Está en la página 1de 9

REVIEW

The control of DNA repair by the cell cycle


Nicole Hustedt and Daniel Durocher

The correct duplication and transmission of genetic material to daughter cells is the primary objective of the cell division cycle.
DNA replication and chromosome segregation present both challenges and opportunities for DNA repair pathways that safeguard
genetic information. As a consequence, there is a profound, two-way connection between DNA repair and cell cycle control. Here,
we review how DNA repair processes, and DNA double-strand break repair in particular, are regulated during the cell cycle to
optimize genomic integrity.

Maintenance of genome stability is essential for normal development, joining (EJ) and homologous recombination (HR). EJ pathways re-ligate
cellular homeostasis and tumour suppression1. It relies on the precise DNA ends together with some or no processing of the ends themselves.
orchestration of DNA replication, chromosome segregation, DNA repair They operate throughout interphase and are inhibited during the pro-
and genome surveillance mechanisms, along with their integration with cess of mitosis. EJ pathways are subdivided into the non-homologous
cell cycle progression and myriad other processes such as transcrip- end-joining pathway (NHEJ) (which is dependent on DNA ligaseIV
tion and metabolism. The modulation of DNA repair by the cell cycle and Ku), and a group of less well elucidated alternative end joining
is largely due to the fact that chromatin, the substrate of DNA repair, (A-EJ) pathways3,4 (dependent on DNA ligaseI/III) (Fig.1). While the
undergoes major transitions as DNA is replicated, compacted and untan- template-independent nature of NHEJ has led many to suggest that it
gled to allow for cell division. At the same time, DNA replication repre- is error prone, it is probably accurate in the majority of cases5. The best
sents both a challenge and an opportunity for DNA repair. defined A-EJ pathway is theta-mediated end joining (TMEJ), where ends
The mitotic cell cycle is defined as the ordered series of events occur- processed to produce 3 overhangs are annealed at short stretches of
ring in distinct phases that ultimately results in the division of a cell into homologous sequences, as small as one nucleotide, to drive deletion of
two daughters2. Two key events punctuate the cell cycle: DNA replica- the intervening sequences and ligation. TMEJ is dependent on PARP1,
tion, when the bulk of the nuclear genome is duplicated, defines S (syn- DNA ligaseIII and the polymerase activity of DNA polymerase (POL,
thesis) phase; and mitosis, defined as the period in which chromosomes encoded by POLQ)3,6. Evidence suggests that POL-mediated TMEJ acts
are condensed, sorted and then equally distributed to daughter cells, as a backup for both NHEJ and HR, since loss of POLQ is lethal in the
delineates M phase. Between the M and S phases are the gap phases G1 context of cells with mutations in both KU/53BP1 and BRCA1/BRCA2,
and G2, respectively 2. Cells can also enter quiescence (or G0), a state which impair NHEJ and HR, respectively 68.
of replicative dormancy. The metronome driving cell cycle progression In contrast, HR comprises repair systems that employ extensive
consists of the periodic activation and inactivation of cyclin-dependent homology and templated DNA synthesis to regenerate the sequence
Ser/Thr kinases (CDKs)2. In mammals, the main cell cycle CDKs (CDK1, surrounding the break site9. The canonical HR pathway that mediates
CDK2, CDK4 and CDK6) partner with specific cyclins to form active the repair of two-ended DSBs is gene conversion (Fig.2), although HR
kinase holoenzymes. CDK1 pairs with cyclins A/B, CDK2 with cyclin is also critical for the repair of one-ended DSBs, such as those produced
A/E and CDK4/6 interact with Dtype cyclins. CDK activity is generally by broken replication forks9. As the ideal template for HR is the sister
low in G1 and rises progressively until it reaches maximal activity upon chromatid, HR is restricted to the S and G2 phases of the cell cycle10.
mitotic entry. Destruction of the A- and B-type cyclins mediated by HRmediated DSB repair requires the formation of extensive single-
the anaphase-promoting complex cyclosome (APC/C), an E3 ubiquitin stranded (ss) DNA through a process termed DNA end resection11 that
ligase triggers mitotic exit, thereby re-establishing the G1 state. requires, for most types of ends, the action of CtIP (ref.12). The role of
CtIP is not yet completely understood, but based on studies in budding
The rise and fall of DNA double-strand break repair yeast 13, it is believed to stimulate the endonuclease activity of MRE11
The most dramatic example of DNA repair regulation by the cell cycle in complex with RAD50 and NBS1 (the MRN complex). After endo-
is certainly the periodic inhibition of DNA double-strand break (DSB) nucleolytic cleavage, resection proceeds bi-directionally 14,15, with the
repair (Figs13). Two main groups of repair pathways mend DSBs: end exonuclease activity of MRN proceeding in the 35 direction, whereas

Nicole Hustedt and Daniel Durocher are at the Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, M5G 1X5,
Canada. Daniel Durocher is also in the Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 3E1, Canada.
e-mail: durocher@lunenfeld.ca

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 1



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEW

Nuclear envelope to homology search, strand invasion and DNA polymerization, processes
by which the sequence surrounding the break site is resynthesized. This

CtIP
BRCA1 pathway generates joint DNA molecules, from D-loops to the four-way
Holliday junction (HJ). HR-mediated DSB repair is completed through
Exo1
RNF8 PTIP
the disassembly of these joint molecules. Extended D-loops can be
P BLM DNA2
RNF168 P
P directly unwound by helicases followed by ligation in a process termed

F1
MDC1 53BP1 synthesis-dependent strand annealing (SDSA)9. Alternatively, when

RI
P
P ATM
Me Me Ub
Ub both ends invade the template, double-HJs are formed. Double-HJs can
M N M N
undergo converging branch migration followed by the processing of the

P
CtIP
R R resulting hemicatenane. This process, termed HJ dissolution, is cata-
EXO1 lysed through the combined action of topoisomeraseIII and the RecQ
HELB RPA BLM DNA2
family helicase BLM bound to RMI1 and RMI2 (the BTR complex)19,20.
P ATM-dependent
Alternatively, nucleases can cleave HJs in a process termed resolution. In
P CDK-dependent
Limited end processing eukaryotes, the main HJ resolvases are the SLX4 complex (comprising
P PLK3-dependent
SLX4SLX1/MUS81EME1 and XPFERCC1) and the GEN1 nucle-
LIGI
LIGIII ase (Yen1 in budding yeast). BTR produces exclusively non-crossover
POLQ
formations that avoid inter-homologue exchanges and thus loss of het-
PARP1
Classical Alternative erozygosity 20,21. In the absence of BLM, cells show an increase in crosso-
non-homologous end joining vers, detectable by sister chromatid exchanges (SCEs) that depend on
end joining (TMEJ)
the structure-specific endonucleases MUS81EME1, SLX1SLX4 and
GEN122,23. When resection reveals direct repeats flanking the break site,
KU70 a RAD52-dependent process termed single-strand annealing (SSA)3,9
KU80
LIGIV can repair the break with the ensuing loss of the intervening sequences.
End joining products:
SSA is, by definition, error prone and thus resection must be balanced
525 bp homology
to promote gene conversion over SSA.

05 bp insertions/deletions Variable size insertion/deletions End resection gets a helping hand from CDKs
By virtue of being the first step shared by the HR and A-EJ processes, and
by being simultaneously inhibitory to NHEJ (in particular to Ku bind-
NHEJ: ON ing 24), DNA end resection is the critical node for the regulation of DSB
A-EJ: ON repair by the cell cycle25. The regulation of resection is often reformulated
HR: OFF (pre-RAD51);
ON (HJ dissolution)
in the context of DSB repair pathway choice, since resection helps set-
OFF (HJ resolution) tle the competition between repair systems that has been observed in a
G1 Early S wide variety of organisms2629. The importance of end stability in DSB
CDK activity repair pathway selection was first recognized in budding yeast 30, and it
was there that CDK activity was found to be essential for activation of
Figure 1 Repair of double-strand breaks in G1 and early Sphase. ATM DNA end resection31,32. The involvement of CDK in eukaryotic resection
is recruited to DSBs through MRN and phosphorylates targets such as is conserved in all species examined, including in human cells33.
nucleosomes (in particular H2AX, resulting in H2AX) (top left). H2AX The rising levels of CDK activity that characterizes Sphase entry
recruits MDC1, also an ATM target. MDC1 phosphorylation recruits the E3
ubiquitin ligase RNF8, which, through recruitment of a second E3 ubiquitin
and progression results in the phosphorylation and activation of the
ligase (RNF168), leads to histone H2A ubiquitylation. This modification, enzymes that catalyse end resection while simultaneously weakening the
together with H4K20 methylation, allows for 53BP1 recruitment (top impediments to this process. In budding yeast, Cdk1 (Cdc28)-dependent
right). 53BP1 is phosphorylated at its Nterminus in an ATM-dependent phosphorylation of the CtIP homologue Sae2 on Ser267 (ref.34) stimu-
manner, creating docking sites for RIF1 and PTIP. 53BP1 blocks resection
at DSBs, thereby channelling DSB repair towards NHEJ. Long-range
lates Mre11 endonuclease activity 13. Cdk1 also stimulates the ability of
resection is also limited through HELB. End joining can be divided into the nucleosome remodeller Fun30 to antagonize the 53BP1 orthologue
two pathways: Ku- and DNA-ligase-IV-dependent classical non-homologous Rad9 (ref.35), an inhibitor of end resection36. In vertebrates, CtIP is
end joining (NHEJ, left), and PARP1-, POL- and LigI/III-dependent DNA phosphorylated on an equivalent residue, Thr847, which is critical for
polymerase-mediated end joining (TMEJ, right). The intensity of shading
in the grey band (bottom) indicates the magnitude of CDK activation in the initiation of DNA end resection37. However, CtIP phosphorylation
G1/early Sphase. P, phosphorylation; Ub, ubiquitylation; Me, methylation; is complex and several other residues are phosphorylated38,39, including
red arrows, resection. Ser327, which is targeted by CDKs to promote binding to BRCA140,41,
possibly to stimulate DNA end resection4244. Additionally, the CDK2-
the redundant activities of EXO1 and the BLM/DNA2 nucleases cata- dependent phosphorylation of Ser276 and Thr315 promotes binding to
lyse long-range 53 resection16. The ensuing ssDNA is first bound the cis-trans prolyl isomerase PIN1 to dampen, rather than stimulate,
by RPA, which must then be replaced by RAD51 to form a nucleofila- ssDNA formation45.
ment competent for HR through the action of BRCA1, PALB2 and the Whereas the phosphorylation of CtIP by CDKs is clearly involved
BRCA2 recombination mediator 17,18. The RAD51 recombinase is central in the cell cycle regulation of end resection, as demonstrated by

2 NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEW

P highlight that additional levels of control must exist to enforce strict cell
HELB cycle regulation of resection and HR, and raise the question of how the
Nuclear enve
lope complex, multi-site phosphorylation of CtIP is integrated to produce the
optimal response to DSBs.
In addition to CtIP regulation, CDK activity promotes end resection
RNF8 PTIP
P P
through the phosphorylation of EXO148 and NBS14951. In budding yeast,
RNF168 P
Dna2 phosphorylation by Cdk1 is also involved in stimulating DNA end

F1
MDC1 53BP1

RI
P
ATM BRCA1
Me
Ub P Me Ub resection52, which suggests that each of the main complexes involved in

P
CtIP
M N the generation of ssDNA at DSB sites are positively regulated by CDK-
P

dependent phosphorylation.
R P
CDKs also promote DNA end resection by weakening the activity of
two distinct blocks to resection: one mediated by the chromatin-binding
P RPA
protein 53BP1 (explored in more detail below), and a second embod-
RPA
BLMDNA2 ied by HELB, an ssDNA translocase. HELB is recruited to DSBs via
EXO1
an interaction with RPA53,54, which suggests a feedback mechanism in
PALB2
BRCA1
which resected ends themselves prohibit resection. The Cterminus of
BRCA2 HELB contains dual nuclear import and export signals that are modu-
RAD51
lated by CDK2 phosphorylation such that nuclear HELB concentration
RAD51 decreases as CDK2 activity rises55,56. As the function of HELB requires
its nuclear localization53, these observations suggest that the HELB-
dependent block to resection weakens as Sphase progresses. Finally,
cyclins play direct, non-canonical roles in promoting HR: cyclinD1
TOPIIIA binds to RAD51 to promote its recruitment to DSBs57, while cyclinA2
BLM
HR products: R1 R2 upregulates MRE11 expression in Sphase by binding to its mRNA and
promoting its translation58.

Dissonance between Sphase and HR


Non-crossover Crossover Although it is often said that HR is activated in Sphase, this is not entirely
accurate: early studies clearly established that HR is suppressed in early
Sphase26,29,59,60. In one example26, NHEJ-defective Ku70 avian DT40
SLX4 EME1
cells are hypersensitive to ionizing radiation in G1 and early Sphase,
SLX1
MUS81 but become increasingly radioresistant upon replication of a large por-
GEN1 tion of their genome (mid-S onwards). Similar results were obtained by
enumerating H2AX foci in cells deficient in DNA-PKcs (ref.60) or by
NHEJ: ON
A-EJ: ON monitoring the recruitment of RAD5261. Together these experiments sug-
HR: ON (pre-RAD51); gest that HR activation and Sphase entry are uncoupled from each other.
ON (HJ dissolution)
This dissonance is not entirely surprising, because early S is similar to G1
OFF (HJ resolution)
in that only a small fraction of the genome is replicated. The question is
Late S G2
therefore whether there is a post-replicative and local activation of HR
CDK activity
behind the fork (Fig.4a), whether the local accumulation of pro-HR fac-
tors on replicated DNA induces global activation of recombination, or
Figure 2 Repair of double-strand breaks in late Sphase. Top: during Sphase,
CDK activity rises, resulting in phosphorylation of NBS1, HELB and CtIP. whether there is simply a timer that activates HR in mid-Sphase (Fig.4b).
NBS1 and CtIP phosphorylation stimulates MRN endonuclease activity and The simplest model for explaining HR activation during Sphase pro-
allows BRCA1 to bind to CtIP. BRCA1 counteracts 53BP1. Phosphorylation poses that replicated chromatin is competent for HR whereas unrepli-
of HELB results in its nuclear export. Middle: long-range resection proceeds
through the combined action of DNA2/BLM and EXO1, also a CDK target.
cated chromatin remains refractory to this type of repair (Fig.4a). The
BRCA1, BRCA2 and PALB2 promote replacement of RPA by RAD51 on recent discovery that the lack of methylation at the Lys20 residue of
ssDNA. Bottom: strand invasion, joint molecule and dHJ formation are steps histoneH4 (H4K20me0) represents a post-replicative chromatin mark62
of HR. dHJs can be dissolved by the BTR complex, resulting in non-crossover has a number of implications for this model. H4K20me0 is specifically
product formation; activity towards dHJ of the resolvases MUS81EME1 and
SLX1SLX4 separately is low. Another dHJ resolvase, GEN1, is excluded
recognized by the TONSL subunit of the MMS22LTONSL com-
from the nucleus. Red arrows, resection; green arrowheads, dHJ dissolution; plex 62, suggesting that this complex labels newly replicated chromatin.
brown arrowheads, dHJ resolution. MMS22LTONSL is a poorly characterized HR factor that promotes
RAD51 loading in response to DNA replication stress6365, suggesting
unscheduled activation of resection in G1 cells through the T847E phos- a direct link between post-replicative chromatin marking and HR. As
phomimetic CtIP mutant 37,46, there is also evidence that CtIP can be H4K20me0 is also incompatible with 53BP1 recruitment66,67, H4K20me0
active in G1 to promote A-EJ. Ser327 and Thr847 can be phosphorylated and its recognition may tip the balance of repair in favour of HR via two
by PLK3 in G1 cells independently of CDK activity 47. These observations independent means.

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 3



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEW

In budding yeast, cohesin must be actively loaded at DSB sites to pro-


P
mote HR, as replication-coupled sister cohesion establishment is insuf-
RNF8 RNF168
ficient to promote repair 71,72. Cohesin factors are also loaded onto DSB
53BP1
sites in mammalian cells, suggesting that they also act at a late stage
P P P in DNA repair 73. Collectively, these observations suggest that cohesion
MDC1 improves the efficiency of the HR reaction rather than playing a direct
ATM
Me P Me role in its activation.
N M M N The post-replicative marking model of HR activation does not eas-
P

ily explain how DNA end resection is activated as Sphase progresses


CtIP

P R R
P

unless one invokes, for example, the titration of a resection inhibitor as


new chromatin is formed. One can rather envisage that the rise in CDK
activity seen in Sphase is interpreted as a sort of timer that triggers a
P P P rise in DNA end resection activity (Fig.4b). This behaviour could be
accomplished through the multisite phosphorylation of CtIP, coupled to
P

HELB No extensive the CDK2-dependent exclusion of HELB from the nucleus. The increase
PALB2 P
resection BRCA1 in HR factor abundance in Sphase may also contribute to an HR timer.
BRCA2
The resection timer concept is different from post-replicative marking
No RAD51 filament formation of chromatin because the former is probabilistic in nature whereas the
latter is deterministic with respect to DSB repair pathway choice.

TOPIIIA 53BP1 and BRCA1 battle for real estate


BLM
HR products:
R1 R2 53BP1 accumulates at DSB sites by recognizing nucleosomes modified
by methylation on histoneH4 Lys20 and ubiquitylation of H2A Lys15
by RNF16866,67,74. 53BP1 promotes NHEJ and opposes HR (and A-EJ)
at least in part by blocking DNA end resection. Limiting end resection
Non-crossover Crossover requires the ATM-dependent phosphorylation of the 53BP1 N-terminal
region, which in turn promotes the recruitment of PTIP and RIF17580,
which are both independently involved in blocking DNA end resection.
P P GEN1
The function of RIF1 in blocking end resection also involves MAD2L2
SLX4 EME1 (also known as REV7)81,82.
SLX1 MUS81 For DNA end resection to be activated in S/G2 cells, cells must weaken
the end-protection activity of 53BP1 in a manner that involves BRCA1
(Fig.2). Indeed, loss of 53BP1 in mice suppresses defective HR and the
NHEJ: OFF
lethality caused by loss-of-function mutations in Brca18385. As 53BP1
A-EJ: OFF (?) inactivation does not suppress the defects associated with mutations in
HR: OFF (pre-RAD51); other HR genes, these results imply that a major function for BRCA1
ON (HJ dissolution)
ON (HJ resolution) is to antagonize 53BP1. Further evidence for this model emerged when
Early mitosis Late mitosis
analyses of 53BP1 localization to DSB sites by structured illumination
CDK activity microscopy found that 53BP1 ionizing-radiation-induced foci were
reshaped by BRCA1 in S/G2 cells86. This may be due to the BRCA1-
Figure 3 Repair of double-strand breaks in mitosis. Top: RNF8 and 53BP1 mediated ubiquitylation of histoneH2A, which enables the nucleosome
phosphorylation in mitosis blocks their recruitment to DSBs. DSB ends may remodeller SMARCAD1 to antagonize 53BP187. BRCA1 also inhibits
be held together until their repair in the next G1 phase. Middle: resection is the recruitment of RIF1 to DSB sites77,78,88 in a manner that involves its
limited by HELB, and the phosphorylation of RPA and BRCA2 contributes
to inhibition of RAD51 replacement. Mitotic late-stage HR intermediates interaction with CtIP and the CDK-dependent phosphorylation of CtIP
must be removed before chromosome segregation. Bottom: CDK1-dependent at residue Ser32777. While some studies suggest that loss of RIF1 or REV7
phosphorylation promotes MUS81EME1SLX1SLX4 holocomplex suppresses the HR deficiency of BRCA1-mutated cells77,81, other studies
formation. GEN1 gains access to chromatin after nuclear envelope
instead point to PTIP being the key 53BP1 effector involved in opposing
breakdown. Unlike BTR-mediated dissolution, dHJ resolution results in
both crossover and non-crossover product formation, depending on the HJ BRCA1-dependent DSB repair 79.
cleavage orientation. Conversely, the recruitment of BRCA1 to DSB sites in G1 cells is
suppressed by a pathway that involves 53BP1 and RIF177,78 (Fig. 1).
Another event associated with DNA replication is the establish- 53BP1RIF1 and BRCA1CtIP are thus engaged in a battle for real
ment of sister chromatid cohesion. Its main effector, cohesin, promotes estate at the site of DNA ends that is refereed by CDKs and histone
HR in a variety of systems6870 and could be a key determinant for the modifications such as H4 (Lys16) and H2A (Lys15) acetylation89,90.
Sphase activation of HR. However, one must discriminate between the While this antagonism represents a mechanism regulating DSB
bias towards sister chromatid recombination caused by the tethering repair pathway choice during the cell cycle, recent evidence suggests
of chromatids versus choice between HR and competing pathways. that 53BP1 and BRCA1 may instead fine-tune each others activity.

4 NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEW

a
Unreplicated chromatin Replicated chromatin

MMS22L MMS22L MMS22L

TONSL TONSL TONSL

53BP1
Me Me Ub Me
53BP1
H4K20me2
MMS22L MMS22L

H4K20me0 Ub TONSL TONSL

NHEJ

sin
he
Co
HR

b G1 S G1 S
mid-S mid-S

M G2 M G2

P
Nu

Nu
cle

CDK activity low CDK sufficiently high HELB

cle
ar

CDK CDK

ar
en

HELB

en
vel

vel
ope

ope
CtIP

P
EXO1 RPA P
P
CtIP

M N M N
P

EXO1
R R

NHEJ HR

Figure 4 Models for cell-cycle-dependent activation of repair by homologous recombination. (a) The post-replicative chromatin marking model. Incorporation
of unmethylated histone H4 Lys20 in newly replicated chromatin. H4K20me0 is recognized by MMS22LTONSL, which promotes HR. H4K20me0 also
precludes binding of 53BP1. Cohesin also labels newly replicated chromatin and promotes HR. (b) The timer model. CDK activity progressively rises during
Sphase, triggering enough phosphorylation of NBS1, CtIP, HELB and EXO1 to stimulate resection and HR.

In particular, 53BP1 limits the extent of resection in S/G2 cells91, thereby the identification and repair of DNA lesions. Consequently, replica-
decreasing the probability of engaging mutagenic RAD52-dependent tion-dependent and -independent repair pathways exist for a subset
SSA. Therefore, 53BP1 can be viewed, paradoxically, as a factor that of lesions. A clear case of this situation is the repair of interstrand
promotes the fidelity of HR91. crosslinks (ICLs). ICLs block replisome progression, which initiates
repair pathways that either involve the glycosylase NEIL3 (ref. 93)
Risks and opportunities of genome scanning by the replisome or by the Fanconi anaemia (FA) group of proteins94. The replication-
Semi-conservative DNA replication is a remarkable process that must dependent FA pathway coordinates incision on each side of the lesion,
be precisely regulated to occur exactly once per cell cycle with mind- translesion DNA synthesis followed by HR, and nucleotide excision
boggling accuracy and speed especially considering that DNA rep- repair (NER), which together regenerate an undamaged DNA duplex.
lication forks must replicate past difficult DNA structures, repetitive While ICLs are a threat to genome replication, they also block tran-
sequences, DNA lesions, RNADNA hybrids, tightly bound proteins scription and so their removal must occur independently of cell cycle
and collisions with transcribing RNA polymerases92. Alternatively, as position. The block to RNA polymerase progression is a plausible
replisomes scan the entire genome, it also provides an opportunity for mechanism for the initiation of replication-independent ICL repair 95

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 5



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEW

alongside the detection of helix-distorting ICLs by the NER pathway 96. depends on the action of endonucleases, such as MUS81EME1, in early
Replication-independent ICL repair is poorly understood, although it has mitosis112,113. MUS81EME1 cleaves a broad range of branched DNA
been shown in Xenopus laevis extracts to involve PCNA ubiquitylation structures23,114,115 that have been proposed to occur at CFSs, although the
and DNA polymerase (ref.97) at a step downstream of lesion unhook- exact nature of these structures is still under investigation.
ing 95. Exactly how replication-dependent and -independent ICL repair In addition to processing late-replication intermediates, cells also
pathways compete or cooperate to remove these toxic lesions is unknown. attempt to complete the replication of at-risk regions such as CFSs. In
Although ICL repair harnesses the genome-scanning properties particular, a POLD3-dependent DNA synthesis pathway, reminiscent
of DNA replication, the repair of most lesions at stalled forks is risky of break-induced replication, occurs at MUS81-processed CFSs in early
because their excision in the context of an unwound template will result mitosis116. In the absence of MUS81EME1, cells display increased lev-
in a DSB. To compensate for the inability of most types of excision repair els of FANCD2-marked, DAPI-positive anaphase bridges along with
pathways to operate in ssDNA, cells have evolved the DNA damage tol- FANCD2-bound ultra-fine bridges (UFBs) that are DAPI-negative but
erance pathway (also known as DNA damage bypass), which is not a coated with the translocase PICH, the BTR complex and RIF1112,113,117119.
DNA repair process perse as it enable replisomes to bypass fork-blocking The localization of these proteins at CFSs may represent a last effort to
lesions. This pathway consists of two general branches: template switch- facilitate sister chromatid disjunction. Loss of these UFB binding and
ing and translesion DNA synthesis, which are both initiated following processing factors leads to micronucleation and spontaneous 53BP1 foci
detection of extended stretches of RPA-coated ssDNA at stalled DNA in the next G1 phase, which are often referred to as 53BP1 bodies. The
replication forks. The formation of ssDNA promotes various flavours function of 53BP1 bodies is still mysterious, but they may mark, protect
of PCNA ubiquitylation that activate either template switching or the or facilitate the repair of these inherited DNA lesions112,113. Collectively,
recruitment of translesion synthesis polymerases98. DNA damage toler- these findings indicate that CFS expression through the action of struc-
ance is often referred to as post-replicative repair, and studies in budding ture-specific nucleases in mitosis is beneficial for chromosome segrega-
yeast have clearly established that it remains competent long after the tion rather than being a genome-destabilizing event.
completion of bulk replication in G299,100 which suggests that daughter
strand gaps may be marked, perhaps by PCNA, until they are repaired. Paradoxical inhibition of DSB repair in Mphase
While cells attempt to complete DSB repair before the onset of chromo-
M phase DNA repair deals with unfinished business some segregation, they might also face new DNA lesions during mitotic
The segregation of chromatids to opposite poles is the core mission of progression. It has been known for decades that from late prophase
mitosis. It is therefore imperative that all links between sister chromatids onwards, cells do not repair mitotic DSBs or delay mitotic progression
are severed at the onset of anaphase. While cohesin removal and sister as a consequence of their detection120,121. There are some exceptions to
chromatid decatenation by topoisomeraseII clears most chromatid link- this situation: limited mitotic DNA repair occurs in response to etopo-
ages101,102, DNA replication intermediates and other types of joint DNA side treatment, perhaps reflecting the high activity of topoisomeraseII in
molecules such as HJs can persist until mitosis23,103. Mitotic cells, in fact, mitosis122,123, and a robust metaphase arrest can be induced by high doses
activate pathways that process such joint molecules to promote chromo- of DNA damage that impair centromere function, thereby activating the
some segregation (Fig.3). spindle assembly checkpoint 124,125.
One such pathway is the mitotic activation of HJ resolution, which Mitotic cells do, however, sense DSBs, and can initiate a DSB signal-
is triggered by the CDK1-dependent phosphorylation of SLX4 and ling cascade that is comparable, in its early stages, to that of interphase
EME1 at the onset of mitosis23. This event promotes the interaction of cells. Indeed, NBS1 and Ku both localize to mitotic DSBs, and breaks
SLX1SLX4, a HJ nickase, with MUS81EME1, which is active against induce H2AX foci that colocalize with MDC1, as expected126132. H2AX
nicked HJs23. The other HJ resolvase, GEN1, is also activated upon phosphorylation occurs in an ATM- and DNA-PK-dependent manner,
mitotic entry as it gains access to chromatin only after nuclear envelope indicating that both kinases respond to mitotic DNA breaks128. However,
breakdown due to its nuclear export sequence104,105. Its budding yeast the signalling cascade downstream of MDC1 is interrupted in mitotic
orthologue, Yen1, is also excluded from the nucleus during interphase cells, as RNF8, RNF168, BRCA1 and 53BP1 all fail to colocalize with
due to phosphorylation that is reversed upon anaphase entry 106,107. As H2AX during this cell cycle stage128,129,133. The first block to this cascade
HJ resolution results in the formation of either non-crossover or less is at the level of RNF8 recruitment due to a CDK1-dependent inhibi-
desirable crossover products, the mitotic activation of HJ resolution is tion of the RNF8MDC1 interaction134. The inhibitory CDK1 site on
a last-ditch attempt to process the recombination intermediates that human RNF8 is not conserved in other species, thus alternative means
escaped the attention of the BTR complex 108. to inhibit RNF8 recruitment in those species must exist. A second block
Late and incomplete DNA replication represents a second class of to this cascade involves 53BP1 phosphorylation in Mphase128,129,135,136
liability for chromosome segregation. This is particularly true for loci in particular, phosphorylation of the Thr1609 and Ser1618 residues
referred to as common fragile sites (CFSs) that are replicated late in the by CDK1 (or p38 kinase) and PLK1, respectively 134,136,137. The phospho-
cell cycle, especially under conditions in which replication is challenged rylation of these two residues inhibits the interaction of 53BP1 with
with low doses of the DNA polymerase inhibitor aphidicolin109,110. CFSs ubiquitylated nucleosomes67 and thus blocks its accumulation on dam-
are said to be expressed when metaphase chromosomes display gaps aged chromatin134,136,137. Mutation of Thr1609 and Ser1618 to non-phos-
or breaks110. CFSs usually reside in very large genes with long introns, phorylatable residues alleviates the impediment to 53BP1 recruitment
and their instability may arise from lack of DNA replication origins, late and partially restores mitotic DSB repair (presumably through NHEJ).
DNA replication intermediates, DNA secondary structures, or collisions The partial nature of mitotic DSB repair activation is likely to be due to
between transcription and replication machineries111. CFS expression another block to mitotic NHEJ that involves XRCC4 phosphorylation122.

6 NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEW

DNA repair stimulated by 53BP1 is, paradoxically, deleterious during ACKNOWLEDGEMENTS


the process of mitosis, as it leads to ionizing radiation hypersensitivity We thank R. Szilard, M. Zimmermann, S. Noordermeer and A. Sartori for critical
reading of the manuscript, as well as D. DAmours and J. Lukas for stimulating
and chromosome segregation defects that are caused by sister-telomere discussions and advice. N.H. is a long-term fellow of the Human Science Frontier
fusions134. Mitotic telomeres often show signs of DNA damage, espe- Program. D.D. is a Canada Research Chair (Tier1) in the Molecular Mechanisms
cially in cells undergoing prolonged mitotic arrest 138 or those that are of Genome Integrity. Funding for work in the laboratory of D.D relating to the
regulation of DNA repair by the cell cycle include CIHR grant FDN143343 and a
concurrently damaged with intrachromosomal DSBs134,139. Interestingly, Grant-in-Aid from the Krembil Foundation.
in both cases, the mitotic kinase AuroraB is critical for the induction
of a telomeric DNA damage response134,138. These observations suggest ADDITIONAL INFORMATION
Reprints and permissions information is available online at www.nature.com/
that repair by NHEJ may be blocked in mitosis in order to tolerate some reprints. Correspondence should be addressed to D.D.
levels of telomere uncapping, at the potential cost of mis-segregating
acentric fragments produced by mitotic DSBs on chromosome arms. The COMPETING FINANCIAL INTERESTS
N.H. declares no competing financial interests. D.D. is a paid advisor and
function of this transient telomere uncapping is unknown but it could be receives funding from CRISPR Therapeutics, and also receives funding from
linked to telomere segregation or to a pathway that triggers the perma- Blueline Bioscience.
nent cell cycle arrest of damaged mitotic cells in the following G1 stage140.
1. Jackson, S.P. & Bartek, J. The DNA-damage response in human biology and disease.
While HJ resolution pathways are activated in mitosis, extensive Nature 461, 10711078 (2009).
DNA end resection, CHK1 activation and RAD51 loading is blocked 2. Murray, A.W. & Hunt, T. The Cell Cycle: An Introduction (Freeman, 1993).
3. Sfeir, A. & Symington, L.S. Microhomology-mediated end joining: a back-up survival
in mitosis141,142. CHK1 inactivation is due to the degradation of Claspin, mechanism or dedicated pathway? Trends Biochem. Sci. 40, 701714 (2015).
its activator, at the onset of mitosis143. RAD51 loading is inhibited in 4. Lieber, M.R. The mechanism of double-strand DNA break repair by the nonhomologous
DNA end-joining pathway. Annu. Rev. Biochem. 79, 181211 (2010).
a CDK1-dependent manner, and ectopic CDK1 activation in Xenopus 5. Betermier, M., Bertrand, P. & Lopez, B.S. Is non-homologous end-joining really an
Sphase extracts abrogates RAD51 chromatin binding 141. Both RPA and inherently error-prone process? PLoS Genet. 10, e1004086 (2014).
6. Wyatt, D.W. etal. Essential roles for polymerase theta-mediated end joining in the
BRCA2 are phosphorylated by cyclin-dependent kinases, which may repair of chromosome breaks. Mol. Cell 63, 662673 (2016).
also contribute to the inhibition of RAD51 filament assembly or stabil- 7. Mateos-Gomez, P.A. etal. Mammalian polymerase theta promotes alternative NHEJ
and suppresses recombination. Nature 518, 254257 (2015).
ity 144146. Elevating CDK1 activity in interphase through WEE1 inhibition 8. Ceccaldi, R. etal. Homologous-recombination-deficient tumours are dependent on
decreases HR efficiency, emphasizing a role for CDK1 in blocking HR Pol-mediated repair. Nature 518, 258262 (2015).
9. Verma, P. & Greenberg, R.A. Noncanonical views of homology-directed DNA repair.
in mitosis147. Genes Dev. 30, 11381154 (2016).
10. Essers, J. et al. Analysis of mouse Rad54 expression and its implications for
homologous recombination. DNA Repair 1, 779793 (2002).
Are mitotic chromosomal fragments held together? 11. Symington, L.S. & Gautier, J. Double-strand break end resection and repair pathway
Broken chromosomes represent a major problem for mitotic cells, choice. Annu. Rev. Genet. 45, 247271 (2011).
12. Sartori, A.A. etal. Human CtIP promotes DNA end resection. Nature 450, 509514
since the segregation of the resulting acentric chromatid fragments (2007).
should be random in the absence of compensatory mechanisms. 13. Cannavo, E. & Cejka, P. Sae2 promotes dsDNA endonuclease activity within Mre11-
Rad50-Xrs2 to resect DNA breaks. Nature 514, 122125 (2014).
Such mechanisms would allow mitotic cells to suppress DNA repair 14. Garcia, V., Phelps, S.E., Gray, S. & Neale, M.J. Bidirectional resection of DNA double-
with minimal consequences. Evidence for their existence is found in strand breaks by Mre11 and Exo1. Nature 479, 241244 (2011).
15. Shibata, A. etal. DNA double-strand break repair pathway choice is directed by distinct
Drosophila melanogaster, in which the ends of mitotic DNA breaks are MRE11 nuclease activities. Mol. Cell 53, 718 (2014).
tethered through a DAPI-stainable DNA bridge. This bridge is covered 16. Nimonkar, A.V. etal. BLM-DNA2-RPA-MRN and EXO1-BLM-RPA-MRN constitute two
DNA end resection machineries for human DNA break repair. Genes Dev. 25, 350362
with the mitotic kinases Polo, AuroraB and BubR1 and, importantly, (2011).
facilitates efficient segregation of acentric chromosome fragments148. It 17. Park, J.Y., Zhang, F. & Andreassen, P.R. PALB2: the hub of a network of tumor
suppressors involved in DNA damage responses. Biochim. Biophys. 1846, 263275
is currently unclear whether a similar mechanism exists in mammals, (2014).
but it has been speculated that MRN could also carry out its DNA end- 18. Roy, R., Chun, J. & Powell, S.N. BRCA1 and BRCA2: different roles in a common
pathway of genome protection. Nat. Rev. Cancer 12, 6878 (2011).
tethering activity during mitosis149. 19. Cejka, P., Plank, J.L., Bachrati, C.Z., Hickson, I.D. & Kowalczykowski, S.C. Rmi1
stimulates decatenation of double Holliday junctions during dissolution by Sgs1-Top3.
Nat. Struct. Mol. Biol. 17, 13771382 (2010).
Conclusion 20. Wu, L. & Hickson, I.D. The Blooms syndrome helicase suppresses crossing over during
The cell division cycle is intimately linked with genome protection, and homologous recombination. Nature 426, 870874 (2003).
21. Ira, G., Malkova, A., Liberi, G., Foiani, M. & Haber, J. E. Srs2 and Sgs1-Top3
this Review only shines light on a few salient examples. We anticipate suppress crossovers during double-strand break repair in yeast. Cell 115, 401411
that the cell cycle regulation of DNA repair will continue to be a focus (2003).
22. Ray, J.H. & German, J. Blooms syndrome and EM9 cells in BrdU-containing medium
of investigation for a number of reasons. Firstly, the development of exhibit similarly elevated frequencies of sister chromatid exchange but dissimilar
targeted cancer therapies that modulate cell cycle progression (CDK, amounts of cellular proliferation and chromosome disruption. Chromosoma 90,
383388 (1984).
WEE1 and mitotic kinase inhibitors150) will impact the response to DNA 23. Wyatt, H.D., Sarbajna, S., Matos, J. & West, S.C. Coordinated actions of SLX1-SLX4
damage, and it may be possible to harness this regulatory intertwining and MUS81-EME1 for Holliday junction resolution in human cells. Mol. Cell 52,
234247 (2013).
for therapeutic purposes. Secondly, the suppression of DNA end resec- 24. Krasner, D.S., Daley, J.M., Sung, P. & Niu, H. Interplay between Ku and replication
tion and HR in G1 (and G0) has profound implications for therapeutic proteinA in the restriction of Exo1-mediated DNA break end resection. J.Biol. Chem.
290, 1880618816 (2015).
gene editing since it is currently impossible to undertake gene targeting 25. Symington, L.S. & Gautier, J. Double-strand break end resection and repair pathway
in non-dividing cells. Manipulating the mechanisms that suppress HR in choice. Annu. Rev. Genet. 45, 247271 (2011).
26. Takata, M. etal. Homologous recombination and non-homologous end-joining pathways
G1 cells may unlock the ability to precisely manipulate genomes in non- of DNA double-strand break repair have overlapping roles in the maintenance of
dividing cells. We also expect that, in the coming years, the regulation of chromosomal integrity in vertebrate cells. EMBO J. 17, 54975508 (1998).
27. Siede, W., Friedl, A. A., Dianova, I., Eckardt-Schupp, F. & Friedberg, E. C. The
other repair pathways by the cell cycle will be revealed, in particular how Saccharomyces cerevisiae Ku autoantigen homologue affects radiosensitivity only in
the cell cycle modulates the various types of ICL repair. the absence of homologous recombination. Genetics 142, 91102 (1996).

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 7



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEW

28. Pierce, A.J., Hu, P., Han, M., Ellis, N. & Jasin, M. Ku DNA end-binding protein 64. Duro, E. et al. Identification of the MMS22L-TONSL complex that promotes
modulates homologous repair of double-strand breaks in mammalian cells. Genes homologous recombination. Mol. Cell 40, 632644 (2010).
Dev. 15, 32373242 (2001). 65. ODonnell, L. etal. The MMS22L-TONSL complex mediates recovery from replication
29. Saintigny, Y. et al. Characterization of homologous recombination induced by stress and homologous recombination. Mol. Cell 40, 619631 (2010).
replication inhibition in mammalian cells. EMBO J. 20, 38613870 (2001). 66. Botuyan, M.V. etal. Structural basis for the methylation state-specific recognition of
30. Frank-Vaillant, M. & Marcand, S. Transient stability of DNA ends allows nonhomologous histone H4K20 by 53BP1 and Crb2 in DNA repair. Cell 127, 13611373 (2006).
end joining to precede homologous recombination. Mol. Cell 10, 11891199 (2002). 67. Fradet-Turcotte, A. etal. 53BP1 is a reader of the DNA-damage-induced H2A Lys15
31. Ira, G. et al. DNA end resection, homologous recombination and DNA damage ubiquitin mark. Nature 499, 5054 (2013).
checkpoint activation require CDK1. Nature 431, 10111017 (2004). 68. Sjogren, C. & Nasmyth, K. Sister chromatid cohesion is required for postreplicative
32. Aylon, Y., Liefshitz, B. & Kupiec, M. The CDK regulates repair of double-strand breaks double-strand break repair in Saccharomyces cerevisiae. Curr. Biol. 11, 991995
by homologous recombination during the cell cycle. EMBO J. 23, 48684875 (2004). (2001).
33. Jazayeri, A. etal. ATM- and cell cycle-dependent regulation of ATR in response to DNA 69. Sonoda, E. etal. Scc1/Rad21/Mcd1 is required for sister chromatid cohesion and
double-strand breaks. Nat. Cell Biol. 8, 3745 (2006). kinetochore function in vertebrate cells. Dev. Cell 1, 759770 (2001).
34. Huertas, P., Cortes-Ledesma, F., Sartori, A.A., Aguilera, A. & Jackson, S.P. CDK 70. Bauerschmidt, C. etal. Cohesin promotes the repair of ionizing radiation-induced
targets Sae2 to control DNA-end resection and homologous recombination. Nature DNA double-strand breaks in replicated chromatin. Nucleic Acids Res. 38, 477487
455, 689692 (2008). (2010).
35. Chen, X. etal. Enrichment of Cdk1-cyclins at DNA double-strand breaks stimulates 71. Strom, L., Lindroos, H.B., Shirahige, K. & Sjogren, C. Postreplicative recruitment of
Fun30 phosphorylation and DNA end resection. Nucleic Acids Res. 44, 27422753 cohesin to double-strand breaks is required for DNA repair. Mol. Cell 16, 10031015
(2016). (2004).
36. Ferrari, M. etal. Functional interplay between the 53BP1-ortholog Rad9 and the 72. Unal, E. etal. DNA damage response pathway uses histone modification to assemble
Mre11 complex regulates resection, end-tethering and repair of a double-strand break. a double-strand break-specific cohesin domain. Mol. Cell 16, 9911002 (2004).
PLoS Genet. 11, e1004928 (2015). 73. Kim, J. S., Krasieva, T. B., LaMorte, V., Taylor, A. M. & Yokomori, K. Specific
37. Huertas, P. & Jackson, S.P. Human CtIP mediates cell cycle control of DNA end recruitment of human cohesin to laser-induced DNA damage. J.Biol. Chem. 277,
resection and double strand break repair. J.Biol. Chem. 284, 95589565 (2009). 4514945153 (2002).
38. Makharashvili, N. & Paull, T. T. CtIP: a DNA damage response protein at the 74. Wilson, M.D. etal. The structural basis of modified nucleosome recognition by 53BP1.
intersection of DNA metabolism. DNA Repair 32, 7581 (2015). Nature 536, 100103 (2016).
39. Wang, H. etal. The interaction of CtIP and Nbs1 connects CDK and ATM to regulate 75. Zimmermann, M., Lottersberger, F., Buonomo, S.B., Sfeir, A. & de Lange, T. 53BP1
HR-mediated double-strand break repair. PLoS Genet. 9, e1003277 (2013). regulates DSB repair using Rif1 to control 5 end resection. Science 339, 700704
40. Yu, X., Wu, L.C., Bowcock, A.M., Aronheim, A. & Baer, R. The C-terminal (BRCT) (2013).
domains of BRCA1 interact invivo with CtIP, a protein implicated in the CtBP pathway 76. Di Virgilio, M. et al. Rif1 prevents resection of DNA breaks and promotes
of transcriptional repression. J.Biol. Chem. 273, 2538825392 (1998). immunoglobulin class switching. Science 339, 711715 (2013).
41. Yu, X. & Chen, J. DNA damage-induced cell cycle checkpoint control requires CtIP, a 77. Escribano-Diaz, C. etal. A cell cycle-dependent regulatory circuit composed of 53BP1-
phosphorylation-dependent binding partner of BRCA1 C-terminal domains. Mol. Cell RIF1 and BRCA1-CtIP controls DNA repair pathway choice. Mol. Cell 49, 872883
Biol. 24, 94789486 (2004). (2013).
42. Stark, J.M., Pierce, A.J., Oh, J., Pastink, A. & Jasin, M. Genetic steps of mammalian 78. Feng, L., Fong, K.W., Wang, J., Wang, W. & Chen, J. RIF1 counteracts BRCA1-mediated
homologous repair with distinct mutagenic consequences. Mol. Cell Biol. 24, end resection during DNA repair. J.Biol. Chem. 288, 1113511143 (2013).
93059316 (2004). 79. Callen, E. et al. 53BP1 Mediates productive and mutagenic DNA repair through
43. Schlegel, B.P., Jodelka, F.M. & Nunez, R. BRCA1 promotes induction of ssDNA by distinct phosphoprotein interactions. Cell 153, 12661280 (2013).
ionizing radiation. Cancer Res. 66, 51815189 (2006). 80. Munoz, I.M., Jowsey, P.A., Toth, R. & Rouse, J. Phospho-epitope binding by the BRCT
44. Cruz-Garcia, A., Lopez-Saavedra, A. & Huertas, P. BRCA1 accelerates CtIP-mediated domains of hPTIP controls multiple aspects of the cellular response to DNA damage.
DNA-end resection. Cell Rep. 9, 451459 (2014). Nucleic Acids Res. 35, 53125322 (2007).
45. Steger, M. etal. Prolyl isomerase PIN1 regulates DNA double-strand break repair by 81. Xu, G. etal. REV7 counteracts DNA double-strand break resection and affects PARP
counteracting DNA end resection. Mol. Cell 50, 333343 (2013). inhibition. Nature 521, 541544 (2015).
46. Orthwein, A. etal. A mechanism for the suppression of homologous recombination in 82. Boersma, V. etal. MAD2L2 controls DNA repair at telomeres and DNA breaks by
G1 cells. Nature 528, 422426 (2015). inhibiting 5 end resection. Nature 521, 537540 (2015).
47. Barton, O. etal. Polo-like kinase 3 regulates CtIP during DNA double-strand break 83. Bouwman, P. etal. 53BP1 loss rescues BRCA1 deficiency and is associated with triple-
repair in G1. J.Cell Biol. 206, 877894 (2014). negative and BRCA-mutated breast cancers. Nat. Struct. Mol. Biol. 17, 688695
48. Tomimatsu, N. etal. Phosphorylation of EXO1 by CDKs 1and 2 regulates DNA end (2010).
resection and repair pathway choice. Nat. Commun. 5, 3561 (2014). 84. Bunting, S.F. etal. 53BP1 inhibits homologous recombination in Brca1-deficient
49. Wohlbold, L. etal. Chemical genetics reveals a specific requirement for Cdk2 activity cells by blocking resection of DNA breaks. Cell 141, 243254 (2010).
in the DNA damage response and identifies Nbs1 as a Cdk2 substrate in human cells. 85. Cao, L. etal. A selective requirement for 53BP1 in the biological response to genomic
PLoS Genet. 8, e1002935 (2012). instability induced by Brca1 deficiency. Mol. Cell 35, 534541 (2009).
50. Falck, J. etal. CDK targeting of NBS1 promotes DNA-end resection, replication restart 86. Chapman, J.R., Sossick, A.J., Boulton, S.J. & Jackson, S.P. BRCA1-associated
and homologous recombination. EMBO Rep. 13, 561568 (2012). exclusion of 53BP1 from DNA damage sites underlies temporal control of DNA repair.
51. Ferretti, L.P., Lafranchi, L. & Sartori, A.A. Controlling DNA-end resection: a new task J.Cell Sci. 125, 35293534 (2012).
for CDKs. Front. Genet. 4, 99 (2013). 87. Densham, R.M. etal. Human BRCA1-BARD1 ubiquitin ligase activity counteracts
52. Chen, X. etal. Cell cycle regulation of DNA double-strand break end resection by Cdk1- chromatin barriers to DNA resection. Nat. Struct. Mol. Biol. 23, 647655 (2016).
dependent Dna2 phosphorylation. Nat. Struct. Mol. Biol. 18, 10151019 (2011). 88. Chapman, J.R. etal. RIF1 is essential for 53BP1-dependent nonhomologous end
53. Tkac, J. etal. HELB is a feedback inhibitor of DNA end resection. Mol. Cell 61, joining and suppression of DNA double-strand break resection. Mol. Cell 49, 858871
405418 (2016). (2013).
54. Guler, G.D. etal. Human DNA helicase B (HDHB) binds to replication protein A and 89. Jacquet, K. etal. The TIP60 complex regulates bivalent chromatin recognition by
facilitates cellular recovery from replication stress. J.Biol. Chem. 287, 64696481 53BP1 through direct H4K20me binding and H2AK15 acetylation. Mol. Cell 62,
(2012). 409421 (2016).
55. Gu, J. etal. Cell cycle-dependent regulation of a human DNA helicase that localizes 90. Tang, J. et al. Acetylation limits 53BP1 association with damaged chromatin to
in DNA damage foci. Mol. Biol. Cell 15, 33203332 (2004). promote homologous recombination. Nat. Struct. Mol. Biol. 20, 317325 (2013).
56. Spencer, S.L. etal. The proliferation-quiescence decision is controlled by a bifurcation 91. Ochs, F. etal. 53BP1 fosters fidelity of homology-directed DNA repair. Nat. Struct.
in CDK2 activity at mitotic exit. Cell 155, 369383 (2013). Mol. Biol. 23, 714721 (2016).
57. Jirawatnotai, S. etal. A function for cyclin D1 in DNA repair uncovered by protein 92. Zeman, M.K. & Cimprich, K.A. Causes and consequences of replication stress. Nat.
interactome analyses in human cancers. Nature 474, 230234 (2011). Cell Biol. 16, 29 (2014).
58. Kanakkanthara, A. etal. Cyclin A2 is an RNA binding protein that controls Mre11 93. Semlow, D.R., Zhang, J., Budzowska, M., Drohat, A.C. & Walter, J.C. Replication-
mRNA translation. Science 353, 15491552 (2016). dependent unhooking of DNA interstrand cross-links by the NEIL3 glycosylase. Cell
59. Cheong, N., Wang, X., Wang, Y. & Iliakis, G. Loss of S-phase-dependent radioresistance 167, 498511 (2016).
in irs-1 cells exposed to Xrays. Mutat. Res. 314, 7785 (1994). 94. Kottemann, M.C. & Smogorzewska, A. Fanconi anaemia and the repair of Watson and
60. Rothkamm, K., Kruger, I., Thompson, L.H. & Lobrich, M. Pathways of DNA double- Crick DNA crosslinks. Nature 493, 356363 (2013).
strand break repair during the mammalian cell cycle. Mol. Cell Biol. 23, 57065715 95. Williams, H.L., Gottesman, M.E. & Gautier, J. The differences between ICL repair
(2003). during and outside of S phase. Trends Biochem. Sci. 38, 386393 (2013).
61. Karanam, K., Kafri, R., Loewer, A. & Lahav, G. Quantitative live cell imaging reveals 96. Marteijn, J.A., Lans, H., Vermeulen, W. & Hoeijmakers, J.H. Understanding nucleotide
a gradual shift between DNA repair mechanisms and a maximal use of HR in mid S excision repair and its roles in cancer and ageing. Nat. Rev. Mol. Cell Biol. 15,
phase. Mol. Cell 47, 320329 (2012). 465481 (2014).
62. Saredi, G. etal. H4K20me0 marks post-replicative chromatin and recruits the TONSL- 97. Williams, H.L., Gottesman, M.E. & Gautier, J. Replication-independent repair of DNA
MMS22L DNA repair complex. Nature 534, 714718 (2016). interstrand crosslinks. Mol. Cell 47, 140147 (2012).
63. Piwko, W. etal. RNAi-based screening identifies the Mms22L-Nfkbil2 complex as a 98. Garcia-Rodriguez, N., Wong, R.P. & Ulrich, H.D. Functions of ubiquitin and SUMO
novel regulator of DNA replication in human cells. EMBO J. 29, 42104222 (2010). in DNA replication and replication Stress. Front. Genet. 7, 87 (2016).

8 NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEW

99. Karras, G. I. & Jentsch, S. The RAD6 DNA damage tolerance pathway operates 124. Mikhailov, A., Cole, R.W. & Rieder, C.L. DNA damage during mitosis in human cells
uncoupled from the replication fork and is functional beyond S phase. Cell 141, delays the metaphase/anaphase transition via the spindle-assembly checkpoint. Curr.
255267 (2010). Biol. 12, 17971806 (2002).
100. Daigaku, Y., Davies, A.A. & Ulrich, H.D. Ubiquitin-dependent DNA damage bypass 125. Musacchio, A. Spindle assembly checkpoint: the third decade. Phil. Trans. R.Soc. B
is separable from genome replication. Nature 465, 951955 (2010). 366, 35953604 (2011).
101. Clarke, D.J., Johnson, R.T. & Downes, C.S. TopoisomeraseII inhibition prevents 126. Rogakou, E.P., Boon, C., Redon, C. & Bonner, W.M. Megabase chromatin domains
anaphase chromatid segregation in mammalian cells independently of the generation involved in DNA double-strand breaks invivo. J.Cell Biol. 146, 905916 (1999).
of DNA strand breaks. J.Cell Sci. 105, 563569 (1993). 127. Kato, T. A., Okayasu, R. & Bedford, J. S. Comparison of the induction and
102. Oliveira, R.A., Hamilton, R.S., Pauli, A., Davis, I. & Nasmyth, K. Cohesin cleavage disappearance of DNA double strand breaks and gamma-H2AX foci after irradiation
and Cdk inhibition trigger formation of daughter nuclei. Nat. Cell Biol. 12, 185192 of chromosomes in G1-phase or in condensed metaphase cells. Mutat. Res. 639,
(2010). 108112 (2008).
103. Wechsler, T., Newman, S. & West, S.C. Aberrant chromosome morphology in human 128. Giunta, S., Belotserkovskaya, R. & Jackson, S.P. DNA damage signaling in response
cells defective for Holliday junction resolution. Nature 471, 642646 (2011). to double-strand breaks during mitosis. J.Cell Biol. 190, 197207 (2010).
104. Matos, J., Blanco, M.G., Maslen, S., Skehel, J.M. & West, S.C. Regulatory control of 129. van Vugt, M.A. etal. A mitotic phosphorylation feedback network connects Cdk1,
the resolution of DNA recombination intermediates during meiosis and mitosis. Cell Plk1, 53BP1, and Chk2 to inactivate the G(2)/M DNA damage checkpoint. PLoS Biol.
147, 158172 (2011). 8, e1000287 (2010).
105. Chan, Y.W. & West, S.C. Spatial control of the GEN1 Holliday junction resolvase 130. Gomez-Godinez, V. etal. Analysis of DNA double-strand break response and chromatin
ensures genome stability. Nat. Commun. 5, 4844 (2014). structure in mitosis using laser microirradiation. Nucleic Acids Res. 38, e202 (2010).
106. Blanco, M.G., Matos, J. & West, S.C. Dual control of Yen1 nuclease activity and 131. Mari, P.O. etal. Dynamic assembly of end-joining complexes requires interaction
cellular localization by Cdk and Cdc14 prevents genome instability. Mol. Cell 54, between Ku70/80 and XRCC4. Proc. Natl Acad. Sci. USA 103, 1859718602 (2006).
94106 (2014). 132. Britton, S., Coates, J. & Jackson, S.P. A new method for high-resolution imaging of
107. Eissler, C.L. etal. The Cdk/cDc14 module controls activation of the Yen1 holliday Ku foci to decipher mechanisms of DNA double-strand break repair. J.Cell Biol. 202,
junction resolvase to promote genome stability. Mol. Cell 54, 8093 (2014). 579595 (2013).
108. Matos, J. & West, S.C. Holliday junction resolution: regulation in space and time. 133. Nelson, G., Buhmann, M. & von Zglinicki, T. DNA damage foci in mitosis are devoid
DNA Repair 19, 176181 (2014). of 53BP1. Cell Cycle 8, 33793383 (2009).
109. Le Beau, M.M. etal. Replication of a common fragile site, FRA3B, occurs late in S 134. Orthwein, A. etal. Mitosis inhibits DNA double-strand break repair to guard against
phase and is delayed further upon induction: implications for the mechanism of fragile telomere fusions. Science 344, 189193 (2014).
site induction. Hum. Mol. Genet. 7, 755761 (1998). 135. Jullien, D., Vagnarelli, P., Earnshaw, W.C. & Adachi, Y. Kinetochore localisation of the
110. Glover, T.W., Berger, C., Coyle, J. & Echo, B. DNA polymerase alpha inhibition by DNA damage response component 53BP1 during mitosis. J.Cell Sci. 115, 7179
aphidicolin induces gaps and breaks at common fragile sites in human chromosomes. (2002).
Hum. Genet. 67, 136142 (1984). 136. Benada, J., Burdova, K., Lidak, T., von Morgen, P. & Macurek, L. Polo-like kinase 1
111. Durkin, S. G. & Glover, T. W. Chromosome fragile sites. Annu. Rev. Genet. 41, inhibits DNA damage response during mitosis. Cell Cycle 14, 219231 (2015).
169192 (2007). 137. Lee, D.H. etal. Dephosphorylation enables the recruitment of 53BP1 to double-strand
112. Ying, S. etal. MUS81 promotes common fragile site expression. Nat. Cell Biol. 15, DNA breaks. Mol. Cell 54, 512525 (2014).
10011007 (2013). 138. Hayashi, M.T., Cesare, A.J., Fitzpatrick, J.A., Lazzerini-Denchi, E. & Karlseder, J.
113. Naim, V., Wilhelm, T., Debatisse, M. & Rosselli, F. ERCC1 and MUS81-EME1 promote A telomere-dependent DNA damage checkpoint induced by prolonged mitotic arrest.
sister chromatid separation by processing late replication intermediates at common Nat. Struct. Mol. Biol. 19, 387394 (2012).
fragile sites during mitosis. Nat. Cell Biol. 15, 10081015 (2013). 139. Gomez, M. et al. PARP1 is a TRF2-associated poly(ADP-ribose)polymerase and
114.Ehmsen, K.T. & Heyer, W.D. Saccharomyces cerevisiae Mus81-Mms4 is a catalytic, protects eroded telomeres. Mol. Biol. Cell 17, 16861696 (2006).
DNA structure-selective endonuclease. Nucleic Acids Res. 36, 21822195 140. Hayashi, M.T., Cesare, A.J., Rivera, T. & Karlseder, J. Cell death during crisis is
(2008). mediated by mitotic telomere deprotection. Nature 522, 492496 (2015).
115. Fricke, W. M., Bastin-Shanower, S. A. & Brill, S. J. Substrate specificity of the 141. Peterson, S.E. etal. Cdk1 uncouples CtIP-dependent resection and Rad51 filament
Saccharomyces cerevisiae Mus81-Mms4 endonuclease. DNA Repair 4, 243251 formation during M-phase double-strand break repair. J.Cell Biol. 194, 705720
(2005). (2011).
116. Minocherhomji, S. etal. Replication stress activates DNA repair synthesis in mitosis. 142. Ayoub, N. etal. The carboxyl terminus of Brca2 links the disassembly of Rad51
Nature 528, 286290 (2015). complexes to mitotic entry. Curr. Biol. 19, 10751085 (2009).
117. Chan, K.L., North, P.S. & Hickson, I.D. BLM is required for faithful chromosome 143. Freire, R., van Vugt, M.A., Mamely, I. & Medema, R.H. Claspin: timing the cell cycle
segregation and its localization defines a class of ultrafine anaphase bridges. EMBO arrest when the genome is damaged. Cell Cycle 5, 28312834 (2006).
J. 26, 33973409 (2007). 144. Oakley, G.G. etal. RPA phosphorylation in mitosis alters DNA binding and protein-
118. Biebricher, A. et al. PICH: a DNA translocase specially adapted for processing protein interactions. Biochemistry 42, 32553264 (2003).
anaphase bridge DNA. Mol. Cell 51, 691701 (2013). 145. Anantha, R. W., Sokolova, E. & Borowiec, J. A. RPA phosphorylation facilitates
119. Hengeveld, R.C. etal. Rif1 is required for resolution of ultrafine DNA bridges in mitotic exit in response to mitotic DNA damage. Proc. Natl Acad. Sci. USA 105,
anaphase to ensure genomic stability. Dev. Cell 34, 466474 (2015). 1290312908 (2008).
120. Zirkle, R.E. & Bloom, W. Irradiation of parts of individual cells. Science 117, 487493 146. Esashi, F. etal. CDK-dependent phosphorylation of BRCA2 as a regulatory mechanism
(1953). for recombinational repair. Nature 434, 598604 (2005).
121. Rieder, C.L. & Cole, R.W. Entry into mitosis in vertebrate somatic cells is guarded by 147. Krajewska, M. etal. Forced activation of Cdk1 via wee1 inhibition impairs homologous
a chromosome damage checkpoint that reverses the cell cycle when triggered during recombination. Oncogene 32, 30013008 (2013).
early but not late prophase. J.Cell Biol. 142, 10131022 (1998). 148. Royou, A., Gagou, M.E., Karess, R. & Sullivan, W. BubR1- and Polo-coated DNA tethers
122. Terasawa, M., Shinohara, A. & Shinohara, M. Canonical non-homologous end facilitate poleward segregation of acentric chromatids. Cell 140, 235245 (2010).
joining in mitosis induces genome instability and is suppressed by Mphasespecific 149. Giunta, S. & Jackson, S.P. Give me a break, but not in mitosis: the mitotic DNA
phosphorylation of XRCC4. PLoS Genet. 10, e1004563 (2014). damage response marks DNA double-strand breaks with early signaling events. Cell
123. Metzger, L. & Iliakis, G. Kinetics of DNA double-strand break repair throughout the Cycle 10, 12151221 (2011).
cell cycle as assayed by pulsed field gel electrophoresis in CHO cells. Int. J.Radiat. 150. OConnor, M.J. Targeting the DNA damage response in cancer. Mol. Cell 60, 547560
Biol. 59, 13251339 (1991). (2015).

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 9



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

También podría gustarte