Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 718(1-3): 383-92, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23978568

ABSTRACT

We have investigated the effects of tCFA15, a non-peptidic compound, on the differentiation of neural stem cell-derived neurospheres, and have found that tCFA15 promotes their differentiation into neurons and reduces their differentiation into astrocytes, in a dose-dependent manner. This response is reminiscent of that resulting from the loss-of-function of Notch signaling after inactivation of the Delta-like 1 (Dll1) gene. Further analysis of the expression of genes from the Notch pathway by reverse transcriptase-PCR revealed that tCFA15 treatment results in a consistent decrease in the level of Notch1 mRNA. We have confirmed this result in other cell lines and propose that it reflects a general effect of the tCFA15 molecule. We discuss the implications of this finding with respect to regulation of Notch activity in neural stem cells, and the possible use of tCFA15 as a therapeutic tool for various pathologies that result from impairment of Notch signaling.


Subject(s)
Cell Differentiation/drug effects , Cyclohexanones/pharmacology , Fatty Alcohols/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Receptor, Notch1/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Gene Expression Regulation/drug effects , Mice , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Notch1/genetics
2.
Exp Neurol ; 225(1): 60-73, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20451518

ABSTRACT

Tenascin-C (Tnc) is a multimodular extracellular matrix glycoprotein that is markedly upregulated in CNS injuries where it is primarily secreted by reactive astrocytes. Different Tnc isoforms can be generated by the insertion of variable combinations of one to seven (in rats) alternatively spliced distinct fibronectin type III (FnIII) domains to the smallest variant. Each spliced FnIII repeat mediates specific actions on neurite outgrowth, neuron migration or adhesion. Hence, different Tnc isoforms might differentially influence CNS repair. We explored the expression pattern of Tnc variants after cortical lesions and after treatment of astrocytes with various cytokines. Using RT-PCR, we observed a strong upregulation of Tnc transcripts containing the spliced FnIII domains B or D in injured tissue at 2-4 days post-lesion (dpl). Looking at specific combinations, we showed a dramatic increase of Tnc isoforms harboring the neurite outgrowth-promoting BD repeat with both the B and D domains being adjacent to each other. Isoforms containing only the axon growth-stimulating spliced domain D were also dramatically enhanced after injury. Injury-induced increase of Tnc proteins comprising the domain D was confirmed by Western Blotting and immunostaining of cortical lesions. In contrast, the FnIII modules C and AD1 were weakly modulated after injury. The growth cone repulsive A1A2A4 domains were poorly expressed in normal and injured tissue but the smallest isoform, which is also repellant, was highly expressed after injury. Expression of the shortest Tnc isoform and of variants containing B, D or BD, was strongly upregulated in cultured astrocytes after TGFbeta1 treatment, suggesting that TGFbeta1 could mediate, at least in part, the injury-induced upregulation of these isoforms. We identified complex injury-induced differential regulations of Tnc isoforms that may well influence axonal regeneration and repair processes in the damaged CNS.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Fibronectins/metabolism , Tenascin/metabolism , Alternative Splicing/genetics , Animals , Animals, Newborn , Astrocytes/pathology , Brain Injuries/genetics , Brain Injuries/pathology , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/physiology , Cells, Cultured , Contactins , Disease Models, Animal , Female , Fibronectins/genetics , Fibronectins/physiology , Nerve Regeneration/physiology , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Rats , Rats, Sprague-Dawley , Tenascin/genetics , Tenascin/physiology
3.
Glia ; 57(8): 860-74, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19053060

ABSTRACT

Prominin-1 (CD133) is a cholesterol-interacting pentaspan membrane glycoprotein specifically associated with plasma membrane protrusions. Prominin-1 is expressed by various stem and progenitor cells, notably neuroepithelial progenitors found in the developing embryonic brain. Here, we further investigated its expression in the murine brain. Biochemical analyses of brain membranes at early stages of development revealed the expression of two distinct splice variants of prominin-1, s1 and s3, which have different cytoplasmic C-terminal domains. The relative abundance of the s3 variant increased toward adulthood, whereas the opposite was observed for the s1 variant. Our combined in situ hybridization and immunohistochemistry revealed the expression of prominin-1 in a subpopulation of Olig-2-positive oligodendroglial cells present within white matter tracts of postnatal and adult brain. Furthermore, immunohistological and biochemical characterization suggested strongly that the s3 variant is a novel component of myelin. Consistent with this, the expression of prominin-1.s3 was significantly reduced in the brain of myelin-deficient mice. Finally, oligodendrocytes expressed selectively the s3 variant whereas GFAP-positive astrocytes expressed the s1 variant in primary glial cell cultures derived from embryonic brains. Collectively, our data demonstrate a complex expression pattern of prominin-1 molecules in developing adult brain. Given that prominin-1 is thought to act as an organizer of plasma membrane protrusions, they further suggest that a specific prominin-1 splice variant might play a role in morphogenesis and/or maintenance of the myelin sheath.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Glycoproteins/metabolism , Neuroglia/metabolism , Peptides/genetics , Peptides/metabolism , AC133 Antigen , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Brain/metabolism , Cells, Cultured , Chlorocebus aethiops , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/metabolism , Kidney/cytology , Mice , Mice, Jimpy , Mice, Mutant Strains , Microscopy, Immunoelectron/methods , Myelin Basic Protein/deficiency , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Nerve Tissue Proteins/metabolism , Neuroglia/ultrastructure , Oligodendrocyte Transcription Factor 2 , Optic Nerve/metabolism , Optic Nerve/ultrastructure , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transfection
4.
J Comp Neurol ; 504(6): 659-79, 2007 Oct 20.
Article in English | MEDLINE | ID: mdl-17722031

ABSTRACT

The chondroitin sulfate proteoglycan DSD-1-PG/phosphacan represents one of four splice variants of receptor-protein-tyrosine-phosphatase-beta/zeta (RPTPbeta/zeta). This receptor is expressed by glial cells and occurs in two isoforms, RPTPbeta(long) and RPTPbeta(short). The secreted forms phosphacan and phosphacan short isoform (PSI) bind to extracellular matrix and adhesion molecules and might mediate astroglial effects on neuronal differentiation. Phosphacan and RPTPbeta(long) both carry the DSD-1 epitope, a glycosaminoglycan modification that is involved in stimulating neurite outgrowth of embryonic rat mesencephalic and hippocampal neurons in a polycationic environment. Additionally, phosphacan inhibits neurite outgrowth of embryonic DRG neurons in the presence of laminin. In the light of these functional properties we examined the expression patterns of the DSD-1 epitope and phosphacan isoforms in the developing mouse visual system. During retinal development the DSD-1 epitope appears around embryonic day (E)13, peaks around postnatal day (P)6, and is downregulated from P9 to adolescence. By comparison, the phosphacan core protein is first detectable at E12, reaches maximal levels around P14, and persists, although at lower levels, to adulthood. The DSD-1 epitope is restricted to the nerve fiber and the inner plexiform layers. In contrast, the phosphacan core protein immunoreactivity extends from the nerve fiber layer to the outer plexiform layer. The level of expression of the phosphacan/RPTPbeta gene was investigated by reverse-transcriptase polymerase chain reaction. These experiments suggest that there is a shift in the expression patterns of the different phosphacan/RPTPbeta isoforms during late embryonic and postnatal development. In situ hybridization experiments support the conclusion that at least one of the phosphacan/RPTPbeta isoforms in the retina is expressed by neurons.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Visual Pathways , Animals , Animals, Newborn , Embryo, Mammalian , Female , Male , Mice , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Visual Pathways/embryology , Visual Pathways/growth & development , Visual Pathways/metabolism
5.
Mol Cell Neurosci ; 36(1): 71-85, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17689979

ABSTRACT

A unique and unresolved property of the central nervous system is that its extracellular matrix lacks fibrillar elements. In the present report, we show that astrocytes secrete triple helices of fibrillar collagens type I, III and V in culture, while no astroglial collagen expression could be detected in vivo. We discovered two inhibitory mechanisms that could underlie this apparent discrepancy. Thus, we uncover a strong inhibitory effect of meningeal cells on astrocytic collagen expression in coculture assays. Furthermore, we present evidence that EGF-receptor activation downregulates collagen expression in astrocytes via an autocrine loop. These investigations provide a rational framework to explain why the brain is devoid of collagen fibers, which is a unique feature that characterizes the structure of the neural extracellular matrix. Moreover, fibrillar collagens were found transiently upregulated in a laser-induced cortical lesion, suggesting that these could contribute to the glial scar that inhibits axonal regeneration.


Subject(s)
Astrocytes/drug effects , Autocrine Communication/physiology , Epidermal Growth Factor/pharmacology , Fibrillar Collagens/metabolism , Gene Expression Regulation/drug effects , Meninges/cytology , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/radiation effects , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques/methods , Culture Media, Conditioned/pharmacology , Cytokines/pharmacology , Gene Expression Regulation/physiology , Lasers/adverse effects , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tyrphostins/pharmacology
6.
Adv Exp Med Biol ; 557: 25-53, 2006.
Article in English | MEDLINE | ID: mdl-16955703

ABSTRACT

Interactions between neurons and glial cells play important roles in regulating key events of development and regeneration of the CNS. Thus, migrating neurons are partly guided by radial glia to their target, and glial scaffolds direct the growth and directional choice of advancing axons, e.g., at the midline. In the adult, reactive astrocytes and myelin components play a pivotal role in the inhibition of regeneration. The past years have shown that astrocytic functions are mediated on the molecular level by extracellular matrix components, which include various glycoproteins and proteoglycans. One important, developmentally regulated chondroitin sulfate proteoglycan is DSD-1-PG/phosphacan, a glial derived proteoglycan which represents a splice variant of the receptor protein tyrosine phosphatase (RPTP)-beta (also known as PTP-zeta). Current evidence suggests that this proteoglycan influences axon growth in development and regeneration, displaying inhibitory or stimulatory effects dependent on the mode of presentation, and the neuronal lineage. These effects seem to be mediated by neuronal receptors of the Ig-CAM superfamily.


Subject(s)
Central Nervous System , Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfates/metabolism , Nerve Tissue Proteins/metabolism , Protein Isoforms/metabolism , Protein Tyrosine Phosphatases/metabolism , Regeneration/physiology , Animals , Cell Line , Central Nervous System/cytology , Central Nervous System/pathology , Central Nervous System/physiology , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfates/genetics , Extracellular Matrix/metabolism , Humans , Ligands , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/physiology , Protein Isoforms/genetics , Protein Tyrosine Phosphatases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Transforming Growth Factor beta/metabolism
7.
Cell Tissue Res ; 321(3): 323-33, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16028071

ABSTRACT

Phosphacan is a chondroitin sulfate proteoglycan representing the secreted extracellular part of a transmembrane receptor protein tyrosine phosphatase (RPTP-beta). These isoforms have been implicated in cell-extracellular matrix signaling events associated with myelination, axon growth, and cell migration in the developing central nervous system and may play critical roles in the context of brain pathologies. Recently, we have reported the identification of a new isoform of phosphacan, the phosphacan short isoform (PSI), the expression of which peaks in the second postnatal week. PSI interacts with the neuronal receptors L1 and F3/contactin and can promote neurite growth of cortical neurons. In this study, we have assessed, by in situ hybridization, the expression profile of PSI in the rat brain at postnatal day 7. PSI is largely expressed in the gray matter of the developing cerebral cortex in which it colocalizes with phosphacan, whereas the expression of RPTPbeta receptor forms is restricted to the ventricular area in which PSI has not been observed. Neurons from all layers of the cortex express PSI. In the cerebellum, on the other hand, no expression of PSI has been detected, although the other phosphacan/RPTP-beta isoforms show strong PSI expression here. Overall, our study suggests that PSI is expressed during the postnatal period in differentiated neurons of the cortex but is absent from structures in which proliferation and migration occur. The significance of these observations is discussed in the context of previous models of phosphacan/RPTP-beta functions.


Subject(s)
Cerebral Cortex/cytology , Chondroitin Sulfate Proteoglycans/metabolism , Neurons/metabolism , Protein Isoforms/metabolism , Animals , Cell Fractionation , Cerebellum/cytology , Cerebellum/metabolism , Cerebral Cortex/metabolism , Chondroitin Sulfate Proteoglycans/genetics , In Situ Hybridization , Neurons/cytology , Protein Isoforms/genetics , Rats , Rats, Wistar , Receptor-Like Protein Tyrosine Phosphatases, Class 5
8.
Eur J Neurosci ; 20(10): 2524-40, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15548197

ABSTRACT

Analysis of Tenascin-C (TN-C) knockout mice revealed novel roles for this extracellular matrix (ECM) protein in regulation of the developmental programme of oligodendrocyte precursor cells (OPCs), their maturation into myelinating oligodendrocytes and sensitivity to growth factors. A major component of the ECM of developing nervous tissue, TN-C was expressed in zones of proliferation, migration and morphogenesis. Examination of TN-C knockout mice showed roles for TN-C in control of OPC proliferation and migration towards zones of myelination [E. Garcion et al. (2001) Development, 128, 2485-2496]. Extending our studies of TN-C effects on OPC development we found that OPCs can endogenously express TN-C protein. This expression covered the whole range of possible TN-C isoforms and could be strongly up-regulated by leukaemia inhibitory factor and ciliary neurotrophic factor, cytokines known to modulate OPC proliferation and survival. Comparative analysis of TN-C knockout OPCs with wild-type OPCs reveals an accelerated rate of maturation in the absence of TN-C, with earlier morphological differentiation and precocious expression of myelin basic protein. TN-C knockout OPCs plated on poly-lysine displayed higher levels of apoptosis than wild-type OPCs and there was also an earlier loss of responsiveness to the protective effects of platelet-derived growth factor (PDGF), indicating that TN-C has anti-apoptotic effects that may be associated with PDGF signalling. The existence of mechanisms to compensate for the absence of TN-C in the knockout is indicated by the development of oligodendrocytes derived from TN-C knockout neurospheres. These were present in equivalent proportions to those found in wild-type neurospheres but displayed enhanced myelin membrane formation.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Oligodendroglia/metabolism , Platelet-Derived Growth Factor/pharmacology , Stem Cells/metabolism , Tenascin/physiology , Animals , Animals, Newborn , Antigens/metabolism , Blotting, Western/methods , Brain/cytology , Brain/embryology , Brain/growth & development , Brain/metabolism , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Chondroitin Sulfates/metabolism , Cytokines/pharmacology , Embryo, Mammalian , Humans , Immunohistochemistry/methods , In Situ Hybridization/methods , In Situ Nick-End Labeling/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/drug effects , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Protein Tyrosine Phosphatases/metabolism , Proteoglycans/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cells/drug effects , Tenascin/genetics , Time Factors
9.
Mol Cell Neurosci ; 24(4): 951-71, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14697661

ABSTRACT

Several chondroitin sulfate proteoglycans (CSPGs) are upregulated after CNS injury and are thought to limit axonal regeneration in the adult mammalian CNS. Therefore, we examined the expression of the CSPG, receptor protein tyrosine phosphatase beta (RPTPbeta)/phosphacan, after a knife lesion to the cerebral cortex and after treatment of glial cultures with regulatory factors. The three splice variants of this CSPG gene, the secreted isoform, phosphacan, and the two transmembrane isoforms, the long and short RPTPbeta, were examined. Western blot and immunostaining analysis of injured and uninjured tissue revealed a transient decrease of phosphacan protein levels, but not of short RPTPbeta, in the injured tissue from 1 to 7 days postlesion (dpl). By real time RT-PCR, we show that phosphacan and long RPTPbeta mRNA levels are transiently down-regulated at 2 dpl, unlike those of short RPTPbeta which increased after 4 dpl. In contrast to the core glycoprotein, the phosphacan chondroitin sulfate (CS) glycosaminoglycan epitope DSD-1 was up-regulated after 7 dpl. Phosphacan was expressed by cultivated astrocytes and oligodendrocyte precursors but was more glycanated in oligodendrocyte precursors, which produce more of DSD-1 epitope than astrocytes. Epidermal growth factor/transforming growth factor alpha strongly increased the astrocytic expression of long RPTPbeta and phosphacan and slightly the short RPTPbeta protein levels, while interferon gamma and tumor necrosis factor alpha reduced astrocytic levels of phosphacan, but not of the receptor forms. Examining the effects of phosphacan on axon growth from rat E17 cortical neurons, we found that phosphacan stimulates outgrowth in a largely CS dependent manner, while it blocks the outgrowth-promoting effects of laminin through an interaction that is not affected by removal of the CS chains. These results demonstrate complex injury-induced modifications in phosphacan expression and glycanation that may well influence axonal regeneration and repair processes in the damaged CNS.


Subject(s)
Brain Injuries/enzymology , Chondroitin Sulfate Proteoglycans/biosynthesis , Glycosaminoglycans/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neuroglia/enzymology , Protein Tyrosine Phosphatases/biosynthesis , Animals , Brain Injuries/genetics , Brain Injuries/metabolism , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/pharmacology , Cytokines/pharmacology , Epitopes/biosynthesis , Epitopes/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glycosaminoglycans/genetics , Humans , Nerve Tissue Proteins/genetics , Neuroglia/drug effects , Neuroglia/metabolism , Protein Tyrosine Phosphatases/genetics , Rats , Rats, Sprague-Dawley , Receptor-Like Protein Tyrosine Phosphatases, Class 5
10.
J Biol Chem ; 278(26): 24164-73, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12700241

ABSTRACT

Phosphacan, one of the principal proteoglycans in the extracellular matrix of the central nervous system, is implicated in neuron-glia interactions associated with neuronal differentiation and myelination. We report here the identification of a novel truncated form of phosphacan, phosphacan short isoform (PSI), that corresponds to the N-terminal carbonic anhydrase- and fibronectin type III-like domains and half of the spacer region. The novel cDNA transcript was isolated by screening of a neonatal brain cDNA expression library using a polyclonal antibody raised against phosphacan. Expression of this transcript in vivo was confirmed by Northern blot hybridization. Analysis of brain protein extracts reveals the presence of a 90-kDa glycosylated protein in the phosphate-buffered saline-insoluble 100000 x g fraction that reacts with antisera against both phosphacan and a recombinant PSI protein and that has the predicted N-terminal sequence. This protein is post-translationally modified with oligosaccharides, including the HNK-1 epitope, but, unlike phosphacan, it is not a proteoglycan. The expression of the PSI protein varies during central nervous system development in a fashion similar to that observed for phosphacan, being first detected around embryonic day 16 and then showing a dramatic increase in expression to plateau around the second week post-natal. Both the native and recombinant PSI protein can interact with the Ig cell adhesion molecules, F3/contactin and L1, and in neurite outgrowth assays, the PSI protein can promote outgrowth of cortical neurons when used as a coated substrate. Hence, the identification of this novel isoform of phosphacan/receptor protein tyrosine phosphatase-beta provides a new component in cell-cell and cell-extracellular matrix signaling events in which these proteins have been implicated.


Subject(s)
Chondroitin Sulfate Proteoglycans/physiology , Neurites , Neurons/chemistry , Receptors, Cell Surface/metabolism , Animals , Base Sequence , Brain/cytology , Brain/embryology , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Embryo, Mammalian , Genetic Variation , Mice , Mice, Inbred Strains , Molecular Sequence Data , Nerve Tissue Proteins , Neurons/ultrastructure , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Processing, Post-Translational , Protein Tyrosine Phosphatases , RNA, Messenger/analysis , RNA, Messenger/isolation & purification , Receptor-Like Protein Tyrosine Phosphatases, Class 5
11.
Glia ; 41(4): 382-92, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12555205

ABSTRACT

The use of monoclonal antibodies has led to much progress in the characterization of extracellular matrix components of the CNS. F1C3 is a monoclonal antibody raised against the astrocytic cell line, Neu7. Analysis by immunoprecipitation and Western blots of the F1C3 antigen in Neu7 cell lysates and conditioned medium reveals a recognition of several protein bands around 140-230 kD. Internal peptide sequence data from these bands indicate that they are highly homologous to fibrillar collagens, and the F1C3 antigen is specifically digested by the collagenase I protease. Other glial cell lines show F1C3 antigen expression including A7, C6, and U373. Cultures of neonatal primary astrocytes also express F1C3 antigen, and Western blot analysis of rat brain extracts from different ages and parts of the brain confirm an in vivo expression of F1C3 protein. The significance of the expression of fibrillar collagen-like proteins by astrocytes is discussed together with its possible implication during developmental processes and in the context of CNS lesions and regeneration.


Subject(s)
Astrocytes/metabolism , Fibrillar Collagens/biosynthesis , Animals , Astrocytes/cytology , Brain/cytology , Brain/metabolism , Cell Line , Cells, Cultured , Mice , Rats
12.
J Neurosci ; 22(15): 6596-609, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12151539

ABSTRACT

Tenascin-C is a multimodular glycoprotein that possesses neurite outgrowth-stimulating properties, and one functional site has been localized to the alternatively spliced fibronectin type III domain D. To identify the neuronal receptor that mediates this effect, neighboring pairs of fibronectin type III domains were expressed as hybrid proteins fused to the Fc fragment of human immunoglobulin. These IgFc fusions were tested for neurite outgrowth-promoting properties on embryonic day 18 rat hippocampal neurons, and both the combinations BD and D6 were shown to promote the elongation of the longest process, the prospective axon. Antibodies to the cell adhesion molecule F3/contactin of the Ig superfamily blocked the BD- but not the D6-dependent effect. Biochemical studies using F3/contactin-IgFc chimeric proteins confirmed that the adhesion molecule selectively reacts with the combination BD but not with other pairs of fibronectin type III repeats of tenascin-C. The alternatively spliced BD cassettes are prominently expressed in the developing hippocampus, as shown by reverse transcription PCR, and colocalize with F3 expression during perinatal periods when axon growth and the establishment of hippocampal connections take place. We conclude that F3/contactin regulates axon growth of hippocampal neurons in response to tenascin-C.


Subject(s)
Alternative Splicing/physiology , Cell Adhesion Molecules, Neuronal/metabolism , Hippocampus/drug effects , Neurons/drug effects , Tenascin/pharmacology , Amino Acid Motifs/physiology , Animals , Cells, Cultured , Contactins , Fibronectins/physiology , Hippocampus/cytology , Hippocampus/embryology , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Immunohistochemistry , Mice , Neurites/drug effects , Neurites/physiology , Neurons/cytology , Neurons/metabolism , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
J Neurocytol ; 31(6-7): 497-505, 2002.
Article in English | MEDLINE | ID: mdl-14501219

ABSTRACT

Molecular studies have demonstrated that the murine AN2 antigen is the mouse homologue of the rat NG2 and human MCSP protein. The molecule is a single-pass transmembrane protein which carries a variable number of glyco- and glycosaminoglycan chains according to cell type and developmental stage. AN2/NG2 has two extracellular Laminin G-like domains which are classically involved in cell adhesion and recognition. It possesses a single PDZ binding motif in the short intracellular tail. The AN2/NG2 antigen is expressed by glial progenitor cells in developing and adult CNS and also by immature Schwann cells. Antibodies against AN2/NG2 inhibit the migration of antigen-positive cells in in vitro assays, suggesting that the molecule plays a role in migration. Many AN2/NG2-positive cells surround synapses in the developing and adult brain. A recently identified intracellular partner of AN2/NG2 is the glutamate receptor interacting protein GRIP, which binds to the GluRB subunit of the AMPA subclass of glutamate receptors. The AN2/NG2 protein may position AMPA receptors on perisynaptic glial cells towards active synapses by binding to a neuronal receptor. Many highly migratory neural tumors including melanomas express AN2/NG2. In the demyelinating disease Multiple Sclerosis, some patients synthesise antibodies against the protein. Such antibodies may play a pathological role by inhibiting the migration of oligodendrocyte progenitor cells to demyelinated axons thus blocking remyelination, as well as possibly interfering with glial neuronal signalling at synapses and nodes of Ranvier.


Subject(s)
Antigens/metabolism , Brain/embryology , Brain/metabolism , Cell Movement/physiology , Neuroglia/metabolism , Proteoglycans/metabolism , Stem Cells/metabolism , Animals , Antigens, Surface/metabolism , Brain/cytology , Humans , Mice , Nerve Regeneration/physiology , Neuroglia/cytology , Rats , Receptors, AMPA/metabolism , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...