Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-32372941

ABSTRACT

Epileptogenesis is the gradual process responsible for converting a healthy brain into an epileptic brain. This process can be triggered by a wide range of factors, including brain injury or tumors, infections, and status epilepticus. Epileptogenesis results in aberrant synaptic plasticity, neuroinflammation and seizure-induced cell death. As Matrix Metalloproteinases (MMPs) play a crucial role in cellular plasticity by remodeling the extracellular matrix (ECM), gelatinases (MMP-2 and MMP-9) were recently highlighted as key players in epileptogenesis. In this work, we engineered a biosensor to report in situ gelatinase activity in a model of epileptogenesis. This biosensor encompasses a gelatinase-sensitive activatable cell penetrating peptide (ACPP) coupled to a TAMRA fluorophore, allowing fluorescence uptake in cells displaying endogenous gelatinase activities. In a preclinical mouse model of temporal lobe epilepsy (TLE), the intrahippocampal kainate injection, ACPPs revealed a localized distribution of gelatinase activities, refining temporal cellular changes during epileptogenesis. The activity was found particularly but not only in the ipsilateral hippocampus, starting from the CA1 area and spreading to dentate gyrus from the early stages throughout chronic epilepsy, notably in neurons and microglial cells. Thus, our work shows that ACPPs are suitable molecular imaging probes for detecting the spatiotemporal pattern of gelatinase activity during epileptogenesis, suggesting their possible use as vectors to target cellular reactive changes with treatment for epileptogenesis.

2.
J Clin Invest ; 130(3): 1168-1184, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32039920

ABSTRACT

Dopamine receptor D1 modulates glutamatergic transmission in cortico-basal ganglia circuits and represents a major target of L-DOPA therapy in Parkinson's disease. Here we show that D1 and metabotropic glutamate type 5 (mGlu5) receptors can form previously unknown heteromeric entities with distinctive functional properties. Interacting with Gq proteins, cell-surface D1-mGlu5 heteromers exacerbated PLC signaling and intracellular calcium release in response to either glutamate or dopamine. In rodent models of Parkinson's disease, D1-mGlu5 nanocomplexes were strongly upregulated in the dopamine-denervated striatum, resulting in a synergistic activation of PLC signaling by D1 and mGlu5 receptor agonists. In turn, D1-mGlu5-dependent PLC signaling was causally linked with excessive activation of extracellular signal-regulated kinases in striatal neurons, leading to dyskinesia in animals treated with L-DOPA or D1 receptor agonists. The discovery of D1-mGlu5 functional heteromers mediating maladaptive molecular and motor responses in the dopamine-denervated striatum may prompt the development of new therapeutic principles for Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , MAP Kinase Signaling System , Neurons/metabolism , Parkinson Disease, Secondary/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Dopamine D1/metabolism , Animals , Corpus Striatum/pathology , HEK293 Cells , Humans , Mice , Mice, Knockout , Multiprotein Complexes/agonists , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurons/pathology , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/pathology , Rats , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/genetics
3.
Neurobiol Dis ; 129: 13-28, 2019 09.
Article in English | MEDLINE | ID: mdl-31051234

ABSTRACT

Finding new targets to control or reduce seizure activity is essential to improve the management of epileptic patients. We hypothesized that activation of the pre-synaptic and inhibitory metabotropic glutamate receptor type 7 (mGlu7) reduces spontaneous seizures. We tested LSP2-9166, a recently developed mGlu7/4 agonist with unprecedented potency on mGlu7 receptors, in two paradigms of epileptogenesis. In a model of chemically induced epileptogenesis (pentylenetetrazole systemic injection), LSP2-9166 induces an anti-epileptogenic effect rarely observed in preclinical studies. In particular, we found a bidirectional modulation of seizure progression by mGlu4 and mGlu7 receptors, the latter preventing kindling. In the intra-hippocampal injection of kainic acid mouse model that mimics the human mesial temporal lobe epilepsy, we found that LSP2-9166 reduces seizure frequency and hippocampal sclerosis. LSP2-9166 also acts as an anti-seizure drug on established seizures in both models tested. Specific modulation of the mGlu7 receptor could represent a novel approach to reduce pathological network remodeling.


Subject(s)
Aminobutyrates/pharmacology , Anticonvulsants/pharmacology , Hippocampus/drug effects , Receptors, Metabotropic Glutamate/agonists , Seizures/metabolism , Animals , Epilepsy/metabolism , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/metabolism , Kindling, Neurologic/drug effects , Mice , Mice, Mutant Strains
4.
J Mol Cell Biol ; 10(5): 423-436, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29762713

ABSTRACT

Metabotropic glutamate receptors are expressed at excitatory synapses and control synaptic transmission in mammalian brain. These receptors are involved in numerous patho-physiological functions. However, little is known about the molecular determinants responsible for their intracellular transport and membrane targeting. Here we investigated the nature of the molecular motor and adaptor protein responsible for trafficking and membrane localization of the group I metabotropic glutamate mGlu1 postsynaptic receptor in cultured hippocampal neurons. In proteomic studies, we identified the synaptosome-associated protein 23 (SNAP23) and the molecular motor Kif5 kinesin as proteins interacting with mGlu1 receptor. We showed that SNAP23, but not Kif5, directly interacts with mGlu1 receptor carboxyl terminus. Using a recombination approach to impair or enhance the interaction between SNAP23 and Kif5, we found that the SNAP23-Kif5 complex controls the trafficking of mGlu1 receptor along microtubules. Additional fluorescence recovery after cleavage experiments allowed us to identify a role of the complex in the receptor cell surface targeting. In conclusion, our study indicates that along dendritic processes Kif5-SNAP23 complex contributes to proper mGlu1 receptor trafficking and cell surface expression.


Subject(s)
Kinesins/metabolism , Microtubules/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Cells, Cultured , Female , Hippocampus/cytology , Kinesins/genetics , Male , Mice, Transgenic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurons/metabolism , Protein Interaction Domains and Motifs , Protein Transport , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , Rats, Wistar , Receptors, Metabotropic Glutamate/genetics
5.
Neuropharmacology ; 115: 92-99, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27276689

ABSTRACT

The orphan Glutamate receptor Delta2 (GluD2) intrinsic ion channel activity is indirectly triggered by glutamate through stimulation of the metabotropic glutamate receptor 1 (mGlu1), in cerebellar Purkinje cells. However, the mechanisms of GluD2 ion channel opening are entirely unknown. In this work, we investigated the signaling pathways underlying the mGlu1-induced GluD2 current, performing whole-cell voltage-clamp recordings from mGlu1 and GluD2 transfected HEK293 cells. We show that the activation of GluD2 channels via DHPG-induced mGlu1 stimulation is Gαq-dependent. Moreover, inhibition of the downstream components of the mGlu1 canonical signaling pathway PLC and PKC with U73122 and GF109203X, respectively, strongly reduced the DHPG-induced GluD2 current. These results were further confirmed on endogenous receptors at the Parallel Fiber - Purkinje Cell cerebellar synapse, indicating that the opening of the GluD2 channel by mGlu1 receptor mobilizes the canonical Gq-PLC-PKC pathway. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.


Subject(s)
Receptors, Glutamate/physiology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Animals , Cerebellum/drug effects , Cerebellum/physiology , Estrenes/pharmacology , HEK293 Cells , Humans , Indoles/pharmacology , Maleimides/pharmacology , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Pyrrolidinones/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Signal Transduction/drug effects
6.
Front Neural Circuits ; 10: 31, 2016.
Article in English | MEDLINE | ID: mdl-27199672

ABSTRACT

Mutation of the metabotropic glutamate receptor type 7 (mGlu7) induces absence-like epileptic seizures, but its precise role in the somatosensory thalamocortical network remains unknown. By combining electrophysiological recordings, optogenetics, and pharmacology, we dissected the contribution of the mGlu7 receptor at mouse thalamic synapses. We found that mGlu7 is functionally expressed at both glutamatergic and GABAergic synapses, where it can inhibit neurotransmission and regulate short-term plasticity. These effects depend on the PDZ-ligand of the receptor, as they are lost in mutant mice. Interestingly, the very low affinity of mGlu7 receptors for glutamate raises the question of how it can be activated, namely at GABAergic synapses and in basal conditions. Inactivation of the receptor activity with the mGlu7 negative allosteric modulator (NAM), ADX71743, enhances thalamic synaptic transmission. In vivo administration of the NAM induces a lethargic state with spindle and/or spike-and-wave discharges accompanied by a behavioral arrest typical of absence epileptic seizures. This provides evidence for mGlu7 receptor-mediated tonic modulation of a physiological function in vivo preventing synchronous and potentially pathological oscillations.


Subject(s)
Cerebral Cortex/cytology , Neural Pathways/physiology , Receptors, Metabotropic Glutamate/metabolism , Thalamus/physiology , Animals , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Cerebral Cortex/physiology , Channelrhodopsins , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GABA Agents/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neurons/physiology , Post-Synaptic Density/drug effects , Post-Synaptic Density/genetics , Receptors, GABA-A/physiology , Receptors, Metabotropic Glutamate/genetics , Synaptic Potentials/drug effects , Synaptic Potentials/genetics
7.
Neurotoxicology ; 54: 140-152, 2016 05.
Article in English | MEDLINE | ID: mdl-27108687

ABSTRACT

Exposure to organophosphorus (OP) compounds, either pesticides or chemical warfare agents, represents a major health problem. As potent irreversible inhibitors of cholinesterase, OP may induce seizures, as in status epilepticus, and occasionally brain lesions. Although these compounds are extremely toxic agents, the search for novel antidotes remains extremely limited. In silico modeling constitutes a useful tool to identify pharmacological targets and to develop efficient therapeutic strategies. In the present work, we developed a new in silico simulator in order to predict the neurotoxicity of irreversible inhibitors of acetyl- and/or butyrylcholinesterase (ChE) as well as the potential neuroprotection provided by antagonists of cholinergic muscarinic and glutamate N-methyl-d-aspartate (NMDA) receptors. The simulator reproduced firing of CA1 hippocampal neurons triggered by exposure to paraoxon (POX), as found in patch-clamp recordings in in vitro mouse hippocampal slices. In the case of POX intoxication, it predicted a preventing action of the muscarinic receptor antagonist atropine sulfate, as well as a synergistic action with the non-competitive NMDA receptor antagonist memantine. These in silico predictions relative to beneficial effects of atropine sulfate combined with memantine were recapitulated experimentally in an in vivo model of POX in adult male Swiss mice using electroencephalic (EEG) recordings. Thus, our simulator is a new powerful tool to identify protective therapeutic strategies against OP central effects, by screening various combinations of muscarinic and NMDA receptor antagonists.


Subject(s)
Computer Simulation , Models, Neurological , Neurotoxicity Syndromes/etiology , Organophosphates/toxicity , Paraoxon/toxicity , Acetylcholinesterase/metabolism , Animals , Brain Waves/drug effects , Cholinesterase Reactivators/pharmacology , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Memantine/pharmacology , Membrane Potentials/drug effects , Mice , Neurons/drug effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/physiopathology , Oximes/pharmacology , Pyridinium Compounds/pharmacology
8.
Elife ; 5: e12430, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26977767

ABSTRACT

Synapses and nuclei are connected by bidirectional communication mechanisms that enable information transfer encoded by macromolecules. Here, we identified RNF10 as a novel synaptonuclear protein messenger. RNF10 is activated by calcium signals at the postsynaptic compartment and elicits discrete changes at the transcriptional level. RNF10 is enriched at the excitatory synapse where it associates with the GluN2A subunit of NMDA receptors (NMDARs). Activation of synaptic GluN2A-containing NMDARs and induction of long term potentiation (LTP) lead to the translocation of RNF10 from dendritic segments and dendritic spines to the nucleus. In particular, we provide evidence for importin-dependent long-distance transport from synapto-dendritic compartments to the nucleus. Notably, RNF10 silencing prevents the maintenance of LTP as well as LTP-dependent structural modifications of dendritic spines.


Subject(s)
Carrier Proteins/metabolism , Hippocampus/physiology , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Animals , Cell Nucleus/metabolism , Protein Transport , Rats
9.
Mol Biol Cell ; 26(11): 2112-27, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25851601

ABSTRACT

By regulating actin cytoskeleton dynamics, Rho GTPases and their activators RhoGEFs are implicated in various aspects of neuronal differentiation, including dendritogenesis and synaptogenesis. Purkinje cells (PCs) of the cerebellum, by developing spectacular dendrites covered with spines, represent an attractive model system in which to decipher the molecular signaling underlying these processes. To identify novel regulators of dendritic spine morphogenesis among members of the poorly characterized DOCK family of RhoGEFs, we performed gene expression profiling of fluorescence-activated cell sorting (FACS)-purified murine PCs at various stages of their postnatal differentiation. We found a strong increase in the expression of the Cdc42-specific GEF DOCK10. Depleting DOCK10 in organotypic cerebellar cultures resulted in dramatic dendritic spine defects in PCs. Accordingly, in mouse hippocampal neurons, depletion of DOCK10 or expression of a DOCK10 GEF-dead mutant led to a strong decrease in spine density and size. Conversely, overexpression of DOCK10 led to increased spine formation. We show that DOCK10 function in spinogenesis is mediated mainly by Cdc42 and its downstream effectors N-WASP and PAK3, although DOCK10 is also able to activate Rac1. Our global approach thus identifies an unprecedented function for DOCK10 as a novel regulator of dendritic spine morphogenesis via a Cdc42-mediated pathway.


Subject(s)
Cerebellum/growth & development , Dendritic Spines/physiology , Guanine Nucleotide Exchange Factors/physiology , Neurogenesis , Neurons/physiology , Purkinje Cells/physiology , Animals , Dendritic Spines/ultrastructure , Female , Flow Cytometry , Gene Expression Profiling , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neuropeptides/metabolism , Purkinje Cells/metabolism , Purkinje Cells/ultrastructure , Signal Transduction , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
10.
J Neurosci ; 35(7): 3155-73, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25698751

ABSTRACT

The charged multivesicular body proteins (Chmp1-7) are an evolutionarily conserved family of cytosolic proteins that transiently assembles into helical polymers that change the curvature of cellular membrane domains. Mutations in human CHMP2B cause frontotemporal dementia, suggesting that this protein may normally control some neuron-specific process. Here, we examined the function, localization, and interactions of neuronal Chmp2b. The protein was highly expressed in mouse brain and could be readily detected in neuronal dendrites and spines. Depletion of endogenous Chmp2b reduced dendritic branching of cultured hippocampal neurons, decreased excitatory synapse density in vitro and in vivo, and abolished activity-induced spine enlargement and synaptic potentiation. To understand the synaptic effects of Chmp2b, we determined its ultrastructural distribution by quantitative immuno-electron microscopy and its biochemical interactions by coimmunoprecipitation and mass spectrometry. In the hippocampus in situ, a subset of neuronal Chmp2b was shown to concentrate beneath the perisynaptic membrane of dendritic spines. In synaptoneurosome lysates, Chmp2b was stably bound to a large complex containing other members of the Chmp family, as well as postsynaptic scaffolds. The supramolecular Chmp assembly detected here corresponds to a stable form of the endosomal sorting complex required for transport-III (ESCRT-III), a ubiquitous cytoplasmic protein complex known to play a central role in remodeling of lipid membranes. We conclude that Chmp2b-containing ESCRT-III complexes are also present at dendritic spines, where they regulate synaptic plasticity. We propose that synaptic ESCRT-III filaments may function as a novel element of the submembrane cytoskeleton of spines.


Subject(s)
Endosomal Sorting Complexes Required for Transport/deficiency , Nerve Tissue Proteins/deficiency , Synapses/physiology , Animals , Cells, Cultured , Computer Simulation , Dendrites/metabolism , Dendrites/ultrastructure , Endosomal Sorting Complexes Required for Transport/genetics , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Hippocampus/cytology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Mutation/genetics , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/ultrastructure , Post-Synaptic Density/metabolism , Post-Synaptic Density/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/ultrastructure , Red Fluorescent Protein
11.
Eur J Neurosci ; 41(2): 157-67, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25377770

ABSTRACT

Type 1 metabotropic glutamate (mGlu1) receptors play a pivotal role in different forms of synaptic plasticity in the cerebellar cortex, e.g. long-term depression at glutamatergic synapses and rebound potentiation at GABAergic synapses. These various forms of plasticity might depend on the subsynaptic arrangement of the receptor in Purkinje cells that can be regulated by protein-protein interactions. This study investigated, by means of the freeze-fracture replica immunogold labelling method, the subcellular localization of mGlu1 receptors in the rodent cerebellum and whether Homer proteins regulate their subsynaptic distribution. We observed a widespread extrasynaptic localization of mGlu1 receptors and confirmed their peri-synaptic enrichment at glutamatergic synapses. Conversely, we detected mGlu1 receptors within the main body of GABAergic synapses onto Purkinje cell dendrites. Although Homer proteins are known to interact with the mGlu1 receptor C-terminus, we could not detect Homer3, the most abundant Homer protein in the cerebellar cortex, at GABAergic synapses by pre-embedding and post-embedding immunoelectron microscopy. We then hypothesized a critical role for Homer proteins in the peri-junctional localization of mGlu1 receptors at glutamatergic synapses. To disrupt Homer-associated protein complexes, mice were tail-vein injected with the membrane-permeable dominant-negative TAT-Homer1a. Freeze-fracture replica immunogold labelling analysis showed no significant alteration in the mGlu1 receptor distribution pattern at parallel fibre-Purkinje cell synapses, suggesting that other scaffolding proteins are involved in the peri-synaptic confinement. The identification of interactors that regulate the subsynaptic localization of the mGlu1 receptor at neurochemically distinct synapses may offer new insight into its trafficking and intracellular signalling.


Subject(s)
Cerebellar Cortex/metabolism , Glutamic Acid/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Blotting, Western , Carrier Proteins/metabolism , Cerebellar Cortex/ultrastructure , Homer Scaffolding Proteins , Immunohistochemistry , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Purkinje Cells/metabolism , Purkinje Cells/ultrastructure , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Synapses/ultrastructure
12.
Hippocampus ; 24(12): 1466-81, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25044816

ABSTRACT

The acute activation of the dopamine D1-like receptors (D1R) is involved in a plethora of functions ranging from increased locomotor activity to the facilitation of consolidation, storage, and retrieval of memories. Although much less characterized, epileptiform activities, usually triggered by disruption of the glutamate and GABA balance, have also been reported to involve the dopaminergic transmission. Using a combination of biochemical, immunohistochemical, electrophysiological, and behavioral approaches we have investigated the consequences of repeated stimulation of D1R using the selective D1R-like agonist SKF81297. Here, we report that repeated systemic administration of SKF81297 induces kindled seizures in mice. These seizure episodes parallel the hyperactivation of the mTOR signaling in the hippocampus, leading to disrupted long-term potentiation (LTP) in the dentate gyrus (DG) and altered recognition memories. The mTOR inhibitor rapamycin delays the development of SKF81297-induced kindled seizures, and rescues LTP in the DG and object recognition. Our results show that repeated stimulation of D1R is sufficient to induce generalized seizures leading to the overactivation of mTOR signaling, disrupted hippocampal plasticity, and impaired long-term recognition memories. This work highlights the interest of mTOR inhibitors as therapeutic strategies to reverse plasticity and cognitive deficits.


Subject(s)
Dentate Gyrus/physiopathology , Memory Disorders/physiopathology , Receptors, Dopamine D1/metabolism , Seizures/physiopathology , TOR Serine-Threonine Kinases/metabolism , Animals , Benzazepines/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Dentate Gyrus/drug effects , Dopamine Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Neuroprotective Agents/pharmacology , Receptors, Dopamine D1/agonists , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Seizures/chemically induced , Seizures/drug therapy , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tissue Culture Techniques
13.
J Cell Sci ; 127(Pt 16): 3451-62, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24938595

ABSTRACT

Quantitative spatio-temporal characterization of protein interactions in living cells remains a major challenge facing modern biology. We have investigated in living neurons the spatial dependence of the stoichiometry of interactions between two core proteins of the N-methyl-D-aspartate (NMDA)-receptor-associated scaffolding complex, GKAP (also known as DLGAP1) and DLC2 (also known as DYNLL2), using a novel variation of fluorescence fluctuation microscopy called two-photon scanning number and brightness (sN&B). We found that dimerization of DLC2 was required for its interaction with GKAP, which, in turn, potentiated GKAP self-association. In the dendritic shaft, the DLC2-GKAP hetero-oligomeric complexes were composed mainly of two DLC2 and two GKAP monomers, whereas, in spines, the hetero-complexes were much larger, with an average of ∼16 DLC2 and ∼13 GKAP monomers. Disruption of the GKAP-DLC2 interaction strongly destabilized the oligomers, decreasing the spine-preferential localization of GKAP and inhibiting NMDA receptor activity. Hence, DLC2 serves a hub function in the control of glutamatergic transmission by ordering GKAP-containing complexes in dendritic spines. Beyond illuminating the role of DLC2-GKAP interactions in glutamatergic signaling, these data underscore the power of the sN&B approach for quantitative spatio-temporal imaging of other important protein complexes.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurons/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dendritic Spines/metabolism , Dimerization , GTPase-Activating Proteins , Humans , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/chemistry , Protein Binding , SAP90-PSD95 Associated Proteins , Sequence Alignment , Synapses/chemistry , Synapses/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
14.
Neurobiol Dis ; 68: 47-56, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24768804

ABSTRACT

Episodic ataxia type-2 (EA2) is a dominantly inherited human neurological disorder caused by loss of function mutations in the CACNA1A gene, which encodes the CaV2.1 subunit of P/Q-type voltage-gated calcium channels. It remains however unknown whether the deficit of cerebellar CaV2.1 in adult is in direct link with the disease. To address this issue, we have used lentiviral based-vector RNA interference (RNAi) to knock-down CaV2.1 expression in the cerebellum of adult mice. We show that suppression of the P/Q-type channels in Purkinje neurons induced motor abnormalities, such as imbalance and ataxic gait. Interestingly, moderate channel suppression caused no basal ataxia, while ß-adrenergic activation and exercise mimicked stress induced motor disorders. Moreover, stress-induced ataxia was stable, non-progressive and totally abolished by acetazolamide, a carbonic anhydrase inhibitor used to treat EA2. Altogether, these data reveal that P/Q-type channel suppression in adult mice supports the episodic status of EA2 disease.


Subject(s)
Ataxia/etiology , Calcium Channels, N-Type/metabolism , Cerebellum/pathology , Nystagmus, Pathologic/etiology , Purkinje Cells/metabolism , RNA Interference/physiology , RNA, Small Interfering/physiology , Animals , Ataxia/genetics , Ataxia/pathology , Ataxia/physiopathology , Calcium Channels, N-Type/genetics , Cerebellum/metabolism , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Movement/physiology , Nystagmus, Pathologic/genetics , Nystagmus, Pathologic/pathology , Nystagmus, Pathologic/physiopathology , Postural Balance/genetics , RNA, Small Interfering/genetics , Transduction, Genetic
15.
J Biol Chem ; 289(5): 2600-9, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24352656

ABSTRACT

Development of dendritic spines is important for synaptic function, and alteration in spine morphogenesis is often associated with mental disorders. Rich2 was an uncharacterized Rho-GAP protein. Here we searched for a role of this protein in spine morphogenesis. We found that it is enriched in dendritic spines of cultured hippocampal pyramidal neurons during early stages of development. Rich2 specifically stimulated the Rac1 GTPase in these neurons. Inhibition of Rac1 by EHT 1864 increased the size and decreased the density of dendritic spines. Similarly, Rich2 overexpression increased the size and decreased the density of dendritic spines, whereas knock-down of the protein by specific si-RNA decreased both size and density of spines. The morphological changes were reflected by the increased amplitude and decreased frequency of miniature EPSCs induced by Rich2 overexpression, while si-RNA treatment decreased both amplitude and frequency of these events. Finally, treatment of neurons with EHT 1864 rescued the phenotype induced by Rich2 knock-down. These results suggested that Rich2 controls dendritic spine morphogenesis and function via inhibition of Rac1.


Subject(s)
Dendritic Spines/enzymology , GTPase-Activating Proteins/metabolism , Neurons/enzymology , Neuropeptides/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , COS Cells , Chlorocebus aethiops , Excitatory Postsynaptic Potentials/physiology , GTPase-Activating Proteins/genetics , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/growth & development , Mice , Morphogenesis/physiology , Neurogenesis/physiology , Neurons/ultrastructure , Neuropeptides/genetics , Patch-Clamp Techniques , Primary Cell Culture , rac1 GTP-Binding Protein/genetics
16.
EMBO Rep ; 15(1): 103-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24357660

ABSTRACT

The orphan GluD2 receptor belongs to the ionotropic glutamate receptor family but does not bind glutamate. Ligand-gated GluD2 currents have never been evidenced, and whether GluD2 operates as an ion channel has been a long-standing question. Here, we show that GluD2 gating is triggered by type 1 metabotropic glutamate receptors, both in a heterologous expression system and in Purkinje cells. Thus, GluD2 is not only an adhesion molecule at synapses but also works as a channel. This gating mechanism reveals new properties of glutamate receptors that emerge from their interaction and opens unexpected perspectives regarding synaptic transmission and plasticity.


Subject(s)
Receptors, Glutamate/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Calcium Signaling , Cerebellum/cytology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials , Glycine/analogs & derivatives , Glycine/pharmacology , HEK293 Cells , Humans , Ion Channel Gating , Male , Mice , Mice, Inbred C57BL , Purkinje Fibers/drug effects , Purkinje Fibers/physiology , Resorcinols/pharmacology
17.
Article in English | MEDLINE | ID: mdl-23908646

ABSTRACT

Networks of signaling molecules are activated in response to environmental changes. How are these signaling networks dynamically integrated in space and time to process particular information? To tackle this issue, biosensors of single signaling pathways have been engineered. Bioluminescence resonance energy transfer (BRET)-based biosensors have proven to be particularly efficient in that matter due to the high sensitivity of this technology to monitor protein-protein interactions or conformational changes in living cells. Extracellular signal-regulated kinases (ERK) are ubiquitously expressed and involved in many diverse cellular functions that might be encoded by the strength and spatio-temporal pattern of ERK activation. We developed a BRET-based sensor of ERK activity, called Rluc8-ERKsubstrate-Venus (REV). As expected, BRET changes of REV were correlated with ERK phosphorylation, which is required for its kinase activity. In neurons, the nature of the stimuli determines the strength, the location, or the moment of ERK activation, thus highlighting how acute modulation of ERK may encode the nature of initial stimulus to specify the consequences of this activation. This study provides evidence for suitability of REV as a new biosensor to address biological questions.

18.
J Neurosci ; 33(23): 9699-715, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23739967

ABSTRACT

Synaptic long-term potentiation (LTP) is a key mechanism involved in learning and memory, and its alteration is associated with mental disorders. Shank3 is a major postsynaptic scaffolding protein that orchestrates dendritic spine morphogenesis, and mutations of this protein lead to mental retardation and autism spectrum disorders. In the present study we investigated the role of a new Shank3-associated protein in LTP. We identified the Rho-GAP interacting CIP4 homolog 2 (Rich2) as a new Shank3 partner by proteomic screen. Using single-cell bioluminescence resonance energy transfer microscopy, we found that Rich2-Shank3 interaction is increased in dendritic spines of mouse cultured hippocampal neurons during LTP. We further characterized Rich2 as an endosomal recycling protein that controls AMPA receptor GluA1 subunit exocytosis and spine morphology. Knock-down of Rich2 with siRNA, or disruption of the Rich2-Shank3 complex using an interfering mimetic peptide, inhibited the dendritic spine enlargement and the increase in GluA1 subunit exocytosis typical of LTP. These results identify Rich2-Shank3 as a new postsynaptic protein complex involved in synaptic plasticity.


Subject(s)
Exocytosis/physiology , GTPase-Activating Proteins/metabolism , Long-Term Potentiation/physiology , Nerve Tissue Proteins/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Amino Acid Sequence , Animals , Dendritic Spines/metabolism , Female , GTPase-Activating Proteins/genetics , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Mice , Microfilament Proteins , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Protein Binding/physiology , Random Allocation , Rats , Rats, Sprague-Dawley
19.
Neuropharmacology ; 66: 151-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22465815

ABSTRACT

Restoring synaptic plasticity in neurodegenerative diseases could prevent neuronal degeneration, as well as motor and cognitive disorders. In Parkinson's disease, synaptic plasticity at corticostriatal synapses is altered. Dendrites of striatal medium spiny neurons (MSNs) receive dopaminergic inputs from the substantia nigra and glutamatergic cortical afferents. Because both glutamate and dopamine are required to induce and sustain MSNs plasticity, the particular molecular mechanisms involved at this synaptic triad are difficult to understand. In the present work, we established a convenient in vitro model of the corticostriatal synapse to study synaptic plasticity. We focused on long-term depression involving group I metabotropic glutamate (mGlu) receptors. We found that in striatal neurons co-cultured with cortical neurons, the absence of dopaminergic stimuli favored the excess of glutamatergic drive from cortical neuron terminals, thus resulting in a constitutive depression of the corticostriatal glutamatergic transmission. Indeed, concomitant blockade of group I mGlu receptors and activation of dopaminergic receptors stably reduced the depression of the synaptic transmission. Thus the dependence on glutamate and dopamine balance of the corticostriatal synapse responsiveness validates the accuracy of this manageable in vitro model to depict the molecular pathways involved in the plasticity at corticostriatal synapses and to test restorative therapeutic approaches in Parkinson's disease. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.


Subject(s)
Cerebral Cortex/physiology , Corpus Striatum/physiology , Dopamine Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptors, Dopamine/physiology , Receptors, Metabotropic Glutamate/physiology , Synaptic Transmission/physiology , Actins/genetics , Animals , Cerebral Cortex/drug effects , Coculture Techniques/methods , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dendritic Spines/ultrastructure , Female , Glutamic Acid/metabolism , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Neuronal Plasticity/physiology , Primary Cell Culture/methods , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Synaptic Transmission/drug effects
20.
J Cell Biol ; 198(2): 251-63, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22801779

ABSTRACT

Scaffolding proteins interact with membrane receptors to control signaling pathways and cellular functions. However, the dynamics and specific roles of interactions between different components of scaffold complexes are poorly understood because of the dearth of methods available to monitor binding interactions. Using a unique combination of single-cell bioluminescence resonance energy transfer imaging in living neurons and electrophysiological recordings, in this paper, we depict the role of glutamate receptor scaffold complex remodeling in space and time to control synaptic transmission. Despite a broad colocalization of the proteins in neurons, we show that spine-confined assembly/disassembly of this scaffold complex, physiologically triggered by sustained activation of synaptic NMDA (N-methyl-d-aspartate) receptors, induces physical association between ionotropic (NMDA) and metabotropic (mGlu5a) synaptic glutamate receptors. This physical interaction results in an mGlu5a receptor-mediated inhibition of NMDA currents, providing an activity-dependent negative feedback loop on NMDA receptor activity. Such protein scaffold remodeling represents a form of homeostatic control of synaptic excitability.


Subject(s)
Dendritic Spines/physiology , Synaptic Transmission/physiology , Animals , HEK293 Cells , Hippocampus/physiology , Homeostasis/physiology , Humans , Rats , Receptors, Glutamate/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...