101
|
Mota C, Coelho C, Leimkühler S, Garattini E, Terao M, Santos-Silva T, Romão MJ. Critical overview on the structure and metabolism of human aldehyde oxidase and its role in pharmacokinetics. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
102
|
Wu L, Han W, Chen Y, Zhang T, Liu J, Zhong S, Liu H, Han C, Zhang Z, Liu S, Tang L. Gender Differences in the Hepatotoxicity and Toxicokinetics of Emodin: The Potential Mechanisms Mediated by UGT2B7 and MRP2. Mol Pharm 2018; 15:3931-3945. [PMID: 30011215 DOI: 10.1021/acs.molpharmaceut.8b00387] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Emodin is a main anthraquinone compound which exists in Chinese traditional medicines including Polygonum multiflorum and Rhubarb. It is documented to have obvious liver and kidney toxicity. This study aims to (a) estimate gender differences of the hepatotoxicity and toxicokinetics in rats after oral administration of emodin (60 and 150 mg/kg/d) for a consecutive 28 days and (b) clarify relative mechanisms caused by glucuronidation and disposition. Hepatotoxicity was significantly higher in female rats than that in male rats, as evidenced by histopathological and biochemical tests. Similarly, the toxicokinetic profiles of emodin have time and gender differences, which could cause time and gender differences in hepatotoxicity. The metabolic and transcriptomics data of 55 human liver and 36 human kidney samples demonstrated that UDP-glucuronosyltransferase 2B7 (UGT2B7) was the predominant enzyme for emodin glucuronidation. A genome-wide association study (GWAS) identified that rs11726899 located within ∼50 kb of the transcript of UGT2B could significantly affect emodin metabolism. Knockdown of UGT2B7 in HepG2 cells significantly decreased emodin glucuronidation and increased cytotoxicity of emodin. The gene expression and protein levels of UGT2B7 were decreased, but those of the multidrug-resistant-protein 2 (MRP2) were increased in HepG2 cells after being treated with 50 μM emodin for 48 h. Long-term use of emodin could decrease the intrinsic clearance (CLint, decreased by 18.5%-35.4%) values of zidovidue (UGT2B7 substrate) glucuronide in both male and female liver microsomes from rats administrated with emodin for 28 days, thus causing the accumulation of emodin. However, higher self-induced MRP2 expression and lower hepatotoxicity were observed in emodin-treated male rats compared to that in female rats. Therefore, gender differences in the hepatotoxicity and toxicokinetics of emodin are potentially mediated by the coupling of UGT2B7 and MRP2 in vivo.
Collapse
Affiliation(s)
- Lili Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology , Southern Medical University , Guangzhou 510515 , China.,Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Weichao Han
- Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Yulian Chen
- Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Tao Zhang
- Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Junjin Liu
- Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Shilong Zhong
- Medical Research Center of Guangdong General Hospital , Guangdong Academy of Medical Sciences , Guangzhou 510515 , China
| | - Han Liu
- Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Congcong Han
- Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Zhongyi Zhang
- Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Shuwen Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology , Southern Medical University , Guangzhou 510515 , China.,Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| | - Lan Tang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology , Southern Medical University , Guangzhou 510515 , China.,Biopharmaceutics, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou 510515 , China
| |
Collapse
|
103
|
Lv X, Zhang JB, Wang XX, Hu WZ, Shi YS, Liu SW, Hao DC, Zhang WD, Ge GB, Hou J, Yang L. Amentoflavone is a potent broad-spectrum inhibitor of human UDP-glucuronosyltransferases. Chem Biol Interact 2018; 284:48-55. [DOI: 10.1016/j.cbi.2018.02.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 02/03/2018] [Accepted: 02/12/2018] [Indexed: 11/25/2022]
|
104
|
Wang Y, Yu YX, Luan Y, An J, Yin DG, Zhang XY. Bioactivation of 1-chloro-2-hydroxy-3-butene, an in vitro metabolite of 1,3-butadiene, by rat liver microsomes. Chem Biol Interact 2018; 282:36-44. [DOI: 10.1016/j.cbi.2018.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 12/03/2017] [Accepted: 01/05/2018] [Indexed: 01/15/2023]
|
105
|
den Braver-Sewradj SP, den Braver MW, Toorneman RM, van Leeuwen S, Zhang Y, Dekker SJ, Vermeulen NPE, Commandeur JNM, Vos JC. Reduction and Scavenging of Chemically Reactive Drug Metabolites by NAD(P)H:Quinone Oxidoreductase 1 and NRH:Quinone Oxidoreductase 2 and Variability in Hepatic Concentrations. Chem Res Toxicol 2018; 31:116-126. [PMID: 29281794 PMCID: PMC5997408 DOI: 10.1021/acs.chemrestox.7b00289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
Detoxicating
enzymes NAD(P)H:quinone oxidoreductase 1 (NQO1) and
NRH:quinone oxidoreductase 2 (NQO2) catalyze the two-electron reduction
of quinone-like compounds. The protective role of the polymorphic
NQO1 and NQO2 enzymes is especially of interest in the liver as the
major site of drug bioactivation to chemically reactive drug metabolites.
In the current study, we quantified the concentrations of NQO1 and
NQO2 in 20 human liver donors and NQO1 and NQO2 activities with quinone-like
drug metabolites. Hepatic NQO1 concentrations ranged from 8 to 213
nM. Using recombinant NQO1, we showed that low nM concentrations of
NQO1 are sufficient to reduce synthetic amodiaquine and carbamazepine
quinone-like metabolites in vitro. Hepatic NQO2 concentrations
ranged from 2 to 31 μM. NQO2 catalyzed the reduction of quinone-like
metabolites derived from acetaminophen, clozapine, 4′-hydroxydiclofenac,
mefenamic acid, amodiaquine, and carbamazepine. The reduction of the
clozapine nitrenium ion supports association studies showing that
NQO2 is a genetic risk factor for clozapine-induced agranulocytosis.
The 5-hydroxydiclofenac quinone imine, which was previously shown
to be reduced by NQO1, was not reduced by NQO2. Tacrine was identified
as a potent NQO2 inhibitor and was applied to further confirm the
catalytic activity of NQO2 in these assays. While the in vivo relevance of NQO2-catalyzed reduction of quinone-like metabolites
remains to be established by identification of the physiologically
relevant co-substrates, our results suggest an additional protective
role of the NQO2 protein by non-enzymatic scavenging of quinone-like
metabolites. Hepatic NQO1 activity in detoxication of quinone-like
metabolites becomes especially important when other detoxication pathways
are exhausted and NQO1 levels are induced.
Collapse
Affiliation(s)
- Shalenie P den Braver-Sewradj
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Michiel W den Braver
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Robin M Toorneman
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Stephanie van Leeuwen
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Yongjie Zhang
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Stefan J Dekker
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Nico P E Vermeulen
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Jan N M Commandeur
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - J Chris Vos
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
106
|
Gong Y, Yuan Y. Resistance mechanisms of Helicobacter pylori and its dual target precise therapy. Crit Rev Microbiol 2018; 44:371-392. [PMID: 29293032 DOI: 10.1080/1040841x.2017.1418285] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Helicobacter pylori drug resistance presents a significant challenge to the successful eradication of this pathogen. To find strategies to improve the eradication efficacy of H. pylori, it is necessary to clarify the resistance mechanisms involved. The mechanisms of H. pylori drug resistance can be investigated from two angles: the pathogen and the host. A comprehensive understanding of the molecular mechanisms of H. pylori resistance based on both pathogen and host would aid the implementation of precise therapy, or ideally "dual target precise therapy" (bacteria and host-specific target therapy). In recent years, with increased understanding of the mechanisms of H. pylori resistance, the focus of eradication has shifted from disease-specific to patient-specific treatment. The implementation of "precision medicine" has also provided a new perspective on the treatment of infectious diseases. In this article, we systematically review current research on H. pylori drug resistance from the perspective of both the pathogen and the host. We also review therapeutic strategies targeted to pathogen and host factors that are aimed at achieving precise treatment of H. pylori.
Collapse
Affiliation(s)
- Yuehua Gong
- a Tumor Etiology and Screening Department of Cancer Institute and General Surgery , the First Hospital of China Medical University , Shenyang , China.,b Key Laboratory of Cancer Etiology and Prevention (China Medical University) Liaoning Provincial Education Department , Shenyang , China.,c National Clinical Research Center for Digestive Diseases , Xi'an , China
| | - Yuan Yuan
- a Tumor Etiology and Screening Department of Cancer Institute and General Surgery , the First Hospital of China Medical University , Shenyang , China.,b Key Laboratory of Cancer Etiology and Prevention (China Medical University) Liaoning Provincial Education Department , Shenyang , China.,c National Clinical Research Center for Digestive Diseases , Xi'an , China
| |
Collapse
|
107
|
In vitro metabolism study of a novel P38 kinase inhibitor: in silico predictions, structure elucidation using MS/MS-I. Future Med Chem 2018; 10:201-220. [DOI: 10.4155/fmc-2017-0126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Metabolism study of PH-797804, a promising newly developed drug for treatment of chronic inflammation which inhibits P38 mitogen-activated protein kinase. Materials & methods: Susceptibility of PH-797804 to metabolism was first investigated using SMARTCyp and Xenosite web servers. Molecular docking of the drug into CYP3A4 crystal structures evaluated binding interactions with active site. The predicted results were confirmed by in vitro incubation with rat S9 fraction. Metabolites of PH-797804 were identified by MS/MS. Results: A hydroxy metabolite and a cysteine/glutathione conjugate were detected. Computational prediction of reactive site of PH-797804 was conducted. Conclusion: The probable cysteine/glutathione adduct is indicative of potential drug chemical reactivity with potential to damage DNA and may provide guidance to the design of analogs with minimum toxicity.
Collapse
|
108
|
Cruciani G, Milani N, Benedetti P, Lepri S, Cesarini L, Baroni M, Spyrakis F, Tortorella S, Mosconi E, Goracci L. From Experiments to a Fast Easy-to-Use Computational Methodology to Predict Human Aldehyde Oxidase Selectivity and Metabolic Reactions. J Med Chem 2017; 61:360-371. [DOI: 10.1021/acs.jmedchem.7b01552] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Gabriele Cruciani
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
| | - Nicolò Milani
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
| | - Paolo Benedetti
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
| | - Susan Lepri
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
| | - Lucia Cesarini
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
| | - Massimo Baroni
- Molecular Discovery Ltd, Centennial
Park, Borehamwood, Hertfordshire, United Kingdom
| | - Francesca Spyrakis
- Department
of Drug Science and Technology, University of Turin, via P. Giuria
9, 10125 Turin, Italy
| | - Sara Tortorella
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
- Molecular Horizon srl, via Montelino
32, 06084 Bettona, Italy
| | - Edoardo Mosconi
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
- Computational
Laboratory for Hybrid/Organic Photovoltaics, National Research Council−Institute of Molecular Science and Technologies, Via Elce
di Sotto 8, I-06123 Perugia, Italy
| | - Laura Goracci
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy
- Consortium for Computational Molecular and Materials Sciences (CMS), via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
109
|
Forkuo AD, Ansah C, Pearson D, Gertsch W, Cirello A, Amaral A, Spear J, Wright CW, Rynn C. Identification of cryptolepine metabolites in rat and human hepatocytes and metabolism and pharmacokinetics of cryptolepine in Sprague Dawley rats. BMC Pharmacol Toxicol 2017; 18:84. [PMID: 29273084 PMCID: PMC5741962 DOI: 10.1186/s40360-017-0188-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 12/05/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND This study aims at characterizing the in vitro metabolism of cryptolepine using human and rat hepatocytes, identifying metabolites in rat plasma and urine after a single cryptolepine dose, and evaluating the single-dose oral and intravenous pharmacokinetics of cryptolepine in male Sprague Dawley (SD) rats. METHODS The in vitro metabolic profiles of cryptolepine were determined by LC-MS/MS following incubation with rat and human hepatocytes. The in vivo metabolic profile of cryptolepine was determined in plasma and urine samples from Sprague Dawley rats following single-dose oral administration of cryptolepine. Pharmacokinetic parameters of cryptolepine were determined in plasma and urine from Sprague Dawley rats after single-dose intravenous and oral administration. RESULTS Nine metabolites were identified in human and rat hepatocytes, resulting from metabolic pathways involving oxidation (M2-M9) and glucuronidation (M1, M2, M4, M8, M9). All human metabolites were found in rat hepatocyte incubations except glucuronide M1. Several metabolites (M2, M6, M9) were also identified in the urine and plasma of rats following oral administration of cryptolepine. Unchanged cryptolepine detected in urine was negligible. The Pharmacokinetic profile of cryptolepine showed a very high plasma clearance and volume of distribution (Vss) resulting in a moderate average plasma half-life of 4.5 h. Oral absorption was fast and plasma exposure and oral bioavailability were low. CONCLUSIONS Cryptolepine metabolism is similar in rat and human in vitro with the exception of direct glucuronidation in human. Clearance in rat and human is likely to include a significant metabolic contribution, with proposed primary human metabolism pathways hydroxylation, dihydrodiol formation and glucuronidation. Cryptolepine showed extensive distribution with a moderate half-life.
Collapse
Affiliation(s)
- Arnold Donkor Forkuo
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Science, College of Health Sciences Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.
| | - Charles Ansah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Science, College of Health Sciences Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - David Pearson
- Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Postfach, CH-4002, Basel, Switzerland
| | - Werner Gertsch
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Postfach, CH-4002, Basel, Switzerland
| | - Amanda Cirello
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research, 250 Massachusetts Ave Cambridge, 02139, Cambridge, MA, USA
| | - Adam Amaral
- Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, 250 Massachusetts Ave Cambridge, 02139, Cambridge, MA, USA
| | - Jaimie Spear
- Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, 250 Massachusetts Ave Cambridge, 02139, Cambridge, MA, USA
| | - Colin W Wright
- School of Pharmacy, University of Bradford, West Yorkshire, BD7 1DP, Bradford, USA
| | - Caroline Rynn
- Metabolism and Pharmacokinetics, Novartis Institute for BioMedical Research, Novartis Pharma AG, Postfach, CH-4002, Basel, Switzerland
| |
Collapse
|
110
|
den Braver-Sewradj SP, den Braver MW, Baze A, Decorde J, Fonsi M, Bachellier P, Vermeulen NPE, Commandeur JNM, Richert L, Vos JC. Direct comparison of UDP-glucuronosyltransferase and cytochrome P450 activities in human liver microsomes, plated and suspended primary human hepatocytes from five liver donors. Eur J Pharm Sci 2017; 109:96-110. [PMID: 28778465 DOI: 10.1016/j.ejps.2017.07.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 11/26/2022]
Abstract
UDP-glucuronosyltransferases (UGTs) and cytochrome P450s (CYPs) are the major enzymes involved in hepatic metabolism of drugs. Hepatic drug metabolism is commonly investigated using human liver microsomes (HLM) or primary human hepatocytes (PHH). We describe the development of a sensitive assay to phenotype activities of six major hepatic UGT isoforms (UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9 and UGT2B7) in intact PHH by analysis of glucuronidation of selective probe substrates. The non-selective, general substrate 7-hydroxycoumarin was included for comparison. For each liver donor preparation (five donors) UGT activities in cryopreserved suspended and plated PHH were compared to HLM prepared from the same donors. Standard CYP reaction phenotyping of seven major isoforms was performed in parallel. For all donors, CYP- and UGT-isoforms activity profiles were comparable in PHH and HLM, indicating that reaction phenotyping with selective probe substrates in intact cells primarily reflects respective CYP or UGT activity. System-dependent effects on UGT and CYP isoform activity were still found. While UGT activity of UGT1A1 was equivalent in plated and suspended PHH, UGT1A3, UGT1A6 and UGT2B7 activity was higher in suspended PHH and UGT1A9 and UGT1A4 activity was higher in plated PHH. The well-known decrease in activity of most CYP isoforms in plated compared to suspended PHH was confirmed. Importantly, we found a significant loss in CYP2C19 and CYP2B6 in HLM, activity being lower than in intact cells. Taken together, these findings implicate that, dependent on the UGT or CYP isoforms involved in the metabolism of a given compound, the outcome of metabolic assays is strongly dependent on the choice of the in vitro system. The currently described UGT- and CYP- activity profiling method can be used as a standard assay in intact cells and can especially aid in reaction phenotyping of in vitro systems for which a limited number of cells are available.
Collapse
Affiliation(s)
- Shalenie P den Braver-Sewradj
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Michiel W den Braver
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Audrey Baze
- Kaly-Cell, 20A Rue du Général Leclerc, Plobsheim, France; UNISTRA, 4 Rue Blaise Pascal, Strasbourg, France
| | | | | | - Philippe Bachellier
- UNISTRA, 4 Rue Blaise Pascal, Strasbourg, France; Centre de Chirurgie Viscérale et de Transplantation, Hôpital de Hautepierre, 67098 Strasbourg, France
| | - Nico P E Vermeulen
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Jan N M Commandeur
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Lysiane Richert
- Kaly-Cell, 20A Rue du Général Leclerc, Plobsheim, France; PEPITE EA4267, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France.
| | - J Chris Vos
- AIMMS-Division of Molecular Toxicology, Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, O
- 2 building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands.
| |
Collapse
|
111
|
Järvinen E, Troberg J, Kidron H, Finel M. Selectivity in the Efflux of Glucuronides by Human Transporters: MRP4 Is Highly Active toward 4-Methylumbelliferone and 1-Naphthol Glucuronides, while MRP3 Exhibits Stereoselective Propranolol Glucuronide Transport. Mol Pharm 2017; 14:3299-3311. [DOI: 10.1021/acs.molpharmaceut.7b00366] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Erkka Järvinen
- Division
of Pharmaceutical Chemistry and Technology,
and ‡Division of Pharmaceutical
Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Johanna Troberg
- Division
of Pharmaceutical Chemistry and Technology,
and ‡Division of Pharmaceutical
Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Heidi Kidron
- Division
of Pharmaceutical Chemistry and Technology,
and ‡Division of Pharmaceutical
Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Moshe Finel
- Division
of Pharmaceutical Chemistry and Technology,
and ‡Division of Pharmaceutical
Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| |
Collapse
|
112
|
Govendir M. Review of some pharmacokinetic and pharmacodynamic properties of anti-infective medicines administered to the koala (Phascolarctos cinereus). J Vet Pharmacol Ther 2017; 41:1-10. [PMID: 28703410 DOI: 10.1111/jvp.12435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/04/2017] [Indexed: 01/12/2023]
Abstract
Although koalas are iconic Australian animals, no pharmacokinetic studies of any first-line medicines used to treat diseased or injured koalas had been published prior to 2010. Traditionally, medicine dosages suggested for this species underwent linear extrapolation from those recommended for domesticated species. The koala, a specialist folivore whose natural diet consists of almost exclusively Eucalyptus spp. foliage has anatomical and physiological adaptations for detoxifying their diet which also affect medicine pharmacokinetic profiles. This review addresses aspects of medicine absorption, clearance, and other indices (such as medicine binding to plasma proteins) of enrofloxacin/marbofloxacin and chloramphenicol used for the systemic treatment of chlamydiosis, and fluconazole ± amphotericin, and posaconazole for the treatment of cryptococcosis. Based on observations from published studies, this review includes suggestions to improve therapeutic outcomes when administering medicines to diseased koalas.
Collapse
Affiliation(s)
- M Govendir
- Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
113
|
de Bruyn Kops C, Friedrich NO, Kirchmair J. Alignment-Based Prediction of Sites of Metabolism. J Chem Inf Model 2017; 57:1258-1264. [PMID: 28520411 DOI: 10.1021/acs.jcim.7b00165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Prediction of metabolically labile atom positions in a molecule (sites of metabolism) is a key component of the simulation of xenobiotic metabolism as a whole, providing crucial information for the development of safe and effective drugs. In 2008, an exploratory study was published in which sites of metabolism were derived based on molecular shape- and chemical feature-based alignment to a molecule whose site of metabolism (SoM) had been determined by experiments. We present a detailed analysis of the breadth of applicability of alignment-based SoM prediction, including transfer of the approach from a structure- to ligand-based method and extension of the applicability of the models from cytochrome P450 2C9 to all cytochrome P450 isozymes involved in drug metabolism. We evaluate the effect of molecular similarity of the query and reference molecules on the ability of this approach to accurately predict SoMs. In addition, we combine the alignment-based method with a leading chemical reactivity model to take reactivity into account. The combined model yielded superior performance in comparison to the alignment-based approach and the reactivity models with an average area under the receiver operating characteristic curve of 0.85 in cross-validation experiments. In particular, early enrichment was improved, as evidenced by higher BEDROC scores (mean BEDROC = 0.59 for α = 20.0, mean BEDROC = 0.73 for α = 80.5).
Collapse
Affiliation(s)
- Christina de Bruyn Kops
- Faculty of Mathematics, Informatics and Natural Sciences, Department of Computer Science, Center for Bioinformatics, Universität Hamburg , Hamburg 20146, Germany
| | - Nils-Ole Friedrich
- Faculty of Mathematics, Informatics and Natural Sciences, Department of Computer Science, Center for Bioinformatics, Universität Hamburg , Hamburg 20146, Germany
| | - Johannes Kirchmair
- Faculty of Mathematics, Informatics and Natural Sciences, Department of Computer Science, Center for Bioinformatics, Universität Hamburg , Hamburg 20146, Germany
| |
Collapse
|
114
|
Yang N, Sun R, Liao X, Aa J, Wang G. UDP-glucuronosyltransferases (UGTs) and their related metabolic cross-talk with internal homeostasis: A systematic review of UGT isoforms for precision medicine. Pharmacol Res 2017; 121:169-183. [PMID: 28479371 DOI: 10.1016/j.phrs.2017.05.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/03/2017] [Accepted: 05/03/2017] [Indexed: 12/11/2022]
Abstract
UDP-glucuronosyltransferases (UGTs) are the primary phase II enzymes catalyzing the conjugation of glucuronic acid to the xenobiotics with polar groups for facilitating their clearance. The UGTs belong to a superfamily that consists of diverse isoforms possessing distinct but overlapping metabolic activity. The abnormality or deficiency of UGTs in vivo is highly associated with some diseases, efficacy and toxicity of drugs, and precisely therapeutic personality. Despite the great effects and fruitful results achieved, to date, the expression and functions of individual UGTs have not been well clarified, the inconsistency of UGTs is often observed in human and experimental animals, and the complex regulation factors affecting UGTs have not been systematically summarized. This article gives an overview of updated reports on UGTs involving the various regulatory factors in terms of the genetic, environmental, pathological, and physiological effects on the functioning of individual UGTs, in turn, the dysfunction of UGTs induced disease risk and endo- or xenobiotic metabolism-related toxicity. The complex cross-talk effect of UGTs with internal homeostasis is systematically summarized and discussed in detail, which would be of great importance for personalized precision medicine.
Collapse
Affiliation(s)
- Na Yang
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Runbin Sun
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoying Liao
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Jiye Aa
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
115
|
Structure-metabolism relationships in human-AOX: Chemical insights from a large database of aza-aromatic and amide compounds. Proc Natl Acad Sci U S A 2017; 114:E3178-E3187. [PMID: 28373537 DOI: 10.1073/pnas.1618881114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aldehyde oxidase (AOX) is a metabolic enzyme catalyzing the oxidation of aldehyde and aza-aromatic compounds and the hydrolysis of amides, moieties frequently shared by the majority of drugs. Despite its key role in human metabolism, to date only fragmentary information about the chemical features responsible for AOX susceptibility are reported and only "very local" structure-metabolism relationships based on a small number of similar compounds have been developed. This study reports a more comprehensive coverage of the chemical space of structures with a high risk of AOX phase I metabolism in humans. More than 270 compounds were studied to identify the site of metabolism and the metabolite(s). Both electronic [supported by density functional theory (DFT) calculations] and exposure effects were considered when rationalizing the structure-metabolism relationship.
Collapse
|
116
|
Romão MJ, Coelho C, Santos-Silva T, Foti A, Terao M, Garattini E, Leimkühler S. Structural basis for the role of mammalian aldehyde oxidases in the metabolism of drugs and xenobiotics. Curr Opin Chem Biol 2017; 37:39-47. [DOI: 10.1016/j.cbpa.2017.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/05/2017] [Accepted: 01/06/2017] [Indexed: 10/20/2022]
|
117
|
Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep 2017; 7:42717. [PMID: 28256516 PMCID: PMC5335600 DOI: 10.1038/srep42717] [Citation(s) in RCA: 7812] [Impact Index Per Article: 976.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/13/2017] [Indexed: 02/06/2023] Open
Abstract
To be effective as a drug, a potent molecule must reach its target in the body in sufficient concentration, and stay there in a bioactive form long enough for the expected biologic events to occur. Drug development involves assessment of absorption, distribution, metabolism and excretion (ADME) increasingly earlier in the discovery process, at a stage when considered compounds are numerous but access to the physical samples is limited. In that context, computer models constitute valid alternatives to experiments. Here, we present the new SwissADME web tool that gives free access to a pool of fast yet robust predictive models for physicochemical properties, pharmacokinetics, drug-likeness and medicinal chemistry friendliness, among which in-house proficient methods such as the BOILED-Egg, iLOGP and Bioavailability Radar. Easy efficient input and interpretation are ensured thanks to a user-friendly interface through the login-free website http://www.swissadme.ch. Specialists, but also nonexpert in cheminformatics or computational chemistry can predict rapidly key parameters for a collection of molecules to support their drug discovery endeavours.
Collapse
Affiliation(s)
- Antoine Daina
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Quartier Sorge, Bâtiment Génopode, CH-1015 Lausanne, Switzerland
| | - Olivier Michielin
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Quartier Sorge, Bâtiment Génopode, CH-1015 Lausanne, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois, CH-1015 Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Vincent Zoete
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Quartier Sorge, Bâtiment Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
118
|
Kratochwil NA, Meille C, Fowler S, Klammers F, Ekiciler A, Molitor B, Simon S, Walter I, McGinnis C, Walther J, Leonard B, Triyatni M, Javanbakht H, Funk C, Schuler F, Lavé T, Parrott NJ. Metabolic Profiling of Human Long-Term Liver Models and Hepatic Clearance Predictions from In Vitro Data Using Nonlinear Mixed-Effects Modeling. AAPS JOURNAL 2017; 19:534-550. [DOI: 10.1208/s12248-016-0019-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/18/2016] [Indexed: 12/15/2022]
|
119
|
Schütze G, Schwarz MJ. Therapeutic Drug Monitoring for individualised risk reduction in psychopharmacotherapy. Trends Analyt Chem 2016. [DOI: 10.1016/j.trac.2016.05.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
120
|
Investigating the substrate binding mechanism of sulfotransferase 2A1 based on substrate tunnel analysis: a molecular dynamics simulation study. J Mol Model 2016; 22:176. [DOI: 10.1007/s00894-016-3041-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/20/2016] [Indexed: 10/21/2022]
|
121
|
Desirable Characteristics of Hepatitis C Treatment Regimens: A Review of What We Have and What We Need. Infect Dis Ther 2016; 5:299-312. [PMID: 27384319 PMCID: PMC5019974 DOI: 10.1007/s40121-016-0118-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Indexed: 12/23/2022] Open
Abstract
There have been dramatic advancements in the treatment of chronic hepatitis C (HCV) infection. This is largely due to the approval of several direct-acting antiviral agents (DAAs) from a variety of medication classes with novel mechanisms of action. These therapies are a welcomed advancement given their improved efficacy and tolerability compared to pegylated interferon and ribavirin (RBV)-based regimens. These convenient, all-oral regimens treat a variety of genotypes and often offer high cure rates in a variety of HCV-infected populations. While there are several benefits associated with these therapies, there are also notable shortcomings. Shortcomings include diminished response or need for adjunctive RBV in difficult-to-treat populations (decompensated cirrhosis, active substance abuse patients, advanced kidney disease, etc.), activity against select genotypes, substantial drug–drug interaction potential, and high cost. Therefore, while current DAA-based therapies have several favorable attributes, each also has its limitations. The purpose of this review is to (1) identify the characteristics of an ideal HCV treatment regimen, (2) describe desirable features of existing regimens, (3) summarize limitations of existing regimens, and (4) introduce promising emerging therapies. This manuscript will serve as a guide for evaluating the caliber of future HCV treatment regimens.
Collapse
|
122
|
Coughtrie MWH. Function and organization of the human cytosolic sulfotransferase (SULT) family. Chem Biol Interact 2016; 259:2-7. [PMID: 27174136 DOI: 10.1016/j.cbi.2016.05.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/02/2016] [Indexed: 12/29/2022]
Abstract
The sulfuryl transfer reaction is of fundamental biological importance. One of the most important manifestations of this process are the reactions catalyzed by members of the cytosolic sulfotransferase (SULT) superfamily. These enzymes transfer the sulfuryl moiety from the universal donor PAPS (3'-phosphoadenosine 5'-phosphosulfate) to a wide variety of substrates with hydroxyl- or amino-groups. Normally a detoxification reaction this facilitates the elimination of a multitude of xenobiotics, although for some molecules sulfation is a bioactivation step. In addition, sulfation plays a key role in endocrine and other signalling pathways since many steroids, sterols, thyroid hormones and catecholamines exist primarily as sulfate conjugates in humans. This article summarizes much of our current knowledge of the organization and function of the human cytosolic sulfotransferases and highlights some of the important interspecies differences that have implications for, among other things, drug development and chemical safety analysis.
Collapse
Affiliation(s)
- Michael W H Coughtrie
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
123
|
Li H, He J, Jia W. The influence of gut microbiota on drug metabolism and toxicity. Expert Opin Drug Metab Toxicol 2015; 12:31-40. [PMID: 26569070 DOI: 10.1517/17425255.2016.1121234] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Gut microbiota plays critical roles in drug metabolism. The variation of gut microbiota contributes to the interindividual differences toward drug therapy including drug-induced toxicity and efficacy. Accordingly, the investigation and elucidation of gut microbial impacts on drug metabolism and toxicity will not only facilitate the way of personalized medicine, but also improve rational drug design. AREAS COVERED This review provides an overview of the microbiota-host co-metabolism on drug metabolism and summarizes 30 clinical drugs that are co-metabolized by host and gut microbiota. Moreover, this review is specifically focused on elucidating the gut microbial modulation of some clinical drugs, in which the gut microbial influences on drug metabolism, drug-induced toxicity and efficacy are discussed. EXPERT OPINION The gut microbial contribution to drug metabolism and toxicity is increasingly recognized, but remains largely unexplored due to the extremely complex relationship between gut microbiota and host. The mechanistic elucidation of gut microbiota in drug metabolism is critical before any practical progress in drug design or personalized medicine could be made by modulating human gut microbiota. Analytical technique innovation is urgently required to strengthen our capability in recognizing microbial functions, including metagenomics, metabolomics and the integration of multidisciplinary knowledge.
Collapse
Affiliation(s)
- Houkai Li
- a Center for Chinese Medical Therapy and Systems Biology , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Jiaojiao He
- a Center for Chinese Medical Therapy and Systems Biology , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Wei Jia
- a Center for Chinese Medical Therapy and Systems Biology , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China.,b Cancer Epidemiology Program , University of Hawaii Cancer Center , Honolulu , HI 96813 , USA
| |
Collapse
|
124
|
Identification and characterization of naturally occurring inhibitors against UDP-glucuronosyltransferase 1A1 in Fructus Psoraleae (Bu-gu-zhi). Toxicol Appl Pharmacol 2015; 289:70-8. [DOI: 10.1016/j.taap.2015.09.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/19/2015] [Accepted: 09/01/2015] [Indexed: 01/18/2023]
|
125
|
The impact of ligands on the structure and flexibility of sulfotransferases: a molecular dynamics simulation study. J Mol Model 2015; 21:190. [DOI: 10.1007/s00894-015-2739-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/15/2015] [Indexed: 01/11/2023]
|
126
|
Lv X, Ge GB, Feng L, Troberg J, Hu LH, Hou J, Cheng HL, Wang P, Liu ZM, Finel M, Cui JN, Yang L. An optimized ratiometric fluorescent probe for sensing human UDP-glucuronosyltransferase 1A1 and its biological applications. Biosens Bioelectron 2015; 72:261-7. [PMID: 25988789 DOI: 10.1016/j.bios.2015.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/15/2015] [Accepted: 05/04/2015] [Indexed: 01/10/2023]
Abstract
This study aimed to develop a practical ratiometric fluorescent probe for highly selective and sensitive detection of human UDP-glucuronosyltransferase 1A1 (UGT1A1), one of the most important phase II enzymes. 4-Hydroxy-1,8-naphthalimide (HN) was selected as the fluorophore for this study because it possesses intramolecular charge transfer (ICT) feature and displays outstanding optical properties. A series of N-substituted derivatives with various hydrophobic, acidic and basic groups were designed and synthesized to evaluate the selectivity of HN derivatives toward UGT1A1. Our results demonstrated that the introduction of an acidic group to HN could significantly improve the selectivity of UGT1A1. Among the synthesized fluorescent probes, NCHN (N-3-carboxy propyl-4-hydroxy-1,8-naphthalimide) displayed the best combination of selectivity, sensitivity and ratiometric fluorescence response following UGT1A1-catalyzed glucuronidation. UGT1A1-catalyzed NCHN-4-O-glucuronidation generated a single fluorescent product with a high quantum yield (Φ=0.688) and brought remarkable changes in both color and fluorescence in comparison with the parental substrate. The newly developed probe has been successfully applied for sensitive measurements of UGT1A1 activities in human liver preparations, as well as for rapid screening of UGT1A1 modulators, using variable enzyme sources. Furthermore, its potential applications for live imaging of endogenous UGT1A1in cells have also been demonstrated.
Collapse
Affiliation(s)
- Xia Lv
- Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Guang-Bo Ge
- Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116012, China
| | - Lei Feng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116012, China
| | - Johanna Troberg
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Liang-Hai Hu
- College of Life Science, Jilin University, Changchun, China
| | - Jie Hou
- Dalian Medical University, Dalian 116044, China
| | | | - Ping Wang
- Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Zhao-Ming Liu
- Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jing-Nan Cui
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116012, China.
| | - Ling Yang
- Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
127
|
Abstract
Drug metabolism can produce metabolites with physicochemical and pharmacological properties that differ substantially from those of the parent drug, and consequently has important implications for both drug safety and efficacy. To reduce the risk of costly clinical-stage attrition due to the metabolic characteristics of drug candidates, there is a need for efficient and reliable ways to predict drug metabolism in vitro, in silico and in vivo. In this Perspective, we provide an overview of the state of the art of experimental and computational approaches for investigating drug metabolism. We highlight the scope and limitations of these methods, and indicate strategies to harvest the synergies that result from combining measurement and prediction of drug metabolism.
Collapse
|
128
|
Zhang PP, Zhao L, Long SY, Tian P. The effect of ligands on the thermal stability of sulfotransferases: a molecular dynamics simulation study. J Mol Model 2015; 21:72. [PMID: 25750022 DOI: 10.1007/s00894-015-2625-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 02/15/2015] [Indexed: 11/24/2022]
Abstract
Human cytosolic sulfotransferases (hSULTs) are important phase II metabolic enzymes. They catalyze transfer of the sulfuryl-group (-SO3) from 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to the hydroxyl or primary amine moieties of a large number of endogenous and xenobiotic substrates. Broad selectivity and specificity of binding and activity within the sulfortransferases family could be detected by thermal denaturation assays, which have been made more and more suitable for high throughput screening based on recent technical advances. Here molecular dynamics simulations were used to explore the effect of the cofactor (PAPS) and substrate (LCA) on the thermal stability of the enzyme. It was found that the apo-enzyme unfolded fastest upon heating. The holo-enzyme with bound substrate LCA unfolded slowest. This thermo-denaturation order is consistent with that observed in experiments. Further it was found that the cofactor and substrate will pronouncedly increase the thermal stability of the active pocket regions that interact directly with the ligands. In addition, cofactor and substrate show noticeable synergy effect on the thermal stability of the enzyme.
Collapse
Affiliation(s)
- Pu-pu Zhang
- School of Life Sciences, Jilin University, Changchun, China
| | | | | | | |
Collapse
|
129
|
Sanoh S, Tayama Y, Sugihara K, Kitamura S, Ohta S. Significance of aldehyde oxidase during drug development: Effects on drug metabolism, pharmacokinetics, toxicity, and efficacy. Drug Metab Pharmacokinet 2015; 30:52-63. [DOI: 10.1016/j.dmpk.2014.10.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/03/2014] [Accepted: 10/03/2014] [Indexed: 12/28/2022]
|
130
|
Bale SS, Vernetti L, Senutovitch N, Jindal R, Hegde M, Gough A, McCarty WJ, Bakan A, Bhushan A, Shun TY, Golberg I, DeBiasio R, Usta BO, Taylor DL, Yarmush ML. In vitro platforms for evaluating liver toxicity. Exp Biol Med (Maywood) 2014; 239:1180-1191. [PMID: 24764241 DOI: 10.1177/1535370214531872] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The liver is a heterogeneous organ with many vital functions, including metabolism of pharmaceutical drugs and is highly susceptible to injury from these substances. The etiology of drug-induced liver disease is still debated although generally regarded as a continuum between an activated immune response and hepatocyte metabolic dysfunction, most often resulting from an intermediate reactive metabolite. This debate stems from the fact that current animal and in vitro models provide limited physiologically relevant information, and their shortcomings have resulted in "silent" hepatotoxic drugs being introduced into clinical trials, garnering huge financial losses for drug companies through withdrawals and late stage clinical failures. As we advance our understanding into the molecular processes leading to liver injury, it is increasingly clear that (a) the pathologic lesion is not only due to liver parenchyma but is also due to the interactions between the hepatocytes and the resident liver immune cells, stellate cells, and endothelial cells; and (b) animal models do not reflect the human cell interactions. Therefore, a predictive human, in vitro model must address the interactions between the major human liver cell types and measure key determinants of injury such as the dosage and metabolism of the drug, the stress response, cholestatic effect, and the immune and fibrotic response. In this mini-review, we first discuss the current state of macro-scale in vitro liver culture systems with examples that have been commercialized. We then introduce the paradigm of microfluidic culture systems that aim to mimic the liver with physiologically relevant dimensions, cellular structure, perfusion, and mass transport by taking advantage of micro and nanofabrication technologies. We review the most prominent liver-on-a-chip platforms in terms of their physiological relevance and drug response. We conclude with a commentary on other critical advances such as the deployment of fluorescence-based biosensors to identify relevant toxicity pathways, as well as computational models to create a predictive tool.
Collapse
Affiliation(s)
- Shyam Sundhar Bale
- Center for Engineering in Medicine (CEM) at Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston MA 02114
| | - Lawrence Vernetti
- University of Pittsburgh Drug Discovery Institute, Pittsburgh PA 15260.,University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh PA 15260
| | - Nina Senutovitch
- University of Pittsburgh Drug Discovery Institute, Pittsburgh PA 15260.,University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh PA 15260
| | - Rohit Jindal
- Center for Engineering in Medicine (CEM) at Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston MA 02114
| | - Manjunath Hegde
- Center for Engineering in Medicine (CEM) at Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston MA 02114
| | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, Pittsburgh PA 15260.,University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh PA 15260
| | - William J McCarty
- Center for Engineering in Medicine (CEM) at Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston MA 02114
| | - Ahmet Bakan
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh PA 15260
| | - Abhinav Bhushan
- Center for Engineering in Medicine (CEM) at Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston MA 02114
| | - Tong Ying Shun
- University of Pittsburgh Drug Discovery Institute, Pittsburgh PA 15260
| | - Inna Golberg
- Center for Engineering in Medicine (CEM) at Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston MA 02114
| | - Richard DeBiasio
- University of Pittsburgh Drug Discovery Institute, Pittsburgh PA 15260
| | - Berk Osman Usta
- Center for Engineering in Medicine (CEM) at Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston MA 02114
| | - D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, Pittsburgh PA 15260.,University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh PA 15260
| | - Martin L Yarmush
- Center for Engineering in Medicine (CEM) at Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston MA 02114
| |
Collapse
|