Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Biol Chem ; 287(26): 22341-53, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22577147

RESUMEN

During nervous system development, neuronal growth, migration, and functional morphogenesis rely on the appropriate control of the subcellular cytoskeleton including microtubule dynamics. Stathmin family proteins play major roles during the various stages of neuronal differentiation, including axonal growth and branching, or dendritic development. We have shown previously that stathmins 2 (SCG10) and 3 (SCLIP) fulfill distinct, independent and complementary regulatory roles in axonal morphogenesis. Although the two proteins have been proposed to display the four conserved phosphorylation sites originally identified in stathmin 1, we show here that they possess distinct phosphorylation sites within their specific proline-rich domains (PRDs) that are differentially regulated by phosphorylation by proline-directed kinases involved in the control of neuronal differentiation. ERK2 or CDK5 phosphorylate the two proteins but with different site specificities. We also show for the first time that, unlike stathmin 2, stathmin 3 is a substrate for glycogen synthase kinase (GSK) 3ß both in vitro and in vivo. Interestingly, stathmin 3 phosphorylated at its GSK-3ß target site displays a specific subcellular localization at neuritic tips and within the actin-rich peripheral zone of the growth cone of differentiating hippocampal neurons in culture. Finally, pharmacological inhibition of GSK-3ß induces a redistribution of stathmin 3, but not stathmin 2, from the periphery toward the Golgi region of neurons. Stathmin proteins can thus be either regulated locally or locally targeted by specific phosphorylation, each phosphoprotein of the stathmin family fulfilling distinct and specific roles in the control of neuronal differentiation.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Neuronas/metabolismo , Prolina/química , Serina/química , Estatmina/metabolismo , Animales , Diferenciación Celular , Glucógeno Sintasa Quinasa 3 beta , Células HeLa , Humanos , Microtúbulos/metabolismo , Modelos Biológicos , Neuritas/metabolismo , Fosforilación , Conejos , Ratas , Ratas Sprague-Dawley , Ratas Wistar
2.
J Biol Chem ; 285(15): 11667-80, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20145240

RESUMEN

In vertebrates, stathmins form a family of proteins possessing two tubulin binding repeats (TBRs), which each binds one soluble tubulin heterodimer. The stathmins thus sequester two tubulins in a phosphorylation-dependent manner, providing a link between signal transduction and microtubule dynamics. In Drosophila, we show here that a single stathmin gene (stai) encodes a family of D-stathmin proteins. Two of the D-stathmins are maternally deposited and then restricted to germ cells, and the other two are detected in the nervous system during embryo development. Like in vertebrates, the nervous system-enriched stathmins contain an N-terminal domain involved in subcellular targeting. All the D-stathmins possess a domain containing three or four predicted TBRs, and we demonstrate here, using complementary biochemical and biophysical methods, that all four predicted TBR domains actually bind tubulin. D-stathmins can indeed bind up to four tubulins, the resulting complex being directly visualized by electron microscopy. Phylogenetic analysis shows that the presence of regulated multiple tubulin sites is a conserved characteristic of stathmins in invertebrates and allows us to predict key residues in stathmin for the binding of tubulin. Altogether, our results reveal that the single Drosophila stathmin gene codes for a stathmin family similar to the multigene vertebrate one, but with particular tubulin binding properties.


Asunto(s)
Unión Proteica , Estatmina/química , Estatmina/genética , Tubulina (Proteína)/química , Animales , Dimerización , Drosophila , Células HeLa , Humanos , Hibridación in Situ , Microtúbulos/metabolismo , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Interferencia de ARN , Proteínas Recombinantes/química , Resonancia por Plasmón de Superficie
3.
Nature ; 435(7041): 519-22, 2005 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-15917812

RESUMEN

Vinblastine is one of several tubulin-targeting Vinca alkaloids that have been responsible for many chemotherapeutic successes since their introduction in the clinic as antitumour drugs. In contrast with the two other classes of small tubulin-binding molecules (Taxol and colchicine), the binding site of vinblastine is largely unknown and the molecular mechanism of this drug has remained elusive. Here we report the X-ray structure of vinblastine bound to tubulin in a complex with the RB3 protein stathmin-like domain (RB3-SLD). Vinblastine introduces a wedge at the interface of two tubulin molecules and thus interferes with tubulin assembly. Together with electron microscopical and biochemical data, the structure explains vinblastine-induced tubulin self-association into spiral aggregates at the expense of microtubule growth. It also shows that vinblastine and the amino-terminal part of RB3-SLD binding sites share a hydrophobic groove on the alpha-tubulin surface that is located at an intermolecular contact in microtubules. This is an attractive target for drugs designed to perturb microtubule dynamics by interfacial interference, for which tubulin seems ideally suited because of its propensity to self-associate.


Asunto(s)
Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Vinblastina/química , Vinblastina/farmacología , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Proteínas de Microtúbulos/química , Modelos Moleculares , Fosfoproteínas/química , Estructura Terciaria de Proteína , Estatmina , Relación Estructura-Actividad , Vinblastina/metabolismo
4.
Mol Cell Neurosci ; 43(1): 15-32, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19660553

RESUMEN

Nervous system function and plasticity rely on the complex architecture of neuronal networks elaborated during development, when neurons acquire their specific and complex shape. During neuronal morphogenesis, the formation and outgrowth of functionally and structurally distinct axons and dendrites require a coordinated and dynamic reorganization of the microtubule cytoskeleton involving numerous regulators. While most of these factors act directly on microtubules to stabilize them or promote their assembly, depolymerization or fragmentation, others are now emerging as essential regulators of neuronal differentiation by controlling tubulin availability and modulating microtubule dynamics. In this review, we recapitulate how the microtubule network is actively regulated during the successive phases of neuronal morphogenesis, and what are the specific roles of the various microtubule-regulating proteins in that process. We then describe the specific signaling pathways and inter-regulations that coordinate the different activities of these proteins to sustain neuronal development in response to environmental cues.


Asunto(s)
Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Neuronas , Animales , Axones/metabolismo , Axones/ultraestructura , Diferenciación Celular/fisiología , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Morfogénesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/fisiología , Transducción de Señal/fisiología , Moduladores de Tubulina/metabolismo
5.
Nature ; 428(6979): 198-202, 2004 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-15014504

RESUMEN

Microtubules are cytoskeletal polymers of tubulin involved in many cellular functions. Their dynamic instability is controlled by numerous compounds and proteins, including colchicine and stathmin family proteins. The way in which microtubule instability is regulated at the molecular level has remained elusive, mainly because of the lack of appropriate structural data. Here, we present the structure, at 3.5 A resolution, of tubulin in complex with colchicine and with the stathmin-like domain (SLD) of RB3. It shows the interaction of RB3-SLD with two tubulin heterodimers in a curved complex capped by the SLD amino-terminal domain, which prevents the incorporation of the complexed tubulin into microtubules. A comparison with the structure of tubulin in protofilaments shows changes in the subunits of tubulin as it switches from its straight conformation to a curved one. These changes correlate with the loss of lateral contacts and provide a rationale for the rapid microtubule depolymerization characteristic of dynamic instability. Moreover, the tubulin-colchicine complex sheds light on the mechanism of colchicine's activity: we show that colchicine binds at a location where it prevents curved tubulin from adopting a straight structure, which inhibits assembly.


Asunto(s)
Colchicina/química , Colchicina/metabolismo , Proteínas de Microtúbulos , Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/metabolismo , Fosfoproteínas/química , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Colchicina/farmacología , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína , Estatmina , Moduladores de Tubulina
6.
J Neurosci ; 28(29): 7387-98, 2008 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-18632943

RESUMEN

Cerebellar Purkinje cells elaborate one of the most complex dendritic arbors among neurons to integrate the numerous signals they receive from the cerebellum circuitry. Their dendritic differentiation undergoes successive, tightly regulated phases of development involving both regressive and growth events. Although many players regulating the late phases of Purkinje cell dendritogenesis have been identified, intracellular factors controlling earlier phases of dendritic development remain mostly unknown. In this study, we explored the biological properties and functions of SCLIP, a protein of the stathmin family, in Purkinje cell dendritic differentiation and cerebellum development. Unlike the other stathmins, SCLIP is strongly expressed in Purkinje cells during cerebellar development and accumulates in their dendritic processes at a critical period of their formation and outgrowth. To reveal SCLIP functions, we developed a lentiviral-mediated approach on cerebellar organotypic cultures to inhibit or increase its expression in Purkinje cells in their tissue environment. Depletion of SCLIP promoted retraction of the Purkinje cell primitive process and then prevented the formation of new dendrites at early stages of postnatal development. It also prevented their elongation and branching at later phases of differentiation. Conversely, SCLIP overexpression promoted dendritic branching and development. Together, our results demonstrate for the first time that SCLIP is crucial for both the formation and proper development of Purkinje cell dendritic arbors. SCLIP appears thus as a novel and specific factor that controls the early phases of Purkinje cell dendritic differentiation during cerebellum development.


Asunto(s)
Diferenciación Celular/fisiología , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Dendritas/metabolismo , Factores de Crecimiento Nervioso/fisiología , Células de Purkinje/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Línea Celular , Cerebelo/anatomía & histología , Cerebelo/embriología , Dendritas/genética , Humanos , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Técnicas de Cultivo de Órganos , Células de Purkinje/citología , Ratas
7.
Biol Cell ; 100(10): 577-89, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18422486

RESUMEN

BACKGROUND INFORMATION: Precise localization of proteins to specialized subcellular domains is fundamental for proper neuronal development and function. The neural microtubule-regulatory phosphoproteins of the stathmin family are such proteins whose specific functions are controlled by subcellular localization. Whereas stathmin is cytosolic, SCG10, SCLIP and RB3/RB3'/RB3'' are localized to the Golgi and vesicle-like structures along neurites and at growth cones. We examined the molecular determinants involved in the regulation of this specific subcellular localization in hippocampal neurons in culture. RESULTS: We show that their conserved N-terminal domain A carrying two palmitoylation sites is dominant over the others for Golgi and vesicle-like localization. Using palmitoylation-deficient GFP (green fluorescent protein) fusion mutants, we demonstrate that domains A of stathmin proteins have the particular ability to control protein targeting to either Golgi or mitochondria, depending on their palmitoylation. This regulation involves the co-operation of two subdomains within domain A, and seems also to be under the control of its SLD (stathmin-like domain) extension. CONCLUSIONS: Our results unravel that, in specific biological conditions, palmitoylation of stathmin proteins might be able to control their targeting to express their functional activities at appropriate subcellular sites. They, more generally, open new perspectives regarding the role of palmitoylation as a signalling mechanism orienting proteins to their functional subcellular compartments.


Asunto(s)
Proteínas Portadoras/metabolismo , Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Estatmina/metabolismo , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Células Cultivadas , Drosophila , Proteínas de Drosophila/metabolismo , Hipocampo/citología , Humanos , Lipoilación , Ratones , Proteínas de Microtúbulos , Datos de Secuencia Molecular , Mutación , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Estructura Terciaria de Proteína , Ratas
8.
Mol Biol Cell ; 13(2): 698-710, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11854423

RESUMEN

Stathmin is a ubiquitous regulatory phosphoprotein, the generic element of a family of neural phosphoproteins in vertebrates that possess the capacity to bind tubulin and interfere with microtubule dynamics. Although stathmin and the other proteins of the family have been associated with numerous cell regulations, their biological roles remain elusive, as in particular inactivation of the stathmin gene in the mouse resulted in no clear deleterious phenotype. We identified stathmin phosphoproteins in Drosophila, encoded by a unique gene sharing the intron/exon structure of the vertebrate stathmin and stathmin family genes. They interfere with microtubule assembly in vitro, and in vivo when expressed in HeLa cells. Drosophila stathmin expression is regulated during embryogenesis: it is high in the migrating germ cells and in the central and peripheral nervous systems, a pattern resembling that of mammalian stathmin. Furthermore, RNA interference inactivation of Drosophila stathmin expression resulted in germ cell migration arrest at stage 14. It also induced important anomalies in nervous system development, such as loss of commissures and longitudinal connectives in the ventral cord, or abnormal chordotonal neuron organization. In conclusion, a single Drosophila gene encodes phosphoproteins homologous to the entire vertebrate stathmin family. We demonstrate for the first time their direct involvement in major biological processes such as development of the reproductive and nervous systems.


Asunto(s)
Drosophila/fisiología , Proteínas de Microtúbulos , Microtúbulos/fisiología , Sistema Nervioso/embriología , Fosfoproteínas/fisiología , Secuencia de Aminoácidos , Animales , Drosophila/embriología , Exones/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Estatmina , Transfección
9.
J Neurosci ; 25(3): 737-47, 2005 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-15659612

RESUMEN

Understanding the biological relevance of reexpression of developmental molecules in pathological conditions is crucial for the development of new therapies. In this study, we report the increased expression of stathmin, a developmentally regulated tubulin-binding protein, in the brains of patients with multiple sclerosis (MS). In physiological conditions, stathmin immunoreactivity was observed in polysialic acid-neural cell adhesion molecule-positive migratory progenitors in the subventricular zone, and its expression progressively decreased as the cells matured into oligodendrocytes (OLs). In MS patients, however, stathmin levels were elevated in 2',3'-cyclic nucleotide 3'-phosphodiesterase-positive OLs, in 10 of 10 bioptic samples analyzed. Increased levels of stathmin were confirmed by Western blot analysis of normal-appearing white matter samples from MS brains. In addition, using mass spectrometry, stathmin was identified as the main component of a specific myelin protein fraction consistently increased in MS preparations compared with controls. To test the biological relevance of increased stathmin levels, primary OL progenitors were transfected using a myc-tagged stathmin cDNA and were allowed to differentiate. Consistent with a distinct role played by this molecule in cells of the OL lineage at different developmental stages, transient transfection in progenitors favored the bipolar migratory phenotype but did not affect survival. However, sustained stathmin levels in differentiating OLs, because of overexpression, resulted in enhanced apoptotic susceptibility. We conclude that stathmin expression in demyelinating disorders could have a dual role. On one hand, by favoring the migratory phenotype of progenitors, it may promote myelin repair. On the other hand, stathmin in mature OLs may indicate cell stress and possibly affect survival.


Asunto(s)
Encéfalo/metabolismo , Enfermedades Desmielinizantes/metabolismo , Proteínas de Microtúbulos/biosíntesis , Oligodendroglía/metabolismo , Fosfoproteínas/biosíntesis , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Enfermedades Desmielinizantes/inducido químicamente , Epilepsia del Lóbulo Temporal/metabolismo , Etidio , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Microtúbulos/fisiología , Esclerosis Múltiple/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Fosfoproteínas/fisiología , Ratas , Estatmina , Células Madre/metabolismo
10.
FEBS J ; 273(3): 577-87, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16420481

RESUMEN

Protein phosphorylation ensures the accurate and controlled expression of the genome, for instance by regulating the activities of pre-mRNA splicing factors. Here we report that splicing factor 1 (SF1), which is involved in an early step of intronic sequence recognition, is highly phosphorylated in mammalian cells on two serines within an SPSP motif at the junction between its U2AF65 and RNA binding domains. We show that SF1 interacts in vitro with the protein kinase KIS, which possesses a 'U2AF homology motif' (UHM) domain. The UHM domain of KIS is required for KIS and SF1 to interact, and for KIS to efficiently phosphorylate SF1 on the SPSP motif. Importantly, SPSP phosphorylation by KIS increases binding of SF1 to U2AF65, and enhances formation of the ternary SF1-U2AF65-RNA complex. These results further suggest that this phosphorylation event has an important role for the function of SF1, and possibly for the structural rearrangements associated with spliceosome assembly and function.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Prolina/metabolismo , Ribonucleoproteínas/metabolismo , Serina/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos/fisiología , Células HeLa , Humanos , Técnicas In Vitro , Fosforilación , Unión Proteica , ARN/metabolismo , Factores de Empalme de ARN , Factor de Empalme U2AF
11.
J Chromatogr A ; 1106(1-2): 181-9, 2006 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-16427064

RESUMEN

Two-dimensional gel electrophoresis (2-DE) and tandem mass spectrometry were successfully used for determination of a phosphorylation site of stathmin induced by heat stress to Jurkat cells of a human T lymphoblastic cell line. The cells were incubated for 30 min at 41 degrees C up to 45 degrees C in a serum free 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) buffered culture medium. The intracellular soluble proteins were separated by 2-DE, and some of the proteins increased their abundance by heat stress. Those proteins were identified to be calmodulin, protein kinase C substrate, thymosin beta-4 and F-actin capping protein beta-subunit by peptide mass fingerprinting (PMF) with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS). On the contrary, protein phosphatase 2C gamma-isoform, nucleophosmin, translationally controlled tumor protein, Rho GDP-dissociation inhibitor-1, eukaryotic translation initiation factors 5A and 3A subunit 2, ubiquitin-like protein SMT 3B and chloride intracellular channel protein-1 were decreased their abundance. A protein spot of M(r) 18,000 and pI 5.9 was markedly increased at temperatures higher than 43 degrees C at which the cells were led to apoptosis. The spot was identified to be stathmin of a signal relay protein which has a function of sequestering microtubule. MALDI-quadrupole ion trap (QIT)-TOF-MS/MS and immunoblotting with a monoclonal antibody specific for a phosphorylation site of stathmin showed that the spot was a phosphorylated stathmin at serine 37 (Ser 37). The phosphorylation was suppressed by treatment of cells with olomoucine of an inhibitor specific for cyclin dependent kinase (Cdk-1). These results strongly suggest that heat stress activates Cdk-1 which phosphorylates Ser 37 on the stathmin molecule. The phosphorylation may cause the functional loss of stathmin for dynamic microtubule assembly and leads Jurkat cells to cell cycle arrest and apoptosis.


Asunto(s)
Electroforesis en Gel Bidimensional/métodos , Trastornos de Estrés por Calor/metabolismo , Serina/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Estatmina/metabolismo , Secuencia de Aminoácidos , Apoptosis , Humanos , Células Jurkat , Datos de Secuencia Molecular , Fosforilación , Estatmina/química
12.
Biochem J ; 378(Pt 3): 877-88, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-14670078

RESUMEN

Stathmin is a ubiquitous 17 kDa cytosolic phosphoprotein proposed to play a general role in the integration and relay of intracellular signalling pathways. It is believed to regulate microtubule dynamics by sequestering tubulin in a complex made of two tubulin heterodimers per stathmin molecule (T2S complex). The other proteins of the stathmin family can also bind two tubulin heterodimers through their SLD (stathmin-like domain), but the different tubulin:SLD complexes display varying stabilities. In this study, we analysed the relative influence of three regions of SLDs on the interaction with tubulin and the mechanistic processes that lead to its sequestration. Tubulin-binding properties of fragments and chimaeras of stathmin and RB3(SLD) were studied in vitro by tubulin polymerization, size-exclusion chromatography and surface plasmon resonance assays. Our results show that the N-terminal region of SLDs favours the binding of the first tubulin heterodimer and that the second C-terminal tubulinbinding site confers the specific stability of a given tubulin:SLD complex. Our results highlight the molecular processes by which tubulin co-operatively interacts with the SLDs. This knowledge may contribute to drug development aimed at disturbing microtubules that could be used for the treatment of cancer.


Asunto(s)
Proteínas de Microtúbulos , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Tubulina (Proteína)/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Sustancias Macromoleculares , Metilaminas/farmacología , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Fosfoproteínas/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Estatmina
13.
Prog Neurobiol ; 126: 1-18, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25449700

RESUMEN

Nervous system development, plasticity and regeneration require numerous, coordinated and finely tuned subcellular mechanisms. Phosphoproteins of the stathmin family, originally identified as intracellular signal relay proteins, are mostly or exclusively expressed in the nervous system with a high level of expression during brain development. Vertebrate stathmins 1-4 all possess a C-terminal "stathmin-like domain" that binds or releases tubulin in a phosphorylation dependent way, and hence participates in the control of microtubule dynamics, an essential process for neuronal differentiation. Contrary to stathmin 1, stathmins 2-4 possess an N-terminal extension whose reversible palmitoylation specifically targets them to the Golgi and intracellular membranes. Regulation of stathmins 2-4 palmitoylation is therefore an important regulatory mechanism that controls their shuttling to various neuronal compartments where they can then act locally. Expression of stathmins is upregulated during neuronal differentiation and plasticity, and altered in numerous neurodegenerative diseases. Experimental perturbation of stathmins expression in Drosophila or in neurons in culture revealed their importance in neuronal growth and differentiation, each stathmin fulfilling at least partially distinct and likely complementary roles. On the other hand, knock-out of stathmins in mice, with the exception of stathmin 2, resulted in mostly mild or no detected phenotype, revealing likely compensations among stathmins. Altogether, through their combinatorial expression and regulation by phosphorylation and by palmitoylation, and through their interactions with tubulin and other neuronal protein targets, the various stathmins appear as essential regulators of neuronal differentiation at the various stages during development and plasticity of the nervous system.


Asunto(s)
Sistema Nervioso/metabolismo , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Fosfoproteínas/metabolismo , Animales , Humanos
14.
Endocrinology ; 144(4): 1464-73, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12639930

RESUMEN

We used the library subtraction technique to identify genes specifically expressed in the rat uterus during early pregnancy. One such gene was that for stathmin, a factor that is associated with tubulin binding and the destabilization of microtubules. Stathmin was expressed at higher levels in implantation sites than in interimplantation sites on d 6 and 7 of pregnancy; the levels on d 6 and 7 were higher in implantation sites than in the entire uterus on d 3-5 of pregnancy or in nonpregnant uteri. Intense expression of stathmin mRNA was primarily limited to the subluminal stromal cells at the implantation site. Expression was also detected in the decidual zones and was accentuated during the period of decidualization (d 7-12). In the delayed implantation pregnant rat model, uterine stathmin expression was low, but increased after implantation induced by administration of 17beta-estradiol to the progesterone-primed animal. Further, decidualization in the pseudopregnant rat, induced by intrauterine infusion of oil, enhanced stathmin expression. Stathmin expression clearly increases in the uterus when stimulated by embryo implantation and decidualization and may play a role in the early stages of pregnancy.


Asunto(s)
Decidua/fisiología , Implantación del Embrión/fisiología , Proteínas de Microtúbulos , Fosfoproteínas/genética , Útero/fisiología , Animales , Estrógenos/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Hibridación in Situ , Progesterona/farmacología , Seudoembarazo/fisiopatología , ARN Mensajero/análisis , Ratas , Estatmina
15.
FEBS Lett ; 529(2-3): 341-5, 2002 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-12372625

RESUMEN

N-terminal acetylation is a protein modification common in eukaryotes, but rare in prokaryotes. Here, we characterized five mammalian stathmin-like subdomains expressed in Escherichia coli by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and nanoESI Q-TOF tandem mass spectrometry. We revealed that RB3(SLD) and RB3'(SLD) are N(alpha)-acetylated, whereas SCG10(SLD) and SCLIP(SLD), although identical up to residue 6, are not, as well as stathmin. To assess the influence of the N-terminal sequences on N(alpha)-acetylation, we exchanged residues 7 and 8 between acetylated RB3(SLD) and unacetylated SCG10(SLD), and showed that it reversed the acetylation pattern. Our results demonstrate that ectopic recombinant proteins can be extensively N(alpha)-acetylated in E. coli, and that the rules governing N(alpha)-acetylation are complex and involve the N-terminal region, as in eukaryotes.


Asunto(s)
Escherichia coli/metabolismo , Proteínas Recombinantes/metabolismo , Acetilación , Secuencia de Aminoácidos , Datos de Secuencia Molecular , Proteínas Recombinantes/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
16.
FEBS Lett ; 576(1-2): 114-8, 2004 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-15474021

RESUMEN

The physiological role of the prion protein is largely unknown. Here, clustering of prion at the surface of GT1-7 cells was observed upon anti-prion antibody treatments. This clustering was associated with a rapid and transient phosphorylation of the mitogen activated protein kinases (MAPKs) extracellular receptor kinases 1 and 2 (ERK1/2), and also of the microtubule-destabilizing protein stathmin at serine 16. The specificity of this antibody-mediated activation was ascertained by its inhibition by prion small interfering RNA. The phosphorylation of ERK1/2 but not that of stathmin was abolished by the MAPK/ERK kinase 1 inhibitor U0126, whereas both signaling pathways were blocked by the specific inhibitor of the epidermal growth factor receptor AG1478, suggesting the likely recruitment of this receptor upon prion clustering.


Asunto(s)
Proteínas de Microtúbulos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/fisiología , Fosfoproteínas/metabolismo , Proteínas PrPC/fisiología , Animales , Anticuerpos Monoclonales/metabolismo , Western Blotting , Butadienos/farmacología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Microscopía Confocal , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Quinazolinas , ARN Interferente Pequeño/antagonistas & inhibidores , Estatmina , Tirfostinos/farmacología
17.
Brain Res Mol Brain Res ; 114(1): 55-64, 2003 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-12782393

RESUMEN

KIS is the only known protein kinase that possesses an RNA recognition motif. This original structure indicates a role for KIS in the maturation of RNAs possibly by phosphorylating and regulating the activities of RNA associated factors. Another function of KIS has recently been unravelled--it negatively regulates the cdk inhibitor p27Kip1 and thus promotes cell cycle progression through G1. In order to explore the functional expression of this kinase, we quantified its mRNA in a wide range of rat and human tissues, during development and in tumors. In both species, the highest level of KIS gene expression was in adult neural tissues. Interestingly, within the adult rat brain, KIS mRNA is enriched in several areas including the substantia nigra compacta and nuclei of the brain stem. Furthermore, KIS gene expression increases dramatically during brain development. Altogether our results point to a ubiquitous function for KIS together with a particular implication during neural differentiation or in the function of mature neural cells. No dysregulation of KIS gene expression was detected in human tumors from breast, bladder, prostate, liver and kidney origins. On the other hand, the KIS gene was overexpressed in NF1-associated plexiform neurofibromas and malignant peripheral nerve sheath tumors (MPNSTs) as compared to dermal neurofibroma which suggests a possible implication of KIS in the genesis of NF1-associated tumors.


Asunto(s)
Tronco Encefálico/enzimología , Regulación Enzimológica de la Expresión Génica , Proteínas Serina-Treonina Quinasas/genética , Sustancia Negra/enzimología , Factores de Edad , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Datos de Secuencia Molecular , Neurofibromatosis/genética , Neurofibromina 1/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/análisis , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Homología de Secuencia de Aminoácido , Proteínas Supresoras de Tumor/metabolismo
18.
Dev Neurobiol ; 74(12): 1226-42, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24909416

RESUMEN

The hippocampus is one of the two areas in the mammalian brain where adult neurogenesis occurs. Adult neurogenesis is well known to be involved in hippocampal physiological functions as well as pathophysiological conditions. Microtubules (MTs), providing intracellular transport, stability, and transmitting force, are indispensable for neurogenesis by facilitating cell division, migration, growth, and differentiation. Although there are several examples of MT-stabilizing proteins regulating different aspects of adult neurogenesis, relatively little is known about the function of MT-destabilizing proteins. Stathmin is such a MT-destabilizing protein largely restricted to the CNS, and in contrast to its developmental family members, stathmin is also expressed at significant levels in the adult brain, notably in areas involved in adult neurogenesis. Here, we show an important role for stathmin during adult neurogenesis in the subgranular zone of the mouse hippocampus. After carefully mapping stathmin expression in the adult dentate gyrus (DG), we investigated its role in hippocampal neurogenesis making use of stathmin knockout mice. Although hippocampus development appears normal in these animals, different aspects of adult neurogenesis are affected. First, the number of proliferating Ki-67+ cells is decreased in stathmin knockout mice, as well as the expression of the immature markers Nestin and PSA-NCAM. However, newborn cells that do survive express more frequently the adult marker NeuN and have a more mature morphology. Furthermore, our data suggest that migration in the DG might be affected. We propose a model in which stathmin controls the transition from neuronal precursors to early postmitotic neurons.


Asunto(s)
Hipocampo/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Estatmina/metabolismo , Animales , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Proteínas de Unión al ADN , Hipocampo/citología , Antígeno Ki-67/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Nestina/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Células-Madre Neurales/citología , Neuronas/citología , Proteínas Nucleares/metabolismo , Ácidos Siálicos/metabolismo , Estatmina/genética
19.
Mol Biol Cell ; 22(11): 1930-42, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21471001

RESUMEN

Protein palmitoylation is a reversible lipid modification that plays critical roles in protein sorting and targeting to specific cellular compartments. The neuronal microtubule-regulatory phosphoproteins of the stathmin family (SCG10/stathmin 2, SCLIP/stathmin 3, and RB3/stathmin 4) are peripheral proteins that fulfill specific and complementary roles in the formation and maturation of the nervous system. All neuronal stathmins are localized at the Golgi complex and at vesicles along axons and dendrites. Their membrane anchoring results from palmitoylation of two close cysteine residues present within their homologous N-terminal targeting domains. By preventing palmitoylation with 2-bromopalmitate or disrupting the integrity of the Golgi with brefeldin A, we were able to show that palmitoylation of stathmins 2 and 3 likely occurs at the Golgi and is crucial for their specific subcellular localization and trafficking. In addition, this membrane binding is promoted by a specific set of palmitoyl transferases that localize with stathmins 2 and 3 at the Golgi, directly interact with them, and enhance their membrane association. The subcellular membrane-associated microtubule-regulatory activity of stathmins might then be fine-tuned by extracellular stimuli controlling their reversible palmitoylation, which can be viewed as a crucial regulatory process for specific and local functions of stathmins in neurons.


Asunto(s)
Aciltransferasas/metabolismo , Aparato de Golgi/metabolismo , Transporte de Proteínas , Estatmina/metabolismo , Animales , Brefeldino A/farmacología , Membrana Celular/metabolismo , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Aparato de Golgi/efectos de los fármacos , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Lipoilación/efectos de los fármacos , Neuronas/metabolismo , Palmitatos/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas
20.
Methods Cell Biol ; 95: 407-47, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20466147

RESUMEN

The description of the molecular mechanisms of interaction between tubulin or microtubules and partners at atomic scale is expected to have critical impacts on the understanding of basic physiological processes. This information will also help the design of future drug candidates that may be used to fight various pathologies such as cancer or neurological diseases. For these reasons, this aspect of tubulin research has been tackled since the seventies using many different methods and at different scales. NMR appears as a unique approach to provide, with atomic resolution, the solution structure and dynamical properties of tubulin/microtubule partners in free and bound states. Though tubulin is not directly amenable to solution NMR, the NMR ligand-based experiments allow one to obtain valuable data on the molecular mechanisms that sustain structure-function relationship, in particular atomic details on the partner binding site. We will first describe herein some basic principles of solution NMR spectroscopy that should not be missed for a comprehensive reading of NMR reports. A series of results will then be presented to illustrate the wealth and variety of NMR experiments and how this approach enlightens tubulin/microtubules interaction with partners.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Fragmentos de Péptidos/metabolismo , Proteínas/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Animales , Humanos , Ligandos , Espectroscopía de Resonancia Magnética/instrumentación , Microtúbulos/química , Microtúbulos/metabolismo , Modelos Moleculares , Peso Molecular , Fragmentos de Péptidos/química , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Proteínas/química , Soluciones , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA