Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Syst Biol ; 18(8): e10473, 2022 08.
Article in English | MEDLINE | ID: mdl-35996956

ABSTRACT

Neuronal stimulation induced by the brain-derived neurotrophic factor (BDNF) triggers gene expression, which is crucial for neuronal survival, differentiation, synaptic plasticity, memory formation, and neurocognitive health. However, its role in chromatin regulation is unclear. Here, using temporal profiling of chromatin accessibility and transcription in mouse primary cortical neurons upon either BDNF stimulation or depolarization (KCl), we identify features that define BDNF-specific chromatin-to-gene expression programs. Enhancer activation is an early event in the regulatory control of BDNF-treated neurons, where the bZIP motif-binding Fos protein pioneered chromatin opening and cooperated with co-regulatory transcription factors (Homeobox, EGRs, and CTCF) to induce transcription. Deleting cis-regulatory sequences affect BDNF-mediated Arc expression, a regulator of synaptic plasticity. BDNF-induced accessible regions are linked to preferential exon usage by neurodevelopmental disorder-related genes and the heritability of neuronal complex traits, which were validated in human iPSC-derived neurons. Thus, we provide a comprehensive view of BDNF-mediated genome regulatory features using comparative genomic approaches to dissect mammalian neuronal stimulation.


Subject(s)
Brain-Derived Neurotrophic Factor , Chromatin , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Chromatin/genetics , Chromatin/metabolism , Humans , Mammals/genetics , Mice , Neurons/metabolism , Transcription Factors/metabolism
2.
Nat Commun ; 9(1): 689, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449567

ABSTRACT

A better understanding of proteostasis in health and disease requires robust methods to determine protein half-lives. Here we improve the precision and accuracy of peptide ion intensity-based quantification, enabling more accurate protein turnover determination in non-dividing cells by dynamic SILAC-based proteomics. This approach allows exact determination of protein half-lives ranging from 10 to >1000 h. We identified 4000-6000 proteins in several non-dividing cell types, corresponding to 9699 unique protein identifications over the entire data set. We observed similar protein half-lives in B-cells, natural killer cells and monocytes, whereas hepatocytes and mouse embryonic neurons show substantial differences. Our data set extends and statistically validates the previous observation that subunits of protein complexes tend to have coherent turnover. Moreover, analysis of different proteasome and nuclear pore complex assemblies suggests that their turnover rate is architecture dependent. These results illustrate that our approach allows investigating protein turnover and its implications in various cell types.


Subject(s)
Cells/metabolism , Proteins/chemistry , Proteins/metabolism , Animals , Cells/chemistry , Cells, Cultured , Humans , Mass Spectrometry , Mice , Peptides/chemistry , Peptides/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteomics
3.
J Neurosci Res ; 95(1-2): 301-310, 2017 01 02.
Article in English | MEDLINE | ID: mdl-27870402

ABSTRACT

There are inherent biological differences between males and females that contribute to sex differences in brain function and to many sex-specific illnesses and disorders. Traditionally, it has been thought that such differences are due largely to hormonal regulation; however, there are also genetic and epigenetic effects caused by the inheritance and unequal dosage of genes located on the X and Y chromosomes. Here we discuss the evidence in favor of a genetic and epigenetic basis for sexually dimorphic behavior, as a consequence of underlying differences in the regulation of genes that drive brain function. A better understanding of sex-specific molecular processes in the brain will provide further insight for the development of novel therapeutic approaches for the treatment of neuropsychiatric disorders characterized by sex differences. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain/physiology , Epigenomics , Gene Expression Regulation/genetics , Sex Characteristics , Animals , Humans , Sex Chromosomes/genetics
4.
J Neurosci ; 36(25): 6771-7, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27335407

ABSTRACT

UNLABELLED: The RNA modification N(6)-methyladenosine (m(6)A) influences mRNA stability and cell-type-specific developmental programming, and is highly abundant in the adult brain. However, it has not been determined whether m(6)A is dynamically regulated by experience. Based on transcriptome-wide profiling of m(6)A, we report that the level of m(6)A increases in the medial prefrontal cortex (mPFC) of mice in response to behavioral experience. The modulation was enriched near the stop codon of mRNAs, including genes related to neuronal plasticity. In primary cortical neurons, in vitro, modulation of m(6)A by the RNA demethylase FTO influenced the degradation profiles of a subset of transcripts with modulated sites. In vivo, the expression of Fto and the m(6)A methyltransferase, Mettl3 correlated with the observed increase in m(6)A levels post-training. Furthermore, targeted knockdown of FTO in the mPFC led to enhanced consolidation of cued fear memory. Thus, together with its role in early development, the dynamic regulation of m(6)A in the adult brain serves as an important epitranscriptomic mechanism associated with behavioral adaptation. SIGNIFICANCE STATEMENT: N(6)-methyladenosine (m(6)A) is the most prevalent internal modification on RNA, however, its cellular dynamics in vivo remains elusive. Here we provide the first demonstration of m(6)A upregulation in the mouse medial prefrontal cortex (mPFC) following behavioral training. Knocking down the m(6)A demethylase FTO in the mPFC, which increases total m(6)A level, results in enhanced consolidation of fear memory. Our findings suggest that m(6)A is regulated in an activity-dependent manner in the adult brain, and may function to fine-tune mRNA turnover during memory-related processes.


Subject(s)
Adenosine/analogs & derivatives , Memory/physiology , Neurons/metabolism , Prefrontal Cortex/cytology , Adenosine/genetics , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Cells, Cultured , Conditioning, Classical/physiology , Cues , Embryo, Mammalian , Exploratory Behavior/physiology , Fear/physiology , Gene Expression Profiling , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proteolysis , RNA, Small Interfering/genetics
5.
Biol Psychiatry ; 78(12): 848-59, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-25792222

ABSTRACT

BACKGROUND: RNA-directed regulation of epigenetic processes has recently emerged as an important feature of mammalian differentiation and development. Perturbation of this regulatory system in the brain may contribute to the development of neuropsychiatric disorders. METHODS: RNA sequencing was used to identify changes in the experience-dependent expression of long noncoding RNAs (lncRNAs) within the medial prefrontal cortex of adult mice. Transcripts were validated by real-time quantitative polymerase chain reaction and a candidate lncRNA, Gomafu, was selected for further investigation. The functional role of this schizophrenia-related lncRNA was explored in vivo by antisense oligonucleotide-mediated gene knockdown in the medial prefrontal cortex, followed by behavioral training and assessment of fear-related anxiety. Long noncoding RNA-directed epigenetic regulation of gene expression was investigated by chromatin and RNA immunoprecipitation assays. RESULTS: RNA sequencing analysis revealed changes in the expression of a significant number of genes related to neural plasticity and stress, as well as the dynamic regulation of lncRNAs. In particular, we detected a significant downregulation of Gomafu lncRNA. Our results revealed that Gomafu plays a role in mediating anxiety-like behavior and suggest that this may occur through an interaction with a key member of the polycomb repressive complex 1, BMI1, which regulates the expression of the schizophrenia-related gene beta crystallin (Crybb1). We also demonstrated a novel role for Crybb1 in mediating fear-induced anxiety-like behavior. CONCLUSIONS: Experience-dependent expression of lncRNAs plays an important role in the epigenetic regulation of adaptive behavior, and the perturbation of Gomafu may be related to anxiety and the development of neuropsychiatric disorders.


Subject(s)
Anxiety/metabolism , Anxiety/physiopathology , Epigenesis, Genetic , Fear/physiology , Prefrontal Cortex/metabolism , RNA, Long Noncoding/metabolism , Animals , Anxiety/genetics , Brain-Derived Neurotrophic Factor/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Conditioning, Classical/physiology , Crystallins/metabolism , Gene Expression Profiling , Homer Scaffolding Proteins , Male , Mice , Mice, Inbred C57BL , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , beta-Crystallin B Chain
6.
Eur J Neurosci ; 40(7): 3032-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25041363

ABSTRACT

Activity-dependent gene expression depends, in part, on transcriptional regulation that is coordinated by rapid changes in the chromatin landscape as well as the covalent modification of DNA. Here we demonstrate that the expression of brain-derived neurotrophic factor (BDNF), a gene that is critically involved in neural plasticity and subject to epigenetic regulation, is regulated by the RNA/DNA editing enzyme, activation-induced cytidine deaminase (AID). Similar to previous reports, we observed an activity-dependent induction of BDNF exon IV mRNA expression, which correlated with a reduction in DNA methylation within the BDNF P4 promoter. Lentiviral-mediated knockdown of AID disrupted these effects and inhibited BDNF exon IV mRNA expression, an effect that was associated with decreased cAMP response element-binding protein occupancy within the BDNF P4 promoter. Thus, together with other epigenetic mechanisms, AID plays a key role in regulating activity-dependent BDNF expression in post-mitotic cortical neurons.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/metabolism , Cytidine Deaminase/genetics , Gene Expression Regulation , Neurons/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Methylation , Mice , Mice, Inbred C57BL , Mitosis , RNA, Messenger/metabolism
7.
Neurobiol Learn Mem ; 105: 125-32, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23806749

ABSTRACT

Dynamic variations in DNA methylation regulate neuronal gene expression in an experience-dependent manner. Although DNA methylation has been implicated in synaptic plasticity, learning and memory, active DNA demethylation is also induced by learning, which suggests that an interaction between the two processes is necessary for cognitive function. Active DNA demethylation is a complex process involving a variety of proteins and epigenetic regulatory enzymes, the understanding of which with respect to its role in the adult brain is in its infancy. We here provide an overview of the current understanding of active DNA demethylation, and describe how this process may establish persistent epigenetic states that are associated with neural plasticity and memory formation.


Subject(s)
DNA Methylation/physiology , Epigenesis, Genetic , Memory/physiology , Neuronal Plasticity/genetics , Animals , Mice
8.
Trends Neurosci ; 36(1): 3-13, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23041052

ABSTRACT

DNA methylation was once considered to be a static epigenetic modification whose primary function was restricted to directing the development of cellular phenotype. However, it is now evident that the methylome is dynamically regulated across the lifespan: during development as a putative mechanism by which early experience leaves a lasting signature on the genome and during adulthood as a function of behavioral adaptation. Here, we propose that experience-dependent variations in DNA methylation, particularly within the context of learning and memory, represent a form of genomic metaplasticity that serves to prime the transcriptional response to later learning-related stimuli and neuronal reactivation.


Subject(s)
Adaptation, Physiological/physiology , Brain/physiology , DNA Methylation/physiology , Learning/physiology , Memory/physiology , Animals , Genomics , Humans
9.
Nat Neurosci ; 14(9): 1115-7, 2011 Aug 14.
Article in English | MEDLINE | ID: mdl-21841775

ABSTRACT

MicroRNAs are small non-coding RNAs that mediate post-transcriptional gene silencing. Fear-extinction learning in C57/Bl6J mice led to increased expression of the brain-specific microRNA miR-128b, which disrupted stability of several plasticity-related target genes and regulated formation of fear-extinction memory. Increased miR-128b activity may therefore facilitate the transition from retrieval of the original fear memory toward the formation of a new fear-extinction memory.


Subject(s)
Brain/metabolism , Extinction, Psychological/physiology , Fear , Memory/physiology , MicroRNAs/metabolism , Animals , Behavior, Animal , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Transformed , Conditioning, Classical , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Nerve Tissue Proteins/metabolism , Potassium Chloride/pharmacology , Protein Phosphatase 1/metabolism , RNA, Small Interfering/metabolism , Reelin Protein , Serine Endopeptidases/metabolism , Trans-Activators/metabolism , Transduction, Genetic/methods , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...