DOI QR코드

DOI QR Code

A Review on Structure, Modifications and Structure-Activity Relation of Quercetin and Its Derivatives

  • Magar, Rubin Thapa (Department of Life Science and Biochemical Engineering, SunMoon University) ;
  • Sohng, Jae Kyung (Department of Life Science and Biochemical Engineering, SunMoon University)
  • Received : 2019.07.01
  • Accepted : 2019.11.08
  • Published : 2020.01.28

Abstract

Quercetin and its derivatives are important metabolites that belong to the flavonol class of flavonoids. Quercetin and some of the conjugates have been approved by the FDA for human use. They are widely distributed among plants and have various biological activities, such as being anticancer, antiviral, and antioxidant. Hence, the biosynthesis of novel derivatives is an important field of research. Glycosylation and methylation are two important modification strategies that have long been used and have resulted in many novel metabolites that are not present in natural sources. A strategy for modifying quercetin in E. coli by means of glycosylation, for example, involves overexpressing respective glycosyltransferases (GTs) in the host and metabolic engineering for increasing nucleoside diphosphate sugar (NDP-sugar). Still others have used microorganisms other than E. coli, such as Streptomyces sp., for the biotransformation process. The overall study of the structural activity relationship has revealed that modification of some residues in quercetin decreased one activity but increased others. This review summarizes all of the information mentioned above.

Keywords

References

  1. Nemeth K, Piskula MK. 2007. Food content, processing, absorption and metabolism of onion flavonoids. Crit. Rev. Food Sci. Nutr. 47: 397-409. https://doi.org/10.1080/10408390600846291
  2. Berardini N, Fezer R, Conrad J, Beifuss U, Carle R, Schieber A. 2005. Screening of mango (Mangifera indica L.) cultivars for their contents of flavonol O- and xanthone C-glycosides, anthocyanins and pectin. J. Agric. Food Chem. 53: 1563-1570. https://doi.org/10.1021/jf0484069
  3. Ramal ho SD, d e Sousa LR, Burger MC, L ima MI, da S il va MF, Fernandes JB, et al. 2015. Evaluation of flavonols and derivatives as human cathepsin B inhibitor. Nat. Prod. Res. 29: 2212-2214. https://doi.org/10.1080/14786419.2014.1002404
  4. Qiu X, Kroeker A, He S, Kozak R, Audet J, Mbikay M, Chretien M. 2016. Prophylactic efficacy of quercetin 3-${\beta}$-OD-glucoside against Ebola virus infection. Antimicrob. Agents Chemother. 60: 5182-5188. https://doi.org/10.1128/AAC.00307-16
  5. Lee S, Sy S, Lee Y, Park Y, Kim BG, Ahn JH, et al. 2011. Rhamnetin production based on the rational design of the poplar O-methyltransferase enzyme and its biological activities. Bioorg. Med. Chem. Lett. 21: 3866-3870. https://doi.org/10.1016/j.bmcl.2011.05.043
  6. Makino T, Shimizu R, Kanemaru M, Suzuki Y, Moriwaki M, Mizukami H. 2009. Enzymatically modified isoquercitrin, alpha-oligoglucosyl quercetin 3-O-glucoside, is absorbed more easily than other quercetin glycosides or aglycone after oral administration in rats. Biol. Pharm. Bull. 32: 2034-2040. https://doi.org/10.1248/bpb.32.2034
  7. Darsandhari S, Bae JY, Shrestha B,Yamaguchi T, Jung HJ, Han JM, et al. 2018. Enzymatic synthesis of novel quercetin sialyllactoside derivatives. Nat. Prod. Res. 6: 1-9.
  8. Lee YS, Huh JY, Nam SH, Kim D, Lee SB. 2013. Synthesis of quercetin-3-O-glucoside from rutin by Penicillium decumbens naringinase. J. Food Sci. 78: C411-415. https://doi.org/10.1111/1750-3841.12056
  9. de Araujo ME, Moreira Franco YE, Alberto TG, Sobreiro MA, Conrado MA, Priolli DG, et al. 2012. Enzymatic deglycosylation of rutin improves its antioxidant and antiproliferative activities. Food Chem. 141: 266-273. https://doi.org/10.1016/j.foodchem.2013.02.127
  10. Lee YS, Woo JB, Ryu SI, Moon SK, Han NS, Lee SB. 2017. Glucosylation of flavonol and flavanones by Bacillus cyclodextrin glucosyltransferase to enhance their solubility and stability. Food Chem. 229: 75-83. https://doi.org/10.1016/j.foodchem.2017.02.057
  11. Rha CS, Choi JM, Jung YS, Kim ER, Ko MJ, Seo DH, et al. 2019. High-efficiency enzymatic production of ${\alpha}$-isoquercitrin glucosides by amylosucrase from Deinococcus geothermalis. Enzym. Microb. Technol. 120: 84-90. https://doi.org/10.1016/j.enzmictec.2018.10.006
  12. Kim JH, Shin KH, Ko JH, Ahn JH. 2006. Glucosylation of flavonols by Escherichia coli expressing glucosyltransferase from rice (Oryza sativa). J. Biosci. Bioeng. 102: 135-137. https://doi.org/10.1263/jbb.102.135
  13. Pandey RP, Malla S, Simkhada D, Kim BG, Sohng JK. 2013. Production of 3-O-xylosyl quercetin in Escherichia coli. Appl. Microbiol. Biotechnol. 97: 1889-1901. https://doi.org/10.1007/s00253-012-4438-9
  14. Yoon JA, Kim BG, Lee WJ, Lim Y, Chong Y, Ahn JH. 2002. Production of a novel quercetin glycoside through metabolic engineering of Escherichia coli. Appl. Environ. Microbiol. 78: 4256-4262. https://doi.org/10.1128/AEM.00275-12
  15. An DG, Yang SM, Kim BG, Ahn JH. 2016. Biosynthesis of two quercetin O-diglycosides in Escherichia coli. J. Ind. Microbiol. Biotechnol. 43: 841-849. https://doi.org/10.1007/s10295-016-1750-x
  16. Choi GS, Kim HJ, Kim EJ, Lee SJ, Lee Y, Ahn JH. 2018. Stepwise synthesis of quercetin bisglycosides using engineered Escherichia coli. J. Microbiol. Biotechnol. 28: 1859-1864. https://doi.org/10.4014/jmb.1807.07043
  17. Han SH, Kim BG, Yoon JA, Chong Y, Ahn JH. 2014. Synthesis of flavonoid O-pentosides by Escherichia coli through engineering of nucleotide sugar synthesis pathways and glycosyltransferase. Appl. Environ. Microbiol. 80: 2754-2762. https://doi.org/10.1128/AEM.03797-13
  18. Kim SY, Lee HR, Park KS, Kim BG, Ahn JH. 2015. Metabolic engineering of Escherichia coli for the biosynthesis of flavonoid O-glucuronides and flavonoid O-galactoside. Appl. Microbiol. Biotechnol. 99: 2233-2242. https://doi.org/10.1007/s00253-014-6282-6
  19. Pandey RP, Jung HY, Parajuli P, Nguyen THT, Bashyal P, Sohng JK. 2019. A synthetic approach for biosynthesis of miquelianin and scutellarin A in Escherichia coli. Appl. Sci. 9: 215. https://doi.org/10.3390/app9020215
  20. Kim BG, Shin KH, Lee Y, Hur HG, Lim Y, Ahn JH. 2005. Multiple regiospecific methylations of a flavonoid by plant O-methyltransferases expressed in E. coli. Biotechnol. Lett. 27: 1861-1864. https://doi.org/10.1007/s10529-005-3893-0
  21. Darsandhari S, Dhakal D, Shrestha B, Parajuli P, Seo JH, Kim TS, et al. 2018. Characterization of regioselective flavonoid O-methyltransferase from the Streptomyces sp. KCTC 0041BP. Enzym. Microb. Technol. 113: 29-36. https://doi.org/10.1016/j.enzmictec.2018.02.007
  22. Xu JQ, Fan N, Yu BY, Wang QQ, Zhang J. 2017. Biotransformation of quercetin by Gliocladium deliquescens NRRL 1086. Chin. J. Nat. Med. 15: 615-624. https://doi.org/10.1016/S1875-5364(17)30089-4
  23. Marvalina C, Azerada R. 2011. Microbial glucuronidation of polyphenols. J. Mol. Catal. B Enzym. 73: 43-52. https://doi.org/10.1016/j.molcatb.2011.07.015
  24. Ma B, Zeng J, Shao L, Zhan J. 2013. Efficient bioconversion of quercetin into a novel glycoside by Streptomyces rimosus subsp. rimosus ATCC 10970. J. Biosci. Bioeng. 115: 24-26. https://doi.org/10.1016/j.jbiosc.2012.07.020
  25. Hosny M, Dhar K, Rosazza JPN. 2001. Hydroxylations and methylations of quercetin, fisetin, and catechin by Streptomyces griseus. J. Nat. Prod. 64: 462-465. https://doi.org/10.1021/np000457m
  26. de MB Costa EM, Pimenta FC, Luz WC, de Ol iveira V. 2008. Selection of filamentous fungi of the Beauveriagenus able to metabolize quercetin like mammalian cells. Braz. J. Microbiol. 39: 405-408. https://doi.org/10.1590/S1517-83822008000200036
  27. Lesjak M, Beara I, Simin I, Pintac D, Majkic T, Bekvalac K, et al. 2018. Antioxidant and anti-inflammatory activities of quercetin and its derivatives. J. Funct. Foods. 40: 68-75. https://doi.org/10.1016/j.jff.2017.10.047
  28. Rice-Evans CA, Miller NJ, Paganga G. 1996. Structureantioxidant activity relationships of flavonoids and phenolic acids. Free Radic. Biol. Med. 20: 933-956. https://doi.org/10.1016/0891-5849(95)02227-9
  29. Santos AC, Uyemura SA, Lopes JL, Bazon JN, Mingatto FE, Curti C. 1998. Effect of naturally occurring flavonoids on lipid peroxidation and membrane permeability transition in mitochondria. Free Radic. Biol. Med. 24: 1455-1461. https://doi.org/10.1016/S0891-5849(98)00003-3
  30. Lee CW, Seo JY, Lee J, Choi JW, Cho S, Bae JY, et al. 2017. 3-O-Glucosylation of quercetin enhances inhibitory effects on the adipocyte differentiation and lipogenesis. Biomed. Pharmacother. 95: 589-598. https://doi.org/10.1016/j.biopha.2017.08.002

Cited by

  1. Global Gene Expression Profiling Reveals Isorhamnetin Induces Hepatic-Lineage Specific Differentiation in Human Amniotic Epithelial Cells vol.8, 2020, https://doi.org/10.3389/fcell.2020.578036
  2. The Pharmacological Activity, Biochemical Properties, and Pharmacokinetics of the Major Natural Polyphenolic Flavonoid: Quercetin vol.9, pp.3, 2020, https://doi.org/10.3390/foods9030374
  3. Narrative review of Mediterranean diet in Cilento: longevity and potential prevention for prostate cancer vol.13, 2020, https://doi.org/10.1177/17562872211026404
  4. Iron Complexes of Flavonoids-Antioxidant Capacity and Beyond vol.22, pp.2, 2020, https://doi.org/10.3390/ijms22020646
  5. SIRT5-Related Desuccinylation Modification Contributes to Quercetin-Induced Protection against Heart Failure and High-Glucose-Prompted Cardiomyocytes Injured through Regulation of Mitochondrial Qualit vol.2021, 2021, https://doi.org/10.1155/2021/5876841
  6. Human Sirtuin Regulators: The “Success” Stories vol.12, 2020, https://doi.org/10.3389/fphys.2021.752117
  7. Therapeutic Potential of Quercetin to Alleviate Endothelial Dysfunction in Age-Related Cardiovascular Diseases vol.8, 2021, https://doi.org/10.3389/fcvm.2021.658400
  8. Rutin: A Potential Antiviral for Repurposing as a SARS-CoV-2 Main Protease (Mpro) Inhibitor vol.16, pp.4, 2020, https://doi.org/10.1177/1934578x21991723
  9. Rutin Is a Low Micromolar Inhibitor of SARS-CoV-2 Main Protease 3CLpro: Implications for Drug Design of Quercetin Analogs vol.9, pp.4, 2020, https://doi.org/10.3390/biomedicines9040375
  10. Quercetin-3-O-glucuronide in the Ethanol Extract of Lotus Leaf (Nelumbo nucifera) Enhances Sleep Quantity and Quality in a Rodent Model via a GABAergic Mechanism vol.26, pp.10, 2020, https://doi.org/10.3390/molecules26103023
  11. Selective β-Mono-Glycosylation of a C15-Hydroxylated Metabolite of the Agricultural Herbicide Cinmethylin Using Leloir Glycosyltransferases vol.69, pp.19, 2021, https://doi.org/10.1021/acs.jafc.1c01321
  12. Rutin and Quercetin Decrease Cholesterol in HepG2 Cells but Not Plasma Cholesterol in Hamsters by Oral Administration vol.26, pp.12, 2020, https://doi.org/10.3390/molecules26123766
  13. Quercetin against MCF7 and CAL51 breast cancer cell lines: apoptosis, gene expression and cytotoxicity of nano-quercetin vol.16, pp.22, 2020, https://doi.org/10.2217/nnm-2021-0070
  14. 2-Hydroxy-5-(3,5,7-trihydroxy-4-oxo-4H-chromen-2-yl)phenyl (E)-3-(4-hydroxy-3-methoxyphenyl)acrylate: Synthesis, In Silico Analysis and In Vitro Pharmacological Evaluation vol.2021, pp.3, 2020, https://doi.org/10.3390/m1258
  15. Food flavonols: Nutraceuticals with complex health benefits and functionalities vol.117, 2020, https://doi.org/10.1016/j.tifs.2021.03.030
  16. Neuroinflammation as a Therapeutic Target in Retinitis Pigmentosa and Quercetin as Its Potential Modulator vol.13, pp.11, 2021, https://doi.org/10.3390/pharmaceutics13111935
  17. Phytochemical profiles of edible flowers of medicinal plants of Dendrobium officinale and Dendrobium devonianum vol.9, pp.12, 2020, https://doi.org/10.1002/fsn3.2602
  18. Biosynthesis of a rosavin natural product in Escherichia coli by glycosyltransferase rational design and artificial pathway construction vol.69, 2020, https://doi.org/10.1016/j.ymben.2021.10.010