Survey
* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project
[CANCER RESEARCH58. 95-101, January 1, 19981 Inhibition of DNA Methylation by 5-Aza-2'-deoxycytidine Suppresses the Growth of Human Tumor Cell Lines' Christina M. Bender, Martha M. Pao, and Peter A. Jones2 Urological Cancer Research Laboratory, USC/Norris Comprehensive Cancer Center, Unis'ersity of Southern California School of Medicine, ii)s Angeles, California 90033 ABSTRACT have been shown to coexist during oncogenic transformation (9, 10), and de novo methylation Alterations in DNAmethylationpatterns accompanythe establishment of immortal cell lines. De novo methylation of CpG islands within the control regions ofgrowth-regulatory genes may Inactivate their transcrip tion, giving cells selective growth advantages in culture. We exposed seven human tumor cell lines and two human fibroblast cell strains to the demethylating agent, 5-aza-2'-deoxycytidine (5-Aza-CdR), to determine whether the silencing of growth-regulatory genes by de novo methylation in immortalized cell lines could be reversed, possibly restoring growth control. After recovery from the Immediate cytotoxic effects of 5-Aza CdR, this agent suppressed cellular growth in all seven tumor lines but not in either fibroblast strain. Because alterations In the p16 (CDKN2/MTSJ) cell cycle regulatory gene are associated with numerous cancers, we analyzed expression of this gene before and after S-Aza-CdR treatment. The gene was reactivated by 5-Aza-CdR treatment in three of four tumor cell lines not expressing p16, whereas the fourth tumor line contained a p16 homozygous deletion. pitS was shown to be hypermethylated only in the cell lines and its up-regulation by 5.Aza-CdR was associated with demethylation of thepl6 promoter. The remaining tumor lines expressed p16 at constant levels before and after S-Aza-CdR treatment and showed minimal pi6 promoter methylation, suggesting that other growth-regula tory genes may have been silenced by de novo methylation in these cells. p16 expression, cell growth inhibition, and G1 cell cycle arrest by S-Aza CdR in the T24 bladder tumor cell line were also heritable after prolonged passage In culture. Furthermore, a dormant pitS gene was reactivated in T24 cells growing in nWnu rats, and 5-Aza-CdR treatment of T24 cells before inoculation into nWnu mice decreased the rate of tumor growth. These results suggest that S-Aza-CdR may slow the growth of tumor cells by reactivating growth-regulatory genes silenced by de nova methylation. INTRODUCTION Tumorigenesis and increased cellular proliferation arise via the accumulation of specific genetic and epigenetic changes that provide tumor cells with selective growth advantages over neighboring normal cells. Identifying molecular alterations (including mutation, deletion, and abnormal methylation) associated with a particular cancer will facilitate the early detection of tumors and the development of ther apies that target these specific alterations. Aberrant DNA methylation patterns accompany the neoplastic process in vivo (1, 2), and de novo methylation of CpG islands is observed during the establishment of immortal cell lines in vitro (3, 4). DNA methylation normally occurs at cytosine residues within CpG dinucleotides which are represented at a lower than expected fre quency throughout the eukaryotic genome (5, 6). In contrast, regions of the genome defined as CpG islands contain the predicted frequency of CpG dinucleotides that are usually unmethylated, except for those on the inactive X chromosome and parentally imprinted genes (7, 8). Global hypomethylation and regional CpG island hypermethylation @ of DNA has been demonstrated in cancer derived cell lines ( 11) and uncultured tumors ( 12, 13). This epigenetic modification may increase cell proliferation via hypermethylation of growth-regulatory genes, especially because de novo methylation within the regulatory sequences of tumor suppressor genes has been proposed to silence their transcription in specific cancers. For exam ple, hypermethylation of the RBJ gene has been observed in retino blastomas (14, 15), and transcriptional silencing has been associated with hypermethylation of the VHL gene in sporadic renal cell card nomas (16), the estrogen receptor gene in breast cancers ( 17), the p15 (INK4B/MTS2) gene in leukemias (18), the p16 gene in human tumor cell lines (19, 20), and the H19 gene in Wilms' tumor (21). Genes silenced by hypermethylation can be reactivated by 5-Aza-CR3 or 5-Aza-CdR (22), which inhibit DNA methylation and are also effec tive antileukemic agents (23, 24). In this report, we analyzed p16 activation because this gene is altered in numerous cancers. The p16 gene on chromosome 9p2 1 encodes a Mr 16,000 cell cycle regulatory protein that belongs to the cyclin-dependent kinase inhibitory protein family and regulates the G1/S-phase cell cycle transition, preventing phosphorylation of the retinoblastoma gene product (25). The p16 gene is especially inter esting because it is frequently lost via homozygous deletion or loss of heterozygosity in many human malignancies (26, 27), and its pro moter and first two exons have the characteristics of CpG islands. Evidence for pitS hypermethylation has also been documented in several human cancers including: head and neck squamous cell car cinoma (28), non-small cell lung cancer (28), bladder transitional cell carcinoma (20), gliomas (29), and nasopharyngeal carcinoma (30). p16 reactivation by 5-Aza-CdR has also been demonstrated in vitro (19, 28, 31). Thus, methylation ofthe 5' regulatory region ofp/6 may represent an alternative means for its inactivation besides intragenic mutation or homozygous deletion. We investigated cellular growth suppression in various human tumor cell lines following treatment with 5-Aza-CdR after complete recovery from the immediate cytotoxic effects of this agent. 5-Aza CdR suppressed cellular growth in all seven tumor cell lines analyzed but not in human fibroblasts treated under the same conditions. p16 expression levels were measured before and after treatment to deter mine whether up-regulation of this gene was associated with cell growth inhibition in the tumor cell lines. 5-Aza-CdR induced pitS expression in three tumor cell lines not expressing this gene, but the three remaining tumor cell lines expressed p16 at steady levels before and after 5-Aza-CdR treatment, implicating that growth-regulatory genes outside the pitS pathway might have been silenced by de novo methylation in these cells. The pitS promoter was also shown to be hypermethylated only in the three cell lines not expressing the gene, and significant demethylation of this region was associated with gene activation by 5-Aza-CdR. In the T24 bladder carcinoma-derived cell line, pitS expression, cellular growth suppression, and 0 arrest were heritable for up to 10 passages following 5-Aza-CdR treatment. A Received 9/16/97; accepted 10/29/97. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18U.S.C.Section1734solelyto indicatethis fact. I Supported by USPHS 2 To requests whom Grant for reprints R37 CA49758 should be from the National addressed, Urological Cancer Institute. Cancer 3 The Research abbreviations used are: 5-Aza-CR, 5-aza-deoxycytidine; 5-Aza-CdR, 5-Aza-2'- deoxycytidine; Ara-C, l-@-D-arabinofuranosylcytosine; RT-PCR, reverse transcription Laboratory, USC/Norris Comprehensive Cancer Center, University of Southern California School of Medicine, 1441 Eastlake Avenue, Los Angeles, California 90033. Phone: (213) 764-0816; Fax: (213) 764-0102. PCR; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; Ms-SNuPE. methylation sensitive single nucleotide extension assay. 95 Downloaded from cancerres.aacrjournals.org on June 16, 2017. © 1998 American Association for Cancer Research. INHIBITION OF DNA METhYLATION RESTORES GROWTH CONTROL Determination dormant p16 gene was also reactivated in T24 cells growing in nu/nu rats, and T24 cells treated with 5-Aza-CdR before introduction into nu/nu mice formed tumors that were significantly smaller of Cell Doubling Time Cells (10―/60-mm dish) were plated approximately 9 days after 5-Aza-CdR treatment, and the cell number/dish was counted with a Coulter counter each than those induced by untreated T24 cells. These results suggest cancer therapies in which growth-regulatory genes (like p16) silenced by de novo methylation are reactivated by 5-Aza-CdR, effectively suppressing the growth of human tumor cells. day for 5 consecutive days. Untreated cells were analyzed under similar conditions as a control. The average cell number from two plates was deter mined, and the mean cell numbers were plotted to define the cell population doubling times. RT-PCR MATERIALS AND METHODS Total RNA was isolated from 2 x 106 cells lysed in 2 ml of buffer Cell Lines containing guanidine isothiocyanate T24, J82, and 5637 (bladder transitional carcinoma-derived cell lines) were tific, Fair Lawn, NJ) and 2-mercaptoethanol obtained from the American Type Culture Collection (Rockville, MD). HCT15 and HCT1 16 (colon carcinoma-derived @ cell lines) were also obtained Inc., Palo Alto, (0.1 M; Sigma). RNA was pre cipitated (I h at —20°C) in 50% isopropanol/50% lysis buffer following from the standard phenol-chloroform extraction of the cell lysate. After centrifugation (10 mm at 10,000 x g), the supernatant was decanted, and the RNA pellet was American Type Culture Collection. SK-Mel-173 and SK-Mel-28 (melanoma cell lines) were obtained from Memorial Sloan Kettering (New York, NY). LD98 (human fibroblast cell strain) was established in our laboratory, and 1-1 (human fibroblast cell strain) was obtained from Children's Hospital (Los Angeles, CA). T24, SK-Mel-l73, SK-Mel-28, and LD98 were cultured in DMEM supplemented with 10% FCS and 5% penicillin/streptomycin. J82, 5637, HCT15, HCT1 16, and T-l were cultured in MEM supplemented (4 M; Life Technologies, CA), N-laurylsarcosine(0.5%;Sigma), sodiumcitrate(25 mM;FisherScien washed twice in 70% ethanol prepared with diethylpyrocarbonate-treated double-distilled water. The RNA pellet was dissolved in 100% diethylpyro carbonate water. Two of total RNA was reverse-transcribed using random hexamers, deoxynucleotide triphosphates (Boehringer Mannheim, Germany), and Superscript II reverse transcnptase (Life Technologies, Inc., Palo Alto, CA) in a 25-@tlreactionas described(20). cDNA was amplifiedwith primers with 10% FCS, 5% penicillin/streptomycin, nonessential amino acids, and sodium specific for either p16 or GAPDH. pitS and GAPDH PCR primer sequences, pyruvate. probe sequences, and PCR conditions were used as described (20). PCRs were performed with cDNA template concentration equivalent to 100 ng of RNA in 5-Aza-CdR and Am-C Treatments 10% DMSO. All reactions were analyzed in the linear range of amplification. PCR products were resolved on 2% agarose gels and subsequently transferred Cell Lines and Cell Strains. Cells were plated (3 X l0@cells/lOO-mm to a nylon membrane (Zetaprobe; Bio-Rad, Richmond, CA) under alkaline dish) and treated 24 h later with 5 X l0@ M, l0@ M, 5 X l0@ M, or lO_6 M conditions. All blots were hybridized with digoxigenin-labeled internal oligo 5-Aza-CdR or l0@ M Ara-C (Sigma Chemical Co., St. Louis, MO). The nucleotide probes (Genius Hybridization Method; Boehringer Mannheim). medium was changed 24 h after drug treatment and every subsequent 3 days. RNA and DNA were isolated 9 days after treatment as described (20). Determination Nude Rats. T24 cells (3 X 106)were injectedvia laparotomydirectlyinto the bladder walls of4-week-old nw'nurats with a 27-gauge needle. 5-Aza-CdR treatments of Cell Cycle Profile Cells (2 x 105/lOO-mm dish) were plated and treated with i0@ M or 5 X l0@ M 5-Aza-CdR or l0@ M Ara-C. Cells were fixed after 7 days with were initiated 4—6 weeks later, allowing time for macroscopic bladder tumors to form. Each animal received one daily i.p. injection of 150, 75% ethanol, treated with RNasin (20 300, 600, or 900 iodide (50 p.g/ml; Sigma). DNA content at each cell cycle stage was deter @gof 5-Aza-CdR for 5 days. Animals were sacrificed 24 h after the last treatment, and total bladder RNA was isolated to analyze pitS expression. Nude Mice. T24 cells (2 X 106) were injecteds.c. into the rightand left flanks of 4-week-old @g/ml),and stained with propidium mined via flow cytometry. Determination of Cytotoxicity nu/nu mice 11 days after treatment with 5-Aza-CdR (5 X l0@ M) in vitro. The cells were passed once in culture to facilitate recovery from the cytotoxic effects of the drug before harvesting for injection. T24 cells Untreated T24 cells were injected s.c. into individual mice under the same conditions as a control. Tumor sizes were measured 4 weeks after injection using a caliper, and total tumor RNA was extracted (200/60-mm dish) were plated in triplicate sets for a colony formation assay. Cells were treated with l0@ M, 5 X l0@ M, or lO_6 M 5-Aza-CdR or l0@ MAra-C 24 h later. Once cell colonies were visible (after 7 days), cells were fixed in 100% methanol and stained with 10% Giemsa stain. The percentage cell survival was assessed by dividing the mean colony number on the treated plates divided by the mean colony number on the to analyze p16 expression via RT-PCR. untreated plates X 100. Quantitation ofpl6 Promoter Methylation RESULTS p16 promoter methylation was measured using a Ms-SNuPE assay as described (32). All cell lines and cell strains were treated with 5 X i0@ M 5-Aza-CdR, and DNA was harvested 9 days after treatment (by standard proteinase K digestion, phenol/chloroform extraction, and ethanol precipitation Effects procedures). Genomic DNA was treated with sodium bisulfite as described by Frommer's group (33, 34) to convert unmethylated cytosines to uracil, leaving 5-methylcytosine unchanged, and a region of the pitS 5' promoter was ampli fled using PCR primers specific for bisulfite-converted DNA as described by Gonzalgo and Jones (32). Single nucleotide primer extension was performed using internal terminating primers immediately specific for the region amplified, with each primer 5' of the CpG site to be assayed. The methylation status of three CpG sites were analyzed, residing in a region approximately 200-bp upstream from exon 1 in the p16 promoter. The internal primer sequences, specific CpG locations, Ms-SNuPE extension conditions, and meth ods for detecting 32P-labeleddCTP or dTTP incorporation (to quantitate the percentage methylation per CpG) are described by Gonzalgo and Jones of 5-Aza-CdR on Cell Growth Inhibition, p16 Expres sion, andpl6 Promoter Methylation in Human Tumor Cell Lines. During the establishment of non tumorigenic or tumorigenic cell lines, hypermethylation within the 5' CpG islands of growth-regulatory genes may cause cells to proliferate more rapidly, giving these cells a selective advantage in culture (3, 4). Various tumor cell lines were, therefore, treated with 5-Aza-CdR to determine whether this inhibitor of DNA methylation could restore growth control to tumor cell lines or alter the growth characteristics of diploid fibroblasts (Fig. 1). After recovery from immediate drug cytotoxicity, significant growth mlii bition was observed with all seven tumor lines treated with 5 X iO@ M 5-Aza-CdR but not with human fibroblasts. Thus tumor cells, but not normal cells, were responsive to the growth-suppressive effects of this DNA methylation inhibitor. The results obtained for the expression of pitS before and after (32). The methylation percentage values determined for the three CpG sites were averaged and recorded as the percentage of pi6 promoter methylation. 96 Downloaded from cancerres.aacrjournals.org on June 16, 2017. © 1998 American Association for Cancer Research. 1N@B@ON OF DNA METHYLATION RESTORES GROWTH CONTROL Fig. 1. Effects of 5-Aza-CdR on growth suppres sion, p16 expression, and p16 methylation in tumor Human Time(Hrs) Expression Growth Tumor 5AzaCdR Suppressionp16 5AzaCdR Cell LineDoubling (+)i (-) (+)Fold (-) (+)% cell lines.Afterrecoveryfromimmediatedrugcy T24 totoxicity (approximately 9—11days), the effects of 5 x i0@ M 5-Aza-CdR on cell growth inhibition wereanalyzedin bladdercancer-derivedcell lines (F24, J82, and 5637), colon cancer-derived cell lines (HCTI5 and HCT1 16), melanoma cell lines (SK Mel-173 and SK-Mel-28), and flbroblast cell strains (LD98 and T-1) by comparing the doubling times of these cells before and after dnig treatment (as de scribed in “Materialsand Methods―).p16 expression @ was analyzed in these cells by RT-PCR analysis following RNA isolation approximately 9 days after drug treatment. p16 methylation levels were also measured in the cells before and after treatment with 5-Aza-CdR. DNA was isolated 9 days after drug treatment, treated with sodium bisulfite, and a region of the p16 promoter (approximately 200-bp up p16 Promoter Methylation 5AzaCdR (-) 56 35. J82 41 65 1.6 5 563721 29 501.7 1.7@J93 8 I: 5 .@ .@ 7.@ 0 I-. HCTI5 48SK@MEL173 g .3 HCT1I6 53 48 701.5 1.5IsSI90 C.)35 stream from exon 1) was amplified with PCR prim ers specific for bisulfite-converted DNA. Methyla tionat eachofthreespecificCpUsin thisregionwas quantitated using Ms-SNuPE as described (32), and the values were averaged and recorded as the per centage of p16 promoter methylation. 5-Aza-CdR treatment suppressed growth in all seven cell lines, @ @ up-regulatedpi6expressionintheT24,HCT15,and HCT116cell lines,anddecreasedp16 promoterhy permethylation in these same three cellilnes. Growth suppressionwas not detectedin the LD98andT-l fibroblasts 71 C I 6@@1 SK-MEL-2844 49 .11 2.2I Strain I LD98 @i 1101!6 1 Io.I8 T-1 @j__32 27 @-- 5-Azu-C4Rtreatmentare also shownin Fig 1.This gene1whichwas 32 27 -@:: I..1 —@3 5 7 and RT-PCR was performed to determine whether p16 was induced by drug treatment, The T24 bladder carcinoma-derived cell line con tains a transcriptionally silent, hypermethylated p16 gene with a wild-type sequence (data not shown),4 Fig 2 shows that l0@@M 5-Aza-CdR induced p16 expression in a dose-dependent manner, The 124 population doubling time increased after treatment with iO@@M 5-Aza-CdR with a dose-dependent effect at higher concentrations (5 X l0@ M and lO_6 M), directly correlating with p16 induction, Cells were also treated with the chemotherapeutic agent Ara-C, which incorporates into replicating DNA but does not inhibit DNA methy lation. Am-C (l0@ M) did not induce growth suppression in T24 cells at a dose equitoxic to 5 X l0@ M S-Aza-CdR (data not shown), showing that p)6 reactivation did not result from the general cyto toxicity of 5-Aza-CdR. selected for study because it is known to frequently undergo de novo methylation during tumorigenesis. was up-regulated in T24, HCT1S, and HCT1 16 cells but not in J82, 5637, and SK-Mel-28 cells or normal fibroblasts. SK.Mel-173 cells contain a p16 homozygous deletion, Because 5-Aza-C4R is known to be highly effective at inducing the expression of genes inappropriately silenced by de novo methylation (35), these results suggested that the growth-suppressive effects of 5-Aza-CdR in cancer cells were due to the up-regulation of p16 in the T24, HCT15, and HCT1 16 cells and possibly other growth regulators in the four other cancer cell types@In contrast, tibroblasts that also showed Initial sensitivity to drug toxicity (data not shown) did not respond to treatment by showing a heritable alteration in cell doubling time, possibly because abnormal methylation patterns may not exist in these normal somatic cells, Fig 1 also shows the quantitative measurements of pitS promoter methylation immediately downstream from a region previously shown Cell Cycle Arrest after Treatment with 3-Aza.CdR, T24, 5637, and LD98 cells treated with 5-Aza-CdR (5 X l0@ M) were analyzed by flow cytometry to identify any changes in their cell cycle profiles Cells were maintained in drug-free medium and harvested two pas sages after treatment to ensure complete recovery from the immediate toxic effects of 5-Aza-CdR, Interestingly. exposure of T24 cells to 5-Aza-CdR resulted in a 2-fold increase in the proportion of cells in G1 (from 34 to 64%; Fig. 3)@In contrast, 5637 cells, which express p16, showed a more subtle increase in the proportion of cells in G1, from 34 to 44%, after treatment with 5-Aza-CdR. LD98 fibroblasts, which also express p16, showed a negligible change in cell cycle profile (Fig 3), Treatment of T24 cells with an equitoxic dose of Ara-C (l0@ M)did not increase the proportion of cells in G1 (data not to contain putative transcription initiation sites (36)@Hypermethyla tion of p)6 was demonstrated in the T24, HCT1@,and HCT1 16 cell lines, and demethylation by 5-Aza-CdR was associated with the up-regulation of p16 exclusively in these three lines, The 5637, J82, and SK-Mel-28 cell lines, which express p16, showed low levels of p16 methylation as cxpected@ These results suggest that the growth suppressive effects of 5-Aza-CdR observed in these cell lines may arise from the demethylation of other growth-regulatory genes besides p16, which are presumably not hypermethylated in the normal fibro blasts, Effects of 5.Aza.CdR on p16 ExpressIon. Previous studies have demonstrated a strong correlation between hypermethylation of the p16 5' CpG island and transcriptional silencing of this gene in 4 M. L Gonzalgo,T. Haysahida,M. M. Pan,C. M. Bender,Y. C. Tsui,F. A. Gonzales, uncultured tumors and cancer-derived cell lines (19, 20, 28, 29, 31). H.0. Nguyen,T.T. Nguyen,andP. A.Jones.Theroleof DNAmethylationinexpression of the pJQ/pIólocus in human bladder cancer cell lines, submitted for publication. T24 cells were treated with increasing concentrations of 5-Aza-CdR. 97 Downloaded from cancerres.aacrjournals.org on June 16, 2017. © 1998 American Association for Cancer Research. INHIBITIONOF DNA METHYLATIONRESTORESGROWTHCONTROL after 5-Aza-CdR treatment to determine whether growth suppression and pitS expression following drug treatment were heritable in vitro. Growth inhibition after treatment with l0@ M 5-Aza-CdR was heri table for nine passages, but cells exposed to 5 X l0@ M 5-Aza-CdR showed growth suppression even after the tenth passage (Fig. 4a). In addition, RT-PCR-based expression analyses showed significant pitS induction by both l0@ M and 5 X l0@ M 5-Aza-CdR (Fig. 4b). Expression levels gradually diminished during each subsequent pas sage up to the tenth passage, possibly arising from de novo methyl ation of p16 or the selection of cells unaffected by the drug. These observations demonstrate that p16 induction and growth suppression by 5-Aza-CdR treatment are heritable and dose dependent, supporting findings by Costello et a!. (29) in which 5-Aza-CdR-mediated pitS expression was heritable in vitro. The induction of pitS by inhibition of DNA methylation may explain why T24 cells treated with 5-Aza CdR proliferated more slowly; however, reactivation of other growth regulatory genes silenced by de novo methylation may have also contributed to this change. 5AzaCdR-Mediatedp16 InductionIn Vitro @3 4 @z 5AzaCdR(M) oUTT@ C.) p16mRNA GAPDHmRNA —0@ —0 Doubling time (h) —0 21 21 26 35 72 Fig. 2. Effects of 5-Aza-CdR on p16 expression and cell doubling times in T24 cells. RNA was isolated from T24 cells 9 days after 5-Aza-CdR (5 X l0@ M)treatment (as described in @Materialsand Methods―).Untreated T24 cells were analyzed under similar conditions as a control. p16 expression was measured via RT-PCR. The cell doubling times were also analyzed 9 days after drug treatment to allow recovery from the cytotoxic effects of the agent. p16 expression and growth suppression were observed after treatment Reactivation with at least l0@ M5-Aza-CdR. GAPDH expression was measured to control for cDNA input and integrity. shown); therefore, the general cytotoxic effects of 5-Aza-CdR did not influence the change in cell cycle profile observed in these cells. Heritability of 5-Aza.CdR-mediated Growth Suppression of p16 by 5-Aza-CdR in Vivo. Bladder tumors and pl6 Expression in Vitro. Cell population doubling times and pitS mRNA levels were measured in T24 cells during successive passages CellType5AzaCdR3%3%Cell 5AzaCdR+ T24@34% Line 63%@@°“5637 Fig. 3. Effects of 5-Aza-CdR on cell cycle pro files in T24, 5637, and LD98 cells. Cells were exposed to 5-Aza-CdR (5 X l0@ M) for 24 h, followed by drug removal. Cells were maintained @34% for 7 days and passaged once before harvesting. On the tenth day after treatment, cells were fixed in 75% ethanol, stained with propidium iodide, and analyzed by flow cytometry to determine relative DNA content at specific cell cycle stages. A pro nounced increase ofcells in G1 was observed in T24 cells treated with 5-Aza-CdR, but a more subtle Cell Line19% 47%LD982% effect was observed in 5637 cells. A negligible effect was observed in LD9S cells. 27%Cell in duced in nu/nu rats, via laparotomy and direct implantation of T24 cells into bladder walls, were analyzed to determine whether 5-Aza CdR could activate pi6 expression in vivo. Approximately 4—6weeks later, when macroscopic bladder tumors were evident, 5-Aza-CdR was administered i.p. every 24 h for 5 days. Animals were sacrificed on the sixth day for the harvesting of total bladder RNA. Reactivation of pitS was detected via RT-PCR in five of the six bladder tumors in animals treated with 5-Aza-CdR (Fig. 5), demonstrating that a dor mant pitS gene could be effectively reactivated by the drug in vivo. 30% @-‘‘4% Strain @ =G2/M =G1 I=s 98 Downloaded from cancerres.aacrjournals.org on June 16, 2017. © 1998 American Association for Cancer Research. INHIBITION OF DNA METHYLATION RESTORES GROWTH CONTROL A 1.65 C 0 I1O-7M 5AzaCdR U 5x1 O-7M 5AzaCdR 1.55 11.0. 0. Cl) 1.45 p.-. II @0 1.35 1.25 L0 + EE 1.15 .E.E 1.05 00 ,@ 11 n o@ 1 2 3 Passage B @ @ 4 5 8 9 10 with 5AzaCdR g o Passage #after Treatment with1O-7M 5AzaCdR +C 0 1 2 3 4 5 6 7 8 9 10 I —@•.....—._ •1 @ .@ @ ¶@ g @ 7 # after Treatment p16 mRNA GAPDH mRNA 6 ‘ft .@ pass.9 S after Treatment with 5x10-7M 5AzaCd@!@@ o ‘i 2 3 4 5 6 7 8 g io p16mRNA j @ GAPDHmRNA —0 I Fig. 4. Heritability of 5-Aza-CdR-mediated p16 induction in T24 cells. In A. T24 cells were treated with either l0@ Mor 5 X l0@ M5-Aza-CdR for 24 h. After drug removal, cells were maintained in culture and passaged every 3 days prior to approaching confluency. Cell doubling times were determined at each passage after drug treatment (as described in “Materials and Methods―). In B, cells were harvested for total RNA isolation, and RT-PCR-based p16 expression analysis was performed during each passage after treatment (as described in “Materialsand Methods―).The passage numbers after treatment are indicated (0—10).Growth suppression and p16 expression (induced by 5-Aza-CdR) were heritable for at least nine passages, and these effects were dose dependent. GAPDH mRNA levels were measured to control for cDNA input and integrity. Additional control experiments were performed in which total bladder T24 RNA from rats treated with PBS showed no detectable p16 expression culture to facilitate recovery from the immediate cytotoxic effects of the drug, and then injected s.c. into nu/nu mice. Table 1 shows the effects of 5-Aza-CdR on the tumorigenicity of T24 cells in vivo. Tumors induced via the injection of untreated T24 cells were approx imately 1.5 mm3 in size, whereas the tumors induced by T24 cells (data not shown). Effects of 5-Aza-CdR on Twnorigenicity of T24 Cells in Vivo. Tumors were induced in nu/nu mice via s.c. injection of either untreated T24 cells or T24 cells treated with 5 X i0@ M 5-Aza-CdR. cells were treated 1 1 days prior to injection, passed 99 Downloaded from cancerres.aacrjournals.org on June 16, 2017. © 1998 American Association for Cancer Research. once in INHIBITION OF DNA METHYLATION RESTORES GROWTh CONTROL 5AzaCdR-Mediatedp16 InductionInVivo 0 @ .@ confirming previous studies by Merlo et a!. (28). We have also demonstrated that both of the cell lines not expressing p16 (T24 and HCT 15) were hypermethylated at this locus, suggesting that p16 methylation may be a common event during the establishment of cell lines in vitro. p16 up-regulation in these three tumor cell lines may also explain how 5-Aza-CdR suppressed cell growth; however, the absence of p16 up-regulation in the remaining four tumor cell lines suggests that other growth-regulatory genes in different pathways may @øj Tumor+5AzaCdR + C@1 2 3 4 56 p16 mRNA be potential targets for de novo methylation. The induction of other —0@ GAPDH mRNA -0@ genes, either alone or in combination with pitS, may also explain the observed growth suppression in the T24, HCT15, and HCT116 cell lines. Although de novo methylation of growth-regulatory genes may accompany the development of primary tumors in vivo, the strong selection pressure during the establishment of these tumors into im mortal cell lines implicates that further de novo methylation of these genes may also occur in vitro. This phenomenon may explain why few primary tumors ever transform into immortal cell lines, and those tumor cells which do survive selection to become established lines often appear to divide more rapidly than they did in vivo. Homozygous deletion or intragenic mutation of growth-regulatory genes may also give cells a selective advantage in vivo or in vitro, leading to increased cellular proliferation. Transcriptional silencing via the de novo methylation of 5' promoter sequences of growth @• ===::::=:@:I Fig. 5. Reactivation of p16 by 5-Aza-CdR in vivo. p16 expression after 5-Aza-CdR treatment was detected via RT-PCR (with primers specific for human p16 cDNA) in T24 cells growing in nu/nu rats. Daily 5-Aza-CdR treatmenLswere performed (4-6 weeks after T24 cell inoculation) via i.p. injection into six individual nu/nu rats for 5 days (columns1—6; asdescribedin “Materials andMethods―). PBS wasinjectedinto additional nu/nu rats as a negative control. Total bladder RNA was harvested for p16 RT-PCR analysis 24 h after the last drug treatment (as described in “Materialsand Methods―),and GAPDH mRNA levels were measured to control for cDNA input and integrity. treated with 5-Aza-CdR averaged 0. 14 mm@in size (Table 1). The p16 gene was also shown to be expressed by RT-PCR analysis in six of six tumors induced by 5-Aza-CdR-treated T24 cells and in zero of six tumors induced by untreated T24 cells (data not shown). The activa tion ofpl6 and/or other growth-regulatory genes may explain why the tumors grew more slowly. These results demonstrate that the herita ble, growth-inhibitory effects of 5-Aza-CdR treatment in vitro are still observed in T24 cells subsequent to their implantation in vivo. regulatory genes represents an alternative mechanism for their mac tivation, and studies have shown that increased methylation of CpG islands accompanies the establishment of immortal tumor cell lines (3, 4). Thus, hemizygous deletion of one allele coupled to hypermethy lation of the remaining allele represents an alternative means for complete inactivation of tumor suppressor genes in either primary tumors or cancer cell lines. This is specifically demonstrated in the T24 cell line, which contains only one copy of chromosome 9 (which harbors the pitS locus), and contains a hypermethylated p16 gene on the remaining allele. Numerous investigations have described reactivation of genes by 5-Aza-CdR, which include loci on the inactive X chromosome (37), the VHL gene (16), the E-cadherin gene (38), and the estrogen receptor gene (17). Our studies ofpl6 reactivation in the T24 bladder tumor cell line represents a good system for investigating the general effects of 5-Aza-CdR during the reactivation of genes silenced by de novo methylation. Other cell lines containing various hypermethylated genes that are inducible by 5-Aza-CdR in vitro also represent good systems for study. The demonstration of pitS expression and growth suppression by 5-Aza-CdR after 41 cell population doublings also suggests the potential of this drug as an effective chemotherapeutic agent. Whether the gradual decrease in pitS expression during subse quent cell passages arose from the de novo methylation of this gene or the selection of cells unaffected by 5-Aza-CdR needs to be determined. 01 arrest was also observed in T24 cells after drug treatment, and this growth inhibition may have been achieved by reactivating a dormant plo gene rather than introducing an exogenous gene as described in previous studies (39, 40). It is possible, however, that the DISCUSSION Our data show that brief treatment of established human tumor cell lines, but not diploid fibroblasts, with 5-Aza-CdR leads to a substan tial increase in the doubling times of surviving cells. Because all seven cell lines analyzed had the same response and 5-Aza-CdR is well known for its ability to induce the expression of genes silenced by de novo methylation, the most likely explanation for the result is that reexpression of silenced growth-regulatory genes inhibited cell growth. Other genes besides growth-regulatory genes may also be activated by 5-Aza-CdR, resulting in growth suppression. Alternative explanations for these phenomena could be the cytotoxic activity of the drug or the induction of other cellular physiological changes (i.e., changes in nucleotide pools), which normally slow cell growth. We think that these explanations are unlikely because cells were allowed to recover from the immediate cytotoxicity of 5-Aza-CdR before the growth kinetics were measured, and in the case of the T24 cells, the effects were apparent for up to 10 passages (41 population doublings) after drug treatment. T24 cells treated with equitoxic doses of Ara-C (which does not inhibit DNA methylation) showed no heritable growth inhibition, confirming that the cytotoxic effects of 5-Aza-CdR did not suppress cell growth. Furthermore, neither fibroblast strain tested showed growth inhibition at equal drug doses, which caused equivalent cytotoxicities and which should cause similar changes in cellular physiology. In three of the seven cases examined, we observed expression of the pitS gene after treatment with 5-Aza-CdR. pitS negatively regulates the G1/S transition of the cell cycle; therefore, loss of pitS function should potentiate the transition of cells into S-phase and increase cellular proliferation. Hypermethylation ofpl6 in various cancers has also been reported in several studies (19, 20, 28—31),and our study demonstrates that a dormant pitS gene can be reactivated in vitro, Table 1 Effects of 5-Aza-CdR on tumorigenicity in vivo T24 cells were treated for 24 h with 5-Aza-CdR (5 X lO@ M)in vitro. These cells were injected s.c. into the right and left flanks of nu/nu mice after 11 days, allowing time for recovery from the immediate cytotoxic effects of the drug. Untreated T24 cells were used as a control. Individual tumors induced by either 5-Aza-CdR-treated or untreated T24 cells were measured 4 weeks after injection. -5AzaCdR+5AzaCdRAverage tumor size Range1.5 mm3)Sample size(n mm@ mm@ (0—2.4 mm3)0.14 = 16)(n (0-0.3 = 14) 100 Downloaded from cancerres.aacrjournals.org on June 16, 2017. © 1998 American Association for Cancer Research. INHIBI11ON OF DNA METHYLATION RESTORES GROWTH CONTROL increased proportion 18. Herman, J. 0., Jen J., Merlo, A., and Baylin, S. B. Hypermethylation-associated inactivation indicates a tumor suppressor role for pI5IINK4B. Cancer Res., 56: 722—727, 1996. 19. Herman, J. G., Merlo, A., Mao, L., Lapidus, R. G., Issa, J-P., Davidson, N. E., of cells in G@ resulted from the nonspecific reactivation of other growth-regulatory genes by 5-Aza-CdR, which @ may act alone or in combination with pitS. To address this issue, we also investigated the cell cycle profile of 5637 cells, which express pitS before and after 5-Aza-CdR treatment. These cells did not exhibit a dramatic increase of cells in G@ and normal fibroblasts treated under the same conditions showed a negligible change in cell cycle profile (Fig. 3). Although these results are consistent with the hypothesis that pitS induction by 5-Aza-CdR mediates G1 arrest in T24 cells, addi Sidransky Tornout, J. M., and Jones, P. A. Methylation of the 5' CpG island of the p16 tumor suppressor gene in normal and transformed human tissues correlates with gene silencing. Cancer Res., 55: 453 1—4537,1995. 21. Zhang, Y., Shields, T., Crenshaw, T., Hao, Y., Moulton, T., and Tycko, B. Imprinting of human H19: allele-specific CpG methylation, loss of the active allele in Wilms' tumor and potential for somatic allele switching. Am. 3. Hum. Genet., 53: 113—124, 1993. 22. Harris, M. Induction of thymidine kinase in enzyme deficient Chinese hamster cells. Cell,29: 483—492, 1982. 23. Jones, P. A.. and Taylor. S. M. Cellular differentiation, cytidine analogs, and DNA methylation.Cell,20: 85—93, 1995. 24. Pinto,A., andZagonel,V. 5-Aza-2'-deoxycytidine (Decitabine)and 5-Aza-cytidine in the treatment of acute myeloid leukemias and myelodysplastic syndromes: past, present, and future trends. Leukemia (Baltimore), I: 51-60, 1993. 25. Seimno, M., Hannon, G. J., and Beach, D. A new regulatory motif in cell-cycle control causing specific inhibition ofcyclin D/CDK4. Nature (Lond.), 386: 704—707, REFERENCES 1993. 26. Nobori, T., Miura, K., Wu, D. J., Lois, A., Takabayashi, K., and Carson, D. A. Deletions of the cyclin dependent kinase 4-inhibitor gene in multiple human cancers. 1. Jones, P. A. DNA methylation errors and cancer. Cancer Rca., 56: 2463-2467, 1996. 2. Baylin, S. B., Makos, M., Wu, J., Yen, R-W. C., deBustros, A., Vertino, P., and Nelkin, B. D. Abnormal patterns of DNA methylation in human neoplasia: potential consequencesfor tumorprogression.CancerCells.3: 383-390,1991. 3. Jones, P.A.,Wolkowicz, M.J.,Rideout, W.M.,ifi, Gonzales, F.G.,Marziasz, C.M., Coetzee, G. A., and Tapscott, S. J. Dc novo methylation of the MyoDi CpG island during the establishment of immortal cell lines. Proc. Nail. Aced. Sci. USA. 87: 6117-6121, 1990. 4. Antequera, F., Boyes, J., and Bird, A. High levels ofde novo methylation and altered Nature (Lond.), 388: 753—756,1994. 27. van der Reit, P., Nawroz, H., Hruban, R. H., Cono, R., Tokino, K., Koch, W., and Sidransky, D. Frequent loss of chromosome 9p2l—22early in head and neck cancer progression. Cancer Res., 54: 1156—1 158, 1994. 28. Merlo, A., Herman, J. G., Mao, L, Lee, D. J., Gabrielson, E., Burger, P. C., Baylin. S. B.,andSidransky,D. 5' CpGislandmethylationis associatedwithtranscriptional silencing of the tumor suppressor pl6/MTS1 in human cancers. Nat. Med.. I: 686—692,1995. 29. Costello, J. F., Berger, M. S., Huang H-T.. and Cavenee, W. K. Silencing of chromatinstructureat CpGislandsin cell lines.Cell,62: 503—514, 1990. 5, Bird, A. P. CpG-rich islands and the function of DNA methylation. Nature (Lond.), 209—213, 1986. pJ6ICDKN2 expression in human gliomas by methylation and chromatin condensa 6. Gardiner-Garden, M., and Frommer, M. CpG islands in vertebrate genomes. J. Mol. Biol., 196: 261-282, 1987. tion. Cancer Res., 56: 2405—2410,1996. 30. Lo, K. W., Cheung, S-T., van Hasselt, A., Tsang Y-S.. Ma, K-F., Chung Y-F., Woo, K. K. S., Lee, J. C. K., and Huang, D. P. Hypermethylation of the pitS gene in nasopharyngeal carcinoma. Cancer Res., 56: 2721—2725,1996. 7, Li, E., Berad, C., and Jaenish, R. DNA methylation in genomic imprinting. Nature (Lond.), 386: 382—385.1993. 8. Ferguson-Smith, A., Sasaki, C. H., Cattanach, B. M., and Surani, M. A. Parental origin specific epigenetic modification of the mouse H19 gene. Nature (Lond.), 382: 31. Otterson,G. A.,Khlief,S. N.,Chen,W.,Coxon,A.B.,andKaye,F.J. CDKN2gene silencing in lung cancer by DNA hypermethylation and kinetics of p16INK4 protein induction by 5-aza-2'-deoxycytidine. Oncogene, Ii: 1211—1216, 1995. 751—755,1993. 9. Feinberg, A. P., and Vogelstein, B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature (Lond.), 301: 89—92,1983. 32. Gonzalgo, M. L., and Jones, P. A. Rapid quantitation of methylation at specific sites using methylation-sensitive single nucleotide primer extension (Ms-SNuPE). Nucleic Acids Res., 25: 2529—2531,1997. 10. Jones,P. A.,Rideout,W.M.,Shen,J. C.,Spruck,C. H.,andTsai,Y.C. Methylation, mutation, and cancer. Bioessays, 14: 33—38,1992. Gonzales, F. G., and Jones, P. A. Progressive increases in the methylation status and heterochromatinization of the Myo Dl CpG island during oncogenic transformation. Mol.Cell.Biol.,14: 6143—6152, 1994. 12. Baylin, S. B., Fearon, E. R., Vogelstein, B., de Bustros, A., Sarkis, S. J., Burke, P. J., Steal, S. P., and Nelkin, B. D. Hypermethylation of the 5' region of the calcitonin gene is a property of human lymphoid and acute myeloid malignancies. Blood, 70: 412—417, 1987. 13. Vertino, P. M., Spillare, E. A., Harris, C. C., and Baylin, S. B. Altered chromosomal methylationpatternsaccompanyoncogene-induced transformationof humanbron chial epitheial cells. Cancer Res., 53: 1684—1689,1993. 14. Greger, V., Passarge, E., Hopping, W., Messmer, E., and Horsethmke, B. Epigenetic changes may contribute to the formation and spontaneous regression of retinoblas toma. Hum. Genet., 83: 155—158,1989. 15. Sakai, T., Togushida, J., Ohtani, N., Yandell, D. W., Rapaport, J. M., and Dryja, T. P. gene is Cancer Res., 55: 4525—4530, 1995. by 5-Aza-CdR in vivo and the decreased tumorigealcity of 5-Aza CdR-treated T24 cells in vivo further suggest the chemotherapeutic potential of 5-Aza-CdR. Reactivation of growth-regulatory genes silenced by de novo methylation suggests clinical applications in which genes inactivated by hypermethylation in human tumors can be induced by 5-Aza-CdR during anticancer therapy. 11. Rideout, W. M., Eversole-Cire, P., Spruck, C. H., Hasted, C. M., Coetzee, G. A., of the CDKN2/p16/MTSI 20. Gonzalez-Zulueta, M., Bender, C. M., Yang, A. S., Nguyen, T., Beart, R. W., Van tional studies must be performed to confirm this. The induction of p16 321: D., and Baylin, S. B. Inactivation frequently associated with aberrant DNA methylation in all common human cancers. 33, Clark, S. J., Harrison, J., Paul, C. L., and Frommer, M. High sensitivity mapping of methylated cytosines. Nucleic Acids Res., 22: 2990—2997, 1994. 34, Frommer, M., McDonald, L E., Millar, D. S., Collis, C. M., Watt, F., Grigg, G. W., Molloy, P. L., and Paul, C. L. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc. NatI. Aced. Sci. USA, 89: 1827—1831, 1992. 35, Jones, P. A. Gene activation by 5-azacytidine. In: A. Razi, H. Cedar, and A. D. Riggs (edt.), DNA Methylation: Biochemistry and Biological Significance, pp. 165—187. New York: Springer-Verlag, 1984. 36. Hara, G., Smith, R., Parry, D., Tahara, H., Stone, S., and Peters, G. Regulation of p16/CDKN2 expression and its implications for cell immortalization and senescence. Mol. Cell. Biol., 16: 859—867,1996. 37. Jones, P. A., Taylor, S. M., Mohandas, T., and Shapiro, L. J, Cell cycle-specific reactivation of an inactive X-chromosome locus by 5-azadeoxycytidine. Proc. Natl. Aced.Sd. USA,79: 1215—1219, 1982. Allele-specific hypermethylation of the retinoblastoma tumor suppressor gene. Am. Hum. Genet., 48: 880—888,1991. 38. Yoshiura, K., Kanai, Y., OChiari,A., Shimoyama, Y., Sugimura, T.. and Hirohashi, tumor-suppressor gene by DNA methylation in renal carcinoma. Proc. NatI. Aced. Sci. USA. 91: 9700—9704, 1994. 17. Ottaviano, Y. L., Issa, J-P., Parl, F. F., Smith, H. S., Baylin, S. B., and Davidson, N. E. inhibition of tumor cell proliferation by the pI6'@4 gene by an adenovirus vector. Cancer Ret., 55: 3250—3253,1995. S. Silencingof the E-cadherininvasion-suppressor gene by CpG methylationin human carcinomas. Proc. Natl. Aced. Sci. USA, 92: 7416—7419,1995. 16. Herman.J. G.,Latif,F.,Weng,Y.,Lerman,M.I.,Zbar,B.,Liu,S.,Samid,D.,Dusts, D-S, Gnarra,3. R., Linehan,W. M., and Baylin,S. B. Silencingof the VHL 39, Jin, X., Nguyen, D., Zhang, W-W, Kiritis, A. P.. and Roth, J. A. Cell cycle arrest and 40. Amp, A., Nishikawa, R., Furnari, F. B., Cavenee, W. K., and Su Huang, H-i. Replacement of the pitS gene suppresses human glioma cell growth. Cancer Res., 55: Methylation of the estrogen receptor gene CpG island marks loss of estrogen receptor expression in human breast cancer cells. Cancer Rca., 54: 2552—2555,1994. 1371—1374, 1995. 101 Downloaded from cancerres.aacrjournals.org on June 16, 2017. © 1998 American Association for Cancer Research. Inhibition of DNA Methylation by 5-Aza-2′-deoxycytidine Suppresses the Growth of Human Tumor Cell Lines Christina M. Bender, Martha M. Pao and Peter A. Jones Cancer Res 1998;58:95-101. Updated version E-mail alerts Reprints and Subscriptions Permissions Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/58/1/95 Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at [email protected]. To request permission to re-use all or part of this article, contact the AACR Publications Department at [email protected]. Downloaded from cancerres.aacrjournals.org on June 16, 2017. © 1998 American Association for Cancer Research.