Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 23(11): 1536-1550, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36271147

RESUMO

CD40 signaling in classical type 1 dendritic cells (cDC1s) is required for CD8 T cell-mediated tumor rejection, but the underlying mechanisms are incompletely understood. Here, we identified CD40-induced genes in cDC1s, including Cd70, Tnfsf9, Ptgs2 and Bcl2l1, and examined their contributions to anti-tumor immunity. cDC1-specific inactivation of CD70 and COX-2, and global CD27 inactivation, only partially impaired tumor rejection or tumor-specific CD8 T cell expansion. Loss of 4-1BB, alone or in Cd27-/- mice, did not further impair anti-tumor immunity. However, cDC1-specific CD40 inactivation reduced cDC1 mitochondrial transmembrane potential and increased caspase activation in tumor-draining lymph nodes, reducing migratory cDC1 numbers in vivo. Similar impairments occurred during in vitro antigen presentation by Cd40-/- cDC1s to CD8+ T cells, which were reversed by re-expression of Bcl2l1. Thus, CD40 signaling in cDC1s not only induces costimulatory ligands for CD8+ T cells but also induces Bcl2l1 that sustains cDC1 survival during priming of anti-tumor responses.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Camundongos , Animais , Antígenos CD40/genética , Apresentação de Antígeno , Células Dendríticas , Camundongos Endogâmicos C57BL
2.
Immunity ; 55(6): 1032-1050.e14, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35704993

RESUMO

Conventional dendritic cells (cDCs), cDC1 and cDC2, act both to initiate immunity and maintain self-tolerance. The tryptophan metabolic enzyme indoleamine 2,3-dioxygenase 1 (IDO1) is used by cDCs in maintaining tolerance, but its role in different subsets remains unclear. At homeostasis, only mature CCR7+ cDC1 expressed IDO1 that was dependent on IRF8. Lipopolysaccharide treatment induced maturation and IDO1-dependent tolerogenic activity in isolated immature cDC1, but not isolated cDC2. However, both human and mouse cDC2 could induce IDO1 and acquire tolerogenic function when co-cultured with mature cDC1 through the action of cDC1-derived l-kynurenine. Accordingly, cDC1-specific inactivation of IDO1 in vivo exacerbated disease in experimental autoimmune encephalomyelitis. This study identifies a previously unrecognized metabolic communication in which IDO1-expressing cDC1 cells extend their immunoregulatory capacity to the cDC2 subset through their production of tryptophan metabolite l-kynurenine. This metabolic axis represents a potential therapeutic target in treating autoimmune demyelinating diseases.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase , Cinurenina , Animais , Células Dendríticas , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Cinurenina/metabolismo , Camundongos , Transdução de Sinais , Triptofano/metabolismo
3.
Immunity ; 53(4): 759-774.e9, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32795402

RESUMO

Development and function of conventional dendritic cell (cDC) subsets, cDC1 and cDC2, depend on transcription factors (TFs) IRF8 and IRF4, respectively. Since IRF8 and IRF4 can each interact with TF BATF3 at AP1-IRF composite elements (AICEs) and with TF PU.1 at Ets-IRF composite elements (EICEs), it is unclear how these factors exert divergent actions. Here, we determined the basis for distinct effects of IRF8 and IRF4 in cDC development. Genes expressed commonly by cDC1 and cDC2 used EICE-dependent enhancers that were redundantly activated by low amounts of either IRF4 or IRF8. By contrast, cDC1-specific genes relied on AICE-dependent enhancers, which required high IRF concentrations, but were activated by either IRF4 or IRF8. IRF8 was specifically required only by a minority of cDC1-specific genes, such as Xcr1, which could distinguish between IRF8 and IRF4 DNA-binding domains. Thus, these results explain how BATF3-dependent Irf8 autoactivation underlies emergence of the cDC1-specific transcriptional program.


Assuntos
Células Dendríticas/metabolismo , Elementos Facilitadores Genéticos/genética , Fatores Reguladores de Interferon/genética , Animais , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Quimiocinas/genética , Transcrição Gênica/genética
4.
Nature ; 584(7822): 624-629, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32788723

RESUMO

Conventional type 1 dendritic cells (cDC1)1 are thought to perform antigen cross-presentation, which is required to prime CD8+ T cells2,3, whereas cDC2 are specialized for priming CD4+ T cells4,5. CD4+ T cells are also considered to help CD8+ T cell responses through a variety of mechanisms6-11, including a process whereby CD4+ T cells 'license' cDC1 for CD8+ T cell priming12. However, this model has not been directly tested in vivo or in the setting of help-dependent tumour rejection. Here we generated an Xcr1Cre mouse strain to evaluate the cellular interactions that mediate tumour rejection in a model requiring CD4+ and CD8+ T cells. As expected, tumour rejection required cDC1 and CD8+ T cell priming required the expression of major histocompatibility class I molecules by cDC1. Unexpectedly, early priming of CD4+ T cells against tumour-derived antigens also required cDC1, and this was not simply because they transport antigens to lymph nodes for processing by cDC2, as selective deletion of major histocompatibility class II molecules in cDC1 also prevented early CD4+ T cell priming. Furthermore, deletion of either major histocompatibility class II or CD40 in cDC1 impaired tumour rejection, consistent with a role for cognate CD4+ T cell interactions and CD40 signalling in cDC1 licensing. Finally, CD40 signalling in cDC1 was critical not only for CD8+ T cell priming, but also for initial CD4+ T cell activation. Thus, in the setting of tumour-derived antigens, cDC1 function as an autonomous platform capable of antigen processing and priming for both CD4+ and CD8+ T cells and of the direct orchestration of their cross-talk that is required for optimal anti-tumour immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Apresentação Cruzada , Células Dendríticas/imunologia , Neoplasias/imunologia , Animais , Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/citologia , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Antígenos de Histocompatibilidade Classe II/imunologia , Camundongos , Transdução de Sinais
6.
Proc Natl Acad Sci U S A ; 115(42): 10726-10731, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30279176

RESUMO

CD4+ T follicular helper (TFH) cells support germinal center (GC) reactions promoting humoral immunity. Dendritic cell (DC) diversification into genetically distinct subsets allows for specialization in promoting responses against several types of pathogens. Whether any classical DC (cDC) subset is required for humoral immunity is unknown, however. We tested several genetic models that selectively ablate distinct DC subsets in mice for their impact on splenic GC reactions. We identified a requirement for Notch2-dependent cDC2s, but not Batf3-dependent cDC1s or Klf4-dependent cDC2s, in promoting TFH and GC B cell formation in response to sheep red blood cells and inactivated Listeria monocytogenes This effect was mediated independent of Il2ra and several Notch2-dependent genes expressed in cDC2s, including Stat4 and Havcr2 Notch2 signaling during cDC2 development also substantially reduced the efficiency of cDC2s for presentation of MHC class II-restricted antigens, limiting the strength of CD4 T cell activation. Together, these results demonstrate a nonredundant role for the Notch2-dependent cDC2 subset in supporting humoral immune responses.


Assuntos
Linfócitos B/imunologia , Células Dendríticas/imunologia , Eritrócitos/imunologia , Centro Germinativo/imunologia , Receptor Notch2/fisiologia , Baço/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Apresentação de Antígeno/imunologia , Linfócitos B/metabolismo , Diferenciação Celular , Células Cultivadas , Células Dendríticas/metabolismo , Centro Germinativo/metabolismo , Imunidade Humoral/imunologia , Fator 4 Semelhante a Kruppel , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovinos , Transdução de Sinais , Baço/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(15): 3957-3962, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28348230

RESUMO

RelB is an NF-κB family transcription factor activated in the noncanonical pathway downstream of NF-κB-inducing kinase (NIK) and TNF receptor family members including lymphotoxin-ß receptor (LTßR) and CD40. Early analysis suggested that RelB is required for classical dendritic cell (cDC) development based on a severe reduction of cDCs in Relb-/- mice associated with profound myeloid expansion and perturbations in B and T cells. Subsequent analysis of radiation chimeras generated from wild-type and Relb-/- bone marrow showed that RelB exerts cell-extrinsic actions on some lineages, but it has remained unclear whether the impact of RelB on cDC development is cell-intrinsic or -extrinsic. Here, we reevaluated the role of RelB in cDC and myeloid development using a series of radiation chimeras. We found that there was no cell-intrinsic requirement for RelB for development of most cDC subsets, except for the Notch2- and LTßR-dependent subset of splenic CD4+ cDC2s. These results identify a relatively restricted role of RelB in DC development. Moreover, the myeloid expansion in Relb-/- mice resulted from hematopoietic-extrinsic actions of RelB. This result suggests that there is an unrecognized but critical role for RelB within the nonhematopoietic niche that controls normal myelopoiesis.


Assuntos
Células Dendríticas/fisiologia , Células Mieloides/fisiologia , Fator de Transcrição RelB/genética , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Sistema Hematopoético/citologia , Sistema Hematopoético/metabolismo , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-beta/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Serina-Treonina Quinases/metabolismo , Baço/citologia , Baço/metabolismo , Fator de Transcrição RelB/metabolismo , Quinase Induzida por NF-kappaB
8.
J Am Soc Nephrol ; 29(1): 138-154, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29217759

RESUMO

Dendritic cells (DCs) are thought to form a dendritic network across barrier surfaces and throughout organs, including the kidney, to perform an important sentinel function. However, previous studies of DC function used markers, such as CD11c or CX3CR1, that are not unique to DCs. Here, we evaluated the role of DCs in renal inflammation using a CD11c reporter mouse line and two mouse lines with DC-specific reporters, Zbtb46-GFP and Snx22-GFP. Multiphoton microscopy of kidney sections confirmed that most of the dendritically shaped CD11c+ cells forming a network throughout the renal interstitium expressed macrophage-specific markers. In contrast, DCs marked by Zbtb46-GFP or Snx22-GFP were less abundant, concentrated around blood vessels, and round in shape. We confirmed this pattern of localization using imaging mass cytometry. Motility measurements showed that resident macrophages were sessile, whereas DCs were motile before and after inflammation. Although uninflamed glomeruli rarely contained DCs, injury with nephrotoxic antibodies resulted in accumulation of ZBTB46 + cells in the periglomerular region. ZBTB46 identifies all classic DCs, which can be categorized into two functional subsets that express either CD103 or CD11b. Depletion of ZBTB46 + cells attenuated the antibody-induced kidney injury, whereas deficiency of the CD103+ subset accelerated injury through a mechanism that involved increased neutrophil infiltration. RNA sequencing 7 days after nephrotoxic antibody injection showed that CD11b+ DCs expressed the neutrophil-attracting cytokine CXCL2, whereas CD103+ DCs expressed high levels of several anti-inflammatory genes. These results provide new insights into the distinct functions of the two major DC subsets in glomerular inflammation.


Assuntos
Células Dendríticas/fisiologia , Glomerulonefrite/imunologia , Glomerulonefrite/patologia , Animais , Antígenos CD/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Antígenos CD11/genética , Antígeno CD11b/genética , Movimento Celular , Quimiocina CXCL2/genética , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Cadeias alfa de Integrinas/metabolismo , Macrófagos , Masculino , Camundongos , Camundongos Knockout , Neutrófilos/patologia , Neutrófilos/fisiologia , Proteínas Repressoras/genética , Análise de Sequência de RNA , Nexinas de Classificação/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma
9.
Cell Rep ; 33(7): 108381, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33207188

RESUMO

Central to anti-tumor immunity are dendritic cells (DCs), which stimulate long-lived protective T cell responses. Recent studies have demonstrated that DCs can achieve a state of hyperactivation, which is associated with inflammasome activities within living cells. Herein, we report that hyperactive DCs have an enhanced ability to migrate to draining lymph nodes and stimulate potent cytotoxic T lymphocyte (CTL) responses. This enhanced migratory activity is dependent on the chemokine receptor CCR7 and is associated with a unique transcriptional program that is not observed in conventionally activated or pyroptotic DCs. We show that hyperactivating stimuli are uniquely capable of inducing durable CTL-mediated anti-tumor immunity against tumors that are sensitive or resistant to PD-1 inhibition. These protective responses are intrinsic to the cDC1 subset of DCs, depend on the inflammasome-dependent cytokine IL-1ß, and enable tumor lysates to serve as immunogens. If these activities are verified in humans, hyperactive DCs may impact immunotherapy.


Assuntos
Imunidade Adaptativa/imunologia , Células Dendríticas/imunologia , Inflamassomos/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Humanos , Imunoterapia , Linfonodos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores CCR7/imunologia , Receptores CCR7/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
10.
Cancer Immunol Res ; 7(1): 29-39, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30482745

RESUMO

The BATF3-dependent cDC1 lineage of conventional dendritic cells (cDC) is required for rejection of immunogenic sarcomas and for rejection of progressive sarcomas during checkpoint blockade therapy. One unique function of the cDC1 lineage is the efficient cross-presentation of tumor-derived neoantigens to CD8+ T cells, but it is not clear that this is the only unique function of cDC1 required for tumor rejection. We previously showed that BATF3 functions during cDC1 lineage commitment to maintain IRF8 expression in the specified cDC1 progenitor. However, since cDC1 progenitors do not develop into mature cDC1s in Batf3 -/- mice, it is still unclear whether BATF3 has additional functions in mature cDC1 cells. A transgenic Irf8-Venus reporter allele increases IRF8 protein concentration sufficiently to allow autonomous cDC1 development in spleens of Batf3 -/- mice. These restored Batf3 -/- cDC1s are transcriptionally similar to control wild-type cDC1s but have reduced expression of a restricted set of cDC1-specific genes. Restored Batf3 -/- cDC1s are able to cross-present cell-associated antigens both in vitro and in vivo However, Batf3 -/- cDC1 exhibit altered characteristics in vivo and are unable to mediate tumor rejection. These results show that BATF3, in addition to regulating Irf8 expression to stabilize cDC1 lineage commitment, also controls expression of a small set of genes required for cDC1-mediated tumor rejection. These BATF3-regulated genes may be useful targets in immunotherapies aimed at promoting tumor rejection.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Células Dendríticas/imunologia , Fibrossarcoma/genética , Rejeição de Enxerto/genética , Fatores Reguladores de Interferon/genética , Proteínas Repressoras/genética , Animais , Linhagem Celular Tumoral , Feminino , Fibrossarcoma/imunologia , Rejeição de Enxerto/imunologia , Linfonodos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Baço/imunologia
11.
J Exp Med ; 215(5): 1417-1435, 2018 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-29572360

RESUMO

The receptor Flt3 and its ligand Flt3L are both critical for dendritic cell (DC) development, but DC deficiency is more severe in Flt3l-/- mice than in Flt3-/- mice. This has led to speculation that Flt3L binds to another receptor that also supports DC development. However, we found that Flt3L administration does not generate DCs in Flt3-/- mice, arguing against a second receptor. Instead, Flt3-/- DC progenitors matured in response to macrophage colony-stimulating factor (M-CSF) or stem cell factor, and deletion of Csf1r in Flt3-/- mice further reduced DC development, indicating that these cytokines could compensate for Flt3. Surprisingly, Flt3-/- DC progenitors displayed enhanced M-CSF signaling, suggesting that loss of Flt3 increased responsiveness to other cytokines. In agreement, deletion of Flt3 in Flt3l-/- mice paradoxically rescued their severe DC deficiency. Thus, multiple cytokines can support DC development, and the discrepancy between Flt3-/- and Flt3l-/- mice results from the increased sensitivity of Flt3-/- progenitors to these cytokines.


Assuntos
Citocinas/metabolismo , Proteínas de Membrana/deficiência , Transdução de Sinais , Tirosina Quinase 3 Semelhante a fms/deficiência , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Deleção de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Células-Tronco/farmacologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Tirosina Quinase 3 Semelhante a fms/metabolismo
12.
Science ; 362(6415): 694-699, 2018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30409884

RESUMO

During the process of cross-presentation, viral or tumor-derived antigens are presented to CD8+ T cells by Batf3-dependent CD8α+/XCR1+ classical dendritic cells (cDC1s). We designed a functional CRISPR screen for previously unknown regulators of cross-presentation, and identified the BEACH domain-containing protein WDFY4 as essential for cross-presentation of cell-associated antigens by cDC1s in mice. However, WDFY4 was not required for major histocompatibility complex class II presentation, nor for cross-presentation by monocyte-derived dendritic cells. In contrast to Batf3 -/- mice, Wdfy4 -/- mice displayed normal lymphoid and nonlymphoid cDC1 populations that produce interleukin-12 and protect against Toxoplasma gondii infection. However, similar to Batf3 -/- mice, Wdfy4 -/- mice failed to prime virus-specific CD8+ T cells in vivo or induce tumor rejection, revealing a critical role for cross-presentation in antiviral and antitumor immunity.


Assuntos
Antígenos de Neoplasias/imunologia , Antígenos Virais/imunologia , Apresentação Cruzada/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Linfócitos T CD8-Positivos/imunologia , Sistemas CRISPR-Cas , Testes Genéticos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Toxoplasma/imunologia , Toxoplasmose/imunologia
13.
Nat Commun ; 9(1): 1828, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29739946

RESUMO

NOTCH signaling is required for the arterial specification and formation of hematopoietic stem cells (HSCs) and lympho-myeloid progenitors in the embryonic aorta-gonad-mesonephros region and extraembryonic vasculature from a distinct lineage of vascular endothelial cells with hemogenic potential. However, the role of NOTCH signaling in hemogenic endothelium (HE) specification from human pluripotent stem cell (hPSC) has not been studied. Here, using a chemically defined hPSC differentiation system combined with the use of DLL1-Fc and DAPT to manipulate NOTCH, we discover that NOTCH activation in hPSC-derived immature HE progenitors leads to formation of CD144+CD43-CD73-DLL4+Runx1 + 23-GFP+ arterial-type HE, which requires NOTCH signaling to undergo endothelial-to-hematopoietic transition and produce definitive lympho-myeloid and erythroid cells. These findings demonstrate that NOTCH-mediated arterialization of HE is an essential prerequisite for establishing definitive lympho-myeloid program and suggest that exploring molecular pathways that lead to arterial specification may aid in vitro approaches to enhance definitive hematopoiesis from hPSCs.


Assuntos
Artérias/citologia , Endotélio Vascular/citologia , Hemangioblastos/citologia , Hematopoese , Neovascularização Fisiológica , Células-Tronco Pluripotentes/citologia , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Antígenos CD/imunologia , Artérias/metabolismo , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Rastreamento de Células/instrumentação , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Endotélio Vascular/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/imunologia , Hemangioblastos/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/imunologia , Células-Tronco Pluripotentes/imunologia
14.
Cell Rep ; 22(13): 3440-3453.e6, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590614

RESUMO

Although the outcome of flavivirus infection can vary from asymptomatic to lethal, environmental factors modulating disease severity are poorly defined. Here, we observed increased susceptibility of mice to severe West Nile (WNV), Dengue, and Zika virus infections after treatment with oral antibiotics (Abx) that depleted the gut microbiota. Abx treatment impaired the development of optimal T cell responses, with decreased levels of WNV-specific CD8+ T cells associated with increased infection and immunopathology. Abx treatments that resulted in enhanced WNV susceptibility generated changes in the overall structure of the gut bacterial community and in the abundance of specific bacterial taxa. As little as 3 days of treatment with ampicillin was sufficient to alter host immunity and WNV outcome. Our results identify oral Abx therapy as a potential environmental determinant of systemic viral disease, and they raise the possibility that perturbation of the gut microbiota may have deleterious consequences for subsequent flavivirus infections.


Assuntos
Antibacterianos/efeitos adversos , Flavivirus/isolamento & purificação , Infecção por Zika virus/tratamento farmacológico , Administração Oral , Aedes , Ampicilina/efeitos adversos , Ampicilina/farmacologia , Animais , Antibacterianos/farmacologia , Ceco/efeitos dos fármacos , Ceco/microbiologia , Chlorocebus aethiops , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Células Vero , Infecção por Zika virus/imunologia , Infecção por Zika virus/microbiologia , Infecção por Zika virus/patologia
15.
Cell Rep ; 15(11): 2462-74, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27264183

RESUMO

Both classical DCs (cDCs) and monocyte-derived DCs (Mo-DCs) are capable of cross-priming CD8(+) T cells in response to cell-associated antigens. We found that Ly-6C(hi)TREML4(-) monocytes can differentiate into Zbtb46(+) Mo-DCs in response to granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) but that Ly-6C(hi)TREML4(+) monocytes were committed to differentiate into Ly-6C(lo)TREML4(+) monocytes. Differentiation of Zbtb46(+) Mo-DCs capable of efficient cross-priming required both GM-CSF and IL-4 and was accompanied by the induction of Batf3 and Irf4. However, monocytes require IRF4, but not BATF3, to differentiate into Zbtb46(+) Mo-DCs capable of cross-priming CD8(+) T cells. Instead, Irf4(-/-) monocytes differentiate into macrophages in response to GM-CSF and IL-4. Thus, cDCs and Mo-DCs require distinct transcriptional programs of differentiation in acquiring the capacity to prime CD8(+) T cells. These differences may be of consideration in the use of therapeutic DC vaccines based on Mo-DCs.


Assuntos
Apresentação Cruzada/genética , Células Dendríticas/imunologia , Monócitos/citologia , Transcrição Gênica , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/metabolismo , Antígenos/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Apresentação Cruzada/efeitos dos fármacos , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fatores Reguladores de Interferon/metabolismo , Interleucina-4/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Receptores Imunológicos/metabolismo , Proteínas Repressoras/deficiência , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
16.
J Exp Med ; 213(13): 2871-2883, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27899443

RESUMO

In this study, to examine cross-presentation by classical dendritic cells (DCs; cDCs), we evaluated the role of RAB43, a protein found to be selectively expressed by Batf3-dependent CD8α+ and CD103+ compared with other DC subsets and immune lineages. Using a specific monoclonal antibody, we localized RAB43 expression to the Golgi apparatus and LAMP1- cytoplasmic vesicles. Mice with germline or conditional deletion of Rab43 are viable and fertile and have normal development of cDCs but show a defect for in vivo and in vitro cross-presentation of cell-associated antigen. This defect is specific to cDCs, as Rab43-deficient monocyte-derived DCs showed no defect in cross-presentation of cell-associated antigen. These results suggest that RAB43 provides a specialized activity used in cross-presentation selectively by CD8α+ DCs but not other antigen-presenting cells.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos CD8/imunologia , Células Dendríticas/imunologia , Monócitos/imunologia , Proteínas rab de Ligação ao GTP/imunologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Antígenos CD8/genética , Regulação da Expressão Gênica/imunologia , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Camundongos , Camundongos Knockout , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Proteínas rab de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA