Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 196
Filter
Add more filters

Publication year range
1.
Cell ; 176(6): 1502-1515.e10, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30799036

ABSTRACT

Several general principles of global 3D genome organization have recently been established, including non-random positioning of chromosomes and genes in the cell nucleus, distinct chromatin compartments, and topologically associating domains (TADs). However, the extent and nature of cell-to-cell and cell-intrinsic variability in genome architecture are still poorly characterized. Here, we systematically probe heterogeneity in genome organization. High-throughput optical mapping of several hundred intra-chromosomal interactions in individual human fibroblasts demonstrates low association frequencies, which are determined by genomic distance, higher-order chromatin architecture, and chromatin environment. The structure of TADs is variable between individual cells, and inter-TAD associations are common. Furthermore, single-cell analysis reveals independent behavior of individual alleles in single nuclei. Our observations reveal extensive variability and heterogeneity in genome organization at the level of individual alleles and demonstrate the coexistence of a broad spectrum of genome configurations in a cell population.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Chromatin/genetics , Genome Components/physiology , Cell Line , Cell Nucleus/genetics , Chromosomes , Fibroblasts/physiology , Genome/genetics , Genome Components/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Single-Cell Analysis
2.
Cell ; 174(1): 172-186.e21, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29958106

ABSTRACT

The fusion oncoprotein CBFß-SMMHC, expressed in leukemia cases with chromosome 16 inversion, drives leukemia development and maintenance by altering the activity of the transcription factor RUNX1. Here, we demonstrate that CBFß-SMMHC maintains cell viability by neutralizing RUNX1-mediated repression of MYC expression. Upon pharmacologic inhibition of the CBFß-SMMHC/RUNX1 interaction, RUNX1 shows increased binding at three MYC distal enhancers, where it represses MYC expression by mediating the replacement of the SWI/SNF complex component BRG1 with the polycomb-repressive complex component RING1B, leading to apoptosis. Combining the CBFß-SMMHC inhibitor with the BET inhibitor JQ1 eliminates inv(16) leukemia in human cells and a mouse model. Enhancer-interaction analysis indicated that the three enhancers are physically connected with the MYC promoter, and genome-editing analysis demonstrated that they are functionally implicated in deregulation of MYC expression. This study reveals a mechanism whereby CBFß-SMMHC drives leukemia maintenance and suggests that inhibitors targeting chromatin activity may prove effective in inv(16) leukemia therapy.


Subject(s)
Apoptosis , Chromatin/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Animals , Apoptosis/drug effects , Azepines/pharmacology , Azepines/therapeutic use , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Inversion/drug effects , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/metabolism , DNA/chemistry , DNA/metabolism , DNA Helicases/metabolism , Disease Models, Animal , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Polycomb Repressive Complex 1/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Triazoles/pharmacology , Triazoles/therapeutic use
3.
Cell ; 169(5): 930-944.e22, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28525758

ABSTRACT

The molecular mechanisms underlying folding of mammalian chromosomes remain poorly understood. The transcription factor CTCF is a candidate regulator of chromosomal structure. Using the auxin-inducible degron system in mouse embryonic stem cells, we show that CTCF is absolutely and dose-dependently required for looping between CTCF target sites and insulation of topologically associating domains (TADs). Restoring CTCF reinstates proper architecture on altered chromosomes, indicating a powerful instructive function for CTCF in chromatin folding. CTCF remains essential for TAD organization in non-dividing cells. Surprisingly, active and inactive genome compartments remain properly segregated upon CTCF depletion, revealing that compartmentalization of mammalian chromosomes emerges independently of proper insulation of TADs. Furthermore, our data support that CTCF mediates transcriptional insulator function through enhancer blocking but not as a direct barrier to heterochromatin spreading. Beyond defining the functions of CTCF in chromosome folding, these results provide new fundamental insights into the rules governing mammalian genome organization.


Subject(s)
Chromosomes, Mammalian/chemistry , Animals , CCCTC-Binding Factor , Cell Cycle , Chromatin/metabolism , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Indoleacetic Acids/pharmacology , Mice , Repressor Proteins/metabolism , Transcription, Genetic
4.
Mol Cell ; 84(8): 1422-1441.e14, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38521067

ABSTRACT

The topological state of chromosomes determines their mechanical properties, dynamics, and function. Recent work indicated that interphase chromosomes are largely free of entanglements. Here, we use Hi-C, polymer simulations, and multi-contact 3C and find that, by contrast, mitotic chromosomes are self-entangled. We explore how a mitotic self-entangled state is converted into an unentangled interphase state during mitotic exit. Most mitotic entanglements are removed during anaphase/telophase, with remaining ones removed during early G1, in a topoisomerase-II-dependent process. Polymer models suggest a two-stage disentanglement pathway: first, decondensation of mitotic chromosomes with remaining condensin loops produces entropic forces that bias topoisomerase II activity toward decatenation. At the second stage, the loops are released, and the formation of new entanglements is prevented by lower topoisomerase II activity, allowing the establishment of unentangled and territorial G1 chromosomes. When mitotic entanglements are not removed in experiments and models, a normal interphase state cannot be acquired.


Subject(s)
Chromosomes , DNA Topoisomerases, Type II , DNA Topoisomerases, Type II/genetics , Chromosomes/genetics , Mitosis/genetics , Interphase/genetics , Polymers
5.
Cell ; 164(6): 1110-1121, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26967279

ABSTRACT

Proper expression of genes requires communication with their regulatory elements that can be located elsewhere along the chromosome. The physics of chromatin fibers imposes a range of constraints on such communication. The molecular and biophysical mechanisms by which chromosomal communication is established, or prevented, have become a topic of intense study, and important roles for the spatial organization of chromosomes are being discovered. Here we present a view of the interphase 3D genome characterized by extensive physical compartmentalization and insulation on the one hand and facilitated long-range interactions on the other. We propose the existence of topological machines dedicated to set up and to exploit a 3D genome organization to both promote and censor communication along and between chromosomes.


Subject(s)
Chromosomes/metabolism , Genome , Adenosine Triphosphatases/metabolism , Animals , CCCTC-Binding Factor , DNA-Binding Proteins/metabolism , Female , Humans , Mitosis , Multiprotein Complexes/metabolism , Repressor Proteins , X Chromosome Inactivation
6.
Cell ; 165(2): 259-61, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058655

ABSTRACT

Genetic material is not randomly organized within the nucleus of a cell. How this organization occurs and why it matters are questions that Cell editor Marta Koch posed to Mitchell Guttman, Job Dekker, and Stavros Lomvardas. Excerpts from this Conversation are presented below, and an audio file of the full discussion is available with the article online.


Subject(s)
Cell Nucleus/chemistry , Chromosomes/chemistry , DNA/chemistry , Animals , Cell Nucleus/genetics , Cell Nucleus/physiology , Chromosomes/genetics , Chromosomes/metabolism , DNA/genetics , DNA/metabolism , Genomics , National Institutes of Health (U.S.) , United States
7.
Mol Cell ; 83(14): 2434-2448.e7, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37402370

ABSTRACT

Insertions and deletions (indels) are common sources of structural variation, and insertions originating from spontaneous DNA lesions are frequent in cancer. We developed a highly sensitive assay called insertion and deletion sequencing (Indel-seq) to monitor rearrangements in human cells at the TRIM37 acceptor locus that reports indels stemming from experimentally induced and spontaneous genome instability. Templated insertions, which derive from sequences genome wide, require contact between donor and acceptor loci, require homologous recombination, and are stimulated by DNA end-processing. Insertions are facilitated by transcription and involve a DNA/RNA hybrid intermediate. Indel-seq reveals that insertions are generated via multiple pathways. The broken acceptor site anneals with a resected DNA break or invades the displaced strand of a transcription bubble or R-loop, followed by DNA synthesis, displacement, and then ligation by non-homologous end joining. Our studies identify transcription-coupled insertions as a critical source of spontaneous genome instability that is distinct from cut-and-paste events.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Humans , DNA End-Joining Repair , DNA/genetics , Genomic Instability , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Mol Cell ; 83(15): 2624-2640, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37419111

ABSTRACT

The four-dimensional nucleome (4DN) consortium studies the architecture of the genome and the nucleus in space and time. We summarize progress by the consortium and highlight the development of technologies for (1) mapping genome folding and identifying roles of nuclear components and bodies, proteins, and RNA, (2) characterizing nuclear organization with time or single-cell resolution, and (3) imaging of nuclear organization. With these tools, the consortium has provided over 2,000 public datasets. Integrative computational models based on these data are starting to reveal connections between genome structure and function. We then present a forward-looking perspective and outline current aims to (1) delineate dynamics of nuclear architecture at different timescales, from minutes to weeks as cells differentiate, in populations and in single cells, (2) characterize cis-determinants and trans-modulators of genome organization, (3) test functional consequences of changes in cis- and trans-regulators, and (4) develop predictive models of genome structure and function.


Subject(s)
Cell Nucleus , Genome , Genome/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/metabolism
9.
Cell ; 162(1): 108-19, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26119342

ABSTRACT

We describe a Hi-C-based method, Micro-C, in which micrococcal nuclease is used instead of restriction enzymes to fragment chromatin, enabling nucleosome resolution chromosome folding maps. Analysis of Micro-C maps for budding yeast reveals abundant self-associating domains similar to those reported in other species, but not previously observed in yeast. These structures, far shorter than topologically associating domains in mammals, typically encompass one to five genes in yeast. Strong boundaries between self-associating domains occur at promoters of highly transcribed genes and regions of rapid histone turnover that are typically bound by the RSC chromatin-remodeling complex. Investigation of chromosome folding in mutants confirms roles for RSC, "gene looping" factor Ssu72, Mediator, H3K56 acetyltransferase Rtt109, and the N-terminal tail of H4 in folding of the yeast genome. This approach provides detailed structural maps of a eukaryotic genome, and our findings provide insights into the machinery underlying chromosome compaction.


Subject(s)
Biochemistry/methods , Chromosomes, Fungal/chemistry , Nucleosomes/chemistry , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics
10.
Cell ; 163(1): 134-47, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26365489

ABSTRACT

Mammalian interphase chromosomes interact with the nuclear lamina (NL) through hundreds of large lamina-associated domains (LADs). We report a method to map NL contacts genome-wide in single human cells. Analysis of nearly 400 maps reveals a core architecture consisting of gene-poor LADs that contact the NL with high cell-to-cell consistency, interspersed by LADs with more variable NL interactions. The variable contacts tend to be cell-type specific and are more sensitive to changes in genome ploidy than the consistent contacts. Single-cell maps indicate that NL contacts involve multivalent interactions over hundreds of kilobases. Moreover, we observe extensive intra-chromosomal coordination of NL contacts, even over tens of megabases. Such coordinated loci exhibit preferential interactions as detected by Hi-C. Finally, the consistency of NL contacts is inversely linked to gene activity in single cells and correlates positively with the heterochromatic histone modification H3K9me3. These results highlight fundamental principles of single-cell chromatin organization. VIDEO ABSTRACT.


Subject(s)
Chromatin/metabolism , Nuclear Lamina/metabolism , Single-Cell Analysis/methods , Cell Line, Tumor , Chromatin/chemistry , Chromosomes/chemistry , Chromosomes/metabolism , Genome-Wide Association Study , Humans , In Situ Hybridization, Fluorescence , Interphase
11.
Nature ; 625(7993): 181-188, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38123679

ABSTRACT

Olfactory receptor (OR) choice provides an extreme example of allelic competition for transcriptional dominance, where every olfactory neuron stably transcribes one of approximately 2,000 or more OR alleles1,2. OR gene choice is mediated by a multichromosomal enhancer hub that activates transcription at a single OR3,4, followed by OR-translation-dependent feedback that stabilizes this choice5,6. Here, using single-cell genomics, we show formation of many competing hubs with variable enhancer composition, only one of which retains euchromatic features and transcriptional competence. Furthermore, we provide evidence that OR transcription recruits enhancers and reinforces enhancer hub activity locally, whereas OR RNA inhibits transcription of competing ORs over distance, promoting transition to transcriptional singularity. Whereas OR transcription is sufficient to break the symmetry between equipotent enhancer hubs, OR translation stabilizes transcription at the prevailing hub, indicating that there may be sequential non-coding and coding mechanisms that are implemented by OR alleles for transcriptional prevalence. We propose that coding OR mRNAs possess non-coding functions that influence nuclear architecture, enhance their own transcription and inhibit transcription from their competitors, with generalizable implications for probabilistic cell fate decisions.


Subject(s)
Olfactory Receptor Neurons , RNA , Receptors, Odorant , Alleles , Cell Lineage , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Regulatory Sequences, Nucleic Acid/genetics , RNA/genetics , Transcription, Genetic , Genomics , Single-Cell Analysis
12.
Cell ; 157(4): 950-63, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24813616

ABSTRACT

A new level of chromosome organization, topologically associating domains (TADs), was recently uncovered by chromosome conformation capture (3C) techniques. To explore TAD structure and function, we developed a polymer model that can extract the full repertoire of chromatin conformations within TADs from population-based 3C data. This model predicts actual physical distances and to what extent chromosomal contacts vary between cells. It also identifies interactions within single TADs that stabilize boundaries between TADs and allows us to identify and genetically validate key structural elements within TADs. Combining the model's predictions with high-resolution DNA FISH and quantitative RNA FISH for TADs within the X-inactivation center (Xic), we dissect the relationship between transcription and spatial proximity to cis-regulatory elements. We demonstrate that contacts between potential regulatory elements occur in the context of fluctuating structures rather than stable loops and propose that such fluctuations may contribute to asymmetric expression in the Xic during X inactivation.


Subject(s)
Chromosomes/chemistry , Transcription, Genetic , X Chromosome Inactivation , Animals , Chromatin/chemistry , Female , In Situ Hybridization, Fluorescence , Male , Mice , Models, Biological , Models, Molecular , RNA, Long Noncoding/metabolism
13.
Nature ; 616(7957): 495-503, 2023 04.
Article in English | MEDLINE | ID: mdl-37046085

ABSTRACT

Skates are cartilaginous fish whose body plan features enlarged wing-like pectoral fins, enabling them to thrive in benthic environments1,2. However, the molecular underpinnings of this unique trait remain unclear. Here we investigate the origin of this phenotypic innovation by developing the little skate Leucoraja erinacea as a genomically enabled model. Analysis of a high-quality chromosome-scale genome sequence for the little skate shows that it preserves many ancestral jawed vertebrate features compared with other sequenced genomes, including numerous ancient microchromosomes. Combining genome comparisons with extensive regulatory datasets in developing fins-including gene expression, chromatin occupancy and three-dimensional conformation-we find skate-specific genomic rearrangements that alter the three-dimensional regulatory landscape of genes that are involved in the planar cell polarity pathway. Functional inhibition of planar cell polarity signalling resulted in a reduction in anterior fin size, confirming that this pathway is a major contributor to batoid fin morphology. We also identified a fin-specific enhancer that interacts with several hoxa genes, consistent with the redeployment of hox gene expression in anterior pectoral fins, and confirmed its potential to activate transcription in the anterior fin using zebrafish reporter assays. Our findings underscore the central role of genome reorganization and regulatory variation in the evolution of phenotypes, shedding light on the molecular origin of an enigmatic trait.


Subject(s)
Animal Fins , Biological Evolution , Genome , Genomics , Skates, Fish , Animals , Animal Fins/anatomy & histology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Skates, Fish/anatomy & histology , Skates, Fish/genetics , Zebrafish/genetics , Genes, Reporter/genetics
14.
Nat Rev Mol Cell Biol ; 17(12): 741-742, 2016 Nov 21.
Article in English | MEDLINE | ID: mdl-27869158

ABSTRACT

The spatial organization of genomes is studied using microscopy- and chromosome conformation capture (3C)-based methods. The two types of methods produce data that are often consistent, but there are cases where they appear discordant. These cases provide opportunities to derive better models of chromatin folding, which can reconcile the datasets.


Subject(s)
Chromosomes/ultrastructure , Chromosome Mapping , Genetic Loci , Genome, Human , Humans , Nucleic Acid Conformation
16.
Cell ; 153(6): 1281-95, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23706625

ABSTRACT

Understanding the topological configurations of chromatin may reveal valuable insights into how the genome and epigenome act in concert to control cell fate during development. Here, we generate high-resolution architecture maps across seven genomic loci in embryonic stem cells and neural progenitor cells. We observe a hierarchy of 3D interactions that undergo marked reorganization at the submegabase scale during differentiation. Distinct combinations of CCCTC-binding factor (CTCF), Mediator, and cohesin show widespread enrichment in chromatin interactions at different length scales. CTCF/cohesin anchor long-range constitutive interactions that might form the topological basis for invariant subdomains. Conversely, Mediator/cohesin bridge short-range enhancer-promoter interactions within and between larger subdomains. Knockdown of Smc1 or Med12 in embryonic stem cells results in disruption of spatial architecture and downregulation of genes found in cohesin-mediated interactions. We conclude that cell-type-specific chromatin organization occurs at the submegabase scale and that architectural proteins shape the genome in hierarchical length scales.


Subject(s)
Cell Lineage , Chromatin/metabolism , Genome , Nuclear Proteins/analysis , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/chemistry , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genome-Wide Association Study , Mediator Complex/genetics , Mediator Complex/metabolism , Mice , Neural Stem Cells/chemistry , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Sequence Analysis, DNA , Cohesins
17.
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
18.
Mol Cell ; 77(2): 324-337.e8, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31704182

ABSTRACT

A major challenge in biology is to understand how complex gene expression patterns are encoded in the genome. While transcriptional enhancers have been studied extensively, few transcriptional silencers have been identified, and they remain poorly understood. Here, we used a novel strategy to screen hundreds of sequences for tissue-specific silencer activity in whole Drosophila embryos. Almost all of the transcriptional silencers that we identified were also active enhancers in other cellular contexts. These elements are bound by more transcription factors than non-silencers. A subset of these silencers forms long-range contacts with promoters. Deletion of a silencer caused derepression of its target gene. Our results challenge the common practice of treating enhancers and silencers as separate classes of regulatory elements and suggest the possibility that thousands or more bifunctional CRMs remain to be discovered in Drosophila and 104-105 in humans.


Subject(s)
Drosophila/genetics , Enhancer Elements, Genetic/genetics , Silencer Elements, Transcriptional/genetics , Transcription, Genetic/genetics , Animals , Animals, Genetically Modified/genetics , Male
19.
Mol Cell ; 78(3): 554-565.e7, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32213324

ABSTRACT

Over the past decade, 3C-related methods have provided remarkable insights into chromosome folding in vivo. To overcome the limited resolution of prior studies, we extend a recently developed Hi-C variant, Micro-C, to map chromosome architecture at nucleosome resolution in human ESCs and fibroblasts. Micro-C robustly captures known features of chromosome folding including compartment organization, topologically associating domains, and interactions between CTCF binding sites. In addition, Micro-C provides a detailed map of nucleosome positions and localizes contact domain boundaries with nucleosomal precision. Compared to Hi-C, Micro-C exhibits an order of magnitude greater dynamic range, allowing the identification of ∼20,000 additional loops in each cell type. Many newly identified peaks are localized along extrusion stripes and form transitive grids, consistent with their anchors being pause sites impeding cohesin-dependent loop extrusion. Our analyses comprise the highest-resolution maps of chromosome folding in human cells to date, providing a valuable resource for studies of chromosome organization.


Subject(s)
Chromosomes, Human/ultrastructure , Animals , CCCTC-Binding Factor/metabolism , Cells, Cultured , Chromatin/chemistry , Chromosomes, Mammalian/ultrastructure , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Humans , Male , Mammals/genetics , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Signal-To-Noise Ratio
20.
Cell ; 148(5): 908-21, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22341456

ABSTRACT

The extent to which the three-dimensional organization of the genome contributes to chromosomal translocations is an important question in cancer genomics. We generated a high-resolution Hi-C spatial organization map of the G1-arrested mouse pro-B cell genome and used high-throughput genome-wide translocation sequencing to map translocations from target DNA double-strand breaks (DSBs) within it. RAG endonuclease-cleaved antigen-receptor loci are dominant translocation partners for target DSBs regardless of genomic position, reflecting high-frequency DSBs at these loci and their colocalization in a fraction of cells. To directly assess spatial proximity contributions, we normalized genomic DSBs via ionizing radiation. Under these conditions, translocations were highly enriched in cis along single chromosomes containing target DSBs and within other chromosomes and subchromosomal domains in a manner directly related to pre-existing spatial proximity. By combining two high-throughput genomic methods in a genetically tractable system, we provide a new lens for viewing cancer genomes.


Subject(s)
Genome , Neoplasms/genetics , Translocation, Genetic , Animals , DNA Breaks, Double-Stranded/radiation effects , G1 Phase , High-Throughput Nucleotide Sequencing , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Precursor Cells, B-Lymphoid/cytology , Receptors, Antigen/genetics
SELECTION OF CITATIONS
SEARCH DETAIL