Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Mol Cell ; 84(8): 1406-1421.e8, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38490199

ABSTRACT

Enhancers bind transcription factors, chromatin regulators, and non-coding transcripts to modulate the expression of target genes. Here, we report 3D genome structures of single mouse ES cells as they are induced to exit pluripotency and transition through a formative stage prior to undergoing neuroectodermal differentiation. We find that there is a remarkable reorganization of 3D genome structure where inter-chromosomal intermingling increases dramatically in the formative state. This intermingling is associated with the formation of a large number of multiway hubs that bring together enhancers and promoters with similar chromatin states from typically 5-8 distant chromosomal sites that are often separated by many Mb from each other. In the formative state, genes important for pluripotency exit establish contacts with emerging enhancers within these multiway hubs, suggesting that the structural changes we have observed may play an important role in modulating transcription and establishing new cell identities.


Subject(s)
Mouse Embryonic Stem Cells , Regulatory Sequences, Nucleic Acid , Mice , Animals , Mouse Embryonic Stem Cells/metabolism , Embryonic Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic
2.
Biol Open ; 13(1)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38149716

ABSTRACT

As cells exit the pluripotent state and begin to commit to a specific lineage they must activate genes appropriate for that lineage while silencing genes associated with pluripotency and preventing activation of lineage-inappropriate genes. The Nucleosome Remodelling and Deacetylation (NuRD) complex is essential for pluripotent cells to successfully undergo lineage commitment. NuRD controls nucleosome density at regulatory sequences to facilitate transcriptional responses, and also has been shown to prevent unscheduled transcription (transcriptional noise) in undifferentiated pluripotent cells. How these activities combine to ensure cells engage a gene expression program suitable for successful lineage commitment has not been determined. Here, we show that NuRD is not required to silence all genes. Rather, it restricts expression of genes primed for activation upon exit from the pluripotent state, but maintains them in a transcriptionally permissive state in self-renewing conditions, which facilitates their subsequent activation upon exit from naïve pluripotency. We further show that NuRD coordinates gene expression changes, which acts to maintain a barrier between different stable states. Thus NuRD-mediated chromatin remodelling serves multiple functions, including reducing transcriptional noise, priming genes for activation and coordinating the transcriptional response to facilitate lineage commitment.


Subject(s)
DNA-Binding Proteins , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Nucleosomes , Cell Differentiation/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics
3.
Stem Cell Res ; 46: 101867, 2020 07.
Article in English | MEDLINE | ID: mdl-32535494

ABSTRACT

Differentiation of mammalian pluripotent cells involves large-scale changes in transcription and, among the molecules that orchestrate these changes, chromatin remodellers are essential to initiate, establish and maintain a new gene regulatory network. The Nucleosome Remodelling and Deacetylation (NuRD) complex is a highly conserved chromatin remodeller which fine-tunes gene expression in embryonic stem cells. While the function of NuRD in mouse pluripotent cells has been well defined, no study yet has defined NuRD function in human pluripotent cells. Here we find that while NuRD activity is required for lineage commitment from primed pluripotency in both human and mouse cells, the nature of this requirement is surprisingly different. While mouse embryonic stem cells (mESC) and epiblast stem cells (mEpiSC) require NuRD to maintain an appropriate differentiation trajectory as judged by gene expression profiling, human induced pluripotent stem cells (hiPSC) lacking NuRD fail to even initiate these trajectories. Further, while NuRD activity is dispensable for self-renewal of mESCs and mEpiSCs, hiPSCs require NuRD to maintain a stable self-renewing state. These studies reveal that failure to properly fine-tune gene expression and/or to reduce transcriptional noise through the action of a highly conserved chromatin remodeller can have different consequences in human and mouse pluripotent stem cells.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Mice , Nucleosomes
4.
EMBO J ; 38(12)2019 06 17.
Article in English | MEDLINE | ID: mdl-31036553

ABSTRACT

Multiprotein chromatin remodelling complexes show remarkable conservation of function amongst metazoans, even though components present in invertebrates are often found as multiple paralogous proteins in vertebrate complexes. In some cases, these paralogues specify distinct biochemical and/or functional activities in vertebrate cells. Here, we set out to define the biochemical and functional diversity encoded by one such group of proteins within the mammalian Nucleosome Remodelling and Deacetylation (NuRD) complex: Mta1, Mta2 and Mta3. We find that, in contrast to what has been described in somatic cells, MTA proteins are not mutually exclusive within embryonic stem (ES) cell NuRD and, despite subtle differences in chromatin binding and biochemical interactions, serve largely redundant functions. ES cells lacking all three MTA proteins exhibit complete NuRD loss of function and are viable, allowing us to identify a previously unreported function for NuRD in reducing transcriptional noise, which is essential for maintaining a proper differentiation trajectory during early stages of lineage commitment.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Transcription, Genetic , Animals , Cells, Cultured , Cellular Reprogramming/genetics , DNA-Binding Proteins/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Signal-To-Noise Ratio , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Transcription, Genetic/physiology
5.
Nat Commun ; 9(1): 4300, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30327463

ABSTRACT

Chromatin structure and function is regulated by reader proteins recognizing histone modifications and/or histone variants. We recently identified that PWWP2A tightly binds to H2A.Z-containing nucleosomes and is involved in mitotic progression and cranial-facial development. Here, using in vitro assays, we show that distinct domains of PWWP2A mediate binding to free linker DNA as well as H3K36me3 nucleosomes. In vivo, PWWP2A strongly recognizes H2A.Z-containing regulatory regions and weakly binds H3K36me3-containing gene bodies. Further, PWWP2A binds to an MTA1-specific subcomplex of the NuRD complex (M1HR), which consists solely of MTA1, HDAC1, and RBBP4/7, and excludes CHD, GATAD2 and MBD proteins. Depletion of PWWP2A leads to an increase of acetylation levels on H3K27 as well as H2A.Z, presumably by impaired chromatin recruitment of M1HR. Thus, this study identifies PWWP2A as a complex chromatin-binding protein that serves to direct the deacetylase complex M1HR to H2A.Z-containing chromatin, thereby promoting changes in histone acetylation levels.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Histone Deacetylases/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Repressor Proteins/metabolism , Acetylation , Animals , Chromosomal Proteins, Non-Histone/genetics , HEK293 Cells , Histone Deacetylases/genetics , Histones/genetics , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Nucleosomes/metabolism , RNA, Small Interfering , Repressor Proteins/genetics , Trans-Activators
6.
Mol Cell ; 71(1): 56-72.e4, 2018 07 05.
Article in English | MEDLINE | ID: mdl-30008319

ABSTRACT

Chromatin remodeling complexes play essential roles in metazoan development through widespread control of gene expression, but the precise molecular mechanisms by which they do this in vivo remain ill defined. Using an inducible system with fine temporal resolution, we show that the nucleosome remodeling and deacetylation (NuRD) complex controls chromatin architecture and the protein binding repertoire at regulatory regions during cell state transitions. This is primarily exerted through its nucleosome remodeling activity while deacetylation at H3K27 follows changes in gene expression. Additionally, NuRD activity influences association of RNA polymerase II at transcription start sites and subsequent nascent transcript production, thereby guiding the establishment of lineage-appropriate transcriptional programs. These findings provide a detailed molecular picture of genome-wide modulation of lineage-specific transcription by an essential chromatin remodeling complex as well as insight into the orchestration of molecular events involved in transcriptional transitions in vivo. VIDEO ABSTRACT.


Subject(s)
Gene Expression Regulation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mouse Embryonic Stem Cells/metabolism , Nucleosomes/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , Acetylation , Animals , Cell Line , Histones/genetics , Histones/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Nucleosomes/genetics , RNA Polymerase II/genetics , Transcription Initiation Site
7.
Nat Commun ; 9(1): 2520, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29955052

ABSTRACT

A major challenge in single-molecule imaging is tracking the dynamics of proteins or complexes for long periods of time in the dense environments found in living cells. Here, we introduce the concept of using FRET to enhance the photophysical properties of photo-modulatable (PM) fluorophores commonly used in such studies. By developing novel single-molecule FRET pairs, consisting of a PM donor fluorophore (either mEos3.2 or PA-JF549) next to a photostable acceptor dye JF646, we demonstrate that FRET competes with normal photobleaching kinetic pathways to increase the photostability of both donor fluorophores. This effect was further enhanced using a triplet-state quencher. Our approach allows us to significantly improve single-molecule tracking of chromatin-binding proteins in live mammalian cells. In addition, it provides a novel way to track the localization and dynamics of protein complexes by labeling one protein with the PM donor and its interaction partner with the acceptor dye.


Subject(s)
Chromatin/chemistry , Microscopy, Fluorescence/methods , Mouse Embryonic Stem Cells/metabolism , Single Molecule Imaging/methods , Animals , Cell Line , Chromatin/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/ultrastructure , Photobleaching
8.
Nat Protoc ; 13(5): 1034-1061, 2018 05.
Article in English | MEDLINE | ID: mdl-29674753

ABSTRACT

Fluorescence imaging and chromosome conformation capture assays such as Hi-C are key tools for studying genome organization. However, traditionally, they have been carried out independently, making integration of the two types of data difficult to perform. By trapping individual cell nuclei inside a well of a 384-well glass-bottom plate with an agarose pad, we have established a protocol that allows both fluorescence imaging and Hi-C processing to be carried out on the same single cell. The protocol identifies 30,000-100,000 chromosome contacts per single haploid genome in parallel with fluorescence images. Contacts can be used to calculate intact genome structures to better than 100-kb resolution, which can then be directly compared with the images. Preparation of 20 single-cell Hi-C libraries using this protocol takes 5 d of bench work by researchers experienced in molecular biology techniques. Image acquisition and analysis require basic understanding of fluorescence microscopy, and some bioinformatics knowledge is required to run the sequence-processing tools described here.


Subject(s)
Chromatin/ultrastructure , Chromosomes/ultrastructure , Molecular Biology/methods , Molecular Conformation , Mouse Embryonic Stem Cells , Optical Imaging/methods , Animals , Cells, Cultured , Imaging, Three-Dimensional/methods , Mice , Single-Cell Analysis/methods
9.
Adv Anat Embryol Cell Biol ; 229: 3-14, 2018.
Article in English | MEDLINE | ID: mdl-29177761

ABSTRACT

The very first cell divisions in mammalian embryogenesis produce a ball of cells, each with the potential to form any cell in the developing embryo or placenta. At some point, the embryo produces enough cells that some are located on the outside of the embryo, while others are completely surrounded by other cells. It is at this point that cells undergo the very first lineage commitment event: outer cells form the trophectoderm and lose the potential to form embryonic lineages, while inner cells form the Inner Cell Mass, which retain embryonic potential. Cell identity is defined by gene expression patterns, and gene expression is largely controlled by how the DNA is packaged into chromatin. A number of protein complexes exist which are able to use the energy of ATP to remodel chromatin: that is, to alter the nucleosome topology of chromatin. Here, we summarise the evidence that chromatin remodellers play essential roles in the successful completion of preimplantation development in mammals and describe recent efforts to understand the molecular mechanisms through which chromatin remodellers facilitate the successful completion of the first cell fate decisions in mammalian embryogenesis.


Subject(s)
Cell Differentiation , Chromatin Assembly and Disassembly , Embryonic Development , Animals , Blastocyst , Chromatin , Embryo, Mammalian , Embryonic Development/genetics
10.
J Exp Med ; 214(10): 3085-3104, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28899870

ABSTRACT

Differentiation of lineage-committed cells from multipotent progenitors requires the establishment of accessible chromatin at lineage-specific transcriptional enhancers and promoters, which is mediated by pioneer transcription factors that recruit activating chromatin remodeling complexes. Here we show that the Mbd3/nucleosome remodeling and deacetylation (NuRD) chromatin remodeling complex opposes this transcriptional pioneering during B cell programming of multipotent lymphoid progenitors by restricting chromatin accessibility at B cell enhancers and promoters. Mbd3/NuRD-deficient lymphoid progenitors therefore prematurely activate a B cell transcriptional program and are biased toward overproduction of pro-B cells at the expense of T cell progenitors. The striking reduction in early thymic T cell progenitors results in compensatory hyperproliferation of immature thymocytes and development of T cell lymphoma. Our results reveal that Mbd3/NuRD can regulate multilineage differentiation by constraining the activation of dormant lineage-specific enhancers and promoters. In this way, Mbd3/NuRD protects the multipotency of lymphoid progenitors, preventing B cell-programming transcription factors from prematurely enacting lineage commitment. Mbd3/NuRD therefore controls the fate of lymphoid progenitors, ensuring appropriate production of lineage-committed progeny and suppressing tumor formation.


