Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nature ; 606(7915): 812-819, 2022 06.
Article in English | MEDLINE | ID: mdl-35676475

ABSTRACT

DNA replication occurs through an intricately regulated series of molecular events and is fundamental for genome stability1,2. At present, it is unknown how the locations of replication origins are determined in the human genome. Here we dissect the role of topologically associating domains (TADs)3-6, subTADs7 and loops8 in the positioning of replication initiation zones (IZs). We stratify TADs and subTADs by the presence of corner-dots indicative of loops and the orientation of CTCF motifs. We find that high-efficiency, early replicating IZs localize to boundaries between adjacent corner-dot TADs anchored by high-density arrays of divergently and convergently oriented CTCF motifs. By contrast, low-efficiency IZs localize to weaker dotless boundaries. Following ablation of cohesin-mediated loop extrusion during G1, high-efficiency IZs become diffuse and delocalized at boundaries with complex CTCF motif orientations. Moreover, G1 knockdown of the cohesin unloading factor WAPL results in gained long-range loops and narrowed localization of IZs at the same boundaries. Finally, targeted deletion or insertion of specific boundaries causes local replication timing shifts consistent with IZ loss or gain, respectively. Our data support a model in which cohesin-mediated loop extrusion and stalling at a subset of genetically encoded TAD and subTAD boundaries is an essential determinant of the locations of replication origins in human S phase.


Subject(s)
Cell Cycle Proteins , Chromatin , Chromosomal Proteins, Non-Histone , Replication Origin , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , Humans , Replication Origin/genetics , S Phase , Cohesins
2.
Nature ; 576(7785): 158-162, 2019 12.
Article in English | MEDLINE | ID: mdl-31776509

ABSTRACT

Features of higher-order chromatin organization-such as A/B compartments, topologically associating domains and chromatin loops-are temporarily disrupted during mitosis1,2. Because these structures are thought to influence gene regulation, it is important to understand how they are re-established after mitosis. Here we examine the dynamics of chromosome reorganization by Hi-C after mitosis in highly purified, synchronous mouse erythroid cell populations. We observed rapid establishment of A/B compartments, followed by their gradual intensification and expansion. Contact domains form from the 'bottom up'-smaller subTADs are formed initially, followed by convergence into multi-domain TAD structures. CTCF is partially retained on mitotic chromosomes and immediately resumes full binding in ana/telophase. By contrast, cohesin is completely evicted from mitotic chromosomes and regains focal binding at a slower rate. The formation of CTCF/cohesin co-anchored structural loops follows the kinetics of cohesin positioning. Stripe-shaped contact patterns-anchored by CTCF-grow in length, which is consistent with a loop-extrusion process after mitosis. Interactions between cis-regulatory elements can form rapidly, with rates exceeding those of CTCF/cohesin-anchored contacts. Notably, we identified a group of rapidly emerging transient contacts between cis-regulatory elements in ana/telophase that are dissolved upon G1 entry, co-incident with the establishment of inner boundaries or nearby interfering chromatin loops. We also describe the relationship between transcription reactivation and architectural features. Our findings indicate that distinct but mutually influential forces drive post-mitotic chromatin reconfiguration.


Subject(s)
Chromatin , G1 Phase , Mitosis , Animals , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Mice , Cohesins
3.
Sci Rep ; 9(1): 9526, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31266973

ABSTRACT

Mammalian genomes are folded into a hierarchy of compartments, topologically associating domains (TADs), subTADs, and long-range looping interactions. The higher-order folding patterns of chromatin contacts within TADs and how they localize to disease-associated single nucleotide variants (daSNVs) remains an open area of investigation. Here, we analyze high-resolution Hi-C data with graph theory to understand possible mesoscale network architecture within chromatin domains. We identify a subset of TADs exhibiting strong core-periphery mesoscale structure in embryonic stem cells, neural progenitor cells, and cortical neurons. Hyper-connected core nodes co-localize with genomic segments engaged in multiple looping interactions and enriched for occupancy of the architectural protein CCCTC binding protein (CTCF). CTCF knockdown and in silico deletion of CTCF-bound core nodes disrupts core-periphery structure, whereas in silico mutation of cell type-specific enhancer or gene nodes has a negligible effect. Importantly, neuropsychiatric daSNVs are significantly more likely to localize with TADs folded into core-periphery networks compared to domains devoid of such structure. Together, our results reveal that a subset of TADs encompasses looping interactions connected into a core-periphery mesoscale network. We hypothesize that daSNVs in the periphery of genome folding networks might preserve global nuclear architecture but cause local topological and functional disruptions contributing to human disease. By contrast, daSNVs co-localized with hyper-connected core nodes might cause severe topological and functional disruptions. Overall, these findings shed new light into the mesoscale network structure of fine scale genome folding within chromatin domains and its link to common genetic variants in human disease.


Subject(s)
Chromosomes/chemistry , Models, Biological , CCCTC-Binding Factor/antagonists & inhibitors , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , Chromosomes/genetics , Chromosomes/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome , Humans , Mental Disorders/genetics , Mental Disorders/pathology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/chemistry , Neurons/metabolism , Polymorphism, Single Nucleotide , RNA Interference
4.
Cell ; 175(1): 224-238.e15, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30173918

ABSTRACT

More than 25 inherited human disorders are caused by the unstable expansion of repetitive DNA sequences termed short tandem repeats (STRs). A fundamental unresolved question is why some STRs are susceptible to pathologic expansion, whereas thousands of repeat tracts across the human genome are relatively stable. Here, we discover that nearly all disease-associated STRs (daSTRs) are located at boundaries demarcating 3D chromatin domains. We identify a subset of boundaries with markedly higher CpG island density compared to the rest of the genome. daSTRs specifically localize to ultra-high-density CpG island boundaries, suggesting they might be hotspots for epigenetic misregulation or topological disruption linked to STR expansion. Fragile X syndrome patients exhibit severe boundary disruption in a manner that correlates with local loss of CTCF occupancy and the degree of FMR1 silencing. Our data uncover higher-order chromatin architecture as a new dimension in understanding repeat expansion disorders.


Subject(s)
Chromatin/genetics , Microsatellite Repeats/physiology , Trinucleotide Repeat Expansion/physiology , Adult , Brain/cytology , Brain/pathology , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/physiology , Cell Line , Chromatin/physiology , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , CpG Islands/genetics , CpG Islands/physiology , DNA/genetics , Disease/etiology , Disease/genetics , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/physiology , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Genome, Human/genetics , Humans , Male , Microsatellite Repeats/genetics , Trinucleotide Repeat Expansion/genetics
5.
Methods Enzymol ; 603: 93-101, 2018.
Article in English | MEDLINE | ID: mdl-29673536

ABSTRACT

Methods for using exogenous fluorophore and general anesthetic 1-aminoanthracene (1-AMA) and its photoactive derivative 1-azidoanthracene (1-AZA) are provided. 1-AMA potentiates GABAA chloride currents and immobilizes Xenopus laevis tadpoles. Cellular and tissue anesthetic distribution can be imaged for quantifying "on-pathway" and "off-pathway" targets. 1-AZA shares targets with 1-AMA and offers further optoanesthetic spatial and temporal control upon near-UV laser irradiation. Furthermore, 1-AZA adduction provides screening of possible relevant anesthetic protein targets and binding site characterization. We highlight several useful imaging and binding assays to demonstrate utility of 1-AMA and its derivative 1-AZA.


Subject(s)
Anesthetics, General/chemistry , Anthracenes/chemistry , Azides/chemistry , Larva/metabolism , Receptors, GABA-A/chemistry , Anesthetics, General/metabolism , Animals , Anthracenes/metabolism , Azides/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Larva/growth & development , Larva/ultrastructure , Lasers , Microscopy, Fluorescence , Photochemical Processes , Protein Binding , Receptors, GABA-A/metabolism , Ultraviolet Rays , Xenopus laevis/anatomy & histology , Xenopus laevis/growth & development , Xenopus laevis/metabolism
6.
Nat Methods ; 15(2): 119-122, 2018 02.
Article in English | MEDLINE | ID: mdl-29334377

ABSTRACT

Mammalian genomes are folded in a hierarchy of compartments, topologically associating domains (TADs), subTADs and looping interactions. Here, we describe 3DNetMod, a graph theory-based method for sensitive and accurate detection of chromatin domains across length scales in Hi-C data. We identify nested, partially overlapping TADs and subTADs genome wide by optimizing network modularity and varying a single resolution parameter. 3DNetMod can be applied broadly to understand genome reconfiguration in development and disease.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Computational Biology/methods , Computer Graphics , Genome, Human , High-Throughput Nucleotide Sequencing , Humans
7.
Mol Cell ; 66(1): 102-116.e7, 2017 Apr 06.
Article in English | MEDLINE | ID: mdl-28388437

ABSTRACT

Bromodomain and extraterminal motif (BET) proteins are pharmacologic targets for the treatment of diverse diseases, yet the roles of individual BET family members remain unclear. We find that BRD2, but not BRD4, co-localizes with the architectural/insulator protein CCCTC-binding factor (CTCF) genome-wide. CTCF recruits BRD2 to co-bound sites whereas BRD2 is dispensable for CTCF occupancy. Disruption of a CTCF/BRD2-occupied element positioned between two unrelated genes enables regulatory influence to spread from one gene to another, suggesting that CTCF and BRD2 form a transcriptional boundary. Accordingly, single-molecule mRNA fluorescence in situ hybridization (FISH) reveals that, upon site-specific CTCF disruption or BRD2 depletion, expression of the two genes becomes increasingly correlated. HiC shows that BRD2 depletion weakens boundaries co-occupied by CTCF and BRD2, but not those that lack BRD2. These findings indicate that BRD2 supports boundary activity, and they raise the possibility that pharmacologic BET inhibitors can influence gene expression in part by perturbing domain boundary function.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Repressor Proteins/metabolism , Transcription, Genetic , Animals , Binding Sites , CCCTC-Binding Factor , CRISPR-Cas Systems , Cell Line , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Editing/methods , In Situ Hybridization, Fluorescence , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Single Molecule Imaging/methods , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
8.
J Am Chem Soc ; 135(14): 5389-98, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23484901

ABSTRACT

Recently, we identified 1-aminoanthracene as a fluorescent general anesthetic. To investigate the mechanism of action, a photoactive analogue, 1-azidoanthracene, was synthesized. Administration of 1-azidoanthracene to albino stage 40-47 tadpoles was found to immobilize animals upon near-UV irradiation of the forebrain region. The immobilization was often reversible, but it was characterized by a longer duration consistent with covalent attachment of the ligand to functionally important targets. IEF/SDS-PAGE examination of irradiated tadpole brain homogenate revealed labeled protein, identified by mass spectrometry as ß-tubulin. In vitro assays with aminoanthracene-cross-linked tubulin indicated inhibition of microtubule polymerization, similar to colchicine. Tandem mass spectrometry confirmed anthracene binding near the colchicine site. Stage 40-47 tadpoles were also incubated 1 h with microtubule stabilizing agents, epothilone D or discodermolide, followed by dosing with 1-aminoanthracene. The effective concentration of 1-aminoanthracene required to immobilize the tadpoles was significantly increased in the presence of either microtubule stabilizing agent. Epothilone D similarly mitigated the effects of a clinical neurosteroid general anesthetic, allopregnanolone, believed to occupy the colchicine site in tubulin. We conclude that neuronal microtubules are "on-pathway" targets for anthracene general anesthetics and may also represent functional targets for some neurosteroid general anesthetics.


Subject(s)
Anesthesia , Anthracenes/pharmacology , Azides/pharmacology , Microtubules/drug effects , Animals , Anthracenes/administration & dosage , Anthracenes/chemistry , Azides/administration & dosage , Azides/chemistry , Larva/drug effects , Microtubules/metabolism , Molecular Structure , Ultraviolet Rays , Xenopus laevis
10.
J Chem Theory Comput ; 6(9): 2804-8, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-26616081

ABSTRACT

Previous work by us showed that in replica exchange molecular dynamics, exchanges should be attempted extremely often, providing gains in efficiency and no undesired effects. Since that time some questions have been raised about the extendability of these claims to the general case. In this work, we answer this question in two ways. First, we perform a study measuring the effect of exchange attempt frequency in explicit solvent simulations including thousands of atoms. This shows, consistent with the previous assertion, that high exchange attempt frequency allows an optimal rate of exploration of configurational space. Second, we present an explanation of many theoretical and technical pitfalls when implementing replica exchange that cause "improper" exchanges resulting in erroneous data, exacerbated by high exchange attempt frequency.

SELECTION OF CITATIONS
SEARCH DETAIL