Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Cell ; 177(5): 1201-1216.e19, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-31031005

RESUMO

Innate immune responses are intricately linked with intracellular metabolism of myeloid cells. Toll-like receptor (TLR) stimulation shifts intracellular metabolism toward glycolysis, while anti-inflammatory signals depend on enhanced mitochondrial respiration. How exogenous metabolic signals affect the immune response is unknown. We demonstrate that TLR-dependent responses of dendritic cells (DCs) are exacerbated by a high-fatty-acid (FA) metabolic environment. FAs suppress the TLR-induced hexokinase activity and perturb tricarboxylic acid cycle metabolism. These metabolic changes enhance mitochondrial reactive oxygen species (mtROS) production and, in turn, the unfolded protein response (UPR), leading to a distinct transcriptomic signature with IL-23 as hallmark. Interestingly, chemical or genetic suppression of glycolysis was sufficient to induce this specific immune response. Conversely, reducing mtROS production or DC-specific deficiency in XBP1 attenuated IL-23 expression and skin inflammation in an IL-23-dependent model of psoriasis. Thus, fine-tuning of innate immunity depends on optimization of metabolic demands and minimization of mtROS-induced UPR.


Assuntos
Microambiente Celular/imunologia , Células Dendríticas/imunologia , Imunidade Inata , Mitocôndrias/imunologia , Espécies Reativas de Oxigênio/imunologia , Resposta a Proteínas não Dobradas/imunologia , Animais , Microambiente Celular/genética , Ciclo do Ácido Cítrico/genética , Ciclo do Ácido Cítrico/imunologia , Células Dendríticas/patologia , Hexoquinase/genética , Hexoquinase/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Resposta a Proteínas não Dobradas/genética , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/imunologia
2.
Immunity ; 54(1): 68-83.e6, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33238133

RESUMO

While antibiotics are intended to specifically target bacteria, most are known to affect host cell physiology. In addition, some antibiotic classes are reported as immunosuppressive for reasons that remain unclear. Here, we show that Linezolid, a ribosomal-targeting antibiotic (RAbo), effectively blocked the course of a T cell-mediated autoimmune disease. Linezolid and other RAbos were strong inhibitors of T helper-17 cell effector function in vitro, showing that this effect was independent of their antibiotic activity. Perturbing mitochondrial translation in differentiating T cells, either with RAbos or through the inhibition of mitochondrial elongation factor G1 (mEF-G1) progressively compromised the integrity of the electron transport chain. Ultimately, this led to deficient oxidative phosphorylation, diminishing nicotinamide adenine dinucleotide concentrations and impairing cytokine production in differentiating T cells. In accordance, mice lacking mEF-G1 in T cells were protected from experimental autoimmune encephalomyelitis, demonstrating that this pathway is crucial in maintaining T cell function and pathogenicity.


Assuntos
Antibacterianos/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Linezolida/uso terapêutico , Mitocôndrias/metabolismo , Peptídeos Cíclicos/uso terapêutico , Ribossomos/metabolismo , Células Th17/fisiologia , Animais , Autoimunidade/efeitos dos fármacos , Diferenciação Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Terapia de Alvo Molecular , Esclerose Múltipla/tratamento farmacológico , NAD/metabolismo , Fosforilação Oxidativa , Fator G para Elongação de Peptídeos/genética , Fator G para Elongação de Peptídeos/metabolismo
4.
Immunity ; 44(6): 1246-8, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27332726

RESUMO

Conventional dendritic cells (cDCs) and plasmacytoid dendritic cells (pDCs) serve non-overlapping functions in immune responses. In this issue of Immunity, Pearce and colleagues (2016) report that pDCs use different metabolic pathways from cDCs to support their specialized function.


Assuntos
Células Dendríticas/imunologia , Imunidade , Humanos , Vírus
5.
Eur J Immunol ; 53(2): e2149691, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36577714

RESUMO

The CD11c+ MHCII+ compartment within GM-CSF cultures consists of a MHCIIlow CD11bhigh population (GM-Macs) and a MHCIIhigh CD11bint population (GM-DCs), with different metabolic profiles. GM-Macs upregulate iNOS and produce nitric oxide (NO) upon TLR activation inhibiting mitochondrial respiration (OXPHOS) while promoting glycolytic metabolism in GM-DCs, which naturally do not express iNOS.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos , Óxido Nítrico , Camundongos , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Células Dendríticas/metabolismo , Diferenciação Celular , Camundongos Endogâmicos C57BL
6.
Eur J Immunol ; 53(11): e2249816, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-36303448

RESUMO

This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. This article provides protocols with top ticks and pitfalls for preparation and successful generation of mouse and human DC from different cellular sources, such as murine BM and HoxB8 cells, as well as human CD34+ cells from cord blood, BM, and peripheral blood or peripheral blood monocytes. We describe murine cDC1, cDC2, and pDC generation with Flt3L and the generation of BM-derived DC with GM-CSF. Protocols for human DC generation focus on CD34+ cell culture on OP9 cell layers for cDC1, cDC2, cDC3, and pDC subset generation and DC generation from peripheral blood monocytes (MoDC). Additional protocols include enrichment of murine DC subsets, CRISPR/Cas9 editing, and clinical grade human DC generation. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.


Assuntos
Células Dendríticas , Monócitos , Animais , Camundongos , Humanos , Antígenos CD34 , Fenótipo , Diferenciação Celular
7.
Eur J Immunol ; 2022 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-36563126

RESUMO

This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Recent studies have provided evidence for an increasing number of phenotypically distinct conventional DC (cDC) subsets that on one hand exhibit a certain functional plasticity, but on the other hand are characterized by their tissue- and context-dependent functional specialization. Here, we describe a selection of assays for the functional characterization of mouse and human cDC. The first two protocols illustrate analysis of cDC endocytosis and metabolism, followed by guidelines for transcriptomic and proteomic characterization of cDC populations. Then, a larger group of assays describes the characterization of cDC migration in vitro, ex vivo, and in vivo. The final guidelines measure cDC inflammasome and antigen (cross)-presentation activity. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.

8.
J Allergy Clin Immunol ; 148(1): 16-32, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33966898

RESUMO

The progression through different steps of T-cell development, activation, and effector function is tightly bound to specific cellular metabolic processes. Previous studies established that T-effector cells have a metabolic bias toward aerobic glycolysis, whereas naive and regulatory T cells mainly rely on oxidative phosphorylation. More recently, the field of immunometabolism has drifted away from the notion that mitochondrial metabolism holds little importance in T-cell activation and function. Of note, T cells possess metabolic promiscuity, which allows them to adapt their nutritional requirements according to the tissue environment. Altogether, the integration of these metabolic pathways culminates in the generation of not only energy but also intermediates, which can regulate epigenetic programs, leading to changes in T-cell fate. In this review, we discuss the recent literature on how glycolysis, amino acid catabolism, and fatty acid oxidation work together with the tricarboxylic acid cycle in the mitochondrion. We also emphasize the importance of the electron transport chain for T-cell immunity. We also discuss novel findings highlighting the role of key enzymes, accessory pathways, and posttranslational protein modifications that distinctively regulate T-cell function and might represent prominent candidates for therapeutic purposes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/imunologia , Ácidos Graxos/imunologia , Glicólise/imunologia , Mitocôndrias/imunologia , NAD/imunologia , Poliaminas/imunologia , Animais , Humanos
9.
Immunol Rev ; 283(1): 213-231, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29664569

RESUMO

CD8+ T cells are key members of the adaptive immune response against infections and cancer. As we discuss in this review, these cells can present diverse metabolic requirements, which have been intensely studied during the past few years. Our current understanding suggests that aerobic glycolysis is a hallmark of activated CD8+ T cells, while naive and memory (Tmem ) cells often rely on oxidative phosphorylation, and thus mitochondrial metabolism is a crucial determinant of CD8+ Tmem cell development. Moreover, it has been proposed that CD8+ Tmem cells have a specific requirement for the oxidation of long-chain fatty acids (LC-FAO), a process modulated in lymphocytes by the enzyme CPT1A. However, this notion relies heavily on the metabolic analysis of in vitro cultures and on chemical inhibition of CPT1A. Therefore, we introduce more recent studies using genetic models to demonstrate that CPT1A-mediated LC-FAO is dispensable for the development of CD8+ T cell memory and protective immunity, and question the use of chemical inhibitors to target this enzyme. We discuss insights obtained from those and other studies analyzing the metabolic characteristics of CD8+ Tmem cells, and emphasize how T cells exhibit flexibility in their choice of metabolic fuel.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Ácidos Graxos/metabolismo , Imunidade Celular , Metabolismo dos Lipídeos , Oxirredutases do Álcool/metabolismo , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Metabolismo Energético , Humanos , Ativação Linfocitária/imunologia , Mitocôndrias/metabolismo , Transdução de Sinais
10.
Semin Immunol ; 28(5): 514-524, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27825556

RESUMO

Recent advances in the field of immunometabolism support the concept that fundamental processes in T cell biology, such as TCR-mediated activation and T helper lineage differentiation, are closely linked to changes in the cellular metabolic programs. Although the major task of the intermediate metabolism is to provide the cell with a constant supply of energy and molecular precursors for the production of biomolecules, the dynamic regulation of metabolic pathways also plays an active role in shaping T cell responses. Key metabolic processes such as glycolysis, fatty acid and mitochondrial metabolism are now recognized as crucial players in T cell activation and differentiation, and their modulation can differentially affect the development of T helper cell lineages. In this review, we describe the diverse metabolic processes that T cells engage during their life cycle from naïve towards effector and memory T cells. We consider in particular how the cellular metabolism may actively support the function of T cells in their different states. Moreover, we discuss how molecular regulators such as mTOR or AMPK link environmental changes to adaptations in the cellular metabolism and elucidate the consequences on T cell differentiation and function.


Assuntos
Diferenciação Celular , Metabolismo Energético , Ativação Linfocitária/imunologia , Redes e Vias Metabólicas , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/fisiologia , Animais , Ácidos Graxos/metabolismo , Glicólise , Humanos , Memória Imunológica , Mitocôndrias/metabolismo , Fenótipo , Transdução de Sinais
11.
Proc Natl Acad Sci U S A ; 113(12): 3329-34, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26941238

RESUMO

Sialic acids are negatively charged nine-carbon carboxylated monosaccharides that often cap glycans on glycosylated proteins and lipids. Because of their strategic location at the cell surface, sialic acids contribute to interactions that are critical for immune homeostasis via interactions with sialic acid-binding Ig-type lectins (siglecs). In particular, these interactions may be of importance in cases where sialic acids may be overexpressed, such as on certain pathogens and tumors. We now demonstrate that modification of antigens with sialic acids (Sia-antigens) regulates the generation of antigen-specific regulatory T (Treg) cells via dendritic cells (DCs). Additionally, DCs that take up Sia-antigen prevent formation of effector CD4(+) and CD8(+)T cells. Importantly, the regulatory properties endowed on DCs upon Sia-antigen uptake are antigen-specific: only T cells responsive to the sialylated antigen become tolerized. In vivo, injection of Sia-antigen-loaded DCs increased de novo Treg-cell numbers and dampened effector T-cell expansion and IFN-γ production. The dual tolerogenic features that Sia-antigen imposed on DCs are Siglec-E-mediated and maintained under inflammatory conditions. Moreover, loading DCs with Sia-antigens not only inhibited the function of in vitro-established Th1 and Th17 effector T cells but also significantly dampened ex vivo myelin-reactive T cells, present in the circulation of mice with experimental autoimmune encephalomyelitis. These data indicate that sialic acid-modified antigens instruct DCs in an antigen-specific tolerogenic programming, enhancing Treg cells and reducing the generation and propagation of inflammatory T cells. Our data suggest that sialylation of antigens provides an attractive way to induce antigen-specific immune tolerance.


Assuntos
Adaptação Fisiológica/imunologia , Antígenos/imunologia , Proliferação de Células , Ácido N-Acetilneuramínico/química , Linfócitos T Reguladores/imunologia , Animais , Antígenos/química , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/citologia
12.
Infect Immun ; 86(6)2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29555679

RESUMO

Giardiasis is one of the most common human intestinal diseases worldwide. Several experimental animal models have been used to evaluate Giardia infections, with gerbils (Meriones unguiculatus) being the most valuable model due to their high susceptibility to Giardia infection, abundant shedding of cysts, and pathophysiological alterations and signs of disease similar to those observed in humans. Here, we report cytokine and antibody profiles both during the course of Giardia infection in gerbils and after immunization with a novel oral vaccine comprising a mixture of purified variant-specific surface proteins (VSPs). Transcript levels of representative cytokines of different immune profiles as well as macro- and microtissue alterations were assessed in Peyer's patches, mesenteric lymph nodes, and spleens. During infection, cytokine responses showed a biphasic profile: an early induction of Th1 (gamma interferon [IFN-γ], interleukin-1ß [IL-1ß], IL-6, and tumor necrosis factor [TNF]), Th17 (IL-17), and Th2 (IL-4) cytokines, together with intestinal alterations typical of inflammation, followed by a shift toward a predominant Th2 (IL-5) response, likely associated with a counterregulatory mechanism. Conversely, immunization with an oral vaccine comprising the entire repertoire of VSPs specifically showed high levels of IL-17, IL-6, IL-4, and IL-5, without obvious signs of inflammation. Both immunized and infected animals developed local (intestinal secretory IgA [S-IgA]) and systemic (serum IgG) humoral immune responses against VSPs; however, only infected animals showed evident signs of giardiasis. This is the first comprehensive report of cytokine expression and anti-Giardia antibody production during infection and VSP vaccination in gerbils, a reliable model of the human disease.


Assuntos
Giardia lamblia/genética , Giardíase/prevenção & controle , Proteínas de Membrana/genética , Vacinas Protozoárias/imunologia , Animais , Feminino , Gerbillinae , Giardíase/parasitologia , Humanos , Masculino , Proteínas de Membrana/imunologia , Organismos Geneticamente Modificados , Organismos Livres de Patógenos Específicos , Vacinação
13.
Trends Immunol ; 36(2): 81-91, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25592731

RESUMO

The specific regulation of cellular metabolic processes is of major importance for directing immune cell differentiation and function. We review recent evidence indicating that changes in basic cellular lipid metabolism have critical effects on T cell proliferation and cell fate decisions. While induction of de novo fatty acid (FA) synthesis is essential for activation-induced proliferation and differentiation of effector T cells, FA catabolism via ß-oxidation is important for the development of CD8(+) T cell memory as well as for the differentiation of CD4(+) regulatory T cells. We consider the influence of lipid metabolism and metabolic intermediates on the regulation of signaling and transcriptional pathways via post-translational modifications, and discuss how an improved understanding of FA metabolism may reveal strategies for manipulating immune responses towards therapeutic outcomes.


Assuntos
Ácidos Graxos/metabolismo , Imunomodulação , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Humanos , Memória Imunológica , Lipídeos/biossíntese , Ativação Linfocitária , Oxirredução , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Serina-Treonina Quinases TOR/metabolismo
14.
Eur J Immunol ; 46(9): 2233-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27338930

RESUMO

Upon antigen-specific or allogeneic activation, T cells sharply increase their metabolic activity to cope with augmented needs for proliferation and effector functions. Therefore, enzymes involved in energy metabolism constitute attractive targets to modulate the activity of pathogenic effector T cells in the setting of graft-versus-host-disease (GVHD). Here, we show that T cells deficient for acetyl-CoA carboxylase 1 (TACC1) are dramatically less pathogenic than wild-type (WT) T cells in a lethal C57BL/6 into BALB/c model of acute GVHD and permitted sustained survival of recipient mice. In line with this clinical observation, higher frequencies of GVHD-suppressing Foxp3(+) regulatory T (Treg) cells were detected in the colon of TACC T-cell recipients. In vitro, T-cell stimulation with allogeneic DCs induced higher proportions of Treg cells but also led to diminished proliferation of TACC1 T cells compared to WT T cells. Furthermore, TACC1 T cells activated by allogeneic DCs showed impaired glycolysis and lipid synthesis. Thus, targeting de novo fatty acid synthesis via acetyl-CoA carboxylase inhibition may be a promising new strategy to prevent GVHD.


Assuntos
Acetil-CoA Carboxilase/antagonistas & inibidores , Ácidos Graxos/biossíntese , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/prevenção & controle , Acetil-CoA Carboxilase/deficiência , Transferência Adotiva , Animais , Biomarcadores , Transplante de Medula Óssea , Diferenciação Celular , Modelos Animais de Doenças , Deleção de Genes , Doença Enxerto-Hospedeiro/mortalidade , Imunofenotipagem , Macrolídeos/farmacologia , Masculino , Camundongos , Fenótipo , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Homólogo
15.
Eur J Immunol ; 45(1): 32-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25403892

RESUMO

Dendritic cells (DCs) are key regulators of both innate and adaptive immunity. During infection, DCs recognise pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs) including the Toll-like receptor (TLR) family. TLRs mainly signal via the adaptor protein MyD88. This signalling pathway is required for immune protection during many infections, which are lethal in the absence of MyD88. However, the cell type specific importance of this pathway during both innate and adaptive immune responses against pathogens in vivo remains ill-defined. We discuss recent findings from conditional KO or gain-of-function mouse models targeting TLR/MyD88 signalling pathways in DCs and other myeloid cells during infection. While the general assumption that MyD88-dependent recognition by DCs is essential for inducing protective immunity holds true in some instances, the results surprisingly indicate a much more complex context-dependent requirement for this pathway in DCs and other myeloid or lymphoid cell-types in vivo. Furthermore, we highlight the advantages of Cre-mediated DC targeting approaches and their possible limitations. We also present future perspectives on the development of new genetic mouse models to target distinct DC subsets in vivo. Such models will serve to understand the functional heterogeneity of DCs in vivo.


Assuntos
Candidíase/imunologia , Células Dendríticas/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Toxoplasmose/imunologia , Imunidade Adaptativa , Animais , Candida albicans/imunologia , Candidíase/genética , Candidíase/microbiologia , Células Dendríticas/microbiologia , Células Dendríticas/parasitologia , Regulação da Expressão Gênica , Imunidade Inata , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/genética , Regiões Promotoras Genéticas , Transdução de Sinais/genética , Receptores Toll-Like/genética , Toxoplasma/imunologia , Toxoplasmose/genética , Toxoplasmose/parasitologia
16.
Immunol Cell Biol ; 94(10): 925-934, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27562063

RESUMO

A continuous increase in the prevalence of autoimmune diseases is to be expected in the aging societies worldwide. Autoimmune disorders not only cause severe disability and chronic pain, but also lead to considerable socio-economic costs. Given that the current treatment options are not curative, have substantial side effects and a high percentage of non-responders, innovative options to the existing therapeutic armament against autoimmune diseases are urgently required. Accumulating evidence suggests that changes in the metabolism of immune cells are associated with, and contribute to the pathogenesis of autoimmunity. Additionally, some autoimmune diseases share alterations in metabolic pathways, key metabolites or metabolic byproducts such as reactive oxygen species. Other examples for metabolic changes in autoimmune settings include modifications in amino acid and cholesterol levels or glucose catabolism. Thus, the emerging field of immunometabolism may hold the potential to discover new therapeutic targets. Here, we discuss recent findings describing metabolic changes in autoimmune arthritis, multiple sclerosis as well as type 1 diabetes, focusing on pathophysiological aspects.


Assuntos
Autoimunidade , Metabolismo/imunologia , Animais , Humanos , Redes e Vias Metabólicas , Modelos Biológicos
17.
Blood ; 124(20): 3081-91, 2014 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25100743

RESUMO

Multiple subsets of FMS-like tyrosine kinase 3 ligand (FLT3L)-dependent dendritic cells (DCs) control T-cell tolerance and immunity. In mice, Batf3-dependent CD103(+) DCs efficiently enter lymph nodes and cross-present antigens, rendering this conserved DC subset a promising target for tolerance induction or vaccination. However, only limited numbers of CD103(+) DCs can be isolated with current methods. Established bone marrow culture protocols efficiently generate monocyte-derived DCs or produce a mixture of FLT3L-dependent DC subsets. We show that CD103(+) DC development requires prolonged culture time and continuous action of both FLT3L and granulocyte macrophage colony-stimulating factor (GM-CSF), explained by a dual effect of GM-CSF on DC precursors and differentiating CD103(+) DCs. Accordingly, we established a novel method to generate large numbers of CD103(+) DCs (iCD103-DCs) with limited presence of other DC subsets. iCD103-DCs develop in a Batf3- and Irf8-dependent fashion, express a CD8α/CD103 DC gene signature, cross-present cell-associated antigens, and respond to TLR3 stimulation. Thus, iCD103-DCs reflect key features of tissue CD103(+) DCs. Importantly, iCD103-DCs express high levels of CCR7 upon maturation and migrate to lymph nodes more efficiently than classical monocyte-derived DCs. Finally, iCD103-DCs induce T cell-mediated protective immunity in vivo. Our study provides insights into CD103(+) DC development and function.


Assuntos
Antígenos CD/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Células Dendríticas/citologia , Células Dendríticas/imunologia , Cadeias alfa de Integrinas/imunologia , Proteínas Repressoras/imunologia , Animais , Antígenos CD/análise , Fatores de Transcrição de Zíper de Leucina Básica/análise , Diferenciação Celular , Células Cultivadas , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Imunidade Celular , Cadeias alfa de Integrinas/análise , Proteínas de Membrana/imunologia , Camundongos , Proteínas Repressoras/análise , Linfócitos T/imunologia , Receptor 3 Toll-Like/imunologia
18.
Eur J Immunol ; 44(5): 1399-409, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24435955

RESUMO

Tuberculosis is a chronic infectious disease caused by Mycobacterium tuberculosis that is responsible for almost 1.5 million deaths per year. Sensing of mycobacteria by the host's immune system relies on different families of receptors present on innate immune cells. Amongst them, several members of the TLR family are involved in the activation of immune cells by mycobacteria, yet the in vivo contribution of individual TLRs to the protective immune response remains controversial. On the contrary, MyD88, the adaptor molecule for most TLRs, plays a non-redundant role in the protection against tuberculosis and mice with a complete germline deletion of MyD88 succumb very early to infection. MyD88 is expressed in both immune and non-immune cells, but it is not clear whether control of mycobacteria requires ubiquitous or cell-type specific MyD88 expression. Therefore, using novel conditional switch-on mouse models, we aimed to investigate the importance of MyD88 signalling in DCs and macrophages for the induction of protective effector mechanisms against mycobacterial infection. We conclude that specific reactivation of MyD88 signalling in CD11c- or lysozyme M-expressing myeloid cells during Mycobacterium bovis Bacille Calmette-Guerin infection is sufficient to restore systemic and local inflammatory cytokine production and to control pathogen burden.


Assuntos
Citocinas/imunologia , Macrófagos/imunologia , Mycobacterium bovis/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia , Tuberculose/imunologia , Animais , Antígeno CD11c/biossíntese , Antígeno CD11c/genética , Antígeno CD11c/imunologia , Doença Crônica , Citocinas/biossíntese , Citocinas/genética , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Modelos Animais de Doenças , Deleção de Genes , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Muramidase/biossíntese , Muramidase/genética , Muramidase/imunologia , Mycobacterium bovis/metabolismo , Fator 88 de Diferenciação Mieloide/biossíntese , Fator 88 de Diferenciação Mieloide/genética , Transdução de Sinais/genética , Tuberculose/genética , Tuberculose/metabolismo , Tuberculose/patologia , Tuberculose/prevenção & controle , Tuberculose/veterinária
19.
Eur J Immunol ; 44(10): 2990-3002, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25042334

RESUMO

Foxp3 specifies the Treg cell lineage and is indispensable for immune tolerance. Accordingly, rare Foxp3 mutations cause lethal autoimmunity. The mechanisms precipitating more prevalent human autoimmune diseases are poorly understood, but involve a combination of genetic and environmental factors. Many autoimmune diseases associate with a partial Treg-cell dysfunction, yet mouse models reflecting such complex pathophysiological processes are rare. Around 95% of Foxp3(+) Treg cells can be specifically depleted in bacterial artifical chromosome (BAC)-transgenic Depletion of REGulatory T cells (DEREG) mice through diphtheria toxin (DT) treatment. However, Treg-cell depletion fails to cause autoimmunity in adult DEREG mice for unclear reasons. By crossing Foxp3(GFP) knock-in mice to DEREG mice, we introduced additional genetic susceptibility that does not affect untreated mice. Strikingly, DT treatment of DEREG × Foxp3(GFP) mice rapidly causes autoimmunity characterized by blepharitis, tissue damage, and autoantibody production. This inflammatory disease is associated with augmented T-cell activation, increased Th2 cytokine production and myeloproliferation, and is caused by defective Treg-cell homeostasis, preventing few DT-insensitive Treg cells from repopulating the niche after Treg-cell depletion. Our study provides important insights into self-tolerance. We further highlight DEREG × Foxp3(GFP) mice as a model to investigate the role of environmental factors in precipitating autoimmunity. This may help to better understand and treat human autoimmunity.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Ativação Linfocitária/imunologia , Linfócitos T Reguladores/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Técnicas de Introdução de Genes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos
20.
Eur J Immunol ; 43(10): 2543-53, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23784881

RESUMO

Vaccination is one of the oldest yet still most effective methods to prevent infectious diseases. However, eradication of intracellular pathogens and treatment of certain diseases like cancer requiring efficient cytotoxic immune responses remain a medical challenge. In mice, a successful approach to induce strong cytotoxic CD8⁺ T-cell (CTL) reactions is to target antigens to DCs using specific antibodies against surface receptors in combination with adjuvants. A major drawback for translating this strategy into one for the clinic is the lack of analogous targets in human DCs. DC-SIGN (DC-specific-ICAM3-grabbing-nonintegrin/CD209) is a C-type lectin receptor with potent endocytic capacity and a highly restricted expression on human immature DCs. Therefore, DC-SIGN represents an ideal candidate for DC targeting. Using transgenic mice that express human DC-SIGN under the control of the murine CD11c promoter (hSIGN mice), we explored the efficacy of anti-DC-SIGN antibodies to target antigens to DCs and induce protective immune responses in vivo. We show that anti-DC-SIGN antibodies conjugated to OVA induced strong and persistent antigen-specific CD4⁺ and CD8⁺ T-cell responses, which efficiently protected from infection with OVA-expressing Listeria monocytogenes. Thus, we propose DC targeting via DC-SIGN as a promising strategy for novel vaccination protocols against intracellular pathogens.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/imunologia , Lectinas Tipo C/metabolismo , Listeria monocytogenes/imunologia , Receptores de Superfície Celular/metabolismo , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/metabolismo , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Antígeno CD11c/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Humanos , Imunidade Ativa , Imunidade Celular , Imunomodulação , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Listeria monocytogenes/genética , Camundongos , Camundongos Transgênicos , Ovalbumina/genética , Ovalbumina/metabolismo , Regiões Promotoras Genéticas/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Transgenes/genética , Vacinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA