Open Access

Inhibitory effects of quercetin and its major metabolite quercetin‑3‑O‑β‑D‑glucoside on human UDP‑glucuronosyltransferase 1A isoforms by liquid chromatography‑tandem mass spectrometry

  • Authors:
    • Rui Zhang
    • Ye Wei
    • Tingyu Yang
    • Xixi Huang
    • Jinping Zhou
    • Chunxiao Yang
    • Jiani Zhou
    • Yani Liu
    • Shaojun Shi
  • View Affiliations

  • Published online on: June 6, 2021     https://doi.org/10.3892/etm.2021.10274
  • Article Number: 842
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Quercetin is a flavonoid that is widely present in plant‑derived food. Quercetin‑3‑O‑β‑D‑glucoside (Q3GA) is a predominant metabolite of quercetin in animal and human plasma. The inhibitory effects of the UDP‑glucuronosyl transferases (UGTs) caused by herbal components may be a key factor for the clinical assessment of herb‑drug interactions (HDIs). The present study aimed to investigate the inhibitory profile of quercetin and Q3GA on recombinant UGT1A isoforms in vitro. The metabolism of the nonspecific substrate 4‑methylumbelliferone (4‑MU) by the UGT1A isoforms was assessed by liquid chromatography‑tandem mass spectrometry. Preliminary screening experiments indicated that quercetin exhibited stronger inhibitory effects on UGT1A1, UGT1A3, UGT1A6 and UGT1A9 enzymes than Q3GA. Kinetic experiments were performed to characterize the type of inhibition caused by quercetin and Q3GA towards these UGT isoforms. Quercetin exerted non‑competitive inhibition on UGT1A1 and UGT1A6, with half maximal inhibitory concentration (IC50) values of 7.47 and 7.07 µM and inhibition kinetic parameter (Ki) values of 2.18 and 28.87 µM, respectively. Quercetin also exhibited competitive inhibition on UGT1A3 and UGT1A9, with IC50 values of 10.58 and 2.81 µM and Ki values of 1.60 and 0.51 µM, respectively. However, Q3GA displayed weak inhibition on UGT1A1, UGT1A3 and UGT1A6 enzymes with IC50 values of 45.21, 106.5 and 51.37 µM, respectively. In the present study, quercetin was a moderate inhibitor of UGT1A1 and UGT1A3, a weak inhibitor of UGT1A6, and a strong inhibitor on UGT1A9. The results of the present study suggested potential HDIs that may occur following quercetin co‑administration with drugs that are mainly metabolized by UGT1A1, UGT1A3 and UGT1A9 enzymes.

Introduction

The flavonoid quercetin (3,3',4',5,7-pentahydroxyflavone; Fig. 1A) is one of the most abundant dietary polyphenols. It is mainly present in fruits and vegetables and ~3-40 mg quercetin is consumed in daily diets (1,2). Quercetin is widespread in the flowers, leaves and fruits of various plants and exhibits a multitude of pharmacological activities, including anti-neoplastic (3-5), anti-oxidative (6,7), anti-inflammatory (8,9), anti-thrombotic (10,11), antiviral (12,13), cardiovascular-protective (14,15) and immune-regulatory (16,17) effects. Isolated quercetin is a dietary supplement and its recommended maximum daily dosage is 1,000 mg (18). Following oral ingestion, quercetin is extensively conjugated with glucuronic acid and/or sulfate in the small intestine and liver. Quercetin-3-O-β-D-glucoside (Q3GA; Fig. 1B) is one of the primary metabolites found in the blood circulation (19). Q3GA exerts various pharmacological properties. Quercetin inhibits the viability of neural stem cells via the Akt signaling pathway. However, Q3GA provides a novel therapeutic potential in neurodegenerative diseases (20). In addition, the anti-inflammatory activity of Q3GA was evaluated by assessing the inhibition of LPS-induced NO release in vitro (21).

The biological activity of quercetin is notably affected by phase II, and not phase I, metabolism enzymes. As a plant-derived polyphenol, quercetin contains more than one free hydroxyl group, which makes it easy to be metabolized by different types of UGT enzyme isoforms, including UGT1As (22,23). UGTs are considered the indispensable enzymes of phase II metabolism and catalyze the conjugation of several endobiotics or xenobiotics with UDP-glucuronic acid in order to produce more hydrophilic metabolites that are easily excreted via the kidneys or the bile and the gut (24,25). CYP450-mediated herb-drug interactions (HDIs) have been previously investigated by in vitro assays using cocktails of probe substrates (26-29). Subsequent studies involving UGT enzymes have demonstrated that drug interactions based on the inhibition of UGTs may lead to clinically important side effects (30,31). Therefore, from a clinical point of view, the study of the inhibition of herbal compounds on UGT-mediated metabolism may aid the understanding of HDIs.

The in vitro UGT enzyme assay utilizes the nonspecific substrate 4-MU as a substrate and has various advantages over the use of human liver microsomes that include several specific probe-substrates (32). In the present study, the inhibition type and inhibitory effects of quercetin and its major metabolite Q3GA were assessed on various UGT isoforms (UGT1A1, UGT1A3, UGT1A6 and UGT1A9) by liquid chromatography-tandem mass spectrometry (LC-MS/MS). This method was used to detect the changes in the concentration levels of 4-methylumbelliferyl-β-D-glucuronides (4-MU-G). This may provide insight into the potential HDIs regarding quercetin and Q3GA, providing the basis for further drug research and safe drug use.

