Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 35(13): 5097-108, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25834037

ABSTRACT

Neuronal histone H3-lysine 4 methylation landscapes are defined by sharp peaks at gene promoters and other cis-regulatory sequences, but molecular and cellular phenotypes after neuron-specific deletion of H3K4 methyl-regulators remain largely unexplored. We report that neuronal ablation of the H3K4-specific methyltransferase, Kmt2a/Mixed-lineage leukemia 1 (Mll1), in mouse postnatal forebrain and adult prefrontal cortex (PFC) is associated with increased anxiety and robust cognitive deficits without locomotor dysfunction. In contrast, only mild behavioral phenotypes were observed after ablation of the Mll1 ortholog Kmt2b/Mll2 in PFC. Impaired working memory after Kmt2a/Mll1 ablation in PFC neurons was associated with loss of training-induced transient waves of Arc immediate early gene expression critical for synaptic plasticity. Medial prefrontal layer V pyramidal neurons, a major output relay of the cortex, demonstrated severely impaired synaptic facilitation and temporal summation, two forms of short-term plasticity essential for working memory. Chromatin immunoprecipitation followed by deep sequencing in Mll1-deficient cortical neurons revealed downregulated expression and loss of the transcriptional mark, trimethyl-H3K4, at <50 loci, including the homeodomain transcription factor Meis2. Small RNA-mediated Meis2 knockdown in PFC was associated with working memory defects similar to those elicited by Mll1 deletion. Therefore, mature prefrontal neurons critically depend on maintenance of Mll1-regulated H3K4 methylation at a subset of genes with an essential role in cognition and emotion.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Memory, Short-Term/physiology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Animals , Behavior, Animal/physiology , Cytoskeletal Proteins/metabolism , Gene Expression , Gene Knockdown Techniques , Homeodomain Proteins/drug effects , Homeodomain Proteins/genetics , Male , Methylation , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Prosencephalon/physiology , Pyramidal Cells/physiology
2.
Arch Gen Psychiatry ; 69(3): 314-24, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22065254

ABSTRACT

CONTEXT: Neuronal dysfunction in cerebral cortex and other brain regions could contribute to the cognitive and behavioral defects in autism. OBJECTIVE: To characterize epigenetic signatures of autism in prefrontal cortex neurons. DESIGN: We performed fluorescence-activated sorting and separation of neuronal and nonneuronal nuclei from postmortem prefrontal cortex, digested the chromatin with micrococcal nuclease, and deeply sequenced the DNA from the mononucleosomes with trimethylated H3K4 (H3K4me3), a histone mark associated with transcriptional regulation. Approximately 15 billion base pairs of H3K4me3-enriched sequences were collected from 32 brains. SETTING: Academic medical center. PARTICIPANTS: A total of 16 subjects diagnosed as having autism and 16 control subjects ranging in age from 0.5 to 70 years. MAIN OUTCOME MEASURES: Identification of genomic loci showing autism-associated H3K4me3 changes in prefrontal cortex neurons. RESULTS: Subjects with autism showed no evidence for generalized disruption of the developmentally regulated remodeling of the H3K4me3 landscape that defines normal prefrontal cortex neurons in early infancy. However, excess spreading of H3K4me3 from the transcription start sites into downstream gene bodies and upstream promoters was observed specifically in neuronal chromatin from 4 of 16 autism cases but not in controls. Variable subsets of autism cases exhibit altered H3K4me3 peaks at numerous genes regulating neuronal connectivity, social behaviors, and cognition, often in conjunction with altered expression of the corresponding transcripts. Autism-associated H3K4me3 peaks were significantly enriched in genes and loci implicated in neurodevelopmental diseases. CONCLUSIONS: Prefrontal cortex neurons from subjects with autism show changes in chromatin structures at hundreds of loci genome-wide, revealing considerable overlap between genetic and epigenetic risk maps of developmental brain disorders.


Subject(s)
Autistic Disorder/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Histones/metabolism , Prefrontal Cortex/metabolism , Adolescent , Adult , Aged , Autistic Disorder/etiology , Autistic Disorder/metabolism , Case-Control Studies , Child , Child, Preschool , Chromatin/genetics , Chromatin/metabolism , DNA Fingerprinting , Female , Flow Cytometry , Histones/genetics , Humans , Male , Middle Aged , Models, Genetic , Neurons/metabolism , Prefrontal Cortex/cytology , Risk Factors , Young Adult
3.
J Neurosci ; 30(21): 7152-67, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20505083

ABSTRACT

Histone methyltransferases specific for the histone H3-lysine 9 residue, including Setdb1 (Set domain, bifurcated 1)/Eset/Kmt1e are associated with repressive chromatin remodeling and expressed in adult brain, but potential effects on neuronal function and behavior remain unexplored. Here, we report that transgenic mice with increased Setdb1 expression in adult forebrain neurons show antidepressant-like phenotypes in behavioral paradigms for anhedonia, despair, and learned helplessness. Chromatin immunoprecipitation in conjunction with DNA tiling arrays (ChIP-chip) revealed that genomic occupancies of neuronal Setdb1 are limited to <1% of annotated genes, which include the NMDA receptor subunit NR2B/Grin2B and other ionotropic glutamate receptor genes. Chromatin conformation capture and Setdb1-ChIP revealed a loop formation tethering the NR2B/Grin2b promoter to the Setdb1 target site positioned 30 kb downstream of the transcription start site. In hippocampus and ventral striatum, two key structures in the neuronal circuitry regulating mood-related behaviors, Setdb1-mediated repressive histone methylation at NR2B/Grin2b was associated with decreased NR2B expression and EPSP insensitivity to pharmacological blockade of NR2B, and accelerated NMDA receptor desensitization consistent with a shift in NR2A/B subunit ratios. In wild-type mice, systemic treatment with the NR2B antagonist, Ro25-6981 [R-(R,S)-alpha-(4-hydroxyphenyl)-beta-methyl-4-(phenylmethyl)-1-piperidine propranol], and hippocampal small interfering RNA-mediated NR2B/Grin2b knockdown resulted in behavioral changes similar to those elicited by the Setdb1 transgene. Together, these findings point to a role for neuronal Setdb1 in the regulation of affective and motivational behaviors through repressive chromatin remodeling at a select set of target genes, resulting in altered NMDA receptor subunit composition and other molecular adaptations.


Subject(s)
Affect/physiology , Behavior, Animal/physiology , Gene Expression Regulation/physiology , Protein Methyltransferases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Adaptation, Ocular/drug effects , Adaptation, Ocular/genetics , Affect/drug effects , Age Factors , Animals , Animals, Newborn , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Chromatin/metabolism , Chromatin Immunoprecipitation/methods , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Electroshock/adverse effects , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Fear/drug effects , Fear/physiology , Food Preferences/drug effects , Food Preferences/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Histone-Lysine N-Methyltransferase , Humans , Immobility Response, Tonic/drug effects , Immobility Response, Tonic/physiology , Maze Learning/drug effects , Maze Learning/physiology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Protein Methyltransferases/genetics , RNA, Small Interfering/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Sucrose/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...