1
|
Zheng R, Wang B, Wang J, Cheng X, Ye Q. Absorption, transportation, residual distribution of chiral 14C-dufulin in garlics and metabolism of its racemate. Food Chem 2025; 463:141151. [PMID: 39255711 DOI: 10.1016/j.foodchem.2024.141151] [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: 06/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
Dufulin is a novel chiral plant antiviral agent. In this study, we investigated the uptake, translocation and accumulation of 14C-dufulin stereoisomers in different tissues of garlic via leaf introduction and root uptake. The behavior of dufulin enantiomers in plants is not stereoselective, and dufulin is more likely to be absorbed by leaves than by roots. The metabolites of 14C-dufulin with high specific activity in garlic were qualitatively and quantitatively analyzed by HPLC-QTOF-MS, and the metabolic pathway involved was elucidated. In the leaf and bulb, dufulin underwent phase I and phase II metabolism and produced four metabolites. The ratios and concentrations of these four metabolites in the bulb, but not in the leaf, met the residue criterion. Overall, our results provide relatively accurate predictions for the risk assessment of dufulin, which will help guide its rational use and ensure its ecological safety and human health.
Collapse
Affiliation(s)
- Ruonan Zheng
- Zhejiang Key Laboratory of Drug Prevention and Control Technology, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China.
| | - Binjie Wang
- Zhejiang Key Laboratory of Drug Prevention and Control Technology, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China.
| | - Jiye Wang
- Zhejiang Key Laboratory of Drug Prevention and Control Technology, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China.
| | - Xi Cheng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture and Rural Affairs, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China.
| | - Qingfu Ye
- Institute of Nuclear Agricultural Sciences, Key Laboratory of Nuclear Agricultural Sciences of Ministry of Agriculture of the PRC and Zhejiang Province, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
2
|
González-Padilla D, Camara MD, Lauschke VM, Zhou Y. Population-scale variability of the human UDP-glycosyltransferase gene family. J Genet Genomics 2024; 51:1228-1236. [PMID: 38969258 DOI: 10.1016/j.jgg.2024.06.018] [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: 05/12/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024]
Abstract
Human UDP-glycosyltransferases (UGTs) are responsible for the glycosylation of a wide variety of endogenous substrates and commonly prescribed drugs. Different genetic polymorphisms in UGT genes are implicated in interindividual differences in drug response and cancer risk. However, the genetic complexity beyond these variants has not been comprehensively assessed. We here leveraged whole-exome and whole-genome sequencing data from 141,456 unrelated individuals across 7 major human populations to provide a comprehensive profile of genetic variability across the human UGT gene family. Overall, 9666 exonic variants were observed, of which 98.9% were rare. To interpret the functional impact of UGT missense variants, we developed a gene family-specific variant effect predictor. This algorithm identified a total of 1208 deleterious variants, most of which were found in African and South Asian populations. Structural analysis corroborated the predicted effects for multiple variations in substrate binding sites. Combined, our analyses provide a systematic overview of UGT variability, which can yield insights into interindividual differences in phase 2 metabolism and facilitate the translation of sequencing data into personalized predictions of UGT substrate disposition.
Collapse
Affiliation(s)
| | - Mahamadou D Camara
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden; Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tübingen, Tübingen, Germany.
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| |
Collapse
|
3
|
Hu DG, Marri S, Hulin JA, Ansaar R, Mackenzie PI, McKinnon RA, Meech R. Activation of Cryptic Donor Splice Sites within the UDP-Glucuronosyltransferase (UGT)1A First-Exon Region Generates Variant Transcripts That Encode UGT1A Proteins with Truncated Aglycone-Binding Domains. Drug Metab Dispos 2024; 52:526-538. [PMID: 38565302 DOI: 10.1124/dmd.123.001565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 02/19/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
The human UDP-glucuronosyltransferases (UGTs) have crucial roles in metabolizing and clearing numerous small lipophilic compounds. The UGT1A locus generates nine UGT1A mRNAs, 65 spliced transcripts, and 34 circular RNAs. In this study, our analysis of published UGT-RNA capture sequencing (CaptureSeq) datasets identified novel splice junctions that predict 24 variant UGT1A transcripts derived from ligation of exon 2 to unique sequences within the UGT1A first-exon region using cryptic donor splice sites. Of these variants, seven (1A1_n1, 1A3_n3, 1A4_n4, 1A5_n1, 1A8_n2, 1A9_n2, 1A10_n7) are predicted to encode UGT1A proteins with truncated aglycone-binding domains. We assessed their expression profiles and deregulation in cancer using four RNA sequencing (RNA-Seq) datasets of paired normal and cancerous drug-metabolizing tissues from large patient cohorts. Variants were generally coexpressed with their canonical counterparts with a higher relative abundance in tumor than in normal tissues. Variants showed tissue-specific expression with high interindividual variability but overall low abundance. However, 1A8_n2 showed high abundance in normal and cancerous colorectal tissues, with levels that approached or surpassed canonical 1A8 mRNA levels in many samples. We cloned 1A8_n2 and showed expression of the predicted protein (1A8_i3) in human embryonic kidney (HEK)293T cells. Glucuronidation assays with 4-methylumbelliferone (4MU) showed that 1A8_i3 had no activity and was unable to inhibit the activity of 1A8_i1 protein. In summary, the activation of cryptic donor splice sites within the UGT1A first-exon region expands the UGT1A transcriptome and proteome. The 1A8_n2 cryptic donor splice site is highly active in colorectal tissues, representing an important cis-regulatory element that negatively regulates the function of the UGT1A8 gene through pre-mRNA splicing. SIGNIFICANT STATEMENT: The UGT1A locus generates nine canonical mRNAs, 65 alternately spliced transcripts, and 34 different circular RNAs. The present study reports a series of novel UDP-glucuronosyltransferase (UGT)1A variants resulting from use of cryptic donor splice sites in both normal and cancerous tissues, several of which are predicted to encode variant UGT1A proteins with truncated aglycone-binding domains. Of these, 1A8_n2 shows exceptionally high abundance in colorectal tissues, highlighting its potential role in the first-pass metabolism in gut through the glucuronidation pathway.
Collapse
Affiliation(s)
- Dong Gui Hu
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Shashikanth Marri
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Julie-Ann Hulin
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Radwan Ansaar
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Peter I Mackenzie
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Ross A McKinnon
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| | - Robyn Meech
- College of Medicine and Public Health, Flinders Health and Medical Research Institute Flinders University, Bedford Park, Australia
| |
Collapse
|
4
|
Wang H, Song J, Hunt BJ, Zuo K, Zhou H, Hayward A, Li B, Xiao Y, Geng X, Bass C, Zhou S. UDP-glycosyltransferases act as key determinants of host plant range in generalist and specialist Spodoptera species. Proc Natl Acad Sci U S A 2024; 121:e2402045121. [PMID: 38683998 PMCID: PMC11087754 DOI: 10.1073/pnas.2402045121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/13/2024] [Indexed: 05/02/2024] Open
Abstract
Phytophagous insects have evolved sophisticated detoxification systems to overcome the antiherbivore chemical defenses produced by many plants. However, how these biotransformation systems differ in generalist and specialist insect species and their role in determining insect host plant range remains an open question. Here, we show that UDP-glucosyltransferases (UGTs) play a key role in determining the host range of insect species within the Spodoptera genus. Comparative genomic analyses of Spodoptera species that differ in host plant breadth identified a relatively conserved number of UGT genes in generalist species but high levels of UGT gene pseudogenization in the specialist Spodoptera picta. CRISPR-Cas9 knockouts of the three main UGT gene clusters of Spodoptera frugiperda revealed that UGT33 genes play an important role in allowing this species to utilize the poaceous plants maize, wheat, and rice, while UGT40 genes facilitate utilization of cotton. Further functional analyses in vivo and in vitro identified the UGT SfUGT33F32 as the key mechanism that allows generalist S. frugiperda to detoxify the benzoxazinoid DIMBOA (2,4-dihydroxy-7-methoxy-2H-1,4-benzoxazin-3(4H)-one), a potent insecticidal phytotoxin produced by poaceous plants. However, while this detoxification capacity is conserved in several generalist Spodoptera species, Spodoptera picta, which specializes on Crinum plants, is unable to detoxify DIMBOA due to a nonfunctionalizing mutation in SpUGT33F34. Collectively, these findings provide insight into the role of insect UGTs in host plant adaptation, the mechanistic basis of evolutionary transitions between generalism and specialism and offer molecular targets for controlling a group of notorious insect pests.
Collapse
Affiliation(s)
- Huidong Wang
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, College of Agriculture, Henan University, Kaifeng475004, Henan, China
| | - Jing Song
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, College of Agriculture, Henan University, Kaifeng475004, Henan, China
| | - Benjamin J. Hunt
- Centre for Ecology and Conservation, University of Exeter, PenrynTR10 9FE, United Kingdom
| | - Kairan Zuo
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, College of Agriculture, Henan University, Kaifeng475004, Henan, China
| | - Huiru Zhou
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, College of Agriculture, Henan University, Kaifeng475004, Henan, China
| | - Angela Hayward
- Centre for Ecology and Conservation, University of Exeter, PenrynTR10 9FE, United Kingdom
| | - Bingbing Li
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, College of Agriculture, Henan University, Kaifeng475004, Henan, China
| | - Yajuan Xiao
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, College of Agriculture, Henan University, Kaifeng475004, Henan, China
| | - Xing Geng
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, College of Agriculture, Henan University, Kaifeng475004, Henan, China
| | - Chris Bass
- Centre for Ecology and Conservation, University of Exeter, PenrynTR10 9FE, United Kingdom
| | - Shutang Zhou
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, College of Agriculture, Henan University, Kaifeng475004, Henan, China
| |
Collapse
|
5
|
Xue Q, Wang Y, Liu Y, Hua H, Zhou X, Xu Y, Zhang Y, Xiong C, Liu X, Yang K, Huang Y. Dysregulated Glucuronidation of Bilirubin Exacerbates Liver Inflammation and Fibrosis in Schistosomiasis Japonica through the NF-κB Signaling Pathway. Pathogens 2024; 13:287. [PMID: 38668242 PMCID: PMC11054532 DOI: 10.3390/pathogens13040287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/29/2024] Open
Abstract
Hepatic fibrosis is an important pathological manifestation of chronic schistosome infection. Patients with advanced schistosomiasis show varying degrees of abnormalities in liver fibrosis indicators and bilirubin metabolism. However, the relationship between hepatic fibrosis in schistosomiasis and dysregulated bilirubin metabolism remains unclear. In this study, we observed a positive correlation between total bilirubin levels and the levels of ALT, AST, LN, and CIV in patients with advanced schistosomiasis. Additionally, we established mouse models at different time points following S. japonicum infection. As the infection time increased, liver fibrosis escalated, while liver UGT1A1 consistently exhibited a low expression, indicating impaired glucuronidation of bilirubin metabolism in mice. In vitro experiments suggested that SEA may be a key inhibitor of hepatic UGT1A1 expression after schistosome infection. Furthermore, a high concentration of bilirubin activated the NF-κB signaling pathway in L-O2 cells in vitro. These findings suggested that the dysregulated glucuronidation of bilirubin caused by S. japonicum infection may play a significant role in schistosomiasis liver fibrosis through the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Qingkai Xue
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, 117 Meiyuan Yangxiang, Wuxi 214064, China; (Q.X.)
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi 214064, China
| | - Yuyan Wang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China;
| | - Yiyun Liu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, 117 Meiyuan Yangxiang, Wuxi 214064, China; (Q.X.)
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi 214064, China
- School of Public Health, Nanjing Medical University, Nanjing 211166, China;
| | - Haiyong Hua
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, 117 Meiyuan Yangxiang, Wuxi 214064, China; (Q.X.)
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi 214064, China
| | - Xiangyu Zhou
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, 117 Meiyuan Yangxiang, Wuxi 214064, China; (Q.X.)
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi 214064, China
| | - Yongliang Xu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, 117 Meiyuan Yangxiang, Wuxi 214064, China; (Q.X.)
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi 214064, China
| | - Ying Zhang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, 117 Meiyuan Yangxiang, Wuxi 214064, China; (Q.X.)
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi 214064, China
| | - Chunrong Xiong
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, 117 Meiyuan Yangxiang, Wuxi 214064, China; (Q.X.)
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi 214064, China
| | - Xinjian Liu
- Department of Pathogen Biology, Key Laboratory of Antibody Techniques of National Health Commission, Nanjing Medical University, Nanjing 211166, China
| | - Kun Yang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, 117 Meiyuan Yangxiang, Wuxi 214064, China; (Q.X.)
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi 214064, China
- School of Public Health, Nanjing Medical University, Nanjing 211166, China;
| | - Yuzheng Huang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China;
| |
Collapse
|
6
|
Wu C, Luo M, Xie D, Zhong S, Xu J, Lu D. Kinetic Characterization of Estradiol Glucuronidation by Liver Microsomes and Expressed UGT Enzymes: The Effects of Organic Solvents. Eur J Drug Metab Pharmacokinet 2024:10.1007/s13318-024-00888-2. [PMID: 38472634 DOI: 10.1007/s13318-024-00888-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND AND OBJECTIVE In vitro glucuronidation of 17β-estradiol (estradiol) is often performed to assess the role of uridine 5'-diphospho-glucuronosyltransferase 1A1 (UGT1A1) in xenobiotic/drug metabolism. The objective of this study was to determine the effects of four commonly used organic solvents [i.e., dimethyl sulfoxide (DMSO), methanol, ethanol, and acetonitrile] on the glucuronidation kinetics of estradiol, which can be glucuronidated at C3 and C17 positions. METHODS The impacts of organic solvents on estradiol glucuronidation were determined by using expressed UGT enzymes and liver microsomes from both human and animals. RESULTS In human liver microsomes (HLM), methanol, ethanol, and acetonitrile significantly altered estradiol glucuronidation kinetics with increased Vmax (up to 2.6-fold) and CLmax (up to 2.8-fold) values. Altered estradiol glucuronidation in HLM was deduced to be attributed to the enhanced metabolic activities of UGT1A1 and UGT2B7, whose activities differ at the two glucuronidation positions. The effects of organic solvents on estradiol glucuronidation were glucuronidation position-, isozyme-, and solvent-specific. Furthermore, both ethanol and acetonitrile have a greater tendency to modify the glucuronidation activity of estradiol in animal liver microsomes. CONCLUSION Organic solvents such as methanol, ethanol, and acetonitrile showed great potential in adjusting the glucuronidation of estradiol. DMSO is the most suitable solvent due to its minimal influence on estradiol glucuronidation. Researchers should be cautious in selecting appropriate solvents to get accurate results when assessing the metabolism of a new chemical entity.
Collapse
Affiliation(s)
- Caimei Wu
- Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232 Waihuan East Road, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, 510006, China
| | - Meixue Luo
- Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232 Waihuan East Road, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, 510006, China
| | - Dihao Xie
- Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232 Waihuan East Road, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, 510006, China
| | - Simin Zhong
- Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232 Waihuan East Road, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, 510006, China
| | - Jiahao Xu
- Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232 Waihuan East Road, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, 510006, China
| | - Danyi Lu
- Institute of Molecular Rhythm and Metabolism, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232 Waihuan East Road, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, 510006, China.
| |
Collapse
|
7
|
Miao YS, Wang JY, Zhuang RR, Huo XK, Yi ZC, Sun XN, Yu ZL, Tian XG, Ning J, Feng L, Ma XC, Lv X. A high-affinity fluorescent probe for human uridine-disphosphate glucuronosyltransferase 1A9 function monitoring under environmental pollutant exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133439. [PMID: 38218035 DOI: 10.1016/j.jhazmat.2024.133439] [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: 10/02/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024]
Abstract
Uridine-disphosphate glucuronosyltransferase 1A9 (UGT1A9), an important detoxification and inactivation enzyme for toxicants, regulates the exposure level of environmental pollutants in the human body and induces various toxicological consequences. However, an effective tool for high-throughput monitoring of UGT1A9 function under exposure to environmental pollutants is still lacking. In this study, 1,3-dichloro-7-hydroxy-9,9-dimethylacridin-2(9H)-one (DDAO) was found to exhibit excellent specificity and high affinity towards human UGT1A9. Remarkable changes in absorption and fluorescence signals after reacting with UGT1A9 were observed, due to the intramolecular charge transfer (ICT) mechanism. Importantly, DDAO was successfully applied to monitor the biological functions of UGT1A9 in response to environmental pollutant exposure not only in microsome samples, but also in living cells by using a high-throughput screening method. Meanwhile, the identified pollutants that disturb UGT1A9 functions were found to significantly influence the exposure level and retention time of bisphenol S/bisphenol A in living cells. Furthermore, the molecular mechanism underlying the inhibition of UGT1A9 by these pollutant-derived disruptors was elucidated by molecular docking and molecular dynamics simulations. Collectively, a fluorescent probe to characterize the responses of UGT1A9 towards environmental pollutants was developed, which was beneficial for elucidating the health hazards of environmental pollutants from a new perspective.
Collapse
Affiliation(s)
- Yi-Sheng Miao
- Second Affiliated Hospital, Dalian Medical University, Dalian 116023, China; College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Jia-Yue Wang
- Second Affiliated Hospital, Dalian Medical University, Dalian 116023, China; College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Rui-Rui Zhuang
- College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xiao-Kui Huo
- Second Affiliated Hospital, Dalian Medical University, Dalian 116023, China
| | - Zi-Chang Yi
- College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xiao-Nan Sun
- College of Integrative Medicine, Dalian Medical University, Dalian 116044, China; School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Zhen-Long Yu
- College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xiang-Ge Tian
- Second Affiliated Hospital, Dalian Medical University, Dalian 116023, China
| | - Jing Ning
- College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Lei Feng
- Second Affiliated Hospital, Dalian Medical University, Dalian 116023, China; Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China.
| | - Xiao-Chi Ma
- Second Affiliated Hospital, Dalian Medical University, Dalian 116023, China; Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Xia Lv
- Second Affiliated Hospital, Dalian Medical University, Dalian 116023, China; College of Integrative Medicine, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
8
|
Hu DG, Marri S, Hulin JA, McKinnon RA, Mackenzie PI, Meech R. A Comprehensive Bioinformatic Analysis of RNA-seq Datasets Reveals a Differential and Variable Expression of Wildtype and Variant UGT1A Transcripts in Human Tissues and Their Deregulation in Cancers. Cancers (Basel) 2024; 16:353. [PMID: 38254842 PMCID: PMC10814044 DOI: 10.3390/cancers16020353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
The UGT1A locus generates over 60 different alternatively spliced transcripts and 30 circular RNAs. To date, v2 and v3 transcripts are the only variant UGT1A transcripts that have been functionally characterized. Both v2 and v3 transcripts encode the same inactive variant UGT1A proteins (i2s) that can negatively regulate glucuronidation activity and influence cancer cell metabolism. However, the abundance and interindividual variability in the expression of v2 and v3 transcripts in human tissues and their potential deregulation in cancers have not been comprehensively assessed. To address this knowledge gap, we quantified the expression levels of v1, v2, and v3 transcripts using RNA-seq datasets with large cohorts of normal tissues and paired normal and tumor tissues from patients with six different cancer types (liver, kidney, colon, stomach, esophagus, and bladder cancer). We found that v2 and v3 abundance varied significantly between different tissue types, and that interindividual variation was also high within the same tissue type. Moreover, the ratio of v2 to v3 variants varied between tissues, implying their differential regulation. Our results showed higher v2 abundance in gastrointestinal tissues than liver and kidney tissues, suggesting a more significant negative regulation of glucuronidation by i2 proteins in gastrointestinal tissues than in liver and kidney tissues. We further showed differential deregulation of wildtype (v1) and variant transcripts (v2, v3) in cancers that generally increased the v2/v1 and/or v3/v1 expression ratios in tumors compared to normal tissues, indicating a more significant role of the variants in tumors. Finally, we report ten novel UGT1A transcripts with novel 3' terminal exons, most of which encode variant proteins with a similar structure to UGT1A_i2 proteins. These findings further emphasize the diversity of the UGT1A transcriptome and proteome.
Collapse
Affiliation(s)
- Dong Gui Hu
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide 5042, Australia; (S.M.); (J.-A.H.); (R.A.M.); (P.I.M.); (R.M.)
| | | | | | | | | | | |
Collapse
|
9
|
Zhou L, Montalvo AD, Collins JM, Wang D. Quantitative analysis of the UDP-glucuronosyltransferase transcriptome in human tissues. Pharmacol Res Perspect 2023; 11:e01154. [PMID: 37983911 PMCID: PMC10659769 DOI: 10.1002/prp2.1154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/22/2023] Open
Abstract
UDP-glucuronosyltransferases (UGTs) are phase II drug metabolizing enzymes that play important roles in the detoxification of endogenous and exogenous substrates. The 22 human UGTs belong to four families (UGT1, UGT2, UGT3, and UGT8) and differ in their expression, substrate specificity, UDP-sugar preference, and physiological functions. Differential expression/activity of the UGTs contributes to interperson variability in drug responses and toxicity, hormone homeostasis, and disease/cancer risks. However, in normal tissues, the tissue-specific expression profiles and transcriptional regulation of the UGTs are still not fully understood. In this study, we comprehensively analyzed the transcriptome of 22 UGTs in 54 human tissues/regions using RNAseq data from GTEx. We then validated the findings in the liver and small intestine samples using real-time PCR. Our results showed large interindividual variability across tissues in the expression of each UGT and the overall composition of UGT pools, consisting of different UGTs and their splice isoforms. Our results also revealed coexpression of the UGTs, Cytochrome P450s, and many transcription factors in the liver, suggesting potential coregulation or functional coordination. Our results provide the groundwork for future studies to detail further the regulation of the expression and activity of the UGTs.
Collapse
Affiliation(s)
- Lucas Zhou
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for PharmacogenomicsUniversity of FloridaGainesvilleFloridaUSA
| | - Abelardo D. Montalvo
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for PharmacogenomicsUniversity of FloridaGainesvilleFloridaUSA
| | - Joseph M. Collins
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for PharmacogenomicsUniversity of FloridaGainesvilleFloridaUSA
| | - Danxin Wang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for PharmacogenomicsUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
10
|
Sulekha A, Osborne MJ, Gasiorek J, Borden KLB. 1H, 13C, 15N Backbone and sidechain chemical shift assignments of the C-terminal domain of human UDP-glucuronosyltransferase 2B17 (UGT2B17-C). BIOMOLECULAR NMR ASSIGNMENTS 2023; 17:67-73. [PMID: 36757531 DOI: 10.1007/s12104-023-10122-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/01/2023] [Indexed: 06/02/2023]
Abstract
UDP-glucuronosyltransferases are the principal enzymes involved in the glucuronidation of metabolites and xenobiotics for physiological clearance in humans. Though glucuronidation is an indispensable process in the phase II metabolic pathway, UGT-mediated glucuronidation of most prescribed drugs (> 55%) and clinical evidence of UGT-associated drug resistance are major concerns for therapeutic development. While UGTs are highly conserved enzymes, they manifest unique substrate and inhibitor specificity which is poorly understood given the dearth of experimentally determined full-length structures. Such information is important not only to conceptualize their specificity but is central to the design of inhibitors specific to a given UGT in order to avoid toxicity associated with pan-UGT inhibitors. Here, we provide the 1H, 13C and 15N backbone (~ 90%) and sidechain (~ 62%) assignments for the C-terminal domain of UGT2B17, which can be used to determine the molecular binding sites of inhibitor and substrate, and to understand the atomic basis for inhibitor selectivity between UGT2B17 and other members of the UGT2B subfamily. Given the physiological relevance of UGT2B17 in the elimination of hormone-based cancer drugs, these assignments will contribute towards dissecting the structural basis for substrate specificity, selective inhibitor recognition and other aspects of enzyme activity with the goal of selectively overcoming glucuronidation-based drug resistance.
Collapse
Affiliation(s)
- Anamika Sulekha
- Department of Pathology and Cell Biology and Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, Pavilion Marcelle‑Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Michael J Osborne
- Department of Pathology and Cell Biology and Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, Pavilion Marcelle‑Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Jadwiga Gasiorek
- Department of Pathology and Cell Biology and Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, Pavilion Marcelle‑Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Katherine L B Borden
- Department of Pathology and Cell Biology and Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, Pavilion Marcelle‑Coutu, Chemin Polytechnique, Montreal, QC, Canada.
| |
Collapse
|
11
|
Yang Z, Xiao T, Lu K. Contribution of UDP-glycosyltransferases to chlorpyrifos resistance in Nilaparvata lugens. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 190:105321. [PMID: 36740334 DOI: 10.1016/j.pestbp.2022.105321] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 06/18/2023]
Abstract
As a multigene superfamily of Phase II detoxification enzymes, uridine diphosphate (UDP)-glycosyltransferases (UGTs) play important roles in the metabolism of xenobiotics including insecticides. In this study, 5-nitrouracil, an inhibitor of UGT enzyme activity, effectively increased the toxicity of chlorpyrifos to the chlorpyrifos-resistant strain of Nilaparvata lugens, one of the most resistant rice pests. The enzyme content of UGT in the resistant strain was significantly higher than that in the susceptible strain. Among 20 identified UGT genes, UGT386H2, UGT386J2, UGT386N2 and UGT386P1 were found significantly overexpressed in the resistant strain and can be effectively induced by chlorpyrifos. These four UGT genes were most highly expressed in the midgut and/or fat body, two main insect detoxification tissues. Amino acid sequence alignments revealed that these four UGTs contained a variable N-terminal substrate-binding domain and a conserved C-terminal sugar donor-binding domain. Furthermore, homology modeling and molecular docking analyses showed that these UGTs could stably bind to chlorpyrifos and chlorpyrifos oxon, with the binding free energies from -19.4 to -110.62 kcal mol-1. Knockdown of UGT386H2 or UGT386P1 by RNA interference dramatically increased the susceptibility of the resistant strain to chlorpyrifos. These findings suggest that overexpression of these two UGT genes contributes to chlorpyrifos resistance in N. lugens.
