<<

Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

Cell Death and Survival Molecular Research Loss of DNA Glycosylase Selectively Resensitizes p53-Mutant and -Deficient Cells to 5-FdU Yan Yan1, Yulan Qing2, John J. Pink2, and Stanton L. Gerson2

Abstract

Thymidylate synthase (TS) inhibitors including fluoropyri- significantly sensitizes p53 KO cells.Thissensitizationcanalso midines [e.g., 5- (5-FU) and 5-Fluorodeoxyuridine be recapitulated by UDG depletion in cells with p53 KD by (5-FdU, floxuridine)] and (e.g., ) are shRNAs. In addition, sensitization is also observed with peme- widely used against solid tumors. Previously, we reported that trexed in p53 KO cells, but not with 5-FU, most likely due to shRNA-mediated knockdown (KD) of uracil DNA glycosylase RNA incorporation. Importantly, in p53 WT cells, the apoptosis (UDG) sensitized cancer cells to 5-FdU. Because p53 has also pathway induced by 5-FdU is activated independent of UDG been shown as a critical determinant of the sensitivity to TS status. However, in p53 KO cells, apoptosis is compromised in inhibitors, we further interrogated 5-FdU cytotoxicity after UDG-expressing cells, but dramatically elevated in UDG- UDG depletion with regard to p53 status. By analyzing a panel depleted cells. Collectively, these results provide evidence that of human cancer cells with known p53 status, it was deter- loss of UDG catalyzes significant cell death signals only in mined that p53-mutated or -deficient cells are highly resistant cancer cells mutant or deficient in p53. to 5-FdU. UDG depletion resensitizes 5-FdU in p53-mutant and -deficient cells, whereas p53 wild-type (WT) cells are not Implications: This study reveals that UDG depletion restores affected under similar conditions. Utilizing paired HCT116 p53 sensitivity to TS inhibitors and has chemotherapeutic potential WT and p53 knockout (KO) cells, it was shown that loss of p53 in the context of mutant or deficient p53. Mol Cancer Res; 16(2); improves cell survival after 5-FdU, and UDG depletion only 212–21. 2017 AACR.

Introduction Fluoropyrimidines are widely used in the treatment of various types of malignancies for their broad antitumor activity. Once (TS) is a key that catalyzes the taken into cells, fluoropyrimidines can be metabolized into fluor- only means for de novo synthesis of deoxythymidine monopho- odeoxyuridine monophosphate (FdUMP) and fluorodeoxyuri- sphate (dTMP; ref. 1). TS utilizes 5,10-methylenetetrahydrofolate dine triphosphate (FdUTP; refs. 2–5). The metabolite FdUMP (5,10-CH THF) as the methyl-group donor and catalyzes the 2 inhibits TS by forming a stable ternary complex with TS and reductive of monophosphate (dUMP) CH THF (6–8), which ultimately leads to the depletion of dTTP to dTMP (1). dTMP is subsequently phosphorylated to deoxythy- 2 and accumulation of deoxyuridine triphosphate (dUTP). The midine triphosphate (dTTP), a critical precursor for DNA repli- resulting imbalance of deoxynucleotide pools favors the utiliza- cation and repair. As TS contains binding sites for the substrate tion of dUTP and FdUTP during DNA replication and leads to the (dUMP) and the cofactor folate (5,10-CH THF), two 2 accumulationofbothuraciland5-FUinDNA (2–5).Multitargeted structurally different classes of inhibitors, nucleotide, or folate antifolates such as pemetrexed have been approved as compo- analogs block the activity of TS (2). The class of fluoropyrimidines nents of first-line therapy in combination with for the including 5-fluorouracil (5-FU) and floxuridine (5-FdU) target treatment of advanced non–small cell lung cancer (9). Pemetrexed the nucleotide-binding site, whereas the antifolates such as peme- inhibits several folate-dependent ; however, TS is its trexed target the folate-binding site of TS. predominant target (10–13). Administration of pemetrexed leads to a global reduction in nucleotide synthesis as well as accumu- lation of dUTP (14). As a result, dUTP is used in DNA synthesis in place of dTTP, generating uracil misincorporation into DNA (15). 1Department of Pharmacology, Case Western Reserve University, Cleveland, 2 Misincorporated uracil and 5-FU are both primarily recognized Ohio. Case Comprehensive Cancer Center, Division of General Medical and repaired by the uracil DNA glycosylase (UDG)–initiated base Sciences-, Case Western Reserve University, Cleveland, Ohio. excision repair pathway (16). Although incorporation of uracil Note: Supplementary data for this article are available at Molecular Cancer and 5-FU into DNA is well documented as a consequence of Research Online (http://mcr.aacrjournals.org/). exposure to TS inhibitors (15), the impact of the downstream Corresponding Author: Stanton L. Gerson, Case Comprehensive Cancer Center, repair pathway directed by UDG on cell survival is not consistent. Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH 44106. It has been hypothesized that -less futile cycles of uracil Phone: 216-844-8562; Fax: 216-844-4975; E-mail: [email protected] misincorporation, excision by UDG, and further dUTP reinsertion doi: 10.1158/1541-7786.MCR-17-0215 result in DNA strand breaks and cell death (17). If thymine-less 2017 American Association for Cancer Research. cell death was dependent on UDG-mediated removal of uracil

212 Mol Cancer Res; 16(2) February 2018

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

UDG Depletion Resensitizes p53-Mutant Cells to 5-FdU

and 5-FU, one would expect a correlation between the cytotoxicity Table 1. Cell lines and strains used in this work of TS inhibitors and UDG expression. However, the majority of Cell line Origin p53 status studies reported that neither overexpression, nor inhibition, of A375 Melanoma wt UDG affected the sensitivity to TS inhibitors in human, mouse, or LoVo Colon cancer wt – RKO Colon cancer wt chicken DT40 cells (16, 18 23). In contrast, recently both our and A2780 Ovarian cancer wt the Karnitz group observed that loss of UDG highly potentiated H460 Large cell lung cancer wt the cytotoxicity of 5-FdU in several cancer cell lines, indicating H1299 Non–small cell lung cancer null that uracil and 5-FU incorporation played a key role in cell killing OVCAR8 Ovarian cancer del 126-132 (24, 25). DLD1 Colon cancer S241F As the mediators of cell killing due to persistent uracil and 5-FU HEC1A Endometrial cancer R248Q lesions in DNA are not clear, we assessed the likely pathways and noted that one of the major differences in these disparate findings is that cancer cells bearing p53 mutations were used in our and sity, Cleveland, OH). Other cancer cell lines were purchased from Karnitz's experimental system, whereas nontransformed or p53 the American Type Culture Collection. Details of the cell lines (wild-type) WT cancer cells were used in the majorities of others used in this study are listed in Table 1. All cells were maintained in (16, 18–20, 22). Mutation of TP53 is the most frequently observed DMEM (Corning 15-017-CV) supplemented with 10% dialyzed gene alteration in (26). Mutations in p53 have been FBS, 2 mmol/L L-glutamine, 1% MEM NEAA, 100 U/mL penicil- shown to influence cellular response to chemotherapeutic agents lin, and 100 mg/mL streptomycin. Cells were incubated at 37Cin such as cisplatin, , and 5-FU (27, 28). Notably, sub- a humidified atmosphere of 95% air and 5% CO2. 5-FdU and 5- stantial evidence reveals that loss of p53, or p53 mutations, is FU were purchased from Sigma-Aldrich, dissolved respectively in linked to resistance to 5-FU due to inability to activate apoptosis Milli-Q water and DMSO, and stored as a 10 mmol/L stock at pathway. For example, a study using isogenic cell systems dem- 80C. Pemetrexed was purchased from LC laboratories and onstrated that deletion of p53 from a p53 WT colon cancer cell prepared fresh for each experiment by dissolving in Milli-Q water. line (HCT116) rendered cells remarkably resistant to apoptosis induced by 5-FU (29). In addition, 5-FU resistance was also Lentiviral shRNA knockdown described in a variety of p53-mutated cancer cells, including p53 or UDG knockdown (KD) was achieved via shRNA trans- colon, bladder, pancreatic, and gastric cancer (30–33). However, duction. Lentiviral vectors LV-THM-shp53 (which also expresses a few studies have reported on the link of p53 status with the GFP reporter) or LV-Bleo-shp53 to perform p53 KD in WT response to other TS inhibitors such as 5-FdU. HCT116 cells were obtained from Dr. Mark Jackson's laboratory Given the divergent cell models with different p53 status used at Case Western Reserve University, Cleveland, OH (38). Lenti- in our and other studies, the following questions remain unan- viral vector targeting GFP (sh-GFP) was used as control. UDG swered: (1) does loss or mutation of p53 render cells resistant to 5- shRNA vectors (shUDG: NM_003362.2-656s21c1, shUDG-2: FdU, and (2) is the potentiated cytotoxicity of 5-FdU after UDG NM_003362.2-758s21c1, and shUDG-3: NM_003362.2- depletion reliant upon p53 status? To gain insight into these 893s21c1) were purchased from Sigma, and a scramble targeting questions, we tested the impact of UDG depletion on 5-FdU shRNA vector (Sigma) was used as paired control. The lentiviral cytotoxicity in a number of cancer cell lines with differing p53 production and infection were performed as previously described status. We found that, in general, loss or mutation of p53 remark- (24). Cells stably infected with LV-THM-p53 were isolated by cell ably reduced the sensitivity to 5-FdU, and depletion of UDG sorting on the basis of their GFP expression. Cells stably infected selectively resensitized p53-deficient or -mutated cancer cells to 5- with LV-Bleo-p53 were selected with zeocin (Sigma). Selection of FdU. In order to understand the underlying mechanism that positive UDG KD cells was assessed with puromycin (Sigma). contributes to the distinct response after UDG depletion, we utilized paired HCT116 cell lines with, or without, deletion of Clonogenic survival assay the TP53 gene and observed that loss of UDG selectively resensi- As described previously (24), cancer cells (200–300 cells/well) tized HCT116 cells with p53 deletion. This resensitization was were seeded in 6-well culture dishes and allowed to adhere also observed with pemetrexed, but to a lesser extent with 5-FU, overnight. For 5-FdU, cells were treated for 24 hours, then gently which mainly causes damage in RNA (21, 34–37). In the presence washed with PBS once, and incubated with fresh media for at least of WT p53, 5-FdU treatment induced activation of the apoptosis 10 days to allow individual colonies to form. For 5-FU or pathway in both UDG competent, or UDG-depleted cells at pemetrexed, cells were treated continuously for at least 10 days comparable levels. However, in the absence of WT p53, apoptosis to form colonies. After 10 to 18 days, the plates were stained with activation was compromised in UDG-expressing cells and dra- methylene blue. Colonies containing 50 cells were counted. The matically elevated in UDG-depleted cells. Collectively, these percentage of survival was determined relative to untreated con- findings suggest that loss, or mutation, of p53 is associated with trol averaged over three independent experiments. 5-FdU resistance, and UDG depletion can significantly restore sensitivity, indicating that UDG may serve as a therapeutic target Western blots and qPCR to improve the clinical effectiveness of 5-FdU. Western blots were performed as previously described (39). Twenty microgram of protein was loaded on SDS-polyacrylamide Materials and Methods gel. The following antibodies were used to detect proteins on the membrane: a-Tubulin (Calbiochem); GAPDH (Santa Cruz Cell lines and drugs Biotechnology); UDG (FL-313; Santa Cruz Biotechnology); HCT116 p53 knockout (KO) cells were a gift from Dr. Guang- cleaved PARP (Asp214)(19F4; Cell Signaling Technology); bin Luo (Department of Genetics, Case Western Reserve Univer- cleaved caspase 3 (Cell Signaling Technology); p53 (FL-393; Santa

www.aacrjournals.org Mol Cancer Res; 16(2) February 2018 213

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

Yan et al.

