Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
2.
Growth Factors ; 18(4): 277-85, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11519826

RESUMEN

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.


Asunto(s)
Proteínas de Unión al Calcio , Hipoxia de la Célula/fisiología , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Células 3T3 , Ácido 5,8,11,14-Eicosatetrainoico/farmacología , Aminopiridinas/farmacología , Animales , Ácido Araquidónico/metabolismo , Línea Celular , Inhibidores de la Ciclooxigenasa/farmacología , Cisteína/química , Dimerización , Factor 1 de Crecimiento de Fibroblastos/química , Factor 1 de Crecimiento de Fibroblastos/genética , Humanos , Inhibidores de la Lipooxigenasa/farmacología , Sustancias Macromoleculares , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Nitrobencenos/farmacología , Estrés Fisiológico/fisiopatología , Sulfonamidas/farmacología , Sinaptotagmina I , Sinaptotagminas , Temperatura , Transfección
3.
J Biol Chem ; 276(27): 25549-57, 2001 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-11432880

RESUMEN

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.


Asunto(s)
Cobre/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteínas S100/metabolismo , Estrés Fisiológico/metabolismo , Animales , Sistema Libre de Células , Cisteína/metabolismo , Detergentes/farmacología , Dimerización , Factor 1 de Crecimiento de Fibroblastos , Humanos , Molibdeno/farmacología , Proteínas del Tejido Nervioso/metabolismo , Oxidación-Reducción , Fragmentos de Péptidos/metabolismo , Conejos , Dodecil Sulfato de Sodio/farmacología , Sinaptotagmina I
4.
J Biol Chem ; 276(25): 22544-52, 2001 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-11410600

RESUMEN

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.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Respuesta al Choque Térmico , Proteínas S100 , Células 3T3 , Animales , Factor 1 de Crecimiento de Fibroblastos , Heparina/metabolismo , Ratones , Biosíntesis de Proteínas , Ovinos , Transcripción Genética
5.
J Biol Chem ; 276(34): 32022-30, 2001 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-11427524

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana/fisiología , Proteína Oncogénica pp60(v-src)/metabolismo , Proteínas/fisiología , Células 3T3 , Animales , Proteínas de Unión al Calcio , Cortactina , Péptidos y Proteínas de Señalización Intercelular , Proteína Jagged-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Microfilamentos/metabolismo , Mutagénesis Sitio-Dirigida , Fenotipo , Fosforilación , Proteínas/genética , Proteínas/metabolismo , Receptores Notch , Proteínas Serrate-Jagged , Espectrometría de Fluorescencia , Transfección , Tirosina/metabolismo
6.
J Biol Chem ; 276(7): 5147-51, 2001 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-11087725

RESUMEN

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.


Asunto(s)
Proteínas de Unión al Calcio , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Respuesta al Choque Térmico , Interleucina-1/fisiología , Precursores de Proteínas/fisiología , Células 3T3 , Aminopiridinas/farmacología , Animales , Brefeldino A/farmacología , Desoxiglucosa/farmacología , Factor 1 de Crecimiento de Fibroblastos , Interleucina-1/genética , Interleucina-1/metabolismo , Cinética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Transporte de Proteínas , Eliminación de Secuencia , Sinaptotagmina I , Sinaptotagminas , Transfección
7.
J Biol Chem ; 275(42): 32753-62, 2000 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-10921913

RESUMEN

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.


Asunto(s)
Actinas/fisiología , Aminopiridinas/farmacología , Movimiento Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Endotelio Vascular/fisiología , Genes src , Músculo Liso Vascular/fisiología , Células 3T3 , Actinas/química , Actinas/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Aorta , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Cortactina , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Factor 1 de Crecimiento de Fibroblastos , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/fisiología , Humanos , Células L , Ratones , Proteínas de Microfilamentos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Fosforilación , Ratas , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/fisiología , Esteroides , Transfección , Venas Umbilicales , Xenopus laevis
8.
Biochem Biophys Res Commun ; 268(3): 853-9, 2000 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-10679295

RESUMEN

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.


Asunto(s)
Proteínas/metabolismo , Células 3T3 , Animales , Secuencia de Bases , Proteínas de Unión al Calcio , Comunicación Celular , Diferenciación Celular , División Celular , Cartilla de ADN/genética , Matriz Extracelular/metabolismo , Expresión Génica , Péptidos y Proteínas de Señalización Intercelular , Proteína Jagged-1 , Proteínas de la Membrana , Ratones , Ratones Desnudos , Neovascularización Fisiológica , Fenotipo , Proteínas/química , Proteínas/genética , Proteínas Serrate-Jagged , Transducción de Señal , Transfección
9.
J Biol Chem ; 273(35): 22209-16, 1998 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-9712834

RESUMEN

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.


Asunto(s)
Proteínas de Unión al Calcio , Factores de Crecimiento de Fibroblastos/metabolismo , Respuesta al Choque Térmico , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células 3T3 , Sulfato de Amonio/química , Animales , Secuencia de Bases , Encéfalo/metabolismo , Bovinos , Medios de Cultivo Condicionados , Cartilla de ADN , Dimerización , Factores de Crecimiento de Fibroblastos/química , Heparina/metabolismo , Glicoproteínas de Membrana/química , Ratones , Proteínas del Tejido Nervioso/química , Oxidación-Reducción , Desnaturalización Proteica , Sinaptotagmina I , Sinaptotagminas
10.
J Cell Biol ; 141(7): 1647-58, 1998 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-9647656

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , División Celular , Movimiento Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Proteínas Tirosina Quinasas Receptoras , Familia-src Quinasas/metabolismo , Células 3T3 , Actinas/fisiología , Animales , Citoesqueleto/fisiología , ADN/biosíntesis , Activación Enzimática , Factores de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica , Cinética , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Ornitina Descarboxilasa/genética , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-myc/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
12.
Pharmacology ; 55(6): 269-78, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9413855

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Distamicinas/farmacología , Inhibidores de Crecimiento/farmacología , Células 3T3/efectos de los fármacos , Animales , Antineoplásicos/química , Línea Celular Transformada/efectos de los fármacos , Línea Celular Transformada/patología , Células Cultivadas , Citocinas/antagonistas & inhibidores , ADN/biosíntesis , Distamicinas/química , Endotelio Vascular/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Ratones Desnudos , Mitosis/efectos de los fármacos , Mitosis/genética , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Suramina/farmacología , Transfección , Venas Umbilicales
13.
FEBS Lett ; 414(2): 419-24, 1997 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-9315732

RESUMEN

The tumor promoter phorbol 12-myristic 13-acetate inhibits the growth of human endothelial cells and induces the formation of capillary-like, tubular structures. We report the novel growth regulatory function of the immediate-early gene, edg-3, which is identical to the IkappaB alpha/MAD-3 gene. We employed phosphothioate oligonucleotides (PTO) directed against the translation initiation site of IkappaB alpha to inhibit its expression. The antisense IkappaB alpha PTO-treated cells exhibited an exaggerated growth response to fibroblast growth factor-1 (FGF-1). In contrast, IL-1-induced growth arrest response was not modulated. These data suggest that the early response gene IkappaB alpha is an endogenous regulator of endothelial cell growth in vitro.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Proteínas I-kappa B , Proteínas Inmediatas-Precoces/biosíntesis , División Celular/efectos de los fármacos , Células Cultivadas , Cicloheximida/farmacología , Endotelio Vascular/efectos de los fármacos , Genes Inmediatos-Precoces/efectos de los fármacos , Humanos , Inhibidor NF-kappaB alfa , Oligonucleótidos Antisentido/farmacología , Reacción en Cadena de la Polimerasa , Biosíntesis de Proteínas , Receptores Lisofosfolípidos , Acetato de Tetradecanoilforbol/farmacología , Tionucleótidos , Venas Umbilicales
14.
J Biol Chem ; 271(51): 32499-502, 1996 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-8955070

RESUMEN

Angiogenesis, or the formation of new blood vessels, plays a central role in a number of physiologic and pathologic conditions, including wound healing, diabetic retinopathy, and solid tumor growth, and endothelial cells can be induced to mimic this process in vitro. Using a modification of the differential display method (Zimrin, A. B., Villeponteau, B., and Maciag, T. (1995) Biochem. Biophys. Res. Commun. 213, 630-638), we isolated the human homolog of the Jagged ligand for the Notch receptor from human endothelial cells exposed to fibrin and demonstrate that the Jagged transcript, but not the Notch 1 or Notch 2 transcripts, are up-regulated by fibrin. Interestingly, the addition of an antisense Jagged oligomer to bovine microvascular endothelial cells grown on a collagen gel resulted in a marked increase in invasion and tube formation in the underlying gel in response to fibroblast growth factor. In contrast, no effect was observed on vascular endothelial growth factor-induced angiogenesis under identical conditions. These data suggest that Jagged-Notch signaling is able to regulate fibroblast growth factor-induced endothelial cell migration in vitro, an early event during angiogenesis in vivo.


Asunto(s)
Endotelio Vascular/fisiología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Neovascularización Fisiológica , Receptores de Superficie Celular/genética , Factores de Transcripción , Proteínas de Unión al Calcio , Células Cultivadas , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteína Jagged-1 , Oligonucleótidos Antisentido , ARN Mensajero/genética , Receptor Notch1 , Receptor Notch2 , Proteínas Serrate-Jagged , Transducción de Señal
15.
J Cell Biol ; 134(3): 783-91, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8707855

RESUMEN

Senescent cells do not proliferate in response to exogenous growth factors, yet the number and affinity of growth factor receptors on the cell surface appear to be similar to presenescent cell populations. To determine whether a defect in receptor signaling exists, we analyzed human umbilical vein endothelial cells (HUVEC) since HUVEC growth is absolutely dependent upon the presence of FGF. We report that in both presenescent and senescent HUVEC populations, FGF-1 induces the expression of cell cycle-specific genes, suggesting that functional FGF receptor (FGFR) may exist on the surface of these cells. However, the tyrosine phosphorylation of FGFR-1 substrates, Src and cortactin, is impaired in senescent HUVEC, and only the presenescent cell populations exhibit a FGF-1-dependent Src tyrosine kinase activity. Moreover, we demonstrate that senescent HUVEC are unable to migrate in response to FGF-1, and these data correlate with an altered organization of focal adhesion sites. These data suggest that the induction of gene expression is insufficient to promote a proliferative or migratory phenotype in senescent HUVEC and that the attenuation of the FGFR-1 signal transduction pathway may be involved in the inability of senescent HUVEC to proliferate and/or migrate.


Asunto(s)
Endotelio Vascular/citología , Factor 1 de Crecimiento de Fibroblastos/farmacología , Proteínas Tirosina Quinasas Receptoras , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/fisiología , Tirosina/metabolismo , Secuencia de Bases , Adhesión Celular , Ciclo Celular , División Celular , Movimiento Celular , Células Cultivadas , Senescencia Celular , Cortactina , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Microfilamentos/metabolismo , Datos de Secuencia Molecular , Fosforilación , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Venas Umbilicales
16.
J Biol Chem ; 271(24): 14198-205, 1996 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-8662999

RESUMEN

The alternatively spliced fibroblast growth factor receptor (FGFR)-1 isoforms, FGFR-1alpha and FGFR-1beta, are characterized by the presence of either three or two Ig-like loops in the extracellular domain and are differentially expressed during embryonic development and tumor progression. We have previously shown that in cells irreversibly committed to DNA synthesis by FGF-1, approximately 15% of cell surface FGFR-1 traffics to a perinuclear locale as a structurally intact and functional tyrosine kinase (Prudovsky, I., Savion, N., Zhan, X., Friesel, R., Xu, J., Hou, J., McKeehan, W. L., and Maciag, T. (1994) J. Biol. Chem. 269, 31720-31724). In order to define the structural requirement for association of FGFR-1 with the nucleus, the expression and trafficking of FGFR-1 in FGFR-1alpha and FGFR-1beta L6 myoblast transfectants was studied. Although FGFR-1alpha was expressed as p145 and p125 forms, FGFR-1beta was expressed as p120 and p100 forms in the L6 myoblast transfectants. Tunicamycin and N-glyconase experiments suggest that these forms of FGFR-1alpha and FGFR-1beta are the result of differential glycosylation. However, only the p145 form of FGFR-1alpha and the p120 form of FGFR-1beta were able to bind FGF-1 and activate tyrosine phosphorylation. Pulse-chase analysis of FGFR-1 biosynthesis suggests that the p125 and p100 proteins are the precursor forms of p145 FGFR-1alpha and p120 FGFR-1beta, respectively. Because ligand-chase analysis demonstrated that FGFR-1beta L6 myoblast transfectants exhibited a reduced efficiency of nuclear translocation of exogenous FGF-1 when compared with FGFR-1alpha transfectants, the intracellular trafficking of the FGFR-1alpha and FGFR-1beta isoforms was studied using an in vitro kinase assay to amplify immunoprecipitated FGFR-1. Indeed, the appearance of the FGFR-1alpha but not FGFR-1beta isoform in the nuclear fraction of L6 myoblast transfectants suggests that the distal Ig-like loop in FGFR-1alpha mediates the differential nuclear association of FGFR-1alpha as a structurally intact and functional tyrosine kinase. Further, the FGFR-1beta L6 myoblast transfectants but not the FGFR-1alpha myoblast transfectants exhibited a pronounced morphologic change in response to exogenous FGF-1. Because this phenotype change involves the induction of a rounded cellular shape, it is possible that the FGFR-1alpha and FGFR-1beta may ultimately exhibit differential trafficking to adhesion sites.


Asunto(s)
Empalme Alternativo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Autorradiografía , Línea Celular , Núcleo Celular/metabolismo , Expresión Génica , Glicosilación , Immunoblotting , Cinética , Ligandos , Metionina/metabolismo , Peso Molecular , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/biosíntesis , Receptores de Factores de Crecimiento de Fibroblastos/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Transducción de Señal , Radioisótopos de Azufre , Transfección , Tunicamicina/farmacología
17.
Biochim Biophys Acta ; 1312(1): 27-38, 1996 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-8679713

RESUMEN

Autocrine/paracrine stimulation of cell growth by members of the fibroblast growth factor (FGF) family of polypeptides is dependent upon extracellular interactions with specific high affinity receptors at the cell surface. Acidic FGF (FGF-1) lacks a classical signal sequence for secretion, suggesting that intrinsic levels of this mitogen may not stimulate cell growth and utilizes a non-classical pathway to gain access to the extracellular compartment. To evaluate the biological potential of intracellular FGF-1 more rigorously, human cDNA sequences for the growth factor were introduced into primary murine embryonic fibroblasts using retrovirally mediated gene transfer. Heparin affinity, Western analysis, mitogenic assays, in situ immunohistochemical techniques, induction of tyrosine phosphorylation and antibody inhibition studies were used to demonstrate functionality of the FGF-1 transgene in this experimental model. Under normal culture conditions, cells constitutively expressing intracellular FGF-1 exhibited a slight growth advantage. In contrast, when maintained in reduced serum, these cells adopted a transformed phenotype and demonstrated an enhanced growth potential, induction of FGF-specific phosphotyrosyl proteins and the nuclear association of the growth factor. Analysis of the conditioned media from these stressed cells indicated that serum starvation induces the secretion of FGF-1 as latent high molecular mass complexes requiring reducing agents to activate its full biological potential.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/biosíntesis , Fibroblastos/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sangre , División Celular , Células Cultivadas , Cortactina , Medios de Cultivo Condicionados/química , ADN Complementario , Factor 1 de Crecimiento de Fibroblastos/análisis , Factor 1 de Crecimiento de Fibroblastos/química , Factor 1 de Crecimiento de Fibroblastos/genética , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/química , Fibroblastos/citología , Técnicas de Transferencia de Gen , Humanos , Ratones , Proteínas de Microfilamentos/análisis , Mitógenos , Datos de Secuencia Molecular , Peso Molecular , Fosforilación , Proteínas Proto-Oncogénicas pp60(c-src)/análisis , Tirosina/metabolismo
18.
J Biol Chem ; 270(49): 29039-42, 1995 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-7493920

RESUMEN

Fibroblast growth factor (FGF)-1 lacks a classical signal sequence to direct its secretion yet utilizes high affinity cell surface receptors to signal its heparin-dependent angiogenic and neurotrophic activities. We have previously reported that FGF-1 is released in response to temperature stress as a latent homodimer through a pathway that is potentiated by the Golgi inhibitor, brefeldin A (Jackson, A., Tarantini, F., Gamble, S., Friedman, S., and Maciag, T. (1995) J. Biol. Chem. 270, 33-36). In an attempt to further characterize this unconventional secretion mechanism, we sought to define the Cys residue(s) critical for FGF-1 dimer formation and release and to determine whether FGF-1 can associate with known phospholipid components of organelle or plasma membranes, which may be disturbed by brefeldin A. Utilizing FGF-1 Cys mutants, we were able to demonstrate that residue Cys30 is critical for FGF-1 release in response to heat shock. In addition, using solid phase phospholipid binding assays we demonstrate that FGF-1 is able to specifically associate with phosphatidylserine (PS). Heparin inhibits the association between FGF-1 and PS, and synthetic peptide competition assays suggest that the PS-binding domain of FGF-1 lies between residues 114 and 137. These observations indicate that FGF-1 may be able to associate with the PS component of organelle and/or plasma membranes and that the domains responsible for FGF-1 homodimer formation and PS binding are structurally distinct.


Asunto(s)
Cisteína/metabolismo , Factor 1 de Crecimiento de Fibroblastos/química , Fosfatidilserinas/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Transfección
19.
Am J Physiol ; 269(5 Pt 2): F653-62, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7503231

RESUMEN

The proximal tubule epithelium regenerates following nephrotoxic damage. To determine the role of fibroblast growth factors (FGFs) in the regeneration of rat proximal tubule epithelial (RPTE) cells, we investigated proliferation, differentiation, and FGF-1 expression in vivo in rat kidney before and after nephrotoxic damage to the proximal tubule epithelium caused by S-(1,1,2,2-tetrafluoroethyl)-L-cysteine administration. In undamaged kidneys, FGF-1 was expressed in distal tubule elements, including cortical and medullary collecting ducts, as well as in blood vessels and glomeruli, but was absent in RPTE. One day after damage, there was an increase in proliferation of surviving proximal tubule epithelial cells and a coincident increase in FGF-1 expression in invading mononuclear cells. After this initial burst of proliferation, FGF-1 expression increased in poorly differentiated vimentin-positive regenerative epithelial cells, indicating that autocrine FGF-1 expression in the regenerative epithelium is a later event in the regeneration process. FGF-1 staining persisted in foci of macrophages, interstitial cells, and nephropathic tubules within areas of interstitial expansion 2 wk after damage. We concluded that transient paracrine and autocrine expression of FGF-1 could play mitogenic and/or morphogenic roles during tubular regeneration. Persistent expression in macrophages, fibroblasts, and nephropathic tubules may be associated with tubular degeneration. FGF-1 expression may be an important contributor to both tubular regeneration and degenerative disease following toxicant exposure.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Túbulos Renales Proximales/fisiología , Regeneración , Animales , Diferenciación Celular , División Celular , Cisteína/análogos & derivados , Células Epiteliales , Epitelio/efectos de los fármacos , Epitelio/fisiología , Hidrocarburos Fluorados , Inmunohistoquímica , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Masculino , Ratas , Ratas Endogámicas F344 , Factores de Tiempo , Vimentina/metabolismo
20.
Biochem Biophys Res Commun ; 213(2): 630-8, 1995 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-7544124

RESUMEN

Endothelial cells can be induced to form a branching network of tubular structures using a variety of cell culture conditions. We have examined this differentiation process using several sets of conditions: plating human umbilical vein endothelial cells (HUVEC) on Matrigel, adding collagen to the apical surface of HUVEC grown on fibronectin, and plating HUVEC on fibrin in the presence of FGF-1. We determined that although the first two conditions produce dramatic morphologic changes in the HUVEC population, gene transcription and translation are not required for the regulation of the process. Rather, post-translational events are involved since the Matrigel-dependent process could be inhibited by the addition of nocodazole, suramin or H7, a protein kinase inhibitor. In contrast, the fibrin matrix-dependent differentiation pathway involved transcriptional and translational events since the addition of either actinomycin D or cycloheximide inhibited this pathway. A modified differential display of RNA extracted from HUVEC after 0, 2, 5, and 24 hours on fibrin revealed expression of a novel cDNA.


Asunto(s)
Diferenciación Celular , Colágeno , Endotelio Vascular/citología , Fibrina , Laminina , Modelos Biológicos , Neovascularización Patológica , Proteoglicanos , Secuencia de Bases , Células Cultivadas , Medios de Cultivo , Cicloheximida/farmacología , Dactinomicina/farmacología , Combinación de Medicamentos , Endotelio Vascular/efectos de los fármacos , Factor 1 de Crecimiento de Fibroblastos/farmacología , Humanos , Datos de Secuencia Molecular , Transcripción Genética , Venas Umbilicales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA