1
|
Li W, Xue Y, Zhang F, Xiao L, Huang Z, Li W, Zhu L, Ge G. In Vitro Ciclopirox Glucuronidation in Liver Microsomes from Humans and Various Experimental Animals. Eur J Drug Metab Pharmacokinet 2024; 49:619-629. [PMID: 38990427 DOI: 10.1007/s13318-024-00907-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND AND OBJECTIVE Ciclopirox is a widely used antifungal drug, redisposition of which has drawn increasing attentions due to multiple promising activities. The drug undergoes extensive glucuronidation, which acts as a major obstacle in the ongoing novel application and still remains poorly understood. The current study aims to phenotype ciclopirox glucuronidation pathway and as well to decipher the related species differences. METHODS Ciclopirox glucuronidation was investigated in liver microsomes from humans (HLM) and various experimental animals. Assays with recombinant uridine diphosphate glucuronosyltransferases (UGTs), enzyme kinetic analyses and selective inhibitors were used to determine the role of individual UGTs in ciclopirox glucuronidation. RESULTS HLM is highly active in ciclopirox glucuronidation with Michaelis-Menten constant (Km), maximum velocity (Vmax), and intrinsic clearance (CLint) values of 139 μM, 7.89 nmol/min/mg, and 56 μL/min/mg, respectively. UGT1A9 displays by far the highest activity, whereas several other isoforms (UGT1A6, UGT1A7, and UGT1A8) catalyze formation of traced glucuronides. Further kinetic analysis demonstrates that UGT1A9 has a closed Km value (167 μM) to HLM. UGT1A9 selective inhibitor (magnolol) can potently inhibit ciclopirox glucuronidation in HLM with the IC50 value of 0.12 μM. The reaction displays remarkable differences across liver microsomes from mice, rats, cynomolgus monkey, minipig, and beagle dog, with the CLint values in the range of 26-369 μL/min/mg. In addition, ciclopirox glucuronidation activities of experimental animals' liver microsomes were less sensitive to magnolol than that of HLM. CONCLUSIONS Ciclopirox glucuronidation displays remarkable species differences with UGT1A9 as a dominant contributor in humans. It is suggested that the pharmacological or toxicological effects of ciclopirox may be UGT1A9 and species dependent.
Collapse
Affiliation(s)
- Wenjing Li
- School of Life Science, Innovation Center of Targeted Development of Medicinal Resources (iCTM), Anqing Normal University, 1318 Jixianbei Road, Anqing, 246133, People's Republic of China
| | - Yufan Xue
- School of Life Science, Innovation Center of Targeted Development of Medicinal Resources (iCTM), Anqing Normal University, 1318 Jixianbei Road, Anqing, 246133, People's Republic of China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology and Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, People's Republic of China
| | - Ling Xiao
- School of Resources and Environment, Anqing Normal University, Anqing, 246311, People's Republic of China
| | - Zhu Huang
- School of Life Science, Innovation Center of Targeted Development of Medicinal Resources (iCTM), Anqing Normal University, 1318 Jixianbei Road, Anqing, 246133, People's Republic of China
| | - Wenjuan Li
- School of Life Science, Innovation Center of Targeted Development of Medicinal Resources (iCTM), Anqing Normal University, 1318 Jixianbei Road, Anqing, 246133, People's Republic of China
| | - Liangliang Zhu
- School of Life Science, Innovation Center of Targeted Development of Medicinal Resources (iCTM), Anqing Normal University, 1318 Jixianbei Road, Anqing, 246133, People's Republic of China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology and Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, People's Republic of China.
| |
Collapse
|
2
|
Liu P, Li Q, Zhu G, Zhang T, Tu D, Zhang F, Finel M, He Y, Ge G. Characterization of the glucuronidating pathway of pectolinarigenin, the major active constituent of the Chinese medicine Daji, in humans and its influence on biological activities. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117280. [PMID: 37797876 DOI: 10.1016/j.jep.2023.117280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/13/2023] [Accepted: 10/03/2023] [Indexed: 10/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Chinese medicine Daji (the aerial part of Cirsium japonicum DC.) and its charred product (Cirsii Japonici Herba Carbonisata) have been widely used as hemostatic agents or diuretic agents to prepare a variety of Chinese herbal formula. Pectolinarigenin (PEC), one of the most abundant constituents in both Daji and its charred product, has been considered as the key effective substance responsible for the major pharmacological activities of Daji, including hemostasis, hepatoprotective, anti-tumor and anti-osteoporosis effects. However, the major metabolic pathways of PEC in humans and the influence of PEC metabolism on its biological activities are poorly understood. AIM OF THE STUDY To characterize the main metabolic pathway(s) and key enzymes of PEC in human biological systems, as well as to reveal the influence of PEC metabolism on its biological activities. MATERIALS AND METHODS The metabolic stability assays of PEC were investigated in human liver microsomes (HLM). The O-glucuronide of PEC was biosynthesized and characterized by nuclear magnetic resonance (NMR) spectroscopy. The key enzymes responsible for O-glucuronidation of PEC in humans were assigned by performing UGT reaction phenotyping, chemical inhibition and enzymatic kinetic assays. The agonist effects of PEC and its O-glucuronide on nuclear factor erythroid2-related factor 2 (Nrf2), Peroxisome proliferator activated receptors (PPARα and PPARβ) were tested at the cellular level. RESULTS PEC could be readily metabolized to form a mono-O-glucuronide in both human liver microsome (HLM) and human intestinal microsome (HIM). The mono-O-glucuronide was bio-synthesized by mouse liver S9 and its structure was fully characterized as PEC-7-O-β-D-glucuronide (PEC-O-7-G). UGT1A1, UGT1A3 and UGT1A9 are key enzymes responsible for PEC-7-O-glucuronidation in HLM, while UGT1A1, UGT1A9 and 1A10 may play key roles in this reaction in HIM. Biological tests revealed that PEC displayed strong agonist effects on Nrf2, PPARα and PPARβ, whereas PEC-7-O-glucuronide showed relatively weak Nrf2 agonist effect and very weak PPAR agonist effects, indicating that PEC-7-O-glucuronidation strongly weaken its agonist effects on Nrf2 and PPAR. CONCLUSIONS Our results demonstrate that 7-O-glucuronidation is the major metabolic pathway of PEC in human tissues, while UGT1A1, 1A3 and 1A9 are key contributing enzymes responsible for PEC-7-O-glucuronidation in human liver. It is also found that PEC 7-O-glucuronidation significantly weakens the Nrf2 and PPAR agonist effects. All these findings are very helpful for the pharmacologists to deep understand the metabolic rates of PEC in humans.
Collapse
Affiliation(s)
- Peiqi Liu
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qian Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guanghao Zhu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tiantian Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dongzhu Tu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Yuqi He
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China.
| | - Guangbo Ge
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
3
|
Miyauchi Y, Kimura A, Sawai M, Fujimoto K, Hirota Y, Tanaka Y, Takechi S, Mackenzie PI, Ishii Y. Use of a Baculovirus-Mammalian Cell Expression-System for Expression of Drug-Metabolizing Enzymes: Optimization of Infection With a Focus on Cytochrome P450 3A4. Front Pharmacol 2022; 13:832931. [PMID: 35295333 PMCID: PMC8919721 DOI: 10.3389/fphar.2022.832931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/28/2022] [Indexed: 11/28/2022] Open
Abstract
Heterologous expression systems are important for analyzing the effects of genetic factors including single nucleotide polymorphisms on the functions of drug-metabolizing enzymes. In this study, we focused on a baculovirus-mammalian cell (Bac-Mam) expression system as a safer and more efficient approach for this purpose. The baculovirus-insect cell expression system is widely utilized in large-scale protein expression. Baculovirus has been shown to also infect certain mammalian cells, although the virus only replicates in insect cells. With this knowledge, baculovirus is now being applied in a mammalian expression system called the Bac-Mam system wherein a gene-modified baculovirus is used whose promotor is replaced with one that can function in mammalian cells. We subcloned open-reading frames of cytochrome P450 3A4 (CYP3A4), UDP-glucuronosyltransferase (UGT) 1A1, and UGT2B7 into a transfer plasmid for the Bac-Mam system, and prepared recombinant Bac-Mam virus. The obtained virus was amplified in insect Sf9 cells and used to infect mammalian COS-1 cells. Expression of CYP3A4, UGT1A1, and UGT2B7 in COS-1 cell homogenates were confirmed by immunoblotting. Optimum infection conditions including the amount of Bac-Mam virus, culture days before collection, and concentration of sodium butyrate, an enhancer of viral-transduction were determined by monitoring CYP3A4 expression. Expressed CYP3A4 showed appropriate activity without supplying hemin/5-aminolevulinic acid or co-expressing with NADPH-cytochrome P450 reductase. Further, we compared gene transfer efficiency between the Bac-Mam system and an established method using recombinant plasmid and transfection reagent. Our results indicate that the Bac-Mam system can be applied to introduce drug-metabolizing enzyme genes into mammalian cells that are widely used in drug metabolism research. The expressed enzymes are expected to undergo appropriate post-translational modification as they are in mammalian bodies. The Bac-Mam system may thus accelerate pharmacogenetics and pharmacogenomics research.
Collapse
Affiliation(s)
- Yuu Miyauchi
- Laboratory of Hygienic Chemistry, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan.,Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akane Kimura
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Madoka Sawai
- Laboratory of Hygienic Chemistry, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Keiko Fujimoto
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuko Hirota
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshitaka Tanaka
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Takechi
- Laboratory of Hygienic Chemistry, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Peter I Mackenzie
- Clinical Pharmacology, College of Medicine and Public Health, Flinders Medical Centre and Flinders University, Adelaide, SA, Australia
| | - Yuji Ishii
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
4
|
Cussotto S, Walsh J, Golubeva AV, Zhdanov AV, Strain CR, Fouhy F, Stanton C, Dinan TG, Hyland NP, Clarke G, Cryan JF, Griffin BT. The gut microbiome influences the bioavailability of olanzapine in rats. EBioMedicine 2021; 66:103307. [PMID: 33819741 PMCID: PMC8047500 DOI: 10.1016/j.ebiom.2021.103307] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/21/2022] Open
Abstract
Background The role of the gut microbiome in the biotransformation of drugs has recently come under scrutiny. It remains unclear whether the gut microbiome directly influences the extent of drug absorbed after oral administration and thus potentially alters clinical pharmacokinetics. Methods In this study, we evaluated whether changes in the gut microbiota of male Sprague Dawley rats, as a result of either antibiotic or probiotic administration, influenced the oral bioavailability of two commonly prescribed antipsychotics, olanzapine and risperidone. Findings The bioavailability of olanzapine, was significantly increased (1.8-fold) in rats that had undergone antibiotic-induced depletion of gut microbiota, whereas the bioavailability of risperidone was unchanged. There was no direct effect of microbiota depletion on the expression of major CYP450 enzymes involved in the metabolism of either drug. However, the expression of UGT1A3 in the duodenum was significantly downregulated. The reduction in faecal enzymatic activity, observed during and after antibiotic administration, did not alter the ex vivo metabolism of olanzapine or risperidone. The relative abundance of Alistipes significantly correlated with the AUC of olanzapine but not risperidone. Interpretation Alistipes may play a role in the observed alterations in olanzapine pharmacokinetics. The gut microbiome might be an important variable determining the systemic bioavailability of orally administered olanzapine. Additional research exploring the potential implication of the gut microbiota on the clinical pharmacokinetics of olanzapine in humans is warranted. Funding This research is supported by APC Microbiome Ireland, a research centre funded by Science Foundation Ireland (SFI), through the Irish Government's National Development Plan (grant no. 12/RC/2273 P2) and by Nature Research-Yakult (The Global Grants for Gut Health; Ref No. 626891).
Collapse
Affiliation(s)
- Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Jacinta Walsh
- APC Microbiome Ireland, University College Cork, Cork, Ireland; School of Pharmacy, University College Cork, Cavanagh Pharmacy Building, Cork, Ireland
| | - Anna V Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Alexander V Zhdanov
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Conall R Strain
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County, Cork, Ireland
| | - Fiona Fouhy
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Niall P Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Brendan T Griffin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; School of Pharmacy, University College Cork, Cavanagh Pharmacy Building, Cork, Ireland.
| |
Collapse
|
5
|
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: 7] [Impact Index Per Article: 1.4] [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
|
6
|
Identification and interspecies characterization of UDP-glucuronosyltransferase isoforms catalyzing acacetin glucuronidation using recombinant UGT enzymes and microsomes. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2019. [DOI: 10.1016/j.jtcms.2019.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
7
|
Ge Y, Chen S, Mu W, Ba Q, Li J, Chen P, Wang X, Wang H. Epigenetic regulation of UDP-Glucuronosyltransferase by microRNA-200a/-183: implications for responses to sorafenib treatment in patients with hepatocellular carcinoma. Cancer Lett 2019; 454:14-25. [PMID: 30910587 DOI: 10.1016/j.canlet.2019.03.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 10/27/2022]
Abstract
Patients receiving sorafenib treatment for hepatocellular carcinoma (HCC) experience different treatment efficacy. Personalized sorafenib treatment should be achieved through the identification of predictors of therapeutic response. In the current study, we found that high UGT1A9 expression indicated better prognosis for HCC patients treated with sorafenib after surgery. In silico analysis predicted microRNA-200a/-183 as potential regulators of the UGT1A gene family via binding to the shared UGT1A9 3'-UTR. A significant inverse correlation between microRNA-200a/-183 and UGT1A9 mRNA level was observed in a panel of HCC specimens. Direct binding was further demonstrated by luciferase reporter gene vector carrying wild-type or binding site truncated UGT1A9 3'-UTR. MicroRNA-200a/-183 downregulated UGT1A9 expression in a dose-dependent manner and significantly reduced sorafenib β-D-glucuronide formation in HCC cells. These data indicated that UGT1A9, under epigenetic regulation of microRNA-200a/-183, could predict patients who might benefit from adjuvant sorafenib treatment after surgery.
Collapse
Affiliation(s)
- Yang Ge
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuzhen Chen
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China; National Center for Liver Cancer, Second Military Medical University, Shanghai, 200438, China
| | - Wei Mu
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Ba
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jingquan Li
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Peizhan Chen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xianming Wang
- Department of General Surgery, Qianfoshan Hospital Affiliated to Shandong University, Shandong, 250014, China.
| | - Hui Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
8
|
Cho PJ, Kim JH, Lee HS, Kim JA, Lee S. Identification of specific UGT1A9-mediated glucuronidation of licoricidin in human liver microsomes. Biopharm Drug Dispos 2019; 40:94-98. [DOI: 10.1002/bdd.2169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/29/2018] [Accepted: 12/16/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Pil Joung Cho
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu 41566 Republic of Korea
| | - Ju-Hyun Kim
- College of Pharmacy; Yeungnam University; Gyeongsan 38541 Republic of Korea
| | - Hye Suk Lee
- BK21 Plus Team for Creative Leader Program for Pharmacomics-based Future, Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy; The Catholic University of Korea; Bucheon 14662 Republic of Korea
| | - Jeong Ah Kim
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu 41566 Republic of Korea
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu 41566 Republic of Korea
| |
Collapse
|
9
|
Pharmacokinetics, Tissue Distribution and Excretion of a Novel Diuretic (PU-48) in Rats. Pharmaceutics 2018; 10:pharmaceutics10030124. [PMID: 30096833 PMCID: PMC6160999 DOI: 10.3390/pharmaceutics10030124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/21/2018] [Accepted: 07/27/2018] [Indexed: 01/02/2023] Open
Abstract
Methyl 3-amino-6-methoxythieno [2,3-b] quinoline-2-carboxylate (PU-48) is a novel diuretic urea transporter inhibitor. The aim of this study is to investigate the profile of plasma pharmacokinetics, tissue distribution, and excretion by oral dosing of PU-48 in rats. Concentrations of PU-48 within biological samples are determined using a validated high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. After oral administration of PU-48 (3, 6, and 12 mg/kg, respectively) in self-nanomicroemulsifying drug delivery system (SNEDDS) formulation, the peak plasma concentrations (Cmax), and the area under the curve (AUC0⁻∞) were increased by the dose-dependent and linear manner, but the marked different of plasma half-life (t1/2) were not observed. This suggests that the pharmacokinetic profile of PU-48 prototype was first-order elimination kinetic characteristics within the oral three doses range in rat plasma. Moreover, the prototype of PU-48 was rapidly and extensively distributed into thirteen tissues, especially higher concentrations were detected in stomach, intestine, liver, kidney, and bladder. The total accumulative excretion of PU-48 in the urine, feces, and bile was less than 2%. This research is the first report on disposition via oral administration of PU-48 in rats, and it provides important information for further development of PU-48 as a diuretic drug candidate.
Collapse
|
10
|
Kahma H, Filppula AM, Neuvonen M, Tarkiainen EK, Tornio A, Holmberg MT, Itkonen MK, Finel M, Neuvonen PJ, Niemi M, Backman JT. Clopidogrel Carboxylic Acid Glucuronidation is Mediated Mainly by UGT2B7, UGT2B4, and UGT2B17: Implications for Pharmacogenetics and Drug-Drug Interactions . Drug Metab Dispos 2017; 46:141-150. [PMID: 29138287 DOI: 10.1124/dmd.117.078162] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/11/2017] [Indexed: 01/04/2023] Open
Abstract
The antiplatelet drug clopidogrel is metabolized to an acyl-β-d-glucuronide, which causes time-dependent inactivation of CYP2C8. Our aim was to characterize the UDP-glucuronosyltransferase (UGT) enzymes that are responsible for the formation of clopidogrel acyl-β-d-glucuronide. Kinetic analyses and targeted inhibition experiments were performed using pooled human liver and intestine microsomes (HLMs and HIMs, respectively) and selected human recombinant UGTs based on preliminary screening. The effects of relevant UGT polymorphisms on the pharmacokinetics of clopidogrel were evaluated in 106 healthy volunteers. UGT2B7 and UGT2B17 exhibited the greatest level of clopidogrel carboxylic acid glucuronidation activities, with a CLint,u of 2.42 and 2.82 µl⋅min-1⋅mg-1, respectively. Of other enzymes displaying activity (UGT1A3, UGT1A9, UGT1A10-H, and UGT2B4), UGT2B4 (CLint,u 0.51 µl⋅min-1⋅mg-1) was estimated to contribute significantly to the hepatic clearance. Nonselective UGT2B inhibitors strongly inhibited clopidogrel acyl-β-d-glucuronide formation in HLMs and HIMs. The UGT2B17 inhibitor imatinib and the UGT2B7 and UGT1A9 inhibitor mefenamic acid inhibited clopidogrel carboxylic acid glucuronidation in HIMs and HLMs, respectively. Incubation of clopidogrel carboxylic acid in HLMs with UDPGA and NADPH resulted in strong inhibition of CYP2C8 activity. In healthy volunteers, the UGT2B17*2 deletion allele was associated with a 10% decrease per copy in the plasma clopidogrel acyl-β-d-glucuronide to clopidogrel carboxylic acid area under the plasma concentration-time curve from 0 to 4 hours (AUC0-4) ratio (P < 0.05). To conclude, clopidogrel carboxylic acid is metabolized mainly by UGT2B7 and UGT2B4 in the liver and by UGT2B17 in the small intestinal wall. The formation of clopidogrel acyl-β-d-glucuronide is impaired in carriers of the UGT2B17 deletion. These findings may have implications regarding the intracellular mechanisms leading to CYP2C8 inactivation by clopidogrel.
Collapse
Affiliation(s)
- Helinä Kahma
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Anne M Filppula
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - E Katriina Tarkiainen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Mikko T Holmberg
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Matti K Itkonen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Moshe Finel
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Pertti J Neuvonen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, and Helsinki University Hospital (H.K., A.M.F., M.Ne., E.K.T., A.T., M.T.H., M.K.I., P.J.N., M.Ni., J.T.B.) and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki (M.F.), Helsinki, Finland
| |
Collapse
|
11
|
Oda S, Kato Y, Hatakeyama M, Iwamura A, Fukami T, Kume T, Yokoi T, Nakajima M. Evaluation of Expression and Glycosylation Status of UGT1A10 in Supersomes and Intestinal Epithelial Cells with a Novel Specific UGT1A10 Monoclonal Antibody. Drug Metab Dispos 2017; 45:1027-1034. [DOI: 10.1124/dmd.117.075291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/29/2017] [Indexed: 12/15/2022] Open
|
12
|
Abstract
Since more than 70% of clinically used drugs are excreted from the body through metabolic processes, drug metabolism is a key determinant of pharmacokinetics, drug response and drug toxicity. Much progress has been made in understanding drug-drug interactions via the inhibition or induction of cytochrome P450s (P450, CYP), as well as the effects of genetic polymorphisms of P450s on pharmacokinetics, and this has facilitated the progress of optimized pharmacotherapy in the clinic. Now, similar information is needed for non-CYP enzymes, especially concerning Phase I enzymes, based on advanced basic and clinical studies. Recently, it was revealed that post-transcriptional regulation by microRNAs or RNA editing plays a significant role in regulating the expression of drug-metabolizing enzymes, thus conferring variability in the detoxification and metabolic activation of drugs or chemicals. Changes in the expression profile of microRNAs in tissues or body fluids can be a biomarker of drug response and toxicity; therefore, such studies could also be useful for drug repositioning. In addition, microRNAs are involved in pharmacogenetics, because single nucleotide polymorphisms in microRNA binding sites of mRNAs, or microRNAs themselves, may cause changes in gene expression. Some microRNA-related polymorphisms could be biomarkers of the clinical outcome of pharmacotherapy. In this review article, recent progress and future directions for drug metabolism studies are discussed.
Collapse
Affiliation(s)
- Miki Nakajima
- Faculty of Pharmaceutical Sciences, Kanazawa University
| |
Collapse
|
13
|
Lu D, Wang S, Xie Q, Guo L, Wu B. Transcriptional Regulation of Human UDP-Glucuronosyltransferase 2B10 by Farnesoid X Receptor in Human Hepatoma HepG2 Cells. Mol Pharm 2017; 14:2899-2907. [PMID: 28267333 DOI: 10.1021/acs.molpharmaceut.6b01103] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Little is known about transcriptional regulators of UDP-glucuronosyltransferase 2B10 (UGT2B10), an enzyme known to glucuronidate many chemicals and drugs such as nicotine and tricyclic antidepressants. Here, we uncovered that UGT2B10 was transcriptionally regulated by farnesoid X receptor (FXR), the bile acid sensing nuclear receptor. GW4064 and chenodeoxycholic acid (two specific FXR agonists) treatment of HepG2 cells led to a significant increase in the mRNA level of UGT2B10. The treated cells also showed enhanced glucuronidation activities toward amitriptyline (an UGT2B10 probe substrate). In reporter gene assays, the extent of UGT2B10 activation by the FXR agonists was positively correlated with the amount of cotransfected FXR. Consistently, knockdown of FXR by shRNA attenuated the induction effect on UGT2B10 expression. Furthermore, a combination of electrophoretic mobility shift assay and chromatin immunoprecipitation showed that the FXR receptor trans-activated UGT2B10 through its specific binding to the -209- to -197-bp region (an IR1 element) of the UGT2B10 promoter. In summary, our results for the first time established FXR as a transcriptional regulator of human UGT2B10.
Collapse
Affiliation(s)
- Danyi Lu
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Shuai Wang
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Qian Xie
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Lianxia Guo
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Baojian Wu
- Division of Pharmaceutics, College of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
14
|
Troberg J, Järvinen E, Ge GB, Yang L, Finel M. UGT1A10 Is a High Activity and Important Extrahepatic Enzyme: Why Has Its Role in Intestinal Glucuronidation Been Frequently Underestimated? Mol Pharm 2016; 14:2875-2883. [DOI: 10.1021/acs.molpharmaceut.6b00852] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Johanna Troberg
- Division
of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland
| | - Erkka Järvinen
- Division
of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland
| | - Guang-Bo Ge
- Laboratory
of Pharmaceutical Resource Discovery, Dalian Institute of Chemical
Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Ling Yang
- Laboratory
of Pharmaceutical Resource Discovery, Dalian Institute of Chemical
Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Moshe Finel
- Division
of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland
| |
Collapse
|
15
|
Sakakibara Y, Katoh M, Kondo Y, Nadai M. Effects of β-Naphthoflavone on Ugt1a6 and Ugt1a7 Expression in Rat Brain. Biol Pharm Bull 2016; 39:78-83. [PMID: 26725430 DOI: 10.1248/bpb.b15-00578] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Uridine 5'-diphosphate-glucuronosyltransferase (UGT) catalyzes a major phase II reaction in a drug-metabolizing enzyme system. Although the UGT1A subfamily is expressed mainly in the liver, it is also expressed in the brain. The purpose of the present study was to elucidate the effect of β-naphthoflavone (BNF), one of the major inducers of drug-metabolizing enzymes, on Ugt1a6 and Ugt1a7 mRNA expression and their glucuronidation in the rat brain. Eight-week-old male Sprague-Dawley rats were treated intraperitoneally with BNF (80 mg/kg), once daily for 7 d. Ugt1a6 and Ugt1a7 mRNA expression increased in the cerebellum and hippocampus (Ugt1a6: 2.1- and 2.3-fold, respectively; Ugt1a7: 1.7- and 2.8-fold, respectively); acetaminophen glucuronidation also increased in the same regions by 4.1- and 2.7-fold, respectively. BNF induced Ugt1a6 and Ugt1a7 mRNA expression and their glucuronidation, and the degree of induction differed among 9 regions. BNF also upregulated CYP1A1, CYP1A2, and CYP1B1 mRNAs in the rat brain. Since the aryl hydrocarbon receptor signaling pathway was activated by BNF, it is indicated that Ugt1a6 and Ugt1a7 were induced via AhR in the rat brain. This study clarified that Ugt1a6 and Ugt1a7 mRNA expression and their enzyme activities were altered by BNF, suggesting that these changes may lead to alteration in the pharmacokinetics of UGT substrate in rat brain.
Collapse
|
16
|
Sakakibara Y, Katoh M, Kondo Y, Nadai M. Effects of Phenobarbital on Expression of UDP-Glucuronosyltransferase 1a6 and 1a7 in Rat Brain. ACTA ACUST UNITED AC 2015; 44:370-7. [PMID: 26684499 DOI: 10.1124/dmd.115.067439] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/11/2015] [Indexed: 11/22/2022]
Abstract
UDP-glucuronosyltransferase (UGT), a phase II drug-metabolizing enzyme, is expressed in the brain and can catalyze glucuronidation of endogenous and exogenous substrates in the brain. Thus, changes in UGT1A expression could affect homeostasis and drug efficacy. Phenobarbital (PB), a typical inducer of drug-metabolizing enzymes, has been reported to induce oxidative stress and epigenetic changes, which could alter UGT expression in the brain. Here, we aimed to clarify the effects of PB on Ugt1a6 and Ugt1a7 gene expression in rat brains. Sprague-Dawley rats were treated intraperitoneally with PB (80 mg/kg), once daily for 7 days. Ugt1a6 and Ugt1a7 mRNA expression levels were increased in the striatum and thalamus (Ugt1a6, 3.0- and 2.9-fold, respectively; Ugt1a7, 2.6- and 2.6-fold, respectively). Acetaminophen glucuronidation was also increased in the medulla oblongata and thalamus by 1.8- and 1.2-fold, respectively. The induction rates within different brain regions were correlated with Ugt1a6 and Ugt1a7 mRNA expression, and the degree of induction also correlated with that of NF-E2-related factor-2 mRNA. Measurement of oxidative stress markers suggested that PB induced oxidative stress in brain regions in which Ugt1a6 and Ugt1a7 mRNAs were increased. Moreover, histone modifications were altered by PB treatment, resulting in increased histone H3 lysine 4 trimethylation in the striatum and thalamus and decreased histone H3 lysine 9 trimethylation in the thalamus. These results suggested that oxidative stress and histone modifications may promote transcriptional activation of Ugt1a6 and Ugt1a7 genes. In summary, Ugt1a6 and Ugt1a7 mRNA levels were increased by PB treatment, which may alter pharmacokinetics in the brain.
Collapse
Affiliation(s)
- Yukiko Sakakibara
- Pharmaceutics, Faculty of Pharmacy, Meijo University; 150 Yagotoyama, Tenpaku-ku, Nagoya 468-8503, Japan
| | - Miki Katoh
- Pharmaceutics, Faculty of Pharmacy, Meijo University; 150 Yagotoyama, Tenpaku-ku, Nagoya 468-8503, Japan
| | - Yuya Kondo
- Pharmaceutics, Faculty of Pharmacy, Meijo University; 150 Yagotoyama, Tenpaku-ku, Nagoya 468-8503, Japan
| | - Masayuki Nadai
- Pharmaceutics, Faculty of Pharmacy, Meijo University; 150 Yagotoyama, Tenpaku-ku, Nagoya 468-8503, Japan
| |
Collapse
|
17
|
Dluzen DF, Lazarus P. MicroRNA regulation of the major drug-metabolizing enzymes and related transcription factors. Drug Metab Rev 2015; 47:320-34. [PMID: 26300547 PMCID: PMC6309899 DOI: 10.3109/03602532.2015.1076438] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Identifying novel mechanisms contributing to patient variability of drug response is a major goal of personalized medicine. Epigenetic regulation of gene expression by microRNA (miRNA) impacts a broad range of cellular processes, but knowledge of its regulation of drug-metabolizing enzymes (DMEs) is more limited. This review provides an introduction to miRNA and their functionality and summarizes known miRNA regulation of DME families, including the cytochrome P450s, UDP-glucuronoslytransferases, glutathione-S-transferases, sulfotransferases and aldo-keto reductases, and the transcription factors known to be involved in DME regulation.
Collapse
Affiliation(s)
- Douglas F Dluzen
- a Laboratory of Epidemiology and Population Sciences , National Institutes of Health , Baltimore , MD , USA and
| | - Philip Lazarus
- b Department of Pharmaceutical Sciences , Washington State University , Spokane , WA , USA
| |
Collapse
|
18
|
Wu Z, Zhang X, Ma Z, Wu B. Establishment of pharmacophore and VolSurf models to predict the substrates of UDP-glucuronosyltransferase1A3. Xenobiotica 2015; 45:653-62. [DOI: 10.3109/00498254.2015.1016136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
19
|
Oda S, Fukami T, Yokoi T, Nakajima M. A comprehensive review of UDP-glucuronosyltransferase and esterases for drug development. Drug Metab Pharmacokinet 2015; 30:30-51. [DOI: 10.1016/j.dmpk.2014.12.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/24/2014] [Accepted: 12/02/2014] [Indexed: 01/24/2023]
|
20
|
Manevski N, Swart P, Balavenkatraman KK, Bertschi B, Camenisch G, Kretz O, Schiller H, Walles M, Ling B, Wettstein R, Schaefer DJ, Itin P, Ashton-Chess J, Pognan F, Wolf A, Litherland K. Phase II metabolism in human skin: skin explants show full coverage for glucuronidation, sulfation, N-acetylation, catechol methylation, and glutathione conjugation. Drug Metab Dispos 2014; 43:126-39. [PMID: 25339109 DOI: 10.1124/dmd.114.060350] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although skin is the largest organ of the human body, cutaneous drug metabolism is often overlooked, and existing experimental models are insufficiently validated. This proof-of-concept study investigated phase II biotransformation of 11 test substrates in fresh full-thickness human skin explants, a model containing all skin cell types. Results show that skin explants have significant capacity for glucuronidation, sulfation, N-acetylation, catechol methylation, and glutathione conjugation. Novel skin metabolites were identified, including acyl glucuronides of indomethacin and diclofenac, glucuronides of 17β-estradiol, N-acetylprocainamide, and methoxy derivatives of 4-nitrocatechol and 2,3-dihydroxynaphthalene. Measured activities for 10 μM substrate incubations spanned a 1000-fold: from the highest 4.758 pmol·mg skin(-1)·h(-1) for p-toluidine N-acetylation to the lowest 0.006 pmol·mg skin(-1)·h(-1) for 17β-estradiol 17-glucuronidation. Interindividual variability was 1.4- to 13.0-fold, the highest being 4-methylumbelliferone and diclofenac glucuronidation. Reaction rates were generally linear up to 4 hours, although 24-hour incubations enabled detection of metabolites in trace amounts. All reactions were unaffected by the inclusion of cosubstrates, and freezing of the fresh skin led to loss of glucuronidation activity. The predicted whole-skin intrinsic metabolic clearances were significantly lower compared with corresponding whole-liver intrinsic clearances, suggesting a relatively limited contribution of the skin to the body's total systemic phase II enzyme-mediated metabolic clearance. Nevertheless, the fresh full-thickness skin explants represent a suitable model to study cutaneous phase II metabolism not only in drug elimination but also in toxicity, as formation of acyl glucuronides and sulfate conjugates could play a role in skin adverse reactions.
Collapse
Affiliation(s)
- Nenad Manevski
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Piet Swart
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Kamal Kumar Balavenkatraman
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Barbara Bertschi
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Gian Camenisch
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Olivier Kretz
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Hilmar Schiller
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Markus Walles
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Barbara Ling
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Reto Wettstein
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Peter Itin
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Joanna Ashton-Chess
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Francois Pognan
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Armin Wolf
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Karine Litherland
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| |
Collapse
|
21
|
Ito Y, Fukami T, Yokoi T, Nakajima M. An orphan esterase ABHD10 modulates probenecid acyl glucuronidation in human liver. Drug Metab Dispos 2014; 42:2109-16. [PMID: 25217485 DOI: 10.1124/dmd.114.059485] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Probenecid, a widely used uricosuric agent, is mainly metabolized to probenecid acyl glucuronide (PRAG), which is considered a causal substance of severe allergic or anaphylactoid reactions. PRAG can be hydrolyzed (deglucuronidated) to probenecid. The purpose of this study was to identify enzymes responsible for probenecid acyl glucuronidation and PRAG deglucuronidation in human livers and to examine the effect of deglucuronidation in PRAG formation. In human liver homogenates (HLHs), the intrinsic clearance (CLint) of PRAG deglucuronidation was much greater (497-fold) than that of probenecid acyl glucuronidation. Evaluation of PRAG formation by recombinant UDP-glucuronosyltransferase (UGT) isoforms and an inhibition study using HLHs as an enzyme source demonstrated that multiple UGT isoforms, including UGT1A1, UGT1A9, and UGT2B7, catalyzed probenecid acyl glucuronidation. We found that recombinant α/β hydrolase domain containing 10 (ABHD10) substantially catalyzed PRAG deglucuronidation activity, whereas carboxylesterases did not. Similar inhibitory patterns by chemicals between HLHs and recombinant ABHD10 supported the major contribution of ABHD10 to PRAG deglucuronidation in human liver. Interestingly, it was demonstrated that the CLint value of probenecid acyl glucuronidation in HLHs was increased by 1.7-fold in the presence of phenylmethylsulfonyl fluoride, which potently inhibited ABHD10 activity. In conclusion, we found that PRAG deglucuronidation catalyzed by ABHD10 suppressively regulates PRAG formation via multiple UGT enzymes in human liver. The balance of activities by these enzymes is important for the formation of PRAG, which may be associated with the adverse reactions observed after probenecid administration.
Collapse
Affiliation(s)
- Yusuke Ito
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Tsuyoshi Yokoi
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| |
Collapse
|
22
|
Wang H, Bian T, Jin T, Chen Y, Lin A, Chen C. Association analysis of UGT1A genotype and haplotype with SN-38 glucuronidation in human livers. Pharmacogenomics 2014; 15:785-98. [PMID: 24897286 DOI: 10.2217/pgs.14.29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. This study comprehensively investigates the effects of UGT1A1 and UGT1A9 genetic polymorphism on SN-38 glucuronidation activity. MATERIALS & METHODS Genetic polymorphisms and combinational haplotypes of UGT1A1 and UGT1A9, SN-38 glucuronidation activities, and protein levels of UGT1A1 and UGT1A9 were determined using a set of over 45 Chinese livers. RESULTS UGT1A1 reduced function variants UGT1A1*6, *28, *60 and *1B exhibited additive effect. The number of UGT1A1 reduced function alleles was associated with decreased SN-38G formation rates and UGT1A protein levels. UGT1A9 I399C>T and UGT1A9*1b, which were highly linked, were associated with increased SN-38 glucuronidation activity and UGT1A protein levels. However, further analysis based on UGT1A9-1A1 haplotypes confirmed that their increased effect was partly due to their close linkage with UGT1A1 reduced function alleles. CONCLUSION UGT1A1 genetic polymorphisms have a more important function in human liver SN-38 glucuronidation activity than UGT1A9. Original submitted 7 November 2013; Revision submitted 30 January 2014.
Collapse
Affiliation(s)
- Huijuan Wang
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, 229 North Taibai Road, Xi'an 710069, China.
| | | | | | | | | | | |
Collapse
|
23
|
Gundert-Remy U, Bernauer U, Blömeke B, Döring B, Fabian E, Goebel C, Hessel S, Jäckh C, Lampen A, Oesch F, Petzinger E, Völkel W, Roos PH. Extrahepatic metabolism at the body's internal–external interfaces. Drug Metab Rev 2014; 46:291-324. [DOI: 10.3109/03602532.2014.900565] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Yamamoto K, Mukai M, Nagaoka K, Hayashi K, Hichiya H, Okada K, Murata M, Shigeyama M, Narimatsu S, Hanioka N. Functional characterization of cynomolgus monkey UDP-glucuronosyltransferase 1A9. Eur J Drug Metab Pharmacokinet 2014; 39:195-202. [DOI: 10.1007/s13318-014-0177-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 01/09/2014] [Indexed: 10/25/2022]
|
25
|
Dluzen DF, Sun D, Salzberg AC, Jones N, Bushey RT, Robertson GP, Lazarus P. Regulation of UDP-glucuronosyltransferase 1A1 expression and activity by microRNA 491-3p. J Pharmacol Exp Ther 2014; 348:465-77. [PMID: 24399855 DOI: 10.1124/jpet.113.210658] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The UDP-glucuronosyltransferase (UGT) 1A enzymes are involved in the phase II metabolism of many important endogenous and exogenous compounds. The nine UGT1A isoforms exhibit high interindividual differences in expression, but their epigenetic regulation is not well understood. The purpose of the present study was to examine microRNA (miRNA) regulation of hepatic UGT1A enzymes and determine whether or not that regulation impacts enzymatic activity. In silico analysis identified miRNA 491-3p (miR-491-3p) as a potential regulator of the UGT1A gene family via binding to the shared UGT1A 3'-untranslated region common to all UGT1A enzymes. Transfection of miR-491-3p mimic into HuH-7 cells significantly repressed UGT1A1 (P < 0.001), UGT1A3 (P < 0.05), and UGT1A6 (P < 0.05) mRNA levels. For UGT1A1, this repression correlated with significantly reduced metabolism of raloxifene into raloxifene-6-glucuronide (ral-6-gluc; P < 0.01) and raloxifene-4'-glucuronide (ral-4'-gluc; P < 0.01). In HuH-7 cells with repressed miR-491-3p expression, there was a significant increase (~80%; P < 0.01) in UGT1A1 mRNA and a corresponding increase in glucuronidation of raloxifene into ral-6-gluc (50%; P < 0.05) and ral-4'-gluc (22%; P < 0.01). Knockdown of endogenous miR-491-3p in HepG2 cells did not significantly alter UGT1A1 mRNA levels but did increase the formation of ral-6-gluc (50%; P < 0.05) and ral-4'-gluc (34%; P < 0.001). A significant inverse correlation between miR-491-3p expression and both UGT1A3 (P < 0.05) and UGT1A6 (P < 0.01) mRNA levels was observed in a panel of normal human liver specimens, with a significant (P < 0.05) increase in UGT1A3 and UGT1A6 mRNA levels observed in miR-491-3p nonexpressing versus expressing liver specimens. These results suggest that miR-491-3p is an important factor in regulating the expression of UGT1A enzymes in vivo.
Collapse
Affiliation(s)
- Douglas F Dluzen
- Departments of Pharmacology (D.F.D., D.S., N.J., R.T.B., G.P.R., P.L.) and Public Health Sciences (A.C.S.), Penn State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington (P.L.)
| | | | | | | | | | | | | |
Collapse
|
26
|
Grosse L, Campeau AS, Caron S, Morin FA, Meunier K, Trottier J, Caron P, Verreault M, Barbier O. Enantiomer selective glucuronidation of the non-steroidal pure anti-androgen bicalutamide by human liver and kidney: role of the human UDP-glucuronosyltransferase (UGT)1A9 enzyme. Basic Clin Pharmacol Toxicol 2013; 113:92-102. [PMID: 23527766 DOI: 10.1111/bcpt.12071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/14/2013] [Indexed: 11/30/2022]
Abstract
Bicalutamide (Casodex(®) ) is a non-steroidal pure anti-androgen used in the treatment of localized prostate cancer. It is a racemate drug, and its activity resides in the (R)-enantiomer, with little in the (S)-enantiomer. A major metabolic pathway for bicalutamide is glucuronidation catalysed by UDP-glucuronosyltransferase (UGT) enzymes. While (S)bicalutamide is directly glucuronidated, (R)bicalutamide requires hydroxylation prior to glucuronidation. The contribution of human tissues and UGT isoforms in the metabolism of these enantiomers has not been extensively investigated. In this study, both (R) and/or (S)bicalutamide were converted into glucuronide (-G) derivatives after incubation of pure and racemic solutions with microsomal extracts from human liver and kidney. Intestinal microsomes exhibited only low reactivity with these substrates. Km values of liver and kidney samples for (S)bicalutamide glucuronidation were similar, and lower than values obtained with the (R)-enantiomer. Among the 16 human UGTs tested, UGT1A8 and UGT1A9 were able to form both (S) and (R)bicalutamide-G from pure or racemic substrates. UGT2B7 was also able to form (R)bicalutamide-G. Kinetic parameters of the recombinant UGT2B7, UGT1A8 and UGT1A9 enzymes support a predominant role of the UGT1A9 isoform in bicalutamide metabolism. Accordingly, (S)bicalutamide inhibited the ability of human liver and kidney microsomes to glucuronidate the UGT1A9 probe substrate, propofol. In conclusion, the present study provides the first comprehensive analysis of in vitro bicalutamide glucuronidation by human tissues and UGTs and identifies UGT1A9 as a major contributor for (R) and (S) glucuronidation in the human liver and kidney.
Collapse
Affiliation(s)
- Laurent Grosse
- Laboratory of molecular pharmacology, CHU-Québec Research Centre and the Faculty of pharmacy, Laval University, Québec, QC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kato Y, Izukawa T, Oda S, Fukami T, Finel M, Yokoi T, Nakajima M. Human UDP-Glucuronosyltransferase (UGT) 2B10 in DrugN-Glucuronidation: Substrate Screening and Comparison with UGT1A3 and UGT1A4. Drug Metab Dispos 2013; 41:1389-97. [DOI: 10.1124/dmd.113.051565] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
28
|
Du J, You T, Chen X, Zhong D. Stereoselective Glucuronidation of Ornidazole in Humans: Predominant Contribution of UDP-Glucuronosyltransferases 1A9 and 2B7. Drug Metab Dispos 2013; 41:1306-18. [DOI: 10.1124/dmd.113.051235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
29
|
Zhang H, Patana AS, Mackenzie PI, Ikushiro S, Goldman A, Finel M. Human UDP-Glucuronosyltransferase Expression in Insect Cells: Ratio of Active to Inactive Recombinant Proteins and the Effects of a C-Terminal His-Tag on Glucuronidation Kinetics. Drug Metab Dispos 2012; 40:1935-44. [DOI: 10.1124/dmd.112.046086] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|