Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.332
Filter
1.
Annu Rev Biochem ; 91: 183-195, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35303789

ABSTRACT

Gene regulation arises out of dynamic competition between nucleosomes, transcription factors, and other chromatin proteins for the opportunity to bind genomic DNA. The timescales of nucleosome assembly and binding of factors to DNA determine the outcomes of this competition at any given locus. Here, we review how these properties of chromatin proteins and the interplay between the dynamics of different factors are critical for gene regulation. We discuss how molecular structures of large chromatin-associated complexes, kinetic measurements, and high resolution mapping of protein-DNA complexes in vivo set the boundary conditions for chromatin dynamics, leading to models of how the steady state behaviors of regulatory elements arise.


Subject(s)
Chromatin , Nucleosomes , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA/genetics , DNA/metabolism , Nucleosomes/genetics , Transcription Factors/genetics
2.
Cell ; 184(16): 4237-4250.e19, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34297924

ABSTRACT

The organization of genomic DNA into defined nucleosomes has long been viewed as a hallmark of eukaryotes. This paradigm has been challenged by the identification of "minimalist" histones in archaea and more recently by the discovery of genes that encode fused remote homologs of the four eukaryotic histones in Marseilleviridae, a subfamily of giant viruses that infect amoebae. We demonstrate that viral doublet histones are essential for viral infectivity, localize to cytoplasmic viral factories after virus infection, and ultimately are found in the mature virions. Cryogenic electron microscopy (cryo-EM) structures of viral nucleosome-like particles show strong similarities to eukaryotic nucleosomes despite the limited sequence identify. The unique connectors that link the histone chains contribute to the observed instability of viral nucleosomes, and some histone tails assume structural roles. Our results further expand the range of "organisms" that require nucleosomes and suggest a specialized function of histones in the biology of these unusual viruses.


Subject(s)
DNA Viruses/metabolism , Histones/metabolism , Nucleosomes/metabolism , Amoeba/virology , Fluorescent Dyes/metabolism , Histones/chemistry , Models, Molecular , Proteomics , Virion/metabolism
3.
Cell ; 182(1): 127-144.e23, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32502394

ABSTRACT

Before zygotic genome activation (ZGA), the quiescent genome undergoes reprogramming to transition into the transcriptionally active state. However, the mechanisms underlying euchromatin establishment during early embryogenesis remain poorly understood. Here, we show that histone H4 lysine 16 acetylation (H4K16ac) is maintained from oocytes to fertilized embryos in Drosophila and mammals. H4K16ac forms large domains that control nucleosome accessibility of promoters prior to ZGA in flies. Maternal depletion of MOF acetyltransferase leading to H4K16ac loss causes aberrant RNA Pol II recruitment, compromises the 3D organization of the active genomic compartments during ZGA, and causes downregulation of post-zygotically expressed genes. Germline depletion of histone deacetylases revealed that other acetyl marks cannot compensate for H4K16ac loss in the oocyte. Moreover, zygotic re-expression of MOF was neither able to restore embryonic viability nor onset of X chromosome dosage compensation. Thus, maternal H4K16ac provides an instructive function to the offspring, priming future gene activation.


Subject(s)
Histones/metabolism , Lysine/metabolism , Transcriptional Activation/genetics , Acetylation , Animals , Base Sequence , Chromosome Segregation/genetics , Conserved Sequence , Dosage Compensation, Genetic , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , Evolution, Molecular , Female , Genome , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Male , Mammals/genetics , Mice , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Oocytes/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , X Chromosome/metabolism , Zygote/metabolism
4.
Cell ; 179(2): 470-484.e21, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31543265

ABSTRACT

Eukaryotic chromatin is highly condensed but dynamically accessible to regulation and organized into subdomains. We demonstrate that reconstituted chromatin undergoes histone tail-driven liquid-liquid phase separation (LLPS) in physiologic salt and when microinjected into cell nuclei, producing dense and dynamic droplets. Linker histone H1 and internucleosome linker lengths shared across eukaryotes promote phase separation of chromatin, tune droplet properties, and coordinate to form condensates of consistent density in manners that parallel chromatin behavior in cells. Histone acetylation by p300 antagonizes chromatin phase separation, dissolving droplets in vitro and decreasing droplet formation in nuclei. In the presence of multi-bromodomain proteins, such as BRD4, highly acetylated chromatin forms a new phase-separated state with droplets of distinct physical properties, which can be immiscible with unmodified chromatin droplets, mimicking nuclear chromatin subdomains. Our data suggest a framework, based on intrinsic phase separation of the chromatin polymer, for understanding the organization and regulation of eukaryotic genomes.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Chromatin/metabolism , E1A-Associated p300 Protein/metabolism , Histones/metabolism , Transcription Factors/metabolism , Acetylation , Animals , Escherichia coli/genetics , HeLa Cells , Humans , Nuclear Proteins/metabolism , Sf9 Cells
5.
Cell ; 176(6): 1490-1501.e12, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30765112

ABSTRACT

Methylation of histone H3 K79 by Dot1L is a hallmark of actively transcribed genes that depends on monoubiquitination of H2B K120 (H2B-Ub) and is an example of histone modification cross-talk that is conserved from yeast to humans. We report here cryo-EM structures of Dot1L bound to ubiquitinated nucleosome that show how H2B-Ub stimulates Dot1L activity and reveal a role for the histone H4 tail in positioning Dot1L. We find that contacts mediated by Dot1L and the H4 tail induce a conformational change in the globular core of histone H3 that reorients K79 from an inaccessible position, thus enabling this side chain to insert into the active site in a position primed for catalysis. Our study provides a comprehensive mechanism of cross-talk between histone ubiquitination and methylation and reveals structural plasticity in histones that makes it possible for histone-modifying enzymes to access residues within the nucleosome core.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Animals , Catalytic Domain , Chromatin/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/ultrastructure , Histones/chemistry , Histones/genetics , Humans , Methylation , Models, Molecular , Nucleosomes/metabolism , Protein Processing, Post-Translational , Receptor Cross-Talk , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitination , Xenopus laevis
6.
Cell ; 177(7): 1781-1796.e25, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31104845

ABSTRACT

DNA N6-adenine methylation (6mA) has recently been described in diverse eukaryotes, spanning unicellular organisms to metazoa. Here, we report a DNA 6mA methyltransferase complex in ciliates, termed MTA1c. It consists of two MT-A70 proteins and two homeobox-like DNA-binding proteins and specifically methylates dsDNA. Disruption of the catalytic subunit, MTA1, in the ciliate Oxytricha leads to genome-wide loss of 6mA and abolishment of the consensus ApT dimethylated motif. Mutants fail to complete the sexual cycle, which normally coincides with peak MTA1 expression. We investigate the impact of 6mA on nucleosome occupancy in vitro by reconstructing complete, full-length Oxytricha chromosomes harboring 6mA in native or ectopic positions. We show that 6mA directly disfavors nucleosomes in vitro in a local, quantitative manner, independent of DNA sequence. Furthermore, the chromatin remodeler ACF can overcome this effect. Our study identifies a diverged DNA N6-adenine methyltransferase and defines the role of 6mA in chromatin organization.


Subject(s)
Multienzyme Complexes/metabolism , Nucleosomes/enzymology , Oxytricha/enzymology , Protozoan Proteins/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Tetrahymena thermophila/enzymology , Multienzyme Complexes/genetics , Nucleosomes/genetics , Oxytricha/genetics , Protozoan Proteins/genetics , Site-Specific DNA-Methyltransferase (Adenine-Specific)/genetics , Tetrahymena thermophila/genetics
7.
Cell ; 179(6): 1342-1356.e23, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31759698

ABSTRACT

Mammalian switch/sucrose non-fermentable (mSWI/SNF) complexes are multi-component machines that remodel chromatin architecture. Dissection of the subunit- and domain-specific contributions to complex activities is needed to advance mechanistic understanding. Here, we examine the molecular, structural, and genome-wide regulatory consequences of recurrent, single-residue mutations in the putative coiled-coil C-terminal domain (CTD) of the SMARCB1 (BAF47) subunit, which cause the intellectual disability disorder Coffin-Siris syndrome (CSS), and are recurrently found in cancers. We find that the SMARCB1 CTD contains a basic α helix that binds directly to the nucleosome acidic patch and that all CSS-associated mutations disrupt this binding. Furthermore, these mutations abrogate mSWI/SNF-mediated nucleosome remodeling activity and enhancer DNA accessibility without changes in genome-wide complex localization. Finally, heterozygous CSS-associated SMARCB1 mutations result in dominant gene regulatory and morphologic changes during iPSC-neuronal differentiation. These studies unmask an evolutionarily conserved structural role for the SMARCB1 CTD that is perturbed in human disease.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , Mutation/genetics , Nucleosomes/metabolism , SMARCB1 Protein/genetics , Transcription Factors/metabolism , Amino Acid Sequence , Enhancer Elements, Genetic/genetics , Female , Genome, Human , HEK293 Cells , HeLa Cells , Heterozygote , Humans , Male , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Domains , SMARCB1 Protein/chemistry , SMARCB1 Protein/metabolism
8.
Cell ; 179(4): 953-963.e11, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31675501

ABSTRACT

Chromatin domains and their associated structures must be faithfully inherited through cellular division to maintain cellular identity. However, accessing the localized strategies preserving chromatin domain inheritance, specifically the transfer of parental, pre-existing nucleosomes with their associated post-translational modifications (PTMs) during DNA replication, is challenging in living cells. We devised an inducible, proximity-dependent labeling system to irreversibly mark replication-dependent H3.1 and H3.2 histone-containing nucleosomes at desired loci in mouse embryonic stem cells so that their fate after DNA replication could be followed. Strikingly, repressed chromatin domains are preserved through local re-deposition of parental nucleosomes. In contrast, nucleosomes decorating active chromatin domains do not exhibit such preservation. Notably, altering cell fate leads to an adjustment of the positional inheritance of parental nucleosomes that reflects the corresponding changes in chromatin structure. These findings point to important mechanisms that contribute to parental nucleosome segregation to preserve cellular identity.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Epigenesis, Genetic , Nucleosomes/genetics , Animals , Cell Differentiation/genetics , Cell Division/genetics , Cell Lineage/genetics , DNA Replication/genetics , Histones/genetics , Mice , Mouse Embryonic Stem Cells/metabolism , Nucleosomes/metabolism , Protein Processing, Post-Translational/genetics
9.
Cell ; 178(3): 624-639.e19, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348889

ABSTRACT

Recent breakthroughs with synthetic budding yeast chromosomes expedite the creation of synthetic mammalian chromosomes and genomes. Mammals, unlike budding yeast, depend on the histone H3 variant, CENP-A, to epigenetically specify the location of the centromere-the locus essential for chromosome segregation. Prior human artificial chromosomes (HACs) required large arrays of centromeric α-satellite repeats harboring binding sites for the DNA sequence-specific binding protein, CENP-B. We report the development of a type of HAC that functions independently of these constraints. Formed by an initial CENP-A nucleosome seeding strategy, a construct lacking repetitive centromeric DNA formed several self-sufficient HACs that showed no uptake of genomic DNA. In contrast to traditional α-satellite HAC formation, the non-repetitive construct can form functional HACs without CENP-B or initial CENP-A nucleosome seeding, revealing distinct paths to centromere formation for different DNA sequence types. Our developments streamline the construction and characterization of HACs to facilitate mammalian synthetic genome efforts.


Subject(s)
Centromere/metabolism , Chromosomes, Artificial, Human/metabolism , DNA, Satellite/metabolism , Binding Sites , Cell Line, Tumor , Centromere/genetics , Centromere Protein A/genetics , Centromere Protein A/metabolism , Centromere Protein B/deficiency , Centromere Protein B/genetics , Centromere Protein B/metabolism , Epigenesis, Genetic , Humans , Nucleosomes/chemistry , Nucleosomes/metabolism , Plasmids/genetics , Plasmids/metabolism
10.
Immunity ; 57(3): 462-477.e9, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38430908

ABSTRACT

Inducible nucleosome remodeling at hundreds of latent enhancers and several promoters shapes the transcriptional response to Toll-like receptor 4 (TLR4) signaling in macrophages. We aimed to define the identities of the transcription factors that promote TLR-induced remodeling. An analysis strategy based on ATAC-seq and single-cell ATAC-seq that enriched for genomic regions most likely to undergo remodeling revealed that the transcription factor nuclear factor κB (NF-κB) bound to all high-confidence peaks marking remodeling during the primary response to the TLR4 ligand, lipid A. Deletion of NF-κB subunits RelA and c-Rel resulted in the loss of remodeling at high-confidence ATAC-seq peaks, and CRISPR-Cas9 mutagenesis of NF-κB-binding motifs impaired remodeling. Remodeling selectivity at defined regions was conferred by collaboration with other inducible factors, including IRF3- and MAP-kinase-induced factors. Thus, NF-κB is unique among TLR4-activated transcription factors in its broad contribution to inducible nucleosome remodeling, alongside its ability to activate poised enhancers and promoters assembled into open chromatin.


Subject(s)
NF-kappa B , Toll-Like Receptor 4 , NF-kappa B/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Nucleosomes , Signal Transduction , Gene Expression Regulation , Transcription Factor RelA/metabolism
11.
Cell ; 175(3): 780-795.e15, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30318142

ABSTRACT

During mitosis, chromatin condensation shapes chromosomes as separate, rigid, and compact sister chromatids to facilitate their segregation. Here, we show that, unlike wild-type yeast chromosomes, non-chromosomal DNA circles and chromosomes lacking a centromere fail to condense during mitosis. The centromere promotes chromosome condensation strictly in cis through recruiting the kinases Aurora B and Bub1, which trigger the autonomous condensation of the entire chromosome. Shugoshin and the deacetylase Hst2 facilitated spreading the condensation signal to the chromosome arms. Targeting Aurora B to DNA circles or centromere-ablated chromosomes or releasing Shugoshin from PP2A-dependent inhibition bypassed the centromere requirement for condensation and enhanced the mitotic stability of DNA circles. Our data indicate that yeast cells license the chromosome-autonomous condensation of their chromatin in a centromere-dependent manner, excluding from this process non-centromeric DNA and thereby inhibiting their propagation.


Subject(s)
Centromere/genetics , Chromosomes, Fungal/genetics , Mitosis , Saccharomyces cerevisiae/genetics , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sirtuin 2/genetics , Sirtuin 2/metabolism
12.
Cell ; 175(4): 1074-1087.e18, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388444

ABSTRACT

Mutation rates along the genome are highly variable and influenced by several chromatin features. Here, we addressed how nucleosomes, the most pervasive chromatin structure in eukaryotes, affect the generation of mutations. We discovered that within nucleosomes, the somatic mutation rate across several tumor cohorts exhibits a strong 10 base pair (bp) periodicity. This periodic pattern tracks the alternation of the DNA minor groove facing toward and away from the histones. The strength and phase of the mutation rate periodicity are determined by the mutational processes active in tumors. We uncovered similar periodic patterns in the genetic variation among human and Arabidopsis populations, also detectable in their divergence from close species, indicating that the same principles underlie germline and somatic mutation rates. We propose that differential DNA damage and repair processes dependent on the minor groove orientation in nucleosome-bound DNA contribute to the 10-bp periodicity in AT/CG content in eukaryotic genomes.


Subject(s)
DNA/genetics , Germ-Line Mutation , Mutation Rate , Nucleosomes/genetics , Arabidopsis/genetics , DNA/chemistry , GC Rich Sequence , Genetic Variation , Nucleic Acid Conformation , Nucleosomes/chemistry
13.
Cell ; 174(4): 818-830.e11, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30057113

ABSTRACT

Rtt109 is a unique histone acetyltransferase acetylating histone H3 lysine 56 (H3K56), a modification critical for DNA replication-coupled nucleosome assembly and genome stability. In cells, histone chaperone Asf1 is essential for H3K56 acetylation, yet the mechanisms for H3K56 specificity and Asf1 requirement remain unknown. We have determined the crystal structure of the Rtt109-Asf1-H3-H4 complex and found that unwinding of histone H3 αN, where K56 is normally located, and stabilization of the very C-terminal ß strand of histone H4 by Asf1 are prerequisites for H3K56 acetylation. Unexpectedly, an interaction between Rtt109 and the central helix of histone H3 is also required. The observed multiprotein, multisite substrate recognition mechanism among histone modification enzymes provides mechanistic understandings of Rtt109 and Asf1 in H3K56 acetylation, as well as valuable insights into substrate recognition by histone modification enzymes in general.


Subject(s)
Aspergillus fumigatus/metabolism , Histone Acetyltransferases/metabolism , Histones/chemistry , Lysine/metabolism , Molecular Chaperones/metabolism , Acetylation , Amino Acid Sequence , Histone Acetyltransferases/chemistry , Histones/metabolism , Lysine/chemistry , Molecular Chaperones/chemistry , Protein Conformation , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Sequence Homology , Substrate Specificity
14.
Mol Cell ; 84(3): 429-446.e17, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38215753

ABSTRACT

Nucleosomes, the basic structural units of chromatin, hinder recruitment and activity of various DNA repair proteins, necessitating modifications that enhance DNA accessibility. Poly(ADP-ribosyl)ation (PARylation) of proteins near damage sites is an essential initiation step in several DNA-repair pathways; however, its effects on nucleosome structural dynamics and organization are unclear. Using NMR, cryoelectron microscopy (cryo-EM), and biochemical assays, we show that PARylation enhances motions of the histone H3 tail and DNA, leaving the configuration of the core intact while also stimulating nuclease digestion and ligation of nicked nucleosomal DNA by LIG3. PARylation disrupted interactions between nucleosomes, preventing self-association. Addition of LIG3 and XRCC1 to PARylated nucleosomes generated condensates that selectively partition DNA repair-associated proteins in a PAR- and phosphorylation-dependent manner in vitro. Our results establish that PARylation influences nucleosomes across different length scales, extending from the atom-level motions of histone tails to the mesoscale formation of condensates with selective compositions.


Subject(s)
Nucleosomes , Poly ADP Ribosylation , Nucleosomes/genetics , Poly ADP Ribosylation/genetics , Poly(ADP-ribose) Polymerases/metabolism , Cryoelectron Microscopy , Biomolecular Condensates , DNA Repair , Histones/genetics , Histones/metabolism , DNA/genetics , DNA/metabolism , DNA Damage , Poly (ADP-Ribose) Polymerase-1/metabolism
15.
Mol Cell ; 84(11): 2053-2069.e9, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38810649

ABSTRACT

Facilitates chromatin transcription (FACT) is a histone chaperone that supports transcription through chromatin in vitro, but its functional roles in vivo remain unclear. Here, we analyze the in vivo functions of FACT with the use of multi-omics analysis after rapid FACT depletion from human cells. We show that FACT depletion destabilizes chromatin and leads to transcriptional defects, including defective promoter-proximal pausing and elongation, and increased premature termination of RNA polymerase II. Unexpectedly, our analysis revealed that promoter-proximal pausing depends not only on the negative elongation factor (NELF) but also on the +1 nucleosome, which is maintained by FACT.


Subject(s)
Chromatin , High Mobility Group Proteins , Nucleosomes , Promoter Regions, Genetic , RNA Polymerase II , Transcription, Genetic , Transcriptional Elongation Factors , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Humans , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , Chromatin/metabolism , Chromatin/genetics , Nucleosomes/metabolism , Nucleosomes/genetics , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , HeLa Cells , Chromatin Assembly and Disassembly , HEK293 Cells , Transcription Elongation, Genetic , Transcription Termination, Genetic
16.
Mol Cell ; 84(2): 194-201, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38016477

ABSTRACT

In eukaryotic genomes, transcriptional machinery and nucleosomes compete for binding to DNA sequences; thus, a crucial aspect of gene regulatory element function is to modulate chromatin accessibility for transcription factor (TF) and RNA polymerase binding. Recent structural studies have revealed multiple modes of TF engagement with nucleosomes, but how initial "pioneering" results in steady-state DNA accessibility for further TF binding and RNA polymerase II (RNAPII) engagement has been unclear. Even less well understood is how distant sites of open chromatin interact with one another, such as when developmental enhancers activate promoters to release RNAPII for productive elongation. Here, we review evidence for the centrality of the conserved SWI/SNF family of nucleosome remodeling complexes, both in pioneering and in mediating enhancer-promoter contacts. Consideration of the nucleosome unwrapping and ATP hydrolysis activities of SWI/SNF complexes, together with their architectural features, may reconcile steady-state TF occupancy with rapid TF dynamics observed by live imaging.


Subject(s)
Nucleosomes , Transcription Factors , Nucleosomes/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin , DNA-Binding Proteins/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Epigenesis, Genetic , Chromatin Assembly and Disassembly
17.
Mol Cell ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38925115

ABSTRACT

The evolutionarily conserved HIRA/Hir histone chaperone complex and ASF1a/Asf1 co-chaperone cooperate to deposit histone (H3/H4)2 tetramers on DNA for replication-independent chromatin assembly. The molecular architecture of the HIRA/Hir complex and its mode of histone deposition have remained unknown. Here, we report the cryo-EM structure of the S. cerevisiae Hir complex with Asf1/H3/H4 at 2.9-6.8 Å resolution. We find that the Hir complex forms an arc-shaped dimer with a Hir1/Hir2/Hir3/Hpc2 stoichiometry of 2/4/2/4. The core of the complex containing two Hir1/Hir2/Hir2 trimers and N-terminal segments of Hir3 forms a central cavity containing two copies of Hpc2, with one engaged by Asf1/H3/H4, in a suitable position to accommodate a histone (H3/H4)2 tetramer, while the C-terminal segments of Hir3 harbor nucleic acid binding activity to wrap DNA around the Hpc2-assisted histone tetramer. The structure suggests a model for how the Hir/Asf1 complex promotes the formation of histone tetramers for their subsequent deposition onto DNA.

18.
Mol Cell ; 84(3): 476-489.e10, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38211589

ABSTRACT

Pioneer transcription factors (TFs) regulate cell fate by establishing transcriptionally primed and active states. However, cell fate control requires the coordination of both lineage-specific gene activation and repression of alternative-lineage programs, a process that is poorly understood. Here, we demonstrate that the pioneer TF FOXA coordinates with PRDM1 TF to recruit nucleosome remodeling and deacetylation (NuRD) complexes and Polycomb repressive complexes (PRCs), which establish highly occupied, accessible nucleosome conformation with bivalent epigenetic states, thereby preventing precocious and alternative-lineage gene expression during human endoderm differentiation. Similarly, the pioneer TF OCT4 coordinates with PRDM14 to form bivalent enhancers and repress cell differentiation programs in human pluripotent stem cells, suggesting that this may be a common and critical function of pioneer TFs. We propose that pioneer and PRDM TFs coordinate to safeguard cell fate through epigenetic repression mechanisms.


Subject(s)
Nucleosomes , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Nucleosomes/genetics , Cell Differentiation/genetics , Polycomb-Group Proteins/metabolism , Epigenesis, Genetic
19.
Mol Cell ; 84(5): 839-853.e12, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38242129

ABSTRACT

RNF168 plays a central role in the DNA damage response (DDR) by ubiquitylating histone H2A at K13 and K15. These modifications direct BRCA1-BARD1 and 53BP1 foci formation in chromatin, essential for cell-cycle-dependent DNA double-strand break (DSB) repair pathway selection. The mechanism by which RNF168 catalyzes the targeted accumulation of H2A ubiquitin conjugates to form repair foci around DSBs remains unclear. Here, using cryoelectron microscopy (cryo-EM), nuclear magnetic resonance (NMR) spectroscopy, and functional assays, we provide a molecular description of the reaction cycle and dynamics of RNF168 as it modifies the nucleosome and recognizes its ubiquitylation products. We demonstrate an interaction of a canonical ubiquitin-binding domain within full-length RNF168, which not only engages ubiquitin but also the nucleosome surface, clarifying how such site-specific ubiquitin recognition propels a signal amplification loop. Beyond offering mechanistic insights into a key DDR protein, our study aids in understanding site specificity in both generating and interpreting chromatin ubiquitylation.


Subject(s)
Nucleosomes , Ubiquitin-Protein Ligases , Nucleosomes/genetics , Cryoelectron Microscopy , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Histones/metabolism , Chromatin/genetics , DNA Repair , Ubiquitin/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , DNA Damage
20.
Cell ; 167(3): 709-721.e12, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27768892

ABSTRACT

Chromatin remodelers regulate genes by organizing nucleosomes around promoters, but their individual contributions are obfuscated by the complex in vivo milieu of factor redundancy and indirect effects. Genome-wide reconstitution of promoter nucleosome organization with purified proteins resolves this problem and is therefore a critical goal. Here, we reconstitute four stages of nucleosome architecture using purified components: yeast genomic DNA, histones, sequence-specific Abf1/Reb1, and remodelers RSC, ISW2, INO80, and ISW1a. We identify direct, specific, and sufficient contributions that in vivo observations validate. First, RSC clears promoters by translating poly(dA:dT) into directional nucleosome removal. Second, partial redundancy is recapitulated where INO80 alone, or ISW2 at Abf1/Reb1sites, positions +1 nucleosomes. Third, INO80 and ISW2 each align downstream nucleosomal arrays. Fourth, ISW1a tightens the spacing to canonical repeat lengths. Such a minimal set of rules and proteins establishes core mechanisms by which promoter chromatin architecture arises through a blend of redundancy and specialization.


Subject(s)
Chromatin Assembly and Disassembly , Nucleosomes/chemistry , Nucleosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Chromatin/chemistry , Chromatin/genetics , DNA, Fungal/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Genome, Fungal , Histones/chemistry , Histones/genetics , Poly dA-dT/chemistry , Protein Biosynthesis , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL