Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Am J Pathol ; 173(1): 205-16, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18583324

RESUMEN

The transforming growth factor alpha (TGFalpha)/epidermal growth factor receptor (EGFR) signaling pathway appears to play a critical role in colon cancer progression, but the cellular and molecular mechanisms that contribute to metastasis remain unknown. KM12C colon cancer cell clones expressing high (C9) or negligible (C10) levels of TGFalpha were implanted into the cecal walls of nude mice. C9 tumors formed autocrine and paracrine EGFR networks, whereas C10 tumors were unable to signal through EGFR. The tumor microenvironment of C9, but not C10, contained cells enriched in vascular endothelial growth factor (VEGF) A, interleukin-8, and matrix metalloproteinases-2 and -9 and had a high vascular surface area. C9 tumors recruited a macrophage population that co-expressed F4/80 and lymphatic vessel endothelial hyaluronic acid receptor and produced VEGFC. The mean lymphatic density of C9 tumors was threefold higher than that of C10 tumors. C9, but not C10, tumor cells metastasized to regional lymph nodes in all mice and to the liver in 5 of 10 mice. Forced expression of TGFalpha in C10 tumor cells led to the generation of autocrine and paracrine EGFR signaling, macrophage recruitment, enhanced blood and lymphatic vascular surface areas, and increased lymphatic metastasis. Collectively, these data show that activation of TGFalpha-EGFR signaling in colon cancer cells creates a microenvironment that is conducive for metastasis, providing a rationale for efforts to inhibit EGFR signaling in TGFalpha-positive colon cancers.


Asunto(s)
Neoplasias del Colon/patología , Receptores ErbB/metabolismo , Metástasis de la Neoplasia/patología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador alfa/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Neovascularización Patológica/patología , Transfección
2.
Mol Cell Biol ; 25(1): 44-59, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15601829

RESUMEN

hTid-1, a human homolog of the Drosophila tumor suppressor l(2)Tid and a novel DnaJ protein, regulates the activity of nuclear factor kappaB (NF-kappaB), but its mechanism is not established. We report here that hTid-1 strongly associated with the cytoplasmic protein complex of NF-kappaB-IkappaB through direct interaction with IkappaBalpha/beta and the IKKalpha/beta subunits of the IkappaB kinase complex. These interactions resulted in suppression of the IKK activity in a J-domain-dependent fashion and led to the cytoplasmic retention and enhanced stability of IkappaB. Overexpression of hTid-1 by using recombinant baculovirus or adenovirus led to inhibition of cell proliferation and induction of apoptosis of human osteosarcoma cells regardless of the p53 expression status. Adherent cultured cells transduced with Ad.hTid-1 detached from the dish surface. Morphological changes consistent with apoptosis and cell death were evident 48 h after Ad.EGFP-hTid-1 transduction. In contrast, cells transduced with Ad.EGFP or Ad.EGFP-hTd-1DeltaN100, a mutant that has the N-terminal J domain deletion and that lost suppressive activity on IKK, continued to proliferate. Similar data were obtained with A375 human melanoma cells. Ad.EGFP or Ad.EGFP-hTd-1DeltaN100 ex vivo-transduced A375 cells injected subcutaneously into nude mice produced growing tumors, whereas Ad.EGFP-hTid-1-transduced cells did not. Collectively, the data suggest that hTid-1 represses the activity of NF-kappaB through physical and functional interactions with the IKK complex and IkappaB and, in doing so, it modulates cell growth and death.


Asunto(s)
Proteínas de Choque Térmico/fisiología , FN-kappa B/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Agar/metabolismo , Animales , Apoptosis , Baculoviridae/metabolismo , Muerte Celular , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Citoplasma/metabolismo , Drosophila , Eliminación de Gen , Regulación de la Expresión Génica , Genes Reporteros , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas del Choque Térmico HSP40 , Proteínas de Choque Térmico/química , Humanos , Proteínas I-kappa B/metabolismo , Inmunoprecipitación , Lentivirus/genética , Masculino , Ratones , Ratones Desnudos , Microscopía Fluorescente , Mutación , Trasplante de Neoplasias , Osteosarcoma/metabolismo , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Sales de Tetrazolio/farmacología , Tiazoles/farmacología , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/metabolismo
3.
Mol Cancer Ther ; 6(10): 2652-63, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17913856

RESUMEN

Epidermal growth factor receptor (EGFR) has been extensively targeted in the treatment of non-small cell lung cancer, producing responses in a small number of patients. To study the role of ligand expression in mediating response to EGFR antagonism, we injected NCI-H441 [EGFR and EGF/transforming growth factor-alpha (TGF-alpha) positive] or PC14-PE6 (EGFR positive and EGF/TGF-alpha negative) human lung adenocarcinoma cells into the lungs of nude mice. We randomized the mice to receive treatment with the EGFR tyrosine kinase inhibitors gefitinib or AEE788 or vehicle. Treatment of mice bearing NCI-H441 but not PC14-PE6 lung tumors resulted in a significant reduction in primary tumor growth, pleural effusion, and lymph node metastasis. Immunohistochemical analyses revealed that NCI-H441 and PC14-PE6 cells expressed EGFR but that the expression of EGF/TGF-alpha was high in NCI-H441 cells and very low in PC14-PE6 cells. Consequently, EGFR was activated in both tumor and tumor-associated endothelial cells in the NCI-H441 tumors but not in the PC14-PE6 tumors. Antagonism of EGFR signaling by treatment of mice with AEE788 decreased proliferation and increased apoptosis of both tumor cells and tumor-associated endothelial cells in NCI-H441 tumors but not in PC14-PE6 tumors. However, after transfection of PC14-PE6 cells with TGF-alpha, lung tumors derived from the transfected cells expressed and activated EGFR in both tumor and tumor-associated endothelial cells and tumors responded to treatment with AEE788. Collectively, these results strongly suggest that the response of human lung cancers growing orthotopically in mice to the inhibition of EGFR signaling is determined by ligand (EGF/TGF-alpha) expression by tumor cells. Our findings provide an additional explanation for the susceptibility of lung cancers to treatment with EGFR tyrosine kinase inhibitors.


Asunto(s)
Adenocarcinoma/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Adenocarcinoma/patología , Animales , Antineoplásicos/farmacología , Western Blotting , Proliferación Celular/efectos de los fármacos , Gefitinib , Dosificación de Gen , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Purinas/farmacología , Quinazolinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Res ; 65(24): 11529-35, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16357162

RESUMEN

Angiogenesis, the development of new blood vessels from preexisting vessels, is crucial to tissue growth, repair, and maintenance. This process begins with the formation of endothelial cell sprouts followed by the proliferation and migration of neighboring endothelial cells along the preformed extensions. The initiating event and mechanism of sprouting is not known. We show that the phenotypic expression of negatively charged membrane surface in apoptotic cells initiates the formation of directional endothelial cell sprouts that extend toward the dying cells by a mechanism that involves endothelial cell membrane hyperpolarization and cytoskeleton reorganization but is independent of diffusible molecules.


Asunto(s)
Apoptosis , Comunicación Celular , Movimiento Celular/fisiología , Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Neovascularización Fisiológica/fisiología , Electricidad Estática , Animales , Células Cultivadas , Técnicas de Cocultivo , Receptores ErbB/antagonistas & inhibidores , Ferritinas/metabolismo , Canales Iónicos/antagonistas & inhibidores , Melanoma/metabolismo , Melanoma/patología , Potenciales de la Membrana , Ratones , Fosfolípidos/metabolismo , Purinas/farmacología , Ratas , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Sefarosa/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
5.
Clin Cancer Res ; 11(13): 4923-33, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16000591

RESUMEN

PURPOSE: We determined whether the administration of the tyrosine kinase inhibitor, AEE788, which targets the epidermal growth factor receptor and the vascular endothelial growth factor receptor, alone or in combination with paclitaxel, can inhibit progressive growth of human ovarian carcinoma in the peritoneal cavity of female nude mice. EXPERIMENTAL DESIGN: Western blot analysis and immunohistochemical analysis identified the optimal dose and schedule of AEE788 therapy. In several different experiments, paclitaxel-sensitive and paclitaxel-resistant human ovarian carcinoma cells were injected into the peritoneal cavity of nude mice. Seven days later, treatment with saline (control), AEE788 alone, paclitaxel alone, or a combination of AEE788 and paclitaxel began and continued for 45 days when the mice were necropsied. In independent survival experiments, the mice were necropsied when they became moribund. RESULTS: Oral administration of AEE788 inhibited phosphorylation of the epidermal growth factor receptor and vascular endothelial growth factor receptor for up to 48 hours. Treatment with AEE788 plus paclitaxel significantly reduced tumor weight and increased survival of mice implanted with paclitaxel-sensitive cell lines compared with control mice or mice treated with AEE788 alone or paclitaxel alone. In mice implanted with paclitaxel-resistant cells, the combination therapy also significantly reduced tumor weight but did not prolong survival. The combination therapy induced apoptosis of both tumor cells and tumor-associated endothelial cells. CONCLUSIONS: The administration of AEE788 and paclitaxel inhibits the progression of human ovarian carcinoma in the peritoneal cavity of female nude mice, in part, by inducing apoptosis of tumor-associated endothelial cells.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Purinas/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Western Blotting , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Desnudos , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/patología , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Antígeno Nuclear de Célula en Proliferación/análisis , Purinas/administración & dosificación , Purinas/uso terapéutico , Análisis de Supervivencia , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
J Natl Cancer Inst ; 95(6): 458-70, 2003 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-12644539

RESUMEN

BACKGROUND: Expression of platelet-derived growth factor (PDGF) and activation (by autophosphorylation) of its receptor (PDGF-R), a tyrosine kinase, are associated with the growth of metastatic prostate tumor cells in the bone parenchyma. The tyrosine kinase inhibitor STI571 blocks the PDGF signaling pathway by inhibiting PDGF-R autophosphorylation. We examined the effects of STI571, given alone or with paclitaxel (Taxol), on tumor growth in a mouse model of prostate cancer metastasis. METHODS: Human prostate cancer PC-3MM2 cells were injected into the tibias of male nude mice. Three days later the mice (20 per group) were randomly assigned to 5 weeks of treatment with oral and injected water (control), daily oral STI571, weekly injected paclitaxel, or STI571 plus paclitaxel. Lesions in bone and the surrounding muscles were then harvested and analyzed by histology, western blotting (for PDGF-R phosphorylation), immunohistochemistry (for expression of proangiogenic molecules), and double immunofluorescence (to identify endothelial cells and apoptotic tumor cells). Growth of bone lesions was monitored by digital radiography. Bone lesions from control mice were used to establish short-term cell cultures for analysis of PDGF-R phosphorylation. All statistical tests were two-sided. RESULTS: PC-3MM2 cells cultured from bone lesions and treated in vitro with STI571 had less phosphorylated PDGF-R than untreated cells. In control mice, bone lesions expressed high levels of PDGF and activated (i.e., phosphorylated) PDGF-R, whereas lesions in the adjacent musculature did not. Activated PDGF-R was present on the surface of endothelial cells within the bone lesions but not in endothelial cells of uninjected bone. Mice treated with STI571 or STI571 plus paclitaxel had a lower tumor incidence, smaller tumors, and less bone lysis and lymph node metastasis than mice treated with water or paclitaxel alone (P<.001 for all). Mice treated with STI571 or STI571 plus paclitaxel had less phosphorylated PDGF-R on tumor cells and tumor-associated endothelial cells, less tumor cell proliferation, statistically significantly more apoptotic tumor cells (all P<.001), and fewer tumor-associated endothelial cells (P<.001) than control mice. CONCLUSIONS: Endothelial cells appear to express phosphorylated PDGF-R when they are exposed to tumor cells that express PDGF. Using STI571 to inhibit PDGF-R phosphorylation may, especially in combination with paclitaxel, produce substantial therapeutic effects against prostate cancer bone metastasis.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Inhibidores Enzimáticos/farmacología , Piperazinas/farmacología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos Fitogénicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Benzamidas , Western Blotting , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/enzimología , Neoplasias Óseas/secundario , División Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mesilato de Imatinib , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Desnudos , Microcirculación/efectos de los fármacos , Neoplasias Experimentales , Paclitaxel/administración & dosificación , Fosforilación/efectos de los fármacos , Piperazinas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Pirimidinas/administración & dosificación , Intensificación de Imagen Radiográfica , Receptores del Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Células Tumorales Cultivadas
7.
Cancer Res ; 63(11): 2971-6, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12782605

RESUMEN

Microvascular endothelial cells play a critical role in tumor progression and metastasis by forming capillary networks that encourage tumor growth and by promoting the attachment of circulating tumor cells to the vascular wall of distant tissues. Efforts to study the molecular mechanisms that mediate these complex processes in different anatomical compartments have been impeded by difficulties in the isolation and propagation of endothelial cells from different organs. To overcome these limitations, we used two-color flow cytometry to identify and select microvascular endothelial cells from primary cultures obtained from different organs of mice whose tissues harbor a temperature-sensitive SV40 large T antigen (H-2K(b)-tsA58 mice; ImmortoMice). The selection strategy targeted cell populations expressing the inducible endothelial cell adhesion molecules, E-selectin and VCAM-1, and proved successful in generating microvascular endothelial cell lines from a number of different organs. When cultured under permissive temperatures (33 degrees C), individual cell lines displayed doubling times consistent with endothelial cells possessing an angiogenic phenotype. The transfer of endothelial cells to nonpermissive temperatures (37 degrees C) resulted in cell differentiation and the induction of a quiescent state. Established cell lines exhibited several inherent endothelial properties, including the expression of constitutive and inducible levels of endothelial cell adhesion molecules E-selectin, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1, internalization of acetylated low-density lipoprotein, and formation of loop structures on Matrigel surfaces. The immortalized endothelial cell lines established from H-2K(b)-tsA58 mice provide, for the first time, a cell culture system to examine factors regulating angiogenesis and tumor cell arrest in different organ systems.


Asunto(s)
Endotelio Vascular/citología , Neoplasias/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Animales , División Celular/fisiología , Línea Celular , Colágeno , Combinación de Medicamentos , Selectina E/biosíntesis , Endotelio Vascular/metabolismo , Femenino , Citometría de Flujo , Antígenos H-2/inmunología , Humanos , Inmunohistoquímica , Molécula 1 de Adhesión Intercelular/biosíntesis , Laminina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Metástasis de la Neoplasia , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Técnicas de Cultivo de Órganos , Proteoglicanos , Molécula 1 de Adhesión Celular Vascular/biosíntesis
8.
Cancer Res ; 64(12): 4201-8, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15205332

RESUMEN

Once prostate cancer metastasizes to bone, conventional chemotherapy is largely ineffective. We hypothesized that inhibition of phosphorylation of the epidermal growth factor receptor (EGF-R) and platelet-derived growth factor receptor (PDGF-R) expressed on tumor cells and tumor-associated endothelial cells, which is associated with tumor progression, in combination with paclitaxel would inhibit experimental prostate cancer bone metastasis and preserve bone structure. We tested this hypothesis in nude mice, using human PC-3MM2 prostate cancer cells. PC-3MM2 cells growing adjacent to bone tissue and endothelial cells within these lesions expressed phosphorylated EGF-R and PDGF-R alpha and -beta on their surfaces. The percentage of positive endothelial cells and the intensity of receptor expression directly correlated with proximity to bone tissue. Oral administration of PKI166 inhibited the phosphorylation of EGF-R but not PDGF-R, whereas oral administration of STI571 inhibited the phosphorylation of PDGF-R but not EGF-R. Combination therapy using oral PKI166 and STI571 with i.p. injections of paclitaxel induced a high level of apoptosis in tumor vascular endothelial cells and tumor cells in parallel with inhibition of tumor growth in the bone, preservation of bone structure, and reduction of lymph node metastasis. Collectively, these data demonstrate that blockade of phosphorylation of EGF-R and PDGF-R coupled with administration of paclitaxel significantly suppresses experimental human prostate cancer bone metastasis.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Receptores ErbB/antagonistas & inhibidores , Paclitaxel/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Benzamidas , Neoplasias Óseas/metabolismo , División Celular/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Receptores ErbB/metabolismo , Receptores ErbB/fisiología , Humanos , Mesilato de Imatinib , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Desnudos , Fosforilación , Piperazinas/administración & dosificación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/metabolismo , Pirimidinas/administración & dosificación , Pirroles/administración & dosificación , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Especificidad por Sustrato , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Res ; 64(11): 3727-30, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15172974

RESUMEN

The activation of the microvascular endothelial cell platelet-derived growth factor (PDGF) receptor (PDGF-R) by PDGF has been implicated in neoplastic angiogenesis. Here, we established cultures of murine bone microvascular endothelial cells and examined their response to stimulation with PDGF BB ligand and to blockade of PDGF-R signaling with the tyrosine kinase inhibitor STI571 (Gleevec). The addition of STI571 to cultures of bone endothelial cells blocked PDGF BB-induced phosphorylation in a dose-dependent manner and completely abrogated the activation of downstream targets Akt and ERK1/2. Coadministration of STI571 and Taxol also induced the activation of procaspase-3 and significant apoptosis. These data suggest that phosphorylation of PDGF-R stimulates survival pathways in bone endothelial cells and that by selectively inhibiting PDGF-R signaling with STI571, the cells are rendered sensitive to Taxol treatment. The therapeutic combination of STI571 and Taxol may be a powerful tool for targeting tumor-associated endothelial cells in the skeletal compartment.


Asunto(s)
Huesos/citología , Huesos/enzimología , Células Endoteliales/citología , Células Endoteliales/enzimología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Becaplermina , Benzamidas , Huesos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Mesilato de Imatinib , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Paclitaxel/farmacología , Fosforilación , Piperazinas/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-sis , Pirimidinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
10.
Neoplasia ; 7(12): 1065-72, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16354589

RESUMEN

Tumor cells and tumor-associated endothelial cells express activated epidermal growth factor receptor (EGFR) due to production of EGF-related ligands in the tumor microenvironment. To investigate the effect of perpetual EGFR activation on endothelial cells, we developed a novel method to generate constitutively active EGFR. We fused the entire intracellular domain of the EGFR to the N-terminus of the CD3zeta component of the T-cell receptor signaling complex. Expression of the chimeric receptor CD3-EGFR in EGFR-deficient human embryonic kidney cells resulted in ligand-independent sustained EGFR phosphorylation and in the induction of Akt, mitogen-activated protein kinase, and signal transducer and activator of transcription 3 (Stat3). Next, CD3-EGFR was stably expressed in murine brain endothelial cells where it signaled for the initiation of angiogenic programs, Stat3 activation, and continuous proliferation. A comparison between brain endothelial cells encoding CD3zeta and CD3-EGFR revealed that proangiogenic phenotype was modulated by the intracellular effector Stat3 and that suppression of this downstream target with the EGFR tyrosine kinase inhibitor PKI166 could revert this phenotype. Thus, our results validate the use of chimeric constitutively active receptors to replicate critical features observed in pathophysiological processes that can expedite the identification of novel therapeutic agents targeting EGFR activation and function.


Asunto(s)
Complejo CD3/metabolismo , Receptores ErbB/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Encéfalo/metabolismo , Complejo CD3/genética , Proliferación Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Humanos , Riñón/metabolismo , Ligandos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fenotipo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
11.
Clin Cancer Res ; 9(17): 6534-44, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695158

RESUMEN

PURPOSE: We evaluated the expression of platelet-derived growth factor (PDGF) ligands and receptors in clinical specimens of human pancreatic adenocarcinomas and determined the therapeutic effect of STI571 (Gleevec), a protein tyrosine kinase inhibitor of PDGF receptor (PDGFR), on human pancreatic carcinoma cells growing in the pancreas and liver of nude mice. EXPERIMENTAL DESIGN: Immunohistochemical staining for PDGF-AA and -BB ligands, PDGFR-alpha and -beta, and phosphorylated PDGFR-alpha and -beta was performed on 31 specimens of human pancreatic cancer and L3.6pl human pancreatic adenocarcinoma cell line. To determine the in vivo effects of STI571, nude mice with L3.6pl cells injected into the pancreas were randomized 7 days later to receive one of the following treatments: sterile water p.o. (control), STI571, gemcitabine, or a combination of STI571 and gemcitabine. RESULTS: In 29 of 31 clinical specimens of human pancreatic adenocarcinoma, both tumor cells and tumor-associated endothelial cells expressed phosphorylated PDGFR-alpha and -beta. L3.6pl cells growing in culture expressed moderate amounts of PDGF-AA and little to no PDGFR-alpha or -beta, whereas L3.6pl cells growing in the pancreas of nude mice expressed a high level of PDGF and receptors. Colocalization immunohistochemical analysis demonstrated expression of activated PDGFR-beta by tumor-associated endothelial cells in both the pancreas and in liver metastases. Tumors of mice treated for 4 weeks with STI571 (50 mg/kg or 100 mg/kg p.o. daily) were slightly smaller than controls. Tumors treated with gemcitabine and STI571 (50 mg/kg) were >70% smaller than tumors in control mice and 36% smaller than those in mice treated with gemcitabine only (P < 0.0002 and P < 0.04, respectively). Combination therapy also inhibited spontaneous metastasis to the liver. Tumors from mice treated with both STI571 and gemcitabine had decreased expression of activated (phosphorylated) PDGFR-alpha and -beta, decreased mean vessel density, decreased cell proliferation, and increased apoptosis of tumor cells. CONCLUSIONS: Collectively, these data show that activated PDGFR on tumor cells and tumor-endothelial cells can be a novel target for therapy of pancreatic carcinoma.


Asunto(s)
Carcinoma/patología , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Piperazinas/farmacología , Pirimidinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Adenocarcinoma/metabolismo , Animales , Antineoplásicos/farmacología , Becaplermina , Benzamidas , Western Blotting , División Celular , Línea Celular Tumoral , Humanos , Mesilato de Imatinib , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ligandos , Ratones , Ratones Desnudos , Microscopía Fluorescente , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-sis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Tiempo
12.
Clin Exp Metastasis ; 20(5): 421-30, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14524531

RESUMEN

To further characterize the role of hepatocyte growth factor-scatter factor (HGF-SF) and its receptor (c-Met) in osteosarcoma (OS), human OS cell lines with low (SAOS-2) and high (SAOS-LM2) metastatic potential, and cell lines derived from spontaneous canine OS were studied. All cell lines were evaluated for c-Met and HGF-SF expression and receptor activation using Northern, RT-PCR, and Western blot analyses, respectively. Functional activity of receptor-ligand interaction was measured using c-Met phosphorylation status, proliferation assays (anchorage-dependent and -independent), Matrigel invasion, modulation of urokinase plasminogen activator (uPA) expression, and cell dispersion (scattering). All cell lines exhibited steady-state mRNA expression of c-Met. The canine OS cell lines also expressed HGF-SF mRNA as determined by RT-PCR analysis. Western analysis showed c-Met protein expression and HGF-stimulated (human) or constitutive (canine) receptor autophosphorylation. Treatment with recombinant human HGF resulted in enhanced proliferation in 3 of 5 OS cell lines and enhanced colony formation in 2 of 5 OS cell lines. Matrigel invasion was significantly enhanced in 3 of the cell lines and uPA levels were significantly increased in the SAOS-2 cells following HGF treatment. Scattering was enhanced in both the SAOS-2 and SAOS-LM2 cells. These data support the involvement of c-Met and HGF-SF in the growth and progression of human and canine OS, and may offer new targets for the development of therapeutic strategies for OS.


Asunto(s)
Osteosarcoma/metabolismo , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Proto-Oncogénicas c-met/fisiología , Animales , Northern Blotting , Western Blotting , División Celular , Colágeno/farmacología , Perros , Combinación de Medicamentos , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Laminina/farmacología , Ligandos , Metástasis de la Neoplasia , Hibridación de Ácido Nucleico , Fosforilación , Proteoglicanos/farmacología , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
13.
Cancer Res ; 69(3): 828-35, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19141644

RESUMEN

Murine melanomas produce site-specific experimental brain metastases that reflect clinical reality. When injected into the internal carotid artery of mice, K-1735 melanoma cells produce metastatic lesions only in the brain parenchyma, whereas B16 melanoma cells and the somatic hybrid cells of B16 x K-1735 melanoma cells produce metastatic lesions only in the leptomeninges and ventricles. In the present study, we identified transforming growth factor-beta2 (TGF-beta2), an isoform of the TGF-beta family, as a molecular determinant of melanoma cell growth in the brain parenchyma. We found that the TGF-beta2 mRNA was highly expressed by the K-1735 cells, whereas the B16 cells or any B16 x K-1735 somatic cell-cell fusion hybrids have low expression. Transfection of the TGF-beta2 gene into B16 cells resulted in the production of microscopic metastatic lesions in the brain parenchyma, without a decrease in metastasis to the leptomeninges or ventricles. TGF-beta2 knockdown in the K-1735 melanoma cells significantly reduced metastasis to the brain parenchyma but did not induce metastasis to the leptomeninges or ventricles. These data show that TGF-beta2 expression by murine melanoma cells is necessary for the establishment and growth of metastases in the brain parenchyma.


Asunto(s)
Neoplasias Encefálicas/secundario , Melanoma Experimental/secundario , Factor de Crecimiento Transformador beta2/biosíntesis , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Femenino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Invasividad Neoplásica , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Transfección , Factor de Crecimiento Transformador beta2/genética
14.
Cancer Cell ; 13(5): 385-93, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18455122

RESUMEN

Expression of the epidermal growth factor receptor (EGFR), a receptor tyrosine kinase associated with cell proliferation and survival, is overactive in many tumors of epithelial origin. Blockade of the kinase activity of EGFR has been used for cancer therapy; however, by itself, it does not seem to reach maximum therapeutic efficacy. We report here that in human cancer cells, the function of kinase-independent EGFR is to prevent autophagic cell death by maintaining intracellular glucose level through interaction and stabilization of the sodium/glucose cotransporter 1 (SGLT1).


Asunto(s)
Receptores ErbB/fisiología , Neoplasias/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transportador 1 de Sodio-Glucosa/fisiología , Autofagia , Muerte Celular , División Celular , Supervivencia Celular , Receptores ErbB/deficiencia , Receptores ErbB/genética , Glucosa/metabolismo , Homeostasis , Humanos , Cinética , Metástasis de la Neoplasia , ARN Interferente Pequeño/genética , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores
15.
Neoplasia ; 9(12): 1066-77, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18084614

RESUMEN

The purpose of our study was to determine whether the dual inhibition of epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) signaling pathways in tumor-associated endothelial cells can inhibit the progressive growth of human colon carcinoma in the cecum of nude mice. SW620CE2 human colon cancer cells growing in culture and orthotopically in the cecum of nude mice expressed a high level of transforming growth factor alpha (TGF-alpha) and vascular endothelial growth factor (VEGF) but were negative for EGFR, human epidermal growth factor receptor 2 (HER2), and VEGFR. Double immunofluorescence staining revealed that tumor-associated endothelial cells expressed EGFR, VEGFR2, phosphorylated EGFR (pEGFR), and phosphorylated VEGFR (pVEGFR). Treatment of mice with either 7H-pyrrolo [2,3-d]-pyrimidine lead scaffold (AEE788; an inhibitor of EGFR and VEGFR tyrosine kinase) or CPT-11 as single agents significantly inhibited the growth of cecal tumors (P < .01); this decrease was even more pronounced with AEE788 combined with CPT-11 (P < .001). AEE788 alone or combined with CPT-11 also inhibited the expression of pEGFR and pVEGFR on tumor-associated endothelial cells, significantly decreased vascularization and tumor cell proliferation, and increased the level of apoptosis in both tumor-associated endothelial cells and tumor cells. These data demonstrate that targeting EGFR and VEGFR signaling on tumor-associated endothelial cells provides a viable approach for the treatment of colon cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Camptotecina/análogos & derivados , Neoplasias del Colon/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Purinas/uso terapéutico , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Camptotecina/administración & dosificación , Camptotecina/farmacología , Camptotecina/uso terapéutico , Neoplasias del Ciego/tratamiento farmacológico , Neoplasias del Ciego/enzimología , Neoplasias del Ciego/patología , Línea Celular Tumoral/trasplante , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Células Endoteliales/enzimología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Irinotecán , Masculino , Ratones , Ratones Desnudos , Proteínas de Neoplasias/análisis , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/administración & dosificación , Purinas/farmacología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Neoplasia ; 8(7): 543-50, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16867216

RESUMEN

Direct injection of murine K-1735 melanoma cells into the subcutis, lung, or brain of syngeneic mice produces amelanotic tumors, whereas intravenous injection into the lateral tail vein or internal carotid artery produces both amelanotic and melanotic foci in the lung and the brain respectively. We hypothesized that loss of adhesion in the circulation may contribute to the melanogenic phenotypes of cells. To test this, we used enforced suspension culture of K-1735 cells by consistent rotating culture of K-1735 cells. We found that the expression of the microphthalmia transcription factor (MITF) and melanin-stimulating hormone receptor (MSHR) were upregulated in cells growing in suspension and were accompanied by inhibitions of AKT and ERK, which were reversed in cells upon regrowth as an adherent monolayer. Inhibition of the AKT pathway was responsible for MITF induction by suspension culture. Stable expression of constitutively active AKT significantly repressed the melanogenesis of K-1735 cells injected via circulation. An amelanotic clone of K-1735 cells was resistant to suspension culture-induced MITF, although the inhibition of AKT pathway was intact. Collectively, these data suggest that the inhibition of AKT pathway due to loss of adhesion within the circulation renders a subpopulation of K-1735 cells to produce melanin.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Melanoma Amelanótico/patología , Melanoma/patología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Animales , Adhesión Celular , Línea Celular Tumoral , Inmunohistoquímica , Melanoma/metabolismo , Melanoma Amelanótico/metabolismo , Ratones , Ratones Endogámicos C3H , Factor de Transcripción Asociado a Microftalmía/metabolismo , Metástasis de la Neoplasia , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Receptores de la Hormona Hipofisaria/metabolismo
17.
Prostate ; 59(2): 167-76, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15042617

RESUMEN

BACKGROUND: Increased expression of the hepatocyte growth factor (HGF) receptor (MET) is associated with high-grade prostatic adenocarcinoma and metastasis. However, the mechanism through which MET signaling contributes to prostate cancer (CaP) metastasis remains unclear. METHODS: Human PC-3 CaP cells and in vivo selected, isogeneic variant cells of increasing metastatic potential (PC-3M, PC-3M-Pro4, and PC-3M-LN4) were used to investigate the effect of HGF on CaP cell growth, protease production, and invasion. Cell-free urokinase-type plasminogen activator (u-PA) expression and function following HGF treatment were analyzed by Western blot, ELISA, and casein/plasminogen zymography. In vitro invasion stimulated by HGF was measured using Matrigel-coated invasion chambers. RESULTS: Both mRNA and functional protein for MET were detected in each of the CaP cell lines. HGF treatment (0-40 ng/ml) weakly increase proliferation, however, HGF induced soluble u-PA protein and activity 3-fold in the metastatic variant cells. HGF significantly stimulated the invasion of highly metastatic PC-3M-LN4 cells through Matrigel and treatment with specific urokinase receptor inhibitors diminished the HGF-stimulated invasion in a dose-dependent manner. CONCLUSIONS: These results demonstrate the biological significance of u-PA up-regulation in response to HGF in highly metastatic hormone refractory CaP cells.


Asunto(s)
Adenocarcinoma/patología , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/farmacología , Invasividad Neoplásica/fisiopatología , Activadores Plasminogénicos/farmacología , Neoplasias de la Próstata/patología , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , Antineoplásicos Hormonales/farmacología , División Celular , Resistencia a Antineoplásicos , Humanos , Masculino , Transducción de Señal , Células Tumorales Cultivadas , Regulación hacia Arriba
18.
Am J Pathol ; 161(3): 929-38, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12213721

RESUMEN

The purpose of this study was to determine whether the expression of epidermal growth factor receptor (EGF-R) and activated EGF-R by tumor-associated endothelial cells is influenced by interaction with specific growth factors in the microenvironment. Different human carcinoma cell lines expressing EGF-R with low or high levels of EGF/transforming growth factor (TGF)-alpha were implanted into orthotopic organs of nude mice. In the EGF/TGF-alpha-positive bladder cancer (253J-BV), pancreatic cancer (L3.6pl), and renal cancer (RBM1-IT) but not in the EGF/TGF-alpha-negative renal cancer SN12-PM6, tumor-associated endothelial cells expressed EGF-R and activated EGF-R. Mice were implanted with human 253J-BV bladder tumors (EGF+) or human SN12-PM6 renal tumors (EGF-). Treatment with oral PKI 166 (a specific inhibitor of EGF-R phosphorylation) alone, intraperitoneal paclitaxel alone (253J-BV), gemcitabine alone (SN12-PM6), or combination of PKI 166 and chemotherapy produced a 60%, 32%, or 81% reduction in the volume of 253J-BV bladder tumors, respectively, and 26%, 23%, or 51% reduction in the volume of SN12-PM6 kidney tumors, respectively. Immunohistochemical analyses demonstrated down-regulation of activated EGF-R in EGF/TGF-alpha-positive and EGF/TGF-alpha-negative lesions from mice treated with PKI 166, although apoptosis of tumor-associated endothelial cells was found only in EGF/TGF-alpha-positive tumors. Collectively, these data suggest that expression of activated EGF-R by tumor-associated endothelial cells provides an important target for therapy.


Asunto(s)
Desoxicitidina/análogos & derivados , Endotelio Vascular/metabolismo , Receptores ErbB/antagonistas & inhibidores , Neoplasias Experimentales/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Desoxicitidina/administración & dosificación , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neovascularización Patológica/tratamiento farmacológico , Paclitaxel/administración & dosificación , Pirimidinas/administración & dosificación , Pirroles/administración & dosificación , Células Tumorales Cultivadas , Gemcitabina
19.
J Cell Biochem ; 92(1): 77-91, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15095405

RESUMEN

To further define the role of insulin-like growth factor-1 (IGF-1) and its receptor (IGF-1R) in osteosarcoma (OS), human OS cell lines with low (SAOS-2) and high (SAOS-LM2) metastatic potential and three canine OS-derived cell lines were studied. Cell lines were evaluated for: IGF-1R expression; expression of IGF binding proteins (IGFBPs); effect of IGF-1 on tumor cell growth, invasion, expression of urokinase plasminogen activator (uPA), and soluble uPA receptor (suPAR), and; ectopic and orthotopic tumorigenicity of the canine OS cells in athymic mice. All cell lines exhibited steady-state mRNA expression of IGF-1R. The SAOS-2 and SAOS-LM2 cells expressed 9,138 and 10,234 cell-associated binding sites, respectively. Canine OS cells expressed from 1,728 to 3,883 binding sites. Two IGF-1-treated cell lines displayed enhanced proliferation. Two cell lines formed colonies in semisolid media, and IGF-1 increased colony number. Matrigel invasion was enhanced in one cell line following IGF-1 treatment. uPA and suPAR were unchanged in SAOS-2 and SAOS-LM2 cells following IGF-1 treatment, but the highly metastatic OS line SAOS-LM2 expressed five times more suPAR and displayed enhanced invasion compared to the parental, low metastatic SAOS-2. IGFBP-5 was detected in four of five cell lines, and IGFBP-3 was detected in two canine OS cell lines. Two canine OS lines were tumorigenic, and one metastasized spontaneously. In conclusion, OS cells express IGF-1R, which can contribute to their growth and invasion. There is suggestive evidence that increasing receptor number may contribute to in vivo tumorigenesis. Additional studies are needed to determine how IGF-1/IGF-1R interactions contribute to the malignant phenotype of OS.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/secundario , Receptor IGF Tipo 1/metabolismo , Animales , Línea Celular , Perros , Expresión Génica , Humanos , Pulmón/patología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Unión Proteica , Receptor IGF Tipo 1/genética , Trasplante Heterólogo , Activador de Plasminógeno de Tipo Uroquinasa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA