Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunol ; 196(10): 4390-9, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27067005

RESUMEN

It has been shown recently that neutrophils are able to produce IL-22 and IL-17, which differentially regulate the pathogenesis of inflammatory bowel disease. However, it is still largely unknown how the neutrophil production of IL-22 and IL-17 is regulated, and their role in the pathogenesis of inflammatory bowel disease. In this study, we found that IL-23 promoted neutrophil production of IL-17 and IL-22. IL-23 stimulated the neutrophil expression of IL-23R as well as rorc and ahr. Retinoid acid receptor-related orphan receptor γ t and aryl-hydrocarbon receptor differentially regulated IL-23 induction of neutrophil IL-17 and IL-22. In addition, IL-23 induced the activation of mTOR in neutrophils. Blockade of the mTOR pathway inhibited IL-23-induced expression of rorc and ahr, as well as IL-17 and IL-22 production. By using a microbiota Ag-specific T cell-mediated colitis model, we demonstrated that depletion of neutrophils, as well as blockade of IL-22, resulted in a significant increase in the severity of colitis, thereby indicating a protective role of neutrophils and IL-22 in chronic colitis. Collectively, our data revealed that neutrophils negatively regulate microbiota Ag-specific T cell induction of colitis, and IL-23 induces neutrophil production of IL-22 and IL-17 through induction of rorc and ahr, which is mediated by the mTOR pathway.


Asunto(s)
Interleucina-17/biosíntesis , Interleucina-23/metabolismo , Interleucinas/biosíntesis , Neutrófilos/metabolismo , Receptores de Interleucina/metabolismo , Serina-Treonina Quinasas TOR/genética , Animales , Ciego/patología , Diferenciación Celular , Colitis/inmunología , Colitis/patología , Colon/patología , Interleucina-23/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Interleucina/genética , Transducción de Señal , Células Th17/inmunología , Interleucina-22
2.
Sci Rep ; 6: 22040, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26907705

RESUMEN

Multiple mechanisms exist in regulation of host responses to massive challenges from microbiota to maintain immune homeostasis in the intestines. Among these is the enriched Th17 cells in the intestines, which regulates intestinal homeostasis through induction of antimicrobial peptides and secretory IgA among others. However, the means by which Th17 cells develop in response to microbiota is still not completely understood. Although both TLR5 and CD172α(+) lamina propria dendritic cells (LPDC) have been shown to promote Th17 cell development, it is still unclear whether TLR5 mediates the CD172α(+)LPDC induction of Th17 cells. By using a microbiota antigen-specific T cell reporter mouse system, we demonstrated that microbiota antigen-specific T cells developed into Th17 cells in the intestinal LP, but not in the spleen when transferred into TCRßxδ(-/-) mice. LPDCs expressed high levels of TLR5, and most CD172α(+)LPDCs also co-expressed TLR5. LPDCs produced high levels of IL-23, IL-6 and TGFß when stimulated with commensal flagellin and promoted Th17 cell development when cultured with full-length CBir1 flagellin but not CBir1 peptide. Wild-type CD172α(+), but not CD172α(-), LPDCs induced Th17 cells, whereas TLR5-deficient LPDC did not induce Th17 cells. Our data thereby demonstrated that TLR5 mediates CD172α(+)LPDC induction of Th17 cells in the intestines.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Receptores Inmunológicos/inmunología , Células Th17/inmunología , Receptor Toll-Like 5/inmunología , Animales , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Flagelina/farmacología , Regulación de la Expresión Génica/inmunología , Homeostasis , Interleucina-23/genética , Interleucina-23/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Mucosa Intestinal/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T alfa-beta/deficiencia , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Transducción de Señal , Células Th17/citología , Receptor Toll-Like 5/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
3.
Eur J Immunol ; 46(5): 1162-7, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26840377

RESUMEN

It has been shown that while commensal bacteria promote Th1, Th17 and Treg cells in lamina propria (LP) in steady-state conditions, they suppress mucosal Th2 cells. However, it is still unclear whether there are specific commensal organisms down-regulating Th2 responses, and the mechanism involved. Here we demonstrate that commensal A4 bacteria, a member of the Lachnospiraceae family, which produce an immunodominant microbiota CBir1 antigen, inhibits LP Th2-cell development. When transferred into the intestines of RAG(-/-) mice, CBir1-specific T cells developed predominately towards Th1 cells and Th17 cells, but to a lesser extent into Th2 cells. The addition of A4 bacterial lysates to CD4(+) T-cell cultures inhibited production of IL-4. A4 bacteria stimulated dendritic cell production of TGF-ß, and blockade of TGF-ß abrogated A4 bacteria inhibition of Th2-cell development in vitro and in vivo. Collectively, our data show that A4 bacteria inhibit Th2-cell differentiation by inducing dendritic cell production of TGF-ß.


Asunto(s)
Células Dendríticas/inmunología , Bacterias Grampositivas/inmunología , Membrana Mucosa/inmunología , Simbiosis , Células Th2/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Bacterias Grampositivas/química , Interleucina-4/biosíntesis , Interleucina-4/inmunología , Activación de Linfocitos , Ratones , Membrana Mucosa/microbiología , Células TH1/inmunología , Células Th17/inmunología , Células Th2/fisiología , Factor de Crecimiento Transformador beta/biosíntesis
4.
Inflamm Allergy Drug Targets ; 14(1): 4-12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26296210

RESUMEN

Inflammatory bowel disease is a chronic, debilitating immunological disorder for which there are few effective treatments. New therapies targeting gut homing molecules, such as CCR9 and α4ß7, are currently in development, with some of these reaching clinical trials. Gut-trophic molecules and their receptors are critical to the development of both tolerant and inflammatory immune responses in the gut. However, we know little regarding the function of homing molecules as it relates to IBD. Data have suggested both pathological and protective roles for gut homing molecules in IBD development and maintenance. In addition, recent research findings have suggested that chemokines can influence T cell differentiation and function. Given the current clinical relevance, it is essential to obtain a better understanding of the role of gut homing molecules in the regulation of IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Integrinas/metabolismo , Intestinos/inmunología , Receptores CCR/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Diferenciación Celular , Movimiento Celular , Ensayos Clínicos como Asunto , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Terapia Molecular Dirigida
5.
PLoS One ; 10(7): e0134100, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26230654

RESUMEN

T cells reactive to microbiota regulate the pathogenesis of inflammatory bowel disease (IBD). As T cell trafficking to intestines is regulated through interactions between highly specific chemokine-chemokine receptors, efforts have been made to develop intestine-specific immunosuppression based on blocking these key processes. CCR9, a gut-trophic chemokine receptor expressed by lymphocytes and dendritic cells, has been implicated in the regulation of IBD through mediating recruitment of T cells to inflamed sites. However, the role of CCR9 in inducing and sustaining inflammation in the context of IBD is poorly understood. In this study, we demonstrate that CCR9 deficiency in effector T cells and Tregs does not affect the development of colitis in a microbiota antigen-specific, T cell-mediated model. However, Treg cells express higher levels of CCR9 compared to those in effector T cells. Interestingly, CCR9 inhibits Treg cell development, in that CCR9-/- mice demonstrate a high level of Foxp3+ Tregs, and ligation of CCR9 by its ligand CCL25 inhibits Treg cell differentiation in vitro. Collectively, our data indicate that in addition to acting as a gut-homing molecule, CCR9 signaling shapes immune responses by inhibiting Treg cell development.


Asunto(s)
Receptores CCR/fisiología , Linfocitos T Reguladores/citología , Animales , Colitis/fisiopatología , Ratones , Ratones Transgénicos , Receptores CCR/genética
6.
Gut ; 64(11): 1755-64, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25281418

RESUMEN

OBJECTIVE: Although both innate and adaptive responses to microbiota have been implicated in the pathogenesis of IBD, it is still largely unknown how they are regulated during intestinal inflammation. In this report, we investigated the role of microRNA (miR)-10a, a small, non-coding RNA, in the regulation of innate and adaptive responses to microbiota in IBD. METHODS: miR-10a expression was analysed in the inflamed mucosa of IBD patients treated with or without antitumour necrosis factor (anti-TNF) monoclonal antibodies (mAb) (infliximab) by qRT-PCR. Human monocyte-derived dendritic cells (DC) and IBD CD4+ T cells were transfected with miR-10a precursor to define their effect on the function of DC and CD4+ T cells. RESULTS: The expression of miR-10a was markedly decreased, while NOD2 and interleukin (IL)-12/IL-23p40 were significantly increased, in the inflamed mucosa of IBD patients compared with those in healthy controls. Commensal bacteria, TNF and interferon-γ inhibited human DC miR-10a expression in vitro. Anti-TNF mAb treatment significantly promoted miR-10a expression, whereas it markedly inhibited NOD2 and IL-12/IL-23p40 in the inflamed mucosa. We further identified NOD2, in addition to IL-12/IL-23p40, as a target of miR-10a. The ectopic expression of the miR-10a precursor inhibited IL-12/IL-23p40 and NOD2 in DC. Moreover, miR-10a was found to markedly suppress IBD T helper (Th)1 and Th17 cell responses. CONCLUSIONS: Our data indicate that miR-10a is decreased in the inflamed mucosa of IBD and downregulates mucosal inflammatory response through inhibition of IL-12/IL-23p40 and NOD2 expression, and blockade of Th1/Th17 cell immune responses. Thus, miR-10a could play a role in the pathogenesis and progression of IBD.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Celular , Enfermedades Inflamatorias del Intestino/inmunología , MicroARNs/fisiología , Células TH1/inmunología , Células Th17/inmunología , Adulto , Animales , Femenino , Humanos , Inmunidad Celular/genética , Enfermedades Inflamatorias del Intestino/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
7.
J Leukoc Biol ; 96(5): 895-905, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25015957

RESUMEN

Tregs play a crucial role in the maintenance of intestinal immune homeostasis. However, significant numbers of Foxp3(+) Tregs accumulate in the inflamed lesions in experimental colitis and in IBD patients. Treg production of the proinflammatory cytokines IFN-γ and/or IL-17 may arguably explain their ineffectiveness in suppressing intestinal inflammation. However, it remains unknown whether iTreg and tTreg produce proinflammatory cytokines and how TLR signaling regulates this process. Here, we found that Foxp3(+)Tregs were increased in the intestines of B6.TLR4(-/-) and B6.IL-10(-/-) mice when compared with WT B6 mice. TLR4(-/-) and IL-10(-/-) resulted in more Tregs within inflamed intestines. The majority of Foxp3(+) Tregs in the spleen was Helios(+)Nrp1(+), whereas most Foxp3(+) Tregs in the intestinal LP were Helios(-)Nrp1(-). More Helios(+)Nrp1(+) Tregs expressed IFN-γ and/or IL-17 than did Helios(-)Nrp1(-) Tregs in the spleen and intestine, which was increased with TLR4(-/-). TLR4 signaling in T cells and APCs inhibited Foxp3(+) induction via MyD88-dependent, TRIF-independent pathways, which was negatively regulated by SOCS3. Collectively, these data demonstrate Helios(+)Nrp1(+) tTregs and Helios(-)Nrp1(-) iTregs produce proinflammatory cytokines in the intestines during inflammation, which was regulated by TLR4 signaling.


Asunto(s)
Inflamación , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Mucosa Intestinal , Intestinos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Citocinas/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/patología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Receptor Toll-Like 4/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Inflamm Allergy Drug Targets ; 13(1): 65-73, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24438006

RESUMEN

The intestines harbor over trillions of commensal bacteria, which co-evolve and form a mutualistic relationship with the host, with microbial-host interaction shaping immune adaption and bacterial communities. The intestinal microbiota not only benefits the host and contributes to the maintenance of intestinal homeostasis, but also causes chronic intestinal inflammation under certain conditions. Thus, understanding the microbiota regulation of inflammatory bowel disease (IBD) will provide great insights into the pathogenesis of IBD as well as potential therapeutics for IBD patients.


Asunto(s)
Inmunidad Mucosa , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Intestinos/inmunología , Microbiota , Animales , Evolución Biológica , Interacción Gen-Ambiente , Interacciones Huésped-Patógeno , Humanos , Inmunomodulación , Intestinos/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...