• Nie Znaleziono Wyników

Atm-haploinsufficiency enhances susceptibility to carcinogen-induced mammary tumors.

N/A
N/A
Protected

Academic year: 2021

Share "Atm-haploinsufficiency enhances susceptibility to carcinogen-induced mammary tumors."

Copied!
8
0
0

Pełen tekst

(1)

Atm-haploinsufficiency enhances susceptibility to carcinogen-induced

mammary tumors

Shu Lu

1,†

, Kate Shen

1,†

, Yaolin Wang

4

, Steven J.Santner

1

,

Jie Chen

5

, S.C.Brooks

1,2

and Y.Alan Wang

1,3,

1Karmanos Cancer Institute,2Department of Biochemistry and Molecular

Biology,3Department of Pathology, Wayne State University, School of Medicine, 110 E. Warren Avenue, Detroit, MI 48201, USA,4Department of

Tumor Biology, Schering-Plough Research Institute, Kenilworth, NJ 07033, USA and5Department of Mathematics, University of Massachusetts, Boston,

MA 02215, USA

To whom correspondence should be addressed. Tel:+1 313 833 0715;

Fax:+1 313 831 7518; E-mail: wangya@kci.wayne.edu

Ataxia-telangiectasia (A-T), which is due to mutations in

the ATM gene, is a rare autosomal recessive genomic

instability syndrome characterized by radiosensitivity

and predisposition to cancer. Epidemiological studies

have suggested that relatives of A-T patients (A-T carriers)

have increased risks of developing breast cancer. We

pro-pose that increased breast cancer risks in A-T carriers may

be due to exposure to various environmental carcinogens

and/or dietary consumption. To test this hypothesis, we

treated a congenic strain of Atm

+/

mice with DMBA

(7,12-dimethylbenz(

a)anthracene), a mammary

carcino-gen, and observed mammary tumor incidence. It was

found that Atm

+/

mice have a 2-fold increase, as well as

early onset, in mammary tumor incidence relative to

wild-type mice (P

< 0.005). The increased mammary

tumor development is correlated with a 3-fold increase

in the development of mammary dysplasia in Atm

+/

com-pared with wild-type mice (P

< 0.05). We also found that

Ras signaling pathway was not activated in DMBA-induced

mammary tumors irrespective of the Atm status. At the

cellular level, Atm-haploinsufficiency confers increased

cellular stress manifested by an increased p53 expression

and a slightly enhanced survival of mammary epithelial

cells in response to radiation. Our results demonstrate that

Atm heterozygotes are predisposed to mammary tumor

development and support the hypothesis that exposure to

environmental carcinogens contributes to the increased

rate of breast cancer development in A-T carriers. Given

that 1% of the general population are ATM heterozygotes

(A-T carriers), this study has great implications in breast

cancer development in this population.

Introduction

ATM, which encodes a serine/threonine protein kinase of

the PIKK family, is known to ensure genomic integrity by

controlling cell cycle checkpoint in response to DNA damage

signals. ATM is required for the phosphorylation and

activa-tion of downstream genes such as p53, hChk2/hCds1, MDM2

and BRCA1. Functional mutations or amplifications of these

genes have frequently been associated with the development of

breast cancer (1–3). The importance of ATM in suppressing

breast cancer development is supported by recent studies

demonstrating that ATM is epigenetically silenced in advanced

breast cancer (4) and that ATM mutations contribute to the

development of familial breast and ovarian cancer (5).

Consistent with these observations, a number of

epidemio-logical studies have suggested that blood relatives of

ataxia-telangiectasia (A-T) patients have increased risks of developing

breast cancer (6–12). Indeed, haploinsufficiency of Atm has

been shown to result in increased sensitivity to

radiation-induced aging phenotypes, such as hair graying and cataracts,

in mouse models (13,14). About 80% of the mutations in

A-T patients are due to truncating mutations that lead to the

loss of ATM protein (15,16). Atm truncating/knockout mouse

models have faithfully recapitulated most of the A-T-associated

phenotypes (17–20). However, none of the truncated Atm

knockout mouse models develop mammary tumors when housed

in a controlled environment. On the other hand,

10% of the

mothers of A-T patients (obligate A-T carriers) are more likely

to develop breast cancers at an early age (8,10–12). These

observations suggest that additional environmental factors may

influence the breast cancer development in A-T carriers.

In light of the varied environmental exposures encountered

by the A-T carriers, it is suggested that A-T carriers are more

sensitive to environmental carcinogens, and hence have higher

incidence of breast cancer than the general population. In this

study, we tested this hypothesis and analyzed the incidence of

carcinogen-induced mammary tumors in an A-T carrier mouse

model system.

Materials and methods

Mice

Male Atm mice in a mixed genetic background [129SvEv x Black Swiss, (20)] were backcrossed with female FVB/N mice to generate FVB.129S6-Atm F1 mice. The F1 mice were then backcrossed again with female FVB/N mice for a total of 13 generations to obtain FVB.129S6(BLKSW)-Atmtm1Ledmice. PCR assay was used to identify Atm heterozygotes with the following primers: M1:50-tgtagataggtcagcattggat-30; M10: 50-gtcaaaattcgatctgctgct-30; Neo1: 50-gctctttactgaaggctctttac-30. PCR amplification was performed in 1· GPCR

buffer [16.6 mM (NH4)2SO4, 67 mM Tris–HCl (pH 8.3), 6.7 mM MgCl2, 5 mM

bME, 1% DMSO, 80 mg/ml BSA, 1 mM dNTP, 150 nM PCR primers and 2 U Taq polymerase]. PCR amplification was initially denatured for 2 min at 94C, followed by 30 cycles of 30 s at 94C, 45 s at 56C and 60 s at 72C. PCR

product was resolved onto 2% agarose gel and wild-type and mutant genotypes were identified as 125 and 400 bp bands, respectively, on agarose gel in the presence of ethidium bromide. Mice were fed ad libitum and housed in a barrier facility with 12-h light cycle.

DMBA mammary carcinogenesis

Wild-type (n¼ 8) and Atm+/mice (n¼ 17) were used for 7,12-dimethyl-benz(a)anthracene (DMBA) treatment in FVB congenic background as described (21). In brief, DMBA (Sigma) was dissolved in olive oil at a Abbreviations:A-T, ataxia-telangiectasia; DMBA,

7,12-dimethylbenz(a)anthracene; LOH, loss of heterozygosity; MAPK, mitogen-activated protein kinase.

These authors contributed equally to this work.

(2)

concentration of 5 mg/ml and administrated to mice6 weeks of age by oral gavage at a dose of 1 mg once a week for five consecutive weeks. Mammary tumor development was followed by weekly palpation of DMBA-treated mice. Whole mount and histological analysis of mammary gland

Whole mount analysis was performed as described (22). For histological analysis of the whole mount mammary gland, the tissue was removed from the slide and placed in xylene for 30 min. The whole mount was then paraffin-embedded and sectioned with a Zeiss microtome. Sections were rehydrated in a series of descending concentrations of alcohol followed by counterstaining in fast green (Sigma). Histological analysis of mammary tumors was performed as described (23).

Protein expression analysis

Frozen mammary glands or mammary tumors were thawed on ice followed by homogenization in NP 40 buffer (150 mM NaCl, 50 mM Tris–HCl, pH 8.0 and 1% IGEPAL CA630) containing a cocktail of proteinase inhibitors (Roche). Cleared lysate were resolved on SDS–PAGE gel and transferred to PVDF membrane. Protein expression was detected by enhanced chemiluminescence following incubations of various antibodies. The following antibodies were used: anti-ATM (MAT3), 1:1000 (kindly provided by Y.Shiloh); anti-Mcm2 (Santa Cruz, N-19, sc-9839), 1:200; anti-Smc1 (Novus, NB-100-204), 1:5000; AhR (Santa Cruz, H-211, sc-5579), 1:500; anti-H-Ras (Santa Cruz, F235, sc-29), 1:500; anti-Raf (Pharmingen, Cat#13981A), 1 mg/ml; anti-MAPKpT202/ Y204 (Cell Signaling, #9101), 1:1000; anti-MAPK (ERK2, Santa Cruz, C-14, sc-154), 1:400; anti-p53-S15 (Cell Signaling, #9284), 1:1000. For quantitative analysis, the scanned gel images were analyzed by ImagePro 5.0 software, and relative levels were normalized against the expression levels of b-actin.

Tumor genomic DNA isolation and Ras mutation assay

Following NP40 extraction of tumor sample as described above, insoluble pellet was briefly rinsed in PBS, and 200ml of lysis buffer (100 mM NaCl, 10 mM EDTA, 20 mM Tris–HCl, pH 7.5 and 0.5% SDS) containing 100mg/ml proteinase K (Roche) was added followed by overnight incubation at 50C.

Saturated NaCl (100ml) was then added to the samples and this was followed by vigorous shaking for 5 min. Tumor samples were spun at 6000 g for 10 min after sitting on ice for 10 min. Supernatant was transferred to a new tube and tumor genomic DNA was precipitated by addition of 100% ethanol. Tumor DNA was dissolved in TE and was used for PCR assay directly without further treatment.

Detection of H-Ras mutation at codon 61 was done essentially as described, with modification (24). In brief, following amplification of exon 2 of HRas, DNA sample was digested with XbaI for 2 h at 37C. Digested samples were

resolved onto 5% polyacylamide gel in 1· TBE buffer to distinguish the parental DNA (207 bp) and the mutated HRas gene (116 and 91 bp) along with thefX174DNA/HaeIII molecular weight marker (Promega).

Atm LOH analysis of mammary tumor DNA was identical to that described for mice genotyping. LOH in the Atm heterozygotes tumor samples would be revealed by the disappearance of 125 bp wild-type PCR product. Sod1 locus was also examined as PCR control with the following primers: SODKOEX4 50 -gaacatcgtgtgatctcactgtcaggagag-30; SODKOEX5 50

-caagcggctcccagcatttc-cagtctttgt-30. Sod1 primers were kindly provided by Dr. Y.Ho. Isolation of mammary epithelial cell lines

Isolation of epithelial cells from mouse mammary gland was similar to that described previously (25). Briefly, the mammary tissues from wild-type or Atm+/mice were chopped fine and the pieces were suspended in DMEM/F12 (Invitrogen) containing 100 U/ml hyaluronidase (Sigma) and 150 U/ml collagenase (Sigma). After overnight incubation at 37C, the organoids were transferred to flasks in attachment medium (DMEM/F12 with 10 mM HEPES, 20% calf serum, pen/strep and Fungizone). After attachment of a significant number of organoids, the flasks were refed with high-calcium growth medium [DMEM/F12 with 10 mM HEPES, 1.05 mM CaCl2, 5%

horse serum (GIBCO), 10mg/ml insulin (Sigma), 20 ng/ml EGF (Upstate Biotechnology), 0.5mg/ml hydrocortisone (Sigma) and 100 ng/ml cholera toxin (Calbiochem)] to allow cells to spread over the plastic. After the attached pieces had started to grow out (about 10 days), the cells were stripped of fibroblasts by a short trypsinization and refed in low calcium growth medium (containing 0.04 mM CaCl2) to start cells growing as floaters. Within a month,

growth in this medium and production of floaters became extremely rapid. After 8 weeks some cells were transferred back into high-calcium medium to determine whether they had spontaneously immortalized. Wild-type (WTME1, WTME2) and Atm+/(AHME1, AHME2) mammary epithelial cell lines were maintained in mammary media (2.5% fetal bovine serum, 5% horse serum, 10 mg/ml insulin, 5 ng/ml EGF and 1· penicillin/streptomycin in DMEM/F-12 media).

Clonogenic survival assay

Clonogenic survival assay was done essentially as described (26), with modi-fication. In brief, exponentially growing cell lines were seeded in triplicate at 300 cells/per 6-well dish and irradiated at the indicated dose the day after plating. Radiation was performed using a137Cs source (Mark 1 Irradiator, J.L.Shepherd & Sons, San Bernardino, CA) with a dose rate of 1.1 Gy/min. The surviving clones were fixed in cold methanol and stained with 0.1% crystal violet 2 weeks post-radiation. The survival fractions were expressed as a percentage of number of irradiated survival clones divided by the number of non-irradiated surviving clones.

Statistical analysis

Mammary tumor-free survival was performed with Kaplan–Meier analysis. Log-rank test was performed to determine statistical significance. A P-value of<0.05 was considered to be statistically significant. The significance of mammary dysplasia in Atm+/mice compared with wild-type was performed by Poisson regression analysis.

Results

In an effort to better understand the function of Atm in

mam-mary tumor development, we generated a congenic strain of

Atm knockout mice by backcrossing 129SvEv x Black Swiss

Atm

tm1Led

mice (20) into FVB/N congenic background for 13

generations. Reciprocal mammary gland transplantation can be

performed with this new strain, which we referred to as

FVB.129S6(BLKSW)-Atm

tm1Led

(Lu and Wang, unpublished

data). To test the hypothesis that A-T heterozygotes are

predisposed to mammary tumorigenesis and to identify the

environmental factors that may influence the development

of breast cancer of A-T heterozygotes in the general

popula-tion, we performed the following experiments with mice

and cells derived from FVB.129S6(BLKSW)-Atm

tm1Led

(Atm

+/

hereafter).

Wild-type and Atm

+/

nulliparous female mice were treated

with DMBA at 6 weeks of age, and mammary tumor

devel-opment was monitored weekly by palpation. Once tumor mass

was identified, the mice were killed and the tumors dissected

for histopathological analysis (Table I). Nearly twice as

many

Atm

heterozygotes

developed

mammary

tumors

(64.7%) as the wild-type mice (37.5%). The relative risk

factor for DMBA-induced mammary tumors is 1.7 for Atm

heterozygotes. Histopathological analysis of tumor samples

indicates a wide variety of mammary tumor histological

types in both wild-type and Atm heterozygotes (Table I and

data not shown). Most importantly, Atm heterozygotes

developed mammary tumors earlier with an average onset

of 189 days compared with 229 days for wild-type mice

(Figure 1A, P

< 0.005 by log-rank test). In contrast, control

untreated wild-type (n

¼ 6) and Atm

+/

(n

¼ 11) mice did

not develop mammary tumors over a 1-year period (data

not shown). Interestingly, following DMBA treatment,

ovarian tumors were observed in both wild-type and Atm

+/

mice, whereas uterine tumors were observed in some of the

Atm

+/

mice but not in wild-type mice (Table I).

Since tissue dysplasia is an early event during

tumorigen-esis, the extent of DMBA-induced dysplasia was determined in

the mammary glands of the treated mice. In these experiments,

free mammary glands were harvested from

tumor-bearing mice and processed for whole mount analysis. Poisson

regression analysis was performed to determine whether

mammary dysplasia might be affected by mammary tumor

development. Our analysis suggests that mammary tumor

development had no effect on mammary dysplasia of adjacent

glands (P

¼ 0.169, Chi-Square test). Over 56% (n ¼ 53) of the

(3)

Atm

+/

mammary glands exhibited severe dysplasia, compared

with 21% (n

¼ 28) of the wild-type mammary glands that

showed only mild dysplasia (Figure 1B and Figure 2, P

¼

0.0248 by Poisson regression analysis). The dysplastic

mam-mary structures in the wild-type mice were more localized

by whole mount analysis and exhibited a single epithelial

layer by histological analysis (Figure 2A–D). For example,

after DMBA treatment, 58% of the Atm heterozygotes with

dysplastic mammary glands appeared to be diffusive, whereas

only 11% of the wild-type with dysplastic mammary glands

had such feature (Figure 2E–L, data not shown).

Histopatho-logical analysis revealed the presence of various stages of

mammary tumorigenesis including hyperplastic alveolar

nodule (HAN) (Figure 2M–P) and mammary intraductal

neoplasia (MIN) (Figure 2G–H) in Atm

+/

mammary glands.

Although increased mammary dysplasia in Atm

+/

mice

could contribute to enhanced mammary tumorigenesis in

those mice, it is not clear whether mammary tumors from

Atm

+/

mice exhibit loss of heterozygosity (LOH) at the

Atm locus. To further characterize the enhanced

carcinogen-induced mammary tumor development in Atm

+/

mice,

genomic DNA from three wild-type and five Atm

+/

mammary

tumors was harvested from fresh frozen tissues. PCR analysis

was performed to determine whether LOH occurred in tumors

from Atm

+/

mice. Our results demonstrated that the wild-type

Atm allele was retained in all five mammary tumors analyzed

(Figure 3A). It is possible that Atm LOH might have occurred

in the tumors and that wild-type PCR products detected may be

derived from normal cells intermingling with tumor cells.

Therefore, we compared PCR products derived from

wild-type and Atm

+/

mammary tissues with that derived from

tumors and found that the intensity of the wild-type bands

were comparable in tissues or tumors derived from Atm

+/

mice (Figure 3B). Moreover, Atm expression was clearly

detectable in all five mammary tumor samples and three

ovar-ian tumor samples derived from Atm

+/

mice (Figure 3C).

Therefore,

increased

mammary

tumor

development

in

Atm

+/

mice is not due to loss of Atm expression from the

wild-type allele. Collectively, these results suggest that

Atm-haploinsufficiency confers increased sensitivity to chemical

carcinogenesis. Interestingly, Mini-Chromosome Maintenance

protein 2 (Mcm2), which was recently described as a target of

ATM/ATR (27,28), was strongly expressed in three of the five

Atm

+/

mammary tumors but none in wild-type tumors.

Fur-thermore, Mcm2 expression was only weakly detected in one

of the three Atm

+/

ovarian tumors (Figure 3B). In addition, the

expression of Structural Maintenance Chromosome 1 (Smc1),

another downstream target of Atm (29,30), appears to be

positively correlated with the levels of Atm protein except

in one wild-type tumor (#2) expressing high amount of

Table I. DMBA-induced tumors in wild-type and Atm+/mice

Age (days) Tumor pathology Mice, Atm+

1 112 Lymphoma

2 142 Mammary adenocarcinoma 3 168 Mammary adenocarcinoma 4 169 Mammary adenosquamous tumor,

uterine adenocarcinoma 5 171 Mammary adenosquamous tumor 6 183 Mammary adenoma

7 185 Mammary papillary tumor 8 202 Mammary cribriform tumor

9 204 Mammary solid tumor, uterine adenocarcinoma 10 205 Mammary adenocarcinoma

11 206 Mammary adenocarcinoma 12 208 Uterine squamous cell carcinoma 13 212 Ovarine granulosa cell tumor 14 215 Ovarine granulosa cell tumor

15 253 Lung adenocarcinoma, gastric adenocarcinoma 16 254 Mammary squamous carcinoma and

mammary solid tumor

17 263 Uterine adenosquamous carcinoma, lung adenocarcinoma

Mice, WT

1 218 Mammary adenocarcinoma 2 232 Mammary adenosquamous tumor 3 237 Mammary solid undifferentiated tumor

and ovary granulosa cell tumor 4 272 Ovarine granulosa cell tumor 5 272 Ovarine granulosa cell tumor

6 272 No tumor

7 272 No tumor

8 272 No tumor

Six wild-type tumors and 22 Atm+/tumors were analyzed. Three of the eight wild-type mice developed mammary tumors compared with 11 of the 17 Atm+/mice.

Fig. 1.Tumor incidence and mammary dysplasia in wild-type and Atm+/ mice. (A) Kaplane–Meier survival analysis of mammary tumor-free mice. DMBA (Sigma) was dissolved in olive oil at a concentration of 5 mg/ml and administrated to mice6 weeks of age by oral gavages at a dose of 1 mg once a week for five consecutive weeks. Wild-type mice, (n¼ 8); Atm+/mice, (n¼ 17). P < 0.005 by log-rank test. (B) Increased mammary dysplasia in Atm+/mice compared with wild-type mice. Whole mount analysis of mammary glands from wild-type or Atm+/mice treated with DMBA. Number of mammary glands analyzed; wild-type mice, n¼ 28; Atm+/mice, n¼ 53. On the basis of the Poisson regression model, the estimated coefficient is 0.9988, which is significant with P-value of 0.0248. It is indicated that Atm+/mice had higher rate of dysplasia.

(4)

Atm. The levels of Atm protein in these tumors were not due to

differential protein loading since Aryl hydrocarbon Receptor

(AhR) and Raf were all expressed similarly in these tumors

(Figures 3B and 4B). It has been demonstrated recently that

ATM is epigenetically silenced in advanced breast cancers (4).

It is possible that variable expression of Atm in the mammary

tumors may also be under epigenetic regulation.

It was shown previously that DMBA-induced skin

carcino-genesis was due to mutation of the Hras gene at codon 61,

which could be detected in over 90% of the DMBA-induced

skin tumor samples (31,24). In addition, mammary tumors

induced by DMBA appear to be correlated with ras mutations

(32–34). To determine whether mammary tumor induced

by DMBA harbours Hras mutation at codon 61 in our

experimental model, an allele-specific Hras mutation assay

was performed (24). This assay takes advantage of the fact

that DMBA-induced Hras mutation creates a novel XbaI

restriction site, which could be conveniently employed to

dis-tinguish the wild-type allele from mutated Hras allele after

PCR amplification of sequence surrounding codon 61. Our data

clearly indicate that Hras mutation at codon 61 was not

involved in either DMBA-induced mammary or ovarian

tumor development since all tumors from wild-type and

Atm

+/

mice display only wild-type Hras allele following

XbaI digestion of the PCR products (Figure 4A). Although

we were unable to detect Hras mutation at codon 61, the

presence of other activating mutations in Ras gene cannot

be excluded. Since Ras mutations would activate

mitogen-activated protein kinase (MAPK) signaling pathway, we

therefore determined the expression of activated MAPK in

these mammary tumors. As shown in Figure 4B,

phosphoryla-tion of MAPK at Thr202 and Tyr204 was variably observed

in mammary and ovarian tumors derived from both wild-type

and Atm

+/

mice. In a limited comparison of Ras signaling

between mammary gland and mammary tumors from the same

wild-type or Atm

+/

mice, we also could not find correlative

evidence of Ras activation and mammary tumor development

Fig. 2. Whole mount and histopathological analysis of mammary dysplasia. (A–D) Wild-type mammary gland. (E–P) Atm+/mammary gland. (A, B, E, F, I, J, M and N) Whole mount analysis of mammary gland. (C, D, G, H, K, L, O and P) Histopathological analysis of the corresponding whole mount mammary gland. (G and H) Representative images showing MIN. (K and L) Representative images of ductal epitheliums hyperplasia were presented. (M–P) Representative images indicating the presence of HAN. Magnifications, A, E, I, M¼ 7.5·; B, F, J, N ¼ 20·; C, G, K, O ¼ 100·; D, H, L, P ¼ 200·.

Fig. 3. AtmLOH analysis in DMBA-induced tumor samples. (A) PCR genotyping of Atm locus in three wild-type mammary tumors and five Atm+/mammary tumors. Three ovarian tumors from Atm+/mice were also examined for Atm LOH. Mut, Atm mutant genotype; WT, wild-type genotype. All tumors retain wild-type Atm, demonstrating no LOH at Atm locus. Control PCR analysis of Sod1 was also performed. (B) PCR genotyping of Atm locus in genomic DNA derived from mouse tails, mammary gland and mammary tumors. (C) Nuclear protein expression in mammary and ovarian tumors by immunoblot analysis. Mcm2 expression can only be detected in some of the mammary tumors derived from Atm+/ mice but none in the tumors from wild-type mice. The expression of Smc1 was strongly elevated in three of the mammary tumors from Atm+/mice but only weakly in one of the three wild-type tumors. The Atm monoclonal antibody used here is not expected to recognize any truncated Atm protein in this mouse strain.

(5)

since phosphorylated MAPK was expressed at a lower level in

the Atm

+/

mammary tumors than that in the mammary glands

(Figure 4C). Although our data did not provide evidence for

a maintenance role of Ras activation in DMBA-induced

mammary tumors, we cannot exclude the possibility that

Ras activation may be required for the initiation of

DMBA-induced mammary tumor development.

It was shown previously that ionizing radiation induces

mammary dysplasia in about 10% of the Atm

+/

mice although

the mechanism of dysplasia induction was not determined (35).

To further characterize the molecular mechanism of

mam-mary tumor development in an Atm

+/

background, we

gen-erated mammary epithelial cell lines from Atm

+/

(AHME1

and AHME2) and wild-type control (WTME1 and WTME2)

littermates. Both wild-type and Atm

+/

cell lines display

epithelial morphology and express pan-cytokeratin marker,

confirming the nature of mammary epithelial cells (data not

shown). To determine whether Atm-haploinsufficiency affects

DNA damage signaling, we examined the induction of p53 in

both wild-type and Atm

+/

mammary cells in response to DNA

damage. Interestingly,

2-fold increased p53

phosphoryla-tion at serine 18 (equivalent to human Ser15) was detected

in AHME1 (2.07 ± 0.58, n

¼ 3) and AHME2 (3.1-fold) Atm

+/

mammary cell lines without DNA damage (Figure 5),

suggest-ing that Atm-haploinsufficiency elevated endogenous DNA

damage in unstressed cells. However, upon DNA damage,

Ser18 phosphorylation was detected to a similar extent in

both wild-type and Atm

+/

mammary epithelial cells in a

range of doses and time points examined (Figure 5). The

expression of Atm was verified in these cells and it was

con-firmed that Atm

+/

cells expressed half the amount of Atm protein

found in wild-type cells (Figure 6A). Interestingly, a slightly

decreased expression of Atm was observed in wild-type

mam-mary epithelial cells in response to irradiation, while a slightly

increased expression of Atm was seen in Atm

+/

mammary

epi-thelial cells after normalization to the expression levels of

b-actin

(Figure 6A).

One of the major features of A-T patients is increased

sensitivity to radiation (3). At the cellular level, the increased

radiation sensitivity can be determined by clonogenic survival

assay. To determine whether Atm-haploinsufficiency could

confer radiosensitivity in mammary epithelial cells, we

per-formed clonogenic survival assay in response to various doses

of ionizing radiation in both wild-type and Atm

+/

mammary

cell lines. Surprisingly, we found that both Atm

+/

mammary

epithelial cell lines (AHME1 and AHME2) were less

sensitive than wild-type mammary epithelial cell lines

(WTME1 and WTME2) to radiation in this clonogenic

survival assay (Figure 6B). Hence, Atm-haploinsufficiency

Fig. 4.Ras signaling in DMBA-induced mammary tumors. All tumor

sample numberings were identical to Figure 3. (A) DMBA-induced mammary and ovarian tumors do not contain activating H-Ras mutation at codon 61. PCR-amplified DNA was digested by XbaI DNA restriction enzyme to distinguish mutated H-Ras from normal H-Ras. WT indicates a 216 bp PCR product of the normal H-Ras on a 5% polyacrylamide gel stained with ethidium bromide. (B) Ras signaling pathway was not altered in mammary tumors derived from either wild-type or Atm+/mice. The expression of Ras, Raf, activated MAPKpT202/Y204 and MAPKp44/42 was determined by immunoblot analysis. (C) Comparison of Ras signaling in mammary gland (MG) and mammary tumor (MT) derived from wild-type and Atm+/mice. Immunoblot analysis was performed as in (B).

Fig. 5.The induction of p53 in response to radiation in Atm+/mammary epithelial cell lines. Wild-type (WTME1, WTME2) and Atm+/(AHME1, AHME2) mammary epithelial cell lines were established from respective mouse mammary gland. Active proliferating cells were irradiated at given doses, and protein extracts were prepared at the time indicated.

Phosphorylated p53 at serine 18 was examined in these cells.b-actin was used as loading control. (A–C) WTME1 and AHME1 cells were irradiated at 1 Gy (A) or 8 Gy (B) of various time points, or (C) irradiated at various doses for 30 min. (D) WTME2 and AHME2 cells were irradiated at given doses and the expression of phosphorylated p53 was determined at indicated time points.

(6)

confers enhanced survival in response to radiation in

mam-mary epithelial cells.

Discussion

It was estimated that 1% of the general population are A-T

carriers who are at an increased risk of breast cancer

devel-opment (6). This hypothesis is supported by the observations

that obligate A-T carriers are more likely to develop breast

cancer at an early age (8,10–12). The majority of the mutations

in A-T patients are truncating mutations, whereas the

remain-ing are missense mutations (1,15). However, the relative

contribution of the truncating and missense mutations of

ATM to breast cancer development in A-T family is not

clear. It is inferred that blood relatives of A-T patients

would most probably have truncating mutations that

predis-pose this population to breast cancer. However, since only a

small percentage of these A-T carriers develop breast cancer at

an early age, it is also probable that missense ATM mutations

preferentially contributed to breast cancer in relatives of A-T

patients. Gatti et al. (15) had proposed a model suggesting

that missense mutations of ATM predispose to cancer in the

heterozygotic or homozygotic state in the general population.

It was shown previously that 7271T>G and IVS10-6T>G

missense mutations have been associated with multiple

cases of breast cancers in the A-T family and in the non-A-T

Australia family (36). However, the association of these two

mutations with breast cancer was not supported by large-scale

family-history-based or population-based screening for these

two mutations in breast cancer patients (37,38). In addition,

ATM mutations are more frequently associated with early

onset familial breast cancer patients (5,39) while these studies

have not been replicated by others (40,41). To reconcile these

differences, we propose that the expression of ATM mutations

is modified by environmental factors, such as local

environ-mental carcinogen exposure or a difference in dietary intake.

Animal models for both truncating mutations and missense

mutations have been developed (17–20,42). Using these

models, it was shown that Atm

+/

mice with truncating

muta-tions are more sensitive to radiation-induced aging problems,

such as cataracts and hair graying (13,14). Mammary epithelial

cells from Atm heterozygotes with truncating mutations appear

to be sensitive to radiation-induced mammary dysplasia

in 10% of the transplanted tissues (35). Interestingly, Atm

+/



mice with truncating mutations have not been shown to

develop mammary tumors. The difference in the occurrence

of mammary tumors between mouse and human in the Atm

+/

background is not clear. However, one important difference

is that all animals studied are housed in similar controlled

environment and fed similar diets. To account for this

differ-ence, we propose that A-T carriers who developed breast

cancers might have encountered additional mutagenic events

from exposure to environmental toxic compounds through

dietary consumption.

A mouse model with missense mutation (ATM-DSRI) in

which three amino acids of ATM at positions 2556–2558

have been deleted has been developed recently (42). This

model is representative of missense mutations observed in

some A-T patients where full-length ATM protein can be

detected. However, ATM protein kinase activity is lost in

this mutant protein (42). Interestingly,

9% of the

heterozy-gotes of ATM-DSRI mice developed tumors, a 3-fold increase

over that of the wild-type mice (43). However, only 2% of the

ATM-DSRI heterozygotes developed mammary tumors (43),

suggesting that additional factors are required for mammary

tumor development in this model. It will be interesting

to determine whether ATM-DSRI mice are also sensitive to

chemical carcinogen-induced mammary tumorigenesis.

In Atm

+/

mice treated with ethylnitrosourea (ENU, a direct

alkylating agent that is thought to produce DNA lesions

with-out DNA double-strand breaks), no increased mammary

tumors were observed within 100 days of treatment (44).

The inability to detect increased tumors induced by ENU in

Atm

+/

mice may be due to a short incubation time or that

Atm

+/

mammary epithelial cells are not sensitive to ENU

treatment. Umesaka et al. (45) have recently reported

that

Atm-haploinsufficiency

promotes

mammary

tumor

development in a p53 heterozygotic background and that

the Atm-haploinsufficiency effect was further enhanced by

ionizing radiation. However, it is not clear whether mammary

dysplasia occurred in this cohort. The importance of ATM in

suppressing breast cancer development is also supported by a

recent study demonstrating that ATM is epigenetically silenced

in advanced breast cancer (4).

In this study, we found that truncating Atm

+/

mice had a

2-fold increased mammary tumor incidence in the

DMBA-induced mammary tumorigenesis model. In addition, we

found that there was about a 3-fold increase in mammary

dysplasia induced by DMBA treatment in Atm

+/

mice

Fig. 6. Atmheterozygotes displayed enhanced survival to radiation.

(A) Atm expression in wild-type (WTME2) or Atm+/(AHME1) mammary epithelial cell line in response to different doses of irradiation. The expression of Atm was determined 1 h post-radiation. The relative expression levels were quantitated using ImagePro 5.0 software after normalization againstb-actin, and fold differences compared with the level of Atm in non-irradiated wild-type cells were shown at bottom of the gel. (B) Mammary epithelial cell lines (WTME1, WTME2, AHME1 and AHME2) were irradiated at given doses and survival clones were scored after 2 weeks in culture. Survival fractions were expressed as percentage of irradiated survival clones normalized to non-irradiated survival clones. The experiment was performed in triplicate and results were presented as mean ± SEM.

(7)

compared with wild-type mice. Importantly, there was more

severe mammary dysplasia in Atm heterozygotes than in

wild-type mice (see Figure 2E–L), which is consistent with

the notion that DMBA may interfere with the hormone

response pathway (46,47). Mammary dysplasia appears to

be specifically associated in mice treated with either DMBA

(this study) or ionizing radiation (35) since untreated Atm

+/

mice do not develop this phenotype (data not shown, also see

ref. 35). The severe mammary dysplasia observed in Atm

heterozygotes implies that Atm

+/

mammary epithelial cells

are more sensitive to hormone-induced proliferation than

wild-type cells. Therefore, increased tumor incidence and dysplasia

in Atm heterozygotes may be partly explained by the combined

actions of increased mutation accumulations (deficient DNA

repair system) and increased sensitivity to hormone-induced

proliferation of mammary epithelial cells. DMBA causes bulky

DNA–protein adducts formation, which could be repaired

by base/nucleotide excision repair. Activation of Ras pathway

has been implicated in DMBA-induced carcinogenesis in

gen-eral (32–34,48,49). The possibility of DMBA-induced Ras

mutation in the initiation of mammary tumorigenesis was

evaluated in this study. Our results suggest that Ras mutation

at codon 61 was not involved in DMBA-induced mammary

tumors irrespective of the status of Atm. However, it is likely

that DMBA may also induce other Ras mutations. If true, then

Ras mutations would activate downstream signaling, and such

events should be detected. Surprisingly, examination of Raf

and MAPK levels did not provide such evidence. Therefore,

our results suggest that DMBA induces distinct signaling

path-ways in tumor progression dependent on cell types. However,

we could not exclude the possibility that activation of Ras

signaling pathway is involved in the initiation of mammary

carcinogenesis but is not required for the maintenance once

mammary tumor mass formed. Alternatively, DMBA-induced

DNA-adducts may interfere with replication fork movement

and therefore may cause replication fork collapse. Restart of

the replication fork is thought to invoke a DNA recombination

step induced by double-strand breaks. It is possible that in Atm

heterozygotes the restart of the replication fork progression is

defective, thereby leading to the accumulation of mutations

and chromosome instability. Consistent with this

interpreta-tion, DMBA and other bulky-adduct-forming agents, such as

PhIP (2-amino-1-methyl-6-phenylimidazo[4,5-b] pyridine),

have been shown to induce aneuploidy in Chinese hamster

cells and colon cancer cells, respectively (50,51).

Mammary epithelial cells from Atm heterozygotes might be

deficient in the correction of environmental mutagenic insults

and hence predisposed to breast cancer. Consistent with this

hypothesis, we found that Atm

+/

mammary epithelial cells

exhibit elevated basal levels of p53 phosphorylation compared

with that in wild-type cells. Constitutive induction of p53

phosphorylation in Atm

+/

mammary cells indicates that

these cells are constantly under low levels of stresses compared

with wild-type cells. Therefore, Atm-haploinsufficiency

con-fers elevated cellular stresses in mammary epithelial cells.

Atm-deficient cells exhibit increased radiation sensitivity.

The cause of radiation sensitivity in Atm-deficient cells is not

well understood although both cell cycle checkpoints and DNA

repair defects could contribute to such effect (26,52). It was

shown previously that ATM heterozygotic lymphocytes

showed intermediate radiation sensitivity using a short-term

survival assay (43,53), whereas radiation sensitivity was not

observed in mouse Atm

+/

ES cells using a clonogenic survival

assay (18). In this study, we were therefore surprised to found

that Atm-haploinsufficiency confers slightly enhanced survival

in mammary epithelial cells in response to DNA damage. The

variations in radiation sensitivity in these different cell

types are not clear at present but could be dependent on cell

context and/or due to the history of cellular stresses.

Atm-haploinsufficiency may predispose mammary epithelial cells

to genome instability and therefore leads to enhanced

tumori-genesis in response to chemical carcinogens. In summary, our

results suggest that gene–environmental interactions may

dic-tate the breast cancer development in ATM heterozygotes.

Acknowledgements

We thank Dr Y.Shiloh for Atm monoclonal antibody, Drs B.Reizis and A.Elson for Atm PCR primer sequences, and Dr Y.Ho for Sod1 PCR primers. This research was partly supported by Grants from Department of Defense (DAMD17-02-1-0619) and NIH (RO1 CA89526) and (P30 ES06639). Conflict of Interest Statement: None declared.

References

1. Concannon,P. (2002) ATM heterozygosity and cancer risk. Nat. Genet., 32, 89–90.

2. Hall,J. (2005) The ataxia-telangiectasia mutated gene and breast cancer: gene expression profiles and sequence variants. Cancer Lett., 227, 105–114. 3. Shiloh,Y. (2003) ATM and related protein kinases: safeguarding genome

integrity. Nat. Rev. Cancer, 3, 155–168.

4. Vo,Q.N., Kim,W.J., Cvitanovic,L., Boudreau,D.A., Ginzinger,D.G. and Brown,K.D. (2004) The ATM gene is a target for epigenetic silencing in locally advanced breast cancer. Oncogene, 23, 9432–9437.

5. Thorstenson,Y.R. and Roxas,A. and Kroiss,R. et al. (2003) Contributions of ATM mutations to familial breast and ovarian cancer. Cancer Res., 63, 3325–3333.

6. Athma,P., Rappaport,R. and Swift,M. (1996) Molecular genotyping shows that ataxia-telangiectasia heterozygotes are predisposed to breast cancer. Cancer Genet. Cytogenet., 92, 130–134.

7. Geoffroy-Perez,B., Janin,N., Ossian,K. et al. (2001) Cancer risk in hetero-zygotes for ataxia-telangiectasia. Int. J. Cancer, 93, 288–293.

8. Inskip,H.M., Kinlen,L.J., Taylor,A.M., Woods,C.G. and Arlett,C.F. (1999) Risk of breast cancer and other cancers in heterozygotes for ataxia-telangiectasia. Br. J. Cancer, 79, 1304–1307.

9. Janin,N., Andrieu,N., Ossian,K., Lauge,A., Croquette,M.F., Griscelli,C., Debre,M., Bressac-de-Paillerets,B., Aurias,A. and Stoppa-Lyonnet,D. (1999) Breast cancer risk in ataxia telangiectasia (AT) heterozygotes: haplotype study in French AT families. Br. J. Cancer, 80, 1042–1045. 10. Olsen,J.H., Hahnemann,J.M., Borresen-Dale,A.L. et al. (2001) Cancer in

patients with ataxia-telangiectasia and in their relatives in the nordic countries. [see comment]. J. Natl Cancer Inst., 93, 121–127.

11. Olsen,J.H., Hahnemann,J.M., Borresen-Dale,A.L. et al. (2005) Breast and other cancers in 1445 blood relatives of 75 Nordic patients with ataxia telangiectasia. Br. J. Cancer, 93, 260–265.

12. Thompson,D., Duedal,S., Kirner,J., McGuffog,L., Last,J., Reiman,A., Byrd,P., Taylor,M. and Easton,D.F. (2005) Cancer risks and mortality in heterozygous ATM mutation carriers. J. Natl Cancer Inst., 97, 813–822. 13. Barlow,C., Eckhaus,M.A., Schaffer,A.A. and Wynshaw-Boris,A. (1999) Atm haploinsufficiency results in increased sensitivity to sublethal doses of ionizing radiation in mice. Nat. Genet., 21, 359–360.

14. Worgul,B.V., Smilenov,L., Brenner,D.J., Junk,A., Zhou,W. and Hall,E.J. (2002) Atm heterozygous mice are more sensitive to radiation-induced cataracts than are their wild-type counterparts. Proc. Natl Acad. Sci. USA, 99, 9836–9839.

15. Gatti,R.A., Tward,A. and Concannon,P. (1999) Cancer risk in ATM heterozygotes: a model of phenotypic and mechanistic differences between missense and truncating mutations. Mol. Genet. Metab., 68, 419–423. 16. Meyn,M.S. (1999) Ataxia-telangiectasia, cancer and the pathobiology of

the ATM gene. Clini. Genet., 55, 289–304.

17. Herzog,K.H., Chong,M.J., Kapsetaki,M., Morgan,J.I. and McKinnon,P.J. (1998) Requirement for Atm in ionizing radiation-induced cell death in the developing central nervous system. Science, 280, 1089–1091.

(8)

18. Xu,Y., Ashley,T., Brainerd,E.E., Bronson,R.T., Meyn,M.S. and Baltimore,D. (1996) Targeted disruption of ATM leads to growth retarda-tion, chromosomal fragmentation during meiosis, immune defects, and thymic lymphoma. [see comment]. Genes Dev., 10, 2411–2422. 19. Barlow,C., Hirotsune,S., Paylor,R. et al. (1996) Atm-deficient mice: a

paradigm of ataxia telangiectasia. Cell, 86, 159–171.

20. Elson,A., Wang,Y., Daugherty,C.J., Morton,C.C., Zhou,F., Campos-Torres,J. and Leder,P. (1996) Pleiotropic defects in ataxia-telangiectasia protein-deficient mice. Proc. Natl Acad. Sci. USA, 93, 13084–13089. 21. Lydon,J.P., Ge,G., Kittrell,F.S., Medina,D. and O’Malley,B.W. (1999)

Murine mammary gland carcinogenesis is critically dependent on progesterone receptor function. Cancer Res., 59, 4276–4284.

22. Wang,Y., Shen,K., Wang,Y. and Brooks,S.C. (2005) Retinoic acid signaling is required for proper morphogenesis of mammary gland. Dev. Dyn., 234, 892–899.

23. Shen,K., Heng,H., Wang,Y., Lu,S., Liu,G., Deng,C.X., Brooks,S.C. and Wang,Y.A. (2005) Atm and p21 cooperate to suppress aneuploidy and subsequent tumor development. Cancer Res., 65, 8747–8753.

24. Nagase,H., Mao,J.H. and Balmain,A. (2003) Allele-specific Hras mutations and genetic alterations at tumor susceptibility loci in skin carcinomas from interspecific hybrid mice. Cancer Res., 63, 4849–4853.

25. Soule,H.D., Maloney,T.M., Wolman,S.R., Peterson,W.D.,Jr, Brenz,R., McGrath,C.M., Russo,J., Pauley,R.J., Jones,R.F. and Brooks,S.C. (1990) Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res., 50, 6075–6086. 26. Xu,B., Kim,S.T., Lim,D.S. and Kastan,M.B. (2002) Two molecularly

distinct G(2)/M checkpoints are induced by ionizing irradiation. Mol. Cell. Biol., 22, 1049–1059.

27. Yoo,H.Y., Shevchenko,A. and Dunphy,W.G. (2004) Mcm2 is a direct substrate of ATM and ATR during DNA damage and DNA replication checkpoint responses. J. Biol. Chem., 279, 53353–53364.

28. Cortez,D., Glick,G. and Elledge,S.J. (2004) Minichromosome maintenance proteins are direct targets of the ATM and ATR checkpoint kinases. [see comment]. Proc. Natl Acad. Sci. USA, 101, 10078–10083.

29. Yazdi,P.T., Wang,Y., Zhao,S., Patel,N., Lee,E.Y. and Qin,J. (2002) SMC1 is a downstream effector in the ATM/NBS1 branch of the human S-phase checkpoint. Genes Dev., 16, 571–582.

30. Kim,S.T., Xu,B. and Kastan,M.B. (2002) Involvement of the cohesin protein, Smc1, in Atm-dependent and independent responses to DNA damage. Genes Dev., 16, 560–570.

31. Mao,J.H., To,M.D., Perez-Losada,J., Wu,D., Del Rosario,R. and Balmain,A. (2004) Mutually exclusive mutations of the Pten and ras pathways in skin tumor progression. Genes Dev., 18, 1800–1805.

32. Kito,K., Kihana,T., Sugita,A., Murao,S., Akehi,S., Sato,M., Tachibana,M., Kimura,S. and Ueda,N. (1996) Incidence of p53 and Ha-ras gene mutations in chemically induced rat mammary carcinomas. Mol. Carcinog., 17, 78–83.

33. Kumar,R., Medina,D. and Sukumar,S. (1990) Activation of H-ras onco-genes in preneoplastic mouse mammary tissues. Oncogene, 5, 1271–1277. 34. Qing,W.G., Conti,C.J., LaBate,M., Johnston,D., Slaga,T.J. and MacLeod,M.C. (1997) Induction of mammary cancer and lymphoma by multiple, low oral doses of 7,12-dimethylbenz[a]anthracene in SENCAR mice. Carcinogenesis, 18, 553–559.

35. Weil,M.M., Kittrell,F.S., Yu,Y., McCarthy,M., Zabriskie,R.C. and Ullrich,R.L. (2001) Radiation induces genomic instability and mammary ductal dysplasia in Atm heterozygous mice. Oncogene, 20, 4409–4411.

36. Chenevix-Trench,G., Spurdle,A.B., Gatei,M. et al. (2002) Dominant negative ATM mutations in breast cancer families. [see comment] [erratum appears in J Natl Cancer Inst 2002 Jun 19; 94: 952] . J Natl Cancer Inst, 94, 205–215.

37. Szabo,C.I., Schutte,M., Broeks,A. et al. (2004) Are ATM mutations 7271T!G and IVS10-6T!G really high-risk breast cancer-susceptibility alleles? Cancer Res., 64, 840–843.

38. Bernstein,J.L., Bernstein,L., Thompson,W.D. et al. (2003) ATM variants 7271T>G and IVS10-6T>G among women with unilateral and bilateral breast cancer. Br. J. Cancer, 89, 1513–1516.

39. Teraoka,S.N., Malone,K.E., Doody,D.R., Suter,N.M., Ostrander,E.A., Daling,J.R. and Concannon,P. (2001) Increased frequency of ATM mutations in breast carcinoma patients with early onset disease and positive family history. Cancer, 92, 479–487.

40. Chen,J., Birkholtz,G.G., Lindblom,P., Rubio,C. and Lindblom,A. (1998) The role of ataxia-telangiectasia heterozygotes in familial breast cancer. Cancer Res., 58, 1376–1379.

41. FitzGerald,M.G., Bean,J.M., Hegde,S.R., Unsal,H., MacDonald,D.J., Harkin,D.P., Finkelstein,D.M., Isselbacher,K.J. and Haber,D.A. (1997) Heterozygous ATM mutations do not contribute to early onset of breast cancer. [see comment]. Nat. Genet., 15, 307–310.

42. Spring,K., Cross,S., Li,C. et al. (2001) Atm knock-in mice harboring an in-frame deletion corresponding to the human ATM 7636del9 common mutation exhibit a variant phenotype. Cancer Res., 61, 4561–4568. 43. Spring,K., Ahangari,F., Scott,S.P. et al. (2002) Mice heterozygous for

mutation in Atm, the gene involved in ataxia-telangiectasia, have heightened susceptibility to cancer. [see comment]. Nat. Genet., 32, 185–190.

44. Karabinis,M.E., Larson,D., Barlow,C., Wynshaw-Boris,A. and Moser,A.R. (2001) Heterozygosity for a mutation in Brca1 or Atm does not increase susceptibility to ENU-induced mammary tumors in Apc(Min)/+ mice. Carcinogenesis, 22, 343–346.

45. Umesaka,S., Fujisawa,K., Sayoko,L. et al. (2005) Atm heterozygous deficiency enhances development of mammary carcinomas in p53 hetero-zygous knockout mice. Breast Cancer Res., 7, R164–R170.

46. Safe,S., Wormke,M. and Samudio,I. (2000) Mechanisms of inhibitory aryl hydrocarbon receptor-estrogen receptor crosstalk in human breast cancer cells. J. Mammary Gland Biol. Neoplasia, 5, 295–306.

47. Klinge,C.M., Bowers,J.L., Kulakosky,P.C., Kamboj,K.K. and Swanson,H.I. (1999) The aryl hydrocarbon receptor (AHR)/AHR nuclear translocator (ARNT) heterodimer interacts with naturally occurring estrogen response elements. Mol. Cell. Endocrinol., 157, 105–119.

48. Balmain,A., Ramsden,M., Bowden,G.T. and Smith,J. (1984) Activation of the mouse cellular Harvey-ras gene in chemically induced benign skin papillomas. Nature, 307, 658–660.

49. Quintanilla,M., Brown,K., Ramsden,M. and Balmain,A. (1986) Carcinogen-specific mutation and amplification of Ha-ras during mouse skin carcinogenesis. Nature, 322, 78–80.

50. Bardelli,A., Cahill,D.P., Lederer,G., Speicher,M.R., Kinzler,K.W., Vogelstein,B. and Lengauer,C. (2001) Carcinogen-specific induction of genetic instability. [see comment]. Proc. Natl Acad. Sci. USA, 98, 5770–5775.

51. Li,R., Yerganian,G., Duesberg,P., Kraemer,A., Willer,A., Rausch,C. and Hehlmann,R. (1997) Aneuploidy correlated 100% with chemical transformation of Chinese hamster cells. Proc. Natl Acad. Sci. USA, 94, 14506–14511.

52. Kuhne,M., Riballo,E., Rief,N., Rothkamm,K., Jeggo,P.A. and Lobrich,M. (2004) A double-strand break repair defect in ATM-deficient cells contributes to radiosensitivity. Cancer Res., 64, 500–508.

53. Angele,S., Romestaing,P., Moullan,N. et al. (2003) ATM haplotypes and cellular response to DNA damage: association with breast cancer risk and clinical radiosensitivity. Cancer Res, 63, 8717–8725.

Received July 11, 2005; revised October 26, 2005; accepted December 6, 2005

Cytaty

Powiązane dokumenty

Ogólny wskaźnik akceptacji choroby w badanej gru- pie wyniósł średnio 25,35 ± 9,25 punktu, co interpretu- je się jako przeciętny poziom przystosowania do cho- roby nowotworowej..

The case is exceptional because the spleen is an organ where lung cancer metastases are not frequently found, while metas- tases to the adrenal gland alone are common.. More

stwierdzili zależność między wystąpieniem wariantu Cys/Cys a podwyższonym ryzykiem wystąpienia RJG [11]. Wykazali, że u osób młodych układ Cys/Cys przyczynia się do pod-

(…) An effective biological agent should not be switched to another bDMARD for non-medical reasons” [12]. Based on these recommendations, the Rheuma- tology Section of the

Wyniki: Uzyskane wyniki badania zależności między postawami i strategiami radzenia sobie z cho- robą nowotworową a jakością życia uwarunkowaną stanem zdrowia wskazują, że: 1)

On the basis of the examined data, some genes, such as GABA, the  serotonin 5-HTT transporter and the  dopamine receptor (DRD2), were discovered to be possibly connected

Do czynników środowiskowych mających najsilniejszy oraz udowodniony wpływ na rozwój UBC należą: palenie tytoniu, aminy aromatyczne, wielopierścieniowe węglowodory aromatyczne,

Wyższy niż w innych wariantach oraz niż w zdrowej tarczycy poziom ekspresji Met obserwuje się w wysokoko- mórkowym wariancie raka brodawkowatego oraz w rakach z