1
|
Foglia M, Guarrera L, Kurosaki M, Cassanmagnago GA, Bolis M, Miduri M, Cereseto A, Umbach A, Craparotta I, Fratelli M, Vallerga A, Paroni G, Zanetti A, Cavallaro AV, Russo L, Garattini E, Terao M. The NIPBL-gene mutation of a Cornelia de Lange Syndrome patient causes deficits in the hepatocyte differentiation of induced Pluripotent Stem Cells via altered chromatin-accessibility. Cell Mol Life Sci 2024; 81:439. [PMID: 39453535 PMCID: PMC11511806 DOI: 10.1007/s00018-024-05481-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 09/24/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
The Cornelia de Lange syndrome (CdLS) is a rare genetic disease, which is characterized by a cohesinopathy. Mutations of the NIPBL gene are observed in 65% of CdLS patients. A novel iPSC (induced Pluripotent Stem Cell) line was reprogrammed from the leukocytes of a CdLS patient carrying a missense mutation of the NIPBL gene. A mutation-corrected isogenic iPSC-line and two iPSC-lines generated from the healthy parents were used as controls. The iPSC lines were differentiated along the hepatocyte-lineage. Comparative immunofluorescence, RNA-seq and ATAC-seq analyses were performed on undifferentiated and differentiated iPSCs. In addition, chromatin organization was studied by ChIP-Seq analysis on the patient derived iPSCs as well as the respective controls. Relative to the mutation-corrected and the healthy-parents iPSCs, the patient-derived counterparts are defective in terms of differentiation along the hepatocyte-lineage. One-third of the genes selectively up-regulated in CdLS-derived iPSCs and hepatic cells are non-protein-coding genes. By converse, most of the selectively down-regulated genes code for transcription factors and proteins regulating neural differentiation. Some of the transcriptionally silenced loci, such as the DPP6 gene on chromosome 7q36.2 and the ZNF gene cluster on chromosome 19p12, are located in closed-chromatin regions. Relative to the corresponding controls, the global transcriptomic differences observed in CdLS undifferentiated iPSCs are associated with altered chromatin accessibility, which was confirmed by ChIP-Seq analysis. Thus, the deficits in the differentiation along the hepatocyte lineage observed in our CdLS patient is likely to be due to a transcriptional dysregulation resulting from a cohesin-dependent alteration of chromatin accessibility.
Collapse
Affiliation(s)
- Marika Foglia
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Luca Guarrera
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Mami Kurosaki
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Giada Andrea Cassanmagnago
- Department of Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Marco Bolis
- Department of Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
- Faculty of Biomedical Sciences, Institute of Oncology Research, USI, 6500, Bellinzona, TI, Switzerland
| | - Matteo Miduri
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Anna Cereseto
- Laboratory of Molecular Virology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Povo, TN, Italy
| | - Alessandro Umbach
- Laboratory of Molecular Virology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Povo, TN, Italy
| | - Ilaria Craparotta
- Department of Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Maddalena Fratelli
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Arianna Vallerga
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Gabriela Paroni
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Adriana Zanetti
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Andrea Vincenzo Cavallaro
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Luca Russo
- Laboratory of Biochemistry and Protein Chemistry, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Enrico Garattini
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Mineko Terao
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| |
Collapse
|
2
|
Mao J, Zeng F, Qin W, Hu M, Xu L, Cheng F, Zhong M, Zhang Y. A joint population pharmacokinetic model to assess the high variability of whole-blood and intracellular tacrolimus in early adult renal transplant recipients. Int Immunopharmacol 2024; 137:112535. [PMID: 38908078 DOI: 10.1016/j.intimp.2024.112535] [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: 04/22/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
Tacrolimus (TAC) has high pharmacokinetic (PK) variability during the early transplantation period. The relationships between whole-blood and intracellular TAC concentrations and clinical outcomes remain controversial. This study identifies the factors affecting the PK variability of TAC and characterizes the relationships between whole-blood and intracellular TAC concentrations. Data regarding whole-blood TAC concentrations of 1,787 samples from 215 renal transplant recipients (<90 days postoperative) across two centers and intracellular TAC concentrations (648 samples) digitized from previous studies were analyzed using nonlinear mixed-effects modeling. The effects of potential covariates were screened, and the distribution of whole-blood to intracellular TAC concentration ratios (RWB:IC) was estimated. The final model was evaluated using bootstrap, goodness of fit, and prediction-corrected visual predictive checks. The optimal dosing regimens and target ranges for each type of immune cell subsets were determined using Monte Carlo simulations. A two-compartment model adequately described the data, and the estimated mean TAC CL/F was 23.6 L·h-1 (relative standard error: 11.5 %). The hematocrit level, CYP3A5*3 carrier status, co-administration with Wuzhi capsules, and tapering prednisolone dose may contribute to the high variability of TAC PK variability during the early post-transplant period. The estimated RWB:IC of all TAC concentrations in peripheral blood mononuclear cells (PBMCs) was 4940, and inter-center variability of PBMCs was observed. The simulated TAC target range in PBMCs was 20.2-85.9 pg·million cells-1. Inter-center variability in intracellular concentrations should be taken into account in further analyses. TAC dosage adjustments can be guided based on PK/PD variability and simulated intracellular concentrations.
Collapse
Affiliation(s)
- Junjun Mao
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China.
| | - Fang Zeng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Road, Wuhan, Hubei 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 1277 Jie Fang Road, Wuhan, Hubei 430022, China
| | - Weiwei Qin
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China
| | - Min Hu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Road, Wuhan, Hubei 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 1277 Jie Fang Road, Wuhan, Hubei 430022, China
| | - Luyang Xu
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China
| | - Fang Cheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Road, Wuhan, Hubei 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 1277 Jie Fang Road, Wuhan, Hubei 430022, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Road, Wuhan, Hubei 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 1277 Jie Fang Road, Wuhan, Hubei 430022, China.
| |
Collapse
|
3
|
Wang Y, Guo D, Winkler R, Lei X, Wang X, Messina J, Luo J, Lu H. Development of novel liver-targeting glucocorticoid prodrugs. MEDICINE IN DRUG DISCOVERY 2024; 21:100172. [PMID: 38390434 PMCID: PMC10883687 DOI: 10.1016/j.medidd.2023.100172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
Background Glucocorticoids (GCs) are widely used in the treatment of inflammatory liver diseases and sepsis, but GC's various side effects on extrahepatic tissues limit their clinical benefits. Liver-targeting GC therapy may have multiple advantages over systemic GC therapy. The purpose of this study was to develop novel liver-targeting GC prodrugs as improved treatment for inflammatory liver diseases and sepsis. Methods A hydrophilic linker or an ultra-hydrophilic zwitterionic linker carboxylic betaine (CB) was used to bridge cholic acid (CA) and dexamethasone (DEX) to generate transporter-dependent liver-targeting GC prodrugs CA-DEX and the highly hydrophilic CA-CB-DEX. The efficacy of liver-targeting DEX prodrugs and DEX were determined in primary human hepatocytes (PHH), macrophages, human whole blood, and/or mice with sepsis induced by cecal ligation and puncture. Results CA-DEX was moderately water soluble, whereas CA-CB-DEX was highly water soluble. CA-CB-DEX and CA-DEX displayed highly transporter-dependent activities in reporter assays. Data mining found marked dysregulation of many GR-target genes important for lipid catabolism, cytoprotection, and inflammation in patients with severe alcoholic hepatitis. These key GR-target genes were similarly and rapidly (within 6 h) induced or down-regulated by CA-CB-DEX and DEX in PHH. CA-CB-DEX had much weaker inhibitory effects than DEX on endotoxin-induced cytokines in mouse macrophages and human whole blood. In contrast, CA-CB-DEX exerted more potent anti-inflammatory effects than DEX in livers of septic mice. Conclusions CA-CB-DEX demonstrated good hepatocyte-selectivity in vitro and better anti-inflammatory effects in vivo. Further test of CA-CB-DEX as a novel liver-targeting GC prodrug for inflammatory liver diseases and sepsis is warranted.
Collapse
Affiliation(s)
- Yazheng Wang
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Dandan Guo
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Rebecca Winkler
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Xiaohong Lei
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Xiaojing Wang
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Jennifer Messina
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Juntao Luo
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| |
Collapse
|
4
|
Lee J, Beers JL, Geffert RM, Jackson KD. A Review of CYP-Mediated Drug Interactions: Mechanisms and In Vitro Drug-Drug Interaction Assessment. Biomolecules 2024; 14:99. [PMID: 38254699 PMCID: PMC10813492 DOI: 10.3390/biom14010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Drug metabolism is a major determinant of drug concentrations in the body. Drug-drug interactions (DDIs) caused by the co-administration of multiple drugs can lead to alteration in the exposure of the victim drug, raising safety or effectiveness concerns. Assessment of the DDI potential starts with in vitro experiments to determine kinetic parameters and identify risks associated with the use of comedication that can inform future clinical studies. The diverse range of experimental models and techniques has significantly contributed to the examination of potential DDIs. Cytochrome P450 (CYP) enzymes are responsible for the biotransformation of many drugs on the market, making them frequently implicated in drug metabolism and DDIs. Consequently, there has been a growing focus on the assessment of DDI risk for CYPs. This review article provides mechanistic insights underlying CYP inhibition/induction and an overview of the in vitro assessment of CYP-mediated DDIs.
Collapse
Affiliation(s)
- Jonghwa Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.L.B.); (R.M.G.)
| | | | | | - Klarissa D. Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.L.B.); (R.M.G.)
| |
Collapse
|
5
|
Authement AK, Isoherranen N. The impact of pregnancy and associated hormones on the pharmacokinetics of Δ 9-tetrahydrocannabinol. Expert Opin Drug Metab Toxicol 2024; 20:73-93. [PMID: 38258511 DOI: 10.1080/17425255.2024.2309213] [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: 10/24/2023] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
INTRODUCTION (-)-Δ9-tetrahydrocannabinol (THC) is the main psychoactive component of cannabis. Cannabis is the most widely used drug of abuse by pregnant individuals, but its maternal-fetal safety is still unclear. The changes in THC disposition during pregnancy may affect THC safety and pharmacology. AREAS COVERED This review summarizes the current literature on THC metabolism and pharmacokinetics in humans. It provides an analysis of how hormonal changes during pregnancy may alter the expression of cannabinoid metabolizing enzymes and THC and its metabolite pharmacokinetics. THC is predominately (>70%) cleared by hepatic metabolism to its psychoactive active metabolite, 11-OH-THC by cytochrome P450 (CYP) 2C9 and to other metabolites (<30%) by CYP3A4. Other physiological processes that change during pregnancy and may alter cannabinoid disposition are also reviewed. EXPERT OPINION THC and its metabolites disposition likely change during pregnancy. Hepatic CYP2C9 and CYP3A4 are induced in pregnant individuals and in vitro by pregnancy hormones. This induction of CYP2C9 and CYP3A4 is predicted to lead to altered THC and 11-OH-THC disposition and pharmacodynamic effects. More in vitro studies of THC metabolism and induction of the enzymes metabolizing cannabinoids are necessary to improve the prediction of THC pharmacokinetics in pregnant individuals.
Collapse
Affiliation(s)
- Aurora K Authement
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
6
|
Zhang Q, Chen Y, Li J, Xia H, Tong Y, Liu Y. Recent Advances in Hepatic Metabolic Regulation by the Nuclear Factor Rev-erbɑ. Curr Drug Metab 2024; 25:2-12. [PMID: 38409696 DOI: 10.2174/0113892002290055240212074758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/28/2024]
Abstract
Rev-erbɑ (NR1D1) is a nuclear receptor superfamily member that plays a vital role in mammalian molecular clocks and metabolism. Rev-erbɑ can regulate the metabolism of drugs and the body's glucose metabolism, lipid metabolism, and adipogenesis. It is even one of the important regulatory factors regulating the occurrence of metabolic diseases (e.g., diabetes, fatty liver). Metabolic enzymes mediate most drug metabolic reactions in the body. Rev-erbɑ has been recognized to regulate drug metabolic enzymes (such as Cyp2b10 and Ugt1a9). Therefore, this paper mainly reviewed that Rev-erbɑ regulates I and II metabolic enzymes in the liver to affect drug pharmacokinetics. The expression of these drug metabolic enzymes (up-regulated or down-regulated) is related to drug exposure and effects/ toxicity. In addition, our discussion extends to Rev-erbɑ regulating some transporters (such as P-gp, Mrp2, and Bcrp), as they also play an essential role in drug metabolism. Finally, we briefly describe the role and mechanism of nuclear receptor Rev-erbɑ in lipid and glucose homeostasis, obesity, and metabolic disorders syndrome. In conclusion, this paper aims to understand better the role and mechanism of Rev-erbɑ in regulating drug metabolism, lipid, glucose homeostasis, obesity, and metabolic disorders syndrome, which explores how to target Rev-erbɑ to guide the design and development of new drugs and provide scientific reference for the molecular mechanism of new drug development, rational drug use, and drug interaction.
Collapse
Affiliation(s)
- Qi Zhang
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yutong Chen
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jingqi Li
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Haishan Xia
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yongbin Tong
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yuyu Liu
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
7
|
Ho YS, Torres-Vergara P, Penny J. Regulation of the ATP-binding cassette transporters ABCB1, ABCG2 and ABCC5 by nuclear receptors in porcine blood-brain barrier endothelial cells. Br J Pharmacol 2023; 180:3092-3109. [PMID: 37476954 DOI: 10.1111/bph.16196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 05/26/2023] [Accepted: 06/22/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND AND PURPOSE Blood-brain barrier (BBB) ABCB1, ABCG2 and ABCC5 transporters influence central therapeutic drug distribution. Transporter expression is regulated by the NR3C1, NR1I3 and NR1I2 nuclear receptors, but their precise roles in brain are poorly understood. We investigated the effects of selective ligand-based activation of NR3C1, NR1I3, NR1I2 and NR2B1 in porcine brain endothelial cells (PBECs). EXPERIMENTAL APPROACH Primary cultures of PBECs were exposed to NR3C1, NR1I3 and NR1I2 ligands and ABCB1, ABCG2 and ABCC5 transporter activities determined by measuring intracellular accumulation of fluorescent probes. Western blotting was used to determine the effects of receptor ligands on expression of ABCB1, ABCG2, ABCC5, NR1I2, NR1I3, NR3C1 and NR2B1. Fluorescent immunocytochemistry was employed to assess the effects of receptor ligands on the cellular localisation of NR1I2 and NR1I3. KEY RESULTS The NR1I2 agonist rifampicin significantly up-regulated ABCG2 activity, which is counteracted by co-treatment with NR1I2 antagonist l-sulforaphane. The NR1I3 agonist 6-(4-chlorophenyl)-imidazo[2,1-b]thiazole-5-carbaldehyde and inverse agonist meclizine significantly down-regulated ABCB1, ABCG2 and ABCC5 activity. NR3C1 agonist dexamethasone significantly increased ABCB1, ABCG2 and ABCC5 activity and ABCG2 and ABCC5 protein expression, which was counteracted by co-treatment with the NR3C1 antagonist mifepristone. This first study demonstrates that NR1I3 and NR3C1 regulate ABCC5 activity and protein expression in BBB endothelial cells. CONCLUSIONS AND IMPLICATIONS In PBECs, expression of key ATP-binding cassette (ABC) transporters and nuclear receptors is differentially regulated by NR1I3, NR1I2, NR3C1 and NR2B1. This will help to better understand the response of the BBB to physiological and pharmacological activation of nuclear receptors.
Collapse
Affiliation(s)
- Yu Siong Ho
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Pablo Torres-Vergara
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Bourdin V, Bigot W, Vanjak A, Burlacu R, Lopes A, Champion K, Depond A, Amador-Borrero B, Sene D, Comarmond C, Mouly S. Drug-Drug Interactions Involving Dexamethasone in Clinical Practice: Myth or Reality? J Clin Med 2023; 12:7120. [PMID: 38002732 PMCID: PMC10672071 DOI: 10.3390/jcm12227120] [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: 09/19/2023] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Concomitant administration of multiple drugs frequently causes severe pharmacokinetic or pharmacodynamic drug-drug interactions (DDIs) resulting in the possibility of enhanced toxicity and/or treatment failure. The activity of cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp), a drug efflux pump sharing localization and substrate affinities with CYP3A4, is a critical determinant of drug clearance, interindividual variability in drug disposition and clinical efficacy, and appears to be involved in the mechanism of numerous clinically relevant DDIs, including those involving dexamethasone. The recent increase in the use of high doses of dexamethasone during the COVID-19 pandemic have emphasized the need for better knowledge of the clinical significance of drug-drug interactions involving dexamethasone in the clinical setting. We therefore aimed to review the already published evidence for various DDIs involving dexamethasone in vitro in cell culture systems and in vivo in animal models and humans.
Collapse
Affiliation(s)
- Venceslas Bourdin
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - William Bigot
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Anthony Vanjak
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Ruxandra Burlacu
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Amanda Lopes
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Karine Champion
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Audrey Depond
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Blanca Amador-Borrero
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Damien Sene
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM U976, Hôpital Saint-Louis, 75010 Paris, France
| | - Chloe Comarmond
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM U976, Hôpital Saint-Louis, 75010 Paris, France
| | - Stéphane Mouly
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM UMR-S1144, Hôpital Fernand Widal, 75010 Paris, France
| |
Collapse
|
9
|
Population Pharmacokinetics of Isavuconazole in Critical Care Patients with COVID-19-Associated Pulmonary Aspergillosis and Monte Carlo Simulations of High Off-Label Doses. J Fungi (Basel) 2023; 9:jof9020211. [PMID: 36836325 PMCID: PMC9960864 DOI: 10.3390/jof9020211] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Isavuconazole is a triazole antifungal agent recently recommended as first-line therapy for invasive pulmonary aspergillosis. With the COVID-19 pandemic, cases of COVID-19-associated pulmonary aspergillosis (CAPA) have been described with a prevalence ranging from 5 to 30%. We developed and validated a population pharmacokinetic (PKpop) model of isavuconazole plasma concentrations in intensive care unit patients with CAPA. Nonlinear mixed-effect modeling Monolix software were used for PK analysis of 65 plasma trough concentrations from 18 patients. PK parameters were best estimated with a one-compartment model. The mean of ISA plasma concentrations was 1.87 [1.29-2.25] mg/L despite prolonged loading dose (72 h for one-third) and a mean maintenance dose of 300 mg per day. Pharmacokinetics (PK) modeling showed that renal replacement therapy (RRT) was significantly associated with under exposure, explaining a part of clearance variability. The Monte Carlo simulations suggested that the recommended dosing regimen did not achieve the trough target of 2 mg/L in a timely manner (72 h). This is the first isavuconazole PKpop model developed for CAPA critical care patients underlying the need of therapeutic drug monitoring, especially for patients under RRT.
Collapse
|
10
|
Li K, Yu XH, Maskey AR, Musa I, Wang ZZ, Garcia V, Guo A, Yang N, Srivastava K, Dunkin D, Li JX, Guo L, Cheng YC, Yuan H, Tiwari R, Li XM. Cytochrome P450 3A4 suppression by epimedium and active compound kaempferol leads to synergistic anti-inflammatory effect with corticosteroid. Front Pharmacol 2023; 13:1042756. [PMID: 36793921 PMCID: PMC9922998 DOI: 10.3389/fphar.2022.1042756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/23/2022] [Indexed: 01/31/2023] Open
Abstract
Introduction: Cytochrome P450 (CYP) 3A4 is a major drug metabolizing enzyme for corticosteroids (CS). Epimedium has been used for asthma and variety of inflammatory conditions with or without CS. It is unknown whether epimedium has an effect on CYP 3A4 and how it interacts with CS. We sought to determine the effects of epimedium on CYP3A4 and whether it affects the anti-inflammatory function of CS and identify the active compound responsible for this effect. Methods: The effect of epimedium on CYP3A4 activity was evaluated using the Vivid CYP high-throughput screening kit. CYP3A4 mRNA expression was determined in human hepatocyte carcinoma (HepG2) cells with or without epimedium, dexamethasone, rifampin, and ketoconazole. TNF-α levels were determined following co-culture of epimedium with dexamethasone in a murine macrophage cell line (Raw 264.7). Active compound (s) derived from epimedium were tested on IL-8 and TNF-α production with or without corticosteroid, on CYP3A4 function and binding affinity. Results: Epimedium inhibited CYP3A4 activity in a dose-dependent manner. Dexamethasone enhanced the expression of CYP3A4 mRNA, while epimedium inhibited the expression of CYP3A4 mRNA and further suppressed dexamethasone enhancement of CYP3A4 mRNA expression in HepG2 cells (p < 0.05). Epimedium and dexamethasone synergistically suppressed TNF-α production by RAW cells (p < 0.001). Eleven epimedium compounds were screened by TCMSP. Among the compounds identified and tested only kaempferol significantly inhibited IL-8 production in a dose dependent manner without any cell cytotoxicity (p < 0.01). Kaempferol in combination with dexamethasone showed complete elimination of TNF-α production (p < 0.001). Furthermore, kaempferol showed a dose dependent inhibition of CYP3A4 activity. Computer docking analysis showed that kaempferol significantly inhibited the catalytic activity of CYP3A4 with a binding affinity of -44.73kJ/mol. Discussion: Inhibition of CYP3A4 function by epimedium and its active compound kaempferol leads to enhancement of CS anti-inflammatory effect.
Collapse
Affiliation(s)
- Ke Li
- Guangdong Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, China
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Xiu-Hua Yu
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
- Central Laboratory, Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Anish R. Maskey
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Ibrahim Musa
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Zhen-Zheng Wang
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Austin Guo
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Nan Yang
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
- General Nutraceutical Technology, Elmsford, NY, United States
| | - Kamal Srivastava
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
- General Nutraceutical Technology, Elmsford, NY, United States
| | - David Dunkin
- Department of Pediatrics, Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jun-Xiong Li
- Guangdong Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, China
| | - Longgang Guo
- Guangzhou ImVin Pharmaceutical Co., Ltd., Guangzhou, China
| | - Yung-Chi Cheng
- Department of Pharmacology, School of Medicine, Yale University, New Haven, China
| | - Haoliang Yuan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Raj Tiwari
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Xiu-Min Li
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
- Department of Otolaryngology, Westchester Medical Center New York Medical College, Valhalla, NY, United States
| |
Collapse
|
11
|
Cheng A, Lei S, Zhu J, Lu J, Paine MF, Xie W, Ma X. Chemical basis of pregnane X receptor activators in the herbal supplement Gancao (licorice)☆. LIVER RESEARCH 2022. [DOI: 10.1016/j.livres.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
12
|
Klyushova LS, Perepechaeva ML, Grishanova AY. The Role of CYP3A in Health and Disease. Biomedicines 2022; 10:2686. [PMID: 36359206 PMCID: PMC9687714 DOI: 10.3390/biomedicines10112686] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
CYP3A is an enzyme subfamily in the cytochrome P450 (CYP) superfamily and includes isoforms CYP3A4, CYP3A5, CYP3A7, and CYP3A43. CYP3A enzymes are indiscriminate toward substrates and are unique in that these enzymes metabolize both endogenous compounds and diverse xenobiotics (including drugs); almost the only common characteristic of these compounds is lipophilicity and a relatively large molecular weight. CYP3A enzymes are widely expressed in human organs and tissues, and consequences of these enzymes' activities play a major role both in normal regulation of physiological levels of endogenous compounds and in various pathological conditions. This review addresses these aspects of regulation of CYP3A enzymes under physiological conditions and their involvement in the initiation and progression of diseases.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, 630117 Novosibirsk, Russia
| | | |
Collapse
|
13
|
Tarek Mahmoud S, Moffid MA, Sayed RM, Mostafa EA. Core shell stationary phase for a novel separation of some COVID-19 used drugs by UPLC-MS/MS Method: Study of grapefruit consumption impact on their pharmacokinetics in rats. Microchem J 2022; 181:107769. [PMID: 35855210 PMCID: PMC9284531 DOI: 10.1016/j.microc.2022.107769] [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: 04/22/2022] [Revised: 06/11/2022] [Accepted: 07/06/2022] [Indexed: 12/15/2022]
Abstract
A sensitive and selective UPLC-MS/MS method was developed for the synchronized determination of four drugs used in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), namely, azithromycin, apixaban, dexamethasone, and favipiravir in rat plasma. using a Poroshell 120 EC-C18 column (50 mm × 4.6 mm, 2.7 m) with a high-resolution ESI tandem mass spectrometer detection with multiple reaction monitoring. We used an Agilent Poroshell column, which is characterized by a stationary phase based on non-porous core particles. With a remarkable improvement in the number of theoretical plates and low column backpressure. In addition, the developed method was employed in studying the potential food-drug interaction of grapefruit juice (GFJ) with the selected drugs which affects their pharmacokinetics in rats. The LC-MS/MS operated in positive and negative ionization mode using two internal standards: moxifloxacin and chlorthalidone, respectively. Liquid- liquid extraction of the cited drugs from rat plasma was accomplished using diethyl ether: dichloromethane (70:30, v/v). The analytes were separated using methanol: 0.1 % formic acid in water (95: 5, v/v) as a mobile phase in isocratic mode of elution pumped at a flow rate of 0.3 mL/min. A detailed validation of the bio-analytical method was performed in accordance with US-FDA and EMA guidelines. Concerning the in vivo pharmacokinetic study, the statistical significance between the results of the test groups receiving GFJ along with the cited drugs and the control group was assessed demonstrating that GFJ increased the plasma concentration of azithromycin, apixaban, and dexamethasone. Accordingly, this food-drug interaction requires cautious ingestion of GFJ in patients using (SARS-CoV-2) medications as it can produce negative effects in the safety of the drug therapy. A potential drug-drug interaction is also suggested between those medications requiring a suitable dose adjustment.
Collapse
Affiliation(s)
- Sally Tarek Mahmoud
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| | - Marwa A Moffid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| | - Rawda M Sayed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| | - Eman A Mostafa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| |
Collapse
|
14
|
Eng ME, Imperio GE, Bloise E, Matthews SG. ATP-binding cassette (ABC) drug transporters in the developing blood-brain barrier: role in fetal brain protection. Cell Mol Life Sci 2022; 79:415. [PMID: 35821142 PMCID: PMC11071850 DOI: 10.1007/s00018-022-04432-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/27/2022] [Accepted: 06/15/2022] [Indexed: 12/19/2022]
Abstract
The blood-brain barrier (BBB) provides essential neuroprotection from environmental toxins and xenobiotics, through high expression of drug efflux transporters in endothelial cells of the cerebral capillaries. However, xenobiotic exposure, stress, and inflammatory stimuli have the potential to disrupt BBB permeability in fetal and post-natal life. Understanding the role and ability of the BBB in protecting the developing brain, particularly with respect to drug/toxin transport, is key to promoting long-term brain health. Drug transporters, particularly P-gp and BCRP are expressed in early gestation at the developing BBB and have a crucial role in developmental homeostasis and fetal brain protection. We have highlighted several factors that modulate drug transporters at the developing BBB, including synthetic glucocorticoid (sGC), cytokines, maternal infection, and growth factors. Some factors have the potential to increase expression and function of drug transporters and increase brain protection (e.g., sGC, transforming growth factor [TGF]-β). However, others inhibit drug transporters expression and function at the BBB, increasing brain exposure to xenobiotics (e.g., tumor necrosis factor [TNF], interleukin [IL]-6), negatively impacting brain development. This has implications for pregnant women and neonates, who represent a vulnerable population and may be exposed to drugs and environmental toxins, many of which are P-gp and BCRP substrates. Thus, alterations in regulated transport across the developing BBB may induce long-term changes in brain health and compromise pregnancy outcome. Furthermore, a large portion of neonatal adverse drug reactions are attributed to agents that target or access the nervous system, such as stimulants (e.g., caffeine), anesthetics (e.g., midazolam), analgesics (e.g., morphine) and antiretrovirals (e.g., Zidovudine); thus, understanding brain protection is key for the development of strategies to protect the fetal and neonatal brain.
Collapse
Affiliation(s)
- Margaret E Eng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | | | - Enrrico Bloise
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Stephen G Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada.
- Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
15
|
Dutta M, Lim JJ, Cui JY. Pregnane X Receptor and the Gut-Liver Axis: A Recent Update. Drug Metab Dispos 2022; 50:478-491. [PMID: 34862253 PMCID: PMC11022899 DOI: 10.1124/dmd.121.000415] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/02/2021] [Indexed: 02/04/2023] Open
Abstract
It is well-known that the pregnane X receptor (PXR)/Nr1i2 is a critical xenobiotic-sensing nuclear receptor enriched in liver and intestine and is responsible for drug-drug interactions, due to its versatile ligand binding domain (LBD) and target genes involved in xenobiotic biotransformation. PXR can be modulated by various xenobiotics including pharmaceuticals, nutraceuticals, dietary factors, and environmental chemicals. Microbial metabolites such as certain secondary bile acids (BAs) and the tryptophan metabolite indole-3-propionic acid (IPA) are endogenous PXR activators. Gut microbiome is increasingly recognized as an important regulator for host xenobiotic biotransformation and intermediary metabolism. PXR regulates and is regulated by the gut-liver axis. This review summarizes recent research advancements leveraging pharmaco- and toxico-metagenomic approaches that have redefined the previous understanding of PXR. Key topics covered in this review include: (1) genome-wide investigations on novel PXR-target genes, novel PXR-DNA interaction patterns, and novel PXR-targeted intestinal bacteria; (2) key PXR-modulating activators and suppressors of exogenous and endogenous sources; (3) novel bidirectional interactions between PXR and gut microbiome under physiologic, pathophysiological, pharmacological, and toxicological conditions; and (4) modifying factors of PXR-signaling including species and sex differences and time (age, critical windows of exposure, and circadian rhythm). The review also discusses critical knowledge gaps and important future research topics centering around PXR. SIGNIFICANCE STATEMENT: This review summarizes recent research advancements leveraging O'mics approaches that have redefined the previous understanding of the xenobiotic-sensing nuclear receptor pregnane X receptor (PXR). Key topics include: (1) genome-wide investigations on novel PXR-targeted host genes and intestinal bacteria as well as novel PXR-DNA interaction patterns; (2) key PXR modulators including microbial metabolites under physiological, pathophysiological, pharmacological, and toxicological conditions; and (3) modifying factors including species, sex, and time.
Collapse
Affiliation(s)
- Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
16
|
Rachmale M, Rajput N, Jadav T, Sahu AK, Tekade RK, Sengupta P. Implication of metabolomics and transporter modulation based strategies to minimize multidrug resistance and enhance site-specific bioavailability: a needful consideration toward modern anticancer drug discovery. Drug Metab Rev 2022; 54:101-119. [PMID: 35254954 DOI: 10.1080/03602532.2022.2048007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Induction of drug-metabolizing enzymes and efflux transporters (DMET) through activation of pregnane x receptor (PXR) is the primary factor involved in almost all bioavailability and drug resistance-related problems of anticancer drugs. PXR is a transcriptional regulator of many metabolizing enzymes and efflux transporters proteins like p-glycoprotein (p-gp), multidrug resistant protein 1 and 2 (MRP 1 and 2), and breast cancer resistant protein (BCRP), etc. Several anticancer drugs are potent activators of PXR receptors and can modulate the gene expression of DMET proteins. Involvement of anticancer drugs in transcriptional regulation of DMET can prompt increased metabolism and efflux of their own or other co-administered drugs, which leads to poor site-specific bioavailability and increased drug resistance. In this review, we have discussed several novel strategies to evade drug-induced PXR activation and p-gp efflux including assessment of PXR ligand and p-gp substrate at early stages of drug discovery. Additionally, we have critically discussed the chemical structure and drug delivery-based approaches to avoid PXR binding and inhibit the p-gp activity of the drugs at their target sites.
Collapse
Affiliation(s)
- Megha Rachmale
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Niraj Rajput
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Tarang Jadav
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Amit Kumar Sahu
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Pinaki Sengupta
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| |
Collapse
|
17
|
Jacobs TG, Marzolini C, Back DJ, Burger DM. Dexamethasone is a dose-dependent perpetrator of drug-drug interactions: implications for use in people living with HIV. J Antimicrob Chemother 2022; 77:568-573. [PMID: 34791318 PMCID: PMC8690014 DOI: 10.1093/jac/dkab412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Global use of dexamethasone in COVID-19 patients has revealed a poor understanding of the drug-drug interaction (DDI) potential of dexamethasone, particularly with antiretroviral agents (ARVs). Dexamethasone is both a substrate and a dose-dependent inducer of cytochrome P450 3A4 (CYP3A4). As many ARVs are substrates and/or inhibitors or inducers of CYP3A4, there is concern about DDIs with dexamethasone either as a perpetrator or a victim. Assessment of DDIs that involve dexamethasone is complex as dexamethasone is used at a range of daily doses (generally 0.5 up to 40 mg) and a treatment course can be short, long, or intermittent. Moreover, DDIs with dexamethasone have been evaluated only for a limited number of drugs. Here, we summarize the available in vitro and in vivo data on the interaction potential of dexamethasone and provide recommendations for the management of DDIs with ARVs, considering various dexamethasone dosages and treatment durations.
Collapse
Affiliation(s)
- Tom G Jacobs
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Catia Marzolini
- Division of Infectious Diseases and Hospital Epidemiology, Departments of Medicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - David J Back
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - David M Burger
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
18
|
Rodrigues JS, Faria-Pereira A, Camões SP, Serras AS, Morais VA, Ruas JL, Miranda JP. Improving human mesenchymal stem cell-derived hepatic cell energy metabolism by manipulating glucose homeostasis and glucocorticoid signaling. Front Endocrinol (Lausanne) 2022; 13:1043543. [PMID: 36714559 PMCID: PMC9880320 DOI: 10.3389/fendo.2022.1043543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/24/2022] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION The development of reliable hepatic in vitro models may provide insights into disease mechanisms, linking hepatocyte dysmetabolism and related pathologies. However, several of the existing models depend on using high concentrations of hepatocyte differentiation-promoting compounds, namely glucose, insulin, and dexamethasone, which is among the reasons that have hampered their use for modeling metabolism-related diseases. This work focused on modulating glucose homeostasis and glucocorticoid concentration to improve the suitability of a mesenchymal stem-cell (MSC)-derived hepatocyte-like cell (HLC) human model for studying hepatic insulin action and disease modeling. METHODS We have investigated the role of insulin, glucose and dexamethasone on mitochondrial function, insulin signaling and carbohydrate metabolism, namely AKT phosphorylation, glycogen storage ability, glycolysis and gluconeogenesis, as well as fatty acid oxidation and bile acid metabolism gene expression in HLCs. In addition, we evaluated cell morphological features, albumin and urea production, the presence of hepatic-specific markers, biotransformation ability and mitochondrial function. RESULTS Using glucose, insulin and dexamethasone levels close to physiological concentrations improved insulin responsiveness in HLCs, as demonstrated by AKT phosphorylation, upregulation of glycolysis and downregulation of Irs2 and gluconeogenesis and fatty acid oxidation pathways. Ammonia detoxification, EROD and UGT activities and sensitivity to paracetamol cytotoxicity were also enhanced under more physiologically relevant conditions. CONCLUSION HLCs kept under reduced concentrations of glucose, insulin and dexamethasone presented an improved hepatic phenotype and insulin sensitivity demonstrating superior potential as an in vitro platform for modeling energy metabolism-related disorders, namely for the investigation of the insulin signaling pathway.
Collapse
Affiliation(s)
- Joana Saraiva Rodrigues
- Research Institute for Medicines (imed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Faria-Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio Póvoas Camões
- Research Institute for Medicines (imed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Sofia Serras
- Research Institute for Medicines (imed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Vanessa Alexandra Morais
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Jorge Lira Ruas
- Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Joana Paiva Miranda
- Research Institute for Medicines (imed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- *Correspondence: Joana Paiva Miranda,
| |
Collapse
|
19
|
Mao J, Qiu X, Qin W, Xu L, Zhang M, Zhong M. Factors Affecting Time-Varying Clearance of Cyclosporine in Adult Renal Transplant Recipients: A Population Pharmacokinetic Perspective. Pharm Res 2021; 38:1873-1887. [PMID: 34750720 DOI: 10.1007/s11095-021-03114-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/20/2021] [Indexed: 11/27/2022]
Abstract
AIM The pharmacokinetic (PK) properties of cyclosporine (CsA) in renal transplant recipients are patient- and time-dependent. Knowledge of this time-related variability is necessary to maintain or achieve CsA target exposure. Here, we aimed to identify factors explaining variabilities in CsA PK properties and characterize time-varying clearance (CL/F) by performing a comprehensive analysis of CsA PK factors using population PK (popPK) modeling of long-term follow-up data from our institution. METHODS In total, 3674 whole-blood CsA concentrations from 183 patients who underwent initial renal transplantation were analyzed using nonlinear mixed-effects modeling. The effects of potential covariates were selected according to a previous study and well-accepted theoretical mechanisms. Model-informed individualized therapeutic regimens were also evaluated. RESULTS A two-compartment model adequately described the data and the estimated mean CsA CL/F was 32.6 L h-1 (relative standard error: 5%). Allometrically scaled body size, hematocrit (HCT) level, CGC haplotype carrier status, and postoperative time may contribute to CsA PK variability. The CsA bioavailability in patients receiving a prednisolone dose (PD) of 80 mg was 20.6% lower than that in patients receiving 20 mg. A significant decrease (52.6%) in CL/F was observed as the HCT increased from 10.5% to 60.5%. The CL/F of the non-CGC haplotype carrier was 14.4% lower than that of the CGC haplotype carrier at 3 months post operation. CONCLUSIONS By monitoring body size, HCT, PD, and CGC haplotype, changes in CsA CL/F over time could be predicted. Such information could be used to optimize CsA therapy. CsA dose adjustments should be considered in different postoperative periods.
Collapse
Affiliation(s)
- Junjun Mao
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Xiaoyan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| | - Weiwei Qin
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| | - Luyang Xu
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Ming Zhang
- Department of Nephrology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| |
Collapse
|
20
|
Fujino C, Sanoh S, Katsura T. Variation in Expression of Cytochrome P450 3A Isoforms and Toxicological Effects: Endo- and Exogenous Substances as Regulatory Factors and Substrates. Biol Pharm Bull 2021; 44:1617-1634. [PMID: 34719640 DOI: 10.1248/bpb.b21-00332] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The CYP3A subfamily, which includes isoforms CYP3A4, CYP3A5, and CYP3A7 in humans, plays important roles in the metabolism of various endogenous and exogenous substances. Gene and protein expression of CYP3A4, CYP3A5, and CYP3A7 show large inter-individual differences, which are caused by many endogenous and exogenous factors. Inter-individual differences can cause negative outcomes, such as adverse drug events and disease development. Therefore, it is important to understand the variations in CYP3A expression caused by endo- and exogenous factors, as well as the variation in the metabolism and kinetics of endo- and exogenous substrates. In this review, we summarize the factors regulating CYP3A expression, such as bile acids, hormones, microRNA, inflammatory cytokines, drugs, environmental chemicals, and dietary factors. In addition, variations in CYP3A expression under pathological conditions, such as coronavirus disease 2019 and liver diseases, are described as examples of the physiological effects of endogenous factors. We also summarize endogenous and exogenous substrates metabolized by CYP3A isoforms, such as cholesterol, bile acids, hormones, arachidonic acid, vitamin D, and drugs. The relationship between the changes in the kinetics of these substrates and the toxicological effects in our bodies are discussed. The usefulness of these substrates and metabolites as endogenous biomarkers for CYP3A activity is also discussed. Notably, we focused on discrimination between CYP3A4, CYP3A5, and CYP3A7 to understand inter-individual differences in CYP3A expression and function.
Collapse
Affiliation(s)
- Chieri Fujino
- Laboratory of Clinical Pharmaceutics and Therapeutics, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,School of Pharmaceutical Sciences, Wakayama Medical University
| | - Toshiya Katsura
- Laboratory of Clinical Pharmaceutics and Therapeutics, College of Pharmaceutical Sciences, Ritsumeikan University
| |
Collapse
|
21
|
Cell-Permeable Succinate Rescues Mitochondrial Respiration in Cellular Models of Amiodarone Toxicity. Int J Mol Sci 2021; 22:ijms222111786. [PMID: 34769217 PMCID: PMC8583998 DOI: 10.3390/ijms222111786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Amiodarone is a potent antiarrhythmic drug and displays substantial liver toxicity in humans. It has previously been demonstrated that amiodarone and its metabolite (desethylamiodarone, DEA) can inhibit mitochondrial function, particularly complexes I (CI) and II (CII) of the electron transport system in various animal tissues and cell types. The present study, performed in human peripheral blood cells, and one liver-derived human cell line, is primarily aimed at assessing the concentration-dependent effects of these drugs on mitochondrial function (respiration and cellular ATP levels). Furthermore, we explore the efficacy of a novel cell-permeable succinate prodrug in alleviating the drug-induced acute mitochondrial dysfunction. Amiodarone and DEA elicit a concentration-dependent impairment of mitochondrial respiration in both intact and permeabilized platelets via the inhibition of both CI- and CII-supported respiration. The inhibitory effect seen in human platelets is also confirmed in mononuclear cells (PBMCs) and HepG2 cells. Additionally, amiodarone elicits a severe concentration-dependent ATP depletion in PBMCs, which cannot be explained solely by mitochondrial inhibition. The succinate prodrug NV118 alleviates the respiratory deficit in platelets and HepG2 cells acutely exposed to amiodarone. In conclusion, amiodarone severely inhibits metabolism in primary human mitochondria, which can be counteracted by increasing mitochondrial function using intracellular delivery of succinate.
Collapse
|
22
|
Kuncharoenwirat N, Chatuphonprasert W, Jarukamjorn K. Differential Impacts of Phenol Red on Benzo[ a]pyrene and Dexamethasone-Modified Cytochrome P450s in Human Cancer Cells. Pak J Biol Sci 2021; 24:790-800. [PMID: 34486298 DOI: 10.3923/pjbs.2021.790.800] [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: 11/15/2022]
Abstract
<b>Background and Objective:</b> Phenol red, the pH indicator in cell culture media, influences the expression of cytochrome P450s (CYPs) in cell lines. This study aimed to examine how phenol red modified CYP induction by benzo[<i>a</i>]pyrene and dexamethasone in human hepatocarcinoma (HepG2), colorectal adenocarcinoma (Caco-2) and choriocarcinoma (BeWo) cells. <b>Materials and Methods:</b> The cells (1×10<sup>5</sup> cells/well in a 24-well plate) were incubated with benzo[<i>a</i>]pyrene (0.1, 1 and 10 μM) or dexamethasone (1, 5 and 10 μM) in either phenol red or phenol red-free media for 24 hrs. The mRNA expression of CYPs was determined by Real-Time Polymerase Chain Reaction (RT/qPCR). <b>Results:</b> Phenol red enhanced expression of benzo[<i>a</i>]pyrene-induced CYP1A2 inHepG2 and BeWo cells and suppressed benzo[<i>a</i>]pyrene-induced CYP2A6 expression in HepG2 and Caco-2 cells, benzo[<i>a</i>]pyrene induced CYP2B6 expression in HepG2 cells and benzo[<i>a</i>]pyrene- and dexamethasone-induced CYP3A4 expression in HepG2 and Caco-2 cells. The expression of CYP3A5 was affected differently in HepG2 and Caco-2 cell lines. Phenol red enhanced benzo[<i>a</i>]pyrene- and dexamethasone-induced CYP3A5 expression in Caco-2 cells but suppressed benzo[<i>a</i>]pyrene- and dexamethasone-induced CYP3A5 expression in HepG2 cells. <b>Conclusion:</b> Phenol red differentially influenced expression of benzo[<i>a</i>]pyrene- and dexamethasone-induced CYP1A2, CYP2A6, CYP2B6, CYP3A4 and CYP3A5 mRNAs in HepG2, Caco-2 and BeWo cells. Therefore, the inclusion of phenol red in cell culture media is of concern in studies of drug and xenobiotic metabolism via CYPs in human cell line models.
Collapse
|
23
|
Bone Mineral Metabolism During Chemotherapy in Childhood Acute Lymphoblastic Leukemia: Unexpected Vitamin D Deficiency From Induction Corticosteroids in Acute Lymphoblastic Leukemia. J Pediatr Hematol Oncol 2021; 43:240-241. [PMID: 33290292 DOI: 10.1097/mph.0000000000002028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
24
|
Smythe MA, Burns C, Liu Q, Garwood CL. Potential Dexamethasone-Direct Oral Anticoagulant Drug Interaction: Is This a Concern in COVID? Ann Pharmacother 2021; 56:319-329. [PMID: 34137279 DOI: 10.1177/10600280211025042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To evaluate the literature on a potential dexamethasone-direct oral anticoagulant (DOAC) drug interaction and provide management considerations with COVID hypercoagulability. DATA SOURCES A search of EMBASE, PubMed, and Google Scholar (January 1990 to May 2021), limited to the English language, using applicable search terms resulted in 137 articles, with 21 relevant articles included. Regulatory agency and clinical guidance documents were also reviewed. STUDY SELECTION AND DATA EXTRACTION Included articles describe in vitro or in vivo animal or human data for dexamethasone induction of cytochrome P450 (CYP) 3A4 or P-glycoprotein (P-gp). DATA SYNTHESIS Dexamethasone has the potential to interact with the DOACs via CYP3A4 and/or P-gp induction. Only apixaban and rivaroxaban have CYP3A4 metabolism. Dexamethasone can increase CYP3A4 activity by up to 70% and reduce the area under the concentration-time curve (AUC) of CYP3A4 substrates by >40%, which is consistent with criteria for a weak CYP inducer. In rodents, dexamethasone P-gp induction is associated with AUC reductions of 20% to 50%. Human data are lacking. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Severe COVID-19 infection is associated with hypercoagulability. Although heparins are the preferred anticoagulants for hospitalized COVID-19 patients, DOACs are being utilized. Dexamethasone is recommended for hospitalized COVID-19 patients requiring supplemental oxygen. The concurrent use of dexamethasone and apixaban or rivaroxaban in such patients carries the potential for reduced anticoagulant effect during a state of heightened thrombotic risk. CONCLUSIONS Concurrent use of dexamethasone and apixaban or rivaroxaban in hospitalized COVID-19 patients with laboratory evidence of COVID coagulopathy should be avoided until higher-quality data are available.
Collapse
Affiliation(s)
- Maureen A Smythe
- Wayne State University, Detroit, MI, USA.,Beaumont Hospital, Royal Oak, MI, USA
| | | | | | - Candice L Garwood
- Wayne State University, Detroit, MI, USA.,Detroit Medical Center, MI, USA
| |
Collapse
|
25
|
Pes K, Friese A, Cox CJ, Laizé V, Fernández I. Biochemical and molecular responses of the Mediterranean mussel (Mytilus galloprovincialis) to short-term exposure to three commonly prescribed drugs. MARINE ENVIRONMENTAL RESEARCH 2021; 168:105309. [PMID: 33798995 DOI: 10.1016/j.marenvres.2021.105309] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 06/12/2023]
Abstract
Pharmaceuticals represent a group of emerging contaminants. The short-term effect (3 and 7 days) of warfarin (1 and 10 mg L-1), dexamethasone (0.392 and 3.92 mg L-1) and imidazole (0.013 and 0.13 mg L-1) exposure was evaluated on mussels (Mytilus galloprovincialis). Total antioxidant status, glutathione reductase, glutathione peroxidase (GPx) and superoxide dismutase enzyme activities, and the expression of genes involved in the xenobiotic response (ATP binding cassette subfamily B member 1 (abcb1) and several nuclear receptor family J (nr1j) isoforms), were evaluated. All nr1j isoforms are suggested to be the xenobiotic receptor orthologs of the NR1I family. All drugs increased GPx activity and altered the expression of particular nr1j isoforms. Dexamethasone exposure also decreased abcb1 expression. These findings raised some concerns regarding the release of these pharmaceuticals into the aquatic environment. Thus, further studies might be needed to perform an accurate environmental risk assessment of these 3 poorly studied drugs.
Collapse
Affiliation(s)
- Katia Pes
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Annika Friese
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Cymon J Cox
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Vincent Laizé
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Ignacio Fernández
- Aquaculture Research Center, Agro-Technological Institute of Castilla y León (ITACyL), Ctra. Arévalo, s/n. 40196 Zamarramala, Segovia, Spain.
| |
Collapse
|
26
|
Li H, Wang YG, Ma ZC, Yun-Hang G, Ling S, Teng-Fei C, Guang-Ping Z, Gao Y. A high-throughput cell-based gaussia luciferase reporter assay for measurement of CYP1A1, CYP2B6, and CYP3A4 induction. Xenobiotica 2021; 51:752-763. [PMID: 33896369 DOI: 10.1080/00498254.2021.1918800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The induction of cytochrome P450s can result in reduced drug efficacy and lead to potential drug-drug interactions. The xenoreceptors-aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR), and pregnane X receptor (PXR)-play key roles in CYP induction by xenobiotics. In order to be able to rapidly screen for the induction of three enzymes (CYP1A1, CYP2B6, and CYP3A4), we generated a stable AhR-responsive HepG2 cell line, a stable CAR-responsive HepG2 cell line, and a stable PXR-responsive HepG2 cell line.To validate these stable xenoreceptor-responsive HepG2 cell lines, we evaluated the induction of the different Gaussia reporter activities, as well as the mRNA and protein expression levels of endogenous CYPs in response to different inducers.The induction of luciferase activity in the stable xenoreceptor-responsive HepG2 cell lines by specific inducers occurred in a concentration dependent manner. There was a positive correlation between the induction of luciferase activities and the induction endogenous CYP mRNA expression levels. These xenoreceptor-responsive HepG2 cell lines were further validated with known CYP1A1, CYP2B6, and CYP3A4 inducers.These stable xenoreceptor-responsive HepG2 cell lines may be used in preclinical research for the rapid and sensitive detection of AhR, CAR, and PXR ligands that induce CYP450 isoforms.
Collapse
Affiliation(s)
- Han Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu-Guang Wang
- Institute of Radiation Medicine Academy of Military Medical Sciences, Beijing, China
| | - Zeng-Chun Ma
- Institute of Radiation Medicine Academy of Military Medical Sciences, Beijing, China
| | - Gao Yun-Hang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Song Ling
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chen Teng-Fei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhang Guang-Ping
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Gao
- Institute of Radiation Medicine Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
27
|
Disease-drug and drug-drug interaction in COVID-19: Risk and assessment. Biomed Pharmacother 2021; 139:111642. [PMID: 33940506 PMCID: PMC8078916 DOI: 10.1016/j.biopha.2021.111642] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is announced as a global pandemic in 2020. Its mortality and morbidity rate are rapidly increasing, with limited medications. The emergent outbreak of COVID-19 prompted by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) keeps spreading. In this infection, a patient's immune response plays pivotal role in the pathogenesis. This inflammatory factor was shown by its mediators that, in severe cases, reach the cytokine at peaks. Hyperinflammatory state may sparks significant imbalances in transporters and drug metabolic machinery, and subsequent alteration of drug pharmacokinetics may result in unexpected therapeutic response. The present scenario has accounted for the requirement for therapeutic opportunities to relive and overcome this pandemic. Despite the diminishing developments of COVID-19, there is no drug still approved to have significant effects with no side effect on the treatment for COVID-19 patients. Based on the evidence, many antiviral and anti-inflammatory drugs have been authorized by the Food and Drug Administration (FDA) to treat the COVID-19 patients even though not knowing the possible drug-drug interactions (DDI). Remdesivir, favipiravir, and molnupiravir are deemed the most hopeful antiviral agents by improving infected patient’s health. Dexamethasone is the first known steroid medicine that saved the lives of seriously ill patients. Some oligopeptides and proteins have also been using. The current review summarizes medication updates to treat COVID-19 patients in an inflammatory state and their interaction with drug transporters and drug-metabolizing enzymes. It gives an opinion on the potential DDI that may permit the individualization of these drugs, thereby enhancing the safety and efficacy.
Collapse
|
28
|
Donia T, Khedr S, Salim EI, Hessien M. Trichostatin A sensitizes hepatoma cells to Taxol more than 5-Aza-dC and dexamethasone. Drug Metab Pers Ther 2021; 36:299-309. [PMID: 34773731 DOI: 10.1515/dmpt-2020-0186] [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: 12/05/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Sherien Khedr
- College of Pharmacy, Arab Academy for Science, Technology & Maritime Transport, Alexandria, Egypt
| | - Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
29
|
Donia T, Khedr S, Salim EI, Hessien M. Trichostatin A sensitizes hepatoma cells to Taxol more than 5-Aza-dC and dexamethasone. Drug Metab Pers Ther 2021; 0:dmdi-2020-0186. [PMID: 33818027 DOI: 10.1515/dmdi-2020-0186] [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: 12/05/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Sherien Khedr
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
30
|
Baburaj G, Thomas L, Rao M. Potential Drug Interactions of Repurposed COVID-19 Drugs with Lung Cancer Pharmacotherapies. Arch Med Res 2021; 52:261-269. [PMID: 33257051 PMCID: PMC7670900 DOI: 10.1016/j.arcmed.2020.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 11/03/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Lung cancer patients are at heightened risk for developing COVID-19 infection as well as complications due to multiple risk factors such as underlying malignancy, anti-cancer treatment induced immunosuppression, additional comorbidities and history of smoking. Recent literatures have reported a significant proportion of lung cancer patients coinfected with COVID-19. Chloroquine, hydroxychloroquine, lopinavir/ritonavir, ribavirin, oseltamivir, remdesivir, favipiravir, and umifenovir represent the major repurposed drugs used as potential experimental agents for COVID-19 whereas azithromycin, dexamethasone, tocilizumab, sarilumab, famotidine and ceftriaxone are some of the supporting agents that are under investigation for COVID-19 management. The rationale of this review is to identify potential drug-drug interactions (DDIs) occurring in lung cancer patients receiving lung cancer medications and repurposed COVID-19 drugs using Micromedex and additional literatures. This review has identified several potential DDIs that could occur with the concomitant treatments of COVID-19 repurposed drugs and lung cancer medications. This information may be utilized by the healthcare professionals for screening and identifying potential DDIs with adverse outcomes, based on their severity and documentation levels and consequently design prophylactic and management strategies for their prevention. Identification, reporting and management of DDIs and dissemination of related information should be a major consideration in the delivery of lung cancer care during this ongoing COVID-19 pandemic for better patient outcomes and updating guidelines for safer prescribing practices in this coinfected condition.
Collapse
Affiliation(s)
- Gayathri Baburaj
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Levin Thomas
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
31
|
Tras B, Eser Faki H, Ozdemir Kutahya Z, Bahcivan E, Dik B, Bozkurt B, Uney K. Treatment and protective effects of metalloproteinase inhibitors alone and in combination with N-Acetyl cysteine plus vitamin E in rats exposed to aflatoxin B 1. Toxicon 2021; 194:79-85. [PMID: 33617885 DOI: 10.1016/j.toxicon.2021.02.012] [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: 11/13/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 11/19/2022]
Abstract
This study was conducted to investigate the effects of matrix metalloproteinase (MMP) inhibitors dexamethasone and minocycline administrations -both single and in combination with N-acetylcysteine (NAC) and vitamin E-on the tissue distribution and lethal dose (LD)50 of aflatoxin (AF)B1 in rats. We performed this study on male Wistar rats (8-10 weeks) in two phases. In the first phase, rats were administered dexamethasone (5 and 20 mg/kg) and minocycline (45 and 90 mg/kg), both as single treatments and in combination with NAC (200 mg/kg) and vitamin E (600 mg/kg); these treatments followed AFB1 administration (2 mg/kg). In the second phase, the therapeutic effect value (TEV) was calculated to determine the treatment effect on the LD50 level of AFB1. The tissue affinity of AFB1 from high to low was liver, kidney, intestine, brain, heart, spleen, lung, testis, and vitreous humor, respectively. Dexamethasone at the 20 mg/kg dose significantly reduced AFB1 concentrations in the plasma and the other tissues, except for the vitreous humor. The effects of minocycline on the plasma and tissue concentrations of AFB1 varied by dose and tissue. The combinations of dexamethasone or minocycline with NAC and vitamin E increased the AFB1 concentrations in the plasma and all tissues, except for vitreous humor and liver. In male rats, the LD50 value of AFB1 was 11.86 mg/kg. The TEV of dexamethasone (20 mg/kg) was calculated to be 1.5. Dexamethasone can be administered in repeated doses at ≥20 mg/kg to increase survival in AFB1 poisoning.
Collapse
Affiliation(s)
- Bunyamin Tras
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031, Konya, Turkey
| | - Hatice Eser Faki
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031, Konya, Turkey
| | - Zeynep Ozdemir Kutahya
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Cukurova, 01930, Adana, Turkey
| | - Emre Bahcivan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Kafkas, 36000, Kars, Turkey
| | - Burak Dik
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031, Konya, Turkey.
| | - Banu Bozkurt
- Department of Ophthalmology, Faculty of Medicine, University of Selcuk, 42031, Konya, Turkey
| | - Kamil Uney
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031, Konya, Turkey
| |
Collapse
|
32
|
Hakkola J, Hukkanen J, Turpeinen M, Pelkonen O. Inhibition and induction of CYP enzymes in humans: an update. Arch Toxicol 2020; 94:3671-3722. [PMID: 33111191 PMCID: PMC7603454 DOI: 10.1007/s00204-020-02936-7] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022]
Abstract
The cytochrome P450 (CYP) enzyme family is the most important enzyme system catalyzing the phase 1 metabolism of pharmaceuticals and other xenobiotics such as herbal remedies and toxic compounds in the environment. The inhibition and induction of CYPs are major mechanisms causing pharmacokinetic drug–drug interactions. This review presents a comprehensive update on the inhibitors and inducers of the specific CYP enzymes in humans. The focus is on the more recent human in vitro and in vivo findings since the publication of our previous review on this topic in 2008. In addition to the general presentation of inhibitory drugs and inducers of human CYP enzymes by drugs, herbal remedies, and toxic compounds, an in-depth view on tyrosine-kinase inhibitors and antiretroviral HIV medications as victims and perpetrators of drug–drug interactions is provided as examples of the current trends in the field. Also, a concise overview of the mechanisms of CYP induction is presented to aid the understanding of the induction phenomena.
Collapse
Affiliation(s)
- Jukka Hakkola
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, POB 5000, 90014, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Janne Hukkanen
- Biocenter Oulu, University of Oulu, Oulu, Finland.,Research Unit of Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Miia Turpeinen
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, POB 5000, 90014, Oulu, Finland.,Administration Center, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Olavi Pelkonen
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, POB 5000, 90014, Oulu, Finland.
| |
Collapse
|
33
|
Mao J, Jiao Z, Qiu X, Zhang M, Zhong M. Incorporating nonlinear kinetics to improve predictive performance of population pharmacokinetic models for ciclosporin in adult renal transplant recipients: A comparison of modelling strategies. Eur J Pharm Sci 2020; 153:105471. [PMID: 32682934 DOI: 10.1016/j.ejps.2020.105471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/18/2020] [Accepted: 07/15/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Ciclosporin has been shown to follow nonlinear pharmacokinetics (PK) in renal transplant recipients who received ciclosporin (NeoralⓇ, Novartis)-based triple immunosuppressive therapy. Some of these nonlinear properties have not been fully considered in population PK (popPK) analysis. Therefore, the aim of this study was to determine the potential influence of nonlinearity and the functional forms of covariates on model predictability as well as to analyse multiple nonlinear factors in the in vivo process. METHODS A total of 2969 ciclosporin whole-blood measurements, including 1328 pre-dose and 1641 2-h post-dose concentrations, were collected from 173 patients who underwent their first renal transplantation. Four popPK models based on different modelling strategies were developed to investigate the discrepancy between empirical and theory-based, linear and nonlinear compartmental kinetic models and empirical formulae on model predictability. Prediction and simulation-based diagnostics (prediction-corrected visual predictive checks) were performed to determine the stability and predictive performance of these four models. RESULTS Model predictability improved when nonlinearity was considered. The theory-based nonlinear model which incorporated nonlinear property based on known theoretical relationships performed better than the other two compartmental models. The nonlinear Michaelis-Menten model showed a remarkable improvement in predictive performance compared to the other three compartmental models. The saturated binding of ciclosporin to erythrocytes, auto-inhibition induced by the inhibitory effects of ciclosporin on cytochrome P450 3A4/P-glycoprotein may have contributed to the nonlinearity. Ciclosporin-prednisolone drug interaction should be given serious consideration in clinical settings. CONCLUSIONS Incorporation of nonlinear properties is likely to be a promising approach for improving ciclosporin model predictability. Theory-based modelling is helpful to improve model predictability. However, ciclosporin nonlinear kinetics resources need further investigation.
Collapse
Affiliation(s)
- Junjun Mao
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China
| | - Zheng Jiao
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China; Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai 200030, China.
| | - Xiaoyan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China
| | - Ming Zhang
- Department of Nephropathy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, China
| |
Collapse
|
34
|
Turolo S, Edefonti A, Ghio L, Testa S, Morello W, Montini G. CYP and SXR gene polymorphisms influence in opposite ways acute rejection rate in pediatric patients with renal transplant. BMC Pediatr 2020; 20:246. [PMID: 32450827 PMCID: PMC7249618 DOI: 10.1186/s12887-020-02152-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
Background We evaluated the role of CYP3A5, ABCB1 and SXR gene polymorphisms in the occurrence of acute kidney rejection in a cohort of pediatric renal transplant recipients. Methods Forty-nine patients were genotyped for CYP3A5, ABCB1 and SXR polymorphisms and evaluated with tacrolimus through levels in a retrospective monocenter study. Results Patients with the A allele of CYP3A5 treated with tacrolimus had a higher risk of acute rejection than those without the A allele, while patients carrying the homozygous GG variant for SXR A7635GG did not show any episode of acute rejection. Conclusion Genetic analysis of polymorphisms implicated in drug metabolism and tacrolimus trough levels may help to forecast the risk of acute rejection and individualize drug dosage in children undergoing renal transplantation.
Collapse
Affiliation(s)
- Stefano Turolo
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico UOC Nefrologia Dialisi e Trapianto pediatrico, Via della, Commenda 9, 20122, Milan, Italy.
| | - Alberto Edefonti
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico UOC Nefrologia Dialisi e Trapianto pediatrico, Via della, Commenda 9, 20122, Milan, Italy
| | - Luciana Ghio
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico UOC Nefrologia Dialisi e Trapianto pediatrico, Via della, Commenda 9, 20122, Milan, Italy
| | - Sara Testa
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico UOC Nefrologia Dialisi e Trapianto pediatrico, Via della, Commenda 9, 20122, Milan, Italy
| | - William Morello
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico UOC Nefrologia Dialisi e Trapianto pediatrico, Via della, Commenda 9, 20122, Milan, Italy
| | - Giovanni Montini
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico UOC Nefrologia Dialisi e Trapianto pediatrico, Via della, Commenda 9, 20122, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
35
|
Yuan X, Lu H, Zhao A, Ding Y, Min Q, Wang R. Transcriptional regulation of CYP3A4 by nuclear receptors in human hepatocytes under hypoxia. Drug Metab Rev 2020; 52:225-234. [PMID: 32270716 DOI: 10.1080/03602532.2020.1733004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The human hepatic cytochrome P-450 3A4 (CYP3A4), recognized as a multifunctional enzyme, has a wide range of substrates including commonly used drugs. Previous investigations demonstrated that the expression of CYP3A4 in human hepatocytes could be regulated by some nuclear receptors (NRs) at transcriptional level under diverse situations. The significance of oxygen on CYP3A4-mediated metabolism seems notable while the regulatory mode of CYP3A4 in the particular case still remains elusive. Recently, striking evidence has emerged that both CYP3A4 and its regulator NR could be inhibited by exposure to hypoxia. Therefore, it is of great importance to elucidate whether and how these NRs act in the transcriptional regulation of CYP3A4 in human hepatocytes under hypoxic conditions. In this review, we mainly summarized transcriptional regulation of the pivotal enzyme CYP3A4 by NRs and explored the possible regulatory pathways of CYP3A4 via these major NRs under hypoxia, expecting to provide favorable evidence for further clinical guidance under such pathological situations.
Collapse
Affiliation(s)
- Xuechun Yuan
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hui Lu
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Anpeng Zhao
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Yidan Ding
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qiong Min
- Pharmacy department, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Rong Wang
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
36
|
Dexamethasone simulates the anticancer effect of nano-formulated paclitaxel in breast cancer cells. Bioorg Chem 2020; 99:103792. [PMID: 32240873 DOI: 10.1016/j.bioorg.2020.103792] [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: 12/17/2019] [Revised: 02/12/2020] [Accepted: 03/22/2020] [Indexed: 02/08/2023]
Abstract
Although the chemosensitizing effect of Dexamethasone (DEX) and its ability to increase the sensitivity of breast cancer cells to chemotherapy were previously reported, this study aimed to explore how far cotreatment of breast cancer cells with paclitaxel (PTX) and DEX mimics the anticancer effect of nanoformulated PTX. To establish this goal, PTX was nanoformulated with poly (lactic-co-glycolic acid) (PLGA) and the nanoparticles (PTX-NPs) were physically authenticated. Breast cancer cells (MCF-7) were treated with PTX or PTX-NPs in presence or absence of low concentration (10 nM) of DEX. Cells viability (assessed by MTT assay), apoptosis (assessed by flow cytometry) and the expression of PTX resistance gene (TRX1) and PTX metabolizing genes (CYP2C8 and CYP3A4) were investigated. The results showed that nanoformulated PTX was validated by nano-size assessment, increased the anionic surface charge and prober conjugation with the biodegradable carrier (PLGA), as indicated by the FTIR spectroscopy. Initially, the IC50 value of PTX was 19.3 μg/ml and cotreatment with DEX minimized it to 5.22 μg/ml, whereas PTX-NPs alone inhibited cell proliferation with IC50 6.67 μg/ml. Also, in presence of DEX, PTX-NPs further decreased the IC50 to 5 μg/ml. In parallel, DEX has increased the responsiveness of cells to PTX without potentiating its apoptotic effect. Moreover, the glucocorticoid (with PTX or PTX-NPs) downregulated TXR1 gene by 26% (P < 0.01) and 28.4% (P < 0.05) respectively. Similarly, the mRNA level of CYP3A4 significantly decreased in presence of DEX. The main PTX metabolizing gene CYP2C8, in contrast, was upregulated, especially in cells cotreated with PTX/DEX (P < 0.001). Conclusively, the study reports that cotreatment of breast cancer cells with submolar concentration of DEX acts as similar as the nanoformulated PTX, possibly through its modulatory effects on the expression of the main PTX metabolizing gene (CYP2C8) and downregulating Taxol resistance gene.
Collapse
|
37
|
Smutny T, Dusek J, Hyrsova L, Nekvindova J, Horvatova A, Micuda S, Gerbal-Chaloin S, Pavek P. The 3'-untranslated region contributes to the pregnane X receptor (PXR) expression down-regulation by PXR ligands and up-regulation by glucocorticoids. Acta Pharm Sin B 2020; 10:136-152. [PMID: 31998607 PMCID: PMC6976988 DOI: 10.1016/j.apsb.2019.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/16/2022] Open
Abstract
Pregnane X receptor (PXR) is the major regulator of xenobiotic metabolism. PXR itself is controlled by various signaling molecules including glucocorticoids. Moreover, negative feed-back regulation has been proposed at the transcriptional level. We examined the involvement of the 3'-untranslated region (3'-UTR) of NR1I2 mRNA and microRNAs in PXR- and glucocorticoid receptor (GR)-mediated regulation of NR1I2 gene expression. PXR ligands were found to significantly downregulate NR1I2 mRNA expression in a set of 14 human hepatocyte cultures. Similarly, PXR was downregulated by PCN in the C57/BL6 mice liver. In mechanistic studies with the full-length 3'-UTR cloned into luciferase reporter or expression vectors, we showed that the 3'-UTR reduces PXR expression. From the miRNAs tested, miR-18a-5p inhibited both NR1I2 expression and CYP3A4 gene induction. Importantly, we observed significant upregulation of miR-18a-5p expression 6 h after treatment with the PXR ligand rifampicin, which indicates a putative mechanism underlying NR1I2 negative feed-back regulation in hepatic cells. Additionally, glucocorticoids upregulated NR1I2 expression not only through the promoter region but also via 3'-UTR regulation, which likely involves downregulation of miR-18a-5p. We conclude that miR-18a-5p is involved in the down-regulation of NR1I2 expression by its ligands and in the upregulation of NR1I2 mRNA expression by glucocorticoids in hepatic cells.
Collapse
Key Words
- 3′-UTR, 3′-untranslated region
- CAR, constitutive androstane receptor
- CYP3A4, cytochrome P450 3A4
- Cytochrome P450 3A4
- DEX, dexamethasone
- DMEs, drug metabolizing enzymes
- DMSO, dimethyl sulfoxide
- ER, estrogen receptor
- GRα, glucocorticoid receptor α
- Gene expression
- Gluc, Gaussia luciferase
- Glucocorticoid
- LBD, ligand binding domain
- MRE, miRNA-response element
- MicroRNA
- NR, nuclear receptor
- PB, phenobarbital
- PCN, pregnenolone 16α-carbonitrile
- PHHs, primary human hepatocytes
- PPARα, peroxisome proliferator-activated receptor α
- PXR, pregnane X receptor
- Pregnane X receptor
- RXRα, retinoid X receptor α
- Regulation
- Rif, rifampicin
- SEAP, secreted alkaline phosphatase
- miRNA, microRNA
Collapse
Affiliation(s)
- Tomas Smutny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove CZ-500 05, Czech Republic
| | - Jan Dusek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove CZ-500 05, Czech Republic
| | - Lucie Hyrsova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove CZ-500 05, Czech Republic
| | - Jana Nekvindova
- Institute of Clinical Biochemistry and Diagnostics, University Hospital Hradec Kralove, Hradec Kralove CZ-500 05, Czech Republic
| | - Alzbeta Horvatova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove CZ-500 05, Czech Republic
| | - Stanislav Micuda
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove CZ-500 03, Czech Republic
| | | | - Petr Pavek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove CZ-500 05, Czech Republic
| |
Collapse
|
38
|
Riddick DS, Mullen Grey AK. Mechanisms of NADPH - cytochrome P450 oxidoreductase induction by dexamethasone in the H4IIE rat hepatoma cell line. Can J Physiol Pharmacol 2019; 98:267-274. [PMID: 31825663 DOI: 10.1139/cjpp-2019-0586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Expression of NADPH - cytochrome P450 oxidoreductase (POR), electron donor for microsomal P450s, is induced in rat liver by dexamethasone (DEX), an activator of the glucocorticoid receptor (GR) and the pregnane X receptor (PXR). DEX induction of POR in rat liver is primarily PXR-mediated, although GR may contribute to mRNA effects. We examined the role of GR and PXR in the DEX induction of POR mRNA and protein in the H4IIE rat hepatoma cell line. The DEX EC50 for a PXR target, CYP3A23, exceeded that for the GR targets tyrosine aminotransferase and PXR as well as POR itself. POR protein levels were induced 3- and 4-fold, respectively, by DEX concentrations activating GR selectively (100 nM) or both GR and PXR (10 μM). POR was induced by triamcinolone acetonide, a selective GR agonist, but not pregnenolone-16α-carbonitrile, a selective PXR agonist. POR induction was blocked by the GR antagonist RU486 but minimally influenced by the PXR antagonist FLB-12. The half-life for POR mRNA was prolonged by DEX at both 100 nM and 10 μM. GR is more important in DEX-induced POR expression in H4IIE cells compared to rat liver in vivo, calling into question the suitability of this cell model for mechanistic studies.
Collapse
Affiliation(s)
- David S Riddick
- Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, ON M5S 1A8, Canada.,Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anne K Mullen Grey
- Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, ON M5S 1A8, Canada.,Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
39
|
Sachar M, Kelly EJ, Unadkat JD. Mechanisms of CYP3A Induction During Pregnancy: Studies in HepaRG Cells. AAPS JOURNAL 2019; 21:45. [PMID: 30919109 DOI: 10.1208/s12248-019-0316-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/01/2019] [Indexed: 01/06/2023]
Abstract
Activity of CYP3A, an enzyme responsible for metabolism of many marketed drugs, is induced by ~ 2-fold in pregnant women. Through studies in sandwich-cultured human hepatocytes (SCHH) and HepaRG cells, our laboratory has shown that this induction is likely mediated by the increase in cortisol plasma concentrations during pregnancy. Cortisol, at plasma concentrations observed during the third trimester (~ 800 nM), either alone or in combination with other pregnancy-related hormones, induces CYP3A activity in SCHH and HepaRG cells when cultured in dexamethasone-free media. To determine the mechanism(s) by which cortisol induces CYP3A activity, HepaRG cells were pre-incubated in dexamethasone-free medium and then incubated for 72 h with cortisol (798 nM). Glucocorticoid receptor (GR), pregnane X receptor (PXR), and CYP3A4 or CYP3A5 were knocked down using siRNA, and mRNA expression of these genes was measured. CYP3A4, and not CYP3A5, was found to be the dominant contributor to total CYP3A activity in control- and cortisol-treated HepaRG cells. Constitutive mRNA expression of CYP3A4 in HepaRG cells was regulated by both PXR and GR whereas constitutive expression of CYP3A5 in HepaRG cells was regulated by GR alone. Cortisol-mediated CYP3A4 induction in HepaRG cells was primarily mediated by GR-dependent PXR induction pathway and to a smaller extent via a PXR-independent pathway. Cortisol-mediated CYP3A5 induction was regulated by GR-dependent PXR-independent pathway. These data indicate that PXR plays a central role in cortisol-mediated induction of CYP3A activity during pregnancy and suggests that other enzymes and transporters, such as CYP2B6 and P-glycoprotein, may also be induced during pregnancy via the same mechanism(s).
Collapse
Affiliation(s)
- Madhav Sachar
- Department of Pharmaceutics, University of Washington, P.O. Box 357610, Seattle, WA, 98195, USA
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, P.O. Box 357610, Seattle, WA, 98195, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, P.O. Box 357610, Seattle, WA, 98195, USA.
| |
Collapse
|
40
|
Belderbos BPS, Hussaarts KGAM, van Harten LJ, Oomen-de Hoop E, de Bruijn P, Hamberg P, van Alphen RJ, Haberkorn BCM, Lolkema MP, de Wit R, van Soest RJ, Mathijssen RHJ. Effects of prednisone on docetaxel pharmacokinetics in men with metastatic prostate cancer: A randomized drug-drug interaction study. Br J Clin Pharmacol 2019; 85:986-992. [PMID: 30737835 DOI: 10.1111/bcp.13889] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 12/24/2022] Open
Abstract
AIMS Docetaxel has been approved for the treatment of metastatic prostate cancer in combination with prednisone. Since prednisone is known to induce the cytochrome P450 iso-enzyme CYP3A4, which is the main metabolizing enzyme of docetaxel in the liver, a potential drug-drug interaction may occur. In this prospective randomized pharmacokinetic cross-over study we investigated docetaxel exposure with concomitant prednisone, compared to docetaxel monotherapy in men with metastatic prostate cancer. METHODS Patients scheduled to receive at least 6 cycles of docetaxel (75 mg/m2 ) and who gave written informed consent were randomized to receive either the 1st 3 cycles, or the last 3 consecutive cycles with prednisone (twice daily 5 mg). Pharmacokinetic blood sampling was performed during cycle 3 and cycle 6. Primary endpoint was difference in docetaxel exposure, calculated as area under the curve (AUC0-inf ) and analysed by means of a linear mixed model. Given the cross-over design the study was powered on 18 patients to answer the primary, pharmacokinetic, endpoint. RESULTS Eighteen evaluable patients were included in the trial. Docetaxel concentration with concomitant prednisone (AUC0-inf 2784 ng*h/mL, 95% confidence interval 2436-3183 ng*h/mL) was similar to the concentration of docetaxel monotherapy (AUC0-inf 2647 ng*h/mL, 95% confidence interval 2377-2949 ng*h/mL). Exploratory analysis showed no toxicity differences between docetaxel monotherapy and docetaxel cycles with prednisone. CONCLUSION No significant difference in docetaxel concentrations was observed. In addition, we found similar toxicity profiles in absence and presence of prednisone. Therefore, from a pharmacokinetic point of view, docetaxel may be administrated with or without prednisone.
Collapse
Affiliation(s)
- Bodine P S Belderbos
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Koen G A M Hussaarts
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Leonie J van Harten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Peter de Bruijn
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Robbert J van Alphen
- Department of Internal Medicine, Elisabeth Twee Steden Ziekenhuis, Tilburg, The Netherlands
| | | | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Robert J van Soest
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
41
|
Abbott KL, Chaudhury CS, Chandran A, Vishveshwara S, Dvorak Z, Jiskrova E, Poulikova K, Vyhlidalova B, Mani S, Pondugula SR. Belinostat, at Its Clinically Relevant Concentrations, Inhibits Rifampicin-Induced CYP3A4 and MDR1 Gene Expression. Mol Pharmacol 2019; 95:324-334. [PMID: 30622215 PMCID: PMC6362450 DOI: 10.1124/mol.118.114587] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/03/2019] [Indexed: 01/26/2023] Open
Abstract
Activation of human pregnane X receptor (hPXR) has been associated with induction of chemoresistance. It has been proposed that such chemoresistance via cytochrome P450/drug transporters can be reversed with the use of antagonists that specifically abrogate agonist-mediated hPXR activation. Unfortunately, proposed antagonists lack the specificity and appropriate pharmacological characteristics that allow these features to be active in the clinic. We propose that, ideally, an hPXR antagonist would be a cancer drug itself that is part of a "cancer drug cocktail" and effective as an hPXR antagonist at therapeutic concentrations. Belinostat (BEL), a histone deacetylase inhibitor approved for the treatment of relapsed/refractory peripheral T-cell lymphoma, and often used in combination with chemotherapy, is an attractive candidate based on its hPXR ligand-like features. We sought to determine whether these features of BEL might allow it to behave as an antagonist in combination chemotherapy regimens that include hPXR activators. BEL represses agonist-activated hPXR target gene expression at its therapeutic concentrations in human primary hepatocytes and LS174T human colon cancer cells. BEL repressed rifampicin-induced gene expression of CYP3A4 and multidrug resistance protein 1, as well as their respective protein activities. BEL decreased rifampicin-induced resistance to SN-38, the active metabolite of irinotecan, in LS174T cells. This finding indicates that BEL could suppress hPXR agonist-induced chemoresistance. BEL attenuated the agonist-induced steroid receptor coactivator-1 interaction with hPXR, and, together with molecular docking studies, the study suggests that BEL directly interacts with multiple sites on hPXR. Taken together, our results suggest that BEL, at its clinically relevant therapeutic concentration, can antagonize hPXR agonist-induced gene expression and chemoresistance.
Collapse
Affiliation(s)
- Kodye L Abbott
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Chloe S Chaudhury
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Aneesh Chandran
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Saraswathi Vishveshwara
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Zdenek Dvorak
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Eva Jiskrova
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Karolina Poulikova
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Barbora Vyhlidalova
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Sridhar Mani
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| | - Satyanarayana R Pondugula
- Department of Anatomy, Physiology and Pharmacology (K.L.A., C.S.C., S.R.P.) and Auburn University Research Initiative in Cancer (K.L.A., C.S.C., S.R.P.), Auburn University, Auburn, Alabama; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India (A.C., S.V.); Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Olomouc, Czech Republic (Z.D., E.J., K.P., B.V.); and Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York (S.M.)
| |
Collapse
|
42
|
Daniel E, Digweed D, Quirke J, Voet B, Ross RJ, Davies M. Hydrocortisone Granules Are Bioequivalent When Sprinkled Onto Food or Given Directly on the Tongue. J Endocr Soc 2019; 3:847-856. [PMID: 30993254 PMCID: PMC6457279 DOI: 10.1210/js.2018-00380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/22/2019] [Indexed: 12/02/2022] Open
Abstract
Background Immediate-release hydrocortisone granules in capsules for opening in pediatric-appropriate doses have recently been licensed for children with adrenal insufficiency. This study evaluated the bioavailability of hydrocortisone granules administered as sprinkles onto soft food and yogurt compared with direct administration to the back of the tongue. Methods Randomized, 3-period crossover study in 18 dexamethasone-suppressed healthy men. In each period, the fasted participants received 5 mg hydrocortisone granules either directly to the back of the tongue or sprinkled onto soft food (applesauce), or yogurt, followed by 240 mL of water. Serum cortisol was measured by liquid chromatography tandem mass spectometry. Results The cortisol geometric mean maximum concentration (Cmax) and area under the curve (AUC) for direct administration, sprinkles onto yogurt, and sprinkles onto soft food were: Cmax 428, 426, 427 nmol/L and AUC0-inf 859, 886, 844 h × nmol/L, and AUC0-t 853, 882, 838 h × nmol/L respectively. The 90% CI for the ratios of Cmax, AUC0-inf and AUC0-t for administration with soft food or yogurt to direct administration were well within the bioequivalent range, 80% to 125%. Median time to Cmax (Tmax) was similar between methods of administration: 0.63 hours administered directly, 0.75 hours on soft food and 0.75 hours on yogurt. No adverse events occurred during the study. Conclusions Hydrocortisone granules administered as sprinkles onto soft food or yogurt but not mixed with these foods are bioequivalent to those administered directly to the back of the tongue. Carers, parents, or patients may choose to administer hydrocortisone granules either directly or sprinkled onto soft food or yogurt.
Collapse
Affiliation(s)
- Eleni Daniel
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Dena Digweed
- Diurnal Ltd, Cardiff Medicentre, Cardiff, United Kingdom
| | - Jo Quirke
- Diurnal Ltd, Cardiff Medicentre, Cardiff, United Kingdom
| | - Bernard Voet
- Diurnal Ltd, Cardiff Medicentre, Cardiff, United Kingdom
| | - Richard J Ross
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Madhu Davies
- Diurnal Ltd, Cardiff Medicentre, Cardiff, United Kingdom
| |
Collapse
|
43
|
Pharmacokinetic interaction of calcitriol with 20(S)-protopanaxadiol in mice: Determined by LC/MS analysis. Eur J Pharm Sci 2019; 130:173-180. [PMID: 30654110 DOI: 10.1016/j.ejps.2019.01.016] [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] [Received: 09/24/2018] [Revised: 12/23/2018] [Accepted: 01/11/2019] [Indexed: 12/31/2022]
Abstract
The physiological and anti-cancer functions of vitamin D3 are accomplished primarily via 1α,25-dihydroxyvitamin D3 (calcitriol), whereas 20(S)-protopanaxadiol (aPPD) is a ginsenoside, which is isolated from Panax ginseng, with potential anti-cancer benefits. In the present study, we report a pharmacokinetic (PK) herb-nutrient interaction between calcitriol and aPPD in mice. A liquid chromatography mass spectrometry (LC/MS) method was developed using 4-phenyl-1,2,4-triazoline-3,5-dione derivatizing agent and we subsequently used the method to quantitate calcitriol in mouse serum. The limit of quantitation was 0.01 ng/ml which is approximately 100 fold lower than the previously reported assay from our laboratory. Calcitriol PK parameters were determined in non-tumor-bearing or C4-2 human prostate tumor-bearing nude mice following oral co-administration of calcitriol either alone or in combination with aPPD. Mice were pretreated with oral aPPD (70 mg/kg) or vehicle control twice daily for seven consecutive days, followed by a single oral dose of 4 μg/kg calcitriol alone or in combination with aPPD. Our PK results demonstrated that co-administration of calcitriol with aPPD (following pre-treatment with vehicle for seven days) resulted in a 35% increase in the area under the curve (AUC0-24 h) and a 41% increase in the maximum serum concentration (Cmax) compared to the calcitriol only group. aPPD therefore significantly increased calcitriol serum exposure. We also saw a reduction in the time required to reach Cmax. In contrast, calcitriol PK in mice co-administered with calcitriol and aPPD as well as those pretreated seven consecutive days with aPPD was no different than that determined for the mice that received vehicle for seven days as pre-treatment. Co-administration of calcitriol with aPPD therefore could increase health benefits of vitamin D3, however any increased risk of hypercalcemia, resulting from this combination approach, requires further investigation. Lastly, we surmise that a cytochrome P450 inhibition-based mechanism may contribute to the observed PK interaction.
Collapse
|
44
|
Identification of genetic variants associated with tacrolimus metabolism in kidney transplant recipients by extreme phenotype sampling and next generation sequencing. THE PHARMACOGENOMICS JOURNAL 2018; 19:375-389. [PMID: 30442921 PMCID: PMC6522337 DOI: 10.1038/s41397-018-0063-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/11/2018] [Accepted: 09/27/2018] [Indexed: 12/26/2022]
Abstract
An extreme phenotype sampling (EPS) model with targeted next-generation sequencing (NGS) identified genetic variants associated with tacrolimus (Tac) metabolism in subjects from the Deterioration of Kidney Allograft Function (DeKAF) Genomics cohort which included 1,442 European Americans (EA) and 345 African Americans (AA). This study included 48 subjects separated into 4 groups of 12 (AA high, AA low, EA high, EA low). Groups were selected by the extreme phenotype of dose-normalized Tac trough concentrations after adjusting for common genetic variants and clinical factors. NGS spanned >3 Mb of 28 genes and identified 18,661 genetic variants (3,961 previously unknown). A group of 125 deleterious variants, by SIFT analysis, were associated with Tac troughs in EAs (burden test, p=0.008), CYB5R2 was associated with Tac troughs in AAs (SKAT, p=0.00079). In CYB5R2, rs61733057 (increased allele frequency in AAs) was predicted to disrupt protein function by SIFT and PolyPhen2 analysis. The variants merit further validation.
Collapse
|
45
|
Wang X, Wang F, Lu Z, Jin X, Zhang Y. Semi-quantitative profiling of bile acids in serum and liver reveals the dosage-related effects of dexamethasone on bile acid metabolism in mice. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1095:65-74. [DOI: 10.1016/j.jchromb.2018.07.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 07/06/2018] [Accepted: 07/15/2018] [Indexed: 12/11/2022]
|
46
|
Prasad B, Bhatt DK, Johnson K, Chapa R, Chu X, Salphati L, Xiao G, Lee C, Hop CECA, Mathias A, Lai Y, Liao M, Humphreys WG, Kumer SC, Unadkat JD. Abundance of Phase 1 and 2 Drug-Metabolizing Enzymes in Alcoholic and Hepatitis C Cirrhotic Livers: A Quantitative Targeted Proteomics Study. Drug Metab Dispos 2018; 46:943-952. [PMID: 29695616 PMCID: PMC5987995 DOI: 10.1124/dmd.118.080523] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/13/2018] [Indexed: 01/12/2023] Open
Abstract
To predict the impact of liver cirrhosis on hepatic drug clearance using physiologically based pharmacokinetic (PBPK) modeling, we compared the protein abundance of various phase 1 and phase 2 drug-metabolizing enzymes (DMEs) in S9 fractions of alcoholic (n = 27) or hepatitis C (HCV, n = 30) cirrhotic versus noncirrhotic (control) livers (n = 25). The S9 total protein content was significantly lower in alcoholic or HCV cirrhotic versus control livers (i.e., 38.3 ± 8.3, 32.3 ± 12.8, vs. 51.1 ± 20.7 mg/g liver, respectively). In general, alcoholic cirrhosis was associated with a larger decrease in the DME abundance than HCV cirrhosis; however, only the abundance of UGT1A4, alcohol dehydrogenase (ADH)1A, and ADH1B was significantly lower in alcoholic versus HCV cirrhotic livers. When normalized to per gram of tissue, the abundance of nine DMEs (UGT1A6, UGT1A4, CYP3A4, UGT2B7, CYP1A2, ADH1A, ADH1B, aldehyde oxidase (AOX)1, and carboxylesterase (CES)1) in alcoholic cirrhosis and five DMEs (UGT1A6, UGT1A4, CYP3A4, UGT2B7, and CYP1A2) in HCV cirrhosis was <25% of that in control livers. The abundance of most DMEs in cirrhotic livers was 25% to 50% of control livers. CES2 abundance was not affected by cirrhosis. Integration of UGT2B7 abundance in cirrhotic livers into the liver cirrhosis (Child Pugh C) model of Simcyp improved the prediction of zidovudine and morphine PK in subjects with Child Pugh C liver cirrhosis. These data demonstrate that protein abundance data, combined with PBPK modeling and simulation, can be a powerful tool to predict drug disposition in special populations.
Collapse
Affiliation(s)
- Bhagwat Prasad
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Deepak Kumar Bhatt
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Katherine Johnson
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Revathi Chapa
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Xiaoyan Chu
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Laurent Salphati
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Guangqing Xiao
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Caroline Lee
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Cornelis E C A Hop
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Anita Mathias
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Yurong Lai
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Mingxiang Liao
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - William G Humphreys
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Sean C Kumer
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| | - Jashvant D Unadkat
- University of Washington, Seattle, Washington (B.P., D.K.B., K.J., R.C., J.D.U.); Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C.); Gilead Sciences, Inc., Foster City, California (A.S.R., A.M.); Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Biogen, Cambridge, Massachusetts (G.X.); Ardea Biosciences, Inc., San Diego, California (C.L.); Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., W.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and University of Kansas Medical Center, Kansas City, Kansas (S.C.K.)
| |
Collapse
|
47
|
Chen C, Soto-Gutierrez A, Baptista PM, Spee B. Biotechnology Challenges to In Vitro Maturation of Hepatic Stem Cells. Gastroenterology 2018; 154:1258-1272. [PMID: 29428334 PMCID: PMC6237283 DOI: 10.1053/j.gastro.2018.01.066] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/05/2018] [Accepted: 01/10/2018] [Indexed: 12/16/2022]
Abstract
The incidence of liver disease is increasing globally. The only curative therapy for severe end-stage liver disease, liver transplantation, is limited by the shortage of organ donors. In vitro models of liver physiology have been developed and new technologies and approaches are progressing rapidly. Stem cells might be used as a source of liver tissue for development of models, therapies, and tissue-engineering applications. However, we have been unable to generate and maintain stable and mature adult liver cells ex vivo. We review factors that promote hepatocyte differentiation and maturation, including growth factors, transcription factors, microRNAs, small molecules, and the microenvironment. We discuss how the hepatic circulation, microbiome, and nutrition affect liver function, and the criteria for considering cells derived from stem cells to be fully mature hepatocytes. We explain the challenges to cell transplantation and consider future technologies for use in hepatic stem cell maturation, including 3-dimensional biofabrication and genome modification.
Collapse
Affiliation(s)
- Chen Chen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; The Royal Netherlands Academy of Arts and Sciences, Hubrecht Institute and University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Pedro M Baptista
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain; Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas, Madrid, Spain; Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain; Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain; Department of Biomedical and Aerospace Engineering, Universidad Carlos III de Madrid, Madrid, Spain
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
48
|
Taneja G, Chu C, Maturu P, Moorthy B, Ghose R. Role of c-Jun-N-Terminal Kinase in Pregnane X Receptor-Mediated Induction of Human Cytochrome P4503A4 In Vitro. Drug Metab Dispos 2018; 46:397-404. [PMID: 29440179 DOI: 10.1124/dmd.117.079160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/01/2018] [Indexed: 12/17/2022] Open
Abstract
Cytochrome P450 CYP3A4 is the most abundant drug-metabolizing enzyme and is responsible for the metabolism of ∼50% of clinically available drugs. Induction of CYP3A4 impacts the disposition of its substrates and leads to harmful clinical consequences, such as failure of therapy. To prevent such undesirable consequences, the molecular mechanisms of regulation of CYP3A4 need to be fully understood. CYP3A4 induction is regulated primarily by the xenobiotic nuclear receptor pregnane-X receptor (PXR). After ligand binding, PXR is translocated to the nucleus, where it binds to the CYP3A4 promoter and induces its gene expression. PXR function is modulated by phosphorylation(s) by multiple kinases. In this study, we determined the role of the c-Jun N-terminal kinase (JNK) in PXR-mediated induction of CYP3A4 enzyme in vitro. Human liver carcinoma cells (HepG2) were transfected with CYP3A4 luciferase and PXR plasmids, followed by treatment with JNK inhibitor (SP600125; SP) and PXR activators rifampicin (RIF) or hyperforin. Our results indicate that SP treatment significantly attenuated PXR-mediated induction of CYP3A4 reporter activity, as well as gene expression and enzyme activity. JNK knockdown by siRNA (targeting both JNK 1 and 2) also attenuated CYP3A4 induction by RIF. Interestingly, SP treatment attenuated JNK activation by RIF. Furthermore, treatment with RIF increased PXR nuclear levels and binding to the CYP3A4 promoter; SP attenuated these effects. This study shows that JNK is a novel mechanistic regulator of CYP3A4 induction by PXR.
Collapse
Affiliation(s)
- Guncha Taneja
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| | - Chun Chu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| | - Paramahamsa Maturu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| | - Bhagavatula Moorthy
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| |
Collapse
|
49
|
Jonsson-Schmunk K, Schafer SC, Croyle MA. Impact of nanomedicine on hepatic cytochrome P450 3A4 activity: things to consider during pre-clinical and clinical studies. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0376-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
50
|
Analysis of common polymorphisms within NR1I2 and NR1I3 genes and tacrolimus dose-adjusted concentration in stable kidney transplant recipients. Pharmacogenet Genomics 2017; 27:372-377. [DOI: 10.1097/fpc.0000000000000301] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|