Literature
Home医源资料库在线期刊分子药理学杂志2005年第67卷第5期

Modulation of Cellular Response to Cisplatin by a Novel Inhibitor of DNA Polymerase

来源:分子药理学杂志
摘要:CellularexperimentsshowedthattreatmentbyMAinthepresenceofcisplatinsensitizedthePol-overexpressingcisplatin-resistant2008C135。Cellularproliferationwasexpressedastheratioofthecellnumberintheuntreatedwellsversusthatinthetreatedwells。EffectoftheThreeInhibi......

点击显示 收起

    Equipe Instabilitee Geeneetique et Cancer, Institut de Pharmacologie et de Biologie Structurale, Unitee Mixte Recherche Centre National de la Recherche Scientifique 5089 (F.B., P.B., Y.C., J.P.C., A.B., J.S.H., C.C.), Centre de Recherche en Pharmacologie-Santee, Unitee Mixte Recherche Centre National de la Recherche Scientifique/P. Fabre 2587 (M.K.), Centre de Criblage Pharmacologique, Unitee Mixte Recherche Centre National de la Recherche Scientifique/P. Fabre 2646 (F.A.)
    Chimie des Substances Naturelles Bioactives, Unitee Mixte Recherche Centre National de la Recherche Scientifique/P. Fabre 2597 (C.L., B.D., G.M.), Institut de Sciences et Technologies du Meedicament de Toulouse 3, Toulouse, France
    Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich, Switzerland (E.F.-H., U.H.)

    Abstract

    DNA polymerase  (Pol ) is an error-prone enzyme whose up-regulation has been shown to be a genetic instability enhancer as well as a contributor to cisplatin resistance in tumor cells. In this work, we describe the isolation of new Pol  inhibitors after high throughput screening of 8448 semipurified natural extracts. In vitro, the selected molecules affect specifically Pol -mediated DNA synthesis compared with replicative extracts from cell nuclei. One of them, masticadienonic acid (MA), is particularly attractive because it perturbs neither the activity of the purified replicative Pol  nor that of nuclear HeLa cell extracts. With an IC50 value of 8 e, MA is the most potent of the Pol  inhibitors found so far. Docking simulation revealed that this molecule could substitute for single-strand DNA in the binding site of Pol  by binding Lys35, Lys68, and Lys60, which are the main residues involved in the interaction Pol /single-strand DNA. Selected inhibitors also affect the Pol -mediated translesion synthesis (TLS) across cisplatin adducts; MA was still the most efficient. Therefore, masticadienonic acid sensitized the cisplatin-resistant 2008C135.25 human tumor cells. Our data suggest that molecules such as masticadienonic acid could be suitable in conjunction with cisplatin to enhance anticancer treatments.

    DNA polymerase  (Pol ) is the smallest human DNA polymerase with a molecular mass of 39 kDa. It contains two domains: first, an N-terminal fragment (8 kDa) that possesses the binding affinity for single-strand and double-strand DNA, 5'-phosphate recognition in gapped DNA, and dRP lyase activity; and second, a C-terminal domain (31 kDa) that contains the catalytic activity. In somatic cells, Pol  is an essential enzyme of the base excision repair (BER) and single-strand break repair pathways (Hubscher et al., 2002). We also demonstrated (Canitrot et al., 1998; Bergoglio et al., 2001) that Pol  overexpression allows cells to tolerate bifunctional DNA damage generated, for example, by cisplatin, which is used in the treatment of many cancers (Pil and Lippard, 1997). Indeed, Pol  facilitates the error-prone translesion replication of these adducts that otherwise would block the replicative machinery and kill the cells (Hoffmann et al., 1995; Canitrot et al., 1998). A large number of mechanisms can contribute to cisplatin resistance, such as decreased drug uptake, enhanced repair of the adducts, or increased levels of detoxifying agents like glutathione (Andrews and Howell, 1990). The resistant phenotype of 2008C135.25 cells used in this study, generated from 2008 human ovarian carcinomas by in vitro selection with cisplatin (Andrews et al., 1985), has been attributed to impaired uptake, decreased DNA platination (Jekunen et al., 1994), and translesion synthesis across cisplatin adducts. Such replicative bypass has been measured either indirectly by quantifying radiolabeled nascent DNA (Mamenta et al., 1994) or more directly by showing that extracts from these cells displayed an enhanced Pol -mediated capacity to replicate a DNA oligonucleotide carrying a cisplatin adduct (Bergoglio et al., 2001). Moreover, down-regulation of pol  by small inducible RNA clearly sensitized cisplatin-resistant HeLa and SKOV-3 cancer cells to cisplatin (M. Albertella, KuDOS Pharmaceuticals, Cambridge, England, UK, unpublished data; communicated in Jacques Monod Conference, Roscoff, France, 2004).

    In addition to its contribution to the mutagenic translesion process, up-regulated Pol  induces an overall untargeted genetic instability by competing with replicative DNA polymerases in DNA transactions where normally it is not involved (Canitrot et al., 2000; Servant et al., 2002a). It thus contributes to the emergence of variant clones with a proliferating phenotype (Canitrot et al., 1998, 1999; Bergoglio et al., 2002, 2003; Servant et al., 2002a). In accordance with these data, inhibition of Pol -mediated DNA synthesis by using dideoxycytidine, which is efficiently incorporated into DNA by this enzyme (Copeland et al., 1992), increased the survival of mice inoculated previously with Pol -overexpressing melanomas (Louat et al., 2001).

    Pol  shares infidelity and translesion synthesis capacity with the newly discovered error-prone DNA polymerases such as Pol , , , , , e? and  (Radman, 1999), which seem also devoted in vivo to help cells to tolerate DNA damage. Among those "adaptive" DNA polymerases, Pol , whose primary sequence is very close to that of Pol , also possesses an intrinsic 5'-deoxyribose-5'-phosphate lyase function (Garcia-Diaz et al., 2001) and a distributive DNA synthesis activity (Garcia-Diaz et al., 2002). Moreover, Pol  can substitute in vitro Pol  in BER, suggesting that it also participates in this pathway (Garcia-Diaz et al., 2001). In this work, we sought natural compounds that can selectively inhibit Pol  as 1) tools to investigate its specific role in vivo by distinguishing it from other DNA polymerases such as replicative or -like DNA polymerase; and 2) new potential drugs that are likely to reverse the Pol -associated tumor resistance to cisplatin. To our knowledge, only two Pol  inhibitors have been considered to date as specific [i.e., prunasin (IC50 of 93 e), isolated from a red Perila and a mugwort (Mizushina et al., 1999), and solanapyrone A (IC50 of 30 e), a plant phytotoxin (Mizushina et al., 2002)]. After screening 8448 semipurified extracts from plants and marine organisms, we purified from Juniperus communis, Pistacia lentiscus, and Mahurea palustris the three inhibitors trans-communic acid (CA), masticadienonic acid (MA), and the novel mahureone A (MH), respectively. MA was the most effective inhibitor (IC50 of 8 e) identified to date. Moreover, it only poorly altered the action of either the replicative Pol  or that of HeLa cell nuclear extracts. In addition, pol -deficient mouse embryonic fibroblasts (MEF) were not sensitive to the action of MA. In contrast, MA alters the response to cisplatin of isogenic wild-type MEF cells, conferring to these cells a pol eC/eC phenotype. Finally, MA inhibited the Pol -mediated translesion DNA synthesis across a cisplatin lesion. We showed by docking that MA could compete with the single-strand DNA (ssDNA) for binding into the ssDNA binding site of Pol .MA potentially binds three lysine residues (Lys35, Lys60, and Lys68) involved in the ssDNA binding activity of the aminoterminal 8-kDa domain (Prasad et al., 1998). Cellular experiments showed that treatment by MA in the presence of cisplatin sensitized the Pol -overexpressing cisplatin-resistant 2008C135.25 human tumor cells, whereas it does not alter the response to cisplatin of pol  minus MEF cells. In summary, we have isolated a molecule that is likely to target the Pol -directed transactions and thus reduce tumoral cisplatin resistance.

    Materials and Methods

    Plant Extracts and Compounds. Extracts from either the stems of J. communis and P. lentiscus or the leaves of M. palustris were prepared by overnight maceration in ethyl acetate (1:10 w/v), filtration, and evaporation. These extracts (50 mg) were first coarsely fractionated on 1.5 mg (6 ml) solid-phase epitaxial SiO2 cartridges in an Upti-clean column (Interchim, Montluon, France) using hexane and a gradient of methanol in chloroform as eluents. Aliquots were dissolved in 100% DMSO and diluted 1:1200 in the enzyme assay. Then, 50 g of the plant material was submitted to a bioguided fractionation by combining chromatographies (normal and reverse C18 phases) to lead to 50 mg of CA, 60 mg of MA, and 400 mg of MH, respectively, as pure compounds. These products were dissolved in appropriated buffer to achieve 10eC2 to 10eC7 M solutions.

    Cells and Cell Extracts. The pol  null and the corresponding wild-type MEFs were purchased from the American Type Culture Collection (Manassas, VA) as described previously (Sobol et al., 1996). They were cultivated in DMEM. Human 2008 and 2008C135.25 cells were grown in RPMI 1640 medium as described previously (Bergoglio et al., 2001). HeLa cells (American Type Culture Collection) were grown as spinner cultures in the same medium. Replicative extracts were prepared from either HeLa or MEF cells as described previously (Roberts et al., 1993) after harvesting cells in the upper part of their log-phase growth.

    Enzymes. Rat Pol  for the high-throughput screening (HTS) was purified as described previously (Skandalis and Loeb, 2001). The 8-kDa domain of human Pol  (residues 1eC88) was cloned into pIVEX2.4d (Roche Diagnostics, Indianapolis, IN) to obtain the p1968 plasmid, which was sequenced and introduced into BL21(DE3) bacteria before purification as described previously (Skandalis and Loeb, 2001). Purification of Pol  was carried out by GTP Technology (Toulouse, France). Human Pol  for replication assays was purchased from Trevigen (Gaithersburg, MD). Pol  was purchased from CHIMERx (Milwaukee, WI). The recombinant four subunits of Pol  were isolated from infected insect baculovirus Sf9 cells as described previously (Podust et al., 2002). Full-length recombinant single-subunit Pol  was purified according to procedures described by Ramadan et al. (2003). Purification of the human immunodeficiency virus reverse transcriptase was done as published previously (Preston et al., 1988).

    Microassay for HTS. For HTS, the DNA polymerase activity was determined as the amount of fluorescein-12-dCTP incorporated into a 60-mer biotinylated oligonucleotide template hybridized to a 5' 17-mer synthetic primer. This substrate was immobilized in a streptavidin-coated combiplate C8 (ThermoLabsystem, Franklin, MA). The standard reaction mixture (100 e蘬) contained 25 mM HEPES, pH 8.5, 5 mM MgCl2, 125 mM NaCl, 25 pmol biotinylated hybridized oligonucleotide, and 5 e蘥 of rat Pol  in the presence of extracts or compounds. The reaction was started with the simultaneous addition of 10 e dNTP and 1 e fluorescein-12-dCTP. Incubation was for 150 min at 37°C, and the products were washed three times with 200 e蘬 of 25 mM HEPES, pH 8.5, 5 mM MgCl2, 125 mM NaCl, and 0.05% (v/v) Tween 20. The fluorescence was measured in a Fluostar fluorimeter (BMG Labtechnologies Inc., Durham, NC). The HTS experiments were run on a Beckman Sagian system (Beckman Coulter, Fullerton, CA). Plate-handling was performed with the Optimized Robot for Chemical Analysis robotic arm (Beckman Coulter). Each positive fraction was manually controlled with the same protocol.

    DNA Replication Assay. Cell-extract preparation and replication conditions for these extracts were described previously (Servant et al., 2002a). All purified DNA polymerases were tested by using specific conditions for their optimal activity. One unit of DNA polymerase corresponds to 1 pmol of dNTP incorporated into acid-insoluble materials at 37°C in 60 min by using as a substrate an activated calf thymus DNA preincubated with DNase I. Calf thymus DNA preincubated with DNase I (activated DNA; Sigma-Aldrich, St. Louis, MO) was used as a template in a final volume of 20 e蘬 at 37°C for various incubation times in the presence of inhibitors. Human Pol  (0.5 units) was added to a reaction buffer containing 25 mM HEPES KOH, pH 8.5, 10 mM MgCl2, 25 mM NaCl, 1 mM DTT, 100 e dATP, 100 e dGTP, 100 e dCTP, 0.2 e藽i dTTP, and 1 e蘥 of template. Wild-type human Pol  (0.5 unit) was used in 50 mM Tris, pH 8.5, 50 mM NaCl, 1 mM DTT, 20 e蘥/ml bovine serum albumin, 0.75 mM MnCl2, 5 e dATP, 5 e dGTP, 5 e dCTP, 0.2 e藽i [3H]dTTP, and 1 e蘥 of template. The assay conditions with human Pol  (0.05 units), Pol  (1.5 unit), Pol  (1 units), or human immunodeficiency virus reverse transcriptase (1 unit) were 50 mM Tris, pH 6.5, 1 mM DTT, 0.25 mg/ml bovine serum albumin, 6 mM MgCl2, 4.6 e蘥/ml proliferating cell nuclear antigen (if Pol ), 100 e dATP, 100 e dGTP, 100 e dCTP, 0.2 e藽i [3H]dTTP, and 1 e蘥 of template. The amounts of DNA polymerases were chosen to obtain comparable nucleotide incorporation without inhibitor. After incubation, reaction was stopped with 10 mM EDTA, and the radioactive DNA products were collected on GF/C glass fiber filters (Whatman, Clifton, NJ). Filters were washed twice with 0.5% trichloroacetic acid solution and then used for determination of radioactivity. IC50 value was calculated by using GraphPad Prism software (GraphPad Software Inc., San Diego, CA).

    Docking of Masticadienonic Acid on Human DNA Polymerase . Molecular modeling was performed by using SYBYL 6.9 (Tripos, St Louis, MO) by running on a Silicon Graphics Octane 2 workstation. Docking calculations were performed using FlexX, version 1.11 (Tripos). The FlexX scoring function was used during the complex construction phase. DrugScore as implemented in FlexX reranked the obtained solutions. Thirty docking solutions were generated. These solutions were checked to find the solution with the best score that matched the original pharmacophore hypothesis. Figures were generated using Grasp (Nicholls et al., 1991) or Pymol (http://www.pymol.org).

    Translesion Synthesis Assay. The unmodified or cisplatin-modified 60-mer template were prepared as described previously (Hoffmann et al., 1995) and hybridized to a 5'-32P-labeled 17-mer primer. Translesion reactions were carried out as described previously (Bergoglio et al., 2001). At the end of the reaction, the samples were denatured for 10 min at 70°C and loaded onto a 15% polyacrylamide/7 M urea/30% formamide gel.

    Proliferation and Clonogeny Assays. For growth-rate analysis, the control MEF and MEF pol eC/eC cells were seeded at 300 and 600 cells/well in six-well dishes, respectively. Cells were allowed to attach overnight at 34°C in a minimal essential medium supplemented with glutamine/10% fetal calf serum/penicillin/streptomycin and treated with cisplatin (Sigma-Aldrich) for 1 h at 37°C and/or masticadienonic acid at various concentrations during 6 days. Cells were then trypsinized and counted. Cellular proliferation was expressed as the ratio of the cell number in the untreated wells versus that in the treated wells.

    Cytotoxicity for the 2008 and 2008C135.25 cells was determined by a clonogenic assay as described previously (Bergoglio et al., 2001). For resistance-reversion assays, cells were treated with inhibitors at DL20 all experiment long, with cisplatin (Sigma-Aldrich) being administrated for 1 h at 37°C 24 h after plating.

    Results

    HTS of Partially Purified Plant Extracts. An automated assay was designed to identify Pol  inhibitors by selecting compounds that decreased the incorporation of fluorescent nucleotides into a DNA template. The ddCTP chain terminator, which inhibits DNA synthesis mediated by Pol  (Bouayadi et al., 1997), was used as a reference (Fig. 1A). The "z' factor" determination proposed by Zhang et al. (1999) is an indicator of the performance of the assay system. In the present case, we measured a z' factor average value of 0.55. During the run, the interplate CV of the DMSO control signal value averaged 7.5% (data not shown). This simple assay enabled the screening of 8448 partially purified natural extract fractions, from which 71 active extracts were identified. Subsequent 32P-labeled primer extension procedure was carried out with Pol  and replicative Chinese hamster ovary cell extracts to select specific inhibitory extracts. Three of them were selected (Fig. 1A): J. communis (EX1), P. lentiscus (EX2), and M. palustris (EX3), from which we isolated and purified the corresponding active molecules trans-communic acid, masticadienonic acid, and mahureone A, respectively.

    Structural Characterization of the Three Inhibitors. The compound isolated from J. communis (Cupressaceae) was found to be a ubiquitous diterpene of the labdane series, CA. Its structure (Fig. 1B) was established by comparison of its spectral data (1 and 2D NMR and mass spectrometry) with that from an authentic sample (Arya et al., 1961). A second molecule was isolated in a straightforward manner from P. lentiscus (Anarcardiaceae), a small tree growing in the Mediterranean basin and the exudate of which, known as mastic, is used as a chewing gum. The active compound was found to be MA (3-oxotirucalla-7,24-dien-26-oic acid), for which the structure (Fig. 1C) was known (Barton and Soane, 1956). It is noteworthy that the isomer isomasticadienonic acid, also present in the extract, which only differs by the position of the double bond in ring B (7 versus 8), is totally inactive in the test (data not shown). The structure is also very close to another Pol  inhibitor isolated by Hecht et al., which is characterized by a rare E double bond in the chain (Deng et al., 2000). The Z configuration of the 24 double bond in the mastic compound was ascertained by the NMR chemical shift of H-24 (6.10 versus 6.89 ppm in the E compound). The third product was isolated from a plant from Guyana, M. palustris, a species belonging to the Guttiferae family and which has so far not been the object of chemical investigations. Mass spectrometry indicated that this molecule, for which we propose the trivial name of mahureone A (MH), had a C35H46O6 composition (exact mass analysis), and the molecular ion was characterized by its decomposition with losses of 69 (isoprene chain) and 128 mass units. UV was not very informative and simply indicated the presence of a phenyl ring (max = 283 nm), whereas infrared displayed vibrations for a complex carbonyl system. Literature search showed that a compound, laxifloranone (Fig. 2B), also isolated from a Guttiferae, Marila laxiflora, with similar data (Bokesch et al., 1999), but direct comparison of the compounds on high-performance liquid chromatography proved that they were different. The 1H and 13C NMR spectra and full analysis of 2D NMR experiments led to the proposed structure (Fig. 1D), salient feature of which were the central six-member ring that contained a rare  triketone (enolised form) and no less than eight contiguous quaternary carbon atoms. The validity of the structure was further supported by the isolation of four other compounds from the same series (data not shown). The differences between mahureone A and laxifloranone may be explained by a ring opening in basic medium followed by ring closure of an intermediate enol in a series of events compatible with the rule of the biosynthesis. They most probably belong to the same optical series.

    Effect of CA, MA, and MH on DNA Polymerase , Other DNA Polymerases, and Replicative Nuclear Extracts. To assess the efficiency of the three inhibitors on Pol , we carried out an in vitro replication assay by using an activated gapped genomic DNA. DNA synthesis signals obtained with Pol  were compared with that of nuclear extracts from HeLa cells, which contain all the replicative DNA polymerases able to support bidirectional and semiconservative genomic DNA replication, and with the purified replicative Pol  and the error-prone Pol  and Pol . As shown in Fig. 2, A to C, all three molecules inhibited Pol  with IC50 values for CA, MA, and MH of 92, 8, and 118 e, respectively. Nuclear extracts from HeLa cells were much more resistant to inhibition by those molecules, with IC50 values of 2, 3, and 2 mM, respectively. Pol  activity was also resistant to MA, with an IC50 value of 60 mM, whereas CA and MH inhibited Pol . Furthermore, MH altered the activity of Pol  and Pol , with these enzymes being less affected than Pol  by the two other inhibitors CA and MA (Fig. 2, AeCC). We also tested the inhibitory action of MA on the other replicative enzyme Pol , involved in the synthesis of RNA/DNA primers, and found that this enzyme is less affected (IC50 = 93 e) than Pol  (Fig. 2B). Taken together, these data indicate that MA is an attractive Pol  inhibitor.

    Effect of MA on Pol  Null Cells. Although pol -deficient null mice are not viable (Sugo et al., 2000), suggesting that correctly performed BER is highly important for maintaining development, the corresponding embryonic cells survive in culture (Sobol et al., 1996). However, pol  is probably required in responding to endogenous oxidative genotoxic stress (Horton et al., 2002), and we predicted that inhibition of this enzyme in cells would affect their survival compared with wild-type cells grown in the same conditions. Therefore, we used a pol  knockout mice fibroblast cell line and its isogenic control wild-type cell and conducted growth analysis for both cell lines in the absence or presence of the inhibitor for 6 days. We found that cells lacking Pol  are less sensitive to MA compared with the control cells, suggesting an alteration of essential Pol -mediated DNA transactions (BER or single-strand break repair). This is illustrated in cellular proliferation experiments as a function of dose of MA, shown in Fig. 2D. As an additional control, we also measured the MA activity on replicative MEF and MEF pol eC/eC cell extracts and found that both extracts are resistant to MA (IC50 values of 15,000 and 4000 e, respectively), probably because Pol  at a basal level does not belong to the replication machinery. From this experiment, we suggest that MA could alter the Pol -mediated process of endogenous oxidative DNA damage.

    MA Best Fits the ssDNA Binding Site of the 8-kDa Domain of DNA Polymerase . The crystal and solution structure of the human Pol  have been determined previously (Sawaya et al., 1997; Maciejewski et al., 2000). We used the Pol  Protein Data Bank file 1BPY without DNA to dock MA using FlexX. This software is designed to identify the positive charges that are likely to bind carboxylate. In silico analysis indicated that MA is likely to interact with the ssDNA binding site of the 8-kDa N-terminal domain of Pol  (Fig. 3, AeCC). Figure 3A shows the ssDNA binding site of Pol  in the presence of the ssDNA. Figure 3B shows MA alone in the ssDNA binding site of Pol . Figure 3C represents the superposition of ssDNA and MA, showing that the inhibitor interacts with Lys60, Lys68, and Lys35. Lys60 makes a hydrogen bond with MA and Lys68 and Lys35 make ionic contact with the carbonyl of MA. The distances between those residues and MA are 2.86, 2.85, and 2.81 , respectively. It is interesting that Lys68 and Lys35 are known to be the main residues recognizing directly ssDNA (Prasad et al., 1998). We checked that the ssDNA binding site of Pol  does not differ in the absence of DNA by superposing the two structures (1BPY and 1DK3 without DNA; data not shown). Moreover, we found that MA does not bind the dNTP site of the active site in the palm domain, and band-shift assays indicated that MA interacts with the 8-kDa domain (data not shown), further suggesting a competitive inhibiting action of MA because of its association with the ssDNA binding site.

    Effect of the Three Inhibitors on the TLS across a Cisplatin Adduct. In vitro, Pol  efficiently replicates past a single Pt-(dGpG) lesion (Fig. 4B; compare lanes 1 and 2) during a 17-mer primer extension reaction on a platinated 60-mer template (Fig. 4A). We investigated the impact of the inhibitors during this specific Pol -mediated bypass synthesis. We determined inhibitory concentrations able to affect the TLS without inhibitory effect on undamaged DNA. As indicated in Fig. 4B (lanes 3eC8), MA is the most effective TLS inhibitor because only 30 e MA was sufficient for altering the bypass (Fig. 4B, lanes 3 and 4), whereas the inhibition was achieved with 100 e CA and 200 e MH (Fig. 4B, lanes 5eC8). It is noteworthy that the presence of the adduct changes the synthesis from distributive to processive in nature. We discussed this aspect in previous articles (Hoffmann et al., 1995, 1996), explaining that when the damage is present, the enzyme dissociates frequently from the adducted bases, resulting in more availability of Pol  to reinitiate primer extension and to extend most of the [3'-OH] termini to the site of the lesion. Furthermore, the synthesis upstream of the lesion is not altered in the presence of inhibitors. However concentrations of the inhibitors used for this TLS experiment could not be compared with those used in the calf thymus-activated DNA assay (Fig. 2), because the nature of DNA substrates is different, and the ratio [Pol /3'hydroxy termini of the DNA substrate] is much lower in the latter case.

    Cell Resistance to Cisplatin in the Presence of Inhibitors. We demonstrated previously (Bergoglio et al., 2001) that a Pol -dependent TLS across platinated DNA cross-links occurred in the 2008C135.25 human tumor cells, known to display a tolerant phenotype toward cisplatin. We proposed that this TLS process could contribute to the cisplatin resistance phenotype exhibited by these cells compared with the parental 2008 cells (Bergoglio et al., 2001). The potential of the three inhibitors to prevent Pol  from bypassing a cisplatin adduct in vitro prompted us to examine whether they could reduce the resistance to cisplatin of the 2008C135.25 cells. Clonogenic experiments were performed by incubating the cells with both cisplatin and a sublethal (80% cell survival) concentration of inhibitors (Fig. 4C). In the presence of MA, the strongest TLS inhibitor in vitro, we observed a significant sensitization of 2008C135.25 cells to cisplatin, whereas the response of parental 2008 cells to cisplatin was not altered. In contrast, MA and MH, which were less efficient for inhibiting TLS in vitro (Fig. 4B), had poor effect.

    Finally we wondered whether the deletion of pol  had any impact on the ability of MA to sensitize the cells to cisplatin. Figure 4D indicates that the response of pol  minus MEF cells to cisplatin was not modified by the presence of MA, whereas such a response is altered in wild-type MEF cells, with cisplatin survival of pol +/+ cells treated with MA being similar to that of pol  minus MEF cells. These data are in accordance with another study recently published, showing that a synthetic Pol  inhibitor did not affect the sensitivity of pol  knockout MEF cells to methyl methane sulfonate, whereas it alters the cell survival of wild-type cells (Hu et al., 2004). Our results further reinforce the in vitro findings showing the specific anti-Pol  inhibitory action of MA.

    Discussion

    We describe here the identification of three new natural molecules that inhibited Pol , an error-prone enzyme frequently overexpressed in tumor cells (Scanlon et al., 1989; Srivastava et al., 1999; Canitrot et al., 2000; Servant et al., 2002b). Among those molecules, we identified a product that we named mahureone A because it was purified from the leaves of M. palustris. The two other compounds identified were trans-communic acid, which is a ubiquitous diterpene of the labdane series isolated from J. communis, and masticadienonic acid, from P. lentiscus. To our knowledge, the latter is the most efficient Pol  inhibitor identified to date, with an IC50 value of 8 e. It is interesting that it altered to a much lesser extent the activity of either nuclear cell extracts or purified replicative Pol  and Pol , or error-prone DNA polymerases  and . MA could thus be of particular interest as a tool able to distinguish the biochemical features of the two closely related enzymes Pol  and Pol . Docking simulation shows that it could be located at the ssDNA-binding site of the 8-kDa N-terminal domain. This domain, also called dRPlyase domain, is only present in the X family DNA polymerases and Pol  from the Y family, therefore possibly explaining the specificity of inhibition compared with the replicative Pol . The ssDNA binding site of Pol  differs from that of Pol , presenting a lower positive charge, which might explain the different IC50 values of MA between Pol  and Pol . Before this work, prunasin was considered to be the best Pol  inhibitor, with an IC50 of 98 e (Mizushina et al., 1999). Our study reports a natural compound whose inhibitory action is more potent and preferentially targets Pol  compared with a wide range of other DNA polymerases. This was confirmed by using a pol -deficient null embryonic cell line, which was less sensitive to the action of MA than its isogenic wild-type cell line. Therefore, masticadienonic acid can be considered to be a potential useful molecular tool for investigating the role of Pol  among all of the X family DNA polymerases in normal somatic cells, such as in helping to distinguish its specific role in base excision repair pathway from Pol .

    The anticancer agent cisplatin is widely used in the treatment of ovarian, testicular, head, and neck carcinomas. Its therapeutic effects result from covalent binding to DNA, thus inhibiting replication and/or transcription (Pil and Lippard, 1997). Nevertheless, the cisplatin lesions are also mutagenic and could thus play a role in the generation of second tumors in patients with cancer treated with this agent. Another major obstacle for the successful treatment of cancer by cisplatin is drug resistance. Most of the studies on platinum resistance have focused on decreased drug uptake, enhanced nucleotide excision repair, or loss of DNA mismatch repair (Andrews and Howell, 1990). It has also been proposed that an alternative tumor resistance may be caused by an increased capacity of the cell to tolerate platinum/DNA lesions. We (Hoffmann et al., 1995; Canitrot et al., 1998) and others (Vaisman et al., 1999; Vaisman and Chaney, 2000) have shown that Pol  has the potential to efficiently catalyze error-prone translesion synthesis in vitro across the major intrastrand cross-link at the N-7 positions of adjacent guanine bases. In addition to alternative mechanisms such as drug uptake and DNA platination (Jekunen et al., 1994), this translesion mechanism has been proposed to contribute to the cisplatin resistance of the human ovarian carcinoma 2008C135.25 cells (Mamenta et al., 1994; Bergoglio et al., 2001), with extracts from these cells displaying enhanced replicative bypass and Pol -mediated translesion synthesis of platinum lesions. We found here that the three selected inhibitors affect the in vitro ability of Pol  to bypass a cisplatin adduct, with MA being the most potent. Therefore, we showed that MA decreased the ability of 2008C135.25 to adapt to cisplatin pressure. In contrast, MA did not affect the response to cisplatin of MEF pol eC/eC cells and confers to wild-type cells a pol eC/eC phenotype of resistance to cisplatin. We were surprised to observe in the control experiments that MA protected wild-type MEF cells from cisplatin toxicity, which are known to be sensitive to this agent (Raaphorst et al., 2002). So far, we cannot explain this discrepancy between cells that overexpress Pol  and those that only express the enzyme at a basal level. However, these data suggest that the intracellular level of Pol  protein in the response of cells to genotoxic stresses and the nature of the cells (tumorigenic and human in the case of 2008C13, embryonic and murine for MEF cells) are certainly of importance in the modulation of the adjuvant effect of masticadienonic acid. Another explanation (Raaphorst et al., 2002) could be that depletion of Pol  and, by extension, base excision repair favor recombination repair and cell survival.

    From a pharmaceutical point of view, masticadienonic acid could thus be considered a potentially useful adjuvant that is likely to reinforce the cisplatin action against tumors for which a Pol  overexpression "signature" would have been first diagnosed. It also could be considered as an antimutagenic drug likely to prevent side effects of cisplatin administration (i.e., emergence of secondary tumors) in the context of new individual therapeutic strategies.

    Acknowledgements

    We acknowledge S. B. Howell (San Diego, CA) for the gift of 2008 and 2008C13 cells. We also thank V. Masson (Centre de Criblage Pharmacologique, Toulouse, France) and K. Ramadan (Institute of Veterinary Biochemistry and Molecular Biology, Zurich, Switzerland) for their technical assistance. C. Moretti (L'Institut de Recherche pour le Deeveloppement, Cayenne, France) collected Mahurea palustris.

    F.B. and P.B. contributed equally to this work, and C.C. and J.S.H. contributed equally to this work.

    doi:10.1124/mol.104.001776.

    References

    Andrews PA and Howell SB (1990) Cellular pharmacology of cisplatin: perspectives on mechanisms of acquired resistance. Cancer Cells 2: 35eC43.

    Andrews PA, Murphy MP, and Howell SB (1985) Differential potentiation of alkylating and platinating agent cytotoxicity in human ovarian carcinoma cells by glutathione depletion. Cancer Res 45: 6250eC6253.

    Arya VP, Enzell C, Erdtman H, and Kubota T (1961) Communic acid, a new diterpene acid from Juniperus communis. Acta Chem Scand 225eC226.

    Barton DHR and Soane E (1956) The constitution and stereochemistry of masticadienonic acid. J Chem Soc 4150eC4157.

    Bergoglio V, Canitrot Y, Hogarth L, Minto L, Howell SB, Cazaux C, and Hoffmann JS (2001) Enhanced expression and activity of DNA polymerase beta in human ovarian tumor cells: impact on sensitivity towards antitumor agents. Oncogene 20: 6181eC6187.

    Bergoglio V, Ferrari E, Hubscher U, Cazaux C, and Hoffmann JS (2003) DNA polymerase beta can incorporate ribonucleotides during DNA synthesis of undamaged and CPD-damaged DNA. J Mol Biol 29: 1017eC1023.

    Bergoglio V, Pillaire MJ, Lacroix-Triki M, Raynaud-Messina B, Bieth A, Canitrot Y, Gares M, Wright M, Delsol G, Loeb L, et al. (2002) Deregulated DNA polymerase beta induces chromosome instability and tumorigenesis. Cancer Res 62: 3511eC3514.

    Bokesch HR, Groweiss A, McKee TC, and Boyd MR (1999) Laxifloranone, a new phloroglucinol derivative from Marila laxiflora. J Nat Prod 62: 1197eC1199.

    Bouayadi K, Hoffmann JS, Fons P, Tiraby M, Reynes JP, and Cazaux C (1997) Overexpression of DNA polymerase  sensitizes mammalian cells to 2',3'-dideoxycytidine and 3'-azido-3'-deoxythymidine. Cancer Res 57: 110eC116.

    Canitrot Y, Cazaux C, Frechet M, Bouayadi K, Lesca C, Salles B, and Hoffmann JS (1998) Overexpression of DNA polymerase beta in cell results in a mutator phenotype and a decreased sensitivity to anticancer drugs. Proc Natl Acad Sci USA 95: 12586eC12590.

    Canitrot Y, Hoffmann JS, Calsou P, Hayakawa H, Salles B, and Cazaux C (2000) Nucleotide excision repair DNA synthesis by excess DNA polymerase beta: a potential source of genetic instability in cancer cells. FASEB J 14: 1765eC1774.

    Canitrot Y, Lautier D, Laurent G, Freechet M, Ahmed A, Turhan AG, Salles B, Cazaux C, and Hoffmann JS (1999) Mutator phenotype of BCR-ABL transfected Ba/F3 cell lines and its association with enhanced expression of DNA polymerase . Oncogene 18: 2876eC2880.

    Copeland WC, Chen MS, and Wang TSF (1992) Human DNA polymerases  and  are able to incorporate anti-HIV deoxynucleotides into DNA. J Biol Chem 267: 21459eC21464.

    Deng JZ, Starck SR, Sun A, Salat M, and Hecht SM (2000) A new 7,8-euphadien-type triterpenoid from Brackenridgea nitida and Bleasdalea bleasdalei that inhibits DNA polymerase beta. J Nat Prod 63: 1357eC1361.

    Garcia-Diaz M, Bebenek K, Kunkel TA, and Blanco L (2001) Identification of an intrinsic 5'-deoxyribose-5'-phosphate lyase activity in human DNA polymerase lambda: a possible role in base excision repair. J Biol Chem 276: 34659eC34663.

    Garcia-Diaz M, Bebenek K, Sabariegos R, Dominguez O, Rodriguez J, Kirchhoff T, Garcia-Palomero E, Picher AJ, Juarez R, Ruiz JF, et al. (2002) DNA polymerase 32 , a novel DNA repair enzyme in human cells. J Biol Chem 277: 13184eC13191.

    Hoffmann JS, Pillaire MJ, Garcia-Estefania D, Lapalu S, and Villani G (1996) In vitro bypass replication of the cisplatin-d(GpG) lesion by calf thymus DNA polymerase  and human immunodeficiency virus type I reverse transcriptase is highly mutagenic. J Biol Chem 271: 15386eC15392.

    Hoffmann JS, Pillaire MJ, Maga G, Podust V, He筨scher U, and Villani G (1995) DNA polymerase  bypass in vitro d(GpG)-cisplatin adduct placed on codon 13 of H-ras gene. Proc Natl Acad Sci USA 92: 5356eC5360.

    Horton JK, Baker A, Vande Berg BJ, Sobol RW, and Wilson SH (2002) Involvement of DNA polymerase beta in protection against cytotoxicity of oxydative damage. DNA Repair 1: 317eC333.

    Hu HY, Horton JK, Gryk MR, Prasad R, Naron JM, Sun DA, Hecht SM, Wilson SH, and Mullen GP (2004) Identification of small molecule synthetic inhibitors of DNA polymerase  by NMR chemical shift mapping. J Biol Chem 279: 39736eC39744.

    Hubscher U, Maga G, and Spadari S (2002) Eukaryotic DNA polymerases. Annu Rev Biochem 71: 133eC163.

    Jekunen AP, Hom DK, Alcaraz JE, Eastman A, and Howell SB (1994) Cellular pharmacology of dichloro(ethylenediamine)platinum(II) in cisplatin-sensitive and resistant human ovarian carcinoma cells. Cancer Res 54: 2680eC2687.

    Louat T, Servant L, Rols MP, Teissie J, Hoffmann JS, and Cazaux C (2001) Antitumor activity of 2',3'-dideoxycytidine nucleotide analog against tumors up-regulating DNA polymerase beta. Mol Pharmacol 60: 553eC558.

    Maciejewski MW, Liu D, Prasad R, Wilson SH, and Mullen GP (2000) Backbone dynamics and refined solution structure of the N-terminal domain of DNA polymerase beta. Correlation with DNA binding and dRP lyase activity. J Mol Biol 296: 229eC253.

    Mamenta EL, Poma EE, Kaufmann WK, Delmastro DA, Grady HL, and Chaney SG (1994) Enhanced replicative bypass of platinum-DNA adducts in cisplatin-resistant human ovarian carcinoma cell lines. Cancer Res 54: 3500eC3505.

    Mizushina Y, Kamisuki S, Kasai N, Shimazaki N, Takemura M, Asahara H, Linn S, Yoshida S, Matsukage A, Koiwai O, et al. (2002) A plant phytotoxin, solanapyrone A, is an inhibitor of DNA polymerase beta and lambda. J Biol Chem 277: 630eC638.

    Mizushina Y, Takahashi N, Ogawa A, Tsurugaya K, Koshino H, Takemura M, Yoshida S, Matsukage A, Sugawara F, and Sakaguchi K (1999) The cyanogenic glucoside, prunasin (D-mandelonitrile-beta-D-glucoside), is a novel inhibitor of DNA polymerase beta. J Biochem 126: 430eC436.

    Nicholls A, Sharp KA, and Honig B (1991) Protein folding and association: insights from the interfacial and thermodynamic properties of hydrocarbons. Proteins 11: 281eC296.

    Pil P and Lippard S (1997) Encyclopedia of Cancer. Academic Press, San Diego.

    Podust VN, Chang LS, Ott R, Dianov GL, and Fanning E (2002) Reconstitution of human DNA polymerase using recombinant baculoviruses. J Biol Chem 277: 3894eC3901.

    Prasad R, Beard WA, Chyan JY, Maciejewski MW, Mullen GP, and Wilson SH (1998) Functional analysis of the amino-terminal 8-kDa domain of DNA polymerase beta as revealed by site-directed mutagenesis. DNA binding and 5'-deoxyribose phosphate lyase activities. J Biol Chem 273: 11121eC11126.

    Preston B, Poiesz B, and Loeb L (1988) Fidelity of HIV-1 reverse transcriptase. Science (Wash DC) 242: 1168eC1171.

    Raaphorst G, Ng C, and Yang D (2002) Comparison of response to radiation, hyperthermia and cisplatin in parental and polymerase beta knockout cells. Int J Hyperthermia 18: 33eC39.

    Radman M (1999) Enzymes of evolutionary change. Nature (Lond) 401: 866eC868.

    Ramadan K, Maga G, Shevelev I, Villani G, Blanco L, and Hubscher U (2003) Human DNA polymerase lambda possesses terminal deoxyribonucleotidyl transferase activity and can elongate RNA primers: implications for novel functions. J Mol Biol 328: 63eC72.

    Roberts J, Nguyen D, and Kunkel T (1993) Frameshift fidelity during replication of double-stranded DNA in HeLa cell extracts. Biochemistry 32: 4083eC4089.

    Sawaya MR, Prasad R, Wilson SH, Kraut J, and Pelletier H (1997) Crystal structures of human DNA polymerase beta complexed with gapped and nicked DNA: evidence for an induced fit mechanism. Biochemistry 36: 11205eC11215.

    Scanlon KJ, Kashani-Sabet M, and Miyachi H (1989) Differential gene expression in human cancer cells resistant to cisplatin. Cancer Investig 7: 581eC587.

    Servant L, Bieth A, Cazaux C, and Hoffmann JS (2002a) Involvement of DNA polymerase beta in DNA replication and mutagenic consequences. J Mol Biol 315: 1039eC1047.

    Servant L, Cazaux C, Bieth A, Iwai S, Hanaoka F, and Hoffmann JS (2002b) A role for DNA polymerase  in mutagenic UV lesion bypass. J Biol Chem 277: 50046eC50053.

    Skandalis A and Loeb LA (2001) Enzymatic properties of rat DNA polymerase beta mutants obtained by randomized mutagenesis. Nucleic Acids Res 29: 2418eC2426.

    Sobol RW, Horton JK, Ke筯n R, Gu H, Singhal RK, Prasad R, Rajewsky K, and Wilson SH (1996) Requirement of mammalian DNA polymerase-beta in base-excision repair. Nature (Lond) 379: 183eC186.

    Srivastava DK, Husain I, Arteaga CL, and Wilson SH (1999) DNA polymerase beta expression differences in selected human tumors and cell lines. Carcinogenesis 20: 1049eC1054.

    Sugo N, Aratani Y, Nagashima Y, Kubota Y, and Koyama H (2000) Neonatal lethality with abnormal neurogenesis in mice deficient in DNA polymerase beta. EMBO (Eur Mol Biol Organ) J 15: 1397eC1404.

    Vaisman A and Chaney SG (2000) The efficiency and fidelity of translesion synthesis past cisplatin and oxaliplatin GpG adducts by human DNA polymerase . J Biol Chem 275: 13017eC13025.

    Vaisman A, Lim SE, Patrick SM, Copeland WC, Hinkle DC, Turchi JJ, and Chaney SG (1999) Effect of DNA polymerases and high mobility group protein 1 on the carrier ligand specificity for translesion synthesis past platinum-DNA adducts. Biochemistry 38: 11026eC11039.

    Zhang JH, Chung TD, and Oldenburg KR (1999) A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J Biomol Screen 4: 67eC73.

作者: F. Boudsocq, P. Benaim, Y. Canitrot, M. Knibiehler 2007-5-15
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具