Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
PLoS Biol ; 22(5): e3002596, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38718086

RÉSUMÉ

Autism spectrum disorders (ASD) frequently accompany macrocephaly, which often involves hydrocephalic enlargement of brain ventricles. Katnal2 is a microtubule-regulatory protein strongly linked to ASD, but it remains unclear whether Katnal2 knockout (KO) in mice leads to microtubule- and ASD-related molecular, synaptic, brain, and behavioral phenotypes. We found that Katnal2-KO mice display ASD-like social communication deficits and age-dependent progressive ventricular enlargements. The latter involves increased length and beating frequency of motile cilia on ependymal cells lining ventricles. Katnal2-KO hippocampal neurons surrounded by enlarged lateral ventricles show progressive synaptic deficits that correlate with ASD-like transcriptomic changes involving synaptic gene down-regulation. Importantly, early postnatal Katnal2 re-expression prevents ciliary, ventricular, and behavioral phenotypes in Katnal2-KO adults, suggesting a causal relationship and a potential treatment. Therefore, Katnal2 negatively regulates ependymal ciliary function and its deletion in mice leads to ependymal ciliary hyperfunction and hydrocephalus accompanying ASD-related behavioral, synaptic, and transcriptomic changes.


Sujet(s)
Trouble du spectre autistique , Cils vibratiles , Épendyme , Souris knockout , Phénotype , Animaux , Mâle , Souris , Trouble du spectre autistique/génétique , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/physiopathologie , Comportement animal , Cils vibratiles/métabolisme , Modèles animaux de maladie humaine , Épendyme/métabolisme , Hippocampe/métabolisme , Hydrocéphalie/génétique , Hydrocéphalie/métabolisme , Hydrocéphalie/anatomopathologie , Hydrocéphalie/physiopathologie , Katanine/métabolisme , Katanine/génétique , Souris de lignée C57BL , Neurones/métabolisme , Synapses/métabolisme , Transcriptome/génétique
2.
Anal Chim Acta ; 1306: 342623, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38692796

RÉSUMÉ

BACKGROUND: Brain-derived exosomes circulate in the bloodstream and other bodily fluids, serving as potential indicators of neurological disease progression. These exosomes present a promising avenue for the early and precise diagnosis of neurodegenerative conditions. Notably, miRNAs found in plasma extracellular vesicles (EVs) offer distinct diagnostic benefits due to their stability, abundance, and resistance to breakdown. RESULTS: In this study, we introduce a method using transferrin conjugated magnetic nanoparticles (TMNs) to isolate these exosomes from the plasma of patients with neurological disorders. This TMNs technique is both quick (<35 min) and cost-effective, requiring no high-priced ingredients or elaborate equipment for EV extraction. Our method successfully isolated EVs from 33 human plasma samples, including those from patients with Parkinson's disease (PD), Multiple Sclerosis (MS), and Dementia. Using quantitative polymerase chain reaction (PCR) analysis, we evaluated the potential of 8 exosomal miRNA profiles as biomarker candidates. Six exosomal miRNA biomarkers (miR-195-5p, miR-495-3p, miR-23b-3P, miR-30c-2-3p, miR-323a-3p, and miR-27a-3p) were consistently linked with all stages of PD. SIGNIFICANCE: The TMNs method provides a practical, cost-efficient way to isolate EVs from biological samples, paving the way for non-invasive neurological diagnoses. Furthermore, the identified miRNA biomarkers in these exosomes may emerge as innovative tools for precise diagnosis in neurological disorders including PD.


Sujet(s)
Exosomes , Nanoparticules de magnétite , microARN , Maladie de Parkinson , Transferrine , Humains , Maladie de Parkinson/diagnostic , Maladie de Parkinson/sang , Exosomes/composition chimique , microARN/sang , Nanoparticules de magnétite/composition chimique , Transferrine/composition chimique , Encéphale/métabolisme , Marqueurs biologiques/sang , Mâle , Femelle
3.
Mol Psychiatry ; 2024 May 04.
Article de Anglais | MEDLINE | ID: mdl-38704508

RÉSUMÉ

Sensory abnormalities are observed in ~90% of individuals with autism spectrum disorders (ASD), but the underlying mechanisms are poorly understood. GluN2B, an NMDA receptor subunit that regulates long-term depression and circuit refinement during brain development, has been strongly implicated in ASD, but whether GRIN2B mutations lead to sensory abnormalities remains unclear. Here, we report that Grin2b-mutant mice show behavioral sensory hypersensitivity and brain hyperconnectivity associated with the anterior cingulate cortex (ACC). Grin2b-mutant mice with a patient-derived C456Y mutation (Grin2bC456Y/+) show sensory hypersensitivity to mechanical, thermal, and electrical stimuli through supraspinal mechanisms. c-fos and functional magnetic resonance imaging indicate that the ACC is hyperactive and hyperconnected with other brain regions under baseline and stimulation conditions. ACC pyramidal neurons show increased excitatory synaptic transmission. Chemogenetic inhibition of ACC pyramidal neurons normalizes ACC hyperconnectivity and sensory hypersensitivity. These results suggest that GluN2B critically regulates ASD-related cortical connectivity and sensory brain functions.

4.
J Neuroinflammation ; 20(1): 225, 2023 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-37794409

RÉSUMÉ

BACKGROUND: Neuromyelitis optica spectrum disorder (NMOSD) stands out among CNS inflammatory demyelinating diseases (CIDDs) due to its unique disease characteristics, including severe clinical attacks with extensive lesions and its association with systemic autoimmune diseases. We aimed to investigate whether characteristics of B cell receptors (BCRs) differ between NMOSD and other CIDDs using high-throughput sequencing. METHODS: From a prospective cohort, we recruited patients with CIDDs and categorized them based on the presence and type of autoantibodies: NMOSD with anti-aquaporin-4 antibodies, myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) with anti-myelin oligodendrocyte glycoprotein antibodies, double-seronegative demyelinating disease (DSN), and healthy controls (HCs). The BCR features, including isotype class, clonality, somatic hypermutation (SHM), and the third complementarity-determining region (CDR3) length, were analyzed and compared among the different disease groups. RESULTS: Blood samples from 33 patients with CIDDs (13 NMOSD, 12 MOGAD, and 8 DSN) and 34 HCs were investigated for BCR sequencing. Patients with NMOSD tended to have more activated BCR features compare to the other disease groups. They showed a lower proportion of unswitched isotypes (IgM and IgD) and a higher proportion of switched isotypes (IgG), increased clonality of BCRs, higher rates of SHM, and shorter lengths of CDR3. Notably, advanced age was identified as a clinical factor associated with these activated BCR features, including increased levels of clonality and SHM rates in the NMOSD group. Conversely, no such clinical factors were found to be associated with activated BCR features in the other CIDD groups. CONCLUSIONS: NMOSD patients, among those with CIDDs, displayed the most pronounced B cell activation, characterized by higher levels of isotype class switching, clonality, SHM rates, and shorter CDR3 lengths. These findings suggest that B cell-mediated humoral immune responses and characteristics in NMOSD patients are distinct from those observed in the other CIDDs, including MOGAD. Age was identified as a clinical factor associated with BCR activation specifically in NMOSD, implying the significance of persistent B cell activation attributed to anti-aquaporin-4 antibodies, even in the absence of clinical relapses throughout an individual's lifetime.


Sujet(s)
Neuromyélite optique , Humains , Aquaporine-4 , Études prospectives , Glycoprotéine MOG , Autoanticorps , Récepteurs pour l'antigène des lymphocytes B
5.
Neuron ; 111(21): 3378-3396.e9, 2023 11 01.
Article de Anglais | MEDLINE | ID: mdl-37657442

RÉSUMÉ

A genetically valid animal model could transform our understanding of schizophrenia (SCZ) disease mechanisms. Rare heterozygous loss-of-function (LoF) mutations in GRIN2A, encoding a subunit of the NMDA receptor, greatly increase the risk of SCZ. By transcriptomic, proteomic, and behavioral analyses, we report that heterozygous Grin2a mutant mice show (1) large-scale gene expression changes across multiple brain regions and in neuronal (excitatory and inhibitory) and non-neuronal cells (astrocytes and oligodendrocytes), (2) evidence of hypoactivity in the prefrontal cortex (PFC) and hyperactivity in the hippocampus and striatum, (3) an elevated dopamine signaling in the striatum and hypersensitivity to amphetamine-induced hyperlocomotion (AIH), (4) altered cholesterol biosynthesis in astrocytes, (5) a reduction in glutamatergic receptor signaling proteins in the synapse, and (6) an aberrant locomotor pattern opposite of that induced by antipsychotic drugs. These findings reveal potential pathophysiologic mechanisms, provide support for both the "hypo-glutamate" and "hyper-dopamine" hypotheses of SCZ, and underscore the utility of Grin2a-deficient mice as a genetic model of SCZ.


Sujet(s)
Dopamine , Protéomique , Récepteurs du N-méthyl-D-aspartate , Animaux , Souris , Encéphale/métabolisme , Dopamine/métabolisme , Névroglie/métabolisme , Neurones/métabolisme , Cortex préfrontal/métabolisme , Modèles animaux de maladie humaine , Récepteurs du N-méthyl-D-aspartate/génétique
6.
Biomater Res ; 27(1): 12, 2023 Feb 16.
Article de Anglais | MEDLINE | ID: mdl-36797805

RÉSUMÉ

BACKGROUND: Brain-derived exosomes released into the blood are considered a liquid biopsy to investigate the pathophysiological state, reflecting the aberrant heterogeneous pathways of pathological progression of the brain in neurological diseases. Brain-derived blood exosomes provide promising prospects for the diagnosis of neurological diseases, with exciting possibilities for the early and sensitive diagnosis of such diseases. However, the capability of traditional exosome isolation assays to specifically isolate blood exosomes and to characterize the brain-derived blood exosomal proteins by high-throughput proteomics for clinical specimens from patients with neurological diseases cannot be assured. We report a magnetic transferrin nanoparticles (MTNs) assay, which combined transferrin and magnetic nanoparticles to isolate brain-derived blood exosomes from clinical samples. METHODS: The principle of the MTNs assay is a ligand-receptor interaction through transferrin on MTNs and transferrin receptor on exosomes, and electrostatic interaction via positively charged MTNs and negatively charged exosomes to isolate brain-derived blood exosomes. In addition, the MTNs assay is simple and rapid (< 35 min) and does not require any large instrument. We confirmed that the MTNs assay accurately and efficiently isolated exosomes from serum samples of humans with neurodegenerative diseases, such as dementia, Parkinson's disease (PD), and multiple sclerosis (MS). Moreover, we isolated exosomes from serum samples of 30 patients with three distinct neurodegenerative diseases and performed unbiased proteomic analysis to explore the pilot value of brain-derived blood protein profiles as biomarkers. RESULTS: Using comparative statistical analysis, we found 21 candidate protein biomarkers that were significantly different among three groups of neurodegenerative diseases. CONCLUSION: The MTNs assay is a convenient approach for the specific and affordable isolation of extracellular vesicles from body fluids for minimally-invasive diagnosis of neurological diseases.

8.
Biol Psychiatry ; 91(11): 934-944, 2022 06 01.
Article de Anglais | MEDLINE | ID: mdl-34556257

RÉSUMÉ

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and repetitive symptoms. A key feature of ASD is early-life manifestations of symptoms, indicative of early pathophysiological mechanisms. In mouse models of ASD, increasing evidence indicates that there are early pathophysiological mechanisms that can be corrected early to prevent phenotypic defects in adults, overcoming the disadvantage of the short-lasting effects that characterize adult-initiated treatments. In addition, the results from gene restorations indicate that ASD-related phenotypes can be rescued in some cases even after the brain has fully matured. These results suggest that we need to consider both temporal and mechanistic aspects in studies of ASD models and carefully compare genetic and nongenetic corrections. Here, we summarize the early and late corrections in mouse models of ASD by genetic and pharmacological interventions and discuss how to better integrate these results to ensure efficient and long-lasting corrections for eventual clinical translation.


Sujet(s)
Trouble du spectre autistique , Animaux , Trouble du spectre autistique/génétique , Trouble du spectre autistique/thérapie , Encéphale , Modèles animaux de maladie humaine , Souris , Phénotype
9.
Curr Biol ; 31(24): 5450-5461.e4, 2021 12 20.
Article de Anglais | MEDLINE | ID: mdl-34687608

RÉSUMÉ

A sparse population of neurons active during a learning event has been identified as memory engram cells. However, cells that are recruited to support memory when experience is repeated have been scarcely explored. Evidence from previous studies provides contradictory views. To address these questions, we employed learning-dependent cell labeling in the lateral amygdala (LA) and applied electrophysiological recording, spine imaging, and optogenetic tools to the labeled neurons with or without retraining. We found that engram cells established from original fear learning became dispensable for memory retrieval specifically with relearning, and this correlated with a reduction of synaptic transmission and loss of dendritic spines in these neurons. Despite such decreased connectivity, direct activation of these neurons resulted in fear-memory recall. We further identified that repeated memory was encoded in neurons active during relearning. These results suggest a shift in neuronal ensembles encoding fear memory in the LA by relearning through disconnection of the existing engram neurons established from original experience.


Sujet(s)
Groupe nucléaire basolatéral , Peur , Animaux , Groupe nucléaire basolatéral/physiologie , Peur/physiologie , Mémoire/physiologie , Souris , Souris de lignée C57BL , Optogénétique
10.
Commun Biol ; 4(1): 1138, 2021 09 29.
Article de Anglais | MEDLINE | ID: mdl-34588597

RÉSUMÉ

Many synaptic adhesion molecules positively regulate synapse development and function, but relatively little is known about negative regulation. SALM4/Lrfn3 (synaptic adhesion-like molecule 4/leucine rich repeat and fibronectin type III domain containing 3) inhibits synapse development by suppressing other SALM family proteins, but whether SALM4 also inhibits synaptic function and specific behaviors remains unclear. Here we show that SALM4-knockout (Lrfn3-/-) male mice display enhanced contextual fear memory consolidation (7-day post-training) but not acquisition or 1-day retention, and exhibit normal cued fear, spatial, and object-recognition memory. The Lrfn3-/- hippocampus show increased currents of GluN2B-containing N-methyl-D-aspartate (NMDA) receptors (GluN2B-NMDARs), but not α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors (AMPARs), which requires the presynaptic receptor tyrosine phosphatase PTPσ. Chronic treatment of Lrfn3-/- mice with fluoxetine, a selective serotonin reuptake inhibitor used to treat excessive fear memory that directly inhibits GluN2B-NMDARs, normalizes NMDAR function and contextual fear memory consolidation in Lrfn3-/- mice, although the GluN2B-specific NMDAR antagonist ifenprodil was not sufficient to reverse the enhanced fear memory consolidation. These results suggest that SALM4 suppresses excessive GluN2B-NMDAR (not AMPAR) function and fear memory consolidation (not acquisition).


Sujet(s)
Molécules d'adhérence cellulaire neuronale/génétique , Peur/physiologie , Consolidation de la mémoire/physiologie , Récepteurs du N-méthyl-D-aspartate/génétique , Animaux , Molécules d'adhérence cellulaire neuronale/métabolisme , Souris , Souris knockout , Récepteurs du N-méthyl-D-aspartate/métabolisme
11.
Nat Commun ; 12(1): 2695, 2021 05 11.
Article de Anglais | MEDLINE | ID: mdl-33976205

RÉSUMÉ

mTOR signaling, involving mTORC1 and mTORC2 complexes, critically regulates neural development and is implicated in various brain disorders. However, we do not fully understand all of the upstream signaling components that can regulate mTOR signaling, especially in neurons. Here, we show a direct, regulated inhibition of mTOR by Tanc2, an adaptor/scaffolding protein with strong neurodevelopmental and psychiatric implications. While Tanc2-null mice show embryonic lethality, Tanc2-haploinsufficient mice survive but display mTORC1/2 hyperactivity accompanying synaptic and behavioral deficits reversed by mTOR-inhibiting rapamycin. Tanc2 interacts with and inhibits mTOR, which is suppressed by mTOR-activating serum or ketamine, a fast-acting antidepressant. Tanc2 and Deptor, also known to inhibit mTORC1/2 minimally affecting neurodevelopment, distinctly inhibit mTOR in early- and late-stage neurons. Lastly, Tanc2 inhibits mTORC1/2 in human neural progenitor cells and neurons. In summary, our findings show that Tanc2 is a mTORC1/2 inhibitor affecting neurodevelopment.


Sujet(s)
Encéphale/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Neurones/métabolisme , Protéines/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Animaux , Encéphale/embryologie , Encéphale/croissance et développement , Cellules cultivées , Cellules HEK293 , Humains , Immunosuppresseurs/pharmacologie , Incapacités d'apprentissage/génétique , Incapacités d'apprentissage/physiopathologie , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Apprentissage du labyrinthe/physiologie , Troubles de la mémoire/génétique , Troubles de la mémoire/physiopathologie , Souris de lignée C57BL , Souris knockout , Plasticité neuronale/effets des médicaments et des substances chimiques , Plasticité neuronale/génétique , Plasticité neuronale/physiologie , Protéines/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Sirolimus/pharmacologie
12.
PLoS Biol ; 18(4): e3000717, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32353004

RÉSUMÉ

Extensive evidence links Glutamate receptor, ionotropic, NMDA2B (GRIN2B), encoding the GluN2B/NR2B subunit of N-methyl-D-aspartate receptors (NMDARs), with various neurodevelopmental disorders, including autism spectrum disorders (ASDs), but the underlying mechanisms remain unclear. In addition, it remains unknown whether mutations in GluN2B, which starts to be expressed early in development, induces early pathophysiology that can be corrected by early treatments for long-lasting effects. We generated and characterized Grin2b-mutant mice that carry a heterozygous, ASD-risk C456Y mutation (Grin2b+/C456Y). In Grin2b+/C456Y mice, GluN2B protein levels were strongly reduced in association with decreased hippocampal NMDAR currents and NMDAR-dependent long-term depression (LTD) but unaltered long-term potentiation, indicative of mutation-induced protein degradation and LTD sensitivity. Behaviorally, Grin2b+/C456Y mice showed normal social interaction but exhibited abnormal anxiolytic-like behavior. Importantly, early, but not late, treatment of young Grin2b+/C456Y mice with the NMDAR agonist D-cycloserine rescued NMDAR currents and LTD in juvenile mice and improved anxiolytic-like behavior in adult mice. Therefore, GluN2B-C456Y haploinsufficiency decreases GluN2B protein levels, NMDAR-dependent LTD, and anxiety-like behavior, and early activation of NMDAR function has long-lasting effects on adult mouse behavior.


Sujet(s)
Anxiété/génétique , Hippocampe/physiologie , Dépression synaptique à long terme/physiologie , Récepteurs du N-méthyl-D-aspartate/génétique , Animaux , Anxiété/physiopathologie , Comportement animal/effets des médicaments et des substances chimiques , Cyclosérine/pharmacologie , Potentiels post-synaptiques excitateurs/génétique , Techniques de knock-in de gènes , Haploinsuffisance/génétique , Hétérozygote , Hippocampe/métabolisme , Dépression synaptique à long terme/effets des médicaments et des substances chimiques , Souches mutantes de souris , Mutation , Protéines de tissu nerveux/métabolisme , Récepteurs du N-méthyl-D-aspartate/agonistes , Récepteurs du N-méthyl-D-aspartate/métabolisme
13.
Elife ; 92020 03 06.
Article de Anglais | MEDLINE | ID: mdl-32142410

RÉSUMÉ

Synaptic adhesion molecules regulate synapse development and function. However, whether and how presynaptic adhesion molecules regulate postsynaptic NMDAR function remains largely unclear. Presynaptic LAR family receptor tyrosine phosphatases (LAR-RPTPs) regulate synapse development through mechanisms that include trans-synaptic adhesion; however, whether they regulate postsynaptic receptor functions remains unknown. Here we report that presynaptic PTPσ, a LAR-RPTP, enhances postsynaptic NMDA receptor (NMDAR) currents and NMDAR-dependent synaptic plasticity in the hippocampus. This regulation does not involve trans-synaptic adhesions of PTPσ, suggesting that the cytoplasmic domains of PTPσ, known to have tyrosine phosphatase activity and mediate protein-protein interactions, are important. In line with this, phosphotyrosine levels of presynaptic proteins, including neurexin-1, are strongly increased in PTPσ-mutant mice. Behaviorally, PTPσ-dependent NMDAR regulation is important for social and reward-related novelty recognition. These results suggest that presynaptic PTPσ regulates postsynaptic NMDAR function through trans-synaptic and direct adhesion-independent mechanisms and novelty recognition in social and reward contexts.


Sujet(s)
Receptor-Like Protein Tyrosine Phosphatases, Class 2/métabolisme , Récepteurs du N-méthyl-D-aspartate/métabolisme , Synapses/physiologie , Animaux , Régulation de l'expression des gènes/physiologie , Souris , Souris transgéniques , Neuroimagerie , Test en champ ouvert , Receptor-Like Protein Tyrosine Phosphatases, Class 2/génétique , Récepteurs du N-méthyl-D-aspartate/génétique , Transmission synaptique/physiologie
14.
PLoS Biol ; 17(6): e2005326, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-31166939

RÉSUMÉ

Netrin-G ligand-3 (NGL-3) is a postsynaptic adhesion molecule known to directly interact with the excitatory postsynaptic scaffolding protein postsynaptic density-95 (PSD-95) and trans-synaptically with leukocyte common antigen-related (LAR) family receptor tyrosine phosphatases to regulate presynaptic differentiation. Although NGL-3 has been implicated in the regulation of excitatory synapse development by in vitro studies, whether it regulates synapse development or function, or any other features of brain development and function, is not known. Here, we report that mice lacking NGL-3 (Ngl3-/- mice) show markedly suppressed normal brain development and postnatal survival and growth. A change of the genetic background of mice from pure to hybrid minimized these developmental effects but modestly suppressed N-methyl-D-aspartate (NMDA) receptor (NMDAR)-mediated synaptic transmission in the hippocampus without affecting synapse development, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor (AMPAR)-mediated basal transmission, and presynaptic release. Intriguingly, long-term depression (LTD) was near-completely abolished in Ngl3-/- mice, and the Akt/glycogen synthase kinase 3ß (GSK3ß) signaling pathway, known to suppress LTD, was abnormally enhanced. In addition, pharmacological inhibition of Akt, but not activation of NMDARs, normalized the suppressed LTD in Ngl3-/- mice, suggesting that Akt hyperactivity suppresses LTD. Ngl3-/- mice displayed several behavioral abnormalities, including hyperactivity, anxiolytic-like behavior, impaired spatial memory, and enhanced seizure susceptibility. Among them, the hyperactivity was rapidly improved by pharmacological NMDAR activation. These results suggest that NGL-3 regulates brain development, Akt/GSK3ß signaling, LTD, and locomotive and cognitive behaviors.


Sujet(s)
Encéphale/embryologie , Encéphale/croissance et développement , Protéines liées au GPI/métabolisme , Protéines de tissu nerveux/métabolisme , Animaux , Encéphale/métabolisme , Protéines liées au GPI/génétique , Glycogen synthase kinase 3 beta/métabolisme , Hippocampe/métabolisme , Ligands , Dépression synaptique à long terme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Protéines de tissu nerveux/génétique , Nétrines/métabolisme , Plasticité neuronale , Neurones/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Récepteur de l'AMPA/métabolisme , Récepteurs du N-méthyl-D-aspartate/métabolisme , Transduction du signal , Synapses/métabolisme , Synapses/physiologie , Transmission synaptique
15.
Front Mol Neurosci ; 12: 145, 2019.
Article de Anglais | MEDLINE | ID: mdl-31249508

RÉSUMÉ

Nav1.2, a voltage-gated sodium channel subunit encoded by the Scn2a gene, has been implicated in various brain disorders, including epilepsy, autism spectrum disorder, intellectual disability, and schizophrenia. Nav1.2 is known to regulate the generation of action potentials in the axon initial segment and their propagation along axonal pathways. Nav1.2 also regulates synaptic integration and plasticity by promoting back-propagation of action potentials to dendrites, but whether Nav1.2 deletion in mice affects neuronal excitability, synaptic transmission, synaptic plasticity, and/or disease-related animal behaviors remains largely unclear. Here, we report that mice heterozygous for the Scn2a gene (Scn2a +/- mice) show decreased neuronal excitability and suppressed excitatory synaptic transmission in the presence of network activity in the hippocampus. In addition, Scn2a +/- mice show suppressed hippocampal long-term potentiation (LTP) in association with impaired spatial learning and memory, but show largely normal locomotor activity, anxiety-like behavior, social interaction, repetitive behavior, and whole-brain excitation. These results suggest that Nav1.2 regulates hippocampal neuronal excitability, excitatory synaptic drive, LTP, and spatial learning and memory in mice.

16.
Cell Rep ; 23(13): 3839-3851, 2018 06 26.
Article de Anglais | MEDLINE | ID: mdl-29949768

RÉSUMÉ

Netrin-G ligand 2 (NGL-2)/LRRC4, implicated in autism spectrum disorders and schizophrenia, is a leucine-rich repeat-containing postsynaptic adhesion molecule that interacts intracellularly with the excitatory postsynaptic scaffolding protein PSD-95 and trans-synaptically with the presynaptic adhesion molecule netrin-G2. Functionally, NGL-2 regulates excitatory synapse development and synaptic transmission. However, whether it regulates synaptic plasticity and disease-related specific behaviors is not known. Here, we report that mice lacking NGL-2 (Lrrc4-/- mice) show suppressed N-Methyl-D-aspartate receptor (NMDAR)-dependent synaptic plasticity in the hippocampus. NGL-2 associates with NMDARs through both PSD-95-dependent and -independent mechanisms. Moreover, Lrrc4-/- mice display mild social interaction deficits and repetitive behaviors that are rapidly improved by pharmacological NMDAR activation. These results suggest that NGL-2 promotes synaptic stabilization of NMDARs, regulates NMDAR-dependent synaptic plasticity, and prevents autistic-like behaviors from developing in mice, supporting the hypothesis that NMDAR dysfunction contributes to autism spectrum disorders.


Sujet(s)
Trouble autistique/anatomopathologie , Protéines de tissu nerveux/génétique , Récepteurs du N-méthyl-D-aspartate/métabolisme , Animaux , Trouble autistique/métabolisme , Cyclosérine/pharmacologie , Modèles animaux de maladie humaine , Homologue-4 de la protéine Disks Large/composition chimique , Homologue-4 de la protéine Disks Large/métabolisme , Appareil de Golgi/métabolisme , Hippocampe/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Protéines de tissu nerveux/déficit , Plasticité neuronale/effets des médicaments et des substances chimiques , Liaison aux protéines , Sous-unités de protéines/composition chimique , Sous-unités de protéines/métabolisme , Récepteurs du N-méthyl-D-aspartate/agonistes , Apprentissage spatial , Synapses/métabolisme , Transmission synaptique/effets des médicaments et des substances chimiques
17.
J Neurosci ; 38(17): 4076-4092, 2018 04 25.
Article de Anglais | MEDLINE | ID: mdl-29572432

RÉSUMÉ

Shank2 is an excitatory postsynaptic scaffolding protein implicated in synaptic regulation and psychiatric disorders including autism spectrum disorders. Conventional Shank2-mutant (Shank2-/-) mice display several autistic-like behaviors, including social deficits, repetitive behaviors, hyperactivity, and anxiety-like behaviors. However, cell-type-specific contributions to these behaviors have remained largely unclear. Here, we deleted Shank2 in specific cell types and found that male mice lacking Shank2 in excitatory neurons (CaMKII-Cre;Shank2fl/fl) show social interaction deficits and mild social communication deficits, hyperactivity, and anxiety-like behaviors. In particular, male mice lacking Shank2 in GABAergic inhibitory neurons (Viaat-Cre;Shank2fl/fl) display social communication deficits, repetitive self-grooming, and mild hyperactivity. These behavioral changes were associated with distinct changes in hippocampal and striatal synaptic transmission in the two mouse lines. These results indicate that cell-type-specific deletions of Shank2 in mice lead to differential synaptic and behavioral abnormalities.SIGNIFICANCE STATEMENT Shank2 is an abundant excitatory postsynaptic scaffolding protein implicated in the regulation of excitatory synapses and diverse psychiatric disorders including autism spectrum disorders. Previous studies have reported in vivo functions of Shank2 mainly using global Shank2-null mice, but it remains largely unclear how individual cell types contribute to Shank2-dependent regulation of neuronal synapses and behaviors. Here, we have characterized conditional Shank2-mutant mice carrying the Shank2 deletion in excitatory and inhibitory neurons. These mouse lines display distinct alterations of synaptic transmission in the hippocampus and striatum that are associated with differential behavioral abnormalities in social, repetitive, locomotor, and anxiety-like domains.


Sujet(s)
Anxiété/génétique , Neurones GABAergiques/métabolisme , Relations interpersonnelles , Protéines de tissu nerveux/génétique , Transmission synaptique , Animaux , Anxiété/physiopathologie , Corps strié/cytologie , Corps strié/métabolisme , Corps strié/physiologie , Neurones GABAergiques/physiologie , Hippocampe/cytologie , Hippocampe/métabolisme , Hippocampe/physiologie , Mâle , Souris , Souris de lignée C57BL , Activité motrice , Protéines de tissu nerveux/métabolisme , Phénotype
18.
Nat Commun ; 6: 7168, 2015 May 18.
Article de Anglais | MEDLINE | ID: mdl-25981743

RÉSUMÉ

Genetic aspects of autism spectrum disorders (ASDs) have recently been extensively explored, but environmental influences that affect ASDs have received considerably less attention. Zinc (Zn) is a nutritional factor implicated in ASDs, but evidence for a strong association and linking mechanism is largely lacking. Here we report that trans-synaptic Zn mobilization rapidly rescues social interaction in two independent mouse models of ASD. In mice lacking Shank2, an excitatory postsynaptic scaffolding protein, postsynaptic Zn elevation induced by clioquinol (a Zn chelator and ionophore) improves social interaction. Postsynaptic Zn is mainly derived from presynaptic pools and activates NMDA receptors (NMDARs) through postsynaptic activation of the tyrosine kinase Src. Clioquinol also improves social interaction in mice haploinsufficient for the transcription factor Tbr1, which accompanies NMDAR activation in the amygdala. These results suggest that trans-synaptic Zn mobilization induced by clioquinol rescues social deficits in mouse models of ASD through postsynaptic Src and NMDAR activation.


Sujet(s)
Trouble autistique/physiopathologie , Récepteurs du N-méthyl-D-aspartate/métabolisme , Synapses/métabolisme , Zinc/composition chimique , Amygdale (système limbique)/métabolisme , Animaux , Comportement animal , Chélateurs/composition chimique , Clioquinol/composition chimique , Croisements génétiques , Protéines de liaison à l'ADN/métabolisme , Dendrites/métabolisme , Modèles animaux de maladie humaine , Électrophysiologie , Femelle , Haploinsuffisance , Hippocampe/métabolisme , Ionophores/composition chimique , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Protéines de tissu nerveux/métabolisme , Comportement social , Transmission synaptique/physiologie , Protéines à domaine boîte-T , src-Family kinases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE