Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(21)2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34768871

ABSTRACT

Replication timing (RT) is a cellular program to coordinate initiation of DNA replication in all origins within the genome. RIF1 (replication timing regulatory factor 1) is a master regulator of RT in human cells. This role of RIF1 is associated with binding G4-quadruplexes and changes in 3D chromatin that may suppress origin activation over a long distance. Many effects of RIF1 in fork reactivation and DNA double-strand (DSB) repair (DSBR) are underlined by its interaction with TP53BP1 (tumor protein p53 binding protein). In G1, RIF1 acts antagonistically to BRCA1 (BRCA1 DNA repair associated), suppressing end resection and homologous recombination repair (HRR) and promoting non-homologous end joining (NHEJ), contributing to DSBR pathway choice. RIF1 is an important element of intra-S-checkpoints to recover damaged replication fork with the involvement of HRR. High-resolution microscopic studies show that RIF1 cooperates with TP53BP1 to preserve 3D structure and epigenetic markers of genomic loci disrupted by DSBs. Apart from TP53BP1, RIF1 interact with many other proteins, including proteins involved in DNA damage response, cell cycle regulation, and chromatin remodeling. As impaired RT, DSBR and fork reactivation are associated with genomic instability, a hallmark of malignant transformation, RIF1 has a diagnostic, prognostic, and therapeutic potential in cancer. Further studies may reveal other aspects of common regulation of RT, DSBR, and fork reactivation by RIF1.


Subject(s)
DNA Repair/physiology , DNA Replication Timing/physiology , Telomere-Binding Proteins/metabolism , BRCA1 Protein/metabolism , Chromatin/metabolism , DNA/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA End-Joining Repair/genetics , DNA End-Joining Repair/physiology , DNA Replication/genetics , DNA Replication/physiology , DNA Replication Timing/genetics , Genomic Instability/genetics , Humans , Recombinational DNA Repair , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/physiology , Tumor Suppressor p53-Binding Protein 1/metabolism
2.
Cell Rep ; 37(1): 109799, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610305

ABSTRACT

Replication timing (RT) associates with genome architecture, while having a mixed relationship to histone marks. By profiling replication at high resolution and assessing broad histone marks across the cell cycle at the resolution of RT with and without genetic perturbation, we address the causal relationship between histone marks and RT. Four primary chromatin states, including an uncharacterized H3K36me2 state, emerge and define 97% of the mappable genome. RT and local replication patterns (e.g., initiation zones) quantitatively associate with chromatin states, histone mark dynamics, and spatial chromatin structure. Manipulation of broad histone marks and enhancer elements by overexpressing the histone H3 lysine 9/36 tri-demethylase KDM4A impacts RT across 11% of the genome. Broad histone modification changes were strong predictors of the observed RT alterations. Lastly, replication within H3K36me2-enriched neighborhoods is sensitive to KDM4A overexpression and is controlled at a megabase scale. These studies establish a role for collective chromatin mark regulation in modulating RT.


Subject(s)
Chromatin/chemistry , DNA Replication Timing/physiology , Cell Line , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Genome , Histone Code/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Methylation , S Phase
3.
Cell Rep ; 36(12): 109722, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34551299

ABSTRACT

DNA replication timing and three-dimensional (3D) genome organization are associated with distinct epigenome patterns across large domains. However, whether alterations in the epigenome, in particular cancer-related DNA hypomethylation, affects higher-order levels of genome architecture is still unclear. Here, using Repli-Seq, single-cell Repli-Seq, and Hi-C, we show that genome-wide methylation loss is associated with both concordant loss of replication timing precision and deregulation of 3D genome organization. Notably, we find distinct disruption in 3D genome compartmentalization, striking gains in cell-to-cell replication timing heterogeneity and loss of allelic replication timing in cancer hypomethylation models, potentially through the gene deregulation of DNA replication and genome organization pathways. Finally, we identify ectopic H3K4me3-H3K9me3 domains from across large hypomethylated domains, where late replication is maintained, which we purport serves to protect against catastrophic genome reorganization and aberrant gene transcription. Our results highlight a potential role for the methylome in the maintenance of 3D genome regulation.


Subject(s)
DNA Methylation , DNA Replication Timing/physiology , Genome, Human , Cell Line, Tumor , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Databases, Genetic , Gene Expression , Histones/metabolism , Humans , Sequence Analysis, DNA/methods
4.
Nat Rev Mol Cell Biol ; 20(12): 721-737, 2019 12.
Article in English | MEDLINE | ID: mdl-31477886

ABSTRACT

The 3D organization of mammalian chromatin was described more than 30 years ago by visualizing sites of DNA synthesis at different times during the S phase of the cell cycle. These early cytogenetic studies revealed structurally stable chromosome domains organized into subnuclear compartments. Active-gene-rich domains in the nuclear interior replicate early, whereas more condensed chromatin domains that are largely at the nuclear and nucleolar periphery replicate later. During the past decade, this spatiotemporal DNA replication programme has been mapped along the genome and found to correlate with epigenetic marks, transcriptional activity and features of 3D genome architecture such as chromosome compartments and topologically associated domains. But the causal relationship between these features and DNA replication timing and the regulatory mechanisms involved have remained an enigma. The recent identification of cis-acting elements regulating the replication time and 3D architecture of individual replication domains and of long non-coding RNAs that coordinate whole chromosome replication provide insights into such mechanisms.


Subject(s)
Cell Cycle/physiology , Chromatin Assembly and Disassembly/physiology , DNA Replication Timing/physiology , Genome, Human/physiology , Heterochromatin/metabolism , Animals , Humans
5.
Nat Commun ; 10(1): 416, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679435

ABSTRACT

DNA replication timing is known to facilitate the establishment of the epigenome, however, the intimate connection between replication timing and changes to the genome and epigenome in cancer remain largely uncharacterised. Here, we perform Repli-Seq and integrated epigenome analyses and demonstrate that genomic regions that undergo long-range epigenetic deregulation in prostate cancer also show concordant differences in replication timing. A subset of altered replication timing domains are conserved across cancers from different tissue origins. Notably, late-replicating regions in cancer cells display a loss of DNA methylation, and a switch in heterochromatin features from H3K9me3-marked constitutive to H3K27me3-marked facultative heterochromatin. Finally, analysis of 214 prostate and 35 breast cancer genomes reveal that late-replicating regions are prone to cis and early-replication to trans chromosomal rearrangements. Together, our data suggests that the nature of chromosomal rearrangement in cancer is related to the spatial and temporal positioning and altered epigenetic states of early-replicating compared to late-replicating loci.


Subject(s)
Chromosome Aberrations , DNA Replication Timing/physiology , Epigenesis, Genetic/physiology , Neoplasms/genetics , Breast Neoplasms , Cell Line, Tumor , DNA Methylation , DNA Replication , Deoxyribonuclease I/analysis , Epigenomics , Female , Gene Expression Regulation, Neoplastic , Genome , Genomics , Heterochromatin , Humans , Male , Prostatic Neoplasms , Whole Genome Sequencing
6.
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
7.
EMBO Rep ; 19(9)2018 09.
Article in English | MEDLINE | ID: mdl-30104203

ABSTRACT

Despite its evolutionarily conserved function in controlling DNA replication, the chromosomal binding sites of the budding yeast Rif1 protein are not well understood. Here, we analyse genome-wide binding of budding yeast Rif1 by chromatin immunoprecipitation, during G1 phase and in S phase with replication progressing normally or blocked by hydroxyurea. Rif1 associates strongly with telomeres through interaction with Rap1. By comparing genomic binding of wild-type Rif1 and truncated Rif1 lacking the Rap1-interaction domain, we identify hundreds of Rap1-dependent and Rap1-independent chromosome interaction sites. Rif1 binds to centromeres, highly transcribed genes and replication origins in a Rap1-independent manner, associating with both early and late-initiating origins. Interestingly, Rif1 also binds around activated origins when replication progression is blocked by hydroxyurea, suggesting association with blocked forks. Using nascent DNA labelling and DNA combing techniques, we find that in cells treated with hydroxyurea, yeast Rif1 stabilises recently synthesised DNA Our results indicate that, in addition to controlling DNA replication initiation, budding yeast Rif1 plays an ongoing role after initiation and controls events at blocked replication forks.


Subject(s)
DNA Replication/physiology , Replication Origin/physiology , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Telomere-Binding Proteins/metabolism , Binding Sites/physiology , Cell Cycle , Cell Cycle Proteins/metabolism , Centromere/metabolism , Chromosomes, Plant/chemistry , DNA/metabolism , DNA Replication Timing/physiology , Minichromosome Maintenance Proteins/metabolism , Mutation , Protein Phosphatase 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , S Phase/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Shelterin Complex , Telomere/metabolism , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/genetics , Transcription Factors/metabolism
8.
Nucleic Acids Res ; 46(19): e112, 2018 11 02.
Article in English | MEDLINE | ID: mdl-29986073

ABSTRACT

Genomes are replicated in a reproducible temporal pattern. Current methods for assaying allele replication timing are time consuming and/or expensive. These include high-throughput sequencing which can be used to measure DNA copy number as a proxy for allele replication timing. Here, we use droplet digital PCR to study DNA replication timing at multiple loci in budding yeast and human cells. We establish that the method has temporal and spatial resolutions comparable to the high-throughput sequencing approaches, while being faster than alternative locus-specific methods. Furthermore, the approach is capable of allele discrimination. We apply this method to determine relative replication timing across timing transition zones in cultured human cells. Finally, multiple samples can be analysed in parallel, allowing us to rapidly screen kinetochore mutants for perturbation to centromere replication timing. Therefore, this approach is well suited to the study of locus-specific replication and the screening of cis- and trans-acting mutants to identify mechanisms that regulate local genome replication timing.


Subject(s)
DNA Replication Timing/physiology , DNA Replication/genetics , Polymerase Chain Reaction/methods , Alleles , Cells, Cultured , DNA Copy Number Variations/genetics , DNA Replication Timing/genetics , Genome, Fungal , Genome, Human , HeLa Cells , Humans , Jurkat Cells , Saccharomyces cerevisiae , Saccharomycetales/genetics , Sensitivity and Specificity
9.
PLoS One ; 13(4): e0195207, 2018.
Article in English | MEDLINE | ID: mdl-29659604

ABSTRACT

Morphologically, polytene chromosomes of Drosophila melanogaster consist of compact "black" bands alternating with less compact "grey" bands and interbands. We developed a comprehensive approach that combines cytological mapping data of FlyBase-annotated genes and novel tools for predicting cytogenetic features of chromosomes on the basis of their protein composition and determined the genomic coordinates for all black bands of polytene chromosome 2R. By a PCNA immunostaining assay, we obtained the replication timetable for all the bands mapped. The results allowed us to compare replication timing between polytene chromosomes in salivary glands and chromosomes from cultured diploid cell lines and to observe a substantial similarity in the global replication patterns at the band resolution level. In both kinds of chromosomes, the intervals between black bands correspond to early replication initiation zones. Black bands are depleted of replication initiation events and are characterized by a gradient of replication timing; therefore, the time of replication completion correlates with the band length. The bands are characterized by low gene density, contain predominantly tissue-specific genes, and are represented by silent chromatin types in various tissues. The borders of black bands correspond well to the borders of topological domains as well as to the borders of the zones showing H3K27me3, SUUR, and LAMIN enrichment. In conclusion, the characteristic pattern of polytene chromosomes reflects partitioning of the Drosophila genome into two global types of domains with contrasting properties. This partitioning is conserved in different tissues and determines replication timing in Drosophila.


Subject(s)
DNA Replication Timing/physiology , Diploidy , Drosophila melanogaster/genetics , Genome, Insect , Polytene Chromosomes/genetics , Animals , Chromosomes, Insect/chemistry , Chromosomes, Insect/genetics , Chromosomes, Insect/metabolism , DNA Replication , Physical Chromosome Mapping , Polytene Chromosomes/metabolism , Salivary Glands/metabolism
10.
Sci Rep ; 8(1): 3198, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29453404

ABSTRACT

The synthesis of middle-to-late-replicating DNA can be affected independently of the rest of the genome by down-regulating the tumor suppressor PREP1 (PKNOX1). Indeed, DNA combing shows that PREP1 down-regulation affects DNA replication rate, increases the number of simultaneously firing origins and the asymmetry of DNA replication, leading to DNA damage. Genome-wide analysis of replication timing by Repli-seq shows that, upon PREP1 down-regulation, 25% of the genome is replicated earlier in the S-phase. The targeted DNA sequences correspond to Lamin-Associated Domains (LADs), and include late-replicating (LRRs) and temporal transition regions (TTRs). Notably, the distribution of PREP1 DNA binding sites and of its target genes indicates that DNA replication defects are independent of the overall PREP1 transcriptional activity. Finally, PREP1 down-regulation causes a substantial decrease in Lamin B1 levels. This suggests that DNA is released from the nuclear lamina earlier than in the control cells and is available for replication, thus explaining timing defects and DNA damage.This is the first evidence that the replication timing of a specific fraction of the human genome is affected by PREP1 tumor suppressor. This previously unknown function might significantly contribute to the genomic instability observed in human tumors.


Subject(s)
DNA Replication Timing/physiology , Genes, Tumor Suppressor/physiology , Genomic Instability , Homeodomain Proteins/physiology , Binding Sites , DNA Damage , DNA Replication Timing/genetics , Gene Expression Regulation , Genome, Human , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lamin Type B/metabolism
11.
Phys Rev E ; 95(5-1): 052403, 2017 May.
Article in English | MEDLINE | ID: mdl-28618495

ABSTRACT

Synthetic biology sets out to implement new functions in cells, and to develop a deeper understanding of biological design principles. Elowitz and Leibler [Nature (London) 403, 335 (2000)NATUAS0028-083610.1038/35002125] showed that by rational design of the reaction network, and using existing biological components, they could create a network that exhibits periodic gene expression, dubbed the repressilator. More recently, Stricker et al. [Nature (London) 456, 516 (2008)NATUAS0028-083610.1038/nature07389] presented another synthetic oscillator, called the dual-feedback oscillator, which is more stable. Detailed studies have been carried out to determine how the stability of these oscillators is affected by the intrinsic noise of the interactions between the components and the stochastic expression of their genes. However, as all biological oscillators reside in growing and dividing cells, an important question is how these oscillators are perturbed by the cell cycle. In previous work we showed that the periodic doubling of the gene copy numbers due to DNA replication can couple not only natural, circadian oscillators to the cell cycle [Paijmans et al., Proc. Natl. Acad. Sci. (USA) 113, 4063 (2016)PNASA60027-842410.1073/pnas.1507291113], but also these synthetic oscillators. Here we expand this study. We find that the strength of the locking between oscillators depends not only on the positions of the genes on the chromosome, but also on the noise in the timing of gene replication: noise tends to weaken the coupling. Yet, even in the limit of high levels of noise in the replication times of the genes, both synthetic oscillators show clear signatures of locking to the cell cycle. This work enhances our understanding of the design of robust biological oscillators inside growing and diving cells.


Subject(s)
Biological Clocks , Cell Cycle/physiology , Cell Enlargement , Models, Biological , Biological Clocks/physiology , Computer Simulation , DNA Replication Timing/physiology , Feedback, Physiological , Genes/physiology , Stochastic Processes
12.
Proc Natl Acad Sci U S A ; 114(20): 5183-5188, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28465435

ABSTRACT

One of the most fascinating features of biological systems is the ability to sustain high accuracy of all major cellular processes despite the stochastic nature of underlying chemical processes. It is widely believed that such low error values are the result of the error-correcting mechanism known as kinetic proofreading. However, it is usually argued that enhancing the accuracy should result in slowing down the process, leading to the so-called speed-accuracy trade-off. We developed a discrete-state stochastic framework that allowed us to investigate the mechanisms of the proofreading using the method of first-passage processes. With this framework, we simultaneously analyzed the speed and accuracy of the two fundamental biological processes, DNA replication and tRNA selection during the translation. The results indicate that these systems tend to optimize speed rather than accuracy, as long as the error level is tolerable. Interestingly, for these processes, certain kinetic parameters lay in the suboptimal region where their perturbations can improve both speed and accuracy. Additional constraints due to the energetic cost of proofreading also play a role in the error correcting process. Our theoretical findings provide a microscopic picture of how complex biological processes are able to function so fast with high accuracy.


Subject(s)
DNA Replication/physiology , Protein Biosynthesis/physiology , Computer Simulation , DNA Replication Timing/physiology , Kinetics , Models, Biological , Models, Theoretical , RNA, Transfer/metabolism , Stochastic Processes , Substrate Specificity , Thermodynamics , Transcription, Genetic/physiology
13.
Adv Exp Med Biol ; 1042: 287-310, 2017.
Article in English | MEDLINE | ID: mdl-29357064

ABSTRACT

Recent studies on G-quadruplex (G4) revealed crucial and conserved functions of G4 in various biological systems. We recently showed that Rif1, a conserved nuclear factor, binds to G4 present in the intergenic regions and plays a major role in spatiotemporal regulation of DNA replication. Rif1 may tether chromatin fibers through binding to G4, generating specific chromatin domains that dictate the replication timing. G4 and its various binding partners are now implicated in many other chromosome regulations, including transcription, replication initiation, recombination, gene rearrangement, and transposition.


Subject(s)
DNA Replication/physiology , G-Quadruplexes , Telomere-Binding Proteins/metabolism , Animals , DNA Breaks, Double-Stranded , DNA Replication Timing/physiology , Humans , Protein Binding , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism
14.
Cell Cycle ; 13(20): 3241-8, 2014.
Article in English | MEDLINE | ID: mdl-25485504

ABSTRACT

Transcriptional events during S-phase are critical for cell cycle progression. Here, by using a nascent RNA capture assay coupled with high-throughput sequencing, we determined the temporal patterns of transcriptional events that occur during S-phase. We show that genes involved in critical S-phase-specific biological processes such as nucleosome assembly and DNA repair have temporal transcription patterns across S-phase that are not evident from total RNA levels. By comparing transcription timing with replication timing in S-phase, we show that early replicating genes show increased transcription late in S-phase whereas late replicating genes are predominantly transcribed early in S-phase. Global anti-correlation between replication and transcription timing was observed only based on nascent RNA but not total RNA. Our data provides a detailed view of ongoing transcriptional events during the S-phase of cell cycle, and supports that transcription and replication are temporally separated.


Subject(s)
DNA Replication/physiology , Cell Cycle/genetics , Cell Cycle/physiology , DNA Replication/genetics , DNA Replication Timing/genetics , DNA Replication Timing/physiology , Humans , S Phase/genetics
15.
Cold Spring Harb Perspect Biol ; 5(8): a010132, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23838440

ABSTRACT

Patterns of replication within eukaryotic genomes correlate with gene expression, chromatin structure, and genome evolution. Recent advances in genome-scale mapping of replication kinetics have allowed these correlations to be explored in many species, cell types, and growth conditions, and these large data sets have allowed quantitative and computational analyses. One striking new correlation to emerge from these analyses is between replication timing and the three-dimensional structure of chromosomes. This correlation, which is significantly stronger than with any single histone modification or chromosome-binding protein, suggests that replication timing is controlled at the level of chromosomal domains. This conclusion dovetails with parallel work on the heterogeneity of origin firing and the competition between origins for limiting activators to suggest a model in which the stochastic probability of individual origin firing is modulated by chromosomal domain structure to produce patterns of replication. Whether these patterns have inherent biological functions or simply reflect higher-order genome structure is an open question.


Subject(s)
Chromatin/genetics , Chromosomes/genetics , DNA Replication Timing/physiology , Eukaryota/genetics , Evolution, Molecular , Gene Expression Regulation/genetics , Genome/genetics , Models, Genetic , Chromosomes/ultrastructure , Computational Biology/methods , Computational Biology/trends , Species Specificity
16.
Semin Cancer Biol ; 23(2): 80-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23327985

ABSTRACT

Normal cellular division requires that the genome be faithfully replicated to ensure that unaltered genomic information is passed from one generation to the next. DNA replication initiates from thousands of origins scattered throughout the genome every cell cycle; however, not all origins initiate replication at the same time. A vast amount of work over the years indicates that different origins along each eukaryotic chromosome are activated in early, middle or late S phase. This temporal control of DNA replication is referred to as the replication-timing program. The replication-timing program represents a very stable epigenetic feature of chromosomes. Recent evidence has indicated that the replication-timing program can influence the spatial distribution of mutagenic events such that certain regions of the genome experience increased spontaneous mutagenesis compared to surrounding regions. This influence has helped shape the genomes of humans and other multicellular organisms and can affect the distribution of mutations in somatic cells. It is also becoming clear that the replication-timing program is deregulated in many disease states, including cancer. Aberrant DNA replication timing is associated with changes in gene expression, changes in epigenetic modifications and an increased frequency of structural rearrangements. Furthermore, certain replication timing changes can directly lead to overt genomic instability and may explain unique mutational signatures that are present in cells that have undergone the recently described processes of "chromothripsis" and "kataegis". In this review, we will discuss how the normal replication timing program, as well as how alterations to this program, can contribute to the evolution of the genomic landscape in normal and cancerous cells.


Subject(s)
DNA Replication Timing/physiology , DNA Replication/genetics , Genomic Instability/genetics , Neoplasms/genetics , Animals , DNA Replication Timing/genetics , Epigenesis, Genetic/physiology , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Time Factors
17.
Biol Reprod ; 87(3): 62, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22674395

ABSTRACT

In eukaryotes, DNA synthesis is preceded by licensing of replication origins. We examined the subcellular localization of two licensing proteins, ORC2 and MCM7, in the mouse zygotes and two-cell embryos. In somatic cells ORC2 remains bound to DNA replication origins throughout the cell cycle, while MCM7 is one of the last proteins to bind to the licensing complex. We found that MCM7 but not ORC2 was bound to DNA in metaphase II oocytes and remained associated with the DNA until S-phase. Shortly after fertilization, ORC2 was detectable at the metaphase II spindle poles and then between the separating chromosomes. Neither protein was present in the sperm cell at fertilization. As the sperm head decondensed, MCM7 was bound to DNA, but no ORC2 was seen. By 4 h after fertilization, both pronuclei contained DNA bound ORC2 and MCM7. As expected, during S-phase of the first zygotic cell cycle, MCM7 was released from the DNA, but ORC2 remained bound. During zygotic mitosis, ORC2 again localized first to the spindle poles, then to the area between the separating chromosomes. ORC2 then formed a ring around the developing two-cell nuclei before entering the nucleus. Only soluble MCM7 was present in the G2 pronuclei, but by zygotic metaphase it was bound to DNA, again apparently before ORC2. In G1 of the two-cell stage, both nuclei had salt-resistant ORC2 and MCM7. These data suggest that licensing follows a unique pattern in the early zygote that differs from what has been described for other mammalian cells that have been studied.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication/physiology , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Origin Recognition Complex/metabolism , Zygote/metabolism , Animals , Chromatin/metabolism , DNA Replication Timing/physiology , Embryo, Mammalian , Female , Fertilization/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Minichromosome Maintenance Complex Component 7 , Models, Biological , Tissue Distribution , Zygote/ultrastructure
18.
Epigenomics ; 3(5): 543-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22126245

ABSTRACT

The Chromatin, Replication and Chromosomal Stability Conference took place on June 20-21 in Stockholm, Sweden. In this article, I outline the broad scientific program of the meeting which reflected the wide diversity in epigenetics research. Distinct histone modifications are linked with specific chromatin structures and intranuclear positioning, thereby impacting replication timing and replication initiation, which in turn are related to gene expression and cell differentiation. Interference in any of these interconnected mechanisms can result in DNA breakage and lead to the activation of repair pathways. The DNA repair mechanisms again are influenced by the chromatin structure. In summary, the conference highlighted the functional implication of epigenetics in chromatin compaction, transcription regulation, replication control and DNA repair. The tight control of all these mechanisms defines the final cellular character.


Subject(s)
Chromatin/physiology , Chromosomal Instability , Congresses as Topic , DNA Repair/physiology , DNA Replication Timing/physiology , DNA Replication/physiology , Epigenomics/methods , Epigenomics/trends
19.
BMC Syst Biol ; 5: 201, 2011 Dec 21.
Article in English | MEDLINE | ID: mdl-22189092

ABSTRACT

BACKGROUND: In Escherichia coli, overlapping rounds of DNA replication allow the bacteria to double in faster times than the time required to copy the genome. The precise timing of initiation of DNA replication is determined by a regulatory circuit that depends on the binding of a critical number of ATP-bound DnaA proteins at the origin of replication, resulting in the melting of the DNA and the assembly of the replication complex. The synthesis of DnaA in the cell is controlled by a growth-rate dependent, negatively autoregulated gene found near the origin of replication. Both the regulatory and initiation activity of DnaA depend on its nucleotide bound state and its availability. RESULTS: In order to investigate the contributions of the different regulatory processes to the timing of initiation of DNA replication at varying growth rates, we formulate a minimal quantitative model of the initiator circuit that includes the key ingredients known to regulate the activity of the DnaA protein. This model describes the average-cell oscillations in DnaA-ATP/DNA during the cell cycle, for varying growth rates. We evaluate the conditions under which this ratio attains the same threshold value at the time of initiation, independently of the growth rate. CONCLUSIONS: We find that a quantitative description of replication initiation by DnaA must rely on the dependency of the basic parameters on growth rate, in order to account for the timing of initiation of DNA replication at different cell doubling times. We isolate two main possible scenarios for this, depending on the roles of DnaA autoregulation and DnaA ATP-hydrolysis regulatory process. One possibility is that the basal rate of regulatory inactivation by ATP hydrolysis must vary with growth rate. Alternatively, some parameters defining promoter activity need to be a function of the growth rate. In either case, the basal rate of gene expression needs to increase with the growth rate, in accordance with the known characteristics of the dnaA promoter. Furthermore, both inactivation and autorepression reduce the amplitude of the cell-cycle oscillations of DnaA-ATP/DNA.


Subject(s)
Bacterial Proteins/metabolism , DNA Replication Timing/physiology , DNA-Binding Proteins/metabolism , Escherichia coli/growth & development , Escherichia coli/genetics , Gene Expression Regulation, Fungal/physiology , Models, Biological , Adenosine Triphosphate/metabolism , Bacterial Proteins/physiology , Computer Simulation , DNA-Binding Proteins/physiology , Gene Expression Regulation, Fungal/genetics , Hydrolysis
20.
PLoS One ; 6(11): e27413, 2011.
Article in English | MEDLINE | ID: mdl-22096571

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is linked to contraction of an array of tandem 3.3-kb repeats (D4Z4) at 4q35.2 from 11-100 copies to 1-10 copies. The extent to which D4Z4 contraction at 4q35.2 affects overall 4q35.2 chromatin organization remains unclear. Because DNA replication timing is highly predictive of long-range chromatin interactions, we generated genome-wide replication-timing profiles for FSHD and control myogenic precursor cells. We compared non-immortalized myoblasts from four FSHD patients and three control individuals to each other and to a variety of other human cell types. This study also represents the first genome-wide comparison of replication timing profiles in non-immortalized human cell cultures. Myoblasts from both control and FSHD individuals all shared a myoblast-specific replication profile. In contrast, male and female individuals were readily distinguished by monoallelic differences in replication timing at DXZ4 and other regions across the X chromosome affected by X inactivation. We conclude that replication timing is a robust cell-type specific feature that is unaffected by FSHD-related D4Z4 contraction.


Subject(s)
DNA Replication Timing/physiology , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts/metabolism , Tandem Repeat Sequences/genetics , Adolescent , Adult , Cells, Cultured , DNA Replication Timing/genetics , Female , Humans , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...