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.
Development ; 150(21)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37938830

ABSTRACT

The histone variant H2A.Z is central to early embryonic development, determining transcriptional competency through chromatin regulation of gene promoters and enhancers. In addition to genic loci, we find that H2A.Z resides at a subset of evolutionarily young repetitive elements, including DNA transposons, long interspersed nuclear elements and long terminal repeats, during early zebrafish development. Moreover, increases in H2A.Z occur when repetitive elements become transcriptionally active. Acquisition of H2A.Z corresponds with a reduction in the levels of the repressive histone modification H3K9me3 and a moderate increase in chromatin accessibility. Notably, however, de-repression of repetitive elements also leads to a significant reduction in H2A.Z over non-repetitive genic loci. Genic loss of H2A.Z is accompanied by transcriptional silencing at adjacent coding sequences, but remarkably, these impacts are mitigated by augmentation of total H2A.Z protein via transgenic overexpression. Our study reveals that levels of H2A.Z protein determine embryonic sensitivity to de-repression of repetitive elements, that repetitive elements can function as a nuclear sink for epigenetic factors and that competition for H2A.Z greatly influences overall transcriptional output during development. These findings uncover general mechanisms in which counteractive biological processes underlie phenotypic outcomes.


Subject(s)
Histones , Zebrafish , Animals , Histones/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Chromatin/genetics , Protein Processing, Post-Translational , Embryonic Development/genetics , Nucleosomes
2.
Cell Rep ; 36(7): 109551, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34407406

ABSTRACT

Rapid removal of histone H2A.Z from neuronal chromatin is a key step in learning-induced gene expression and memory formation, but mechanisms underlying learning-induced H2A.Z removal are unclear. Anp32e was recently identified as an H2A.Z-specific histone chaperone that removes H2A.Z from nucleosomes in dividing cells, but its role in non-dividing neurons is unclear. Moreover, prior studies investigated Anp32e function under steady-state rather than stimulus-induced conditions. Here, we show that Anp32e regulates H2A.Z binding in neurons under steady-state conditions, with lesser impact on stimulus-induced H2A.Z removal. Functionally, Anp32e depletion leads to H2A.Z-dependent impairment in transcription and dendritic arborization in cultured hippocampal neurons, as well as impaired recall of contextual fear memory and transcriptional regulation. Together, these data indicate that Anp32e regulates behavioral and morphological outcomes by preventing H2A.Z accumulation in chromatin rather than by regulating activity-mediated H2A.Z dynamics.


Subject(s)
Dendrites/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Memory , Molecular Chaperones/metabolism , Neurons/metabolism , Transcription, Genetic , Animals , Chromatin/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
Nat Commun ; 11(1): 5063, 2020 10 08.
Article in English | MEDLINE | ID: mdl-33033242

ABSTRACT

Genome-wide chromatin state underlies gene expression potential and cellular function. Epigenetic features and nucleosome positioning contribute to the accessibility of DNA, but widespread regulators of chromatin state are largely unknown. Our study investigates how coordination of ANP32E and H2A.Z contributes to genome-wide chromatin state in mouse fibroblasts. We define H2A.Z as a universal chromatin accessibility factor, and demonstrate that ANP32E antagonizes H2A.Z accumulation to restrict chromatin accessibility genome-wide. In the absence of ANP32E, H2A.Z accumulates at promoters in a hierarchical manner. H2A.Z initially localizes downstream of the transcription start site, and if H2A.Z is already present downstream, additional H2A.Z accumulates upstream. This hierarchical H2A.Z accumulation coincides with improved nucleosome positioning, heightened transcription factor binding, and increased expression of neighboring genes. Thus, ANP32E dramatically influences genome-wide chromatin accessibility through subtle refinement of H2A.Z patterns, providing a means to reprogram chromatin state and to hone gene expression levels.


Subject(s)
Chromatin/metabolism , Genome , Molecular Chaperones/metabolism , Animals , Cell Differentiation/genetics , DNA Helicases/metabolism , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Histones/metabolism , Mice , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...