Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurochem ; 163(6): 478-499, 2022 12.
Article in English | MEDLINE | ID: mdl-36153691

ABSTRACT

The developmental process of central nervous system (CNS) myelin sheath formation is characterized by well-coordinated cellular activities ultimately ensuring rapid and synchronized neural communication. During this process, myelinating CNS cells, namely oligodendrocytes (OLGs), undergo distinct steps of differentiation, whereby the progression of earlier maturation stages of OLGs represents a critical step toward the timely establishment of myelinated axonal circuits. Given the complexity of functional integration, it is not surprising that OLG maturation is controlled by a yet fully to be defined set of both negative and positive modulators. In this context, we provide here first evidence for a role of lysophosphatidic acid (LPA) signaling via the G protein-coupled receptor LPA6 as a negative modulatory regulator of myelination-associated gene expression in OLGs. More specifically, the cell surface accessibility of LPA6 was found to be restricted to the earlier maturation stages of differentiating OLGs, and OLG maturation was found to occur precociously in Lpar6 knockout mice. To further substantiate these findings, a novel small molecule ligand with selectivity for preferentially LPA6 and LPA6 agonist characteristics was functionally characterized in vitro in primary cultures of rat OLGs and in vivo in the developing zebrafish. Utilizing this approach, a negative modulatory role of LPA6 signaling in OLG maturation could be corroborated. During development, such a functional role of LPA6 signaling likely serves to ensure timely coordination of circuit formation and myelination. Under pathological conditions as seen in the major human demyelinating disease multiple sclerosis (MS), however, persistent LPA6 expression and signaling in OLGs can be seen as an inhibitor of myelin repair. Thus, it is of interest that LPA6 protein levels appear elevated in MS brain samples, thereby suggesting that LPA6 signaling may represent a potential new druggable pathway suitable to promote myelin repair in MS.


Subject(s)
Oligodendroglia , Zebrafish , Mice , Animals , Rats , Humans , Oligodendroglia/metabolism , Myelin Sheath/metabolism , Neurogenesis/physiology , Cell Differentiation/physiology , Receptors, Lysophosphatidic Acid
2.
Front Cell Neurosci ; 16: 905299, 2022.
Article in English | MEDLINE | ID: mdl-35722615

ABSTRACT

The sodium-dependent glutamate transporter GLT-1 (EAAT2, SLC1A2) has been well-described as an important regulator of extracellular glutamate homeostasis in the central nervous system (CNS), a function that is performed mainly through its presence on astrocytes. There is, however, increasing evidence for the expression of GLT-1 in CNS cells other than astrocytes and in functional roles that are mediated by mechanisms downstream of glutamate uptake. In this context, GLT-1 expression has been reported for both neurons and oligodendrocytes (OLGs), and neuronal presynaptic presence of GLT-1 has been implicated in the regulation of glutamate uptake, gene expression, and mitochondrial function. Much less is currently known about the functional roles of GLT-1 expressed by OLGs. The data presented here provide first evidence that GLT-1 expressed by maturing OLGs contributes to the modulation of developmental myelination in the CNS. More specifically, using inducible and conditional knockout mice in which GLT-1 was deleted in maturing OLGs during a peak period of myelination (between 2 and 4 weeks of age) revealed hypomyelinated characteristics in the corpus callosum of preferentially male mice. These characteristics included reduced percentages of smaller diameter myelinated axons and reduced myelin thickness. Interestingly, this myelination phenotype was not found to be associated with major changes in myelin gene expression. Taken together, the data presented here demonstrate that GLT-1 expressed by maturing OLGs is involved in the modulation of the morphological aspects associated with CNS myelination in at least the corpus callosum and during a developmental window that appears of particular vulnerability in males compared to females.

3.
Glia ; 60(10): 1605-18, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22821873

ABSTRACT

During development, progenitors that are committed to differentiate into oligodendrocytes, the myelinating cells of the central nervous system (CNS), are generated within discrete regions of the neuroepithelium. More specifically, within the developing spinal cord and hindbrain ventrally located progenitor cells that are characterized by the expression of the transcription factor olig2 give temporally rise to first motor neurons and then oligodendrocyte progenitors. The regulation of this temporal neuron-glial switch has been found complex and little is known about the extrinsic factors regulating it. Our studies described here identified a zebrafish ortholog to mammalian atx, which displays evolutionarily conserved expression pattern characteristics. Most interestingly, atx was found to be expressed by cells of the cephalic floor plate during a time period when ventrally-derived oligodendrocyte progenitors arise in the developing hindbrain of the zebrafish. Knock-down of atx expression resulted in a delay and/or inhibition of the timely appearance of oligodendrocyte progenitors and subsequent developmental stages of the oligodendrocyte lineage. This effect of atx knock-down was not accompanied by changes in the number of olig2-positive progenitor cells, the overall morphology of the axonal network or the number of somatic abducens motor neurons. Thus, our studies identified Atx as an extrinsic factor that is likely secreted by cells from the floor plate and that is involved in regulating specifically the progression of olig2-positive progenitor cells into lineage committed oligodendrocyte progenitors.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Oligodendroglia/physiology , Phosphoric Diester Hydrolases/metabolism , Rhombencephalon/cytology , Rhombencephalon/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/drug effects , Body Patterning/genetics , Cell Differentiation/genetics , Cloning, Molecular , Embryo, Nonmammalian , Functional Laterality/genetics , Gene Expression Regulation, Developmental/drug effects , Morpholinos/pharmacology , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/drug effects , Phosphoric Diester Hydrolases/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Mol Cell Neurosci ; 37(2): 412-24, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18164210

ABSTRACT

Development of a complex process network by maturing oligodendrocytes is a critical but currently poorly characterized step toward myelination. Here, we demonstrate that the matricellular oligodendrocyte-derived protein phosphodiesterase-Ialpha/autotaxin (PD-Ialpha/ATX) and especially its MORFO domain are able to promote this developmental step. In particular, the single EF hand-like motif located within PD-Ialpha/ATX's MORFO domain was found to stimulate the outgrowth of higher order branches but not process elongation. This motif was also observed to be critical for the stimulatory effect of PD-Ialpha/ATX's MORFO domain on the reorganization of focal adhesions located at the leading edge of oligodendroglial protrusions. Collectively, our data suggest that PD-Ialpha/ATX promotes oligodendroglial process network formation and expansion via the cooperative action of multiple functional sites located within the MORFO domain and more specifically, a novel signaling pathway mediated by the single EF hand-like motif and regulating the correlated events of process outgrowth and focal adhesion organization.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/growth & development , Central Nervous System/metabolism , Focal Adhesions/metabolism , Oligodendroglia/metabolism , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/metabolism , Animals , Animals, Newborn , Cell Communication/physiology , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Cells, Cultured , Central Nervous System/cytology , Female , Oligodendroglia/cytology , Phosphoric Diester Hydrolases/chemistry , Protein Structure, Tertiary/physiology , Pyrophosphatases/chemistry , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
5.
Glia ; 53(5): 563-6, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16355369

ABSTRACT

During development, postmigratory, premyelinating oligodendrocytes extend processes that navigate through the central nervous system (CNS) environment, where they recognize a number of extracellular cues, including axonal segments to be myelinated. Ultimately this recognition event leads to the formation of the CNS myelin sheath. However, the morphological structures and molecular mechanisms that control such oligodendroglial pathfinding are poorly understood. Here we show that postmigratory, premyelinating oligodendrocyte processes possess at their distal tips expansions that ultrastructurally resemble growth cones of postmigratory neurons and that we will refer to as OLG-growth cones. OLG-growth cones are highly motile and capable of mediating process outgrowth, retraction, and branching. In addition, they express regulators of cytoskeletal organization, GAP43 and cofilin, that are known to mediate neuronal growth cone navigation. In a choice situation, processes of postmigratory, premyelinating oligodendrocytes and their OLG-growth cones have the ability to selectively avoid a nonpermissive substrate, that is, collagen IV. Thus, our findings provide, for the first time, a detailed characterization of sensorimotor structures present at the tips of postmigratory, premyelinating oligodendrocyte processes. Furthermore, the data presented here suggest that, although the cellular mechanisms involved in growth cone steering may be similar for postmigratory neuronal and oligodendroglial cells, extracellular cues may be interpreted in a cell-type-specific fashion.


Subject(s)
Growth Cones/physiology , Motor Neurons/physiology , Myelin Sheath/physiology , Neurons, Afferent/physiology , Oligodendroglia/physiology , Actin Depolymerizing Factors/metabolism , Animals , Cell Movement , Cells, Cultured , GAP-43 Protein/biosynthesis , Growth Cones/metabolism , Growth Cones/ultrastructure , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Neurons, Afferent/metabolism , Neurons, Afferent/ultrastructure , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Rats
6.
Mol Cell Neurosci ; 27(2): 140-50, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15485770

ABSTRACT

Myelination within the central nervous system (CNS) involves substantial morphogenesis of oligodendrocytes requiring plastic changes in oligodendrocyte-extracellular matrix (ECM) interactions, that is, adhesion. Our previous studies indicated that a regulator of such adhesive plasticity is oligodendrocyte-released phosphodiesterase-I alpha/autotaxin (PD-I alpha/ATX). We report here, that PD-I alpha/ATX's adhesion antagonism is mediated by a protein fragment different from the one that stimulates tumor cell motility. Furthermore, PD-I alpha/ATX's adhesion-antagonizing fragment causes a reorganized distribution of the focal adhesion components vinculin and paxillin and an integrin-dependent reduction in focal adhesion kinase (FAK) phosphorylation at tyrosine residue 925 (pFAK-925). In vivo, a similar reduction in pFAK-925 occurs at the onset of myelination when PD-I alpha/ATX expression is significantly upregulated. Most importantly, it can also be induced by the application of exogenous PD-I alpha/ATX. Our data, therefore, suggest that PD-I alpha/ATX participates in the regulation of myelination via a novel signaling pathway leading to changes in integrin-dependent focal adhesion assembly and consequently oligodendrocyte-ECM interactions.


Subject(s)
Cytoskeleton/metabolism , Glucose-6-Phosphate Isomerase/metabolism , Glycoproteins/metabolism , Multienzyme Complexes/metabolism , Nerve Fibers, Myelinated/metabolism , Phosphoric Diester Hydrolases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 1 , Cytoskeleton/drug effects , Female , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Glucose-6-Phosphate Isomerase/pharmacology , Glycoproteins/pharmacology , Multienzyme Complexes/pharmacology , Nerve Fibers, Myelinated/drug effects , Phosphodiesterase I , Phosphoric Diester Hydrolases/physiology , Phosphorylation/drug effects , Pregnancy , Pyrophosphatases , Rats , Rats, Sprague-Dawley
7.
J Neurosci Res ; 67(2): 174-84, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11782961

ABSTRACT

Cell cultures prepared from oligodendrocytes directly obtained from adult rat brain are composed of mature cells that lose their cell processes and myelin membrane during their isolation and therefore represent a very useful model to investigate the factors that could stimulate their recovery. We have observed that mature oligodendrocytes isolated from adult animals remain as round cells that lack processes for the first 3-4 days in culture. At the end of this lag period, however, the majority of the adult oligodendrocytes show a remarkable recovery, rapidly growing complex and extensive cell processes. Interestingly, the end of this lag period is accompanied by a dramatic upregulation in the expression of thyroid hormone (T(3)) receptor (TR). The functional importance of this increase in TR levels is supported by the observation that the majority of the cells cultured in the presence of T(3) show significantly more extensive and complex process outgrowth than the control cells in cultures lacking this hormone. In addition, this reactivation of the adult cells was also preceded by an increased expression of glucocorticoid receptor (GR) and cyclic AMP-response element binding protein (CREB), two transcription factors that together with TR appear to play important roles in the control of neonatal oligodendrocyte development. Thus, it is possible to hypothesize that upregulation of these proteins may be part of the metabolic changes that occur during the lag period required for recovery of the adult oligodendrocytes. These observations raise the question of whether these transcription factors may play any significant role during remyelination after demyelinating lesions of adult CNS.


Subject(s)
Cell Differentiation/physiology , Demyelinating Diseases/metabolism , Nerve Regeneration/physiology , Oligodendroglia/metabolism , Recovery of Function/physiology , Transcription Factors/metabolism , Triiodothyronine/pharmacology , Up-Regulation/physiology , Animals , Animals, Newborn , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/growth & development , Central Nervous System/metabolism , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Demyelinating Diseases/physiopathology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Immunohistochemistry , Myelin Basic Protein/drug effects , Myelin Basic Protein/metabolism , Nerve Regeneration/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Thyroid Hormone/drug effects , Receptors, Thyroid Hormone/metabolism , Recovery of Function/drug effects , Time Factors , Transcription Factors/drug effects , Triiodothyronine/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...