Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(21): e2321388121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38748583

ABSTRACT

Protocadherin19 (PCDH19)-related epilepsy syndrome is a rare disorder characterized by early-onset epilepsy, intellectual disability, and autistic behaviors. PCDH19 is located on the X chromosome and encodes a calcium-dependent single-pass transmembrane protein, which regulates cell-to-cell adhesion through homophilic binding. In human, 90% of heterozygous females, containing PCDH19 wild-type and mutant cells due to random X inactivation, are affected, whereas mutant males, containing only mutant cells, are typically not. The current view, the cellular interference, is that the altered interactions between wild-type and mutant cells during development, rather than loss of function itself, are responsible. However, studies using Pcdh19 knockout mice showed that the complete loss of function also causes autism-like behaviors both in males and females, suggesting that other functions of PCDH19 may also contribute to pathogenesis. To address whether mosaicism is required for PCDH19-related epilepsy, we generated Xenopus tropicalis tadpoles with complete or mosaic loss of function by injecting antisense morpholino oligonucleotides into the blastomeres of neural lineage at different stages of development. We found that either mosaic or complete knockdown results in seizure-like behaviors, which could be rescued by antiseizure medication, and repetitive behaviors. Our results suggest that the loss of PCDH19 function itself, in addition to cellular interference, may also contribute to PCDH19-related epilepsy.


Subject(s)
Cadherins , Epilepsy , Mosaicism , Protocadherins , Xenopus , Animals , Cadherins/genetics , Cadherins/metabolism , Female , Epilepsy/genetics , Epilepsy/metabolism , Male , Behavior, Animal , Humans
2.
Bioinformatics ; 39(10)2023 10 03.
Article in English | MEDLINE | ID: mdl-37713469

ABSTRACT

MOTIVATION: Efficient assessment of the blood-brain barrier (BBB) penetration ability of a drug compound is one of the major hurdles in central nervous system drug discovery since experimental methods are costly and time-consuming. To advance and elevate the success rate of neurotherapeutic drug discovery, it is essential to develop an accurate computational quantitative model to determine the absolute logBB value (a logarithmic ratio of the concentration of a drug in the brain to its concentration in the blood) of a drug candidate. RESULTS: Here, we developed a quantitative model (LogBB_Pred) capable of predicting a logBB value of a query compound. The model achieved an R2 of 0.61 on an independent test dataset and outperformed other publicly available quantitative models. When compared with the available qualitative (classification) models that only classified whether a compound is BBB-permeable or not, our model achieved the same accuracy (0.85) with the best qualitative model and far-outperformed other qualitative models (accuracies between 0.64 and 0.70). For further evaluation, our model, quantitative models, and the qualitative models were evaluated on a real-world central nervous system drug screening library. Our model showed an accuracy of 0.97 while the other models showed an accuracy in the range of 0.29-0.83. Consequently, our model can accurately classify BBB-permeable compounds as well as predict the absolute logBB values of drug candidates. AVAILABILITY AND IMPLEMENTATION: Web server is freely available on the web at http://ssbio.cau.ac.kr/software/logbb_pred/. The data used in this study are available to download at http://ssbio.cau.ac.kr/software/logbb_pred/dataset.zip.


Subject(s)
Blood-Brain Barrier , Brain , Blood-Brain Barrier/physiology , Biological Transport , Permeability , Central Nervous System Agents
3.
Elife ; 112022 11 01.
Article in English | MEDLINE | ID: mdl-36317872

ABSTRACT

Social deficit is a major feature of neuropsychiatric disorders, including autism spectrum disorders, schizophrenia, and attention-deficit/hyperactivity disorder, but its neural mechanisms remain unclear. Here, we examined neuronal discharge characteristics in the medial prefrontal cortex (mPFC) of IRSp53/Baiap2-mutant mice, which show social deficits, during social approach. We found a decrease in the proportion of IRSp53-mutant excitatory mPFC neurons encoding social information, but not that encoding non-social information. In addition, the firing activity of IRSp53-mutant neurons was less differential between social and non-social targets. IRSp53-mutant excitatory mPFC neurons displayed an increase in baseline neuronal firing, but decreases in the variability and dynamic range of firing as well as burst firing during social and non-social target approaches compared to wild-type controls. Treatment of memantine, an NMDA receptor antagonist that rescues social deficit in IRSp53-mutant mice, alleviates the reduced burst firing of IRSp53-mutant pyramidal mPFC neurons. These results suggest that suppressed neuronal activity dynamics and burst firing may underlie impaired cortical encoding of social information and social behaviors in IRSp53-mutant mice.


Subject(s)
Neurons , Schizophrenia , Animals , Mice , Neurons/physiology , Pyramidal Cells/metabolism , Prefrontal Cortex/physiology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
4.
Nat Commun ; 12(1): 5116, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34433814

ABSTRACT

NMDA receptor (NMDAR) and GABA neuronal dysfunctions are observed in animal models of autism spectrum disorders, but how these dysfunctions impair social cognition and behavior remains unclear. We report here that NMDARs in cortical parvalbumin (Pv)-positive interneurons cooperate with gap junctions to promote high-frequency (>80 Hz) Pv neuronal burst firing and social cognition. Shank2-/- mice, displaying improved sociability upon NMDAR activation, show impaired cortical social representation and inhibitory neuronal burst firing. Cortical Shank2-/- Pv neurons show decreased NMDAR activity, which suppresses the cooperation between NMDARs and gap junctions (GJs) for normal burst firing. Shank2-/- Pv neurons show compensatory increases in GJ activity that are not sufficient for social rescue. However, optogenetic boosting of Pv neuronal bursts, requiring GJs, rescues cortical social cognition in Shank2-/- mice, similar to the NMDAR-dependent social rescue. Therefore, NMDARs and gap junctions cooperate to promote cortical Pv neuronal bursts and social cognition.


Subject(s)
Gap Junctions/metabolism , Interneurons/physiology , Nerve Tissue Proteins/metabolism , Social Cognition , Synapses/physiology , Animals , Gap Junctions/genetics , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Parvalbumins/genetics , Parvalbumins/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Social Behavior , Synapses/genetics
5.
Front Mol Neurosci ; 14: 683196, 2021.
Article in English | MEDLINE | ID: mdl-34177464

ABSTRACT

Shank2 is an abundant postsynaptic scaffolding protein that is known to regulate excitatory synapse assembly and synaptic transmission and has been implicated in various neurodevelopmental disorders, including autism spectrum disorders (ASD). Previous studies on Shank2-mutant mice provided mechanistic insights into their autistic-like phenotypes, but it remains unclear how transcriptomic patterns are changed in brain regions of the mutant mice in age- and gene dosage-dependent manners. To this end, we performed RNA-Seq analyses of the transcripts from the prefrontal cortex (PFC) of heterozygous and homozygous Shank2-mutant mice lacking exons 6 and 7 at juvenile (week 3) and adult (week 12) stages. Juvenile heterozygous Shank2-mutant mice showed upregulation of glutamate synapse-related genes, downregulation of ribosomal and mitochondrial genes, and transcriptomic changes that are opposite to those observed in ASD (anti-ASD) such as upregulation of ASD_down (downregulated in ASD), GABA neuron-related, and oligodendrocyte-related genes. Juvenile homozygous Shank2 mice showed upregulation of chromatin-related genes and transcriptomic changes that are in line with those occurring in ASD (pro-ASD) such as downregulation of ASD_down, GABA neuron-related, and oligodendrocyte-related genes. Adult heterozygous and homozygous Shank2-mutant mice both exhibited downregulation of ribosomal and mitochondrial genes and pro-ASD transcriptomic changes. Therefore, the gene dosage- and age-dependent effects of Shank2 deletions in mice include differential transcriptomic changes across distinct functional contexts, including synapses, chromatin, ribosomes, mitochondria, GABA neurons, and oligodendrocytes.

6.
Biochem Biophys Res Commun ; 516(4): 1204-1210, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31296385

ABSTRACT

ßPix activates Rho family small GTPases, Rac1 and Cdc42 as a guanine nucleotide exchange factor. Although overexpression of ßPix in cultured neurons indicates that ßPix is involved in spine morphogenesis and synapse formation in vitro, the in vivo role of ßPix in the neuron is not well understood. Recently, we generated ßPix knockout mice that showed lethality at embryonic day 9.5. Here, we investigate the neuronal role of ßPix using ßPix heterozygous mice that are viable and fertile. ßPix heterozygous mice show decreased expression levels of ßPix proteins in various tissues including the brain. Cultured hippocampal neurons from ßPix heterozygous mice show a decrease in neurite length and complexity as well as synaptic density. Both excitatory and inhibitory synapse densities are decreased in these neurons. Golgi-staining of hippocampal tissues from the brain of these mice show reduced dendritic complexity and spine density in the hippocampal neurons. Expression levels of NMDA- and AMPA-receptor subunits and Git1 protein in hippocampal tissues are also decreased in these mice. Behaviorally, ßPix heterozygous mice exhibit impaired social interaction. Altogether, these results indicate that ßPix is required for neurite morphogenesis and synapse formation, and the reduced expression of ßPix proteins results in a defect in social behavior.


Subject(s)
Hippocampus/pathology , Neurons/pathology , Rho Guanine Nucleotide Exchange Factors/genetics , Animals , Cells, Cultured , Dendrites/metabolism , Dendrites/pathology , Gene Deletion , Heterozygote , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/metabolism , Neurites/pathology , Neurons/metabolism , Social Behavior , Synapses/metabolism , Synapses/pathology
7.
PLoS Biol ; 17(6): e2005326, 2019 06.
Article in English | MEDLINE | ID: mdl-31166939

ABSTRACT

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.


Subject(s)
Brain/embryology , Brain/growth & development , GPI-Linked Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Brain/metabolism , GPI-Linked Proteins/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Ligands , Long-Term Synaptic Depression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Netrins/metabolism , Neuronal Plasticity , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Synapses/metabolism , Synapses/physiology , Synaptic Transmission
8.
Biol Psychiatry ; 85(7): 534-543, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30466882

ABSTRACT

BACKGROUND: Autism spectrum disorder involves neurodevelopmental dysregulations that lead to visible symptoms at early stages of life. Many autism spectrum disorder-related mechanisms suggested by animal studies are supported by demonstrated improvement in autistic-like phenotypes in adult animals following experimental reversal of dysregulated mechanisms. However, whether such mechanisms also act at earlier stages to cause autistic-like phenotypes is unclear. METHODS: We used Shank2-/- mice carrying a mutation identified in human autism spectrum disorder (exons 6 and 7 deletion) and combined electrophysiological and behavioral analyses to see whether early pathophysiology at pup stages is different from late pathophysiology at juvenile and adult stages and whether correcting early pathophysiology can normalize late pathophysiology and abnormal behaviors in juvenile and adult mice. RESULTS: Early correction of a dysregulated mechanism in young mice prevents manifestation of autistic-like social behaviors in adult mice. Shank2-/- mice, known to display N-methyl-D-aspartate receptor (NMDAR) hypofunction and autistic-like behaviors at postweaning stages after postnatal day 21 (P21), show the opposite synaptic phenotype-NMDAR hyperfunction-at an earlier preweaning stage (∼P14). Moreover, this NMDAR hyperfunction at P14 rapidly shifts to NMDAR hypofunction after weaning (∼P24). Chronic suppression of the early NMDAR hyperfunction by the NMDAR antagonist memantine (P7-P21) prevents NMDAR hypofunction and autistic-like social behaviors from manifesting at later stages (∼P28 and P56). CONCLUSIONS: Early NMDAR hyperfunction leads to late NMDAR hypofunction and autistic-like social behaviors in Shank2-/- mice, and early correction of NMDAR dysfunction has the long-lasting effect of preventing autistic-like social behaviors from developing at later stages.


Subject(s)
Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/physiopathology , Behavior, Animal/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Social Behavior , Age Factors , Animals , Behavior, Animal/physiology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology
9.
Nat Neurosci ; 21(9): 1218-1228, 2018 09.
Article in English | MEDLINE | ID: mdl-30104731

ABSTRACT

Autism spectrum disorders (ASDs) are four times more common in males than in females, but the underlying mechanisms are poorly understood. We characterized sexually dimorphic changes in mice carrying a heterozygous mutation in Chd8 (Chd8+/N2373K) that was first identified in human CHD8 (Asn2373LysfsX2), a strong ASD-risk gene that encodes a chromatin remodeler. Notably, although male mutant mice displayed a range of abnormal behaviors during pup, juvenile, and adult stages, including enhanced mother-seeking ultrasonic vocalization, enhanced attachment to reunited mothers, and isolation-induced self-grooming, their female counterparts do not. This behavioral divergence was associated with sexually dimorphic changes in neuronal activity, synaptic transmission, and transcriptomic profiles. Specifically, female mice displayed suppressed baseline neuronal excitation, enhanced inhibitory synaptic transmission and neuronal firing, and increased expression of genes associated with extracellular vesicles and the extracellular matrix. Our results suggest that a human CHD8 mutation leads to sexually dimorphic changes ranging from transcription to behavior in mice.


Subject(s)
Behavior, Animal/physiology , DNA-Binding Proteins/biosynthesis , Gene Expression/physiology , Neurons/physiology , Sex Characteristics , Animals , Anxiety, Separation/genetics , Anxiety, Separation/psychology , DNA-Binding Proteins/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Female , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Object Attachment , Signal Transduction/physiology , Social Behavior , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Transcriptome , Vocalization, Animal
10.
Front Mol Neurosci ; 11: 209, 2018.
Article in English | MEDLINE | ID: mdl-29970987

ABSTRACT

Shank2 is an abundant postsynaptic scaffolding protein implicated in neurodevelopmental and psychiatric disorders, including autism spectrum disorders (ASD). Deletion of Shank2 in mice has been shown to induce social deficits, repetitive behaviors, and hyperactivity, but the identity of the cell types that contribute to these phenotypes has remained unclear. Here, we report a conditional mouse line with a Shank2 deletion restricted to parvalbumin (PV)-positive neurons (Pv-Cre;Shank2fl/fl mice). These mice display moderate hyperactivity in both novel and familiar environments and enhanced self-grooming in novel, but not familiar, environments. In contrast, they showed normal levels of social interaction, anxiety-like behavior, and learning and memory. Basal brain rhythms in Pv-Cre;Shank2fl/fl mice, measured by electroencephalography, were normal, but susceptibility to pentylenetetrazole (PTZ)-induced seizures was decreased. These results suggest that Shank2 deletion in PV-positive neurons leads to hyperactivity, enhanced self-grooming and suppressed brain excitation.

11.
J Neurosci ; 38(17): 4076-4092, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29572432

ABSTRACT

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.


Subject(s)
Anxiety/genetics , GABAergic Neurons/metabolism , Interpersonal Relations , Nerve Tissue Proteins/genetics , Synaptic Transmission , Animals , Anxiety/physiopathology , Corpus Striatum/cytology , Corpus Striatum/metabolism , Corpus Striatum/physiology , GABAergic Neurons/physiology , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Motor Activity , Nerve Tissue Proteins/metabolism , Phenotype
12.
Biol Psychiatry ; 81(10): 838-847, 2017 05 15.
Article in English | MEDLINE | ID: mdl-27450033

ABSTRACT

Imbalances between excitation and inhibition in synaptic transmission and neural circuits have been implicated in autism spectrum disorders. Excitation and inhibition imbalances are frequently observed in animal models of autism spectrum disorders, and their correction normalizes key autistic-like phenotypes in these animals. These results suggest that excitation and inhibition imbalances may contribute to the development and maintenance of autism spectrum disorders and represent an important therapeutic target.


Subject(s)
Autism Spectrum Disorder/pathology , Disease Models, Animal , Neural Inhibition/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Autism Spectrum Disorder/physiopathology , Humans
13.
J Neurosci ; 36(26): 6926-36, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27358451

ABSTRACT

UNLABELLED: Although the medial prefrontal cortex (mPFC) is known to play a crucial role in rodent social behavior, little is known about mPFC neural correlates of social behavior. In the present study, we examined single-neuron activity in the mPFC of mice performing a modified version of the three-chamber test. We found that a subset of mPFC neurons elevate discharge rates when approaching a stranger mouse but not when approaching an inanimate object or an empty chamber. Our results reveal mPFC neural activity that is correlated with social approach behavior in a widely used social-interaction paradigm. These findings might be helpful for future investigations of mPFC neural processes underlying social interaction in health and disease. SIGNIFICANCE STATEMENT: Although the prefrontal cortex is known to play a crucial role in rodent social behavior, little is known about prefrontal neural correlates of social behavior. This study shows that the activity of a subset of prefrontal neurons increases in association with social approach behavior during a three-chamber test-a widely used behavioral paradigm. Such responses might be a signature of prefrontal neural processes underlying social approach behavior.


Subject(s)
Interpersonal Relations , Neurons/physiology , Prefrontal Cortex/cytology , Action Potentials/physiology , Analysis of Variance , Animals , Choice Behavior , Male , Mice , Mice, Inbred C57BL
14.
Sci Rep ; 5: 13342, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26302668

ABSTRACT

When subjected to stress, some individuals develop maladaptive symptoms whereas others retain normal behavior. The medial prefrontal cortex (mPFC) is known to control these adaptive responses to stress. Here, we show that mPFC neurons in the left hemisphere control stress effects on social behavior. Mice made socially avoidant by the stress of chronic social defeats showed depressed neural activity in the left mPFC. Photoactivation of these neurons reversed social avoidance and restored social activity. Despite social defeats, resilient mice with normal sociability showed normal firing rates in the left mPFC; however, photoinhibition of these neurons induced social avoidance. The same photomodulation administered to the right mPFC caused no significant effects. These results explain how stressed individuals develop maladaptive behaviors through left cortical depression, as reported in mood and anxiety disorders.


Subject(s)
Dominance, Cerebral , Nerve Net/physiopathology , Prefrontal Cortex/physiopathology , Social Behavior Disorders/physiopathology , Social Behavior , Stress, Psychological/physiopathology , Adaptation, Physiological , Animals , Avoidance Learning , Behavior, Animal , Male , Mice , Mice, Inbred C57BL , Social Behavior Disorders/complications , Stress, Psychological/complications
15.
Nat Neurosci ; 18(3): 435-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25622145

ABSTRACT

Social deficits are observed in diverse psychiatric disorders, including autism spectrum disorders and schizophrenia. We found that mice lacking the excitatory synaptic signaling scaffold IRSp53 (also known as BAIAP2) showed impaired social interaction and communication. Treatment of IRSp53(-/-) mice, which display enhanced NMDA receptor (NMDAR) function in the hippocampus, with memantine, an NMDAR antagonist, or MPEP, a metabotropic glutamate receptor 5 antagonist that indirectly inhibits NMDAR function, normalized social interaction. This social rescue was accompanied by normalization of NMDAR function and plasticity in the hippocampus and neuronal firing in the medial prefrontal cortex. These results, together with the reduced NMDAR function implicated in social impairments, suggest that deviation of NMDAR function in either direction leads to social deficits and that correcting the deviation has beneficial effects.


Subject(s)
Gene Expression Regulation/physiology , Mutation/genetics , Nerve Tissue Proteins/genetics , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Social Behavior Disorders/genetics , Animals , Animals, Newborn , Case-Control Studies , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Gene Expression Regulation/drug effects , Grooming/drug effects , Grooming/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Social Behavior Disorders/drug therapy , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...