Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO Rep ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890452

ABSTRACT

Heterochromatin stability is crucial for progenitor proliferation during early neurogenesis. It relays on the maintenance of local hubs of H3K9me. However, understanding the formation of efficient localized levels of H3K9me remains limited. To address this question, we used neural stem cells to analyze the function of the H3K9me2 demethylase PHF2, which is crucial for progenitor proliferation. Through mass-spectroscopy and genome-wide assays, we show that PHF2 interacts with heterochromatin components and is enriched at pericentromeric heterochromatin (PcH) boundaries where it maintains transcriptional activity. This binding is essential for silencing the satellite repeats, preventing DNA damage and genome instability. PHF2's depletion increases the transcription of heterochromatic repeats, accompanied by a decrease in H3K9me3 levels and alterations in PcH organization. We further show that PHF2's PHD and catalytic domains are crucial for maintaining PcH stability, thereby safeguarding genome integrity. These results highlight the multifaceted nature of PHF2's functions in maintaining heterochromatin stability and regulating gene expression during neural development. Our study unravels the intricate relationship between heterochromatin stability and progenitor proliferation during mammalian neurogenesis.

2.
Development ; 148(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34081130

ABSTRACT

Epigenetic factors have been shown to play a crucial role in X-linked intellectual disability (XLID). Here, we investigate the contribution of the XLID-associated histone demethylase PHF8 to astrocyte differentiation and function. Using genome-wide analyses and biochemical assays in mouse astrocytic cultures, we reveal a regulatory crosstalk between PHF8 and the Notch signaling pathway that balances the expression of the master astrocytic gene Nfia. Moreover, PHF8 regulates key synaptic genes in astrocytes by maintaining low levels of H4K20me3. Accordingly, astrocytic-PHF8 depletion has a striking effect on neuronal synapse formation and maturation in vitro. These data reveal that PHF8 is crucial in astrocyte development to maintain chromatin homeostasis and limit heterochromatin formation at synaptogenic genes. Our studies provide insights into the involvement of epigenetics in intellectual disability.


Subject(s)
Astrocytes/metabolism , Cell Differentiation , Gene Expression Regulation , Histone Demethylases/genetics , Transcription Factors/genetics , Animals , Astrocytes/cytology , Binding Sites , Biomarkers , Cell Differentiation/genetics , Cell Proliferation , Gene Expression Profiling , Histone Demethylases/metabolism , Histones/metabolism , Mice , Models, Biological , Neurogenesis , Neurons/metabolism , Protein Binding , Synapses/metabolism , Transcription Factors/metabolism , Transcription, Genetic
3.
Proc Natl Acad Sci U S A ; 116(39): 19464-19473, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31488723

ABSTRACT

Histone H3 lysine 9 methylation (H3K9me) is essential for cellular homeostasis; however, its contribution to development is not well established. Here, we demonstrate that the H3K9me2 demethylase PHF2 is essential for neural progenitor proliferation in vitro and for early neurogenesis in the chicken spinal cord. Using genome-wide analyses and biochemical assays we show that PHF2 controls the expression of critical cell cycle progression genes, particularly those related to DNA replication, by keeping low levels of H3K9me3 at promoters. Accordingly, PHF2 depletion induces R-loop accumulation that leads to extensive DNA damage and cell cycle arrest. These data reveal a role of PHF2 as a guarantor of genome stability that allows proper expansion of neural progenitors during development.


Subject(s)
DNA Damage , Histone Demethylases/metabolism , Homeodomain Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Chick Embryo , DNA Methylation , Embryonic Stem Cells , Epigenesis, Genetic , Genome-Wide Association Study , Histone Demethylases/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Neural Stem Cells/enzymology , Neurogenesis/physiology , Promoter Regions, Genetic , Transcription Factors/metabolism
4.
Nucleic Acids Res ; 46(7): 3351-3365, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29438503

ABSTRACT

During neurogenesis, dynamic developmental cues, transcription factors and histone modifying enzymes regulate the gene expression programs by modulating the activity of neural-specific enhancers. How transient developmental signals coordinate transcription factor recruitment to enhancers and to which extent chromatin modifiers contribute to enhancer activity is starting to be uncovered. Here, we take advantage of neural stem cells as a model to unravel the mechanisms underlying neural enhancer activation in response to the TGFß signaling. Genome-wide experiments demonstrate that the proneural factor ASCL1 assists SMAD3 in the binding to a subset of enhancers. Once located at the enhancers, SMAD3 recruits the histone demethylase JMJD3 and the remodeling factor CHD8, creating the appropriate chromatin landscape to allow enhancer transcription and posterior gene activation. Finally, to analyze the phenotypical traits owed to cis-regulatory regions, we use CRISPR-Cas9 technology to demonstrate that the TGFß-responsive Neurog2 enhancer is essential for proper neuronal polarization.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Enhancer Elements, Genetic/genetics , Neurogenesis/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Lineage/genetics , Cell Polarity/genetics , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Promoter Regions, Genetic , Signal Transduction/genetics , Transcription Factors/genetics
5.
Nucleic Acids Res ; 45(7): 3800-3811, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28100697

ABSTRACT

A precise immune response is essential for cellular homeostasis and animal survival. The paramount importance of its control is reflected by the fact that its non-specific activation leads to inflammatory events that ultimately contribute to the appearance of many chronic diseases. However, the molecular mechanisms preventing non-specific activation and allowing a quick response upon signal activation are not yet fully understood. In this paper we uncover a new function of PHF8 blocking signal independent activation of immune gene promoters. Affinity purifications coupled with mass spectrometry analysis identified SIN3A and HDAC1 corepressors as new PHF8 interacting partners. Further molecular analysis demonstrated that prior to interferon gamma (IFNγ) stimulation, PHF8 is bound to a subset of IFNγ-responsive promoters. Through the association with HDAC1 and SIN3A, PHF8 keeps the promoters in a silent state, maintaining low levels of H4K20me1. Upon IFNγ treatment, PHF8 is phosphorylated by ERK2 and evicted from the promoters, correlating with an increase in H4K20me1 and transcriptional activation. Our data strongly indicate that in addition to its well-characterized function as a coactivator, PHF8 safeguards transcription to allow an accurate immune response.


Subject(s)
Histone Demethylases/metabolism , Interferon-gamma/pharmacology , Transcription Factors/metabolism , Transcriptional Activation , Cell Line , Chromatin/metabolism , Gene Silencing , Histone Deacetylase 1/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
SELECTION OF CITATIONS
SEARCH DETAIL
...