You are on page 1of 13

Molecular Cell

Review

Forging Ahead through Darkness:


PCNA, Still the Principal Conductor
at the Replication Fork
Katherine N. Choe1,2 and George-Lucian Moldovan1,3,*
1Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
2Present address: Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute,
Harvard Medical School, Boston, MA 02215, USA
3Lead Contact

*Correspondence: glm29@psu.edu
http://dx.doi.org/10.1016/j.molcel.2016.12.020

Proliferating cell nuclear antigen (PCNA) lies at the center of the faithful duplication of eukaryotic genomes.
With its distinctive doughnut-shaped molecular structure, PCNA was originally studied for its role in stimu-
lating DNA polymerases. However, we now know that PCNA does much more than promote processive
DNA synthesis. Because of the complexity of the events involved, cellular DNA replication poses major
threats to genomic integrity. Whatever predicament lies ahead for the replication fork, PCNA is there to
orchestrate the events necessary to handle it. Through its many protein interactions and various post-trans-
lational modifications, PCNA has far-reaching impacts on a myriad of cellular functions.

General Structural and Functional Characteristics PIP-box of the CDK inhibitor p21 is one of the strongest
of PCNA PCNA-interacting peptides (Bruning and Shamoo, 2004). This
Proliferating cell nuclear antigen (PCNA) is a ring-shaped homo- ability to finely tune the interaction affinity through residues other
trimer that encircles the DNA (Figure 1); hence, it is also referred to than those conserved in the consensus sequence provides the
as a sliding clamp (Boehm et al., 2016a; Dieckman et al., 2012; first level of regulation of the competition for PCNA binding.
Moldovan et al., 2007; Warbrick, 2000). PCNA has a highly Indeed, modifying the PIP-box of crucial replication and repair
conserved 6-fold symmetry because each of the monomers is proteins to increase the strength of PCNA interaction results in
composed of two similarly folded globular regions united by a impaired replication and repair (Fridman et al., 2010), indicating
flexible interdomain connecting loop (IDCL). The face of PCNA that competition through binding strength is a critical regulatory
pointing in the direction of DNA synthesis is known as the front mechanism.
face, and it is the site of interaction for most binding partners. A PCNA is an essential co-factor for DNA polymerases during
staggering number of proteins interact directly with PCNA. replication (O’Donnell et al., 2013; Siddiqui et al., 2013). PCNA
Thus, they must compete with each other for dancing with the tethers polymerases to DNA and dramatically increases their
sliding clamp (De Biasio and Blanco, 2013; Maga and Hubscher, processivity (the average number of nucleotides added before
2003; Mailand et al., 2013). Most interacting partners bind PCNA dissociation from DNA). Similar to other DNA replication factors,
through a conserved sequence termed the PIP (PCNA-interact- PCNA is essential for viability in all organisms. PCNA also partic-
ing peptide)-box, which inserts itself into a hydrophobic pocket ipates in non-replicative DNA synthesis events, such as those
on the front face, beneath the IDCLs. The consensus PIP occurring during DNA repair. Repair DNA synthesis events are
sequence is Q-x-x-J-x-x-w-w, in which J is a moderately common to a variety of DNA repair processes, including nucleo-
hydrophobic amino acid (L, V, I, or M) and w is an aromatic residue tide excision repair, homologous recombination, and mismatch
(Y or F). Many partners have degenerate sequences missing repair. But the functions of PCNA extend well beyond DNA syn-
some of the core amino acids and are still able to bind PCNA. thesis. Whereas it is devoid of enzymatic activity itself, PCNA
A reverse PIP-box is also able to support PCNA interaction (Ped- exerts a strong influence on the metabolism of DNA and
ley et al., 2014). In addition, a second PCNA-interacting motif, chromatin by recruiting various enzymes to DNA. PCNA not
termed APIM (AlkB homolog 2 PCNA-interacting motif) is wide- only participates in localizing these factors to their sites of action,
spread among DNA repair proteins and is defined as K/R-F/Y/ but in many instances, it also directly activates their enzymatic
W-L/I/V/A-L/I/V/A-K/R (Gilljam et al., 2009). The APIM interaction activities.
surface on PCNA partially overlaps with that used by the PIP-box. The repertoire of PCNA functions is dramatically amplified by
The diversity of sequence variations of PCNA-binding motifs its post-translational modifications (Hoege et al., 2002; Moldovan
(Table 1) allows for differential binding strengths. For example, et al., 2007). Because PCNA lacks enzymatic activity and instead
the specialized PIP-box Q-x-x-J-T-D-w-w provides a higher af- exerts its effects through protein-protein interactions, regulated
finity for PCNA (Havens and Walter, 2009). This indicates that addition and removal of these moieties to PCNA provides an ideal
the PIP-box motifs lacking the TD sequence are suboptimal mechanism for controlling PCNA functions. These modifications
because they provide lower binding affinity. The TD-containing generally do not affect PCNA structure. Instead, they provide an

380 Molecular Cell 65, February 2, 2017 ª 2016 Elsevier Inc.


Molecular Cell

Review
Figure 1. PCNA Structure
(A) Front view of PCNA. Each monomer is pre-
sented in a different color. Arrows indicate the
position of K164, the residue targeted by ubiq-
uitination and SUMOylation. Grey arrowhead in-
dicates the interdomain connecting loop (IDCL) on
one of the monomers.
(B) Front view of PCNA showing the interaction of
one of the monomers with a p21-derived PIP-box
peptide (in purple, marked by arrowhead).
(C) Side view. Arrows indicate K164 residue on two
of the monomers. Grey arrowhead indicates the
IDCL on one of the monomers. The front face is
also indicated.

additional surface for interaction with specific binding factors— proceeds continuously. PCNA interaction with Polε is weak,
the second level of regulating PCNA interactions (Mailand et al., and PCNA may even dissociate and remain behind the fork to
2013). This is the case even for bulky modifications, such as ubiq- mark the leading strand for post-replicative events, such as
uitination or the ubiquitin-like protein SUMO (Tsutakawa et al., mismatch repair.
2015). Structures of mono-ubiquitinated PCNA using X-ray crys- Because DNA synthesis can only occur in the 50 –30 direction,
tallography, electron microscopy, and small-angle X-ray scat- replication on the lagging strand is discontinuous. On this strand,
tering (Lau et al., 2015; Tsutakawa et al., 2015; Zhang et al., PCNA interacts with multiple PIP-box-containing subunits of
2012) revealed several different conformations, including one Pold, as well as with the Pold-associated protein PDIP46
with ubiquitin extending away from PCNA, as well as a ‘‘docked’’ (Wang et al., 2016). Primers are being synthesized every 100–
conformation, in which ubiquitin interacts extensively with the 200 nt to generate Okazaki fragments. The end of the previous
PCNA surface, forming a contiguous binding interface. It is Okazaki fragment functions as a molecular break that slows
possible that these different conformations allow ubiquitinated down Pold 10-fold. The polymerase partly displaces the primer
PCNA the necessary plasticity to interact with different binding end, forming a flap structure that is cleaved off by FEN1, 1 nt
partners. In contrast, SUMO is simply tethered to PCNA without at a time (Stodola and Burgers, 2016). Eventually, DNA ligase I
additional interactions, resulting in an extended, flexible tag, seals the resulting nick. Both enzymes interact with PCNA, which
which acts as an independent interaction module (Armstrong activates their activities (Zheng and Shen, 2011). Formation of a
et al., 2012; Tsutakawa et al., 2015). As detailed below, PCNA stable, active Okazaki fragment maturation complex in which
post-translational modifications are generally associated with different monomers of the PCNA trimer interact with Pold,
particular functional outcomes (Table 2). FEN1, and DNA ligase I, respectively (a so-called ‘‘PCNA tool
belt’’; Figure 2) was proposed many years ago and recently
DNA Replication demonstrated experimentally (Stodola and Burgers, 2016). On
DNA replication is initiated at distinct replication origins, which the other hand, mutant PCNA trimers with a single binding site
are marked by binding of the pre-replicative protein complex in are perfectly capable of supporting Okazaki fragment maturation
G1 phase of the cell cycle (O’Donnell et al., 2013; Siddiqui (Dovrat et al., 2014), arguing that tool belts may not be absolutely
et al., 2013). Origin firing during S phase involves the assembly required. Interestingly, post-translational modifications of FEN1
of two back-to-back replication forks that proceed in opposing seem to play an important role in regulating these interactions
directions. Each replication fork is headed by the Cdc45, (Zheng and Shen, 2011).
Mcm2-7, and GINS (CMG) helicase, which unwinds the DNA. Because it is loaded on the primer template junction at each
This allows synthesis of the primer by the DNA polymerase a Okazaki fragment, PCNA accumulates on the lagging strand.
complex, which contains one catalytic subunit responsible for Unloading of PCNA requires the activity of an RFC-like complex,
synthesis of the initial 10-nt RNA primer, and a separate DNA in which the catalytic subunit RFC1 is replaced by ATAD5 (Elg1 in
polymerase subunit that extends the RNA primer with about yeast; Kubota et al., 2013; Lee et al., 2013). Underlining the
20 nt. PCNA is loaded at the primer template junction by the importance of removing PCNA from DNA, loss of Elg1 unloading
multi-subunit replication factor C (RFC) complex, which can activity results in DNA repair defects and chromosome insta-
open the PCNA ring and clamp it on the DNA. Whereas the bility. Re-expression of Elg1 during G2 and M phase, but not dur-
fork assembly takes place on a DNA region devoid of nucleo- ing S phase, can restore genomic stability, indicating that PCNA
somes, PCNA loading seems to require a chromatin context retention on DNA beyond DNA replication and into G2/M is toxic
marked by methylation of histone H3 at lysine K56, which is cata- (Johnson et al., 2016). Interestingly, Elg1 is itself inhibited during
lyzed by the methyltransferase G9a (Yu et al., 2012). In vitro G1 to allow PCNA loading on DNA at the G1/S transition (Huang
reconstitution of the replisome complex assembly showed et al., 2016).
that, in the presence of PCNA-RFC, the primase is switched PCNA as Co-factor for Regulated Protein Degradation
with the high-fidelity replicative DNA polymerases ε (on the lead- during Replication
ing strand) and d (on the lagging strand; Georgescu et al., 2014, One essential activity of PCNA is to promote degradation of a
2015). On the leading strand, DNA synthesis catalyzed by Polε subset of its binding partners. A large number of substrates

Molecular Cell 65, February 2, 2017 381


Molecular Cell

Review

Table 1. PCNA Interaction Motifs replication speed. Under these conditions, PCNA is retained
on chromatin to allow CAF-1 recruitment once histones become
Interaction Motif Sequence Example
available (Mejlvang et al., 2014). Not only do nucleosomes need
PIP-box Q x x L/V/I/M x x QRSIMSFF (in DNA
to be assembled on nascent DNA, but their histone marks also
F/Y F/Y ligase I)
need to be preserved so that the parental epigenetic status is
Degenerate Missing one or two KHTLDIFF (in Polk)
maintained in the daughter cells. Parental histones ahead of
PIP-box of the core QRNHETAF replication forks are relocated to nascent DNA behind the repli-
amino acids (in MCL1)
cation fork, providing a possible mechanism for transferring
QTKVEFPE (in PDIP1) epigenetic modifications. However, there is evidence that most
Specialized PIP- Q x x L/V/I/M T D QTSITDFF (in p21) histones loaded on the nascent DNA lack modifications present
box for strong F/Y F/Y on the nucleosomes ahead of the fork, such as H3K4me3 or
interaction H3K27me3 modifications. Instead, epigenetic inheritance relies
PIP-degron Q x x L/V/I/M T D QRRVTDFFARRR on the presence of the enzymes responsible for such modifica-
F/Y F/Y x x x K/R (in CDT1) tions (histone methylation, acetylation, and ubiquitination) in
Inverted PIP-box F/Y F/Y x x L/V/I/M FFAGIVWQ (in Akt) close proximity to PCNA during replication (Petruk et al., 2012,
MxxQ 2013). It is likely that these enzymes interact with PCNA. More-
APIM K/R F/Y/W L/I/V/A NKFLARE (in over, DNMT1, an enzyme responsible for DNA methylation at
L/I/V/A K/R RAD51B) CpG islands, binds directly to PCNA (Mortusewicz et al., 2005).
The consensus residues are shown underlined. Regulation of Cell Proliferation and Apoptosis
Because of its essential role in DNA replication, regulation of
PCNA steady-state levels represents an important mechanism
have been identified for this PCNA-targeted degradation. The to control proliferation in response to extracellular conditions.
CDK inhibitors p21 and Xic1 are degraded to allow normal cell- Growth factor signaling (epidermal growth factor [EGF] through
cycle progression (Abbas et al., 2008; Kim et al., 2010). Degrada- EGF receptor [EGFR] kinase and hepatocyte growth factor-like
tion of the accessory p12 subunit of Pold alters its enzymatic protein (HGFL) through c-Abl kinase) results in phosphorylation
activity (Zhang et al., 2013). The thymine DNA glycosylase TDG of PCNA at Tyr211, which increases its chromatin association
is degraded to prevent unwanted DNA demethylation (Shibata and protects it from degradation (Wang et al., 2006; Zhao
et al., 2014; Slenn et al., 2014). Degradation of the anti-recombi- et al., 2014). Several other signaling molecules have been
nogenic FBH1 helicase regulates DNA repair (Bacquin et al., shown to regulate proliferation through controlling PCNA levels,
2013). The replication licensing factors CDC6 and CDT1 are including ERK8 (Groehler and Lannigan, 2010) and 14-3-3z (Gao
degraded to prevent re-initiation of DNA replication (re-replica- et al., 2015). In addition, p21, one of the most potent negative
tion; Arias and Walter, 2006; Clijsters and Wolthuis, 2014). regulators of proliferation, inhibits not only CDKs but also
Degradation of the histone methyltransferase Set8 prevents PCNA through a tight PIP-box-mediated association that out-
untimely chromatin compaction (Centore et al., 2010; Oda competes DNA polymerases and other interactors (Gulbis
et al., 2010). et al., 1996; Moldovan et al., 2007).
Degradation of these substrates occurs only when they are PCNA is an important modulator of apoptosis through interac-
bound to PCNA on chromatin during S phase or after DNA dam- tions with both pro-apoptotic (ING1b) and anti-apoptotic (p53,
age exposure and requires ubiquitination of the substrate by the Gadd45, MyD118, and CR6) regulators (Moldovan et al., 2007).
CRL4-CDT2 ubiquitin ligase. The PCNA interactors degraded More recently, an unexpected role of cytoplasmic PCNA in regu-
through this mechanism possess a special type of PIP-box, lating apoptosis has emerged where PCNA binds to procas-
termed PIP-degron: Q-x-x-J-T-D-w-w-x-x-x-B, where B is a pases-3, -8, -9, and -10, blocking their activation and inhibiting
basic residue (K or R; Havens and Walter, 2009). Importantly, apoptosis (Witko-Sarsat et al., 2010). This anti-apoptotic activity
this basic residue recruits the CRL4-CDT2 ubiquitin ligase but is regulated by S-nitrosylation of cytoplasmic PCNA, which
only when the substrate is bound to chromatin-loaded DNA. blocks interaction with caspase-9, thus allowing activation of
The mechanistic basis for this is still unclear. Once ubiquitinated this caspase (Yin et al., 2015). Interestingly, cytosolic PCNA
by CDT2, the substrates are removed from chromatin by the also participates in the activation of the Akt pathway, possibly
ubiquitin-targeted chaperone p97, a hexameric AAA-ATPase through its interactions with APIM-motif-containing components
complex known to remodel ubiquitin complexes, and delivered of this pathway. Indeed, inhibition of these interactions reduces
to the proteasome for degradation (Raman et al., 2011). In order Akt activation and cell growth (Olaisen et al., 2015).
to allow re-expression of the substrates in G2 phase, CDK1
phosphorylates CDT2 in late S phase, preventing its recruitment Translesion Synthesis
to chromatin-bound PCNA (Rizzardi et al., 2015). One of the major activities of PCNA is to promote tolerance of
PCNA and Chromatin Assembly DNA damage during DNA replication. DNA lesions block the pro-
Nascent DNA is rapidly coated with nucleosomes in a co-replica- gression of high-fidelity replicative DNA polymerases d and ε. To
tional process that requires binding of the histone chaperone bypass them, cells employ specialized low-fidelity polymerases
CAF-1 to PCNA at replication forks (Zhang et al., 2000). Interest- (such as Polh, Polk, and Rev1)—a process termed translesion
ingly, chromatin assembly can regulate the speed of replication synthesis (TLS) (Cipolla et al., 2016; Jansen et al., 2015). TLS is
fork progression: reducing histone pools results in decreased a double-edged sword: whereas bypass of DNA lesions allows

382 Molecular Cell 65, February 2, 2017


Molecular Cell

Review

Table 2. Post-translational Modifications of Human PCNA


Modification Target Site Enzyme Outcome at the Molecular Level Biological Function
Phosphorylation Y211 EGFR, c-Abl (kinases) Protects against PCNA Promotes cell proliferation
degradation
Inhibits MutS binding Inhibits MMR
Mono-ubiquitination K164 (and potentially RAD18, CDT2 (E3 ligases) Recruits TLS polymerases Promotes TLS
others)
K63-linked multi- K164 HLTF, SHPRH (E3 ligases) Recruits the translocase Promotes error-free
ubiquitination ZRANB3 bypass?
ISGylation K168 EFP (E3 ligase) Recruits USP10 to deubiquitinate Turns off TLS
PCNA
SUMOylation K164 UBC9 (E2-conjugating Recruits HR inhibitor PARI Inhibits HR
enzyme; no E3 ligase
necessary?)
Acetylation K13, K14, K20, K77, CBP, p300 (acetyl- Promotes removal of PCNA from Promotes genomic
K80, K284 transferases) DNA after NER stability
S-nitrosylation C81, C162 (nitrosylases?) Blocks interaction with caspase-9 Promotes apoptosis

cells to continue their proliferation program without replication Turning off TLS is an important aspect of the pathway,
arrest, low-fidelity polymerases are mutagenic on both normal because unrestrained use of low-fidelity TLS polymerases
and damaged DNA templates and thus must be kept in check. would introduce mismatches throughout the genome. PCNA
The regulation of TLS relies on post-translational modification deubiquitination by USP1 was one of the first mechanisms
of PCNA (Bienko et al., 2005; Guo et al., 2006; Hoege et al., described as a negative regulator of eukaryotic TLS (Huang
2002; Kannouche et al., 2004; Plosky et al., 2006; Watanabe et al., 2006), and since then, several other mechanisms have
et al., 2004). Upon fork stalling at DNA lesions, PCNA mono- been described. The protein SPARTAN binds ubiquitinated
ubiquitination at Lys 164 by the ubiquitin ligase RAD18 recruits PCNA at stalled forks and is important for maintaining ubiqui-
TLS polymerases to bypass the lesion (Figure 3). The molecular tin-PCNA levels through a yet-unclear mechanism (Centore
basis for this recruitment is the presence of separate PCNA-in- et al., 2012; Ghosal et al., 2012; Juhasz et al., 2012). At the
teracting motifs and ubiquitin-interacting domains in most TLS same time, SPARTAN plays a crucial role in switching off TLS
polymerases, providing higher binding affinity for the ubiquiti- by recruiting the ubiquitin-selective chaperone p97 to remove
nated form of PCNA. In contrast, replicative DNA polymerases Polh from DNA (Davis et al., 2012; Mosbech et al., 2012). The
only possess PCNA-binding motifs and lack ubiquitin-interacting small PCNA-interacting protein PAF15 also participates in
domains. Indeed, it was recently shown that PCNA ubiquitination turning off TLS by tightly associating with PCNA following lesion
does not affect the assembly or processivity of the Pold complex bypass, out-competing TLS polymerases (De Biasio et al., 2015;
(Hedglin et al., 2016). Povlsen et al., 2012).
Fine-Tuning TLS TLS is also regulated by additional PCNA modification. After
The various TLS polymerases differ in their abilities to bypass DNA damage, PCNA can be modified with the ubiquitin-like
different lesions; some of them can even ensure accurate molecule ISG15 (ISGylation) through the recruitment of the
bypass of specific lesions, such as in the case of Polh-mediated ISG15 E3 ligase EFP to ubiquitinated PCNA. This modification
bypass of UV-induced thymidine dimers. Mechanisms for spe- then recruits the deubiquitinating enzyme USP10 to remove
cific recruitment of a polymerase to a lesion are not yet known. ubiquitin from PCNA and restore high-fidelity DNA synthesis
Recent models suggest that this may be a stochastic process: (Park et al., 2014).
because PCNA is a trimer, tool belts can be formed in which Additional layers of complexity have been added in recent
each monomer binds a different polymerase, albeit with low years. The CRL4-CDT2 ubiquitin ligase can also ubiquitinate
affinity. This ensures a high local concentration of various TLS PCNA at K164 to promote TLS, but unlike RAD18, it seems to
polymerases that can individually sample the lesion and eventu- be associated with endogenous replication stress rather than
ally engage it (Boehm et al., 2016b). Such tool belts have been DNA damage exposure (Terai et al., 2010). PCNA ubiquitination
observed even in the absence of ubiquitination. Indeed, under can also be increased by a variety of factors, including the repair
certain conditions, polymerases can perform TLS in the proteins FANCD2 (Chen et al., 2016) and PTIP (Göhler et al.,
absence of PCNA ubiquitination (Acharya et al., 2010; Despras 2008); the ADP-ribosyltransferase PARP10 (Nicolae et al.,
et al., 2012; Hendel et al., 2011; Krijger et al., 2011; Wit et al., 2014); the signaling molecules SIVA1 (Han et al., 2014) and
2015). However, PCNA ubiquitination not only increases effi- MAGE-A4 (Gao et al., 2016); the ubiquitin ligases BRCA1 (Tian
ciency but also alters the mutation spectra of TLS (Hendel et al., 2013) and RNF8 (Zhang et al., 2008); the checkpoint pro-
et al., 2011). It is likely that the addition of ubiquitin provides teins CHK1 and claspin (Yang et al., 2008), and others. Whereas
an additional surface that the polymerases can attach to with it is unclear how each of these factors fit in the bigger picture,
varying affinities. they likely provide fine-tuning mechanisms, ensuring that the

Molecular Cell 65, February 2, 2017 383


Molecular Cell

Review
Figure 2. Example of PCNA Tool Belt: The
Lig1 Pol Pol Lig1 Okazaki Fragment Maturation Complex
FEN1 FEN1
Pold, FEN1, and DNA ligase I bind PCNA simul-
taneously and each interact with a monomer of the
PCNA trimer to facilitate enzyme switching during
Okazaki fragment maturation.
Lig1 Pol
FEN1

(1) DNA synthesis (2) Flap cleavage (3) Ligation

proper balance of TLS is achieved, depending on cell type, Non-canonical Roles of PCNA Ubiquitination
growth conditions and proliferation signals, cell-cycle stage, PCNA ubiquitination can be detected outside of S phase in human
chromatin status, amount of damage, etc. cells under certain conditions. For example, PCNA ubiquitination
Timing of PCNA Ubiquitination and TLS can be induced by UV in quiescent (G0-arrested) cells (Ogi et al.,
PCNA ubiquitination is normally detected during S phase in both 2010; Yang et al., 2013b). This event seems to promote recruit-
yeast and human cells (Hoege et al., 2002; Huang et al., 2006; ment of the TLS Polk to UV lesions for repair synthesis (Ogi
Terai et al., 2010). Mechanistic studies have shown that uncou- et al., 2010). Oxidative damage also induces PCNA ubiquitination
pling between the helicase machinery and the stalled polymer- outside S phase (Zlatanou et al., 2011). Whereas it is still depen-
ase results in accumulation of single-stranded DNA that recruits dent on RAD18, this ubiquitination event involves a non-canonical
RAD18 to ubiquitinate PCNA (Chang and Cimprich, 2009; Davies activity of the mismatch repair complex MSH2-MSH6, which is
et al., 2008). Chromatin remodeling by the ZBTB1-KAP1 com- required to initiate removal of the damaged strand by the
plex promotes RAD18 recruitment and PCNA ubiquitination, nuclease EXO1. In case DNA lesions are present on the remaining
perhaps by increasing RAD18 accessibility to DNA (Kim et al., strand, which may frequently be the case, PCNA ubiquitination
2014). Switching the replicative polymerase with a TLS polymer- promotes a TLS-like lesion bypass event using low-fidelity poly-
ase seems to be a straightforward and time-saving mechanism merases. Interestingly, even though this process still uses the ca-
for restarting a stalled replication fork and continuation of nonical RAD18 ubiquitin ligase, an alternative deubiquitinating
DNA replication. Thus, PCNA ubiquitination and TLS were enzyme, namely USP7, is employed to remove ubiquitin from
thought to occur co-replicationally, taking place instantaneously PCNA under these conditions (Kashiwaba et al., 2015).
following fork stalling—a process sometimes referred to as ‘‘on PCNA ubiquitination was recently shown to participate in acti-
the fly’’ (Hedglin and Benkovic, 2015; Yang et al., 2013a). vation of a novel replication stress checkpoint, which is mediated
Whereas there is evidence for PCNA-ubiquitination-mediated by ataxia telangiectasia mutated (ATM) (unlike the classic repli-
polymerase switching in vitro (Masuda et al., 2010), the process cation stress checkpoint activated by ATM and Rad3-related
can occur independent of PCNA ubiquitination in vivo (Edmunds [ATR]). Ubiquitinated PCNA recruits WRNIP protein, which in
et al., 2008). Consistent with this, genetic experiments in yeast turn binds to the ATM-activating cofactor ATMIN. This pathway
showed that PCNA ubiquitination can also occur in G2 phase is important for coordinating replication completion with cell-
without impacting damage tolerance (Daigaku et al., 2010; Kar- cycle progression, and in its absence, cells enter mitosis with
ras and Jentsch, 2010). These results instead point toward a under-replicated DNA (Kanu et al., 2016). Finally, recent genetic
post-replicational model for TLS, where the fork is re-established experiments in yeast suggest a role for PCNA ubiquitination and
downstream of the lesion, leaving behind a short unreplicated TLS in Okazaki fragment maturation, at least in the context of
region with the lesion to be repaired or bypassed at a later FEN1 inactivation (Becker et al., 2015).
time in S phase or even G2 (Figure 3). Mechanistic details of PCNA Poly-ubiquitination
such a process are missing, and many important questions The PCNA ubiquitination events described above to regulate
remain unanswered. Is the integrity of the stalled fork main- TLS are mono-ubiquitination events. However, PCNA can also
tained? Is PCNA ubiquitinated upon stalling or rather later, right be ubiquitinated by K63-linked multi-ubiquitin chains at Lys
before bypass? Why resort to TLS bypass when, behind the fork, 164. This modification is significant in yeast but present at
the lesion can be bypassed in an error-free manner, for example, much lower levels in human cells (Hoege et al., 2002). PCNA
through recombination? Nevertheless, such a post-replicative poly-ubiquitination is believed to initiate an alternative, error-
mechanism would provide the cell with additional flexibility to free lesion bypass mechanism involving the use of the newly
deal with a larger number of lesions and would perhaps provide replicated sister chromatid as template (known as template
the opportunity to sample the local environment of each individ- switching [TS]). Whereas many details of TS are still missing,
ual lesion before deciding on error-free versus error-prone recent electron microscopy studies identified structures resem-
bypass. The two possibilities may not be mutually exclusive bling double Holliday junctions as critical intermediates of
(Waters et al., 2009). Interestingly, p53 was recently shown to TS—suggesting a mechanism analogous to recombination
bind PCNA at stalled forks and suppress the extension from (Giannattasio et al., 2014). Because DNA replication is more
these forks (Hampp et al., 2016), perhaps providing a mecha- faithful in early S phase, it has been proposed that TS may be
nism for marking those particular stalled forks that will be preferentially used over TLS early in replication (Branzei and Psa-
resolved post-replicationally. khye, 2016; Stamatoyannopoulos et al., 2009).

384 Molecular Cell 65, February 2, 2017


Molecular Cell

Review
Figure 3. Two Non-exclusive Models for
Co-replicational Post-replicational the Timing of TLS
In the co-replicational model, also termed ‘‘on-
the-fly,’’ the polymerase switch occurs instanta-
1. 1. neously upon stalling, following PCNA ubiquiti-
P
Pol P
Pol nation. In the post-replicational model, the fork is
reassembled downstream of the lesion, leaving
behind a gap that is filled later in S phase or G2 by
TLS polymerases. Whether PCNA is left behind at
the gap or is loaded de novo is not clear.
ub
2. 2.
P
Pol

ases can perform DNA synthesis in the


D-loop extension step. Processivity of
ub ub Pold and Polk is enhanced by PCNA,
3. 3. TLS pol
P
Pol whereas it has no effect on the extension
by Polh. This suggests that PCNA can
affect the outcome of HR by regulating
the polymerase choice for D-loop exten-
ub sion (Sebesta et al., 2013). PCNA also
4. TLS pol 4. promotes HR at the end resection step.
PCNA loads onto DNA strands at dou-
ble-strand breaks and serves as a proc-
essivity factor for EXO1 during its end
resection activity (Chen et al., 2013).
5. PCNA can also inhibit HR through its
P
Pol
SUMO modification, which is a predomi-
nant modification in yeast. Whereas it
has been much harder to detect in
human cells, it is clear that PCNA
6. SUMOylation has a conserved role in in-
hibiting homologous recombination (Gali
et al., 2012; Hoege et al., 2002; Moldovan
et al., 2007, 2012). This activity may serve
as a means to regulate how stalled repli-
cation forks restart. HR-based mecha-
How PCNA poly-ubiquitination promotes template switching nisms can restart stalled replication forks, but this process has
is not yet understood. The ZRANB3 translocase is recruited to the potential to initiate chromosome rearrangements (Carr and
poly-ubiquitinated PCNA and remodels the DNA structure at Lambert, 2013) and likely requires prolonged replication arrest.
the stalled fork (Ciccia et al., 2012; Weston et al., 2012; Yuan In contrast, TLS, whereas locally mutagenic, would provide a
et al., 2012). Mgs1 (the yeast homolog of WRNIP1) also binds faster and more straightforward way to restart the forks.
poly-ubiquitinated PCNA and appears to interfere with Pold Because both ubiquitination and SUMOylation occur at the
functions (Saugar et al., 2012). It is still unclear how these activ- same lysine residue, K164, they would be mutually exclusive
ities promote error-free lesion bypass. Further complicating the on a PCNA monomer (though not necessarily on a trimeric
picture, the E3 ubiquitin ligases SHPRH and HLTF that are molecule). In any case, the two modifications functionally coop-
involved in PCNA poly-ubiquitination in human cells (Motegi erate to promote TLS over HR. Because PCNA SUMOylation is
et al., 2008) also participate in TLS (Lin et al., 2011). found constitutively during S phase (and not only in response
to DNA damage treatment, as is the case for ubiquitination),
Homologous Recombination SUMOylation-mediated HR suppression may also be involved
Homologous recombination (HR) repairs DNA double-strand in suppressing unwanted recombination events between newly
breaks. Following DNA end resection at the break, the result- synthesized sister chromatids during normal fork progression.
ing single-stranded DNA overhang is coated by multiple mole- In yeast, PCNA SUMOylation recruits the helicase Srs2, a
cules of the recombinase RAD51 (Prakash et al., 2015). This recombination inhibitor, which contains separate modules for in-
nucleoprotein filament then invades the sister chromatid. When teracting with PCNA (PIP-box) and SUMO (SIM motif; Armstrong
homology is found, the target strand is unwound and a displace- et al., 2012; Hoege et al., 2002; Papouli et al., 2005; Pfander et al.,
ment loop is formed. This structure is extended by DNA polymer- 2005). Srs2 inhibits recombination by removing RAD51 from sin-
ases past the break site, after which the elongated strand is gle-stranded DNA, thereby disassembling an essential recombi-
re-annealed to its original partner. The resulting gap is filled by nation intermediate structure. In human cells, SUMOylated PCNA
regular repair DNA synthesis. Both replicative and TLS polymer- recruits the recombination inhibitor PARI, a Srs2 homolog that

Molecular Cell 65, February 2, 2017 385


Molecular Cell

Review

lacks helicase activity. Similar to Srs2, PARI binds to RAD51 and more nicks are present because of discontinuous DNA syn-
has an inhibitory effect toward RAD51 nucleofilaments (Moldo- thesis. Replication errors are recognized by MutSa (MSH2-
van et al., 2012). PARI also inhibits HR at a later stage, during MSH6; identifies base mismatches and small insertion/deletion
the DNA synthesis step (Burkovics et al., 2016). Independent of loops) and MutSb (MSH2-MSH3; recognizes larger insertion/
its SUMOylation, PCNA also interacts with a number of other deletion loops) heterodimers. Once the mismatch is recognized,
recombination inhibitors. This includes RTEL1, a telomeric the MutS heterodimers form clamps that dissociate from DNA
recombination inhibitor that disrupts RAD51 post-invasion fila- and slide away. Eventually, a complex with MutLa (MLH1-
ments (D-loops; Vannier et al., 2013), and FBH1, which promotes PMS2) and PCNA is formed. PCNA activates the endonuclease
degradation of RAD51 (Chu et al., 2015). activity of MutLa on the nascent strand; the resulting nick is
further processed by the EXO1 exonuclease or through repeat
Other Genome Stability Mechanisms nickase activity by MutLa (Ganai and Johansson, 2016; Goellner
Nucleotide Excision Repair et al., 2014). Repair DNA synthesis completes the process.
Nucleotide excision repair (NER) removes helix-distorting DNA PCNA also binds MutSa, which seems to promote PCNA reten-
lesions and bulky adducts (Marteijn et al., 2014). This is achieved tion on chromatin after replication to facilitate MMR (Kawasoe
through the concerted efforts of about 30 proteins that act et al., 2016). Intriguingly, PCNA phosphorylation at Y211 by
sequentially to detect the lesion (XPC-RAD23B complex), open EGFR was shown to inhibit binding of PCNA to MutS complexes,
up the helix (TFIIH, XPB, and XPD), dually incise the damaged resulting in reduced MMR efficiency and increased mutagenesis
strand 30 nt apart (endonucleases ERCC1-XPF on the 50 side (Ortega et al., 2015). Because, as described above, this
and XPG on the 30 side), and remove the lesion-containing oligo- phosphorylation is a proliferation signal, these findings provide
nucleotide. PCNA-dependent repair DNA synthesis fills the gap another example of proliferation taking place at the expense of
to complete the reaction. PCNA also interacts, through an genome fidelity.
APIM motif, with XPA, a scaffold protein involved in organizing The Replication Stress Response
events downstream of helix opening (Gilljam et al., 2012). More- Prolonged replication fork stalling generates aberrant fork struc-
over, PCNA interacts with XPG and targets it for CDT1-mediated tures containing long stretches of single-stranded DNA. Such
degradation to allow repair DNA synthesis that bulky XPG mole- structures activate a stress response, mediated by checkpoint
cules may sterically hinder (Han et al., 2015). Finally, PCNA acti- kinases, which helps stabilize and restart the forks, thus prevent-
vates the enzymatic activity of XPF (Hutton et al., 2010). ing generation of DNA damage and genomic instability (Zeman
Following completion of NER by DNA synthesis, PCNA is acet- and Cimprich, 2014). In addition to the ubiquitinated PCNA-
ylated by CBP and p300 at lysine residues that lie in the DNA-in- dependent ATM checkpoint described above, PCNA interacts
teracting inner hole of PCNA. Acetylated PCNA is removed from with two ubiquitin ligases, TRAIP and HUWE1, and these interac-
chromatin and subjected to proteasomal degradation (Cazzalini tions are required for efficient fork progression under replication
et al., 2014). This prevents retention of PCNA on chromatin after stress conditions (Choe et al., 2016; Hoffmann et al., 2016). In
NER events, which may be damaging, as noted above. both cases, PCNA was shown to not be a substrate for these li-
Base Excision Repair gases. Instead, HUWE1 ubiquitinates the histone H2AX, a variant
Base excision repair (BER) repairs small lesions and damaged of H2A that is essential for DNA damage detection and signaling.
bases (Krokan and Bjørås, 2013). BER is initiated by the recog- Indeed, ubiquitination of H2AX by this novel PCNA-HUWE1
nition and excision of damaged bases by damage-specific pathway promotes its phosphorylation by ATR. Phosphorylated
DNA glycosylases, followed by cleavage 50 of the abasic site H2AX, also known as gH2AX, in turn recruits repair proteins to
by the AP-endonuclease 1 (APE1). PCNA interacts with and deal with broken forks (Choe et al., 2016). A relevant substrate
stimulates the activities of several glycosylases (including for TRAIP has not been identified so far, although its inactivation
UNG2, NTH1, and MPG) as well as APE1 (Moldovan et al., also reduces gH2AX formation (Harley et al., 2016). It was pro-
2007). Repair of the gap is achieved by two distinct mechanisms: posed that TRAIP may participate in removing PCNA from stalled
short-patch repair involves a 1 nt re-synthesis step by DNA po- forks under certain conditions (Feng et al., 2016). Indeed, it has
lymerase b and nick sealing by XRCC1-DNA ligase III. On the been previously shown that, upon treatment with replication in-
other hand, long-patch repair proceeds by synthesis of a stretch hibitors, such as hydroxyurea, ATR-dependent fork remodeling
of 2–20 nt by Pold in concert with PCNA and RFC, creating a flap results in active unloading of PCNA (Dungrawala et al., 2015;
structure that is engaged by the FEN-1-DNA ligase I-PCNA Yu et al., 2014). The exact role of this unloading is unclear,
complex. but it may facilitate formation of novel DNA structures important
Mismatch Repair for fork restart under these conditions. It is not known how
Mismatch repair (MMR) repairs replication-generated errors, common these events are under endogenous replication stress
such as mismatches and insertion/deletions loops. Crucial for conditions.
this process is the ability to discriminate between the template
and the nascent strands so that the nascent strand can be cor- PCNA and Human Disease
rected (Ganai and Johansson, 2016). The recognition of the Reflecting its role in promoting genomic stability, several close
nascent strand is based on the presence of nicks generated dur- companions of PCNA have been found to be associated with
ing replication, or during removal of erroneously incorporated cancer predisposition syndromes, including ATAD5 for ovarian
ribonucleotides, which have not yet been sealed. Indeed, cancer (Kuchenbaecker et al., 2015) and SPARTAN for hepato-
mismatch correction occurs faster on the lagging strand, where cellular carcinoma (Lessel et al., 2014). High PARI expression

386 Molecular Cell 65, February 2, 2017


Molecular Cell

Review

Table 3. PCNA Inhibitors in Cancer Therapy


Inhibitor Properties Mechanism of Action Effects
T2AA (Punchihewa T3 thyroid hormone Binds the PCNA cavity that Inhibits PCNA interaction with p21 (in vitro) and
et al., 2012) derivative interacts with PIP-box motifs; Pold (in cells). Treatment results in inhibition of cell
Interferes with PCNA-PIP-box growth and proliferation, increased replication
interactions stress, and reduced TLS. Combination with
Does not affect PCNA-DNA cisplatin increases cellular DNA damage and
binding reduces cell growth in U2OS cells.
PCNA-I1 (Tan et al., Small-molecule inhibitor Stabilizes the PCNA trimer Treatment reduces PCNA binding to chromatin,
2012) by binding Arg146 through an possibly through trimer stabilization. Inhibits
O-N hydrogen bond on one growth in various human and mouse cancer cell
monomer of PCNA and to Asp86 lines with a potency 9-fold higher than in
through an N-O hydrogen bond untransformed cells. Leads to cell-cycle arrest.
on its adjacent monomer
€ller et al.,
ATX-101 (Mu APIM-containing peptide Interferes with PCNA-APIM Induces apoptosis in multiple myeloma and
2013) motif interactions primary cancer cells in a caspase-dependent
manner. Potentiates the cytotoxicity of melphalan
in multiple myeloma cell lines and in a xenograft
multiple myeloma mouse model.
R9-caPeptide (Smith Cell-permeable peptide Interferes with PCNA-PIP motif Preferentially targets PCNA-protein interactions in
et al., 2015) interactions cancer cells over normal cells. Disrupts PCNA
association to chromatin and Pold, inhibiting DNA
replication in vitro and cell growth. Enhances
sensitivity to cisplatin in cancer cells. Inhibits tumor
growth in mice.
Y211F Peptide (Zhao Peptide covering the Y211 Inhibits PCNA phosphorylation Induces S phase arrest, inhibition of DNA
et al., 2011) region of PCNA at Y211 synthesis, and enhanced cell death in human
prostate cancer cell lines. Decreases tumor growth
in PC3-derived xenograft tumors in nude mice.

in tumors is associated with increased sensitivity to genotoxic (Harley et al., 2016; Shahrour et al., 2016). Similar to the PCNA
cancer therapy, presumably through its HR-inhibiting activity mutations, cells from these patients show a strong DNA repair
(Pitroda et al., 2014). Mice expressing the K164R mutant as defect but a relatively minor proliferation defect.
the only source of PCNA show altered somatic hypermutation Targeting PCNA in Cancer Therapy
at the immunoglobulin G (IgG) loci (Roa et al., 2008), suggesting Because of its essential role in promoting proliferation, PCNA is
that human B cell deficiency syndromes may also be caused by an obvious target for cancer therapy (Wang, 2014). However,
defective PCNA ubiquitination. However, until very recently, no because PCNA is not an enzyme and instead acts through its
PCNA mutation has been reported in human disease, reflecting protein interactions, it has been very difficult to design drugs
perhaps the fact that most mutations are expected to destroy that inhibit PCNA. In recent years, a number of studies identified
the complex fold of PCNA and thus result in inactivation of this small molecules that block PCNA-Pold interaction or PCNA
essential protein. trimer formation with proliferation inhibitory effects in vitro (Pun-
A recent report presented the first human syndrome caused by chihewa et al., 2012; Tan et al., 2012; Zhao et al., 2011; Table 3).
PCNA mutation (Baple et al., 2014). The patients, from a single A cell-permeable peptide containing the APIM motif, which
extended family, share a homozygous missense PCNA muta- blocks binding of partners with the APIM motif, showed prom-
tion, Ser228Ile. Ser228 lies close to the IDCL, and the mutation ising results in treating myeloma and bladder cancer in mouse
results in a large conformational change that affects interactions xenograft studies (Mu €ller et al., 2013). Similarly, a peptide span-
with PIP-box proteins, including Fen1, DNA ligase 1, and XPG ning the IDCL, which presumably inhibits PCNA interaction with
(Duffy et al., 2016). For reasons still not completely clear, the mu- PIP-box-containing partners, could inhibit cancer growth in neu-
tation affects DNA repair rather than DNA replication functions of roblastoma and breast cancer mouse xenograft models (Smith
PCNA (Green et al., 2014). This helps explain why the patients et al., 2015). These studies suggest that PCNA targeting in can-
with this mutation survive but suffer from immunodeficiency, cer may not be far away (Wang, 2014).
photosensitivity, and neurodegeneration symptoms (including
hearing loss, ataxia, cognitive decline, and other brain defects). Conclusions
Indeed, many DNA-repair-related syndromes, such as ataxia tel- Studies over the past decade have unveiled many hidden
angiectasia (ATM mutations), exhibit clinical characteristics that aspects of PCNA. Identification of new binding partners and
include neurodegeneration (Rass et al., 2007). More recently, ho- new functions, including non-canonical roles outside the
mozygous missense mutations in the PCNA-binding partners nucleus, greatly expanded the range of PCNA activities. More-
TRAIP and PARP10 have been associated with microcephaly over, it has become clear that, within the same functional

Molecular Cell 65, February 2, 2017 387


Molecular Cell

Review

pathway, PCNA can play both activating and inhibitory roles, as Baple, E.L., Chambers, H., Cross, H.E., Fawcett, H., Nakazawa, Y., Chioza,
B.A., Harlalka, G.V., Mansour, S., Sreekantan-Nair, A., Patton, M.A., et al.
detailed above for HR and MMR. The switch from activation to (2014). Hypomorphic PCNA mutation underlies a human DNA repair disorder.
inhibition is achieved, at least in these two cases, through J. Clin. Invest. 124, 3137–3146.
post-translational modifications (SUMOylation and phosphory-
Becker, J.R., Pons, C., Nguyen, H.D., Costanzo, M., Boone, C., Myers, C.L.,
lation, respectively). and Bielinsky, A.K. (2015). Genetic interactions implicating postreplicative
How the numerous PCNA interactions are regulated continues repair in Okazaki fragment processing. PLoS Genet. 11, e1005659.
to puzzle. It is likely that dynamic exchanges of the functional
Bergoglio, V., Boyer, A.S., Walsh, E., Naim, V., Legube, G., Lee, M.Y., Rey, L.,
complexes loaded on PCNA are required throughout replication. Rosselli, F., Cazaux, C., Eckert, K.A., and Hoffmann, J.S. (2013). DNA synthe-
The composition of PCNA complexes will vary from fork to fork, sis by Pol h promotes fragile site stability by preventing under-replicated DNA
in mitosis. J. Cell Biol. 201, 395–408.
depending on chromatin structure, DNA sequence, and replica-
tion timing. For example, common fragile sites in the genome act Bienko, M., Green, C.M., Crosetto, N., Rudolf, F., Zapart, G., Coull, B., Kan-
as natural fork barriers, and their replication, which generally nouche, P., Wider, G., Peter, M., Lehmann, A.R., et al. (2005). Ubiquitin-bind-
ing domains in Y-family polymerases regulate translesion synthesis. Science
takes place in late S phase, requires TLS rather than replicative 310, 1821–1824.
polymerases (Bergoglio et al., 2013). Other polymerase switch-
ing events may be required during the replication of heavily chro- Boehm, E.M., Gildenberg, M.S., and Washington, M.T. (2016a). The many
roles of PCNA in eukaryotic DNA replication. Enzymes 39, 231–254.
matinized DNA sequences and of other regions replicating late in
S phase, when mutation rates are higher (Stamatoyannopoulos Boehm, E.M., Spies, M., and Washington, M.T. (2016b). PCNA tool belts and
et al., 2009). Moreover, replication of various types of chromatin polymerase bridges form during translesion synthesis. Nucleic Acids Res. 44,
8250–8260.
may require a different set of chromatin factors bound to PCNA.
What regulates the dynamics of PCNA complexes is not clear, Branzei, D., and Psakhye, I. (2016). DNA damage tolerance. Curr. Opin. Cell
Biol. 40, 137–144.
but it is likely that post-translational modifications play some
role. Quantitative proteomic studies may be able to reveal the Bruning, J.B., and Shamoo, Y. (2004). Structural and thermodynamic analysis
composition of the various PCNA networks during replication of human PCNA with peptides derived from DNA polymerase-delta p66 sub-
unit and flap endonuclease-1. Structure 12, 2209–2219.
and repair. Next-generation sequencing approaches may be
used to investigate the impact of these complexes on replication Burkovics, P., Dome, L., Juhasz, S., Altmannova, V., Sebesta, M., Pacesa, M.,
fidelity. Finally, molecular mechanistic studies are needed to Fugger, K., Sorensen, C.S., Lee, M.Y., Haracska, L., and Krejci, L. (2016). The
PCNA-associated protein PARI negatively regulates homologous recombina-
shed light on the more enigmatic PCNA-dependent processes, tion via the inhibition of DNA repair synthesis. Nucleic Acids Res. 44,
including template switching, and strand discrimination during 3176–3189.
MMR. Without a doubt, secret facets of PCNA are waiting to
Carr, A.M., and Lambert, S. (2013). Replication stress-induced genome insta-
be discovered. bility: the dark side of replication maintenance by homologous recombination.
J. Mol. Biol. 425, 4733–4744.

AUTHOR CONTRIBUTIONS Cazzalini, O., Sommatis, S., Tillhon, M., Dutto, I., Bachi, A., Rapp, A., Nardo, T.,
Scovassi, A.I., Necchi, D., Cardoso, M.C., et al. (2014). CBP and p300 acety-
K.N.C. and G.L.M. wrote the manuscript. late PCNA to link its degradation with nucleotide excision repair synthesis.
Nucleic Acids Res. 42, 8433–8448.

ACKNOWLEDGMENTS Centore, R.C., Havens, C.G., Manning, A.L., Li, J.M., Flynn, R.L., Tse, A., Jin,
J., Dyson, N.J., Walter, J.C., and Zou, L. (2010). CRL4(Cdt2)-mediated
destruction of the histone methyltransferase Set8 prevents premature chro-
We would like to thank Michael O’Connor, Mark Hedglin, Tanay Thakar, Kris- matin compaction in S phase. Mol. Cell 40, 22–33.
ten Clements, and Claudia Nicolae for comments on the manuscript. G.-L.M. is
supported by NIH 1R01ES026184, Department of Defense CA140303, and the Centore, R.C., Yazinski, S.A., Tse, A., and Zou, L. (2012). Spartan/C1orf124, a
St. Baldrick’s Foundation. This paper is dedicated to the memory of Prof. Ste- reader of PCNA ubiquitylation and a regulator of UV-induced DNA damage
fan Jentsch, irreplaceable maestro of cellular biology. response. Mol. Cell 46, 625–635.

Chang, D.J., and Cimprich, K.A. (2009). DNA damage tolerance: when it’s OK
REFERENCES to make mistakes. Nat. Chem. Biol. 5, 82–90.

Abbas, T., Sivaprasad, U., Terai, K., Amador, V., Pagano, M., and Dutta, A. Chen, X., Paudyal, S.C., Chin, R.I., and You, Z. (2013). PCNA promotes proc-
(2008). PCNA-dependent regulation of p21 ubiquitylation and degradation essive DNA end resection by Exo1. Nucleic Acids Res. 41, 9325–9338.
via the CRL4Cdt2 ubiquitin ligase complex. Genes Dev. 22, 2496–2506.
Chen, X., Bosques, L., Sung, P., and Kupfer, G.M. (2016). A novel role for non-
Acharya, N., Yoon, J.H., Hurwitz, J., Prakash, L., and Prakash, S. (2010). ubiquitinated FANCD2 in response to hydroxyurea-induced DNA damage.
DNA polymerase eta lacking the ubiquitin-binding domain promotes replica- Oncogene 35, 22–34.
tive lesion bypass in humans cells. Proc. Natl. Acad. Sci. USA 107,
10401–10405. Choe, K.N., Nicolae, C.M., Constantin, D., Imamura Kawasawa, Y., Delgado-
Diaz, M.R., De, S., Freire, R., Smits, V.A., and Moldovan, G.L. (2016). HUWE1
Arias, E.E., and Walter, J.C. (2006). PCNA functions as a molecular platform to interacts with PCNA to alleviate replication stress. EMBO Rep. 17, 874–886.
trigger Cdt1 destruction and prevent re-replication. Nat. Cell Biol. 8, 84–90.
Chu, W.K., Payne, M.J., Beli, P., Hanada, K., Choudhary, C., and Hickson, I.D.
Armstrong, A.A., Mohideen, F., and Lima, C.D. (2012). Recognition of SUMO- (2015). FBH1 influences DNA replication fork stability and homologous recom-
modified PCNA requires tandem receptor motifs in Srs2. Nature 483, 59–63. bination through ubiquitylation of RAD51. Nat. Commun. 6, 5931.

Bacquin, A., Pouvelle, C., Siaud, N., Perderiset, M., Salomé-Desnoulez, S., Ciccia, A., Nimonkar, A.V., Hu, Y., Hajdu, I., Achar, Y.J., Izhar, L., Petit, S.A.,
Tellier-Lebegue, C., Lopez, B., Charbonnier, J.B., and Kannouche, P.L. Adamson, B., Yoon, J.C., Kowalczykowski, S.C., et al. (2012). Polyubiquiti-
(2013). The helicase FBH1 is tightly regulated by PCNA via CRL4(Cdt2)-medi- nated PCNA recruits the ZRANB3 translocase to maintain genomic integrity
ated proteolysis in human cells. Nucleic Acids Res. 41, 6501–6513. after replication stress. Mol. Cell 47, 396–409.

388 Molecular Cell 65, February 2, 2017


Molecular Cell

Review
Cipolla, L., Maffia, A., Bertoletti, F., and Sabbioneda, S. (2016). The regulation Georgescu, R.E., Schauer, G.D., Yao, N.Y., Langston, L.D., Yurieva, O.,
of DNA damage tolerance by ubiquitin and ubiquitin-like modifiers. Front. Zhang, D., Finkelstein, J., and O’Donnell, M.E. (2015). Reconstitution of a eu-
Genet. 7, 105. karyotic replisome reveals suppression mechanisms that define leading/lag-
ging strand operation. eLife 4, e04988.
Clijsters, L., and Wolthuis, R. (2014). PIP-box-mediated degradation prohibits
re-accumulation of Cdc6 during S phase. J. Cell Sci. 127, 1336–1345. Ghosal, G., Leung, J.W., Nair, B.C., Fong, K.W., and Chen, J. (2012). Prolifer-
ating cell nuclear antigen (PCNA)-binding protein C1orf124 is a regulator of
Daigaku, Y., Davies, A.A., and Ulrich, H.D. (2010). Ubiquitin-dependent translesion synthesis. J. Biol. Chem. 287, 34225–34233.
DNA damage bypass is separable from genome replication. Nature 465,
951–955. Giannattasio, M., Zwicky, K., Follonier, C., Foiani, M., Lopes, M., and Branzei,
D. (2014). Visualization of recombination-mediated damage bypass by tem-
Davies, A.A., Huttner, D., Daigaku, Y., Chen, S., and Ulrich, H.D. (2008). Acti- plate switching. Nat. Struct. Mol. Biol. 21, 884–892.
vation of ubiquitin-dependent DNA damage bypass is mediated by replication
protein a. Mol. Cell 29, 625–636. Gilljam, K.M., Feyzi, E., Aas, P.A., Sousa, M.M., Mu€ller, R., Vågbø, C.B., Cat-
terall, T.C., Liabakk, N.B., Slupphaug, G., Drabløs, F., et al. (2009). Identifica-
€thke, I., and Rouse,
Davis, E.J., Lachaud, C., Appleton, P., Macartney, T.J., Na tion of a novel, widespread, and functionally important PCNA-binding motif.
J. (2012). DVC1 (C1orf124) recruits the p97 protein segregase to sites of DNA J. Cell Biol. 186, 645–654.
damage. Nat. Struct. Mol. Biol. 19, 1093–1100.
Gilljam, K.M., Mu €ller, R., Liabakk, N.B., and Otterlei, M. (2012). Nucleotide
De Biasio, A., and Blanco, F.J. (2013). Proliferating cell nuclear antigen struc- excision repair is associated with the replisome and its efficiency depends
ture and interactions: too many partners for one dancer? Adv. Protein Chem. on a direct interaction between XPA and PCNA. PLoS ONE 7, e49199.
Struct. Biol. 91, 1–36.
Goellner, E.M., Smith, C.E., Campbell, C.S., Hombauer, H., Desai, A., Putnam,
De Biasio, A., de Opakua, A.I., Mortuza, G.B., Molina, R., Cordeiro, T.N., Cas- C.D., and Kolodner, R.D. (2014). PCNA and Msh2-Msh6 activate an Mlh1-
tillo, F., Villate, M., Merino, N., Delgado, S., Gil-Cartón, D., et al. (2015). Struc- Pms1 endonuclease pathway required for Exo1-independent mismatch repair.
ture of p15(PAF)-PCNA complex and implications for clamp sliding during DNA Mol. Cell 55, 291–304.
replication and repair. Nat. Commun. 6, 6439.
Göhler, T., Munoz, I.M., Rouse, J., and Blow, J.J. (2008). PTIP/Swift is required
Despras, E., Delrieu, N., Garandeau, C., Ahmed-Seghir, S., and Kannouche, for efficient PCNA ubiquitination in response to DNA damage. DNA Repair
P.L. (2012). Regulation of the specialized DNA polymerase eta: revisiting the (Amst.) 7, 775–787.
biological relevance of its PCNA- and ubiquitin-binding motifs. Environ. Mol.
Mutagen. 53, 752–765. Green, C.M., Baple, E.L., and Crosby, A.H. (2014). PCNA mutation affects DNA
repair not replication. Cell Cycle 13, 3157–3158.
Dieckman, L.M., Freudenthal, B.D., and Washington, M.T. (2012). PCNA struc-
ture and function: insights from structures of PCNA complexes and post-trans- Groehler, A.L., and Lannigan, D.A. (2010). A chromatin-bound kinase, ERK8,
lationally modified PCNA. Subcell. Biochem. 62, 281–299. protects genomic integrity by inhibiting HDM2-mediated degradation of the
DNA clamp PCNA. J. Cell Biol. 190, 575–586.
Dovrat, D., Stodola, J.L., Burgers, P.M., and Aharoni, A. (2014). Sequential
switching of binding partners on PCNA during in vitro Okazaki fragment matu- Gulbis, J.M., Kelman, Z., Hurwitz, J., O’Donnell, M., and Kuriyan, J. (1996).
ration. Proc. Natl. Acad. Sci. USA 111, 14118–14123. Structure of the C-terminal region of p21(WAF1/CIP1) complexed with human
PCNA. Cell 87, 297–306.
Duffy, C.M., Hilbert, B.J., and Kelch, B.A. (2016). A disease-causing variant in
PCNA disrupts a promiscuous protein binding site. J. Mol. Biol. 428, Guo, C., Tang, T.S., Bienko, M., Parker, J.L., Bielen, A.B., Sonoda, E., Takeda,
1023–1040. S., Ulrich, H.D., Dikic, I., and Friedberg, E.C. (2006). Ubiquitin-binding motifs in
REV1 protein are required for its role in the tolerance of DNA damage. Mol.
Dungrawala, H., Rose, K.L., Bhat, K.P., Mohni, K.N., Glick, G.G., Couch, F.B., Cell. Biol. 26, 8892–8900.
and Cortez, D. (2015). The replication checkpoint prevents two types of fork
collapse without regulating replisome stability. Mol. Cell 59, 998–1010. Hampp, S., Kiessling, T., Buechle, K., Mansilla, S.F., Thomale, J., Rall, M., Ahn,
€ller, L. (2016). DNA damage toler-
J., Pospiech, H., Gottifredi, V., and Wiesmu
Edmunds, C.E., Simpson, L.J., and Sale, J.E. (2008). PCNA ubiquitination and ance pathway involving DNA polymerase i and the tumor suppressor p53 reg-
REV1 define temporally distinct mechanisms for controlling translesion syn- ulates DNA replication fork progression. Proc. Natl. Acad. Sci. USA 113,
thesis in the avian cell line DT40. Mol. Cell 30, 519–529. E4311–E4319.

Feng, W., Guo, Y., Huang, J., Deng, Y., Zang, J., and Huen, M.S. (2016). TRAIP Han, J., Liu, T., Huen, M.S., Hu, L., Chen, Z., and Huang, J. (2014). SIVA1 di-
regulates replication fork recovery and progression via PCNA. Cell Discov. rects the E3 ubiquitin ligase RAD18 for PCNA monoubiquitination. J. Cell Biol.
2, 16016. 205, 811–827.

Fridman, Y., Palgi, N., Dovrat, D., Ben-Aroya, S., Hieter, P., and Aharoni, A. Han, C., Wani, G., Zhao, R., Qian, J., Sharma, N., He, J., Zhu, Q., Wang, Q.E.,
(2010). Subtle alterations in PCNA-partner interactions severely impair DNA and Wani, A.A. (2015). Cdt2-mediated XPG degradation promotes gap-filling
replication and repair. PLoS Biol. 8, e1000507. DNA synthesis in nucleotide excision repair. Cell Cycle 14, 1103–1115.

Gali, H., Juhasz, S., Morocz, M., Hajdu, I., Fatyol, K., Szukacsov, V., Burkovics, Harley, M.E., Murina, O., Leitch, A., Higgs, M.R., Bicknell, L.S., Yigit, G., Black-
P., and Haracska, L. (2012). Role of SUMO modification of human PCNA at ford, A.N., Zlatanou, A., Mackenzie, K.J., Reddy, K., et al. (2016). TRAIP pro-
stalled replication fork. Nucleic Acids Res. 40, 6049–6059. motes DNA damage response during genome replication and is mutated in
primordial dwarfism. Nat. Genet. 48, 36–43.
Ganai, R.A., and Johansson, E. (2016). DNA replication-a matter of fidelity.
Mol. Cell 62, 745–755. Havens, C.G., and Walter, J.C. (2009). Docking of a specialized PIP Box onto
chromatin-bound PCNA creates a degron for the ubiquitin ligase CRL4Cdt2.
Gao, X., Dan, S., Xie, Y., Qin, H., Tang, D., Liu, X., He, Q.Y., and Liu, L. (2015). Mol. Cell 35, 93–104.
14-3-3z reduces DNA damage by interacting with and stabilizing proliferating
cell nuclear antigen. J. Cell. Biochem. 116, 158–169. Hedglin, M., and Benkovic, S.J. (2015). Regulation of Rad6/Rad18 activity dur-
ing DNA damage tolerance. Annu. Rev. Biophys. 44, 207–228.
Gao, Y., Mutter-Rottmayer, E., Greenwalt, A.M., Goldfarb, D., Yan, F., Yang,
Y., Martinez-Chacin, R.C., Pearce, K.H., Tateishi, S., Major, M.B., and Vaziri, Hedglin, M., Pandey, B., and Benkovic, S.J. (2016). Stability of the human po-
C. (2016). A neomorphic cancer cell-specific role of MAGE-A4 in trans-lesion lymerase d holoenzyme and its implications in lagging strand DNA synthesis.
synthesis. Nat. Commun. 7, 12105. Proc. Natl. Acad. Sci. USA 113, E1777–E1786.

Georgescu, R.E., Langston, L., Yao, N.Y., Yurieva, O., Zhang, D., Finkelstein, Hendel, A., Krijger, P.H., Diamant, N., Goren, Z., Langerak, P., Kim, J., Reiss-
J., Agarwal, T., and O’Donnell, M.E. (2014). Mechanism of asymmetric poly- ner, T., Lee, K.Y., Geacintov, N.E., Carell, T., et al. (2011). PCNA ubiquitination
merase assembly at the eukaryotic replication fork. Nat. Struct. Mol. Biol. is important, but not essential for translesion DNA synthesis in mammalian
21, 664–670. cells. PLoS Genet. 7, e1002262.

Molecular Cell 65, February 2, 2017 389


Molecular Cell

Review
Hoege, C., Pfander, B., Moldovan, G.L., Pyrowolakis, G., and Jentsch, S. tigators; Australian Cancer Study (Ovarian Cancer Investigators); Australian
(2002). RAD6-dependent DNA repair is linked to modification of PCNA by ubiq- Ovarian Cancer Study Group; Consortium of Investigators of Modifiers of
uitin and SUMO. Nature 419, 135–141. BRCA1 and BRCA2 (2015). Identification of six new susceptibility loci for inva-
sive epithelial ovarian cancer. Nat. Genet. 47, 164–171.
Hoffmann, S., Smedegaard, S., Nakamura, K., Mortuza, G.B., Ra €schle, M.,
Ibañez de Opakua, A., Oka, Y., Feng, Y., Blanco, F.J., Mann, M., et al. Lau, W.C., Li, Y., Zhang, Q., and Huen, M.S. (2015). Molecular architecture of
(2016). TRAIP is a PCNA-binding ubiquitin ligase that protects genome stability the Ub-PCNA/Pol h complex bound to DNA. Sci. Rep. 5, 15759.
after replication stress. J. Cell Biol. 212, 63–75.
Lee, K.Y., Fu, H., Aladjem, M.I., and Myung, K. (2013). ATAD5 regulates the
Huang, T.T., Nijman, S.M., Mirchandani, K.D., Galardy, P.J., Cohn, M.A., Haas, lifespan of DNA replication factories by modulating PCNA level on the chro-
W., Gygi, S.P., Ploegh, H.L., Bernards, R., and D’Andrea, A.D. (2006). Regula- matin. J. Cell Biol. 200, 31–44.
tion of monoubiquitinated PCNA by DUB autocleavage. Nat. Cell Biol. 8,
339–347. Lessel, D., Vaz, B., Halder, S., Lockhart, P.J., Marinovic-Terzic, I., Lopez-Mos-
queda, J., Philipp, M., Sim, J.C., Smith, K.R., Oehler, J., et al. (2014). Mutations
Huang, F., Saraf, A., Florens, L., Kusch, T., Swanson, S.K., Szerszen, L.T., Li, in SPRTN cause early onset hepatocellular carcinoma, genomic instability and
G., Dutta, A., Washburn, M.P., Abmayr, S.M., and Workman, J.L. (2016). The progeroid features. Nat. Genet. 46, 1239–1244.
Enok acetyltransferase complex interacts with Elg1 and negatively regulates
PCNA unloading to promote the G1/S transition. Genes Dev. 30, 1198–1210. Lin, J.R., Zeman, M.K., Chen, J.Y., Yee, M.C., and Cimprich, K.A. (2011).
SHPRH and HLTF act in a damage-specific manner to coordinate different
Hutton, R.D., Craggs, T.D., White, M.F., and Penedo, J.C. (2010). PCNA and forms of postreplication repair and prevent mutagenesis. Mol. Cell 42,
XPF cooperate to distort DNA substrates. Nucleic Acids Res. 38, 1664–1675. 237–249.

Jansen, J.G., Tsaalbi-Shtylik, A., and de Wind, N. (2015). Roles of mutagenic Maga, G., and Hubscher, U. (2003). Proliferating cell nuclear antigen (PCNA):
translesion synthesis in mammalian genome stability, health and disease. a dancer with many partners. J. Cell Sci. 116, 3051–3060.
DNA Repair (Amst.) 29, 56–64.
Mailand, N., Gibbs-Seymour, I., and Bekker-Jensen, S. (2013). Regulation of
Johnson, C., Gali, V.K., Takahashi, T.S., and Kubota, T. (2016). PCNA retention PCNA-protein interactions for genome stability. Nat. Rev. Mol. Cell Biol. 14,
on DNA into G2/M phase causes genome instability in cells lacking Elg1. Cell 269–282.
Rep. 16, 684–695.
Marteijn, J.A., Lans, H., Vermeulen, W., and Hoeijmakers, J.H. (2014). Under-
Juhasz, S., Balogh, D., Hajdu, I., Burkovics, P., Villamil, M.A., Zhuang, Z., and standing nucleotide excision repair and its roles in cancer and ageing. Nat.
Haracska, L. (2012). Characterization of human Spartan/C1orf124, an ubiqui- Rev. Mol. Cell Biol. 15, 465–481.
tin-PCNA interacting regulator of DNA damage tolerance. Nucleic Acids Res.
40, 10795–10808. Masuda, Y., Piao, J., and Kamiya, K. (2010). DNA replication-coupled PCNA
mono-ubiquitination and polymerase switching in a human in vitro system.
Kannouche, P.L., Wing, J., and Lehmann, A.R. (2004). Interaction of human J. Mol. Biol. 396, 487–500.
DNA polymerase eta with monoubiquitinated PCNA: a possible mechanism
for the polymerase switch in response to DNA damage. Mol. Cell 14, 491–500. Mejlvang, J., Feng, Y., Alabert, C., Neelsen, K.J., Jasencakova, Z., Zhao, X.,
Lees, M., Sandelin, A., Pasero, P., Lopes, M., and Groth, A. (2014). New his-
Kanu, N., Zhang, T., Burrell, R.A., Chakraborty, A., Cronshaw, J., DaCosta, C., tone supply regulates replication fork speed and PCNA unloading. J. Cell
Grönroos, E., Pemberton, H.N., Anderton, E., Gonzalez, L., et al. (2016). Biol. 204, 29–43.
RAD18, WRNIP1 and ATMIN promote ATM signalling in response to replica-
tion stress. Oncogene 35, 4009–4019. Moldovan, G.L., Pfander, B., and Jentsch, S. (2007). PCNA, the maestro of the
replication fork. Cell 129, 665–679.
Karras, G.I., and Jentsch, S. (2010). The RAD6 DNA damage tolerance
pathway operates uncoupled from the replication fork and is functional beyond Moldovan, G.L., Dejsuphong, D., Petalcorin, M.I., Hofmann, K., Takeda, S.,
S phase. Cell 141, 255–267. Boulton, S.J., and D’Andrea, A.D. (2012). Inhibition of homologous recombina-
tion by the PCNA-interacting protein PARI. Mol. Cell 45, 75–86.
Kashiwaba, S., Kanao, R., Masuda, Y., Kusumoto-Matsuo, R., Hanaoka, F.,
and Masutani, C. (2015). USP7 is a suppressor of PCNA ubiquitination Mortusewicz, O., Schermelleh, L., Walter, J., Cardoso, M.C., and Leonhardt,
and oxidative-stress-induced mutagenesis in human cells. Cell Rep. 13, H. (2005). Recruitment of DNA methyltransferase I to DNA repair sites. Proc.
2072–2080. Natl. Acad. Sci. USA 102, 8905–8909.

Kawasoe, Y., Tsurimoto, T., Nakagawa, T., Masukata, H., and Takahashi, T.S. Mosbech, A., Gibbs-Seymour, I., Kagias, K., Thorslund, T., Beli, P., Povlsen,
(2016). MutSa maintains the mismatch repair capability by inhibiting PCNA L., Nielsen, S.V., Smedegaard, S., Sedgwick, G., Lukas, C., et al. (2012).
unloading. eLife 5, e15155. DVC1 (C1orf124) is a DNA damage-targeting p97 adaptor that promotes ubiq-
uitin-dependent responses to replication blocks. Nat. Struct. Mol. Biol. 19,
Kim, D.H., Budhavarapu, V.N., Herrera, C.R., Nam, H.W., Kim, Y.S., and Yew, 1084–1092.
P.R. (2010). The CRL4Cdt2 ubiquitin ligase mediates the proteolysis of cyclin-
dependent kinase inhibitor Xic1 through a direct association with PCNA. Mol. Motegi, A., Liaw, H.J., Lee, K.Y., Roest, H.P., Maas, A., Wu, X., Moinova, H.,
Cell. Biol. 30, 4120–4133. Markowitz, S.D., Ding, H., Hoeijmakers, J.H., and Myung, K. (2008). Poly-
ubiquitination of proliferating cell nuclear antigen by HLTF and SHPRH pre-
Kim, H., Dejsuphong, D., Adelmant, G., Ceccaldi, R., Yang, K., Marto, J.A., and vents genomic instability from stalled replication forks. Proc. Natl. Acad. Sci.
D’Andrea, A.D. (2014). Transcriptional repressor ZBTB1 promotes chromatin USA 105, 12411–12416.
remodeling and translesion DNA synthesis. Mol. Cell 54, 107–118.
Mu€ller, R., Misund, K., Holien, T., Bachke, S., Gilljam, K.M., Våtsveen, T.K., Rø,
Krijger, P.H., van den Berk, P.C., Wit, N., Langerak, P., Jansen, J.G., Reynaud, T.B., Bellacchio, E., Sundan, A., and Otterlei, M. (2013). Targeting proliferating
C.A., de Wind, N., and Jacobs, H. (2011). PCNA ubiquitination-independent cell nuclear antigen and its protein interactions induces apoptosis in multiple
activation of polymerase h during somatic hypermutation and DNA damage myeloma cells. PLoS ONE 8, e70430.
tolerance. DNA Repair (Amst.) 10, 1051–1059.
Nicolae, C.M., Aho, E.R., Vlahos, A.H., Choe, K.N., De, S., Karras, G.I., and
Krokan, H.E., and Bjørås, M. (2013). Base excision repair. Cold Spring Harb. Moldovan, G.L. (2014). The ADP-ribosyltransferase PARP10/ARTD10 inter-
Perspect. Biol. 5, a012583. acts with proliferating cell nuclear antigen (PCNA) and is required for DNA
damage tolerance. J. Biol. Chem. 289, 13627–13637.
Kubota, T., Nishimura, K., Kanemaki, M.T., and Donaldson, A.D. (2013). The
Elg1 replication factor C-like complex functions in PCNA unloading during O’Donnell, M., Langston, L., and Stillman, B. (2013). Principles and concepts
DNA replication. Mol. Cell 50, 273–280. of DNA replication in bacteria, archaea, and eukarya. Cold Spring Harb. Per-
spect. Biol. 5, a010108.
Kuchenbaecker, K.B., Ramus, S.J., Tyrer, J., Lee, A., Shen, H.C., Beesley, J.,
Lawrenson, K., McGuffog, L., Healey, S., Lee, J.M., et al.; EMBRACE; GEMO €bner, M.R., Beck, D.B., Vermeulen, M., Hurwitz, J., Spector, D.L.,
Oda, H., Hu
Study Collaborators; Breast Cancer Family Registry; HEBON; KConFab Inves- and Reinberg, D. (2010). Regulation of the histone H4 monomethylase PR-

390 Molecular Cell 65, February 2, 2017


Molecular Cell

Review
Set7 by CRL4(Cdt2)-mediated PCNA-dependent degradation during DNA and is required for meiotic progression. Proc. Natl. Acad. Sci. USA 105,
damage. Mol. Cell 40, 364–376. 16248–16253.

Ogi, T., Limsirichaikul, S., Overmeer, R.M., Volker, M., Takenaka, K., Cloney, Saugar, I., Parker, J.L., Zhao, S., and Ulrich, H.D. (2012). The genome mainte-
R., Nakazawa, Y., Niimi, A., Miki, Y., Jaspers, N.G., et al. (2010). Three DNA nance factor Mgs1 is targeted to sites of replication stress by ubiquitylated
polymerases, recruited by different mechanisms, carry out NER repair synthe- PCNA. Nucleic Acids Res. 40, 245–257.
sis in human cells. Mol. Cell 37, 714–727.
Sebesta, M., Burkovics, P., Juhasz, S., Zhang, S., Szabo, J.E., Lee, M.Y., Har-
€ller, R., Nedal, A., and Otterlei, M. (2015). PCNA-interacting
Olaisen, C., Mu acska, L., and Krejci, L. (2013). Role of PCNA and TLS polymerases in D-loop
peptides reduce Akt phosphorylation and TLR-mediated cytokine secretion extension during homologous recombination in humans. DNA Repair (Amst.)
suggesting a role of PCNA in cellular signaling. Cell. Signal. 27, 1478–1487. 12, 691–698.

Ortega, J., Li, J.Y., Lee, S., Tong, D., Gu, L., and Li, G.M. (2015). Phosphory- Shahrour, M.A., Nicolae, C.M., Edvardson, S., Ashhab, M., Galvan, A.M., Con-
lation of PCNA by EGFR inhibits mismatch repair and promotes misincorpora- stantin, D., Abu-Libdeh, B., Moldovan, G.L., and Elpeleg, O. (2016). PARP10
tion during DNA synthesis. Proc. Natl. Acad. Sci. USA 112, 5667–5672. deficiency manifests by severe developmental delay and DNA repair defect.
Neurogenetics 17, 227–232.
Papouli, E., Chen, S., Davies, A.A., Huttner, D., Krejci, L., Sung, P., and Ulrich,
H.D. (2005). Crosstalk between SUMO and ubiquitin on PCNA is mediated by Shibata, E., Dar, A., and Dutta, A. (2014). CRL4Cdt2 E3 ubiquitin ligase and
recruitment of the helicase Srs2p. Mol. Cell 19, 123–133. proliferating cell nuclear antigen (PCNA) cooperate to degrade thymine DNA
glycosylase in S phase. J. Biol. Chem. 289, 23056–23064.
Park, J.M., Yang, S.W., Yu, K.R., Ka, S.H., Lee, S.W., Seol, J.H., Jeon, Y.J.,
and Chung, C.H. (2014). Modification of PCNA by ISG15 plays a crucial role Siddiqui, K., On, K.F., and Diffley, J.F. (2013). Regulating DNA replication in eu-
in termination of error-prone translesion DNA synthesis. Mol. Cell 54, 626–638. karya. Cold Spring Harb. Perspect. Biol. 5, a012930.

Pedley, A.M., Lill, M.A., and Davisson, V.J. (2014). Flexibility of PCNA-protein Slenn, T.J., Morris, B., Havens, C.G., Freeman, R.M., Jr., Takahashi, T.S., and
interface accommodates differential binding partners. PLoS ONE 9, e102481. Walter, J.C. (2014). Thymine DNA glycosylase is a CRL4Cdt2 substrate. J. Biol.
Chem. 289, 23043–23055.
Petruk, S., Sedkov, Y., Johnston, D.M., Hodgson, J.W., Black, K.L., Kover-
mann, S.K., Beck, S., Canaani, E., Brock, H.W., and Mazo, A. (2012). TrxG Smith, S.J., Gu, L., Phipps, E.A., Dobrolecki, L.E., Mabrey, K.S., Gulley, P., Dil-
and PcG proteins but not methylated histones remain associated with DNA lehay, K.L., Dong, Z., Fields, G.B., Chen, Y.R., et al. (2015). A peptide
through replication. Cell 150, 922–933. mimicking a region in proliferating cell nuclear antigen specific to key protein
interactions is cytotoxic to breast cancer. Mol. Pharmacol. 87, 263–276.
Petruk, S., Black, K.L., Kovermann, S.K., Brock, H.W., and Mazo, A. (2013).
Stepwise histone modifications are mediated by multiple enzymes that rapidly Stamatoyannopoulos, J.A., Adzhubei, I., Thurman, R.E., Kryukov, G.V., Mirkin,
associate with nascent DNA during replication. Nat. Commun. 4, 2841. S.M., and Sunyaev, S.R. (2009). Human mutation rate associated with DNA
replication timing. Nat. Genet. 41, 393–395.
Pfander, B., Moldovan, G.L., Sacher, M., Hoege, C., and Jentsch, S. (2005).
SUMO-modified PCNA recruits Srs2 to prevent recombination during S phase. Stodola, J.L., and Burgers, P.M. (2016). Resolving individual steps of Okazaki-
Nature 436, 428–433. fragment maturation at a millisecond timescale. Nat. Struct. Mol. Biol. 23,
402–408.
Pitroda, S.P., Pashtan, I.M., Logan, H.L., Budke, B., Darga, T.E., Weichsel-
baum, R.R., and Connell, P.P. (2014). DNA repair pathway gene expression Tan, Z., Wortman, M., Dillehay, K.L., Seibel, W.L., Evelyn, C.R., Smith, S.J.,
score correlates with repair proficiency and tumor sensitivity to chemotherapy. Malkas, L.H., Zheng, Y., Lu, S., and Dong, Z. (2012). Small-molecule targeting
Sci. Transl. Med. 6, 229ra42. of proliferating cell nuclear antigen chromatin association inhibits tumor cell
growth. Mol. Pharmacol. 81, 811–819.
Plosky, B.S., Vidal, A.E., Fernández de Henestrosa, A.R., McLenigan, M.P.,
McDonald, J.P., Mead, S., and Woodgate, R. (2006). Controlling the subcellu- Terai, K., Abbas, T., Jazaeri, A.A., and Dutta, A. (2010). CRL4(Cdt2) E3 ubiqui-
lar localization of DNA polymerases iota and eta via interactions with ubiquitin. tin ligase monoubiquitinates PCNA to promote translesion DNA synthesis.
EMBO J. 25, 2847–2855. Mol. Cell 37, 143–149.

Povlsen, L.K., Beli, P., Wagner, S.A., Poulsen, S.L., Sylvestersen, K.B., Poul- Tian, F., Sharma, S., Zou, J., Lin, S.Y., Wang, B., Rezvani, K., Wang, H., Parvin,
sen, J.W., Nielsen, M.L., Bekker-Jensen, S., Mailand, N., and Choudhary, C. J.D., Ludwig, T., Canman, C.E., and Zhang, D. (2013). BRCA1 promotes
(2012). Systems-wide analysis of ubiquitylation dynamics reveals a key role the ubiquitination of PCNA and recruitment of translesion polymerases
for PAF15 ubiquitylation in DNA-damage bypass. Nat. Cell Biol. 14, in response to replication blockade. Proc. Natl. Acad. Sci. USA 110,
1089–1098. 13558–13563.

Prakash, R., Zhang, Y., Feng, W., and Jasin, M. (2015). Homologous recombi- Tsutakawa, S.E., Yan, C., Xu, X., Weinacht, C.P., Freudenthal, B.D., Yang, K.,
nation and human health: the roles of BRCA1, BRCA2, and associated pro- Zhuang, Z., Washington, M.T., Tainer, J.A., and Ivanov, I. (2015). Structurally
teins. Cold Spring Harb. Perspect. Biol. 7, a016600. distinct ubiquitin- and sumo-modified PCNA: implications for their distinct
roles in the DNA damage response. Structure 23, 724–733.
Punchihewa, C., Inoue, A., Hishiki, A., Fujikawa, Y., Connelly, M., Evison, B.,
Shao, Y., Heath, R., Kuraoka, I., Rodrigues, P., et al. (2012). Identification of Vannier, J.B., Sandhu, S., Petalcorin, M.I., Wu, X., Nabi, Z., Ding, H., and Boul-
small molecule proliferating cell nuclear antigen (PCNA) inhibitor that disrupts ton, S.J. (2013). RTEL1 is a replisome-associated helicase that promotes telo-
interactions with PIP-box proteins and inhibits DNA replication. J. Biol. Chem. mere and genome-wide replication. Science 342, 239–242.
287, 14289–14300.
Wang, S.C. (2014). PCNA: a silent housekeeper or a potential therapeutic
Raman, M., Havens, C.G., Walter, J.C., and Harper, J.W. (2011). A genome- target? Trends Pharmacol. Sci. 35, 178–186.
wide screen identifies p97 as an essential regulator of DNA damage-depen-
dent CDT1 destruction. Mol. Cell 44, 72–84. Wang, S.C., Nakajima, Y., Yu, Y.L., Xia, W., Chen, C.T., Yang, C.C., McIntush,
E.W., Li, L.Y., Hawke, D.H., Kobayashi, R., and Hung, M.C. (2006). Tyrosine
Rass, U., Ahel, I., and West, S.C. (2007). Defective DNA repair and neurode- phosphorylation controls PCNA function through protein stability. Nat. Cell
generative disease. Cell 130, 991–1004. Biol. 8, 1359–1368.

Rizzardi, L.F., Coleman, K.E., Varma, D., Matson, J.P., Oh, S., and Cook, J.G. Wang, X., Zhang, S., Zheng, R., Yue, F., Lin, S.H., Rahmeh, A.A., Lee, E.Y.,
(2015). CDK1-dependent inhibition of the E3 ubiquitin ligase CRL4CDT2 en- Zhang, Z., and Lee, M.Y. (2016). PDIP46 (DNA polymerase d interacting protein
sures robust transition from S Phase to Mitosis. J. Biol. Chem. 290, 556–567. 46) is an activating factor for human DNA polymerase d. Oncotarget 7,
6294–6313.
Roa, S., Avdievich, E., Peled, J.U., Maccarthy, T., Werling, U., Kuang, F.L.,
Kan, R., Zhao, C., Bergman, A., Cohen, P.E., et al. (2008). Ubiquitylated Warbrick, E. (2000). The puzzle of PCNA’s many partners. BioEssays 22,
PCNA plays a role in somatic hypermutation and class-switch recombination 997–1006.

Molecular Cell 65, February 2, 2017 391


Molecular Cell

Review
Watanabe, K., Tateishi, S., Kawasuji, M., Tsurimoto, T., Inoue, H., and Yamai- cation forks and PCNA unloading from lagging strands when forks stall. Mol.
zumi, M. (2004). Rad18 guides poleta to replication stalling sites through phys- Cell 56, 551–563.
ical interaction and PCNA monoubiquitination. EMBO J. 23, 3886–3896.
Yuan, J., Ghosal, G., and Chen, J. (2012). The HARP-like domain-containing
Waters, L.S., Minesinger, B.K., Wiltrout, M.E., D’Souza, S., Woodruff, R.V., protein AH2/ZRANB3 binds to PCNA and participates in cellular response to
and Walker, G.C. (2009). Eukaryotic translesion polymerases and their roles replication stress. Mol. Cell 47, 410–421.
and regulation in DNA damage tolerance. Microbiol. Mol. Biol. Rev. 73,
134–154. Zeman, M.K., and Cimprich, K.A. (2014). Causes and consequences of repli-
cation stress. Nat. Cell Biol. 16, 2–9.
Weston, R., Peeters, H., and Ahel, D. (2012). ZRANB3 is a structure-specific
ATP-dependent endonuclease involved in replication stress response. Genes
Zhang, Z., Shibahara, K., and Stillman, B. (2000). PCNA connects DNA replica-
Dev. 26, 1558–1572.
tion to epigenetic inheritance in yeast. Nature 408, 221–225.
Wit, N., Buoninfante, O.A., van den Berk, P.C., Jansen, J.G., Hogenbirk, M.A.,
de Wind, N., and Jacobs, H. (2015). Roles of PCNA ubiquitination and TLS Zhang, S., Chea, J., Meng, X., Zhou, Y., Lee, E.Y., and Lee, M.Y. (2008). PCNA
polymerases k and h in the bypass of methyl methanesulfonate-induced is ubiquitinated by RNF8. Cell Cycle 7, 3399–3404.
DNA damage. Nucleic Acids Res. 43, 282–294.
Zhang, Z., Zhang, S., Lin, S.H., Wang, X., Wu, L., Lee, E.Y., and Lee, M.Y.
Witko-Sarsat, V., Mocek, J., Bouayad, D., Tamassia, N., Ribeil, J.A., Candalh, (2012). Structure of monoubiquitinated PCNA: implications for DNA polymer-
C., Davezac, N., Reuter, N., Mouthon, L., Hermine, O., et al. (2010). Prolifer- ase switching and Okazaki fragment maturation. Cell Cycle 11, 2128–2136.
ating cell nuclear antigen acts as a cytoplasmic platform controlling human
neutrophil survival. J. Exp. Med. 207, 2631–2645. Zhang, S., Zhao, H., Darzynkiewicz, Z., Zhou, P., Zhang, Z., Lee, E.Y., and Lee,
M.Y. (2013). A novel function of CRL4(Cdt2): regulation of the subunit structure
Yang, X.H., Shiotani, B., Classon, M., and Zou, L. (2008). Chk1 and Claspin of DNA polymerase d in response to DNA damage and during the S phase.
potentiate PCNA ubiquitination. Genes Dev. 22, 1147–1152. J. Biol. Chem. 288, 29550–29561.

Yang, K., Weinacht, C.P., and Zhuang, Z. (2013a). Regulatory role of ubiquitin Zhao, H., Lo, Y.H., Ma, L., Waltz, S.E., Gray, J.K., Hung, M.C., and Wang, S.C.
in eukaryotic DNA translesion synthesis. Biochemistry 52, 3217–3228. (2011). Targeting tyrosine phosphorylation of PCNA inhibits prostate cancer
growth. Mol. Cancer Ther. 10, 29–36.
Yang, Y., Durando, M., Smith-Roe, S.L., Sproul, C., Greenwalt, A.M., Kauf-
mann, W., Oh, S., Hendrickson, E.A., and Vaziri, C. (2013b). Cell cycle
Zhao, H., Chen, M.S., Lo, Y.H., Waltz, S.E., Wang, J., Ho, P.C., Vasiliauskas,
stage-specific roles of Rad18 in tolerance and repair of oxidative DNA dam-
J., Plattner, R., Wang, Y.L., and Wang, S.C. (2014). The Ron receptor tyrosine
age. Nucleic Acids Res. 41, 2296–2312.
kinase activates c-Abl to promote cell proliferation through tyrosine phosphor-
Yin, L., Xie, Y., Yin, S., Lv, X., Zhang, J., Gu, Z., Sun, H., and Liu, S. (2015). The ylation of PCNA in breast cancer. Oncogene 33, 1429–1437.
S-nitrosylation status of PCNA localized in cytosol impacts the apoptotic
pathway in a Parkinson’s disease paradigm. PLoS ONE 10, e0117546. Zheng, L., and Shen, B. (2011). Okazaki fragment maturation: nucleases take
centre stage. J. Mol. Cell Biol. 3, 23–30.
Yu, Y., Song, C., Zhang, Q., DiMaggio, P.A., Garcia, B.A., York, A., Carey, M.F.,
and Grunstein, M. (2012). Histone H3 lysine 56 methylation regulates DNA Zlatanou, A., Despras, E., Braz-Petta, T., Boubakour-Azzouz, I., Pouvelle, C.,
replication through its interaction with PCNA. Mol. Cell 46, 7–17. Stewart, G.S., Nakajima, S., Yasui, A., Ishchenko, A.A., and Kannouche, P.L.
(2011). The hMsh2-hMsh6 complex acts in concert with monoubiquitinated
Yu, C., Gan, H., Han, J., Zhou, Z.X., Jia, S., Chabes, A., Farrugia, G., Ordog, T., PCNA and Pol h in response to oxidative DNA damage in human cells. Mol.
and Zhang, Z. (2014). Strand-specific analysis shows protein binding at repli- Cell 43, 649–662.

392 Molecular Cell 65, February 2, 2017

You might also like