Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Glia ; 60(3): 393-403, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22379615

ABSTRACT

The analysis of the molecular mechanisms involved in the initial interaction between neurons and Schwann cells is a key issue in understanding the myelination process. We recently identified Cthrc1 (Collagen triple helix repeat containing 1) as a gene upregulated in Schwann cells upon interaction with the axon. Cthrc1 encodes a secreted protein previously shown to be involved in migration and proliferation in different cell types. We performed a functional analysis of Cthrc1 in Schwann cells by loss-of- and gain-of-function approaches using RNA interference knockdown in cell culture and a transgenic mouse line that overexpresses the gene. This work establishes that Cthrc1 enhances Schwann cell proliferation but prevents myelination. In particular, time-course analysis of myelin formation intransgenic animals reveals that overexpression of Cthrc1 in Schwann cells leads to a delay in myelin formation with cells maintaining a proliferative state. Our data, therefore, demonstrate that Cthrc1 plays a negative regulatory role, fine-tuning the onset of peripheral myelination.


Subject(s)
Extracellular Matrix Proteins/metabolism , Gene Expression Regulation/genetics , Schwann Cells/metabolism , Animals , Bromodeoxyuridine/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation , Cell Survival/drug effects , Cell Survival/genetics , Early Growth Response Protein 2/genetics , Embryo, Mammalian , Extracellular Matrix Proteins/genetics , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Electron , Myelin Basic Protein/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Schwann Cells/ultrastructure , Time Factors , Transfection
2.
PLoS One ; 6(1): e14533, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21267074

ABSTRACT

Chronic acquired neuropathies of unknown origin are classified as chronic inflammatory demyelinating polyneuropathies (CIDP) and chronic idiopathic axonal polyneuropathies (CIAP). The diagnosis can be very difficult, although it has important therapeutic implications since CIDP can be improved by immunomodulating treatment. The aim of this study was to examine the possible abnormalities of nodal and paranodal regions in these two types of neuropathies. Longitudinal sections of superficial peroneal nerves were obtained from biopsy material from 12 patients with CIDP and 10 patients with CIAP and studied by immunofluorescence and in some cases electron microscopy. Electron microscopy revealed multiple alterations in the nodal and paranodal regions which predominated in Schwann cells in CIDP and in axons in CIAP. In CIDP paranodin/Caspr immunofluorescence was more widespread than in control nerves, extending along the axon in internodes where it appeared intense. Nodal channels Nav and KCNQ2 were less altered but were also detected in the internodes. In CIAP paranodes, paranodin labeling was irregular and/or decreased. To test the consequences of acquired primary Schwann cells alteration on axonal proteins, we used a mouse model based on induced deletion of the transcription factor Krox-20 gene. In the demyelinated sciatic nerves of these mice we observed alterations similar to those found in CIDP by immunofluorescence, and immunoblotting demonstrated increased levels of paranodin. Finally we examined whether the alterations in paranodin immunoreactivity could have a diagnosis value. In a sample of 16 biopsies, the study of paranodin immunofluorescence by blind evaluators led to correct diagnosis in 70 ± 4% of the cases. This study characterizes for the first time the abnormalities of nodes of Ranvier in CIAP and CIDP, and the altered expression and distribution of nodal and paranodal proteins. Marked differences were observed between CIDP and CIAP and the alterations in paranodin immunofluorescence may be an interesting tool for their differential diagnosis.


Subject(s)
Polyneuropathies/pathology , Ranvier's Nodes/pathology , Animals , Axons , Cell Adhesion Molecules, Neuronal/analysis , Chronic Disease , Fluorescent Antibody Technique , Humans , Mice , Microscopy, Electron , Nerve Tissue Proteins/analysis , Schwann Cells/pathology
3.
Glia ; 59(3): 351-62, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21264944

ABSTRACT

The initial interaction between the Schwann cell and the axon is a complex and poorly understood aspect of the myelination process. To investigate the molecular mechanisms involved in this interaction and to identify novel genes required for myelination, we performed an RNA profiling analysis, comparing Schwann cells cultured alone or in the presence of neurons. This led to the selection of 30 genes, mostly upregulated on Schwann cell-axon interaction. Most of the identified proteins are associated with the extracellular space or signal transduction systems, consistent with possible roles in Schwann cell-axon interaction. We performed a functional analysis of one of these genes, Dok4 (downstream of kinase-4), which encodes a membrane-associated tyrosine kinase substrate. Silencing RNA-mediated knock-down of Dok4 severely affected in vitro myelination. Moreover, Dok4 is required at early stages in the myelination process, including the initial interaction with the axon, and is also involved in Schwann cell migration and proliferation. Finally, this analysis establishes the interest of our gene collection in further understanding of the molecular mechanisms involved in Schwann cell-axon interaction.


Subject(s)
Axons/metabolism , Cell Communication/physiology , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Myelin Sheath/physiology , Nerve Tissue Proteins/physiology , Phosphoproteins/physiology , Schwann Cells/metabolism , Adaptor Proteins, Signal Transducing , Animals , Axons/physiology , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Schwann Cells/cytology
4.
J Neurosci ; 30(17): 5958-67, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20427655

ABSTRACT

CNS/PNS interfaces constitute cell boundaries, because they delimit territories with different neuronal and glial contents. Despite their potential interest in regenerative medicine, the mechanisms restricting oligodendrocytes and astrocytes to the CNS and Schwann cells to the PNS in mammals are not known. To investigate the involvement of peripheral glia and myelin in the maintenance of the CNS/PNS boundary, we have first made use of different mouse mutants. We show that depletion of Schwann cells and boundary cap cells or inactivation of Krox20/Egr2, a master regulatory gene for myelination in Schwann cells, results in transgression of the CNS/PNS boundary by astrocytes and oligodendrocytes and in myelination of nerve root axons by oligodendrocytes. In contrast, such migration does not occur with the Trembler(J) mutation, which prevents PNS myelination without affecting Krox20 expression. Altogether, these data suggest that maintenance of the CNS/PNS boundary requires a Krox20 function separable from myelination control. Finally, we have analyzed a human patient affected by a congenital amyelinating neuropathy, associated with the absence of the KROX20 protein in Schwann cells. In this case, the nerve roots were also invaded by oligodendrocytes and astrocytes. This indicates that transgression of the CNS/PNS boundary by central glia can occur in pathological situations in humans and suggests that the underlying mechanisms are common with the mouse.


Subject(s)
Central Nervous System/physiology , Early Growth Response Protein 2/metabolism , Neuroglia/physiology , Peripheral Nervous System/physiology , Animals , Astrocytes/physiology , Astrocytes/ultrastructure , Axons/physiology , Axons/ultrastructure , Cell Movement/physiology , Central Nervous System/ultrastructure , Early Growth Response Protein 2/genetics , Humans , Infant , Mice , Mice, Transgenic , Myelin Sheath/physiology , Myelin Sheath/ultrastructure , Neuroglia/ultrastructure , Oligodendroglia/physiology , Oligodendroglia/ultrastructure , Peripheral Nervous System/ultrastructure , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Schwann Cells/physiology , Schwann Cells/ultrastructure , Spinal Nerve Roots/physiology , Spinal Nerve Roots/ultrastructure
5.
Bull Acad Natl Med ; 194(4-5): 743-4, 2010.
Article in French | MEDLINE | ID: mdl-21568047

ABSTRACT

CNS/PNS interfaces constitute cell boundaries, defining territories with different neuronal and glial contents. Despite their potential implications for regenerative medicine, the mechanisms that restrict oligodendrocytes and astrocytes to the CNS and Schwann cells to the PNS are not known in mammals. To investigate the involvement of peripheral glia and myelin in CNS/PNS boundary maintenance, we first studied mutant mice. We found that inactivation of Krox20/Egr2, a master regulatory gene for myelination in Schwann cells, resulted in CNS/PNS boundary transgression by astrocytes and oligodendrocytes, and in myelination of nerve root axons by oligodendrocytes. In contrast, no such migration was observed in mice with the Trembler(J) mutation, which prevents PNS myelination without affecting Krox20 expression. These findings suggest that CNS/PNS boundary maintenance requires a new Krox20 function independent of myelination control. We also examined a patient with congenital amyelinating neuropathy, whose Schwann cells lack KROX20 protein. Interestingly, the patient's nerve roots were also invaded by oligodendrocytes and astrocytes, indicating that CNS/PNS boundary transgression by central glia can occur in human pathological situations and that the underlying mechanisms are the same as in mutant mice.


Subject(s)
Central Nervous System/physiology , Early Growth Response Protein 2/physiology , Peripheral Nervous System/physiology , Humans
6.
J Neurosci ; 28(23): 5891-900, 2008 Jun 04.
Article in English | MEDLINE | ID: mdl-18524893

ABSTRACT

Krox20/Egr2 is a zinc finger transcription factor that plays essential roles in several developmental processes, including peripheral nervous system myelination by Schwann cells, where it acts as a master gene regulator. Krox20 is known to interact with cofactors of the Nab family and a mutation affecting isoleucine 268, which prevents this interaction, has been shown to result in congenital hypomyelinating neuropathy in humans. To further investigate the role of this interaction, we have introduced such a mutation, Krox20(I268F), in the mouse germ line. Clinical, immunohistochemical, and ultrastructural analyses of the homozygous mutants reveal that they develop a severe hypomyelination phenotype that mimics the human syndrome. Furthermore, a time-course analysis of the disease indicates that it follows a biphasic evolution, the hypomyelination phase being followed by a dramatic demyelination. Although for the regulation of most analyzed Krox20 target genes the mutation behaves as a loss of function, this is not the case for a few of them. This differential effect indicates that the molecular function of the Krox20-Nab interaction is target dependent and might explain the degradation of the residual myelin, because of imbalances in its composition. In conclusion, this work provides a novel and useful model for severe human peripheral neuropathies.


Subject(s)
Amino Acid Substitution/genetics , Early Growth Response Protein 2/genetics , Neoplasm Proteins/genetics , Peripheral Nervous System Diseases/genetics , Repressor Proteins/genetics , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Early Growth Response Protein 2/metabolism , Female , Male , Mice , Mice, Mutant Strains , Neoplasm Proteins/metabolism , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/physiology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Protein Binding/genetics , Repressor Proteins/metabolism , Time Factors
7.
Proc Natl Acad Sci U S A ; 104(11): 4694-9, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360586

ABSTRACT

In multiple sclerosis (MS), oligodendrocyte and myelin destruction lead to demyelination with subsequent axonal loss. Experimental demyelination in rodents has highlighted the activation of the subventricular zone (SVZ) and the involvement of progenitor cells expressing the polysialylated form of neural cell adhesion molecule (PSA-NCAM) in the repair process. In this article, we studied the distribution of early PSA-NCAM(+) progenitors in the SVZ and MS lesions in human postmortem brains. Compared with controls, MS SVZ showed a 2- to 3-fold increase in cell density and proliferation, which correlated with enhanced numbers of PSA-NCAM(+) and glial fibrillary acidic protein-positive (GFAP(+)) cells. PSA-NCAM(+) progenitors mainly were Sox9(+), and a few expressed Sox10 and Olig2, markers of oligodendroglial specification. PSA-NCAM(+) progenitors expressing Sox10 and Olig2 also were detected in demyelinated MS lesions. In active and chronic active lesions, the number of PSA-NCAM(+) progenitors was 8-fold higher compared with chronic silent lesions, shadow plaques, and normal-appearing white matter. In active and chronic active lesions, PSA-NCAM(+) progenitors were more frequent in periventricular lesions (30-50%) than in lesions remote from the ventricular wall. These data indicate that, as in rodents, activation of gliogenesis in the SVZ occurs in MS and suggest the mobilization of SVZ-derived early glial progenitors to periventricular lesions, where they could give rise to oligodendrocyte precursors. These early glial progenitors could be a potential target for therapeutic strategies designed to promote myelin repair in MS.


Subject(s)
Brain/metabolism , Multiple Sclerosis/metabolism , Neuroglia/cytology , Stem Cells/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , DNA-Binding Proteins/metabolism , Disease Progression , High Mobility Group Proteins/metabolism , Humans , Models, Biological , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Oligodendrocyte Transcription Factor 2 , SOX9 Transcription Factor , SOXE Transcription Factors , Transcription Factors/metabolism , Transcription, Genetic
8.
J Neurosci ; 26(38): 9771-9, 2006 Sep 20.
Article in English | MEDLINE | ID: mdl-16988048

ABSTRACT

Onset of myelination in Schwann cells is governed by several transcription factors, including Krox20/Egr2, and mutations affecting Krox20 result in various human hereditary peripheral neuropathies, including congenital hypomyelinating neuropathy (CHN) and Charcot-Marie-Tooth disease (CMT). Similar molecular information is not available on the process of myelin maintenance. We have generated conditional Krox20 mutations in the mouse that allowed us to develop models for CHN and CMT. In the latter case, specific inactivation of Krox20 in adult Schwann cells results in severe demyelination, involving rapid Schwann cell dedifferentiation and increased proliferation, followed by an attempt to remyelinate and a block at the promyelinating stage. These data establish that Krox20 is not only required for the onset of myelination but that it is also crucial for the maintenance of the myelinating state. Furthermore, myelin maintenance appears as a very dynamic process in which Krox20 may constitute a molecular switch between Schwann cell myelination and demyelination programs.


Subject(s)
Alleles , Early Growth Response Protein 2/biosynthesis , Gene Expression Regulation/physiology , Myelin Sheath/metabolism , Peripheral Nerves/metabolism , Animals , Cell Proliferation , Early Growth Response Protein 2/genetics , Mice , Mice, Transgenic , Mutation , Myelin Sheath/genetics , Myelin Sheath/ultrastructure , Peripheral Nerves/ultrastructure
9.
J Biol Chem ; 281(8): 5300-9, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16332675

ABSTRACT

Notch1 and beta1-integrins are cell surface receptors involved in the recognition of the niche that surrounds stem cells through cell-cell and cell-extracellular matrix interactions, respectively. Notch1 is also involved in the control of cell fate choices in the developing central nervous system (Lewis, J. (1998) Semin. Cell Dev. Biol. 9, 583-589). Here we report that Notch and beta1-integrins are co-expressed and that these proteins cooperate with the epidermal growth factor receptor in neural progenitors. We describe data that suggests that beta1-integrins may affect Notch signaling through 1) physical interaction (sequestration) of the Notch intracellular domain fragment by the cytoplasmic tail of the beta1-integrin and 2) affecting trafficking of the Notch intracellular domain via caveolin-mediated mechanisms. Our findings suggest that caveolin 1-containing lipid rafts play a role in the coordination and coupling of beta1-integrin, Notch1, and tyrosine kinase receptor signaling pathways. We speculate that this will require the presence of the adequate beta1-activating extracellular matrix or growth factors in restricted regions of the central nervous system and namely in neurogenic niches.


Subject(s)
ErbB Receptors/metabolism , Integrin beta1/metabolism , Neurons/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism , Animals , Brain/metabolism , Caveolin 1/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Central Nervous System/metabolism , Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/metabolism , Glutathione Transferase/metabolism , Growth Substances/metabolism , Immunohistochemistry , Immunoprecipitation , Manganese/chemistry , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors
10.
J Neurosci ; 24(15): 3816-25, 2004 Apr 14.
Article in English | MEDLINE | ID: mdl-15084663

ABSTRACT

Newly formed oligodendrocytes in the CNS derive survival cues from their target axons. These cues are provided in part by laminins expressed on the axon, which are recognized by alpha6beta1 integrin on the oligdendrocyte and amplify platelet-derived growth factor (PDGF) signaling through the phosphatidylinositol 3'-kinase (PI3K) pathway. The alpha6beta1 integrin is localized in oligodendrocyte lipid rafts. We show here using the sphingolipid synthesis inhibitor fumonisin-B1 to deplete rafts that this localization is important for normal survival signaling, because depletion increases oligodendrocyte apoptosis and inhibits PI3K signaling. We have shown previously that PDGF-mediated integrin activation is an important component of oligodendrocyte proliferation signaling, and here we present evidence that a similar mechanism operates in survival signaling. Integrin activation using manganese increases raft localization and rescues the effects of both raft depletion and PDGF removal on survival and PI3K signaling. Together, these results point to an essential role for rafts in oligodendrocyte survival signaling on the basis of the provision of a favorable environment for growth factor-mediated integrin activation.


Subject(s)
Integrins/metabolism , Membrane Microdomains/metabolism , Oligodendroglia/metabolism , Animals , Animals, Newborn , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fumonisins/pharmacology , Integrin alpha6beta1/drug effects , Integrin alpha6beta1/metabolism , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/metabolism , Integrins/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Manganese/pharmacology , Membrane Microdomains/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
11.
J Clin Invest ; 112(4): 544-53, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12925695

ABSTRACT

We studied the immunological basis for the very potent encephalitogenicity of myelin/oligodendrocyte glycoprotein (MOG), a minor component of myelin in the CNS that is widely used to induce experimental autoimmune encephalomyelitis (EAE). For this purpose, we generated a mutant mouse lacking a functional mog gene. This MOG-deficient mouse presents no clinical or histological abnormalities, permitting us to directly assess the role of MOG as a target autoantigen in EAE. In contrast to WT mice, which developed severe EAE following immunization with whole myelin, MOG-deficient mice had a mild phenotype, demonstrating that the anti-MOG response is a major pathogenic component of the autoimmune response directed against myelin. Moreover, while MOG transcripts are expressed in lymphoid organs in minute amounts, both MOG-deficient and WT mice show similar T and B cell responses against the extracellular domain of MOG, including the immunodominant MOG 35-55 T cell epitope. Furthermore, no differences in the fine specificity of the T cell responses to overlapping peptides covering the complete mouse MOG sequence were observed between MOG+/+ and MOG-/- mice. In addition, upon adoptive transfer, MOG-specific T cells from WT mice and those from MOG-deficient mice are equally pathogenic. This total lack of immune tolerance to MOG in WT C57BL/6 mice may be responsible for the high pathogenicity of the anti-MOG immune response as well as the high susceptibility of most animal strains to MOG-induced EAE.


Subject(s)
Immune Tolerance , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/physiology , Animals , B-Lymphocytes/immunology , Blotting, Northern , Blotting, Western , Brain/metabolism , Cell Division , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron , Models, Genetic , Myelin Proteins , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein , Peptides/chemistry , Phenotype , Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Tissue Distribution
13.
Curr Biol ; 13(2): 151-5, 2003 Jan 21.
Article in English | MEDLINE | ID: mdl-12546790

ABSTRACT

Individual growth factors can regulate multiple aspects of behavior within a single cell during differentiation, with each signaling pathway controlled independently and also responsive to other receptors such as cell surface integrins. The mechanisms by which this is achieved remain poorly understood. Here we use myelin-forming oligodendrocytes and their precursors to examine the role of lipid rafts, cholesterol and sphingolipid-rich microdomains of the cell membrane implicated in cell signaling. In these cells, the growth factor PDGF has sequential and independent roles in proliferation and survival. We show that the oligodendrocyte PDGFalpha receptor becomes sequestered in a raft compartment at the developmental stage when PDGF ceases to promote proliferation, but is now required for survival. We also show that laminin-2, which is expressed on axons in the CNS and which provides a target-dependent signal for oligodendrocyte survival by amplification of PDGFalphaR signaling, induces clustering of the laminin binding integrin alpha6beta1 with the PDGFalphaR-containing lipid raft domains. This extracellular matrix-induced colocalization of integrin and growth factor receptor generates a signaling environment within the raft for survival-promoting PI3K/Akt activity. These results demonstrate novel signaling roles for lipid rafts that ensure the separation and amplification of growth factor signaling pathways during development.


Subject(s)
Integrins/metabolism , Membrane Microdomains/metabolism , Oligodendroglia/metabolism , Platelet-Derived Growth Factor/metabolism , Protein Serine-Threonine Kinases , Animals , Cell Differentiation , In Vitro Techniques , Integrin alpha6beta1/metabolism , Laminin/metabolism , Oligodendroglia/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 99(20): 13211-6, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12235363

ABSTRACT

The destiny of the mitotically active cells of the subventricular zone (SVZ) in adult rodents is to migrate to the olfactory bulb, where they contribute to the replacement of granular and periglomerular neurons. However, these adult neural progenitors also can be mobilized in periventricular white matter and triggered to differentiate into astrocytes and oligodendrocytes in response to lysolecithin-induced demyelination. To mimic the environmental conditions of multiple sclerosis, we assessed the proliferation, migration, and differentiation potential of adult SVZ progenitor cells in response to experimental autoimmune encephalomyelitis (EAE) in mice. Inflammation and demyelination were observed in all mouse brains after EAE induction. EAE induced cell proliferation throughout the brain and especially within the lesions. Proliferating cells were neural progenitors, astrocytes, and oligodendrocyte precursors. EAE enhanced the migration of SVZ-derived neural progenitors to the olfactory bulb and triggered their mobilization in the periventricular white matter. The mobilized cells gave rise to neurons, astrocytes, and oligodendrocytes in the olfactory bulb but essentially to astrocytes and oligodendrocytes in the lesioned white matter. Our data indicate that the adult mouse SVZ is a source of newly generated oligodendrocytes and thus may contribute, along with oligodendrocyte precursors, to the replacement of oligodendrocytes in inflammatory demyelinating diseases of the central nervous system such as multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Neurons/cytology , Neurons/metabolism , Oligodendroglia/cytology , Prosencephalon/metabolism , Stem Cells/cytology , Animals , Bromodeoxyuridine/pharmacology , Cell Division , Cell Movement , Female , Immunohistochemistry , Inflammation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Stem Cells/metabolism , Time Factors
15.
J Neurosci Res ; 69(6): 763-71, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12205670

ABSTRACT

Precursor cells of the adult mouse subventricular zone (SVZ) are mobilized and recruited by a lysolecithin (LPC)-induced demyelination of the corpus callosum. Because age decreases the proliferation of the SVZ neural precursors as well as the potential for myelin repair of the adult central nervous system, we have compared the ability of young and aged adult neural precursors to respond to LPC-induced demyelination. With age, the SVZ cells lost their capacity to proliferate and to be recruited by the lesion. Whereas a single injection of fibroblast growth factor-2 or transforming growth factor-alpha stimulated the proliferation of SVZ and rostral migratory stream precursors in both groups of animals after demyelination, they favored recruitment at the lesion in young mice but not in aged ones. In vitro experiments using neurospheres derived from young and aged animals indicated that both populations have the same migratory performances. Our in vivo data thus suggest that aged neural precursors may loose their intrinsic capacities to respond to demyelination-induced signals. Alternatively, their function may be altered by modification of the aged extracellular environment.


Subject(s)
Corpus Callosum/cytology , Demyelinating Diseases/drug therapy , Fibroblast Growth Factor 2/pharmacology , Stem Cells/cytology , Transforming Growth Factor alpha/pharmacology , Age Factors , Animals , Cell Division/drug effects , Cell Movement/drug effects , Demyelinating Diseases/chemically induced , Lateral Ventricles/cytology , Lysophosphatidylcholines , Mice , Mice, Inbred Strains , Neurons/cytology , Stem Cells/drug effects
16.
Mol Cell Neurosci ; 19(2): 225-38, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11860275

ABSTRACT

Using the model of lysolecithin-induced demyelination of the corpus callosum in wild-type, NCAM-deficient, and endoneuraminidase-injected mice, we have analyzed the consequences of the loss of expression of NCAM or PSA residues on the migration and proliferation capacities of neural precursors of the subventricular zone (SVZ). We showed that the absence of PSA or NCAM delayed migration of neural precursors to the olfactory bulb and consequently enhanced their recruitment at the lesion site. Moreover, after demyelination, the lack of NCAM but not PSA promoted proliferation in the SVZ and the lesion while the lack of PSA favored the differentiation of the traced cells into the oligodendroglial fate both in the SVZ and in the lesion. As previously demonstrated in vitro (L. Decker et al., 2000, Mol. Cell. Neurosci. 16, 422-439), these data illustrate the involvement of PSA and NCAM in neural precursor motility and differentiation in the normal and injured central nervous system, suggesting distinct roles for these two molecules under pathophysiological conditions.


Subject(s)
Cell Differentiation/genetics , Cell Movement/genetics , Membrane Glycoproteins , Neural Cell Adhesion Molecules/deficiency , Neurons/metabolism , Sialic Acids/deficiency , Stem Cells/metabolism , Telencephalon/growth & development , Animals , Antigens, CD/metabolism , Astrocytes/cytology , Astrocytes/metabolism , CD24 Antigen , Cell Division/genetics , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glycoside Hydrolases/pharmacology , Immunohistochemistry , Lateral Ventricles/cytology , Lateral Ventricles/growth & development , Lateral Ventricles/metabolism , Lysophosphatidylcholines/pharmacology , Mice , Mice, Knockout , Nerve Regeneration/genetics , Neural Cell Adhesion Molecules/genetics , Sialic Acids/genetics , Stem Cells/cytology , Telencephalon/cytology , Telencephalon/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...