Subject(s)
B-Lymphocytes/metabolism , Carcinogenesis/metabolism , Cell Lineage/physiology , DNA-Binding Proteins/physiology , Lymphocytes/physiology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Gene Expression Regulation/physiology , Lymphoma, T-Cell/etiology , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/physiology , Thymocytes/metabolism , Thymocytes/physiology
11.
Nature ; 544(7648): 59-64, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28289288

ABSTRACT

The folding of genomic DNA from the beads-on-a-string-like structure of nucleosomes into higher-order assemblies is crucially linked to nuclear processes. Here we calculate 3D structures of entire mammalian genomes using data from a new chromosome conformation capture procedure that allows us to first image and then process single cells. The technique enables genome folding to be examined at a scale of less than 100 kb, and chromosome structures to be validated. The structures of individual topological-associated domains and loops vary substantially from cell to cell. By contrast, A and B compartments, lamina-associated domains and active enhancers and promoters are organized in a consistent way on a genome-wide basis in every cell, suggesting that they could drive chromosome and genome folding. By studying genes regulated by pluripotency factor and nucleosome remodelling deacetylase (NuRD), we illustrate how the determination of single-cell genome structure provides a new approach for investigating biological processes.


Subject(s)
Chromatin Assembly and Disassembly , Genome , Molecular Imaging/methods , Nucleosomes/chemistry , Single-Cell Analysis/methods , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Mammalian/chemistry , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Enhancer Elements, Genetic , G1 Phase , Gene Expression Regulation , Gene Regulatory Networks , Genome/genetics , Haploidy , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Models, Molecular , Molecular Conformation , Molecular Imaging/standards , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Reproducibility of Results , Single-Cell Analysis/standards , Cohesins
12.
Elife ; 62017 03 20.
Article in English | MEDLINE | ID: mdl-28318487

ABSTRACT

Turning genes on and off is essential for development and homeostasis, yet little is known about the sequence and causal role of chromatin state changes during the repression of active genes. This is surprising, as defective gene silencing underlies developmental abnormalities and disease. Here we delineate the sequence and functional contribution of transcriptional repression mechanisms at high temporal resolution. Inducible entry of the NuRD-interacting transcriptional regulator Ikaros into mouse pre-B cell nuclei triggered immediate binding to target gene promoters. Rapid RNAP2 eviction, transcriptional shutdown, nucleosome invasion, and reduced transcriptional activator binding required chromatin remodeling by NuRD-associated Mi2beta/CHD4, but were independent of HDAC activity. Histone deacetylation occurred after transcriptional repression. Nevertheless, HDAC activity contributed to stable gene silencing. Hence, high resolution mapping of transcriptional repression reveals complex and interdependent mechanisms that underpin rapid transitions between transcriptional states, and elucidates the temporal order, functional role and mechanistic separation of NuRD-associated enzymatic activities.


Subject(s)
Down-Regulation , Gene Silencing , Transcription, Genetic , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Histone Deacetylases/metabolism , Ikaros Transcription Factor/metabolism , Mice , Precursor Cells, B-Lymphoid/physiology , RNA Polymerase II/metabolism , Time Factors
13.
Development ; 143(17): 3074-84, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27471257

ABSTRACT

Sall4 is an essential transcription factor for early mammalian development and is frequently overexpressed in cancer. Although it is reported to play an important role in embryonic stem cell (ESC) self-renewal, whether it is an essential pluripotency factor has been disputed. Here, we show that Sall4 is dispensable for mouse ESC pluripotency. Sall4 is an enhancer-binding protein that prevents precocious activation of the neural gene expression programme in ESCs but is not required for maintenance of the pluripotency gene regulatory network. Although a proportion of Sall4 protein physically associates with the Nucleosome Remodelling and Deacetylase (NuRD) complex, Sall4 neither recruits NuRD to chromatin nor influences transcription via NuRD; rather, free Sall4 protein regulates transcription independently of NuRD. We propose a model whereby enhancer binding by Sall4 and other pluripotency-associated transcription factors is responsible for maintaining the balance between transcriptional programmes in pluripotent cells.


Subject(s)
DNA-Binding Proteins/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Chromatin Immunoprecipitation , Computational Biology , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Mass Spectrometry , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Nucleosomes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
14.
Development ; 142(15): 2586-97, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26116663

ABSTRACT

Chromatin remodelling proteins are essential for different aspects of metazoan biology, yet functional details of why these proteins are important are lacking. Although it is possible to describe the biochemistry of how they remodel chromatin, their chromatin-binding profiles in cell lines, and gene expression changes upon loss of a given protein, in very few cases can this easily translate into an understanding of how the function of that protein actually influences a developmental process. Here, we investigate how the chromatin remodelling protein CHD4 facilitates the first lineage decision in mammalian embryogenesis. Embryos lacking CHD4 can form a morphologically normal early blastocyst, but are unable to successfully complete the first lineage decision and form functional trophectoderm (TE). In the absence of a functional TE, Chd4 mutant blastocysts do not implant and are hence not viable. By measuring transcript levels in single cells from early embryos, we show that CHD4 influences the frequency at which unspecified cells in preimplantation stage embryos express lineage markers prior to the execution of this first lineage decision. In the absence of CHD4, this frequency is increased in 16-cell embryos, and by the blastocyst stage cells fail to properly adopt a TE gene expression programme. We propose that CHD4 allows cells to undertake lineage commitment in vivo by modulating the frequency with which lineage-specification genes are expressed. This provides novel insight into both how lineage decisions are made in mammalian cells, and how a chromatin remodelling protein functions to facilitate lineage commitment.


Subject(s)
Blastocyst/physiology , Cell Differentiation/physiology , Cell Lineage/physiology , DNA Helicases/metabolism , Gene Expression Regulation, Developmental/physiology , Animals , Chromatin Assembly and Disassembly/genetics , Crosses, Genetic , DNA Primers/genetics , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Multiplex Polymerase Chain Reaction , Single-Cell Analysis
15.
Neural Dev ; 10: 13, 2015 May 02.
Article in English | MEDLINE | ID: mdl-25934499

ABSTRACT

BACKGROUND: Chromatin-modifying complexes have key roles in regulating various aspects of neural stem cell biology, including self-renewal and neurogenesis. The methyl binding domain 3/nucleosome remodelling and deacetylation (MBD3/NuRD) co-repressor complex facilitates lineage commitment of pluripotent cells in early mouse embryos and is important for stem cell homeostasis in blood and skin, but its function in neurogenesis had not been described. Here, we show for the first time that MBD3/NuRD function is essential for normal neurogenesis in mice. RESULTS: Deletion of MBD3, a structural component of the NuRD complex, in the developing mouse central nervous system resulted in reduced cortical thickness, defects in the proper specification of cortical projection neuron subtypes and neonatal lethality. These phenotypes are due to alterations in PAX6+ apical progenitor cell outputs, as well as aberrant terminal neuronal differentiation programmes of cortical plate neurons. Normal numbers of PAX6+ apical neural progenitor cells were generated in the MBD3/NuRD-mutant cortex; however, the PAX6+ apical progenitor cells generate EOMES+ basal progenitor cells in reduced numbers. Cortical progenitor cells lacking MBD3/NuRD activity generate neurons that express both deep- and upper-layer markers. Using laser capture microdissection, gene expression profiling and chromatin immunoprecipitation, we provide evidence that MBD3/NuRD functions to control gene expression patterns during neural development. CONCLUSIONS: Our data suggest that although MBD3/NuRD is not required for neural stem cell lineage commitment, it is required to repress inappropriate transcription in both progenitor cells and neurons to facilitate appropriate cell lineage choice and differentiation programmes.


Subject(s)
Cerebral Cortex/cytology , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Neural Stem Cells/cytology , Neurogenesis/physiology , Transcription Factors/physiology , Animals , Cell Count , Cell Cycle , Cell Lineage , Cerebral Cortex/abnormalities , Cerebral Cortex/embryology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Eye Proteins/physiology , Gene Expression Profiling , Homeodomain Proteins/physiology , Mice , Mice, Knockout , Neurogenesis/genetics , Neurons/classification , Neurons/cytology , Nucleosomes/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/physiology , Repressor Proteins/physiology , T-Box Domain Proteins/analysis , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Transgenes
16.
FEBS J ; 282(9): 1692-702, 2015 May.
Article in English | MEDLINE | ID: mdl-25354247

ABSTRACT

Proteins that modify the structure of chromatin are known to be important for various aspects of metazoan biology including development, disease and possibly ageing. Yet functional details of why these proteins are important, i.e. how their action influences a given biological process, are lacking. While it is now possible to describe the biochemistry of how these proteins remodel chromatin, their chromatin binding profiles in cell lines, or gene expression changes upon loss of a given protein, in very few cases has this easily translated into an understanding of how the function of that protein actually influences a developmental process. Given that many chromatin modifying proteins will largely exert their influence through control of gene expression, it is useful to consider developmental processes as changes in the gene regulatory network (GRN), with each cell type exhibiting a unique gene expression profile. In this essay we consider the impact of two abundant and highly conserved chromatin modifying complexes, namely the nucleosome remodelling and deacetylation (NuRD) complex and the polycomb repressive complex 2 (PRC2), on the change in GRNs associated with lineage commitment during early mammalian development. We propose that while the NuRD complex limits the stability of cell states and defines the developmental trajectory between two stable states, PRC2 activity is important for stabilizing a new GRN once established. Although these two complexes display different biochemical activities, chromatin binding profiles and mutant phenotypes, we propose a model to explain how they cooperate to facilitate the transition through cell states that is development.


Subject(s)
Cell Lineage , Chromatin/metabolism , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Mice
17.
Cell Stem Cell ; 15(1): 102-10, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24835571

ABSTRACT

The Nucleosome Remodeling and Deacetylase (NuRD) complex is essential for embryonic development and pluripotent stem cell differentiation. In this study, we investigated whether NuRD is also involved in the reverse biological process of induction of pluripotency in neural stem cells. By knocking out MBD3, an essential scaffold subunit of the NuRD complex, at different time points in reprogramming, we found that efficient formation of reprogramming intermediates and induced pluripotent stem cells from neural stem cells requires NuRD activity. We also show that reprogramming of epiblast-derived stem cells to naive pluripotency requires NuRD complex function and that increased MBD3/NuRD levels can enhance reprogramming efficiency when coexpressed with the reprogramming factor NANOG. Our results therefore show that the MBD3/NuRD complex plays a key role in reprogramming in certain contexts and that a chromatin complex required for cell differentiation can also promote reversion back to a naive pluripotent cell state.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Neural Stem Cells/physiology , Pluripotent Stem Cells/physiology , Animals , Cell Dedifferentiation/genetics , Cell Differentiation/genetics , Cell Line , Cellular Reprogramming/genetics , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Gene Knockout Techniques , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Nanog Homeobox Protein
19.
Biochem Soc Trans ; 41(3): 777-82, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23697937

ABSTRACT

The CHD4 (chromodomain-helicase-DNA-binding 4) (or Mi-2ß) protein is a founding component of the NuRD (nucleosome remodelling and deacetylation) complex. NuRD has long been known to function in transcriptional regulation, and is conserved throughout the animal and plant kingdoms. In recent years, evidence has steadily accumulated indicating that CHD4 can both function outside of the NuRD complex and also play important roles in cellular processes other than transcriptional regulation. A number of loss-of-function studies have identified important roles for CHD4 in the DNA-damage response and in cell cycle progression through S-phase and into G2. Furthermore, as part of NuRD, it participates in regulating acetylation levels of p53, thereby indirectly regulating the G1/S cell cycle checkpoint. Although CHD4 has a somewhat complicated relationship with the cell cycle, recent evidence indicates that CHD4 may exert some tumour-suppressor functions in human carcinogenesis. CHD4 is a defining member of the NuRD complex, but evidence is accumulating that CHD4 also plays important NuRD-independent roles in the DNA-damage response and cell cycle progression, as well as in transcriptional regulation.


Subject(s)
Autoantigens/metabolism , Cell Cycle/physiology , DNA Damage/physiology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Animals , Autoantigens/genetics , Cell Cycle/genetics , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , DNA Damage/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism
20.
Development ; 140(3): 505-12, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23293282

ABSTRACT

Through decades of research it has been established that some chromatin-modifying proteins can repress transcription, and thus are generally termed 'repressors'. Although classic repressors undoubtedly silence transcription, genome-wide studies have shown that many repressors are associated with actively transcribed loci and that this is a widespread phenomenon. Here, we review the evidence for the presence of repressors at actively transcribed regions and assess what roles they might be playing. We propose that the modulation of expression levels by chromatin-modifying, co-repressor complexes provides transcriptional fine-tuning that drives development.


Subject(s)
Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Animals , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enzyme Activation , Histone Deacetylases/genetics , Lysine/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL