Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Prog Neurobiol ; 126: 1-18, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25449700

ABSTRACT

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.


Subject(s)
Nervous System/metabolism , Neurogenesis/physiology , Neuronal Plasticity/physiology , Neurons/metabolism , Phosphoproteins/metabolism , Animals , Humans
2.
Dev Neurobiol ; 74(12): 1226-42, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24909416

ABSTRACT

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.


Subject(s)
Hippocampus/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Stathmin/metabolism , Animals , Cell Movement/physiology , Cell Survival/physiology , DNA-Binding Proteins , Hippocampus/cytology , Ki-67 Antigen/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nestin/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Nuclear Proteins/metabolism , Sialic Acids/metabolism , Stathmin/genetics
3.
J Biol Chem ; 287(26): 22341-53, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22577147

ABSTRACT

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.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Neurons/metabolism , Proline/chemistry , Serine/chemistry , Stathmin/metabolism , Animals , Cell Differentiation , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , Microtubules/metabolism , Models, Biological , Neurites/metabolism , Phosphorylation , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar
4.
Mol Biol Cell ; 22(11): 1930-42, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21471001

ABSTRACT

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.


Subject(s)
Acyltransferases/metabolism , Golgi Apparatus/metabolism , Protein Transport , Stathmin/metabolism , Animals , Brefeldin A/pharmacology , Cell Membrane/metabolism , Cells, Cultured , Fluorescent Antibody Technique, Indirect , Golgi Apparatus/drug effects , Hippocampus/cytology , Hippocampus/metabolism , Humans , Lipoylation/drug effects , Neurons/metabolism , Palmitates/pharmacology , Protein Processing, Post-Translational/drug effects , Rats
5.
Methods Cell Biol ; 95: 407-47, 2010.
Article in English | MEDLINE | ID: mdl-20466147

ABSTRACT

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.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Peptide Fragments/metabolism , Proteins/metabolism , Tubulin/chemistry , Tubulin/metabolism , Animals , Humans , Ligands , Magnetic Resonance Spectroscopy/instrumentation , Microtubules/chemistry , Microtubules/metabolism , Models, Molecular , Molecular Weight , Peptide Fragments/chemistry , Peptides/chemistry , Peptides/metabolism , Protein Binding , Proteins/chemistry , Solutions , Tubulin Modulators/chemistry , Tubulin Modulators/metabolism
6.
J Biol Chem ; 285(15): 11667-80, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20145240

ABSTRACT

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.


Subject(s)
Protein Binding , Stathmin/chemistry , Stathmin/genetics , Tubulin/chemistry , Animals , Dimerization , Drosophila , HeLa Cells , Humans , In Situ Hybridization , Microtubules/metabolism , Protein Interaction Mapping , Protein Structure, Tertiary , RNA Interference , Recombinant Proteins/chemistry , Surface Plasmon Resonance
7.
Mol Cell Neurosci ; 43(1): 15-32, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19660553

ABSTRACT

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.


Subject(s)
Cytoskeleton/metabolism , Microtubules/metabolism , Neurons , Animals , Axons/metabolism , Axons/ultrastructure , Cell Differentiation/physiology , Humans , Microtubule-Associated Proteins/metabolism , Morphogenesis/physiology , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/physiology , Signal Transduction/physiology , Tubulin Modulators/metabolism
8.
J Biol Chem ; 284(11): 6909-17, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19131341

ABSTRACT

Microtubules are cytoskeletal components involved in multiple cell functions such as mitosis, motility, or intracellular traffic. In vivo, these polymers made of alphabeta-tubulin nucleate mostly from the centrosome to establish the interphasic microtubule network or, during mitosis, the mitotic spindle. Centrosomal P4.1-associated protein (CPAP; also named CENPJ) is a centrosomal protein involved in the assembly of centrioles and important for the centrosome function. This protein contains a microtubule-destabilizing region referred to as PN2-3. Here we decrypt the microtubule destabilization activity of PN2-3 at the molecular level and show that it results from the sequestration of tubulin by PN2-3 in a non-polymerizable 1:1 complex. We also map the tubulin/PN2-3 interaction both on the PN2-3 sequence and on the tubulin surface. NMR and CD data on free PN2-3 in solution show that this is an intrinsically unstructured protein that comprises a 23-amino acid residue alpha-helix. This helix is embedded in a 76-residue region that interacts strongly with tubulin. The interference of PN2-3 with well characterized tubulin properties, namely GTPase activity, nucleotide exchange, vinblastine-induced self-assembly, and stathmin family protein binding, highlights the beta subunit surface located at the intermolecular longitudinal interface when tubulin is embedded in a microtubule as a tubulin/PN2-3 interaction area. These findings characterize the PN2-3 fragment of CPAP as a protein with an unprecedented tubulin sequestering mechanism distinct from that of stathmin family proteins.


Subject(s)
Centrioles/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism , Animals , Circular Dichroism , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary/physiology , Sheep
9.
J Mol Biol ; 381(3): 748-62, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18588901

ABSTRACT

The protein kinase KIS is made by the juxtaposition of a unique kinase domain and a C-terminal domain with a U2AF homology motif (UHM), a sequence motif for protein interaction initially identified in the heterodimeric pre-mRNA splicing factor U2AF. This domain of KIS is closely related to the C-terminal UHM domain of the U2AF large subunit, U2AF(65). KIS phosphorylates the splicing factor SF1, which in turn enhances SF1 binding to U2AF(65) and the 3' splice site, an event known to take place at an early step of spliceosome assembly. Here, the analysis of the subcellular localization of mutated forms of KIS indicates that the kinase domain of KIS is the necessary domain for its nuclear localization. As in the case of U2AF(65), the UHM-containing C-terminal domain of KIS is required for binding to the splicing factors SF1 and SF3b155. The efficiency of KIS binding to SF1 and SF3b155 is similar to that of U2AF(65) in pull-down assays. These results further support the functional link of KIS with splicing factors. Interestingly, when compared to other UHM-containing proteins, KIS presents a different specificity for the UHM docking sites that are present in the N-terminal region of SF3b155, thus providing a new insight into the variety of interactions mediated by UHM domains.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribonucleoproteins/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Animals , CHO Cells , Cricetinae , Cricetulus , Cytoplasm/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , RNA Splicing Factors , Rats , Splicing Factor U2AF
10.
J Neurosci ; 28(29): 7387-98, 2008 Jul 16.
Article in English | MEDLINE | ID: mdl-18632943

ABSTRACT

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.


Subject(s)
Cell Differentiation/physiology , Cerebellum/growth & development , Cerebellum/metabolism , Dendrites/metabolism , Nerve Growth Factors/physiology , Purkinje Cells/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Line , Cerebellum/anatomy & histology , Cerebellum/embryology , Dendrites/genetics , Humans , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Organ Culture Techniques , Purkinje Cells/cytology , Rats
11.
Biol Cell ; 100(10): 577-89, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18422486

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Neurons/metabolism , Stathmin/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cells, Cultured , Drosophila , Drosophila Proteins/metabolism , Hippocampus/cytology , Humans , Lipoylation , Mice , Microtubule Proteins , Molecular Sequence Data , Mutation , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Protein Structure, Tertiary , Rats
12.
Mol Cell Neurosci ; 34(2): 137-46, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17145186

ABSTRACT

Stathmin family phosphoproteins participate in the control of microtubule dynamics and have been proposed to be involved in the control of neuronal differentiation. We examined the biological properties and functions of SCLIP, the stathmin family protein most similar to SCG10, a widely studied and recognized neuronal differentiation marker. SCLIP, like SCG10, is present from the earliest stages of hippocampal neuron differentiation in culture at vesicle-like structures following dynamic microtubules. Its inhibition by RNA interference resulted in increased axonal branching, revealing a novel biological role for SCLIP, distinct from SCG10 whose down-regulation in the same conditions promoted growth cone expansion. The enhanced axonal branching resulted from the formation of collateral lamellar protrusions, with cytoskeleton reorganization typical of normal branching. In addition to revealing a novel function for SCLIP in axonal morphogenesis, our results demonstrate for the first time that stathmin family proteins fulfill different and complementary roles during neuronal differentiation.


Subject(s)
Axons/metabolism , Hippocampus/cytology , Nerve Growth Factors/physiology , Neurons/cytology , Stathmin/physiology , Animals , Axons/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cytochalasin D/pharmacology , Cytoskeleton/metabolism , Drug Interactions , Embryo, Mammalian , Fluorescent Antibody Technique/methods , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/biosynthesis , Growth Cones/drug effects , Growth Cones/physiology , Membrane Proteins/metabolism , Neurons/drug effects , Nocodazole/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA, Small Interfering/pharmacology , Rats , Transfection/methods
13.
J Neurochem ; 99(1): 237-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16925597

ABSTRACT

Regulation of cytoskeletal dynamics is essential to neuronal plasticity during development and adulthood. Dysregulation of these mechanisms may contribute to neuropsychiatric and neurodegenerative diseases. The neuronal protein kinase, cyclin-dependent kinase 5 (Cdk5), is involved in multiple aspects of neuronal function, including regulation of cytoskeleton. A neuroproteomic search identified the tubulin-binding protein, stathmin, as a novel Cdk5 substrate. Stathmin was phosphorylated by Cdk5 in vitro at Ser25 and Ser38, previously identified as mitogen-activated protein kinase (MAPK) and p38 MAPKdelta sites. Cdk5 predominantly phosphorylated Ser38, while MAPK and p38 MAPKdelta predominantly phosphorylated Ser25. Stathmin was phosphorylated at both sites in mouse brain, with higher levels in cortex and striatum. Cdk5 knockout mice exhibited decreased phospho-Ser38 levels. During development, phospho-Ser25 and -Ser38 levels peaked at post-natal day 7, followed by reduction in total stathmin. Inhibition of protein phosphatases in striatal slices caused an increase in phospho-Ser25 and a decrease in total stathmin. Interestingly, the prefrontal cortex of schizophrenic patients had increased phospho-Ser25 levels. In contrast, total and phospho-Ser25 stoichiometries were decreased in the hippocampus of Alzheimer's patients. Thus, microtubule regulatory mechanisms involving the phosphorylation of stathmin may contribute to developmental synaptic pruning and structural plasticity, and may be involved in neuropsychiatric and neurodegenerative disorders.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Mitogen-Activated Protein Kinases/metabolism , Stathmin/metabolism , Animals , DNA Primers , Mass Spectrometry , Molecular Sequence Data , Phosphorylation , Polymerase Chain Reaction , Rats , Stathmin/genetics , Tubulin/metabolism
14.
J Chromatogr A ; 1106(1-2): 181-9, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16427064

ABSTRACT

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.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Heat Stress Disorders/metabolism , Serine/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Stathmin/metabolism , Amino Acid Sequence , Apoptosis , Humans , Jurkat Cells , Molecular Sequence Data , Phosphorylation , Stathmin/chemistry
15.
FEBS J ; 273(3): 577-87, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16420481

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proline/metabolism , Ribonucleoproteins/metabolism , Serine/metabolism , Transcription Factors/metabolism , Amino Acid Motifs/physiology , HeLa Cells , Humans , In Vitro Techniques , Phosphorylation , Protein Binding , RNA/metabolism , RNA Splicing Factors , Splicing Factor U2AF
16.
Biochemistry ; 44(44): 14616-25, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16262261

ABSTRACT

Microtubules are major cytoskeletal components involved in numerous cellular functions such as mitosis, cell motility, or intracellular traffic. These cylindrical polymers of alphabeta-tubulin assemble in a closely regulated dynamic manner. We have shown that the stathmin family proteins sequester tubulin in a nonpolymerizable ternary complex, through their stathmin-like domains (SLD) and thus contribute to the regulation of microtubule dynamics. We demonstrate here that short peptides derived from the N-terminal part of SLDs impede tubulin polymerization with various efficiencies and that phosphorylation of the most potent of these peptides reduces its efficiency as in full-length stathmin. To understand the mechanism of action of these peptides, we undertook a NMR-based structural analysis of the peptide-tubulin interaction with the most efficient peptide (I19L). Our results show that, while disordered when free in solution, I19L folds into a beta-hairpin upon binding to tubulin. We further identified, by means of saturation transfer difference NMR, hydrophobic residues located on the beta2-strand of I19L that are involved in its tubulin binding. These structural data were used together with tubulin atomic coordinates from the tubulin/RB3-SLD crystal structure to model the I19L/tubulin interaction. The model agrees with I19L acting through an autonomous tubulin capping capability to impede tubulin polymerization and provides information to help understand the variation of efficiency against tubulin polymerization among the peptides tested. Altogether these results enlighten the mechanism of tubulin sequestration by SLDs, while they pave the way for the development of protein-based compounds aimed at interfering with tubulin polymerization.


Subject(s)
Microtubules/metabolism , Peptides/metabolism , Protein Structure, Secondary , Stathmin/metabolism , Tubulin/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Cattle , Humans , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Protein Structure, Tertiary , Sequence Alignment , Stathmin/chemistry , Tubulin/chemistry
17.
J Cell Sci ; 118(Pt 10): 2313-23, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15870110

ABSTRACT

The appropriate targeting of membrane-associated proteins involves a diversity of motifs including post-translational modifications and specific protein sequences. Phosphoproteins of the stathmin family are important regulators of microtubule dynamics, in particular in the developing and mature nervous system. Whereas stathmin is cytosolic, SCG10, SCLIP and the splice variants RB3/RB3'/RB3'' are associated with Golgi and vesicular membranes, through their palmitoylated N-terminal A domains. In order to identify essential motifs involved in this specific targeting, we examined the subcellular distribution of various subdomains derived from domain A of SCG10 fused with GFP. We show that the Golgi localization of SCG10 results from the cooperation of two motifs: a membrane-anchoring palmitoylation motif and a newly identified Golgi-specifying sequence. The latter displayed no targeting activity by itself, but retained a Golgi-specifying activity when associated with another membrane-anchoring palmitoylation motif derived from the protein GAP-43. We further identified critical residues for the specific Golgi targeting of domain A. Altogether, our results give new insight into the regulation of the subcellular localization of stathmin family proteins, an important feature of their physiological functions in differentiating and mature neural cells. More generally we provide new information on essential mechanisms of functional protein subcellular targeting.


Subject(s)
GAP-43 Protein/metabolism , Golgi Apparatus/metabolism , Nerve Growth Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Carrier Proteins , Cell Membrane/physiology , Cells, Cultured , Dogs , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins , Microtubule Proteins , Molecular Sequence Data , Mutation , Nerve Growth Factors/genetics , Protein Structure, Tertiary , Protein Transport , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stathmin
18.
Nature ; 435(7041): 519-22, 2005 May 26.
Article in English | MEDLINE | ID: mdl-15917812

ABSTRACT

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.


Subject(s)
Tubulin/chemistry , Tubulin/metabolism , Vinblastine/chemistry , Vinblastine/pharmacology , Binding Sites , Crystallography, X-Ray , Dimerization , Hydrophobic and Hydrophilic Interactions , Kinetics , Microtubule Proteins/chemistry , Models, Molecular , Phosphoproteins/chemistry , Protein Structure, Tertiary , Stathmin , Structure-Activity Relationship , Vinblastine/metabolism
19.
J Neurosci ; 25(3): 737-47, 2005 Jan 19.
Article in English | MEDLINE | ID: mdl-15659612

ABSTRACT

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.


Subject(s)
Brain/metabolism , Demyelinating Diseases/metabolism , Microtubule Proteins/biosynthesis , Oligodendroglia/metabolism , Phosphoproteins/biosynthesis , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cells, Cultured , Demyelinating Diseases/chemically induced , Epilepsy, Temporal Lobe/metabolism , Ethidium , Humans , Mice , Mice, Inbred C57BL , Microtubule Proteins/physiology , Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Oligodendroglia/cytology , Phosphoproteins/physiology , Rats , Stathmin , Stem Cells/metabolism
20.
FEBS Lett ; 576(1-2): 114-8, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15474021

ABSTRACT

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.


Subject(s)
Microtubule Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/physiology , Phosphoproteins/metabolism , PrPC Proteins/physiology , Animals , Antibodies, Monoclonal/metabolism , Blotting, Western , Butadienes/pharmacology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique, Indirect , Mice , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Nitriles/pharmacology , Phosphorylation/drug effects , Quinazolines , RNA, Small Interfering/antagonists & inhibitors , Stathmin , Tyrphostins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...