183 results on '"Finel M"'
Search Results
2. Efflux transport of nicotine, cotinine and trans-3'-hydroxycotinine glucuronides by human hepatic transporters
- Author
-
Jarvinen, E., Sjostedt, N., Koenderink, J.B., Kidron, H., Finel, M., Jarvinen, E., Sjostedt, N., Koenderink, J.B., Kidron, H., and Finel, M.
- Abstract
Contains fulltext : 215788.pdf (publisher's version ) (Closed access), Nicotine is the addiction causing alkaloid in tobacco, and it is used in smoking cessation therapies. Although the metabolic pathways of nicotine are well known and mainly occur in the liver, the transport of nicotine and its metabolites is poorly characterized. The highly hydrophilic nature and urinary excretion of nicotine glucuronide metabolites indicate that hepatic basolateral efflux transporters mediate their excretion. We aimed here to find the transporters responsible for the hepatic excretion of nicotine, cotinine and trans-3'-hydroxycotinine (OH-cotinine) glucuronides. To this end, we tested their transport by multidrug resistance-associated proteins 1 (MRP1, ABCC1) and MRP3-6 (ABCC3-6), which are located on the basolateral membranes of hepatocytes, as well as MRP2 (ABCC2), breast cancer resistance protein (BCRP, ABCG2) and multidrug resistance protein 1 (MDR1, P-gp, ABCB1) that are expressed in the apical membranes of these cells. ATP-dependent transport of these glucuronides was evaluated in inside-out membrane vesicles expressing the transporter of interest. In addition, potential interactions of both the glucuronides and parent compounds with selected transporters were tested by inhibition assays. Considerable ATP-dependent transport was observed only for OH-cotinine glucuronide by MRP3. The kinetics of this transport activity was characterized, resulting in an estimated Km value of 895 micromol/L. No significant transport was found for nicotine or cotinine glucuronides by any of the tested transporters at either 5 or 50 micromol/L substrate concentration. Furthermore, neither nicotine, cotinine nor OH-cotinine inhibited MRP2-4, BCRP or MDR1. In this study, we directly examined, for the first time, efflux transport of the three hydrophilic nicotine glucuronide metabolites by the major human hepatic efflux transporters. Despite multiple transporters studied here, our results indicate that an unknown transporter may be responsible for the hepatic excreti
- Published
- 2019
3. Glucuronidation of Clopidogrel Carboxylic Acid is Mainly Mediated by Human Udp-Glucuronosyltransferases UGT2B7 and UGT2B17
- Author
-
Kahma, H., primary, Filppula, A.M., additional, Neuvonen, M., additional, Tornio, A., additional, Tarkiainen, E.K., additional, Holmberg, M.T., additional, Itkonen, M.K., additional, Finel, M., additional, Neuvonen, P.J., additional, Niemi, M., additional, and Backman, J.T., additional
- Published
- 2017
- Full Text
- View/download PDF
4. Influence of different metabolic reactions on endocrine activities of bisphenol S
- Author
-
Gramec Skledar, D., primary, Trontelj, J., additional, Klopčič, I., additional, Dolenc, M. Sollner, additional, Schmidt, J., additional, Finel, M., additional, and Mašič, L. Peterlin, additional
- Published
- 2016
- Full Text
- View/download PDF
5. The First Aspartic Acid of the DQxD Motif for Human UDP-Glucuronosyltransferase 1A10 Interacts with UDP-Glucuronic Acid during Catalysis
- Author
-
Redinbo, M. R., Goldman, A., Finel, M., Patana, A.-S., Zielinska, A. K., Miller, G. P., Radominska-Pandya, A., Miley, M. J., Bratton, S. M., and Xiong, Y.
- Abstract
All UDP-glucuronosyltransferase enzymes (UGTs) share a common cofactor, UDP-glucuronic acid (UDP-GlcUA). The binding site for UDP-GlcUA is localized to the C-terminal domain of UGTs on the basis of amino acid sequence homology analysis and crystal structures of glycosyltransferases, including the C-terminal domain of human UGT2B7. We hypothesized that the 393DQMD-NAK399 region of human UGT1A10 interacts with the glucuronic acid moiety of UDP-GlcUA. Using site-directed mutagenesis and enzymatic analysis, we demonstrated that the D393A mutation abolished the glucuronidation activity of UGT1A10 toward all substrates. The effects of the alanine mutation at Q394, D396, and K399 on glucuronidation activities were substrate-dependent. Previously, we examined the importance of these residues in UGT2B7. Although D393 (D398 in UGT2B7) is similarly critical for UDP-GlcUA binding in both enzymes, the effects of Q394 (Q399 in UGT2B7) to Ala mutation on activity were significant but different between UGT1A10 and UGT2B7. A model of the UDP-GlcUA binding site suggests that the contribution of other residues to cosubstrate binding may explain these differences between UGT1A10 and UGT2B7. We thus postulate that D393 is critical for the binding of glucuronic acid and that proximal residues, e.g., Q394 (Q399 in UGT2B7), play a subtle role in cosubstrate binding in UGT1A10 and UGT2B7. Hence, this study provides important new information needed for the identification and understanding of the binding sites of UGTs, a major step forward in elucidating their molecular mechanism.
- Published
- 2007
- Full Text
- View/download PDF
6. The effect of differentiation on the expression of UGT isoenzymes in Caco-2 cell line
- Author
-
Siissalo, S., primary, Zhang, H., additional, Hirvonen, J., additional, and Finel, M., additional
- Published
- 2008
- Full Text
- View/download PDF
7. Mutation analysis in UGT1A9 suggests a relationship between substrate and catalytic residues in UDP-glucuronosyltransferases
- Author
-
Patana, A.-S., primary, Kurkela, M., additional, Finel, M., additional, and Goldman, A., additional
- Published
- 2008
- Full Text
- View/download PDF
8. High Throughput Screening Assay for UDP-Glucuronosyltransferase 1A1 Glucuronidation Profiling
- Author
-
Trubetskoy, O.V., primary, Finel, M., additional, Kurkela, M., additional, Fitzgerald, M., additional, Peters, N.R., additional, Hoffman, F.M., additional, and Trubetskoy, V.S., additional
- Published
- 2007
- Full Text
- View/download PDF
9. Microplate screening assay to identify inhibitors of human catechol-O-methyltransferase
- Author
-
KURKELA, M, primary, SIISKONEN, A, additional, FINEL, M, additional, TAMMELA, P, additional, TASKINEN, J, additional, and VUORELA, P, additional
- Published
- 2004
- Full Text
- View/download PDF
10. Diphenyleneiodonium inhibits reduction of iron-sulfur clusters in the mitochondrial NADH-ubiquinone oxidoreductase (Complex I).
- Author
-
Majander, A., primary, Finel, M., additional, and Wikström, M., additional
- Published
- 1994
- Full Text
- View/download PDF
11. F1F0-ATP synthase from bovine heart mitochondria: development of the purification of a monodisperse oligomycin-sensitive ATPase
- Author
-
Lutter, R, primary, Saraste, M, additional, van Walraven, H S, additional, Runswick, M J, additional, Finel, M, additional, Deatherage, J F, additional, and Walker, J E, additional
- Published
- 1993
- Full Text
- View/download PDF
12. NADH:ubiquinone oxidoreductase from bovine heart mitochondria. cDNA sequences of the import precursors of the nuclear-encoded 39 kDa and 42 kDa subunits
- Author
-
Fearnley, I M, primary, Finel, M, additional, Skehel, J M, additional, and Walker, J E, additional
- Published
- 1991
- Full Text
- View/download PDF
13. Purification of the 45 kDa, membrane bound NADH dehydrogenase of Escherichia coli (NDH-2) and analysis of its interaction with ubiquinone analogues
- Author
-
Bjorklof, K., Zickermann, V., and Finel, M.
- Published
- 2000
- Full Text
- View/download PDF
14. Deletion of the gene for subunit III leads to defective assembly of bacterial cytochrome oxidase.
- Author
-
Haltia, T., Finel, M., Harms, N., Nakari, T., Raitio, M., Wikström, M., and Saraste, M.
- Abstract
COIII is one of the major subunits in the mitochondrial and a bacterial cytochrome c oxidase, cytochrome aa3. It does not contain any of the enzyme's redox‐active metal centres and can be removed from the enzyme without major changes in its established functions. We have deleted the COIII gene from Paracoccus denitrificans. The mutant still expresses spectroscopically detectable enzyme almost as the wild‐type, but its cytochrome c oxidase activity is much lower. From 50 to 80% of cytochrome a is reduced and its absorption maximum is 2‐3 nm blue‐shifted. The EPR signal of ferric cytochrome a is heterogeneous indicating the presence of multiple cytochrome a species. Proteolysis of the membrane‐bound oxidase shows new cleavage sites both in COI and COII. DEAE‐chromatography of solubilized enzyme yields fractions that contain a COI + COII complex and in addition haem‐binding, free COI as well as free COII. The mutant phenotype can be complemented by introducing the COIII gene back to cells in a plasmid vector. We conclude that cytochrome oxidase assembles inefficiently in the absence of COIII and that this subunit may facilitate a late step in the assembly. The different oxidase species in the mutant represent either accumulating intermediates of the assembly pathway or dissociation products of a labile COI + COII complex and its conformational variants.
- Published
- 1989
- Full Text
- View/download PDF
15. Genome-wide analyses of neonatal jaundice reveal a marked departure from adult bilirubin metabolism.
- Author
-
Solé-Navais P, Juodakis J, Ytterberg K, Wu X, Bradfield JP, Vaudel M, LaBella AL, Helgeland Ø, Flatley C, Geller F, Finel M, Zhao M, Lazarus P, Hakonarson H, Magnus P, Andreassen OA, Njølstad PR, Grant SFA, Feenstra B, Muglia LJ, Johansson S, Zhang G, and Jacobsson B
- Subjects
- Humans, Infant, Newborn, Adult, Female, Male, Polymorphism, Single Nucleotide, Genetic Predisposition to Disease, Norway, Quantitative Trait Loci, Alleles, Mutation, Missense, Liver metabolism, White People genetics, Jaundice, Neonatal genetics, Jaundice, Neonatal metabolism, Genome-Wide Association Study, Bilirubin metabolism, Glucuronosyltransferase genetics, Glucuronosyltransferase metabolism
- Abstract
Jaundice affects almost all neonates in their first days of life and is caused by the accumulation of bilirubin. Although the core biochemistry of bilirubin metabolism is well understood, it is not clear why some neonates experience more severe jaundice and require treatment with phototherapy. Here, we present the first genome-wide association study of neonatal jaundice to date in nearly 30,000 parent-offspring trios from Norway (cases ≈ 2000). The alternate allele of a common missense variant affecting the sequence of UGT1A4 reduces the susceptibility to jaundice five-fold, which replicated in separate cohorts of neonates of African American and European ancestries. eQTL colocalization analyses indicate that the association may be driven by regulation of UGT1A1 in the intestines, but not in the liver. Our results reveal marked differences in the genetic variants involved in neonatal jaundice compared to those regulating bilirubin levels in adults, suggesting distinct genetic mechanisms for the same biological pathways., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
16. Human UDP-glucuronosyltransferase 1As catalyze aristolochic acid D O-glucuronidation to form a lesser nephrotoxic glucuronide.
- Author
-
Tu DZ, Liu PQ, Zhu GH, Zeng HR, Deng YY, Huang J, Niu XT, Liu YF, Hu J, Liang XM, Finel M, Wang P, and Ge GB
- Subjects
- Humans, Glucuronides metabolism, Microsomes, Liver metabolism, Reactive Oxygen Species metabolism, Glucuronosyltransferase metabolism, Kinetics, Catalysis, Uridine Diphosphate metabolism, Aristolochic Acids toxicity, Mitochondrial Diseases
- Abstract
Ethnopharmacological Relevance: Aristolochic acids (AAs) are naturally occurring nitro phenanthrene carboxylic acids primarily found in plants of the Aristolochiaceae family. Aristolochic acid D (AAD) is a major constituent in the roots and rhizomes of the Chinese herb Xixin (the roots and rhizomes of Asarum heterotropoides F. Schmidt), which is a key material for preparing a suite of marketed Chinese medicines. Structurally, AAD is nearly identical to the nephrotoxic aristolochic acid I (AAI), with an additional phenolic group at the C-6 site. Although the nephrotoxicity and metabolic pathways of AAI have been well-investigated, the metabolic pathway(s) of AAD in humans and the influence of AAD metabolism on its nephrotoxicity has not been investigated yet., Aim of the Study: To identify the major metabolites of AAD in human tissues and to characterize AAD O-glucuronidation kinetics in different enzyme sources, as well as to explore the influence of AAD O-glucuronidation on its nephrotoxicity., Materials and Methods: The O-glucuronide of AAD was biosynthesized and its chemical structure was fully characterized by both
1 H-NMR and13 C-NMR. Reaction phenotyping assays, chemical inhibition assays, and enzyme kinetics analyses were conducted to assess the crucial enzymes involved in AAD O-glucuronidation in humans. Docking simulations were performed to mimic the catalytic conformations of AAD in human UDP-glucuronosyltransferases (UGTs), while the predicted binding energies and distances between the deprotonated C-6 phenolic group of AAD and the glucuronyl moiety of UDPGA in each tested human UGT isoenzyme were measured. The mitochondrial membrane potentials (MMP) and reactive oxygen species (ROS) levels in HK-2 cells treated with either AAI, or AAD, or AAD O-glucuronide were tested, to elucidate the impact of O-glucuronidation on the nephrotoxicity of AAD., Results: AAD could be rapidly metabolized in human liver and intestinal microsomes (HLM and HIM, respectively) to form a mono-glucuronide, which was purified and fully characterized as AAD-6-O-β-D-glucuronide (AADG) by NMR. UGT1A1 was the predominant enzyme responsible for AAD-6-O-glucuronidation, while UGT1A9 contributed to a lesser extent. AAD-6-O-glucuronidation in HLM, HIM, UGT1A1 and UGT1A9 followed Michaelis-Menten kinetics, with the Km values of 4.27 μM, 9.05 μM, 3.87 μM, and 7.00 μM, respectively. Docking simulations suggested that AAD was accessible to the catalytic cavity of UGT1A1 or UGT1A9 and formed catalytic conformations. Further investigations showed that both AAI and AAD could trigger the elevated intracellular ROS levels and induce mitochondrial dysfunction and in HK-2 cells, but AADG was hardly to trigger ROS accumulation and mitochondrial dysfunction., Conclusion: Collectively, UGT1A-catalyzed AAD 6-O-glucuronidation represents a crucial detoxification pathway of this naturally occurring AAI analogs in humans, which is very different from that of AAI., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
17. Anthraquinone biocolourant dermocybin is metabolized whereas dermorubin is not in in vitro liver fractions and recombinant metabolic enzymes.
- Author
-
Yli-Öyrä J, Juvonen RO, Lehtonen M, Herrala M, Finel M, Räisänen R, and Rysä J
- Subjects
- Humans, Animals, Rats, Mice, Swine, Male, Recombinant Proteins metabolism, Liver metabolism, Liver enzymology, Cytosol metabolism, Oxidation-Reduction, Glucuronides metabolism, Microsomes, Liver metabolism, Glucuronosyltransferase metabolism, Cytochrome P-450 Enzyme System metabolism, Anthraquinones metabolism
- Abstract
Fungal anthraquinones dermocybin and dermorubin are attractive alternatives for synthetic dyes but their metabolism is largely unknown. We conducted a qualitative in vitro study to identify their metabolism using human liver microsomes and cytosol, as well as recombinant human cytochrome P450 (CYP), UDP-glucuronosyltransferase (UGT) and sulfotransferase (SULT) enzymes. Additionally, liver microsomal and cytosolic fractions from rat, mouse and pig were used. Following incubations of the biocolourants with the enzymes in the presence of nicotinamide adenine dinucleotide phosphate, UDP-glucuronic acid, 3'-phosphoadenosine-5'-phosphosulfate (PAPS) or S-adenosyl methionine (SAM) to enable CYP oxidation, glucuronidation, sulfonation or methylation, we observed several oxidation and conjugation metabolites for dermocybin but none for dermorubin. Human CYP1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4 and 3A7 catalysed dermocybin oxidation. The formation of dermocybin glucuronides was catalysed by human UGT1A1, 1A3, 1A7, 1A8, 1A9, 1A10 and 2B15. Human SULT1B1, 1C2 and 2A1 sulfonated dermocybin. Dermocybin oxidation was faster than conjugation in human liver microsomes. Species differences were seen in dermocybin glucuronidation between human, rat, mouse and pig. In conclusion, many CYP and conjugation enzymes metabolized dermocybin, whereas dermorubin was not metabolized in human liver fractions in vitro. The results indicate that dermocybin would be metabolized in humans in vivo., (© 2024 The Authors. Basic & Clinical Pharmacology & Toxicology published by John Wiley & Sons Ltd on behalf of Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society).)
- Published
- 2024
- Full Text
- View/download PDF
18. Characterization of the glucuronidating pathway of pectolinarigenin, the major active constituent of the Chinese medicine Daji, in humans and its influence on biological activities.
- Author
-
Liu P, Li Q, Zhu G, Zhang T, Tu D, Zhang F, Finel M, He Y, and Ge G
- Subjects
- Humans, Mice, Animals, Medicine, Chinese Traditional, NF-E2-Related Factor 2 metabolism, PPAR alpha metabolism, Glucuronosyltransferase metabolism, Microsomes, Liver metabolism, Kinetics, Glucuronides metabolism, PPAR-beta metabolism
- Abstract
Ethnopharmacological Relevance: The Chinese medicine Daji (the aerial part of Cirsium japonicum DC.) and its charred product (Cirsii Japonici Herba Carbonisata) have been widely used as hemostatic agents or diuretic agents to prepare a variety of Chinese herbal formula. Pectolinarigenin (PEC), one of the most abundant constituents in both Daji and its charred product, has been considered as the key effective substance responsible for the major pharmacological activities of Daji, including hemostasis, hepatoprotective, anti-tumor and anti-osteoporosis effects. However, the major metabolic pathways of PEC in humans and the influence of PEC metabolism on its biological activities are poorly understood., Aim of the Study: To characterize the main metabolic pathway(s) and key enzymes of PEC in human biological systems, as well as to reveal the influence of PEC metabolism on its biological activities., Materials and Methods: The metabolic stability assays of PEC were investigated in human liver microsomes (HLM). The O-glucuronide of PEC was biosynthesized and characterized by nuclear magnetic resonance (NMR) spectroscopy. The key enzymes responsible for O-glucuronidation of PEC in humans were assigned by performing UGT reaction phenotyping, chemical inhibition and enzymatic kinetic assays. The agonist effects of PEC and its O-glucuronide on nuclear factor erythroid2-related factor 2 (Nrf2), Peroxisome proliferator activated receptors (PPARα and PPARβ) were tested at the cellular level., Results: PEC could be readily metabolized to form a mono-O-glucuronide in both human liver microsome (HLM) and human intestinal microsome (HIM). The mono-O-glucuronide was bio-synthesized by mouse liver S9 and its structure was fully characterized as PEC-7-O-β-D-glucuronide (PEC-O-7-G). UGT1A1, UGT1A3 and UGT1A9 are key enzymes responsible for PEC-7-O-glucuronidation in HLM, while UGT1A1, UGT1A9 and 1A10 may play key roles in this reaction in HIM. Biological tests revealed that PEC displayed strong agonist effects on Nrf2, PPARα and PPARβ, whereas PEC-7-O-glucuronide showed relatively weak Nrf2 agonist effect and very weak PPAR agonist effects, indicating that PEC-7-O-glucuronidation strongly weaken its agonist effects on Nrf2 and PPAR., Conclusions: Our results demonstrate that 7-O-glucuronidation is the major metabolic pathway of PEC in human tissues, while UGT1A1, 1A3 and 1A9 are key contributing enzymes responsible for PEC-7-O-glucuronidation in human liver. It is also found that PEC 7-O-glucuronidation significantly weakens the Nrf2 and PPAR agonist effects. All these findings are very helpful for the pharmacologists to deep understand the metabolic rates of PEC in humans., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
19. Fluorescence-Based High-Throughput Assays for Investigating Cytochrome P450 Enzyme-Mediated Drug-Drug Interactions.
- Author
-
He R, Dai Z, Finel M, Zhang F, Tu D, Yang L, and Ge G
- Subjects
- Humans, Drug Interactions, Herb-Drug Interactions, Cytochrome P-450 Enzyme System metabolism, High-Throughput Screening Assays
- Abstract
The cytochrome P450 enzymes (CYPs), a group of heme-containing enzymes, catalyze oxidative metabolism of a wide range of drugs and xenobiotics, as well as different endogenous molecules. Strong inhibition of human CYPs is the most common cause of clinically associated pharmacokinetic drug-drug/herb-drug interactions (DDIs/HDIs), which may result in serious adverse drug reactions, even toxicity. Accurate and rapid assessing of the inhibition potentials on CYP activities for therapeutic agents is crucial for the prediction of clinically relevant DDIs/HDIs. Over the past few decades, significant efforts have been invested into developing optical substrates for the human CYPs, generating a variety of powerful tools for high-throughput assays to detect CYP activities in biologic specimens and for screening of CYP inhibitors. This minireview focuses on recent advances in optical substrates developments for human CYPs, as well as their applications in screening CYP inhibitors and DDIs/HDIs studies. The examples for rational design and optimization of highly specific optical substrates for the target CYP enzyme, as well as applications in investigating CYP-mediated DDIs, are illustrated. Finally, the challenges and future perspectives in this field are proposed. Collectively, this review summarizes the reported optical-based biochemical assays for highly efficient CYP activities detection, which strongly facilitated the discovery of CYP inhibitors and the investigations on CYP-mediated DDIs. SIGNIFICANCE STATEMENT: Optical substrates for cytochrome P450 enzymes (CYPs) have emerged as powerful tools for the construction of high-throughput assays for screening of CYP inhibitors. This mini-review covers the advances and challenges in the development of highly specific optical substrates for sensing human CYP isoenzymes, as well as their applications in constructing fluorescence-based high-throughput assays for investigating CYP-mediated drug-drug interactions., (Copyright © 2023 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2023
- Full Text
- View/download PDF
20. Optical substrates for drug-metabolizing enzymes: Recent advances and future perspectives.
- Author
-
Jin Q, Wu J, Wu Y, Li H, Finel M, Wang D, and Ge G
- Abstract
Drug-metabolizing enzymes (DMEs), a diverse group of enzymes responsible for the metabolic elimination of drugs and other xenobiotics, have been recognized as the critical determinants to drug safety and efficacy. Deciphering and understanding the key roles of individual DMEs in drug metabolism and toxicity, as well as characterizing the interactions of central DMEs with xenobiotics require reliable, practical and highly specific tools for sensing the activities of these enzymes in biological systems. In the last few decades, the scientists have developed a variety of optical substrates for sensing human DMEs, parts of them have been successfully used for studying target enzyme(s) in tissue preparations and living systems. Herein, molecular design principals and recent advances in the development and applications of optical substrates for human DMEs have been reviewed systematically. Furthermore, the challenges and future perspectives in this field are also highlighted. The presented information offers a group of practical approaches and imaging tools for sensing DMEs activities in complex biological systems, which strongly facilitates high-throughput screening the modulators of target DMEs and studies on drug/herb‒drug interactions, as well as promotes the fundamental researches for exploring the relevance of DMEs to human diseases and drug treatment outcomes., (© 2022 Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences. Production and hosting by Elsevier B.V.)
- Published
- 2022
- Full Text
- View/download PDF
21. A broad-spectrum substrate for the human UDP-glucuronosyltransferases and its use for investigating glucuronidation inhibitors.
- Author
-
Zhou QH, Qin WW, Finel M, He QQ, Tu DZ, Wang CR, and Ge GB
- Subjects
- Animals, Benzodioxoles chemistry, Dogs, Drug Evaluation, Preclinical methods, Drug Interactions, Enzyme Inhibitors metabolism, Female, Glucuronides chemistry, Glucuronides metabolism, Glucuronosyltransferase chemistry, Humans, Isoflavones chemistry, Kinetics, Macaca fascicularis, Male, Mice, Microsomes, Liver metabolism, Rabbits, Rats, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Substrate Specificity, Benzodioxoles metabolism, Enzyme Inhibitors pharmacology, Glucuronosyltransferase antagonists & inhibitors, Glucuronosyltransferase metabolism, Isoflavones metabolism
- Abstract
Strong inhibition of the human UDP-glucuronosyltransferase enzymes (UGTs) may lead to undesirable effects, including hyperbilirubinaemia and drug/herb-drug interactions. Currently, there is no good way to examine the inhibitory effects and specificities of compounds toward all the important human UGTs, side-by-side and under identical conditions. Herein, we report a new, broad-spectrum substrate for human UGTs and its uses in screening and characterizing of UGT inhibitors. Following screening a variety of phenolic compound(s), we have found that methylophiopogonanone A (MOA) can be readily O-glucuronidated by all tested human UGTs, including the typical N-glucuronidating enzymes UGT1A4 and UGT2B10. MOA-O-glucuronidation yielded a single mono-O-glucuronide that was biosynthesized and purified for structural characterization and for constructing an LC-UV based MOA-O-glucuronidation activity assay, which was then used for investigating MOA-O-glucuronidation kinetics in recombinant human UGTs. The derived K
m values were crucial for selecting the most suitable assay conditions for assessing inhibitory potentials and specificity of test compound(s). Furthermore, the inhibitory effects and specificities of four known UGT inhibitors were reinvestigated by using MOA as the substrate for all tested UGTs. Collectively, MOA is a broad-spectrum substrate for the human UGTs, which offers a new and practical tool for assessing inhibitory effects and specificities of UGT inhibitors., (Copyright © 2021 Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
22. A fluorescence-based microplate assay for high-throughput screening and evaluation of human UGT inhibitors.
- Author
-
Zhou QH, Lv X, Tian ZH, Finel M, Feng L, Huo PC, Zhu YD, Lu Y, Hou J, and Ge GB
- Subjects
- Drug Interactions, Glucuronides, Glucuronosyltransferase, Humans, Microsomes, Liver, Enzyme Inhibitors pharmacology, High-Throughput Screening Assays
- Abstract
Human UDP-glucuronosyltransferase enzymes (hUGTs), one of the most important classes of conjugative enzymes, are responsible for the glucuronidation and detoxification of a variety of endogenous substances and xenobiotics. Inhibition of hUGTs may cause undesirable effects or adverse drug-drug interactions (DDI) via modulating the glucuronidation rates of endogenous toxins or the drugs that are primarily conjugated by the inhibited hUGTs. Herein, to screen hUGTs inhibitors in a more efficient way, a novel fluorescence-based microplate assay has been developed by utilizing a fluorogenic substrate. Following screening of series of 4-hydroxy-1,8-naphthalimide derivatives, we found that 4-HN-335 is a particularly good substrate for a panel of hUGTs. Under physiological conditions, 4-HN-335 can be readily O-glucuronidated by ten hUGTs, such reactions generate a single O-glucuronide with a high quantum yield (Ф = 0.79) and bring remarkable changes in fluorescence emission. Subsequently, a fluorescence-based microplate assay is developed to simultaneously measure the inhibitory effects of selected compound(s) on ten hUGTs. The newly developed fluorescence-based microplate assay is time- and cost-saving, easy to manage and can be adapted for 96-well microplate format with the Z-factor of 0.92. We further demonstrate the utility of the fluorescence-based assay for high-throughput screening of two compound libraries, resulting in the identification of several potent UGT inhibitors, including natural products and FDA-approved drugs. Collectively, this study reports a novel fluorescence-based microplate assay for simultaneously sensing the residual activities of ten hUGTs, which strongly facilitates the identification and characterization of UGT inhibitors from drugs or herbal constituents and the investigations on UGT-mediated DDI., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Neobavaisoflavone Induces Bilirubin Metabolizing Enzyme UGT1A1 via PPARα and PPARγ.
- Author
-
Zhu YD, Guan XQ, Chen J, Peng S, Finel M, Zhao YY, Wang RM, Bi HC, Lei M, Wang DD, and Ge GB
- Abstract
UDP-glucuronosyltransferase 1A1 (UGT1A1) is an essential enzyme in mammals that is responsible for detoxification and metabolic clearance of the endogenous toxin bilirubin and a variety of xenobiotics, including some crucial therapeutic drugs. Discovery of potent and safe UGT1A1 inducers will provide an alternative therapy for ameliorating hyperbilirubinaemia and drug-induced hepatoxicity. This study aims to find efficacious UGT1A1 inducer(s) from natural flavonoids, and to reveal the mechanism involved in up-regulating of this key conjugative enzyme by the flavonoid(s) with strong UGT1A1 induction activity. Among all the tested flavonoids, neobavaisoflavone (NBIF) displayed the most potent UGT1A1 induction activity, while its inductive effects were confirmed by both western blot and glucuronidation activity assays. A panel of nuclear receptor reporter assays demonstrated that NBIF activated PPARα and PPARγ in a dose-dependent manner. Meanwhile, we also found that NBIF could up-regulate the expression of PPARα and PPARγ in hepatic cells, suggesting that the induction of UGT1A1 by NBIF was mainly mediated by PPARs. In silico simulations showed that NBIF could stably bind on pocket II of PPARα and PPARγ. Collectively, our results demonstrated that NBIF is a natural inducer of UGT1A1, while this agent induced UGT1A1 mainly via activating and up-regulating PPARα and PPARγ. These findings suggested that NBIF can be used as a promising lead compound for the development of more efficacious UGT1A1 inducers to treat hyperbilirubinaemia and UGT1A1-associated drug toxicities., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Zhu, Guan, Chen, Peng, Finel, Zhao, Wang, Bi, Lei, Wang and Ge.)
- Published
- 2021
- Full Text
- View/download PDF
24. An ultra-sensitive and easy-to-use assay for sensing human UGT1A1 activities in biological systems.
- Author
-
Zhu YD, Pang HL, Zhou QH, Qin ZF, Jin Q, Finel M, Wang YN, Qin WW, Lu Y, Wang DD, and Ge GB
- Abstract
The human UDP-glucuronosyltransferase 1A1 (UGT1A1), one of the most essential conjugative enzymes, is responsible for the metabolism and detoxification of bilirubin and other endogenous substances, as well as many different xenobiotic compounds. Deciphering UGT1A1 relevance to human diseases and characterizing the effects of small molecules on the activities of UGT1A1 requires reliable tools for probing the function of this key enzyme in complex biological matrices. Herein, an easy-to-use assay for highly-selective and sensitive monitoring of UGT1A1 activities in various biological matrices, using liquid chromatography with fluorescence detection (LC-FD), has been developed and validated. The newly developed LC-FD based assay has been confirmed in terms of sensitivity, specificity, precision, quantitative linear range and stability. One of its main advantages is lowering the limits of detection and quantification by about 100-fold in comparison to the previous assay that used the same probe substrate, enabling reliable quantification of lower amounts of active enzyme than any other method. The precision test demonstrated that both intra- and inter-day variations for this assay were less than 5.5%. Furthermore, the newly developed assay has also been successfully used to screen and characterize the regulatory effects of small molecules on the expression level of UGT1A1 in living cells. Overall, an easy-to-use LC-FD based assay has been developed for ultra-sensitive UGT1A1 activities measurements in various biological systems, providing an inexpensive and practical approach for exploring the role of UGT1A1 in human diseases, interactions with xenobiotics, and characterization modulatory effects of small molecules on this conjugative enzyme., Competing Interests: The authors declare that there are no conflicts of interest., (© 2020 Xi'an Jiaotong University. Production and hosting by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
25. Human efflux transport of testosterone, epitestosterone and other androgen glucuronides.
- Author
-
Järvinen E, Kidron H, and Finel M
- Subjects
- ATP Binding Cassette Transporter, Subfamily B metabolism, ATP Binding Cassette Transporter, Subfamily G, Member 2 metabolism, Biological Transport, Humans, Models, Molecular, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins metabolism, Neoplasm Proteins metabolism, Epitestosterone metabolism, Glucuronides metabolism, Liver metabolism, Testosterone metabolism
- Abstract
Several drug-metabolizing enzymes are known to control androgen homeostasis in humans. UDP-glucuronosyltransferases convert androgens to glucuronide conjugates in the liver and intestine, which enables subsequent elimination of these conjugated androgens via urine. The most important androgen is testosterone, while others are the testosterone metabolites androsterone and etiocholanolone, and the testosterone precursor dehydroepiandrosterone. Epitestosterone is another endogenous androgen, which is included as a crucial marker in urine doping tests. Since glucuronide conjugates are hydrophilic, efflux transporters mediate their excretion from tissues. In this study, we employed the membrane vesicle assay to identify the efflux transporters for glucuronides of androsterone, dehydroepiandrosterone, epitestosterone, etiocholanolone and testosterone. The human hepatic and intestinal transporters MRP2 (ABCC2), MRP3 (ABCC3), MRP4 (ABCC4), BCRP (ABCG2) and MDR1 (ABCB1) were studied in vitro. Of these transporters, only MRP2 and MRP3 transported the androgen glucuronides investigated. In kinetic analyses, MRP3 transported glucuronides of androsterone, epitestosterone and etiocholanolone at low K
m values, between 0.4 and 4 μM, while the Km values for glucuronides of testosterone and dehydroepiandrosterone were 14 and 51 μM, respectively. MRP2 transported the glucuronides at lower affinity, as indicated by Km values over 100 μM. Interestingly, the MRP2-mediated transport of androsterone and epitestosterone glucuronides was best described by sigmoidal kinetics. The inability of BCRP to transport any of the androgen glucuronides investigated is drastically different from its highly active transport of several estrogen conjugates. Our results explain the transporter-mediated disposition of androgen glucuronides in humans, and shed light on differences between the human efflux transporters MRP2, MRP3, MRP4, BCRP and MDR1., Competing Interests: Declaration of Competing Interest None., (Copyright © 2019 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
26. In vitro glucuronidation of 7-hydroxycoumarin derivatives in intestine and liver microsomes of Beagle dogs.
- Author
-
Juvonen RO, Heikkinen AT, Kärkkäinen O, Jehangir R, Huuskonen J, Troberg J, Raunio H, Pentikäinen OT, and Finel M
- Subjects
- Animals, Dogs, Humans, Microsomes metabolism, Colon metabolism, Glucuronides metabolism, Glucuronosyltransferase metabolism, Intestine, Small metabolism, Liver metabolism, Umbelliferones metabolism
- Abstract
Beagle dog is a standard animal model for evaluating nonclinical pharmacokinetics of new drug candidates. Glucuronidation in intestine and liver is an important first-pass drug metabolic pathway, especially for phenolic compounds. This study evaluated the glucuronidation characteristics of several 7-hydroxycoumarin derivatives in beagle dog's intestine and liver in vitro. To this end, glucuronidation rates of 7-hydroxycoumarin (compound 1), 7-hydroxy-4-trifluoromethylcoumarin (2), 6-methoxy-7-hydroxycoumarin (3), 7-hydroxy-3-(4-tolyl)coumarin (4), 3-(4-fluorophenyl)coumarin (5), 7-hydroxy-3-(4-hydroxyphenyl)coumarin (6), 7-hydroxy-3-(4-methoxyphenyl)coumarin (7), and 7-hydroxy-3-(1H-1,2,4-tirazole)coumarin (8) were determined in dog's intestine and liver microsomes, as well as recombinant dog UGT1A enzymes. The glucuronidation rates of 1, 2 and 3 were 3-10 times higher in liver than in small intestine microsomes, whereas glucuronidation rates of 5, 6, 7 and 8 were similar in microsomes from both tissues. In the colon, glucuronidation of 1 and 2 was 3-5 times faster than in small intestine. dUGT1A11 glucuronidated efficiently all the substrates and was more efficient catalyst for 8 than any other dUGT1A. Other active enzymes were dUGT1A2 that glucuronidated efficiently 2, 3, 4, 5, 6 and 7, while dUGT1A10 glucuronidated efficiently 1, 2, 3, 4, 5 and 7. Kinetic analyses revealed that the compounds' K
m values varied between 1.1 (dUGT1A10 and 2) and 250 µM (dUGT1A7 and 4). The results further strengthen the concept that dog intestine has high capacity for glucuronidation, and that different dUGT1As mediate glucuronidation with distinct substrates selectivity in dog and human., (Copyright © 2019 Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
27. Efflux transport of nicotine, cotinine and trans-3'-hydroxycotinine glucuronides by human hepatic transporters.
- Author
-
Järvinen E, Sjöstedt N, Koenderink JB, Kidron H, and Finel M
- Subjects
- ATP Binding Cassette Transporter, Subfamily B metabolism, ATP Binding Cassette Transporter, Subfamily G, Member 2 metabolism, Biological Transport, Glucuronides metabolism, Humans, Liver metabolism, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins metabolism, Neoplasm Proteins metabolism, ATP-Binding Cassette Transporters metabolism, Cotinine analogs & derivatives, Cotinine metabolism, Hepatocytes metabolism, Nicotine metabolism
- Abstract
Nicotine is the addiction causing alkaloid in tobacco, and it is used in smoking cessation therapies. Although the metabolic pathways of nicotine are well known and mainly occur in the liver, the transport of nicotine and its metabolites is poorly characterized. The highly hydrophilic nature and urinary excretion of nicotine glucuronide metabolites indicate that hepatic basolateral efflux transporters mediate their excretion. We aimed here to find the transporters responsible for the hepatic excretion of nicotine, cotinine and trans-3'-hydroxycotinine (OH-cotinine) glucuronides. To this end, we tested their transport by multidrug resistance-associated proteins 1 (MRP1, ABCC1) and MRP3-6 (ABCC3-6), which are located on the basolateral membranes of hepatocytes, as well as MRP2 (ABCC2), breast cancer resistance protein (BCRP, ABCG2) and multidrug resistance protein 1 (MDR1, P-gp, ABCB1) that are expressed in the apical membranes of these cells. ATP-dependent transport of these glucuronides was evaluated in inside-out membrane vesicles expressing the transporter of interest. In addition, potential interactions of both the glucuronides and parent compounds with selected transporters were tested by inhibition assays. Considerable ATP-dependent transport was observed only for OH-cotinine glucuronide by MRP3. The kinetics of this transport activity was characterized, resulting in an estimated K
m value of 895 µmol/L. No significant transport was found for nicotine or cotinine glucuronides by any of the tested transporters at either 5 or 50 µmol/L substrate concentration. Furthermore, neither nicotine, cotinine nor OH-cotinine inhibited MRP2-4, BCRP or MDR1. In this study, we directly examined, for the first time, efflux transport of the three hydrophilic nicotine glucuronide metabolites by the major human hepatic efflux transporters. Despite multiple transporters studied here, our results indicate that an unknown transporter may be responsible for the hepatic excretion of nicotine and cotinine glucuronides., (© 2019 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society).)- Published
- 2019
- Full Text
- View/download PDF
28. Characterization of human UGT2A3 expression using a prepared specific antibody against UGT2A3.
- Author
-
Gotoh-Saito S, Abe T, Furukawa Y, Oda S, Yokoi T, Finel M, Hatakeyama M, Fukami T, and Nakajima M
- Subjects
- Antigen-Antibody Reactions, Glucuronosyltransferase genetics, Glucuronosyltransferase metabolism, Humans, Microsomes immunology, Microsomes metabolism, RNA, Messenger genetics, RNA, Messenger immunology, RNA, Messenger metabolism, Recombinant Proteins genetics, Recombinant Proteins immunology, Recombinant Proteins metabolism, Tumor Cells, Cultured, Antibodies immunology, Glucuronosyltransferase immunology
- Abstract
UDP-Glucuronosyltransferase (UGT) 2A3 belongs to a UGT superfamily of phase II drug-metabolizing enzymes that catalyzes the glucuronidation of many endobiotics and xenobiotics. Previous studies have demonstrated that UGT2A3 is expressed in the human liver, small intestine, and kidney at the mRNA level; however, its protein expression has not been determined. Evaluation of the protein expression of UGT2A3 would be useful to determine its role at the tissue level. In this study, we prepared a specific antibody against human UGT2A3 and evaluated the relative expression of UGT2A3 in the human liver, small intestine, and kidney. Western blot analysis indicated that this antibody is specific to UGT2A3 because it did not cross-react with other human UGT isoforms or rodent UGTs. UGT2A3 expression in the human small intestine was higher than that in the liver and kidney. Via treatment with endoglycosidase, it was clearly demonstrated that UGT2A3 was N-glycosylated. UGT2A3 protein levels were significantly correlated with UGT2A3 mRNA levels in a panel of 28 human liver samples (r = 0.64, p < 0.001). In conclusion, we successfully prepared a specific antibody against UGT2A3. This antibody would be useful to evaluate the physiological, pharmacological, and toxicological roles of UGT2A3 in human tissues., (Copyright © 2019 The Japanese Society for the Study of Xenobiotics. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
29. Inhibition of human carboxylesterases by magnolol: Kinetic analyses and mechanism.
- Author
-
Song YQ, Weng ZM, Dou TY, Finel M, Wang YQ, Ding LL, Jin Q, Wang DD, Fang SQ, Cao YF, Hou J, and Ge GB
- Subjects
- Binding Sites, Biocatalysis, Biphenyl Compounds chemistry, Carboxylic Ester Hydrolases antagonists & inhibitors, Catalytic Domain, Drugs, Chinese Herbal chemistry, Drugs, Chinese Herbal metabolism, Hep G2 Cells, Humans, Hydrolysis, Kinetics, Lignans chemistry, Molecular Docking Simulation, Biphenyl Compounds metabolism, Carboxylic Ester Hydrolases metabolism, Lignans metabolism
- Abstract
Magnolol, the most abundant bioactive constituent of the Chinese herb Magnolia officinalis, has been found with multiple biological activities, including anti-oxidative, anti-inflammatory and enzyme-regulatory activities. In this study, the inhibitory effects and inhibition mechanism of magnolol on human carboxylesterases (hCEs), the key enzymes responsible for the hydrolytic metabolism of a variety of endogenous esters as well as ester-bearing drugs, have been well-investigated. The results demonstrate that magnolol strongly inhibits hCE1-mediated hydrolysis of various substrates, whereas the inhibition of hCE2 by magnolol is substrate-dependent, ranging from strong to moderate. Inhibition of intracellular hCE1 and hCE2 by magnolol was also investigated in living HepG2 cells, and the results showed that magnolol could strongly inhibit intracellular hCE1, while the inhibition of intracellular hCE2 was weak. Inhibition kinetic analyses and docking simulations revealed that magnolol inhibited both hCE1 and hCE2 in a mixed manner, which could be partially attributed to its binding at two distinct ligand-binding sites in each carboxylesterase, including the catalytic cavity and the regulatory domain. In addition, the potential risk of the metabolic interactions of magnolol via hCE1 inhibition was predicted on the basis of a series of available pharmacokinetic data and the inhibition constants. All these findings are very helpful in deciphering the metabolic interactions between magnolol and hCEs, and also very useful for avoiding deleterious interactions via inhibition of hCEs., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
30. Case report by Toce and co-authors: Have all the reasons for poor morphine glucuronidation been addressed?
- Author
-
Finel M and Järvinen E
- Subjects
- Administration, Intravenous, Child, Glucuronosyltransferase, Humans, Male, Morphine, Sleep Apnea, Central
- Published
- 2019
- Full Text
- View/download PDF
31. Metabolism of Scoparone in Experimental Animals and Humans.
- Author
-
Juvonen RO, Novák F, Emmanouilidou E, Auriola S, Timonen J, Heikkinen AT, Küblbeck J, Finel M, and Raunio H
- Subjects
- Animals, Coumarins pharmacokinetics, Dogs, Drugs, Chinese Herbal pharmacokinetics, Female, Humans, Male, Mice, Mice, Inbred DBA, Microsomes, Liver metabolism, Oxidation-Reduction, Rabbits, Rats, Rats, Wistar, Swine, Coumarins metabolism, Drugs, Chinese Herbal metabolism
- Abstract
Scoparone, a major constituent of the Chinese herbal medicine Yin Chen Hao, expresses beneficial effects in experimental models of various diseases. The intrinsic doses and effects of scoparone are dependent on its metabolism, both in humans and animals. We evaluated in detail the metabolism of scoparone in human, mouse, rat, pig, dog, and rabbit liver microsomes in vitro and in humans in vivo . Oxidation of scoparone to isoscopoletin via 6-O-demethylation was the major metabolic pathway in liver microsomes from humans, mouse, rat, pig and dog, whereas 7-O-demethylation to scopoletin was the main reaction in rabbit. The scoparone oxidation rates in liver microsomes were 0.8 - 1.2 µmol/(min*g protein) in mouse, pig, and rabbit, 0.2 - 0.4 µmol/(min*g protein) in man and dog, and less than 0.1 µmol/(min*g) in rat. In liver microsomes of all species, isoscopoletin was oxidized to 3-[4-methoxy-ρ-(3, 6)-benzoquinone]-2-propenoate and esculetin, which was formed also in the oxidation of scopoletin. Human CYP2A13 exhibited the highest rate of isoscopoletin and scopoletin oxidation, followed by CYP1A1 and CYP1A2. Glucuronidation of isoscopoletin and scopoletin was catalyzed by the human UGT1A1, UGT1A6, UGT1A7, UGT1A8, UGT1A9, UGT1A10, and UGT2B17. Dog was most similar to man in scoparone metabolism. Isoscopoletin glucuronide and sulfate conjugates were the major scoparone in vivo metabolites in humans, and they were completely excreted within 24 h in urine. Scoparone and its metabolites did not activate key nuclear receptors regulating CYP and UGT enzymes. These results outline comprehensively the metabolic pathways of scoparone in man and key preclinical animal species., Competing Interests: The authors declare no conflicts of interest., (Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2019
- Full Text
- View/download PDF
32. Inhibition of UGT1A1 by natural and synthetic flavonoids.
- Author
-
Liu XY, Lv X, Wang P, Ai CZ, Zhou QH, Finel M, Fan B, Cao YF, Tang H, and Ge GB
- Subjects
- Catalytic Domain, Flavones chemistry, Flavones pharmacology, Flavonoids chemistry, Fluorescent Dyes metabolism, Glucuronosyltransferase chemistry, Glucuronosyltransferase metabolism, Humans, Inhibitory Concentration 50, Kaempferols chemistry, Kaempferols pharmacology, Microsomes, Liver drug effects, Microsomes, Liver metabolism, Molecular Docking Simulation, Substrate Specificity drug effects, Flavonoids pharmacology, Glucuronosyltransferase antagonists & inhibitors
- Abstract
Flavonoids are widely distributed phytochemicals in vegetables, fruits and medicinal plants. Recent studies demonstrate that some natural flavonoids are potent inhibitors of the human UDP-glucuronosyltransferase 1A1 (UGT1A1), a key enzyme in detoxification of endogenous harmful compounds such as bilirubin. In this study, the inhibitory effects of 56 natural and synthetic flavonoids on UGT1A1 were assayed, while the structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated. The results demonstrated that the C-3 and C-7 hydroxyl groups on the flavone skeleton would enhance UGT1A1 inhibition, while flavonoid glycosides displayed weaker inhibitory effects than their corresponding aglycones. Further investigation on inhibition kinetics of two strong flavonoid-type UGT1A1 inhibitors, acacetin and kaempferol, yielded interesting results. Both flavonoids were competitive inhibitors against UGT1A1-mediated NHPN-O-glucuronidation, but were mixed and competitive inhibitors toward UGT1A1-mediated NCHN-O-glucuronidation, respectively. Furthermore, docking simulations showed that the binding areas of NHPN, kaempferol and acacetin on UGT1A1 were highly overlapping, and convergence with the binding area of bilirubin within UGT1A1. In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition., (Copyright © 2018. Published by Elsevier B.V.)
- Published
- 2019
- Full Text
- View/download PDF
33. Recent progress and challenges in screening and characterization of UGT1A1 inhibitors.
- Author
-
Lv X, Xia Y, Finel M, Wu J, Ge G, and Yang L
- Abstract
Uridine-diphosphate glucuronosyltransferase 1A1 (UGT1A1) is an important conjugative enzyme in mammals that is responsible for the conjugation and detoxification of both endogenous and xenobiotic compounds. Strong inhibition of UGT1A1 may trigger adverse drug/herb-drug interactions, or result in metabolic disorders of endobiotic metabolism. Therefore, both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have recommended assaying the inhibitory potential of drugs under development on the human UGT1A1 prior to approval. This review focuses on the significance, progress and challenges in discovery and characterization of UGT1A1 inhibitors. Recent advances in the development of UGT1A1 probes and their application for screening UGT1A1 inhibitors are summarized and discussed in this review for the first time. Furthermore, a long list of UGT1A1 inhibitors, including information on their inhibition potency, inhibition mode, and affinity, has been prepared and analyzed. Challenges and future directions in this field are highlighted in the final section. The information and knowledge that are presented in this review provide guidance for rational use of drugs/herbs in order to avoid the occurrence of adverse effects via UGT1A1 inhibition, as well as presenting methods for rapid screening and characterization of UGT1A1 inhibitors and for facilitating investigations on UGT1A1-ligand interactions.
- Published
- 2019
- Full Text
- View/download PDF
34. Endocrine activities and adipogenic effects of bisphenol AF and its main metabolite.
- Author
-
Skledar DG, Carino A, Trontelj J, Troberg J, Distrutti E, Marchianò S, Tomašič T, Zega A, Finel M, Fiorucci S, and Mašič LP
- Subjects
- Animals, Humans, Mice, Signal Transduction, Benzhydryl Compounds metabolism, Glucuronides metabolism, Phenols metabolism
- Abstract
Bisphenol AF (BPAF) is a fluorinated analog of bisphenol A (BPA), and it is a more potent estrogen receptor (ER) agonist. BPAF is mainly metabolized to BPAF-glucuronide (BPAF-G), which has been reported to lack ER agonist activity and is believed to be biologically inactive. The main goal of the current study was to examine the influence of the metabolism of BPAF via glucuronidation on its ER activity and adipogenesis. Also, as metabolites can have different biological activities, the effects of BPAF-G on other nuclear receptors were evaluated. First, in-vitro BPAF glucuronidation was investigated using recombinant human enzymes. Specific reporter-gene assays were used to determine BPAF and BPAF-G effects on estrogen, androgen, glucocorticoid, and thyroid receptor pathways, and on PXR, FXR, and PPARγ pathways. Their effects on lipid accumulation and differentiation were determined in murine 3T3L1 preadipocytes using Nile Red, with mRNA expression analysis of the adipogenic markers adiponectin, Fabp4, Cebpα, and PPARγ. BPAF showed strong agonistic activity for hERα and moderate antagonistic activities for androgen and thyroid receptors, and for PXR. BPAF-G was antagonistic for PXR and PPARγ. BPAF (0.1 μM) and BPAF-G (1.0 μM) induced lipid accumulation and increased expression of key adipogenic markers in murine preadipocytes. BPAF-G is therefore not an inactive metabolite of BPAF. Further toxicological and epidemiological investigations of BPAF effects on human health are warranted, to provide better understanding of the metabolic end-elimination of BPAF., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
35. A Novel Pathogenic UGT1A1 Variant in a Sudanese Child with Type 1 Crigler-Najjar Syndrome.
- Author
-
Elfar W, Järvinen E, Ji W, Mosorin J, Sega AG, Iuga AC, Lobritto SJ, Konstantino M, Chan A, Finel M, and Lakhani SA
- Subjects
- Child, Preschool, Crigler-Najjar Syndrome surgery, Genetic Testing, Homozygote, Humans, Liver Transplantation, Male, Sequence Deletion, Sudan, Crigler-Najjar Syndrome genetics, Glucuronosyltransferase genetics
- Abstract
Uridine diphosphate glucuronosyltransferases (UGTs) are key enzymes responsible for the body's ability to process a variety of endogenous and exogenous compounds. Significant gains in understanding UGT function have come from the analysis of variants seen in patients. We cared for a Sudanese child who showed clinical features of type 1 Crigler-Najjar syndrome (CN-1), namely severe unconjugated hyperbilirubinemia leading to liver transplantation. CN-1 is an autosomal recessive disorder caused by damaging mutations in the gene for UGT1A1, the hepatic enzyme responsible for bilirubin conjugation in humans. Clinical genetic testing was unable to identify a known pathogenic UGT1A1 mutation in this child. Instead, a novel homozygous variant resulting in an in-frame deletion, p.Val275del, was noted. Sanger sequencing demonstrated that this variant segregated with the disease phenotype in this family. We further performed functional testing using recombinantly expressed UGT1A1 with and without the patient variant, demonstrating that p.Val275del results in a complete lack of glucuronidation activity, a hallmark of CN-1. Sequence analysis of this region shows a high degree of conservation across all known catalytically active human UGTs, further suggesting that it plays a key role in the enzymatic function of UGTs. Finally, we note that the patient's ethnicity likely played a role in his variant being previously undescribed and advocate for greater diversity and inclusion in genomic medicine., (Copyright © 2018 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2019
- Full Text
- View/download PDF
36. Data on biosynthesis of BPAF glucuronide, enzyme kinetics of BPAF glucuronidation, and molecular modeling.
- Author
-
Gramec Skledar D, Trontelj J, Troberg J, Tomašič T, Zega A, Finel M, and Peterlin Mašič L
- Abstract
Bisphenol AF (BPAF) is in the body mainly metabolized to the corresponding bisphenol AF glucuronide (BPAF-G). While BPAF-G is not commercially available, enzyme-assisted synthesis of BPAF-G using the human recombinant enzyme UGT2A1, purification of BPAF-G by solid phase extraction and semi-preparative HPLC and chemical characterization of BPAF-G by NMR and LC-MS/MS were performed and are described here. Furthermore, BPAF glucuronidation kinetics with the UGT enzymes that showed the highest glucuronidation activity in previous studies (i.e hepatic UGTs 1A3, 2B7, and 2B17, intestinal UGT 1A10 and UGT 2A1 that is present in airways) was performed and data is presented. Hepatic enzymes exhibited high affinities toward BPAF, while extrahepatic UGTs 2A1 and 1A10 showed the high v
max values (3.3 and 3.0 nmol/min/mg, respectively). To understand molecular interactions of BPA, BPAF and BPAF-G with ligand biding sites of several nuclear receptors, molecular modeling was performed and data on the binding modes of BPAF, BPA, and BPAF-G in the ligand-binding sites of nuclear receptors are presented. This article is related to "Endocrine activities and adipogenic effects of bisphenol AF and its main metabolite" (Skledar et al., 2019).- Published
- 2018
- Full Text
- View/download PDF
37. Efflux transport of estrogen glucuronides by human MRP2, MRP3, MRP4 and BCRP.
- Author
-
Järvinen E, Deng F, Kidron H, and Finel M
- Subjects
- Biological Transport, Humans, Multidrug Resistance-Associated Protein 2, ATP Binding Cassette Transporter, Subfamily G, Member 2 metabolism, Estrogens metabolism, Estrogens, Conjugated (USP) metabolism, Glucuronides metabolism, Multidrug Resistance-Associated Proteins metabolism, Neoplasm Proteins metabolism
- Abstract
Estrone, estradiol and estriol are endogenous human estrogens that are rapidly conjugated with glucuronic acid in both intestinal and hepatic epithelial cells. The resulting glucuronides, estrone-3-glucuronide (E
1 -G), estradiol-3- and 17-glucuronides (E2 -3G and E2 -17G), as well as estriol-3- and 16-glucuronides (E3 -3G and E3 -16G) are found in human plasma and urine. Unlike E2 -17G, the efflux transport of other estrogen glucuronides by human transporters has not yet been investigated comprehensively. We have studied the transport of E1 -G, E2 -3G, E3 -3G, E3 -16G and estrone-3-sulfate (E1 -S), another important estrogen conjugate, using the vesicular transport assay with recombinant human MRP2, MRP3, MRP4, MDR1 and BCRP that were expressed in insect cells. The transport screening assays revealed that whereas E1 -S was a good and specific substrate for BCRP, the less transporter-specific conjugates, E1 -G and E2 -3G, were still transported by BCRP at 10-fold higher rates than E1 -S. BCRP also transported E3 -16G at higher rates than the studied MRPs, while it transported E3 -3G at lower rates than MRP3. MRP2 exhibited lower or equal transport rates of E1 -G, E2 -3G, E3 -3G and E3 -16G in comparison to MRP3 and BCRP in the screening assays, mainly due to its high Km values, between 180 and 790 μM. MRP3 transported all the tested glucuronides at rather similar rates, at Km values below 20 μM, but lower Vmax values than other transporters. In the case of E3 -3G, MRP3 was the most active transporter in the screening assay. MRP4 transported only E3 -16G at considerable rates, while none of the tested estrogen conjugates was transported by MDR1 at higher rates than control vesicles. These new results, in combination with previously reported in vivo human data, stimulate our understanding on the substrate specificity and role of efflux transporters in disposition of estrogen glucuronides in humans., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
38. Molecular Docking-Based Design and Development of a Highly Selective Probe Substrate for UDP-glucuronosyltransferase 1A10.
- Author
-
Juvonen RO, Rauhamäki S, Kortet S, Niinivehmas S, Troberg J, Petsalo A, Huuskonen J, Raunio H, Finel M, and Pentikäinen OT
- Subjects
- Fluorescent Dyes metabolism, Glucuronides metabolism, Glucuronosyltransferase chemistry, Glucuronosyltransferase genetics, Humans, Microsomes, Molecular Probes chemistry, Molecular Probes metabolism, Mutagenesis, Site-Directed, Mutation, Substrate Specificity, Umbelliferones chemistry, Umbelliferones metabolism, Drug Design, Fluorescent Dyes chemistry, Glucuronosyltransferase metabolism, Molecular Docking Simulation, Molecular Imaging methods
- Abstract
Intestinal and hepatic glucuronidation by the UDP-glucuronosyltransferases (UGTs) greatly affect the bioavailability of phenolic compounds. UGT1A10 catalyzes glucuronidation reactions in the intestine, but not in the liver. Here, our aim was to develop selective, fluorescent substrates to easily elucidate UGT1A10 function. To this end, homology models were constructed and used to design new substrates, and subsequently, six novel C3-substituted (4-fluorophenyl, 4-hydroxyphenyl, 4-methoxyphenyl, 4-(dimethylamino)phenyl, 4-methylphenyl, or triazole) 7-hydroxycoumarin derivatives were synthesized from inexpensive starting materials. All tested compounds could be glucuronidated to nonfluorescent glucuronides by UGT1A10, four of them highly selectively by this enzyme. A new UGT1A10 mutant, 1A10-H210M, was prepared on the basis of the newly constructed model. Glucuronidation kinetics of the new compounds, in both wild-type and mutant UGT1A10 enzymes, revealed variable effects of the mutation. All six new C3-substituted 7-hydroxycoumarins were glucuronidated faster by human intestine than by liver microsomes, supporting the results obtained with recombinant UGTs. The most selective 4-(dimethylamino)phenyl and triazole C3-substituted 7-hydroxycoumarins could be very useful substrates in studying the function and expression of the human UGT1A10.
- Published
- 2018
- Full Text
- View/download PDF
39. Clopidogrel Carboxylic Acid Glucuronidation is Mediated Mainly by UGT2B7, UGT2B4, and UGT2B17: Implications for Pharmacogenetics and Drug-Drug Interactions .
- Author
-
Kahma H, Filppula AM, Neuvonen M, Tarkiainen EK, Tornio A, Holmberg MT, Itkonen MK, Finel M, Neuvonen PJ, Niemi M, and Backman JT
- Subjects
- Drug Interactions genetics, Glucuronosyltransferase genetics, Humans, Intestinal Mucosa metabolism, Kinetics, Liver metabolism, Microsomes, Liver metabolism, Minor Histocompatibility Antigens genetics, Pharmacogenetics methods, Ticlopidine metabolism, Glucuronides metabolism, Glucuronosyltransferase metabolism, Minor Histocompatibility Antigens metabolism, Ticlopidine analogs & derivatives
- Abstract
The antiplatelet drug clopidogrel is metabolized to an acyl- β -d-glucuronide, which causes time-dependent inactivation of CYP2C8. Our aim was to characterize the UDP-glucuronosyltransferase (UGT) enzymes that are responsible for the formation of clopidogrel acyl- β -d-glucuronide. Kinetic analyses and targeted inhibition experiments were performed using pooled human liver and intestine microsomes (HLMs and HIMs, respectively) and selected human recombinant UGTs based on preliminary screening. The effects of relevant UGT polymorphisms on the pharmacokinetics of clopidogrel were evaluated in 106 healthy volunteers. UGT2B7 and UGT2B17 exhibited the greatest level of clopidogrel carboxylic acid glucuronidation activities, with a CL
int,u of 2.42 and 2.82 µ l⋅min-1 ⋅mg-1 , respectively. Of other enzymes displaying activity (UGT1A3, UGT1A9, UGT1A10-H, and UGT2B4), UGT2B4 (CLint,u 0.51 µ l⋅min-1 ⋅mg-1 ) was estimated to contribute significantly to the hepatic clearance. Nonselective UGT2B inhibitors strongly inhibited clopidogrel acyl- β -d-glucuronide formation in HLMs and HIMs. The UGT2B17 inhibitor imatinib and the UGT2B7 and UGT1A9 inhibitor mefenamic acid inhibited clopidogrel carboxylic acid glucuronidation in HIMs and HLMs, respectively. Incubation of clopidogrel carboxylic acid in HLMs with UDPGA and NADPH resulted in strong inhibition of CYP2C8 activity. In healthy volunteers, the UGT2B17*2 deletion allele was associated with a 10% decrease per copy in the plasma clopidogrel acyl- β -d-glucuronide to clopidogrel carboxylic acid area under the plasma concentration-time curve from 0 to 4 hours (AUC0-4 ) ratio ( P < 0.05). To conclude, clopidogrel carboxylic acid is metabolized mainly by UGT2B7 and UGT2B4 in the liver and by UGT2B17 in the small intestinal wall. The formation of clopidogrel acyl- β -d-glucuronide is impaired in carriers of the UGT2B17 deletion. These findings may have implications regarding the intracellular mechanisms leading to CYP2C8 inactivation by clopidogrel., (Copyright © 2018 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2018
- Full Text
- View/download PDF
40. Identification and characterization of human UDP-glucuronosyltransferases responsible for xanthotoxol glucuronidation.
- Author
-
He G, Troberg J, Lv X, Xia YL, Zhu LL, Ning J, Ge GB, Finel M, and Yang L
- Subjects
- Humans, Kinetics, Microsomes, Liver metabolism, Furocoumarins metabolism, Glucuronosyltransferase metabolism
- Abstract
1. Xanthotoxol is a furanocoumarin that possesses many pharmacological activities and in this study its in vitro glucuronidation was studied. 2. Xanthotoxol can be rapidly metabolized to a mono-glucuronide in both human intestine microsomes (HIM) and human liver microsomes (HLM); the structure of the metabolite was confirmed by NMR spectroscopy. 3. Reaction phenotyping with 12 commercial recombinant human UGTs, as well as with the Helsinki laboratory UGT1A10 that carry a C-terminal His-tag (UGT1A10-H), revealed that UGT1A10-H catalyzes xanthotoxol glucuronidation at the highest rate, followed by UGT1A8. The other enzymes, namely UGT1A3, UGT1A1, UGT1A6, UGT1A10 (commercial), and UGT2B7 displayed moderate-to-low reaction rates. 4. In kinetic analyses, HIM exhibited much higher affinity for xanthotoxol, along with high V
max and mild substrate inhibition, whereas the kinetics in HLM was biphasic. UGT1A1 (high Km value), UGT1A10-H (low Km value), and UGT1A8 exhibited mild substrate inhibition. 5. Considering the above findings and the current knowledge on UGTs expression in HIM, it is likely that UGT1A10 is mainly responsible for xanthotoxol glucuronidation in the human small intestine, with some contribution from UGT1A1. In the liver, this reaction is mainly catalyzed by UGT1A1 and UGT2B7. 6. Glucuronidation appears to be the major metabolic pathway of xanthotoxol in human.- Published
- 2018
- Full Text
- View/download PDF
41. Selectivity in the Efflux of Glucuronides by Human Transporters: MRP4 Is Highly Active toward 4-Methylumbelliferone and 1-Naphthol Glucuronides, while MRP3 Exhibits Stereoselective Propranolol Glucuronide Transport.
- Author
-
Järvinen E, Troberg J, Kidron H, and Finel M
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2 metabolism, Animals, Biological Assay, Biological Transport, Active, Estradiol analogs & derivatives, Estradiol pharmacokinetics, Glucuronates metabolism, Humans, Hymecromone analogs & derivatives, Multidrug Resistance-Associated Protein 2, Neoplasm Proteins metabolism, Propranolol chemical synthesis, Propranolol pharmacokinetics, Pyrenes metabolism, Recombinant Proteins metabolism, Sf9 Cells, Spodoptera, Stereoisomerism, Glucuronates pharmacokinetics, Glucuronides pharmacokinetics, Hymecromone pharmacokinetics, Multidrug Resistance-Associated Proteins metabolism, Propranolol analogs & derivatives
- Abstract
Xenobiotic and endobiotic glucuronides, which are generated in hepatic and intestinal epithelial cells, are excreted via efflux transporters. Multidrug resistance proteins 2-4 (MRP2-MRP4) and the breast cancer resistance protein (BCRP) are efflux transporters that are expressed in these polarized cells, on either the basolateral or apical membranes. Their localization, along with expression levels, affects the glucuronide excretion pathways. We have studied the transport of three planar cyclic glucuronides and glucuronides of the two propranolol enantiomers, by the vesicular transport assay, using vesicles from baculovirus-infected insect cells expressing human MRP2, MRP3, MRP4, or BCRP. The transport of estradiol-17β-glucuronide by recombinant MRP2-4 and BCRP, as demonstrated by kinetic values, were within the ranges previously reported. Our results revealed high transport rates and apparent affinity of MRP4 toward the glucuronides of 4-methylumbelliferone, 1-naphthol, and 1-hydroxypyrene (K
m values of 168, 13, and 3 μM, respectively) in comparison to MRP3 (Km values of 278, 98, and 8 μM, respectively). MRP3 exhibited lower rates, but stereoselective transport of propranolol glucuronides, with higher affinity toward the R-enantiomer than the S-enantiomer (Km values 154 vs 434 μM). The glucuronide of propranolol R-enantiomer was not significantly transported by either MRP2, MRP4, or BCRP. Of the tested small glucuronides in this study, BCRP transported only 1-hydroxypyrene glucuronide, at very high rates and high apparent affinity (Vmax and Km values of 4400 pmol/mg/min and 11 μM). The transport activity of MRP2 with all of the studied small glucuronides was relatively very low, even though it transported the reference compound, estradiol-17β-glucuronide, at a high rate (Vmax = 3500 pmol/mg/min). Our results provide new information, at the molecular level, of efflux transport of the tested glucuronides, which could explain their disposition in vivo, as well as provide new tools for in vitro studies of MRP3, MRP4, and BCRP.- Published
- 2017
- Full Text
- View/download PDF
42. UGT1A10 Is a High Activity and Important Extrahepatic Enzyme: Why Has Its Role in Intestinal Glucuronidation Been Frequently Underestimated?
- Author
-
Troberg J, Järvinen E, Ge GB, Yang L, and Finel M
- Subjects
- Animals, Blotting, Western, Camptothecin analogs & derivatives, Camptothecin metabolism, Chromatography, High Pressure Liquid, Coumarins metabolism, Diclofenac metabolism, Estradiol metabolism, Estrone metabolism, Flavonoids metabolism, Humans, Hymecromone metabolism, Irinotecan, Kinetics, Microsomes, Liver metabolism, Glucuronosyltransferase metabolism, Intestinal Mucosa metabolism
- Abstract
The aim of this work was to highlight a considerable and broad problem in UGT1A10 activity assessment that has led to underestimation of its role in intestinal glucuronidation of drugs and other xenobiotics. The reason appears to be poor activity of the commercial UGT1A10 that is used by many laboratories, and here we have tested it by comparison with our recombinant His-tagged UGT1A10 (designated as UGT1A10-H), both expressed in insect cells. The glucuronidation rates of morphine, estradiol, estrone, SN-38, diclofenac, 4-methylumbelliferone, 7-amino-4-methylcoumarin, N-(3-carboxypropyl)-4-hydroxy-1,8-naphthalimide, and bavachinin were assayed. The results revealed that the activity of commercial UGT1A10 was low, very low, and in the cases of morphine, estrone, 7-methyl-4-aminocoumarin, and bavachinin it was below the detection limit. On the other hand, under the same conditions, UGT1A10-H exhibited high glucuronidation rates toward all these compounds. Moreover, using estradiol, morphine, and estrone, in the presence and absence of suitable inhibitors, nilotinib or atractylenolide I, it was demonstrated that UGT1A10-H, but not the commercial UGT1A10, provides a good tool to study the role of native UGT1A10 in the human intestine. The results also suggest that much of the data in the literature on UGT1A10 activity may have to be re-evaluated.
- Published
- 2017
- Full Text
- View/download PDF
43. Bisphenol-A glucuronidation in human liver and breast: identification of UDP-glucuronosyltransferases (UGTs) and influence of genetic polymorphisms.
- Author
-
Street CM, Zhu Z, Finel M, and Court MH
- Subjects
- Breast metabolism, Glucuronosyltransferase metabolism, Humans, Liver metabolism, Polymorphism, Genetic, Benzhydryl Compounds metabolism, Glucuronosyltransferase genetics, Hazardous Substances metabolism, Phenols metabolism
- Abstract
1. Bisphenol-A is a ubiquitous environmental contaminant that is primarily metabolized by glucuronidation and associated with various human diseases including breast cancer. Here we identified UDP-glucuronosyltransferases (UGTs) and genetic polymorphisms responsible for interindividual variability in bisphenol-A glucuronidation in human liver and breast. 2. Hepatic UGTs showing the highest bisphenol-A glucuronidation activity included UGT2B15 and UGT1A9. Relative activity factor normalization indicated that UGT2B15 contributes >80% of activity at bisphenol-A concentrations under 5 μM, while UGT1A9 contributes up to 50% of activity at higher concentrations. 3. Bisphenol-A glucuronidation by liver microsomes (46 donors) ranged from 0.25 to 4.3 nmoles/min/mg protein. Two-fold higher glucuronidation (p = 0.018) was observed in UGT1A9 *22/*22 livers compared with *1/*1 and *1/*22 livers. However, no associations were observed for UGT2B15*2 or UGT1A1*28 genotypes. 4. Bisphenol-A glucuronidation by breast microsomes (15 donors) ranged from <0.2 to 56 fmoles/min/mg protein. Breast mRNA expression of UGTs capable of glucuronidating bisphenol-A was highest for UGT1A1, followed by UGT2B4, UGT1A9, UGT1A10, UGT2B7 and UGT2B15. Bisphenol-A glucuronidation was over 10-fold lower in breast tissues with the UGT1A1*28 allele compared with tissues without this allele (p = 0.006). 5. UGT2B15 and UGT1A9 contribute to glucuronidation variability in liver, while UGT1A1 is important in breast.
- Published
- 2017
- Full Text
- View/download PDF
44. Influence of metabolism on endocrine activities of bisphenol S.
- Author
-
Skledar DG, Schmidt J, Fic A, Klopčič I, Trontelj J, Dolenc MS, Finel M, and Mašič LP
- Subjects
- Animals, Biotransformation, Cell Line, Cytochrome P-450 CYP2C9 genetics, Cytochrome P-450 CYP2C9 metabolism, Cytochrome P-450 CYP3A genetics, Cytochrome P-450 CYP3A metabolism, Endocrine Disruptors toxicity, Humans, Hydroxylation, Inactivation, Metabolic, Microsomes, Liver drug effects, Microsomes, Liver enzymology, Microsomes, Liver metabolism, Phenols toxicity, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae metabolism, Sulfones toxicity, Endocrine Disruptors metabolism, Glucuronides metabolism, Phenols metabolism, Receptors, Estrogen metabolism, Receptors, Thyroid Hormone antagonists & inhibitors, Sulfones metabolism
- Abstract
Bisphenol S (BPS; bis[4-hydroxyphenyl]sulfone) is commonly used as a replacement for bisphenol A in numerous consumer products. The main goal of this study was to examine the influence of different metabolic reactions that BPS undergoes on the endocrine activity. We demonstrate that hydroxylation of the aromatic ring of BPS, catalyzed mainly by the cytochrome P450 enzymes CYP3A4 and CYP2C9, is its major in-vitro phase I biotransformation. Nevertheless, coupled oxidative-conjugative reactions analyses revealed that glucuronidation and formation of BPS glucuronide is the predominant BPS metabolic pathway. BPS reactive metabolites that can be tracked as glutathione conjugates were not detected in the present study. Two in-vitro systems were used to evaluate the endocrine activity of BPS and its two main metabolites, BPS glucuronide and hydroxylated BPS 4-(4-hydroxy-benzenesulfonyl)-benzene-1,2-diol (BPSM1). In addition, we have tested two structural analogs of BPS, bis[4-(2-hydroxyetoxy)phenyl]sulfone (BHEPS) and 4,4-sulfonylbis(2-methylphenol) (dBPS). The test systems were yeast cells, for evaluating estrogenic and androgenic activities, and the GH3.TRE-Luc reporter cell line for measuring thyroid hormone activity. BPS and BPSM1 were weak agonists of the estrogen receptor, EC50 values of 8.4 × 10(-5) M and 6.7 × 10(-4) M, respectively. Additionally, BPSM1 exhibited weak antagonistic activity toward the thyroid hormone receptor, with an IC50 of 4.3 × 10(-5) M. In contrast to BPSM1, BPS glucuronide was inactive in these assays, inhibiting neither the estrogen nor the thyroid hormone receptors. Hence, glucuronidation appears to be the most important pathway for both BPS metabolism and detoxification., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. Biosynthesis of Drug Glucuronide Metabolites in the Budding Yeast Saccharomyces cerevisiae.
- Author
-
Ikushiro S, Nishikawa M, Masuyama Y, Shouji T, Fujii M, Hamada M, Nakajima N, Finel M, Yasuda K, Kamakura M, and Sakaki T
- Subjects
- Animals, Carboxylic Acids metabolism, Glucuronosyltransferase metabolism, Humans, Oxidoreductases metabolism, Umbelliferones metabolism, Glucuronides metabolism, Saccharomyces cerevisiae metabolism, Saccharomycetales metabolism
- Abstract
Glucuronidation is one of the most common pathways in mammals for detoxification and elimination of hydrophobic xenobiotic compounds, including many drugs. Metabolites, however, can form active or toxic compounds, such as acyl glucuronides, and their safety assessment is often needed. The absence of efficient means for in vitro synthesis of correct glucuronide metabolites frequently limits such toxicological analyses. To overcome this hurdle we have developed a new approach, the essence of which is a coexpression system containing a human, or another mammalian UDP-glucuronosyltransferases (UGTs), as well as UDP-glucose-6-dehydrogenase (UGDH), within the budding yeast, Saccharomyces cerevisiae. The system was first tested using resting yeast cells coexpressing UGDH and human UGT1A6, 7-hydroxycoumarin as the substrate, in a reaction medium containing 8% glucose, serving as a source of UDP-glucuronic acid. Glucuronides were readily formed and recovered from the medium. Subsequently, by selecting suitable mammalian UGT enzyme for the coexpression system we could obtain the desired glucuronides of various compounds, including molecules with multiple conjugation sites and acyl glucuronides of several carboxylic acid containing drugs, namely, mefenamic acid, flufenamic acid, and zomepirac. In conclusion, a new and flexible yeast system with mammalian UGTs has been developed that exhibits a capacity for efficient production of various glucuronides, including acyl glucuronides.
- Published
- 2016
- Full Text
- View/download PDF
46. Glucuronidation of estrone and 16α-hydroxyestrone by human UGT enzymes: The key roles of UGT1A10 and UGT2B7.
- Author
-
Kallionpää RA, Järvinen E, and Finel M
- Subjects
- Intestines enzymology, Kinetics, Microsomes, Liver enzymology, Substrate Specificity, Estrone metabolism, Glucuronides metabolism, Glucuronosyltransferase metabolism, Hydroxyestrones metabolism
- Abstract
The glucuronidation of estrone and 16α-hydroxyestrone by recombinant human UDP-glucuronosyltransferase enzymes (UGTs) of subfamilies 1A, 2A and 2B was studied. Microsomes from human liver and small intestine were also tested for the glucuronidation of these two estrogens. The results revealed that UGT1A10 is by far the most active enzyme in estrone glucuronidation. UGT1A10 also exhibited high rate of 16α-hydroxyestrone conjugation at the 3-OH, whereas UGT2B7 catalyzed its glucuronidation at high rates at the 16-OH. Human liver microsomes exhibited high rates of 16α-hydroxyestrone-16-glucuronide formation, but very low formation rates of either 16α-hydroxyestrone-3-glucuronide or estrone glucuronide. On the other hand, human intestine microsomes catalyzed the formation of all these 3 different glucuronides at high rates. Kinetic analyses revealed very low Km value for 16α-hydroxyestrone glucuronidation by UGT2B7, below 4 μM, suggesting higher affinity than commonly found among UGTs and their substrates. In further studies with UGT1A10, mutant F93G exhibited increased glucuronidation rates of 16α-hydroxyestrone, but not estrone, whereas mutations in F90 did not reveal any activity with either estrogen. Taken together, the results of this study significantly expand our understanding on the metabolism of estrogens and their interactions with the human UGTs., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
47. The Polymorphic Variant P24T of UDP-Glucuronosyltransferase 1A4 and Its Unusual Consequences.
- Author
-
Troberg J and Finel M
- Subjects
- Animals, HEK293 Cells, Humans, Microsomes, Liver drug effects, Microsomes, Liver enzymology, Trifluoperazine metabolism, Trifluoperazine pharmacology, Genetic Variation genetics, Glucuronosyltransferase genetics, Polymorphism, Genetic genetics
- Abstract
The P24T polymorphic variant of the human UDP-glucuronosyltransferase 1A4 (UGT1A4*2, 70C>A) occurs within the signal peptide, five amino acids upstream of the cleavage site and the start of the mature protein. Bioinformatic analysis of the variant suggested that the signal peptide of part of the translated protein is cleaved two residues upstream of the regular site, whereas the rest is cleaved as usual. To test this, recombinant UGT1A4-P24T, with a C-terminal His-tag, was expressed in sf9 insect cells and affinity-purified for N-terminal protein sequencing. The results were in agreement with the in silico prediction. About half of the mutant protein was cleaved at the regular site, between S28 and G29, whereas the other half was cleaved two amino acids upstream, between A26 and E27. The glucuronidation of two substrates, dexmedetomidine and trifluoperazine, was assayed using membrane-enriched UGT1A4-P24T and wild-type UGT1A4. The variant exhibited much lower glucuronidation rates, but kinetic analyses revealed large differences between them only in the Vmax values. The Km values for both substrates were not affected by the mutation and its consequences. This might suggest that the unusual signal peptide cleavage in UGT1A4-P24T somehow disturbs protein folding. Moreover, it raises the possibility that the effect of UGT1A4-P24T on the glucuronidation rate in mammalian expression systems would be mild since they contain more effective post-translation protein control systems in the endoplasmic reticulum. In summary, our results reveal the effect of a polymorphic mutation on the signal sequence cleavage and thereby also the mature UGT., (Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
48. An optimized ratiometric fluorescent probe for sensing human UDP-glucuronosyltransferase 1A1 and its biological applications.
- Author
-
Lv X, Ge GB, Feng L, Troberg J, Hu LH, Hou J, Cheng HL, Wang P, Liu ZM, Finel M, Cui JN, and Yang L
- Subjects
- Biosensing Techniques methods, Drug Evaluation, Preclinical methods, Fluorescent Dyes metabolism, Glucuronosyltransferase antagonists & inhibitors, Glucuronosyltransferase metabolism, Hep G2 Cells, Humans, Microscopy, Fluorescence methods, Naphthalimides metabolism, Optical Imaging methods, Enzyme Assays methods, Fluorescent Dyes chemistry, Glucuronosyltransferase analysis, Microsomes, Liver enzymology, Naphthalimides chemistry
- Abstract
This study aimed to develop a practical ratiometric fluorescent probe for highly selective and sensitive detection of human UDP-glucuronosyltransferase 1A1 (UGT1A1), one of the most important phase II enzymes. 4-Hydroxy-1,8-naphthalimide (HN) was selected as the fluorophore for this study because it possesses intramolecular charge transfer (ICT) feature and displays outstanding optical properties. A series of N-substituted derivatives with various hydrophobic, acidic and basic groups were designed and synthesized to evaluate the selectivity of HN derivatives toward UGT1A1. Our results demonstrated that the introduction of an acidic group to HN could significantly improve the selectivity of UGT1A1. Among the synthesized fluorescent probes, NCHN (N-3-carboxy propyl-4-hydroxy-1,8-naphthalimide) displayed the best combination of selectivity, sensitivity and ratiometric fluorescence response following UGT1A1-catalyzed glucuronidation. UGT1A1-catalyzed NCHN-4-O-glucuronidation generated a single fluorescent product with a high quantum yield (Φ=0.688) and brought remarkable changes in both color and fluorescence in comparison with the parental substrate. The newly developed probe has been successfully applied for sensitive measurements of UGT1A1 activities in human liver preparations, as well as for rapid screening of UGT1A1 modulators, using variable enzyme sources. Furthermore, its potential applications for live imaging of endogenous UGT1A1in cells have also been demonstrated., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
49. Exploring the structure-activity relationships of ABCC2 modulators using a screening approach.
- Author
-
Wissel G, Kudryavtsev P, Ghemtio L, Tammela P, Wipf P, Yliperttula M, Finel M, Urtti A, Kidron H, and Xhaard H
- Subjects
- Animals, Biological Transport drug effects, Estradiol analogs & derivatives, Estradiol metabolism, Fluoresceins metabolism, Gene Expression, High-Throughput Screening Assays, Molecular Probes metabolism, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins genetics, Multidrug Resistance-Associated Proteins metabolism, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sf9 Cells, Small Molecule Libraries chemistry, Spodoptera, Structure-Activity Relationship, Transport Vesicles metabolism, Multidrug Resistance-Associated Proteins agonists, Multidrug Resistance-Associated Proteins antagonists & inhibitors, Small Molecule Libraries pharmacology, Transport Vesicles drug effects
- Abstract
ABCC2 is a transporter with key influence on liver and kidney pharmacokinetics. In order to explore the structure-activity relationships of compounds that modulate ABCC2, and by doing so gain insights into drug-drug interactions, we screened a library of 432 compounds for modulators of radiolabeled β-estradiol 17-(β-d-glucuronide) (EG) and fluorescent 5(6)-carboxy-2',7'-dichlorofluorescein transport (CDCF) in membrane vesicles. Following the primary screen at 80μM, dose-response curves were used to investigate in detail 86 compounds, identifying 16 low μM inhibitors and providing data about the structure-activity relationships in four series containing 19, 24, 10, and eight analogues. Measurements with the CDCF probe were consistently more robust than for the EG probe. Only one compound was clearly probe-selective with a 50-fold difference in the IC50s obtained by the two assays. We built 24 classification models using the SVM and fused-XY Kohonen methods, revealing molecular descriptors related to number of rings, solubility and lipophilicity as important to distinguish inhibitors from inactive compounds. This study is to the best of our knowledge the first to provide details about structure-activity relationships in ABCC2 modulation., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
50. A UGT2B10 splicing polymorphism common in african populations may greatly increase drug exposure.
- Author
-
Fowler S, Kletzl H, Finel M, Manevski N, Schmid P, Tuerck D, Norcross RD, Hoener MC, Spleiss O, and Iglesias VA
- Subjects
- Cells, Cultured, Chromatography, High Pressure Liquid, Databases, Nucleic Acid, Glucuronides metabolism, Humans, Microsomes, Liver drug effects, Microsomes, Liver metabolism, Oxazoles administration & dosage, Substrate Specificity, Tandem Mass Spectrometry, Black People genetics, Gene Silencing, Glucuronosyltransferase genetics, Oxazoles pharmacokinetics, Polymorphism, Single Nucleotide
- Abstract
RO5263397 [(S)-4-(3-fluoro-2-methyl-phenyl)-4,5-dihydro-oxazol-2-ylamine], a new compound that showed promising results in animal models of schizophrenia, is mainly metabolized in humans by N-glucuronidation. Enzyme studies, using the (then) available commercial uridine 5'-diphosphate-glucuronosyltransferases (UGTs), suggested that UGT1A4 is responsible for its conjugation. In the first clinical trial, in which RO5263397 was administered orally to healthy human volunteers, a 136-fold above-average systemic exposure to the parent compound was found in one of the participants. Further administration in this trial identified two more such poor metabolizers, all three of African origin. Additional in vitro studies with recombinant UGTs showed that the contribution of UGT2B10 to RO5263397 glucuronidation is much higher than UGT1A4 at clinically relevant concentrations. DNA sequencing in all of these poor metabolizers identified a previously uncharacterized splice site mutation that prevents assembly of full-length UGT2B10 mRNA and thus functional UGT2B10 protein expression. Further DNA database analyses revealed the UGT2B10 splice site mutation to be highly frequent in individuals of African origin (45%), moderately frequent in Asians (8%) and almost unrepresented in Caucasians (<1%). A prospective study using hepatocytes from 20 individual African donors demonstrated a >100-fold lower intrinsic clearance of RO5263397 in cells homozygous for the splice site variant allele. Our results highlight the need to include UGT2B10 when screening the human UGTs for the enzymes involved in the glucuronidation of a new compound, particularly when there is a possibility of N-glucuronidation. Moreover, this study demonstrates the importance of considering different ethnicities during drug development., (Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.