Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nat Commun ; 12(1): 1669, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33723257

RESUMEN

Immune checkpoint inhibitors are used for treating patients with metastatic melanoma. Since the response to treatment is variable, biomarkers are urgently needed to identify patients who may benefit from such therapy. Here, we combine single-cell RNA-sequencing and multiparameter flow cytometry to assess changes in circulating CD8+ T cells in 28 patients with metastatic melanoma starting anti-PD-1 therapy, followed for 6 months: 17 responded to therapy, whilst 11 did not. Proportions of activated and proliferating CD8+ T cells and of mucosal-associated invariant T (MAIT) cells are significantly higher in responders, prior to and throughout therapy duration. MAIT cells from responders express higher level of CXCR4 and produce more granzyme B. In silico analysis support MAIT presence in the tumor microenvironment. Finally, patients with >1.7% of MAIT among peripheral CD8+ population show a better response to treatment. Our results thus suggest that MAIT cells may be considered a biomarker for patients responding to anti-PD-1 therapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Células T Invariantes Asociadas a Mucosa/inmunología , Células T Invariantes Asociadas a Mucosa/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Anciano , Anciano de 80 o más Años , Biomarcadores , Linfocitos T CD8-positivos/inmunología , Femenino , Granzimas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Receptores CXCR4/metabolismo
2.
Nat Immunol ; 21(12): 1552-1562, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046887

RESUMEN

T cell memory relies on the generation of antigen-specific progenitors with stem-like properties. However, the identity of these progenitors has remained unclear, precluding a full understanding of the differentiation trajectories that underpin the heterogeneity of antigen-experienced T cells. We used a systematic approach guided by single-cell RNA-sequencing data to map the organizational structure of the human CD8+ memory T cell pool under physiological conditions. We identified two previously unrecognized subsets of clonally, epigenetically, functionally, phenotypically and transcriptionally distinct stem-like CD8+ memory T cells. Progenitors lacking the inhibitory receptors programmed death-1 (PD-1) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) were committed to a functional lineage, whereas progenitors expressing PD-1 and TIGIT were committed to a dysfunctional, exhausted-like lineage. Collectively, these data reveal the existence of parallel differentiation programs in the human CD8+ memory T cell pool, with potentially broad implications for the development of immunotherapies and vaccines.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Memoria Inmunológica , Células Progenitoras Linfoides/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Biomarcadores , Diferenciación Celular/inmunología , Biología Computacional/métodos , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunofenotipificación , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Ratones , Homeostasis del Telómero
3.
Sci Transl Med ; 12(548)2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32554710

RESUMEN

Local delivery of anticancer agents has the potential to maximize treatment efficacy and minimize the acute and long-term systemic toxicities. Here, we used unsupervised systematic evolution of ligands by exponential enrichment to identify four RNA aptamers that specifically recognized mouse and human myeloid cells infiltrating tumors but not their peripheral or circulating counterparts in multiple mouse models and from patients with head and neck squamous cell carcinoma (HNSCC). The use of these aptamers conjugated to doxorubicin enhanced the accumulation and bystander release of the chemotherapeutic drug in both primary and metastatic tumor sites in breast and fibrosarcoma mouse models. In the 4T1 mammary carcinoma model, these doxorubicin-conjugated aptamers outperformed Doxil, the first clinically approved highly optimized nanoparticle for targeted chemotherapy, promoting tumor regression after just three administrations with no detected changes in weight loss or blood chemistry. These RNA aptamers recognized tumor infiltrating myeloid cells in a variety of mouse tumors in vivo and from human HNSCC ex vivo. This work suggests the use of RNA aptamers for the detection of myeloid-derived suppressor cells in humans and for a targeted delivery of chemotherapy to the tumor microenvironment in multiple malignancies.


Asunto(s)
Antineoplásicos , Aptámeros de Nucleótidos , Neoplasias de Cabeza y Cuello , Células Supresoras de Origen Mieloide , Animales , Línea Celular Tumoral , Humanos , Indicadores y Reactivos , Ratones , Carcinoma de Células Escamosas de Cabeza y Cuello , Microambiente Tumoral
4.
J Immunol ; 203(12): 3179-3189, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31740485

RESUMEN

In mice, the ability of naive T (TN) cells to mount an effector response correlates with TCR sensitivity for self-derived Ags, which can be quantified indirectly by measuring surface expression levels of CD5. Equivalent findings have not been reported previously in humans. We identified two discrete subsets of human CD8+ TN cells, defined by the absence or presence of the chemokine receptor CXCR3. The more abundant CXCR3+ TN cell subset displayed an effector-like transcriptional profile and expressed TCRs with physicochemical characteristics indicative of enhanced interactions with peptide-HLA class I Ags. Moreover, CXCR3+ TN cells frequently produced IL-2 and TNF in response to nonspecific activation directly ex vivo and differentiated readily into Ag-specific effector cells in vitro. Comparative analyses further revealed that human CXCR3+ TN cells were transcriptionally equivalent to murine CXCR3+ TN cells, which expressed high levels of CD5. These findings provide support for the notion that effector differentiation is shaped by heterogeneity in the preimmune repertoire of human CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Receptores CXCR3/metabolismo , Adulto , Factores de Edad , Anciano , Animales , Biomarcadores , Células Cultivadas , Femenino , Humanos , Memoria Inmunológica , Inmunofenotipificación , Activación de Linfocitos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto Joven
5.
Front Immunol ; 10: 1973, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31481962

RESUMEN

Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the first step in the kynurenine pathway of tryptophan (Trp) degradation that produces several biologically active Trp metabolites. L-kynurenine (Kyn), the first byproduct by IDO1, promotes immunoregulatory effects via activation of the Aryl hydrocarbon Receptor (AhR) in dendritic cells (DCs) and T lymphocytes. We here identified the nuclear coactivator 7 (NCOA7) as a molecular target of 3-hydroxyanthranilic acid (3-HAA), a Trp metabolite produced downstream of Kyn along the kynurenine pathway. In cells overexpressing NCOA7 and AhR, the presence of 3-HAA increased the association of the two molecules and enhanced Kyn-driven, AhR-dependent gene transcription. Physiologically, conventional (cDCs) but not plasmacytoid DCs or other immune cells expressed high levels of NCOA7. In cocultures of CD4+ T cells with cDCs, the co-addition of Kyn and 3-HAA significantly increased the induction of Foxp3+ regulatory T cells and the production of immunosuppressive transforming growth factor ß in an NCOA7-dependent fashion. Thus, the co-presence of NCOA7 and the Trp metabolite 3-HAA can selectively enhance the activation of ubiquitary AhR in cDCs and consequent immunoregulatory effects. Because NCOA7 is often overexpressed and/or mutated in tumor microenvironments, our current data may provide evidence for a new immune check-point mechanism based on Trp metabolism and AhR.


Asunto(s)
Ácido 3-Hidroxiantranílico/metabolismo , Células Dendríticas/metabolismo , Coactivadores de Receptor Nuclear/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Células Dendríticas/inmunología , Femenino , Humanos , Quinurenina/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Coactivadores de Receptor Nuclear/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Linfocitos T Reguladores/inmunología
6.
Nat Protoc ; 14(7): 1946-1969, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31160786

RESUMEN

The interrogation of single cells is revolutionizing biology, especially our understanding of the immune system. Flow cytometry is still one of the most versatile and high-throughput approaches for single-cell analysis, and its capability has been recently extended to detect up to 28 colors, thus approaching the utility of cytometry by time of flight (CyTOF). However, flow cytometry suffers from autofluorescence and spreading error (SE) generated by errors in the measurement of photons mainly at red and far-red wavelengths, which limit barcoding and the detection of dim markers. Consequently, development of 28-color fluorescent antibody panels for flow cytometry is laborious and time consuming. Here, we describe the steps that are required to successfully achieve 28-color measurement capability. To do this, we provide a reference map of the fluorescence spreading errors in the 28-color space to simplify panel design and predict the success of fluorescent antibody combinations. Finally, we provide detailed instructions for the computational analysis of such complex data by existing, popular algorithms (PhenoGraph and FlowSOM). We exemplify our approach by designing a high-dimensional panel to characterize the immune system, but we anticipate that our approach can be used to design any high-dimensional flow cytometry panel of choice. The full protocol takes a few days to complete, depending on the time spent on panel design and data analysis.


Asunto(s)
Anticuerpos Monoclonales , Citometría de Flujo/métodos , Técnica del Anticuerpo Fluorescente/métodos , Análisis de la Célula Individual/métodos , Algoritmos , Biomarcadores , Tampones (Química) , Colorantes , Biología Computacional/métodos , Factores de Transcripción Forkhead/inmunología , Humanos , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Linfocitos T/citología , Linfocitos T/inmunología
7.
J Exp Med ; 216(2): 317-336, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30655308

RESUMEN

Altered control of T follicular helper (Tfh) cells can lead to generation of autoantibodies and autoimmune manifestations. Signaling pathways that selectively limit pathogenic responses without affecting the protective function of Tfh cells are unknown. Here we show that the ATP-gated ionotropic P2X7 receptor restricts the expansion of aberrant Tfh cells and the generation of self-reactive antibodies in experimental murine lupus, but its activity is dispensable for the expansion of antigen-specific Tfh cells during vaccination. P2X7 stimulation promotes caspase-mediated pyroptosis of Tfh cells and controls the development of pathogenic ICOS+ IFN-γ-secreting cells. Circulating Tfh cells from patients with systemic lupus erythematosus (SLE) but not primary antiphospholipid syndrome (PAPS), a nonlupus systemic autoimmune disease, were hyporesponsive to P2X7 stimulation and resistant to P2X7-mediated inhibition of cytokine-driven expansion. These data point to the P2X7 receptor as a checkpoint regulator of Tfh cells; thus, restoring P2X7 activity in SLE patients could selectively limit the progressive amplification of pathogenic autoantibodies, which deteriorate patients' conditions.


Asunto(s)
Lupus Eritematoso Sistémico/inmunología , Receptores Purinérgicos P2X7/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Modelos Animales de Enfermedad , Interferón gamma/genética , Interferón gamma/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Noqueados , Piroptosis/genética , Piroptosis/inmunología , Receptores Purinérgicos P2X7/genética , Linfocitos T Colaboradores-Inductores/patología
8.
J Exp Med ; 215(10): 2520-2535, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30154266

RESUMEN

CD8+ T cells infiltrating tumors are largely dysfunctional, but whether a subset maintains superior functionality remains ill defined. By high-dimensional single cell analysis of millions of CD8+ T cells from 53 individuals with lung cancer, we defined those subsets that are enriched in tumors compared with cancer-free tissues and blood. Besides exhausted and activated cells, we identified CXCR5+ TIM-3- CD8+ T cells with a partial exhausted phenotype, while retaining gene networks responsible for stem-like plasticity and cytotoxicity, as revealed by single cell sequencing of the whole transcriptome. Ex vivo, CXCR5+ TIM-3- CD8+ T cells displayed enhanced self-renewal and multipotency compared with more differentiated subsets and were largely polyfunctional. Analysis of inhibitory and costimulatory receptors revealed PD-1, TIGIT, and CD27 as possible targets of immunotherapy. We thus demonstrate a hierarchy of differentiation in the context of T cell exhaustion in human cancer similar to that of chronically infected mice, which is further shown to disappear with disease progression.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias Pulmonares/inmunología , Células Madre/inmunología , Animales , Linfocitos T CD8-positivos/patología , Femenino , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Proteínas de Neoplasias/inmunología , Receptores CXCR5/inmunología , Receptores Inmunológicos/inmunología , Células Madre/patología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
10.
Cancer Cell ; 30(3): 377-390, 2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27622331

RESUMEN

Effective cancer immunotherapy requires overcoming immunosuppressive tumor microenvironments. We found that local nitric oxide (NO) production by tumor-infiltrating myeloid cells is important for adoptively transferred CD8(+) cytotoxic T cells to destroy tumors. These myeloid cells are phenotypically similar to inducible nitric oxide synthase (NOS2)- and tumor necrosis factor (TNF)-producing dendritic cells (DC), or Tip-DCs. Depletion of immunosuppressive, colony stimulating factor 1 receptor (CSF-1R)-dependent arginase 1(+) myeloid cells enhanced NO-dependent tumor killing. Tumor elimination via NOS2 required the CD40-CD40L pathway. We also uncovered a strong correlation between survival of colorectal cancer patients and NOS2, CD40, and TNF expression in their tumors. Our results identify a network of pro-tumor factors that can be targeted to boost cancer immunotherapies.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/inmunología , Neoplasias/terapia , Óxido Nítrico Sintasa de Tipo II/inmunología , Linfocitos T Citotóxicos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Arginasa/biosíntesis , Arginasa/inmunología , Antígenos CD40/inmunología , Ligando de CD40/inmunología , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/biosíntesis
11.
Eur J Immunol ; 44(11): 3307-19, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25113564

RESUMEN

By restraining T-cell activation and promoting Treg-cell expansion, myeloid-derived suppressor cells (MDSCs) and tolerogenic DCs can control self-reactive and antigraft effector T cells in autoimmunity and transplantation. Their therapeutic use and characterization, however, is limited by their scarce availability in the peripheral blood of tumor-free donors. In the present study, we describe and characterize a novel population of human myeloid suppressor cells, named fibrocytic MDSC, which are differentiated from umbilical cord blood precursors by 4-day culture with FDA-approved cytokines (recombinant human-GM-CSF and recombinant human-G-CSF). This MDSC subset, characterized by the expression of MDSC-, DC-, and fibrocyte-associated markers, promotes Treg-cell expansion and induces normoglycemia in a xenogeneic mouse model of Type 1 diabetes. In order to exert their protolerogenic function, fibrocytic MDSCs require direct contact with activated T cells, which leads to the production and secretion of IDO. This new myeloid subset may have an important role in the in vitro and in vivo production of Treg cells for the treatment of autoimmune diseases, and in either the prevention or control of allograft rejection.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Activación de Linfocitos/inmunología , Células Mieloides/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular , Diabetes Mellitus Tipo 1/inmunología , Femenino , Sangre Fetal/citología , Perfilación de la Expresión Génica , Rechazo de Injerto/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células HEK293 , Humanos , Tolerancia Inmunológica , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Mieloides/citología , Proteínas Recombinantes/farmacología , Linfocitos T Reguladores/trasplante
12.
Nature ; 511(7508): 184-90, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24930766

RESUMEN

Disease tolerance is the ability of the host to reduce the effect of infection on host fitness. Analysis of disease tolerance pathways could provide new approaches for treating infections and other inflammatory diseases. Typically, an initial exposure to bacterial lipopolysaccharide (LPS) induces a state of refractoriness to further LPS challenge (endotoxin tolerance). We found that a first exposure of mice to LPS activated the ligand-operated transcription factor aryl hydrocarbon receptor (AhR) and the hepatic enzyme tryptophan 2,3-dioxygenase, which provided an activating ligand to the former, to downregulate early inflammatory gene expression. However, on LPS rechallenge, AhR engaged in long-term regulation of systemic inflammation only in the presence of indoleamine 2,3-dioxygenase 1 (IDO1). AhR-complex-associated Src kinase activity promoted IDO1 phosphorylation and signalling ability. The resulting endotoxin-tolerant state was found to protect mice against immunopathology in Gram-negative and Gram-positive infections, pointing to a role for AhR in contributing to host fitness.


Asunto(s)
Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Resistencia a la Enfermedad/efectos de los fármacos , Endotoxemia/genética , Endotoxemia/inmunología , Endotoxemia/metabolismo , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inflamación/enzimología , Inflamación/genética , Inflamación/metabolismo , Quinurenina/metabolismo , Lipopolisacáridos/farmacología , Ratones , Fosforilación , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal , Triptófano Oxigenasa/metabolismo , Familia-src Quinasas/metabolismo
13.
PLoS One ; 9(2): e87680, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498352

RESUMEN

Monocytes/macrophages are key players in all phases of physiological and pathological inflammation. To understanding the regulation of macrophage functional differentiation during inflammation, we designed an in vitro model that recapitulates the different phases of the reaction (recruitment, initiation, development, and resolution), based on human primary blood monocytes exposed to sequential changes in microenvironmental conditions. All reaction phases were profiled by transcriptomic microarray analysis. Distinct clusters of genes were identified that are differentially regulated through the different phases of inflammation. The gene sets defined by GSEA analysis revealed that the inflammatory phase was enriched in inflammatory pathways, while the resolution phase comprised pathways related to metabolism and gene rearrangement. By comparing gene clusters differentially expressed in monocytes vs. M1 and vs. M2 macrophages extracted from an in-house created meta-database, it was shown that cells in the model resemble M1 during the inflammatory phase and M2 during resolution. The validation of inflammatory and transcriptional factors by qPCR and ELISA confirmed the transcriptomic profiles in the different phases of inflammation. The accurate description of the development of the human inflammatory reaction provided by this in vitro kinetic model can help in identifying regulatory mechanisms in physiological conditions and during pathological derangements.


Asunto(s)
Diferenciación Celular/genética , Perfilación de la Expresión Génica , Macrófagos/metabolismo , Monocitos/metabolismo , Adulto , Células Cultivadas , Microambiente Celular/genética , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Análisis por Conglomerados , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Macrófagos/citología , Masculino , Persona de Mediana Edad , Monocitos/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Adulto Joven
14.
Biochim Biophys Acta ; 1843(5): 955-64, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24472656

RESUMEN

In spite of the numerous reports implicating MafB transcription factor in the molecular control of monocyte-macrophage differentiation, the precise genetic program underlying this activity has been, to date, poorly understood. To clarify this issue, we planned a number of experiments that were mainly conducted on human primary macrophages. In this regard, a preliminary gene function study, based on MafB inactivation and over-expression, indicated MMP9 and IL-7R genes as possible targets of the investigated transcription factor. Bioinformatics analysis of their promoter regions disclosed the presence of several putative MARE elements and a combined approach of EMSA and luciferase assay subsequently demonstrated that expression of both genes is indeed activated by MafB through a direct transcription mechanism. Additional investigation, performed with similar procedures to elucidate the biological relevance of our observation, revealed that MafB is a downstream target of the IL-10/STAT3 signaling pathway, normally inducing the macrophage de-activation process. Taken together our data support the existence of a signaling cascade by which stimulation of macrophages with the IL-10 cytokine determines a sequential activation of STAT3 and MafB transcription factors, in turn leading to an up-regulated expression of MMP9 and IL-7R genes.


Asunto(s)
Interleucina-10/metabolismo , Activación de Macrófagos , Factor de Transcripción MafB/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Secuencia de Bases , Línea Celular , Sondas de ADN , Silenciador del Gen , Humanos , Factor de Transcripción MafB/genética , Metaloproteinasa 7 de la Matriz/genética , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Receptores de Interleucina-7/genética
15.
Nat Immunol ; 12(9): 870-8, 2011 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-21804557

RESUMEN

Regulation of tryptophan metabolism by indoleamine 2,3-dioxygenase (IDO) in dendritic cells (DCs) is a highly versatile modulator of immunity. In inflammation, interferon-γ is the main inducer of IDO for the prevention of hyperinflammatory responses, yet IDO is also responsible for self-tolerance effects in the longer term. Here we show that treatment of mouse plasmacytoid DCs (pDCs) with transforming growth factor-ß (TGF-ß) conferred regulatory effects on IDO that were mechanistically separable from its enzymic activity. We found that IDO was involved in intracellular signaling events responsible for the self-amplification and maintenance of a stably regulatory phenotype in pDCs. Thus, IDO has a tonic, nonenzymic function that contributes to TGF-ß-driven tolerance in noninflammatory contexts.


Asunto(s)
Inmunidad Adaptativa , Células Dendríticas , Tolerancia Inmunológica , Indolamina-Pirrol 2,3,-Dioxigenasa , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Humanos , Hipersensibilidad/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Triptófano/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA