Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Infect Dis ; 226(12): 2054-2063, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-35543281

RESUMO

BACKGROUND: Respiratory syncytial virus (RSV) is an important cause of disease in older adults. We evaluated the safety and immunogenicity of a stabilized RSV prefusion F subunit (RSVpreF) vaccine candidate with/without adjuvant in adults aged 65-85 years. METHODS: Primary cohort participants were equally randomized to 1 of 7 RSVpreF formulations: 60 µg with either Al(OH)3 or CpG/Al(OH)3, 120 µg with either Al(OH)3 or CpG/Al(OH)3, 240 µg with either Al(OH)3 or CpG/Al(OH)3, 240 µg unadjuvanted, or placebo, administered concomitantly with high-dose seasonal inactivated influenza vaccine (SIIV). Participants in the month 0,2 cohort were randomized to RSVpreF 240 µg with CpG/Al(OH)3 or placebo, administered at months 0 and 2. RESULTS: All RSVpreF vaccine candidates elicited robust and persistent serum neutralizing responses when administered alone or with SIIV. There was no notable difference in neutralizing response between the formulations, including those containing CpG. In the month 0,2 cohort, there was no booster effect of dose 2. SIIV responses were similar or slightly lower with concomitant administration of RSVpreF. Most systemic and local reactions were mild and more frequent after RSVpreF than placebo. CONCLUSIONS: RSVpreF formulations were well tolerated and elicited robust neutralizing responses in older adults; however, CpG/Al(OH)3 did not further enhance responses. Clinical Trials Registration. NCT03572062.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Humano , Humanos , Idoso , Proteínas Virais de Fusão , Anticorpos Neutralizantes , Anticorpos Antivirais , Adjuvantes Imunológicos , Adjuvantes Farmacêuticos
2.
J Infect Dis ; 226(4): 585-594, 2022 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-35413121

RESUMO

The development of a vaccine to prevent congenital human cytomegalovirus (HCMV) disease is a public health priority. We tested rhesus CMV (RhCMV) prototypes of HCMV vaccine candidates in a seronegative macaque oral challenge model. Immunogens included a recombinant pentameric complex (PC; gH/gL/pUL128/pUL130/pUL131A), a postfusion gB ectodomain, and a DNA plasmid that encodes pp65-2. Immunization with QS21-adjuvanted PC alone or with the other immunogens elicited neutralizing titers comparable to those elicited by RhCMV infection. Similarly, immunization with all 3 immunogens elicited pp65-specific cytotoxic T-cell responses comparable to those elicited by RhCMV infection. RhCMV readily infected immunized animals and was detected in saliva, blood, and urine after challenge in quantities similar to those in placebo-immunized animals. If HCMV evades vaccine-elicited immunity in humans as RhCMV evaded immunity in macaques, a HCMV vaccine must elicit immunity superior to, or different from, that elicited by the prototype RhCMV vaccine to block horizontal transmission.


Assuntos
Infecções por Citomegalovirus , Vacinas contra Citomegalovirus , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Citomegalovirus , Humanos , Macaca mulatta , Proteínas do Envelope Viral
3.
Proc Natl Acad Sci U S A ; 117(38): 23730-23741, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32879009

RESUMO

Although plasmacytoid dendritic cells (pDCs) have been shown to play a critical role in generating viral immunity and promoting tolerance to suppress antitumor immunity, whether and how pDCs cross-prime CD8 T cells in vivo remain controversial. Using a pDC-targeted vaccine model to deliver antigens specifically to pDCs, we have demonstrated that pDC-targeted vaccination led to strong cross-priming and durable CD8 T cell immunity. Surprisingly, cross-presenting pDCs required conventional DCs (cDCs) to achieve cross-priming in vivo by transferring antigens to cDCs. Taking advantage of an in vitro system where only pDCs had access to antigens, we further demonstrated that cross-presenting pDCs were unable to efficiently prime CD8 T cells by themselves, but conferred antigen-naive cDCs the capability of cross-priming CD8 T cells by transferring antigens to cDCs. Although both cDC1s and cDC2s exhibited similar efficiency in acquiring antigens from pDCs, cDC1s but not cDC2s were required for cross-priming upon pDC-targeted vaccination, suggesting that cDC1s played a critical role in pDC-mediated cross-priming independent of their function in antigen presentation. Antigen transfer from pDCs to cDCs was mediated by previously unreported pDC-derived exosomes (pDCexos), that were also produced by pDCs under various conditions. Importantly, all these pDCexos primed naive antigen-specific CD8 T cells only in the presence of bystander cDCs, similarly to cross-presenting pDCs, thus identifying pDCexo-mediated antigen transfer to cDCs as a mechanism for pDCs to achieve cross-priming. In summary, our data suggest that pDCs employ a unique mechanism of pDCexo-mediated antigen transfer to cDCs for cross-priming.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Apresentação Cruzada/imunologia , Células Dendríticas/metabolismo , Exossomos/metabolismo , Animais , Apresentação de Antígeno/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Exossomos/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL
4.
Immunity ; 45(6): 1205-1218, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28002729

RESUMO

Inflammation triggers the differentiation of Ly6Chi monocytes into microbicidal macrophages or monocyte-derived dendritic cells (moDCs). Yet, it is unclear whether environmental inflammatory cues control the polarization of monocytes toward each of these fates or whether specialized monocyte progenitor subsets exist before inflammation. Here, we have shown that naive monocytes are phenotypically heterogeneous and contain an NR4A1- and Flt3L-independent, CCR2-dependent, Flt3+CD11c-MHCII+PU.1hi subset. This subset acted as a precursor for FcγRIII+PD-L2+CD209a+, GM-CSF-dependent moDCs but was distal from the DC lineage, as shown by fate-mapping experiments using Zbtb46. By contrast, Flt3-CD11c-MHCII-PU.1lo monocytes differentiated into FcγRIII+PD-L2-CD209a-iNOS+ macrophages upon microbial stimulation. Importantly, Sfpi1 haploinsufficiency genetically distinguished the precursor activities of monocytes toward moDCs or microbicidal macrophages. Indeed, Sfpi1+/- mice had reduced Flt3+CD11c-MHCII+ monocytes and GM-CSF-dependent FcγRIII+PD-L2+CD209a+ moDCs but generated iNOS+ macrophages more efficiently. Therefore, intercellular disparities of PU.1 expression within naive monocytes segregate progenitor activity for inflammatory iNOS+ macrophages or moDCs.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Transferência Adotiva , Animais , Antígenos Ly/imunologia , Separação Celular , Células Dendríticas/citologia , Citometria de Fluxo , Macrófagos/citologia , Camundongos , Monócitos/citologia , Óxido Nítrico Sintase Tipo II/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase
5.
Cell Metab ; 23(5): 852-66, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27166946

RESUMO

Plasmacytoid dendritic cells (pDCs) are unique bone-marrow-derived cells that produce large amounts of type I interferon in response to microbial stimulation. Furthermore, pDCs also promote T cell tolerance in sterile-inflammation conditions. However, the immunomodulatory role of aortic pDCs in atherosclerosis has been poorly understood. Here, we identified functional mouse and human pDCs in the aortic intima and showed that selective, inducible pDC depletion in mice exacerbates atherosclerosis. Aortic pDCs expressed CCR9 and indoleamine 2,3-dioxygenase 1 (IDO-1), an enzyme involved in driving the generation of regulatory T cells (Tregs). As a consequence, loss of pDCs resulted in decreased numbers of Tregs and reduced IL-10 levels in the aorta. Moreover, antigen presentation by pDCs expanded antigen-specific Tregs in the atherosclerotic aorta. Notably, Tregs ablation affected pDC homeostasis in diseased aorta. Accordingly, pDCs in human atherosclerotic aortas colocalized with Tregs. Collectively, we identified a mechanism of atheroprotection mediated by tolerogenic aortic pDCs.


Assuntos
Aorta/patologia , Aterosclerose/enzimologia , Aterosclerose/prevenção & controle , Células Dendríticas/enzimologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Anticorpos/farmacologia , Aterosclerose/imunologia , Aterosclerose/patologia , Medula Óssea/patologia , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Epitopos , Homeostase/efeitos dos fármacos , Humanos , Interferon Tipo I/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Receptores de LDL/metabolismo , Fatores de Tempo , Receptor Toll-Like 9/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo
6.
J Immunol Methods ; 434: 32-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27073171

RESUMO

Conventional dendritic cells (cDCs) are essential immune cells linking the innate and adaptive immune system. cDC depletion in mice is an important method to study the function of these cells in vivo. Here we report an inducible in vivo system for cDC depletion in which excision of a loxP flanked Stop signal enables expression of the human diphtheria toxin receptor (DTR) under the control of Zbtb46 (zDC(lSlDTR)). cDCs can be specifically depleted by combining zDC(lSlDTR) mice with a Csf1r(Cre) driver line. In addition, we show that zDC(Cre) mice can be used to produce cDC specific conditional knockout mice (Irf8, Irf4, Notch2) which lack specific subsets of cDCs.


Assuntos
Células da Medula Óssea/citologia , Células Dendríticas/imunologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/imunologia , Animais , Células da Medula Óssea/imunologia , Cruzamentos Genéticos , Células Dendríticas/citologia , Citometria de Fluxo/métodos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Baço/imunologia
7.
Nature ; 507(7491): 243-7, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24509714

RESUMO

The transcription factors c-Myc and N-Myc--encoded by Myc and Mycn, respectively--regulate cellular growth and are required for embryonic development. A third paralogue, Mycl1, is dispensable for normal embryonic development but its biological function has remained unclear. To examine the in vivo function of Mycl1 in mice, we generated an inactivating Mycl1(gfp) allele that also reports Mycl1 expression. We find that Mycl1 is selectively expressed in dendritic cells (DCs) of the immune system and controlled by IRF8, and that during DC development, Mycl1 expression is initiated in the common DC progenitor concurrent with reduction in c-Myc expression. Mature DCs lack expression of c-Myc and N-Myc but maintain L-Myc expression even in the presence of inflammatory signals such as granulocyte-macrophage colony-stimulating factor. All DC subsets develop in Mycl1-deficient mice, but some subsets such as migratory CD103(+) conventional DCs in the lung and liver are greatly reduced at steady state. Importantly, loss of L-Myc by DCs causes a significant decrease in in vivo T-cell priming during infection by Listeria monocytogenes and vesicular stomatitis virus. The replacement of c-Myc by L-Myc in immature DCs may provide for Myc transcriptional activity in the setting of inflammation that is required for optimal T-cell priming.


Assuntos
Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Linfócitos T/imunologia , Animais , Antígenos CD/metabolismo , Divisão Celular , Células Dendríticas/citologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Cadeias alfa de Integrinas/metabolismo , Fatores Reguladores de Interferon/metabolismo , Listeria monocytogenes/imunologia , Fígado/citologia , Fígado/imunologia , Pulmão/citologia , Pulmão/imunologia , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-myc/deficiência , Transcrição Gênica , Vesiculovirus/imunologia
8.
J Exp Med ; 210(10): 2025-39, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-24043764

RESUMO

Dendritic cells (DCs), monocytes, and macrophages are closely related phagocytes that share many phenotypic features and, in some cases, a common developmental origin. Although the requirement for DCs in initiating adaptive immune responses is well appreciated, the role of monocytes and macrophages remains largely undefined, in part because of the lack of genetic tools enabling their specific depletion. Here, we describe a two-gene approach that requires overlapping expression of LysM and Csf1r to define and deplete monocytes and macrophages. The role of monocytes and macrophages in immunity to pathogens was tested by their selective depletion during infection with Citrobacter rodentium. Although neither cell type was required to initiate immunity, monocytes and macrophages contributed to the adaptive immune response by secreting IL-12, which induced Th1 polarization and IFN-γ secretion. Thus, whereas DCs are indispensable for priming naive CD4(+) T cells, monocytes and macrophages participate in intestinal immunity by producing mediators that direct T cell polarization.


Assuntos
Citrobacter rodentium/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Macrófagos/imunologia , Monócitos/imunologia , Subpopulações de Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Movimento Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/imunologia , Ordem dos Genes , Interleucina-12/biossíntese , Interleucina-12/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Muramidase/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Subpopulações de Linfócitos T/metabolismo
9.
Crit Rev Immunol ; 32(6): 489-501, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23428225

RESUMO

Dendritic cells are professional antigen presenting cells linking the innate and the adaptive immune system. Antibody mediated antigen delivery to functionally distinct DC subpopulations is a promising approach to induce or inhibit antigen specific immune responses in vivo and is useful for designing therapeutic vaccination strategies. This review focuses on the state of the art of this technique and describes recent findings of antigen targeting to the subpopulation of plasmacytoid dendritic cells and the possibilities to induce immunity or attenuate autoimmunity by delivering antigens specifically to this cell type.


Assuntos
Apresentação de Antígeno , Vacinas Anticâncer , Células Dendríticas/imunologia , Animais , Humanos , Tolerância Imunológica , Imunidade Celular , Imunomodulação
10.
J Immunol ; 186(12): 6718-25, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21555533

RESUMO

Plasmacytoid dendritic cells (PDCs) are capable of presenting Ags to T cells in a tolerogenic or immunogenic manner depending on the formulation of the Ag and the mode of stimulation. It has not been investigated whether effective adaptive immune responses useful for vaccination can be induced by Ab-mediated Ag targeting to PDCs in vivo. In this study, we show that Ag delivered to murine PDCs via bone marrow stromal cell Ag 2 (BST2)/CD317 in combination with TLR agonists as adjuvants is specifically presented by PDCs in vivo and elicits strong cellular and humoral immune responses. These include IFN-γ production by CD4(+) T cells and high Ab titers with a broad range of IgG isotypes. In addition, BST2-mediated Ag delivery in the presence of polyinosinic-polycytidylic acid as adjuvant induces cytotoxic T lymphocytes that are functional in vivo. A single immunization with Ag-fused anti-BST2 Ab together with polyinosinic-polycytidylic acid as adjuvant is sufficient to trigger protective immunity against subsequent viral infection and tumor growth. We conclude that despite the potential tolerogenic properties of PDCs, Ag targeting to PDCs in combination with TLR agonists as adjuvants is an effective vaccination strategy.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos CD/imunologia , Células Dendríticas/imunologia , Imunidade Celular/imunologia , Glicoproteínas de Membrana/imunologia , Vacinação/métodos , Animais , Anticorpos/administração & dosagem , Anticorpos/uso terapêutico , Antígenos/administração & dosagem , Antígenos/uso terapêutico , Citotoxicidade Imunológica , Camundongos , Proteínas Recombinantes de Fusão/administração & dosagem
11.
Blood ; 117(24): 6562-70, 2011 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-21508410

RESUMO

Whereas the final differentiation of conventional dendritic cells (CDCs) from committed precursors occurs locally in secondary lymphoid or peripheral tissues, plasmacytoid dendritic cells (PDCs) are thought to fully develop in the bone marrow from common DC progenitors before migrating to the periphery. In our study, we define, for the first time, a subpopulation of CCR9(-) major histocompatibility complex class II(low) PDCs in murine bone marrow, which express E2-2 and are immediate precursors of CCR9(+) fully differentiated PDCs. However, CCR9(-) PDCs have the plasticity to acquire the phenotype and function of CD11b(+) CD8α(-) major histocompatibility complex class II(high) CDC-like cells under the influence of soluble factors produced by intestinal epithelial cells or recombinant GM-CSF. This deviation from the PDC lineage commitment is regulated on the level of transcription factors reflected by down-regulation of E2-2 and up-regulation of ID2, PU.1, and BATF3. Thus, CCR9(-) PDCs are immediate PDC precursors that can be reprogrammed to differentiate into CDC-like cells with higher antigen-presenting and cytokine-producing capacity under the influence of the local tissue microenvironment.


Assuntos
Diferenciação Celular , Células Dendríticas/citologia , Células Dendríticas/fisiologia , Receptores CCR/metabolismo , Células-Tronco/fisiologia , Animais , Apresentação de Antígeno/fisiologia , Antígeno CD11b/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Separação Celular/métodos , Células Cultivadas , Células Dendríticas/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Células-Tronco/citologia , Células-Tronco/metabolismo
12.
Eur J Immunol ; 41(5): 1334-43, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21469103

RESUMO

Exogenous and endogenous RNA ligands of Toll-like receptor (TLR) 7 which are present during viral infection or autoimmune diseases such as systemic lupus erythematosus (SLE) directly activate DCs and B cells and thus support the generation of effector T and B lymphocytes. However, the generation of effective antiviral or autoreactive adaptive immune responses requires blocking of immunosuppression by Tregs. In this study, we show that TLR7 ligands reduce the number of Tregs generated de novo from naïve murine T cells in vitro and in vivo. In the presence of TLR7-activated splenic DCs, Foxp3 was transiently induced in naïve T cells by TGF-ß but was downregulated at later time points. Neutralization experiments revealed that loss of Foxp3 after initial induction was mostly dependent on IL-6 produced in the DC-T-cell cocultures containing TLR7 ligands. Thus, under the influence of TLR7 ligands fewer Tregs were generated and these expressed lower levels of Foxp3 correlating with a reduced capacity to suppress responder T-cell proliferation. Thus, we provide evidence that TLR7 ligands affect Treg-dependent immune regulation and may thereby contribute to the development of autoimmune diseases such as systemic lupus erythematosus.


Assuntos
Células Dendríticas/imunologia , Fatores de Transcrição Forkhead/metabolismo , Glicoproteínas de Membrana/imunologia , Linfócitos T Reguladores/imunologia , Receptor 7 Toll-Like/imunologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Animais , Diferenciação Celular , Técnicas de Cocultura , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Tolerância Imunológica , Interleucina-6/imunologia , Ligantes , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , RNA/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA