<<

Proc. Nati. Acad. Sci. USA Vol. 88, pp. 8572-8576, October 1991 Medical Sciences Stimulation of dihydrofolate promoter activity by antimetabolic drugs HELEN B. EASTMAN, ANDREW G. SWICK*, M. CHRISTINE SCHMITTt, AND JANE CLIFFORD AZIZKHAN Lineberger Cancer Research Center and the Departments of Pharmacology and Pediatrics, University of North Carolina, Chapel Hill, NC 27599-7295 Communicated by Gertrude B. Elion, June 17, 1991 (receivedfor review September 19, 1990)

ABSTRACT (DHFR; EC 1.5.1.3) Transcriptional activation ofthe DHFR gene by MTX is an is required in folate for the synthesis of purines, equally plausible hypothesis to explain the increased mRNA thymidine, and glycine. Although there have been several and levels, and this possibility has not been carefully reports ofinduction ofDHFR enzyme by methotrexate (MTX), addressed. We have previously characterized the protein/ a drug that competitively inhibits DHFR, there are no studies DNA interactions in the DHFR promoter that are required reported that examine the effect of MTX on DHFR gene for efficient and accurate transcription (11-13). There are transcription. We have examined the effect of MTX and other several GC elements (shown in Fig. 1), consensus binding inhibitors of DNA synthesis on DHFR transcription using a sites for the transcription factor Spl, present in the hamster transient expression assay. MTX stimulates transient expres- DHFR promoter that are essential for DHFR transcription sion in a concentration-dependent manner from a hamster (11) and that control transcription start site utilization (12). DHFR promoter construct containing 150 base pairs 5' to the This (G+C)-rich promoter motif is common to many so- start of transcription. Addition of either tetrahydrofolate or called housekeeping genes as well as several growth factor hypoxanthine plus thymidine prevents the promoter induction and growth factor receptor genes and oncogenes (ref. 11 and in response to MTX, suggestiug that stimulation by MTX references therein). There are also two overlapping recogni- results from inhibition of these metabolites. Furthermore, two tion sites for the transcription factor E2F that lie immediately other antimetabolic drugs-fluorodeoxyuridine and hydroxy- 3' to the major start of transcription; mutation of these sites urea-als stimulate the DHFR promoter in a concentration- results in a significant reduction of transcription (13). The dependent manner. In contrast, aphidicolin, which blocks cell functional significance of a sequence is often growth through inhibition of DNA polymerase a, has no effect suggested by evolutionary conservation; the 5' flanking re- on theDHFR promoter. The potential relevance ofthese results gions of the human (14), murine (15), and hamster (16, 17) to cross-resistance to chemotherapeutic agents and to the DHFR genes exhibit a high degree of sequence conservation, process of gene amplification is discussed. particularly within the binding sites for the transcription factors Spl and E2F. Because the DHFR promoter is similar Dihydrofolate reductase (DHFR; EC 1.5.1.3) catalyzes the to those ofmany housekeeping genes, it is possible that many reduction of folate to dihydrofolate and then to tetrahydro- other genes in this class may also be transcriptionally acti- folate. Reduced folates are required cofactors for de novo vated by antimetabolic drugs, such as those discussed herein. synthesis of thymidylate, purines, and glycine. DHFR is We have examined the effects of treatment with MTX and required for maintenance ofcell growth and is thus expressed other inhibitors of DNA synthesis on DHFR promoter ac- in virtually all growing cells. The chemotherapeutic agent tivity to address the question of whether these agents tran- methotrexate (MTX) is a dihydrofolate analog that binds scriptionally activate the DHFR gene. In this paper we report DHFR, thereby inhibiting its enzymatic activity and prevent- that treatment of Chinese hamster ovary (CHO) cells with ing . Chronic MTX treatment can lead to MTX MTX increases expression from a DHFR promoter chloram- resistance; one mechanism whereby cells become resistant is phenicol acetyltransferase (CAT) construct. We show that amplification of the DHFR gene, which results in increased 5-meth- DHFR mRNA and protein levels (1). Very little is known stimulation by MTX can be inhibited by addition of about the early events that occur between MTX treatment yltetrahydrofolate, which obviates the requirement for and gene amplification. DHFR to reduce folates. This increase can also be prevented Several laboratories have reported that DHFR enzyme by addition of hypoxanthine plus thymidine, which supplies levels are elevated by treatment of cells with MTX (2-4) or the two end-products ofthe biosynthetic pathway involved in hydroxyurea (4) and that the stimulation by hydroxyurea is DNA replication that is inhibited by MTX. We also examined due to elevated mRNA levels (4). The drug-induced increase the effect of three other DNA synthesis inhibitors in DHFR mRNA levels may result from increased transcrip- aphidicolin, fluorodeoxyuridine, and hydroxyurea-on the tion, decreased mRNA degradation, or accumulation of DHFR promoter. We present data to demonstrate that treat- mRNA due to blockage of cells at the G1/S boundary of the ment of cells with aphidicolin does not affect DHFR pro- cell cycle, when DHFR gene transcription is maximal (5, 6). moter activity, whereas hydroxyurea or fluorodeoxyuridine The stimulation ofDHFR by MTX and hydroxyurea may also treatments stimulate the DHFR promoter in a concentration- be the result of inhibition of DNA synthesis, resulting in dependent manner. Thus, the DHFR promoter is stimulated rereplication of the gene (i.e., gene amplification) and sub- by at least three different antimetabolites, and the stimulation sequent elevated expression from the increased number of is not simply the result of inhibition of cell growth. transcription templates (7). Consistent with this idea, hy- droxyurea, which blocks DNA replication, and hypoxia and Abbreviations: DHFR, dihydrofolate reductase; MTX, methotrex- UV irradiation, which damage DNA, all have been found to ate; CAT, chloramphenicol acetyltransferase; CHO, Chinese ham- increase the frequency of DHFR gene amplification (8-10). ster ovary; AdMLP, adenovirus major late promoter; DMSO, di- methyl sulfoxide; ANOVA, analysis of variance. *Present address: Department of Biological Chemistry, Johns Hop- The publication costs of this article were defrayed in part by page charge kins University, Baltimore, MD 21205. payment. This article must therefore be hereby marked "advertisement" tPresent address: Vanderbilt University School of Medicine, Nash- in accordance with 18 U.S.C. §1734 solely to indicate this fact. ville, TN 37215. 8572 Downloaded by guest on September 29, 2021 Medical Sciences: Eastman et al. Proc. Natl. Acad. Sci. USA 88 (1991) 8573

1 11 1 LE2F-e IIn I I II I ------ATG 0o r. 0 FIG. 1. DHFR promoter transient expression vector. pDHF/CAT was constructed by cloning the hamster DHFR promoter fragment from nucleotide position -210 to -23 (relative to ATG = position 1) 5' to the bacterial CAT gene and simian 40 poly(A) signal in a pUC18 vector. The major start site of DHFR transcription (bold arrow) is at nucleotide position -63, and nucleotide position -107 is the minor start site. GC boxes are indicated by stippled boxes and are numbered in the text I-IV, proximal to distal; open boxes indicate additional conserved sequence elements. Binding sites for the transcription factor E2F are indicated by the hatched box. EXPERIMENTAL PROCEDURES CAT) reported in this manuscript contained DNA sequence from the positions -210 to -23 bp relative to ATG. The Construction of Clones. The DHFR promoter constructs were derived from a genomic CHO clone previously de- sequence elements in this construct that are highly conserved scribed (16). The construct that is used in the present studies, in the mouse and human DHFR promoters (shown in Fig. 1) designated pDHF/CAT, contains hamster DHFR promoter consist of four GC boxes (stippled boxes), two 20-bp regions sequence from positions -210 to -23 base pairs (bp) relative 3' to boxes I and II (open boxes), and two overlapping to the start of translation (i.e., -147 to +40 relative to the recognition sites for the transcription factor E2F (hatched major transcription start) cloned 5' to the coding sequence for box). Each of these elements has been shown to be func- the bacterial CAT gene fused to a poly(A) sequence from the tionally important for constitutive activity of the hamster simian virus 40 large tumor antigen (11). The clone used as a DHFR promoter (11-13). Maximal promoter activity is ob- control consists of the adenovirus major late promoter fused tained with this construct; constructs with additional 5' to CAT (AdMLP/CAT) (18) in the same expression vector sequence are equally active in transfection experiments (11), construct. whereas deletion or mutation of GC boxes I, II, or III Cell Culture and Transfections. CHO cells were grown in significantly reduces transcriptional efficiency (12). monolayer in Eagle's minimal essential medium (MEM) To determine the effect of MTX on the DHFR promoter, supplemented with 5% fetal bovine serum, nonessential CHO cells were transfected with pDHF/CAT and treated amino acids, glutamine, and penicillin/streptomycin. The with MTX at 0.05, 0.1, 0.15, 0.2, and 0.25 /ug/ml. A linear doubling time of CHO cells is -20 hr under these growth increase in CAT activity was observed in response to MTX conditions. Twenty-four hours prior to transfection, cells treatment from 0.05 to 0.2 pug/ml (Fig. 2). In cells treated with were plated at -5 x 105 per 10-cm plate in Eagle's MEM 0.2 ,gg of MTX per ml of medium, there was an -5-fold supplemented with 5% dialyzed fetal bovine serum (dFBS). stimulation of CAT expression in this experiment. The max- Promoter-CAT constructs were transfected by the calcium in the 20 times that MTX has been tested phosphate coprecipitation method essentially as described by imal stimulation Graham and van der Eb (19). Within each experiment, a ranged from 4- to 20-fold. single DNA precipitate at a concentration of 10 Ag/ml was To determine whether general transcription is stimulated formed for each DNA construct. The individual plates were by MTX treatment, the AdMLP (18), which bears no se- rinsed and the monolayer was drained; 0.5 ml of DNA quence homology to the DHFR promoter, was tested. As precipitate (5 ,ug) was added dropwise to the individual shown in Fig. 2, CAT expression under the control of the plates. Complete medium containing 5% dFBS was added AdMLP was not stimulated by MTX treatment ofCHO cells. after 30 min at room temperature. Four hours later, the Thus, stimulation of the DHFR promoter by MTX is not the monolayer was washed and complete medium containing result of a general transcriptional response. dFBS was added to the cells. Test agents were added directly Metabolic Signals. In an effort to understand the signal(s) to the medium as indicated in the figure legends. MTX was involved in the elevation ofDHFR promoter activity, various obtained from the Drug Synthesis and Chemistry Branch of substances that obviate the requirement for certain metabolic the National Cancer Institute; MTX was reconstituted in 0.1 cofactors were added in addition to MTX. Addition of 2 ,uM M NaOH in a 50 mg/ml stock solution that was diluted in water for use and stored in the dark at -20°C. Aphidicolin, 6.0 hydroxyurea, and fluorodeoxyuridine were obtained from Sigma. Aphidicolin was reconstituted in dimethyl sulfoxide 5.0 DHFR (DMSO) at 10 mg/ml; DMSO was added to cells in the > volumes used for aphidicolin treatment to control for vehicle :t 4.0 - effects. Hydroxyurea and fluorodeoxyuridine were reconsti- tuted in water at 1 M and 100 mM, respectively. 3.0- Assay for CAT Activity. Following a 48-hr incubation, cells 0 were harvested and subjected to freeze-thaw lysis. Lysates .;5lY 2.0- were assayed for protein content by the Bradford (20) assay. AdMLP CAT activity was measured by a fluor-diffusion assay using 1.0e [acetyl-3H]acetyl CoA (200 mCi/mmol; 1 Ci = 37 GBq; NEN), essentially as described by Neumann et al. (21), 0.0 - except that 0.4 ,Ci of [3H]acetyl CoA plus 40 AM unlabeled 0.00 0.05 0.10 0.15 0.20 0.25 acetyl CoA were used per assay. Twenty micrograms of protein cell extract was assayed, and CAT activity was Methotrexate (jug/ml) calculated as pmol ofproduct acetylated per ,ug ofprotein. To make comparisons between experiments, data are presented FIG. 2. Effect of MTX on transient expression from the DHFR as relative CAT activity, with wild-type untreated CAT promoter. pDHF/CAT or AdMLP/CAT plasmid constructs (5 ,g each) were transfected by the calcium phosphate procedure into expression normalized to 1. CHO cells. MTX was added to the medium at the concentrations indicated. After 48 hr, cells were harvested and assayed for protein RESULTS concentration and CAT activity was measured in cellular lysates on equal amounts of protein. CAT activity of untreated cells was Effects of MTX on DHFR Promoter Activity. The hamster assigned a relative value of 1. These results are representative of DHFR promoter construct used in the experiments (pDHF/ three repetitions of this experiment. Downloaded by guest on September 29, 2021 8574 Medical Sciences: Eastman et al. Proc. Natl. Acad. Sci. USA 88 (1991) 5-methyltetrahydrofolate, which circumvents the cellular re- A quirement for DHFR, restored cell growth (Fig. 3B) and 10 - --.__ ..-

blocked the stimulation ofDHFR promoter activity (Fig. 3A). r 5-Methyltetrahydrofolate restores tetrahydrofolate pools, MT X (Ig rnI 8 0 0 which are specifically blocked by MTX; however, 5-meth- F: E.025 yltetrahydrofolate also competitively inhibits MTX transport 6 El .05 into cells (22,23). To address the possibility that the inhibition -El -10 20 ofCAT activity was due to decreased MTX uptake rather than C) 4 to activation of the promoter, we added hypoxanthine and H- thymidine, alone and in combination with MTX, to circumvent 2 Er the inhibition of purine and thymidine biosynthesis, respec- LU tively, by MTX. Neither hypoxanthine nor thymidine has been 0 shown to affect MTX transport, but both of these compounds C H 4 F HX Thy HX+ThV have been shown to rescue cells from MTX treatment (24). Both 17.6 ,uM hypoxanthine and 100 gM thymidine alone inhibited stimulation of the DHFR promoter by MTX, hypo- B xanthine to a greater extent than thymidine. When both agents 1.5 - ---- were added together, stimulation by MTX was completely MTX (uLg ml) blocked, suggesting that the stimulated transcription is medi- MO E .025 ated by decreased purine and thymidine metabolism (Fig. 3A). z 1.0 l .05 When added in the absence of MTX, neither 5-methyltetrahy- Li .10 drofolate, thymidine, nor hypoxanthine significantly affected -n 20 CAT expression (data not shown). U' 0.5 Relatioship Between Effects on Cell Growth and Promoter Stimniation. Treatment ofcells with MTX alone stimulated the DHFR promoter in a concentration-dependent manner (Figs. Lu 2 and 3A, groupC). In an effort to determine whether the effect 0. of MTX was due to cell killing and a stress-related response, C H 4F HX Thy HX.Jhy especially at the higher concentrations, cells treated as shown and the protein content ofthe cellular FIG. 3. Effect of 5-methyltetrahydrofolate (H4F), hypoxanthine in Fig. 3A were counted, (HX), and/or thymidine (Thy) on MTX stimulation of the DHFR lysate was compared to the cell number. We have found that promoter and on cell growth. CHO cells were transfected with there is a linear relationship between cell number and protein pDHF/CAT (5 ,ug) by the calcium phosphate transfection method. concentration (data not shown); we therefore routinely deter- Four hours later, the following reagents were added to the medium mined protein concentration as a measure of cell growth. as indicated: 0.025-0.2,ug of MTX per ml; 2 jAM H4F; 17.6,uM HX; These data are graphed normalizing the protein content (or cell 100 AtM Thy. All agents were added at 0 and 24 hr. C, control number) ofthe untreated sample to 1.0 to readily compare one (treatment with MTX alone). (A) Results ofthe CAT assay performed using equal amounts of protein in cell lysates 48 hr after transfection. experiment with another. As can be seen in Fig. 3B, when cells For each treatment group, CAT activity for cells not treated with are treated with MTX alone (group C) cell growth is blocked MTX was assigned a relative value of 1. Absolute values for CAT completely by MTX at 0.025,ug/ml, and very little additional activity in this experiment ranged from 12 pmol of acetylated product effect on cell number is seen at the higher concentrations, formed per /Ag of protein in the untreated control to 99.5 pmol/g at where there is a dose-dependent increase in CAT activity (Fig. 0.20 jtg of MTX per ml. (B) Effect ofthese treatments on cell growth. 3A, group C). From 0.025 to 0.2 gg/ml, the cell number is Protein concentrations in the cellular lysates were determined by the -20% of the untreated sample. This result is consistent with Bradford assay (20) and normalized to a control value of 1.0 as cell growth during the 48-hr treatment, since during described for CAT assays. Protein concentration is a measure of cell blocked number as discussed in Results. These results are representative of this period the cell number of the untreated sample increased three repetitions of this experiment. Two-way analysis of variance 5-fold, corresponding to 2.3 doublings. Thus, within the range (ANOVA) ofthe data inA revealed that the MTX-treated samples are where a linear relationship between drug concentration and significantly different from one another and from the untreated promoter activity is observed, MTX blocked proliferation sample (P c 0.05) at all of the concentrations tested. Furthermore, equally but did not kill cells. Addition of the various metab- the addition of H4F, HX, or HX plus Thy with MTX at all of the olites did not equivalently affect proliferation and DHFR concentrations eliminated this difference (P s 0.05), whereas the promoter activity, suggesting that the effect ofMTX on DHFR addition of Thy had no significant effect as compared with MTX transcription is not due simply to inhibition of cell growth. alone at all of the concentrations tested, with the exception of0.2 ,ug Effect of Other Inhibitors of DNA Synthesis. To determine of MTX per ml (P c 0.05). if the DHFR promoter was stimulated by other inhibitors of DNA synthesis and cell growth, we tested the effect of three Taken together, these data suggest that DHFR promoter other agents on the DHFR promoter. Cells were transfected activity is significantly stimulated in cells treated with MTX, with pDHF/CAT and treated with aphidicolin (10-100 ,ug/ hydroxyurea, and fluorodeoxyuridine. This stimulation is not ml), an inhibitor of DNA polymerase a. Aphidicolin blocked simply the result of inhibition of DNA synthesis and cell cell proliferation (Fig. 4B), but had no effect on CAT expres- proliferation; aphidicolin, which blocks proliferation as ef- sion from the DHFR promoter (Fig. 4A). The effects of two fectively as MTX, does not affect promoter activity. Fur- other antimetabolites on the DHFR promoter were also thermore, MTX stimulation of CAT activity is dose depen- tested. Fluorodeoxyuridine, an inhibitor of thymidylate syn- dent at drug concentrations that have an equal antiprolifer- thetase, stimulated the DHFR promoter in a concentration- ative effect. dependent manner (Fig. SA). Hydroxyurea, an inhibitor of reductase, in the range of 0.05-1 mM, also DISCUSSION stimulated the DHFR promoter in a concentration-dependent We have explored the response of the DHFR promoter to manner to a maximum of 2.5-fold (Fig. SC). Both of these MTX, and to other inhibitors of DNA synthesis, and found agents inhibited cell growth (Fig. 5 B and D), although not as that MTX as well as the antimetabolites fluorodeoxyuridine completely as MTX or aphidicolin. and hydroxyurea stimulate the DHFR promoter, whereas Downloaded by guest on September 29, 2021 Medical Sciences: Eastman et al. Proc. Natl. Acad. Sci. USA 88 (1991) 8575

A B A B 3 >- 1.2 _..us 1.2 LL ,

-) v.. 0.3 -. 0.61.0 -j0.1.01-: LU U 0.4>04 L 0. C F D r, LU L Lu .. cc 0.0 0. cc 0 A 25 5 D 0 10 50 100 0 10 50 100 FUdR DOSEi M. FUdR DOSEi.:rl: APC DOSE (Gig ml) APC DOSE ..g ml) D FIG. 4. Effect of aphidicolin (APC) on transient expression from 2- 3. W 1.2. the DHFR promoter and on cell growth. CHO cells were transfected S 1.0. with pDHF/CAT (5 Eg) by the calcium phosphate transfection Z 0.8. method. Four hours later, APC was added at the indicated concen- C-) 0'W, 0.6 trations. An equal volume ofDMSO, which was the vehicle for APC, w was added to a duplicate transfected plate. Cells were harvested and 21 Xlsll > 0.4. HH\nN lysates were prepared and assayed for protein concentration and \ 0.2 uJL-J CAT activity. (A) CAT activity in cells treated with the indicated 0. u0IC 0.0 concentration of APC, after subtracting the effect of DMSO alone. o .05 .1 .5 1 5 0 .05 .1 .5 1 5 DMSO stimulated CAT activity up to 2-fold at the highest amount HU DOSE (mM) HU DOSE (mM) (100 Aul in 10 ml). The absolute value ofCAT activity was conversion of 7.8 pmol of acetyl CoA per jug of cellular lysate in the control and FIG. 5. Effect of fluorodeoxyuridine (FUdR) and hydroxyurea the treated samples. These results are representative of three repe- (HU) on the DHFR promoter. CHO cells transfected with pDHF/ titions ofthis experiment. Two-way ANOVA revealed no significant CAT (5 Ag) by the calcium phosphate transfection method were differences in CAT activity between the APC-treated samples and treated for 48 hr with FUdR or HU at the indicated concentrations. the control at any of the APC concentrations tested (P < 0.05). (B) Cellular lysates were assayed for protein concentration and CAT Relative protein content of the cellular lysates at the indicated activity. Data shown are from one experiment; these results have concentration of APC, which was directly related to cell number. been observed in three separate experiments with the range of Cell number was 1o of control at the highest concentration ofAPC. induction with both drugs being 2- to 3-fold. (A) Effect of FUdR on CAT activity; the range of activity was 14.9 pmol of acetylated aphidicolin has no effect. DHFR enzyme levels have been product formed per ug of protein in the control to 31.9 pmol/,ug of protein in the cells treated with 5 ALM FUdR. Two-way ANOVA shown by others to be elevated in response to acute treatment demonstrated a significant effect of FUdR on CAT activity at 1 and of cells with MTX (2-4). The most common mechanism by 5 ,tM vs. control (P c 0.05). (B) Effect of FUdR on cell growth; the which DHFR enzyme is overproduced in MTX-resistant cells absolute values were 3.3 Aug/ml in the untreated sample and 1.5 is thought to be gene amplification; however, it is clear from jug/ml in the cells treated with 5 .tM FUdR. This represents -50%o our studies that there may also be an induction of DHFR inhibition of growth; complete growth inhibition was not observed transcription in response to MTX treatment of cells. even at higher drug concentrations. (C) Effect of HU on CAT Since DHFR plays an essential role in metabolism, we expression. In this experiment the untreated cells converted 14.9 examined metabolic that could mediate pmol of[3H]acetyl CoA per ag ofprotein and cells treated with 1 mM some possible signals HU converted 35.8 pmol/i&g. Two-way ANOVA demonstrated a enhanced DHFR transcription. DHFR is the only enzyme significant effect of HU at 0.5 and 1 mM vs. control (P < 0.07). (D) capable ofregenerating intracellular pools ofreduced folates, Cell growth was inhibited 50%, equivalent to the effect of FUdR. which are cofactors for glycine, purine, and thymidine bio- synthesis. Therefore, inactivation ofDHFR by MTX leads to mechanism by which the three antimetabolic agents (MTX, lower cellular tetrahydrofolate levels and subsequent reduc- fluorodeoxyuridine, and hydroxyurea) stimulate transcrip- tion ofcell growth (25). The effects ofMTX can be overcome tion. Since the three agents that we found stimulated the by 5-methyltetrahydrofolate (26). Addition of 5-methyltet- DHFR promoter are inhibitors of nucleotide biosynthesis, it rahydrofolate blocked the stimulation ofCAT activity and the is possible that stimulation of DHFR transcription occurs inhibition of cell growth by MTX. Since the effect of folate through a common pathway. The promoters for thymidylate could have been the result of decreased uptake of MTX (22, synthetase (27) and ribonucleotide reductase (28), like 23), we also examined the effect of addition of intermediates DHFR, have multiple GC boxes (putative Spl binding sites), whose biosynthesis is blocked by MTX. Addition of hypo- like TATAA xanthine alone or of both thymidine and hypoxanthine com- and thymidylate synthetase, DHFR, lacks both pletely prevented the stimulation, whereas addition of thy- and CCAAT motifs. We have shown that the GC boxes ofthe midine alone reduced the stimulation from 8-fold to 2-fold. hamsterDHFR promoter control the rate oftranscription and Hypoxanthine and thymidine separately partially blocked the start site selection (11, 12). It is interesting to speculate that antiproliferative effect of MTX, whereas addition of both all of these drugs may affect common transcription factors to hypoxanthine and thymidine restored normal growth. These activate transcription of a class of genes. results suggest that the antiproliferative effect of MTX and The DHFR promoter fragment used in the described ex- the effect ofthe drug on DHFR transcription are not mediated periments contains 40 bp 3' to the major start oftranscription by completely overlapping mechanisms. that could conceivably contribute to specific posttranscrip- We also examined the ability ofthree other DNA synthesis tional effects on CAT mRNA since this sequence is present inhibitors-aphidicolin, hydroxyurea, and fluorodeoxyuri- in the DHFR transcripts. Within this 40-nucleotide region, dine-to stimulate transcription of the DHFR promoter. the only sequence element conserved among the hamster, Aphidicolin blocks DNA replication by inhibition of DNA mouse, and human genes is the region corresponding to a polymerase a. Fluorodeoxyuridine is an inhibitor of thymi- for the transcription factor E2F. Although mu- dylate synthetase, and hydroxyurea inhibits ribonucleotide tation of the E2F region has been shown to affect transcrip- reductase. Aphidicolin did not affect the DHFR promoter, tion (13), there is no evidence to support a role for E2F in whereas hydroxyurea and fluorodeoxyuridine stimulated binding to RNA or stabilizing RNA. A mutation that abol- DHFR transcription. All of these agents had an antiprolifer- ished E2F interaction with the E2F sites decreased basal ative effect, suggesting that inhibition ofcell growth is not the promoter activity but did not affect relative MTX inducibility Downloaded by guest on September 29, 2021 8576 Medical Sciences: Eastman et al. Proc. Natl. Acad. Sci. USA 88 (1991) (data not shown). The CAT coding sequence, simian virus 40 1. Alt, F. W., Kellems, R. E., Bertino, J. R. & Schimke, R. T. region, and plasmid vector are identical in both the DHFR (1978) J. Biol. Chem. 253, 1357-1370. 2. Hillcoat, B. C., Swett, V. & Bertino, J. R. (1967) Proc. Natl. CAT construct and the AdMLP/CAT construct, which in Acad. Sci. USA 58, 1632-1637. these studies did not respond to MTX. Taken together, these 3. Domin, B. A., Grill, S. P., Bustow, K. F. & Cheng, Y. C. data suggest that MTX inducibility is not conferred by DHFR (1982) Mol. Pharmacol. 21, 478-482. promoter sequence 3' to the start oftranscription. Additional 4. Johnston, R. N., Feder, S. P., Hill, A. B., Sherwood, S. W. & posttranscriptional orother effects ofMTX onDHFR expres- Schimke, R. T. (1986) Mot. Cell. Biot. 6, 3373-3381. 5. Santiago, C., Collins, M. & Johnson, L. F. (1984) J. Cell. sion in cells are possible but do not appear to be involved in Physiot. 118, 79-86. the response observed in this study. Experiments are nec- 6. Farnham, P. J. & Schimke, R. T. (1985) J. Biol. Chem. 260, essary- to identify the specific DHFR promoter element that 7675-7680. mediates the effect of MTX and to study the effect of MTX 7. Mariani, B. A. & Schimke, R. T. (1984) J. Biol. Chem. 259, on the factor that binds to this element. 1901-1910. 8. Brown, P., Tlsty, T. D. & Schimke, R. T. (1983) Mol. Cell. The fact that MTX and other antimetabolites can increase Biol. 3, 1097-1107. transcription from the DHFR promoter is potentially relevant 9. Tlsty, T. D., Brown, P. C. & Schimke, R. T. (1984) Mol. Cell. to the issue of cellular cross-resistance to chemotherapeutic Biol. 4, 1050-1056. agents. Cross-resistance to drugs is aconfounding problem in 10. Sherwood, S. W., Schumacher, R. I. & Schimke, R. T. (1988) cancer chemotherapy; the clinical approach has been to Mol. Cell. Biol. 8, 2822-2827. 11. Swick, A, G., Blake, M. C., Kahn, J. W. & Azizkhan, J. C. empirically combine chemotherapeutic agents. It is clear that (1989) Nucleic Acids Res. 17, 9291-9304. selection of cells for resistance to two different drugs results 12. Blake, M. C., Jambou, R. C., Swick, A. G., Kahn, J. W. & in a frequency of cross-resistance that is often much greater Azizkhan, J. C. (1990) Mol. Cell. Biol. 10, 6632-6641. than the product of the two individual frequencies (29, 30). 13. Blake, M. C. & Azizkhan, J. C. (1989) Mol. Cell. Biol. 9, There are many possible mechanisms by which cross- 4994-5002. resistance can arise. We propose a mechanism whereby 14. Chen, M. J., Shimada, T., Moulton, A. D., Cline, A., Hum- phries, R. J., Maizel, J. & Nienhuis, A. W. (1984) J. Biol. several different drugs increase the activity or level of a Chem. 259, 3933-3943. common transcription factor, which could then stimulate 15. Farnham, P. J., Abrams, J. M. & Schimke, R. T. (1985) Proc. transcription of a range of genes with common promoter Natl. Acad. Sci. USA 82, 3978-3982. elements. Cells that are resistant to these agents may have 16. Azizkhan, J. C., Vaughn, J., Christy, R. J. & Hamlin, J. H. increased the basal activity of this transcription factor(s), (1986) Biochemistry 25, 6228-6236. which would enable them to circumvent the inhibitory effects 17. Mitchell, P. J., Carothers, A. M., Han, J. H., Harding, J. D., Kas, E., Venolia, L. & Chasin, L. A. (1986) Mol. Cell. Biol. 6, of a range of drugs by increasing the transcription of genes 425-440. whose products are inhibited. 18. Carthew, R. W., Chodosh, L. A. & Sharp, P. A. (1985) Cell43, Our data are consistent with the hypothesis that elevated 439-448. DHFR transcription may be an early response to MTX 19. Graham, F. L. & van der Eb, A. J. (1973) Virology 52,456-467. treatment. Stimulation of DHFR- transcription by MTX 20. Bradford, M. M. (1976) Anal. Biochem. 72, 248-254. would provide a mechanism to increase enzyme levels 21. Neumann, J. R., Morency, C. A. & Russian, K. D. (1987) prior Biotechniques 5, 444-448. to gene amplification. The increased transcription ofthe gene 22. Goldman, I. D., Lichtenstein, N. S. & Oliverio, V. T. (1968) J. could be directly involved in subsequent gene amplification. Biol. Chem. 243, 5007-5017. This hypothesis is supported by the observation that several 23. Sirotnak, F. M. & Dosbach, R. C. (1976) Cancer Res. 36, ofthe agents that increase DHFR mRNA levels also increase 1151-1158. the frequency of gene amplification (refs. 4, 10, 31; J.C.A. 24. Taylor, I. W. & Tattersall, M. H. N. (1981) Cancer Res. 41, and It has been that the 1549-1558. A.G.S., unpublished). reported 25. Blakely, R. L. (1969) Biochemistry ofFolic Acid and Reduced increase in gene amplification in response to hydroxyurea is Pteridines (North-Holland, Amsterdam). prevented by simultaneous addition of cycloheximide, sug- 26. Goodman, L. S. & Gilman, A. (1970) The Pharmacological gesting that a factor that facilitates gene amplification is Basis of Therapeutics (Macmillan, London). stimulated during the treatment (10). Furthermore, transcrip- 27. Deng, T., Li, D., Jehn, C.-H. & Johnson, L. F. (1986) J. Biol. tionally active genes are better templates for recombination Chem. 261, 16000-16005. (32-35) and DNA repair (36) and may be better templates for 28. Thelander, M. & Thelander, L. (1989) EMBO J. 8, 2475-2479. 29. Guilotto, E., Knights, C. & Stark, G. R. (1987) Cell 48, the process of gene amplification. 837-845. 30. Rice, G. C., Ling, V. & Schimke, R. T. (1987) Proc. Natl. H.B.E. and A.G.S. contributed equally to this manuscript. We are Acad. Sci. USA 84, 9261-9264. indebted to Barbara Lipes for preliminary experiments and to 31. Farnham, P. J. & Schimke, R. T. (1986) Mol. Cell. Biol. 6, Michael Wade and Drs. Robert Jambou and Albert Baldwin for 2392-2401. helpful suggestions on experiments and the manuscript. We also 32. Lutkzer, S., Rothman, P., Pollack, R., Coffman, R. & Alt, acknowledge Drs. Philip Sharp and Albert Baldwin for the AdMLP F. W. (1988) Cell 53, 177-184. construct. This work was supported by research grants from the 33. Stewart, S. E. & Roeder, S. (1989) Mol. Cell. Biol. 9, 3464- American Cancer Society (CD-317 and CD-317A,B) and from the 3472. March of Dimes Birth Defects Foundation (1-1158). Additional 34. Thomas, B. J. & Rothstein, R. (1989) Cell 56, 619-630. support was received from the American Cancer Society (JFRA-168, 35. Corcoran, C. M., Corey, S. & Adams, J. M. (1985) Cell 40, J.C.A.) and from the National Institutes of Health [NCI CA09156 71-79. (A.G.S.) and NIEHS ES 07126 granted to the Curriculum in Toxi- 36. Mellon, I., Spivak, G. & Hanawalt, P. C. (1987) Cell 51, cology (H.B.E.)]. 241-249. Downloaded by guest on September 29, 2021