Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(18): 7312-7, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589885

RESUMEN

Hypoxia, or low oxygen tension, is a major regulator of tumor development and aggressiveness. However, how cancer cells adapt to hypoxia and communicate with their surrounding microenvironment during tumor development remain important questions. Here, we show that secreted vesicles with exosome characteristics mediate hypoxia-dependent intercellular signaling of the highly malignant brain tumor glioblastoma multiforme (GBM). In vitro hypoxia experiments with glioma cells and studies with patient materials reveal the enrichment in exosomes of hypoxia-regulated mRNAs and proteins (e.g., matrix metalloproteinases, IL-8, PDGFs, caveolin 1, and lysyl oxidase), several of which were associated with poor glioma patient prognosis. We show that exosomes derived from GBM cells grown at hypoxic compared with normoxic conditions are potent inducers of angiogenesis ex vivo and in vitro through phenotypic modulation of endothelial cells. Interestingly, endothelial cells were programmed by GBM cell-derived hypoxic exosomes to secrete several potent growth factors and cytokines and to stimulate pericyte PI3K/AKT signaling activation and migration. Moreover, exosomes derived from hypoxic compared with normoxic conditions showed increased autocrine, promigratory activation of GBM cells. These findings were correlated with significantly enhanced induction by hypoxic compared with normoxic exosomes of tumor vascularization, pericyte vessel coverage, GBM cell proliferation, as well as decreased tumor hypoxia in a mouse xenograft model. We conclude that the proteome and mRNA profiles of exosome vesicles closely reflect the oxygenation status of donor glioma cells and patient tumors, and that the exosomal pathway constitutes a potentially targetable driver of hypoxia-dependent intercellular signaling during tumor development.


Asunto(s)
Vasos Sanguíneos/patología , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Exosomas/metabolismo , Glioma/irrigación sanguínea , Glioma/patología , Animales , Comunicación Autocrina , Neoplasias Encefálicas/genética , Hipoxia de la Célula/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Humanos , Ratones , Ratones SCID , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Comunicación Paracrina , Pericitos/metabolismo , Pericitos/patología , Proteoma/metabolismo , Transducción de Señal/genética , Donantes de Tejidos , Transcriptoma/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Methods Mol Biol ; 720: 327-38, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21318883

RESUMEN

The polyamines are polycationic compounds essential for cellular proliferation and transformation. In addition to a well-defined biosynthesis pathway, polyamines are internalized into cells by as yet incompletely defined mechanisms. Numerous reports have shown that efficient polyamine uptake depends on the presence of polyanionic, cell surface-associated heparan sulfate proteoglycans (HSPGs). In this chapter, we provide protocols for studying HSPG-mediated uptake of polyamines in various cell lines, and provide instructions for the use of two different genetic models of HSPG deficiency. We describe the enzymatic reduction of cell surface HSPG through Heparinase III lyase treatment as well as the use of phage display-derived single chain variable fragment (scFv) anti-HS antibodies to block HSPGs at the cell surface. Finally, we provide a protocol for the quantitative verification of loss or reduction of cell surface HSPGs and a detailed description of polyamine uptake measurement.


Asunto(s)
Bioquímica/métodos , Proteoglicanos de Heparán Sulfato/metabolismo , Poliaminas/metabolismo , Adenoviridae/genética , Animales , Anticuerpos/farmacología , Células CHO , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Citometría de Flujo , Vectores Genéticos/genética , Células HeLa , Proteoglicanos de Heparán Sulfato/biosíntesis , Humanos , Integrasas/metabolismo , Ratones , Polisacárido Liasas/metabolismo , Reproducibilidad de los Resultados , Coloración y Etiquetado , Transducción Genética
3.
Exp Cell Res ; 316(16): 2683-91, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20594968

RESUMEN

The polyamines are essential for cancer cell proliferation during tumorigenesis. Targeted inhibition of ornithine decarboxylase (ODC), i.e. a key enzyme of polyamine biosynthesis, by alpha-difluoromethylornithine (DFMO) has shown anti-neoplastic activity in various experimental models. This activity has mainly been attributed to the anti-proliferative effect of DFMO in cancer cells. Here, we provide evidence that unperturbed ODC activity is a requirement for proper microvessel sprouting ex vivo as well as the migration of primary human endothelial cells. DFMO-mediated ODC inhibition was reversed by extracellular polyamine supplementation, showing that anti-angiogenic effects of DFMO were specifically related to polyamine levels. ODC inhibition was associated with an abnormal morphology of the actin cytoskeleton during cell spreading and migration. Moreover, our data suggest that de-regulated actin cytoskeleton dynamics in DFMO treated endothelial cells may be related to constitutive activation of the small GTPase CDC42, i.e. a well-known regulator of cell motility and actin cytoskeleton remodeling. These insights into the potential role of polyamines in angiogenesis should stimulate further studies testing the combined anti-tumor effect of polyamine inhibition and established anti-angiogenic therapies in vivo.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Endotelio Vascular/efectos de los fármacos , Neovascularización Fisiológica , Inhibidores de la Ornitina Descarboxilasa , Poliaminas/farmacología , Animales , Western Blotting , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Eflornitina/farmacología , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos C57BL , Microcirculación , Ornitina Descarboxilasa/metabolismo , Venas Umbilicales , Cicatrización de Heridas , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo
4.
In Vitro Cell Dev Biol Anim ; 46(7): 577-84, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20383663

RESUMEN

Angiogenesis is a hallmark of expanding tissue e.g. during embryogenesis and wound healing in physiology as well as in diseases such as cancer and atherosclerosis. Key steps of the angiogenic process involve growth factor-mediated stimulation of endothelial cell sprouting and tube formation. Heparan sulphate proteoglycans (HSPGs) have been implicated as important co-receptors of several pro-angiogenic proteins. The importance of HSPGs in physiology was underscored by the finding that knockout of the gene encoding HS polymerase, EXT-1, resulted in early embryonic lethality. Here, we describe the establishment of HS-deficient endothelial cells from sprouting aortas as well as from the lungs of EXT-1(flox/flox) mice. Recombination of the loxP-flanked EXT-1 locus by Cre-expressing adenovirus was demonstrated at the mRNA level. Moreover, depletion of HS polysaccharides was verified by flow cytometry and fluorescence microscopy methodology using phage display-derived anti-HS antibodies. In summary, we provide a genetic model to unravel the functional role of HSPGs specifically in primary endothelial cells during early steps of angiogenesis. Our studies are applicable to most loxP-based transgenic mouse strains, and may thus be of general importance in the angiogenesis field.


Asunto(s)
Aorta/crecimiento & desarrollo , Técnicas de Cultivo de Célula/métodos , Células Endoteliales/metabolismo , Heparitina Sulfato/deficiencia , Pulmón/irrigación sanguínea , N-Acetilglucosaminiltransferasas/genética , Neovascularización Fisiológica , Adenoviridae/genética , Animales , Células Cultivadas , Células Endoteliales/enzimología , Vectores Genéticos/genética , Heparitina Sulfato/metabolismo , Integrasas/metabolismo , Pulmón/enzimología , Ratones , Ratones Transgénicos , N-Acetilglucosaminiltransferasas/metabolismo , Recombinación Genética/genética
5.
J Biol Chem ; 284(47): 32959-67, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19783663

RESUMEN

Cellular uptake of several viruses and polybasic macromolecules requires the expression of cell-surface heparan sulfate proteoglycan (HSPG) through as yet ill defined mechanisms. We unexpectedly found that among several cell-surface-binding single chain variable fragment (scFv) anti-HS antibody (alphaHS) clones, only one, AO4B08, efficiently translocated macromolecular cargo to intracellular vesicles through induction of HSPG endocytosis. Interestingly, AO4B08-induced PG internalization was strictly dependent on HS 2-O-sulfation and appeared independent of intact N-sulfation. AO4B08 and human immunodeficiency virus (HIV)-Tat, i.e. a well known cell-penetrating peptide, were shown to compete for the internalizing PG population. To obtain a more detailed characterization of this pathway, we have developed a procedure for the isolation of endocytic vesicles by conjugating AO4B08 with superparamagnetic nanoparticles. [(35)S]sulfate-labeled HSPG was found to accumulate in isolated, AO4B08-containing vesicles, providing the first biochemical evidence for intact HSPG co-internalization with its ligand. Further analysis revealed the existence of both syndecan, i.e. a transmembrane HSPG, and glycosyl-phosphatidyl-inositol-anchored glypican in purified vesicles. Importantly, internalized syndecan and glypican were found to co-localize in AO4B08-containing vesicles. Our data establish HSPGs as true internalizing receptors of macromolecular cargo and indicate that the sorting of cell-surface HSPG to endocytic vesicles is determined by a specific HS epitope that can be carried by both syndecan and glypican core protein.


Asunto(s)
Membrana Celular/metabolismo , Epítopos/química , Glipicanos/química , Proteoglicanos de Heparán Sulfato/metabolismo , Fragmentos de Inmunoglobulinas/metabolismo , Sindecanos/química , Animales , Células CHO , Cricetinae , Cricetulus , Endocitosis , Antígenos VIH/química , Células HeLa , Humanos , Nanopartículas/química , Biblioteca de Péptidos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
6.
Biochem Biophys Res Commun ; 380(2): 413-8, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19250631

RESUMEN

Hypoxia-dependent angiogenesis is an inherent feature of solid tumors, and a better understanding of the molecular mechanisms of hypoxic cell-death should provide additional targets for cancer therapy. Here, we show a novel role of the polyamines in endothelial cell (EC) survival during hypoxia. Polyamine depletion by specific inhibition of ornithine decarboxylase was shown to protect ECs from hypoxia-induced apoptosis. Inhibition of the polyamines resulted in a significant induction of PI3K/AKT and its down-stream target MCL-1, i.e. an anti-apoptotic member of the BCL-2 family. Specific inhibitors of PI3K reversed the decrease of hypoxia-induced apoptosis as well as the induction of MCL-1 in polyamine-deprived cells. Moreover, siRNA-mediated down-regulation of MCL-1 was found to counter-act the protective effect of polyamine inhibition. We conclude that the polyamines regulate hypoxia-induced apoptosis in ECs through PI3K/AKT and MCL-1 dependent pathways. Our results may have important implications for the modulation of hypoxia-driven neovascularization.


Asunto(s)
Poliaminas Biogénicas/metabolismo , Células Endoteliales/fisiología , Neovascularización Patológica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Apoptosis , Poliaminas Biogénicas/antagonistas & inhibidores , Hipoxia de la Célula , Supervivencia Celular , Regulación hacia Abajo , Células Endoteliales/metabolismo , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética
7.
Cancer Res ; 68(22): 9291-301, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19010902

RESUMEN

Hypoxia is a hallmark of solid tumors, which may offer opportunities for targeted therapies of cancer; however, the mechanisms that link hypoxia to malignant transformation and tumor progression are not fully understood. Here, we show that up-regulation of the polyamine system promotes cancer cell survival during hypoxic stress. Hypoxia was found to induce polyamine transport and the key enzyme of polyamine biosynthesis, ornithine decarboxylase (ODC), in a variety of cancer cell lines. Increased ODC protein expression was shown in hypoxic, GLUT-1-expressing regions of tumor spheroids and experimental tumors, as well as in clinical tumor specimens. Hypoxic induction of the polyamine system was dependent on antizyme inhibitor (i.e., a key positive regulator of ODC and polyamine transport), as shown by RNA interference experiments. Interestingly, depletion of the polyamines during hypoxia resulted in increased apoptosis, which indicates an essential role of the polyamines in cancer cell adaptation to hypoxic stress. These results were supported by experiments in an in vivo glioma tumor model, showing significantly enhanced antitumor effects of the antiangiogenic, humanized anti-vascular endothelial growth factor (VEGF) antibody bevacizumab when used in combination with the well-established, irreversible inhibitor of ODC, alpha-difluoromethylornithine. Our results provide important insights into the hypoxic stress response in malignant cells and implicate combined targeting of VEGF and ODC as an alternative strategy to treat cancer disease.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Poliaminas Biogénicas/fisiología , Hipoxia de la Célula , Eflornitina/administración & dosificación , Neoplasias/tratamiento farmacológico , Inhibidores de la Ornitina Descarboxilasa , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados , Bevacizumab , Proteínas Portadoras/fisiología , Células Cultivadas , Quimioterapia Combinada , Eflornitina/farmacología , Femenino , Transportador de Glucosa de Tipo 1/análisis , Humanos , Ratones , Ratones SCID , Ornitina Descarboxilasa/metabolismo , Espermina/farmacocinética
8.
Int J Oncol ; 32(4): 749-56, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18360702

RESUMEN

The growth-promoting polyamines are polybasic compounds that efficiently enter cancer cells by as yet incompletely defined mechanisms. Strategies to inhibit their internalization may have important implications in the management of tumor disease. Here, we show that cellular binding and uptake of polyamines are inhibited by a single chain variable fragment anti-heparan sulfate (HS) antibody. Polyamine uptake was inhibited in a dose-dependent manner, and was associated with compensatory up-regulation of ornithine decarboxylase (ODC), i.e. the key enzyme of the polyamine biosynthesis pathway. Conversely, depletion of intracellular polyamines by the specific ODC-inhibitor alpha-difluoromethylornithine (DFMO) resulted in increased cellular binding of polyamine and anti-HS antibody. Importantly, anti-HS antibody also efficiently targeted DFMO-induced polyamine uptake, and combined polyamine biosynthesis inhibition by DFMO, and uptake inhibition by anti-HS antibody attenuated tumor cell proliferation in vitro. In conclusion, cell-surface HS proteoglycan is a relevant target for antibody-mediated inhibition of the uptake of polyamines, and polyamine-dependent cell proliferation.


Asunto(s)
Poliaminas Biogénicas/antagonistas & inhibidores , Heparitina Sulfato/inmunología , Fragmentos de Inmunoglobulinas/farmacología , Animales , Poliaminas Biogénicas/fisiología , Transporte Biológico , Células CHO , Proliferación Celular , Cricetinae , Cricetulus , Eflornitina/farmacología , Células HeLa , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...