1
|
Corachea AJM, Ferrer RJE, Ty LPB, Aquino LAC, Morta MT, Macalindong SS, Uy GLB, Odoño EG, Llames JHS, Tablizo FA, Cutiongco-Dela Paz EMC, Dofitas RB, Velarde MC. Lymphovascular Invasion Is Associated With Doxorubicin Resistance in Breast Cancer. J Transl Med 2025; 105:104115. [PMID: 39978641 DOI: 10.1016/j.labinv.2025.104115] [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: 08/07/2024] [Revised: 01/30/2025] [Accepted: 02/08/2025] [Indexed: 02/22/2025] Open
Abstract
Lymphovascular invasion (LVI), the invasion of tumor cells into the lymphatic or vascular space, is an early indicator of potential metastasis, with its presence in breast cancer independently predicting poorer outcomes even after neoadjuvant chemotherapy. However, a major limitation is that LVI detection currently relies on postsurgical evaluation. To address this, we determined whether LVI+ breast tumors contain a unique gene signature that could facilitate earlier detection. Here, we conducted an integrative analysis of the gene profile between LVI+ and LVI- primary breast tumors from various sources, including published data and our own research, using both microarray and RNA-seq data. Our analysis revealed protein binding and vesicle-related genes to be the most enriched categories in LVI+ vs LVI- tumors. Furthermore, LVI+ tumors showed enrichment for xenobiotic metabolism genes, particularly drug metabolism enzymes, such as cytochrome P450 and uridine 5'-diphospho-glucuronosyltransferases. An elastic net regression model containing 13 of these uridine 5'-diphospho-glucuronosyltransferases and cytochrome P450 genes can predict LVI status with 92% accuracy. This suggests a potential link to drug resistance, which was further confirmed by the finding that patients with LVI+ tumors had a significantly lower clinical response rate than individuals with LVI- tumors. We also observed this resistance in patient-derived organoids, with LVI+ organoids exhibiting lower sensitivity to doxorubicin, implying that doxorubicin might be less effective for LVI+ breast cancer, potentially contributing to poorer outcomes. Overall, our study unlocked an exciting opportunity for personalized medicine, in that, therapy efficacy and patient outcomes can be improved by incorporating the LVI-associated gene signature into treatment plans.
Collapse
Affiliation(s)
- Allen Joy M Corachea
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Regina Joyce E Ferrer
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Lance Patrick B Ty
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Lizzie Anne C Aquino
- Department of Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Madeleine T Morta
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Shiela S Macalindong
- Department of Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Gemma Leonora B Uy
- Department of Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Eugene G Odoño
- Department of Pathology, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Jo-Hannah S Llames
- Philippine Genome Center, University of the Philippines, Diliman, Quezon City, Philippines
| | - Francis A Tablizo
- Philippine Genome Center, University of the Philippines, Diliman, Quezon City, Philippines
| | | | - Rodney B Dofitas
- Department of Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Michael C Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines.
| |
Collapse
|
2
|
Rivera-Herrera AL, Rouleau M, Singbo MNU, Cuppens T, Prunier J, Droit A, Simonyan D, Guillemette C. Sexual dimorphism in metabolomic and phenotypic spectra of UGT deficiency: findings from the Canadian Longitudinal Study on Aging. Biol Sex Differ 2025; 16:26. [PMID: 40264209 PMCID: PMC12013048 DOI: 10.1186/s13293-025-00708-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/29/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Two of the most frequently deleted genes in the human genome are the UDP-glycosyltransferases UGT2B17 and UGT2B28. They encode metabolic enzymes of the glucuronidation pathway that plays a pivotal role in the maintenance of cellular homeostasis for a variety of small molecule metabolites. These deletions may impact health, yet their effects remain poorly understood. We evaluated the impact of UGT deficiency on the plasma metabolome and examined the association between altered metabolites and health outcomes. METHODS The metabolomic profiles of 4262 proficient gene carriers were compared with those of 352 UGT2B17-deficient, 97 UGT2B28-deficient, and 20 double-gene-deficient individuals from the Canadian Longitudinal Study on Aging. Significant metabolites found in these comparisons were analyzed for their associations with common diseases. RESULTS The unexpectedly broad molecular divergence found in UGT-deficient metabolomes, which affected > 10% of metabolites, implies their significant influence across various metabolite classes-particularly lipids and amino acids - extending beyond their known substrates. The metabolic profiles of UGT2B17-deficient men and UGT2B28-deficient women were most impacted, with UGT2B17 deficiency affecting various metabolites linked to metabolic diseases, arthritis, and osteoporosis. Metabolites impacted by a UGT2B28 deficiency such as amino acids, were linked to metabolic disorders in women. CONCLUSION The findings significantly advance our understanding of the metabolic landscape associated with these frequently deleted genes in the human genome, which may influence susceptibility to various diseases in a sex-specific manner, laying the groundwork for determining their pathological mechanisms and impact on human health.
Collapse
Affiliation(s)
- Ana Lucia Rivera-Herrera
- Faculty of Pharmacy, Université Laval, Québec, QC, G1V 0A6, Canada
- Université Laval Cancer Research Center, Québec, QC, G1R 3S3, Canada
- Centre Hospitalier Universitaire (CHU) de Québec Research Center - Université Laval, R4701.5, 2705 Blvd Laurier Québec, Québec, QC, G1V 4G2, Canada
| | - Michèle Rouleau
- Faculty of Pharmacy, Université Laval, Québec, QC, G1V 0A6, Canada
- Université Laval Cancer Research Center, Québec, QC, G1R 3S3, Canada
- Centre Hospitalier Universitaire (CHU) de Québec Research Center - Université Laval, R4701.5, 2705 Blvd Laurier Québec, Québec, QC, G1V 4G2, Canada
| | - Mahukpe Narcisse Ulrich Singbo
- Centre Hospitalier Universitaire (CHU) de Québec Research Center - Université Laval, R4701.5, 2705 Blvd Laurier Québec, Québec, QC, G1V 4G2, Canada
| | - Tania Cuppens
- Université Laval Cancer Research Center, Québec, QC, G1R 3S3, Canada
- Centre Hospitalier Universitaire (CHU) de Québec Research Center - Université Laval, R4701.5, 2705 Blvd Laurier Québec, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Julien Prunier
- Université Laval Cancer Research Center, Québec, QC, G1R 3S3, Canada
- Centre Hospitalier Universitaire (CHU) de Québec Research Center - Université Laval, R4701.5, 2705 Blvd Laurier Québec, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Arnaud Droit
- Université Laval Cancer Research Center, Québec, QC, G1R 3S3, Canada
- Centre Hospitalier Universitaire (CHU) de Québec Research Center - Université Laval, R4701.5, 2705 Blvd Laurier Québec, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - David Simonyan
- Centre Hospitalier Universitaire (CHU) de Québec Research Center - Université Laval, R4701.5, 2705 Blvd Laurier Québec, Québec, QC, G1V 4G2, Canada
| | - Chantal Guillemette
- Faculty of Pharmacy, Université Laval, Québec, QC, G1V 0A6, Canada.
- Université Laval Cancer Research Center, Québec, QC, G1R 3S3, Canada.
- Centre Hospitalier Universitaire (CHU) de Québec Research Center - Université Laval, R4701.5, 2705 Blvd Laurier Québec, Québec, QC, G1V 4G2, Canada.
| |
Collapse
|
3
|
Xue J, Li Q, Wang Y, Yin R, Zhang J. Insight into the structure, oligomerization, and the role in drug resistance of human UDP-glucuronosyltransferases. Arch Toxicol 2025; 99:1153-1165. [PMID: 39812829 DOI: 10.1007/s00204-024-03929-6] [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: 10/12/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025]
Abstract
Human UDP-glucuronosyltransferases (UGTs) are pivotal phase II metabolic enzymes facilitating the transfer of glucuronic acid from UDP-glucuronic acid (UDPGA) to various substrates. UGTs are classic type I transmembrane glycoproteins, mainly localized in the endoplasmic reticulum (ER) membrane. This review comprehensively explores UGTs, encompassing gene expression, functional characteristics, substrate specificity, and metabolic mechanisms. A recent analysis of C-terminal structures, compared with original data, underscores the pivotal role of α3, α4, and β4 functional domains in selectively recognizing diverse glycosyl donors. Accumulating evidence suggests that UGTs function as homo- and heterodimers, with oligomers likely stabilizing UGTs and modulating their activity. The review sheds light on the implications of UGT oligomerization on substrate glucuronidation and the interplay between protein-protein interaction and glucuronidation activity. UGT-mediated drug resistance, often underestimated, emerges as a clinically relevant form of chemical resistance, with delineated outcomes in tumors and other diseases. This review provides a multifaceted exploration of the physiological significance of UGTs, spanning genetics, proteins, oligomerization, drug resistance, and more, offering insights into their metabolic mechanisms. Understanding interactions between UGT isoforms is crucial for predicting drug-drug interactions, preventing drug toxicity, and enabling precision treatment.
Collapse
Affiliation(s)
- Jia Xue
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuyi Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yao Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruxi Yin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jian Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Bennani Y, Ben Hassine K, Gencaslan M, Boudal‐Khoshbeen M, Samer C, Ansari M, Daali Y, Satyanarayana Uppugunduri CR. In vitro screening of UGT2B10 in silico prioritized putative ligands from drugs used in the pediatric hematopoietic stem cell transplantation setting. Pharmacol Res Perspect 2024; 12:e70011. [PMID: 39611692 PMCID: PMC11605732 DOI: 10.1002/prp2.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 11/30/2024] Open
Abstract
UGT2B10 is a phase II drug metabolizing enzyme with limited information on its role in the metabolism of drugs, especially in the pediatric hematopoietic stem cell transplantation setting. Previously, we investigated UGT2B10's role through in silico analyses and prioritized acetaminophen (APAP), lorazepam (LOR), mycophenolic acid (MPA), and voriconazole N-oxide (VCZ N-oxide) for in vitro investigations. In this report, we present in vitro screening of these candidates and of voriconazole (VCZ) to assess their potential to be substrates and/or inhibitors of UGT2B10. Enzyme kinetics experiments included recombinant UGT2B10 and analytical methods based on ultra high-performance liquid chromatography coupled to mass spectrometry (UHPLC-MS). To determine potential substrates, candidates were incubated at various therapeutically observed concentrations with recombinant UGT2B10 to identify the corresponding glucuronide metabolite. Inhibition capacity was tested using the selective probe cotinine for its glucuronidation to cotinine N-ß-d-glucuronide. IC50 was determined for compounds exhibiting inhibition. Among the tested compounds, LOR (IC50 = 0.01 μM, R2 = 0.9257) and MPA (IC50 = 0.38 mM, R2 = 0.9212) exhibited inhibition potential for UGT2B10. None of the other tested compounds featured inhibition potential and none of the compounds tested exhibited metabolism through UGT2B10. Further exploration on the clinical relevance of this inhibition using modeling strategies, overlapping nature with other UGT isoforms, and screening other molecules for their inhibition potential on UGT2B10 is warranted.
Collapse
Affiliation(s)
- Yahia Bennani
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Geneva Lausanne School of PharmacyUniversity of GenevaGenevaSwitzerland
| | - Khalil Ben Hassine
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and ObstetricsUniversity of GenevaGenevaSwitzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
| | | | - Mary Boudal‐Khoshbeen
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and ObstetricsUniversity of GenevaGenevaSwitzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
| | - Caroline Samer
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Geneva Lausanne School of PharmacyUniversity of GenevaGenevaSwitzerland
- Swiss Center for Applied Human ToxicologyGenevaSwitzerland
- Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Marc Ansari
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and ObstetricsUniversity of GenevaGenevaSwitzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and ToxicologyGeneva University HospitalsGenevaSwitzerland
- Geneva Lausanne School of PharmacyUniversity of GenevaGenevaSwitzerland
- Swiss Center for Applied Human ToxicologyGenevaSwitzerland
- Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Chakradhara Rao Satyanarayana Uppugunduri
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and ObstetricsUniversity of GenevaGenevaSwitzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
- Department of Medical OncologyJawaharlal Institute of Postgraduate Medical Education and ResearchPuducherryIndia
| |
Collapse
|
5
|
Lapham K, Ferguson N, Niosi M, Goosen TC. Clotrimazole Identified as a Selective UGT2B4 Inhibitor Using Canagliflozin-2'- O-Glucuronide Formation as a Selective UGT2B4 Probe Reaction. Drug Metab Dispos 2024; 52:1083-1093. [PMID: 39142826 DOI: 10.1124/dmd.124.001812] [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: 05/20/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
UGT2B4 is a highly expressed drug-metabolizing enzyme in the liver contributing to the glucuronidation of several drugs. To enable quantitatively assessing UGT2B4 contribution toward metabolic clearance, a potent and selective UGT2B4 inhibitor that can be used for reaction phenotyping was sought. Initially, a canagliflozin-2'-O-glucuronyl transferase activity assay was developed in recombinant UGT2B4 and human liver microsomes (HLM) [±2% bovine serum albumin (BSA)]. Canagliflozin-2'-O-glucuronidation (C2OG) substrate concentration at half-maximal velocity value in recombinant UGT2B4 and HLM were similar. C2OG formation intrinsic clearance was five- to seven-fold higher in incubations containing 2% BSA, suggesting UGT2B4 susceptibility to the inhibitory unsaturated long-chain fatty acids released during the incubation. Monitoring for C2OG formation, 180 compounds were evaluated for UGT2B4 inhibition potency in the presence and absence of 2% BSA. Compounds that exhibited an apparent UGT2B4 IC50 of < 1 μM in HLM with 2% BSA were evaluated for inhibition of UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2B7, UGT2B10, UGT2B15, and UGT2B17 catalytic activities to establish selectivity suitable for supporting UGT reaction phenotyping. In this study, clotrimazole was identified as a potent UGT2B4 inhibitor (HLM apparent IC50 of 11 to 35 nM ± 2% BSA). Moreover, clotrimazole exhibited selectivity for UGT2B4 inhibition (>24-fold) over the other UGT enzymes evaluated. Additionally, during this study it was discovered that the previously described UGT2B7 inhibitors 16α- and 16β-phenyllongifolol also inhibit UGT2B4. Clotrimazole, a potent and selective UGT2B4 inhibitor, will prove essential during UGT reaction phenotyping. SIGNIFICANCE STATEMENT: To mechanistically evaluate drug interactions, it is essential to understand the contribution of individual enzymes to the metabolic clearance of a drug. The present study describes the development of a UGT2B4 activity assay that enabled the discovery of the highly selective and potent UGT2B4 inhibitor clotrimazole. Clotrimazole can be used in UGT reaction phenotyping studies to estimate fractional contribution of UGT2B4.
Collapse
Affiliation(s)
- Kimberly Lapham
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Groton, Connecticut
| | - Nicholas Ferguson
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Groton, Connecticut
| | - Mark Niosi
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Groton, Connecticut
| | - Theunis C Goosen
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Groton, Connecticut
| |
Collapse
|
6
|
Yin H, Wang Z, Lv X, Wang Z, Wang Y, Fan W, Li S, Jiang L, Cao J, Liu Y. Inhibition of human UDP-glucuronosyltransferase enzyme by entrectinib: Implications for drug-drug interactions. Chem Biol Interact 2024; 395:111023. [PMID: 38677539 DOI: 10.1016/j.cbi.2024.111023] [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: 02/21/2024] [Revised: 04/13/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
As a new type of oral tyrosine kinase inhibitor, entrectinib can act on multiple targets and exert efficacy and has been approved for the treatment of non-small cell lung cancer (NSCLC) and solid tumors. However, whether entrectinib affects the activities of recombinant human UDP-glucuronosyltransferases (UGTs) remains unclear. Herein, we aimed to investigate the inhibitory effects of entrectinib on human UGTs and to assess the potential risk of causing drug-drug interactions (DDIs) based on the inhibition against UGTs. High-performance liquid chromatography (HPLC) was used to evaluate the inhibitory effects of entrectinib on UGTs according to the product formation rate of UGT substrate with or without entrectinib, and the inhibition kinetics experiment was conducted to assess the inhibitory type of entrectinib on UGTs. Our results showed that entrectinib exhibited extensive inhibitory effects on most human UGTs, and especially inhibited the activities of UGT1A7, UGT1A8, and UGT2B15 with Ki (Inhibition constant) of lower than 5 μM (0.95-4.38 μM). Furthermore, the results from quantitative prediction research suggested that the combination of entrectinib at 600 mg/day with substrates primarily metabolized by hepatic UGT2B15 or intestinal UGT1A7 and UGT1A8 might cause clinical DDIs. Thus, special attention should be paid to avoid adverse reactions induced by DDIs when co-administration of entrectinib and drugs metabolized by UGTs.
Collapse
Affiliation(s)
- Hang Yin
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhe Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xin Lv
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhen Wang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Ying Wang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Wenxuan Fan
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Shuang Li
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Lili Jiang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Jun Cao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, 116044, China.
| | - Yong Liu
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China.
| |
Collapse
|
7
|
Wang L, Wang L, Sun X, Fu L, Wang X, Wang X, Chen L, Huang Y. Detection of uridine diphosphate glucuronosyltransferase 1A1 for pancreatic cancer imaging and treatment via a "turn-on" fluorescent probe. Analyst 2024; 149:2877-2886. [PMID: 38567989 DOI: 10.1039/d4an00035h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) is expressed ubiquitously in cancer cells and can metabolize exogenous substances. Studies show higher UGT1A1 levels in pancreatic cancer cells than normal cells. Therefore, we need a method to monitor the activity level of UGT1A1 in pancreatic cancer cells and in vivo. Here, we report a fluorescent probe, BCy-panc, for UGT1A1 imaging in cells and in vivo. Compared with other molecular probes, this probe is readily prepared, with high selectivity and sensitivity for the detection of UGT1A1. Our results show that BCy-panc rapidly detects UGT1A1 in pancreatic cancer. In addition, there is an urgent need for evidence to clarify the relationship between UGT1A1 and pancreatic cancer development. The present investigation found that the increase of UGT1A1 by chrysin was effective in inducing apoptosis in pancreatic cancer cells. These results indicate that the synergistic effect of chrysin and cisplatin at the cellular level is superior to that of cisplatin alone. The UGT1A1 level may be a biomarker for early diagnosis of cancer. Meanwhile, UGT1A1 plays a crucial role in pancreatic cancer, and the combination of chrysin and cisplatin may provide effective ideas for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Lingxiao Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Lingyun Wang
- Jinan Zhangqiu District People's Hospital, Jinan 250000, China
| | - Xiao Sun
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Lili Fu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Xinlei Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Xiaoyan Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Lingxin Chen
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yan Huang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| |
Collapse
|
8
|
Cortez N, Villegas C, Burgos V, Ortiz L, Cabrera-Pardo JR, Paz C. Therapeutic Potential of Chlorogenic Acid in Chemoresistance and Chemoprotection in Cancer Treatment. Int J Mol Sci 2024; 25:5189. [PMID: 38791228 PMCID: PMC11121551 DOI: 10.3390/ijms25105189] [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: 03/13/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Chemotherapeutic drugs are indispensable in cancer treatment, but their effectiveness is often lessened because of non-selective toxicity to healthy tissues, which triggers inflammatory pathways that are harmful to vital organs. In addition, tumors' resistance to drugs causes failures in treatment. Chlorogenic acid (5-caffeoylquinic acid, CGA), found in plants and vegetables, is promising in anticancer mechanisms. In vitro and animal studies have indicated that CGA can overcome resistance to conventional chemotherapeutics and alleviate chemotherapy-induced toxicity by scavenging free radicals effectively. This review is a summary of current information about CGA, including its natural sources, biosynthesis, metabolism, toxicology, role in combatting chemoresistance, and protective effects against chemotherapy-induced toxicity. It also emphasizes the potential of CGA as a pharmacological adjuvant in cancer treatment with drugs such as 5-fluorouracil, cisplatin, oxaliplatin, doxorubicin, regorafenib, and radiotherapy. By analyzing more than 140 papers from PubMed, Google Scholar, and SciFinder, we hope to find the therapeutic potential of CGA in improving cancer therapy.
Collapse
Affiliation(s)
- Nicole Cortez
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (N.C.); (C.V.)
| | - Cecilia Villegas
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (N.C.); (C.V.)
| | - Viviana Burgos
- Departamento de Ciencias Biológicas y Químicas, Facultad de Recursos Naturales, Universidad Católica de Temuco, Rudecindo Ortega, Temuco 4780000, Chile;
| | - Leandro Ortiz
- Instituto de Ciencias Químicas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile;
| | - Jaime R. Cabrera-Pardo
- Laboratorio de Química Aplicada y Sustentable, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000000, Chile;
| | - Cristian Paz
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (N.C.); (C.V.)
| |
Collapse
|
9
|
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
|
10
|
Lv X, Wang Z, Wang Z, Yin H, Xia Y, Jiang L, Liu Y. Avapritinib Carries the Risk of Drug Interaction via Inhibition of UDP-Glucuronyltransferase (UGT) 1A1. Curr Drug Metab 2024; 25:197-204. [PMID: 38803186 DOI: 10.2174/0113892002288312240521092054] [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: 11/28/2023] [Revised: 03/04/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Avapritinib is the only drug for adult patients with PDGFRA exon 18 mutated unresectable or metastatic gastrointestinal stromal tumor (GIST). Although avapritinib has been approved by the FDA for four years, little is known about the risk of drug-drug interactions (DDIs) via UDP-glucuronyltransferases (UGTs) inhibition. OBJECTIVE The aim of the present study was to systematically evaluate the inhibitory effects of avapritinib against UGTs and to quantitatively estimate its potential DDIs risk in vivo. METHODS Recombinant human UGTs were employed to catalyze the glucuronidation of substrates in a range of concentrations of avapritinib. The kinetics analysis was performed to evaluate the inhibition types of avapritinib against UGTs. The quantitative prediction of DDIs was done using in vitro-in vivo extrapolation (IVIVE). RESULTS Avapritinib had a potent competitive inhibitory effect on UGT1A1. Quantitative prediction results showed that avapritinib administered at clinical doses might result in a 14.85% increase in area under the curve (AUC) of drugs primarily cleared by UGT1A1. Moreover, the Rgut value was calculated to be 18.44. CONCLUSION Avapritinib has the potential to cause intestinal DDIs via the inhibition of UGT1A1. Additional attention should be paid when avapritinib is coadministered with UGT1A1 substrates.
Collapse
Affiliation(s)
- Xin Lv
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhen Wang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhe Wang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hang Yin
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yangliu Xia
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Lili Jiang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yong Liu
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| |
Collapse
|
11
|
Yang F, Wenzel M, Bureik M, Parr MK. Glucuronidation Pathways of 5- and 7-Hydroxypropranolol: Determination of Glucuronide Structures and Enzyme Selectivity. Molecules 2023; 28:7783. [PMID: 38067513 PMCID: PMC10707847 DOI: 10.3390/molecules28237783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Propranolol, a non-selective beta-blocker medication, has been utilized in the treatment of cardiovascular diseases for several decades. Its hydroxynaphthyl metabolites have been recognized to possess varying degrees of beta-blocker activity due to the unaltered side-chain. This study achieved the successful separation and identification of diastereomeric glucuronic metabolites derived from 4-, 5-, and 7-hydroxypropranolol (4-OHP, 5-OHP, and 7-OHP) in human urine. Subsequently, reaction phenotyping of 5- and 7-hydroxypropranolol by different uridine 5'-diphospho-glucuronosyltransferases (UGTs) was carried out, with a comparison to the glucuronidation of 4-hydroxypropranolol (4-OHP). Among the 19 UGT enzymes examined, UGT1A1, UGT1A3, UGT1A7, UGT1A8, UGT1A9, UGT1A10, UGT2A1, and UGT2A2 were found to be involved in the glucuronidation of 5-OHP. Furthermore, UGT1A6 exhibited glucuronidation activity towards 7-OHP, along with the aforementioned eight UGTs. Results obtained by glucuronidation of corresponding methoxypropranolols and MS/MS analysis of 1,2-dimethylimidazole-4-sulfonyl (DMIS) derivatives of hydroxypropranolol glucuronides suggest that both the aromatic and aliphatic hydroxy groups of the hydroxypropranolols may be glucuronidated in vitro. However, the analysis of human urine samples collected after the administration of propranolol leads us to conclude that aromatic-linked glucuronidation is the preferred pathway under physiological conditions.
Collapse
Affiliation(s)
- Fan Yang
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; (F.Y.); (M.W.)
| | - Maxi Wenzel
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; (F.Y.); (M.W.)
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China;
| | - Maria Kristina Parr
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; (F.Y.); (M.W.)
| |
Collapse
|
12
|
Ye W, Wang Z, Lv X, Yin H, Jiang L, Wang Z, Liu Y. Potential risk of drug-drug interactions of ponatinib via inhibition against human UDP-glucuronosyltransferases. Toxicol In Vitro 2023; 92:105664. [PMID: 37597759 DOI: 10.1016/j.tiv.2023.105664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/10/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Ponatinib is an efficient oral tyrosine kinase inhibitor (TKI) for T315I-positive Ph + ALL and T315I-positive chronic myeloid leukemia (CML) or BCR-ABL when no other TKIs can be prescribed. In this research, we evaluated the inhibitory effects of ponatinib on human recombinant UDP-glucuronosyltransferases (UGTs) and predicted the magnitude of potential drug-drug interaction (DDI) risk of co-treatment with ponatinib and UGTs substrates by using in vitro-in vivo extrapolation (IVIVE) method. Our study presented that ponatinib showed a broad-spectrum inhibition against UGTs. Particularly, ponatinib exhibited potent inhibitory effects towards UGT1A7, UGT1A1, and UGT1A9 with IC50 values of 0.37, 0.41, and 0.89 μM, respectively, which might lead to clinically significant DDI.
Collapse
Affiliation(s)
- Weiyi Ye
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Xin Lv
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Hang Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhe Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
13
|
Yang F, Sharma SS, Bureik M, Parr MK. Mutual Modulation of the Activities of Human CYP2D6 and Four UGTs during the Metabolism of Propranolol. Curr Issues Mol Biol 2023; 45:7130-7146. [PMID: 37754235 PMCID: PMC10527876 DOI: 10.3390/cimb45090451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/28/2023] Open
Abstract
Cytochromes P450 (CYP) and UDP-glucuronosyltransferases (UGT) are two enzyme families that play an important role in drug metabolism, catalyzing either the functionalization or glucuronidation of xenobiotics. However, their mutual interactions are poorly understood. In this study, the functional interactions of human CYP2D6 with four human UGTs (UGT1A7, UGT1A8, UGT1A9, and UGT2A1) were investigated using our previously established co-expression model system in the fission yeast Schizosaccharomyces pombe. The substrate employed was propranolol because it is well metabolized by CYP2D6. Moreover, the CYP2D6 metabolite 4-hydroxypropranolol is a known substrate for the four UGTs included in this study. Co-expression of either UGT1A7, UGT1A8, or UGT1A9 was found to increase the activity of CYP2D6 by a factor of 3.3, 2.1 or 2.8, respectively, for the conversion of propranolol to 4-hydroxypropranolol. In contrast, UGT2A1 co-expression did not change CYP2D6 activity. On the other hand, the activities of all four UGTs were completely suppressed by co-expression of CYP2D6. This data corroborates our previous report that CYP2D6 is involved in functional CYP-UGT interactions and suggest that such interactions can contribute to both adverse drug reactions and changes in drug efficacy.
Collapse
Affiliation(s)
- Fan Yang
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany;
| | - Sangeeta Shrestha Sharma
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (S.S.S.); (M.B.)
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (S.S.S.); (M.B.)
| | - Maria Kristina Parr
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany;
| |
Collapse
|
14
|
Miners JO, Polasek TM, Hulin JA, Rowland A, Meech R. Drug-drug interactions that alter the exposure of glucuronidated drugs: Scope, UDP-glucuronosyltransferase (UGT) enzyme selectivity, mechanisms (inhibition and induction), and clinical significance. Pharmacol Ther 2023:108459. [PMID: 37263383 DOI: 10.1016/j.pharmthera.2023.108459] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Drug-drug interactions (DDIs) arising from the perturbation of drug metabolising enzyme activities represent both a clinical problem and a potential economic loss for the pharmaceutical industry. DDIs involving glucuronidated drugs have historically attracted little attention and there is a perception that interactions are of minor clinical relevance. This review critically examines the scope and aetiology of DDIs that result in altered exposure of glucuronidated drugs. Interaction mechanisms, namely inhibition and induction of UDP-glucuronosyltransferase (UGT) enzymes and the potential interplay with drug transporters, are reviewed in detail, as is the clinical significance of known DDIs. Altered victim drug exposure arising from modulation of UGT enzyme activities is relatively common and, notably, the incidence and importance of UGT induction as a DDI mechanism is greater than generally believed. Numerous DDIs are clinically relevant, resulting in either loss of efficacy or an increased risk of adverse effects, necessitating dose individualisation. Several generalisations relating to the likelihood of DDIs can be drawn from the known substrate and inhibitor selectivities of UGT enzymes, highlighting the importance of comprehensive reaction phenotyping studies at an early stage of drug development. Further, rigorous assessment of the DDI liability of new chemical entities that undergo glucuronidation to a significant extent has been recommended recently by regulatory guidance. Although evidence-based approaches exist for the in vitro characterisation of UGT enzyme inhibition and induction, the availability of drugs considered appropriate for use as 'probe' substrates in clinical DDI studies is limited and this should be research priority.
Collapse
Affiliation(s)
- John O Miners
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Thomas M Polasek
- Certara, Princeton, NJ, USA; Centre for Medicines Use and Safety, Monash University, Melbourne, Australia
| | - Julie-Ann Hulin
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Andrew Rowland
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Robyn Meech
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
15
|
Kumar S, Sherman MY. Resistance to TOP-1 Inhibitors: Good Old Drugs Still Can Surprise Us. Int J Mol Sci 2023; 24:ijms24087233. [PMID: 37108395 PMCID: PMC10138578 DOI: 10.3390/ijms24087233] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Irinotecan (SN-38) is a potent and broad-spectrum anticancer drug that targets DNA topoisomerase I (Top1). It exerts its cytotoxic effects by binding to the Top1-DNA complex and preventing the re-ligation of the DNA strand, leading to the formation of lethal DNA breaks. Following the initial response to irinotecan, secondary resistance is acquired relatively rapidly, compromising its efficacy. There are several mechanisms contributing to the resistance, which affect the irinotecan metabolism or the target protein. In addition, we have demonstrated a major resistance mechanism associated with the elimination of hundreds of thousands of Top1 binding sites on DNA that can arise from the repair of prior Top1-dependent DNA cleavages. Here, we outline the major mechanisms of irinotecan resistance and highlight recent advancements in the field. We discuss the impact of resistance mechanisms on clinical outcomes and the potential strategies to overcome resistance to irinotecan. The elucidation of the underlying mechanisms of irinotecan resistance can provide valuable insights for the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Santosh Kumar
- Department of Molecular Biology, Ariel University, Ariel 40700, Israel
| | - Michael Y Sherman
- Department of Molecular Biology, Ariel University, Ariel 40700, Israel
| |
Collapse
|
16
|
Sharma SS, Sharma S, Zhao J, Bureik M. Mutual Influence of Human Cytochrome P450 Enzymes and UDP-Glucuronosyltransferases on Their Respective Activities in Recombinant Fission Yeast. Biomedicines 2023; 11:biomedicines11020281. [PMID: 36830817 PMCID: PMC9953201 DOI: 10.3390/biomedicines11020281] [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: 12/06/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023] Open
Abstract
Cytochromes P450 (CYPs) and UDP-glucuronosyltransferases (UGTs) are the most important human drug metabolizing enzymes, but their mutual interactions are poorly understood. In this study, we recombinantly co-expressed of each one of the 19 human members of the UGT families 1 and 2 with either CYP2C9, CYP2D6, or CYP4Z1 in fission yeast. Using these strains, we monitored a total of 72 interactions: 57 cases where we tested the influence of UGT co-expression on CYP activity and 15 cases of the opposite approach. In the majority of cases (88%), UGT co-expression had a statistically significant (p < 0.05) effect on P450 activity (58% positive and 30% negative). Strong changes were observed in nine cases, including one case with an activity increase by a factor of 23 (CYP2C9 activity in the presence of UGT2A3) but also four cases with a complete loss of activity. When monitoring the effect of CYP co-expression on the activity of five UGTs, activity changes were generally not so pronounced and, if observed, always detrimental. UGT2B7 activity was not influenced by CYP co-expression, while the other UGTs were affected to varying degrees. These data suggest the notion that mutual influence of CYPs and UGTs on each other's activity is a widespread phenomenon.
Collapse
|
17
|
Junk SV, Schaeffeler E, Zimmermann M, Möricke A, Beier R, Schütte P, Fedders B, Alten J, Hinze L, Klein N, Kulozik A, Muckenthaler MU, Koehler R, Borkhardt A, Vijayakrishnan J, Ellinghaus D, Forster M, Franke A, Wintering A, Kratz CP, Schrappe M, Schwab M, Houlston RS, Cario G, Stanulla M. Chemotherapy-related hyperbilirubinemia in pediatric acute lymphoblastic leukemia: a genome-wide association study from the AIEOP-BFM ALL study group. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:21. [PMID: 36639636 PMCID: PMC9838013 DOI: 10.1186/s13046-022-02585-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/26/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Characterization of clinical phenotypes in context with tumor and host genomic information can aid in the development of more effective and less toxic risk-adapted and targeted treatment strategies. To analyze the impact of therapy-related hyperbilirubinemia on treatment outcome and to identify contributing genetic risk factors of this well-recognized adverse effect we evaluated serum bilirubin levels in 1547 pediatric patients with acute lymphoblastic leukemia (ALL) and conducted a genome-wide association study (GWAS). PATIENTS AND METHODS Patients were treated in multicenter trial AIEOP-BFM ALL 2000 for pediatric ALL. Bilirubin toxicity was graded 0 to 4 according to the Common Toxicity Criteria (CTC) of the National Cancer Institute. In the GWAS discovery cohort, including 650 of the 1547 individuals, genotype frequencies of 745,895 single nucleotide variants were compared between 435 patients with hyperbilirubinemia (CTC grades 1-4) during induction/consolidation treatment and 215 patients without it (grade 0). Replication analyses included 224 patients from the same trial. RESULTS Compared to patients with no (grade 0) or moderate hyperbilirubinemia (grades 1-2) during induction/consolidation, patients with grades 3-4 had a poorer 5-year event free survival (76.6 ± 3% versus 87.7 ± 1% for grades 1-2, P = 0.003; 85.2 ± 2% for grade 0, P < 0.001) and a higher cumulative incidence of relapse (15.6 ± 3% versus 9.0 ± 1% for grades 1-2, P = 0.08; 11.1 ± 1% for grade 0, P = 0.007). GWAS identified a strong association of the rs6744284 variant T allele in the UGT1A gene cluster with risk of hyperbilirubinemia (allelic odds ratio (OR) = 2.1, P = 7 × 10- 8). TT-homozygotes had a 6.5-fold increased risk of hyperbilirubinemia (grades 1-4; 95% confidence interval (CI) = 2.9-14.6, P = 7 × 10- 6) and a 16.4-fold higher risk of grade 3-4 hyperbilirubinemia (95% CI 6.1-43.8, P = 2 × 10- 8). Replication analyses confirmed these associations with joint analysis yielding genome-wide significance (allelic OR = 2.1, P = 6 × 10- 11; 95% CI 1.7-2.7). Moreover, rs6744284 genotypes were strongly linked to the Gilbert's syndrome-associated UGT1A1*28/*37 allele (r2 = 0.70), providing functional support for study findings. Of clinical importance, the rs6744284 TT genotype counterbalanced the adverse prognostic impact of high hyperbilirubinemia on therapy outcome. CONCLUSIONS Chemotherapy-related hyperbilirubinemia is a prognostic factor for treatment outcome in pediatric ALL and genetic variation in UGT1A aids in predicting the clinical impact of hyperbilirubinemia. TRIAL REGISTRATION http://www. CLINICALTRIALS gov ; #NCT00430118.
Collapse
Affiliation(s)
- Stefanie V. Junk
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Elke Schaeffeler
- Margarete-Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Martin Zimmermann
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Anja Möricke
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Rita Beier
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Peter Schütte
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Birthe Fedders
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Julia Alten
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Laura Hinze
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Norman Klein
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Andreas Kulozik
- grid.7700.00000 0001 2190 4373Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Martina U. Muckenthaler
- grid.7700.00000 0001 2190 4373Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Rolf Koehler
- grid.7700.00000 0001 2190 4373Department of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Arndt Borkhardt
- grid.411327.20000 0001 2176 9917Clinic for Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jayaram Vijayakrishnan
- grid.18886.3fDivision of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, UK
| | - David Ellinghaus
- grid.9764.c0000 0001 2153 9986Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Michael Forster
- grid.9764.c0000 0001 2153 9986Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Andre Franke
- grid.9764.c0000 0001 2153 9986Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Astrid Wintering
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Christian P. Kratz
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Martin Schrappe
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Schwab
- Margarete-Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany ,grid.10392.390000 0001 2190 1447Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University of Tuebingen, Tuebingen, Germany ,grid.10392.390000 0001 2190 1447Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany ,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany
| | - Richard S. Houlston
- grid.18886.3fDivision of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Gunnar Cario
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Martin Stanulla
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
18
|
Tu MJ, Yu AM. Recent Advances in Novel Recombinant RNAs for Studying Post-transcriptional Gene Regulation in Drug Metabolism and Disposition. Curr Drug Metab 2023; 24:175-189. [PMID: 37170982 PMCID: PMC10825985 DOI: 10.2174/1389200224666230425232433] [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: 10/31/2022] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 05/13/2023]
Abstract
Drug-metabolizing enzymes and transporters are major determinants of the absorption, disposition, metabolism, and excretion (ADME) of drugs, and changes in ADME gene expression or function may alter the pharmacokinetics/ pharmacodynamics (PK/PD) and further influence drug safety and therapeutic outcomes. ADME gene functions are controlled by diverse factors, such as genetic polymorphism, transcriptional regulation, and coadministered medications. MicroRNAs (miRNAs) are a superfamily of regulatory small noncoding RNAs that are transcribed from the genome to regulate target gene expression at the post-transcriptional level. The roles of miRNAs in controlling ADME gene expression have been demonstrated, and such miRNAs may consequently influence cellular drug metabolism and disposition capacity. Several types of miRNA mimics and small interfering RNA (siRNA) reagents have been developed and widely used for ADME research. In this review article, we first provide a brief introduction to the mechanistic actions of miRNAs in post-transcriptional gene regulation of drug-metabolizing enzymes, transporters, and transcription factors. After summarizing conventional small RNA production methods, we highlight the latest advances in novel recombinant RNA technologies and applications of the resultant bioengineered RNA (BioRNA) agents to ADME studies. BioRNAs produced in living cells are not only powerful tools for general biological and biomedical research but also potential therapeutic agents amenable to clinical investigations.
Collapse
Affiliation(s)
- Mei-Juan Tu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
19
|
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
|
20
|
Vishnubalaji R, Shaath H, Al-Alwan M, Abdelalim EM, Alajez NM. Reciprocal interplays between MicroRNAs and pluripotency transcription factors in dictating stemness features in human cancers. Semin Cancer Biol 2022; 87:1-16. [PMID: 36354097 DOI: 10.1016/j.semcancer.2022.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
The interplay between microRNAs (miRNAs) and pluripotency transcription factors (TFs) orchestrates the acquisition of cancer stem cell (CSC) features during the course of malignant transformation, rendering them essential cancer cell dependencies and therapeutic vulnerabilities. In this review, we discuss emerging themes in tumor heterogeneity, including the clonal evolution and the CSC models and their implications in resistance to cancer therapies, and then provide thorough coverage on the roles played by key TFs in maintaining normal and malignant stem cell pluripotency and plasticity. In addition, we discuss the reciprocal interactions between miRNAs and MYC, OCT4, NANOG, SOX2, and KLF4 pluripotency TFs and their contributions to tumorigenesis. We provide our view on the potential to interfere with key miRNA-TF networks through the use of RNA-based therapeutics as single agents or in combination with other therapeutic strategies, to abrogate the CSC state and render tumor cells more responsive to standard and targeted therapies.
Collapse
Affiliation(s)
- Radhakrishnan Vishnubalaji
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Hibah Shaath
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Monther Al-Alwan
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Essam M Abdelalim
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Nehad M Alajez
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
| |
Collapse
|
21
|
Rausch M, Samodelov SL, Visentin M, Kullak-Ublick GA. The Farnesoid X Receptor as a Master Regulator of Hepatotoxicity. Int J Mol Sci 2022; 23:ijms232213967. [PMID: 36430444 PMCID: PMC9695947 DOI: 10.3390/ijms232213967] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The nuclear receptor farnesoid X receptor (FXR, NR1H4) is a bile acid (BA) sensor that links the enterohepatic circuit that regulates BA metabolism and elimination to systemic lipid homeostasis. Furthermore, FXR represents a real guardian of the hepatic function, preserving, in a multifactorial fashion, the integrity and function of hepatocytes from chronic and acute insults. This review summarizes how FXR modulates the expression of pathway-specific as well as polyspecific transporters and enzymes, thereby acting at the interface of BA, lipid and drug metabolism, and influencing the onset and progression of hepatotoxicity of varying etiopathogeneses. Furthermore, this review article provides an overview of the advances and the clinical development of FXR agonists in the treatment of liver diseases.
Collapse
|
22
|
SNPs in 3'UTR miRNA Target Sequences Associated with Individual Drug Susceptibility. Int J Mol Sci 2022; 23:ijms232213725. [PMID: 36430200 PMCID: PMC9692299 DOI: 10.3390/ijms232213725] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
The complementary interaction of microRNAs (miRNAs) with their binding sites in the 3'untranslated regions (3'UTRs) of target gene mRNAs represses translation, playing a leading role in gene expression control. MiRNA recognition elements (MREs) in the 3'UTRs of genes often contain single nucleotide polymorphisms (SNPs), which can change the binding affinity for target miRNAs leading to dysregulated gene expression. Accumulated data suggest that these SNPs can be associated with various human pathologies (cancer, diabetes, neuropsychiatric disorders, and cardiovascular diseases) by disturbing the interaction of miRNAs with their MREs located in mRNA 3'UTRs. Numerous data show the role of SNPs in 3'UTR MREs in individual drug susceptibility and drug resistance mechanisms. In this review, we brief the data on such SNPs focusing on the most rigorously proven cases. Some SNPs belong to conventional genes from the drug-metabolizing system (in particular, the genes coding for cytochromes P450 (CYP 450), phase II enzymes (SULT1A1 and UGT1A), and ABCB3 transporter and their expression regulators (PXR and GATA4)). Other examples of SNPs are related to the genes involved in DNA repair, RNA editing, and specific drug metabolisms. We discuss the gene-by-gene studies and genome-wide approaches utilized or potentially utilizable to detect the MRE SNPs associated with individual response to drugs.
Collapse
|
23
|
Kondo M, Ikenaka Y, Nakayama SMM, Kawai YK, Ishizuka M. Duplication, Loss, and Evolutionary Features of Specific UDP-Glucuronosyltransferase Genes in Carnivora (Mammalia, Laurasiatheria). Animals (Basel) 2022; 12:2954. [PMID: 36359081 PMCID: PMC9658400 DOI: 10.3390/ani12212954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 12/26/2024] Open
Abstract
UDP-glucuronosyltransferases (UGTs) are one of the most important enzymes for xenobiotic metabolism or detoxification. Through duplication and loss of genes, mammals evolved the species-specific variety of UGT isoforms. Among mammals, Carnivora is one of the orders that includes various carnivorous species, yet there is huge variation of food habitat. Recently, lower activity of UGT1A and 2B were shown in Felidae and pinnipeds, suggesting evolutional loss of these isoforms. However, comprehensive analysis for genetic or evolutional features are still missing. This study was conducted to reveal evolutional history of UGTs in Carnivoran species. We found specific gene expansion of UGT1As in Canidae, brown bear and black bear. We also found similar genetic duplication in UGT2Bs in Canidae, and some Mustelidae and Ursidae. In addition, we discovered contraction or complete loss of UGT1A7-12 in phocids, some otariids, felids, and some Mustelids. These studies indicate that even closely related species have completely different evolution of UGTs and further imply the difficulty of extrapolation of the pharmacokinetics and toxicokinetic result of experimental animals into wildlife carnivorans.
Collapse
Affiliation(s)
- Mitsuki Kondo
- Laboratory of Toxicology, Department of Environmental Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan
| | - Yoshinori Ikenaka
- Laboratory of Toxicology, Department of Environmental Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan
- Water Research Group, Unit for Environmental Sciences and Management, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa
- Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- One Health Research Center, Hokkaido University, Sapporo 060-0818, Japan
| | - Shouta M. M. Nakayama
- Laboratory of Toxicology, Department of Environmental Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan
- Biomedical Sciences Department, School of Veterinary Medicine, The University of Zambia, P.O. Box 32379, Lusaka 10101, Zambia
| | - Yusuke K. Kawai
- Laboratory of Toxicology, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Japan
| | - Mayumi Ishizuka
- Laboratory of Toxicology, Department of Environmental Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan
| |
Collapse
|
24
|
Audet-Delage Y, Rouleau M, Villeneuve L, Guillemette C. The Glycosyltransferase Pathway: An Integrated Analysis of the Cell Metabolome. Metabolites 2022; 12:metabo12101006. [PMID: 36295907 PMCID: PMC9609030 DOI: 10.3390/metabo12101006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022] Open
Abstract
Nucleotide sugar-dependent glycosyltransferases (UGTs) are critical to the homeostasis of endogenous metabolites and the detoxification of xenobiotics. Their impact on the cell metabolome remains unknown. Cellular metabolic changes resulting from human UGT expression were profiled by untargeted metabolomics. The abundant UGT1A1 and UGT2B7 were studied as UGT prototypes along with their alternative (alt.) splicing-derived isoforms displaying structural differences. Nineteen biochemical routes were modified, beyond known UGT substrates. Significant variations in glycolysis and pyrimidine pathways, and precursors of the co-substrate UDP-glucuronic acid were observed. Bioactive lipids such as arachidonic acid and endocannabinoids were highly enriched by up to 13.3-fold (p < 0.01) in cells expressing the canonical enzymes. Alt. UGT2B7 induced drastic and unique metabolic perturbations, including higher glucose (18-fold) levels and tricarboxylic acid cycle (TCA) cycle metabolites and abrogated the effects of the UGT2B7 canonical enzyme when co-expressed. UGT1A1 proteins promoted the accumulation of branched-chain amino acids (BCAA) and TCA metabolites upstream of the mitochondrial oxoglutarate dehydrogenase complex (OGDC). Alt. UGT1A1 exacerbated these changes, likely through its interaction with the OGDC component oxoglutarate dehydrogenase-like (OGDHL). This study expands the breadth of biochemical pathways associated with UGT expression and establishes extensive connectivity between UGT enzymes, alt. proteins and other metabolic processes.
Collapse
Affiliation(s)
- Yannick Audet-Delage
- Centre Hospitalier Universitaire de Québec Research Center—Université Laval, Faculty of Pharmacy, and Université Laval Cancer Research Center (CRC), R4720, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
| | - Michèle Rouleau
- Centre Hospitalier Universitaire de Québec Research Center—Université Laval, Faculty of Pharmacy, and Université Laval Cancer Research Center (CRC), R4720, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
| | - Lyne Villeneuve
- Centre Hospitalier Universitaire de Québec Research Center—Université Laval, Faculty of Pharmacy, and Université Laval Cancer Research Center (CRC), R4720, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
| | - Chantal Guillemette
- Centre Hospitalier Universitaire de Québec Research Center—Université Laval, Faculty of Pharmacy, and Université Laval Cancer Research Center (CRC), R4720, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
- Canada Research Chair in Pharmacogenomics, Université Laval, Québec, QC G1V 4G2, Canada
- Correspondence: ; Tel.: +1-(418)-654-2296
| |
Collapse
|
25
|
Tesolin P, Bertinetto FE, Sonaglia A, Cappellani S, Concas MP, Morgan A, Ferrero NM, Zabotti A, Gasparini P, Amoroso A, Quartuccio L, Girotto G. High Throughput Genetic Characterisation of Caucasian Patients Affected by Multi-Drug Resistant Rheumatoid or Psoriatic Arthritis. J Pers Med 2022; 12:jpm12101618. [PMID: 36294757 PMCID: PMC9605087 DOI: 10.3390/jpm12101618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/14/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022] Open
Abstract
Rheumatoid and psoriatic arthritis (RA and PsA) are inflammatory rheumatic disorders characterised by a multifactorial etiology. To date, the genetic contributions to the disease onset, severity and drug response are not clearly defined, and despite the development of novel targeted therapies, ~10% of patients still display poor treatment responses. We characterised a selected cohort of eleven non-responder patients aiming to define the genetic contribution to drug resistance. An accurate clinical examination of the patients was coupled with several high-throughput genetic testing, including HLA typing, SNPs-array and Whole Exome Sequencing (WES). The analyses revealed that all the subjects carry very rare HLA phenotypes which contain HLA alleles associated with RA development (e.g., HLA-DRB1*04, DRB1*10:01 and DRB1*01). Additionally, six patients also carry PsA risk alleles (e.g., HLA-B*27:02 and B*38:01). WES analysis and SNPs-array revealed 23 damaging variants with 18 novel “drug-resistance” RA/PsA candidate genes. Eight patients carry likely pathogenic variants within common genes (CYP21A2, DVL1, PRKDC, ORAI1, UGT2B17, MSR1). Furthermore, “private” damaging variants were identified within 12 additional genes (WNT10A, ABCB7, SERPING1, GNRHR, NCAPD3, CLCF1, HACE1, NCAPD2, ESR1, SAMHD1, CYP27A1, CCDC88C). This multistep approach highlighted novel RA/PsA candidate genes and genotype-phenotype correlations potentially useful for clinicians in selecting the best therapeutic strategy.
Collapse
Affiliation(s)
- Paola Tesolin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Francesca Eleonora Bertinetto
- Department of Medical Sciences, University of Turin, and Immunogenetic and Transplant Biology Service, University Hospital “Città della Salute e della Scienza”, 10124 Turin, Italy
| | - Arianna Sonaglia
- Division of Rheumatology, Department of Medicine (DAME), ASUFC, University of Udine, 33100 Udine, Italy
| | - Stefania Cappellani
- Institute for Maternal and Child Health—IRCCS, Burlo Garofolo, 34137 Trieste, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health—IRCCS, Burlo Garofolo, 34137 Trieste, Italy
- Correspondence: ; Tel.: +39-0403785539
| | - Anna Morgan
- Institute for Maternal and Child Health—IRCCS, Burlo Garofolo, 34137 Trieste, Italy
| | - Norma Maria Ferrero
- Department of Medical Sciences, University of Turin, and Immunogenetic and Transplant Biology Service, University Hospital “Città della Salute e della Scienza”, 10124 Turin, Italy
| | - Alen Zabotti
- Division of Rheumatology, Department of Medicine (DAME), ASUFC, University of Udine, 33100 Udine, Italy
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
- Institute for Maternal and Child Health—IRCCS, Burlo Garofolo, 34137 Trieste, Italy
| | - Antonio Amoroso
- Department of Medical Sciences, University of Turin, and Immunogenetic and Transplant Biology Service, University Hospital “Città della Salute e della Scienza”, 10124 Turin, Italy
| | - Luca Quartuccio
- Division of Rheumatology, Department of Medicine (DAME), ASUFC, University of Udine, 33100 Udine, Italy
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
- Institute for Maternal and Child Health—IRCCS, Burlo Garofolo, 34137 Trieste, Italy
| |
Collapse
|
26
|
Wang Z, Jiang L, Wang X, Yin H, Wang Z, Lv X, Liu Y. Cabozantinib Carries the Risk of Drug-Drug Interactions via Inhibition of UDPglucuronosyltransferase (UGT) 1A9. Curr Drug Metab 2022; 23:912-919. [PMID: 36306450 DOI: 10.2174/1389200224666221028140652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/12/2022] [Accepted: 09/28/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Cabozantinib is a multiple receptor tyrosine kinases inhibitor (TKI) approved to treat progressive, metastatic medullary thyroid cancer, advanced renal cell carcinoma, and hepatocellular carcinoma. Drugdrug interactions (DDIs) for cabozantinib have been identified involving the role of cytochromes P450. Although the previous study reported that cabozantinib showed a slight inhibition of UDP-glucuronosyltransferase (UGT) 1A1 at the highest concentration tested, there are no reports on the potential for UGTs-mediated-DDIs. Hence, the current study aims to address this knowledge gap. OBJECTIVE This study aimed to investigate the inhibitory effect of cabozantinib on human UGTs and to quantitatively evaluate the DDI potential via UGT inhibition. METHODS The inhibitory effects of cabozantinib on UGTs were determined by measuring the formation rates for 4- methylumbelliferone (4-MU) glucuronide and trifluoperazine N-glucuronide using recombinant human UGT isoforms in the absence or presence of cabozantinib. Inhibition kinetic studies were conducted to determine the type of inhibition of cabozantinib on UGTs and the corresponding inhibition constant (Ki) value. In vitro-in vivo extrapolation (IVIVE) was further employed to predict the potential risk of DDI in vivo. RESULTS Cabozantinib displayed potent inhibition of UGT1A1, 1A3, 1A4, 1A6, 1A7, 1A8, 1A9, 1A10, 2B7, and 2B15. Cabozantinib exhibited noncompetitive inhibition towards UGT1A1 and 1A3 and inhibition towards UGT1A7 and 1A9. The Ki,u values (mean ± standard deviation) were calculated to be 2.15±0.11 μM, 0.83±0.05 μM, 0.75±0.04 μM and 0.18 ± 0.10 μM for UGT1A1, 1A3, 1A7 and 1A9, respectively. Co-administration of cabozantinib at the clinically approved dose of 60 mg/day or 140 mg/day may result in approximately a 26% to 60% increase in the systemic exposure of drugs predominantly cleared by UGT1A9, implying a high risk of DDIs. CONCLUSION Cabozantinib has the potential to cause DDIs via the inhibition of UGT1A9; therefore, additional attention should be paid to the safety of the combined use of cabozantinib and drugs metabolized by UGT1A9.
Collapse
Affiliation(s)
- Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Xiaoyu Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Hang Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Xin Lv
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| |
Collapse
|
27
|
Computational prediction for the metabolism of human UDP-glucuronosyltransferase 1A1 substrates. Comput Biol Med 2022; 149:105959. [DOI: 10.1016/j.compbiomed.2022.105959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/31/2022] [Accepted: 08/06/2022] [Indexed: 11/15/2022]
|
28
|
Yang F, Liu S, Wolber G, Bureik M, Parr MK. Complete Reaction Phenotyping of Propranolol and 4-Hydroxypropranolol with the 19 Enzymes of the Human UGT1 and UGT2 Families. Int J Mol Sci 2022; 23:ijms23137476. [PMID: 35806479 PMCID: PMC9267274 DOI: 10.3390/ijms23137476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 02/01/2023] Open
Abstract
Propranolol is a competitive non-selective beta-receptor antagonist that is available on the market as a racemic mixture. In the present study, glucuronidation of propranolol and its equipotent phase I metabolite 4-hydroxypropranolol by all 19 members of the human UGT1 and UGT2 families was monitored. UGT1A7, UGT1A9, UGT1A10 and UGT2A1 were found to glucuronidate propranolol, with UGT1A7, UGT1A9 and UGT2A1 mainly acting on (S)-propranolol, while UGT1A10 displays the opposite stereoselectivity. UGT1A7, UGT1A9 and UGT2A1 were also found to glucuronidate 4-hydroxypropranolol. In contrast to propranolol, 4-hydroxypropranolol was found to be glucuronidated by UGT1A8 but not by UGT1A10. Additional biotransformations with 4-methoxypropanolol demonstrated different regioselectivities of these UGTs with respect to the aliphatic and aromatic hydroxy groups of the substrate. Modeling and molecular docking studies were performed to explain the stereoselective glucuronidation of the substrates under study.
Collapse
Affiliation(s)
- Fan Yang
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany;
| | - Sijie Liu
- Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; (S.L.); (G.W.)
| | - Gerhard Wolber
- Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; (S.L.); (G.W.)
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China;
| | - Maria Kristina Parr
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany;
- Correspondence:
| |
Collapse
|
29
|
Atasilp C, Biswas M, Jinda P, Nuntharadthanaphong N, Rachanakul J, Hongkaew Y, Vanwong N, Saokaew S, Sukasem C. Association of UGT1A1*6, UGT1A1*28, or ABCC2 c.3972C>T genetic polymorphisms with irinotecan-induced toxicity in Asian cancer patients: Meta-analysis. Clin Transl Sci 2022; 15:1613-1633. [PMID: 35506159 PMCID: PMC9283744 DOI: 10.1111/cts.13277] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 11/30/2022] Open
Abstract
Effects of UGT1A1*6 and UGT1A1*28 genetic polymorphisms on irinotecan-induced severe toxicities in Asian cancer patients are inconclusive. Also, ABCC2 c.3972C>T may affect toxicity of irinotecan. The aim was to assess the aggregated risk of neutropenia or diarrhea in Asian cancer patients taking irinotecan and inherited UGT1A1*6, UGT1A1*28, or ABCC2 c.3972C>T genetic variants. A PubMed literature search for eligible studies was conducted. Odds ratios (ORs) were measured using RevMan software where p values <0.05 were statistically significant. Patients that inherited both UGT1A1*6 and UGT1A1*28 genetic variants (heterozygous: UGT1A1*1/*6 + *1/*28 and homozygous: UGT1A1*6/*6 + *28/*28) were significantly associated with increased risk of neutropenia and diarrhea compared to patients with UGT1A1*1/*1 (neutropenia: OR 2.89; 95% CI 1.97-4.23; p < 0.00001; diarrhea: OR 2.26; 95% CI 1.71-2.99; p < 0.00001). Patients carrying homozygous variants had much stronger effects in developing toxicities (neutropenia: OR 6.23; 95% CI 3.11-12.47; p < 0.00001; diarrhea: OR 3.21; 95% CI 2.13-4.85; p < 0.00001) than those with heterozygous variants. However, patients carrying the ABCC2 c.3972C>T genetic variant were not significantly associated with neutropenia (OR 1.67; 95% CI 0.98-2.84; p = 0.06) and were significantly associated with a reduction in irinotecan-induced diarrhea (OR 0.31; 95% CI 0.11-0.81; p = 0.02). Asian cancer patients should undergo screening for both UGT1A1*6 and UGT1A1*28 genetic variants to reduce substantially irinotecan-induced severe toxicities.
Collapse
Affiliation(s)
- Chalirmporn Atasilp
- Chulabhorn International College of MedicineThammasat UniversityPathum ThaniThailand
| | - Mohitosh Biswas
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
- Department of PharmacyUniversity of RajshahiRajshahiBangladesh
| | - Pimonpan Jinda
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
| | - Nutthan Nuntharadthanaphong
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
| | - Jiratha Rachanakul
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
| | - Yaowaluck Hongkaew
- Advance Research and Development LaboratoryBumrungrad International HospitalBangkokThailand
| | - Natchaya Vanwong
- Department of Clinical Chemistry, Faculty of Allied Health SciencesChulalongkorn UniversityBangkokThailand
| | - Surasak Saokaew
- Division of Pharmacy Practice, Department of Pharmaceutical CareSchool of Pharmaceutical Sciences, University of PhayaoPhayaoThailand
- Center of Health Outcomes Research and Therapeutic Safety (COHORTS)School of Pharmaceutical Sciences, University of PhayaoPhayaoThailand
- Unit of Excellence on Clinical Outcomes Research and Integration (UNICORN)School of Pharmaceutical Sciences, University of PhayaoPhayaoThailand
| | - Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
- Pharmacogenomics and Precision Medicine, The Preventive Genomics & Family Check‐up Services CenterBumrungrad International HospitalBangkokThailand
| |
Collapse
|
30
|
Caron P, Van Long FN, Rouleau M, Bujold E, Fortin P, Mohammadi S, Lévesque É, Breton S, Guillemette C. A liquid chromatography-mass spectrometry assay for the quantification of nucleotide sugars in human plasma and urine specimens and its clinical application. J Chromatogr A 2022; 1677:463296. [DOI: 10.1016/j.chroma.2022.463296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/16/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
|
31
|
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: 7] [Impact Index Per Article: 2.3] [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
|
32
|
Järvinen E, Deng F, Kiander W, Sinokki A, Kidron H, Sjöstedt N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front Pharmacol 2022; 12:802539. [PMID: 35095509 PMCID: PMC8793843 DOI: 10.3389/fphar.2021.802539] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Glucuronidation and sulfation are the most typical phase II metabolic reactions of drugs. The resulting glucuronide and sulfate conjugates are generally considered inactive and safe. They may, however, be the most prominent drug-related material in the circulation and excreta of humans. The glucuronide and sulfate metabolites of drugs typically have limited cell membrane permeability and subsequently, their distribution and excretion from the human body requires transport proteins. Uptake transporters, such as organic anion transporters (OATs and OATPs), mediate the uptake of conjugates into the liver and kidney, while efflux transporters, such as multidrug resistance proteins (MRPs) and breast cancer resistance protein (BCRP), mediate expulsion of conjugates into bile, urine and the intestinal lumen. Understanding the active transport of conjugated drug metabolites is important for predicting the fate of a drug in the body and its safety and efficacy. The aim of this review is to compile the understanding of transporter-mediated disposition of phase II conjugates. We review the literature on hepatic, intestinal and renal uptake transporters participating in the transport of glucuronide and sulfate metabolites of drugs, other xenobiotics and endobiotics. In addition, we provide an update on the involvement of efflux transporters in the disposition of glucuronide and sulfate metabolites. Finally, we discuss the interplay between uptake and efflux transport in the intestine, liver and kidneys as well as the role of transporters in glucuronide and sulfate conjugate toxicity, drug interactions, pharmacogenetics and species differences.
Collapse
Affiliation(s)
- Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alli Sinokki
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
33
|
Palabiyik-Yucelik SS, Moser S, Becker K, Halici Z, Bayir Y, Stonig M, Schennach H, Fuchs D, Gostner JM, Kurz K. Oxyresveratrol modulates the immune response in vitro. Pteridines 2021. [DOI: 10.1515/pteridines-2020-0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
The naturally occurring stilbenoid oxyresveratrol was shown to influence inflammatory and metabolic processes. During cellular immune activation, tryptophan breakdown and neopterin formation via the enzymes indoleamine 2,3-dioxygenase-1 (IDO-1) and GTP-cyclohydrolase, respectively, are induced. Neopterin and the kynurenine to tryptophan ratio are reliable and pertinent biomarkers of Th1-type immune response and are also used in vitro to monitor effects of active plant ingredients on peripheral blood mononuclear cells (PBMCs). We investigated the effects of oxyresveratrol on the activity of the above-mentioned pathways in mitogen-stimulated human PBMC and in the myelomonocytic cell line THP-1. Oxyresveratrol exerted suppressive effects on tryptophan breakdown in both stimulated cell models. Of note, in PBMC, tryptophan breakdown was induced at lower concentrations (5–20 µM) and suppressed at higher treatment concentrations only. Neopterin formation was decreased dose-dependently in stimulated PBMC. In unstimulated PBMC similar, albeit lesser effects were observed. Data indicate that oxyresveratrol exerts distinct and concentration-dependent effects on different immune cell types. IDO-1 is targeted by oxyresveratrol and its activity can be modulated in both directions. Detailed investigations of the interactions would be interesting to fully explore the activity of this phytocompound.
Collapse
Affiliation(s)
- Saziye Sezin Palabiyik-Yucelik
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Atatürk University , Erzurum , Turkey
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck , Innsbruck , Austria
- Clinical Research, Development and Design Application and Research Center, Atatürk University , Erzurum , Turkey
| | - Simone Moser
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians University of Munich , Munich , Germany
| | - Kathrin Becker
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck , Innsbruck , Austria
| | - Zekai Halici
- Clinical Research, Development and Design Application and Research Center, Atatürk University , Erzurum , Turkey
- Department of Pharmacology, Faculty of Medicine, Atatürk University , Erzurum , Turkey
| | - Yasin Bayir
- Department of Biochemistry, Faculty of Pharmacy, Atatürk University , Erzurum , Turkey
| | - Marlies Stonig
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck , Innsbruck , Austria
| | - Harald Schennach
- Central Institute of Blood Transfusion and Immunology, University Hospital of Innsbruck , Innsbruck , Austria
| | - Dietmar Fuchs
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck , Innsbruck , Austria
| | - Johanna M. Gostner
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck , Innsbruck , Austria
| | - Katharina Kurz
- Department of Internal Medicine II, Infectious Diseases, Pneumology, Rheumatology, Medical University of Innsbruck , Anichstrasse 35 , 6020 Innsbruck , Austria
| |
Collapse
|
34
|
Marshall JC, Liang Y, Sahasrabudhe V, Tensfeldt T, Fediuk DJ, Zhou S, Krishna R, Dawra VK, Wood LS, Sweeney K. Meta-Analysis of Noncompartmental Pharmacokinetic Parameters of Ertugliflozin to Evaluate Dose Proportionality and UGT1A9 Polymorphism Effect on Exposure. J Clin Pharmacol 2021; 61:1220-1231. [PMID: 33813736 PMCID: PMC8453771 DOI: 10.1002/jcph.1866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/29/2021] [Indexed: 12/16/2022]
Abstract
Ertugliflozin, a sodium‐glucose cotransporter 2 inhibitor, is primarily metabolized via glucuronidation by the uridine 5′‐diphospho‐glucuronosyltransferase (UGT) isoform UGT1A9. This noncompartmental meta‐analysis of ertugliflozin pharmacokinetics evaluated the relationship between ertugliflozin exposure and dose, and the effect of UGT1A9 genotype on ertugliflozin exposure. Pharmacokinetic data from 25 phase 1 studies were pooled. Structural models for dose proportionality described the relationship between ertugliflozin area under the plasma concentration‐time curve (AUC) or maximum observed plasma concentration (Cmax) and dose. A structural model for the UGT1A9 genotype described the relationship between ertugliflozin AUC and dose, with genotype information on 3 UGT1A9 polymorphisms (UGT1A9‐2152, UGT1A9*3, UGT1A9*1b) evaluated as covariates from the full model. Ertugliflozin AUC and Cmax increased in a dose‐proportional manner over the dose range of 0.5‐300 mg, and population‐predicted AUC and Cmax values for the 5‐ and 15‐mg ertugliflozin tablets administered in the fasted state demonstrated good agreement with the observed data. The largest change in ertugliflozin AUC was in subjects carrying the UGT1A9*3 heterozygous variant, with population‐predicted AUC (90% confidence interval) values of 485 ng·h/mL (458 to 510 ng·h/mL) and 1560 ng·h/mL (1480 to 1630 ng·h/mL) for ertugliflozin 5 and 15 mg, respectively, compared with 436 ng·h/mL (418 to 455 ng·h/mL) and 1410 ng·h/mL (1350 to 1480 ng·h/mL), respectively, in wild‐type subjects. Overall, the mean effects of the selected UGT1A9 variants on ertugliflozin AUC were within ±10% of the wild type. UGT1A9 genotype did not have any clinically meaningful effects on ertugliflozin exposure in healthy subjects. No ertugliflozin dose adjustment would be required in patients with the UGT1A9 variants assessed in this study.
Collapse
Affiliation(s)
| | | | | | | | | | - Susan Zhou
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | | | | | | |
Collapse
|
35
|
Morelli C, Formica V, Riondino S, Russo A, Ferroni P, Guadagni F, Roselli M. Irinotecan or Oxaliplatin: Which is the First Move for the Mate? Curr Med Chem 2021; 28:3158-3172. [PMID: 33069191 DOI: 10.2174/0929867327666201016124950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/02/2020] [Accepted: 09/06/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVES The aim of the present review is to discuss the potential link between RAS, BRAF and microsatellite instability (MSI) mutational patterns and chemotherapeutic agent efficacy [Irinotecan (IRI) vs. Oxaliplatin (OXA)], and how this can potentially influence the choice of the chemotherapy backbone. METHODS Following a review of the research literature, all pertinent articles published in the core journals were selected for the study. The inclusion criteria regarded relevant clinical and pre-clinical studies on the topic of interest (Relationship of OXA and IRI to KRAS/BRAF mutations and MSI). RESULTS Excision repair cross complementation group 1 (ERCC1) expression is inhibited by KRAS mutation, making tumor cells more sensitive to OXA. Results from OPUS, COIN and PRIME trials support that no conclusive data are available for BRAF mutant population because of the small number of patients. Enhanced IRI cytotoxicity to MSI cell lines is due to the participation of some of the mismatch repair (MMR) components in various DNA repair processes and their role in the maintenance of the pro-apoptotic effect of IRI and G2/M cell arrest. CONCLUSION OXA and IRI are indispensable drugs for mCRC treatment and their selection must be as careful as that of targeted agents. We suggest taking into consideration the interaction between known genomic alterations and OXA and IRI activity to personalize chemotherapy in mCRC patients.
Collapse
Affiliation(s)
- Cristina Morelli
- Department of Systems Medicine, Medical Oncology Unit, Tor Vergata Clinical Center, Tor Vergata University of Rome, Viale Oxford 81, 00133, Rome, Italy
| | - Vincenzo Formica
- Department of Systems Medicine, Medical Oncology Unit, Tor Vergata Clinical Center, Tor Vergata University of Rome, Viale Oxford 81, 00133, Rome, Italy
| | - Silvia Riondino
- Department of Systems Medicine, Medical Oncology Unit, Tor Vergata Clinical Center, Tor Vergata University of Rome, Viale Oxford 81, 00133, Rome, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Patrizia Ferroni
- BioBIM (InterInstitutional Multidisciplinary Biobank), IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Fiorella Guadagni
- BioBIM (InterInstitutional Multidisciplinary Biobank), IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Mario Roselli
- Department of Systems Medicine, Medical Oncology Unit, Tor Vergata Clinical Center, Tor Vergata University of Rome, Viale Oxford 81, 00133, Rome, Italy
| |
Collapse
|
36
|
|
37
|
Meng CL, Zhao W, Zhong DN. Epigenetics and microRNAs in UGT1As. Hum Genomics 2021; 15:30. [PMID: 34034810 PMCID: PMC8147421 DOI: 10.1186/s40246-021-00331-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/10/2021] [Indexed: 11/10/2022] Open
Abstract
UDP-glucuronosyltransferases (UGTs) are the main phase II drug-metabolizing enzymes mediating the most extensive glucuronidation-binding reaction in the human body. The UGT1A family is involved in more than half of glucuronidation reactions. However, significant differences exist in the distribution of UGT1As in vivo and the expression of UGT1As among individuals, and these differences are related to the occurrence of disease and differences in metabolism. In addition to genetic polymorphisms, there is now interest in the contribution of epigenetics and noncoding RNAs (especially miRNAs) to this differential change. Epigenetics regulates UGT1As pretranscriptionally through DNA methylation and histone modification, and miRNAs are considered the key mechanism of posttranscriptional regulation of UGT1As. Both epigenetic inheritance and miRNAs are involved in the differences in sex expression and in vivo distribution of UGT1As. Moreover, epigenetic changes early in life have been shown to affect gene expression throughout life. Here, we review and summarize the current regulatory role of epigenetics in the UGT1A family and discuss the relationship among epigenetics and UGT1A-related diseases and treatment, with references for future research.
Collapse
Affiliation(s)
- Cui-Lan Meng
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning City, Guangxi, China
| | - Wei Zhao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning City, Guangxi, China
| | - Dan-Ni Zhong
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning City, Guangxi, China.
| |
Collapse
|
38
|
Wang Z, Wang X, Wang Z, Jia Y, Feng Y, Jiang L, Xia Y, Cao J, Liu Y. In vitro inhibition of human UDP-glucuronosyltransferase (UGT) 1A1 by osimertinib, and prediction of in vivo drug-drug interactions. Toxicol Lett 2021; 348:10-17. [PMID: 34044055 DOI: 10.1016/j.toxlet.2021.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 11/25/2022]
Abstract
Osimertinib is the only third-generation epidermal growth factor receptor tyrosine-kinase inhibitor (EGFR-TKI) approved by Food and Drug Administration (FDA). This study aimed to know the inhibitory effect of osimertinib on human UDP-glucosyltransferases (UGTs) and human liver microsomes (HLMs), as well as to identify its potential to cause drug-drug interaction (DDI) arising from the modulation of UGT activity. High inhibitory effect of osimertinib was shown towards UGT1A1, 1A3, 1A6, 1A7, 1A8, 1A10, 2B7 and 2B15. Especially, osimertinib exhibited competitive inhibition against UGT1A1 with a Ki,u of 0.87 ± 0.12 μM. It also noncompetitively inhibited SN-38 glucuronidation in pooled HLMs with a Ki,u of 3.32 ± 0.25 μM. Results from quantitative prediction study indicated that osimertinib administered at 80 mg/day may result in a 4.83 % increase in the AUC of drugs mainly metabolized by UGT1A1, implying low risk of DDI via liver metabolism. However, the ratios of [I]gut/Ki,u are much higher than 11 in HLMs and recombinant UGT1A1, indicating a risk for interaction in intestine. The effects of osimertinib on intestinal UGT should be paid more attention on to avoid unnecessary clinical DDI risks.
Collapse
Affiliation(s)
- Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Xiaoyu Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yaqin Jia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yuyi Feng
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yangliu Xia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Jun Cao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, 116044, China.
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China.
| |
Collapse
|
39
|
Nardone-White DT, Bissada JE, Abouda AA, Jackson KD. Detoxication versus Bioactivation Pathways of Lapatinib In Vitro: UGT1A1 Catalyzes the Hepatic Glucuronidation of Debenzylated Lapatinib. Drug Metab Dispos 2021; 49:233-244. [PMID: 33376146 PMCID: PMC7883098 DOI: 10.1124/dmd.120.000236] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/20/2020] [Indexed: 12/13/2022] Open
Abstract
O-Dealkylation of the tyrosine kinase inhibitor lapatinib by cytochrome P450 3A enzymes is implicated in the development of lapatinib-induced hepatotoxicity. Conjugative metabolism of debenzylated lapatinib (M1) via glucuronidation and sulfation is thought to be a major detoxication pathway for lapatinib in preclinical species (rat and dog), limiting formation of the quinoneimine reactive metabolite. Glucuronidation of M1 by human recombinant UDP-glucuronosyltransferases (UGTs) has been reported in vitro; however, the relative UGT enzyme contributions are unknown, and the interspecies differences in the conjugation versus bioactivation pathways of M1 have not been fully elucidated. In the present study, reaction phenotyping experiments using human recombinant UGT enzymes and enzyme-selective chemical inhibitors demonstrated that UGT1A1 was the major hepatic UGT enzyme involved in lapatinib M1 glucuronidation. Formation of the M1-glucuronide by human liver microsomes from UGT1A1-genotyped donors was significantly correlated with UGT1A1 activity as measured by 17β-estradiol 3-glucuronidation (R 2 = 0.90). Interspecies differences were found in the biotransformation of M1 in human, rat, and dog liver microsomal and 9000g supernatant (S9) fractions via glucuronidation, sulfation, aldehyde oxidase-mediated oxidation, and bioactivation to the quinoneimine trapped as a glutathione (GSH) conjugate. Moreover, we demonstrated the sequential metabolism of lapatinib in primary human hepatocytes to the M1-glucuronide, M1-sulfate, and quinoneimine-GSH conjugate. M1 glucuronidation was highly correlated with the rates of M1 formation, suggesting that O-dealkylation may be the rate-limiting step in lapatinib biotransformation. Interindividual variability in the formation and clearance pathways of lapatinib M1 likely influences the hepatic exposure to reactive metabolites and may affect the risk for hepatotoxicity. SIGNIFICANCE STATEMENT: We used an integrated approach to examine the interindividual and interspecies differences in detoxication versus bioactivation pathways of lapatinib, which is associated with idiosyncratic hepatotoxicity. In addition to cytochrome P450 (P450)-mediated bioactivation, we report that multiple non-P450 pathways are involved in the biotransformation of the primary phenolic metabolite of lapatinib in vitro, including glucuronidation, sulfation, and aldehyde oxidase mediated oxidation. UGT1A1 was identified as the major hepatic enzyme involved in debenzylated lapatinib glucuronidation, which may limit hepatic exposure to the potentially toxic quinoneimine.
Collapse
Affiliation(s)
- Dasean T Nardone-White
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (D.T.N.-W., K.D.J.) and Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee (J.E.B., A.A.A.)
| | - Jennifer E Bissada
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (D.T.N.-W., K.D.J.) and Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee (J.E.B., A.A.A.)
| | - Arsany A Abouda
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (D.T.N.-W., K.D.J.) and Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee (J.E.B., A.A.A.)
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (D.T.N.-W., K.D.J.) and Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee (J.E.B., A.A.A.)
| |
Collapse
|
40
|
Zhu YD, Guan XQ, Chen J, Peng S, Finel M, Zhao YY, Wang RM, Bi HC, Lei M, Wang DD, Ge GB. Neobavaisoflavone Induces Bilirubin Metabolizing Enzyme UGT1A1 via PPARα and PPARγ. Front Pharmacol 2021; 11:628314. [PMID: 33628187 PMCID: PMC7897654 DOI: 10.3389/fphar.2020.628314] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022] Open
Abstract
UDP-glucuronosyltransferase 1A1 (UGT1A1) is an essential enzyme in mammals that is responsible for detoxification and metabolic clearance of the endogenous toxin bilirubin and a variety of xenobiotics, including some crucial therapeutic drugs. Discovery of potent and safe UGT1A1 inducers will provide an alternative therapy for ameliorating hyperbilirubinaemia and drug-induced hepatoxicity. This study aims to find efficacious UGT1A1 inducer(s) from natural flavonoids, and to reveal the mechanism involved in up-regulating of this key conjugative enzyme by the flavonoid(s) with strong UGT1A1 induction activity. Among all the tested flavonoids, neobavaisoflavone (NBIF) displayed the most potent UGT1A1 induction activity, while its inductive effects were confirmed by both western blot and glucuronidation activity assays. A panel of nuclear receptor reporter assays demonstrated that NBIF activated PPARα and PPARγ in a dose-dependent manner. Meanwhile, we also found that NBIF could up-regulate the expression of PPARα and PPARγ in hepatic cells, suggesting that the induction of UGT1A1 by NBIF was mainly mediated by PPARs. In silico simulations showed that NBIF could stably bind on pocket II of PPARα and PPARγ. Collectively, our results demonstrated that NBIF is a natural inducer of UGT1A1, while this agent induced UGT1A1 mainly via activating and up-regulating PPARα and PPARγ. These findings suggested that NBIF can be used as a promising lead compound for the development of more efficacious UGT1A1 inducers to treat hyperbilirubinaemia and UGT1A1-associated drug toxicities.
Collapse
Affiliation(s)
- Ya-Di Zhu
- Trauma Emergency Center, The Seventh Affiliated People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Qing Guan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Peng
- Trauma Emergency Center, The Seventh Affiliated People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ying-Yuan Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Rui-Min Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hui-Chang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ming Lei
- Trauma Emergency Center, The Seventh Affiliated People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan-Dan Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
41
|
Raju B, Choudhary S, Narendra G, Verma H, Silakari O. Molecular modeling approaches to address drug-metabolizing enzymes (DMEs) mediated chemoresistance: a review. Drug Metab Rev 2021; 53:45-75. [PMID: 33535824 DOI: 10.1080/03602532.2021.1874406] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Resistance against clinically approved anticancer drugs is the main roadblock in cancer treatment. Drug metabolizing enzymes (DMEs) that are capable of metabolizing a variety of xenobiotic get overexpressed in malignant cells, therefore, catalyzing drug inactivation. As evident from the literature reports, the levels of DMEs increase in cancer cells that ultimately lead to drug inactivation followed by drug resistance. To puzzle out this issue, several strategies inclusive of analog designing, prodrug designing, and inhibitor designing have been forged. On that front, the implementation of computational tools can be considered a fascinating approach to address the problem of chemoresistance. Various research groups have adopted different molecular modeling tools for the investigation of DMEs mediated toxicity problems. However, the utilization of these in-silico tools in maneuvering the DME mediated chemoresistance is least considered and yet to be explored. These tools can be employed in the designing of such chemotherapeutic agents that are devoid of the resistance problem. The current review canvasses various molecular modeling approaches that can be implemented to address this issue. Special focus was laid on the development of specific inhibitors of DMEs. Additionally, the strategies to bypass the DMEs mediated drug metabolism were also contemplated in this report that includes analogs and pro-drugs designing. Different strategies discussed in the review will be beneficial in designing novel chemotherapeutic agents that depreciate the resistance problem.
Collapse
Affiliation(s)
- Baddipadige Raju
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Gera Narendra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
42
|
Zhou J, Argikar UA, Miners JO. Enzyme Kinetics of Uridine Diphosphate Glucuronosyltransferases (UGTs). Methods Mol Biol 2021; 2342:301-338. [PMID: 34272700 DOI: 10.1007/978-1-0716-1554-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glucuronidation, catalyzed by uridine diphosphate glucuronosyltransferases (UGTs), is an important process for the metabolism and clearance of many lipophilic chemicals, including drugs, environmental chemicals, and endogenous compounds. Glucuronidation is a bisubstrate reaction that requires the aglycone and the cofactor, UDP-GlcUA. Accumulating evidence suggests that the bisubstrate reaction follows a compulsory-order ternary mechanism. To simplify the kinetic modeling of glucuronidation reactions in vitro, UDP-GlcUA is usually added to incubations in large excess. Many factors have been shown to influence UGT activity and kinetics in vitro, and these must be accounted for during experimental design and data interpretation. While the assessment of drug-drug interactions resulting from UGT inhibition has been challenging in the past, the increasing availability of UGT enzyme-selective substrate and inhibitor "probes" provides the prospect for more reliable reaction phenotyping and assessment of drug-drug interaction potential. Although extrapolation of the in vitro intrinsic clearance of a glucuronidated drug often underpredicts in vivo clearance, careful selection of in vitro experimental conditions and inclusion of extrahepatic glucuronidation may improve the predictivity of in vitro-in vivo extrapolation. Physiologically based pharmacokinetic (PBPK) modeling has also shown to be of value for predicting PK of drugs eliminated by glucuronidation.
Collapse
Affiliation(s)
- Jin Zhou
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA.
| | - Upendra A Argikar
- Translational Medicine, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA
| | - John O Miners
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
43
|
Connors NJ, Mazer-Amirshahi M, Motov S, Kim HK. Relative addictive potential of opioid analgesic agents. Pain Manag 2020; 11:201-215. [PMID: 33300384 DOI: 10.2217/pmt-2020-0048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Opioid overdoses and deaths continue to be a problem in the USA with a significant portion related to prescribed opioid analgesic agents. The role of pharmacogentic factors in opioid addiction is an active area of research. While all opioid analgesic agents have some addictive potential, it is clear that there are some with greater addictive potential. Oxycodone is the most widely abused opioid analgesic and it appears to predispose to chronic use with high likability by users. Fentanyl and hydromorphone are both very lipophilic allowing rapid penetration into the CNS, but are not rated as highly as other agents. Providers should consider the risk of addiction with the opioids they prescribe and give those with a lower addictive potential.
Collapse
Affiliation(s)
- Nicholas J Connors
- HCA Healthcare, Trident Medical Center, Charleston, SC 29406, USA.,Palmetto Poison Center, Columbia, SC 29201, USA
| | - Maryann Mazer-Amirshahi
- Department of Emergency Medicine, MedStar Washington Hospital Center, Washington, DC 20010, USA
| | - Sergey Motov
- Department of Emergency Medicine, Maimonides Medical Center, Brooklyn, NY 11219, USA
| | - Hong K Kim
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
44
|
Li X, Lu Y, Ou X, Zeng S, Wang Y, Qi X, Zhu L, Liu Z. Changes and sex- and age-related differences in the expression of drug metabolizing enzymes in a KRAS-mutant mouse model of lung cancer. PeerJ 2020; 8:e10182. [PMID: 33240601 PMCID: PMC7680056 DOI: 10.7717/peerj.10182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/23/2020] [Indexed: 01/16/2023] Open
Abstract
Background This study aimed to systematically profile the alterations and sex- and age-related differences in the drug metabolizing enzymes (DMEs) in a KRAS-mutant mouse model of lung cancer (KRAS mice). Methodology In this study, the LC-MS/MS approach and a probe substrate method were used to detect the alterations in 21 isoforms of DMEs, as well as the enzymatic activities of five isoforms, respectively. Western blotting was applied to study the protein expression of four related receptors. Results The proteins contents of CYP2C29 and CYP3A11, were significantly downregulated in the livers of male KRAS mice at 26 weeks (3.7- and 4.4-fold, respectively, p < 0.05). SULT1A1 and SULT1D1 were upregulated by 1.8- to 7.0- fold at 20 (p = 0.015 and 0.017, respectively) and 26 weeks (p = 0.055 and 0.031, respectively). There were positive correlations between protein expression and enzyme activity for CYP2E1, UGT1A9, SULT1A1 and SULT1D1 (r2 ≥ 0.5, p < 0.001). Western blotting analysis revealed the downregulation of AHR, FXR and PPARα protein expression in male KRAS mice at 26 weeks. For sex-related differences, CYP2E1 was male-predominant and UGT1A2 was female-predominant in the kidney. UGT1A1 and UGT1A5 expression was female-predominant, whereas UGT2B1 exhibited male-predominant expression in liver tissue. For the tissue distribution of DMEs, 21 subtypes of DMEs were all expressed in liver tissue. In the intestine, the expression levels of CYP2C29, CYP27A1, UGT1A2, 1A5, 1A6a, 1A9, 2B1, 2B5 and 2B36 were under the limitation of quantification. The subtypes of CYP7A1, 1B1, 2E1 and UGT1A1, 2A3, 2B34 were detected in kidney tissue. Conclusions This study, for the first time, unveils the variations and sex- and age-related differences in DMEs in C57 BL/6 (WT) mice and KRAS mice.
Collapse
Affiliation(s)
- Xiaoyan Li
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiyan Lu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojun Ou
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sijing Zeng
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoxiao Qi
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lijun Zhu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
45
|
Kim YA, Sarto Basso R, Wojtowicz D, Liu AS, Hochbaum DS, Vandin F, Przytycka TM. Identifying Drug Sensitivity Subnetworks with NETPHIX. iScience 2020; 23:101619. [PMID: 33089107 PMCID: PMC7566085 DOI: 10.1016/j.isci.2020.101619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 09/08/2020] [Accepted: 09/24/2020] [Indexed: 12/29/2022] Open
Abstract
Phenotypic heterogeneity in cancer is often caused by different patterns of genetic alterations. Understanding such phenotype-genotype relationships is fundamental for the advance of personalized medicine. We develop a computational method, named NETPHIX (NETwork-to-PHenotype association with eXclusivity) to identify subnetworks of genes whose genetic alterations are associated with drug response or other continuous cancer phenotypes. Leveraging interaction information among genes and properties of cancer mutations such as mutual exclusivity, we formulate the problem as an integer linear program and solve it optimally to obtain a subnetwork of associated genes. Applied to a large-scale drug screening dataset, NETPHIX uncovered gene modules significantly associated with drug responses. Utilizing interaction information, NETPHIX modules are functionally coherent and can thus provide important insights into drug action. In addition, we show that modules identified by NETPHIX together with their association patterns can be leveraged to suggest drug combinations.
Collapse
Affiliation(s)
- Yoo-Ah Kim
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD 20894, USA
| | - Rebecca Sarto Basso
- Department of Industrial Engineering and Operations Research, University of California at Berkeley, Berkeley, CA 94709, USA
| | - Damian Wojtowicz
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD 20894, USA
| | - Amanda S Liu
- Montgomery Blair High School, Silver Spring, MD 20901, USA
| | - Dorit S Hochbaum
- Department of Industrial Engineering and Operations Research, University of California at Berkeley, Berkeley, CA 94709, USA
| | - Fabio Vandin
- Department of Information Engineering, University of Padova, Padova 35131, Italy
| | - Teresa M Przytycka
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD 20894, USA
| |
Collapse
|
46
|
Miners JO, Rowland A, Novak JJ, Lapham K, Goosen TC. Evidence-based strategies for the characterisation of human drug and chemical glucuronidation in vitro and UDP-glucuronosyltransferase reaction phenotyping. Pharmacol Ther 2020; 218:107689. [PMID: 32980440 DOI: 10.1016/j.pharmthera.2020.107689] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/26/2022]
Abstract
Enzymes of the UDP-glucuronosyltransferase (UGT) superfamily contribute to the elimination of drugs from almost all therapeutic classes. Awareness of the importance of glucuronidation as a drug clearance mechanism along with increased knowledge of the enzymology of drug and chemical metabolism has stimulated interest in the development and application of approaches for the characterisation of human drug glucuronidation in vitro, in particular reaction phenotyping (the fractional contribution of the individual UGT enzymes responsible for the glucuronidation of a given drug), assessment of metabolic stability, and UGT enzyme inhibition by drugs and other xenobiotics. In turn, this has permitted the implementation of in vitro - in vivo extrapolation approaches for the prediction of drug metabolic clearance, intestinal availability, and drug-drug interaction liability, all of which are of considerable importance in pre-clinical drug development. Indeed, regulatory agencies (FDA and EMA) require UGT reaction phenotyping for new chemical entities if glucuronidation accounts for ≥25% of total metabolism. In vitro studies are most commonly performed with recombinant UGT enzymes and human liver microsomes (HLM) as the enzyme sources. Despite the widespread use of in vitro approaches for the characterisation of drug and chemical glucuronidation by HLM and recombinant enzymes, evidence-based guidelines relating to experimental approaches are lacking. Here we present evidence-based strategies for the characterisation of drug and chemical glucuronidation in vitro, and for UGT reaction phenotyping. We anticipate that the strategies will inform practice, encourage development of standardised experimental procedures where feasible, and guide ongoing research in the field.
Collapse
Affiliation(s)
- John O Miners
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Andrew Rowland
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | | | | | | |
Collapse
|
47
|
Neuvonen M, Hirvensalo P, Tornio A, Rago B, West M, Lazzaro S, Mathialagan S, Varma M, Cerny MA, Costales C, Ramanathan R, Rodrigues AD, Niemi M. Identification of Glycochenodeoxycholate 3-O-Glucuronide and Glycodeoxycholate 3-O-Glucuronide as Highly Sensitive and Specific OATP1B1 Biomarkers. Clin Pharmacol Ther 2020; 109:646-657. [PMID: 32961594 PMCID: PMC7983942 DOI: 10.1002/cpt.2053] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/07/2020] [Indexed: 11/09/2022]
Abstract
The aim of this study was to investigate the sensitivity and specificity of endogenous glycochenodeoxycholate and glycodeoxycholate 3-O-glucuronides (GCDCA-3G and GDCA-3G) as substrates for organic anion transporting polypeptide 1B1 (OATP1B1) in humans. We measured fasting levels of plasma GCDCA-3G and GDCA-3G using liquid chromatography-tandem mass spectrometry in 356 healthy volunteers. The mean plasma levels of both compounds were ~ 50% lower in women than in men (P = 2.25 × 10-18 and P = 4.73 × 10-9 ). In a microarray-based genome-wide association study, the SLCO1B1 rs4149056 (c.521T>C, p.Val174Ala) variation showed the strongest association with the plasma GCDCA-3G (P = 3.09 × 10-30 ) and GDCA-3G (P = 1.60 × 10-17 ) concentrations. The mean plasma concentration of GCDCA-3G was 9.2-fold (P = 8.77 × 10-31 ) and that of GDCA-3G was 6.4-fold (P = 2.45x10-13 ) higher in individuals with the SLCO1B1 c.521C/C genotype than in those with the c.521T/T genotype. No other variants showed independent genome-wide significant associations with GCDCA-3G or GDCA-3G. GCDCA-3G was highly efficacious in detecting the SLCO1B1 c.521C/C genotype with an area under the receiver operating characteristic curve of 0.996 (P < 0.0001). The sensitivity (98-99%) and specificity (100%) peaked at a cutoff value of 180 ng/mL for men and 90 ng/mL for women. In a haplotype-based analysis, SLCO1B1*5 and *15 were associated with reduced, and SLCO1B1*1B, *14, and *35 with increased OATP1B1 function. In vitro, both GCDCA-3G and GDCA-3G showed at least 6 times higher uptake by OATP1B1 than OATP1B3 or OATP2B1. These data indicate that the hepatic uptake of GCDCA-3G and GDCA-3G is predominantly mediated by OATP1B1. GCDCA-3G, in particular, is a highly sensitive and specific OATP1B1 biomarker in humans.
Collapse
Affiliation(s)
- Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Päivi Hirvensalo
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Brian Rago
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Mark West
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Sarah Lazzaro
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | | | - Manthena Varma
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Matthew A Cerny
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Chester Costales
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Ragu Ramanathan
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - A David Rodrigues
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| |
Collapse
|
48
|
Miyauchi Y, Kurohara K, Kimura A, Esaki M, Fujimoto K, Hirota Y, Takechi S, Mackenzie PI, Ishii Y, Tanaka Y. The carboxyl-terminal di-lysine motif is essential for catalytic activity of UDP-glucuronosyltransferase 1A9. Drug Metab Pharmacokinet 2020; 35:466-474. [PMID: 32883578 DOI: 10.1016/j.dmpk.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 11/25/2022]
Abstract
UDP-Glucuronosyltransferase (UGT) is a type I membrane protein localized to the endoplasmic reticulum (ER). UGT has a di-lysine motif (KKXX/KXKXX) in its cytoplasmic domain, which is defined as an ER retention signal. However, our previous study has revealed that UGT2B7, one of the major UGT isoform in human, localizes to the ER in a manner that is independent of this motif. In this study, we focused on another UGT isoform, UGT1A9, and investigated the role of the di-lysine motif in its ER localization, glucuronidation activity, and homo-oligomer formation. Immunofluorescence microscopy indicated that the cytoplasmic domain of UGT1A9 functioned as an ER retention signal in a chimeric protein with CD4, but UGT1A9 itself could localize to the ER in a di-lysine motif-independent manner. In addition, UGT1A9 formed homo-oligomers in the absence of the motif. However, deletion of the di-lysine motif or substitution of lysines in the motif for alanines, severely impaired glucuronidation activity of UGT1A9. This is the first study that re-defines the cytoplasmic di-lysine motif of UGT as an essential peptide for retaining glucuronidation capacity.
Collapse
Affiliation(s)
- Yuu Miyauchi
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan; Laboratory of Hygienic Chemistry, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan.
| | - Ken Kurohara
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akane Kimura
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Madoka Esaki
- Laboratory of Hygienic Chemistry, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Keiko Fujimoto
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuko Hirota
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Takechi
- Laboratory of Hygienic Chemistry, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Peter I Mackenzie
- Clinical Pharmacology, College of Medicine and Public Health, Flinders Medical Centre and Flinders University, Adelaide, Australia
| | - Yuji Ishii
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan; Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | - Yoshitaka Tanaka
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
49
|
Sharma S, Durairaj P, Bureik M. Rapid and convenient biotransformation procedure for human drug metabolizing enzymes using permeabilized fission yeast cells. Anal Biochem 2020; 607:113704. [PMID: 32697953 DOI: 10.1016/j.ab.2020.113704] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/14/2022]
Abstract
The development of convenient assays for the in vitro study of drug metabolizing enzymes (DMEs) such as cytochromes P450 (CYPs) and UDP-glucuronosyltransferases (UGTs) greatly facilitates metabolism studies of candidate drug compounds and other xenobiotics. We have developed and optimized an experimental approach that combines the advantages of recombinant expression in yeast with a microsomal-like biotransformation and thus allows for rapid and convenient enzymatic assays. Recombinant strains of the fission yeast Schizosaccharomyces pombe have previously been demonstrated to functionally express human CYPs and UGTs. Permeabilization of such cells with Triton X-100 results in the formation of enzyme bags, which can be used as biocatalysts. This protocol describes the preparation of such enzyme bags (3 h) and their application in enzyme activity assays (4 h) utilizing either pro-luminescent substrates and luminescence measurements or non-luminescent substrates and liquid chromatography coupled to mass spectrometry (LC-MS). Both applications provide practical tools for investigating CYP and UGT reactions in vitro without the need for additional sophisticated instrumentation or expertise.
Collapse
Affiliation(s)
- Shishir Sharma
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
| | - Pradeepraj Durairaj
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
50
|
Hirvensalo P, Tornio A, Launiainen T, Paile‐Hyvärinen M, Tapaninen T, Neuvonen M, Backman JT, Niemi M. UGT1A3 and Sex Are Major Determinants of Telmisartan Pharmacokinetics—A Comprehensive Pharmacogenomic Study. Clin Pharmacol Ther 2020; 108:885-895. [DOI: 10.1002/cpt.1928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Päivi Hirvensalo
- Department of Clinical Pharmacology University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital Helsinki Finland
- Individualized Drug Therapy Research Program Faculty of Medicine University of Helsinki Helsinki Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital Helsinki Finland
- Individualized Drug Therapy Research Program Faculty of Medicine University of Helsinki Helsinki Finland
| | - Terhi Launiainen
- Department of Clinical Pharmacology University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital Helsinki Finland
- Individualized Drug Therapy Research Program Faculty of Medicine University of Helsinki Helsinki Finland
| | - Maria Paile‐Hyvärinen
- Department of Clinical Pharmacology University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital Helsinki Finland
- Individualized Drug Therapy Research Program Faculty of Medicine University of Helsinki Helsinki Finland
| | - Tuija Tapaninen
- Department of Clinical Pharmacology University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital Helsinki Finland
- Individualized Drug Therapy Research Program Faculty of Medicine University of Helsinki Helsinki Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital Helsinki Finland
- Individualized Drug Therapy Research Program Faculty of Medicine University of Helsinki Helsinki Finland
| | - Janne T. Backman
- Department of Clinical Pharmacology University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital Helsinki Finland
- Individualized Drug Therapy Research Program Faculty of Medicine University of Helsinki Helsinki Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital Helsinki Finland
- Individualized Drug Therapy Research Program Faculty of Medicine University of Helsinki Helsinki Finland
| |
Collapse
|