Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Neoplasma ; 66(6): 963-970, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31607128

RESUMEN

Triple negative breast cancer (TNBC) is a particularly aggressive subtype of breast cancer (BC) for which limited therapeutic options are available. Recently, ß-blockers (BBs) have been suggested to have favorable effects in the treatment of BC. The aim of this systematic review was to collect evidence from preclinical and clinical studies concerning the scientific evidence for the repurposing of BBs in TNBC treatment. PubMed database was searched to retrieve studies of interest published up to 30/01/2018. All preclinical studies using TNBC in vitro and in vivo models and assessing the effect of any molecule with sympatholytic or sympathomimetic activity on adrenergic receptors were included. Clinical studies concerning BBs were considered eligible. The Newcastle-Ottawa scale was used for the quality assessment of clinical studies. A total of 614 study references were retrieved. Forty-six preclinical studies were included. In in vitro studies, propranolol, a non-selective BB, significantly decreased proliferation, migration and invasion of TNBC cells. Consistently, in in vivo studies, propranolol inhibited metastasis, angiogenesis and tumor growth. Clinical studies, reporting evidence from a total of four distinct retrospective observational cohort studies, showed a beneficial effect of BBs in TNBC treatment. The overall quality of the clinical evidence collected was low. Preclinical evidence collected in this systematic review are in line with the results reported in the clinical studies retrieved, pointing towards a beneficial effect of BB in the treatment of TNBC. However, given the overall low quality of available evidence, no definite conclusion may be drawn.


Asunto(s)
Antagonistas Adrenérgicos beta/uso terapéutico , Reposicionamiento de Medicamentos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Humanos , Neovascularización Patológica , Estudios Observacionales como Asunto , Estudios Retrospectivos
2.
J Cell Biol ; 119(3): 629-41, 1992 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1383237

RESUMEN

Hepatocyte Growth Factor (HGF, also known as Scatter Factor) is a powerful mitogen or motility factor in different cells, acting through the tyrosine kinase receptor encoded by the MET protooncogene. Endothelial cells express the MET gene and expose at the cell surface the mature protein (p190MET) made of a 50 kD (alpha) subunit disulfide linked to a 145-kD (beta) subunit. HGF binding to endothelial cells identifies two sites with different affinities. The higher affinity binding site (Kd = 0.35 nM) corresponds to the p190MET receptor. Sub-nanomolar concentrations of HGF, but not of a recombinant inactive precursor, stimulate the receptor kinase activity, cell proliferation and motility. HGF induces repairs of a wound in endothelial cell monolayer. HGF stimulates the scatter of endothelial cells grown on three-dimensional collagen gels, inducing an elongated phenotype. In the rabbit cornea, highly purified HGF promotes neovascularization at sub-nanomolar concentrations. HGF lacks activities related to hemostasis-thrombosis, inflammation and endothelial cells accessory functions. These data show that HGF is an in vivo potent angiogenic factor and in vitro induces endothelial cells to proliferate and migrate.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Endotelio Vascular/fisiología , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Neovascularización Patológica , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Animales , Línea Celular , Células Cultivadas , Córnea/citología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/genética , Humanos , Cinética , Sustancias Macromoleculares , Ratones , Mutagénesis Sitio-Dirigida , Proteínas Proto-Oncogénicas c-met , Proto-Oncogenes , Conejos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Venas Umbilicales
3.
Oxid Med Cell Longev ; 2018: 9765027, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538807

RESUMEN

Endothelial cells (ECs) are dynamic cells that turn from growth into senescence, the latter being associated with cellular dysfunction, altered metabolism, and age-related cardiovascular diseases. Aldehyde dehydrogenase 2 (ALDH2) is a mitochondrial enzyme metabolizing acetaldehyde and other toxic aldehydes, such as 4-hydroxynonenal (4-HNE). In conditions in which lipid peroxidation products and reactive oxygen species (ROS) are accumulated, ECs become dysfunctional and significantly contribute to the progression of vascular-dependent diseases. The aim of the present study has been to investigate whether inhibition of ALDH2 alters endothelial functions together with the impairment of bioenergetic functions, accelerating the acquisition of a senescent phenotype. HUVECs transfected with siRNA targeting ALDH2 or treated with daidzin, an ALDH2 inhibitor, were used in this study. We observed an alteration in cell morphology associated with endothelial dysfunctions. Loss of ALDH2 reduced cell proliferation and migration and increased paracellular permeability. To assess bioenergetic function in intact ECs, extracellular flux analysis was carried out to establish oxygen consumption rates (OCR). We observed a decrease in mitochondrial respiration and reserve capacity that coincided with SA-ß-Gal accumulation and an increase in p21 and p53 expression in siALDH2 or daidzin-treated HUVECs. Treatment with N-acetyl-L-cysteine (NAC) reduced endothelial dysfunctions mediated by siALDH2, indicating that oxidative stress downstream to siALDH2 plays an instrumental role. Our results highlight that ALDH2 impairment accelerates the acquisition of a premature senescent phenotype, a change likely to be associated with the observed reduction of mitochondrial respiration and reserve capacity.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/metabolismo , Respiración de la Célula/fisiología , Senescencia Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Mitocondrias/metabolismo , Humanos
4.
Cell Death Differ ; 13(7): 1088-96, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16410806

RESUMEN

Recent evidences suggest that Abeta peptides modulate endothelial cell (EC) functions. At low concentrations, Abeta1-40 enhances the pro-angiogenic activity of FGF-2, whereas deposition of excess Abeta causes EC dysfunction and cerebral amyloid angiopathy (CAA). We investigated whether FGF-2 attenuates EC dysfunction caused by pathological Abeta levels. We studied Abeta1-40 on EC survival, as well as on signals responsible of their angiogenic phenotype. At 5-50 microM Abeta1-40 reduced EC population, caused apoptosis, downregulated FGF-2 production, inhibited FGF-2 binding to heparin, and FGFR1 phosphorylation. Toxic effects were owing to lack of FGF-2 stimulation, as EC overexpressing FGF-2 displayed extraordinary resistance to Abeta1-40 injuries. The FGF-2 mechanism responsible for reversing damages, involves the downstream enhancement of Akt, a pathway independent of eNOS activation. In conclusion, we demonstrate that FGF-2 protects EC from the effects of excess Abeta1-40, suggesting that it may attenuate the consequences of Abeta deposition in pathologies as CAA.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Células Endoteliales/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fragmentos de Péptidos/farmacología , Animales , Western Blotting , Células CHO , Caspasa 3 , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Cricetulus , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Activación Enzimática/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
5.
FEBS Lett ; 581(4): 702-6, 2007 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-17274989

RESUMEN

Oxidative stress is one of the factor contributing to blood brain barrier degeneration. This phenomenon is observed during pathological conditions such as Alzheimer's disease or cerebral amyloid angiopathy in which brain haemorrhages are very frequent. Both diseases are characterized by beta amyloid peptide deposition either in neurons or in vessels. Oxidative stress leads to impairment of mitochondrial functions and apoptotic cell death subsequent to caspases activation. In this paper we demonstrate that BH4 domain of Bcl-xl administrated to endothelial cells as the conjugated form with TAT peptide, reverts Abeta-induced apoptotic cell death by activating a survival programme which is Akt/endothelial nitric oxide synthase dependent.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Endotelio Vascular/efectos de los fármacos , Productos del Gen tat/farmacología , Fragmentos de Péptidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Activación Enzimática/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxidación-Reducción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo
6.
J Clin Invest ; 99(11): 2625-34, 1997 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9169492

RESUMEN

Systemic administration of the nitric oxide (NO) synthase inhibitor Nomega-nitro--arginine methyl ester (L-NAME) to rabbits bearing a corneal implant blocked vascular endothelial growth factor (VEGF), but not basic fibroblast growth factor (bFGF)-induced angiogenesis. L-NAME completely blocked angiogenesis induced by VEGF-transfected MCF-7 breast carcinoma cells and the cells remained dormant in the cornea. Postcapillary endothelial cell migration and growth induced by VEGF were blocked by both the NO synthase inhibitor Nomega-mono-methyl--arginine and by the guanylate cyclase inhibitor LY 83583. We conclude that NO is a downstream imperative of VEGF-, but not bFGF-induced angiogenesis, and propose that the NO synthase/guanylate cyclase pathway is a potential target for controlling tumor angiogenesis in response to VEGF. Our studies support recent evidence that VEGF and bFGF induce angiogenesis by different mechanistic pathways using the alphavbeta5 and alphavbeta3 integrins, respectively.


Asunto(s)
Córnea/irrigación sanguínea , Factores de Crecimiento Endotelial/fisiología , Inhibidores Enzimáticos/administración & dosificación , Factor 2 de Crecimiento de Fibroblastos/fisiología , Linfocinas/fisiología , NG-Nitroarginina Metil Éster/administración & dosificación , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa/fisiología , Animales , Trasplante de Córnea , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Conejos , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
7.
J Clin Invest ; 91(6): 2504-12, 1993 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7685771

RESUMEN

To elucidate mechanisms underlying neovascularization that accompanies certain chronic immune/inflammatory disorders, the effects of interferon-alpha (IFN-alpha) and interleukin 2 (IL-2) on endothelial cell (EC) growth in vitro and angiogenesis in vivo were studied. Preincubation of cultured human ECs with IFN-alpha, followed by exposure to IL-2, resulted in effective stimulation of cell growth, whereas either cytokine alone had only a slight effect. The combination of IFN-alpha/IL-2 induced an angiogenic response in the rabbit cornea. IL-2 receptor expression was enhanced on IFN-alpha-treated ECs: p55 was increased and p70 was induced. 125I-IL-2 binding to ECs treated with IFN-alpha was enhanced (Kd from approximately 7 nM to approximately 260 pM with IFN-alpha), and anti-p55 IgG blocked 125I-IL-2/EC interaction as well as IL-2-mediated EC proliferation. Consistent with these findings in cell culture, immunohistologic studies demonstrated p55 and p70 antigen in the vasculature of rheumatoid joints, but not in normal joint tissue. Exposure of cultured ECs to IFN-alpha increased levels of intracellular EC basic fibroblast growth factor (bFGF), and subsequent addition of IL-2 led to bFGF release into the medium. The observation that anti-bFGF IgG largely blocked EC proliferation in response to IFN-alpha/IL-2 suggested that bFGF was a critical agent in this setting. These data suggest a mechanism rendering ECs responsive to IL-2 which may be relevant in immune/inflammatory disorders: IFN-alpha-mediated induction of functional EC receptors for IL-2, which drives cell proliferation by a mechanism dependent on increased synthesis and release of bFGF.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Interferón-alfa/farmacología , Interleucina-2/farmacología , Neovascularización Patológica , Animales , Artritis Reumatoide/patología , División Celular , Células Cultivadas , Córnea/irrigación sanguínea , Sinergismo Farmacológico , Endotelio Vascular/crecimiento & desarrollo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Articulaciones/patología , Conejos , Receptores de Interleucina-2/metabolismo , Venas Umbilicales/citología
8.
J Clin Invest ; 94(5): 2036-44, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7525653

RESUMEN

We evaluated the effects of nitric oxide (NO) generators and endogenous production of NO elicited by substance P (SP) in the angiogenesis process. Angiogenesis was monitored in the rabbit cornea in vivo and in vitro by measuring the growth and migration of endothelial cells isolated from coronary postcapillary venules. The angiogenesis promoted in the rabbit cornea by [Sar9]-SP-sulfone, a stable and selective agonist for the tachykinin NK1 receptor, and by prostaglandin E1 (PGE1), was potentiated by sodium nitroprusside (SNP). Conversely, the NO synthase inhibitor N omega-nitro-L-arginine methyl ester (L-NAME), given systemically, inhibited angiogenesis elicited by [Sar9]-SP-sulfone and by PGE1. Endothelial cells exposed to SNP exhibited an increase in thymidine incorporation and in total cell number. Exposure of the cells to NO generating drugs, such as SNP, isosorbide dinitrate, and glyceryl trinitrate, produced a dose-dependent increase in endothelial cell migration. Capillary endothelial cell proliferation and migration produced by SP were abolished by pretreatment with the NO synthase inhibitors N omega-mono-methyl-L-arginine (L-NMMA), N omega-nitro-L-arginine (L-NNA), and L-NAME. Exposure of the cells to SP activated the calcium-dependent NO synthase. Angiogenesis and endothelial cell growth and migration induced by basic fibroblast growth factor were not affected by NO synthase inhibitors. These data indicate that NO production induced by vasoactive agents, such as SP, functions as an autocrine regulator of the microvascular events necessary for neovascularization and mediates angiogenesis.


Asunto(s)
Endotelio Vascular/citología , Neovascularización Patológica/etiología , Óxido Nítrico/fisiología , Sustancia P/farmacología , Aminoácido Oxidorreductasas/antagonistas & inhibidores , Animales , Arginina/análogos & derivados , Arginina/farmacología , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , GMP Cíclico/biosíntesis , GMP Cíclico/sangre , ADN/biosíntesis , Endotelio Vascular/efectos de los fármacos , NG-Nitroarginina Metil Éster , Óxido Nítrico Sintasa , Nitroprusiato/farmacología , Agregación Plaquetaria/efectos de los fármacos , Conejos
9.
J Clin Invest ; 87(3): 986-95, 1991 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1705569

RESUMEN

This study was designed to identify the set of functions activated in cultured endothelial cells by the hematopoietic growth factors, granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage-colony-stimulating factor (GM-CSF), and to compare them with those elicited by prototypic cytokines active on these cells. Moreover, indications as to the in vivo relevance of in vitro effects were obtained. G-CSF and GM-CSF induced endothelial cells to proliferate and migrate. In contrast, unlike appropriate reference cytokines (IL-1 and tumor necrosis factor, IFN-gamma), G-CSF and GM-CSF did not modulate endothelial cell functions related to hemostasis-thrombosis (production of procoagulant activity and of platelet activating factor), inflammation (expression of leukocyte adhesion molecule-1 and production of platelet activating factor), and accessory function (expression of class II antigens of MHC). Other colony-stimulating factors (IL-3 and macrophage-colony-stimulating factor) were inactive on all functions tested. In comparison to basic fibroblast growth factor (bFGF), G-CSF and GM-CSF induced lower maximal proliferation of endothelial cells, whereas migration was of the same order of magnitude. G-CSF and GM-CSF stimulated repair of mechanically wounded endothelial monolayers. Exposure to both cytokines induced shape changes and cytoskeletal reorganization consistent with a migratory phenotype. To explore the in vivo relevance of the in vitro effects of these cytokines on endothelium, we studied the angiogenic activity of human G-CSF in the rabbit cornea. G-CSF, but not the heat-inactivated molecule, had definite angiogenic activity, without any sign of inflammatory reactions. G-CSF was less active than bFGF. However, the combination of a nonangiogenic dose of bFGF with G-CSF resulted in an angiogenic response higher than that elicited by either individual cytokines. Thus, G-CSF and GM-CSF induce endothelial cells to express an activation/differentiation program (including proliferation and migration) related to angiogenesis.


Asunto(s)
Endotelio Vascular/fisiología , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Animales , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Citocinas/farmacología , Citoesqueleto/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas In Vitro , Neovascularización Patológica , Conejos , Cicatrización de Heridas
10.
Mol Biol Cell ; 10(2): 313-27, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9950679

RESUMEN

Exogenous gangliosides affect the angiogenic activity of fibroblast growth factor-2 (FGF-2), but their mechanism of action has not been elucidated. Here, a possible direct interaction of sialo-glycolipids with FGF-2 has been investigated. Size exclusion chromatography demonstrates that native, but not heat-denatured, 125I-FGF-2 binds to micelles formed by gangliosides GT1b, GD1b, or GM1. Also, gangliosides protect native FGF-2 from trypsin digestion at micromolar concentrations, the order of relative potency being GT1b > GD1b > GM1 = GM2 = sulfatide > GM3 = galactosyl-ceramide, whereas asialo-GM1, neuraminic acid, and N-acetylneuramin-lactose were ineffective. Scatchard plot analysis of the binding data of fluorochrome-labeled GM1 to immobilized FGF-2 indicates that FGF-2/GM1 interaction occurs with a Kd equal to 6 microM. This interaction is inhibited by the sialic acid-binding peptide mastoparan and by the synthetic fragments FGF-2(112-129) and, to a lesser extent, FGF-2(130-155), whereas peptides FGF-2(10-33), FGF-2(39-59), FGF-2(86-96), and the basic peptide HIV-1 Tat(41-60) were ineffective. These data identify the COOH terminus of FGF-2 as a putative ganglioside-binding region. Exogenous gangliosides inhibit the binding of 125I-FGF-2 to high-affinity tyrosine-kinase FGF-receptors (FGFRs) of endothelial GM 7373 cells at micromolar concentrations. The order of relative potency was GT1b > GD1b > GM1 > sulfatide a = sialo-GM1. Accordingly, GT1b,GD1b, GM1, and GM2, but not GM3 and asialo-GM1, prevent the binding of 125I-FGF-2 to a soluble, recombinant form of extracellular FGFR-1. Conversely, the soluble receptor and free heparin inhibit the interaction of fluorochrome-labeled GM1 to immobilized FGF-2. In agreement with their FGFR antagonist activity, free gangliosides inhibit the mitogenic activity exerted by FGF-2 on endothelial cells in the same range of concentrations. Also in this case, GT1b was the most effective among the gangliosides tested while asialo-GM1, neuraminic acid, N-acetylneuramin-lactose, galactosyl-ceramide, and sulfatide were ineffective. In conclusion, the data demonstrate the capacity of exogenous gangliosides to interact with FGF-2. This interaction involves the COOH terminus of the FGF-2 molecule and depends on the structure of the oligosaccharide chain and on the presence of sialic acid residue(s) in the ganglioside molecule. Exogenous gangliosides act as FGF-2 antagonists when added to endothelial cell cultures. Since gangliosides are extensively shed by tumor cells and reach elevated levels in the serum of tumor-bearing patients, our data suggest that exogenous gangliosides may affect endothelial cell function by a direct interaction with FGF-2, thus modulating tumor neovascularization.


Asunto(s)
Endotelio Vascular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Gangliósidos/metabolismo , Animales , Sitios de Unión , Secuencia de Carbohidratos , Bovinos , Línea Celular Transformada , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/farmacología , Gangliósidos/química , Gangliósidos/farmacología , Humanos , Cinética , Datos de Secuencia Molecular , Neovascularización Patológica/etiología , Proteínas Tirosina Quinasas/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
11.
Clin Exp Metastasis ; 23(3-4): 187-201, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17028923

RESUMEN

Endothelial cells express two dependent intercellular adhesion molecules: vascular endothelial (VE)-cadherin, specific for endothelial cells, and N-cadherin, also present in neuronal, lens, skeletal and heart muscle cells, osteoblasts, pericytes and fibroblasts. While there exists a vast amount of evidence that VE-cadherin promotes angiogenesis, the role of N-cadherin still remains to be elucidated. We found that a soluble 90-kDa fragment N-cadherin promotes angiogenesis in the rabbit cornea assay and in the chorioallantoic assay when cleaved enzymatically from the extracellular domain of N-cadherin. Soluble N-cadherin stimulates migration of endothelial cells in the wound healing assay and stimulates phosphorylation of extracellular regulated kinase. In vitro experiments with PD173074 and knock-down of N-cadherin and fibroblast growth factor (FGF)-receptor, showed that the pro-angiogenic effect of soluble N-cadherin is N-cadherin- and FGF-receptor-dependent. Our results suggest that soluble N-cadherin stimulates migration of endothelial cells through the FGF-receptor.


Asunto(s)
Cadherinas/farmacología , Endotelio Vascular/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Córnea/irrigación sanguínea , Humanos , Modelos Biológicos , Fosforilación , Conejos , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Factores de Tiempo , Cicatrización de Heridas
12.
J Natl Cancer Inst ; 69(2): 483-7, 1982 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6180208

RESUMEN

In vivo cell populations at high risk of neoplastic transformation have been shown to acquire the ability to induce new formation of vessels. The present experiments tested whether the same change occurred during neoplastic transformation in vitro. In four cell populations (human HBL 100 mammary epithelium, BALB/c fibroblasts, C57BL-MG epithelium, and Syrian golden hamster embryo cells), angiogenic capacity appeared during their cultivation in vitro and was evident long before a neoplastic transformation could be recognized. The data were interpreted to support the hypothesis that acquisition of angiogenic capacity by a cell population normally devoid of this capacity indicates an increased risk of neoplastic transformation.


Asunto(s)
Transformación Celular Neoplásica/patología , Neovascularización Patológica , Animales , Línea Celular , Córnea/irrigación sanguínea , Cricetinae , Femenino , Humanos , Masculino , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Riesgo
13.
J Natl Cancer Inst ; 69(2): 475-82, 1982 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6180207

RESUMEN

The interstitial fluid of MTW9A and Walker carcinomas and their ethanol extract induced strong angiogenic response in the rabbit (New Zealand White) corneal test. The fluid collected in vivo was rich in E-type prostaglandins, and prostaglandin E1 (PGE1) in particular was strongly angiogenic at the lowest dose as compared with the angiogenic responses of prostaglandins E2, I2, and F2 alpha. Neoplastic fibroblasts also induced a strong angiogenic response, but in indomethacin-treated rabbits neovascularization failed to occur. Copper was concentrated in the cornea during PGE1-induced neovascularization, and copper-deficient rabbits were unable to mount an angiogenic response in the corneal test. Ceruloplasmin, the copper carrier of plasma, was found to be angiogenic at high doses. In indomethacin-treated rabbits, however, ceruloplasmin at the same high doses failed to induce angiogenesis. The experiments are interpreted to indicate that angiogenesis is the end result of a sequence of events, two of which are PGE1 production and copper mobilization in the tissue where neovascularization occurs.


Asunto(s)
Cobre/fisiología , Neovascularización Patológica , Prostaglandinas E/fisiología , Alprostadil , Animales , Carcinoma 256 de Walker/metabolismo , Ceruloplasmina/farmacología , Cobre/análisis , Cobre/farmacología , Córnea/análisis , Córnea/irrigación sanguínea , Dinoprost , Dinoprostona , Epoprostenol/farmacología , Femenino , Indometacina/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/metabolismo , Neovascularización Patológica/efectos de los fármacos , Prostaglandinas E/farmacología , Prostaglandinas F/farmacología , Conejos , Ratas , Ratas Endogámicas
14.
J Natl Cancer Inst ; 69(5): 1183-8, 1982 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-6182332

RESUMEN

The ability to induce new formation of capillaries in the cornea was tested for ceruloplasmin, the copper carrier of serum, for fragments of the ceruloplasmin molecule with and without copper, for heparin, and for glycyl-L-histidyl-L-lysine, bound or not bound to copper ions. Male or female 2- to 3-kg New Zealand White rabbits were used. These experiments were prompted by the previous observation of copper accumulation in the cornea during angiogenesis and by the inability of copper-deficient rabbits to mount an angiogenic response. The results showed that the three different molecules were all able to induce angiogenesis provided that they were bound to copper. Fragments of the ceruloplasmin molecule also induced angiogenesis but only when copper was bound to the peptides. The data are interpreted to indicate that copper ions are involved in the sequence of events leading to angiogenesis and that the carrier molecules may be of quite a different nature.


Asunto(s)
Ceruloplasmina/farmacología , Cobre/farmacología , Córnea/irrigación sanguínea , Neovascularización Patológica , Animales , Córnea/efectos de los fármacos , Femenino , Sustancias de Crecimiento/farmacología , Heparina/farmacología , Masculino , Oligopéptidos/farmacología , Conejos
15.
J Natl Cancer Inst ; 90(8): 587-96, 1998 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9554441

RESUMEN

BACKGROUND: Angiogenesis (formation of new blood vessels) is associated with tumor growth and metastasis in patients with solid tumors, including those of the head and neck. Nitric oxide (NO) production may contribute to these processes. We assessed the role of the NO pathway in angiogenesis and tumor progression in patients with head and neck cancer. METHODS: Biochemical assays were used to measure NO synthase (NOS) activity and cyclic guanosine monophosphate (cGMP) levels in specimens of tumor and normal mucosa obtained from 27 patients. Microvessels in tumor specimens were identified by CD-31-specific immunohistochemical staining. Associations between microvessel densities, levels of NOS, and cGMP were examined by use of two-sided statistical tests. Tumor specimens and human squamous carcinoma A-431 cells were grown as explants on the corneas of rabbits, and the effect of the NOS inhibitor N(omega)-nitro-L-arginine-methyl ester (L-NAME) was tested. RESULTS: Levels of total NOS, inducible NOS, and cGMP were higher in tumor specimens than in specimens of normal mucosa (all P<.0001). Tumor specimens from patients with lymph node metastases presented a higher total NOS activity (P = .005) and were markedly more vascularized than tumor specimens from patients with no lymph node involvement (P = .0002). Microvessel density at the tumor edge was an independent predictor of metastasis for this series of patients (odds ratio = 1.19; 95% confidence interval = 1.07-2.89; P = .04). A-431 cells and tumor specimens exhibiting high levels of NOS activity induced angiogenesis in the rabbit cornea assay; when NO production was blocked, tumor angiogenesis and growth were repressed. CONCLUSIONS: The NO pathway appears to play a key role in tumor angiogenesis and spread in patients with head and neck cancer.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Neovascularización Patológica/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/metabolismo , Adulto , Anciano , Carcinoma de Células Escamosas/enzimología , GMP Cíclico/metabolismo , Progresión de la Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Neoplasias de Cabeza y Cuello/enzimología , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Análisis Multivariante , NG-Nitroarginina Metil Éster/farmacología , Estadificación de Neoplasias , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II , Reacción en Cadena de la Polimerasa/métodos , Transcripción Genética , Células Tumorales Cultivadas
16.
J Natl Cancer Inst ; 87(3): 213-9, 1995 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-7535859

RESUMEN

BACKGROUND: Vascular endothelial growth factor (VEGF) is a secreted endothelial-specific growth factor that is angiogenic in vivo. It is commonly expressed in a range of carcinomas. PURPOSE: The study was designed to investigate the effect of constitutive expression of VEGF on tumor formation by estrogen-dependent human MCF-7 breast carcinoma cells. METHODS: A full-length complementary DNA encoding the shortest isoform of VEGF (VEGF121) was stably transfected into MCF-7 cells. Transfected clones were screened for VEGF121 messenger RNA (mRNA) expression by ribonuclease protection analysis and for secretion of VEGF121 protein by Western blot analysis. Secretion of biologically active VEGF121 by transfectants was confirmed by 1) a competitive radioreceptor-binding assay, 2) stimulation of the growth of microvascular endothelial cells in vitro, and 3) potent angiogenic activity in the rabbit corneal assay. Tumor models were then established by subcutaneously implanting wild-type or VEGF121-transfected MCF-7 cells, together with either mouse BALB/3T3 clone A31 fibroblasts or human MDA-435S breast carcinoma cells, into ovariectomized nude mice either with or without a separately implanted slow-release estrogen pellet. Tumor vascularity was quantitatively assessed by capillary vessel counting after staining with the pan-endothelial marker CD31. RESULTS: Stable VEGF121-overexpressing MCF-7 cells were isolated and designated V12 cells. When implanted into the rabbit cornea, V12 cells elicited a strong directional outgrowth of capillaries. The growth rate of V12 cells in vitro was indistinguishable from that of MCF-7 wild-type cells. V12 cells formed faster growing tumors than did wild-type cells (P < .01) when xenografted subcutaneously into nude mice with either 3T3 fibroblasts or MDA-435S cells. Tumors formed from V12 cells were more vascular (P < .01) and showed a heterogeneous distribution of vessels when compared with the homogeneous distribution seen in tumors formed from wild-type cells. VEGF121 overexpression had no effect on hormone dependence or tamoxifen sensitivity of tumor formation by MCF-7 cells in mice. No macroscopic evidence for metastasis from subcutaneous implants was obtained. CONCLUSIONS: VEGF121 expression by breast carcinoma cells confers a growth advantage in vivo but not in vitro. Tumors formed by V12 transfectants were more vascular than those formed by wild-type MCF-7 cells, and we surmise that the growth advantage arises from increased tumor vascularization induced by VEGF121. IMPLICATIONS: Tumor formation by V12 cells could provide a useful model for the assessment of anti-angiogenic drugs.


Asunto(s)
Neoplasias de la Mama/patología , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular , Linfocinas/fisiología , Células 3T3 , Secuencia de Aminoácidos , Animales , Western Blotting , Neoplasias de la Mama/irrigación sanguínea , ADN Complementario , Factores de Crecimiento Endotelial/biosíntesis , Humanos , Linfocinas/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Neovascularización Patológica/fisiopatología , Conejos , Proteínas Recombinantes/biosíntesis , Transfección , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
17.
Cancer Res ; 41(12 Pt 1): 5060-3, 1981 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-6171345

RESUMEN

The capacity to induce new formation of capillaries was tested in cells attached to plastics. It is known that large plastic coverslips implanted s.c. in CBA mice produce sarcomas more rapidly and in a greater number than do small coverslips. We observed that within a few weeks after implantation the cells attached to the large coverslips showed an angiogenic capacity about 5-fold greater than that of the cells attached to the small coverslips. Months before a sarcoma was evident, angiogenesis induced by the cells attached to the large coverslips predicted the high risk of neoplastic transformation by large coverslips.


Asunto(s)
Transformación Celular Neoplásica/inducido químicamente , Neovascularización Patológica/efectos de los fármacos , Plásticos/farmacología , Sarcoma Experimental/irrigación sanguínea , Animales , Femenino , Reacción a Cuerpo Extraño/complicaciones , Ratones , Ratones Endogámicos CBA , Conejos , Sarcoma Experimental/inducido químicamente
18.
Cancer Res ; 57(9): 1814-9, 1997 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9135027

RESUMEN

Recent analysis of bladder tumors has correlated expression of the neurokine midkine (MK) with poor patient survival. To examine a role for MK and the related pleiotrophin (PTN) in tumorigenesis, they were overexpressed in MCF-7 breast carcinoma cells. Expression had no effect on in vitro growth but conferred a growth advantage in vivo. Enhanced tumor growth correlated with increased vascular density and endothelial proliferation, implicating an angiogenic role for MK and PTN. Angiogenic activity of MK and PTN was confirmed in the rabbit corneal assay. Our data therefore identify two novel targets for antiangiogenic drug development.


Asunto(s)
Proteínas Portadoras/fisiología , Citocinas/fisiología , Sustancias de Crecimiento/fisiología , Neovascularización Fisiológica , Animales , División Celular , Endotelio Vascular/citología , Femenino , Humanos , Midkina , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Conejos , Transfección , Células Tumorales Cultivadas
19.
Cancer Res ; 59(10): 2417-24, 1999 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10344752

RESUMEN

Angiogenesis has been identified as an important target for antineoplastic therapy. The use of purine analogue antimetabolites in combination chemotherapy of solid tumors has been proposed. To assess the possibility that selected purine analogues may affect tumor neovascularization, 6-methylmercaptopurine riboside (6-MMPR), 6-methylmercaptopurine, 2-aminopurine, and adenosine were evaluated for the capacity to inhibit angiogenesis in vitro and in vivo. 6-MMPR inhibited fibroblast growth factor-2 (FGF2)-induced proliferation and delayed the repair of mechanically wounded monolayer in endothelial GM 7373 cell cultures. 6-MMPR also inhibited the formation of solid sprouts within fibrin gel by FGF2-treated murine brain microvascular endothelial cells and the formation of capillary-like structures on Matrigel by murine aortic endothelial cells transfected with FGF2 cDNA. 6-MMPR affected FGF2-induced intracellular signaling in murine aortic endothelial cells by inhibiting the phosphorylation of extracellular signal-regulated kinase-2. The other molecules were ineffective in all of the assays. In vivo, 6-MMPR inhibited vascularization in the chick embryo chorioallantoic membrane and prevented blood vessel formation induced by human endometrial adenocarcinoma specimens grafted onto the chorioallantoic membrane. Also, topical administration of 6-MMPR caused the regression of newly formed blood vessels in the rabbit cornea. Thus, 6-MMPR specifically inhibits both the early and the late phases of the angiogenesis process in vitro and exerts a potent anti-angiogenic activity in vivo. These results provide a new rationale for the use of selected purine analogues in combination therapy of solid cancer.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Metiltioinosina/farmacología , Neovascularización Fisiológica/efectos de los fármacos , 2-Aminopurina/farmacología , Adenocarcinoma/irrigación sanguínea , Adenosina/farmacología , Alantoides/irrigación sanguínea , Alantoides/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Aorta/citología , Encéfalo/irrigación sanguínea , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Bovinos , Células Cultivadas , Embrión de Pollo , Corion/irrigación sanguínea , Corion/efectos de los fármacos , Córnea/irrigación sanguínea , Córnea/efectos de los fármacos , Depresión Química , Neoplasias Endometriales/irrigación sanguínea , Endotelio Vascular/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Mercaptopurina/análogos & derivados , Mercaptopurina/farmacología , Ratones , Proteína Quinasa 1 Activada por Mitógenos , Trasplante de Neoplasias , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Conejos , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transfección
20.
Cancer Res ; 55(20): 4729-38, 1995 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-7553656

RESUMEN

The human endometrial adenocarcinoma HEC-1-B cell line was transfected with an expression vector harboring the human basic fibroblast growth factor (bFGF) cDNA under the control of the human beta-actin gene promoter. Stable transfectants were obtained in which a constitutive, limited overexpression of M(r) 24,000, 22,000, and 18,000 bFGF isoforms was observed. When transfectants were screened for the capacity to release the growth factor, significant amounts of bFGF were present in the conditioned medium and extracellular matrix of the bFGF-B9 clone but not of the bFGF-A8 clone, even though both cell lines produced similar levels of intracellular bFGF. When compared to parental cells, bFGF-B9 cells showed down-regulation of tyrosine kinase fibroblast growth factor receptors along with up-regulation of urokinase-type plasminogen activator expression which was abolished by incubation of the cell cultures with neutralizing anti-bFGF antibody. In vivo, bFGF-B9 cells formed highly vascularized tumors growing faster than parental cells when injected s.c. in nude mice. Also, they were more potent than nontransfected cells in inducing an angiogenic response in the rabbit cornea assay. In contrast, the bFGF-A8 cell phenotype was indistinguishable from parental cells both in vitro and in vivo. In conclusion, clonal differences exist within the HEC-1-B cell line in the capacity to release bFGF. bFGF export by human endometrial adenocarcinoma cells results in autocrine and paracrine effects that confer a growth advantage in vivo associated with increased neovascularization.


Asunto(s)
Adenocarcinoma Papilar/patología , Neoplasias Endometriales/patología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neovascularización Patológica/patología , Anciano , Animales , División Celular , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Expresión Génica , Humanos , Técnicas In Vitro , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/patología , ARN Mensajero/genética , Transfección , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda