Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Ann N Y Acad Sci ; 1445(1): 62-73, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30945313

RESUMEN

One successful class of cancer immunotherapies, immune checkpoint inhibitory antibodies, disrupts key pathways that regulate immune checkpoints, such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). These agents unleash the potency of antigen-experienced T cells that have already been induced as a consequence of the existing tumor. But only 20% of cancers naturally induce T cells. For most cancers, vaccines are require to induce and mobilize T effector cells (Teffs ) to traffick into tumors. We evaluated the effects of anti-CTLA-4 given in combination with an antigen-specific dendritic cell vaccine on intratumoral Teffs in a murine pancreatic cancer model. The dendritic cell-targeted tumor antigen plus anti-CTLA-4 significantly increased the number of vaccine-induced CD4+ Teffs within the tumor. This increase was accompanied by a reduction in the size of the peripheral CD4+ Teff pool. We also found that IL-3 production by activated CD4+ T cells was significantly increased with this combination. Importantly, the CD4+ Teff response was attenuated in Il3-/- mice, suggesting mediation of the effect by IL-3. Finally, the induced T cell infiltration was associated with activation of the tumor endothelium by T cell-derived IL-3. Our findings collectively provide a new insight into the mechanism driving Teff infiltration and vascular activation in a murine pancreatic cancer model, specifically identifying a new role for IL-3 in the anticancer immune response.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Antígeno CTLA-4/antagonistas & inhibidores , Vacunas contra el Cáncer/inmunología , Interleucina-3/inmunología , Neoplasias Pancreáticas/patología , Animales , Linfocitos T CD4-Positivos/inmunología , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Interleucina-3/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Pancreáticas/inmunología
2.
Immun Inflamm Dis ; 7(2): 55-67, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-28474788

RESUMEN

INTRODUCTION: Targeting antigens to dendritic cells (DCs) in vivo via a DC-restricted endocytic receptor, DEC205, has been validated to enhance immunity in several vaccine platforms. Particularly atttractive is selected delivery of proteins to DCs in vivo because it enables proteins to be more immunogenic and provides a cheaper and effective way for repeated immunizations. METHODS: In this study, we tested the efficacy of a single chain antibody to DEC205 (scDEC) to deliver protein antigens selectively to DCs in vivo and to induce protective immunity. RESULTS: In comparison to soluble Ovalbumin (OVA) antigen, when recombinant scDEC:OVA protein was injected subcutaneously (s.c.) into mice, the OVA protein was selectively presented by DCs to both TCR transgenic CD8+ and CD4+ T cells approximately 500 and 100 times more efficient than soluble OVA, respectively, and could persist for seven days following s.c. injection of the scDEC205:OVA. Similarly selective targeting of HIV Gag P24 to DCs in vivo using scDEC-Gag protein plus polyICLC vaccine resulted in strong, long lasting, polyfuntional CD4+ T cells in mice which were protective against airway challenge by a recombinant vaccinia-gag virus. CONCLUSION: Thus targeting protein antigens to DCs using scDEC can be used either alone or in combination with other strategies for effective immunization.


Asunto(s)
Vacunas contra el SIDA/inmunología , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Células Dendríticas/inmunología , Anticuerpos de Cadena Única/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA/administración & dosificación , Adyuvantes Inmunológicos , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células CHO , Línea Celular , Cricetulus , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , VIH/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Infecciones por VIH/prevención & control , Humanos , Inmunización , Inmunogenicidad Vacunal/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Anticuerpos de Cadena Única/farmacología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología
4.
PLoS One ; 11(1): e0146412, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26745371

RESUMEN

Cytokines secreted from dendritic cells (DCs) play an important role in the regulation of T helper (Th) cell differentiation and activation into effector cells. Therefore, controlling cytokine secretion from DCs may potentially regulate Th differentiation/activation. DCs also induce de-novo generation of regulatory T cells (Treg) that modulate the immune response. In the current study we used the mixed leukocyte reaction (MLR) to investigate the effect of allospecific Treg on IL-12, TNFα and IL-6 secretion by DCs. Treg cells were found to markedly down-regulate IL-12 secretion from DCs following stimulation with TLR7/8 agonist. This down-regulation of IL-12 was neither due to a direct suppression of its production by the DCs nor a result of marked DC death. We found that IL-12 was rather actively consumed by Treg cells. IL-12 consumption was mediated by a subpopulation of IL-12Rß2-expressing Treg cells and was dependent on MHC class-II expressed on dendritic cells. Furthermore, IL-12 consumption by Tregs increased their suppressive effect on T cell proliferation and Th1 activation. These results provide a new pathway of Th1 response regulation where IL-12 secreted by DCs is consumed by a sub-population of IL-12Rß2-expressing Treg cells. Consumption of IL-12 by Tregs not only reduces the availability of IL-12 to Th effector cells but also enhances the Treg immunosuppressive effect. This DC-induced IL-12Rß2-expressing Treg subpopulation may have a therapeutic advantage in suppressing Th1 mediated autoimmunity.


Asunto(s)
Linaje de la Célula/inmunología , Células Dendríticas/citología , Interleucina-12/genética , Receptores de Interleucina-12/genética , Linfocitos T Reguladores/citología , Células TH1/citología , Animales , Transporte Biológico , Diferenciación Celular , Linaje de la Célula/genética , Proliferación Celular , Células Dendríticas/inmunología , Femenino , Regulación de la Expresión Génica , Inmunofenotipificación , Interleucina-12/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Prueba de Cultivo Mixto de Linfocitos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Receptores de Interleucina-12/inmunología , Transducción de Señal , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Balance Th1 - Th2 , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/genética , Receptor Toll-Like 8/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
5.
PLoS One ; 10(4): e0124828, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25915045

RESUMEN

Airways infection with Mycobacterium tuberculosis (Mtb) is contained mostly by T cell responses, however, Mtb has developed evasion mechanisms which affect antigen presenting cell (APC) maturation/recruitment delaying the onset of Ag-specific T cell responses. Hypothetically, bypassing the natural infection routes by delivering antigens directly to APCs may overcome the pathogen's naturally evolved evasion mechanisms, thus facilitating the induction of protective immune responses. We generated a murine monoclonal fusion antibody (α-DEC-ESAT) to deliver Early Secretory Antigen Target (ESAT)-6 directly to DEC205+ APCs and to assess its in vivo effects on protection associated responses (IFN-γ production, in vivo CTL killing, and pulmonary mycobacterial load). Treatment with α-DEC-ESAT alone induced ESAT-6-specific IFN-γ producing CD4+ T cells and prime-boost immunization prior to Mtb infection resulted in early influx (d14 post-infection) and increased IFN-γ+ production by specific T cells in the lungs, compared to scarce IFN-γ production in control mice. In vivo CTL killing was quantified in relevant tissues upon transferring target cells loaded with mycobacterial antigens. During infection, α-DEC-ESAT-treated mice showed increased target cell killing in the lungs, where histology revealed cellular infiltrate and considerably reduced bacterial burden. Targeting the mycobacterial antigen ESAT-6 to DEC205+ APCs before infection expands specific T cell clones responsible for early T cell responses (IFN-γ production and CTL activity) and substantially reduces lung bacterial burden. Delivering mycobacterial antigens directly to APCs provides a unique approach to study in vivo the role of APCs and specific T cell responses to assess their potential anti-mycobacterial functions.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Antígenos CD/genética , Antígenos CD/metabolismo , Carga Bacteriana , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Línea Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Citometría de Flujo , Inmunización , Interferón gamma/biosíntesis , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Masculino , Ratones , Antígenos de Histocompatibilidad Menor , Mycobacterium tuberculosis/patogenicidad , Péptidos/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Tuberculosis Pulmonar/metabolismo , Tuberculosis Pulmonar/patología
6.
EMBO Mol Med ; 6(9): 1191-204, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25085878

RESUMEN

Skin dendritic cells (DCs) control the immunogenicity of cutaneously administered vaccines. Antigens targeted to DCs via the C-type lectin Langerin/CD207 are cross-presented to CD8(+) T cells in vivo. We investigated the relative roles of Langerhans cells (LCs) and Langerin(+) dermal DCs (dDCs) in different vaccination settings. Poly(I:C) and anti-CD40 agonist antibody promoted cytotoxic responses upon intradermal immunization with ovalbumin (OVA)-coupled anti-Langerin antibodies (Langerin/OVA). This correlated with CD70 upregulation in Langerin(+) dDCs, but not LCs. In chimeric mice where Langerin targeting was restricted to dDCs, CD8(+) T-cell memory was enhanced. Conversely, providing Langerin/OVA exclusively to LCs failed to prime cytotoxicity, despite initial antigen cross-presentation to CD8(+) T cells. Langerin/OVA combined with imiquimod could not prime CD8(+) T cells and resulted in poor cytotoxicity in subsequent responses. This tolerance induction required targeting and maturation of LCs. Altogether, Langerin(+) dDCs prime long-lasting cytotoxic responses, while cross-presentation by LCs negatively influences CD8(+) T-cell priming. Moreover, this highlights that DCs exposed to TLR agonists can still induce tolerance and supports the existence of qualitatively different DC maturation programs.


Asunto(s)
Antígenos de Superficie/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Células de Langerhans/inmunología , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/inmunología , Animales , Presentación de Antígeno , Reactividad Cruzada , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos
7.
J Exp Med ; 211(9): 1875-91, 2014 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-25135299

RESUMEN

DCs are critical for initiating immunity. The current paradigm in vaccine biology is that DCs migrating from peripheral tissue and classical lymphoid-resident DCs (cDCs) cooperate in the draining LNs to initiate priming and proliferation of T cells. Here, we observe subcutaneous immunity is Fms-like tyrosine kinase 3 ligand (Flt3L) dependent. Flt3L is rapidly secreted after immunization; Flt3 deletion reduces T cell responses by 50%. Flt3L enhances global T cell and humoral immunity as well as both the numbers and antigen capture capacity of migratory DCs (migDCs) and LN-resident cDCs. Surprisingly, however, we find immunity is controlled by cDCs and actively tempered in vivo by migDCs. Deletion of Langerin(+) DC or blockade of DC migration improves immunity. Consistent with an immune-regulatory role, transcriptomic analyses reveals different skin migDC subsets in both mouse and human cluster together, and share immune-suppressing gene expression and regulatory pathways. These data reveal that protective immunity to protein vaccines is controlled by Flt3L-dependent, LN-resident cDCs.


Asunto(s)
Células Dendríticas/inmunología , Proteínas de la Membrana/inmunología , Vacunas/inmunología , Animales , Presentación de Antígeno , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Células Dendríticas/clasificación , Femenino , Expresión Génica , Humanos , Inmunidad Humoral/genética , Inyecciones Intradérmicas , Inyecciones Subcutáneas , Interferón gamma/biosíntesis , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ligandos , Masculino , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/inmunología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Proteínas/inmunología , Subgrupos de Linfocitos T/inmunología , Factores de Transcripción/inmunología , Vacunas/administración & dosificación
8.
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
9.
J Invest Dermatol ; 134(5): 1265-1275, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24288007

RESUMEN

Skin-derived dendritic cells (DCs) are potent antigen-presenting cells with critical roles in both adaptive immunity and tolerance to self. Skin DCs carry antigens and constitutively migrate to the skin-draining lymph nodes (LNs). In mice, Langerin-CD11b- dermal DCs are a low-frequency, heterogeneous, migratory DC subset that traffics to LNs (Langerin-CD11b- migDCs). Here, we build on the observation that Langerin-CD11b- migDCs are Fms-like tyrosine kinase 3 ligand (Flt3L) dependent and strongly Flt3L responsive, which may relate them to classical DCs. Examination of DC capture of FITC from painted skin, DC isolation from skin explant culture, and from the skin of CCR7 knockout mice, which accumulate migDCs, demonstrate these cells are cutaneous residents. Langerin-CD11b- Flt3L-responsive DCs are largely CD24(+) and CX3CR1(low) and can be depleted from Zbtb46-DTR mice, suggesting classical DC lineage. Langerin-CD11b- migDCs present antigen with equal efficiency to other DC subsets ex vivo, including classical CD8α cDCs and Langerin+CD103+ dermal DCs. Finally, transcriptome analysis suggests a close relationship with other skin DCs, and a lineage relationship with other classical DCs. This work demonstrates that Langerin- CD11b- dermal DCs, a previously overlooked cell subset, may be an important contributor to the cutaneous immune environment.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/inmunología , Proteínas de la Membrana/inmunología , Piel/citología , Piel/inmunología , Inmunidad Adaptativa/inmunología , Animales , Antígenos de Superficie/inmunología , Antígenos de Superficie/metabolismo , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Receptor 1 de Quimiocinas CX3C , Movimiento Celular/inmunología , Femenino , Tolerancia Inmunológica/inmunología , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Lectinas de Unión a Manosa/inmunología , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR7/genética , Receptores de Quimiocina/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
10.
J Exp Med ; 210(9): 1871-88, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23960190

RESUMEN

Developing efficacious vaccines against enteric diseases is a global challenge that requires a better understanding of cellular recruitment dynamics at the mucosal surfaces. The current paradigm of T cell homing to the gastrointestinal (GI) tract involves the induction of α4ß7 and CCR9 by Peyer's patch and mesenteric lymph node (MLN) dendritic cells (DCs) in a retinoic acid-dependent manner. This paradigm, however, cannot be reconciled with reports of GI T cell responses after intranasal (i.n.) delivery of antigens that do not directly target the GI lymphoid tissue. To explore alternative pathways of cellular migration, we have investigated the ability of DCs from mucosal and nonmucosal tissues to recruit lymphocytes to the GI tract. Unexpectedly, we found that lung DCs, like CD103(+) MLN DCs, up-regulate the gut-homing integrin α4ß7 in vitro and in vivo, and induce T cell migration to the GI tract in vivo. Consistent with a role for this pathway in generating mucosal immune responses, lung DC targeting by i.n. immunization induced protective immunity against enteric challenge with a highly pathogenic strain of Salmonella. The present report demonstrates novel functional evidence of mucosal cross talk mediated by DCs, which has the potential to inform the design of novel vaccines against mucosal pathogens.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/patología , Inmunidad Mucosa/inmunología , Pulmón/patología , Linfocitos T/inmunología , Administración Intranasal , Traslado Adoptivo , Animales , Antígenos CD/metabolismo , Antígenos de Superficie/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Movimiento Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Clorhidrato de Fingolimod , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Inmunidad Mucosa/efectos de los fármacos , Inmunización , Integrinas/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Glicoles de Propileno/farmacología , Receptores CCR/metabolismo , Salmonella/efectos de los fármacos , Salmonella/inmunología , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Salmonelosis Animal/patología , Salmonelosis Animal/prevención & control , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Factor de Crecimiento Transformador beta/farmacología , Tretinoina/farmacología
11.
PLoS One ; 8(6): e67453, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840706

RESUMEN

Efficient vaccination against the parasite Leishmania major, the causative agent of human cutaneous leishmaniasis, requires development of type 1 T-helper (Th1) CD4(+) T cell immunity. Because of their unique capacity to initiate and modulate immune responses, dendritic cells (DCs) are attractive targets for development of novel vaccines. In this study, for the first time, we investigated the capacity of a DC-targeted vaccine to induce protective responses against L. major. To this end, we genetically engineered the N-terminal portion of the stress-inducible 1 protein of L. major (LmSTI1a) into anti-DEC205/CD205 (DEC) monoclonal antibody (mAb) and thereby delivered the conjugated protein to DEC(+) DCs in situ in the intact animal. Delivery of LmSTI1a to adjuvant-matured DCs increased the frequency of antigen-specific CD4(+) T cells producing IFN-γ(+), IL-2(+), and TNF-α(+) in two different strains of mice (C57BL/6 and Balb/c), while such responses were not observed with the same doses of a control Ig-LmSTI1a mAb without receptor affinity or with non-targeted LmSTI1a protein. Using a peptide library for LmSTI1a, we identified at least two distinct CD4(+) T cell mimetopes in each MHC class II haplotype, consistent with the induction of broad immunity. When we compared T cell immune responses generated after targeting DCs with LmSTI1a or other L. major antigens, including LACK (Leishmania receptor for activated C kinase) and LeIF (Leishmania eukaryotic ribosomal elongation and initiation factor 4a), we found that LmSTI1a was superior for generation of IFN-γ-producing CD4(+) T cells, which correlated with higher protection of susceptible Balb/c mice to a challenge with L. major. For the first time, this study demonstrates the potential of a DC-targeted vaccine as a novel approach for cutaneous leishmaniasis, an increasing public health concern that has no currently available effective treatment.


Asunto(s)
Antígenos de Protozoos/inmunología , Células Dendríticas/inmunología , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Proteínas Protozoarias/inmunología , Células TH1/inmunología , Animales , Proliferación Celular , Células Dendríticas/parasitología , Femenino , Inmunización , Interferón gamma/metabolismo , Leishmaniasis Cutánea/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
12.
Vaccine ; 31(19): 2366-71, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23499580

RESUMEN

Dengue virus (DENV) is the causal agent of severe disease and, in some cases, mortality in humans, but no licensed vaccines against dengue are available. An effective vaccine against dengue requires long-term humoral and cellular immune responses. Several viral proteins have been the subjects of intense research, especially the envelope (E) protein, aimed at developing a vaccine. Domain III of the envelope protein (EDIII) has been identified as a potential candidate because it is involved in binding to host cell receptors and contains epitopes that elicit virus neutralizing antibodies. However, this domain is not sufficiently antigenic when is expressed and administered as antigen to elicit a strong immune response. One alternative to enhance immunogenicity is to target the antigen to dendritic cells to induce T-cells for broad antibody responses. In this work, a single chain antibody fragment (scFv) raised against the DEC-205 receptor fused with the EDIII was successfully expressed in Nicotiana benthamiana. The recombinant protein was expressed and purified from the plant and evaluated in BALB/c mice to test its immunogenicity and ability to induce neutralizing antibodies against DENV. The mice immunized with the recombinant protein produced specific and strong humoral immune responses to DENV. Only two immunizations were required to generate a memory response to DENV without the presence of adjuvants. Also, recognition of the recombinant protein with sera from DENV-infected patients was observed. These findings suggest that this strategy has potential for development of an effective vaccine against DENV.


Asunto(s)
Antígenos CD/inmunología , Vacunas contra el Dengue/inmunología , Dengue/prevención & control , Lectinas Tipo C/inmunología , Receptores de Superficie Celular/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Formación de Anticuerpos , Dengue/inmunología , Virus del Dengue/inmunología , Virus del Dengue/metabolismo , Epítopos/inmunología , Epítopos/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Antígenos de Histocompatibilidad Menor , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
13.
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
14.
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
15.
J Clin Invest ; 123(2): 844-54, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23298832

RESUMEN

Harnessing DCs for immunotherapies in vivo requires the elucidation of the physiological role of distinct DC populations. Migratory DCs traffic from peripheral tissues to draining lymph nodes charged with tissue self antigens. We hypothesized that these DC populations have a specialized role in the maintenance of peripheral tolerance, specifically, to generate suppressive Foxp3+ Tregs. To examine the differential capacity of migratory DCs versus blood-derived lymphoid-resident DCs for Treg generation in vivo, we targeted a self antigen, myelin oligodendrocyte glycoprotein, using antibodies against cell surface receptors differentially expressed in these DC populations. Using this approach together with mouse models that lack specific DC populations, we found that migratory DCs have a superior ability to generate Tregs in vivo, which in turn drastically improve the outcome of experimental autoimmune encephalomyelitis. These results provide a rationale for the development of novel therapies targeting migratory DCs for the treatment of autoimmune diseases.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Periférica/inmunología , Animales , Antígenos CD/inmunología , Antígenos de Superficie/inmunología , Autoantígenos , Movimiento Celular/inmunología , Células Dendríticas/clasificación , Células Dendríticas/fisiología , Encefalomielitis Autoinmune Experimental/inmunología , Factores de Transcripción Forkhead/metabolismo , Células de Langerhans/inmunología , Células de Langerhans/fisiología , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/antagonistas & inhibidores , Lectinas de Unión a Manosa/inmunología , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Glicoproteína Mielina-Oligodendrócito/inmunología , Tolerancia Periférica/fisiología , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Linfocitos T Reguladores/inmunología
16.
Vaccine ; 30(45): 6359-67, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22947140

RESUMEN

It is of great interest to develop a pneumonic plague vaccine that would induce combined humoral and cellular immunity in the lung. Here we investigate a novel approach based on targeting of dendritic cells using the DEC-205/CD205 receptor (DEC) via the intranasal route as way to improve mucosal cellular immunity to the vaccine. Intranasal administration of Yersinia pestis LcrV (V) protein fused to anti-DEC antibody together with poly IC as an adjuvant induced high frequencies of IFN-γ secreting CD4(+) T cells in the airway and lung as well as pulmonary IgG and IgA antibodies. Anti-DEC:LcrV was more efficient to induce IFN-γ/TNF-α/IL-2 secreting polyfunctional CD4(+) T cells when compared to non-targeted soluble protein vaccine. In addition, the intranasal route of immunization with anti-DEC:LcrV was associated with improved survival upon pulmonary challenge with the virulent CO92 Y. pestis. Taken together, these data indicate that targeting dendritic cells via the mucosal route is a potential new avenue for the development of a mucosal vaccine against pneumonic plague.


Asunto(s)
Antígenos CD/inmunología , Células Dendríticas/inmunología , Inmunidad Mucosa , Lectinas Tipo C/inmunología , Pulmón/inmunología , Peste/prevención & control , Receptores de Superficie Celular/inmunología , Adyuvantes Inmunológicos/farmacología , Administración Intranasal , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunidad Celular , Inmunidad Humoral , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Vacuna contra la Peste/inmunología , Poli I-C/inmunología , Poli I-C/farmacología , Proteínas Citotóxicas Formadoras de Poros/inmunología , Yersinia pestis/patogenicidad
17.
PLoS One ; 7(7): e41897, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22860026

RESUMEN

Mass Spectrometry (MS) is becoming a preferred method to identify class I and class II peptides presented on major histocompability complexes (MHC) on antigen presenting cells (APC). We describe a combined computational and MS approach to identify exogenous MHC II peptides presented on mouse spleen dendritic cells (DCs). This approach enables rapid, effective screening of a large number of possible peptides by a computer-assisted strategy that utilizes the extraordinary human ability for pattern recognition. To test the efficacy of the approach, a mixture of epitope peptide mimics (mimetopes) from HIV gag p24 sequence were added exogenously to Fms-like tyrosine kinase 3 ligand (Flt3L)-mobilized splenic DCs. We identified the exogenously added peptide, VDRFYKTLRAEQASQ, and a second peptide, DRFYKLTRAEQASQ, derived from the original exogenously added 15-mer peptide. Furthermore, we demonstrated that our strategy works efficiently with HIV gag p24 protein when delivered, as vaccine protein, to Flt3L expanded mouse splenic DCs in vitro through the DEC-205 receptor. We found that the same MHC II-bound HIV gag p24 peptides, VDRFYKTLRAEQASQ and DRFYKLTRAEQASQ, were naturally processed from anti-DEC-205 HIV gag p24 protein and presented on DCs. The two identified VDRFYKTLRAEQASQ and DRFYKLTRAEQASQ MHC II-bound HIV gag p24 peptides elicited CD4(+) T-cell mediated responses in vitro. Their presentation by DCs to antigen-specific T cells was inhibited by chloroquine (CQ), indicating that optimal presentation of these exogenously added peptides required uptake and vesicular trafficking in mature DCs. These results support the application of our strategy to identify and characterize peptide epitopes derived from vaccine proteins processed by DCs and thus has the potential to greatly accelerate DC-based vaccine development.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/inmunología , Proteína p24 del Núcleo del VIH/inmunología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno , Antígenos Virales/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Epítopos de Linfocito T/metabolismo , Proteína p24 del Núcleo del VIH/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo
18.
J Immunol Methods ; 384(1-2): 184-90, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-22841832

RESUMEN

DEC205/CD205, an endocytic receptor of C-type multilectin, is expressed highly in dendritic cells (DCs). DEC205 was shown to efficiently deliver vaccine antigens in surrogate ligands to the antigen processing and presentation machinery of DCs, which resulted in the development of DC-targeted vaccines employing anti-DC monoclonal antibodies (mAbs). During our studies to characterize a variety of anti-DC mAbs including anti-DEC205 by flow cytometric analysis, we discovered that a secondary anti-immunoglobulin antibody conjugated with PE-Cy5.5 bound strongly to the cells expressing mouse DEC205 (mDEC205) without incubation of a primary anti-mDEC205 mAb. In the present study we demonstrate that various antibodies and streptavidin conjugated with PE-Cy5.5 bind to the mDEC205-expressing cells including CHO, KIT6, and HEK293 cells. The interaction between the PE-Cy5.5 conjugates and the cells expressing mDEC205 appears distinctive, since none of the PE-Cy5.5 conjugates bind to the cells that express human DEC205 on surface. Besides, only PE-Cy5.5 conjugates bind strongly to mDEC205-expressing cells; PerCP-Cy5.5, APC-Cy5.5, and Cy5.5 conjugates bind weakly; PE, PE-Cy5, Cy5, FITC, or Alexa488 conjugates do not bind to mDEC205-expressing cells. Therefore the use of PE-Cy5.5 conjugates, widely utilized in multicolor flow cytometry, requires precaution against nonspecific binding to mDEC205-positive cells.


Asunto(s)
Antígenos CD/inmunología , Inmunoconjugados/inmunología , Lectinas Tipo C/inmunología , Receptores de Superficie Celular/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Unión Competitiva/inmunología , Células CHO , Carbocianinas/química , Cricetinae , Cricetulus , Células Dendríticas/inmunología , Citometría de Flujo , Expresión Génica , Células HEK293 , Humanos , Hibridomas , Inmunoconjugados/química , Inmunoconjugados/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Ratones , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Unión Proteica/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Especificidad de la Especie , Estreptavidina/química
19.
J Exp Med ; 209(5): 1011-28, 2012 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-22547652

RESUMEN

Delta-like ligand 4 (Dll4)-Notch signaling is essential for T cell development and alternative thymic lineage decisions. How Dll4-Notch signaling affects pro-T cell fate and thymic dendritic cell (tDC) development is unknown. We found that Dll4 pharmacological blockade induces accumulation of tDCs and CD4(+)CD25(+)FoxP3(+) regulatory T cells (T(reg) cells) in the thymic cortex. Both genetic inactivation models and anti-Dll4 antibody (Ab) treatment promote de novo natural T(reg) cell expansion by a DC-dependent mechanism that requires major histocompatibility complex II expression on DCs. Anti-Dll4 treatment converts CD4(-)CD8(-)c-kit(+)CD44(+)CD25(-) (DN1) T cell progenitors to immature DCs that induce ex vivo differentiation of naive CD4(+) T cells into T(reg) cells. Induction of these tolerogenic DN1-derived tDCs and the ensuing expansion of T(reg) cells are Fms-like tyrosine kinase 3 (Flt3) independent, occur in the context of transcriptional up-regulation of PU.1, Irf-4, Irf-8, and CSF-1, genes critical for DC differentiation, and are abrogated in thymectomized mice. Anti-Dll4 treatment fully prevents type 1 diabetes (T1D) via a T(reg) cell-mediated mechanism and inhibits CD8(+) T cell pancreatic islet infiltration. Furthermore, a single injection of anti-Dll4 Ab reverses established T1D. Disease remission and recurrence are correlated with increased T(reg) cell numbers in the pancreas-draining lymph nodes. These results identify Dll4-Notch as a novel Flt3-alternative pathway important for regulating tDC-mediated T(reg) cell homeostasis and autoimmunity.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/fisiología , Diabetes Mellitus Tipo 1/prevención & control , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Secretasas de la Proteína Precursora del Amiloide/deficiencia , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Anticuerpos/farmacología , Western Blotting , Proteínas de Unión al Calcio , Cartilla de ADN/genética , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Genes MHC Clase II/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Páncreas/patología , Reacción en Cadena de la Polimerasa , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Timo/citología , Timo/inmunología , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...