Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 19(11): 4863-74, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18784255

RESUMO

Angiogenesis is controlled by several regulatory mechanisms, including the Notch and fibroblast growth factor (FGF) signaling pathways. FGF1, a prototype member of FGF family, lacks a signal peptide and is released through an endoplasmic reticulum-Golgi-independent mechanism. A soluble extracellular domain of the Notch ligand Jagged1 (sJ1) inhibits Notch signaling and induces FGF1 release. Thrombin, a key protease of the blood coagulation cascade and a potent inducer of angiogenesis, stimulates rapid FGF1 release through a mechanism dependent on the major thrombin receptor protease-activated receptor (PAR) 1. This study demonstrates that thrombin cleaves Jagged1 in its extracellular domain. The sJ1 form produced as a result of thrombin cleavage inhibits Notch-mediated CBF1/Suppressor of Hairless [(Su(H)]/Lag-1-dependent transcription and induces FGF1 expression and release. The overexpression of Jagged1 in PAR1 null cells results in a rapid thrombin-induced export of FGF1. These data demonstrate the existence of novel cross-talk between thrombin, FGF, and Notch signaling pathways, which play important roles in vascular formation and remodeling.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Fármacos Cardiovasculares/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/metabolismo , Trombina/farmacologia , Animais , Proteínas de Ligação ao Cálcio/química , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fator 1 de Crescimento de Fibroblastos/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Proteína Jagged-1 , Proteínas de Membrana/química , Camundongos , Peso Molecular , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/metabolismo , Crista Neural/citologia , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Receptor PAR-1/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores Notch/metabolismo , Receptores de Trombina , Proteínas Serrate-Jagged , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
2.
Stem Cells ; 25(12): 3133-42, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17761753

RESUMO

The activation of Notch signaling in neural crest stem cells (NCSCs) results in the rapid loss of neurogenic potential and differentiation into glia. We now show that the attenuation of endogenous Notch signaling within expanding NCSC clones by the Notch ligand soluble Jagged1 (sJ1), maintains NCSCs in a clonal self-renewing state in vitro without affecting their sensitivity to instructive differentiation signals observed previously during NCSC self-renewal. sJ1 functions as a competitive inhibitor of Notch signaling to modulate endogenous cell-cell communication to levels sufficient to inhibit neural differentiation but insufficient to instruct gliogenic differentiation. Attenuated Notch signaling promotes the induction and nonclassic release of fibroblast growth factor 1 (FGF1). The functions of sJ1 and FGF1 signaling are complementary, as abrogation of FGF signaling diminishes the ability of sJ1 to promote NCSC expansion, yet the secondary NCSCs maintain the dosage sensitivity of the founder. These results validate and build upon previous studies on the role of Notch signaling in stem cell self-renewal and suggest that the differentiation bias or self-renewal potential of NCSCs is intrinsically linked to the level of endogenous Notch signaling. This should provide a unique opportunity for the expansion of NCSCs ex vivo without altering their differentiation bias for clinical cell replacement or transplant strategies in tissue repair. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Diferenciação Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Proteínas de Membrana/fisiologia , Crista Neural/citologia , Crista Neural/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Animais , Proteínas de Ligação ao Cálcio/genética , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Células Clonais/citologia , Células Clonais/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Proteínas de Membrana/genética , Inibição Neural/genética , Inibição Neural/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Ratos , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/genética , Receptor Notch1/fisiologia , Proteínas Serrate-Jagged , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Solubilidade , Células-Tronco/metabolismo
3.
Exp Cell Res ; 313(15): 3308-18, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17643421

RESUMO

Sphingosine kinase 1 catalyzes the formation of sphingosine-1-phosphate, a lipid mediator involved in the regulation of angiogenesis. Sphingosine kinase 1 is constitutively released from cells, even though it lacks a classical signal peptide sequence. Because copper-dependent non-classical stress-induced release of FGF1 also regulates angiogenesis, we questioned whether sphingosine kinase 1 is involved in the FGF1 release pathway. We report that (i) the coexpression of sphingosine kinase 1 with FGF1 inhibited the release of sphingosine kinase 1 at 37 degrees C; (ii) sphingosine kinase 1 was released at 42 degrees C in complex with FGF1; (iii) sphingosine kinase 1 null cells failed to release FGF1 at stress; (iv) sphingosine kinase 1 is a high affinity copper-binding protein which formed a complex with FGF1 in a cell-free system, and (v) sphingosine kinase 1 over expression rescued the release of FGF1 from inhibition by the copper chelator, tetrathiomolybdate. We propose that sphingosine kinase 1 is a component of the copper-dependent FGF1 release pathway.


Assuntos
Cobre/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Células Cultivadas , Quelantes/farmacologia , Clonagem Molecular , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Molibdênio/farmacologia , Células NIH 3T3 , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Transporte Proteico , Temperatura
4.
Biochem Biophys Res Commun ; 350(3): 604-9, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-17027650

RESUMO

Thrombin induces cell proliferation and migration during vascular injury. We report that thrombin rapidly stimulated expression and release of the pro-angiogenic polypeptide fibroblast growth factor 1 (FGF1). Thrombin failed to induce FGF1 release from protease-activated receptor 1 (PAR1) null fibroblasts, indicating that this effect was dependent on PAR1. Similarly to thrombin, FGF1 expression and release were induced by TRAP, a specific oligopeptide agonist of PAR1. These results identify a novel aspect of the crosstalk between FGF and thrombin signaling pathways which both play important roles in tissue repair and angiogenesis.


Assuntos
Fator 1 de Crescimento de Fibroblastos/metabolismo , Receptor PAR-1/metabolismo , Transdução de Sinais/fisiologia , Trombina/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Camundongos , Células NIH 3T3 , Transdução de Sinais/efeitos dos fármacos
5.
Biochem Biophys Res Commun ; 349(1): 192-9, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16930531

RESUMO

Fibroblast growth factor (FGF)1 is released from cells as a constituent of a complex that contains the small calcium binding protein S100A13, and the p40 kDa form of synaptotagmin (Syt)1, through an ER-Golgi-independent stress-induced pathway. FGF1 and the other components of its secretory complex are signal peptide-less proteins. We examined their capability to interact with lipid bilayers by studying protein-induced carboxyfluorescein release from liposomes of different phospholipid (pL) compositions. FGF1, p40 Syt1, and S100A13 induced destabilization of liposomes composed of acidic but not of zwitterionic pL. We produced mutants of FGF1 and p40 Syt1, in which specific basic amino acid residues in the regions that bind acidic pL were substituted. The ability of these mutants to induce liposomes destabilization was strongly attenuated, and they exhibited drastically diminished spontaneous and stress-induced release. Apparently, the non-classical release of FGF1 and p40 Syt1 involves destabilization of membranes containing acidic pL.


Assuntos
Fator 1 de Crescimento de Fibroblastos/química , Sinaptotagmina I/química , Animais , Membrana Celular/metabolismo , Complexo de Golgi/metabolismo , Bicamadas Lipídicas/química , Lipossomos/química , Camundongos , Modelos Biológicos , Células NIH 3T3 , Proteínas Recombinantes/química
6.
FEBS Lett ; 579(25): 5798-5802, 2005 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-16225865

RESUMO

Notch signaling involves proteolytic cleavage of the transmembrane Notch receptor after binding to its transmembrane ligands, Delta or Jagged; and the resultant soluble intracellular domain of Notch stimulates a cascade of transcriptional events. The Delta1 ligand also undergoes proteolytic cleavage upon Notch binding, resulting in the production of a free intracellular domain. We demonstrate that the expression of the intracellular domain of Delta1 results in a non-proliferating senescent-like cell phenotype which is dependent on the expression of the cell cycle inhibitor, p21, and is abolished by co-expression of constitutively active Notch1. These data suggest a new intracellular role for Delta1.


Assuntos
Proliferação de Células , Proteínas de Membrana/metabolismo , Animais , Senescência Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Células NIH 3T3 , Estrutura Terciária de Proteína
7.
Langmuir ; 20(21): 8961-5, 2004 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-15461473

RESUMO

Structural deformations of lipid hybrid bilayer membranes induced by signal peptideless (SPL) proteins have been studied for the first time using the inherently surface specific nonlinear optical technique of sum frequency generation vibrational spectroscopy. Specifically, deformations of 1,2-distearoylphosphatidylglycerol(DSPG) membranes induced by interaction with FGF-1, a SPL protein which is released asa function of cellular stress through a nonclassical pathway, have been investigated. FGF-1 was found to induce lipid alkyl chain deformations in previously highly ordered DSPG membranes at the extremely low concentration of 1 nM at 60 degrees C. The deformation process was shown to exhibit a degree of reversibility upon removal of the protein by rinsing with buffer solution.


Assuntos
Fator 1 de Crescimento de Fibroblastos/química , Bicamadas Lipídicas/química , Membranas Artificiais , Fosfatidilgliceróis/química , Conformação Proteica , Espectrofotometria Infravermelho/métodos , Propriedades de Superfície , Vibração
8.
J Biol Chem ; 279(14): 13285-8, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-14769803

RESUMO

The interactions between Notch (N) receptors and their transmembrane ligands, Jagged1 (JI) and Delta1 (Dl1), mediate signaling events between neighboring cells that are crucial during embryonal development and in adults. Since the non-transmembrane extracellular form of J1 acts as an antagonist of N activation in NIH 3T3 mouse fibroblast cells and induces fibroblast growth factor 1 (FGF1)-dependent transformation (Small, D., Kovalenko, D., Soldi, R., Mandinova, A., Kolev, V., Trifonova, R., Bagala, C., Kacer, D., Battelli, C., Liaw, L., Prudovsky, I., and Maciag, T. (2003) J. Biol. Chem. 278, 16405-16413), we examined the potential redundant functions of the two subfamilies of Notch ligands and report that while the soluble (s) forms of both Dl1 and J1 act as N signaling antagonists in NIH 3T3 cells, they do display disparate functions. While sJ1 induced an attenuation of cell motility which is accompanied by a decrease in actin stress fibers and an increase in adherence junctions, sDl1 does not. However, sJ1, like sDl1, induces a NIH 3T3 cell tranformed phenotype mediated by FGF signaling. Because the inhibition of N signaling by sJ1 and sDl1 is rescued by dominant-negative Src expression, we suggest that there may be cooperation between the Notch and Src signaling pathways.


Assuntos
Movimento Celular/fisiologia , Proteínas de Membrana/metabolismo , Proteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Adesões Focais/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Proteína Jagged-1 , Proteínas de Membrana/genética , Camundongos , Células NIH 3T3 , Fenótipo , Proteínas/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores Notch , Proteínas Serrate-Jagged , Transdução de Sinais/fisiologia , Fibras de Estresse/fisiologia , Transfecção
9.
Blood ; 103(6): 2114-20, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-14630795

RESUMO

Endothelial cell adhesive interactions are mediated by both fibrinogen and fibrin, and growth is stimulated by fibroblast growth factor 2 (FGF-2). We have shown previously that FGF-2 binds specifically and with high affinity to fibrinogen and fibrin and that fibrinogen potentiates the proliferative capacity of FGF-2 and also protects it from proteolytic degradation. To further characterize this interaction we have performed FGF-2 mutagenesis to identify the interactive site. Because FGF-1 has a similar structure to FGF-2 but does not bind to fibrinogen, we used a strategy of cassette and site-directed mutagenesis, exchanging residues from FGF-1 and FGF-2 and correlating structural changes with fibrinogen binding. Two cassette interchange mutants, 2212 and 2211, contained either the third cassette or both the third and fourth cassettes from FGF-1, and neither exhibited any affinity for fibrinogen. Exchange of 5 residues (Phe95, Ser100, Asn102, Arg107, and Arg109) from FGF-2 into the corresponding sites in the third cassette of FGF-1 imparted high-affinity binding with apparent dissociation constants (Kd) of 5.3 nM and 8.6 nM, respectively, compared with 1.3 nM for wild-type FGF-2. We conclude that these 5 residues define a high-affinity binding site in FGF-2 for fibrinogen.


Assuntos
Fibrinogênio/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/química , Humanos , Dados de Sequência Molecular , Mutagênese , Estrutura Terciária de Proteína
10.
J Cell Sci ; 116(Pt 24): 4871-81, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14625381

RESUMO

Non-classical protein release independent of the ER-Golgi pathway has been reported for an increasing number of proteins lacking an N-terminal signal sequence. The export of FGF1 and IL-1alpha, two pro-angiogenic polypeptides, provides two such examples. In both cases, export is based on the Cu2+-dependent formation of multiprotein complexes containing the S100A13 protein and might involve translocation of the protein across the membrane as a 'molten globule'. FGF1 and IL-1alpha are involved in pathological processes such as restenosis and tumor formation. Inhibition of their export by Cu2+ chelators is thus an effective strategy for treatment of several diseases.


Assuntos
Retículo Endoplasmático/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Interleucina-1/metabolismo , Sinais Direcionadores de Proteínas/fisiologia , Animais , Hipóxia Celular/fisiologia , Membrana Celular/metabolismo , Cobre/metabolismo , Humanos , Fluidez de Membrana/fisiologia , Modelos Estruturais , Neovascularização Patológica/metabolismo , Transporte Proteico , Proteínas S100
11.
Cell ; 115(2): 163-75, 2003 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-14567914

RESUMO

Axon-derived molecules are temporally and spatially required as positive or negative signals to coordinate oligodendrocyte differentiation. Increasing evidence suggests that, in addition to the inhibitory Jagged1/Notch1 signaling cascade, other pathways act via Notch to mediate oligodendrocyte differentiation. The GPI-linked neural cell recognition molecule F3/contactin is clustered during development at the paranodal region, a vital site for axoglial interaction. Here, we show that F3/contactin acts as a functional ligand of Notch. This trans-extracellular interaction triggers gamma-secretase-dependent nuclear translocation of the Notch intracellular domain. F3/Notch signaling promotes oligodendrocyte precursor cell differentiation and upregulates the myelin-related protein MAG in OLN-93 cells. This can be blocked by dominant negative Notch1, Notch2, and two Deltex1 mutants lacking the RING-H2 finger motif, but not by dominant-negative RBP-J or Hes1 antisense oligonucleotides. Expression of constitutively active Notch1 or Notch2 does not upregulate MAG. Thus, F3/contactin specifically initiates a Notch/Deltex1 signaling pathway that promotes oligodendrocyte maturation and myelination.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas de Membrana/metabolismo , Oligodendroglia/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Células CHO , Moléculas de Adesão Celular Neuronais/farmacologia , Diferenciação Celular , Técnicas de Cocultura , Contactinas , Cricetinae , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Células HeLa , Proteínas de Homeodomínio/metabolismo , Humanos , Ligantes , Proteínas de Membrana/genética , Modelos Biológicos , Mutação , Glicoproteína Associada a Mielina/metabolismo , Oligodendroglia/citologia , Ratos , Receptores de Superfície Celular/metabolismo , Receptores Notch , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1 , Ativação Transcricional , Ubiquitina-Proteína Ligases , Regulação para Cima
13.
J Cell Sci ; 116(Pt 13): 2687-96, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12746488

RESUMO

Copper is involved in the promotion of angiogenic and inflammatory events in vivo and, although recent clinical data has demonstrated the potential of Cu2+ chelators for the treatment of cancer in man, the mechanism for this activity remains unknown. We have previously demonstrated that the signal peptide-less angiogenic polypeptide, FGF1, uses intracellular Cu2+ to facilitate the formation of a multiprotein aggregate that enables the release of FGF1 in response to stress and that the expression of the precursor form but not the mature form of IL-1alpha represses the stress-induced export of FGF1 from NIH 3T3 cells. We report here that IL-1alpha is a Cu2+-binding protein and human U937 cells, like NIH 3T3 cells, release IL-1alpha in response to temperature stress in a Cu2+-dependent manner. We also report that the stress-induced export of IL-1alpha involves the intracellular association with the Cu2+-binding protein, S100A13. In addition, the expression of a S100A13 mutant lacking a sequence novel to this gene product functions as a dominant-negative repressor of IL-1alpha release, whereas the expression of wild-type S100A13 functions to eliminate the requirement for stress-induced transcription. Lastly, we present biophysical evidence that IL-1alpha may be endowed with molten globule character, which may facilitate its release through the plasma membrane. Because Cu2+ chelation also represses the release of FGF1, the ability of Cu2+ chelators to potentially serve as effective clinical anti-cancer agents may be related to their ability to limit the export of these proinflammatory and angiogenic signal peptide-less polypeptides into the extracellular compartment.


Assuntos
Cobre/metabolismo , Interleucina-1/metabolismo , Neovascularização Patológica/metabolismo , Proteínas S100/fisiologia , Estresse Fisiológico/metabolismo , Animais , Antineoplásicos/farmacologia , Quelantes/farmacologia , Cobre/farmacologia , Citosol/metabolismo , Líquido Extracelular/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Humanos , Camundongos , Mutação/genética , Células NIH 3T3 , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/fisiopatologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas S100/deficiência , Proteínas S100/genética , Estresse Fisiológico/fisiopatologia , Temperatura , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Células U937
14.
Proc Natl Acad Sci U S A ; 100(11): 6700-5, 2003 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-12754378

RESUMO

The induction of an acute inflammatory response followed by the release of polypeptide cytokines and growth factors from peripheral blood monocytes has been implicated in mediating the response to vascular injury. Because the Cu2+-binding proteins IL-1alpha and fibroblast growth factor 1 are exported into the extracellular compartment in a stress-dependent manner by using intracellular Cu2+ to facilitate the formation of S100A13 heterotetrameric complexes and these signal peptideless polypeptides have been implicated as regulators of vascular injury in vivo, we examined the ability of Cu2+ chelation to repress neointimal thickening in response to injury. We observed that the oral administration of the Cu2+ chelator tetrathiomolybdate was able to reduce neointimal thickening after balloon injury in the rat. Interestingly, although immunohistochemical analysis of control neointimal sections exhibited prominent staining for MAC1, IL-1alpha, S100A13, and the acidic phospholipid phosphatidylserine, similar sections obtained from tetrathiomolybdate-treated animals did not. Further, adenoviral gene transfer of the IL-1 receptor antagonist during vascular injury also significantly reduced the area of neointimal thickening. Our data suggest that intracellular copper may be involved in mediating the response to injury in vivo by its ability to regulate the stress-induced release of IL-1alpha by using the nonclassical export mechanism employed by human peripheral blood mononuclear cells in vitro.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Quelantes/farmacologia , Cobre/química , Molibdênio/farmacologia , Adenoviridae/genética , Animais , Vasos Sanguíneos/lesões , Vasos Sanguíneos/metabolismo , Ceruloplasmina/metabolismo , Quelantes/química , Humanos , Imuno-Histoquímica , Masculino , Molibdênio/química , Ratos , Ratos Sprague-Dawley , Transfecção
15.
J Biol Chem ; 278(18): 16405-13, 2003 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-12598523

RESUMO

Aberrant activations of the Notch and fibroblast growth factor receptor (FGFR) signaling pathways have been correlated with neoplastic growth in humans and other mammals. Here we report that the suppression of Notch signaling in NIH 3T3 cells by the expression of either the extracellular domain of the Notch ligand Jagged1 or dominant-negative forms of Notch1 and Notch2 results in the appearance of an exaggerated fibroblast growth factor (FGF)-dependent transformed phenotype characterized by anchorage-independent growth in soft agar. Anchorage-independent growth exhibited by Notch-repressed NIH 3T3 cells may result from prolonged FGFR stimulation caused by both an increase in the expression of prototypic and oncogenic FGF gene family members and the nonclassical export of FGF1 into the extracellular compartment. Interestingly, FGF exerts a negative effect on Notch by suppressing CSL (CBF-1/RBP-Jk/KBF2 in mammals, Su(H) in Drosophila and Xenopus, and Lag-2 in Caenorhabditis elegans)-dependent transcription, and the ectopic expression of constitutively active forms of Notch1 or Notch2 abrogates FGF1 release and the phenotypic effects of FGFR stimulation. These data suggest that communication between the Notch and FGFR pathways may represent an important reciprocal autoregulatory mechanism for the regulation of normal cell growth.


Assuntos
Transformação Celular Neoplásica , Fator 1 de Crescimento de Fibroblastos/fisiologia , Proteínas de Membrana/fisiologia , Proteínas , Células 3T3 , Animais , Proteínas de Ligação ao Cálcio , Divisão Celular , Proteínas de Drosophila , Fator 1 de Crescimento de Fibroblastos/genética , Fibroblastos/citologia , Peptídeos e Proteínas de Sinalização Intercelular , Proteína Jagged-1 , Glicoproteínas de Membrana/fisiologia , Camundongos , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Receptores Notch , Proteínas Serrate-Jagged , Proteínas de Xenopus
16.
J Cell Biol ; 158(2): 201-8, 2002 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-12135982

RESUMO

The release of signal peptideless proteins occurs through nonclassical export pathways and the release of fibroblast growth factor (FGF)1 in response to cellular stress is well documented. Although biochemical evidence suggests that the formation of a multiprotein complex containing S100A13 and Synaptotagmin (Syt)1 is important for the release of FGF1, it is unclear where this intracellular complex is assembled. As a result, we employed real-time analysis using confocal fluorescence microscopy to study the spatio-temporal aspects of this nonclassical export pathway and demonstrate that heat shock stimulates the redistribution of FGF1 from a diffuse cytosolic pattern to a locale near the inner surface of the plasma membrane where it colocalized with S100A13 and Syt1. In addition, coexpression of dominant-negative mutant forms of S100A13 and Syt1, which both repress the release of FGF1, failed to inhibit the stress-induced peripheral redistribution of intracellular FGF1. However, amlexanox, a compound that is known to attenuate actin stress fiber formation and FGF1 release, was able to repress this process. These data suggest that the assembly of the intracellular complex involved in the release of FGF1 occurs near the inner surface of the plasma membrane and is dependent on the F-actin cytoskeleton.


Assuntos
Proteínas de Ligação ao Cálcio , Fator 1 de Crescimento de Fibroblastos , Células 3T3 , Animais , Fator 1 de Crescimento de Fibroblastos/química , Fator 1 de Crescimento de Fibroblastos/metabolismo , Membranas Intracelulares/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Camundongos , Microscopia Confocal , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Transporte Proteico , Proteínas S100/química , Proteínas S100/metabolismo , Sinaptotagmina I , Sinaptotagminas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...