Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 266
Filter
Add more filters

Country/Region as subject
Publication year range
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 ; 185(17): 3083-3085, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35985286

ABSTRACT

In some plants and animals, microtubules attach across the length of the chromosome in mitosis, forming a holocentromere instead of a single centromeric locus. A new study in Cell shows that in the holocentric beak sedge Rhynchospora, holocentromeres also impact genomic architecture, epigenome organization, and karyotype evolution.


Subject(s)
Centromere , Cyperaceae , Animals , Centromere/genetics , Cyperaceae/genetics , Microtubules , Mitosis , Plants/genetics
3.
Immunity ; 57(2): 287-302.e12, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38354704

ABSTRACT

The interaction of the tumor necrosis factor receptor (TNFR) family member CD27 on naive CD8+ T (Tn) cells with homotrimeric CD70 on antigen-presenting cells (APCs) is necessary for T cell memory fate determination. Here, we examined CD27 signaling during Tn cell activation and differentiation. In conjunction with T cell receptor (TCR) stimulation, ligation of CD27 by a synthetic trimeric CD70 ligand triggered CD27 internalization and degradation, suggesting active regulation of this signaling axis. Internalized CD27 recruited the signaling adaptor TRAF2 and the phosphatase SHP-1, thereby modulating TCR and CD28 signals. CD27-mediated modulation of TCR signals promoted transcription factor circuits that induced memory rather than effector associated gene programs, which are induced by CD28 costimulation. CD27-costimulated chimeric antigen receptor (CAR)-engineered T cells exhibited improved tumor control compared with CD28-costimulated CAR-T cells. Thus, CD27 signaling during Tn cell activation promotes memory properties with relevance to T cell immunotherapy.


Subject(s)
CD28 Antigens , Gene Regulatory Networks , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , CD28 Antigens/metabolism , Signal Transduction , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , CD27 Ligand/genetics , CD27 Ligand/metabolism , CD8-Positive T-Lymphocytes
4.
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
5.
Cell ; 165(3): 580-92, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27062929

ABSTRACT

Every nucleosome across the genome must be disrupted and reformed when the replication fork passes, but how chromatin organization is re-established following replication is unknown. To address this problem, we have developed Mapping In vivo Nascent Chromatin with EdU and sequencing (MINCE-seq) to characterize the genome-wide location of nucleosomes and other chromatin proteins behind replication forks at high temporal and spatial resolution. We find that the characteristic chromatin landscape at Drosophila promoters and enhancers is lost upon replication. The most conspicuous changes are at promoters that have high levels of RNA polymerase II (RNAPII) stalling and DNA accessibility and show specific enrichment for the BRM remodeler. Enhancer chromatin is also disrupted during replication, suggesting a role for transcription factor (TF) competition in nucleosome re-establishment. Thus, the characteristic nucleosome landscape emerges from a uniformly packaged genome by the action of TFs, RNAPII, and remodelers minutes after replication fork passage.


Subject(s)
DNA Replication , Nucleosomes/metabolism , S Phase , Transcription Factors/metabolism , Transcription, Genetic , Animals , Chromatin/metabolism , Drosophila melanogaster , Promoter Regions, Genetic , RNA Polymerase II/metabolism
6.
Mol Cell ; 82(23): 4458-4470.e5, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36370708

ABSTRACT

The two doublet histones of Marseillevirus are distantly related to the four eukaryotic core histones and wrap 121 base pairs of DNA to form remarkably similar nucleosomes. By permeabilizing Marseillevirus virions and performing genome-wide nuclease digestion, chemical cleavage, and mass spectrometry assays, we find that the higher-order organization of Marseillevirus chromatin fundamentally differs from that of eukaryotes. Marseillevirus nucleosomes fully protect DNA within virions as closely abutted 121-bp DNA-wrapped cores without linker DNA or phasing along genes. Likewise, we observed that nucleosomes reconstituted onto multi-copy tandem repeats of a nucleosome-positioning sequence are tightly packed. Dense promiscuous packing of fully wrapped nucleosomes rather than "beads on a string" with genic punctuation represents a distinct mode of DNA packaging by histones. We suggest that doublet histones have evolved for viral genome protection and may resemble an early stage of histone differentiation leading to the eukaryotic octameric nucleosome.


Subject(s)
Giant Viruses , Nucleosomes , Nucleosomes/genetics , Histones/genetics , Giant Viruses/genetics , DNA/genetics , Virion/genetics , Genome, Viral
7.
Nat Rev Mol Cell Biol ; 18(2): 115-126, 2017 02.
Article in English | MEDLINE | ID: mdl-27924075

ABSTRACT

Most histones are assembled into nucleosomes behind the replication fork to package newly synthesized DNA. By contrast, histone variants, which are encoded by separate genes, are typically incorporated throughout the cell cycle. Histone variants can profoundly change chromatin properties, which in turn affect DNA replication and repair, transcription, and chromosome packaging and segregation. Recent advances in the study of histone replacement have elucidated the dynamic processes by which particular histone variants become substrates of histone chaperones, ATP-dependent chromatin remodellers and histone-modifying enzymes. Here, we review histone variant dynamics and the effects of replacing DNA synthesis-coupled histones with their replication-independent variants on the chromatin landscape.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Animals , Chromatin/genetics , DNA Replication , Genetic Variation , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/genetics , Humans , Nucleosomes/metabolism , Plants/genetics , Transcription, Genetic , Yeasts/genetics
8.
Trends Biochem Sci ; 48(12): 1071-1082, 2023 12.
Article in English | MEDLINE | ID: mdl-37777391

ABSTRACT

Giant viruses (Nucleocytoviricota) have a largely conserved lifecycle, yet how they cram their large genomes into viral capsids is mostly unknown. The major capsid protein and the packaging ATPase (pATPase) comprise a highly conserved morphogenesis module in giant viruses, yet some giant viruses dispense with an icosahedral capsid, and others encode multiple versions of pATPases, including conjoined ATPase doublets, or encode none. Some giant viruses have acquired DNA-condensing proteins to compact their genomes, including sheath-like structures encasing folded DNA or densely packed viral nucleosomes that show a resemblance to eukaryotic nucleosomes at the telomeres. Here, we review what is known and unknown about these ATPases and condensing proteins, and place these variations in the context of viral lifecycles.


Subject(s)
Nucleosomes , Viral Genome Packaging , Capsid Proteins/chemistry , Capsid Proteins/genetics , DNA , Adenosine Triphosphatases/genetics , Genome, Viral , Virus Assembly/genetics
9.
Mol Cell ; 73(2): 238-249.e3, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30554944

ABSTRACT

The classic view of nucleosome organization at active promoters is that two well-positioned nucleosomes flank a nucleosome-depleted region (NDR). However, this view has been recently disputed by contradictory reports as to whether wider (≳150 bp) NDRs instead contain unstable, micrococcal nuclease-sensitive ("fragile") nucleosomal particles. To determine the composition of fragile particles, we introduce CUT&RUN.ChIP, in which targeted nuclease cleavage and release is followed by chromatin immunoprecipitation. We find that fragile particles represent the occupancy of the RSC (remodeling the structure of chromatin) nucleosome remodeling complex and RSC-bound, partially unwrapped nucleosomal intermediates. We also find that general regulatory factors (GRFs) bind to partially unwrapped nucleosomes at these promoters. We propose that RSC binding and its action cause nucleosomes to unravel, facilitate subsequent binding of GRFs, and constitute a dynamic cycle of nucleosome deposition and clearance at the subset of wide Pol II promoter NDRs.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation/methods , DNA, Fungal/metabolism , DNA-Binding Proteins/metabolism , Micrococcal Nuclease/metabolism , Nucleosomes/enzymology , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Transcription Factors/metabolism , Binding Sites , DNA, Fungal/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , Nucleic Acid Conformation , Nucleosomes/genetics , Protein Binding , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
11.
Mol Cell ; 75(3): 562-575.e5, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31253573

ABSTRACT

Although the in vitro structural and in vivo spatial characteristics of transcription factor (TF) binding are well defined, TF interactions with chromatin and other companion TFs during development are poorly understood. To analyze such interactions in vivo, we profiled several TFs across a time course of human embryonic stem cell differentiation and studied their interactions with nucleosomes and co-occurring TFs by enhanced chromatin occupancy (EChO), a computational strategy for classifying TF interactions with chromatin. EChO shows that multiple individual TFs can employ either direct DNA binding or "pioneer" nucleosome binding at different enhancer targets. Nucleosome binding is not exclusively confined to inaccessible chromatin but rather correlated with local binding of other TFs and degeneracy at key bases in the pioneer factor target motif responsible for direct DNA binding. Our strategy reveals a dynamic exchange of TFs at enhancers across developmental time that is aided by pioneer nucleosome binding.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Transcription Factors/genetics , Binding Sites , DNA-Binding Proteins/genetics , Humans , Nucleosomes/genetics
12.
Genes Dev ; 33(17-18): 1252-1264, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31395740

ABSTRACT

Although MAX is regarded as an obligate dimerization partner for MYC, its function in normal development and neoplasia is poorly defined. We show that B-cell-specific deletion of Max has a modest effect on B-cell development but completely abrogates Eµ-Myc-driven lymphomagenesis. While Max loss affects only a few hundred genes in normal B cells, it leads to the global down-regulation of Myc-activated genes in premalignant Eµ-Myc cells. We show that the balance between MYC-MAX and MNT-MAX interactions in B cells shifts in premalignant B cells toward a MYC-driven transcriptional program. Moreover, we found that MAX loss leads to a significant reduction in MYC protein levels and down-regulation of direct transcriptional targets, including regulators of MYC stability. This phenomenon is also observed in multiple cell lines treated with MYC-MAX dimerization inhibitors. Our work uncovers a layer of Myc autoregulation critical for lymphomagenesis yet partly dispensable for normal development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Active Transport, Cell Nucleus , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Kynurenine/genetics , Kynurenine/metabolism , Lymphoma/physiopathology , Mice , Organoids/growth & development , Organoids/physiopathology , Oximes/pharmacology , Sulfonamides/pharmacology
13.
Genome Res ; 33(1): 1-17, 2023 01.
Article in English | MEDLINE | ID: mdl-36650052

ABSTRACT

Vertebrate genomes are partitioned into chromatin domains or topologically associating domains (TADs), which are typically bound by head-to-head pairs of CTCF binding sites. Transcription at domain boundaries correlates with better insulation; however, it is not known whether the boundary transcripts themselves contribute to boundary function. Here we characterize boundary-associated RNAs genome-wide, focusing on the disease-relevant INK4a/ARF and MYC TAD. Using CTCF site deletions and boundary-associated RNA knockdowns, we observe that boundary-associated RNAs facilitate recruitment and clustering of CTCF at TAD borders. The resulting CTCF enrichment enhances TAD insulation, enhancer-promoter interactions, and TAD gene expression. Importantly, knockdown of boundary-associated RNAs results in loss of boundary insulation function. Using enhancer deletions and CRISPRi of promoters, we show that active TAD enhancers, but not promoters, induce boundary-associated RNA transcription, thus defining a novel class of regulatory enhancer RNAs.


Subject(s)
Chromatin , RNA , Chromatin/genetics , CCCTC-Binding Factor/metabolism , Binding Sites , Promoter Regions, Genetic , Enhancer Elements, Genetic
14.
Mol Cell ; 71(2): 193-194, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30028998

ABSTRACT

Two papers in Molecular Cell (Kubik et al., 2018; Yan et al., 2018) explore the mechanisms by which transcription factors bind their sites in chromatin, providing fresh insights into the much-debated question of how transcription factors can be "pioneers."


Subject(s)
Saccharomycetales , Transcription Factors/genetics , Chromatin , Chromatin Assembly and Disassembly , Nucleosomes
15.
Genes Dev ; 32(1): 20-25, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29386331

ABSTRACT

We combined classical salt fractionation with chromatin immunoprecipitation to recover human centromeric chromatin under native conditions. We found that >85% of the total centromeric chromatin is insoluble under conditions typically used for native chromatin extraction. To map both soluble and insoluble chromatin in situ, we combined CUT&RUN (cleavage under targets and release using nuclease), a targeted nuclease method, with salt fractionation. Using this approach, we observed unexpected structural and conformational variations of centromere protein A (CENP-A)-containing complexes on different α-satellite dimeric units within highly homogenous arrays. Our results suggest that slight α-satellite sequence differences control the structure and occupancy of the associated centromeric chromatin complex.


Subject(s)
Centromere Protein A/chemistry , Centromere/chemistry , Chromatin/chemistry , Centromere Protein A/isolation & purification , Centromere Protein A/metabolism , Centromere Protein B/chemistry , Centromere Protein B/metabolism , Chemical Fractionation , Chromatin/isolation & purification , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , DNA, Satellite/chemistry , Humans , K562 Cells , Solubility
16.
Genome Res ; 32(4): 608-615, 2022 04.
Article in English | MEDLINE | ID: mdl-35361623

ABSTRACT

Centromeres, the chromosomal loci where spindle fibers attach during cell division to segregate chromosomes, are typically found within satellite arrays in plants and animals. Satellite arrays have been difficult to analyze because they comprise megabases of tandem head-to-tail highly repeated DNA sequences. Much evidence suggests that centromeres are epigenetically defined by the location of nucleosomes containing the centromere-specific histone H3 variant cenH3, independently of the DNA sequences where they are located; however, the reason that cenH3 nucleosomes are generally found on rapidly evolving satellite arrays has remained unclear. Recently, long-read sequencing technology has clarified the structures of satellite arrays and sparked rethinking of how they evolve, and new experiments and analyses have helped bring both understanding and further speculation about the role these highly repeated sequences play in centromere identification.


Subject(s)
Centromere , Nucleosomes , Centromere/genetics , Epigenesis, Genetic , Epigenomics , Histones/genetics , Histones/metabolism , Nucleosomes/genetics
17.
Nat Methods ; 19(11): 1383-1392, 2022 11.
Article in English | MEDLINE | ID: mdl-36192462

ABSTRACT

Whereas techniques to map chromatin-bound proteins are well developed, mapping chromatin-associated RNAs remains a challenge. Here, we describe Reverse Transcribe and Tagment (RT&Tag), in which RNAs associated with a chromatin epitope are targeted by an antibody followed by a protein A-Tn5 transposome. Localized reverse transcription generates RNA/cDNA hybrids that are subsequently tagmented by Tn5 transposases for downstream sequencing. We demonstrate the utility of RT&Tag in Drosophila cells for capturing the noncoding RNA roX2 with the dosage compensation complex and maturing transcripts associated with silencing histone modifications. We also show that RT&Tag can detect N6-methyladenosine-modified mRNAs, and show that genes producing methylated transcripts are characterized by extensive promoter pausing of RNA polymerase II. The high efficiency of in situ antibody tethering and tagmentation makes RT&Tag especially suitable for rapid low-cost profiling of chromatin-associated RNAs.


Subject(s)
Chromatin , RNA , Animals , Chromatin/genetics , RNA/genetics , Histone Code , Drosophila/genetics , DNA, Complementary , Antibodies
18.
Nat Rev Genet ; 20(5): 283-297, 2019 05.
Article in English | MEDLINE | ID: mdl-30886348

ABSTRACT

Sophisticated gene-regulatory mechanisms probably evolved in prokaryotes billions of years before the emergence of modern eukaryotes, which inherited the same basic enzymatic machineries. However, the epigenomic landscapes of eukaryotes are dominated by nucleosomes, which have acquired roles in genome packaging, mitotic condensation and silencing parasitic genomic elements. Although the molecular mechanisms by which nucleosomes are displaced and modified have been described, just how transcription factors, histone variants and modifications and chromatin regulators act on nucleosomes to regulate transcription is the subject of considerable ongoing study. We explore the extent to which these transcriptional regulatory components function in the context of the evolutionarily ancient role of chromatin as a barrier to processes acting on DNA and how chromatin proteins have diversified to carry out evolutionarily recent functions that accompanied the emergence of differentiation and development in multicellular eukaryotes.


Subject(s)
Chromatin Assembly and Disassembly , DNA/genetics , Genome , Nucleosomes/genetics , Transcription, Genetic , Animals , Biological Evolution , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/history , Chromosomal Proteins, Non-Histone/metabolism , DNA/history , DNA/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Genomics/methods , Histones/genetics , Histones/history , Histones/metabolism , History, 21st Century , History, Ancient , Humans , Nucleosomes/chemistry , Nucleosomes/metabolism , Prokaryotic Cells/cytology , Prokaryotic Cells/metabolism , Transcription Factors/genetics , Transcription Factors/history , Transcription Factors/metabolism
19.
Mol Cell ; 68(6): 1038-1053.e4, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29225036

ABSTRACT

Nucleosomes are disrupted during transcription and other active processes, but the structural intermediates during nucleosome disruption in vivo are unknown. To identify intermediates, we mapped subnucleosomal protections in Drosophila cells using Micrococcal Nuclease followed by sequencing. At the first nucleosome position downstream of the transcription start site, we identified unwrapped intermediates, including hexasomes that lack either proximal or distal contacts. Inhibiting topoisomerases or depleting histone chaperones increased unwrapping, whereas inhibiting release of paused RNAPII or reducing RNAPII elongation decreased unwrapping. Our results indicate that positive torsion generated by elongating RNAPII causes transient loss of histone-DNA contacts. Using this mapping approach, we found that nucleosomes flanking human CTCF insulation sites are similarly disrupted. We also identified diagnostic subnucleosomal particle remnants in cell-free human DNA data as a relic of transcribed genes from apoptosing cells. Thus identification of subnucleosomal fragments from nuclease protection data represents a general strategy for structural epigenomics.


Subject(s)
Chromatin Assembly and Disassembly , Drosophila melanogaster/metabolism , Epigenomics , Gene Expression Regulation , Histones/chemistry , Nucleosomes/chemistry , Transcription, Genetic , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Histones/genetics , Histones/metabolism , Micrococcal Nuclease/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription Initiation Site
20.
Trends Biochem Sci ; 45(1): 13-26, 2020 01.
Article in English | MEDLINE | ID: mdl-31630896

ABSTRACT

Gene regulation in eukaryotes requires the controlled access of sequence-specific transcription factors (TFs) to their sites in a chromatin landscape dominated by nucleosomes. Nucleosomes are refractory to TF binding, and often must be removed from regulatory regions. Recent genomic studies together with in vitro measurements suggest that the nucleosome barrier to TF binding is modulated by dynamic nucleosome unwrapping governed by ATP-dependent chromatin remodelers. Genome-wide occupancy and the regulation of subnucleosomal intermediates have gained recent attention with the application of high-resolution approaches for precision mapping of protein-DNA interactions. We summarize here recent findings on nucleosome substructures and TF binding dynamics, and highlight how unwrapped nucleosomal intermediates provide a novel signature of active chromatin.


Subject(s)
Epigenome/genetics , Nucleosomes/genetics , Nucleosomes/metabolism , Humans , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL