Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
Biomater Sci ; 8(1): 39-63, 2019 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-31799977

RESUMEN

The term secretome, which traditionally strictly refers to single proteins, should be expanded to also include the great variety of nanoparticles secreted by cells (secNPs) into the extracellular space, which ranges from high-density lipoproteins of a few nanometers to extracellular vesicles and fat globules of hundreds of nanometers. Widening the definition is urged by the ever-increasing understanding of the role of secNPs as regulators/mediators of key physiological and pathological processes, which also puts them in the running as breakthrough cell-free therapeutics and diagnostics. "Made by cells for cells", secNPs are envisioned as a sweeping paradigm shift in nanomedicine, promising to overcome the limitations of synthetic nanoparticles by unsurpassed circulation and targeting abilities, precision and sustainability. From a longer/wider perspective, advanced manipulation would possibly make secNPs available as building blocks for future "biogenic" nanotechnology. However, the current knowledge is fragmented and sectorial (the majority of the studies being focused on a specific biological and/or medical aspect of a given secNP class or subclass), the understanding of the nanoscale and interfacial properties is limited and the development of bioprocesses and regulatory initiatives is in the early days. We believe that new multidisciplinary competencies and synergistic efforts need to be attracted and augmented to move forward. This review will contribute to the effort by attempting for the first time to rationally gather and elaborate secNPs and their traits into a unique concise framework - from biogenesis to colloidal properties, engineering and clinical translation - disclosing the overall view and easing comparative analysis and future exploitation.


Asunto(s)
Vesículas Extracelulares/metabolismo , Lipoproteínas/metabolismo , Nanopartículas/metabolismo , Animales , Vesículas Extracelulares/química , Humanos , Lipoproteínas/química , Conformación Molecular , Nanomedicina , Nanopartículas/química
2.
Oncogene ; 36(47): 6531-6541, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-28783175

RESUMEN

Vascular endothelial growth factor receptor-2 (VEGFR2) is the main pro-angiogenic receptor expressed by endothelial cells (ECs). Using surface plasmon resonance, immunoprecipitation, enzymatic digestion, immunofluorescence and cross-linking experiments with specific sugar-binding lectins, we demonstrated that VEGFR2 bears both α,1-fucose and α(2,6)-linked sialic acid (NeuAc). However, only the latter is required for VEGF binding to VEGFR2 and consequent VEGF-dependent VEGFR2 activation and motogenic response in ECs. Notably, downregulation of ß-galactoside α(2,6)-sialyltransferase expression by short hairpin RNA transduction inhibits VEGFR2 α(2,6) sialylation that is paralleled by an increase of ß-galactoside α(2,3)-sialyltransferase expression. This results in an ex-novo α(2,3)-NeuAc sialylation of the receptor that functionally replaces the lacking α(2,6)-NeuAc, thus allowing VEGF/VEGFR2 interaction. In keeping with the role of VEGFR2 sialylation in angiogenesis, the α(2,6)-NeuAc-binding lectin Sambucus nigra (SNA) prevents VEGF-dependent VEGFR2 autophosphorylation and EC motility, proliferation and motogenesis. In addition, SNA exerts a VEGF-antagonist activity in tridimensional angiogenesis models in vitro and in the chick-embryo chorioallantoic membrane neovascularization assay and mouse matrigel plug assay in vivo. In conclusion, VEGFR2-associated NeuAc plays an important role in modulating VEGF/VEGFR2 interaction, EC pro-angiogenic activation and neovessel formation. VEGFR2 sialylation may represent a target for the treatment of angiogenesis-dependent diseases.


Asunto(s)
Neovascularización Patológica/metabolismo , Lectinas de Plantas/farmacología , Procesamiento Proteico-Postraduccional , Proteínas Inactivadoras de Ribosomas/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Membrana Corioalantoides , Regulación hacia Abajo , Células Endoteliales/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Galactósidos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Ácido N-Acetilneuramínico/metabolismo , Neovascularización Fisiológica , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Resonancia por Plasmón de Superficie , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta-D-Galactósido alfa 2-6-Sialiltransferasa , beta-Galactosida alfa-2,3-Sialiltransferasa
3.
J Clin Pathol ; 67(11): 968-73, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25078331

RESUMEN

AIMS: Adrenocortical carcinoma (ACC) carries a poor prognosis and current systemic cytotoxic therapies result in only modest improvement in overall survival. In this retrospective study, we performed a comprehensive genomic profiling of 29 consecutive ACC samples to identify potential targets of therapy not currently searched for in routine clinical practice. METHODS: DNA from 29 ACC was sequenced to high, uniform coverage (Illumina HiSeq) and analysed for genomic alterations (GAs). RESULTS: At least one GA was found in 22 (76%) ACC (mean 2.6 alterations per ACC). The most frequent GAs were in TP53 (34%), NF1 (14%), CDKN2A (14%), MEN1 (14%), CTNNB1 (10%) and ATM (10%). APC, CCND2, CDK4, DAXX, DNMT3A, KDM5C, LRP1B, MSH2 and RB1 were each altered in two cases (7%) and EGFR, ERBB4, KRAS, MDM2, NRAS, PDGFRB, PIK3CA, PTEN and PTCH1 were each altered in a single case (3%). In 17 (59%) of ACC, at least one GA was associated with an available therapeutic or a mechanism-based clinical trial. CONCLUSIONS: Next-generation sequencing can discover targets of therapy for relapsed and metastatic ACC and shows promise to improve outcomes for this aggressive form of cancer.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/tratamiento farmacológico , Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/tratamiento farmacológico , Carcinoma Corticosuprarrenal/genética , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Terapia Molecular Dirigida , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Biopsia , Diseño de Fármacos , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Selección de Paciente , Medicina de Precisión , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Adulto Joven
5.
Curr Pharm Des ; 15(30): 3577-89, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19860702

RESUMEN

Angiogenesis and inflammation are closely integrated processes. Fibroblast growth factor-2 (FGF2) is a prototypic angiogenesis inducer belonging to the family of the heparin-binding FGF growth factors. FGF2 exerts its pro-angiogenic activity by interacting with various endothelial cell surface receptors, including tyrosine kinase receptors, heparan-sulfate proteoglycans, and integrins. A tight cross-talk exists between FGF2 and the inflammatory response in the modulation of blood vessel growth. Pentraxins act as soluble pattern recognition receptors with a wide range of functions in various pathophysiological conditions. The long-pentraxin PTX3 shares the C-terminal pentraxin-domain with short-pentraxins and possesses a unique N-terminal domain. These structural features indicate that PTX3 may have distinct biological/ligand recognition properties when compared to short-pentraxins. Co-expression of PTX3 and FGF2 has been observed in different inflammation/angiogenesis-dependent diseases. PTX3 binds FGF2 with high affinity and specificity. The interaction prevents the binding of FGF2 to its cognate tyrosine kinase receptors, leading to inhibition of the angiogenic activity of the growth factor. This suggests that PTX3 may exert a modulatory function by limiting the angiogenic activity of FGF2. An integrated approach that utilized PTX3 fragments, monoclonal antibodies, and surface plasmon resonance analysis has identified the FGF2-binding domain in the unique N-terminal extension of PTX3. On this basis, PTX3-derived synthetic peptides have been designed endowed with a significant antiangiogenic activity in vitro and in vivo. They may provide the basis for the development of novel antiangiogenic FGF2 antagonists.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Proteína C-Reactiva/farmacología , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Proteínas del Tejido Nervioso/farmacología , Péptidos/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Proteína C-Reactiva/química , Humanos , Modelos Moleculares , Conformación Molecular , Neovascularización Patológica/tratamiento farmacológico , Proteínas del Tejido Nervioso/química , Péptidos/química
6.
Curr Pharm Des ; 13(20): 2025-44, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17627537

RESUMEN

Angiogenesis, the process of new blood vessel formation from pre-existing ones, plays a key role in various physiological and pathological conditions, including embryonic development, wound repair, inflammation, and tumor growth. The 1980s saw for the first time the identification, purification, and sequencing of the two prototypic heparin-binding angiogenic fibroblast growth factors (FGF) 1 and 2. Since then, 22 structurally-related members of the FGF family and different classes of FGF receptors have been identified. Several experimental evidences point to a role for various FGFs in the neovascularization process that takes place in inflammation, angioproliferative diseases, and tumor growth. Thus, the FGF/FGF receptor system represents a target for the development of anti-angiogenic therapies. Purpose of this review is to summarize the different modalities that have been approached to impair the pro-angiogenic activity of the FGF/FGF receptor system and discuss their possible therapeutic implications.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Diseño de Fármacos , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Animales , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Neovascularización Fisiológica , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
7.
J Pathol ; 211(5): 541-549, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17310502

RESUMEN

The role of angiogenesis in tumour progression is a major subject in modern oncology and a correlation between angiogenesis and poor outcome has been demonstrated for human neuroblastomas. However, the role of angiogenesis in the maturation phase of neuroblastic tumours has never been considered. Human carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), a potent pro-angiogenic factor and mediator of vascular endothelial growth factor (VEGF)-induced angiogenesis, plays a crucial role during the activation phase of angiogenesis and it has been shown to be expressed in the microvessels of the developing central nervous system as well as in newly formed immature blood vessels in many different tumours and under physiological conditions. The present study has investigated the role of CEACAM1/VEGF-mediated angiogenesis across the whole spectrum of neuroblastic tumours, from undifferentiated to fully differentiated mature ganglioneuromas. CEACAM1 is peculiarly expressed in the microvessels of areas of active tumour maturation among differentiating neuroblastic/ganglion cells, whereas it is completely absent in the vessels of poorly differentiated/undifferentiated as well as in entirely mature Schwannian-rich areas. Interestingly, VEGF expression has been found in differentiating neuroblastic/ganglion cells adjacent to CEACAM1-positive microvessels. In keeping with these observations, VEGF expression was found in human neuroblastoma SH-SY5Y cells during differentiation after retinoic acid treatment. Moreover, conditioned medium from SH-SY5Y cells collected at different stages of differentiation induced progressive in vitro up-regulation of CEACAM1 expression in human umbilical vein endothelial cells (HUVECs) that was abrogated by the specific VEGF receptor-2/KDR inhibitor SU5416. Taken together, these data point to a role for CEACAM1/VEGF cross-talk during the maturation phase of neuroblastic tumours. This may mimic physiological events leading to maturation of the vasculature in the developing normal central nervous system. On the other hand, in poorly differentiated/undifferentiated lesions, VEGF-sustained angiogenesis does not reproduce physiological steps, but rather is associated with tumour aggressiveness and may involve other molecular pathways.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/fisiopatología , Antígenos CD/fisiología , Moléculas de Adhesión Celular/fisiología , Neoplasias del Mediastino/fisiopatología , Neovascularización Patológica/fisiopatología , Neuroblastoma/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Adolescente , Neoplasias de las Glándulas Suprarrenales/irrigación sanguínea , Glándulas Suprarrenales/irrigación sanguínea , Diferenciación Celular/fisiología , Células Cultivadas , Niño , Preescolar , Células Endoteliales/química , Femenino , Ganglios/química , Humanos , Inmunohistoquímica/métodos , Lactante , Masculino , Neoplasias del Mediastino/irrigación sanguínea , Mediastino/irrigación sanguínea , Microcirculación , Neuroblastoma/irrigación sanguínea , Regulación hacia Arriba/fisiología
8.
Leukemia ; 18(2): 331-6, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14671634

RESUMEN

The role of erythropoietin (Epo) in angiogenesis has not been completely clarified. Epo induces endothelial cell proliferation and migration and stimulates angiogenesis on rat aortic rings in vitro and in vivo in the chick embryo chorioallantoic membrane (CAM) assay. The aim of the present study was to evaluate the ultrastructural aspects of angiogenesis in the CAM vasculature after recombinant human Epo (rHuEpo) exposure. The results demonstrated that after rHuEpo stimulation, the generation of new blood vessels occurred more frequently following an intussusceptive microvascular growth (IMG) mechanism. We have performed our experiments between days 8 and 12 of incubation, that is, when in the normal condition the capillary network expands mainly by IMG, and because it is generally accepted that implants made from days 8 to 10 are strongly angiogenic. This response is peculiar of rHuEpo, because it is abolished when an Epo-blocking antibody was coadministered with Epo.


Asunto(s)
Eritropoyetina/farmacología , Intususcepción/inducido químicamente , Neovascularización Fisiológica/efectos de los fármacos , Animales , Embrión de Pollo , Embrión no Mamífero/irrigación sanguínea , Células Endoteliales/química , Células Endoteliales/ultraestructura , Humanos , Microcirculación/efectos de los fármacos , Microcirculación/embriología , Microcirculación/ultraestructura , Microscopía Electrónica , Receptores de Eritropoyetina/análisis , Proteínas Recombinantes , Factores de Tiempo
9.
Eur J Clin Invest ; 33(10): 891-6, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14511361

RESUMEN

Erythropoietin (Epo) is produced by the fetal liver and adult kidney and is an essential stimulator of erythropoiesis. It has, however, been shown to modulate host cellular signal transduction pathway to perform many other functions. New sites of Epo production have been found, such as the female reproductive organs and central nervous system. This review summarizes the involvement of Epo in the regulation of angiogenesis in both normal and pathological conditions.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Eritropoyetina/fisiología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Encéfalo/irrigación sanguínea , Femenino , Genitales Femeninos/irrigación sanguínea , Humanos , Neoplasias/irrigación sanguínea
10.
FEBS Lett ; 547(1-3): 183-8, 2003 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-12860410

RESUMEN

Cytosolic sialidase Neu2 has been implicated in myoblast differentiation. Here we observed a significant upregulation of Neu2 expression during differentiation of murine C2C12 myoblasts. This was evidenced both as an increase in Neu2 mRNA steady-state levels and in the cytosolic sialidase enzymatic activity. To understand the biological significance of Neu2 upregulation in myoblast differentiation, C2C12 cells were stably transfected with the rat cytosolic sialidase Neu2 cDNA. Neu2 overexpressing clones were characterized by a marked decrement of cell proliferation and by the capacity to undergo spontaneous myoblast differentiation also when maintained under standard growth conditions. This was evidenced by the formation of myogenin-positive myotubes and by a significant decrease in the nuclear levels of cyclin D1 protein. No differentiation was on the contrary observed in parental and mock-transfected cells under the same experimental conditions. The results indicate that Neu2 upregulation per se is sufficient to trigger myoblast differentiation in C2C12 cells.


Asunto(s)
Diferenciación Celular/fisiología , Mioblastos/citología , Neuraminidasa/genética , Animales , Secuencia de Bases , Línea Celular , Citosol/enzimología , Cartilla de ADN , Cinética , Ratones , Neuraminidasa/metabolismo , Ratas , Proteínas Recombinantes/metabolismo , Transfección
11.
Curr Pharm Des ; 9(7): 553-66, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12570803

RESUMEN

Angiogenesis is the process of generating new capillary blood vessels. Uncontrolled endothelial cell proliferation is observed in tumor neovascularization and in angioproliferative diseases. Tumors cannot growth as a mass above few mm(3) unless a new blood supply is induced. It derives that the control of the neovascularization process may affect tumor growth and may represent a novel approach to tumor therapy. Angiogenesis is controlled by a balance between proangiogenic and antiangiogenic factors. The angiogenic switch represents the net result of the activity of angiogenic stimulators and inhibitors, suggesting that counteracting even a single major angiogenic factor could shift the balance towards inhibition. Heparan sulfate proteoglycans are involved in the modulation of the neovascularization that takes place in different physiological and pathological conditions. This modulation occurs through the interaction with angiogenic growth factors or with negative regulators of angiogenesis. Thus, the study of the biochemical bases of this interaction may help to design glycosaminoglycan analogs endowed with angiostatic properties. The purpose of this review is to provide an overview of the structure/function of heparan sulfate proteoglycans in endothelial cells and to summarize the angiostatic properties of synthetic heparin-like compounds, chemically modified heparins, and biotechnological heparins.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Heparina/análogos & derivados , Heparina/farmacología , Neovascularización Patológica/tratamiento farmacológico , Inductores de la Angiogénesis/antagonistas & inhibidores , Animales , Adhesión Celular/fisiología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiología , Proteoglicanos de Heparán Sulfato/metabolismo , Proteoglicanos de Heparán Sulfato/fisiología , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/fisiopatología
12.
J Cell Mol Med ; 6(3): 439-46, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12417062

RESUMEN

The CAM is an extraembryonic membrane which serves as a gas exchange surface and its respiratory function is provided by an extensive capillary network. The development of the vascular system of the CAM is a complex, highly regulated process that depends on genetic and epigenetic factors expressed by endothelial and non-endothelial cells. In spite of the evidence that several growth factors are angiogenic in the CAM assay, poorly investigated is their role in the development of the CAM's vascular system. This article reviews our studies concerning the role of exogenous and endogenous fibroblast growth factor-2 (FGF-2) in the CAM vascularization. The findings in all these studies support the importance of FGF-2 as an autocrine paracrine stimulator of angiogenesis and its key role in the development of the vascular system in the avian embryo.


Asunto(s)
Alantoides/irrigación sanguínea , Corion/irrigación sanguínea , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neovascularización Fisiológica , Alantoides/citología , Alantoides/fisiología , Animales , Comunicación Autocrina , Embrión de Pollo , Corion/citología , Corion/fisiología , Endotelio Vascular/citología , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Humanos , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
13.
Leukemia ; 16(8): 1490-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12145690

RESUMEN

The antimetabolite 6-thioguanine (6-TG) is utilized in the management of acute myelogenous leukemia (AML). Angiogenesis is a possible therapeutic target in hematologic tumors. Thus, we addressed the possibility that 6-TG may also act as an anti-angiogenic molecule. 6-TG inhibited endothelial cell proliferation triggered by fibroblast growth factor-2 (FGF2) and vascular endothelial growth factor (VEGF) and delayed the repair of a mechanically wounded endothelial cell monolayer. Also, 6-TG inhibited sprouting within fibrin gel, morphogenesis on Matrigel, and collagen gel invasion by endothelial cells. 2-Aminopurine was ineffective. In vivo, 6-TG inhibited basal, VEGF-induced, and FGF2-induced vascularization in the chick embryo chorioallantoic membrane and prevented neovascularization triggered by leukemia LIK cells or their conditioned medium. Finally, bone marrow vascularization in AML patients was decreased to control values in the early remission phase and persisted unvaried after 8-12 months of maintenance therapy with 6-TG. Thus, 6-TG inhibits different steps of the angiogenesis process in vitro and exerts a potent anti-angiogenic activity in vivo. Its anti-angiogenic activity, together with its antimetabolite activity towards tumor cells, may contribute to its action during maintenance therapy in AML. These results suggest a new rationale for the use of purine analogs in the management of AML.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Endotelio Vascular/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Tioguanina/farmacología , 2-Aminopurina/farmacología , Enfermedad Aguda , Anciano , Alantoides/irrigación sanguínea , Alantoides/efectos de los fármacos , Anemia/tratamiento farmacológico , Anemia/patología , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Médula Ósea/irrigación sanguínea , Bovinos , Línea Celular Transformada , Embrión de Pollo , Corion/irrigación sanguínea , Corion/efectos de los fármacos , Citarabina/administración & dosificación , Daunorrubicina/administración & dosificación , Evaluación de Medicamentos , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/citología , Etopósido/administración & dosificación , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Estudios de Seguimiento , Humanos , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/patología , Linfocinas/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Inducción de Remisión , Estrés Mecánico , Tioguanina/administración & dosificación , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
15.
Exp Hematol ; 29(8): 993-1003, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11495705

RESUMEN

OBJECTIVE: The mechanisms used by human lymphoproliferative diseases to invade locally and metastasize are thought to be similar to those developed by solid tumors, including cell proliferation and secretion of extracellular matrix-degrading enzymes following adhesion to extracellular matrix proteins. Hence, the ability of Namalwa (Burkitt's lymphoma), U266 (multiple myeloma), and CEM (T-cell lymphoblastic leukemia) cells to interact with the extracellular matrix components vitronectin and fibronectin was determined. Fresh bone marrow plasma cells from patients with multiple myeloma also were studied. MATERIALS AND METHODS: Engagement of alpha(v)beta(3) integrin, formation and protein composition of focal adhesion contacts on the cell surface, phosphorylation of several signal transduction proteins in the contacts, cell proliferation, and enzyme secretion were studied following adhesion to vitronectin and fibronectin. RESULTS: All three lines adhered to immobilized vitronectin and fibronectin. Adhesion was fully prevented by neutralizing monoclonal anti-alpha(v)beta(3) integrin antibody. Integrin engagement caused the formation of phosphorylated pp60(src)/focal adhesion kinase complexes and the aggregation of focal adhesion plaques containing the beta(3) integrin subunit, the cytoskeletal proteins vinculin, cortactin, and paxillin, the tyrosine kinases focal adhesion kinase and pp60(src), the adapter protein Grb-2, and the mitogen-activated protein kinase ERK-2. Free and immobilized vitronectin and fibronectin stimulated the proliferation of cells under serum-free conditions and the production and release of urokinase-type plasminogen activator, and increased the release of the activated forms of matrix metalloproteinase-2 and matrix metalloproteinase-9 in an alpha(v)beta(3) integrin-dependent manner. Similar results were obtained in myeloma plasma cells. CONCLUSIONS: The demonstrated ability of lymphoid tumor cells to interact with the extracellular matrix components vitronectin and fibronectin via alpha(v)beta(3) integrin can be interpreted as evidence of a novel mechanism for their invasion and spreading. This interaction allows them to adhere to the substratum and enhances their proliferation and protease secretion.


Asunto(s)
Células de la Médula Ósea/patología , Adhesión Celular/fisiología , División Celular/fisiología , Endopeptidasas/metabolismo , Mieloma Múltiple/patología , Células Plasmáticas/patología , Receptores de Vitronectina/fisiología , Anticuerpos Monoclonales/farmacología , Activación Enzimática , Fibronectinas , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Humanos , Cinética , Linfoma , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Receptores de Vitronectina/inmunología , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Vitronectina
16.
J Biol Chem ; 276(41): 37900-8, 2001 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-11473122

RESUMEN

The angiogenic basic fibroblast growth factor (FGF2) interacts with tyrosine kinase receptors (FGFRs) and heparan sulfate proteoglycans (HSPGs) in endothelial cells. Here, we report the FGF2 antagonist and antiangiogenic activity of novel sulfated derivatives of the Escherichia coli K5 polysaccharide. K5 polysaccharide was chemically sulfated in N- and/or O-position after N-deacetylation. O-Sulfated and N,O-sulfated K5 derivatives with a low degree and a high degree of sulfation compete with heparin for binding to 125I-FGF2 with different potency. Accordingly, they abrogate the formation of the HSPG.FGF2.FGFR ternary complex, as evidenced by their capacity to prevent FGF2-mediated cell-cell attachment of FGFR1-overexpressing HSPG-deficient Chinese hamster ovary (CHO) cells to wild-type CHO cells. They also inhibited 125I-FGF2 binding to FGFR1-overexpressing HSPG-bearing CHO cells and adult bovine aortic endothelial cells. K5 derivatives also inhibited FGF2-mediated cell proliferation in endothelial GM 7373 cells and in human umbilical vein endothelial (HUVE) cells. In all these assays, the N-sulfated K5 derivative and unmodified K5 were poorly effective. Also, highly O-sulfated and N,O-sulfated K5 derivatives prevented the sprouting of FGF2-transfected endothelial FGF2-T-MAE cells in fibrin gel and spontaneous angiogenesis in vitro on Matrigel of FGF2-T-MAE and HUVE cells. Finally, the highly N,O-sulfated K5 derivative exerted a potent antiangiogenic activity on the chick embryo chorioallantoic membrane. These data demonstrate the possibility of generating FGF2 antagonists endowed with antiangiogenic activity by specific chemical sulfation of bacterial K5 polysaccharide. In particular, the highly N,O-sulfated K5 derivative may provide the basis for the design of novel angiostatic compounds.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Endotelio Vascular/efectos de los fármacos , Escherichia coli/metabolismo , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Polisacáridos/farmacología , Animales , Células CHO , Secuencia de Carbohidratos , Bovinos , Células Cultivadas , Embrión de Pollo , Cricetinae , Endotelio Vascular/citología , Humanos , Polisacáridos/química , Proteínas Recombinantes/antagonistas & inhibidores
17.
J Vasc Res ; 38(4): 389-97, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11455210

RESUMEN

Fibroblast growth factor-2 (FGF2) and vascular endothelial growth factor (VEGF) exert their angiogenic activity by interacting with endothelial cells in a distinct manner. In this study, we investigated the morphological features of endothelial cells of the chick embryo chorioallantoic membrane (CAM) microvasculature after stimulation with FGF2 or VEGF. In order to provide a continuous delivery of the growth factor, we utilized a recently developed gelatin sponge/CAM assay in which a limited number of FGF2- or VEGF-transfected cells were adsorbed onto gelatin sponges and applied on the top of the CAM on day 8 of development. Their angiogenic activity was compared to that exerted by a single bolus of the corresponding growth factor. All the angiogenic stimuli induced a comparable vasoproliferative response, as demonstrated by the appearance of similar numbers of immature blood vessels within the sponge on day 12. No angiogenic response was observed in CAMs implanted with the corresponding parental cell lines or vehicle. Electron microscopy demonstrated that VEGF-overexpressing cells modified the phenotype of the endothelium of the blood vessels at the boundary between the implant and the surrounding CAM mesenchyme. The endothelial lining of 30% of these vessels showed segmental attenuations, was frequently interrupted and became fenestrated, mimicking what is observed in tumor vasculature. In contrast, the vessels consisted of continuous endothelium sealed by tight junctions in all the other experimental conditions. These results indicate that FGF2 and VEGF interact with endothelial cells of the CAM in a distinct manner. Both growth factors induce a potent angiogenic response, but only VEGF delivered in a continuous manner by its transfectants can modify the phenotype of the otherwise quiescent endothelium of CAM blood microvessels. The gelatin sponge/CAM assay may constitute a new model to study the mechanisms leading to endothelial fenestration in tumor growth.


Asunto(s)
Alantoides/irrigación sanguínea , Corion/irrigación sanguínea , Factores de Crecimiento Endotelial/administración & dosificación , Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Linfocinas/administración & dosificación , Neovascularización Fisiológica , Adsorción , Animales , Aorta , Capilares/fisiología , Capilares/ultraestructura , Línea Celular , Trasplante de Células/métodos , Embrión de Pollo , Factores de Crecimiento Endotelial/genética , Endotelio Vascular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Gelatina , Expresión Génica , Linfocinas/genética , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/administración & dosificación , Factores de Tiempo , Transfección , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
18.
Oncogene ; 20(21): 2655-63, 2001 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-11420677

RESUMEN

Recombinant Fibroblast Growth Factor-4 (FGF4) and FGF2 induce extracellular signal-regulated kinase-1/2 activation and DNA synthesis in murine aortic endothelial (MAE) cells. These cells co-express the IIIc/Ig-3 loops and the novel glycosaminoglycan-modified IIIc/Ig-2 loops isoforms of FGF receptor-2 (FGFR2). The affinity of FGF4/FGFR2 interaction is 20-30 times lower than that of FGF2 and is enhanced by heparin. Overexpression of FGF2 or FGF4 cDNA in MAE cells results in a transformed phenotype and increased proliferative capacity, more evident for FGF2 than FGF4 transfectants. Both transfectants induce angiogenesis when applied on the top of the chick embryo chorioallantoic membrane. However, in contrast with FGF2-transfected cells, FGF4 transfectants show a limited capacity to growth under anchorage-independent conditions and lack the ability to invade 3D fibrin gel and to undergo morphogenesis in vitro. Also, they fail to induce hemangiomas when injected into the allantoic sac of the chick embryo. In conclusion, although exogenous FGF2 and FGF4 exert a similar response in MAE cells, significant differences are observed in the biological behavior of FGF4 versus FGF2 transfectants, indicating that the expression of the various members of the FGF family can differently affect the behavior of endothelial cells and, possibly, of other cell types, including tumor cells.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos/fisiología , Proteínas Proto-Oncogénicas/farmacología , Proteínas Proto-Oncogénicas/fisiología , Células 3T3/fisiología , Alantoides/irrigación sanguínea , Animales , Línea Celular , Embrión de Pollo , Corion/irrigación sanguínea , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor 4 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/biosíntesis , Factores de Crecimiento de Fibroblastos/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Neovascularización Fisiológica/fisiología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Recombinantes/farmacología , Transfección
19.
Cancer Res ; 61(13): 5057-64, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11431341

RESUMEN

Cidofovir [(S)-HPMPC; (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine] is an antiviral drug that has been approved for the treatment of cytomegalovirus retinitis in AIDS patients. Cidofovir also possesses potent inhibitory activity against various human papillomavirus-induced tumors in animal models and patients. In addition, cidofovir inhibits the development of murine polyomavirus-induced hemangiomas in rats by an as-yet-uncharacterized, antivirus-independent mechanism. Here we report the inhibitory effect of cidofovir on the development of virus-independent vascular tumors originated by basic fibroblast growth factor (FGF2)-overexpressing endothelial cells (FGF2-T-MAE cells). In vitro, cidofovir was cytostatic to FGF2-T-MAE cells at a 50% cytostatic concentration of 6.7 microg/ml. Cidofovir concentrations >25 microg/ml resulted in cytotoxicity because of induction of apoptosis. Cidofovir did not affect FGF2-T-MAE cell sprouting in three-dimensional fibrin gel and morphogenesis on Matrigel at noncytotoxic concentrations. In vivo, cidofovir (100 microg/egg) completely suppressed hemangioma formation on the chick chorioallantoic membrane (CAM) induced by intra-allantoic injection of FGF2-T-MAE cells, without affecting the formation of normal CAM vessels. Accordingly, cidofovir applied locally at 200 microg/disc, reduced neovascularization on the CAM by only 35%. Intratumoral or systemic administration of cidofovir caused a significant inhibition of the growth of s.c., i.p., or intracerebral FGF2-T-MAE xenografts in nude mice and severe combined immunodeficient mice. Drug-induced apoptosis was observed in FGF2-T-MAE tumors as soon as 2 days after the beginning of treatment. In conclusion, cidofovir appears to inhibit the growth of endothelium-derived tumors via induction of apoptosis without exerting a direct antiangiogenic activity.


Asunto(s)
Antineoplásicos/farmacología , Citosina/farmacología , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Hemangioendotelioma/tratamiento farmacológico , Organofosfonatos , Compuestos Organofosforados/farmacología , Animales , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/tratamiento farmacológico , División Celular/efectos de los fármacos , Embrión de Pollo , Cidofovir , Citosina/análogos & derivados , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/fisiología , Hemangioendotelioma/irrigación sanguínea , Hemangioendotelioma/patología , Ratones , Ratones Desnudos , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Transfección , Células Tumorales Cultivadas
20.
Endothelium ; 8(1): 65-74, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11409852

RESUMEN

Extracellular Tat protein, the transactivating factor of the human immunodeficiency virus type 1 (HIV-1), modulates gene expression, growth, and angiogenic activity in endothelial cells by interacting with the vascular endothelial growth factor (VEGF) receptor-2 (Flk-1/KDR). Recombinant Tat protein, produced as glutathione-S-transferase chimera (GST-Tat), activates mitogen-activated protein kinase (MAPK) ERK(1/2) in human, murine, and bovine endothelial cells whereas GST is ineffective. In bovine aortic endothelial cells, GST-Tat and the 165 amino acid VEGF isoform (VEGF165) induce transient ERK(1/2) phosphorylation with similar potency and kinetics. The synthetic peptide Tat(41-60), but not peptides Tat(1-21) and Tat(71-86), causes ERK(1/2) phosphorylation, thus implicating Tat/KDR interaction in the activation of this signalling pathway. Accordingly, GST-Tat induces ERK(1/2) phosphorylation in KDR-transfected porcine aortic endothelial cells but not in parental cells. MAPK kinase inhibitors PD098059 and U0126 prevent ERK(1/2) phosphorylation by Tat. However, they do not affect the angiogenic activity exerted by Tat in the murine Matrigel plug and chick embryo chorioallantoic membrane assays. Blocking of MAPK kinase activity impairs instead the angiogenic response to VEGF165 and to fibroblast growth factor-2 (FGF-2). Our data demonstrate that ERK(1/2) activation following the interaction of HIV-1 Tat protein with endothelial cell Flk-1/KDR receptor does not represent an absolute requirement for a full angiogenic response to this growth factor that appears to utilize mechanism(s) at least in part distinct from those triggered by other prototypic angiogenic growth factors.


Asunto(s)
Endotelio Vascular/fisiología , Productos del Gen tat/metabolismo , VIH-1/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Animales , Butadienos/farmacología , Bovinos , Células Cultivadas , Colágeno , Combinación de Medicamentos , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Productos del Gen tat/farmacología , Humanos , Cinética , Laminina , Ratones , Proteína Quinasa 3 Activada por Mitógenos , Nitrilos/farmacología , Fragmentos de Péptidos/farmacología , Fosforilación , Proteoglicanos , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...