Cruz Biotechnology); and p21 (Santa Cruz Biotechnology). For Results qRT-PCR, total RNA from cells was extracted by using the RNeasy p53 mutation or deficiency affords 5-FdU resistance among Plus Mini Kit (Qiagen). cDNA synthesis was performed by using different types of cancer cells the SuperScript III First Strand Kit (Life Technologies). qPCR was Given that p53 mutations or deficiencies are frequently achieved with validated TaqMAN MGB FAMTM dye–labeled observed in cancers, studies have demonstrated that mutations probes (Applied Biosystems) for nuclear UDG on an ABI 7500 of p53 reduce 5-FU cytotoxicity (29–33). To understand Fast Real-time PCR System (Applied Biosystems). b-Actin was whether these mutations also altertheresponseto5-FdU,a used as an endogenous control, and relative gene expression was DD panel of human cancer cell lines from colon, lung, ovarian, calculated as 2 Ct. skin, and endometrium with intrinsically differing p53 status Flow cytometric assay of apoptosis was utilized in this study. The p53 status of each cell line is Cells were seeded in 6-well tissue culture plates (1.5 105 cells/ listed in Table 1. To determine p53 protein functionality in fi well) and allowed to attach overnight. Cells were then treated with p53 WT and p53-mutant (Mut) or -de cient cancer cell lines, 25 nmol/L 5-FdU for 24 hours, washed twice with PBS, and we assessed p53 levels and expression of p21, a widely accept- replenished with drug-free medium at 48, 72, and 96 hours. After ed initiator of p53-activated signaling (40), 24 hours after recovery, the cells floating in the medium were collected. The administration of 8 Gy gamma irradiation. All the p53 WT adherent cells were trypsinized, pelleted, washed in ice-cold PBS, cancer cell lines used in this work induced p21 expression after and resuspended in 1X binding buffer according to the manu- irradiation, indicating functional p53 in these cell lines (Sup- facturer's instructions (FITC Annexin V Apoptosis Detection Kit; plementary Fig. S1). In order to establish the relationship BD Pharmingen). Cells were then stained with FITC–Annexin V between p53 status and 5-FdU sensitivity, we evaluated the and propidium iodide (PI) for 15 minutes at room temperature in cytotoxicity of 5-FdU in these cell lines by clonogenic survival the dark. Annexin V–FITC detects translocation of phosphatidy- assay. As shown in Fig. 1A, the cell lines tested displayed a linositol from the inner to the out of cell membrane during early spectrum of 5-FdU sensitivities with IC50 values ranging from apoptosis, and PI can enter the cells in late apoptosis or necrosis. 1.32 0.33 to 269.55 0.73 nmol/L for A2780 and H1299 Untreated cells were used as control for the double staining. The lines, respectively. Importantly, we observed that, in general, fi cells were analyzed immediately after staining using an Attune cell lines with p53 mutation or de ciency (Fig. 1A, solid lines) fi NXT instrument and FlowJo software. For each measurement, at were signi cantly more resistant to 5-FdU than p53 WT least 20,000 cells were counted. cells (Fig. 1A, dashed lines), with the exception of A375 which has WT p53 but an IC50 of 110.81 1.80 nmol/L. In addition, Statistical analysis except for A375, the IC50 values for the p53 WT cancer lines Statistical significance between two treatment groups was ana- clustered together at a lower dose range (<10 nmol/L), whereas lyzed using unpaired two-tailed Student t test. Significance was p53-mutant or -deficient lines clustered at a higher range assigned for a P value < 0.05. Standard software GraphPad Prism (>100 nmol/L; Fig. 1B). These observations are consistent with and Excel 2013 (Microsoft Corp.) were used for all statistical the hypothesis that p53 mutation or deficiency is associated analysis. with resistance to 5-FdU.

Figure 1. 5-FdU resistance in different types of cancer cells with p53 mutation or deficiency. A, Clonogenic survival assay in cancer cell lines shown in Table 1 in response to increasing doses of 5-FdU. Cell lines with WT p53, dashed lines; cell lines with deficient (or mutant) p53, solid lines. The results represent three independent experiments that were done in duplicate each time.

B, IC50 values of 5-FdU for cancer cell lines listed in Table 1 with WT p53 or deficient (or mutant) p53 status, respectively.

214 Mol Cancer Res; 16(2) February 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

UDG Depletion Resensitizes p53-Mutant Cells to 5-FdU

UDG depletion sensitizes cancer cells with p53 mutation or loss of WT p53 protein expression can alter the response to 5-FdU. deficiency to 5-FdU exposure To address this, we utilized paired HCT116 colon cancer cell lines Previously, the discordant findings on sensitization to 5-FdU with or without genetic TP53 deletion and tested their sensitivity following UDG depletion were reported using cell models with to 5-FdU, and the loss of p53 expression was evaluated by Western differing p53 status (16, 18–25). To understand whether the blot (Fig. 3A). Using a clonogenic survival assay, we demonstrated divergent responses could be attributed to p53 status, we explored that p53 KO cells were more resistant to 5-FdU than p53 WT cells whether UDG depletion could sensitize p53-mutant or -deficient (Fig. 3B). KD of p53 by shRNA recapitulates the resistance cancer cells to 5-FdU differentially. For these experiments, we used observed in p53 KO cells (Fig. 3B), indicating that p53 status is shRNA to deplete UDG in various cancer cell lines with differing a key mediator of the response of HCT116 cells to 5-FdU. p53 status, as listed in Table 1. UDG stable KD was evaluated by To understand whether loss of p53 protein will affect the Western blot (Fig. 2A and B, insert). Based on a clonogenic response to 5-FdU after UDG depletion, we knocked down UDG survival assay, we observed that UDG depletion selectively sen- by shRNA in both HCT116 p53 WT and p53 KO cells. UDG KD sitized cells with p53 mutation or deficiency to 5-FdU exposure levels were shown to be greater than 90% as evaluated by Western (Fig. 2A). However, in p53 WT cell lines, UDG depletion did not blot and qPCR (Fig. 3C and D). In agreement with our data using alter the cytotoxicity of 5-FdU (Fig. 2B). Collectively, these results p53-mutant cells, UDG depletion greatly enhanced cytotoxicity of demonstrate that UDG depletion resensitizes p53-mutant or 5-FdU in p53 KO cells but did not significantly affect p53 WT cells -deficient cancer cells, providing a novel therapeutic target for (Fig. 3E and F), indicating that p53 is involved in regulating the patients with p53-mutant tumors. response to 5-FdU following UDG depletion. To exclude the off- target effect of a single shRNA, we also utilized two other shRNAs 5-FdU resistance in p53 KO or KD cells is reversed by UDG that target UDG in HCT116 p53 WT and p53 KO cells and depletion observed similar effect (Supplementary Fig. S2). In addition, Because many studies have identified gain of various functions depletion of UDG also potentiated 5-FdU cytotoxicity in two for specific p53-mutated proteins (41, 42), we next asked whether HCT116 cancer cells with different shRNAs targeted to p53

Figure 2. UDG depletion selectively sensitizes cells with p53 mutation or deficiency to 5-FdU. Stable cancer cell lines infected with nontargeted scramble control shRNA (shSCR) or UDG-directed shRNA (shUDG) were analyzed by Western blot to examine UDG levels (insert). Clonogenic survival assays of UDG-expressing (shSCR) and UDG-depleted (shUDG) cancer cells with (A) mutant or deficient p53, or (B) WT p53 that are treated with increasing doses of 5-FdU. Values indicate mean values S.E.M. The results represent three independent experiments that were done in duplicate each time (, P < 0.01).

www.aacrjournals.org Mol Cancer Res; 16(2) February 2018 215

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

Yan et al.

Figure 3. 5-FdU resistance due to loss of p53 is reversed by UDG depletion. A, p53 expression levels were analyzed by Western blot in HCT116 cells with wild- type p53 (p53WT), knockout of p53 (p53KO), shGFP-expressing vector (shGFP), and shp53-expressing vector (shp53); , nonspecific bands. B, Clonogenic survival assay for increasing doses of 5-FdU in HCT116 p53WT, p53KO, shGFP-infected, and shp53-infected cells. HCT116 p53WT and p53KO cells stably infected with nontargeted scramble control shRNA (shSCR) or UDG-directed shRNA (shUDG) were analyzed by Western blot (C) and qPCR (D) to examine UDG levels. Clonogenic survival assay for increasing doses of 5-FdU in HCT116 (E) p53WT cells, with shSCR, or with shUDG; and (F) p53KO cells, with shSCR, or with shUDG. Viable colonies (>50 cells) stained with methylene blue after 10 days of culture were counted. The results represent three independent experiments that were done in duplicate each time (, P < 0.01).

(Fig. 4A–E). Collectively, these results confirm that loss of p53 UDG-expressing and UDG-depleted cells in the presence of p53 protein renders cells resistant to 5-FdU, and UDG depletion after pemetrexed or 5-FU treatment (Fig. 5A and B). However, in selectively resensitizes p53 KO and KD cells to 5-FdU. the absence of p53, UDG depletion sensitized cells to pemetrexed (Fig. 5C), whereas loss of UDG only moderately sensitized cells to UDG depletion selectively sensitizes p53 KO cancer cells to 5-FU at high concentrations (Fig. 5D), reaffirming that the pri- pemetrexed and 5-FU mary cytotoxic effect of 5-FU depends on RNA incoporation. Although all TS inhibitors have the ability to block TS, disrupt- Together, these results indicate that UDG depletion also sensitizes ing DNA replication and leading to uracil incorporation into cells without p53 to other TS inhibitors, mainly through gener- DNA, differences among distinct TS inhibitors have been reported ation of DNA damage. in terms of their other metabolism-mediated cytotoxic pathways (2). For example, pemetrexed polyglutamate derivatives also 5-FdU activates cell death in p53 KO cancer cells with depleted demonstrate inhibitory activity for other folate-dependent UDG enzymes such as glycinamide ribonucleotide, but to a lesser extent To understand whether 5-FdU resistance observed in p53 KO (10–13). Moreover, unlike 5-FdU, which mainly exerts its cyto- cells is due to a failure to activate cell death pathways, we toxicity due to effects at the DNA level (24), studies have revealed monitored cell death progression by Annexin V and PI staining. that the cytotoxicity of 5-FU is primarily RNA-mediated, as 5-FU is Cells were exposed to 5-FdU for 24 hours, washed with PBS, metabolized to fluorouridine triphosphate which affects multiple and then allowed to recover in drug-free medium for a total of RNA processes following its incorporation into (21, 34–37). 48, 72, and 96 hours (Fig. 6A). In cells with WT p53, 5-FdU In order to address the question of whether p53 status is respon- caused significant cell death (Annexin V and PI positive) at sible for the differences in sensitivity to other TS inhibitors, 48 hours which was retained at 72 and 96 hours in both including pemetrexed and 5-FU, in UDG-depleted cells, we eval- UDG-expressing and UDG-depleted cells (Fig. 6B and C). uated cell viability following drug exposure in UDG-depleted p53 However, in the absence of p53, cell death caused by 5-FdU WT and p53 KO cancer cells. Similar to our observations with was significantly lower in UDG-expressing cells, whereas in 5-FdU, no significant survival differences were found between UDG-depleted cells, cell death was detected at 24 hours and

216 Mol Cancer Res; 16(2) February 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

UDG Depletion Resensitizes p53-Mutant Cells to 5-FdU

Figure 4. p53 KD resensitizes cancer cells with UDG depletion to 5-FdU. A, HCT116 cells stably infected with shGFP or shp53 (shp53-THM or shp53-Bleo) shRNAs were analyzed by the Western blot to examine p53 KD levels (, nonspecific bands). B, HCT116 cells expressing shGFP or shp53 (shp53-THM or shp53-Bleo) were further infected with nontargeted scramble control shRNA (shSCR) or UDG-directed shRNA (shUDG). UDG mRNA levels were determined by qPCR. Clonogenic survival assay for increasing doses of 5-FdU in (C) shGFP, (D) shp53-THM, and (E) shp53-Bleo–infected HCT116 cells alone, with shSCR, or with shUDG. Viable colonies (>50 cells) stained with methylene blue after 10 days of culture were counted. The results represent three independent experiments that were done in duplicate each time (, P < 0.01). significantly elevated at 48 to 96 hours (Fig. 6B and C). These to loss of p53 is responsible for the enhanced cell survival datasuggestthat5-FdU–induced cell death is dependent upon observed in p53 KO cells. However, in p53 KO cell with coinci- p53, supporting the observation that drug resistance can be dent UDG depletion, 5-FdU selectively activates a p53-indepen- observed as a result of abrogation of the p53-mediated cell dent apoptotic pathway through a mechanism which needs death pathway. Importantly, UDG depletion significantly further investigation. potentiates death of cells lacking WT p53 activity through a p53-independent pathway. Discussion To further elucidate whether the cell death caused by 5-FdU is due to apoptosis, we examined expression of proteins involved in In this study, we utilized multiple cancer cells bearing differing the activation of the apoptotic pathway. In WT p53 cells, we p53 status with or without UDG expression. We observed that loss observed that p53 expression was induced at 24 hours, and the of UDG selectively resensitized cancer cells with p53 mutation or induction remained for 96 hours in both shSCR and shUDG cells deficiency to 5-FdU, but did not alter the response of p53 WT cells. following 5-FdU exposure (Fig. 6D). The expression of cleaved These results demonstrate that UDG, through its function of PARP, a hallmark of apoptotic cell death, was induced at 48 hours removing uracil or 5-FU, plays a major role in the effect of 5-FdU and persisted through 72 and 96 hours in p53 WT cells regardless on the response of cells lacking WT p53 activity. Our findings of whether UDG was present or not (Fig. 6D). In addition, cleaved resolve the unexplained discrepancy observed in a number of caspase 3 was also detected in both UDG-expressing or -depleted prior studies regarding the role of UDG in sensitivity to TS p53 WT cells (Fig. 6D). In the absence of p53, induction of cleaved inhibitors. Prior studies revealed that either loss of UDG PARP or caspase 3 was not readily detected in cells expressing enhanced the cytotoxicity of 5-FdU or pemetrexed in cancer cells UDG after 5-FdU exposure (Fig. 6E), whereas both were robustly (24, 25), or overexpression or inhibition of UDG had no effect induced from 48 to 96 hours in cells depleted of UDG (Fig. 6E). on the sensitivity of human or mouse cells to TS inhibition Taken together, our results suggest that 5-FdU–induced apoptosis (16, 18–20, 22). The difference, we propose, is dependent on is mediated through p53, and the lack of apoptosis activation due p53 status.

www.aacrjournals.org Mol Cancer Res; 16(2) February 2018 217

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

Yan et al.

Figure 5. UDG depletion selectively sensitizes p53 KO cells to pemetrexed and 5-FU. Clonogenic survival assay in HCT116 p53 WT cells (shSCR and shUDG) treated with increasing doses of (A) pemetrexed and (B) 5-FU. Clonogenic survival assay in HCT116 p53 KO cells (shSCR and shUDG) treated with increasing doses of (C) pemetrexed and (D) 5-FU. Viable colonies (>50 cells) stained with methylene blue after 10 days of culture were counted. The results represent three independent experiments that were done in duplicate (, P < 0.01).

p53 plays a key role in determining the sensitivity of cells to Our results demonstrated that inhibition of UDG selectively 5-FU. A number of studies reported that enhanced 5-FU resistance sensitized p53-mutant and -deficient cancer cells to 5-FdU, but has been observed in cells bearing TP53 deletion or mutations did not alter the response in p53 WT cells. Importantly, we have (29–33). However, unlike other TS inhibitors, 5-FU exposure observed that apoptosis following 5-FdU is efficiently induced in caused only slightly potentiated cytotoxicity at higher doses in the presence of p53 but highly compromised in cells lacking p53, UDG-depleted, p53 KO cell lines. Recently, several studies have indicating that the activation of the 5-FdU–induced cell death observed that the cytotoxicity of 5-FU is more dependent on its pathway is dependent on p53. Further studies with different p53 incorporation into RNA than its inhibition of TS, diminishing its WT cell lines also revealed cells highly sensitive to 5-FdU with IC50 effect on DNA (21, 34–37). In addition, activation of p53 fol- values lower than 10 nmol/L. One exception we observed was in lowing 5-FU exposure has been identified as working through the A375 melanoma cells line, which has a WT TP53 gene. A375 RNA mechanisms (43, 44). Because UDG recognizes only DNA was relatively insensitive to 5-FdU and had an IC50 of 110.81 lesions, it is not surprising that depletion of UDG does not 1.80 nmol/L. Clearly, more knowledge is needed regarding the significantly alter cellular responses to agents that primarily affect p53-mediated cell death pathway and how 5-FdU, with or with- RNA function. Together, this suggests that the increased cytotox- out UDG, causes damage and triggers cell death. In response to icity of 5-FdU and pemetrexed observed in UDG-depleted cells is 5-FdU, cells lacking WT p53, combined with UDG depletion, primarily due to uracil and 5-FU incorporation into DNA. activate cell death in a p53-independent manner, which reverses The present results illustrate that cells with p53 mutation or chemoresistance and selectively resensitizes these cancer cells to deficiency are significantly resistant to 5-FdU in comparison with 5-FdU. p53 WT cells. It is clear that many different mutant p53s also The current findings of this article focus on the role of p53 in acquire oncogenic functions that are distinct from the activities of apoptosis and show that in the presence of p53, both UDG WT– WT p53 (41, 42). Some p53 mutants provide enhanced resistance and UDG-depleted cells activate apoptosis at similar levels; how- to apoptosis induced by a variety of treatments, including certain ever, in the absence of p53, apoptosis induction is compromised chemotherapeutic drugs (27, 45). In particular, one study iden- in the UDG WT cells but significantly increased in UDG-depleted tified that p53 mutants activate expression of dUTPase (46), which cells. These data explain that p53 WT cells have 5-FdU IC50 values has been related to the resistance to TS inhibitors (47–49). Our less than 10 nmol/L, whereas p53-mutant or -deficient cells have results on a select group of p53 mutants as well as p53 KO cell lines IC50 values higher than 100 nmol/L. The result is consistent with a revealed resistance to 5-FdU treatment. However, the p53 KO cell previous publication from Janet Houghton's group (30) that cells line is much less resistant to 5-FdU than other p53-mutant cell with WT p53 displayed acute apoptosis, whereas cells with mutant lines, suggesting the potential for enhanced resistance due to p53 showed delayed or compromised apoptosis following fluor- gained functions for certain p53 mutants. opyrimidine treatment. Based on these results, we propose the

218 Mol Cancer Res; 16(2) February 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

UDG Depletion Resensitizes p53-Mutant Cells to 5-FdU

Figure 6. UDG depletion induces cell death caused by 5-FdU in p53 KO cancer cells. A, Schematic diagram of the treatment for HCT116 p53WT (shSCR and shUDG) and p53KO (shSCR and shUDG) cells with 25 nmol/L 5-FdU for 24 hours, washed, and replenished with drug-free medium at indicated time points. B, Untreated (Unt) or treated cells were subjected to FITC– Annexin V and PI staining and analyzed by flow cytometry. Representative flow plots of three independent experiments are shown. C, Cell death is expressed as the percentage of Annexin V-positive cells. Values indicate mean values SD. All experiments were performed independently for 3 times (, P < 0.01). Protein expression involved in regulation of apoptotic cell death in response to 5-FdU was detected in HCT116 (D) p53 WT (shSCR and shUDG) and (E) p53 KO (shSCR and shUDG) cells (, nonspecific bands).

mechanism of cell death that: first, DNA damage due to loss of already been activated in the presence of WT p53 following UDG enhanced the apoptosis in p53-mutant or -deficient cancer 5-FdU–induced stress. Second, loss of UDG increases levels of cells, but did not change the apoptosis in p53 WT cells which has persistent uracil and 5-FU incorporation at the replication forks

www.aacrjournals.org Mol Cancer Res; 16(2) February 2018 219

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

Yan et al.

that without p53 results in early S phase arrest and disintegration Authors' Contributions of the replication fork progression, as we have shown previously Conception and design: Y. Yan, Y. Qing, S.L. Gerson (24). Further support for this mechanism comes from an article Development of methodology: S.L. Gerson published recently by Swati Palit Deb's lab (50), which has shown Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Yan that lack of WT p53 increases DNA origin firing compared with fi Analysis and interpretation of data (e.g., statistical analysis, biostatistics, p53 WT cells. In our system, more DNA origin rings would result computational analysis): Y. Yan, Y. Qing, J.J. Pink, S.L. Gerson in both more 5-FU and uracil incorporation and further disrup- Writing, review, and/or revision of the manuscript: Y. Yan, Y. Qing, J.J. Pink, tion of these replication forks that improve the killing effect in S.L. Gerson p53-mutant or -deficient cancer cells. Taken together, these results Study supervision: S.L. Gerson provide an explanation for the discordant findings in previous published data regarding the role of UDG in mediating the The costs of publication of this article were defrayed in part by the cytotoxicity of TS inhibitors and suggest that UDG is an attractive payment of page charges. This article must therefore be hereby marked therapeutic target in cancer cells with p53 mutation or deficiency, advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate to enhance their response to TS inhibitors. this fact.

Disclosure of Potential Conflicts of Interest Received April 21, 2017; revised August 2, 2017; accepted October 26, 2017; No potential conflicts of interest were disclosed. published OnlineFirst November 8, 2017.

References 1. Carreras CW, Santi DV. The catalytic mechanism and structure of thymi- 18. Welsh SJ, Hobbs S, Aherne GW. Expression of uracil DNA glycosylase dylate synthase. Annu Rev Biochem 1995;64:721–62. (UDG) does not affect cellular sensitivity to thymidylate synthase (TS) 2. Wilson PM, Danenberg PV, Johnston PG, Lenz HJ, Ladner RD. Standing the inhibition. Eur J Cancer 2003;39:378–87. test of time: targeting thymidylate biosynthesis in cancer therapy. Nat Rev 19. Andersen S, Heine T, Sneve R, Konig€ I, Krokan HE, Epe B, et al. Incorpo- Clin Oncol 2014;11:282–98. ration of dUMP into DNA is a major source of spontaneous DNA damage, 3. Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: mechanisms of while excision of uracil is not required for cytotoxicity of fluoropyrimidines action and clinical strategies. Nat Rev Cancer 2003;3:330–8. in mouse embryonic fibroblasts. Carcinogenesis 2005;26:547–55. 4. Malet-Martino M, Martino R. Clinical studies of three oral prodrugs of 5- 20. Luo Y, Walla M, Wyatt MD. Uracil incorporation into genomic DNA does fluorouracil (, UFT, S-1): a review. Oncologist 2002;7:288– not predict toxicity caused by chemotherapeutic inhibition of thymidylate 323. synthase. DNA Repair 2008;7:162–9. 5. Wyatt MD, Wilson DM 3rd. Participation of DNA repair in the response to 21. Pettersen HS, Visnes T, Vagbø CB, Svaasand EK, Doseth B, Slupphaug G, 5-fluorouracil. Cell Mol Life Sci 2009;66:788–99. et al. UNG-initiated base excision repair is the major repair route for 5- 6. Santi DV, McHenry CS. 5-Fluoro-20-deoxyuridylate: covalent complex with fluorouracil in DNA, but 5-fluorouracil cytotoxicity depends mainly on thymidylate synthetase. Proc Natl Acad Sci 1972;69:1855–7. RNA incorporation. Nucleic Acids Res 2011;39:8430–44. 7. Matthews DA, Appelt K, Oatley SJ, Xuong NH. Crystal structure of Escher- 22. Kemmerich K, Dingler FA, Rada C, Neuberger MS. Germline ablation ichia coli thymidylate synthase containing bound 5-fluoro-20-deoxyuridy- of SMUG1 DNA glycosylase causes loss of 5-hydroxymethyluracil- late and 10-propargyl-5, 8-dideazafolate. J Mol Biol 1990;214:923–36. and UNG-backup uracil-excision activities and increases cancer 8. Kaiyawet N, Rungrotmongkol T, Hannongbua S. Effect of halogen sub- predisposition of Ung/ Msh2/ mice. Nucleic Acids Res 2012;40: stitutions on dUMP to stability of thymidylate synthase/dUMP/mTHF 6016–25. ternary complex using molecular dynamics simulation. J Chem Inf Model 23. Nagaria P, Svilar D, Brown AR, Wang XH, Sobol RW, Wyatt MD. SMUG1 2013;53:1315–23. but not UNG DNA glycosylase contributes to the cellular response to 9. Scagliotti GV, Parikh P, Von Pawel J, Biesma B, Vansteenkiste J, Manegold recovery from 5-fluorouracil induced replication stress. Mutat Res 2013; C, et al. Phase III study comparing cisplatin plus with cisplatin 743:26–32. plus pemetrexed in -naive patients with advanced-stage 24. Yan Y, Han X, Qing Y, Condie AG, Gorityala S, Yang S, et al. Inhibition of non–small-cell lung cancer. J Clin Oncol 2008;26:3543–51. uracil DNA glycosylase sensitizes cancer cells to 5-fluorodeoxyuridine 10. Grindey GB, Shih C, Barnett CJ, Pearce HL, Engelhardt JA, Todd GC, et al. through replication fork collapse-induced DNA damage. Oncotarget LY231514, a novel pyrrolopyrimidine that inhibits thymidylate 2016;7:59299–313. synthase (TS). Proc Am Assoc Cancer Res 1992;33:411 25. Huehls AM, Huntoon CJ, Joshi PM, Baehr CA, Wagner JM, Wang X, et al. 11. Shih C, Chen VJ, Gossett LS, Gates SB, MacKellar WC, Habeck LL, et al. Genomically incorporated 5-fluorouracil that escapes UNG-initiated base LY231514, a pyrrolo [2, 3-d] -based antifolate that inhibits excision repair blocks DNA replication and activates homologous recom- multiple folate-requiring enzymes. Cancer Res 1997;57:1116–23. bination. Mol Pharmacol 2016;89:53–62. 12. Hanauske AR, Chen V, Paoletti P, Niyikiza C. Pemetrexed disodium: a 26. Olivier M, Hollstein M, Hainaut P. TP53 mutations in human cancers: novel antifolate clinically active against multiple solid tumors. Oncologist origins, consequences, and clinical use. Cold Spring Harb Perspect Biol 2001;6:363–73. 2010;2:a001008. 13. Goldman ID, Zhao R. Molecular, biochemical, and cellular pharmacology 27. Freed-Pastor WA, Prives C. Mutant p53: one name, many proteins. Genes of pemetrexed. Semin Oncol 2002;29:(6 Suppl 18)3–17. Dev 2012;26:1268–86. 14. Van Triest B, Pinedo HM, Giaccone G, Peters GJ. Downstream molecular 28. Tchelebi L, Ashamalla H, Graves PR. Mutant p53 and the response to determinants of response to 5-fluorouracil and antifolate thymidylate chemotherapy and radiation. Subcell Biochem 2014;85:133–59. synthase inhibitors. Ann Oncol 2000;11:385–91. 29. Bunz F, Hwang PM, Torrance C, Waldman T, Zhang Y, Dillehay L, et al. 15. Berger SH, Pittman DL, Wyatt MD. Uracil in DNA: consequences for Disruption of p53 in human cancer cells alters the responses to therapeutic carcinogenesis and chemotherapy. Biochem Pharmacol 2008;76:697–706. agents. J Clin Invest 1999;104:263–9. 16. Grogan BC, Parker JB, Guminski AF, Stivers JT. Effect of the thymidylate 30. Petak I, Tillman DM, Houghton JA. p53 dependence of Fas induction and synthase inhibitors on dUTP and TTP pool levels and the activities of DNA acute apoptosis in response to 5-fluorouracil-leucovorin in human colon repair glycosylases on uracil and 5-fluorouracil in DNA. Biochemistry carcinoma cell lines. Clin Cancer Res 2000;6:4432–41. 2011;50:618–27. 31. Stravopodis DJ, Karkoulis PK, Konstantakou EG, Melachroinou S, 17. Ladner RD. The role of dUTPase and uracil-DNA repair in cancer chemo- Thanasopoulou A, Aravantinos G, et al. Thymidylate synthase inhibition therapy. Curr Protein Pept Sci 2001;2:361–70. induces p53-dependent and p53-independent apoptotic responses in

220 Mol Cancer Res; 16(2) February 2018 Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

UDG Depletion Resensitizes p53-Mutant Cells to 5-FdU

human urinary bladder cancer cells. J Cancer Res Clin Oncol 2011; 40. Abbas T, Dutta A. p21 in cancer: intricate networks and multiple activities. 137:359–74. Nat Rev Cancer 2009;9:400–14. 32. Eisold S, Linnebacher M, Ryschich E, Antolovic D, Hinz U, Klar E, et al. The 41. Muller PA, Vousden KH. p53 mutations in cancer. Nat Cell Biol 2013; effect of adenovirus expressing wild-type p53 on 5-fluorouracil chemo- 15:2–8. sensitivity is related to p53 status in pancreatic cancer cell lines. World J 42. Aschauer L, Muller PA. Novel targets and interaction partners of mutant Gastroenterol 2004;10:3583–9. p53 gain-of-function. Biochem Soc Trans 2016;44:460–6. 33. Osaki M, Tatebe S, Goto A, Hayashi H, Oshimura M, Ito H. 5-Fluorouracil 43. Sun XX, Dai MS, Lu H. 5-fluorouracil activation of p53 involves an MDM2- (5-FU) induced apoptosis in gastric cancer cell lines: role of the p53 gene. ribosomal protein interaction. J Biol Chem 2007;282:8052–9. Apoptosis 1997;2:221–6. 44. Akpinar B, Bracht EV, Reijnders D, Safarikova B, Jelinkova I, Grandien A, 34. Geoffroy FJ, Allegra CJ, Sinha B, Grem JL. Enhanced cytotoxicity with et al. 5-Fluorouracil-induced RNA stress engages a TRAIL-DISC-dependent interleukin-1 alpha and 5-fluorouracil in HCT116 colon cancer cells. Oncol apoptosis axis facilitated by p53. Oncotarget 2015;6:43679. Res 1993;6:581–91. 45. Oren M, Rotter V. Mutant p53 gain-of-function in cancer. Cold Spring Harb 35. Pritchard DM, Watson AJ, Potten CS, Jackman AL, Hickman JA. Inhibition Perspect Biol 2010;2:a001107. by uridine but not of p53-dependent intestinal apoptosis 46. Pugacheva EN, Ivanov AV, Kravchenko JE, Kopnin BP, Levine AJ, initiated by 5-fluorouracil: evidence for the involvement of RNA pertur- Chumakov PM. Novel gain of function activity of p53 mutants: activation bation. Proc Natl Acad Sci USA 1997;94:1795–9. of the dUTPase gene expression leading to resistance to 5-fluorouracil. 36. Brody JR, Hucl T, Costantino CL, Eshleman JR, Gallmeier E, Zhu H, et al. Oncogene 2002;21:4595. Limits to thymidylate synthase and TP53 genes as predictive determinants 47. Parsels LA, Parsels JD, Wagner LM, Loney TL, Radany EH, Maybaum J. for fluoropyrimidine sensitivity and further evidence for RNA-based tox- Mechanism and pharmacological specificity of dUTPase-mediated protec- icity as a major influence. Cancer Res 2009;69:984–91. tion from DNA damage and cytotoxicity in human tumor cells. Cancer 37. Huehls AM, Wagner JM, Huntoon CJ, Geng L, Erlichman C, Patel AG, et al. Chemother Pharmacol 1998;42:357–62. Poly (ADP-Ribose) polymerase inhibition synergizes with 5-fluorodeox- 48. Koehler SE, Ladner RD. Small interfering RNA-mediated suppression of yuridine but not 5-fluorouracil in ovarian cancer cells. Cancer Res 2011; dUTPase sensitizes cancer cell lines to thymidylate synthase inhibition. 71:4944–54. Mol Pharmacol 2004;66:620–6. 38. Cipriano R, Patton JT, Mayo LD, Jackson MW. Inactivation of p53 signaling 49. Wilson PM, LaBonte MJ, Lenz HJ, Mack PC, Ladner RD. Inhibition of by p73 or PTEN ablation results in a transformed phenotype that remains dUTPase induces synthetic lethality with thymidylate synthase–targeted susceptible to Nutlin-3 mediated apoptosis. 2010;9:1373–9. therapies in non–small cell lung cancer. Mol Cancer Ther 2012;11: 39. Yan L, Bulgar A, Miao Y, Mahajan V, Donze JR, Gerson SL, et al. Combined 616–28. treatment with and methoxyamine: blocking apurininc/ 50. Singh S, Vaughan CA, Frum RA, Grossman SR, Deb S, Palit Deb S. Mutant pyrimidinic site repair coupled with targeting topoisomerase IIa. Clin p53 establishes targetable tumor dependency by promoting unscheduled Cancer Res 2007;13:1532–9. replication. J Clin Invest 2017;127:1839–55.

www.aacrjournals.org Mol Cancer Res; 16(2) February 2018 221

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 8, 2017; DOI: 10.1158/1541-7786.MCR-17-0215

Loss of Uracil DNA Glycosylase Selectively Resensitizes p53-Mutant and -Deficient Cells to 5-FdU

Yan Yan, Yulan Qing, John J. Pink, et al.

Mol Cancer Res 2018;16:212-221. Published OnlineFirst November 8, 2017.

Updated version Access the most recent version of this article at: doi:10.1158/1541-7786.MCR-17-0215

Supplementary Access the most recent supplemental material at: Material http://mcr.aacrjournals.org/content/suppl/2017/11/08/1541-7786.MCR-17-0215.DC1

Cited articles This article cites 50 articles, 17 of which you can access for free at: http://mcr.aacrjournals.org/content/16/2/212.full#ref-list-1

Citing articles This article has been cited by 3 HighWire-hosted articles. Access the articles at: http://mcr.aacrjournals.org/content/16/2/212.full#related-urls

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://mcr.aacrjournals.org/content/16/2/212. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from mcr.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research.