Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 197(1): 258-67, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16289052

ABSTRACT

Thyroid hormone plays an important role in regulating the development and regeneration of the nervous system. Our previous work showed that local administration of triiodothyronine (T3) at the level of transected rat sciatic nerve increased the number and diameter of regenerated axons, but the mechanism underlying the improved regeneration is still unclear. Here, we have investigated the effect of T3 on the expression of SCG10, a regulator of microtubule dynamics in growth cones. After transection of adult rat sciatic nerves, silicone tubes were implanted and filled with T3 or phosphate-buffered solution. At various time points following surgery, the expression of SCG10 protein and mRNA was analyzed. Semi-quantitative Western blot analysis revealed that sciatic nerve transection induced a more than 20-fold upregulation of SCG10 protein in proximal nerve segments at 1 day post-lesion, while at this time point, SCG10 mRNA in dorsal root ganglion neurons was not increased yet. The increase in SCG10 protein and mRNA could be observed over 30 days. Local T3 treatment significantly enhanced the increase in SCG10 protein levels about two-fold in the different segments of transected nerve during the regeneration period. Also SCG10 mRNA levels in lumbar ganglia were enhanced. Immunohistochemical analysis showed that T3 treatment not only increased the number of SCG10 positive axons but also the intensity of their staining. These results suggest that SCG10 is involved in the regulation of regeneration. The stimulating effect of T3 on SCG10 expression could provide a mechanism by which T3 enhances peripheral nerve regeneration.


Subject(s)
Nerve Growth Factors/biosynthesis , Nerve Regeneration/drug effects , Sciatic Nerve/physiology , Thyroid Hormones/pharmacology , Animals , Blotting, Western , Carrier Proteins , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Membrane Proteins , Microtubule Proteins , Nerve Tissue Proteins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Sciatic Nerve/cytology , Sciatic Nerve/drug effects , Triiodothyronine/pharmacology , Up-Regulation/drug effects
2.
Cell Motil Cytoskeleton ; 62(2): 124-32, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16145686

ABSTRACT

The cytoskeleton, composed of actin filaments, intermediate filaments, and microtubules, is a highly dynamic supramolecular network actively involved in many essential biological mechanisms such as cellular structure, transport, movements, differentiation, and signaling. As a first step to characterize the biophysical changes associated with cytoskeleton functions, we have developed finite elements models of the organization of the cell that has allowed us to interpret atomic force microscopy (AFM) data at a higher resolution than that in previous work. Thus, by assuming that living cells behave mechanically as multilayered structures, we have been able to identify superficial and deep effects that could be related to actin and microtubule disassembly, respectively. In Cos-7 cells, actin destabilization with Cytochalasin D induced a decrease of the visco-elasticity close to the membrane surface, while destabilizing microtubules with Nocodazole produced a stiffness decrease only in deeper parts of the cell. In both cases, these effects were reversible. Cell softening was measurable with AFM at concentrations of the destabilizing agents that did not induce detectable effects on the cytoskeleton network when viewing the cells with fluorescent confocal microscopy. All experimental results could be simulated by our models. This technology opens the door to the study of the biophysical properties of signaling domains extending from the cell surface to deeper parts of the cell.


Subject(s)
Cytoskeleton/physiology , Actins/antagonists & inhibitors , Animals , Biomechanical Phenomena , COS Cells , Chlorocebus aethiops , Computer Simulation , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Genes, Reporter , Microscopy, Confocal , Microtubules/drug effects , Microtubules/physiology , Models, Biological , Transfection
3.
Neuroscience ; 135(4): 1231-9, 2005.
Article in English | MEDLINE | ID: mdl-16165289

ABSTRACT

Investigation of the elements underlying synapse replacement after brain injury is essential for predicting the neural compensation that can be achieved after various types of damage. The growth-associated proteins superior cervical ganglion-10 and growth-associated protein-43 have previously been linked with structural changes in the corticostriatal system in response to unilateral deafferentation. To examine the regulation of this response, unilateral cortical aspiration lesion was carried out in combination with ipsilateral 6-hydroxydopamine lesion of the substantia nigra, and the time course of the contralateral cortical molecular response was followed. Unilateral cortical aspiration lesion in rats corresponds with an upregulation of superior cervical ganglion-10 mRNA at 3 and 10 days post-lesion, and protein, sustained from three to at least 27 days following lesion. With the addition of substantia nigra lesion, the response shifts to an upregulation of growth-associated protein-43 mRNA at 3 and 10 days post-lesion, and protein after 10 days. Nigral lesion alone does not alter contralateral expression of either gene. Likewise, motor function assessment using the rotorod test revealed no significant long-term deficits in animals that sustained only nigrostriatal damage, but cortical lesion was associated with a temporary deficit which was sustained when nigrostriatal input was also removed. Growth-associated protein-43 and superior cervical ganglion-10, two presynaptic genes that are postulated to play roles in lesion-induced sprouting, are differentially upregulated in corticostriatal neurons after cortical versus combined cortical/nigral lesions. The shift in contralateral gene response from superior cervical ganglion-10 to growth-associated protein-43 upregulation and associated behavioral deficit following combined cortical and nigral denervation suggest that nigrostriatal afferents regulate cortical lesion-induced gene expression and ultimate functional outcome.


Subject(s)
Brain Injuries/metabolism , Cerebral Cortex/metabolism , GAP-43 Protein/biosynthesis , Nerve Growth Factors/biosynthesis , Substantia Nigra/metabolism , Animals , Blotting, Western , Brain Injuries/physiopathology , Carrier Proteins , Cerebral Cortex/injuries , Functional Laterality , Gene Expression , In Situ Hybridization , Male , Membrane Proteins , Microtubule Proteins , Motor Activity/physiology , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Substantia Nigra/injuries , Up-Regulation
4.
Exp Neurol ; 192(2): 340-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15755551

ABSTRACT

The expression of the transcription factor ATF3 in the brain was examined by immunohistochemistry during axonal regeneration induced by the implantation of pieces of peripheral nerve into the thalamus of adult rats. After 3 days, ATF3 immunoreactivity was present in many cells within approximately 500 mum of the graft. In addition, ATF3-positive cell nuclei were found in the thalamic reticular nucleus (TRN) and medial geniculate nuclear complex (MGN), from which most regenerating axons originate. CNS cells with ATF3-positive nuclei were predominantly neurons and did not show signs of apoptosis. The number of ATF3-positive cells had declined by 7 days and further by 1 month after grafting when most ATF3-positive cells were found in the TRN and MGN. 14 days or more after grafting, some ATF3-positive nuclei were distorted and may have been apoptotic. In some experiments of 1 month duration, neurons which had regenerated axons to the distal ends of grafts were retrogradely labeled with DiAsp. ATF3-positive neurons in these animals were located in regions of the TRN and MGN containing retrogradely labeled neurons and the great majority were also labeled with DiAsp. SCG10 and c-Jun were found in neurons in the same regions as retrogradely labeled and ATF3-positive cells. Thus, ATF3 is transiently upregulated by injured CNS neurons, but prolonged expression is part of the pattern of gene expression associated with axonal regeneration. The co-expression of ATF3 with c-jun suggests that interactions between these transcription factors may be important for controlling the program of gene expression necessary for regeneration.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Neurons/metabolism , Peripheral Nerves/transplantation , Thalamus/cytology , Transcription Factors/metabolism , Activating Transcription Factor 3 , Animals , Axons/transplantation , Carrier Proteins , Female , Immunohistochemistry/methods , Membrane Proteins , Microtubule Proteins , Nerve Growth Factors/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Pyridinium Compounds , Rats , Rats, Sprague-Dawley , Thalamus/transplantation , Time Factors , Tissue Transplantation/methods , Up-Regulation/physiology
5.
Mol Cell Neurosci ; 20(4): 595-615, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12213442

ABSTRACT

We have compared SCG10 and CAP-23 expression with that of GAP-43 during axonal regeneration in the peripheral and central nervous systems (PNS, CNS) of adult rats. SCG10, CAP-23, and GAP-43 mRNAs were strongly upregulated by motor and dorsal root ganglion (DRG) neurons following sciatic nerve crush, but not after dorsal rhizotomy. When the sciatic nerve was cut and ligated to prevent reinnervation of targets, expression of all three mRNAs was prolonged. Neurons in the thalamic reticular nucleus and deep cerebellar nuclei transiently upregulated these mRNAs after axotomy, and showed prolonged upregulation of all three molecules when regenerating axons into peripheral nerve grafts inserted into the thalamus of cerebellum. Neurons in the dorsal thalamus and cerebellar cortex showed poor regenerative capacity and most did not upregulate any of these mRNAs. Thus, in both PNS and CNS neurons, the transcription of SCG10, CAP-23, and GAP-43 mRNAs is coregulated following axotomy and during regeneration. Signals from living peripheral nerve appear to maintain expression of all three mRNAs in regenerating neurons, and in PNS neurons downregulation correlates with target reinnervation. Thus, SCG10 and CAP-23, as well as GAP-43, are likely to be important neuronal determinants of regenerative ability.


Subject(s)
Axons/physiology , Calmodulin-Binding Proteins , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Regeneration/physiology , Nerve Tissue Proteins , Transcription, Genetic/physiology , Up-Regulation , Animals , Carrier Proteins , Cerebellum/physiopathology , Cerebellum/surgery , Cytoskeletal Proteins/genetics , Female , GAP-43 Protein/genetics , Ganglia, Spinal/injuries , Ganglia, Spinal/metabolism , Membrane Proteins , Microtubule Proteins , Nerve Crush , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reference Values , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Spinal Cord/metabolism , Thalamic Nuclei/physiopathology , Thalamus/metabolism , Thalamus/surgery , Tibial Nerve/metabolism , Tibial Nerve/transplantation , Wounds and Injuries/metabolism
6.
FEBS Lett ; 508(2): 259-64, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11718727

ABSTRACT

The neuronal growth-associated protein SCG10 is enriched in the growth cones of neurons where it destabilizes microtubules and thus contributes to the dynamic assembly and disassembly of microtubules. Since its microtubule-destabilizing activity is regulated by phosphorylation, SCG10 may link extracellular signals to rearrangements of the neuronal cytoskeleton. To identify signal transduction pathways that may lead to SCG10 phosphorylation, we tested a series of serine-threonine-directed protein kinases that phosphorylate SCG10 in vitro. We demonstrate that purified SCG10 can be phosphorylated by two subclasses of mitogen-activated protein (MAP) kinases, c-Jun N-terminal/stress-activated protein kinase (JNK/SAPK) and p38 MAP kinase. Moreover, SCG10 was found to bind tightly and specifically to JNK3/SAPKbeta. JNK3/SAPKbeta phosphorylation occurs at Ser-62 and Ser-73, residues that result in reduced microtubule-destabilizing activity for SCG10. Endogenous SCG10 also undergoes increased phosphorylation in sympathetic neurons at times of JNK3/SAPKbeta activation following deprivation from nerve growth factor. Together these observations indicate that activation of JNK/SAPKs provides a pathway for phosphorylation of SCG10 and control of growth cone microtubule formation following neuronal exposure to cellular stresses.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Amino Acids/analysis , Animals , Blotting, Western , COS Cells , Calcium-Binding Proteins , Carrier Proteins , Growth Cones/metabolism , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Mass Spectrometry , Membrane Proteins , Mice , Microtubule Proteins , Mitogen-Activated Protein Kinase 10 , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Sequence Data , Nerve Growth Factors/chemistry , Phosphorylation , Protein Binding , Rats , Rats, Sprague-Dawley , Stathmin , Substrate Specificity , p38 Mitogen-Activated Protein Kinases
7.
J Comp Neurol ; 433(2): 239-54, 2001 Apr 30.
Article in English | MEDLINE | ID: mdl-11283962

ABSTRACT

The membrane-associated protein SCG10 is expressed specifically by neuronal cells. Recent experiments have suggested that it promotes neurite outgrowth by increasing microtubule dynamics in growth cones. SCG10 is related to the ubiquitous but neuron-enriched cytosolic protein stathmin. To better understand the role played by SCG10 and stathmin in vivo, we have analyzed the expression and localization of these proteins in both the olfactory epithelium and the olfactory bulb in developing and adult rats, as well as in adult bulbectomized rats. The olfactory epithelium is exceptional in that olfactory receptor neurons constantly regenerate and reinnervate the olfactory bulb throughout animal life-span. SCG10 and stathmin expression in the olfactory receptor neurons was found to be regulated during embryonic and postnatal development and to correlate with neuronal maturation. Whereas SCG10 expression was restricted to immature olfactory receptor neurons (GAP-43-positive, olfactory marker protein-negative), stathmin was also expressed by the basal cells. In the olfactory bulb of postnatal and adult rats, a moderate to strong SCG10 immunoreactivity was present in the olfactory nerve layer, whereas no labeling was detected in the glomerular layer. Olfactory glomeruli also showed no apparent immunoreactivity for several cytoskeletal proteins such as tubulin and microtubule-associated proteins. In unilaterally bulbectomized rats, SCG10 and stathmin were seen to be up-regulated in the regenerating olfactory epithelium at postsurgery stages corresponding to olfactory axon regeneration. Our data strongly suggest that, in vivo, both SCG10 and stathmin may play a role in axonal outgrowth during ontogenesis as well as during axonal regeneration.


Subject(s)
Axons/physiology , Microtubule Proteins , Nerve Growth Factors/metabolism , Nerve Regeneration/physiology , Olfactory Pathways/physiology , Phosphoproteins/metabolism , Rats/metabolism , Aging/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Carrier Proteins , Cytoskeletal Proteins/metabolism , Embryo, Mammalian/physiology , Embryonic and Fetal Development , GAP-43 Protein/metabolism , Membrane Proteins , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Olfactory Bulb/physiology , Olfactory Mucosa/metabolism , Rats/embryology , Rats, Wistar , Stathmin , Up-Regulation
8.
J Cell Biochem ; 80(3): 346-52, 2001.
Article in English | MEDLINE | ID: mdl-11135364

ABSTRACT

Stathmin/Op18 is a highly conserved 19 kDa cytosolic phosphoprotein. Human and chicken stathmin share 93% identity with only 11 amino acid substitutions. One of the substituted amino acids is serine 25, which is a glycine in chicken stathmin. In human stathmin, serine 25 is the main phosphorylation site for MAP kinase. In this study, we have compared the phosphorylation of human and chicken stathmin. The proteins were expressed in Sf9 cells using the baculovirus expression system and purified for in vitro phosphorylation assays. Phosphorylation with MAP kinase showed that chicken stathmin was phosphorylated 10 times less than human stathmin. To identify the phosphorylation sites we used liquid chromatography/mass spectrometry (LC/MS/MS). The only amino acid found phosphorylated was serine 38, which corresponds to the minor phosphorylation site in human stathmin. Phosphorylation with p34(cdc2)- and cGMP-dependent protein kinases gave almost identical phosphorylation levels in the two stathmins.


Subject(s)
Microtubule Proteins , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , Amino Acid Sequence , Animals , Humans , Mass Spectrometry , Molecular Sequence Data , Molecular Weight , Phosphoproteins/chemistry , Phosphoproteins/isolation & purification , Phosphorylation , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Stathmin
9.
J Struct Biol ; 131(2): 156-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11042086

ABSTRACT

Crystals of a complex formed between the alpha/beta-tubulin heterodimer and SCG10, a neuron-specific growth-associated protein, have been obtained by the hanging drop method. They belong to the space group P2(1)2(1)2(1), with unit cell parameters a = 56 A, b = 353 A, c = 466 A and four molecular complexes in the asymmetric unit. A complete X-ray diffraction data set to 6.1 A resolution has been collected using synchrotron radiation. This represents a challenging opportunity to study at a molecular level the structure-function relationships between a microtubule-destabilizing protein, SCG10, and tubulin.


Subject(s)
Nerve Growth Factors/chemistry , Nerve Growth Factors/metabolism , Tubulin/chemistry , Tubulin/metabolism , Animals , Brain/metabolism , Cattle , Crystallization , Dimerization , Macromolecular Substances , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Structure, Quaternary , X-Ray Diffraction
10.
Eur J Neurosci ; 12(7): 2224-34, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10947801

ABSTRACT

SCG10 is a membrane-associated, microtubule-destabilizing protein of neuronal growth cones. Using immunoelectron microscopy, we show that in the developing cortex of mice, SCG10 is specifically localized to the trans face Golgi complex and apparently associated with vesicular structures in putative growth cones. Consistent with this, subcellular fractionation of rat forebrain extracts demonstrates that the protein is enriched in the fractions containing the Golgi apparatus and growth cone particles. In isolated growth cone particles, SCG10 was found to be particularly concentrated in the growth cone vesicle fraction. To evaluate the molecular determinants of the specific targeting of SCG10 to growth cones, we have transfected PC12 cells and primary neurons in culture with mutant and fusion cDNA constructs. Deletion of the amino-terminal domain or mutations within this domain that prevented palmitoylation at cysteines 22 and 24 abolished Golgi localization as well as growth cone targeting, suggesting that palmitoylation of the amino-terminal domain is a necessary signal for Golgi sorting and possibly transport of SCG10 to growth cones. Fusion proteins consisting of the amino-terminal domain of SCG10 and the cytosolic proteins stathmin or glutathione-S-transferase colocalized with a Golgi marker, alpha-mannosidase II, and accumulated in growth cones of both axons and dendrites. These results reveal a novel axonal/dendritic growth cone targeting sequence that involves palmitoylation.


Subject(s)
Golgi Apparatus/chemistry , Growth Cones/chemistry , Membrane Proteins , Nerve Growth Factors/analysis , Nerve Growth Factors/genetics , Animals , Calcium-Binding Proteins , Carrier Proteins , Cysteine/metabolism , Fluorescent Antibody Technique , Gene Deletion , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Growth Cones/metabolism , Growth Cones/ultrastructure , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred Strains , Microscopy, Immunoelectron , Microtubule Proteins , Mutagenesis/physiology , Nerve Growth Factors/chemistry , Nerve Tissue Proteins/analysis , PC12 Cells , Palmitic Acid/metabolism , Protein Sorting Signals/genetics , Protein Structure, Tertiary , Rats , Stathmin , Subcellular Fractions/chemistry , Synaptophysin/analysis , Synaptosomal-Associated Protein 25 , Transfection
12.
J Biol Chem ; 273(14): 8439-46, 1998 Apr 03.
Article in English | MEDLINE | ID: mdl-9525956

ABSTRACT

SCG10 is a neuron-specific, membrane-associated protein that is highly concentrated in growth cones of developing neurons. Previous studies have suggested that it is a regulator of microtubule dynamics and that it may influence microtubule polymerization in growth cones. Here, we demonstrate that in vivo, SCG10 exists in both phosphorylated and unphosphorylated forms. By two-dimensional gel electrophoresis, two phosphoisoforms were detected in neonatal rat brain. Using in vitro phosphorylated recombinant protein, four phosphorylation sites were identified in the SCG10 sequence. Ser-50 and Ser-97 were the target sites for protein kinase A, Ser-62 and Ser-73 for mitogen-activated protein kinase and Ser-73 for cyclin-dependent kinase. We also show that overexpression of SCG10 induces a disruption of the microtubule network in COS-7 cells. By expressing different phosphorylation site mutants, we have dissected the roles of the individual phosphorylation sites in regulating its microtubule-destabilizing activity. We show that nonphosphorylatable mutants have increased activity, whereas mutants in which phosphorylation is mimicked by serine-to-aspartate substitutions have decreased activity. These data suggest that the microtubule-destabilizing activity of SCG10 is regulated by phosphorylation, and that SCG10 may link signal transduction of growth or guidance cues involving serine/threonine protein kinases to alterations of microtubule dynamics in the growth cone.


Subject(s)
Brain/metabolism , Microtubules/metabolism , Nerve Growth Factors/metabolism , Amino Acid Sequence , Animals , Brain/ultrastructure , Carrier Proteins , Electrophoresis, Gel, Two-Dimensional , Membrane Proteins , Microtubule Proteins , Microtubules/ultrastructure , Molecular Sequence Data , Mutation , Nerve Growth Factors/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Rats , Rats, Wistar
13.
FEBS Lett ; 416(2): 149-52, 1997 Oct 20.
Article in English | MEDLINE | ID: mdl-9369201

ABSTRACT

Stathmin is a regulator of microtubule dynamics which undergoes extensive phosphorylation during the cell cycle as well as in response to various extracellular factors. Four serine residues are targets for protein kinases: Ser-25 and Ser-38 for proline-directed kinases such as mitogen-activated protein kinase and cyclin-dependent protein kinase, and Ser-16 and Ser-63 for cAMP-dependent protein kinase. We studied the effect of phosphorylation on the microtubule-destabilizing activity of stathmin and on its interaction with tubulin in vitro. We show that triple phosphorylation on Ser-16, Ser-25, and Ser-38 efficiently inhibits its activity and prevents its binding to tubulin.


Subject(s)
Microtubule Proteins , Microtubules/physiology , Phosphoproteins/metabolism , Protein Kinases/metabolism , Tubulin/metabolism , Animals , Binding Sites , Brain/ultrastructure , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle , Cloning, Molecular , Cross-Linking Reagents , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinases/metabolism , Humans , Kinetics , Microtubules/ultrastructure , Phosphorylation , Phosphoserine , Proline , Recombinant Proteins/metabolism , Serine , Stathmin , Swine
14.
Protein Expr Purif ; 9(3): 363-71, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9126608

ABSTRACT

SCG10 is a neuron-specific, developmentally regulated protein which is highly enriched in growth cones. Sequence homology indicates that it is related to the phosphoprotein stathmin or Op18, an in vitro and in vivo substrate for several serine/threonine kinases which are involved in a variety of signaling pathways. As a first step to examine the biochemical properties of SCG10, the protein was expressed in Escherichia coli and purified to apparent homogeneity. The purified protein was used in in vitro phosphorylation assays. SCG10 was phosphorylated by MAP kinase, cAMP-dependent protein kinase, cGMP-dependent protein kinase, p34cdc2 kinase, DNA-dependent protein kinase, Ca2+/calmodulin kinase II, and casein kinase II. The protein was not a substrate for casein kinase I and protein kinase C. SCG10 was phosphorylated by src tyrosine kinase, which demonstrates that the protein can be phosphorylated in vitro on a tyrosine residue. Our data suggest that SCG10 is a phosphoprotein which might be involved in signal transduction in neurons.


Subject(s)
Microtubule Proteins , Nerve Growth Factors/isolation & purification , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins , DNA, Recombinant/genetics , Escherichia coli/genetics , Gene Expression , Membrane Proteins , Molecular Sequence Data , Molecular Weight , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurons/metabolism , Phosphoproteins/genetics , Phosphorylation , Plasmids/genetics , Rats , Sequence Homology, Amino Acid , Signal Transduction , Stathmin
15.
Protein Expr Purif ; 9(2): 295-300, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9056495

ABSTRACT

SCG10 is a neuron-specific growth-associated protein with high sequence homology to the ubiquitous phosphoprotein stathmin/Op18. The main structural difference between the two proteins is the 34-amino-acid N-terminal extension of SCG10, which is responsible for the membrane attachment. Full length SCG10 has been purified and shows limited solubility, in contrast to stathmin, which is a highly soluble protein. In order to obtain a more soluble form of SCG10 which would be better suited for biochemical and structural studies, we deleted the N-terminal extension and expressed the C-terminal portion of the protein. Two forms of N-terminal-truncated SCG10 (delta SCG10 and delta SCG10r) were purified to homogeneity in a four-step purification procedure. delta SCG10 starts at amino acid 35 and delta SCG10r at amino acid 48 in the SCG10 sequence, giving proteins of 16,899 and 15,189 kDa, respectively. The truncated SCG10 was highly soluble up to concentrations of 20 mg/ml. The proteins were like the full length SCG10 substrate for serine/threonine protein kinases, including MAP kinase, PKA, and p34cdc2 kinase. With these highly soluble forms of SCG10 biochemical and structural studies of this multiphosphoprotein become feasible.


Subject(s)
Membrane Proteins/biosynthesis , Membrane Proteins/isolation & purification , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/isolation & purification , Phosphoproteins/biosynthesis , Phosphoproteins/isolation & purification , Amino Acid Sequence , Animals , Carrier Proteins , Chromatography, Agarose , Chromatography, High Pressure Liquid , Circular Dichroism , Cloning, Molecular , Escherichia coli/chemistry , Escherichia coli/genetics , Genetic Vectors/metabolism , Membrane Proteins/chemistry , Microtubule Proteins , Molecular Sequence Data , Nerve Growth Factors/chemistry , Phosphoproteins/chemistry , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Solubility , Superior Cervical Ganglion/chemistry
16.
J Biol Chem ; 272(8): 5175-82, 1997 Feb 21.
Article in English | MEDLINE | ID: mdl-9030585

ABSTRACT

SCG10 is a neuronal growth-associated protein that is concentrated in the growth cones of developing neurons. SCG10 shows a high degree of sequence homology to the ubiquitous phosphoprotein stathmin, which has been recently identified as a factor that destabilizes microtubules by increasing their catastrophe rate. Whereas stathmin is a soluble cytosolic protein, SCG10 is membrane-associated, indicating that the protein acts in a distinct subcellular compartment. Identifying the precise intracellular distribution of SCG10 as well as the mechanisms responsible for its specific targeting will contribute to elucidating its function. The main structural feature distinguishing the two proteins is that SCG10 contains an NH2-terminal extension of 34 amino acids. In this study, we have examined the intracellular distribution of SCG10 in PC12 cells and in transfected COS-7 cells and the role of the NH2-terminal domain in membrane-binding and intracellular targeting. SCG10 was found to be localized to the Golgi complex region. We show that the NH2-terminal region (residues 1-34) was necessary for membrane targeting and Golgi localization. Fusion proteins consisting of the NH2-terminal 34 amino acids of SCG10 and the related protein stathmin or the unrelated protein, beta-galactosidase, accumulated in the Golgi, demonstrating that this sequence was sufficient for Golgi localization. Biosynthetic labeling of transfected COS-7 cells with [3H]palmitic acid revealed that two cysteine residues contained within the NH2-terminal domain were sites of palmitoylation.


Subject(s)
Golgi Apparatus/metabolism , Nerve Growth Factors/metabolism , Animals , Binding Sites , Biological Transport , COS Cells , Carrier Proteins , Membrane Proteins , Microtubule Proteins , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , PC12 Cells , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
17.
Proc Natl Acad Sci U S A ; 94(2): 741-5, 1997 Jan 21.
Article in English | MEDLINE | ID: mdl-9012855

ABSTRACT

Dynamic assembly and disassembly of microtubules is essential for cell division, cell movements, and intracellular transport. In the developing nervous system, microtubule dynamics play a fundamental role during neurite outgrowth, elongation, and branching, but the molecular mechanisms involved are unknown. SCG10 is a neuron-specific protein that is membrane-associated and highly enriched in growth cones. Here we show that SCG10 binds to microtubules, inhibits their assembly, and can induce microtubule disassembly. We also show that SCG10 overexpression enhances neurite outgrowth in a stably transfected neuronal cell line. These data identify SCG10 as a key regulator of neurite extension through regulation of microtubule instability.


Subject(s)
Microtubules/physiology , Nerve Growth Factors/physiology , Nerve Tissue Proteins/physiology , Tubulin/physiology , Animals , Carrier Proteins , Cell Differentiation , Fluorescent Antibody Technique, Indirect , Light , Macromolecular Substances , Membrane Proteins , Microtubule Proteins , Neurites/ultrastructure , PC12 Cells , Rats , Scattering, Radiation , Swine
18.
J Neurosci Res ; 50(6): 1000-9, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9452014

ABSTRACT

The neuron-specific protein SCG10 and the ubiquitous protein stathmin are two members of a family of microtubule-destabilizing factors that may regulate microtubule dynamics in response to extracellular signals. To gain insight into the function of these proteins in the nervous system, we have compared their intracellular distribution in cortical neurons developing in culture. We have used double-immunofluorescence microscopy with specific antibodies for stathmin and SCG10 in combination with antibodies for axonal, microtubule, and synaptic marker proteins. Stathmin and SCG10 were coexpressed in individual neurons. While both proteins were highly expressed in developing cultures during differentiation, their subcellular localization was strikingly different. Stathmin showed a cytosolic distribution, mainly in cell bodies, whereas SCG10 strongly labeled the growth cones of axons and dendrites. During neurite outgrowth, SCG10 appeared as a single concentrated spot in a region of the growth cone where the microtubules are known to be particularly dynamic. Disassembly of labile microtubules by nocodazole caused a dispersal of the SCG10 staining into punctate structures, indicating that its subcellular localization is microtubule-dependent. Upon maturation and synapse formation, the levels of both stathmin and SCG10 decreased to become undetectable. These observations demonstrate that the expression of both proteins is associated with neurite outgrowth and suggest that they perform their roles in this process in distinct subcellular compartments.


Subject(s)
Cerebral Cortex/chemistry , Microtubule Proteins , Nerve Growth Factors/analysis , Neurons/chemistry , Phosphoproteins/analysis , Animals , Carrier Proteins , Cells, Cultured , Cellular Senescence/physiology , Cerebral Cortex/cytology , Down-Regulation , Fluorescent Antibody Technique , Membrane Proteins , Microtubules/chemistry , Rats , Stathmin , Synapses/metabolism
19.
J Cell Biol ; 133(6): 1383-90, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8682872

ABSTRACT

Stathmin is a ubiquitous cytosolic protein which undergoes extensive phosphorylation in response to a variety of external signals. It is highly abundant in developing neurons. The use of antisense oligonucleotides which selectively block stathmin expression has allowed us to study directly its role in rat PC12 cells. We show that stathmin depletion prevents nerve growth factor (NGF)-stimulated differentiation of PC12 cells into sympathetic-like neurons although the expression of several NGF-inducible genes was not affected. Furthermore, we found that stathmin phosphorylation in PC12 cells which is induced by NGF depends on mitogen-activated protein kinase (MAPK) activity. We conclude that stathmin is an essential component of the NGF-induced MAPK signaling pathway and performs a key role during differentiation of developing neurons.


Subject(s)
Membrane Glycoproteins , Microtubule Proteins , Nerve Growth Factors/physiology , Neurons/cytology , Phosphoproteins/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Carrier Proteins , Cell Differentiation/drug effects , Cell Differentiation/physiology , Enzyme Inhibitors/pharmacology , Eye Proteins/biosynthesis , Flavonoids/pharmacology , Gene Expression/drug effects , Intermediate Filament Proteins/biosynthesis , Membrane Proteins , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/biosynthesis , Neurites , Neuropeptides , Oligonucleotides, Antisense/pharmacology , PC12 Cells , Peripherins , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Rats , Signal Transduction/physiology , Stathmin , Thionucleotides/pharmacology
20.
J Biol Chem ; 271(14): 8260-5, 1996 Apr 05.
Article in English | MEDLINE | ID: mdl-8626520

ABSTRACT

M-CAT motifs mediate muscle-specific transcriptional activity via interaction with binding factors that are antigenically and biochemically related to vertebrate transcription enhancer factor-1 (TEF-1), a member of the TEA/ATTS domain family of transcription factors. M-CAT binding activities present in cardiac and skeletal muscle tissues cannot be fully accounted for by existing cloned isoforms of TEF-1. TEF-1-related cDNAs isolated from heart libraries indicate that at least three classes of TEF-1-related cDNAs are expressed in these and other tissues. One class are homologues of the human TEF-1 originally cloned from HeLa cells (Xiao, J. H., Davidson, I., Matthes, H., Garnier, J. M., and Chambon, P. (1991) Cell 65, 551-568). A second class represents homologues of the avian TEF-1-related gene previously isolated (Stewart, A. F., Larkin, S. B., Farrance, I. K., Mar, J. H., Hall, D. E., and Ordahl, C. P. (1994) J. Biol. Chem. 269, 3147-3150). The third class consists of a novel, divergent TEF-1 cDNA, named DTEF-1, and its preliminary characterization is described here. Two isoforms of DTEF-1 (DTEF-1A and DTEF-1B) were isolated as 1.9-kilobase pair clones with putative open reading frames of 433 and 432 amino acids whose differences are attributable to alternative splicing at the C terminus of the TEA DNA binding domain. Cardiac muscle contains high levels of DTEF-1 transcripts, but unexpectedly low levels are detected in skeletal muscle. DTEF-1 transcripts are present at intermediate levels in gizzard and lung, and at low levels in kidney. DTEF-1A is a sequence-specific M-CAT-binding factor. The distinct spatial pattern of expression, and unusual amino acid sequence in its DNA binding domain, may indicate a particular role for DTEF-1 in cell-specific gene regulation. Recent work also suggests that at least one more TEF-1-related gene exists in vertebrates. We propose a naming system for the four TEF-1 gene family members identified to date that preserves existing nomenclature and provides a means for extending that nomenclature as additional family members may be identified.


Subject(s)
DNA-Binding Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Chickens , DNA Primers/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Molecular Sequence Data , Multigene Family , Myocardium/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid , TEA Domain Transcription Factors , Transcription Factors/chemistry , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...