Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.520
Filter
1.
Methods Mol Biol ; 2856: 63-70, 2025.
Article in English | MEDLINE | ID: mdl-39283446

ABSTRACT

Three-dimensional (3D) chromosome structures are closely related to various chromosomal functions, and deep analysis of the structures is crucial for the elucidation of the functions. In recent years, chromosome conformation capture (3C) techniques combined with next-generation sequencing analysis have been developed to comprehensively reveal 3D chromosome structures. Micro-C is one such method that can detect the structures at nucleosome resolution. In this chapter, I provide a basic method for Micro-C analysis. I present and discuss a series of data analyses ranging from mapping to basic downstream analyses, including loop detection.


Subject(s)
High-Throughput Nucleotide Sequencing , Software , Workflow , High-Throughput Nucleotide Sequencing/methods , Humans , Chromosomes/genetics , Computational Biology/methods , Chromosome Mapping/methods , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism
2.
Mol Cell ; 84(18): 3371-3373, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39303676

ABSTRACT

In this issue of Molecular Cell, Engeholm et al.1 present cryo-EM structures of the chromatin remodeler Chd1 bound to a hexasome-nucleosome complex, an intermediate state during transcription either with or without FACT to restore the missing H2A-H2B dimer. These two binding modes reveal how Chd1 and FACT cooperate in nucleosome re-establishment during transcription.


Subject(s)
Cryoelectron Microscopy , DNA-Binding Proteins , Nucleosomes , Nucleosomes/metabolism , Nucleosomes/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Chromatin Assembly and Disassembly , Histones/metabolism , Histones/genetics , Humans , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Protein Binding , Transcription, Genetic , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/genetics , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/chemistry
3.
Mol Cell ; 84(18): 3438-3454.e8, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39232583

ABSTRACT

Spreading of H3K27me3 is crucial for the maintenance of mitotically inheritable Polycomb-mediated chromatin silencing in animals and plants. However, how Polycomb repressive complex 2 (PRC2) accesses unmodified nucleosomes in spreading regions for spreading H3K27me3 remains unclear. Here, we show in Arabidopsis thaliana that the chromatin remodeler PICKLE (PKL) plays a specialized role in H3K27me3 spreading to safeguard cell identity during differentiation. PKL specifically localizes to H3K27me3 spreading regions but not to nucleation sites and physically associates with PRC2. Loss of PKL disrupts the occupancy of the PRC2 catalytic subunit CLF in spreading regions and leads to aberrant dedifferentiation. Nucleosome density increase endowed by the ATPase function of PKL ensures that unmodified nucleosomes are accessible to PRC2 catalytic activity for H3K27me3 spreading. Our findings demonstrate that PKL-dependent nucleosome compaction is critical for PRC2-mediated H3K27me3 read-and-write function in H3K27me3 spreading, thus revealing a mechanism by which repressive chromatin domains are established and propagated.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cell Differentiation , Chromatin Assembly and Disassembly , Histones , Nucleosomes , Polycomb Repressive Complex 2 , Nucleosomes/metabolism , Nucleosomes/genetics , Arabidopsis/genetics , Arabidopsis/metabolism , Histones/metabolism , Histones/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/genetics , Gene Expression Regulation, Plant , Chromatin/metabolism , Chromatin/genetics
4.
Mol Cell ; 84(18): 3423-3437.e8, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39270644

ABSTRACT

To maintain the nucleosome organization of transcribed genes, ATP-dependent chromatin remodelers collaborate with histone chaperones. Here, we show that at the 5' ends of yeast genes, RNA polymerase II (RNAPII) generates hexasomes that occur directly adjacent to nucleosomes. The resulting hexasome-nucleosome complexes are then resolved by Chd1. We present two cryoelectron microscopy (cryo-EM) structures of Chd1 bound to a hexasome-nucleosome complex before and after restoration of the missing inner H2A/H2B dimer by FACT. Chd1 uniquely interacts with the complex, positioning its ATPase domain to shift the hexasome away from the nucleosome. In the absence of the inner H2A/H2B dimer, its DNA-binding domain (DBD) packs against the ATPase domain, suggesting an inhibited state. Restoration of the dimer by FACT triggers a rearrangement that displaces the DBD and stimulates Chd1 remodeling. Our results demonstrate how chromatin remodelers interact with a complex nucleosome assembly and suggest how Chd1 and FACT jointly support transcription by RNAPII.


