Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 36(47): 6531-6541, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-28783175

RESUMO

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


Assuntos
Neovascularização Patológica/metabolismo , Lectinas de Plantas/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Inativadoras de Ribossomos/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Membrana Corioalantoide , Regulação para Baixo , Células Endoteliais/metabolismo , Feminino , Imunofluorescência , Galactosídeos , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ácido N-Acetilneuramínico/metabolismo , Neovascularização Fisiológica , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno , Sialiltransferases/genética , Sialiltransferases/metabolismo , Ressonância de Plasmônio de Superfície , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta-D-Galactosídeo alfa 2-6-Sialiltransferase , beta-Galactosídeo alfa-2,3-Sialiltransferase
2.
Oncogene ; 36(18): 2609-2618, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27819680

RESUMO

Syndecan-1 is a heparan sulfate proteoglycan (HSPG) commonly upregulated in AIDS-related B lymphoid malignancies. Tat is the main HIV-1 transactivating factor that has a major role in the pathogenesis of AIDS-related lymphomas (ARL) by engaging heparan sulfate proteoglycans (HSPGs), chemokine receptors and integrins at the lymphoid cell (LC) surface. Here B-lymphoid Namalwa cell clones that do not express or overexpress syndecan-1 (EV-Ncs and SYN-Ncs, respectively) were compared for their responsiveness with Tat: in the absence of syndecan-1, Tat induces a limited EV-Nc migration via C-X-C motif chemokine receptor 4 (CXCR4), G-proteins and Rac. Syndecan-1 overexpression increases SYN-Nc responsiveness to Tat and makes this response independent from CXCR4 and G-protein and dependent instead on pp60src phosphorylation. Tat-induced SYN-Nc migration and pp60src phosphorylation require the engagement of αvß3 integrin and consequent pp125FAK phosphorylation. This complex set of Tat-driven activations is orchestrated by the direct interaction of syndecan-1 with pp60src and its simultaneous coupling with αvß3. The Tat/syndecan-1/αvß3 interplay is retained in vivo and is shared also by other syndecan-1+ B-LCs, including BJAB cells, whose responsiveness to Tat is inhibited by syndecan-1 knockdown. In conclusion, overexpression of syndecan-1 confers to B-LCs an increased capacity to migrate in response to Tat, owing to a switch from a CXCR4/G-protein/Rac to a syndecan-1/αvß3/pp60src/pp125FAK signal transduction pathway that depends on the formation of a complex in which syndecan-1 interacts with Tat via its HS-chains, with αvß3 via its core protein ectodomain and with pp60src via its intracellular tail. These findings have implications in ARL progression and may help in identifying new therapeutical targets for the treatment of AIDS-associated neoplasia.


Assuntos
Quinase 1 de Adesão Focal/genética , Integrina alfaVbeta3/genética , Linfócitos/metabolismo , Neoplasias/genética , Sindecana-1/genética , Adesão Celular/genética , Regulação Neoplásica da Expressão Gênica , HIV-1/genética , Humanos , Linfócitos/patologia , Complexos Multiproteicos/genética , Neoplasias/patologia , Fosforilação , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Receptores CXCR4/genética , Transdução de Sinais/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/administração & dosagem , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
3.
Radiol Med ; 117(3): 354-68, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22020424

RESUMO

PURPOSE: We assessed the usefulness of contrast-enhanced magnetic resonance cholangiography (CE-MRC) with liver-specific contrast agent in evaluating the biliary tree after hepatic surgery. MATERIALS AND METHODS: A total of 142 patients with suspected biliary complications after liver surgery underwent hepatobiliary MR before and after administration of gadolinium ethoxy benzylic diethylenetriamine pentaacetic acid (Gd-EOB-DTPA). Unenhanced MR cholangiopancreatography (MRCP) and postcontrast MRC were obtained in all patients. Blinded image evaluation and semiquantitative analysis comparing MRCP and CE-MRC were performed by two experienced radiologists. RESULTS: In all cases, optimal postcontrast visualisation of the biliary tract was obtained. In 22 patients, a postsurgical biliary complication was confirmed. MRCP detected 64% of lesions, but in 36% of cases, an alteration was only suspected but not clearly defined. CE-MRC allowed definite diagnosis in 100% of cases. CONCLUSIONS: Hepatobiliary-specific contrast agents allow for accurate and extensive study of biliary tract alterations, especially in assessing postsurgical complications.


Assuntos
Doenças Biliares/diagnóstico , Colangiopancreatografia por Ressonância Magnética , Meios de Contraste , Gadolínio DTPA , Fígado/cirurgia , Humanos , Complicações Pós-Operatórias
4.
Hum Mol Genet ; 10(19): 2165-70, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11590133

RESUMO

The phenotypes in myotonic dystrophy types 1 and 2 (DM1 and DM2) are similar, suggesting a shared pathophysiologic mechanism. DM1 is caused by expansion of a CTG repeat in the DMPK gene. Pathogenic effects of this mutation are likely to be mediated, at least in part, by the expanded CUG repeat in mutant mRNA. The mutant transcripts are retained in the nucleus in multiple discrete foci. We investigated the possibility that DM2 is also caused by expansion of a CTG repeat or related sequence. Analysis of DNA by repeat expansion detection methods, and RNA by ribonuclease protection, did not show an expanded CTG or CUG repeat in DM2. However, hybridization of muscle sections with fluorescence-labeled CAG-repeat oligonucleotides showed nuclear foci in DM2 similar to those seen in DM1. Nuclear foci were present in all patients with symptomatic DM1 (n = 9) or DM2 (n = 9) but not in any disease controls or healthy subjects (n = 23). The foci were not seen with CUG- or GUC-repeat probes. Foci in DM2 were distinguished from DM1 by lower stability of the probe-target duplex, suggesting that a sequence related to the DM1 CUG expansion accumulates in the DM2 nucleus. Muscleblind proteins, which interact with expanded CUG repeats in vitro, localized to the nuclear foci in both DM1 and DM2. These results support the idea that nuclear accumulation of mutant RNA is pathogenic in DM1, suggest that a similar disease process occurs in DM2, and point to a role for muscleblind in the pathogenesis of both disorders.


Assuntos
Proteínas de Drosophila , Distrofia Miotônica/genética , Proteínas Nucleares/genética , RNA/metabolismo , Adulto , Idoso , Animais , Núcleo Celular/metabolismo , Drosophila , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Proteínas Nucleares/metabolismo , Ribonucleases/metabolismo , Repetições de Trinucleotídeos/genética
5.
J Biol Chem ; 276(41): 37900-8, 2001 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-11473122

RESUMO

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


Assuntos
Inibidores da Angiogênese/farmacologia , Endotélio Vascular/efeitos dos fármacos , Escherichia coli/metabolismo , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Polissacarídeos/farmacologia , Animais , Células CHO , Sequência de Carboidratos , Bovinos , Células Cultivadas , Embrião de Galinha , Cricetinae , Endotélio Vascular/citologia , Humanos , Polissacarídeos/química , Proteínas Recombinantes/antagonistas & inibidores
6.
Endothelium ; 8(1): 65-74, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11409852

RESUMO

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


Assuntos
Endotélio Vascular/fisiologia , Produtos do Gene tat/metabolismo , HIV-1/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neovascularização Fisiológica/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Animais , Butadienos/farmacologia , Bovinos , Células Cultivadas , Colágeno , Combinação de Medicamentos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Produtos do Gene tat/farmacologia , Humanos , Cinética , Laminina , Camundongos , Proteína Quinase 3 Ativada por Mitógeno , Nitrilas/farmacologia , Fragmentos de Peptídeos/farmacologia , Fosforilação , Proteoglicanas , Receptores de Fatores de Crescimento do Endotélio Vascular , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana
7.
J Biol Chem ; 276(25): 22420-5, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11304529

RESUMO

HIV-1 Tat protein, released from HIV-infected cells, may act as a pleiotropic heparin-binding growth factor. From this observation, extracellular Tat has been implicated in the pathogenesis of AIDS and of AIDS-associated pathologies. Here we demonstrate that the heparin analog pentosan polysulfate (PPS) inhibits the interaction of glutathione S-transferase (GST)-Tat protein with heparin immobilized to a BIAcore sensor chip. Competition experiments showed that Tat-PPS interaction occurs with high affinity (K(d) = 9.0 nm). Also, GST.Tat prevents the binding of [(3)H]heparin to GST.Tat immobilized to glutathione-agarose beads. In vitro, PPS inhibits GST.Tat internalization and, consequently, HIV-1 long terminal repeat transactivation in HL3T1 cells. Also, PPS inhibits cell surface interaction and mitogenic activity of GST.Tat in murine adenocarcinoma T53 Tat-less cells. In all assays, PPS exerts its Tat antagonist activity with an ID(50) equal to approximately 1.0 nm. In vivo, PPS inhibits the neovascularization induced by GST.Tat or by Tat-overexpressing T53 cells in the chick embryo chorioallantoic membrane. In conclusion, PPS binds Tat protein and inhibits its cell surface interaction, internalization, and biological activity in vitro and in vivo. PPS may represent a prototypic molecule for the development of novel Tat antagonists with therapeutic implications in AIDS and AIDS-associated pathologies, including Kaposi's sarcoma.


Assuntos
Produtos do Gene tat/antagonistas & inibidores , HIV-1/metabolismo , Poliéster Sulfúrico de Pentosana/farmacologia , Animais , Linhagem Celular , Embrião de Galinha , Endocitose , Produtos do Gene tat/metabolismo , Repetição Terminal Longa de HIV/genética , Humanos , Poliéster Sulfúrico de Pentosana/metabolismo , Ligação Proteica , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Ativação Transcricional/efeitos dos fármacos , Produtos do Gene tat do Vírus da Imunodeficiência Humana
8.
FASEB J ; 14(13): 1917-30, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11023976

RESUMO

Tat protein, a trans-activating factor of the human immunodeficiency virus type 1, acts also as an extracellular molecule modulating gene expression, cell survival, growth, transformation, and angiogenesis. Here we demonstrate that human thrombospondin-1 (TSP), a plasma glycoprotein and constituent of the extracellular matrix, binds to glutathione-S-transferase (GST)-Tat protein but not to GST. Scatchard plot analysis of the binding of free GST-Tat to immobilized TSP reveals a high-affinity interaction (Kd equal to 25 nM). Accordingly, TSP inhibits cell internalization and HIV-1 LTR trans-activating activity of extracellular Tat in HL3T1 cells with ID50 equal to 10-30 nM. Also, TSP inhibits cell interaction and mitogenic activity of extracellular Tat in T53 Tat-less cells. TSP is instead ineffective when administered after the interaction of Tat with cell surface heparan-sulfate proteoglycans has occurred, in keeping with its ability to prevent but not disrupt Tat/heparin interaction in vitro. Finally, TSP inhibits the autocrine loop of stimulation exerted by endogenous Tat in parental T53 cells. Accordingly, TSP overexpression inhibits cell proliferation, angiogenic activity, and tumorigenic capacity of stable T53 transfectants. Our data demonstrate the ability of TSP to bind to Tat protein and to affect its LTR trans-activating, mitogenic, angiogenic, and tumorigenic activity. These findings suggest that TSP may be implicated in the progression of AIDS and in AIDS-associated pathologies by modulating the bioavailability and biological activity of extracellular Tat.


Assuntos
Produtos do Gene tat/metabolismo , HIV-1 , Trombospondina 1/metabolismo , Adenocarcinoma/irrigação sanguínea , Animais , Antineoplásicos/metabolismo , Disponibilidade Biológica , Embrião de Galinha , Produtos do Gene tat/genética , Repetição Terminal Longa de HIV , Células HeLa , Heparina/farmacologia , Humanos , Camundongos , Camundongos Transgênicos , Mitógenos/metabolismo , Neovascularização Patológica , Ligação Proteica/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Neoplasias Cutâneas/irrigação sanguínea , Ativação Transcricional , Produtos do Gene tat do Vírus da Imunodeficiência Humana
9.
EMBO J ; 19(17): 4439-48, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10970838

RESUMO

Myotonic dystrophy (DM1) is an autosomal dominant neuromuscular disorder associated with a (CTG)(n) expansion in the 3'-untranslated region of the DM1 protein kinase (DMPK) gene. To explain disease pathogenesis, the RNA dominance model proposes that the DM1 mutation produces a gain-of-function at the RNA level in which CUG repeats form RNA hairpins that sequester nuclear factors required for proper muscle development and maintenance. Here, we identify the triplet repeat expansion (EXP) RNA-binding proteins as candidate sequestered factors. As predicted by the RNA dominance model, binding of the EXP proteins is specific for dsCUG RNAs and proportional to the size of the triplet repeat expansion. Remarkably, the EXP proteins are homologous to the Drosophila muscleblind proteins required for terminal differentiation of muscle and photoreceptor cells. EXP expression is also activated during mammalian myoblast differentiation, but the EXP proteins accumulate in nuclear foci in DM1 cells. We propose that DM1 disease is caused by aberrant recruitment of the EXP proteins to the DMPK transcript (CUG)(n) expansion.


Assuntos
Proteínas de Drosophila , Distrofia Miotônica/genética , Proteínas Nucleares/genética , Repetições de Trinucleotídeos , Sequência de Aminoácidos , Animais , Sequência de Bases , Núcleo Celular/metabolismo , DNA/metabolismo , Primers do DNA , Drosophila , Células HeLa , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Homologia de Sequência de Aminoácidos
10.
Adv Exp Med Biol ; 476: 7-34, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10949652

RESUMO

Angiogenesis is the process of generating new capillary blood vessels. Uncontrolled endothelial cell proliferation is observed in tumour neovascularization. Several growth factors and cytokines have been shown to stimulate endothelial cell proliferation in vitro and in vivo and among them FGF2 was one of the first to be characterised. FGF2 is a Mr 18,000 heparin-binding cationic polypeptide that induces proliferation, migration, and protease production in endothelial cells in culture and neovascularization in vivo. FGF2 interacts with endothelial cells through two distinct classes of receptors, the high affinity tyrosine-kinase receptors (FGFRs) and low affinity heparan sulfate proteoglycans (HSPGs) present on the cell surface and in the extracellular matrix. Besides experimental evidence for paracrine mode of action for FGF2, some observations raise the hypothesis that FGF2 may also play an autocrine role in endothelial cells. FGF2 may therefore represent a target for anti-angiogenic therapies. In order to assess the angiostatic potential of different classes of compounds, novel experimental models have been developed based on the autocrine and/or the paracrine capacity of FGF2.


Assuntos
Fator 2 de Crescimento de Fibroblastos/fisiologia , Neovascularização Patológica/metabolismo , Inibidores da Angiogênese , Animais , Endotélio Vascular/citologia , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Expressão Gênica , Hemangioma/metabolismo , Hemangioma/patologia , Humanos , Camundongos , Modelos Biológicos , Polímeros , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia , Ácidos Sulfônicos , Transfecção , Células Tumorais Cultivadas
11.
Nucleic Acids Res ; 27(17): 3534-42, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10446244

RESUMO

Myotonic dystrophy (DM) is associated with a (CTG) (n) triplet repeat expansion in the 3'-untranslated region of the myotonic dystrophy protein kinase (DMPK) gene. Using electron microscopy, we visualized large RNAs containing up to 130 CUG repeats and studied the binding of purified CUG-binding protein (CUG-BP) to these RNAs. Electron microscopic examination revealed perfect double-stranded (ds)RNA segments whose lengths were that expected for duplex RNA. The RNA dominant mutation model for DM pathogenesis predicts that the expansion mutation acts at the RNA level by forming long dsRNAs that sequester certain RNA-binding proteins. To test this model, we examined the subcellular distribution and RNA-binding properties of CUG-BP. While previous studies have demonstrated that mutant DMPK transcripts accumu-late in nuclear foci, the localization pattern of CUG-BP in both normal and DM cells was similar. Although CUG-BP in nuclear extracts preferentially photocrosslinked to DMPK transcripts, this binding was not proportional to (CUG) (n) repeat size. Moreover, CUG-BP localized to the base of the RNA hairpin and not along the stem, as visualized by electron micro-scopy. These results provide the first visual evidence that the DM expansion forms an RNA hairpin structure and suggest that CUG-BP is unlikely to be a sequestered factor.


Assuntos
Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Regiões 3' não Traduzidas/genética , Proteínas CELF1 , Células Cultivadas , Fibroblastos/metabolismo , Biblioteca Gênica , Células HeLa , Humanos , Microscopia Eletrônica , Modelos Genéticos , Miotonina Proteína Quinase , Conformação de Ácido Nucleico , Plasmídeos , Ligação Proteica , Proteínas Serina-Treonina Quinases/ultraestrutura , RNA de Cadeia Dupla/ultraestrutura , Proteínas de Ligação a RNA/ultraestrutura , Proteínas Recombinantes de Fusão/metabolismo , Ribonucleoproteínas/ultraestrutura , Repetições de Trinucleotídeos/genética
12.
Mol Biol Cell ; 10(2): 313-27, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9950679

RESUMO

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.


Assuntos
Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Gangliosídeos/metabolismo , Animais , Sítios de Ligação , Sequência de Carboidratos , Bovinos , Linhagem Celular Transformada , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/farmacologia , Gangliosídeos/química , Gangliosídeos/farmacologia , Humanos , Cinética , Dados de Sequência Molecular , Neovascularização Patológica/etiologia , Proteínas Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
13.
J Biol Chem ; 273(26): 16027-37, 1998 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-9632653

RESUMO

Heparin binds extracellular HIV-1 Tat protein and modulates its HIV long terminal repeat (LTR)-transactivating activity (M. Rusnati, D. Coltrini, P. Oreste, G. Zoppetti, A. Albini, D. Noonan, F. d'Adda di Fagagna, M. Giacca, and M. Presta (1997) J. Biol. Chem. 272, 11313-11320). On this basis, the glutathione S-transferase (GST)-TatR49/52/53/55/56/57A mutant, in which six arginine residues within the basic domain of Tat were mutagenized to alanine residues, was compared with GST-Tat for its capacity to bind immobilized heparin. Dissociation of the GST-TatR49/52/53/55/56/57A.heparin complex occurred at ionic strength significantly lower than that required to dissociate the GST-Tat.heparin complex. Accordingly, heparin binds immobilized GST-Tat and GST-TatR49/52/53/55/56/57A with a dissociation constant equal to 0.3 and 1.0 microM, respectively. Also, the synthetic basic domain Tat-(41-60) competes with GST-Tat for heparin binding. Suramin inhibits [3H]heparin/Tat interaction, 125I-GST-Tat internalization, and the LTR-transactivating activity of extracellular Tat in HL3T1 cells and prevents 125I-GST-Tat binding and cell proliferation in Tat-overexpressing T53 cells. The suramin derivative 14C-PNU 145156E binds immobilized GST-Tat with a dissociation constant 5 times higher than heparin and is unable to bind GST-TatR49/52/53/55/56/57A. Although heparin was an antagonist more potent than suramin, modifications of the backbone structure in selected suramin derivatives originated Tat antagonists whose potency was close to that shown by heparin. In conclusion, suramin derivatives bind the basic domain of Tat, prevent Tat/heparin and Tat/cell surface interactions, and inhibit the biological activity of extracellular Tat. Our data demonstrate that tailored polysulfonated compounds represent potent extracellular Tat inhibitors of possible therapeutic value.


Assuntos
Produtos do Gene tat/metabolismo , HIV-1 , Heparina/metabolismo , Suramina/análogos & derivados , Animais , Divisão Celular , Distamicinas/metabolismo , Produtos do Gene tat/antagonistas & inibidores , Repetição Terminal Longa de HIV , Humanos , Camundongos , Camundongos Transgênicos , Modelos Químicos , Mimetismo Molecular , Suramina/metabolismo , Ativação Transcricional , Células Tumorais Cultivadas , Produtos do Gene tat do Vírus da Imunodeficiência Humana
14.
Mol Biol Cell ; 7(3): 369-81, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8868466

RESUMO

Basic fibroblast growth factor (FGF-2) induces cell proliferation and urokinase-type plasminogen activator (uPA) production in fetal bovine aortic endothelial GM 7373 cells. In the present paper we investigated the role of the interaction of FGF-2 with tyrosine-kinase (TK) FGF receptors (FGFRs) in mediating uPA up-regulation in these cells. The results show that FGF-2 antagonists suramin, protamine, heparin, the synthetic peptide FGF-2(112-155), and a soluble form of FGFR-1 do not inhibit FGF-2-mediated uPA up-regulation at concentrations that affect growth factor binding to cell surface receptors and mitogenic activity. In contrast, tyrosine phosphorylation inhibitors and overexpression of a dominant negative TK- mutant of FGFR-1 abolish the uPA-inducing activity of FGF-2, indicating that FGFR and its TK activity are essential in mediating uPA induction. Accordingly, FGF-2 induces uPA up-regulation in Chinese hamster ovary cells transfected with wild-type FGFR-1, -2, -3, or -4 but not with TK- FGFR-1 mutant. Small unilamellar phosphatidyl choline:cholesterol vesicles loaded with FGF-2 increased uPA production in GM 7373 cells in the absence of a mitogenic response. Liposome-encapsulated FGF-2 showed a limited but significant capacity, relative to free FGF-2, to interact with FGFR both at 4 degrees C and 37 degrees C and to be internalized within the cell. uPA up-regulation by liposome-encapsulated FGF-2 was quenched by neutralizing anti-FGF-2 antibodies, indicating that the activity of liposome-delivered FGF-2 is mediated by an extracellular action of the growth factor. Taken together, the data indicate that a distinct interaction of FGF-2 with FGFR, quantitatively and/or qualitatively different from the one that leads to mitogenicity, is responsible for the uPA-inducing activity of the growth factor.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Aorta/citologia , Aorta/embriologia , Células CHO , Bovinos , Linhagem Celular , Cricetinae , Endotélio Vascular/citologia , Fator 2 de Crescimento de Fibroblastos/agonistas , Humanos , Lipossomos/metabolismo , Mitógenos , Proteínas Tirosina Quinases/metabolismo
15.
Dev Biol ; 170(1): 39-49, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7601314

RESUMO

Chorioallantoic membrane (CAM) and chorioallantoic fluid (CAF) of the chick embryo were studied for the presence of immunoreactive and biologically active basic fibroblast growth factor (bFGF) from Day 6 to Day 18 of incubation. An immunoreactive M(r) 16,000 bFGF-like molecule was detected both in CAM and in CAF. 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 biologically active and immunoreactive bFGF vary in CAM and CAF during embryonic development, maximal concentrations being observed between Days 10 and 14 of incubation. At all time points investigated, absolute concentrations of bFGF were significantly higher in CAM (ranging from 25 to 183 ng/g of wet tissue) than in CAF (ranging from 0.2 to 4 ng/ml). In a parallel series of experiments performed at Day 8 and evaluated at Day 12 of chick embryo development, human recombinant bFGF and neutralizing anti-bFGF antibody were investigated for their capacity to affect the vasoproliferative processes of the CAM. The two molecules either were applied onto the surface of the CAM or were injected into the allantoic sac. When bFGF or anti-bFGF antibodies were absorbed on methylcellulose discs and applied on the top of the CAM, they exerted a strong angiogenic or anti-angiogenic effect, respectively. On the contrary, when bFGF or the corresponding neutralizing antibody was injected into the allantoic sac, no modifications of the vasoproliferative processes of the CAM were observed at either the macroscopic or the microscopic level. These results provide evidence indicating that endogenous bFGF has a rate-limiting role in the vascularization of the CAM during chick embryogenesis. bFGF located within the CAM, rather than that present in the CAF, appears to be involved in this developmental process.


Assuntos
Alantoide/irrigação sanguínea , Córion/irrigação sanguínea , Fator 2 de Crescimento de Fibroblastos/fisiologia , Alantoide/efeitos dos fármacos , Alantoide/metabolismo , Animais , Anticorpos/farmacologia , Aorta , Líquidos Corporais/metabolismo , Capilares/embriologia , Bovinos , Embrião de Galinha , Córion/efeitos dos fármacos , Córion/metabolismo , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/imunologia , Humanos , Ativadores de Plasminogênio/biossíntese , Proteínas Recombinantes/farmacologia
16.
Int J Dev Neurosci ; 13(1): 29-39, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7793308

RESUMO

The presence of basic fibroblast growth factor (bFGF) and FGF receptors was investigated in microglia cells derived from human fetal brain long-term cultures. Production of bFGF was suggested through the capability of microglial extracts to stimulate plasminogen activator (PA) synthesis in endothelial cells. The identity of PA-stimulating activity with bFGF was confirmed by its high affinity for heparin and its cross-reactivity with polyclonal antibodies to human recombinant bFGF. These antibodies recognized a cell-associated M(r) 18,000 protein as well as trace amounts of the M(r) 24,000 bFGF isoform in Western blot. All microglial cells showed bFGF immunoreactivity in the cytoplasm and, sometimes, in the nucleus. Scatchard plot analysis of 125I-bFGF binding data revealed the presence of low affinity heparansulphate proteoglycans (380,000 +/- 60,000 sites/cell; Kd = 730 +/- 200 nM) and of high affinity tyrosine-kinase receptors (10,300 + 2500 sites/cell; Kd = 30 +/- 9 pM). Immunocytochemistry confirmed the presence of FGF receptor (1/flg) on the cell surface of some, but not all microglial cells, with prevalent association to ameboid microglia. Transcripts for FGF receptors 1, 2, 3 and 4 were found in microglia by Northern blot analysis. Co-expression of bFGF and its receptors in human fetal microglia suggests an autocrine role of bFGF in these cells.


Assuntos
Feto/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Microglia/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Células Cultivadas , Proteínas Filagrinas , Humanos , Imuno-Histoquímica , Distribuição Tecidual , Transcrição Gênica
17.
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
18.
J Cell Physiol ; 154(1): 152-61, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8419401

RESUMO

We have shown (Presta et al., Cell Regul., 2:719-726, 1991) that a long-lasting interaction of basic fibroblast growth factor (bFGF) with endothelial GM 7373 cells is required to induce cell proliferation. In the present work, we have investigated the interaction of 125I-bFGF with GM 7373 cells, its pathway of internalization, and its intracellular fate under the same experimental conditions previously utilized to assess the mitogenic activity of the growth factor. Cell cultures were incubated with 10 ng/ml 125I-bFGF for 2 h at 4 degrees C. Then, cells were shifted to 37 degrees C without changing the medium. A rapid down-regulation of high affinity sites, paralleled by a rapid internalization of 125I-bFGF, was observed during the first 1-2 h at 37 degrees C. After 6-8 h, also low affinity sites down-regulate. This was paralleled by a continuous internalization of 125I-bFGF and by a slow disappearance of the growth factor from the culture medium. This suggests that GM 7373 cells activate, when exposed to bFGF for a long period of time, a late internalization pathway mediated by low affinity sites. This hypothesis was supported by the following experimental evidence: 1) soluble heparin inhibited the prolonged internalization of 125I-bFGF and its binding to low affinity sites with the same potency; 2) treatment of GM 7373 cells with heparinase, which removes most of the low affinity sites, also inhibited the prolonged internalization of 125I-bFGF. 125I-bFGF internalized via low affinity sites was partially protected from lysosomal degradation. This was the case also when 125I-bFGF was internalized in the presence of soluble heparin, suggesting that the complexes bFGF-cell surface glycosaminoglycan and bFGF-soluble heparin are maintained during the internalization of the growth factor. Moreover, the capacity of soluble heparin to inhibit the mitogenic activity of bFGF also when added to cell cultures several hours after the growth factor indicates that the requirement for a prolonged interaction of bFGF with GM 7373 cells in order to induce cell proliferation might be related to the late internalization of the growth factor via low affinity sites.


Assuntos
Endocitose , Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Heparina/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Bovinos , Linhagem Celular , Membrana Celular/metabolismo , Endotélio Vascular/citologia , Humanos , Cinética
19.
Growth Factors ; 9(4): 269-78, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8148156

RESUMO

Residues 27-31 (Lys-Asp-Pro-Lys-Arg) of the 155-amino acid form of basic fibroblast growth factor (bFGF) are in good agreement with a consensus sequence for nuclear translocation. To evaluate the role of this sequence in mediating the intracellular localization and biological activity of bFGF, basic residues Lys-27, Lys-30, and Arg-31 were changed to neutral glutamine residues by site-directed mutagenesis of the human bFGF cDNA. The bFGF mutant (M1Q-bFGF) was expressed in eukaryotic cells and in prokaryotic cells, from which it was purified to homogeneity. Transient expression of bFGF cDNA and of M1Q-bFGF cDNA in simian COS-1 cells followed by immunolocalization and by subcellular fractionation indicated that both molecules localize in the nucleus, as well as in the cytoplasm of transfected cells, and interact with nuclear chromatin and with eukaryote DNA in a similar manner. Prokaryotic expression of M1Q-bFGF cDNA yields a polypeptide endowed with a receptor-binding capacity and mitogenic activity similar to that exerted by wild-type bFGF. However, recombinant M1Q-bFGF showed a drastically reduced capacity to induce the production of urokinase-type plasminogen activator (uPA) in endothelial cells. The uPA-inducing activity of M1Q-bFGF was fully restored by the presence of soluble heparin in the culture medium. In conclusion, the sequence bFGF(27-31) does not appear to represent a nuclear translocation and/or retention sequence for bFGF. However, neutralization of its basic residues seems to modify the tertiary structure of the growth factor, thus affecting some of its biological properties.


Assuntos
Núcleo Celular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Transporte Biológico , Bovinos , Linhagem Celular , Clonagem Molecular , Sequência Consenso , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Immunoblotting , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...