ArticlePDF Available

Dynamic loading and redistribution of the MCM2-7 helicase complex through the cell cycle

Authors:

Abstract

Eukaryotic replication origins are defined by the ORC-dependent loading of the Mcm2-7 helicase complex onto chromatin in G1. Paradoxically, there is a vast excess of Mcm2-7 relative to ORC assembled onto chromatin in G1. These excess Mcm2-7 complexes exhibit little co-localization with ORC or replication foci and can function as dormant origins. We dissected the mechanisms regulating the assembly and distribution of the Mcm2-7 complex in the Drosophila genome. We found that in the absence of cyclin E/Cdk2 activity, there was a 10-fold decrease in chromatin-associated Mcm2-7 relative to the levels found at the G1/S transition. The minimal amounts of Mcm2-7 loaded in the absence of cyclin E/Cdk2 activity were strictly localized to ORC binding sites. In contrast, cyclin E/Cdk2 activity was required for maximal loading of Mcm2-7 and a dramatic genome-wide reorganization of the distribution of Mcm2-7 that is shaped by active transcription. Thus, increasing cyclin E/Cdk2 activity over the course of G1 is not only critical for Mcm2-7 loading, but also for the distribution of the Mcm2-7 helicase prior to S-phase entry. © 2015 The Authors.
Article
Dynamic loading and redistribution of the Mcm2-7
helicase complex through the cell cycle
Sara K Powell
, Heather K MacAlpine
, Joseph A Prinz, Yulong Li, Jason A Belsky & David M MacAlpine
*
Abstract
Eukaryotic replication origins are defined by the ORC-dependent
loading of the Mcm2-7helicase complex onto chromatin in G1.
Paradoxically, there is a vast excess of Mcm2-7relative to ORC
assembled onto chromatin in G1. These excess Mcm2-7complexes
exhibit little co-localization with ORC or replication foci and can
function as dormant origins. We dissected the mechanisms regu-
lating the assembly and distribution of the Mcm2-7complex in the
Drosophila genome. We found that in the absence of cyclin E/Cdk2
activity, there was a 10-fold decrease in chromatin-associated
Mcm2-7relative to the levels found at the G1/S transition. The
minimal amounts of Mcm2-7loaded in the absence of cyclin
E/Cdk2activity were strictly localized to ORC binding sites. In
contrast, cyclin E/Cdk2activity was required for maximal loading
of Mcm2-7and a dramatic genome-wide reorganization of the
distribution of Mcm2-7that is shaped by active transcription.
Thus, increasing cyclin E/Cdk2activity over the course of G1is not
only critical for Mcm2-7loading, but also for the distribution of
the Mcm2-7helicase prior to S-phase entry.
Keywords cell cycle; chromatin; DNA replication; Mcm2-7
Subject Categories Cell Cycle; Chromatin, Epigenetics, Genomics &
Functional Genomics; DNA Replication, Repair & Recombination
DOI 10.15252/embj.201488307 |Received 26 February 2014 | Revised 27
November 2014 | Accepted 2December 2014 | Published online 2January 2015
The EMBO Journal (2015)34:531543
Introduction
The duplication of a eukaryotic genome within the confines of
S-phase is a remarkable event. First, the sheer scale of the process
tens to thousands of million base pairs of DNAneeds to be precisely
copied once and only once within just a few hours. In addition, DNA
replication forks must initiate within and progress through diverse
local chromatin environments including accessible euchromatin
and repressive heterochromatin. And finally, the process has to be
dynamic and capable of responding to environmental and develop-
mental cues. Thus, with every cell cycle, thousands of DNA replica-
tion start sites (origins) must be selected and activated in a regulated
and coordinated manner to ensure that the entire genome is faithfully
duplicated (reviewed in Masai et al,2010).
Each potential origin of replication is marked by the origin recog-
nition complex (ORC) (Bell & Stillman, 1992; Rao & Stillman, 1995).
In G1, the Mcm2-7 complex, the replicative helicase, is loaded as a
double hexamer at ORC binding sites in an ORC-, Cdc6-, and Cdt1-
dependent manner to form the pre-replicative complex (pre-RC)
(Evrin et al, 2009; Remus et al, 2009; Gambus et al, 2011). The
assembly of the pre-RC in G1 “licenses” the origin for potential
activation in the subsequent S-phase. As a cell enters S-phase,
cyclin-dependent kinase (CDK) and Dbf4-dependent kinase (DDK)
activate the Mcm2-7 helicase by the recruitment of Cdc45 and the
GINS complex to form the CMG (Cdc45, Mcm2-7, and GINS) holo-
helicase complex (reviewed in Tanaka & Araki, 2013). After initia-
tion of DNA replication, the Mcm2-7 helicase, as part of the CMG
complex, travels with and unwinds the DNA ahead of the replica-
tion fork (Aparicio et al, 1997; Labib, 2000; Pacek & Walter, 2004;
Pacek et al, 2006; Sekedat et al, 2010).
In theory, a double hexamer of Mcm2-7 loaded at each origin
should be sufficient to replicate the genome, with a single hexamer
of Mcm2-7 traveling with each bidirectional DNA replication fork.
However, nuclear Mcm2-7 protein levels are in vast excess relative
to ORC or the number of replication origins (Burkhart et al, 1995;
Lei et al, 1996; Donovan et al, 1997; Mahbubani et al, 1997;
Edwards et al, 2002). In vitro, multiple double hexamers of Mcm2-7
are able to be loaded at ORC binding sites by reiterative rounds of
ATP hydrolysis (Bowers et al, 2004; Evrin et al, 2013). Electron
microscopy revealed that these double hexamers are distributed
throughout the DNA template (Evrin et al, 2009; Remus et al,
2009), suggesting the ability of the Mcm2-7 complex to passively
translocate away from ORC binding sites. Similarly, in vivo immuno-
fluorescence and chromatin association studies have revealed that
Mcm2-7 are broadly distributed throughout the nucleus and exhibit
little co-localization with ORC (Madine et al, 1995; Krude et al,
1996; Romanowski et al, 1996b; Ritzi et al, 1998; Edwards et al,
2002; Harvey & Newport, 2003). Together, these seemingly paradox-
ical observations regarding the location and quantity of Mcm2-7
have been termed the “MCM Paradox” (Takahashi et al, 2005).
Although the mechanisms regulating the loading of multiple
Mcm2-7 complexes and their distribution throughout the genome
are unclear, increasing data suggest that the excess Mcm2-7 are
Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
*Corresponding author. Tel: +1 919 681 6077; E-mail: david.macalpine@duke.edu
These authors contributed equally
ª2015 The Authors The EMBO Journal Vol 34 |No 4|2015 531
Published online: January 2, 2015
important for maintaining genome stability during replicative stress.
Mcm2-7 levels can be depleted by more than 90% with little, if any,
impact on progression through S-phase (Woodward et al, 2006;
Crevel et al, 2007; Ge et al, 2007; Ibarra et al, 2008). However, in
the absence of the full complement of Mcm2-7, there is a marked
reduction in the utilization of dormant origins and an increase in
cell death when cells encounter replicative stress during S-phase
(Crevel & Cotterill, 1991; Woodward et al, 2006; Ge et al, 2007;
Ibarra et al, 2008). Thus, the full complement of Mcm2-7 is critical
for the activation of dormant replication origins and function to
preserve genome integrity during replicative stress.
We set out to examine this “MCM Paradox” from a biochemical
and genome-wide perspective by using chromatin association assays
and genome-wide chromatin immunoprecipitation (ChIP) to quan-
tify the loading and distribution of Mcm2-7 at different points in the
cell cycle of Drosophila Kc cells. Specific questions we set out to
address included: (i) How does Mcm2-7 loading onto chromatin
progress during G1? (ii) Do Mcm2-7 complexes load adjacent to
ORC? (iii) Where are Mcm2-7 distributed throughout the genome?
and (iv) Do other chromosomal features (e.g. transcription units)
impact the distribution of Mcm2-7?
Results
Mcm2-7loading is regulated by cyclin E/Cdk2activity
Mcm2-7 chromatin association is dynamic throughout the cell cycle.
Mcm2-7 are loaded onto chromatin in G1, travel ahead of the repli-
cation fork in S-phase, and are removed from the chromatin as
S-phase progresses (reviewed in Masai et al, 2010). In order to
examine the dynamics of Mcm2-7 chromatin association, we devel-
oped approaches to arrest Drosophila Kc cells at specific points in
the cell cycle when Mcm2-7 would presumably be associated or not
associated with ORC at potential origins of replication. We were
particularly interested in identifying a defined cell cycle point imme-
diately before Mcm2-7 helicase activation and its movement with
the DNA replication fork.
We reasoned that inhibition of cyclin E/Cdk2 kinase activity
would be sufficient to arrest cells in G1 immediately prior to origin
activation. We first depleted cyclin E and Cdk2 separately using
RNAi in Drosophila Kc cells. In Drosophila, cyclin E is the regula-
tory subunit of the cyclin-dependent kinase 2 (Cdk2), both of
which, like their mammalian homologs, function together to drive
cells out of G1 and into S-phase (Dulic et al, 1992; Ohtsubo &
Roberts, 1993; Knoblich et al, 1994). We also overexpressed Dacapo,
a p27 homolog and potent inhibitor of cyclin E/Cdk2 kinase
activity (Lane et al, 1996), from a copper-responsive metallothion-
ein promoter. As controls, cells were arrested in early G1 by RNAi
depletion of Dup/Cdt1, a key replication licensing factor required
for Mcm2-7 loading (Whittaker et al, 2000) and at the G1/S transi-
tion by treatment with 1 mM hydroxyurea (HU). Dup/Cdt1 RNAi,
cyclin E RNAi, Cdk2 RNAi, Dacapo overexpression (+Dacapo), and
HU treatment all resulted in a cell cycle arrest with the majority
(7085%) of cells having a 2C DNA content by flow cytometry
(Fig 1A). In contrast, only 3040% of the cells from an asynchro-
nous population of Drosophila cells exhibit 2C DNA content
(Supplementary Fig S1D).
We next assessed the relative amounts of nuclear chromatin-
associated Mcm2-7 in each of these conditions by chromatin
fractionation (Fig 1B). A polyclonal antibody specific to Drosophila
Orc2 (Austin et al, 1999) and a monoclonal antibody (AS1.1) that
recognizes all six Mcm2-7 subunits (Chen et al, 2007) were used to
assess the chromatin-associated and whole cell extract levels of ORC
and Mcm2-7. As expected, cells lacking the licensing factor, Dup/
Cdt1, exhibited no detectable chromatin-associated Mcm2-7 (Fig 1B,
lane 1). The background levels of Mcm2-7 chromatin association in
the Dup/Cdt1-depleted cells were similar to the levels observed in
G2-arrested cells where the presence of geminin prevents pre-RC
formation (McGarry & Kirschner, 1998) (Supplementary Fig S1B). In
contrast, cells arrested at the G1/S transition by HU treatment had a
robust accumulation of nuclear Mcm2-7 (Fig 1B, lane 5). Interest-
ingly, cyclin E and Cdk2 RNAi cells exhibited an intermediate
phenotype (Fig 1B, lanes 2 and 3) with considerably less nuclear
chromatin-associated Mcm2-7 than HU-arrested cells. We observed
a slight increase in the amount of chromatin-associated Mcm2-7 in
+Dacapo cells (Fig 1B, lane 4) over both cyclin E and Cdk2 RNAi
cells. However, the amount of chromatin-bound Mcm2-7 in +Dacapo
cells was still markedly reduced compared to levels seen in
HU-treated cells. In contrast to the cell cycle fluctuations in chroma-
tin-associated Mcm2-7 levels, ORC remained constant for each
condition surveyed. Importantly, the cell cycle differences observed
in Mcm2-7 chromatin association were not simply due to changes in
protein levels as the levels of ORC and Mcm2-7 remained constant
in the non-nuclear cytoplasmic fraction. The chromatin-associated
Mcm2-7/ORC ratio was calculated for each condition, normalized to
the level in HU to obtain a fold difference, and plotted on a log
10
scale (Fig 1C). A 40-fold difference in chromatin-associated Mcm2-7
was observed between Dup/Cdt1 RNAi and HU and 9-fold to 15-fold
differences in loading were observed between HU and cells with
impaired cyclin E/Cdk2 kinase activity (cyclin E RNAi, Cdk2 RNAi,
and +Dacapo). Together, our results suggest that while minimal
loading of the Mcm2-7 complex can occur in the absence of cyclin
E/Cdk2 activity, the loading of the full complement of Mcm2-7
requires cyclin E/Cdk2 kinase activity.
Maximal loading of Mcm2-7coincides with entry into S-phase
Our results suggest that Mcm2-7 levels continue to increase during
G1 concomitant with increasing Cdk2 kinase activity prior to entry
into S-phase. However, it remained possible that the maximal
Mcm2-7 loading we observed was perhaps a consequence of replica-
tive stress resulting from HU treatment and the activation of
dormant replication origins (Anglana et al, 2003). Thus, we wanted
to assess Mcm2-7 loading without perturbing S-phase or activating
the intra-S-phase checkpoint.
To monitor pre-RC assembly as cells synchronously enter
S-phase from G1, we induced overexpression of Dacapo to block
cyclin E/Cdk2 kinase activity and prevent the full complement of
Mcm2-7 from being assembled on the chromatin in G1. We then
released the cells from the Dacapo arrest by removing the inducer
(Cu
2+
) of Dacapo expression. Entry into the next cell cycle was
prevented by the addition of the mitotic inhibitor, colcemid, to the
medium. As cells progressed into S-phase, we monitored DNA
content, Dacapo protein levels, and the chromatin-associated levels
of Mcm2-7 and ORC (Fig 2). We found that most cells entered
The EMBO Journal Vol 34 |No 4|2015 ª2015 The Authors
The EMBO Journal Transcription shapes the distribution of Mcm2-7Sara K Powell et al
532
Published online: January 2, 2015
S-phase by 3 h and had completed DNA replication by 9 h. Entry
into S-phase at 3 h coincided with a decrease in Dacapo expression
and a marked increase (~9-fold) in Mcm2-7 association with the
chromatin. The increase in Mcm2-7 chromatin association at the
onset of S-phase entry was transient as Mcm2-7 were likely removed
from chromatin with passage of the DNA replication fork (Kuipers
et al, 2011). Together, these results demonstrate that the cyclin E/
Cdk2 kinase-dependent loading of the full complement of Mcm2-7 is
a regulated process that occurs during a normal cell cycle.
We also examined Mcm2-7 nuclear localization by immunofluo-
rescence in an asynchronous cell population (Supplementary Fig
S2). In order to visualize chromatin-associated Mcm2-7 in the
nucleus, it is necessary to remove the cytoplasmic and non-
chromatin-associated Mcm2-7 population (which are the majority)
by permeabilizing and washing the cells prior to fixation (Claycomb
et al, 2002). Unfortunately, the permeabilization prior to fixation
makes it impossible to monitor non-chromatin-associated factors
such as cyclin E or Cdk2 levels. In the absence of a suitable cell
cycle marker for cells in G1 or early S-phase, we instead used mono-
methylation of lysine 20 on histone H4 (H4K20me1) as a marker of
cells in late S-phase or G2/M. H4K20me1 is a well-characterized cell
cycle-regulated chromatin mark that is generated by PR-Set7/Set8 in
very late S-phase and persists on the chromatin up until mitosis
(Abbas et al, 2010; Oda et al, 2010) (Supplementary Fig S2A). Thus,
cells in late S-phase and G2/M will be identifiable by elevated levels
of H4K20me1 and, in contrast, cells in G1 and early to mid-S-phase
will be marked by very low levels of H4K20me1. We found that
H4K20me1 and Mcm2-7 staining cell populations were mutually
exclusive with very little overlap consistent with Mcm2-7 being
removed from chromatin by the end of S-phase (Supplementary Fig
+ Dacapo HU
Cdk2
RNAi
Cyclin E
RNAi
Dup/Cdt1
RNAi
Counts
Nuclear
chromatin
fraction
Mcm2-7
Orc2
Mcm2-7
Orc2
Cytoplasmic
fraction
*
Dup/Cdt1 RNAi
Cyclin E RNAi
Cdk2 RNAi
+Dacapo
HU
A
BC
2C 4C 2C 4C 2C 4C 2C 4C 2C 4C
Dup/Cdt1 RNAi
Cyclin E RNAi
Cdk2 RNAi
+ Dacapo
HU
1
10
100
Normalized Mcm2-7/ORC ratio (log 10)
1 2 3 4 5
Figure 1. Cyclin E/Cdk2activity modulates Mcm2-7loading during G1.
A FACS profiles of DNA content for cells arrested at different points in the cell cycle by Dup/Cdt1RNAi, cyclin E RNAi, Cdk2RNAi, Dacapo overexpression (+Dacapo), and
1mM HU.
B Analysis of Mcm2-7nuclear chromatin association at different points in the cell cycle. Nuclear chromatin fractions and cytoplasmic extracts were assayed for Orc2
and Mcm2-7by Western blot. A non-specific band is indicated by an asterisk (*).
C Quantification of the ratio of chromatin-bound Mcm2-7relative to Orc2(log
10
scale) for a minimum of 11 replicates (mean SD).
Mcm2-7
Orc2
Mcm2-7
Orc2
Dacapo
Hours post-Dacapo release
0 1 3 5 7 9 11 13
Nuclear
chromatin
fraction
Cytoplasmic
fraction
Figure 2. The full complement of Mcm2-7is loaded during an
unperturbed G1/S transition.
Cell cycle analysis by FACS of cells arrested in G1by Dacapo overexpression
followed by release back into the cell cycle for 13 h (top). Western blot analysis of
Mcm2-7and Orc2for the nuclear chromatin and cytoplasmic fractions of
Mcm2-7, Orc2, and Dacapo.
ª2015 The Authors The EMBO Journal Vol 34 |No 4|2015
Sara K Powell et al Transcription shapes the distribution of Mcm2-7The EMBO Journal
533
Published online: January 2, 2015
S2B). The remaining H4K20me1-negative cells displayed a range of
Mcm2-7 staining from barely detectable to a robust signal, presum-
ably representing cells in early G1 (low cyclin E/Cdk2 activity) and
cells in late G1/early S-phase (high cyclin E/Cdk2 activity), respec-
tively (Supplementary Fig S2C). Thus, even in a dividing population
of cells, we can clearly detect multiple stages of Mcm2-7 loading in
G1 and early S-phase cells. These findings also parallel an increase in
chromatin-associated Mcm2-7 from late M phase into and peaking at
the G1/S transition (Kuipers et al,2011;Symeonidouet al, 2013).
The cyclin E/Cdk2kinase-dependent phase of Mcm2-7loading
requires the canonical pre-RC assembly pathway
It is well documented that ORC, Cdc6, and Cdt1 are able to load
multiple Mcm2-7 double hexamers onto DNA templates in vitro
(Bowers et al, 2004; Evrin et al, 2009; Remus et al, 2009). In vivo,
the situation is more complex with ORC, Cdc6, and Cdt1 dynami-
cally associating with chromatin (McNairn et al, 2005; Xouri et al,
2007; Sonneville et al, 2012) and ATP hydrolysis driving both the
loading and release of the Mcm2-7 helicase complex (Frigola et al,
2013). It was evident from our biochemical studies that Dup/Cdt1
was required to load minimal amounts of Mcm2-7 in a cyclin
E-independent manner (Fig 1). However, it remained unclear
whether the full complement of Mcm2-7 loading we observed,
which is stimulated by cyclin E/Cdk2 kinase activity, was dependent
on the canonical pre-RC assembly pathway (e.g. Cdc6 and
Dup/Cdt1).
To determine whether the full complement of Mcm2-7 loading
we observed in HU-arrested cells is dependent on the established
pre-RC assembly pathway, we depleted cells of Cdc6 and Dup/Cdt1
immediately after the cyclin E/Cdk2-independent pre-RC assembly
step (Fig 3A). Specifically, we arrested cells in early G1 by RNAi
depletion of cyclin E for 24 h followed by treatment with either
control (pUC) or Cdc6 and Dup/Cdt1 dsRNA for 24 h. Cyclin E was
subsequently restored by overexpression of RNAi-resistant FLAG-
cyclin E. To assess Mcm2-7 loading at a static point in the cell cycle
immediately after entry into S-phase, the cells were released into
medium containing 1 mM HU. As expected, the majority of cells
from each condition arrested with a 2C DNA content (Fig 3B).
We found that cells depleted of Dup/Cdt1 and Cdc6 were unable
to recruit additional Mcm2-7 when cyclin E expression and progres-
sion through G1 were restored by FLAG-cyclin E overexpression
Kc FLAG-cyclin E-
inducible cells
Cyclin E RNAi
pUC RNAi Dup/Cdt1 &
Cdc6 RNAi
Induce FLAG-cyclin E & 1 mM HU
HU
Cyclin E RNAi
Dup/Cdt1
RNAi pUC RNAi Dup/Cdt1 & Cdc6 RNAi
Counts
2C 4C 2C 4C 2C 4C 2C 4C 2C 4C 2C 4C
+FLAG-E +FLAG-E
Mcm2-7
Orc2
Cyclin E RNAi
pUC RNAi
Dup/Cdt1 RNAi
FLAG-cyclin E
Cdc6 RNAi
1 mM HU
Mcm2-7
Orc2
Dup/Cdt1
Cyclin E
1 2 3 4 5 6
B
AC
-+
++
++
-+
+-
--
-+
--
-+
++
--
-+
--+-
+-
-+
+-
++
Cytoplasmic
fraction
Nuclear
chromatin
fraction
Figure 3. The cyclin E/Cdk2kinase-dependent phase of Mcm2-7loading requires the canonical pre-RC assembly pathway.
A Schematic of the experiment.
B FACS profiles of DNA content for each cell population assayed.
C Western blot analysis for Orc2and Mcm2-7in the nuclear chromatin fraction and Orc2, Mcm2-7, Dup/Cdt1, and cyclin E in the cytoplasmic fraction for each
condition assayed.
The EMBO Journal Vol 34 |No 4|2015 ª2015 The Authors
The EMBO Journal Transcription shapes the distribution of Mcm2-7Sara K Powell et al
534
Published online: January 2, 2015
(Fig 3C, lanes 4 and 5). In contrast, control RNAi (pUC) cells were
able to load additional Mcm2-7 when driven through G1 by FLAG-
cyclin E overexpression (Fig 3C, lane 3). The increase of chromatin-
associated Mcm2-7 was not due to a difference in FLAG-cyclin E
overexpression. These results indicate that Dup/Cdt1 and Cdc6 are
needed to load the full complement of Mcm2-7 onto chromatin
throughout G1 in a regulated manner.
Genome-wide distribution of Mcm2-7is determined by
cyclin E/Cdk2activity and local transcription
Earlier immunofluorescence-based studies noted that Mcm2-7 do
not strictly co-localize with ORC in the nucleus during S-phase
(Madine et al, 1995; Krude et al, 1996; Romanowski et al, 1996b).
The dichotomy in Mcm2-7 chromatin association that we observed
between cells arrested by cyclin E RNAi and cells arrested at
the G1/S transition by HU treatment prompted us to investigate
the genome-wide distribution of Mcm2-7 relative to ORC.
Specifically, we used chromatin immunoprecipitation to address
where Mcm2-7 was localized in relation to ORC in cyclin E- and
HU-arrested cells.
We used genome-wide mapping experiments to localize ORC in
asynchronous cells (MacAlpine et al, 2010; Roy et al, 2010)
(Fig 4A) and Mcm2-7 in cyclin E RNAi (Fig 4B) treated cells. We
identified 5,135 ORC peaks and 3,792 Mcm2-7 peaks throughout
the Drosophila Kc genome. The concordance between ORC and
Mcm2-7 in cyclin E RNAi treated cells was >93%, consistent with
the critical role of ORC in pre-RC assembly (Fig 4C). Not surpris-
ingly, we found that ORC and Mcm2-7 were specifically enriched
at early activating origins of DNA replication mapped by the
incorporation of a nucleotide analog, BrdU, during an HU arrest
(Supplementary Fig S3).
Strikingly, we found a very different pattern of Mcm2-7 localiza-
tion at the G1/S transition during an HU arrest (Fig 5A). In contrast
to the tight co-localization with ORC observed in the cyclin
E-depleted cells, we observed a “binary” pattern of Mcm2-7 localiza-
tion across the genome. Specifically, we observed broad chromo-
somal regions containing Mcm2-7 signal punctuated by the absence
of Mcm2-7 localization. The dramatic change in Mcm2-7 was not
due to a global change in chromatin configuration in the HU-
arrested cells because ORC binding remained indistinguishable
between the asynchronous and HU-arrested cells (Supplementary
Fig S4). Together with our biochemical results, these data suggest
that the full complement of Mcm2-7 we observed at the G1/S transi-
tion has re-distributed from ORC binding sites. We do not believe
that Mcm2-7 are completely coating the DNA, but rather that we are
observing the likelihood of detecting Mcm2-7 signal within specific
genomic features (see below).
The binary distribution of Mcm2-7 across the genome prompted
us to investigate genomic features that may be associated with the
broad regions of high or low Mcm2-7 levels along the chromosome.
The Mcm2-7 localization pattern relative to annotated genomic
features suggested that Mcm2-7 may be displaced by actively tran-
scribed genes (Fig 5A). To quantitatively assess the Mcm2-7 distri-
bution relative to transcription units, we generated histograms of
Mcm2-7 enrichment for transcribed and non-transcribed genes
(Fig 5B) and found a bimodal pattern of Mcm2-7 enrichment.
Specifically, active genes had no or very little Mcm2-7 signal,
whereas inactive or non-transcribed genes exhibited an elevated
Mcm2-7 signal (P<1.02 ×10
257
;t=40.16). We also considered
1627 284
3508
ORC Mcm2-7
C
1.00 2.25 3.50 4.75 6.00
Chromosome 2L (Mb)
-20246
Mcm2-7 CycE RNAi
B
1.00 2.25 3.50 4.75 6.00
Chromosome 2L (Mb)
-20246
ORC
A
Figure 4. Mcm2-7loading is restricted to ORC binding sites in the absence of cyclin E.
A Genome-wide analysis of ORC localization by ChIP-chip. ORC enrichment from asynchronous cells is depicted for a 5-Mb section of chromosome 2L.
B Genome-wide analysis of Mcm2-7localization in early G1by ChIP-chip. Mcm2-7enrichment from cyclin E RNAi-depleted cells is depicted for a 5-Mb section of
chromosome 2L.
C Venn diagram depicting the overlap between ORC and Mcm2-7peaks.
ª2015 The Authors The EMBO Journal Vol 34 |No 4|2015
Sara K Powell et al Transcription shapes the distribution of Mcm2-7The EMBO Journal
535
Published online: January 2, 2015
ED
CB
A
Early
Late
0
3
03
0
05 30 35 0
0
050100
Percent of Meta Gene
Mcm2-7 HU Enrichment
Mcm2-7 Late S-Phase ChIP Signal
Mcm2-7 Late S-Phase ChIP Signal
Figure 5. The Mcm2-7chromatin distribution in G1is dependent on cyclin E/Cdk2activity and the transcription machinery.
A Genome-wide analysis of Mcm2-7localization at the G1/S transition by ChIP-chip. Mcm2-7enrichment from HU-arrested cells is depicted for a 5-Mb section of
chromosome 2L. Inset: transcribed (green) and non-transcribed (red) genes are indicated above with genes on the positive strand on the top and those on the
negative on the bottom.
B Bimodal distribution of Mcm2-7enrichment over transcribed and non-transcribed genes. Histogram showing the distribution of probe scores found within
transcribed (green) and non-transcribed (red) genes.
CMeta-gene analysis of Mcm2-7enrichment for different deciles of gene expression and their aggregated probe intensities.
D Mcm2-7is displaced from chromatin by DNA replication. Genome-wide analysis of Mcm2-7localization in late S-phase (6h post HU release) by ChIP-chip. Mcm2-7
enrichment is depicted for a 10-Mb section of chromosome 2L (filled gray), replication timing profile (black line) and early (yellow) and late (purple) replication timing
domains.
E Box-plots representing late S-phase Mcm2-7ChIP signal found within early (246) or late (167) replication domains.
The EMBO Journal Vol 34 |No 4|2015 ª2015 The Authors
The EMBO Journal Transcription shapes the distribution of Mcm2-7Sara K Powell et al
536
Published online: January 2, 2015
that the bimodal distribution of Mcm2-7 enrichment between active
and inactive genes might be due to the activation of early origins
that are enriched near actively transcribed genes. However, we
found that the bimodal distribution for Mcm2-7 was not dependent
on early origin activity, but instead was a specific feature of anno-
tated transcripts (Supplementary Fig S5).
The enrichment of Mcm2-7 signal at non-transcribed genes was
indistinguishable from intergenic levels (Supplementary Fig S6),
suggesting that active transcription was responsible for displacing
Mcm2-7 from chromatin. We reasoned that if active transcription
was sufficient to displace Mcm2-7 from chromatin, then in the
absence of additional Mcm2-7 loading, we should expect that both
low and high transcription levels should be able to displace Mcm2-7
from chromatin. To address this, we binned the 14,594 genes into
deciles based on their relative gene expression levels and plotted the
Mcm2-7 enrichment relative to a “meta”-gene body (Fig 5C). We
found that Mcm2-7 levels were depleted over the entire transcrip-
tion unit for actively transcribed genes. The seven most expressed
deciles (green and gray lines) exhibited the same degree of Mcm2-7
depletion, suggesting that any amount of transcription is sufficient
to displace Mcm2-7 from the chromatin.
We sought to directly test the requirement for active transcription
in the displacement of the Mcm2-7 complex from gene bodies. As
we only observe differences in Mcm2-7 distribution during an HU
arrest when further pre-RC assembly is blocked, we would need to
specifically alter transcription during the progression from G1 to S,
which would likely interfere with the transcription of key cell
cycle-regulated genes (e.g. E2F-regulated genes). As an alternative
approach, we examined the genome-wide localization of Mcm2-7 in
two different Drosophila cell lines (Kc167 and S2) treated with HU.
Although much of the transcriptome is the same between these cell
lines (Cherbas et al, 2011), we identified approximately 100 genes
that were significantly transcribed in one cell line and off in the
other. As before, we found that the Mcm2-7 complex was excluded
from actively transcribed genes in both cell lines (Supplementary
Fig S7). However, when we specifically examined those genes that
were only transcribed in one cell line, we observed a significant
(MannWhitneyWilcoxon test; P<7×10
5
) decrease in Mcm2-7
occupancy that was specific for the transcribed cell line. We
conclude that transcription is required to displace the Mcm2-7
complexes from gene bodies. Together, these results suggest that
individual Mcm2-7 complexes can be displaced by active transcrip-
tion and that after the G1/S transition, they cannot be re-established
or translocate into these regions.
The Mcm2-7 helicase complex is inherently dynamic throughout
the cell cycle; it is loaded onto chromatin, travels with the replica-
tion fork, and is rapidly removed from the chromatin by passage of
the replication fork (Madine et al, 1995; Krude et al, 1996;
Romanowski et al, 1996a; Kuipers et al, 2011). Our biochemical and
genomic data indicate that the full complement of Mcm2-7 is loaded
onto chromatin in late G1 and redistributes throughout non-tran-
scribed genes and intergenic sequences by the G1/S transition. If this
broad distribution of chromatin-associated Mcm2-7 at the G1/S tran-
sition represents the true biological distribution of the Mcm2-7
complex, then we would expect that these Mcm2-7 complexes would
be displaced by replication during S-phase. To test this hypothesis,
we arrested cells at the G1/S transition by treatment with 1 mM HU
and then released them synchronously into S-phase. We surveyed
the genome-wide distribution of Mcm2-7 near the end of S-phase
(6 h after HU release) (Fig 5D, dark gray). We found that Mcm2-7
had been displaced from early replicating regions of the genome and
only remained at those sequences copied in late S-phase (Fig 5D,
black line and purple segments). To quantify these results, we used
our prior segmentation of the genome into early (yellow) and late
(purple) replicating domains (Lubelsky et al, 2014) and compared it
to the distribution of Mcm2-7 at the end of S-phase. We found a
significant enrichment of Mcm2-7 associated with late replicating
domains relative to early domains (Fig 5E, P<2.37 ×10
68
,
t=18.22). Together, these data argue that the broad distribution of
Mcm2-7 we observe at the G1/S transition represents the full and
functional complement of Mcm2-7 on the chromatin.
Discussion
In order to ensure the fidelity of the DNA replication program and
genome integrity, there are many more Mcm2-7 helicase complexes
loaded onto DNA than potential start sites marked by ORC. We have
examined this “MCM Paradox” using both genomic and biochemical
approaches to understand the mechanisms by which the full
complement of excess Mcm2-7 are loaded and distributed through-
out the genome to preserve genomic integrity. We found that in
early G1, minimal levels of Mcm2-7 were loaded onto chromatin at
ORC binding sites independent of cyclin E/Cdk2 activity (Fig 6).
Concurrent with and dependent on increasing cyclin E/Cdk2 activity
during G1, we observed an increase in Mcm2-7 loading that culmi-
nated in a 10-fold higher level of chromatin-associated Mcm2-7 at
the G1/S transition. Both the cyclin E/Cdk2 activity-independent
Mcm2-7 loading and cyclin E/Cdk2 activity-dependent Mcm2-7
loading were dependent on the canonical pre-RC assembly pathway.
Strikingly, the full complement of Mcm2-7 when assembled onto
chromatin was not restricted to sequences immediately adjacent to
ORC binding sites, but rather distributed throughout the genome
and shaped by active transcription.
Precise regulation of cyclin E/Cdk2kinase activity is critical for
pre-RC assembly and genomic stability
Pre-RC assembly begins in telophase (Dimitrova et al, 1999), contin-
ues through G1, and culminates just prior to entry into S-phase
(Symeonidou et al, 2013). The precipitous drop in CDK activity as
cells exit mitosis likely leads to the limited amounts of Mcm2-7 being
assembled onto chromatin that we detect specifically at ORC binding
sites. As the cells progress into G1, increasing levels of Cdk2 activity
contribute to cyclin E/Cdk2-dependent Mcm2-7 loading. Indeed, a
gradual increase of Cdk2 activity occurs following exit from mitosis
in cycling human cells (Spencer et al, 2013). Conversely, at some
point, increasing CDK activity leads to the inhibition of pre-RC
assembly and ultimately origin activation (reviewed in Masai et al,
2010). In S. cerevisiae, CDK activity targeted toward pre-RC compo-
nents directly impairs pre-RC assembly (Drury et al, 1997; Elsasser
et al, 1999; Nguyen et al, 2001); however, in higher eukaryotes, the
evidence for a direct role is less clear. In human cell culture, phos-
phorylation of Cdt1 by CDK actively promotes its degradation by the
SCF-Skp2 E3 ligase (Liu et al, 2004; Nishitani et al, 2006). However,
this appears to be a minor pathway as the bulk of Cdt1 is degraded in
ª2015 The Authors The EMBO Journal Vol 34 |No 4|2015
Sara K Powell et al Transcription shapes the distribution of Mcm2-7The EMBO Journal
537
Published online: January 2, 2015
S-phase by PCNA-coupled Cul4-Ddb1 destruction (Arias & Walter,
2006; Jin et al, 2006; Senga et al, 2006).
How does cyclin E/Cdk2 activity promote the loading of the full
Mcm2-7 complement? Prior experiments performed on quiescent
mammalian cells re-entering the cell cycle suggested that cyclin E/
Cdk2 kinase activity stabilizes Cdc6 and Cdc7, two factors critical
for pre-RC assembly and initiation, respectively (Mailand & Diffley,
2005; Chuang et al, 2009). In contrast, cycling cells become compe-
tent to assemble the pre-RC immediately after the metaphase to
anaphase transition (Clijsters et al, 2013). Following exit from
anaphase, Cdc6 is degraded by APC/C-Cdh1; however, there is a
short window between late G1 and S where Cdc6 protein may accu-
mulate before being degraded in S-phase by a PIP-box and Crl4-Cdt2
mediated mechanism (Clijsters & Wolthuis, 2014). This second
wave of Cdc6 expression in late G1 is likely dependent on cyclin E/
Cdk2 activity and E2f-regulated gene expression and may serve to
promote loading the full complement of Mcm2-7 on the chromatin.
Alternatively, the Mcm2-7 complexes, which are loaded proximal to
the ORC binding sites in a cyclin E- and Cdk2-independent manner,
may serve to promote additional Mcm2-7 loading upon exposure to
increasing cyclin E/Cdk2 kinase activity prior to S-phase by a direct
Mcm2-7cyclin ECdt1 interaction (Geng et al, 2007). The transition
from a cyclin E/Cdk2 kinase-independent mode to cyclin E/Cdk2
kinase-dependent mode of Mcm2-7 loading would facilitate a feed
forward mechanism to amplify Mcm2-7 loading as G1 progresses.
The precise regulation of cyclin E/Cdk2 kinase activity during G1
is critical for pre-RC assembly and genome stability. Cyclin E/Cdk2
kinase activity is required to load the full Mcm2-7 complement, yet
deregulation of cyclin E/Cdk2 kinase activity results in a shortening
of G1, a lengthening of S-phase, and genomic instability (Ohtsubo &
Roberts, 1993; Resnitzky et al, 1994; Wimmel et al, 1994; Spruck
et al, 1999). The genomic instability and lengthening of S-phase
appear to be due to a defect in pre-RC assembly (Ekholm-Reed et al,
2004). We speculate that deregulation of cyclin E/Cdk2 kinase activ-
ity would result in the cells transitioning from mitosis being immedi-
ately exposed to high levels of cyclin E/Cdk2 kinase activity
instead of the gradual increase that normally occurs over the course
of G1 (Spencer et al, 2013). Thus, insufficient pre-RCs would be
Cyclin E/Cdk2 Activity
transcribed non-transcribed
ORC
2
3
4
56
7
Cdc6
Dup/Cdt1
Mcm2-7
Figure 6. Model of pre-RC assembly and redistribution of Mcm2-7in G1.
Mcm2-7is loaded at ORC binding sites immediately after entry into G1.AsG1progresses, increasing cyclin E/Cdk2activity promotes the loading of additional Mcm2-7
complexes resulting in the full complement of Mcm2-7being loaded by the end of G1. All Mcm2-7loading is dependent on Cdc6and Cdt1. Prior to or coinciding with the entry
into S-phase, the full complement of Mcm2-7redistributes along the chromosomes and is displaced from transcribed genes.
The EMBO Journal Vol 34 |No 4|2015 ª2015 The Authors
The EMBO Journal Transcription shapes the distribution of Mcm2-7Sara K Powell et al
538
Published online: January 2, 2015
assembled on chromatin as the cyclin E-deregulated cells rapidly
enter S-phase and may not properly redistribute throughout the
genome. In vivo, mutations that impact Mcm2-7 levels and pre-RC
assembly have been linked to genome instability and tumorigenesis
(Pruitt et al, 2007; Shima et al, 2007; Chuang et al, 2010).
Genome-wide redistribution of Mcm2-7
Our chromatin immunoprecipitation studies of Mcm2-7 localization
provide the first genome-wide view of the Mcm2-7 distribution in a
higher eukaryote and also reveal a dramatic reorganization of
Mcm2-7 during late G1. In the absence of cyclin E/Cdk2 kinase
activity (early G1), we find that there is near perfect concordance
between ORC and Mcm2-7 peaks. A similar distribution of overlap-
ping ORC and Mcm2-7 peaks is also observed in S. cerevisiae
(Wyrick et al, 2001; Eaton et al, 2010). Not surprisingly, these
genome-wide datasets from S. cerevisiae reinforce the essential role of
ORC in pre-RC assembly and suggest that Mcm2-7 loading is a local
phenomenon restricted to the adjacent sequences. In contrast, in
Drosophila, we see a dramatic change in the distribution of Mcm2-7
that occurs by the G1/S transition. Not only are more Mcm2-7 loaded
onto the chromatin, but they are also broadly enriched along the
entire chromosome and not restricted to ORC binding sites.
The distribution of the full Mcm2-7 complement along the
genome is shaped by active transcription. Mcm2-7 signal is enriched
in both intergenic and non-transcribed sequences, but depleted from
actively transcribed transcripts. The displacement of Mcm2-7 is not
localized to exons, but rather occurs over the entirety of the tran-
script suggesting that local sequence bias (e.g. increased GC content
in exons) is not a contributing factor. We do not envision that the
full complement of Mcm2-7 is coating these intergenic and non-
transcribed regions, but rather represent an increased probability of
finding an Mcm2-7 complex in static regions of the genome relative
to those that are actively undergoing transcription.
We propose that Mcm2-7 residing in transcribed regions are
displaced by the passage of RNA Pol II during transcription. The
displacement of Mcm2-7 during transcription is analogous to the
removal of Mcm2-7 from inactive origins during passage of the
replication fork in S-phase (Madine et al, 1995; Krude et al, 1996;
Romanowski et al, 1996a; Kuipers et al, 2011). The bimodal distri-
bution of Mcm2-7 at intergenic and non-transcribed genes relative
to active genes may only be established after the transition into
S-phase. Prior to S-phase and the cessation of pre-RC assembly,
Mcm2-7 are likely in a cycle of loading, translocation, and subse-
quent eviction in transcribed regions; however, once pre-RC assem-
bly is inhibited they are no longer able to re-occupy transcribed
regions following eviction by active transcription.
The mechanism(s) by which the Mcm2-7 enrichment transitions
from ORC-specific localization to a broad and distributed pattern
along the chromosome is not clear. We envision at least two possi-
bilitiestranslocation along the DNA away from ORC or the loading
of Mcm2-7 at distal sites by chromosomal looping. In vitro reconsti-
tution of Mcm2-7 loading on circular DNA templates revealed
Mcm2-7 double hexamers distributed randomly on the template irre-
spective of the location of the ACS or ORC binding site (Evrin et al,
2009; Remus et al, 2009). Thus, in vitro, Mcm2-7 double hexamers
can be loaded and are free to translocate along the dsDNA template.
However, it is very difficult to imagine how, in vivo, Mcm2-7
complexes could translocate along the DNA given the chromatin
obstacles such as nucleosomes, DNA binding proteins, and active
transcription. Nucleosomes would have to be displaced and re-
assembled throughout the genome requiring ATP-dependent chro-
matin remodeling activities and histone chaperones (Alabert &
Groth, 2012). Alternatively, the loading of Mcm2-7 at sites distal to
ORC may be achieved by chromatin looping mediated by cohesin
complexes that are close to ORC in Drosophila (MacAlpine et al,
2010). In mammalian cells, cohesin has been shown to be required
for looping chromatin at replication factories (Guillou et al, 2010).
Defining an origin of replication
The search for sequence-based replicators, similar to the ARS
element in S. cerevisiae, has been a holy grail for the mammalian
replication field. In vitro, any sequence can be replicated in Xenopus
extracts and plasmid-based assays have also exhibited very promis-
cuous replication (Krysan & Calos, 1991). Recently, the analysis of
mammalian replication intermediates by next-generation sequencing
identified G4-quadruplex sequences as potential replicators (Cayrou
et al, 2011; Besnard et al, 2012; Valton et al, 2014). Despite the
identification of G4 quadruplex structures as origins of replication,
there is little concordance between datasets, indicating that these
degenerate structures, which occur every few kilobases, are not
sufficient for origin specification. Work from the Hamlin group
using two-dimensional gel electrophoresis demonstrated that many
genomic fragments have the potential to harbor inefficient replica-
tion origins (Mesner et al, 2006). We propose that the broad distri-
bution of the full complement of Mcm2-7 we observe contributes to
the apparent promiscuity of metazoan origin selection. In vitro
experiments in Xenopus have shown that ORC is not required for
initiation after pre-RC assembly (Hua & Newport, 1998; Rowles
et al, 1999). Thus, those Mcm2-7 helicases located distally from
ORC would still have the potential to be activated and could likely
function as dormant origins in the presence of replicative stress.
In our prior studies, we have demonstrated that early origins of
replication, which are resistant to HU, are enriched for ORC binding
sites (MacAlpine et al, 2010). These ORC binding sites also serve as
focal points for the cyclin E-independent Mcm2-7 loading. Together,
these data suggest that Mcm2-7 loaded proximal to ORC binding sites
have an increased likelihood of activation during early S-phase. In
S. cerevisiae, Cdc45 specifically associates with early activating origins
in G1 of the cell cycle (Aparicio et al, 1999). Thus, during early G1 in
Drosophila, Cdc45 may specifically associate with the few Mcm2-7
complexes loaded proximal to ORC binding sites, increasing the likeli-
hood of activation of this subset of pre-RCs during entry into S-phase.
Materials and Methods
Drosophila cell culture
Cells were cultured in 150-mm plates at an approximate density of
1×10
6
cells/ml in Schneider’s Insect Cell Medium (Invitrogen)
supplemented with 10% heat-inactivated FBS (Hyclone) and 1%
penicillin/streptomycin/glutamine (Invitrogen). Dacapo was overex-
pressed for 48 h in the presence of 500 lM copper sulfate.
FLAG-tagged cyclin E was overexpressed in the presence of 500 lM
ª2015 The Authors The EMBO Journal Vol 34 |No 4|2015
Sara K Powell et al Transcription shapes the distribution of Mcm2-7The EMBO Journal
539
Published online: January 2, 2015
copper sulfate in the medium. To arrest at G1/S, cells were incu-
bated with 1 mM HU for 24 h (unless otherwise noted). G2-arrested
cells were treated with 3% DMSO and assayed after 48 h.
dsRNA synthesis and RNAi
Primers were designed with a 50T7 sequence (TTAATACGACTC-
ACTATAGGGAGA) to amplify a 500900 bp region in the gene of
interest. The region was amplified using standard PCR followed by
gel extraction (Qiagen Qiaquick gel extraction) and the PCR product
was subsequently used as a template for dsRNA production (Pro-
mega T7 Ribomax express large scale expression). The dsRNA was
purified by phenol/chloroform extraction followed by ethanol
precipitation. Primers used for each target: Dup/Cdt1 dsRNA, F:
50ctatcagtatcaagaacaggcg, R: 50tgctttccaccagactg; cyclin E dsRNA,
F: 50gccatccgtcacataagca, R: 50atcgtggaagcaagcagac; Cdk2 dsRNA, F:
50tgtggccctcaaaaagattc, R: 50gaagtaagcgtgctgcagtg; Cdc6 dsRNA, F:
50gccacagcacctgatcagtccttcgcg, R: 50gcagtttacaagaaactatgcacc; pUC
dsRNA, F: 50agctcactcaaaggcggtaa, R: 50gcctacatacctcgctctgc.
Cells were washed with serum-free Schneider’s insect medium
(Invitrogen), resuspended to 1 ×10
6
cells/ml in serum-free insect
medium, and then replated. dsRNA (15 lg/1 ×10
6
cells/ml) was
added to the medium and incubated for 1 h at 25°C. Medium with
2×serum was added to make the total medium 1×in respect to
serum. Cyclin E, Cdk2 and pUC RNAi cells were assessed 48 h after
RNAi, Dup/Cdt1 and Cdc6 RNAi cells were assessed after 24 h.
FACS preparation and analysis
Cells were harvested by centrifugation, washed with ice-cold 1×
PBS, resuspended in residual 1×PBS, and fixed overnight at 4°C
with ice-cold 100% ethanol. Next, the cells were centrifuged, the
supernatant removed, and washed with 1×PBS1% fetal bovine
serum. The cells were resuspended in a final volume of 800 ll1×
PBS1% fetal bovine serum with 50 mg/ml of propidium iodide
and 0.1 mg/ml RNaseA. The cells were incubated at 37°C for 1 h.
Equal numbers of cells per condition were assayed using a FACS-
Canto machine running BD FACSDiva software and R bioconductor
flowCore package to generate histograms.
Chromatin fractionation
Fractionation of the chromatin was optimized for Drosophila cell
lines from established protocols (Hancock, 1974; Ritzi et al, 1998).
Cells were harvested and then washed with ice-cold 1×PBS twice.
Cells were resuspended in hypotonic buffer [10 mM HEPES-KOH
(pH 7.5), 20 mM KCl, 0.25 mM EDTA, and a Roche protease mini
tab], kept on ice for 4 min then centrifuged for 4 min at 700 gat
4°C. The pellet was resuspended in lysis buffer [10 mM HEPES-KOH
(pH 7.5), 70 mM NaCl, 20 mM KCl, 5 mM MgCl
2
, 2 mM CaCl
2
,
0.5% NP-40, and a Roche protease inhibitor tab] and underlayed
with 1 volume of 30% sucrose. The lysate was centrifuged for
3 min at 3,000 gat 4°C. The supernatant was set aside as the whole
cell lysate sample. The pellet was resuspended in lysis buffer and
centrifuged with a sucrose cushion. The pellet was washed with low
salt buffer [20 mM HEPES/KOH (pH 7.5), 0.5 mM MgCl
2
, 0.3 M
sucrose, KCl, and Roche protease inhibitor tab] and centrifuged at
4°C for 3 min at 3,000 g. The pellet was washed again in low salt
buffer and resuspended in 2×SDS +b-ME loading buffer. To verify
that the proteins associated with the nuclear pellet were soluble,
they were washed in high salt buffer (480 mM HEPES/KOH (pH
7.5), 0.5 mM MgCl
2
, 0.3 M sucrose, KCl, and Roche protease inhibi-
tor tab) (Supplementary Fig S8A). Alternatively, to solubilize the
chromatin-bound fraction, pellets were resuspended in low salt
buffer supplemented with 5 mM CaCl
2
and treated with 45 U of
micrococcal nuclease for 10 min prior to quenching with 5 mM
EGTA (Supplementary Fig S8B).
Immunofluorescence
For double labeling of Mcm2-7 and H4K20me1, cells were treated
with 0.5% Triton X-100 in PBS for 1 min, washed, fixed with 4%
paraformaldehyde in PBS for 10 min, and blocked with 3% BSA in
PBS-T (0.1% Triton in PBS). Cells were stained with monoclonal
Mcm2-7 antibody (AS1.1, 1:100) and anti-H4K20me1 antibody
(ab9051, 1:1,000), followed by secondary detection with Alexa Fluor
568 goat anti-mouse and Alexa Fluor 633 goat anti-rabbit antibodies
(1:500). Images were acquired with Zeiss Axio Imager wide field
fluorescence microscope, and the median intensities of individual
cells (n=277) from each channel were measured by ImageJ. The
plot of H4K20me1 signal versus Mcm2-7 signal was generated in R.
Western blot analysis
Mcm2-7 monoclonal mouse antibody (AS1.1) was used at 1:100
(Chen et al, 2007), Orc2 rabbit polyclonal antibody was used at
1:3,000 (Austin et al, 1999), Dacapo (NP-1) used at 1:1,000 (Iowa
Hybridoma Bank), Dup/Cdt1 guinea pig raised antibody was used at
1:5,000, and cyclin E antibody raised in rabbits was used at 1:500
(d-300 Santa Cruz sc-33748). Secondary antibodies: goat anti-mouse
IRDye 800CW IgG (LiCor), goat anti-rabbit Alexa Fluor 680 IgG (Invi-
trogen), and goat anti-guinea pig IRDye 800 (Rockland Immuno-
chemicals), were all used at 1:10,000. Westerns were visualized and
quantified using LiCor infrared technology and gray scaled in ImageJ.
ChIP-chip sample preparation
Samples were prepared as described previously (MacAlpine et al,
2010). Mcm2-7 (AS1.1) antibody was used at a 1/25 dilution. All
experiments were performed in duplicate.
ChIP-Chip and ChIP-Seq analysis
Within-replicate probe intensities were determined and between-
slide intensities normalized via the R (R Development Core Team,
2008) package limma (Smyth, 2004). Replicated probe intensities
were determined via MA2C (Song et al, 2007). Subsequently, all
analyses were done in R using the combined replicate probe intensi-
ties. Gene bodies were obtained from (Graveley et al, 2011). Mcm2-7
ChIP-seq and corresponding input samples were performed on
HU-arrested Kc and S2 cells. log2 RPKM ratios of Mcm ChIP-seq to
input sample were calculated over all Drosophila gene models from
the 5.12 annotation release, excluding any genes overlapping a Kc
or S2 early origin. Genes were then subsequently subsetted into
untranscribed (<1 RPKM) and transcribed (>4 RPKM) groups
based on expression data from Cherbas et al (2011). log2 ratios of
The EMBO Journal Vol 34 |No 4|2015 ª2015 The Authors
The EMBO Journal Transcription shapes the distribution of Mcm2-7Sara K Powell et al
540
Published online: January 2, 2015
Mcm2-7 ChIP-seq to input samples were then compared between
the cell lines for four gene classes: untranscribed genes in both Kc
and S2 (4,593 genes), transcribed genes in both Kc and S2 (4,804
genes), untranscribed in Kc and transcribed in S2 (91 genes), and
transcribed in Kc and untranscribed in S2 (128 genes). The Mann
WhitneyWilcoxon Test was used to evaluate differences in the log2
ratio distributions between Kc and S2.
FLAG-cyclin E stable transfection
SLIC cloning (Li & Elledge, 2012) was used to introduce a FLAG
fusion tag in frame at either the N-terminus or C-terminus of cyclin
E cDNA (clone LD22682) in the pMK-CTAP plasmid (pMK-CTAP
was a gift from Artavanis laboratory). Briefly, the plasmid was
digested with a single restriction site enzyme, N term =XhoI, C
term =SpeI. The 50ends were treated with T4 DNA polymerase
(NEB) for 45 min at room temperature. Approximately 150 ng of
vector in a 1:1 ratio was annealed to FLAG primers in 1×ligation
mix (Invitrogen) at 37°C for 30 min. The reactions were trans-
formed into max efficiency DH5acells (Invitrogen). Effectene trans-
fection reagent kit (Qiagen) protocol was followed for suspension
cells in 100-mm dish, with the only exception being that 150 ll
Effectene reagent was added. Cells were plated at 1 ×10
6
cells/ml
and incubated at 25°C for 2 days. Hygromycin B (Sigma) was then
added to 0.125 lg/ml.
Data access
All genomic data are publicly available at NCBI GEO and SRA data
repositories with the following accessions numbers: GSE17282,
GSE17283, GSE41349, GSE63915, SRP050273.
Supplementary information for this article is available online:
http://emboj.embopress.org
Acknowledgements
We thank Bob Duronio, Don Fox, and MacAlpine laboratory members for criti-
cal comments on the manuscript. Don Rio for the Dacapo construct, Steve Bell
for Mcm2-7antibody, and the Artavanis laboratory for pMK plasmid. This work
was supported by an American Cancer Society Research Scholar Grant
(120222-RSG-11-048-01-DMC) and the National Institutes of Health
(1R01GM104097-01A1).
Author contributions
SKP and DMM designed the experiments and project. SKP performed the
biochemical experiments, HKM performed the genome-wide experiments, JAP
and JAB executed the bioinformatic analysis, and YL performed the immuno-
fluorescence experiments. SKP, HKM, and DMM wrote the manuscript.
Conflict of interest
The authors declare that they have no conflict of interest.
References
Abbas T, Shibata E, Park J, Jha S, Karnani N, Dutta A (2010) CRL4(Cdt2)
regulates cell proliferation and histone gene expression by targeting PR-
Set7/Set8for degradation. Mol Cell 40:921
Alabert C, Groth A (2012) Chromatin replication and epigenome maintenance.
Nat Rev Mol Cell Biol 13:153 167
Anglana M, Apiou F, Bensimon A, Debatisse M (2003) Dynamics of DNA
replication in mammalian somatic cells: nucleotide pool modulates origin
choice and interorigin spacing. Cell 114:385 394
Aparicio OM, Weinstein DM, Bell SP (1997) Components and dynamics of
DNA replication complexes in S. cerevisiae: redistribution of MCM proteins
and Cdc45p during S phase. Cell 91:59 69
Aparicio OM, Stout AM, Bell SP (1999) Differential assembly of Cdc45p and
DNA polymerases at early and late origins of DNA replication. Proc Natl
Acad Sci USA 96:9130 9135
Arias EE, Walter JC (2006) PCNA functions as a molecular platform to
trigger Cdt1destruction and prevent re-replication. Nat Cell Biol 8:
84 90
Austin RJ, Orr-Weaver TL, Bell SP (1999)Drosophila ORC specifically binds to
ACE3, an origin of DNA replication control element. Genes Dev 13:
2639 2649
Bell SP, Stillman B (1992) ATP-dependent recognition of eukaryotic origins of
DNA replication by a multiprotein complex. Nature 357:128 134
Besnard E, Babled A, Lapasset L, Milhavet O, Parrinello H, Dantec C, Marin JM,
Lemaitre JM (2012) Unraveling cell type-specific and reprogrammable
human replication origin signatures associated with G-quadruplex
consensus motifs. Nat Struct Mol Biol 19:837 844
Bowers JL, Randell JC, Chen S, Bell SP (2004) ATP hydrolysis by ORC catalyzes
reiterative Mcm2-7assembly at a defined origin of replication. Mol Cell
16:967 978
Burkhart R, Schulte D, Hu D, Musahl C, Gohring F, Knippers R (1995)
Interactions of human nuclear proteins P1Mcm3and P1Cdc46.Eur J
Biochem 228:431 438
Cayrou C, Coulombe P, Vigneron A, Stanojcic S, Ganier O, Peiffer I, Rivals E,
Puy A, Laurent-Chabalier S, Desprat R, Mechali M (2011) Genome-scale
analysis of metazoan replication origins reveals their organization in
specific but flexible sites defined by conserved features. Genome Res 21:
1438 1449
Chen S, de Vries MA, Bell SP (2007) Orc6is required for dynamic recruitment
of Cdt1during repeated Mcm2-7loading. Genes Dev 21:2897 2907
Cherbas L, Willingham A, Zhang D, Yang L, Zou Y, Eads BD, Carlson JW,
Landolin JM, Kapranov P, Dumais J, Samsonova A, Choi JH, Roberts J, Davis
CA, Tang H, van Baren MJ, Ghosh S, Dobin A, Bell K, Lin W et al (2011) The
transcriptional diversity of 25 Drosophila cell lines. Genome Res 21:
301 314
Chuang LC, Teixeira LK, Wohlschlegel JA, Henze M, Yates JR, Mendez J, Reed
SI (2009) Phosphorylation of Mcm2by Cdc7promotes pre-replication
complex assembly during cell-cycle re-entry. Mol Cell 35:206 216
Chuang CH, Wallace MD, Abratte C, Southard T, Schimenti JC (2010)
Incremental genetic perturbations to MCM2-7expression and subcellular
distribution reveal exquisite sensitivity of mice to DNA replication stress.
PLoS Genet 6:e1001110
Claycomb JM, MacAlpine DM, Evans JG, Bell SP, Orr-Weaver TL (2002)
Visualization of replication initiation and elongation in Drosophila.J Cell
Biol 159:225 236
Clijsters L, Ogink J, Wolthuis R (2013) The spindle checkpoint, APC/C(Cdc20),
and APC/C(Cdh1) play distinct roles in connecting mitosis to S phase. J Cell
Biol 201:1013 1026
Clijsters L, Wolthuis R (2014) PIP-box-mediated degradation prohibits re-
accumulation of Cdc6during S phase. J Cell Sci 127:1336 1345
Crevel G, Cotterill S (1991) DNA replication in cell-free extracts from
Drosophila melanogaster.EMBO J 10:4361 4369
ª2015 The Authors The EMBO Journal Vol 34 |No 4|2015
Sara K Powell et al Transcription shapes the distribution of Mcm2-7The EMBO Journal
541
Published online: January 2, 2015
Crevel G, Hashimoto R, Vass S, Sherkow J, Yamaguchi M, Heck MM, Cotterill S
(2007) Differential requirements for MCM proteins in DNA replication in
Drosophila S2cells. PLoS ONE 2:e833
Dimitrova DS, Todorov IT, Melendy T, Gilbert DM (1999)Mcm2, but not RPA,
is a component of the mammalian early G1-phase prereplication complex.
J Cell Biol 146:709 722
Donovan S, Harwood J, Drury LS, Diffley JF (1997) Cdc6p-dependent loading of
Mcm proteins onto pre-replicative chromatin in budding yeast. Proc Natl
Acad Sci USA 94:5611 5616
Drury LS, Perkins G, Diffley JF (1997) The Cdc4/34/53 pathway targets Cdc6p
for proteolysis in budding yeast. EMBO J 16:5966 5976
Dulic V, Lees E, Reed SI (1992) Association of human cyclin E with a periodic
G1-S phase protein kinase. Science 257:1958 1961
Eaton ML, Galani K, Kang S, Bell SP, MacAlpine DM (2010) Conserved
nucleosome positioning defines replication origins. Genes Dev 24:748 753
Edwards MC, Tutter AV, Cvetic C, Gilbert CH, Prokhorova TA, Walter JC (2002)
MCM2-7complexes bind chromatin in a distributed pattern surrounding
the origin recognition complex in Xenopus egg extracts. J Biol Chem 277:
33049 33057
Ekholm-Reed S, Spruck CH, Sangfelt O, van Drogen F, Mueller-Holzner E,
Widschwendter M, Zetterberg A, Reed SI (2004) Mutation of hCDC4
leads to cell cycle deregulation of cyclin E in cancer. Cancer Res 64:
795 800
Elsasser S, Chi Y, Yang P, Campbell JL (1999) Phosphorylation controls
timing of Cdc6p destruction: a biochemical analysis. Mol Biol Cell 10:
3263 3277
Evrin C, Clarke P, Zech J, Lurz R, Sun J, Uhle S, Li H, Stillman B, Speck C
(2009) A double-hexameric MCM2-7complex is loaded onto origin DNA
during licensing of eukaryotic DNA replication. Proc Natl Acad Sci USA 106:
20240 20245
Evrin C, Fernandez-Cid A, Zech J, Herrera MC, Riera A, Clarke P, Brill S, Lurz R,
Speck C (2013) In the absence of ATPase activity, pre-RC formation is blocked
prior to MCM2-7hexamer dimerization. Nucleic Acids Res 41:3162 3172
Frigola J, Remus D, Mehanna A, Diffley JF (2013) ATPase-dependent quality
control of DNA replication origin licensing. Nature 495:339 343
Gambus A, Khoudoli GA, Jones RC, Blow JJ (2011) MCM2-7form double hexamers
at licensed origins in Xenopus egg extract. J Biol Chem 286:1185511864
Ge XQ, Jackson DA, Blow JJ (2007) Dormant origins licensed by excess Mcm2-
7are required for human cells to survive replicative stress. Genes Dev 21:
3331 3341
Geng Y, Lee YM, Welcker M, Swanger J, Zagozdzon A, Winer JD, Roberts JM,
Kaldis P, Clurman BE, Sicinski P (2007) Kinase-independent function of
cyclin E. Mol Cell 25:127 139
Graveley BR, Brooks AN, Carlson JW, Duff MO, Landolin JM, Yang L, Artieri CG,
van Baren MJ, Boley N, Booth BW, Brown JB, Cherbas L, Davis CA, Dobin A,
Li R, Lin W, Malone JH, Mattiuzzo NR, Miller D, Sturgill D et al (2011)The
developmental transcriptome of Drosophila melanogaster.Nature 471:
473 479
Guillou E, Ibarra A, Coulon V, Casado-Vela J, Rico D, Casal I, Schwob E, Losada
A, Mendez J (2010) Cohesin organizes chromatin loops at DNA replication
factories. Genes Dev 24:2812 2822
Hancock R (1974) Interphase chromosomal deoxyribonucleoprotein isolated
as a discrete structure from cultured cells. J Mol Biol 86:649 663
Harvey KJ, Newport J (2003) CpG methylation of DNA restricts prereplication
complex assembly in xenopus egg extracts. Mol Cell Biol 23:6769 6779
Hua XH, Newport J (1998) Identification of a preinitiation step in DNA
replication that is independent of origin recognition complex and cdc6,
but dependent on cdk2.J Cell Biol 140:271 281
Ibarra A, Schwob E, Mendez J (2008) Excess MCM proteins protect human
cells from replicative stress by licensing backup origins of replication. Proc
Natl Acad Sci USA 105:8956 8961
Jin J, Arias EE, Chen J, Harper JW, Walter JC (2006) A family of diverse Cul4-
Ddb1-interacting proteins includes Cdt2, which is required for S phase
destruction of the replication factor Cdt1.Mol Cell 23:709 721
Knoblich JA, Sauer K, Jones L, Richardson H, Saint R, Lehner CF (1994) Cyclin
E controls S phase progression and its down-regulation during Drosophila
embryogenesis is required for the arrest of cell proliferation. Cell 77:
107 120
Krude T, Musahl C, Laskey RA, Knippers R (1996)Human replication proteins
hCdc21, hCdc46 and P1Mcm3bind chromatin uniformly before S-phase
and are displaced locally during DNA replication. J Cell Sci 109:309 318
Krysan PJ, Calos MP (1991) Replication initiates at multiple locations on an
autonomously replicating plasmid in human cells. Mol Cell Biol 11:
1464 1472
Kuipers MA, Stasevich TJ, Sasaki T, Wilson KA, Hazelwood KL, McNally JG,
Davidson MW, Gilbert DM (2011) Highly stable loading of Mcm proteins
onto chromatin in living cells requires replication to unload. J Cell Biol
192:29 41
Labib K (2000) Uninterrupted MCM2-7function required for DNA replication
fork progression. Science 288:1643 1647
Lane ME, Sauer K, Wallace K, Jan YN, Lehner CF, Vaessin H (1996) Dacapo, a
cyclin-dependent kinase inhibitor, stops cell proliferation during
Drosophila development. Cell 87:1225 1235
Lei M, Kawasaki Y, Tye BK (1996) Physical interactions among Mcm proteins
and effects of Mcm dosage on DNA replication in Saccharomyces
cerevisiae.Mol Cell Biol 16:5081 5090
Li MZ, Elledge SJ (2012) SLIC: a method for sequence- and ligation-
independent cloning. Methods Mol Biol 852:51 59
Liu E, Li X, Yan F, Zhao Q, Wu X (2004) Cyclin-dependent kinases
phosphorylate human Cdt1and induce its degradation. J Biol Chem 279:
17283 17288
Lubelsky Y, Prinz JA, DeNapoli L, Li Y, Belsky JA, MacAlpine DM (2014) DNA
replication and transcription programs respond to the same chromatin
cues. Genome Res 24:1102 1114
MacAlpine HK, Gordan R, Powell SK, Hartemink AJ, MacAlpine DM (2010)
Drosophila ORC localizes to open chromatin and marks sites of cohesin
complex loading. Genome Res 20:201 211
Madine MA, Khoo CY, Mills AD, Musahl C, Laskey RA (1995) The nuclear
envelope prevents reinitiation of replication by regulating the binding of
MCM3to chromatin in Xenopus egg extracts. Curr Biol 5:1270 1279
Mahbubani HM, Chong JP, Chevalier S, Thommes P, Blow JJ (1997 ) Cell cycle
regulation of the replication licensing system: involvement of a Cdk-
dependent inhibitor. J Cell Biol 136:125 135
Mailand N, Diffley JF (2005) CDKs promote DNA replication origin licensing in
human cells by protecting Cdc6from APC/C-dependent proteolysis. Cell
122:915 926
Masai H, Matsumoto S, You Z, Yoshizawa-Sugata N, Oda M (2010) Eukaryotic
chromosome DNA replication: where, when, and how? Annu Rev Biochem
79:89 130
McGarry TJ, Kirschner MW (1998) Geminin, an inhibitor of DNA replication, is
degraded during mitosis. Cell 93:1043 1053
McNairn AJ, Okuno Y, Misteli T, Gilbert DM (2005) Chinese hamster ORC
subunits dynamically associate with chromatin throughout the cell-cycle.
Exp Cell Res 308:345 356
Mesner LD, Crawford EL, Hamlin JL (2006) Isolating apparently pure libraries
of replication origins from complex genomes. Mol Cell 21:719 726
The EMBO Journal Vol 34 |No 4|2015 ª2015 The Authors
The EMBO Journal Transcription shapes the distribution of Mcm2-7Sara K Powell et al
542
Published online: January 2, 2015
Nguyen VQ, Co C, Li JJ (2001) Cyclin-dependent kinases prevent DNA re-
replication through multiple mechanisms. Nature 411:1068 1073
Nishitani H, Sugimoto N, Roukos V, Nakanishi Y, Saijo M, Obuse C, Tsurimoto
T, Nakayama KI, Nakayama K, Fujita M, Lygerou Z, Nishimoto T (2006)
Two E3ubiquitin ligases, SCF-Skp2and DDB1-Cul4, target human Cdt1for
proteolysis. EMBO J 25:1126 1136
Oda H, Hubner MR, Beck DB, Vermeulen M, Hurwitz J, Spector DL, Reinberg D
(2010) Regulation of the histone H4monomethylase PR-Set7by CRL4
(Cdt2)-mediated PCNA-dependent degradation during DNA damage. Mol
Cell 40:364 376
Ohtsubo M, Roberts JM (1993) Cyclin-dependent regulation of G1in
mammalian fibroblasts. Science 259:1908 1912
Pacek M, Walter JC (2004) A requirement for MCM7and Cdc45 in chromosome
unwinding during eukaryotic DNA replication. EMBO J 23:3667 3676
Pacek M, Tutter AV, Kubota Y, Takisawa H, Walter JC (2006) Localization of
MCM2-7, Cdc45, and GINS to the site of DNA unwinding during
eukaryotic DNA replication. Mol Cell 21:581 587
Pruitt SC, Bailey KJ, Freeland A (2007) Reduced Mcm2expression results in
severe stem/progenitor cell deficiency and cancer. Stem Cells 25:
3121 3132
RDevelopment Core Team (2008)R: A Language and Environment for
Statistical Computing. Vienna, Austria: R Foundation for Statistical
Computing. ISBN 3-900051-07-0, URL http://www.R-project.org
Rao H, Stillman B (1995) The origin recognition complex interacts with a
bipartite DNA binding site within yeast replicators. Proc Natl Acad Sci USA
92:2224 2228
Remus D, Beuron F, Tolun G, Griffith JD, Morris EP, Diffley JF (2009) Concerted
loading of Mcm2-7double hexamers around DNA during DNA replication
origin licensing. Cell 139:719 730
Resnitzky D, Gossen M, Bujard H, Reed SI (1994) Acceleration of the G1/S
phase transition by expression of cyclins D1and E with an inducible
system. Mol Cell Biol 14:1669 1679
Ritzi M, Baack M, Musahl C, Romanowski P, Laskey RA, Knippers R (1998)
Human minichromosome maintenance proteins and human origin
recognition complex 2protein on chromatin. J Biol Chem 273:
24543 24549
Romanowski P, Madine MA, Laskey RA (1996a) XMCM7, a novel member of
the Xenopus MCM family, interacts with XMCM3and colocalizes with it
throughout replication. Proc Natl Acad Sci USA 93:10189 10194
Romanowski P, Madine MA, Rowles A, Blow JJ, Laskey RA (1996b) The Xenopus
origin recognition complex is essential for DNA replication and MCM
binding to chromatin. Curr Biol 6:1416 1425
Rowles A, Tada S, Blow JJ (1999)Changes in association of the Xenopus origin
recognition complex with chromatin on licensing of replication origins.
J Cell Sci 112:2011 2018
Roy S, Ernst J, Kharchenko PV, Kheradpour P, Negre N, Eaton ML, Landolin
JM, Bristow CA, Ma L, Lin MF, Washietl S, Arshinoff BI, Ay F, Meyer PE,
Robine N, Washington NL, Di Stefano L, Berezikov E, Brown CD, Candeias
Ret al (2010) Identification of functional elements and regulatory circuits
by Drosophila modENCODE. Science 330:1787 1797
Sekedat MD, Fenyo D, Rogers RS, Tackett AJ, Aitchison JD, Chait BT (2010 )
GINS motion reveals replication fork progression is remarkably uniform
throughout the yeast genome. Mol Syst Biol 6:353
Senga T, Sivaprasad U, Zhu W, Park JH, Arias EE, Walter JC, Dutta A (2006)
PCNA is a cofactor for Cdt1degradation by CUL4/DDB1-mediated N-
terminal ubiquitination. J Biol Chem 281:6246 6252
Shima N, Alcaraz A, Liachko I, Buske TR, Andrews CA, Munroe RJ, Hartford
SA, Tye BK, Schimenti JC (2007) A viable allele of Mcm4causes
chromosome instability and mammary adenocarcinomas in mice. Nat
Genet 39:93 98
Smyth GK (2004) Linear models and empirical Bayes methods for assessing
differential expression in microarray experiments. Stat Appl Genet Mol Biol
3:3
Song JS, Johnson WE, Zhu X, Zhang X, Li W, Manrai AK, Liu JS, Chen R, Liu XS
(2007) Model-based analysis of two-color arrays (MA2C). Genome Biol 8:
R178
Sonneville R, Querenet M, Craig A, Gartner A, Blow JJ (2012) The dynamics of
replication licensing in live Caenorhabditis elegans embryos. J Cell Biol
196:233 246
Spencer SL, Cappell SD, Tsai FC, Overton KW, Wang CL, Meyer T (2013) The
proliferation-quiescence decision is controlled by a bifurcation in CDK2
activity at mitotic exit. Cell 155:369 383
Spruck CH, Won KA, Reed SI (1999) Deregulated cyclin E induces chromosome
instability. Nature 401:297 300
Symeonidou IE, Kotsantis P, Roukos V, Rapsomaniki MA, Grecco HE, Bastiaens
P, Taraviras S, Lygerou Z (2013) Multi-step loading of human
minichromosome maintenance proteins in live human cells. J Biol Chem
288:35852 35867
Takahashi TS, Wigley DB, Walter JC (2005) Pumps, paradoxes and
ploughshares: mechanism of the MCM2-7DNA helicase. Trends Biochem
Sci 30:437 444
Tanaka S, Araki H (2013) Helicase activation and establishment of replication
forks at chromosomal origins of replication. Cold Spring Harb Perspect Biol
5:a010371
Valton AL, Hassan-Zadeh V, Lema I, Boggetto N, Alberti P, Saintome C, Riou
JF, Prioleau MN (2014)G4motifs affect origin positioning and efficiency
in two vertebrate replicators. EMBO J 33:732 746
Whittaker AJ, Royzman I, Orr-Weaver TL (2000)Drosophila double parked: a
conserved, essential replication protein that colocalizes with the origin
recognition complex and links DNA replication with mitosis and the
down-regulation of S phase transcripts. Genes Dev 14:1765 1776
Wimmel A, Lucibello FC, Sewing A, Adolph S, Muller R (1994) Inducible
acceleration of G1progression through tetracycline-regulated expression
of human cyclin E. Oncogene 9:995 997
Woodward AM, Gohler T, Luciani MG, Oehlmann M, Ge X, Gartner A, Jackson
DA, Blow JJ (2006) Excess Mcm2-7license dormant origins of replication
that can be used under conditions of replicative stress. J Cell Biol 173:
673 683
Wyrick JJ, Aparicio JG, Chen T, Barnett JD, Jennings EG, Young RA, Bell SP,
Aparicio OM (2001) Genome-wide distribution of ORC and MCM proteins
in S. cerevisiae: high-resolution mapping of replication origins. Science 294:
2357 2360
Xouri G, Squire A, Dimaki M, Geverts B, Verveer PJ, Taraviras S, Nishitani H,
Houtsmuller AB, Bastiaens PI, Lygerou Z (2007) Cdt1associates
dynamically with chromatin throughout G1and recruits Geminin onto
chromatin. EMBO J 26:1303 1314
ª2015 The Authors The EMBO Journal Vol 34 |No 4|2015
Sara K Powell et al Transcription shapes the distribution of Mcm2-7The EMBO Journal
543
Published online: January 2, 2015
... However, it is worth noting that despite this, nearly 100% of the origins in yeast are within 1 kb of the ORC-binding sites (Figure 3f). Similar observations have been made in Drosophila, where cyclin E/cdk2 kinase activity promotes the loading of a vast excess of MCM2-7 on chromatin relative to ORC, and the MCM2-7 complexes move away from their loading sites due to the activity of the transcriptional apparatus (Powell et al., 2015). ...
Article
Full-text available
Based on experimentally determined average inter-origin distances of ~100 kb, DNA replication initiates from ~50,000 origins on human chromosomes in each cell cycle. The origins are believed to be specified by binding of factors like the origin recognition complex (ORC) or CTCF or other features like G-quadruplexes. We have performed an integrative analysis of 113 genome-wide human origin profiles (from five different techniques) and five ORC-binding profiles to critically evaluate whether the most reproducible origins are specified by these features. Out of ~7.5 million union origins identified by all datasets, only 0.27% (20,250 shared origins) were reproducibly obtained in at least 20 independent SNS-seq datasets and contained in initiation zones identified by each of three other techniques, suggesting extensive variability in origin usage and identification. Also, 21% of the shared origins overlap with transcriptional promoters, posing a conundrum. Although the shared origins overlap more than union origins with constitutive CTCF-binding sites, G-quadruplex sites, and activating histone marks, these overlaps are comparable or less than that of known transcription start sites, so that these features could be enriched in origins because of the overlap of origins with epigenetically open, promoter-like sequences. Only 6.4% of the 20,250 shared origins were within 1 kb from any of the ~13,000 reproducible ORC-binding sites in human cancer cells, and only 4.5% were within 1 kb of the ~11,000 union MCM2-7-binding sites in contrast to the nearly 100% overlap in the two comparisons in the yeast, Saccharomyces cerevisiae . Thus, in human cancer cell lines, replication origins appear to be specified by highly variable stochastic events dependent on the high epigenetic accessibility around promoters, without extensive overlap between the most reproducible origins and currently known ORC- or MCM-binding sites.
... Previous studies have revealed that forcing origin firing by inhibiting ATR (ATM-and Rad3-related kinase) or CHK1 (Checkpoint kinase 1) leads to a 3-4-fold higher accumulation of CDC45 or TIMELESS in replication domains 44,74 . Our calculations are consistent with numerous studies indicating that despite the variations between cell types and developmental stages, DNA-loaded MCM complexes greatly exceed the number of origins typically employed during the S phase [75][76][77][78] . ...
Article
Full-text available
Accurate and complete replication of genetic information is a fundamental process of every cell division. The replication licensing is the first essential step that lays the foundation for error-free genome duplication. During licensing, minichromosome maintenance protein complexes, the molecular motors of DNA replication, are loaded to genomic sites called replication origins. The correct quantity and functioning of licensed origins are necessary to prevent genome instability associated with severe diseases, including cancer. Here, we delve into recent discoveries that shed light on the novel functions of licensed origins, the pathways necessary for their proper maintenance, and their implications for cancer therapies.
... Nevertheless, the main task of mammalian ORC identical to its yeast counterpart is to load replicative helicases onto DNA during the late M and G1 phases of the cell cycle [29]. The temporal gap between licensing and firing provides a time window during which dynamic events such as collision with transcription complexes can relocate the MCM double hexamer [125][126][127][128], thereby allowing for flexibility in specifying helicase loading sites. As a consequence, this may contribute to the coordination of DNA replication and transcriptional programs during development and cell fate transitions, which is crucial for the cell fate and life cycles of higher eukaryotes. ...
Article
Full-text available
In eukaryotic genomes, hundreds to thousands of potential start sites of DNA replication named origins are dispersed across each of the linear chromosomes. During S-phase, only a subset of origins is selected in a stochastic manner to assemble bidirectional replication forks and initiate DNA synthesis. Despite substantial progress in our understanding of this complex process, a comprehensive 'identity code' that defines origins based on specific nucleotide sequences, DNA structural features, the local chromatin environment, or 3D genome architecture is still missing. In this article, we review the genetic and epigenetic features of replication origins in yeast and metazoan chromosomes and highlight recent insights into how this flexibility in origin usage contributes to nuclear organization, cell growth, differentiation, and genome stability.
... This mechanism contributes to destabilization of early-replicating fragile sites (ERFSs), a category of hard-to-replicate regions associated with small, highly expressed and early-replicating genes [13][14][15][16] . The second is pre-RC displacement and/or disassembly by ongoing transcription machinery, a phenomenon observed in budding yeast 17,18 , Drosophila 19 and human cells [20][21][22] . The resulting reorganization of the initiation program tends to co-orientate replication and transcription, minimizing head-on encounters 20,21,[23][24][25][26][27][28] . ...
Article
Full-text available
Genome integrity requires replication to be completed before chromosome segregation. The DNA-replication checkpoint (DRC) contributes to this coordination by inhibiting CDK1, which delays mitotic onset. Under-replication of common fragile sites (CFSs), however, escapes surveillance, resulting in mitotic chromosome breaks. Here we asked whether loose DRC activation induced by modest stresses commonly used to destabilize CFSs could explain this leakage. We found that tightening DRC activation or CDK1 inhibition stabilizes CFSs in human cells. Repli-Seq and molecular combing analyses showed a burst of replication initiations implemented in mid S-phase across a subset of late-replicating sequences, including CFSs, while the bulk genome was unaffected. CFS rescue and extra-initiations required CDC6 and CDT1 availability in S-phase, implying that CDK1 inhibition permits mistimed origin licensing and firing. In addition to delaying mitotic onset, tight DRC activation therefore supports replication completion of late origin-poor domains at risk of under-replication, two complementary roles preserving genome stability.
... Given that hORC shows little or no sequence specificity in DNA binding, the sites for replication licensing are determined mainly by local chromatin context and transcription status in human cells 23,51,52 . Consistent with previous studies 43,49,53 , the hMCM-DH binding sites are found at an extremely high density within non-transcribed regions, forming broad zones widespread over the human genome. Furthermore, we found that hORC and H2A.Z show distinct patterns in relation to the MCM-DH clusters at early RDs. ...
Article
In eukaryotes, DNA replication initiation requires assembly and activation of the minichromosome maintenance (MCM) 2-7 double hexamer (DH) to melt origin DNA strands. However, the mechanism for this initial melting is unknown. Here, we report a 2.59-Å cryo-electron microscopy structure of the human MCM-DH (hMCM-DH), also known as the pre-replication complex. In this structure, the hMCM-DH with a constricted central channel untwists and stretches the DNA strands such that almost a half turn of the bound duplex DNA is distorted with 1 base pair completely separated, generating an initial open structure (IOS) at the hexamer junction. Disturbing the IOS inhibits DH formation and replication initiation. Mapping of hMCM-DH footprints indicates that IOSs are distributed across the genome in large clusters aligning well with initiation zones designed for stochastic origin firing. This work unravels an intrinsic mechanism that couples DH formation with initial DNA melting to license replication initiation in human cells.
Article
The accurate transmission of genetic information is crucial for the reproduction and evolution of life. To ensure the faithful transmission of genetic information from parents to offspring, organisms have developed a precise DNA replication regulation system. In eukaryotes, during the G1 phase of the cell cycle, the Origin Recognition Complex (ORC) firstly recognizes the specific regions on the chromosome, and then recruits CDC6, CDT1 and MCM complex to form a pre-replication complex (pre-RC), which marks the replication origins. As the cells enter the S phase, the replication origins are selectively activated. The correct selection and activation of DNA replication origins are of utmost importance for the process of DNA replication. Here we describe the processes involved in the selection and activation of replication origins, as well as the epigenetic regulation mechanisms of DNA replication initiation in eukaryotic organism, with a particular focus on histone variants and modifications.
Preprint
DNA replication initiates from ∼50,000 origins on human chromosomes in each cell-cycle and the origins are hypothesized to be specified by binding of factors like the Origin Recognition Complex (ORC) or CTCF or other features like G-quadruplexes. We have performed an integrative analysis of 113 genome-wide human origin profiles (from five different techniques) and 5 ORC-binding site datasets to critically evaluate whether the most reproducible origins are specified by these features. Out of ∼7.5 million 300 bp chromosomal fragments reported to harbor origins by all the datasets, only 0.27% were reproducibly detected by four techniques (20,250 shared origins), suggesting extensive variability in origin usage and identification in different circumstances. 21% of the shared origins overlap with transcriptional promoters, posing a conundrum. Although the shared origins overlap more than union origins with constitutive CTCF binding sites, G-quadruplex sites and activating histone marks, these overlaps are comparable or less than that of known Transcription Start Sites, so that these features could be enriched in origins because of the overlap of origins with epigenetically open, promoter-like sequences. Only 6.4% of the 20,250 shared origins were within 1 kb from any of the ∼13,000 reproducible ORC binding sites in human cancer cells, in contrast to the nearly 100% overlap between the two in the yeast, S. cerevisiae. Thus, in human cancer cell-lines, replication origins appear to be specified by highly variable stochastic events dependent on the high epigenetic accessibility around promoters, without extensive overlap between the most reproducible origins and ORC-binding sites.
Article
Transcription and replication both require large macromolecular complexes to act on a DNA template, yet these machineries cannot simultaneously act on the same DNA sequence. Conflicts between the replication and transcription machineries (transcription–replication conflicts, or TRCs) are widespread in both prokaryotes and eukaryotes and have the capacity to both cause DNA damage and compromise complete, faithful replication of the genome. This review will highlight recent studies investigating the genomic locations of TRCs and the mechanisms by which they may be prevented, mitigated, or resolved. We address work from both model organisms and mammalian systems but predominantly focus on multicellular eukaryotes owing to the additional complexities inherent in the coordination of replication and transcription in the context of cell type–specific gene expression and higher-order chromatin organization. Expected final online publication date for the Annual Review of Genetics, Volume 57 is November 2023. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Cancer has been linked to the uncontrolled proliferation of cells and the overexpression of cell-cycle genes. The cell cycle machinery plays a crucial role in the regulation of the apoptosis to mitosis to growth phase progression. The mechanisms of the cell cycle also play an important role in preventing DNA damage. There are multiple members of the protein kinase family that are involved in the activities of the cell cycle. Essential cyclins effectively regulate cyclin-dependent kinases (CDKs), which are themselves adversely regulated by naturally occurring CDK inhibitors. Despite the fact that various compounds can effectively block the cell cycle kinases and being investigated for their potential to fight cancer. This chapter explains the detail of cell cycle and checkpoint regulators, that are crucial to the malignant cellular process. The known CDKs inhibitors and their mechanism of action in various cancers have also been addressed as a step toward the development of a possibly novel technique for the design of new drugs against cell cycle kinase proteins.
Article
Studying the dynamics of genome replication in mammalian cells has been historically challenging. To reveal the location of replication initiation and termination in the human genome, we developed Okazaki fragment sequencing (OK-seq), a quantitative approach based on the isolation and strand-specific sequencing of Okazaki fragments, the lagging strand replication intermediates. OK-seq quantitates the proportion of leftward- and rightward-oriented forks at every genomic locus and reveals the location and efficiency of replication initiation and termination events. Here we provide the detailed experimental procedures for performing OK-seq in unperturbed cultured human cells and budding yeast and the bioinformatics pipelines for data processing and computation of replication fork directionality. Furthermore, we present the analytical approach based on a hidden Markov model, which allows automated detection of ascending, descending and flat replication fork directionality segments revealing the zones of replication initiation, termination and unidirectional fork movement across the entire genome. These tools are essential for the accurate interpretation of human and yeast replication programs. The experiments and the data processing can be accomplished within six days. Besides revealing the genome replication program in fine detail, OK-seq has been instrumental in numerous studies unravelling mechanisms of genome stability, epigenome maintenance and genome evolution. A protocol for quantitative genome-wide detection of replication initiation, fork progression and termination, based on purification and strand-specific sequencing of Okazaki fragments isolated from mammalian or yeast cells.
Article
Full-text available
DNA replication is a dynamic process that occurs in a temporal order along each of the chromosomes. A consequence of the temporally coordinated activation of replication origins is the establishment of broad domains (>100 kb) that replicate either early or late in S phase. This partitioning of the genome into early and late replication domains is important for maintaining genome stability, gene dosage, and epigenetic inheritance; however, the molecular mechanisms that define and establish these domains are poorly understood. The modENCODE Project provided an opportunity to investigate the chromatin features that define the Drosophila replication timing program in multiple cell lines. The majority of early and late replicating domains in the Drosophila genome were static across all cell lines; however, a small subset of domains was dynamic and exhibited differences in replication timing between the cell lines. Both origin selection and activation contribute to defining the DNA replication program. Our results suggest that static early and late replicating domains were defined at the level of origin selection (ORC binding) and likely mediated by chromatin accessibility. In contrast, dynamic domains exhibited low ORC densities in both cell types, suggesting that origin activation and not origin selection governs the plasticity of the DNA replication program. Finally, we show that the male-specific early replication of the X chromosome is dependent on the dosage compensation complex (DCC), suggesting that the transcription and replication programs respond to the same chromatin cues. Specifically, MOF-mediated hyperacetylation of H4K16 on the X chromosome promotes both the up-regulation of male-specific transcription and origin activation.
Article
Full-text available
Cdc6 and Cdt1 initiate DNA replication licensing when cells exit mitosis. In cycling cells, Cdc6 is efficiently degraded from anaphase onwards as a result of APC/C(Cdh1) activity. When APC/C(Cdh1) is switched off again at the end of G1 phase, Cdc6 could thus reappear, risking DNA re-licensing as long as Cdt1 is still present. Here, we carefully investigated the dynamics of Cdt1 and Cdc6 in cycling cells. We reveal a novel APC/C(Cdh1) independent degradation pathway that prevents nuclear Cdc6 re-accumulation at the G1/S transition and during S-phase. Similar to Cdt1, nuclear clearance of Cdc6 depends on an N-terminal PIP-box and CRL4-Cdt2. When cells reach G2 phase, Cdc6 re-appears rapidly but at this time Cdt1 is mostly absent and Cdc6 expression is limited to the cytoplasm. We propose that Cdk1 contributes to the nuclear export of Cdc6 at the S-to-G2 transition. Summarized, our results show that different Cdc6 control mechanisms restrain erroneous DNA replication licensing during G1 phase, S phase and G2 phase.
Article
Full-text available
Once-per-cell cycle replication is regulated through the assembly onto chromatin of multi-subunit protein complexes which license DNA for a further round of replication. Licensing consists of the loading of the hexameric MCM2-7 complex onto chromatin during G1, and is dependent on the licensing factor Cdt1. In vitro experiments have suggested a two-step binding mode for MCM proteins, with transient initial interactions converted to stable chromatin loading. Here, we assess MCM loading in live human cells, using an in vivo licensing assay based on Fluorescence Recovery After Photobleaching (FRAP) of GFP tagged MCM protein subunits through the cell cycle. We show that in telophase, MCM2 and MCM4 maintain transient interactions with chromatin, exhibiting kinetics similar to Cdt1. These are converted to stable interactions from early G1 phase. The immobile fraction of MCM2 and MCM4 increases during the G1 phase, suggestive of reiterative licensing. In late G1 phase, a large fraction of MCM proteins are loaded onto chromatin, with maximal licensing observed just prior to S-phase onset. Fluorescence Loss In Photobleaching (FLIP) experiments show subnuclear concentration of MCM-chromatin interactions which differ as G1 progresses and do not co-localize with sites of DNA synthesis in S phase.
Article
Full-text available
DNA replication depends on a preceding licensing event by Cdt1 and Cdc6. In animal cells, relicensing after S phase but before mitosis is prevented by the Cdt1 inhibitor geminin and mitotic cyclin activity. Here, we show that geminin, like cyclin B1 and securin, is a bona fide target of the spindle checkpoint and APC/C(Cdc20). Cyclin B1 and geminin are degraded simultaneously during metaphase, which directs Cdt1 accumulation on segregating sister chromatids. Subsequent activation of APC/C(Cdh1) leads to degradation of Cdc6 well before Cdt1 becomes unstable in a replication-coupled manner. In mitosis, the spindle checkpoint supports Cdt1 accumulation, which promotes S phase onset. We conclude that the spindle checkpoint, APC/C(Cdc20), and APC/C(Cdh1) act successively to ensure that the disappearance of licensing inhibitors coincides exactly with a peak of Cdt1 and Cdc6. Whereas cell cycle entry from quiescence requires Cdc6 resynthesis, our results indicate that proliferating cells use a window of time in mitosis, before Cdc6 is degraded, as an earlier opportunity to direct S phase.
Article
DNA replication ensures the accurate duplication of the genome at each cell cycle. It begins at specific sites called replication origins. Genome-wide studies in vertebrates have recently identified a consensus G-rich motif potentially able to form G-quadruplexes (G4) in most replication origins. However, there is no experimental evidence to demonstrate that G4 are actually required for replication initiation. We show here, with two model origins, that G4 motifs are required for replication initiation. Two G4 motifs cooperate in one of our model origins. The other contains only one critical G4, and its orientation determines the precise position of the replication start site. Point mutations affecting the stability of this G4 in vitro also impair origin function. Finally, this G4 is not sufficient for origin activity and must cooperate with a 200-bp cis-regulatory element. In conclusion, our study strongly supports the predicted essential role of G4 in replication initiation.
Article
Tissue homeostasis in metazoans is regulated by transitions of cells between quiescence and proliferation. The hallmark of proliferating populations is progression through the cell cycle, which is driven by cyclin-dependent kinase (CDK) activity. Here, we introduce a live-cell sensor for CDK2 activity and unexpectedly found that proliferating cells bifurcate into two populations as they exit mitosis. Many cells immediately commit to the next cell cycle by building up CDK2 activity from an intermediate level, while other cells lack CDK2 activity and enter a transient state of quiescence. This bifurcation is directly controlled by the CDK inhibitor p21 and is regulated by mitogens during a restriction window at the end of the previous cell cycle. Thus, cells decide at the end of mitosis to either start the next cell cycle by immediately building up CDK2 activity or to enter a transient G0-like state by suppressing CDK2 activity.
Article
Many replication proteins assemble on the pre-RC-formed replication origins and constitute the pre-initiation complex (pre-IC). This complex formation facilitates the conversion of Mcm2-7 in the pre-RC to an active DNA helicase, the Cdc45-Mcm-GINS (CMG) complex. Two protein kinases, cyclin-dependent kinase (CDK) and Dbf4-dependent kinase (DDK), work to complete the formation of the pre-IC. Each kinase is responsible for a distinct step of the process in yeast; Cdc45 associates with origins in a DDK-dependent manner, whereas the association of GINS with origins depends on CDK. These associations with origins also require specific initiation proteins: Sld3 for Cdc45; and Dpb11, Sld2, and Sld3 for GINS. Functional homologs of these proteins exist in metazoa, although pre-IC formation cannot be separated by requirement of DDK and CDK because of experimental limitations. Once the replicative helicase is activated, the origin DNA is unwound, and bidirectional replication forks are established.
Article
The regulated loading of the Mcm2-7 DNA helicase (comprising six related subunits, Mcm2 to Mcm7) into pre-replicative complexes at multiple replication origins ensures precise once per cell cycle replication in eukaryotic cells. The origin recognition complex (ORC), Cdc6 and Cdt1 load Mcm2-7 into a double hexamer bound around duplex DNA in an ATP-dependent reaction, but the molecular mechanism of this origin 'licensing' is still poorly understood. Here we show that both Mcm2-7 hexamers in Saccharomyces cerevisiae are recruited to origins by an essential, conserved carboxy-terminal domain of Mcm3 that interacts with and stimulates the ATPase activity of ORC-Cdc6. ATP hydrolysis can promote Mcm2-7 loading, but can also promote Mcm2-7 release if components are missing or if ORC has been inactivated by cyclin-dependent kinase phosphorylation. Our work provides new insights into how origins are licensed and reveals a novel ATPase-dependent mechanism contributing to precise once per cell cycle replication.