Collapse
Affiliation(s)
- Zhiming Yang
- Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, China
| | - Tianxiang Xiao
- Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, China
| | - Kai Lu
- Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
12
|
Zheng R, Shao S, Li X, Zhang W, Zhang S, Yu Z, Ye Q. Understanding the metabolism of the novel plant antiviral agent dufulin by different positional 14C labeling in cherry radishes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 858:159396. [PMID: 36244481 DOI: 10.1016/j.scitotenv.2022.159396] [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: 09/08/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
Dufulin is a new type of plant antiviral agent. However, its metabolism in plants, which is very important for environmental risk assessment, is still unclear. In this study, we used 14C markers at different positions and high-performance liquid chromatography-quadrupole time-of-flight-mass spectrometry (HPLC-QTOF-MS) to qualitatively and quantitatively analyze dufulin metabolites in cherry radish. By combining ion pairs with unique abundance ratios, we clarified the metabolite structures, inferred the metabolic pathway of dufulin, and clarified the criteria for residues. The extractable residue of dufulin from cherry radish stem and leaf tissues was above 98 % and that in the succulent root was above 87 %. In the stem and leaf tissues and succulent root, dufulin underwent both phase I and phase II metabolism, and four metabolites were produced, including a conjugate of glucose malonate and hydroxylated dufulin, which was confirmed by comparison with a standard. However, the proportions and concentrations of the four metabolites did not meet the residue criteria, so only the dufulin precursor compound was included as a residue. This study provides reliable data for evaluating the impacts of dufulin on the environment and human health and for objectively examining the safety of dufulin.
Collapse
Affiliation(s)
- Ruonan Zheng
- Institute of Nuclear Agricultural Sciences, Key Laboratory of Nuclear Agricultural Sciences of Ministry of Agriculture of the PRC and Zhejiang Province, Zhejiang University, Hangzhou 310058, China.
| | - Siyao Shao
- Institute of Nuclear Agricultural Sciences, Key Laboratory of Nuclear Agricultural Sciences of Ministry of Agriculture of the PRC and Zhejiang Province, Zhejiang University, Hangzhou 310058, China.
| | - Xiaofeng Li
- Institute of Catalysis, Key Laboratory of Applied Chemistry of Zhejiang Province, Zhejiang University, Hangzhou 310058, China.
| | - Weiwei Zhang
- Institute of Nuclear Agricultural Sciences, Key Laboratory of Nuclear Agricultural Sciences of Ministry of Agriculture of the PRC and Zhejiang Province, Zhejiang University, Hangzhou 310058, China.
| | - Sufen Zhang
- Institute of Nuclear Agricultural Sciences, Key Laboratory of Nuclear Agricultural Sciences of Ministry of Agriculture of the PRC and Zhejiang Province, Zhejiang University, Hangzhou 310058, China.
| | - Zhiyang Yu
- Institute of Nuclear Agricultural Sciences, Key Laboratory of Nuclear Agricultural Sciences of Ministry of Agriculture of the PRC and Zhejiang Province, Zhejiang University, Hangzhou 310058, China.
| | - Qingfu Ye
- Institute of Nuclear Agricultural Sciences, Key Laboratory of Nuclear Agricultural Sciences of Ministry of Agriculture of the PRC and Zhejiang Province, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
13
|
Liu W, Li J, Zhao R, Lu Y, Huang P. The Uridine diphosphate (UDP)-glycosyltransferases (UGTs) superfamily: the role in tumor cell metabolism. Front Oncol 2023; 12:1088458. [PMID: 36741721 PMCID: PMC9892627 DOI: 10.3389/fonc.2022.1088458] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/09/2022] [Indexed: 01/20/2023] Open
Abstract
UDP-glycosyltransferases (UGTs), important enzymes in biotransformation, control the levels and distribution of numerous endogenous signaling molecules and the metabolism of a wide range of endogenous and exogenous chemicals. The UGT superfamily in mammals consists of the UGT1, UGT2, UGT3, and UGT8 families. UGTs are rate-limiting enzymes in the glucuronate pathway, and in tumors, they are either overexpressed or underexpressed. Alterations in their metabolism can affect gluconeogenesis and lipid metabolism pathways, leading to alterations in tumor cell metabolism, which affect cancer development and prognosis. Glucuronidation is the most common mammalian conjugation pathway. Most of its reactions are mainly catalyzed by UGT1A, UGT2A and UGT2B. The body excretes UGT-bound small lipophilic molecules through the bile, urine, or feces. UGTs conjugate a variety of tiny lipophilic molecules to sugars, such as galactose, xylose, acetylglucosamine, glucuronic acid, and glucose, thereby inactivating and making water-soluble substrates, such as carcinogens, medicines, steroids, lipids, fatty acids, and bile acids. This review summarizes the roles of members of the four UGT enzyme families in tumor function, metabolism, and multiple regulatory mechanisms, and its Inhibitors and inducers. The function of UGTs in lipid metabolism, drug metabolism, and hormone metabolism in tumor cells are among the most important topics covered.
Collapse
Affiliation(s)
| | | | | | - Yao Lu
- *Correspondence: Yao Lu, ; Panpan Huang,
| | | |
Collapse
|
14
|
Significance of UGT1A6, UGT1A9, and UGT2B7 genetic variants and their mRNA expression in the clinical outcome of renal cell carcinoma. Mol Cell Biochem 2022:10.1007/s11010-022-04637-4. [PMID: 36571650 DOI: 10.1007/s11010-022-04637-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 12/07/2022] [Indexed: 12/27/2022]
Abstract
UDP-glucuronosyltransferase (UGT) metabolizes a number of endogenous and exogenous substrates. Renal cells express high amounts of UGT; however, the significance of UGT in patients with renal cell carcinoma (RCC) remains unknown. In this study, we profile the mRNA expression of UGT subtypes (UGT1A6, UGT1A9, and UGT2B7) and their genetic variants in the kidney tissue of 125 Japanese patients with RCC (Okayama University Hospital, Japan). In addition, we elucidate the association between the UGT variants and UGT mRNA expression levels and clinical outcomes in these patients. The three representative genetic variants, namely, UGT1A6 541A > G, UGT1A9 i399C > T, and UGT2B7-161C > T, were genotyped, and their mRNA expression levels in each tissue were determined. We found that the mRNA expression of the three UGTs (UGT1A6, UGT1A9, and UGT2B7) are significantly downregulated in RCC tissues. Moreover, in patients with RCC, the UGT2B7-161C > T variant and high UGT2B7 mRNA expression are significantly correlated with preferable cancer-specific survival (CSS) and overall survival (OS), respectively. As such, the UGT2B7-161C > T variant and UGT2B7 mRNA expression level were identified as significant independent prognostic factors of CSS and CSS/OS, respectively. Taken together, these findings indicate that UGT2B7 has a role in RCC progression and may, therefore, represent a potential prognostic biomarker for patients with RCC.
Collapse
|
15
|
Dias VHV, Mattos JJ, Bastolla CLV, Lüchmann KH, Bainy ACD. Characterisation of UDP-glucuronosyltransferase activity in sea turtle Chelonia mydas. Xenobiotica 2022; 52:1011-1019. [PMID: 36594659 DOI: 10.1080/00498254.2022.2164750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Uridine diphosphate glucuronosyltransferase (UGT) enzymes conjugate many lipophilic chemicals, such as drugs, environmental contaminants, and endogenous compounds, promoting their excretion. The complexity of UGT kinetics, and the location of enzyme active site in endoplasmic reticulum lumen, requires an accurate optimisation of enzyme assays.In the present study, we characterised UGT activity in liver microsomes of green turtles (Chelonia mydas), an endangered species. The conditions for measuring UGT activity were standardised through spectrofluorimetric methods, using the substrates 4-methylumbelliferone (4-MU) and uridine diphosphate glucuronic acid (UDPGA) at 30 °C and pH 7.4.The green turtles showed UGT activity at the saturating concentrations of substrates of 250 µM to 4-MU and 7 mM to UDPGA. The alamethicin, Brij®58, bovine serum albumin (BSA), and magnesium increased UGT activity. The assay using alamethicin (22 µg per mg of protein), magnesium (1 mM), and BSA (0.25%) reached the highest Vmax (1203 pmol·min-1mg·protein-1). Lithocholic acid and diclofenac inhibited UGT activity in green turtles.This study is the first report of UGT activity in the liver of green turtles and provides a base for future studies to understand the mechanisms of toxicity by exposure to contaminants in this charismatic species.
Collapse
Affiliation(s)
- Vera Helena V Dias
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry-LABCAI, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Jacó J Mattos
- Aquaculture Pathology Research Center-NEPAQ, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Camila L V Bastolla
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry-LABCAI, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Karim H Lüchmann
- Department of Scientific and Technological Education, Santa Catarina State University, Florianópolis, Brazil
| | - Afonso C D Bainy
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry-LABCAI, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
16
|
The Somatic Mutation Landscape of UDP-Glycosyltransferase ( UGT) Genes in Human Cancers. Cancers (Basel) 2022; 14:cancers14225708. [PMID: 36428799 PMCID: PMC9688768 DOI: 10.3390/cancers14225708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The human UDP-glycosyltransferase (UGTs) superfamily has a critical role in the metabolism of anticancer drugs and numerous pro/anti-cancer molecules (e.g., steroids, lipids, fatty acids, bile acids and carcinogens). Recent studies have shown wide and abundant expression of UGT genes in human cancers. However, the extent to which UGT genes acquire somatic mutations within tumors remains to be systematically investigated. In the present study, our comprehensive analysis of the somatic mutation profiles of 10,069 tumors from 33 different TCGA cancer types identified 3427 somatic mutations in UGT genes. Overall, nearly 18% (1802/10,069) of the assessed tumors had mutations in UGT genes with huge variations in mutation frequency across different cancer types, ranging from over 25% in five cancers (COAD, LUAD, LUSC, SKCM and UCSC) to less than 5% in eight cancers (LAML, MESO, PCPG, PAAD, PRAD, TGCT, THYM and UVM). All 22 UGT genes showed somatic mutations in tumors, with UGT2B4, UGT3A1 and UGT3A2 showing the largest number of mutations (289, 307 and 255 mutations, respectively). Nearly 65% (2260/3427) of the mutations were missense, frame-shift and nonsense mutations that have been predicted to code for variant UGT proteins. Furthermore, about 10% (362/3427) of the mutations occurred in non-coding regions (5' UTR, 3' UTR and splice sites) that may be able to alter the efficiency of translation initiation, miRNA regulation or the splicing of UGT transcripts. In conclusion, our data show widespread somatic mutations of UGT genes in human cancers that may affect the capacity of cancer cells to metabolize anticancer drugs and endobiotics that control pro/anti-cancer signaling pathways. This highlights their potential utility as biomarkers for predicting therapeutic efficacy and clinical outcomes.
Collapse
|
17
|
Luo MY, Zhou Y, Gu WM, Wang C, Shen NX, Dong JK, Lei HM, Tang YB, Liang Q, Zou JH, Xu L, Ma P, Zhuang G, Bi L, Xu L, Zhu L, Chen HZ, Shen Y. Metabolic and Nonmetabolic Functions of PSAT1 Coordinate Signaling Cascades to Confer EGFR Inhibitor Resistance and Drive Progression in Lung Adenocarcinoma. Cancer Res 2022; 82:3516-3531. [PMID: 36193649 DOI: 10.1158/0008-5472.can-21-4074] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 05/03/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022]
Abstract
Emerging evidence demonstrates that the dysregulated metabolic enzymes can accelerate tumorigenesis and progression via both metabolic and nonmetabolic functions. Further elucidation of the role of metabolic enzymes in EGFR inhibitor resistance and metastasis, two of the leading causes of death in lung adenocarcinoma, could help improve patient outcomes. Here, we found that aberrant upregulation of phosphoserine aminotransferase 1 (PSAT1) confers erlotinib resistance and tumor metastasis in lung adenocarcinoma. Depletion of PSAT1 restored sensitivity to erlotinib and synergistically augmented the tumoricidal effect. Mechanistically, inhibition of PSAT1 activated the ROS-dependent JNK/c-Jun pathway to induce cell apoptosis. In addition, PSAT1 interacted with IQGAP1, subsequently activating STAT3-mediated cell migration independent of its metabolic activity. Clinical analyses showed that PSAT1 expression positively correlated with the progression of human lung adenocarcinoma. Collectively, these findings reveal the multifunctionality of PSAT1 in promoting tumor malignancy through its metabolic and nonmetabolic activities. SIGNIFICANCE Metabolic and nonmetabolic functions of PSAT1 confer EGFR inhibitor resistance and promote metastasis in lung adenocarcinoma, suggesting therapeutic targeting of PSAT1 may attenuate the malignant features of lung cancer.
Collapse
Affiliation(s)
- Ming-Yu Luo
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Ye Zhou
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Wei-Ming Gu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Cheng Wang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Ning-Xiang Shen
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Jiang-Kai Dong
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Hui-Min Lei
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Ya-Bin Tang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Qian Liang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Jing-Hua Zou
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Lu Xu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Pengfei Ma
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglei Zhuang
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Bi
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Xu
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| |
Collapse
|
18
|
Hao Z, Xu J, Zhao H, Zhou W, Liu Z, He S, Yin X, Zhang B, Wang Z, Zhou X. The inhibition of tamoxifen on UGT2B gene expression and enzyme activity in rat liver contribute to the estrogen homeostasis dysregulation. BMC Pharmacol Toxicol 2022; 23:33. [PMID: 35642027 PMCID: PMC9158366 DOI: 10.1186/s40360-022-00574-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 05/24/2022] [Indexed: 11/12/2022] Open
Abstract
Background Tamoxifen treatment may induce dysregulation of estrogen homeostasis, leading to the occurrence of related adverse reactions. However, the potential mechanisms are still unclear. The purpose of the present study was to uncover whether tamoxifen treatment would act on estrogen metabolism-related biological enzymes and the regulatory effect on estrogen homeostasis to clarify the key factors and potential mechanisms of adverse reactions caused by long-term use of tamoxifen. Method Female SD rats were administrated with tamoxifen CMC-Na solution (p.o.) once daily for four weeks and then housed at room temperature. Serum, breast, liver, uterus, and ovarian tissues were obtained, and the effects of tamoxifen administration on estrogen homeostasis, the expression, and activity of estrogen metabolic enzyme were evaluated. Results Compared with the control group, the estrogen homeostasis was disturbed and the expression and activity of UGT2B1 (homology with human UGT2B7) were significantly reduced in the rats administrated with tamoxifen. The inhibitory effect of tamoxifen on UGT2B7 was dominated by hydrophobic and π-π stacking interactions, resulting in a concentration-dependent inhibition of UGT2B7 activity by tamoxifen and the imbalance of ligand-activated transcription factors, leading to abnormal regulation of UGT2B and disturbance of estrogen homeostasis, which in turn led to adverse reactions of tamoxifen. Conclusion We established links between estrogen metabolism and tamoxifen administration and we proposed that the UGT2B inhibition was involved in the disturbance of estrogen homeostasis and the occurrence of tamoxifen-related adverse reactions. Supplementary Information The online version contains supplementary material available at 10.1186/s40360-022-00574-6.
Collapse
Affiliation(s)
- Zhixiang Hao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China.,Department of Pharmacy, Xuzhou First People's hospital, 221000, Xuzhou, China
| | - Jiahao Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Han Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Wei Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Zhao Liu
- Department of Thyroid and Breast Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shiqing He
- Department of Thyroid and Breast Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Bei Zhang
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Zhongjian Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Xueyan Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, College of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China.
| |
Collapse
|
19
|
Dimunová D, Matoušková P, Podlipná R, Boušová I, Skálová L. The role of UDP-glycosyltransferases in xenobiotic-resistance. Drug Metab Rev 2022; 54:282-298. [DOI: 10.1080/03602532.2022.2083632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Diana Dimunová
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Petra Matoušková
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Radka Podlipná
- Laboratory of Plant Biotechnologies, Institute of Experimental Botany, Czech Academy of Sciences, 165 02 Praha 6 - Lysolaje, Czech Republic
| | - Iva Boušová
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Lenka Skálová
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| |
Collapse
|
20
|
Hu DG, Mackenzie PI, Hulin JA, McKinnon RA, Meech R. Regulation of human UDP-glycosyltransferase ( UGT) genes by miRNAs. Drug Metab Rev 2022; 54:120-140. [PMID: 35275773 DOI: 10.1080/03602532.2022.2048846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The human UGT gene superfamily is divided into four subfamilies (UGT1, UGT2, UGT3 and UGT8) that encodes 22 functional enzymes. UGTs are critical for the metabolism and clearance of numerous endogenous and exogenous compounds, including steroid hormones, bile acids, bilirubin, fatty acids, carcinogens, and therapeutic drugs. Therefore, the expression and activities of UGTs are tightly regulated by multiple processes at the transcriptional, post-transcriptional and post-translational levels. During recent years, nearly twenty studies have investigated the post-transcriptional regulation of UGT genes by miRNAs using human cancer cell lines (predominantly liver cancer). Overall, 14 of the 22 UGT mRNAs (1A1, 1A3, 1A4, 1A6, 1A8, 1A9, 1A10, 2A1, 2B4, 2B7, 2B10, 2B15, 2B17, UGT8) have been shown to be regulated by various miRNAs through binding to their respective 3' untranslated regions (3'UTRs). Three 3'UTRs (UGT1A, UGT2B7 and UGT2B15) contain the largest number of functional miRNA target sites; in particular, the UGT1A 3'UTR contains binding sites for 12 miRNAs (548d-5p, 183-5p, 214-5p, 486-3p, 200a-3p, 491-3p, 141-3p, 298, 103b, 376b-3p, 21-3p, 1286). Although all nine UGT1A family members have the same 3'UTR, these miRNA target sites appear to be functional in an isoform-specific and cellular context-dependent manner. Collectively, these observations demonstrate that miRNAs represent important post-transcriptional regulators of the UGT gene superfamily. In this article, we present a comprehensive review of reported UGT/miRNA regulation studies, describe polymorphisms within functional miRNA target sites that may affect their functionalities, and discuss potential cooperative and competitive regulation of UGT mRNAs by miRNAs through adjacently located miRNA target sites.
Collapse
Affiliation(s)
- Dong Gui Hu
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Peter I Mackenzie
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Julie-Ann Hulin
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Ross A McKinnon
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Robyn Meech
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| |
Collapse
|
21
|
Davydov DR, Dangi B, Yue G, Ahire DS, Prasad B, Zgoda VG. Exploring the Interactome of Cytochrome P450 2E1 in Human Liver Microsomes with Chemical Crosslinking Mass Spectrometry. Biomolecules 2022; 12:biom12020185. [PMID: 35204686 PMCID: PMC8869672 DOI: 10.3390/biom12020185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/05/2022] Open
Abstract
Aiming to elucidate the system-wide effects of the alcohol-induced increase in the content of cytochrome P450 2E1 (CYP2E1) on drug metabolism, we explored the array of its protein-protein interactions (interactome) in human liver microsomes (HLM) with chemical crosslinking mass spectrometry (CXMS). Our strategy employs membrane incorporation of purified CYP2E1 modified with photoreactive crosslinkers benzophenone-4-maleimide and 4-(N-succinimidylcarboxy)benzophenone. Exposure of bait-incorporated HLM samples to light was followed by isolating the His-tagged bait protein and its crosslinked aggregates on Ni-NTA agarose. Analyzing the individual bands of SDS-PAGE slabs of thereby isolated protein with the toolset of untargeted proteomics, we detected the crosslinked dimeric and trimeric complexes of CYP2E1 with other drug-metabolizing enzymes. Among the most extensively crosslinked partners of CYP2E1 are the cytochromes P450 2A6, 2C8, 3A4, 4A11, and 4F2, UDP-glucuronosyltransferases (UGTs) 1A and 2B, fatty aldehyde dehydrogenase (ALDH3A2), epoxide hydrolase 1 (EPHX1), disulfide oxidase 1α (ERO1L), and ribophorin II (RPN2). These results demonstrate the exploratory power of the proposed CXMS strategy and corroborate the concept of tight functional integration in the human drug-metabolizing ensemble through protein-protein interactions of the constituting enzymes.
Collapse
Affiliation(s)
- Dmitri R. Davydov
- Department of Chemistry, Washington State University, Pullman, WA 99164, USA;
- Correspondence:
| | - Bikash Dangi
- Department of Chemistry, Washington State University, Pullman, WA 99164, USA;
| | - Guihua Yue
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (G.Y.); (D.S.A.); (B.P.)
| | - Deepak S. Ahire
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (G.Y.); (D.S.A.); (B.P.)
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (G.Y.); (D.S.A.); (B.P.)
| | - Victor G. Zgoda
- Orekhovich Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia;
| |
Collapse
|
22
|
Osborne MJ, Rahardjo AK, Volpon L, Borden KLB. 1H, 13C and 15N chemical shift assignments of the C-terminal domain of human UDP-Glucuronosyltransferase 2B7 (UGT2B7-C). BIOMOLECULAR NMR ASSIGNMENTS 2021; 15:323-328. [PMID: 33870481 PMCID: PMC8549657 DOI: 10.1007/s12104-021-10024-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/10/2021] [Indexed: 06/02/2023]
Abstract
The human UDP-glucuronosyltransferase (UGT) family of enzymes catalyze the covalent addition of glucuronic acid to a wide range of compounds, generally rendering them inactive. Although important for clearance of environmental toxins and metabolites, UGT activation can lead to inappropriate glucuronidation of therapeutics underlying drug resistance. Indeed, 50% of medications are glucuronidated. To better understand this mode of resistance, we studied the UGT2B7 enzyme associated with glucuronidation of cancer drugs such as Tamoxifen and Sorafenib. We report 1H, 13C and 15N backbone (> 90%) and side-chain assignments (~ 78% completeness according to CYANA) for the C-terminal domain of UGT2B7 (UGT2B7-C). Given the biomedical importance of this family of enzymes, our assignments will provide a key tool for improving understanding of the biochemical basis for substrate selectivity and other aspects of enzyme activity. This in turn will inform on drug design to overcome UGT-related drug resistance.
Collapse
Affiliation(s)
- Michael J Osborne
- Department of Pathology and Cell Biology, Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, Pavillion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Amanda K Rahardjo
- Department of Pathology and Cell Biology, Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, Pavillion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Laurent Volpon
- Department of Pathology and Cell Biology, Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, Pavillion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Katherine L B Borden
- Department of Pathology and Cell Biology, Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, Pavillion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada.
| |
Collapse
|
23
|
Hu DG, Marri S, Mackenzie PI, Hulin JA, McKinnon RA, Meech R. The Expression Profiles and Deregulation of UDP-Glycosyltransferase ( UGT) Genes in Human Cancers and Their Association with Clinical Outcomes. Cancers (Basel) 2021; 13:4491. [PMID: 34503303 PMCID: PMC8430925 DOI: 10.3390/cancers13174491] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022] Open
Abstract
The human UDP-glycosyltransferase (UGTs) superfamily has 22 functional enzymes that play a critical role in the metabolism of small lipophilic compounds, including carcinogens, drugs, steroids, lipids, fatty acids, and bile acids. The expression profiles of UGT genes in human cancers and their impact on cancer patient survival remains to be systematically investigated. In the present study, a comprehensive analysis of the RNAseq and clinical datasets of 9514 patients from 33 different TCGA (the Genome Cancer Atlas) cancers demonstrated cancer-specific UGT expression profiles with high interindividual variability among and within individual cancers. Notably, cancers derived from drug metabolizing tissues (liver, kidney, gut, pancreas) expressed the largest number of UGT genes (COAD, KIRC, KIRP, LIHC, PAAD); six UGT genes (1A6, 1A9, 1A10, 2A3, 2B7, UGT8) showed high expression in five or more different cancers. Kaplan-Meier plots and logrank tests revealed that six UGT genes were significantly associated with increased overall survival (OS) rates [UGT1A1 (LUSC), UGT1A6 (ACC), UGT1A7 (ACC), UGT2A3 (KIRC), UGT2B15 (BLCA, SKCM)] or decreased OS rates [UGT2B15 (LGG), UGT8 (UVM)] in specific cancers. Finally, differential expression analysis of 611 patients from 12 TCGA cancers identified 16 UGT genes (1A1, 1A3, 1A6, 1A7, 1A8, 1A9, 1A10, 2A1, 2A3, 2B4, 2B7, 2B11, 2B15, 3A1, 3A2, UGT8) that were up/downregulated in at least one cancer relative to normal tissues. In conclusion, our data show widespread expression of UGT genes in cancers, highlighting the capacity for intratumoural drug metabolism through the UGT conjugation pathway. The data also suggests the potentials for specific UGT genes to serve as prognostic biomarkers or therapeutic targets in cancers.
Collapse
Affiliation(s)
- Dong Gui Hu
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Shashikanth Marri
- Dicipline of Molecular Medicine and Pathology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia;
| | - Peter I. Mackenzie
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Julie-Ann Hulin
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Ross A. McKinnon
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Robyn Meech
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| |
Collapse
|
24
|
Takahashi RH, Forrest WF, Smith AD, Badee J, Qiu N, Schmidt S, Collier AC, Parrott N, Fowler S. Characterization of Hepatic UDP-Glucuronosyltransferase Enzyme Abundance-Activity Correlations and Population Variability Using a Proteomics Approach and Comparison with Cytochrome P450 Enzymes. Drug Metab Dispos 2021; 49:760-769. [PMID: 34187837 DOI: 10.1124/dmd.121.000474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/24/2021] [Indexed: 11/22/2022] Open
Abstract
The expression of ten major drug-metabolizing UDP-glucuronosyltransferase (UGT) enzymes in a panel of 130 human hepatic microsomal samples was measured using a liquid chromatography-tandem mass spectrometry-based approach. Simultaneously, ten cytochromes P450 and P450 reductase were also measured, and activity-expression relationships were assessed for comparison. The resulting data sets demonstrated that, with the exception of UGT2B17, 10th to 90th percentiles of UGT expression spanned 3- to 8-fold ranges. These ranges were small relative to ranges of reported mean UGT enzyme expression across different laboratories. We tested correlation of UGT expression with enzymatic activities using selective probe substrates. A high degree of abundance-activity correlation (Spearman's rank correlation coefficient > 0.6) was observed for UGT1As (1A1, 3, 4, 6) and cytochromes P450. In contrast, protein abundance and activity did not correlate strongly for UGT1A9 and UGT2B enzymes (2B4, 7, 10, 15, and 17). Protein abundance was strongly correlated for UGTs 2B7, 2B10, and 2B15. We suggest a number of factors may contribute to these differences including incomplete selectivity of probe substrates, correlated expression of these UGT2B isoforms, and the impact of splice and polymorphic variants on the peptides used in proteomics analysis, and exemplify this in the case of UGT2B10. Extensive correlation analyses identified important criteria for validating the fidelity of proteomics and enzymatic activity approaches for assessing UGT variability, population differences, and ontogenetic changes. SIGNIFICANCE STATEMENT: Protein expression data allow detailed assessment of interindividual variability and enzyme ontogeny. This study has observed that expression and enzyme activity are well correlated for hepatic UGT1A enzymes and cytochromes P450. However, for the UGT2B family, caution is advised when assuming correlation of expression and activity as is often done in physiologically based pharmacokinetic modeling. This can be due to incomplete probe substrate specificities, but may also be related to presence of inactive UGT protein materials and the effect of splicing variations.
Collapse
Affiliation(s)
- Ryan H Takahashi
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - William F Forrest
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Alexander D Smith
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Justine Badee
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - NaHong Qiu
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Stephan Schmidt
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Abby C Collier
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Neil Parrott
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| | - Stephen Fowler
- Department of Drug Metabolism and Pharmacokinetics (R.H.T.) and Department of OMNI Bioinformatics (W.F.F.), Genentech, Inc., South San Francisco, California; Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (N.Q., N.P., S.F.); Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada (A.D.S., A.C.C.)
| |
Collapse
|
25
|
Hu DG, Mackenzie PI, Hulin JA, McKinnon RA, Meech R. Circular RNAs of UDP-Glycosyltransferase ( UGT) Genes Expand the Complexity and Diversity of the UGT Transcriptome. Mol Pharmacol 2021; 99:488-503. [PMID: 33824186 DOI: 10.1124/molpharm.120.000225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/26/2021] [Indexed: 12/27/2022] Open
Abstract
The human UDP-glycosyltransferase (UGT) gene superfamily generates 22 canonical transcripts coding for functional enzymes and also produces nearly 150 variant UGT transcripts through alternative splicing and intergenic splicing. In the present study, our analysis of circRNA databases identified backsplicing events that predicted 85 circRNAs from UGT genes, with 33, 11, and 19 circRNAs from UGT1A, UGT2B4, UGT8, respectively. Most of these UGT circRNAs were reported by one database and had low abundance in cell- or tissue-specific contexts. Using reverse-transcriptase polymerase chain reaction with divergent primers and cDNA samples from human tissues and cell lines, we found 13 circRNAs from four UGT genes: UGT1A (three), UGT2B7 (one), UGT2B10 (one), and UGT8 (eight). Notably, all eight UGT8 circRNAs contain open reading frames that include the canonical start AUG codon and encode variant proteins that all have the common 274-amino acidN-terminal region of wild-type UGT8 protein. We further showed that one UGT8 circRNA (circ_UGT8-1) was broadly expressed in human tissues and cell lines, resistant to RNase R digestion, and predominately present in the cytoplasm. We cloned five UGT8 circRNAs into the Zinc finger with KRAB and SCAN domains 1 vector and transfected them into HEK293T cells. All these vectors produced both circRNAsand linear transcripts with varying circular/linear ratios (0.17-1.14).Western blotting and mass spectrometry assays revealed that only linear transcripts and not circRNAs were translated. In conclusion, our findings of nearly 100 circRNAs greatly expand the complexity and diversity of the UGT transcriptome; however, UGT circRNAs are expressed at a very low level in specific cellular contexts, and their biologic functions remain to be determined. SIGNIFICANCE STATEMENT: The human UGT gene transcriptome comprises 22 canonical transcripts coding for functional enzymes and approximately 150 alternatively spliced and chimeric variant transcripts. The present study identified nearly 100 circRNAs from UGT genes, thus greatly expanding the complexity and diversity of the UGT transcriptome. UGT circRNAs were expressed broadly in human tissues and cell lines; however, most showed very low abundance in tissue- and cell-specific contexts, and therefore their biological functions remain to be investigated.
Collapse
Affiliation(s)
- Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Peter I Mackenzie
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Julie-Ann Hulin
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Ross A McKinnon
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Robyn Meech
- Department of Clinical Pharmacology and Flinders Cancer Centre, Flinders University, College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
26
|
Yin J, Li F, Zhou Y, Mou M, Lu Y, Chen K, Xue J, Luo Y, Fu J, He X, Gao J, Zeng S, Yu L, Zhu F. INTEDE: interactome of drug-metabolizing enzymes. Nucleic Acids Res 2021; 49:D1233-D1243. [PMID: 33045737 PMCID: PMC7779056 DOI: 10.1093/nar/gkaa755] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/19/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Drug-metabolizing enzymes (DMEs) are critical determinant of drug safety and efficacy, and the interactome of DMEs has attracted extensive attention. There are 3 major interaction types in an interactome: microbiome-DME interaction (MICBIO), xenobiotics-DME interaction (XEOTIC) and host protein-DME interaction (HOSPPI). The interaction data of each type are essential for drug metabolism, and the collective consideration of multiple types has implication for the future practice of precision medicine. However, no database was designed to systematically provide the data of all types of DME interactions. Here, a database of the Interactome of Drug-Metabolizing Enzymes (INTEDE) was therefore constructed to offer these interaction data. First, 1047 unique DMEs (448 host and 599 microbial) were confirmed, for the first time, using their metabolizing drugs. Second, for these newly confirmed DMEs, all types of their interactions (3359 MICBIOs between 225 microbial species and 185 DMEs; 47 778 XEOTICs between 4150 xenobiotics and 501 DMEs; 7849 HOSPPIs between 565 human proteins and 566 DMEs) were comprehensively collected and then provided, which enabled the crosstalk analysis among multiple types. Because of the huge amount of accumulated data, the INTEDE made it possible to generalize key features for revealing disease etiology and optimizing clinical treatment. INTEDE is freely accessible at: https://idrblab.org/intede/.
Collapse
Affiliation(s)
- Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinjing Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kangli Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jia Xue
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianbo Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xu He
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Lushan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| |
Collapse
|
27
|
Kellerová P, Navrátilová M, Nguyen LT, Dimunová D, Raisová Stuchlíková L, Štěrbová K, Skálová L, Matoušková P. UDP-Glycosyltransferases and Albendazole Metabolism in the Juvenile Stages of Haemonchus contortus. Front Physiol 2020; 11:594116. [PMID: 33324241 PMCID: PMC7726322 DOI: 10.3389/fphys.2020.594116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/05/2020] [Indexed: 11/17/2022] Open
Abstract
The nematode Haemonchus contortus, a gastrointestinal parasite of ruminants, can severely burden livestock production. Although anthelmintics are the mainstay in the treatment of haemonchosis, their efficacy diminishes due to drug-resistance development in H. contortus. An increased anthelmintics inactivation via biotransformation belongs to a significant drug-resistance mechanism in H. contortus. UDP-glycosyltransferases (UGTs) participate in the metabolic inactivation of anthelmintics and other xenobiotic substrates through their conjugation with activated sugar, which drives the elimination of the xenobiotics due to enhanced solubility. The UGTs family, in terms of the biotransformation of commonly used anthelmintics, has been well described in adults as a target stage. In contrast, the free-living juvenile stages of H. contortus have attracted less attention. The expression of UGTs considerably varies throughout the life cycle of the juvenile nematodes, suggesting their different roles. Furthermore, the constitutive expression in a susceptible strain with two resistant strains shows several resistance-related changes in UGTs expression, and the exposure of juvenile stages of H. contortus to albendazole (ABZ) and ABZ-sulfoxide (ABZSO; in sublethal concentrations) leads to the increased expression of several UGTs. The anthelmintic drug ABZ and its primary metabolite ABZSO biotransformation, tested in the juvenile stages, shows significant differences between susceptible and resistant strain. Moreover, higher amounts of glycosidated metabolites of ABZ are formed in the resistant strain. Our results show similarly, as in adults, the UGTs and glycosidations significant for resistance-related differences in ABZ biotransformation and warrant further investigation in their individual functions.
Collapse
Affiliation(s)
- Pavlína Kellerová
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, Hradec Králové, Czechia
| | - Martina Navrátilová
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, Hradec Králové, Czechia
| | - Linh Thuy Nguyen
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, Hradec Králové, Czechia
| | - Diana Dimunová
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, Hradec Králové, Czechia
| | - Lucie Raisová Stuchlíková
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, Hradec Králové, Czechia
| | - Karolína Štěrbová
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, Hradec Králové, Czechia
| | - Lenka Skálová
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, Hradec Králové, Czechia
| | - Petra Matoušková
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, Hradec Králové, Czechia
| |
Collapse
|
28
|
The Expression Profiles of ADME Genes in Human Cancers and Their Associations with Clinical Outcomes. Cancers (Basel) 2020; 12:cancers12113369. [PMID: 33202946 PMCID: PMC7697355 DOI: 10.3390/cancers12113369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
ADME genes are a group of genes that are involved in drug absorption, distribution, metabolism, and excretion (ADME). The expression profiles of ADME genes within tumours is proposed to impact on cancer patient survival; however, this has not been systematically examined. In this study, our comprehensive analyses of pan-cancer datasets from the Cancer Genome Atlas (TCGA) revealed differential intratumoral expression profiles for ADME genes in 21 different cancer types. Most genes also showed high interindividual variability within cancer-specific patient cohorts. Using Kaplan-Meier plots and logrank tests, we showed that intratumoral expression levels of twenty of the thirty-two core ADME genes were associated with overall survival (OS) in these cancers. Of these genes, five showed significant association with unfavourable OS in three cancers, including SKCM (ABCC2, GSTP1), KIRC (CYP2D6, CYP2E1), PAAD (UGT2B7); sixteen showed significant associations with favourable OS in twelve cancers, including BLCA (UGT2B15), BRCA (CYP2D6), COAD (NAT1), HNSC (ABCB1), KIRC (ABCG2, CYP3A4, SLC22A2, SLC22A6), KIRP (SLC22A2), LIHC (CYP2C19, CYP2C8, CYP2C9, CYP3A5, SLC22A1), LUAD (SLC15A2), LUSC (UGT1A1), PAAD (ABCB1), SARC (ABCB1), and SKCM (ABCB1, DYPD). Overall, these data provide compelling evidence supporting ADME genes as prognostic biomarkers and potential therapeutic targets. We propose that intratumoral expression of ADME genes may impact cancer patient survival by multiple mechanisms that can include metabolizing/transporting anticancer drugs, activating anticancer drugs, and metabolizing/transporting a variety of endogenous molecules involved in metabolically fuelling cancer cells and/or controlling pro-growth signalling pathways.
Collapse
|
29
|
Mikolajczyk K, Kaczmarek R, Czerwinski M. How glycosylation affects glycosylation: the role of N-glycans in glycosyltransferase activity. Glycobiology 2020; 30:941-969. [PMID: 32363402 DOI: 10.1093/glycob/cwaa041] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022] Open
Abstract
N-glycosylation is one of the most important posttranslational modifications of proteins. It plays important roles in the biogenesis and functions of proteins by influencing their folding, intracellular localization, stability and solubility. N-glycans are synthesized by glycosyltransferases, a complex group of ubiquitous enzymes that occur in most kingdoms of life. A growing body of evidence shows that N-glycans may influence processing and functions of glycosyltransferases, including their secretion, stability and substrate/acceptor affinity. Changes in these properties may have a profound impact on glycosyltransferase activity. Indeed, some glycosyltransferases have to be glycosylated themselves for full activity. N-glycans and glycosyltransferases play roles in the pathogenesis of many diseases (including cancers), so studies on glycosyltransferases may contribute to the development of new therapy methods and novel glycoengineered enzymes with improved properties. In this review, we focus on the role of N-glycosylation in the activity of glycosyltransferases and attempt to summarize all available data about this phenomenon.
Collapse
Affiliation(s)
- Krzysztof Mikolajczyk
- Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland
| | - Radoslaw Kaczmarek
- Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland
| | - Marcin Czerwinski
- Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland
| |
Collapse
|
30
|
Mertansine Inhibits mRNA Expression and Enzyme Activities of Cytochrome P450s and Uridine 5′-Diphospho-Glucuronosyltransferases in Human Hepatocytes and Liver Microsomes. Pharmaceutics 2020; 12:pharmaceutics12030220. [PMID: 32131538 PMCID: PMC7150891 DOI: 10.3390/pharmaceutics12030220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 02/28/2020] [Accepted: 03/01/2020] [Indexed: 11/16/2022] Open
Abstract
Mertansine, a tubulin inhibitor, is used as the cytotoxic component of antibody–drug conjugates (ADCs) for cancer therapy. The effects of mertansine on uridine 5′-diphospho-glucuronosyltransferase (UGT) activities in human liver microsomes and its effects on the mRNA expression of cytochrome P450s (CYPs) and UGTs in human hepatocytes were evaluated to assess the potential for drug–drug interactions (DDIs). Mertansine potently inhibited UGT1A1-catalyzed SN-38 glucuronidation, UGT1A3-catalyzed chenodeoxycholic acid 24-acyl-β-glucuronidation, and UGT1A4-catalyzed trifluoperazine N-β-d-glucuronidation, with Ki values of 13.5 µM, 4.3 µM, and 21.2 µM, respectively, but no inhibition of UGT1A6, UGT1A9, and UGT2B7 enzyme activities was observed in human liver microsomes. A 48 h treatment of mertansine (1.25–2500 nM) in human hepatocytes resulted in the dose-dependent suppression of mRNA levels of CYP1A2, CYP2B6, CYP3A4, CYP2C8, CYP2C9, CYP2C19, UGT1A1, and UGT1A9, with IC50 values of 93.7 ± 109.1, 36.8 ± 18.3, 160.6 ± 167.4, 32.1 ± 14.9, 578.4 ± 452.0, 539.5 ± 233.4, 856.7 ± 781.9, and 54.1 ± 29.1 nM, respectively, and decreased the activities of CYP1A2-mediated phenacetin O-deethylase, CYP2B6-mediated bupropion hydroxylase, and CYP3A4-mediated midazolam 1′-hydroxylase. These in vitro DDI potentials of mertansine with CYP1A2, CYP2B6, CYP2C8/9/19, CYP3A4, UGT1A1, and UGT1A9 substrates suggest that it is necessary to carefully characterize the DDI potentials of ADC candidates with mertansine as a payload in the clinic.
Collapse
|
31
|
Hetero-oligomer formation of mouse UDP-glucuronosyltransferase (UGT) 2b1 and 1a1 results in the gain of glucuronidation activity towards morphine, an activity which is absent in homo-oligomers of either UGT. Biochem Biophys Res Commun 2020; 525:348-353. [PMID: 32093886 DOI: 10.1016/j.bbrc.2020.02.075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/11/2020] [Indexed: 01/11/2023]
Abstract
UDP-Glucuronosyltransferase (UGT, Ugt) is a major drug metabolizing enzyme family involved in the glucuronidation and subsequent elimination of drugs and small lipophilic molecules. UGT forms homo- and hetero-oligomers that enhance or suppress UGT activity. In our previous study, we characterized mouse Ugt1a1 and all the Ugt isoform belonging to the Ugt2b subfamily and revealed that mouse Ugt2b1 and Ugt1a1 cannot metabolize morphine. Mouse Ugt2b1 had been believed to function similarly to rat UGT2B1, which plays a major role in morphine glucuronidation in rat liver. Thus, in this study, we hypothesized that hetero-oligomerization with another Ugt isoform may affect Ugt2b1 catalytic ability. We co-expressed Ugt1a1 and Ugt2b1 in a baculovirus-insect cell system, and confirmed hetero-oligomer formation by co-immunoprecipitation. As reported previously, microsomes singly expressing Ugt1a1 or Ugt2b1 were inactive towards the glucuronidation of morphine. Interestingly, in contrast, morphine-3-glucuronide, a major metabolite of morphine was formed, when Ugt2b1 and Ugt1a1 were co-expressed. This effect of hetero-oligomerization of Ugt1a1 and Ugt2b1 was also observed for 17β-estradiol glucuronidation. This is the first report demonstrating that UGT acquires a novel catalytic ability by forming oligomers. Protein-protein interaction of Ugts may contribute to robust detoxification of xenobiotics by altering the substrate diversity of the enzymes.
Collapse
|