Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
PLoS Pathog ; 14(2): e1006869, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29470558

RESUMO

The intestinal immune system must be able to respond to a wide variety of infectious organisms while maintaining tolerance to non-pathogenic microbes and food antigens. The Vitamin A metabolite all-trans-retinoic acid (atRA) has been implicated in the regulation of this balance, partially by regulating innate lymphoid cell (ILC) responses in the intestine. However, the molecular mechanisms of atRA-dependent intestinal immunity and homeostasis remain elusive. Here we define a role for the transcriptional repressor Hypermethylated in cancer 1 (HIC1, ZBTB29) in the regulation of ILC responses in the intestine. Intestinal ILCs express HIC1 in a vitamin A-dependent manner. In the absence of HIC1, group 3 ILCs (ILC3s) that produce IL-22 are lost, resulting in increased susceptibility to infection with the bacterial pathogen Citrobacter rodentium. Thus, atRA-dependent expression of HIC1 in ILC3s regulates intestinal homeostasis and protective immunity.


Assuntos
Imunidade Inata , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Fatores de Transcrição Kruppel-Like/fisiologia , Linfócitos/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Homeostase/genética , Homeostase/imunologia , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Intestinos/microbiologia , Fatores de Transcrição Kruppel-Like/genética , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tretinoína/metabolismo
2.
PLoS Pathog ; 12(9): e1005876, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27598373

RESUMO

The intestine is a common site for a variety of pathogenic infections. Helminth infections continue to be major causes of disease worldwide, and are a significant burden on health care systems. Lysine methyltransferases are part of a family of novel attractive targets for drug discovery. SETD7 is a member of the Suppressor of variegation 3-9-Enhancer of zeste-Trithorax (SET) domain-containing family of lysine methyltransferases, and has been shown to methylate and alter the function of a wide variety of proteins in vitro. A few of these putative methylation targets have been shown to be important in resistance against pathogens. We therefore sought to study the role of SETD7 during parasitic infections. We find that Setd7-/- mice display increased resistance to infection with the helminth Trichuris muris but not Heligmosomoides polygyrus bakeri. Resistance to T. muris relies on an appropriate type 2 immune response that in turn prompts intestinal epithelial cells (IECs) to alter differentiation and proliferation kinetics. Here we show that SETD7 does not affect immune cell responses during infection. Instead, we found that IEC-specific deletion of Setd7 renders mice resistant to T. muris by controlling IEC turnover, an important aspect of anti-helminth immune responses. We further show that SETD7 controls IEC turnover by modulating developmental signaling pathways such as Hippo/YAP and Wnt/ß-Catenin. We show that the Hippo pathway specifically is relevant during T. muris infection as verteporfin (a YAP inhibitor) treated mice became susceptible to T. muris. We conclude that SETD7 plays an important role in IEC biology during infection.


Assuntos
Intestinos/imunologia , Proteínas Metiltransferases/metabolismo , Transdução de Sinais , Tricuríase/imunologia , Trichuris/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Proliferação de Células , Citocinas/metabolismo , Resistência à Doença , Células Epiteliais/parasitologia , Células Epiteliais/fisiologia , Deleção de Genes , Histona-Lisina N-Metiltransferase , Humanos , Intestinos/parasitologia , Intestinos/fisiologia , Camundongos , Especificidade de Órgãos , Fosfoproteínas/metabolismo , Porfirinas/efeitos adversos , Proteínas Metiltransferases/genética , Tricuríase/parasitologia , Tricuríase/patologia , Verteporfina , Proteínas de Sinalização YAP , beta Catenina/metabolismo
3.
Eur J Immunol ; 46(1): 122-30, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26518471

RESUMO

In mouse models of infection with the gastrointestinal parasite Trichuris muris, appropriate dendritic-cell (DC) Ag sampling, migration, and presentation to T cells are necessary to mount a protective Th2-polarized adaptive immune response, which is needed to clear infection. SH2-containing inositol 5'-phosphatase 1 (SHIP-1) has been shown to be an important regulator of DC function in vitro through the negative regulation of the phosphoinositide 3-kinase (PI3K) pathway, but its role in vivo is relatively unexplored. In the current work, mice with a specific deletion of SHIP-1 in DCs (Ship1(ΔDC) ) were infected with the parasite T. muris. Ship1(ΔDC) mice were susceptible to infection due to ineffective priming of Th2-polarized responses. This is likely due to an increased production of interleukin (IL) 12p40 by SHIP-1-deficient DCs, as in vivo antibody blockade of IL-12p40 was able to facilitate the clearing of infection in Ship1(ΔDC) mice. Our results describe a critical role for SHIP-1 in regulating the ability of DCs to efficiently prime Th2-type responses.


Assuntos
Células Dendríticas/imunologia , Ativação Linfocitária/imunologia , Monoéster Fosfórico Hidrolases/imunologia , Células Th2/imunologia , Tricuríase/imunologia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Inositol Polifosfato 5-Fosfatases , Camundongos , Camundongos Mutantes , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trichuris/imunologia
4.
Eur J Immunol ; 46(11): 2587-2596, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27594558

RESUMO

Proinflammatory cytokines produced during immune responses to infectious stimuli are well-characterized to have secondary effects on the function of hematopoietic progenitor cells in the BM. However, these effects on the BM are poorly characterized during chronic infection with intestinal helminth parasites. In this study, we use the Trichuris muris model of infection and show that Th1 cell-associated, but not acute Th2 cell-associated, responses to chronic T. muris infection cause a major, transient expansion of CD48- CD150- multipotent progenitor cells in the BM that is dependent on the presence of adaptive immune cells and IFN-γ signaling. Chronic T. muris infection also broadly stimulated proliferation of BM progenitor cells including CD48- CD150+ hematopoietic stem cells. This shift in progenitor activity during chronic T. muris infection correlated with a functional increase in myeloid colony formation in vitro as well as neutrophilia in the BM and peripheral blood. In parallel, we observed an accumulation of CD4+ , CD8+ , and CD4- CD8- (double negative) T cells that expressed IFN-γ, displaying activated and central memory-type phenotypes in the bone marrow during chronic infection. Thus, these results demonstrate that Th1 cell-driven responses in the intestine during chronic helminth infection potently influence upstream hematopoietic processes in the BM via IFN-γ.


Assuntos
Medula Óssea/imunologia , Hematopoese/imunologia , Interferon gama/imunologia , Células Th1/imunologia , Células Th2/imunologia , Tricuríase/sangue , Tricuríase/imunologia , Animais , Doença Crônica , Modelos Animais de Doenças , Células-Tronco Hematopoéticas/fisiologia , Memória Imunológica , Interferon gama/biossíntese , Interferon gama/genética , Intestinos/imunologia , Camundongos , Tricuríase/parasitologia , Trichuris/imunologia , Trichuris/fisiologia
5.
Infect Immun ; 84(2): 491-501, 2016 02.
Artigo em Inglês | MEDLINE | ID: mdl-26644379

RESUMO

Immunological cross talk between mucosal tissues such as the intestine and the lung is poorly defined during homeostasis and disease. Here, we show that a low-dose infection with the intestinally restricted helminth parasite Trichuris muris results in the production of Th1 cell-dependent gamma interferon (IFN-γ) and myeloid cell-derived interleukin-10 (IL-10) in the lung without causing overt airway pathology. This cross-mucosal immune response in the lung inhibits the development of papain-induced allergic airway inflammation, an innate cell-mediated type 2 airway inflammatory disease. Thus, we identify convergent and nonredundant roles of adaptive and innate immunity in mediating cross-mucosal suppression of type 2 airway inflammation during low-dose helminth-induced intestinal inflammation. These results provide further insight in identifying novel intersecting immune pathways elicited by gut-to-lung mucosal cross talk.


Assuntos
Enteropatias Parasitárias/imunologia , Intestinos/imunologia , Intestinos/parasitologia , Pulmão/imunologia , Hipersensibilidade Respiratória/imunologia , Tricuríase/imunologia , Trichuris/imunologia , Imunidade Adaptativa , Animais , Antígenos de Dermatophagoides/imunologia , Asma/imunologia , Asma/prevenção & controle , Linfócitos T CD4-Positivos/imunologia , Comunicação Celular , Relação Dose-Resposta Imunológica , Interações Hospedeiro-Parasita , Imunidade Inata , Imunidade nas Mucosas , Inflamação/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-10/biossíntese , Interleucina-10/imunologia , Enteropatias Parasitárias/parasitologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Papaína , Tricuríase/parasitologia , Trichuris/patogenicidade
6.
J Allergy Clin Immunol ; 136(3): 725-736.e2, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25746967

RESUMO

BACKGROUND: Inpp5d (Src homology 2 domain-containing inositol-5-phosphatase [Ship1])-deficient mice experience spontaneous airway inflammation and have enhanced sensitivity to allergen-induced airway inflammation. OBJECTIVE: We hypothesized that lineage-specific deletion of Ship1 expression in cells known to be crucial for adaptive TH2 responses would uncover distinct roles that could either positively or negatively regulate susceptibility to allergic airway inflammation (AAI). METHODS: Ship1 expression was deleted in B cells, T cells, or dendritic cells (DCs), and the resulting Ship1(ΔB cell), Ship1(ΔT cell), Ship1(ΔDC), or Ship1(F/F) (wild-type) control mice were evaluated in a model of house dust mite (HDM)-induced AAI. RESULTS: Unlike germline panhematopoietic Ship1 deletion, deletion of Ship1 selectively in either the B-cell, T-cell, or DC lineages did not result in spontaneous airway inflammation. Strikingly, although loss of Ship1 in the B-cell lineage did not affect HDM-induced AAI, loss of Ship1 in either of the T-cell or DC lineages protected mice from AAI by skewing the typical TH2 immune response toward a TH1 response. CONCLUSIONS: Although panhematopoietic deletion of Ship1 leads to spontaneous lung inflammation, selective deletion of Ship1 in T cells or DCs impairs the formation of an adaptive TH2 response and protects animals from HDM-induced AAI.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Linhagem da Célula/imunologia , Células Dendríticas/imunologia , Monoéster Fosfórico Hidrolases/imunologia , Pneumonia/imunologia , Linfócitos T/imunologia , Imunidade Adaptativa , Alérgenos/administração & dosagem , Alérgenos/imunologia , Animais , Antígenos de Dermatophagoides/administração & dosagem , Antígenos de Dermatophagoides/imunologia , Linfócitos B/imunologia , Linfócitos B/patologia , Hiper-Reatividade Brônquica/induzido quimicamente , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/patologia , Linhagem da Célula/genética , Células Dendríticas/patologia , Expressão Gênica , Inositol Polifosfato 5-Fosfatases , Camundongos , Camundongos Knockout , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Pneumonia/induzido quimicamente , Pneumonia/genética , Pneumonia/patologia , Pyroglyphidae/química , Linfócitos T/patologia , Equilíbrio Th1-Th2
7.
Immunol Cell Biol ; 93(3): 245-52, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25582341

RESUMO

Repressive epigenetic modifications such as dimethylation and trimethylation histone H3 at lysine 9 (H3K9me2 and H3K9me3) and H3K27me3 have been shown to be critical for embryonic stem (ES) cell differentiation by silencing cell lineage-promiscuous genes. CD4(+) T helper (T(H)) cell differentiation is a powerful model to study the molecular mechanisms associated with cellular lineage choice in adult cells. Naïve T(H) cells have the capacity to differentiate into one of the several phenotypically and functionally distinct and stable lineages. Although some repressive epigenetic mechanisms have a critical role in T(H) cell differentiation in a similar manner to that in ES cells, it is clear that there are disparate functions for certain modifications between ES cells and T(H) cells. Here we review the role of repressive histone modifications in the differentiation and function of T(H) cells in health and disease.


Assuntos
Diferenciação Celular , Repressão Epigenética , Histonas/metabolismo , Inflamação/genética , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Antígenos CD4/metabolismo , Humanos , Inflamação/imunologia , Metilação
8.
J Allergy Clin Immunol ; 133(4): 1142-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24679471

RESUMO

BACKGROUND: Allergic inflammation involves the sensitization of naive CD4(+) T cells to allergens, resulting in a TH2-skewed inflammatory response. Although antigen presentation by dendritic cells to T cells in the lymph node is crucial for TH2 cell development, the innate signals that initiate adaptive type 2 inflammation and the role of group 2 innate lymphoid cells (ILC2s) are poorly understood. OBJECTIVE: We sought to investigate the influence of ILC2s and the route of priming on the development of an adaptive type 2 immune response to lung allergens. METHODS: Wild-type and ILC2-deficient mice were exposed intranasally or systemically to the TH2-inducing antigens house dust mite or ovalbumin in a model of allergic airway inflammation or the TH17-inducing bacterial antigen Saccharopolyspora rectivirgula in a model of hypersensitivity pneumonitis. The formation of an adaptive immune response was evaluated based on serum antibody titers and production of T cell-derived cytokines (IL-4, IL-5, IL-13 and IL-17A). RESULTS: We find that lung ILC2s play a critical role in priming the adaptive type 2 immune response to inhaled allergens, including the recruitment of eosinophils, TH2 cytokine production and serum IgE levels. Surprisingly, systemic priming with ovalbumin, with or without adjuvants, circumvents the requirement for ILC2s in inducing TH2-driven lung inflammation. ILC2s were also found to be dispensable for the sensitization to TH1- or TH17-inducing antigens. CONCLUSION: These data highlight a critical role for ILC2s in the development of adaptive type 2 responses to local, but not systemic, antigen exposure.


Assuntos
Alérgenos/imunologia , Imunidade Inata , Subpopulações de Linfócitos/imunologia , Células Th2/imunologia , Imunidade Adaptativa , Animais , Asma/imunologia , Asma/metabolismo , Asma/patologia , Interleucina-5/biossíntese , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos/metabolismo , Camundongos , Camundongos Transgênicos , Pyroglyphidae/imunologia , Células Th17/imunologia , Células Th17/metabolismo , Células Th2/metabolismo
9.
J Immunol ; 188(8): 3839-50, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22430739

RESUMO

Although SHIP is a well-established suppressor of IgE plus Ag-induced degranulation and cytokine production in bone marrow-derived mast cells (BMMCs), little is known about its role in connective tissue (CTMCs) or mucosal (MMCs) mast cells. In this study, we compared SHIP's role in the development as well as the IgE plus Ag and TLR-induced activation of CTMCs, MMCs, and BMMCs and found that SHIP delays the maturation of all three mast cell subsets and, surprisingly, that it is a positive regulator of IgE-induced BMMC survival. We also found that SHIP represses IgE plus Ag-induced degranulation of all three mast cell subsets and that TLR agonists do not trigger their degranulation, whether SHIP is present or not, nor do they enhance IgE plus Ag-induced degranulation. In terms of cytokine production, we found that in MMCs and BMMCs, which are poor producers of TLR-induced cytokines, SHIP is a potent negative regulator of IgE plus Ag-induced IL-6 and TNF-α production. Surprisingly, however, in splenic or peritoneal derived CTMCs, which are poor producers of IgE plus Ag-induced cytokines, SHIP is a potent positive regulator of TLR-induced cytokine production. Lastly, cell signaling and cytokine production studies with and without LY294002, wortmannin, and PI3Kα inhibitor-2, as well as with PI3K p85α(-/-) BMMCs and CTMCs, are consistent with SHIP positively regulating TLR-induced cytokine production via an adaptor-mediated pathway while negatively regulating IgE plus Ag-induced cytokine production by repressing the PI3K pathway.


Assuntos
Imunoglobulina E/imunologia , Mastócitos/imunologia , Mucosa/imunologia , Monoéster Fosfórico Hidrolases/imunologia , Animais , Antígenos/imunologia , Degranulação Celular/imunologia , Diferenciação Celular , Linhagem da Célula/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Regulação da Expressão Gênica , Inositol Polifosfato 5-Fosfatases , Interleucina-6/biossíntese , Interleucina-6/imunologia , Mastócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa/citologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Inibidores de Fosfoinositídeo-3 Quinase , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Monoéster Fosfórico Hidrolases/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
10.
J Immunol ; 186(7): 3858-65, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21339365

RESUMO

We previously showed that monophosphoryl lipid A (MLA) activates TLR4 in dendritic cells (DCs) in a Toll/IL-1R domain-containing adaptor inducing IFN-ß (TRIF)-biased manner: MLA produced from Salmonella minnesota Re595 induced signaling events and expression of gene products that were primarily TRIF dependent, whereas MyD88-dependent signaling was impaired. Moreover, when tested in TRIF-intact/MyD88-deficient DCs, synthetic MLA of the Escherichia coli chemotype (sMLA) showed the same activity as its diphosphoryl, inflammatory counterpart (synthetic diphosphoryl lipid A), indicating that TRIF-mediated signaling is fully induced by sMLA. Unexpectedly, we found that the transcript level of one proinflammatory cytokine was increased in sMLA-treated cells by MyD88 deficiency to the higher level induced by synthetic diphosphoryl lipid A, which suggested MyD88 may paradoxically help restrain proinflammatory signaling by TRIF-biased sMLA. In this article, we demonstrate that sMLA induces MyD88 recruitment to TLR4 and activates the anti-inflammatory lipid phosphatase SHIP1 in an MyD88-dependent manner. At the same time, MyD88-dependent signaling activity at the level of IL-1R-associated kinase 1 is markedly reduced. Increased SHIP1 activity is associated with reductions in sMLA-induced IκB kinase α/ß and IFN regulatory factor 3 activation and with restrained expression of their downstream targets, endothelin-1 and IFN-ß, respectively. Results of this study identify a pattern that is desirable in the context of vaccine adjuvant design: TRIF-biased sMLA can stimulate partial MyD88 activity, with MyD88-dependent SHIP1 helping to reduce proinflammatory signaling in DCs.


Assuntos
Adjuvantes Imunológicos/fisiologia , Células Dendríticas/imunologia , Mediadores da Inflamação/fisiologia , Lipídeo A/análogos & derivados , Fator 88 de Diferenciação Mieloide/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/metabolismo , Adjuvantes Imunológicos/antagonistas & inibidores , Adjuvantes Imunológicos/metabolismo , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/microbiologia , Células da Medula Óssea/patologia , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Escherichia coli/imunologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Inositol Polifosfato 5-Fosfatases , Lipídeo A/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Salmonella/imunologia , Transdução de Sinais/genética , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/fisiologia
11.
J Immunol ; 186(1): 323-32, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21131429

RESUMO

We report that SHIP(-/-) mice, compared to SHIP(+/+) mice, are Th2 skewed with elevated serum IgE and twice as many splenic CD4(+) Th2 cells that, when stimulated with anti-CD3, produce more IL-4 and less IFN-γ. Exploring the reason for this Th2 skewing, we found that freshly isolated SHIP(-/-) splenic and bone marrow basophils are present in elevated numbers and secrete far more IL-4 in response to IL-3 or to FcεRI stimulation than do WT basophils. These SHIP(-/-) basophils markedly skew wild-type macrophage colony stimulating factor-derived macrophages toward an M2 phenotype, stimulate OT-II CD4(+) Th cells to differentiate into Th2 cells, and trigger SHIP(+/+) B cells to become IgE-producing cells. All these effects are completely abrogated with neutralizing anti-IL-4 Ab. Exploring the cell signaling pathways responsible for hyperproduction of IL-4 by SHIP(-/-) basophils, we found that IL-3-induced activation of the PI3K pathway is significantly enhanced and that PI3K inhibitors, especially a p110α inhibitor, dramatically suppresses IL-4 production from these cells. In vivo studies, in which basophils were depleted from mast cell-deficient SHIP(+/+) and SHIP(-/-) mice, confirmed the central role that basophils play in the Th2 skewing of naive SHIP-deficient mice. Taken together, these studies demonstrate that SHIP is a potent negative regulator of IL-4 production from basophils and thus may be a novel therapeutic target for Th1- and Th2-related diseases.


Assuntos
Basófilos/imunologia , Diferenciação Celular/imunologia , Interleucina-4/antagonistas & inibidores , Interleucina-4/biossíntese , Monoéster Fosfórico Hidrolases/fisiologia , Proteínas Repressoras/fisiologia , Células Th2/citologia , Células Th2/imunologia , Animais , Basófilos/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Células Cultivadas , Imunoglobulina E/biossíntese , Imunoglobulina E/sangue , Inositol Polifosfato 5-Fosfatases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Células Th2/metabolismo
12.
J Immunol ; 185(8): 4545-53, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20844190

RESUMO

Resident tissue macrophages (Mφs) continually survey the microenvironment, ingesting Ags and presenting them on their surface for recognition by T cells. Because these Ags can be either host cell- or pathogen-derived, Mφs must be able to distinguish whether a particular Ag should provoke an immune response or be tolerated. However, the mechanisms that determine whether Mφs promote or inhibit T cell activation are not well understood. To investigate this, we first determined the mechanism by which murine resident peritoneal Mφs suppress in vitro T cell proliferation in the absence of pathogens and then explored the effects of different pathogen-derived molecules on Mφ immunosuppression. Our results suggest that, in response to IFN-γ, which is secreted by TCR-activated T cells, resident peritoneal Mφs acquire immunosuppressive properties that are mediated by NO. However, pretreatment of Mφs with LPS or dsRNA, but not CpG or peptidoglycan, eliminates their suppressive properties, in part via the induction of autocrine-acting IFN-ß. These results suggest TLR agonists that activate TRIF, and consequently induce IFN-ß, but not those that exclusively signal through MyD88, abrogate the immunosuppressive properties of Mφs, and thus promote T cell expansion and elimination of invading microorganisms.


Assuntos
Tolerância Imunológica/imunologia , Interferon beta/biossíntese , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Animais , Western Blotting , Proliferação de Células , Separação Celular , Citometria de Fluxo , Interferon beta/imunologia , Lipopolissacarídeos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , RNA de Cadeia Dupla/imunologia , Linfócitos T/metabolismo , Receptores Toll-Like/agonistas
13.
J Immunol ; 184(6): 2805-13, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20154203

RESUMO

Although several groups have investigated the role of SHIP in macrophage (M) development and function, SHIP's contribution to the generation, maturation, and innate immune activation of dendritic cells (DCs) is poorly understood. We show herein that SHIP negatively regulates the generation of DCs from bone marrow precursors in vitro and in vivo, as illustrated by the enhanced expansion of DCs from SHIP(-/-) GM-CSF cultures, as well as increased numbers of DCs in the spleens of SHIP-deficient mice. Interestingly, however, these SHIP(-/-) DCs display a relatively immature phenotype and secrete substantially lower levels of IL-12 after TLR ligand stimulation than wild type DCs. This, in turn, leads to a dramatically reduced stimulation of Ag-specific T cell proliferation and Th1 cell responses in vitro and in vivo. This immature phenotype of SHIP(-/-) DCs could be reversed with the PI3K inhibitors LY294002 and wortmannin, suggesting that SHIP promotes DC maturation by reducing the levels of the PI3K second messenger phosphatidylinositol-3,4,5-trisphosphate. These results are consistent with SHIP being a negative regulator of GM-CSF-derived DC generation but a positive regulator of GM-CSF-derived DC maturation and function.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Células Dendríticas/citologia , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Imunidade Inata/genética , Inositol Polifosfato 5-Fosfatases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Células-Tronco/citologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Receptores Toll-Like/fisiologia , Regulação para Cima/genética , Regulação para Cima/imunologia
14.
Blood ; 113(13): 2945-54, 2009 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-19139077

RESUMO

Gram-negative bacterial infections, unlike viral infections, do not typically protect against subsequent viral infections. This is puzzling given that lipopolysaccharide (LPS) and double-stranded (ds) RNA both activate the TIR domain-containing adaptor-inducing interferon beta (TRIF) pathway and, thus, are both capable of eliciting an antiviral response by stimulating type I interferon (IFN) production. We demonstrate herein that SH2-containing inositol-5'-phosphatase (SHIP) protein levels are dramatically increased in murine macrophages via the MyD88-dependent pathway, by up-regulating autocrine-acting transforming growth factor-beta (TGFbeta). The increased SHIP then mediates, via inhibition of the phosphatidylinositol-3-kinase (PI3K) pathway, cytosine-phosphate-guanosine (CPG)- and LPS-induced tolerance and cross-tolerance and restrains IFN-beta production induced by a subsequent exposure to LPS or dsRNA. Intriguingly, we found, using isoform-specific PI3K inhibitors, that LPS- or cytosine-phosphate-guanosine-induced interleukin-6 (IL-6) is positively regulated by p110alpha, -gamma, and -delta but negatively regulated by p110beta. This may explain some of the controversy concerning the role of PI3K in Toll-like receptor-induced cytokine production. Consistent with our in vitro findings, SHIP(-/-) mice overproduce IFN-beta in response to LPS, and this leads to antiviral hypothermia. Thus, up-regulation of SHIP in response to Gram-negative bacterial infections probably explains the inability of such infections to protect against subsequent viral infections.


Assuntos
Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Lipopolissacarídeos/farmacologia , Monoéster Fosfórico Hidrolases/genética , Vírus/imunologia , Animais , Células Cultivadas , Ilhas de CpG/imunologia , Ilhas de CpG/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Hipotermia/genética , Hipotermia/imunologia , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/genética , Inositol Polifosfato 5-Fosfatases , Interferon beta/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/fisiologia , Monoéster Fosfórico Hidrolases/metabolismo , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia
15.
J Immunol ; 182(9): 5507-14, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380799

RESUMO

Reports showing that W/W(v) mice are protected from experimental autoimmune encephalomyelitis (EAE, a murine model of multiple sclerosis), have implicated mast cells as an essential component in disease susceptibility, but the role of mast cell trafficking has not been addressed. In this study, we have used both mast cell transplantation and genetic mutations (Cd34(-/-), W/W(v), W(sh)/W(sh)) to investigate the role of mast cell trafficking in EAE in detail. We show, for the first time, that bone marrow-derived mast cells are actively recruited to the CNS during EAE. Unexpectedly, however, we found that EAE develops unabated in two independent genetic backgrounds in the complete absence of mast cells or bone marrow-derived mast cell reconstitution. We conclude that although mast cells do accumulate in the brain and CNS during demyelinating disease via peripheral mast cell trafficking, they are completely dispensable for development of disease.


Assuntos
Movimento Celular/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Predisposição Genética para Doença , Mastócitos/imunologia , Mastócitos/patologia , Medula Espinal/imunologia , Medula Espinal/patologia , Transferência Adotiva , Sequência de Aminoácidos , Animais , Antígenos CD34/genética , Antígenos CD34/fisiologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Movimento Celular/genética , Encefalomielite Autoimune Experimental/genética , Feminino , Mastócitos/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Dados de Sequência Molecular , Medula Espinal/metabolismo
16.
J Immunol ; 183(1): 228-36, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19542434

RESUMO

SHIP1 inhibits immune receptor signaling through hydrolysis of the PI3K product phosphatidylinositol 3,4,5-trisphosphate, forming phosphatidylinositol 3,4-bisphosphate. In mast cells, SHIP1 represses FcepsilonRI- and cytokine-mediated activation in vitro, but little is known regarding the function of SHIP1 in mast cells in vivo or the susceptibility of Ship1(-/-) mice to mast cell-associated diseases. In this study, we found that Ship1(-/-) mice have systemic mast cell hyperplasia, increased serum levels of IL-6, TNF, and IL-5, and heightened anaphylactic response. Further, by reconstituting mast cell-deficient mice with Ship1(+/+) or Ship1(-/-) mast cells, we found that the above defects were due to loss of SHIP1 in mast cells. Additionally, we found that mice reconstituted with Ship1(-/-) mast cells suffered worse allergic asthma pathology than those reconstituted with Ship1(+/+) mast cells. In summary, our data show that SHIP1 represses allergic inflammation and mast cell hyperplasia in vivo and exerts these effects specifically in mast cells.


Assuntos
Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Hipersensibilidade/patologia , Hipersensibilidade/prevenção & controle , Mediadores da Inflamação/fisiologia , Mastócitos/enzimologia , Mastócitos/patologia , Monoéster Fosfórico Hidrolases/fisiologia , Anafilaxia/enzimologia , Anafilaxia/genética , Anafilaxia/patologia , Animais , Células Cultivadas , Citocinas/fisiologia , Feminino , Hiperplasia/enzimologia , Hiperplasia/genética , Hiperplasia/prevenção & controle , Hipersensibilidade/enzimologia , Hipersensibilidade/genética , Inositol Polifosfato 5-Fosfatases , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética
17.
J Immunol ; 183(6): 3652-60, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19710468

RESUMO

There is a great deal of interest in determining what regulates the generation of classically activated (M1) vs alternatively activated (M2) macrophages (Mphis) because of the opposing effects that these two Mphi subsets have on tumor progression. We show herein that IL-3 and, to a lesser extent, GM-CSF skew murine Mphi progenitors toward an M2 phenotype, especially in the absence of SHIP. Specifically, the addition of these cytokines, with or without M-CSF, to adherence- or lineage-depleted (Lin(-)) SHIP(-/-) bone marrow (BM) cells induces high levels of the M2 markers, arginase I, and Ym1 in the resulting mature Mphis. These in vitro-derived mature Mphis also display other M2 characteristics, including an inability to enhance anti-CD3-stimulated splenic T cell secretion of IFN-gamma and low IL-12 and high IL-10 production in response to LPS. Not surprisingly, given that IL-3 and GM-CSF utilize STAT5 to trigger many downstream signaling pathways, this M2 phenotype is suppressed when STAT5(-/-) BM cells are used. Unexpectedly, however, this M2 phenotype is also suppressed when STAT6(-/-) BM cells are used, suggesting that IL-4- or IL-13-induced signaling might be involved. Consistent with this, we found that IL-3 and GM-CSF stimulate the production of IL-4, especially from SHIP(-/-) Lin(-) BM cells, and that neutralizing anti-IL-4 Abs block IL-3-induced M2 skewing. Moreover, we found that basophil progenitors within the Lin(-) BM are responsible for this IL-3- and GM-CSF-induced IL-4 production, and that SHIP represses M2 skewing not by preventing skewing within Mphis themselves but by inhibiting IL-4 production from basophils.


Assuntos
Basófilos/metabolismo , Diferenciação Celular/imunologia , Interleucina-3/farmacologia , Interleucina-4/biossíntese , Macrófagos/citologia , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Inositol Polifosfato 5-Fosfatases , Ativação de Macrófagos , Camundongos , Células-Tronco/citologia
18.
Front Biosci ; 12: 2836-48, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17485263

RESUMO

The SH2-containing inositol-5'-phosphatase, SHIP, represses the proliferation, survival, and activation of hematopoietic cells, in large part by translocating to membranes following extracellular stimulation and hydrolysing the phosphatidylinositol-3-kinase (PI3K)-generated second messenger PI-3,4,5-P3 (PIP3) to PI-3,4-P2. SHIP-/- mice have, as a result, an increased number of monocyte/macrophages because their progenitors display enhanced survival and proliferation, as well as more rapid differentiation. Interestingly, SHIP-/- mice do not display lipopolysaccharide (LPS)- or CpG oligonucleotide-induced tolerance because this blunting of inflammatory mediator production is contingent upon LPS- and CpG-induced upregulation of SHIP in their macrophages and mast cells. This upregulation is mediated via the production of autocrine-acting TGFbeta which is induced via the MyD88-dependent pathway. The increased levels of SHIP then inhibit both MyD88-dependent and independent signaling. Intriguingly, SHIP-/- peritoneal and alveolar macrophages produce less nitric oxide (NO) than wild-type macrophages because they have constitutively high arginase I levels and this enzyme competes with inducible nitric oxide synthase (iNOS) for the substrate L-arginine. It is likely that, in the face of chronically elevated PIP3 levels in their myeloid progenitors, SHIP-/- mice display a skewed development away from M1 (killer) macrophages towards M2 (healing) macrophages. This suggests that SHIP plays a critical role in programming macrophages.


Assuntos
Macrófagos/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Tolerância Imunológica/fisiologia , Inositol Polifosfato 5-Fosfatases , Camundongos , Camundongos Knockout , Fenótipo , Monoéster Fosfórico Hidrolases/química , Monoéster Fosfórico Hidrolases/genética
19.
J Leukoc Biol ; 101(4): 893-900, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28034915

RESUMO

The incidence of inflammatory bowel diseases (IBDs) has steadily increased in recent decades-a phenomenon that cannot be explained by genetic mutations alone. Other factors, including the composition of the intestinal microbiome, are potentially important contributors to the increased occurrence of this group of diseases. Previous reports have shown a correlation between early-life antibiotic (Abx) treatment and an increased incidence of IBD. In this report, we investigated the effects of early-life Abx treatments on the pathogenicity of CD4+ T cells using an experimental T cell transfer model of IBD. Our results show that CD4+ T cells isolated from adult mice that had been treated with Abx during gestation and in early life induced a faster onset of IBD in Rag1-deficient mice compared with CD4+ T cells of untreated mice. Ex vivo functional analyses of IBD-inducing CD4+ T cells did not show significant differences in their immunologic potential ex vivo, despite their in vivo phenotype. However, genome-wide gene-expression analysis revealed that these cells displayed dysregulated expression of genes associated with cell-cycle regulation, metabolism, and cellular stress. Analysis of Abx-treated CD4+ T cell donors showed systemically elevated levels of the stress hormone corticosterone throughout life compared with untreated donors. The cohousing of Abx-treated mice with untreated mice decreased serum corticosterone, and a consequent transfer of the cells from cohoused mice into Rag1-deficient mice restored the onset and severity of disease to that of untreated animals. Thus, our results suggest that early-life Abx treatment results in a stress response with high levels of corticosterone that influences CD4+ T cell function.


Assuntos
Antibacterianos/farmacologia , Linfócitos T CD4-Positivos/imunologia , Inflamação/imunologia , Inflamação/patologia , Mucosa Intestinal/patologia , Animais , Antibacterianos/uso terapêutico , Linfócitos T CD4-Positivos/efeitos dos fármacos , Corticosterona/sangue , Citocinas/genética , Citocinas/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Genoma , Abrigo para Animais , Inflamação/sangue , Doenças Inflamatórias Intestinais/sangue , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/metabolismo
20.
Sci Immunol ; 1(3)2016 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-28670633

RESUMO

Fibrosis is the result of dysregulated tissue regeneration and is characterized by excessive accumulation of matrix proteins that become detrimental to tissue function. In Crohn's disease, this manifests itself as recurrent gastrointestinal strictures for which there is no effective therapy beyond surgical intervention. Using a model of infection-induced chronic gut inflammation, we show that Rora-deficient mice are protected from fibrosis; infected intestinal tissues display diminished pathology, attenuated collagen deposition and reduced fibroblast accumulation. Although Rora is best known for its role in ILC2 development, we find that Salmonella-induced fibrosis is independent of eosinophils, STAT6 signaling and Th2 cytokine production arguing that this process is largely ILC2-independent. Instead, we observe reduced levels of ILC3- and T cell-derived IL-17A and IL-22 in infected gut tissues. Furthermore, using Rorasg/sg /Rag1-/- bone marrow chimeric mice, we show that restoring ILC function is sufficient to re-establish IL-17A and IL-22 production and a profibrotic phenotype. Our results show that RORα-dependent ILC3 functions are pivotal in mediating gut fibrosis and they offer an avenue for therapeutic intervention in Crohn's-like diseases.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA