Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell ; 176(4): 816-830.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30595451

ABSTRACT

The temporal order of DNA replication (replication timing [RT]) is highly coupled with genome architecture, but cis-elements regulating either remain elusive. We created a series of CRISPR-mediated deletions and inversions of a pluripotency-associated topologically associating domain (TAD) in mouse ESCs. CTCF-associated domain boundaries were dispensable for RT. CTCF protein depletion weakened most TAD boundaries but had no effect on RT or A/B compartmentalization genome-wide. By contrast, deletion of three intra-TAD CTCF-independent 3D contact sites caused a domain-wide early-to-late RT shift, an A-to-B compartment switch, weakening of TAD architecture, and loss of transcription. The dispensability of TAD boundaries and the necessity of these "early replication control elements" (ERCEs) was validated by deletions and inversions at additional domains. Our results demonstrate that discrete cis-regulatory elements orchestrate domain-wide RT, A/B compartmentalization, TAD architecture, and transcription, revealing fundamental principles linking genome structure and function.


Subject(s)
DNA Replication Timing/physiology , DNA Replication/genetics , DNA Replication/physiology , Animals , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Chromatin , DNA/genetics , DNA Replication Timing/genetics , Embryonic Stem Cells , Enhancer Elements, Genetic/genetics , Mammals/genetics , Mammals/metabolism , Mice , Repressor Proteins/metabolism , Spatio-Temporal Analysis
2.
Mol Cell ; 82(14): 2541-2543, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35868255

ABSTRACT

Dequeker and colleagues performed elegant in vivo, in silico, and in vitro experiments to demonstrate that the MCM complex, an essential DNA replication factor, is an obstacle for the DNA loop formation by cohesin.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cell Cycle Proteins/genetics , Cell Nucleus , Chromatin , Chromosomal Proteins, Non-Histone/genetics , Cohesins
3.
Mol Cell ; 78(3): 522-538.e9, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32220303

ABSTRACT

To understand the role of the extensive senescence-associated 3D genome reorganization, we generated genome-wide chromatin interaction maps, epigenome, replication-timing, whole-genome bisulfite sequencing, and gene expression profiles from cells entering replicative senescence (RS) or upon oncogene-induced senescence (OIS). We identify senescence-associated heterochromatin domains (SAHDs). Differential intra- versus inter-SAHD interactions lead to the formation of senescence-associated heterochromatin foci (SAHFs) in OIS but not in RS. This OIS-specific configuration brings active genes located in genomic regions adjacent to SAHDs in close spatial proximity and favors their expression. We also identify DNMT1 as a factor that induces SAHFs by promoting HMGA2 expression. Upon DNMT1 depletion, OIS cells transition to a 3D genome conformation akin to that of cells in replicative senescence. These data show how multi-omics and imaging can identify critical features of RS and OIS and discover determinants of acute senescence and SAHF formation.


Subject(s)
Cellular Senescence/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Genome, Human , Oncogenes , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Fibroblasts , Heterochromatin/genetics , Humans , In Situ Hybridization, Fluorescence
4.
Nature ; 583(7818): 699-710, 2020 07.
Article in English | MEDLINE | ID: mdl-32728249

ABSTRACT

The human and mouse genomes contain instructions that specify RNAs and proteins and govern the timing, magnitude, and cellular context of their production. To better delineate these elements, phase III of the Encyclopedia of DNA Elements (ENCODE) Project has expanded analysis of the cell and tissue repertoires of RNA transcription, chromatin structure and modification, DNA methylation, chromatin looping, and occupancy by transcription factors and RNA-binding proteins. Here we summarize these efforts, which have produced 5,992 new experimental datasets, including systematic determinations across mouse fetal development. All data are available through the ENCODE data portal (https://www.encodeproject.org), including phase II ENCODE1 and Roadmap Epigenomics2 data. We have developed a registry of 926,535 human and 339,815 mouse candidate cis-regulatory elements, covering 7.9 and 3.4% of their respective genomes, by integrating selected datatypes associated with gene regulation, and constructed a web-based server (SCREEN; http://screen.encodeproject.org) to provide flexible, user-defined access to this resource. Collectively, the ENCODE data and registry provide an expansive resource for the scientific community to build a better understanding of the organization and function of the human and mouse genomes.


Subject(s)
DNA/genetics , Databases, Genetic , Genome/genetics , Genomics , Molecular Sequence Annotation , Registries , Regulatory Sequences, Nucleic Acid/genetics , Animals , Chromatin/genetics , Chromatin/metabolism , DNA/chemistry , DNA Footprinting , DNA Methylation/genetics , DNA Replication Timing , Deoxyribonuclease I/metabolism , Genome, Human , Histones/metabolism , Humans , Mice , Mice, Transgenic , RNA-Binding Proteins/genetics , Transcription, Genetic/genetics , Transposases/metabolism
5.
Mol Cell ; 62(5): 756-65, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27259206

ABSTRACT

Complete duplication of large metazoan chromosomes requires thousands of potential initiation sites, only a small fraction of which are selected in each cell cycle. Assembly of the replication machinery is highly conserved and tightly regulated during the cell cycle, but the sites of initiation are highly flexible, and their temporal order of firing is regulated at the level of large-scale multi-replicon domains. Importantly, the number of replication forks must be quickly adjusted in response to replication stress to prevent genome instability. Here we argue that large genomes are divided into domains for exactly this reason. Once established, domain structure abrogates the need for precise initiation sites and creates a scaffold for the evolution of other chromosome functions.


Subject(s)
DNA Replication , DNA/biosynthesis , Genome , S Phase , Animals , Base Sequence , Cell Lineage , Chromatin Assembly and Disassembly , DNA/chemistry , DNA/genetics , DNA Damage , Gene Expression Regulation, Developmental , Genomic Instability , Genotype , Humans , Models, Genetic , Nucleic Acid Conformation , Phenotype , Replication Origin , Stochastic Processes , Structure-Activity Relationship , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
6.
Chromosome Res ; 30(4): 401-414, 2022 12.
Article in English | MEDLINE | ID: mdl-35781769

ABSTRACT

The human genome is divided into functional units that replicate at specific times during S-phase. This temporal program is known as replication timing (RT) and is coordinated with the spatial organization of the genome and transcriptional activity. RT is also cell type-specific, dynamically regulated during development, and alterations in RT are observed in multiple diseases. Thus, the precise measure of RT is critical to understand the role of RT in gene function regulation. Distinct methods for assaying the RT program exist; however, conventional methods require thousands of cells as input, prohibiting its applicability to samples with limited cell numbers such as those from disease patients or from early developing embryos. Although single-cell RT analyses have been developed, these methods are low throughput, require generation of numerous libraries, increased sequencing costs, and produce low resolution data. Here, we developed an improved method to measure RT genome-wide that enables high-resolution analysis of low input samples. This method incorporates direct cell sorting into lysis buffer, as well as DNA fragmentation and library preparation in a single tube, resulting in higher yields, increased quality, and reproducibility with decreased costs. We also performed a systematic data processing analysis to provide standardized parameters for RT measurement. This optimized method facilitates RT analysis and will enable its application to a broad range of studies investigating the role of RT in gene expression, nuclear architecture, and disease.


Subject(s)
DNA Replication Timing , Genome, Human , Humans , Reproducibility of Results , Gene Library , High-Throughput Nucleotide Sequencing/methods , DNA Replication
8.
Genome Res ; 29(9): 1415-1428, 2019 09.
Article in English | MEDLINE | ID: mdl-31434679

ABSTRACT

DNA replication occurs in a defined temporal order known as the replication timing (RT) program and is regulated during development, coordinated with 3D genome organization and transcriptional activity. However, transcription and RT are not sufficiently coordinated to predict each other, suggesting an indirect relationship. Here, we exploit genome-wide RT profiles from 15 human cell types and intermediate differentiation stages derived from human embryonic stem cells to construct different types of RT regulatory networks. First, we constructed networks based on the coordinated RT changes during cell fate commitment to create highly complex RT networks composed of thousands of interactions that form specific functional subnetwork communities. We also constructed directional regulatory networks based on the order of RT changes within cell lineages, and identified master regulators of differentiation pathways. Finally, we explored relationships between RT networks and transcriptional regulatory networks (TRNs) by combining them into more complex circuitries of composite and bipartite networks. Results identified novel trans interactions linking transcription factors that are core to the regulatory circuitry of each cell type to RT changes occurring in those cell types. These core transcription factors were found to bind cooperatively to sites in the affected replication domains, providing provocative evidence that they constitute biologically significant directional interactions. Our findings suggest a regulatory link between the establishment of cell-type-specific TRNs and RT control during lineage specification.


Subject(s)
DNA Replication Timing , Embryonic Stem Cells/cytology , Transcription Factors/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , DNA/metabolism , Embryonic Stem Cells/chemistry , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Transcription, Genetic
9.
Genome Res ; 28(6): 800-811, 2018 06.
Article in English | MEDLINE | ID: mdl-29735606

ABSTRACT

DNA replication occurs in a defined temporal order known as the replication-timing (RT) program. RT is regulated during development in discrete chromosomal units, coordinated with transcriptional activity and 3D genome organization. Here, we derived distinct cell types from F1 hybrid musculus × castaneus mouse crosses and exploited the high single-nucleotide polymorphism (SNP) density to characterize allelic differences in RT (Repli-seq), genome organization (Hi-C and promoter-capture Hi-C), gene expression (total nuclear RNA-seq), and chromatin accessibility (ATAC-seq). We also present HARP, a new computational tool for sorting SNPs in phased genomes to efficiently measure allele-specific genome-wide data. Analysis of six different hybrid mESC clones with different genomes (C57BL/6, 129/sv, and CAST/Ei), parental configurations, and gender revealed significant RT asynchrony between alleles across ∼12% of the autosomal genome linked to subspecies genomes but not to parental origin, growth conditions, or gender. RT asynchrony in mESCs strongly correlated with changes in Hi-C compartments between alleles but not as strongly with SNP density, gene expression, imprinting, or chromatin accessibility. We then tracked mESC RT asynchronous regions during development by analyzing differentiated cell types, including extraembryonic endoderm stem (XEN) cells, four male and female primary mouse embryonic fibroblasts (MEFs), and neural precursor cells (NPCs) differentiated in vitro from mESCs with opposite parental configurations. We found that RT asynchrony and allelic discordance in Hi-C compartments seen in mESCs were largely lost in all differentiated cell types, accompanied by novel sites of allelic asynchrony at a considerably smaller proportion of the genome, suggesting that genome organization of homologs converges to similar folding patterns during cell fate commitment.


Subject(s)
DNA Replication Timing/genetics , DNA Replication/genetics , Genome/genetics , Neural Stem Cells/cytology , Alleles , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Female , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Male , Mice , Mouse Embryonic Stem Cells/cytology , Promoter Regions, Genetic
10.
Bioinformatics ; 35(13): 2167-2176, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30475980

ABSTRACT

MOTIVATION: The replication timing (RT) program has been linked to many key biological processes including cell fate commitment, 3D chromatin organization and transcription regulation. Significant technology progress now allows to characterize the RT program in the entire human genome in a high-throughput and high-resolution fashion. These experiments suggest that RT changes dynamically during development in coordination with gene activity. Since RT is such a fundamental biological process, we believe that an effective quantitative profile of the local RT program from a diverse set of cell types in various developmental stages and lineages can provide crucial biological insights for a genomic locus. RESULTS: In this study, we explored recurrent and spatially coherent combinatorial profiles from 42 RT programs collected from multiple lineages at diverse differentiation states. We found that a Hidden Markov Model with 15 hidden states provide a good model to describe these genome-wide RT profiling data. Each of the hidden state represents a unique combination of RT profiles across different cell types which we refer to as 'RT states'. To understand the biological properties of these RT states, we inspected their relationship with chromatin states, gene expression, functional annotation and 3D chromosomal organization. We found that the newly defined RT states possess interesting genome-wide functional properties that add complementary information to the existing annotation of the human genome. AVAILABILITY AND IMPLEMENTATION: R scripts for inferring HMM models and Perl scripts for further analysis are available https://github.com/PouletAxel/script_HMM_Replication_timing. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
DNA Replication Timing , Genome, Human , Cell Differentiation , Chromatin , Genomics , Humans
11.
Proc Natl Acad Sci U S A ; 114(51): E10972-E10980, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29196523

ABSTRACT

Progeroid syndromes are rare genetic disorders that phenotypically resemble natural aging. Different causal mutations have been identified, but no molecular alterations have been identified that are in common to these diseases. DNA replication timing (RT) is a robust cell type-specific epigenetic feature highly conserved in the same cell types from different individuals but altered in disease. Here, we characterized DNA RT program alterations in Hutchinson-Gilford progeria syndrome (HGPS) and Rothmund-Thomson syndrome (RTS) patients compared with natural aging and cellular senescence. Our results identified a progeroid-specific RT signature that is common to cells from three HGPS and three RTS patients and distinguishes them from healthy individuals across a wide range of ages. Among the RT abnormalities, we identified the tumor protein p63 gene (TP63) as a gene marker for progeroid syndromes. By using the redifferentiation of four patient-derived induced pluripotent stem cells as a model for the onset of progeroid syndromes, we tracked the progression of RT abnormalities during development, revealing altered RT of the TP63 gene as an early event in disease progression of both HGPS and RTS. Moreover, the RT abnormalities in progeroid patients were associated with altered isoform expression of TP63 Our findings demonstrate the value of RT studies to identify biomarkers not detected by other methods, reveal abnormal TP63 RT as an early event in progeroid disease progression, and suggest TP63 gene regulation as a potential therapeutic target.


Subject(s)
DNA Replication Timing , Progeria/genetics , Aged, 80 and over , Biomarkers , Child , Fibroblasts/metabolism , Gene Expression , Genomics/methods , Humans , Infant, Newborn , Lamin Type A/genetics , Lamin Type A/metabolism , Progeria/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
12.
Genome Res ; 25(8): 1091-103, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26055160

ABSTRACT

Duplication of the genome in mammalian cells occurs in a defined temporal order referred to as its replication-timing (RT) program. RT changes dynamically during development, regulated in units of 400-800 kb referred to as replication domains (RDs). Changes in RT are generally coordinated with transcriptional competence and changes in subnuclear position. We generated genome-wide RT profiles for 26 distinct human cell types, including embryonic stem cell (hESC)-derived, primary cells and established cell lines representing intermediate stages of endoderm, mesoderm, ectoderm, and neural crest (NC) development. We identified clusters of RDs that replicate at unique times in each stage (RT signatures) and confirmed global consolidation of the genome into larger synchronously replicating segments during differentiation. Surprisingly, transcriptome data revealed that the well-accepted correlation between early replication and transcriptional activity was restricted to RT-constitutive genes, whereas two-thirds of the genes that switched RT during differentiation were strongly expressed when late replicating in one or more cell types. Closer inspection revealed that transcription of this class of genes was frequently restricted to the lineage in which the RT switch occurred, but was induced prior to a late-to-early RT switch and/or down-regulated after an early-to-late RT switch. Analysis of transcriptional regulatory networks showed that this class of genes contains strong regulators of genes that were only expressed when early replicating. These results provide intriguing new insight into the complex relationship between transcription and RT regulation during human development.


Subject(s)
Cell Lineage , DNA Replication Timing , Gene Expression Profiling/methods , Pluripotent Stem Cells/physiology , Cell Differentiation , Cells, Cultured , Cluster Analysis , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genome, Human , Humans , Pluripotent Stem Cells/cytology
13.
J Cell Biochem ; 118(8): 2151-2160, 2017 08.
Article in English | MEDLINE | ID: mdl-27996173

ABSTRACT

In metazoans, nuclear DNA is organized during the interphase in negatively supercoiled loops anchored to a compartment or substructure known as the nuclear matrix. The interactions between DNA and the nuclear matrix (NM) are of higher affinity than those between DNA and chromatin proteins since the last ones do not resist the procedures for extracting the NM. The structural interactions DNA-NM constitute a set of topological relationships that define a nuclear higher order structure (NHOS) although there are further higher order levels of organization within the nucleus. So far, the evidence derived from studies with primary hepatocytes and naïve B lymphocytes indicates that the NHOS is cell-type specific at the local and at the large-scale level, and so it has been suggested that such NHOS is primary determined by structural and thermodynamic constraints. We carried out a comparative characterization of the NHOS of postmitotic cortical neurons with that of hepatocytes and naïve B lymphocytes. Our results indicate that the NHOS of neurons is completely different at the large scale and at the local level from that one observed in hepatocytes or in naïve B lymphocytes, confirming on the one hand that the set of structural DNA-NM interactions is cell-type specific and supporting, on the other hand the notion that structural constraints that impinge on chromosomal DNA and the NM are more important for determining this NHOS than functional constraints related to replication and/or transcription. J. Cell. Biochem. 118: 2151-2160, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hepatocytes/metabolism , Neurons/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , DNA/metabolism , Hepatocytes/cytology , Kinetics , Neurons/cytology , Nuclear Matrix/metabolism , Rats , Rats, Wistar
14.
Adv Exp Med Biol ; 1042: 229-257, 2017.
Article in English | MEDLINE | ID: mdl-29357061

ABSTRACT

DNA replication occurs in a defined temporal order during S phase, known as the replication timing programme, which is regulated not only during the cell cycle but also during the process of development and differentiation. The units of replication timing regulation, known as replication domains (RDs), frequently comprise several nearly synchronously firing replication origins. Replication domains correspond to topologically associating domains (TADs) mapped by chromatin conformation capture methods and are likely to be the molecular equivalents of replication foci observed using cytogenetic methods. Both TAD and replication foci are considered to be stable structural units of chromosomes, conserved through the cell cycle and development, and accordingly, the boundaries of RDs also appear to be stable in different cell types. During both normal development and progression of disease, distinct cell states are characterized by unique replication timing signatures, with approximately half of genomic RDs switching replication timing between these cell states. Advances in functional genomics provide hope that we can soon gain an understanding of the cause and consequence of the replication timing programme and its myriad correlations with chromatin context and gene regulation.


Subject(s)
Chromatin , DNA Replication/physiology , Genome/genetics , Replicon/physiology , Animals , Binding Sites/genetics , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA Replication Timing , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genome/physiology , Humans
15.
PLoS Genet ; 10(5): e1004290, 2014 May.
Article in English | MEDLINE | ID: mdl-24785991

ABSTRACT

Discovery of lineage-specific somatic copy number variation (CNV) in mammals has led to debate over whether CNVs are mutations that propagate disease or whether they are a normal, and even essential, aspect of cell biology. We show that 1,000 N polyploid trophoblast giant cells (TGCs) of the mouse placenta contain 47 regions, totaling 138 Megabases, where genomic copies are underrepresented (UR). UR domains originate from a subset of late-replicating heterochromatic regions containing gene deserts and genes involved in cell adhesion and neurogenesis. While lineage-specific CNVs have been identified in mammalian cells, classically in the immune system where V(D)J recombination occurs, we demonstrate that CNVs form during gestation in the placenta by an underreplication mechanism, not by recombination nor deletion. Our results reveal that large scale CNVs are a normal feature of the mammalian placental genome, which are regulated systematically during embryogenesis and are propagated by a mechanism of underreplication.


Subject(s)
DNA Copy Number Variations , Genome , Placenta/metabolism , Animals , Cell Adhesion/genetics , Cell Differentiation/genetics , Female , Gene Deletion , Humans , Neurogenesis , Polyploidy , Pregnancy , Stochastic Processes
16.
Curr Protoc ; 3(11): e945, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38009262

ABSTRACT

Replication timing is significantly correlated with gene expression and chromatin organization, changes dynamically during cell differentiation, and is altered in diseased states. Genome-wide analysis of replication timing is performed in actively replicating cells by Repli-seq. Current methods for Repli-seq require cells to be fixed in large numbers. This is a barrier for sample types that are sensitive to fixation or are in very limited numbers. In this article, we outline optimized methods to process live cells and intact nuclei for Repli-seq. Our protocol enables the processing of a smaller number of cells per sample and reduces processing time and sample loss while obtaining high-quality data. Further, for samples that tend to form clumps and are difficult to dissociate into a single-cell suspension, we also outline methods for isolation, staining, and processing of nuclei for Repli-seq. The Repli-seq data obtained from live cells and intact nuclei are comparable to those obtained from the standard protocols. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Live cell isolation and staining Alternate Protocol: Nuclei isolation and staining.


Subject(s)
Cell Nucleus , Coloring Agents , Cell Nucleus/genetics , DNA Replication Timing , Cell Separation , Genome
17.
Cell Transplant ; 32: 9636897231158728, 2023.
Article in English | MEDLINE | ID: mdl-36929807

ABSTRACT

Currently, there is a significant shortage of transplantable organs for patients in need. Interspecies chimerism and blastocyst complementation are alternatives for generating transplantable human organs in host animals such as pigs to meet this shortage. While successful interspecies chimerism and organ generation have been observed between evolutionarily close species such as rat and mouse, barriers still exist for more distant species pairs such as human-mouse, marmoset-mouse, human-pig, and others. One of the proposed barriers to chimerism is the difference in developmental stages between the donor cells and the host embryo at the time the cells are introduced into the host embryo. Hence, there is a logical effort to stage-match the donor cells with the host embryos for enhancing interspecies chimerism. In this study, we used an in silico approach to simultaneously stage-match the early developing embryos of four species, including human, marmoset, mouse, and pig based on transcriptome similarities. We used an unsupervised clustering algorithm to simultaneously stage-match all four species as well as Spearman's correlation analyses to stage-match pairs of donor-host species. From our stage-matching analyses, we found that the four stages that best matched with each other are the human blastocyst (E6/E7), the gastrulating mouse embryo (E6-E6.75), the marmoset late inner cell mass, and the pig late blastocyst. We further demonstrated that human pluripotent stem cells best matched with the mouse post-implantation stages. We also performed ontology analysis of the genes upregulated and commonly expressed between donor-host species pairs at their best matched stages. The stage-matching results predicted by this study will inform in vivo and in vitro interspecies chimerism and blastocyst complementation studies and can be used to match donor cells with host embryos between multiple species pairs to enhance chimerism for organogenesis.


Subject(s)
Callithrix , Pluripotent Stem Cells , Swine , Mice , Animals , Humans , Rats , Chimerism , Embryo, Mammalian , Blastocyst
18.
Front Genet ; 13: 907547, 2022.
Article in English | MEDLINE | ID: mdl-36506300

ABSTRACT

Common fragile sites (CFSs) are specific regions of all individuals' genome that are predisposed to DNA double strand breaks (DSBs) and undergo subsequent rearrangements. CFS formation can be induced in vitro by mild level of DNA replication stress, such as DNA polymerase inhibition or nucleotide pool disturbance. The mechanisms of CFS formation have been linked to DNA replication timing control, transcription activities, as well as chromatin organization. However, it is unclear what specific cis- or trans-factors regulate the interplay between replication and transcription that determine CFS formation. We recently reported genome-wide mapping of DNA DSBs under replication stress induced by aphidicolin in human lymphoblastoids for the first time. Here, we systematically compared these DSBs with regards to nearby epigenomic features mapped in the same cell line from published studies. We demonstrate that aphidicolin-induced DSBs are strongly correlated with histone 3 lysine 36 trimethylation, a marker for active transcription. We further demonstrate that this DSB signature is a composite effect by the dual treatment of aphidicolin and its solvent, dimethylsulfoxide, the latter of which potently induces transcription on its own. We also present complementing evidence for the association between DSBs and 3D chromosome architectural domains with high density gene cluster and active transcription. Additionally, we show that while DSBs were detected at all but one of the fourteen finely mapped CFSs, they were not enriched in the CFS core sequences and rather demarcated the CFS core region. Related to this point, DSB density was not higher in large genes of greater than 300 kb, contrary to reported enrichment of CFS sites at these large genes. Finally, replication timing analyses demonstrate that the CFS core region contain initiation events, suggesting that altered replication dynamics are responsible for CFS formation in relatively higher level of replication stress.

19.
BMC Cell Biol ; 12: 3, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21244708

ABSTRACT

BACKGROUND: In the interphase nucleus of metazoan cells DNA is organized in supercoiled loops anchored to a nuclear matrix (NM). There is varied evidence indicating that DNA replication occurs in replication factories organized upon the NM and that DNA loops may correspond to the actual replicons in vivo. In normal rat liver the hepatocytes are arrested in G0 but they synchronously re-enter the cell cycle after partial-hepatectomy leading to liver regeneration in vivo. We have previously determined in quiescent rat hepatocytes that a 162 kbp genomic region containing members of the albumin gene family is organized into five structural DNA loops. RESULTS: In the present work we tracked down the movement relative to the NM of DNA sequences located at different points within such five structural DNA loops during the S phase and after the return to cellular quiescence during liver regeneration. Our results indicate that looped DNA moves sequentially towards the NM during replication and then returns to its original position in newly quiescent cells, once the liver regeneration has been achieved. CONCLUSIONS: Looped DNA moves in a sequential fashion, as if reeled in, towards the NM during DNA replication in vivo thus supporting the notion that the DNA template is pulled progressively towards the replication factories on the NM so as to be replicated. These results provide further evidence that the structural DNA loops correspond to the actual replicons in vivo.


Subject(s)
DNA Replication , DNA/metabolism , Nuclear Matrix/metabolism , Animals , Cells, Cultured , Deoxyribonuclease I/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Kinetics , Liver Regeneration , Male , Rats , Rats, Wistar , S Phase
20.
Nat Commun ; 11(1): 3613, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32680994

ABSTRACT

Common fragile sites (CFSs) are regions susceptible to replication stress and are hotspots for chromosomal instability in cancer. Several features were suggested to underlie CFS instability, however, these features are prevalent across the genome. Therefore, the molecular mechanisms underlying CFS instability remain unclear. Here, we explore the transcriptional profile and DNA replication timing (RT) under mild replication stress in the context of the 3D genome organization. The results reveal a fragility signature, comprised of a TAD boundary overlapping a highly transcribed large gene with APH-induced RT-delay. This signature enables precise mapping of core fragility regions in known CFSs and identification of novel fragile sites. CFS stability may be compromised by incomplete DNA replication and repair in TAD boundaries core fragility regions leading to genomic instability. The identified fragility signature will allow for a more comprehensive mapping of CFSs and pave the way for investigating mechanisms promoting genomic instability in cancer.


Subject(s)
Chromosome Fragile Sites/genetics , DNA Replication Timing/genetics , Genome, Human , Genomic Instability , Aphidicolin/pharmacology , Cell Line , Chromatin Immunoprecipitation Sequencing , Chromosome Mapping/methods , DNA/chemistry , DNA Replication Timing/drug effects , Fibroblasts , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/genetics , Nucleic Acid Conformation , Sensitivity and Specificity , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL