Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 30: 1-22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22136168

RESUMEN

A properly functioning adaptive immune system signifies the best features of life. It is diverse beyond compare, tolerant without fail, and capable of behaving appropriately with a myriad of infections and other challenges. Dendritic cells are required to explain how this remarkable system is energized and directed. I frame this article in terms of the major decisions that my colleagues and I have made in dendritic cell science and some of the guiding themes at the time the decisions were made. As a result of progress worldwide, there is now evidence of a central role for dendritic cells in initiating antigen-specific immunity and tolerance. The in vivo distribution and development of a previously unrecognized white cell lineage is better understood, as is the importance of dendritic cell maturation to link innate and adaptive immunity in response to many stimuli. Our current focus is on antigen uptake receptors on dendritic cells. These receptors enable experiments involving selective targeting of antigens in situ and new approaches to vaccine design in preclinical and clinical systems.


Asunto(s)
Alergia e Inmunología/tendencias , Células Dendríticas/inmunología , Alergia e Inmunología/historia , Animales , Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Tolerancia Inmunológica/inmunología , Inmunidad Celular/inmunología , Lectinas Tipo C/metabolismo , Antígenos de Histocompatibilidad Menor , Modelos Animales , Receptores de Superficie Celular/metabolismo , Vacunas/inmunología
2.
Cell ; 143(3): 416-29, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-21029863

RESUMEN

Dendritic cells (DCs), critical antigen-presenting cells for immune control, normally derive from bone marrow precursors distinct from monocytes. It is not yet established if the large reservoir of monocytes can develop into cells with critical features of DCs in vivo. We now show that fully differentiated monocyte-derived DCs (Mo-DCs) develop in mice and DC-SIGN/CD209a marks the cells. Mo-DCs are recruited from blood monocytes into lymph nodes by lipopolysaccharide and live or dead gram-negative bacteria. Mobilization requires TLR4 and its CD14 coreceptor and Trif. When tested for antigen-presenting function, Mo-DCs are as active as classical DCs, including cross-presentation of proteins and live gram-negative bacteria on MHC I in vivo. Fully differentiated Mo-DCs acquire DC morphology and localize to T cell areas via L-selectin and CCR7. Thus the blood monocyte reservoir becomes the dominant presenting cell in response to select microbes, yielding DC-SIGN(+) cells with critical functions of DCs.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Células Dendríticas/citología , Escherichia coli/inmunología , Lectinas Tipo C/metabolismo , Monocitos/citología , Receptores de Superficie Celular/metabolismo , Animales , Presentación de Antígeno , Moléculas de Adhesión Celular/inmunología , Células Dendríticas/inmunología , Selectina L/inmunología , Lectinas Tipo C/inmunología , Receptores de Lipopolisacáridos/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/inmunología , Receptores CCR7/inmunología , Receptores de Superficie Celular/inmunología , Linfocitos T/inmunología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología
4.
Immunity ; 35(5): 819-31, 2011 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-22078798

RESUMEN

Early events in atherosclerosis occur in the aortic intima and involve monocytes that become macrophages. We looked for these cells in the steady state adult mouse aorta, and surprisingly, we found a dominance of dendritic cells (DCs) in the intima. In contrast to aortic adventitial macrophages, CD11c(+)MHC II(hi) DCs were poorly phagocytic but were immune stimulatory. DCs were of two types primarily: classical Flt3-Flt3L signaling-dependent, CD103(+)CD11b(-) DCs and macrophage-colony stimulating factor (M-CSF)-dependent, CD14(+)CD11b(+)DC-SIGN(+) monocyte-derived DCs. Both types expanded during atherosclerosis. By crossing Flt3(-/-) to Ldlr(-/-) atherosclerosis-prone mice, we developed a selective and marked deficiency of classical CD103(+) aortic DCs, and they were associated with exacerbated atherosclerosis without alterations in blood lipids. Concomitantly, the Flt3(-/-)Ldlr(-/-) mice had fewer Foxp3(+) Treg cells and increased inflammatory cytokine mRNAs in the aorta. Therefore, functional DCs are dominant in normal aortic intima and, in contrast to macrophages, CD103(+) classical DCs are associated with atherosclerosis protection.


Asunto(s)
Aterosclerosis/inmunología , Células Dendríticas/inmunología , Transducción de Señal , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Antígenos CD/metabolismo , Aorta/efectos de los fármacos , Aorta/inmunología , Aterosclerosis/genética , Aterosclerosis/patología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/inmunología , Procedimientos de Reducción del Leucocitos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Tirosina Quinasa 3 Similar a fms/genética
5.
Immunity ; 29(3): 319-24, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18799140

RESUMEN

Dendritic cells (DCs) are the antigen presenting cells that initiate and regulate immunity. By studying these cells in vivo, we will be able to move beyond standard approaches and design vaccines that directly harness the elaborate properties of DCs to control immunity.


Asunto(s)
Presentación de Antígeno/inmunología , Citocinas/inmunología , Células Dendríticas/inmunología , Vacunas/inmunología , Animales , Células Dendríticas/fisiología , Humanos , Inmunidad Mucosa
6.
PLoS Biol ; 12(1): e1001759, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24409099

RESUMEN

Type I interferons (IFNs) play an important role in direct antiviral defense as well as linking the innate and adaptive immune responses. On dendritic cells (DCs), IFNs facilitate their activation and contribute to CD8(+) and CD4(+) T cell priming. However, the precise molecular mechanism by which IFNs regulate maturation and immunogenicity of DCs in vivo has not been studied in depth. Here we show that, after in vivo stimulation with the TLR ligand poly IC, IFNs dominate transcriptional changes in DCs. In contrast to direct TLR3/mda5 signaling, IFNs are required for upregulation of all pathways associated with DC immunogenicity. In addition, metabolic pathways, particularly the switch from oxidative phosphorylation to glycolysis, are also regulated by IFNs and required for DC maturation. These data provide evidence for a metabolic reprogramming concomitant with DC maturation and offer a novel mechanism by which IFNs modulate DC maturation.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Glucólisis/efectos de los fármacos , Interferón-alfa/genética , Fosforilación Oxidativa/efectos de los fármacos , Poli I-C/farmacología , Inmunidad Adaptativa , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad Innata , Inyecciones Intraperitoneales , Helicasa Inducida por Interferón IFIH1 , Interferón-alfa/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Transcripción Genética
7.
Blood ; 121(25): 5034-44, 2013 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-23482932

RESUMEN

Functional differences between human dendritic cell (DC) subsets and the potential benefits of targeting them with vaccines remain poorly defined. Here we describe that mice with reconstituted human immune system components (huNSG mice) develop all human conventional and plasmacytoid DC compartments in lymphoid organs. Testing different Toll-like receptor agonists for DC maturation in vivo, we found that IL-12p70 and interferon (IFN)-α production correlated with the maturation of CD141+ (BDCA3+) conventional DCs in huNSG mice. Furthermore, depletion of CD141+ DCs before stimulation significantly reduced IFN-α levels in vivo. This DC subset produced similar total amounts but different subtypes of IFN-α in response to synthetic double-stranded RNA compared with plasmacytoid DCs in response to a single-stranded RNA equivalent. Moreover, synthetic double-stranded RNA as adjuvant and antigen targeting to the endocytic receptor DEC-205, a combination that focuses antigen presentation for T-cell priming on CD141+ DCs, stimulated antigen-specific human CD4+ T-cell responses. Thus, the human CD141+ DC subset is a prominent source of IFN-α and interleukin-12 production and should be further evaluated for vaccine development.


Asunto(s)
Antígenos CD/inmunología , Células Dendríticas/inmunología , Interferón-alfa/biosíntesis , Lectinas Tipo C/inmunología , Activación de Linfocitos/inmunología , ARN Bicatenario/inmunología , Receptores de Superficie Celular/inmunología , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/citología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Interferón-alfa/inmunología , Ratones , Antígenos de Histocompatibilidad Menor
8.
Nat Chem Biol ; 9(4): 250-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23416331

RESUMEN

Targeted delivery of antigens to dendritic cells (DCs) is a promising vaccination strategy. However, to ensure immunity, the approach depends on coadministration of an adjuvant. Here we ask whether targeting of both adjuvant and antigen to DCs is sufficient to induce immunity. Using a protein ligation method, we develop a general approach for linking the immune stimulant, poly dA:dT (pdA:dT), to a monoclonal antibody (mAb) specific for DEC205 (DEC). We show that DEC-specific mAbs deliver pdA:dT to DCs for the efficient production of type I interferon in human monocyte-derived DCs and in mice. Notably, adaptive T-cell immunity is elicited when mAbs specific for DEC-pdA:dT are used as the activation stimuli and are administered together with a DC-targeted antigen. Collectively, our studies indicate that DCs can integrate innate and adaptive immunity in vivo and suggest that dual delivery of antigen and adjuvant to DCs might be an efficient approach to vaccine development.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Anticuerpos Monoclonales/inmunología , Antígenos CD/inmunología , Antígenos/inmunología , Células Dendríticas/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inmunoconjugados/inmunología , Lectinas Tipo C/inmunología , Poli dA-dT/inmunología , Receptores de Superficie Celular/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/química , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Antígenos/administración & dosificación , Antígenos/química , Antígenos CD/administración & dosificación , Antígenos CD/química , Células Dendríticas/inmunología , Sistemas de Liberación de Medicamentos , Vectores Genéticos , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Lectinas Tipo C/administración & dosificación , Lectinas Tipo C/química , Ratones , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Plásmidos , Poli dA-dT/administración & dosificación , Poli dA-dT/química , Receptores de Superficie Celular/administración & dosificación , Receptores de Superficie Celular/química
10.
J Immunol ; 188(3): 1147-55, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22210914

RESUMEN

Members of the triggering expressed on myeloid cells (Trem) receptor family fine-tune inflammatory responses. We previously identified one of these receptors, called Treml4, expressed mainly in the spleen, as well as at high levels by CD8α(+) dendritic cells and macrophages. Like other Trem family members, Treml4 has an Ig-like extracellular domain and a short cytoplasmic tail that associates with the adaptor DAP12. To follow up on our initial results that Treml4-Fc fusion proteins bind necrotic cells, we generated a knockout mouse to assess the role of Treml4 in the uptake and presentation of dying cells in vivo. Loss of Treml4 expression did not impair uptake of dying cells by CD8α(+) dendritic cells or cross-presentation of cell-associated Ag to CD8(+) T cells, suggesting overlapping function between Treml4 and other receptors in vivo. To further investigate Treml4 function, we took advantage of a newly generated mAb against Treml4 and engineered its H chain to express three different Ags (i.e., OVA, HIV GAGp24, and the extracellular domain of the breast cancer protein HER2). OVA directed to Treml4 was efficiently presented to CD8(+) and CD4(+) T cells in vivo. Anti-Treml4-GAGp24 mAbs, given along with a maturation stimulus, induced Th1 Ag-specific responses that were not observed in Treml4 knockout mice. Also, HER2 targeting using anti-Treml4 mAbs elicited combined CD4(+) and CD8(+) T cell immunity, and both T cells participated in resistance to a transplantable tumor. Therefore, Treml4 participates in Ag presentation in vivo, and targeting Ags with anti-Treml4 Abs enhances immunization of otherwise naive mice.


Asunto(s)
Presentación de Antígeno/inmunología , Receptor ErbB-2/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Inmunidad Celular , Inmunización , Ratones , Ratones Noqueados , Sustancias Protectoras , Ingeniería de Proteínas
11.
Proc Natl Acad Sci U S A ; 108(6): 2384-9, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21262813

RESUMEN

Improved protein-based vaccines should facilitate the goal of effective vaccines against HIV and other pathogens. With respect to T cells, the efficiency of immunization, or "immunogenicity," is improved by targeting vaccine proteins to maturing dendritic cells (DCs) within mAbs to DC receptors. Here, we compared the capacity of Langerin/CD207, DEC205/CD205, and Clec9A receptors, each expressed on the CD8(+) DC subset in mice, to bring about immunization of microbial-specific T cells from the polyclonal repertoire, using HIV gag-p24 protein as an antigen. α-Langerin mAb targeted splenic CD8(+) DCs selectively in vivo, whereas α-DEC205 and α-Clec9A mAbs targeted additional cell types. When the mAb heavy chains were engineered to express gag-p24, the α-Langerin, α-DEC205, and α-Clec9A fusion mAbs given along with a maturation stimulus induced comparable levels of gag-specific T helper 1 (Th1) and CD8(+) T cells in BALB/c × C57BL/6 F1 mice. These immune T cells were more numerous than targeting the CD8(-) DC subset with α-DCIR2-gag-p24. In an in vivo assay in which gag-primed T cells were used to report the early stages of T-cell responses, α-Langerin, α-DEC205, and α-Clec9A also mediated cross-presentation to primed CD8(+) T cells if, in parallel to antigen uptake, the DCs were stimulated with α-CD40. α-Langerin, α-DEC205, and α-Clec9A targeting greatly enhanced T-cell immunization relative to nonbinding control mAb or nontargeted HIV gag-p24 protein. Therefore, when the appropriate subset of DCs is targeted with a vaccine protein, several different receptors expressed by that subset are able to initiate combined Th1 and CD8(+) immunity.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Antígenos de Superficie/inmunología , Antígenos CD8 , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Proteína p24 del Núcleo del VIH/inmunología , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/inmunología , Receptores de Superficie Celular/inmunología , Receptores Inmunológicos/inmunología , Células TH1/inmunología , Vacunas contra el SIDA/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Proteína p24 del Núcleo del VIH/farmacología , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Antígenos de Histocompatibilidad Menor
12.
Proc Natl Acad Sci U S A ; 108(17): 7131-6, 2011 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-21467219

RESUMEN

Protein vaccines, if rendered immunogenic, would facilitate vaccine development against HIV and other pathogens. We compared in nonhuman primates (NHPs) immune responses to HIV Gag p24 within 3G9 antibody to DEC205 ("DEC-HIV Gag p24"), an uptake receptor on dendritic cells, to nontargeted protein, with or without poly ICLC, a synthetic double stranded RNA, as adjuvant. Priming s.c. with 60 µg of both HIV Gag p24 vaccines elicited potent CD4(+) T cells secreting IL-2, IFN-γ, and TNF-α, which also proliferated. The responses increased with each of three immunizations and recognized multiple Gag peptides. DEC-HIV Gag p24 showed better cross-priming for CD8(+) T cells, whereas the avidity of anti-Gag antibodies was ∼10-fold higher with nontargeted Gag 24 protein. For both protein vaccines, poly ICLC was essential for T- and B-cell immunity. To determine whether adaptive responses could be further enhanced, animals were boosted with New York vaccinia virus (NYVAC)-HIV Gag/Pol/Nef. Gag-specific CD4(+) and CD8(+) T-cell responses increased markedly after priming with both protein vaccines and poly ICLC. These data reveal qualitative differences in antibody and T-cell responses to DEC-HIV Gag p24 and Gag p24 protein and show that prime boost with protein and adjuvant followed by NYVAC elicits potent cellular immunity.


Asunto(s)
Vacunas contra el SIDA/inmunología , Adyuvantes Inmunológicos/farmacología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteína p24 del Núcleo del VIH/inmunología , ARN Bicatenario/farmacología , Virus Vaccinia/inmunología , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/farmacología , Animales , Linfocitos B/inmunología , Citocinas/inmunología , Femenino , Anticuerpos Anti-VIH/inmunología , Proteína p24 del Núcleo del VIH/farmacología , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Inmunidad Celular/inmunología , Macaca mulatta , Masculino , ARN Bicatenario/inmunología , Virus Vaccinia/genética
13.
J Exp Med ; 204(6): 1359-69, 2007 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-17502666

RESUMEN

The ability of dendritic cells (DCs) to activate immunity is linked to their maturation status. In prior studies, we have shown that selective antibody-mediated blockade of inhibitory FcgammaRIIB receptor on human DCs in the presence of activating immunoglobulin (Ig) ligands leads to DC maturation and enhanced immunity to antibody-coated tumor cells. We show that Fcgamma receptor (FcgammaR)-mediated activation of human monocytes and monocyte-derived DCs is associated with a distinct gene expression pattern, including several inflammation-associated chemokines, as well as type 1 interferon (IFN) response genes, including the activation of signal transducer and activator of transcription 1 (STAT1). FcgammaR-mediated STAT1 activation is rapid and requires activating FcgammaRs. However, this IFN response is observed without a detectable increase in the expression of type I IFNs themselves or the need to add exogenous IFNs. Induction of IFN response genes plays an important role in FcgammaR-mediated effects on DCs, as suppression of STAT1 by RNA interference inhibited FcgammaR-mediated DC maturation. These data suggest that the balance of activating/inhibitory FcgammaRs may regulate IFN signaling in myeloid cells. Manipulation of FcgammaR balance on DCs and monocytes may provide a novel approach to regulating IFN-mediated pathways in autoimmunity and human cancer.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Interferón Tipo I/inmunología , Monocitos/inmunología , Receptores de IgG/antagonistas & inhibidores , Transducción de Señal/inmunología , Anticuerpos/farmacología , Western Blotting , Citocinas/metabolismo , Células Dendríticas/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Interferón Tipo I/metabolismo , Monocitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , Factor de Transcripción STAT1/genética
14.
J Exp Med ; 204(1): 191-201, 2007 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-17210729

RESUMEN

Most treatments that prevent autoimmune diabetes in nonobese diabetic (NOD) mice require intervention at early pathogenic stages, when insulitis is first developing. We tested whether dendritic cell (DC)-expanded, islet antigen-specific CD4+ CD25+ suppressor T cells could treat diabetes at later stages of disease, when most of the insulin-producing islet beta cells had been destroyed by infiltrating lymphocytes. CD4+ CD25+ CD62L+ regulatory T cells (T reg cells) from BDC2.5 T cell receptor transgenic mice were expanded with antigen-pulsed DCs and IL-2, and were then injected into NOD mice. A single dose of as few as 5x10(4) of these islet-specific T reg cells blocked diabetes development in prediabetic 13-wk-old NOD mice. The T reg cells also induced long-lasting reversal of hyperglycemia in 50% of mice in which overt diabetes had developed. Successfully treated diabetic mice had similar responses to glucose challenge compared with nondiabetic NOD mice. The successfully treated mice retained diabetogenic T cells, but also had substantially increased Foxp3+ cells in draining pancreatic lymph nodes. However, these Foxp3+ cells were derived from the recipient mice and not the injected T reg cells, suggesting a role for endogenous T reg cells in maintaining tolerance after treatment. Therefore, inoculation of DC-expanded, antigen-specific suppressor T cells has considerable efficacy in ameliorating ongoing diabetes in NOD mice.


Asunto(s)
Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/terapia , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Islotes Pancreáticos/inmunología , Selectina L/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Femenino , Factores de Transcripción Forkhead/metabolismo , Insulina/biosíntesis , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Estado Prediabético/sangre , Estado Prediabético/inmunología , Estado Prediabético/terapia , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante
15.
J Exp Med ; 204(5): 1095-106, 2007 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-17438065

RESUMEN

Interferon (IFN)-gamma, a cytokine critical for resistance to infection and tumors, is produced by CD4(+) helper T lymphocytes after stimulation by cultured dendritic cells (DCs) that secrete a cofactor, interleukin (IL)-12. We have identified a major IL-12-independent pathway whereby DCs induce IFN-gamma-secreting T helper (Th)1 CD4(+) T cells in vivo. This pathway requires the membrane-associated tumor necrosis family member CD70 and was identified by targeting the LACK antigen from Leishmania major within an antibody to CD205 (DEC-205), an uptake receptor on a subset of DCs. Another major DC subset, targeted with 33D1 anti-DCIR2 antibody, also induced IFN-gamma in vivo but required IL-12, not CD70. Isolated CD205(+) DCs expressed cell surface CD70 when presenting antigen to T cell receptor transgenic T cells, and this distinction was independent of maturation stimuli. CD70 was also essential for CD205(+) DC function in vivo. Detection of the IL-12-independent IFN-gamma pathway was obscured with nontargeted LACK, which was presented by both DC subsets. This in situ analysis points to CD70 as a decision maker for Th1 differentiation by CD205(+) DCs, even in Th2-prone BALB/c animals and potentially in vaccine design. The results indicate that two DC subsets have innate propensities to differentially affect the Th1/Th2 balance in vivo and by distinct mechanisms.


Asunto(s)
Ligando CD27/metabolismo , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Interferón gamma/metabolismo , Subgrupos Linfocitarios/inmunología , Transducción de Señal/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos CD/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/inmunología , Ensayo de Inmunoadsorción Enzimática , Interleucina-12/metabolismo , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Receptores de Superficie Celular/inmunología
16.
Eur J Immunol ; 42(1): 101-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22002164

RESUMEN

Protein-based vaccines offer safety and cost advantages but require adjuvants to induce immunity. Here we examined the adjuvant capacity of glucopyranosyl lipid A (GLA), a new synthetic non-toxic analogue of lipopolysaccharide. In mice, in comparison with non-formulated LPS and monophosphoryl lipid A, formulated GLA induced higher antibody titers and generated Type 1 T-cell responses to HIV gag-p24 protein in spleen and lymph nodes, which was dependent on TLR4 expression. Immunization was greatly improved by targeting HIV gag p24 to DCs with an antibody to DEC-205, a DC receptor for antigen uptake and processing. Subcutaneous immunization induced antigen-specific T-cell responses in the intestinal lamina propria. Immunity did not develop in mice transiently depleted of DCs. To understand how GLA works, we studied DCs directly from vaccinated mice. Within 4 h, GLA caused DCs to upregulate CD86 and CD40 and produce cytokines including IL-12p70 in vivo. Importantly, DCs removed from mice 4 h after vaccination became immunogenic, capable of inducing T-cell immunity upon injection into naïve mice. These data indicate that a synthetic and clinically feasible TLR4 agonist rapidly stimulates full maturation of DCs in vivo, allowing for adaptive immunity to develop many weeks to months later.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Células Dendríticas/efectos de los fármacos , Lípido A/análogos & derivados , Receptor Toll-Like 4/agonistas , Vacunas de Subunidad/inmunología , Animales , Anticuerpos Antivirales/sangre , Células Dendríticas/inmunología , Células Dendríticas/virología , VIH/inmunología , Proteína p24 del Núcleo del VIH/inmunología , Lípido A/farmacología , Tejido Linfoide/inmunología , Tejido Linfoide/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organismos Libres de Patógenos Específicos , Receptor Toll-Like 4/inmunología , Vacunas de Subunidad/farmacología
17.
Nature ; 449(7161): 419-26, 2007 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-17898760

RESUMEN

Dendritic cells (DCs) orchestrate a repertoire of immune responses that bring about resistance to infection and silencing or tolerance to self. In the settings of infection and cancer, microbes and tumours can exploit DCs to evade immunity, but DCs also can generate resistance, a capacity that is readily enhanced with DC-targeted vaccines. During allergy, autoimmunity and transplant rejection, DCs instigate unwanted responses that cause disease, but, again, DCs can be harnessed to silence these conditions with novel therapies. Here we present some medical implications of DC biology that account for illness and provide opportunities for prevention and therapy.


Asunto(s)
Células Dendríticas/inmunología , Medicina , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/terapia , Células Dendríticas/citología , Humanos , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/fisiopatología , Enfermedades del Sistema Inmune/terapia , Medicina/métodos , Neoplasias/inmunología , Neoplasias/terapia , Inmunología del Trasplante
18.
Proc Natl Acad Sci U S A ; 107(9): 4281-6, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20160099

RESUMEN

To improve the efficacy of T cell-based vaccination, we pursued the principle that CD4(+) T cells provide help for functional CD8(+) T cell immunity. To do so, we administered HIV gag to mice successively as protein and DNA vaccines. To achieve strong CD4(+) T cell immunity, the protein vaccine was targeted selectively to DEC-205, a receptor for antigen presentation on dendritic cells. This targeting helped CD8(+) T cell immunity develop to a subsequent DNA vaccine and improved protection to intranasal challenge with recombinant vaccinia gag virus, including more rapid accumulation of CD8(+) T cells in the lung. The helper effect of dendritic cell-targeted protein vaccine was mimicked by immunization with specific MHC II binding HIV gag peptides but not peptides from a disparate Yersinia pestis microbe. CD4(+) helper cells upon adoptive transfer allowed wild-type, but not CD40(-/-), recipient mice to respond better to the DNA vaccine. The transfer also enabled recipients to more rapidly accumulate gag-specific CD8(+) T cells in the lung following challenge with vaccinia gag virus. Thus, complementary prime boost vaccination, in which prime and boost favor distinct types of T cell immunity, improves plasmid DNA immunization, including mobilization of CD8(+) T cells to sites of infection.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos T CD8-positivos/inmunología , Productos del Gen gag/inmunología , Vacunas de ADN/inmunología , Vacunas contra el SIDA/administración & dosificación , Administración Intranasal , Secuencia de Aminoácidos , Animales , Productos del Gen gag/química , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Vacunas de ADN/administración & dosificación
19.
Breast Cancer Res ; 14(2): R39, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22397502

RESUMEN

INTRODUCTION: Given their relative simplicity of manufacture and ability to be injected repeatedly, vaccines in a protein format are attractive for breast and other cancers. However, soluble human epidermal growth factor receptor (HER2)/neu protein as a vaccine has not been immunogenic. When protein is directly targeted to antigen uptake receptors, such as DEC205 (DEC), efficient processing and presentation of antigen take place. The aim of this study was to determine the immunogenicity of a HER2 protein vaccine that directly targets to DEC+ dendritic cells (DCs) in a mouse breast cancer model. METHODS: We genetically engineered the HER2 extracellular domain into a monoclonal antibody specific for DEC (DEC-HER2). Mice of various genetic backgrounds were immunized with DEC-HER2 in combination with DC maturation stimuli (poly IC ± CD40 Ab). Vaccine-induced T cell immunity was determined by analyzing the ability of CD4+/CD8+ T cell to produce interferon (IFN)-gamma and proliferate upon antigen rechallenge. Sera were assessed for the presence of antigen specific antibody (Ab). For vaccine efficacy, FVB/N mice were immunized with DEC-HER2 in combination with poly IC and protection against neu-expressing mammary tumors was assessed. Protection mechanisms and tumor-specific T cell responses were also evaluated. RESULTS: We demonstrate that DEC-HER2 fusion mAb, but not Ctrl Ig-HER2, elicits strong, broad and multifunctional CD4+ T cell immunity, CD8+ T cell responses, and humoral immunity specific for HER2 antigen. Cross-reactivity to rat neu protein was also observed. Importantly, mice xeno-primed with DEC-HER2 were protected from a neu-expressing mammary tumor challenge. Both CD4+ and CD8+ T cells mediated the tumor protection. Robust anti-tumor T cell immunity was detected in tumor protected mice. CONCLUSIONS: Immunization of mice with HER2 protein vaccine targeting DEC+ DCs in vivo induced high levels of T- and B-cell immunity. Non-targeted HER2 protein was poorly immunogenic for CD4+ and CD8+ T cells. This vaccination approach provided long-term survival benefit for mice challenged with neu-expressing tumor following as little as 2.7 µg of HER2 protein incorporated in the vaccine. Vaccine-induced CD4+ and CD8+ T cells were both essential for tumor protection. This immunization strategy demonstrates great potential towards the development of vaccines for breast cancer patients.


Asunto(s)
Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Receptor ErbB-2/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Neoplasias de la Mama/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Femenino , Humanos , Inmunidad Humoral , Interferón gamma/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Poli I-C/inmunología , Poli I-C/farmacología , Estructura Terciaria de Proteína/genética , Receptor ErbB-2/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Linfocitos T/inmunología
20.
J Exp Med ; 203(13): 2887-93, 2006 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-17145955

RESUMEN

HIV-1 infects immature dendritic cells (iDCs), but infection is inefficient compared with activated CD4+ T cells and only involves a small subset of iDCs. We analyzed whether this could be attributed to specific cellular restrictions during the viral life cycle. To study env-independent restriction to HIV-1 infection, we used a single-round infection assay with HIV-1 pseudotyped with vesicular stomatitis virus G protein (HIV-VSVG). Small interfering RNA-mediated depletion of APOBEC3G/3F (A3G/3F), but not TRIM5alpha, enhanced HIV-1 infection of iDCs, indicating that A3G/3F controls the sensitivity of iDCs to HIV-1 infection. Furthermore, sequences of HIV reverse transcripts revealed G-to-A hypermutation of HIV genomes during iDC infection, demonstrating A3G/3F cytidine deaminase activity in iDCs. When we separated the fraction of iDCs that was susceptible to HIV, we found the cells to be deficient in A3G messenger RNA and protein. We also noted that during DC maturation, which further reduces susceptibility to infection, A3G levels increased. These findings highlight a role for A3G/3F in explaining the resistance of most DCs to HIV-1 infection, as well as the susceptibility of a fraction of iDCs. An increase in the A3G/3F-mediated intrinsic resistance of iDCs could result in a block of HIV infection at its mucosal point of entry.


Asunto(s)
Citosina Desaminasa/fisiología , Células Dendríticas/virología , VIH-1/crecimiento & desarrollo , Monocitos/citología , Nucleósido Desaminasas/fisiología , Proteínas Represoras/fisiología , Desaminasa APOBEC-3G , Factores de Restricción Antivirales , Secuencia de Bases , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Citidina Desaminasa , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , ADN Viral/genética , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Citometría de Flujo , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , VIH-1/genética , Células HeLa , Humanos , Células Jurkat , Lipopolisacáridos/farmacología , Glicoproteínas de Membrana/genética , Datos de Secuencia Molecular , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , Mutación Puntual , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Homología de Secuencia de Ácido Nucleico , Transfección , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteínas del Envoltorio Viral/genética , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA