Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 180, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36635295

ABSTRACT

The potential of pluripotent cells to respond to developmental cues and trigger cell differentiation is enhanced during the G1 phase of the cell cycle, but the molecular mechanisms involved are poorly understood. Variations in polycomb activity during interphase progression have been hypothesized to regulate the cell-cycle-phase-dependent transcriptional activation of differentiation genes during lineage transition in pluripotent cells. Here, we show that recruitment of Polycomb Repressive Complex 1 (PRC1) and associated molecular functions, ubiquitination of H2AK119 and three-dimensional chromatin interactions, are enhanced during S and G2 phases compared to the G1 phase. In agreement with the accumulation of PRC1 at target promoters upon G1 phase exit, cells in S and G2 phases show firmer transcriptional repression of developmental regulator genes that is drastically perturbed upon genetic ablation of the PRC1 catalytic subunit RING1B. Importantly, depletion of RING1B during retinoic acid stimulation interferes with the preference of mouse embryonic stem cells (mESCs) to induce the transcriptional activation of differentiation genes in G1 phase. We propose that incremental enrolment of polycomb repressive activity during interphase progression reduces the tendency of cells to respond to developmental cues during S and G2 phases, facilitating activation of cell differentiation in the G1 phase of the pluripotent cell cycle.


Subject(s)
Histones , Pluripotent Stem Cells , Polycomb Repressive Complex 1 , Animals , Mice , Cell Differentiation/genetics , Chromatin/genetics , Histones/metabolism , Interphase , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Pluripotent Stem Cells/cytology
2.
Development ; 149(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35686641

ABSTRACT

Enhancers control the establishment of spatiotemporal gene expression patterns throughout development. Over the past decade, the development of new technologies has improved our capacity to link enhancers with their target genes based on their colocalization within the same topological domains. However, the mechanisms that regulate how enhancers specifically activate some genes but not others within a given domain remain unclear. In this Review, we discuss recent insights into the factors controlling enhancer specificity, including the genetic composition of enhancers and promoters, the linear and 3D distance between enhancers and their target genes, and cell-type specific chromatin landscapes. We also discuss how elucidating the molecular principles of enhancer specificity might help us to better understand and predict the pathological consequences of human genetic, epigenetic and structural variants.


Subject(s)
Chromatin , Enhancer Elements, Genetic , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Epigenomics , Humans , Promoter Regions, Genetic/genetics
3.
STAR Protoc ; 3(3): 101492, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35769926

ABSTRACT

cis-regulatory elements (CREs) orchestrate the spatiotemporal control of gene expression. The regulatory activity of CREs is typically assessed by reporter assays, in which CREs are studied outside their endogenous context. To circumvent this problem, we developed a CRISPR-Cas9 knockin approach to study CREs in a scar-free genomic context. Here, we describe the design, transfection, and screening protocol to insert CREs in mouse embryonic stem cells. Our strategy can provide important insights into the sufficiency of CREs for gene expression control. For complete details on the use and execution of this protocol, please refer to Pachano et al. (2021).


Subject(s)
Mouse Embryonic Stem Cells , Regulatory Sequences, Nucleic Acid , Animals , Genomics , Mice , Transfection
4.
Nat Commun ; 12(1): 4344, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34272393

ABSTRACT

Poised enhancers (PEs) represent a genetically distinct set of distal regulatory elements that control the expression of major developmental genes. Before becoming activated in differentiating cells, PEs are already bookmarked in pluripotent cells with unique chromatin and topological features that could contribute to their privileged regulatory properties. However, since PEs were originally characterized in embryonic stem cells (ESC), it is currently unknown whether PEs are functionally conserved in vivo. Here, we show that the chromatin and 3D structural features of PEs are conserved among mouse pluripotent cells both in vitro and in vivo. We also uncovered that the interactions between PEs and their target genes are globally controlled by the combined action of Polycomb, Trithorax and architectural proteins. Moreover, distal regulatory sequences located close to developmental genes and displaying the typical genetic (i.e. CpG islands) and chromatin (i.e. high accessibility and H3K27me3 levels) features of PEs are commonly found across vertebrates. These putative PEs show high sequence conservation within specific vertebrate clades, with only a few being evolutionary conserved across all vertebrates. Lastly, by genetically disrupting PEs in mouse and chicken embryos, we demonstrate that these regulatory elements play essential roles during the induction of major developmental genes in vivo.


Subject(s)
Chromatin/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental/genetics , Histones/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Chick Embryo , Chromatin/genetics , Chromatin Immunoprecipitation Sequencing , CpG Islands , Embryonic Stem Cells/drug effects , Epigenesis, Genetic , Gene Deletion , Gene Expression Regulation, Developmental/drug effects , Germ Layers/metabolism , Homozygote , Mice , Phylogeny , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Transcription Factors/genetics
5.
Nat Genet ; 53(7): 1036-1049, 2021 07.
Article in English | MEDLINE | ID: mdl-34183853

ABSTRACT

CpG islands (CGIs) represent a widespread feature of vertebrate genomes, being associated with ~70% of all gene promoters. CGIs control transcription initiation by conferring nearby promoters with unique chromatin properties. In addition, there are thousands of distal or orphan CGIs (oCGIs) whose functional relevance is barely known. Here we show that oCGIs are an essential component of poised enhancers that augment their long-range regulatory activity and control the responsiveness of their target genes. Using a knock-in strategy in mouse embryonic stem cells, we introduced poised enhancers with or without oCGIs within topologically associating domains harboring genes with different types of promoters. Analysis of the resulting cell lines revealed that oCGIs act as tethering elements that promote the physical and functional communication between poised enhancers and distally located genes, particularly those with large CGI clusters in their promoters. Therefore, by acting as genetic determinants of gene-enhancer compatibility, CGIs can contribute to gene expression control under both physiological and potentially pathological conditions.


Subject(s)
CpG Islands , DNA Methylation , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Expression Regulation , Animals , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/metabolism , Gene Knock-In Techniques , Mice , Promoter Regions, Genetic
6.
Brief Funct Genomics ; 18(6): 358-366, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31673701

ABSTRACT

Polycomb group proteins (PcGs) control the epigenetic and transcriptional state of developmental genes and regulatory elements during mammalian embryogenesis. Moreover, PcGs can also contribute to 3D genome organization, adding an additional layer of complexity to their regulatory functions. Understanding the mechanistic basis and the dynamics of PcG-dependent chromatin structures will help us untangle the full complexity of PcG function during development. Since most studies concerning the 3D organization of PcG-bound chromatin in mammals have been performed in embryonic stem cells (ESCs), here we will focus on this cell type characterized by its unique self-renewal and pluripotency properties. More specifically, we will highlight recent findings and discuss open questions regarding how PcG-dependent changes in 3D chromatin architecture control gene expression, cellular identity and differentiation potential in ESCs. We believe that this can serve to illustrate the diverse regulatory mechanisms by which PcG proteins control the proper execution of gene expression programs during mammalian embryogenesis.


Subject(s)
Chromatin/metabolism , DNA Packaging/physiology , Embryonic Stem Cells/metabolism , Genome/physiology , Polycomb-Group Proteins/physiology , Animals , Chromatin/chemistry , Humans , Nucleic Acid Conformation , Polycomb-Group Proteins/metabolism , Protein Domains/physiology , Protein Folding
7.
Cell Microbiol ; 19(6)2017 06.
Article in English | MEDLINE | ID: mdl-28054438

ABSTRACT

Trichomonas vaginalis is a common sexually transmitted parasite that colonizes the human urogenital tract. Infections range from asymptomatic to highly inflammatory, depending on the host and the parasite strain. Different T. vaginalis strains vary greatly in their adherence and cytolytic capacities. These phenotypic differences might be attributed to differentially expressed genes as a consequence of extra-genetic variation, such as epigenetic modifications. In this study, we explored the role of histone acetylation in regulating gene transcription and pathogenesis in T. vaginalis. Here, we show that histone 3 lysine acetylation (H3KAc) is enriched in nucleosomes positioned around the transcription start site of active genes (BAP1 and BAP2) in a highly adherent parasite strain; compared with the low acetylation abundance in contrast to that observed in a less-adherent strain that expresses these genes at low levels. Additionally, exposition of less-adherent strain with a specific histone deacetylases inhibitor, trichostatin A, upregulated the transcription of BAP1 and BAP2 genes in concomitance with an increase in H3KAc abundance and chromatin accessibility around their transcription start sites. Moreover, we demonstrated that the binding of initiator binding protein, the transcription factor responsible for the initiation of transcription of ~75% of known T. vaginalis genes, depends on the histone acetylation state around the metazoan-like initiator to which initiator binding protein binds. Finally, we found that trichostatin A treatment increased parasite aggregation and adherence to host cells. Our data demonstrated for the first time that H3KAc is a permissive histone modification that functions to mediate both transcription and pathogenesis of the parasite T. vaginalis.


Subject(s)
Cell Adhesion/drug effects , Cell Aggregation/drug effects , Histones/metabolism , Trichomonas Vaginitis/pathology , Trichomonas vaginalis/genetics , Trichomonas vaginalis/pathogenicity , Acetylation/drug effects , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Aggregation/physiology , Cell Line, Tumor , Cervix Uteri/cytology , Cervix Uteri/metabolism , Cervix Uteri/parasitology , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , HeLa Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Metalloendopeptidases/genetics , Protein Binding/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Trichomonas Vaginitis/parasitology , Trichomonas vaginalis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...