Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 449
Filtrar
1.
Mucosal Immunol ; 9(4): 859-72, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26555706

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a life-threatening inflammatory respiratory disorder, often induced by cigarette smoke (CS) exposure. The development of effective therapies is impaired by a lack of understanding of the underlining mechanisms. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine with inflammatory and apoptotic properties. We interrogated a mouse model of CS-induced experimental COPD and human tissues to identify a novel role for TRAIL in COPD pathogenesis. CS exposure of wild-type mice increased TRAIL and its receptor messenger RNA (mRNA) expression and protein levels, as well as the number of TRAIL(+)CD11b(+) monocytes in the lung. TRAIL and its receptor mRNA were also increased in human COPD. CS-exposed TRAIL-deficient mice had decreased pulmonary inflammation, pro-inflammatory mediators, emphysema-like alveolar enlargement, and improved lung function. TRAIL-deficient mice also developed spontaneous small airway changes with increased epithelial cell thickness and collagen deposition, independent of CS exposure. Importantly, therapeutic neutralization of TRAIL, after the establishment of early-stage experimental COPD, reduced pulmonary inflammation, emphysema-like alveolar enlargement, and small airway changes. These data provide further evidence for TRAIL being a pivotal inflammatory factor in respiratory diseases, and the first preclinical evidence to suggest that therapeutic agents that target TRAIL may be effective in COPD therapy.


Asunto(s)
Inflamación/inmunología , Pulmón/inmunología , Monocitos/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , ARN Mensajero/genética , Mucosa Respiratoria/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Apoptosis , Modelos Animales de Enfermedad , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fumar/efectos adversos , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Regulación hacia Arriba
2.
Leukemia ; 29(3): 576-85, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25234168

RESUMEN

In leukemogenesis, Notch signaling can be up and downregulated in a context-dependent manner. The transcription factor hairy and enhancer of split-1 (Hes1) is well-characterized as a downstream target of Notch signaling. Hes1 encodes a basic helix-loop-helix-type protein, and represses target gene expression. Here, we report that deletion of the Hes1 gene in mice promotes acute myeloid leukemia (AML) development induced by the MLL-AF9 fusion protein. We then found that Hes1 directly bound to the promoter region of the FMS-like tyrosine kinase 3 (FLT3) gene and downregulated the promoter activity. FLT3 was consequently upregulated in MLL-AF9-expressing immortalized and leukemia cells with a Hes1- or RBPJ-null background. MLL-AF9-expressing Hes1-null AML cells showed enhanced proliferation and ERK phosphorylation following FLT3 ligand stimulation. FLT3 inhibition efficiently abrogated proliferation of MLL-AF9-induced Hes1-null AML cells. Furthermore, an agonistic anti-Notch2 antibody induced apoptosis of MLL-AF9-induced AML cells in a Hes1-wild type but not a Hes1-null background. We also accessed two independent databases containing messenger RNA (mRNA) expression profiles and found that the expression level of FLT3 mRNA was negatively correlated with those of HES1 in patient AML samples. These observations demonstrate that Hes1 mediates tumor suppressive roles of Notch signaling in AML development, probably by downregulating FLT3 expression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/genética , Leucemia Mieloide Aguda/genética , Tirosina Quinasa 3 Similar a fms/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Proliferación Celular , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Ratones , Ratones Transgénicos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Análisis de Supervivencia , Factor de Transcripción HES-1 , Tirosina Quinasa 3 Similar a fms/metabolismo
3.
Leukemia ; 29(1): 145-56, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24791857

RESUMEN

The systemic inflammatory response observed during acute graft-versus-host disease (aGVHD) is driven by proinflammatory cytokines, a 'cytokine storm'. The function of plasmin in regulating the inflammatory response is not fully understood, and its role in the development of aGVHD remains unresolved. Here we show that plasmin is activated during the early phase of aGVHD in mice, and its activation correlated with aGVHD severity in humans. Pharmacological plasmin inhibition protected against aGVHD-associated lethality in mice. Mechanistically, plasmin inhibition impaired the infiltration of inflammatory cells, the release of membrane-associated proinflammatory cytokines including tumor necrosis factor-α (TNF-α) and Fas-ligand directly, or indirectly via matrix metalloproteinases (MMPs) and alters monocyte chemoattractant protein-1 (MCP-1) signaling. We propose that plasmin and potentially MMP-9 inhibition offers a novel therapeutic strategy to control the deadly cytokine storm in patients with aGVHD, thereby preventing tissue destruction.


Asunto(s)
Fibrinolisina/antagonistas & inhibidores , Enfermedad Injerto contra Huésped/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Secuencia de Bases , Transporte Biológico , Línea Celular , Cartilla de ADN , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Enfermedad Injerto contra Huésped/enzimología , Enfermedad Injerto contra Huésped/mortalidad , Humanos , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Índice de Severidad de la Enfermedad
4.
Mucosal Immunol ; 7(5): 1199-208, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24569802

RESUMEN

Allergic asthma can vanish over time either spontaneously or induced by allergen-specific immunotherapy. In mice with established airway allergic inflammation, chronic intranasal (IN) allergen challenges decreases progressively airway allergic inflammation. Here we compared the contribution of different regulatory pathways that could be associated with this phenomenon, known as local inhalational tolerance. We found that inhalational tolerance was not associated with increased number of regulatory T cells or suppressive cytokines. Instead, it was associated with increased apoptosis of airway inflammatory leukocytes revealed by annexin-V staining and the expression of apical caspase 8 and effector caspase 3. Also, the transition from acute to chronic phase was associated with a shift in the expression of pro-allergic to pro-apoptotic molecules. The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) was found to be a key molecule in mediating resolution of allergic inflammation because anti-TRAIL treatment blocked apoptosis and increased the infiltration of T helper type 2 (Th2) cells and eosinophils. Notably, repeated IN treatment with recombinant TRAIL in established airway allergic inflammation augmented leukocyte apoptosis and decreased the frequency of interleukin-5-producing Th2 cells and eosinophils to airways. Our data indicate that TRAIL signaling is sufficient for downmodulation of allergic airway disease, suggesting a potential therapeutic use of TRAIL for asthma treatment.


Asunto(s)
Alérgenos/inmunología , Hipersensibilidad Respiratoria/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Apoptosis/inmunología , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica/inmunología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Pulmón/inmunología , Pulmón/fisiopatología , Ratones , Ratones Noqueados , Proteínas Recombinantes/genética , Hipersensibilidad Respiratoria/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Células Th2/inmunología
5.
Mucosal Immunol ; 7(4): 786-801, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24220301

RESUMEN

The vitamin A (VA) metabolite retinoic acid (RA) affects the properties of T cells and dendritic cells (DCs). In VA-deficient mice, we observed that mesenteric lymph node (MLN)-DCs induce a distinct inflammatory T helper type 2 (Th2)-cell subset that particularly produces high levels of interleukin (IL)-13 and tumor necrosis factor-α (TNF-α). This subset expressed homing receptors for skin and inflammatory sites, and was mainly induced by B220(-)CD8α(-)CD11b(+)CD103(-) MLN-DCs in an IL-6- and OX40 ligand-dependent manner, whereas RA inhibited this induction. The corresponding MLN-DC subset of VA-sufficient mice induced a similar T-cell subset in the presence of RA receptor antagonists. IL-6 induced this subset differentiation from naive CD4(+) T cells upon activation with antibodies against CD3 and CD28. Transforming growth factor-ß inhibited this induction, and reciprocally enhanced Th17 induction. Treatment with an agonistic anti-OX40 antibody and normal MLN-DCs enhanced the induction of general inflammatory Th2 cells. In VA-deficient mice, proximal colon epithelial cells produced TNF-α that may have enhanced OX40 ligand expression in MLN-DCs. The repeated oral administrations of a T cell-dependent antigen primed VA-deficient mice for IL-13-dependent strong immunoglobulin G1 (IgG1) responses and IgE responses that caused skin allergy. These results suggest that RA inhibits allergic responses to oral antigens by preventing MLN-DCs from inducing IL-13-producing inflammatory Th2 cells.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Interleucina-13/biosíntesis , Ganglios Linfáticos/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Tretinoina/farmacología , Administración Oral , Animales , Antígenos/administración & dosificación , Antígenos/inmunología , Ligando de CD40/metabolismo , Diferenciación Celular/inmunología , Colon/inmunología , Colon/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Isotipos de Inmunoglobulinas/inmunología , Inmunofenotipificación , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mesenterio/inmunología , Mesenterio/metabolismo , Ratones , Fenotipo , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Células Th2/citología , Factor de Necrosis Tumoral alfa/metabolismo , Deficiencia de Vitamina A/inmunología , Deficiencia de Vitamina A/metabolismo
6.
Mucosal Immunol ; 7(3): 478-88, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24045576

RESUMEN

Respiratory infections in early life can lead to chronic respiratory disease. Chlamydia infections are common causes of respiratory disease, particularly pneumonia in neonates, and are linked to permanent reductions in pulmonary function and the induction of asthma. However, the immune responses that protect against early-life infection and the mechanisms that lead to chronic lung disease are incompletely understood. Here we identify novel roles for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in promoting Chlamydia respiratory infection-induced pathology in early life, and subsequent chronic lung disease. By infecting TRAIL-deficient neonatal mice and using neutralizing antibodies against this factor and its receptors in wild-type mice, we demonstrate that TRAIL is critical in promoting infection-induced histopathology, inflammation, and mucus hypersecretion, as well as subsequent alveolar enlargement and impaired lung function. This suggests that therapeutic agents that target TRAIL or its receptors may be effective treatments for early-life respiratory infections and associated chronic lung disease.


Asunto(s)
Neumonía/metabolismo , Infecciones del Sistema Respiratorio/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes/farmacología , Apoptosis/genética , Infecciones por Chlamydia/metabolismo , Chlamydia muridarum , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica , Ratones , Ratones Noqueados , Moco/metabolismo , FN-kappa B/metabolismo , Neumonía/genética , Neumonía/microbiología , Neumonía/patología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/metabolismo , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/microbiología , Ligando Inductor de Apoptosis Relacionado con TNF/deficiencia , Ligando Inductor de Apoptosis Relacionado con TNF/genética
8.
Clin Exp Immunol ; 172(3): 500-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23600839

RESUMEN

Recent basic and clinical studies have shown that the programmed death ligand (PD-L)/PD-1 pathway has a significant role in tumour immunity, and its blockade has a therapeutic potential against several human cancers. We hypothesized that anti-angiogeneic treatment might augment the efficacy of PD-1 blockade. To this end, we evaluated combining the blockade of PD-1 and vascular endothelial growth factor receptor 2 (VEGFR2) in a murine cancer model using Colon-26 adenocarcinoma. Interestingly, simultaneous treatment with anti-PD-1 and anti-VEGFR2 monoclonal antibodies (mAbs) inhibited tumour growth synergistically in vivo without overt toxicity. Blocking VEGFR2 inhibited tumour neovascularization significantly, as demonstrated by the reduced number of microvessels, while PD-1 blockade had no impact on tumour angiogenesis. PD-1 blockade might promote T cell infiltration into tumours and significantly enhanced local immune activation, as shown by the up-regulation of several proinflammatory cytokine expressions. Importantly, VEGFR2 blockade did not interfere with T cell infiltration and immunological activation induced by PD-1 blockade. In conclusion, simultaneous blockade of PD-1 and VEGFR2 induced a synergistic in-vivo anti-tumour effect, possibly through different mechanisms that might not be mutually exclusive. This unique therapeutic strategy may hold significant promise for future clinical application.


Asunto(s)
Neoplasias Experimentales/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/inmunología , Adenocarcinoma/terapia , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Anticuerpos Monoclonales/administración & dosificación , Línea Celular Tumoral , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Sinergismo Farmacológico , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/inmunología , Neovascularización Patológica/prevención & control , Receptor de Muerte Celular Programada 1/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
9.
Oncogene ; 32(13): 1670-81, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-22665066

RESUMEN

Allogeneic stem cell transplantation (allo-SCT) is a potentially curative therapy for chronic myeloid leukemia and Philadelphia chromosome-positive (Ph(+)) acute lymphoblastic leukemia, and the graft-vs-leukemia (GVL) effect can eradicate residual leukemia after allo-SCT. Ph(+) leukemia cells frequently express death-inducing receptors (DR4 and DR5) for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), which is one of the cytotoxic ligands expressed on cytotoxic T cells and natural killer cells mediating the GVL effect. Here we demonstrate that imatinib specifically downregulated DR4 and DR5 expression in cell lines and clinical samples of Ph(+) leukemia. Second-generation tyrosine kinase inhibitors (dasatinib and nilotinib) and short hairpin RNA against bcr-abl also downregulated DR4 and DR5 expression in Ph(+) leukemia cells, and transfection of bcr-abl into a Ph(-) leukemia cell line induced DR4 and DR5 expression, which was abrogated by imatinib treatment. Accordingly, Ph(+) leukemia cells that had been pretreated with imatinib showed resistance to the pro-apoptotic activity of recombinant human soluble TRAIL. These observations demonstrate that BCR-ABL is critically involved in the leukemia-specific expression of DR4 and DR5 and in the susceptibility of Ph(+) leukemia to TRAIL-mediated anti-leukemic activity, providing new insight into the mechanisms of the tumor-specific cytotoxic activities of TRAIL.


Asunto(s)
Proteínas de Fusión bcr-abl/fisiología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Receptores de Muerte Celular/genética , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Línea Celular Tumoral , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Cromosoma Filadelfia , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/farmacología , Receptores de Muerte Celular/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
10.
Mucosal Immunol ; 6(4): 728-39, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23149662

RESUMEN

Studies examining the role of programmed death 1 (PD-1) ligand 2 (PD-L2)/PD-1 in asthma have yielded conflicting results. To clarify its role, we examined the PD-L2 expression in biopsies from human asthmatics and the lungs of aeroallergen-treated mice. PD-L2 expression in bronchial biopsies correlated with the severity of asthma. In mice, allergen exposure increased PD-L2 expression on pulmonary myeloid dendritic cells (DCs), and PD-L2 blockade diminished allergen-induced airway hyperresponsiveness (AHR). By contrast, PD-1 blockade had no impact, suggesting that PD-L2 promotes AHR in a PD-1-independent manner. Decreased AHR was associated with enhanced serum interleukin (IL)-12 p40, and in vitro stimulation of DCs with allergen and PD-L2-Fc reduced IL-12 p70 production, suggesting that PD-L2 inhibits allergen-driven IL-12 production. In our model, IL-12 did not diminish T helper type 2 responses but rather directly antagonized IL-13-inducible gene expression, highlighting a novel role for IL-12 in regulation of IL-13 signaling. Thus, allergen-driven enhancement of PD-L2 signaling through a PD-1-independent mechanism limits IL-12 secretion, exacerbating AHR.


Asunto(s)
Asma/inmunología , Asma/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interleucina-12/biosíntesis , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Alérgenos/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Asma/tratamiento farmacológico , Asma/genética , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inmunoglobulina G/inmunología , Subunidad p40 de la Interleucina-12/metabolismo , Interleucina-13/metabolismo , Interleucina-13/farmacología , Masculino , Ratones , Moco/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Proteína 2 Ligando de Muerte Celular Programada 1/agonistas , Proteína 2 Ligando de Muerte Celular Programada 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
11.
Leukemia ; 26(12): 2483-93, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22743623

RESUMEN

t(17;19)-acute lymphoblastic leukemia (ALL) shows extremely poor prognosis. E2A-HLF derived from t(17;19) blocks apoptosis induced by the intrinsic mitochondrial pathway and has a central role in leukemogenesis and chemoresistance. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is expressed on cytotoxic T cells and natural killer cells and binds with death receptors (DR4/DR5), inducing apoptosis by dual activation of intrinsic and extrinsic pathways, and TRAIL mediates the graft-versus-leukemia (GVL) effect after allogeneic stem cell transplantation (allo-SCT). We found that cell lines and patients' samples of t(17;19)-ALL expressed death receptors for TRAIL, and recombinant soluble TRAIL immediately induced apoptosis into t(17;19)-ALL cell lines. E2A-HLF induced gene expression of DR4/DR5, which was dependent on the DNA-binding and transactivation activities of E2A-HLF through the 5' upstream region of the start site at least in the DR4 gene. Introduction of E2A-HLF into non-t(17;19)-ALL cell line upregulated DR4 and DR5 expression, and sensitized to proapoptotic activity of recombinant soluble TRAIL. Finally, a newly diagnosed t(17;19)-ALL patient underwent allo-SCT immediately after induction of first complete remission, and the patient has survived without relapse for over 3-1/2 years after allo-SCT. These findings suggest that E2A-HLF sensitizes t(17;19)-ALL to the GVL effect by upregulating death receptors for TRAIL.


Asunto(s)
Apoptosis/efectos de los fármacos , Cromosomas Humanos Par 17/genética , Cromosomas Humanos Par 19/genética , Proteínas de Unión al ADN/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Factores de Transcripción/genética , Translocación Genética/genética , Western Blotting , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Humanos , Luciferasas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas , Regulación hacia Arriba
12.
Mucosal Immunol ; 5(2): 216-25, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22294048

RESUMEN

Many prion diseases are orally acquired. Our data show that after oral exposure, early prion replication upon follicular dendritic cells (FDC) in Peyer's patches is obligatory for the efficient spread of disease to the brain (termed neuroinvasion). For prions to replicate on FDC within Peyer's patches after ingestion of a contaminated meal, they must first cross the gut epithelium. However, the mechanism through which prions are conveyed into Peyer's patches is uncertain. Within the follicle-associated epithelium overlying Peyer's patches are microfold cells (M cells), unique epithelial cells specialized for the transcytosis of particles. We show that following M cell-depletion, early prion accumulation upon FDC in Peyer's patches is blocked. Furthermore, in the absence of M cells at the time of oral exposure, neuroinvasion and disease development are likewise blocked. These data suggest M cells are important sites of prion uptake from the gut lumen into Peyer's patches.


Asunto(s)
Encéfalo/metabolismo , Células Dendríticas Foliculares/inmunología , Enterocitos/patología , Ganglios Linfáticos Agregados/inmunología , Enfermedades por Prión/inmunología , Priones/inmunología , Ligando RANK/inmunología , Administración Oral , Animales , Encéfalo/inmunología , Encéfalo/patología , Diferenciación Celular/genética , Células Cultivadas , Células Dendríticas Foliculares/patología , Progresión de la Enfermedad , Ingestión de Alimentos , Enterocitos/inmunología , Enterocitos/metabolismo , Exposición a Riesgos Ambientales/efectos adversos , Carne , Ratones , Ratones Noqueados , Ganglios Linfáticos Agregados/patología , Enfermedades por Prión/patología , Enfermedades por Prión/prevención & control , Enfermedades por Prión/transmisión , Transcitosis/genética , Transcitosis/inmunología
13.
Gene Ther ; 19(11): 1075-84, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22071968

RESUMEN

Myeloablative transplantation of bone marrow (BM) engineered to express myelin oligodendrocyte glycoprotein (MOG) establishes central intrathymic tolerance and completely prevents MOG-induced experimental autoimmune encephalomyelitis (EAE) in mice. Here we asked whether non-myeloablative transplantation of MOG expressing BM (pMOG-bone marrow transplantation (BMT)) can also provide the same protection. Using stepwise reduction of irradiation doses, 275 cGy irradiation with pMOG-BMT protected 100% of mice from EAE development even with two subsequent re-challenge with MOG. Irradiation doses <275 cGy produced dose-dependent partial protection with significant disease protection still evident at 50 cGy. Splenocytes from 275 cGy recipients proliferated to MOG stimulation in vitro, indicating that MOG-reactive cells are present in the periphery but failed to induce disease. MOG-stimulated splenocytes produced little or no interleukin-17, interferon-γ, granulocyte-monocyte colony stimulating factor and tumor necrosis factor-α compared with EAE control. Adoptive transfer of CD4 T cells from EAE-resistant mice into Rag2(-/-) mice devoid of MOG expression resulted in MOG-induced EAE in ~74% of mice. Treatment of EAE-resistant mice with anti-programmed death 1 (PD-1) monoclonal antibody-induced EAE in 67% of mice. We conclude that non-myeloablative transplantation of self-antigen expressing BM induces robust peripheral tolerance that completely prevented EAE development. Our findings implicate clonal anergy and the PD-1 pathway in the maintenance of peripheral tolerance.


Asunto(s)
Autoantígenos/genética , Autoantígenos/inmunología , Autoinmunidad/genética , Trasplante de Médula Ósea , Tolerancia Periférica/genética , Acondicionamiento Pretrasplante , Traslado Adoptivo , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Antígenos de Diferenciación/inmunología , Linfocitos T CD4-Positivos/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Relación Dosis-Respuesta en la Radiación , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Mediadores de Inflamación/inmunología , Ratones , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/genética , Glicoproteína Mielina-Oligodendrócito/inmunología , Receptor de Muerte Celular Programada 1 , Médula Espinal/patología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/inmunología , Quimera por Trasplante , Irradiación Corporal Total
14.
Leukemia ; 26(2): 332-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21931322

RESUMEN

Activation of the fibrinolytic system during lymphoma progression is a well-documented clinical phenomenon. But the mechanism by which the fibrinolytic system can modulate lymphoma progression has been elusive. The main fibrinolytic enzyme, plasminogen (Plg)/plasmin (Plm), can activate matrix metalloproteinases (MMPs), such as MMP-9, which has been linked to various malignancies. Here we provide the evidence that blockade of Plg reduces T-cell lymphoma growth by inhibiting MMP-9-dependent recruitment of CD11b(+)F4/80(+) myeloid cells locally within the lymphoma tissue. Genetic Plg deficiency and drug-mediated Plm blockade delayed T-cell lymphoma growth and diminished MMP-9-dependent CD11b(+)F4/80(+) myeloid cell infiltration into lymphoma tissues. A neutralizing antibody against CD11b inhibited T-cell lymphoma growth in vivo, which indicates that CD11b(+) myeloid cells have a role in T-cell lymphoma growth. Plg deficiency in T-cell lymphoma-bearing mice resulted in reduced plasma levels of the growth factors vascular endothelial growth-A and Kit ligand, both of which are known to enhance myeloid cell proliferation. Collectively, the data presented in this study demonstrate a previously undescribed role of Plm in lymphoproliferative disorders and provide strong evidence that specific blockade of Plg represents a promising approach for the regulation of T-cell lymphoma growth.


Asunto(s)
Antifibrinolíticos/farmacología , Antígeno CD11b/inmunología , Linfoma de Células T/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Linfoma de Células T/enzimología , Linfoma de Células T/inmunología , Metaloproteinasa 9 de la Matriz/genética , Ratones , Plasminógeno/genética , Plasminógeno/fisiología , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Allergy ; 67(3): 328-35, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22175699

RESUMEN

BACKGROUND: Eosinophils and lymphocytes are pathogenically important in allergic inflammation and sensitive to Fas-mediated apoptosis. Fas ligand (FasL) activity therefore should play a role in regulating the allergic immune response. We aimed to characterize the role of FasL expression in airway eosinophilia in Aspergillus fumigatus (Af)-induced sensitization and to determine whether FasL neutralization alters the inflammatory response. METHODS: Sensitized Balb/c mice were killed before (day 0) and 1, 7 and 10 days after a single intranasal challenge with Af. Animals received either neutralizing antibody to FasL (clone MFL4) or irrelevant hamster IgG via intraperitoneal injection on days -1 and 5. FasL expression, BAL and tissue inflammatory cell and cytokine profile, and apoptosis were assessed. RESULTS: Postchallenge FasL gene expression in BAL cells and TUNEL positivity in the airways coincided with the height of inflammatory cell influx on day 1, while soluble FasL protein was released on day 7, preceding resolution of the inflammatory changes. Although eosinophil numbers showed a negative correlation with soluble FasL levels in the airways, MBP(+) eosinophils remained TUNEL negative in the submucosal tissue, throughout the 10-day period after Af challenge. Systemic FasL neutralization significantly enhanced BAL and tissue eosinophil counts. This effect was associated with increased activation of T cells and release of IL-5, IL-9, and GM-CSF in the BAL fluid of mice, indicating an involvement of pro-eosinophilic survival pathways. CONCLUSIONS: FasL activity may play an active role in resolving eosinophilic inflammation through regulating T cells and pro-eosinophilic cytokine release during the allergic airway response.


Asunto(s)
Asma/inmunología , Asma/fisiopatología , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Proteína Ligando Fas/inmunología , Inflamación/inmunología , Animales , Aspergillus fumigatus/inmunología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Cricetinae , Eosinofilia/inmunología , Eosinofilia/patología , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Femenino , Humanos , Recuento de Leucocitos , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización
16.
Am J Transplant ; 10(1): 40-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19889124

RESUMEN

The programmed death-1 (PD-1)/B7-H1 pathway acts as an important negative regulator of immune responses. We herein investigated the role of the PD-1/B7-H1 pathway in establishing an immunological spontaneous tolerance status in mouse liver allografting. B7-H1 is highly expressed on the donor-derived tissue cells and it is also associated with the apoptosis of infiltrating T cells in the allografts. Strikingly, a blockade of the PD-1/B7-H1 pathway via anti-B7-H1mAb or using B7-H1 knockout mice as a donor led to severe cell infiltration as well as hemorrhaging and necrosis, thus resulting in mortality within 12 days. Furthermore, the expression of the FasL, perforin, granzyme B, iNOS and OPN mRNA in the liver allografts increased in the antibody-treated group in comparison to the controls. Taken together, these data revealed that the B7-H1 upregulation on the tissue cells of liver allografts thus plays an important role in the apoptosis of infiltrating cells, which might play a critical role of the induction of the spontaneous tolerance after hepatic transplantation in mice.


Asunto(s)
Antígenos de Superficie/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Antígeno B7-1/inmunología , Trasplante de Hígado/inmunología , Glicoproteínas de Membrana/inmunología , Péptidos/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Apoptosis , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Antígeno B7-1/genética , Antígeno B7-H1 , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Supervivencia de Injerto/inmunología , Tolerancia Inmunológica , Trasplante de Hígado/patología , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/antagonistas & inhibidores , Péptidos/deficiencia , Péptidos/genética , Receptor de Muerte Celular Programada 1 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T/inmunología , Linfocitos T/patología , Quimera por Trasplante/inmunología , Trasplante Homólogo
17.
Br J Cancer ; 101(10): 1709-16, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19844235

RESUMEN

BACKGROUND: B7-H3 is a new member of the B7 ligand family and regulates T-cell responses in various conditions. However, the role of B7-H3 in tumour immunity is largely unknown. The purpose of this study was to evaluate the clinical significance of B7-H3 expression in human pancreatic cancer and the therapeutic potential for cancer immunotherapy. METHODS: We investigated B7-H3 expression in 59 patients with pancreatic cancer by immunohistochemistry and real-time PCR. Furthermore, we examined the anti-tumour effect of B7-H3-blocking monoclonal antibody in vivo in a murine pancreatic cancer model. RESULTS: Tumour-related B7-H3 expression was abundant in most human pancreatic cancer tissues and was significantly higher compared with that in non-cancer tissue or normal pancreas. Moreover, its expression was significantly more intense in cases with lymph node metastasis and advanced pathological stage. B7-H3 blockade promoted CD8(+) T-cell infiltration into the tumour and induced a substantial anti-tumour effect on murine pancreatic cancer. In addition, the combination of gemcitabine with B7-H3 blockade showed a synergistic anti-tumour effect without overt toxicity. CONCLUSION: Our data show for the first time that B7-H3 may have a critical role in pancreatic cancer and provide the rationale for developing a novel cancer immunotherapy against this fatal disease.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD/biosíntesis , Antígenos CD/inmunología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales/inmunología , Antígenos CD/genética , Antígenos B7 , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neoplasias Pancreáticas/genética , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Gene Ther ; 16(11): 1329-39, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19626052

RESUMEN

The induction of strong cell-mediated immunity against targeted cancer cells is difficult, and often requires specific vaccination schema and the appropriate adjuvants to be effective. The chemokine RANTES has been studied as a vaccine adjuvant in cancer therapy, but specific applications remain to be determined. For gene-based vaccination against B16 melanoma in C57BL/6JNarl mice, initial priming with mouse RANTES cDNA followed 24 h later by human gp100 DNA vaccination, and later boosting with a viral vector expressing mRANTES and hgp100 strongly suppressed B16/hgp100 primary tumors and lung metastasis. The inclusion of mRANTES in this vaccination regimen gave significantly better suppression of tumor growth, substantially enhanced mouse survival, and led to greater cytotoxic activity of splenocytes against B16/hgp100 cells than vaccination against hgp100 alone. B16/hgp100 melanoma cells were resistant to the ligands TRAIL and FasL in vitro but sensitized to them in vivo owing to the priming effect of cytokines in response to vaccination. Our data demonstrate that co-vaccination with chemokine (mRANTES) and tumor-specific (hgp100) genes in a specific time sequence is more effective at suppressing tumor growth and metastasis than hgp100 alone, and this effect may be mediated by sensitization of tumor cells to death ligands.


Asunto(s)
Vacunas contra el Cáncer , Quimiocina CCL5/metabolismo , Terapia Genética/métodos , Melanoma Experimental/prevención & control , Glicoproteínas de Membrana/metabolismo , Adyuvantes Inmunológicos/genética , Animales , Vacunas contra el Cáncer/inmunología , Quimiocina CCL5/genética , Citotoxicidad Inmunológica , ADN Complementario/genética , Femenino , Expresión Génica , Humanos , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Piel/inmunología , Transgenes , Células Tumorales Cultivadas , Vacunas de ADN/inmunología , Antígeno gp100 del Melanoma
19.
Br J Ophthalmol ; 93(1): 110-5, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18782797

RESUMEN

BACKGROUND/AIMS: Tumour-necrosis-factor-related apoptosis-inducing ligand (TRAIL) plays a role in the development of allergic asthma. The study aimed to determine whether TRAIL also participates in the development of experimental allergic conjunctivitis (EC), another allergic disease model. METHODS: EC was induced in BALB/c mice by active immunisation with ragweed (RW) followed by RW challenge. To investigate whether TRAIL in the conjunctiva plays a role in the development of EC, conjunctival TRAIL expression in EC-developing mice was evaluated by immunohistochemistry. Additionally, the effect of subconjunctival injection of recombinant TRAIL on conjunctival inflammation was examined. To investigate whether TRAIL expressed in systemic immunocompetent cells plays a role in the development of EC, anti-TRAIL blocking Ab or anti-TRAIL receptor agonistic Ab was intraperitoneally injected into EC-developing mice, and conjunctival eosinophil infiltration was evaluated. RESULTS: Conjunctival TRAIL expression was not increased by EC induction. Moreover, subconjunctival injection of TRAIL protein in naive mice did not induce conjunctival inflammation. Thus, TRAIL in the conjunctiva is less likely to participate in the development of EC. Systemic treatment with anti-TRAIL blocking Ab in EC-developing mice did not affect the severity of EC. However, systemic treatment during the induction phase of EC with an agonistic Ab for the TRAIL receptor significantly augmented the severity of EC and increased Ag-recall splenocyte IFN-gamma production in vitro. CONCLUSIONS: These results indicate that TRAIL receptor-expressing cells in lymphoid organ participate in the development of EC.


Asunto(s)
Conjuntiva/inmunología , Conjuntivitis Alérgica/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Alérgenos , Ambrosia , Animales , Separación Celular , Conjuntiva/metabolismo , Conjuntivitis Alérgica/metabolismo , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Polen , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
20.
Am J Transplant ; 8(11): 2272-82, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18785958

RESUMEN

Memory T cells are the very essence of adaptive immunity with their rapid and efficient response to antigen rechallenge and long-term persistence. However, it is becoming increasingly evident that when primed with self or transplanted tissue, these cells play a key role in causing and perpetuating tissue damage. Furthermore, current treatments, which efficiently control the naive response, have limited effects on primed T cells. We have used a treatment based on a combination of antibodies specific for molecules expressed by activated T lymphocytes to selectively remove these cells. This approach, which we termed multi-hit therapy, leads to cumulative binding of antibodies to the target T cells and a striking prolongation of skin graft survival in presensitized recipients in a stringent skin transplant model. The findings are consistent with the depletion of graft-specific CD4+ and CD8+ T cells, although other modes of action, such as T-cell regulation and altered migration could play a role. In conclusion, our therapeutic strategy controls primed T cells which are a major driving force in the pathology of many autoimmune diseases and in transplant rejection.


Asunto(s)
Supervivencia de Injerto , Activación de Linfocitos , Linfocitos T/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular , Femenino , Memoria Inmunológica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Bazo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...