Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 15(1): e0228251, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31978191

RESUMO

Resistance to chronic Toxoplasma gondii infection requires ongoing recruitment of T cells to the brain. Thus, the factors that promote, sustain, and regulate the T cell response to the parasite in the brain are of great interest. The costimulatory molecule ICOS (inducible T cell costimulator) has been reported to act largely through the PI3K pathway in T cells, and can play pro-inflammatory or pro-regulatory roles depending on the inflammatory context and T cell type being studied. During infection with T. gondii, ICOS promotes early T cell responses, while in the chronic stage of infection ICOS plays a regulatory role by limiting T cell responses in the brain. We sought to characterize the role of ICOS signaling through PI3K during chronic infection using two models of ICOS deficiency: total ICOS knockout (KO) mice and ICOS YF mice that are unable to activate PI3K signaling. Overall, ICOS KO and ICOS YF mice had similar severe defects in parasite-specific IgG production and parasite control compared to WT mice. Additionally, we observed expanded effector T cell populations and a loss of Treg frequency in the brains of both ICOS KO and ICOS YF mice. When comparing the remaining Treg populations in infected mice, ICOS KO Tregs expressed WT levels of Foxp3 and CD25, while ICOS YF Tregs expressed significantly less Foxp3 and CD25 compared to both WT and ICOS KO mice. Together, these results suggest that PI3K-independent signaling downstream of ICOS plays an important role in Treg stability in the context of chronic inflammation.


Assuntos
Encéfalo/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Toxoplasmose/patologia , Animais , Anticorpos Antiprotozoários/sangue , Encéfalo/citologia , Encéfalo/imunologia , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/deficiência , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Leucócitos Mononucleares/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Sítio-Dirigida , Fosfatidilinositol 3-Quinases/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Toxoplasmose/metabolismo , Toxoplasmose/parasitologia
2.
PLoS One ; 14(7): e0219449, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31283790

RESUMO

Signaling through the inducible costimulator ICOS is required for the homeostasis and function of various immune cell populations, with an outstanding role in the generation and maintenance of germinal centers. Very recently, it has been suggested that the clinical phenotype of ICOS-deficient patients is much broader than initially anticipated and the innate immune response might be also affected. However, the role of the ICOS/ICOS-Ligand axis in the homeostasis and development of innate NK cells is not known, and reports on its participation in NK cell activation are scarce. NK cells may express low levels of ICOS that are markedly enhanced upon activation. We show here that ICOS-deficient (ICOS-KO) mice present low NK cell numbers and defects in the homeostasis of these cells, with delayed maturation and altered expression of the developmental NK cell markers CD122, NK1.1, CD11b or CD27. Our experiments in mixed bone marrow chimera mice indicate that, both, cell-intrinsic defects of ICOS-KO NK and deficiencies in the milieu of these mice contribute to the altered phenotype. ICOS-deficient NK cells show impaired production of IFN-γ and cytotoxicity, and a final outcome of defects in NK cell-mediated effector function during the response to poly(I:C) or vaccinia virus infection in vivo. Interestingly, we show that murine innate cells like IL-2-cultured NK and bone marrow-derived dendritic cells can simultaneously express ICOS and ICOS-Ligand; both molecules are functional in NK intracellular signaling, enhancing early phosphorylation of Akt and Erk, or IFN-γ secretion in IL-2-activated NK cells. Our study shows the functional importance of the ICOS/ICOS-L pair in NK cell homeostasis, differentiation and activity and suggests novel therapeutic targets for NK manipulation.


Assuntos
Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Células Matadoras Naturais/metabolismo , Animais , Apoptose , Antígeno CD11b/metabolismo , Diferenciação Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Ligante Coestimulador de Linfócitos T Induzíveis/genética , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/deficiência , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Interferon gama/metabolismo , Interleucina-2/farmacologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Poli I-C/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Vacínia/imunologia , Vacínia/patologia
3.
J Immunol ; 202(4): 1039-1044, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30642977

RESUMO

The ICOS pathway has been implicated in the development and functions of regulatory T (Treg) cells, including those producing IL-10. Treg cell-derived IL-10 is indispensable for the establishment and maintenance of intestinal immune homeostasis. We examined the possible involvement of the ICOS pathway in the accumulation of murine colonic Foxp3- and/or IL-10-expressing cells. We show that ICOS deficiency does not impair induction of IL-10 by intestinal CD4 T cells but, instead, triggers substantial reductions in gut-resident and peripherally derived Foxp3+ Treg cells. ICOS deficiency is associated with reduced demethylation of Foxp3 CNS2 and enhanced loss of Foxp3. This instability significantly limits the ability of ICOS-deficient Treg cells to reverse ongoing inflammation. Collectively, our results identify a novel role for ICOS costimulation in imprinting the functional stability of Foxp3 that is required for the retention of full Treg cell function in the periphery.


Assuntos
Regulação para Baixo , Fatores de Transcrição Forkhead/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Interleucina-10/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Regulação para Baixo/imunologia , Fatores de Transcrição Forkhead/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/deficiência , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-10/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T Reguladores/imunologia
4.
J Infect Dis ; 218(4): 659-668, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-29378030

RESUMO

Staphylococcus aureus is a major cause of both community- and healthcare-acquired pneumonias. Inducible costimulator (ICOS) is part of the CD28 family of proteins and is a target for immune checkpoint therapy. We found ICOS highly expressed on activated CD4 cells in response to S. aureus. In the absence of ICOS, mice had improved survival in a pneumonia model with the methicillin-resistant Staphylococcus aureus (MRSA) strain USA300 and significant reductions in bacterial burden in a nonlethal acute pneumonia model. Infected Icos-/- mice had major reductions in several proinflammatory cytokines, neutrophils, inflammatory monocytes, and eosinophils compared to infected wild-type mice, while there was improved expression of CD11c and macrophage receptor with collagenous structure on the surface of alveolar macrophages. Early during infection infected Icos-/- mice had increased numbers of alveolar macrophages and expression of several surface markers on alveolar macrophages and neutrophils. ICOS signaling also contributed to the pathogenesis of the airway pathogens Klebsiella pneumoniae, Pseudomonas aeruginosa, and Streptococcus pneumoniae, and neutralizing antibody to ICOS led to improved clearance of S. aureus from the airway. Our results indicate that ICOS plays a significant role in orchestrating the innate immune response to S. aureus and other airway pathogens, and could be a potential immunomodulatory target to attenuate S. aureus-related immunopathology.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Pneumonia Estafilocócica/patologia , Animais , Carga Bacteriana , Modelos Animais de Doenças , Fatores Imunológicos/análise , Proteína Coestimuladora de Linfócitos T Induzíveis/deficiência , Infecções por Klebsiella/patologia , Pulmão/patologia , Macrófagos Alveolares/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções Pneumocócicas/patologia , Pneumonia Estafilocócica/microbiologia , Infecções por Pseudomonas/patologia , Análise de Sobrevida
5.
J Immunol ; 191(1): 200-7, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23729441

RESUMO

We and others have previously shown that ICOS plays an important role in inducing acute graft-versus-host disease (GVHD) in murine models of allogeneic bone marrow transplantation. ICOS potentiates TCR-mediated PI3K activation and intracellular calcium mobilization. However, ICOS signal transduction pathways involved in GVHD remain unknown. In this study, we examined the contribution of ICOS-PI3K signaling in the pathogenic potential of T cells using a knock-in mouse strain, ICOS-YF, which selectively lost the ability to activate PI3K. We found that when total T cells were used as alloreactive T cells, ICOS-YF T cells caused less severe GVHD compared with ICOS wild-type T cells, but they induced much more aggressive disease than ICOS knockout T cells. This intermediate level of pathogenic capacity of ICOS-YF T cells was correlated with similar levels of IFN-γ-producing CD8 T cells that developed in the recipients of ICOS-WT or ICOS-YF T cells. We further evaluated the role of ICOS-PI3K signaling in CD4 versus CD8 T cell compartment using GVHD models that are exclusively driven by CD4 or CD8 T cells. Remarkably, ICOS-YF CD8 T cells caused disease similar to ICOS wild-type CD8 T cells, whereas ICOS-YF CD4 T cells behaved very similarly to their ICOS knockout counterparts. Consistent with their in vivo pathogenic potential, CD8 T cells responded to ICOS ligation in vitro by PI3K-independent calcium flux, T cell activation, and proliferation. Thus, in acute GVHD in mice, CD4 T cells heavily rely on ICOS-PI3K signaling pathways; in contrast, CD8 T cells can use PI3K-independent ICOS signaling pathways, possibly through calcium.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/fisiologia , Ativação Linfocitária/imunologia , Fosfatidilinositol 3-Quinase/fisiologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Doença Aguda , Animais , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Doença Enxerto-Hospedeiro/enzimologia , Proteína Coestimuladora de Linfócitos T Induzíveis/deficiência , Ativação Linfocitária/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais/genética , Subpopulações de Linfócitos T/enzimologia , Subpopulações de Linfócitos T/metabolismo
6.
Eur J Immunol ; 43(3): 705-15, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23319295

RESUMO

Foxp3(+) regulatory T (Treg) cells are key immune regulators during helminth infections, and identifying the mechanisms governing their induction is of principal importance for the design of treatments for helminth infections, allergies and autoimmunity. Little is yet known regarding the co-stimulatory environment that favours the development of Foxp3(+) Treg-cell responses during helminth infections. As recent evidence implicates the co-stimulatory receptor ICOS in defining Foxp3(+) Treg-cell functions, we investigated the role of ICOS in helminth-induced Foxp3(+) Treg-cell responses. Infection of ICOS(-/-) mice with Heligmosomoides polygyrus or Schistosoma mansoni led to a reduced expansion and maintenance of Foxp3(+) Treg cells. Moreover, during H. polygyrus infection, ICOS deficiency resulted in increased Foxp3(+) Treg-cell apoptosis, a Foxp3(+) Treg-cell specific impairment in IL-10 production, and a failure to mount putatively adaptive Helios(-) Foxp3(+) Treg-cell responses within the intestinal lamina propria. Impaired lamina propria Foxp3(+) Treg-cell responses were associated with increased production of IL-4 and IL-13 by CD4(+) T cells, demonstrating that ICOS dominantly downregulates Type 2 responses at the infection site, sharply contrasting with its Type 2-promoting effects within lymphoid tissue. Thus, ICOS regulates Type 2 immunity in a tissue-specific manner, and plays a key role in driving Foxp3(+) Treg-cell expansion and function during helminth infections.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Helmintíase/genética , Helmintíase/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Interleucina-10/biossíntese , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Apoptose/genética , Feminino , Proteína Coestimuladora de Linfócitos T Induzíveis/deficiência , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mucosa/imunologia , Mucosa/parasitologia , Nematospiroides dubius/imunologia , Células Th2/imunologia
7.
J Immunol ; 189(1): 234-44, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22661090

RESUMO

We have established a comprehensive in vivo mouse model for the CD4(+) T cell response to an "innocuous" versus "dangerous" exogenous Ag and developed an in vivo test for tolerance. In this model, specific gene-expression signatures, distinctive upregulation of early T cell-communication molecules, and differential expansion of effector T cells (Teff) and regulatory T cells (Treg) were identified as central correlates of T cell tolerance and T cell immunity. Different from essentially all other T cell-activation molecules, ICOS was found to be induced in the immunity response and not by T cells activated under tolerogenic conditions. If expressed, ICOS did not act as a general T cell costimulator but selectively caused a massive expansion of effector CD4(+) T cells, leaving the regulatory CD4(+) T cell compartment largely undisturbed. Thus, ICOS strongly contributed to the dramatic change in the balance between Ag-specific Teff and Treg from ∼1:1 at steady state to 21:1 at the height of the immune response. This newly defined role for the balance of Teff to Treg, together with its known key function in T cell help for B cells, establishes ICOS as a central mediator of immunity. Given its exceptionally selective induction on CD4(+) T cells under inflammatory, but not tolerogenic, conditions, ICOS emerges as a pivotal effector molecule in the early decision between tolerance and immunity to exogenous Ag.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Tolerância Imunológica , Proteína Coestimuladora de Linfócitos T Induzíveis/fisiologia , Imunidade Adaptativa/genética , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Tolerância Imunológica/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/deficiência , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA