Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Cancer Res ; 67(7): 3064-73, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17409413

RESUMEN

The Src homology and collagen (Src) family of adaptor proteins comprises six Shc-like proteins encoded by three loci in mammals (Shc, Rai, and Sli). Shc-like proteins are tyrosine kinase substrates, which regulate diverse signaling pathways and cellular functions, including Ras and proliferation (p52/p46Shc), phosphatidylinositol 3-kinase and survival (p54Rai), and mitochondrial permeability transition and apoptosis (p66Shc). Here, we report the identification, cloning, and sequence characterization of a new member of the Shc family that we termed RaLP. RaLP encodes a 69-kDa protein characterized by the CH2-PTB-CH1-SH2 modularity, typical of the Shc protein family, and expressed, among adult tissues, only in melanomas. Analysis of RaLP expression during the melanoma progression revealed low expression in normal melanocytes and benign nevi, whereas high levels of RaLP protein were found at the transition from radial growth phase to vertical growth phase and metastatic melanomas, when tumor cells acquire migratory competence and invasive potential. Notably, silencing of RaLP expression in metastatic melanomas by RNA interference reduced tumorigenesis in vivo. Analysis of RaLP in melanoma signal transduction pathways revealed that (a) when ectopically expressed in RaLP-negative melanocytes and nonmetastatic melanoma cells, it functions as a substrate of activated insulin-like growth factor-1 and epidermal growth factor receptors and increases Ras/mitogen-activated protein kinase (MAPK) signaling and cell migration, whereas (b) its silencing in RaLP-positive melanoma cells abrogates cell migration in vitro, without affecting MAPK signaling, suggesting that RaLP activates both Ras-dependent and Ras-independent migratory pathways in melanomas. These findings indicate that RaLP is a specific marker of metastatic melanomas, a critical determinant in the acquisition of the migratory phenotype by melanoma cells, and a potential target for novel anti-melanoma therapeutic strategies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular/fisiología , Melanoma/enzimología , Melanoma/patología , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Clonación Molecular , Regulación hacia Abajo , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Sistema de Señalización de MAP Quinasas , Melanoma/genética , Melanoma/secundario , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor IGF Tipo 1/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Transfección , Familia-src Quinasas/genética
2.
Mol Cell Biol ; 27(10): 3769-79, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17339329

RESUMEN

Polycomb group (PcG) proteins form multiprotein complexes, called Polycomb repressive complexes (PRCs). PRC2 contains the PcG proteins EZH2, SUZ12, and EED and represses transcription through methylation of lysine (K) 27 of histone H3 (H3). Suz12 is essential for PRC2 activity and its inactivation results in early lethality of mouse embryos. Here, we demonstrate that Suz12(-/-) mouse embryonic stem (ES) cells can be established and expanded in tissue culture. The Suz12(-/-) ES cells are characterized by global loss of H3K27 trimethylation (H3K27me3) and higher expression levels of differentiation-specific genes. Moreover, Suz12(-/-) ES cells are impaired in proper differentiation, resulting in a lack of repression of ES cell markers as well as activation of differentiation-specific genes. Finally, we demonstrate that the PcGs are actively recruited to several genes during ES cell differentiation, which despite an increase in H3K27me3 levels is not always sufficient to prevent transcriptional activation. In summary, we demonstrate that Suz12 is required for the establishment of specific expression programs required for ES cell differentiation. Furthermore, we provide evidence that PcGs have different mechanisms to regulate transcription during cellular differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas Represoras/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Lisina/metabolismo , Metilación , Ratones , Ratones Noqueados , Complejos Multiproteicos , Proteína Homeótica Nanog , Neuronas/citología , Neuronas/fisiología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Transcripción Genética
3.
Adv Exp Med Biol ; 522: 83-97, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12674213

RESUMEN

Measurement of tumor angiogenesis to predict and/or to assess the efficacy of antiangiogenic therapies is mainly based on the evaluation of microvessel density (MVD). We developed a novel flow cytometry procedure to measure circulating endothelial cells (CECs) and circulating endothelial cells progenitors (CECPs) in either preclinical and clinical studies. Preclinical studies were performed on an animal model of human lymphoma. A trend toward higher CECs values was observed on day 7 and 14 after transplant, and differences vs controls were highly significant on day 21 (p = 0.0061). A strong correlation was found between CECs and tumor volume (r = 0.942, p = 0.004) and between CECs and tumor-generated VEGF (r = 0.669, p = 0.02). In mice given cyclophosphamide, most of circulating apoptotic cells were hematopoietic and not endothelial. Conversely, in mice given endostatin, all of the increase in apoptotic cells was in the endothelial cell compartment. In a parallel study, we looked for CECs in the peripheral blood of 20 healthy controls and 76 newly diagnosed cancer patients by means of four-color flow cytometry. In breast cancer (n = 46) and lymphoma (n = 30) patients, both resting and activated CECs were increased by 5 fold (P < 0.0008 vs control). CECs significantly correlated with plasma levels of VCAM-1 and VEGF. Resting and activated CECs were similar to healthy controls in 7 lymphoma patients achieving complete remission after chemotherapy, and activated CECs were found to decrease in 13 breast cancer patients evaluated before and 24h after quadrantectomy. In conclusion, our findings indicate a close relation between CEC increase and tumor progression, and support CECs evaluation as a clinically relevant, non invasive angiogenesis marker. Furthermore, this assay offers insight into anti-angiogenic activity of different drugs.


Asunto(s)
Biomarcadores de Tumor/análisis , Endotelio Vascular/citología , Citometría de Flujo/métodos , Microcirculación/crecimiento & desarrollo , Microcirculación/patología , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/patología , Células Madre/citología , Inhibidores de la Angiogénesis/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citotoxinas/farmacología , Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/sangre , Endotelio Vascular/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Linfocinas/sangre , Linfoma/patología , Linfoma/fisiopatología , Masculino , Ratones , Ratones SCID , Microcirculación/fisiopatología , Neoplasias/fisiopatología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Células Madre/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Clin Cancer Res ; 9(1): 377-82, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12538491

RESUMEN

PURPOSE: We investigated the effect of endostatin on differentiation, mobilization, and clonogenic potential of circulating endothelial cell (EC) progenitors, and whether the effect of endostatin was improved by continuous infusion (CI) versus bolus administration. EXPERIMENTAL DESIGN: Four-color flow cytometry and clonogenic EC cultures were used to study EC progenitors in tumor-free mice, tumor-bearing immunodeficient mice, and immunodeficient mice xenotransplanted with human bone marrow (BM) cells. RESULTS: Endostatin significantly reduced the number of circulating EC progenitors in tumor-free BALB/c mice. The effect of endostatin on EC progenitors was enhanced significantly in mice treated with CI drug treatment. When immunodeficient mice xenotransplanted with human BM cells were treated with CI of endostatin we observed a significant decrease in the engraftment and differentiation of human BM-derived EC progenitors. Numbers of circulating EC progenitors increased 7-fold in immunodeficient mice bearing human lymphoma. In this preclinical model, treatment with CI of endostatin inhibited host murine EC progenitor mobilization and human tumor growth. Furthermore, the clonogenic potential of EC progenitors was impaired severely. CONCLUSIONS: Endostatin is a potent inhibitor of the mobilization and clonogenic potential of human and murine EC progenitors, and its preclinical activity is increased significantly in CI compared with bolus administration. These observations might be useful in the design of future clinical trials.


Asunto(s)
Antineoplásicos/uso terapéutico , Colágeno/administración & dosificación , Colágeno/uso terapéutico , Endotelio Vascular/citología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/uso terapéutico , Animales , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Colágeno/metabolismo , Endostatinas , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Trasplante de Neoplasias , Fragmentos de Péptidos/metabolismo , Células Madre , Factores de Tiempo
5.
Blood ; 100(8): 2989-95, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12351412

RESUMEN

Acute promyelocytic leukemia (APL) is associated with chromosomal translocations resulting in fusion proteins of the retinoic acid receptor (RAR). Here, we report a novel murine model system for APL, based on the transduction of purified murine hematopoietic progenitors (lin(-)) using high-titer retroviral vectors encoding promyelocytic leukemia-RAR (PML-RAR), and the green fluorescent protein (GFP) as a marker. PML-RAR-expressing lin(-) cells were impaired in their ability to undergo terminal myeloid differentiation and showed increased proliferative potential in vitro. Inoculation of transduced lin(-) cells into syngeneic, irradiated mice resulted in the development of retinoic acid-sensitive promyelocytic leukemias at high frequency (> 80%) and short latency (approximately 4 months). Morphologic and immunophenotypic analysis revealed no gross abnormalities of the preleukemic bone marrows. However, hematopoietic progenitors from PML-RAR preleukemic mice showed a severe impairment in their ability to undergo myeloid differentiation in vitro. This result, together with the monoclonality or oligoclonality of the leukemic blasts, supports a "multiple-hit" model, where the fusion protein causes a "preleukemic" phase, and leukemia occurs after additional genetic lesions. This model system faithfully reproduces the main characteristics of human APL and represents a versatile tool for the in vitro and in vivo study of mechanisms of leukemogenesis and the design of protocols for differentiation treatment.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia Promielocítica Aguda/genética , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/genética , Animales , Células de la Médula Ósea/patología , Diferenciación Celular , Supervivencia Celular , Hematopoyesis/efectos de la radiación , Leucemia Promielocítica Aguda/patología , Linfocitos/inmunología , Linfocitos/fisiología , Ratones , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Recombinantes/metabolismo , Retroviridae , Tasa de Supervivencia , Factores de Tiempo , Transfección
6.
Exp Hematol ; 30(8): 905-14, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12160842

RESUMEN

OBJECTIVE: We studied the in vitro and in vivo hematopoietic potential of human stem cells residing in muscle tissue collected from adults with head and neck cancer. MATERIALS AND METHODS: Adherent muscle cells were cultured in F12 medium with 10% fetal bovine serum and transplanted into immunodeficient mice. RESULTS: On day 12 we obtained a median of 500,000 adherent cells per gram muscle sample. Thy-1, endoglin, HER2/neu, and P1H12 were expressed in the majority of cells. CD34, VEGFR2, c-kit, VCAM-1, and CXCR4 were expressed in 0.5-1.5%, 1-5%, 1-15%, 9-15%, and 30% of cells, respectively. Immunodeficient mice transplanted with fresh muscle cells or less than 500,000 cultured cells showed little or no human engraftment. In mice transplanted with more than 500,000 cultured cells, up to 14% human CD45(+) hematopoietic cells (including myeloid and lymphoid subsets) were detected by flow cytometry. Engraftment was confirmed by polymerase chain reaction, Southern blotting, and DNA sequencing. Liver, muscle, and spleen evaluated for human DNA were positive in the majority of mice showing hematopoietic engraftment in the bone marrow. In vivo hematopoietic engraftment potential was maintained in cultured CD45(-) muscle cells transduced with the green fluorescence protein gene. CONCLUSIONS: Human stem cells residing in muscle tissue can generate multilineage hematopoiesis in immunodeficient mice. Surprisingly, this hematopoietic potential increased in cultured versus fresh cells from muscle tissue.


Asunto(s)
Células Madre Hematopoyéticas/citología , Músculo Esquelético/citología , Adulto , Animales , Adhesión Celular , Células Cultivadas/citología , Células Cultivadas/trasplante , Ensayo de Unidades Formadoras de Colonias , Femenino , Genes RAG-1 , Genes Reporteros , Supervivencia de Injerto , Neoplasias de Cabeza y Cuello/patología , Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Antígenos Comunes de Leucocito/biosíntesis , Antígenos Comunes de Leucocito/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , Factor de Transcripción PAX7 , Quimera por Radiación , Trasplante Heterólogo
7.
Mol Cell Biol ; 22(15): 5527-38, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12101245

RESUMEN

We report here the characterization of a mutant mouse line with a specific gene trap event in the Mdm4 locus. Absence of Mdm4 expression results in embryonic lethality (10.5 days postcoitum [dpc]), which was rescued by transferring the Mdm4 mutation into a Trp53-null background. Mutant embryos were characterized by overall growth deficiency, anemia, improper neural tube closure, and dilation of lateral ventricles. In situ analysis demonstrated increased levels of p21(CIP1/Waf1) and lower levels of Cyclin E and proliferating cell nuclear antigen expression. Consistent with lack of 5-bromo-2'-deoxyuridine incorporation, these data suggest a block of mutant embryo cells in the G(1) phase of the cell cycle. Accordingly, Mdm4-deficient mouse embryonic fibroblasts manifested a greatly reduced proliferative capacity in culture. Moreover, extensive p53-dependent cell death was specifically detected in the developing central nervous system of the Mdm4 mutant embryos. These findings unambiguously assign a critical role for Mdm4 as a negative regulator of p53 and suggest that Mdm4 could contribute to neoplasias retaining wild-type Trp53. Finally, we provide evidence indicating that Mdm4 plays no role on cell proliferation or cell cycle control that is distinct from its ability to modulate p53 function.


Asunto(s)
Anomalías Múltiples , Embrión de Mamíferos/anomalías , Neuronas/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Anomalías Múltiples/genética , Animales , Bromodesoxiuridina , Muerte Celular/genética , División Celular/genética , Células Cultivadas , Ciclina E/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Fibroblastos/citología , Fase G1 , Genes Letales , Ventrículos Laterales/anomalías , Ratones , Ratones Mutantes , Defectos del Tubo Neural/genética , Neuronas/patología , Fenotipo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , Rombencéfalo/metabolismo , Rombencéfalo/patología , Médula Espinal/metabolismo , Médula Espinal/patología , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA