Publications

Found 31 results
Filters: Author is R.J. Oliveira  [Clear All Filters]
2016
M. F. Magosso, Carvalho, P. C., Shneider, B. U. C., Pessatto, L. R., Pesarini, J. R., Silva, P. V. B., Correa, W. A., Kassuya, C. A. L., Muzzi, R. M., Oliveira, R. J., Magosso, M. F., Carvalho, P. C., Shneider, B. U. C., Pessatto, L. R., Pesarini, J. R., Silva, P. V. B., Correa, W. A., Kassuya, C. A. L., Muzzi, R. M., and Oliveira, R. J., Acrocomia aculeata prevents toxicogenetic damage caused by the antitumor agent cyclophosphamide, vol. 15, p. -, 2016.
M. F. Magosso, Carvalho, P. C., Shneider, B. U. C., Pessatto, L. R., Pesarini, J. R., Silva, P. V. B., Correa, W. A., Kassuya, C. A. L., Muzzi, R. M., Oliveira, R. J., Magosso, M. F., Carvalho, P. C., Shneider, B. U. C., Pessatto, L. R., Pesarini, J. R., Silva, P. V. B., Correa, W. A., Kassuya, C. A. L., Muzzi, R. M., and Oliveira, R. J., Acrocomia aculeata prevents toxicogenetic damage caused by the antitumor agent cyclophosphamide, vol. 15, p. -, 2016.
M. D. Martello, David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., Oliveira, R. J., Martello, M. D., David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., Oliveira, R. J., Martello, M. D., David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., and Oliveira, R. J., Campomanesia adamantium extract induces DNA damage, apoptosis, and affects cyclophosphamide metabolism, vol. 15, p. -, 2016.
M. D. Martello, David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., Oliveira, R. J., Martello, M. D., David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., Oliveira, R. J., Martello, M. D., David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., and Oliveira, R. J., Campomanesia adamantium extract induces DNA damage, apoptosis, and affects cyclophosphamide metabolism, vol. 15, p. -, 2016.
M. D. Martello, David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., Oliveira, R. J., Martello, M. D., David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., Oliveira, R. J., Martello, M. D., David, N., Matuo, R., Carvalho, P. C., Navarro, S. D., Monreal, A. C. D., Cunha-Laura, A. L., Cardoso, C. A. L., Kassuya, C. A. L., and Oliveira, R. J., Campomanesia adamantium extract induces DNA damage, apoptosis, and affects cyclophosphamide metabolism, vol. 15, p. -, 2016.
L. C. Hermeto, DeRossi, R., Oliveira, R. J., Pesarini, J. R., Antoniolli-Silva, A. C. M. B., Jardim, P. H. A., Santana, Á. E., Deffune, E., Rinaldi, J. C., Justulin, L. A., Hermeto, L. C., DeRossi, R., Oliveira, R. J., Pesarini, J. R., Antoniolli-Silva, A. C. M. B., Jardim, P. H. A., Santana, Á. E., Deffune, E., Rinaldi, J. C., and Justulin, L. A., Effects of intra-articular injection of mesenchymal stem cells associated with platelet-rich plasma in a rabbit model of osteoarthritis, vol. 15, p. -, 2016.
L. C. Hermeto, DeRossi, R., Oliveira, R. J., Pesarini, J. R., Antoniolli-Silva, A. C. M. B., Jardim, P. H. A., Santana, Á. E., Deffune, E., Rinaldi, J. C., Justulin, L. A., Hermeto, L. C., DeRossi, R., Oliveira, R. J., Pesarini, J. R., Antoniolli-Silva, A. C. M. B., Jardim, P. H. A., Santana, Á. E., Deffune, E., Rinaldi, J. C., and Justulin, L. A., Effects of intra-articular injection of mesenchymal stem cells associated with platelet-rich plasma in a rabbit model of osteoarthritis, vol. 15, p. -, 2016.
R. J. Oliveira, de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., Antoniolli-Silva, A. C. M. B., Oliveira, R. J., de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., Antoniolli-Silva, A. C. M. B., Oliveira, R. J., de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., and Antoniolli-Silva, A. C. M. B., Moquiniastrum polymorphum subsp floccosum extract: screening for mutagenic and antimutagenic activity, vol. 15, no. 4, p. -, 2016.
Conflicts of interest The authors declare no conflicts of interest. ACKNOWLEDGMENTS Research supported by FUNDECT (Fundação de Apoio ao Desenvolvimento do Ensino, Ciência e Tecnologia do Estado de Mato Grosso do Sul), CAPES (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior), and CNPq (Conselho Nacional de Desenvolvimento Científico e Tecnológico). REFERENCES Bohlmann F, Zdero C, Schmeda-Hirschmann G, Jakupovic J, et al (1986). Dimeric guainolides and other constituents from Gochnatia species. Phytochemistry 44: 1175-1178. http://dx.doi.org/10.1016/S0031-9422(00)81575-7 Bueno NR, Castilho RO, Costa RB, Pott A, et al (2005). Medicinal plants used by the Kaiowá and Guarani indigenous populations in the Caarapó reserve, Mato Grosso do Sul, Brazil. Acta Bot. Bras. 19: 39-44. http://dx.doi.org/10.1590/S0102-33062005000100005 Cantero WB, Takahachi NA, Mauro MO, Pesarini JR, et al (2015). Genomic lesions and colorectal carcinogenesis: the effects of protein-calorie restriction and inulin supplementation on deficiency statuses. Genet. Mol. Res. 14: 2422-2435. http://dx.doi.org/10.4238/2015.March.27.27 Catalan CAN, Vega MI, Lopez ME, Cuenca M del R, et al (2003). Coumarins and a kaurane from Gochnatia polymorpha ssp. Polymorpha from Paraguay. Biochem. Syst. Ecol. 31: 417-422. http://dx.doi.org/10.1016/S0305-1978(02)00163-1 Cooper EL, et al (2004). Drug discovery, CAM and natural products. Evid. Based Complement. Alternat. Med. 1: 215-217. http://dx.doi.org/10.1093/ecam/neh032 Cragg GM, Newman DJ, et al (2013). Natural products: a continuing source of novel drug leads. Biochim. Biophys. Acta 1830: 3670-3695. http://dx.doi.org/10.1016/j.bbagen.2013.02.008 David Nd, Mauro MdeO, Gonçalves CA, Pesarini JR, et al (2014). Gochnatia polymorpha ssp. floccosa: bioprospecting of an anti-inflammatory phytotherapy for use during pregnancy. J. Ethnopharmacol. 