Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Theranostics ; 11(4): 1594-1608, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33408769

RESUMEN

The Notch pathway is highly active in almost all patients with T-cell acute lymphoblastic leukemia (T-ALL), but the implication of Notch ligands in T-ALL remains underexplored. Methods: We used a genetic mouse model of Notch ligand delta like 4 (DLL4)-driven T-ALL and performed thymectomies and splenectomies in those animals. We also used several patient-derived T-ALL (PDTALL) models, including one with DLL4 expression on the membrane and we treated PDTALL cells in vitro and in vivo with demcizumab, a blocking antibody against human DLL4 currently being tested in clinical trials in patients with solid cancer. Results: We show that surgical removal of the spleen abrogated T-ALL development in our preclinical DLL4-driven T-ALL mouse model. Mechanistically, we found that the spleen, and not the thymus, promoted the accumulation of circulating CD4+CD8+ T cells before T-ALL onset, suggesting that DLL4-driven T-ALL derives from these cells. Then, we identified a small subset of T-ALL patients showing higher levels of DLL4 expression. Moreover, in mice xenografted with a DLL4-positive PDTALL model, treatment with demcizumab had the same therapeutic effect as global Notch pathway inhibition using the potent γ-secretase inhibitor dibenzazepine. This result demonstrates that, in this PDTALL model, Notch pathway activity depends on DLL4 signaling, thus validating our preclinical mouse model. Conclusion: DLL4 expression in human leukemic cells can be a source of Notch activity in T-ALL, and the spleen plays a major role in a genetic mouse model of DLL4-driven T-ALL.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio/metabolismo , Regulación Neoplásica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptores Notch/metabolismo , Bazo/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Receptores Notch/genética , Bazo/metabolismo , Bazo/cirugía , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Immunother Cancer ; 8(2)2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33127657

RESUMEN

BACKGROUND: Cancer immunotherapy has evolved from interferon-alpha (IFNα) and interleukin-2 in the 1980s to CTLA-4 and PD-1/PD-L1 checkpoint inhibitors (CPIs), the latter highlighting the importance of enhancing T-cell functions. While the search for novel immunomodulatory pathways continues, combination therapies augmenting multiple pathways can also increase efficacy. The association of autoimmune-related adverse events with clinical efficacy following CPI treatment has been inferred and suggests that breaking tolerance thresholds associated with autoimmunity may affect host immune responses for effective cancer immunotherapy. RESULTS: Here, we show that loss of autoimmune associated PTPN22, a key desensitization node for multiple signaling pathways, including IFNα receptor (IFNAR) and T-cell receptor, can augment tumor responses. Implantation of syngeneic tumors in Ptpn22-/- mice led to expansion and activation of peripheral and intratumoral T cells and, in turn, spontaneous tumor regression as well as enhanced responses in combination with anti-PD-L1 treatment. Using genetically modified mice expressing a catalytically inactive PTPN22 or the autoimmunity-associated human single-nucleotide polymorphism variant, augmentation of antitumor immunity was dependent on PTPN22 phosphatase activity and partially on its adaptor functions. Further, antitumor responses were dependent on both CD4+ and CD8+T cells and, in part, IFNAR function. Finally, we demonstrate that the autoimmune susceptibility Ptpn22(C1858T) variant is associated with lower risk of developing non-melanoma skin cancers, improved overall survival and increased risk for development of hyperthyroidism or hypothyroidism following atezolizumab (anti-PD-L1) treatment. CONCLUSIONS: Together, these data suggest that inhibition of PTPN22 phosphatase activity may provide an effective therapeutic option for cancer immunotherapy and that exploring genetic variants that shift immune tolerance thresholds may serve as a paradigm for finding new cancer immunotherapy targets.


Asunto(s)
Autoinmunidad/genética , Inmunoterapia/métodos , Neoplasias/terapia , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo , Animales , Humanos , Masculino , Ratones
3.
Nat Cancer ; 1(7): 681-691, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-35122038

RESUMEN

Inhibiting the programmed death-1 (PD-1) pathway is one of the most effective approaches to cancer immunotherapy, but its mechanistic basis remains incompletely understood. Binding of PD-1 to its ligand PD-L1 suppresses T-cell function in part by inhibiting CD28 signaling. Tumor cells and infiltrating myeloid cells can express PD-L1, with myeloid cells being of particular interest as they also express B7-1, a ligand for CD28 and PD-L1. Here we demonstrate that dendritic cells (DCs) represent a critical source of PD-L1, despite being vastly outnumbered by PD-L1+ macrophages. Deletion of PD-L1 in DCs, but not macrophages, greatly restricted tumor growth and led to enhanced antitumor CD8+ T-cell responses. Our data identify a unique role for DCs in the PD-L1-PD-1 regulatory axis and have implications for understanding the therapeutic mechanism of checkpoint blockade, which has long been assumed to reflect the reversal of T-cell exhaustion induced by PD-L1+ tumor cells.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Antígeno B7-H1/genética , Antígenos CD28/metabolismo , Células Dendríticas , Humanos , Ligandos , Neoplasias/genética , Receptor de Muerte Celular Programada 1/genética
4.
Cancer Immunol Res ; 7(6): 963-976, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31064777

RESUMEN

Exhausted T cells have been described in cancer patients and murine tumor models largely based on their expression of various inhibitory receptors. Understanding of the functional attributes of these cells is limited. Here, we report that among CD8+ T cells in commonly used syngeneic tumor models, the coexpression of inhibitory receptors PD-1, LAG3, and TIM3 defined a group of highly activated and functional effector cells. Coexpression of these receptors further enriched for antigen-specific cells with increased T-cell receptor clonality. Anti-PD-L1 treatment increased the number and activation of these triple-positive CD8+ T cells without affecting the density of PD-1- cells. The intratumoral density of CD8+ T cells coexpressing inhibitory receptors negatively correlated with tumor burden. The density ratio and pretreatment phenotype of CD8+ T cells coexpressing inhibitory receptors was positively correlated with response across a variety of tumor models. Our results demonstrate that coexpression of inhibitory receptors is not a signifier of exhausted T cells, but rather can define a group of activated and functional effector cells in syngeneic tumor models. In the cancer setting, these cells could represent a heterogeneous population of not only exhausted but also highly activated cells responsive to treatment.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptores Coestimuladores e Inhibidores de Linfocitos T/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Antígeno B7-H1/antagonistas & inhibidores , Biomarcadores de Tumor , Línea Celular Tumoral , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Isoinjertos , Ratones , Neoplasias/patología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
5.
Cancer Res ; 79(7): 1493-1506, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30679180

RESUMEN

Checkpoint inhibitors like anti-PD1/PD-L1 have demonstrated significant therapeutic efficacy in a subset of patients partly through reinvigoration of CD8 T cells. However, their impact on myeloid cells remains largely unknown. Here, we report that anti-PD-L1 treatment favorably impacts the phenotype and function of tumor macrophages by polarizing the macrophage compartment toward a more proinflammatory phenotype. This phenotype was characterized by a decrease in Arginase-I (ARG1) expression and an increase in iNOS, MHCII, and CD40 expression. Whole-transcriptome profiling further confirmed extensive polarization of both tumor monocytes and macrophages from a suppressive to a proinflammatory, immunostimulatory phenotype. This polarization was driven mainly through IFNγ and was associated with enhanced T-cell activity. Transfer of monocytes into anti-PD-L1-treated tumor-bearing mice led to macrophage differentiation into a more proinflammatory phenotype, with an increase in CD8 T cells expressing granzyme-B and an increase in the CD8/Treg ratio compared with control-treated mice. Although in responsive tumor models, anti-PD-L1 treatment remodeled the macrophage compartment with beneficial effects on T cells, both macrophage reprogramming and depletion were needed to maximize anti-PD-L1 responses in a tumor immune contexture with high macrophage burden. Our results demonstrate that anti-PD-L1 treatment can favorably remodel the macrophage compartment in responsive tumor models toward a more proinflammatory phenotype, mainly through increased IFNγ levels. They also suggest that directly targeting these cells with reprogramming and depleting agents may further augment the breadth and depth of response to anti-PD-L1 treatment in less responsive or more macrophage-dense tumor microenvironments. SIGNIFICANCE: This work demonstrates that increased IFNγ signaling following anti-PD-L1 treatment can remodel the macrophage compartment to enhance T-cell responses.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/7/1493/F1.large.jpg.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Macrófagos/metabolismo , Neoplasias/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Polaridad Celular , Proliferación Celular , Femenino , Humanos , Interferón gamma/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Microambiente Tumoral
6.
J Immunol ; 201(8): 2273-2286, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30209192

RESUMEN

Immunotherapy with checkpoint inhibitors has proved to be highly effective, with durable responses in a subset of patients. Given their encouraging clinical activity, checkpoint inhibitors are increasingly being tested in clinical trials in combination with chemotherapy. In many instances, there is little understanding of how chemotherapy might influence the quality of the immune response generated by checkpoint inhibitors. In this study, we evaluated the impact of chemotherapy alone or in combination with anti-PD-L1 in a responsive syngeneic tumor model. Although multiple classes of chemotherapy treatment reduced immune cell numbers and activity in peripheral tissues, chemotherapy did not antagonize but in many cases augmented the antitumor activity mediated by anti-PD-L1. This dichotomy between the detrimental effects in peripheral tissues and enhanced antitumor activity was largely explained by the reduced dependence on incoming cells for antitumor efficacy in already established tumors. The effects of the various chemotherapies were also agent specific, and synergy with anti-PD-L1 was achieved by different mechanisms that ultimately helped establish a new threshold for response. These results rationalize the combination of chemotherapy with immunotherapy and suggest that, despite the negative systemic effects of chemotherapy, effective combinations can be obtained through distinct mechanisms acting within the tumor.


Asunto(s)
Adenocarcinoma/terapia , Anticuerpos Monoclonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfocitos T CD8-positivos/efectos de los fármacos , Neoplasias del Colon/terapia , Inmunoterapia/métodos , Adenocarcinoma/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Femenino , Citometría de Flujo , Humanos , Inmunidad Celular , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Carga Tumoral/efectos de los fármacos
7.
Cancer Res ; 77(22): 6400-6414, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28951461

RESUMEN

Perineural invasion (PNI) is an ominous event strongly linked to poor clinical outcome. Cells residing within peripheral nerves collaborate with cancer cells to enable PNI, but the contributing conditions within the tumor microenvironment are not well understood. Here, we show that CCR2-expressing inflammatory monocytes (IM) are preferentially recruited to sites of PNI, where they differentiate into macrophages and potentiate nerve invasion through a cathepsin B-mediated process. A series of adoptive transfer experiments with genetically engineered donors and recipients demonstrated that IM recruitment to nerves was driven by CCL2 released from Schwann cells at the site of PNI, but not CCL7, an alternate ligand for CCR2. Interruption of either CCL2-CCR2 signaling or cathepsin B function significantly impaired PNI in vivo Correlative studies in human specimens demonstrated that cathepsin B-producing macrophages were enriched in invaded nerves, which was associated with increased local tumor recurrence. These findings deepen our understanding of PNI pathogenesis and illuminate how PNI is driven in part by corruption of a nerve repair program. Further, they support the exploration of inhibiting IM recruitment and function as a targeted therapy for PNI. Cancer Res; 77(22); 6400-14. ©2017 AACR.


Asunto(s)
Catepsina B/metabolismo , Quimiocina CCL2/metabolismo , Monocitos/metabolismo , Neoplasias Pancreáticas/metabolismo , Nervios Periféricos/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Quimiocina CCL2/genética , Humanos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Monocitos/patología , Invasividad Neoplásica , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Nervios Periféricos/patología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Células de Schwann/metabolismo , Trasplante Heterólogo
8.
Cell ; 165(3): 679-89, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27040495

RESUMEN

Increasing antibiotic resistance among bacterial pathogens has rendered some infections untreatable with available antibiotics. Klebsiella pneumoniae, a bacterial pathogen that has acquired high-level antibiotic resistance, is a common cause of pulmonary infections. Optimal clearance of K. pneumoniae from the host lung requires TNF and IL-17A. Herein, we demonstrate that inflammatory monocytes are rapidly recruited to the lungs of K. pneumoniae-infected mice and produce TNF, which markedly increases the frequency of IL-17-producing innate lymphoid cells. While pulmonary clearance of K. pneumoniae is preserved in neutrophil-depleted mice, monocyte depletion or TNF deficiency impairs IL-17A-dependent resolution of pneumonia. Monocyte-mediated bacterial uptake and killing is enhanced by ILC production of IL-17A, indicating that innate lymphocytes engage in a positive-feedback loop with monocytes that promotes clearance of pneumonia. Innate immune defense against a highly antibiotic-resistant bacterial pathogen depends on crosstalk between inflammatory monocytes and innate lymphocytes that is mediated by TNF and IL-17A.


Asunto(s)
Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/fisiología , Animales , Inflamación/inmunología , Interleucina-17/inmunología , Infecciones por Klebsiella/microbiología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Linfocitos/inmunología , Ratones , Monocitos/inmunología , Factor de Necrosis Tumoral alfa/inmunología
9.
Nat Immunol ; 17(4): 379-86, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26901151

RESUMEN

The T cell antigen receptor (TCR) is unique in that its affinity for ligand is unknown before encounter and can vary by orders of magnitude. How the immune system regulates individual T cells that display very different reactivity to antigen remains unclear. Here we found that activated CD4(+) T cells, at the peak of clonal expansion, persistently downregulated their TCR expression in proportion to the strength of the initial antigen recognition. This programmed response increased the threshold for cytokine production and recall proliferation in a clone-specific manner and ultimately excluded clones with the highest antigen reactivity. Thus, programmed downregulation of TCR expression represents a negative feedback mechanism for constraining T cell effector function with a suitable time delay to thereby allow pathogen control while avoiding excess inflammatory damage.


Asunto(s)
Regulación hacia Abajo , Listeriosis/inmunología , Receptores de Antígenos de Linfocitos T/genética , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Immunoblotting , Listeria monocytogenes , Activación de Linfocitos , Ratones , Ratones Transgénicos , Mycobacterium tuberculosis , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T , Transcriptoma
10.
Cell Host Microbe ; 18(1): 27-37, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26159718

RESUMEN

Infection with the opportunistic enteric pathogen Clostridium difficile is an increasingly common clinical complication that follows antibiotic treatment-induced gut microbiota perturbation. Innate lymphoid cells (ILCs) are early responders to enteric pathogens; however, their role during C. difficile infection is undefined. To identify immune pathways that mediate recovery from C. difficile infection, we challenged C57BL/6, Rag1(-/-) (which lack T and B cells), and Rag2(-/-)Il2rg(-/-) (Ragγc(-/-)) mice (which additionally lack ILCs) with C. difficile. In contrast to Rag1(-/-) mice, ILC-deficient Ragγc(-/-) mice rapidly succumbed to infection. Rag1(-/-) but not Ragγc(-/-) mice upregulate expression of ILC1- or ILC3-associated proteins following C. difficile infection. Protection against infection was restored by transferring ILCs into Ragγc(-/-) mice. While ILC3s made a minor contribution to resistance, loss of IFN-γ or T-bet-expressing ILC1s in Rag1(-/-) mice increased susceptibility to C. difficile. These data demonstrate a critical role for ILC1s in defense against C. difficile.


Asunto(s)
Clostridioides difficile/inmunología , Infecciones por Clostridium/inmunología , Resistencia a la Enfermedad , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Animales , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia
11.
Infect Immun ; 83(9): 3418-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26056382

RESUMEN

Klebsiella pneumoniae is a common respiratory pathogen, with some strains having developed broad resistance to clinically available antibiotics. Humans can become infected with many different K. pneumoniae strains that vary in genetic background, antibiotic susceptibility, capsule composition, and mucoid phenotype. Genome comparisons have revealed differences between K. pneumoniae strains, but the impact of genomic variability on immune-mediated clearance of pneumonia remains unclear. Experimental studies of pneumonia in mice have used the rodent-adapted 43816 strain of K. pneumoniae and demonstrated that neutrophils are essential for optimal host defense. It remains unclear, however, whether CCR2(+) monocytes contribute to K. pneumoniae clearance from the lung. We selectively depleted neutrophils, CCR2(+) monocytes, or both from immunocompetent mice and determined susceptibility to infection by the 43816 strain and 4 newly isolated clinical K. pneumoniae strains. The clinical K. pneumoniae strains, including one carbapenem-resistant ST258 strain, are less virulent than 43816. Optimal clearance of each of the 5 strains required either neutrophils or CCR2(+) monocytes. Selective neutrophil depletion markedly worsened infection with K. pneumoniae strain 43816 and three clinical isolates but did not increase susceptibility of mice to infection with the carbapenem-resistant K. pneumoniae ST258 strain. Depletion of CCR2(+) monocytes delayed recovery from infection with each of the 5 K. pneumoniae strains, revealing a contribution of these cells to bacterial clearance from the lung. Our findings demonstrate strain-dependent variation in the contributions of neutrophils and CCR2(+) monocytes to clearance of K. pneumoniae pulmonary infection.


Asunto(s)
Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/inmunología , Monocitos/inmunología , Neutrófilos/inmunología , Infecciones del Sistema Respiratorio/microbiología , Animales , Modelos Animales de Enfermedad , Infecciones por Klebsiella/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/inmunología , Infecciones del Sistema Respiratorio/inmunología
12.
Immunobiology ; 220(2): 210-4, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25214476

RESUMEN

Monocytes are a subset of circulating blood cells with remarkable plasticity. They can develop into a wide range of terminally differentiated cells and perform versatile functions during infection, tumor formation and in the setting of chronic inflammation. This review focuses on the role of monocytes during microbial infection and summarizes our understanding of the diverse roles that monocytes play in defense against different pathogens.


Asunto(s)
Inflamación/inmunología , Inflamación/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos/inmunología , Quimiotaxis/inmunología , Humanos , Inmunidad Innata , Inmunomodulación , Inflamación/microbiología , Monocitos/microbiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Necrosis Tumoral/metabolismo
13.
Nature ; 510(7503): 152-6, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24739972

RESUMEN

T-helper-17 (TH17) cells have critical roles in mucosal defence and in autoimmune disease pathogenesis. They are most abundant in the small intestine lamina propria, where their presence requires colonization of mice with microbiota. Segmented filamentous bacteria (SFB) are sufficient to induce TH17 cells and to promote TH17-dependent autoimmune disease in animal models. However, the specificity of TH17 cells, the mechanism of their induction by distinct bacteria, and the means by which they foster tissue-specific inflammation remain unknown. Here we show that the T-cell antigen receptor (TCR) repertoire of intestinal TH17 cells in SFB-colonized mice has minimal overlap with that of other intestinal CD4(+) T cells and that most TH17 cells, but not other T cells, recognize antigens encoded by SFB. T cells with antigen receptors specific for SFB-encoded peptides differentiated into RORγt-expressing TH17 cells, even if SFB-colonized mice also harboured a strong TH1 cell inducer, Listeria monocytogenes, in their intestine. The match of T-cell effector function with antigen specificity is thus determined by the type of bacteria that produce the antigen. These findings have significant implications for understanding how commensal microbiota contribute to organ-specific autoimmunity and for developing novel mucosal vaccines.


Asunto(s)
Antígenos Bacterianos/inmunología , Bacterias Grampositivas/inmunología , Intestinos/inmunología , Simbiosis , Células Th17/inmunología , Animales , Antígenos Bacterianos/química , Vacunas Bacterianas , Diferenciación Celular , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Bacterias Grampositivas/química , Hibridomas/inmunología , Inmunidad Mucosa/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestino Delgado/citología , Intestino Delgado/inmunología , Intestinos/citología , Listeria monocytogenes/inmunología , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Células Th17/citología
14.
J Exp Med ; 210(12): 2755-71, 2013 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-24218137

RESUMEN

The mechanisms involved in the maintenance of memory IgE responses are poorly understood, and the role played by germinal center (GC) IgE(+) cells in memory responses is particularly unclear. IgE(+) B cell differentiation is characterized by a transient GC phase, a bias toward the plasma cell (PC) fate, and dependence on sequential switching for the production of high-affinity IgE. We show here that IgE(+) GC B cells are unfit to undergo the conventional GC differentiation program due to impaired B cell receptor function and increased apoptosis. IgE(+) GC cells fail to populate the GC light zone and are unable to contribute to the memory and long-lived PC compartments. Furthermore, we demonstrate that direct and sequential switching are linked to distinct B cell differentiation fates: direct switching generates IgE(+) GC cells, whereas sequential switching gives rise to IgE(+) PCs. We propose a comprehensive model for the generation and memory of IgE responses.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulina E/metabolismo , Memoria Inmunológica , Modelos Inmunológicos , Animales , Apoptosis , Linfocitos B/citología , Diferenciación Celular , Centro Germinal/citología , Centro Germinal/inmunología , Proteínas Fluorescentes Verdes/genética , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Nippostrongylus , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Infecciones por Strongylida/inmunología
15.
PLoS One ; 8(12): e84841, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24386421

RESUMEN

Notch signaling is essential for the development of T cell progenitors through the interaction of NOTCH1 receptor on their surface with the ligand, Delta-like 4 (DLL4), which is expressed by the thymic epithelial cells. Notch signaling is quickly shut down once the cells pass ß-selection, and CD4/CD8 double positive (DP) cells are unresponsive to Notch. Over the past two decades a number of papers reported that over-activation of Notch signaling causes T cell acute lymphoblastic leukemia (T-ALL), a cancer that prominently features circulating monoclonal CD4/CD8 double positive T cells in different mouse models. However, the possible outcomes of Notch over-activation at different stages of T cell development are unknown, and the fine timing of Notch signaling that results in T-ALL is poorly understood. Here we report, by using a murine model that ectopically expresses DLL4 on developing T cells, that the T-ALL onset is highly dependent on a sustained Notch activity throughout the DP stage, which induces additional mutations to further boost the signaling. In contrast, a shorter period of Notch activation that terminates at the DP stage causes a polyclonal, non-transmissible lymphoproliferative disorder that is also lethal. These observations resolved the discrepancy of previous papers on DLL4 driven hematological diseases in mice, and show the critical importance of the timing and duration of Notch activity.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Regulación Leucémica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteínas de la Membrana/biosíntesis , Neoplasias Experimentales/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Proteínas de Unión al Calcio , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptores Notch/genética , Receptores Notch/metabolismo
16.
Adv Immunol ; 116: 113-41, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23063075

RESUMEN

IgE antibodies are involved in allergic reactions. High affinity IgE antibodies can cause anaphylaxis when cross-linked by minute amounts of antigen. The issue of how the IgE response is initiated and maintained is addressed in this review. A model has been proposed by which IgE(+) cells expressing antibodies that bind with high affinity to their antigens are generated through an IgG1 intermediate, which goes through affinity maturation in germinal centers (GC) before undergoing sequential switching to IgE. Mice deficient in IgG1 produce IgE at almost normal levels, but the IgE antibodies produced in IgG1-deficient mice lack the antigen-binding strength and the somatic mutations associated with affinity maturation. A GFP reporter strain, which expresses a modified IgE molecule, was recently developed and was utilized to challenge the sequential switching model. Several molecules that are highly expressed in GC can antagonize class switching to IgE in GC antagonize partially class switching to IgE; in addition, GC IgE(+) cells are gradually lost from GC as the immune response progresses, as shown with another recently developed, Venus-expressing IgE reporter mouse strain. In contrast, as a population, IgG1 cells thrive in the GC environment. Membrane IgE-expressing plasmablasts and plasma cells (PC) were recognized as a major component of the IgE response in secondary lymphoid organs. The swift development of IgE cells toward the PC fate, together with the affinity maturation of the IgE response via an IgG intermediate, represent the most salient features of the IgE immune responses, which make them distinct from IgG responses.


Asunto(s)
Hipersensibilidad/inmunología , Inmunoglobulina E/inmunología , Anafilaxia/inmunología , Animales , Humanos , Cambio de Clase de Inmunoglobulina , Inmunoglobulina E/genética , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Células Plasmáticas/inmunología
17.
J Exp Med ; 209(10): 1723-42, S1, 2012 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-22966001

RESUMEN

Foxp3 activity is essential for the normal function of the immune system. Two types of regulatory T (T reg) cells express Foxp3, thymus-generated natural T reg (nT reg) cells, and peripherally generated adaptive T reg (iT reg) cells. These cell types have complementary functions. Until now, it has not been possible to distinguish iT reg from nT reg cells in vivo based solely on surface markers. We report here that Neuropilin 1 (Nrp1) is expressed at high levels by most nT reg cells; in contrast, mucosa-generated iT reg and other noninflammatory iT reg cells express low levels of Nrp1. We found that Nrp1 expression is under the control of TGF-ß. By tracing nT reg and iT reg cells, we could establish that some tumors have a very large proportion of infiltrating iT reg cells. iT reg cells obtained from highly inflammatory environments, such as the spinal cords of mice with spontaneous autoimmune encephalomyelitis (EAE) and the lungs of mice with chronic asthma, express Nrp1. In the same animals, iT reg cells in secondary lymphoid organs remain Nrp1(low). We also determined that, in spontaneous EAE, iT reg cells help to establish a chronic phase of the disease.


Asunto(s)
Membrana Mucosa/inmunología , Neuropilina-1/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Timo/inmunología , Animales , Linaje de la Célula , Membrana Celular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/inmunología , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/microbiología , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Metagenoma/inmunología , Ratones , Ratones Transgénicos , Membrana Mucosa/metabolismo , Neuropilina-1/genética , Timo/metabolismo , Factor de Crecimiento Transformador beta/farmacología
19.
J Exp Med ; 209(2): 353-64, 2012 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-22249450

RESUMEN

IgE antibodies with high affinity for their antigens can be stably cross-linked at low concentrations by trace amounts of antigen, whereas IgE antibodies with low affinity bind their antigens weakly. In this study, we find that there are two distinct pathways to generate high and low affinity IgE. High affinity IgE is generated through sequential class switching (µâ†’γ→ε) in which an intermediary IgG phase is necessary for the affinity maturation of the IgE response, where the IgE inherits somatic hypermutations and high affinity from the IgG1 phase. In contrast, low affinity IgE is generated through direct class switching (µâ†’ε) and is much less mutated. Mice deficient in IgG1 production cannot produce high affinity IgE, even after repeated immunizations. We demonstrate that a small amount of high affinity IgE can cause anaphylaxis and is pathogenic. Low affinity IgE competes with high affinity IgE for binding to Fcε receptors and prevents anaphylaxis and is thus beneficial.


Asunto(s)
Anafilaxia/inmunología , Afinidad de Anticuerpos/inmunología , Cambio de Clase de Inmunoglobulina/fisiología , Inmunoglobulina E/biosíntesis , Animales , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Receptores de IgE/inmunología , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...