Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 239
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Adv Biol Regul ; 53(1): 87-96, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23266087

RESUMEN

Receptor tyrosine kinases (RTKs) function through protein kinase entities located in the intracellular domain of each protomer. Following activation by ligand binding, they selectively form phosphotyrosine residues by autocatalytic modification. Some of these sites are involved in maintaining the active conformation of the kinase, while others become docking sites for various adaptor/effector/scaffold proteins, which, after complexing with the receptor, then initiate further responses through cascades of post-translational modifications and the generation of lipid second messengers. Although there is substantial overlap in the pathways and activities stimulated by this superfamily, the molecular features of the endodomains of the sub-families and the moieties that they interact with to perpetrate their signals are surprisingly distinct, which may play a significant role in the regulation and responses of the individual RTK types. Some use large scaffold proteins as the basis for most, if not all, of their signal-generating interactions, while others have numerous receptor endodomain phosphotyrosine sites that are quite overlapping in specificity. The members of the Trk family of receptors each have several tyrosine residues that are phosphorylated following stimulation, including those in the kinase activation loop, but there are only two established sites (Y490 and Y785 on TrkA) that are known to be directly involved in signal propagation. Taking advantage of this limited repertoire of docking sites, we have applied phosphoproteomic methods to dissect the signaling responses of both the native protein and derivatives that have had these two sites modified. Interestingly, a clear subset that was not dependent on either docking site was identified. A comparison with a similar set of data for EGFR indicates a considerable degree of similarity in the downstream signaling profile between these two RTKs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Procesamiento Proteico-Postraduccional , Receptor trkA/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Regulación de la Expresión Génica , Humanos , Ligandos , Fosfoproteínas/genética , Fosforilación , Fosfotirosina/genética , Regiones Promotoras Genéticas , Unión Proteica , Proteoma , Receptor trkA/genética
2.
Mol Cell Proteomics ; 3(5): 429-40, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14752058

RESUMEN

Protein sulfonation on serine and threonine residues is described for the first time. This post-translational modification is shown to occur in proteins isolated from organisms representing a broad span of eukaryote evolution, including the invertebrate mollusk Lymnaea stagnalis, the unicellular malaria parasite Plasmodium falciparum, and humans. Detection and structural characterization of this novel post-translational modification was carried out using liquid chromatography coupled to electrospray tandem mass spectrometry on proteins including a neuronal intermediate filament and a myosin light chain from the snail, a cathepsin-C-like enzyme from the parasite, and the cytoplasmic domain of the human orphan receptor tyrosine kinase Ror-2. These findings suggest that sulfonation of serine and threonine may be involved in multiple functions including protein assembly and signal transduction.


Asunto(s)
Cadenas Ligeras de Miosina/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Proteínas Protozoarias/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Cromatografía Liquida , Clonación Molecular , Humanos , Lymnaea/metabolismo , Espectrometría de Masas , Péptidos/metabolismo , Plasmodium falciparum/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa , Serina/metabolismo , Treonina/metabolismo
3.
J Biol Chem ; 276(40): 37529-36, 2001 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-11459840

RESUMEN

To assess the contribution of the intracellular domain tyrosine residues to the signaling capacity of fibroblast growth factor receptor 1 (FGFR1), stably transfected chimeras bearing the ectodomain of the platelet-derived growth factor receptor (PDGFR) and the endodomain of FGFR1 were systematically altered by a tyrosine to phenylalanine bloc and individual conversions. The 15 tyrosine residues of the endodomain of this construct (PFR1) were divided into four linear segments (labeled A, B, C, and D) that contained 4, 4, 2, and 5 tyrosine residues, respectively. When stimulated by platelet-derived growth factor, derivatives in which the A, B, or A + B blocs of tyrosines were mutated were about two-thirds as active as the unmodified chimera at 48 h but achieved full activity by 96 h in a neurite outgrowth assay in transfected PC12 cells. Elimination of only the two activation loop tyrosines (C bloc) also inactivated the receptor. All derivatives in which 4 (or 5) of the D bloc tyrosines were mutated were inactive in producing differentiation but showed low levels of kinase activity in in vitro assays. Derivatives in which 1, 2, or 3 tyrosines of the D bloc in different combinations were systematically changed demonstrated that 2 residues (Tyr(677) and Tyr(701), using hFGFR1 numbering) were essential for bioactivity, but the remaining 3 residues, including Tyr(766), the previously identified site for phospholipase C gamma (PLC gamma) activation, were not. Differentiation activity was paralleled by the activation (phosphorylation) of FRS2, SOS, and ERK1/2. PLC gamma activity was dependent on the presence of Tyr(766) but also required Tyr(677) and/or Tyr(701). Although fully active chimeras did not require PLC gamma, the responses of chimeras showing reduced activation of FRS2 were significantly enhanced by this activity. These results establish that PFR1 does not utilize any tyrosine residues, phosphorylated or not, to activate FRS2. However, it does require Tyr(677) and/or Tyr(701), which may function to stabilize the active conformation directly or indirectly.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/fisiología , Tirosina/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , División Celular , Activación Enzimática , Isoenzimas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Moleculares , Mutagénesis , Neuritas/efectos de los fármacos , Neuritas/patología , Células PC12 , Péptidos/farmacología , Fosfolipasa C gamma , Fosfoproteínas/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Ratas , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Fosfolipasas de Tipo C/metabolismo , Tirosina/metabolismo
4.
J Neurosci Res ; 63(4): 341-6, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11170184

RESUMEN

We previously identified the urokinase plasminogen activator receptor (UPAR) as a gene induced by nerve growth factor (NGF), but not by epidermal growth factor (EGF), in PC12 cells (Farias-Eisner et al. [2000] J. Neurosci. 20:230-239). Antisense oligonucleotides for the UPAR mRNA or an antibody directed against UPAR protein, added simultaneously with NGF, block NGF-induced morphological and biochemical differentiation of PC12 cells. In this report, we show that anti-UPAR antibody blocks morphological differentiation and the expression of two NGF-specific secondary response genes, collagenase-1 and transin, in PC12 cells only during the first 2 hr following NGF exposure. These data suggest that induced UPAR expression is required only over a short period of time following exposure to NGF for the differentiation program in PC12 cells to proceed. For two models of "primed" PC12 cells, we found that UPAR expression and function are not required for NGF-induced differentiation. UPAR and the secondary response genes collagenase-1 and transin are not induced in "primed" PC12 cells in response to NGF, and anti-UPAR antibody does not block morphological differentiation in these cells. Our data suggests that UPAR is required only transiently during the "priming" of PC12 cells in NGF-induced PC12 cell differentiation.


Asunto(s)
Factor de Crecimiento Nervioso/farmacología , Neuronas/citología , Neuronas/fisiología , Receptores de Superficie Celular/genética , Animales , Anticuerpos/farmacología , Diferenciación Celular/efectos de los fármacos , Colagenasas/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Metaloproteinasa 3 de la Matriz/genética , Pruebas de Neutralización , Células PC12 , Ratas , Receptores de Superficie Celular/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa
5.
J Neurosci ; 20(20): 7556-63, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11027214

RESUMEN

Nerve growth factor binds to the TrkA and p75(NTR) (p75) and generates signals leading to neuronal cell survival, differentiation, and programmed cell death. Here we describe a series of experiments involving selective activation of either TrkA or p75 in which distinct cell-signaling intermediates promote different cellular consequences. We analyzed pheochromocytoma 12 (PC12) cells stably expressing chimeras consisting of the extracellular domain of PDGF receptor (PDGFR) fused to the transmembrane and cytoplasmic segments of p75 or TrkA. Because PC12 cells lack endogenous PDGFR, addition of PDGF to these cell lines permits selective activation of the p75 or TrkA responses without stimulating endogenous receptors. Although both p75 and TrkA activated nuclear factor-kappaB (NF-kappaB), we show that distinct proximal-signaling intermediates are used by each receptor. A dominant-negative mutant of TRAF6 blocked p75- but not TrkA-mediated induction of NF-kappaB. Conversely a dominant-negative mutant of Shc inhibited TrkA but not p75 activation of NF-kappaB. Both of these distinct signaling pathways subsequently converge, leading to activation of the IkappaB kinase complex. Moreover, the activation of NF-kappaB by these distinct pathways after stimulation of either TrkA or p75 leads to different physiological consequences. Blocking p75-mediated activation of NF-kappaB by ecdysone-inducible expression of a nondegradable mutant of IkappaBalpha significantly enhanced apoptosis. In contrast, blocking NF-kappaB induction via TrkA significantly inhibited neurite process formation in PC12 cells. Together these findings indicate that, although both of these receptors lead to the activation of NF-kappaB, they proceed via distinct proximal-signaling intermediates and contribute to different cellular outcomes.


Asunto(s)
Proteínas I-kappa B , FN-kappa B/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Neuritas/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/genética , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Genes Dominantes , Inhibidor NF-kappaB alfa , FN-kappa B/farmacología , Factor de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Células PC12/efectos de los fármacos , Células PC12/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
6.
FASEB J ; 14(7): 973-81, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10783152

RESUMEN

The discoidin domain receptor (DDR1) is characterized by a discoidin I motif in the extracellular domain, an unusually long cytoplasmic juxtamembrane (JM) region, and a kinase domain that is 45% identical to that of the NGF receptor, TrkA. DDR1 also has a major splice form, which has a 37 amino acid insert in the JM region with a consensus Shc PTB site that is lacking in the shorter receptor. One class of ligands for the DDR receptors has recently been identified as being derived from the collagen family, but neither native PC12 cells, which express modest amounts of DDR1, nor transfected PC12 cells, which express much larger amounts of DDR1, respond to this ligand. A chimeric receptor, containing the extracellular domain of hPDGFRbeta fused to the transmembrane and intracellular regions of DDR1, also fails to mediate neuronal-like differentiation in stably transfected PC12 cells and is only weakly autophosphorylated. However, chimeric receptors, which are composed of combinations of intracellular regions from DDR1 and TrkA (with the extracellular domain of hPDGFRbeta), in some cases provided ligand (PDGF) -inducible receptor responses. Those with the TrkA kinase domain and the DDR1 JM regions were able to produce differentiation to varying degrees, whereas the opposite combination did not. Analysis of the signaling responses of the two chimeras with DDR1 JM sequences (with and without the insert) indicated that the shorter sequence bound and activated FRS2 whereas the insert-containing form activated Shc instead. Both activated PLCgamma through the carboxyl-terminal tyrosine of the TrkA domain (Y785 in TrkA residue numbering). Mutation of this site (Y-->F) eliminated PLCgamma activation (indicating there are no other cryptic binding sites for PLCgamma in the DDR1 sequences) and markedly reduced the differentiative activity of the receptor. This is in contrast to TrkA (or PDGFRbeta/TrkA chimeras), where ablation of this pathway has no notable effect on PC12 cell morphogenic responses. Thus, the activation of FRS2 and Shc (leading to MAPK activation) is weaker in the DDR1/TrkA chimeras than in TrkA alone, and the PLCgamma contribution becomes essential for full response. Nonetheless, both DDR1 JM regions contain potentially usable signaling sites, albeit they apparently are not activated directly in DDR1 (or DDR1 chimeras) in a ligand-dependent fashion. These findings suggest that the DDR1 receptors do have signaling capacity but may require additional components or altered conditions to fully activate their kinase domains and/or sustain the activation of the JM sites.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras , Receptor trkA/metabolismo , Receptores Mitogénicos/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Animales , Membrana Celular/metabolismo , Receptores con Dominio Discoidina , Activación Enzimática , Isoenzimas/metabolismo , Células PC12 , Fosfolipasa C gamma , Fosforilación , Ratas , Receptor trkA/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Recombinantes de Fusión/genética , Especificidad por Sustrato , Fosfolipasas de Tipo C/metabolismo
7.
Biochim Biophys Acta ; 1477(1-2): 253-66, 2000 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-10708862

RESUMEN

Mouse alpha- and gamma-nerve growth factor (NGF) are glandular kallikreins that form a non-covalent complex (7S NGF) with beta-NGF. gamma-NGF is an active arginine-specific esteropeptidase; the alpha-subunit is catalytically inactive and has a zymogen-like conformation. Site-directed mutagenesis of alpha-NGF to alter the N-terminus and three residues in loop 7, a region that contributes to the catalytic center, restored substantial catalytic activity against N-benzoyl arginine-p-nitroanilide as substrate in two derivatives although they were not as active as recombinant gamma-NGF. Seven of the 15 derivatives that remained more alpha-like were able to substitute for native alpha-NGF in reforming 7S complexes; the other eight derivatives that were more gamma-like showed greatly reduced ability to do so. However, the most gamma-like alpha-NGF derivative could not substitute for native gamma-NGF in 7S complex formation. These findings suggest that the alpha-NGF backbone can be corrected to a functional enzyme by the addition of a normal N-terminal structure and two catalytic site substitutions and that the 7S complex requires one kallikrein subunit in the zymogen form and one in an active conformation.


Asunto(s)
Endopeptidasas/química , Factor de Crecimiento Nervioso/química , Secuencia de Aminoácidos , Animales , Catálisis , Línea Celular , Cromatografía en Gel , Precursores Enzimáticos/química , Humanos , Espectrometría de Masas , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Factor de Crecimiento Nervioso/biosíntesis , Factor de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/química , Plásmidos , Conformación Proteica , Proteínas Recombinantes/química , Glándula Submandibular/enzimología
8.
J Biol Chem ; 275(3): 2147-56, 2000 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-10636920

RESUMEN

The role of signal transducer and activator of transcription (STAT) signaling pathways in the interleukin-6 (IL-6)-induced morphological differentiation of PC12-E2 cells was assessed using wild type and dominant negative mutants of Stat1 and Stat3, containing Tyr --> Phe (YF), Ser --> Ala (SA), and the double mutations (DM), respectively. FS3-YF or FS3-DM markedly inhibited the IL-6-induced response, but overexpression of FS3-SA caused only a modest inhibition. Expression of all Stat3 mutants had no effect on NGF-induced neurite outgrowth. Overexpression of wild type Stat1 protein inhibited IL-6 activated DNA binding complexes containing Stat3 homodimers, which may explain the partial negative effect of Stat1 on IL-6-induced neurite outgrowth. Specificity of these STAT constructs was confirmed using luciferase reporter gene assays, which showed that IL-6-activated transcription was blocked by expression of FS3-YF and FS3-DM and that FS1 enhanced the interferon gamma-activated transcription. Thus, in PC12-E2 cells, Stat3 homodimers are preferentially activated by IL-6, indicating a role for Stat3 in the regulation of cellular differentiation. Furthermore, IL-6 induced robust neurite outgrowth in PC12-E2 cells expressing dominant negative forms of RAS or SHC or in cells pretreated with the mitogen-activated protein kinase mitogen-activated protein kinase kinase inhibitor, PD98059. Thus, activation of the Stat3 signaling pathway, but not RAS/ERK dependent pathways, is essential for differentiation of PC12-E2 cells by IL-6.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Proteínas de Unión al ADN/metabolismo , Interleucina-6/metabolismo , Transactivadores/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Regulación de la Expresión Génica , Genes Dominantes , Humanos , Interferón gamma/metabolismo , Mutagénesis Sitio-Dirigida , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Neuritas/metabolismo , Células PC12 , Fosforilación , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ratas , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Serina/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Factores de Tiempo , Transactivadores/genética , Tirosina/metabolismo
10.
J Biol Chem ; 274(29): 20047, 1999 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-10400608
11.
Proc Natl Acad Sci U S A ; 96(13): 7178-83, 1999 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-10377388

RESUMEN

Stably transfected PC12 cell lines expressing similar amounts of chimeric receptors composed of the extracellular domain of the human platelet-derived growth factor (PDGF)beta receptor and the transmembrane and intracellular domains of the fibroblast growth factor receptors (FGFRs) 1, 3, and 4 undergo ligand-induced differentiation. The FGFR1 chimera (PFR1) is the most potent of the three, and PFR4 requires more frequent (every 24 hr) addition of ligand to maintain the response. Both PFR1 and -3 also show significant ligand-independent autophosphorylation but PFR4 does not. All of the chimeras activated phospholipase Cgamma, Shc, FGFR substrate (FRS)2, and the mitogen-activated protein kinases, ERK1 and 2. PFR4 was moderately weaker in stimulating these effects as well; PFR1 and -3 were comparable. None of the chimeras induced Sos association or were coprecipitated with Shc. Cotransfection of a dominant-negative Shc derivative, with tyrosine at 239, 240, and 317 replaced with phenylalanine, in the PFR-expressing cells was without effect on PDGF-induced neurite outgrowth. The same derivative substantially inhibited the response of these cells to NGF. These results indicate that FGFR1, 3, and 4 (i) are capable of signaling in a similar fashion; (ii) primarily use FRS2 and, perhaps, PLCgamma; and (iii) do not utilize Shc. The results also suggest that the principal difference between FGFR1, 3, and 4 is in the strength of the tyrosine kinase activity and that qualitative differences in signaling capacity are likely to be less important.


Asunto(s)
Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Ligandos , Células PC12 , Fosforilación , Ratas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/genética , Transfección
12.
J Biol Chem ; 274(19): 13403-9, 1999 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-10224104

RESUMEN

In eukaryotes, two isozymes (I and II) of methionine aminopeptidase (MetAP) catalyze the removal of the initiator methionine if the penultimate residue has a small radius of gyration (glycine, alanine, serine, threonine, proline, valine, and cysteine). Using site-directed mutagenesis, recombinant yeast MetAP I derivatives that are able to cleave N-terminal methionine from substrates that have larger penultimate residues have been expressed. A Met to Ala change at 329 (Met206 in Escherichia coli enzyme) produces an average catalytic efficiency 1.5-fold higher than the native enzyme on normal substrates and cleaves substrates containing penultimate asparagine, glutamine, isoleucine, leucine, methionine, and phenylalanine. Interestingly, the native enzyme also has significant activity with the asparagine peptide not previously identified as a substrate. Mutation of Gln356 (Gln233 in E. coli MetAP) to alanine results in a catalytic efficiency about one-third that of native with normal substrates but which can cleave methionine from substrates with penultimate histidine, asparagine, glutamine, leucine, methionine, phenylalanine, and tryptophan. Mutation of Ser195 to alanine had no effect on substrate specificity. None of the altered enzymes produced cleaved substrates with a fully charged residue (lysine, arginine, aspartic acid, or glutamic acid) or tyrosine in the penultimate position.


Asunto(s)
Aminopeptidasas/metabolismo , Saccharomyces cerevisiae/enzimología , Aminopeptidasas/química , Aminopeptidasas/genética , Secuencia de Bases , Catálisis , Cartilla de ADN , Cinética , Metionil Aminopeptidasas , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
13.
Biotechnol Appl Biochem ; 29(2): 157-63, 1999 04.
Artículo en Inglés | MEDLINE | ID: mdl-10075912

RESUMEN

Methionine aminopeptidase (MetAP), in two isoenzymic forms, is responsible for hydrolysis of the initiator methionine residues from the majority of newly synthesized proteins. It is an essential gene product and is ubiquitously found in archaea, eubacteria, and lower and higher eukaryotes. MetAP also has a potentially important role in the production of recombinant proteins, since failure to correctly remove initiator methionine residues can result in a product that is inactive or immunogenic. A rapid two-step purification of recombinant yeast (Saccharomyces cerevisiae) MetAP I that produces a product that is more than 95% pure and maintains a high level of activity (kcat 320-1556 min-1) has been developed. In addition, a versatile, accurate and sensitive assay for MetAP activity is presented. In contrast with previous methods, which usually use short tripeptides and require a post-reaction derivatization step, this assay uses peptides ranging in size from four to eight residues and utilizes UV detection of a tryptophan residue at the C-terminus. As little as 1 pmol of yeast MetAP I can be detected, with 5 microM peptide in a 100 microl reaction. The combination of a rapid purification protocol and a significantly improved activity assay will allow for a detailed examination of MetAP structure-function relationships and may lead to improved enzyme reagents for use in recombinant-protein production.


Asunto(s)
Aminopeptidasas/aislamiento & purificación , Saccharomyces cerevisiae/enzimología , Secuencia de Aminoácidos , Aminopeptidasas/química , Aminopeptidasas/metabolismo , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Clonación Molecular , Cartilla de ADN , Electroforesis en Gel de Poliacrilamida , Metionil Aminopeptidasas , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Sensibilidad y Especificidad , Especificidad por Sustrato
14.
J Biol Chem ; 273(52): 35250-9, 1998 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-9857065

RESUMEN

The effect of six point mutations causing various human skeletal dysplasias, occurring in the transmembrane (TM) and kinase domains (KD) of fibroblast growth factor receptor 3, were introduced into a chimera composed of the extracellular domain of human platelet-derived growth factor beta and the TM and intracellular domains of hFGFR3. Stable transfectants in rat PC12 cells showed distinct differences in the two classes of mutations. The cells containing TM mutants displayed normal expression and activation but higher responsiveness to lower doses of ligand. The KD mutants showed significantly altered expression patterns. Normal amounts of a lower Mr receptor (p130) reflecting incomplete glycosylation, but only greatly decreased amounts of the mature (p170) form, were observed. However, the latter material showed normal ligand-dependent activation. In contrast, the p130 form, which is regularly observed in the expression of both native and chimeric receptors, exhibits strong ligand-independent tyrosine phosphorylation, particularly with the K650E mutation. Expression of two of the KD mutants (K650M and K650E), under control of an inducible metallothionein promoter, indicated that this receptor was sufficiently autoactivated to produce at least partial differentiation and, in the case of the K650E mutation, to induce ligand-independent neurite outgrowth. A model is presented that suggests that the low Mr (p130) KD mutants can, under the right conditions, signal intracellularly, but when they are fully glycosylated and move to the cell surface they adopt a normal, inhibited conformation, in the form of ligand-independent dimers, that neutralizes the effects of the mutations. When ligands bind, these dimeric receptors are activated in a normal manner. This model suggests that unliganded dimers may be a common intermediate in receptor tyrosine kinase signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Mutación , Osteocondrodisplasias/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Tirosina Quinasas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Diferenciación Celular , Proteína Adaptadora GRB2 , Humanos , Isoenzimas/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Neuritas , Células PC12/citología , Fosfolipasa C gamma , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Proteínas/metabolismo , Ratas , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Proteínas Son Of Sevenless , Fosfolipasas de Tipo C/metabolismo
15.
Protein Sci ; 7(12): 2684-7, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9865965

RESUMEN

Yeast methionine aminopeptidase I (MetAP I) is one of two enzymes in Saccharomyces cerevisiae that is responsible for cotranslational cleavage of initiator methionines. It has previously been classified as a Co2+ metalloprotease in all prokaryotic and eukaryotic forms studied. However, treatment of recombinant apo-MetAP I with 12.5 microM Zn2+ produces an enzyme that is as active as that reconstituted with 200 microM Co2+. In the presence of physiological concentrations of reduced glutathione (GSH), Co-MetAP I is inactive, while the activity of Zn-MetAP I is increased more than 1.7-fold over Zn-MetAP I assayed in the absence of GSH. Given that the in vivo concentration of Zn2+ is at least 1,000-fold higher than that of Co2+, and that Co2+ is insoluble in physiological concentrations of GSH, it is probable that yeast MetAP I is actually a Zn2+ metalloprotease. Furthermore, unless there are extraordinary conditions that insulate or sequester them from this reducing milieu, that have yet to be identified, there are not likely to be any cytoplasmic enzymes that use free Co2+.


Asunto(s)
Aminopeptidasas/metabolismo , Cobalto/metabolismo , Levaduras/enzimología , Zinc/metabolismo , Aminopeptidasas/efectos de los fármacos , Aminopeptidasas/genética , Unión Competitiva , Cationes , Activación Enzimática , Proteínas Fúngicas/efectos de los fármacos , Proteínas Fúngicas/metabolismo , Glutatión/metabolismo , Glutatión/farmacología , Metionil Aminopeptidasas , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
Immunol Cell Biol ; 76(5): 406-13, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9797459

RESUMEN

Rat phaeochromocytoma (PC12) cells respond to many growth factors and produce different phenotypes, including neurite outgrowth. Receptor tyrosine kinases (RTK), which activate multiple signalling pathways in response to ligand binding, initiate many of these. One such family of receptors, the fibroblast growth factor receptor (FGFR), has four different members and expresses at least three of these in PC12 cells. A chimeric tyrosine kinase receptor, consisting of the extracellular domain of human plasma-derived growth factor receptor-beta (hPDGFR-beta) and the transmembrane and intracellular region of FGFR1 (designated PFR1), was constructed and was stably transfected into cloned PC12 cell lines. This chimera, which can be activated without stimulating endogenous RTK including other FGFR, induces neurite outgrowth in a PDGF-dependent manner. By altering the protocol for preparing the retroviral vectors, cells with a wide range of expression levels can be obtained. The amount of these chimeric receptors seems to correlate with the time and the intensity of response as observed in neurite outgrowth assays. Analysis of proteins implicated in FGFR1 signalling indicates that upon stimulation, a tyrosine phosphorylated protein designated FRS2 associates with SOS, Grb2 and the receptor. The chimeric receptor appears entirely similar to that observed for the stimulation of native PC12 cells with FGF2. These results support the view that FRS2 is the dominant FGFR1 signalling entity in PC12 cells.


Asunto(s)
Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/fisiología , Animales , Northern Blotting , Western Blotting , Línea Celular , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Vectores Genéticos , Humanos , Factores de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Células PC12 , Reacción en Cadena de la Polimerasa , Proteínas Tirosina Quinasas/metabolismo , Ratas , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento/análisis , Proteínas Recombinantes de Fusión/genética
17.
Trends Biochem Sci ; 23(7): 263-7, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9697417

RESUMEN

Removal of the initiator methionine and/or acetylation of the alpha-amino group are among the earliest possible chemical modifications that occur during protein synthesis in eukaryotes. These events are catalyzed by methionine aminopeptidase and N alpha-acetyltransferase, respectively. Recent advances in the isolation and characterization of these enzymes indicate that they exist as isoforms that vary in cellular location, function, and evolutionary origins.


Asunto(s)
Acetiltransferasas/metabolismo , Aminopeptidasas/metabolismo , Acetiltransferasas/química , Acetiltransferasas/genética , Aminopeptidasas/química , Aminopeptidasas/genética , Animales , Evolución Molecular , Humanos , Metionil Aminopeptidasas , Modelos Moleculares , Conformación Proteica , Procesamiento Proteico-Postraduccional
18.
J Biol Chem ; 272(35): 22293-9, 1997 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-9268379

RESUMEN

By transient expression of both truncated forms of p52(SHCA) and those with point mutations in 293T cells, it has been shown that, in addition to Tyr-317, Tyr-239/240 is a major site of phosphorylation that serves as a docking site for Grb2.Sos1 complexes. In addition, analysis of epidermal growth factor (EGF)-induced activation of mitogen-activated protein kinase in 293T cells showed that the overexpression Shc SH2 or phosphotyrosine binding (PTB) domains of ShcA alone has a more potent negative effect than the overexpression of the forms of ShcA lacking Tyr-317 or Tyr 239/240 or both. In transiently transfected PC12 cells, the ShcA PTB domain and tyrosine phosphorylation in the CH1 domain, especially on Tyr-239/240, are crucial for mediating nerve growth factor (NGF)-induced neurite outgrowth. These findings suggest that the EGF and NGF (TrkA) receptor can utilize Shc in different ways to promote their activity. For EGF-induced mitogen-activated protein kinase activation in 293T cells, both Shc PTB and SH2 domains are essential for optimal activation, indicating that a mechanism independent of Grb2 engagement with Shc may exist. For NGF-induced neurite outgrowth in PC12 cells, Shc PTB plays an essential role, and phosphorylation on Tyr-239/240, but not on Tyr-317, is required.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Factores de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas/metabolismo , Tirosina/metabolismo , Animales , Sitios de Unión , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , División Celular , Activación Enzimática , Proteína Adaptadora GRB2 , Humanos , Riñón/citología , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Mutagénesis , Células PC12 , Ratas , Proteínas Adaptadoras de la Señalización Shc , Proteínas Son Of Sevenless , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Dominios Homologos src
20.
Mol Cell Biol ; 17(7): 4169-77, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9199352

RESUMEN

Mutations in the gene for human fibroblast growth factor receptor 3 (hFGFR3) cause a variety of skeletal dysplasias, including the most common genetic form of dwarfism, achondroplasia (ACH). Evidence indicates that these phenotypes are not due to simple haploinsufficiency of FGFR3 but are more likely related to a role in negatively regulating skeletal growth. The effects of one of these mutations on FGFR3 signaling were examined by constructing chimeric receptors composed of the extracellular domain of human platelet-derived growth factor receptor beta (hPDGFR beta) and the transmembrane and intracellular domains of hFGFR3 or of an ACH (G375C) mutant. Following stable transfection in PC12 cells, which lack platelet-derived growth factor (PDGF) receptors, all clonal cell lines, with either type of chimera, showed strong neurite outgrowth in the presence of PDGF but not in its absence. Antiphosphotyrosine immunoblots showed ligand-dependent autophosphorylation, and both receptor types stimulated strong phosphorylation of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase, an event associated with the differentiative response of these cells. In addition, ligand-dependent phosphorylation of phospholipase Cgamma and Shc was also observed. All of these responses were comparable to those observed from ligand activation, such as by nerve growth factor, of the native PC12 cells used to prepare the stable transfectants. The cells with the chimera bearing the ACH mutation were more rapidly responsive to ligand with less sustained MAPK activation, indicative of a preactivated or primed condition and consistent with the view that these mutations weaken ligand control of FGFR3 function. However, the full effect of the mutation likely depends in part on structural features of the extracellular domain. Although FGFR3 has been suggested to act as a negative regulator of long-bone growth in chrondrocytes, it produces differentiative signals similar to those of FGFR1, to which only positive effects have been ascribed, in PC12 cells. Therefore, its regulatory effects on bone growth likely result from cellular contexts and not the induction of a unique FGFR3 signaling pathway.


Asunto(s)
Acondroplasia/genética , Proteínas Tirosina Quinasas , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Animales , Cartílago/fisiología , Diferenciación Celular , Humanos , Neuritas/ultraestructura , Células PC12 , Fosforilación , Fosfotirosina/metabolismo , Ratas , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Proteínas Recombinantes de Fusión , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA