Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Breast Cancer Res Treat ; 132(1): 75-85, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21553120

RESUMEN

Mammosphere culture has been used widely for the enrichment of mammary epithelial stem cells and breast cancer stem cells (CSCs). Epithelial-to-mesenchymal transition (EMT) also induces stem cell features in normal and transformed mammary cells. We examined whether mammosphere culture conditions per se induced EMT in the epithelial MCF-7 breast cancer cell line. MCF-7 cells were cultured as mammospheres for 5 weeks, with dispersal and reseeding at the end of each week. This mammosphere culture induced a complete EMT by 3 weeks. Return of the cells to standard adherent culture conditions in serum-supplemented media generated a cell population (called MCF-7(M) cells), which displays a stable mesenchymal and CSC-like CD(44+)/CD(24-/low) phenotype. EMT was accompanied by a stable, marked increase in EMT-associated transcription factors and mesenchymal markers, and a decrease in epithelial markers and estrogen receptor α (ERα). MCF-7(M) cells showed increased motility, proliferation and chemoresistance in vitro, and produced larger tumors in immunodeficient mice with or without estrogen supplementation. MicroRNA analysis showed suppression of miR-200c, miR-203, and miR-205; and increases in miR-222 and miR-221. Antisense hairpin RNA inhibitor targeting miR-221 resulted in re-expression of ERα in MCF-7(M) cells. This study provides the first example of mammosphere culture conditions inducing EMT and of EMT regulating microRNAs that target ERα.


Asunto(s)
Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Esferoides Celulares/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Ratones , Ratones SCID , MicroARNs/genética , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Fenotipo , Carga Tumoral/genética
2.
Oncogene ; 29(48): 6378-89, 2010 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-20729917

RESUMEN

The v-Crk oncogene product consists of two protein interaction modules, a Src homology 2 (SH2) domain and a Src homology 3 (SH3) domain. Overexpression of CrkI, the cellular homolog of v-Crk, transforms mouse fibroblasts, and elevated CrkI expression is observed in several human cancers. The SH2 and SH3 domains of Crk are required for transformation, but the identity of the critical cellular binding partners is not known. A number of candidate Crk SH3-binding proteins have been identified, including the nonreceptor tyrosine kinases c-Abl and Arg, and the guanine nucleotide exchange proteins C3G, SOS1 and DOCK180. The aim of this study is to determine which of these are required for transformation by CrkI. We found that short hairpin RNA-mediated knockdown of C3G or SOS1 suppressed anchorage-independent growth of NIH-3T3 cells overexpressing CrkI, whereas knockdown of SOS1 alone was sufficient to suppress tumor formation by these cells in nude mice. Knockdown of C3G was sufficient to revert morphological changes induced by CrkI expression. By contrast, knockdown of Abl family kinases or their inhibition with imatinib enhanced anchorage-independent growth and tumorigenesis induced by Crk. These results show that SOS1 is essential for CrkI-induced fibroblast transformation, and also reveal a surprising negative role for Abl kinases in Crk transformation.


Asunto(s)
Transformación Celular Neoplásica , Proteínas Proto-Oncogénicas c-crk/fisiología , Dominios Homologos src , Animales , Apoptosis , Factor 2 Liberador de Guanina Nucleótido/fisiología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Desnudos , Células 3T3 NIH , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/fisiología , Proteínas Proto-Oncogénicas c-crk/química , Proteína SOS1/fisiología , Transducción de Señal
3.
Oncogene ; 25(16): 2379-92, 2006 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-16369494

RESUMEN

The process of cancer cell invasion involves degradation of the extracellular matrix (ECM) by proteases, integrin adhesion and cell motility. The role of ECM degrading proteases on the hypoxia-induced invasion of breast carcinoma cells was investigated. Hypoxia markedly increased the invasion capacity of MDA-MB-231 and MDA-MB-435 breast carcinoma cell lines. Matrix metalloproteinase (MMP) inhibitors blocked the hypoxia-induced invasion, whereas other protease inhibitors had no effect. Antibodies or siRNAs blocking either membrane type-1 MMP (MT1-MMP) or MMP-2 were effective in reducing the hypoxia-induced invasion. Serum-free reconstitution experiments confirmed the involvement of the MT1-MMP/MMP-2/tissue inhibitor of metalloproteinase-2 complex in this hypoxia-induced response. Overexpression of MT1-MMP in a poorly invasive breast cancer cell line, T47-D, promoted hypoxia-induced invasion and MMP-2 activation. Cell surface accumulation and activation of MT1-MMP without apparent regulation at the mRNA or protein levels indicated a post-translational adaptive response to hypoxia. Inhibition of the small GTPase RhoA eliminated the hypoxia-induced invasion and blocked the localization of MT1-MMP to the plasma membrane. Zymographic and molecular analysis of human breast tumors showed a strong correlation between hypoxic microenvironments and MMP-2 activation without changes in MT1-MMP expression. Our studies suggest that hypoxic tumor microenvironments promote breast cancer invasion through an MT1-MMP-dependent mechanism.


Asunto(s)
Neoplasias de la Mama/patología , Hipoxia de la Célula , Metaloproteinasa 2 de la Matriz/fisiología , Metaloproteinasas de la Matriz/fisiología , Línea Celular Tumoral , Activación Enzimática , Femenino , Humanos , Metaloproteinasas de la Matriz Asociadas a la Membrana , Invasividad Neoplásica , Interferencia de ARN , ARN Mensajero/análisis , Inhibidor Tisular de Metaloproteinasa-2/fisiología , Proteína de Unión al GTP rhoA/fisiología
4.
Growth Factors ; 19(1): 19-34, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11678207

RESUMEN

Aberrant expression of vascular endothelial growth factor (VEGF) has been demonstrated to be associated with most human solid tumors. Here we report that TGF-beta potently induces VEGF expression in human HT-1080 fibrosarcomas primarily through transcriptional activation with no significant changes in mRNA turnover. The tyrosine kinase inhibitor genistein and AP-1 inhibitor curcumin significantly blocked TGF-beta induction of VEGF expression while SP-1 and MKK1 inhibitors did not. TGF-beta enhanced both AP-1 and HIF-1 DNA binding activities whereas SP-1, AP-2 and NF-1 did not show major changes. Transcriptional reporter assays provided further evidence that TGF-beta augmented both AP-1 and HIF-1 activities. Moreover, TGF-beta-treated HT-1080 cells contained higher levels of HIF-1alpha and c-jun proteins in nuclear extracts. TGF-beta and hypoxia synergistically induced VEGF mRNA expression. Given the fact that most tumors respond to hypoxic stress with increased VEGF expression via HIF-1-dependent transcription, this study identifies for the first time that TGF-beta also increases VEGF mRNA in an AP-l/HIF-1-dependent mechanism and may potentiate the hypoxic response.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factores de Crecimiento Endotelial/biosíntesis , Linfocinas/biosíntesis , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Western Blotting , Hipoxia de la Célula , Factores de Crecimiento Endotelial/genética , Inhibidores Enzimáticos/farmacología , Genes Reporteros , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Linfocinas/genética , Proteínas Proto-Oncogénicas c-jun/análisis , ARN Mensajero/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
5.
Lab Invest ; 81(1): 61-75, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11204275

RESUMEN

Angiogenesis is a key component of human cancer progression and metastasis. In an effort to recapitulate early events in tumor-induced angiogenesis, we have employed a subcutaneous Matrigel implant model using immunodeficient mice as hosts. Matrigel-containing fibroblast growth factor 2 (FGF-2; 1.2 microg/ml) induced stromal cell infiltration into the Matrigel/skin interface within 4 days and maximal neovascularization at 7 days. Cells staining positive for the endothelial cell marker, platelet-endothelial cell adhesion molecule 1 (PECAM-1), were present in neovessels and in isolated cells within the Matrigel matrix. Immunohistochemical analysis revealed high levels of vascular endothelial growth factor (VEGF) deposited in the stromal interface present only in the FGF-2-containing but not in control Matrigel implants. VEGF expression was confirmed with in situ hybridization. High VEGF mRNA levels were observed in the infiltrating stromal cells but not in endothelial or endothelial precursors as defined by PECAM-1 staining. In vitro analysis of FGF-2-treated embryonic fibroblasts, Balb/c 3T3 cells, showed an induction of VEGF transcription, mRNA synthesis, and protein secretion as defined by transcriptional reporter, Northern blot, and ELISA assays. The FGF-2-induced VEGF expression was not dependent on select matrix adherence or signaling components because VEGF mRNA expression induced by FGF-2 was equally activated on serum, basement membrane, and fibronectin matrix substrates. Systemic application of anti-VEGF antibodies significantly repressed FGF-2-induced angiogenesis over control antibody by 88% (p < 0.001). These data support an FGF-2 angiogenic model that is dependent on endothelial cell activation, stromal cell infiltration, and VEGF expression by the infiltrating stromal cell population.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Linfocinas/metabolismo , Neovascularización Patológica , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células 3T3 , Animales , Factores de Crecimiento Endotelial/genética , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Linfocinas/genética , Ratones , Ratones Desnudos , Neoplasias/irrigación sanguínea , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
6.
Am J Pathol ; 156(1): 159-67, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10623663

RESUMEN

Inhibition of the vascular endothelial growth factor (VEGF) receptor Flk-1 has been shown to prevent invasion of experimental squamous cell carcinomas (SCC). To directly investigate the role of VEGF in tumor invasion, we stably transfected human SCC-13 cells, which are characterized by a noninvasive phenotype in vivo, with expression vectors containing murine VEGF(164) in sense (SCC/VEGF+) or antisense (SCC/VEGF-) orientation or with vector alone (SCC/vec). SCC/vec cells formed slowly growing, well-differentiated tumors with well-defined borders between tumor and stroma, after intradermal or subcutaneous injection. In contrast, SCC/VEGF+ tumors were characterized by rapid tumor growth, with small cell groups and single cells invading into the surrounding tissue, and by admixture of blood vessels and tumor cells in areas of tumor invasion. We detected an increase in tumor vessel density and size in VEGF-overexpressing tumors, resulting in a more than fourfold increase in total vascular areas. In contrast, SCC/VEGF- clones formed noninvasive, sharply circumscribed tumors with reduced vascular density. These findings demonstrate that selective VEGF overexpression was sufficient to induce tumor invasiveness, and they provide further evidence for an active role of the tumor stroma in cancer progression.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Linfocinas/metabolismo , Animales , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/patología , División Celular , ADN Complementario/genética , Factores de Crecimiento Endotelial/genética , Humanos , Enfermedades del Sistema Inmune/genética , Linfocinas/genética , Ratones , Ratones Endogámicos BALB C/genética , Invasividad Neoplásica/genética , Trasplante de Neoplasias , Neovascularización Patológica/patología , Oligonucleótidos/farmacología , Oligonucleótidos Antisentido/farmacología , Fenotipo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Transfección , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
7.
Cell ; 99(3): 301-12, 1999 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-10555146

RESUMEN

Vascular endothelial cells undergo morphogenesis into capillary networks in response to angiogenic factors. We show here that sphingosine-1-phosphate (SPP), a platelet-derived bioactive lipid, activates the EDG-1 and -3 subtypes of G protein-coupled receptors on endothelial cells to regulate angiogenesis. SPP induces the Gi/mitogen-activated protein kinase/cell survival pathway and the small GTPase Rho- and Raccoupled adherens junction assembly. Both EDG-1-and EDG-3-regulated signaling pathways are required for endothelial cell morphogenesis into capillary-like networks. Indeed, SPP synergized with polypeptide angiogenic growth factors in the formation of mature neovessels in vivo. These data define SPP as a novel regulator of angiogenesis.


Asunto(s)
Cadherinas/fisiología , Adhesión Celular/fisiología , Endotelio Vascular/fisiología , Proteínas I-kappa B , Uniones Intercelulares/fisiología , Lisofosfolípidos , Neovascularización Fisiológica/fisiología , Receptores Acoplados a Proteínas G , Esfingosina/análogos & derivados , Animales , Antígenos CD , Cadherinas/análisis , Calcio/metabolismo , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/efectos de los fármacos , Femenino , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/fisiología , Uniones Intercelulares/efectos de los fármacos , Ratones , Ratones Desnudos , Modelos Biológicos , Morfogénesis/efectos de los fármacos , Inhibidor NF-kappaB alfa , Neovascularización Fisiológica/efectos de los fármacos , Oocitos/fisiología , Receptores de Superficie Celular/fisiología , Receptores Lisofosfolípidos , Proteínas Recombinantes/metabolismo , Esfingosina/farmacología , Venas Umbilicales , Xenopus laevis
8.
Cancer Res ; 59(20): 5392-7, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10537325

RESUMEN

Gangliosides are sialated glycosphingolipids present on the plasma membranes of all vertebrate cells. Tumors shed gangliosides into the extracellular microenvironment, which may influence tumor-host cell interactions. We have investigated the role of gangliosides on the growth and angiogenesis of the EPEN experimental mouse brain tumor. EPEN cells express only ganglioside G(M3), and the solid tumors formed in vivo are sparsely vascularized with extensive necrosis. We stably transfected the EPEN cells with the cDNA for N-acetylgalactosaminyl transferase, a key enzyme for the synthesis of complex gangliosides. In addition to G(M3), the transfected cell line (EPEN-GNT) expressed complex gangliosides G(M2), G(M1), and G(D1a). The EPEN-GNT tumor was more densely vascularized with less necrosis and grew more rapidly than the nontransfected EPEN or mock-transfected (EPEN-V) control tumors. Also, VEGF gene expression was higher in the EPEN-GNT tumor than in the control tumors. The synthesis of complex gangliosides in the EPEN-GNT tumor cells also stimulated vascularization in an in vivo Matrigel assay for angiogenesis. These results indicate that the ratio of G(M3) to complex gangliosides can influence the growth and angiogenic properties of the EPEN experimental brain tumor and are consistent with previous findings in other systems. We conclude that gangliosides may be important modulators of brain tumor angiogenesis.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Gangliósidos/fisiología , Neovascularización Patológica/etiología , Animales , Factores de Crecimiento Endotelial/genética , Gangliósidos/genética , Regulación Neoplásica de la Expresión Génica , Linfocinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Índice Mitótico , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
9.
J Biol Chem ; 274(22): 15407-14, 1999 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-10336429

RESUMEN

Aberrant expression of the potent angiogenic cytokine, vascular endothelial growth factor (VEGF), has been demonstrated to be associated with most human solid tumors. Both transcriptional and post-transcriptional mechanisms have been shown to modulate VEGF expression in a multitude of cell types. Here we report that when protein kinase C (PKC) pathways were activated in human glioblastoma U373 cells by phorbol 12-myristate 13-acetate (PMA), VEGF mRNA expression was up-regulated via a post-transcriptional mRNA stabilization mechanism. PMA treatment exhibited no increase in VEGF-specific transcriptional activation as determined by run-off transcription assays and VEGF promoter-luciferase reporter assays. However, PMA increased VEGF mRNA half-life from 0.8 to 3.6 h which was blocked by PKC inhibitors but not by protein kinase A or cyclic nucleotide-dependent protein kinase inhibitors. When U373 cells were transfected with antisense oligonucleotide sequences to the translation start sites of PKC-alpha, -beta, -gamma, -delta, -epsilon, or -zeta isoforms, both PKC-alpha and -zeta antisense oligonucleotides showed substantial inhibition of PMA-induced VEGF mRNA. In addition, overexpression of PKC-zeta resulted in a strong constitutive up-regulation of VEGF mRNA expression. This study demonstrates for the first time that specific PKC isoforms regulate VEGF mRNA expression through post-transcriptional mechanisms.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Isoenzimas/metabolismo , Linfocinas/genética , Proteína Quinasa C/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Glioblastoma/genética , Humanos , Oligonucleótidos Antisentido/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Estaurosporina/farmacología , Activación Transcripcional , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
10.
Am J Physiol ; 276(5): E849-55, 1999 05.
Artículo en Inglés | MEDLINE | ID: mdl-10329978

RESUMEN

Physiological stressors such as sepsis and tissue damage initiate an acute immune response and cause transient systemic insulin resistance. This study was conducted to determine whether tumor necrosis factor-alpha (TNF-alpha), a cytokine produced by immune cells during skeletal muscle damage, decreases insulin responsiveness at the cellular level. To examine the molecular mechanisms associated with TNF-alpha and insulin action, we measured insulin receptor substrate (IRS)-1- and IRS-2-mediated phosphatidylinositol 3-kinase (PI 3-kinase) activation, IRS-1-PI 3-kinase binding, IRS-1 tyrosine phosphorylation, and the phosphorylation of two mitogen-activated protein kinases (MAPK, known as p42(MAPK) and p44(MAPK)) in cultured C2C12 myotubes. Furthermore, we determined the effects of TNF-alpha on insulin-stimulated 2-deoxyglucose (2-DG) uptake. We observed that TNF-alpha impaired insulin stimulation of IRS-1- and IRS-2-mediated PI 3-kinase activation by 54 and 55% (P < 0.05), respectively. In addition, TNF-alpha decreased insulin-stimulated IRS-1 tyrosine phosphorylation by 40% (P < 0.05). Furthermore, TNF-alpha repressed insulin-induced p42(MAPK) and p44(MAPK) tyrosine phosphorylation by 81% (P < 0.01). TNF-alpha impairment of insulin signaling activation was accompanied by a decrease (P < 0.05) in 2-DG uptake in the muscle cells (60 +/- 4 vs. 44 +/- 6 pmol. min-1. mg-1). These data suggest that increases in TNF-alpha may cause insulin resistance in skeletal muscle by inhibiting IRS-1- and IRS-2-mediated PI 3-kinase activation as well as p42(MAPK) and p44(MAPK) tyrosine phosphorylation, leading to impaired insulin-stimulated glucose uptake.


Asunto(s)
Glucosa/metabolismo , Insulina/farmacología , Proteínas Quinasas Activadas por Mitógenos , Músculos/efectos de los fármacos , Músculos/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Desoxiglucosa/metabolismo , Activación Enzimática , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/farmacología , Fosforilación , Fosfotirosina/metabolismo
11.
J Biol Chem ; 274(3): 1359-65, 1999 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9880507

RESUMEN

A 126-base region of human vascular endothelial growth factor (VEGF) 3'-untranslated region, which we identified as the hypoxia stability region, forms seven hypoxia-inducible RNA-protein complexes with apparent molecular masses ranging from 40 to 90 kDa in RNA-UV-cross-linking assays. In this study, we show that proteins that form the 60-kDa RNA-protein complex with the hypoxia stability region were present in both cytoplasmic and nuclear compartments. We purified the protein associated in the 60-kDa complex and identified it as heterogeneous nuclear ribonucleoprotein L (hnRNP L) by protein sequencing. Removal of hnRNP L by immunoprecipitation specifically abolished formation of the 60-kDa complex. Synthetic deoxyribonucleotide competition studies defined the RNA-binding site of hnRNP L as a 21-base-long sequence, 5'-CACCCACCCACAUACAUACAU-3'. Immunoprecipitation of hnRNP L followed by reverse transcription-polymerase chain reaction showed that hnRNP L specifically interacts with VEGF mRNA in hypoxic cells in vivo. Furthermore, when M21 cells transfected with antisense oligodeoxyribonucleotide to the hnRNP L RNA-binding site, the VEGF mRNA half-life was significantly reduced under hypoxic conditions. Thus, we propose that specific association of hnRNP L with VEGF mRNA under hypoxia may play an important role in hypoxia-induced post-transcriptional regulation of VEGF mRNA expression.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Hipoxia/metabolismo , Linfocinas/genética , ARN Nuclear Heterogéneo/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Secuencia de Bases , Sitios de Unión , Electroforesis en Gel de Poliacrilamida , Ribonucleoproteína Heterogénea-Nuclear Grupo L , Ribonucleoproteínas Nucleares Heterogéneas , Humanos , Datos de Secuencia Molecular , Peso Molecular , Oligodesoxirribonucleótidos Antisentido/metabolismo , Procesamiento Postranscripcional del ARN , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
12.
J Biol Chem ; 273(41): 26277-80, 1998 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-9756852

RESUMEN

The transcription factor Sp1 is ubiquitously expressed and plays a significant role in the constitutive and induced expression of a variety of mammalian genes and may even contribute to tumorigenesis. Here, we describe a novel pathway whereby Sp1 promotes the transcription of vascular permeability factor/vascular endothelial growth factor (VPF/VEGF), a potent angiogenic factor, by interacting directly and specifically with protein kinase C zeta (PKC zeta) isoform in renal cell carcinoma. PKC zeta binds and phosphorylates the zinc finger region of Sp1. Moreover, in the presence of the wild type von Hippel-Lindau gene product, the interaction of Sp1 with PKC zeta is inhibited, and in this manner steady state levels of Sp1 phosphorylation are decreased significantly. Co-transfection of renal cell carcinoma cells and human fibrosarcoma cells with a plasmid overexpressing PKC zeta and VPF/VEGF promoter luciferase constructs results in activation of Sp1-mediated transcription, whereas expression of a dominant-negative mutant of PKC zeta repressed this activation. Taken together, our results suggest a new pathway of cell signaling through PKC zeta and provide an insight into PKC zeta and Sp1-dependent transcriptional regulation of VPF/VEGF expression and thus tumor angiogenesis.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Ligasas , Linfocinas/genética , Proteína Quinasa C/metabolismo , Factor de Transcripción Sp1/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Fibrosarcoma/enzimología , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
13.
J Exp Med ; 188(6): 1135-45, 1998 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-9743532

RESUMEN

Vascular permeability factor/vascular endothelial cell growth factor (VPF/VEGF) can both potently enhance vascular permeability and induce proliferation of vascular endothelial cells. We report here that mouse or human mast cells can produce and secrete VPF/VEGF. Mouse mast cells release VPF/VEGF upon stimulation through Fcepsilon receptor I (FcepsilonRI) or c-kit, or after challenge with the protein kinase C activator, phorbol myristate acetate, or the calcium ionophore, A23187; such mast cells can rapidly release VPF/VEGF, apparently from a preformed pool, and can then sustain release by secreting newly synthesized protein. Notably, the Fc epsilonRI-dependent secretion of VPF/VEGF by either mouse or human mast cells can be significantly increased in cells which have undergone upregulation of Fc epsilonRI surface expression by a 4-d preincubation with immunoglobulin E. These findings establish that at least one cell type, the mast cell, can be stimulated to secrete VPF/VEGF upon immunologically specific activation via a member of the multichain immune recognition receptor family. Our observations also identify a new mechanism by which mast cells can contribute to enhanced vascular permeability and/or angiogenesis, in both allergic diseases and other settings.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Inmunoglobulina E/fisiología , Linfocinas/metabolismo , Mastocitos/metabolismo , Receptores de IgE/biosíntesis , Regulación hacia Arriba/inmunología , Animales , Calcimicina/farmacología , Línea Celular , Células Cultivadas , Dinitrofenoles/inmunología , Dinitrofenoles/farmacología , Factores de Crecimiento Endotelial/biosíntesis , Factores de Crecimiento Endotelial/genética , Femenino , Haptenos/farmacología , Humanos , Linfocinas/biosíntesis , Linfocinas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de IgE/fisiología , Albúmina Sérica/inmunología , Albúmina Sérica/farmacología , Factor de Células Madre/farmacología , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética/inmunología , Cordón Umbilical/citología , Cordón Umbilical/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
14.
Cancer Res ; 58(18): 4185-92, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9751633

RESUMEN

Vascular endothelial growth factor (VEGF), also known as vascular permeability factor, has been investigated as a potent mediator of brain tumor angiogenesis and tumor growth. We evaluated the effect of VEGF expression on the pathophysiology of tumor growth in the brain. Human SK-MEL-2 melanoma cells, with minimal VEGF expression, were stably transfected with either sense or antisense mouse VEGF cDNA and used to produce intracerebral xenografts. Vascular permeability, blood volume, blood flow, and tumor fluorodeoxyglucose metabolism were assessed using tissue sampling and quantitative autoradiography. Tumor proliferation was assessed by measuring bromodeoxyuridine labeling indices. Tumor vascular density and morphological status of the blood-brain barrier were evaluated by immunohistochemistry. SK-MEL-2 cells transfected with sense VEGF (V+) expressed large amounts of mouse and human VEGF protein; V+ cells formed well-vascularized, rapidly growing tumors with minimal tumor necrosis. V+ tumors had substantial and significant increases in blood volume, blood flow, vascular permeability, and fluorodeoxyglucose metabolism compared to wild-type and/or V- (antisense VEGF) tumors. VEGF antisense transfected V- expressed no detectable VEGF protein and formed minimally vascularized tumors. V- tumors had a very low initial growth rate with central necrosis; blood volume, blood flow, vascular permeability, and glucose metabolism levels were low compared to wild-type and V+ tumors. A substantial inhibition of intracerebral tumor growth, as well as a decrease in tumor vascularity, blood flow, and vascular permeability may be achieved by down-regulation of endogenous VEGF expression in tumor tissue. VEGF-targeted antiangiogenic gene therapy could be an effective component of a combined strategy to treat VEGF-producing brain tumors.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Factores de Crecimiento Endotelial/metabolismo , Linfocinas/metabolismo , Melanoma/irrigación sanguínea , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica , Animales , Volumen Sanguíneo , Neoplasias Encefálicas/metabolismo , Permeabilidad Capilar , Circulación Cerebrovascular , Factores de Crecimiento Endotelial/genética , Ensayo de Inmunoadsorción Enzimática , Fluorodesoxiglucosa F18/metabolismo , Humanos , Linfocinas/genética , Melanoma/metabolismo , Ratones , Proteínas de Neoplasias/genética , ARN sin Sentido/metabolismo , Transfección , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
15.
J Invest Dermatol ; 111(1): 1-6, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9665379

RESUMEN

Vascular endothelial growth factor (VEGF) has been implicated in the pathologic angiogenesis observed in psoriasis and other chronic inflammatory skin diseases that are characterized by enhanced expression of VEGF by epidermal keratinocytes and of VEGF receptors by tortuous microvessels in the upper dermis. To investigate the functional importance of chronic VEGF overexpression in vivo, we used a keratin 14 promoter expression cassette containing the gene for murine VEGF164 to selectively target VEGF expression to basal epidermal keratinocytes in transgenic mice. These mice demonstrated an increased density of tortuous cutaneous blood capillaries with elevated expression levels of the high affinity VEGF receptors, VEGFR-1 and VEGFR-2, most prominently during the neonatal period. In contrast, no abnormalities of lymphatic vessels were detected. In addition, the number of mast cells in the upper dermis was significantly increased in transgenic skin. Intravital fluorescence microscopy revealed highly increased leukocyte rolling and adhesion in postcapillary skin venules that were both inhibited after injection of blocking antibodies against E- and P-selectin. Combined blocking antibodies against intercellular adhesion molecule-1 and lymphocyte function-associated antigen-1 were without effect, whereas an anti-vascular cell adhesion molecule-1/VLA-4 antibody combination almost completely normalized the enhanced leukocyte adhesion in transgenic mice. This study reveals VEGF as a growth factor specific for blood vessels, but not lymphatic vessels, and demonstrates that chronic orthotopic overexpression of VEGF in the epidermis is sufficient to induce cardinal features of chronic skin inflammation, providing a molecular link between angiogenesis, mast cell accumulation, and leukocyte recruitment to sites of inflammation.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Leucocitos/fisiología , Linfocinas/fisiología , Piel/irrigación sanguínea , Animales , Adhesión Celular , Movimiento Celular , Factores de Crecimiento Endotelial/genética , Linfocinas/genética , Mastocitos/fisiología , Ratones , Ratones Transgénicos , Microcirculación , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
16.
Mol Biol Cell ; 9(2): 469-81, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9450968

RESUMEN

Hypoxia is a prominent feature of malignant tumors that are characterized by angiogenesis and vascular hyperpermeability. Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) has been shown to be up-regulated in the vicinity of necrotic tumor areas, and hypoxia potently induces VPF/VEGF expression in several tumor cell lines in vitro. Here we report that hypoxia-induced VPF/VEGF expression is mediated by increased transcription and mRNA stability in human M21 melanoma cells. RNA-binding/electrophoretic mobility shift assays identified a single 125-bp AU-rich element in the 3' untranslated region that formed hypoxia-inducible RNA-protein complexes. Hypoxia-induced expression of chimeric luciferase reporter constructs containing this 125-bp AU-rich hypoxia stability region were significantly higher than constructs containing an adjacent 3' untranslated region element without RNA-binding activity. Using UV-cross-linking studies, we have identified a series of hypoxia-induced proteins of 90/88 kDa, 72 kDa, 60 kDa, 56 kDa, and 46 kDa that bound to the hypoxia stability region element. The 90/88-kDa and 60-kDa species were specifically competed by excess hypoxia stability region RNA. Thus, increased VPF/VEGF mRNA stability induced by hypoxia is mediated, at least in part, by specific interactions between a defined mRNA stability sequence in the 3' untranslated region and distinct mRNA-binding proteins in human tumor cells.


Asunto(s)
Hipoxia de la Célula/genética , Factores de Crecimiento Endotelial/genética , Linfocinas/genética , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Secuencia de Bases , Clonación Molecular , ADN Complementario/genética , Regulación Neoplásica de la Expresión Génica/genética , Semivida , Humanos , Melanoma , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN , ARN Mensajero/química , ARN Mensajero/genética , ARN Neoplásico/química , ARN Neoplásico/genética , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ADN , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
17.
Int J Exp Pathol ; 79(6): 347-57, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10319016

RESUMEN

The molecular mechanism underlying oxygen sensing in mammalian cells has been extensively investigated in the areas of glucose transport, glycolysis, erythropoiesis, angiogenesis and catecholamine metabolism. Expression of functionally operative representative proteins in these specific areas, such as the glucose transporter 1, glycolytic enzymes, erythropoietin, vascular endothelial growth factor and tyrosine hydroxylase are all induced by hypoxia. Recent studies demonstrated that both transcriptional activation and post-transcriptional mechanisms are important to the hypoxia-mediated regulation of gene expression. In this article, the cis-acting elements and trans-acting factors involved in the transcriptional activation of gene expression will be reviewed. In addition, the mechanisms of post-transcriptional mRNA stabilization will also be addressed. We will discuss whether these two processes of regulation of hypoxia-responsive genes are mechanistically linked and co-operative in nature.


Asunto(s)
Hipoxia de la Célula/genética , Regulación de la Expresión Génica/fisiología , Mamíferos/genética , Animales , Humanos , ARN Mensajero/genética , Factores de Transcripción/fisiología
18.
J Biol Chem ; 272(44): 27509-12, 1997 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-9346879

RESUMEN

Mutation or loss of function of the von Hippel-Lindau (VHL) tumor suppressor gene is regularly found in sporadic renal cell carcinomas (RCC), well vascularized malignant tumors that characteristically overexpress vascular permeability factor/vascular endothelial growth factor (VPF/VEGF). The wild-type VHL (wt-VHL) gene product acts to suppress VPF/VEGF expression, which is overexpressed when wt-VHL is inactive. The present study investigated the pathways by which VHL regulates VPF/VEGF expression. We found that inhibition of protein kinase C (PKC) represses VPF/VEGF expression in RCC cells that regularly overexpress VPF/VEGF. The wt-VHL expressed by stably transfected RCC cells forms cytoplasmic complexes with two specific PKC isoforms, zeta and delta, and prevents their translocation to the cell membrane where they otherwise would engage in signaling steps that lead to VPF/VEGF overexpression. Other experiments implicated mitogen-activated protein kinase (MAPK) phosphorylation as a downstream step in PKC regulation of VPF/VEGF expression. Taken together, these data demonstrate that wt-VHL, by neutralizing PKC isoforms zeta and delta and thereby inhibiting MAPK activation, plays an important role in preventing aberrant VPF/VEGF overexpression and the angiogenesis that results from such overexpression.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Factores de Crecimiento Endotelial/genética , Neoplasias Renales/metabolismo , Ligasas , Linfocinas/genética , Proteína Quinasa C/antagonistas & inhibidores , Proteínas/fisiología , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas , Transporte Biológico , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Inhibidores Enzimáticos/farmacología , Genes Supresores de Tumor , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Fosforilación , Transducción de Señal , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
19.
J Invest Dermatol ; 108(3): 263-8, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9036922

RESUMEN

Tissue hypoxia is a characteristic feature of malignant tumors and healing wounds, conditions that are associated with angiogenesis and with increased expression of vascular permeability factor (VPF; also called vascular endothelial growth factor, VEGF), a selective endothelial cell mitogen inducing microvascular hyperpermeability in vivo. We investigated the regulation of VPF/VEGF and its receptors by tissue hypoxia in normal human skin explants and in cultured skin cells in vitro. VPF/VEGF mRNA expression was dramatically upregulated in epidermal keratinocytes, dermal fibroblasts, and dermal microvessels after 24 h of skin organ culture. Hypoxia also enhanced the expression of VPF/VEGF in cultured epidermal keratinocytes and dermal microvascular endothelial cells (predominantly VPF/VEGF121 and VPF/VEGF165) and in dermal fibroblasts (additional upregulation of VPF/VEGF189). The expression of the VPF/VEGF receptor Flt-1 was selectively induced on dermal microvessels in skin explant cultures and in dermal endothelial cell monolayer cultures under hypoxic conditions. In contrast, the KDR receptor was downregulated by hypoxia. These results suggest that hypoxia likely regulates cutaneous angiogenesis and microvascular permeability by two distinct mechanisms: (i) Induction of VPF/VEGF in epithelial and mesenchymal cells, including endothelial cells. (ii) Differential modulation of VPF/VEGF receptor expression by microvascular endothelial cells. These mechanisms may be of importance in the pathogenesis of healing wounds and some malignant tumors that are commonly characterized by hypoxia and overexpression of VPF/VEGF.


Asunto(s)
Factores de Crecimiento Endotelial/biosíntesis , Hipoxia/fisiopatología , Linfocinas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores de Factores de Crecimiento/genética , Células Cultivadas , Endotelio Vascular/química , Endotelio Vascular/citología , Fibroblastos/química , Fibroblastos/citología , Expresión Génica , Humanos , Recién Nacido , Queratinocitos/química , Queratinocitos/citología , Masculino , Técnicas de Cultivo de Órganos , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular , Piel , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
Proc Natl Acad Sci U S A ; 94(25): 13612-7, 1997 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-9391074

RESUMEN

Vascular endothelial growth factor (VEGF), also known as vascular permeability factor, is a cytokine of central importance for the angiogenesis associated with cancers and other pathologies. Because angiogenesis often involves endothelial cell (EC) migration and proliferation within a collagen-rich extracellular matrix, we investigated the possibility that VEGF promotes neovascularization through regulation of collagen receptor expression. VEGF induced a 5- to 7-fold increase in dermal microvascular EC surface protein expression of two collagen receptors-the alpha1beta1 and alpha2beta1 integrins-through induction of mRNAs encoding the alpha1 and alpha2 subunits. In contrast, VEGF did not induce increased expression of the alpha3beta1 integrin, which also has been implicated in collagen binding. Integrin alpha1-blocking and alpha2-blocking antibodies (Ab) each partially inhibited attachment of microvascular EC to collagen I, and alpha1-blocking Ab also inhibited attachment to collagen IV and laminin-1. Induction of alpha1beta1 and alpha2beta1 expression by VEGF promoted cell spreading on collagen I gels which was abolished by a combination of alpha1-blocking and alpha2-blocking Abs. In vivo, a combination of alpha1-blocking and alpha2-blocking Abs markedly inhibited VEGF-driven angiogenesis; average cross-sectional area of individual new blood vessels was reduced 90% and average total new vascular area was reduced 82% without detectable effects on the pre-existing vasculature. These data indicate that induction of alpha1beta1 and alpha2beta1 expression by EC is an important mechanism by which VEGF promotes angiogenesis and that alpha1beta1 and alpha2beta1 antagonists may prove effective in inhibiting VEGF-driven angiogenesis in cancers and other important pathologies.


Asunto(s)
Factores de Crecimiento Endotelial/farmacología , Integrinas/fisiología , Linfocinas/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Animales , Anticuerpos/farmacología , Adhesión Celular , Células Cultivadas , Colágeno/metabolismo , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Humanos , Técnicas In Vitro , Integrina alfa1beta1 , Integrinas/antagonistas & inhibidores , Integrinas/genética , Linfocinas/fisiología , Ratones , Ratones Desnudos , ARN Mensajero/genética , Receptores de Colágeno , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA