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.
Artigo em Inglês | MEDLINE | ID: mdl-26555706

RESUMO

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.


Assuntos
Inflamação/imunologia , Pulmão/imunologia , Monócitos/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , RNA Mensageiro/genética , Mucosa Respiratória/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fumar/efeitos adversos , Ligante Indutor de Apoptose Relacionado a TNF/genética , Regulação para Cima
2.
Leukemia ; 29(3): 576-85, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25234168

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação Leucêmica da Expressão Gênica , Proteínas de Homeodomínio/genética , Leucemia Mieloide Aguda/genética , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Proliferação de Células , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Transgênicos , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais , Análise de Sobrevida , Fatores de Transcrição HES-1 , Tirosina Quinase 3 Semelhante a fms/metabolismo
3.
Leukemia ; 29(1): 145-56, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24791857

RESUMO

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.


Assuntos
Fibrinolisina/antagonistas & inibidores , Doença Enxerto-Hospedeiro/prevenção & controle , Mediadores da Inflamação/antagonistas & inibidores , Metaloproteinase 9 da Matriz/metabolismo , Animais , Sequência de Bases , Transporte Biológico , Linhagem Celular , Primers do DNA , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Doença Enxerto-Hospedeiro/enzimologia , Doença Enxerto-Hospedeiro/mortalidade , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Índice de Gravidade de Doença
4.
Mucosal Immunol ; 7(5): 1199-208, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24569802

RESUMO

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.


Assuntos
Alérgenos/imunologia , Hipersensibilidade Respiratória/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Apoptose/imunologia , Caspase 3/genética , Caspase 3/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Doença Crônica , Feminino , Regulação da Expressão Gênica/imunologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Pulmão/imunologia , Pulmão/fisiopatologia , Camundongos , Camundongos Knockout , Proteínas Recombinantes/genética , Hipersensibilidade Respiratória/tratamento farmacológico , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Células Th2/imunologia
5.
Mucosal Immunol ; 7(4): 786-801, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24220301

RESUMO

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.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Interleucina-13/biossíntese , Linfonodos/imunologia , Células Th2/imunologia , Células Th2/metabolismo , Tretinoína/farmacologia , Administração Oral , Animais , Antígenos/administração & dosagem , Antígenos/imunologia , Ligante de CD40/metabolismo , Diferenciação Celular/imunologia , Colo/imunologia , Colo/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Isotipos de Imunoglobulinas/imunologia , Imunofenotipagem , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mesentério/imunologia , Mesentério/metabolismo , Camundongos , Fenótipo , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Células Th2/citologia , Fator de Necrose Tumoral alfa/metabolismo , Deficiência de Vitamina A/imunologia , Deficiência de Vitamina A/metabolismo
6.
Mucosal Immunol ; 7(3): 478-88, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24045576

RESUMO

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.


Assuntos
Pneumonia/metabolismo , Infecções Respiratórias/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos Neutralizantes/farmacologia , Apoptose/genética , Infecções por Chlamydia/metabolismo , Chlamydia muridarum , Modelos Animais de Doenças , Progressão da Doença , Expressão Gênica , Camundongos , Camundongos Knockout , Muco/metabolismo , NF-kappa B/metabolismo , Pneumonia/genética , Pneumonia/microbiologia , Pneumonia/patologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/metabolismo , Infecções Respiratórias/genética , Infecções Respiratórias/microbiologia , Ligante Indutor de Apoptose Relacionado a TNF/deficiência , Ligante Indutor de Apoptose Relacionado a TNF/genética
8.
Clin Exp Immunol ; 172(3): 500-6, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23600839

RESUMO

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.


Assuntos
Neoplasias Experimentais/terapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Inibidores da Angiogênese/administração & dosagem , Animais , Anticorpos Monoclonais/administração & dosagem , Linhagem Celular Tumoral , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Sinergismo Farmacológico , Feminino , Humanos , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/imunologia , Neovascularização Patológica/prevenção & controle , Receptor de Morte Celular Programada 1/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
9.
Oncogene ; 32(13): 1670-81, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22665066

RESUMO

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.


Assuntos
Proteínas de Fusão bcr-abl/fisiologia , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Receptores de Morte Celular/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Linhagem Celular Tumoral , Proteínas de Fusão bcr-abl/genética , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Cromossomo Filadélfia , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/farmacologia , Receptores de Morte Celular/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
10.
Mucosal Immunol ; 6(4): 728-39, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23149662

RESUMO

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.


Assuntos
Asma/imunologia , Asma/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interleucina-12/biossíntese , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Alérgenos/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Asma/tratamento farmacológico , Asma/genética , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Imunoglobulina G/imunologia , Subunidade p40 da Interleucina-12/metabolismo , Interleucina-13/metabolismo , Interleucina-13/farmacologia , Masculino , Camundongos , Muco/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Proteína 2 Ligante de Morte Celular Programada 1/agonistas , Proteína 2 Ligante de Morte Celular Programada 1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
11.
Leukemia ; 26(12): 2483-93, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22743623

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Cromossomos Humanos Par 17/genética , Cromossomos Humanos Par 19/genética , Proteínas de Ligação a DNA/genética , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fatores de Transcrição/genética , Translocação Genética/genética , Western Blotting , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Luciferases/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo , Ativação Transcricional , Células Tumorais Cultivadas , Regulação para Cima
12.
Mucosal Immunol ; 5(2): 216-25, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22294048

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Células Dendríticas Foliculares/imunologia , Enterócitos/patologia , Nódulos Linfáticos Agregados/imunologia , Doenças Priônicas/imunologia , Príons/imunologia , Ligante RANK/imunologia , Administração Oral , Animais , Encéfalo/imunologia , Encéfalo/patologia , Diferenciação Celular/genética , Células Cultivadas , Células Dendríticas Foliculares/patologia , Progressão da Doença , Ingestão de Alimentos , Enterócitos/imunologia , Enterócitos/metabolismo , Exposição Ambiental/efeitos adversos , Carne , Camundongos , Camundongos Knockout , Nódulos Linfáticos Agregados/patologia , Doenças Priônicas/patologia , Doenças Priônicas/prevenção & controle , Doenças Priônicas/transmissão , Transcitose/genética , Transcitose/imunologia
13.
Allergy ; 67(3): 328-35, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22175699

RESUMO

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.


Assuntos
Asma/imunologia , Asma/fisiopatologia , Modelos Animais de Doenças , Eosinófilos/imunologia , Proteína Ligante Fas/imunologia , Inflamação/imunologia , Animais , Aspergillus fumigatus/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Cricetinae , Eosinofilia/imunologia , Eosinofilia/patologia , Proteína Ligante Fas/genética , Proteína Ligante Fas/metabolismo , Feminino , Humanos , Contagem de Leucócitos , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização
14.
Gene Ther ; 19(11): 1075-84, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22071968

RESUMO

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.


Assuntos
Autoantígenos/genética , Autoantígenos/imunologia , Autoimunidade/genética , Transplante de Medula Óssea , Tolerância Periférica/genética , Condicionamento Pré-Transplante , Transferência Adotiva , Animais , Anticorpos/administração & dosagem , Anticorpos/imunologia , Antígenos de Diferenciação/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/biossíntese , Citocinas/imunologia , Relação Dose-Resposta à Radiação , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Mediadores da Inflamação/imunologia , Camundongos , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/genética , Glicoproteína Mielina-Oligodendrócito/imunologia , Receptor de Morte Celular Programada 1 , Medula Espinal/patologia , Baço/imunologia , Baço/metabolismo , Linfócitos T Reguladores/imunologia , Quimeras de Transplante , Irradiação Corporal Total
15.
Leukemia ; 26(2): 332-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21931322

RESUMO

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.


