Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Histochem Cell Biol ; 145(6): 629-36, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26748643

RESUMEN

The embryonic origin of lymphatic endothelial cells (LECs) has been a matter of controversy since more than a century. However, recent studies in mice have supported the concept that embryonic lymphangiogenesis is a complex process consisting of growth of lymphatics from specific venous segments as well as the integration of lymphangioblasts into the lymphatic networks. Similarly, the mechanisms of adult lymphangiogenesis are poorly understood and have rarely been studied. We have recently shown that endothelial progenitor cells isolated from the lung of adult mice have the capacity to form both blood vessels and lymphatics when grafted with Matrigel plugs into the skin of syngeneic mice. Here, we followed up on these experiments and studied the behavior of host leukocytes during lymphangiogenesis in the Matrigel plugs. We observed a striking co-localization of CD45(+) leukocytes with the developing lymphatics. Numerous CD45(+) cells expressed the LEC marker podoplanin and were obviously integrated into the lining of lymphatic capillaries. This indicates that, similar to inflammation-induced lymphangiogenesis in man, circulating CD45(+) cells of adult mice are capable of initiating lymphangiogenesis and of adopting a lymphvasculogenic cellular differentiation program. The data are discussed in the context of embryonic and inflammation-induced lymphangiogenesis.


Asunto(s)
Antígenos Comunes de Leucocito/inmunología , Leucocitos/inmunología , Vasos Linfáticos/inmunología , Animales , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/inmunología , Leucocitos/citología , Vasos Linfáticos/citología , Ratones , Ratones Endogámicos C57BL
2.
J Cell Biol ; 132(6): 1177-88, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8601593

RESUMEN

In angiogenesis associated with tissue repair and disease, fibrin and inflammatory mediators are often involved. We have used three-dimensional fibrin matrices to investigate the humoral requirements of human microvascular endothelial cells (hMVEC) to form capillary-like tubular structures. bFGF and VEGF165 were unable to induce tubular structures by themselves. Simultaneous addition of one or both of these factors with TNFalpha induced outgrowth of tubules, the effect being the strongest when bFGF, VEGF165, and TNFalpha were added simultaneously. Exogenously added u-PA, but not its nonproteolytic amino-terminal fragment, could replace TNFalpha, suggesting that TNFalpha-induced u-PA synthesis was involved. Soluble u-PA receptor (u-PAR) or antibodies that inhibited u-PA activity prevented the formation of tubular structures by 59-99%. epsilon-ACA and trasylol which inhibit the formation and activity of plasmin reduced the extent of tube formation by 71-95%. TNFalpha or u-PA did not induce tubular structures without additional growth factors. bFGF and VEGF165 enhanced of the u-PAR by 72 and 46%, but TNFalpha itself also increased u-PAR in hMVEC by 30%. Induction of mitogenesis was not the major contribution of bFGF and VEGF165 because the cell number did not change significantly in the presence of TNFalpha, and tyrphostin A47, which inhibited mitosis completely, reduced the formation of tubular structures only by 28-36%. These data show that induction of cell-bound u-PA activity by the cytokine TNFalpha is required in addition to the angiogenic factors VEGF165 and/or bFGF to induce in vitro formation of capillary-like structures by hMVEC in fibrin matrices. These data may provide insight in the mechanism of angiogenesis as occurs in pathological conditions.


Asunto(s)
Capilares/citología , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/efectos de los fármacos , Fibrina , Factor 2 de Crecimiento de Fibroblastos/farmacología , Linfocinas/farmacología , Neovascularización Fisiológica/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , División Celular/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Endotelio Vascular/ultraestructura , Fibrinolisina/fisiología , Humanos , Morfogénesis/efectos de los fármacos , Neovascularización Patológica/fisiopatología , Activadores Plasminogénicos/fisiología , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
3.
Clin Oral Implants Res ; 20(1): 24-30, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19126104

RESUMEN

OBJECTIVES: The present study was conducted to test the hypothesis that preshaped polylactic acid (PLA) implants loaded with recombinant human bone morphogenic protein 2 (rhBMP-2) can induce bone formation in a rat ectopic model. MATERIALS AND METHODS: Two groups of porous cylindrical poly-DL-lactic acid implants of 8-mm diameter were produced by gas foaming with CO(2), incorporating 48 and 96 microg rhBMP-2, respectively, into each implant. Blank PLA implants were used as controls. The release of BMPs and the induction of alkaline phosphatase were assessed in vitro. Osteoinduction in vivo was tested by insertion of 15 implants from each group into the gluteal muscles of Wistar rats. Five implants from each group were retrieved after 6, 13 and 26 weeks and assessed using flat panel volume detector computed tomography and light microscopy. RESULTS: Both groups of implants showed increased release of rhBMP-2 during the first 24-48 h, with a slightly higher amount being released from the implants with 48 microg. Release during subsequent intervals was <100 ng/72 h in the low-concentration group and >100 ng in the group with 96 microg rhBMP-2. Implants with 95 microg rhBMP-2 exhibited bone formation in vivo on the outside of the implants across the observation period of 26 weeks with invasion of bone into the pores, whereas implants with 48 microg rhBMP-2 failed to induce the formation of bone tissue. No bone formation was found in the control implants. CONCLUSIONS: The results suggest that release rates of rhBMP-2 for ectopic bone induction have to be >100 ng/72 h to maintain the osteoinductive activity of the tested porous PLA implants. This slow release system may have impact on alveolar bone augmentation procedures when used as individually preformed osteoinductive implants.


Asunto(s)
Implantes Absorbibles , Proteínas Morfogenéticas Óseas/farmacología , Portadores de Fármacos , Osteogénesis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Factor de Crecimiento Transformador beta/farmacología , Animales , Proteína Morfogenética Ósea 2 , Nalgas , Preparaciones de Acción Retardada , Relación Dosis-Respuesta a Droga , Humanos , Ácido Láctico , Masculino , Osificación Heterotópica , Poliésteres , Polímeros , Ratas , Ratas Wistar
4.
J Biomed Mater Res A ; 83(2): 455-62, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17477390

RESUMEN

The aim of the present report was to test a system for controlled release of recombinant human bone morphogenic protein (rhBMP2) incorporated into polylactic acid (PLA) implants. Incorporation of rhBMP2 into the polymer was accomplished by mixing rhBMP2 solution with granular powder of amorphous poly-DL-lactic acid, subsequent lyophilization, and high pressure CO(2) treatment at 100 bar. Porous cylindrical implants of 8 mm diameter and 3 mm thickness were fabricated with 100, 200, 400, and 800 microg BMP2/g polymer and submitted to in vitro testing. Polymer degradation was assessed during immersion of PLA implants into PBS for 176 days by measuring the inherent viscosity at days 0, 99, and 131. BMP2 release was evaluated by immersion of both the lyophilized powder and the implants into cell culture medium for up to 27 days. BMP2 release was assessed using a custom made ELISA. The biological activity of the released growth factor was determined by measuring the induction of alkaline phosphatase (AP) in C2C12 cells. There was a significant retardation in the release of BMP2 from the implants compared to the granular powder. Detectable amounts of BMP2 were found for all concentrations of BMP2 until the end of the observation period. Significant induction of AP was detected by BMP released from the implants after 3, 6, and 9 days. The present in-vitro study has shown that incorporation of rhBMP2 into PLA implants with subsequent slow release of biologically active growth factor is possible.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Preparaciones de Acción Retardada/metabolismo , Ácido Láctico/metabolismo , Polímeros/metabolismo , Proteínas Recombinantes/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Implantes Absorbibles , Fosfatasa Alcalina/biosíntesis , Animales , Proteína Morfogenética Ósea 2 , Línea Celular , Inducción Enzimática , Gases , Humanos , Concentración de Iones de Hidrógeno , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Microscopía Electrónica de Rastreo , Poliésteres
5.
Leukemia ; 31(2): 361-372, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27535218

RESUMEN

The interaction between vascular endothelial cells (ECs) and cancer cells is of vital importance to understand tumor dissemination. A paradigmatic cancer to study cell-cell interactions is classical Hodgkin Lymphoma (cHL) owing to its complex microenvironment. The role of the interplay between cHL and ECs remains poorly understood. Here we identify canonical WNT pathway activity as important for the mutual interactions between cHL cells and ECs. We demonstrate that local canonical WNT signaling activates cHL cell chemotaxis toward ECs, adhesion to EC layers and cell invasion using not only the Wnt-inhibitor Dickkopf, tankyrases and casein kinase 1 inhibitors but also knockdown of the lymphocyte enhancer binding-factor 1 (LEF-1) and ß-catenin in cHL cells. Furthermore, LEF-1- and ß-catenin-regulated cHL secretome promoted EC migration, sprouting and vascular tube formation involving vascular endothelial growth factor A (VEGF-A). Importantly, high VEGFA expression is associated with a worse overall survival of cHL patients. These findings strongly support the concept that WNTs might function as a regulator of lymphoma dissemination by affecting cHL cell chemotaxis and promoting EC behavior and thus angiogenesis through paracrine interactions.


Asunto(s)
Comunicación Celular , Células Endoteliales/metabolismo , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/patología , Microambiente Tumoral , Vía de Señalización Wnt , Adhesión Celular/genética , Línea Celular , Movimiento Celular/genética , Quimiocina CCL19/metabolismo , Quimiotaxis/genética , Quimiotaxis/inmunología , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/inmunología , Humanos , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Neovascularización Patológica , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
FASEB J ; 19(14): 2035-6, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16215210

RESUMEN

Pericyte loss and capillary regression are characteristic for incipient diabetic retinopathy. Pericyte recruitment is involved in vessel maturation, and ligand-receptor systems contributing to pericyte recruitment are survival factors for endothelial cells in pericyte-free in vitro systems. We studied pericyte recruitment in relation to the susceptibility toward hyperoxia-induced vascular remodeling using the pericyte reporter X-LacZ mouse and the mouse model of retinopathy of prematurity (ROP). Pericytes were found in close proximity to vessels, both during formation of the superficial and the deep capillary layers. When exposure of mice to the ROP was delayed by 24 h, i.e., after the deep retinal layer had formed [at postnatal (p) day 8], preretinal neovascularizations were substantially diminished at p18. Mice with a delayed ROP exposure had 50% reduced avascular zones. Formation of the deep capillary layers at p8 was associated with a combined up-regulation of angiopoietin-1 and PDGF-B, while VEGF was almost unchanged during the transition from a susceptible to a resistant capillary network. Inhibition of Tie-2 function either by soluble Tie-2 or by a sulindac analog, an inhibitor of Tie-2 phosphorylation, resensitized retinal vessels to neovascularizations due to a reduction of the deep capillary network. Inhibition of Tie-2 function had no effect on pericyte recruitment. Our data indicate that the final maturation of the retinal vasculature and its resistance to regressive signals such as hyperoxia depend on the completion of the multilayer structure, in particular the deep capillary layers, and are independent of the coverage by pericytes.


Asunto(s)
Capilares/metabolismo , Endotelio Vascular/citología , Retina/citología , Angiopoyetina 1/biosíntesis , Animales , Capilares/citología , Supervivencia Celular , Densitometría , Retinopatía Diabética/patología , Genes Reporteros , Hipoxia , Immunoblotting , Operón Lac , Ligandos , Ratones , Neovascularización Patológica , Pericitos/citología , Pericitos/metabolismo , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor TIE-2/metabolismo , Retina/embriología , Vasos Retinianos/patología , Factores de Tiempo , Regulación hacia Arriba
7.
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
8.
Cancer Res ; 52(17): 4821-3, 1992 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-1511446

RESUMEN

Platelet-derived growth factor and phorbol ester cause an increase in vascular endothelial growth factor (VEGF) mRNA expression in control NIH 3T3 fibroblasts and NIH 3T3 fibroblasts overexpressing human protein kinase C (PKC) alpha. In the case of phorbol ester-induced VEGF expression, the VEGF mRNA levels were significantly higher in cells overexpressing human PKC alpha as compared to control cells. In cells stimulated with platelet-derived growth factor or phorbol ester, induction of expression was lost after down-regulation of PKC. This indicates that PKC is involved in the signal transduction leading to VEGF expression.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Linfocinas/genética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Quinasa C/fisiología , Células 3T3 , Animales , Expresión Génica/efectos de los fármacos , Ratones , ARN Mensajero/genética , Acetato de Tetradecanoilforbol/farmacología , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Transfección , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
9.
Cancer Res ; 59(16): 4129-35, 1999 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10463618

RESUMEN

The goal of this investigation was to measure changes in vascular permeability, pore cutoff size, and number of transvascular transport pathways as a function of time and in response to vascular endothelial growth factor (VEGF), placenta growth factor (PIGF-1 and PIGF-2), or basic fibroblast growth factor (bFGF). Two human and two murine tumors were implanted in the dorsal skin chamber or cranial window. Vascular permeability to BSA (approximately 7 nm in diameter) and extravasation of polyethylene glycol-stabilized long-circulating liposomes (100-400 nm) and latex microspheres (approximately 800 nm) were determined by intravital microscopy. Vascular permeability was found to be temporally heterogeneous. VEGF superfusion (100 ng/ml) significantly increased vascular permeability to albumin in normal s.c. vessels, whereas a 30-fold higher dose of VEGF (3000 ng/ml) was required to increase permeability in pial vessels, suggesting that different tissues exhibit different dose thresholds for VEGF activity. Furthermore, VEGF superfusion (1000 ng/ml) increased vascular permeability to albumin in a hypopermeable human glioma xenograft in cranial window, whereas VEGF superfusion (10-1000 ng/ml) failed to increase permeability in a variety of hyperpermeable tumors grown in dorsal skin chamber. Interestingly, low-dose VEGF treatment (10 ng/ml) doubled the maximum pore size (from 400 to 800 nm) and significantly increased the frequency of large (400 nm) pores in human colon carcinoma xenografts. PIGF-1, PIGF-2, or bFGF did not show any significant effect on permeability or pore size in tumors. These findings suggest that exogenous VEGF may be useful for augmenting the transvascular delivery of larger antineoplastic agents such as gene targeting vectors and encapsulated drug carriers (typical range, 100-300 nm) into tumors.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Factores de Crecimiento Endotelial/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Linfocinas/farmacología , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica , Proteínas Gestacionales/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Humanos , Liposomas , Sustancias Macromoleculares , Ratones , Microesferas , Factor de Crecimiento Placentario , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
10.
Oncogene ; 9(3): 963-9, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8108142

RESUMEN

Many tumor cells produce vascular endothelial growth factor (VEGF), which is thought to be a pivotal mediator of tumor neoangiogenesis. Expression of the VEGF gene can be induced by tumor promoting phorbol esters, such as 12-O-tetradecanoylphorbol-13-acetate (TPA), which activate protein kinase C (PKC). Here we show that in transient transfection assays a mutated form of the murine p53 tumor suppressor gene (ala135-->val) induces expression of VEGF mRNA and potentiates TPA stimulated VEGF mRNA expression. In NIH 3T3 cells which stably overexpress the temperature sensitive p53 (ala135-->val), displaying mutant phenotype at 37 degrees C and wildtype phenotype at 32.5 degrees C, induction of VEGF mRNA and protein by activated PKC is strongly synergistic with mutant, but not wildtype p53. Mutant p53 specifically increases TPA induction of VEGF without affecting the expression of other TPA inducible genes. TPA dependent VEGF expression is also enhanced by human p53 mutated at amino acid 175. Thus, our data link PKC and p53, the gene most frequently altered in human tumors, with the regulation of tumor angiogenesis.


Asunto(s)
Factores de Crecimiento Endotelial/biosíntesis , Genes p53 , Linfocinas/biosíntesis , Mutación , Proteína Quinasa C/metabolismo , Células 3T3 , Animales , Factores de Crecimiento Endotelial/genética , Activación Enzimática , Humanos , Linfocinas/genética , Ratones , ARN Mensajero/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
11.
Circulation ; 100(6): 583-6, 1999 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-10441093

RESUMEN

BACKGROUND: Endothelial receptor tyrosine kinases include 3 members of the vascular endothelial growth factor receptor (VEGFR) family and 2 members of the angiopoietin receptor (Tie) family. In addition, the VEGF(165) isoform binds to neuropilin-1 (NP-1), a receptor for collapsins/semaphorins. The importance of these receptors for vasculogenesis and angiogenesis has been shown in gene-targeted mice, but so far, little is known about their exact expression patterns in the human vasculature. METHODS AND RESULTS: Frozen sections of human fetal heart were stained immunohistochemically with receptor-specific monoclonal (VEGFR, Tie) or polyclonal (NP-1) antibodies. The following patterns were observed: The endocardium was positive for VEGFR-1, VEGFR-2, NP-1, Tie-1, and Tie-2 but negative for VEGFR-3. The coronary vessels were positive for Tie-1, Tie-2, VEGFR-1, and NP-1 and negative for VEGFR-2 and VEGFR-3. Myocardial capillaries and epicardial blood vessels stained for VEGFR-1, VEGFR-2, NP-1, and Tie-1; myocardial capillaries and epicardial veins weakly for Tie-2; and epicardial lymphatic vessels for VEGFR-2 and VEGFR-3, weakly for Tie-1 and Tie-2, but not for VEGFR-1 or NP-1. CONCLUSIONS: The results demonstrate differential expression of the endothelial growth factor receptors in distinct types of vessels in the human heart. This information is useful for the understanding of their roles in physiological and pathological processes and for their diagnostic and therapeutic application in cardiovascular medicine.


Asunto(s)
Corazón Fetal/química , Proteínas Fetales/análisis , Proteínas Musculares/análisis , Proteínas del Tejido Nervioso/análisis , Proteínas Proto-Oncogénicas/análisis , Proteínas Tirosina Quinasas Receptoras/análisis , Receptores de Superficie Celular/análisis , Receptores de Factores de Crecimiento/análisis , Capilares/química , Capilares/embriología , Circulación Coronaria , Endocardio/química , Factores de Crecimiento Endotelial/fisiología , Secciones por Congelación , Humanos , Linfocinas/fisiología , Miocardio/química , Neovascularización Fisiológica/fisiología , Neuropilina-1 , Pericardio/química , Pericardio/embriología , Receptor TIE-1 , Receptor TIE-2 , Receptores TIE , Receptores de Factores de Crecimiento Endotelial Vascular , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Receptor 3 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
12.
Eur J Cell Biol ; 63(1): 84-95, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8005109

RESUMEN

The cortex corticis of the neonatal rabbit kidney consists of developing nephrons, vessels, collecting duct ampullae and the nephrogenic mesenchyme. Inductive interactions between embryonic mesenchyme and collecting duct ampullae lead to the coordinated development of the nephrons and the collecting duct system. The factors regulating nephrogenesis and vascular development within this tissue region are unknown. In order to analyze the hormonal regulation of vascular development an organotypic culture system was established. Cortex explants from neonatal rabbit kidneys were prepared, mounted in a set of holding rings and cultured under serum-free conditions for 14 days in conventional culture plates or under permanent medium perfusion in a newly developed culture container. The detection of endothelial cells was carried out by means of two monoclonal antibodies. Within the renal cortex corticis EnPo 1 detected developing vasculature as well as podocytes and a subset of mesenchymal cells. EC1 displayed exclusive specificity for endothelial cells. The antibody did not discriminate between arteries and veins. Endothelial cells of different developmental stages were labeled with the same intensity. A combination of both antibodies allowed the discrimination between developing endothelial cells and podocytes. Following 14 days of culture under permanent medium exchange, excellent tissue preservation as well as endothelial cell proliferation was observed in cortex explants. In contrast, tissue kept in stationary culture revealed a high degree of disintegration. Endothelial antigen expression was also severely disturbed. Tissue maintenance under stationary conditions was improved by the application of a hormone mixture consisting of aldosterone and 1,25-hydroxyvitamin D3. However, the high degree of spatial organization shown by developing endothelial cells in vivo was maintained exclusively in explants cultured in the presence of hormone under permanent perfusion.


Asunto(s)
Corteza Renal/irrigación sanguínea , Corteza Renal/citología , Aldosterona/farmacología , Animales , Anticuerpos Monoclonales , Células Cultivadas , Colecalciferol/farmacología , Medio de Cultivo Libre de Suero/farmacología , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Linfocinas/farmacología , Microcirculación , Perfusión , Conejos , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
J Clin Endocrinol Metab ; 77(6): 1723-5, 1993 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8263163

RESUMEN

Vascularization is a prerequisit for corpus luteum formation. Angiogenesis is supposed to be regulated by vascular growth factors. Vascular endothelial growth factor (VEGF) induces specifically endothelial cell proliferation as well as angiogenesis and increases capillary permeability. With this study we demonstrate for the first time the presence of VEGF-mRNA in human luteinized granulosa cells by Northern blot technique. Granulosa cells were obtained from 11 individual patients undergoing ovarian stimulation for in vitro fertilization procedure. Two transcripts of VEGF-mRNA at 3.9Kb and at 4.3Kb could be detected in each case. Specific transcripts were expressed in different amounts. These results indicate that VEGF may promote at least in part vascularization of the human corpus luteum.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Células de la Granulosa/metabolismo , Linfocinas/genética , ARN Mensajero/análisis , Células Cultivadas , Cuerpo Lúteo/irrigación sanguínea , Femenino , Fertilización In Vitro , Expresión Génica/efectos de los fármacos , Humanos , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
14.
J Clin Endocrinol Metab ; 80(6): 1967-71, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7775647

RESUMEN

Ovarian hyperstimulation syndrome (OHSS) is a severe complication arising from controlled ovarian stimulation treatment. This iatrogenic condition is potentially lethal and occurs in 0.3-5% of stimulated ovarian cycles. hCG exacerbates OHSS. The pathophysiology of OHSS is still unknown; therefore, treatment regimens are aimed at ameliorating symptoms. Prominent features of OHSS are an elevated risk of thromboembolism due to enhanced production of von Willebrand factor by endothelial cells and ascites, or pulmonary edema due to increased vascular permeability followed by third space fluid accumulation. Both of these sequelae can be evoked by vascular endothelial growth factor (VEGF), also known as vascular permeability factor (VPF). High concentrations of VEGF/VPF have been demonstrated in ascitic fluid from patients with OHSS, but the source of VEGF/VPF in these patients remained unidentified. Here we report that the messenger ribonucleic acid expression of VEGF/VPF in human luteinized granulosa cells (GCs) is dose and time dependently enhanced by hCG in vitro. Furthermore, VEGF/VPF proteins are produced by GCs. Our results suggest that the effects of hCG on the development and course of OHSS may be mediated by the production of VEGF/VPF by GCs.


Asunto(s)
Gonadotropina Coriónica/farmacología , Factores de Crecimiento Endotelial/genética , Expresión Génica/efectos de los fármacos , Células de la Granulosa/metabolismo , Linfocinas/genética , Síndrome de Hiperestimulación Ovárica/metabolismo , Adulto , Células Cultivadas , Gonadotropina Coriónica/efectos adversos , Femenino , Fertilización In Vitro , Humanos , Cinética , Síndrome de Hiperestimulación Ovárica/inducido químicamente , ARN Mensajero/metabolismo , Receptores de HFE/metabolismo , Receptores de HL/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
15.
FEBS Lett ; 223(2): 243-6, 1987 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-3666150

RESUMEN

A cDNA clone of about 2300 base pairs was prepared from the human osteosarcoma cell line U-2 OS by hybridization with a 22-mer oligonucleotide complementary to the NH2-terminus of PDGF-A. Restriction and sequence analysis showed that this clone contains the entire coding region for PDGF-A and a long 3'-untranslated region which is only distantly related to that in the mRNA of PDGF-B.


Asunto(s)
Factor de Crecimiento Derivado de Plaquetas/genética , Secuencia de Aminoácidos , Secuencia de Bases , Clonación Molecular , Humanos , Datos de Secuencia Molecular , Peso Molecular , Biosíntesis de Proteínas , ARN Mensajero/genética , Células Tumorales Cultivadas
16.
FEBS Lett ; 213(1): 89-94, 1987 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-3470198

RESUMEN

It was shown previously [(1984) EMBO J. 3, 453-459] that after treatment of the human erythroleukemia cell line HEL with phorbol ester and dimethyl sulfoxide there was a marked increase in the amounts of megakaryocytic markers, especially of platelet alpha-granule proteins and platelet glycoproteins. In order to investigate this differentiation process further we have studied the expression of the mRNA encoding PDGF-A and PDGF-B (c-sis). Upon addition of the phorbol ester to the culture medium the expression of the c-sis transcript was enhanced about 7-fold over a period of 4 days. With dimethyl sulfoxide there was no significant stimulation of the expression. Addition of cycloheximide to HEL cells treated for a short period with phorbol ester superinduced the expression of the c-sis gene. The HEL cells did not express the A-chain mRNA even in the presence of phorbol ester or dimethyl sulfoxide. This leads us to propose that synthesis of the PDGF-A chain and PDGF-B chain is differentially regulated in the megakaryocytic-like HEL cell line.


Asunto(s)
Leucemia Eritroblástica Aguda/metabolismo , Ésteres del Forbol/farmacología , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Diferenciación Celular , Línea Celular , Dimetilsulfóxido/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Factor de Crecimiento Derivado de Plaquetas/genética , ARN Mensajero/metabolismo
17.
FEBS Lett ; 198(2): 344-8, 1986 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-3456904

RESUMEN

A cDNA clone of about 2500 base pairs was prepared from the human osteosarcoma cell line U-2 OS by hybridizing with a v-sis probe. Sequence analysis showed that this cDNA contains the coding region for the PDGF-B chain. It is discussed that the mitogen secreted by these osteosarcoma cells contains the PDGF-B chain and is probably a homodimer of two B-chains.


Asunto(s)
Osteosarcoma/análisis , Factor de Crecimiento Derivado de Plaquetas/genética , ARN Mensajero/análisis , Secuencia de Bases , Línea Celular , ADN/análisis , Humanos , Oncogenes
18.
Angiogenesis ; 1(1): 117-30, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-14517398

RESUMEN

The aim of this study was to characterize the capillary density, progression and persistence of new capillaries induced by different isoforms of vascular endothelial growth factor (VEGF)-A. They were produced and purified using the same protocol and assessed in the same experimental model, the rabbit cornea assay. Monogenic homodimers for VEGF121 and VEGF165 together with the heterodimer VEGF121/165 were tested as slow release polymer pellets implanted into the avascular rabbit cornea and examined up to 18 days post-implantation. The implants consistently stimulated angiogenesis in the absence of inflammation. The VEGF121 isoform produced the strongest increase of new capillary vessels which rapidly and persistently progressed into the corneal stroma. VEGF165 promoted the growth of a smaller number of capillaries which ten-ded to regress over time. Heterodimers of VEGF121/165 produced intermediate in vivo activities between the two homodimers. In vitro endothelial cell proliferation, mobilization and adhesion were promoted by all VEGF isoforms under serum-free or serum-reduced conditions with the same order of potency. Anti-soluble KDR (sKDR) antibody completely inhibited the effects of all the isoforms. These results indicate that monogenic homodimer preparations of VEGF121 and VEGF165 can display distinct biological effects which are functionally retained after the heterodimeric assembly of VEGF121 and VEGF165. The observed different biological behavior of the VEGF isoforms reveals the possibility that in vivo the assembly of dimers derived from splicing of a single gene may yield molecules with either different matrix or receptor interaction, stability or diffusion rate according to specific needs.

