Fibroblasts -deficient Murine Embryonic Msh2 Mutator ...

1 downloads 0 Views 1MB Size Report
Sep 1, 1997 - Fibroblasts. -deficient Murine Embryonic. Msh2. Mutator Phenotype in. Updated Version http://cancerres.aacrjournals.org/content/57/17/3765.
Mutator Phenotype in Msh2-deficient Murine Embryonic Fibroblasts Armin H. Reitmair, Robert Risley, Robert G. Bristow, et al. Cancer Res 1997;57:3765-3771. Published online September 1, 1997.

Updated Version

Citing Articles

E-mail alerts Reprints and Subscriptions Permissions

Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/57/17/3765

This article has been cited by 18 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/57/17/3765#related-urls

Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at [email protected]. To request permission to re-use all or part of this article, contact the AACR Publications Department at [email protected].

Downloaded from cancerres.aacrjournals.org on July 13, 2011 Copyright © 1997 American Association for Cancer Research

ICANCER RESEARCH57, 3765-3771. September I. 19971

Mutator Phenotype in Msh2-deficient

Murine Embryonic Fibroblasts'

Armin H. Reitmair,2 Robert Risley,2 Robert G. Bristow,2 Teresa Wilson,2 Ami Ganesh, Anne Jang, James Peacock, Samuel Benchimol, Richard P Hill, Tak W. Mak, Richard Fishel, and Mark Meuth3 Ontario Cancer Institute/Amgen Institute, Departments of Medical Biophysics and Immunology, University of Toronto, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada [A. H. R., T. W. M.]; Departments of Oncological Sciences and Radiation Oncology. Eccles Institute of Human Genetics, Universirv of Utah, Salt Lake City, Utah 84! 12 [R. R., A. G., M. MI: Ontario Cancer Insitute, MSG 2M9, Canada JR. G. B., A. J., R. P. Hi;

Departments of Medical Biophysics and Radiation Oncology, University of Toronto, 610 University DNA Repair and Molecular Carcinogenesis Program, Kimmel Cancer Center, Thomas Jefferson

Avenue, Toronto, Ontario University, Philadelphia,

Pennsylvania 19107 (T. W., R. Fl; and Ontario Cancer Institute, Department of Medical Biophysics. University of Toronto, 6/0 University Avenue. Toronto, Ontario MSG 2M9, Canada (J. P., S. B.J

ABSTRACT

copy is mutated or lost as an early event in the development of a tumor. This second event renders cells mismatch repair deficient (7, Embryonic fibroblast cell lines were established from mice deficient, 8), presumably leading to the rapid accumulation of mutations that heterozygous, or proficient for Msh2, one of the three known DNA mis drive tumor development. Two other genes encoding homologues of match repair genes involved in hereditary nonpolyposis colon cancer (HNPCC). Cell lines were established by transfection of primary mouse the E. coli mutS gene (hMSH3 and hMSH6) appear to be involved in the repair of some types of DNA replication errors or damage (9—11). embryo fibroblasts with E7 and Ras oncogenes or mutantp53. Spontane ously immortalized cells derived from the primary cultures were also Mutations in these genes have been found in some tumor cell lines, studied.To determinewhetherthesecellsdevelopeda mutatorphenotype but not HNPCC patients (12—14).Finally a fifth homologue of the similar to that found in colon cancer cells deficient in mismatch repair, we mismatch repair genes, hPMSJ, was identified based on its homology measured mutation rates, microsatellite instability, and sensitivities to a with MutL. However only one germline alteration of this gene has range of DNA-damaging agents. The mutator phenotype detected in the been identified and no apparent relationship to tumorigenesis has been E7 and Ras or mutantpS3-lmmortalized Msh2'-/mouse cells was sim reported (6). liar to that found in humanmismatchrepair-deficientcolorectalcard Cell lines originating from tumors deficient in mismatch repair aetna cell lines. Mutation rates to ouabain resistance were increased have a distinctive mutator phenotype. The rate of mutations that result 8-42-fold relative to lines from Msh2+/+ mice, and microsatellite mats in the loss or gain of repeat units in microsatellites is dramatically bility was detectable in 12—18%of subclones derived from the Msh2—/— increased (7, 15, 16). Studies from several laboratories provide strong line but was undetectable in subclones developed from the Msh2+I+ line. evidence that this microsatellite instability is diagnostic of the mis Furthermore, E7 and Ras or spontaneously immortalized Msh2—/- cells were significantly more resistant to the cytotoxic effects of 6-thioguanine match repair deficiency in colorectal carcinoma cell lines (7, 8). This relative to Msh2+/+ cells. In contrast, these lines showed various re DNA sequence instability is not limited to simple sequence repeat spouses to UV light and cis-platinum, suggesting that mismatch repair structures. Mismatch repair-deficient cell lines have a similar (60— deficiency was not the sole determinant for sensitivity to these DNA 600-fold) increase in the rate of spontaneous mutations at selectable damaging agents. Particular attention was paid to the properties of cells loci such as those encoding the punne salvage enzymes HPRT (16— heterozygous for the Msh2 mutant gene, which would mimic the situation 18) or APRT (19). of an HNPCC carrier. However, our studies failed to reveal any properties In addition to an increased rate of spontaneous mutation, mismatch of these cells that might provide a growth advantage or predispose them repair-deficient tumor cell lines are altered in their response to a wide for the acquisition of further mutations. This observation is consistent range of DNA-damaging agents. Mismatch repair-deficient colorectal with the model that Inactivation of the wild-type Msh2 allele is a critical step for tumorigenesis

in HNPCC

carcinoma

patients.

INTRODUCTION Recent work revealed a distinctive class of colorectal carcinomas having a high level of microsatellite repeat instability (1). In heredi tary forms of these cancers (HNPCC4 and Muir-Tone syndrome), microsatellite instability is largely associated with mutations in one of two human homologues of the Escherichia coli mismatch repair genes MutS and MutL (hMSH2, hMLHJ; Refs. 2—5).Rare germline muta tions of the MutL homologue hPMS2 also have been found associated with HNPCC (6). HNPCC patients inherit a mutation of one of the alleles encoding a mismatch repair gene, and the second wild-type

proteins Received 12/18/96; accepted 7/2/97. The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. I A. H. R.

was

supported

by

a stipend

of

the

German

Academic

Exchange

Service

(DAAD-Doktorandenstipendium aus Miueln des zweiten Hochschulsonderprogramms). This work was funded in part by grants from the Medical Research Council of Canada and

the National Cancer Institute of Canada (to R. B., S. B., R. P. H., and T. W. M.). National Cancer Institute Grants ROl CA62244 and ROl CA22188 supported the work of M. M.;

Grants ROl CA56542 and ROl CA 67007 supported the work of R. F. 2 These

authors

3 To whom

contributed

requests

equally

for reprints

to this

should

work.

be addressed.

Phone:

(801) 585-5258;

Fax:

(801) 585-3501; E-mail: [email protected]. 4 The

abbreviations

used

are:

HNPCC,

hereditary

nonpolyposis

colon

cancer:

HPRT.

hypoxanthine guanine phosphoribosyl transferase; APRT, adenine phosphoribosyl trans ferase; 6-tg, 6-thioguanine.

cell lines are resistant

to DNA-alkylating

agents

or base

analogues, such as 6-tg. More recently, evidence has been presented that cis-platinum-resistant cell lines may also be deficient in mismatch repair (20—22).Lesions produced by these agents are recognized both by the hMSH2 protein alone and the hMSH2/hMSH6 heterodimer (22—24)and may induce a futile mismatch repair cycle that ultimately leads to cell death (17, 25). The loss of mismatch repair would prevent the initiation of this futile repair and therefore increase cell survival. In contrast, some mismatch repair-deficient colorectal carcinoma cell lines are more sensitive to the cytotoxic effects of UV irradiation (26). This increased sensitivity is accompanied by the loss of the preferen tial repair of UV-induced lesions on the transcribed strand of coding sequences in human cells and suggests that the mismatch repair also play some role in the transcription

coupled

repair

path

way (26). Unfortunately, the colon cancer cell lines used in these character izations

have

numerous

alterations

of proto-oncogenes

and tumor

suppressors that may contribute to the final mutator phenotype. The assignment of a mutator phenotype to a specific mismatch repair gene is further complicated by the recent observation that a single tumor cell line may harbor mutations of more than one mismatch repair gene (14) or of other genes that govern mutation rates (27). The recently described mice containing knockouts of mismatch repair genes (28— 32) potentially offer a powerful approach for determining the effects of mismatch repair gene deficiencies in the absence of other alter ations. Homozygous mice deficient in Msh2, Pms2, or MIhI are viable

3765

Downloaded from cancerres.aacrjournals.org on July 13, 2011 Copyright © 1997 American Association for Cancer Research

MUTATOR PHENOTYPE IN M5H2-DEFICIENT MOUSE FIBROBL.ASTS

but primarily develop lymphomas between 2 and 12 months of age (28—31). Intestinal tumors are also observed in the majority of Msh2—I— mice that survive to >6 months (33). About 7% of animals develop a variety of skin neoplasms similar to those found in humans suffering from Muir-Torre syndrome. In the work reported here, we characterized immortalized embryo fibroblasts obtained from Msh2+/+, +1—, and —I— mice to determine whether they develop a mutator phenotype similar to that found in human colorectal carci noma cell lines. Dc Wind et al. (29) showed that mouse ES cells with knockouts of Msh2 developed an increased frequency of Hprt mutants and resistance to DNA alkylating agents. We have made more quan titative measurements of mutation rate at two genetic loci that allow us to compare the level of mutation in different cell lines. We show that mouse embryo fibroblasts derived from Msh2—I-- cells develop a mutator phenotype similar to that found in human tumor cell lines deficient in mismatch repair. However, important differences in sen sitivity to DNA-damaging agents in the immortalized cell lines were detected. Considering the high penetrance of HNPCC (34), we paid particular attention to embryo fibroblasts derived from Msh2 het erozygous mice for evidence of effects on spontaneous mutation rates or sensitivity to DNA-damaging agents, but no significant changes were detectable.

A

+1+ +1-. -1-

208

-

118

-

86 -

51 -

B

+1++1--I-‘5 10 20'‘510 20'‘510 20 @.tg protein

@

.

7

e

‘@‘

MATERIALS AND METHODS Recombinant Vectors. Mp53pro193contains a 16-kbEcoRI fragmentin pUCl8 carrying the entire mouse p53 gene and bearing a single point mutation at codon 193 that converts an arginine residue to a proline (35). The HPVI6 E7

coding sequence is present on the pJ4fll6.E7 plasmid (36). Activated c-Ha-ras sequences are present on pEJ6.fl (37). Plasmid pHMR272 contains the hygro mycin resistance gene (38).

Embryonic

Fibroblasts. Heterozygote (Msh2@)

..

mice of mixed

C57BIJ6J and 129/Ola genetic background (Fl generation) were mated. A timed pregnant female was sacrificed after 13 days of gestation, and individual fetuses were removed and washed repeatedly with PBS. Murine embryonic fibroblasts from these littermate embryos were prepared as described previ ously (35), and their Msh2 status was determined using a PCR-based geno typing assay (33). Cells to be transfected were pooled according to genotype and plated

from primary

cell cultures

at a cell density

of 3.0 X 10' cells per

60-mm dish, giving rise to secondary cultures and transfected the following

day. Transformations were performed as described previously (39) and con tamed 10 iLgof NIH3T3 carrier DNA, I @g of pHMR272, and either 10 @g of pEJfl.fl with 5 [email protected]

or 20 @.tgofMpS3prol93.

Approximately

24 h

after transfection, the cells were trypsinized and plated in fresh medium. Selection in a-MEM containing 15 @g/mlhygromycin commenced 48 h after transfection. Colonies were identified visually by microscope approximately 2 weeks later. After 14 days in selective medium, cells from individual hygro mycin-resistant

colonies

were isolated

with cloning

cylinders

and transferred

to 24-well plates containing 2 ml of selective medium. When cultures reached confluency they were passaged at a ratio of 1:5, and clones that survived the 10th passage were considered immortal.

The immortalized mouse fibroblast lines were subsequently maintained on a-MEM supplemented with 10% FCS. Drug or radiation sensitivity was examined in three E7- and ras-immortalized fibroblast lines derived from three different colonies obtained from the transformation of Msh2+/+ embryo fibroblasts, one line derived from Msh2+/— embryo fibroblasts, and two lines

Fig. I. Western blot and Gff binding activity analysis of E7/Ras-immortalized cells derived from Msh2+/+ , +1—,and —I— mice. A, Westem blot analysis of Msh2 content in the three cell lines. B, GiT binding activity. Various amounts of extracts prepared from

the three mouse lines were incubated with the 32P oligoduplex containing the G/T mismatch. The resulting protein-DNA complexes were analyzed using a gel mobility shift assay.

derived from Msh2—/— fibroblasts. Cells were treated 16—18h after being seeded at densities that would yield 25—70surviving colonies at the time of fixation. Graded doses of ionizing radiation (i.e., 0—10Gy) were delivered @

fixed and stained with a methylene blue solution made up in 50% ethanol.

lamp at a dose rate of 8

Colonies with more than 50 cells were scored, and survival was expressed as the relative plating efficiency of treated cells to the control. in vitro parameters

J/m2 per mm. For treatment with cis-platinum, cells were exposed to various

were intercompared between the cell lines with the nonparametric Mann

concentrations of drug mixed in culture medium for 72 h. Following exposure, cells were washed in PBS, and fresh medium was added. To determine 6-tg sensitivity, cells were exposed continuously to various concentrations of the drugs in growth medium. For all treatments, after 8—10days, colonies were

Whitney test using the MINITAB

using

37Cs -y-rays under

irradiation

aerobic

of cells was performed

conditions

and at room

using a germicidal

temperature.

UV

general statistics program (Minitab Inc.,

PA). Western Blot and Mismatch Binding Analysis. Approximately5 X 106 cells were harvested by treatment with trypsin followed by two washes with

3766

Downloaded from cancerres.aacrjournals.org on July 13, 2011 Copyright © 1997 American Association for Cancer Research

MUTATOR PHENOTYPE IN MSH2-DEFICIENTMOUSE FIBROBLASTS

(5.6%) Msh24' and 3 of 15 (20%) Msh2@ colonies survived the tenth passage. Therefore, the immortalization frequency by a mutant

Table 1 Microsatellite instability in E7/Ras-immortalized mouse embryofibroblast cell lines

(EF2O1)cell DNA was purified from subclones derived from Msh2+/+ (EFIOO)or —I— ofclones lines and used for amplification of the four microsatellite loci. The proportion showing presented.Locus novel alleles is EF2O1D3mit203

p53 allele did not appearto depend upon Msh2 status in this expert

EF100

2/18D6mit59 3/18D7mit62 2/18DlPmit36

0/17 0/14 0/16 0/17

3/17

PBS. The cell pellets were resuspended in 150 pi of high-salt buffer (50 rust potassium phosphate, pH 7.5, 300 mM NaCl, 0. 1 rust EDTA, 1 mM DTT, and 10% glycerol) and subjected to three freeze-thaw cycles. The samples were

centrifuged, and the supernatant dialyzed against the above buffer containing 50 mM NaCI. Protein

concentrations

of the total cell extracts

were determined

using the Bradford method (Bio-Rad). Ten @gof +1+, +/—, and —I— total cell extracts were electrophoresed on a 7% SDS-polyacrylamide gel and

transferred to nitrocellulose. Anti-hMSH2 polyclonal antibody (40) was used at a concentration of 3 ,.@g/ml,and Msh2 was detected by chemiluminescence as directed by the manufacturer

(Boehringer-Mannheim,

Indianapolis,

IN).

For the mismatch binding assay, 32P-labeledDNA duplex containing a G@ mismatch was prepared as described previously oligonucleotides

used

to construct

(41). The sequences of the

this substrate

were

as follows:

top, 5'-

CGGCGAATfCCACCAAGCTFGATCGCTCGAGGTACCAGG-3'; bottom, 5'-CCTGGTACCTCGAGCGATCGAGCTFOGTGQAATFCGCCG-3'. Mis matched DNA binding assays were carried out in 30-id reactions containing 5, 10, or 20 i.@gof whole cell protein extract, DNA binding buffer [50 mivi potassium phosphate, pH 7.5, 50 mM NaCl, 0.1 mM EDTA, 1 mM DTF, 10%

glycerol, 200 ng of poly(dI'dC)poly(dFdC)J, 1 ng of 32P-labeledDNA duplex and incubated at 37°Cfor 10 mm. The binding reactions were loaded onto a 5% polyacrylamide gel (1.5 mm thick, 22-cm-long, and 14 cm wide) contain ing 2.5% glycerol

that was run at 35 mA for 3 h. The gel was then dried and

analyzed

on a Molecular

obtained

from the Kimmel

Measurement

Dynamics

Phosphorlmager.

Cancer

of Mutation

Institute

Rates.

Nucleic

Oligonucleotides Acids

were

Facility.

Mutation rates were measured by Luria

Delbruck fluctuation tests as described previously (16). Replica cultures were inoculated with 100 cells and then grown to a final density of 1—4X 106. All

of the cells from each replica were then plated in selective medium containing either 5 !.Lg/ml6-tg or 2 inst ouabain. Colonies growing after 10 days were fixed, stained, and scored. Mutation rates were calculated by the method of the mean. Microsatellite MSH2+/+

Instability.

or MSH2—/—

microsatellite

Subclones

mice

loci were analyzed

were

grown

derived

from cell lines derived

out for purification

for each subclone.

Primers

of DNA.

from Four

for the amplifi

cation of these microsatellites were obtained from the mouse genome data base.

RESULTS

ment. Several hygromycin-resistant colonies that were cotransfected with activated Ras and HPVJ6 E7 survived at least 10 passages. The transfected clones used in these experiments were carried as long as 40 generations with no decrease in growth rate or plating efficiency. Msh2 Expression and Mismatch Binding. E7/Ras-transfected cell lines were first examined for Msh2 expression by Western blot analysis to be certain that the immortalized cells maintained the expected levels of this protein. Fig. 1A shows that Msh2 was absent in cells derived from the —I— mice and that cells derived from the +1— mice had lower levels of Msh2 relative to the +1+ cells. Off mis match binding activity in the three lines showed a similar pattern (Fig. 1B). The E7/Ras-immortalized cell line derived from the Msh2—/— mice showed a loss of binding activity. The line derived from the +1— mice had lower levels of binding relative to that found in the Msh2+/+ line. Microsatellite Instability. The frequency of cells containing novel microsatellite alleles at four different loci was determined using DNA purified from cultures grown out from single cell clones. Four loci were examined in 14—18subclones isolated from +1+ or —I--cells obtained from the E7/Ras-transfected lines. Novel alleles were de tected at each of the loci tested in 12—18%of the subclones isolated from the Msh2-deficient line (Table 1). In contrast, no novel alleles were detected in subclones isolated from the Msh2+/+ cells (overall frequency, 2n DNA content. Although the number of active X chromosomes may not be altered in these cells, this problem was avoided by measuring the rate of mutation to ouabain resistance. Such mutations act dominantly, so they are not obscured in polyploid cells (42). Furthermore, recovery of 6-tg-resistant cells in the Msh2 proficient and deficient fibroblasts may be affected by the differing sensitivity of these cells to the selective agent (see below). Table 2 shows that the rate of mutation to ouabain resistance in the Msh2—/—line was elevated 12-fold relative to the Msh2+/+ line and 25-fold relative Msh2+I— line. The lower

p53-l5 +1+ 3.8X l0@

+1— 4.4 X lO_8

3.1X

forplating Mutation rates are expressed as mutations/cell/generation and are not corrected efficiency. b

not tested.

3767

Downloaded from cancerres.aacrjournals.org on July 13, 2011 Copyright © 1997 American Association for Cancer Research

MUTATOR PHENOTYPE IN MSH2-DEFICIENTMOUSE FIBROBLASTS

A

B

C

.1

.91

U La

‘U be

be

.E

.5 a :a

Fig. 2. Sensitivity of E7/Ras-immortalized mouse embryo fibroblasts derived from Msh2+/+, +1—, and —I— mice to DNA-damaging agents. A, sensitivity to 6-tg; B, sensitivity to UV light; C, sensitivity to cis-platinum; D, sensitivity to ioniz

ing radiation. The results represent mean survival 6-thioguanine

values based on 2—6independent experiments. The

@g/m1)

Uv (j/m2)

range of plating efficiencies of the E7/Ras-immor talized mouse embryo fibroblasts was 33—I10% for

the Msh2+/+ cells, 8—52%for the Msh2+/— cells, and 29—130%for the Msh2—/— cells. , values for the Msh2—/— cells; L@,Msh2+/—cells; ., Msh2+/+ cells. Data presented are for one

C

D

E7/ras-transformed line derived from Msh2+/+, +1—, or —I— mice. Two additional clones from Msh2+/+ mice and one additional clone from Msh2—/—mice gave similar results (data not

shown). C .@

LI

‘U be .@

.5 a

.0001 0.0

0.2

0.4 0.6 0.8 cisplatinu.m (IIM)

rate of mutation in the Msh2 +1—line may be a reflection of the lower plating efficiency of these cells. Plating efficiencies ranged from 33 to 110% for the Msh2+/+ cells, from 8 to 52% for the Msh2+/— cells, and from 29 to 130% for the Msh2—I—cells. Mutant p53-transfected embryo fibroblasts were also examined for mutation

rates

to ouabain

resistance

to determine

whether

the gene(s)

used for immortalization influenced mutation rates. Mutant p53-trans fected embryo fibroblast clones derived from —I— mice showed a very similar response in mutation rate: an 8-fold increase in the rate of mutation to ouabain resistance relative to p53-transfected fibroblasts derived from Msh2+/+ mice and a 70-fold increase compared to cells derived from the +1—mice (Table 2). Sensitivity to Damaging Agents. To determine whether cells de rived from the Msh2—/—,+1—, or +1+ mice displayed altered sensitivity to DNA-damaging agents similar to that shown by the mismatch repair-deficient human colorectal carcinoma cell lines, we determined response of these cells to 6-tg, UV light, cis-platinum, and ionizing radiation. In these experiments, both E7/Ras-transfected fi broblasts and spontaneously immortalized lines growing out of the primary

mouse

embryo

fibroblast

cultures

were

used.

Both

types

1.0

1.2

4

6

8

10

12

Radiation(Gy)

levels of resistance found in human colon cancer cell lines. Sponta neously and E7/Ras-immortalized cells derived from Msh2+/— mice showed no significant change in sensitivity to 6-tg relative to lines derived from the +1+ mice. Different results were obtained for spontaneously immortalized and E7/Ras-transfected mouse embryo fibroblasts in response to UV, ionizing

radiation,

or cis-platinum

treatment

(Figs.

2 and

3). Sponta

neously immortalized cultures derived from Msh2—I—mice were significantly more resistant (more than 2-fold) to the cytotoxic effects of UV compared with cells derived from +1+ or +1— mice (P = 0.02). E7/Ras-immortalized cells from the —I— mice showed no difference in UV sensitivity compared to those derived from +1+ mice. Spontaneously immortalized mouse fibroblasts derived from Msh2—/—and +1—embryos were significantly more resistant to cis-platinum than those from +1+ embryos (P = 0.02 and 0.05, respectively). E7/Ras-transfected fibroblasts from +1+ or —I— em bryos showed no difference in sensitivity to cis-platinum. E7/Ras and spontaneously

immortalized

cell lines derived

from all three types of

mice showed similar responses to ionizing radiation.

of

Msh2—/—lines showed a statistically significant (3—50-fold; DISCUSSION P = 0.04 and 0.01 for the E7/Ras and spontaneously immortalized cells, respectively) resistance to the cytotoxic effects of 6-tg relative We examined embryonic fibroblast cell lines derived from to the Msh2+/+ or +1—lines (Figs. 2 and 3). This is similar to the Msh2—/— , +1—,and +1+ mice to determine whether they developed 3768

Downloaded from cancerres.aacrjournals.org on July 13, 2011 Copyright © 1997 American Association for Cancer Research

MUTATOR PHENOTYPE IN MSH2-DEHCIENT MOUSE FIBROBLASTS

A

B

I

@

.1 .@

.01

a. 4' ‘4

.5

Fig. 3. Sensitivity of spontaneously immortal ized mouse

embryo

cells derived

from Msh2+/+,

.001

+1—,and —I—mice to DNA-damaging agents.

0

The resultsrepresentmeansurvivalvaluesbased on 3—6independent experiments. A, sensitivity to 6-tg; B, sensitivity to UV light; C, sensitivity to cis-platinum;

4

8

6-thioguanine

12

16

20

UVQ/m2)

(jag/mi)

D, sensitivity to ionizing radiation.

The results represent mean survival values based on 2—6independent experiments. The range of plating efficiencies of the spontaneously immortal ized mouse embryo fibroblasts was 0.27—6.2%for the Msh2+/+ cells, 0.31—3.5%for the Msh2+/— cells, and 0.4—31%for the Msh2—/— cells. U, values for the Msh2—/— cells;

C

D

@,Msh2+/— cells;

., Msh2+/+ cells.

.1

.1

.@

.5

I

.01

.001

0

2

4

6

8

10

12

Radiation (Gy) cisplatinum

a mutator phenotype similar to that found in human colorectal carcinoma cell lines deficient in mismatch repair. Colon cancer cell lines have numerous alterations of proto-oncogenes and tumor suppressors that could influence this phenotype. The use of the mouse embryo cell lines appears to circumvent this difficulty, although the lines used have been immortalized to facilitate the measurements of mutation rate and sensitivity to agents that induce the futile repair cycle. Our data indicate that the E7/Ras-immor talized mouse line derived from Msh2—/-—mice develops micro satellite instability, resistance to the cytotoxic effects of 6-tg, and a 6—12-fold increase in mutation rate. Thus, by these criteria the mutator phenotype in the immortalized Msh2—/—embryo fibro blast lines is similar to that found in the repair-deficient human tumor cell lines (7, 8, 16, 18). There are quantitative differences in the elevation of mutation rates in the mouse Msh2-deficient cells and the rates reported for the human colorectal carcinoma cell lines deficient in hMLHJ or hMSH6. Mutation rates at the HPRT locus in the human tumor cell lines are elevated 100—700-fold (16, 18), whereas the rates of mutation to ouabain resistance are elevated 24—30-fold.5 The Hprt mutation rate reported here for the 5 B. Richards

and

M.

Meuth,

unpublished

data.

(pM)

Msh2—/—lines is very similar to the rates measured for the human tumor cell lines. However, the rate of Hprt mutation in the mouse Msh2+I+ lines appears to be about 10-fold higher than the rate for mismatch repair-proficient human cell lines. Mismatch repair-deficient mouse fibroblasts were resistant to 6-tg, much like the repair-deficient colon cancer cell lines (43). However, the E7/Ras-transformed mouse fibroblast lines derived from Msh2—I-- mice showed no change in sensitivity to cis-platinum, UV light, or ionizing radiation. Spontaneously immortalized mouse cells derived from the Msh2—/—mice were significantly more resistant to cis-platinum and UV light but not ionizing radiation. Presumably, cells deficient in Msh2 were unable to recognize the platinum adducts and initiate the futile repair cycle. The resistance to UV light is unexpected, given the increased sensitivity of human mismatch repair deficient cell lines reported previously (26). Nevertheless, the differ ences in the responses in the two sets of mouse embryo fibroblast lines suggest that the altered sensitivity to some of these DNA-damaging agents may be a product of immortalization or alterations of growth controls, as well as mismatch repair status. For example, previous studies indicate that mutations ofp53 may induce resistance to DNA damaging agents, such as UV light, by eliminating p53-dependent apoptosis (44). Thus, some of the phenotypic alterations found in the

3769

Downloaded from cancerres.aacrjournals.org on July 13, 2011 Copyright © 1997 American Association for Cancer Research

MUTATOR PHENOTYPE IN MSH2-DEFICIENTMOUSE FIBROBLASTS

mismatch repair-deficient human tumor cell lines may not be solely the result of the repair deficiency. Because of the high penetrance of HNPCC (34), another question that we addressed in these studies was whether the Msh2 heterozy gotes displayed any changes in mutation rate or sensitivity to DNA damaging agents that might predispose these cells to loss of the remaining Msh2 wild-type allele. No increase in mutation rate was seen in these heterozygous cells; in fact, they had consistently lower mutation

rates

than

the

Msh2+/+

cells.

It is also

notable

that

the

Msh2 heterozygous mouse cells showed no alteration in sensitivity to

Uv relativeto theMsh2+/+ cells.Theseresultsshouldbe considered in light of the recent demonstration of a deficiency in transcription coupled repair in a human lymphoblastoid cell strain heterozygous for hMSH2 and proficient in mismatch repair (26). Thus, except for the increased resistance to cis-platinum seen in the spontaneously trans formed mouse cells, our studies fail to reveal any phenotype in the Msh2-heterozygous mouse cells that might provide a selective growth advantage or predispose to the acquisition of further mutations in mismatch

repair

genes

or genes

that control

cell growth.

These studies support the idea that the loss of mismatch repair results

in an increase

in spontaneous

mutation

rate. Secondary

alter

ations of replication fidelity, other repair genes, or genes regulating the cell cycle may further enhance this mutator phenotype. Presum ably, this mutator phenotype increases the likelihood of mutations in tumor suppressor or proto-oncogenes that are necessary for carcino genesis. The detection of mutations in tumor suppressor genes at sites similar to hot spots of mutation in the HPRT gene in mismatch repair-deficient colon cancer cell lines further supports this proposal (16, 45—47).The questions of why tumor development is restricted to only a few tissue types despite the expression of Msh2 in a wide range of different tissues (40, 48, 49) and why only limited numbers of tumors were seen throughout the intestines of individual Msh2-defi cient animals,

although

every

crypt stem cell is homozygous

gene defect (unlike human HNPCC heterozygotes), resolved.

for the

Venter, J. C., Dunlops, M. G., Hamilton, S., Petersen, G. M., de Ia Chapelle, A., Vogelstein, B., and Kinzler, K. W. Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature (Lond.), 371: 75—80,1994. 7. Parsons, R., Li, G-M., Longley, M. J., Fang, W-H., Papadopoulos, N., Jen, J., de la

Chapelle, A., Kinzler, K. W., Vogelstein, B., and Modrich, P. Hypermutability and mismatch repair deficiency in RER+ tumor cells. Cell, 75: 1227—1236,1993. 8. Boyer, J. C., Umar, A., Risinger, J. I., Lipford, J. R., Kane, M., Yin, S., Barrett, J. C.,

Kolodner, R. D., and Kunkel, T. A. Microsatellite instability, mismatch repair deficiency, and genetic defects in human cancer cell lines. Cancer Res., 55: 6063—

6070, 1995. 9. Drummond, J. T., Li, G-M., Langley, M. J., and Modrich, P. Isolation of a hMSH2— p160 heterodimer that restores DNA mismatch repair to tumor cells. Science (Wash ington DC), 268: 1909—1911, 1995.

10. Palombo, F., Galhinari, P., Iaccarmno,I., Lettieri, T., Hughes, M., D'Arrigo, A., Truong, 0., Hsuan, J. J., and Jiricny, J. GTBP, a 160 kilodalton protein essential for mismatch binding activity in human cells. Science (Washington DC), 268: 1912— 1914,1995.

1I. Acharya, S., Wilson, T., Gradia, S., Kane, M. F., Guerette, S., Marsischky, G. T., Kolodner, R., and Fishel, R. hMSH2 forms specific mispair binding complexes with hMSH3 and hMSH6. Proc. Natl. Acad. Sci. USA, 93: 13629—13634,1996. 12. Papadopoulos, N., Nicolaides, N. C., Liu, B., Parsons, R., Lengauer, C., Palombo, F., D'Arrigo, A., Markowitz, S., Willson, J. K. V., Kinzler, K. W., Jiricny, J., and Vogelstein, B. Mutations of GTBP in genetically unstable cells. Science (Washington DC), 268: 1915—1917,1995. 13. Risinger, J. I., Umar, A., Boyd, J., Berchuck, A., Kunkel, T. A., and Barret, J. C. Mutation of MSH3 in endometrial cancer and evidence for its functional role in heteroduplex repair. Nat. Genet., 14: 102—105,1996.

14. Malkhosyan, S., Rampino, N., Yamamoto, H., and Perucho, M. Frameshift mutator mutations. Nature (Lond.), 382: 499—500,1996. 15. Shibata, D., Peinado, M. A., Ionov, Y., Malkhosyan, S., and Perucho, M. Genomic instability in repeated sequences is an early somatic event in colorectal tumorigenesis

that persists after transformation. Nat. Genet., 6: 273—281,1994. 16. Bhattacharyya, N. P., Skandalis, A., Ganesh, A., Groden, J., and Meuth, M. Mutator phenotypes in human colorectal carcinoma cell lines. Proc. Nail. Acad. Sci. USA, 91: 6319—6323, 1994. 17. Kat, A., Thilly, W. G., Fang, W-H., Longley, M. J., Li, G-M., and Modrich, P. An alkylation-tolerant, mutator human cell line is deficient in strand-specific mismatch

repair. Proc. Nail. Acad. Sci. USA, 90: 6424—6428,1993. 18. Eshleman, J. R., Lang, E. Z., Bowerfind, G. K., Parsons, R., Vogelstein, B., Wilson,

J. K. V., Veigl, M. L., Sedwick, W. D., and Markowitz, S. D. Increased mutation rate at the hprt locus accompanies microsatellite instability in colon cancer. Oncogene, 10: 33—37, 1995.

19. Phear, G., Bhauacharyya, N. P., and Meuth, M. Loss of heterozygosity and base substitution at the APRT locus in mismatch repair-proficient

and -deficient colorectal

carcinoma cell lines. Mol. Cell. Biol., 16: 6516—6523,1996.

remain to be

20. Anthony. D. A., Mcllwrath, A., Gallagher, W., Edlin, A., and Brown, R. Microsat ellite instability, apoptosis and loss of p53 function in drug-resistant tumor cells.

Cancer Res., 56: 1374—1381,1996. 21. Aebi, S., Kurdi-Haidar, B., Gordon, R., Cenni, B., Theng, H., Fink, D., Christen, R. D., Bolund, C. R., Koi, M., Fishel, R., and Howell, S. B. Loss of DNA mismatch repair in acquired resistance to cisplatin. Cancer Res., 56: 3087—3090, 1996. 22. Fink, D., Nebel, S., Aebi, S., Zheng, H., Cenni, B., Nehme, A., Christen, R. D., and

ACKNOWLEDGMENTS We acknowledge Homayoun Vaziri and José de Ia Pompa for stimulating

Howell, S. B. The role of DNA mismatch repair in platinum drug resistance. Cancer Res., 56: 4881—4886, 1996. 23. Mello, J. A., Acharya, S., Fishel, R., and Essigman, J. M. The mismatch repair protein

discussions.

hMSH2 binds selectively to DNA adducts of the anticancer drug cisplatin. Chem. Biol., 3: 579—589, 1996. 24. Duckett, D. R., Drummond, J. T., Murchie, A. I. H., Reardon, J. T., Sancar, A., Lilley,

REFERENCES 1. de la Chapelle, A., and Peltomaki, P. Genetics of hereditary colon cancer. Annu. Rev. Genet., 29: 329—348,1995. 2. Fishel, R., Lescoe, M. K., Rao, M. R. S., Copeland, N. G.. Jenkins, N. A., Garber,

J., Kane, M., and Kolodner, R. The human mutation gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell, 75: 1027—1038, 1993. 3. Leach, F. S., Nicolaides, N. C., Papadopoulos, N., Liu, B., Jen, J., Parsons, R., Peltomaki, P., Sistonen, P., Aaltonen, L. A., Nystrosm-Lahti, M., Guan, X-Y., Zhang, J., Meltzer, P. 5., Yu, J-W., Kao, F-T., Chen, D. J., Cerosaletti, K. M., Foumier, R. E. K., Todd, S., Lewis, T., Leach, R. J., Naylor, S. L., Weissenbach, J., Mecklin,

J-P., Jarvinen, H., Petersen, G. M., Hamilton, S. R., Green, J., Jass, J., Watson, P., Lynch, H. T., Trent, J. M., de Ia Chapelle, A., Kinzler, K. W., and Vogelstein, B.

Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell, 75: 1215—1225, 1993. 4. Bronner, C. E., Baker, S. M., Morrison, P. T., Warren, G., Smith, L. G., Lescoe, M. K., Kane, M., Earabino, C., Lipford, J., Lindblom, A., Tannergard, P., Bollag, R. J., Godwin, A. R., Ward, D. C., Nordenskjold, M., Fishel, R., Kolodner, R., and Liskay, R. M. Mutation in the DNA mismatch repair gene homologue hMLHJ is associated with hereditary non-polyposis colon cancer. Nature (Lond.), 368: 258— 261, 1994. 5. Papadopoulos, N., Nicolaides, N. C., Wei, Y-F., Ruben, S. M., Carter, K. C., Rosen, C. A., Haseltine, W. A., Fleischmann, R. D., Fraser, C. M., Adams, M. D., Venter,

J. C., Hamilton, S. R., Petersen, G. M., Watson, P., Lynch, H. T., Peltomaki, P., Mecklin, J-P., de Ia Chapelle, A., Kinzler, K. W., and Vogelstein, B. Mutation of a mutL homolog in hereditary colon cancer. Science (Washington DC), 263: 1625—

1629, 1994. 6. Nicolaides, N. C., Papadopoulos, N., Liu, B., Wei, Y-F., Carter, K. C., Ruben, S. M., Rosen, C. A., Haseltine, W. A., Fleischmann, R. D., Fraser, C. M., Adams, M. D.,

D. M. J., and Modrich, P. Human MutSa recognizes damaged DNA base pairs containing O'@-methylguanine, O'-methylthymine, or the cisplatin-d(GpG) adduct. Proc. Natl. Acad. Sci. USA, 93: 6443—6447, 1996. 25. Karran, P., and Bignami, M. DNA damaged tolerance, mismatch repair and genome instability. BioEssays, 16: 833—839, 1994. 26. Mellon, I., Rajpal, D. K., Koi, M., Boland, C. R., and Champe, G. N. Transcription coupled repair deficiency and mutations in human mismatch repair genes. Science

(Washington DC), 272: 557—560,1996. 27. da Costa, L. T., Liu, B., El-Deity, W. S., Hamilton, S. R., Kinzler, K. W., Vogelstein, B., Markowitz, S., Wilson, J. K. V., de Ia Chapelle, A., Downey, K. M., and So, A. G. Polymerase 8 variants in RER colorectal tumours. Nat. Genet., 9: 10—11,1995. 28. Reitmair, A. H., Schmits, R., Ewel, A., Bapat. B., Redston, M., Waterhouse, P., Mittrucker, H-W., Wakeham, A., Liu, B., Thomason, A., Griesser, H., Gallinger, S., Balhausen, W. G., Fishel, R., and Mak, T. W. MSH2 deficient mice are viable and susceptible to lymphoid tumours. Nat. Genet., 11: 64—70,1995. 29. de Wind, N., Dekker, M., Berns, A., Radman, M., and te Ride, H. Inactivation of the

mouse MSH2 gene results in mismatch repair deficiency, methylation tolerance, hyperrecombination, and predisposition to cancer. Cell, 82: 321—330,1995. 30. Baker, S. M., Bronner, C. E., Zhang, L., Plug, A. W., Robatzek, M., Warren, G., Elliott, E. A., Yu, J., Ashley, T., Arnheim, N., Flavell, R. A., and Liskay, R. M. Male mice defective in the DNA mismatch repair gene PMS2 exhibit abnormal chromo some synapsis in meiosis. Cell, 82: 309—319, 1995. 31. Baker, S. M., Plug, A. W., Prolla, T. A., Bronner, C. E., Harris, A. C., Yao, X.,

Christie, D-M., Monell, C., Arnheim, N., Bradley, A., Ashley, T., and Liskay, R. M. Involvement of mouse Mlhl in DNA mismatch repair and meiotic crossing over. Nat. Genet., 13: 336—342,1996. 32. Edelman, W., Cohen, P. E., Kane, M., Lau, K., Morrow, B., Bemsen, S., Umar, A.,

Kunkel, T., Cattoretti, G., Chaganti, R., Pollard, J. W., Kolodner, R. D., and Kucher

3770

Downloaded from cancerres.aacrjournals.org on July 13, 2011 Copyright © 1997 American Association for Cancer Research

MUTATOR PHENOTYPE IN MSH2-DEFICIENTMOUSE FIBROBLASTS lapati, R. Meiotic pachytene arrest in MLH-l-deficient

mice. Cell, 85: 1125-1134,

1996. 33. Reitmair, A. H., Redston, M., Cai, J. C., Chuang, T. C. Y., Bjerknes, M., Cheng,

42. Siminovitch, L. On the nature of hereditable variation in cultured somatic cells. Cell, 7: 1—11,1976. 43. Hawn, M. T., Umar, A., Carethers, J. M., Marra, G., Kunkel, T. A., Boland, C. R., and

Koi, M. Evidence for a connection between the mismatch repair system and the G2 H., Hay, K., Gallinger, S., Bapat, B., and Mak, T. W. Spontaneous intestinal cell cycle checkpoint. Cancer Rca., 55: 3721—3725,1995. carcinomas and skin neoplasms in Msh2-deficient mice. Cancer Res., 56: 3842— 44. Ford, J. M., and Hanawalt, P. C. Li-Fraumeni syndrome fibroblasts homozygous for 3849,1996. 34. Dunlop, M. G., Farrington, S. M., Carothers, A. D., Wyllie, A. H., Sharp, L., Liu, B., p53 mutations are deficient in global DNA repair but exhibit normal transcription Kinzler, K. W., and Vogelstein, B. Cancer risk associated with germline DNA coupled repair and enhanced UV resistance. Proc. Natl. Acad. Sci. USA, 92: 8876— mismatch repair gene mutations. Hum. Mol. Genet., 6: 105—1 10: 1997.

35. Rovinski, B., and Benchimol, S. Immortalization of rat embryo fibroblasts by the cellular p53 oncogene. Oncogene, 2: 445—452, 1988. 36. Storey, A., Nm, D., Murray, A., Osborn, K., Banks, L., and Crawford, L. Comparison of the in vitro transforming activities of human papillomavirus types. EMBO J., 7: 1815—1820, 1988. 37. Shih, C., and Weinberg, R. A. Isolation of a transforming sequence from a human

bladdercarcinomacell line.Cell,29: 161—169, 1982. 38. Bernard, H. U., Kraemmer, G., and Roewekamp, W. G. Construction of a fusion gene

thatconfersresistanceagainsthygromycinB to mammaliancellsinculture.Exp.Cell Res., 158:237—243, 1985. 39. Munroe, D. G., Peacock, J. W., and Benchimol, S. Inactivation of the cellular p53 gene is a common feature of Friend virus-induced erythroleukemia: relationship of inactivation to dominant transforming alleles. Mol. Cell. Biol., JO: 3307—3313,1990. 40. Wilson, T. M., Ewel, A., Duguid, J. R., Eble, J. N., Lescoe, M. K., Fishel, R., and

Kelley, M. R. Differential cellular expression of the human MSH2 repair enzyme in small and large intestine. Cancer Res., 55: 5146—5150,1995. 41. Fishel, R., Ewel, A., Lee, S., Lescoe, M. K., and Griffith, J. Binding of mismatched

8880, 1995. 45. Lazar, V., Grandjouan,

S., Bognel, C., Couturier, D., Rougier, P., Bellet, D., and

Bressac-de Paillerets, B. Accumulation of multiple mutations in tumor suppressor genes during tumorigenesis in HNPCC patients. Hum. Mol. Genet., 3: 2257—2260, 1994. 46. Markowitz,

S., Wang, J., Myeroff,

L., Parsons,

R., Sun, L., Lutterbaugh,

J.,

Vogelstein, B., Brattain, M., and Willson, J. K. V. Inactivation of the type II TGF-p receptor in colon cancer cells with microsatellite instability. Science (Washington DC), 268: 1336—1338, 1995. 47. Huang, J., Papadopoulos, N., McKinley, A. J., Farrington, S. M., Curtis. L. J., Wyllie, A. H., Zheng, S., Willson, J. K. V., Markowitz, S. D., Morin, P., Kinzler, K. W., Vogelstein, B., and Dunlop, M. G. APC mutations in colorectal tumors with mismatch

repair deficiency. Proc. NatI. Aced. Sci. USA, 93: 9049—9054,1996. 48. Varlet, I., Pallard, C., Radman, M., Moreau, J., and de Wind, N. Cloning and

expression of the Xenopus and mouse Msh2 DNA mismatch repair genes. Nucleic Acids Res., 22: 5723—5728,1994. 49. Leach, F. S., Polyak, K., Burrell, M., Johnson, K. A., Hill, D., Dunlop. M. G.. Wyllie, A. H., de Ia Chapelle, A., Hamilton, S. R., Kinzler, K. W., and Vogelstein. B.

Expression of the human mismatch repair genes in normal and neoplastic tissues. Cancer Res., 56: 235—240,1996.

microsatellite DNA sequences by the human MSH2 protein. Science (Washington DC), 266: 1403—1405,1994.

3771

Downloaded from cancerres.aacrjournals.org on July 13, 2011 Copyright © 1997 American Association for Cancer Research