Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Neuron ; 110(10): 1728-1741.e7, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35294900

ABSTRACT

The architecture whereby activity across many brain regions integrates to encode individual appetitive social behavior remains unknown. Here we measure electrical activity from eight brain regions as mice engage in a social preference assay. We then use machine learning to discover a network that encodes the extent to which individual mice engage another mouse. This network is organized by theta oscillations leading from prelimbic cortex and amygdala that converge on the ventral tegmental area. Network activity is synchronized with cellular firing, and frequency-specific activation of a circuit within this network increases social behavior. Finally, the network generalizes, on a mouse-by-mouse basis, to encode individual differences in social behavior in healthy animals but fails to encode individual behavior in a 'high confidence' genetic model of autism. Thus, our findings reveal the architecture whereby the brain integrates distributed activity across timescales to encode an appetitive brain state underlying individual differences in social behavior.


Subject(s)
Appetitive Behavior , Brain , Amygdala , Animals , Brain/physiology , Mice , Social Behavior , Ventral Tegmental Area
2.
Cereb Cortex ; 31(4): 1998-2012, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33230530

ABSTRACT

Emerging evidence suggests that epigenetic mechanisms regulate aberrant gene transcription in stress-associated mental disorders. However, it remains to be elucidated about the role of DNA methylation and its catalyzing enzymes, DNA methyltransferases (DNMTs), in this process. Here, we found that male rats exposed to chronic (2-week) unpredictable stress exhibited a substantial reduction of Dnmt3a after stress cessation in the prefrontal cortex (PFC), a key target region of stress. Treatment of unstressed control rats with DNMT inhibitors recapitulated the effect of chronic unpredictable stress on decreased AMPAR expression and function in PFC. In contrast, overexpression of Dnmt3a in PFC of stressed animals prevented the loss of glutamatergic responses. Moreover, the stress-induced behavioral abnormalities, including the impaired recognition memory, heightened aggression, and hyperlocomotion, were partially attenuated by Dnmt3a expression in PFC of stressed animals. Finally, we found that there were genome-wide DNA methylation changes and transcriptome alterations in PFC of stressed rats, both of which were enriched at several neural pathways, including glutamatergic synapse and microtubule-associated protein kinase signaling. These results have therefore recognized the potential role of DNA epigenetic modification in stress-induced disturbance of synaptic functions and cognitive and emotional processes.


Subject(s)
DNA Methyltransferase 3A/metabolism , Locomotion/physiology , Prefrontal Cortex/enzymology , Stress, Psychological/enzymology , Stress, Psychological/psychology , Synapses/enzymology , Animals , Chronic Disease , DNA Methyltransferase 3A/antagonists & inhibitors , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Locomotion/drug effects , Male , Mice , Phthalimides/pharmacology , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
3.
JCI Insight ; 3(23)2018 12 06.
Article in English | MEDLINE | ID: mdl-30518695

ABSTRACT

OXTR modulates a variety of behaviors in mammals, including social memory and recognition. Genetic and epigenetic dysregulation of OXTR has been suggested to be implicated in neuropsychiatric disorders, including autism spectrum disorder (ASD). While the involvement of DNA methylation is suggested, the mechanism underlying epigenetic regulation of OXTR is largely unknown. This has hampered the experimental design and interpretation of the results of epigenetic studies of OXTR in neuropsychiatric disorders. From the generation and characterization of a new line of Tet1 mutant mice - by deleting the largest coding exon 4 (Tet1Δe4) - we discovered for the first time to our knowledge that Oxtr has an array of mRNA isoforms and a complex transcriptional regulation. Select isoforms of Oxtr are significantly reduced in the brain of Tet1Δe4-/- mice. Accordingly, CpG islands of Oxtr are hypermethylated during early development and persist into adulthood. Consistent with the reduced express of OXTR, Tet1Δe4-/- mice display impaired maternal care, social behavior, and synaptic responses to oxytocin stimulation. Our findings elucidate a mechanism mediated by TET1 protein in regulating Oxtr expression by preventing DNA hypermethylation of Oxtr. The discovery of epigenetic dysregulation of Oxtr in TET1-deficient mouse brain supports the necessity of a reassessment of existing findings and a value of future studies of OXTR in neuropsychiatric disorders.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Proto-Oncogene Proteins/genetics , Receptors, Oxytocin/genetics , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Behavior, Animal/physiology , Brain/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Exons , Female , Histones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Proto-Oncogene Proteins/metabolism , RNA Isoforms/metabolism , Receptors, Oxytocin/metabolism , Social Behavior , Transcriptome
4.
Neuropsychopharmacology ; 43(8): 1779-1788, 2018 07.
Article in English | MEDLINE | ID: mdl-29760409

ABSTRACT

Autism is a neurodevelopmental disorder characterized by social deficits and repetitive behaviors. Genetic screening has identified synaptic, transcriptional, and chromatin genes disrupted in autistic patients. Haploinsufficiency of Shank3, which encodes a scaffold protein at glutamatergic synapses, is causally linked to autism. Using a Shank3-deficient mouse model that exhibits prominent autism-like phenotypes, we have found that histone acetylation in the prefrontal cortex (PFC) is abnormally low, which can be reversed by MS-275 (also known as Entinostat, SNDX-275), a class I histone deacetylase (HDAC) inhibitor that is selectively potent in PFC. A brief (3-day) treatment with MS-275 (i.p.) led to the sustained (11 days) rescue of autistic social preference deficits in Shank3-deficient mice, without altering locomotion, motor coordination, anxiety, or the increased grooming. MS-275 treatment also rescued the diminished NMDAR surface expression and NMDAR function induced by Shank3 deficiency. Moreover, F-actin at synapses was restored and the transcription of actin regulators was elevated by MS-275 treatment of Shank3-deficient mice, which may contribute to the recovery of actin-based NMDAR synaptic delivery. Taken together, these results suggest that MS-275 treatment could normalize the aberrant epigenetic regulation of genes, leading to the amelioration of synaptic and social deficits associated with autism.


Subject(s)
Autistic Disorder/drug therapy , Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Pyridines/pharmacology , Social Behavior , Synapses/drug effects , Actins/metabolism , Animals , Autistic Disorder/physiopathology , Brain/drug effects , Brain/physiopathology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Disease Models, Animal , Epigenesis, Genetic/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Rats , Synapses/physiology
5.
Transl Psychiatry ; 8(1): 94, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29700290

ABSTRACT

We previously reported a new line of Shank3 mutant mice which led to a complete loss of Shank3 by deleting exons 4-22 (Δe4-22) globally. Δe4-22 mice display robust ASD-like behaviors including impaired social interaction and communication, increased stereotypical behavior and excessive grooming, and a profound deficit in instrumental learning. However, the anatomical and neural circuitry underlying these behaviors are unknown. We generated mice with Shank3 selectively deleted in forebrain, striatum, and striatal D1 and D2 cells. These mice were used to interrogate the circuit/brain-region and cell-type specific role of Shank3 in the expression of autism-related behaviors. Whole-cell patch recording and biochemical analyses were used to study the synaptic function and molecular changes in specific brain regions. We found perseverative exploratory behaviors in mice with deletion of Shank3 in striatal inhibitory neurons. Conversely, self-grooming induced lesions were observed in mice with deletion of Shank3 in excitatory neurons of forebrain. However, social, communicative, and instrumental learning behaviors were largely unaffected in these mice, unlike what is seen in global Δe4-22 mice. We discovered unique patterns of change for the biochemical and electrophysiological findings in respective brain regions that reflect the complex nature of transcriptional regulation of Shank3. Reductions in Homer1b/c and membrane hyper-excitability were observed in striatal loss of Shank3. By comparison, Shank3 deletion in hippocampal neurons resulted in increased NMDAR-currents and GluN2B-containing NMDARs. These results together suggest that Shank3 may differentially regulate neural circuits that control behavior. Our study supports a dissociation of Shank3 functions in cortical and striatal neurons in ASD-related behaviors, and it illustrates the complexity of neural circuit mechanisms underlying these behaviors.


Subject(s)
Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/psychology , Corpus Striatum/physiopathology , Nerve Tissue Proteins/physiology , Prosencephalon/physiopathology , Animals , Behavior, Animal , Corpus Striatum/metabolism , Disease Models, Animal , Excitatory Postsynaptic Potentials , Hippocampus/metabolism , Hippocampus/physiopathology , Homer Scaffolding Proteins/metabolism , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/genetics , Neurons/physiology , Phenotype , Prosencephalon/metabolism , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Social Behavior , Synapses/metabolism
6.
Am J Med Genet B Neuropsychiatr Genet ; 177(4): 426-433, 2018 06.
Article in English | MEDLINE | ID: mdl-29704315

ABSTRACT

Genetic mutations in genes encoding proteins involved in epigenetic machinery have been reported in individuals with autism spectrum disorder (ASD), intellectual disability, congenital heart disease, and other disorders. H1 histone linker protein, the basic component in nucleosome packaging and chromatin organization, has not been implicated in human disease until recently. We report a de novo deleterious mutation of histone cluster 1 H1 family member e (HIST1H1E; c.435dupC; p.Thr146Hisfs*50), encoding H1 histone linker protein H1.4, in a 10-year-old boy with autism and intellectual disability diagnosed through clinical whole exome sequencing. The c.435dupC at the 3' end of the mRNA leads to a frameshift and truncation of the positive charge in the carboxy-terminus of the protein. An expression study demonstrates the mutation leads to reduced protein expression, supporting haploinsufficiency of HIST1H1E protein and loss of function as an underlying mechanism of dysfunction in the brain. Taken together with other recent cases with mutations of HIST1H1E in intellectual disability, the evidence supporting the link to causality in disease is strong. Our finding implicates the deficiency of H1 linker histone protein in autism. The systematic review of candidate genes implicated in ASD revealed that 42 of 215 (19.5%) genes are directly involved in epigenetic regulations and the majority of these genes belong to histone writers, readers, and erasers. While the mechanism of how haploinsufficiency of HIST1H1E causes autism is entirely unknown, our report underscores the importance of further study of the function of this protein and other histone linker proteins in brain development.


Subject(s)
Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Histones/genetics , Autism Spectrum Disorder/physiopathology , Child , Epigenesis, Genetic/genetics , Epigenomics/methods , Genetic Predisposition to Disease , Humans , Male , Mutation , Exome Sequencing
7.
JCI Insight ; 2(20)2017 10 19.
Article in English | MEDLINE | ID: mdl-29046483

ABSTRACT

Genetic defects in the synaptic scaffolding protein gene, SHANK2, are linked to a variety of neuropsychiatric disorders, including autism spectrum disorders, schizophrenia, intellectual disability, and bipolar disorder, but the molecular mechanisms underlying the pleotropic effects of SHANK2 mutations are poorly understood. We generated and characterized a line of Shank2 mutant mice by deleting exon 24 (Δe24). Shank2Δe24-/- mice engage in significantly increased locomotor activity, display abnormal reward-seeking behavior, are anhedonic, have perturbations in circadian rhythms, and show deficits in social and cognitive behaviors. While these phenotypes recapitulate the pleotropic behaviors associated with human SHANK2-related disorders, major behavioral features in these mice are reminiscent of bipolar disorder. For instance, their hyperactivity was augmented with amphetamine but was normalized with the mood stabilizers lithium and valproate. Shank2 deficiency limited to the forebrain recapitulated the bipolar mania phenotype. The composition and functions of NMDA and AMPA receptors were altered at Shank2-deficient synapses, hinting toward the mechanism underlying these behavioral abnormalities. Human genetic findings support construct validity, and the behavioral features in Shank2 Δe24 mice support face and predictive validities of this model for bipolar mania. Further genetic studies to understand the contribution of SHANK2 deficiencies in bipolar disorder are warranted.


Subject(s)
Bipolar Disorder/genetics , Motor Activity/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Amphetamine/pharmacology , Anhedonia , Animals , Antimanic Agents/therapeutic use , Behavior, Animal , Central Nervous System Stimulants/pharmacology , Chronobiology Disorders/drug therapy , Chronobiology Disorders/genetics , Cognitive Dysfunction/genetics , Female , Hippocampus/metabolism , Hippocampus/ultrastructure , Lithium Compounds/therapeutic use , Male , Mice , Mice, Knockout , Motor Activity/drug effects , N-Methylaspartate/metabolism , Phenotype , Prosencephalon/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Social Behavior Disorders/genetics , Synapses/metabolism
8.
Nat Commun ; 7: 11459, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27161151

ABSTRACT

Human neuroimaging studies suggest that aberrant neural connectivity underlies behavioural deficits in autism spectrum disorders (ASDs), but the molecular and neural circuit mechanisms underlying ASDs remain elusive. Here, we describe a complete knockout mouse model of the autism-associated Shank3 gene, with a deletion of exons 4-22 (Δe4-22). Both mGluR5-Homer scaffolds and mGluR5-mediated signalling are selectively altered in striatal neurons. These changes are associated with perturbed function at striatal synapses, abnormal brain morphology, aberrant structural connectivity and ASD-like behaviour. In vivo recording reveals that the cortico-striatal-thalamic circuit is tonically hyperactive in mutants, but becomes hypoactive during social behaviour. Manipulation of mGluR5 activity attenuates excessive grooming and instrumental learning differentially, and rescues impaired striatal synaptic plasticity in Δe4-22(-/-) mice. These findings show that deficiency of Shank3 can impair mGluR5-Homer scaffolding, resulting in cortico-striatal circuit abnormalities that underlie deficits in learning and ASD-like behaviours. These data suggest causal links between genetic, molecular, and circuit mechanisms underlying the pathophysiology of ASDs.


Subject(s)
Autism Spectrum Disorder/physiopathology , Cerebral Cortex/physiopathology , Corpus Striatum/physiopathology , Homer Scaffolding Proteins/metabolism , Nerve Tissue Proteins/deficiency , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Behavior, Animal , Cerebral Cortex/pathology , Corpus Striatum/pathology , Female , Humans , Long-Term Synaptic Depression , Male , Mice , Mice, Knockout , Microfilament Proteins , Models, Neurological , Nerve Net/pathology , Nerve Net/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Sequence Deletion , Social Behavior
9.
J Neurosci ; 36(7): 2119-30, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26888924

ABSTRACT

Stress and the major stress hormone corticosterone induce profound influences in the brain. Altered histone modification and transcriptional dysfunction have been implicated in stress-related mental disorders. We previously found that repeated stress caused an impairment of prefrontal cortex (PFC)-mediated cognitive functions by increasing the ubiquitination and degradation of AMPA-type glutamate receptors via a mechanism depending on the E3 ubiquitin ligase Nedd4. Here, we demonstrated that in PFC of repeatedly stressed rats, active glucocorticoid receptor had the increased binding to the glucocorticoid response element of histone deacetylase 2 (HDAC2) promoter, resulting in the upregulation of HDAC2. Inhibition or knock-down of HDAC2 blocked the stress-induced impairment of synaptic transmission, AMPAR expression, and recognition memory. Furthermore, we found that, in stressed animals, the HDAC2-dependent downregulation of histone methyltransferase Ehmt2 (G9a) led to the loss of repressive histone methylation at the Nedd4-1 promoter and the transcriptional activation of Nedd4. These results have provided an epigenetic mechanism and a potential treatment strategy for the detrimental effects of chronic stress. SIGNIFICANCE STATEMENT: Prolonged stress exposure can induce altered histone modification and transcriptional dysfunction, which may underlie the profound influence of stress in regulating brain functions. We report an important finding about the epigenetic mechanism controlling the detrimental effects of repeated stress on synaptic transmission and cognitive function. First, it has revealed the stress-induced alteration of key epigenetic regulators HDAC2 and Ehmt2, which determines the synaptic and behavioral effects of repeated stress. Second, it has uncovered the stress-induced histone modification of the target gene Nedd4, an E3 ligase that is critically involved in the ubiquitination and degradation of AMPA receptors and cognition. Third, it has provided the epigenetic approach, HDAC2 inhibition or knock-down, to rescue synaptic and cognitive functions in stressed animals.


Subject(s)
Cognition Disorders/etiology , Endosomal Sorting Complexes Required for Transport , Histones/metabolism , Receptors, AMPA , Stress, Psychological/complications , Ubiquitin-Protein Ligases , Animals , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Gene Knockdown Techniques , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histones/chemistry , Male , Nedd4 Ubiquitin Protein Ligases , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Recognition, Psychology/drug effects , Stress, Psychological/metabolism , Synaptic Transmission/drug effects , Ubiquitination/genetics
10.
Cell Rep ; 11(9): 1400-1413, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26027926

ABSTRACT

Haploinsufficiency of the Shank3 gene, which encodes a scaffolding protein at glutamatergic synapses, is a highly prevalent and penetrant risk factor for autism. Using combined behavioral, electrophysiological, biochemical, imaging, and molecular approaches, we find that Shank3-deficient mice exhibit autism-like social deficits and repetitive behaviors, as well as the significantly diminished NMDA receptor (NMDAR) synaptic function and synaptic distribution in prefrontal cortex. Concomitantly, Shank3-deficient mice have a marked loss of cortical actin filaments, which is associated with the reduced Rac1/PAK activity and increased activity of cofilin, the major actin depolymerizing factor. The social deficits and NMDAR hypofunction are rescued by inhibiting cofilin or activating Rac1 in Shank3-deficient mice and are induced by inhibiting PAK or Rac1 in wild-type mice. These results indicate that the aberrant regulation of synaptic actin filaments and loss of synaptic NMDARs contribute to the manifestation of autism-like phenotypes. Thus, targeting actin regulators provides a strategy for autism treatment.


Subject(s)
Actin Depolymerizing Factors/metabolism , Autistic Disorder/metabolism , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/physiopathology , Animals , Autistic Disorder/genetics , Autistic Disorder/physiopathology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuropeptides/metabolism , Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
11.
Biol Psychiatry ; 76(12): 953-62, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-24832867

ABSTRACT

BACKGROUND: Methylphenidate (MPH), a psychostimulant drug used to treat attention-deficit/hyperactivity disorder, produces the effects of increasing alertness and improving attention. However, misuse of MPH has been associated with an increased risk of aggression and psychosis. We sought to determine the molecular mechanism underlying the complex actions of MPH. METHODS: Adolescent (4-week-old) rats were given one injection of MPH at different doses. The impact of MPH on glutamatergic signaling in pyramidal neurons of prefrontal cortex was measured. Behavioral changes induced by MPH were also examined in parallel. RESULTS: Administration of low-dose (.5 mg/kg) MPH selectively potentiated N-methyl-D-aspartate receptor (NMDAR)-mediated excitatory postsynaptic currents (EPSCs) via adrenergic receptor activation, whereas high-dose (10 mg/kg) MPH suppressed both NMDAR-mediated and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor-mediated EPSCs. The dual effects of MPH on EPSCs were associated with bidirectional changes in the surface level of glutamate receptor subunits. Behavioral tests also indicated that low-dose MPH facilitated prefrontal cortex-mediated temporal order recognition memory and attention. Animals injected with high-dose MPH exhibited significantly elevated locomotive activity. Inhibiting the function of synaptosomal-associated protein 25, a key SNARE protein involved in NMDAR exocytosis, blocked the increase of NMDAR-mediated EPSCs by low-dose MPH. In animals exposed to repeated stress, administration of low-dose MPH effectively restored NMDAR function and temporal order recognition memory via a mechanism dependent on synaptosomal-associated protein 25. CONCLUSIONS: These results provide a potential mechanism underlying the cognitive-enhancing effects of low-dose MPH as well as the psychosis-inducing effects of high-dose MPH.


Subject(s)
Attention/drug effects , Central Nervous System Stimulants/pharmacology , Methylphenidate/pharmacology , Motor Activity/drug effects , Receptors, Glutamate/metabolism , Recognition, Psychology/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Analysis of Variance , Animals , Benzazepines/pharmacology , Biophysics , Discrimination, Psychological/drug effects , Dopamine Antagonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Gene Expression Regulation/drug effects , In Vitro Techniques , Male , Maprotiline/pharmacology , Neurons/drug effects , Peptides/pharmacology , Piperazines/pharmacology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , RNA, Messenger/metabolism , Rats , Receptors, Glutamate/genetics , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/metabolism , Yohimbine/pharmacology
12.
J Neurosci ; 33(40): 15767-78, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24089484

ABSTRACT

Shank3, which encodes a scaffolding protein at glutamatergic synapses, is a genetic risk factor for autism. In this study, we examined the impact of Shank3 deficiency on the NMDA-type glutamate receptor, a key player in cognition and mental illnesses. We found that knockdown of Shank3 with a small interfering RNA (siRNA) caused a significant reduction of NMDAR-mediated ionic or synaptic current, as well as the surface expression of NR1 subunits, in rat cortical cultures. The effect of Shank3 siRNA on NMDAR currents was blocked by an actin stabilizer, and was occluded by an actin destabilizer, suggesting the involvement of actin cytoskeleton. Since actin dynamics is regulated by the GTPase Rac1 and downstream effector p21-activated kinase (PAK), we further examined Shank3 regulation of NMDARs when Rac1 or PAK was manipulated. We found that the reducing effect of Shank3 siRNA on NMDAR currents was mimicked and occluded by specific inhibitors for Rac1 or PAK, and was blocked by constitutively active Rac1 or PAK. Immunocytochemical data showed a strong reduction of F-actin clusters after Shank3 knockdown, which was occluded by a PAK inhibitor. Inhibiting cofilin, the primary downstream target of PAK and a major actin depolymerizing factor, prevented Shank3 siRNA from reducing NMDAR currents and F-actin clusters. Together, these results suggest that Shank3 deficiency induces NMDAR hypofunction by interfering with the Rac1/PAK/cofilin/actin signaling, leading to the loss of NMDAR membrane delivery or stability. It provides a potential mechanism for the role of Shank3 in cognitive deficit in autism.


Subject(s)
Actins/metabolism , Cerebral Cortex/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , HEK293 Cells , Humans , Nerve Tissue Proteins/genetics , Neurons/cytology , RNA Interference , RNA, Small Interfering , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Synapses/metabolism , Synaptic Transmission/physiology , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
13.
J Physiol ; 591(16): 3935-47, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23774277

ABSTRACT

The group II metabotropic glutamate receptors (group II mGluRs) have emerged as the new drug targets for the treatment of mental disorders like schizophrenia. To understand the potential mechanisms underlying the antipsychotic effects of group II mGluRs, we examined their impact on NMDA receptors (NMDARs), since NMDAR hypofunction has been implicated in schizophrenia. The activation of group II mGluRs caused a significant enhancement of NMDAR currents in cortical pyramidal neurons, which was associated with increased NMDAR surface expression and synaptic localization. We further examined whether these effects of group II mGluRs are through the regulation of NMDAR exocytosis via SNARE proteins, a family of proteins involved in vesicle fusion. We found that the enhancing effect of APDC, a selective agonist of group II mGluRs, on NMDAR currents was abolished when botulinum toxin was delivered into the recorded neurons to disrupt the SNARE complex. Inhibiting the function of two key SNARE proteins, SNAP-25 and syntaxin 4, also eliminated the effect of APDC on NMDAR currents. Moreover, the application of APDC increased the activity of Rab4, a small Rab GTPase mediating fast recycling from early endosomes to the plasma membrane, and enhanced the interaction between syntaxin 4 and Rab4. Knockdown of Rab4 or expression of dominant-negative Rab4 attenuated the effect of APDC on NMDAR currents. Taken together, these results have identified key molecules involved in the group II mGluR-induced potentiation of NMDAR exocytosis and function.


Subject(s)
Frontal Lobe/physiology , Pyramidal Cells/physiology , Receptors, Metabotropic Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/physiology , SNARE Proteins/physiology , Animals , Frontal Lobe/embryology , In Vitro Techniques , Rats , Synaptosomal-Associated Protein 25/physiology
14.
J Biol Chem ; 286(39): 33719-28, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21832090

ABSTRACT

Emerging evidence from studies of Huntington disease (HD) pathophysiology suggests that huntingtin (htt) and its associated protein HAP1 participate in intracellular trafficking and synaptic function. However, it is largely unknown whether AMPA receptor trafficking, which is crucial for controlling the efficacy of synaptic excitation, is affected by the mutant huntingtin with polyglutamine expansion (polyQ-htt). In this study, we found that expressing polyQ-htt in neuronal cultures significantly decreased the amplitude and frequency of AMPAR-mediated miniature excitatory postsynaptic current (mEPSC), while expressing wild-type huntingtin (WT-htt) increased mEPSC. AMPAR-mediated synaptic transmission was also impaired in a transgenic mouse model of HD expressing polyQ-htt. The effect of polyQ-htt on mEPSC was mimicked by knockdown of HAP1 and occluded by the dominant negative HAP1. Moreover, we found that huntingtin affected mESPC via a mechanism depending on the kinesin motor protein, KIF5, which controls the transport of GluR2-containing AMPARs along microtubules in dendrites. The GluR2/KIF5/HAP1 complex was disrupted and dissociated from microtubules in the HD mouse model. Together, these data suggest that AMPAR trafficking and function is impaired by mutant huntingtin, presumably due to the interference of KIF5-mediated microtubule-based transport of AMPA receptors. The diminished strength of glutamatergic transmission could contribute to the deficits in movement control and cognitive processes in HD conditions.


Subject(s)
Mutation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Receptors, AMPA/metabolism , Synaptic Potentials , Synaptic Transmission , Animals , HEK293 Cells , Humans , Huntingtin Protein , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , Peptides/genetics , Peptides/metabolism , Protein Transport/genetics , Rats , Receptors, AMPA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...