1
|
Yumoto Y, Endo T, Harada H, Kobayashi K, Nakabayashi T, Abe Y. High-throughput assay to simultaneously evaluate activation of CYP3A and the direct and time-dependent inhibition of CYP3A, CYP2C9, and CYP2D6 using liquid chromatography-tandem mass spectrometry. Xenobiotica 2024; 54:45-56. [PMID: 38265764 DOI: 10.1080/00498254.2024.2308818] [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: 10/25/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024]
Abstract
In the early stages of drug discovery, adequate evaluation of the potential drug-drug interactions (DDIs) of drug candidates is important. Several CYP3A activators are known to lead to underestimation of DDIs. These compounds affect midazolam 1'-hydroxylation but not midazolam 4-hydroxylation.We used both metabolic reactions of midazolam to evaluate the activation and inhibition of CYP3A activators simultaneously. For our CYP inhibition assay using cocktail probe substrates, simultaneous liquid chromatography-tandem mass spectrometry monitoring of 1'-hydroxymidazolam and 4-hydroxymidazolam for CYP3A was established in addition to monitoring of 4-hydroxydiclofenac and 1'-hydroxybufuralol for CYP2C9 and CYP2D6.The results of our cocktail inhibition assay were well correlated with those of a single inhibition assay, as were the estimated inhibition parameters for typical CYP3A inhibitors. In our assay, a proprietary compound that activated midazolam 1'-hydroxylation and tended to inhibit 4-hydroxylation was evaluated along with known CYP3A activators. All compounds were well characterised by comparison of the results of midazolam 1'- and 4-hydroxylation.In conclusion, our CYP cocktail inhibition assay can detect CYP3A activation and assess the direct and time-dependent inhibition potentials for CYP3A, CYP2C9, and CYP2D6. This method is expected to be very efficient in the early stages of drug discovery.
Collapse
Affiliation(s)
- Yu Yumoto
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Takuro Endo
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Hiroshi Harada
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Kaoru Kobayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Takeshi Nakabayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Yoshikazu Abe
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| |
Collapse
|
2
|
Pelkonen O, Abass K, Parra Morte JM, Panzarea M, Testai E, Rudaz S, Louisse J, Gundert-Remy U, Wolterink G, Jean-Lou CM D, Coecke S, Bernasconi C. Metabolites in the regulatory risk assessment of pesticides in the EU. FRONTIERS IN TOXICOLOGY 2023; 5:1304885. [PMID: 38188093 PMCID: PMC10770266 DOI: 10.3389/ftox.2023.1304885] [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: 09/30/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
A large majority of chemicals is converted into metabolites through xenobiotic-metabolising enzymes. Metabolites may present a spectrum of characteristics varying from similar to vastly different compared with the parent compound in terms of both toxicokinetics and toxicodynamics. In the pesticide arena, the role of metabolism and metabolites is increasingly recognised as a significant factor particularly for the design and interpretation of mammalian toxicological studies and in the toxicity assessment of pesticide/metabolite-associated issues for hazard characterization and risk assessment purposes, including the role of metabolites as parts in various residues in ecotoxicological adversities. This is of particular relevance to pesticide metabolites that are unique to humans in comparison with metabolites found in in vitro or in vivo animal studies, but also to disproportionate metabolites (quantitative differences) between humans and mammalian species. Presence of unique or disproportionate metabolites may underlie potential toxicological concerns. This review aims to present the current state-of-the-art of comparative metabolism and metabolites in pesticide research for hazard and risk assessment, including One Health perspectives, and future research needs based on the experiences gained at the European Food Safety Authority.
Collapse
Affiliation(s)
- Olavi Pelkonen
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Khaled Abass
- Department of Environmental Health Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research (SIMR), University of Sharjah, Sharjah, United Arab Emirates
- Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
| | | | | | - Emanuela Testai
- Mechanisms, Biomarkers and Models Unit, Environment and Health Department, Istituto Superiore di Sanità, Rome, Italy
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
| | - Jochem Louisse
- EFSA, European Food Safety Authority, Parma, Italy
- Wageningen Food Safety Research (WFSR), Wageningen, Netherlands
| | - Ursula Gundert-Remy
- Institute of Clinical Pharmacology and Toxicology, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gerrit Wolterink
- Centre for Prevention, Lifestyle and Health, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | | | - Sandra Coecke
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | |
Collapse
|
3
|
Masimirembwa C, Ramsay M, Botha J, Ellis E, Etheredge H, Hurrell T, Kanji CR, Kapungu NN, Maher H, Mthembu B, Naidoo J, Scholefield J, Rambarran S, van der Schyff F, Smyth N, Strobele B, Thelingwani RS, Loveland J, Fabian J. The African Liver Tissue Biorepository Consortium: Capacitating Population-Appropriate Drug Metabolism, Pharmacokinetics, and Pharmacogenetics Research in Drug Discovery and Development. Drug Metab Dispos 2023; 51:1551-1560. [PMID: 37751997 DOI: 10.1124/dmd.123.001400] [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: 05/24/2023] [Revised: 08/21/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Pharmaceutical companies subject all new molecular entities to a series of in vitro metabolic characterizations that guide the selection and/or design of compounds predicted to have favorable pharmacokinetic properties in humans. Current drug metabolism research is based on liver tissue predominantly obtained from people of European origin, with limited access to tissue from people of African origin. Given the interindividual and interpopulation genomic variability in genes encoding drug-metabolizing enzymes, efficacy and safety of some drugs are poorly predicted for African populations. To address this gap, we have established the first comprehensive liver tissue biorepository inclusive of people of African origin. The African Liver Tissue Biorepository Consortium currently includes three institutions in South Africa and one in Zimbabwe, with plans to expand to other African countries. The program has collected 67 liver samples as of July 2023. DNA from the donors was genotyped for 120 variants in 46 pharmacogenes and revealed variants that are uniquely found in African populations, including the low-activity, African-specific CYP2C9*5 and *8 variants relevant to the metabolism of diclofenac. Larger liver tissue samples were used to isolate primary human hepatocytes. Viability of the hepatocytes and microsomal fractions was demonstrated by the activity of selected cytochrome P450s. This resource will be used to ensure the safety and efficacy of existing and new drugs in African populations. This will be done by characterizing compounds for properties such as drug clearance, metabolite and enzyme identification, and drug-drug and drug-gene interactions. SIGNIFICANCE STATEMENT: Standard optimization of the drug metabolism of new molecular entities in the pharmaceutical industry uses subcellular fractions such as microsomes and isolated primary hepatocytes, being done mainly with tissue from donors of European origin. Pharmacogenetics research has shown that variants in genes coding for drug-metabolizing enzymes have interindividual and interpopulation differences. We established an African liver tissue biorepository that will be useful in ensuring drug discovery and development research takes into account drug responses in people of African origin.
Collapse
Affiliation(s)
- Collen Masimirembwa
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Michele Ramsay
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Jean Botha
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Ewa Ellis
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Harriet Etheredge
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Tracey Hurrell
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Comfort Ropafadzo Kanji
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Nyasha Nicole Kapungu
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Heather Maher
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Busisiwe Mthembu
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Jerolen Naidoo
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Janine Scholefield
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Sharan Rambarran
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Francisca van der Schyff
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Natalie Smyth
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Bernd Strobele
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Roslyn Stella Thelingwani
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Jerome Loveland
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - June Fabian
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| |
Collapse
|
4
|
Keuper-Navis M, Walles M, Poller B, Myszczyszyn A, van der Made TK, Donkers J, Eslami Amirabadi H, Wilmer MJ, Aan S, Spee B, Masereeuw R, van de Steeg E. The application of organ-on-chip models for the prediction of human pharmacokinetic profiles during drug development. Pharmacol Res 2023; 195:106853. [PMID: 37473876 DOI: 10.1016/j.phrs.2023.106853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Organ-on-chip (OoC) technology has led to in vitro models with many new possibilities compared to conventional in vitro and in vivo models. In this review, the potential of OoC models to improve the prediction of human oral bioavailability and intrinsic clearance is discussed, with a focus on the functionality of the models and the application in current drug development practice. Multi-OoC models demonstrating the application for pharmacokinetic (PK) studies are summarized and existing challenges are identified. Physiological parameters for a minimal viable platform of a multi-OoC model to study PK are provided, together with PK specific read-outs and recommendations for relevant reference compounds to validate the model. Finally, the translation to in vivo PK profiles is discussed, which will be required to routinely apply OoC models during drug development.
Collapse
Affiliation(s)
- Marit Keuper-Navis
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Markus Walles
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Birk Poller
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Adam Myszczyszyn
- Faculty of Veterinary Medicine & Regenerative Medicine Center Utrecht (RMCU), Utrecht University, Utrecht, the Netherlands
| | - Thomas K van der Made
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Joanne Donkers
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | | | | | - Saskia Aan
- Stichting Proefdiervrij, Den Haag, the Netherlands
| | - Bart Spee
- Faculty of Veterinary Medicine & Regenerative Medicine Center Utrecht (RMCU), Utrecht University, Utrecht, the Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands.
| |
Collapse
|
5
|
Lin F, Lin X, Wang X, Mei G, Chen B, Yao H, Huang L. Inhibitory effect of Selaginella doederleinii hieron on human cytochrome P450. Front Pharmacol 2023; 14:1108867. [PMID: 36874034 PMCID: PMC9975586 DOI: 10.3389/fphar.2023.1108867] [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: 11/26/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Introduction: Selaginella doederleinii Hieron is a traditional Chinese herbal medicine, the ethyl acetate extract from Selaginella doederleinii (SDEA) showed favorable anticancer potentials. However, the effect of SDEA on human cytochrome P450 enzymes (CYP450) remains unclear. To predict the herb-drug interaction (HDI) and lay the groundwork for further clinical trials, the inhibitory effect of SDEA and its four constituents (Amentoflavone, Palmatine, Apigenin, Delicaflavone) on seven CYP450 isoforms were investigated by using the established CYP450 cocktail assay based on LC-MS/MS. Methods: Appropriate substrates for seven tested CYP450 isoforms were selected to establish a reliable cocktail CYP450 assay based on LC-MS/MS. The contents of four constituents (Amentoflavone, Palmatine, Apigenin, Delicaflavone) in SDEA were determined as well. Then, the validated CYP450 cocktail assay was applied to test the inhibitory potential of SDEA and four constituents on CYP450 isoforms. Results: SDEA showed strong inhibitory effect on CYP2C9 and CYP2C8 (IC50 ≈ 1 μg/ml), moderate inhibitory effect against CYP2C19, CYP2E1 and CYP3A (IC50 < 10 μg/ml). Among the four constituents, Amentoflavone had the highest content in the extract (13.65%) and strongest inhibitory effect (IC50 < 5 μM), especially for CYP2C9, CYP2C8 and CYP3A. Amentoflavone also showed time-dependent inhibition on CYP2C19 and CYP2D6. Apigenin and Palmatine both showed concentration-dependent inhibition. Apigenin inhibited CYP1A2, CYP2C8, CYP2C9, CYP2E1 and CYP3A. Palmatine inhibited CYP3A and had a weak inhibitory effect on CYP2E1. As for Delicaflavone, which has the potential to develop as an anti-cancer agent, showed no obvious inhibitory effect on CYP450 enzymes. Conclusion: Amentoflavone may be one of the main reasons for the inhibition of SDEA on CYP450 enzymes, the potential HDI should be considered when SDEA or Amentoflavone were used with other clinical drugs. On the contrast, Delicaflavone is more suitable to develop as a drug for clinical use, considering the low level of CYP450 metabolic inhibition.
Collapse
Affiliation(s)
- Fei Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xinhua Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xuewen Wang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Guanghui Mei
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bing Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Hong Yao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Lingyi Huang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
6
|
Schleiff MA, Crosby S, Blue M, Schleiff BM, Boysen G, Miller GP. CYP2C9 and 3A4 play opposing roles in bioactivation and detoxification of diphenylamine NSAIDs. Biochem Pharmacol 2021; 194:114824. [PMID: 34748821 DOI: 10.1016/j.bcp.2021.114824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/25/2022]
Abstract
Diphenylamine NSAIDs are taken frequently for chronic pain conditions, yet their use may potentiate hepatotoxicity risks through poorly characterized metabolic mechanisms. Our previous work revealed that seven marketed or withdrawn diphenylamine NSAIDs undergo bioactivation into quinone-species metabolites, whose reaction specificities depended on halogenation and the type of acidic group on the diphenylamine. Herein, we identified cytochromes P450 responsible for those bioactivations, determined reaction specificities, and estimated relative contributions of enzymes to overall hepatic bioactivations and detoxifications. A qualitative activity screen revealed CYP2C8, 2C9, 2C19, and 3A4 played roles in drug bioactivation. Subsequent steady-state studies with recombinant CYPs recapitulated the importance of halogenation and acidic group type on bioactivations but importantly, showed patterns unique to each CYP. CYP2C9, 2C19 and 3A4 bioactivated all NSAIDs with CYP2C9 dominating all possible bioactivation pathways. For each CYP, specificities for overall oxidative metabolism were not impacted significantly by differences in NSAID structures but the values themselves differed among the enzymes such that CYP2C9 and 3A4 were more efficient than others. When considering hepatic CYP abundance, CYP2C9 almost exclusively accounted for diphenylamine NSAID bioactivations, whereas CYP3A4 provided a critical counterbalance favoring their overall detoxification. Preference for either outcome would depend on molecular structures favoring metabolism by the CYPs as well as the influence of clinical factors altering their expression and/or activity. While focused on NSAIDs, these findings have broader implications on bioactivation risks given the expansion of the diphenylamine scaffold to other drug classes such as targeted cancer therapeutics.
Collapse
Affiliation(s)
- Mary Alexandra Schleiff
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Samantha Crosby
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Madison Blue
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Benjamin Mark Schleiff
- Independent Researcher, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Gunnar Boysen
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Grover Paul Miller
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| |
Collapse
|
7
|
Habenschus MD, Carrão DB, de Albuquerque NCP, Perovani IS, Moreira da Silva R, Nardini V, Lopes NP, Dias LG, Moraes de Oliveira AR. In vitro enantioselective inhibition of the main human CYP450 enzymes involved in drug metabolism by the chiral pesticide tebuconazole. Toxicol Lett 2021; 351:1-9. [PMID: 34407455 DOI: 10.1016/j.toxlet.2021.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/23/2021] [Accepted: 08/13/2021] [Indexed: 11/29/2022]
Abstract
Tebuconazole (TEB) is a chiral triazole fungicide worldwide employed to control plant pathogens and preserve wood. People can be exposed to TEB either through diet and occupational contamination. This work investigates the in vitro inhibitory potential of rac-TEB, S-(+)-TEB, and R-(-)-TEB over the main cytochrome P450 enzymes (CYP450) using human liver microsomes to predict TEB in vivo inhibition potential. The IC50 values showed that in vitro inhibition was enantioselective for CYP2C9, CYP2C19, and CYP2D6, but not for CYP3A4/5. Despite enantioselectivity, rac-TEB and its single enantiomers were always classified in the same category. The inhibition mechanisms and constants were determined for rac-TEB and it has shown to be a mixed inhibitor of CYP3A4/5 (Ki = 1.3 ± 0.3 μM, αKi = 3.2 ± 0.5 μM; Ki = 0.6 ± 0.3 μM, αKi = 1.3 ± 0.3 μM) and CYP2C9 (Ki = 0.7 ± 0.1 μM, αKi = 2.7 ± 0.5 μM), and a competitive inhibitor of CYP2D6 (Ki = 11.9 ± 0.7 μM) and CYP2C19 (Ki = 0.23 ± 0.02 μM), respectively, suggesting that in some cases, rac-TEB has a higher or comparable inhibitory potential than well-known strong inhibitors of CYP450 enzymes, especially for CYP2C9 and CYP2C19. In vitro-in vivo extrapolations (IVIVE) were conducted based on the results and data available in the literature about TEB absorption and metabolism. R1 values were estimated based on the Food and Drug Administration guideline and suggested that in a chronic oral exposure scenario considering the acceptable daily intake dose proposed by the European Food and Safety Authority, the hypothesis of rac-TEB to inhibit the activities of CYP3A4/5, CYP2C9, and CYP2C19 in vivo and cause pesticide-drug interactions cannot be disregarded.
Collapse
Affiliation(s)
- Maísa Daniela Habenschus
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Daniel Blascke Carrão
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Nayara Cristina Perez de Albuquerque
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Icaro Salgado Perovani
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Rodrigo Moreira da Silva
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14090-903, Ribeirão Preto, SP, Brazil
| | - Viviani Nardini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Norberto Peporine Lopes
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14090-903, Ribeirão Preto, SP, Brazil
| | - Luís Gustavo Dias
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Anderson Rodrigo Moraes de Oliveira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil; National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Unesp, Institute of Chemistry, P.O. Box 355, 14800-900, Araraquara, SP, Brazil.
| |
Collapse
|
8
|
Ilexsaponin A1: In vitro metabolites identification and evaluation of inhibitory drug-drug interactions. Drug Metab Pharmacokinet 2021; 40:100415. [PMID: 34461570 DOI: 10.1016/j.dmpk.2021.100415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/23/2021] [Accepted: 07/23/2021] [Indexed: 11/23/2022]
Abstract
As a triterpene saponin, ilexsaponin A1 is one of the most abundant, representative and active components in plants of Ilex pubescens, used in the treatment of cardiovascular diseases. This study aimed to identify the metabolites of ilexsaponin A1 and evaluate its in vitro inhibitory drug-drug interaction (DDI) potential by using human liver microsomes (HLM) and cytochrome P450 enzymes (CYPs)-specific probes, with all the qualitative and quantitative analysis performed by LC-MS/MS. As a result, two metabolites generated through the metabolic pathways of glucuronic acid conjugation and glucose conjugation were first time detected in the HLM. An inhibitory DDI evaluating system consisting of 7 major CYP enzymes involving 8 CYP-catalyzed reactions was established, validated and then used for the DDI evaluation. Our data suggested ilexsaponin A1 and its metabolite, ilexgenin A, are not direct or mechanism-based inhibitors of CYP1A2, 2B6, 2C8, 2C9, 2D6, 2E1 or 3A4/5 at 0.05-10 μM. A significant decreased remaining activity of CYP2B6 (from 77.89 % to 23.19 %) was observed in a dose-dependent manner when increased the concentration of ilexsaponin A1 from 50 to 500 μM. Collectively, our data demonstrate ilexsaponin A1 is unlikely to cause DDIs by inhibiting co-administered drugs metabolized by these CYP enzymes.
Collapse
|
9
|
Shi Y, Meng D, Wang S, Geng P, Xu T, Zhou Q, Zhou Y, Li W, Chen X. Effects of Avitinib on CYP450 Enzyme Activity in vitro and in vivo in Rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3661-3673. [PMID: 34456561 PMCID: PMC8387736 DOI: 10.2147/dddt.s323186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/05/2021] [Indexed: 12/23/2022]
Abstract
Purpose Avitinib is the first third-generation epithelial growth factor receptor (EGFR) inhibitor independently developed in China and is mainly used for treating non-small cell lung cancer. However, pharmacokinetic details are limited. This study explored the in vivo and in vitro effects of avitinib on cytochrome CYP450 enzymes metabolic activity. Methods A rapid and sensitive ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was developed and validated for determining six probe substrates and their metabolites. Avitinib influence on activity levels of CYP isozymes was examined in vitro using human and rat liver microsomes (HLMs/RLMs). For in vivo studies, rats were pretreated with 30 mg/kg avitinib once daily for 7 days (avitinib multiple-doses group), 30 mg/kg avitinib on day 7 (avitinib single-dose group), or an equivalent amount of CMC-Na once daily for 7 days (control group), followed by intragastrical administration of the probe substrates (1 mg/kg tolbutamide and 10 mg/kg phenacetin, bupropion, chlorzoxazone, dextromethorphan, and midazolam). Plasma pharmacokinetics and IC50 values of the probe substrates were then compared. Pharmacokinetic parameters were determined using non-compartmental analysis implemented in a pharmacokinetic program. Results In vitro experiments revealed different inhibitory effects of avitinib on the six probe substrates with various IC50 values (bupropion, 6.39/22.64 μM; phenacetin, 15.79/48.36 μM; chlorzoxazone, 23.15/57.09 μM; midazolam, 27.64/59.6 μM; tolbutamide, 42.18/6.91 μM; dextromethorphan, 44.39/56.57 μM, in RLMs and HLMs respectively). In vivo analysis revealed significant differences (P <0.05) in distinct pharmacokinetic parameters (AUC(0-t), AUC (0-∞), Cmax, MRT(0-t), MRT (0-∞), and CLz/F) for the six probe substrates after avitinib pretreatment. Conclusion A sensitive and reliable UPLC-MS/MS method was established to determine the concentration of six probe substrates in rat plasma. Avitinib had inhibitory effects on CYP450 enzymes, especially cyp2b1, cyp1a2 in RLMs, CYP2C9 in HLMs, and cyp1a2, cyp2b1, cyp2d1, and cyp2e1 in vivo. Our data recommend caution when avitinib was taken simultaneously with drugs metabolized by CYP450 enzymes.
Collapse
Affiliation(s)
- Yong Shi
- Comprehensive Breast Health Center, Department of Thyroid and Breast Surgery, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, 323000, People's Republic of China
| | - Deru Meng
- Comprehensive Breast Health Center, Department of Thyroid and Breast Surgery, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, 323000, People's Republic of China
| | - Shuanghu Wang
- Comprehensive Breast Health Center, Department of Thyroid and Breast Surgery, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, 323000, People's Republic of China
| | - Peiwu Geng
- Comprehensive Breast Health Center, Department of Thyroid and Breast Surgery, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, 323000, People's Republic of China
| | - Tao Xu
- Department of Pharmacy, Ningbo First Hospital, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Quan Zhou
- Comprehensive Breast Health Center, Department of Thyroid and Breast Surgery, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, 323000, People's Republic of China
| | - Yunfang Zhou
- Comprehensive Breast Health Center, Department of Thyroid and Breast Surgery, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, 323000, People's Republic of China
| | - Wanshu Li
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Xugao Chen
- Department of Radiology, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, 323000, People's Republic of China
| |
Collapse
|
10
|
El-Wafaey DI, Nafea OE, Faruk EM. Naringenin alleviates hepatic injury in zinc oxide nanoparticles exposed rats: impact on oxido-inflammatory stress and apoptotic cell death. Toxicol Mech Methods 2021; 32:58-66. [PMID: 34348583 DOI: 10.1080/15376516.2021.1965275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Human exposure to nanoparticles became unavoidable secondary to their massive involvement in a multitude of industrial applications. Zinc oxide nanoparticles (ZnONPs) are one of the most commonly used metal oxide nanoparticles in biological applications. Naringenin (NAR), a citrus-derived flavonoid, has favorable biological properties that promote human health. The present study was carried out to investigate the possible defensive role of NAR versus ZnONPs provoked hepatic injury in rats through an evaluation of liver enzymes, hepatic biomarkers of oxidative stress, inflammatory process, apoptotic cell death along with histopathological examination of liver tissue. Therefore, 32 adult rats were randomly divided into four equal groups as control, NAR, ZnONPs and co-treated ZnONPs with NAR groups. All treatments were administered for 14 days. Our results showed that ZnONPs induced hepatic injury as documented by the marked increased in hepatic enzymes activities, disturbed hepatic oxidant/antioxidant balance, increased hepatic inflammatory reactions, in addition to, extensive hepatic morphological alterations, marked collagen fibers accumulation as well as overexpression of apoptotic BAX and the noticeable intensified positive nuclear staining for nuclear factor Kabba-b in hepatic tissues. Concurrent NAR supplement to ZnONPs- treated rats significantly declined liver enzymes activities, restored oxidant/antioxidant balance, reversed inflammation, induced fewer collagen fibers accumulation, and antagonized BAX-mediated apoptotic cell death in hepatic tissues. We concluded that concurrent NAR supplement to ZnONPs treated rats improved hepatic function and structure by its antioxidant, anti-inflammatory and antiapoptotic potentials.
Collapse
Affiliation(s)
- Dalia Ibrahim El-Wafaey
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ola Elsayed Nafea
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.,Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Eman Mohamed Faruk
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt.,Department of Anatomy, Faculty of Medicine, Umm Al Qura University, Mecca, Saudi Arabia
| |
Collapse
|
11
|
Kahma H, Aurinsalo L, Neuvonen M, Katajamäki J, Paludetto MN, Viinamäki J, Launiainen T, Filppula AM, Tornio A, Niemi M, Backman JT. An automated cocktail method for in vitro assessment of direct and time-dependent inhibition of nine major cytochrome P450 enzymes - application to establishing CYP2C8 inhibitor selectivity. Eur J Pharm Sci 2021; 162:105810. [PMID: 33753217 DOI: 10.1016/j.ejps.2021.105810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
We developed an in vitro high-throughput cocktail assay with nine major drug-metabolizing CYP enzymes, optimized for screening of time-dependent inhibition. The method was applied to determine the selectivity of the time-dependent CYP2C8 inhibitors gemfibrozil 1-O-β-glucuronide and clopidogrel acyl-β-D-glucuronide. In vitro incubations with CYP selective probe substrates and pooled human liver microsomes were conducted in 96-well plates with automated liquid handler techniques and metabolite concentrations were measured with quantitative UHPLC-MS/MS analysis. After determination of inter-substrate interactions and Km values for each reaction, probe substrates were divided into cocktails I (tacrine/CYP1A2, bupropion/CYP2B6, amodiaquine/CYP2C8, tolbutamide/CYP2C9 and midazolam/CYP3A4/5) and II (coumarin/CYP2A6, S-mephenytoin/CYP2C19, dextromethorphan/CYP2D6 and astemizole/CYP2J2). Time-dependent inhibitors (furafylline/CYP1A2, selegiline/CYP2A6, clopidogrel/CYP2B6, gemfibrozil 1-O-β-glucuronide/CYP2C8, tienilic acid/CYP2C9, ticlopidine/CYP2C19, paroxetine/CYP2D6 and ritonavir/CYP3A) and direct inhibitor (terfenadine/CYP2J2) showed similar inhibition with single substrate and cocktail methods. Established time-dependent inhibitors caused IC50 fold shifts ranging from 2.2 to 30 with the cocktail method. Under time-dependent inhibition conditions, gemfibrozil 1-O-β-glucuronide was a strong (>90% inhibition) and selective (<< 20% inhibition of other CYPs) inhibitor of CYP2C8 at concentrations ranging from 60 to 300 μM, while the selectivity of clopidogrel acyl-β-D-glucuronide was limited at concentrations above its IC80 for CYP2C8. The time-dependent IC50 values of these glucuronides for CYP2C8 were 8.1 and 38 µM, respectively. In conclusion, a reliable cocktail method including the nine most important drug-metabolizing CYP enzymes was developed, optimized and validated for detecting time-dependent inhibition. Moreover, gemfibrozil 1-O-β-glucuronide was established as a selective inhibitor of CYP2C8 for use as a diagnostic inhibitor in in vitro studies.
Collapse
Affiliation(s)
- Helinä Kahma
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Laura Aurinsalo
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jani Katajamäki
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Marie-Noëlle Paludetto
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jenni Viinamäki
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Terhi Launiainen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Anne M Filppula
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
12
|
Enantioselective inhibition of human CYP2C19 by the chiral pesticide ethofumesate: Prediction of pesticide-drug interactions in humans. Chem Biol Interact 2021; 345:109552. [PMID: 34147487 DOI: 10.1016/j.cbi.2021.109552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/20/2021] [Accepted: 06/08/2021] [Indexed: 11/20/2022]
Abstract
Ethofumesate is a chiral herbicide that may display enantioselective behavior in humans. For this reason, the enantioselective potential of ethofumesate and its main metabolite ethofumesate-2-hydroxy to cause pesticide-drug interactions on cytochrome P450 forms (CYPs) has been evaluated by using human liver microsomes. Among the evaluated CYPs, CYP2C19 had its activity decreased by the ethofumesate racemic mixture (rac-ETO), (+)-ethofumesate ((+)-ETO), and (-)-ethofumesate ((-)-ETO). CYP2C19 inhibition was not time-dependent, but a strong inhibition potential was observed for rac-ETO (IC50 = 5 ± 1 μmol L-1), (+)-ETO (IC50 = 1.6 ± 0.4 μmol L-1), and (-)-ETO (IC50 = 1.8 ± 0.4 μmol L-1). The reversible inhibition mechanism was competitive, and the inhibition constant (Ki) values for rac-ETO (2.6 ± 0.4 μmol L-1), (+)-ETO (1.5 ± 0.2 μmol L-1), and (-)-ETO (0.7 ± 0.1 μmol L-1) were comparable to the Ki values of strong CYP2C19 inhibitors. Inhibition of CYP2C19 by ethofumesate was enantioselective, being almost twice higher for (-)-ETO than for (+)-ETO, which indicates that this enantiomer may be a more potent inhibitor of this CYP form. For an in vitro-in vivo correlation, the Food and Drug Administration's (FDA) guideline on the assessment of drug-drug interactions used in the early stages of drug development was used. The FDA's R1 values were estimated on the basis of the obtained ethofumesate Ki and distribution volume, metabolism, unbound plasma fraction, gastrointestinal and dermal absorption data available in the literature. The correlation revealed that ethofumesate probably inhibits CYP2C19 in vivo for both chronic (oral) and occupational (dermal) exposure scenarios.
Collapse
|
13
|
Shibata Y, Tamemoto Y, Singh SP, Yoshitomo A, Hozuki S, Sato H, Hisaka A. Plausible drug interaction between cyclophosphamide and voriconazole via inhibition of CYP2B6. Drug Metab Pharmacokinet 2021; 39:100396. [PMID: 33992954 DOI: 10.1016/j.dmpk.2021.100396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/30/2021] [Accepted: 04/06/2021] [Indexed: 02/07/2023]
Abstract
The inhibitory activities of eight cytochrome P450 (CYP) isoenzymes for representative or suspected inhibitors of CYPs, including pesticides, were evaluated simultaneously using an in vitro cocktail incubation method to demonstrate the importance of systematic evaluation of CYP inhibitory risks in drug interaction (DI). Potent inhibition of CYP2B6 was noticeable for some azoles, including voriconazole. When voriconazole and cyclophosphamide were co-administered in mice, cyclophosphamide-induced alopecia and leukopenia were significantly suppressed by approximately 50% with increased blood concentrations of cyclophosphamide. The formation of an active metabolite of cyclophosphamide was suppressed effectively by voriconazole in the mouse liver microsomes. Surveys of adverse event reporting databases in Japan (JADER) and the U.S. (FAERS) showed that the proportional reporting ratios of neutropenia, hemorrhagic cystitis, and alopecia for cyclophosphamide, which is principally activated by CYP2B6 in humans, were mostly reduced, or tended to be reduced when azoles, including voriconazole, were prescribed in combination. It is highly likely that DIs between cyclophosphamide and azoles occur in the clinical setting. This study also suggests that more proper consideration of CYP2B6-mediated DIs is warranted. The combination of the in vitro cocktail method and a survey of adverse event reporting databases was a useful method to comprehensively detect pharmacokinetic DIs.
Collapse
Affiliation(s)
- Yukihiro Shibata
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan.
| | - Yuta Tamemoto
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan.
| | - Sheelendra Pratap Singh
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan; CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India.
| | - Aoi Yoshitomo
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan.
| | - Shizuka Hozuki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan.
| | - Hiromi Sato
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan.
| | - Akihiro Hisaka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan.
| |
Collapse
|
14
|
Wang J, Chen F, Jiang H, Xu J, Meng D, Geng P, Dai D, Hu J, Zhou Y, Zhou Q, Wang S. Inhibition and Induction by Poziotinib of Different Rat Cytochrome P450 Enzymes In Vivo and in an In Vitro Cocktail Method. Front Pharmacol 2021; 11:593518. [PMID: 33746741 PMCID: PMC7970697 DOI: 10.3389/fphar.2020.593518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/14/2020] [Indexed: 01/10/2023] Open
Abstract
Poziotinib is an orally active, irreversible, pan-HER tyrosine kinase inhibitor used to treat non-small cell lung cancer, breast cancer, and gastric cancer. Poziotinib is currently under clinical investigation, and understanding its drug-drug interactions is extremely important for its future development and clinical application. The cocktail method is most suitable for evaluating the activity of cytochrome P450 enzymes (CYPs). As poziotinib is partially metabolized by CYPs, cocktail probes are used to study the interaction between drugs metabolized by each CYP subtype. Midazolam, bupropion, dextromethorphan, tolbutamide, chlorzoxazone, phenacetin, and their metabolites were used to examine the effects of poziotinib on the activity of cyp1a2, 2b1, 2d1, 2c11, 2e1, and 3a1/2, respectively. The in vitro experiment was carried out by using rat liver microsomes (RLMs), whereas the in vivo experiment involved the comparison of the pharmacokinetic parameters of the probes after co-administration with poziotinib to rats to those of control rats treated with only probes. UPLC-MS/MS was used to detect the probes and their metabolites in rat plasma and rat liver microsomes. The in vitro results revealed that the half-maximal inhibitory concentration values of bupropion and tolbutamide in RLMs were 8.79 and 20.17 μM, respectively, indicating that poziotinib showed varying degrees of inhibition toward cyp2b1 and cyp2c11. Poziotinib was a competitive inhibitor of cyp2b1 and cyp2c11, with Ki values of 16.18 and 17.66 μM, respectively. No time- or concentration-dependence of inhibition by poziotinib was observed toward cyp2b1 and cyp2c11 in RLMs. Additionally, no obvious inhibitory effects were observed on the activity of cyp1a2, cyp2d1, cyp2e1, and cyp3a1/2. In vivo analysis revealed that bupropion, tolbutamide, phenacetin, and chlorzoxazone showed significantly different pharmacokinetic parameters after administration (p < 0.05); there was no significant difference in the pharmacokinetic parameters of dextromethorphan and midazolam. These results show that poziotinib inhibited cyp2b1 and cyp2c11, but induced cyp1a2 and cyp2e1 in rats. Thus, poziotinib inhibited cyp2b1 and cyp2c11 activity in rats, suggesting the possibility of interactions between poziotinib and these CYP substrates and the need for caution when combining them in clinical settings.
Collapse
Affiliation(s)
- Jinhui Wang
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| | - Feifei Chen
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, China
| | - Hui Jiang
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, China
| | - Jia Xu
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, China
| | - Deru Meng
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, China
| | - Peiwu Geng
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, China
| | - Dapeng Dai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingbo Hu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| | - Yunfang Zhou
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, China
| | - Quan Zhou
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, China
| | - Shuanghu Wang
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, China
| |
Collapse
|
15
|
Sodhi JK, Halladay JS. Case Study 9: Probe-Dependent Binding Explains Lack of CYP2C9 Inactivation by 1-Aminobenzotriazole (ABT). Methods Mol Biol 2021; 2342:765-779. [PMID: 34272716 DOI: 10.1007/978-1-0716-1554-6_28] [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] [Indexed: 06/13/2023]
Abstract
The potential for new chemical entities to inhibit the major cytochrome P450 (CYP) isoforms is routinely evaluated to minimize the risk of developing drugs with drug-drug interaction liabilities. CYP inhibition assays are routinely performed in a high-throughput format to efficiently screen large numbers of compounds. In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). Mechanistic reversible and time-dependent inhibition experiments revealed that the ineffective CYP2C9 inactivation by ABT was also probe-dependent, with utilization of (S)-warfarin as the probe substrate resulting in more potent CYP2C9 inhibition by ABT compared to diclofenac. Addition of (S)-warfarin to the reversible and time-dependent inhibition experiments between ABT and diclofenac resulted in an attenuation of the inhibitory effects of ABT on CYP2C9-mediated diclofenac metabolism. Molecular docking studies further confirmed that (S)-warfarin and diclofenac preferentially bind in different regions of the CYP2C9 active site, with (S)-warfarin occupying a distal "warfarin-binding pocket" and diclofenac occupying a binding site close to the active heme moiety. ABT preferentially binds in the distal warfarin-binding pocket, supporting that diclofenac is minimally deterred from access to the CYP2C9 active site in the presence of ABT, thus resulting in minimal inactivation. Simultaneously docking of (S)-warfarin and ABT revealed that (S)-warfarin outcompetes ABT for the distal binding site and results in the binding of ABT to the CYP2C9 active site, supporting the observations of potent inactivation of CYP2C9 when (S)-warfarin is the probe substrate.These results highlight that probe selection is crucial when evaluating CYP inhibition potential, and it is recommended that multiple probes be utilized for CYP2C9, similar to the approach routinely employed for CYP3A4. Further, utilization of ABT as a pan-inhibitor of CYP activity for investigational compounds, both in vitro and in vivo, should be accompanied with the understanding that residual CYP-mediated oxidative metabolism could potentially be observed for CYP2C9 substrates and should not necessarily be attributed to non-P450-mediated metabolism.
Collapse
Affiliation(s)
- Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, CA, USA.
| | | |
Collapse
|
16
|
Dangi B, Davydova NY, Vavilov NE, Zgoda VG, Davydov DR. Nonadditivity in human microsomal drug metabolism revealed in a study with coumarin 152, a polyspecific cytochrome P450 substrate. Xenobiotica 2020; 50:1393-1405. [PMID: 32662751 PMCID: PMC7740640 DOI: 10.1080/00498254.2020.1775913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022]
Abstract
We closely characterized 7-Dimethylamino-4-trifluromethylcoumarin (Coumarin 152, C152), a substrate metabolized by multiple P450 species, to establish a new fluorogenic probe for the studies of functional integration in the cytochrome P450 ensemble. Scanning fluorescence spectroscopy and LC/MS-MS were used to characterize the products of N-demethylation of C152 and optimize their fluorometric detection. The metabolism of C152 by the individual P450 species was characterized using the microsomes containing cDNA-expressed enzymes. C152 metabolism in human liver microsomes (HLM) was studied in a preparation with quantified content of eleven P450 species. C152 is metabolized by CYP2B6, CYP3A4, CYP3A5, CYP2C19, CYP1A2, CYP2C9, and CYP2C8 listed in the order of decreasing turnover. The affinities exhibited by CYP3A5, CYP2C9, and CYP2C8 were lower than those characteristic to the other enzymes. The presumption of additivity suggests the participation of CYP3A4, CYP2B6, and CYP2C19 to be 84, 8, and 0.2%, respectively. Contrary to this prediction, inhibitory analysis identified CYP2C19 as the principal C152-metabolizing enzyme. We thoroughly characterize C152 for the studies of drug metabolism in HLM and demonstrate the limitations of the proportional projection approach by providing an example, where the involvement of individual P450 species cannot be predicted from their content.
Collapse
Affiliation(s)
- Bikash Dangi
- Department of Chemistry, Washington State University, Pullman, WA, 99164
| | | | | | - Victor G. Zgoda
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
- Skolkovo Institute of Science and Technology, 143025 Skolkovo, Moscow region, Russia
| | - Dmitri R. Davydov
- Department of Chemistry, Washington State University, Pullman, WA, 99164
| |
Collapse
|
17
|
de Albuquerque NCP, Carrão DB, Habenschus MD, Fonseca FS, Moreira da Silva R, Lopes NP, Rocha BA, Barbosa Júnior F, de Oliveira ARM. Risk assessment of the chiral pesticide fenamiphos in a human model: Cytochrome P450 phenotyping and inhibition studies. Food Chem Toxicol 2020; 146:111826. [PMID: 33127494 DOI: 10.1016/j.fct.2020.111826] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 12/23/2022]
Abstract
Fenamiphos (FS) is a chiral organophosphate pesticide that is used to control nematodes in several crops. Enantioselective differences may be observed in FS activity, bioaccumulation, metabolism, and toxicity. Humans may be exposed to FS through occupational and chronic (food, water, and environmental) exposure. FS may cause undesirable CYP450 pesticide-drug interactions, which may impact human health. Here, the CYP450 isoforms involved in enantioselective FS metabolism were identified, and CYP450 inhibition by rac-FS, (+)-FS, and (-)-FS was evaluated to obtain reliable information on enantioselective FS risk assessment in humans. CYP3A4 and CYP2E1 metabolized FS enantiomers, and CYP2B6 may participate in rac-FS metabolism. In addition, rac-FS, (+)-FS, and (-)-FS were reversible competitive CYP1A2, CYP2C19, and CYP3A4/5 inhibitors. High stereoselective inhibition potential was verified; rac-FS and (-)-FS strongly inhibited and (+)-FS moderately inhibited CYP1A2. Stereoselective differences were also detected for CYP2C19 and CYP3A4/5, which were strongly inhibited by rac-FS, (+)-FS, and (-)-FS. Our results indicated a high potential for CYP450 drug-pesticide interactions, which may affect human health. The lack of stereoselective research on the effect of chiral pesticides on the activity of CYP450 isoforms highlights the importance of assessing the risks of such pesticides in humans.
Collapse
Affiliation(s)
- Nayara Cristina Perez de Albuquerque
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Daniel Blascke Carrão
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Maísa Daniela Habenschus
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Franciele Saraiva Fonseca
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Rodrigo Moreira da Silva
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14090-903, Ribeirão Preto, SP, Brazil
| | - Norberto Peporine Lopes
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14090-903, Ribeirão Preto, SP, Brazil
| | - Bruno Alves Rocha
- Departamento de Química, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, 09972-270, Campus Diadema, SP, Brazil
| | - Fernando Barbosa Júnior
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14040-903, Ribeirão Preto, SP, Brazil
| | - Anderson Rodrigo Moraes de Oliveira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil; National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Unesp, Institute of Chemistry, P.O. Box 355, 14800-900, Araraquara, SP, Brazil.
| |
Collapse
|
18
|
Evaluation of acacetin inhibition potential against cytochrome P450 in vitro and in vivo. Chem Biol Interact 2020; 329:109147. [PMID: 32738202 DOI: 10.1016/j.cbi.2020.109147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/16/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023]
Abstract
Acacetin is a natural flavonoid that is widely distributed in plants and possesses numerous pharmacological activities. The aim of the present study was to investigate the effects of acacetin on the activities of the cytochrome P450 family members CYP1A2, CYP2B1, CYP2C11, CYP2D1, CYP2E1, and CYP3A2 in rat liver microsomes in vitro and rats in vivo to evaluate potential herb-drug interactions by using a cocktail approach. Phenacetin, bupropion, tolbutamide, dextromethorphan, chlorzoxazone, and midazolam were chosen as the probe substrates. An ultra-performance liquid chromatography-tandem mass spectrometry method was developed for the simultaneous detection of the probe substrates and their metabolites. In vitro, the mode of acacetin inhibition of CYP2B1, CYP2C11, and CYP2E1 was competitive, while mixed inhibition was observed for CYP1A2 and CYP3A2. The Ki values in this study were less than 8.32 μM. In vivo, the mixed probe substrates were administered by gavage after daily intraperitoneal injection with 50 mg/kg acacetin or saline for 2 weeks. The main pharmacokinetic parameters, area under the plasma concentration-time curve (AUC), plasma clearance (CL), and maximum plasma concentration (Cmax) of the probe substrates were significantly different in the experimental group than in the control group. Overall, the in vitro and in vivo results indicated that acacetin would be at high risk to cause toxicity and drug interactions via cytochrome P450 inhibition.
Collapse
|
19
|
Jang KJ, Otieno MA, Ronxhi J, Lim HK, Ewart L, Kodella KR, Petropolis DB, Kulkarni G, Rubins JE, Conegliano D, Nawroth J, Simic D, Lam W, Singer M, Barale E, Singh B, Sonee M, Streeter AJ, Manthey C, Jones B, Srivastava A, Andersson LC, Williams D, Park H, Barrile R, Sliz J, Herland A, Haney S, Karalis K, Ingber DE, Hamilton GA. Reproducing human and cross-species drug toxicities using a Liver-Chip. Sci Transl Med 2020; 11:11/517/eaax5516. [PMID: 31694927 DOI: 10.1126/scitranslmed.aax5516] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
Nonclinical rodent and nonrodent toxicity models used to support clinical trials of candidate drugs may produce discordant results or fail to predict complications in humans, contributing to drug failures in the clinic. Here, we applied microengineered Organs-on-Chips technology to design a rat, dog, and human Liver-Chip containing species-specific primary hepatocytes interfaced with liver sinusoidal endothelial cells, with or without Kupffer cells and hepatic stellate cells, cultured under physiological fluid flow. The Liver-Chip detected diverse phenotypes of liver toxicity, including hepatocellular injury, steatosis, cholestasis, and fibrosis, and species-specific toxicities when treated with tool compounds. A multispecies Liver-Chip may provide a useful platform for prediction of liver toxicity and inform human relevance of liver toxicities detected in animal studies to better determine safety and human risk.
Collapse
Affiliation(s)
| | - Monicah A Otieno
- Janssen Pharmaceutical Research and Development, Nonclinical Safety, 1400 Welsh and McKean Road, Spring House, PA 19477, USA.
| | - Janey Ronxhi
- Emulate Inc., 27 Drydock Avenue, Boston, MA 02210, USA
| | - Heng-Keang Lim
- Janssen Pharmaceutical Research and Development, Drug Metabolism and Pharmacokinetics, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Lorna Ewart
- Clinical Pharmacology and Safety Sciences Department, Biopharmaceuticals Science Unit, AstraZeneca, Cambridge, CB4 0WG, UK
| | | | | | | | | | | | - Janna Nawroth
- Emulate Inc., 27 Drydock Avenue, Boston, MA 02210, USA
| | - Damir Simic
- Janssen Pharmaceutical Research and Development, Nonclinical Safety, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Wing Lam
- Janssen Pharmaceutical Research and Development, Drug Metabolism and Pharmacokinetics, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Monica Singer
- Janssen Pharmaceutical Research and Development, Nonclinical Safety, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Erio Barale
- Janssen Pharmaceutical Research and Development, Nonclinical Safety, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Bhanu Singh
- Janssen Pharmaceutical Research and Development, Nonclinical Safety, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Manisha Sonee
- Janssen Pharmaceutical Research and Development, Nonclinical Safety, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Anthony J Streeter
- Janssen Pharmaceutical Research and Development, Nonclinical Safety, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Carl Manthey
- Janssen Pharmaceutical Research and Development, IPD Biology, 1400 Welsh and McKean Road, Spring House, PA 19477, USA
| | - Barry Jones
- Clinical Pharmacology and Safety Sciences Department, Biopharmaceuticals Science Unit, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Abhishek Srivastava
- Clinical Pharmacology and Safety Sciences Department, Biopharmaceuticals Science Unit, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Linda C Andersson
- Clinical Pharmacology and Safety Sciences Department, Biopharmaceuticals Science Unit, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Dominic Williams
- Clinical Pharmacology and Safety Sciences Department, Biopharmaceuticals Science Unit, AstraZeneca, Cambridge, CB4 0WG, UK
| | | | | | - Josiah Sliz
- Emulate Inc., 27 Drydock Avenue, Boston, MA 02210, USA
| | - Anna Herland
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | | | - Katia Karalis
- Emulate Inc., 27 Drydock Avenue, Boston, MA 02210, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA.,Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA 02139, USA
| | | |
Collapse
|
20
|
Large Volume Direct Injection Ultra-High Performance Liquid Chromatography-Tandem Mass Spectrometry-Based Comparative Pharmacokinetic Study between Single and Combinatory Uses of Carthamus tinctorius Extract and Notoginseng Total Saponins. Pharmaceutics 2020; 12:pharmaceutics12020180. [PMID: 32093170 PMCID: PMC7076352 DOI: 10.3390/pharmaceutics12020180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 02/08/2020] [Accepted: 02/17/2020] [Indexed: 11/17/2022] Open
Abstract
The combination of Carthamus tinctorius extract (CTE) and notoginseng total saponins (NGTS), namely, CNP, presents a synergistic effect on myocardial ischemia protection. Herein, comparative pharmacokinetic studies between CNP and CTE/NGTS were conducted to clarify their synergistic mechanisms. A large volume direct injection ultra-high performance liquid chromatography-tandem mass spectrometry (LVDI-UHPLC-MS/MS) platform was developed for sensitively assaying the multi-component pharmacokinetic and in vitro cocktail assay of cytochrome p450 (CYP450) before and after compatibility of CTE and NGTS. The pharmacokinetic profiles of six predominantly efficacious components of CNP, including hydroxysafflor yellow A (HSYA); ginsenosides Rg1 (GRg1), Re (GRe), Rb1 (GRb1), and Rd (GRd); and notoginsenoside R1 (NGR1), were obtained, and the results disclosed that CNP could increase the exposure levels of HSYA, GRg1, GRe, GRb1, and NGR1 at varying degrees. The in vitro cocktail assay demonstrated that CNP exhibited more potent inhibition on CYP1A2 than CTE and NGTS, and GRg1, GRb1, GRd, quercetin, kaempferol, and 6-hydroxykaempferol were found to be the major inhibitory compounds. The developed pharmacokinetic interaction-based strategy provides a viable orientation for the compatibility investigation of herb medicines.
Collapse
|
21
|
Pharmacogenomics of poor drug metabolism in Greyhounds: Cytochrome P450 (CYP) 2B11 genetic variation, breed distribution, and functional characterization. Sci Rep 2020; 10:69. [PMID: 31919457 PMCID: PMC6952448 DOI: 10.1038/s41598-019-56660-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/13/2019] [Indexed: 01/11/2023] Open
Abstract
Greyhounds recover more slowly from certain injectable anesthetics than other dog breeds. Previous studies implicate cytochrome P450 (CYP) 2B11 as an important clearance mechanism for these drugs and suggest Greyhounds are deficient in CYP2B11. However, no CYP2B11 gene mutations have been identified that explain this deficiency in Greyhounds. The objectives of this study were to provide additional evidence for CYP2B11 deficiency in Greyhounds, determine the mechanisms underlying this deficiency, and identify CYP2B11 mutations that contribute to this phenotype in Greyhounds. Greyhound livers metabolized CYP2B11 substrates slower, possessed lower CYP2B11 protein abundance, but had similar or higher mRNA expression than other breeds. Gene resequencing identified three CYP2B11 haplotypes, H1 (reference), H2, and H3 that were differentiated by mutations in the gene 3'-untranslated region (3'-UTR). Compared with 63 other dog breeds, Greyhounds had the highest CYP2B11-H3 allele frequency, while CYP2B11-H2 was widely distributed across most breeds. Using 3'-UTR luciferase reporter constructs, CYP2B11-H3 showed markedly lower gene expression (over 70%) compared to CYP2B11-H1 while CYP2B11-H2 expression was intermediate. Truncated mRNA transcripts were observed in CYP2B11-H2 and CYP2B11-H3 but not CYP2B11-H1 transfected cells. Our results implicate CYP2B11 3'-UTR mutations as a cause of decreased CYP2B11 enzyme expression in Greyhounds through reduced translational efficiency.
Collapse
|
22
|
Krogstad V, Peric A, Robertsen I, Kringen MK, Wegler C, Angeles PC, Hjelmesæth J, Karlsson C, Andersson S, Artursson P, Åsberg A, Andersson TB, Christensen H. A Comparative Analysis of Cytochrome P450 Activities in Paired Liver and Small Intestinal Samples from Patients with Obesity. Drug Metab Dispos 2019; 48:8-17. [DOI: 10.1124/dmd.119.087940] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022] Open
|
23
|
Excretion, Metabolism and Cytochrome P450 Inhibition of Methyl 3,4-Dihydroxybenzoate (MDHB): A Potential Candidate to Treat Neurodegenerative Diseases. Eur J Drug Metab Pharmacokinet 2019; 45:51-69. [PMID: 31602595 DOI: 10.1007/s13318-019-00576-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND OBJECTIVES Methyl 3,4-dihydroxybenzoate (MDHB) has the potential to prevent neurodegenerative diseases (NDDs). The present work investigated its excretion, metabolism, and cytochrome P450-based drug-drug interactions (DDIs). METHODS After intragastric administration of MDHB, the parent drug was assayed in the urine and faeces of mice. Metabolites of MDHB in the urine, faeces, brain, plasma and liver were detected by liquid chromatography-hybrid quadrupole time-of-flight mass spectrometry (LC-QTOF/MS). A cocktail approach was used to evaluate the inhibition of cytochrome P450 isoforms by MDHB. RESULTS The cumulative excretion permille of MDHB in the urine and faeces were found to be 0.67 ± 0.31 and 0.49 ± 0.44‰, respectively. A total of 96 metabolites of MDHB were identified, and all IC50 (half-maximal inhibitory concentration) values of MDHB towards cytochrome P450 isoforms were > 100 μM. CONCLUSIONS The results suggest that MDHB has a low parent drug cumulative excretion percentage and that MDHB has multiple metabolites and is mainly metabolized through the loss of -CH2 and -CO2, the loss of -CH2O, ester bond hydrolysis, the loss of -O and -CO2, isomerization, methylation, sulfate conjugation, the loss of -CH2O and -O and glycine conjugation, glycine conjugation, the loss of two -O groups and alanine conjugation, the loss of -CH2O and -O and glucose conjugation, glucuronidation, glucose conjugation, etc., in vivo. Finally, MDHB has a low probability of cytochrome P450-based DDIs.
Collapse
|
24
|
Yang Q, Sun J, Li C, Zhang H, Xu W, Liu C, Zheng X. Comparative research on the metabolism of metoprolol by four CYP2D6 allelic variants in vitro with LC-MS/MS. J Pharm Biomed Anal 2019; 174:479-485. [PMID: 31228851 DOI: 10.1016/j.jpba.2019.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/28/2019] [Accepted: 06/10/2019] [Indexed: 11/18/2022]
Abstract
Specific study about the effect of cytochrome P450 2D6 (CYP2D6) polymorphisms on the metabolism of clinic drugs is of great significance for drug safety investigation. Here, the interaction between CYP2D6 variants (*1, *2, *10, *39) and metoprolol (MET) was intensively researched in vitro from the aspect of drug-enzyme kinetic study. To obtain quantitative data, α-hydroxymetoprolol (main metabolite of MET) was selected as an ideal analyte and an LC-MS/MS method was adopted for sample determination. Firstly, by selecting suitable internal standard and optimizing separation condition, the LC-MS/MS method was established and validated. Then, the drug-enzyme incubation system was optimized by two parameters: incubation time and amount of enzyme. Lastly, the interaction between CYP2D6 allelic variants and MET was characterized by Km, Vmax and CLint. As a result, four CYP2D6 enzymes displayed diverse Km or Vmax towards MET and the values of CLint showed a wide range from 8.91 to 100%. Relative to CYP2D6*1 (CLint*1 = 100%), CYP2D6*2 demonstrated the second high catalytic activity (CLint*2/*1 = 74.87%) while CYP2D6*39 (CLint*39/*1 = 29.65%) and CYP2D6*10 (CLint*10/*1 = 8.91%) showed minimal catalytic activity. This comprehensive in vitro data suggested the prominent influence of CYP2D6 polymorphisms on the metabolism of MET, which could offer valuable information for personalized administration of MET in clinic.
Collapse
Affiliation(s)
- Qingqing Yang
- Department of Pharmacology, Tianjin Medical University, Tianjin, 300070, China
| | - Jing Sun
- Department of Pharmacology, Tianjin Medical University, Tianjin, 300070, China
| | - Chuan Li
- Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193, China
| | - Haizhi Zhang
- Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193, China.
| | - Weiren Xu
- Department of Pharmacology, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193, China.
| | - Changxiao Liu
- Tianjin Center for New Drug Evaluation and Research, State Key Laboratory of Drug Delivery Technology and Pharmaceutics, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193, China
| | - Xuemin Zheng
- Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193, China
| |
Collapse
|
25
|
Li Q, Sun M, Li G, Qiu L, Huang Z, Gong J, Huang J, Li G, Si L. The sub-chronic impact of mPEG2k-PCLx polymeric nanocarriers on cytochrome P450 enzymes after intravenous administration in rats. Eur J Pharm Biopharm 2019; 142:101-113. [DOI: 10.1016/j.ejpb.2019.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/09/2019] [Accepted: 06/17/2019] [Indexed: 01/21/2023]
|
26
|
Long N, Tang H, Lin L, Li J, Guo L, Sun F, Dai M. Effects of infection of MRSA on the expression and activity of renal cytochrome P450s in mice. J Toxicol Sci 2019; 44:299-307. [PMID: 30944282 DOI: 10.2131/jts.44.299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) leads to serious infections, but it is not known whether it changes the expression of kidney drug metabolizing enzymes during infection. The mice were infected with different doses of MRSA and the oxidative stress and inflammation levels in the kidney were examined. The mRNA expression and activity of cytochrome P450 enzyme was analysed. Mice infected with high levels of MRSA showed a decrease in renal antioxidant capability and an elevated level of oxidative metabolites, which was accompanied by the release of inflammatory cytokines. The levels of interleukin 1β, tumour necrosis factor alpha, and macrophage inflammatory protein-1α were significantly increased along with the levels of nitric oxide and malondialdehyde. On day 7, mRNA expression of Cyp1a2, 2d22, and 3a11 were decreased by the high level of MRSA, but the low level of MRSA increased their expressions. Cyp2e1 mRNA expression was increased by MRSA in the kidney of mice. High dose of MRSA infection increased the oxidative stress and inflammatory response in mouse kidney, leading to the decrease in the expression of renal drug-metabolizing enzymes and no recovery within 7 days.
Collapse
Affiliation(s)
- Nana Long
- School of Laboratory Medicine, Chengdu Medical College, China.,Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, China
| | - Huaqiao Tang
- School of Laboratory Medicine, Chengdu Medical College, China.,Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, China
| | - Lin Lin
- School of Laboratory Medicine, Chengdu Medical College, China.,Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, China
| | - Jianlong Li
- School of Laboratory Medicine, Chengdu Medical College, China.,Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, China
| | - Lijuan Guo
- School of Laboratory Medicine, Chengdu Medical College, China.,Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, China
| | - Fenghui Sun
- School of Laboratory Medicine, Chengdu Medical College, China.,Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, China
| | - Min Dai
- School of Laboratory Medicine, Chengdu Medical College, China.,Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, China
| |
Collapse
|
27
|
Carrão DB, Habenchus MD, de Albuquerque NCP, da Silva RM, Lopes NP, de Oliveira ARM. In vitro inhibition of human CYP2D6 by the chiral pesticide fipronil and its metabolite fipronil sulfone: Prediction of pesticide-drug interactions. Toxicol Lett 2019; 313:196-204. [PMID: 31278966 DOI: 10.1016/j.toxlet.2019.07.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022]
Abstract
Fipronil is a chiral insecticide employed worldwide in crops, control of public hygiene and control of veterinary pests. Humans can be exposed to fipronil through occupational, food, and environmental contamination. Therefore, the risk assessment of fipronil in humans is important to protect human health. Fipronil sulfone is the major metabolite formed during fipronil metabolism by humans. Since the CYP450 enzymes are the main ones involved in drug metabolism, the evaluation of their inhibition by fipronil and its main metabolite is important to predict drug-pesticide interactions. The aim of this work was to investigate the inhibition effects of rac-fipronil, S-fipronil, R-fipronil and fipronil sulfone on the main human CYP450 isoforms. The results showed that CYP2D6 is the only CYP450 isoform inhibited by these xenobiotics. In addition, no enantioselective differences were observed in the inhibition of CYP450 isoforms by fipronil and its individuals' enantiomers. Rac-fipronil, S-fipronil and R-fipronil are moderate CYP2D6 inhibitors showing a competitive inhibition profile. On the other hand, the metabolite fipronil sulfone showed to be a strong inhibitor of CYP2D6 also by competitive inhibition. These results highlight the importance of metabolite evaluation on pesticide safety since the metabolism of fipronil into fipronil sulfone increases the risk of pesticide-drug interactions for drugs metabolized by CYP2D6.
Collapse
Affiliation(s)
- Daniel Blascke Carrão
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Maísa Daniela Habenchus
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Nayara Cristina Perez de Albuquerque
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Rodrigo Moreira da Silva
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14090-903, Ribeirão Preto, SP, Brazil
| | - Norberto Peporine Lopes
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14090-903, Ribeirão Preto, SP, Brazil
| | - Anderson Rodrigo Moraes de Oliveira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil; National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Unesp, Institute of Chemistry, P.O. Box 355, 14800-900, Araraquara, SP, Brazil.
| |
Collapse
|
28
|
Ji S, He DD, Su ZY, Du Y, Wang YJ, Gao SK, Guo MZ, Tang DQ. P450 enzymes-based metabolic interactions between monarch drugs and the other constituent herbs: A strategy to explore compatibility mechanism of Sangju-Yin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 58:152866. [PMID: 30831464 DOI: 10.1016/j.phymed.2019.152866] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/11/2019] [Accepted: 02/17/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Herbal compatibility of compound formulas can enhance therapeutic effects or reduce side effects of the monarch drugs, but majority of compatibility mechanisms are still unknown. Sangju-Yin, a well-known Chinese compound formula, is currently used to treat common cold in clinical. PURPOSE In this study, we proposed a strategy to explore the compatibility mechanism of Sangju-Yin by investigating P450 enzymes-based metabolic interactions between monarch drugs and the other constituent herbs. METHODS Under the guidance of traditional Chinese medicine theory, the constituent herbs of Sangju-Yin were divided into four groups, including monarch drugs, monarch drugs with addition of minister drugs, monarch drugs with addition of minister and adjuvant drugs, as well as the whole recipe, namely monarch drugs with addition of minister, adjuvant and conductant drugs. Their effects on rats in vivo P450 (CYP1A2, CYP2A3, CYP2C6, CYP2C11 and CYP3A1) activities after oral administration were evaluated using probe drug assay based on LC-MS/MS. Moreover, effects of the four groups of herbs on mRNA expression of P450 enzymes after oral administration, as well as in vitro P450 activities after co-incubation, were investigated to explore the underlying mechanisms. RESULTS Comparing with monarch drugs, addition of different constituent herbs significantly enhanced CYP1A2 and CYP2C6 activities, and inhibited CYP2A3 and CYP3A1 activities, indicating their possible influences on plasma concentrations of active constituents in the monarch drugs. Mechanism study suggested that these herbs affected P450 activities by transcriptional regulation and/or direct interaction with the enzymes. CONCLUSION This study clarified the compatibility mechanism of Sangju-Yin from the aspect of P450 enzymes-based metabolic interactions, which would benefit better understanding of the therapeutic basis of Sangju-Yin.
Collapse
Affiliation(s)
- Shuai Ji
- Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou 221004, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Dan-Dan He
- Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou 221004, China
| | - Zhen-Yu Su
- Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou 221004, China
| | - Yan Du
- Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou 221004, China
| | - Yu-Jie Wang
- Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou 221004, China
| | - Shi-Kai Gao
- Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou 221004, China
| | - Meng-Zhe Guo
- Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou 221004, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Dao-Quan Tang
- Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou 221004, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
29
|
Kim HJ, Lee H, Ji HK, Lee T, Liu KH. Screening of ten cytochrome P450 enzyme activities with 12 probe substrates in human liver microsomes using cocktail incubation and liquid chromatography-tandem mass spectrometry. Biopharm Drug Dispos 2019; 40:101-111. [PMID: 30730576 DOI: 10.1002/bdd.2174] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 11/09/2022]
Abstract
Testing for potential drug interactions of new chemical entities is essential when developing a novel drug. In this study, an assay was designed to evaluate drug interactions with 10 major human cytochrome P450 (P450) enzymes incubated in liver microsomes, involving 12 probe substrates with two cocktail incubation sets used in a single liquid chromatography-tandem mass spectrometry (LC-MS/MS) run. The P450 substrate composition in each cocktail set was optimized to minimize solvent effects and mutual drug interactions among substrates as follows: cocktail A was composed of phenacetin for CYP1A2, bupropion for CYP2B6, amodiaquine for CYP2C8, diclofenac for CYP2C9, S-mephenytoin for CYP2C19, and dextromethorphan for CYP2D6; cocktail B was composed of coumarin for CYP2A6, chlorzoxazone for CYP2E1, astemizole for CYP2J2, and midazolam, nifedipine, and testosterone for CYP3A. Multiple probe substrates were used for CYP3A owing to the multiple substrate-binding sites and substrate-dependent inhibition. After incubation in human liver microsomes, each incubation mixture was pooled and all probe metabolites were simultaneously analysed in a single LC-MS/MS run. Polarity switching was used to acquire the negative-ion mode for hydroxychlorzoxazone and positive-ion mode for the remaining analytes. The method was validated by comparing the inhibition data obtained from incubation of each individual probe substrate alone and with the substrate cocktails. The half-maximal inhibitory concentration values obtained from the cocktail and individual incubations were well correlated and in agreement with previously reported values. This new method will be useful in assessing the drug interaction potential of new chemical entities during new drug development.
Collapse
Affiliation(s)
- Hyun-Ji Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, South Korea.,BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Kyungpook National University, Daegu, 41566, South Korea
| | - Hyunyoung Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, South Korea.,BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Kyungpook National University, Daegu, 41566, South Korea
| | - Hyeon-Kyeong Ji
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, South Korea.,BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Kyungpook National University, Daegu, 41566, South Korea
| | - Taeho Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Kwang-Hyeon Liu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, South Korea.,BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Kyungpook National University, Daegu, 41566, South Korea
| |
Collapse
|
30
|
Cusinato DAC, Filgueira GCDO, Rocha A, Cintra MAC, Lanchote VL, Coelho EB. LC-MS/MS analysis of the plasma concentrations of a cocktail of 5 cytochrome P450 and P-glycoprotein probe substrates and their metabolites using subtherapeutic doses. J Pharm Biomed Anal 2019; 164:430-441. [DOI: 10.1016/j.jpba.2018.10.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/14/2018] [Accepted: 10/17/2018] [Indexed: 11/29/2022]
|
31
|
Tang H, Long N, Dai M, Lin L, Li J, Sun F, Guo L. Effect of citral on mouse hepatic cytochrome P450 enzymes. PHARMACEUTICAL BIOLOGY 2018; 56:337-343. [PMID: 29969356 PMCID: PMC6130695 DOI: 10.1080/13880209.2018.1470191] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
CONTEXT Citral is used as a potential natural treatment for various infectious diseases. OBJECTIVE To examine the effect of citral on the mRNA expression and activities of cytochrome P450 (CYP450) enzymes and establish the relationship between citral-induced liver injury and oxidative stress. MATERIALS AND METHODS ICR mice were randomly divided into citral (20, 200, and 2000 mg/kglow), Tween-80, and control groups (0.9% saline), 10 mice in each group. The citral-treated groups were intragastrically administered citral for 3 d, control groups treated with 0.5% Tween-80 and 0.9% saline in the same way. Liver injury and CYP450 enzymes were analyzed by analyzing the histopathological changes and the changes of related enzymes. RESULTS Citral treatment (2000 mg/kg) for 3 d increased serum glutamic pyruvic transaminase and glutamic oxaloacetic transaminase levels, as well as glutathione, gydroxyl radicals, malonaldehyde and total superoxide dismutase contents, but decreased the content of total antioxidant capacity. In doses of 20 and 200 mg/kg groups mice, the contents of NO were decreased significantly and other changes were similar to the 2000 mg/kg group mice, but the liver damage was most severe in the 2000 mg/kg group. Citral induced the mRNA expression and activities of CYP450 1A2, 2D22, and 2E1 in the liver of mice at doses of 20 and 200 mg/kg. There were no changes in testing indexes in Tween-80 treated group mice. Due to its toxic effects, the CYP induction effect of citral negatively correlated with its dose. Although the mRNA expression of CYP450 3A11 was induced by citral, its activity was not affected by low and moderate doses of citral. CYP450 3A11 activity was significantly decreased by high-dose citral. CONCLUSIONS Citral is hepatotoxic and induced oxidative stress in higher dose, which has a negative effect on CYP450 enzymes. These data suggest caution needs to be taken in order to avoid citral-drug interactions in human beings.
Collapse
Affiliation(s)
- Huaqiao Tang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
- Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food, Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Nana Long
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
- Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food, Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Min Dai
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
- Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food, Chengdu Medical College, Chengdu, Sichuan, PR China
- CONTACT Min Dai School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Lin Lin
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
- Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food, Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Jianlong Li
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
- Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food, Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Fenghui Sun
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
- Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food, Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Lijuan Guo
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China
- Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food, Chengdu Medical College, Chengdu, Sichuan, PR China
| |
Collapse
|
32
|
Badée J, Qiu N, Parrott N, Collier AC, Schmidt S, Fowler S. Optimization of Experimental Conditions of Automated Glucuronidation Assays in Human Liver Microsomes Using a Cocktail Approach and Ultra-High Performance Liquid Chromatography-Tandem Mass Spectrometry. Drug Metab Dispos 2018; 47:124-134. [PMID: 30478159 DOI: 10.1124/dmd.118.084301] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/20/2018] [Indexed: 11/22/2022] Open
Abstract
UDP-glucuronosyltransferase (UGT)-mediated metabolism is possibly the most important conjugation reaction for marketed drugs. However, there are currently no generally accepted standard incubation conditions for UGT microsomal assays, and substantial differences in experimental design and methodology between laboratories hinder cross-study comparison of in vitro activities. This study aimed to define optimal experimental conditions to determine glucuronidation activity of multiple UGT isoforms simultaneously using human liver microsomes. Hepatic glucuronidation activities of UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2B4, UGT2B7, UGT2B10, UGT2B15, and UGT2B17 were determined using cocktail incubations of 10 UGT probe substrates. Buffer components and cosubstrates were assessed over a range of concentrations including magnesium chloride (MgCl2; 0-10 mM) and uridine 5'-diphosphoglucuronic acid (UDPGA; 1-25 mM) with either Tris-HCl or potassium phosphate buffer (100 mM, pH 7.4). Greater microsomal glucuronidation activity by different hepatic UGT isoforms was obtained using 10 mM MgCl2 and 5 mM UDPGA with 100 mM Tris-HCl buffer. The influence of bovine serum albumin (BSA; 0.1%-2% w/v) on glucuronidation activity was also assessed. Enzyme- and substrate-dependent effects of BSA were observed, resulting in decreased total activity of UGT1A1, UGT1A3, and UGT2B17 and increased total UGT1A9 and UGT2B7 activity. The inclusion of BSA did not significantly reduce the between-subject variability of UGT activity. Future in vitro UGT profiling studies under the proposed optimized experimental conditions would allow high-quality positive control data to be generated across laboratories, with effective control of a high degree of between-donor variability for UGT activity and for chemical optimization toward lower-clearance drug molecules in a pharmaceutical drug discovery setting.
Collapse
Affiliation(s)
- Justine Badée
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Nahong Qiu
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Neil Parrott
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Abby C Collier
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Stephen Fowler
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| |
Collapse
|
33
|
Study on the inhibitory effect of furafylline and troleandomycin in the 7-methoxyresorufin-O-demethylase and nifedipine oxidase activities in hepatic microsomes from four poultry species using high-performance liquid chromatography coupled with fluorescence and ultraviolet detection. J Pharm Biomed Anal 2018; 164:148-154. [PMID: 30390556 DOI: 10.1016/j.jpba.2018.10.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 10/28/2022]
Abstract
The present study reports the in vitro studies with furafylline and troleandomycin (TAO) as specific inhibitors of activities 7-methoxyresorufin-O-demethylase (MROD) and nifedipine oxidase, catalyzed by cytochrome P450 1 A2 (CYP1 A2) and 3A4 human enzymes, respectively, in hepatic microsomes of quail, duck, turkey and chicken. The results suggest that in chicken and quail the MROD activity is carried out by orthologs CYP1 A4 and 1 A5, meanwhile in duck and turkey by a CYP1 A5 ortholog. The nifedipine oxidase activity is carried out by orthologs of the CYP3A family in the four bird species. The use of furafylline and TAO significantly decreased these activities (P < 0.05) and suggested that the biotransformation of resorufin methyl ether (RME) may be related to more than one avian ortholog.
Collapse
|
34
|
Esteves F, Campelo D, Urban P, Bozonnet S, Lautier T, Rueff J, Truan G, Kranendonk M. Human cytochrome P450 expression in bacteria: Whole-cell high-throughput activity assay for CYP1A2, 2A6 and 3A4. Biochem Pharmacol 2018; 158:134-140. [PMID: 30308189 DOI: 10.1016/j.bcp.2018.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022]
Abstract
Cytochrome P450s (CYPs) are key enzymes involved in drug and xenobiotic metabolism. A wide array of in vitro methodologies, including recombinant sources, are currently been used to assess CYP catalysis, to identify the metabolic profile of compounds, potential drug-drug interactions, protein-protein interactions in the CYP enzyme complex and the role of polymorphic enzymes. We report here on a bacterial whole-cells high-throughput method for the activity evaluation of human CYP1A2, 2A6, and 3A4, when sustained by NADPH cytochrome P450 oxidoreductase (CPR), in the absence or presence of cytochrome b5 (CYB5). This new assay consists of a microplate real-time fluorometric method, with direct measurement of metabolite formation, in a suspension of Escherichia coli BTC-CYP bacteria, a human CYP competent tester strain when incubated with specific fluorogenic substrates. Overall, the maximum turnover (kcat) velocities of the three human CYPs resulting from the whole-BTC cells assays were similar to those obtained when applying the corresponding standard reference membrane fractions assays. CYP activity screening with co-expression of CYB5 suggests an enhancing effect of CYB5 on the kcat of specific isoforms, when using the whole-BTC cells assay. Our results demonstrate that this new approach can offer an efficient high-throughput method for screening of CYP1A2, 2A6 and 3A4 activity and can be potentially applicable for other human CYPs. This can be of particular use for timely and efficient screening of chemical libraries or mutant libraries of CYP enzyme complex proteins, without the necessity for labor intensive isolation of subcellular fractions.
Collapse
Affiliation(s)
- Francisco Esteves
- Center for Toxicogenomics and Human Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal. http://www.fcm.unl.pt
| | - Diana Campelo
- Center for Toxicogenomics and Human Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Philippe Urban
- LISBP, Université de Toulouse, CNRS, INRA, INSA, Toulouse, France
| | - Sophie Bozonnet
- LISBP, Université de Toulouse, CNRS, INRA, INSA, Toulouse, France
| | - Thomas Lautier
- LISBP, Université de Toulouse, CNRS, INRA, INSA, Toulouse, France
| | - José Rueff
- Center for Toxicogenomics and Human Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Gilles Truan
- LISBP, Université de Toulouse, CNRS, INRA, INSA, Toulouse, France
| | - Michel Kranendonk
- Center for Toxicogenomics and Human Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
35
|
Xu M, Tang H, Zhou X, Chen H, Dong Q, Zhang Y, Ye G, Shi F, Lv C, Jing B, He C, Zhao L, Li Y. Effects and mechanisms of sub-chronic exposure to copper nanoparticles on renal cytochrome P450 enzymes in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 63:135-146. [PMID: 30100346 DOI: 10.1016/j.etap.2018.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 06/08/2023]
Abstract
Copper nanoparticles (Cu NPs) have widespread application because of their special physicochemical characteristics, however we need to more clearly study the toxicity mechanism of Cu NPs to ensure its safe use in pharmaceutical and animal feed. Thus, the aim of this study was to evaluate the effects and mechanisms of sub-chronic exposure to Cu NPs on renal CYP450 s of rats. In this study, we investigated the effects of Cu NPs on renal oxidative stress, cytokines and histopathology of rats. We found that Cu NPs (200 mg/kg) significantly disordered the function and structure of the kidney and caused a dose-dependent increase in oxidative stress and cytokines, which significantly decreased the levels of mRNA, protein, and activity of CYP450 s. Micro-coppers (Cu MPs) and Cu ions have similar effects, but their effects on CYP450 s were weaker than Cu NPs. The expression of nuclear receptors were inhibited and the expression of Akt, STAT3/5, CREB, p70S6K, NF-κB, P38 and ERK1/2 were activated when the inhibition effects of CYP450 s activity were observed in renal of rats. Therefore, we believe that Cu NPs can activate the STAT, NF-κB and MAPK signaling pathways to down-regulate the expression and activity of CYP450 s by inducing oxidative stress and inflammatory response in rat kidney.
Collapse
Affiliation(s)
- Min Xu
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Huaqiao Tang
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - XueRong Zhou
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Helin Chen
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Qi Dong
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Yuanli Zhang
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Gang Ye
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Fei Shi
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Cheng Lv
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Bo Jing
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Changliang He
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Ling Zhao
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Yinglun Li
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China.
| |
Collapse
|
36
|
Tarantino G, Capone D, Contaldi P, Gianno A, Teresa M, Tufano A. Phenotyping of CYP 4501A2 Activity by Total Overnight Salivary Caffeine Assessment (TOSCA) in Patients on Warfarin Treatment: A Cross-Sectional Study. Clin Appl Thromb Hemost 2018; 24:928-935. [PMID: 28992765 PMCID: PMC6714717 DOI: 10.1177/1076029617733040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Warfarin is an oral anticoagulant, commonly used for primary and secondary prevention of venous and arterial thromboembolic events. The drug is characterized by narrow therapeutic index, widespread individual variability in clinical response, and high rates of adverse events, particularly bleeding complications. For these reasons, a close monitoring of the dosage, using the frequent assessment of coagulation status by means of International Normalized Ratio value, is mandatory. Warfarin is metabolized by hepatic cytochrome P-450. High CYP 450 activity may lead to low drug concentration and requires high warfarin doses to reach efficacy; conversely, low CYP 450 activity is responsible for high drug concentration and needs for low doses to avoid potential toxicity risks. The major isoforms of CYP involved in the metabolism of warfarin sodium are CYP1A2 (for the R-warfarin) and CYP2C9 (for the S-warfarin). The probes for testing CYP1A2 are phenacetin and caffeine while for CYP2C9 tolbutamide. Although S-warfarin has major activity, it was decided to exclude its phenotyping for ethical issues, being mandatory to use a drug (tolbutamide). Instead, it was chosen to test the 1A2 isoform, as the activity of the latter isoform could be investigated by using caffeine contained in the caffeinated beverages. Specifically, a single-point concentration of salivary caffeine (total overnight salivary caffeine assessment [TOSCA]) after an overnight period of the caffeinated beverages abstinence was utilized. In the present study, 75 nonsmoker patients regularly receiving warfarin sodium were enrolled. The results have showed a significant association of the warfarin dose with TOSCA values (coefficient = -0.15, standard error = 0.04, 95% confidence interval = -0.24 to -0.06, t = -3.23, P = .002). In conclusion, the phenotyping of CYP1A2 by TOSCA could be useful, if further proven, to help manage patients on warfarin in order to lessen severe adverse events.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, “Federico II” University, Naples, Italy
| | - Domenico Capone
- Integrated Care Department of Clinical Neurosciences, Anesthesiology and Drug-Use, Section of Clinical Pharmacology, “Federico II” University, Naples, Italy
| | - Paola Contaldi
- Department of Clinical Medicine and Surgery, “Federico II” University, Naples, Italy
| | - Adriana Gianno
- Department of Clinical Medicine and Surgery, “Federico II” University, Naples, Italy
| | - Mosca Teresa
- Integrated Care Department of Clinical Neurosciences, Anesthesiology and Drug-Use, Section of Clinical Pharmacology, “Federico II” University, Naples, Italy
| | - Antonella Tufano
- Department of Clinical Medicine and Surgery, “Federico II” University, Naples, Italy
| |
Collapse
|
37
|
Quesnot N, Bucher S, Gade C, Vlach M, Vene E, Valença S, Gicquel T, Holst H, Robin MA, Loyer P. Production of chlorzoxazone glucuronides via cytochrome P4502E1 dependent and independent pathways in human hepatocytes. Arch Toxicol 2018; 92:3077-3091. [PMID: 30151596 DOI: 10.1007/s00204-018-2300-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/23/2018] [Indexed: 12/28/2022]
Abstract
CYP2E1 activity is measured in vitro and in vivo via hydroxylation of the Chlorzoxazone (CHZ) producing the 6-hydroxychlorzoxazone (OH-CHZ) further metabolized as a glucuronide excreted in urine. Thus, the quantification of the OH-CHZ following enzymatic hydrolysis of CHZ-derived glucuronide appears to be a reliable assay to measure the CYP2E1 activity without direct detection of this glucuronide. However, OH-CHZ hydrolyzed from urinary glucuronide accounts for less than 80% of the CHZ administrated dose in humans leading to postulate the production of other unidentified metabolites. Moreover, the Uridine 5'-diphospho-glucuronosyltransferase (UGT) involved in the hepatic glucuronidation of OH-CHZ has not yet been identified. In this study, we used recombinant HepG2 cells expressing CYP2E1, metabolically competent HepaRG cells, primary hepatocytes and precision-cut human liver slices to identify metabolites of CHZ (300 μM) by high pressure liquid chromatography-UV and liquid-chromatography-mass spectrometry analyses. Herein, we report the detection of the CHZ-O-glucuronide (CHZ-O-Glc) derived from OH-CHZ in culture media but also in mouse and human urine and we identified a novel CHZ metabolite, the CHZ-N-glucuronide (CHZ-N-Glc), which is resistant to enzymatic hydrolysis and produced independently of CHZ hydroxylation by CYP2E1. Moreover, we demonstrate that UGT1A1, 1A6 and 1A9 proteins catalyze the synthesis of CHZ-O-Glc while CHZ-N-Glc is produced by UGT1A9 specifically. Together, we demonstrated that hydrolysis of CHZ-O-Glc is required to reliably quantify CYP2E1 activity because of the rapid transformation of OH-CHZ into CHZ-O-Glc and identified the CHZ-N-Glc produced independently of the CYP2E1 activity. Our results also raise the questions of the contribution of CHZ-N-Glc in the overall CHZ metabolism and of the quantification of CHZ glucuronides in vitro and in vivo for measuring UGT1A activities.
Collapse
Affiliation(s)
- Nicolas Quesnot
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, 35000, Rennes, France
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Simon Bucher
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, 35000, Rennes, France
| | - Christina Gade
- Department of Clinical Pharmacology, Bispebjerg Hospital, 23 Bispebjerg Bakke, 2400, Copenhagen, NV, Denmark
| | - Manuel Vlach
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, 35000, Rennes, France
| | - Elise Vene
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, 35000, Rennes, France
| | - Samuel Valença
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thomas Gicquel
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, 35000, Rennes, France
| | - Helle Holst
- Department of Clinical Pharmacology, Bispebjerg Hospital, 23 Bispebjerg Bakke, 2400, Copenhagen, NV, Denmark
| | - Marie-Anne Robin
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, 35000, Rennes, France
| | - Pascal Loyer
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, 35000, Rennes, France.
- Institut NuMeCan, Inserm U1241, Hôpital Pontchaillou, 35033, Rennes, France.
| |
Collapse
|
38
|
Muradás TC, Abbadi BL, Villela AD, Macchi FS, Bergo PF, de Freitas TF, Sperotto NDM, Timmers LFSM, Norberto de Souza O, Picada JN, Fachini J, da Silva JB, de Albuquerque NCP, Habenschus MD, Carrão DB, Rocha BA, Barbosa Junior F, de Oliveira ARM, Mascarello A, Neuenfeldf P, Nunes RJ, Morbidoni HR, Campos MM, Basso LA, Rodrigues-Junior VS. Pre-clinical evaluation of quinoxaline-derived chalcones in tuberculosis. PLoS One 2018; 13:e0202568. [PMID: 30114296 PMCID: PMC6095594 DOI: 10.1371/journal.pone.0202568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
New effective compounds for tuberculosis treatment are needed. This study evaluated the effects of a series of quinoxaline-derived chalcones against laboratorial strains and clinical isolates of M. tuberculosis. Six molecules, namely N5, N9, N10, N15, N16, and N23 inhibited the growth of the M. tuberculosis H37Rv laboratorial strain. The three compounds (N9, N15 and N23) with the lowest MIC values were further tested against clinical isolates and laboratory strains with mutations in katG or inhA genes. From these data, N9 was selected as the lead compound for further investigation. Importantly, this chalcone displayed a synergistic effect when combined with moxifloxacin. Noteworthy, the anti-tubercular effects of N9 did not rely on inhibition of mycolic acids synthesis, circumventing important mechanisms of resistance. Interactions with cytochrome P450 isoforms and toxic effects were assessed in silico and in vitro. The chalcone N9 was not predicted to elicit any mutagenic, genotoxic, irritant, or reproductive effects, according to in silico analysis. Additionally, N9 did not cause mutagenicity or genotoxicity, as revealed by Salmonella/microsome and alkaline comet assays, respectively. Moreover, N9 did not inhibit the cytochrome P450 isoforms CYP3A4/5, CYP2C9, and CYP2C19. N9 can be considered a potential lead molecule for development of a new anti-tubercular therapeutic agent.
Collapse
Affiliation(s)
- Thaís C. Muradás
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre, Brazil
| | - Bruno L. Abbadi
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Porto Alegre, Brazil
| | - Anne D. Villela
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Fernanda S. Macchi
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Porto Alegre, Brazil
| | - Pedro F. Bergo
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Talita F. de Freitas
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre, Brazil
| | - Nathalia D. M. Sperotto
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre, Brazil
| | - Luis F. S. M. Timmers
- Laboratório de Bioinformática, Modelagem e Simulação de Biossistemas, PUCRS, Porto Alegre, Brazil
| | - Osmar Norberto de Souza
- Laboratório de Bioinformática, Modelagem e Simulação de Biossistemas, PUCRS, Porto Alegre, Brazil
| | - Jaqueline N. Picada
- Laboratory of Toxicological Genetics, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Jean Fachini
- Laboratory of Toxicological Genetics, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | | | - Nayara C. P. de Albuquerque
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Maísa D. Habenschus
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Daniel B. Carrão
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Bruno A. Rocha
- Laboratório de Toxicologia e Essencialidade de Metais, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Fernando Barbosa Junior
- Laboratório de Toxicologia e Essencialidade de Metais, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Anderson R. M. de Oliveira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Alessandra Mascarello
- Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Patrícia Neuenfeldf
- Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Ricardo J. Nunes
- Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Héctor R. Morbidoni
- Laboratorio de Microbiología Molecular, Facultad de Ciencias Médicas, Universidad Nacional de Rosario (UNR), Rosario, Argentina
| | - Maria M. Campos
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre, Brazil
- Centro de Pesquisa de Toxicologia e Farmacologia, PUCRS, Porto Alegre, Brazil
| | - Luiz A. Basso
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Porto Alegre, Brazil
| | - Valnês S. Rodrigues-Junior
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| |
Collapse
|
39
|
MS methods to study macromolecule-ligand interaction: Applications in drug discovery. Methods 2018; 144:152-174. [PMID: 29890284 DOI: 10.1016/j.ymeth.2018.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/01/2018] [Accepted: 06/03/2018] [Indexed: 12/12/2022] Open
Abstract
The interaction of small compounds (i.e. ligands) with macromolecules or macromolecule assemblies (i.e. targets) is the mechanism of action of most of the drugs available today. Mass spectrometry is a popular technique for the interrogation of macromolecule-ligand interactions and therefore is also widely used in drug discovery and development. Thanks to its versatility, mass spectrometry is used for multiple purposes such as biomarker screening, identification of the mechanism of action, ligand structure optimization or toxicity assessment. The evolution and automation of the instruments now allows the development of high throughput methods with high sensitivity and a minimized false discovery rate. Herein, all these approaches are described with a focus on the methods for studying macromolecule-ligand interaction aimed at defining the structure-activity relationships of drug candidates, along with their mechanism of action, metabolism and toxicity.
Collapse
|
40
|
Effect of Gambogenic Acid on Cytochrome P450 1A2, 2B1 and 2E1, and Constitutive Androstane Receptor in Rats. Eur J Drug Metab Pharmacokinet 2018; 43:655-664. [DOI: 10.1007/s13318-018-0477-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Application of in vitro CYP and transporter assays to predict clinical drug–drug interactions. Bioanalysis 2018; 10:619-623. [DOI: 10.4155/bio-2017-0266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
42
|
LC-MS/MS method for the simultaneous quantification of intestinal CYP and UGT activity. J Pharm Biomed Anal 2018; 155:194-201. [PMID: 29649788 DOI: 10.1016/j.jpba.2018.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/18/2023]
Abstract
Many orally administered drugs are subject to first-pass metabolism by cytochrome P450 (CYP) enzymes and uridine 5'-diphospho-glucuronosyltransferases (UGT). While their hepatic activity is well characterized, respective information about the intestine are very scare due to limited availability of tissue, very low microsomal protein content and the heterogeneity of the individual segments. As a consequence, determination of enzyme kinetic parameters is challenging. It was therefore the aim of this study to develop a sensitive liquid chromatography tandem mass spectrometry method for the simultaneous quantification of CYP and UGT metabolites formed by clinically relevant intestinal biotransformation enzymes: 4-hydroxydiclofenac (CYP2C9), 5-hydroxyomeprazole (CYP2C19), dextrorphan (CYP2D6), 1-hydroxymidazolam (CYP3A), ezetimibe glucuronide (UGT1A) and naloxone glucuronide (UGT2B7). After precipitation of microsomal protein with acetonitrile, analytes were chromatographically separated on a C18 column with gradient elution using acetonitrile and water, both containing 0.1% formic acid and detected with a tandem mass spectrometer operating in positive mode with electron spray ionization. The assay was validated according to current bioanalytical guidelines regarding linearity, accuracy, precision, stability, recovery and matrix effects spanning an analytical range from 1 to 200 nmol/L for each analyte. The developed method was successfully applied to a proof of concept experiment using pooled human jejunal microsomes (50 μg protein/mL) in order to determine enzyme kinetic parameters. Formation of all monitored metabolites followed Michaelis-Menten kinetics and allowed calculation of KM and Vmax values. The developed method may be useful for characterization of enzymatic activity in the human intestine which may allow more precise insights into the intestinal contribution to first pass metabolism of drugs.
Collapse
|
43
|
Huguet J, Gaudette F, Michaud V, Turgeon J. Development and validation of probe drug cocktails for the characterization of CYP450-mediated metabolism by human heart microsomes. Xenobiotica 2018; 49:187-199. [PMID: 29448869 DOI: 10.1080/00498254.2018.1438684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
1. The objective of our study was to develop and validate a cocktail approach to allow the simultaneous characterization of various CYP450-mediated oxidations by human heart microsomes for nine probe drug substrates, namely, 7-ethoxyresorufin, bupropion, repaglinide, tolbutamide, bufuralol, chlorzoxazone, ebastine, midazolam and dodecanoic acid. 2. The first validation step was conducted using recombinant human CYP450 isoenzymes by comparing activity measured for each probe drug as a function of (1) buffer used, (2) selectivity towards specific isoenzymes and (3) drug interactions between probes. Activity was all measured by validated LC-MSMS methods. 3. Two cocktails were then constituted with seven of the nine drugs and subjected to kinetic validation. Finally, all probe drugs were incubated with human heart microsomes prepared from ventricular tissues obtained from 12 patients undergoing cardiac transplantation. 4. Validated cocktail #1 including bupropion, chlorzoxazone, ebastine and midazolam was used to characterize CYP2B6-, 2E1-, 2J2- and 3A5-mediated metabolism in human hearts. 5. Cocktail #2 which includes bufuralol, 7-ethoxyresorufin and repaglinide failed the validation step. Substrates in cocktail #2 as well as tolbutamide and dodecanoic acid had to be incubated separately because of their physico-chemical characteristics (solubility and ionization) or drug interactions. 6. Activity in HHM was the highest towards ebastine, chlorzoxazone and tolbutamide.
Collapse
Affiliation(s)
- Jade Huguet
- a CRCHUM, University of Montreal , Montreal , Canada.,b Faculty of Pharmacy , University of Montreal , Montreal , Canada
| | | | - Veronique Michaud
- a CRCHUM, University of Montreal , Montreal , Canada.,b Faculty of Pharmacy , University of Montreal , Montreal , Canada.,c College of Pharmacy , University of Florida, Lake Nona Campus , Orlando , FL , USA
| | - Jacques Turgeon
- c College of Pharmacy , University of Florida, Lake Nona Campus , Orlando , FL , USA
| |
Collapse
|
44
|
Tang H, Long N, Lin L, Liu Y, Li J, Sun F, Guo L, Zhang F, Dai M. Effect of MRSA on CYP450: dynamic changes of cytokines, oxidative stress, and drug-metabolizing enzymes in mice infected with MRSA. Infect Drug Resist 2018; 11:229-238. [PMID: 29491713 PMCID: PMC5815478 DOI: 10.2147/idr.s153871] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) is a very damaging and widespread pathogen, which is associated with many diseases and causes serious infections. MRSA infection can modulate the effects of drugs, which may occur through an influence on cytochrome P450 (CYP450), the drug-metabolizing enzyme in the liver. In this study, we evaluated the underlying mechanism of drug failure or poisoning in MRSA infection. Materials and methods Mice were infected with three different doses of MRSA and the changes in CYP450 expression, cytokines, and oxidative stress markers were evaluated. Results The administration of an attack dose of MRSA caused serious symptoms of infection and resulted in a 40% mortality rate in the mice. MRSA induced strong inflammation and oxidative stress in the mice, predominantly caused by significant increases in interleukin (IL)-1β, IL-4, IL-6, macrophage inflammatory protein, glutathione S-transferase (GST), and malondialdehyde, and decreases in oxygen radical absorbance capacity and glutathione levels in the liver. The expression of IL-2, tumor necrosis factor-α, and GST was briefly suppressed, but increased on days 3 and 7. The increased inflammation and oxidative stress further induced a significant decrease in the mRNA levels and activities of CYP450 1A2, 2D22, 2E1, and 3A1 in MRSA-infected mice within the first day of infection. Conclusion These results show that MRSA infection leads to inflammation and oxidative stress, and reduces the expression levels and activities of drug metabolism enzymes, which decreased drug metabolism in patients infected with MRSA. Therefore, to avoid a drug overdose, the plasma concentration of patients with MRSA infection should be continuously monitored.
Collapse
Affiliation(s)
- Huaqiao Tang
- School of Laboratory Medicine.,Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food
| | - Nana Long
- School of Laboratory Medicine.,Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food
| | - Lin Lin
- School of Laboratory Medicine.,Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food
| | - Yao Liu
- School of Laboratory Medicine
| | - Jianlong Li
- School of Laboratory Medicine.,Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food
| | - Fenghui Sun
- School of Laboratory Medicine.,Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food
| | - Lijuan Guo
- School of Laboratory Medicine.,Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food
| | - Fen Zhang
- School of Humanities and Information Management, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Min Dai
- School of Laboratory Medicine.,Laboratory of Veterinary Drug Residue Prevention and Control Technology of Animal-derived Food
| |
Collapse
|
45
|
den Braver-Sewradj SP, den Braver MW, Baze A, Decorde J, Fonsi M, Bachellier P, Vermeulen NPE, Commandeur JNM, Richert L, Vos JC. Direct comparison of UDP-glucuronosyltransferase and cytochrome P450 activities in human liver microsomes, plated and suspended primary human hepatocytes from five liver donors. Eur J Pharm Sci 2017; 109:96-110. [PMID: 28778465 DOI: 10.1016/j.ejps.2017.07.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 11/26/2022]
Abstract
UDP-glucuronosyltransferases (UGTs) and cytochrome P450s (CYPs) are the major enzymes involved in hepatic metabolism of drugs. Hepatic drug metabolism is commonly investigated using human liver microsomes (HLM) or primary human hepatocytes (PHH). We describe the development of a sensitive assay to phenotype activities of six major hepatic UGT isoforms (UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9 and UGT2B7) in intact PHH by analysis of glucuronidation of selective probe substrates. The non-selective, general substrate 7-hydroxycoumarin was included for comparison. For each liver donor preparation (five donors) UGT activities in cryopreserved suspended and plated PHH were compared to HLM prepared from the same donors. Standard CYP reaction phenotyping of seven major isoforms was performed in parallel. For all donors, CYP- and UGT-isoforms activity profiles were comparable in PHH and HLM, indicating that reaction phenotyping with selective probe substrates in intact cells primarily reflects respective CYP or UGT activity. System-dependent effects on UGT and CYP isoform activity were still found. While UGT activity of UGT1A1 was equivalent in plated and suspended PHH, UGT1A3, UGT1A6 and UGT2B7 activity was higher in suspended PHH and UGT1A9 and UGT1A4 activity was higher in plated PHH. The well-known decrease in activity of most CYP isoforms in plated compared to suspended PHH was confirmed. Importantly, we found a significant loss in CYP2C19 and CYP2B6 in HLM, activity being lower than in intact cells. Taken together, these findings implicate that, dependent on the UGT or CYP isoforms involved in the metabolism of a given compound, the outcome of metabolic assays is strongly dependent on the choice of the in vitro system. The currently described UGT- and CYP- activity profiling method can be used as a standard assay in intact cells and can especially aid in reaction phenotyping of in vitro systems for which a limited number of cells are available.
Collapse
Affiliation(s)
- Shalenie P den Braver-Sewradj
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Michiel W den Braver
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Audrey Baze
- Kaly-Cell, 20A Rue du Général Leclerc, Plobsheim, France; UNISTRA, 4 Rue Blaise Pascal, Strasbourg, France
| | | | | | - Philippe Bachellier
- UNISTRA, 4 Rue Blaise Pascal, Strasbourg, France; Centre de Chirurgie Viscérale et de Transplantation, Hôpital de Hautepierre, 67098 Strasbourg, France
| | - Nico P E Vermeulen
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Jan N M Commandeur
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Lysiane Richert
- Kaly-Cell, 20A Rue du Général Leclerc, Plobsheim, France; PEPITE EA4267, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France.
| | - J Chris Vos
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands.
| |
Collapse
|
46
|
Pouyfung P, Sarapusit S, Rongnoparut P. Effects of Vernonia cinerea Compounds on Drug-metabolizing Cytochrome P450s in Human Liver Microsomes. Phytother Res 2017; 31:1916-1925. [PMID: 28994497 DOI: 10.1002/ptr.5939] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 08/27/2017] [Accepted: 09/06/2017] [Indexed: 11/11/2022]
Abstract
Vernonia cinerea has been widely used in traditional medicines for various diseases and shown to aid in smoking abstinence and has anticancer properties. V. cinerea bioactive compounds, including flavonoids and hirsutinolide-type sesquiterpene lactones, have shown an inhibition effect on the nicotine-metabolizing cytochrome P450 2A6 (CYP2A6) enzyme and hirsutinolides reported suppressing cancer growth. In this study, V. cinerea ethanol extract and its bioactive compounds, including four flavonoids and four hirsutinolides, were investigated for an inhibitory effect on human liver microsomal CYPs 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4 using cocktail inhibition assays combined with LC-MS/MS analysis. Among tested flavonoids, chrysoeriol was more potent in inhibition on CYP2A6 and CYP1A2 than other liver CYPs, with better binding efficiency toward CYP2A6 than CYP1A2 (Ki values in competitive mode of 1.93 ± 0.05 versus 3.39 ± 0.21 μM, respectively). Hirsutinolides were prominent inhibitors of CYP2A6 and CYP2D6, with IC50 values of 12-23 and 15-41 μM, respectively. These hirsutinolides demonstrated time-dependent inhibition, an indication of mechanism-based inactivation, toward CYP2A6. Quantitative prediction of microsomal metabolism of these flavonoids and hirsutinolides, including half-lives and hepatic clearance rate, was examined. These findings may have implications for further in vivo studies of V. cinerea. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Phisit Pouyfung
- Department of Biochemistry, Faculty of Science, Mahidol University, 272 Rama 6 Rd, Ratchathewi, Bangkok, 10400, Thailand
| | - Songklod Sarapusit
- Department of Biochemistry and Center for Innovation in Chemistry, Faculty of Science, Burapha University, 169 Long-Hard Bangsaen Rd, Muang, Chonburi, 20131, Thailand
| | - Pornpimol Rongnoparut
- Department of Biochemistry, Faculty of Science, Mahidol University, 272 Rama 6 Rd, Ratchathewi, Bangkok, 10400, Thailand
| |
Collapse
|
47
|
Abstract
After oral intake of drugs, drugs go through the first pass metabolism in the gut and the liver, which greatly affects the final outcome of the drugs' efficacy and side effects. The first pass metabolism is a complex process involving the gut and the liver tissue, with transport and reaction occurring simultaneously at various locations, which makes it difficult to be reproduced in vitro with conventional cell culture systems. In an effort to tackle this challenge, here we have developed a microfluidic gut-liver chip that can reproduce the dynamics of the first pass metabolism. The microfluidic chip consists of two separate layers for gut epithelial cells (Caco-2) and the liver cells (HepG2), and is designed so that drugs go through a sequential absorption in the gut chamber and metabolic reaction in the liver chamber. We fabricated the chip and showed that the two different cell lines can be successfully co-cultured on chip. When the two cells are cultured on chip, changes in the physiological function of Caco-2 and HepG2 cells were noted. The cytochrome P450 metabolic activity of both cells were significantly enhanced, and the absorptive property of Caco-2 cells on chip also changed in response to the presence of flow. Finally, first pass metabolism of a flavonoid, apigenin, was evaluated as a model compound, and co-culture of gut and liver cells on chip resulted in a metabolic profile that is closer to the reported profile than a monoculture of gut cells. This microfluidic gut-liver chip can potentially be a useful platform to study the complex first pass metabolism of drugs in vitro.
Collapse
|
48
|
Sun Y, Liu Y, Zhang X, Wan C, Lyu T, Zhang L. Effects of m-nisoldipine on the activity and mRNA expression of four CYP isozymes in rats. Xenobiotica 2017; 48:676-683. [DOI: 10.1080/00498254.2017.1358831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Yupeng Sun
- Department of Pharmaceutical Analysis and
| | - Yanyan Liu
- Department of Pharmaceutical Analysis and
| | - Xia Zhang
- Department of Pharmaceutical Analysis and
| | | | - Tao Lyu
- Pharmaceutical Experimental Center, School of Pharmacy, Hebei Medical University, Shijiazhuang, PR China
| | | |
Collapse
|
49
|
Saad M, Matheeussen A, Bijttebier S, Verbueken E, Pype C, Casteleyn C, Van Ginneken C, Apers S, Maes L, Cos P, Van Cruchten S. In vitro CYP-mediated drug metabolism in the zebrafish (embryo) using human reference compounds. Toxicol In Vitro 2017; 42:329-336. [DOI: 10.1016/j.tiv.2017.05.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 10/19/2022]
|
50
|
Helvenstein M, Hambÿe S, Blankert B. Hepatocyte-based flow analytical bioreactor for online xenobiotics metabolism bioprediction. Nanobiomedicine (Rij) 2017; 4:1849543517702898. [PMID: 29942392 PMCID: PMC6009796 DOI: 10.1177/1849543517702898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/25/2017] [Indexed: 11/23/2022] Open
Abstract
The research for new in vitro screening tools for predictive metabolic profiling of drug candidates is of major interest in the pharmaceutical field. The main motivation is to avoid late rejection in drug development and to deliver safer drugs to the market. Thanks to the superparamagnetic properties of iron oxide nanoparticles, a flow bioreactor has been developed which is able to perform xenobiotic metabolism studies. The selected cell line (HepaRG) maintained its metabolic competencies once iron oxide nanoparticles were internalized. Based on magnetically trapped cells in a homemade immobilization chamber, through which a flow of circulating phase was injected to transport nutrients and/or the studied xenobiotic, off-line and online (when coupled to a high-performance liquid chromatography chain) metabolic assays were developed using diclofenac as a reference compound. The diclofenac demonstrated a similar metabolization profile chromatogram, both with the newly developed setup and with the control situation. Highly versatile, this pioneering and innovative instrumental design paves the way for a new approach in predictive metabolism studies.
Collapse
Affiliation(s)
- M Helvenstein
- Laboratory of Pharmaceutical Analysis, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons - UMONS, Mons, Belgium
| | - S Hambÿe
- Laboratory of Pharmaceutical Analysis, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons - UMONS, Mons, Belgium
| | - B Blankert
- Laboratory of Pharmaceutical Analysis, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons - UMONS, Mons, Belgium
| |
Collapse
|