Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Research Article

Synthesis of Eugenol Derivatives and Evaluation of their Antifungal Activity Against Fusarium solani f. sp. piperis

Author(s): Sarah C. Maximino, Jessyca A.P. Dutra, Ricardo P. Rodrigues, Rita C.R. Gonçalves, Pedro A.B. Morais, José A. Ventura, Ricardo P. Schuenck, Valdemar Lacerda Júnior, Rodrigo R. Kitagawa and Warley S. Borges*

Volume 26, Issue 14, 2020

Page: [1532 - 1542] Pages: 11

DOI: 10.2174/1381612826666200403120448

Price: $65

Abstract

Background: Fusarium solani f. sp. piperis is a phytopathogen that causes one of the most destructive diseases in black pepper crops, resulting in significant economic and crop production losses. Consequently, the control of this fungal disease is a matter of current and relevant interest in agriculture.

Objective: The objective was to synthesize eugenol derivatives with antifungal activity.

Methods: In this study, using bimolecular nucleophilic substitution and click chemistry approaches, four new and three known eugenol derivatives were obtained. The eugenol derivatives were characterized and their antifungal and cytotoxic effects were evaluated.

Results: Eugenol derivative 4 (2-(4-allyl-2-methoxyphenoxy)-3-chloronaphthalene-1,4-dione) was the most active against F. solani f. sp. piperis and showed acceptable cytotoxicity. Compound 4 was two-fold more effective than tebuconazole in an antifungal assay and presented similar cytotoxicity in macrophages. The in silico study of β-glucosidase suggests a potential interaction of 4 with amino acid residues by a cation-π interaction with residue Arg177 followed by a hydrogen bond with Glu596, indicating an important role in the interactions with 4, justifying the antifungal action of this compound. In addition, the cytotoxicity after metabolism was evaluated as a mimic assay with the S9 fraction in HepG2 cells. Compound 4 demonstrated maintenance of cytotoxicity, showing IC50 values of 11.18 ± 0.5 and 9.04 ± 0.2 μg mL-1 without and with the S9 fraction, respectively. In contrast, eugenol (257.9 ± 0.4 and 133.5 ± 0.8 μg mL-1), tebuconazole (34.94 ± 0.2 and 26.76 ± 0.17 μg mL-1) and especially carbendazim (251.0 ± 0.30 and 34.7 ± 0.10 μg mL-1) showed greater cytotoxicity after hepatic biotransformation.

Conclusion: The results suggest that 4 is a potential candidate for use in the design of new and effective compounds that could control this pathogen.

Keywords: Antifungal, eugenol derivatives, cytotoxicity, Piper nigrum, Fusarium solani f. sp. piperis, macrophages.

[1]
da Silva JK, Silva JR, Nascimento SB, et al. Antifungal activity and computational study of constituents from Piper divaricatum essential oil against Fusarium infection in black pepper. Molecules 2014; 19(11): 17926-42.
[http://dx.doi.org/10.3390/molecules191117926] [PMID: 25375334]
[2]
Joy N, Abraham Z, Soniya EV. A preliminary assessment of genetic relationships among agronomically important cultivars of black pepper. BMC Genet 2007; 8: 42-8.
[http://dx.doi.org/10.1186/1471-2156-8-42] [PMID: 17603884]
[3]
Yogesh MS. Production and export performance of black pepper. Int J Humanit Soc Sci Invent 2013; 2: 36-44.
[4]
Food and Agricultural commodities production: Countries by commodity. Food and Agricultural Organization of the United Nations (FAOSTAT) Available at:. www.fao.org
[5]
Levantamento Sistemático da Produção Agrícola. Instituto Brasileiro de Geografia e Estatística (IBGE) Available from:. www.ibge.org
[6]
Ventura JA, Costa H. Manejo da fusariose da pimenta-do-reino no estado do Espírito-Santo. Incaper 2004; 16: 1-10.
[7]
Rodríguez DJ, Trejo-González FA, Rodríguez-García R, et al. Antifungal activity in vitro of Rhus muelleri against Fusarium oxysporum f. sp. lycopersici. Ind Crops Prod 2015; 75: 150-8.
[http://dx.doi.org/10.1016/j.indcrop.2015.05.048]
[8]
Duarte MLR, Archer SA. In vitro toxin production by Fusarium solani f. sp. piperis. Fitopatol Bras 2005; 28: 229-35.
[http://dx.doi.org/10.1590/S0100-41582003000300002]
[9]
Ventura JA, Milanez D. Fusariose da pimenta-do-reino e seu controle. Empresa Capixaba Pesquisa Agropecuária 1983; 23: 1-17.
[10]
D’Addazio V, Aparecida R, Salomão A, et al. Evaluation of in vitro Fusarium solani f. sp. piperis by different products in Brazil. Afr J Microbiol Res 2016; 10: 1992-8.
[http://dx.doi.org/10.5897/AJMR2016.8292]
[11]
Albuquerque FC, Duarte MLR, Benchimol RL, Endo T. Resistência de piperaceas nativas da Amazônia a infecção causada por Nectria haematococca f. sp. piperis. Acta Amazon 2001; 31: 341-1.
[http://dx.doi.org/10.1590/1809-43922001313348]
[12]
Erisléia-Meireles N, Xavier LP, Ramos AR. Phenylpropanoids produced by Piper divaricatum: a resistant species to infection by Fusarium solani f. sp. piperis, the pathogenic agent of fusariosis in black pepper plant. J Plant Pathol Microbiol 2016; 7: 1-6.
[13]
Spolti P, Del Ponte EM, Dong Y, Cummings JA, Bergstrom GC. Triazole sensitivity in a contemporary population of Fusarium graminearum from New York wheat and competitiveness of a tebuconazole-resistant isolate. Plant Dis 2014; 98(5): 607-13.
[http://dx.doi.org/10.1094/PDIS-10-13-1051-RE] [PMID: 30708550]
[14]
Singh S, Singh N, Kumar V, et al. Toxicity, monitoring and biodegradation of the fungicide carbendazim. Environ Chem Lett 2016; 14: 317-29.
[http://dx.doi.org/10.1007/s10311-016-0566-2]
[15]
Di Renzo F, Bacchetta R, Bizzo A, Giavini E, Menegola E. Is the amphibian X. laevis WEC a good alternative method to rodent WEC teratogenicity assay? The example of the three triazole derivative fungicides Triadimefon, Tebuconazole, Cyproconazole. Reprod Toxicol 2011; 32(2): 220-6.
[http://dx.doi.org/10.1016/j.reprotox.2011.05.001] [PMID: 21601633]
[16]
Ma Y, Xu F, Chen C, Li Q, Wang M. The beneficial use of essential oils from buds and fruit of Syzygium aromaticum to combat pathogenic fungi of Panax notoginseng. Ind Crops Prod 2019; 133: 185-92.
[http://dx.doi.org/10.1016/j.indcrop.2019.03.029]
[17]
Sales MDC, Costa HB, Fernandes PMB, Ventura JA, Meira DD. Antifungal activity of plant extracts with potential to control plant pathogens in pineapple. Asian Pac J Trop Biomed 2016; 6: 26-31.
[http://dx.doi.org/10.1016/j.apjtb.2015.09.026]
[18]
Sharma A, Rajendran S, Srivastava A, Sharma S, Kundu B. Antifungal activities of selected essential oils against Fusarium oxysporum f. sp. lycopersici 1322, with emphasis on Syzygium aromaticum essential oil. J Biosci Bioeng 2017; 123(3): 308-13.
[http://dx.doi.org/10.1016/j.jbiosc.2016.09.011] [PMID: 27876218]
[19]
Xie Y, Yang Z, Cao D, Rong F, Ding H, Zhang D. Antitermitic and antifungal activities of eugenol and its congeners from the flower buds of Syzgium aromaticum (clove). Ind Crops Prod 2015; 77: 780-6.
[http://dx.doi.org/10.1016/j.indcrop.2015.09.044]
[20]
Da Silva FFM, Queiroz José F, Lemos TLG, do Nascimento PGG, Costa AK, de M, Paiva LMM de. Eugenol derivatives: synthesis, characterization, and evaluation of antibacterial and antioxidant activities. Chem Cent J 2018; 12: 1-9.
[http://dx.doi.org/10.1186/s13065-018-0407-4]
[21]
Abrão PHO, Pizi RB, de Souza TB, et al. Synthesis and biological evaluation of new eugenol mannich bases as promising antifungal agents. Chem Biol Drug Des 2015; 86(4): 459-65.
[http://dx.doi.org/10.1111/cbdd.12504] [PMID: 25556966]
[22]
Manganyi MC, Regnier T, Olivier EI. Antimicrobial activities of selected essential oils against Fusarium oxysporum isolates and their biofilms. S Afr J Bot 2015; 99: 115-21.
[http://dx.doi.org/10.1016/j.sajb.2015.03.192]
[23]
Polzin GM, Stanfill SB, Brown CR, Ashley DL, Watson CH. Determination of eugenol, anethole, and coumarin in the mainstream cigarette smoke of Indonesian clove cigarettes. Food Chem Toxicol 2007; 45(10): 1948-53.
[http://dx.doi.org/10.1016/j.fct.2007.04.012] [PMID: 17583404]
[24]
Wenqiang G, Shufen L, Ruixiang Y, Shaokun T, Can Q. Comparison of essential oils of clove buds extracted with supercritical carbon dioxide and other three traditional extraction methods. Food Chem 2007; 101: 1558-64.
[http://dx.doi.org/10.1016/j.foodchem.2006.04.009]
[25]
Brito KB, Francisco CS, Ferreira D, et al. Identifying new isatin derivatives with GSK-3β inhibition capacity trought molecular docking and bioassays. J Braz Chem Soc 2019; 31(3)
[http://dx.doi.org/10.21577/0103-5053.20190206]
[26]
Rahim NHCA, Asari A, Ismail N, Osman H. Synthesis and antibacterial study of eugenol derivatives. Asian J Chem 2017; 29: 22-6.
[http://dx.doi.org/10.14233/ajchem.2017.20100]
[27]
Teixeira RR, Gazolla PAR, da Silva AM, et al. Synthesis and leishmanicidal activity of eugenol derivatives bearing 1,2,3-triazole functionalities. Eur J Med Chem 2018; 146: 274-86.
[http://dx.doi.org/10.1016/j.ejmech.2018.01.046] [PMID: 29407957]
[28]
Clinical and laboratory standards institute reference method for broth dilution antifungal susceptibility testing of filamentous fungi; approved standard M38-A2. 2nd ed. Villanova, PA, USA: National Committee for Clinical Laboratory Standards 2002.
[29]
Xie X, Xu X, Sun C, Yu Z. Down-regulation of KIAP promotes cyclophosphamide-induced apoptosis of hepatoma HEPG2 cells 2017; 10: 4443-9.
[30]
Prakash B, Singh P, Kedia A, Dubey NK. Assessment of some essential oils as food preservatives based on antifungal, antiaflatoxin, antioxidant activities and in vivo efficacy in food system. Food Res Int 2012; 49: 201-8.
[http://dx.doi.org/10.1016/j.foodres.2012.08.020]
[31]
Camacho C, Coulouris G, Avagyan V, et al. BLAST+: architecture and applications. BMC Bioinformatics 2009; 10: 421.
[http://dx.doi.org/10.1186/1471-2105-10-421] [PMID: 20003500]
[32]
Guex N, Peitsch MC, Schwede T. Automated comparative protein structure modeling with SWISS-MODEL and Swiss-PdbViewer: a historical perspective. Electrophoresis 2009; 30(Suppl. 1): S162-73.
[http://dx.doi.org/10.1002/elps.200900140] [PMID: 19517507]
[33]
Benkert P, Biasini M, Schwede T. Toward the estimation of the absolute quality of individual protein structure models. Bioinformatics 2011; 27(3): 343-50.
[http://dx.doi.org/10.1093/bioinformatics/btq662] [PMID: 21134891]
[34]
Jones G, Willett P, Glen RC, Leach AR, Taylor R. Development and validation of a genetic algorithm for flexible docking. J Mol Biol 1997; 267(3): 727-48.
[http://dx.doi.org/10.1006/jmbi.1996.0897] [PMID: 9126849]
[35]
Verdonk ML, Cole JC, Hartshorn MJ, Murray CW, Taylor RD. Improved protein-ligand docking using GOLD. Proteins 2003; 52(4): 609-23.
[http://dx.doi.org/10.1002/prot.10465] [PMID: 12910460]
[36]
Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983; 65(1-2): 55-63.
[http://dx.doi.org/10.1016/0022-1759(83)90303-4] [PMID: 6606682]
[37]
de Cássia Ribeiro Gonçalves R, Rezende Kitagawa R, Aparecida Varanda E, Stella Gonçalves Raddi M, Andrea Leite C, Regina Pombeiro Sponchiado S. Effect of biotransformation by liver S9 enzymes on the mutagenicity and cytotoxicity of melanin extracted from Aspergillus nidulans. Pharm Biol 2016; 54(6): 1014-21.
[http://dx.doi.org/10.3109/13880209.2015.1091846] [PMID: 26459656]
[38]
Choi JM, Oh SJ, Lee JY, et al. Prediction of drug-induced liver injury in hepg2 cells cultured with human liver microsomes. Chem Res Toxicol 2015; 28(5): 872-85.
[http://dx.doi.org/10.1021/tx500504n] [PMID: 25860621]
[39]
Xie Y, Huang Q, Wang Z, Cao H, Zhang D. Structure-activity relationships of cinnamaldehyde and eugenol derivatives against plant pathogenic fungi. Ind Crops Prod 2017; 97: 388-94.
[http://dx.doi.org/10.1016/j.indcrop.2016.12.043]
[40]
Seseni L, Regnier T, Roux-van der Merwe MP, Mogale E, Badenhorst J. Control of Fusarium spp. causing damping-off of pine seedlings by means of selected essential oils. Ind Crops Prod 2015; 76: 329-32.
[http://dx.doi.org/10.1016/j.indcrop.2015.07.002]
[41]
Zore GB, Thakre AD, Jadhav S, Karuppayil SM, Candida A. Terpenoids inhibit Candida albicans growth by affecting membrane integrity and arrest of cell cycle. Phytomedicine 2011; 18(13): 1181-90.
[http://dx.doi.org/10.1016/j.phymed.2011.03.008] [PMID: 21596542]
[42]
Pavela R. Efficacy of naphthoquinones as insecticides against the house fly, Musca domestica L. Ind Crop Prod 13 43: 745-50.
[43]
Sánchez-Calvo JM, Barbero GR, Guerrero-Vásquez G, et al. Synthesis, antibacterial and antifungal activities of naphthoquinone derivatives: a structure–activity relationship study. Med Chem Res 2016; 25: 1274-85.
[http://dx.doi.org/10.1007/s00044-016-1550-x]
[44]
Anaissi-Afonso L, Oramas-Royo S, Ayra-Plasencia J, et al. Lawsone, Juglone, and β-Lapachone derivatives with enhanced mitochondrial-based toxicity. ACS Chem Biol 2018; 13(8): 1950-7.
[http://dx.doi.org/10.1021/acschembio.8b00306] [PMID: 29878754]
[45]
Leyva E, López LI, de la Cruz RFG, Espinosa-González CG. Synthesis and studies of the antifungal activity of 2-anilino-/2,3-dianilino-/2-phenoxy- and 2,3-diphenoxy-1,4-naphthoquinones. Res Chem Intermed 2017; 43: 1813-27.
[http://dx.doi.org/10.1007/s11164-016-2732-3]
[46]
Nascimento SB, Cascardo JC, Menezes IC, et al. Identifying sequences potentially related to resistance response of Piper tuberculatum to Fusarium solani f. sp. piperis by suppression subtractive hybridization. Protein Pept Lett 2009; 16(12): 1429-34.
[http://dx.doi.org/10.2174/092986609789839368] [PMID: 20001904]
[47]
de Souza CRB, Brígida ABS, dos Santos RC, de Monteiro Costa CN, Darnet SH, Harada ML. Identification of sequences expressed during compatible black pepper-Fusarium solani f. sp. piperis interaction. Acta Physiol Plant 2011; 33: 2553-60.
[http://dx.doi.org/10.1007/s11738-011-0788-7]
[48]
Yoshida E, Hidaka M, Fushinobu S, et al. Purification, crystallization and preliminary X-ray analysis of β-glucosidase from Kluyveromyces marxianus NBRC1777. Acta Crystallogr Sect F Struct Biol Cryst Commun 2009; 65(Pt 11): 1190-2.
[http://dx.doi.org/10.1107/S1744309109042948] [PMID: 19923748]
[49]
Kuiper HA. The role of toxicology in the evaluation of new agrochemicals. J Environ Sci Health B 1996; 31(3): 353-63.
[http://dx.doi.org/10.1080/03601239609372996] [PMID: 8642181]
[50]
Kitagawa RR, Raddi MSG, Santos LC, Vilegas W. A new cytotoxic naphthoquinone from Paepalanthus latipes. Chem Pharm Bull (Tokyo) 2004; 52(12): 1487-8.
[http://dx.doi.org/10.1248/cpb.52.1487] [PMID: 15577252]
[51]
Kitagawa RR, da Fonseca LM, Ximenes VF, Khalil NM, Vilegas W, Raddi MS. Ascorbic acid potentiates the cytotoxicity of the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin. Phytochemistry 2008; 69(11): 2205-8.
[http://dx.doi.org/10.1016/j.phytochem.2008.05.007] [PMID: 18617198]
[52]
Sousa ET, Lopes WA, Andrade JB. Fontes, formação, reatividade e determinação de quinonas na atmosfera. Quim Nova 2016; 39: 486-95.
[53]
Abdel Bar FM, Khanfar MA, Elnagar AY, et al. Design and pharmacophore modeling of biaryl methyl eugenol analogs as breast cancer invasion inhibitors. Bioorg Med Chem 2010; 18(2): 496-507.
[http://dx.doi.org/10.1016/j.bmc.2009.12.019] [PMID: 20034800]
[54]
Richardson SJ, Bai A, Kulkarni AA, Moghaddam MF. Efficiency in drug discovery: liver S9 fraction assay as a screen for metabolic stability. Drug Metab Lett 2016; 10(2): 83-90.
[http://dx.doi.org/10.2174/1872312810666160223121836] [PMID: 26902079]
[55]
Brandon EFA, Raap CD, Meijerman I, Beijnen JH, Schellens JHM. An update on in vitro test methods in human hepatic drug biotransformation research: pros and cons. Toxicol Appl Pharmacol 2003; 189(3): 233-46.
[http://dx.doi.org/10.1016/S0041-008X(03)00128-5] [PMID: 12791308]
[56]
Kitagawa RR, Vilegas W, Varanda EA, Raddi MSG. Evaluation of mutagenicity and metabolism-mediated cytotoxicity of the naphthoquinone 5-methoxy-3,4-dehydroxanthomegnin from Paepalanthus latipes. Brazilian J Pharmacogn 2015; 25: 16-21.
[http://dx.doi.org/10.1016/j.bjp.2014.12.001]
[57]
Babich H, Stern A, Borenfreund E. Eugenol cytotoxicity evaluated with continuous cell lines. Toxicol In Vitro 1993; 7(2): 105-9.
[http://dx.doi.org/10.1016/0887-2333(93)90119-P] [PMID: 20732177]
[58]
Burkey JL, Sauer JM, McQueen CA, Sipes IG. Cytotoxicity and genotoxicity of methyleugenol and related congeners-- a mechanism of activation for methyleugenol. Mutat Res 2000; 453(1): 25-33.
[http://dx.doi.org/10.1016/S0027-5107(00)00070-1] [PMID: 11006409]
[59]
Maralhas A, Monteiro A, Martins C, et al. Genotoxicity and endoreduplication inducing activity of the food flavouring eugenol. Mutagenesis 2006; 21(3): 199-204.
[http://dx.doi.org/10.1093/mutage/gel017] [PMID: 16595588]
[60]
Cuendet M, Oteham CP, Moon RC, Pezzuto JM. Quinone reductase induction as a biomarker for cancer chemoprevention. J Nat Prod 2006; 69(3): 460-3.
[http://dx.doi.org/10.1021/np050362q] [PMID: 16562858]
[61]
Knebel C, Heise T, Zanger UM, Lampen A, Marx-Stoelting P, Braeuning A. The azole fungicide tebuconazole affects human CYP1A1 and CYP1A2 expression by an aryl hydrocarbon receptor-dependent pathway. Food Chem Toxicol 2019; 123: 481-91.
[http://dx.doi.org/10.1016/j.fct.2018.11.039] [PMID: 30458266]
[62]
Schmidt F, Marx-Stoelting P, Haider W, et al. Combination effects of azole fungicides in male rats in a broad dose range. Toxicology 2016; 355-356: 54-63.
[http://dx.doi.org/10.1016/j.tox.2016.05.018] [PMID: 27234313]
[63]
Schmidt G, Piponov H, Chuang D, Gonzalez M. Hand infections in the immunocompromised patient: an update. J Hand Surg Am 2019; 44(2): 144-9.
[http://dx.doi.org/10.1016/j.jhsa.2018.07.001] [PMID: 30145028]
[64]
Roviello GN, Roviello V, Musumeci D, Pedone C. Synthesis of a novel benzodifuran derivative and its molecular recognition of poly rA RNA. Biol Chem 2013; 394(9): 1235-9.
[http://dx.doi.org/10.1515/hsz-2013-0154] [PMID: 23729622]
[65]
Carella A, Roviello V, Iannitti R, et al. Evaluating the biological properties of synthetic 4-nitrophenyl functionalized benzofuran derivatives with telomeric DNA binding and antiproliferative activities. Int J Biol Macromol 2019; 121: 77-88.
[http://dx.doi.org/10.1016/j.ijbiomac.2018.09.153] [PMID: 30261256]
[66]
Roviello GN, Moccia M, Sapio R, et al. Synthesis, characterization and hybridization studies of new nucleo-γ-peptides based on diaminobutyric acid. J Pept Sci 2006; 12(12): 829-35.
[http://dx.doi.org/10.1002/psc.819] [PMID: 17131297]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy