Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
2.
Cell Rep ; 39(2): 110667, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35417707

ABSTRACT

Cortical wiring relies on guidepost cells and activity-dependent processes that are thought to act sequentially. Here, we show that the construction of layer 1 (L1), a main site of top-down integration, is regulated by crosstalk between transient Cajal-Retzius cells (CRc) and spontaneous activity of the thalamus, a main driver of bottom-up information. While activity was known to regulate CRc migration and elimination, we found that prenatal spontaneous thalamic activity and NMDA receptors selectively control CRc early density, without affecting their demise. CRc density, in turn, regulates the distribution of upper layer interneurons and excitatory synapses, thereby drastically impairing the apical dendrite activity of output pyramidal neurons. In contrast, postnatal sensory-evoked activity had a limited impact on L1 and selectively perturbed basal dendrites synaptogenesis. Collectively, our study highlights a remarkable interplay between thalamic activity and CRc in L1 functional wiring, with major implications for our understanding of cortical development.


Subject(s)
Interneurons , Pyramidal Cells , Dendrites/physiology , Interneurons/physiology , Neurons/physiology , Thalamus
3.
Cell Rep ; 37(12): 110133, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34936875

ABSTRACT

Intracellular calcium signaling underlies the astroglial control of synaptic transmission and plasticity. Mitochondria-endoplasmic reticulum contacts (MERCs) are key determinants of calcium dynamics, but their functional impact on astroglial regulation of brain information processing is unexplored. We found that the activation of astrocyte mitochondrial-associated type-1 cannabinoid (mtCB1) receptors determines MERC-dependent intracellular calcium signaling and synaptic integration. The stimulation of mtCB1 receptors promotes calcium transfer from the endoplasmic reticulum to mitochondria through a specific molecular cascade, involving the mitochondrial calcium uniporter (MCU). Physiologically, mtCB1-dependent mitochondrial calcium uptake determines the dynamics of cytosolic calcium events in astrocytes upon endocannabinoid mobilization. Accordingly, electrophysiological recordings in hippocampal slices showed that conditional genetic exclusion of mtCB1 receptors or dominant-negative MCU expression in astrocytes blocks lateral synaptic potentiation, through which astrocytes integrate the activity of distant synapses. Altogether, these data reveal an endocannabinoid link between astroglial MERCs and the regulation of brain network functions.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Cannabinoids/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Receptors, Cannabinoid/physiology , Synapses/physiology , Animals , Astrocytes/cytology , Calcium Channels/physiology , Calcium Signaling , Cells, Cultured , Hippocampus/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Synaptic Transmission
4.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33619110

ABSTRACT

The organization of sensory maps in the cerebral cortex depends on experience, which drives homeostatic and long-term synaptic plasticity of cortico-cortical circuits. In the mouse primary somatosensory cortex (S1) afferents from the higher-order, posterior medial thalamic nucleus (POm) gate synaptic plasticity in layer (L) 2/3 pyramidal neurons via disinhibition and the production of dendritic plateau potentials. Here we address whether these thalamocortically mediated responses play a role in whisker map plasticity in S1. We find that trimming all but two whiskers causes a partial fusion of the representations of the two spared whiskers, concomitantly with an increase in the occurrence of POm-driven N-methyl-D-aspartate receptor-dependent plateau potentials. Blocking the plateau potentials restores the archetypical organization of the sensory map. Our results reveal a mechanism for experience-dependent cortical map plasticity in which higher-order thalamocortically mediated plateau potentials facilitate the fusion of normally segregated cortical representations.


Subject(s)
Action Potentials/physiology , Evoked Potentials, Somatosensory/physiology , Nerve Net/physiology , Somatosensory Cortex/physiology , Thalamus/physiology , Vibrissae/physiology , Action Potentials/drug effects , Animals , Brain Mapping/methods , Dizocilpine Maleate/pharmacology , Evoked Potentials, Somatosensory/drug effects , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Gene Expression , Male , Mice , Mice, Inbred C57BL , Nerve Net/anatomy & histology , Neuronal Plasticity/drug effects , Optical Imaging , Patch-Clamp Techniques , Picrotoxin/pharmacology , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Somatosensory Cortex/anatomy & histology , Thalamus/anatomy & histology , Vibrissae/injuries
5.
Elife ; 92020 11 30.
Article in English | MEDLINE | ID: mdl-33252331

ABSTRACT

Survival depends on the ability of animals to select the appropriate behavior in response to threat and safety sensory cues. However, the synaptic and circuit mechanisms by which the brain learns to encode accurate predictors of threat and safety remain largely unexplored. Here, we show that frontal association cortex (FrA) pyramidal neurons of mice integrate auditory cues and basolateral amygdala (BLA) inputs non-linearly in a NMDAR-dependent manner. We found that the response of FrA pyramidal neurons was more pronounced to Gaussian noise than to pure frequency tones, and that the activation of BLA-to-FrA axons was the strongest in between conditioning pairings. Blocking BLA-to-FrA signaling specifically at the time of presentation of Gaussian noise (but not 8 kHz tone) between conditioning trials impaired the formation of auditory fear memories. Taken together, our data reveal a circuit mechanism that facilitates the formation of fear traces in the FrA, thus providing a new framework for probing discriminative learning and related disorders.


Subject(s)
Acoustic Stimulation/adverse effects , Amygdala/physiology , Fear/physiology , Frontal Lobe/physiology , Learning/physiology , Animals , Calcium/metabolism , Conditioning, Classical/physiology , Male , Mice , Microscopy, Confocal , Neuronal Plasticity/physiology , Optogenetics , Patch-Clamp Techniques
6.
Cell Rep ; 32(9): 108097, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32877679

ABSTRACT

Cortical plasticity improves behaviors and helps recover lost functions after injury. However, the underlying synaptic mechanisms remain unclear. In mice, we show that trimming all but one whisker enhances sensory responses from the spared whisker in the barrel cortex and occludes whisker-mediated synaptic potentiation (w-Pot) in vivo. In addition, whisker-dependent behaviors that are initially impaired by single-whisker experience (SWE) rapidly recover when associated cortical regions remap. Cross-linking the surface GluA2 subunit of AMPA receptors (AMPARs) suppresses the expression of w-Pot, presumably by blocking AMPAR surface diffusion, in mice with all whiskers intact, indicating that synaptic potentiation in vivo requires AMPAR trafficking. We use this approach to demonstrate that w-Pot is required for SWE-mediated strengthening of synaptic inputs and initiates the recovery of previously learned skills during the early phases of SWE. Taken together, our data reveal that w-Pot mediates cortical remapping and behavioral improvement upon partial sensory deafferentation.


Subject(s)
Neuronal Plasticity/genetics , Receptors, AMPA/metabolism , Animals , Humans , Mice , Sensory Deprivation/physiology
7.
Neuron ; 105(5): 764-766, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32135087

ABSTRACT

In this issue of Neuron, Roth et al. (2020) report that the content of GluA1-containing AMPAR at spines and dendrites in vivo in the motor and visual cortex increases proportionally to the learning of a motor task. Visual cortex activity is necessary for increasing AMPAR content and learning in light.


Subject(s)
Neuronal Plasticity , Receptors, AMPA , Dendrites , Neurons , Protein Transport
8.
Front Mol Neurosci ; 12: 238, 2019.
Article in English | MEDLINE | ID: mdl-31611774

ABSTRACT

Intelligence is the ability to learn appropriate responses to stimuli and the capacity to master new skills. Synaptic integration at the dendritic level is thought to be essential for this ability through linear and non-linear processing, by allowing neurons to be tuned to relevant information and to maximize adaptive behavior. Showing that dendrites are able to generate local computations that influence how animals perceive the world, form a new memory or learn a new skill was a break-through in neuroscience, since in the past they were seen as passive elements of the neurons, just funneling information to the soma. Here, we provide an overview of the role of dendritic integration in improving the neuronal network and behavioral performance. We focus on how NMDA spikes are generated and their role in neuronal computation for optimal behavioral output based on recent in vivo studies on rodents.

9.
Brain Struct Funct ; 220(6): 3673-82, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25158900

ABSTRACT

The process of learning mainly depends on the ability to store new information, while the ability to retrieve this information and express appropriate behaviors are also crucial for the adaptation of individuals to environmental cues. Thereby, all three components contribute to the cognitive fitness of an individual. While a lack of behavioral adaptation is a recurrent trait of intellectually disabled patients, discriminating between memory formation, memory retrieval or behavioral expression deficits is not easy to establish. Here, we report some deficits in contextual fear behavior in knockout mice for the intellectual disability gene Il1rapl1. Functional in vivo experiments revealed that the lack of conditioned response resulted from a local inhibitory to excitatory (I/E) imbalance in basolateral amygdala (BLA) consecutive to a loss of excitatory drive onto BLA principal cells by caudal hippocampus axonal projections. A normalization of the fear behavior was obtained in adult mutant mice following opsin-based in vivo synaptic priming of hippocampo-BLA synapses in adult il1rapl1 knockout mice, indicating that synaptic efficacy at hippocampo-BLA projections is crucial for contextual fear memory expression. Importantly, because this restoration was obtained after the learning phase, our results suggest that some of the genetically encoded cognitive deficits in humans may originate from a lack of restitution of genuinely formed memories rather than an exclusive inability to store new memories.


Subject(s)
Amygdala/physiology , Fear/physiology , Hippocampus/physiology , Intellectual Disability/physiopathology , Interleukin-1 Receptor Accessory Protein/physiology , Animals , Conditioning, Classical/physiology , Disease Models, Animal , Intellectual Disability/genetics , Interleukin-1 Receptor Accessory Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Synapses/physiology , Synaptic Potentials
10.
Nature ; 515(7525): 116-9, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25174710

ABSTRACT

Long-term synaptic potentiation (LTP) is thought to be a key process in cortical synaptic network plasticity and memory formation. Hebbian forms of LTP depend on strong postsynaptic depolarization, which in many models is generated by action potentials that propagate back from the soma into dendrites. However, local dendritic depolarization has been shown to mediate these forms of LTP as well. As pyramidal cells in supragranular layers of the somatosensory cortex spike infrequently, it is unclear which of the two mechanisms prevails for those cells in vivo. Using whole-cell recordings in the mouse somatosensory cortex in vivo, we demonstrate that rhythmic sensory whisker stimulation efficiently induces synaptic LTP in layer 2/3 (L2/3) pyramidal cells in the absence of somatic spikes. The induction of LTP depended on the occurrence of NMDAR (N-methyl-d-aspartate receptor)-mediated long-lasting depolarizations, which bear similarities to dendritic plateau potentials. In addition, we show that whisker stimuli recruit synaptic networks that originate from the posteromedial complex of the thalamus (POm). Photostimulation of channelrhodopsin-2 expressing POm neurons generated NMDAR-mediated plateau potentials, whereas the inhibition of POm activity during rhythmic whisker stimulation suppressed the generation of those potentials and prevented whisker-evoked LTP. Taken together, our data provide evidence for sensory-driven synaptic LTP in vivo, in the absence of somatic spiking. Instead, LTP is mediated by plateau potentials that are generated through the cooperative activity of lemniscal and paralemniscal synaptic circuitry.


Subject(s)
Dendrites/physiology , Long-Term Potentiation , Somatosensory Cortex/cytology , Somatosensory Cortex/physiology , Action Potentials , Animals , Channelrhodopsins , Male , Mice , Mice, Inbred C57BL , Physical Stimulation , Receptors, N-Methyl-D-Aspartate/metabolism , Thalamus/cytology , Thalamus/physiology , Vibrissae/physiology
11.
Nature ; 511(7510): 471-4, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24828045

ABSTRACT

During development, thalamocortical (TC) input has a critical role in the spatial delineation and patterning of cortical areas, yet the underlying cellular and molecular mechanisms that drive cortical neuron differentiation are poorly understood. In the primary (S1) and secondary (S2) somatosensory cortex, layer 4 (L4) neurons receive mutually exclusive input originating from two thalamic nuclei: the ventrobasalis (VB), which conveys tactile input, and the posterior nucleus (Po), which conveys modulatory and nociceptive input. Recently, we have shown that L4 neuron identity is not fully committed postnatally, implying a capacity for TC input to influence differentiation during cortical circuit assembly. Here we investigate whether the cell-type-specific molecular and functional identity of L4 neurons is instructed by the origin of their TC input. Genetic ablation of the VB at birth resulted in an anatomical and functional rewiring of Po projections onto L4 neurons in S1. This induced acquisition of Po input led to a respecification of postsynaptic L4 neurons, which developed functional molecular features of Po-target neurons while repressing VB-target traits. Respecified L4 neurons were able to respond both to touch and to noxious stimuli, in sharp contrast to the normal segregation of these sensory modalities in distinct cortical circuits. These findings reveal a behaviourally relevant TC-input-type-specific control over the molecular and functional differentiation of postsynaptic L4 neurons and cognate intracortical circuits, which instructs the development of modality-specific neuronal and circuit properties during corticogenesis.


Subject(s)
Cell Differentiation , Neural Pathways/physiology , Neurons/cytology , Neurons/physiology , Post-Synaptic Density/physiology , Somatosensory Cortex/physiology , Thalamic Nuclei/physiology , Animals , Axons/drug effects , Axons/physiology , Capsaicin/pharmacology , Cell Differentiation/drug effects , Female , Male , Mice, Inbred C57BL , Neural Pathways/drug effects , Neurons/drug effects , Noxae/pharmacology , Optogenetics , Post-Synaptic Density/drug effects , Somatosensory Cortex/cytology , Somatosensory Cortex/drug effects , Synaptic Potentials/drug effects , Thalamic Nuclei/cytology , Thalamic Nuclei/drug effects , Touch/physiology , Vibrissae/drug effects , Vibrissae/physiology
12.
PLoS Biol ; 12(3): e1001820, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24667537

ABSTRACT

Cognitive and behavioral disorders are thought to be a result of neuronal dysfunction, but the underlying molecular defects remain largely unknown. An important signaling pathway involved in the regulation of neuronal function is the cyclic AMP/Protein kinase A pathway. We here show an essential role for coronin 1, which is encoded in a genomic region associated with neurobehavioral dysfunction, in the modulation of cyclic AMP/PKA signaling. We found that coronin 1 is specifically expressed in excitatory but not inhibitory neurons and that coronin 1 deficiency results in loss of excitatory synapses and severe neurobehavioral disabilities, including reduced anxiety, social deficits, increased aggression, and learning defects. Electrophysiological analysis of excitatory synaptic transmission in amygdala revealed that coronin 1 was essential for cyclic-AMP-protein kinase A-dependent presynaptic plasticity. We further show that upon cell surface stimulation, coronin 1 interacted with the G protein subtype Gαs to stimulate the cAMP/PKA pathway. The absence of coronin 1 or expression of coronin 1 mutants unable to interact with Gαs resulted in a marked reduction in cAMP signaling. Strikingly, synaptic plasticity and behavioral defects of coronin 1-deficient mice were restored by in vivo infusion of a membrane-permeable cAMP analogue. Together these results identify coronin 1 as being important for cognition and behavior through its activity in promoting cAMP/PKA-dependent synaptic plasticity and may open novel avenues for the dissection of signal transduction pathways involved in neurobehavioral processes.


Subject(s)
Behavior, Animal , Cognition/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Microfilament Proteins/physiology , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/genetics , Animals , Brain/metabolism , Brain/pathology , Humans , Memory , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Signal Transduction , Social Behavior
13.
Philos Trans R Soc Lond B Biol Sci ; 369(1633): 20130160, 2014 Jan 05.
Article in English | MEDLINE | ID: mdl-24298161

ABSTRACT

Loss-of-function mutations in the gene encoding for the RhoGAP protein of oligophrenin-1 (OPHN1) lead to cognitive disabilities (CDs) in humans, yet the underlying mechanisms are not known. Here, we show that in mice constitutive lack of Ophn1 is associated with dysregulation of the cyclic adenosine monophosphate/phosphate kinase A (cAMP/PKA) signalling pathway in a brain-area-specific manner. Consistent with a key role of cAMP/PKA signalling in regulating presynaptic function and plasticity, we found that PKA-dependent presynaptic plasticity was completely abolished in affected brain regions, including hippocampus and amygdala. At the behavioural level, lack of OPHN1 resulted in hippocampus- and amygdala-related learning disabilities which could be fully rescued by the ROCK/PKA kinase inhibitor fasudil. Together, our data identify OPHN1 as a key regulator of presynaptic function and suggest that, in addition to reported postsynaptic deficits, loss of presynaptic plasticity contributes to the pathophysiology of CDs.


Subject(s)
Cytoskeletal Proteins/deficiency , GTPase-Activating Proteins/deficiency , Learning Disabilities/genetics , Neuronal Plasticity/physiology , Nuclear Proteins/deficiency , Presynaptic Terminals/physiology , Signal Transduction/physiology , Animals , Blotting, Western , Conditioning, Psychological , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeletal Proteins/genetics , Electric Stimulation , GTPase-Activating Proteins/genetics , Learning Disabilities/physiopathology , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics
14.
J Neurosci ; 33(34): 13805-19, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23966701

ABSTRACT

Intellectual disorders (IDs) have been regularly associated with morphological and functional deficits at glutamatergic synapses in both humans and rodents. How these synaptic deficits may lead to the variety of learning and memory deficits defining ID is still unknown. Here we studied the functional and behavioral consequences of the ID gene il1rapl1 deficiency in mice and reported that il1rapl1 constitutive deletion alters cued fear memory formation. Combined in vivo and in vitro approaches allowed us to unveil a causal relationship between a marked inhibitory/excitatory (I/E) imbalance in dedicated amygdala neuronal subcircuits and behavioral deficits. Cell-targeted recordings further demonstrated a morpho-functional impact of the mutation at thalamic projections contacting principal cells, whereas the same afferents on interneurons are unaffected by the lack of Il1rapl1. We thus propose that excitatory synapses have a heterogeneous vulnerability to il1rapl1 gene constitutive mutation and that alteration of a subset of excitatory synapses in neuronal circuits is sufficient to generate permanent cognitive deficits.


Subject(s)
Excitatory Postsynaptic Potentials/genetics , Intellectual Disability/complications , Memory Disorders/etiology , Amygdala/cytology , Anesthetics, Local/pharmacology , Animals , Association Learning/physiology , Cerebral Cortex/cytology , Channelrhodopsins , Conditioning, Psychological/physiology , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Disease Models, Animal , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Fear/physiology , GABA Antagonists/pharmacology , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , In Vitro Techniques , Intellectual Disability/genetics , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neurons/physiology , Neurons/ultrastructure
15.
Neuron ; 75(3): 490-502, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22884332

ABSTRACT

Functional maps in the cerebral cortex reorganize in response to changes in experience, but the synaptic underpinnings remain uncertain. Here, we demonstrate that layer (L) 2/3 pyramidal cell synapses in mouse barrel cortex can be potentiated upon pairing of whisker-evoked postsynaptic potentials (PSPs) with action potentials (APs). This spike-timing-dependent long-term potentiation (STD-LTP) was only effective for PSPs evoked by deflections of a whisker in the neuron's receptive field center, and not its surround. Trimming of all except two whiskers rapidly opened the possibility to drive STD-LTP by the spared surround whisker. This facilitated STD-LTP was associated with a strong decrease in the surrounding whisker-evoked inhibitory conductance and partially occluded picrotoxin-mediated LTP facilitation. Taken together, our data demonstrate that sensory deprivation-mediated disinhibition facilitates STD-LTP from the sensory surround, which may promote correlation- and experience-dependent expansion of receptive fields.


Subject(s)
Long-Term Potentiation/physiology , Somatosensory Cortex/physiology , Action Potentials/physiology , Animals , Mice , Neurons/physiology , Physical Stimulation , Sensory Deprivation/physiology , Vibrissae/innervation
16.
Neuron ; 74(2): 214-7, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22542175

ABSTRACT

In this issue of Neuron, Chen et al. (2012) and van Versendaal et al. (2012) used fluorescently tagged gephyrin to track inhibitory synapses in the mouse visual cortex in vivo. Their studies show that visual experience-dependent plasticity is associated with clustered and location-specific pruning of inhibitory synapses.

17.
PLoS One ; 7(1): e29763, 2012.
Article in English | MEDLINE | ID: mdl-22291894

ABSTRACT

The GDI1 gene encodes αGDI, which retrieves inactive GDP-bound RAB from membranes to form a cytosolic pool awaiting vesicular release. Mutations in GDI1 are responsible for X-linked Intellectual Disability. Characterization of the Gdi1-null mice has revealed alterations in the total number and distribution of hippocampal and cortical synaptic vesicles, hippocampal short-term synaptic plasticity and specific short-term memory deficits in adult mice, which are possibly caused by alterations of different synaptic vesicle recycling pathways controlled by several RAB GTPases. However, interpretation of these studies is complicated by the complete ablation of Gdi1 in all cells in the brain throughout development. In this study, we generated conditionally gene-targeted mice in which the knockout of Gdi1 is restricted to the forebrain, hippocampus, cortex and amygdala and occurs only during postnatal development. Adult mutant mice reproduce the short-term memory deficit previously reported in Gdi1-null mice. Surprisingly, the delayed ablation of Gdi1 worsens the pre-synaptic phenotype at cortico-amygdala synaptic connections compared to Gdi1-null mice. These results suggest a pivotal role of αGDI via specific RAB GTPases acting specifically in forebrain regions at the pre-synaptic sites involved in memory formation.


Subject(s)
Amygdala/metabolism , Guanine Nucleotide Dissociation Inhibitors/genetics , Presynaptic Terminals/metabolism , Prosencephalon/metabolism , Synaptic Transmission/genetics , Age Factors , Amygdala/physiology , Animals , Cognition Disorders/genetics , Cognition Disorders/metabolism , Cognition Disorders/pathology , Female , Guanine Nucleotide Dissociation Inhibitors/metabolism , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/genetics , Synapses/genetics , Synapses/metabolism , Synaptic Transmission/physiology
18.
PLoS One ; 7(2): e30917, 2012.
Article in English | MEDLINE | ID: mdl-22363515

ABSTRACT

Tumor Necrosis Factor Receptor-Associated Factors (TRAFs) are major signal transducers for the TNF and interleukin-1/Toll-like receptor superfamilies. However, TRAF4 does not fit the paradigm of TRAF function in immune and inflammatory responses. Its physiological and molecular functions remain poorly understood. Behavorial analyses show that TRAF4-deficient mice (TRAF4-KO) exhibit altered locomotion coordination typical of ataxia. TRAF4-KO central nervous system (CNS) ultrastructure shows strong myelin perturbation including disorganized layers and disturbances in paranode organization. TRAF4 was previously reported to be expressed by CNS neurons. Using primary cell culture, we now show that TRAF4 is also expressed by oligodendrocytes, at all stages of their differentiation. Moreover, histology and electron microscopy show degeneration of a high number of Purkinje cells in TRAF4-KO mice, that was confirmed by increased expression of the Bax pro-apoptotic marker (immunofluorescence), TUNEL analysis, and caspase-3 activation and PARP1 cleavage (western blotting). Consistent with this phenotype, MAG and NogoA, two myelin-induced neurite outgrowth inhibitors, and their neuron partners, NgR and p75NTR were overexpressed (Q-RT-PCR and western blotting). The strong increased phosphorylation of Rock2, a RhoA downstream target, indicated that the NgR/p75NTR/RhoA signaling pathway, known to induce actin cytoskeleton rearrangement that favors axon regeneration inhibition and neuron apoptosis, is activated in the absence of TRAF4 (western blotting). Altogether, these results provide conclusive evidence for the pivotal contribution of TRAF4 to myelination and to cerebellar homeostasis, and link the loss of TRAF4 function to demyelinating or neurodegenerative diseases.


Subject(s)
Central Nervous System/metabolism , Homeostasis , Myelin Sheath/metabolism , TNF Receptor-Associated Factor 4/metabolism , Aging/pathology , Animals , Behavior, Animal/physiology , Blotting, Western , Body Weight/physiology , Cells, Cultured , Central Nervous System/pathology , Central Nervous System/physiopathology , Fluorescent Antibody Technique , GPI-Linked Proteins/metabolism , Locomotion/physiology , Mice , Mice, Knockout , Myelin Proteins/metabolism , Myelin Sheath/pathology , Myelin-Associated Glycoprotein/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurites/metabolism , Neurons/metabolism , Neurons/pathology , Nogo Proteins , Oligodendroglia/metabolism , Oligodendroglia/pathology , Purkinje Cells/metabolism , Purkinje Cells/pathology , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , TNF Receptor-Associated Factor 4/deficiency , rhoA GTP-Binding Protein/metabolism
19.
PLoS One ; 7(1): e30180, 2012.
Article in English | MEDLINE | ID: mdl-22276158

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors are associated with many forms of synaptic plasticity. Their expression level and subunit composition undergo developmental changes in several brain regions. In the mouse cerebellum, beside a developmental switch between NR2B and NR2A/C subunits in granule cells, functional postsynaptic NMDA receptors are seen in Purkinje cells of neonate and adult but not juvenile rat and mice. A presynaptic effect of NMDA on GABA release by cerebellar interneurons was identified recently. Nevertheless whereas NMDA receptor subunits are detected on parallel fiber terminals, a presynaptic effect of NMDA on spontaneous release of glutamate has not been demonstrated. Using mouse cerebellar cultures and patch-clamp recordings we show that NMDA facilitates glutamate release onto Purkinje cells in young cultures via a presynaptic mechanism, whereas NMDA activates extrasynaptic receptors in Purkinje cells recorded in old cultures. The presynaptic effect of NMDA on glutamate release is also observed in Purkinje cells recorded in acute slices prepared from juvenile but not from adult mice and requires a specific protocol of NMDA application.


Subject(s)
Cerebellar Cortex/cytology , N-Methylaspartate/pharmacology , Purkinje Cells/metabolism , Animals , Electrophysiology , Glutamic Acid/metabolism , Mice , Organ Culture Techniques , Patch-Clamp Techniques , Purkinje Cells/drug effects , Rats , Receptors, N-Methyl-D-Aspartate/metabolism
20.
PLoS One ; 5(7): e11399, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20625482

ABSTRACT

Rett syndrome (RTT) is a neuro-developmental disorder caused by loss of function of Mecp2--methyl-CpG-binding protein 2--an epigenetic factor controlling DNA transcription. In mice, removal of Mecp2 in the forebrain recapitulates most of behavioral deficits found in global Mecp2 deficient mice, including amygdala-related hyper-anxiety and lack of social interaction, pointing a role of Mecp2 in emotional learning. Yet very little is known about the establishment and maintenance of synaptic function in the adult amygdala and the role of Mecp2 in these processes. Here, we performed a longitudinal examination of synaptic properties at excitatory projections to principal cells of the lateral nucleus of the amygdala (LA) in Mecp2 mutant mice and their wild-type littermates. We first show that during animal life, Cortico-LA projections switch from a tonic to a phasic mode, whereas Thalamo-LA synapses are phasic at all ages. In parallel, we observed a specific elimination of Cortico-LA synapses and a decrease in their ability of generating presynaptic long term potentiation. In absence of Mecp2, both synaptic maturation and synaptic elimination were exaggerated albeit still specific to cortical projections. Surprisingly, associative LTP was unaffected at Mecp2 deficient synapses suggesting that synaptic maintenance rather than activity-dependent synaptic learning may be causal in RTT physiopathology. Finally, because the timing of synaptic evolution was preserved, we propose that some of the developmental effects of Mecp2 may be exerted within an endogenous program and restricted to synapses which maturate during animal life.


Subject(s)
Amygdala/metabolism , Methyl-CpG-Binding Protein 2/physiology , Rett Syndrome/metabolism , Rett Syndrome/physiopathology , Animals , Disease Models, Animal , In Vitro Techniques , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Rett Syndrome/genetics , Synaptic Potentials/genetics , Synaptic Potentials/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...