Assuntos
Antifibrinolíticos/farmacologia , Antígeno CD11b/imunologia , Linfoma de Células T/patologia , Metaloproteinase 9 da Matriz/metabolismo , Animais , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Linfoma de Células T/enzimologia , Linfoma de Células T/imunologia , Metaloproteinase 9 da Matriz/genética , Camundongos , Plasminogênio/genética , Plasminogênio/fisiologia , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Am J Transplant ; 10(1): 40-6, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19889124

RESUMO

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.


Assuntos
Antígenos de Superfície/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Antígeno B7-1/imunologia , Transplante de Fígado/imunologia , Glicoproteínas de Membrana/imunologia , Peptídeos/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Apoptose , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Antígeno B7-1/genética , Antígeno B7-H1 , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/imunologia , Tolerância Imunológica , Transplante de Fígado/patologia , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/antagonistas & inibidores , Peptídeos/deficiência , Peptídeos/genética , Receptor de Morte Celular Programada 1 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/imunologia , Linfócitos T/patologia , Quimeras de Transplante/imunologia , Transplante Homólogo
17.
Br J Cancer ; 101(10): 1709-16, 2009 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-19844235

RESUMO

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.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD/biossíntese , Antígenos CD/imunologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/terapia , Receptores Imunológicos/biossíntese , Receptores Imunológicos/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD/genética , Antígenos B7 , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neoplasias Pancreáticas/genética , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Gene Ther ; 16(11): 1329-39, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19626052

RESUMO

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.


Assuntos
Vacinas Anticâncer , Quimiocina CCL5/metabolismo , Terapia Genética/métodos , Melanoma Experimental/prevenção & controle , Glicoproteínas de Membrana/metabolismo , Adjuvantes Imunológicos/genética , Animais , Vacinas Anticâncer/imunologia , Quimiocina CCL5/genética , Citotoxicidade Imunológica , DNA Complementar/genética , Feminino , Expressão Gênica , Humanos , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Pele/imunologia , Transgenes , Células Tumorais Cultivadas , Vacinas de DNA/imunologia , Antígeno gp100 de Melanoma
19.
Br J Ophthalmol ; 93(1): 110-5, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18782797

RESUMO

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.


Assuntos
Túnica Conjuntiva/imunologia , Conjuntivite Alérgica/imunologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Alérgenos , Ambrosia , Animais , Separação Celular , Túnica Conjuntiva/metabolismo , Conjuntivite Alérgica/metabolismo , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Pólen , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
20.
J Pathol ; 216(4): 505-13, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18810759

RESUMO

Granulomatous experimental autoimmune thyroiditis (G-EAT) is induced by mouse thyroglobulin (MTG)-sensitized splenocytes activated in vitro with MTG and IL-12. Thyroid lesions reach maximal severity 20 days after cell transfer, and usually resolve or progress to fibrosis by day 60 depending on the extent of thyroid damage at day 20. Our previous studies indicated that neutralization of TNF-alpha or FasL had no effect on G-EAT induction, but neutralization of TNF-alpha promoted, while neutralization of FasL inhibited, G-EAT resolution. TNF-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily. This study was undertaken to define the role of endogenous TRAIL in G-EAT development and/or resolution. Neutralization of endogenous TRAIL had little effect on G-EAT induction, but significantly inhibited G-EAT resolution and increased thyroid fibrosis. This correlated with higher expression of pro-inflammatory cytokines and preferential expression of the pro-apoptotic molecule TRAIL, and anti-apoptotic molecules FLIP and Bcl-xL on inflammatory cells in thyroids of anti-TRAIL-treated recipients. The results suggest that endogenous TRAIL is not required for G-EAT development in recipients, but is critical for G-EAT resolution. Endogenous TRAIL might promote resolution, at least in part, through modulation of the balance between pro- and anti-inflammatory cytokines, and the expression pattern of pro- and anti-apoptotic molecules of thyroid epithelial cells (TECs) and inflammatory cells.


Assuntos
Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Tireoidite Autoimune/metabolismo , Tireoidite Subaguda/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Apoptose , Células Cultivadas , Inflamação , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Microscopia de Fluorescência , Modelos Animais , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Tireoidite Autoimune/tratamento farmacológico , Tireoidite Subaguda/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...