Subject(s)
Chromatin Assembly and Disassembly , Cryoelectron Microscopy , DNA-Binding Proteins , High Mobility Group Proteins , Histones , Nucleosomes , RNA Polymerase II , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription, Genetic , Transcriptional Elongation Factors , Nucleosomes/metabolism , Nucleosomes/genetics , Nucleosomes/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Histones/metabolism , Histones/genetics , Protein Binding , Models, Molecular , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics
5.
J Vis Exp ; (211)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39311556

ABSTRACT

Nucleosomes constitute the primary unit of eukaryotic chromatin and have been the focus of numerous informative single-molecule investigations regarding their biophysical properties and interactions with chromatin-binding proteins. Nucleosome reconstitution on DNA for these studies typically involves a salt dialysis procedure that provides precise control over the placement and number of nucleosomes formed along a DNA tether. However, this protocol is time-consuming and requires a substantial amount of DNA and histone octamers as inputs. To offer an alternative strategy, an in situ nucleosome reconstitution method for single-molecule force and fluorescence microscopy that utilizes the histone chaperone Nap1 is described. This method enables users to assemble nucleosomes on any DNA template without the need for strong nucleosome positioning sequences, adjust nucleosome density on demand, and use fewer reagents. In situ nucleosome formation occurs within seconds, offering a simpler experimental workflow and a convenient transition into single-molecule measurements. Examples of two downstream assays for probing nucleosome mechanics and visualizing the behavior of individual proteins on chromatin are further described.


Subject(s)
Microscopy, Fluorescence , Nucleosomes , Nucleosomes/chemistry , Nucleosomes/metabolism , Nucleosomes/genetics , Microscopy, Fluorescence/methods , Nucleosome Assembly Protein 1/chemistry , Nucleosome Assembly Protein 1/metabolism , Nucleosome Assembly Protein 1/genetics , Single Molecule Imaging/methods , DNA/chemistry , DNA/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry
6.
Commun Biol ; 7(1): 1144, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39277674

ABSTRACT

The nucleosome including H2A.B, a mammalian-specific H2A variant, plays pivotal roles in spermatogenesis, embryogenesis, and oncogenesis, indicating unique involvement in transcriptional regulation distinct from canonical H2A nucleosomes. Despite its significance, the exact regulatory mechanism remains elusive. This study utilized solid-state nanopores to investigate DNA unwinding dynamics, applying local force between DNA and histones. Comparative analysis of canonical H2A and H2A.B nucleosomes demonstrated that the H2A.B variant required a lower voltage for complete DNA unwinding. Furthermore, synchronization analysis and molecular dynamics simulations indicate that the H2A.B variant rapidly unwinds DNA, causing the H2A-H2B dimer to dissociate from DNA immediately upon disassembly of the histone octamer. In contrast, canonical H2A nucleosomes unwind DNA at a slower rate, suggesting that the H2A-H2B dimer undergoes a state of stacking at the pore. These findings suggest that nucleosomal DNA in the H2A.B nucleosomes undergoes a DNA unwinding process involving histone octamer disassembly distinct from that of canonical H2A nucleosomes, enabling smoother unwinding. The integrated approach of MD simulations and nanopore measurements is expected to evolve into a versatile tool for studying molecular interactions, not only within nucleosomes but also through the forced dissociation of DNA-protein complexes.


Subject(s)
DNA , Histones , Molecular Dynamics Simulation , Nucleosomes , Nucleosomes/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Histones/metabolism , Histones/chemistry , Histones/genetics , DNA/metabolism , DNA/chemistry , DNA/genetics , Animals , Nucleic Acid Conformation , Nanopores
7.
Commun Biol ; 7(1): 1086, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232115

ABSTRACT

Cell-free DNA (cfDNA) has emerged as a pivotal player in precision medicine, revolutionizing the diagnostic and therapeutic landscape. While its clinical applications have significantly increased in recent years, current cfDNA assays have limited ability to identify the active transcriptional programs that govern complex disease phenotypes and capture the heterogeneity of the disease. To address these limitations, we have developed a non-invasive platform to enrich and examine the active chromatin fragments (cfDNAac) in peripheral blood. The deconvolution of the cfDNAac signal from traditional nucleosomal chromatin fragments (cfDNAnuc) yields a catalog of features linking these circulating chromatin signals in blood to specific regulatory elements across the genome, including enhancers, promoters, and highly transcribed genes, mirroring the epigenetic data from the ENCODE project. Notably, these cfDNAac counts correlate strongly with RNA polymerase II activity and exhibit distinct expression patterns for known circadian genes. Additionally, cfDNAac signals across gene bodies and promoters show strong correlations with whole blood gene expression levels defined by GTEx. This study illustrates the utility of cfDNAac analysis for investigating epigenomics and gene expression, underscoring its potential for a wide range of clinical applications in precision medicine.


Subject(s)
Cell-Free Nucleic Acids , Chromatin , Chromatin/genetics , Chromatin/metabolism , Humans , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Promoter Regions, Genetic , Epigenesis, Genetic , Epigenomics/methods , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Nucleosomes/metabolism , Nucleosomes/genetics
8.
Nucleic Acids Res ; 52(17): 10194-10219, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39142653

ABSTRACT

The chromatin-remodeling enzyme helicase lymphoid-specific (HELLS) interacts with cell division cycle-associated 7 (CDCA7) on nucleosomes and is involved in the regulation of DNA methylation in higher organisms. Mutations in these genes cause immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome, which also results in DNA hypomethylation of satellite repeat regions. We investigated the functional domains of human CDCA7 in HELLS using several mutant CDCA7 proteins. The central region is critical for binding to HELLS, activation of ATPase, and nucleosome sliding activities of HELLS-CDCA7. The N-terminal region tends to inhibit ATPase activity. The C-terminal 4CXXC-type zinc finger domain contributes to CpG and hemimethylated CpG DNA preference for DNA-dependent HELLS-CDCA7 ATPase activity. Furthermore, CDCA7 showed a binding preference to DNA containing hemimethylated CpG, and replication-dependent pericentromeric heterochromatin foci formation of CDCA7 with HELLS was observed in mouse embryonic stem cells; however, all these phenotypes were lost in the case of an ICF syndrome mutant of CDCA7 mutated in the zinc finger domain. Thus, CDCA7 most likely plays a role in the recruitment of HELLS, activates its chromatin remodeling function, and efficiently induces DNA methylation, especially at hemimethylated replication sites.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases , DNA Methylation , Zinc Fingers , Humans , Animals , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/chemistry , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/metabolism , CpG Islands/genetics , DNA/metabolism , DNA/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Mutation , Protein Binding , Nucleosomes/metabolism , Nucleosomes/genetics , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Protein Domains , Mouse Embryonic Stem Cells/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Heterochromatin/metabolism , Heterochromatin/genetics , Face/abnormalities , Nuclear Proteins
9.
Mol Cell ; 84(18): 3455-3468.e6, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39208807

ABSTRACT

Mammalian gene expression is controlled by transcription factors (TFs) that engage sequence motifs in a chromatinized genome, where nucleosomes can restrict DNA access. Yet, how nucleosomes affect individual TFs remains unclear. Here, we measure the ability of over one hundred TF motifs to recruit TFs in a defined chromosomal locus in mouse embryonic stem cells. This identifies a set sufficient to enable the binding of TFs with diverse tissue specificities, functions, and DNA-binding domains. These chromatin-competent factors are further classified when challenged to engage motifs within a highly phased nucleosome. The pluripotency factors OCT4-SOX2 preferentially engage non-nucleosomal and entry-exit motifs, but not nucleosome-internal sites, a preference that also guides binding genome wide. By contrast, factors such as BANP, REST, or CTCF engage throughout, causing nucleosomal displacement. This supports that TFs vary widely in their sensitivity to nucleosomes and that genome access is TF specific and influenced by nucleosome position in the cell.


Subject(s)
Mouse Embryonic Stem Cells , Nucleosomes , Transcription Factors , Nucleosomes/metabolism , Nucleosomes/genetics , Animals , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Mouse Embryonic Stem Cells/metabolism , Binding Sites , Protein Binding , Genome/genetics , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Chromatin/metabolism , Chromatin/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Chromatin Assembly and Disassembly
10.
J Cell Biol ; 223(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39186086

ABSTRACT

Chromosome compaction is a key feature of mitosis and critical for accurate chromosome segregation. However, a precise quantitative analysis of chromosome geometry during mitotic progression is lacking. Here, we use volume electron microscopy to map, with nanometer precision, chromosomes from prometaphase through telophase in human RPE1 cells. During prometaphase, chromosomes acquire a smoother surface, their arms shorten, and the primary centromeric constriction is formed. The chromatin is progressively compacted, ultimately reaching a remarkable nucleosome concentration of over 750 µM in late prometaphase that remains relatively constant during metaphase and early anaphase. Surprisingly, chromosomes then increase their volume in late anaphase prior to deposition of the nuclear envelope. The plateau of total chromosome volume from late prometaphase through early anaphase described here is consistent with proposals that the final stages of chromatin condensation in mitosis involve a limit density, such as might be expected for a process involving phase separation.


Subject(s)
Anaphase , Nucleosomes , Prometaphase , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Nucleosomes/genetics , Humans , Cell Line , Chromosomes, Human/metabolism , Chromosomes, Human/genetics , Chromatin/metabolism , Chromatin/genetics , Mitosis , Centromere/metabolism , Centromere/ultrastructure , Centromere/genetics
11.
Mol Cell ; 84(17): 3209-3222.e5, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39191261

ABSTRACT

RNA polymerases must initiate and pause within a complex chromatin environment, surrounded by nucleosomes and other transcriptional machinery. This environment creates a spatial arrangement along individual chromatin fibers ripe for both competition and coordination, yet these relationships remain largely unknown owing to the inherent limitations of traditional structural and sequencing methodologies. To address this, we employed long-read chromatin fiber sequencing (Fiber-seq) in Drosophila to visualize RNA polymerase (Pol) within its native chromatin context with single-molecule precision along up to 30 kb fibers. We demonstrate that Fiber-seq enables the identification of individual Pol II, nucleosome, and transcription factor footprints, revealing Pol II pausing-driven destabilization of downstream nucleosomes. Furthermore, we demonstrate pervasive direct distance-dependent transcriptional coupling between nearby Pol II genes, Pol III genes, and transcribed enhancers, modulated by local chromatin architecture. Overall, transcription initiation reshapes surrounding nucleosome architecture and couples nearby transcriptional machinery along individual chromatin fibers.


Subject(s)
Chromatin , Drosophila melanogaster , Nucleosomes , Transcription, Genetic , Animals , Nucleosomes/metabolism , Nucleosomes/genetics , Chromatin/metabolism , Chromatin/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/enzymology , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Chromatin Assembly and Disassembly , RNA Polymerase III/metabolism , RNA Polymerase III/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics
12.
Mol Cell ; 84(17): 3175-3191.e8, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39096900

ABSTRACT

Heterochromatin enforces transcriptional gene silencing and can be epigenetically inherited, but the underlying mechanisms remain unclear. Here, we show that histone deacetylation, a conserved feature of heterochromatin domains, blocks SWI/SNF subfamily remodelers involved in chromatin unraveling, thereby stabilizing modified nucleosomes that preserve gene silencing. Histone hyperacetylation, resulting from either the loss of histone deacetylase (HDAC) activity or the direct targeting of a histone acetyltransferase to heterochromatin, permits remodeler access, leading to silencing defects. The requirement for HDAC in heterochromatin silencing can be bypassed by impeding SWI/SNF activity. Highlighting the crucial role of remodelers, merely targeting SWI/SNF to heterochromatin, even in cells with functional HDAC, increases nucleosome turnover, causing defective gene silencing and compromised epigenetic inheritance. This study elucidates a fundamental mechanism whereby histone hypoacetylation, maintained by high HDAC levels in heterochromatic regions, ensures stable gene silencing and epigenetic inheritance, providing insights into genome regulatory mechanisms relevant to human diseases.


Subject(s)
Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Silencing , Heterochromatin , Histone Deacetylases , Histones , Nucleosomes , Heterochromatin/metabolism , Heterochromatin/genetics , Nucleosomes/metabolism , Nucleosomes/genetics , Histones/metabolism , Histones/genetics , Acetylation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Humans , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Animals
13.
Mol Cell ; 84(15): 2856-2869.e9, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121843

ABSTRACT

RNA polymerase II (RNA Pol II)-mediated transcription is a critical, highly regulated process aided by protein complexes at distinct steps. Here, to investigate RNA Pol II and transcription-factor-binding and dissociation dynamics, we generated endogenous photoactivatable-GFP (PA-GFP) and HaloTag knockins using CRISPR-Cas9, allowing us to track a population of molecules at the induced Hsp70 loci in Drosophila melanogaster polytene chromosomes. We found that early in the heat-shock response, little RNA Pol II and DRB sensitivity-inducing factor (DSIF) are reused for iterative rounds of transcription. Surprisingly, although PAF1 and Spt6 are found throughout the gene body by chromatin immunoprecipitation (ChIP) assays, they show markedly different binding behaviors. Additionally, we found that PAF1 and Spt6 are only recruited after positive transcription elongation factor (P-TEFb)-mediated phosphorylation and RNA Pol II promoter-proximal pause escape. Finally, we observed that PAF1 may be expendable for transcription of highly expressed genes where nucleosome density is low. Thus, our live-cell imaging data provide key constraints to mechanistic models of transcription regulation.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , RNA Polymerase II , Transcription, Genetic , Transcriptional Elongation Factors , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , Positive Transcriptional Elongation Factor B/metabolism , Positive Transcriptional Elongation Factor B/genetics , Promoter Regions, Genetic , CRISPR-Cas Systems , Transcription Factors/metabolism , Transcription Factors/genetics , Polytene Chromosomes/genetics , Polytene Chromosomes/metabolism , Gene Expression Regulation , Phosphorylation , Protein Binding , Heat-Shock Response/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nucleosomes/metabolism , Nucleosomes/genetics
14.
PLoS Genet ; 20(8): e1011366, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102423

ABSTRACT

In Saccharomyces cerevisiae, the forkhead (Fkh) transcription factor Fkh1 (forkhead homolog) enhances the activity of many DNA replication origins that act in early S-phase (early origins). Current models posit that Fkh1 acts directly to promote these origins' activity by binding to origin-adjacent Fkh1 binding sites (FKH sites). However, the post-DNA binding functions that Fkh1 uses to promote early origin activity are poorly understood. Fkh1 contains a conserved FHA (forkhead associated) domain, a protein-binding module with specificity for phosphothreonine (pT)-containing partner proteins. At a small subset of yeast origins, the Fkh1-FHA domain enhances the ORC (origin recognition complex)-origin binding step, the G1-phase event that initiates the origin cycle. However, the importance of the Fkh1-FHA domain to either chromosomal replication or ORC-origin interactions at genome scale is unclear. Here, S-phase SortSeq experiments were used to compare genome replication in proliferating FKH1 and fkh1-R80A mutant cells. The Fkh1-FHA domain promoted the activity of ≈ 100 origins that act in early to mid- S-phase, including the majority of centromere-associated origins, while simultaneously inhibiting ≈ 100 late origins. Thus, in the absence of a functional Fkh1-FHA domain, the temporal landscape of the yeast genome was flattened. Origins are associated with a positioned nucleosome array that frames a nucleosome depleted region (NDR) over the origin, and ORC-origin binding is necessary but not sufficient for this chromatin organization. To ask whether the Fkh1-FHA domain had an impact on this chromatin architecture at origins, ORC ChIPSeq data generated from proliferating cells and MNaseSeq data generated from G1-arrested and proliferating cell populations were assessed. Origin groups that were differentially regulated by the Fkh1-FHA domain were characterized by distinct effects of this domain on ORC-origin binding and G1-phase chromatin. Thus, the Fkh1-FHA domain controlled the distinct chromatin architecture at early origins in G1-phase and regulated origin activity in S-phase.


Subject(s)
Chromatin , DNA Replication , G1 Phase , Origin Recognition Complex , Replication Origin , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Replication Origin/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , DNA Replication/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Chromatin/genetics , Chromatin/metabolism , Origin Recognition Complex/genetics , Origin Recognition Complex/metabolism , G1 Phase/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , S Phase/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Protein Domains/genetics , Binding Sites , Protein Binding , Chromosomes, Fungal/genetics , Chromosomes, Fungal/metabolism , Nucleosomes/metabolism , Nucleosomes/genetics
15.
Sci Adv ; 10(34): eadp5753, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178260

ABSTRACT

Mutations of the SNF2 family ATPase HELLS and its activator CDCA7 cause immunodeficiency, centromeric instability, and facial anomalies syndrome, characterized by DNA hypomethylation at heterochromatin. It remains unclear why CDCA7-HELLS is the sole nucleosome remodeling complex whose deficiency abrogates the maintenance of DNA methylation. We here identify the unique zinc-finger domain of CDCA7 as an evolutionarily conserved hemimethylation-sensing zinc finger (HMZF) domain. Cryo-electron microscopy structural analysis of the CDCA7-nucleosome complex reveals that the HMZF domain can recognize hemimethylated CpG in the outward-facing DNA major groove within the nucleosome core particle, whereas UHRF1, the critical activator of the maintenance methyltransferase DNMT1, cannot. CDCA7 recruits HELLS to hemimethylated chromatin and facilitates UHRF1-mediated H3 ubiquitylation associated with replication-uncoupled maintenance DNA methylation. We propose that the CDCA7-HELLS nucleosome remodeling complex assists the maintenance of DNA methylation on chromatin by sensing hemimethylated CpG that is otherwise inaccessible to UHRF1 and DNMT1.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA Methylation , Nucleosomes , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Nucleosomes/metabolism , Nucleosomes/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cryoelectron Microscopy , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/chemistry , CpG Islands , Ubiquitination , Evolution, Molecular , DNA/metabolism , DNA/chemistry , DNA/genetics , Zinc Fingers , Chromatin/metabolism , Chromatin/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/chemistry , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/chemistry , Eukaryota/genetics , Eukaryota/metabolism , Protein Binding , Histones/metabolism , Histones/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/chemistry
16.
Nat Commun ; 15(1): 7386, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39191772

ABSTRACT

Germline pathogenic TP53 variants predispose individuals to a high lifetime risk of developing multiple cancers and are the hallmark feature of Li-Fraumeni syndrome (LFS). Our group has previously shown that LFS patients harbor shorter plasma cell-free DNA fragmentation; independent of cancer status. To understand the functional underpinning of cfDNA fragmentation in LFS, we conducted a fragmentomic analysis of 199 cfDNA samples from 82 TP53 mutation carriers and 30 healthy TP53-wildtype controls. We find that LFS individuals exhibit an increased prevalence of A/T nucleotides at fragment ends, dysregulated nucleosome positioning at p53 binding sites, and loci-specific changes in chromatin accessibility at development-associated transcription factor binding sites and at cancer-associated open chromatin regions. Machine learning classification resulted in robust differentiation between TP53 mutant versus wildtype cfDNA samples (AUC-ROC = 0.710-1.000) and intra-patient longitudinal analysis of ctDNA fragmentation signal enabled early cancer detection. These results suggest that cfDNA fragmentation may be a useful diagnostic tool in LFS patients and provides an important baseline for cancer early detection.


Subject(s)
Cell-Free Nucleic Acids , DNA Fragmentation , Germ-Line Mutation , Li-Fraumeni Syndrome , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Male , Female , Li-Fraumeni Syndrome/genetics , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/blood , Adult , Young Adult , Middle Aged , Circulating Tumor DNA/genetics , Circulating Tumor DNA/blood , Adolescent , Neoplasms/genetics , Neoplasms/pathology , Chromatin/genetics , Chromatin/metabolism , Machine Learning , Heterozygote , Child , Nucleosomes/metabolism , Nucleosomes/genetics , Early Detection of Cancer
17.
Mol Cell ; 84(16): 3011-3025.e7, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39116874

ABSTRACT

The histone variant macroH2A is generally linked to transcriptionally inactive chromatin, but how macroH2A regulates chromatin structure and functions in the transcriptional process remains elusive. This study reveals that while the integration of human macroH2A1.2 into nucleosomes does not affect their stability or folding dynamics, it notably hinders the maintenance of facilitates chromatin transcription's (FACT's) function. We show that FACT effectively diminishes the stability of macroH2A1.2-nucleosomes and expedites their depletion subsequent to the initial unfolding process. Furthermore, we identify the residue S139 in macroH2A1.2 as a critical switch to modulate FACT's function in nucleosome maintenance. Genome-wide analyses demonstrate that FACT-mediated depletion of macroH2A-nucleosomes allows the correct localization of macroH2A, while the S139 mutation reshapes macroH2A distribution and influences stimulation-induced transcription and cellular response in macrophages. Our findings provide mechanistic insights into the intricate interplay between macroH2A and FACT at the nucleosome level and elucidate their collective role in transcriptional regulation and immune response of macrophages.


Subject(s)
Histones , Nucleosomes , Transcription, Genetic , Transcriptional Elongation Factors , Humans , Nucleosomes/metabolism , Nucleosomes/genetics , Histones/metabolism , Histones/genetics , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Macrophages/metabolism , Mutation , Chromatin Assembly and Disassembly , Mice , Chromatin/metabolism , Chromatin/genetics , Gene Expression Regulation , RAW 264.7 Cells , Protein Binding , HEK293 Cells
18.
Mol Cell ; 84(16): 3061-3079.e10, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39121853

ABSTRACT

Mouse FOXA1 and GATA4 are prototypes of pioneer factors, initiating liver cell development by binding to the N1 nucleosome in the enhancer of the ALB1 gene. Using cryoelectron microscopy (cryo-EM), we determined the structures of the free N1 nucleosome and its complexes with FOXA1 and GATA4, both individually and in combination. We found that the DNA-binding domains of FOXA1 and GATA4 mainly recognize the linker DNA and an internal site in the nucleosome, respectively, whereas their intrinsically disordered regions interact with the acidic patch on histone H2A-H2B. FOXA1 efficiently enhances GATA4 binding by repositioning the N1 nucleosome. In vivo DNA editing and bioinformatics analyses suggest that the co-binding mode of FOXA1 and GATA4 plays important roles in regulating genes involved in liver cell functions. Our results reveal the mechanism whereby FOXA1 and GATA4 cooperatively bind to the nucleosome through nucleosome repositioning, opening chromatin by bending linker DNA and obstructing nucleosome packing.


Subject(s)
Cryoelectron Microscopy , GATA4 Transcription Factor , Hepatocyte Nuclear Factor 3-alpha , Nucleosomes , Protein Binding , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Nucleosomes/metabolism , Nucleosomes/genetics , Nucleosomes/ultrastructure , Animals , GATA4 Transcription Factor/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/chemistry , Mice , Chromatin/metabolism , Chromatin/genetics , Histones/metabolism , Histones/genetics , Histones/chemistry , Binding Sites , DNA/metabolism , DNA/genetics , DNA/chemistry , Chromatin Assembly and Disassembly , Humans
19.
BMC Res Notes ; 17(1): 219, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103906

ABSTRACT

OBJECTIVE: In past work in budding yeast, we identified a nucleosomal region required for proper interactions between the histone chaperone complex yFACT and transcribed genes. Specific histone mutations within this region cause a shift in yFACT occupancy towards the 3' end of genes, a defect that we have attributed to impaired yFACT dissociation from DNA following transcription. In this work we wished to assess the contributions of DNA sequences at the 3' end of genes in promoting yFACT dissociation upon transcription termination. RESULTS: We generated fourteen different alleles of the constitutively expressed yeast gene PMA1, each lacking a distinct DNA fragment across its 3' end, and assessed their effects on occupancy of the yFACT component Spt16. Whereas most of these alleles conferred no defects on Spt16 occupancy, one did cause a modest increase in Spt16 binding at the gene's 3' end. Interestingly, the same allele also caused minor retention of RNA Polymerase II (Pol II) and altered nucleosome occupancy across the same region of the gene. These results suggest that specific DNA sequences at the 3' ends of genes can play roles in promoting efficient yFACT and Pol II dissociation from genes and can also contribute to proper chromatin architecture.


Subject(s)
Nucleosomes , RNA Polymerase II , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Nucleosomes/metabolism , Nucleosomes/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , Histone Chaperones/genetics , Histone Chaperones/metabolism , DNA, Fungal/genetics , DNA, Fungal/metabolism , Alleles , Base Sequence , Gene Expression Regulation, Fungal , Transcription, Genetic
20.
Mol Cell ; 84(16): 3080-3097.e9, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39043178

ABSTRACT

Alternative transcription start sites can affect transcript isoform diversity and translation levels. In a recently described form of gene regulation, coordinated transcriptional and translational interference results in transcript isoform-dependent changes in protein expression. Specifically, a long undecoded transcript isoform (LUTI) is transcribed from a gene-distal promoter, interfering with expression of the gene-proximal promoter. Although transcriptional and chromatin features associated with LUTI expression have been described, the mechanism underlying LUTI-based transcriptional interference is not well understood. Using an unbiased genetic approach followed by functional genomics, we uncovered that the Swi/Snf chromatin remodeling complex is required for co-transcriptional nucleosome remodeling that leads to LUTI-based repression. We identified genes with tandem promoters that rely on Swi/Snf function for transcriptional interference during protein folding stress, including LUTI-regulated genes. This study provides clear evidence for Swi/Snf playing a direct role in gene repression via a cis transcriptional interference mechanism.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone , Nucleosomes , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , Transcription, Genetic , Transcription Factors/metabolism , Transcription Factors/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Nucleosomes/metabolism , Nucleosomes/genetics , Gene Expression Regulation, Fungal , Transcription Initiation Site , Chromatin/metabolism , Chromatin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL