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










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 10: 411, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30949163

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease that affects ~1% of the world's population. B cells and autoantibodies play an important role in the pathogenesis of RA. The P2RX7 receptor is an ATP-gated cation channel and its activation results in the release of pro-inflammatory molecules. Thus, antagonists of P2RX7 have been considered to have potential as novel anti-inflammatory therapies. Although originally identified for its role in innate immunity, P2RX7 has recently been found to negatively control Peyer's patches (PP) T follicular helper cells (Tfh), which specialize in helping B cells, under homeostatic conditions. We have previously demonstrated that PP Tfh cells are required for the augmentation of autoimmune arthritis mediated by gut commensal segmented filamentous bacteria (SFB). Thus, we hypothesized that P2RX7 is required to control autoimmune disease by keeping the Tfh cell response in check. To test our hypothesis, we analyzed the impact of P2RX7 deficiency in vivo using both the original K/BxN autoimmune arthritis model and T cell transfers in the K/BxN system. We also examined the impact of P2RX7 ablation on autoimmune development in the presence of the gut microbiota SFB. Our data illustrate that contrary to exerting an anti-inflammatory effect, P2RX7 deficiency actually enhances autoimmune arthritis. Interestingly, SFB colonization can negate the difference in disease severity between WT and P2RX7-deficient mice. We further demonstrated that P2RX7 ablation in the absence of SFB caused reduced apoptotic Tfh cells and enhanced the Tfh response, leading to an increase in autoantibody production. It has been shown that activation of TIGIT, a well-known T cell exhaustion marker, up-regulates anti-apoptotic molecules and promotes T cell survival. We demonstrated that the reduced apoptotic phenotype of P2rx7-/- Tfh cells is associated with their increased expression of TIGIT. This suggested that while P2RX7 was regulating the Tfh population by promoting cell death, TIGIT may have been opposing P2RX7 by inhibiting cell death. Together, these results demonstrated that systemic administration of general P2RX7 antagonists may have detrimental effects in autoimmune therapies, especially in Tfh cell-dependent autoimmune diseases, and cell-specific targeting of P2RX7 should be considered in order to achieve efficacy for P2RX7-related therapy.


Assuntos
Artrite Reumatoide/imunologia , Microbioma Gastrointestinal/imunologia , Receptores Purinérgicos P2X7/deficiência , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Apoptose/imunologia , Artrite Reumatoide/microbiologia , Autoanticorpos/imunologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Nódulos Linfáticos Agregados/imunologia , Receptores Imunológicos/metabolismo , Receptores Purinérgicos P2X7/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
2.
Immunology ; 156(4): 305-318, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30560993

RESUMO

The immune system is essential for maintaining a delicate balance between eliminating pathogens and maintaining tolerance to self-tissues to avoid autoimmunity. An enormous and complex community of gut microbiota provides essential health benefits to the host, particularly by regulating immune homeostasis. Many of the metabolites derived from commensals can impact host health by directly regulating the immune system. Many autoimmune diseases arise from an imbalance between pathogenic effector T cells and regulatory T (Treg) cells. Recent interest has emerged in understanding how cross-talk between gut microbiota and the host immune system promotes autoimmune development by controlling the differentiation and plasticity of T helper and Treg cells. At the molecular level, our recent study, along with others, demonstrates that asymptomatic colonization by commensal bacteria in the gut is capable of triggering autoimmune disease by molecular mimicking self-antigen and skewing the expression of dual T-cell receptors on T cells. Dysbiosis, an imbalance of the gut microbiota, is involved in autoimmune development in both mice and humans. Although it is well known that dysbiosis can impact diseases occurring within the gut, growing literature suggests that dysbiosis also causes the development of gut-distal/non-gut autoimmunity. In this review, we discuss recent advances in understanding the potential molecular mechanisms whereby gut microbiota induces autoimmunity, and the evidence that the gut microbiota triggers gut-distal autoimmune diseases.


Assuntos
Autoimunidade/imunologia , Microbioma Gastrointestinal/imunologia , Linfócitos T/imunologia , Animais , Disbiose/imunologia , Humanos
3.
Artigo em Inglês | MEDLINE | ID: mdl-29755958

RESUMO

Streptococcus pneumoniae causes infection-related mortality worldwide. Immunocompromised individuals, including young children, the elderly, and those with immunodeficiency, are especially vulnerable, yet little is known regarding S. pneumoniae-related pathogenesis and protection in immunocompromised hosts. Recently, strong interest has emerged in the gut microbiota's impact on lung diseases, or the "gut-lung axis." However, the mechanisms of gut microbiota protection against gut-distal lung diseases like pneumonia remain unclear. We investigated the role of the gut commensal, segmented filamentous bacteria (SFB), against pneumococcal pneumonia in immunocompetent and immunocompromised mouse models. For the latter, we chose the Rag-/- model, with adaptive immune deficiency. Immunocompetent adaptive protection against S. pneumoniae infection is based on antibodies against pneumococcal capsular polysaccharides, prototypical T cell independent-II (TI-II) antigens. Although SFB colonization enhanced TI-II antibodies in C57BL/6 mice, our data suggest that SFB did not further protect these immunocompetent animals. Indeed, basal B cell activity in hosts without SFB is sufficient for essential protection against S. pneumoniae. However, in immunocompromised Rag-/- mice, we demonstrate a gut-lung axis of communication, as SFB influenced lung protection by regulating innate immunity. Neutrophil resolution is crucial to recovery, since an unchecked neutrophil response causes severe tissue damage. We found no early neutrophil recruitment differences between hosts with or without SFB; however, we observed a significant drop in lung neutrophils in the resolution phase of S. pneumoniae infection, which corresponded with lower CD47 expression, a molecule that inhibits phagocytosis of apoptotic cells, in SFB-colonized Rag-/- mice. SFB promoted a shift in lung neutrophil phenotype from inflammatory neutrophils expressing high levels of CD18 and low levels of CD62L, to pro-resolution neutrophils with low CD18 and high CD62L. Blocking CD47 in SFB(-) mice increased pro-resolution neutrophils, suggesting CD47 down-regulation may be one neutrophil-modulating mechanism SFB utilizes. The SFB-induced lung neutrophil phenotype remained similar with heat-inactivated S. pneumoniae treatment, indicating these SFB-induced changes in neutrophil phenotype during the resolution phase are not simply secondary to better bacterial clearance in SFB(+) than SFB(-) mice. Together, these data demonstrate that the gut commensal SFB may provide much-needed protection in immunocompromised hosts in part by promoting neutrophil resolution post lung infection.


Assuntos
Anticorpos Antibacterianos/imunologia , Microbioma Gastrointestinal/imunologia , Neutrófilos/imunologia , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/imunologia , Animais , Cápsulas Bacterianas/imunologia , Antígeno CD47/metabolismo , Modelos Animais de Doenças , Hospedeiro Imunocomprometido , Selectina L , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/imunologia , Fagocitose/imunologia , Polissacarídeos Bacterianos/imunologia
4.
Ann N Y Acad Sci ; 1417(1): 57-70, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28984367

RESUMO

To maintain health, the immune system must maintain a delicate balance between eliminating invading pathogens and avoiding immune disorders such as autoimmunity and allergies. The gut microbiota provide essential health benefits to the host, particularly by regulating immune homeostasis. Dysbiosis, an alteration and imbalance of the gut microbiota, is associated with the development of several autoimmune diseases in both mice and humans. In this review, we discuss recent advances in understanding how certain factors, such as age and gender, affect the gut microbiota, which in turn can influence the development of autoimmune diseases. The age factor in microbiota-dependent immune disorders indicates a window of opportunity for future diagnostic and therapeutic approaches. We also discuss unique commensal bacteria with strong immunomodulatory activity. Finally, we provide an overview of the potential molecular mechanisms whereby gut microbiota induce autoimmunity, as well as the evidence that gut microbiota trigger extraintestinal diseases by inducing the migration of gut-derived immune cells. Elucidating the interaction of gut microbiota and the host immune system will help us understand the pathogenesis of immune disorders, and provide us with new foundations to develop novel immuno- or microbe-targeted therapies.


Assuntos
Interações entre Hospedeiro e Microrganismos/imunologia , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/microbiologia , Fatores Etários , Animais , Autoimunidade , Disbiose/imunologia , Disbiose/microbiologia , Epigênese Genética , Ácidos Graxos Voláteis/imunologia , Feminino , Microbioma Gastrointestinal/imunologia , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Doenças do Sistema Imunitário/genética , Imunidade Inata , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Modelos Imunológicos , Mimetismo Molecular/imunologia , Fatores Sexuais , Tretinoína/imunologia , Ácido Úrico/imunologia
5.
Cell Host Microbe ; 22(5): 697-704.e4, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29120746

RESUMO

Lung complications are a major cause of rheumatoid arthritis-related mortality. Involvement of gut microbiota in lung diseases by the gut-lung axis has been widely observed, but the underlying mechanism remains mostly unknown. Using an autoimmune arthritis model, we show that a constituent of the gut microbiota, segmented filamentous bacteria (SFB), distantly provoke lung pathology. SFB induce autoantibodies in lung during the pre-arthritic phase, and SFB-dependent lung pathology requires the T helper 17 (Th17) responses. SFB-induced gut Th17 cells are preferentially recruited to lung over spleen due to robust expression in the lung of the Th17 chemoattractant, CCL20. Additionally, we found that in peripheral tissues, SFB selectively expand dual T cell receptor (TCR)-expressing Th17 cells recognizing both an SFB epitope and self-antigen, thus augmenting autoimmunity. This study reveals mechanisms for commensal-mediated gut-lung crosstalk and dual TCR-based autoimmunity.


Assuntos
Doenças Autoimunes/imunologia , Doenças Autoimunes/microbiologia , Autoimunidade , Bactérias/imunologia , Microbioma Gastrointestinal/imunologia , Pulmão/imunologia , Células Th17/imunologia , Animais , Apoptose/imunologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/microbiologia , Autoanticorpos , Bactérias/patogenicidade , Diferenciação Celular/imunologia , Proliferação de Células , Quimiocina CCL20/metabolismo , Modelos Animais de Doenças , Fezes/microbiologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Baço , Simbiose , Células Th17/metabolismo
6.
Arthritis Res Ther ; 19(1): 188, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28810929

RESUMO

BACKGROUND: Age is an important risk factor for rheumatoid arthritis (RA), which often develops in middle age. However, how age-associated changes in immunity impact RA is poorly understood. Gut microbiota are known to be involved in the pathogenesis of RA, but the effects of microbiota in older subjects remain mostly unknown. METHODS: We used segmented filamentous bacteria (SFB), a gut commensal species with immunomodulatory effects, and K/BxN mice, a T cell receptor (TCR) transgenic model, to study the effect of age and microbiota on autoimmune arthritis. Comparing young and middle-aged K/BxN T cells of the same TCR specificity allows us to study T cells with an age focus eliminating a key variable: TCR repertoire alteration with age. In addition to joints, we also studied pathological changes in the lung, an important extra-articular RA manifestation. We used flow cytometry to evaluate T follicular helper (Tfh) and T helper 17 (Th17) cells, as they both contribute to autoantibody production, a key disease index in both RA and K/BxN arthritis. RESULTS: Middle-aged K/BxN mice had aggravated arthritis and pathological changes in the lung compared to young mice. Middle-aged mice displayed a strong accumulation of Tfh but not Th17 cells, and had defective Th17 differentiation and low expression of interleukin-23, a critical cytokine for Th17 maintenance. Although a soaring Tfh cell population accompanied by robust germinal center B cell responses were found in middle-aged mice, there was decreased cycling of Tfh cells, and SFB only induced the non-Tfh cells to upregulate Bcl-6, the Tfh master transcription factor, in the young but not the middle-aged group. Finally, the accumulated Tfh cells in middle-aged mice had an effector phenotype (CD62LloCD44hi). CONCLUSION: Age-dependent Tfh cell accumulation may play a crucial role in the increased autoimmune disease phenotype in middle-age. SFB, a potent stimulus for inducing Tfh differentiation, fails to promote Tfh differentiation in middle-aged K/BxN mice, suggesting that most of the middle-aged Tfh cells with an effector phenotype are Tfh effector memory cells induced at an earlier age. Our results also indicate that exposure to immunomodulatory commensals may allow the young host to develop an overactive immune system reminiscent of that found in the middle-aged host.


Assuntos
Artrite Reumatoide/imunologia , Doenças Autoimunes/imunologia , Microbioma Gastrointestinal/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Células Th17/imunologia , Fatores Etários , Animais , Artrite Reumatoide/genética , Doenças Autoimunes/genética , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Proliferação de Células/genética , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Células Th17/metabolismo
7.
J Immunol ; 198(5): 1855-1864, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28130500

RESUMO

Rheumatoid arthritis is an autoimmune disorder that affects the joints and other organs. Pulmonary complications contribute significantly to rheumatoid arthritis mortality. Retinoic acid and its synthetic compound AM80 play roles in immunoregulation but their effect on mucosal autoimmunity remains largely unknown. T follicular helper (Tfh) and Th17 cells are known to promote inflammation and autoantibody production. Using the K/BxN autoimmune arthritis model, we elucidate a novel mechanism whereby oral AM80 administration suppressed lung mucosa-associated Tfh and autoantibody responses by increasing the gut-homing α4ß7 integrin expression on Tfh cells. This diverted Tfh cells from systemic (non-gut) inflamed sites such as the lung into the gut-associated lymphoid tissues, Peyer's patches, and thus reduced the systemic autoantibodies. AM80 also inhibited the lung Th17 response. AM80's effect in the lungs was readily applied to the joints as AM80 also inhibited Tfh and Th17 responses in the spleen, the major autoantibody producing site known to correlate with K/BxN arthritis severity. Finally, we used anti-ß7 treatment as an alternative approach, demonstrating that manipulating T cell migration between the gut and systemic sites alters the systemic disease outcome. The ß7 blockade prevented both Tfh and Th17 cells from entering the non-immunopathogenic site, the gut, and retained these T effector cells in the systemic sites, leading to augmented arthritis. These data suggest a dual beneficial effect of AM80, targeting both Tfh and Th17 cells, and warrant strict safety monitoring of gut-homing perturbing agents used in treating intestinal inflammation.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Autoimunidade/efeitos dos fármacos , Benzoatos/uso terapêutico , Pulmão/imunologia , Tetra-Hidronaftalenos/uso terapêutico , Células Th17/imunologia , Animais , Artrite Reumatoide/imunologia , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/prevenção & controle , Autoimunidade/imunologia , Benzoatos/administração & dosagem , Benzoatos/efeitos adversos , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Integrinas/deficiência , Integrinas/genética , Integrinas/imunologia , Intestinos/imunologia , Pulmão/citologia , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Nódulos Linfáticos Agregados/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Tetra-Hidronaftalenos/administração & dosagem , Tetra-Hidronaftalenos/efeitos adversos
8.
Immunity ; 44(4): 875-88, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27096318

RESUMO

Gut microbiota profoundly affect gut and systemic diseases, but the mechanism whereby microbiota affect systemic diseases is unclear. It is not known whether specific microbiota regulate T follicular helper (Tfh) cells, whose excessive responses can inflict antibody-mediated autoimmunity. Using the K/BxN autoimmune arthritis model, we demonstrated that Peyer's patch (PP) Tfh cells were essential for gut commensal segmented filamentous bacteria (SFB)-induced systemic arthritis despite the production of auto-antibodies predominantly occurring in systemic lymphoid tissues, not PPs. We determined that SFB, by driving differentiation and egress of PP Tfh cells into systemic sites, boosted systemic Tfh cell and auto-antibody responses that exacerbated arthritis. SFB induced PP Tfh cell differentiation by limiting the access of interleukin 2 to CD4(+) T cells, thereby enhancing Tfh cell master regulator Bcl-6 in a dendritic cell-dependent manner. These findings showed that gut microbiota remotely regulated a systemic disease by driving the induction and egress of gut Tfh cells.


Assuntos
Artrite/imunologia , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Microbioma Gastrointestinal/imunologia , Nódulos Linfáticos Agregados/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Autoanticorpos/imunologia , Doenças Autoimunes/imunologia , Linfócitos B/imunologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/imunologia , Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Nódulos Linfáticos Agregados/citologia , Proteínas Proto-Oncogênicas c-bcl-6 , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T Auxiliares-Indutores/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...