Acessibilidade / Reportar erro

Evaluation of mutagenicity and metabolism-mediated cytotoxicity of the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin from Paepalanthus latipes

Abstract

A large number of quinones have been associated with antitumor, antibacterial, antimalarial, and antifungal activities. Results of previous studies of 5-methoxy-3,4-dehydroxanthomegnin, a naphthoquinone isolated fromPaepalanthus latipes Silveira, Eriocaulaceae, revealed antitumor, antibacterial, immunomodulatory, and antioxidant activities. In this study, we assessed the mutagenicity and metabolism-mediated cytotoxicity of 5-methoxy-3,4-dehydroxanthomegnin by using the Ames test and a microculture neutral red assay incorporating an S9 fraction (hepatic microsomal fraction and cofactors), respectively. We also evaluated the mutagenic activity inSalmonella typhimurium strains TA100, TA98, TA102, and TA97a, as well as the cytotoxic effect on McCoy cells with and without metabolic activation in both tests. Results indicated that naphthoquinone does not cause mutations by substitution or by addition and deletion of bases in the deoxyribonucleic acid sequence with and without metabolic activation. As previously demonstrated, the in vitro cytotoxicity of 5-methoxy-3,4-dehydroxanthomegnin to McCoy cells showed a significant cytotoxic index (CI50) of 11.9 μg/ml. This index was not altered by addition of the S9 fraction, indicating that the S9 mixture failed to metabolically modify the compound. Our results, allied with more specific biological assays in the future, would contribute to the safe use of 5-methoxy-3,4-dehydroxanthomegnin, compound that has showed in previous studies beneficial properties as a potential anticancer drug.

Cytotoxicity; 5-Methoxy-3,4-dehydroxanthomegnin; Mutagenicity


Introduction

“For many centuries, plants have provided a rich source of therapeutic agents and bases for synthetic drugs. Despite the development of organic synthesis, 25% of the drugs prescribed worldwide are derived from plant sources, showing that plant species are still an important source of new drugs” (Sacoman et al., 2008Sacoman, J.L., Monteiro, K.M., Possenti, A., Figueira, G.M., Foglio, M.A., Carvalho,.J.E., 2008. Cytotoxicity and antitumoral activity of dichloromethane extract and its fractions from Pothomorphe umbellata. Braz. J. Med. Biol. Res. 41, 411-415.). Much research has been conducted on plants in popular use, with the objective of identifying natural products with therapeutic potential (Balunas and Kinghorn, 2005Balunas, M.A., Kinghorn, A.D., 2005. Drug discovery from medicinal plants. Life Sci. 78,431-441.; Gurib-Fakim, 2006Gurib-Fakim, A., 2006. Medicinal plants: traditions of yesterday and drugs of tomorrow. Mol. Asp. Med. 27,1-93.; Newman and Cragg, 2012Newman, D.J., Cragg, G.M., 2012. NaturaI products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 75,311-335.).

The Eriocaulaceae family, commonly found in the States of Bahia and Minas Gerais, Brazil, has been the source of several biologically active compounds.Paepalanthus, with about 500 species, is one of its principal genus. A number of studies have demonstrated that almost all species ofPaepalanthus subgenus Platycaulon possess naphthopyranone derivatives, including paepalantine and 8,8′-paepalantine dimer isolated from Paepalanthus bromelioides (Vilegas et al., 1990Vilegas, W., Roque, N.F., Salatino, A., Giesbrecht, A.M., Davino, S., 1990. Isocoumarin from Paepalanthus bromelioides. Phytochemistry 29, 2299-2301.; Coelho et al., 2000Coelho, R.G., Vilegas, W., Devienne, K.F., Raddi, M.S.G., 2000. A new cytotoxic naphthopyrone dimer from Paepalanthus bromelioides. Fitoterapia 71, 497-500.), planifolin isolated from Paepalanthus planifolius (Santos et al., 2001Santos, L.C., Piacente, S., Pizza, C., Albert, K., Dachtler, M., Vilegas, W., 2001. Planifolin, a new naphthopyranone dimer and Xavonoids fromPaepalanthus planifolius. J. Nat. Prod. 64, 122-124.; Varanda et al., 2006Varanda, E.A., Varella, S.D., Rampazo, R.A., Kitagawa, R.R., Raddi, M.S.G., Vilegas, W., Santos, L.C., 2006. Mutagenic and cytotoxic effect of planifolin: a naphthopyranone dimer isolated from Paepalanthus planifolius. Toxicol. In Vitro 20, 664-668.), and 5-methoxy-3,4-dehydroxanthomegnin isolated from Paepalanthus latipes Silveira, Eriocaulaceae, (Kitagawa et al., 2004, 2008Kitagawa, R.R., Raddi, M.S.G., Santos, L.C., Vilegas, W., 2004. A new cytotoxic naphthoquinone from Paepalanthus latipes. Chem. Pharm. Bull. 52, 1487-1488.).

The compound 5-methoxy-3,4-dehydroxanthomegnin (1) is a naphthoquinone with potential therapeutic applications. Previous studies have shown that this compound has antitumor and immunomodulatory effects (Kitagawa et al., 2011Kitagawa, R.R., Vilegas, W., Carlos, I.Z., Raddi, M.S.G., 2011. Antitumor and immunomodulatory effects of the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin. Braz. J. Pharmacogn. 21, 1084-1088.), as well as anti-Helicobacter pylori and antioxidant properties (Kitagawa et al., 2012Kitagawa, R.R., Bonacorsi, C., Fonseca, L.M., Vilegas, W., Raddi, M.S.G., 2012. Anti-Helicobacter pylori activity and oxidative burst inhibition by the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin fromPaepalanthus latipes. Braz. J. Pharmacogn. 22, 53-59.). The antitumor effect of 5-methoxy-3,4-dehydroxanthomegnin may be enhanced by association with ascorbic acid as demonstrated by a significant cytotoxic index (CI) for McCoy cells. The enhanced effect is probably due to hydrogen peroxide generated by ascorbate-driven 5-methoxy-3,4-dehydroxanthomegnin redox cycling (Kitagawa et al., 2008Kitagawa, R.R., Fonseca, L.M., Ximenes, V.F., Khalil, N.M., Vilegas, W., Raddi, M.S.G., 2008. Ascorbic acid potentiates the cytotoxicity of the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin. Phytochemistry 69, 2205-2208.).


Currently, interest is focused on cytotoxic compounds that appear to exert a beneficial effect on key mechanisms involved in the pathogenesis of cancer and infection diseases. Many active compounds that work by interfering with the function of DNA seem to play a decisive role in antitumor activity (Harvey, 2008Harvey, A.L., 2008. Natural products in drug discovery. Drug Discov. Today 13, 894-901.; Ma and Wang, 2009Ma, X., Wang, Z., 2009. Anticancer drug discovery in the future: an evolutionary perspective. Drug Discov. Today 14, 1136-1142.; Sudan and Rupasinghe, 2014Sudan, S., Rupasinghe, H.P., 2014. Quercetin-3-O-glucoside induces human DNA topoisomerase II inhibition, cell cycle arrest and apoptosis in hepatocellular carcinoma cells. Anticancer Res. 34, 1691-1699.).

Short-term tests that detect genetic damage have allowed evaluation of the carcinogenic risks of chemicals to humans. The Ames assay, which is recommended for testing the mutagenicity of chemical compounds with potential pharmacological applications (Varanda et al., 2006Varanda, E.A., Varella, S.D., Rampazo, R.A., Kitagawa, R.R., Raddi, M.S.G., Vilegas, W., Santos, L.C., 2006. Mutagenic and cytotoxic effect of planifolin: a naphthopyranone dimer isolated from Paepalanthus planifolius. Toxicol. In Vitro 20, 664-668.; Resende et al., 2012Resende, F.A., Vilegas, W., Santos, L.C., Varanda, E.A., 2012. Mutagenicity of flavonoids assayed by bacterial reverse mutation (Ames) test. Molecules 17, 5255-5268.; Aardema, 2013Aardema, M.J., 2013. The holy grail in genetic toxicology: follow-up approaches for positive results in the Ames assay. Environ. Mol. Mutagen. 54,617-620.), was used in the present study to evaluatein vitro the mutagenic effect of 5-methoxy-3,4-dehydroxanthomegnin (1).

In vitro cytotoxicity tests simulate injury to cells from the tested substances, which may be caused by a number of incomplete mechanisms, during periods of exposure that are realistic for acute toxicity (Benbow et al., 2010Benbow, J.W., Aubrecht,J., Banker, M.J., Nettleton, D., Aleo, M.D., 2010. Predicting safety toleration of pharmaceutical chemical leads: cytotoxicity correlations to exploratory toxicity studies. Toxicol. Lett. 197, 175-182.). The central point regarding in vitro/in vivo comparisons refers to xenobiotic-metabolizing capacity. Bioactivation is an important consideration in in vitrocytotoxicity assays, since in vivo the test agent may be biotransformed. The incorporation of the S9 microsomal fraction has been used in the study of metabolic activation of chemicals in a variety of cell culture models (Soares et al., 2006Soares, V.C., Varanda, E.A., Raddi, M.S.G., 2006. In vitro basal and metabolism-mediated cytotoxicity of flavonoids. Food Chem. Toxicol. 44, 835-838.; Ponsoni et al., 2013Ponsoni, K., Raddi, M.S.G., Almeida, D.V., Almeida, A.E., Alécio, A.C., 2013. Effects of liver S9 enzymes on somalargine and solasodine cytotoxicity and mass spectrometric fragmentation. Eur. Food Res. Technol. 237, 179-184.). The test is applicable to the analysis of toxic ranges, for the detection of biotransformation of parent compounds, and for the evaluation of the cytotoxic effects of chemotherapeutic agents (Borenfreund and Puerner, 1987Borenfreund, E., Puerner, J.A., 1987. Short-term quantitativein vitro cytotoxicity assay involving an S-9 activating system. Cancer Lett. 34, 243-248.). In this study, we investigated in vitro metabolism-mediated cytotoxicity of 5-methoxy-3,4-dehydroxanthomegnin using the S9 system. For comparison, we evaluated cyclophosphamide, an indirect-acting cytotoxicant employed in antineoplastic therapy, as the control of biotransformation induction by the hepatic S9 fraction (Hill and Hill, 1984Hill, H.Z., Hill, G.L., 1984. In vitro activation of cyclophosphamide for an in vitro chemosensitivity assay. J. Surg. Oncol. 26, 225-229.).

Material and methods

Plant material

Paepalanthus latipes Silveira, Eriocaulaceae, was collected at Serra do Cipó in the Cadeia do Espinhaço, Minas Gerais, Brazil, and authenticated by Professor Paulo Takeo Sano from the Institute of Biosciences, University of São Paulo. The voucher specimen (CFSC, 13846) is on file at the Herbarium in the Department of Botany, Institute of Biosciences, University of São Paulo, Brazil.

Chemicals and culture media

Eagle medium was purchased from Adolfo Lutz (São Paulo, Brazil), and fetal bovine serum from Cultilab (Campinas, Brazil). Dimethyl sulfoxide (DMSO), nicotinamide adenine dinucleotide phosphate sodium salt (NADP), d-glucose-6-phosphate disodium salt, magnesium chloride (MgCl2), l-histidine monohydrate, d-biotin, sodium azide, 2-anthramine, and 2-aminofluorene were purchased from Sigma Chemical Co. (St. Louis, MO, USA). Oxoid Nutrient Broth No. 2 (Oxoid; Basingstoke, UK) and Bacto Agar (BD Bacto™; Sparks, MD, USA) were used as bacterial media. d-glucose, magnesium sulfate, citric acid monohydrate, anhydrous dibasic potassium phosphate, sodium ammonium phosphate, monobasic sodium phosphate, dibasic sodium phosphate, and sodium chloride were purchased from Merck (Whitehouse Station, NJ, USA). Neutral red (NR) was obtained from Riedel-de-Haën AG (Seelze, Hannover, Germany). The 5-methoxy-3,4-dehydroxanthomegnin, isolated and characterized as previously described (Kitagawa et al., 2004Kitagawa, R.R., Raddi, M.S.G., Santos, L.C., Vilegas, W., 2004. A new cytotoxic naphthoquinone from Paepalanthus latipes. Chem. Pharm. Bull. 52, 1487-1488.), was stored as stock solution at 10 mg/ml in DMSO.

Metabolic activation system (S9 mixture)

The S9 fraction, prepared from livers of Sprague-Dawley rats treated with the polychlorinated biphenyl mixture Aroclor 1254 (500 mg/kg), was purchased from Molecular Toxicology, Inc. (Boone, NC, USA). The metabolic activation system consisted of S9 fraction (4%), 0.4 M MgCl2 (1%), 1.65 M KCl (1%), 1 M d-glucose-6-phosphate disodium (0.5%), 0.1 M NADP (4%), 0.2 M phosphate buffer (50%), and sterile distilled water (39.5%) (Maron and Ames, 1983Maron, D.M., Ames, B.N., 1983. Revised methods for theSalmonella mutagenicity test. Mutat. Res. 113, 173-225.).

Salmonella mutagenic assay

Mutagenic activity was tested by Salmonella/microsome assay, using the Salmonella typhimurium tester strains TA97a, TA98, TA100, and TA102 (kindly provided by B.N. Ames; Berkeley, CA, USA), with and without metabolization by the preincubation method (Maron and Ames, 1983Maron, D.M., Ames, B.N., 1983. Revised methods for theSalmonella mutagenicity test. Mutat. Res. 113, 173-225.). The strains from frozen cultures were grown overnight for 12–14 h in Oxoid Nutrient Broth No. 2. The metabolic activation mixture (S9) was freshly prepared before each test. Various concentrations of 5-methoxy-3,4-dehydroxanthomegnin dissolved in DMSO were tested: 62.5, 125.0, 250.0, 500.0, and 750.0 μg/plate for strains TA98, TA97a, and TA100; 6.25, 12.5, 25.0, 50.0, and 75.0 μg/plate for strain TA102. These concentrations were selected based on a preliminary toxicity test. In all subsequent assays, the upper limit of the dose range tested was either the highest nontoxic dose or the lowest toxic dose determined in this preliminary assay. Toxicity was apparent as a reduction in the number of His+ revertants or as an alteration in the auxotrophic background (i.e., background lawn). The various concentrations of 5-methoxy-3,4-dehydroxanthomegnin were added to 0.5 ml of 0.2 M phosphate buffer (pH 7.4) or to 0.5 ml of 4% S9 mixture combined with 0.1 ml of bacterial culture and then incubated at 37 °C for 20–30 min. After this time, 2 ml of top agar was added to the mixture and poured on to a plate containing minimal agar. The plates were incubated at 37 °C for 48 h and the revertant colonies were counted manually. All experiments were analyzed in triplicate. The results were analyzed with the Salanal statistical software package, adopting the Bernstein et al. (1982)Bernstein, L., Kaldor, J., Mccann, J., Pike, M.C., 1982. An empirical approach to the statistical analysis of mutagenesis data from theSalmonella test. Mutat. Res. 97, 267-281. model. The data (revertants/plate) were assessed by analysis of variance (ANOVA), followed by linear regression. The mutagenic index (MI) was also calculated for each concentration tested, this being the average number of revertants per plate with the test compound divided by the average number of revertants per plate with the negative (solvent) control. A sample was considered mutagenic when a dose–response relationship was detected and a 2-fold increase in the number of mutants (MI ≥ 2) was observed with at least 1 concentration (Santos et al., 2006Santos, F.V., Colus, I.M.S., Silva, M.A., Vilegas, W., Varanda, E.A., 2006. Assessment of DNA damage induced by extracts and fractions ofStrychnos pseudoquina, a Brazilian medicinal plant with antiulcerogenic activity. Food Chem. Toxicol. 44, 1585-1589.). The standard mutagens used as positive controls in experiments without the S9 mixture were 4-nitro-O-phenylenediamine (10.0 μg/plate) for TA98 and TA97a, sodium azide (1.25 μg/plate) for TA100, and daunomycin (3.0 μg/plate) for TA102. In the experiments with metabolic activation, 2-anthramine (2.5 μg/plate) was used with TA98, TA97a, and TA100, and 2-aminofluorene (10.0 μg/plate) with TA102. DMSO served as the negative (solvent) control (75.0 μl/plate).

Cytotoxicity assay

McCoy cells (CCL1–ATCC/USA, from the cell culture section of the Adolfo Lutz Institute, São Paulo, Brazil) were maintained in Eagle medium with 7.5% fetal bovine serum. After trypsinization, 0.2 ml of medium containing approximately 104 cells/ml were seeded into 96-well microtiter tissue culture plates and incubated at 37 °C. After 24 h, the Eagle medium was removed and the cells were placed in unmodified medium (control) or in medium modified with various concentrations of 5-methoxy-3,4-dehydroxanthomegnin (5, 10, 20, 40, 50, 80, and 100 μg/ml) or cyclophosphamide (25, 50, 100, 150 μg/ml) with and without the S9 mixture at 10%. After incubating for another 24 h, the medium was removed and the plates were prepared for the NR assay (Borenfreund and Puerner, 1985Borenfreund, E., Puerner, J.A., 1985. Toxicity determined in vitro by morphological alterations and neutral red absorption. Toxicol. Lett. 24, 119-124.). After brief agitation, the plates were transferred to a microplate reader (Spectra and Rainbow [Shell] Readers; Tecan, Austria) and the optical density of each well was measured using a 540 nm filter and a 620 nm reference wavelength. All experiments were performed at least four times, using three wells for each concentration of compound tested. The cytotoxicity data were standardized by determining absorbance and calculating the corresponding chemical concentrations. Linear regression analysis with a 95% confidence limit was used to define dose–response curves and to compute the concentrations of chemical agents needed to reduce absorbance of the NR by 50% (IC50), called cytotoxic midpoint (Barile, 1994Barile, F.A., 1994. Introduction to In Vitro Cytotoxicology: Mechanisms and Methods. CRC Press, Inc., Boca Raton.).

Results

Tables 1 and 2 show the mutagenicity results for 5-methoxy-3,4-dehydroxanthomegnin which demonstrate that this compound does not possess mutagenic activity for the strains TA100, TA97a, and TA98, with MI less than 2.0 at all tested concentrations with or without metabolic activation. The TA102 strain was found to be more sensitive to the toxic effects of 5-methoxy-3,4-dehydroxanthomegnin; thus, it was necessary to decrease the concentrations used with TA102 relative to the other strains. In fact, the lowest concentration used in the experiments with TA100, TA97a, and TA98 was close to the highest concentration used with TA102. Nevertheless, the TA102 strain did not display mutagenicity in the absence or presence of metabolic activation.

Table 1
Mutagenic activity and mutagenic index (MI) of 5-methoxy-3,4-dehydroxanthomegnin (naphthoquinone) at various concentrations in strains TA100, TA98, and TA97a of Salmonella typhimuriumin the presence (+S9) and absence (−S9) of metabolic activation.
Table 2
Mutagenic activity and mutagenic index (MI) of 5-methoxy-3,4-dehydroxanthomegnin (naphthoquinone) at various concentrations in strain TA102 of Salmonella typhimurium in the presence (+S9) and absence (−S9) of metabolic activation.

The concentration–response curve for cytotoxicity was established for 5-methoxy-3,4-dehydroxanthomegnin in the presence and absence of an S9 mixture as an external metabolizing system (Fig. 1). Table 3 shows the IC50 of 5-methoxy-3,4-dehydroxanthomegnin and cyclophosphamide (control) for McCoy cells in the presence and absence of the S9 system. The IC50 obtained for 5-methoxy-3,4-dehydroxanthomegnin in the presence of the S9 mixture did not differ from that attained without the metabolizing system. Exposure of the McCoy cells to cyclophosphamide with and without S9 confirms the efficacy of the hepatic microsomal fraction for in vitro metabolic activation assay.

Fig. 1
Concentration-effect relationship of 5-methoxy-3,4-dehydroxanthomegnin (A) and cyclophosphamide (B) on McCoy cells with (red line) and without the S9 (blackline) metabolic activation system. Each point and bar represents the mean ± SD for at least three independent experiments carried out in triplicate. (For interpretation of thereferences to color in this text, the reader is referred to the web version of the article.)
Table 3
Cytotoxic midpoint (μg/ml) on McCoy cells for 5-methoxy-3,4-dehydroxanthomegnin and cyclophosphamide without and with the S9 metabolic activation.

Discussion

During the past few decades, plant research has revealed several chemical compounds with important pharmacological activities. Some of these compounds have been incorporated into drugs such as antineoplastics-vinblastine and vincristine isolated from Catharanthus roseus, camptothecin derivatives obtained fromCamptotheca acuminata, derivatives of podophyllotoxin from the rhizomes of Podophyllum peltatum and P. hexandrum, and taxol extracted from Taxus brevifolia (Cragg and Newman, 2005Cragg, G.M., Newman, D.J., 2005. Plants as a source of anti-cancer agents. J. Ethnopharmacol. 100, 72-79.; Srivastava et al., 2005Srivastava, V., Negi, A.S., Kumar, J.K., Gupta, M.M., Khanuja, S.P.S., 2005. Plant-based anticancer molecules: a chemical and biological profile of some important leads. Bioorg. Med. Chem. 13,5892-5908.; Basmadjian et al., 2014Basmadjian, C., Zhao, Q., Bentouhami, E., Djehal, A., Nebigil, C.G., Johnson, R.A., Serova, M., Gramont, A., Faivre, S., Raymond, E., Désaubry, L.G., 2014. Cancer wars: natural products strike back. Front. Chem. 2, 1-18.). However, the literature also describes many plants containing mutagenic compounds, such as furocoumarins, tannins, anthraquinones, alkaloids, and flavonoids (Rietjens et al., 2005Rietjens, I.M.C.M., Boersma, M.G., Vanderwoude, H., Jeurissen, S.M.F., Schutte, E.M., Alink, G.M., 2005. Flavonoids and akenylbenzenes: mechanisms of mutagenic action and carcinogenic risk. Mutat. Res. 574, 124-138.; Nesslany et al., 2009Nesslany, F., Simar-Meintières, S., Ficheux, H., Marzin, D., 2009. Aloe-emodin-induced DNA fragmentation in the mouse in vivocomet assay. Mutat. Res. 678, 13-19.; Guterres et al., 2013Guterres, Z.R., da Silva, A.F., Garcez, W.S., Garcez, F.R., Fernandes, C.A., Garcez, F.R., 2013. Mutagenicity and recombinagenicity ofOcotea acutifolia (Lauraceae) aporphinoid alkaloids. Mutat. Res. 757,91-96.; Mininel et al., 2014Mininel, F.J., Junior, C.S.L., Espanha, L.G., Resende, F.A., Varanda, E.A., Leite, C.Q.F., Vilegas, W., Santos, L.C., 2014. Characterization and quantification of compounds in the hydroalcoholic extract of the leaves fromTerminalia catappa Linn (Combretaceae) and their mutagenic activity. Evid. Based Compl. Alt., 1-11.). This evidence draws attention to the importance of studying the genetic risks of plant compounds, since the presence of mutagens in medicines can be dangerous to human health.

Previous studies have shown that species belonging to Paepalanthussubgenus Platycaulon possess naphthopyranone derivatives. The naphthopyranone paepalantine isolated from P. vellozioidesexhibited strong mutagenicity and cytotoxicity (Varanda et al., 1997Varanda, E.A., Raddi, M.S.G., Dias, F.L., Araujo, M.C.S., Gibran, S.C.A., Takahashi, C.S., Vilegas, W., 1997. Mutagenic and cytotoxic activity of an isocoumarin (Paepalantine) isolated from Paepalanthus vellozioides. Teratog. Carcinog. Mutagen. 17, 85-95.). The genotoxic potential of paepalantine was also demonstrated by Tavares et al. (1999)Tavares, D.C., Varanda, E.A., Andrade, F.P.D., Vilegas, W., Takahashi, C.S., 1999. Evaluation of the genotoxic potential of the isocoumarin paepalantine in vivo and in vitro mammalian system. J. Ethnopharmacol. 68, 115-120. inin vivo assays with bone marrow cells of Wistar rats. The mutagenic activity of planifolin isolated from P. planifolius was tested through Salmonella/microsome assays, with results indicating that this naphthopyranone dimer caused mutations by substitution and by addition and deletion of bases in the DNA sequence. Moreover, its mutagenic potential increased in the presence of metabolization. These results, allied to the chemical structure, suggest that planifolin may act as an intercalating agent in the DNA molecule (Varanda et al., 2006Varanda, E.A., Varella, S.D., Rampazo, R.A., Kitagawa, R.R., Raddi, M.S.G., Vilegas, W., Santos, L.C., 2006. Mutagenic and cytotoxic effect of planifolin: a naphthopyranone dimer isolated from Paepalanthus planifolius. Toxicol. In Vitro 20, 664-668.). In the present study, we assessed the mutagenic activity of 5-methoxy-3,4-dehydroxanthomegnin (1) using Salmonella/microsome assays. The compound 5-methoxy-3,4-dehydroxanthomegnin was not mutagenic to strain TA98, which is used to detect frameshift mutations. Substitution of DNA bases in strain TA100 and oxidative DNA damage in strain TA102 were not detected. No mutagenicity was observed in the TA97a strain, which detects frameshift mutations that are sensitive to heavy metal mutagens. The results were also negative for all the S. typhimuriumstrains tested with the S9 mixture.

However, data reported in the literature reveal that some quinones, including naphthoquinones, present mutagenicity after metabolization. Chesis et al. (1984)Chesis, P.L., Levin, D.E., Smith, M.T., Emster, L., Ames, B.N., 1984. Mutagenicity of quinones: pathways of metabolic activation and detoxification. Proc. Natl. Acad. Sci. 81,1696-1700. concluded that the mutagenicity of quinones was mainly due to one-electron reduction of quinones to semiquinonesvia the formation of superoxide anion radical (O2 ) and, subsequently, hydrogen peroxide (H2O2).Tikkanen et al. (1983)Tikkanen, L., Matsushima, T., Natori, S., Yoahihira, K., 1983. Mutagenicity of natural naphthoquinones and benzoquinone in theSalmonella/microsome test. Mutat. Res. 124, 25-34. observed that naphthoquinones with 1 or 2 hydroxyl and/or methyl substituents are mutagenic with metabolic activation. Nevertheless, it seems that the position of substituents, as well as the number of substituents, is important for mutagenicity. This point seems to explain the absence of mutagenicity of 5-methoxy-3,4-dehydroxanthomegnin, which has hydroxyl and methyl substituents and did not present mutagenicity in tests with or without metabolization.

Considerable interest has been focused on short-term in vitrocytotoxicity assays with cultured cells for evaluation of acute toxicities of chemical agents and pilot studies in drug development. Such assays would not only curtail the use of animals for median lethal dose (LD50) and similar tests, but would serve as an economical approach to the rapid screening of xenobiotics (Greene et al., 2010Greene, N., Aleo, M.D., Louise-May, S., Price, D.A., Will, Y., 2010. Using an in vitro cytotoxicity assay to aid in compound selection for in vivo safety studies. Bioorg. Med. Chem. Lett. 20,5308-5312.).

Cytotoxicity due to direct-acting chemicals is readily demonstrable in vitro. However, the toxicity of many chemicals is dependent upon metabolic activation, usually catalyzed by the microsomal cytochrome P-450-dependent monooxygenase system, and the majority of cell lines currently used in in vitro cytotoxicity tests possess little intrinsic drug-metabolic activity. Consequently, problems arise when metabolism-mediated cytotoxic events are studied in vitro. One possible answer to this problem is the coincubation of cultured cells with metabolically active rodent liver fractions, in a manner similar to their use in in vitro mutagenesis assays (Gonzalez, 2005Gonzalez, F.J., 2005. Role of cytochromes P450 in chemical toxicity and oxidative stress: studies with CYP2E1. Mutat. Res. 569,101-110.; Li et al., 2012Li, A.P., Uzgare, A., LaForge, Y.S., 2012. Definition of metabolism-dependent xenobiotic toxicity with co-cultures of human hepatocytes and mouse 3T3 fibroblasts in the novel integrated discrete multiple organ co-culture (IdMOC) experimental system: results with model toxicants aflatoxin B1, cyclophosphamide and tamoxifen. Chem. Biol. Interact. 199, 1-8.; Cole et al., 2014Cole, S.D., Madren-Whalley, J.S., Li, A.P., Dorsey, R., Salem, H., 2014. High content analysis of an in vitro model for metabolic toxicity: results with the model toxicants 4-aminophenol and cyclophosphamide. J. Biomol. Screen. 19, 1402-1408.).

In previous studies, 5-methoxy-3,4-dehydroxanthomegnin showed significant in vitro cytotoxicity to McCoy cells in the NR microculture test compared with cis-diamminedichloroplatinum (cisplatin), one of the most widely used chemotherapeutic drugs (Kitagawa et al., 2004Kitagawa, R.R., Raddi, M.S.G., Santos, L.C., Vilegas, W., 2004. A new cytotoxic naphthoquinone from Paepalanthus latipes. Chem. Pharm. Bull. 52, 1487-1488.). Our results indicated that combined treatment with 5-methoxy-3,4-dehydroxanthomegnin and hepatic S9 microsomal fraction did not alter the cytotoxicity of this naphthoquinone.

One hypothesis explaining the same cytotoxic potential of 5-methoxy-3,4-dehydroxanthomegnin with and without the S9 microsomal system is that this compound does not act as a substrate for the enzymatic system. Previous studies explored redox cycling of 5-methoxy-3,4-dehydroxanthomegnin in a nonenzymatic system. In this study, we used the NR assay to evaluate the ability of ascorbic acid associated with 5-methoxy-3,4-dehydroxanthomegnin to cause cell death in the same cell line. The synergic effect of ascorbic acid on 5-methoxy-3,4-dehydroxanthomegnin (1) resulted in a CI that was seven times lower than the index for 5-methoxy-3,4-dehydroxanthomegnin alone added to the McCoy cell line. The observed synergic effect was most probably due to H2O2 generated by ascorbate-driven 5-methoxy-3,4-dehydroxanthomegnin redox cycling (Kitagawa et al., 2008Kitagawa, R.R., Fonseca, L.M., Ximenes, V.F., Khalil, N.M., Vilegas, W., Raddi, M.S.G., 2008. Ascorbic acid potentiates the cytotoxicity of the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin. Phytochemistry 69, 2205-2208.), indicating that the association of 5-methoxy-3,4-dehydroxanthomegnin with ascorbic acid may be promising in the treatment of solid tumors that are deficient in antioxidant defenses.

The results of this study investigating the mutagenic activity and metabolism-mediated cytotoxicity of 5-methoxy-3,4-dehydroxanthomegnin, allied in the future with more specific biological assays, will contribute to the safe use of 5-methoxy-3,4-dehydroxanthomegnin, signifying its beneficial properties as a potential anticancer drug.

Acknowledgements

This study was supported by a grant of CNPq to R.R.K., to FAPESP for financial aid to W.V. and financial assistance from the PADC-UNESP.

References

  • Aardema, M.J., 2013. The holy grail in genetic toxicology: follow-up approaches for positive results in the Ames assay. Environ. Mol. Mutagen. 54,617-620.
  • Balunas, M.A., Kinghorn, A.D., 2005. Drug discovery from medicinal plants. Life Sci. 78,431-441.
  • Barile, F.A., 1994. Introduction to In Vitro Cytotoxicology: Mechanisms and Methods. CRC Press, Inc., Boca Raton.
  • Basmadjian, C., Zhao, Q., Bentouhami, E., Djehal, A., Nebigil, C.G., Johnson, R.A., Serova, M., Gramont, A., Faivre, S., Raymond, E., Désaubry, L.G., 2014. Cancer wars: natural products strike back. Front. Chem. 2, 1-18.
  • Benbow, J.W., Aubrecht,J., Banker, M.J., Nettleton, D., Aleo, M.D., 2010. Predicting safety toleration of pharmaceutical chemical leads: cytotoxicity correlations to exploratory toxicity studies. Toxicol. Lett. 197, 175-182.
  • Bernstein, L., Kaldor, J., Mccann, J., Pike, M.C., 1982. An empirical approach to the statistical analysis of mutagenesis data from theSalmonella test. Mutat. Res. 97, 267-281.
  • Borenfreund, E., Puerner, J.A., 1985. Toxicity determined in vitro by morphological alterations and neutral red absorption. Toxicol. Lett. 24, 119-124.
  • Borenfreund, E., Puerner, J.A., 1987. Short-term quantitativein vitro cytotoxicity assay involving an S-9 activating system. Cancer Lett. 34, 243-248.
  • Chesis, P.L., Levin, D.E., Smith, M.T., Emster, L., Ames, B.N., 1984. Mutagenicity of quinones: pathways of metabolic activation and detoxification. Proc. Natl. Acad. Sci. 81,1696-1700.
  • Coelho, R.G., Vilegas, W., Devienne, K.F., Raddi, M.S.G., 2000. A new cytotoxic naphthopyrone dimer from Paepalanthus bromelioides Fitoterapia 71, 497-500.
  • Cole, S.D., Madren-Whalley, J.S., Li, A.P., Dorsey, R., Salem, H., 2014. High content analysis of an in vitro model for metabolic toxicity: results with the model toxicants 4-aminophenol and cyclophosphamide. J. Biomol. Screen. 19, 1402-1408.
  • Cragg, G.M., Newman, D.J., 2005. Plants as a source of anti-cancer agents. J. Ethnopharmacol. 100, 72-79.
  • Gonzalez, F.J., 2005. Role of cytochromes P450 in chemical toxicity and oxidative stress: studies with CYP2E1. Mutat. Res. 569,101-110.
  • Greene, N., Aleo, M.D., Louise-May, S., Price, D.A., Will, Y., 2010. Using an in vitro cytotoxicity assay to aid in compound selection for in vivo safety studies. Bioorg. Med. Chem. Lett. 20,5308-5312.
  • Gurib-Fakim, A., 2006. Medicinal plants: traditions of yesterday and drugs of tomorrow. Mol. Asp. Med. 27,1-93.
  • Guterres, Z.R., da Silva, A.F., Garcez, W.S., Garcez, F.R., Fernandes, C.A., Garcez, F.R., 2013. Mutagenicity and recombinagenicity ofOcotea acutifolia (Lauraceae) aporphinoid alkaloids. Mutat. Res. 757,91-96.
  • Harvey, A.L., 2008. Natural products in drug discovery. Drug Discov. Today 13, 894-901.
  • Hill, H.Z., Hill, G.L., 1984. In vitro activation of cyclophosphamide for an in vitro chemosensitivity assay. J. Surg. Oncol. 26, 225-229.
  • Kitagawa, R.R., Raddi, M.S.G., Santos, L.C., Vilegas, W., 2004. A new cytotoxic naphthoquinone from Paepalanthus latipes Chem. Pharm. Bull. 52, 1487-1488.
  • Kitagawa, R.R., Fonseca, L.M., Ximenes, V.F., Khalil, N.M., Vilegas, W., Raddi, M.S.G., 2008. Ascorbic acid potentiates the cytotoxicity of the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin. Phytochemistry 69, 2205-2208.
  • Kitagawa, R.R., Vilegas, W., Carlos, I.Z., Raddi, M.S.G., 2011. Antitumor and immunomodulatory effects of the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin. Braz. J. Pharmacogn. 21, 1084-1088.
  • Kitagawa, R.R., Bonacorsi, C., Fonseca, L.M., Vilegas, W., Raddi, M.S.G., 2012. Anti-Helicobacter pylori activity and oxidative burst inhibition by the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin fromPaepalanthus latipes Braz. J. Pharmacogn. 22, 53-59.
  • Li, A.P., Uzgare, A., LaForge, Y.S., 2012. Definition of metabolism-dependent xenobiotic toxicity with co-cultures of human hepatocytes and mouse 3T3 fibroblasts in the novel integrated discrete multiple organ co-culture (IdMOC) experimental system: results with model toxicants aflatoxin B1, cyclophosphamide and tamoxifen. Chem. Biol. Interact. 199, 1-8.
  • Ma, X., Wang, Z., 2009. Anticancer drug discovery in the future: an evolutionary perspective. Drug Discov. Today 14, 1136-1142.
  • Maron, D.M., Ames, B.N., 1983. Revised methods for theSalmonella mutagenicity test. Mutat. Res. 113, 173-225.
  • Mininel, F.J., Junior, C.S.L., Espanha, L.G., Resende, F.A., Varanda, E.A., Leite, C.Q.F., Vilegas, W., Santos, L.C., 2014. Characterization and quantification of compounds in the hydroalcoholic extract of the leaves fromTerminalia catappa Linn (Combretaceae) and their mutagenic activity. Evid. Based Compl. Alt., 1-11.
  • Nesslany, F., Simar-Meintières, S., Ficheux, H., Marzin, D., 2009. Aloe-emodin-induced DNA fragmentation in the mouse in vivocomet assay. Mutat. Res. 678, 13-19.
  • Newman, D.J., Cragg, G.M., 2012. NaturaI products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 75,311-335.
  • Ponsoni, K., Raddi, M.S.G., Almeida, D.V., Almeida, A.E., Alécio, A.C., 2013. Effects of liver S9 enzymes on somalargine and solasodine cytotoxicity and mass spectrometric fragmentation. Eur. Food Res. Technol. 237, 179-184.
  • Resende, F.A., Vilegas, W., Santos, L.C., Varanda, E.A., 2012. Mutagenicity of flavonoids assayed by bacterial reverse mutation (Ames) test. Molecules 17, 5255-5268.
  • Rietjens, I.M.C.M., Boersma, M.G., Vanderwoude, H., Jeurissen, S.M.F., Schutte, E.M., Alink, G.M., 2005. Flavonoids and akenylbenzenes: mechanisms of mutagenic action and carcinogenic risk. Mutat. Res. 574, 124-138.
  • Sacoman, J.L., Monteiro, K.M., Possenti, A., Figueira, G.M., Foglio, M.A., Carvalho,.J.E., 2008. Cytotoxicity and antitumoral activity of dichloromethane extract and its fractions from Pothomorphe umbellata Braz. J. Med. Biol. Res. 41, 411-415.
  • Santos, L.C., Piacente, S., Pizza, C., Albert, K., Dachtler, M., Vilegas, W., 2001. Planifolin, a new naphthopyranone dimer and Xavonoids fromPaepalanthus planifolius J. Nat. Prod. 64, 122-124.
  • Santos, F.V., Colus, I.M.S., Silva, M.A., Vilegas, W., Varanda, E.A., 2006. Assessment of DNA damage induced by extracts and fractions ofStrychnos pseudoquina, a Brazilian medicinal plant with antiulcerogenic activity. Food Chem. Toxicol. 44, 1585-1589.
  • Soares, V.C., Varanda, E.A., Raddi, M.S.G., 2006. In vitro basal and metabolism-mediated cytotoxicity of flavonoids. Food Chem. Toxicol. 44, 835-838.
  • Srivastava, V., Negi, A.S., Kumar, J.K., Gupta, M.M., Khanuja, S.P.S., 2005. Plant-based anticancer molecules: a chemical and biological profile of some important leads. Bioorg. Med. Chem. 13,5892-5908.
  • Sudan, S., Rupasinghe, H.P., 2014. Quercetin-3-O-glucoside induces human DNA topoisomerase II inhibition, cell cycle arrest and apoptosis in hepatocellular carcinoma cells. Anticancer Res. 34, 1691-1699.
  • Tavares, D.C., Varanda, E.A., Andrade, F.P.D., Vilegas, W., Takahashi, C.S., 1999. Evaluation of the genotoxic potential of the isocoumarin paepalantine in vivo and in vitro mammalian system. J. Ethnopharmacol. 68, 115-120.
  • Tikkanen, L., Matsushima, T., Natori, S., Yoahihira, K., 1983. Mutagenicity of natural naphthoquinones and benzoquinone in theSalmonella/microsome test. Mutat. Res. 124, 25-34.
  • Varanda, E.A., Raddi, M.S.G., Dias, F.L., Araujo, M.C.S., Gibran, S.C.A., Takahashi, C.S., Vilegas, W., 1997. Mutagenic and cytotoxic activity of an isocoumarin (Paepalantine) isolated from Paepalanthus vellozioides Teratog. Carcinog. Mutagen. 17, 85-95.
  • Varanda, E.A., Varella, S.D., Rampazo, R.A., Kitagawa, R.R., Raddi, M.S.G., Vilegas, W., Santos, L.C., 2006. Mutagenic and cytotoxic effect of planifolin: a naphthopyranone dimer isolated from Paepalanthus planifolius Toxicol. In Vitro 20, 664-668.
  • Vilegas, W., Roque, N.F., Salatino, A., Giesbrecht, A.M., Davino, S., 1990. Isocoumarin from Paepalanthus bromelioides Phytochemistry 29, 2299-2301.
  • Erratum

    In the above paper, on page 17, first column, where the chemical structure1:

    It should be:

Publication Dates

  • Publication in this collection
    Jan-Feb 2015

History

  • Received
    22 Apr 2014
  • Accepted
    19 Dec 2014
Sociedade Brasileira de Farmacognosia Universidade Federal do Paraná, Laboratório de Farmacognosia, Rua Pref. Lothario Meissner, 632 - Jd. Botânico, 80210-170, Curitiba, PR, Brasil, Tel/FAX (41) 3360-4062 - Curitiba - PR - Brazil
E-mail: revista@sbfgnosia.org.br