Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 280
Filter
Add more filters

Publication year range
1.
Nat Rev Mol Cell Biol ; 16(6): 360-74, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25999062

ABSTRACT

DNA replication begins with the assembly of pre-replication complexes (pre-RCs) at thousands of DNA replication origins during the G1 phase of the cell cycle. At the G1-S-phase transition, pre-RCs are converted into pre-initiation complexes, in which the replicative helicase is activated, leading to DNA unwinding and initiation of DNA synthesis. However, only a subset of origins are activated during any S phase. Recent insights into the mechanisms underlying this choice reveal how flexibility in origin usage and temporal activation are linked to chromosome structure and organization, cell growth and differentiation, and replication stress.


Subject(s)
DNA Replication/physiology , DNA/biosynthesis , G1 Phase/physiology , Replication Origin/physiology , S Phase/physiology , Animals , Cell Differentiation/physiology , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , DNA/genetics , Humans
2.
Mol Cell ; 70(6): 1067-1080.e12, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29944888

ABSTRACT

The replisome must overcome DNA damage to ensure complete chromosome replication. Here, we describe the earliest events in this process by reconstituting collisions between a eukaryotic replisome, assembled with purified proteins, and DNA damage. Lagging-strand lesions are bypassed without delay, leaving daughter-strand gaps roughly the size of an Okazaki fragment. In contrast, leading-strand polymerase stalling significantly impacts replication fork progression. We reveal that the core replisome itself can bypass leading-strand damage by re-priming synthesis beyond it. Surprisingly, this restart activity is rare, mainly due to inefficient leading-strand re-priming, rather than single-stranded DNA exposure or primer extension. We find several unanticipated mechanistic distinctions between leading- and lagging-strand priming that we propose control the replisome's initial response to DNA damage. Notably, leading-strand restart was specifically stimulated by RPA depletion, which can occur under conditions of replication stress. Our results have implications for pathway choice at stalled forks and priming at DNA replication origins.


Subject(s)
DNA Repair/physiology , DNA Replication/physiology , DNA/metabolism , DNA Damage/physiology , DNA Primase/metabolism , DNA Repair/genetics , DNA, Single-Stranded/metabolism , Eukaryota/genetics , Eukaryotic Cells/metabolism , Replication Origin/genetics , Replication Origin/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
3.
Genes Dev ; 31(11): 1073-1088, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28717046

ABSTRACT

DNA replication results in the doubling of the genome prior to cell division. This process requires the assembly of 50 or more protein factors into a replication fork. Here, we review recent structural and biochemical insights that start to explain how specific proteins recognize DNA replication origins, load the replicative helicase on DNA, unwind DNA, synthesize new DNA strands, and reassemble chromatin. We focus on the minichromosome maintenance (MCM2-7) proteins, which form the core of the eukaryotic replication fork, as this complex undergoes major structural rearrangements in order to engage with DNA, regulate its DNA-unwinding activity, and maintain genome stability.


Subject(s)
DNA Replication/physiology , Animals , Chromatin/metabolism , DNA Helicases/metabolism , DNA Replication/genetics , Evolution, Molecular , Genomic Instability/genetics , Humans , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/metabolism , Replication Origin/physiology
4.
Genes Dev ; 31(23-24): 2405-2415, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29330352

ABSTRACT

Initiation of eukaryotic chromosome replication follows a spatiotemporal program. The current model suggests that replication origins compete for a limited pool of initiation factors. However, it remains to be answered how these limiting factors are preferentially recruited to early origins. Here, we report that Dbf4 is enriched at early origins through its interaction with forkhead transcription factors Fkh1 and Fkh2. This interaction is mediated by the Dbf4 C terminus and was successfully reconstituted in vitro. An interaction-defective mutant, dbf4ΔC, phenocopies fkh alleles in terms of origin firing. Remarkably, genome-wide replication profiles reveal that the direct fusion of the DNA-binding domain (DBD) of Fkh1 to Dbf4 restores the Fkh-dependent origin firing but interferes specifically with the pericentromeric origin activation. Furthermore, Dbf4 interacts directly with Sld3 and promotes the recruitment of downstream limiting factors. These data suggest that Fkh1 targets Dbf4 to a subset of noncentromeric origins to promote early replication in a manner that is reminiscent of the recruitment of Dbf4 to pericentromeric origins by Ctf19.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Transcription Factors/metabolism , Replication Origin/physiology , Saccharomyces cerevisiae Proteins/metabolism , Cell Cycle Proteins/genetics , DNA Replication/genetics , DNA-Binding Proteins/metabolism , Genome, Fungal/genetics , Mutation , Nuclear Proteins/metabolism , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Replication Origin/genetics , Saccharomyces cerevisiae Proteins/genetics
5.
Genes Dev ; 28(1): 71-84, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24395248

ABSTRACT

At all but the slowest growth rates, Escherichia coli cell cycles overlap, and its nucleoid is segregated to daughter cells as a forked DNA circle with replication ongoing-a state fundamentally different from eukaryotes. We have solved the chromosome organization, structural dynamics, and segregation of this constantly replicating chromosome. It is locally condensed to form a branched donut, compressed so that the least replicated DNA spans the cell center and the newest DNA extends toward the cell poles. Three narrow zones at the cell center and quarters contain both the replication forks and nascent DNA and serve to segregate the duplicated chromosomal information as it flows outward. The overall pattern is smoothly self-replicating, except when the duplicated terminus region is released from the septum and recoils to the center of a sister nucleoid. In circular cross-section of the cell, the left and right arms of the chromosome form separate, parallel structures that lie in each cell half along the radial cell axis. In contrast, replication forks and origin and terminus regions are found mostly at the center of the cross section, balanced by the parallel chromosome arms. The structure is consistent with the model in which the nucleoid is a constrained ring polymer that develops by spontaneous thermodynamics. The ring polymer pattern extrapolates to higher growth rates and also provides a structural basis for the form of the chromosome during very slow growth.


Subject(s)
Chromosomes, Bacterial/genetics , Chromosomes, Bacterial/metabolism , DNA Replication/physiology , Escherichia coli/genetics , Escherichia coli/metabolism , Replication Origin/physiology , DNA Replication/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Polymers/metabolism
6.
Genes Dev ; 28(15): 1653-66, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25085418

ABSTRACT

The regulated loading of the replicative helicase minichromosome maintenance proteins 2-7 (MCM2-7) onto replication origins is a prerequisite for replication fork establishment and genomic stability. Origin recognition complex (ORC), Cdc6, and Cdt1 assemble two MCM2-7 hexamers into one double hexamer around dsDNA. Although the MCM2-7 hexamer can adopt a ring shape with a gap between Mcm2 and Mcm5, it is unknown which Mcm interface functions as the DNA entry gate during regulated helicase loading. Here, we establish that the Saccharomyces cerevisiae MCM2-7 hexamer assumes a closed ring structure, suggesting that helicase loading requires active ring opening. Using a chemical biology approach, we show that ORC-Cdc6-Cdt1-dependent helicase loading occurs through a unique DNA entry gate comprised of the Mcm2 and Mcm5 subunits. Controlled inhibition of DNA insertion triggers ATPase-driven complex disassembly in vitro, while in vivo analysis establishes that Mcm2/Mcm5 gate opening is essential for both helicase loading onto chromatin and cell cycle progression. Importantly, we demonstrate that the MCM2-7 helicase becomes loaded onto DNA as a single hexamer during ORC/Cdc6/Cdt1/MCM2-7 complex formation prior to MCM2-7 double hexamer formation. Our study establishes the existence of a unique DNA entry gate for regulated helicase loading, revealing key mechanisms in helicase loading, which has important implications for helicase activation.


Subject(s)
DNA, Fungal/metabolism , Minichromosome Maintenance Proteins/metabolism , Protein Subunits/metabolism , Saccharomyces cerevisiae/enzymology , Adenosine Triphosphate/metabolism , Cell Cycle , Chromosomes, Fungal/metabolism , Enzyme Activation , Hydrolysis , Minichromosome Maintenance Proteins/chemistry , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/ultrastructure , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Protein Subunits/chemistry , Protein Subunits/genetics , Replication Origin/physiology , Saccharomyces cerevisiae/genetics
7.
PLoS Pathog ; 15(12): e1008228, 2019 12.
Article in English | MEDLINE | ID: mdl-31841561

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic herpesvirus and WHO class 1 carcinogen that resides in B lymphocytes of nearly all humans. While silent in most, EBV can cause endemic Burkitt lymphoma in children and post-transplant lymphoproliferative disorders/lymphomas in immunocompromised hosts. The pathogenesis of such lymphomas is multifactorial but to a large extent depends on EBV's ability to aggressively drive cellular DNA replication and B cell proliferation despite cell-intrinsic barriers to replication. One such barrier is oncogenic replication stress which hinders the progression of DNA replication forks. To understand how EBV successfully overcomes replication stress, we examined cellular replication forks in EBV-transformed B cells using iPOND (isolation of Proteins on Nascent DNA)-mass spectrometry and identified several cellular proteins that had not previously been linked to DNA replication. Of eight candidate replisome-associated proteins that we validated at forks in EBV-transformed cells and Burkitt lymphoma-derived cells, three zinc finger proteins (ZFPs) were upregulated early in B cells newly-infected with EBV in culture as well as expressed at high levels in EBV-infected B blasts in the blood of immunocompromised transplant recipients. Expressed highly in S- and G2-phase cells, knockdown of each ZFP resulted in stalling of proliferating cells in the S-phase, cleavage of caspase 3, and cell death. These proteins, newly-identified at replication forks of EBV-transformed and Burkitt lymphoma cells therefore contribute to cell survival and cell cycle progression, and represent novel targets for intervention of EBV-lymphomas while simultaneously offering a window into how the replication machinery may be similarly modified in other cancers.


Subject(s)
B-Lymphocytes/virology , Cell Transformation, Viral/physiology , Epstein-Barr Virus Infections/metabolism , Replication Origin/physiology , Zinc Fingers/physiology , B-Lymphocytes/pathology , Burkitt Lymphoma/virology , Cell Proliferation/physiology , Herpesvirus 4, Human , Humans
8.
Plant Physiol ; 183(1): 206-220, 2020 05.
Article in English | MEDLINE | ID: mdl-32205451

ABSTRACT

The selection and firing of DNA replication origins play key roles in ensuring that eukaryotes accurately replicate their genomes. This process is not well documented in plants due in large measure to difficulties in working with plant systems. We developed a new functional assay to label and map very early replicating loci that must, by definition, include at least a subset of replication origins. Arabidopsis (Arabidopsis thaliana) cells were briefly labeled with 5-ethynyl-2'-deoxy-uridine, and nuclei were subjected to two-parameter flow sorting. We identified more than 5500 loci as initiation regions (IRs), the first regions to replicate in very early S phase. These were classified as strong or weak IRs based on the strength of their replication signals. Strong initiation regions were evenly spaced along chromosomal arms and depleted in centromeres, while weak initiation regions were enriched in centromeric regions. IRs are AT-rich sequences flanked by more GC-rich regions and located predominantly in intergenic regions. Nuclease sensitivity assays indicated that IRs are associated with accessible chromatin. Based on these observations, initiation of plant DNA replication shows some similarity to, but is also distinct from, initiation in other well-studied eukaryotic systems.


Subject(s)
Arabidopsis/metabolism , Chromatin/metabolism , DNA, Plant/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , DNA Replication/genetics , DNA Replication/physiology , DNA, Plant/physiology , Replication Origin/genetics , Replication Origin/physiology
9.
Nature ; 519(7544): 431-5, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25739503

ABSTRACT

Eukaryotic cells initiate DNA replication from multiple origins, which must be tightly regulated to promote precise genome duplication in every cell cycle. To accomplish this, initiation is partitioned into two temporally discrete steps: a double hexameric minichromosome maintenance (MCM) complex is first loaded at replication origins during G1 phase, and then converted to the active CMG (Cdc45-MCM-GINS) helicase during S phase. Here we describe the reconstitution of budding yeast DNA replication initiation with 16 purified replication factors, made from 42 polypeptides. Origin-dependent initiation recapitulates regulation seen in vivo. Cyclin-dependent kinase (CDK) inhibits MCM loading by phosphorylating the origin recognition complex (ORC) and promotes CMG formation by phosphorylating Sld2 and Sld3. Dbf4-dependent kinase (DDK) promotes replication by phosphorylating MCM, and can act either before or after CDK. These experiments define the minimum complement of proteins, protein kinase substrates and co-factors required for regulated eukaryotic DNA replication.


Subject(s)
DNA Replication , Replication Origin/physiology , Saccharomyces cerevisiae Proteins/isolation & purification , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Minichromosome Maintenance Proteins/metabolism , Multienzyme Complexes/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Replication Origin/genetics , Replication Protein A/metabolism , Saccharomyces cerevisiae/enzymology
10.
EMBO J ; 35(2): 176-92, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26620551

ABSTRACT

During DNA replication, thousands of replication origins are activated across the genome. Chromatin architecture contributes to origin specification and usage, yet it remains unclear which chromatin features impact on DNA replication. Here, we perform a RNAi screen for chromatin regulators implicated in replication control by measuring RPA accumulation upon replication stress. We identify six factors required for normal rates of DNA replication and characterize a function of the bromodomain and PHD finger-containing protein 3 (BRPF3) in replication initiation. BRPF3 forms a complex with HBO1 that specifically acetylates histone H3K14, and genomewide analysis shows high enrichment of BRPF3, HBO1 and H3K14ac at ORC1-binding sites and replication origins found in the vicinity of TSSs. Consistent with this, BRPF3 is necessary for H3K14ac at selected origins and efficient origin activation. CDC45 recruitment, but not MCM2-7 loading, is impaired in BRPF3-depleted cells, identifying a BRPF3-dependent function of HBO1 in origin activation that is complementary to its role in licencing. We thus propose that BRPF3-HBO1 acetylation of histone H3K14 around TSS facilitates efficient activation of nearby replication origins.


Subject(s)
Cell Cycle/physiology , Histone Acetyltransferases/metabolism , Histones/metabolism , Replication Origin/physiology , Acetylation , Cell Cycle/genetics , Cell Line , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA Replication/genetics , DNA Replication/physiology , Histone Acetyltransferases/genetics , Humans , Immunohistochemistry , Replication Origin/genetics
11.
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
12.
Genes Dev ; 26(18): 2050-62, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22987637

ABSTRACT

In eukaryotes, the replication of chromosome DNA is coordinated by a replication timing program that temporally regulates the firing of individual replication origins. However, the molecular mechanism underlying the program remains elusive. Here, we report that the telomere-binding protein Taz1 plays a crucial role in the control of replication timing in fission yeast. A DNA element located proximal to a late origin in the chromosome arm represses initiation from the origin in early S phase. Systematic deletion and substitution experiments demonstrated that two tandem telomeric repeats are essential for this repression. The telomeric repeats recruit Taz1, a counterpart of human TRF1 and TRF2, to the locus. Genome-wide analysis revealed that Taz1 regulates about half of chromosomal late origins, including those in subtelomeres. The Taz1-mediated mechanism prevents Dbf4-dependent kinase (DDK)-dependent Sld3 loading onto the origins. Our results demonstrate that the replication timing program in fission yeast uses the internal telomeric repeats and binding of Taz1.


Subject(s)
DNA Replication/physiology , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/physiology , Telomere-Binding Proteins/metabolism , Base Sequence , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA-Binding Proteins/metabolism , Molecular Sequence Data , Protein Binding , Protein Transport , Replication Origin/physiology , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Telomere-Binding Proteins/genetics
13.
Genes Dev ; 26(2): 137-50, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22279046

ABSTRACT

One of the long-standing questions in eukaryotic DNA replication is the mechanisms that determine where and when a particular segment of the genome is replicated. Cdc7/Hsk1 is a conserved kinase required for initiation of DNA replication and may affect the site selection and timing of origin firing. We identified rif1Δ, a null mutant of rif1(+), a conserved telomere-binding factor, as an efficient bypass mutant of fission yeast hsk1. Extensive deregulation of dormant origins over a wide range of the chromosomes occurs in rif1Δ in the presence or absence of hydroxyurea (HU). At the same time, many early-firing, efficient origins are suppressed or delayed in firing timing in rif1Δ. Rif1 binds not only to telomeres, but also to many specific locations on the arm segments that only partially overlap with the prereplicative complex assembly sites, although Rif1 tends to bind in the vicinity of the late/dormant origins activated in rif1Δ. The binding to the arm segments occurs through M to G1 phase in a manner independent of Taz1 and appears to be essential for the replication timing program during the normal cell cycle. Our data demonstrate that Rif1 is a critical determinant of the origin activation program on the fission yeast chromosomes.


Subject(s)
DNA Replication Timing/genetics , Replication Origin/physiology , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Telomere-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Survival/physiology , Centromere/metabolism , DNA Replication/genetics , G1 Phase , Gene Deletion , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/genetics , Shelterin Complex , Telomere/metabolism , Telomere-Binding Proteins/genetics
14.
Mol Microbiol ; 107(1): 68-80, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29024073

ABSTRACT

Few studies have described chromosomal dynamics in bacterial cells with more than two complete chromosome copies or described changes with respect to development in polyploid cells. We examined the arrangement of chromosomal loci in the very large, highly polyploid, uncultivated intestinal symbiont Epulopiscium sp. type B using fluorescent in situ hybridization. We found that in new offspring, chromosome replication origins (oriCs) are arranged in a three-dimensional array throughout the cytoplasm. As development progresses, most oriCs become peripherally located. Siblings within a mother cell have similar numbers of oriCs. When chromosome orientation was assessed in situ by labeling two chromosomal regions, no specific pattern was detected. The Epulopiscium genome codes for many of the conserved positional guide proteins used for chromosome segregation in bacteria. Based on this study, we present a model that conserved chromosomal maintenance proteins, combined with entropic demixing, provide the forces necessary for distributing oriCs. Without the positional regulation afforded by radial confinement, chromosomes are more randomly oriented in Epulopiscium than in most small rod-shaped cells. Furthermore, we suggest that the random orientation of individual chromosomes in large polyploid cells would not hamper reproductive success as it would in smaller cells with more limited genomic resources.


Subject(s)
Chromosome Segregation/physiology , Clostridiales/metabolism , Replication Origin/physiology , Bacteria/genetics , Bacterial Proteins/metabolism , Clostridiales/genetics , DNA Replication/genetics , DNA, Bacterial/metabolism , In Situ Hybridization, Fluorescence , Polyploidy , Replication Origin/genetics
15.
Genes Dev ; 25(6): 621-33, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21406556

ABSTRACT

Although many chemotherapy drugs activate the intra-S-phase checkpoint pathway to block S-phase progression, not much is known about how and where the intra-S-phase checkpoint regulates origins of replication in human chromosomes. A genomic analysis of replication in human cells in the presence of hydroxyurea (HU) revealed that only the earliest origins fire, but the forks stall within 2 kb and neighboring clusters of dormant origins are activated. The initiation events are located near expressed genes with a preference for transcription start and end sites, and when they are located in intergenic regions they are located near regulatory factor-binding regions (RFBR). The activation of clustered neo-origins by HU suggests that there are many potential replication initiation sites in permissive parts of the genome, most of which are not used in a normal S phase. Consistent with this redundancy, we see multiple sites bound to MCM3 (representative of the helicase) in the region flanking three out of three origins studied in detail. Bypass of the intra-S-phase checkpoint by caffeine activates many new origins in mid- and late-replicating parts of the genome. The intra-S-phase checkpoint suppresses origin firing after the loading of Mcm10, but before the recruitment of Cdc45 and AND-1/CTF4; i.e., after helicase loading but before helicase activation and polymerase loading. Interestingly, Cdc45 recruitment upon checkpoint bypass was accompanied by the restoration of global Cdk2 kinase activity and decrease in both global and origin-bound histone H3 Lys 4 trimethylation (H3K4me3), consistent with the suggestion that both of these factors are important for Cdc45 recruitment.


Subject(s)
Replication Origin/physiology , S Phase/physiology , Caffeine/pharmacology , Cell Cycle Proteins/metabolism , Cell Proliferation , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Hydroxyurea/pharmacology , Minichromosome Maintenance Proteins , Phosphodiesterase Inhibitors/pharmacology , Transcription Initiation Site/drug effects
16.
J Biol Chem ; 292(52): 21417-21430, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29074622

ABSTRACT

In eukaryotes, DNA replication initiates from multiple origins of replication for timely genome duplication. These sites are selected by origin licensing, during which the core enzyme of the eukaryotic DNA replicative helicase, the Mcm2-7 (minichromosome maintenance) complex, is loaded at each origin. This origin licensing requires loading two Mcm2-7 helicases around origin DNA in a head-to-head orientation. Current models suggest that the origin-recognition complex (ORC) and cell-division cycle 6 (Cdc6) proteins recognize and encircle origin DNA and assemble an Mcm2-7 double-hexamer around adjacent double-stranded DNA. To test this model and assess the location of Mcm2-7 initial loading, we placed DNA-protein roadblocks at defined positions adjacent to the essential ORC-binding site within Saccharomyces cerevisiae origin DNA. Roadblocks were made either by covalent cross-linking of the HpaII methyltransferase to DNA or through binding of a transcription activator-like effector (TALE) protein. Contrary to the sites of Mcm2-7 recruitment being precisely defined, only single roadblocks that inhibited ORC-DNA binding showed helicase loading defects. We observed inhibition of helicase loading without inhibition of ORC-DNA binding only when roadblocks were placed on both sides of the origin to restrict sliding of a helicase-loading intermediate. Consistent with a sliding helicase-loading intermediate, when either one of the flanking roadblocks was eliminated, the remaining roadblock had no effect on helicase loading. Interestingly, either origin-flanking nucleosomes or roadblocks resulted in helicase loading being dependent on an additional origin sequence known to be a weaker ORC-DNA-binding site. Together, our findings support a model in which sliding helicase-loading intermediates increase the flexibility of the DNA sequence requirements for origin licensing.


Subject(s)
Arabidopsis Proteins/metabolism , Cell Cycle Proteins/metabolism , Minichromosome Maintenance Proteins/metabolism , Binding Sites , Crystallography, X-Ray , DNA Replication/genetics , DNA Replication/physiology , Minichromosome Maintenance Complex Component 7/metabolism , Minichromosome Maintenance Proteins/physiology , Origin Recognition Complex/genetics , Protein Binding , Protein Domains , Replication Origin/genetics , Replication Origin/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
17.
J Biol Chem ; 292(15): 6056-6075, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28223353

ABSTRACT

A central step in the initiation of chromosomal DNA replication in eukaryotes is the assembly of pre-replicative complex (pre-RC) at late M and early G1 phase of the cell cycles. Since 1973, four proteins or protein complexes, including cell division control protein 6 (Cdc6)/Cdc18, minichromosome maintenance protein complex, origin recognition complex (ORC), and Cdt1, are known components of the pre-RC. Previously, we reported that a non-ORC protein binds to the essential element Δ9 of the Schizosaccharomyces pombe DNA-replication origin ARS3001. In this study, we identified that the non-ORC protein is Sap1. Like ORC, Sap1 binds to DNA origins during cell growth cycles. But unlike ORC, which binds to asymmetric AT-rich sequences through its nine AT-hook motifs, Sap1 preferentially binds to a DNA sequence of 5'-(A/T) n (C/G)(A/T)9-10(G/C)(A/T) n -3' (n ≥ 1). We also found that Sap1 and ORC physically interact. We further demonstrated that Sap1 is required for the assembly of the pre-RC because of its essential role in recruiting Cdc18 to DNA origins. Thus, we conclude that Sap1 is a replication-initiation factor that directly participates in the assembly of the pre-RC. DNA-replication origins in fission yeast are defined by possessing two essential elements with one bound by ORC and the other by Sap1.


Subject(s)
DNA Replication/physiology , DNA, Fungal/biosynthesis , DNA-Binding Proteins/metabolism , Nucleotide Motifs/physiology , Replication Origin/physiology , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA, Fungal/genetics , DNA-Binding Proteins/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics
18.
BMC Genomics ; 19(1): 623, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-30134926

ABSTRACT

BACKGROUND: Genomic regions repressed for DNA replication, resulting in either delayed replication in S phase or underreplication in polyploid cells, are thought to be controlled by inhibition of replication origin activation. Studies in Drosophila polytene cells, however, raised the possibility that impeding replication fork progression also plays a major role. RESULTS: We exploited genomic regions underreplicated (URs) with tissue specificity in Drosophila polytene cells to analyze mechanisms of replication repression. By localizing the Origin Recognition Complex (ORC) in the genome of the larval fat body and comparing this to ORC binding in the salivary gland, we found that sites of ORC binding show extensive tissue specificity. In contrast, there are common domains nearly devoid of ORC in the salivary gland and fat body that also have reduced density of ORC binding sites in diploid cells. Strikingly, domains lacking ORC can still be replicated in some polytene tissues, showing absence of ORC and origins is insufficient to repress replication. Analysis of the width and location of the URs with respect to ORC position indicates that whether or not a genomic region lacking ORC is replicated is controlled by whether replication forks formed outside the region are inhibited. CONCLUSIONS: These studies demonstrate that inhibition of replication fork progression can block replication across genomic regions that constitutively lack ORC. Replication fork progression can be inhibited in both tissue-specific and genome region-specific ways. Consequently, when evaluating sources of genome instability it is important to consider altered control of replication forks in response to differentiation.


Subject(s)
Cell Differentiation/genetics , Chromosome Structures , DNA Replication/genetics , Organogenesis/genetics , Origin Recognition Complex/metabolism , Replication Origin/physiology , Animals , Binding Sites , Chromosome Structures/chemistry , Chromosome Structures/genetics , Chromosome Structures/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Larva , Organ Specificity/genetics
19.
EMBO J ; 33(6): 605-20, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24566989

ABSTRACT

Eukaryotic DNA replication initiates from multiple replication origins. To ensure each origin fires just once per cell cycle, initiation is divided into two biochemically discrete steps: the Mcm2-7 helicase is first loaded into prereplicative complexes (pre-RCs) as an inactive double hexamer by the origin recognition complex (ORC), Cdt1 and Cdc6; the helicase is then activated by a set of "firing factors." Here, we show that plasmids containing pre-RCs assembled with purified proteins support complete and semi-conservative replication in extracts from budding yeast cells overexpressing firing factors. Replication requires cyclin-dependent kinase (CDK) and Dbf4-dependent kinase (DDK). DDK phosphorylation of Mcm2-7 does not by itself promote separation of the double hexamer, but is required for the recruitment of firing factors and replisome components in the extract. Plasmid replication does not require a functional replication origin; however, in the presence of competitor DNA and limiting ORC concentrations, replication becomes origin-dependent in this system. These experiments indicate that Mcm2-7 double hexamers can be precursors of replication and provide insight into the nature of eukaryotic DNA replication origins.


Subject(s)
DNA Replication/physiology , Enzyme Activation/physiology , Minichromosome Maintenance Proteins/metabolism , Multiprotein Complexes/physiology , Replication Origin/physiology , Cell Cycle Proteins/metabolism , Mass Spectrometry , Models, Biological , Models, Molecular , Phosphorylation , Plasmids/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomycetales
20.
Nature ; 492(7428): 205-9, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23201686

ABSTRACT

Replicative DNA helicases generally unwind DNA as a single hexamer that encircles and translocates along one strand of the duplex while excluding the complementary strand (known as steric exclusion). By contrast, large T antigen, the replicative DNA helicase of the simian virus 40 (SV40), is reported to function as a pair of stacked hexamers that pumps double-stranded DNA through its central channel while laterally extruding single-stranded DNA. Here we use single-molecule and ensemble assays to show that large T antigen assembled on the SV40 origin unwinds DNA efficiently as a single hexamer that translocates on single-stranded DNA in the 3'-to-5' direction. Unexpectedly, large T antigen unwinds DNA past a DNA-protein crosslink on the translocation strand, suggesting that the large T antigen ring can open to bypass bulky adducts. Together, our data underscore the profound conservation among replicative helicase mechanisms, and reveal a new level of plasticity in the interactions of replicative helicases with DNA damage.


Subject(s)
DNA Helicases/metabolism , Simian virus 40/enzymology , Antigens, Viral, Tumor/metabolism , DNA Replication , DNA, Single-Stranded/metabolism , DNA, Viral/metabolism , Replication Origin/physiology , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL