Screening of mammalian DNA polymerase and topoisomerase inhibitors from Garcinia mangostana L. and analysis of human cancer cell proliferation and apoptosis

  • Authors:
    • Takefumi Onodera
    • Yukiko Takenaka
    • Sachiko Kozaki
    • Takao Tanahashi
    • Yoshiyuki Mizushina
  • View Affiliations

  • Published online on: January 5, 2016     https://doi.org/10.3892/ijo.2016.3321
  • Pages: 1145-1154
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

We purified and identified eight xanthones from mangosteen (Garcinia mangostana L.) and investigated whether these compounds inhibited the activities of mammalian DNA polymerases (Pols) and human DNA topoisomerases (Topos). β-Mangostin was the strongest inhibitor of both mammalian Pols and human Topos among the isolated xanthones, with 50% inhibitory concentration (IC50) values of 6.4-39.6 and 8.5-10 µM, respectively. Thermal transition analysis indicated that β-mangostin did not directly bind to double-stranded DNA, suggesting that this compound directly bound the enzyme protein rather than the DNA substrate. β-Mangostin showed the strongest suppression of human cervical cancer HeLa cell proliferation among the eight compounds tested, with a 50% lethal dose (LD50) of 27.2 µM. This compound halted cell cycle in S phase at 12-h treatment and induced apoptosis. These results suggest that decreased proliferation by β-mangostin may be a result of the inhibition of cellular Pols rather than Topos, and β-mangostin might be an anticancer chemotherapeutic agent.

Introduction

Cancer is a major public health problem worldwide. Epidemiologic and animal studies have demonstrated an association with the inclusion of vegetables and fruits containing chemopreventive compounds in the diet and a reduced risk of cancer development (1,2). Our laboratory has spent over 20 years screening vegetables and fruits for natural phytochem-ical products that inhibit DNA metabolic enzymes, primarily mammalian DNA polymerases (Pols) and human DNA topoi-somerases (Topos).

Pols (DNA-dependent DNA polymerase, EC 2.7.7.7) catalyze the addition of deoxyribonucleotides to the 3′-hydroxyl terminus of primed double-stranded DNA (dsDNA) molecules (3). The human genome encodes at least 15 Pols that function in cellular DNA synthesis (4,5). Eukaryotic cells contain three replicative Pols (α, δ and ɛ), a single mitochon-drial Pol (γ), and at least 11 non-replicative Pols [β, ζ, η, θ, ι, κ, λ, μ, ν, terminal deoxynucleotidyl transferase (TdT), and REV1] (6,7). Pol structure is highly conserved, and the catalytic subunit shows little variance among species. Sequence homology classifies eukaryotic Pols into four main families: A, B, X and Y (6). Family A includes mitochondrial Pol γ as well as Pols θ and ν; family B includes the three replicative Pols α, δ and ɛ and also Pol ζ; family X comprises Pols β, λ and μ, as well as TdT; and family Y includes Pols η, ι and κ, in addition to REV1 (7).

Topos are nuclear enzymes that alter DNA topology and are required for the replication, transcription, recombination and segregation of daughter chromosomes (8). Eukaryotic cells have two types of Topos, I and II. Topo I catalyzes the passage of a DNA strand through a transient single-strand break in the absence of any high-energy cofactor. In contrast, Topo II catalyzes the passage of dsDNA through a transient double-strand break in the presence of ATP.

Selective inhibitors of Pols and Topos can suppress the proliferation of normal and cancerous human cells or exhibit cytotoxic effects (911). These inhibitors are potentially useful as anticancer, antiviral, antiparasitic or antipregnancy agents. In screening for these enzyme inhibitors in the ethanol extracts of various food materials, we found that the extract of mangosteen (Garcinia mangostana L.) from various parts of the fruit, such as the fruit, pericarp, leaves and rind, strongly inhibited the activities of mammalian Pols. We purified major components and identified eight xanthones by NMR and mass spectra analysis.

The mangosteen tree has been cultivated for centuries in tropical areas of the world. The tree is presumed to have originated in Southeast Asia or Indonesia and has largely remained indigenous to Malay Peninsula, Myanmar, Thailand, Cambodia, Vietnam and the Moluccas (12). The white, inner pulp of the mangosteen fruit is highly praised as one of the best tasting of all tropical fruits. This fruit has been dubbed the ‘queen of fruit’ in its native Thailand. The mangosteen rind, leaves and bark have been used as folk medicine for thousands of years (13). The thick mangosteen rind has been and is used for treating catarrh, cystitis, diarrhea, dysentery, eczema, fever, intestinal ailments, pruritis and other skin ailments. The mangosteen leaves are also used by some natives in teas and for diarrhea, dysentery, fever and thrush. Concentrates of mangosteen bark are used for genito-urinary afflictions and stomatosis.

Over 200 xanthones are currently known to exist in nature and ~50 of them are found in the mangosteen. The xanthones from mangosteen are gaining more interest because of their remarkable pharmacological effects, including not only anticancer activities (14,15), but also analgesic (16), antioxidant (17), anti-inflammatory (18), anti-allergy (19), antibacterial (20), anti-tuberculosis (21), antifungal (22), antiviral (23), cardioprotective (24), neuroprotective (25) and immunomodulation (26) effects. However, the underlying molecular mechanisms of these compounds and their effects on the activities of DNA metabolic enzymes have not been reported.

Therefore, the present study focused on the isolated eight xanthones from mangosteen and investigated these compounds for their inhibitory effects in vitro on mammalian Pol and human Topo activities.

