+ All Categories
Home > Documents > DNA Double-Strand Breaks, p53, and Apoptosis …...[CANCER RESEARCH 58, 3111-3115. July 15, 1998|...

DNA Double-Strand Breaks, p53, and Apoptosis …...[CANCER RESEARCH 58, 3111-3115. July 15, 1998|...

Date post: 31-Mar-2020
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
6
[CANCER RESEARCH 58, 3111-3115. July 15, 1998| DNA Double-Strand Breaks, p53, and Apoptosis during Lymphomagenesis in seid/seid Mice1 Kay E. Gurley, Khoa Vo, and Christopher J. Kemp2 Fred Hulchimon Cancer Research Center, Seattle. Washington 98109-1024 ABSTRACT The tumor-suppressing phenotype of p53 is thought to be due to its accumulation in response to DNA damage and resultant cell cycle arrest or apoptosis. seid/seid mice are defective in DNA double-strand break repair due to a mutation in DNA-dependent protein kinase (DNAPK). Treatment of seid/seid mice with y radiation or .V-i-lliyl-A'-iiitrosourea resulted in —¿86% incidence of T-cell lymphomas, compared with <6% in wild-type mice. The incidence of other tumor types was not increased in seid/seid mice, suggesting that the types of DNA double-strand break that are unrepaired in these mice are not strongly carcinogenic. To determine whether mutations in DNAPK and p53 interact, we examined mice defi cient in both genes. Both seid/seid p53—/—and seid/seid p53+/— mice spontaneously developed lymphomas at shorter latency than did mice with either defect alone. Loss of the wild-type p53 alÃ-elewas observed in 100% of tumors from seid/seid p53+/— mice, indicating strong selection against p53. In contrast, p53 was not inactivated in lymphomas from seid/seid p53+/+ mice. Exposure of these tumor-bearing mice to y radiation re sulted in p53 protein accumulation and high levels of apoptosis in all tumors that were not observed in tumors from seid/seid /«.?+/- mice. Thus, there was a bifurcation of molecular pathways to tumorigenesis. When p53 was heterozygous in the germ line, loss of the wild-type alÃ-ele occurred, and the tumors became apoptosis resistant. When p53 was wild type in the germ line, p53 was not inactivated, and the tumors remained highly apoptosis sensitive. INTRODUCTION The p53 tumor suppressor protein appears to be a central coordi nator of the cellular response to DNA damage. The levels of p53 protein are normally very low, but they increase in some cell types following exposure to DNA-damaging agents (1). DNA dsbs3 consti tute a major class of DNA lesions that lead to p53 accumulation (2). Some but not all cell types respond to these increased levels of p53 by arresting in G, phase of the cell cycle or by undergoing apoptosis (1, 3, 4). Cells with nonfunctional p53 fail to undergo either response, indicating that p53 is required (3, 5-7). Increased genetic instability is also observed in cells lacking functional p53, and these properties are believed to be central to its role as a tumor suppressor gene (8-12). In the majority of animal or human tumors that have sustained p53 mutations, the nature of the selective force against p53 function is not known. In mice, significant accumulation of p53 protein in response to whole-body y radiation is limited to thymic and splenic lympho cytes, osteocytes, a subset of keratinocytes, epithelial cells in the bottom of the small intestinal crypts, and, to a lesser extent, some other tissues (1, 3, 4, 13). However, most cells in the intact animal do not accumulate immunohistochemically detectable p53, and they do not undergo G, arrest or apoptosis in response to DNA damage. It is Received 12/31/97; accepted 5/18/98. 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. 1This work was funded in part by NIH Grant CA70414 (to C. J. K.). 2 To whom requests for reprints should be addressed, at Fred Hutchinson Cancer Research Center. Cl-015. 1100 Fairview Avenue North, P.O. Box 19024, Seattle, WA 98109-1024. Phone: (206)667-4252; Fax: (206)667-5815; E-mail: [email protected]. 3 The abbreviations used are: dsb, doubie-strand break; DNAPK, DNA-dependent protein kinase; DNAPK^, DNAPK catalytic subunit; SC1D, severe combined immuno- deficient; ENU, /V-elhyl-JV-nitrosourea; LOH, loss of heterozygosity; MNU, /V-methyl-Af- nitrosourea. not known whether tissues that cannot induce p53 select against p53 during tumorigenesis or whether p53 is selected against only in those tissues that are capable of accumulating high levels of p53. To address the roles of the DNA damage-p53 accumulation-apop- tosis pathway in tumorigenesis, we elected to study the seid/seid mutant mouse. These mice are defective in DNA dsb repair, and as a consequence, their cells are radiosensitive and developing lympho cytes cannot complete V(D)J antigen receptor gene rearrangement. The latter leads to maturation arrest of developing lymphocytes and absence of functionally mature T and B lymphocytes (14). The genetic defect in these mice was recently identified as a mutation in the gene encoding the DNAPKCS (15). This large protein was originally iden tified biochemically as a serine-threonine kinase that required double- stranded DNA ends for its activity. Activated DNAPK phosphorylates many proteins in vitro, although the relevant in vivo targets have yet to be identified. It is not known precisely how DNAPK participates in dsb repair, but following the generation of free DNA ends during V(D)J recombination or following DNA damage, the proteins Ku70 and Ku80 bind to the ends and recruit DNAPKCS, leading to its activation and subsequent rejoining of the DNA ends (16). The DNA dsb repair defect appears to exist in all cells of seid/seid mice because myeloid cells, fibroblasts, intestinal crypt cells, epithelial cells, sper- matogonial stem cells, and fibrosarcoma cells have all been shown to exhibit increased radiation sensitivity (17-20). The evidence described above, notably, the DNA dsb connection, predicts a functional relationship between DNAPK and p53. Although p53 protein is phosphorylated by DNAPK in vitro, p53 induction, G, cell cycle arrest, and apoptosis in response to DNA damage all occur normally in SCID cells and seid/seid mice, indicating that DNAPK is not a required regulator of these responses (13, 21, 22). In seid/seid mice, developing T lymphocytes are arrested at the double-negative CD4~CD8~ stage due to the failure to complete T-cell receptor rearrangement (23). When seid/seid mice were crossed to p53 knockout mice to generate seid/seid p53—/— mice, CD4+CD8+ cells were detected, indicating that p53 participates in the maturation arrest of seid T-lymphocyte precursors, and in the absence of p53, some cells are able to progress to the double-positive stage (23, 24). Additionally, mutations in p53 and DNAPK^ were shown to interact during tumorigenesis because seid/seid p53-/- mice developed lymphomas with reduced latency, as compared to either seid/seid or p53—/—mice alone (22, 23). These results are consistent with a model in which p53 would respond to unrepaired dsbs in SCID lymphocytes and block matura tion by cell cycle arrest or apoptosis (22, 23). In the absence of p53, these cells would survive and, thus, be at much greater risk for transformation. On the basis of the genetic interaction between DNAPK and p53 and current understanding of p53 function, one might expect a strong selective pressure to mutate p53 during tumor igenesis in seid/seid mice. Here, we show that spontaneous lympho mas rapidly developed in seid/seidp53+/- mice, and all these tumors had lost the remaining wild-type p53 alÃ-ele,confirming this predic tion. In contrast, all lymphomas examined from seid/seid p53+/+ mice retained the DNA damage-p53 accumulation-apoptosis pathway. Presumably, mutation or inactivation of a second, functionally unre- 3111 on April 8, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
Transcript
Page 1: DNA Double-Strand Breaks, p53, and Apoptosis …...[CANCER RESEARCH 58, 3111-3115. July 15, 1998| DNA Double-Strand Breaks, p53, and Apoptosis during Lymphomagenesis in seid/seid Mice1

[CANCER RESEARCH 58, 3111-3115. July 15, 1998|

DNA Double-Strand Breaks, p53, and Apoptosis during Lymphomagenesis inseid/seid Mice1

Kay E. Gurley, Khoa Vo, and Christopher J. Kemp2

Fred Hulchimon Cancer Research Center, Seattle. Washington 98109-1024

ABSTRACT

The tumor-suppressing phenotype of p53 is thought to be due to its

accumulation in response to DNA damage and resultant cell cycle arrestor apoptosis. seid/seid mice are defective in DNA double-strand breakrepair due to a mutation in DNA-dependent protein kinase (DNAPK).Treatment of seid/seid mice with y radiation or .V-i-lliyl-A'-iiitrosourea

resulted in —¿�86%incidence of T-cell lymphomas, compared with <6% inwild-type mice. The incidence of other tumor types was not increased inseid/seid mice, suggesting that the types of DNA double-strand break that

are unrepaired in these mice are not strongly carcinogenic. To determinewhether mutations in DNAPK and p53 interact, we examined mice deficient in both genes. Both seid/seid p53—/—and seid/seid p53+/— mice

spontaneously developed lymphomas at shorter latency than did mice witheither defect alone. Loss of the wild-type p53 alíelewas observed in 100%of tumors from seid/seid p53+/— mice, indicating strong selection against

p53. In contrast, p53 was not inactivated in lymphomas from seid/seidp53+/+ mice. Exposure of these tumor-bearing mice to y radiation re

sulted in p53 protein accumulation and high levels of apoptosis in alltumors that were not observed in tumors from seid/seid /«.?+/- mice.

Thus, there was a bifurcation of molecular pathways to tumorigenesis.When p53 was heterozygous in the germ line, loss of the wild-type alíele

occurred, and the tumors became apoptosis resistant. When p53 was wildtype in the germ line, p53 was not inactivated, and the tumors remainedhighly apoptosis sensitive.

INTRODUCTION

The p53 tumor suppressor protein appears to be a central coordinator of the cellular response to DNA damage. The levels of p53protein are normally very low, but they increase in some cell typesfollowing exposure to DNA-damaging agents (1). DNA dsbs3 consti

tute a major class of DNA lesions that lead to p53 accumulation (2).Some but not all cell types respond to these increased levels of p53 byarresting in G, phase of the cell cycle or by undergoing apoptosis (1,3, 4). Cells with nonfunctional p53 fail to undergo either response,indicating that p53 is required (3, 5-7). Increased genetic instability is

also observed in cells lacking functional p53, and these properties arebelieved to be central to its role as a tumor suppressor gene (8-12).

In the majority of animal or human tumors that have sustained p53mutations, the nature of the selective force against p53 function is notknown. In mice, significant accumulation of p53 protein in responseto whole-body y radiation is limited to thymic and splenic lympho

cytes, osteocytes, a subset of keratinocytes, epithelial cells in thebottom of the small intestinal crypts, and, to a lesser extent, someother tissues (1, 3, 4, 13). However, most cells in the intact animal donot accumulate immunohistochemically detectable p53, and they donot undergo G, arrest or apoptosis in response to DNA damage. It is

Received 12/31/97; accepted 5/18/98.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 with18 U.S.C. Section 1734 solely to indicate this fact.

1This work was funded in part by NIH Grant CA70414 (to C. J. K.).2 To whom requests for reprints should be addressed, at Fred Hutchinson Cancer

Research Center. Cl-015. 1100 Fairview Avenue North, P.O. Box 19024, Seattle, WA98109-1024. Phone: (206)667-4252; Fax: (206)667-5815; E-mail: [email protected].

3 The abbreviations used are: dsb, doubie-strand break; DNAPK, DNA-dependentprotein kinase; DNAPK^, DNAPK catalytic subunit; SC1D, severe combined immuno-deficient; ENU, /V-elhyl-JV-nitrosourea; LOH, loss of heterozygosity; MNU, /V-methyl-Af-

nitrosourea.

not known whether tissues that cannot induce p53 select against p53during tumorigenesis or whether p53 is selected against only in thosetissues that are capable of accumulating high levels of p53.

To address the roles of the DNA damage-p53 accumulation-apop-

tosis pathway in tumorigenesis, we elected to study the seid/seidmutant mouse. These mice are defective in DNA dsb repair, and as aconsequence, their cells are radiosensitive and developing lymphocytes cannot complete V(D)J antigen receptor gene rearrangement.The latter leads to maturation arrest of developing lymphocytes andabsence of functionally mature T and B lymphocytes (14). The geneticdefect in these mice was recently identified as a mutation in the geneencoding the DNAPKCS (15). This large protein was originally identified biochemically as a serine-threonine kinase that required double-

stranded DNA ends for its activity. Activated DNAPK phosphorylatesmany proteins in vitro, although the relevant in vivo targets have yetto be identified. It is not known precisely how DNAPK participates indsb repair, but following the generation of free DNA ends duringV(D)J recombination or following DNA damage, the proteins Ku70and Ku80 bind to the ends and recruit DNAPKCS, leading to itsactivation and subsequent rejoining of the DNA ends (16). The DNAdsb repair defect appears to exist in all cells of seid/seid mice becausemyeloid cells, fibroblasts, intestinal crypt cells, epithelial cells, sper-

matogonial stem cells, and fibrosarcoma cells have all been shown toexhibit increased radiation sensitivity (17-20).

The evidence described above, notably, the DNA dsb connection,predicts a functional relationship between DNAPK and p53. Althoughp53 protein is phosphorylated by DNAPK in vitro, p53 induction, G,cell cycle arrest, and apoptosis in response to DNA damage all occurnormally in SCID cells and seid/seid mice, indicating that DNAPK isnot a required regulator of these responses (13, 21, 22).

In seid/seid mice, developing T lymphocytes are arrested at thedouble-negative CD4~CD8~ stage due to the failure to complete

T-cell receptor rearrangement (23). When seid/seid mice were crossedto p53 knockout mice to generate seid/seid p53—/— mice,CD4+CD8+ cells were detected, indicating that p53 participates in

the maturation arrest of seid T-lymphocyte precursors, and in theabsence of p53, some cells are able to progress to the double-positive

stage (23, 24). Additionally, mutations in p53 and DNAPK^ wereshown to interact during tumorigenesis because seid/seid p53-/-

mice developed lymphomas with reduced latency, as compared toeither seid/seid or p53—/—mice alone (22, 23).

These results are consistent with a model in which p53 wouldrespond to unrepaired dsbs in SCID lymphocytes and block maturation by cell cycle arrest or apoptosis (22, 23). In the absence of p53,these cells would survive and, thus, be at much greater risk fortransformation. On the basis of the genetic interaction betweenDNAPK and p53 and current understanding of p53 function, onemight expect a strong selective pressure to mutate p53 during tumorigenesis in seid/seid mice. Here, we show that spontaneous lymphomas rapidly developed in seid/seidp53+/- mice, and all these tumors

had lost the remaining wild-type p53 alíele,confirming this predic

tion. In contrast, all lymphomas examined from seid/seid p53+/+mice retained the DNA damage-p53 accumulation-apoptosis pathway.Presumably, mutation or inactivation of a second, functionally unre-

3111

on April 8, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 2: DNA Double-Strand Breaks, p53, and Apoptosis …...[CANCER RESEARCH 58, 3111-3115. July 15, 1998| DNA Double-Strand Breaks, p53, and Apoptosis during Lymphomagenesis in seid/seid Mice1

LYMPHOMA(il;Nl:SIS IN SCIO MICK

lated pathway that did not involve loss of the DNA damage-apoptosis

response was favored if p53 was wild type in the germ line.

MATERIALS AND METHODS

Tumor Induction. Balb/cByJSmn scid/l, C3HSmn.C-.cciW/J, Balb/cByJ.

and C3H mice were purchased from The Jackson Laboratory and ¡>53deficientmice were obtained from Larry Donehower (25) and bred in-house. Allseid/seid mice were maintained in germ-free microisolator cages and fed

autoclaved food and water ad lihiiiiin. Balb/cByJSmn scid/l mice were crossedto C3HSmn.C-w/W/J to generate experimental C3C F, scid/scid/1 mice. Similarly, Balb/cByJ mice were crossed to C3H mice to generate C3C F, wild-type

controls. For the radiation experiment, mice were exposed between 24 and 48 hafter birth to 1 Gy ( 100 rad) of whole-body y radiation from a l37Cs source at

a dose rate of 330 cGy/min. For the ENU experiment, mice were injected at12-14 days of age with ENU i.p. (Sigma Chemical Co., St. Louis, MO; 0.5

/ig/g body weight) dissolved in trioctanoin (Arcos). Mice were observed dailyand sacrificed when they showed signs of tumor development. To generatedouble-deficient mice, CiHSmn.C-scid mice were crossed to C57BL6/Jp53—/- mice to generate C3B6 F, scid/+ p53+/- mice, which were intercrossed to generate F2 mice. The seid/seid p53+/- mice from this cross were

crossed again to generate seid/seid mice of all three p53 genotypes: thus, theyare of mixed C3H and C57BL/6J background. The seid/seid mice were

identified by examining peripheral blood smears for mature lymphocytes, and/>5J genotype was determined by PCR analysis of toe DNA (26).

Tumor Analysis. Tumor tissue was both frozen and fixed in formalin forroutine processing and staining with H&E. In vivo apoptotic index wasdetermined by irradiating tumor-bearing mice with 4 Gy of y radiation,

sacrificing the mice at several time points, and preparing tissues as above. Theapoptotic index was the mean value of observed apoptotic bodies from threeX40 microscope fields. In most tumors, the number of apoptotic bodies wasvery similar throughout the entire tumor section. There were —¿�1000cells per

X40 field, so an apoptotic index of 1000 indicates a virtually completeapoptotic response. p53 immunostaining was performed as described previously using anti-p53 CM-5 antibody (Novacastra Labs), visualized with 3,3'-

diaminobenzidine (Sigma) and NiCl. and counterstained with methyl green(13). Lymphoid immunostaining was performed by cutting 6-/am frozen tumorsections, staining with primary antibody to CD3 (Serotec) for T-cell identification or CD45R (PharMingen) for B-cell identification, and visualizing with

antirat FITC (Caltag). LOH of p53 in tumors was performed by Southern blotanalysis as described (26), except that the probe was labeled with digoxigenin.followed by anti-digoxigenin alkaline phosphatase (Boehringer Mannheim).

RESULTS

seid/seid Mice Are Prone to Radiation-induced T-Cell Lympho-

mas but not Other Tumor Types, seid/seid mice have only beenreported to show predisposition to T-cell lymphoma development (19.

23, 27, 28). This very narrow tissue specificity for a panorganismalDNA repair defect prompted us to examine radiation and carcinogen-

induced tumor susceptibility. We argued that treating seid/seid micewith broad-range carcinogens such as radiation (29) and ENU (30)

might reveal other tissues that were predisposed.Sixty-four C3CF, seid/seid/] mice (Balb/cByJSmn seid/

J X C3HSmn.C-.sr;W/J F,. referred to as seid/seid mice) were exposedto a single dose of 1 Gy of y radiation within 48 h of birth. Fifty-fiveof 64 (86%) of the mice developed thymic or disseminated lympho-

mas with a very short latency (Fig. 1). The tumor mass was most oftenwithin the thymus, with occasional spleen and lymph node involvement and metastasis to liver, kidneys, or lungs. Seven of seven tumorstested were of T-cell origin, as determined by anti-CD3 immunoflu-

orescence (data not shown). No other tumors types were observed. Fornine of the mice that became sick or died during the experiment, thecause of death could not be ascertained by necropsy. This exclusiveT-cell lymphoma susceptibility is similar to published reports for

seid/seid mice (19, 27, 28, 31).

100

20 30weeks of age

Fig. 1. ENU- and y radiation-induced tumors in scid/stid mice. scUl/.-icidor wild-typemice were exposed to 1 Gy of whole-body y radiation within 24-48 h after birth or

injected with ENU (0.5 fig/g body weight) at 12 days of age. acid/scid control mice wereuntreated. Mice were sacrificed when they showed signs of tumor development. The dataare plotted as Kaplan-Meier survival plots. Mice thai died of nontumor causes were not

included in the analysis.

The irradiated seid/seid mice may have been predisposed to tumor-

igenesis in other tissues, but this was not observed because theydeveloped lymphomas at an early age. This predisposition might havebeen revealed had they had lived longer. Seven of 10 (70%) untreatedC3CF, seid/seid mice, which were concurrently maintained in ourcolony, developed lymphoma. Full necropsy of those mice revealedonly one liver tumor and one lung tumor, although some of these micelived up to 20 months of age. Five of 23 untreated scid/+ controlmice, also observed for 20 months, developed lung tumors, and 1developed a liver tumor. Thus, seid/seid mice are only predisposed tospontaneous lymphomagenesis.

seid/seid Mice Are Prone to ENU-induced T-Cell Lymphomasbut not Other Tumor Types. ENU is a broad-spectrum carcinogen,

inducing tumors of the lung, liver, and elsewhere (30) and is thoughtto induce tumors by causing point mutations in target oncogenes (32).We wished to determine whether the seid defect would cooperate withENU mutagenesis to increase these or other tumor types or theirdegree of malignant progression. We treated 64 C3CF, scid/scid/i and51 wild-type control C3CF, (Balb/cByJ X C3H F,) mice with ENUat 12 days of age. Unexpectedly, 59 of 64 (92%) of the ENU-treated

seid/seid mice developed lymphomas with a very short latency (Fig.1). Significantly, the tumor induction kinetics were indistinguishablefrom the irradiated seid/seid mice. Upon gross and microscopic examination, the ENU-induced lymphomas were very similar in appear

ance to those from the irradiated mice, primarily occupying thethymus and occasionally involving the spleen and lymph nodes. Sevenof seven of the tumors were CD3^, indicating that, as above, the

tumors were of T-cell origin. Only 3 of 51 (6%) of the ENU-treatedwild-type controls developed lymphomas during the course of thestudy. In the longer surviving ENU-treated seid/seid and wild-type

animals, a number of lung and liver tumors were observed, seid/seidmice sacrificed between 20-30 weeks of age averaged 1 lung tumorper animal (52 tumors in 53 mice), whereas wild-type mice sacrificed

between 20 and 36 weeks of age averaged 1.4 lung tumors per animal(32 tumors in 23 mice). This indicated that the SCID defect did notenhance lung tumor development. Because the liver tumor multiplicity increased sharply with age and the mice were sacrificed at differentages, it was not possible to directly compare between groups.

Spontaneous B-Cell Lymphomagenesis in seid/seid p53—/—

Mice Is Accelerated. Appropriate crosses were set up to generateseid/seidp53— /—,seid/seidp53+/—, and seid/seid p53+/+ mice on

a mixed C3HSmn.C X C57BL6/J genetic background. Eleven of 11

3112

on April 8, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 3: DNA Double-Strand Breaks, p53, and Apoptosis …...[CANCER RESEARCH 58, 3111-3115. July 15, 1998| DNA Double-Strand Breaks, p53, and Apoptosis during Lymphomagenesis in seid/seid Mice1

LYMPHOMAGENESIS IN SCIO MICE

(100%) of the seid/seid p53—/—mice developed lymphomas with very

short latency (Table 1). The median age to tumor appearance was 8weeks, and all of the mice had succumbed by 15 weeks of age. The tumormass was located in the thymus, spleen, or lymph nodes with frequentmetastasis to liver, kidneys, or lungs. Tumor latency was longer in thesingle mutants: only 9 of 18 (50%) of the sdd/scid p53+/+ mice fromthe above cross developed lymphomas by 40 weeks of age. Single mutantp53—/- mice developed lymphomas and sarcomas, as well as a number

of other tumor types with a median age of >20 weeks (25, 33). Althoughthose p53—/- mice were not on the same genetic background as theseid/seid mice described here, the tumor induction kinetics of p53—I—

mice has not significantly varied between different laboratories and ondifferent genetic backgrounds (25, 33-36). Thus, tumor latency wasdecreased in the double-deficient seid/seid p53—/—mice relative tosingle-mutant seid/seid or p53—/—mice. These results are in close

agreement with results from two other laboratories (22, 23), attesting tothe robustness of the DNAPK-p53 interaction.

In contrast to the almost exclusive T-cell origin of lymphomas fromseid/seid or p53—/—mice (19, 22), five of six lymphomas examinedfrom seid/seid p53—/—mice were of B-cell origin, as determined byanti-CD45R immunofluorescence (data not shown). Guidos et al. (23)also observed primarily pre-B cell lymphomas in seid/seid p53—/—

mice.Spontaneous T-Cell Lymphomagenesis in seid/seid p53+/—

Mice Is Accelerated and There Is Selection for Loss of p53 Function. Seventeen of 51 (33%) of the seid/seid p53+/- mice sponta

neously developed lymphomas by 20 weeks of age versus only 2 of 18(11%) of the seid/seid p53+/+ mice (Table 1). In total, 40 of 51(78%) of the seid/seid p53+/— mice developed lymphomas by 40

weeks of age, in contrast to 9 of 18 (50%) of the seid/seid p53+/+mice. The median age for tumor development in p53+/— mice was

>50 weeks of age (33, 37). Thus, a 50% reduction inp53 gene dosage

also interacted with the seid defect to increase lymphomagenesis.Seven of seven of these lymphomas were CD3+, indicating T-cell

origin, similar to the seid/seid p53+/+ tumors.Tumor DNAs were analyzed by Southern blot to determine the fate

of the wild-type p53 alíele(26). Thirty of 30 (100%) of these tumorsshowed LOH of the wild-type alíele(Fig. 2). Most tumors showed

nearly complete LOH, whereas two showed partial LOH, indicatingan evolutionary intermediate stage.

Spontaneous or Induced T-Cell Lymphomagenesis in seid/seid

p53+/+ Mice Does Not Select for Loss of p53 Function. A prediction from the above results was that, during tumorigenesis inseid/seidp53+/+ mice, there would also be strong selective pressurein favor of mutant p53, leading to functional inactivation of p53 intumors. We examined p53 function in lymphomas from seid/seid miceof all three p53 genotypes by measuring y radiation-induced p53accumulation and apoptosis. Tumor-bearing mice were exposed to 4

Gy of y radiation and sacrificed 1, 2, or 4 h later. In the absence ofradiation, there was little or no detectable p53 immunostaining (Fig.4ß),and the apoptotic index was very low in 42 of 44 tumorsexamined from all three genotypes of mice (Figs. 3 and 4A). Two tumors

Table 1 Spontaneous lymphoma incidence in seid/seid p53-deßcient mice

Mice of all three p53 genotypes on a sdd/scid background were littermates generatedfrom a seid/seid p53 +/- cross on a mixed CSHSmn.C and C57BL/6J background. Mice

were examined daily and sacrificed when they showed signs of tumor development.Values are number of mice with lymphoma at the indicated age/total number of mice.

Age (weeks)

HTTTTTTTTTW

Genotypeseid/seid

p53—/—seid/seid p53+/-

scid/scid p53+/+109/11(82%)1/51 (2%)0/18(0%)2011/11

(100%)17/51 (33%)2/18(11%)4040/51

(78%)9/18(50%)

wt

Fig. 2. Loss of the wild-type ¡>53alíelein lymphomas from seid/seid p53+/- mice.Southern blot analysis reveals loss of the wild-type p53 alíelein all tumors examined. /)#.p53 pseudogene, which maps to a separate chromosome; wt. wild-type p53 alíele;mu,mutant or "knockout"p53 alíele;Wand W, normal tail DNA fromp53+/— and +/+ mice,

respectively; T, DNA from tumors from seid/seid p53 +/- mice. Note the tumor (fourthfrom left) with partial retention of the wild-type alíele.

a<:200

O

1000 i A: no treatment

| 800^c

•¿�ieooo

scidp53-/- scidp53+/- scidp53-r/+

1000

| 800^c£ 600 -

f 400-

C 200 -

0

B:4Gy4h

seidpii-/- 5CÃŒdp53+/-scidp53+/+

Fig. 3. Radiation-induced apoptosis in lymphomas from seid/seid /753-deficient mice.Tumor-bearing mice were untreated (A) or exposed to 4 Gy of y radiation (fi) and

sacrificed 4 h later, and apoptotic index was determined. Apoptotic index is the numberof apoptotic bodies per x40 microscope field. Dala points, individual tumors fromindividual mice. Apoptotic index was very low in all but two untreated tumors fromseid/seid mice, regardless of p53 genotype (A). Radiation increased apoptosis only inseid/seid p53+/+ tumors; little or no increase was seen in seid/seid p53-deficiem tumors

m

that were recorded as unirradiated showed a virtually complete apoptoticresponse, similar to those in the irradiated groups, which could be aphysiological reaction in these particular animals, due to, for example,tumor hypoxia. At 1, 2, or 4 h postradiation, lymphoma cells from all 23seid/seid p53+/+ mice examined showed nuclear p53 protein accumulation (Fig. 4D). By 4 h, 22 of 22 lymphomas from irradiated sdd/scidp53+/+ mice showed very high levels of apoptosis, as compared tolymphomas from unirradiated mice (Figs. 3 and 4C).

In sharp contrast, all 10 irradiated tumors examined from seid/seidp53—/—or seid/seidp53+/- mice showed very little or no induction

of apoptosis over background levels (Figs. 3 and 4/). p53 protein waslargely undetectable in irradiated seid/seid p53+/— tumors, which isconsistent with the results above showing loss of the wild-type p53

alíele.Two tumors showed a mixed population, with distinct, focalareas of high apoptosis and others of low apoptosis. In serial sectionsof these tumors, focal areas of p53 protein expression (Fig. 4H) or no

3113

on April 8, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 4: DNA Double-Strand Breaks, p53, and Apoptosis …...[CANCER RESEARCH 58, 3111-3115. July 15, 1998| DNA Double-Strand Breaks, p53, and Apoptosis during Lymphomagenesis in seid/seid Mice1

I.YMI'HOMAÅ’NKSIS IN SCIO MICE

B

Fig. 4. Radiation-induced p53 accumulation and apoplosis in lyniphomas from sciti/Miti niicL1.Histológica! sections of lyniphomas from an unirradiated .ïcitl/xcidmouse M)and 4 h after administration of 4 Gy of radiation to seid/seid (C} and seid/seid p5j—/—(I)mice are shown. A virtually complete apoptotic response is seen in the irradiated <¡titl/scitílymphoma (C). compared with the seid/seid p53—/—lymphoma (/). fi. p53 immuno-

staining of lymphomas from seid/seid mice without irradiation; D. 2 h after 4 Gy ofwhole-body radiation. Increased p53 nuclear accumulation is apparent in O. £-//. sametumor from a sciil/scid />5.f+/- mouse 4 h alter 4 Gy of whole-body radiation. E and F.

from the same central core region of the tumor, showing no p53 accumulation and lowlevels of apoptosis. C and H. from the outer edge, showing marked p53 accumulation (//)and apoptosis (G). J. lymphoma from a p53—/—mouse 48 h after 4 Gy of radiation,

showing little apoptosis and milotic recovery.

expression (Fig. 4F) were observed that spatially coincided with areasof high apoptosis (Fig. 4G) or low apoptosis (Fig. 4£).These tumorswere most likely caught in the process of clonal evolution in favor ofcells with complete loss of p53, which is supported by the partial LOHdetected by Southern blot analysis (Fig. 2). We conclude that theDNA damage-p53 induction-apoptosis pathway was inactivated inlymphomas from seid/seid p53+/— mice due to LOH of p53 but was

not inactivated in lymphomas from seid/seid p53+/+ mice, in whichit remained highly efficient.

DISCUSSION

DNA dsbs and Tumorigenesis. Indirect evidence indicates thatthe mutagenic and carcinogenic effects of y radiation are mediated byDNA dsbs (38). Because seid/seid mice have a panorganismal defectin DNA dsb repair, they provide a useful model to test the role of

radiation, dsbs, and cancer. Cumulative observations from two laboratories and this study4 indicate that, of >130 irradiated seid/seid

mice, none developed malignancies other than lymphomas, and all ofthose examined had a T-cell origin. Thus immature T lymphocytes areuniquely sensitive to spontaneous or radiation-induced tumorigenesis

due to DNAPK deficiency, whereas other cell types are not.To further address this very narrow tissue predisposition, we treated

seid/seid mice with END, an alkylating agent and broad-range carcino

gen. As with radiation, nearly 100% of these mice developed lymphomas,and again, other tumor types were not increased. Thus, the SCID dsbdefect does not cooperate with ENU-mediated tumorigenesis in most

tissues, with the marked exception of lymphomagenesis. The almostidentical tumor induction kinetics with radiation and ENU (Fig. 1) wasremarkable and implies that both treatments, despite their different mechanisms of mutagenesis. induced similar events with similar frequency.

The methylating agent MNU has a slightly different mutagenicspectrum than does ENU and also induces lymphomas at high frequency in seid/seid mice.5 Why are immature T lymphocytes, which

lack functional DNAPK. uniquely predisposed to transformation bythese diverse agents?

Lymphocyte precursors are the only somatic cell type to undergolarge-scale programmed genetic rearrangements, which are initiatedby the Rag-I and Rag-2 gene activities. These free DNA ends,

normally rejoined by DNAPK, would remain unjoined in seid/seidmice and, perhaps, increase illegitimate recombination with onco-

genic targets. In fact, chromosomal translocations linking antigenreceptor genes to proto-oncogenes are frequently observed in lymph-

oid malignancies (39).Additionally, the lymphocyte maturation arrest or other unknown

phenotypes of DNAPK deficiency may play a role. For example, inaddition to inducing dsbs, treatment of seid/seid mice with radiation orMNU partially rescued lymphocyte maturation, resulting in the rapidappearance of CD4+CD8+ thymocytes and increased thymic cellularity

(24, 28)/ Interestingly, this effect was specific to T cells because B cellmaturation was not affected. If ENU induced a similar response, thismaturation rescue might contribute to the unique T-cell predisposition inirradiated, ENU- and MNU-treated seid/seid mice.

p53 might also contribute to the lack of a generalized tumor predisposition in seid/seid mice. However, the only tumor type observed inseid/seid p53—/—mice was lymphoma, mostly pre-B cell lymphoma

(Refs. 22 and 23 and this study). Thus, generalized tumor suppression byp53 does not explain the narrow tumor spectrum of seid/seid mice.

Finally, we have shown that xcid/xcid mice are not predisposed to twostage chemical carcinogenesis of the skin or to chemically induced livertumors.'1 Taken together, these results indicate that unrepaired dsbs due to

DNAPK deficiency in seid/seid mice is not a major predisposing factorfor tumorigenesis in most tissues. SCID cells are able to repair a subsetof radiation-induced dsbs, perhaps due to homologous recombination or

other repair pathways (16). Perhaps these alternative repair pathways arecritical for dsb-mediated carcinogenesis.

Bifurcation of Molecular Pathways to Tumorigenesis Depending on p53 Germ-Line Status. We and others (22, 23) observed adecrease in tumor latency in seid/seid p53—/—mice. We also ob

served a very high frequency of LOH of p53 in tumors from seid/seidp53+/- mice and consequent loss of DNA damage-induced apopto

sis. The 100% frequency of LOH is higher than that seen in spontaneous lymphomas fmmp53+/— mice, in which the frequency is only

55-70% (33, 37). Thus, the DNAPK defect increases the frequency of

LOH of p53 in tumors, and this proves that there is very strong

4 C. Guidos, personal communication.*"S. Gerson. personal communication.

6 C. J. Kemp, unpublished results.

3114

on April 8, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 5: DNA Double-Strand Breaks, p53, and Apoptosis …...[CANCER RESEARCH 58, 3111-3115. July 15, 1998| DNA Double-Strand Breaks, p53, and Apoptosis during Lymphomagenesis in seid/seid Mice1

LYMPHOMAOBNESIS IN SCIO MICE

selective pressure against p53 function during lymphomagenesis on aseid/seid mutant background. This also implies that loss of the DNAdamage-p53-apoptosis pathway might be a critical rate-limiting step

for lymphomagenesis in these mice.However, in seid/seid p53+/+ mice, this pathway was not inacti

vated. Despite the fact that these tumors arose very rapidly and werehighly malignant, they remained exceedingly radiation sensitive, withmany showing a virtually complete apoptotic response. Apparently, inthese mice, tumors evolved down an entirely different molecularpathway, which did not involve p53 or other components of the DNAdamage-apoptosis response. Future studies will be directed towardidentifying components of this alternate rate-limiting pathway, which

may involve other mechanisms of apoptosis, for example, thoserelated to c-myc or bcl-2, or may not involve deregulated apoptosis.

The difference in frequency of inactivating p53 in tumors from seid/scidp53+/- versus seid/seid p53+/+ mice could be trivially explained

by the fact that one p53 alíelewas already lost in all somatic cells in theformer mice. Complete loss of p53 in tumors would occur at a frequencyrelated to the spontaneous rate of loss of the second alíeleand subsequentclonal selection. In the seid/seid p53+/+ mice, both alíeleswould haveto be inactivated within the same cell lineage, which would occur withmuch lower probability. In this model, loss of either alíelewould occur atequal probability. However, the very large difference in the frequency ofp53 loss suggests another model in which loss of the second p53 alíeleoccurs at higher probability than loss of the first alíele,due to p53haploinsufficiency. We previously showed by karyotype analysis of bonemarrow cells taken directly from untreated mice that only 2% of the cellsfrom wild-type mice were aneuploid, compared to 26% from p53+/-mice and 52% from p53—/—mice (11). These aberrations were not

clonal, in that each karyotype differed between cells examined, indicatingthat a 50% reduction in p53 gene dosage resulted in significantly increased karyotypic instability. This haploinsufficient phenotype providesa plausible mechanism whereby a mutation in one p53 alíelewouldincrease the probability of loss of the remaining wild-type alíeleduring

tumor cell evolution.

ACKNOWLEDGMENTS

We thank our colleagues at the Fred Hutchinson Cancer Center for valuablecritiques of the manuscript. We also thank Drs. C. Guidos, S. Gersten, and D.Willerford for sharing unpublished results.

REFERENCES

1. MacCallum. D. E.. Hupp. T. R.. Midgley, C. A.. Stuart. D.. Campbell. S. J.. Harper,A., Walsh. F. S., Wright, E. G., Balmain. A.. Lane. D. P.. and Hall, P. A. The p53response to ionising radiation in adult and developing murine tissues. Oncogene, 13:2575-2587, 1996.

2. Nelson. W. G., and Kastan. M. B. DNA strand breaks: the DNA template alterationsthat trigger p53-dependent DNA damage response pathways. Mol. Cell. Biol., 14:1815-1823, 1994.

3. Merritt, A. J., Potten, C. S.. Kemp. C. J., Hickman. J. A.. Lane, D. P.. and Hall. P. A.The role of spontaneous and radiation-induced apoptosis in the gastrointestinal tractof normal and p53-deficien! mice. Cancer Res.. 54: 614-617, 1994.

4. Midgley. C. A., Owens. B., Briscoe. C. V.. Thomas. D. B.. Lane. D. P.. and Hall, P. A.Coupling between y irradiation. p53 induction and the apoptotic response dependsupon cell type in vivo. J. Cell Sci.. 108: 1843-1848, 1995.

5. Lowe, S. W., Schmitt. E. M., Smith. S. W.. Osborne. B. A., and Jacks, T. p53 isrequired for radiation-induced apoptosis in mouse thymocytes. Nature (Lond.), .J62:847-849, 1993.

6. Clarke. A. R., Purdie, C. A., Harrison, D. J., Morris. R. G., Bird. C. C., Hooper, M. L.,and Wyllie, A. H. Thymocyte apoptosis induced by p53-dependent and independentpathways. Nature (Lond.). 362: 849-852, 1993.

7. Clarke, A. R., Gledhill, S., Hooper, M. L.. Bird. C. C., and Wyllie, A. H. p53dependence of early apoptotic and proliferative responses within the mouse intestinalepithelium following y-irradiation. Oncogene, 9: 1767-1773, 1994.

8. Yin. Y.. Tainsky. M. A.. Bischoff, F. Z., Strong, L. C.. and Wahl, G. M. Wild-typep53 restores cell cycle control and inhibits gene amplification in cells with mutant p53alíeles.Cell, 70: 937-948. 1992.

9. Livingstone, L. R., White, A., Sprouse, J.. Livanos, E., Jacks, T., and Tlsty, T. D.Altered cell cycle arrest and gene amplification potential accompany loss of wild-typep53. Cell, 70: 923-935, 1992.

10.

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

Harvey. M.. Sands. A. T., Weiss, R. S.. Hegi, M. E.. Wiseman, R. W., Pantazis. P..Tainsky, M. A.. Bradley, A., and Donehower, L. A. In vitro growth characteristics ofembryo fibroblasts isolated from p53-dcficient mice. Oncogene. H: 2457-2467. 1993.Bouffler, S. D.. Kemp, C. J.. Balmain. A., and Cox, R. Spontaneous and ioni/.ingradiation-induced chromosomal abnormalities in p53-deficient mice. Cancer Res., 55:3883-3889. 1995.Cross, S. M., Sanche/.. C. A.. Morgan, C. A.. Schimke. M. K., Ramel, S.. Idzerda.R. L., and Reid, B. J. A p53-dependent mouse spindle checkpoint. Science (Washington DC). 267: 1353-1356. 1995.Gurley. K. E.. and Kemp. C. J. p53 induction, cell cycle checkpoints, and apoptosisin DNAPK deficient seid mice. Carcinogenesis (Lond.). 17: 2537-2542, 1996.Jackson, S P.. and Jeggo. P. A. DNA double-strand break repair and V(D)J recombination: involvement of DNA-PK. Trends Biochem. Sci.. 20: 412-417, 1995.Araki, R.. Fujimori, A.. Hamatani. K.. Mila, K.. Saito. T.. Mori. M.. Fukurnura. R..Morimyo. M.. Muto. M., Itoh, M.. Tatuimi, K.. and Abe. M. Nonsense mutation atTyr-4046 in the DNA-dependent protein kinase catalytic subunit of severe combinedimmune deficiency mice. Proc. Nati. Acad. Sci. USA, 94: 2438-2443, 1997.Lieber, M. R.. Grawunder. U.. Wu. X., and Yaneva. M. Tying loose ends: roles of Kuand DNA-dependent protein kinase in the repair of double-strand breaks. Curr. Opin.Genet. Dev.. 7: 99-104. 1997.Budach, W.. Hartford. A.. Gioioso, D., Freeman. J.. Taghian. A., and Suit. H. D.Tumors arising in SCID mice share enhanced radiation sensitivity of SCID normaltissues. Cancer Res.. 52: 6292-6296. 1992.Van Buul. P. P. W., De Rooij. D. G.. Zandman. I. M.. Grigorova. M.. and VanDuyn-Goedhart, A. X-ray-induced chromosomal aberrations and cell killing in somatic and germ cells of the seid mouse. Int. J. Radial. Biol., 67: 549-555. 1995.Custer. R. P., Bosma. G. C.. and Bosma. M. J. Severe combined immunodeficiency(SCID) in the mouse. Pathology, reconstitution, neoplasms. Am. J. Pathol., 120:464-477. 1985.Biedermann. K. A., Sun. J. R., Giaccia. A. J.. Tosto, L. M., and Brown. J. M. sadmutation in mice confers hypersensitivity to ionizing radiation and a deficiency inDNA double-strand break repair. Proc. Nati. Acad. Sci. USA. SS: 1394-1397. 1991.Huang, L.. Clarkin. K. C.. and Wahl. G. M. p53-dependent cell cycle arrests arepreserved in DNA-activated protein kinase-deficient mouse fibrohlasls. Cancer Res..56: 2940-2944. 1996.Nacht. M.. Strasser. A.. Chan. Y. R.. Harris. A. W.. Schlisse!. M.. Bronson. R. T., andJacks, T. Mutations in the />5.fand .vciVigenes cooperate in tumorigenesis. Genes De\..10: 2055-2066. 1996.Guidos, C. J.. Williams, C. J.. Grandal. I.. Knowles. G.. Huang, M. T. F., and Danska.J. S. V(D)J recombination activates a p53-dependent DNA damage checkpoint in seidlymphocyte precursors. Genes Dev.. 10: 2038-2054, 1996.Bogue. M. A., Zhu. C., Aguilar-Cordova, E.. Donehower. L. A., and Roth. D. B. p53is required for both radiation-induced differentiation and rescue of V(D)J rearrangements in seid mouse thymocytes. Genes Dev.. 10: 553-565. 1996.Donehower, L. A.. Harvey. M.. Slagle. B. L.. McArthur. M. J.. Montgomery. C. A.,Jr.. Bute!. J. S.. and Bradley. A. Mice deficient for p53 are developmentally normalbut susceptible to spontaneous tumours. Nature (Lond.). 356: 215-221. 1992.Kemp, C. J.. Donehower, L. A., Bradley, A., and Balmain. A. Reduction of ¡>53genedosage does not increase initiation or promotion but enhances malignant progressionof chemically induced skin tumors. Cell, 74: 813-822. 1993.Lieberman. M.. Hansteen. G. A.. Waller, E. K.. Weissman, I. L.. and Sen-Majumdar.A. Unexpected effects of the severe combined immunodeficiency mutation on murinelymphomagenesis. J. Exp. Med.. /76: 399-405, 1992.Danska. J. S.. Pflumio. F., Williams. C. J.. Huner. O.. Dick, J. E.. and Guidos. C. J.Rescue of T cell-specific V(D)J recombination in SCID mice by DNA-damagingagents. Science (Washington DC), 266: 450-455. 1994.Vesselinovitch. S. D., Simmons. E. L.. Mihailovich. N.. Rao, K. V. N.. and Lombard.L. S. The effect of age, fractionation. and dose on radiation Carcinogenesis in varioustissues of mice. Cancer Res.. 31: 2133-2142, 1971.Vesselinovitch. S. D.. Rao. K. V. N.. Mihailovich. N., Rice. J. M.. and Lombard. L. S.Development of broad spectrum of tumors by ethylnitrosourea in mice and themodifying role of age, sex. and strain. Cancer Res., 34: 2530-2538. 1974.Murphy, W. J.. Durum. S. K., Anver, M. R., Ferris. D. K.. McVicar. D. W.. O'Shea,

J. J.. Ruscelli. S. K...Smith. M. R.. Young, H. A., and Longo. D. L. Induction of T-celldifferentiation and lymphomagenesis in the thymus of mice with severe combinedimmune deficiency (SCID). J. Immunol.. 153: 1004-1014, 1994.Skopek, T. R., Walker. V. E.. Cochrane, J. E.. Craft. T. R.. and Cariello, N. F.Mutational spectrum at the Hprt locus in splenic T cells of B6C3FI mice exposed to/V-ethyl-A'-nitrosourea. Proc. Nati. Acad. Sci. USA, 89: 7866-7870, 1992.Kemp. C. J., Wheldon, T., and Balmain. A. p53 deficient mice are extremelysusceptible to radiation-induced tumorigenesis. Nat. Genet.. 8: 66-69. 1994.Harvey, M.. Vogel, H., Morris, D., Bradley, A.. Bernstein. A., and Donehower. L. A.A mutant p53 transgene accelerates tumour development in heterozygous but notnullizygous p53-deficient mice. Nat. Genet.. 9: 305-311. 1995.Jacks. T.. Remington. L., Williams. B. O., Schmitt, E. M.. Halachmi. S., Bronson,R. T., and Weinberg. R. A. Tumor spectrum analysis in p53-mutant mice. Curr. Biol..4: 1-7. 1994.Purdie, C. A., Harrison, D. J., Peter, A.. Dobbie, L., White, S., Howie, S. E. M.,Salter. D. M.. Bird, C. C.. Wyllie. A. H.. Hooper, M. L.. and Clarke. A. R. Tumourincidence, spectrum and ploidy in mice with a large deletion in the p53 gene.Oncogene. 9: 603-609. 1994.Harvey. M.. McArthur. M. J., Montgomery. C. A.. Jr.. Butel. J. S., Bradley. A., andDonehower. L. A. Spontaneous and carcinogen-induced tumorigenesis in p53-defi-cient mice. Nat. Genet.. 5: 225-229. 1993.Ward. J. F. Radiation mutagenesis: the initial DNA lesion responsible. Radiât.Res..142: 362-368, 1995.Rabbitts. T. H. Chromosomal translocations in human cancer. Nature (Lond.). 372:143-149, 1994.

3115

on April 8, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 6: DNA Double-Strand Breaks, p53, and Apoptosis …...[CANCER RESEARCH 58, 3111-3115. July 15, 1998| DNA Double-Strand Breaks, p53, and Apoptosis during Lymphomagenesis in seid/seid Mice1

1998;58:3111-3115. Cancer Res   Kay E. Gurley, Khoa Vo and Christopher J. Kemp 

Micescid/scidLymphomagenesis in DNA Double-Strand Breaks, p53, and Apoptosis during

  Updated version

  http://cancerres.aacrjournals.org/content/58/14/3111

Access the most recent version of this article at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/58/14/3111To request permission to re-use all or part of this article, use this link

on April 8, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from


Recommended