Next Article in Journal
Ionic Liquids as Organocatalysts for Nucleophilic Fluorination: Concepts and Perspectives
Next Article in Special Issue
A Potential Role of Plant/Macrofungi/Algae-Derived Non-Starch Polysaccharide in Colitis Curing: Review of Possible Mechanisms of Action
Previous Article in Journal
Daphnane-Type Diterpenes from Stelleropsis tianschanica and Their Antitumor Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Luteolinidin and Petunidin 3-O-Glucoside: A Competitive Inhibitor of Tyrosinase

1
Department of Pharmaceutical Engineering, Sangji University, 83 Sangjidae-gil, Wonju 26339, Korea
2
Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Korea
3
School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen 518107, China
4
Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
5
BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(17), 5703; https://doi.org/10.3390/molecules27175703
Submission received: 5 August 2022 / Revised: 1 September 2022 / Accepted: 2 September 2022 / Published: 4 September 2022
(This article belongs to the Special Issue Key Role of Natural Bioactive Compounds in Health and Diseases)

Abstract

:
The enzyme tyrosinase plays a key role in the early stages of melanin biosynthesis. This study evaluated the inhibitory activity of anthocyanidin (1) and anthocyanins (26) on the catalytic reaction. Of the six derivatives examined, 13 showed inhibitory activity with IC50 values of 3.7 ± 0.1, 10.3 ± 1.0, and 41.3 ± 3.2 μM, respectively. Based on enzyme kinetics, 13 were confirmed to be competitive inhibitors with Ki values of 2.8, 9.0, and 51.9 μM, respectively. Molecular docking analysis revealed the formation of a binary encounter complex between 13 and the tyrosinase catalytic site. Luteolinidin (1) and petunidin 3-O-glucoside (2) may serve as tyrosinase inhibitors to block melanin production.

1. Introduction

Tyrosinase (EC 1.14.18.1) is a multifunctional copper-containing enzyme that plays a role in melanin biosynthesis in mammals, plants, insects, and microorganisms [1]. It catalyzes two reactions: the hydroxylation of tyrosine to L-3,4-dihydroxyphenylalanine and the subsequent oxidation of L-3,4-dihydroxyphenylalanine to dopaquinone [2]. Melanin is produced in skin melanocytes to protect the skin from ultraviolet (UV) radiation [3]. Melanin overproduction leads to age spots, freckles, senile lentigines, solar lentigo, hyperpigmentation, and melisma [1,3], while overexposure of the skin to UV radiation causes malignant melanoma [4]. These disorders may be directly related to the tyrosinase catalytic reaction [4]. To address these problems, tyrosinase inhibitors with high activity and low toxicity have been developed from natural products [1], such as methyl hesperidin [5], broussoflavonol J [6], and dieckol [7].
Anthocyanins are polyphenols that dissolve in water [8] and are flavonoids biosynthesized from phenylalanine and malonyl-CoA along with flavanols [9]. They exist in edible plants, such as purple corn, black soybean, blueberry, and grape, and are responsible for the plants’ red, purple, and blue colors [8,9]. The carbon skeleton of anthocyanins is composed of C-6 (A-ring), C-3 (C-ring), and C-6 (B-ring) carbons [10]. Their colors depend on the amount of hydroxylation in the B-ring [10]. Their maximum absorption occurs at 450–550 nm [10]. Cyandin, delphinindin, and pelargonidin glycosides such as cyanidin 3-O-glucoside, delphinidin 3-O-glucoside, and pelargonidin 3-O-glucoside are the most abundant naturally occurring anthocyanins, which are widely found in Morus nigra, Berberis vulgaris, and Ipomea batatas [11]. Anthocyanins possess a variety of bioactivities, such as anti-inflammation [12], anti-cancer [13], and renal-protective [14] effects. Cyanidin 3-O-glycoside and peonidin 3-O-glucoside suppress matrix metalloproteinase expression in IL-1β-stimulated human articular chondrocytes [12]. Delphinidin, which is a derivative of an anthocyanin, inhibits glyoxalase 1 and is overexpressed in tumor cells [13]. Cyanidin 3-O-glucoside is protected against diabetic nephropathy by decreasing renal TNF-α mRNA and NF-κB mRNA levels in rats [14].

2. Results

2.1. Inhibitory Activity of Antocyanins on Tyrosinase

This study evaluated the inhibitory effects of one anthocyanidin and five anthocyanins on tyrosinase: luteolinidin (1), petunidin 3-O-glucoside (2), delphinidin 3-O-galactoside (3), kuromanin (cyanidin 3-O-glucoside) (4), delphinidin 3-O-glucoside (5), and callistephin (pelargonidin 3-O-glucoside) (6) (Figure 1). At 50 μM, compounds 13 had inhibitory ratios exceeding 55% of the control value (Table 1), showing dose-dependent inhibitory activity, with IC50 values of 3.7 ± 0.8, 10.3 ± 1.0, and 41.3 ± 3.2 μM, respectively (Figure 1 and Table 1).

2.2. Enzyme Kinetics of the Compounds on Tyrosinase

To reveal the mechanism of binding between compounds 13 and the enzyme, an enzyme kinetic study was performed using various substrate concentrations. The v0 was calculated at ~10% of the substrate conversion rate by tyrosinase. The results are shown as Lineweaver–Burk plots (Figure 2A–C and Table 1). Compounds 13 all had different slopes (Km/Vmax) and different y-axis intercepts (Vmax), demonstrating that they bound to the enzyme reaction site competitively. Using Dixon plots, 13 were calculated to have inhibition constants (Ki) of 2.8, 9.0, and 51.9 μM, respectively (Figure 2D–F and Table 1).

2.3. Molecular Docking of the Compounds with Tyrosinase

To determine the binding orientation of inhibitors in tyrosinase, a molecular simulation analysis was performed using AutoDock 4.2. Based on an enzyme kinetic study, a grid containing the active site with two copper ions was used to simulate the interactions of the inhibitors with the tyrosinase catalytic site. As shown in Figure 3A−C and Table 2, inhibitors 1 and 3 formed two different clusters according to the binding position. However, their Autodock scores were similar. Based on enzyme theory, competitive inhibitors bind mainly to the catalytic site. Out of 50 ranks, cluster 1 next to the catalytic site was created only by ranks 1–3 (1) and 1–5 (3) [15].
These findings suggested that cluster 2 may be the site that interacts with inhibitors 1 and 3. As indicated in Figure 3A–C and Table 2, 13 were stably anchored in the active site of tyrosinase with respective Autodock scores of −5.36, −4.96, and −4.61 kcal/mol. Inhibitor 1 formed four hydrogen bonds with three amino acids (2.71 Å from His244, 2.65 and 2.72 Å from Glu256, and 2.53 Å from Gly281). The five hydroxyl groups of inhibitor 2 were located at distances of 2.78 Å from Asn260, 3.13 Å from Arg268, 2.94 Å from Gly281, and 2.73 Å and 3.15 Å from Ser282. Inhibitor 3 formed three hydrogen bonds: 2.83 Å from Asn260 and two 3.14 Å from Arg268.

3. Experimental Section

3.1. Chemical Reagents

Tyrosinase (T3824), kojic acid (K3125), and L-tyrosine (T3754) were purchased from Sigma–Aldrich (St. Louis, MO, USA). UV-Vis spectra were obtained using the TECAN infinite 200 PRO® spectrophotometer (Zurich, Switzerland). Luteolinidin (1), petunidin 3-O-glucoside (2), delphinidin 3-O-galactoside (3), kuromanin (cyanidin 3-O-glucoside) (4), delphinidin 3-O-glucoside (5), and callistephin (pelargonidin 3-O-glucoside) (6) were purchased from LGC Standards (1154-78-5, Teddington Middlesex, UK) and Sigma–Aldrich (2, 30638; 3, 04301; 4, 52976; 5, 73705; 6, 79576; St. Louis, MO, USA).

3.2. Tyrosinase Assay

To assess the inhibitory effect of compounds on tyrosinase, 130 μL tyrosinase in 0.05 mM phosphate buffer (pH 6.8) was aliquoted into 96-well plates [16], and 20 μL of each compound at concentrations ranging from 0.5 to 0.0075 mM was added. Next, 50 μL 1.2 mM L-tyrosine in phosphate buffer was diluted to calculate the inhibitory activity. Finally, 50 μL 10–0.62 mM l-tyrosine in buffer was added to analyze the initial velocity (v0). Twenty minutes after starting the reaction, the amount of product was detected at 475 nm. The inhibitory activity was calculated using the following equation: Inhibitory activity rate (%) = [(ΔC − ΔS)/ΔC] × 100, where C and S are the intensity of control and inhibitor after 20 min, respectively.

3.3. Molecular Docking

The three-dimensional structure of the protein encoded by 2Y9X was determined from the Research Collaboratory for Structural Bioinformatics homepage. Hydrogen atoms were added to this and assigned Gasteiger charges using AutoDockTools. Ligands were built and their energy minimized by MM2 using Chem3D Pro. A grid containing the active site was established (X = 60, Y = 60, Z = 60). A DPS file was constructed to set up a Lamarckian genetic algorithm for ligand docking with the receptor (50 runs, maximum number set as long). The results were presented using Chimera (San Francisco, CA, USA) and LIGPLOT (Cambridge, UK).

3.4. Statistical Analysis

All inhibitory concentration data were obtained from independent experiments carried out in triplicate. Results are shown as the mean ± standard error of the mean (SEM). The results were subjected to analysis using Sigma plot 10.0 (Systat Software Inc., San Jose, CA, USA).

4. Discussion

The anthocyanin-rich fractions of blueberry extract decreased the proliferation of B16-F10 melanoma murine cells by inducing apoptosis [17]. The major anthocyanins in black rice (peonidin 3-O-glucoside and cyanidin 3-O-glucoside) were confirmed to decrease MMP-2 and u-PA secretion in SCC-, Huh-7, and HeLa cells [18]. Recently, the ethyl acetate fraction from Arctium lappa L. leaves, which contained trans-5-caffeoylquic acid, rutin, kaempferol 3-O-rutinoside, 3,5-di-O-caffeoylquic acid, and 4,5-di-O-caffeoylqunic acid, was found to decrease tyrosinase activity and melanin levels [19]. This downregulated the expression of phosphorylated c-Jun N-terminal kinase, microphthalmia-associated transcription factor, tyrosinase-related protein 1, and tyrosinase in α-MSH-induced B16.F10 cells [19].
Luteolinidin (1) from Sorghum bicolor [20] and petunidin 3-O-glucoside (2) from Vitis vinifera L. [21] were the most potent inhibitors on tyrosinase, whereas delphinidin 3-O-galactoside [22] (3) showed moderate inhibitory activity. Many studies have examined the inhibitory effects of anthocyanin-rich fractions on tyrosinase [23,24], whereas we examined the tyrosinase inhibitory activity of single anthocyanins from plants. As a result, the anthocyanidin (1) and anthocyanins (23) showed the inhibitory activity within the concentration range of micromole on tyrosinase. Some flavonoids have recently been reported to interact directly with the tyrosinase catalytic site [25]. In addition, inhibitors 13, as competitive inhibitors, were confirmed to be bound into a common active site in an in vitro assay. The molecular simulation calculated that they were anchored into the cavity hole in tyrosinase. In particular, two inhibitors, 1 and 3, formed cluster 1 stably in the left position next to the active site. Based on enzyme kinetic theory, it can be confirmed that this cluster, formed by the two inhibitors, had a logical error. Therefore, cluster 2, formed by the two inhibitors on the catalytic site, was determined to be the predicted binding calculated by the Autodock program. According to the above results, it was confirmed that glycosides 2 and 3 form a similar Autodock score and binding poses. Inhibitors 2 and 3, which are similar to the backbone of inhibitor 1, were bound as different poses in the catalytic site by their sugars.

5. Conclusions

Many studies have examined the tyrosinase inhibitory activity of anthocyanin extracts from plants [26]. We evaluated how these compounds inhibit the catalytic reaction. Of the one anthocyanidin and five anthocyanins evaluated, luteolinidin (1), petunidin 3-O-glucoside (2), and delphinidin 3-O-galactoside (3) acted as tyrosinase inhibitors at micromolar concentrations. These compounds bound competitively, with Ki values of 2.8, 9.0, and 51.9 μM, respectively. A molecular simulation of the binding of the compounds to the catalytic site of tyrosinase confirmed that inhibitors 1 and 2 interact with tyrosinase with an excellent inhibitory effect and low Autodock binding energy. Therefore, we suggest that luteolinidin (1) and petunidin 3-O-glucoside (2) should be the lead compounds for developing new tyrosinase inhibitors.

Author Contributions

Conceptualization, J.H.K. and S.Y.Y.; formal analysis, J.H.K. and S.Y.Y.; data curation, X.S.; writing—original draft preparation, J.H.K. and S.Y.Y.; writing—review and editing, J.A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (No. NRF-2022R1C1C1004636) and (No. NRF-2020R1A5A2017323).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are no longer available.

References

  1. Chen, J.; Li, Q.; Ye, Y.; Huang, Z.; Ruan, Z.; Jin, N. Phloretin as both a substrate and inhibitor of tyrosinase: Inhibitory activity and mechanism. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 226, 117642. [Google Scholar] [CrossRef] [PubMed]
  2. Bahuguna, A.; Bharadwaj, S.; Chauhan, A.K.; Kang, S.C. Inhibitory insights of strawberry (Fragaria × ananassa var. Seolhyang) root extract on tyrosinase activity using computational and in vitro analysis. Int. J. Biol. Macromol. 2020, 165, 2773–2788. [Google Scholar] [CrossRef] [PubMed]
  3. Shi, F.; Xie, L.; Lin, Q.; Tong, C.; Fu, Q.; Xu, J.; Xiao, J.; Shi, S. Profiling of tyrosinase inhibitors in mango leaves for a sustainable agro-industry. Food Chem. 2020, 312, 126042. [Google Scholar] [CrossRef] [PubMed]
  4. Barros, M.R.; Menezes, T.M.; da Silva, L.P.; Pires, D.S.; Princival, J.L.; Seabra, G.; Neves, J.L. Furan inhibitory activity against tyrosinase and impact on B16F10 cell toxicity. Int. J. Biol. Macromol. 2019, 136, 1034–1041. [Google Scholar] [CrossRef]
  5. Shu, P.; Fei, Y.; Li, J.; Liu, A.; Zhang, L.; Niu, H.; Liu, W.; Wei, X.; Xiao, F.; Xu, Z. Two new phenylethanoid glycosides from Ginkgo biloba leaves and their tyrosinase inhibitory activities. Carbohydr. Res. 2020, 494, 108059. [Google Scholar] [CrossRef]
  6. Tian, J.-L.; Liu, T.-L.; Xue, J.-J.; Hong, W.; Zhang, Y.; Zhang, D.-X.; Cui, C.-C.; Liu, M.-C.; Niu, S.-L. Flavanoids derivatives from the root bark of Broussonetia papyrifera as a tyrosinase inhibitor. Ind. Crop. Prod. 2019, 138, 111445. [Google Scholar] [CrossRef]
  7. Kang, S.-M.; Heo, S.-J.; Kim, K.-N.; Lee, S.-H.; Yang, H.-M.; Kim, A.-D.; Jeon, Y.-J. Molecular docking studies of a phlorotannin, dieckol isolated from Ecklonia cava with tyrosinase inhibitory activity. Bioorganic Med. Chem. 2012, 20, 311–316. [Google Scholar] [CrossRef]
  8. Jiang, T.; Zhou, J.; Liu, W.; Tao, W.; He, J.; Jin, W.; Guo, H.; Yang, N.; Li, Y. The anti-inflammatory potential of protein-bound anthocyanin compounds from purple sweet potato in LPS-induced RAW264.7 macrophages. Food Res. Int. 2020, 137, 109647. [Google Scholar] [CrossRef]
  9. Lin, Y.; Zhang, L.; Zhang, J.; Zhang, J.; Zhang, Y.; Wang, Y.; Chen, Q.; Luo, Y.; Zhang, Y.; Li, M.; et al. Identi-fication of anthocyanins-related glutathione S-transferase (GST) genes in the genome of cultivated strawberry (Fragaria x ananassa). Int. J. Mol. Sci. 2020, 21, 8708. [Google Scholar] [CrossRef]
  10. Glover, B.J.; Martin, C. Anthocyanins. Curr. Biol. 2012, 22, R147–R150. [Google Scholar] [CrossRef] [Green Version]
  11. Oladzadabbasabadi, N.; Nafchi, A.M.; Ghasemlou, M.; Ariffin, F.; Singh, Z.; Al-Hassan, A.A. Natural anthocyanins: Sources, extraction, characterization, and suitability for smart packaging. Food Packag. Shelf. 2022, 33, 100872. [Google Scholar] [CrossRef]
  12. Wongwichai, T.; Teeyakasem, P.; Pruksakorn, D.; Kongtawelert, P.; Pothacharoen, P. Anthocyanins and metabolites from purple rice inhibit IL-1β-induced matrix metalloproteinases expression in human articular chondrocytes through the NF-kβ and ERK/MAPK pathway. Biomed. Pharmacoth. 2019, 112, 108610. [Google Scholar] [CrossRef] [PubMed]
  13. Takasawa, R.; Saeki, K.; Tao, A.; Yoshimori, A.; Uchiro, H.; Fujiwara, M.; Tanuma, S.-I. Delphinidin, a dietary anthocyanidin in berry fruits, inhibits human glyoxalase I. Bioorganic Med. Chem. 2010, 18, 7029–7033. [Google Scholar] [CrossRef] [PubMed]
  14. Qi, S.S.; He, J.l.; Dong, L.C.; Yuan, L.P.; Wu, J.L.; Zu, Y.X.; Zheng, H.X. cyaniding-3-glucoside from black rice prevents renal dysfunction and renal fibrosis in streptozotoci-diabetic rats. J. Funct. Foods 2020, 72, 104062. [Google Scholar] [CrossRef]
  15. Kim, J.H.; Jo, Y.D.; Kim, H.-Y.; Kim, B.-R.; Nam, B. In vitro and in silico insights into she inhibitors with amide-scaffold from the leaves of Capsicum chinense Jacq. Comput. Struct. Biotec. 2018, 16, 404–411. [Google Scholar] [CrossRef]
  16. Yang, H.; Wang, Z.; Song, W.; Zhao, Z.; Zhao, Y. Isolation of proanthocyanidins from Pinus thunbergii needles and tyrosinase inhibition activity. Process Biochem. 2021, 100, 245–251. [Google Scholar] [CrossRef]
  17. Bunea, A.; Rugină, D.; Sconţa, Z.; Pop, R.M.; Pintea, A.; Socaciu, C.; Tăbăran, F.; Grootaert, C.; Struijs, K.; VanCamp, J. An-thocyanin determination in blue berry extracts from various cultivars and their antiproliferative and apoptotic properties in b16-F10 metastatic murine melanoma cells. Phytochemistry 2013, 95, 436–444. [Google Scholar] [CrossRef]
  18. Chen, P.-N.; Kuo, W.-H.; Chiang, C.-L.; Chiou, H.-L.; Hsieh, Y.-S.; Chu, S.-C. Black rice anthocyanins inhibit cancer cells invasion via repressions of MMPs and u-PA expression. Chem. Interactions 2006, 163, 218–229. [Google Scholar] [CrossRef]
  19. Lee, C.J.; Park, S.K.; Kang, J.Y.; Kim, J.M.; Yoom, S.K.; Han, H.J.; Kim, D.-O.; Heo, H.J. Melanogenesis regulatory activity of the ethyl acetate fraction from Arctium lappa L. leaf on α-MSH–induced B16/F10 melanoma cells. Ind. Crop. Prod. 2019, 138, 111581. [Google Scholar] [CrossRef]
  20. Tugizimana, F.; Steenkamp, P.A.; Piater, L.A.; Labuschagne, L.; Dubery, I.A. Unravellng the metabolic reconfiguration of the post-challenge primed state in Sorghum bicolor responding to colletotrichum sublineolum infection. Metabolites 2019, 9, 194. [Google Scholar] [CrossRef] [Green Version]
  21. Picariello, G.; Ferranti, P.; Garro, G.; Manganiello, G.; Chianese, L.; Coppola, R.; Addeo, F. Profiling of anthocyanins for the taxonomic assessment of ancient purebred V. vinifera red grape varieties. Food Chem. 2014, 146, 15–22. [Google Scholar] [CrossRef]
  22. Ulaszewska, M.; Garcia-Aloy, G.M.; Vázquez-Manjarrez, N.; Soria-Florido, M.T.; Llorach, R.; Mattivi, F.; Manach, C. Food intake biomarkers for berries and grapes. Genes Nutr. 2020, 15, 17. [Google Scholar] [CrossRef] [PubMed]
  23. Jhan, J.K.; Chung, Y.C.; Chen, G.H.; Chang, C.-H.; Lu, Y.-C.; Hsu, C.-K. Anthocyanin contents in the seed coat of black soya bean and their anti-human tyrosinase activity and antioxidative activity. Int. J. Cosmet. Sci. 2016, 38, 319–324. [Google Scholar] [CrossRef] [PubMed]
  24. Fawole, O.A.; Makunga, N.P.; Opara, U.L. Antibacterial, antioxidant and tyrosinase-inhibiton activities of pomegranate fruit peel methanolic extract. BMC Complement Altern. Med. 2012, 12, 200. [Google Scholar] [CrossRef] [PubMed]
  25. Koyu, H.; Kazan, A.; Ozturk, T.K.; Yesil-Celiktas, O.; Haznedaroglu, M.Z. Optimizing subscritical water extraction of Morus nigra L. fruits for maximization of tyrosinase inhibitory activity. J. Supercrit. Fluid. 2017, 127, 15–22. [Google Scholar] [CrossRef]
  26. Koyu, H.; Kazan, A.; Demir, S.; Haznedaroglu, M.Z.; Yesil-Celiktas, O. Optimization of microwave assisted extraction of Morus nigra L. fruits maximizing tyrosinase inhibitory activity with isolation of bioactive constituents. Food Chem. 2018, 248, 183–191. [Google Scholar] [CrossRef]
Figure 1. The structures of compounds 16.
Figure 1. The structures of compounds 16.
Molecules 27 05703 g001
Figure 2. Lineweaver–Burk (AC) and Dixon (DF) plots of inhibitors 13, the results were subjected to analysis using Sigma plot 10.0.
Figure 2. Lineweaver–Burk (AC) and Dixon (DF) plots of inhibitors 13, the results were subjected to analysis using Sigma plot 10.0.
Molecules 27 05703 g002
Figure 3. The best binding poses (AC) of compounds 13 on tyrosinase (orange structure is best pose of cluster 1 of inhibitors 1 and 3, and yellow structure is best pose of cluster 2 of inhibitors 1 and 3, and cluster 1 of inhibitor 2). The green dotted line represents hydrogen bonds’ interaction between inhibitor 13 (DF) and enzyme, respectively.
Figure 3. The best binding poses (AC) of compounds 13 on tyrosinase (orange structure is best pose of cluster 1 of inhibitors 1 and 3, and yellow structure is best pose of cluster 2 of inhibitors 1 and 3, and cluster 1 of inhibitor 2). The green dotted line represents hydrogen bonds’ interaction between inhibitor 13 (DF) and enzyme, respectively.
Molecules 27 05703 g003
Table 1. Inhibitory activity of compounds 16 on tyrosinase.
Table 1. Inhibitory activity of compounds 16 on tyrosinase.
Compound aInhibitory Ratio at 50 μMIC50 (μM) dBinding Mode (ki, μM)
1176.5 ± 11.63.7 ± 0.8Competitive inhibitor (2.8 ± 0.5)
269.2 ± 1.010.3 ± 1.0Competitive inhibitor (9.0 ± 2.1)
355.2 ± 0.941.3 ± 3.2Competitive inhibitor (51.9 ± 3.8)
4−1.9 ± 40.0N.T.N.T. c
547.5 ± 3.2N.T.N.T. c
66.5 ± 2.1N.T.N.T. c
Kojic acid b 31.4 ± 1.1
a Compounds were tested three times. b Positive control c N.T.: not test. d Mean ± SEM.
Table 2. Hydrogen bonds analysis of the compounds with tyrosinase.
Table 2. Hydrogen bonds analysis of the compounds with tyrosinase.
Autodock Energy (kcal/mol)Hydrogen Bonds (Å)
1−5.36His244(2.71), Glu256(2.65, 2.72), Gly281(2.53)
2−4.96Asn260(2.78), Arg268(3.13), Gly281(2.94), Ser282(2.73,3.15)
3−4.61Asn260(2.83), Arg268(3.14, 3.14)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yang, S.Y.; Kim, J.H.; Su, X.; Kim, J.A. The Luteolinidin and Petunidin 3-O-Glucoside: A Competitive Inhibitor of Tyrosinase. Molecules 2022, 27, 5703. https://doi.org/10.3390/molecules27175703

AMA Style

Yang SY, Kim JH, Su X, Kim JA. The Luteolinidin and Petunidin 3-O-Glucoside: A Competitive Inhibitor of Tyrosinase. Molecules. 2022; 27(17):5703. https://doi.org/10.3390/molecules27175703

Chicago/Turabian Style

Yang, Seo Young, Jang Hoon Kim, Xiangdong Su, and Jeong Ah Kim. 2022. "The Luteolinidin and Petunidin 3-O-Glucoside: A Competitive Inhibitor of Tyrosinase" Molecules 27, no. 17: 5703. https://doi.org/10.3390/molecules27175703

Article Metrics

Back to TopTop