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.
Hum Mol Genet ; 27(6): 1039-1054, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29346572

ABSTRACT

Visual system development is light-experience dependent, which strongly implicates epigenetic mechanisms in light-regulated maturation. Among many epigenetic processes, genomic imprinting is an epigenetic mechanism through which monoallelic gene expression occurs in a parent-of-origin-specific manner. It is unknown if genomic imprinting contributes to visual system development. We profiled the transcriptome and imprintome during critical periods of mouse visual system development under normal- and dark-rearing conditions using B6/CAST F1 hybrid mice. We identified experience-regulated, isoform-specific and brain-region-specific imprinted genes. We also found imprinted microRNAs were predominantly clustered into the Dlk1-Dio3 imprinted locus with light experience affecting some imprinted miRNA expression. Our findings provide the first comprehensive analysis of light-experience regulation of the transcriptome and imprintome during critical periods of visual system development. Our results may contribute to therapeutic strategies for visual impairments and circadian rhythm disorders resulting from a dysfunctional imprintome.


Subject(s)
Adaptation, Ocular/genetics , Eye/embryology , Animals , DNA Methylation , Epigenesis, Genetic/genetics , Gene Expression Profiling , Genomic Imprinting , Mice , Mice, Inbred Strains/embryology , Mice, Inbred Strains/genetics , MicroRNAs/genetics , Ocular Physiological Phenomena/genetics , Spatio-Temporal Analysis , Superior Colliculi/embryology , Transcriptome
2.
PLoS One ; 11(9): e0163663, 2016.
Article in English | MEDLINE | ID: mdl-27662371

ABSTRACT

Genomic imprinting is an epigenetic mechanism causing monoallelic expression in a parent-of-origin-specific manner. Disruption of imprinted genes causes various neurological and psychiatric disorders. However, the role of imprinted genes in the brain is largely unknown. Different cell types within distinct brain regions can influence the genomic imprinting status, but imprinted genes in single cell types within distinct brain regions have not been characterized on a genome-wide scale. To address this critical question, we used a multi-stage approach, which combined genetically engineered mice with fluorescence-based laser capture microdissection (LCM) to capture excitatory neurons, inhibitory neurons and astrocytes as single cells in layer 2/3 of mouse visual cortex. RNA sequencing determined parental expression patterns on a genome-wide scale in the captured cells within specific brain regions. The expression level of cell-type-specific genes for excitatory neurons (13 genes), inhibitory neurons (16 genes) and astrocytes (20 genes) confirmed the LCM-captured cells maintained their cellular identities. The parent-of-origin-specific expression pattern of imprinted genes, including maternally expressed Meg3 and paternally expressed Peg3, provided evidence that the status of known imprinted genes was also maintained. Although our platform remains to be improved, our findings demonstrate the parental expression pattern can be analysed not only at the level of a single cell type but also at the level of specific cortical layers. Our approach has the potential to reveal novel regulatory modules associated with plasticity through genomic imprinting mechanisms in different cell types, not only in the visual cortex but also in other brain regions.

3.
Sci Rep ; 5: 17383, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26619789

ABSTRACT

RBFOX3 mutations are linked to epilepsy and cognitive impairments, but the underlying pathophysiology of these disorders is poorly understood. Here we report replication of human symptoms in a mouse model with disrupted Rbfox3. Rbfox3 knockout mice displayed increased seizure susceptibility and decreased anxiety-related behaviors. Focusing on hippocampal phenotypes, we found Rbfox3 knockout mice showed increased expression of plasticity genes Egr4 and Arc, and the synaptic transmission and plasticity were defective in the mutant perforant pathway. The mutant dentate granules cells exhibited an increased frequency, but normal amplitude, of excitatory synaptic events, and this change was associated with an increase in the neurotransmitter release probability and dendritic spine density. Together, our results demonstrate anatomical and functional abnormality in Rbfox3 knockout mice, and may provide mechanistic insights for RBFOX3-related human brain disorders.


Subject(s)
Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Synaptic Transmission , Animals , Anxiety/genetics , Anxiety/metabolism , Anxiety/pathology , Anxiety/physiopathology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , DNA-Binding Proteins , Disease Models, Animal , Early Growth Response Transcription Factors/biosynthesis , Early Growth Response Transcription Factors/genetics , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/physiopathology , Hippocampus/physiopathology , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...