• No results found

VU Research Portal

N/A
N/A
Protected

Academic year: 2021

Share "VU Research Portal"

Copied!
19
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

VU Research Portal

Functional analysis of MSH2/MSH6 variants: In tune or off key?

Wielders, E.A.L.

2016

document version

Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)

Wielders, E. A. L. (2016). Functional analysis of MSH2/MSH6 variants: In tune or off key?.

General rights

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain

• You may freely distribute the URL identifying the publication in the public portal ?

Take down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address:

(2)

Chapter 5

The MSH2 C-terminal 60 amino acids are

essential for interaction with MSH6 and

effective DNA mismatch repair

(3)

104

(4)

5

Introduction

Lynch syndrome (LS) is characterized by a strong predis-position to tumor development in the gastrointestinal and genitourinary tracts 1. Cancer predisposition is caused

by inherited defects in the DNA mismatch repair (MMR) system that counteracts erroneous incorporation of nucleotides during DNA replication 2,3.

Besides the correction of replication errors, MMR has two other functions. MMR activity mediates the toxicity of certain types of DNA damaging agents, in particular methylating compounds4,5,

and it prevents recombination between sequences that are homologous but not fully identical 6,7. Thus, the absence of

MMR is characterized by increased spontaneous and DNA damage induced mutagenesis, tolerance to methylating agents and increased recombination between homologous but not identical DNA sequences 6. Each of these may

contribute to early onset tumor development in LS patients. Such

tumors are characterized by microsatellite instability, i.e., the expansion or contraction of repetitive sequences like (A)n or (CA)n (n is an

integer, generally between 5-50), as DNA polymerase slippage errors that are frequently made at microsatellites are no longer recognized and repaired by MMR 2,3.

The first step in MMR is the recognition of the mismatch by a heterodimeric protein complex composed of the central MMR protein MSH2 and either MSH6 or MSH3 8.

While MSH2/MSH6 (MutSα) pre-dominantly recognizes base-base mismatches and unpaired single- or dinucleotides, MSH2/MSH3 (MutSβ) targets loops of 2-5 nucleotides 9,10.

(5)

106

5

a new, error-free strand completes the MMR reaction.

The majority of MMR gene mutations in LS families affects MSH2 or MLH1 and comprises defects that are obviously deleterious for gene function, such as deletions and frame-shifts. However, missense mutations that only affect a single amino acid have also been detected in the human population. In the absence of functional information about the phenotypic consequences, it is often difficult to determine whether a missense mutation really impacts MMR activity and is the underlying cause of cancer. To study such Variants of Uncertain clinical Significance (VUS; also termed unclassified variants, UV), we have developed a functional assay in which the variant allele is introduced into the endogenous MMR gene of mouse embryonic stem cells (ESCs). Mutant cells are then subjected to a number of functional assays, allowing the unambiguous assessment of the cellular consequences of a MMR gene variant 11.

Here we have used this protocol to study a truncation mutant of mouse

MSH2 that causes deletion of the 60

terminal amino acids downstream of codon 876. This variant is of interest for two reasons. First, in several suspected LS families a 2 base-pair deletion has been detected in the sequence AGAGAG that comprises codons 877 and 878 12-17. The reading

frame shift causes a premature stop codon at codon 880 deleting the 57 C-terminal amino acids (Figure 1A).

These families generally showed co-segregation of the mutation with disease and a high incidence of early onset MSI tumors that often showed loss of MSH2 staining. Second, the biochemical function and significance of the MSH2 C-terminus is unclear. In the 3-D structure of MutSα, the C-termini of MSH2 and MSH6 appeared disordered and therefore their contribution to the MSH2/MSH6 interaction could not be determined 18.

However, the MutSβ crystal structure did reveal the C-termini of MSH2 and MSH3 19. These regions formed

α-helices, three in MSH2 and two in MSH3, which contributed to dimer stabilization by hydrophobic inter-actions and salt bridges (Supple-mentary Figure S1). Besides these interactions, each of these dimerization domains (DMD) also interacts with the ATPase domain of the opposite subunit, which is thought to influence ATP binding 19. To solve the structure of the

853 amino-acids-containing MutS protein of Escherichia coli, it was necessary to truncate the protein at codon 800 to obtain crystals 20. The 53

C-terminal amino acids of MutS were found to promote tetramer formation that negatively interfered with protein crystallization. The truncated protein MutS-ΔC800 formed only dimers that showed normal ATPase activity, recognized DNA mismatches as effectively as wild-type protein, and could efficiently restore mismatch repair activity in a MutS-deficient E. coli strain 20. However, others did report a

(6)

5

ΔC800 expressing E. coli, in particular reduced capacity to prevent recombination between homologous but not-identical DNA sequences 21,22. A

structure of the MutS C-terminus has been solved 22 and showed two

α-helices that can roughly be superimposed on the MSH3 C-terminal α-helices (despite poor sequence conservation) and may thus form similar interactions to stabilize the full-length protein dimer 19.

We show here that deletion of the MSH2 C-terminus severely affected the stability of the MSH2/MSH6 hetero-dimer and consequently strongly attenuated DNA MMR. The C-terminal truncation MSH2 mutant predisposed mice to tumor formation albeit slightly less severely than when MSH2 was fully abrogated. Mutations deleting the MSH2 C-terminus can therefore unambiguously be considered as pathogenic and as a cause of LS.

Figure 1. Generation of a C-terminal truncation of MSH2. (A) C-terminal sequences of E. coli MutS, and human and mouse MSH2. Deleted amino acids in E. coli, found in LS patients, and generated in mouse ESCs are indicated in red. Asterisks indicate identity between human and mouse MSH2 (red) or all three proteins (black). (B) Oligonucleotide (ssODN) used to insert TAAA (left panel) and sequence of the E877X insertion allele (right panel). (C) Oligonucleotide (ssODN) used to substitute GAA for TGA. An additional silent A->C substitution was added to increase the targeting efficiency (left panel). Sequence of the E877X substitution allele is shown on the right.

R K X L

Msh2+ allele Msh2E877Xi allele

B

R E X L

C

R E E L

Msh2+ allele Msh2E877Xs allele exon 16

exon 15

*

A

(7)

108

5

Results

Generation of MSH2 truncation mutants

To study the consequences of an MSH2 truncation mutant resembling those found in LS families (Figure 1A), we introduced a stop codon in Msh2 at codon position 877 in murine embryonic stem cells (ESCs). This was done in two ways. Initially, we inserted a TAAA sequence just behind leucine 876 by oligonucleotide-directed gene modification as described before 23.

This mutant was made in Msh3-/- ESCs,

as this effectively allowed 4-nucleotide insertions, and designated MSH2-E877Xi (i indicates insertion but effectively codon 877 was replaced for a TAA stop codon; Figure 1B). However, since the absence of MSH3 may affect the phenotype of the MSH2 truncation, we also made a similar mutation in wild-type ESCs by substituting the E877 codon for a TGA stop codon, generating E877Xs (s indicates substitution, Figure 1C). Both mutations were rendered homozygous as described in the Methods section. Furthermore, in MSH2-E877Xs cells,

Msh6 was inactivated using conventional gene targeting 24. We

subsequently studied the phenotype of the truncated MSH2 protein in three different ESC lines: Msh2EXs/EXs,

Msh2EXs/EXs;Msh6-/- and Msh2EXi/EXi;

Msh3-/- (E877X is abbreviated as EX).

MSH2 truncation affects MSH6 and MSH3 binding

Deleting 60 C-terminal amino acids from MSH2 generated a shorter protein

that was present at lower levels than wild-type MSH2 protein (Figure 2A). The insertion and the substitution mutants showed the same protein size and level and therefore the indications i and s are omitted hereafter. In both

Msh2EX/EX and Msh2EX/EX;Msh3-/- mutant

cells, MSH6 levels were strongly reduced albeit slightly elevated above the MSH6 level seen in Msh2-/- cells.

This indicates that the truncation severely destabilized MSH2/MSH6 interaction. MSH3 levels were also reduced in Msh2EX/EX and

Msh2EX/EX;Msh6-/- ESCs, but

approxi-mately to the same extent as the mutant MSH2 proteins, suggesting truncated MSH2 was still capable of binding and stabilizing MSH3 (Figure 2A). However, whereas wild-type MSH2 stabilized MSH3 more effectively in Msh6-/- cells than in Msh6+/+ cells

(which is indicative for competition between MSH3 and MSH6 for MSH2 binding), this was not seen for the MSH2-E877X variant: disruption of

Msh6 in Msh2EX/EX cells did not increase

the level of MSH3 protein (Figure 2A). Also MSH6 levels were not increased in

Msh2EX/EX;Msh3-/- cells.

(8)

5

Msh 2 +/+ Msh 2 EX/ + Msh 6 -/ -Msh 3 -/ -Msh 2 -/ -MSH3 MSH2 γ-Tubulin MSH6 Msh2 EX/EX Msh 6 -/ -Msh 3 -/ -WT EX ARP WT EX ARP MSH2-HIS-tag IP Total lysate 2 -/-MSH2 - HIS MSH2 MSH3 MSH6 MSH6 MSH2 - HIS 2-/-3 -/-2 -/-MSH2 A B

Figure 2. MSH heterodimer formation in mutant ESCs. (A) Western blot showing MSH2, truncated MSH2, MSH3 and MSH6 protein levels in mutant ESCs. γ-Tubulin was used as loading control. (B) Western blot showing MSH3 and MSH6 protein levels co-precipitated with His-tagged MSH2-WT or

MSH2-E877X expressed in Msh2-/- or Msh2

-/-;Msh3-/- ESCs (upper panel). His-tagged ARP

and no expression vector (-) were used as controls. Expression of WT and MSH2-E877X in total cell lysates are shown in the lower panel.

Functional consequences of MSH2 truncation

The capacity of MSH2-E877X protein to support mismatch repair was assessed using four functional assays.

Microsatellite instability (MSI)

An estimate of the level of MSI was obtained by measuring the length of three dinucleotide-repeat markers in 30 individual cell clones obtained from 109 cells that were derived from a

single cell of each genotype. Figure 3A shows strongly increased MSI in

Msh2EX/EX cells compared to wild-type

cells, although the level remained 3-fold lower compared to Msh2-/- cells,

indicating some residual MMR capacity of MSH2-E877X protein. Furthermore, in the absence of either MSH6 or MSH3, MSI increased, but remained lower than in Msh2-/- cells. This indicates that

residual MSH2-E877X/MSH6 and MSH2-E877X/MSH3 complexes exist, which both have retained a modest capacity to restore dinucleotide slippage errors.

In Msh3-/- cells, we could make use

of an alternative strategy to measure MSI, as we had previously introduced into these cells a reporter gene consisting of a (CA)15C sequence

disrupting the reading frame of a neomycin resistance gene (neo) 9. The

appearance G418-resistant colonies due to the addition of CA or deletion of (CA)2 restoring neo activity served as

readout for MMR activity. The slippage rate in Msh2EX/EX;Msh3-/- mutant cells

(9)

110

5

0 10 20 30 40 0 2 4 6 8 10 12 14 MSI HPRT u n st a b le mi cro sa te lli te s (% ) 6 -T G re si st a n t co lo n ie s (1 0 ) -6 co rre ct ly ta rg e te d co lo n ie s (% ) 0 10 20 30 40 50 60 100% homologous 99.4% homologous Msh 2+/ + Msh 2 -/-Msh 3 Msh 6 Msh 3 Msh 6 -/- -/- -/- -/-Msh2EX/EX Msh 2+/ + Msh 2 -/-Msh 3 Msh 6 Msh 3 -/- -/- -/-Msh2EX/EX 6-TG concentration (μM) 0,01 0,1 1 100 su rvi vi n g co lo n ie s (% ) 0 20 40 60 80 100 su rvi vi n g co lo n ie s (% ) 0 20 40 60 80 100 0,01 0,1 1 10 100 10 A B C D MNNG concentration (μM) E 40 35 30 25 20 15 10 5 0 mu ta ti o n ra te (x1 0 )-5 Msh 3 -/-Msh 3 -/-Msh 3 -/-Msh 6 -/-Msh 2 EX/E X Msh2+/+ Msh2 -/-Msh3 -/-Msh2 +/-Msh2EX/EX Msh2EX/EX Msh3 -/-Msh2EX/+ Msh3 -/-Msh2EX/+ Msh2+/+ Msh2 -/-Msh3 -/-Msh2 +/-Msh2EX/EX Msh2EX/EX Msh3 -/-Msh2EX/+ Msh3 -/-Msh2EX/+

Figure 3. Mutator phenotype of mutant ESCs. (A) Frequencies of replication errors in mutant ESCs. Black bars indicate the percentage of microsatellites with altered length (left Y-axis). Grey bars show the frequency of 6-TG-resistant colonies indicative for Hprt mutations (right Y-axis). (B) Suppression of recombination between homologous but non-identical DNA sequences. Black bars indicate targeting efficiencies of a 100% identical gene-targeting vector; white bars indicate the efficiencies of the non-isogenic vector differing from the target

sequence at 0.6% of its base pairs. Targeting efficiencies in Msh3-/-, Msh2-/- and Msh6-/- cells

were taken from De Wind et al. 1995 6 and De Wind et al. 1999 24 and shown as controls.

(C) Clonogenic survival of wild-type and mutant ESCs upon exposure to increasing concentrations of MNNG. (D) Clonogenic survival of wild-type and mutant ESCs upon exposure

to increasing concentrations of 6-TG. (E) Slippage rate at a (CA)15C microsatellite sequence

(10)

5

Msh3-/- cells but was five-fold lower

than in fully MMR-deficient

Msh6-/-;Msh3-/- cells (Figure 3E). This

experiment confirms retention of some MMR capacity of residual MSH2-E877X/MSH6 protein.

Mutagenesis at the Hprt gene

Inactivation of the X-linked Hprt gene confers resistance to the nucleotide analogue 6-thioguanine (6-TG). Hence, the appearance of 6-TG-resistant colonies serves as readout for the rate of spontaneous mutagenesis. MSH2-E877X protein increased the mutation frequency at Hprt with respect to wild-type MSH2 protein, but again to a level that was approximately 4-fold lower as seen in fully MSH2-deficient cells (Figure 3A). Concomitant inactivation of Msh6, but not Msh3, increased the mutation frequency back to the level seen in Msh2-/- cells. Thus, also in this

assay, MSH2-E877X/MSH6 had retained some capacity to suppress spontaneous mutagenesis.

MMR-dependent anti-recombination

We have previously shown that MMR is capable of suppressing homologous recombination in a gene targeting experiment where the targeting vector differs ±0.6% from its chromosomal target sequence6. Thus, MMR

sup-pressed non-isogenic gene targeting 35-fold, whereas in Msh2-/- or Msh6

-/-cells, the non-isogenic vector performed as effectively as the perfect-match vector (Figure 3B). No relieve of suppression was seen in Msh3-/- cells,

demonstrating that MMR-dependent anti-recombination solely relied on MSH2/MSH6 activity. Strikingly, in

both Msh2EX/EX and Msh2EX/EX;Msh3

-/-ESCs, non-isogenic targeting was suppressed, albeit less effectively than in MSH2 wild-type cells. This result indicates some anti-recombination activity of residual MSH2-E877X/MSH6 complex.

Response to DNA methylating agents

We previously showed that cell death in response to the methylating agents N-methyl-N’-nitro-N-nitrosoguanidine (MNNG) and 6-TG relied on MSH2/MSH6 but not MSH2/MSH3 activity 24. Indeed, we found high

sensitivity of ESCs expressing wild-type MSH2 protein, with modest haploinsufficiency. In contrast, Msh2

-/-cells were highly tolerant to MNNG and 6-TG as were all cell types expressing MSH2-E877X protein (Figure 3C and 3D). Apparently, the level and/or activity of MSH2-E877X/MSH6 was not sufficient to mediate cell death in response to methylating agents.

Msh2E877X/+ cells behaved identical to

Msh2+/- cells, indicating that

MSH2-E877X did not act dominant negatively. Tumor development in MSH2-E877X mice

The Msh2E877Xi allele was introduced

into the germ line of mice and cohorts of Msh2EX/EX, Msh2EX/-, Msh2EX/+, Msh2

+/-and Msh2-/- mice were generated and

compared for spontaneous tumor development. Msh2EX/EX and Msh2

EX/-mice were strongly predisposed to development of thymic lymphoma, although the incidence appeared somewhat lower than in Msh2

(11)

112

5

Msh2EX/- mice, respectively, and 87% in

Msh2-/- mice (Table 1).

The latency of tumor development in MSH2-EX mice was somewhat longer than in Msh2-/- mice (Figure 4). Also the

incidence of intestinal tumors appeared lower: 0, 8 and 17% in Msh2EX/EX,

Msh2EX/- and Msh2-/- mice, respectively.

Taken together, the Msh2EX allele

strongly predisposed to tumor development although some residual MMR activity may retard tumorigenesis and suppress intestinal tumor development, in particular when two copies of the Msh2EX allele were

present.

Table 1. Tumor incidence in Msh2-mutant-mice

Genotype Number of

mice

Number of animals with tumor

thymus intestine skin Other

Msh2-/- 30 26 5 1 1

Msh2EX/- 25 18 2 2 3

Msh2EX/EX 25 15 0 1 2

Msh2EX/+ 25 0 1 1 5

Msh2+/- 23 0 1 2 10

Figure 4. MSH2-E877X predisposes to tumorigenesis.

Kaplan-Meier curves of cohorts of Msh2+/- (n=23), Msh2-/- (n=30), Msh2EX/+ (n=25), Msh2EX/EX

(n=25) and Msh2EX/- (n= 25). Animals were sacrificed upon signs of illness and examined for

tumor development.

Survival time (days)

(12)

5

Discussion

Our results clearly demonstrate that the 60 C-terminal amino acids of MSH2 are critical for heterodimer formation, in particular with MSH6. Since the stability of MSH6 and MSH3 relies on their interaction with MSH2, the strongly reduced level of MSH6 in MSH2-E877X expressing cells on Western blots is indicative for poor interaction. The interaction with MSH3 appeared less affected by MSH2 truncation. While in the MSH2/MSH6 crystal structure the C-termini of both proteins were not visible, and hence their contribution to dimer formation could not be determined 18, the

MSH2/MSH3 structure revealed the C-termini of both proteins to be involved in dimerization via interacting α-helices19. Based on secondary structure

predictions, it is likely that also in MutSα the C-termini of MSH2 and MSH6 contribute to dimer formation. Our results indicate that these interactions are more critical in MutSα than in MutSβ.

Two of our assays, those measuring tolerance to methylating DNA damaging agents and homologous recombination between slightly diverged DNA sequences, solely measured the activity of MSH2/MSH6 but not of MSH2/MSH3, as the latter complex does not play a role in these two MMR functions. We previously showed that Msh6-/- cells were tolerant

to methylating agents and permissive for non-isogenic gene targeting, whereas Msh3-/- cells behaved as

wild-type 24. Therefore, in Msh2EX/EX cells,

these two assays reflect the activity of the MSH2-E877X/MSH6 heterodimer. As summarized in Table 2, all cell types solely expressing MSH2-E877X were tolerant to 6-TG and MNNG. However, we previously reported the existence of a threshold level of wild-type MSH2 that was required for mediating toxicity of methyl ting agents. This threshold has been found to lie above 10% of wild-type level 25 and the level of

MSH2-E877X/MSH6 was clearly lower. Thus, tolerance to 6-TG/MNNG may

Table 2. Functional assessment of MSH2-E877X

Genotype MSH dimer MSI MNNG Hprt Homologous recombinatio

2/3a 2/6a (%) /6-TGb (10-6) iso/non-iso Repression WT >>>> >>>> 1 S 0.5 35/1c 35 x Msh2-/- (3+/6+) 0 0 32 R 10 22/25c 0.9 x EXs (3+/6+) >> > 10 R 3 52/15 3.5 x EXs (3+/6-) >> 0 17 (nd) 9 (nd) (nd) EXi (3-/6+) 0 > 19 R 1.5 26/3 8.7 x Msh6-/- >>>> 0 6 R 0 18/18c 1.0 x Msh3-/- 0 >>>> 2 S 2 50/0c 50 x

a > indicates level of 2/3 or 2/6 complex compared to wild-type (>>>>) b S, sensitive; R. resistant

(13)

114

5

be due to low MSH2-E877X/MSH6 levels and does not exclude (residual) MMR activity. Evidence for residual MSH2-E877X/MSH6 activity came from the recombination experiment: recom-bination with the non-isogenic targeting vector was still 3.5x repressed in

Msh2EX/EX cells (as opposed to 0.9x in

Msh2-/- cells). We expected that in

Msh3-/- cells, the level of

MSH2-E877X/MSH6 dimer would increase, however, this was not confirmed by Western blotting. Nonetheless, in

Msh3-/- cells, anti-recombination activity

of MSH2-E877X/MSH6 dimer was increased (from 3.5x to 8.7x), suggesting that the formation of MSH2-E877X/MSH6 dimers did benefit from the absence of MSH3. Why the increased anti-recombination activity of MSH2-E877X/MSH6 in Msh3-/- cells was

not reflected by increased MSH6 protein levels in Msh2EX/EX;Msh3-/- cells

is unclear. Also in the immune-precipitation experiment, MSH2-E877X protein did not bind more MSH6 in cells devoid of MSH3 (Figure 2B). Perhaps, stable MSH2-E877X/MSH6 dimers can only be formed in the presence of mismatched DNA, explaining their residual activity but precluding their detection by the methods we used.

Consistent with previous experiments10, dinucleotide slippage

errors can be restored by MSH2/MSH6 as well as MSH2/MSH3 (Table 2). Truncation of MSH2 increased the dinucleotide slippage frequency from 2 to 19% in the absence of MSH3, and from 6 to 17% in the absence of MSH6. As full abrogation of MMR in Msh2

-/-cells caused a slippage frequency of 32%, both MSH2-E877X/MSH6 and MSH2-E877X/MSH3 had residual capacity to restore dinucleotide slippage errors; although the activity of these complexes was modest, the activity of MSH2-E877X/MSH3 appeared somewhat higher than of MSH2-E877X/MSH6. Residual MMR activity of the MSH2-E877X/MSH6 dimer was confirmed by measuring slippage rates at the (CA)15C-neo

reporter in Msh3-/- cells. The Hprt

mutagenesis assay also supports some mismatch repair capacity of MSH2-E877X/MSH6, although the figures were too low to draw a firm conclusion. The functional assays identified the MSH2 truncation mutant as severely affecting, but not completely abolishing DNA MMR activity. Experiments in mice were consistent with this conclusion. MSH2-E877X-expressing mice were strongly predisposed to lymphoid tumor development, but the incidence of tumor development was lower and the time of onset somewhat later than in Msh2-/- animals. In this respect,

Msh2EX/EX mice resembled Msh6

-/-mice24: in both genotypes the incidence

(14)

5

The protocol we have used here and in previous reports, i.e., introducing a MMR mutation at the endogenous gene in mouse ESCs followed by functional assays that interrogate cellular MMR functions, provides unambiguous information about the pathogenicity of MMR VUS. This approach will be much facilitated and accelerated by novel gene modification technology making use of template-directed repair of site-specific DNA double-strand breaks induced by CRISPR/Cas9 nuclease.

Acknowledgements

We thank dr. Allan Bradley for providing the Msh2-/-;Msh3-/- ESC line,

(15)

116

5

Materials and Methods

Generation of mutant embryonic stem cells

Mouse MSH2 protein was truncated just before the glutamic acid at position 877 by introduction of TAAA, thereby deleting the 60 C-terminal amino acids from the protein. This mutant allele, Msh2EXi, was made in Msh3-/- embryonic stem cells (ESCs), as this effectively

allowed insertions by oligonucleotide-directed gene modification 9. A homozygous cell line

was subsequently obtained by labeling the targeted chromosome with a hygromycin (hyg) resistance marker using a Pim1-hyg targeting vector. This allowed for duplication of the targeted allele and concomitant loss of the wild-type allele by selecting cells at high concentrations of Hygromycin B, similarly as described before 11.

In addition, we substituted glutamic acid 877 for a TGA stop codon in wild-type ESCs. The

Msh2EXs allele was rendered homozygous by using the strategy as described above. In Msh2EXs/EXs cells, Msh6 was disrupted by conventional gene targeting 24 and rendered

homozygous by labeling the targeted chromosome with a neomycin (neo) maker using a

Pim1-neo targeting vector 11.

Functional MMR assays

Mutant ESCs were subjected to four functional assays as described in Wielders et al. 11.

Slippage rates in Msh3-/-, Msh3-/-;Msh6-/- and Msh3-/-;Msh2EX/EX cells were measured using the

single-copy (CA)15C-neo reporter gene integrated at the Rosa26 locus, as described in

Dekker et al. 9.

Immunoprecipitation

His-tagged cDNAs of full length and truncated Msh2 were introduced into Msh2-/- and

Msh2-/-;Msh3-/- ESCs (the latter a kind gift of dr. Allen Bradley). Proteins co-precipitating

with an anti-His antibody were visualized by Western blotting as described 11.

Mouse experiment

(16)

5

References

1 Lynch, H. T. & Lynch, J. Lynch syndrome: genetics, natural history, genetic counseling, and prevention. J Clin Oncol 18, 19S-31S (2000).

2 Ionov, Y., Peinado, M. A., Malkhosyan, S., Shibata, D. & Perucho, M. Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature 363, 558-561, doi:10.1038/363558a0 (1993).

3 Nash, G. M. et al. Automated, multiplex assay for high-frequency microsatellite instability in colorectal cancer. J Clin Oncol 21, 3105-3112, doi:10.1200/JCO.2003.11.133 (2003).

4 Karran, P. Mechanisms of tolerance to DNA damaging therapeutic drugs. Carcinogenesis 22, 1931-1937 (2001).

5 Stojic, L. et al. Mismatch repair-dependent G2 checkpoint induced by low doses of SN1 type methylating agents requires the ATR kinase. Genes Dev 18, 1331-1344, doi:10.1101/gad.29440418/11/1331 (2004).

6 de Wind, N., Dekker, M., Berns, A., Radman, M. & te Riele, H. Inactivation of the mouse Msh2 gene results in mismatch repair deficiency, methylation tolerance, hyperrecombination, and predisposition to cancer. Cell 82, 321-330, doi:0092-8674(95)90319-4 (1995).

7 Rayssiguier, C., Thaler, D. S. & Radman, M. The barrier to recombination between Escherichia coli and Salmonella typhimurium is disrupted in mismatch-repair mutants. Nature 342, 396-401, doi:10.1038/342396a0 (1989).

8 Gradia, S. et al. hMSH2-hMSH6 forms a hydrolysis-independent sliding clamp on mismatched DNA.

Molecular cell 3, 255-261 (1999).

9 Dekker, M. et al. Transient suppression of MLH1 allows effective single-nucleotide substitution by single-stranded DNA oligonucleotides. Mutation research 715, 52-60, doi:10.1016/j.mrfmmm.2011.07.008 (2011).

10 Umar, A. et al. Functional overlap in mismatch repair by human MSH3 and MSH6. Genetics 148, 1637-1646 (1998).

11 Wielders, E. A., Dekker, R. J., Holt, I., Morris, G. E. & te Riele, H. Characterization of MSH2 variants by endogenous gene modification in mouse embryonic stem cells. Hum Mutat 32, 389-396, doi:10.1002/humu.21448 (2011).

12 Bianchi, F. et al. Effectiveness of the CRCAPRO program in identifying patients suspected for HNPCC. Clinical genetics 71, 158-164, doi:10.1111/j.1399-0004.2007.00746.x (2007).

13 Durno, C., Aronson, M., Bapat, B., Cohen, Z. & Gallinger, S. Family history and molecular features of children, adolescents, and young adults with colorectal carcinoma. Gut 54, 1146-1150, doi:10.1136/gut.2005.066092 (2005).

14 Millar, A. L. et al. Mismatch repair gene defects contribute to the genetic basis of double primary cancers of the colorectum and endometrium. Hum Mol Genet 8, 823-829 (1999).

15 Miyaki, M. et al. Germ line mutations of hMSH2 and hMLH1 genes in Japanese families with hereditary nonpolyposis colorectal cancer (HNPCC): usefulness of DNA analysis for screening and diagnosis of HNPCC patients. Journal of molecular medicine 73, 515-520 (1995).

16 Terdiman, J. P. et al. Efficient detection of hereditary nonpolyposis colorectal cancer gene carriers by screening for tumor microsatellite instability before germline genetic testing. Gastroenterology

120, 21-30 (2001).

17 Yuan, Y., Han, H. J., Zheng, S. & Park, J. G. Germline mutations of hMLH1 and hMSH2 genes in patients with suspected hereditary nonpolyposis colorectal cancer and sporadic early-onset colorectal cancer. Dis Colon Rectum 41, 434-440 (1998).

18 Warren, J. J. et al. Structure of the human MutSalpha DNA lesion recognition complex. Molecular

cell 26, 579-592, doi:10.1016/j.molcel.2007.04.018 (2007).

19 Gupta, S., Gellert, M. & Yang, W. Mechanism of mismatch recognition revealed by human MutSbeta bound to unpaired DNA loops. Nature structural & molecular biology 19, 72-78, doi:10.1038/nsmb.2175 (2012).

20 Lamers, M. H. et al. The crystal structure of DNA mismatch repair protein MutS binding to a G x T mismatch. Nature 407, 711-717, doi:10.1038/35037523 (2000).

(17)

118

5

22 Mendillo, M. L., Putnam, C. D. & Kolodner, R. D. Escherichia coli MutS tetramerization domain structure reveals that stable dimers but not tetramers are essential for DNA mismatch repair in vivo. J Biol Chem 282, 16345-16354, doi:10.1074/jbc.M700858200 (2007).

23 Dekker, M. et al. Effective oligonucleotide-mediated gene disruption in ES cells lacking the mismatch repair protein MSH3. Gene therapy 13, 686-694, doi:10.1038/sj.gt.3302689 (2006). 24 de Wind, N. et al. HNPCC-like cancer predisposition in mice through simultaneous loss of Msh3 and

Msh6 mismatch-repair protein functions. Nat Genet 23, 359-362, doi:10.1038/15544 [doi] (1999). 25 Claij, N. & Te Riele, H. Methylation tolerance in mismatch repair proficient cells with low MSH2

(18)

5

Supplementary information

Supplementary Figure 1. Three-dimensional structure of MSH2/MSH3 dimer. The image is based on

Gupta et al. 19 and shows MSH2 on the right and MSH3 on the left. The brown and grey helices at the

(19)

Referenties

GERELATEERDE DOCUMENTEN

Oligonucleotide-directed mutation screening: a functional test to classify mismatch repair gene variants of uncertain significance..

Oligonucleotide-directed mutation screening: a functional test to classify mismatch repair gene variants of uncertain significance..

Special focus is required for professions that fall outside the scope of the Individual Health Care Professions Act (Wet BIG), such as carers, and where there is a lack of

Naast een inschatting van het toekomstig saldo is het dus noodzakelijk om uitgangspunten op te stellen voor de techni- sche resultaten, voerprijzen, opbrengstprijzen en toegere-

[twee blurbs van Clare Lennart en Willy Roggeman] Met deze herziene uitgave van haar debuut zijn nu alle boeken (...) in een nieuwe, door Meulenhoff verzorgde editie leverbaar:

Results of this study showed that these processes can also contribute to (the lack of) transparency during recruitment and selection processes because in multiple cases, it

Veldpodzolgronden; leemarm en zwak lemig fijn zand Kamppodzolgronden; leemarm en zwak lemig fijn zand Hoge zwarte enkeerdgronden; grof zand Akkereerdgronden; leemarm en zwak lemig

Als we dus naar de karakterisering van de vondelingen in de komedies Casina en Cistellaria van Plautus en Heautontimorumenos van Terentius kijken, zien we dat het telkens