You are on page 1of 11

MINIREVIEW

DNA mismatch repair system


Classical and fresh roles
Sung-Hoon Jun, Tae Gyun Kim and Changill Ban
Department of Chemistry and Division of Molecular & Life Science, Pohang University of Science and Technology, Korea

Key words The molecular mechanisms of the DNA mismatch repair (MMR) system
antibody diversification; DNA damage have been uncovered over the last decade, especially in prokaryotes. The
response; DNA mismatch repair; MutL;
results obtained for prokaryotic MMR proteins have provided a frame-
MutS
work for the study of the MMR system in eukaryotic organisms, such as
Correspondence yeast, mouse and human, because the functions of MMR proteins have
C. Ban, Department of Chemistry, Pohang been conserved during evolution from bacteria to humans. However, muta-
University of Science and Technology, tions in eukaryotic MMR genes result in pleiotropic phenotypes in addition
Pohang 790–784, Korea to MMR defects, suggesting that eukaryotic MMR proteins have evolved
Fax: +82 54 2793399 to gain more diverse and specific roles in multicellular organisms. Here, we
Tel: +82 54 2792127
summarize recent advances in the understanding of both prokaryotic and
E-mail: ciban@postech.ac.kr
eukaryotic MMR systems and describe various new functions of MMR
(Received 12 December 2005, accepted proteins that have been intensively researched during the last few years,
10 February 2006) including DNA damage surveillance and diversification of antibodies.

doi:10.1111/j.1742-4658.2006.05190.x

The mismatch repair (MMR) system is essential to all


Prokaryotic mismatch repair
organisms because it maintains the stability of the gen-
ome during repeated duplication. It is composed of a Essential components of the MMR system – MutS,
few well-conserved proteins whose functions in the MutL, MutH and Uvr – were identified in Escherichia
postreplicative repair of mismatched DNA have been coli through the genetic studies of mutants that showed
characterized by co-ordinated genetic, biochemical and elevated mutation levels [1,2]. MMR reactions have
structural approaches. Various functions, in addition also been reconstituted with purified components in
to mismatch repair during replication, have been E. coli [3], which drove extensive studies on prokaryotic
reported for MMR proteins such as antirecombination MMR systems.
activity between divergent sequences, promotion of MutS detects mismatches in DNA duplexes and initi-
meiotic crossover, DNA damage surveillance and ates the MMR machinery. A microscopic study sugges-
diversification of immunoglobulins (Fig. 1). Recent ted a possible mechanism for how MutS discriminates
research has provided a great deal of information between heteroduplex and homoduplex DNA [4].
about how MMR proteins are involved in these According to this proposal, nonspecifically bound MutS
diverse processes. bends DNA to search for a mismatch. If it recognizes a

Abbreviations
AID, activation-induced cytidine deaminase; ATM, ataxia telangiectasia mutated; ATR, ATM and Rad3-related; Chk1, checkpoint kinase 1;
Chk2, checkpoint kinase 2; CSR, class switch recombination; LC20, MutL C-terminal 20 kDa; LN40, MutL N-terminal 40 kDa; MLH, MutL
homolog; MMR, mismatch repair; MSH, MutS homolog; PCNA, proliferating cell nuclear antigen; PMS, postmeiotic segregation; RPA,
replication protein A; RFC, replication factor C; S, switch; SHM, somatic hypermutation; V, variable.

FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS 1609
Various functions of mismatch repair proteins S.-H. Jun et al.

Fig. 1. Various functions of mismatch repair


(MMR) proteins. MMR proteins are involved
in diverse genetic pathways through interac-
tions with different proteins. MMR proteins
increase replication fidelity by repairing
errors generated during replication. Prolifer-
ating cell nuclear antigen (PCNA) and replica-
tion factor C (RFC) work with MMR proteins
during mismatch repair in replication. Various
kinds of DNA damage trigger MMR protein-
dependent DNA damage responses that are
implemented through the activation of
ataxia telangiectasia mutated and Rad3-relat-
ed (ATR) and p53. Antibody diversification is
formed by mutations in immunoglobulin
genes that are introduced by MMR proteins
in conjunction with activation-induced cyti-
dine deaminase (AID) and DNA polymerase
g. In addition, MMR proteins regulate recom-
bination and promote meiotic crossover.
The functions of MMR proteins in green
boxes are discussed in this article, whereas
those in red boxes are not.

specific mismatch, MutS undergoes a conformational interactions and hydrogen bonds between the DNA
change and unbends the bent DNA. Crystallographic backbone and side chains of MutS are sequence inde-
studies of Thermus aquaticus and E. coli MutS pendent [5,6]. After the recognition of mismatched
complexed with mismatched DNA provided the molecu- DNA, MutS initiates the MMR system through direct
lar details of mismatch recognition [5–7], suggesting that or indirect interactions with other proteins, including
a homodimer of MutS binds asymmetrically to hetero- MutL, MutH and UvrD. Although an exact answer to
duplex DNA (Fig. 2A). MutS has two functional this puzzle is yet to be found, a few groups have sugges-
domains (a DNA-binding domain and an ATPase ⁄ ted various models for detailed molecular events during
dimerization domain) and the asymmetry in the MMR reactions (Fig. 3) [11].
ATPase ⁄ dimerization domain was also reported to be The function of MutL in the MMR system is to
essential in the MMR process in vivo [8]. These two make a connection between the recognition of a mis-
domains are widely separated from each other, but match and the excision of the mismatch from the
affect each other by conformational changes that are strand within which it is contained [12]. To do this, a
induced by the binding of DNA or ATP [9]. This inter- MutL homodimer interacts with MutS [13] and stimu-
action is a key molecular mechanism for modulating the lates the endonuclease activity of MutH [14]. MutL
function of the MutS protein in the MMR process. Only also loads UvrD onto the DNA. UvrD is a DNA
two residues, both in the same subunit of MutS, take helicase II that unwinds the DNA duplex from the
part in the sequence-specific interaction with a mis- nick generated by MutH [15,16]. MutL is a member of
matched base. One, a conserved glutamate (Glu41 in the GHKL superfamily of ATPases, which includes gy-
T. aquaticus MutS and Glu38 in E. coli MutS), forms a rase, a type II topoisomerase, Hsp90, histidine kinase
hydrogen bond with the mismatched base. Recently, this and MutL [17]. A biochemical study demonstrated that
hydrogen bond was suggested to induce an inhibition MutL has ATPase activity [17,18]. Crystallographic
of the ATPase activity of MutS, helping to form a stable studies have demonstrated that ATP binding drives
MutS–ATP–DNA intermediate of the downstream dimerization of the N-terminal domain of the protein
repair process [10]. The other specific interaction is (Fig. 2B) [18], and the accompanying structural chan-
between an aromatic ring stack of a conserved phenyl- ges may play key roles in co-ordinating the initial steps
alanine (Phe39 in T. aquaticus MutS and Phe36 in of mismatch recognition with downstream processing
E. coli MutS) and the mismatched base. In contrast to steps. A model for the intact MutL protein, which
these sequence–specific interactions, van der Waals includes a large central cavity, was suggested based on

1610 FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS
S.-H. Jun et al. Various functions of mismatch repair proteins

Fig. 2. Structures of MutS, MutL and MutH. (A) Crystal structure of the Thermus aquaticus MutS heteroduplex DNA complex (PDB acces-
sion code: 1EWQ). The MutS homodimer is formed by asymmetric subunits that are represented by ribbon diagrams in green and purple.
The heteroduplex DNA is a space-filling model. Two adjacent large channels with dimensions of  30 · 20 Å and  40 · 20 Å penetrate the
disk-like protein structure, and the latter is occupied by the heteroduplex DNA. The DNA is kinked sharply towards the major groove by
 60 at the unpaired base. Only one subunit (in purple) interacts with the unpaired base, thereby breaking the molecular twofold symmetry
of the homodimer. (B) Crystal structure of the N-terminal 40 kDa fragment (LN40) of Escherichia coli MutL complexed with ADPnP (PDB
accession code: 1B63). The structure of LN40 is homologous to that of an ATPase-containing fragment of DNA gyrase. ADPnP drives the
dimerization of LN40, and the dimer interface is well ordered and made entirely of the segments that were disordered in the apoprotein. (C)
A crystal structure of MutH (PDB accession code: 1AZO). The structure resembles a clamp, with a large cleft dividing the molecule into two
halves. Each half forms a subdomain that contains similar structural elements. The two subdomains share a hydrophobic interface and are
connected by three polypeptide linkers. The active site is located at an interface between two subdomains, and DNA binds in the cleft that
is 15–18 Å wide and 12–14 Å deep.

the structures of the N-terminal domain (LN40) and synthesized strand between the nick and the mismatch
the C-terminal domain (LC20), which were reported is carried out by four redundant single-strand DNA-
separately [17–19]. A biochemical assay, using various specific exonucleases: the 3¢ fi 5¢ exonucleases ExoI
mutant MutL proteins, suggested that LC20 is involved and ExoX and the 5¢ fi 3¢ exonucleases RecJ and
in the DNA-binding activity of MutL. An increase in ExoVII [22]. DNA polymerase III, single-stranded
the DNA-binding activity of MutL also resulted in DNA-binding protein and DNA ligase carry out repair
higher UvrD helicase activity [19]. synthesis [3].
MutH is a member of the type II family of res-
triction endonucleases and cleaves at hemimethyl-
Eukaryotic mismatch repair
ated GATC sites for excision of mismatch-containing
strands [20]. The nicking activity of MutH is stimula- All eukaryotic organisms, including yeast, mouse and
ted in a mismatch-dependent manner by MutS, MutL human, have MutS homologs (MSHs) and MutL
and ATP [20]. A structural study suggested that the homologs (MLHs). The eukaryotic MMR system has
C-terminal helix of MutH might act as a molecular been well conserved during the evolutionary process
lever through which MutS and MutL communicate [3,23]. However, in contrast to MutS and MutL in
and activate MutH (Fig. 2C) [21]. The nick generated bacteria, which function as homodimers, in eukaryotes
by MutH serves as a point of entry for single-stranded MSHs and MLHs form heterodimers with multiple
DNA-binding protein and UvrD ⁄ helicase II, whose proteins. Five highly conserved MSHs (MSH2 to
loading at the nick is facilitated via protein–protein MSH6) are present in both yeast and mammals.
interactions with MutL [15,16]. Excision of the newly MSH1, which is present in mitochondria, exists only in

FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS 1611
Various functions of mismatch repair proteins S.-H. Jun et al.

MSH2 and MSH6, than defects in the two other MutL


homologs [30]. Also, MLH1 interacted with the other
three MutL homologs in a yeast two-hybrid analysis
[31]. Overall, yeast MLH1 ⁄ PMS1 and mammalian
MLH1 ⁄ PMS2 heterodimers (each known as MutLa)
play a major role in mutation avoidance, and the other
two heterodimers of MutL homologs take part in the
repair of specific classes of mismatches [32]. The bio-
chemical activities and structure of MutL homologs
are closely related to those of prokaryotic MutL pro-
teins, especially in the N-terminal domain. The X-ray
crystallographic structure of the conserved N-terminal
40-kDa fragment of human PMS2 resembles that of
the ATPase fragment of E. coli MutL [33].
Extensive genetic studies in yeast have failed to find
orthologs of MutH and UvrD in the MMR system,
Fig. 3. Models for the assembly of the DNA mismatch repair
and there may be no homolog of these two proteins in
complex in a schematic drawing. A mismatch base is detected the eukaryotic genome [34]. Therefore, some diver-
by MutS, and ATP-bound MutS recruits MutL. In model I, the gence in the MMR system from strand discrimination
MutS–MutL complex stays at the mismatch site and activates and the nicking process might occur between prokary-
MutH at some distance. MutS leaves the mismatch site, after bind- otes and eukaryotes. A recent increase in our know-
ing ATP, in both model II and model III. ATP is used as an energy ledge of the eukaryotic MMR system provides some
source for translocation of MutS in model II (translocation model)
understanding of this divergence.
but it acts as a molecular switch of MutS in model III, like GTP of
G-proteins (molecular switch model).
In mammalian cell extracts, mismatches provoke ini-
tiation of excision at pre-existing nicks in exogenous
DNA substrates with high efficiency and specificity
yeast [24]. MSH4 and MSH5 show reproductive tis- [35,36]. The molecular nature of eukaryotic MMR
sue-specific expression, and null mutations of these could be assessed using cell extract assays in vitro, and
genes do not confer mutator phenotypes because they components of the eukaryotic MMR system have been
are involved in meiotic recombination but not postrep- identified with depletion and complementation assays
lication repair [25]. Genetic and biochemical studies using cell extracts. One protein, identified in this way,
have indicated that MSH2 is required for all mismatch is proliferating cell nuclear antigen (PCNA). PCNA is
correction in nuclear DNA, whereas MSH3 and known to function as a processivity factor for replica-
MSH6 are required for the repair of some distinct and tive polymerase, but some mutations in the PCNA
overlapping types of mismatched DNA during replica- gene result in mutator phenotypes [37], and its interac-
tion [26]. These three MutS homologs make two tions with MSH2 and MLH1 [38], and with MSH6
heterodimers: MutSa (MSH2 ⁄ MSH6) and MutSb [39], suggest that it functions in MMR. PCNA has
(MSH2 ⁄ MSH3). The former plays the major role in biochemical activity that increases the binding of
recognition of mismatched DNA in eukaryotic MMR. MutSa to mismatched DNA; the interactions between
That is, MutSa functions in the repair of base–base PCNA and MSH6 are essential for this biochemical
mispairs as well as a range of insertion ⁄ deletion loop activity, which suggests that PCNA might play a role
mispairs, whereas MutSb primarily functions in the in MMR at the mispair recognition stage [39]. PCNA
repair of insertion ⁄ deletion loop mispairs [27,28]. has been proposed to function in the mismatch recog-
MutL homologs in eukaryotic organisms were iden- nition stage of MMR by helping MutSa search for
tified as genes whose amino acid sequences showed mismatched DNA [40] or increasing the mismatch-
high similarity with prokaryotic MutL proteins, or binding specificity of MutSa [39]. One intriguing point
whose mutation phenotypes were increased levels of about the role of PCNA in eukaryotic MMR is that
postmeiotic segregation (PMS) that resulted from a the requirement for PCNA depends on the direction of
failure to repair mismatches in meiotic recombination the nick in the in vitro MMR assay. Although PCNA
intermediates [29]. There are four homologs of MutL is required for mismatch-provoked excision directed
in both yeast and mammals. In a genetic analysis, by a 3¢ strand break in HeLa nuclear extracts, it
defects in MLH1 and PMS1 in yeast resulted in more is not essential for excision directed by a 5¢ nick
severe mutator phenotypes, reminiscent of those of [41,42]. Moreover, whereas 3¢ nick-directed excision is

1612 FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS
S.-H. Jun et al. Various functions of mismatch repair proteins

completely abolished by the inhibition of PCNA, 5¢ DNA damage in normal cells showed that N-methyl-
nick-directed excision is affected only minimally [42]. N¢-nitro-N-nitrosoguanidine, an alkylating agent, trig-
Finally, a mismatch-provoked 5¢ fi 3¢excision reaction gers MMR-dependent G2 ⁄ M arrest [54], which is
can be reconstituted in a purified system that compri- followed by the induction of MMR-dependent apopto-
ses only MutSa, MutLa, ExoI and replication protein sis [55].
A (RPA), without PCNA, and the process is similar The role of MMR proteins in response to DNA
to that observed in nuclear extracts [41]. RPA, the damage can be inferred from the interactions of MMR
eukaryotic single-stranded DNA-binding protein, has proteins with the tumor suppressor protein, p53, and
been shown to enhance excision and stabilize excision p53-related proteins. p53 acts as a major point in a
intermediates in crude fractions [43,44]. The activities complex network that responds to diverse cellular
of ExoI are described below. stresses, including DNA damage [56]. Once stabilized
Genetic studies in yeast, and biochemical studies of and activated by genotoxic stress, p53 can either acti-
MMR activity in cell extracts, indicate that eukaryotes vate or repress a wide array of different gene targets
use a mechanism similar to prokaryotes, with both by binding to their promoter regions, which in turn
3¢ fi 5¢ and 5¢ fi 3¢ exonuclease activities for mis- can regulate cell cycle, cell death and other outcomes
match correction [45]. ExoI, a 5¢ fi 3¢ exonuclease, [57]. The p53 homologs p63 and p73 induce p53-inde-
was found to play a role in mutation avoidance and pendent apoptosis as well as affect trans-activation
mismatch repair in yeast [46], and its physical inter- of certain target genes by p53 [58,59]. Treatment of
action with MSH2 and MLH1 also support a role in human cells with methylating agents results in phos-
MMR [47]. Intriguingly, the mammalian ExoI was phorylation of p53 and induction of apoptosis, a
reported to be involved in both 5¢- and 3¢ nick-directed response that depends on the presence of functional
excision in extracts of mammalian cells [48], but how hMutSa and hMutLa [60]. UVB-induced apoptosis is
ExoI can have a 3¢ fi 5¢ exonuclease activity was significantly reduced in MSH2-deficient cells, and it
unclear. Recent research by the Modrich group pro- correlates with decreased activation of p53, which sug-
vides a plausible answer to this question [49]. They gests that MSH2 may act upstream of p53 to induce
reconstituted mismatch-provoked excision, directed by post-UVB apoptosis [61]. Cisplatin-caused DNA dam-
a strand break located either 3¢ or 5¢ to the mispair, in age increases the stability of p73, which induces apop-
a defined human system using purified human proteins. tosis that is dependent on functional hMLH1 protein
In the presence of the eukaryotic clamp loader replica- [62]. Moreover, cisplatin stimulates the interaction
tion factor C (RFC) and PCNA, 3¢ fi 5¢ excision was between PMS2 and p73, which is required for the acti-
supported by MutSa, MutLa, ExoI and RPA. More- vation of p73 and subsequent induction of apoptosis
over, RFC and PCNA act to suppress 5¢ fi 3¢ excision [63]. PMS2 and p73 can also interact with each other,
when the strand break that directs hydrolysis is located independently of MLH1, suggesting that MMR pro-
3¢ to the mismatch, which suggests that the polarity of teins have specific roles in the DNA damage response.
mismatch-provoked excision by ExoI is regulated by Taken together, these reports indicate that MMR pro-
PCNA and RFC. Once the strand is excised beyond teins may play roles in multiple steps of the DNA
the mismatch, DNA resynthesis occurs by the activity damage response, as damage sensors and adaptors of
of polymerase d [50] in the presence of PCNA [51] and the pathways (Fig. 4).
RPA [43,44]. The remaining nick is then sealed by The roles of MMR proteins in the response to DNA
an as-yet-unidentified ligase, completing the repair damage are further supported by the failure of MMR
process. mutants to trigger G2 ⁄ M arrest in response to the
methylator N-methyl-N¢-nitro-N-nitrosoguanidine and
similar alkylators [64]. The G2 ⁄ M checkpoint prevents
MMR proteins in the DNA damage
cells from initiating mitosis when they experience
response
DNA damage during G2, or when they progress into
The involvement of MMR proteins in the DNA dam- G2 with unrepaired damage incurred during the previ-
age response first became apparent when it was discov- ous S or G1 phases [65]. A study with a cell line lack-
ered that MMR-defective bacterial and mammalian ing hMLH1 expression and an inducible hMLH1
cells are resistant to cell death caused by alkylating expression system showed that methylation-induced
agents [52]. MMR-deficient cells are also resistant to G2 ⁄ M arrest requires a full complement of hMLH1
other DNA-damaging agents, including methylation (expression level similar to that of the wild type),
agents, cisplatin and UV radiation [53]. Subsequent whereas MMR proficiency was restored, even at low
studies on the roles of MMR proteins in response to hMLH1 concentrations ( 10% of wild-type expres-

FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS 1613
Various functions of mismatch repair proteins S.-H. Jun et al.

interaction between MMR proteins and checkpoint


proteins also suggests direct roles for MMR proteins
in the DNA damage response. Both in vitro and in vivo
approaches show that MSH2 binds to Chk1 and Chk2
[68], that MLH1 associates with ATM [70] and that
these interactions are enhanced after treatment with a
methylating agent [68]. MSH2 protein physically inter-
acts with ATR in the damage response to DNA
methylation, and their interaction is required for the
phosphorylation of Chk1 [71]. ATR also serves as a
haploinsufficient tumor suppressor in MMR-deficient
cells, suggesting the genetic interaction of these pro-
teins [72]. Taken together, these findings suggest that
MMR proteins function early in the pathway that
leads from DNA methylating agents to G2 ⁄ M arrest
(Fig. 4).
The molecular mechanism of the involvement of
MMR proteins in various DNA damage responses
is unclear. Given the original function of the MMR
system in detecting and repairing errors that occur
during replication, the MMR protein complex could
serve as a sensor for DNA damage [71]. A large com-
plex, named BRCA1-associated genome surveillance
complex, which includes tumor suppressors and the
MMR ⁄ DNA damage-repair proteins MSH2, MSH6,
Fig. 4. A simplified model of DNA damage response pathways
MLH1, ATM, Bloom’s syndrome, and RAD50–
that are dependent on mismatch repair (MMR) proteins. MMR MRE11–NBS1, has also been suggested to be a poss-
proteins bind to damaged DNA and recruit various signal-transducing ible sensor for DNA damage [73]. The roles of MMR
kinases, including ataxia telangiectasia mutated (ATM), ATM and proteins in the DNA damage response may not be
Rad3-related (ATR), and checkpoint kinase 1 ⁄ checkpoint kinase 2 simple from the viewpoint of their various relation-
(Chk1 ⁄ Chk2). They in turn stabilize and activate p53, a key compo- ships with other regulators of the DNA damage
nent in DNA damage responses, such as cell cycle checkpoint
response, especially with ATM and p53. For instance,
activation and programmed cell death (apoptosis). p73, a p53
homolog, is also a transducer of the MMR protein-dependent DNA
hMLH1 and hPMS2 were identified as direct target
damage response, and postmeiotic segregation 2 (PMS2) is known genes of p53 [74]. Cisplatin induces the accumulation
to bind and stabilize p73. The p38 mitogen-activated protein (MAP) of hPMS1, hPMS2 and hMLH1 through ATM-medi-
kinase pathway connects MMR proteins and p53 ⁄ p73 in this ated protein stabilization, and the induced level of
pathway. c-Abl is a tyrosine kinase that acts upstream of p73 and these MMR proteins is important for the phosphorylat-
stabilizes it [59]. ion of p53 by ATM in the response to DNA damage
[75]. MMR proteins and p53 therefore may act as a
sion) [66], suggesting that these two responses are kind of positive feedback regulation for the DNA
carried out by different genetic pathways. The compo- damage response, or a more complicated network may
nents that transduce the G2 ⁄ M checkpoint signal regulate their activity and expression.
pathway, such as ataxia telangiectasia mutated (ATM),
ATM and Rad3-related (ATR), checkpoint kinase 1
MMR proteins in antibody
(Chk1) and checkpoint kinase 2 (Chk2), are activated
diversification
in the MMR system-dependent G2 ⁄ M arrest induced
by DNA methylation [67,68]. ATR and Chk1 path- In addition to the initial generation of antibody diver-
ways are essential for this response [67]. The mitogen- sity by gene rearrangement during B-cell development
activated protein (MAP) kinase, p38a, is activated in [76], specific antigen recognition triggers a second wave
MMR-proficient cells exposed to the methylating of antibody diversification through somatic hypermu-
agent, temozolomide, but not in MLH1 knockdown tation (SHM) and class switch recombination (CSR).
cells [69], suggesting that the p38 MAP kinase pathway SHM introduces multiple single-nucleotide substitut-
links the MMR system to the G2 ⁄ M checkpoint. The ions into variable (V) regions of immunoglobulin

1614 FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS
S.-H. Jun et al. Various functions of mismatch repair proteins

genes and CSR is a region-specific intrachromosomal to deaminate dC to dU subsequently in ssDNA


recombination that replaces the Cl form of the immu- [82,83]. Transcription of the Ig gene in the V and
noglobulin (Ig) heavy chain constant region (CH) gene switch (S) regions is required for SHM and CSR,
with other CH genes, resulting in a switch of the Ig iso- respectively [84], because AID deaminates cytidine
type from IgM to IgG, IgE, or IgA [77]. The mole- residues in single-stranded DNA located in the tran-
cular processes of SHM and CSR, and the proteins scription bubble of the V and S regions [85,86],
involved in these processes, have been investigated in (Fig. 5). AID-induced mutations of cytidine explain
detail over the last few years. Advances in gene target- some SHM and mutations in CSR, but up to half of
ing techniques have led to the availability of mice with the mutations of the V and S regions are independent
loss-of-function mutations in MMR genes, and recent of AID. Phenotypic analyses of MSH2- and MSH6-
studies using these mice have suggested that MMR defective mice showed that the spectra of SHM were
proteins are directly involved in antibody diversifica- different in these mice than in wild-type mice [78,79],
tion. MSH2-deficient mice accumulated fivefold fewer indicating that MSH2 and MSH6 are required for
mutations in the V region of antibody genes [78]. mutations at AT base pairs during SHM and CSR
MSH6 deficiency caused similar effects, but MSH3 [78,79]. These results suggested that mutations in
deficiency did not [79], suggesting that MutSa plays an SHM and CSR are achieved in two steps: in the first
essential role in SHM. Similarly, mice with loss- step, AID generates mutations in GC base pairs, and
of-function mutations in MSH2 or MSH6 have a in the second step, the MMR system is recruited to
decreased frequency of CSR, but those with MSH3 the mismatched DNA and resynthesizes the DNA
do not [80]. Mice carrying a mutation in the MSH2 strand with the help of an error-prone polymerase,
ATPase domain are deficient in SHM and CSR, sug- such as pol g (Fig. 5) [87]. This model is supported
gesting that the ATPase activity of MSH2 is essential by a report that MSHa not only binds to a U:G
for antibody diversification [81]. It will be interesting mispair, but also physically interacts with DNA poly-
to understand how MMR proteins are involved in the merase g and functionally stimulates its catalytic
processes of SHM and CSR, which require the induc- activity [88]. Moreover, the phenotypes of mice
tion of mutations. mutant for ExoI are similar to those of MSH2– ⁄ –
Both SHM and CSR start with the activity of acti- mice, with reduced SHM and CSR, and ExoI and
vation-induced cytidine deaminase (AID), which is a MLH1 physically interact with mutating variable
homolog of the RNA editing enzyme, but is known regions [89].

Fig. 5. A model of somatic hypermutation that is dependent on the mismatch repair (MMR) protein. (A) During transcription of the immuno-
globulin gene in the variable (V) region, activation-induced cytidine deaminase (AID) deaminates cytidine residues in single-stranded DNA to
produce UG mismatches. (B) MutSa and MutLa are recruited to the mismatched DNA, and activate ExoI. (C) The gaps generated by the
activity of ExoI are refilled by error-prone DNA polymerase g, resulting in mutations in AT base pairs. (D) The diversity of the V regions of
antibody genes is thus accomplished by the formation of mutations by a mechanism that depends on MMR proteins.

FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS 1615
Various functions of mismatch repair proteins S.-H. Jun et al.

Conclusion 8 Lamers MH, Winterwerp HH & Sixma TK (2003) The


alternating ATPase domains of MutS control DNA mis-
The MMR system was originally discovered as a match repair. EMBO J 22, 746–756.
mechanism that maintains the integrity of the genome 9 Lamers MH, Georgijevic D, Lebbink JH, Winterwerp
during replication. Increasingly, however, components HH, Agianian B, de Wind N & Sixma TK (2004) ATP
of the system are being found to participate in diverse increases the affinity between MutS ATPase domains.
cellular processes, including the repair of DNA dam- J Biol Chem 279, 43879–43885.
age and antibody diversification. How MMR proteins 10 Lebbink JH, Georgijevic D, Natrajan G, Fish A, Winter-
are regulated to perform these various functions will werp HH, Sixma TK & de Wind N (2006) Dual role of
be an important question for the co-ordinated under- MutS glutamate 38 in DNA mismatch discrimination
standing of MMR proteins. Searching for the unidenti- and in the authorization of repair. EMBO J 25, 409–419.
fied components of the MMR system will also provide 11 Kunkel TA & Erie DA (2005) DNA mismatch repair.
further information in the growing body of research Annu Rev Biochem 74, 681–710.
on the mechanism of the MMR system. Finally, 12 Sancar A & Hearst JE (1993) Molecular matchmakers.
understanding the MMR system will provide insights Science 259, 1415–1420.
into cancer development related to the defects in 13 Galio L, Bouquet C & Brooks P (1999) ATP hydro-
MMR genes and the treatment of tumors, both hered- lysis-dependent formation of a dynamic ternary nucleo-
itary and sporadic, with defective MMR. protein complex with MutS and MutL. Nucleic Acids
Res 27, 2325–2331.
14 Hall MC & Matson SW (1999) The Escherichia coli
Acknowledgements MutL protein physically interacts with MutH and sti-
mulates the MutH-associated endonuclease activity.
This work was supported by the Center for Integrated
J Biol Chem 274, 1306–1312.
Molecular Systems through KOSEF, the POSRIP res-
15 Dao V & Modrich P (1998) Mismatch-, MutS-, MutL-,
earch grant (1RC0402301), and the Center for Innova- and helicase II-dependent unwinding from the single-
tive Bio-Physio Technology at BNU (grant number: strand break of an incised heteroduplex. J Biol Chem
02-PJ3-PG6-EV05-0001). 273, 9202–9207.
16 Hall MC, Jordan JR & Matson SW (1998) Evidence for
References a physical interaction between the Escherichia coli
methyl-directed mismatch repair proteins MutL and
1 Wagner R & Meselson M (1976) Repair tracts in mis- UvrD. EMBO J 17, 1535–1541.
matched DNA heteroduplexes. Proc Natl Acad Sci USA 17 Ban C & Yang W (1998) Crystal structure and ATPase
73, 4135–4139. activity of MutL: implications for DNA repair and
2 Cox EC, Degnen GE & Scheppe ML (1972) Mutator mutagenesis. Cell 95, 541–552.
gene studies in Escherichia coli: the mutS gene. Genetics 18 Ban C, Junop M & Yang W (1999) Transformation of
72, 551–567. MutL by ATP binding and hydrolysis: a switch in DNA
3 Modrich P & Lahue R (1996) Mismatch repair in repli- mismatch repair. Cell 97, 85–97.
cation fidelity, genetic recombination, and cancer biol- 19 Guarne A, Ramon-Maiques S, Wolff EM, Ghirlando
ogy. Annu Rev Biochem 65, 101–133. R, Hu X, Miller JH & Yang W (2004) Structure of the
4 Wang H, Yang Y, Du Schofield MJC, Fridman Y, Lee MutL C-terminal domain: a model of intact MutL and
SD & Larson ED (2003) DNA bending and unbending its roles in mismatch repair. EMBO J 23, 4134–4145.
by MutS govern mismatch recognition and specificity. 20 Au KG, Welsh K & Modrich P (1992) Initiation of
Proc Natl Acad Sci USA 100, 14822–14827. methyl-directed mismatch repair. J Biol Chem 267,
5 Obmolova G, Ban C, Hsieh P & Yang W (2000) 12142–12148.
Crystal structures of mismatch repair protein MutS 21 Ban C & Yang W (1998) Structural basis for MutH
and its complex with a substrate DNA. Nature 407, activation in E. coli mismatch repair and relationship of
703–710. MutH to restriction endonucleases. EMBO J 17, 1526–
6 Lamers MH, Perrakis A, Enzlin JH, Winterwerp HH, 1534.
de Wind N & Sixma TK (2000) The crystal structure of 22 Burdett V, Baitinger C, Viswanathan M, Lovett ST &
DNA mismatch repair protein MutS binding to a GT Modrich P (2001) In vivo requirement for RecJ, ExoVII,
mismatch. Nature 407, 711–717. ExoI, and ExoX in methyl-directed mismatch repair.
7 Junop MS, Obmolova G, Rausch K, Hsieh P & Yang Proc Natl Acad Sci USA 98, 6765–6770.
W (2001) Composite active site of an ABC ATPase: 23 Kolodner RD (1996) Biochemistry and genetics of euk-
MutS uses ATP to verify mismatch recognition and aryotic mismatch repair. Genes Dev 10, 1433–1442.
authorize DNA repair. Mol Cell 7, 1–12.

1616 FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS
S.-H. Jun et al. Various functions of mismatch repair proteins

24 Reenan RAG & Kolodner RD (1992) Characterization 38 Umar A, Buermeyer AB, Simon JA, Thomas DC, Clark
of insertion mutations in the Saccharomyces cerevisiae AB, Liskay RM & Kunkel TA (1996) Requirement for
MSH1 and MSH2 genes: evidence for separate mito- PCNA in DNA mismatch repair at a step preceding
chondrial and nuclear functions. Genetics 132, 975–985. DNA resynthesis. Cell 87, 65–73.
25 Ross-Macdonald P & Roeder GS (1994) Mutation of a 39 Flores-Rozas H, Clark D & Kolodner RD (2000) Prolif-
meiosis-specific MutS homolog decreases crossing over erating cell nuclear antigen and Msh2p-Msh6p interact
but not mismatch correction. Cell 79, 1069–1080. to form an active mispair recognition complex. Nat
26 Marsischky GT, Filosi N, Kane MF & Kolodner R Genet 26, 375–378.
(1996) Redundancy of Saccharomyces cerevisiae MSH3 40 Lau PJ & Kolodner RD (2003) Trnasfer of the MSH2-
and MSH6 in MSH2-dependent mismatch repair. Genes MSH6 complex from proliferating cell nuclear antigen
Dev 10, 407–420. to mispaired bases in DNA. J Biol Chem 278, 14–17.
27 Genschel J, Littman SJ, Drummond JT & Modrich P 41 Genschel J & Modrich P (2003) Mechanism of 5¢ direc-
(1998) Isolation of MutSb from human cells and com- ted excision in human mismatch repair. Mol Cell 12,
parison of the mismatch repair specificities of MutSb 1077–1086.
and MutSa. J Biol Chem 273, 19895–19901. 42 Guo S, Presnell SR, Yuan F, Zhang Y, Gu L & Li GM
28 Umar A, Risinger JI, Glaab WE, Tindall KR, Barrett (2004) Differential requirement for proliferating cell
JC & Kunkel TA (1998) Functional overlap in mis- nuclear antigen in 5¢-and 3¢ nick-directed excision in
match repair by human MSH3 and MSH6. Genetics human mismatch repair. J Biol Chem 279, 16912–16917.
148, 1637–1646. 43 Lin YL, Shivji MKK, Chen C, Kolodner R, Wood RD
29 Kramer W, Kramer B, Williamson MS & Fogel S & Dutta A (1998) The evolutionarily conserved zinc fin-
(1989) Cloning and nucleotide sequence of DNA mis- ger motif in the largest subunit of human replication
match repair gene PMS1 from Saccharomyces cerevi- protein A is required for DNA replication and mis-
siae: homology of PMS1 to procaryotic MutL and match repair but not for nucleotide excision repair.
HexB. J Bacteriol 171, 5339–5346. J Biol Chem 273, 1453–1461.
30 Prolla T, Christie DM & Liskay RM (1994) Dual 44 Ramilo C, Gu L, Guo S, Zhang X, Patrick SM, Turchi
requirement in yeast DNA mismatch repair for MLH1 JJ & Li GM (2002) Partial reconstitution of human
and PMS1, two homologs of the bacterial mutL gene. DNA mismatch repair in vitro: characterization of the
Mol Cell Biol 14, 407–415. role of human replication protein A. Mol Cell Biol 22,
31 Wang TF, Kleckner N & Hunter N (1999) Functional 2037–2046.
specificity of MutL homologs in yeast: Evidence for 45 Tran HT, Gordenin DA & Resnick MA (1999) The
three Mlh1-based heterocomplexes with distinct roles 3¢ fi 5¢ exonucleases of DNA polymerases d and e and
during meiosis in recombination and mismatch correc- the 5¢ fi 3¢ exonuclease Exo1 have major roles in post-
tion. Proc Natl Acad Sci USA 96, 13914–13919. replication mutation avoidance in Saccharomyces cere-
32 Harfe BD, Minesinger BK & Jinks-Robertson S (2000) visiae. Mol Cell Biol 19, 2000–2007.
Discrete in vivo roles for the MutL homologs Mlh2p 46 Huang KN & Symington LS (1993) A 5¢ fi 3¢ exonuc-
and Mlh3p in the removal of frameshift intermediates in lease from Saccharomyces cerevisiae is required for
budding yeast. Curr Biol 10, 145–148. in vitro recombination between linear DNA molecules
33 Guarne A, Junop MS & Yang W (2001) Structure and with overlapping homology. Mol Cell Biol 13, 3125–3134.
function of the N-terminal 40 kDa fragment of human 47 Tran PT, Simon JA & Liskay RM (2001) Interactions
PMS2: a monomeric GHL ATPase. EMBO J 20, 5521– of Exo1p with components of MutLa in Saccharomyces
5531. cerevisiae. Proc Natl Acad Sci USA 98, 9760–9765.
34 Hafe BD & Robertson SJ (2000) DNA mismatch repair 48 Genschel J, Bazemore LR & Modrich P (2002) Human
and genetic instability. Annu Rev Genet 34, 359–399. exonuclease I is required for 5¢- and 3¢ mismatch repair.
35 Holmes J, Clark S & Modrich P (1990) Strand-specific J Biol Chem 277, 13302–13311.
mismatch correction in nuclear extracts of human and 49 Dzantiev L, Constantin N, Genschel J, Iyer RR, Burg-
Drosophila melanogaster cell lines. Proc Natl Acad Sci ers PMJ & Modrich P (2004) A defined human system
USA 87, 5837–5841. that supports bidirectional mismatch-provoked excision.
36 Iams K, Larson ED & Drummond JT (2002) DNA Mol Cell 15, 31–41.
template requirements for human mismatch repair 50 Longley MJ, Pierce AJ & Modrich P (1997) DNA poly-
in vitro. J Biol Chem 277, 30805–30814. merase d is required for human mismatch repair in vitro.
37 Ayyagari R, Impellizzeri KJ, Yoder BL, Gary SL & J Biol Chem 272, 10917–10921.
Bergers PM (1995) A multinational analysis of the yeast 51 Gu L, Hong Y, McCulloch S, Watanabe H & Li GM
proliferating cell nuclear antigen indicates distinct roles (1998) ATP-dependent interaction of human mismatch
in DNA replication and DNA repair. Mol Cell Biol 15, repair proteins and dual role of PCNA in mismatch
4420–4429. repair. Nucleic Acids Res 26, 1173–1178.

FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS 1617
Various functions of mismatch repair proteins S.-H. Jun et al.

52 Branch P, Aquilina G, Bignami M & Karran P (1993) Methylation-induced G(2) ⁄ M arrest requires a full com-
Defective mismatch binding and a mutator phenotype plement of the mismatch repair protein hMLH1. EMBO
in cells tolerant to DNA damage. Nature 362, 652–654. J 22, 2245–2254.
53 Fink D, Aebi S & Howell SB (1998) The role of DNA 67 Stojic L, Mojas N, Cejka P, Di Pietro M, Ferrari S,
mismatch repair in drug resistance. Clin Cancer Res 4, Marra G & Jiricny J (2004) Mismatch repair-dependent
1–6. G2 checkpoint induced by low doses of SN1 type
54 Hawn MT, Umar A, Carethers JM, Marra G, Kunkel methylating agents requires the ATR kinase. Genes Dev
TA, Boland CR & Koi M (1995) Evidence for a connec- 18, 1331–1344.
tion between the mismatch repair system and the G2 68 Adamson AW, Beardsley DI, Kim W, Gao Y, Baskaran
cell cycle checkpoint. Cancer Res 55, 3721–3725. R & Brown KD (2005) Methylator-induced, mismatch
55 Tominaga Y, Tsuzuki T, Shiraishi A, Kawate H & repair-dependent G2 arrest is activated through Chk1
Sekiguchi M (1997) Alkylation-induced apoptosis of and Chk2. Mol Biol Cell 16, 1513–1526.
embryonic stem cells in which the gene for DNA-repair, 69 Hirose Y, Katayama M, Stokoe D, Kogan DAH, Ber-
methyltransferase, had been disrupted by gene targeting. ger MS & Pieper RO (2003) The p38 mitogen-activated
Carcinogenesis 18, 889–896. protein kinase pathway links the DNA mismatch repair
56 Ko LJ & Prives C (1996) P53: puzzle and paradigm. system to the G2 checkpoint and to resistance to che-
Genes Dev 10, 1054–1072. motherapeutic DNA-methylating agents. Mol Cell Biol
57 Johnstone RW, Ruefli AA & Lowe SW (2002) Apopto- 23, 8306–8315.
sis: a link between cancer genetics and chemotherapy. 70 Brown KD, Rathi A, Kamath R, Beardsley DI, Zhan
Cell 108, 153–164. Q, Mannino JL & Baskaran R (2003) The mismatch
58 Flores ER, Tsai KY, Crowley D, Sengupta S, Yang A, repair system is required for S-phase checkpoint activa-
McKeon F & Jacks T (2002) P63 and p73 are required tion. Nat Genet 33, 80–84.
for p53-dependent apoptosis in response to DNA 71 Wang Y & Qin J (2003) MSH2 and ATR form a signal-
damage. Nature 416, 560–564. ing module and regulate two brnaches of the damage
59 Jost C, Marin M & KaelinW (1997) p73 is a human response to DNA methylation. Proc Natl Acad Sci USA
p53-related protein that can induce apoptosis. Nature 100, 15387–15392.
389, 191–194. 72 Fang Y, Tsao CC, Goodman BK, Furumai R, Tirado
60 Duckett DR, Bronstein SM, Taya Y & Modrich P CA, Abraham RT & Wang XF (2004) ATR functions
(1999) hMutSa and hMutLa-dependent phosphorylation as a gene dosage-dependent tumor suppressor on a mis-
of p53 in response to DNA methylator damage. Proc match repair-deficient background. EMBO J 23, 3164–
Natl Acad Sci USA 96, 12384–12388. 3174.
61 Peters AC, Young LC, Maeda T, Tron VA & Andrew 73 Wang Y, Cortez D, Yazdi P, Neff N, Elledge SJ &
SE (2003) Mammalian DNA mismatch repair protects Qin J (2000) BASC, a super complex of BRCA1-asso-
cells from UVB-induced DNA damage by facilitating ciated proteins involved in the recognition and
apoptosis and p53 activation. DNA Repair (Amster- repair of aberrant DNA structures. Genes Dev 14,
dam) 2, 427–435. 927–939.
62 Gong JG, Costanzo A, Yang HQ, Melino G, Kaelin 74 Chen J & Sadowski I (2005) Identification of the mis-
WG Jr, Levrero M & Wang JY (1999) The tyrosine match repair genes PMS2 and MLH1 as p53 target
kinase c-Abl regulates p73 in apoptosis response to cis- genes by using serial analysis of binding elements. Proc
platin-induced DNA damage. Nature 399, 806–809. Natl Acad Sci USA 102, 4813–4818.
63 Shimodaira H, Yamashita AY, Kolodner RD & Wang 75 Luo Y, Lin F & Lin W (2004) ATM-mediated stabiliza-
JYJ (2003) Interaction of mismatch repair protein tion of hMutL DNA mismatch repair proteins aug-
PMS2 and the p53-related transcription factor p73 in ments p53 activation during DNA damage. Mol Cell
apoptosis response to cisplatin. Proc Natl Acad Sci Biol 24, 6430–6444.
USA 100, 2420–2425. 76 Tonegawa S (1983) Somatic generation of antibody
64 Kat A, Thilly WG, Fang WH, Longley MJ, Li GM & diversity. Nature 302, 575–581.
Modrich P (1993) An alkylation-tolerant, mutator 77 MacLennan IC (1994) Germinal centers. Annu Rev
human cell line is deficient in strandspecific mismatch Immunol 12, 117–139.
repair. Proc Natl Acad Sci USA 90, 6424–6428. 78 Rada C, Ehrenstein MR, Neuberger MS & Milstein C
65 Nyberg KA, Michelson RJ, Putnam CW & Weinert TA (1998) Hot spot focusing of somatic hypermutation in
(2002) Toward maintaining the genome: DNA damage MSH-deficient mice suggests two stages of mutational
and replication checkpoints. Annu Rev Genet 36, 617– targeting. Immunity 9, 135–141.
656. 79 Wiesendanger M, Kneitz B, Edelmann W & Scharff
66 Cejka P, Stojic L, Mojas N, Russell AM, Heinimann K, MD (2000) Somatic mutation in MSH3, MSH6, and
Cannavo E, di Pietro M, Marra G & Jiricny J (2003) MSH3 ⁄ MSH6-deficient mice reveals a role for the

1618 FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS
S.-H. Jun et al. Various functions of mismatch repair proteins

MSH2-MSH6 heterodimer in modulating the base sub- 85 Bransteitter R, Pham P, Shcarff MD & Goodman MF
stitution pattern. J Exp Med 191, 579–584. (2003) Activation-induced cytidine deaminase deami-
80 Li Z, Scherer SJ, Ronai D, Ussel MDI, Peled JU, Bar- nates deoxycytidine on single-stranded DNA but
dwell PD, Zhuang M, Lee K, Martin A, Edelmann W requires the action of Rnase. Proc Natl Acad Sci USA
et al. (2004) Examination of Msh6- and Msh3-deficient 100, 4102–4107.
mice in class switching reveals overlapping and distinct 86 Chaudhuri J, Tian M, Khuong C, Chua K, Pinaud E &
roles of MutS homologues in antibody diversification. Alt FW (2003) Transcription-targeted DNA deamina-
J Exp Med 200, 47–59. tion by the AID antibody diversification enzyme. Nature
81 Martin A, Li Z, Lin D, Bardwell PD, Iglesias-Ussel 422, 726–730.
MD, Edelmann W & Scharff MD (2003) Msh2 ATPase 87 Zeng X, Winter DB, Kasmer C, Kraemer KH, Lehmann
activity is essential for somatic hypermutation at A-T AR & Gearhart PJ (2001) DNA polymerase eta is an A-T
basepairs and for efficient class switch recombination. mutator in somatic hypermutation of immunoglobulin
J Exp Med 198, 1171–1178. variable genes. Nat Immunol 2, 537–541.
82 Muramatsu M, Kinoshita K, Fagarasan S, Yamada S, 88 Wilson TM, Vaisman A, Martomo SA, Sullivan P, Lan
Shinkai Y & Honjo T (2000) Class switch recombina- L, Hanaoka F, Yasui A, Woodgate R & Gearhart PJ
tion and hypermutation require activation-induced cyti- (2005) MSH2–MSH6 stimulates DNA polymerase g,
dine deaminase (AID), a potential RNA editing suggesting a role for A: T mutations in antibody genes.
enzyme. Cell 102, 553–563. J Exp Med 201, 637–645.
83 Pham P, Bransteitter R, Petruska J & Goodman M 89 Bardwell PD, Woo CJ, Wei K, Li Z, Martin A, Sack
(2003) Processive AID-catalysed cytosine deamination SZ, Parris T, Edelmann W & Scharff MD (2004)
on single-stranded DNA simulates somatic hypermuta- Altered somatic hypermutation and reduced class-switch
tion. Nature 424, 103–107. recombination in exonuclease 1-mutant mice. Nat
84 Storb U (1998) Progress in understanding the mechan- Immunol 5, 224–229.
ism and consequences of somatic hypermutation.
Immunol Rev 162, 5–11.

FEBS Journal 273 (2006) 1609–1619 ª 2006 The Authors Journal compilation ª 2006 FEBS 1619

You might also like