Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunity ; 44(4): 924-38, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27096321

RESUMEN

Large numbers of melanoma lesions develop resistance to targeted inhibition of mutant BRAF or fail to respond to checkpoint blockade. We explored whether modulation of intratumoral antigen-presenting cells (APCs) could increase responses to these therapies. Using mouse melanoma models, we found that CD103(+) dendritic cells (DCs) were the only APCs transporting intact antigens to the lymph nodes and priming tumor-specific CD8(+) T cells. CD103(+) DCs were required to promote anti-tumoral effects upon blockade of the checkpoint ligand PD-L1; however, PD-L1 inhibition only led to partial responses. Systemic administration of the growth factor FLT3L followed by intratumoral poly I:C injections expanded and activated CD103(+) DC progenitors in the tumor, enhancing responses to BRAF and PD-L1 blockade and protecting mice from tumor rechallenge. Thus, the paucity of activated CD103(+) DCs in tumors limits checkpoint-blockade efficacy and combined FLT3L and poly I:C therapy can enhance tumor responses to checkpoint and BRAF blockade.


Asunto(s)
Antígenos CD/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Cadenas alfa de Integrinas/metabolismo , Melanoma Experimental/inmunología , Poli I-C/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/farmacología , Animales , Presentación de Antígeno/inmunología , Línea Celular Tumoral , Células Dendríticas/citología , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Oncoimmunology ; 4(4): e998538, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26137416

RESUMEN

Whereas preclinical investigations and clinical studies have established that CD8+ T cells can profoundly affect cancer progression, the underlying mechanisms are still elusive. Challenging the prevalent view that the beneficial effect of CD8+ T cells in cancer is solely attributable to their cytotoxic activity, several reports have indicated that the ability of CD8+ T cells to promote tumor regression is dependent on their cytokine secretion profile and their ability to self-renew. Evidence has also shown that the tumor microenvironment can disarm CD8+ T cell immunity, leading to the emergence of dysfunctional CD8+ T cells. The existence of different types of CD8+ T cells in cancer calls for a more precise definition of the CD8+ T cell immune phenotypes in cancer and the abandonment of the generic terms "pro-tumor" and "antitumor." Based on recent studies investigating the functions of CD8+ T cells in cancer, we here propose some guidelines to precisely define the functional states of CD8+ T cells in cancer.

3.
J Immunol ; 192(12): 5586-98, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24829414

RESUMEN

Blood monocytes from children with systemic lupus erythematosus (SLE) behave similar to dendritic cells (DCs), and SLE serum induces healthy monocytes to differentiate into DCs in a type I IFN-dependent manner. In this study, we found that these monocytes display significant transcriptional changes, including a prominent IFN signature, compared with healthy controls. Few of those changes, however, explain DC function. Exposure to allogeneic T cells in vitro reprograms SLE monocytes to acquire DC phenotype and function, and this correlates with both IFN-inducible (IP10) and proinflammatory cytokine (IL-1ß and IL6) expression. Furthermore, we found that both IFN and SLE serum induce the upregulation of CCR7 transcription in these cells. CCR7 protein expression, however, requires a second signal provided by TLR agonists such as LPS. Thus, SLE serum "primes" a subset of monocytes to readily (<24 h) respond to TLR agonists and acquire migratory DC properties. Our findings might explain how microbial infections exacerbate lupus.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Lupus Eritematoso Sistémico/inmunología , Adolescente , Adulto , Niño , Citocinas/inmunología , Células Dendríticas/patología , Femenino , Humanos , Lipopolisacáridos/farmacología , Lupus Eritematoso Sistémico/patología , Receptores CCR7/inmunología , Receptores Toll-Like/agonistas , Receptores Toll-Like/inmunología
4.
Sci Transl Med ; 5(176): 176ra32, 2013 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-23486778

RESUMEN

Seasonal influenza vaccine protects 60 to 90% of healthy young adults from influenza infection. The immunological events that lead to the induction of protective antibody responses remain poorly understood in humans. We identified the type of CD4+ T cells associated with protective antibody responses after seasonal influenza vaccinations. The administration of trivalent split-virus influenza vaccines induced a temporary increase of CD4+ T cells expressing ICOS, which peaked at day 7, as did plasmablasts. The induction of ICOS was largely restricted to CD4+ T cells coexpressing the chemokine receptors CXCR3 and CXCR5, a subpopulation of circulating memory T follicular helper cells. Up to 60% of these ICOS+CXCR3+CXCR5+CD4+ T cells were specific for influenza antigens and expressed interleukin-2 (IL-2), IL-10, IL-21, and interferon-γ upon antigen stimulation. The increase of ICOS+CXCR3+CXCR5+CD4+ T cells in blood correlated with the increase of preexisting antibody titers, but not with the induction of primary antibody responses. Consistently, purified ICOS+CXCR3+CXCR5+CD4+ T cells efficiently induced memory B cells, but not naïve B cells, to differentiate into plasma cells that produce influenza-specific antibodies ex vivo. Thus, the emergence of blood ICOS+CXCR3+CXCR5+CD4+ T cells correlates with the development of protective antibody responses generated by memory B cells upon seasonal influenza vaccination.


Asunto(s)
Formación de Anticuerpos/inmunología , Vacunas contra la Influenza/inmunología , Receptores CXCR3/metabolismo , Receptores CXCR5/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Adulto , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Ligando de CD40/metabolismo , Células Cultivadas , Niño , Citocinas/metabolismo , Femenino , Citometría de Flujo , Humanos , Masculino , Linfocitos T Colaboradores-Inductores/metabolismo
5.
J Exp Med ; 209(1): 109-21, 2012 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-22213806

RESUMEN

Dendritic cells (DCs) can initiate and shape host immune responses toward either immunity or tolerance by their effects on antigen-specific CD4(+) T cells. DC-asialoglycoprotein receptor (DC-ASGPR), a lectinlike receptor, is a known scavenger receptor. Here, we report that targeting antigens to human DCs via DC-ASGPR, but not lectin-like oxidized-LDL receptor, Dectin-1, or DC-specific ICAM-3-grabbing nonintegrin favors the generation of antigen-specific suppressive CD4(+) T cells that produce interleukin 10 (IL-10). These findings apply to both self- and foreign antigens, as well as memory and naive CD4(+) T cells. The generation of such IL-10-producing CD4(+) T cells requires p38/extracellular signal-regulated kinase phosphorylation and IL-10 induction in DCs. We further demonstrate that immunization of nonhuman primates with antigens fused to anti-DC-ASGPR monoclonal antibody generates antigen-specific CD4(+) T cells that produce IL-10 in vivo. This study provides a new strategy for the establishment of antigen-specific IL-10-producing suppressive T cells in vivo by targeting whole protein antigens to DCs via DC-ASGPR.


Asunto(s)
Antígenos/inmunología , Receptor de Asialoglicoproteína/metabolismo , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Interleucina-10/biosíntesis , Animales , Antígenos/metabolismo , Células Dendríticas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ligandos , Macaca , Masculino , Receptores Depuradores de Clase E/metabolismo , Transducción de Señal
6.
J Immunol ; 185(6): 3504-13, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20729328

RESUMEN

Dectin-1, a C-type lectin recognizing fungal and mycobacterial pathogens, can deliver intracellular signals that activate dendritic cells (DCs), resulting in initiation of immune responses and expansion of Th17 CD4(+) T cell responses. In this paper, we studied the roles of human Dectin-1 (hDectin-1) expressed on DCs in the induction and activation of Ag-specific CD8(+) T cell responses. We first generated an agonistic anti-hDectin-1 mAb, which recognizes the hDectin-1 Glu(143)-Ile(162) region. It bound to in vitro monocyte-derived DCs and to in vivo CD1c(+)CD1a(+) dermal DCs but not to epidermal Langerhans cells. Anti-hDectin-1-mediated DC activation resulted in upregulation of costimulatory molecules and secretion of multiple cytokines and chemokines in a Syk-dependent manner. DCs activated with the anti-hDectin-1 mAb could significantly enhance both neo and foreign Ag-specific CD8(+) T cell responses by promoting both the expansion of CD8(+) T cells and their functional activities. We further demonstrated that delivering Ags to DCs via hDectin-1 using anti-hDectin-1-Ag conjugates resulted in potent Ag-specific CD8(+) T cell responses. Thus, hDectin-1 expressed on DCs can contribute to the induction and activation of cellular immunity against intracellular pathogens, such as mycobacteria, that are recognized by DCs via Dectin-1. Vaccines based on delivering Ags to DCs with an agonistic anti-hDectin-1 mAb could elicit CD8(+) T cell-mediated immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Especificidad de Anticuerpos/genética , Sitios de Unión de Anticuerpos/genética , Línea Celular , Células Cultivadas , Epítopos de Linfocito T/genética , Humanos , Lectinas Tipo C , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/inmunología , Proteínas Recombinantes de Fusión/farmacología
7.
J Immunother ; 31(9): 793-805, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18833008

RESUMEN

Cancer immunotherapy seeks to mobilize a patient's immune system for therapeutic benefit. It can be passive, that is, transfer of immune effector cells (T cells) or proteins (antibodies), or active, that is, vaccination. Early clinical trials testing vaccination with ex vivo generated dendritic cells (DCs) pulsed with tumor antigens provide a proof-of-principle that therapeutic immunity can be elicited. Yet, the clinical benefit measured by regression of established tumors in patients with stage IV cancer has been observed in a fraction of patients only. The next generation of DC vaccines is expected to generate large numbers of high avidity effector CD8 T cells and to overcome regulatory T cells and suppressive environment established by tumors, a major obstacle in metastatic disease. Therapeutic vaccination protocols will combine improved DC vaccines with chemotherapy to exploit immunogenic chemotherapy regimens. We foresee adjuvant vaccination in patients with resected tumors but at high risk of relapse to be based on in vivo targeting of DCs with fusion proteins containing anti-DCs antibodies, antigens from tumor stem/propagating cells, and DC activators.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Células Dendríticas/metabolismo , Inmunoterapia , Neoplasias/terapia , Subgrupos de Linfocitos T/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Terapia Combinada , Células Dendríticas/patología , Humanos , Estadificación de Neoplasias , Neoplasias/patología , Subgrupos de Linfocitos T/patología , Escape del Tumor/inmunología
8.
Blood ; 107(7): 2613-8, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16317096

RESUMEN

Host response to viral infection involves distinct effectors of innate and adaptive immunity, whose mobilization needs to be coordinated to ensure protection. Here we show that influenza virus triggers, in human blood dendritic-cell (DC) subsets (ie, plasmacytoid and myeloid DCs), a coordinated chemokine (CK) secretion program with 3 successive waves. The first one, occurring at early time points (2 to 4 hours), includes CKs potentially attracting effector cells such as neutrophils, cytotoxic T cells, and natural killer (NK) cells (CXCL16, CXCL1, CXCL2, and CXCL3). The second one occurs within 8 to 12 hours and includes CKs attracting effector memory T cells (CXCL8, CCL3, CCL4, CCL5, CXCL9, CXCL10, and CXCL11). The third wave, which occurs after 24 to 48 hours, when DCs have reached the lymphoid organs, includes CCL19, CCL22, and CXCL13, which attract naive T and B lymphocytes. Thus, human blood DC subsets carry a common program of CK production, which allows for a coordinated attraction of the different immune effectors in response to viral infection.


Asunto(s)
Citocinas/genética , Células Dendríticas/inmunología , Células Dendríticas/virología , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Movimiento Celular , Citocinas/clasificación , Células Dendríticas/clasificación , Células Epiteliales/inmunología , Humanos , Memoria Inmunológica , Cinética , Análisis de Secuencia por Matrices de Oligonucleótidos , Orthomyxoviridae/inmunología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...