154: 370-379. http://dx.doi.org/10.1016/j.jep.2014.04.005 Farias ACM, Da Silva JR, Tomassini TCB, et al (1984). Constituents of Mochinea polymorpha. J. Nat. Prod. 47: 363-364. http://dx.doi.org/10.1021/np50032a021 Fedel-Miyasato LES, Formagio ASN, Auharek SA, Kassuya CAL, et al (2014a). Antigenotoxic and antimutagenic effects of Schinus terebinthifolius Raddi in Allium cepa and Swiss mice: a comparative study. Genet. Mol. Res. 13: 3411-3425. http://dx.doi.org/10.4238/2014.April.30.2 Fedel-Miyasato LES, Kassuya CAL, Auharek SA, Formagio ASN, et al (2014b). Evaluation of anti-inflammatory, immunomodulatory, chemopreventive and wound healing potentials from Schinus terebinthifolius methanolic extract. Braz. J. Pharmacog. 24: 565-575. Felicidade I, Lima JD, Pesarini JR, Monreal ACD, et al (2014a). Mutagenic and antimutagenic effects of aqueous extract of rosemary (Rosmarinus officinalis L.) on meristematic cells of Allium cepa. Genet. Mol. Res. 13: 9986-9996. http://dx.doi.org/10.4238/2014.November.28.3 Felicidade I, Lima JD, Pesarini JR, Monreal ACD, et al (2014b). Mutagenic and antimutagenic effects of crude hydroalcoholic extract of rosemary (Rosmarinus officinalis L.) on cultured meristematic cells Allium cepa. VRI Phytomedicine 2: 30-39. Fiskesjö G, et al (1993). The Allium test in wastewater monitoring. Environ. Toxicol. 8: 291-298. Leme DM, Marin-Morales MA, et al (2009). Allium cepa test in environmental monitoring: a review on its application. Mutat. Res. 682: 71-81. http://dx.doi.org/10.1016/j.mrrev.2009.06.002 López ME, Giordano OS, López LA, et al (2002). Sesquiterpene lactone dehydroleucodine selectively induces transient arrest in G2 in Allium cepa root meristematic cells. Protoplasma 219: 82-88. http://dx.doi.org/10.1007/s007090200008 Magosso MF, Carvalho PC, Shneider BU, Pessatto LR, et al (2016). Acrocomia aculeata prevents toxicogenetic damage caused by the antitumor agent cyclophosphamide. Genet. Mol. Res. 15. http://dx.doi.org/10.4238/gmr.15027816 Manoharan K, Banerjee MR, et al (1985). beta-Carotene reduces sister chromatid exchanges induced by chemical carcinogens in mouse mammary cells in organ culture. Cell Biol. Int. Rep. 9: 783-789. http://dx.doi.org/10.1016/0309-1651(85)90096-7 Martello MD, David N, Matuo R, Carvalho PC, et al (2016). Campomanesia adamantium extract induces DNA damage, apoptosis, and affects cyclophosphamide metabolism. Genet. Mol. Res. 15. http://dx.doi.org/10.4238/gmr.15027678 Martins GG, Lívero FA, Stolf AM, Kopruszinski CM, et al (2015). Sesquiterpene lactones of Moquiniastrum polymorphum subsp. floccosum have antineoplastic effects in Walker-256 tumor-bearing rats. Chem. Biol. Interact. 228: 46-56. http://dx.doi.org/10.1016/j.cbi.2015.01.018 Mauro MO, Pesarini JR, Marin-Morales MA, Monreal MT, et al (2014). Evaluation of the antimutagenic activity and mode of action of the fructooligosaccharide inulin in the meristematic cells of Allium cepa culture. Genet. Mol. Res. 13: 4808-4819. http://dx.doi.org/10.4238/2014.February.14.14 Moreira AS, Spitzer V, Schapoval EE, Schenkel EP, et al (2000). Antiinflammatory activity of extracts and fractions from the leaves of Gochnatia polymorpha. Phytother. Res. 14: 638-640. http://dx.doi.org/10.1002/1099-1573(200012)14:8<638::AID-PTR681>3.0.CO;2-Q Nantes CI, Pesarini JR, Mauro MO, Monreal ACD, et al (2014). Evaluation of the antimutagenic activity and mode of action of carrageenan fiber in cultured meristematic cells of Allium cepa. Genet. Mol. Res. 13: 9523-9532. http://dx.doi.org/10.4238/2014.November.12.1 Navarro SD, Mauro MO, Pesarini JR, Ogo FM, et al (2015). Resistant starch: a functional food that prevents DNA damage and chemical carcinogenesis. Genet. Mol. Res. 14: 1679-1691. http://dx.doi.org/10.4238/2015.March.6.14 Oliveira RJ, Ribeiro LR, da Silva AF, Matuo R, et al (2006). Evaluation of antimutagenic activity and mechanisms of action of β-glucan from barley, in CHO-k1 and HTC cell lines using the micronucleus test. Toxicol. In Vitro 20: 1225-1233. http://dx.doi.org/10.1016/j.tiv.2006.04.001 Oliveira RJ, Matuo R, da Silva AF, Matiazi HJ, et al (2007). Protective effect of β-glucan extracted from Saccharomyces cerevisiae, against DNA damage and cytotoxicity in wild-type (k1) and repair-deficient (xrs5) CHO cells. Toxicol. In Vitro 21: 41-52. http://dx.doi.org/10.1016/j.tiv.2006.07.018 Oliveira RJ, Fronza LS, Honda RE, Aquino MT, et al (2015). Effects of Avena sativa L. supplementation on ponderal development, reproductive performance and embryo-fetal development of pregnant rats exposed to cyclophosphamide. PECIBES 1: 1-8. Piornedo R dos R, de Souza P, Stefanello MÉ, Strapasson RL, et al (2011). Anti-inflammatory activity of extracts and 11,13-dihydrozaluzanin C from Gochnatia polymorpha ssp. floccosa trunk bark in mice. J. Ethnopharmacol. 133: 1077-1084. http://dx.doi.org/10.1016/j.jep.2010.11.040 Rang HP, Dale MM, Ritter JM, Flower RJ, et al. (2012). Rang & Dale's pharmacology. 7th edn. Churchill Livingstone, Edinburgh. Rank J, Nielsen MH, et al (1997). Allium cepa anaphase-telophase root tip chromosome aberration assay on N-methyl-N-nitrosourea, maleic hydrazide, sodium azide, and ethyl methanesulfonate. Mutat. Res. 390: 121-127. http://dx.doi.org/10.1016/S0165-1218(97)00008-6 Rocha RS, Kassuya CAL, Formagio AS, Mauro M de O, et al (2016). Analysis of the anti-inflammatory and chemopreventive potential and description of the antimutagenic mode of action of the Annona crassiflora methanolic extract. Pharm. Biol. 54: 35-47. http://dx.doi.org/10.3109/13880209.2015.1014567 Roque N (2014). Moquiniastrum. Lista de espécies da flora do Brasil. Jardim Botânico do Rio de Janeiro. Available at [http://floradobrasil.jbrj.gov.br/jabot/floradobrasil/FB130872]. Accessed June 28, 2016. Sacilotto ACB, Vichnewski W, Herz W, et al (1997). Ent-kaurene diterpenes from Gochnatia polymorpha var. polymorpha. Phytochemistry 44: 659-661. http://dx.doi.org/10.1016/S0031-9422(96)00601-2 Schlemper V, Freitas SA, Schlemper SEM, et al (2011). Antispasmodic effects of hydroalcoholic extract from Gochnatia polymorpha ssp. floccosa in the guinea pig ileum. Res. J. Med. Plant 5: 288-294. http://dx.doi.org/10.3923/rjmp.2011.288.294 Schneider BUC, Meza A, Beatriz A, Pesarini JR, et al (2016). Cardanol: toxicogenetic assessment and its effects when combined with cyclophosphamide. Genet. Mol. Biol. 39: 279-289. http://dx.doi.org/10.1590/1678-4685-GMB-2015-0170 Snustad P and Simmons MJ (2013). Fundamentos de genética. 6th edn. Guanabara Koogan, Rio de Janeiro. Strapasson RL, Cervi AC, Carvalho JE, Ruiz AL, et al (2012). Bioactivity-guided isolation of cytotoxic sesquiterpene lactones of Gochnatia polymorpha ssp. floccosa. Phytother. Res. 26: 1053-1056. http://dx.doi.org/10.1002/ptr.3693 Zar HJ, Udwadia ZF, et al (2013). Advances in tuberculosis 2011-2012. Thorax 68: 283-287. http://dx.doi.org/10.1136/thoraxjnl-2012-203127
R. J. Oliveira, de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., Antoniolli-Silva, A. C. M. B., Oliveira, R. J., de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., Antoniolli-Silva, A. C. M. B., Oliveira, R. J., de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., and Antoniolli-Silva, A. C. M. B., Moquiniastrum polymorphum subsp floccosum extract: screening for mutagenic and antimutagenic activity, vol. 15, no. 4, p. -, 2016.
Conflicts of interest The authors declare no conflicts of interest. ACKNOWLEDGMENTS Research supported by FUNDECT (Fundação de Apoio ao Desenvolvimento do Ensino, Ciência e Tecnologia do Estado de Mato Grosso do Sul), CAPES (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior), and CNPq (Conselho Nacional de Desenvolvimento Científico e Tecnológico). REFERENCES Bohlmann F, Zdero C, Schmeda-Hirschmann G, Jakupovic J, et al (1986). Dimeric guainolides and other constituents from Gochnatia species. Phytochemistry 44: 1175-1178. http://dx.doi.org/10.1016/S0031-9422(00)81575-7 Bueno NR, Castilho RO, Costa RB, Pott A, et al (2005). Medicinal plants used by the Kaiowá and Guarani indigenous populations in the Caarapó reserve, Mato Grosso do Sul, Brazil. Acta Bot. Bras. 19: 39-44. http://dx.doi.org/10.1590/S0102-33062005000100005 Cantero WB, Takahachi NA, Mauro MO, Pesarini JR, et al (2015). Genomic lesions and colorectal carcinogenesis: the effects of protein-calorie restriction and inulin supplementation on deficiency statuses. Genet. Mol. Res. 14: 2422-2435. http://dx.doi.org/10.4238/2015.March.27.27 Catalan CAN, Vega MI, Lopez ME, Cuenca M del R, et al (2003). Coumarins and a kaurane from Gochnatia polymorpha ssp. Polymorpha from Paraguay. Biochem. Syst. Ecol. 31: 417-422. http://dx.doi.org/10.1016/S0305-1978(02)00163-1 Cooper EL, et al (2004). Drug discovery, CAM and natural products. Evid. Based Complement. Alternat. Med. 1: 215-217. http://dx.doi.org/10.1093/ecam/neh032 Cragg GM, Newman DJ, et al (2013). Natural products: a continuing source of novel drug leads. Biochim. Biophys. Acta 1830: 3670-3695. http://dx.doi.org/10.1016/j.bbagen.2013.02.008 David Nd, Mauro MdeO, Gonçalves CA, Pesarini JR, et al (2014). Gochnatia polymorpha ssp. floccosa: bioprospecting of an anti-inflammatory phytotherapy for use during pregnancy. J. Ethnopharmacol. 154: 370-379. http://dx.doi.org/10.1016/j.jep.2014.04.005 Farias ACM, Da Silva JR, Tomassini TCB, et al (1984). Constituents of Mochinea polymorpha. J. Nat. Prod. 47: 363-364. http://dx.doi.org/10.1021/np50032a021 Fedel-Miyasato LES, Formagio ASN, Auharek SA, Kassuya CAL, et al (2014a). Antigenotoxic and antimutagenic effects of Schinus terebinthifolius Raddi in Allium cepa and Swiss mice: a comparative study. Genet. Mol. Res. 13: 3411-3425. http://dx.doi.org/10.4238/2014.April.30.2 Fedel-Miyasato LES, Kassuya CAL, Auharek SA, Formagio ASN, et al (2014b). Evaluation of anti-inflammatory, immunomodulatory, chemopreventive and wound healing potentials from Schinus terebinthifolius methanolic extract. Braz. J. Pharmacog. 24: 565-575. Felicidade I, Lima JD, Pesarini JR, Monreal ACD, et al (2014a). Mutagenic and antimutagenic effects of aqueous extract of rosemary (Rosmarinus officinalis L.) on meristematic cells of Allium cepa. Genet. Mol. Res. 13: 9986-9996. http://dx.doi.org/10.4238/2014.November.28.3 Felicidade I, Lima JD, Pesarini JR, Monreal ACD, et al (2014b). Mutagenic and antimutagenic effects of crude hydroalcoholic extract of rosemary (Rosmarinus officinalis L.) on cultured meristematic cells Allium cepa. VRI Phytomedicine 2: 30-39. Fiskesjö G, et al (1993). The Allium test in wastewater monitoring. Environ. Toxicol. 8: 291-298. Leme DM, Marin-Morales MA, et al (2009). Allium cepa test in environmental monitoring: a review on its application. Mutat. Res. 682: 71-81. http://dx.doi.org/10.1016/j.mrrev.2009.06.002 López ME, Giordano OS, López LA, et al (2002). Sesquiterpene lactone dehydroleucodine selectively induces transient arrest in G2 in Allium cepa root meristematic cells. Protoplasma 219: 82-88. http://dx.doi.org/10.1007/s007090200008 Magosso MF, Carvalho PC, Shneider BU, Pessatto LR, et al (2016). Acrocomia aculeata prevents toxicogenetic damage caused by the antitumor agent cyclophosphamide. Genet. Mol. Res. 15. http://dx.doi.org/10.4238/gmr.15027816 Manoharan K, Banerjee MR, et al (1985). beta-Carotene reduces sister chromatid exchanges induced by chemical carcinogens in mouse mammary cells in organ culture. Cell Biol. Int. Rep. 9: 783-789. http://dx.doi.org/10.1016/0309-1651(85)90096-7 Martello MD, David N, Matuo R, Carvalho PC, et al (2016). Campomanesia adamantium extract induces DNA damage, apoptosis, and affects cyclophosphamide metabolism. Genet. Mol. Res. 15. http://dx.doi.org/10.4238/gmr.15027678 Martins GG, Lívero FA, Stolf AM, Kopruszinski CM, et al (2015). Sesquiterpene lactones of Moquiniastrum polymorphum subsp. floccosum have antineoplastic effects in Walker-256 tumor-bearing rats. Chem. Biol. Interact. 228: 46-56. http://dx.doi.org/10.1016/j.cbi.2015.01.018 Mauro MO, Pesarini JR, Marin-Morales MA, Monreal MT, et al (2014). Evaluation of the antimutagenic activity and mode of action of the fructooligosaccharide inulin in the meristematic cells of Allium cepa culture. Genet. Mol. Res. 13: 4808-4819. http://dx.doi.org/10.4238/2014.February.14.14 Moreira AS, Spitzer V, Schapoval EE, Schenkel EP, et al (2000). Antiinflammatory activity of extracts and fractions from the leaves of Gochnatia polymorpha. Phytother. Res. 14: 638-640. http://dx.doi.org/10.1002/1099-1573(200012)14:8<638::AID-PTR681>3.0.CO;2-Q Nantes CI, Pesarini JR, Mauro MO, Monreal ACD, et al (2014). Evaluation of the antimutagenic activity and mode of action of carrageenan fiber in cultured meristematic cells of Allium cepa. Genet. Mol. Res. 13: 9523-9532. http://dx.doi.org/10.4238/2014.November.12.1 Navarro SD, Mauro MO, Pesarini JR, Ogo FM, et al (2015). Resistant starch: a functional food that prevents DNA damage and chemical carcinogenesis. Genet. Mol. Res. 14: 1679-1691. http://dx.doi.org/10.4238/2015.March.6.14 Oliveira RJ, Ribeiro LR, da Silva AF, Matuo R, et al (2006). Evaluation of antimutagenic activity and mechanisms of action of β-glucan from barley, in CHO-k1 and HTC cell lines using the micronucleus test. Toxicol. In Vitro 20: 1225-1233. http://dx.doi.org/10.1016/j.tiv.2006.04.001 Oliveira RJ, Matuo R, da Silva AF, Matiazi HJ, et al (2007). Protective effect of β-glucan extracted from Saccharomyces cerevisiae, against DNA damage and cytotoxicity in wild-type (k1) and repair-deficient (xrs5) CHO cells. Toxicol. In Vitro 21: 41-52. http://dx.doi.org/10.1016/j.tiv.2006.07.018 Oliveira RJ, Fronza LS, Honda RE, Aquino MT, et al (2015). Effects of Avena sativa L. supplementation on ponderal development, reproductive performance and embryo-fetal development of pregnant rats exposed to cyclophosphamide. PECIBES 1: 1-8. Piornedo R dos R, de Souza P, Stefanello MÉ, Strapasson RL, et al (2011). Anti-inflammatory activity of extracts and 11,13-dihydrozaluzanin C from Gochnatia polymorpha ssp. floccosa trunk bark in mice. J. Ethnopharmacol. 133: 1077-1084. http://dx.doi.org/10.1016/j.jep.2010.11.040 Rang HP, Dale MM, Ritter JM, Flower RJ, et al. (2012). Rang & Dale's pharmacology. 7th edn. Churchill Livingstone, Edinburgh. Rank J, Nielsen MH, et al (1997). Allium cepa anaphase-telophase root tip chromosome aberration assay on N-methyl-N-nitrosourea, maleic hydrazide, sodium azide, and ethyl methanesulfonate. Mutat. Res. 390: 121-127. http://dx.doi.org/10.1016/S0165-1218(97)00008-6 Rocha RS, Kassuya CAL, Formagio AS, Mauro M de O, et al (2016). Analysis of the anti-inflammatory and chemopreventive potential and description of the antimutagenic mode of action of the Annona crassiflora methanolic extract. Pharm. Biol. 54: 35-47. http://dx.doi.org/10.3109/13880209.2015.1014567 Roque N (2014). Moquiniastrum. Lista de espécies da flora do Brasil. Jardim Botânico do Rio de Janeiro. Available at [http://floradobrasil.jbrj.gov.br/jabot/floradobrasil/FB130872]. Accessed June 28, 2016. Sacilotto ACB, Vichnewski W, Herz W, et al (1997). Ent-kaurene diterpenes from Gochnatia polymorpha var. polymorpha. Phytochemistry 44: 659-661. http://dx.doi.org/10.1016/S0031-9422(96)00601-2 Schlemper V, Freitas SA, Schlemper SEM, et al (2011). Antispasmodic effects of hydroalcoholic extract from Gochnatia polymorpha ssp. floccosa in the guinea pig ileum. Res. J. Med. Plant 5: 288-294. http://dx.doi.org/10.3923/rjmp.2011.288.294 Schneider BUC, Meza A, Beatriz A, Pesarini JR, et al (2016). Cardanol: toxicogenetic assessment and its effects when combined with cyclophosphamide. Genet. Mol. Biol. 39: 279-289. http://dx.doi.org/10.1590/1678-4685-GMB-2015-0170 Snustad P and Simmons MJ (2013). Fundamentos de genética. 6th edn. Guanabara Koogan, Rio de Janeiro. Strapasson RL, Cervi AC, Carvalho JE, Ruiz AL, et al (2012). Bioactivity-guided isolation of cytotoxic sesquiterpene lactones of Gochnatia polymorpha ssp. floccosa. Phytother. Res. 26: 1053-1056. http://dx.doi.org/10.1002/ptr.3693 Zar HJ, Udwadia ZF, et al (2013). Advances in tuberculosis 2011-2012. Thorax 68: 283-287. http://dx.doi.org/10.1136/thoraxjnl-2012-203127
R. J. Oliveira, de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., Antoniolli-Silva, A. C. M. B., Oliveira, R. J., de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., Antoniolli-Silva, A. C. M. B., Oliveira, R. J., de David, N., Pesarini, J. R., Nogueira, T. D., Kassuya, C. A. L., Strapasson, R. L. B., Stefanello, M. E. A., Monreal, A. C. D., Matuo, R., and Antoniolli-Silva, A. C. M. B., Moquiniastrum polymorphum subsp floccosum extract: screening for mutagenic and antimutagenic activity, vol. 15, no. 4, p. -, 2016.
Conflicts of interest The authors declare no conflicts of interest. ACKNOWLEDGMENTS Research supported by FUNDECT (Fundação de Apoio ao Desenvolvimento do Ensino, Ciência e Tecnologia do Estado de Mato Grosso do Sul), CAPES (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior), and CNPq (Conselho Nacional de Desenvolvimento Científico e Tecnológico). REFERENCES Bohlmann F, Zdero C, Schmeda-Hirschmann G, Jakupovic J, et al (1986). Dimeric guainolides and other constituents from Gochnatia species. Phytochemistry 44: 1175-1178. http://dx.doi.org/10.1016/S0031-9422(00)81575-7 Bueno NR, Castilho RO, Costa RB, Pott A, et al (2005). Medicinal plants used by the Kaiowá and Guarani indigenous populations in the Caarapó reserve, Mato Grosso do Sul, Brazil. Acta Bot. Bras. 19: 39-44. http://dx.doi.org/10.1590/S0102-33062005000100005 Cantero WB, Takahachi NA, Mauro MO, Pesarini JR, et al (2015). Genomic lesions and colorectal carcinogenesis: the effects of protein-calorie restriction and inulin supplementation on deficiency statuses. Genet. Mol. Res. 14: 2422-2435. http://dx.doi.org/10.4238/2015.March.27.27 Catalan CAN, Vega MI, Lopez ME, Cuenca M del R, et al (2003). Coumarins and a kaurane from Gochnatia polymorpha ssp. Polymorpha from Paraguay. Biochem. Syst. Ecol. 31: 417-422. http://dx.doi.org/10.1016/S0305-1978(02)00163-1 Cooper EL, et al (2004). Drug discovery, CAM and natural products. Evid. Based Complement. Alternat. Med. 1: 215-217. http://dx.doi.org/10.1093/ecam/neh032 Cragg GM, Newman DJ, et al (2013). Natural products: a continuing source of novel drug leads. Biochim. Biophys. Acta 1830: 3670-3695. http://dx.doi.org/10.1016/j.bbagen.2013.02.008 David Nd, Mauro MdeO, Gonçalves CA, Pesarini JR, et al (2014). Gochnatia polymorpha ssp. floccosa: bioprospecting of an anti-inflammatory phytotherapy for use during pregnancy. J. Ethnopharmacol. 154: 370-379. http://dx.doi.org/10.1016/j.jep.2014.04.005 Farias ACM, Da Silva JR, Tomassini TCB, et al (1984). Constituents of Mochinea polymorpha. J. Nat. Prod. 47: 363-364. http://dx.doi.org/10.1021/np50032a021 Fedel-Miyasato LES, Formagio ASN, Auharek SA, Kassuya CAL, et al (2014a). Antigenotoxic and antimutagenic effects of Schinus terebinthifolius Raddi in Allium cepa and Swiss mice: a comparative study. Genet. Mol. Res. 13: 3411-3425. http://dx.doi.org/10.4238/2014.April.30.2 Fedel-Miyasato LES, Kassuya CAL, Auharek SA, Formagio ASN, et al (2014b). Evaluation of anti-inflammatory, immunomodulatory, chemopreventive and wound healing potentials from Schinus terebinthifolius methanolic extract. Braz. J. Pharmacog. 24: 565-575. Felicidade I, Lima JD, Pesarini JR, Monreal ACD, et al (2014a). Mutagenic and antimutagenic effects of aqueous extract of rosemary (Rosmarinus officinalis L.) on meristematic cells of Allium cepa. Genet. Mol. Res. 13: 9986-9996. http://dx.doi.org/10.4238/2014.November.28.3 Felicidade I, Lima JD, Pesarini JR, Monreal ACD, et al (2014b). Mutagenic and antimutagenic effects of crude hydroalcoholic extract of rosemary (Rosmarinus officinalis L.) on cultured meristematic cells Allium cepa. VRI Phytomedicine 2: 30-39. Fiskesjö G, et al (1993). The Allium test in wastewater monitoring. Environ. Toxicol. 8: 291-298. Leme DM, Marin-Morales MA, et al (2009). Allium cepa test in environmental monitoring: a review on its application. Mutat. Res. 682: 71-81. http://dx.doi.org/10.1016/j.mrrev.2009.06.002 López ME, Giordano OS, López LA, et al (2002). Sesquiterpene lactone dehydroleucodine selectively induces transient arrest in G2 in Allium cepa root meristematic cells. Protoplasma 219: 82-88. http://dx.doi.org/10.1007/s007090200008 Magosso MF, Carvalho PC, Shneider BU, Pessatto LR, et al (2016). Acrocomia aculeata prevents toxicogenetic damage caused by the antitumor agent cyclophosphamide. Genet. Mol. Res. 15. http://dx.doi.org/10.4238/gmr.15027816 Manoharan K, Banerjee MR, et al (1985). beta-Carotene reduces sister chromatid exchanges induced by chemical carcinogens in mouse mammary cells in organ culture. Cell Biol. Int. Rep. 9: 783-789. http://dx.doi.org/10.1016/0309-1651(85)90096-7 Martello MD, David N, Matuo R, Carvalho PC, et al (2016). Campomanesia adamantium extract induces DNA damage, apoptosis, and affects cyclophosphamide metabolism. Genet. Mol. Res. 15. http://dx.doi.org/10.4238/gmr.15027678 Martins GG, Lívero FA, Stolf AM, Kopruszinski CM, et al (2015). Sesquiterpene lactones of Moquiniastrum polymorphum subsp. floccosum have antineoplastic effects in Walker-256 tumor-bearing rats. Chem. Biol. Interact. 228: 46-56. http://dx.doi.org/10.1016/j.cbi.2015.01.018 Mauro MO, Pesarini JR, Marin-Morales MA, Monreal MT, et al (2014). Evaluation of the antimutagenic activity and mode of action of the fructooligosaccharide inulin in the meristematic cells of Allium cepa culture. Genet. Mol. Res. 13: 4808-4819. http://dx.doi.org/10.4238/2014.February.14.14 Moreira AS, Spitzer V, Schapoval EE, Schenkel EP, et al (2000). Antiinflammatory activity of extracts and fractions from the leaves of Gochnatia polymorpha. Phytother. Res. 14: 638-640. http://dx.doi.org/10.1002/1099-1573(200012)14:8<638::AID-PTR681>3.0.CO;2-Q Nantes CI, Pesarini JR, Mauro MO, Monreal ACD, et al (2014). Evaluation of the antimutagenic activity and mode of action of carrageenan fiber in cultured meristematic cells of Allium cepa. Genet. Mol. Res. 13: 9523-9532. http://dx.doi.org/10.4238/2014.November.12.1 Navarro SD, Mauro MO, Pesarini JR, Ogo FM, et al (2015). Resistant starch: a functional food that prevents DNA damage and chemical carcinogenesis. Genet. Mol. Res. 14: 1679-1691. http://dx.doi.org/10.4238/2015.March.6.14 Oliveira RJ, Ribeiro LR, da Silva AF, Matuo R, et al (2006). Evaluation of antimutagenic activity and mechanisms of action of β-glucan from barley, in CHO-k1 and HTC cell lines using the micronucleus test. Toxicol. In Vitro 20: 1225-1233. http://dx.doi.org/10.1016/j.tiv.2006.04.001 Oliveira RJ, Matuo R, da Silva AF, Matiazi HJ, et al (2007). Protective effect of β-glucan extracted from Saccharomyces cerevisiae, against DNA damage and cytotoxicity in wild-type (k1) and repair-deficient (xrs5) CHO cells. Toxicol. In Vitro 21: 41-52. http://dx.doi.org/10.1016/j.tiv.2006.07.018 Oliveira RJ, Fronza LS, Honda RE, Aquino MT, et al (2015). Effects of Avena sativa L. supplementation on ponderal development, reproductive performance and embryo-fetal development of pregnant rats exposed to cyclophosphamide. PECIBES 1: 1-8. Piornedo R dos R, de Souza P, Stefanello MÉ, Strapasson RL, et al (2011). Anti-inflammatory activity of extracts and 11,13-dihydrozaluzanin C from Gochnatia polymorpha ssp. floccosa trunk bark in mice. J. Ethnopharmacol. 133: 1077-1084. http://dx.doi.org/10.1016/j.jep.2010.11.040 Rang HP, Dale MM, Ritter JM, Flower RJ, et al. (2012). Rang & Dale's pharmacology. 7th edn. Churchill Livingstone, Edinburgh. Rank J, Nielsen MH, et al (1997). Allium cepa anaphase-telophase root tip chromosome aberration assay on N-methyl-N-nitrosourea, maleic hydrazide, sodium azide, and ethyl methanesulfonate. Mutat. Res. 390: 121-127. http://dx.doi.org/10.1016/S0165-1218(97)00008-6 Rocha RS, Kassuya CAL, Formagio AS, Mauro M de O, et al (2016). Analysis of the anti-inflammatory and chemopreventive potential and description of the antimutagenic mode of action of the Annona crassiflora methanolic extract. Pharm. Biol. 54: 35-47. http://dx.doi.org/10.3109/13880209.2015.1014567 Roque N (2014). Moquiniastrum. Lista de espécies da flora do Brasil. Jardim Botânico do Rio de Janeiro. Available at [http://floradobrasil.jbrj.gov.br/jabot/floradobrasil/FB130872]. Accessed June 28, 2016. Sacilotto ACB, Vichnewski W, Herz W, et al (1997). Ent-kaurene diterpenes from Gochnatia polymorpha var. polymorpha. Phytochemistry 44: 659-661. http://dx.doi.org/10.1016/S0031-9422(96)00601-2 Schlemper V, Freitas SA, Schlemper SEM, et al (2011). Antispasmodic effects of hydroalcoholic extract from Gochnatia polymorpha ssp. floccosa in the guinea pig ileum. Res. J. Med. Plant 5: 288-294. http://dx.doi.org/10.3923/rjmp.2011.288.294 Schneider BUC, Meza A, Beatriz A, Pesarini JR, et al (2016). Cardanol: toxicogenetic assessment and its effects when combined with cyclophosphamide. Genet. Mol. Biol. 39: 279-289. http://dx.doi.org/10.1590/1678-4685-GMB-2015-0170 Snustad P and Simmons MJ (2013). Fundamentos de genética. 6th edn. Guanabara Koogan, Rio de Janeiro. Strapasson RL, Cervi AC, Carvalho JE, Ruiz AL, et al (2012). Bioactivity-guided isolation of cytotoxic sesquiterpene lactones of Gochnatia polymorpha ssp. floccosa. Phytother. Res. 26: 1053-1056. http://dx.doi.org/10.1002/ptr.3693 Zar HJ, Udwadia ZF, et al (2013). Advances in tuberculosis 2011-2012. Thorax 68: 283-287. http://dx.doi.org/10.1136/thoraxjnl-2012-203127
H. R. S. Coelho, Berno, C. R., Falcão, G. R., Hildebrand, C. R., Oliveira, R. J., Antoniolli-Silva, A. C. M. B., Coelho, H. R. S., Berno, C. R., Falcão, G. R., Hildebrand, C. R., Oliveira, R. J., and Antoniolli-Silva, A. C. M. B., Twisting of the spermatic cord: ischemia and reperfusion, toxicogenetic evaluation, and the effects of phosphatidylcholine in pre-clinical trials, vol. 15, p. -, 2016.
H. R. S. Coelho, Berno, C. R., Falcão, G. R., Hildebrand, C. R., Oliveira, R. J., Antoniolli-Silva, A. C. M. B., Coelho, H. R. S., Berno, C. R., Falcão, G. R., Hildebrand, C. R., Oliveira, R. J., and Antoniolli-Silva, A. C. M. B., Twisting of the spermatic cord: ischemia and reperfusion, toxicogenetic evaluation, and the effects of phosphatidylcholine in pre-clinical trials, vol. 15, p. -, 2016.
2014
J. R. Pesarini, Victorelli, S. G., Vicentini, A. P., Ferreira, L. K., Mauro, M. O., Matuo, R., Oliveira, J. R., Antoniolli, A. C. M. B., Mantovani, M. S., and Oliveira, R. J., Antigenotoxic and antimutagenic effects of glutamine supplementation on mice treated with cisplatin, vol. 13, pp. 4820-4830, 2014.
L. E. S. Fedel-Miyasato, Formagio, A. S. N., Auharek, S. A., Kassuya, C. A. L., Navarro, S. D., Cunha-Laura, A. L., Monreal, A. C. D., Vieira, M. C., and Oliveira, R. J., Antigenotoxic and antimutagenic effects of Schinus terebinthifolius Raddi in Allium cepa and Swiss mice: A comparative study, vol. 13, pp. 3411-3425, 2014.
A. L. Cunha-Laura, Auharek, S. A., Oliveira, R. J., Siqueira, J. M., Vieira, M. C., Leite, V. S., and Portugal, L. C., Effects of Maytenus ilicifolia on reproduction and embryo-fetal development in Wistar rats, vol. 13, pp. 3711-3720, 2014.
R. J. Oliveira, Pesarini, J. R., Mauro, M. O., Fronza, L. S., Victorelli, S. G., Cantero, W. B., Sena, M. C., and Antoniolli, A. C. M. B., Effects of phenylalanine on reproductive performance and teratogenesis in mice, vol. 13, pp. 5606-5616, 2014.
C. A. Gonçalves, Silva, N. L., Mauro, M. O., David, N., Cunha-Laura, A. L., Auharek, S. A., Monreal, A. C. D., Vieira, M. C., Silva, D. B., Santos, F. J. L., Siqueira, J. M., and Oliveira, R. J., Evaluation of mutagenic, teratogenic, and immunomodulatory effects of Annona nutans hydromethanolic fraction on pregnant mice, vol. 13, pp. 4392-4405, 2014.
C. I. Nantes, Pesarini, J. R., Mauro, M. O., Monreal, A. C. D., Ramires, A. D., and Oliveira, R. J., Evaluation of the antimutagenic activity and mode of action of carrageenan fiber in cultured meristematic cells of Allium cepa, vol. 13, pp. 9523-9532, 2014.
M. O. Mauro, Pesarini, J. R., Marin-Morales, M. A., Monreal, M. T. F. D., Monreal, A. C. D., Mantovani, M. S., and Oliveira, R. J., Evaluation of the antimutagenic activity and mode of action of the fructooligosaccharide inulin in the meristematic cells of Allium cepa culture, vol. 13, pp. 4808-4819, 2014.
I. Felicidade, Lima, J. D., Pesarini, J. R., Monreal, A. C. D., Mantovani, M. S., Ribeiro, L. R., and Oliveira, R. J., Mutagenic and antimutagenic effects of aqueous extract of rosemary (Rosmarinus officinalis L.) on meristematic cells of Allium cepa, vol. 13, pp. 9986-9996, 2014.
M. H. C. Vieira, Oliveira, R. J., Eça, L. P. M., Pereira, I. S. O., Hermeto, L. C., Matuo, R., Fernandes, W. S., Silva, R. A., and Antoniolli, A. C. M. B., Therapeutic potential of mesenchymal stem cells to treat Achilles tendon injuries, vol. 13, pp. 10434-10449, 2014.
2013
J. R. Pesarini, Zaninetti, P. T., Mauro, M. O., Carreira, C. M., Dichi, J. B., Ribeiro, L. R., Mantovani, M. S., and Oliveira, R. J., Antimutagenic and anticarcinogenic effects of wheat bran in vivo, vol. 12, pp. 1646-1659, 2013.
M. O. Mauro, Monreal, M. T. F. D., Silva, M. T. P., Pesarini, J. R., Mantovani, M. S., Ribeiro, L. R., Dichi, J. B., Carreira, C. M., and Oliveira, R. J., Evaluation of the antimutagenic and anticarcinogenic effects of inulin in vivo, vol. 12, pp. 2281-2293, 2013.
A. M. Niwa, Oliveira, R. J., and Mantovani, M. S., Evaluation of the mutagenicity and antimutagenicity of soy phytoestrogens using micronucleus and comet assays of the peripheral blood of mice, vol. 12, pp. 519-527, 2013.
Adlercreutz H, Mazur W, Bartels P, Elomaa V, et al. (2000). Phytoestrogens and prostate disease. J. Nutr. 130: 658S-659S. PMid:10702603   Bedani R and Rossi EA (2005). Isoflavonas: bioquímica, fisiologia e implicações para a saúde. B CEPPA 23: 231-264.   Boersma BJ, Barnes S, Kirk M, Wang CC, et al. (2001). Soy isoflavonoids and cancer - metabolism at the target site. Mutat. Res. 480-481: 121-127. http://dx.doi.org/10.1016/S0027-5107(01)00175-0   Di Virgilio AL, Iwami K, Watjen W, Kahl R, et al. (2004). Genotoxicity of the isoflavones genistein, daidzein and equol in V79 cells. Toxicol. Lett. 151: 151-162. http://dx.doi.org/10.1016/j.toxlet.2004.04.005 PMid:15177650   Djuric Z, Chen G, Doerge DR, Heilbrun LK, et al. (2001). Effect of soy isoflavone supplementation on markers of oxidative stress in men and women. Cancer Lett. 172: 1-6. http://dx.doi.org/10.1016/S0304-3835(01)00627-9   Ebert MN, Beyer-Sehlmeyer G, Liegibel UM, Kautenburger T, et al. (2001). Butyrate induces glutathione S-transferase in human colon cells and protects from genetic damage by 4-hydroxy-2-nonenal. Nutr. Cancer 41: 156-164. PMid:12094619   Esteves EA and Monteiro JBR (2001). Efeitos benéficos das isoflavonas de soja em doenças crônicas. Rev. Nutr. 14: 43-52. http://dx.doi.org/10.1590/S1415-52732001000100007   Fenech M (2000). The in vitro micronucleus technique. Mutat. Res. 455: 81-95. http://dx.doi.org/10.1016/S0027-5107(00)00065-8   Ferguson LR, Philpott M and Karunasinghe N (2004). Dietary cancer and prevention using antimutagens. Toxicology 198: 147-159. http://dx.doi.org/10.1016/j.tox.2004.01.035 PMid:15138038   Ferrari RA and Demiate IM (2001). Isoflavonas de soja: uma breve revisão. Publicativo UEPG: Biol. Health Sci. 7: 39-46.   Fotakis G and Timbrell JA (2006). In vitro cytotoxicity assays: comparison of LDH, neutral red, MTT and protein assay in hepatoma cell lines following exposure to cadmium chloride. Toxicol. Lett. 160: 171-177. http://dx.doi.org/10.1016/j.toxlet.2005.07.001 PMid:16111842   Gontijo AMMC and Tice R (2003). Teste do Cometa para a Detecção de Dano no DNA e Reparo em Células Individualizadas. In: Mutagênese Ambiental (Ribeiro LR, Salvadori DMF and Marques EK, eds.). Ulbra, Canoas, 247-271.   Hayashi M, Morita T, Kodama Y, Sofuni T, et al. (1990). The micronucleus assay with mouse peripheral blood reticulocytes using acridine orange-coated slides. Mutat. Res. 245: 245-249. http://dx.doi.org/10.1016/0165-7992(90)90153-B   Izumi T, Piskula MK, Osawa S, Obata A, et al. (2000). Soy isoflavone aglycones are absorbed faster and in higher amounts than their glucosides in humans. J. Nutr. 130: 1695-1699. PMid:10867038   Klein CB and King AA (2007). Genistein genotoxicity: critical considerations of in vitro exposure dose. Toxicol. Appl. Pharmacol. 224: 1-11. http://dx.doi.org/10.1016/j.taap.2007.06.022 PMid:17688899   Kulling SE, Lehmann L and Metzler M (2002). Oxidative metabolism and genotoxic potential of major isoflavone phytoestrogens. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 777: 211-218. http://dx.doi.org/10.1016/S1570-0232(02)00215-5   Mendes J (2008). Modulação do Efeito Mutagênico por Fitoestrógeno in vitro e in vivo. Master's thesis, UEL, Londrina.   Mendes J, Tsuboy MS, Marcarini JC, Hoffmann-Campo CB, et al. (2012). Modulation of the mutagenic effect of benzo[a] pyrene and bleomycin by isoflavone extracts in a rat hepatome cell line. Semina: Ciênc. Biol. Saúde 33: 11-20.   Michael MR, Wolz E, Davidovich A and Bausch J (2006). Genetic toxicity studies with genistein. Food Chem. Toxicol. 44: 42-55. http://dx.doi.org/10.1016/j.fct.2005.06.004 PMid:16198038   Moon YJ, Wang X and Morris ME (2006). Dietary flavonoids: effects on xenobiotic and carcinogen metabolism. Toxicol. In Vitro 20: 187-210. http://dx.doi.org/10.1016/j.tiv.2005.06.048 PMid:16289744   Peterson G and Barnes S (1991). Genistein inhibition of the growth of human breast cancer cells: independence from estrogen receptors and the multi-drug resistance gene. Biochem. Biophys. Res. Commun. 179: 661-667. http://dx.doi.org/10.1016/0006-291X(91)91423-A   Pool-Zobel B, Veeriah S and Bohmer FD (2005). Modulation of xenobiotic metabolising enzymes by anticarcinogens -- focus on glutathione S-transferases and their role as targets of dietary chemoprevention in colorectal carcinogenesis. Mutat. Res. 591: 74-92. http://dx.doi.org/10.1016/j.mrfmmm.2005.04.020 PMid:16083918   Ribeiro LR and Salvadori DM (2003). Dietary components may prevent mutation-related diseases in humans. Mutat. Res. 544: 195-201. http://dx.doi.org/10.1016/j.mrrev.2003.06.019 PMid:14644321   Setchell KD (2000). Absorption and metabolism of soy isoflavones-from food to dietary supplements and adults to infants. J. Nutr. 130: 654S-655S. PMid:10702601   Singh NP, McCoy MT, Tice RR and Schneider EL (1988). A simple technique for quantitation of low levels of DNA damage in individual cells. Exp. Cell Res. 175: 184-191. http://dx.doi.org/10.1016/0014-4827(88)90265-0   Stopper H, Schmitt E and Kobras K (2005). Genotoxicity of phytoestrogens. Mutat. Res. 574: 139-155. http://dx.doi.org/10.1016/j.mrfmmm.2005.01.029 PMid:15914213
R. J. Oliveira, Sassaki, E. S., Monreal, A. C. D., Monreal, M. T. F. D., Pesarini, J. R., Mauro, M. O., Matuo, R., Silva, A. F., Zobiole, N. N., Siqueira, J. M., Ribeiro, L. R., and Mantovani, M. S., Pre-treatment with glutamine reduces genetic damage due to cancer treatment with cisplatin, vol. 12, pp. 6040-6051, 2013.