Materials and methods

Chemicals and reagents

4-MU and 4-MU-G were purchased from Shanghai Yuanye Bio-Technology Co., Ltd. Q3GA was purchased from Chengdu Sino Standards Bio-Tech Co., Ltd. 7-Hydroxycoumarin was obtained from Dalian Meilun Biotechnology Co., Ltd. Quercetin and uridine-5'-diphosphoglucuronic acid (UDPGA; trisodium salt) were purchased from Sigma-Aldrich (Merck KGaA). Recombinant human UGT isoforms (UGT1A1, UGT1A3, UGT1A6 and UGT1A9) were expressed in baculovirus-infected insect cells that, where were obtained from Corning, Inc. All other reagents were of the highest analytical grade commercially available. The specific reagents were sourced from companies mentioned previously (33).

Inhibition of recombinant UGTs-catalyzed 4-MU glucuronidation by quercetin and Q3GA

The experimental protocol and incubation has been accurately presented in previous studies (34-36). Typical incubations were performed in 200 µl reaction mixture containing 5 mM UDPGA, 5 mM MgCl2, 50 mM Tris-HCl buffer (pH 7.4), 4-MU and recombinant UGTs. In addition, a series of quercetin concentrations and Q3GA were dissolved in dimethyl sulfoxide (DMSO) and the final concentration of DMSO in the total mixture was <0.5% (v/v). Table I indicates the final concentration of UGT1A isoforms and 4-Mu, and the incubation time used for each UGT enzyme. Following pre-incubation at 37˚C for 5 min, the UDPGA was added in the incubation system to initiate the reaction. The samples were incubated for the aforementioned incubation time periods and the reaction was terminated by the addition of 200 µl ice-cold methanol, containing 500 ng·ml-1 7-hydroxycoumarin as an internal standard. Subsequently, the samples were centrifuged at 20,000 x g for 10 min at 4˚C, and 100 µl supernatant was obtained and injected into the LC-MS/MS system for analysis.

Table I

Substrate concentration, enzyme concentration and incubation time of each UGT enzyme.

Table I

Substrate concentration, enzyme concentration and incubation time of each UGT enzyme.

UGT enzymeEnzyme concentration, mg/mlIncubation time, minSubstrate concentration, µM
UGT1A10.12512030
UGT1A30.051201,200
UGT1A60.02530110
UGT1A90.053030

[i] UGT, UDP-glucuronosyltransferase.

Detection of 4-MU-G by LC-MS/MS

4-MU-G and 7-hydroxycoumarin (internal standard) were analyzed on an API-4000 triple quadruple mass spectrometer (Applied Biosystems; Thermo Fisher Scientific, Inc.) coupled with a Waters ACQUITY Ultra Performance Liquid Chromatograph (Waters Corporation). The separation was performed on an Inertsil ODS-SP column (100x2.1 mm; 3 µm; GL Sciences) with a column temperature of 40˚C. The mobile phase consisted of ultrapure water, containing 0.1% formic acid (A) and acetonitrile containing 0.1% formic acid (B). The following gradient conditions were used: 0-4.00 min, 5-80% B; 4.00-4.10 min, 80-5% B; 4.10-7.00 min, 5% B. The flow rate used was 0.2 ml/min, and the LC retention times of 4-MU-G and 7-hydroxycoumarin were 3.7 and 3.53 min, respectively. The turbo ion spray interface was operated at -4,500 V and the ion source temperature was set at 500℃ in the negative electrospray ionization mode. The multiple reaction monitoring (MRM) mode was employed for quantification using specific precursor/product ion transition. The precursor/product ion transitions were monitored at m/z 351.1→175.1 and 161.0→89.0 for 4-MU-G and 7-hydroxycoumarin, respectively. The optimized working parameters for mass detection of 4-MU-G and 7-hydroxycoumarin were as follows: i) Declustering potential, -50 and -80 V; ii) collision energy, -19 and -40 V; iii) curtain gas, 30 psi; iv) collision activated dissociation gas, 8 psi; v) Gas1, 55 psi and Gas2, 55 psi. The peak areas for all analytes were automatically integrated using the Analyst software (version 1.5.1; Applied Biosystems).

The specificity of this method was optimal. The linear range was estimated to be 50-5,000 ng/ml with the lower limit of quantification at 50 ng/ml. The RSD% of the intra-assay and inter-assay precisions were both <10%. The extraction recovery ranged between 100.99 and 106.34%. The internal standard normalized matrix factors for the low-, moderate- and high-quality control samples were 1.02, 1.07 and 0.99, respectively. The residues were negligible, and the samples were placed in a sampler at 4˚C for 9 h and left at room temperature for 2 h.

Determination of inhibition kinetic parameters of quercetin and Q3GA on recombinant UGTs

The glucuronidation velocity was determined at various 4-MU, quercetin or Q3GA concentrations. A preliminary screening experiment was performed to assess the inhibitory effects of quercetin and Q3GA. A total of 50 µM was selected for quercetin and Q3GA as the experimental group concentration. The remaining activity of UGTs=average concentration of 4-MUGUGT1A enzyme-quercetin/average concentration of 4-MUGUGT1A enzyme-blank x100%. The remaining activity of the blank group without quercetin and Q3GA was 100% in the incubation system. Five or six concentration ranges of quercetin and Q3GA were selected to assess the IC50 values for the UGT1A enzymes. The concentration ranges of quercetin used were as follows: 0-10 µM for UGT1A1, 0-20 µM for UGT1A3 and 0-50 µM for UGT1A6 and UGT1A9. In addition, 0-50 µM Q3GA was selected for UGT1A1, UGT1A3 and UGT1A6. The IC50 values of quercetin and Q3GA towards UGT1A enzyme activities were calculated by nonlinear regression analysis using the GraphPad Prism 5 (GraphPad Software, Inc.). When the remaining activities of these enzymes in the experimental groups were <50% of the control group, the Ki was calculated. The Ki resulted from fitting data into competitive inhibition, non-competitive inhibition, or mixed inhibition models. The type of inhibition was assessed graphically from the Lineweaver-Burk and Dixon plots (37).

Results

Inhibitory activities of quercetin and Q3GA on recombinant UGT1A isoforms

As shown in Table II, in the presence of 50 µM quercetin, the remaining activity of UGT1A1, UGT1A3, UGT1A6 and UGT1A9 enzymes were <8.4, <30.8, 23.1 and 11.7%, respectively, while the remaining activity of Q3GA on UGT1A1, UGT1A3, UGT1A6 and UGT1A9 was 36.1, 33.9, 28.4 and 74.2%, respectively. As the remaining activity of UGT1A isoforms was <50%, except for UGTIA9, quercetin and Q3GA exhibited inhibitory effects on UGT1A1, UGT1A3, UGT1A6 and UGT1A9 enzymes.

Table II

Preliminary inhibition screening of quercetin and Q3GA toward activities of recombinant UGT1A isoforms.

Table II

Preliminary inhibition screening of quercetin and Q3GA toward activities of recombinant UGT1A isoforms.

 Remaining enzyme activity
UGT1A enzymeQuercetin, (%)Q3GA, (%)
UGT1A1<8.436.1
UGT1A3<30.833.9
UGT1A623.128.4
UGT1A911.774.2

[i] Q3GA, quercetin-3-O-β-D-glucoside; UGT, UDP-glucuronosyltransferase.

In the presence of 50 µM quercetin, the final concentrations of the metabolite 4-MUG were too low to detect in UGT1A1 and UGT1A3 incubation systems. However, the lowest limit of quantitation for 4-MUG was 50 ng/ml in the present study. The remaining activity of UGT1A1 enzymes <50 ng/ml/593 ng/mlx100%=8.4%, and the remaining activity of UGT1A1 enzymes <50 ng/ml/162.3 ng/mlx100%=30.8%. Therefore, the remaining activities of UGT1A1 and UGT1A3 were less than 8.4 and 30.8%, respectively.

Inhibition type and kinetics of quercetin towards UGT1A1, UGT1A3, UGT1A6 and UGT1A9

The inhibitory parameters of quercetin on these UGT isoforms were characterized/calculated by further kinetic experiments. Quercetin inhibited UGT1A1, UGT1A3, UGT1A6 and UGT1A9 activity in a dose-dependent manner and the IC50 values were 7.47, 10.58, 7.07 and 2.81 µM, respectively (Figs. 2A, 3A, 4A and 5A). Furthermore, the inhibition types were determined from Dixon and Lineweaver-Burk plots. As shown in Fig. 2B and D and in Fig. 4B and D, the inhibitory effect of quercetin on UGT1A1 and UGT1A6-catalyzed 4-MU glucuronidation was characterized as non-competitive inhibition, while the inhibitory activity of quercetin on UGT1A3 and UGT1A6 was characterized as competitive inhibition (Figs. 3B and D; 5B and D). The calculated Ki were 2.18, 1.60, 28.87 and 0.51 µM for the inhibitory effect of quercetin on UGT1A1, UGT1A3, UGT1A6 and UGT1A9, respectively (Figs. 2C, 3C, 4C and 5C).

Inhibition type and kinetics of Q3GA towards UGT1A1, UGT1A3 and UGT1A6

Preliminary screening experiments indicated that Q3GA exhibited inhibitory effects on UGT1A1, UGT1A3 and UGT1A6. The parameters of the kinetic experiments are presented in Fig. 6. The inhibitory activity of Q3GA on UGT1A1, UGT1A3 and UGT1A6 enzymes was dose-dependent, with IC50 values of 45.21, 106.5 and 51.37 µM, respectively. Therefore, Q3GA may display weakly inhibit UGT1A1, UGT1A3 and UGT1A6.

Discussion

In recent years, dietary supplements and alternative medicine therapies have become increasingly accepted. A previous study (38) has demonstrated that the absorption and disposition of the active ingredients of dietary supplements are significant in determining their biological activity. In addition, accumulated evidence with regard to HDIs between drug-metabolizing enzymes and drug transporters may be used to predict the pharmacokinetic profile of drugs and the underlying HDIs (39,40). These potential interactions may cause significant risks to the patients, particularly for drugs with narrow therapeutic indices.

It has been demonstrated that quercetin may prevent cyclosporine A-induced nephrotoxicity and hepatotoxicity (41,42) as it exerts various effects on the pharmacokinetics of cyclosporine A. However, the underlying mechanisms for these interactions remain unclear. Our previous studies have demonstrated that Q3GA increases the Cmax, AUC0-t and AUC0-∞ of cyclosporine A (19,43). In order to further elucidate the underlying mechanisms of quercetin and Q3GA on the pharmacokinetics of cyclosporine A, the inhibitory effects of quercetin and Q3GA on the enzyme activity of recombinant UGT1A isoforms were investigated in vitro.

The inhibitory potential of the compounds was determined based on their Ki values and they were classified as potent inhibitors (Ki<1 µM), moderate inhibitors (1 µM < Ki<10 µM) and weak inhibitors (Ki>10 µM). Based on the IC50 values of Q3GA on UGT1A1, UGT1A3 and UGT1A6 enzymes that were much higher than 10 µM, and the fact that the remaining activity of UGT1A9 was >50% following Q3GA incubation, these data suggested that Q3GA displayed limited inhibitory effects on these UGT isoforms. Therefore, the subsequent enzymatic kinetics experiments were not performed, so the inhibition constant Ki of Q3GA on UGT1A enzymes could not be calculated. However, quercetin was more active in suppressing the activity of UGT isoforms compared with Q3GA. Its inhibitory effect on UGT1A1 and UGT1A6 was noncompetitive, with IC50 values of 7.47 and 7.07 µM, respectively, and Ki values of 2.18 and 28.87 µM, respectively. By contrast to these two isoforms, quercetin inhibited UGT1A3 and UGT1A9 competitively with IC50 values of 10.58 and 2.81 µM, respectively, and Ki values of 1.60 and 0.51 µM, respectively. These results indicated that quercetin was a moderate inhibitor of UGT1A1 and UGT1A3, and a strong inhibitor of UGT1A9, while Q3GA exhibited weak inhibitory effects on the activity of UGT1A1, UGT1A3, UGT1A6 and UGT1A9 isoforms. There was a small slope in Fig. 4C, which may be due to the fact that quercetin only exerted a weak inhibitory effect on UGT1A6. Furthermore, in the UGT1A3 incubation system, the inhibitory effect of 5 µM quercetin did not have a linear relationship with the inhibitory effect of other concentrations of quercetin, which may be the reason for the small slope in Fig. 3C.

Quercetin and Q3GA exhibited similar inhibitory effects on the protein and mRNA expression levels of UGT1A1 in the small intestine and the liver. This is consistent with our preliminary data and previously published studies (19,43). It has also been reported that quercetin inhibits glucuronidation of ethanol in human liver microsomes and in recombinant UGT1A1, UGT1A3, UGT 1A4, UGT1A6 and UGT1A9 enzymes (44). However, quercetin has been reported to induce UGT1A6 mRNA expression in human intestinal tissues in vitro (45). Notably, quercetin induces UGT1A expression in Caco-2 cells (46). It has also been reported that this flavonoid increases UGT enzyme activities in hepatic, and small and large intestinal tissues of male Wistar rats (47). A previous study has reported that the age-associated differences in the UGT-catalyzed glucuronidation of quercetin depend on the intestinal segment, particularly the proximal and distal segments (48). In brief, humans and animals express different protein and activity levels of UGT1A isoforms.

There are limitations to the present study, including the fact that quercetin or Q3GA were added separately to the UGT1A incubation systems evaluate their inhibitory effects. However, to the best of our knowledge, there are no effective methods to distinguish the role of the quercetin/Q3GA vs. quercetin plus Q3GA, as quercetin may be metabolized to Q3GA by UGT1A in the incubation (49-51), and Q3GA may be hydrolyzed to quercetin in the incubation in activated mouse macrophages (49). The results of the present study demonstrated that the inhibitory effects of quercetin and Q3GA on UGT1A enzymes were significantly different. Additionally, compared with quercetin, the inhibitory effect of Q3GA on UGT1A enzymes was very weak. Therefore, we hypothesized that Q3GA had a separate inhibitory effect on the UGT1A enzymes, rather than inhibiting the UGT1A enzymes through hydrolysis to quercetin.

A previous study (52) has reported that 3-hydroxyflavone has a higher catalytic rate than 7-hydroxyflavone, due to differences in the hydroxyl positions. Therefore, following conjugation of the C-3 hydroxyl group with the glucuronic acid to yield Q3GA, a decrease in the catalytic rate of Q3GA may be noted. In addition, Boersma et al (53) reported that quercetin inhibited UGT1A6 less efficiently than luteolin, which was possibly due to the lack of the C-3 hydroxyl group. This result suggested that flavonoids without the C-3 hydroxyl group exhibit a significant inhibitory effect on the UGT1A6-catalyzed reaction. This blocking effect is more pronounced for UGT1A6 when a larger group is conjugated with the C-3 hydroxyl group. In general, when Q3GA and quercetin are combined with UGT1As, the steric hindrance effect of Q3GA greatly decreases the rate and extent of this catalytic reaction.

In conclusion, the results of the present study indicated that the inhibitory effect of quercetin on UGT1A1, UGT1A3, UGT1A6 and UGT1A9 was more potent than that of Q3GA. Furthermore, the results demonstrated that quercetin was a moderate inhibitor of UGT1A1 and UGT1A3, and a strong inhibitor of UGT1A9, and therefore HDIs may occur when quercetin is co-administered with drugs that are mainly metabolized by UGT1A1, UGT1A3 and UGT1A9.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the National Natural Science Foundation of China (grant nos. 81874326 and 81503161); The Chinese Medicine Research Project of Health Commission of Hubei Province (grant no. ZY2019Z004); and the National Key R&D Program of China (grant no. 2017YFC0909900).

Availability of data and materials

All data generated and/or analyzed during this study are included in this published article.

Authors' contributions

RZ, YL and SS designed all the experiments. YW and TY performed the incubation experiments. Statistical analysis was performed by XH, JZ, JNZ and CY. YL and SS confirm the authenticity of all the raw data. YW, RZ, YL and SS wrote the manuscript. All authors read and approved the final manuscript. RZ, YL and SS designed all the experiments. YW and TY performed the incubation experiments. Statistical analysis was performed by XH, JZ, JNZ and CY. YL and SS confirm the authenticity of all the raw data. YW, RZ, YL and SS wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Babaei F, Mirzababaei M and Nassiri-Asl M: Quercetin in food: Possible mechanisms of its effect on memory. J Food Sci. 83:2280–2287. 2018.PubMed/NCBI View Article : Google Scholar

2 

Costa LG, Garrick JM, Roque PJ and Pellacani C: Mechanisms of neuroprotection by quercetin: Counteracting oxidative stress and more. Oxid Med Cell Longev. 2016(2986796)2016.PubMed/NCBI View Article : Google Scholar

3 

Jana N, Břetislav G, Pavel S and Pavla U: Potential of the flavonoid quercetin to prevent and treat cancer-current status of research. Klin Onkol. 31:184–190. 2018.PubMed/NCBI View Article : Google Scholar

4 

Primikyri A, Sayyad N, Quilici G, Vrettos EI, Lim K, Chi SW, Musco G, Gerothanassis IP and Tzakos AG: Probing the interaction of a quercetin bioconjugate with Bcl-2 in living human cancer cells with in-cell NMR spectroscopy. FEBS Lett. 592:3367–3379. 2018.PubMed/NCBI View Article : Google Scholar

5 

Sharmila G, Athirai T, Kiruthiga B, Senthilkumar K, Elumalai P, Arunkumar R and Arunakaran J: Chemopreventive effect of quercetin in MNU and testosterone induced prostate cancer of sprague-dawley rats. Nutr Cancer. 66:38–46. 2014.PubMed/NCBI View Article : Google Scholar

6 

Tang SH, Li R, Tan J, Wang Y and Jiang ZT: One pot synthesis of water-soluble quercetin derived multifunctional nanoparticles with photothermal and antioxidation capabilities. Colloids Surf B Biointerfaces. 183(110429)2019.PubMed/NCBI View Article : Google Scholar

7 

Zheng YZ, Deng G, Liang Q, Chen DF, Guo R and Lai RC: Antioxidant activity of quercetin and its glucosides from propolis: A theoretical study. Sci Rep. 7(7543)2017.PubMed/NCBI View Article : Google Scholar

8 

Newton K and Dixit VM: Signaling in innate immunity and inflammation. Cold Spring Harb Perspect Biol. 4(a006049)2012.PubMed/NCBI View Article : Google Scholar

9 

Byun EB, Yang MS, Choi HG, Sung NY, Song DS, Sin SJ and Byun EH: Quercetin negatively regulates TLR4 signaling induced by lipopolysaccharide through tollip expression. Biochem Biophys Res Commun. 431:698–705. 2013.PubMed/NCBI View Article : Google Scholar

10 

Lee SM, Moon J, Chung JH, Cha YJ and Shin MJ: Effect of quercetin-rich onion peel extracts on arterial thrombosis in rats. Food Chem Toxicol. 57:99–105. 2013.PubMed/NCBI View Article : Google Scholar

11 

Pan W, Chang MJ, Booyse FM, Grenett HE, Bradley KM, Wolkowicz PE, Shang Q and Tabengwa EM: Quercetin induced tissue-type plasminogen activator expression is mediated through Sp1 and p38 mitogen-activated protein kinase in human endothelial cells. J Thromb Haemost. 6:976–985. 2008.PubMed/NCBI View Article : Google Scholar

12 

Qiu X, Kroeker A, He S, Kozak R, Audet J, Mbikay M and Chrétien M: Prophylactic efficacy of quercetin 3-β-O-d-glucoside against ebola virus infection. Antimicrob Agents Chemother. 60:5182–5188. 2016.PubMed/NCBI View Article : Google Scholar

13 

Gonzalez O, Fontanes V, Raychaudhuri S, Loo R, Loo J, Arumugaswami V, Sun R, Dasgupta A and French SW: The heat shock protein inhibitor quercetin attenuates hepatitis C virus production. Hepatology. 50:1756–1764. 2009.PubMed/NCBI View Article : Google Scholar

14 

Liu H, Guo X, Chu Y and Lu S: Heart protective effects and mechanism of quercetin preconditioning on anti-myocardial ischemia reperfusion (IR) injuries in rats. Gene. 545:149–155. 2014.PubMed/NCBI View Article : Google Scholar

15 

Lin R, Liu J, Gan W and Ding C: Protective effect of quercetin on the homocysteine-injured human umbilical vein vascular endothelial cell line (ECV304). Basic Clin Pharmacol Toxicol. 101:197–202. 2007.PubMed/NCBI View Article : Google Scholar

16 

Kobori M, Takahashi Y, Sakurai M, Akimoto Y, Tsushida T, Oike H and Ippoushi K: Quercetin suppresses immune cell accumulation and improves mitochondrial gene expression in adipose tissue of diet-induced obese mice. Mol Nutr Food Res. 60:300–312. 2016.PubMed/NCBI View Article : Google Scholar

17 

Singh D, Tanwar H, Jayashankar B, Sharma J, Murthy S, Chanda S, Singh SB and Ganju L: Quercetin exhibits adjuvant activity by enhancing Th2 immune response in ovalbumin immunized mice. Biomed Pharmacother. 90:354–360. 2017.PubMed/NCBI View Article : Google Scholar

18 

Andres S, Pevny S, Ziegenhagen R, Bakhiya N, Schäfer B, Hirsch-Ernst KI and Lampen A: Safety aspects of the use of quercetin as a dietary supplement. Mol Nutr Food Res. 62:2018.PubMed/NCBI View Article : Google Scholar

19 

Yang T, Liu Y, Huang X, Zhang R, Yang C, Zhou J, Zhang Y, Wan J and Shi S: Quercetin-3-O-β-D-glucoside decreases the bioavailability of cyclosporin A through regulation of drug metabolizing enzymes, transporters and nuclear receptors in rats. Mol Med Rep. 18:2599–2612. 2018.PubMed/NCBI View Article : Google Scholar

20 

Baral S, Pariyar R, Kim J, Lee HS and Seo J: Quercetin-3-O-glucuronide promotes the proliferation and migration of neural stem cells. Neurobiol Aging. 52:39–52. 2017.PubMed/NCBI View Article : Google Scholar

21 

Li F, Sun XY, Li XW, Yang T and Qi LW: Enrichment and separation of quercetin-3-O-β-d-glucuronide from lotus leaves (nelumbo nucifera gaertn.) and evaluation of its anti-inflammatory effect. J Chromatogr B Analyt Technol Biomed Life Sci. 1040:186–191. 2017.PubMed/NCBI View Article : Google Scholar

22 

Oliveira EJ and Watson DG: In vitro glucuronidation of kaempferol and quercetin by human UGT-1A9 microsomes. FEBS Lett. 471:1–6. 2000.PubMed/NCBI View Article : Google Scholar

23 

Zhou H, Shi R, Ma B, Ma Y, Wang C, Wu D, Wang X and Cheng N: CYP450 1A2 and multiple UGT1A isoforms are responsible for jatrorrhizine metabolism in human liver microsomes. Biopharm Drug Dispos. 34:176–185. 2013.PubMed/NCBI View Article : Google Scholar

24 

Stingl JC, Bartels H, Viviani R, Lehmann ML and Brockmöller J: Relevance of UDP-glucuronosyltransferase polymorphisms for drug dosing: A quantitative systematic review. Pharmacol Ther. 141:92–116. 2014.PubMed/NCBI View Article : Google Scholar

25 

Miners JO, Mackenzie PI and Knights KM: The prediction of drug-glucuronidation parameters in humans: UDP-glucuronosyltransferase enzyme-selective substrate and inhibitor probes for reaction phenotyping and in vitro-in vivo extrapolation of drug clearance and drug-drug interaction potential. Drug Metab Rev. 42:196–208. 2010.PubMed/NCBI View Article : Google Scholar

26 

Jeong HU, Lee JY, Kwon SS, Kim JH, Kim YM, Hong SW, Yeon SH, Lee SM, Cho YY and Lee HS: Metabolism-mediated drug interaction potential of HS-23, a new herbal drug for the treatment of sepsis in human hepatocytes and liver microsomes. Arch Pharm Res. 38:171–177. 2015.PubMed/NCBI View Article : Google Scholar

27 

Tan ML and Lim LE: The effects of Andrographis paniculata (Burm.f.) Nees extract and diterpenoids on the CYP450 isoforms' activities, a review of possible herb-drug interaction risks. Drug Chem Toxicol. 38:241–253. 2015.PubMed/NCBI View Article : Google Scholar

28 

Li G, Huang K, Nikolic D and van Breemen RB: High-throughput cytochrome P450 cocktail inhibition assay for assessing drug-drug and drug-botanical interactions. Drug Metab Dispos. 43:1670–1678. 2015.PubMed/NCBI View Article : Google Scholar

29 

Weissenstein U, Kunz M, Oufir M, Wang JT, Hamburger M, Urech K, Regueiro U and Baumgartner S: Absence of herb-drug interactions of mistletoe with the tamoxifen metabolite (E/Z)-endoxifen and cytochrome P450 3A4/5 and 2D6 in vitro. BMC Complement Altern Med. 19(23)2019.PubMed/NCBI View Article : Google Scholar

30 

Tuteja S, Pyrsopoulos NT, Wolowich WR, Khanmoradi K, Levi DM, Selvaggi G, Weisbaum G, Tzakis AG and Schiff ER: Simvastatin-ezetimibe-induced hepatic failure necessitating liver transplantation. Pharmacotherapy. 28:1188–1193. 2008.PubMed/NCBI View Article : Google Scholar

31 

Magee CN, Medani SA, Leavey SF, Conlon PJ and Clarkson MR: Severe rhabdomyolysis as a consequence of the interaction of fusidic acid and atorvastatin. Am J Kidney Dis. 56:e11–e15. 2010.PubMed/NCBI View Article : Google Scholar

32 

Huang T, Fang ZZ and Yang L: Strong inhibitory effect of medroxyprogesterone acetate (MPA) on UDP-glucuronosyltransferase (UGT) 2B7 might induce drug-drug interactions. Pharmazie. 65:919–921. 2010.PubMed/NCBI

33 

Lu H, Fang ZZ, Cao YF, Hu CM, Hong M, Sun XY, Li H, Liu Y, Fu X and Sun H: Isoliquiritigenin showed strong inhibitory effects towards multiple UDP-glucuronosyltransferase (UGT) isoform-catalyzed 4-methylumbelliferone (4-MU) glucuronidation. Fitoterapia. 84:208–212. 2013.PubMed/NCBI View Article : Google Scholar

34 

Liu C, Cao YF, Fang ZZ, Zhang YY, Hu CM, Sun XY, Huang T, Zeng J, Fan XR and Mo H: Strong inhibition of deoxyschizandrin and schisantherin A toward UDP-glucuronosyltransferase (UGT) 1A3 indicating UGT inhibition-based herb-drug interaction. Fitoterapia. 83:1415–1419. 2012.PubMed/NCBI View Article : Google Scholar

35 

Jiang HM, Fang ZZ, Cao YF, Hu CM, Sun XY, Hong M, Yang L, Ge GB, Liu Y, Zhang YY, et al: New insights for the risk of bisphenol A: Inhibition of UDP-glucuronosyltransferases (UGTs). Chemosphere. 93:1189–1193. 2013.PubMed/NCBI View Article : Google Scholar

36 

Sun D, Zhang CZ, Ran RX, Cao YF, Du Z, Fu ZW, Huang CT, Zhao ZY, Zhang WH and Fang ZZ: In vitro comparative study of the inhibitory effects of mangiferin and its aglycone norathyriol towards UDP-glucuronosyl transferase (UGT) isoforms. Molecules. 22(1008)2017.PubMed/NCBI View Article : Google Scholar

37 

Zhu L, Ge G, Liu Y, He G, Liang S, Fang Z, Dong P, Cao Y and Yang L: Potent and selective inhibition of magnolol on catalytic activities of UGT1A7 and 1A9. Xenobiotica. 42:1001–1008. 2012.PubMed/NCBI View Article : Google Scholar

38 

Miclaus MO, Filip X, Filip C, Martin FA and Grosu IG: Highly sensitive solid forms discrimination on the whole tablet of the active ingredients in quercetin dietary supplements by NMR crystallography approaches. J Pharm Biomed Anal. 124:274–280. 2016.PubMed/NCBI View Article : Google Scholar

39 

Shi S and Li Y: Interplay of drug-metabolizing enzymes and transporters in drug absorption and disposition. Curr Drug Metab. 15:915–941. 2014.PubMed/NCBI View Article : Google Scholar

40 

Wu B: Pharmacokinetic interplay of phase II metabolism and transport: A theoretical study. J Pharm Sci. 101:381–393. 2012.PubMed/NCBI View Article : Google Scholar

41 

Zal F, Mostafavi-Pour Z and Vessal M: Comparison of the effects of vitamin E and/or quercetin in attenuating chronic cyclosporine A-induced nephrotoxicity in male rats. Clin Exp Pharmacol Physiol. 34:720–724. 2007.PubMed/NCBI View Article : Google Scholar

42 

Mostafavi-Pour Z, Zal F, Monabati A and Vessal M: Protective effects of a combination of quercetin and vitamin E against cyclosporine A-induced oxidative stress and hepatotoxicity in rats. Hepatol Res. 38:385–392. 2008.PubMed/NCBI View Article : Google Scholar

43 

Liu Y, Luo X, Yang C, Yang T, Zhou J and Shi S: Impact of quercetin-induced changes in drug-metabolizing enzyme and transporter expression on the pharmacokinetics of cyclosporine in rats. Mol Med Rep. 14:3073–3085. 2016.PubMed/NCBI View Article : Google Scholar

44 

Stachel N and Skopp G: Formation and inhibition of ethyl glucuronide and ethyl sulfate. Forensic Sci Int. 265:61–64. 2016.PubMed/NCBI View Article : Google Scholar

45 

van de Kerkhof EG, de Graaf IA, Ungell AL and Groothuis GM: Induction of metabolism and transport in human intestine: Validation of precision-cut slices as a tool to study induction of drug metabolism in human intestine in vitro. Drug Metab Dispos. 36:604–613. 2008.PubMed/NCBI View Article : Google Scholar

46 

Galijatovic A, Walle UK and Walle T: Induction of UDP-glucuronosyltransferase by the flavonoids chrysin and quercetin in Caco-2 cells. Pharm Res. 17:21–26. 2000.PubMed/NCBI View Article : Google Scholar

47 

van der Logt EM, Roelofs HM, Nagengast FM and Peters WH: Induction of rat hepatic and intestinal UDP-glucuronosyltransferases by naturally occurring dietary anticarcinogens. Carcinogenesis. 24:1651–1656. 2003.PubMed/NCBI View Article : Google Scholar

48 

Bolling BW, Court MH, Blumberg JB and Chen CY: Microsomal quercetin glucuronidation in rat small intestine depends on age and segment. Drug Metab Dispos. 39:1406–1414. 2011.PubMed/NCBI View Article : Google Scholar

49 

Kawai Y, Nishikawa T, Shiba Y, Saito S, Murota K, Shibata N, Kobayashi M, Kanayama M, Uchida K and Terao J: Macrophage as a target of quercetin glucuronides in human atherosclerotic arteries: Implication in the anti-atherosclerotic mechanism of dietary flavonoids. J Biol Chem. 283:9424–9434. 2008.PubMed/NCBI View Article : Google Scholar

50 

Ishizawa K, Yoshizumi M, Kawai Y, Terao J, Kihira Y, Ikeda Y, Tomita S, Minakuchi K, Tsuchiya K and Tamaki T: Pharmacology in health food: Metabolism of quercetin in vivo and its protective effect against arteriosclerosis. J Pharmacol Sci. 115:466–470. 2011.PubMed/NCBI View Article : Google Scholar

51 

Chao CL, Hou YC, Chao PD, Weng CS and Ho FM: The antioxidant effects of quercetin metabolites on the prevention of high glucose-induced apoptosis of human umbilical vein endothelial cells. Brit J Nutr. 101:1165–1170. 2009.PubMed/NCBI View Article : Google Scholar

52 

Senafi SB, Clarke DJ and Burchell B: Investigation of the substrate specificity of a cloned expressed human bilirubin UDP-glucuro-nosyltransferase: UDP-sugar specificity and involvement in steroid and xenobiotic glucuronidation. Biochem J. 303:233–240. 1994.PubMed/NCBI View Article : Google Scholar

53 

Boersma MG, van der Woude H, Bogaards J, Boeren S, Vervoort J, Cnubben NH, van Iersel ML, van Bladeren PJ and Rietjens IM: Regioselectivity of phase II metabolism of luteolin and quercetin by UDP-glucuronosyl transferases. Chem Res Toxicol. 15:662–670. 2002.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2021
Volume 22 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang R, Wei Y, Yang T, Huang X, Zhou J, Yang C, Zhou J, Liu Y and Shi S: Inhibitory effects of quercetin and its major metabolite quercetin‑3‑O‑β‑D‑glucoside on human UDP‑glucuronosyltransferase 1A isoforms by liquid chromatography‑tandem mass spectrometry. Exp Ther Med 22: 842, 2021
APA
Zhang, R., Wei, Y., Yang, T., Huang, X., Zhou, J., Yang, C. ... Shi, S. (2021). Inhibitory effects of quercetin and its major metabolite quercetin‑3‑O‑β‑D‑glucoside on human UDP‑glucuronosyltransferase 1A isoforms by liquid chromatography‑tandem mass spectrometry. Experimental and Therapeutic Medicine, 22, 842. https://doi.org/10.3892/etm.2021.10274
MLA
Zhang, R., Wei, Y., Yang, T., Huang, X., Zhou, J., Yang, C., Zhou, J., Liu, Y., Shi, S."Inhibitory effects of quercetin and its major metabolite quercetin‑3‑O‑β‑D‑glucoside on human UDP‑glucuronosyltransferase 1A isoforms by liquid chromatography‑tandem mass spectrometry". Experimental and Therapeutic Medicine 22.2 (2021): 842.
Chicago
Zhang, R., Wei, Y., Yang, T., Huang, X., Zhou, J., Yang, C., Zhou, J., Liu, Y., Shi, S."Inhibitory effects of quercetin and its major metabolite quercetin‑3‑O‑β‑D‑glucoside on human UDP‑glucuronosyltransferase 1A isoforms by liquid chromatography‑tandem mass spectrometry". Experimental and Therapeutic Medicine 22, no. 2 (2021): 842. https://doi.org/10.3892/etm.2021.10274