Insights into the Pharmacological Effects of Flavonoids: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Roman P. Terekhov.

Flavonoids are widely occurring secondary metabolites of plants. Currently, there is a trend of article numbers increasing, which focuses on the computer modeling of flavonoid interactions with biological targets. Such studies help to accumulatethe data on lead compounds that can find medicinal implementation, including COVID-19. Flavanonol taxifolin demonstrated wound-healing activity. Luteolin, apigenin, and wogonin, which can be classified as flavones, show induced neutrophil apoptosis and have potential as neutrophil apoptosis-inducing anti-inflammatory, proresolution agents.

  • flavonoids
  • phytomedicine
  • taxifolin
  • molecular modeling
  • COVID-19

1. Structure—Biological Activity Relationship: Qualitative Analysis

The parent structure of flavonoids is 1,3-diphenylpropane, and the aromatic fragments are designated as ring A and ring B [67][1]. The majority of flavonoid groups are characterized by the heterocycle (ring C) containing oxygen. This ring may be aromatic (flavones, flavonols, etc.) or not (flavanones, flavanonols, etc.). As the rule, carbonyl and several hydroxyl functional groups are present in the molecular structure of flavonoids that can act as a pharmacophores.
The phenolic hydroxyl groups of the studied natural compounds serve as H-bond donors. In cases when the hydrophobic interactions play a key role, the presence of the methoxy group leads to an increase of affinity to the target compared with the hydroxyl group [68][2]. Due to aromatic rings, the π,π-interactions with the side residues of heterocyclic and aromatic α-amino acids (tryptophan, histidine, phenylalanine, and tyrosine) are possible [69][3]Figure 1 demonstrates all types of interactions.
Figure 1.
 Interaction of taxifolin and P-glycoprotein.
It was found that the antiangiogenic potential of the flavonoid depends on the presence of a C2-C3 double bond [70][4]; the hydroxyl group in the position 3′ of the ring C contributes to an increase in antioxidant, anti-inflammatory, and antitumor activity [71][5]. If, along with the multiple C2-C3 bonds, a catechol group is present in the ring B, then such a molecule demonstrates a high affinity for the angiotensin-converting enzyme [72][6]. Substituents 3-OH, 5-OH, 6-OMe, 6-OH, 7-OH, 3′-OH, and 4′-OMe were identified as key fragments of the molecules when interacting with multidrug resistance-associated protein 2 (MRP2) [55][7].
It was also interesting to determine the specificity of the interaction of flavonoid groups. Thus, flavones (6-hydroxyluteolin, scutellarein), flavonols (kaempferol), and flavanones (naringenin, eridioctyol) exhibit a high affinity to the estrogen receptor α (ERα), which has been proven in both AutoDock and Glide software. Representatives of these groups of flavonoids can be recommended in the development of antitumor drugs for the treatment of breast cancer [71,73][5][8]. Interaction with this protein target results in several types of patient management, such as estrogen hormone replacement therapy and preventive care for breast cancer [74][9]. Flavones (baicalein, ladanein), flavonols (quercetin), and their glycosylated forms (baicalin) interact with the E protein of various strains of the dengue virus causing fever with a similar name [54][10]. Such ligands may be used in the treatment of this disease [75][11]. It is worth noting that the width of the confidence interval of the scoring function calculated for flavones is quite large. This indicates a different degree of protein-ligand binding within this group. Flavones (5-hydroxyflavone) and flavonols (quercetin) have a high affinity for the potassium channel Kir6.1, acting on which some cardiovascular diseases can be treated [56][12]. Flavones (luteolin, apigenin) can serve as the basis of drugs that control the pathogenicity of Helicobacter pylori due to their ability to bind to one of the main virulence factors of bacteria of this species—vacuolating cytotoxin protein (VacA) [76][13]. Flavonols (quercetin), their glycosides (avicularin, hyperoside), and flavanonols (taxifolin) with comparable effects function as arginase inhibitors, which is a potential target for the development of new approaches to the treatment of leishmaniasis [77][14]. Flavan-3-ols (catechin, epicatechin) are characterized by the best values of the scoring function when binding to the CA II-F complex in comparison with flavones, flavanones, and flavanonols and are of interest for the treatment of fluorosis [78][15]. According to the silico results, flavanones (eriodictyol) and flavanonols (taxifolin) are able to inhibit transcription factors Tec1 and Rfg1 because they can be used in the treatment of infection caused by Candida albicans fungus [79][16].

2. Structure—Biological Activity Relationship: Quantitative Analysis

Meta-analyses of scoring functions calculated during molecular docking was studied in [54,56,71,73,76,77,78,79][5][8][10][12][13][14][15][16]. General information about the average affinities of each flavonoid group to the biological targets is presented in Table 1 and Table 2 for AutoDock and Glide software, respectively.
Table 1.
 Comparison of the average affinity of flavonoid groups to target proteins in the AutoDock.

References

  1. Kurkin, V.A. Phenylpropanoids from medicinal plants: Distribution, classification, structural analysis, and biological activity. Chem. Nat. Compd 2003, 39, 123–153.
  2. Shanmugasundaram, J.; Subramanian, V.; Nadipelly, J.; Kathirvelu, P.; Sayeli, V.; Cheriyan, B.V. Anxiolytic–like activity of 5–methoxyflavone in mice with involvement of GABAergic and serotonergic systems—In vivo and in silico evidences. Eur. Neuropsychopharmacol. 2020, 36, 100–110.
  3. Taldaev, A.K.; Terekhov, R.P.; Selivanova, I.A. Interaction of Taxifolin and P-Glycoprotein in silico. In Book of Abstracts XI International Conference on Chemistry for Young Scientists; Izdatelstvo VVM: Moscow, Russia, 2019; p. 436.
  4. Gacche, R.N.; Shegokar, H.D.; Gond, D.S.; Yang, Z.; Jadhav, A.D. Evaluation of selected flavonoids as antiangiogenic, anticancer, and radical scavenging agents: An experimental and in silico analysis. Cell Biochem. Biophys. 2011, 61, 651–663.
  5. Yugandhar, P.; Kumar, K.K.; Neeraja, P.; Savithramma, N. Isolation, characterization and in silico docking studies of synergistic estrogen receptor a anticancer polyphenols from Syzygium alternifolium (Wt.) Walp. J. Intercult. Ethnopharmacol. 2017, 6, 296.
  6. Guerrero, L.; Castillo, J.; Quiñones, M.; Garcia-Vallvé, S.; Arola, L.; Pujadas, G.; Muguerza, B. Inhibition of angiotensin-converting enzyme activity by flavonoids: Structure-activity relationship studies. PLoS ONE 2012, 7, e49493.
  7. Fang, Y.; Cao, W.; Liang, F.; Xia, M.; Pan, S.; Xu, X. Structure affinity relationship and docking studies of flavonoids as substrates of multidrug-resistant associated protein 2 (MRP2) in MDCK/MRP2 cells. Food Chem. 2019, 291, 101–109.
  8. Ye, H.; Shaw, I.C. Food flavonoid ligand structure/estrogen receptor-α affinity relationships—Toxicity or food functionality? Food Chem. Toxicol. 2019, 129, 328–336.
  9. Katzenellenbogen, B.S.; Choi, I.; Delage-Mourroux, R.; Ediger, T.R.; Martini, P.G.V.; Montano, M.; Sun, J.; Weis, K.; Katzenellenbogen, J.A. Molecular mechanisms of estrogen action: Selective ligands and receptor pharmacology. J. Steroid Biochem. Mol. Biol. 2000, 74, 279–285.
  10. Ismail, N.A.; Jusoh, S.A. Molecular docking and molecular dynamics simulation studies to predict flavonoid binding on the surface of DENV2 E protein. Interdiscip. Sci. Comput. Life Sci. 2016, 9, 499–511.
  11. Anasir, M.I.; Ramanathan, B.; Poh, C.L. Structure-based design of antivirals against envelope glycoprotein of dengue virus. Viruses 2020, 12, 367.
  12. Trezza, A.; Cicaloni, V.; Porciatti, P.; Langella, A.; Fusi, F.; Saponara, S.; Spiga, O. From in silico to in vitro: A trip to reveal flavonoid binding on the Rattus norvegicus Kir6.1 ATP-sensitive inward rectifier potassium channel. PeerJ 2018, 6, e4680.
  13. Jouimyi, M.R.; Bounder, G.; Essaidi, I.; Boura, H.; Zerouali, K.; Lebrazi, H.; Kettani, A.; Maachi, F. Molecular docking of a set of flavonoid compounds with Helicobacter pylori virulence factors CagA and VacA. J. Herbmed. Pharmacol. 2020, 9, 412–419.
  14. Da Silva, E.R.; Brogi, S.; Lucon-Júnior, J.F.; Campiani, G.; Gemma, S.; Maquiaveli, C.D.C. Dietary polyphenols rutin, taxifolin and quercetin related compounds target Leishmania amazonensis arginase. Food Funct. 2019, 10, 3172–3180.
  15. Validandi, V.; Khandare, A.L. Reduction of fluoride toxicity by tamarind components: An in silico study. Fluoride 2018, 51, 122–136.
  16. Mishra, S.; Singh, S.; Misra, K. Restraining pathogenicity in Candida albicans by taxifolin as an inhibitor of Ras1-pka pathway. Mycopathologia 2017, 182, 953–965.
  17. Liu, H.; He, S.; Wang, T.; Orang-Ojong, B.; Lu, Q.; Zhang, Z.; Pan, L.; Chai, X.; Wu, H.; Fan, G.; et al. Selected phytoestrogens distinguish roles of ERα transactivation and ligand binding for anti-inflammatory activity. Endocrinology 2018, 159, 3351–3364.
  18. Davis, C.K.; Nasla, K.; Anjana, A.K.; Rajanikant, G.K. Taxifolin as dual inhibitor of Mtb DNA gyrase and isoleucyl-tRNA synthetase: In silico molecular docking, dynamics simulation and in vitro assays. Silico Pharmacol. 2018, 6, 8.
  19. Raj, U.; Varadwaj, P.K. Flavonoids as multi-target inhibitors for proteins associated with ebola virus: In silico discovery using virtual screening and molecular docking studies. Interdiscip. Sci. Comput. Life Sci. 2015, 8, 132–141.
  20. Meenambiga, S.S.; Rajagopal, K. Antibiofilm activity and molecular docking studies of bioactive secondary metabolites from endophytic fungus Aspergillus nidulans on oral Candida albicans. Artic. J. Appl. Pharm. Sci. 2018, 8, 37–45.
  21. Rajendran, P.; Maheshwari, U.; Muthukrishnan, A.; Muthuswamy, R.; Anand, K.; Ravindran, B.; Dhanaraj, P.; Balamuralikrishnan, B.; Chang, S.W.; Chung, W.J. Myricetin: Versatile plant based flavonoid for cancer treatment by inducing cell cycle arrest and ROS–reliant mitochondria-facilitated apoptosis in A549 lung cancer cells and in silico prediction. Mol. Cell. Biochem. 2020, 476, 57–68.
  22. Haque, M.W.; Pattanayak, S.P. Taxifolin inhibits 7,12-dimethylbenz(a)anthracene-induced breast carcinogenesis by regulating AhR/CYP1A1 signaling pathway. Pharmacogn. Mag. 2017, 13, S749.
  23. Sun, J.; Ge, F.; Wang, Y.; Dong, Y.; Shan, Y.; Zhu, Q.; Wu, X.; Wu, C.; Ge, R.S. Taxifolin is a rat and human 11β-hydroxysteroid dehydrogenase 1 inhibitor as a possible drug to treat the metabolic syndrome. J. Funct. Foods 2018, 49, 181–187.
  24. Xu, Z.; Li, K.; Pan, T.; Liu, J.; Li, B.; Li, C.; Wang, S.; Diao, Y.; Liu, X. Lonicerin, an anti-algE flavonoid against Pseudomonas aeruginosa virulence screened from Shuanghuanglian formula by molecule docking based strategy. J. Ethnopharmacol. 2019, 239, 111909.
  25. Lu, J.J.; Zhou, F.M.; Hu, X.J.; Fang, J.J.; Liu, C.X.; Zhu, B.Q.; Ding, Z.S. Molecular docking simulation and in vitro studies on estrogenic activities of flavonoids from leaves of Carya cathayensis Sarg. Steroids 2020, 163, 108726.
  26. Esfahani, A.N.; Mirzaei, M. Flavonoid derivatives for monoamine oxidase–A inhibition. Adv. J. Chem. Sect. B 2019, 1, 17–22.
  27. Orlova, S.V.; Tatarinov, V.V.; Nikitina, E.A.; Sheremeta, A.V.; Ivlev, V.A.; Vasil’ev, V.G.; Paliy, K.V.; Goryainov, S.V. Molecular-biological problems of drug design and mechanism of drug action bioavailability and safety of dihydroquercetin (Review). Pharm. Chem. J. 2021, 55, 3–8.
  28. Weidmann, A.E. Dihydroquercetin: More than just an impurity? Eur. J. Pharmacol. 2012, 684, 19–26.
  29. Das, A.; Baidya, R.; Chakraborty, T.; Samanta, A.K.; Roy, S. Pharmacological basis and new insights of taxifolin: A comprehensive review. Biomed. Pharmacother. 2021, 142, 112004.
  30. Turck, D.; Bresson, J.; Burlingame, B.; Dean, T.; Fairweather-Tait, S.; Heinonen, M.; Hirsch-Ernst, K.I.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.; et al. Statement on the safety of taxifolin-rich extract from Dahurian Larch (Larix gmelinii). EFSA J. 2017, 15, e05059.
  31. Gogoi, N.; Chowdhury, P.; Goswami, A.K.; Das, A.; Chetia, D.; Gogoi, B. Computational guided identification of a citrus flavonoid as potential inhibitor of SARS-CoV-2 main protease. Mol. Divers. 2021, 25, 1745–1759.
  32. Fischer, A.; Sellner, M.; Neranjan, S.; Smieško, M.; Lill, M.A. Potential inhibitors for novel coronavirus protease identified by virtual screening of 606 million compounds. Int. J. Mol. Sci. 2020, 21, 3626.
  33. Yang, C.; Wang, Z.; Mi, Y.; Gao, M.; Lv, J.; Meng, Y.; Yang, B.; Kuang, H. UHPLC-MS/MS determination, pharmacokinetic and bioavailability study of taxifolin in rat plasma after oral administration of its nanodispersion. Molecules 2016, 21, 494.
  34. Zu, Y.; Wu, W.; Zhao, X.; Li, Y.; Wang, W.; Zhong, C. Enhancement of solubility, antioxidant ability and bioavailability of taxifolin nanoparticles by liquid antisolvent precipitation technique. Int. J. Pharm. 2014, 471, 366–376.
  35. Yang, P.; Xu, F.; Li, H.F.; Wang, Y.; Li, F.C.; Shang, M.Y.; Liu, G.X.; Wang, X.; Cai, S.Q. Detection of 191 taxifolin metabolites and their distribution in rats using HPLC-ESI-IT-TOF-MSn. Molecules 2016, 21, 1209.
  36. Terekhov, R.P.; Selivanova, I.A. Molecular modeling of the interaction of the dihydroquercetin and its metabolites with cyclooxygenase-2. Bull. Exp. Biol. Med. 2019, 18, 101–106.
  37. Terekhov, R.P.; Selivanova, I.A.; Tyukavkina, N.A.; Shylov, G.V.; Utenishev, A.N.; Porozov, Y.B. Taxifolin tubes: Crystal engineering and characteristics. Acta Crystallogr. Sect. B Struct. Sci. Cryst. Eng. Mater. 2019, 75, 175–182.
  38. WHO Coronavirus (COVID-19) Dashboard|WHO Coronavirus (COVID-19) Dashboard with Vaccination Data. Available online: https://covid19.who.int/ (accessed on 9 March 2022).
  39. Jain, A.S.; Sushma, P.; Dharmashekar, C.; Beelagi, M.S.; Prasad, S.K.; Shivamallu, C.; Prasad, A.; Syed, A.; Marraiki, N.; Prasad, K.S. In silico evaluation of flavonoids as effective antiviral agents on the spike glycoprotein of SARS-CoV-2. Saudi J. Biol. Sci. 2021, 28, 1040–1051.
  40. Rudrapal, M.; Issahaku, A.R.; Agoni, C.; Bendale, A.R.; Nagar, A.; Soliman, M.E.S.; Lokwani, D. In silico screening of phytopolyphenolics for the identification of bioactive compounds as novel protease inhibitors effective against SARS-CoV-2. J. Biomol. Struct. Dynam. 2021, 1–17.
  41. Guler, H.I.; Tatar, G.; Yildiz, O.; Belduz, A.O.; Kolayli, S. Investigation of potential inhibitor properties of ethanolic propolis extracts against ACE-II receptors for COVID-19 treatment by molecular docking study. Arch. Microbiol. 2021, 203, 3557–3564.
  42. Taldaev, A.K.; Terekhov, R.P.; Selivanova, I.A. Flavonoids as potential inhibitors of SARS-CoV-2 infection: In silico study. Bull. Sib. Med. 2022, 21, 103–108.
  43. Shohan, M.; Nashibi, R.; Mahmoudian-Sani, M.R.; Abolnezhadian, F.; Ghafourian, M.; Alavi, S.M.; Sharhani, A.; Khodadadi, A. The therapeutic efficacy of quercetin in combination with antiviral drugs in hospitalized COVID-19 patients: A randomized controlled trial. Eur. J. Pharmacol. 2022, 914, 174615.
More
Video Production Service