Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 81(1): 181, 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38615095

ABSTRACT

In vertebrates, oligodendrocytes (OLs) are glial cells of the central nervous system (CNS) responsible for the formation of the myelin sheath that surrounds the axons of neurons. The myelin sheath plays a crucial role in the transmission of neuronal information by promoting the rapid saltatory conduction of action potentials and providing neurons with structural and metabolic support. Saltatory conduction, first described in the peripheral nervous system (PNS), is now generally recognized as a universal evolutionary innovation to respond quickly to the environment: myelin helps us think and act fast. Nevertheless, the role of myelin in the central nervous system, especially in the brain, may not be primarily focused on accelerating conduction speed but rather on ensuring precision. Its principal function could be to coordinate various neuronal networks, promoting their synchronization through oscillations (or rhythms) relevant for specific information processing tasks. Interestingly, myelin has been directly involved in different types of cognitive processes relying on brain oscillations, and myelin plasticity is currently considered to be part of the fundamental mechanisms for memory formation and maintenance. However, despite ample evidence showing the involvement of myelin in cognition and neurodevelopmental disorders characterized by cognitive impairments, the link between myelin, brain oscillations, cognition and disease is not yet fully understood. In this review, we aim to highlight what is known and what remains to be explored to understand the role of myelin in high order brain processes.


Subject(s)
Myelin Sheath , Neurodevelopmental Disorders , Animals , Cognition , Central Nervous System , Brain
3.
Cell Mol Life Sci ; 81(1): 15, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38194116

ABSTRACT

Although intracellular Ca2+ signals of oligodendroglia, the myelin-forming cells of the central nervous system, regulate vital cellular processes including myelination, few studies on oligodendroglia Ca2+ signal dynamics have been carried out and existing software solutions are not adapted to the analysis of the complex Ca2+ signal characteristics of these cells. Here, we provide a comprehensive solution to analyze oligodendroglia Ca2+ imaging data at the population and single-cell levels. We describe a new analytical pipeline containing two free, open source and cross-platform software programs, Occam and post-prOccam, that enable the fully automated analysis of one- and two-photon Ca2+ imaging datasets from oligodendroglia obtained by either ex vivo or in vivo Ca2+ imaging techniques. Easily configurable, our software solution is optimized to obtain unbiased results from large datasets acquired with different imaging techniques. Compared to other recent software, our solution proved to be fast, low memory demanding and faithful in the analysis of oligodendroglial Ca2+ signals in all tested imaging conditions. Our versatile and accessible Ca2+ imaging data analysis tool will facilitate the elucidation of Ca2+-mediated mechanisms in oligodendroglia. Its configurability should also ensure its suitability with new use cases such as other glial cell types or even cells outside the CNS.


Subject(s)
Calcium , Oligodendroglia , Workflow , Myelin Sheath , Neuroglia
4.
Front Cell Neurosci ; 15: 645240, 2021.
Article in English | MEDLINE | ID: mdl-33708075

ABSTRACT

Enhanced neuronal activity in the healthy brain can induce de novo myelination and behavioral changes. As neuronal activity can be achieved using non-invasive measures, it may be of interest to utilize the innate ability of neuronal activity to instruct myelination as a novel strategy for myelin repair in demyelinating disorders such as multiple sclerosis (MS). Preclinical studies indicate that stimulation of neuronal activity in demyelinated lesions indeed has the potential to improve remyelination and that the stimulation paradigm is an important determinant of success. However, future studies will need to reveal the most efficient stimulation protocols as well as the biological mechanisms implicated. Nonetheless, clinical studies have already explored non-invasive brain stimulation as an attractive therapeutic approach that ameliorates MS symptomatology. However, whether symptom improvement is due to improved myelin repair remains unclear. In this mini-review, we discuss the neurobiological basis and potential of enhancing neuronal activity as a novel therapeutic approach in MS.

5.
Methods Mol Biol ; 2191: 135-149, 2021.
Article in English | MEDLINE | ID: mdl-32865743

ABSTRACT

In just over 10 years, the use of optogenetic technologies in neuroscience has become widespread, having today a tremendous impact on our understanding of brain function. An extensive number of studies have implemented a variety of tools allowing for the manipulation of neurons with light, including light-activated ion channels or G protein-coupled receptors, among other innovations. In this context, the proper calibration of photostimulation in vivo remains crucial to dissect brain circuitry or investigate the effect of neuronal activity on specific subpopulations of neurons and glia. Depending on the scientific question, the design of specific stimulation protocols must consider from the choice of the animal model to the light stimulation pattern to be delivered. In this chapter, we describe a detailed framework to investigate neuron-glia interactions in both mouse pups and adults using an optogenetic approach.


Subject(s)
Channelrhodopsins/genetics , Neuroglia/metabolism , Neurons/metabolism , Optogenetics/methods , Animals , Brain/metabolism , Brain/physiology , Humans , Mice , Neuroglia/pathology , Neurons/pathology , Patch-Clamp Techniques/methods , Rats
6.
Sci Adv ; 6(49)2020 12.
Article in English | MEDLINE | ID: mdl-33277253

ABSTRACT

Remyelination failure in multiple sclerosis (MS) is associated with a migration/differentiation block of oligodendroglia. The reason for this block is highly debated. It could result from disease-related extrinsic or intrinsic regulators in oligodendroglial biology. To avoid confounding immune-mediated extrinsic effect, we used an immune-deficient mouse model to compare induced pluripotent stem cell-derived oligodendroglia from MS and healthy donors following engraftment in the developing CNS. We show that the MS-progeny behaves and differentiates into oligodendrocytes to the same extent as controls. They generate equal amounts of myelin, with bona fide nodes of Ranvier, and promote equal restoration of their host slow conduction. MS-progeny expressed oligodendrocyte- and astrocyte-specific connexins and established functional connections with donor and host glia. Thus, MS oligodendroglia, regardless of major immune manipulators, are intrinsically capable of myelination and making functional axo-glia/glia-glia connections, reinforcing the view that the MS oligodendrocyte differentiation block is not from major intrinsic oligodendroglial deficits.

7.
Nat Commun ; 11(1): 5151, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33051462

ABSTRACT

Myelination of projection neurons by oligodendrocytes is key to optimize action potential conduction over long distances. However, a large fraction of myelin enwraps the axons of parvalbumin-positive fast-spiking interneurons (FSI), exclusively involved in local cortical circuits. Whether FSI myelination contributes to the fine-tuning of intracortical networks is unknown. Here we demonstrate that FSI myelination is required for the establishment and maintenance of the powerful FSI-mediated feedforward inhibition of cortical sensory circuits. The disruption of GABAergic synaptic signaling of oligodendrocyte precursor cells prior to myelination onset resulted in severe FSI myelination defects characterized by longer internodes and nodes, aberrant myelination of branch points and proximal axon malformation. Consequently, high-frequency FSI discharges as well as FSI-dependent postsynaptic latencies and strengths of excitatory neurons were reduced. These dysfunctions generated a strong excitation-inhibition imbalance that correlated with whisker-dependent texture discrimination impairments. FSI myelination is therefore critical for the function of mature cortical inhibitory circuits.


Subject(s)
Cerebellar Cortex/cytology , Interneurons/physiology , Myelin Sheath/metabolism , Neural Inhibition , Parvalbumins/metabolism , Animals , Axons/metabolism , Cerebellar Cortex/metabolism , Female , Male , Mice , Mice, Transgenic , Oligodendroglia/physiology , Parvalbumins/genetics
8.
Neurosci Lett ; 715: 134615, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31711979

ABSTRACT

In the cerebral cortex, GABAergic interneurons and oligodendrocyte lineage cells share different characteristics and interact despite being neurons and glial cells, respectively. These two distinct cell types share common embryonic origins and are born from precursors expressing similar transcription factors. Moreover, they highly interact with each other through different communication mechanisms during development. Notably, cortical oligodendrocyte precursor cells (OPCs) receive a major and transient GABAergic synaptic input, preferentially from parvalbumin-expressing interneurons, a specific interneuron subtype recently recognized as highly myelinated. In this review, we highlight the similarities and interactions between GABAergic interneurons and oligodendrocyte lineage cells in the cerebral cortex and suggest potential roles of this intimate interneuron-oligodendroglia relationship in cortical construction. We also propose new lines of research to understand the role of the close link between interneurons and oligodendroglia during cortical development and in pathological conditions such as schizophrenia.


Subject(s)
Cell Lineage , Cerebral Cortex/cytology , GABAergic Neurons/physiology , Interneurons/physiology , Oligodendrocyte Precursor Cells/physiology , Oligodendroglia/physiology , Animals , Cerebral Cortex/physiology , GABAergic Neurons/cytology , Humans , Interneurons/cytology , Oligodendrocyte Precursor Cells/cytology , Oligodendroglia/cytology
9.
Nat Commun ; 10(1): 4249, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31534164

ABSTRACT

The first wave of oligodendrocyte precursor cells (firstOPCs) and most GABAergic interneurons share common embryonic origins. Cortical firstOPCs are thought to be replaced by other OPC populations shortly after birth, maintaining a consistent OPC density and making postnatal interactions between firstOPCs and ontogenetically-related interneurons unlikely. Challenging these ideas, we show that a cortical firstOPC subpopulation survives and forms functional cell clusters with lineage-related interneurons. Favored by a common embryonic origin, these clusters display unexpected preferential synaptic connectivity and are anatomically maintained after firstOPCs differentiate into myelinating oligodendrocytes. While the concomitant rescue of interneurons and firstOPCs committed to die causes an exacerbated neuronal inhibition, it abolishes interneuron-firstOPC high synaptic connectivity. Further, the number of other oligodendroglia populations increases through a non-cell-autonomous mechanism, impacting myelination. These findings demonstrate unprecedented roles of interneuron and firstOPC apoptosis in regulating lineage-related cell interactions and the homeostatic oligodendroglia density.


Subject(s)
Apoptosis/physiology , Interneurons/metabolism , Neurogenesis/physiology , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Animals , Central Nervous System/cytology , Central Nervous System/embryology , Female , GABAergic Neurons/cytology , Homeodomain Proteins/metabolism , Interneurons/cytology , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Oligodendroglia/cytology
10.
JCI Insight ; 52019 03 21.
Article in English | MEDLINE | ID: mdl-30896448

ABSTRACT

In demyelinating diseases such as Multiple Sclerosis (MS), demyelination of neuronal fibers impairs impulse conduction and causes axon degeneration. While neuronal activity stimulates oligodendrocyte production and myelination in normal conditions, it remains unclear whether the activity of demyelinated axons restores their loss-of-function in a harmful environment. To investigate this question, we established a model to induce a moderate optogenetic stimulation of demyelinated axons in the corpus callosum at the level of the motor cortex in which cortical circuit activation and locomotor effects were reduced in adult freely moving mice. We demonstrate that a moderate activation of demyelinated axons enhances the differentiation of oligodendrocyte precursor cells onto mature oligodendrocytes, but only under a repeated stimulation paradigm. This activity-dependent increase in the oligodendrocyte pool promotes an extensive remyelination and functional restoration of conduction, as revealed by ultrastructural analyses and compound action potential recordings. Our findings reveal the need of preserving an appropriate neuronal activity in the damaged tissue to promote oligodendrocyte differentiation and remyelination, likely by enhancing axon-oligodendroglia interactions. Our results provide new perspectives for translational research using neuromodulation in demyelinating diseases.


Subject(s)
Demyelinating Diseases/metabolism , Myelin Sheath/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Animals , Axons/metabolism , Brain , Cell Differentiation , Corpus Callosum , Demyelinating Diseases/pathology , Disease Models, Animal , Female , Light , Male , Mice , Mice, Inbred C57BL , Neurons/radiation effects , Remyelination
11.
Elife ; 82019 12 31.
Article in English | MEDLINE | ID: mdl-31891351

ABSTRACT

Programmed cell death and early activity contribute to the emergence of functional cortical circuits. While most neuronal populations are scaled-down by death, some subpopulations are entirely eliminated, raising the question of the importance of such demise for cortical wiring. Here, we addressed this issue by focusing on Cajal-Retzius neurons (CRs), key players in cortical development that are eliminated in postnatal mice in part via Bax-dependent apoptosis. Using Bax-conditional mutants and CR hyperpolarization, we show that the survival of electrically active subsets of CRs triggers an increase in both dendrite complexity and spine density of upper layer pyramidal neurons, leading to an excitation/inhibition imbalance. The survival of these CRs is induced by hyperpolarization, highlighting an interplay between early activity and neuronal elimination. Taken together, our study reveals a novel activity-dependent programmed cell death process required for the removal of transient immature neurons and the proper wiring of functional cortical circuits.


Subject(s)
Apoptosis/genetics , Neurogenesis/genetics , Pyramidal Cells/metabolism , bcl-2-Associated X Protein/genetics , Animals , Animals, Newborn , Cell Polarity/genetics , Cerebral Cortex/metabolism , Electric Stimulation , Interstitial Cells of Cajal/metabolism , Mice , Mutant Proteins/genetics , Pyramidal Cells/pathology
12.
Front Cell Neurosci ; 12: 477, 2018.
Article in English | MEDLINE | ID: mdl-30574070

ABSTRACT

Optogenetic and pharmacogenetic techniques have been effective to analyze the role of neuronal activity in controlling oligodendroglia lineage cells in behaving juvenile and adult mice. This kind of studies is also of high interest during early postnatal (PN) development since important changes in oligodendroglia dynamics occur during the first two PN weeks. Yet, neuronal manipulation is difficult to implement at an early age because high-level, specific protein expression is less reliable in neonatal mice. Here, we describe a protocol allowing for an optogenetic stimulation of neurons in awake mouse pups with the purpose of investigating the effect of neuronal activity on oligodendroglia dynamics during early PN stages. Since GABAergic interneurons contact oligodendrocyte precursor cells (OPCs) through bona fide synapses and maintain a close relationship with these progenitors during cortical development, we used this relevant example of neuron-oligodendroglia interaction to implement a proof-of-principle optogenetic approach. First, we tested Nkx2.1-Cre and Parvalbumin (PV)-Cre lines to drive the expression of the photosensitive ion channel channelrhodopsin-2 (ChR2) in subpopulations of interneurons at different developmental stages. By using patch-clamp recordings and photostimulation of ChR2-positive interneurons in acute somatosensory cortical slices, we analyzed the level of functional expression of ChR2 in these neurons. We found that ChR2 expression was insufficient in PV-Cre mouse at PN day 10 (PN10) and that this channel needs to be expressed from embryonic stages (as in the Nkx2.1-Cre line) to allow for a reliable photoactivation in mouse pups. Then, we implemented a stereotaxic surgery to place a mini-optic fiber at the cortical surface in order to photostimulate ChR2-positive interneurons at PN10. In vivo field potentials were recorded in Layer V to verify that photostimulation reaches deep cortical layers. Finally, we analyzed the effect of the photostimulation on the layer V oligodendroglia population by conventional immunostainings. Neither the total density nor a proliferative fraction of OPCs were affected by increasing interneuron activity in vivo, complementing previous findings showing the lack of effect of GABAergic synaptic activity on OPC proliferation. The methodology described here should provide a framework for future investigation of the role of early cellular interactions during PN brain maturation.

13.
Elife ; 62017 09 06.
Article in English | MEDLINE | ID: mdl-28875931

ABSTRACT

In the adult brain, both neurons and oligodendrocytes can be generated from neural stem cells located within the Sub-Ventricular Zone (SVZ). Physiological signals regulating neuronal versus glial fate are largely unknown. Here we report that a thyroid hormone (T3)-free window, with or without a demyelinating insult, provides a favorable environment for SVZ-derived oligodendrocyte progenitor generation. After demyelination, oligodendrocytes derived from these newly-formed progenitors provide functional remyelination, restoring normal conduction. The cellular basis for neuronal versus glial determination in progenitors involves asymmetric partitioning of EGFR and TRα1, expression of which favor glio- and neuro-genesis, respectively. Moreover, EGFR+ oligodendrocyte progenitors, but not neuroblasts, express high levels of a T3-inactivating deiodinase, Dio3. Thus, TRα absence with high levels of Dio3 provides double-pronged blockage of T3 action during glial lineage commitment. These findings not only transform our understanding of how T3 orchestrates adult brain lineage decisions, but also provide potential insight into demyelinating disorders.


Subject(s)
Brain/cytology , Brain/drug effects , Cell Differentiation/drug effects , Hypothyroidism , Oligodendroglia/physiology , Remyelination , Adult , Animals , ErbB Receptors/metabolism , Humans , Iodide Peroxidase/metabolism , Mice , Thyroid Hormone Receptors alpha/metabolism
14.
Glia ; 65(11): 1821-1832, 2017 11.
Article in English | MEDLINE | ID: mdl-28795438

ABSTRACT

In the brain, neurons establish bona fide synapses onto oligodendrocyte precursor cells (OPCs), but the function of these neuron-glia synapses remains unresolved. A leading hypothesis suggests that these synapses regulate OPC proliferation and differentiation. However, a causal link between synaptic activity and OPC cellular dynamics is still missing. In the developing somatosensory cortex, OPCs receive a major type of synapse from GABAergic interneurons that is mediated by postsynaptic γ2-containing GABAA receptors. Here we genetically silenced these receptors in OPCs during the critical period of cortical oligodendrogenesis. We found that the inactivation of γ2-mediated synapses does not impact OPC proliferation and differentiation or the propensity of OPCs to myelinate their presynaptic interneurons. However, this inactivation causes a progressive and specific depletion of the OPC pool that lacks γ2-mediated synaptic activity without affecting the oligodendrocyte production. Our results show that, during cortical development, the γ2-mediated interneuron-to-OPC synapses do not play a role in oligodendrogenesis and suggest that these synapses finely tune OPC self-maintenance capacity. They also open the interesting possibility that a particular synaptic signaling onto OPCs plays a specific role in OPC function according to the neurotransmitter released, the identity of presynaptic neurons or the postsynaptic receptors involved.


Subject(s)
Cerebral Cortex/cytology , Neurogenesis/physiology , Neurons/physiology , Oligodendrocyte Precursor Cells/physiology , Oligodendroglia/physiology , Synapses/physiology , Animals , Animals, Newborn , Antigens/genetics , Antigens/metabolism , Autophagy-Related Proteins , Calcium/metabolism , Cell Count , Cell Differentiation/physiology , Cerebral Cortex/growth & development , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Myelin Basic Protein/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Statistics, Nonparametric , Synapses/genetics
15.
Cell Rep ; 17(12): 3133-3141, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28009284

ABSTRACT

Cajal-Retzius cells (CRs), the first-born neurons in the developing cerebral cortex, coordinate crucial steps in the construction of functional circuits. CRs are thought to be transient, as they disappear during early postnatal life in both mice and humans, where their abnormal persistence is associated with pathological conditions. Embryonic CRs comprise at least three molecularly and functionally distinct subtypes: septum, ventral pallium/pallial-subpallial boundary (PSB), and hem. However, whether subtype-specific features exist postnatally and through which mechanisms they disappear remain unknown. We report that CR subtypes display unique distributions and dynamics of death in the postnatal mouse cortex. Surprisingly, although all CR subtypes undergo cell death, septum, but not hem, CRs die in a Bax-dependent manner. Bax-inactivated rescued septum-CRs maintain immature electrophysiological properties. These results underlie the existence of an exquisitely refined control of developmental cell death and provide a model to test the effect of maintaining immature circuits in the adult neocortex.


Subject(s)
Cell Death/genetics , Cerebral Cortex/metabolism , Neurons/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Lineage/genetics , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Embryo, Mammalian , Humans , Mice
16.
Nat Commun ; 6: 7844, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26238238

ABSTRACT

The myelin sheath on vertebrate axons is critical for neural impulse transmission, but whether electrically active axons are preferentially myelinated by glial cells, and if so, whether axo-glial synapses are involved, are long-standing questions of significance to nervous system development, plasticity and disease. Here we show using an in vitro system that oligodendrocytes preferentially myelinate electrically active axons, but synapses from axons onto myelin-forming oligodendroglial cells are not required. Instead, vesicular release at nonsynaptic axo-glial junctions induces myelination. Axons releasing neurotransmitter from vesicles that accumulate in axon varicosities induces a local rise in cytoplasmic calcium in glial cell processes at these nonsynaptic functional junctions, and this signalling stimulates local translation of myelin basic protein to initiate myelination.


Subject(s)
Axons/metabolism , Exocytosis , Myelin Sheath/metabolism , Neurotransmitter Agents/metabolism , Oligodendroglia/metabolism , Action Potentials , Animals , Calcium , Calcium Signaling , Gene Expression Regulation , In Vitro Techniques , Intercellular Junctions , Mice , Myelin Basic Protein/genetics , Neuroglia/metabolism , Neurons , Signal Transduction
17.
Front Cell Neurosci ; 9: 77, 2015.
Article in English | MEDLINE | ID: mdl-25852473

ABSTRACT

Oligodendrocyte precursor cells (OPCs) are a major source of remyelinating oligodendrocytes in demyelinating diseases such as Multiple Sclerosis (MS). While OPCs are innervated by unmyelinated axons in the normal brain, the fate of such synaptic contacts after demyelination is still unclear. By combining electrophysiology and immunostainings in different transgenic mice expressing fluorescent reporters, we studied the synaptic innervation of OPCs in the model of lysolecithin (LPC)-induced demyelination of corpus callosum. Synaptic innervation of reactivated OPCs in the lesion was revealed by the presence of AMPA receptor-mediated synaptic currents, VGluT1+ axon-OPC contacts in 3D confocal reconstructions and synaptic junctions observed by electron microscopy. Moreover, 3D confocal reconstructions of VGluT1 and NG2 immunolabeling showed the existence of glutamatergic axon-OPC contacts in post-mortem MS lesions. Interestingly, patch-clamp recordings in LPC-induced lesions demonstrated a drastic decrease in spontaneous synaptic activity of OPCs early after demyelination that was not caused by an impaired conduction of compound action potentials. A reduction in synaptic connectivity was confirmed by the lack of VGluT1+ axon-OPC contacts in virtually all rapidly proliferating OPCs stained with EdU (50-ethynyl-20-deoxyuridine). At the end of the massive proliferation phase in lesions, the proportion of innervated OPCs rapidly recovers, although the frequency of spontaneous synaptic currents did not reach control levels. In conclusion, our results demonstrate that newly-generated OPCs do not receive synaptic inputs during their active proliferation after demyelination, but gain synapses during the remyelination process. Hence, glutamatergic synaptic inputs may contribute to inhibit OPC proliferation and might have a physiopathological relevance in demyelinating disorders.

18.
Elife ; 42015 Apr 22.
Article in English | MEDLINE | ID: mdl-25902404

ABSTRACT

NG2 cells, oligodendrocyte progenitors, receive a major synaptic input from interneurons in the developing neocortex. It is presumed that these precursors integrate cortical networks where they act as sensors of neuronal activity. We show that NG2 cells of the developing somatosensory cortex form a transient and structured synaptic network with interneurons that follows its own rules of connectivity. Fast-spiking interneurons, highly connected to NG2 cells, target proximal subcellular domains containing GABAA receptors with γ2 subunits. Conversely, non-fast-spiking interneurons, poorly connected with these progenitors, target distal sites lacking this subunit. In the network, interneuron-NG2 cell connectivity maps exhibit a local spatial arrangement reflecting innervation only by the nearest interneurons. This microcircuit architecture shows a connectivity peak at PN10, coinciding with a switch to massive oligodendrocyte differentiation. Hence, GABAergic innervation of NG2 cells is temporally and spatially regulated from the subcellular to the network level in coordination with the onset of oligodendrogenesis.


Subject(s)
Interneurons/ultrastructure , Neocortex/cytology , Neural Stem Cells/ultrastructure , Oligodendroglia/ultrastructure , Somatosensory Cortex/cytology , Action Potentials/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation , Gene Expression , Genes, Reporter , Interneurons/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Microtomy , Neocortex/growth & development , Neocortex/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Oligodendroglia/metabolism , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Somatosensory Cortex/growth & development , Somatosensory Cortex/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
19.
Cereb Cortex ; 25(4): 1114-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24217990

ABSTRACT

NG2 cells, a main pool of glial progenitors, express γ-aminobutyric acid A (GABA(A)) receptors (GABA(A)Rs), the functional and molecular properties of which are largely unknown. We recently reported that transmission between GABAergic interneurons and NG2 cells drastically changes during development of the somatosensory cortex, switching from synaptic to extrasynaptic communication. Since synaptic and extrasynaptic GABA(A)Rs of neurons differ in their subunit composition, we hypothesize that GABA(A)Rs of NG2 cells undergo molecular changes during cortical development accompanying the switch of transmission modes. Single-cell RT-PCR and the effects of zolpidem and α5IA on evoked GABAergic currents reveal the predominance of functional α1- and α5-containing GABA(A)Rs at interneuron-NG2 cell synapses in the second postnatal week, while the α5 expression declines later in development when responses are exclusively extrasynaptic. Importantly, pharmacological and molecular analyses demonstrate that γ2, a subunit contributing to the clustering of GABA(A)Rs at postsynaptic sites in neurons, is down-regulated in NG2 cells in a cell type-specific manner in concomitance with the decline of synaptic activity and the switch of transmission mode. In keeping with the synaptic nature of γ2 in neurons, the down-regulation of this subunit is an important molecular hallmark of the change of transmission modes between interneurons and NG2 cells during development.


Subject(s)
Neocortex/growth & development , Neural Stem Cells/physiology , Oligodendroglia/physiology , Receptors, GABA-A/metabolism , Synapses/physiology , Animals , Cytoplasm/drug effects , Cytoplasm/metabolism , Down-Regulation , Electric Stimulation , GABA-A Receptor Agonists/pharmacology , Interneurons/drug effects , Interneurons/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Transgenic , Neocortex/drug effects , Neocortex/physiology , Neural Stem Cells/drug effects , Oligodendroglia/drug effects , Patch-Clamp Techniques , Polymerase Chain Reaction , Pyridines/pharmacology , RNA, Messenger/metabolism , Single-Cell Analysis , Somatosensory Cortex/drug effects , Somatosensory Cortex/growth & development , Somatosensory Cortex/physiology , Synapses/drug effects , Zolpidem , gamma-Aminobutyric Acid/metabolism
20.
Neuroscientist ; 21(3): 266-76, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24722526

ABSTRACT

The surprising discovery of bona fide synapses between neurons and oligodendrocytes precursor cells (OPCs) 15 years ago placed these progenitors as real partners of neurons in the CNS. The role of these synapses has not been established yet, but a main hypothesis is that neuron-OPC synaptic activity is a signaling pathway controlling OPC proliferation/differentiation, influencing the myelination process. However, new evidences describing non-synaptic mechanisms of communication between neurons and OPCs have revealed that neuron-OPC interactions are more complex than expected. The activation of extrasynaptic receptors by ambient neurotransmitter or local spillover and the ability of OPCs to sense neuronal activity through a potassium channel suggest that distinct modes of communication mediate different functions of OPCs in the CNS. This review discusses different mechanisms used by OPCs to interact with neurons and their potential roles during postnatal development and in brain disorders.


Subject(s)
Brain/physiology , Cell Communication , Neurons/physiology , Oligodendroglia/physiology , Stem Cells/physiology , Animals , Brain/growth & development , Calcium Signaling , GABAergic Neurons/physiology , Glutamic Acid/metabolism , Humans , Synapses/physiology , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...