Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arthritis Rheumatol ; 70(12): 1971-1983, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29975009

RESUMO

OBJECTIVE: Intestinal microbiota are associated with the development of inflammatory arthritis. The aim of this study was to dissect intestinal mucosal immune responses in the preclinical phase of arthritis and determine whether the presence of Th17 cells, beyond involvement of the cytokine interleukin-17 (IL-17), is required for arthritis development, and whether the involvement of Th17 cells in arthritis depends on the composition of the host microbiota. METHODS: Mucosal T cell production of IL-17, interferon-γ, tumor necrosis factor α (TNFα), IL-22, and granulocyte-macrophage colony-stimulating factor (GM-CSF) was analyzed by flow cytometry and Luminex assay before arthritis onset in mice immunized to develop collagen-induced arthritis (CIA). Pathogenic features of arthritis in mice with CIA and mice with antigen-induced arthritis were compared between Th17 cell-deficient (CD4-Cre+ Rorcflox/flox ) and Th17 cell-sufficient (CD4-Cre- Rorcflox/flox ) mice. In addition, the impact of intestinal microbiota on the Th17 cell dependence of CIA was assessed. RESULTS: Lamina propria CD4 T cells were activated before the onset of arthritis in mice with CIA, with marked up-regulation of several cytokines, including IL-17A, TNFα, and GM-CSF. CD4-Cre+ Rorcflox/flox  mice showed a specific reduction in intestinal mucosal levels of Th17 cells and partially reduced levels of IL-17-producing CD8 T cells. However, total levels of IL-17A, mostly produced by γδ T cells and neutrophils, were unaffected. The severity of arthritis was significantly reduced in Th17 cell-deficient mice, suggesting that Th17 cells have additional, IL-17A-independent roles in inflammatory arthritis. Accordingly, antigen-stimulated T cells from Th17 cell-deficient mice produced less IL-17A, IL-17F, and GM-CSF. Importantly, the dependence of CIA on the involvement of Th17 cells was mitigated in the presence of an alternative microbiome. CONCLUSION: These data from murine models suggest that activation of mucosal immunity precedes the development of arthritis, and also that Th17 cells have a microbiota-dependent role in arthritis. Therefore, a microbiome-guided stratification of patients might improve the efficacy of Th17-targeted therapies.


Assuntos
Artrite Experimental/imunologia , Artrite Experimental/microbiologia , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/microbiologia , Células Th17/microbiologia , Animais , Citocinas/imunologia , Modelos Animais de Doenças , Imunidade nas Mucosas , Camundongos , Células Th17/imunologia
2.
Sci Rep ; 7(1): 15613, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29142301

RESUMO

Perturbations of the intestinal microbiome have been observed in patients with new-onset and chronic autoimmune inflammatory arthritis. However, it is currently unknown whether these alterations precede the development of arthritis or are rather a consequence of disease. Modulation of intestinal microbiota by oral antibiotics or germ-free condition can prevent arthritis in mice. Yet, the therapeutic potential of modulation of the microbiota after the onset of arthritis is not well characterized. We here show that the intestinal microbial community undergoes marked changes in the preclinical phase of collagen induced arthritis (CIA). The abundance of the phylum Bacteroidetes, specifically families S24-7 and Bacteroidaceae was reduced, whereas Firmicutes and Proteobacteria, such as Ruminococcaceae, Lachnospiraceae and Desulfovibrinocaceae, were expanded during the immune-priming phase of arthritis. In addition, we found that the abundance of lamina propria Th17, but not Th1, cells is highly correlated with the severity of arthritis. Elimination of the intestinal microbiota during established arthritis specifically reduced intestinal Th17 cells and attenuated arthritis. These effects were associated with reduced serum amyloid A expression in ileum and synovial tissue. Our observations suggest that intestinal microbiota perturbations precede arthritis, and that modulation of the intestinal microbiota after the onset of arthritis may offer therapeutic opportunities.


Assuntos
Artrite Experimental/microbiologia , Microbioma Gastrointestinal/genética , Inflamação/microbiologia , Células Th17/imunologia , Animais , Artrite Experimental/genética , Artrite Experimental/imunologia , Bacteroidetes/genética , Modelos Animais de Doenças , Firmicutes/genética , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Camundongos , Proteobactérias/genética , RNA Ribossômico 16S/genética , Proteína Amiloide A Sérica/metabolismo , Células Th1/imunologia , Células Th17/microbiologia
3.
J Immunol ; 196(10): 4390-9, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27067005

RESUMO

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.


Assuntos
Interleucina-17/biossíntese , Interleucina-23/metabolismo , Interleucinas/biossíntese , Neutrófilos/metabolismo , Receptores de Interleucina/metabolismo , Serina-Treonina Quinases TOR/genética , Animais , Ceco/patologia , Diferenciação Celular , Colite/imunologia , Colite/patologia , Colo/patologia , Interleucina-23/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores de Interleucina/genética , Transdução de Sinais , Células Th17/imunologia , Interleucina 22
4.
Sci Rep ; 6: 22040, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26907705

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Receptores Imunológicos/imunologia , Células Th17/imunologia , Receptor 5 Toll-Like/imunologia , Animais , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Flagelina/farmacologia , Regulação da Expressão Gênica/imunologia , Homeostase , Interleucina-23/genética , Interleucina-23/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Mucosa Intestinal/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cultura Primária de Células , Receptores de Antígenos de Linfócitos T alfa-beta/deficiência , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/deficiência , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Transdução de Sinais , Células Th17/citologia , Receptor 5 Toll-Like/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia
5.
Eur J Immunol ; 46(5): 1162-7, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26840377

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Bactérias Gram-Positivas/imunologia , Mucosa/imunologia , Simbiose , Células Th2/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Diferenciação Celular , Células Cultivadas , Bactérias Gram-Positivas/química , Interleucina-4/biossíntese , Interleucina-4/imunologia , Ativação Linfocitária , Camundongos , Mucosa/microbiologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/fisiologia , Fator de Crescimento Transformador beta/biossíntese
6.
Inflamm Allergy Drug Targets ; 14(1): 4-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26296210

RESUMO

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.


Assuntos
Doenças Inflamatórias Intestinais/imunologia , Integrinas/metabolismo , Intestinos/imunologia , Receptores CCR/metabolismo , Receptores de Retorno de Linfócitos/metabolismo , Animais , Anti-Inflamatórios/uso terapêutico , Diferenciação Celular , Movimento Celular , Ensaios Clínicos como Assunto , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Terapia de Alvo Molecular
7.
PLoS One ; 10(7): e0134100, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26230654

RESUMO

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.


Assuntos
Receptores CCR/fisiologia , Linfócitos T Reguladores/citologia , Animais , Colite/fisiopatologia , Camundongos , Camundongos Transgênicos , Receptores CCR/genética
8.
Gut ; 64(11): 1755-64, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25281418

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Imunidade Celular , Doenças Inflamatórias Intestinais/imunologia , MicroRNAs/fisiologia , Células Th1/imunologia , Células Th17/imunologia , Adulto , Animais , Feminino , Humanos , Imunidade Celular/genética , Doenças Inflamatórias Intestinais/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Adulto Jovem
9.
J Leukoc Biol ; 96(5): 895-905, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25015957

RESUMO

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.


Assuntos
Inflamação , Interferon gama/biossíntese , Interleucina-17/biossíntese , Mucosa Intestinal , Intestinos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Colite/genética , Colite/imunologia , Colite/metabolismo , Citocinas/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/patologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Receptor 4 Toll-Like/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Inflamm Allergy Drug Targets ; 13(1): 65-73, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24438006

RESUMO

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.


Assuntos
Imunidade nas Mucosas , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Intestinos/imunologia , Microbiota , Animais , Evolução Biológica , Interação Gene-Ambiente , Interações Hospedeiro-Patógeno , Humanos , Imunomodulação , Intestinos/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...