Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

País de afiliación
Intervalo de año de publicación
1.
J Immunol ; 187(4): 1986-97, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21746963

RESUMEN

Cytosolic phospholipase A(2)α (cPLA(2)α) is the rate-limiting enzyme for release of arachidonic acid, which is converted primarily to PGs via the cyclooxygenase 1 and 2 pathways and to leukotrienes via the 5-lipoxygenase pathway. We used adoptive transfer and relapsing-remitting forms of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, in two different strains of mice (SJL or C57BL/6) to demonstrate that blockade of cPLA(2)α with a highly specific small-molecule inhibitor during the tissue-damage effector phase abrogates the clinical manifestation of disease. Using the adoptive transfer model in SJL mice, we demonstrated that the blockade of cPLA(2)α during the effector phase of disease was more efficacious in ameliorating the disease pathogenesis than the blockade of each of the downstream enzymes, cyclooxygenase-1/2 and 5-lipooxygenase. Similarly, blockade of cPLA(2)α was highly efficacious in ameliorating disease pathogenesis during the effector phase of EAE in the adoptive transfer model of EAE in C57BL/6 mice. Investigation of the mechanism of action indicates that cPLA(2)α inhibitors act on APCs to diminish their ability to induce Ag-specific effector T cell proliferation and proinflammatory cytokine production. Furthermore, cPLA(2)α inhibitors may prevent activation of CNS-resident microglia and may increase oligodendrocyte survival. Finally, in a relapsing-remitting model of EAE in SJL mice, therapeutic administration of a cPLA(2)α inhibitor, starting from the peak of disease or during remission, completely protected the mice from subsequent relapses.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Inhibidores Enzimáticos/farmacología , Fosfolipasas A2 Grupo IV/antagonistas & inhibidores , Esclerosis Múltiple/prevención & control , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/enzimología , Células Presentadoras de Antígenos/patología , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/inmunología , Araquidonato 5-Lipooxigenasa/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/inmunología , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2 , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Fosfolipasas A2 Grupo IV/genética , Fosfolipasas A2 Grupo IV/inmunología , Fosfolipasas A2 Grupo IV/metabolismo , Humanos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Microglía/enzimología , Microglía/inmunología , Microglía/patología , Esclerosis Múltiple/enzimología , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Oligodendroglía/enzimología , Oligodendroglía/inmunología , Oligodendroglía/patología , Linfocitos T/enzimología , Linfocitos T/inmunología , Linfocitos T/patología
2.
J Leukoc Biol ; 81(3): 825-34, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17148690

RESUMEN

Although IFN-alpha forms the foundation of therapy for chronic hepatitis C, only a minority of patients has a sustained response to IFN-alpha alone. The antiviral activities of IFN-alpha formed the rationale for its use in viral hepatitis. However, IFN-alpha and the other Type I IFNs are also pleiotropic immune regulators. Type I IFNs can promote IFN-gamma production by activating STAT4 but can also inhibit production of IL-12, a potent activator of STAT4 and IFN-gamma production. The efficacy of IFN-alpha in the treatment of hepatitis C may therefore depend in part on the balance of IFN-gamma-inducing and IL-12-suppressing effects. We characterized the effects of pegylated IFN-alpha therapy for hepatitis C on the capacity of patients' PBMC to produce IL-12 and IFN-gamma ex vivo. Cells from patients with a sustained virological response to therapy had significantly greater levels of IFN-alpha-driven IFN-gamma production prior to treatment than those from nonresponding patients. No differences in pretreatment IL-12 productive capacity were seen between patient groups. However, therapy with IFN-alpha led to suppression of inducible IL-12 production throughout the course of therapy in both groups of patients.


Asunto(s)
Hepatitis C Crónica/tratamiento farmacológico , Interferón-alfa/administración & dosificación , Interferón gamma/fisiología , Interleucina-12/fisiología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Niño , Preescolar , Femenino , Hepatitis C Crónica/inmunología , Humanos , Inyecciones Subcutáneas , Interferón-alfa/uso terapéutico , Interferón beta/administración & dosificación , Interferón beta/uso terapéutico , Interferón gamma/biosíntesis , Interleucina-12/biosíntesis , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Proyectos Piloto , Relación Estructura-Actividad
3.
Comb Chem High Throughput Screen ; 9(7): 565-70, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16925516

RESUMEN

Phosphoinositide 3-kinases (PI3Ks) comprise a family of kinases that transfer the terminal phosphate of adenosine triphosphate to phosphoinositides at the 3-hydroxyl of the inositol ring to form phosphoinositide (3,4,5) triphosphate (PIP3). The PI3Ks have been shown to play key roles in cell growth, motility, morphology, and survival and thus are of interest as targets in anti-inflammatory and anti-oncogenic drug development. To facilitate identification of novel and selective inhibitors of PI3Ks, we have developed a TR-FRET assay that uses directly labeled reagents. The assay makes use of the high affinity binding of phosphoinositides to a Pleckstrin homology (PH) domain in the general receptor for phosphoinositides 1 (Grp1) protein. It monitors PIP3 produced from the enzymatic reaction by measuring its competition with Bodipy-FL-labeled PIP3 for binding to Terbium chelate-labeled Grp1. By using directly labeled reagents, this assay configuration offers higher sensitivity and faster binding/dissociation kinetics than existing non-radioactive assays, which are critical for competitive assay formats. The assay is homogenous, robust (Z' = 0.88), and simple and, thus, compatible with high throughput screening (HTS) processes.


Asunto(s)
Bioensayo/métodos , Quelantes/química , Transferencia Resonante de Energía de Fluorescencia/métodos , Fosfatidilinositol 3-Quinasas/análisis , Terbio/química , Técnicas Químicas Combinatorias , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositoles/metabolismo , Unión Proteica , Receptores Citoplasmáticos y Nucleares/metabolismo
4.
Cancer Res ; 62(20): 5727-35, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12384531

RESUMEN

The therapeutic efficacy of combined antiangiogenic and immune therapy was tested against weakly immunogenic and highly metastatic 4T1 breast tumor using SU6668, an angiogenesis inhibitor and recombinant murine (rm) B7.2-IgG fusion protein, an immunostimulator. SU6668 inhibits the tyrosine kinase activity of three angiogenic receptors VEGFR2 (Flk-1/KDR), PDGFRbeta, and FGFR1, which play a crucial role in tumor-induced vascularization. rmB7.2-IgG is a fusion protein of the extracellular domain of B7.2, and the hinge and constant domains of murine IgG2a. Intermolecule disulfide linkages are maintained so that it forms a dimer. Our studies showed that three weekly immunizations of BALB/c mice bearing 0.5-0.8 cm 4T1 breast tumors with rmB7.2-IgG and irradiated 4T1 tumor cells (B7.2-IgG/TC) resulted in a significant inhibition of tumor growth and formation of pulmonary metastases. T-cell depletion experiments revealed that both CD4(+) and CD8(+) T lymphocytes are required for stimulation of the antitumor and antimetastatic immune response by B7.2-IgG/TC. Treatment of mice with SU6668 substantially inhibited tumor vascularization but did not inhibit tumor infiltration by T lymphocytes or the T-cell response to rmB7.2-IgG therapy. On the contrary, tumors in mice immunized with B7.2-IgG/TC and treated with SU6668 had higher numbers of tumor infiltrating T cells than tumors of other groups. SU6668 treatments initiated either on day 3 or day 10 after inoculation of 4T1 tumor cells resulted in a significant inhibition of tumor growth. Similarly, three weekly immunizations with B7.2-IgG/TC starting either on day 7 or 12 inhibited growth of 4T1 tumors. However, the most potent antitumor effects were observed in mice treated with a combination of SU6668 and B7.2-IgG/TC. Analysis of pulmonary metastases revealed that combined therapy also had a more potent antimetastatic effect than each modality alone. These results indicate that antiangiogenic and immune therapies using SU6668 and B7.2-IgG/TC are compatible, and manifest complementary antitumor and antimetastatic effects. Combined antiangiogenic and immune therapy might represent a new strategy for cancer treatment.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antígenos CD/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Vacunas contra el Cáncer/farmacología , Inmunoglobulina G/farmacología , Indoles/farmacología , Neoplasias Mamarias Experimentales/terapia , Glicoproteínas de Membrana/farmacología , Pirroles/farmacología , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Antígenos CD/administración & dosificación , Antígenos CD/inmunología , Antígeno B7-2 , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Terapia Combinada , Femenino , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/inmunología , Indoles/administración & dosificación , Interferón gamma/biosíntesis , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/inmunología , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Oxindoles , Propionatos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirroles/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo
6.
J Exp Ther Oncol ; 2(2): 93-9, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12415625

RESUMEN

Interleukin-12 (IL-12) has the capacity to activate cytotoxic lymphocytes, stimulate natural killer cells, induce the production of INF-gamma, and be synergistic with IL-2. We have evaluated this cytokine in an experimental model for metastatic melanoma that approximates the major clinical stages of metastatic dissemination. To develop primary melanoma tumors, mice were injected subcutaneously with 5 x 10(5) cells in a volume of 25 microliters into the middle of the tail (11). In a month, mice were started to be treated for 4 weeks with recombinant murine IL-12 (R mIL-12) at the following doses: 0, 0.5, 2.5, 5.0, 15.0, and 50 micrograms/kg. Diameters of the primary melanoma tumors were measured at weekly intervals. At the end of 13 weeks (9 weeks from the start of treatment with R mIL-12), all surviving mice were sacrificed. Pathological examination of lung metastases (macroscopy) was done with all dead or sacrificed mice. Treatment of mice bearing melanoma at a dose of 300 ng/mouse (15 micrograms/kg) inhibited development of primary tumors in 40% of mice. The primary tumor diameters were significantly lower in the group treated with 300 ng/mouse (15 micrograms/kg) in comparison to controls. At the end of the observation period, groups treated with 0.5, 2.5, 15.0, and 50 micrograms/kg had mean primary tumor diameters smaller than the control group. Evaluation of IL-12 therapy on primary tumor growth, mean diameters of primary tumors, survival rate, and development of lung metastases showed that the best results were observed using 300 ng/mouse (15 micrograms/kg) R mIL-12.


Asunto(s)
Interleucina-12/uso terapéutico , Neoplasias Pulmonares/secundario , Melanoma Experimental/tratamiento farmacológico , Animales , Melanoma Experimental/patología , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Tasa de Supervivencia
7.
Int Immunopharmacol ; 3(4): 457-68, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12689651

RESUMEN

IL-12 modulates T cell responses between helper T cells Th2 and Th1; however, the therapeutic potential of IL-12 for allergic diseases either directly or as an adjuvant in allergen therapy has been controversial. The role of intranasal IL-12 as an adjuvant in modulating the grass pollen allergen (GAL) therapy-induced systemic immune response and lung-specific inflammation and airway reactivity was examined in this study using a mouse model of established allergic asthma. The effects of intranasal or nebulized IL-12 with or without intranasal anti-IFN-gamma antibody were examined in groups of control and allergen-sensitized or -challenged mice. T cell cytokine patterns, antibody response profiles, pulmonary inflammation and airway reactivity were examined. Intranasal IL-12 was found to be more effective in the Th2-Th1 shifting of immune response and anti-inflammatory activity in the lung compared to nebulized IL-12 at the given doses. Intranasal IL-12 significantly decreased production of IFN-gamma, eotaxin and LTC4/D4/E4 in the lung and decreased eosinophil infiltration, resulting in attenuated airway hyper-responsiveness in GAL-sensitized (GS) mice. In contrast, intranasal IL-12 significantly increased IFN-gamma production in the thoracic lymph node cultures and decreased the IL-5/IFN-gamma ratio, suggesting a Th2-Th1 shift. Also, intranasal IL-12 increased GAL-specific IgG2a antibody response, while the IgE response remained unaffected. The systemic effects of IL-12 were IFN-gamma dependent. IL-12 induces differential expression of its own receptor beta1 and beta2 subunits in the lung tissues to augment IL-12 responsiveness. Together, these results demonstrate that intranasal IL-12 is effective in shifting the systemic immune response in the direction of Th1 in IFN-gamma-dependent manner, while decreasing pulmonary inflammation and airway reactivity independent of IFN-gamma. Thus, intranasal delivery of IL-12 may provide an approach for the treatment of asthma and may be useful as an adjuvant in local nasal immunotherapy (IT) and in asthma.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Formación de Anticuerpos/efectos de los fármacos , Asma/inmunología , Interleucina-12/uso terapéutico , Pulmón/patología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Administración por Inhalación , Administración Intranasal , Resistencia de las Vías Respiratorias/efectos de los fármacos , Resistencia de las Vías Respiratorias/inmunología , Alérgenos/administración & dosificación , Alérgenos/inmunología , Animales , Formación de Anticuerpos/inmunología , Asma/tratamiento farmacológico , Asma/patología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/inmunología , Inflamación , Interferón gamma/análisis , Interferón gamma/inmunología , Interleucina-12/administración & dosificación , Interleucina-12/efectos adversos , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos DBA , Nebulizadores y Vaporizadores , Polen/inmunología , Receptores de Interleucina/biosíntesis , Receptores de Interleucina/inmunología , Receptores de Interleucina-12 , Células TH1/inmunología , Células Th2/inmunología
8.
Gene ; 452(2): 63-71, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20036323

RESUMEN

Acidic mammalian chitinase (AMCase), an enzyme implicated in the pathology of asthma, is capable of chitin cleavage at a low pH optimum. The corresponding gene (CHIA) can be found in genome databases of a variety of mammals, but the enzyme properties of only the human and mouse proteins were extensively studied. We wanted to compare enzymes of closely related species, such as humans and macaques. In our attempt to study macaque AMCase, we searched for CHIA-like genes in human and macaque genomes. We found that both genomes contain several additional CHIA-like sequences. In humans, CHIA-L1 (hCHIA-L1) is an apparent pseudogene and has the highest homology to CHIA. To determine which of the two genes is functional in monkeys, we assessed their tissue expression levels. In our experiments, CHIA-L1 expression was not detected in human stomach tissue, while CHIA was expressed at high levels. However, in the cynomolgus macaque stomach tissue, the expression pattern of these two genes was reversed: CHIA-L1 was expressed at high levels and CHIA was undetectable. We hypothesized that in macaques CHIA-L1 (mCHIA-L1), and not CHIA, is a gene encoding an acidic chitinase, and cloned it, using the sequence of human CHIA-L1 as a guide for the primer design. We named the new enzyme MACase (Macaca Acidic Chitinase) to emphasize its differences from AMCase. MACase shares a similar tissue expression pattern and pH optimum with human AMCase, but is 50 times more active in our enzymatic activity assay. DNA sequence of the mCHIA-L1 has higher percentage identity to the human pseudogene hCHIA-L1 (91.7%) than to hCHIA (84%). Our results suggest alternate evolutionary paths for human and monkey acidic chitinases.


Asunto(s)
Quitinasas/genética , Evolución Molecular , Macaca fascicularis/genética , Macaca mulatta/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Quitina/metabolismo , Quitinasas/química , Quitinasas/metabolismo , Secuencia Conservada , Bases de Datos Genéticas , Regulación Enzimológica de la Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Especificidad de Órganos , Alineación de Secuencia
9.
Am J Physiol Lung Cell Mol Physiol ; 296(2): L236-47, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19060229

RESUMEN

Mast cell-fibroblast interactions may contribute to fibrosis in asthma and other disease states. Fibroblast contraction is known to be stimulated by coculture with the human mast cell line, HMC-1, or by mast cell-derived agents. Matrix metalloproteinases (MMPs) can also mediate contraction, but the MMP-dependence of mast cell-induced fibroblast contractility is not established, and the consequences of mast cell activation within the coculture system have not been fully explored. We demonstrate that activation of primary human mast cells (pHMC) with IgE receptor cross-linking, or activation of HMC-1 with C5a, enhanced contractility of human lung fibroblasts in a three-dimensional collagen lattice system. This enhanced contractility was inhibited by the pan-MMP antagonist, batimastat, and was transferrable in the conditioned medium of activated mast cells. Exogenously added MMPs promoted gel contraction by mediating the proteolytic activation of latent transforming growth factor-beta (TGF-beta). Consistent with this, fibroblast contraction induced by mast cell activation was enhanced by addition of excess latent TGF-beta to the cultures. Batimastat inhibited this response, suggesting that MMPs capable of activating latent TGF-beta were released following mast cell activation in coculture with fibroblasts. Collagen production was also stimulated by activated mast cells in an MMP-dependent manner. MMP-2 and MMP-3 content of the gels increased in the presence of activated mast cells, and inhibition of these enzymes blocked the contractile response. These findings demonstrate the MMP dependence of mast cell-induced fibroblast contraction and collagen production.


Asunto(s)
Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Mastocitos/fisiología , Metaloproteinasas de la Matriz/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/efectos de los fármacos , Humanos , Inmunoglobulina E/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Metaloproteinasas de la Matriz/genética , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Inhibidores de Proteasas/farmacología , Tiofenos/farmacología , Factor de Crecimiento Transformador beta/metabolismo
10.
Angiogenesis ; 8(1): 13-23, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16132614

RESUMEN

Experimental studies of antiangiogenic or immune therapy of cancer have generated a great deal of optimism. However, the results of clinical testing of these therapies are below expectations. We hypothesized that the antitumor efficacy can be increased when immune destruction of tumor cell is combined with destruction of tumor vasculature by antiangiogenic drugs. In the present study the therapeutic efficacy of combined antiangiogenic and immune therapy has been tested against the highly aggressive, MHC class I negative murine RM1 prostate tumor. SU6668 was used as the antiangiogenic drug and recombinant murine B7.2-IgG fusion protein was used to stimulate T cell-mediated immune destruction of tumor cells. SU6668 is an inhibitor of the tyrosine kinase activity of three angiogenic receptors VEGFR2 (Flk-1/KDR), PDGFRbeta and FGFR1 that play a crucial role in tumor-induced vascularization. Our studies show that B7.2-IgG treatment of mice with established RM1 prostate tumors resulted in a significant inhibition of tumor growth. Both CD4+ and CD8+ T cells were responsible for this effect. SU6668 therapy substantially inhibited tumor vascularization and tumor growth. When tumor-bearing mice were treated with SU6668 in combination with B7.2-IgG, the antitumor effects were substantially higher than in mice treated separately with SU6668 or B7.2-IgG. Prolonged treatment of mice with SU6668 did not inhibit the immunoreactivity of T lymphocytes. On the contrary, T cells from mice treated with a combination of SU6668 and B7.2-IgG showed higher proliferative responses and cytokine production following anti-CD3 stimulation than T cells of mice treated separately with these modalities. These results indicate that antiangiogenic and immune therapies using SU6668 and B7.2-IgG are compatible and manifest complementary antitumor effects. Combined antiangiogenic and immune therapy might represent a new strategy for cancer treatment.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inmunoterapia , Indoles/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirroles/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Complejo CD3 , Línea Celular Tumoral , Inmunoglobulina G/uso terapéutico , Masculino , Ratones , Oxindoles , Propionatos , Neoplasias de la Próstata/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA