Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.742
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 89: 159-187, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32176523

ABSTRACT

This review focuses on imaging DNA and single RNA molecules in living cells to define eukaryotic functional organization and dynamic processes. The latest advances in technologies to visualize individual DNA loci and RNAs in real time are discussed. Single-molecule fluorescence microscopy provides the spatial and temporal resolution to reveal mechanisms regulating fundamental cell functions. Novel insights into the regulation of nuclear architecture, transcription, posttranscriptional RNA processing, and RNA localization provided by multicolor fluorescence microscopy are reviewed. A perspective on the future use of live imaging technologies and overcoming their current limitations is provided.


Subject(s)
Cell Nucleus/ultrastructure , Chromatin/ultrastructure , DNA/ultrastructure , Gene Expression Regulation , RNA, Messenger/ultrastructure , RNA, Small Untranslated/ultrastructure , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/genetics , DNA/metabolism , DNA Replication , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Humans , Microscopy, Fluorescence , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Single Molecule Imaging/instrumentation , Single Molecule Imaging/methods , Staining and Labeling/methods , Telomere/metabolism , Telomere/ultrastructure , Transcription, Genetic
2.
Cell ; 176(3): 520-534.e25, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30661750

ABSTRACT

Elucidating the global and local rules that govern genome-wide, hierarchical chromatin architecture remains a critical challenge. Current high-throughput chromosome conformation capture (Hi-C) technologies have identified large-scale chromatin structural motifs, such as topologically associating domains and looping. However, structural rules at the smallest or nucleosome scale remain poorly understood. Here, we coupled nucleosome-resolved Hi-C technology with simulated annealing-molecular dynamics (SA-MD) simulation to reveal 3D spatial distributions of nucleosomes and their genome-wide orientation in chromatin. Our method, called Hi-CO, revealed distinct nucleosome folding motifs across the yeast genome. Our results uncovered two types of basic secondary structural motifs in nucleosome folding: α-tetrahedron and ß-rhombus analogous to α helix and ß sheet motifs in protein folding. Using mutants and cell-cycle-synchronized cells, we further uncovered motifs with specific nucleosome positioning and orientation coupled to epigenetic features at individual loci. By illuminating molecular-level structure-function relationships in eukaryotic chromatin, our findings establish organizational principles of nucleosome folding.


Subject(s)
Chromatin/ultrastructure , Nucleosomes/ultrastructure , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Chromosomes/metabolism , Chromosomes/ultrastructure , Nucleosomes/genetics , Nucleosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Initiation Site
3.
Cell ; 170(6): 1045-1047, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28886374

ABSTRACT

It's the stuff of life, and we're fascinated by DNA and how it's packaged into chromatin and compacted into chromosomes. Advances in looking at chromatin organization in cells are letting us see this polymer, its packing, and its function with fresh eyes.


Subject(s)
Chromatin/chemistry , Chromatin/ultrastructure , Cryoelectron Microscopy/methods , DNA Packaging , Tomography/methods , Animals , Cell Nucleus/chemistry , Cell Nucleus/ultrastructure , Chromosomes/chemistry , Chromosomes/ultrastructure , DNA/chemistry , Humans , Nucleosomes/chemistry
4.
Nature ; 627(8005): 890-897, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38448592

ABSTRACT

In eukaryotes, DNA compacts into chromatin through nucleosomes1,2. Replication of the eukaryotic genome must be coupled to the transmission of the epigenome encoded in the chromatin3,4. Here we report cryo-electron microscopy structures of yeast (Saccharomyces cerevisiae) replisomes associated with the FACT (facilitates chromatin transactions) complex (comprising Spt16 and Pob3) and an evicted histone hexamer. In these structures, FACT is positioned at the front end of the replisome by engaging with the parental DNA duplex to capture the histones through the middle domain and the acidic carboxyl-terminal domain of Spt16. The H2A-H2B dimer chaperoned by the carboxyl-terminal domain of Spt16 is stably tethered to the H3-H4 tetramer, while the vacant H2A-H2B site is occupied by the histone-binding domain of Mcm2. The Mcm2 histone-binding domain wraps around the DNA-binding surface of one H3-H4 dimer and extends across the tetramerization interface of the H3-H4 tetramer to the binding site of Spt16 middle domain before becoming disordered. This arrangement leaves the remaining DNA-binding surface of the other H3-H4 dimer exposed to additional interactions for further processing. The Mcm2 histone-binding domain and its downstream linker region are nested on top of Tof1, relocating the parental histones to the replisome front for transfer to the newly synthesized lagging-strand DNA. Our findings offer crucial structural insights into the mechanism of replication-coupled histone recycling for maintaining epigenetic inheritance.


Subject(s)
Chromatin , DNA Replication , Epistasis, Genetic , Histones , Saccharomyces cerevisiae , Binding Sites , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Cryoelectron Microscopy , DNA Replication/genetics , DNA, Fungal/biosynthesis , DNA, Fungal/chemistry , DNA, Fungal/metabolism , DNA, Fungal/ultrastructure , Epistasis, Genetic/genetics , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Multienzyme Complexes/ultrastructure , Nucleosomes/chemistry , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Protein Binding , Protein Domains , Protein Multimerization , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure
5.
Nat Rev Mol Cell Biol ; 17(12): 743-755, 2016 12.
Article in English | MEDLINE | ID: mdl-27580841

ABSTRACT

Chromosomes of eukaryotes adopt highly dynamic and complex hierarchical structures in the nucleus. The three-dimensional (3D) organization of chromosomes profoundly affects DNA replication, transcription and the repair of DNA damage. Thus, a thorough understanding of nuclear architecture is fundamental to the study of nuclear processes in eukaryotic cells. Recent years have seen rapid proliferation of technologies to investigate genome organization and function. Here, we review experimental and computational methodologies for 3D genome analysis, with special focus on recent advances in high-throughput chromatin conformation capture (3C) techniques and data analysis.


Subject(s)
Chromatin/ultrastructure , Animals , Chromosome Mapping , Chromosomes/ultrastructure , Computer Simulation , Humans , Models, Molecular
6.
Nature ; 609(7929): 1048-1055, 2022 09.
Article in English | MEDLINE | ID: mdl-36104563

ABSTRACT

Telomeres, the ends of eukaryotic chromosomes, play pivotal parts in ageing and cancer and are targets of DNA damage and the DNA damage response1-5. Little is known about the structure of telomeric chromatin at the molecular level. Here we used negative stain electron microscopy and single-molecule magnetic tweezers to characterize 3-kbp-long telomeric chromatin fibres. We also obtained the cryogenic electron microscopy structure of the condensed telomeric tetranucleosome and its dinucleosome unit. The structure displayed close stacking of nucleosomes with a columnar arrangement, and an unusually short nucleosome repeat  length that comprised about 132 bp DNA wound in a continuous superhelix around histone octamers. This columnar structure is primarily stabilized by the H2A carboxy-terminal and histone amino-terminal tails in a synergistic manner. The columnar conformation results in exposure of the DNA helix, which may make it susceptible to both DNA damage and the DNA damage response. The conformation also exists in an alternative open state, in which one nucleosome is unstacked and flipped out, which exposes the acidic patch of the histone surface. The structural features revealed in this work suggest mechanisms by which protein factors involved in telomere maintenance can access telomeric chromatin in its compact form.


Subject(s)
Chromatin , DNA , Histones , Molecular Conformation , Telomere , Chromatin/chemistry , Chromatin/genetics , Chromatin/ultrastructure , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , DNA Damage , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Humans , Microscopy, Electron , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/ultrastructure , Single Molecule Imaging , Telomere/chemistry , Telomere/genetics , Telomere/ultrastructure
7.
Annu Rev Cell Dev Biol ; 30: 561-80, 2014.
Article in English | MEDLINE | ID: mdl-25000994

ABSTRACT

In mammals, the process of X-chromosome inactivation ensures equivalent levels of X-linked gene expression between males and females through the silencing of one of the two X chromosomes in female cells. The process is established early in development and is initiated by a unique locus, which produces a long noncoding RNA, Xist. The Xist transcript triggers gene silencing in cis by coating the future inactive X chromosome. It also induces a cascade of chromatin changes, including posttranslational histone modifications and DNA methylation, and leads to the stable repression of all X-linked genes throughout development and adult life. We review here recent progress in our understanding of the molecular mechanisms involved in the initiation of Xist expression, the propagation of the Xist RNA along the chromosome, and the cis-elements and trans-acting factors involved in the maintenance of the repressed state. We also describe the diverse strategies used by nonplacental mammals for X-chromosome dosage compensation and highlight the common features and differences between eutherians and metatherians, in particular regarding the involvement of long noncoding RNAs.


Subject(s)
Gene Silencing , RNA, Long Noncoding/genetics , X Chromosome Inactivation/genetics , Animals , Chromatin/genetics , Chromatin/ultrastructure , Chromosome Mapping , Chromosomes, Human, X/genetics , Embryonic Stem Cells/ultrastructure , Evolution, Molecular , Female , Genomic Imprinting , Humans , Long Interspersed Nucleotide Elements , Male , Marsupialia/genetics , Mice , Sex Determination Processes , Transcription Factors/genetics , X Chromosome/genetics , X Chromosome/ultrastructure
8.
Annu Rev Cell Dev Biol ; 30: 581-613, 2014.
Article in English | MEDLINE | ID: mdl-25150012

ABSTRACT

Embryogenesis depends on a highly coordinated cascade of genetically encoded events. In animals, maternal factors contributed by the egg cytoplasm initially control development, whereas the zygotic nuclear genome is quiescent. Subsequently, the genome is activated, embryonic gene products are mobilized, and maternal factors are cleared. This transfer of developmental control is called the maternal-to-zygotic transition (MZT). In this review, we discuss recent advances toward understanding the scope, timing, and mechanisms that underlie zygotic genome activation at the MZT in animals. We describe high-throughput techniques to measure the embryonic transcriptome and explore how regulation of the cell cycle, chromatin, and transcription factors together elicits specific patterns of embryonic gene expression. Finally, we illustrate the interplay between zygotic transcription and maternal clearance and show how these two activities combine to reprogram two terminally differentiated gametes into a totipotent embryo.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , RNA, Messenger, Stored/genetics , Transcription, Genetic , Zygote/metabolism , Animals , Cell Cycle , Chromatin/genetics , Chromatin/ultrastructure , Drosophila Proteins/physiology , Egg Proteins/genetics , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental/drug effects , High-Throughput Nucleotide Sequencing , Histones/physiology , Humans , Models, Genetic , Oocytes/metabolism , Pregnancy , RNA Stability , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcriptome , Xenopus Proteins/physiology , Zebrafish Proteins/physiology
9.
Mol Cell ; 79(3): 521-534.e15, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32592681

ABSTRACT

Genome-wide mapping of chromatin interactions at high resolution remains experimentally and computationally challenging. Here we used a low-input "easy Hi-C" protocol to map the 3D genome architecture in human neurogenesis and brain tissues and also demonstrated that a rigorous Hi-C bias-correction pipeline (HiCorr) can significantly improve the sensitivity and robustness of Hi-C loop identification at sub-TAD level, especially the enhancer-promoter (E-P) interactions. We used HiCorr to compare the high-resolution maps of chromatin interactions from 10 tissue or cell types with a focus on neurogenesis and brain tissues. We found that dynamic chromatin loops are better hallmarks for cellular differentiation than compartment switching. HiCorr allowed direct observation of cell-type- and differentiation-specific E-P aggregates spanning large neighborhoods, suggesting a mechanism that stabilizes enhancer contacts during development. Interestingly, we concluded that Hi-C loop outperforms eQTL in explaining neurological GWAS results, revealing a unique value of high-resolution 3D genome maps in elucidating the disease etiology.


Subject(s)
Chromatin/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genome, Human , Neurogenesis/genetics , Promoter Regions, Genetic , Adult , Cell Line , Cerebrum/cytology , Cerebrum/growth & development , Cerebrum/metabolism , Chromatin/ultrastructure , Chromosome Mapping , Fetus , Histones/genetics , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/metabolism , Temporal Lobe/cytology , Temporal Lobe/growth & development , Temporal Lobe/metabolism , Transcription Factors/classification , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Mol Cell ; 74(6): 1110-1122, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31226276

ABSTRACT

During embryogenesis, precise gene transcription in space and time requires that distal enhancers and promoters communicate by physical proximity within gene regulatory landscapes. To achieve this, regulatory landscapes fold in nuclear space, creating complex 3D structures that influence enhancer-promoter communication and gene expression and that, when disrupted, can cause disease. Here, we provide an overview of how enhancers and promoters construct regulatory landscapes and how multiple scales of 3D chromatin structure sculpt their communication. We focus on emerging views of what enhancer-promoter contacts and chromatin domains physically represent and how two antagonistic fundamental forces-loop extrusion and homotypic attraction-likely form them. We also examine how these same forces spatially separate regulatory landscapes by functional state, thereby creating higher-order compartments that reconfigure during development to enable proper enhancer-promoter communication.


Subject(s)
Chromatin/ultrastructure , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Genome , Promoter Regions, Genetic , Transcription, Genetic , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chromatin/metabolism , Embryo, Mammalian , Embryonic Development/genetics , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Humans , Molecular Conformation
11.
Nature ; 587(7835): 678-682, 2020 11.
Article in English | MEDLINE | ID: mdl-32911480

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is an innate immune sensor for cytosolic microbial DNA1. After binding DNA, cGAS synthesizes the messenger 2'3'-cyclic GMP-AMP (cGAMP)2-4, which triggers cell-autonomous defence and the production of type I interferons and pro-inflammatory cytokines via the activation of STING5. In addition to responding to cytosolic microbial DNA, cGAS also recognizes mislocalized cytosolic self-DNA and has been implicated in autoimmunity and sterile inflammation6,7. Specificity towards pathogen- or damage-associated DNA was thought to be caused by cytosolic confinement. However, recent findings place cGAS robustly in the nucleus8-10, where tight tethering of chromatin is important to prevent autoreactivity to self-DNA8. Here we show how cGAS is sequestered and inhibited by chromatin. We provide a cryo-electron microscopy structure of the cGAS catalytic domain bound to a nucleosome, which shows that cGAS does not interact with the nucleosomal DNA, but instead interacts with histone 2A-histone 2B, and is tightly anchored to the 'acidic patch'. The interaction buries the cGAS DNA-binding site B, and blocks the formation of active cGAS dimers. The acidic patch robustly outcompetes agonistic DNA for binding to cGAS, which suggests that nucleosome sequestration can efficiently inhibit cGAS, even when accessible DNA is nearby, such as in actively transcribed genomic regions. Our results show how nuclear cGAS is sequestered by chromatin and provides a mechanism for preventing autoreactivity to nuclear self-DNA.


Subject(s)
Catalytic Domain , Chromatin/chemistry , Chromatin/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/chemistry , Amino Acid Sequence , Animals , Autoantigens/chemistry , Autoantigens/immunology , Autoantigens/metabolism , Autoantigens/ultrastructure , Binding Sites , Binding, Competitive , Chromatin/genetics , Chromatin/ultrastructure , Cryoelectron Microscopy , DNA/chemistry , DNA/immunology , DNA/metabolism , DNA/ultrastructure , Enzyme Activation , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Humans , Hydrophobic and Hydrophilic Interactions , Immunity, Innate , Mice , Models, Molecular , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/ultrastructure , Protein Multimerization , THP-1 Cells
12.
Annu Rev Cell Dev Biol ; 28: 113-35, 2012.
Article in English | MEDLINE | ID: mdl-22804576

ABSTRACT

Cell size varies widely among different organisms as well as within the same organism in different tissue types and during development, which places variable metabolic and functional demands on organelles and internal structures. A fundamental question is how essential subcellular components scale to accommodate cell size differences. Nuclear transport has emerged as a conserved means of scaling nuclear size. A meiotic spindle scaling factor has been identified as the microtubule-severing protein katanin, which is differentially regulated by phosphorylation in two different-sized frog species. Anaphase mechanisms and levels of chromatin compaction both act to coordinate cell size with spindle and chromosome dimensions to ensure accurate genome distribution during cell division. Scaling relationships and mechanisms for many membrane-bound compartments remain largely unknown and are complicated by their heterogeneity and dynamic nature. This review summarizes cell and organelle size relationships and the experimental approaches that have elucidated mechanisms of intracellular scaling.


Subject(s)
Cell Size , Animals , Cell Division , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Nucleus Size , Chromatin/metabolism , Chromatin/ultrastructure , Humans , Nuclear Pore/metabolism , Nuclear Pore/ultrastructure , Yeasts/cytology , Yeasts/physiology , Yeasts/ultrastructure
13.
Annu Rev Biochem ; 79: 1-35, 2010.
Article in English | MEDLINE | ID: mdl-20192760

ABSTRACT

Early influences led me first to medical school with a view to microbiology, but I felt the lack of a deeper foundation and changed to chemistry, which in turn led me to physics and mathematics. I moved to the University of Cape Town to work on the X-ray crystallography of some small organic compounds. I developed a new method of using molecular structure factors to solve the crystal structure, which won me a research studentship to Trinity College Cambridge and the Cavendish Laboratory. There I worked on the austenite-pearlite transition in steel. This is governed by the dissipation of latent heat, and I ended up numerically solving partial differential equations. I used the idea of nucleation and growth during the phase change, which had its echo when I later tackled the assembly of Tobacco mosaic virus (TMV) from its constituent RNA and protein subunits. I wanted to move on to X-ray structure analysis of large biological molecules and obtained a Nuffield Fellowship to work in J.D. Bernal's department at Birkbeck College, London. There, I met Rosalind Franklin, who had taken up the study of TMV. I was able to interpret some of Franklin's beautiful X-ray diffraction patterns of the virus particle. From then on, my fate was sealed. After Franklin's untimely death in 1958, I moved in 1962 to the newly built MRC Laboratory of Molecular Biology in Cambridge, which, under Max Perutz, housed the original MRC unit from the Cavendish Laboratory. I was thus privileged to join the Laboratory at an early stage in its expansion and consequently able to take advantage of, and to help build up, its then unique environment of intellectual and technological sophistication. There I have remained ever since.


Subject(s)
Microbiology/history , Chromatin/ultrastructure , History, 20th Century , Lithuania , Microscopy, Electron , South Africa , Tobacco Mosaic Virus/ultrastructure , X-Ray Diffraction
14.
Annu Rev Genet ; 51: 23-44, 2017 11 27.
Article in English | MEDLINE | ID: mdl-28853923

ABSTRACT

The three-dimensional (3D) genome structure is highly ordered by a hierarchy of organizing events ranging from enhancer-promoter or gene-gene contacts to chromosomal territorial arrangement. It is becoming clear that the cohesin and condensin complexes are key molecular machines that organize the 3D genome structure. These complexes are highly conserved from simple systems, e.g., yeast cells, to the much more complex human system. Therefore, knowledge from the budding and fission yeast systems illuminates highly conserved molecular mechanisms of how cohesin and condensin establish the functional 3D genome structures. Here I discuss how these complexes are recruited across the yeast genomes, mediate distinct genome-organizing events such as gene contacts and topological domain formation, and participate in important nuclear activities including transcriptional regulation and chromosomal dynamics.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , Genome, Fungal , Multiprotein Complexes/genetics , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatin/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Fungal/chemistry , Chromosomes, Fungal/ultrastructure , Conserved Sequence , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Multiprotein Complexes/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Schizosaccharomyces/metabolism , Schizosaccharomyces/ultrastructure , Transcription, Genetic , Cohesins
15.
Mol Cell ; 66(3): 384-397.e8, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28475873

ABSTRACT

Linker histones associate with nucleosomes to promote the formation of higher-order chromatin structure, but the underlying molecular details are unclear. We investigated the structure of a 197 bp nucleosome bearing symmetric 25 bp linker DNA arms in complex with vertebrate linker histone H1. We determined electron cryo-microscopy (cryo-EM) and crystal structures of unbound and H1-bound nucleosomes and validated these structures by site-directed protein cross-linking and hydroxyl radical footprinting experiments. Histone H1 shifts the conformational landscape of the nucleosome by drawing the two linkers together and reducing their flexibility. The H1 C-terminal domain (CTD) localizes primarily to a single linker, while the H1 globular domain contacts the nucleosome dyad and both linkers, associating more closely with the CTD-distal linker. These findings reveal that H1 imparts a strong degree of asymmetry to the nucleosome, which is likely to influence the assembly and architecture of higher-order structures.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA/metabolism , Histones/metabolism , Nucleosomes/metabolism , Animals , Base Pairing , Binding Sites , Chromatin/chemistry , Chromatin/genetics , Chromatin/ultrastructure , Cryoelectron Microscopy , DNA/chemistry , DNA/genetics , Histones/chemistry , Humans , Models, Molecular , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/ultrastructure , Protein Binding , Protein Interaction Domains and Motifs , Structure-Activity Relationship , Time Factors , Xenopus laevis/genetics , Xenopus laevis/metabolism
16.
Trends Genet ; 37(3): 224-234, 2021 03.
Article in English | MEDLINE | ID: mdl-32921511

ABSTRACT

Dysfunctions of nuclear processes including transcription and DNA repair lead to severe human diseases. Gaining an understanding of how these processes operate in the crowded context of chromatin can be particularly challenging. Mediator is a large multiprotein complex conserved in eukaryotes with a key coactivator role in the regulation of RNA polymerase (Pol) II transcription. Despite intensive studies, the molecular mechanisms underlying Mediator function remain to be fully understood. Novel findings have provided insights into the relationship between Mediator and chromatin architecture, revealed its role in connecting transcription with DNA repair and proposed an emerging mechanism of phase separation involving Mediator condensates. Recent developments in the field suggest multiple functions of Mediator going beyond transcriptional processes per se that would explain its involvement in various human pathologies.


Subject(s)
Chromatin/genetics , Mediator Complex/genetics , RNA Polymerase II/genetics , Transcription, Genetic/genetics , Chromatin/ultrastructure , DNA Repair/genetics , Humans , Mediator Complex/ultrastructure , RNA Polymerase II/ultrastructure
17.
Mol Cell ; 64(4): 790-802, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27840028

ABSTRACT

Recent studies have revealed the importance of Ki-67 and the chromosome periphery in chromosome structure and segregation, but little is known about this elusive chromosome compartment. Here we used correlative light and serial block-face scanning electron microscopy, which we term 3D-CLEM, to model the entire mitotic chromosome complement at ultra-structural resolution. Prophase chromosomes exhibit a highly irregular surface appearance with a volume smaller than metaphase chromosomes. This may be because of the absence of the periphery, which associates with chromosomes only after nucleolar disassembly later in prophase. Indeed, the nucleolar volume almost entirely accounts for the extra volume found in metaphase chromosomes. Analysis of wild-type and Ki-67-depleted chromosomes reveals that the periphery comprises 30%-47% of the entire chromosome volume and more than 33% of the protein mass of isolated mitotic chromosomes determined by quantitative proteomics. Thus, chromatin makes up a surprisingly small percentage of the total mass of metaphase chromosomes.


Subject(s)
Chromatin/ultrastructure , Chromosomes/ultrastructure , Metaphase , Microscopy, Electron, Scanning/methods , Prophase , Cell Line, Transformed , Cell Nucleolus/chemistry , Cell Nucleolus/ultrastructure , Chromatin/chemistry , Chromosomes/chemistry , Gene Expression , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Retinal Pigment Epithelium/chemistry , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure
18.
Trends Genet ; 36(4): 273-287, 2020 04.
Article in English | MEDLINE | ID: mdl-32007290

ABSTRACT

It is now widely appreciated that the spatial organization of the genome is nonrandom, and its complex 3D folding has important consequences for many genome processes. Recent developments in multiplexed, super-resolution microscopy have enabled an unprecedented view of the polymeric structure of chromatin - from the loose folds of whole chromosomes to the detailed loops of cis-regulatory elements that regulate gene expression. Facilitated by the use of robotics, microfluidics, and improved approaches to super-resolution, thousands to hundreds of thousands of individual cells can now be analyzed in an individual experiment. This has led to new insights into the nature of genomic structural features identified by sequencing, such as topologically associated domains (TADs), and the nature of enhancer-promoter interactions underlying transcriptional regulation. We review these recent improvements.


Subject(s)
Chromatin/ultrastructure , Chromosomes/ultrastructure , Molecular Imaging/trends , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Chromosomes/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Humans , Promoter Regions, Genetic/genetics
19.
EMBO J ; 38(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30609992

ABSTRACT

Cryo-electron tomography and small-angle X-ray scattering were used to investigate the chromatin folding in metaphase chromosomes. The tomographic 3D reconstructions show that frozen-hydrated chromatin emanated from chromosomes is planar and forms multilayered plates. The layer thickness was measured accounting for the contrast transfer function fringes at the plate edges, yielding a width of ~ 7.5 nm, which is compatible with the dimensions of a monolayer of nucleosomes slightly tilted with respect to the layer surface. Individual nucleosomes are visible decorating distorted plates, but typical plates are very dense and nucleosomes are not identifiable as individual units, indicating that they are tightly packed. Two layers in contact are ~ 13 nm thick, which is thinner than the sum of two independent layers, suggesting that nucleosomes in the layers interdigitate. X-ray scattering of whole chromosomes shows a main scattering peak at ~ 6 nm, which can be correlated with the distance between layers and between interdigitating nucleosomes interacting through their faces. These observations support a model where compact chromosomes are composed of many chromatin layers stacked along the chromosome axis.


Subject(s)
Chromatin/ultrastructure , Chromosome Structures/ultrastructure , Chromosomes, Human/ultrastructure , Metaphase , Nucleosomes/ultrastructure , Electron Microscope Tomography , Frozen Sections , HeLa Cells , Humans
20.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33634313

ABSTRACT

Three-dimensional (3D) architecture of the chromosomes is of crucial importance for transcription regulation and DNA replication. Various high-throughput chromosome conformation capture-based methods have revealed that CTCF-mediated chromatin loops are a major component of 3D architecture. However, CTCF-mediated chromatin loops are cell type specific, and most chromatin interaction capture techniques are time-consuming and labor-intensive, which restricts their usage on a very large number of cell types. Genomic sequence-based computational models are sophisticated enough to capture important features of chromatin architecture and help to identify chromatin loops. In this work, we develop Deep-loop, a convolutional neural network model, to integrate k-tuple nucleotide frequency component, nucleotide pair spectrum encoding, position conservation, position scoring function and natural vector features for the prediction of chromatin loops. By a series of examination based on cross-validation, Deep-loop shows excellent performance in the identification of the chromatin loops from different cell types. The source code of Deep-loop is freely available at the repository https://github.com/linDing-group/Deep-loop.


Subject(s)
CCCTC-Binding Factor/genetics , Chromatin/metabolism , Genome, Human , Neural Networks, Computer , CCCTC-Binding Factor/metabolism , Chromatin/ultrastructure , Datasets as Topic , Gene Expression Regulation , Humans , K562 Cells , MCF-7 Cells , Molecular Conformation , Nucleotide Motifs , Software
SELECTION OF CITATIONS
SEARCH DETAIL