19.
J Interferon Cytokine Res ; 20(5): 511-7, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10841080

RESUMEN

Vascular endothelial growth factor (VEGF) plays an important role in the production of ascitic fluid associated with malignant tumor growth. In an experimental model for malignant ascites formation, mice were inoculated intraperitoneally with syngeneic mouse sarcoma tumor cells. Ascites development was not prevented by administering tumor necrosis factor (TNF) simultaneously with the tumor cell inoculation. When the malignant ascites was first drained and renewal of ascites was monitored, however, a TNF dose-dependent inhibition of ascitic fluid accumulation was observed. Northern blot analyses indicated transient downregulation by TNF on the expression of VEGF mRNA in tumor cells. Monoclonal antibody, (mAb) DC101 generated against the mouse VEGF receptor Flk-1 prevented the recurrence of malignant ascites in mice similar to TNF inhibition. In addition, exogenous soluble human Flt-1 used as an inhibitor of endogenous VEGF binding also inhibited ascites recurrence. These data demonstrate that the observed inhibitory effect of TNF on reestablishment of malignant ascites can be achieved equally by inhibition of the interaction of VEGF with its receptor Flk-1.


Asunto(s)
Ascitis/etiología , Ascitis/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Linfocinas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Ascitis/prevención & control , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Factores de Crecimiento Endotelial/genética , Femenino , Humanos , Linfocinas/antagonistas & inhibidores , Linfocinas/genética , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular , Recurrencia , Sarcoma Experimental/genética , Sarcoma Experimental/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
J Immunol Methods ; 226(1-2): 169-77, 1999 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-10410982

RESUMEN

Vascular endothelial growth factor (VEGF) is an important factor for endothelial cell proliferation and a key regulator of blood vessel development in embryos and angiogenesis in adult tissues. Its biological activity is mediated by two receptor tyrosine kinases, VEGFR-1 (Flt-1) and VEGFR-2 (KDR). In contrast to VEGFR-2, a naturally occurring soluble form of the VEGFR-1 (sVEGFR-1) is produced by endothelial cells by differential splicing of the flt-1 gene, and it is a secreted gene product. In order to develop a specific enzyme-linked immunosorbent assay (ELISA) for the measurement of sVEGFR-1, we established five anti-human receptor antibodies and characterized them in detail. These antibodies recognize different epitopes located within the seven Ig-like domains of the extracellular receptor protein but have no neutralizing activity in ligand binding assays. Together with a polyclonal antiserum, a specific human sVEGFR-1 ELISA was developed using the mAb #190.11. The ELISA can detect human sVEGFR-1 with a minimum detection limit of 1 ng/ml. The ELISA does not show any cross-reactivity with other related soluble receptors. Using this assay, human sVEGFR-1 was measured in the supernatant of different VEGFR-1 expressing cell types. No sVEGFR-1 protein was detectable after heparin Sepharose treatment or size-exclusion filtration (< 30 kDa). The ELISA assay for sVEGFR-1 was also used to measure the amount of the soluble receptor in amniotic fluid samples of patients undergoing amniocentesis during the course of normal pregnancies. The concentration of the samples was in the range of 5-35 ng/ml. This ELISA could be useful powerful tool for investigations concerning the physiological function of the soluble receptor under normal and pathophysiological conditions.Furthermore, it may facilitate studies of the mechanisms of receptor production.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Proteínas Proto-Oncogénicas/análisis , Proteínas Tirosina Quinasas Receptoras/análisis , Animales , Anticuerpos Monoclonales/inmunología , Células Cultivadas , Reacciones Cruzadas , Medios de Cultivo , Endotelio Vascular/citología , Humanos , Ligandos , Ratones , Conejos , Solubilidad , Receptor 1 de Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA