ARTICLE
Received 21 Oct 2015 | Accepted 26 Apr 2016 | Published 8 Jun 2016
Ya Zhang1,*, Liang Huang1,*, Haiqing Fu1, Owen K. Smith1, Chii Mei Lin1, Koichi Utani1, Mishal Rao1, William C. Reinhold1, Christophe E. Redon1, Michael Ryan2, RyangGuk Kim2, Yang You1, Harlington Hanna1, Yves Boisclair3, Qiaoming Long3 & Mirit I. Aladjem1
Mammalian chromosome replication starts from distinct sites; however, the principles governing initiation site selection are unclear because proteins essential for DNA replication do not exhibit sequence-specic DNA binding. Here we identify a replication-initiation determinant (RepID) protein that binds a subset of replication-initiation sites. A large fraction of RepID-binding sites share a common G-rich motif and exhibit elevated replication initiation. RepID is required for initiation of DNA replication from RepID-bound replication origins, including the origin at the human beta-globin (HBB) locus. At HBB, RepID is involved in an interaction between the replication origin (Rep-P) and the locus control region. RepID-depleted murine embryonic broblasts exhibit abnormal replication fork progression and fewer replication-initiation events. These observations are consistent with a model, suggesting that RepID facilitates replication initiation at a distinct group of human replication origins.
DOI: 10.1038/ncomms11748 OPEN
A replicator-specic binding protein essential for site-specic initiation of DNA replication in mammalian cells
1 Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
2 In Silico Solutions, Fairfax, Virginia 22033, USA. 3 Department of Animal Science, Cornell University, Ithaca, New York 14853-4801, USA. * These authors contributed equally to this work. Correspondence and requests for materials should be addressed to M.I.A. (email: mailto:[email protected]
Web End [email protected] ).
NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 1
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748
All eukaryotic proliferating cells duplicate their entire genome with high delity during the S phase of the cell cycle. This duplication requires precise coordination
between DNA replication, cell cycle progression, chromatin remodelling and transcription. In eukaryotes, DNA replication initiates from distinct sites within the genome (replication origins), which can vary depending on the cell type and developmental stage17. To initiate DNA replication, a licensing step is required8. Following mitosis, chromatin-bound origin recognition complex (ORC) proteins recruit the licensing factors CDC6 and CDT1 to facilitate the loading of minichromosome maintenance helicases, and the formation of a pre-replication complex911. Specic ORC-binding determines replication origin activity in yeast; however, in metazoans, proteins that are essential for replication do not bind to specic DNA sequences12,13. Pre-replication complexes modify nucleosome-positioning in both Drosophila14 and vertebrates15, and the ORC1 member of the pre-replication complex binds preferentially to open chromatin16. However, these associations are not sufcient to determine the locations of all replication-initiation sites, and it is therefore unclear how nonspecic binding of pre-replication complexes might determine where and when metazoan replication initiates. In addition, the factors that coordinate replication with other cellular processes (that is, cell cycle progression) require further characterization.
There are indications that additional proteins, which are not members of the pre-replication complex, participate in specic DNAprotein interactions occurring at replication-initiation sites. Of particular interest are proteins that bind replicator sequences. Replicators are dened as DNA sequences that contain genetic information, which allows them to function as replication origins17. To date, only a few replicators have been identied in the metazoan genome. Although replication-initiation sites share common characteristics18, no consensus sequences have been identied among known replicators3,8,10,18,19. Only a small subset of the potential replication origins initiate DNA replication in each cell cycle1,3,10, and the choice of origins that initiate each cell cycle is yet unclear. Proteins that interact with particular replicators may thus play a role in determining where and when replication starts. Examples include the interaction of the c-MYC replicator with a DNA-unwinding element-specic protein that recruits CDC45 (ref. 20), interactions of RecQ1 and RecQ4 helicases with pre-initiation complexes20,21, interactions of the histone H3 lysine 9 methyltransferase-associated ORCA protein with pre-replication complexes22 and the role of histone acetylase HBO1 in the activation of CDT1 (ref. 23). Although no sequence-specic DNA-binding proteins were yet shown to determine replicator-specic initiation in mammalian cells, these studies indicate that distinct proteins might interact with subsets of origins, and recruit the general replication machinery to those sites.
The diverse family of WD40-repeat-containing proteins (DDB1- and CUL4-associated factors (DCAFs)) includes Pleckstrin Homology domain-Interacting Protein (PHIP), also known as DCAF14, which associates with insulin receptor substrate (IRS)-1 and IRS-2 (ref. 24). DCAF14/PHIP has also been shown to stimulate cell proliferation and to inhibit apoptosis25,26, and it can serve as a marker for aggressive metastatic melanoma27. One member of the pre-replication complex, CDT1, is a known substrate for a DCAF, CDT2, which mediates its ubiquitination by Cullin 4 (CUL4) and Damage-specic DNA-Binding protein 1 (DDB1)28 and facilitates its degradation in a p97-dependent pathway29,30.
The human beta-globin locus (HBB) contains two intensely studied replicators residing in the replication-initiation regions (IRs)3135. This IR is used in both erythroid and non-erythroid
cells, but initiates DNA replication during early stages of the S phase in erythroid cells and later during the S phase in non-erythroid cells11,3640. Each of the two replicators (Rep-P and RepI) within HBB IR can initiate DNA replication at both native and ectopic sites31,34. Each replicator contains an AT-rich sequence and an asymmetric purine, pyrimidine (AG) sequence, with both sequences required for replication initiation34,35. The HBB IR was used to drive replication of human articial chromosomes32 and as a model replication origin in evolutionary, biochemical and functional studies21,35,4144. The HBB IR, therefore, provides an excellent system to study replicator-binding proteins as well as an opportunity to study replication timing.
To better understand sequence-specic replication initiation, we used the HBB IR as a model to capture replicator-binding proteins. We were particularly interested in proteins that bind to the essential asymmetric purine:pyrimidine (AG) region of the Rep-P replicator. Here we identify a protein RepID (replication-initiation determinant), also known as PHIP or DCAF14, which binds AG and is required for the initiation of DNA replication from Rep-P. Mutations in the RepID-binding sites disable replication initiation from Rep-P and RepID deciency affects cell growth and reduces the frequency of replication initiation events genome-wide. RepID-binding is not restricted to Rep-P, as this protein binds other replication initiation sites that share a common sequence motif. These observations suggest that RepID may play multiple roles during the DNA replication process, functioning at specic categories of replication origins. As a member of the DCAF family and an IRS-1/2 interacting protein, RepID may also serve as a link between DNA replication and metabolic signalling pathways.
ResultsThe Rep-P asymmetric region binds RepID. Previous studies have shown that the Rep-P replicator at the HBB locus (Fig. 1a) contains a 45-bp asymmetric purine:pyrimidine or an AG-rich (AG) region essential for its replicator and anti-silencer functions35. We reasoned that proteins capable of binding this domain would likely play key roles in replication initiation. We used an electrophoretic mobility shift assay (EMSA) to detect AG-binding activities. When oligonucleotides matching the AG domain were incubated with nuclear protein extracts from K562 cells, two shifted DNAprotein complexes were identied (Fig. 1b), indicating that at least two protein complexes (here termed AG1 and AG2, with the AG1 complex demonstrating the faster mobility) could bind to AG in vitro.
We performed intensive mutagenesis of the 45-bp AG wild-type (WT) oligo to identify the specic protein-binding sites (Supplementary Fig. 1a). We found that nucleotide substitutions G10T and G12T eliminated the AG1 complex, while nucleotide substitutions T28G and G30T disrupted the AG2 complex (Fig. 1b and Supplementary Fig. 1a). The substitutions that disrupted the AG1 complex replaced two guanines and also disrupted a potential G-quadruplex at the asymmetric domain, but other substitutions that eliminated G-quadruplexes (for example, GQM, GQEM variants, Supplementary Fig. 1a) did not affect AG1 binding. Specic competitors, but not mutant competitors, could eliminate the observed mobility shifts in a dosage-dependent manner, suggesting that those mobility shifts represented sequence-specic DNAprotein interactions (Fig. 1c, compare lanes 36 with AG1 competitor with lanes 79 with a nonspecic competitor). AG-binding activities could be detected in non-erythroid cell lines including human T-cell leukaemia (Jurkat) cells45 and human colorectal carcinoma (HCT116) cells (Supplementary Fig. 1b). These observations are in line with previous in vivo footprinting analyses46, which revealed
2 NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748 ARTICLE
a
LCR
2.6 kb
Rep-P
AT AG
AG WT AG1 mut AG2 mut AG mut
AG1 site AG2 site
10 kb
b
Labelled AG2 mut oligos Labelled AG1 mut oligos
Labelled AG WT oligos K562 nuclear extract
AG1 complex
Free oligos
+ +
+
c
Unlabelled AG2 mut oligos
Unlabelled AG mut oligos
Unlabelled AG2 mut oligos
Unlabelled AG mut oligos K562 nuclear extract
AG1 complex
Free oligos
+
+ + + + + +
+ +
+ + +
+ + +
AG2 complex
Figure 1 | Two distinct DNAprotein interactions at replicator sequences. (a) Map of the HBB locus (top row), Rep-P (second row) and sequences of the intact (AG WT) and mutant (AG1, AG2 and AG mut) oligonucleotides used in this study. Only one strand is shown. The unshaded nucleotides indicate changes from the AG WT oligo. (b) EMSA analyses were used to measure interactions between proteins from K562 cells and biotin-labelled oligos of AG WT, mutated AG1 and mutated AG2 with sequences shown in a. Two DNAprotein complexes were detected with AG WToligos, but only one complex was detected for AG1 mutant oligos (lower motilityinteraction at the AG2 site) and AG2 mutant oligos (higher motilityinteraction with the AG1 site). Arrowheads point to specic activities termed AG1 and AG2 and to free oligonucleotides. (c) Specicity of AG1 complex formation. Biotin-labelled double-stranded AG2-mutated oligonucleotides, which contain an intact AG1 site and a mutated AG2 site, interacted with K562 nuclear protein extracts in the presence and absence of specic competing unlabelled oligonucleotides (AG2) and nonspecic competing unlabelled oligonucleotides (AG mut, which could not participate in either AG1 or AG2 complexes). Increasing concentrations of unlabelled AG2, but not AG mut, competed for the AG1 complex. The molecular ratios of specic competitor and probe were 1:1, 1:10 and 1:100, and unspecic competitor and probe were 1:1 and 1:100. The symbol indicates
that the reagent was added to the binding reaction, whereas the symbol indicates that the reagent was not included.
protection at the sites corresponding to G10T and G12T (termed AG1) and T28G and G30T (termed AG2).
Since the AG sequence is essential for replication initiation at Rep-P sites34,35, we set out to nd the protein or proteins that interacted with AG1 and/or AG2 as we reasoned that those proteins likely play an important role in regulating replication initiation at Rep-P. In a separate study46 we identied proteins that bound the AG2 site and demonstrated that their functions were related to gene expression. In the current study, therefore, we have concentrated on the AG1 site-binding proteins.
We performed a modied biotin pull-down assay to enrich for AG1-binding proteins (Supplementary Fig. 2a). Oligonucleotides with the AG1 site mutated were added to the assay as competitors to minimize nonspecic DNA-binding and AG2-binding proteins. The resulting protein samples were sequenced using mass spectrometry (Tandem MS/peptide mapping). Of the proteins identied in the initial screen, we used the CellMiner tool47 to select a group of 10 proteins expressed in the K562
cell line that exhibited a high level of coexpression with replication-associated genes (Supplementary Table 1) based on the expression patterns of those genes across the NCI-60 cancer cell collection (see Methods). We then used antibodies against members of the selected group in EMSA assays to screen for a protein that binds the AG oligonucleotides but not AG oligonucleotides harbouring the AG1 mutation. We have identied one such protein candidate RepID, also known as the DCAF14, member of the Ddb1- and Cul4-associated factor family, and as PHIP, a Pleckstrin Homology domain-Interacting Protein.
To test the specicity of RepID antibodies, we have depleted RepID from 2,451 13T melanoma cells48, which are known to overexpress RepID27. Antibodies directed against RepID recognized a single 206-kDa protein on an immunoblot of total proteins from 2,451 13T melanoma cells, but not in cells with doxycycline-mediated induction of a short hairpin RNA (shRNA) directed against RepID (Supplementary Fig. 2b). As shown in
NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 3
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748
Supplementary Fig. 2c, RepID depletion resulted in sharply decreased binding. We have further observed that antibodies directed against RepID were able to supershift the proteinDNA
complex with AG-containing oligonucleotides in EMSA assays (Fig. 2a) but that the complex could not be supershifted using control antibodies, including prebleed (IgG) and antibodies
a b
ORC2 antibody ReplD antibody K562 nuclear extract
+ +
+
+ +
+
Chromosome
Length
Coordinates
RefSeq
Replication:NS
ReplD ChIP
Supershift
5,246 kb 9.6 kb
5,255 kb
AG1 complex
Free oligos
c
d
Cell cycle progression
Enrichment of ReplD-bound
chromatin
8
6
4
2
0
Rep-P WT Rep-P AG1 Rep-P AG
10
8
6
4
2
bG61.3 AG hCollagen hLB2NOri
strand DNAEnrichment of ReplD-bound
chromatin
*
*
*
*
*
*
AG
bG59.8
bG61.3
LacZ
* *
* **
bG59.8 bG61.3 AG
AG
LacZ
*
**
**
**
Amp
Hyg Rep-P LacZ-Zeocin ORF
FRT FRT
0 G1 ES MS LS G2/MCell cycle stages
e f
ReplD WT
ReplD KO
ReplD Rescue
Relative abundance of nascent
strand DNA
20 18 16 14 12 10
8 6 4 2 0
**
Relative abundance of nascent
50
LacZ
bG59.8
bG61.3
FLAG
***
206
36
206
ReplD
GAPDH
45 40 35 30 25 20 15 10
5 0
***
**
**
***
***
***
ReplD KO
********
ReplD rescue
Rep-P WT Rep-P AG1 Rep-P AG
Cell lines
AG
bG59.8 bG61.3
ReplD WT
Primers/probe
Figure 2 | RepID interacts with replication-initiation sites. (a) An antibody against RepID supershifted a DNAprotein complex. EMSAs in K562 nuclear protein extracts are shown in the presence and absence of the indicated antibodies. (The additional band above the AG1 complex is not specic.) (b) ChIP-Seq analysis of RepID binding the HBB locus. Top row, a chromosome map. Below the map, First row, nascent strands reads obtained from K562 cells, second row, RepID ChIP-Seq reads obtained from K562 cells aligned to the indicated region (see Methods for details). (c) ChIP analysis of RepID binding in Simian CV-1 cells harbouring Rep-P variants inserted by site-specic recombination into a constant site. Rep-P WT, unmutated Rep-P; Rep-P AG1, Rep-P carrying the AG1 mutation; Rep-P DAG, Rep-P with the entire AG domain deleted. The LacZ primer/probe served as a negative control. All data were normalized versus amplication by the LacZ primer/probe. FRT, the fLP recombinase target (FRT) site; Amp, ampicillin; Hyg, hygromycin; LacZ-Zeocin ORF, the
LacZ-Zeocin open reading frame. Statistical signicance (Po0.05) was calculated (t-test) versus Rep-P WT. (d) ChIP of RepID binding in K562 cells at different phases of the cell cycle. Primers and probe: bG59.8, bG61.3 and AG from the human Rep-P, hCollagen (human collagen VI), hLB2Nori (a non-initiating sequence near the LMNB2). Statistical signicance (**Po0.01 or *Po0.05) was calculated versus G1. (e) The abundance of HBB sequences in nascent DNA strands from CV-1 cells harbouring Rep-P variants as described in the legend to c. Data were normalized to sequences amplied by the LacZ primer/probe.
Statistical signicance (****Po0.001 or ***Po0.005) was calculated versus Rep-P WT. (f) Depletion of RepID prevented replication initiation at beta-globin origin, but initiation was restored by re-introducing RepID. The abundance of sequences from the HBB locus was measured in nascent DNA strands isolated from U2OS cells harbouring RepID siRNA or expressing of pCMV-RepID-3 FLAG. Statistical signicance (***Po0.005 or **Po0.01) was calculated as
indicated. RepID expression levels were detected using indicated antibodies as shown. Each chart in cf shows results from a representative experiment (n 3).
4 NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748 ARTICLE
directed against pre-replication complex component ORC2 (Supplementary Fig. 2d and Fig. 2a). Using puried RepID fragments expressed in E. coli, we identied the AG interaction domain within amino acids 9231,126 on the RepID sequence (Supplementary Fig. 2e). These observations suggested that RepID was an AG1 site-specic binding protein and was required to form the AG1 complex in vitro.
To investigate whether RepID could bind the asymmetric region (corresponding to the AG oligonucleotide) in vivo, we used chromatin immunoprecipitation (ChIP) followed by sequencing (ChIP-Seq) to detect the binding of RepID at Rep-P (Fig. 2b in K562 and Supplementary Fig. 2f in U2OS cells; original blots are shown in Supplementary Fig. 7). In all examples of ChIP-Seq alignments (for example, Fig. 2b), the top track shows an ideogram of a portion of the relevant chromosome. The region of interest is marked with a circled box. The chromosomal coordinates of the analysed regions are shown beneath the ideogram. The RefSeq alignment of the region of interest is shown below the coordinates. The top experimental track (right below the RefSeq alignment) represents nascent-strand proles (Replication: NS). RepID ChIP patterns (RepID ChIP) are shown below the replication proles. As shown in Fig. 2b, replication-initiation patterns aligned with RepID binding at the HBB locus (human chromosome 11). ChIP assays employing real-time PCR conrmed that RepID-bound chromatin was enriched at bG61.3 (a sequence at the 30 end of Rep-P), the Rep-P AG asymmetric region and the HBB locus control region (LCR; Supplementary Fig. 3a; see Supplementary Table 2 for a list of primers and Supplementary Table 3 for a list of cell lines used in the study). LCR is required for both transcription and initiation of DNA replication at the HBB locus35,38,46,49 and is known to interact directly with Rep-P44,46,50.
To evaluate the sequence specicity of RepID binding, we introduced the AG1 mutations into Rep-P by site-directed mutagenesis, and then used site-specic recombination to generate CV-1 simian cells and murine erythroleukaemia (MEL) cells that carried either Rep-P WT or Rep-P AG1 mutant transgene cassettes (designated as Rep-PWT and Rep-PAG1) at constant genomic locations31,34. This way, all mutants were analysed at identical sites to neutralize chromosomal position effects. Since integration at the MEL cells exhibited orientation-specic gene silencing35, we selected transgenes in which the Rep-P variants were inserted in the permissive orientation that was not prone to transcriptional inactivation. We then analysed RepID binding to these sequences at the ectopic sites. ChIP analyses with antibodies directed against RepID indicated that RepID bound to the AG region of Rep-P WT, but not to the Rep-P AG1 mutant in either simian (CV-1) cells or murine cells (MEL; Fig. 2c and Supplementary Fig. 3b, respectively).
We then tested whether Rep-PRepID interaction occurred throughout the cell cycle; ChIP experiments were performed. Asynchronous cells were fractionated by centrifugal elutriation and the cell cycle phases (G1, Early S, Middle S, Late S and G2/M) of the fractions were determined by propidium iodide staining followed by uorescence-activated cell sorting (FACS) analysis (top portions of Fig. 2d and Supplementary Fig. 3c). In K562 cells, which replicate the beta-globin locus in early S phase, RepID binding was restricted to the G1 and early S phases of the cell cycle (Fig. 2d). RepID binding occurred in the G1 and mid-S phases in lymphoma cells, which replicate the beta-globin locus later during the S phase (Supplementary Fig. 3c).
Preventing RepIDorigin interactions reduced initiation. We next sought to determine whether sequence mutations that affected RepID binding would also affect replication initiation.
The abundance of nascent strands DNA at the Rep-P region inserted in the CV-1 system was measured by real-time PCR (Fig. 2e). High levels of nascent DNA strands located at Rep-P indicated efcient replication initiation at this site. As a negative control, we used Rep-P DAG, a Rep-P variant that lacked its entire AG domain and was not a functional replicator35. The GQEM mutants (Supplementary Fig. 1) that could not form G-quadruplexes but retained the ability to bind RepID was able to initiate replication46. We found that nascent DNA abundance of Rep-P WT sequences was higher than that of Rep-P AG1 mutants inserted at the same location. As expected, the control Rep-P DAG did not initiate replication. These data suggest that the AG1 complex is essential for efcient replication initiation within Rep-P.
We used CRISPR (clustered regularly interspaced short palindromic repeats) -Cas9 to stably deplete (knockout) RepID in HCT116 cells. We tested the knockout efciency using immunoblotting against RepID antibody (Fig. 2f, inset). Nascent DNA abundance of Rep-P in RepID-depleted cells was signicantly reduced. This reduction was prevented when we transiently overexpressed Flag-tagged RepID in the knockout cells (Fig. 2f). By contrast, RepID deciency did not affect nascent DNA abundance at another origin located within the DBF4 locus (Supplementary Fig. 3d). This result supported the hypothesis that RepID plays a role in facilitating replication initiation at some but not all origins.
Genome-wide colocalization of RepID with initiation sites. We used ChIP-Seq to assess whether a FLAG-tagged species of RepID associated with genome-wide replication-initiation events in U2OS cells. In this analysis, RepID-binding peaks called with a genomic DNA control from the same cells identied 24,222 RepID-binding sites. We found that 82.3% of RepID-binding sites localized within 2 kb of replication-initiation sites, whereas 15.4% of replication-initiation sites localized within 2 kb of a RepID-bound region. (The cutoff at 2 kb was based on the size of the isolated nascent strands, which ranged between 0.5 and 1 kb as described in the Methods section.) When this analysis was expanded to consider a 5-kb distance, 86% of RepID-bound regions (20,841 of the total 24,222 sites) colocalized with replication-initiation events and 20.7% of replication-initiation sites colocalized with RepID-bound regions. With the same analysis window, 34.4% of RepID-bound regions colocalized with transcription start sites, a distribution similar to the reported distribution of replication-initiation sites4. ChIP-Seq experiment using an endogenous antibody against RepID in K562 cells (Supplementary Fig. 4a) also suggested that replication-initiation events were highly enriched in RepID-binding sites.
The replication-initiation ratio, reecting the enrichment in replication-initiation events, was the highest observed, thus far, for any DNA-binding protein including transcription factors that were previously shown to associate with the initiation of DNA replication. As shown in the detailed analyses in Supplementary Fig. 4b,c, genome-wide enrichment for replication-initiation events in K562 cells was highest at the locations of RepID binding and diminished with distance from RepID-binding sites. Replication-initiation events also colocalized to a lower extent with c-Jun-binding sites, as reported previously4,51, but did not colocalize with other transcriptional regulators (for example, SIRT6, which exhibited similar colocalization to replication-initiation peaks and simulated randomized peaks).
We next asked whether RepID binding affected initiation activity. To that end, we measured the frequency of replication initiation along with RepID ChIP-Seq in cells with unaltered RepID and in cells that were subjected to CRISPR-mediated RepID depletion. Examples of screenshots aligning nascent-
NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 5
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748
strand proles and ChIP-Seq analyses are shown in Fig. 3a,c,e and in Supplementary Fig. 4dh. In all screenshots, the top, middle and bottom Integrated Genome Viewer (IGV) tracks (right below the RefSeq alignment) represent replication-initiation proles in RepID-procient cells (Replication: WT NS), RepID ChIP-Seq patterns (RepID ChIP) and replication-initiation proles in RepID-depleted cells (Replication: RepID KO NS), respectively. As shown in Fig. 3a (also Supplementary Fig. 4d,g), RepID bound replication origins and replication origins that were not bound by RepID could be found throughout the genome, and could sometimes be located at adjacent regions. To determine the effect of RepID depletion on replication initiation, we next plotted the genome-wide frequency of initiation in RepID KO cells for all 20-kb genomic regions anking replication origins in RepID WT cells (including those that both were and were not bound by RepID). We observed replication-initiation events in both WT and KO cells (Fig. 3b). We then plotted the genome-wide initiation frequency solely in RepID-bound origins (identied by RepID binding in WT cellsfor examples, see Fig. 3c and Supplementary Fig. 4e,h). In this subgroup of origins, the frequency of initiation in KO cells was very low, with no notable colocalization (Fig. 3d). Conversely, replication origins that were not associated with RepID in WT cells (for example, see Fig. 3e and Supplementary Fig. 4f,g) were able to initiate replication in both RepID WT and KO cells (Fig. 3f). These observations suggested that RepID was present at a subset of replication origins and was essential for initiation, specically at those origins.
We next asked whether RepID-binding sites shared common sequence motifs. First, we identied a subset of RepID-bound regions for further analysis. This subset of RepID-bound regions was required to span a region shorter than 400 bp, to be located more than 1 kb away from a neighbouring RepID-bound region and to have a peak score greater than or equal to 150. We have identied 268 RepID-bound regions that t these criteria. A MEME-ChIP analysis for de novo motif (http://meme-suite.org/tools/meme-chip
Web End =http://meme-suite.org/tools/meme-chip) identied several motifs. The top motif that was output by MEME-ChIP was truncated to a 12-bp sequence that matched a segment of the AG region from the Rep-P replicator, which contains the AG1 site (Fig. 3g).
To ask whether this motif was enriched within the RepID-binding sites in the context of the entire genome, we compared the subset of 268 RepID-bound regions to three randomized les, each containing 268 sequences from random genomic loci that were of the same length and GC content as the sequences in the original le. As seen in Supplementary Table 4,71.64% of the RepID-bound regions contained this motif, while 1622% of the randomized sequences contained this motif. As expected, 64.18% of these RepID-bound regions were within 2 kb of a replication origin, whereas only 2025% of the randomized sequences were within 2 kb of a replication origin. Consistent with a signicant but partial overlap of the motif with replication origins, this sequence was found at the HBB, CTCF, JunB origins, but not at the DHFR origin (Fig. 2b and Supplementary Fig. 4gi). Of those sequences in each le that were nearby a replication origin, 70.35% of the RepID-bound regions and 3043% of the regions in the randomized les contained the motif. The high association in the randomized les most likely reected the fact that the randomized origin regions contained a high frequency of GC-rich sequences. RepID regions that were not associated with replication origins exhibited 71.88% association with the motif, and randomized non-origin sequences exhibited between 12 and 16% association. These observations are consistent with a role for the motif in facilitating RepID binding but not in the decision to initiate DNA replication.
Abnormal DNA replication in RepID-decient MEFs. We used single bre analyses (DNA combing) to determine the genome-wide consequences of RepID depletion. When replication patterns in RepID WT and RepID / murine
embryonic broblasts (MEFs)52 were compared, we observed differences in replication fork speed and distance between replication origins (Fig. 4). In RepID WT cells, the median distance between origins was 102.6 kb, and the median fork speed was 1.570 kb min 1 (Fig. 4a,b,d). In RepID / cells, the
median distance between replication origins was 128.3 kb, and the median fork speed was 1.725 kb min 1 (Fig. 4a,c,e). In RepID-decient cells, therefore, there were signicantly fewer replication-initiation events (compared with WT cells). This reduction in initiation events was associated with a compensatory increase in replication fork speed. Although replication origin distances were longer in RepID-decient MEFs, RepID-decient MEFs continued to initiate replication, consistent with the fact that those cells progressed through the S phase and that RepID-decient mice were viable. We hypothesize that the small differences in inter-origin distance we observed reected the fact that other origins would compensate for initiation deciency, as previously reported53. Despite the small differences, notably the differences between replication proles in RepID WT and RepID / MEF cells were statistically signicant (P-values are
0.0218 and 0.0061 for inter-origin distance and fork speed, respectively).
We also assessed the extent of stalled replication, measured as the frequency of asymmetric replication forks, in RepID WT and RepID / cells (Fig. 5). We dened a replication fork as
asymmetric if one side of the fork was 33% longer than the other side (for example, see Fig. 5a). We frequently observed asymmetric replication forks in RepID / cells (31% compared
with 8% in RepID WT cells), suggesting that these cells experienced frequent replication fork stalling (Fig. 5b,c). The percentage of DNA bres exhibiting a replication signal provides another indication of replication activity. In RepID / cells,
a signicantly lower fraction of the DNA bres (7.86%) exhibited a replication signal than bres from WT cells (15.6%, Supplementary Fig. 5a), consistent with a decreased proliferation rate previously reported in RepID-decient cells26. FACS analyses indicated that RepID / cell cultures had fewer cells in the
S phase (and G2/M phase) than WT cells (Supplementary Fig. 5b,c). These observations suggested that, in addition to the observed low initiation rate during the S phase, RepID deciency might also result in a lower frequency of cells entering the S phase. Taken together, our data demonstrated that RepID was required for proper initiation of DNA replication and proper elongation of replication forks. In the absence of RepID, DNA replication initiation and cell cycle progression were both partially impaired.
RepID participates in a distal Rep-P interaction with LCR. We used ChIP-chromosome conformation capture (ChIP-3C) to investigate whether RepID associated with LCR sequences that directly interacted with Rep-P at the HBB locus. In this procedure, crosslinked RepID-bound chromatin was isolated, digested with a restriction enzyme (HindIII) and re-ligated with T4 ligase. With this procedure, if two distant cis-elements are interacting with each other, the ligation will link the two sequences together (Fig. 6a). As shown in Fig. 6b, we were able to amplify a DNA fragment when we amplied crosslinked RepID-bound chromatin with a primer from the HS2 site of the HBB locus control region and another primer from Rep-P. Amplication using the Rep-P anchor with other primer pairs spanning the locus exhibited signicantly lower amplication values, suggesting that the procedure identied an interaction between HS2 and Rep-P
6 NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748 ARTICLE
a
b
RefSeq
Replication: WT NS
RepID ChIP
Replication: RepID KO NS
Colocalized initiation in RepIDKO
40,000
30,000
20,000
10,000
10,000 10,000
5,000 5,000
0
CLIP3 THAP8
0
Distance from centre (all WT replication origin)
c
d
RefSeq
Replication: WT NS
RepID ChIP
Replication: RepID KO NS
Colocalized initiation in RepIDKO
40,000
30,000
20,000
10,000
10,000 10,000
5,000 5,000
0
PSMC6 STYX
0
Distance from centre (ReplD-associated WT origins)
e RefSeq
Replication: WT NS
RepID ChIP
Replication: RepID KO NS
COL5A3
f
Colocalized initiation in RepIDKO
40,000
30,000
20,000
10,000
010,000 10,000
5,000 5,000
0Distance from centre (ReplD non-associated WT origins)
g 2
Bits
1
0
1 2 3 4 5 6 7 8 9 10
11
12
13
14
G
G
C
C
G
N
G
G
N
G
G
G
G
G
Motif
From AG
C
C
C
C
C
C
G
A
A
A
A
C
A
G
G
A
G
G
G
A
G
AG1 site
Figure 3 | Genome-wide colocalization of RepID with replication-initiation sites. (a) A screenshot of a sample genomic region showing replication-initiation proles (NS-Seq) and protein-binding (ChIP-Seq) data. Top track, below the RepSeq genes: nascent-strand patterns from cells with WT RepID (Replication: WT NS). Middle track: ChIP-Seq patterns in WT cells (RepID ChIP). Lowest track: nascent-strand patterns from cells depleted of RepID (Replication: RepID KO NS). The shaded region delineates a RepID-binding origin adjacent to an origin that does not bind RepID. An expanded screenshot of the same region is shown in Supplementary Fig. 4d. (b) The distribution of replication-initiation events in RepID KO cells (overall, 58,656 NS-Seq peaks) that colocalized with initiation events in WT cells, plotted as a function of the distance from the centre of WT origins (overall, 78,859 NS-Seq peaks). (c) A screenshot of a sample genomic region showing replication-initiation proles as in a. The shaded region delineates a replication origin that binds RepID in WT cells and does not initiate replication in KO cells. An expanded screenshot of the same region is shown in Supplementary Fig. 4e. (d) The distribution of replication-initiation events in KO cells plotted as a function of the distance from the centre of origins that were bound by RepID in WT cells (14,716 NS-Seq peaks). (e) A screenshot of a sample genomic region showing replication-initiation proles as in a. The shaded region delineates a replication origin that does not bind RepID in WTcells and initiates replication in both WTand KO cells. An expanded screenshot of the same region is shown in Supplementary Fig. 4f. (f) The distribution of replication-initiation events in KO cells along genomic regions anking replication origins that initiated replication but did not bind RepID in WTcells (64,143 peaks). (g) Consensus sequence for RepID binding identied using a subset of RepID-bound regions. Consensus is aligned with a 12-bp motif, which matches the AG1 site. Data showing the abundance of the motif in RepID-bound regions or randomized les are presented in Supplementary Table 4.
NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 7
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748
a
30 min 30 min
IdU CldU
IdU (green) + CldU (red)
ssDNA
RepID WT
IdU (green) + CldU (red) ssDNA
Ori1 Ori2 Ori3
Inter-origin distance
RepID /
Ori1 Ori2 Inter-origin distance
IdU (green) + CldU (red) ssDNA
b
Median: 102.6
RepID WT
d
Median: 1.570
RepID WT
25
20
10
5
0
N = 66
N = 175
Frequency, %
Frequency, %
20
10
5
0
0.3
0.5
0.7
0.9
1.1
1.3
1.5
1.7
1.9
2.1
2.3
2.5
2.7
2.9
3.1
3.3
3.5
3.7
3.9
4.1
4.3
4.5
15
15
25
50
75
100
125
150
175
200
225
250
275
300
Origin distance, kb
Origin distance, kb
Fork speed (kb min1)
Fork speed (kb min1)
c
Median: 128.3
e
20
10
5
0
0.3
Median: 1.725
N = 90
N = 381
25
20
10
5
0
15
RepID /
RepID /
15
Frequency, %
Frequency, %
25
50
75
100
125
150
175
200
225
250
275
300
0.5
0.7
0.9
1.1
1.3
1.5
1.7
1.9
2.1
2.3
2.5
2.7
2.9
3.1
3.3
3.5
3.7
3.9
4.1
4.3
4.5
RepID WT versus RepID / Mann
Whitney test: P = 0.0218
RepID WT versus RepID / Mann
Whitney test: P = 0.0061
Figure 4 | Depletion of RepID decreases the frequency of replication-initiation events. (a) Cells were sequentially labelled with IdU followed by CldU. Top panel, a typical eld with replication signals (IdU detected in green and CldU detected in red). Second panel, the same eld with all bres labelled with an antibody detecting single-strand DNA (ssDNA; grey). Third and fourth panels, an example of CldU-IdU (third) ssDNA (fourth) bre tracks from RepID WT MEFs. Fifth and sixth panels, an example of CldU-IdU (fth) ssDNA (sixth) bre tracks from RepID / MEFs. Illustrations of replication fork patterns
are shown below the ssDNA track. The lengths of bres label associated with ldU and CIdU incorporation and inter-origin distances were measured (see Methods), and rates of replication fork progression were calculated based on these values. Ori, origin; ssDNA, DNA detected by anti-single strand antibody. (b,c) Measurements of the distribution of distances between replication origins in DNA bres from WT MEFs and RepID / MEFs.
(d,e) Measurements of the distribution of replication fork progression rates for WT and RepID / MEFs. The differences between measurements
from bres obtained from wild-type and RepID-decient MEFs were signicant at Po0.05 (P 0.0218 for inter-origin distance and P 0.0061 for
replication fork speed as calculated using the MannWhitney test). Normality test by KolmogorovSmirnov test showed that the distributions of data for be are not normal (Po0.01).
8 NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748 ARTICLE
a
Symmetric fork
Ori
Left fork Right fork
Ori
Left fork Right fork
Ori
Left fork Right fork
Asymmetric fork
Ori
Left fork Right fork
b c
70
60
50
40
30
20
10
0 0 10 20 30 40 50 60 70
Left fork (kb)
RepID WT
Right fork (kb)
70
60
50
40
30
20
10
RepID /
Right fork (kb)
Asymmetric forks: 8% N = 50
Asymmetric forks: 31% N = 58
0 0 10 20 30 40 50 60 70
Left fork (kb)
Figure 5 | RepID-decient MEFs exhibit replication fork asymmetry. (a) Examples of DNA bres derived from wild-type and RepID / MEFs that
contain symmetric and asymmetric replication forks. (b,c) Scatter plots of left and right fork lengths in RepID WT (b) and RepID / (c) cells. The
percentages of the asymmetric forks (outside the red lines) and the number of replication forks measured in both cells are presented on the plots, demonstrating that 31% of forks were asymmetric in RepID / MEFs, whereas 8% of forks were asymmetric in RepID WT MEFs.
in RepID-bound chromatin (Fig. 6a). As expected, this interaction was not observed when we used chromatin from RepID-depleted cells (RepID KO). ChIP-3C of RepID-bound chromatin from K562 cells yielded a 139-bp PCR product amplied with Rep-P and HS2 primers (Supplementary Fig. 6a). To test whether the amplied fragment indicated an interaction between Rep-P and HS2, we cloned and sequenced the amplication product. As expected, the amplied fragment contained both HS2 and Rep-P sequences linked at a HindIII site (Supplementary Fig. 6a). Chromatin loops were detected at both HS2 and HS4 within the HBB locus in K562 cells (Supplementary Fig. 6b). Interactions between HS4 and Rep-P were not observed in RepID-associated chromatin (compare Fig. 6b with Supplementary Fig. 6b)54. Since immunoprecipitation with an antibody directed against RepID detected the HS2Rep-P interaction in both K562 and U2OS cells, our results suggest that the HBB locus in both erythroid and non-erythroid cells exhibits a RepID-associated interaction between LCR and Rep-P at HS2.
DiscussionIn this study, we report that the RepID protein binds distinct mammalian replication origins and is required for sequence-specic initiation of DNA replication at these origins. RepID was rst identied by its interaction with a single replicator sequence, which is essential for replication initiation at the HBB locus. RepID exhibits genome-wide enrichment at replication-initiation sites. Cells depleted of RepID exhibited diminished initiation frequency, slower elongation of replication forks and frequent replication fork-stalling events. Together, these observations support the notion that distinct DNAprotein interactions at specic groups of replicators dictate replication initiation, and that RepID is a mediator of such interactions.
Although mammalian replicators can initiate DNA replication at ectopic sites, there is an ongoing debate whether replication-initiation sites are determined by specic DNA sequences or
solely by chromatin structures1,3,10. Chromatin modications, for example, dimethylation of histone H3 lysine K79 (H3K79Me2), can associate with a distinct fraction of replication-initiation sites in the human genome, and can mark replicated chromatin during the S phase to prevent re-replication and preserve genomic stability55. The lack of binding specicity by components of pre-replication complexes suggests that, in addition to the requirement for those complexes to initiate DNA replication, distinct replicator-interacting proteins might be required to initiate DNA replication in a sequence-specic manner at particular loci3,56. Our results suggest that RepID is one such protein that interacts with a subgroup of origin sequences.
The locations and timing of replication-initiation events are often affected by interactions with cis-acting distal genomic elements1,3,19,54,57 including promoters, enhancers and insulators. These interactions can form chromatin loops to determine where and when replication initiates and likely coordinate replication with transcription. At the human HBB locus, interaction of Rep-P with the LCR is essential for initiation of DNA replication49. As summarized in Fig. 7, the RepID-binding site at the HBB locus is adjacent to the binding site of the LCR-associated remodelling complex (LARC), which regulates transcription46. Despite the close proximity of transcription and replication complexes at the AG element, RepID is a sequence-specic replicator-interacting protein that does not act as a transcription factor at the HBB locus, as the antisilencing activity of the AG element is not affected by mutations that prevent RepID binding46. Consistent with a separation of function between the two AG-binding proteins, AG2 mutations, which prevent LARC binding46, do not prevent initiation of DNA replication, and prevention of RepID binding does not affect transcriptional activity. These observations rule out a possible causal relationship between RepID and LARC binding. The involvement of RepID in the interaction between Rep-P and LCR suggests a possible mechanism for dictating replication-initiation events. RepID may thus play a role in coordinating transcription
NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 9
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748
a
LCR 5 4 3 2 1
HS4 HS3 HS2
IR
AG45
HS2
AG
(1) Crosslink and digest
HS2
AG
b
700
600
500
Relative crosslinking frequency
WT ReplD
(2) ChIP and ligation
RepID
KO ReplD
HS2
400
300
200
100
(3) PCR
AG
= HindIII digest site
Y
= Anti-RepID antibody
= PCR primer
RepID
0
HS4 HS2
LCR
HBE1 HBG2 HBG1 HBD Rep-P
P13 P12 P11
P10 P9
P8 P7 P6 Probe Anchor
Figure 6 | RepID is present in a complex between LCR and Rep-P in early replicating HBB loci. (a) Schematic illustration of the beta-globin locus and the outline of ChIP-3C procedure. Cells were lysed and digested with HindIII. Crosslinked chromatin fragments were immunoprecipitated with anti-RepID antibody and ligated. Crosslinks were reversed and DNA was isolated and amplied by PCR. The primers and TaqMan probe used for the ChIP-3C analysis are listed in the Methods section. The red arrows indicate the location of AG and HS2 on the beta-globin locus, which are more than 50 kb away from each other. (b) ChIP-3C analysis of long-distance RepID-associated chromatin interactions at the HBB locus in U2OS WT and RepID knockout cells. The 3C primers correspond to sequences near the downstream sticky ends of the 3C fragments. Primer/probe combinations were designated p6 to p13, and their locations are indicated as half arrows. The x axis shows the positions of the restriction fragments on the genomic scale (vertical bars). The diagram represents the extent of PCR amplication of each primer/probe pair. Values represent the average of triplicate samples (bars represent s.e.s). Data were obtained after subtraction of no ligation controls and were normalized to the AG site loading control. Primer efciencies were normalized using a single bacterial articial chromosome (BAC) clone covering the genome segment under study.
and replication at the beta-globin locus and similar RepID-binding replicators through its tandem bromodomains.
We have identied a common GC-rich motif for RepID-binding, which is evident in the AG region of the Rep-P replicator and resembles motifs previously identied for replication-initiation sites in mice and Drosophila7. Although this motif is GC-rich, the ability to bind RepID is distinct from the ability to form G-quadruplex structures18 or other motifs that were associated with many replication origins7,51. Indeed, RepID-bound regions contained this motif whether they contributed to replication initiation or not. These studies support the notion that RepID is a sequence-specic DNA-binding protein that contributes to replication initiation at a subset of replication origins; however, other factors such as histone modications and differentiation state might also affect the frequency of initiation.
RepID is a member of the DCAF family of proteins that interact with Cullin-RING-based E3 ubiquitin ligases28. WD40 repeat-containing proteins, including RepID (DCAF14) BRWD3 and BRWD1 (DCAF19), have diverse functions in eukaryotes that are often associated with cell cycle progression. Examples include LRWD1/ORCA facilitating ORC-binding to chromatin22, RFWD3 associating with replication protein A following damage repair58, DCAF2 (CDT2), mediating DNA-damage-induced p97-mediated Cdt1 proteolysis29 and DCAF1 (VprBP) facilitating cellular proliferation59. Similar to RepID, WD40-repeat-containing DCAF proteins may, therefore, act as adaptors for specic protein chromatin interactions.
The replication deciencies observed in the absence of RepID are consistent with the requirement of RepID for pancreatic b-cell proliferation25 and strongly suggest a role for RepID in the regulation of DNA replication. We observed that both the initiation and elongation steps of DNA synthesis seem affected in RepID-decient MEFs, which initiate DNA replication at a low frequency and exhibit frequent replication fork stalling. These data are consistent with previous observations, suggesting that replication-initiation frequencies and elongation rates are interconnected. Recent studies demonstrate that a low frequency of active replication origins (genome-wide and in fragile sites) might trigger genomic instability, and, conversely, that the pace of replication could dictate the frequency of initiation events53,6062. Although it is formally possible that RepID exerts unrelated effects on initiation and elongation, the colocalization we observed between RepID and replication origins and the requirement for RepID for initiation at a group of replication origins lend support to the hypothesis that RepID deciency reduces the frequency of initiation events and this low frequency, in turn, affects genomic stability.
Although RepID deciency affected replication-initiation rates, notably not all replication-initiation events were disabled, suggesting that many replication-initiation events did not require RepID. Although our observations could not formally rule out an indirect role of RepID in replication (for example, by affecting the activity or facilitating transcription of replication factors that regulate replication in a subgroup of RepID-binding sites),
10 NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748 ARTICLE
LCR
Rep-P
AT
AG
AG
Rep-ID (DCAF14)
LCR-IP (LARC)
(Regulating replication)
(Regulating transcription)
Figure 7 | Spatial organization of DNAprotein interactions within Rep-P. The asymmetric region within the Rep-P replication of the HBB locus is involved in two distinct DNAprotein interactions. An interaction with the LARC complex facilitates transcription and maintains an open chromatin conformation in erythroid cells46, whereas the interaction of adjacent sequences with RepID facilitates the initiation of DNA replication from Rep-P. Top diagram, a schematic of the HBB locus, illustrating the location of the Rep-P replicator; middle, a schematic of the Rep-P replicator, illustrating the location of the AG sequence (sequence shown in Fig. 1a); lower diagram, two adjacent proteinDNA interactions within AG.
the data are consistent with the hypothesis that RepID facilitates initiation at RepID-binding origins. It is likely, therefore, that metazoan replication origins can be divided into different categories, each associated with a specic modier protein that determines origin usage according to cell type and developmental stage. RepID may thus be the rst member of a series of proteins, which we propose to name RepIDs that interact with particular subsets of replication origins to determine replicator activity. Such proteins might facilitate interactions between the cell cycle-regulatory network and chromatin to determine where and when DNA replication starts and how replication coordinates with transcription and other chromatin transactions.
Methods
Cell lines and culture conditions. We grew all cells in DMEM (Invitrogen,Cat. no. 10564-011) supplemented with 10% heat-inactivated fetal calf serum in a 37 C/5% CO2 incubator. We added 1% penicillinstreptomycin (Invitrogen,
Cat. no. 15140-163) and 1% Fungizone (Invitrogen, Cat. no. 15290-018) to the culture media as needed. All cells tested negative for mycoplasma. The selection drug, Zeocin (Invitrogen, Cat. No. R250-01), was added to CV-1 cells to a nal concentration of 100 mg ml 1 before plasmid transfection. For RepID knockout stable clone selection, we added puromycin (Invitrogen, Cat. No. A11138-03) at a nal concentration of 0.3125 mg ml 1 to HCT116 cells and 1 mg ml 1 to U2OS cells after plasmid transfection. Mouse embryonic broblasts were isolated from
RepID-procient and null mice26. CV-1 and RL4 cells harbouring Rep-P WT or mutant Rep-P were originally obtained from American Type Culture Collection (ATCC) and modied to facilitate site-specic insertions of dened sequences by FLP-mediated recombination31. Melanoma cell line 2451 13T (ref. 48) was a gift from Dr Yardena Samuels (NHGRI). All other cancer cell lines were obtained from ATCC (http://www.atcc.org
Web End =www.atcc.org).
Cell cycle fractionation was performed by centrifugal elutriation of asynchronously growing K562 cells. The purity of the fractions was measured using ow cytometry.
EMSA. The oligonucleotide substrates used in the EMSA assays included the 45-bp asymmetric region (AG WT), the AG1 mutant oligo and the AG2 mutant oligo (Fig. 1). For EMSA analysis, biotin-labelled forward and reverse oligonucleotides were mixed at a nal concentration of 100 pM, and then boiled at 100 C for 1 min. After annealing, the oligonucleotides were incubated with 20 mg nuclear extract in 1 binding buffer (10 mM Tris-HCl, 2.5% glycerol, 0.05% NP-40, 25 mM KCl,
1 mg poly (dI.dC) and 1 mM dithiothreitol (DTT)) for 30 min at room temperature. Reactions were subsequently subjected to electrophoresis using a 6% DNA
retardation gel (Invitrogen Cat. no. EC6365BOX) on ice for 1.5 h and then transferred to a positively charged nylon membrane and ultraviolet-crosslinked. We used the LightShift Chemiluminescent EMSA kit (Pierce, Cat. no. 20148) for biotin-labelled DNA detection. For competition assays, biotin-labelled double-strand AG2 mutant oligonucleotides were mixed at a nal concentration of 100 pM with unlabelled double-strand mutant oligonucleotides at the nal concentrations of 100 pM, 1 nM and 10 nM. For supershift assays, nuclear extracts were incubated with antibodies before the labelled annealing oligonucleotides were added. Antibodies included RepID (A302-055A, Bethyl Laboratories Inc.) and ORC2 (559266, BD Biosciences). Antibodies were validated by the use of specic shRNA to deplete RepID in 2451 13T melanoma cells.
shRNA knockdown. Specic silencing of endogenous RepID was achieved using an inducible shRNA-expressing vector, pSingle-tTS-shRNA (Clontech). shRNAs were inserted into the plasmid using the XhoI and HindIII cloning sites and were delivered into 2451 13T melanoma cells48. Stable clones were selected, and cells conditionally expressing shRNA directed against RepID were induced (or not) with doxycycline for 16 days.
RepID knockout stable cells by the CRISPR-CAS9 system. A 20-bp guide sequence (50-GTGATAAAATGATCCGAGTC-30) targeting DNA within the fourth exon of RepID was selected from a published database of predicted high-specicity protospacer-PAM target sites in the human exome. Two complementary oligos (50-CACCGTGATAAAATGATCCGAGTC-30 and 50-AAACGACTCGGA
TCATTTTATCAC-30) containing the RepID guide sequence and BbsI ligation adapters were synthesized by Eurons MWG Operon (Alabama, USA). Oligo (100 mM, in a total volume of 10 ml) was annealed and ligated into the BbsI-digested pX330 vector. The sequence of the construct was veried by sequencing. For stable selection, HCT116 or U2OS cells were cultured in six-well dishes to 7080% conuence. Cells were co-transfected with 2 mg of RepID single guide RNA (sgRNA) plasmid plus 2 mg of linearized pCR2.1 vector harbouring a puromycin-resistance gene and 10 ml of Lipofectamine 2000 (Life Technologies) per well. Twenty-four hours post transfection, 10% of transfected cells was seeding to 10-cm dishes in a serial dilution. In the next day, cells were cultured in the medium with appropriate concentration of puromycin for selection.
Nascent-strand DNA analysis. Nascent-strand DNA31,34 was extracted from asynchronous cells. This DNA was fractionated on a neutral sucrose gradient. DNA fractions (0.51 kb) were collected and treated with l exonuclease to remove non-RNA-primed genomic DNA fragments. Nascent-strand DNA was quantied with real-time PCR using an ABI 7900 thermocycler (primers and probes used for real-time PCR are listed in Supplementary Table 2). AG1 and AG2 mutations were introduced by site-directed mutagenesis46.
ChIP analysis. ChIP analyses were performed with 1% formaldehyde-xed K562, U2OS RepID 3XFlag, CV-1 and RL4 cells using the Millipore ChIP assay kit (Cat. no. 17295). Antibodies included normal rabbit IgG (sc-2027), anti-Flag (F3165; Sigma-Aldrich) and anti-PHIP (sc-68354; Santa Cruz Biotechnologies). ChIP samples were analysed with real-time PCR using an ABI 7900 thermocycler, with primers/probes listed in Supplementary Table 2. ChIP experiments were performed on at least two biological replicates for each cell line, and PCR amplications were performed in triplicates. All ChIP data were expressed as nanogram-amplied DNA (calculated based on standardized curves of genomic DNA) divided by the number of molecules amplied from the same preparation of Input and mock samples.
Peak-calling of ChIP-seq and NS-seq. For nascent-strand and ChIP-Seq experiments, regions that were signicantly enriched were identied using two peak-calling programmes. For RepID ChIP, the MACS peak-calling programme http://liulab.dfci.harvard.edu/MACS/
Web End =http://liulab.dfci.harvard.edu/MACS/ was used, comparing reads from the ChIP-seq experiment to genomic input reads from the same cell line, with default parameters and a P-value 1e 9. Overall, 24,222 regions enriched for RepID
were identied. For U2OS nascent-strand peak-calling, the SICER programme (http://home.gwu.edu/~wpeng/Software.htm
Web End =http://home.gwu.edu/Bwpeng/Software.htm ) was used. This programme was chosen because of the presence of wide initiation zones. Nascent-strand reads were called by comparison with U2OS genomic reads using a window size of 200 bp, gap size of 600 bp and false discovery rate (FDR) of 0.01. Overall, 92,814 regions enriched for replication initiation were identied. Screenshots of example genomic loci were captured using the IGV genome browser (https://www.broadinstitute.org/igv/
Web End =https://www.broadinstitute.org/ https://www.broadinstitute.org/igv/
Web End =igv/ ). Intersections between two les of enriched regions were identied using a custom script (available on request). The genome-wide colocalization analyses comparing bed les of RepID-bound regions and replication-initiation sites were performed using GenomeInspector with a 20-kb window size.
Identication of a consensus sequence. A subset of RepID-enriched regions from FLAG ChIP-Seq experiments in U2OS RepID-3 FLAG stable cells was
identied with the following criteria: enriched regions were shorter than 400 bp,
NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 11
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748
located at least 1 kb away from the nearest RepID peaks, and have peak scores greater than 150. These criteria identied 268 RepID regions that were submitted to MEME-ChIP, with parameters asking for a sequence between 6 and 22 bp. The rst motif was a 21-bp motif, which was truncated to a 12-bp motif that could be found in the AG sequence of the Rep-P replicator containing the AG1 site.
A custom string search script was used to identify the percentage of sequences containing the motif. A custom script was used to create random les that had the same number of sequences of the same length, but from random genomic loci. (Both scripts are available on request.)
DNA bre analyses. DNA combing analysis of replicating DNA was performed as follows using previously published methods63. RepID WT and RepID / cells
were pulse-labelled with 20 mM IdU (Sigma, Cat. no. I-7125) for 30 min, and then with 50 mM CldU (MP biomedical, Cat. no. 105478) for 30 min. Following the CldU pulse, the cells were embedded in low-melting agarose plugs, and were lysed in the plug with lysis buffer (1 mg ml 1 proteinase K, 50 mM EDTA, 1%
N-lauroyl-sarcosine, 10 mM Tris-Cl, pH 8.0) at 50 C overnight. After digesting the plug with b-agarase (New England Biolabs, Beverly, MA), DNA was combed to silanized surfaces (Microsurfaces Inc.). Replicating DNA was detected with anti-IdU (BD, Cat. no. 347580), anti-CldU (Accuratechmecal, Cat. no. OBT0030) and anti-single stranded DNA (Chemicon, Cat. no. MAB3034) antibodies. Images were captured using the Attovision software and the epiuorescence microscope Pathway (Becton Dickinson). For replication fork rate measurements, only internal replication signals on single-strand DNA (not colocalizing with DNA bre bundles, not located at the end of a strand) were selected for analyses. Samples have included a BAC as a molecular marker, allowing us to assess the uniformity of bre-stretching and convert labelled signal length (in microns) to bre length(in kb). Signals were marked for evaluation by blind measurers (not knowing which samples they were measuring); signal length was measured using the Image J software (open source from National Cancer Institute, NIH) followed by automatic compilation of signal lengths into an Excel worksheet. Replication fork velocities were calculated using elongating fork signals only (initiating forks were eliminated). Fork velocities and origin distances were calculated using a constant of 2 kb mm 1. Experiments were performed at least in duplicate using independent biological isolations of DNA bres for each experimental condition. Statistical analyses were preformed with the Prism 5.0 software (GraphPad Software Inc.) using the non-parametric MannWhitney rank-sum test.
Cell cycle analysis. Cells were pulse-labelled with 20 mM 5-ethynyl uridine (EdU) for 45 min before cell harvest. EdU staining was performed using the Click-iT EdU kit (Invitrogen, C10424) according to the manufacturers protocol. 4,6-Diamidino-2-phenylindole or propidium iodide (PI) were used for DNA staining. BD LSRFortessa cell analyser with the FACSDiva software was used for cell cycle analysis.
Nuclear protein extract preparation. Cells were harvested and incubated in sucrose buffer containing NP-40 (320 mM sucrose, 10 mM Tris-HCl pH 8.0,3 mM CaCl2, 2 mM MgOAc, 0.1 mM EDTA, 0.5% NP-40, 1 mM DTT, 0.5 mM phenylmethylsulphonyl uoride (PMSF), protease inhibitor cocktail (Sigma,Cat. P8340) and phosphatase inhibitor cocktail (Roche, Cat. 04906845001)). Nuclei were harvested by centrifugation, washed and resuspended in low-salt buffer(20 mM HEPES pH 7.9, 20 mM KCl, 0.2 mM EDTA, 25% glycerol (v/v), 0.5 mM DTT, 0.5 mM PMSF, protease inhibitor cocktail and phosphatase inhibitor cocktail). And then an equal volume of high-salt buffer (20 mM HEPES pH 7.9, 800 mM KCl, 0.2 mM EDTA, 25% glycerol (v/v), 1% NP-40, 0.5 mM DTT, 0.5 mM PMSF, protease inhibitor cocktail and phosphatase inhibitor cocktail) was added. The suspension was incubated at 4 C for 45 min with rotation, and then were centrifuged it at 14,000 g for 15 min at 4 C. The supernatant was the nuclear soluble fraction, and the pellet was the insoluble chromatin-bound fraction. Before use, protein concentrations were determined with a Bio-Rad DC protein assay kit (Bio-Rad, Cat. no. 500-0112).
Biotin pull-down assay and mass spectrometry. The forward and reverse strands of biotin-labelled AG WT oligos (2.5 mg each) were mixed and incubated for 10 min at room temperature, and then 15 ml 1 binding buffer (10 mM
Tris-HCl, 2.5% glycerol, 0.05% NP-40, 25 mM KCl, 1 mg poly (dI.dC), 1 mM DTT and proteinase inhibitor cocktail) was added. And then 0.5 ml of the Dynabeads M-280 Streptavidin (Invitrogen, Cat. no. 112.05D), 250 mg of the unlabelled competitor AG1 oligo mixture and 500 mg of K562 nuclear protein extract were added. The Dynabeads were collected by using a magnet after 2 h of incubation at room temperature. After washing the beads with 1 binding buffer for three
times, the proteins were eluted by adding 40 ml of 2 SDS sample buffer and ran
on a 412% Tris-Glycine SDSPAGE gel with coomassie-blue. Specic bands were sent for mass spectrometry analysis at the Advanced Technology Program of the National Cancer Institute at Frederick. Of the 117 proteins identied in the initial screen, proteins identied based on a single peptide, low Mascot scores and known contaminants such as keratin were eliminated, and a top group of 10 proteins was selected for further analysis using the CellMiner tool (http://discover.nci.nih.gov
Web End =http://discover.nci.nih.gov; see below for gene expression analyses).
Gene expression pattern analysis. Gene expression data used in the current study can be accessed at CellMiner, at http://discover.nci.nih.gov
Web End =http://discover.nci.nih.gov 47 or from the GEO data repository (accession numbers GSE22821, GSE5846, GSE5949, GSE5720 and GSE32474). To obtain the transcription prole, the NCI-60 cancer cell lines were obtained from the Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis64. Expression levels for transcripts were determined with data for probes from ve platforms and normalization (http://discover.nci.nih.gov/cellminer/
Web End =http://discover.nci.nih.gov/ http://discover.nci.nih.gov/cellminer/
Web End =cellminer/ ). Affymetrix (Affymetrix Inc., Sunnyvale, CA) Human Genome U95 Set (HG-U95); the Human Genome U133 (HG-U133); the Human Genome U133 Plus2.0 Arrays (HG-U133 Plus 2.0); and the GeneChip Human Exon 1.0 ST array (GH Exon 1.0 ST) were included. Agilent (Agilent Technologies Inc., Santa Clara, CA) Whole Human Genome Oligo Microarray was also included. Composite probe set intensity values were transformed to z-scores through subtraction of their60 cell line means and division by their s.d.s (calculations performed in Java). Cross-correlations of the z-score values were carried out in Java.
ChIP-3C analysis. K562 or U2OS cells (107 per sample) were xed in 1% formaldehyde, quenched with 0.125 M glycine and then lysed with 0.2% Nonidet P-40 on ice for 1 h at 4 C. Nuclei were collected by centrifugation, resuspended in NEB buffer 2 (New England Biolabs) containing 0.3% SDS, treated at 37 C for 1 h and quenched with 2% Triton X-100 at 37 C for 1 h, and then digested with HindIII at 37 C overnight. The digested chromatin was immunoprecipitated with an antibody against RepID (A302-055A; Bethyl Laboratories Inc) or IgG control before proceeding to ligation. Subsequently, the beads were washed and resuspended in 200 ml of ligation buffer, and DNA was ligated at 16 C overnight.
Next, crosslinking was reversed and DNA was extracted by phenol/chloroform and ethanol precipitation. The interaction between HS2 and Rep-P was tested by PCR amplication (30 ng DNA template per reaction). The primers and probes used for PCR are listed in Supplementary Table 2. The 3C-qPCR assay primers were designed using the SnapGene Viewer Software. Real-time quantitative PCR was performed on ABI 7900 thermocycler using Taqman Premix (Invitrogen). Primer efciencies were normalized using a single BAC (RP11622D14) clone covering the entire human -globin domain. 3C-qPCR data were normalized versus the internal primer pair AG (Supplementary Table 2) located in the HBB locus. The qPCR procedure (50 C for 2 min followed by 95 C for 10 min and 40 cycles of 95 C for 15 s, 60 C for 1 min) was performed according to the manufacturers instructions.
Chromosome conformation capture. Approximately 107 cells were xed in 1% formaldehyde, quenched with 0.125 M glycine and then lysed with 0.2% Nonidet P-40 on ice for 1 h at 4 C. Nuclei were collected by centrifugation, resuspended in NEB buffer 2 (New England Biolabs) containing 0.3% SDS, treated at 37 C for 1 h and quenched with 2% Triton X-100 at 37 C for 1 h, and then digested with HindIII at 37 C overnight. On the second day, DNA was ligated at 16 C for 4 h in 7 ml of ligation buffer to minimize the intra-DNA ligation. Crosslinking was reversed and DNA extracted by phenol/chloroform. As a negative control, digested DNA was directly reverse-crosslinked without ligation. For the 3C assay, the HindIII-cut and re-ligated BAC RP11622D14 clone covering the entire human -globin domain was used for the 3C control templates. To correct for differences in digestion and crosslinking efciencies between the different samples, 3C data were normalized towards loading controls (GAPDH primers) and a set of primers derived from the ERCC3 gene65. Interaction among Rep-P origin and the human beta-globin locus was quantied with real-time PCR using an ABI 7900 thermocycler as described above (primers and probes used for real-time PCR are listed in Supplementary Table 2).
Data availability. The data sets for the ChIP-seq and nascent-strand next-generation sequencing are available from the GEO under Accession codes GSE28911.
References
1. Mechali, M. Eukaryotic DNA replication origins: many choices for appropriate answers. Nat. Rev. Mol. Cell Biol. 11, 728738 (2010).
2. Bielinsky, A. K. Replication origins: why do we need so many? Cell Cycle 2,
307309 (2003).
3. Aladjem, M. I. Replication in context: dynamic regulation of DNA replication patterns in metazoans. Nat. Rev. Genet. 8, 588600 (2007).
4. Martin, M. M. et al. Genome-wide depletion of replication initiation events in highly transcribed regions. Genome Res. 21, 18221832 (2011).
5. Gilbert, D. M. et al. Space and time in the nucleus: developmental control of replication timing and chromosome architecture. Cold Spring Harb. Symp. Quant. Biol. 75, 143153 (2010).
6. Lunyak, V. V., Ezrokhi, M., Smith, H. S. & Gerbi, S. A. Developmental changes in the Sciara II/9A initiation zone for DNA replication. Mol. Cell Biol. 22, 84268437 (2002).
7. Cayrou, C. et al. Genome-scale analysis of metazoan replication origins reveals their organization in specic but exible sites dened by conserved features. Genome Res. 21, 14381449 (2011).
12 NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748 ARTICLE
8. DePamphilis, M. L. et al. Regulating the licensing of DNA replication origins in metazoa. Curr. Opin. Cell. Biol. 18, 231239 (2006).
9. Blow, J. J. & Dutta, A. Preventing re-replication of chromosomal DNA. Nat. Rev. Mol. Cell Biol. 6, 476486 (2005).
10. Masai, H., Matsumoto, S., You, Z., Yoshizawa-Sugata, N. & Oda, M. Eukaryotic chromosome DNA replication: where, when, and how? Annu. Rev. Biochem. 79, 89130 (2010).
11. Mendez, J. & Stillman, B. Perpetuating the double helix: molecular machines at eukaryotic DNA replication origins. Bioessays 25, 11581167 (2003).
12. Vashee, S. et al. Sequence-independent DNA binding and replication initiation by the human origin recognition complex. Genes Dev. 17, 18941908 (2003).
13. Remus, D., Beall, E. L. & Botchan, M. R. DNA topology, not DNA sequence, is a critical determinant for Drosophila ORC-DNA binding. EMBO J. 23, 897907 (2004).
14. Eaton, M. L., Galani, K., Kang, S., Bell, S. P. & MacAlpine, D. M. Conserved nucleosome positioning denes replication origins. Genes Dev. 24, 748753 (2010).
15. Lubelsky, Y. et al. Pre-replication complex proteins assemble at regions of low nucleosome occupancy within the Chinese hamster dihydrofolate reductase initiation zone. Nucleic Acids Res. 39, 31413155 (2011).
16. Dellino, G. I. et al. Genome-wide mapping of human DNA-replication origins: Levels of transcription at ORC1 sites regulate origin selection and replication timing. Genome Res. 23, 111 (2013).
17. Jacob, F. & Brenner, S. On the regulation of DNA synthesis in bacteria: the hypothesis of the replicon. C. R. Hebd Seances Acad. Sci. 256, 298300 (1963).
18. Besnard, E. et al. Unraveling cell type-specic and reprogrammable human replication origin signatures associated with G-quadruplex consensus motifs. Nat. Struct. Mol. Biol. 19, 837844 (2012).
19. Schepers, A. & Papior, P. Why are we where we are? Understanding replication origins and initiation sites in eukaryotes using ChIP-approaches. Chromosome Res. 18, 6377 (2010).
20. Chowdhury, A. et al. The DNA unwinding element binding protein DUE-B interacts with Cdc45 in preinitiation complex formation. Mol. Cell Biol. 30, 14951507 (2010).
21. Thangavel, S. et al. Human RECQ1 and RECQ4 helicases play distinct roles in DNA replication initiation. Mol. Cell Biol. 30, 13821396 (2010).
22. Giri, S. et al. The preRC protein ORCA organizes heterochromatin by assembling histone H3 lysine 9 methyltransferases on chromatin. eLife 4, e06496 (2015).
23. Miotto, B. & Struhl, K. JNK1 phosphorylation of Cdt1 inhibits recruitment of HBO1 histone acetylase and blocks replication licensing in response to stress. Mol. Cell 44, 6271 (2011).
24. Farhang-Fallah, J., Yin, X., Trentin, G., Cheng, A. M. & Rozakis-Adcock, M. Cloning and characterization of PHIP, a novel insulin receptor substrate-1 pleckstrin homology domain interacting protein. J. Biol. Chem. 275, 4049240497 (2000).
25. Podcheko, A. et al. Identication of a WD40 repeat-containing isoform of PHIP as a novel regulator of beta-cell growth and survival. Mol. Cell Biol. 27, 64846496 (2007).
26. Li, S. et al. The full-length isoform of the mouse pleckstrin homology domain-interacting protein (PHIP) is required for postnatal growth. FEBS Lett. 584, 41214127 (2010).
27. De Semir, D. et al. Pleckstrin homology domain-interacting protein (PHIP) as a marker and mediator of melanoma metastasis. Proc. Natl Acad. Sci. USA 109, 70677072 (2012).
28. Jin, J., Arias, E. E., Chen, J., Harper, J. W. & Walter, J. C. A family of diverse Cul4-Ddb1-interacting proteins includes Cdt2, which is required for S phase destruction of the replication factor Cdt1. Mol. Cell 23, 709721 (2006).
29. Raman, M., Havens, C. G., Walter, J. C. & Harper, J. W. A genome-wide screen identies p97 as an essential regulator of DNA damage-dependent CDT1 destruction. Mol. Cell 44, 7284 (2011).
30. Franz, A. et al. CDC-48/p97 coordinates CDT-1 degradation with GINS chromatin dissociation to ensure faithful DNA replication. Mol. Cell 44, 8596 (2011).
31. Aladjem, M. I., Rodewald, L. W., Kolman, J. L. & Wahl, G. M. Genetic dissection of a mammalian replicator in the human beta-globin locus. Science 281, 10051009 (1998).
32. Henning, K. A. et al. Human articial chromosomes generated by modication of a yeast articial chromosome containing both human alpha satelliteand single-copy DNA sequences. Proc. Natl Acad. Sci. USA 96, 592597 (1999).
33. Kitsberg, D., Selig, S., Keshet, I. & Cedar, H. Replication structure of the human beta-globin gene domain. Nature 366, 588590 (1993).
34. Wang, L. et al. The human beta-globin replication initiation region consists of two modular independent replicators. Mol. Cell Biol. 24, 33733386 (2004).
35. Fu, H. et al. Preventing gene silencing with human replicators. Nat. Biotechnol. 24, 572576 (2006).
36. Dhar, V., Mager, D., Iqbal, A. & Schildkraut, C. L. The coordinate replication of the human beta-globin gene domain reects its transcriptional activity and nuclease hypersensitivity. Mol. Cell Biol. 8, 49584965 (1988).
37. Driscoll, M. C., Dobkin, C. S. & Alter, B. P. Gamma delta beta-thalassemia due to a de novo mutation deleting the 5 beta-globin gene activation-region hypersensitive sites. Proc. Natl Acad. Sci. USA 86, 74707474 (1989).
38. Forrester, W. C. et al. A deletion of the human beta-globin locus activation region causes a major alteration in chromatin structure and replication across the entire beta-globin locus. Genes Dev. 4, 16371649 (1990).
39. Epner, E., Forrester, W. C. & Groudine, M. Asynchronous DNA replication within the human beta-globin gene locus. Proc. Natl Acad. Sci. USA 85, 80818085 (1988).
40. Simon, I. et al. Developmental regulation of DNA replication timing at the human beta globin locus. EMBO J. 20, 61506157 (2001).
41. Kamath, S. & Leffak, M. Multiple sites of replication initiation in the human beta-globin gene locus. Nucleic Acids Res. 29, 809817 (2001).
42. Djeliova, V., Russev, G. & Anachkova, B. DNase I sensitive site in the core region of the human beta-globin origin of replication. J. Cell Biochem. 87, 279283 (2002).
43. Francino, M. P. & Ochman, H. Strand symmetry around the beta-globin origin of replication in primates. Mol. Biol. Evol. 17, 416422 (2000).
44. Karmakar, S., Mahajan, M. C., Schulz, V., Boyapaty, G. & Weissman, S. M.
A multiprotein complex necessary for both transcription and DNA replication at the beta-globin locus. EMBO J. 29, 32603271 (2010).
45. Schneider, U., Schwenk, H. U. & Bornkamm, G. Characterization of EBV-genome negative null and T cell lines derived from children with acute lymphoblastic leukemia and leukemic transformed non-Hodgkin lymphoma. Int. J. Cancer 19, 621626 (1977).
46. Huang, L. et al. Prevention of transcriptional silencing by a replicator-binding complex consisting of SWI/SNF, MeCP1, and hnRNP C1/C2. Mol. Cell Biol. 31, 34723484 (2011).
47. Shankavaram, U. T. et al. CellMiner: a relational database and query tool for the NCI-60 cancer cell lines. BMC Genomics 10, 277 (2009).
48. Wei, X. et al. Exome sequencing identies GRIN2A as frequently mutated in melanoma. Nat. Genet. 43, 442446 (2011).
49. Aladjem, M. I. et al. Participation of the human beta-globin locus control region in initiation of DNA replication. Science 270, 815819 (1995).
50. Noordermeer, D. & de Laat, W. Joining the loops: beta-globin gene regulation. IUBMB Life 60, 824833 (2008).
51. Cadoret, J. C. et al. Genome-wide studies highlight indirect links between human replication origins and gene regulation. Proc. Natl Acad. Sci. USA 105, 1583715842 (2008).
52. Cayrou, C., Coulombe, P. & Mechali, M. Programming DNA replication origins and chromosome organization. Chromosome Res. 18, 137145 (2010).
53. Seiler, J. A., Conti, C., Syed, A., Aladjem, M. I. & Pommier, Y. The intra-S-phase checkpoint affects both DNA replication initiation and elongation: single-cell and -DNA ber analyses. Mol. Cell Biol. 27, 58065818 (2007).54. Yun, W. J. et al. The hematopoietic regulator TAL1 is required for chromatin looping between the beta-globin LCR and human gamma-globin genes to activate transcription. Nucleic Acids Res. 42, 42834293 (2014).
55. Fu, H. et al. Methylation of histone H3 on lysine 79 associates with a group of replication origins and helps limit DNA replication once per cell cycle. PLoS Genet. 9, e1003542 (2013).
56. DePamphilis, M. L. Replication origins in metazoan chromosomes: fact or ction? Bioessays 21, 516 (1999).
57. Donley, N., Smith, L. & Thayer, M. J. ASAR15, a cis-acting locus that controls chromosome-wide replication timing and stability of human chromosome 15. PLoS Genet. 11, e1004923 (2015).
58. Liu, S. et al. RING nger and WD repeat domain 3 (RFWD3) associates with replication protein A (RPA) and facilitates RPA-mediated DNA damage response. J. Biol. Chem. 286, 2231422322 (2011).
59. McCall, C. M. et al. Human immunodeciency virus type 1 Vpr-binding protein VprBP, a WD40 protein associated with the DDB1-CUL4 E3 ubiquitin ligase, is essential for DNA replication and embryonic development. Mol. Cell Biol. 28, 56215633 (2008).
60. Blow, J. J., Ge, X. Q. & Jackson, D. A. How dormant origins promote complete genome replication. Trends Biochem. Sci. 36, 405414 (2011).
61. Debatisse, M., Le Tallec, B., Letessier, A., Dutrillaux, B. & Brison, O. Common fragile sites: mechanisms of instability revisited. Trends Genet. 28, 2232 (2012).
62. Fu, H. et al. The DNA repair endonuclease Mus81 facilitates fastDNA replication in the absence of exogenous damage. Nat. Commun. 6, 6746 (2015).
63. Shimura, T. et al. Blooms syndrome helicase and Mus81 are required to induce transient double-strand DNA breaks in response to DNA replication stress.J. Mol. Biol. 375, 11521164 (2008).64. Scherf, U. et al. A gene expression database for the molecular pharmacology of cancer. Nat. Genet. 24, 236244 (2000).
NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 13
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11748
65. Markova, E. N., Kantidze, O. L. & Razin, S. V. Transcriptional regulation and spatial organisation of the human AML1/RUNX1 gene. J. Cell. Biochem. 112, 19972005 (2011).
Acknowledgements
We thank Dr John Schimenti for kindly sharing RepID-decient murine embryo broblasts and Ms Vidushani Jaylal for help with electrophoretic mobility assays. We are grateful for Dr James Doroshow for facilitating the bioinformatics pipeline data analysis. We thank Dr Yardena Samuels for sharing the melanoma cell line 2451 13T. We thank Dr Randall Smith for generation of the bed le randomization script and Ms Catherine Redmond for help in motif analyses. We thank Dr Sangmin Jang, Ms Amy Conner and Ms Anna Marks for reading and commenting on the manuscript. The intramural programme of the CCR, National Cancer Institute, National Institutes of Health, funded this work.
Author contributions
Y.Z., L.H. and M.I.A. designed experiments. Y.Z., L.H., O.K.S., H.F., K.U. C.-M.L., M.S. (Rao), W.C.R., C.E.R., Y.Y. and H.H. performed experiments and analysed data. O.K.S., C.E.R., M.R. (Ryan) and R.K. performed NGS sequencing analyses. Y.B. and Q.L. provided MEF cells. Y.Z., L.H. and M.I.A. wrote the manuscript.
Additional information
Accession codes: The data sets for the ChIP-seq and nascent-strand next-generation sequencing are available from the GEO under Accession codes GSE28911.
Supplementary Information accompanies this paper at http://www.nature.com/naturecommunications
Web End =http://www.nature.com/ http://www.nature.com/naturecommunications
Web End =naturecommunications
Competing nancial interests: The authors declare no competing nancial interests.
Reprints and permission information is available online at http://npg.nature.com/reprintsandpermissions/
Web End =http://npg.nature.com/ http://npg.nature.com/reprintsandpermissions/
Web End =reprintsandpermissions/
How to cite this article: Zhang, Y. et al. A replicator-specic binding protein essential for site-specic initiation of DNA replication in mammalian cells. Nat. Commun. 7:11748 doi: 10.1038/ncomms11748 (2016).
This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the articles Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
Web End =http://creativecommons.org/licenses/by/4.0/
14 NATURE COMMUNICATIONS | 7:11748 | DOI: 10.1038/ncomms11748 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright Nature Publishing Group Jun 2016
Abstract
Mammalian chromosome replication starts from distinct sites; however, the principles governing initiation site selection are unclear because proteins essential for DNA replication do not exhibit sequence-specific DNA binding. Here we identify a replication-initiation determinant (RepID) protein that binds a subset of replication-initiation sites. A large fraction of RepID-binding sites share a common G-rich motif and exhibit elevated replication initiation. RepID is required for initiation of DNA replication from RepID-bound replication origins, including the origin at the human beta-globin (HBB) locus. At HBB, RepID is involved in an interaction between the replication origin (Rep-P) and the locus control region. RepID-depleted murine embryonic fibroblasts exhibit abnormal replication fork progression and fewer replication-initiation events. These observations are consistent with a model, suggesting that RepID facilitates replication initiation at a distinct group of human replication origins.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer