1
|
Zhang H, Yang L, Shen D, Zhu Y, Zhang L. Identification of Bromophenols' glucuronidation and its induction on UDP- glucuronosyltransferases isoforms. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116281. [PMID: 38581907 DOI: 10.1016/j.ecoenv.2024.116281] [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: 01/28/2024] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/08/2024]
Abstract
Bromophenols (BPs) are prominent environmental pollutants extensively utilized in aquaculture, pharmaceuticals, and chemical manufacturing. This study aims to identify UDP- glucuronosyltransferases (UGTs) isoforms involved in the metabolic elimination of BPs. Mono-glucuronides of BPs were detected in human liver microsomes (HLMs) incubated with the co-factor uridine-diphosphate glucuronic acid (UDPGA). The glucuronidation metabolism reactions catalyzed by HLMs followed Michaelis-Menten or substrate inhibition kinetics. Recombinant enzymes and inhibition experiments with chemical reagents were employed to phenotype the principal UGT isoforms participating in BP glucuronidation. UGT1A6 emerged as the major enzyme in the glucuronidation of 4-Bromophenol (4-BP), while UGT1A1, UGT1A6, and UGT1A8 were identified as the most essential isoforms for metabolizing 2,4-dibromophenol (2,4-DBP). UGT1A1, UGT1A8, and UGT2B4 were deemed the most critical isoforms in the catalysis of 2,4,6-tribromophenol (2,4,6-TBP) glucuronidation. Species differences were investigated using the liver microsomes of pig (PLM), rat (RLM), monkey (MyLM), and dog (DLM). Additionally, 2,4,6-TBP effects on the expression of UGT1A1 and UGT2B7 in HepG2 cells were evaluated. The results demonstrated potential induction of UGT1A1 and UGT2B7 upon exposure to 2,4,6-TBP at a concentration of 50 μM. Collectively, these findings contribute to elucidating the metabolic elimination and toxicity of BPs.
Collapse
Affiliation(s)
- Haoqian Zhang
- Department of Obstetrics and Gynecology, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Cervical Disease, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China; National Clinical Research Center for Obstetrics and Gynecology, Henan Branch, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Cervical Disease, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China; National Clinical Research Center for Obstetrics and Gynecology, Henan Branch, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dandan Shen
- Department of Obstetrics and Gynecology, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Cervical Disease, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China; National Clinical Research Center for Obstetrics and Gynecology, Henan Branch, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanhang Zhu
- Zhengzhou Key Laboratory of Cervical Disease, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China; National Clinical Research Center for Obstetrics and Gynecology, Henan Branch, The third Affliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lihua Zhang
- Department of Pediatric Urology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2
|
Ye W, Wang Z, Lv X, Yin H, Jiang L, Wang Z, Liu Y. Potential risk of drug-drug interactions of ponatinib via inhibition against human UDP-glucuronosyltransferases. Toxicol In Vitro 2023; 92:105664. [PMID: 37597759 DOI: 10.1016/j.tiv.2023.105664] [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: 03/15/2023] [Revised: 07/10/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Ponatinib is an efficient oral tyrosine kinase inhibitor (TKI) for T315I-positive Ph + ALL and T315I-positive chronic myeloid leukemia (CML) or BCR-ABL when no other TKIs can be prescribed. In this research, we evaluated the inhibitory effects of ponatinib on human recombinant UDP-glucuronosyltransferases (UGTs) and predicted the magnitude of potential drug-drug interaction (DDI) risk of co-treatment with ponatinib and UGTs substrates by using in vitro-in vivo extrapolation (IVIVE) method. Our study presented that ponatinib showed a broad-spectrum inhibition against UGTs. Particularly, ponatinib exhibited potent inhibitory effects towards UGT1A7, UGT1A1, and UGT1A9 with IC50 values of 0.37, 0.41, and 0.89 μM, respectively, which might lead to clinically significant DDI.
Collapse
Affiliation(s)
- Weiyi Ye
- 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
| | - Hang Yin
- 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
| | - Zhe Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
3
|
Onder A, Trendafilova A. A REVIEW ON ANOMALIN: A NATURAL BIOACTIVE PYRANOCOUMARIN FROM THE PAST TO THE FUTURE. Chem Biodivers 2022; 19:e202200167. [PMID: 35544308 DOI: 10.1002/cbdv.202200167] [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: 02/21/2022] [Accepted: 05/10/2022] [Indexed: 11/09/2022]
Abstract
Anomalin is a seselin-type pyranocoumarin isolated for the first time from Angelica anomala Avé-Lal, but is also found in several other plant species, especially in Apiaceae. This lipophilic molecule possesses pharmacologically beneficial activities for human health. The major scientific databases Scopus, Web of Science, Google Scholar, and PubMed up to the end of 2021 and the combining terms anomalin, praeruptorin, isolation, structure elucidation, and biological activity were used in the research of this review. This review focuses on the sources, structural properties, and biological functions of anomalin and provides future trends in the investigation of anomalin, particularly in therapies for many common diseases such as anti-inflammatory and neurodegenerative illnesses. As a potential bioactive molecule, prospective studies on anomalin should be done through supported clinical trials. At the end, this review confirms the significant pharmacological potential of anomalin.
Collapse
Affiliation(s)
- Alev Onder
- Ankara University Faculty of Pharmacy: Ankara Universitesi Eczacilik Fakultesi, Department of Pharmacognosy, Tandoğan, 06100, Ankara, BULGARIA
| | - Antoaneta Trendafilova
- Bulgarian Academy of Sciences: B'lgarska akademia na naukite, Institute of Organic Chemistry with Centre of Phytochemistry, Acad. G. Bonchev, bl. 9, Not Available, 1113, Sofia, BULGARIA
| |
Collapse
|
4
|
Dmitriev AV, Lagunin AA, Karasev DА, Rudik AV, Pogodin PV, Filimonov DA, Poroikov VV. Prediction of Drug-Drug Interactions Related to Inhibition or Induction of Drug-Metabolizing Enzymes. Curr Top Med Chem 2019; 19:319-336. [PMID: 30674264 DOI: 10.2174/1568026619666190123160406] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/02/2019] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Drug-drug interaction (DDI) is the phenomenon of alteration of the pharmacological activity of a drug(s) when another drug(s) is co-administered in cases of so-called polypharmacy. There are three types of DDIs: pharmacokinetic (PK), pharmacodynamic, and pharmaceutical. PK is the most frequent type of DDI, which often appears as a result of the inhibition or induction of drug-metabolising enzymes (DME). In this review, we summarise in silico methods that may be applied for the prediction of the inhibition or induction of DMEs and describe appropriate computational methods for DDI prediction, showing the current situation and perspectives of these approaches in medicinal and pharmaceutical chemistry. We review sources of information on DDI, which can be used in pharmaceutical investigations and medicinal practice and/or for the creation of computational models. The problem of the inaccuracy and redundancy of these data are discussed. We provide information on the state-of-the-art physiologically- based pharmacokinetic modelling (PBPK) approaches and DME-based in silico methods. In the section on ligand-based methods, we describe pharmacophore models, molecular field analysis, quantitative structure-activity relationships (QSAR), and similarity analysis applied to the prediction of DDI related to the inhibition or induction of DME. In conclusion, we discuss the problems of DDI severity assessment, mention factors that influence severity, and highlight the issues, perspectives and practical using of in silico methods.
Collapse
Affiliation(s)
| | - Alexey A Lagunin
- Institute of Biomedical Chemistry, Moscow, Russian Federation.,Pirogov Russian National Research Medical University, Moscow, RussiaN Federation
| | | | | | - Pavel V Pogodin
- Institute of Biomedical Chemistry, Moscow, Russian Federation
| | | | | |
Collapse
|
5
|
Du Z, Wang G, Cao YF, Hu CM, Yang K, Liu YZ, Zhang CZ, Zhang WH, Zhu ZT, Sun HZ, Sun XY, Hong M, Fang ZZ. Everolimus-inhibited multiple isoforms of UDP-glucuronosyltransferases (UGTs). Xenobiotica 2017; 48:452-458. [DOI: 10.1080/00498254.2017.1335917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Zuo Du
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
- National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China,
| | - Yun-Feng Cao
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou, Liaoning, China,
| | - Cui-Min Hu
- Tianjin Life Science Research Center, Department of Microbiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China,
| | - Kun Yang
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
- National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, China
| | - Yong-Zhe Liu
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
- National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, China
| | - Chun-Ze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China,
| | - Wei-Hua Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China,
| | - Zhi-Tu Zhu
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou, Liaoning, China,
| | - Hong-Zhi Sun
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou, Liaoning, China,
| | | | - Mo Hong
- RSKT Biopharma Inc, Liaoning, China, and
| | - Zhong-Ze Fang
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
- National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, China
| |
Collapse
|
6
|
Sun D, Zhang CZ, Ran RX, Cao YF, Du Z, Fu ZW, Huang CT, Zhao ZY, Zhang WH, Fang ZZ. In Vitro Comparative Study of the Inhibitory Effects of Mangiferin and Its Aglycone Norathyriol towards UDP-Glucuronosyl Transferase (UGT) Isoforms. Molecules 2017. [PMID: 28621744 PMCID: PMC6152678 DOI: 10.3390/molecules22061008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mangiferin (MGF), the predominant constituent of extracts of the mango plant Mangifera Indica L., has been investigated extensively because of its remarkable pharmacological effects. In vitro recombinant UGTs-catalyzed glucuronidation of 4-methylumbelliferone (4-MU) was used to investigate the inhibition of mangiferin and aglycone norathyriol towards various isoforms of UGTs in our study, which evaluated the inhibitory capacity of MGF and its aglycone norathyriol (NTR) towards UDP-glucuronosyltransferase (UGT) isoforms. Initial screening experiment showed that deglycosylation of MGF into NTR strongly increased the inhibitory effects towards almost all the tested UGT isoforms at a concentration of 100 μM. Kinetic experiments were performed to further characterize the inhibition of UGT1A3, UGT1A7 and UGT1A9 by NTR. NTR competitively inhibited UGT1A3, UGT1A7 and UGT1A9, with an IC50 value of 8.2, 4.4, and 12.3 μM, and a Ki value of 1.6, 2.0, and 2.8 μM, respectively. In silico docking showed that only NTR could dock into the activity cavity of UGT1A3, UGT1A7 and UGT1A9. The binding free energy of NTR to UGT1A3, 1A7, 1A9 were −7.4, −7.9 and −4.0 kcal/mol, respectively. Based on the inhibition evaluation standard ([I]/Ki < 0.1, low possibility; 0.1 < [I]/Ki < 1, medium possibility; [I]/Ki > 1, high possibility), an in vivo herb–drug interaction between MGF/NTR and drugs mainly undergoing UGT1A3-, UGT1A7- or UGT1A9-catalyzed metabolism might occur when the plasma concentration of NTR is above 1.6, 2.0 and 2.8 μM, respectively.
Collapse
Affiliation(s)
- Dan Sun
- College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Chun-Ze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, China.
| | - Rui-Xue Ran
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Yun-Feng Cao
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
| | - Zuo Du
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
- Department of Toxicology, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Zhi-Wei Fu
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
- Department of Toxicology, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Chun-Ting Huang
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
- Department of Toxicology, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Zhen-Ying Zhao
- Tianjin Union Medical Center, 190 Jieyuan Road, Hongqiao District, Tianjin 300121, China.
| | - Wei-Hua Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, China.
| | - Zhong-Ze Fang
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
- Department of Toxicology, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| |
Collapse
|
7
|
Evaluation and Comparison of the Inhibition Effect of Astragaloside IV and Aglycone Cycloastragenol on Various UDP-Glucuronosyltransferase (UGT) Isoforms. Molecules 2016; 21:molecules21121616. [PMID: 27916843 PMCID: PMC6274106 DOI: 10.3390/molecules21121616] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/08/2016] [Accepted: 11/22/2016] [Indexed: 12/17/2022] Open
Abstract
As one of the main active ingredients from Radix Astragali (RA), orally dosed astragaloside IV (AST) is easily transformed to sapogenin-cycloastragenol (CAG) by deglycosylation in the gastrointestinal tract. Because the potential adverse effects of AST and CAG remain unclear, the present study in this article was carried out to investigate the inhibition effects of AST and CAG on UDP-glucuronosyltransferases (UGTs) to explore potential clinical toxicity. An in vitro UGTs incubation mixture was employed to study the inhibition of AST and CAG towards UGT isoforms. Concentrations of 100 μM for each compound were used to initially screen the inhibitory efficiency. Deglycosylation of AST to CAG could strongly increase the inhibitory effects towards almost all of the tested UGT isoforms, with an IC50 of 0.84 μM and 11.28 μM for UGT1A8 and UGT2B7, respectively. Ulteriorly, the inhibition type and kinetics of CAG towards UGT1A8 and UGT2B7 were evaluated depending on the initial screening results. Data fitting using Dixon and Lineweaver-Burk plots demonstrated that CAG competitively inhibited UGT1A8 and noncompetitively inhibited UGT2B7. From the second plot drawn with the slopes from the Lineweaver-Burk plot versus the concentrations of CAG, the inhibition constant (Ki) was calculated to be 0.034 μM and 20.98 μM for the inhibition of UGT1A8 and UGT2B7, respectively. Based on the [I]/Ki standard ([I]/Ki < 0.1, low possibility; 1 > [I]/Ki > 0.1, medium possibility; [I]/Ki > 1, high possibility), it was successfully predicted here that an in vivo herb-drug interaction between AST/CAG and drugs mainly undergoing UGT1A8- or UGT2B7-catalyzed metabolism might occur when the plasma concentration of CAG is above 0.034 μM and 20.98 μM, respectively.
Collapse
|