunología , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
17.
J Exp Med ; 216(4): 831-846, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30877171

RESUMEN

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.


Asunto(s)
Diferenciación Celular/inmunología , Células Epiteliales/metabolismo , Inmunidad Mucosa/inmunología , Inmunoglobulina A/metabolismo , Mucosa Intestinal/inmunología , Factores de Transcripción SOXE/metabolismo , Destete , Animales , Antígenos/inmunología , Células HEK293 , Humanos , Mucosa Intestinal/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/inmunología , Factores de Transcripción SOXE/genética
18.
Vet Immunol Immunopathol ; 124(3-4): 201-8, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18440649

RESUMEN

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.


Asunto(s)
Citocinas/biosíntesis , Adenohipófisis/citología , Adenohipófisis/inmunología , Células Madre/citología , Células Madre/inmunología , Porcinos/metabolismo , Animales , Citocinas/genética , Femenino , Inmunohistoquímica/veterinaria , Inflamación/inmunología , Inflamación/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria
19.
J Exp Med ; 215(2): 501-519, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29339448

RESUMEN

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.


Asunto(s)
Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , FN-kappa B/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides/citología , Organoides/inmunología , Organoides/metabolismo , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/deficiencia , Factor 6 Asociado a Receptor de TNF/genética
20.
Res Vet Sci ; 82(2): 141-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16949627

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Bovinos/anatomía & histología , Íleon/citología , Yeyuno/citología , Proteínas de Microfilamentos/metabolismo , Animales , Íleon/metabolismo , Íleon/ultraestructura , Inmunohistoquímica/veterinaria , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestructura , Yeyuno/metabolismo , Yeyuno/ultraestructura , Microscopía Electrónica de Rastreo/veterinaria , Microscopía Electrónica de Transmisión/veterinaria , Microvellosidades/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA