51
|
Uno Y, Takahira R, Murayama N, Ishii Y, Ikenaka Y, Ishizuka M, Yamazaki H, Ikushiro S. Molecular and functional characterization of UDP-glucuronosyltransferase 1A in cynomolgus macaques. Biochem Pharmacol 2018; 155:172-181. [DOI: 10.1016/j.bcp.2018.06.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/22/2018] [Indexed: 12/19/2022]
|
52
|
Bock KW. From TCDD-mediated toxicity to searches of physiologic AHR functions. Biochem Pharmacol 2018; 155:419-424. [PMID: 30055148 DOI: 10.1016/j.bcp.2018.07.032] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022]
Abstract
TCDD-mediated toxicity of human individuals together with animal studies led to identification of the aryl hydrocarbon receptor (AHR). It was characterized as multifunctional ligand-activated transcription factor and environmental sensor. Comparison of human toxic responses and animal models provide hints to physiologic AHR functions including chemical and microbial defense, homeostasis of stem/progenitor cells and modulation of the immune system in barrier organs such as skin and the gastrointestinal tract. Extrapolation from animals to humans is difficult due to marked species differences and dependence of AHR function on the cellular context. Nevertheless, therapeutic possibilities of AHR agonists and antagonists are in development. The AHR remains challenging and fascinating.
Collapse
Affiliation(s)
- Karl Walter Bock
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstrasse 56, D-72074 Tübingen, Germany.
| |
Collapse
|
53
|
Hu DG, Hulin JA, Wijayakumara DD, McKinnon RA, Mackenzie PI, Meech R. Intergenic Splicing between Four Adjacent UGT Genes (2B15, 2B29P2, 2B17, 2B29P1) Gives Rise to Variant UGT Proteins That Inhibit Glucuronidation via Protein-Protein Interactions. Mol Pharmacol 2018; 94:938-952. [DOI: 10.1124/mol.118.111773] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/08/2018] [Indexed: 01/12/2023] Open
|
54
|
Adam AAA, van der Mark VA, Donkers JM, Wildenberg ME, Oude Elferink RPJ, Chamuleau RAFM, Hoekstra R. A practice-changing culture method relying on shaking substantially increases mitochondrial energy metabolism and functionality of human liver cell lines. PLoS One 2018; 13:e0193664. [PMID: 29672606 PMCID: PMC5908182 DOI: 10.1371/journal.pone.0193664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/15/2018] [Indexed: 12/22/2022] Open
Abstract
Practice-changing culturing techniques of hepatocytes are highly required to increase their differentiation. Previously, we found that human liver cell lines HepaRG and C3A acquire higher functionality and increased mitochondrial biogenesis when cultured in the AMC-Bioartificial liver (BAL). Dynamic medium flow (DMF) is one of the major contributors to this stimulatory effect. Recently, we found that DMF-culturing by shaking of HepaRG monolayers resulted in higher mitochondrial biogenesis. Here we further investigated the effect of DMF-culturing on energy metabolism and hepatic functionality of HepaRG and C3A monolayers. HepaRG and C3A DMF-monolayers were incubated with orbital shaking at 60 rpm during the differentiation phase, while control monolayers were maintained statically. Subsequently, energy metabolism and hepatic functionality were compared between static and DMF-cultures. DMF-culturing of HepaRG cells substantially increased hepatic differentiation; transcript levels of hepatic structural genes and hepatic transcription regulators were increased up to 15-fold (Cytochrome P450 3A4) and nuclear translocation of hepatic transcription factor CEBPα was stimulated. Accordingly, hepatic functions were positively affected, including ammonia elimination, urea production, bile acid production, and CYP3A4 activity. DMF-culturing shifted energy metabolism from aerobic glycolysis towards oxidative phosphorylation, as indicated by a decline in lactate production and glucose consumption, and an increase in oxygen consumption. Similarly, DMF-culturing increased mitochondrial energy metabolism and hepatic functionality of C3A cells. In conclusion, simple shaking of monolayer cultures substantially improves mitochondrial energy metabolism and hepatic differentiation of human liver cell lines. This practice-changing culture method may prove to prolong the in-vitro maintenance of primary hepatocytes and increase hepatic differentiation of stem cells.
Collapse
Affiliation(s)
- Aziza A. A. Adam
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Vincent A. van der Mark
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Experimental Surgical Laboratory, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Joanne M. Donkers
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Manon E. Wildenberg
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Department Of Gastroenterology and Hepatology, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Ronald P. J. Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Robert A. F. M. Chamuleau
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Ruurdtje Hoekstra
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Experimental Surgical Laboratory, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
55
|
Gufford BT, Robarge JD, Eadon MT, Gao H, Lin H, Liu Y, Desta Z, Skaar TC. Rifampin modulation of xeno- and endobiotic conjugating enzyme mRNA expression and associated microRNAs in human hepatocytes. Pharmacol Res Perspect 2018; 6:e00386. [PMID: 29610665 PMCID: PMC5869567 DOI: 10.1002/prp2.386] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/12/2018] [Indexed: 01/06/2023] Open
Abstract
Rifampin is a pleiotropic inducer of multiple drug metabolizing enzymes and transporters. This work utilized a global approach to evaluate rifampin effects on conjugating enzyme gene expression with relevance to human xeno‐ and endo‐biotic metabolism. Primary human hepatocytes from 7 subjects were treated with rifampin (10 μmol/L, 24 hours). Standard methods for RNA‐seq library construction, EZBead preparation, and NextGen sequencing were used to measure UDP‐glucuronosyl transferase UGT, sulfonyltransferase SULT, N acetyltransferase NAT, and glutathione‐S‐transferase GST mRNA expression compared to vehicle control (0.01% MeOH). Rifampin‐induced (>1.25‐fold) mRNA expression of 13 clinically important phase II drug metabolizing genes and repressed (>1.25‐fold) the expression of 3 genes (P < .05). Rifampin‐induced miRNA expression changes correlated with mRNA changes and miRNAs were identified that may modulate conjugating enzyme expression. NAT2 gene expression was most strongly repressed (1.3‐fold) by rifampin while UGT1A4 and UGT1A1 genes were most strongly induced (7.9‐ and 4.8‐fold, respectively). Physiologically based pharmacokinetic modeling (PBPK) was used to simulate the clinical consequences of rifampin induction of CYP3A4‐ and UGT1A4‐mediated midazolam metabolism. Simulations evaluating isolated UGT1A4 induction predicted increased midazolam N‐glucuronide exposure (~4‐fold) with minimal reductions in parent midazolam exposure (~10%). Simulations accounting for simultaneous induction of both CYP3A4 and UGT1A4 predicted a ~10‐fold decrease in parent midazolam exposure with only a ~2‐fold decrease in midazolam N‐glucuronide metabolite exposure. These data reveal differential effects of rifampin on the human conjugating enzyme transcriptome and potential associations with miRNAs that form the basis for future mechanistic studies to elucidate the interplay of conjugating enzyme regulatory elements.
Collapse
Affiliation(s)
- Brandon T Gufford
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| | - Jason D Robarge
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| | - Michael T Eadon
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| | - Hongyu Gao
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN
| | - Hai Lin
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN
| | - Yunlong Liu
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN
| | - Zeruesenay Desta
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| | - Todd C Skaar
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| |
Collapse
|
56
|
Juvonen RO, Rauhamäki S, Kortet S, Niinivehmas S, Troberg J, Petsalo A, Huuskonen J, Raunio H, Finel M, Pentikäinen OT. Molecular Docking-Based Design and Development of a Highly Selective Probe Substrate for UDP-glucuronosyltransferase 1A10. Mol Pharm 2018; 15:923-933. [PMID: 29421866 PMCID: PMC6150735 DOI: 10.1021/acs.molpharmaceut.7b00871] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
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.
Collapse
Affiliation(s)
- Risto O Juvonen
- School of Pharmacy, Faculty of Health Sciences , University of Eastern Finland , Box 1627, FI-70211 Kuopio , Finland
| | | | | | | | - Johanna Troberg
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy , University of Helsinki , P.O. Box 56, FI-00014 Helsinki , Finland
| | - Aleksanteri Petsalo
- School of Pharmacy, Faculty of Health Sciences , University of Eastern Finland , Box 1627, FI-70211 Kuopio , Finland
| | | | - Hannu Raunio
- School of Pharmacy, Faculty of Health Sciences , University of Eastern Finland , Box 1627, FI-70211 Kuopio , Finland
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy , University of Helsinki , P.O. Box 56, FI-00014 Helsinki , Finland
| | - Olli T Pentikäinen
- Institute of Biomedicine, Faculty of Medicine , University of Turku , FI-20014 Turku , Finland
| |
Collapse
|
57
|
Wijayakumara DD, Mackenzie PI, McKinnon RA, Hu DG, Meech R. Regulation of UDP-Glucuronosyltransferase 2B15 by miR-331-5p in Prostate Cancer Cells Involves Canonical and Noncanonical Target Sites. J Pharmacol Exp Ther 2018; 365:48-59. [PMID: 29367276 DOI: 10.1124/jpet.117.245936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/30/2017] [Indexed: 01/08/2023] Open
Abstract
UGT2B15 is an important androgen-metabolizing UDP-glucuronosyltransferase (UGT) and the mechanisms controlling its expression are of considerable interest. Recent studies showed that miR-376c regulates UGT2B15 in prostate cancer cells via a canonical target site in the 3' untranslated region (3'UTR). The UGT2B15 3'UTR also contains a canonical miR-331-5p target site; previous work indicated that deleting this site reduced, but did not abolish, the ability of miR-331-5p to repress a luciferase reporter carrying the UGT2B15 3'UTR We report here the discovery and characterization of a second, noncanonical miR-331-5p target site in the UGT2B15 3'UTR miR-331-5p-mediated repression of a UGT2B15 3'UTR-reporter was partly inhibited by mutating either of the two miR-331-5p target sites separately, but completely abolished by mutating the two sites simultaneously, indicating that the two sites act cooperatively. miR-331-5p mimics significantly reduced both UGT2B15 mRNA levels and glucuronidation activity in prostate cancer cells, confirming that the native transcript is a miR-331-5p target. Transfection of either miR-331-5p or miR-376c mimics repressed the activity of the UGT2B15 3'UTR-reporter; however, cotransfection of both microRNAs (miRNAs) further reduced activity, indicating cooperative regulation by these two miRNAs. A significant negative correlation between miR-331 and UGT2B15 mRNA levels was observed in a tissue RNA panel, and analysis of The Cancer Genome Atlas (TCGA) hepatocellular carcinoma data set provided further evidence that miR-331 may play an important role in regulation of UGT2B15 in vivo. There was no significant correlation between miR-331 and UGT2B15 mRNA levels in the TCGA prostate adenocarcinoma cohort, which may reflect the complexity of androgen-mediated regulation in determining UGT2B15 levels in prostate cancer. Finally, we show that miR-331-5p does not regulate UGT2B17, providing the first evidence for a post-transcriptional mechanism that differentially regulates these two important androgen-metabolizing UGTs.
Collapse
Affiliation(s)
- Dhilushi D Wijayakumara
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Peter I Mackenzie
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Ross A McKinnon
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Robyn Meech
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
58
|
Wang P, Nie YL, Wang SJ, Yang LL, Yang WH, Li JF, Li XT, Zhang LR. Regulation of UGT1A expression by miR-298 in human livers from the Han Chinese population and in human cell lines. Epigenomics 2018; 10:43-57. [DOI: 10.2217/epi-2017-0068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: This study aimed to investigate the role of miRNAs in UGT1A regulation. Materials & methods: Based on bioinformatic prediction results, luciferase reporter assay and cell-transfection experiments were performed to study effects of miR-298 on UGT1A expression. Correlation study was conducted in human livers. Results: miR-298 overexpression reduced mRNA level of UGT1A1 and UGT1A4 in HepG2 and LS174T cells, and that of UGT1A3 and UGT1A9 in LS174T cells. miR-298 repression increased mRNA level of UGT1A4 in HepG2 and LS174T cells, and that of UGT1A1 and UGT1A3 in LS174T cells. Inverse correlations between miR-298, as well as miR-491–3p, and UGT1A3 and 1A4 mRNA levels were observed in livers. Conclusion: The study demonstrates that miR-298 and miR-491–3p downregulates UGT1A expression.
Collapse
Affiliation(s)
- Pei Wang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Ya-li Nie
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Shu-jie Wang
- Department of Pharmacology, School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Lin-lin Yang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Wei-hong Yang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Jiang-feng Li
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Xiao-tian Li
- Department of Pharmacology, School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Li-rong Zhang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
59
|
Rouleau M, Tourancheau A, Girard-Bock C, Villeneuve L, Vaucher J, Duperré AM, Audet-Delage Y, Gilbert I, Popa I, Droit A, Guillemette C. Divergent Expression and Metabolic Functions of Human Glucuronosyltransferases through Alternative Splicing. Cell Rep 2017; 17:114-124. [PMID: 27681425 DOI: 10.1016/j.celrep.2016.08.077] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/15/2016] [Accepted: 08/23/2016] [Indexed: 11/17/2022] Open
Abstract
Maintenance of cellular homeostasis and xenobiotic detoxification is mediated by 19 human UDP-glucuronosyltransferase enzymes (UGTs) encoded by ten genes that comprise the glucuronidation pathway. Deep RNA sequencing of major metabolic organs exposes a substantial expansion of the UGT transcriptome by alternative splicing, with variants representing 20% to 60% of canonical transcript expression. Nearly a fifth of expressed variants comprise in-frame sequences that may create distinct structural and functional features. Follow-up cell-based assays reveal biological functions for these alternative UGT proteins. Some isoforms were found to inhibit or induce inactivation of drugs and steroids in addition to perturbing global cell metabolism (energy, amino acids, nucleotides), cell adhesion, and proliferation. This work highlights the biological relevance of alternative UGT expression, which we propose increases protein diversity through the evolution of metabolic regulators from specific enzymes.
Collapse
Affiliation(s)
- Michèle Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada
| | - Alan Tourancheau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada
| | - Camille Girard-Bock
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada
| | - Lyne Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada
| | - Jonathan Vaucher
- Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
| | - Anne-Marie Duperré
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada
| | - Yannick Audet-Delage
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada
| | - Isabelle Gilbert
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada
| | - Ion Popa
- Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
| | - Arnaud Droit
- Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
60
|
Hirvensalo P, Tornio A, Neuvonen M, Tapaninen T, Paile-Hyvärinen M, Kärjä V, Männistö VT, Pihlajamäki J, Backman JT, Niemi M. Comprehensive Pharmacogenomic Study Reveals an Important Role of UGT1A3 in Montelukast Pharmacokinetics. Clin Pharmacol Ther 2017; 104:158-168. [PMID: 28940478 PMCID: PMC6033076 DOI: 10.1002/cpt.891] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/01/2017] [Accepted: 09/19/2017] [Indexed: 12/17/2022]
Abstract
To identify the genetic basis of interindividual variability in montelukast exposure, we determined its pharmacokinetics and sequenced 379 pharmacokinetic genes in 191 healthy volunteers. An intronic single nucleotide variation (SNV), strongly linked with UGT1A3*2, associated with reduced area under the plasma concentration–time curve (AUC0‐∞) of montelukast (by 18% per copy of the minor allele; P = 1.83 × 10−10). UGT1A3*2 was associated with increased AUC0‐∞ of montelukast acyl‐glucuronide M1 and decreased AUC0‐∞ of hydroxymetabolites M5R, M5S, and M6 (P < 10−9). Furthermore, SNVs in SLCO1B1 and ABCC9 were associated with the AUC0‐∞ of M1 and M5R, respectively. In addition, a candidate gene analysis suggested that CYP2C8 and ABCC9 SNVs also affect the AUC0‐∞ of montelukast. The found UGT1A3 and ABCC9 variants associated with increased expression of the respective genes in human liver samples. Montelukast and its hydroxymetabolites were glucuronidated by UGT1A3 in vitro. These results indicate that UGT1A3 plays an important role in montelukast pharmacokinetics, especially in UGT1A3*2 carriers.
Collapse
Affiliation(s)
- Päivi Hirvensalo
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuija Tapaninen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Maria Paile-Hyvärinen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Vesa Kärjä
- Department of Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Ville T Männistö
- Department of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jussi Pihlajamäki
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Clinical Nutrition and Obesity Center, Kuopio University Hospital, Kuopio, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
61
|
Labriet A, De Mattia E, Cecchin E, Lévesque É, Jonker D, Couture F, Buonadonna A, D'Andrea M, Villeneuve L, Toffoli G, Guillemette C. Improved Progression-Free Survival in Irinotecan-Treated Metastatic Colorectal Cancer Patients Carrying the HNF1A Coding Variant p.I27L. Front Pharmacol 2017; 8:712. [PMID: 29066969 PMCID: PMC5641335 DOI: 10.3389/fphar.2017.00712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/22/2017] [Indexed: 01/15/2023] Open
Abstract
Hepatocyte nuclear factor 1-alpha (HNF1A) is a liver-enriched transcription factor that plays a key role in many aspects of hepatic functions including detoxification processes. We examined whether HNF1A polymorphisms are associated with clinical outcomes in two independent cohorts combining 417 European ancestry patients with metastatic colorectal cancer (mCRC) treated with irinotecan-based chemotherapy. The intronic rs2244608A>G marker was predictive of an improved progression-free survival with a trend in the Canadian cohort and reaching significance in the Italian cohort, with hazard ratios (HR) of 0.74 and 0.72, P = 0.076 and 0.038, respectively. A strong association between rs2244608A>G and improved PFS was found in the combined analysis of both cohorts (HR = 0.72; P = 0.002). Consistent with an altered HNF1A function, mCRC carriers of the rs2244608G minor allele displayed enhanced drug exposure by 45% (P = 0.032) compared to non-carriers. In Caucasians, rs2244608A>G is in strong linkage with the coding variant rs1169288c.79A>C (HNF1A p.I27L). In healthy donors, we observed an altered hepatic (ABCC1, P = 0.009, ABCC2, P = 0.048 and CYP3A5, P = 0.001; n = 89) and intestinal (TOP1, P = 0.004; n = 75) gene expression associated with the rs1169288C allele. In addition, the rs1169288C polymorphism could significantly increase the ABCC1 promoter activity by 27% (P = 0.008) and 15% (P = 0.041) in the human kidney HEK293 and the human liver HepG2 cell lines, respectively. Our findings suggest that the HNF1A rs2244608, or the tightly linked functional coding variant p.I27L, might be a potential prognostic marker with irinotecan-based regimens.
Collapse
Affiliation(s)
- Adrien Labriet
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Elena De Mattia
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Éric Lévesque
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Derek Jonker
- Division of Medical Oncology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Félix Couture
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Angela Buonadonna
- Medical Oncology Unit, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Mario D'Andrea
- Medical Oncology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Lyne Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| |
Collapse
|
62
|
Hu M, Wu B, Liu Z. Bioavailability of Polyphenols and Flavonoids in the Era of Precision Medicine. Mol Pharm 2017; 14:2861-2863. [DOI: 10.1021/acs.molpharmaceut.7b00545] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ming Hu
- Department of Pharmacological
and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas 77030, United States
| | - Baojian Wu
- College of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Zhongqiu Liu
- International Institute for
Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P. R. China
| |
Collapse
|
63
|
Zhou X, Zheng Z, Xu C, Wang J, Min M, Zhao Y, Wang X, Gong Y, Yin J, Guo M, Guo D, Zheng J, Zhang B, Yin X. Disturbance of Mammary UDP-Glucuronosyltransferase Represses Estrogen Metabolism and Exacerbates Experimental Breast Cancer. J Pharm Sci 2017; 106:2152-2162. [PMID: 28479355 DOI: 10.1016/j.xphs.2017.04.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/20/2017] [Accepted: 04/26/2017] [Indexed: 02/01/2023]
Abstract
The progression of breast cancer is closely related to the levels of estrogens within the body. UDP-glucuronosyltransferase (UGT) is an important class of phase II metabolizing enzymes, playing a pivotal role in detoxifying steroid hormone. In the present study, we aim at uncovering the potential dysregulation pattern of UGT and its role in estrogen metabolism and in the pathogenesis of breast cancer. Female Sprague-Dawley rats were treated with 100 mg/kg dimethylbenz(a)anthracene (DMBA) to induce breast cancer. Our results showed that the expression and activity of UGT in mammary tissues were downregulated significantly in DMBA rats. Consistent with this, levels of estradiol, 4-hydroxylated estradiol, and 2-hydroxylated estradiol were increased in both mammary tissues and serum, supporting a notable accumulation of toxic estrogen species in the target tissue of breast cancer. In addition, we also observed the decreased cell migration, cell proliferation, and DNA damage in UGT-transfected MCF-7 cells, suggesting a protective role of UGT against estrogen-induced mammary carcinogenesis. Taken together, these results indicated that accumulation of estrogens induced by UGT deficiency is a critical factor to induce the development of breast cancer. UGT contributes to estrogen elimination, and its glucuronidation capacity influences the estrogen signaling pathway and the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Xueyan Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Ziqiang Zheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Chang Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Juan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Mengjun Min
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yun Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xi Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yinhan Gong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Jiale Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Meng Guo
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Bei Zhang
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China.
| |
Collapse
|
64
|
Wijayakumara DD, Mackenzie PI, McKinnon RA, Hu DG, Meech R. Regulation of UDP-Glucuronosyltransferases UGT2B4 and UGT2B7 by MicroRNAs in Liver Cancer Cells. J Pharmacol Exp Ther 2017; 361:386-397. [DOI: 10.1124/jpet.116.239707] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/21/2017] [Indexed: 01/30/2023] Open
|
65
|
Lu D, Wang S, Xie Q, Guo L, Wu B. Transcriptional Regulation of Human UDP-Glucuronosyltransferase 2B10 by Farnesoid X Receptor in Human Hepatoma HepG2 Cells. Mol Pharm 2017; 14:2899-2907. [PMID: 28267333 DOI: 10.1021/acs.molpharmaceut.6b01103] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Little is known about transcriptional regulators of UDP-glucuronosyltransferase 2B10 (UGT2B10), an enzyme known to glucuronidate many chemicals and drugs such as nicotine and tricyclic antidepressants. Here, we uncovered that UGT2B10 was transcriptionally regulated by farnesoid X receptor (FXR), the bile acid sensing nuclear receptor. GW4064 and chenodeoxycholic acid (two specific FXR agonists) treatment of HepG2 cells led to a significant increase in the mRNA level of UGT2B10. The treated cells also showed enhanced glucuronidation activities toward amitriptyline (an UGT2B10 probe substrate). In reporter gene assays, the extent of UGT2B10 activation by the FXR agonists was positively correlated with the amount of cotransfected FXR. Consistently, knockdown of FXR by shRNA attenuated the induction effect on UGT2B10 expression. Furthermore, a combination of electrophoretic mobility shift assay and chromatin immunoprecipitation showed that the FXR receptor trans-activated UGT2B10 through its specific binding to the -209- to -197-bp region (an IR1 element) of the UGT2B10 promoter. In summary, our results for the first time established FXR as a transcriptional regulator of human UGT2B10.
Collapse
Affiliation(s)
- Danyi Lu
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Shuai Wang
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Qian Xie
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Lianxia Guo
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Baojian Wu
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
66
|
Scotcher D, Billington S, Brown J, Jones CR, Brown CDA, Rostami-Hodjegan A, Galetin A. Microsomal and Cytosolic Scaling Factors in Dog and Human Kidney Cortex and Application for In Vitro-In Vivo Extrapolation of Renal Metabolic Clearance. Drug Metab Dispos 2017; 45:556-568. [PMID: 28270564 PMCID: PMC5399648 DOI: 10.1124/dmd.117.075242] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 02/27/2017] [Indexed: 12/17/2022] Open
Abstract
In vitro-in vivo extrapolation of drug metabolism data obtained in enriched preparations of subcellular fractions rely on robust estimates of physiologically relevant scaling factors for the prediction of clearance in vivo. The purpose of the current study was to measure the microsomal and cytosolic protein per gram of kidney (MPPGK and CPPGK) in dog and human kidney cortex using appropriate protein recovery marker and evaluate functional activity of human cortex microsomes. Cytochrome P450 (CYP) content and glucose-6-phosphatase (G6Pase) activity were used as microsomal protein markers, whereas glutathione-S-transferase activity was a cytosolic marker. Functional activity of human microsomal samples was assessed by measuring mycophenolic acid glucuronidation. MPPGK was 33.9 and 44.0 mg/g in dog kidney cortex, and 41.1 and 63.6 mg/g in dog liver (n = 17), using P450 content and G6Pase activity, respectively. No trends were noted between kidney, liver, and intestinal scalars from the same animals. Species differences were evident, as human MPPGK and CPPGK were 26.2 and 53.3 mg/g in kidney cortex (n = 38), respectively. MPPGK was 2-fold greater than the commonly used in vitro-in vivo extrapolation scalar; this difference was attributed mainly to tissue source (mixed kidney regions versus cortex). Robust human MPPGK and CPPGK scalars were measured for the first time. The work emphasized the importance of regional differences (cortex versus whole kidney–specific MPPGK, tissue weight, and blood flow) and a need to account for these to improve assessment of renal metabolic clearance and its extrapolation to in vivo.
Collapse
Affiliation(s)
- Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester (D.S., A.R.-H., A.G.); Newcastle University, Newcastle (S.B., C.D.A.B.); Biobank, Central Manchester University Hospitals NHS Foundation Trust, Manchester (J.B.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield (A.R.-H.), United Kingdom
| | - Sarah Billington
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester (D.S., A.R.-H., A.G.); Newcastle University, Newcastle (S.B., C.D.A.B.); Biobank, Central Manchester University Hospitals NHS Foundation Trust, Manchester (J.B.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield (A.R.-H.), United Kingdom
| | - Jay Brown
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester (D.S., A.R.-H., A.G.); Newcastle University, Newcastle (S.B., C.D.A.B.); Biobank, Central Manchester University Hospitals NHS Foundation Trust, Manchester (J.B.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield (A.R.-H.), United Kingdom
| | - Christopher R Jones
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester (D.S., A.R.-H., A.G.); Newcastle University, Newcastle (S.B., C.D.A.B.); Biobank, Central Manchester University Hospitals NHS Foundation Trust, Manchester (J.B.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield (A.R.-H.), United Kingdom
| | - Colin D A Brown
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester (D.S., A.R.-H., A.G.); Newcastle University, Newcastle (S.B., C.D.A.B.); Biobank, Central Manchester University Hospitals NHS Foundation Trust, Manchester (J.B.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield (A.R.-H.), United Kingdom
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester (D.S., A.R.-H., A.G.); Newcastle University, Newcastle (S.B., C.D.A.B.); Biobank, Central Manchester University Hospitals NHS Foundation Trust, Manchester (J.B.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield (A.R.-H.), United Kingdom
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester (D.S., A.R.-H., A.G.); Newcastle University, Newcastle (S.B., C.D.A.B.); Biobank, Central Manchester University Hospitals NHS Foundation Trust, Manchester (J.B.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield (A.R.-H.), United Kingdom
| |
Collapse
|
67
|
Rouleau M, Audet-Delage Y, Desjardins S, Rouleau M, Girard-Bock C, Guillemette C. Endogenous Protein Interactome of Human UDP-Glucuronosyltransferases Exposed by Untargeted Proteomics. Front Pharmacol 2017; 8:23. [PMID: 28217095 PMCID: PMC5290407 DOI: 10.3389/fphar.2017.00023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/12/2017] [Indexed: 01/19/2023] Open
Abstract
The conjugative metabolism mediated by UDP-glucuronosyltransferase enzymes (UGTs) significantly influences the bioavailability and biological responses of endogenous molecule substrates and xenobiotics including drugs. UGTs participate in the regulation of cellular homeostasis by limiting stress induced by toxic molecules, and by controlling hormonal signaling networks. Glucuronidation is highly regulated at genomic, transcriptional, post-transcriptional and post-translational levels. However, the UGT protein interaction network, which is likely to influence glucuronidation, has received little attention. We investigated the endogenous protein interactome of human UGT1A enzymes in main drug metabolizing non-malignant tissues where UGT expression is most prevalent, using an unbiased proteomics approach. Mass spectrometry analysis of affinity-purified UGT1A enzymes and associated protein complexes in liver, kidney and intestine tissues revealed an intricate interactome linking UGT1A enzymes to multiple metabolic pathways. Several proteins of pharmacological importance such as transferases (including UGT2 enzymes), transporters and dehydrogenases were identified, upholding a potential coordinated cellular response to small lipophilic molecules and drugs. Furthermore, a significant cluster of functionally related enzymes involved in fatty acid β-oxidation, as well as in the glycolysis and glycogenolysis pathways were enriched in UGT1A enzymes complexes. Several partnerships were confirmed by co-immunoprecipitations and co-localization by confocal microscopy. An enhanced accumulation of lipid droplets in a kidney cell model overexpressing the UGT1A9 enzyme supported the presence of a functional interplay. Our work provides unprecedented evidence for a functional interaction between glucuronidation and bioenergetic metabolism.
Collapse
Affiliation(s)
- Michèle Rouleau
- Pharmacogenomics Laboratory, Canada Research Chair in Pharmacogenomics, Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center, Laval University Québec, QC, Canada
| | - Yannick Audet-Delage
- Pharmacogenomics Laboratory, Canada Research Chair in Pharmacogenomics, Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center, Laval University Québec, QC, Canada
| | - Sylvie Desjardins
- Pharmacogenomics Laboratory, Canada Research Chair in Pharmacogenomics, Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center, Laval University Québec, QC, Canada
| | - Mélanie Rouleau
- Pharmacogenomics Laboratory, Canada Research Chair in Pharmacogenomics, Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center, Laval University Québec, QC, Canada
| | - Camille Girard-Bock
- Pharmacogenomics Laboratory, Canada Research Chair in Pharmacogenomics, Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center, Laval University Québec, QC, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Canada Research Chair in Pharmacogenomics, Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center, Laval University Québec, QC, Canada
| |
Collapse
|
68
|
Advances in drug metabolism and pharmacogenetics research in Australia. Pharmacol Res 2017; 116:7-19. [DOI: 10.1016/j.phrs.2016.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 01/04/2023]
|
69
|
Bock KW. From dioxin toxicity to putative physiologic functions of the human Ah receptor in homeostasis of stem/progenitor cells. Biochem Pharmacol 2017; 123:1-7. [DOI: 10.1016/j.bcp.2016.06.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/23/2016] [Indexed: 01/16/2023]
|
70
|
Neumann E, Mehboob H, Ramírez J, Mirkov S, Zhang M, Liu W. Age-Dependent Hepatic UDP-Glucuronosyltransferase Gene Expression and Activity in Children. Front Pharmacol 2016; 7:437. [PMID: 27899892 PMCID: PMC5110524 DOI: 10.3389/fphar.2016.00437] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 11/01/2016] [Indexed: 11/22/2022] Open
Abstract
UDP-glucuronosyltransferases (UGTs) are important phase II drug metabolism enzymes. The aim of this study was to explore the relationship between age and changes in mRNA expression and activity of major human hepatic UGTs, as well as to understand the potential regulatory mechanism underlying this relationship. Using previously generated data, we investigated age-dependent mRNA expression levels of 11 hepatic UGTs (UGT1A1, UGT1A3, UGT1A4, UGT1A5, UGT1A6, UGT1A9, UGT2B4, UGT2B7, UGT2B10, UGT2B15, and UGT2B17) and 16 transcription factors (AHR, AR, CAR, ESR2, FXR, GCCR, HNF1a, HNF3a, HNF3b, HNF4a, PPARA, PPARG, PPARGC, PXR, SP1, and STAT3) in liver tissue of donors (n = 38) ranging from 0 to 25 years of age. We also examined the correlation between age and microsomal activities using 14 known UGT drug substrates in the liver samples (n = 19) of children donors. We found a statistically significant increase (nominal p < 0.05) in the expression of UGT1A1, UGT1A3, UGT1A4, UGT1A5, UGT1A6, UGT2B7, and UGT2B17, as well as glucuronidation activities of serotonin, testosterone, and vorinostat during the first 25 years of life. Expression of estrogen receptor 1 and pregnane X receptor, two strong UGT transcriptional regulators, were significantly correlated with both age and UGT mRNA expression (p ≤ 0.05). These results suggest that both UGT expression and activity increase during childhood and adolescence, possibly driven in part by hormonal signaling. Our findings may help explain inter-patient variability in response to medications among children.
Collapse
Affiliation(s)
- Elizabeth Neumann
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University West Lafayette, IN, USA
| | - Huma Mehboob
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue UniversityWest Lafayette, IN, USA; Department of Biochemistry, University of AgricultureFaisalabad, Pakistan
| | - Jacqueline Ramírez
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago Chicago, IL, USA
| | - Snezana Mirkov
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago Chicago, IL, USA
| | - Min Zhang
- Department of Statistics, College of Science, Purdue UniversityWest Lafayette, IN, USA; Beijing Institute for Brain Disorders, Capital Medical UniversityBeijing, China
| | - Wanqing Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University West Lafayette, IN, USA
| |
Collapse
|
71
|
Hepatic expression of transcription factors affecting developmental regulation of UGT1A1 in the Han Chinese population. Eur J Clin Pharmacol 2016; 73:29-37. [PMID: 27704169 DOI: 10.1007/s00228-016-2137-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/21/2016] [Indexed: 02/08/2023]
Abstract
PURPOSE Complete or partial inactivity of UGT1A1, the unique enzyme responsible for bilirubin glucuronidation, is commonly associated with hyperbilirubinemia. We investigated the dynamic expression of UGT1A1, and that of the transcription factors (TFs) involved in its developmental regulation, during human hepatic growth in Han Chinese individuals. METHODS Eighty-eight prenatal, pediatric, and adult liver samples were obtained from Han Chinese individuals. Quantitative real-time polymerase chain reaction was used to evaluate mRNA expression of UGT1A1 and TFs including PXR, CAR, HNF1A, HNF4A, PPARA, etc. UGT1A1 protein levels and metabolic activity were determined by western blotting and high-performance liquid chromatography. Direct sequencing was employed to genotype UGT1A1*6 (211G˃A) and UGT1A1*28 (TA6˃TA7) polymorphisms. RESULTS UGT1A1 expression was minimal in prenatal samples, but significantly elevated during pediatric and adult stages. mRNA and protein levels and metabolic activity were prominently increased (120-, 20-, and 10-fold, respectively) in pediatric and adult livers compared to prenatal samples. Furthermore, expression did not differ appreciably between pediatric and adult periods. Dynamic expression of TFs, including PXR, CAR, HNF1A, HNF4A, and PPARA, was consistent with UGT1A1 levels at each developmental stage. A pronounced correlation between expression of these TFs and that of UGT1A1 (P < 0.001) was observed. Moreover, UGT1A1*6 and UGT1A1*28 polymorphisms reduced levels of UGT1A1 by up to 40-60 %. CONCLUSIONS Hepatic expression of transcription factors is associated with developmental regulation of UGT1A1 in the Han Chinese population. Moreover, UGT1A1 polymorphisms are associated with reduced expression of UGT1A1 mRNA and protein, as well as enzyme activity.
Collapse
|
72
|
Vrzal R, Illes P, Dvorak Z. Transplant drugs affect the expression of phase II and antioxidant enzymes in human carcinoma cells HepG2 but not in primary cultures of human hepatocytes: In vitro comparative study. Pharmacol Rep 2016; 68:1008-14. [DOI: 10.1016/j.pharep.2016.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/02/2016] [Accepted: 06/02/2016] [Indexed: 10/21/2022]
|
73
|
Knights KM, Spencer SM, Fallon JK, Chau N, Smith PC, Miners JO. Scaling factors for the in vitro-in vivo extrapolation (IV-IVE) of renal drug and xenobiotic glucuronidation clearance. Br J Clin Pharmacol 2016; 81:1153-64. [PMID: 26808419 PMCID: PMC4876189 DOI: 10.1111/bcp.12889] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/21/2022] Open
Abstract
AIM To determine the scaling factors required for inclusion of renal drug glucuronidation clearance in the prediction of total clearance via glucuronidation (CLUGT ). METHODS Microsomal protein per gram of kidney (MPPGK) was determined for human 'mixed' kidney (n = 5) microsomes (MKM). The glucuronidation activities of deferiprone (DEF), propofol (PRO) and zidovudine (AZT) by MKM and paired cortical (KCM) and medullary (KMM) microsomes were measured, along with the UGT 1A6, 1A9 and 2B7 protein contents of each enzyme source. Unbound intrinsic clearances (CLint,u,UGT ) for PRO and morphine (MOR; 3- and 6-) glucuronidation by MKM, human liver microsomes (HLM) and recombinant UGT1A9 and 2B7 were additionally determined. Data were scaled using in vitro-in vivo extrapolation (IV-IVE) approaches to assess the influence of renal CLint,u,UGT on the prediction accuracy of the calculated CLUGT values of PRO and MOR. RESULTS MPPGK was 9.3 ± 2.0 mg g(-1) (mean ± SD). The respective rates of DEF (UGT1A6), PRO (UGT1A9) and AZT (UGT2B7) glucuronidation by KCM were 1.4-, 5.2- and 10.5-fold higher than those for KMM. UGT 1A6, 1A9 and 2B7 were the only enzymes expressed in kidney. Consistent with the activity data, the abundance of each of these enzymes was greater in KCM than in KMM. The abundance of UGT1A9 in MKM (61.3 pmol mg(-1) ) was 2.7 fold higher than that reported for HLM. CONCLUSIONS Scaled renal PRO glucuronidation CLint,u,UGT was double that of liver. Renal CLint,u,UGT should be accounted for in the IV-IVE of UGT1A9 and considered for UGT1A6 and 2B7 substrates.
Collapse
Affiliation(s)
- Kathleen M. Knights
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, School of MedicineFlinders UniversityAdelaideSouth AustraliaAustralia5001
| | - Shane M. Spencer
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, School of MedicineFlinders UniversityAdelaideSouth AustraliaAustralia5001
| | - John K. Fallon
- Division of Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth Carolina27599USA
| | - Nuy Chau
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, School of MedicineFlinders UniversityAdelaideSouth AustraliaAustralia5001
| | - Philip C. Smith
- Division of Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth Carolina27599USA
| | - John O. Miners
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, School of MedicineFlinders UniversityAdelaideSouth AustraliaAustralia5001
| |
Collapse
|
74
|
Hu DG, Mackenzie PI, McKinnon RA, Meech R. Genetic polymorphisms of human UDP-glucuronosyltransferase (UGT) genes and cancer risk. Drug Metab Rev 2016; 48:47-69. [DOI: 10.3109/03602532.2015.1131292] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
75
|
Abstract
Nucleotide changes in gene regulatory elements can have a major effect on interindividual differences in drug response. For example, by reviewing all published pharmacogenomic genome-wide association studies, we show here that 96.4% of the associated single nucleotide polymorphisms reside in noncoding regions. We discuss how sequencing technologies are improving our ability to identify drug response-associated regulatory elements genome-wide and to annotate nucleotide variants within them. We highlight specific examples of how nucleotide changes in these elements can affect drug response and illustrate the techniques used to find them and functionally characterize them. Finally, we also discuss challenges in the field of drug-responsive regulatory elements that need to be considered in order to translate these findings into the clinic.
Collapse
Affiliation(s)
- Marcelo R Luizon
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
76
|
Wijayakumara DD, Hu DG, Meech R, McKinnon RA, Mackenzie PI. Regulation of Human UGT2B15 and UGT2B17 by miR-376c in Prostate Cancer Cell Lines. J Pharmacol Exp Ther 2015; 354:417-25. [DOI: 10.1124/jpet.115.226118] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/09/2015] [Indexed: 11/22/2022] Open
|
77
|
Margaillan G, Rouleau M, Klein K, Fallon JK, Caron P, Villeneuve L, Smith PC, Zanger UM, Guillemette C. Multiplexed Targeted Quantitative Proteomics Predicts Hepatic Glucuronidation Potential. Drug Metab Dispos 2015; 43:1331-5. [PMID: 26076694 DOI: 10.1124/dmd.115.065391] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 06/15/2015] [Indexed: 11/22/2022] Open
Abstract
Phase II metabolism is prominently governed by UDP-glucuronosyltransferases (UGTs) in humans. These enzymes regulate the bioactivity of many drugs and endogenous small molecules in many organs, including the liver, a major site of regulation by the glucuronidation pathway. This study determined the expression of hepatic UGTs by targeted proteomics in 48 liver samples and by measuring the glucuronidation activity using probe substrates. It demonstrates the sensitivity and accuracy of nano-ultra-performance liquid chromatography with tandem mass spectrometry to establish the complex expression profiles of 14 hepatic UGTs in a single analysis. UGT2B7 is the most abundant UGT in our collection of livers, expressed at 69 pmol/mg microsomal proteins, whereas UGT1A1, UGT1A4, UGT2B4, and UGT2B15 are similarly abundant, averaging 30-34 pmol/mg proteins. The average relative abundance of these five UGTs represents 81% of the measured hepatic UGTs. Our data further highlight the strong relationships in the expression of several UGTs. Most notably, UGT1A4 correlates with most measured UGTs, and the expression levels of UGT2B4/UGT2B7 displayed the strongest correlation. However, significant interindividual variability is observed for all UGTs, both at the level of enzyme concentrations and activity (coefficient of variation: 45%-184%). The reliability of targeted proteomics quantification is supported by the high correlation between UGT concentration and activity. Collectively, these findings expand our understanding of hepatic UGT profiles by establishing absolute hepatic concentrations of 14 UGTs and further suggest coregulated expression between most abundant hepatic UGTs. Data support the value of multiplexed targeted quantitative proteomics to accurately assess specific UGT concentrations in liver samples and hepatic glucuronidation potential.
Collapse
Affiliation(s)
- Guillaume Margaillan
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| | - Michèle Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| | - Kathrin Klein
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| | - John K Fallon
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| | - Patrick Caron
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| | - Lyne Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| | - Philip C Smith
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| | - Ulrich M Zanger
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec and Faculty of Pharmacy, Université Laval, Québec, Canada (G.M., M.R., P.C., L.V., C.G.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); Canada Research Chair in Pharmacogenomics (C.G.)
| |
Collapse
|
78
|
Bock KW. Roles of human UDP-glucuronosyltransferases in clearance and homeostasis of endogenous substrates, and functional implications. Biochem Pharmacol 2015; 96:77-82. [PMID: 25937523 DOI: 10.1016/j.bcp.2015.04.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 04/24/2015] [Indexed: 11/19/2022]
Abstract
Human UDP-glucuronosyltransferases (UGTs) are major phase II enzymes in the drug metabolism system. Despite major advances in characterization of UGT gene family members, their role in clearance and homeostasis of endogenous substrates is insufficiently understood. Endobiotic substrates including bilirubin, serotonin, eicosanoids, steroid hormones, bile acids, thyroxine and fat-soluble vitamins A and D are discussed. Species- and tissue/cell-dependent regulation of UGT expression by ligand-activated transcription factors is often involved in endobiotic homeostasis. However, roles of particular UGTs are often difficult to delineate since they function together with other enzymes and transporters. Better knowledge of endobiotic UGT substrates and consequences of their conjugation may help to understand evolutionary conserved UGT functions.
Collapse
Affiliation(s)
- Karl Walter Bock
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology. University of Tübingen, Wilhelmstrasse 56, D-72074 Tübingen, Germany.
| |
Collapse
|
79
|
Riches Z, Collier AC. Posttranscriptional regulation of uridine diphosphate glucuronosyltransferases. Expert Opin Drug Metab Toxicol 2015; 11:949-65. [PMID: 25797307 DOI: 10.1517/17425255.2015.1028355] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The uridine diphosphate (UDP)-glucuronosyltransferase (UGT) superfamily of enzymes (EC 2.4.1.17) conjugates glucuronic acid to an aglycone substrate to make them more polar and readily excreted. In general, this reaction terminates the activities of chemicals, drugs and toxins, although occasionally a more active or toxic species is produced. AREAS COVERED In addition to their well-known transcriptional responsiveness, UGTs are also regulated by posttranscriptional mechanisms. Here, the authors review these mechanisms, including latency, modulation of co-substrate accessibility and binding, dimerization and oligomerization, protein-protein interactions, allosteric inhibition and activation, posttranslational structural and functional modifications and developmental switching for UGTs. EXPERT OPINION Posttranscriptional regulation of UGTs has traditionally received less attention than nuclear regulation, in part because mechanisms involving ribosomes and endoplasmic reticula are challenging to investigate. Most promising of the posttranscriptional mechanisms reviewed are likely to be effects on co-substrate (UDP-glucuronic acid) transport and availability and structure-function changes to UGT proteins through, for example, glycosylation and phosphorylation. Although classical biochemistry continues to illuminate many aspects of UGT function, advances in proteomics and structural biology are beginning to assist in the determination of posttranscriptional regulation mechanisms for UGTs.
Collapse
Affiliation(s)
- Zoe Riches
- University of British Columbia, Faculty of Pharmaceutical Sciences , 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3 , Canada +1 604 827 2380 ;
| | | |
Collapse
|
80
|
Hu DG, Mackenzie PI, Lu L, Meech R, McKinnon RA. Induction of Human UDP-Glucuronosyltransferase 2B7 Gene Expression by Cytotoxic Anticancer Drugs in Liver Cancer HepG2 Cells. Drug Metab Dispos 2015; 43:660-8. [DOI: 10.1124/dmd.114.062380] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|