Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 291(6): H2692-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16731651

RESUMO

Stress-induced release of IL-1alpha and fibroblast growth factor-1 is dependent on intracellular copper and is a major driver of neointimal hyperplasia. Therefore, we assessed the effect of tetrathiomolybdate (TTM), a clinically proven copper chelator, on in-stent restenosis. Nine pigs were treated with TTM (5 mg/kg po) twice daily for 2 wk before stent implantation and for 4 wk thereafter, and nine pigs served as controls. In-stent restenosis was assessed by quantitative coronary angiography (QCA), intravascular ultrasound (IVUS), and histomorphometry. Serum ceruloplasmin activity was used as a surrogate marker of copper bioavailability. In TTM-treated animals, ceruloplasmin dropped 70 +/- 10% below baseline levels. Baseline characteristics were comparable in TTM-treated and control animals. At 4-wk follow-up, all parameters relevant to in-stent restenosis were significantly reduced in TTM-treated animals: minimal lumen diameter by QCA was 2.03 +/- 0.57 and 1.47 +/- 0.45 mm in TTM-treated and control animals, respectively (P < 0.05), percent stenosis diameter was 39% less in TTM-treated animals (27.1 +/- 16.6% vs. 44.5 +/- 16.1%, P < 0.05), minimal lumen area by IVUS was 60% larger in TTM-treated animals (4.27 +/- 1.56 vs. 2.67 +/- 1.19 mm(2), P < 0.05), and neointimal volume by histomorphometry was 37% less in TTM-treated animals (34.9 +/- 11.5 vs. 55.2 +/- 19.6 mm(3), P < 0.05). We conclude that systemic copper chelation with a clinically approved chelator significantly inhibits in-stent restenosis.


Assuntos
Quelantes/farmacologia , Cobre/metabolismo , Reestenose Coronária/prevenção & controle , Vasos Coronários/fisiopatologia , Molibdênio/farmacologia , Stents , Animais , Ceruloplasmina/metabolismo , Quelantes/metabolismo , Terapia por Quelação/métodos , Angiografia Coronária , Reestenose Coronária/patologia , Reestenose Coronária/fisiopatologia , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/patologia , Modelos Animais de Doenças , Masculino , Molibdênio/metabolismo , Suínos , Fatores de Tempo , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/patologia , Ultrassonografia de Intervenção
2.
Biochem Biophys Res Commun ; 310(4): 1041-7, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-14559220

RESUMO

Although the extravesicular p40 domain of the transmembrane protein, p65 synaptotagmin (Syt) 1, is essential for the non-classical export of the signal peptide-less structure, FGF1, it was not possible to identify a specific intracellular protease responsible for the processing of p65 Syt1. Surprisingly, analysis of the p65 Syt1 coding sequence revealed the presence of two potential alternative ATG codons corresponding to Met103 and Met113 both of which were flanked by Kozak sequences. Indeed, in vitro translation of a Met103Ile but not a Met113Ile p65 Syt1 point mutant exhibited reduced expression of p40 Syt1 and the double p65 Syt1 Met103Ile and Met113Ile point mutant was unable to translate the p40 Syt1 isoform. Since the expression of the p65 Syt1 double point mutant inhibited the stress-induced release of FGF1, it is likely that the alternative translation of the p65 Syt1 transcript at Met103 may be involved in the generation of intracellular p40 Syt1, a critical component of the FGF1 release pathway.


Assuntos
Proteínas de Ligação ao Cálcio , Fator 1 de Crescimento de Fibroblastos/metabolismo , Glicoproteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Biossíntese de Proteínas , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Complementar , Glicoproteínas de Membrana/fisiologia , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Células NIH 3T3 , Proteínas do Tecido Nervoso/fisiologia , Sinaptotagmina I , Sinaptotagminas
4.
Am J Pathol ; 159(3): 875-83, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11549580

RESUMO

The Jagged/Notch signaling pathways control cell fate determination and differentiation, and their dysfunction is associated with human pathologies involving cardiovascular abnormalities. To determine the presence of these genes during vascular response to injury, we analyzed expression of Jagged1, Jagged2, and Notch1 through 4 after balloon catheter denudation of the rat carotid artery. Although low levels of Jagged1, Jagged2, and constitutive expression of Notch1 were seen in uninjured endothelium, expression of all was significantly increased in injured vascular cells. High Jagged1 expression was restricted to the regenerating endothelial wound edge, whereas Notch transcripts were abundant in endothelial and smooth muscle cells. To understand the basis for Jagged/Notch control of cellular phenotype, we studied an in vitro model of NIH3T3 cells transfected with a secreted form of the extracellular domain of Jagged1. We report that the soluble Jagged1 protein caused decreased cell-matrix adhesion and cell migration defects. Cadherin-mediated intercellular junctions as well as focal adhesions were modified in soluble Jagged1 transfectants, demonstrating that cell-cell contacts and adhesion plaques may be targets of Jagged/Notch activity. We suggest that Jagged regulation of cell-cell and cell-matrix interactions may contribute to the control of cell migration in situations of tissue remodeling in vivo.


Assuntos
Lesões das Artérias Carótidas/genética , Expressão Gênica , Proteínas de Membrana/genética , Família Multigênica , Ferimentos não Penetrantes/genética , Células 3T3 , Animais , Proteínas de Ligação ao Cálcio , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/fisiopatologia , Comunicação Celular/fisiologia , Movimento Celular/fisiologia , Junções Célula-Matriz/fisiologia , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intercelular , Proteína Jagged-1 , Proteína Jagged-2 , Camundongos , Fenótipo , Proteínas/fisiologia , Ratos , Receptores Notch , Proteínas Serrate-Jagged , Ferimentos não Penetrantes/patologia , Ferimentos não Penetrantes/fisiopatologia
5.
Growth Factors ; 18(4): 277-85, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11519826

RESUMO

The signal peptide-less FGF gene family prototype, FGF1 is released in response to temperature stress in vitro as a latent reducing agent-sensitive homodimer non-covalently complexed with the extravesicular p40 domain of p65 synaptotagmin (Syt)1. Because FGF1 is well recognized as an angiogenesis factor in vivo and angiogenesis is known to be induced by hypoxia, we examined the release of FGF1 and p40 Syt1 under conditions of hypoxia and temperature stress using a chemostatic microcarrier cell culture system. We report that like the pathway used by FGF1 and p40 Syt1 release under temperature stress, hypoxia also induces the release of FGF1 and p40 Syt1 with similar kinetic and pharmacologic properties including the requirement for functional cysteine residues. Lastly, FGF1 and p40 Syt1 release in response to hypoxia and temperature stress is sensitive to lipoxygenase and cyclooxygenase inhibitors suggesting that arachidonic acid metabolism may play an important role in the mechanism of FGF1 release in vitro.


Assuntos
Proteínas de Ligação ao Cálcio , Hipóxia Celular/fisiologia , Fator 1 de Crescimento de Fibroblastos/metabolismo , Células 3T3 , Ácido 5,8,11,14-Eicosatetrainoico/farmacologia , Aminopiridinas/farmacologia , Animais , Ácido Araquidônico/metabolismo , Linhagem Celular , Inibidores de Ciclo-Oxigenase/farmacologia , Cisteína/química , Dimerização , Fator 1 de Crescimento de Fibroblastos/química , Fator 1 de Crescimento de Fibroblastos/genética , Humanos , Inibidores de Lipoxigenase/farmacologia , Substâncias Macromoleculares , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Nitrobenzenos/farmacologia , Estresse Fisiológico/fisiopatologia , Sulfonamidas/farmacologia , Sinaptotagmina I , Sinaptotagminas , Temperatura , Transfecção
6.
J Biol Chem ; 276(27): 25549-57, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11432880

RESUMO

Fibroblast growth factor (FGF) 1 is known to be released in response to stress conditions as a component of a multiprotein aggregate containing the p40 extravescicular domain of p65 synaptotagmin (Syt) 1 and S100A13. Since FGF1 is a Cu2+-binding protein and Cu2+ is known to induce its dimerization, we evaluated the capacity of recombinant FGF1, p40 Syt1, and S100A13 to interact in a cell-free system and the role of Cu2+ in this interaction. We report that FGF1, p40 Syt1, and S100A13 are able to bind Cu2+ with similar affinity and to interact in the presence of Cu2+ to form a multiprotein aggregate which is resistant to low concentrations of SDS and sensitive to reducing conditions and ultracentrifugation. The formation of this aggregate in the presence of Cu2+ is dependent on the presence of S100A13 and is mediated by cysteine-independent interactions between S100A13 and either FGF1 or p40 Syt1. Interestingly, S100A13 is also able to interact in the presence of Cu2+ with Cys-free FGF1 and this observation may account for the ability of S100A13 to export Cys-free FGF1 in response to stress. Lastly, tetrathiomolybdate, a Cu2+ chelator, significantly represses in a dose-dependent manner the heat shock-induced release of FGF1 and S100A13. These data suggest that S100A13 may be involved in the assembly of the multiprotein aggregate required for the release of FGF1 and that Cu2+ oxidation may be an essential post-translational intracellular modifier of this process.


Assuntos
Cobre/farmacologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteínas S100/metabolismo , Estresse Fisiológico/metabolismo , Animais , Sistema Livre de Células , Cisteína/metabolismo , Detergentes/farmacologia , Dimerização , Fator 1 de Crescimento de Fibroblastos , Humanos , Molibdênio/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Oxirredução , Fragmentos de Peptídeos/metabolismo , Coelhos , Dodecilsulfato de Sódio/farmacologia , Sinaptotagmina I
7.
J Biol Chem ; 276(34): 32022-30, 2001 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-11427524

RESUMO

We have previously demonstrated that the expression of the soluble extracellular domain of the transmembrane ligand for Notch receptors, Jagged 1 (sJ1), in NIH 3T3 cells results in the formation of a matrix-dependent chord-like phenotype, the loss of contact inhibition of growth, and an inhibition of pro-alpha 1(I) collagen expression. In an effort to define the mechanism by which sJ1 induces this phenotype, we report that sJ1 transfectants display biochemical and cytoskeletal alterations consistent with the activation of Src. Indeed, cotransfection of sJ1 transfectants with a dominant-negative mutant of Src resulted in the loss of matrix-dependent chord formation and correlated with the restoration of type I collagen expression and contact inhibition of growth. We also report that the sJ1-mediated induction of Src activity and related phenotypes, including chord formation, may result from the inhibition of endogenous Jagged 1-mediated Notch signaling since it was not possible to detect an sJ1-dependent induction of CSL-dependent transcription in these cells. Interestingly, NIH 3T3 cells transfected with dominant-negative (but not constitutively active) mutants of either Notch 1 or Notch 2 displayed a similar Src-related phenotype as the sJ1 transfectants. These data suggest that the ability of sJ1 to mediate chord formation is Src-dependent and requires the repression of endogenous Jagged 1-mediated Notch signaling, which is tolerant to the destabilization of the actin cytoskeleton, a mediator of cell migration.


Assuntos
Proteínas de Membrana/fisiologia , Proteína Oncogênica pp60(v-src)/metabolismo , Proteínas/fisiologia , Células 3T3 , Animais , Proteínas de Ligação ao Cálcio , Cortactina , Peptídeos e Proteínas de Sinalização Intercelular , Proteína Jagged-1 , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , Mutagênese Sítio-Dirigida , Fenótipo , Fosforilação , Proteínas/genética , Proteínas/metabolismo , Receptores Notch , Proteínas Serrate-Jagged , Espectrometria de Fluorescência , Transfecção , Tirosina/metabolismo
8.
J Biol Chem ; 276(25): 22544-52, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11410600

RESUMO

S100A13, a member of the S100 gene family of Ca(2+)-binding proteins has been previously characterized as a component of a brain-derived heparin-binding multiprotein aggregate/complex containing fibroblast growth factor 1 (FGF1). We report that while expression of S100A13 in NIH 3T3 cells results in the constitutive release of S100A13 into the extracellular compartment at 37 degrees C, co-expression of S100A13 with FGF1 represses the constitutive release of S100A13 and enables NIH 3T3 cells to release S100A13 in response to temperature stress. S100A13 release in response to stress occurs with kinetics similar to that observed for the stress-induced release of FGF1, but S100A13 expression is able to reverse the sensitivity of FGF1 release to inhibitors of transcription and translation. The release of FGF1 and S100A13 in response to heat shock results in the solubility of FGF1 at 100% (w/v) ammonium sulfate saturation, and the expression of a S100A13 deletion mutant lacking its novel basic residue-rich domain acts as a dominant negative effector of FGF1 release in vitro. Surprisingly, the expression of S100A13 also results in the stress-induced release of a Cys-free FGF1 mutant, which is normally not released from NIH 3T3 cells in response to heat shock. These data suggest that S100A13 may be a component of the pathway for the release of the signal peptide-less polypeptide, FGF1, and may involve a role for S100A13 in the formation of a noncovalent FGF1 homodimer.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Resposta ao Choque Térmico , Proteínas S100 , Células 3T3 , Animais , Fator 1 de Crescimento de Fibroblastos , Heparina/metabolismo , Camundongos , Biossíntese de Proteínas , Ovinos , Transcrição Gênica
9.
J Biol Chem ; 276(7): 5147-51, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11087725

RESUMO

Interleukin (IL)1alpha mediates proinflammatory events through its extracellular interaction with the IL1 type I receptor. However, IL1alpha does not contain a conventional signal peptide sequence that provides access to the endoplasmic reticulum-Golgi apparatus for secretion. Thus, we have studied the release of the precursor (p) and mature (m) forms of IL1alpha from NIH 3T3 cells. We have demonstrated that mIL1alpha but not pIL1alpha was released in response to heat shock with biochemical and pharmacological properties similar to those reported for the stress-mediated release pathway utilized by fibroblast growth factor (FGF)1. However, unlike the FGF1 release pathway, the IL1alpha release pathway appears to function independently of synaptotagmin (Syt)1 because the expression of a dominant-negative form of Syt1, which represses the release of FGF1, did not inhibit the release of mIL1alpha in response to temperature stress. Interestingly, whereas the expression of both mIL1alpha and FGF1 in NIH 3T3 cells did not impair the stress-induced release of either polypeptide, the expression of both pIL1alpha and FGF1 repressed the release of FGF1 in response to temperature stress. These data suggest that the release of mIL1alpha requires proteolytic processing of its precursor form and that mIL1alpha and FGF1 may utilize similar but distinct mechanisms for export.


Assuntos
Proteínas de Ligação ao Cálcio , Fator 2 de Crescimento de Fibroblastos/metabolismo , Resposta ao Choque Térmico , Interleucina-1/fisiologia , Precursores de Proteínas/fisiologia , Células 3T3 , Aminopiridinas/farmacologia , Animais , Brefeldina A/farmacologia , Desoxiglucose/farmacologia , Fator 1 de Crescimento de Fibroblastos , Interleucina-1/genética , Interleucina-1/metabolismo , Cinética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Transporte Proteico , Deleção de Sequência , Sinaptotagmina I , Sinaptotagminas , Transfecção
11.
J Biol Chem ; 275(42): 32753-62, 2000 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-10921913

RESUMO

Amlexanox binds S100A13 and inhibits the release of fibroblast growth factor 1 (FGF1). Because members of the S100 gene family are known to be involved with the function of the cytoskeleton, we examined the ability of amlexanox to modify the cytoskeleton and report that amlexanox induces a dramatic reduction in the presence of actin stress fibers and the appearance of a random, non-oriented distribution of focal adhesion sites. Correspondingly, amlexanox induces the complete and reversible non-apoptotic inhibition of cell migration and proliferation, and although amlexanox does not induce either the down-regulation of F-actin levels or the depolymerization of actin filaments, it does induce the tyrosine phosphorylation of cortactin, a Src substrate known to regulate actin bundling. In addition, a dominant negative form of Src is able to partially rescue cells from the effect of amlexanox on both the actin cytoskeleton and cell migration. In contrast, the inhibition of cell proliferation by amlexanox correlates with the inhibition of cyclin D1 expression without interference of the receptor tyrosine kinase/mitogen-activated protein kinase signaling pathway. Last, the ability of amlexanox to inhibit FGF1 release is reversible and correlates with the restoration of the actin cytoskeleton, suggesting a role for the actin cytoskeleton in the FGF1 release pathway.


Assuntos
Actinas/fisiologia , Aminopiridinas/farmacologia , Movimento Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Endotélio Vascular/fisiologia , Genes src , Músculo Liso Vascular/fisiologia , Células 3T3 , Actinas/química , Actinas/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Aorta , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Cortactina , Citoesqueleto/fisiologia , Citoesqueleto/ultraestrutura , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fator 1 de Crescimento de Fibroblastos , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/fisiologia , Humanos , Células L , Camundongos , Proteínas dos Microfilamentos/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Fosforilação , Ratos , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Superfície Celular/fisiologia , Esteroides , Transfecção , Veias Umbilicais , Xenopus laevis
12.
Biochem Biophys Res Commun ; 268(3): 853-9, 2000 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-10679295

RESUMO

Jagged-Notch interactions regulate a transmembrane ligand-receptor signaling pathway involved in the regulation of cell fate determination as well as myoblast and endothelial cell differentiation. To further examine the role of the transmembrane ligand, Jagged-1, in the regulation of cell differentiation, we stably transfected NIH 3T3 cells with a truncated form of Jagged(J)-1, which results in the secretion of a soluble(s) form of the protein. Comparison of gene expression by serial analysis demonstrated that among the 227 transcripts differentially regulated in the sJ-1 transfectants, the expression of the pro-alpha-2(I) collagen transcript and pro-alpha-1(I) collagen translation product was predominantly repressed in sJ-1 transfectants. When plated on extracellular matrices, sJ-1 transfectants formed prominent chord-like structures on type I collagen but not on fibrin, fibronectin, or vitronectin. While the sJ-1 transfectants exhibited growth kinetics similar to control cells and were unable to grow in soft agar, the cells were less sensitive to contact inhibition of growth in vitro and sJ-1 allografts formed tissue masses in nude mice after a prolonged latency period and exhibited an abundance of host-derived microvascular endothelial cells. These data suggest that J-1 may be able to modulate, in a matrix-dependent manner, the organization of cell to cell interactions including its ability to promote the development of chord-like structures.


Assuntos
Proteínas/metabolismo , Células 3T3 , Animais , Sequência de Bases , Proteínas de Ligação ao Cálcio , Comunicação Celular , Diferenciação Celular , Divisão Celular , Primers do DNA/genética , Matriz Extracelular/metabolismo , Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular , Proteína Jagged-1 , Proteínas de Membrana , Camundongos , Camundongos Nus , Neovascularização Fisiológica , Fenótipo , Proteínas/química , Proteínas/genética , Proteínas Serrate-Jagged , Transdução de Sinais , Transfecção
14.
J Cell Physiol ; 177(1): 123-9, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9731752

RESUMO

Sensory cells in the cochlea of the rat transiently express acidic fibroblast growth factor (FGF-1) during the developmental period of terminal innervation in the sensory epithelium. To explore the potential role of FGF-1 in terminal innervation events, the response of cochlear ganglion neurons to FGF-1 was evaluated in culture. Explants from the spiral ganglion of postnatal day 5 rats were cultured in the presence of exogenous FGF-1, with or without heparin. FGF-1 in the culture medium produced a dose-dependent increase in the number and length of neurites produced by spiral ganglion neurons, a response that was enhanced by heparin. To assess the effects of FGF-1 produced by a focal, cellular source, additional explants were cocultured with 3T3 cell transfectants that secrete FGF-1. Neurites that came into contact with FGF-1 secreting cells branched, formed bouton-like terminal swellings on the surface of the transfectants, and stopped extending. The results suggest that FGF-1 may stimulate neurite extension into the sensory epithelium of the cochlea and that focal production of FGF-1 may contribute to the formation of contacts on sensory cells by developing neurites.


Assuntos
Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/farmacocinética , Células Ciliadas Auditivas/metabolismo , Neuritos/metabolismo , Gânglio Espiral da Cóclea/citologia , Células 3T3 , Animais , Meios de Cultura/farmacologia , Expressão Gênica/fisiologia , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/ultraestrutura , Heparitina Sulfato/farmacologia , Camundongos , Neuritos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Transfecção/métodos
15.
J Biol Chem ; 273(35): 22217-23, 1998 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-9712835

RESUMO

By using p65 synaptotagmin-1 and fibroblast growth factor (FGF)-1:beta-galactosidase (beta-gal) NIH 3T3 cell co-transfectants, we demonstrate that a proteolytic fragment consisting of the extravesicular domain of synaptotagmin-1 is released into the extracellular compartment in response to temperature stress with similar kinetics and pharmacological properties as FGF-1:beta-gal. Using a deletion mutant that lacks 95 amino acids from the extravesicular domain of synaptotagmin-1, neither synaptotagmin-1 nor FGF-1:beta-gal are able to access the stress-induced release pathway. Furthermore, the p40 extravesicular fragment of synaptotagmin-1 is constitutively released in p40 synaptotagmin-1 NIH 3T3 cell transfectants, and this release is potentiated when the cells are subjected to temperature stress. These data demonstrate that the p40 fragment derived from synaptotagmin-1 is able to utilize the FGF-1 non-classical exocytotic pathway and that the release of FGF-1 is dependent on synaptotagmin-1.


Assuntos
Proteínas de Ligação ao Cálcio , Fatores de Crescimento de Fibroblastos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células 3T3 , Animais , Sequência de Bases , Primers do DNA , Fatores de Crescimento de Fibroblastos/genética , Resposta ao Choque Térmico , Cinética , Camundongos , Ratos , Sinaptotagmina I , Sinaptotagminas , beta-Galactosidase/genética
16.
J Biol Chem ; 273(35): 22209-16, 1998 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-9712834

RESUMO

The heparin-binding fibroblast growth factor (FGF) prototypes lack a classical signal sequence, yet their presence is required in the extracellular compartment for the activation of cell-surface receptor-dependent signaling. Early studies with FGF-1 demonstrated its presence in bovine brain as a novel high molecular weight complex, and subsequent studies identified a second heparin-binding protein that co-purified with FGF-1. Polypeptide sequence analysis revealed that this heparin-binding protein corresponded to the extravesicular domain of bovine synaptotagmin (Syn)-1, a transmembrane component of synaptic vesicles involved in the regulation of organelle traffic. Since FGF-1 is released in response to heat shock as a mitogenically inactive Cys-30 homodimer, we sought to determine whether this heparin-binding protein was involved in the release of FGF-1. We report that a proteolytic fragment of the extravesicular domain of Syn-1 is associated with FGF-1 in the extracellular compartment of FGF-1-transfected NIH 3T3 cells following temperature stress. By using heparin-Sepharose affinity to discriminate between the monomer and homodimer forms of FGF-1 and resolution by conventional and limited denaturant gel shift immunoblot analysis, it was possible to identify FGF-1 and Syn-1 as potential components of a denaturant- and reducing agent-sensitive extracellular complex. It was also possible to demonstrate that the expression of an antisense-Syn-1 gene represses the release of FGF-1 in response to heat shock. These data indicate that FGF-1 may be able to utilize the cytosolic face of conventional exocytotic vesicles to traffic to the inner surface of the plasma membrane where it may gain access to the extracellular compartment as a complex with Syn-1.


Assuntos
Proteínas de Ligação ao Cálcio , Fatores de Crescimento de Fibroblastos/metabolismo , Resposta ao Choque Térmico , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células 3T3 , Sulfato de Amônio/química , Animais , Sequência de Bases , Encéfalo/metabolismo , Bovinos , Meios de Cultivo Condicionados , Primers do DNA , Dimerização , Fatores de Crescimento de Fibroblastos/química , Heparina/metabolismo , Glicoproteínas de Membrana/química , Camundongos , Proteínas do Tecido Nervoso/química , Oxirredução , Desnaturação Proteica , Sinaptotagmina I , Sinaptotagminas
17.
J Biol Chem ; 273(35): 22224-31, 1998 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-9712836

RESUMO

We have previously characterized the release of the signal peptide sequence-less fibroblast growth factor (FGF) prototype, FGF-1, in vitro as a stress-induced pathway in which FGF-1 is released as a latent homodimer with the p40 extravesicular domain of p65 synaptotagmin (Syn)-1. To determine the biologic relevance of the FGF-1 release pathway in vivo, we sought to resolve and characterize from ovine brain a purified fraction that contained both FGF-1 and p40 Syn-1 and report that the brain-derived FGF-1:p40 Syn-1 aggregate is associated with the calcium-binding protein, S100A13. Since S100A13 binds the anti-inflammatory compound amlexanox and FGF-1 is involved in inflammation, we examined the effects of amlexanox on the release of FGF-1 and p40 Syn-1 in response to stress in vitro. We report that while amlexanox was able to repress the heat shock-induced release of FGF-1 and p40 Syn-1 in a concentration-dependent manner, it had no effect on the constitutive release of p40 Syn-1 from p40 Syn-1 NIH 3T3 cell transfectants. These data suggest the following: (i) FGF-1 is associated with Syn-1 and S100A13 in vivo; (ii) S100A13 may be involved in the regulation of FGF-1 and p40 Syn-1 release in response to temperature stress in vitro; and (iii) the FGF-1 release pathway may be accessible to pharmacologic regulation.


Assuntos
Proteínas de Ligação ao Cálcio , Fatores de Crescimento de Fibroblastos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas S100/metabolismo , Administração Tópica , Aminopiridinas/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Encéfalo/metabolismo , Fatores de Crescimento de Fibroblastos/química , Resposta ao Choque Térmico , Antagonistas dos Receptores Histamínicos H1/farmacologia , Desnaturação Proteica , Ovinos , Sinaptotagmina I , Sinaptotagminas
18.
J Cell Biol ; 141(7): 1647-58, 1998 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-9647656

RESUMO

FGF regulates both cell migration and proliferation by receptor-dependent induction of immediate-early gene expression and tyrosine phosphorylation of intracellular polypeptides. Because little is known about the disparate nature of intracellular signaling pathways, which are able to discriminate between cell migration and proliferation, we used a washout strategy to examine the relationship between immediate-early gene expression and tyrosine phosphorylation with respect to the potential of cells either to migrate or to initiate DNA synthesis in response to FGF-1. We demonstrate that transient exposure to FGF-1 results in a significant decrease in Fos transcript expression and a decrease in tyrosine phosphorylation of the FGFR-1, p42(mapk), and p44(mapk). Consistent with these biochemical effects, we demonstrate that attenuation in the level of DNA synthesis such that a 1.5-h withdrawal is sufficient to return the population to a state similar to quiescence. In contrast, the level of Myc mRNA, the activity of Src, the tyrosine phosphorylation of cortactin, and the FGF-1-induced redistribution of cortactin and F-actin were unaffected by transient FGF-1 stimulation. These biochemical responses are consistent with an implied uncompromised migratory potential of the cells in response to growth factor withdrawal. These results suggest a correlation between Fos expression and the mitogen-activated protein kinase pathway with initiation of DNA synthesis and a correlation between high levels of Myc mRNA and Src kinase activity with the regulation of cell migration.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Divisão Celular , Movimento Celular , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Receptores Proteína Tirosina Quinases , Quinases da Família src/metabolismo , Células 3T3 , Actinas/fisiologia , Animais , Citoesqueleto/fisiologia , DNA/biossíntese , Ativação Enzimática , Fatores de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica , Cinética , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Ornitina Descarboxilase/genética , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-myc/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
20.
Pharmacology ; 55(6): 269-78, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9413855

RESUMO

Interference with growth factor-receptor interactions may have particular relevance in efforts to intervene clinically in both autocrine and paracrine aspects of malignancy. Suramin is a synthetic anticancer agent that works, in part, by blocking the binding of growth factors to their receptors. While initial clinical trials have been encouraging, its use in clinical applications is associated with significant toxicities. Suradista is a novel sulfonated distamycin derivative that is also effective at complexing and inactivating growth factors and cytokines while remaining relatively nontoxic. The goal of this study was to compare the antineoplastic properties of suramin and Suradista. To achieve this, the effects of these compounds on growth factor induced mitogenesis in normal mouse fibroblasts and human umbilical vein endothelial cells were examined, as well as their ability to inhibit the growth of NIH/3T3 cells that had been transformed by the introduction of a fibroblast growth factor (FGF) 1 coding region (residues 1-154) fused to the signal peptide of the hst/KS3 gene (sp-hst/KS3:FGF1-154). In each case, Suradista was more effective than suramin in inhibiting mitogenesis in normal cells, as well as the growth of the transformed cells. Furthermore, Suradista was also shown to be as effective as suramin at inhibiting the growth of sp-hst/KS3:FGF1-154-transformed NIH/3T3 xenografts grown in athymic nude mice when given at only 50% the dosage used for suramin (50 mg/kg for Suradista versus 100 mg/kg for suramin). In summary, these results indicate that novel compounds acting like suramin may be developed as effective antineoplastic agents and may also prove to be of clinical benefit.


Assuntos
Antineoplásicos/farmacologia , Distamicinas/farmacologia , Inibidores do Crescimento/farmacologia , Células 3T3/efeitos dos fármacos , Animais , Antineoplásicos/química , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/patologia , Células Cultivadas , Citocinas/antagonistas & inibidores , DNA/biossíntese , Distamicinas/química , Endotélio Vascular/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos , Camundongos Nus , Mitose/efeitos dos fármacos , Mitose/genética , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Suramina/farmacologia , Transfecção , Veias Umbilicais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...