749
Views
16
CrossRef citations to date
0
Altmetric
Review Article

Substrate selectivity of human intestinal UDP-glucuronosyltransferases (UGTs): in silico and in vitro insights

, , &
Pages 231-252 | Received 27 Sep 2012, Accepted 14 Jan 2013, Published online: 06 Mar 2013

References

  • Ako R, Dong D, Wu B. (2012). 3D-QSAR studies on UDP-glucuronosyltransferase 2B7 substrates using the pharmacophore and VolSurf approaches. Xenobiotica 42:1–10
  • Albert C, Vallée M, Beaudry G, et al. (1999). The monkey and human uridine diphosphate-glucuronosyltransferase UGT1A9, expressed in steroid target tissues, are estrogen-conjugating enzymes. Endocrinology 140:3292–302
  • Ammon S, von Richter O, Hofmann U, et al. (2000). In vitro interaction of codeine and diclofenac. Drug Metab Dispos 28:1149–52
  • Ando Y, Saka H, Ando M, et al. (2000). Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: A pharmacogenetic analysis. Cancer Res 60:6921–6
  • Ando Y, Ueoka H, Sugiyama T, et al. (2002). Polymorphisms of UDP-glucuronosyltransferase and pharmacokinetics of irinotecan. Ther Drug Monit 24:111–16
  • Banhegyi G, Garzo T, Fulceri R, et al. (1993). Latency is the major determinant of UDP-glucuronosyltransferase activity in isolated hepatocytes. FEBS Lett 328:149–52
  • Barbier O, Girard H, Inoue Y, et al. (2005). Hepatic expression of the UGT1A9 gene is governed by hepatocyte nuclear factor 4α. Mol Pharmacol 67:241–9
  • Barre L, Fournel-Gigleux S, Finel M, et al. (2007). Substrate specificity of the human UDP-glucuronosyltransferase UGT2B4 and UGT2B7. FEBS J 274:1256–64
  • Basu NK, Kole L, Owens IS. (2003). Evidence for phosphorylation requirement for human bilirubin UDP-glucuronosyltransferase (UGT1A1) activity. Biochem Biophys Res Commun 303:98–104
  • Beaulieu M, Lévesque E, Tchernof A, et al. (1997). Chromosomal localization, structure, and regulation of the UGT2B17 gene, encoding a C19 steroid metabolizing enzyme. DNA Cell Biol 16:1143–54
  • Beaumont K. (2003). The importance of gut wall metabolism in determining drug bioavailability. In: Waterbeemd HVD, Lennernas H, Artursson P, eds. Drug bioavailability: Estimation of solubility, permeability and absorption. New York: Wiley-VCH Verlag, 311--28
  • Bélanger A, Hum DW, Beaulieu M, et al. (1998). Characterization and regulation of UDP-glucuronosyltransferases in steroid target tissues. J Steroid Biochem 65:301–10
  • Bélanger A, Pelletier G, Labrie F, et al. (2003). Inactivation of androgens by UDP-glucuronosyltransferase enzymes in humans. Trends Endocrinol Metab 14:473–9
  • Bellemare J, Rouleau M, Harvey M, Guillemette C. (2010). Modulation of the human glucuronosyltransferase UGT1A pathway by splice isoform polypeptides is mediated through protein--protein interactions. J Biol Chem 285:3600–7
  • Benet LZ. (1993). Pharmacokinetics of sustained-release etodolac. Rheumatol Int 13:S3–5
  • Bernard O, Guillemette C. (2004). The main role of UGT1A9 in the hepatic metabolism of mycophenolic acid and the effects of naturally occurring variants. Drug Metab Dispos 32:775–8
  • Bernard O, Tojcic J, Journault K, et al. (2006). Influence of nonsynonymous polymorphisms of UGT1A8 and UGT2B7 metabolizing enzymes on the formation of phenolic and acyl glucuronides of mycophenolic acid. Drug Metab Dispos 34:1539–45
  • Betonico GN, Abbud-Filho M, Goloni-Bertollo EM, et al. (2008). Influence of UDP-glucuronosyltransferase polymorphisms on mycophenolate mofetil-induced side effects in kidney transplant patients. Transplant P 40:708–10
  • Bhasker CR, McKinnon W, Stone A, et al. (2000). Genetic polymorphism of UDP-glucuronosyltransferase 2B7 (UGT2B7) at amino acid 268: Ethnic diversity of alleles and potential clinical significance. Pharmacogenetics 10:679–85
  • Bichlmaier I, Siiskonen A, Finel M, Yli-Kauhaluoma J. (2006). Stereochemical sensitivity of the human UDP-glucuronosyltransferases 2B7 and 2B17. J Med Chem 49:1818–27
  • Boersma MG, van der Woude H, Bogaards J, et al. (2002). Regioselectivity of phase II metabolism of luteolin and quercetin by UDP-glucuronosyl transferases. Chem Res Toxicol 15:662–70
  • Bosma PJ. (2003). Inherited disorders of bilirubin metabolism. J Hepatol 38:107–17
  • Bossuyt X, Blanckaert N. (1995). Mechanism of stimulation of microsomal UDP-glucuronosyltransferase by UDP-N-acetylglucosamine. Biochem J 305:321–8
  • Bossuyt X, Blanckaert N. (1997). Carrier-mediated transport of uridine diphosphoglucuronic acid across the endoplasmic reticulum membrane is a prerequisite for UDP-glucuronosyltransferase activity in rat liver. Biochem J 323:645–8
  • Bourne Y, Henrissat B. (2001). Glycoside hydrolases and glycosyltransferases: Families and functional modules. Curr Opin Struct Biol 11:593–600
  • Bowles J, Knight D, Smith C, et al. (2006). Retinoid signaling determines germ cell fate in mice. Science 312:596–600
  • Brand W, Boersma MG, Bik H, et al. (2010). Phase II metabolism of hesperetin by individual UDP-glucuronosyltransferases and sulfotransferases and rat and human tissue samples. Drug Metab Dispos 38:617–25
  • Brazier-Hicks M, Edwards LA, Edwards R. (2007). Selection of plants for roles in phytoremediation: The importance of glucosylation. Plant Biotechnol J 5:627–35
  • Breton C, Imberty A. (1999). Structure/function studies of glycosyltransferases. Curr Opin Struct Biol 9:563–71
  • Breton C, Šnajdrová L, Jeanneau C, et al. (2006). Structures and mechanisms of glycosyltransferases. Glycobiology 16:29R–37R
  • Burchell B. (2003). Genetic variation of human UDP-glucuronosyltransferase: Implications in disease and drug glucuronidation. Am J Pharmacogenomics 3:37–52
  • Burchell B, Lockley DJ, Staines A, et al. (2005). Substrate specificity of human hepatic UDP-glucuronosyltransferases. Method Enzymol 400:46–57
  • Campbell JA, Davies GJ, Bulone V, Henrissat B. (1997). A classification of nucleotide-diphospho-sugar glycosyltransferases based on amino acid sequence similarities. Biochem J 326:929–39
  • Carlini LE, Meropol NJ, Bever J, et al. (2005). UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Clin Cancer Res 3:1226–36
  • Cheng Z, Radominska-Pandya A, Tephly TR. (1998). Cloning and expression of human UDP-glucuronosyltransferase (UGT) 1A8. Arch Biochem Biophys 356:301–5
  • Cheng Z, Radominska-Pandya A, Tephly TR. (1999). Studies on the substrate specificity of human intestinal UDP-glucuronosyltransferases 1A8 and 1A10. Drug Metab Dispos 27:1165–70
  • Ciotti M, Cho JW, George J, Owens IS. (1998). Required buried α-helical structure in the bilirubin UDP-glucuronosyltransferase, UGT1A1, contains a nonreplaceable phenylalanine. Biochemistry 37:11018–25
  • Ciotti M, Marrone A, Potter C, Owens IS. (1997). Genetic polymorphism in the human UGT1A6 (planar phenol) UDP-glucuronosyltransferase: Pharmacological implications. Pharmacogenetics 7:485–95
  • Coleman J, Blake-Kalff M, Davies E. (1997). Detoxification of xenobiotics by plants: Chemical modification and vacuolar compartmentation. Trends Plant Sci 2:144–51
  • Collier AC, Tingle MD, Paxton JW, et al. (2002). Metabolizing enzyme localization and activities in the first trimester human placenta: The effect of maternal and gestational age, smoking and alcohol consumption. Hum Reprod 17:2564–72
  • Coutinho PM, Deleury E, Davies GJ, Henrissat B. (2003). An evolving hierarchical family classification for glycosyltransferases. J Mol Biol 328:307–17
  • Cruciani G, Carosati E, De Boeck B, et al. (2005). MetaSite: Understanding metabolism in human cytochromes from the perspective of the chemist. J Med Chem 48:6970–9
  • de Graaf C, Vermeulen NP, Feenstra KA. (2005). Cytochrome P450 in silico: An integrative modeling approach. J Med Chem 48:2725–55
  • de Leon J. (2003). Glucuronidation enzymes, genes, and psychiatry. Int J Neuropsychopharmacol 6:57–72
  • Dehal SS, Gagne PV, Crespi CL, Patten CJ. (2001). Characterization of a probe substrate and an inhibitor of UDP glucuronosyltransferase (UGT) 1A4 activity in human liver microsomes (HLM) and cDNA-expressed UGT-enzymes. Drug Metab Rev 33:162
  • Dellinger RW, Fang JL, Chen G, et al. (2006). Importance of UDP-glucuronosyltransferase 1A10 (UGT1A10) in the detoxification of polycyclic aromatic hydrocarbons: Decreased glucuronidative activity of the UGT1A10139Lys isoform. Drug Metab Dispos 34:943–9
  • DiMasi JA, Hansen RW, Grabowski HG. (2003). The price of innovation: New estimates of drug development costs. J Health Econ 22:151–85
  • Donato MT, Montero S, Castell JV, et al. (2010). Validated assay for studying activity profiles of human liver UGTs after drug exposure: Inhibition and induction studies. Anal Bioanal Chem 396:2251–63
  • Dong D, Wu B. (2012). In silico modeling of UDP-glucuronosyltransferase 1A10 substrates using the VolSurf approach. J Pharm Sci 101:3531–9
  • Duan SX, Guillemette C, Journault K, et al. (2002). Stereoselective conjugation of oxazepam by human UDP-glucuronosyltransferases (UGTs): S-oxazepam is glucuronidated by UGT2B15, while R-oxazepam is glucuronidated by UGT2B7 and UGT1A9. Drug Metab Dispos 30:1257–65
  • Duan SX, von Moltke LL, Greenblatt DJ, et al. (2001). Interindividual variability in acetaminophen glucuronidation by human liver microsomes: Identification of relevant acetaminophen UDP-glucuronosyltransferase isoforms. J Pharmacol Exp Ther 299:998–1006
  • Dubois SG, Beaulieu M, Lévesque É, et al. (1999). Alteration of human UDP-glucuronosyltransferase UGT2B17 regio-specificity by a single amino acid substitution. J Mol Biol 289:29–39
  • Dutton GJ, Storey ID. (1953). The isolation of a compound of uridine diphosphate and glucuronic acid from liver. Biochem J 53:37–38
  • El Mouelhi M, Beck S, Bock KW. (1993). Stereoselective glucuronidation of (R)-and (S)-naproxen by recombinant rat phenol UDP-glucuronosyltransferase (UGT1A1) and its human orthologue. Biochem Pharmacol 46:1298–300
  • Esteban A, Pérez-Mateo M. (1999). Heterogeneity of paracetamol metabolism in Gilbert’s syndrome. Eur J Drug Metab Pharmacokinet 24:9–13
  • Ethell BT, Anderson GD, Burchell, B. (2003). The effect of valproic acid on drug and steroid glucuronidation by expressed human UDP-glucuronosyltransferases. Biochem Pharmacol 65:1441–9
  • Ethell BT, Ekins S, Wang J, Burchell B. (2002). Quantitative structure activity relationships for the glucuronidation of simple phenols by expressed human UGT1A6 and UGT1A9. Drug Metab Dispos 30:734–8
  • Finel M, Kurkela M. (2008). The UDP-glucuronosyltransferases as oligomeric enzymes. Curr Drug Metab 9:70–6
  • Fisher MB, Paine MF, Strelevitz TJ, Wrighton SA. (2001). The role of hepatic and extrahepatic UDP-glucuronosyltransferases in human drug metabolism. Drug Metab Rev 33:273–97
  • Fisher MB, VandenBranden M, Findlay K, et al. (2000). Tissue distribution and interindividual variation in human UDP-glucuronosyltransferase activity: Relationship between UGT1A1 promoter genotype and variability in a liver bank. Pharmacogenetics 10:727–39
  • Fujino H, Yamada I, Shimada S, et al. (2003). Metabolic fate of pitavastatin, a new inhibitor of HMG-CoA reductase: Human UDP-glucuronosyltransferase enzymes involved in lactonization. Xenobiotica 33:27–41
  • Fujiwara R, Nakajima M, Yamanaka H, et al. (2007). Effects of coexpression of UGT1A9 on enzymatic activities of human UGT1A isoforms. Drug Metab Dispos 35:747–57
  • Fujiwara R, Nakajima M, Yamamoto T, et al. (2009a). In silico and in vitro approaches to elucidate the thermal stability of human UDP-glucuronosyltransferase (UGT) 1A9. Drug Metab Pharmacokinet 24:235–44
  • Fujiwara R, Nakajima M, Yamanaka H, Yokoi, T. (2009b). Key amino acid residues responsible for the differences in substrate specificity of human UDP-glucuronosyltransferase (UGT) 1A9 and UGT1A8. Drug MetabDispos 37:41–6
  • Ghosal A, Hapangama N, Yuan Y, et al. (2004). Identification of human UDP-glucuronosyltransferase enzyme(s) responsible for the glucuronidation of ezetimibe (Zetia). Drug Metab Dispos 32:314–20
  • Ghosh SS, Sappal BS, Kalpana GV, et al. (2001). Homodimerization of human bilirubin-uridine-diphosphoglucuronate glucuronosyl-transferase-1 (UGT1A1) and its functional implications. J Biol Chem 276:42108–15
  • Glare PA, Walsh TD. (1991). Clinical pharmacokinetics of morphine. Ther Drug Monit 13:1–23
  • Grancharov K, Naydenova Z, Lozeva S, Golovinsky E. (2001). Natural and synthetic inhibitors of UDP-glucuronosyltransferase. Pharmacol Ther 89:171–86
  • Green MD, Oturu EM, Tephly TR. (1994). Stable expression of a human liver UDP-glucuronosyltransferase (UGT2B15) with activity toward steroid and xenobiotic substrates. Drug Metab Dispos 22:799–805
  • Guillemette C. (2003). Pharmacogenomics of human UDP-glucuronosyltransferase enzymes. Pharmacogenomics J 3:136–58
  • Guillemette C, Lévesque E, Beaulieu M, et al. (1997). Differential regulation of two uridine diphospho-glucuronosyltransferases, UGT2B15 and UGT2B17, in human prostate LNCaP cells. Endocrinology 138:2998–3005
  • Guillemette C, Ritter JK, Auyeung, DJ, et al. (2000). Structural heterogeneity at the UDP-glucuronosyltransferase 1 locus: Functional consequences of three novel missense mutations in the human UGT1A7 gene. Pharmacogenetics 10:629–44
  • Hagenauer B, Salamon A, Thalhammer T, et al. (2001). In vitro glucuronidation of the cyclin-dependent kinase inhibitor flavopiridol by rat and human liver microsomes: Involvement of UDP-glucuronosyltransferases 1A1 and 1A9. Drug Metab Dispos 29:407–14
  • Hao Q, Krishnaswamy S, Bekaii-Saab T, et al. (2004). UDP-glucuronosyltransferase (UGT) 2B15 pharmacogenetics: UGT2B15 D85Y genotype and gender are major determinants of oxazepam glucuronidation by human liver. J Pharmacol Exp Ther 310:656–65
  • Hariparsad N, Sane RS, Strom SC, Desai PB. (2006). In vitro methods in human drug biotransformation research: Implications for cancer chemotherapy. Toxicol In Vitro 20:135–53
  • Harrington WR, Sengupta S, Katzenellenbogen BS. (2006). Estrogen regulation of the glucuronidation enzyme UGT2B15 in estrogen receptor-positive breast cancer cells. Endocrinology 147:3843–50
  • Herman RJ, Chaudhary A, Szakacs CB. (1994). Disposition of lorazepam in Gilbert’s syndrome: Effects of fasting, feeding, and enterohepatic circulation. J Clin Pharmacol 34:978–84
  • Hiller A, Nguyen N, Strassburg CP, et al. (1999). Retigabine N-glucuronidation and its potential role in enterohepatic circulation. Drug Metab Dispos 27:605–12
  • Hoener B, Benet LZ. (2002). Factors influencing drug absorption and drug availability. In: Banker GS, Rhodes CT, eds. Modern pharmaceutics. 4th ed. New York: Marcel Dekker, 93–117
  • Hu Y, Walker S. (2002). Remarkable structural similarities between diverse glycosyltransferases. Chem Biol 9:1287–96
  • Huang YH, Galijatovic A, Nguyen N, et al. (2002). Identification and functional characterization of UDP-glucuronosyltransferases UGT1A8*1, UGT1A8*2, and UGT1A8*3. Pharmacogenetics 12:287–97
  • Iida A, Saito S, Sekine A, et al. (2002). Catalog of 86 single-nucleotide polymorphisms (SNPs) in three uridine diphosphate glycosyltransferase genes: UGT2A1, UGT2B15, and UGT8. J Hum Genet 47:505–10
  • Ishii Y, Nurrochmad A, Yamada H. (2010). Modulation of UDP-glucuronosyltransferase activity by endogenous compounds. Drug Metab Pharmacokinet 25:134–48
  • Itäaho K, Laakkonen L, Finel M. (2010). How many and which amino acids are responsible for the large activity differences between the highly homologous UDP-glucuronosyltransferases (UGT) 1A9 and UGT1A10? Drug Metab Dispos 38:687–96
  • Itäaho K, Uutela P, Kostiainen R, et al. (2009). Dopamine is a low-affinity and high-specificity substrate for the human UDP-glucuronosyltransferase 1A10. Drug Metab Dispos 37:768–75
  • Ito M, Yamamoto K, Sato H, et al. (2001). Inhibitory effect of troglitazone on glucuronidation catalyzed by human uridine diphosphate-glucuronosyltransferase 1A6. Eur J Clin Pharmacol 56:893–95
  • Iyer L, Das S, Janisch L, et al. (2002). UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2:43–7
  • Iyer L, Hall D, Das S, et al. (1999). Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Clin Pharmacol Ther 65:576–82
  • Jakobsson J, Ekström L, Inotsume N, et al. (2006). Large differences in testosterone excretion in Korean and Swedish men are strongly associated with a UDP-glucuronosyl transferase 2B17 polymorphism. J Clin Endocrinol Metab 91:687–93
  • Jenkinson C, Petroczi A, Barker J, Naughton DP. (2012). Dietary green and white teas suppress UDP-glucuronosyltransferase UGT2B17 mediated testosterone glucuronidation. Steroids 77:691–5
  • Jeong EJ, Liu Y, Lin H, Hu M. (2005). Species-and disposition model-dependent metabolism of raloxifene in gut and liver: Role of UGT1A10. Drug Metab Dispos 33:785–94
  • Jeong H, Choi S, Song JW, et al. (2008). Regulation of UDP-glucuronosyltransferase (UGT) 1A1 by progesterone and its impact on labetalol elimination. Xenobiotica 38:62–75
  • Jinno H, Saeki M, Tanaka-Kagawa T. (2003). Functional characterization of wild-type andvariant (T202I and M59I) human UDP-glucuronosyltransferase 1A10. Drug Metab Dispos 31:528–32
  • Kaji H, Kume T. (2005). Regioselective glucuronidation of denopamine: Marked species differences and identification of human UDP-glucuronosyltransferase isoform. Drug Metab Dispos 33:403–12
  • Kato Y, Ikushiro S, Emi Y, et al. (2008). Hepatic UDP-glucuronosyltransferases responsible for glucuronidation of thyroxine in humans. Drug Metab Dispos 36:51–5
  • Kemp DC, Fan PW, Stevens JC. (2002). Characterization of raloxifene glucuronidation in vitro: Contribution of intestinal metabolism to presystemic clearance. Drug Metab Dispos 30:694–700
  • Kiang TKL, Ensom MHH, Chang TKH. (2005). UDP-glucuronosyltransferases and clinical drug-drug interactions. Pharmacol Therapeut 106:97–132
  • King C, Tang W, Ngui J, et al. (2001). Characterization of rat and human UDP-glucuronosyltransferases responsible for the in vitro glucuronidation of diclofenac. Toxicol Sci 61:49–53
  • King CD, Green MD, Rios GR, et al. (1996). The glucuronidation of exogenous and endogenous compounds by stably expressed rat and human UDP-glucuronosyltransferase 1.1. Arch Biochem Biophys 332:92–100
  • King CD, Rios GR, Green MD, Tephly TR. (2000). UDP-glucuronosyltransferases. Curr Drug Metab 1:143–61
  • Kobayashi T, Sleeman JE, Coughtrie MWH, Burchell B. (2006). Molecular and functional characterization of microsomal UDP-glucuronic acid uptake by members of the nucleotide sugar transporter (NST) family. Biochem J 400:281–9
  • Kola I, Landis J. (2004). Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov 3:711–16
  • Kosoglou T, Statkevich P, Johnson-Levonas AO, et al. (2005). Ezetimibe: A review of its metabolism, pharmacokinetics and drug interactions. Clin Pharmacokinet 44:467–94
  • Krämer SD, Testa B. (2008). The biochemistry of drug metabolism—an introduction: Inter-individual factors affecting drug metabolism. Chem Biodivers 5:2465–578
  • Krishnaswamy S, Duan SX, von Moltke LL, Greenblatt DJ. (2003a). Validation of serotonin (5-hydroxtryptamine) as an in vitro substrate probe for human UDP-glucuronosyltransferase (UGT) 1A6. Drug Metab Dispos 31:133–9
  • Krishnaswamy S, Hao Q, Duan SX, et al. (2003b). Evaluation of 3′-azido-3′-deoxythymidine, morphine, and codeine as probe substrates for UDP-glucuronosyltransferase 2b7 (UGT2b7) in human liver microsomes: Specificity and influence of the UGT2b7*2 polymorphism. Drug Metab Dispos 31:1125–33
  • Krishnaswamy S, Hao Q, von Moltke LL, Greenblatt DJ. (2004). Evaluation of 5-hydroxytryptophol and other endogenous serotonin (5-hydroxytryptamine) analogs as substrates for UDP-glucuronosyltransferase 1A6. Drug Metab Dispos 32:862–9
  • Kurkela M, García-Horsman JA, Luukkanen L, et al. (2003). Expression and characterization of recombinant human UDP-glucuronosyltransferases (UGTs). J Biol Chem 278:3536–44
  • Kurkela M, Hirvonen J, Kostiainen R, Finel M. (2004). The interactions between the N-terminal and C-terminal domains of the human UDP-glucuronosyltransferases are partly isoform-specific, and may involve both monomers. Biochem Pharmacol 68:2443–50
  • Laakkonen L, Finel M. (2010). A molecular model of the human UDP-glucuronosyltransferase 1A1, its membrane orientation, and the interactions between different parts of the enzyme. Mol Pharmacol 77:931–9
  • Lairson LL, Henrissat B, Davies GJ, Withers SG. (2008). Glycosyltransferases: Structures, functions, and mechanisms. Biochemistry 77:521–55
  • Lampe JW, Bigler J, Bush AC, Potter JD. (2000). Prevalence of polymorphisms in the human UDP-glucuronosyltransferase 2B family: UGT2B4 (D458E), UGT2B7 (H268Y), and UGT2B15 (D85Y). Cancer Epidemiol Biomarkers Prev 9:329–33
  • Lampe JW, Bigler J, Horner NK, Potter JD. (1999). UDP-glucuronosyltransferase (UGT1A1*28 and UGT1A6*2) polymorphisms in Caucasians and Asians: Relationships to serum bilirubin concentrations. Pharmacogenetics 9:341–9
  • Langowski J, Long A. (2002). Computer systems for the prediction of xenobiotic metabolism. Adv Drug Deliver Rev 54:407–15
  • Lautala P, Ethell BT, Taskinen J, Burchell B. (2000). The specificity of glucuronidation of entacapone and tolcapone by recombinant human UDP-glucuronosyltransferases. Drug Metab Dispos 28:1385–9
  • Lévesque É, Beaulieu M, Hum DW, Bélanger A. (1999). Characterization and substrate specificity of UGT2B4 (E458): A UDP-glucuronosyltransferase encoded by a polymorphic gene. Pharmacogenetics 9:207–16
  • Lewinsky RH, Smith PA, Mackenzie PI. (2005). Glucuronidation of bioflavonoids by human UGT1A10: Structure-function relationships. Xenobiotica 35:117–29
  • Lewis BC, Mackenzie PI, Elliot DJ, et al. (2007). Amino terminal domains of human UDP-glucuronosyltransferases (UGT) 2B7 and 2B15 associated with substrate selectivity and autoactivation. Biochem Pharmacol 73:1463–73
  • Li D, Fournel-Gigleux S, Barré L, et al. (2007). Identification of aspartic acid and histidine residues mediating the reaction mechanism and the substrate specificity of the human UDP-glucuronosyltransferases 1A. J Biol Chem 282:36514–24
  • Lin JH, Wong BK. (2002). Complexities of glucuronidation affecting in vitro--in vivo extrapolation. Curr Drug Metab 3:623–46
  • Lindstrom TD, Whitaker NG, Whitaker GW. (1984). Disposition and metabolism of a new benzothiophene antiestrogen in rats, dogs, and monkeys. Xenobiotica 14:841–7
  • Liu HX, Liu Y, Zhang JW, et al. (2008). UDP-glucuronosyltransferase 1A6 is the major isozyme responsible for protocatechuic aldehyde glucuronidation in human liver microsomes. Drug Metab Dispos 36:1562–9
  • Liu Y, She M, Wu Z, Dai R. (2011). The inhibition study of human UDP-glucuronosyltransferases with cytochrome P450 selective substrates and inhibitors. J Enzyme Inhib Med Chem 26:386–93
  • Locuson CW, Tracy TS. (2007). Comparative modelling of the human UDP-glucuronosyltransferases: Insights into structure and mechanism. Xenobiotica 37:155–68
  • Loureiro AI, Bonifacio MJ, Fernandes-Lopes C, et al. (2006). Human metabolism of nebicapone (BIA 3-202), a novel catechol-O-methyltransferase inhibitor: Characterization of in vitro glucuronidation. Drug Metab Dispos 34:1856–62
  • Mackenzie PI, Bock KW, Burchell B, et al. (2005). Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics 15:677–85
  • Mackenzie PI, Gregory PA, Gardner-Stephen DA, et al. (2003). Regulation of UDP glucuronosyltransferase genes. Curr Drug Metab 4:249–57
  • Mackenzie PI, Owens IS, Burchell B, et al. (1997). The UDP glycosyltransferase gene superfamily: Recommended nomenclature update based on evolutionary divergence. Pharmacogenetics 7:255–69
  • Mackenzie PI, Rodbourn L. (1990). Organization of the rat UDP-glucuronosyltransferase, UDPGTr-2, gene, and characterization of its promoter. J Biol Chem 265:11328–32
  • MacLeod SL, Nowell S, Plaxco J, Lang NP. (2000). An allele-specific polymerase chain reaction method for the determination of the D85Y polymorphism in the human UDP-glucuronosyltransferase 2B15 gene in a case-control study of prostate cancer. Ann Surg Oncol 7:777–82
  • Magdalou J, Fournel-Gigleux S, Ouzzine M. (2010). Insights on membrane topology and structure/function of UDP-glucuronosyltransferases. Drug Metab Rev 42:159–66
  • Malfatti MA, Felton JS. (2004). Human UDP-glucuronosyltransferase 1A1 is the primary enzyme responsible for the N-glucuronidation of N-hydroxy-PhIP in vitro. Chem Res Toxicol 17:1137–44
  • Mano Y, Usui T, Kamimura H. (2006). Identification of human UDP-glucuronosyltransferase responsible for the glucuronidation of niflumic acid in human liver. Pharm Res 23:1502–8
  • Mano Y, Usui T, Kamimura H. (2007). The UDP-glucuronosyltransferase 2B7 isozyme is responsible for gemfibrozil glucuronidation in the human liver. Drug Metab Dispos 35:2040–4
  • Marsh S, McLeod HL. (2004). Cancer pharmacogenetics. Br J Cancer 90:8–11
  • Martineau I, Tchernof A, Béanger A. (2004). Amino acid residue ILE211 is essential for the enzymatic activity of human UDP-glucuronosyltransferase 1A10 (UGT1A10). Drug Metab Dispos 32:455–9
  • Meech R, Mackenzie PI. (1997). UDP-glucuronosyltransferase, the role of the amino terminus in dimerization. J Biol Chem 272:26913–17
  • Miley MJ, Zielinska AK, Keenan JE, et al. (2007). Crystal structure of the cofactor-binding domain of the human phase II drug-metabolism enzyme UDP-glucuronosyltransferase 2B7. J Mol Biol 369:498–511
  • Miners JO, Bowalgaha K, Elliot DJ, et al. (2011). Characterization of niflumic acid as a selective inhibitor of human liver microsomal UDP-glucuronosyltransferase 1A9: Application to the reaction phenotyping of acetaminophen glucuronidation. Drug Metab Dispos 39:644–52
  • Miners JO, Knights KM, Houston JB, Mackenzie PI. (2006). In vitro-in vivo correlation for drugs and other compounds eliminated by glucuronidation in humans: Pitfalls and promises. Biochem Pharmacol 71:1531–9
  • Miners JO, Mackenzie PI. (1991). Drug glucuronidation in humans. Pharmacol Ther 51:347–69
  • Miners JO, McKinnon RA, Mackenzie PI. (2002). Genetic polymorphisms of UDP-glucuronosyltransferases and their functional significance. Toxicology 181:453–6
  • Miners JO, Smith PA, Sorich MJ, et al. (2004). Predicting human drug glucuronidation parameters: Application of in vitro and in silico modeling approaches. Annu Rev Pharmacol Toxicol 44:1–25
  • Modolo LV, Blount JW, Achnine L, et al. (2007). A functional genomics approach to (iso)flavonoid glycosylation in the model legume Medicago truncatula. Plant Mol Biol 64:499–518
  • Motohashi H, Yamamoto M. (2004). Nrf2–Keap1 defines a physiologically important stress response mechanism. Trends Mol Med 10:549–57
  • Münzel PA, Schmohl S, Heel H, et al. (1999). Induction of human UDP glucuronosyltransferases (UGT1A6, UGT1A9, and UGT2B7) by t-butylhydroquinone and 2,3,7,8-tetrachlorodibenzo-p-dioxin in Caco-2 cells. Drug Metab Dispos 27:569–73
  • Murai T, Samata N, Iwabuchi H, Ikeda T. (2006). Human UDP-glucuronosyltransferase, UGT1A8, glucuronidates dihydrotestosterone to a monoglucuronide and further to a structurally novel diglucuronide. Drug Metab Dispos 34:1102–8
  • Nakajima M, Koga T, Sakai H, et al. (2010). N-Glycosylation plays a role in protein folding of human UGT1A9. Biochem Pharmacol 79:1165–72
  • Nassar AEF, Kamel AM. (2004). Metabolite characterization—one key to success in drug discovery. PharmaTech 2004:46–51
  • Nishimura M, Koeda A, Morikawa H, et al. (2009). Tissue-specific mRNA expression profiles of drug-metabolizing enzymes and transporters in the cynomolgus monkey. Drug Metab Pharmacokinet 24:139–44
  • Nishiyama T, Ogura K, Nakano H, et al. (2002). Reverse geometrical selectivity in glucuronidation and sulfation of cis-and trans-4-hydroxytamoxifens by human liver UDP-glucuronosyltransferases and sulfotransferases. Biochem Pharmacol 63:1817–30
  • Nishiyama T, Kobori T, Arai K, et al. (2006). Identification of human UDP-glucuronosyltransferase isoform (s) responsible for the C-glucuronidation of phenylbutazone. Arch Biochem Biophys 454:72–9
  • Offen W, Martinez-Fleites C, Yang M, et al. (2006). Structure of a flavonoid glucosyltransferase reveals the basis for plant natural product modification. EMBO J 25:1396–405
  • Oguri T, Takahashi T, Miyazaki M, et al. (2004). UGT1A10 is responsible for SN-38 glucuronidation and its expression in human lung cancers. Anticancer Res 24:2893–6
  • Ohno A, Saito Y, Hanioka N, et al. (2004). Involvement of human hepatic UGT1A1, UGT2B4, and UGT2B7 in the glucuronidation of carvedilol. Drug Metab Dispos 32:235–9
  • Ohno S, Nakajin S. (2009). Determination of mRNA expression of human UDP-glucuronosyltransferases and application for localization in various human tissues by real-time reverse transcriptase-polymerase chain reaction. Drug Metab Dispos 37:32–40
  • Okolicsanyi L, Venuti M, Strazzabosco M, et al. (1985). Pharmacokinetics of Josamycin in patients with liver cirrhosis and Gilbert’s syndrome after repeated doses. Int J Clin Pharmacol Ther Toxicol 8:434–8
  • Oliveira EJ, Watson DG. (2000). In vitro glucuronidation of kaempferol and quercetin by human UGT-1A9 microsomes. FEBS Lett 471:1–6
  • Ouzzine M, Antonio L, Burchell B, et al. (2000). Importance of histidine residues for the function of the human liver UDP-glucuronosyltransferase UGT1A6: Evidence for the catalytic role of histidine 370. Mol Pharmacol 58:1609–15
  • Ouzzine M, Barré L, Netter P, et al. (2003). The human UDP-glucuronosyltransferases: Structural aspects and drug glucuronidation. Drug Metab Rev 35:287–303
  • Ouzzine M, Magdalou J, Burchell B, Fournel-Gigleux S. (1999). An internal signal sequence mediates the targeting and retention of the human UDP-glucuronosyltransferase 1A6 to the endoplasmic reticulum. J Biol Chem 274:31401–9
  • Paquette S, Møller BL, Bak S. (2003). On the origin of family 1 plant glycosyltransferases. Phytochemistry 62:399–413
  • Park J, Chen L, Shade K, et al. (2004). Asp85tyr polymorphism in the udp-glucuronosyltransferase (UGT) 2B15 gene and the risk of prostate cancer. J Urol 171:2484–8
  • Patana AS, Kurkela M, Finel M, Goldman A. (2008). Mutation analysis in UGT1A9 suggests a relationship between substrate and catalytic residues in UDP-glucuronosyltransferases. Protein Eng Des Sel 21:537–43
  • Posner J, Cohen AF, Land G, et al. (1989). The pharmacokinetics of lamotrigine (BW430C) in healthy subjects with unconjugated hyperbilirubinaemia (Gilbert’s syndrome). Br J Clin Pharmacol 28:117–20
  • Quiding H, Anderson P, Bondesson U, et al. (1986). Plasma concentrations of codeine and its metabolite, morphine, after single and repeated oral administration. Eur J Clin Pharmacol 30:673–7
  • Radominska-Pandya A. (2012). Compositions comprising specific UGT inhibitors and methods of use thereof. Google Patents: 2012. Publication no.: US 2012/0184536 A1, filed on 27 February 2012
  • Radominska-Pandya A, Czernik PJ, Little JM, et al. (1999). Structural and functional studies of UDP-glucuronosyltransferases. Drug Metab Rev 31:817–99
  • Radominska-Pandya A, Bratton SM, Redinbo MR, Miley MJ. (2010). The crystal structure of human UDP-glucuronosyltransferase 2B7 C-terminal end is the first mammalian UGT target to be revealed: The significance for human UGTs from both the 1A and 2B families. Drug Metab Rev 42:133–44
  • Rae JM, Johnson MD, Lippman ME, Flockhart DA. (2001). Rifampin is a selective, pleiotropic inducer of drug metabolism genes in human hepatocytes: Studies with cDNA and oligonucleotide expression arrays. J Pharmacol Exp Ther 299:849–57
  • Remmel RP, Nagar S, Argikar UA. (2007). Conjugative metabolism of drugs. In: Zhang D, Zhu M, Humphreys WG, eds. Drug metabolism in drug design and development. Hoboken (NJ): Wiley-Interscience, John Wiley and Sons, 37–8
  • Ritter JK. (2007). Intestinal UGTs as potential modifiers of pharmacokinetics and biological responses to drugs and xenobiotics. Expert Opin Drug Metab 3:93–107
  • Ritter JK, Kessler FK, Thompson MT, et al. (1999). Expression and inducibility of the human bilirubin UDP-glucuronosyltransferase UGT1A1 in liver and cultured primary hepatocytes: Evidence for both genetic and environmental influences. Hepatology 30:476–84
  • Ross J, Li Y, Lim EK, Bowles DJ. (2001). Higher plant glycosyltransferases. Genome Biol 2:3004.1–6
  • Rouleau M, Collin P, Bellemare J, et al. (2012). Protein--protein interactions between the bilirubin-conjugating UDP-glucuronosyltransferase UGT1A1 and its shorter isoform 2 regulatory partner derived from alternative splicing. Biochem J 450:107–14
  • Sabolovic N, Heydel JM, Li X, et al. (2004). Carboxyl nonsteroidal anti-inflammatory drugs are efficiently glucuronidated by microsomes of the human gastrointestinal tract. Biochim Biophys Acta 1675:120–9
  • Sabolovic N, Humbert AC, Radominska-Pandya A, Magdalou J. (2006). Resveratrol is efficiently glucuronidated by UDP-glucuronosyltransferases in the human gastrointestinal tract and in Caco-2 cells. Biopharm Drug Dispos 27:181–9
  • Sabolovic N, Magdalou J, Netter P, Abid A. (2000). Nonsteroidal anti-inflammatory drugs and phenols glucuronidation in Caco-2 cells: Identification of the UDP-glucuronosyltransferases UGT1A6, 1A3, and 2B7. Life Sci 67:185–96
  • Saeki M, Ozawa S, Saito Y. (2002). Three novel single nucleotide polymorphisms in UGT1A10. Drug Metab Pharmacokinet 17:488–90
  • Sakaguchi K, Green M, Stock N, et al. (2004). Glucuronidation of carboxylic acid containing compounds by UDP-glucuronosyltransferase isoforms. Arch Biochem Biophys 424:219–25
  • Schuster D, Laggner C, Langer T. (2005). Why drugs fail—a study on side effects in new chemical entities. Curr Pharm Des 11:3545–59
  • Senay C, Jedlitschky G, Terrier N, et al. (2002). The importance of cysteine 126 in the human liver UDP-glucuronosyltransferase UGT1A6. Biochim Biophys Acta 1597:90–6
  • Shao H, He X, Achnine L, et al. (2005). Crystal structures of a multifunctional triterpene/flavonoid glycosyltransferase from Medicago truncatula. Plant Cell 17:3141–54
  • Shelby MK, Cherrington NJ, Vansell NR, Klaassen CD. (2003). Tissue mRNA expression of the rat UDP-glucuronosyltransferase gene family. Drug Metab Dispos 31:326–33
  • Shiratani H, Katoh M, Nakajima M, Yokoi T. (2008). Species differences in UDP-glucuronosyltransferase activities in mice and rats. Drug Metab Dispos 36:1745–52
  • Singh SS. (2006). Preclinical pharmacokinetics: An approach towards safer and efficacious drugs. Curr Drug Metab 7:165–82
  • Smith CM, Faucette SR, Wang H, LeCluyse EL. (2005). Modulation of UDP-glucuronosyltransferase 1A1 in primary human hepatocytes by prototypical inducers. J Biochem Mol Toxicol 19:96–108
  • Smith DA. (1994). Design of drugs through a consideration of drug metabolism and pharmacokinetics. Eur J Drug Metab Pharmacokinet 19:193–9
  • Smith PA, Sorich MJ, Low LSC, et al. (2004). Towards integrated ADME prediction: Past, present and future directions for modelling metabolism by UDPglucuronosyltransferases. J Mol Graph Mod 22:507–17
  • Soars MG, Ring BJ, Wrighton SA. (2003). The effect of incubation conditions on the enzyme kinetics of UDP-glucuronosyltransferases. Drug Metab Dispos 31:762–7
  • Sorich MJ, Smith PA, McKinnon RA, Miners JO. (2002). Pharmacophore and quantitative structure activity relationship modelling of UDP-glucuronosyltransferase 1A1 (UGT1A1) substrates. Pharmacogenetics 12:635–45
  • Sorich MJ, Miners JO, McKinnon RA, Smith PA. (2004). Multiple pharmacophores for the investigation of human UDP-glucuronosyltransferase isoform substrate selectivity. Mol Pharmacol 65:301–8
  • Staines AG, Coughtrie MWH, Burchell B. (2004). N-glucuronidation of carbamazepine in human tissues is mediated by UGT2B7. J Pharmacol Exp Ther 311:1131–7
  • Starlard-Davenport A, Lyn-Cook B, Radominska-Pandya A. (2008). Identification of UDP-glucuronosyltransferase 1A10 in non-malignant and malignant human breast tissues. Steroids 73:611–20
  • Sten T, Qvisen S, Uutela P, et al. (2006). Prominent but reverse stereoselectivity in propranolol glucuronidation by human UDP-glucuronosyltransferases 1A9 and 1A10. Drug Metab Dispos 34:1488–94
  • Stone AN, Mackenzie PI, Galetin A, et al. (2003). Isoform selectivity and kinetics of morphine 3- and 6-glucuronidation by human UDP-glucuronosyltransferases: Evidence for atypical glucuronidation kinetics by UGT2B7. Drug Metab Dispos 31:1086–9
  • Strassburg CP, Barut A, Obermayer-Straub P, et al. (2001). Identification of cyclosporine A and tacrolimus glucuronidation in human liver and the gastrointestinal tract by a differentially expressed UDP-glucuronosyltransferase: UGT2B7. J Hepatol 34:865–72
  • Strassburg CP, Manns MP, Tukey RH. (1998). Expression of the UDP-glucuronosyltransferase 1A locus in human colon. J Biol Chem 273:8719–26
  • Strassburg CP, Oldhafer K, Manns MP, Tukey RH. (1997). Differential expression of the UGT1A locus in human liver, biliary, and gastric tissue: Identification of UGT1A7 and UGT1A10 transcripts in extrahepatic tissue. Mol Pharmacol 52:212–20
  • Sugatani J. (2012). Function, genetic polymorphism, and transcriptional regulation of human UDP-glucuronosyltransferase (UGT) 1A1. Drug Metab Pharmacokinet 2012 Oct 23 [Epub ahead of print]
  • Sugatani J, Yamakawa K, Tonda E, et al. (2004). The induction of human UDP-glucuronosyltransferase 1A1 mediated through a distal enhancer module by flavonoids and xenobiotics. Biochem Pharmacol 67:989–1000
  • Sun D, Chen G, Dellinger RW, et al. (2010). Characterization of 17-dihydroexemestane glucuronidation: Potential role of the UGT2B17 deletion in exemestane pharmacogenetics. Pharmacogenet Genomics 20:575–85
  • Sun D, Sharma AK, Dellinger RW, et al. (2007). Glucuronidation of active tamoxifen metabolites by the human UDP glucuronosyltransferases. Drug Metab Dispos 35:2006–14
  • Takaoka Y, Ohta M, Takeuchi A, et al. (2010). Ligand orientation governs conjugation capacity of UDP-glucuronosyltransferase 1A1. J Biochem 148:25–8
  • Tang C, Hochman JH, Ma B, et al. (2003). Acyl glucuronidation and glucosidation of a new and selective endothelin ETA receptor antagonist in human liver microsomes. Drug Metab Dispos 31:37–45
  • Tephly TR, Burchell B. (1990). UDP-glucuronosyltransferases: A family of detoxifying enzymes. Trends Pharmacol Sci 11:276–9
  • Testa B, Balmat AL, Long A, Judson P. (2005). Predicting drug metabolism—an evaluation of the expert system METEOR. Chem Biodivers 2:872–85
  • Tsoutsikos P, Miners JO, Stapleton A, et al. (2004). Evidence that unsaturated fatty acids are potent inhibitors of renal UDP-glucuronosyltransferases (UGT): Kinetic studies using human kidney cortical microsomes and recombinant UGT1A9 and UGT2B7. Biochem Pharmacol 67:191–9
  • Tukey RH, Strassburg CP. (2000). Human UDP-glucuronosyltransferases: Metabolism, expression, and disease. Annu Rev Pharmacol Toxicol 40:581–616
  • Turgeon D, Carrier JS, Chouinard S, Bélanger A. (2003). Glucuronidation activity of the UGT2B17 enzyme toward xenobiotics. Drug Metab Dispos 31:670–6
  • Uchaipichat V, Mackenzie PI, Guo XH, et al. (2004). Human UDP-glucuronosyltransferases: Isoform selectivity and kinetics of 4-methylumbelliferone and 1-naphthol glucuronidation, effects of organic solvents, and inhibition by diclofenac and probenecid. Drug Metab Dispos 32:413–23
  • Ullrich D, Sieg A, Blume R, et al. (1987). Normal pathways for glucuronidation, sulphation, and oxidation of paracetamol in Gilbert’s syndrome. Eur J Clin Invest 17:237–40
  • Ünligil UM, Rini JM. (2000). Glycosyltransferase structure and mechanism. Curr Opin Struct Biol 10:510–17
  • van de Waterbeemd H, Gifford E. (2003). ADMET in silico modelling: Towards prediction paradise? Nat Rev Drug Discov 2:192–204
  • Villeneuve L, Girard H, Fortier LC. (2003). Novel functional polymorphisms in the UGT1A7 and UGT1A9 glucuronidating enzymes in Caucasian and African-American subjects and their impact on the metabolism of 7-ethyl-10-hydroxycamptothecin and flavopiridol anticancer drugs. J Pharmacol Exp Ther 307:117–28
  • Walle T, Hsieh F, DeLegge MH, et al. (2004). High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab Dispos 32:1377–82
  • Walle T, Otake Y, Galijatovic A, et al. (2000). Induction of UDP-glucuronosyltransferase UGT1A1 by the flavonoid chrysin in the human hepatoma cell line hep G2. Drug Metab Dispos 28:1077–82
  • Watanabe Y, Nakajima M, Ohashi N, et al. (2003). Glucuronidation of etoposide in human liver microsomes is specifically catalyzed by UDP-glucuronosyltransferase 1A1. Drug Metab Dispos 31:589–95
  • Watanabe Y, Nakajima M, Yokoi T. (2002). Troglitazone glucuronidation in human liver and intestine microsomes: High catalytic activity of UGT1A8 and UGT1A10. Drug Metab Dispos 30:1462–9
  • Webb LJ, Miles KK, Auyeung DJ, et al. (2005). Analysis of substrate specificities and tissue expression of rat UDP-glucuronosyltransferases UGT1A7 and UGT1A8. Drug Metab Dispos 33:77–82
  • Wells PG, Mackenzie PI, Chowdhury JR, et al. (2004). Glucuronidation and the UDP-glucuronosyltransferases in health and disease. Drug Metab Dispos 32:281–90
  • Wen Z, Tallman MN, Ali SY, Smith PC. (2007). UDP-glucuronosyltransferase 1A1 is the principal enzyme responsible for etoposide glucuronidation in human liver and intestinal microsomes: Structural characterization of phenolic and alcoholic glucuronides of etoposide and estimation of enzyme kinetics. Drug Metab Dispos 35:371–80
  • Wiggins CAR, Munro S. (1998). Activity of the yeast MNN1 α-1, 3-mannosyltransferase requires a motif conserved in many other families of glycosyltransferases. Proc Natl Acad Sci U S A 95:7945–50
  • Williams JA, Ring BJ, Cantrell VE, et al. (2002). Differential modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed estradiol-3-glucuronidation by the addition of UGT1A1 substrates and other compounds to human liver microsomes. Drug Metab Dispos 30:1266–73
  • Wilson W, Pardo-Manuel de Villena F, Lyn-Cook BD, et al. (2004). Characterization of a common deletion polymorphism of the UGT2B17 gene linked to UGT2B15. Genomics 84:707–14
  • Wu B, Morrow JK, Singh R, et al. (2011). Three-dimensional quantitative structure-activity relationship studies on UGT1A9-mediated 3-O-glucuronidation of natural flavonols using a pharmacophore-based comparative molecular field analysis model. J Pharmacol Exp Ther 336:403–13
  • Xiong Y, Bernardi D, Bratton S, et al. (2006). Phenylalanine 90 and 93 are localized within the phenol binding site of human UDP-glucuronosyltransferase 1A10 as determined by photoaffinity labeling, mass spectrometry, and site-directed mutagenesis. Biochemistry 45:2322–32
  • Xiong Y, Patana AS, Miley MJ, et al. (2008). The first aspartic acid of the DQxD motif for human UDP-glucuronosyltransferase 1A10 interacts with UDP-glucuronic acid during catalysis. Drug Metab Dispos 36:517–22
  • Yamanaka H, Nakajima M, Katoh M, Yokoi T. (2007). Glucuronidation of thyroxine in human liver, jejunum, and kidney microsomes. Drug Metab Dispos 35:1642–8
  • Yin H, Bennett G, Jones JP. (1994). Mechanistic studies of uridine diphosphate glucuronosyltransferase. Chem Biol Interact 90:47–58
  • Yong WP, Ramirez J, Innocenti F, Ratain MJ. (2005). Effects of ketoconazole on glucuronidation by UDP-glucuronosyltransferase enzymes. Clin Cancer Res 11:6699–704
  • Yueh MF, Huang YH, Hiller A, et al. (2003). Involvement of the xenobiotic response element (XRE) in Ah receptor-mediated induction of human UDP-glucuronosyltransferase 1A1. J Biol Chem 278:15001–6
  • Zakim D, Dannenberg AJ. (1992). How does the microsomal membrane regulate UDP-glucuronosyltransferases? Biochem Pharmacol 43:1385–93
  • Zamek-Gliszczynski MJ, Hoffmaster KA, Nezasa K, et al. (2006). Integration of hepatic drug transporters and phase II metabolizing enzymes: Mechanisms of hepatic excretion of sulfate, glucuronide, and glutathione metabolites. Eur J Pharm Sci 27:447–86
  • Zhang D, Cui D, Gambardella J, et al. (2007b). Characterization of the UDP glucuronosyltransferase activity of human liver microsomes genotyped for the UGT1A1*28 polymorphism. Drug Metab Dispos 35:2270–80
  • Zhang JY, Zhan J, Cook CS, et al. (2003). Involvement of human UGT2B7 and 2B15 in rofecoxib metabolism. Drug Metab Dispos 31:652–8
  • Zhang L, Lin G, Chang Q, Zuo Z. (2005). Role of intestinal first-pass metabolism of baicalein in its absorption process. Pharm Res 22:1050–8
  • Zhang L, Lin G, Zuo Z. (2007a). Involvement of UDP-glucuronosyltransferases in the extensive liver and intestinal first-pass metabolism of flavonoid baicalein. Pharm Res 24:81–9
  • Zheng Z, Park JY, Guillemette C, et al. (2001). Tobacco carcinogen-detoxifying enzyme UGT1A7 and its association with orolaryngeal cancer risk. J Natl Cancer Inst 93:1411–18
  • Zhou J, Zhang J, Xie W. (2005). Xenobiotic nuclear receptor-mediated regulation of UDP-glucuronosyltransferases. Curr Drug Metab 6:289–98
  • Zhu BT, Taneja N, Loder DP, et al. (1998). Effects of tea polyphenols and flavonoids on liver microsomal glucuronidation of estradiol and estrone. J Steroid Biochem Mol Biol 64:207–15
  • Zucker K, Tsaroucha A, Olson L, et al. (1999). Evidence that tacrolimus augments the bioavailability of mycophenolate mofetil through the inhibition of mycophenolic acid glucuronidation. Ther Drug Monit 21:35–43

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.