Materials and methods

Materials

A chemically synthesized DNA template, poly(dA), and calf thymus DNA were purchased from Sigma-Aldrich Inc. and a customized oligo(dT)18 DNA primer was produced by Sigma-Aldrich Japan K.K. (Hokkaido, Japan). Radioactive nucleotide [3H]-labeled 2′-deoxythymidine-5′-triphosphate (dTTP; 43 Ci/mmol) was obtained from Moravek Biochemicals Inc. (Brea, CA, USA). Supercoiled pHOT-1 DNA was obtained from TopoGEN Inc. (Port Orange, FL, USA). All other reagents were analytical grade from Nacalai Tesque Inc. (Kyoto, Japan).

Isolation of xanthones from mangosteen

Some xanthone compounds were isolated from the leaves of mangosteen (Garcinia mangostana L.), which were collected in Vietnam and provided by Dr Duy Hoang Le (Kobe Pharmaceutical University). The chemical structures were determined by spectroscopic analyses and comparison with reported data (2730). The identified eight compounds are α-mangostin (1), β-mangostin (2), 3,6-di-O-methyl-γ-mangostin (3), fuscax-anthone C (4), gartanin (5), 8-deoxygartanin (6), mangostanin (7) and morusignin I (8) (Fig. 1).

Enzymes

Pol α was purified from calf thymus by immunoaffinity column chromatography, as described by Tamai et al (31). Recombinant rat Pol β was purified from Escherichia coli JMpβ5, as described by Date et al (32). The human Pol γ catalytic gene was cloned into pFastBac, with the histidine-tagged enzyme expressed using the BAC-TO-BAC HT Baculovirus Expression System according to the supplier's instructions (Life Technologies, Inc., Gaithersburg, MD, USA), and purified using ProBound resin (Invitrogen Japan K.K, Tokyo, Japan) (33). Human Pols δ and ɛ were purified by nuclear fractionation of human peripheral blood cancer cells (Molt-4) using the second subunit of Pols δ and ɛ-conjugated affinity column chromatography, respectively (34). A truncated form of human Pol η (residues 1–511) tagged with His6 at the C-terminus was expressed in E. coli and purified as described by Kusumoto et al (35). Recombinant mouse Pol ι tagged with His6 at the C-terminus was expressed and purified by Ni-NTA column chromatography (36). A truncated form of Pol κ (residues 1–560) with a His6-tag at the C-terminus was expressed in E. coli and purified as described by Ohashi et al (37). Recombinant human His-Pol λ was expressed and purified according to a method described by Shimazaki et al (38). Calf TdT, T7 RNA polymerase and T4 polynucleotide kinase were purchased from Takara Bio Inc. (Kyoto, Japan). Purified human placenta Topos I and II were purchased from TopoGEN Inc. Bovine pancreas deoxyribonuclease I was obtained from Stratagene Cloning Systems (La Jolla, CA, USA).

Measurement of Pol activity

The reaction mixtures for calf Pol α and rat Pol β have been described (39,40). Those for human Pol γ and for human Pols δ and ɛ were as described by Umeda et al (33) and Ogawa et al (41), respectively. The reaction mixtures for mammalian Pols η, ι and κ were the same as that for calf Pol α, and the reaction mixture for human Pol λ was the same as for rat Pol β. For Pols, poly(dA)/oligo(dT)18 (A/T=2/1) and tritium-labeled 2′-deoxythymidine 5′-triphos-phate ([3H]-dTTP) were used as the DNA template-primer and nucleotide [i.e., 2′-deoxynucleoside 5′-triphosphate (dNTP)] substrates, respectively. For the TdT reactions, oligo(dT)18 (3′-OH) and dTTP were used as the DNA primer substrate and nucleotide substrate, respectively.

The eight isolated xanthone compounds were dissolved in distilled dimethyl sulfoxide (DMSO) at various concentrations and sonicated for 30 sec. Aliquots of 4 μl of sonicated samples were mixed with 16 μl of each enzyme (final amount, 0.05 units) in 50 mM Tris-HCl (pH 7.5), containing 1 mM dithiothreitol, 50% glycerol and 0.1 mM ethylenediaminetetraacetic acid (EDTA), and kept at 0ºC for 10 min. These inhibitor-enzyme mixtures (8 μl) were added to 16 μl of each of the enzyme standard reaction mixtures and incubation carried out at 37ºC for 60 min. Activity without the inhibitor was considered 100% and the remaining activity at each inhibitor concentration was determined relative to this value. One unit of Pol activity was defined as the amount of enzyme that catalyzed incorporation of 1 nmol dNTP [measured using [3H]-dTTP] into synthetic DNA template-primers in 60 min at 37ºC under the normal reaction conditions for each enzyme (39,40).

Measurement of Topo activity

Purified human placental Topos I and II were purchased from TopoGen Inc. The catalytic activity of Topo I was determined by detecting supercoiled plasmid DNA (Form I) in its nicked form (Form II) (42). The Topo I reaction was performed in a 20-μl reaction mixture that contained 10 mM Tris-HCl (pH 7.9), pHOT-1 DNA (250 ng), 1 mM EDTA, 150 mM NaCl, 0.1% bovine serum albumin (BSA), 0.l mM spermidine, 5% glycerol, 2 μl of one of the eight isolated xanthone compounds dissolved in DMSO, and 2 units of Topo I. Topo II catalytic activity was analyzed in the same manner, except the reaction mixture contained 50 mM Tris-HCl (pH 8.0), 120 mM KCl, 10 mM MgCl2, 0.5 mM ATP, 0.5 mM dithiothreitol, supercoiled pHOT-1 DNA (250 ng), and 2 units of Topo II (42). Reaction mixtures were incubated at 37ºC for 30 min, followed by digestion with 1% sodium dodecyl sulfate (SDS) and 1 mg/ml proteinase K. After digestion, 2 μl loading buffer (5% sarkosyl, 0.0025% bromophenol blue and 25% glycerol) was added. The same procedure was followed for mobility shift assay assessment of enzyme DNA binding, except SDS denaturation and proteinase K digestion were omitted. Reaction mixtures were subjected to 1% agarose gel electrophoresis in Tris/borate/EDTA buffer. Agarose gels were stained with ethidium bromide (EtBr) and DNA band shifts from Form I to Form II were detected using an enhanced chemiluminescence detection system (Perkin-Elmer Life Sciences Inc., Waltham, MA, USA). Zero-D scan (Version 1.0, M & S Instruments Trading Inc., Osaka, Japan) was used for densitometric quantitation.

Other enzyme assays

The activities of T7 RNA polymerase, T4 polynucleotide kinase and bovine deoxyribonuclease I were measured using standard assays according to the manufacturer's specifications as described by Nakayama and Saneyoshi (43), Soltis and Uhlenbeck (44) and Lu and Sakaguchi (45), respectively.

Thermal transition of DNA

Thermal transition profiles of dsDNA to single-stranded DNA (ssDNA) with or without α-mangostin were obtained with a spectrophotometer (UV-2500; Shimadzu Corp., Kyoto, Japan) equipped with a thermoelectric cell holder was used as previously described (46). Calf thymus DNA (6 μg/ml) was dissolved in 0.1 M sodium phosphate buffer (pH 7.0) containing 1% DMSO. The solution temperature was equilibrated at 75ºC for 10 min, and then increased by 1ºC at 2-min intervals for each measurement point. Any change in the absorbance (260 nm) of the test compound itself at each temperature point was automatically subtracted from that of DNA plus the compound in the spectrophotometer.

Cell culture and measurement of cancer cell viability

The human cervical cancer cell line HeLa was obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). HeLa cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 100 units/ml penicillin and 100 μg/ml streptomycin. Cells were cultured at 37ºC in a humidified incubator containing 5% CO2/95% air. For assessment of cell growth, cells were plated at 2×103 cells/well in 96-well microplates, cultured for 12 h and various concentrations of the eight isolated xanthone compounds added. The compounds were dissolved in DMSO to produce 10-mM stock solutions, which were further diluted to appropriate final concentrations with growth medium containing 0.5% DMSO immediately prior to use. Cell viability was determined by the WST-1 assay following 24-h incubation (47).

Cell cycle analysis

Cellular DNA content for cell cycle analysis was determined as follows: aliquots of 3×105 HeLa cells were added to a 35-mm dish and incubated with the medium that contained the test compound for 0 to 24 h. Cells were then washed with ice-cold PBS three times, fixed with 70% (v/v) ethanol, and stored at −20ºC. DNA was stained with a PI (3,8-diamino-5-[3-(diethylmethylammonio)propyl]-6-phenyl-phenanthridinium diiodide) staining solution for at least 10 min at room temperature in the dark. Fluorescence intensity was measured using a BD FACSCalibur flow cytometer in combination with CellQuest software (Becton-Dickinson Co., Tokyo, Japan).

Measurement of caspase-3 activity

The enzymatic activity of caspase-3 was measured using EnzChek Caspase-3 Assay Kit #2 (Life Technologies Japan Co., Ltd., Tokyo, Japan), according to the instruction manual. HeLa cells (1×106) after inducing apoptosis with test compound were washed with PBS and lysed. Then, the caspase-3 activity in the extracts was determined with a fluorometric assay. The fluorescent product of the substrate Z-DEVD-Rhodamine 110 generated by caspase-3 in the cell extract was detected in a fluorescent microplate reader (SH-9000 Lab; Hitachi High-Technologies Corp., Tokyo, Japan) using an excitation of 496 nm and emission of 520 nm. To determine the background fluorescence of the substrate, test compound without the Z-DEVD-R110 substrate for negative controls, and mixture of activity buffer and Z-DEVD-R110 substrate for only substrate controls were also executed.

4′,6-Diamidino-2-phenylindole (DAPI) staining

Aliquots of 2.5×104 HeLa cells were plated in each well of an 8-well chamber slide (Thermo Fisher Scientific K.K., Tokyo, Japan). Then, the cells were incubated with or without the test compound (27.2 and 54.4 μM) for 12 and 24 h at 37ºC with 5% CO2. After washed with PBS twice, the cells were fixed with 4% paraformaldehyde (pH 7.4) for 15 min. Then, the cells were stained with DAPI for 1 min, and the percentage of apoptotic cells was counted under a fluorescence microscope (Olympus IX70; Olympus, Tokyo, Japan).

Statistical analysis

The data are expressed as the mean value ± the standard deviation (SD) of the mean of at least three independent determinations for each experiment. Statistical significance between each experimental group was analyzed using Student's t-test, and a probability level of 0.01 and 0.05 was used as the criterion of significance.

Results

Effect of the isolated xanthone compounds 1-8 on the activities of mammalian Pols

Selective inhibitors of mammalian Pols have been investigated with the goal to develop chemothera-peutic drugs for anticancer, antivirus and anti-inflammation treatments (48). In our ongoing screening for natural inhibitors of mammalian Pols, we found that the extract of mangosteen (Garcinia mangostana L.) had inhibitory activity against mammalian Pols. The isolated xanthone compounds 1-8 (Fig. 1) were selected and prepared for this study.

The inhibitory activity of each compound toward mammalian Pols was investigated using calf Pol α, rat Pol β, and human Pols γ and κ. For mammalian Pols, Pols α, β, γ and κ were used as the representatives of Pol families B, X, A and Y, respectively (6,7). Assessment of the relative activity of each Pol at a set concentration (10 μM) of the eight test compounds showed that β-mangostin (2) was the strongest inhibitor of these Pol species among the compounds tested (Fig. 2). In particular, this compound showed the strongest inhibitory activity against Pol α among the four mammalian Pols. α-Mangostin (1) and 3,6-di-O-methyl-γ-mangostin (3) slightly inhibited Pols α, γ, and κ activities and Pol κ activity, respectively. These results suggested that the common backbone structure with prenyl groups at C-2 and C-8 of compounds 1-3 might be important for Pol inhibition, and both the hydroxyl group at C-6 and the methoxy group at C-3, which are present in β-mangostin (2), must be much more important for Pol inhibitory activity.

Effect of the isolated xanthone compounds 1-8 on the activities of human Topos I and II

Next, the inhibitory effects of the eight isolated xanthones (10 μM each) were examined against human Topos I and II, which have ssDNA and dsDNA nicking activity, respectively (8). β-Mangostin (2) was the strongest inhibitor of both Topos I and II, and the inhibitory effect of this compound on Topo II was more potent than that on Topo I (Fig. 3). In contrast, mangostanin (7) and morusignin I (8) did not influence the activities of these Topos. Topo inhibition could be ranked as β-mangostin (2) >α-mangostin (1) >3,6-di-O-methyl-γ-mangostin (3) = fuscaxanthone C (4) = gartanin (5) = 8-deoxygartanin (6) >mangostanin (7) = morusignin I (8). Since β-mangostin (2) showed the strongest inhibitory activities on both mammalian Pols and human Topos, we focused on this compound in the subsequent study.

Effects of β-mangostin on the activities of various mammalian Pols and other DNA metabolic enzymes

Evaluation of the inhibition of DNA metabolic enzyme activities in vitro by β-mangostin revealed that this compound inhibited the activities of all eleven mammalian Pols tested from all four families. Calf Pol α and calf TdT were the strongest and weakest inhibited, respectively, with 50% inhibitory concentration (IC50) values of 6.4 and 39.6 μM, respectively (Table I). The inhibition by β-mangostin against mammalian Pol families could be ranked as B-family Pols >A-family Pol >Y-family Pols > X-family Pols. By comparison, the IC50 values of aphidicolin, a known eukaryotic DNA replicative Pols α, δ and ɛ inhibitor, were 20, 13 and 16 μM, respectively (49), showing that the Pol inhibitory activity of β-mangostin was >1.5-fold higher than that of aphidicolin. β-Mangostin inhibited the activity of human Topos I and II with IC50 values of 10.0 and 8.5 μM, respectively. Topotecan and doxorubicin, which are Topo I and Topo II inhibitors, respectively, also inhibited the nicking activities of Topos I and II with IC50 values of 45 and 60 μM, respectively (data not shown). The data show that the inhibitory effect on Topos I and II of β-mangostin was more potent than that of topotecan and doxorubicin, respectively. The inhibition of human Topos activities by β-mangostin was approximately the same order of potency compared with that of the activities of mammalian Pols except for X-family of Pols (Table I).

Table I

IC50 values of β-mangostin on the activities of mammalian Pols, human Topos and various DNA metabolic enzymes.

Table I

IC50 values of β-mangostin on the activities of mammalian Pols, human Topos and various DNA metabolic enzymes.

EnzymeIC50 values (μM)
Mammalian Pols
 A-Family of Pol
  Human Pol γ8.6±0.51
 B-Family of Pols
  Calf Pol α6.4±0.38
  Human Pol δ7.4±0.44
  Human Pol ɛ8.5±0.50
 X-Family of Pols
  Rat Pol β34.8±2.1
  Human Pol λ28.5±1.7
  Human Pol μ38.2±2.2
  Calf TdT39.6±2.3
 Y-Family of Pols
  Human Pol η9.3±0.55
  Mouse Pol ι8.7±0.52
  Human Pol κ9.2±0.54
Human Topos
 Human Topo I10±1.0
 Human Topo II8.5±0.9
Other DNA metabolic enzymes
 T7 RNA polymerase>200
 T4 polynucleotide kinase>200
 Bovine deoxyribonuclease I>200

[i] Compounds were incubated with each enzyme (0.05 units). Data, mean ± SD (n=3).

In contrast, β-mangostin did not influence the activities of other DNA metabolic enzymes, such as T7 RNA polymerase, T4 polynucleotide kinase, and bovine deoxyribonuclease I (Table I). These results indicate that β-mangostin can be specifically classified as an inhibitor of mammalian Pols and Topos.

Influence of β-mangostin on the hyperchromicity of dsDNA

Specific assays were performed to determine whether β-mangostin-induced inhibition resulted from the ability of the compound to bind to DNA or the enzyme. The interaction of β-mangostin with dsDNA was investigated by studying its thermal transition. The melting temperature (Tm) of dsDNA in the presence of an excess of β-mangostin (200 μM) was observed using a spectrophotometer equipped with a thermoelectric cell holder. As shown in Fig. 4, a thermal transition (i.e., Tm) from 75 to 90ºC was not observed within the concentration range used in the assay, whereas when a typical intercalating compound, such as EtBr (15 μM), was used as a positive control, an obvious thermal transition was observed.

We then assessed whether the inhibitory effect of β-mangostin on mammalian Pols and Topos resulted from non-specific adhesion to the enzyme or from selective binding to specific sites. Neither excessive nucleic acid [in the form of Poly(rC)] nor protein (BSA) significantly influenced β-mangostin-induced both Pol and Topo inhibition (data not shown). This suggests that β-mangostin selectively bound to the Pol and Topo molecules. These observations reveal that β-mangostin does not act as a DNA intercalating agent, and rather it directly binds the enzyme to inhibit its activity.

Collectively, these results suggested that β-mangostin might be a potent and selective inhibitor of mammalian Pols and Topos. We next investigated whether Pol and/or Topo inhibition by β-mangostin resulted in decreases in human cancer cell proliferation.

Effect of the isolated xanthone compounds 1-8 on cultured human cancer cells

Pols and Topos have recently emerged as important cellular targets for the development of anticancer agents. Therefore, we also investigated whether the eight isolated xanthones had any cytotoxic effect on HeLa cells. At 100 μM, all the compounds except for 3,6-di-O-methyl-γ-mangostin (3) potently suppressed the rate of HeLa cell growth to <80% that of control (Fig. 5). At 10 μM, β-mangostin (2) could moderately suppress HeLa cell growth, but other compounds did not influence cell proliferation. The 50% lethal dose (LD50) of β-mangostin (2) was 27.2 μM, which is ~2-fold higher than the IC50 for DNA replicative Pols (X-family Pols α, δ and ɛ) and Topos. This suggests that β-mangostin may be able to penetrate the cell membrane and reach the nucleus, where it may then inhibit Pols and/or Topos activities to suppress cell growth. Similar findings for all eight xanthones were observed in the HCT116 human colon carcinoma cell line (data not shown). These results reveal that Pol and Topo inhibition (Figs. 2 and 3) and human cancer cell cytotoxicity (Fig. 5) can be induced by xanthones from mangosteen. Therefore, Pol/Topo inhibition by β-mangostin represents a potentially useful anticancer effect.

Next we analyzed whether β-mangostin could affect the cell cycle distribution of HeLa cells. The cell cycle fraction was recorded after 0, 6, 12 and 24 h of treatment with a concentration of β-mangostin at 54.4 μM (the double density of LD50). The ratio of cells in each of the three cell cycle phases (G1, S and G2/M) is shown in Fig. 6A. Treatment with β-mangostin significantly increased the population of cells in S and G1 phase at 12 and 24 h, respectively, but did not influence the rate of G2/M phase. Dehydroaltenusin, which is a mammalian Pol α specific inhibitor, arrested the cell cycle in the G1 and S phases (50), and etoposide, which is a classical Topo II inhibitor, arrested the cell cycle in the G2/M phase (data not shown). These results suggest that β-mangostin may be an effective inhibitor of DNA replicative Pols, such as Pol α, that arrests the cell cycle at the G1 and S phases.

To examine whether the cell cycle arrest of HeLa cells treated with β-mangostin was due to apoptosis, caspase-3 activity was analyzed using fluorescent substrate (Fig. 6B). When HeLa cells were treated with the compound at 27.2 (=LD50 value) and 54.4 μM, the cells maximally increased caspase-3 activity for 24 and 12 h, respectively (Fig. 6B). In addition, preincubation with a caspase-3 inhibitor, DEVD-CHO, completely blocked this activity by β-mangostin (data not shown), suggesting that the apoptosis by this compound was mediated through caspase-3 activation.

Fig. 6C and D shows the extent of apoptotic cells by DAPI staining. The percentage of HeLa apoptotic cells after treatment with 27.2 and 54.4 μM of β-mangostin was markedly high for 24 and 12 h, respectively (Fig. 6C). These findings suggest that the human cancer cytotoxic effect of β-mangostin must involve a combination of cell proliferation arrest and apoptotic cell death.

Discussion

This is the first study to examine the inhibitory effects of xanthones isolated from mangosteen (G. mangostana L.) (i.e., compounds 1-8 in Fig. 1) on the activities of mammalian Pol species. β-Mangostin was the strongest inhibitor of mammalian Pols α, β, γ and κ as the representatives of Pol families B, X, A and Y, respectively (Fig. 2), and human Topos I and II (Fig. 3) among the compounds investigated. Its human cancer cytotoxicity (Fig. 5) was realized through the inhibition of the DNA metabolic enzymes Pols and Topos, which are essential for DNA replication, repair and recombination as well as cell division.

Topo inhibitors, such as adriamycin, amsacrine, ellipticine, saintopin, streptonigrin and terpentecin, are intercalating agents that are thought to bind directly to the DNA molecule and subsequently indirectly inhibit both types of Topo activity (51). These chemicals inhibit the DNA chain rejoining reactions catalyzed by Topos by stabilizing a tight Topo protein-DNA complex termed the ‘cleavable complex’ (52,53). The possible binding of β-mangostin to DNA was examined by measuring the Tm of dsDNA, and no β-mangostin was found to bind to dsDNA (Fig. 4). Topo inhibitors are categorized into two classes, ‘suppressors’, which are believed to interact directly with the enzyme, and ‘poisons’, which stimulate DNA cleavage and intercalation (54,55). β-Mangostin may be considered a suppressor of Topo functions rather than a conventional poison as this compound does not appear to stabilize Topo protein-DNA covalent complexes, as do the above-mentioned agents. Therefore, β-mangostin could be a new type of Topo inhibitor.

The fifteen Pols encoded by mammalian genomes are specialized for different functions, including DNA replication, DNA repair, recombination and translesion synthesis (6). Some Pol inhibitors, such as cytosine arabinoside (AraC), are anti-cancer drugs, because these compounds inhibit the activities of DNA replicative Pols α, δ and ɛ, suppress DNA synthesis, arrest cells at S phase, and induce apoptosis in cancer cells that have higher cellular Pol activity than normal cells (56). β-Mangostin suppressed proliferation of HeLa cell growth (Fig. 5) and arrested the cell cycle at S phase (Fig. 6A). These effects occur via the inhibition of Pols, especially DNA replicative B-family Pol species (Fig. 2 and Table I), suggesting that this compound affects the cellular DNA synthesis of cancer cells. Together this suggests that foods containing β-mangostin have potential positive effects in the prevention of cancer and promotion of health.

Acknowledgements

The present study was supported in part by the Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan, and the Japan-Supported Program for the Strategic Research Foundation at Private Universities, 2012–2016. T.T. acknowledges Kobe Pharmaceutical University Collaboration Fund.

Abbreviations:

Pol

DNA polymerase (E.C.2.7.7.7)

Topo

DNA topoisomerase

dsDNA

double-stranded DNA

TdT

terminal deoxynucleotidyl transferase

dTTP

2′-deoxythymidine-5′-tripho-sphate

dNTP

2′-deoxynucleoside 5′-triphosphate

DMSO

dimethyl sulfoxide

EDTA

ethylenediaminetetraacetic acid

BSA

bovine serum albumin

SDS

sodium dodecyl sulfate

EtBr

ethidium bromide

ssDNA

single-stranded DNA

DAPI

4′,6-diamidino-2-phenylindole

SD

standard deviation

IC50

50% inhibitory concentration

Tm

melting temperature

LD50

50% lethal dose

References

1 

Liu RH: Potential synergy of phytochemicals in cancer prevention: Mechanism of action. J Nutr. 134(Suppl): 3479S–3485S. 2004.PubMed/NCBI

2 

Surh YJ: Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer. 3:768–780. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Kornberg A and Baker TA: DNA replication. 2nd edition. W.D. Freeman and Co; New York: pp. 197–225. 1992

4 

Hubscher U, Maga G and Spadari S: Eukaryotic DNA polymerases. Annu Rev Biochem. 71:133–163. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Bebenek K and Kunkel TA: DNA repair and replication. Advances in Protein Chemistry. Yang W: Elsevier; San Diego: pp. 137–165. 2004, View Article : Google Scholar

6 

Lange SS, Takata K and Wood RD: DNA polymerases and cancer. Nat Rev Cancer. 11:96–110. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Loeb LA and Monnat RJ Jr: DNA polymerases and human disease. Nat Rev Genet. 9:594–604. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Wang JC: DNA topoisomerases. Annu Rev Biochem. 65:635–692. 1996. View Article : Google Scholar : PubMed/NCBI

9 

Liu LF: DNA topoisomerase poisons as antitumor drugs. Annu Rev Biochem. 58:351–375. 1989. View Article : Google Scholar : PubMed/NCBI

10 

Sakaguchi K, Sugawara F and Mizushina Y: Inhibitors of eukaryotic DNA polymerases. Seikagaku. 74:244–251. 2002.(In Japanese). PubMed/NCBI

11 

Berdis AJ: DNA polymerases as therapeutic targets. Biochemistry. 47:8253–8260. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Ji X, Avula B and Khan IA: Quantitative and qualitative determination of six xanthones in Garcinia mangostana L. by LC-PDA and LC-ESI-MS. J Pharm Biomed Anal. 43:1270–1276. 2007. View Article : Google Scholar

13 

Wexler B: Mangosteen. Woodland Publishing; Utah: 2007

14 

Akao Y, Nakagawa Y, Iinuma M and Nozawa Y: Anti-cancer effects of xanthones from pericarps of mangosteen. Int J Mol Sci. 9:355–370. 2008. View Article : Google Scholar

15 

Yoo JH, Kang K, Jho EH, Chin YW, Kim J and Nho CW: α- and γ-Mangostin inhibit the proliferation of colon cancer cells via β-catenin gene regulation in Wnt/cGMP signalling. Food Chem. 129:1559–1566. 2011. View Article : Google Scholar

16 

Cui J, Hu W, Cai Z, Liu Y, Li S, Tao W and Xiang H: New medicinal properties of mangostins: Analgesic activity and pharmacological characterization of active ingredients from the fruit hull of Garcinia mangostana L. Pharmacol Biochem Behav. 95:166–172. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Jung HA, Su BN, Keller WJ, Mehta RG and Kinghorn AD: Antioxidant xanthones from the pericarp of Garcinia mangostana (Mangosteen). J Agric Food Chem. 54:2077–2082. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Chen LG, Yang LL and Wang CC: Anti-inflammatory activity of mangostins from Garcinia mangostana. Food Chem Toxicol. 46:688–693. 2008. View Article : Google Scholar

19 

Nakatani K, Atsumi M, Arakawa T, Oosawa K, Shimura S, Nakahata N and Ohizumi Y: Inhibitions of histamine release and prostaglandin E2 synthesis by mangosteen, a Thai medicinal plant. Biol Pharm Bull. 25:1137–1141. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Sakagami Y, Iinuma M, Piyasena KG and Dharmaratne HR: Antibacterial activity of α-mangostin against vancomycin resistant Enterococci (VRE) and synergism with antibiotics. Phytomedicine. 12:203–208. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Suksamrarn S, Suwannapoch N, Phakhodee W, Thanuhiranlert J, Ratananukul P, Chimnoi N and Suksamrarn A: Antimycobacterial activity of prenylated xanthones from the fruits of Garcinia mangostana. Chem Pharm Bull (Tokyo). 51:857–859. 2003. View Article : Google Scholar

22 

Kaomongkolgit R, Jamdee K and Chaisomboon N: Antifungal activity of alpha-mangostin against Candida albicans. J Oral Sci. 51:401–406. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Chen SX, Wan M and Loh BN: Active constituents against HIV-1 protease from Garcinia mangostana. Planta Med. 62:381–382. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Devi Sampath P and Vijayaraghavan K: Cardioprotective effect of alpha-mangostin, a xanthone derivative from mangosteen on tissue defense system against isoproterenol-induced myocardial infarction in rats. J Biochem Mol Toxicol. 21:336–339. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Weecharangsan W, Opanasopit P, Sukma M, Ngawhirunpat T, Sotanaphun U and Siripong P: Antioxidative and neuroprotective activities of extracts from the fruit hull of mangosteen (Garcinia mangostana Linn.). Med Princ Pract. 15:281–287. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Tang YP, Li PG, Kondo M, Ji HP, Kou Y and Ou B: Effect of a mangosteen dietary supplement on human immune function: A randomized, double-blind, placebo-controlled trial. J Med Food. 12:755–763. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Ryu HW, Curtis-Long MJ, Jung S, Jin YM, Cho JK, Ryu YB, Lee WS and Park KH: Xanthones with neuraminidase inhibitory activity from the seedcases of Garcinia mangostana. Bioorg Med Chem. 18:6258–6264. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Ito C, Itoigawa M, Takakura T, Ruangrungsi N, Enjo F, Tokuda H, Nishino H and Furukawa H: Chemical constituents of Garcinia fusca: Structure elucidation of eight new xanthones and their cancer chemopreventive activity. J Nat Prod. 66:200–205. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Hano Y, Okamoto T, Suzuki K, Negishi M and Nomura T: Constituents of the Moraceae plants. 17 Components of the root bark of Morus insignis Bur 3 Structures of three new isopre-nylated xanthones morusignins I, J, and K and an isoprenylated flavone morusignin L. Heterocycles. 36:1359–1366. 1993. View Article : Google Scholar

30 

Bennett GJ, Lee HH and Das NP: Biosynthesis of mangostin. Part 1 The origin of the xanthone skeleton. J Chem Soc, Perkin Trans. 1(10): 2671–2676. 1990. View Article : Google Scholar

31 

Tamai K, Kojima K, Hanaichi T, Masaki S, Suzuki M, Umekawa H and Yoshida S: Structural study of immunoaffinity-purified DNA polymerase α-DNA primase complex from calf thymus. Biochim Biophys Acta. 950:263–273. 1988. View Article : Google Scholar : PubMed/NCBI

32 

Date T, Yamaguchi M, Hirose F, Nishimoto Y, Tanihara K and Matsukage A: Expression of active rat DNA polymerase β in Escherichia coli. Biochemistry. 27:2983–2990. 1988. View Article : Google Scholar : PubMed/NCBI

33 

Umeda S, Muta T, Ohsato T, Takamatsu C, Hamasaki N and Kang D: The D-loop structure of human mtDNA is destabilized directly by 1-methyl-4-phenylpyridinium ion (MPP+), a parkinsonism-causing toxin. Eur J Biochem. 267:200–206. 2000. View Article : Google Scholar

34 

Oshige M, Takeuchi R, Ruike T, Kuroda K and Sakaguchi K: Subunit protein-affinity isolation of Drosophila DNA polymerase catalytic subunit. Protein Expr Purif. 35:248–256. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Kusumoto R, Masutani C, Shimmyo S, Iwai S and Hanaoka F: DNA binding properties of human DNA polymerase et al: Implications for fidelity and polymerase switching of translesion synthesis. Genes Cells. 9:1139–1150. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Biertümpfel C, Zhao Y, Kondo Y, Ramón-Maiques S, Gregory M, Lee JY, Masutani C, Lehmann AR, Hanaoka F and Yang W: Structure and mechanism of human DNA polymerase eta. Nature. 465:1044–1048. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Ohashi E, Murakumo Y, Kanjo N, Akagi J, Masutani C, Hanaoka F and Ohmori H: Interaction of hREV1 with three human Y-family DNA polymerases. Genes Cells. 9:523–531. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Shimazaki N, Yoshida K, Kobayashi T, Toji S, Tamai K and Koiwai O: Over-expression of human DNA polymerase lambda in E. coli and characterization of the recombinant enzyme. Genes Cells. 7:639–651. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Mizushina Y, Tanaka N, Yagi H, Kurosawa T, Onoue M, Seto H, Horie T, Aoyagi N, Yamaoka M, Matsukage A, et al: Fatty acids selectively inhibit eukaryotic DNA polymerase activities in vitro. Biochim Biophys Acta. 1308:256–262. 1996. View Article : Google Scholar : PubMed/NCBI

40 

Mizushina Y, Yoshida S, Matsukage A and Sakaguchi K: The inhibitory action of fatty acids on DNA polymerase β. Biochim Biophys Acta. 1336:509–521. 1997. View Article : Google Scholar : PubMed/NCBI

41 

Ogawa A, Murate T, Suzuki M, Nimura Y and Yoshida S: Lithocholic acid, a putative tumor promoter, inhibits mammalian DNA polymerase β. Jpn J Cancer Res. 89:1154–1159. 1998. View Article : Google Scholar

42 

Yonezawa Y, Tsuzuki T, Eitsuka T, Miyazawa T, Hada T, Uryu K, Murakami-Nakai C, Ikawa H, Kuriyama I, Takemura M, et al: Inhibitory effect of conjugated eicosapentaenoic acid on human DNA topoisomerases I and II. Arch Biochem Biophys. 435:197–206. 2005. View Article : Google Scholar : PubMed/NCBI

43 

Nakayama C and Saneyoshi M: Inhibitory effects of 9-β-D-xylofuranosyladenine 5′-triphosphate on DNA-dependent RNA polymerase I and II from cherry salmon (Oncorhynchus masou). J Biochem. 97:1385–1389. 1985.PubMed/NCBI

44 

Soltis DA and Uhlenbeck OC: Isolation and characterization of two mutant forms of T4 polynucleotide kinase. J Biol Chem. 257:11332–11339. 1982.PubMed/NCBI

45 

Lu BC and Sakaguchi K: An endo-exonuclease from meiotic tissues of the basidiomycete Coprinus cinereus. Its purification and characterization. J Biol Chem. 266:21060–21066. 1991.PubMed/NCBI

46 

Mizushina Y, Murakami C, Ohta K, Takikawa H, Mori K, Yoshida H, Sugawara F and Sakaguchi K: Selective inhibition of the activities of both eukaryotic DNA polymerases and DNA topoisomerases by elenic acid. Biochem Pharmacol. 63:399–407. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Ishiyama M, Tominaga H, Shiga M, Sasamoto K, Ohkura Y and Ueno K: A combined assay of cell viability and in vitro cytotoxicity with a highly water-soluble tetrazolium salt, neutral red and crystal violet. Biol Pharm Bull. 19:1518–1520. 1996. View Article : Google Scholar : PubMed/NCBI

48 

Mizushina Y: Specific inhibitors of mammalian DNA polymerase species. Biosci Biotechnol Biochem. 73:1239–1251. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Mizushina Y, Kamisuki S, Mizuno T, Takemura M, Asahara H, Linn S, Yamaguchi T, Matsukage A, Hanaoka F, Yoshida S, et al: Dehydroaltenusin, a mammalian DNA polymerase α inhibitor. J Biol Chem. 275:33957–33961. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Murakami-Nakai C, Maeda N, Yonezawa Y, Kuriyama I, Kamisuki S, Takahashi S, Sugawara F, Yoshida H, Sakaguchi K and Mizushina Y: The effects of dehydroaltenusin, a novel mammalian DNA polymerase α inhibitor, on cell proliferation and cell cycle progression. Biochim Biophys Acta. 1674:193–199. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Gatto B, Capranico G and Palumbo M: Drugs acting on DNA topoisomerases: Recent advances and future perspectives. Curr Pharm Des. 5:195–215. 1999.PubMed/NCBI

52 

Larsen AK, Escargueil AE and Skladanowski A: Catalytic topoisomerase II inhibitors in cancer therapy. Pharmacol Ther. 99:167–181. 2003. View Article : Google Scholar : PubMed/NCBI

53 

Teicher BA: Next generation topoisomerase I inhibitors: Rationale and biomarker strategies. Biochem Pharmacol. 75:1262–1271. 2008. View Article : Google Scholar

54 

Bailly C: Topoisomerase I poisons and suppressors as anticancer drugs. Curr Med Chem. 7:39–58. 2000. View Article : Google Scholar : PubMed/NCBI

55 

Pommier Y, Leo E, Zhang H and Marchand C: DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem Biol. 17:421–433. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Park JK, Lee JS, Lee HH, Choi IS and Park SD: Accumulation of polycyclic aromatic hydrocarbon-induced single strand breaks is attributed to slower rejoining processes by DNA polymerase inhibitor, cytosine arabinoside in CHO-K1 cells. Life Sci. 48:1255–1261. 1991. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 48 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Onodera T, Takenaka Y, Kozaki S, Tanahashi T and Mizushina Y: Screening of mammalian DNA polymerase and topoisomerase inhibitors from Garcinia mangostana L. and analysis of human cancer cell proliferation and apoptosis. Int J Oncol 48: 1145-1154, 2016
APA
Onodera, T., Takenaka, Y., Kozaki, S., Tanahashi, T., & Mizushina, Y. (2016). Screening of mammalian DNA polymerase and topoisomerase inhibitors from Garcinia mangostana L. and analysis of human cancer cell proliferation and apoptosis. International Journal of Oncology, 48, 1145-1154. https://doi.org/10.3892/ijo.2016.3321
MLA
Onodera, T., Takenaka, Y., Kozaki, S., Tanahashi, T., Mizushina, Y."Screening of mammalian DNA polymerase and topoisomerase inhibitors from Garcinia mangostana L. and analysis of human cancer cell proliferation and apoptosis". International Journal of Oncology 48.3 (2016): 1145-1154.
Chicago
Onodera, T., Takenaka, Y., Kozaki, S., Tanahashi, T., Mizushina, Y."Screening of mammalian DNA polymerase and topoisomerase inhibitors from Garcinia mangostana L. and analysis of human cancer cell proliferation and apoptosis". International Journal of Oncology 48, no. 3 (2016): 1145-1154. https://doi.org/10.3892/ijo.2016.3321