1
|
Tagawa K, Maruo Y. Dolutegravir metabolism: impact of genetic variations on uridine diphosphate glucuronosyltransferase subfamilies. Xenobiotica 2025:1-8. [PMID: 39815789 DOI: 10.1080/00498254.2025.2453983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
Dolutegravir (DTG) is a key drug used to treat human immunodeficiency virus type-1 (HIV-1) infections. Adverse events (AEs) of DTG treatment, including headache, anxiety, depression, insomnia, and abnormal dreams, are influenced by sex, body weight, age, and serum bilirubin levels. DTG is mainly metabolised by members of the uridine diphosphate glucuronosyltransferase 1A subfamilies (UGT1As), especially UGT1A1.Some studies suggest a relationship between UGT1A1 variants and AEs. The aim of this study was to identify UGT1A isoforms that exhibit DTG glucuronidation activity and determine the relationship between UGT1A variants and DTG glucuronidation in vitro.UGT1A1, UGT1A3, UGT1A9, and UGT1A10 exhibited DTG glucuronidation activity, of which UGT1A1 was the most active. Furthermore, variants of these isoforms showed decreased DTG glucuronidation activity. The different variants of UGT1As, such as UGT1A1.6, UGT1A1.7, UGT1A1.35, UGT1A1.63, UGT1A3.5, UGT1A9.2, UGT1A10M59I, and UGT1A10T202I, showed reduced glucuronidation activity towards DTG in comparison with the wild-type UGT1As.This study elucidates the relationship between UGT1A variants and the levels of glucuronidation associated with each variant.Checking for UGT1As may be helpful in predicting potential toxicities and adverse effects related to DTG treatment.
Collapse
Affiliation(s)
- Kouji Tagawa
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
- Department of Pediatrics, Sapporo Tokushukai Hospital, Hokkaido, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
2
|
Bennani Y, Ben Hassine K, Gencaslan M, Boudal‐Khoshbeen M, Samer C, Ansari M, Daali Y, Satyanarayana Uppugunduri CR. In vitro screening of UGT2B10 in silico prioritized putative ligands from drugs used in the pediatric hematopoietic stem cell transplantation setting. Pharmacol Res Perspect 2024; 12:e70011. [PMID: 39611692 PMCID: PMC11605732 DOI: 10.1002/prp2.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 11/30/2024] Open
Abstract
UGT2B10 is a phase II drug metabolizing enzyme with limited information on its role in the metabolism of drugs, especially in the pediatric hematopoietic stem cell transplantation setting. Previously, we investigated UGT2B10's role through in silico analyses and prioritized acetaminophen (APAP), lorazepam (LOR), mycophenolic acid (MPA), and voriconazole N-oxide (VCZ N-oxide) for in vitro investigations. In this report, we present in vitro screening of these candidates and of voriconazole (VCZ) to assess their potential to be substrates and/or inhibitors of UGT2B10. Enzyme kinetics experiments included recombinant UGT2B10 and analytical methods based on ultra high-performance liquid chromatography coupled to mass spectrometry (UHPLC-MS). To determine potential substrates, candidates were incubated at various therapeutically observed concentrations with recombinant UGT2B10 to identify the corresponding glucuronide metabolite. Inhibition capacity was tested using the selective probe cotinine for its glucuronidation to cotinine N-ß-d-glucuronide. IC50 was determined for compounds exhibiting inhibition. Among the tested compounds, LOR (IC50 = 0.01 μM, R2 = 0.9257) and MPA (IC50 = 0.38 mM, R2 = 0.9212) exhibited inhibition potential for UGT2B10. None of the other tested compounds featured inhibition potential and none of the compounds tested exhibited metabolism through UGT2B10. Further exploration on the clinical relevance of this inhibition using modeling strategies, overlapping nature with other UGT isoforms, and screening other molecules for their inhibition potential on UGT2B10 is warranted.
Collapse
Affiliation(s)
- Yahia Bennani
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Geneva Lausanne School of PharmacyUniversity of GenevaGenevaSwitzerland
| | - Khalil Ben Hassine
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and ObstetricsUniversity of GenevaGenevaSwitzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
| | | | - Mary Boudal‐Khoshbeen
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and ObstetricsUniversity of GenevaGenevaSwitzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
| | - Caroline Samer
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Geneva Lausanne School of PharmacyUniversity of GenevaGenevaSwitzerland
- Swiss Center for Applied Human ToxicologyGenevaSwitzerland
- Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Marc Ansari
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and ObstetricsUniversity of GenevaGenevaSwitzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Geneva Lausanne School of PharmacyUniversity of GenevaGenevaSwitzerland
- Swiss Center for Applied Human ToxicologyGenevaSwitzerland
- Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Chakradhara Rao Satyanarayana Uppugunduri
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and ObstetricsUniversity of GenevaGenevaSwitzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
- Department of Medical OncologyJawaharlal Institute of Postgraduate Medical Education and ResearchPuducherryIndia
| |
Collapse
|
3
|
Ito S, Iwata Y, Otsuka M, Kaneko Y, Ogata S, Yagi R, Uemura T, Masuda T, Saito T, Saido T, Ohtsuki S. Progressive amyloid-β accumulation in the brain leads to altered protein expressions in the liver and kidneys of APP knock-in mice. J Pharm Sci 2024; 114:103596. [PMID: 39489376 DOI: 10.1016/j.xphs.2024.10.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
Impaired hepatic and renal function influence Alzheimer's disease (AD) progression; however, whether AD progression affects these important organ functions remains unclear. Here, we investigated the impact of AD progression, characterized by brain amyloid-beta (Aβ) accumulation, on liver and kidney function of AppNL-G-F/NL-G-F (APP-KI) mice using quantitative proteomics. SWATH-based quantitative proteomics revealed changes in mitochondrial, drug metabolism, and pharmacokinetic-related proteins in mouse liver and kidneys during the early (2-month-old) and intermediate (5-month-old) stages of Aβ accumulation. Notably, in 5-month-old APP-KI mouse liver, 25 phase I/II metabolizing enzymes (8 CYPs, 7 UGTs, 7 CESs, and 3 SLCs) and five transporters (2 ABCs and 3 SLCs) were significantly altered; specifically, Ugt1a9 and Slc33a1 protein abundances increased, whereas Ugt1a1 and Abcc3 protein abundances decreased. In the kidneys, 13 phase I/II metabolizing enzymes and 10 ABC-SLC transporters were altered, including Ugt1a6, Ugt1a7, Slc22a7, and Abcb1a. Additionally, plasma proteins, such as albumin and alpha-1-acid glycoprotein, which are critical for drug binding and distribution, were also altered. These results underscore the significant role of progressive brain Aβ accumulation in modifying hepatic and renal protein abundances, potentially influencing drug metabolism and disposition in AD. Our findings provide novel insights into the complex relationship between AD progression and organ dysfunction.
Collapse
Affiliation(s)
- Shingo Ito
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| | - Yumi Iwata
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Mitsumi Otsuka
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yui Kaneko
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Seiryo Ogata
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai 980-8575, Japan
| | - Ryotaro Yagi
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Tatsuki Uemura
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Institute for Advanced Biosciences, Keio University, Nihonkoku 403-1, Daihouji, Tsuruoka 997-0017, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Takaomi Saido
- RIKEN Center for Brain Science, Laboratory for Proteolytic Neuroscience, 2-1 Hirosawa, Wako 351-0198 Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| |
Collapse
|
4
|
Tang LWT, Lapham K, Goosen TC. UGT2B10 is the Major UDP-Glucuronosyltransferase 2B Isoform Involved in the Metabolism of Lamotrigine and is Implicated in the Drug-Drug Interaction with Valproic Acid. AAPS J 2024; 26:107. [PMID: 39322784 DOI: 10.1208/s12248-024-00978-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/08/2024] [Indexed: 09/27/2024] Open
Abstract
Lamotrigine is a phenyltriazine anticonvulsant that is primarily metabolized by phase II UDP-glucuronosyltransferases (UGT) to a quaternary N2-glucuronide, which accounts for ~ 90% of the excreted dose in humans. While there is consensus that UGT1A4 plays a predominant role in the formation of the N2-glucuronide, there is compelling evidence in the literature to suggest that the metabolism of lamotrigine is catalyzed by another UGT isoform. However, the exact identity of the UGT isoform that contribute to the formation of this glucuronide remains uncertain. In this study, we harnessed a robust reaction phenotyping strategy to delineate the identities and its associated fraction metabolized (fm) of the UGTs involved in lamotrigine N2-glucuronidation. Foremost, human recombinant UGT mapping experiments revealed that the N2-glucuronide is catalyzed by multiple UGT isoforms. (i.e., UGT1A1, 1A3, 1A4, 1A9, 2B4, 2B7, and 2B10). Thereafter, scaling the apparent intrinsic clearances obtained from the enzyme kinetic experiments with our in-house liver-derived relative expression factors (REF) and relative activity factors (RAF) revealed that, in addition to UGT1A4, UGT2B10 was involved in the N2-glucuronidation of lamotrigine. This was further confirmed via chemical inhibition in human liver microsomes with the UGT1A4-selective inhibitor hecogenin and the UGT2B10-selective inhibitor desloratadine. By integrating various orthogonal approaches (i.e., REF- and RAF-scaling, and chemical inhibition), we quantitatively determined that the fm for UGT1A4 and UGT2B10 ranged from 0.42 - 0.64 and 0.32 - 0.57, respectively. Finally, we also provided nascent evidence that the pharmacokinetic interaction between lamotrigine and valproic acid likely arose from the in vivo inhibition of its UGT2B10-mediated pathway.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Pfizer Inc., 445 Eastern Point Rd, Groton, CT, 06340, USA.
| | - Kimberly Lapham
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Pfizer Inc., 445 Eastern Point Rd, Groton, CT, 06340, USA
| | - Theunis C Goosen
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Pfizer Inc., 445 Eastern Point Rd, Groton, CT, 06340, USA
| |
Collapse
|
5
|
Hu DG, Marri S, Hulin JA, Ansaar R, Mackenzie PI, McKinnon RA, Meech R. Activation of Cryptic Donor Splice Sites within the UDP-Glucuronosyltransferase (UGT)1A First-Exon Region Generates Variant Transcripts That Encode UGT1A Proteins with Truncated Aglycone-Binding Domains. Drug Metab Dispos 2024; 52:526-538. [PMID: 38565302 DOI: 10.1124/dmd.123.001565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 02/19/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
The human UDP-glucuronosyltransferases (UGTs) have crucial roles in metabolizing and clearing numerous small lipophilic compounds. The UGT1A locus generates nine UGT1A mRNAs, 65 spliced transcripts, and 34 circular RNAs. In this study, our analysis of published UGT-RNA capture sequencing (CaptureSeq) datasets identified novel splice junctions that predict 24 variant UGT1A transcripts derived from ligation of exon 2 to unique sequences within the UGT1A first-exon region using cryptic donor splice sites. Of these variants, seven (1A1_n1, 1A3_n3, 1A4_n4, 1A5_n1, 1A8_n2, 1A9_n2, 1A10_n7) are predicted to encode UGT1A proteins with truncated aglycone-binding domains. We assessed their expression profiles and deregulation in cancer using four RNA sequencing (RNA-Seq) datasets of paired normal and cancerous drug-metabolizing tissues from large patient cohorts. Variants were generally coexpressed with their canonical counterparts with a higher relative abundance in tumor than in normal tissues. Variants showed tissue-specific expression with high interindividual variability but overall low abundance. However, 1A8_n2 showed high abundance in normal and cancerous colorectal tissues, with levels that approached or surpassed canonical 1A8 mRNA levels in many samples. We cloned 1A8_n2 and showed expression of the predicted protein (1A8_i3) in human embryonic kidney (HEK)293T cells. Glucuronidation assays with 4-methylumbelliferone (4MU) showed that 1A8_i3 had no activity and was unable to inhibit the activity of 1A8_i1 protein. In summary, the activation of cryptic donor splice sites within the UGT1A first-exon region expands the UGT1A transcriptome and proteome. The 1A8_n2 cryptic donor splice site is highly active in colorectal tissues, representing an important cis-regulatory element that negatively regulates the function of the UGT1A8 gene through pre-mRNA splicing. SIGNIFICANT STATEMENT: The UGT1A locus generates nine canonical mRNAs, 65 alternately spliced transcripts, and 34 different circular RNAs. The present study reports a series of novel UDP-glucuronosyltransferase (UGT)1A variants resulting from use of cryptic donor splice sites in both normal and cancerous tissues, several of which are predicted to encode variant UGT1A proteins with truncated aglycone-binding domains. Of these, 1A8_n2 shows exceptionally high abundance in colorectal tissues, highlighting its potential role in the first-pass metabolism in gut through the glucuronidation pathway.
Collapse
Affiliation(s)
- Dong Gui Hu
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Shashikanth Marri
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Julie-Ann Hulin
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Radwan Ansaar
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Peter I Mackenzie
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Ross A McKinnon
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Robyn Meech
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| |
Collapse
|
6
|
Rouleau M, Villeneuve L, Allain EP, McCabe-Leroux J, Tremblay S, Nguyen Van Long F, Uchil A, Joly-Beauparlant C, Droit A, Guillemette C. Non-canonical transcriptional regulation of the poor prognostic factor UGT2B17 in chronic lymphocytic leukemic and normal B cells. BMC Cancer 2024; 24:410. [PMID: 38566115 PMCID: PMC10985967 DOI: 10.1186/s12885-024-12143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND High expression of the glycosyltransferase UGT2B17 represents an independent adverse prognostic marker in chronic lymphocytic leukemia (CLL). It also constitutes a predictive marker for therapeutic response and a drug resistance mechanism. The key determinants driving expression of the UGT2B17 gene in normal and leukemic B-cells remain undefined. The UGT2B17 transcriptome is complex and is comprised of at least 10 alternative transcripts, identified by previous RNA-sequencing of liver and intestine. We hypothesized that the transcriptional program regulating UGT2B17 in B-lymphocytes is distinct from the canonical expression previously characterized in the liver. RESULTS RNA-sequencing and genomics data revealed a specific genomic landscape at the UGT2B17 locus in normal and leukemic B-cells. RNA-sequencing and quantitative PCR data indicated that the UGT2B17 enzyme is solely encoded by alternative transcripts expressed in CLL patient cells and not by the canonical transcript widely expressed in the liver and intestine. Chromatin accessible regions (ATAC-Seq) in CLL cells mapped with alternative promoters and non-coding exons, which may be derived from endogenous retrotransposon elements. By luciferase reporter assays, we identified key cis-regulatory STAT3, RELA and interferon regulatory factor (IRF) binding sequences driving the expression of UGT2B17 in lymphoblastoid and leukemic B-cells. Electrophoretic mobility shift assays and pharmacological inhibition demonstrated key roles for the CLL prosurvival transcription factors STAT3 and NF-κB in the leukemic expression of UGT2B17. CONCLUSIONS UGT2B17 expression in B-CLL is driven by key regulators of CLL progression. Our data suggest that a NF-κB/STAT3/IRF/UGT2B17 axis may represent a novel B-cell pathway promoting disease progression and drug resistance.
Collapse
Affiliation(s)
- Michèle Rouleau
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Lyne Villeneuve
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Eric P Allain
- Molecular Genetics Laboratory, Vitalité Health Network, Dr. Georges-L.-Dumont University Hospital Center, Moncton, NB, Canada
| | - Jules McCabe-Leroux
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Sophie Tremblay
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Flora Nguyen Van Long
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Ashwini Uchil
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada
- Cancer research center of Université Laval, Québec, Canada
| | - Charles Joly-Beauparlant
- Cancer research center of Université Laval, Québec, Canada
- CRCHUQc-UL and Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Arnaud Droit
- Cancer research center of Université Laval, Québec, Canada
- CRCHUQc-UL and Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Chantal Guillemette
- Faculty of Pharmacy, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc- UL), Université Laval, Québec, QC, Canada.
- Cancer research center of Université Laval, Québec, Canada.
- Canada Research Chair in Pharmacogenomics, Faculty of Pharmacy, Université Laval, Québec, QC, Canada.
| |
Collapse
|
7
|
Hu DG, Marri S, Hulin JA, McKinnon RA, Mackenzie PI, Meech R. A Comprehensive Bioinformatic Analysis of RNA-seq Datasets Reveals a Differential and Variable Expression of Wildtype and Variant UGT1A Transcripts in Human Tissues and Their Deregulation in Cancers. Cancers (Basel) 2024; 16:353. [PMID: 38254842 PMCID: PMC10814044 DOI: 10.3390/cancers16020353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
The UGT1A locus generates over 60 different alternatively spliced transcripts and 30 circular RNAs. To date, v2 and v3 transcripts are the only variant UGT1A transcripts that have been functionally characterized. Both v2 and v3 transcripts encode the same inactive variant UGT1A proteins (i2s) that can negatively regulate glucuronidation activity and influence cancer cell metabolism. However, the abundance and interindividual variability in the expression of v2 and v3 transcripts in human tissues and their potential deregulation in cancers have not been comprehensively assessed. To address this knowledge gap, we quantified the expression levels of v1, v2, and v3 transcripts using RNA-seq datasets with large cohorts of normal tissues and paired normal and tumor tissues from patients with six different cancer types (liver, kidney, colon, stomach, esophagus, and bladder cancer). We found that v2 and v3 abundance varied significantly between different tissue types, and that interindividual variation was also high within the same tissue type. Moreover, the ratio of v2 to v3 variants varied between tissues, implying their differential regulation. Our results showed higher v2 abundance in gastrointestinal tissues than liver and kidney tissues, suggesting a more significant negative regulation of glucuronidation by i2 proteins in gastrointestinal tissues than in liver and kidney tissues. We further showed differential deregulation of wildtype (v1) and variant transcripts (v2, v3) in cancers that generally increased the v2/v1 and/or v3/v1 expression ratios in tumors compared to normal tissues, indicating a more significant role of the variants in tumors. Finally, we report ten novel UGT1A transcripts with novel 3' terminal exons, most of which encode variant proteins with a similar structure to UGT1A_i2 proteins. These findings further emphasize the diversity of the UGT1A transcriptome and proteome.
Collapse
Affiliation(s)
- Dong Gui Hu
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide 5042, Australia; (S.M.); (J.-A.H.); (R.A.M.); (P.I.M.); (R.M.)
| | | | | | | | | | | |
Collapse
|
8
|
Zhou L, Montalvo AD, Collins JM, Wang D. Quantitative analysis of the UDP-glucuronosyltransferase transcriptome in human tissues. Pharmacol Res Perspect 2023; 11:e01154. [PMID: 37983911 PMCID: PMC10659769 DOI: 10.1002/prp2.1154] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/22/2023] Open
Abstract
UDP-glucuronosyltransferases (UGTs) are phase II drug metabolizing enzymes that play important roles in the detoxification of endogenous and exogenous substrates. The 22 human UGTs belong to four families (UGT1, UGT2, UGT3, and UGT8) and differ in their expression, substrate specificity, UDP-sugar preference, and physiological functions. Differential expression/activity of the UGTs contributes to interperson variability in drug responses and toxicity, hormone homeostasis, and disease/cancer risks. However, in normal tissues, the tissue-specific expression profiles and transcriptional regulation of the UGTs are still not fully understood. In this study, we comprehensively analyzed the transcriptome of 22 UGTs in 54 human tissues/regions using RNAseq data from GTEx. We then validated the findings in the liver and small intestine samples using real-time PCR. Our results showed large interindividual variability across tissues in the expression of each UGT and the overall composition of UGT pools, consisting of different UGTs and their splice isoforms. Our results also revealed coexpression of the UGTs, Cytochrome P450s, and many transcription factors in the liver, suggesting potential coregulation or functional coordination. Our results provide the groundwork for future studies to detail further the regulation of the expression and activity of the UGTs.
Collapse
Affiliation(s)
- Lucas Zhou
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for PharmacogenomicsUniversity of FloridaGainesvilleFloridaUSA
| | - Abelardo D. Montalvo
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for PharmacogenomicsUniversity of FloridaGainesvilleFloridaUSA
| | - Joseph M. Collins
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for PharmacogenomicsUniversity of FloridaGainesvilleFloridaUSA
| | - Danxin Wang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for PharmacogenomicsUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
9
|
Collins JM, Wang D. A Comprehensive Evaluation of the Effects of RNA-Editing Proteins ADAR and ADARB1 on the Expression of the Drug-Metabolizing Enzymes in HepaRG Cells. Drug Metab Dispos 2023; 51:1508-1514. [PMID: 37532539 PMCID: PMC10586505 DOI: 10.1124/dmd.123.001396] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/10/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023] Open
Abstract
Two RNA-editing proteins, the adenosine deaminase acting on RNA, ADAR, and ADARB1, broadly regulate gene expression in editing-dependent and editing-independent manners. Previous studies showed that the expression of the drug-metabolizing cytochrome P450s (P450s) and UDP glucuronosyltransferases (UGTs) changes upon knockdown (KD) of ADAR or ADARB1 in different hepatic cell lines. To systematically survey the effects of these two ADARs on the expression of P450s and UGTs, we used small interfering RNA in HepaRG cells and tested the association between the expression of the P450s and ADARs in a liver sample cohort (n = 246). KD of ADAR increased the expression of the CYP3As and CYP2C9 and reduced the expression of the others, whereas KD of ADARB1 reduced the expression of nearly all genes tested. ADAR KD also suppressed the induction of most P450s, whereas ADARB1 KD had mixed effects depending on the inducer/gene combination. P450 expression was positively associated with both ADARs in liver samples, consistent with the KD results. However, after adjusting for the expression of transcription factors (TFs) known to regulate P450 expression, the associations disappeared, indicating that the effects of ADAR or ADARB1 primarily occur through TFs. Moreover, we found that the expression of normally spliced CYP3A5 transcripts is increased in both KDs, indicating a direct effect of the ADARs on promoting the usage of the cryptic splice site generated by CYP3A5*3. Taken together, our results revealed the nonoverlapping regulatory effects of ADAR and ADARB1 and supported their broad roles in controlling the expression of drug-metabolizing enzymes in the liver. SIGNIFICANCE STATEMENT: Here, this study systematically surveyed the roles of ADAR and ADARB1 in both basal and induced expression of drug-metabolizing enzymes and assessed their coexpression in liver samples. This study's results support that ADAR and ADARB1 regulate the expression of the drug-metabolizing enzymes in the liver, suggesting that factors affecting ADAR expression also have the potential to impact drug metabolism.
Collapse
Affiliation(s)
- Joseph M Collins
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for Pharmacogenomics, University of Florida, Gainesville, Florida
| | - Danxin Wang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for Pharmacogenomics, University of Florida, Gainesville, Florida
| |
Collapse
|
10
|
Tagawa K, Maruo Y, Mimura Y, Ikushiro S. Effects of common genetic variants of human uridine diphosphate glucuronosyltransferase subfamilies on irinotecan glucuronidation. Toxicol Mech Methods 2023; 33:197-205. [PMID: 35930428 DOI: 10.1080/15376516.2022.2109229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The adverse effects (diarrhea and neutropenia) of irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) are associated with genetic variants of uridine diphosphate glucuronosyltransferase 1A subfamilies (UGT1As). UGT1As are enzymes that metabolize the active form of irinotecan, 7-ethyl-10 hydroxycamptothecin (SN-38), by glucuronidation in the liver. They are widely known as predictive factors of severe adverse effects, such as neutropenia and diarrhea. Some studies have suggested that variants of UGT1As affect SN-38 glucuronidation activities, thus exerting severe adverse effects. We aimed to identify UGT1A isoforms that show SN-38 glucuronidation activity and determine the relationship between UGT1A variants and SN-38 glucuronidation in vitro. We found that UGT1A1 and UGT1A6-UGT1A10 displayed SN-38 glucuronidation activity. Among these, UGT1A1 was the most active. Furthermore, the variants of these isoforms showed decreased SN-38 glucuronidation activity. In our study, we compared the different variants of UGT1As, such as UGT1A1.6, UGT1A1.7, UGT1A1.27, UGT1A1.35, UGT1A7.3, UGT1A8.4, UGT1A10M59I, and UGT1A10T202I, to determine the differences in the reduction of glucuronidation. Our study elucidates the relationship between UGT1A variants and the level of glucuronidation associated with each variant. Therefore, testing can be done before the initiation of irinotecan treatment to predict potential toxicities and adverse effects.
Collapse
Affiliation(s)
- Kouji Tagawa
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Yu Mimura
- Department of Pediatrics, Toyosato Hospital, Shiga, Japan
| | - Shinichi Ikushiro
- Department of Biotechnology, Toyama Prefectural University, Toyama, Japan
| |
Collapse
|
11
|
Audet-Delage Y, Rouleau M, Villeneuve L, Guillemette C. The Glycosyltransferase Pathway: An Integrated Analysis of the Cell Metabolome. Metabolites 2022; 12:metabo12101006. [PMID: 36295907 PMCID: PMC9609030 DOI: 10.3390/metabo12101006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022] Open
Abstract
Nucleotide sugar-dependent glycosyltransferases (UGTs) are critical to the homeostasis of endogenous metabolites and the detoxification of xenobiotics. Their impact on the cell metabolome remains unknown. Cellular metabolic changes resulting from human UGT expression were profiled by untargeted metabolomics. The abundant UGT1A1 and UGT2B7 were studied as UGT prototypes along with their alternative (alt.) splicing-derived isoforms displaying structural differences. Nineteen biochemical routes were modified, beyond known UGT substrates. Significant variations in glycolysis and pyrimidine pathways, and precursors of the co-substrate UDP-glucuronic acid were observed. Bioactive lipids such as arachidonic acid and endocannabinoids were highly enriched by up to 13.3-fold (p < 0.01) in cells expressing the canonical enzymes. Alt. UGT2B7 induced drastic and unique metabolic perturbations, including higher glucose (18-fold) levels and tricarboxylic acid cycle (TCA) cycle metabolites and abrogated the effects of the UGT2B7 canonical enzyme when co-expressed. UGT1A1 proteins promoted the accumulation of branched-chain amino acids (BCAA) and TCA metabolites upstream of the mitochondrial oxoglutarate dehydrogenase complex (OGDC). Alt. UGT1A1 exacerbated these changes, likely through its interaction with the OGDC component oxoglutarate dehydrogenase-like (OGDHL). This study expands the breadth of biochemical pathways associated with UGT expression and establishes extensive connectivity between UGT enzymes, alt. proteins and other metabolic processes.
Collapse
Affiliation(s)
- Yannick Audet-Delage
- Centre Hospitalier Universitaire de Québec Research Center—Université Laval, Faculty of Pharmacy, and Université Laval Cancer Research Center (CRC), R4720, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
| | - Michèle Rouleau
- Centre Hospitalier Universitaire de Québec Research Center—Université Laval, Faculty of Pharmacy, and Université Laval Cancer Research Center (CRC), R4720, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
| | - Lyne Villeneuve
- Centre Hospitalier Universitaire de Québec Research Center—Université Laval, Faculty of Pharmacy, and Université Laval Cancer Research Center (CRC), R4720, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
| | - Chantal Guillemette
- Centre Hospitalier Universitaire de Québec Research Center—Université Laval, Faculty of Pharmacy, and Université Laval Cancer Research Center (CRC), R4720, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
- Canada Research Chair in Pharmacogenomics, Université Laval, Québec, QC G1V 4G2, Canada
- Correspondence: ; Tel.: +1-(418)-654-2296
| |
Collapse
|
12
|
Wang Z, Jiang L, Wang X, Yin H, Wang Z, Lv X, Liu Y. Cabozantinib Carries the Risk of Drug-Drug Interactions via Inhibition of UDPglucuronosyltransferase (UGT) 1A9. Curr Drug Metab 2022; 23:912-919. [PMID: 36306450 DOI: 10.2174/1389200224666221028140652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/12/2022] [Accepted: 09/28/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Cabozantinib is a multiple receptor tyrosine kinases inhibitor (TKI) approved to treat progressive, metastatic medullary thyroid cancer, advanced renal cell carcinoma, and hepatocellular carcinoma. Drugdrug interactions (DDIs) for cabozantinib have been identified involving the role of cytochromes P450. Although the previous study reported that cabozantinib showed a slight inhibition of UDP-glucuronosyltransferase (UGT) 1A1 at the highest concentration tested, there are no reports on the potential for UGTs-mediated-DDIs. Hence, the current study aims to address this knowledge gap. OBJECTIVE This study aimed to investigate the inhibitory effect of cabozantinib on human UGTs and to quantitatively evaluate the DDI potential via UGT inhibition. METHODS The inhibitory effects of cabozantinib on UGTs were determined by measuring the formation rates for 4- methylumbelliferone (4-MU) glucuronide and trifluoperazine N-glucuronide using recombinant human UGT isoforms in the absence or presence of cabozantinib. Inhibition kinetic studies were conducted to determine the type of inhibition of cabozantinib on UGTs and the corresponding inhibition constant (Ki) value. In vitro-in vivo extrapolation (IVIVE) was further employed to predict the potential risk of DDI in vivo. RESULTS Cabozantinib displayed potent inhibition of UGT1A1, 1A3, 1A4, 1A6, 1A7, 1A8, 1A9, 1A10, 2B7, and 2B15. Cabozantinib exhibited noncompetitive inhibition towards UGT1A1 and 1A3 and inhibition towards UGT1A7 and 1A9. The Ki,u values (mean ± standard deviation) were calculated to be 2.15±0.11 μM, 0.83±0.05 μM, 0.75±0.04 μM and 0.18 ± 0.10 μM for UGT1A1, 1A3, 1A7 and 1A9, respectively. Co-administration of cabozantinib at the clinically approved dose of 60 mg/day or 140 mg/day may result in approximately a 26% to 60% increase in the systemic exposure of drugs predominantly cleared by UGT1A9, implying a high risk of DDIs. CONCLUSION Cabozantinib has the potential to cause DDIs via the inhibition of UGT1A9; therefore, additional attention should be paid to the safety of the combined use of cabozantinib and drugs metabolized by UGT1A9.
Collapse
Affiliation(s)
- Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Xiaoyu Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Hang Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Xin Lv
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| |
Collapse
|
13
|
Yu Z, Hu X, Zhou L, Chen H, Xing Y, Han C, Ding H, Han L, Pan G, Fu Z. Studies on Chemical Characterization of Ginkgo Amillaria Oral Solution and Its Drug–Drug Interaction With Piceatannol 3′-O-β-D-Glucopyranoside for Injection. Front Pharmacol 2022; 13:932646. [PMID: 35928280 PMCID: PMC9344054 DOI: 10.3389/fphar.2022.932646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
Ginkgo Amillaria oral solution (GAO) is commonly used for the treatment of cardiovascular and cerebrovascular diseases in China. Piceatannol-3′-O-β-D-glucopyranoside for injection (PGI) is mainly used for the prevention and treatment of ischemic cerebrovascular diseases. With the spread of cerebrovascular disease, the possibility of combining the two drugs has increased; however, there is no research on the drug–drug interaction (DDI) between these two medicines. In this paper, an ultrahigh-performance liquid chromatography/quadrupole–orbitrap mass spectrometry (UHPLC/Q-Orbitrap MS) method was established to characterize the chemical constituents of GAO first; 62 compounds were identified or tentatively identified based on their retention time (RT), MS, and MS/MS data. Nine main compounds were determined by ultrahigh-performance liquid chromatography/triple quadrupole mass spectrometry (UPLC-QQQ-MS). Furthermore, incubation with liver microsomes in vitro was fulfilled; the results showed that GAO had a significant inhibitory effect on UGT1A9 and UGT2B7 (p < 0.05), and PGI was mainly metabolized by UGT1A9. The identification results of in vivo metabolites of PGI showed that PGI mainly undergoes a phase II binding reaction mediated by UDP-glucuronosyltransferase (UGT) and sulfotransferase (SULT) in vivo. Therefore, pharmacokinetic studies were performed to investigate the DDI between GAO and PGI. The results showed that the AUC (p < 0.05) and T1/2 (p < 0.05) of PGI in vivo were significantly increased when administered together with GAO, whereas the CL was significantly decreased (p < 0.05). The exploration of in vitro and in vivo experiments showed that there was a DDI between GAO and PGI.
Collapse
Affiliation(s)
- Zhenyan Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaohan Hu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Zhou
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huliang Chen
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanchao Xing
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chunyue Han
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Ding
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lifeng Han
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guixiang Pan
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Guixiang Pan, ; Zhifei Fu,
| | - Zhifei Fu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Guixiang Pan, ; Zhifei Fu,
| |
Collapse
|
14
|
Müller L, Keuter L, Bücksteeg D, Uebel T, Wilken M, Schürmann L, Behrens M, Humpf HU, Esselen M. Metabolic conjugation reduces in vitro toxicity of the flavonoid nevadensin. Food Chem Toxicol 2022; 164:113006. [PMID: 35436549 DOI: 10.1016/j.fct.2022.113006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/18/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
Abstract
The present study focuses on the association between metabolic capacity and toxicity of the natural occurring flavonoid nevadensin in vitro. Human colon (HT29), liver (HepG2) and bone marrow (KG1) carcinoma cells were used and strong cell line dependent differences in toxic effect strength were found. HepG2 and KG1 cells were more sensitive against nevadensin treatment in comparison to HT29 cells. High resolution mass spectrometry experiments showed that nevadensin is rapidly glucuronidated in HT29 cells, whereas KG1 cells do not metabolize nevadensin, thus glucuronidation was supposed to be a crucial metabolic pathway in vitro. To proof this suggestion, nevadensin glucuronides were isolated from pig liver microsomes und structurally elucidated via NMR spectroscopy. In HepG2 cells a cellular enrichment of nevadensin itself as well as nevadensin-7-O-glucuronide was determined by tandem mass spectrometry. A proteomic screening of uridine 5'-diphospho (UDP)-glucuronosyltransferase (UGT) in HT29 and HepG2 cells provided first hints that the isoforms UGT1A6 and UGT1A1 are responsible for nevadensin glucuronidation. Additionally, nevadensin was found to be a potent SULT inhibitor in HepG2 cells. In sum, the present study clearly illustrates the importance of obtaining detailed information about metabolic competence of cell lines which should be considered in the evaluation of toxic endpoints.
Collapse
Affiliation(s)
- Lena Müller
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany
| | - Lucas Keuter
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany
| | - David Bücksteeg
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany
| | - Thomas Uebel
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany
| | - Markus Wilken
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany
| | - Lina Schürmann
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany
| | - Matthias Behrens
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany
| | - Hans-Ulrich Humpf
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany
| | - Melanie Esselen
- University of Münster, Institute of Food Chemistry, Corrensstraße 45, 48149, Münster, Germany.
| |
Collapse
|
15
|
Hu DG, Mackenzie PI, Hulin JA, McKinnon RA, Meech R. Regulation of human UDP-glycosyltransferase ( UGT) genes by miRNAs. Drug Metab Rev 2022; 54:120-140. [PMID: 35275773 DOI: 10.1080/03602532.2022.2048846] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The human UGT gene superfamily is divided into four subfamilies (UGT1, UGT2, UGT3 and UGT8) that encodes 22 functional enzymes. UGTs are critical for the metabolism and clearance of numerous endogenous and exogenous compounds, including steroid hormones, bile acids, bilirubin, fatty acids, carcinogens, and therapeutic drugs. Therefore, the expression and activities of UGTs are tightly regulated by multiple processes at the transcriptional, post-transcriptional and post-translational levels. During recent years, nearly twenty studies have investigated the post-transcriptional regulation of UGT genes by miRNAs using human cancer cell lines (predominantly liver cancer). Overall, 14 of the 22 UGT mRNAs (1A1, 1A3, 1A4, 1A6, 1A8, 1A9, 1A10, 2A1, 2B4, 2B7, 2B10, 2B15, 2B17, UGT8) have been shown to be regulated by various miRNAs through binding to their respective 3' untranslated regions (3'UTRs). Three 3'UTRs (UGT1A, UGT2B7 and UGT2B15) contain the largest number of functional miRNA target sites; in particular, the UGT1A 3'UTR contains binding sites for 12 miRNAs (548d-5p, 183-5p, 214-5p, 486-3p, 200a-3p, 491-3p, 141-3p, 298, 103b, 376b-3p, 21-3p, 1286). Although all nine UGT1A family members have the same 3'UTR, these miRNA target sites appear to be functional in an isoform-specific and cellular context-dependent manner. Collectively, these observations demonstrate that miRNAs represent important post-transcriptional regulators of the UGT gene superfamily. In this article, we present a comprehensive review of reported UGT/miRNA regulation studies, describe polymorphisms within functional miRNA target sites that may affect their functionalities, and discuss potential cooperative and competitive regulation of UGT mRNAs by miRNAs through adjacently located miRNA target sites.
Collapse
Affiliation(s)
- Dong Gui Hu
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Peter I Mackenzie
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Julie-Ann Hulin
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Ross A McKinnon
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Robyn Meech
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| |
Collapse
|
16
|
Hu DG, Marri S, Mackenzie PI, Hulin JA, McKinnon RA, Meech R. The Expression Profiles and Deregulation of UDP-Glycosyltransferase ( UGT) Genes in Human Cancers and Their Association with Clinical Outcomes. Cancers (Basel) 2021; 13:4491. [PMID: 34503303 PMCID: PMC8430925 DOI: 10.3390/cancers13174491] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022] Open
Abstract
The human UDP-glycosyltransferase (UGTs) superfamily has 22 functional enzymes that play a critical role in the metabolism of small lipophilic compounds, including carcinogens, drugs, steroids, lipids, fatty acids, and bile acids. The expression profiles of UGT genes in human cancers and their impact on cancer patient survival remains to be systematically investigated. In the present study, a comprehensive analysis of the RNAseq and clinical datasets of 9514 patients from 33 different TCGA (the Genome Cancer Atlas) cancers demonstrated cancer-specific UGT expression profiles with high interindividual variability among and within individual cancers. Notably, cancers derived from drug metabolizing tissues (liver, kidney, gut, pancreas) expressed the largest number of UGT genes (COAD, KIRC, KIRP, LIHC, PAAD); six UGT genes (1A6, 1A9, 1A10, 2A3, 2B7, UGT8) showed high expression in five or more different cancers. Kaplan-Meier plots and logrank tests revealed that six UGT genes were significantly associated with increased overall survival (OS) rates [UGT1A1 (LUSC), UGT1A6 (ACC), UGT1A7 (ACC), UGT2A3 (KIRC), UGT2B15 (BLCA, SKCM)] or decreased OS rates [UGT2B15 (LGG), UGT8 (UVM)] in specific cancers. Finally, differential expression analysis of 611 patients from 12 TCGA cancers identified 16 UGT genes (1A1, 1A3, 1A6, 1A7, 1A8, 1A9, 1A10, 2A1, 2A3, 2B4, 2B7, 2B11, 2B15, 3A1, 3A2, UGT8) that were up/downregulated in at least one cancer relative to normal tissues. In conclusion, our data show widespread expression of UGT genes in cancers, highlighting the capacity for intratumoural drug metabolism through the UGT conjugation pathway. The data also suggests the potentials for specific UGT genes to serve as prognostic biomarkers or therapeutic targets in cancers.
Collapse
Affiliation(s)
- Dong Gui Hu
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Shashikanth Marri
- Dicipline of Molecular Medicine and Pathology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia;
| | - Peter I. Mackenzie
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Julie-Ann Hulin
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Ross A. McKinnon
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Robyn Meech
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| |
Collapse
|
17
|
Takahashi RH, Forrest WF, Smith AD, Badee J, Qiu N, Schmidt S, Collier AC, Parrott N, Fowler S. Characterization of Hepatic UDP-Glucuronosyltransferase Enzyme Abundance-Activity Correlations and Population Variability Using a Proteomics Approach and Comparison with Cytochrome P450 Enzymes. Drug Metab Dispos 2021; 49:760-769. [PMID: 34187837 DOI: 10.1124/dmd.121.000474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/24/2021] [Indexed: 11/22/2022] Open
Abstract
The expression of ten major drug-metabolizing UDP-glucuronosyltransferase (UGT) enzymes in a panel of 130 human hepatic microsomal samples was measured using a liquid chromatography-tandem mass spectrometry-based approach. Simultaneously, ten cytochromes P450 and P450 reductase were also measured, and activity-expression relationships were assessed for comparison. The resulting data sets demonstrated that, with the exception of UGT2B17, 10th to 90th percentiles of UGT expression spanned 3- to 8-fold ranges. These ranges were small relative to ranges of reported mean UGT enzyme expression across different laboratories. We tested correlation of UGT expression with enzymatic activities using selective probe substrates. A high degree of abundance-activity correlation (Spearman's rank correlation coefficient > 0.6) was observed for UGT1As (1A1, 3, 4, 6) and cytochromes P450. In contrast, protein abundance and activity did not correlate strongly for UGT1A9 and UGT2B enzymes (2B4, 7, 10, 15, and 17). Protein abundance was strongly correlated for UGTs 2B7, 2B10, and 2B15. We suggest a number of factors may contribute to these differences including incomplete selectivity of probe substrates, correlated expression of these UGT2B isoforms, and the impact of splice and polymorphic variants on the peptides used in proteomics analysis, and exemplify this in the case of UGT2B10. Extensive correlation analyses identified important criteria for validating the fidelity of proteomics and enzymatic activity approaches for assessing UGT variability, population differences, and ontogenetic changes. SIGNIFICANCE STATEMENT: Protein expression data allow detailed assessment of interindividual variability and enzyme ontogeny. This study has observed that expression and enzyme activity are well correlated for hepatic UGT1A enzymes and cytochromes P450. However, for the UGT2B family, caution is advised when assuming correlation of expression and activity as is often done in physiologically based pharmacokinetic modeling. This can be due to incomplete probe substrate specificities, but may also be related to presence of inactive UGT protein materials and the effect of splicing variations.
Collapse
Affiliation(s)
- Ryan H Takahashi
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - William F Forrest
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Alexander D Smith
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Justine Badee
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - NaHong Qiu
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Stephan Schmidt
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Abby C Collier
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Neil Parrott
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Stephen Fowler
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| |
Collapse
|
18
|
Basit A, Neradugomma NK, Wolford C, Fan PW, Murray B, Takahashi RH, Khojasteh SC, Smith BJ, Heyward S, Totah RA, Kelly EJ, Prasad B. Characterization of Differential Tissue Abundance of Major Non-CYP Enzymes in Human. Mol Pharm 2020; 17:4114-4124. [PMID: 32955894 DOI: 10.1021/acs.molpharmaceut.0c00559] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The availability of assays that predict the contribution of cytochrome P450 (CYP) metabolism allows for the design of new chemical entities (NCEs) with minimal oxidative metabolism. These NCEs are often substrates of non-CYP drug-metabolizing enzymes (DMEs), such as UDP-glucuronosyltransferases (UGTs), sulfotransferases (SULTs), carboxylesterases (CESs), and aldehyde oxidase (AO). Nearly 30% of clinically approved drugs are metabolized by non-CYP enzymes. However, knowledge about the differential hepatic versus extrahepatic abundance of non-CYP DMEs is limited. In this study, we detected and quantified the protein abundance of eighteen non-CYP DMEs (AO, CES1 and 2, ten UGTs, and five SULTs) across five different human tissues. AO was most abundantly expressed in the liver and to a lesser extent in the kidney; however, it was not detected in the intestine, heart, or lung. CESs were ubiquitously expressed with CES1 being predominant in the liver, while CES2 was enriched in the small intestine. Consistent with the literature, UGT1A4, UGT2B4, and UGT2B15 demonstrated liver-specific expression, whereas UGT1A10 expression was specific to the intestine. UGT1A1 and UGT1A3 were expressed in both the liver and intestine; UGT1A9 was expressed in the liver and kidney; and UGT2B17 levels were significantly higher in the intestine than in the liver. All five SULTs were detected in the liver and intestine, and SULT1A1 and 1A3 were detected in the lung. Kidney abundance was the most variable among the studied tissues, and overall, high interindividual variability (>15-fold) was observed for UGT2B17, CES2 (intestine), SULT1A1 (liver), UGT1A9, UGT2B7, and CES1 (kidney). These differential tissue abundance data can be integrated into physiologically based pharmacokinetic (PBPK) models for the prediction of non-CYP drug metabolism and toxicity in hepatic and extrahepatic tissues.
Collapse
Affiliation(s)
- Abdul Basit
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Naveen K Neradugomma
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, United States
| | - Christopher Wolford
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, United States
| | - Peter W Fan
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Bernard Murray
- Drug Metabolism and Pharmacokinetics Department, Gilead Sciences Inc., 324 Lakeside Drive, Foster City, California 94404, United States
| | - Ryan H Takahashi
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, MS 412a, South San Francisco, California 94080, United States
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, MS 412a, South San Francisco, California 94080, United States
| | - Bill J Smith
- Drug Metabolism and Pharmacokinetics Department, Gilead Sciences Inc., 324 Lakeside Drive, Foster City, California 94404, United States
| | - Scott Heyward
- BioIVT Inc., Baltimore, Maryland 21227, United States
| | - Rheem A Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, United States
| | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| |
Collapse
|
19
|
Wang Z, Wang X, Jia Y, Yin H, Feng Y, Jiang L, Cao J, Liu Y. Inhibition of human UDP‐glucuronosyltransferase enzymes by midostaurin and ruxolitinib: implications for drug–drug interactions. Biopharm Drug Dispos 2020; 41:231-238. [DOI: 10.1002/bdd.2241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/25/2020] [Accepted: 04/30/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Zhe Wang
- School of Life and Pharmaceutical SciencesDalian University of Technology Panjin 124221 China
| | - Xiaoyu Wang
- School of Life and Pharmaceutical SciencesDalian University of Technology Panjin 124221 China
| | - Yaqin Jia
- School of Life and Pharmaceutical SciencesDalian University of Technology Panjin 124221 China
| | - Hang Yin
- School of Life and Pharmaceutical SciencesDalian University of Technology Panjin 124221 China
| | - Yuyi Feng
- School of Life and Pharmaceutical SciencesDalian University of Technology Panjin 124221 China
| | - Lili Jiang
- School of Life and Pharmaceutical SciencesDalian University of Technology Panjin 124221 China
| | - Jun Cao
- Department of Occupational and Environmental HealthDalian Medical University Dalian 116044 China
| | - Yong Liu
- School of Life and Pharmaceutical SciencesDalian University of Technology Panjin 124221 China
| |
Collapse
|
20
|
Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)-Based Proteomics of Drug-Metabolizing Enzymes and Transporters. Molecules 2020; 25:molecules25112718. [PMID: 32545386 PMCID: PMC7321193 DOI: 10.3390/molecules25112718] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based proteomics is a powerful tool for identifying and quantifying proteins in biological samples, outperforming conventional antibody-based methods in many aspects. LC-MS/MS-based proteomics studies have revealed the protein abundances of many drug-metabolizing enzymes and transporters (DMETs) in tissues relevant to drug metabolism and disposition. Previous studies have consistently demonstrated marked interindividual variability in DMET protein expression, suggesting that varied DMET function is an important contributing factor for interindividual variability in pharmacokinetics (PK) and pharmacodynamics (PD) of medications. Moreover, differential DMET expression profiles were observed across different species and in vitro models. Therefore, caution must be exercised when extrapolating animal and in vitro DMET proteomics findings to humans. In recent years, DMET proteomics has been increasingly utilized for the development of physiologically based pharmacokinetic models, and DMET proteins have also been proposed as biomarkers for prediction of the PK and PD of the corresponding substrate drugs. In sum, despite the existence of many challenges in the analytical technology and data analysis methods of LC-MS/MS-based proteomics, DMET proteomics holds great potential to advance our understanding of PK behavior at the individual level and to optimize treatment regimens via the DMET protein biomarker-guided precision pharmacotherapy.
Collapse
|
21
|
Emami-Khoyi A, Parbhu SP, Ross JG, Murphy EC, Bothwell J, Monsanto DM, Vuuren BJV, Teske PR, Paterson AM. De Novo Transcriptome Assembly and Annotation of Liver and Brain Tissues of Common Brushtail Possums ( Trichosurus vulpecula) in New Zealand: Transcriptome Diversity after Decades of Population Control. Genes (Basel) 2020; 11:genes11040436. [PMID: 32316496 PMCID: PMC7230921 DOI: 10.3390/genes11040436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022] Open
Abstract
The common brushtail possum (Trichosurus vulpecula), introduced from Australia in the mid-nineteenth century, is an invasive species in New Zealand where it is widespread and forms the largest self-sustained reservoir of bovine tuberculosis (Mycobacterium bovis) among wild populations. Conservation and agricultural authorities regularly apply a series of population control measures to suppress brushtail possum populations. The evolutionary consequence of more than half a century of intensive population control operations on the species’ genomic diversity and population structure is hindered by a paucity of available genomic resources. This study is the first to characterise the functional content and diversity of brushtail possum liver and brain cerebral cortex transcriptomes. Raw sequences from hepatic cells and cerebral cortex were assembled into 58,001 and 64,735 transcripts respectively. Functional annotation and polymorphism assignment of the assembled transcripts demonstrated a considerable level of variation in the core metabolic pathways that represent potential targets for selection pressure exerted by chemical toxicants. This study suggests that the brushtail possum population in New Zealand harbours considerable variation in metabolic pathways that could potentially promote the development of tolerance against chemical toxicants.
Collapse
Affiliation(s)
- Arsalan Emami-Khoyi
- Center for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Auckland Park 2006, South Africa
- Department of Pest-management and Conservation, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Shilpa Pradeep Parbhu
- Center for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Auckland Park 2006, South Africa
| | - James G Ross
- Department of Pest-management and Conservation, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Elaine C Murphy
- Department of Pest-management and Conservation, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Jennifer Bothwell
- Department of Pest-management and Conservation, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Daniela M Monsanto
- Center for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Auckland Park 2006, South Africa
| | - Bettine Jansen van Vuuren
- Center for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Auckland Park 2006, South Africa
| | - Peter R Teske
- Center for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Auckland Park 2006, South Africa
| | - Adrian M Paterson
- Department of Pest-management and Conservation, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| |
Collapse
|
22
|
Emerging roles for UDP-glucuronosyltransferases in drug resistance and cancer progression. Br J Cancer 2020; 122:1277-1287. [PMID: 32047295 PMCID: PMC7188667 DOI: 10.1038/s41416-019-0722-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/06/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
The best-known role of UDP-glucuronosyltransferase enzymes (UGTs) in cancer is the metabolic inactivation of drug therapies. By conjugating glucuronic acid to lipophilic drugs, UGTs impair the biological activity and enhance the water solubility of these agents, driving their elimination. Multiple clinical observations support an expanding role for UGTs as modulators of the drug response and in mediating drug resistance in numerous cancer types. However, accumulating evidence also suggests an influence of the UGT pathway on cancer progression. Dysregulation of the expression and activity of UGTs has been associated with the progression of several cancers, arguing for UGTs as possible mediators of oncogenic pathways and/or disease accelerators in a drug-naive context. The consequences of altered UGT activity on tumour biology are incompletely understood. They might be associated with perturbed levels of bioactive endogenous metabolites such as steroids and bioactive lipids that are inactivated by UGTs or through non-enzymatic mechanisms, thereby eliciting oncogenic signalling cascades. This review highlights the evidence supporting dual roles for the UGT pathway, affecting cancer progression and drug resistance. Pharmacogenomic testing of UGT profiles in patients and the development of therapeutic options that impair UGT actions could provide useful prognostic and predictive biomarkers and enhance the efficacy of anti-cancer drugs.
Collapse
|
23
|
Beyerle J, Holowatyj AN, Haffa M, Frei E, Gigic B, Schrotz-King P, Boehm J, Habermann N, Stiborova M, Scherer D, Kölsch T, Skender S, Becker N, Herpel E, Schneider M, Ulrich A, Schirmacher P, Chang-Claude J, Brenner H, Hoffmeister M, Haug U, Owen RW, Ulrich CM. Expression Patterns of Xenobiotic-Metabolizing Enzymes in Tumor and Adjacent Normal Mucosa Tissues among Patients with Colorectal Cancer: The ColoCare Study. Cancer Epidemiol Biomarkers Prev 2019; 29:460-469. [PMID: 31740522 DOI: 10.1158/1055-9965.epi-19-0449] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/15/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Xenobiotic-metabolizing enzymes (XME) play a critical role in the activation and detoxification of several carcinogens. However, the role of XMEs in colorectal carcinogenesis is unclear. METHODS We investigated the expression of XMEs in human colorectal tissues among patients with stage I-IV colorectal cancer (n = 71) from the ColoCare Study. Transcriptomic profiling using paired colorectal tumor and adjacent normal mucosa tissues of XMEs (GSTM1, GSTA1, UGT1A8, UGT1A10, CYP3A4, CYP2C9, GSTP1, and CYP2W1) by RNA microarray was compared using Wilcoxon rank-sum tests. We assessed associations between clinicopathologic, dietary, and lifestyle factors and XME expression with linear regression models. RESULTS GSTM1, GSTA1, UGT1A8, UGT1A10, and CYP3A4 were all statistically significantly downregulated in colorectal tumor relative to normal mucosa tissues (all P ≤ 0.03). Women had significantly higher expression of GSTM1 in normal tissues compared with men (β = 0.37, P = 0.02). By tumor site, CYP2C9 expression was lower in normal mucosa among patients with rectal cancer versus colon cancer cases (β = -0.21, P = 0.0005). Smokers demonstrated higher CYP2C9 expression levels in normal mucosa (β = 0.17, P = 0.02) when compared with nonsmokers. Individuals who used NSAIDs had higher GSTP1 tumor expression compared with non-NSAID users (β = 0.17, P = 0.03). Higher consumption of cooked vegetables (>1×/week) was associated with higher CYP3A4 expression in colorectal tumor tissues (β = 0.14, P = 0.007). CONCLUSIONS XMEs have lower expression in colorectal tumor relative to normal mucosa tissues and may modify colorectal carcinogenesis via associations with clinicopathologic, lifestyle, and dietary factors. IMPACT Better understanding into the role of drug-metabolizing enzymes in colorectal cancer may reveal biological differences that contribute to cancer development, as well as treatment response, leading to clinical implications in colorectal cancer prevention and management.
Collapse
Affiliation(s)
- Jolantha Beyerle
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Andreana N Holowatyj
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah.,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Mariam Haffa
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Eva Frei
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Petra Schrotz-King
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Juergen Boehm
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah.,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Nina Habermann
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Dominique Scherer
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Torsten Kölsch
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Stephanie Skender
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Nikolaus Becker
- NCT Cancer Registry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, University Hospital, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | | | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Haug
- Department of Clinical Epidemiology, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany.,Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany
| | - Robert W Owen
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Cornelia M Ulrich
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah. .,Huntsman Cancer Institute, Salt Lake City, Utah.,Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
24
|
Hu DG, Hulin JUA, Nair PC, Haines AZ, McKinnon RA, Mackenzie PI, Meech R. The UGTome: The expanding diversity of UDP glycosyltransferases and its impact on small molecule metabolism. Pharmacol Ther 2019; 204:107414. [PMID: 31647974 DOI: 10.1016/j.pharmthera.2019.107414] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/17/2019] [Indexed: 01/23/2023]
Abstract
The UDP glycosyltransferase (UGT) superfamily of enzymes is responsible for the metabolism and clearance of thousands of lipophilic chemicals including drugs, toxins and endogenous signaling molecules. They provide a protective interface between the organism and its chemical-rich environment, as well as controlling critical signaling pathways to maintain healthy tissue function. UGTs are associated with drug responses and interactions, as well as a wide range of diseases including cancer. The human genome contains 22 UGT genes; however as befitting their exceptionally diverse substrate ranges and biological activities, the output of these UGT genes is functionally diversified by multiple processes including alternative splicing, post-translational modification, homo- and hetero-oligomerization, and interactions with other proteins. All UGT genes are subject to extensive alternative splicing generating variant/truncated UGT proteins with altered functions including the capacity to dominantly modulate/inhibit cognate full-length forms. Heterotypic oligomerization of different UGTs can alter kinetic properties relative to monotypic complexes, and potentially produce novel substrate specificities. Moreover, the recently profiled interactions of UGTs with non-UGT proteins may facilitate coordination between different metabolic processes, as well as providing opportunities for UGTs to engage in novel 'moonlighting' functions. Herein we provide a detailed and comprehensive review of all known modes of UGT functional diversification and propose a UGTome model to describe the resulting expansion of metabolic capacity and its potential to modulate drug/xenobiotic responses and cell behaviours in normal and disease contexts.
Collapse
Affiliation(s)
- Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - J Ulie-Ann Hulin
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Pramod C Nair
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Alex Z Haines
- Department of Clinical Pharmacology and Flinders Cancer Centre, 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 Cancer Centre, 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 Cancer Centre, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Robyn Meech
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia.
| |
Collapse
|
25
|
Zhang Z, Liu D, Jiang J, Song X, Zou X, Chu S, Xie K, Dai J, Chen N, Sheng L, Li Y. Metabolism of IMM-H004 and Its Pharmacokinetic-Pharmacodynamic Analysis in Cerebral Ischemia/Reperfusion Injured Rats. Front Pharmacol 2019; 10:631. [PMID: 31249524 PMCID: PMC6584114 DOI: 10.3389/fphar.2019.00631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/17/2019] [Indexed: 12/23/2022] Open
Abstract
IMM-H004, a derivative of coumarin, is a promising candidate for the treatment of cerebral ischemia. The pharmacodynamic mechanisms of IMM-H004 are still under exploration. The present study was conducted to explore the pharmacoactive substances of IMM-H004 from the perspective of drug metabolism. Four metabolites of IMM-H004 including demethylated metabolites M1 and M2, glucuronide conjugate IMM-H004G (M3), and sulfated conjugate M4 were found in rats in vivo. IMM-H004G was the major metabolite in rats and cultured human hepatocytes, and uridine diphosphate-glucuronosyltransferase (UGT) was found to catalyze the metabolism of IMM-H004 in human liver microsomes (HLMs) and rat liver microsomes (RLMs) with high capacity (V max at 3.25 and 5.04 nmol/min/mg protein). Among 13 recombinant human UGT isoforms, UGT1A7, 1A9, 1A8, and 1A1 appeared to be primarily responsible for IMM-H004G formation. The exposure and duration of IMM-H004G (28,948 h × ng/ml of area under the plasma concentration-time curve (AUC), 6.61 h of t 1/2β) was much higher than that of the parent drug (1,638 h × ng/ml of AUC, 0.42 h of t 1/2β) in transient middle cerebral artery occlusion/reperfusion (MCAO/R) rats, consistent with the malondialdehyde (MDA) inhibition effect for at least 10 h. Further pharmacological study revealed that IMM-H004G exhibited a similar neuroprotective activity to that of the parent drug on both oxygen-glucose deprivation injured PC12 cells and transient MCAO/R injured rats. These results demonstrate that both prototype and IMM-H004G are the active pharmaceutical substances, and IMM-H004G, at least in part, contributes to the maintenance of anti-cerebral ischemia efficacy of IMM-H004.
Collapse
Affiliation(s)
- Ziqian Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dandan Liu
- State Key Laboratory of Bioactive Substances and Function Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianwei Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Bioactive Substances and Function Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiuyun Song
- State Key Laboratory of Bioactive Substances and Function Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaowen Zou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Function Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kebo Xie
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jungui Dai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Naihong Chen
- State Key Laboratory of Bioactive Substances and Function Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Sheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Woelflingseder L, Warth B, Vierheilig I, Schwartz-Zimmermann H, Hametner C, Nagl V, Novak B, Šarkanj B, Berthiller F, Adam G, Marko D. The Fusarium metabolite culmorin suppresses the in vitro glucuronidation of deoxynivalenol. Arch Toxicol 2019; 93:1729-1743. [PMID: 31049613 PMCID: PMC6620244 DOI: 10.1007/s00204-019-02459-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/18/2019] [Indexed: 12/29/2022]
Abstract
Glucuronidation is a major phase II conjugation pathway in mammals, playing an important role in the detoxification and biotransformation of xenobiotics including mycotoxins such as deoxynivalenol (DON). Culmorin (CUL), a potentially co-occurring Fusarium metabolite, was recently found to inhibit the corresponding detoxification reaction in plants, namely DON-glucoside formation, raising the question whether CUL might affect also the mammalian counterpart. Using cell-free conditions, CUL when present equimolar (67 µM) or in fivefold excess, suppressed DON glucuronidation by human liver microsomes, reducing the formation of DON-15-glucuronide by 15 and 50%, and DON-3-glucuronide by 30 and 50%, respectively. Substantial inhibitory effects on DON glucuronidation up to 100% were found using the human recombinant uridine 5'-diphospho-glucuronosyltransferases (UGT) 2B4 and 2B7, applying a tenfold excess of CUL (100 µM). In addition, we observed the formation of a novel metabolite of CUL, CUL-11-glucuronide, identified for the first time in vitro as well as in vivo in piglet and human urine samples. Despite the observed potency of CUL to inhibit glucuronidation, no significant synergistic toxicity on cell viability was observed in combinations of CUL (0.1-100 µM) and DON (0.01-10 µM) in HT-29 and HepG2 cells, presumably reflecting the limited capacity of the tested cell lines for DON glucuronidation. However, in humans, glucuronidation is known to represent the main detoxification pathway for DON. The present results, including the identification of CUL-11-glucuronide in urine samples of piglets and humans, underline the necessity of further studies on the relevance of CUL as a potentially co-occurring modulator of DON toxicokinetics in vivo.
Collapse
Affiliation(s)
- Lydia Woelflingseder
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstrasse 38, 1090 Vienna, Austria
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstrasse 38, 1090 Vienna, Austria
| | - Immina Vierheilig
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstrasse 38, 1090 Vienna, Austria
| | - Heidi Schwartz-Zimmermann
- Christian Doppler Laboratory for Mycotoxin Metabolism and Center for Analytical Chemistry, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Strasse 20, 3430 Tulln, Austria
| | - Christian Hametner
- Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9/163, 1060 Vienna, Austria
| | - Veronika Nagl
- BIOMIN Research Center, Technopark 1, 3430 Tulln, Austria
| | - Barbara Novak
- BIOMIN Research Center, Technopark 1, 3430 Tulln, Austria
| | - Bojan Šarkanj
- Department of Applied Chemistry and Ecology, Faculty of Food Technology, Josip Juraj Strossmayer University of Osijek, Franje Kuhača 20, 31000 Osijek, Croatia
| | - Franz Berthiller
- Christian Doppler Laboratory for Mycotoxin Metabolism and Center for Analytical Chemistry, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Strasse 20, 3430 Tulln, Austria
| | - Gerhard Adam
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Strasse 24, 3430 Tulln, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstrasse 38, 1090 Vienna, Austria
| |
Collapse
|
27
|
Émond JP, Labriet A, Desjardins S, Rouleau M, Villeneuve L, Hovington H, Brisson H, Lacombe L, Simonyan D, Caron P, Périgny M, Têtu B, Fallon JK, Klein K, Smith PC, Zanger UM, Guillemette C, Lévesque E. Factors Affecting Interindividual Variability of Hepatic UGT2B17 Protein Expression Examined Using a Novel Specific Monoclonal Antibody. Drug Metab Dispos 2019; 47:444-452. [PMID: 30819787 DOI: 10.1124/dmd.119.086330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 02/21/2019] [Indexed: 08/10/2024] Open
Abstract
Accurate quantification of the metabolic enzyme uridine diphospho-glucuronosyltransferase (UGT) UGT2B17 has been hampered by the high sequence identity with other UGT2B enzymes (as high as 94%) and by the lack of a specific antibody. Knowing the significance of the UGT2B17 pathway in drug and hormone metabolism and cancer, we developed a specific monoclonal antibody (EL-2B17mAb), initially validated by the lack of detection in liver microsomes of an individual carrying no UGT2B17 gene copy and in supersomes expressing UGT2B enzymes. Immunohistochemical detection in livers revealed strong labeling of bile ducts and variable labeling of hepatocytes. Expression levels assessed by immunoblotting were highly correlated to mass spectrometry-based quantification (r = 0.93), and three major expression patterns (absent, low, or high) were evidenced. Livers with very low expression were carriers of the functional rs59678213 G variant, located in the binding site for the transcription factor forkhead box A1 (FOXA1) of the UGT2B17 promoter. The highest level of expression was observed for individuals carrying at least one rs59678213 A allele. Multiple regression analysis indicated that the number of gene copies explained only 8% of UGT2B17 protein expression, 49% when adding rs59678213, reaching 54% when including sex. The novel EL-2B17mAb antibody allowed specific UGT2B17 quantification and exposed different patterns of hepatic expression. It further suggests that FOXA1 is a key driver of UGT2B17 expression in the liver. The availability of this molecular tool will help characterize the UGT2B17 level in various disease states and establish more precisely the contribution of the UGT2B17 enzyme to drug and hormone metabolism.
Collapse
Affiliation(s)
- Jean-Philippe Émond
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Adrien Labriet
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Sylvie Desjardins
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Michèle Rouleau
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Lyne Villeneuve
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Hélène Hovington
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Hervé Brisson
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Louis Lacombe
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - David Simonyan
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Patrick Caron
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Martine Périgny
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Bernard Têtu
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - John K Fallon
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Kathrin Klein
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Philip C Smith
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Ulrich M Zanger
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Chantal Guillemette
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Eric Lévesque
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| |
Collapse
|
28
|
Meech R, Hu DG, McKinnon RA, Mubarokah SN, Haines AZ, Nair PC, Rowland A, Mackenzie PI. The UDP-Glycosyltransferase (UGT) Superfamily: New Members, New Functions, and Novel Paradigms. Physiol Rev 2019; 99:1153-1222. [DOI: 10.1152/physrev.00058.2017] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UDP-glycosyltransferases (UGTs) catalyze the covalent addition of sugars to a broad range of lipophilic molecules. This biotransformation plays a critical role in elimination of a broad range of exogenous chemicals and by-products of endogenous metabolism, and also controls the levels and distribution of many endogenous signaling molecules. In mammals, the superfamily comprises four families: UGT1, UGT2, UGT3, and UGT8. UGT1 and UGT2 enzymes have important roles in pharmacology and toxicology including contributing to interindividual differences in drug disposition as well as to cancer risk. These UGTs are highly expressed in organs of detoxification (e.g., liver, kidney, intestine) and can be induced by pathways that sense demand for detoxification and for modulation of endobiotic signaling molecules. The functions of the UGT3 and UGT8 family enzymes have only been characterized relatively recently; these enzymes show different UDP-sugar preferences to that of UGT1 and UGT2 enzymes, and to date, their contributions to drug metabolism appear to be relatively minor. This review summarizes and provides critical analysis of the current state of research into all four families of UGT enzymes. Key areas discussed include the roles of UGTs in drug metabolism, cancer risk, and regulation of signaling, as well as the transcriptional and posttranscriptional control of UGT expression and function. The latter part of this review provides an in-depth analysis of the known and predicted functions of UGT3 and UGT8 enzymes, focused on their likely roles in modulation of levels of endogenous signaling pathways.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - Siti Nurul Mubarokah
- 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
| | - Alex Z. Haines
- 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
| | - Pramod C. Nair
- 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
| | - Andrew Rowland
- 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
| |
Collapse
|
29
|
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. [PMID: 29959221 DOI: 10.1124/mol.118.111773] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/08/2018] [Indexed: 02/14/2025] Open
Abstract
Recent studies have investigated alternative splicing profiles of UDP-glucuronosyltransferase (UGT) genes and identified over 130 different alternatively spliced UGT transcripts. Although UGT genes are highly clustered, the formation of chimeric transcripts by intergenic splicing between two or more UGT genes has not yet been reported. This study identified 12 chimeric transcripts (chimeras A-L) containing exons from two or three genes of the four neighboring UGT genes (UGT2B15, UGT2B29P2, UGT2B17, and UGT2B29P1) in human liver and prostate cancer cells. These chimeras typically contain the first five exons of UGT2B15 or UGT2B17 (exons 1-5) spliced to a terminal exon (exon 6) from a downstream UGT gene. Hence they encode truncated UGTs with novel C-terminal peptides. Functional assays of representative chimeric UGT proteins (termed chimeric UGT2B15 and chimeric UGT2B17) showed that they are inactive and can repress the activity of wild-type UGTs. Coimmunoprecipitation assays demonstrated heterotypic interactions between chimeric UGT2B15 (or chimeric UGT2B17) and the UGT2B7 protein. Thus oligomerization of the chimeric UGTs with wild-type UGTs may explain their inhibitory activity. Studies in breast and prostate cancer cells showed that both wild-type and chimeric UGT2B15 and UGT2B17 transcripts are regulated in a similar way at the transcriptional level by sex hormones through their canonical promoters but are differentially regulated at the post-transcriptional level by micro-RNA 376c via their unique 3'-untranslated regions. In conclusion, the formation of chimeric transcripts by intergenic splicing among UGT genes represents a novel mechanism contributing to the diversity of the human UGT transcriptome and proteome. The differential post-transcriptional regulation of wild-type and variant transcripts by micro-RNAs may contribute to their deregulated expression in cancer.
Collapse
Affiliation(s)
- Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Bedford Park, South Australia, Australia
| | - Julie-Ann Hulin
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Bedford Park, South Australia, Australia
| | - Dhilushi D Wijayakumara
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, 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, 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, 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, Bedford Park, South Australia, Australia
| |
Collapse
|
30
|
Labriet A, Allain EP, Rouleau M, Audet-Delage Y, Villeneuve L, Guillemette C. Post-transcriptional Regulation of UGT2B10 Hepatic Expression and Activity by Alternative Splicing. Drug Metab Dispos 2018; 46:514-524. [PMID: 29438977 PMCID: PMC5894810 DOI: 10.1124/dmd.117.079921] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/31/2018] [Indexed: 12/17/2022] Open
Abstract
The detoxification enzyme UDP-glucuronosyltransferase UGT2B10 is specialized in the N-linked glucuronidation of many drugs and xenobiotics. Preferred substrates possess tertiary aliphatic amines and heterocyclic amines, such as tobacco carcinogens and several antidepressants and antipsychotics. We hypothesized that alternative splicing (AS) constitutes a means to regulate steady-state levels of UGT2B10 and enzyme activity. We established the transcriptome of UGT2B10 in normal and tumoral tissues of multiple individuals. The highest expression was in the liver, where 10 AS transcripts represented 50% of the UGT2B10 transcriptome in 50 normal livers and 44 hepatocellular carcinomas. One abundant class of transcripts involves a novel exonic sequence and leads to two alternative (alt.) variants with novel in-frame C termini of 10 or 65 amino acids. Their hepatic expression was highly variable among individuals, correlated with canonical transcript levels, and was 3.5-fold higher in tumors. Evidence for their translation in liver tissues was acquired by mass spectrometry. In cell models, they colocalized with the enzyme and influenced the conjugation of amitriptyline and levomedetomidine by repressing or activating the enzyme (40%-70%; P < 0.01) in a cell context-specific manner. A high turnover rate for the alt. proteins, regulated by the proteasome, was observed in contrast to the more stable UGT2B10 enzyme. Moreover, a drug-induced remodeling of UGT2B10 splicing was demonstrated in the HepaRG hepatic cell model, which favored alt. variants expression over the canonical transcript. Our findings support a significant contribution of AS in the regulation of UGT2B10 expression in the liver with an impact on enzyme activity.
Collapse
Affiliation(s)
- Adrien Labriet
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Eric P Allain
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Michèle Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Yannick Audet-Delage
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Lyne Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| |
Collapse
|
31
|
Audet-Delage Y, Rouleau M, Rouleau M, Roberge J, Miard S, Picard F, Têtu B, Guillemette C. Cross-Talk between Alternatively Spliced UGT1A Isoforms and Colon Cancer Cell Metabolism. Mol Pharmacol 2017; 91:167-177. [PMID: 28049773 DOI: 10.1124/mol.116.106161] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 12/29/2016] [Indexed: 12/16/2023] Open
Abstract
Alternative splicing at the human glucuronosyltransferase 1 gene locus (UGT1) produces alternate isoforms UGT1A_i2s that control glucuronidation activity through protein-protein interactions. Here, we hypothesized that UGT1A_i2s function as a complex protein network connecting other metabolic pathways with an influence on cancer cell metabolism. This is based on a pathway enrichment analysis of proteomic data that identified several high-confidence candidate interaction proteins of UGT1A_i2 proteins in human tissues-namely, the rate-limiting enzyme of glycolysis pyruvate kinase (PKM), which plays a critical role in cancer cell metabolism and tumor growth. The partnership of UGT1A_i2 and PKM2 was confirmed by coimmunoprecipitation in the HT115 colon cancer cells and was supported by a partial colocalization of these two proteins. In support of a functional role for this partnership, depletion of UGT1A_i2 proteins in HT115 cells enforced the Warburg effect, with a higher glycolytic rate at the expense of mitochondrial respiration, and led to lactate accumulation. Untargeted metabolomics further revealed a significantly altered cellular content of 58 metabolites, including many intermediates derived from the glycolysis and tricarboxylic acid cycle pathways. These metabolic changes were associated with a greater migration potential. The potential relevance of our observations is supported by the down-regulation of UGT1A_i2 mRNA in colon tumors compared with normal tissues. Alternate UGT1A variants may thus be part of the expanding compendium of metabolic pathways involved in cancer biology directly contributing to the oncogenic phenotype of colon cancer cells. Findings uncover new aspects of UGT functions diverging from their transferase activity.
Collapse
Affiliation(s)
- Yannick Audet-Delage
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Michèle Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Mélanie Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Joannie Roberge
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Stéphanie Miard
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Frédéric Picard
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Bernard Têtu
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| |
Collapse
|