Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 146(6): 918-30, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21925315

RESUMEN

Inhibitors of DNA binding (IDs) antagonize basic-helix-loop-helix (bHLH) transcription factors to inhibit differentiation and maintain stem cell fate. ID ubiquitination and proteasomal degradation occur in differentiated tissues, but IDs in many neoplasms appear to escape degradation. We show that the deubiquitinating enzyme USP1 promotes ID protein stability and stem cell-like characteristics in osteosarcoma. USP1 bound, deubiquitinated, and thereby stabilized ID1, ID2, and ID3. A subset of primary human osteosarcomas coordinately overexpressed USP1 and ID proteins. USP1 knockdown in osteosarcoma cells precipitated ID protein destabilization, cell-cycle arrest, and osteogenic differentiation. Conversely, ectopic USP1 expression in mesenchymal stem cells stabilized ID proteins, inhibited osteoblastic differentiation, and enhanced proliferation. Consistent with USP1 functioning in normal mesenchymal stem cells, USP1-deficient mice were osteopenic. Our observations implicate USP1 in preservation of the stem cell state that characterizes osteosarcoma and identify USP1 as a target for differentiation therapy.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Neoplásicas/citología , Osteosarcoma/patología , Animales , Proteínas de Arabidopsis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteasas Ubiquitina-Específicas , Ubiquitinación
2.
Drug Metab Dispos ; 44(12): 1881-1889, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27638506

RESUMEN

Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. Limited treatment options have only marginally impacted patient survival over the past decades. The phophatidylinositol 3-kinase (PI3K) pathway, frequently altered in GBM, represents a potential target for the treatment of this glioma. 5-(6,6-Dimethyl-4-morpholino-8,9-dihydro-6H-[1,4]oxazino[4,3-e]purin-2-yl)pyrimidin-2-amine (GDC-0084) is a PI3K inhibitor that was specifically optimized to cross the blood-brain barrier. The goals of our studies were to characterize the brain distribution, pharmacodynamic (PD) effect, and efficacy of GDC-0084 in orthotopic xenograft models of GBM. GDC-0084 was tested in vitro to assess its sensitivity to the efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) and in vivo in mice to evaluate its effects on the PI3K pathway in intact brain. Mice bearing U87 or GS2 intracranial tumors were treated with GDC-0084 to assess its brain distribution by matrix-assisted laser desorption ionization (MALDI) imaging and measure its PD effects and efficacy in GBM orthotopic models. Studies in transfected cells indicated that GDC-0084 was not a substrate of P-gp or BCRP. GDC-0084 markedly inhibited the PI3K pathway in mouse brain, causing up to 90% suppression of the pAkt signal. MALDI imaging showed GDC-0084 distributed evenly in brain and intracranial U87 and GS2 tumors. GDC-0084 achieved significant tumor growth inhibition of 70% and 40% against the U87 and GS2 orthotopic models, respectively. GDC-0084 distribution throughout the brain and intracranial tumors led to potent inhibition of the PI3K pathway. Its efficacy in orthotopic models of GBM suggests that it could be effective in the treatment of GBM. GDC-0084 is currently in phase I clinical trials.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Glioblastoma/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular , Línea Celular Tumoral , Perros , Femenino , Glioblastoma/tratamiento farmacológico , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Indazoles/metabolismo , Indazoles/farmacología , Células de Riñón Canino Madin Darby , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/farmacología
3.
Development ; 137(22): 3753-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20978073

RESUMEN

In many organ systems such as the skin, gastrointestinal tract and hematopoietic system, homeostasis is dependent on the continuous generation of differentiated progeny from stem cells. The rodent incisor, unlike human teeth, grows throughout the life of the animal and provides a prime example of an organ that rapidly deteriorates if newly differentiated cells cease to form from adult stem cells. Hedgehog (Hh) signaling has been proposed to regulate self-renewal, survival, proliferation and/or differentiation of stem cells in several systems, but to date there is little evidence supporting a role for Hh signaling in adult stem cells. We used in vivo genetic lineage tracing to identify Hh-responsive stem cells in the mouse incisor and we show that sonic hedgehog (SHH), which is produced by the differentiating progeny of the stem cells, signals to several regions of the incisor. Using a hedgehog pathway inhibitor (HPI), we demonstrate that Hh signaling is not required for stem cell survival but is essential for the generation of ameloblasts, one of the major differentiated cell types in the tooth, from the stem cells. These results therefore reveal the existence of a positive-feedback loop in which differentiating progeny produce the signal that in turn allows them to be generated from stem cells.


Asunto(s)
Células Madre Adultas/metabolismo , Ameloblastos/citología , Proteínas Hedgehog/metabolismo , Incisivo/crecimiento & desarrollo , Ratones/fisiología , Transducción de Señal , Ameloblastos/metabolismo , Animales , Diferenciación Celular , Células Epiteliales/metabolismo , Femenino , Proteínas Hedgehog/antagonistas & inhibidores , Incisivo/citología
4.
J Pathol ; 227(4): 417-30, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22611036

RESUMEN

Resistance to anti-angiogenic therapy can occur via several potential mechanisms. Unexpectedly, recent studies showed that short-term inhibition of either VEGF or VEGFR enhanced tumour invasiveness and metastatic spread in preclinical models. In an effort to evaluate the translational relevance of these findings, we examined the consequences of long-term anti-VEGF monoclonal antibody therapy in several well-validated genetically engineered mouse tumour models of either neuroendocrine or epithelial origin. Anti-VEGF therapy decreased tumour burden and increased overall survival, either as a single agent or in combination with chemotherapy, in all four models examined. Importantly, neither short- nor long-term exposure to anti-VEGF therapy altered the incidence of metastasis in any of these autochthonous models, consistent with retrospective analyses of clinical trials. In contrast, we observed that sunitinib treatment recapitulated previously reported effects on tumour invasiveness and metastasis in a pancreatic neuroendocrine tumour (PNET) model. Consistent with these results, sunitinib treatment resulted in an up-regulation of the hypoxia marker GLUT1 in PNETs, whereas anti-VEGF did not. These results indicate that anti-VEGF mediates anti-tumour effects and therapeutic benefits without a paradoxical increase in metastasis. Moreover, these data underscore the concept that drugs targeting VEGF ligands and receptors may affect tumour metastasis in a context-dependent manner and are mechanistically distinct from one another.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Antiidiotipos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Metástasis de la Neoplasia/tratamiento farmacológico , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/inmunología , Adenocarcinoma/genética , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Modelos Animales de Enfermedad , Quimioterapia Combinada , Ingeniería Genética , Indoles/uso terapéutico , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Ratones , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Pirroles/uso terapéutico , Carcinoma Pulmonar de Células Pequeñas/genética , Sunitinib , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
5.
Proc Natl Acad Sci U S A ; 107(50): 21248-55, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21081700

RESUMEN

Priming of the organ-specific premetastatic sites is thought to be an important yet incompletely understood step during metastasis. In this study, we show that the metastatic tumors we examined overexpress granulocyte-colony stimulating factor (G-CSF), which expands and mobilizes Ly6G+Ly6C+ granulocytes and facilitates their subsequent homing at distant organs even before the arrival of tumor cells. Moreover, G-CSF-mobilized Ly6G+Ly6C+ cells produce the Bv8 protein, which has been implicated in angiogenesis and mobilization of myeloid cells. Anti-G-CSF or anti-Bv8 antibodies significantly reduced lung metastasis. Transplantation of Bv8 null fetal liver cells into lethally irradiated hosts also reduced metastasis. We identified an unexpected role for Bv8: the ability to stimulate tumor cell migration through activation of one of the Bv8 receptors, prokineticin receptor (PKR)-1. Finally, we show that administration of recombinant G-CSF is sufficient to increase the numbers of Ly6G+Ly6C+ cells in organ-specific metastatic sites and results in enhanced metastatic ability of several tumors.


Asunto(s)
Antígenos Ly/inmunología , Factor Estimulante de Colonias de Granulocitos/farmacología , Granulocitos/efectos de los fármacos , Granulocitos/inmunología , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular , Femenino , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/genética , Granulocitos/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Análisis por Micromatrices , Trasplante de Neoplasias , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
6.
Biochim Biophys Acta ; 1773(6): 1007-14, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17509701

RESUMEN

The factors that regulate trophoblast invasion of the uterine vasculature are incompletely understood. In this paper we show that macaque trophoblasts express the mucin, MUC1, and that it is involved in trophoblast-endothelial interaction. Immunocytochemistry, Western blotting and RT-PCR analyses confirmed that MUC1 was expressed by isolated early gestation macaque trophoblasts. MUC1 was also detected in endovascular trophoblasts in sections of placental-decidual tissue during early gestation. A blocking antibody against MUC1 reduced trophoblast adhesion to uterine endothelial cells and also blocked trophoblast transendothelial migration. MUC1 is known to bind to Intercellular Adhesion Molecule-1 (ICAM-1) in other systems. Incubation in the presence of a blocking antibody against Intercellular Adhesion Molecule-1 (ICAM-1) or recombinant ICAM-1 modestly, but significantly, reduced transendothelial trophoblast migration. These results are consistent with the idea that MUC1 is involved in trophoblast adhesion to uterine endothelial cells and in trophoblast transendothelial migration.


Asunto(s)
Comunicación Celular/fisiología , Movimiento Celular/fisiología , Decidua/fisiología , Endotelio Vascular/fisiología , Mucina-1/biosíntesis , Embarazo/fisiología , Trofoblastos/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Adhesión Celular/fisiología , Comunicación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Decidua/citología , Células Endoteliales/citología , Células Endoteliales/fisiología , Endotelio Vascular/citología , Femenino , Molécula 1 de Adhesión Intercelular/metabolismo , Macaca mulatta , Trofoblastos/citología
7.
Mol Cancer Ther ; 14(1): 40-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25376606

RESUMEN

Activating mutations in the KRAS oncogene occur in approximately 90% of pancreatic cancers, resulting in aberrant activation of the MAPK and the PI3K pathways, driving malignant progression. Significant efforts to develop targeted inhibitors of nodes within these pathways are underway and several are currently in clinical trials for patients with KRAS-mutant tumors, including patients with pancreatic cancer. To model MEK and PI3K inhibition in late-stage pancreatic cancer, we conducted preclinical trials with a mutant Kras-driven genetically engineered mouse model that faithfully recapitulates human pancreatic ductal adenocarcinoma development. Treatment of advanced disease with either a MEK (GDC-0973) or PI3K inhibitor (GDC-0941) alone showed modest tumor growth inhibition and did not significantly enhance overall survival. However, combination of the two agents resulted in a significant survival advantage as compared with control tumor-bearing mice. To model the clinical scenario, we also evaluated the combination of these targeted agents with gemcitabine, the current standard-of-care chemotherapy for pancreatic cancer. The addition of MEK or PI3K inhibition to gemcitabine, or the triple combination regimen, incrementally enhanced overall survival as compared with gemcitabine alone. These results are reminiscent of the survival advantage conferred in this model and in patients by the combination of gemcitabine and erlotinib, an approved therapeutic regimen for advanced nonresectable pancreatic cancer. Taken together, these data indicate that inhibition of MEK and PI3K alone or in combination with chemotherapy do not confer a dramatic improvement as compared with currently available therapies for patients with pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Azetidinas/administración & dosificación , Azetidinas/farmacología , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Indazoles/administración & dosificación , Indazoles/farmacología , Ratones , Modelos Biológicos , Mutación , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Piperidinas/administración & dosificación , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Nivel de Atención , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
8.
Neoplasia ; 15(7): 694-711, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23814482

RESUMEN

The phosphatidylinositol 3-kinase (PI3K) pathway is a central mediator of vascular endothelial growth factor (VEGF)-driven angiogenesis. The discovery of small molecule inhibitors that selectively target PI3K or PI3K and mammalian target of rapamycin (mTOR) provides an opportunity to pharmacologically determine the contribution of these key signaling nodes in VEGF-A-driven tumor angiogenesis in vivo. This study used an array of micro-vascular imaging techniques to monitor the antivascular effects of selective class I PI3K, mTOR, or dual PI3K/mTOR inhibitors in colorectal and prostate cancer xenograft models. Micro-computed tomography (micro-CT) angiography, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), vessel size index (VSI) MRI, and DCE ultrasound (DCE-U/S) were employed to quantitatively evaluate the vascular (structural and physiological) response to these inhibitors. GDC-0980, a dual PI3K/mTOR inhibitor, was found to reduce micro-CT angiography vascular density, while VSI MRI demonstrated a significant reduction in vessel density and an increase in mean vessel size, consistent with a loss of small functional vessels and a substantial antivascular response. DCE-MRI showed that GDC-0980 produces a strong functional response by decreasing the vascular permeability/perfusion-related parameter, K (trans). Interestingly, comparable antivascular effects were observed for both GDC-980 and GNE-490 (a selective class I PI3K inhibitor). In addition, mTOR-selective inhibitors did not affect vascular density, suggesting that PI3K inhibition is sufficient to generate structural changes, characteristic of a robust antivascular response. This study supports the use of noninvasive microvascular imaging techniques (DCE-MRI, VSI MRI, DCE-U/S) as pharmacodynamic assays to quantitatively measure the activity of PI3K and dual PI3K/mTOR inhibitors in vivo.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Inhibidores Enzimáticos , Neoplasias/diagnóstico , Neovascularización Patológica/diagnóstico , Angiografía/métodos , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Imagen Multimodal , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Pirimidinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Carga Tumoral/efectos de los fármacos , Ultrasonografía/métodos , Microtomografía por Rayos X/métodos
9.
Dev Cell ; 22(2): 403-17, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22340501

RESUMEN

Signaling events that regulate central nervous system (CNS) angiogenesis and blood-brain barrier (BBB) formation are only beginning to be elucidated. By evaluating the gene expression profile of mouse vasculature, we identified DR6/TNFRSF21 and TROY/TNFRSF19 as regulators of CNS-specific angiogenesis in both zebrafish and mice. Furthermore, these two death receptors interact both genetically and physically and are required for vascular endothelial growth factor (VEGF)-mediated JNK activation and subsequent human brain endothelial sprouting in vitro. Increasing beta-catenin levels in brain endothelium upregulate DR6 and TROY, indicating that these death receptors are downstream target genes of Wnt/beta-catenin signaling, which has been shown to be required for BBB development. These findings define a role for death receptors DR6 and TROY in CNS-specific vascular development.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/metabolismo , Neovascularización Fisiológica , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Biomarcadores/metabolismo , Western Blotting , Comunicación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunoprecipitación , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
10.
J Cell Biol ; 193(5): 935-51, 2011 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-21606205

RESUMEN

Melanoma inhibitory activity member 3 (MIA3/TANGO1) [corrected] is an evolutionarily conserved endoplasmic reticulum resident transmembrane protein. Recent in vitro studies have shown that it is required for the loading of collagen VII, but not collagen I, into COPII-coated transport vesicles. In this paper, we show that mice lacking Mia3 are defective for the secretion of numerous collagens, including collagens I, II, III, IV, VII, and IX, from chondrocytes, fibroblasts, endothelial cells, and mural cells. Collagen deposition by these cell types is abnormal, and extracellular matrix composition is compromised. These changes are associated with intracellular accumulation of collagen and the induction of a strong unfolded protein response, primarily within the developing skeleton. Chondrocyte maturation and bone mineralization are severely compromised in Mia3-null embryos, leading to dwarfism and neonatal lethality. Thus, Mia3's role in protein secretion is much broader than previously realized, and it may, in fact, be required for the efficient secretion of all collagen molecules in higher organisms.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/deficiencia , Colágeno/metabolismo , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
PLoS One ; 5(9): e12682, 2010 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-20856934

RESUMEN

ß-Catenin-dependent Wnt signaling is initiated as Wnt binds to both the receptor FZD and coreceptor LRP5/6, which then assembles a multimeric complex at the cytoplasmic membrane face to recruit and inactivate the kinase GSK3. The large number and sequence diversity of Wnt isoforms suggest the possibility of domain-specific ligand-coreceptor interactions, and distinct binding sites on LRP6 for Wnt3a and Wnt9b have recently been identified in vitro. Whether mechanistically different interactions between Wnts and coreceptors might mediate signaling remains to be determined. It is also not clear whether coreceptor homodimerization induced extracellularly can activate Wnt signaling, as is the case for receptor tyrosine kinases. We generated monoclonal antibodies against LRP6 with the unexpected ability to inhibit signaling by some Wnt isoforms and potentiate signaling by other isoforms. In cell culture, two antibodies characterized further show reciprocal activities on most Wnts, with one antibody antagonizing and the other potentiating. We demonstrate that these antibodies bind to different regions of LRP6 protein, and inhibition of signaling results from blocking Wnt binding. Antibody-mediated dimerization of LRP6 can potentiate signaling only when a Wnt isoform is also able to bind the complex, presumably recruiting FZD. Endogenous autocrine Wnt signaling in different tumor cell lines can be either antagonized or enhanced by the LRP6 antibodies, indicating expression of different Wnt isoforms. As anticipated from the roles of Wnt signaling in cancer and bone development, antibody activities can also be observed in mice for inhibition of tumor growth and in organ culture for enhancement of bone mineral density. Collectively, our results indicate that separate binding sites for different subsets of Wnt isoforms determine the inhibition or potentiation of signaling conferred by LRP6 antibodies. This complexity of coreceptor-ligand interactions may allow for differential regulation of signaling by Wnt isoforms during development, and can be exploited with antibodies to differentially manipulate Wnt signaling in specific tissues or disease states.


Asunto(s)
Anticuerpos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Proteínas Relacionadas con Receptor de LDL/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/metabolismo , Animales , Línea Celular , Humanos , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Ratones , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Acoplados a Proteínas G/genética , Especificidad de la Especie , Regulación hacia Arriba/efectos de los fármacos , Proteínas Wnt/genética
12.
Clin Cancer Res ; 15(21): 6674-82, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19861458

RESUMEN

PURPOSE: Little is known concerning the onset, duration, and magnitude of direct therapeutic effects of anti-vascular endothelial growth factor (VEGF) therapies. Such knowledge would help guide the rational development of targeted therapeutics from bench to bedside and optimize use of imaging technologies that quantify tumor function in early-phase clinical trials. EXPERIMENTAL DESIGN: Preclinical studies were done using ex vivo microcomputed tomography and in vivo ultrasound imaging to characterize tumor vasculature in a human HM-7 colorectal xenograft model treated with the anti-VEGF antibody G6-31. Clinical evaluation was by quantitative magnetic resonance imaging in 10 patients with metastatic colorectal cancer treated with bevacizumab. RESULTS: Microcomputed tomography experiments showed reduction in perfused vessels within 24 to 48 h of G6-31 drug administration (P

Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/tratamiento farmacológico , Diagnóstico por Imagen , Factor A de Crecimiento Endotelial Vascular/inmunología , Adolescente , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Bevacizumab , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Am J Reprod Immunol ; 59(2): 167-81, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18211542

RESUMEN

PROBLEM: We have used an in vitro co-culture system consisting of early gestation macaque trophoblasts cultured on top of human uterine microvascular endothelial cells (UtMVECs) to investigate the inflammatory response of endothelial cells to trophoblasts under shear stress conditions. METHOD: of study Uterine microvascular endothelial cells and trophoblasts were co-cultured in a parallel plate chamber under shear stress (15 dyn/cm(2)) conditions. The distribution and expression of endothelial intercellular adhesion molecule-1 (ICAM-1) was quantified by immunofluorescence image analysis and flow cytometry. Endothelial regulated upon activation normal T-cell expressed and secreted (RANTES) secretion was measured by enzyme-linked immunosorbent assay and permeability was assessed using fluorescein isothiocyanate-dextran. RESULTS: Intercellular adhesion molecule-1, but not vascular cell adhesion molecule-1 or platelet endothelial cell adhesion molecule-1, was re-distributed towards the downstream edge of endothelial cells when the cells were co-cultured with trophoblasts under shear stress conditions. Changes in ICAM-1 distribution were also observed when UtMVECs were co-cultured with trophoblast-conditioned medium under shear stress conditions. Incubation of UtMVECs with trophoblast-conditioned medium increased endothelial permeability, RANTES secretion, and trophoblast adhesion. CONCLUSION: These data support the idea that trophoblasts induce an inflammatory response in uterine endothelial cells that could enhance trophoblast invasion and transmigration.


Asunto(s)
Molécula 1 de Adhesión Intercelular/inmunología , Trofoblastos/inmunología , Útero/inmunología , Animales , Adhesión Celular/inmunología , Movimiento Celular/inmunología , Quimiocina CCL5/inmunología , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/inmunología , Femenino , Humanos , Inmunohistoquímica , Macaca mulatta , Microscopía Fluorescente , Microscopía de Contraste de Fase , Embarazo , Estrés Mecánico , Trofoblastos/citología , Útero/citología
14.
Leuk Lymphoma ; 49(10): 1963-75, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18949619

RESUMEN

Myelodysplastic syndromes (MDS) are common causes of ineffective hematopoiesis and cytopenias in the elderly. Various myelosuppressive and proinflammatory cytokines have been implicated in the high rates of apoptosis and hematopoietic suppression seen in MDS. We have previously shown that p38 MAPK is overactivated in MDS hematopoietic progenitors, which led to current clinical studies of the selective p38alpha inhibitor, SCIO-469, in this disease. We now demonstrate that the myelosuppressive cytokines TNFalpha and IL-1beta are secreted by bone marrow (BM) cells in a p38 MAPK-dependent manner. Their secretion is stimulated by paracrine interactions between BM stromal and mononuclear cells and cytokine induction correlates with CD34+ stem cell apoptosis in an inflammation-simulated in vitro bone marrow microenvironment. Treatment with SCIO-469 inhibits TNF secretion in primary MDS bone marrow cells and protects cytogenetically normal progenitors from apoptosis ex vivo. Furthermore, p38 inhibition diminishes the expression of TNFalpha or IL-1beta-induced proinflammatory chemokines in BM stromal cells. These data indicate that p38 inhibition has anti-inflammatory effects on the bone marrow microenvironment that complements its cytoprotective effect on progenitor survival. These findings support clinical investigation of p38alpha as a potential therapeutic target in MDS and other related diseases characterised by inflammatory bone marrow failure.


Asunto(s)
Médula Ósea/patología , Mediadores de Inflamación/antagonistas & inhibidores , Síndromes Mielodisplásicos/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Anciano , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Humanos , Indoles/farmacología , Inflamación/etiología , Interleucina-1beta/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Comunicación Paracrina/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA