Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
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
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.
Sci Immunol ; 3(25)2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29980621

RESUMEN

Science Immunology's second anniversary invites a pause for editorial reflections.


Asunto(s)
Alergia e Inmunología , Publicaciones Periódicas como Asunto , Aniversarios y Eventos Especiales
6.
7.
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
8.
Int Immunol ; 29(10): 471-478, 2017 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-29186424

RESUMEN

Nasopharynx-associated lymphoid tissue (NALT) is one of the major constituents of the mucosa-associated lymphoid tissue (MALT), and has the ability to induce antigen-specific immune responses. However, the molecular mechanisms responsible for antigen uptake from the nasal cavity into the NALT remain largely unknown. Immunohistochemical analysis showed that CCL9 and CCL20 were co-localized with glycoprotein 2 (GP2) in the epithelium covering NALT, suggesting the existence of M cells in NALT. In analogy with the reduced number of Peyer's patch M cells in CCR6-deficient mice, the number of NALT M cells was drastically decreased in CCR6-deficient mice compared with the wild-type mice. Translocation of nasally administered Salmonella enterica serovar Typhimurium into NALT via NALT M cells was impaired in CCR6-deficient mice, whereas S. Typhimurium demonstrated consistent co-localization with NALT M cells in wild-type mice. When wild-type mice were nasally administered with an attenuated vaccine strain of S. Typhimurium, the mice were protected from a subsequent challenge with wild-type S. Typhimurium. Antigen-specific fecal and nasal IgA was detected after nasal immunization with the attenuated vaccine strain of S. Typhimurium only in wild-type mice but not in CCR6-deficient mice. Taken together, these observations demonstrate that NALT M cells are important as a first line of defense against infection by enabling activation of the common mucosal immune system (CMIS).


Asunto(s)
Células Epiteliales/inmunología , Inmunidad Mucosa/inmunología , Tejido Linfoide/inmunología , Nasofaringe/inmunología , Animales , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
9.
Immunology ; 152(4): 613-627, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28746740

RESUMEN

The intestinal lamina propria (LP) contains antigen-presenting cells with features of dendritic cells and macrophages, collectively referred to as mononuclear phagocytes (MNPs). Association of MNPs with the epithelium is thought to play an important role in multiple facets of intestinal immunity including imprinting MNPs with the ability to induce IgA production, inducing the expression of gut homing molecules on T cells, facilitating the capture of luminal antigens and microbes, and subsequent immune responses in the mesenteric lymph node (MLN). However, the factors promoting this process in the steady state are largely unknown, and in vivo models to test and confirm the importance of LP-MNP association with the epithelium for these outcomes are unexplored. Evaluation of epithelial expression of chemoattractants in mice where MNP-epithelial associations were impaired suggested CCL20 as a candidate promoting epithelial association. Expression of CCR6, the only known receptor for CCL20, was required for MNPs to associate with the epithelium. LP-MNPs from CCR6-/- mice did not display defects in acquiring antigen and stimulating T-cell responses in ex vivo assays or in responses to antigen administered systemically. However, LP-MNPs from CCR6-deficient mice were impaired at acquiring luminal and epithelial antigens, inducing IgA production in B cells, inducing immune responses in the MLN, and capturing and trafficking luminal commensal bacteria to the MLN. These findings identify a crucial role for CCR6 in promoting LP-MNPs to associate with the intestinal epithelium in the steady state to perform multiple functions promoting gut immune homeostasis.


Asunto(s)
Células Dendríticas/inmunología , Impresión Genómica/inmunología , Vigilancia Inmunológica , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Receptores CCR6/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Quimiocina CCL20/genética , Quimiocina CCL20/inmunología , Células Dendríticas/citología , Humanos , Macrófagos/citología , Ratones , Ratones Noqueados , Receptores CCR6/genética , Linfocitos T/citología , Linfocitos T/inmunología
10.
J Exp Med ; 214(6): 1607-1618, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28512157

RESUMEN

Interleukin-22 (IL-22) acts protectively and harmfully on intestinal tissue depending on the situation; therefore, IL-22 signaling needs to be tightly regulated. IL-22 binding protein (IL-22BP) binds IL-22 to inhibit IL-22 signaling. It is expressed in intestinal and lymphoid tissues, although its precise distribution and roles have remained unclear. In this study, we show that IL-22BP is highly expressed by CD11b+CD8α- dendritic cells in the subepithelial dome region of Peyer's patches (PPs). We found that IL-22BP blocks IL-22 signaling in the follicle-associated epithelium (FAE) covering PPs, indicating that IL-22BP plays a role in regulating the characteristics of the FAE. As expected, FAE of IL-22BP-deficient (Il22ra2-/-) mice exhibited altered properties such as the enhanced expression of mucus and antimicrobial proteins as well as prominent fucosylation, which are normally suppressed in FAE. Additionally, Il22ra2-/- mice exhibited the decreased uptake of bacterial antigens into PPs without affecting M cell function. Our present study thus demonstrates that IL-22BP promotes bacterial uptake into PPs by influencing FAE gene expression and function.


Asunto(s)
Antígenos Bacterianos/inmunología , Epitelio/inmunología , Ganglios Linfáticos Agregados/inmunología , Receptores de Interleucina/metabolismo , Animales , Diferenciación Celular , Recuento de Colonia Microbiana , Células Dendríticas/inmunología , Endocitosis , Células Epiteliales/inmunología , Interleucinas/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal , Interleucina-22
11.
Proc Natl Acad Sci U S A ; 114(8): 1976-1981, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28159893

RESUMEN

A significant challenge to HIV eradication is the elimination of viral reservoirs in germinal center (GC) T follicular helper (Tfh) cells. However, GCs are considered to be immune privileged for antiviral CD8 T cells. Here, we show a population of simian immunodeficiency virus (SIV)-specific CD8 T cells express CXCR5 (C-X-C chemokine receptor type 5, a chemokine receptor required for homing to GCs) and expand in lymph nodes (LNs) following pathogenic SIV infection in a cohort of vaccinated macaques. This expansion was greater in animals that exhibited superior control of SIV. The CXCR5+ SIV-specific CD8 T cells demonstrated enhanced polyfunctionality, restricted expansion of antigen-pulsed Tfh cells in vitro, and possessed a unique gene expression pattern related to Tfh and Th2 cells. The increase in CXCR5+ CD8 T cells was associated with the presence of higher frequencies of SIV-specific CD8 T cells in the GC. Following TCR-driven stimulation in vitro, CXCR5+ but not CXCR5- CD8 T cells generated both CXCR5+ as well as CXCR5- cells. However, the addition of TGF-ß to CXCR5- CD8 T cells induced a population of CXCR5+ CD8 T cells, suggesting that this cytokine may be important in modulating these CXCR5+ CD8 T cells in vivo. Thus, CXCR5+ CD8 T cells represent a unique subset of antiviral CD8 T cells that expand in LNs during chronic SIV infection and may play a significant role in the control of pathogenic SIV infection.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Centro Germinal/citología , Receptores CXCR5/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Enfermedad Crónica , Macaca mulatta , Masculino
12.
PLoS Pathog ; 12(12): e1006075, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27973593

RESUMEN

Many natural prion diseases of humans and animals are considered to be acquired through oral consumption of contaminated food or pasture. Determining the route by which prions establish host infection will identify the important factors that influence oral prion disease susceptibility and to which intervention strategies can be developed. After exposure, the early accumulation and replication of prions within small intestinal Peyer's patches is essential for the efficient spread of disease to the brain. To replicate within Peyer's patches, the prions must first cross the gut epithelium. M cells are specialised epithelial cells within the epithelia covering Peyer's patches that transcytose particulate antigens and microorganisms. M cell-development is dependent upon RANKL-RANK-signalling, and mice in which RANK is deleted only in the gut epithelium completely lack M cells. In the specific absence of M cells in these mice, the accumulation of prions within Peyer's patches and the spread of disease to the brain was blocked, demonstrating a critical role for M cells in the initial transfer of prions across the gut epithelium in order to establish host infection. Since pathogens, inflammatory stimuli and aging can modify M cell-density in the gut, these factors may also influence oral prion disease susceptibility. Mice were therefore treated with RANKL to enhance M cell density in the gut. We show that prion uptake from the gut lumen was enhanced in RANKL-treated mice, resulting in shortened survival times and increased disease susceptibility, equivalent to a 10-fold higher infectious titre of prions. Together these data demonstrate that M cells are the critical gatekeepers of oral prion infection, whose density in the gut epithelium directly limits or enhances disease susceptibility. Our data suggest that factors which alter M cell-density in the gut epithelium may be important risk factors which influence host susceptibility to orally acquired prion diseases.


Asunto(s)
Susceptibilidad a Enfermedades , Células Epiteliales , Mucosa Intestinal , Enfermedades por Prión/metabolismo , Animales , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcitosis/fisiología
13.
Am J Physiol Cell Physiol ; 311(3): C498-507, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27413168

RESUMEN

Microfold (M) cells are phagocytic intestinal epithelial cells in the follicle-associated epithelium of Peyer's patches that transport particulate antigens from the gut lumen into the subepithelial dome. Differentiation of M cells from epithelial stem cells in intestinal crypts requires the cytokine receptor activator of NF-κB ligand (RANKL) and the transcription factor Spi-B. We used three-dimensional enteroid cultures established with small intestinal crypts from mice as a model system to investigate signaling pathways involved in M cell differentiation and the influence of other cytokines on RANKL-induced M cell differentiation. Addition of RANKL to enteroids induced expression of multiple M cell-associated genes, including Spib, Ccl9 [chemokine (C-C motif) ligand 9], Tnfaip2 (TNF-α-induced protein 2), Anxa5 (annexin A5), and Marcksl1 (myristoylated alanine-rich protein kinase C substrate) in 1 day. The mature M cell marker glycoprotein 2 (Gp2) was strongly induced by 3 days and expressed by 11% of cells in enteroids. The noncanonical NF-κB pathway was required for RANKL-induced M cell differentiation in enteroids, as addition of RANKL to enteroids from mice with a null mutation in the mitogen-activated protein kinase kinase kinase 14 (Map3k14) gene encoding NF-κB-inducing kinase failed to induce M cell-associated genes. While the cytokine TNF-α alone had little, if any, effect on expression of M cell-associated genes, addition of TNF-α to RANKL consistently resulted in three- to sixfold higher levels of multiple M cell-associated genes than RANKL alone. One contributing mechanism is the rapid induction by TNF-α of Relb and Nfkb2 (NF-κB subunit 2), genes encoding the two subunits of the noncanonical NF-κB heterodimer. We conclude that endogenous activators of canonical NF-κB signaling present in the gut-associated lymphoid tissue microenvironment, including TNF-α, can play a supportive role in the RANKL-dependent differentiation of M cells in the follicle-associated epithelium.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiología , Intestinos/fisiología , Ligando RANK/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Femenino , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Células Madre/fisiología
14.
PLoS One ; 11(4): e0153402, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27100824

RESUMEN

M-cells (microfold cells) are thought to be a primary conduit of intestinal antigen trafficking. Using an established neutralizing anti-RANKL (Receptor Activator of NF-κB Ligand) antibody treatment to transiently deplete M-cells in vivo, we sought to determine whether intestinal M-cells were required for the effective induction of protective immunity following oral vaccination with ΔiglB (a defined live attenuated Francisella novicida mutant). M-cell depleted, ΔiglB-vaccinated mice exhibited increased (but not significant) morbidity and mortality following a subsequent homotypic or heterotypic pulmonary F. tularensis challenge. No significant differences in splenic IFN-γ, IL-2, or IL-17 or serum antibody (IgG1, IgG2a, IgA) production were observed compared to non-depleted, ΔiglB-vaccinated animals suggesting complementary mechanisms for ΔiglB entry. Thus, we examined other possible routes of gastrointestinal antigen sampling following oral vaccination and found that ΔiglB co-localized to villus goblet cells and enterocytes. These results provide insight into the role of M-cells and complementary pathways in intestinal antigen trafficking that may be involved in the generation of optimal immunity following oral vaccination.


Asunto(s)
Vacunas Bacterianas/inmunología , Francisella tularensis/inmunología , Intestinos/citología , Intestinos/inmunología , Tularemia/inmunología , Tularemia/prevención & control , Vacunas Atenuadas/inmunología , Animales , Femenino , Inmunidad , Interferón gamma/inmunología , Interleucina-17/inmunología , Interleucina-2/inmunología , Intestinos/microbiología , Ratones , Ratones Endogámicos BALB C , Bazo/inmunología , Bazo/microbiología
15.
Immunol Lett ; 171: 5-14, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26773232

RESUMEN

Antibodies play an important role in therapy and investigative biomedical research. The TNF-family member Receptor Activator of NF-κB (RANK) is known for its role in bone homeostasis and is increasingly recognized as a central player in immune regulation and epithelial cell activation. However, the study of RANK biology has been hampered by missing or insufficient characterization of high affinity tools that recognize RANK. Here, we present a careful description and comparison of two antibodies, RANK-02 obtained by phage display (Newa, 2014 [1]) and R12-31 generated by immunization (Kamijo, 2006 [2]). We found that both antibodies recognized mouse RANK with high affinity, while RANK-02 and R12-31 recognized human RANK with high and lower affinities, respectively. Using a cell apoptosis assay based on stimulation of a RANK:Fas fusion protein, and a cellular NF-κB signaling assay, we showed that R12-31 was agonist for both species. R12-31 interfered little or not at all with the binding of RANKL to RANK, in contrast to RANK-02 that efficiently prevented this interaction. Depending on the assay and species, RANK-02 was either a weak agonist or a partial antagonist of RANK. Both antibodies recognized human Langerhans cells, previously shown to express RANK, while dermal dendritic cells were poorly labeled. In vivo R12-31 agonist activity was demonstrated by its ability to induce the formation of intestinal villous microfold cells in mice. This characterization of two monoclonal antibodies should now allow better evaluation of their application as therapeutic reagents and investigative tools.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Células Epiteliales/fisiología , Epítopos/metabolismo , Intestinos/efectos de los fármacos , Células de Langerhans/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Afinidad de Anticuerpos , Diferenciación Celular/efectos de los fármacos , Técnicas de Visualización de Superficie Celular , Células Epiteliales/efectos de los fármacos , Epítopos/inmunología , Células HEK293 , Humanos , Inmunización Secundaria , Inmunomodulación , Intestinos/citología , Células Jurkat , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptor Activador del Factor Nuclear kappa-B/inmunología , Transducción de Señal
16.
Nat Commun ; 6: 8282, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26392000

RESUMEN

HIV infection causes bone loss. We previously reported that immunosuppression-mediated B-cell production of receptor activator of NF-κB ligand (RANKL) coupled with decline in osteoprotegerin correlate with decreased bone mineral density (BMD) in untreated HIV infection. Paradoxically, antiretroviral therapy (ART) worsens bone loss although existing data suggest that such loss is largely independent of specific antiretroviral regimen. This led us to hypothesize that skeletal deterioration following HIV disease reversal with ART may be related to T-cell repopulation and/or immune reconstitution. Here we transplant T cells into immunocompromised mice to mimic ART-induced T-cell expansion. T-cell reconstitution elicits RANKL and TNFα production by B cells and/or T cells, accompanied by enhanced bone resorption and BMD loss. Reconstitution of TNFα- or RANKL-null T-cells and pharmacological TNFα antagonist all protect cortical, but not trabecular bone, revealing complex effects of T-cell reconstitution on bone turnover. These findings suggest T-cell repopulation and/or immune reconstitution as putative mechanisms for bone loss following ART initiation.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , VIH-1 , Osteoporosis/inducido químicamente , Linfocitos T/fisiología , Traslado Adoptivo , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/inmunología , Resorción Ósea , Recuento de Linfocito CD4 , Femenino , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
Nat Nanotechnol ; 10(4): 361-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25751305

RESUMEN

In humans and other mammals it is known that calcium and phosphate ions are secreted from the distal small intestine into the lumen. However, why this secretion occurs is unclear. Here, we show that the process leads to the formation of amorphous magnesium-substituted calcium phosphate nanoparticles that trap soluble macromolecules, such as bacterial peptidoglycan and orally fed protein antigens, in the lumen and transport them to immune cells of the intestinal tissue. The macromolecule-containing nanoparticles utilize epithelial M cells to enter Peyer's patches, small areas of the intestine concentrated with particle-scavenging immune cells. In wild-type mice, intestinal immune cells containing these naturally formed nanoparticles expressed the immune tolerance-associated molecule 'programmed death-ligand 1', whereas in NOD1/2 double knockout mice, which cannot recognize peptidoglycan, programmed death-ligand 1 was undetected. Our results explain a role for constitutively formed calcium phosphate nanoparticles in the gut lumen and show how this helps to shape intestinal immune homeostasis.


Asunto(s)
Antígenos/inmunología , Intestinos/citología , Intestinos/inmunología , Peptidoglicano/inmunología , Ganglios Linfáticos Agregados/inmunología , Fosfatos/inmunología , Animales , Calcio/inmunología , Fosfatos de Calcio/inmunología , Células Cultivadas , Humanos , Intestinos/química , Ratones , Ratones Endogámicos BALB C , Minerales/inmunología , Chaperonas Moleculares/inmunología , Nanopartículas/química , Nanopartículas/ultraestructura , Tamaño de la Partícula , Ganglios Linfáticos Agregados/citología
18.
Science ; 346(6211): 861-5, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25395539

RESUMEN

Activators of innate immunity may have the potential to combat a broad range of infectious agents. We report that treatment with bacterial flagellin prevented rotavirus (RV) infection in mice and cured chronically RV-infected mice. Protection was independent of adaptive immunity and interferon (IFN, type I and II) and required flagellin receptors Toll-like receptor 5 (TLR5) and NOD-like receptor C4 (NLRC4). Flagellin-induced activation of TLR5 on dendritic cells elicited production of the cytokine interleukin-22 (IL-22), which induced a protective gene expression program in intestinal epithelial cells. Flagellin also induced NLRC4-dependent production of IL-18 and immediate elimination of RV-infected cells. Administration of IL-22 and IL-18 to mice fully recapitulated the capacity of flagellin to prevent or eliminate RV infection and thus holds promise as a broad-spectrum antiviral agent.


Asunto(s)
Flagelina/administración & dosificación , Inmunidad Innata , Interleucina-18/inmunología , Interleucinas/inmunología , Infecciones por Rotavirus/prevención & control , Receptor Toll-Like 5/fisiología , Animales , Diarrea/inmunología , Diarrea/terapia , Diarrea/virología , Modelos Animales de Enfermedad , Heces/virología , Flagelina/inmunología , Proteínas de Homeodominio/genética , Interleucina-18/administración & dosificación , Interleucina-18/genética , Interleucinas/administración & dosificación , Interleucinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/terapia , Receptor Toll-Like 5/genética , Esparcimiento de Virus , Interleucina-22
19.
J Virol ; 88(12): 6934-43, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24696493

RESUMEN

UNLABELLED: Microfold (M) cells are specialized intestinal epithelial cells that internalize particulate antigens and aid in the establishment of immune responses to enteric pathogens. M cells have also been suggested as a portal for pathogen entry into the host. While virus particles have been observed in M cells, it is not known whether viruses use M cells to initiate a productive infection. Noroviruses (NoVs) are single-stranded RNA viruses that infect host organisms via the fecal-oral route. Murine NoV (MNV) infects intestinal macrophages and dendritic cells and provides a tractable experimental system for understanding how an enteric virus overcomes the intestinal epithelial barrier to infect underlying target cells. We found that replication of two divergent MNV strains was reduced in mice depleted of M cells. Reoviruses are double-stranded RNA viruses that infect hosts via respiratory or enteric routes. In contrast to MNV, reovirus infects enterocytes in the intestine. Despite differences in cell tropism, reovirus infection was also reduced in M cell-depleted mice. These data demonstrate that M cells are required for the pathogenesis of two unrelated enteric viruses that replicate in different cell types within the intestine. IMPORTANCE: To successfully infect their hosts, pathogens that infect via the gastrointestinal tract must overcome the multilayered system of host defenses. Microfold (M) cells are specialized intestinal epithelial cells that internalize particulate antigens and aid in the establishment of immune responses to enteric pathogens. Virus particles have been observed within M cells. However, it is not known whether viruses use M cells to initiate a productive infection. To address this question, we use MNV and reovirus, two enteric viruses that replicate in different cell types in the intestine, intestinal epithelial cells for reovirus and intestinal mononuclear phagocytes for MNV. Interestingly, MNV- and reovirus-infected mice depleted of M cells showed reduced viral loads in the intestine. Thus, our work demonstrates the importance of M cells in the pathogenesis of enteric viruses irrespective of the target cell type in which the virus replicates.


Asunto(s)
Infecciones por Caliciviridae/virología , Células Epiteliales/virología , Intestinos/virología , Norovirus/fisiología , Infecciones por Reoviridae/virología , Reoviridae/fisiología , Replicación Viral , Animales , Línea Celular , Humanos , Intestinos/citología , Ratones , Ratones Endogámicos BALB C
20.
Inflammation ; 37(3): 893-907, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24413902

RESUMEN

Murine hepatic Cyp4a mRNAs are markedly downregulated during inflammation. Here, we investigated the roles of Cyp4a10 and Cyp4a14 in the response to infection with C. rodentium. Absence of either Cyp4a gene attenuated or abrogated the changes in spleen weight, colon crypt length, hepatic cytokine, and acute phase protein mRNAs, and serum acute phase proteins and cytokines caused by infection. Cyp4a10(-/-) mice on a low-salt diet had a similar hepatic acute phase response as those mice on a high-salt diet, suggesting that hypertension associated with this genotype is not the cause of their altered inflammatory response. In contrast, wild-type, Cyp4a10(-/-), and Cyp4a14(-/-) mice showed similar responses to injected LPS. These results implicate Cyp4a10 and Cyp4a14 in the regulation of the host inflammatory response to enteropathogenic bacterial infection but not to acute aseptic inflammation. Understanding the mechanism of this role may lead to novel therapeutic approaches in some inflammatory diseases.


Asunto(s)
Citrobacter rodentium/inmunología , Sistema Enzimático del Citocromo P-450/genética , Infecciones por Enterobacteriaceae/inmunología , Inflamación/inmunología , Animales , Familia 4 del Citocromo P450 , Citocinas/biosíntesis , Citocinas/sangre , Femenino , Hipertensión/inmunología , Inflamación/microbiología , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , Sodio en la Dieta/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...