Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 11(12): 4295-308, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11102524

RESUMO

The latent transforming growth factor-beta-binding protein-1 (LTBP-1) belongs to a family of extracellular glycoproteins that includes three additional isoforms (LTBP-2, -3, and -4) and the matrix proteins fibrillin-1 and -2. Originally described as a TGF-beta-masking protein, LTBP-1 is involved both in the sequestration of latent TGF-beta in the extracellular matrix and the regulation of its activation in the extracellular environment. Whereas the expression of LTBP-1 has been analyzed in normal and malignant cells and rodent and human tissues, little is known about LTBP-1 in embryonic development. To address this question, we used murine embryonic stem (ES) cells to analyze the appearance and role of LTBP-1 during ES cell differentiation. In vitro, ES cells aggregate to form embryoid bodies (EBs), which differentiate into multiple cell lineages. We analyzed LTBP-1 gene expression and LTBP-1 fiber appearance with respect to the emergence and distribution of cell types in differentiating EBs. LTBP-1 expression increased during the first 12 d in culture, appeared to remain constant between d 12 and 24, and declined thereafter. By immunostaining, fibrillar LTBP-1 was observed in those regions of the culture containing endothelial, smooth muscle, and epithelial cells. We found that inclusion of a polyclonal antibody to LTBP-1 during EB differentiation suppressed the expression of the endothelial specific genes ICAM-2 and von Willebrand factor and delayed the organization of differentiated endothelial cells into cord-like structures within the growing EBs. The same effect was observed when cultures were treated with either antibodies to TGF-beta or the latency associated peptide, which neutralize TGF-beta. Conversely, the organization of endothelial cells was enhanced by incubation with TGF-beta 1. These results suggest that during differentiation of ES cells LTBP-1 facilitates endothelial cell organization via a TGF-beta-dependent mechanism.


Assuntos
Proteínas de Transporte/fisiologia , Endotélio/embriologia , Peptídeos e Proteínas de Sinalização Intracelular , Células-Tronco/citologia , Animais , Anticorpos/imunologia , Biomarcadores , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Diferenciação Celular , Células Cultivadas , Endotélio/citologia , Endotélio/metabolismo , Matriz Extracelular/metabolismo , Expressão Gênica , Proteínas de Ligação a TGF-beta Latente , Camundongos , Células-Tronco/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/fisiologia
2.
J Cell Sci ; 112 ( Pt 23): 4213-21, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10564640

RESUMO

In vitro experimental evidences suggest that the proteolytic degradation of the extracellular matrix (ECM) by activation of the urokinase-type plasminogen activator (uPA)/plasmin system may affect growth factor activity and bioavailability. However, no direct in vivo observations were available to support this hypothesis. Here we demonstrate that endothelial GM 7373 cells overexpressing human uPA (uPA-R5 cells) cause the release of (125)I-labeled fibroblast growth factor-2 (FGF2) from endothelial ECM in a plasmin-dependent manner. Accordingly, uPA-R5 cells are angiogenic in vivo when applied on the top of the chorioallantoic membrane (CAM) of the chick embryo. In contrast, mock-transfected Neo2 cells are unable to release ECM-bound (125)I-FGF2 and are poorly angiogenic. Neovascularization elicited by uPA-R5 cells is significantly reduced by neutralizing anti-FGF2 antibodies to values similar to those observed in Neo2 cell-treated CAMs. Accordingly, purified human uPA stimulates neovascularization of the CAM in the absence of an inflammatory response. The angiogenic activity of uPA is significantly inhibited by neutralizing anti-FGF2 antibodies or by pretreatment with phenylmethylsulfonyl fluoride. The non-catalytic, receptor-binding amino-terminal fragment of uPA is instead non angiogenic. Taken together, the data indicate that uPA is able to induce angiogenesis in vivo via a plasmin-dependent degradation of ECM that causes the mobilization of stored endogenous FGF2.


Assuntos
Endotélio Vascular/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Neovascularização Fisiológica/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Alantoide/irrigação sanguínea , Animais , Moléculas de Adesão Celular/farmacologia , Linhagem Celular , Embrião de Galinha , Córion/irrigação sanguínea , Endotélio Vascular/citologia , Matriz Extracelular/fisiologia , Fibroblastos/fisiologia , Humanos , Cinética , Neovascularização Fisiológica/efeitos dos fármacos , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/genética
3.
J Cell Physiol ; 181(2): 273-84, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10497306

RESUMO

The single-copy fibroblast growth factor 2 (FGF-2) gene encodes four coexpressed isoforms of different molecular masses. The 18-kDa FGF-2 is primarily localized in the cytoplasm, whereas the higher molecular mass isoforms (HMW FGF-2) localize to the nucleus and nucleolus. The overexpression of either 18-kDa FGF-2 or HMW FGF-2 promotes cell transformation in a dose-dependent manner. In NIH 3T3 cells, the selective overexpression of HMW FGF-2 but not of 18-kDa FGF-2 confers upon the cells the unique phenotype of growth in low serum-containing medium. Thus, the distinct intracellular localization and the level of expression of FGF-2 are pivotal requirements for the differential effects of FGF-2 isoforms on the cellular phenotype. On this basis, we established a doxycycline-regulatable FGF-2 expression system that permitted us to regulate the expression of each isoform in a time- and dose-dependent manner. We analyzed the growth properties of cells in the presence and absence of doxycycline in both normal and low serum-containing medium and in soft agar. The doxycycline-activated expression of 18-kDa FGF-2 did not allow growth in low serum medium. The growth of cells expressing HMW FGF-2 was increased by doxycycline under all three conditions, and a relationship between the level of HMW FGF-2 expression and cell growth was observed for all three conditions. This doxycycline-regulatable FGF-2 expression system provides a mechanism to analyze changes in FGF-2 targeted pathways and genes and to characterize pathways specifically activated by either the 18-kDa FGF-2 or the HMW FGF-2 isoforms.


Assuntos
Doxiciclina/farmacologia , Fator 2 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/fisiologia , Células 3T3 , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Células Clonais , Relação Dose-Resposta a Droga , Fator 2 de Crescimento de Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Camundongos , Isoformas de Proteínas/genética , Transfecção
4.
Mol Biol Cell ; 10(5): 1429-44, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10233154

RESUMO

Human basic fibroblast growth factor (FGF-2) occurs in four isoforms: a low molecular weight (LMW FGF-2, 18 kDa) and three high molecular weight (HMW FGF-2, 22, 22.5, and 24 kDa) forms. LMW FGF-2 is primarily cytoplasmic and functions in an autocrine manner, whereas HMW FGF-2s are nuclear and exert activities through an intracrine, perhaps nuclear, pathway. Selective overexpression of HMW FGF-2 forms in fibroblasts promotes growth in low serum, whereas overexpression of LMW FGF-2 does not. The HMW FGF-2 forms have two functional domains: an amino-terminal extension and a common 18-kDa amino acid sequence. To investigate the role of these regions in the intracrine signaling of HMW FGF-2, we produced stable transfectants of NIH 3T3 fibroblasts overexpressing either individual HMW FGF-2 forms or artificially nuclear-targeted LMW FGF-2. All of these forms of FGF-2 localize to the nucleus/nucleolus and induce growth in low serum. The nuclear forms of FGF-2 trigger a mitogenic stimulus under serum starvation conditions and do not specifically protect the cells from apoptosis. These data indicate the existence of a specific role for nuclear FGF-2 and suggest that LMW FGF-2 represents the biological messenger in both the autocrine/paracrine and intracrine FGF-2 pathways.


Assuntos
Núcleo Celular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Células 3T3/efeitos dos fármacos , Células 3T3/metabolismo , Sequência de Aminoácidos , Animais , Apoptose , Bromodesoxiuridina/metabolismo , Divisão Celular/fisiologia , Clonagem Molecular , Ensaio de Unidades Formadoras de Colônias , DNA/biossíntese , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Camundongos , Dados de Sequência Molecular , Peso Molecular , Fenótipo , Isoformas de Proteínas
5.
J Pathol ; 189(4): 590-9, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10629563

RESUMO

A close relationship exists between angiogenesis and the formation of vascular lesions. The development of the vascular system in the chick embryo chorioallantoic membrane (CAM) may thus represent a model to study the effects of the deregulation of endothelial cell behaviour. Alterations of the developing vascular tree of the CAM were observed after exposure to murine aortic endothelial (MAE) cells overexpressing human fibroblast growth factor-2 (FGF2) cDNA (pZipFGF2 MAE cells), or to their conditioned medium (CM). pZipFGF2 MAE cells injected into the allantoic sac or applied on to the CAM of day 8-9 chick embryos induce neovascularization and the appearance of haemangioma-like lesions. This activity was not prevented by anti-FGF2 antibodies. The CM from pZipFGF2 MAE cells was also active when adsorbed into a gelatin sponge and applied on to the CAM, both in the absence and in the presence of anti-FGF2 antibodies. No effects on vessel development were exerted by parental MAE cells, FGF2-transfected NIH 3T3 fibroblasts, or their conditioned media. In vitro, pZipFGF2 MAE cell CM caused parental MAE cells to invade fibrin gels and to undergo morphogenesis on Matrigel. This activity was not mimicked by recombinant FGF2 nor affected by anti-FGF2 antibodies, and depended on a M (r) approximately 45 000 heat-labile heparin-binding factor. Size exclusion chromatography of pZipFGF2 MAE cell CM demonstrated that the in vitro activity co-purified with an in vivo angiogenic capacity. Thus, FGF2 overexpression in mouse endothelial cells induces the production of an angiogenic activity distinct from FGF2, which may contribute to the genesis of angioproliferative lesions.


Assuntos
Alantoide/irrigação sanguínea , Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Neovascularização Fisiológica , Alantoide/efeitos dos fármacos , Alantoide/ultraestrutura , Animais , Células Cultivadas , Embrião de Galinha , Cromatografia de Afinidade , Cromatografia em Gel , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hemangioma/etiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica
6.
Biochem Biophys Res Commun ; 252(2): 524-8, 1998 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-9826564

RESUMO

By using the yeast two-hybrid system, we identified the ribosomal protein L6/TAXREB107 as an intracellular partner for FGF-2. L6/TAXREB107 also mediates the DNA binding of the HTLV-1 transactivator Tax. In vitro binding experiments indicated that both the high-molecular-weight forms (HMW) and the 18-kDa form of FGF-2 bind to L6/TAXREB107. Deletion analysis suggested that L6/TAXREB107 has two binding sites for HMW FGF-2 and one binding site for 18-kDa FGF-2, implying that the unique N-terminal extension of the HMW FGF-2 is one of the binding domains for L6/TAXREB107. Transfection assays showed that high expression of either HMW or 18 kDa FGF-2 stimulates Tax-mediated transactivation in NIH 3T3 cells. This result suggests a possible role of FGF-2 in Tax-mediated HTLV-1 transformation as well as FGF-2 binding to ribosomes and/or their precursors.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Produtos do Gene tax/metabolismo , Proteínas Ribossômicas/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Primers do DNA/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/genética , Produtos do Gene tax/química , Produtos do Gene tax/genética , Técnicas In Vitro , Camundongos , Peso Molecular , Reação em Cadeia da Polimerase , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Deleção de Sequência , Ativação Transcricional , Transfecção
7.
J Cell Biol ; 141(7): 1659-73, 1998 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-9647657

RESUMO

FGF-2 and VEGF are potent angiogenesis inducers in vivo and in vitro. Here we show that FGF-2 induces VEGF expression in vascular endothelial cells through autocrine and paracrine mechanisms. Addition of recombinant FGF-2 to cultured endothelial cells or upregulation of endogenous FGF-2 results in increased VEGF expression. Neutralizing monoclonal antibody to VEGF inhibits FGF-2-induced endothelial cell proliferation. Endogenous 18-kD FGF-2 production upregulates VEGF expression through extracellular interaction with cell membrane receptors; high-Mr FGF-2 (22-24-kD) acts via intracellular mechanism(s). During angiogenesis induced by FGF-2 in the mouse cornea, the endothelial cells of forming capillaries express VEGF mRNA and protein. Systemic administration of neutralizing VEGF antibody dramatically reduces FGF-2-induced angiogenesis. Because occasional fibroblasts or other cell types present in the corneal stroma show no significant expression of VEGF mRNA, these findings demonstrate that endothelial cell-derived VEGF is an important autocrine mediator of FGF-2-induced angiogenesis. Thus, angiogenesis in vivo can be modulated by a novel mechanism that involves the autocrine action of vascular endothelial cell-derived FGF-2 and VEGF.


Assuntos
Comunicação Autócrina/fisiologia , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/fisiologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Linfocinas/metabolismo , Neovascularização Fisiológica/fisiologia , Células 3T3 , Animais , Capilares/fisiologia , Bovinos , Divisão Celular , Células Cultivadas , Endotélio Vascular/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos , Comunicação Parácrina/fisiologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
8.
Mol Biol Cell ; 8(12): 2449-61, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9398667

RESUMO

Fibroblast growth factor-2 (FGF-2) immobilized on non-tissue culture plastic promotes adhesion and spreading of bovine and human endothelial cells that are inhibited by anti-FGF-2 antibody. Heat-inactivated FGF-2 retains its cell-adhesive activity despite its incapacity to bind to tyrosine-kinase FGF receptors or to cell-surface heparan sulfate proteoglycans. Recombinant glutathione-S-transferase-FGF-2 chimeras and synthetic FGF-2 fragments identify two cell-adhesive domains in FGF-2 corresponding to amino acid sequences 38-61 and 82-101. Both regions are distinct from the FGF-receptor-binding domain of FGF-2 and contain a DGR sequence that is the inverse of the RGD cell-recognition sequence. Calcium deprivation, RGD-containing eptapeptides, soluble vitronectin (VN), but not fibronectin (FN), inhibit cell adhesion to FGF-2. Conversely, soluble FGF-2 prevents cell adhesion to VN but not FN, thus implicating VN receptor in the cell-adhesive activity of FGF-2. Accordingly, monoclonal and polyclonal anti-alphavbeta3 antibodies prevent cell adhesion to FGF-2. Also, purified human alphavbeta3 binds to immobilized FGF-2 in a cation-dependent manner, and this interaction is competed by soluble VN but not by soluble FN. Finally, anti-alphavbeta3 monoclonal and polyclonal antibodies specifically inhibit mitogenesis and urokinase-type plasminogen activator (uPA) up-regulation induced by free FGF-2 in endothelial cells adherent to tissue culture plastic. These data demonstrate that FGF-2 interacts with alphavbeta3 integrin and that this interaction mediates the capacity of the angiogenic growth factor to induce cell adhesion, mitogenesis, and uPA up-regulation in endothelial cells.


Assuntos
Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Receptores de Vitronectina/metabolismo , Motivos de Aminoácidos , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Cálcio/farmacologia , Bovinos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , Endotélio Vascular/metabolismo , Endotélio Vascular/ultraestrutura , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/química , Fibronectinas/metabolismo , Fibronectinas/farmacologia , Heparina/farmacologia , Humanos , Mitógenos/antagonistas & inibidores , Mitógenos/química , Mitógenos/metabolismo , Mitógenos/farmacologia , Oligopeptídeos/farmacologia , Plásticos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Vitronectina/antagonistas & inibidores , Receptores de Vitronectina/imunologia , Receptores de Vitronectina/isolamento & purificação , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Deleção de Sequência/genética , Solubilidade , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Vitronectina/metabolismo , Vitronectina/farmacologia
9.
Microvasc Res ; 53(3): 254-60, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9211403

RESUMO

Fetal bovine aortic endothelial GM 7373 cells were transfected with a viral expression vector harboring the human urokinase-type plasminogen activator (uPA) gene. The stable transfectant clone uPA-R5 overexpressed and secreted human uPA as shown by Northern blot analysis, immunoprecipitation of metabolically labeled proteins, plasmin chromogenic assays, and SDS-PAGE zymography of cell extracts and conditioned media. The uPA-R5 cells were analyzed for their invasive capacity in vitro in the Matrigel chemoinvasion assay in the presence of serine- or metalloprotease inhibitors. uPA overexpression enhanced the invasive capacity of GM 7373 cells through a mechanism which differs from that mediated by metalloproteases. Endothelial cell uPA transfectants may represent an useful experimental model to investigate the role of uPA in angiogenesis and angioproliferative diseases.


Assuntos
Endotélio Vascular/enzimologia , Endotélio Vascular/fisiologia , Neovascularização Fisiológica , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Animais , Aorta , Bovinos , Linhagem Celular , Endotélio Vascular/efeitos dos fármacos , Feto , Humanos , Neovascularização Fisiológica/genética , Ativador de Plasminogênio Tipo Uroquinase/genética
10.
J Vasc Res ; 34(6): 455-63, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9425998

RESUMO

Several methods for the in vivo study of angiogenesis are available, and each angiogenic assay presents distinct advantages and disadvantages. In this study, we present a new method for the quantitation of angiogenesis and antiangiogenesis in the chick embryo chorioallantoic membrane (CAM), based on the implantation of gelatin sponges on the top of growing CAM, on day 8 of incubation. After implantation, the sponges were treated with a stimulator (recombinant human basic fibroblast growth factor, FGF2) or an inhibitor (a rabbit polyclonal anti-FGF2 antibody) of blood vessel formation. Blood vessels growing vertically into the sponge and at the boundary between sponge and surrounding CAM mesenchyme were counted by a morphometric method on day 12. In addition, to assess whether the gelatin sponge is an appropriate vehicle to deliver cultured cells and evaluate their angiogenic potential, mouse aortic endothelial cells were cotransfected with human FGF2 and the Escherichia coli beta-galactosidase (beta-GAL) reporter gene. Stable transfectants were absorbed by the sponge, and evaluation of the angiogenic response was paralleled by beta-GAL staining to visualize implanted cells. This technique may facilitate the discovery and development of agonists or antagonists of angiogenesis.


Assuntos
Alantoide/irrigação sanguínea , Córion/irrigação sanguínea , Neovascularização Fisiológica/fisiologia , Animais , Anticorpos/imunologia , Linhagem Celular Transformada/metabolismo , Embrião de Galinha , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Fator 2 de Crescimento de Fibroblastos/imunologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Gelatina , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Próteses e Implantes , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia
11.
J Neurosci ; 16(7): 2220-5, 1996 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8601802

RESUMO

Tissue plasminogen activator (tPA), a serine protease that converts inactive plasminogen to active plasmin, is produced in the rat and mouse hippocampus and participates in neuronal plasticity. To help define the role of tPA in the nervous system, we have analyzed the regulation of its expression in the neuronal cell line PC12. In control cultures, tPA activity is exclusively cell-associated, and no activity is measurable in the culture medium. When the cells are treated with depolarizing agents, such as KCI, tPA activity becomes detectable in the medium. The increased secreted tPA activity is not accompanied by an increase in tPA mRNA levels, and it is not blocked by protein synthesis inhibitors. In contrast, tPA release is abolished by Ca2+ channel blockers, suggesting that chemically induced membrane depolarization stimulates the secretion of preformed enzyme. Moreover, KCI has a similar effect in vivo when administered to the murine brain via an osmotic pump: tPA activity increases along the CA2-CA3 regions and dentate gyrus of the hippocampal formation. These results demonstrate a neuronal activity-dependent secretory mechanism that can rapidly increase the amount of tPA in neuronal tissue.


Assuntos
Cálcio/metabolismo , Potenciais da Membrana/fisiologia , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Northern Blotting , Bloqueadores dos Canais de Cálcio/farmacologia , Cricetinae , Expressão Gênica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/fisiologia , Plasticidade Neuronal/fisiologia , Células PC12/metabolismo , Células PC12/fisiologia , Cloreto de Potássio/farmacologia , RNA Mensageiro/análise , Ratos , Ativador de Plasminogênio Tecidual/genética
12.
Cell Growth Differ ; 7(2): 147-60, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8822198

RESUMO

Basic fibroblast growth factor (bFGF) is expressed in vascular endothelium during tumor neovascularization and angioproliferative diseases. The ultimate significance of this observation is poorly understood. We have investigated the biological consequences of endothelial cell activation by endogenous bFGF in a mouse aortic endothelial cell line stably transfected with a retroviral expression vector harboring a human bFGF cDNA. Selected clones expressing M(r) 24,000, M(r) 22,000, and/or M(r) 18,000 bFGF isoforms were characterized by a transformed morphology and an increased saturation density. bFGF transfectants showed invasive behavior and sprouting activity in three-dimensional fibrin gels and formed a complex network of branching cord-like structures connecting foci of infiltrating cells when seeded on laminin-rich basement membrane matrix (Matrigel). The invasive and morphogenetic behavior was prevented by anti-bFGF antibody, revealing the autocrine modality of the process. The biological consequences of this autocrine activation were investigated in vivo. bFGF-transfected cells gave rise to highly vascularized lesions resembling Kaposi's sarcoma when injected in nude mice and induced angiogenesis in avascular rabbit cornea. When injected into the allantoic sac of the chick embryo, they caused an increase in vascular density and formation of hemangiomas in the chorioallantoic membrane. In conclusion, bFGF-overexpressing endothelial cells acquired an angiogenic phenotype and recruit quiescent endothelium originating angioproliferative lesions in vivo. These findings demonstrate that bFGF overexpression exerts an autocrine role for endothelial cells and support the notion that tumor neovascularization and angioproliferative diseases can be triggered by stimuli that induce vascular endothelium to produce its own autocrine factor(s).


Assuntos
Endotélio Vascular/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Neovascularização Patológica/fisiopatologia , Células 3T3/citologia , Células 3T3/fisiologia , Animais , Aorta/citologia , Tamanho Celular/efeitos dos fármacos , Tamanho Celular/fisiologia , Transformação Celular Viral , Embrião de Galinha , Colágeno/farmacologia , DNA Complementar/genética , Combinação de Medicamentos , Endotélio Corneano/citologia , Endotélio Vascular/citologia , Endotélio Vascular/ultraestrutura , Matriz Extracelular , Fibrina/farmacologia , Humanos , Injeções Intravenosas , Laminina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Eletrônica , Óvulo/ultraestrutura , Proteoglicanas/farmacologia , Coelhos , Retroviridae/genética
14.
Int J Oncol ; 8(3): 567-73, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21544398

RESUMO

Basic fibroblast growth factor (bFGF) is expressed in the vascular endothelium of human brain tumors. To investigate the biological consequences of a possible autocrine modality of microvascular endothelial cell activation by endogenous bFGF in these tumors, mouse brain microvascular endothelial cells were stably transfected with a retroviral expression vector harboring a human bFGF cDNA. When grown on tissue culture plastic, bFGF-transfected clones show a transformed morphology and increased saturation density. bFGF-transfectants have an invasive behavior when seeded on three-dimensional fibrin gel and originate endothelial cell sprouts when embedded within fibrin. Also, bFGF-transfected cells undergo morphogenetic organization and produce a complex network of branching cord-like structures connecting foci of infiltrating cells when seeded on Matrigel, a laminin-rich extracellular matrix material. In contrast, parental and mock-transfected cells do not invade fibrin gels nor organize on Matrigel. These findings demonstrate that bFGF overexpression induces an angiogenic phenotype in brain microvascular endothelial cells characterized by an invasive behavior and morphogenic potential. They support the notion that neovascularization of brain tumors can be triggered by stimuli that induce vascular endothelium to produce its own autocrine factor(s).

15.
Cancer Res ; 55(20): 4729-38, 1995 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-7553656

RESUMO

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.


Assuntos
Adenocarcinoma Papilar/patologia , Neoplasias do Endométrio/patologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Neovascularização Patológica/patologia , Idoso , Animais , Divisão Celular , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Expressão Gênica , Humanos , Técnicas In Vitro , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/patologia , RNA Mensageiro/genética , Transfecção , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
16.
Nature ; 377(6547): 340-4, 1995 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-7566088

RESUMO

Neuronal degeneration in the hippocampus, a region of the brain important for acquisition of memory in humans, occurs in various pathological conditions, including Alzheimer's disease, brain ischaemia and epilepsy. When neuronal activity is stimulated in the adult rat and mouse hippocampus, tissue plasminogen activator (tPA), a serine protease that converts inactive plasminogen to the active protease plasmin, is transcriptionally induced. The activity of tPA in neural tissue is correlated with neurite outgrowth, regeneration and migration, suggesting that it might be involved in neuronal plasticity. Here we show that tPA is produced primarily by microglia in the hippocampus. Using excitotoxins to induce neuronal cell loss, we demonstrate that tPA-deficient mice are resistant to neuronal degeneration. These mice are also less susceptible to pharmacologically induced seizures than wild-type mice. These findings identify a role for tPA in neuronal degeneration and seizure.


Assuntos
Agonistas de Aminoácidos Excitatórios/farmacologia , Ácido Caínico/farmacologia , Degeneração Neural/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Resistência a Medicamentos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Convulsões/induzido quimicamente , Convulsões/metabolismo , Ativador de Plasminogênio Tecidual/deficiência , Ativador de Plasminogênio Tecidual/genética
17.
J Cell Physiol ; 162(3): 400-9, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7860647

RESUMO

The mechanism of induction of urokinase-type plasminogen activator (uPA) by basic fibroblast growth factor (bFGF) was explored in fetal bovine aortic endothelial GM 7373 cells. A three- to four-fold increase in the steady-state levels of uPA mRNA was observed after 6 h of incubation of the cell cultures with bFGF. Accordingly, nuclear run-on experiments showed a 2-2.4-fold increase in the rate of uPA gene transcription during the first 4 h of treatment with the growth factor. bFGF did not affect uPA mRNA stability, as evaluated by chase experiments with the mRNA synthesis inhibitor actinomycin D. Upregulation of uPA mRNA was followed by a delayed increase in uPA protein synthesis paralleled by an increase in secreted and cell-associated uPA activity. Twelve h were required before accumulated uPA mRNA was translated into the corresponding protein. During this time interval, the continuous presence of biologically active bFGF in the extracellular environment represented an absolute requirement for uPA mRNA translation. Substitution of residues Lys-27, Lys-30, and Arg-31 to glutamine residues in the bFGF molecule resulted in a mutant (M1Q-bFGF) that caused uPA mRNA accumulation in the absence of a significant increase in cell-associated uPA activity. M1Q-bFGF also induced an increase in cell-associated uPA activity only when added to the cell cultures in the presence of soluble heparin. These results provide evidence that bFGF can affect uPA expression in endothelial GM 7373 cells both at transcriptional and posttranscriptional translational levels. They also show the possibility to dissociate upregulation of uPA mRNA from upregulation of uPA activity by mutagenesis of the bFGF molecule.


Assuntos
Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Bovinos , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Mensageiro/genética , Proteínas Recombinantes , Relação Estrutura-Atividade , Suramina/farmacologia , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos
18.
J Cell Physiol ; 161(1): 149-59, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7929600

RESUMO

The single-copy gene of human basic fibroblast growth factor (bFGF) encodes four co-expressed isoforms, with an apparent molecular weight (M(r)) of 24 kD, 22.5 kD, 22 kD, and 18 kD, co-translated from a single mRNA. As a tool for the study of the role exerted by the different bFGF isoforms in the biology of endothelial cells, human recombinant 24-kD bFGF was produced and purified from transformed Escherichia coli cells. To this purpose, the novel CUG start codon present in human bFGF cDNA and responsible for the synthesis of 24-kD bFGF was mutagenized to the classic AUG start codon. Transient expression of the mutagenized cDNA in simian COS-1 cells, followed by immunolocalization and subcellular fractionation, resulted in the synthesis of high levels of 24-kD bFGF, which localizes in the cell nucleus as an intact protein. When the same 24-kD bFGF cDNA was expressed in E. coli, the recombinant protein was purified to homogeneity by heparin-Sepharose and ion-exchange chromatography. Recombinant 24-kD bFGF was similar to recombinant 18-kD bFGF in receptor-binding activity and in inducing cell proliferation, plasminogen activator production, and chemotactic movement in cultured endothelial cells. In agreement with the in vitro observations, 24-kD bFGF and 18-kD bFGF exerted a similar angiogenic response when assayed in vivo in the rabbit cornea. Experiments performed with the radiolabeled molecule demonstrated that 24-kD bFGF has an intrinsic ability to bind to high-affinity receptors when added to endothelial GM 7373 cell cultures. Receptor-bound 24-kD bFGF is internalized within the cell and associates with the nucleus with kinetics similar to 18-kD bFGF. Internalized 24-kD bFGF is first processed to the 18-kD form via a chloroquine-insensitive pathway and then to smaller fragments into the lysosomal compartment. At variance with the data obtained in transfected COS-1 cells, only limited amounts of exogenous internalized 24-kD bFGF associates with the nucleus in the intact form, mostly of the nuclear-bound molecule being represented by the processed 18-kD protein and by smaller degradation products. In conclusion, human recombinant 24-kD bFGF exerts a biological response in endothelial cells similar to 18-kD bFGF both in vitro and in vivo. Our data point to a different intracellular behavior of the high-molecular-weight bFGF isoform when added exogenously to cultured cells or when produced endogenously in transfected cells.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Animais , Bovinos , Linhagem Celular , Endotélio Vascular/citologia , Fator 2 de Crescimento de Fibroblastos/isolamento & purificação , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Isomerismo , Peso Molecular , Proteínas Recombinantes
19.
Growth Factors ; 8(1): 49-60, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8448039

RESUMO

Immunolocalization of basic fibroblast growth factor (bFGF) was investigated in NIH 3T3 cells transfected with a cDNA encoding for the 18 kD form of human bFGF (18 kD-bFGF) or with a bFGF cDNA encoding for both 18 kD-bFGF and NH2-terminal extended high molecular weight forms of bFGF (HMW-bFGFs). Nuclear and cytoplasmic bFGF-immunoreactivity was observed in both transfectants. Nuclear bFGF immunoreactivity was evenly distributed during interfase and associated with condensed chromosomes throughout the mitotic cycle. Cell fractionation, followed by Western blot analysis, confirmed the presence of 18 kD-bFGF and of HMW-bFGFs in the nucleus of transfected cells. Also, both 18-kD bFGF and HMW-bFGFs copurified with nuclear chromatin. After trypsin digestion, chromatin-bound bFGFs showed a rapid degradation of the nuclear-targeting NH2-terminal extension of HMW-bFGFs which were converted to the 18 kD form. On the contrary, 18 kD-bFGF appeared to be trypsin-resistant when bound to nuclear chromatin or to isolated eukaryote DNA. Thus, our data indicate that: i) both 18 kD-bFGF and HMW-bFGFs localize into the nucleus of transfected NIH 3T3 cells and bind to nuclear chromatin; ii) the interaction of all bFGF isoforms with nuclear chromatin is mediated by one or more sequences present within the 18 kD form; iii) the chromatin-binding domain of HMW-bFGFs is distinct from their nuclear-targeting domain.


Assuntos
Núcleo Celular/metabolismo , Cromatina/metabolismo , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células 3T3 , Animais , Western Blotting , DNA/genética , Eletroforese em Gel de Poliacrilamida , Fator 2 de Crescimento de Fibroblastos/genética , Camundongos , Peso Molecular , Fragmentos de Peptídeos/metabolismo , Transfecção , Tripsina/metabolismo
20.
Int J Cancer ; 53(1): 5-10, 1993 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-8380057

RESUMO

Biopsies of human normal adrenal medulla, adrenal pheochromocytoma, and chemodectoma were studied for the presence of basic fibroblast growth factor (bFGF). An immunoreactive M(r) 18,000 bFGF-like molecule was detected both in normal and neoplastic tissues. This molecule was identified as bFGF on the basis of its molecular weight, its affinity for heparin, and its capacity to induce plasminogen activator production in cultured endothelial GM 7373 cells. The levels of immunoreactive and biologically active bFGF appeared to be significantly higher in the extracts of adrenal pheochromocytoma and chemodectoma than in the extracts of normal adrenal medulla. bFGF immunostaining was detectable in the nuclei of chief (Type-I) cells and of endothelial cells both in normal adrenal medulla and in pheochromocytoma. Cytoplasmic bFGF positivity of endothelial cells was also observed in pheochromocytoma but not in normal tissue. The data are in keeping with the hypothesis that bFGF may exert autocrine and paracrine functions in the growth and neovascularization of human pheochromocytoma.


Assuntos
Neoplasias das Glândulas Suprarrenais/química , Medula Suprarrenal/química , Fator 2 de Crescimento de Fibroblastos/análise , Paraganglioma Extrassuprarrenal/química , Feocromocitoma/química , Adulto , Idoso , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...