1
|
Tillmann AC, Peters DJM, Rostami-Hodjegan A, Wilson P, Norman J, Barber J, Al-Majdoub ZM. Changes in Protein Expression of Renal Drug Transporters and Drug-Metabolizing Enzymes in Autosomal Dominant Polycystic Kidney Disease Patients. Clin Pharmacol Ther 2025. [PMID: 40371605 DOI: 10.1002/cpt.3715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Autosomal dominant polycystic kidney disease is the most prevalent inherited kidney disease and leads to bilateral kidney enlargement and progressive loss of renal function, often over decades. Comorbidities include hypertension, flank pain, and bacterial infections. The condition often necessitates prolonged multidrug therapy. Given the kidneys' critical role in drug excretion, the progressive functional impairment in the disease can lead to complications such as drug overdosing and unexpected levels of drug-drug interactions. Studies of drug-metabolizing enzyme and transporter expression in this patient group remain scarce. We conducted comprehensive global liquid chromatography-tandem mass spectrometry proteomic analyses of microsomal and cytosolic fractions from early-stage (chronic kidney disease stage: 13, n = 16) and end-stage autosomal dominant polycystic kidney disease patients (chronic kidney disease stage: 5, n = 14), comparing them with age-matched healthy controls (n = 11). In the early-stage ADPKD samples, most drug-metabolizing enzymes and drug transporters did not differ significantly from the healthy controls. Exceptions were EPHX2 and SULT1C2 in the cytosolic fraction, which showed a more than 2-fold decrease in abundance (P < 0.05). In contrast, the end-stage ADPKD kidney samples showed a decrease in the abundance of most measured proteins. Several drug-metabolizing enzymes, including CYP4F2, UGT1A6, UGT1A9, and UGT2B7, exhibited statistically significant reductions (P < 0.05). Among the drug transporters, OAT1, OAT3, and OCT2 were below the limit of quantification in most ES-ADPKD samples. MDR1 was the only efflux drug transporter consistently measured, with an average abundance of 1.24 pmol/mg microsomal protein across all samples.
Collapse
Affiliation(s)
- Annika C Tillmann
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
- Certara Predictive Technologies (CPT), Sheffield, UK
| | - Patricia Wilson
- UCL Centre for Kidney and Bladder Health, University College London (UCL) Royal Free Hospital, London, UK
- PKD Charity UK, London, UK
| | - Jill Norman
- UCL Centre for Kidney and Bladder Health, University College London (UCL) Royal Free Hospital, London, UK
- PKD Charity UK, London, UK
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Singh DK, Ahire D, Davydov DR, Prasad B. Differential Tissue Abundance of Membrane-Bound Drug Metabolizing Enzymes and Transporter Proteins by Global Proteomics. Drug Metab Dispos 2024; 52:1152-1160. [PMID: 38641346 PMCID: PMC11495667 DOI: 10.1124/dmd.124.001477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/21/2024] Open
Abstract
Protein abundance data of drug-metabolizing enzymes and transporters (DMETs) are useful for scaling in vitro and animal data to humans for accurate prediction and interpretation of drug clearance and toxicity. Targeted DMET proteomics that relies on synthetic stable isotope-labeled surrogate peptides as calibrators is routinely used for the quantification of selected proteins; however, the technique is limited to the quantification of a small number of proteins. Although the global proteomics-based total protein approach (TPA) is emerging as a better alternative for large-scale protein quantification, the conventional TPA does not consider differential sequence coverage by identifying unique peptides across proteins. Here, we optimized the TPA approach by correcting protein abundance data by the sequence coverage, which was applied to quantify 54 DMETs for characterization of 1) differential tissue DMET abundance in the human liver, kidney, and intestine, and 2) interindividual variability of DMET proteins in individual intestinal samples (n = 13). Uridine diphosphate-glucuronosyltransferase 2B7 (UGT2B7), microsomal glutathione S-transferases (MGST1, MGST2, and MGST3) carboxylesterase 2 (CES2), and multidrug resistance-associated protein 2 (MRP2) were expressed in all three tissues, whereas, as expected, four cytochrome P450s (CYP3A4, CYP3A5, CYP2C9, and CYP4F2), UGT1A1, UGT2B17, CES1, flavin-containing monooxygenase 5, MRP3, and P-glycoprotein were present in the liver and intestine. The top three DMET proteins in individual tissues were: CES1>CYP2E1>UGT2B7 (liver), CES2>UGT2B17>CYP3A4 (intestine), and MGST1>UGT1A6>MGST2 (kidney). CYP3A4, CYP3A5, UGT2B17, CES2, and MGST2 showed high interindividual variability in the intestine. These data are relevant for enhancing in vitro to in vivo extrapolation of drug absorption and disposition and can be used to enhance the accuracy of physiologically based pharmacokinetic prediction of systemic and tissue concentration of drugs. SIGNIFICANCE STATEMENT: This study quantified the abundance and compositions of drug-metabolizing enzymes and transporters in pooled human liver, intestine, and kidney microsomes as well as individual intestinal microsomes using an optimized global proteomics approach. The data revealed large intertissue differences in the abundance of these proteins and high intestinal interindividual variability in the levels of cytochrome P450s (e.g., CYP3A4 and CYP3A5), uridine diphosphate-glucuronosyltransferase 2B17, carboxylesterase 2, and microsomal glutathione S-transferase 2. These data are applicable for the prediction of first-pass metabolism and tissue-specific drug clearance.
Collapse
Affiliation(s)
- Dilip Kumar Singh
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., D.A., B.P.); and Department of Chemistry, Washington State University, Pullman, Washington (D.R.D.)
| | - Deepak Ahire
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., D.A., B.P.); and Department of Chemistry, Washington State University, Pullman, Washington (D.R.D.)
| | - Dmitri R Davydov
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., D.A., B.P.); and Department of Chemistry, Washington State University, Pullman, Washington (D.R.D.)
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., D.A., B.P.); and Department of Chemistry, Washington State University, Pullman, Washington (D.R.D.)
| |
Collapse
|
3
|
Hou S, Liu H, Hu Y, Zhang J, Deng X, Li Z, Zhang Y, Li X, Li Y, Ma L, Yao J, Chen X. Discovery of a novel homocysteine thiolactone hydrolase and the catalytic activity of its natural variants. Protein Sci 2024; 33:e5098. [PMID: 38980003 PMCID: PMC11232049 DOI: 10.1002/pro.5098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Homocysteine thiolactone (HTL), a toxic metabolite of homocysteine (Hcy) in hyperhomocysteinemia (HHcy), is known to modify protein structure and function, leading to protein damage through formation of N-Hcy-protein. HTL has been highly linked to HHcy-associated cardiovascular and neurodegenerative diseases. The protective role of HTL hydrolases against HTL-associated vascular toxicity and neurotoxicity have been reported. Although several endogeneous enzymes capable of hydrolyzing HTL have been identified, the primary enzyme responsible for its metabolism remains unclear. In this study, three human carboxylesterases were screened to explore new HTL hydrolase and human carboxylesterase 1 (hCES1) demonstrates the highest catalytic activity against HTL. Given the abundance of hCES1 in the liver and the clinical significance of its single-nucleotide polymorphisms (SNPs), six common hCES1 nonsynonymous coding SNP (nsSNPs) variants were examined and characterized for their kinetic parameters. Variants E220G and G143E displayed 7.3-fold and 13.2-fold lower catalytic activities than its wild-type counterpart. In addition, the detailed catalytic mechanism of hCES1 for HTL hydrolysis was computational investigated and elucidated by Quantum mechanics/molecular mechanics (QM/MM) molecular dynamics (MD) method. The function of residues E220 and G143 in sustaining its hydrolytic activity of hCES1 was analyzed, and the calculated energy difference aligns well with experimental-derived results, supporting the validity of our computational insights. These findings provide insights into the potential protective role of hCES1 against HTL-associated toxicity, and warrant future studies on the possible association between specific genetic variants of hCES1 with impaired catalytic function and clinical susceptibility of HTL-associated cardiovascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Shurong Hou
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| | - Huan Liu
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| | - Yihui Hu
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| | - Jie Zhang
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| | - Xingyu Deng
- Shanghai Key Laboratory of New Drug DesignSchool of Pharmacy, East China University of Science and TechnologyShanghaiChina
| | - Zhenzhen Li
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| | - Yun Zhang
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| | - Xiaoxuan Li
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| | - Yishuang Li
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| | - Lei Ma
- Shanghai Key Laboratory of New Drug DesignSchool of Pharmacy, East China University of Science and TechnologyShanghaiChina
| | - Jianzhuang Yao
- School of Biological Science and Technology, University of JinanJinanChina
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
4
|
Hammid A, Honkakoski P. Ocular drug-metabolizing enzymes: focus on esterases. Drug Metab Rev 2024; 56:175-189. [PMID: 38888291 DOI: 10.1080/03602532.2024.2368247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
This review describes current knowledge on the expression of ocular phase I and II drug-metabolizing enzymes in the main animal species used in ocular drug development and in humans, with a focus on ocular esterases and their prodrug substrates. The eye possesses a unique metabolic profile, exhibiting a lower and restricted expression of major cytochrome P450s (CYPs) and most transferases apart from glutathione S-transferases (GST) when compared to the liver. In contrast, hydrolytic enzymes are abundant in many ocular tissues. These enzymes have attracted interest because of their role in prodrug activation and drug elimination. A literature survey suggests profound variations in tissue expression levels and activities between different species but also points out significant gaps in knowledge. These uncertainties highlight a need for more detailed characterization of enzymes in individual ocular tissues and across species to aid future translational studies in ophthalmic drug research. Thus, an in-depth analysis of ocular drug metabolism and species differences is crucial for ocular drug development.
Collapse
Affiliation(s)
- Anam Hammid
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
5
|
Lin X, Liu M, Yi Q, Zhou Y, Su J, Qing B, Lu Y, Pu C, Lan W, Zou L, Wang J. Design, synthesis, and evaluation of a carboxylesterase detection probe with therapeutic effects. Talanta 2024; 274:126060. [PMID: 38604044 DOI: 10.1016/j.talanta.2024.126060] [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: 12/24/2023] [Revised: 03/20/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
In this study, a lysosomal targeting fluorescent probe recognition on CEs was designed and synthesized. The obtained probe BF2-cur-Mor demonstrated excellent selectivity, sensitivity, pH-independence, and enzyme affinity towards CEs within 5 min. BF2-cur-Mor could enable recognition of intracellular CEs and elucidate that the CEs content of different cancer cells follows the rule of HepG2 > HCT-116 > A549 > HeLa, and the CEs expression level of hepatoma cancer cells far exceeds that of normal hepatic cells, being in good agreement with the previous reports. The ability of BF2-cur-Mor to monitor CEs in vivo was confirmed by zebrafish experiment. BF2-cur-Mor exhibits some pharmacological activity in that it can induce apoptosis in hepatocellular carcinoma cells but is weaker in normal hepatocyte cells, being expected to be a potential "diagnostic and therapeutic integration" tool for the clinical diagnosis of CEs-related diseases.
Collapse
Affiliation(s)
- Xia Lin
- Guangxi Key Laboratory of Special Biomedicine, Medical College, Guangxi University, Nanning, 530004, China; Guangxi Health Science College, Nanning, 530023, China; School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, China
| | - Min Liu
- Guangxi Key Laboratory of Special Biomedicine, Medical College, Guangxi University, Nanning, 530004, China
| | - Qingyuan Yi
- Guangxi Key Laboratory of Special Biomedicine, Medical College, Guangxi University, Nanning, 530004, China
| | - Ying Zhou
- Guangxi Key Laboratory of Special Biomedicine, Medical College, Guangxi University, Nanning, 530004, China
| | - Jinchan Su
- Guangxi Key Laboratory of Special Biomedicine, Medical College, Guangxi University, Nanning, 530004, China
| | - Binyang Qing
- College of Life Science and Technology, Guangxi University, Nanning, 530004, China
| | - Yaqi Lu
- Guangxi Key Laboratory of Special Biomedicine, Medical College, Guangxi University, Nanning, 530004, China
| | - Chunxiao Pu
- College of Life Science and Technology, Guangxi University, Nanning, 530004, China
| | - Weisen Lan
- College of Agriculture, Guangxi University, Nanning, 530004, China
| | - Lianjia Zou
- Guangxi Health Science College, Nanning, 530023, China.
| | - Jianyi Wang
- Guangxi Key Laboratory of Special Biomedicine, Medical College, Guangxi University, Nanning, 530004, China; School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, China.
| |
Collapse
|
6
|
Nagaoka M, Sakai Y, Nakajima M, Fukami T. Role of carboxylesterase and arylacetamide deacetylase in drug metabolism, physiology, and pathology. Biochem Pharmacol 2024; 223:116128. [PMID: 38492781 DOI: 10.1016/j.bcp.2024.116128] [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: 12/01/2023] [Revised: 01/20/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Carboxylesterases (CES1 and CES2) and arylacetamide deacetylase (AADAC), which are expressed primarily in the liver and/or gastrointestinal tract, hydrolyze drugs containing ester and amide bonds in their chemical structure. These enzymes often catalyze the conversion of prodrugs, including the COVID-19 drugs remdesivir and molnupiravir, to their pharmacologically active forms. Information on the substrate specificity and inhibitory properties of these enzymes, which would be useful for drug development and toxicity avoidance, has accumulated. Recently,in vitroandin vivostudies have shown that these enzymes are involved not only in drug hydrolysis but also in lipid metabolism. CES1 and CES2 are capable of hydrolyzing triacylglycerol, and the deletion of their orthologous genes in mice has been associated with impaired lipid metabolism and hepatic steatosis. Adeno-associated virus-mediated human CES overexpression decreases hepatic triacylglycerol levels and increases fatty acid oxidation in mice. It has also been shown that overexpression of CES enzymes or AADAC in cultured cells suppresses the intracellular accumulation of triacylglycerol. Recent reports indicate that AADAC can be up- or downregulated in tumors of various organs, and its varied expression is associated with poor prognosis in patients with cancer. Thus, CES and AADAC not only determine drug efficacy and toxicity but are also involved in pathophysiology. This review summarizes recent findings on the roles of CES and AADAC in drug metabolism, physiology, and pathology.
Collapse
Affiliation(s)
- Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
7
|
Sharma A, Jin L, Wang X, Wang YT, Stresser DM. Developing an adult stem cell derived microphysiological intestinal system for predicting oral prodrug bioconversion and permeability in humans. LAB ON A CHIP 2024; 24:339-355. [PMID: 38099395 DOI: 10.1039/d3lc00843f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Microphysiological systems (MPS) incorporating human intestinal organoids have shown the potential to faithfully model intestinal biology with the promise to accelerate development of oral prodrugs. We hypothesized that an MPS model incorporating flow, shear stress, and vasculature could provide more reliable measures of prodrug bioconversion and permeability. Following construction of jejunal and duodenal organoid MPS derived from 3 donors, we determined the area under the concentration-time (AUC) curve for the active drug in the vascular channel and characterized the enzymology of prodrug bioconversion. Fosamprenavir underwent phosphatase mediated hydrolysis to amprenavir while dabigatran etexilate (DABE) exhibited proper CES2- and, as anticipated, not CES1-mediated de-esterification, followed by permeation of amprenavir to the vascular channel. When experiments were conducted in the presence of bio-converting enzyme inhibitors (orthovanadate for alkaline phosphatase; bis(p-nitrophenyl)phosphate for carboxylesterase), the AUC of the active drug decreased accordingly in the vascular channel. In addition to functional analysis, the MPS was characterized through imaging and proteomic analysis. Imaging revealed proper expression and localization of epithelial, endothelial, tight junction and catalytic enzyme markers. Global proteomic analysis was used to analyze the MPS model and 3 comparator sources: an organoid-based transwell model (which was also evaluated for function), Matrigel embedded organoids and finally jejunal and duodenal cadaver tissues collected from 3 donors. Hierarchical clustering analysis (HCA) and principal component analysis (PCA) of global proteomic data demonstrated that all organoid-based models exhibited strong similarity and were distinct from tissues. Intestinal organoids in the MPS model exhibited strong similarity to human tissue for key epithelial markers via HCA. Quantitative proteomic analysis showed higher expression of key prodrug converting and drug metabolizing enzymes in MPS-derived organoids compared to tissues, organoids in Matrigel, and organoids on transwells. When comparing organoids from MPS and transwells, expression of intestinal alkaline phosphatase (ALPI), carboxylesterase (CES)2, cytochrome P450 3A4 (CYP3A4) and sucrase isomaltase (SI) was 2.97-, 1.2-, 11.3-, and 27.7-fold higher for duodenum and 7.7-, 4.6-, 18.1-, and 112.2-fold higher for jejunum organoids in MPS, respectively. The MPS approach can provide a more physiological system than enzymes, organoids, and organoids on transwells for pharmacokinetic analysis of prodrugs that account for 10% of all commercial medicines.
Collapse
Affiliation(s)
- Abhinav Sharma
- Quantitative, Translational & ADME Sciences, AbbVie, Inc, 1 North Waukegan Road, North Chicago, IL 60064, USA.
| | - Liang Jin
- AbbVie Bioresearch Center, Worcester, MA, USA
| | - Xue Wang
- AbbVie Bioresearch Center, Worcester, MA, USA
| | - Yue-Ting Wang
- Quantitative, Translational & ADME Sciences, AbbVie, Inc, 1 North Waukegan Road, North Chicago, IL 60064, USA.
| | - David M Stresser
- Quantitative, Translational & ADME Sciences, AbbVie, Inc, 1 North Waukegan Road, North Chicago, IL 60064, USA.
| |
Collapse
|
8
|
Li HX, Sun MR, Zhang Y, Song LL, Zhang F, Song YQ, Hou XD, Ge GB. Human Carboxylesterase 1A Plays a Predominant Role in Hydrolysis of the Anti-Dyslipidemia Agent Fenofibrate in Humans. Drug Metab Dispos 2023; 51:1490-1498. [PMID: 37550069 DOI: 10.1124/dmd.123.001365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023] Open
Abstract
Fenofibrate, a marketed peroxisome proliferator-activated receptor-α (PPARα) agonist, has been widely used for treating severe hypertriglyceridemia and mixed dyslipidemia. As a canonical prodrug, fenofibrate can be rapidly hydrolyzed to release the active metabolite (fenofibric acid) in vivo, but the crucial enzyme(s) responsible for fenofibrate hydrolysis and the related hydrolytic kinetics have not been well-investigated. This study aimed to assign the key organs and crucial enzymes involved in fenofibrate hydrolysis in humans, as well as reveal the impact of fenofibrate hydrolysis on its non-PPAR-mediated biologic activities. Our results demonstrated that fenofibrate could be rapidly hydrolyzed in the preparations from both human liver and lung to release fenofibric acid. Reaction phenotyping assays coupling with chemical inhibition assays showed that human carboxylesterase 1A (hCES1A) played a predominant role in fenofibrate hydrolysis in human liver and lung, while human carboxylesterase 2A (hCES2A) and human monoacylglycerol esterase (hMAGL) contributed to a very lesser extent. Kinetic analyses showed that fenofibrate could be rapidly hydrolyzed by hCES1A in human liver preparations, while the inherent clearance of hCES1A-catalyzed fenofibrate hydrolysis is much higher (>200-fold) than than that of hCES2A or hMAGL. Biologic assays demonstrated that both fenofibrate and fenofibric acid showed very closed Nrf2 agonist effects, but fenofibrate hydrolysis strongly weakens its inhibitory effects against both hCES2A and hNtoum. Collectively, our findings reveal that the liver is the major organ and hCES1A is the predominant enzyme-catalyzing fenofibrate hydrolysis in humans, while fenofibrate hydrolysis significantly reduces inhibitory effects of fenofibrate against serine hydrolases. SIGNIFICANCE STATEMENT: Fenofibrate can be completely converted to fenofibric acid in humans and subsequently exert its pharmacological effects, but the hydrolytic pathways of fenofibrate in humans have not been well-investigated. This study reported that the liver was the predominant organ and human carboxylesterase 1A was the crucial enzyme involved in fenofibrate hydrolysis in humans.
Collapse
Affiliation(s)
- Hong-Xin Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (H.-X.L., M.-R.S., Y.Z., L.-L.S., F.Z., Y.-Q.S., X.-D.H., G.-B.G.) and Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (L.-L.S.)
| | - Meng-Ru Sun
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (H.-X.L., M.-R.S., Y.Z., L.-L.S., F.Z., Y.-Q.S., X.-D.H., G.-B.G.) and Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (L.-L.S.)
| | - Ya Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (H.-X.L., M.-R.S., Y.Z., L.-L.S., F.Z., Y.-Q.S., X.-D.H., G.-B.G.) and Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (L.-L.S.)
| | - Li-Lin Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (H.-X.L., M.-R.S., Y.Z., L.-L.S., F.Z., Y.-Q.S., X.-D.H., G.-B.G.) and Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (L.-L.S.)
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (H.-X.L., M.-R.S., Y.Z., L.-L.S., F.Z., Y.-Q.S., X.-D.H., G.-B.G.) and Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (L.-L.S.)
| | - Yun-Qing Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (H.-X.L., M.-R.S., Y.Z., L.-L.S., F.Z., Y.-Q.S., X.-D.H., G.-B.G.) and Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (L.-L.S.)
| | - Xu-Dong Hou
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (H.-X.L., M.-R.S., Y.Z., L.-L.S., F.Z., Y.-Q.S., X.-D.H., G.-B.G.) and Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (L.-L.S.)
| | - Guang-Bo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (H.-X.L., M.-R.S., Y.Z., L.-L.S., F.Z., Y.-Q.S., X.-D.H., G.-B.G.) and Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (L.-L.S.)
| |
Collapse
|
9
|
Song J, Yu J, Sun K, Chen Z, Xing X, Yang Y, Sun C, Wang Z. Preparation of a Highly Selective “Off-On” Rhodamine-Based Fluorescent Probe for the Specific Determination of Carboxylesterase 2 and Cell Imaging. ANAL LETT 2023. [DOI: 10.1080/00032719.2023.2175213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Affiliation(s)
- Jia Song
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Jiaying Yu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Kai Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Zhixin Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Xiaoxiao Xing
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Yumeng Yang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Chunyu Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Zhifei Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
Hammid A, Fallon JK, Lassila T, Vieiro P, Balla A, Gonzalez F, Urtti A, Smith PC, Tolonen A, Honkakoski P. Activity and Expression of Carboxylesterases and Arylacetamide Deacetylase in Human Ocular Tissues. Drug Metab Dispos 2022; 50:1483-1492. [PMID: 36195336 DOI: 10.1124/dmd.122.000993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/23/2022] [Indexed: 11/22/2022] Open
Abstract
As a multitissue organ, the eye possesses unique anatomy and physiology, including differential expression of drug-metabolizing enzymes. Several hydrolytic enzymes that play a major role in drug metabolism and bioactivation of prodrugs have been detected in ocular tissues, but data on their quantitative expression is scarce. Also, many ophthalmic drugs are prone to hydrolysis. Metabolic characterization of individual ocular tissues is useful for the drug development process, and therefore, seven individual ocular tissues from human eyes were analyzed for the activity and expression of carboxylesterases (CESs) and arylacetamide deacetylase (AADAC). Generic and selective human esterase substrates 4-nitrophenyl acetate (most esterases), D-luciferin methyl ester (CES1), fluorescein diacetate and procaine (CES2), and phenacetin (AADAC) were applied to determine the enzymes' specific activities. Enzyme kinetics and inhibition studies were performed with isoform-selective inhibitors digitonin (CES1) and verapamil and diltiazem (CES2). Enzyme contents were determined using quantitative targeted proteomics, and CES2 expression was confirmed by western blotting. The expression and activity of human CES1 among ocular tissues varied by >10-fold, with the highest levels found in the retina and iris-ciliary body. In contrast, human CES2 expression appeared lower and more similar between tissues, whereas AADAC could not be detected. Inhibition studies showed that hydrolysis of fluorescein diacetate is also catalyzed by enzymes other than CES2. This study provides, for the first time, quantitative information on the tissue-dependent expression of human ocular esterases, which can be useful for the development of ocular drugs, prodrugs, and in pharmacokinetic modeling of the eye. SIGNIFICANCE STATEMENT: Novel and comprehensive data on the protein expression and activities of carboxylesterases from individual human eye tissues are generated. In combination with previous reports on preclinical species, this study will improve the understanding of interspecies differences in ocular drug metabolism and aid the development of ocular pharmacokinetics models.
Collapse
Affiliation(s)
- Anam Hammid
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - John K Fallon
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Toni Lassila
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Paula Vieiro
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Anusha Balla
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Francisco Gonzalez
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Arto Urtti
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Philip C Smith
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Ari Tolonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| |
Collapse
|
11
|
Laeer S, Cawello W, Burckhardt BB, Ablonczy L, Bajcetic M, Breur JMPJ, Dalinghaus M, Male C, de Wildt SN, Breitkreutz J, Faisal M, Keatley-Clarke A, Klingmann I, Lagler FB. Enalapril and Enalaprilat Pharmacokinetics in Children with Heart Failure Due to Dilated Cardiomyopathy and Congestive Heart Failure after Administration of an Orodispersible Enalapril Minitablet (LENA-Studies). Pharmaceutics 2022; 14:pharmaceutics14061163. [PMID: 35745735 PMCID: PMC9228797 DOI: 10.3390/pharmaceutics14061163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 12/29/2022] Open
Abstract
Angiotensin-converting enzyme inhibitors (ACEI), such as enalapril, are a cornerstone of treatment for pediatric heart failure which is still used off-label. Using a novel age-appropriate formulation of enalapril orodispersible minitablets (ODMTs), phase II/III open-label, multicenter pharmacokinetic (PK) bridging studies were performed in pediatric patients with heart failure due to dilated cardiomyopathy (DCM) and congenital heart disease (CHD) in five participating European countries. Children were treated for 8 weeks with ODMTs according to an age-appropriate dosing schedule. The primary objective was to describe PK parameters (area under the curve (AUC), maximal concentration (Cmax), time to reach maximal concentration (t-max)) of enalapril and its active metabolite enalaprilat. Of 102 patients, 89 patients (n = 26, DCM; n = 63 CHD) were included in the primary PK endpoint analysis. Rate and extent of enalapril and its active metabolite enalaprilat were described and etiology and age could be identified as potential PK modifying factors. The dosing schedule appeared to be tolerated well and did not result in any significant drug-related serious adverse events. The PK analysis and the lack of severe safety events supports the applied age-appropriate dosing schedule for the enalapril ODMTs.
Collapse
Affiliation(s)
- Stephanie Laeer
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-Universitaet Düsseldorf, 40225 Duesseldorf, Germany; (W.C.); (B.B.B.); (M.F.)
- Correspondence: ; Tel.: +49-211-8110740
| | - Willi Cawello
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-Universitaet Düsseldorf, 40225 Duesseldorf, Germany; (W.C.); (B.B.B.); (M.F.)
| | - Bjoern B. Burckhardt
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-Universitaet Düsseldorf, 40225 Duesseldorf, Germany; (W.C.); (B.B.B.); (M.F.)
| | - László Ablonczy
- Goettsegen György Hungarian Institute of Cardiology (HPHC), 1450 Budapest, Hungary;
| | - Milica Bajcetic
- Univerzitetska Dečja Klinika (UDK), University Children Hospital, School of Medicine, University of Belgrade, 11129 Belgrade, Serbia;
| | - Johannes M. P. J. Breur
- University Medical Center Utrecht, Wilhelmina Children’s Hospital, 3584 CX Utrecht, The Netherlands;
| | - Michiel Dalinghaus
- Division of Pediatric Cardiology, Erasmus MC Sophia Children’s Hospital, 3000 CA Rotterdam, The Netherlands;
| | - Christoph Male
- Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Saskia N. de Wildt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children’s Hospital, 3015 GJ Rotterdam, The Netherlands;
- Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | | | - Muhammed Faisal
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-Universitaet Düsseldorf, 40225 Duesseldorf, Germany; (W.C.); (B.B.B.); (M.F.)
| | | | | | | |
Collapse
|
12
|
Data-independent acquisition (DIA): An emerging proteomics technology for analysis of drug-metabolizing enzymes and transporters. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 39:49-56. [PMID: 34906325 DOI: 10.1016/j.ddtec.2021.06.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/22/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023]
Abstract
Data-independent acquisition (DIA) proteomics is a recently-developed global mass spectrometry (MS)-based proteomics strategy. In a DIA method, precursor ions are isolated into pre-defined isolation windows and fragmented; all fragmented ions in each window are then analyzed by a high-resolution mass spectrometer. DIA proteomics analysis is characterized by a broad protein coverage, high reproducibility, and accuracy, and its combination with advances in other techniques such as sample preparation and computational data analysis could lead to further improvements in assay performances. DIA technology has been increasingly utilized in various proteomics studies, including quantifying drug-metabolizing enzymes and transporters. Quantitative proteomics study of drug-metabolizing enzymes and transporters could lead to a better understanding of pharmacokinetics and pharmacodynamics and facilitate drug development. This review summarizes the application of DIA technology in proteomic analysis of drug-metabolizing enzymes and transporters.
Collapse
|
13
|
Liu J, Shang X, Huang S, Xu Y, Lu J, Zhang Y, Liu Z, Wang X. Construction and Characterization of CRISPR/Cas9 Knockout Rat Model of Carboxylesterase 2a Gene. Mol Pharmacol 2021; 100:480-490. [PMID: 34503976 DOI: 10.1124/molpharm.121.000357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/30/2021] [Indexed: 11/22/2022] Open
Abstract
Carboxylesterase (CES) 2, an important metabolic enzyme, plays a critical role in drug biotransformation and lipid metabolism. Although CES2 is very important, few animal models have been generated to study its properties and functions. Rat Ces2 is similar to human CES2A-CES3A-CES4A gene cluster, with highly similar gene structure, function, and substrate. In this report, CRISPR-associated protein-9 (CRISPR/Cas9) technology was first used to knock out rat Ces2a, which is a main subtype of Ces2 mostly distributed in the liver and intestine. This model showed the absence of CES2A protein expression in the liver. Further pharmacokinetic studies of diltiazem, a typical substrate of CES2A, confirmed the loss of function of CES2A both in vivo and in vitro. At the same time, the expression of CES2C and CES2J protein in the liver decreased significantly. The body and liver weight of Ces2a knockout rats also increased, but the food intake did not change. Moreover, the deficiency of Ces2a led to obesity, insulin resistance, and liver fat accumulation, which are consistent with the symptoms of nonalcoholic fatty liver disease (NAFLD). Therefore, this rat model is not only a powerful tool to study drug metabolism mediated by CES2 but also a good disease model to study NAFLD. SIGNIFICANCE STATEMENT: Human carboxylesterase (CES) 2 plays a key role in the first-pass hydrolysis metabolism of most oral prodrugs as well as lipid metabolism. In this study, CRISPR/Cas9 technology was used to knock out Ces2a gene in rats for the first time. This model can be used not only in the study of drug metabolism and pharmacokinetics but also as a disease model of nonalcoholic fatty liver disease (NAFLD) and other metabolic disorders.
Collapse
Affiliation(s)
- Jie Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Xuyang Shang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Shengbo Huang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Yuan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Jian Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Yuanjin Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Zongjun Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| |
Collapse
|
14
|
Honda S, Fukami T, Hirosawa K, Tsujiguchi T, Zhang Y, Nakano M, Uehara S, Uno Y, Yamazaki H, Nakajima M. Differences in Hydrolase Activities in the Liver and Small Intestine between Marmosets and Humans. Drug Metab Dispos 2021; 49:718-728. [PMID: 34135089 DOI: 10.1124/dmd.121.000513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022] Open
Abstract
For drug development, species differences in drug-metabolism reactions present obstacles for predicting pharmacokinetics in humans. We characterized the species differences in hydrolases among humans and mice, rats, dogs, and cynomolgus monkeys. In this study, to expand the series of such studies, we attempted to characterize marmoset hydrolases. We measured hydrolase activities for 24 compounds using marmoset liver and intestinal microsomes, as well as recombinant marmoset carboxylesterase (CES) 1, CES2, and arylacetamide deacetylase (AADAC). The contributions of CES1, CES2, and AADAC to hydrolysis in marmoset liver microsomes were estimated by correcting the activities by using the ratios of hydrolase protein levels in the liver microsomes and those in recombinant systems. For six out of eight human CES1 substrates, the activities in marmoset liver microsomes were lower than those in human liver microsomes. For two human CES2 substrates and three out of seven human AADAC substrates, the activities in marmoset liver microsomes were higher than those in human liver microsomes. Notably, among the three rifamycins, only rifabutin was hydrolyzed by marmoset tissue microsomes and recombinant AADAC. The activities for all substrates in marmoset intestinal microsomes tended to be lower than those in liver microsomes, which suggests that the first-pass effects of the CES and AADAC substrates are due to hepatic hydrolysis. In most cases, the sums of the values of the contributions of CES1, CES2, and AADAC were below 100%, which indicated the involvement of other hydrolases in marmosets. In conclusion, we clarified the substrate preferences of hydrolases in marmosets. SIGNIFICANCE STATEMENT: This study confirmed that there are large differences in hydrolase activities between humans and marmosets by characterizing marmoset hydrolase activities for compounds that are substrates of human CES1, CES2, or arylacetamide deacetylase. The data obtained in this study may be useful for considering whether marmosets are appropriate for examining the pharmacokinetics and efficacies of new chemical entities in preclinical studies.
Collapse
Affiliation(s)
- Shiori Honda
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Keiya Hirosawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Takuya Tsujiguchi
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Yongjie Zhang
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Shotaro Uehara
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Yasuhiro Uno
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Hiroshi Yamazaki
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| |
Collapse
|
15
|
Feng J, Lu H, Yang Y, Huang W, Cheng H, Kong H, Li L. SERS-ELISA determination of human carboxylesterase 1 using metal-organic framework doped with gold nanoparticles as SERS substrate. Mikrochim Acta 2021; 188:280. [PMID: 34331134 DOI: 10.1007/s00604-021-04928-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/05/2021] [Indexed: 10/20/2022]
Abstract
By in situ synthesis of gold nanoparticles (AuNPs) within the acid-etched (AE) MIL-101 (Cr) framework, AE-MIL-101 (Cr) nanocomposites embedded with AuNPs (AuNP/AE-MIL-101 (Cr)) were prepared as surface-enhanced Raman scattering (SERS) substrate. AuNPs are uniformly distributed and stabilized inside the metal-organic framework (MOF), thus forming more SERS hotspots. The SERS performance of AuNP/AE-MIL-101 (Cr) was evaluated using 4-mercaptophenylboronic acid (4-MPBA), 4-mercaptobenzoic acid (4-MBA), benzidine, and rhodamine 6G (R6G). The SERS substrate displays satisfying stability with very low background signal. When benzidine is used as the Raman reporter, the limit of detection (LOD) can reach 6.7 × 10-13 mol·L-1, and the relative standard deviation (RSD) of the intra- and inter-batch repetitive tests is less than 5.2%. On this basis, we developed a method for the detection of human carboxylesterase 1 (hCE 1) in human serum using AuNP/AE-MIL-101 (Cr) nanocomposite as SERS substrate and enzyme-linked immunosorbent assay (ELISA) colorimetric substrate as SERS marker. This method was used to determine hCE 1 in clinical serum samples without complicated sample pretreatment, and the detection results were consistent with the data determined by ELISA. In the concentration range 0.1-120 ng·mL-1, the SERS signal intensity of benzidine at 1609 cm-1 gradually decreases with the increase of hCE 1 concentration (R2 = 0.9948). The average recoveries of hCE 1 in human serum are in the range 84 to 108%, with RSDs lower than 7.7%. By using AuNP/acid etching-MIL-101(Cr) metal organic framework (MOF) as SERS substrate and enzyme-linked immunosorbent assay (ELISA) colorimetric substrate as the SERS marker, a rapid and sensitive method for the determination of human carboxylesterase 1 (hCE1) in human serum samples has been developed.
Collapse
Affiliation(s)
- Jun Feng
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, Guangxi, People's Republic of China.,State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, 541004, People's Republic of China
| | - Hao Lu
- Guangxi Key Laboratory of Green Processing of Sugar Resources, College of Biological and Chemical Engineering, Guangxi University of Science and Technology, No.268 Donghuan Road, Chengzhong District, Liuzhou City, 545006, Guangxi Zhuang Autonomous Region, China.,Provine and Ministry Co-sponsored Collaborative Innovation Center of Sugarcane and Sugar Industry, Nanning, 530004, Guangxi, People's Republic of China
| | - Yu Yang
- Guangxi Key Laboratory of Green Processing of Sugar Resources, College of Biological and Chemical Engineering, Guangxi University of Science and Technology, No.268 Donghuan Road, Chengzhong District, Liuzhou City, 545006, Guangxi Zhuang Autonomous Region, China.,Provine and Ministry Co-sponsored Collaborative Innovation Center of Sugarcane and Sugar Industry, Nanning, 530004, Guangxi, People's Republic of China
| | - Wenyi Huang
- Guangxi Key Laboratory of Green Processing of Sugar Resources, College of Biological and Chemical Engineering, Guangxi University of Science and Technology, No.268 Donghuan Road, Chengzhong District, Liuzhou City, 545006, Guangxi Zhuang Autonomous Region, China.,Provine and Ministry Co-sponsored Collaborative Innovation Center of Sugarcane and Sugar Industry, Nanning, 530004, Guangxi, People's Republic of China
| | - Hao Cheng
- Guangxi Key Laboratory of Green Processing of Sugar Resources, College of Biological and Chemical Engineering, Guangxi University of Science and Technology, No.268 Donghuan Road, Chengzhong District, Liuzhou City, 545006, Guangxi Zhuang Autonomous Region, China.,Provine and Ministry Co-sponsored Collaborative Innovation Center of Sugarcane and Sugar Industry, Nanning, 530004, Guangxi, People's Republic of China
| | - Hongxing Kong
- Guangxi Key Laboratory of Green Processing of Sugar Resources, College of Biological and Chemical Engineering, Guangxi University of Science and Technology, No.268 Donghuan Road, Chengzhong District, Liuzhou City, 545006, Guangxi Zhuang Autonomous Region, China. .,Provine and Ministry Co-sponsored Collaborative Innovation Center of Sugarcane and Sugar Industry, Nanning, 530004, Guangxi, People's Republic of China.
| | - Lijun Li
- Guangxi Key Laboratory of Green Processing of Sugar Resources, College of Biological and Chemical Engineering, Guangxi University of Science and Technology, No.268 Donghuan Road, Chengzhong District, Liuzhou City, 545006, Guangxi Zhuang Autonomous Region, China. .,Provine and Ministry Co-sponsored Collaborative Innovation Center of Sugarcane and Sugar Industry, Nanning, 530004, Guangxi, People's Republic of China.
| |
Collapse
|
16
|
Mallick P, Moreau M, Song G, Efremenko AY, Pendse SN, Creek MR, Osimitz TG, Hines RN, Hinderliter P, Clewell HJ, Lake BG, Yoon M. Development and Application of a Life-Stage Physiologically Based Pharmacokinetic (PBPK) Model to the Assessment of Internal Dose of Pyrethroids in Humans. Toxicol Sci 2021; 173:86-99. [PMID: 31593217 PMCID: PMC6944222 DOI: 10.1093/toxsci/kfz211] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
To address concerns around age-related sensitivity to pyrethroids, a life-stage physiologically based pharmacokinetic (PBPK) model, supported by in vitro to in vivo extrapolation (IVIVE) was developed. The model was used to predict age-dependent changes in target tissue exposure of 8 pyrethroids; deltamethrin (DLM), cis-permethrin (CPM), trans-permethrin, esfenvalerate, cyphenothrin, cyhalothrin, cyfluthrin, and bifenthrin. A single model structure was used based on previous work in the rat. Intrinsic clearance (CLint) of each individual cytochrome P450 or carboxylesterase (CES) enzyme that are active for a given pyrethroid were measured in vitro, then biologically scaled to obtain in vivo age-specific total hepatic CLint. These IVIVE results indicate that, except for bifenthrin, CES enzymes are largely responsible for human hepatic metabolism (>50% contribution). Given the high efficiency and rapid maturation of CESs, clearance of the pyrethroids is very efficient across ages, leading to a blood flow-limited metabolism. Together with age-specific physiological parameters, in particular liver blood flow, the efficient metabolic clearance of pyrethroids across ages results in comparable to or even lower internal exposure in the target tissue (brain) in children than that in adults in response to the same level of exposure to a given pyrethroid (Cmax ratio in brain between 1- and 25-year old = 0.69, 0.93, and 0.94 for DLM, bifenthrin, and CPM, respectively). Our study demonstrated that a life-stage PBPK modeling approach, coupled with IVIVE, provides a robust framework for evaluating age-related differences in pharmacokinetics and internal target tissue exposure in humans for the pyrethroid class of chemicals.
Collapse
Affiliation(s)
| | - Marjory Moreau
- ScitoVation, LLC, Research Triangle Park, North Carolina 27709
| | - Gina Song
- ScitoVation, LLC, Research Triangle Park, North Carolina 27709.,ToxStrategies, Cary, North Carolina 27511
| | | | - Salil N Pendse
- ScitoVation, LLC, Research Triangle Park, North Carolina 27709
| | - Moire R Creek
- Moire Creek Toxicology Consulting Services, Lincoln, California 95648
| | | | - Ronald N Hines
- US EPA, ORD, NHEERL, Research Triangle Park, North Carolina 27709
| | | | | | - Brian G Lake
- Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
| | - Miyoung Yoon
- ScitoVation, LLC, Research Triangle Park, North Carolina 27709.,ToxStrategies, Cary, North Carolina 27511
| |
Collapse
|
17
|
Skottvoll F, Hansen FA, Harrison S, Boger IS, Mrsa A, Restan MS, Stein M, Lundanes E, Pedersen-Bjergaard S, Aizenshtadt A, Krauss S, Sullivan G, Bogen IL, Wilson SR. Electromembrane Extraction and Mass Spectrometry for Liver Organoid Drug Metabolism Studies. Anal Chem 2021; 93:3576-3585. [PMID: 33534551 PMCID: PMC8023518 DOI: 10.1021/acs.analchem.0c05082] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
Liver organoids are emerging tools for precision drug development and toxicity screening. We demonstrate that electromembrane extraction (EME) based on electrophoresis across an oil membrane is suited for segregating selected organoid-derived drug metabolites prior to mass spectrometry (MS)-based measurements. EME allowed drugs and drug metabolites to be separated from cell medium components (albumin, etc.) that could interfere with subsequent measurements. Multiwell EME (parallel-EME) holding 100 μL solutions allowed for simple and repeatable monitoring of heroin phase I metabolism kinetics. Organoid parallel-EME extracts were compatible with ultrahigh-performance liquid chromatography (UHPLC) used to separate the analytes prior to detection. Taken together, liver organoids are well-matched with EME followed by MS-based measurements.
Collapse
Affiliation(s)
- Frøydis
Sved Skottvoll
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Frederik André Hansen
- Department
of Pharmacy, University of Oslo, P.O. Box 1068, Blindern, NO-0316 Oslo, Norway
| | - Sean Harrison
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Ida Sneis Boger
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Ago Mrsa
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Magnus Saed Restan
- Department
of Pharmacy, University of Oslo, P.O. Box 1068, Blindern, NO-0316 Oslo, Norway
| | - Matthias Stein
- Institute
of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, Beethovenstr.
55, DE-38106 Braunschweig, Germany
| | - Elsa Lundanes
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
| | - Stig Pedersen-Bjergaard
- Department
of Pharmacy, University of Oslo, P.O. Box 1068, Blindern, NO-0316 Oslo, Norway
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Aleksandra Aizenshtadt
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Stefan Krauss
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
- Department
of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 1110, Blindern, 0317, Oslo, Norway
| | - Gareth Sullivan
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
- Department
of Pediatric Research, Oslo University Hospital
and University of Oslo, P.O. Box 1112,
Blindern, 0317 Oslo, Norway
| | - Inger Lise Bogen
- Section
for Drug Abuse Research, Department of Forensic Sciences, Oslo University Hospital, P.O. Box 4950, Nydalen, NO-0424 Oslo, Norway
- Institute
of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1103,
Blindern, NO-0317 Oslo, Norway
| | - Steven Ray Wilson
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| |
Collapse
|
18
|
Quantitative analysis of mRNA expression levels of aldo-keto reductase and short-chain dehydrogenase/reductase isoforms in human livers. Drug Metab Pharmacokinet 2020; 35:539-547. [PMID: 33036882 DOI: 10.1016/j.dmpk.2020.08.004] [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: 07/21/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 01/06/2023]
Abstract
The aldo-keto reductase (AKR) and short-chain dehydrogenase/reductase (SDR) superfamilies are responsible for the reduction in compounds containing the aldehyde, ketone, and quinone groups. In humans, 12 AKR isoforms (AKR1A1, AKR1B1, AKR1B10, AKR1B15, AKR1C1, AKR1C2, AKR1C3, AKR1C4, AKR1D1, AKR1E2, AKR7A2, and AKR7A3) and 6 SDR isoforms (CBR1, CBR3, CBR4, HSD11B1, DHRS4, and DCXR) have been found to catalyze the reduction in xenobiotics, but their hepatic expression levels are unclear. The purpose of this study is to determine the absolute mRNA expression levels of these 18 isoforms in the human liver. In 22 human livers, all isoforms, except for AKR1B15, are expressed, and AKR1C2 (on average 1.6 × 106 copy/μg total RNA), AKR1C3 (1.3 × 106), AKR1C1 (1.3 × 106), CBR1 (9.7 × 105), and HSD11B1 (1.1 × 106) are abundant, representing 67% of the total expression of reductases in the liver. The expression levels of AKR1C2, AKR1C3, AKR1C1, CBR1, and HSD11B1 are significantly correlated with each other, except between AKR1C2 and CBR1, suggesting that they might be regulated by common factor(s). In conclusion, this study comprehensively determined the absolute expression of mRNA expression of each AKR and SDR isoform in the human liver.
Collapse
|
19
|
Hedges L, Brown S, MacLeod AK, Moreau M, Yoon M, Creek MR, Osimitz TG, Lake BG. Metabolism of bifenthrin, β-cyfluthrin, λ-cyhalothrin, cyphenothrin and esfenvalerate by rat and human cytochrome P450 and carboxylesterase enzymes. Xenobiotica 2020; 50:1434-1442. [DOI: 10.1080/00498254.2020.1795745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Laura Hedges
- Concept Life Sciences (formerly CXR Biosciences Ltd.), Dundee, UK
| | - Susan Brown
- Concept Life Sciences (formerly CXR Biosciences Ltd.), Dundee, UK
| | | | | | - Miyoung Yoon
- ScitoVation, LLC, Research Triangle Park, NC, USA
| | - Moire R. Creek
- Moire Creek Toxicology Consulting Services, Lincoln, CA, USA
| | | | - Brian G. Lake
- Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
| |
Collapse
|
20
|
Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)-Based Proteomics of Drug-Metabolizing Enzymes and Transporters. Molecules 2020; 25:molecules25112718. [PMID: 32545386 PMCID: PMC7321193 DOI: 10.3390/molecules25112718] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based proteomics is a powerful tool for identifying and quantifying proteins in biological samples, outperforming conventional antibody-based methods in many aspects. LC-MS/MS-based proteomics studies have revealed the protein abundances of many drug-metabolizing enzymes and transporters (DMETs) in tissues relevant to drug metabolism and disposition. Previous studies have consistently demonstrated marked interindividual variability in DMET protein expression, suggesting that varied DMET function is an important contributing factor for interindividual variability in pharmacokinetics (PK) and pharmacodynamics (PD) of medications. Moreover, differential DMET expression profiles were observed across different species and in vitro models. Therefore, caution must be exercised when extrapolating animal and in vitro DMET proteomics findings to humans. In recent years, DMET proteomics has been increasingly utilized for the development of physiologically based pharmacokinetic models, and DMET proteins have also been proposed as biomarkers for prediction of the PK and PD of the corresponding substrate drugs. In sum, despite the existence of many challenges in the analytical technology and data analysis methods of LC-MS/MS-based proteomics, DMET proteomics holds great potential to advance our understanding of PK behavior at the individual level and to optimize treatment regimens via the DMET protein biomarker-guided precision pharmacotherapy.
Collapse
|
21
|
Bhatt P, Bhatt K, Huang Y, Lin Z, Chen S. Esterase is a powerful tool for the biodegradation of pyrethroid insecticides. CHEMOSPHERE 2020; 244:125507. [PMID: 31835049 DOI: 10.1016/j.chemosphere.2019.125507] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/28/2019] [Indexed: 06/10/2023]
Abstract
Agricultural and household applications of pyrethroid insecticides have significantly increased residual concentrations in living cells and environments. The enhanced concentration is toxic for living beings. Pyrethroid hydrolase enzyme (pyrethroid catalyzing esterase) regulates pyrethroid degradation, and has been well reported in various organisms (bacteria, fungi, insects and animals). Hydrolysis mechanisms of these esterases are different from others and properly function at factors viz., optimum temperature, pH and physicochemical environment. Active site of the enzyme contains common amino acids that play important role in pyrethroid catalysis. Immobilization technology emphasizes the development of better reusable efficiency of pyrethroid hydrolases to carry out large-scale applications for complete degradation of pyrethroids from the environments. In this review we have attempted to provide insights of pyrethroid-degrading esterases in different living systems along with complete mechanisms.
Collapse
Affiliation(s)
- Pankaj Bhatt
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Kalpana Bhatt
- Department of Botany and Microbiology, Gurukula Kangri University, Haridwar 249404, Uttarakhand, India
| | - Yaohua Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Ziqiu Lin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Shaohua Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China.
| |
Collapse
|
22
|
Chen Y, Ke M, Xu J, Lin C. Simulation of the Pharmacokinetics of Oseltamivir and Its Active Metabolite in Normal Populations and Patients with Hepatic Cirrhosis Using Physiologically Based Pharmacokinetic Modeling. AAPS PharmSciTech 2020; 21:98. [PMID: 32128656 DOI: 10.1208/s12249-020-1638-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/12/2020] [Indexed: 12/15/2022] Open
Abstract
Oseltamivir is a neuraminidase inhibitor widely used to treat and prevent influenza A and B infections, although its safety and pharmacokinetics have not been evaluated in patients with severe hepatic impairment. A physiologically based pharmacokinetic (PBPK) model of the prodrug oseltamivir and its active metabolite, oseltamivir carboxylate (OC), was established and validated to simulate their disposition in adults and predict the exposure in patients with Child-Pugh C cirrhosis (CP-C). The simulated results from PBPK modeling and the observed data after oral administration of various oseltamivir regimens were consistent according to the fold error values of less than 2. Furthermore, the clinical observations published in the literature were comparable with our pharmacokinetic predictions. In patients with CP-C, the oseltamivir Cmax was approximately 2-fold increased, and its AUC was approximately 6-fold higher compared with those in normal subjects. In contrast, the AUC of OC in CP-C patients did not differ significantly from that in normal subjects, whereas its Cmax was reduced by approximately 30% in the patients. Examination of drug exposure in different health conditions indicated that the oseltamivir exposure was significantly increased in conditions with elevated cirrhosis severity, which might be associated with a higher risk of adverse drug effects, e.g., neuropsychiatric adverse events (NPAEs). In conclusion, the pharmacokinetics of oseltamivir and OC were correctly predicted by PBPK modeling. The model further predicted that the pharmacokinetics of oseltamivir might be altered in liver cirrhosis, depending on the degree of severity.
Collapse
|
23
|
Alsmadi MM, Alfarah MQ, Albderat J, Alsalaita G, AlMardini R, Hamadi S, Al‐Ghazawi A, Abu‐Duhair O, Idkaidek N. The development of a population physiologically based pharmacokinetic model for mycophenolic mofetil and mycophenolic acid in humans using data from plasma, saliva, and kidney tissue. Biopharm Drug Dispos 2019; 40:325-340. [DOI: 10.1002/bdd.2206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 09/22/2019] [Accepted: 10/09/2019] [Indexed: 02/06/2023]
Affiliation(s)
| | | | - Jawaher Albderat
- Queen Rania Abdullah Children Hospital, Royal Medical Services Amman Jordan
| | - Ghazi Alsalaita
- Queen Rania Abdullah Children Hospital, Royal Medical Services Amman Jordan
| | - Reham AlMardini
- Queen Rania Abdullah Children Hospital, Royal Medical Services Amman Jordan
| | - Salim Hamadi
- Deparment of Pharmaceutical Technology, Faculty of PharmacyUniversity of Petra Amman Jordan
| | | | - Omar Abu‐Duhair
- Deparment of Pharmaceutical Technology, Faculty of PharmacyUniversity of Petra Amman Jordan
| | - Nasir Idkaidek
- Deparment of Pharmaceutical Technology, Faculty of PharmacyUniversity of Petra Amman Jordan
| |
Collapse
|
24
|
Kailass K, Sadovski O, Capello M, Kang Y, Fleming JB, Hanash SM, Beharry AA. Measuring human carboxylesterase 2 activity in pancreatic cancer patient-derived xenografts using a ratiometric fluorescent chemosensor. Chem Sci 2019; 10:8428-8437. [PMID: 31803422 PMCID: PMC6844279 DOI: 10.1039/c9sc00283a] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/28/2019] [Indexed: 12/17/2022] Open
Abstract
Irinotecan-based therapy is a common treatment for pancreatic cancer. To elicit its anticancer activity, the drug requires first the hydrolysis action of the enzyme human carboxylesterase 2 (hCES2). It has been established that pancreatic cancer patients have various levels of hCES2, whereby patients having low levels respond poorer to Irinotecan than patients with higher levels, suggesting that hCES2 can be used to predict response. However, current methods that measure hCES2 activity are inaccurate, complex or lengthy, thus being incompatible for use in a clinical setting. Here, we developed a small molecule ratiometric fluorescent chemosensor that accurately measures hCES2 activity in a single-step within complex mixtures. Our chemosensor is highly selective for hCES2 over hCES1, cell permeable and can measure hCES2 activity in pancreatic cancer patient-derived xenografts. Given the simplicity, accuracy and tissue compatibility of our assay, we anticipate our chemosensor can be used to predict patient response to Irinotecan-based therapy.
Collapse
Affiliation(s)
- Karishma Kailass
- Department of Chemical and Physical Sciences , University of Toronto Mississauga , Mississauga , ON L5L 1C6 , Canada .
| | - Oleg Sadovski
- Department of Chemical and Physical Sciences , University of Toronto Mississauga , Mississauga , ON L5L 1C6 , Canada .
| | - Michela Capello
- Department of Clinical Cancer Prevention , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Ya'an Kang
- Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Jason B Fleming
- Department of Gastrointestinal Oncology , H. Lee Moffitt Cancer Center , Tampa , FL , USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Andrew A Beharry
- Department of Chemical and Physical Sciences , University of Toronto Mississauga , Mississauga , ON L5L 1C6 , Canada .
| |
Collapse
|
25
|
Song PF, Zhu YD, Ma HY, Wang YN, Wang DD, Zou LW, Ge GB, Yang L. Discovery of natural pentacyclic triterpenoids as potent and selective inhibitors against human carboxylesterase 1. Fitoterapia 2019; 137:104199. [DOI: 10.1016/j.fitote.2019.104199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/30/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022]
|
26
|
Di L. The Impact of Carboxylesterases in Drug Metabolism and Pharmacokinetics. Curr Drug Metab 2019; 20:91-102. [PMID: 30129408 PMCID: PMC6635651 DOI: 10.2174/1389200219666180821094502] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/03/2018] [Accepted: 08/08/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Carboxylesterases (CES) play a critical role in catalyzing hydrolysis of esters, amides, carbamates and thioesters, as well as bioconverting prodrugs and soft drugs. The unique tissue distribution of CES enzymes provides great opportunities to design prodrugs or soft drugs for tissue targeting. Marked species differences in CES tissue distribution and catalytic activity are particularly challenging in human translation. METHODS Review and summarization of CES fundamentals and applications in drug discovery and development. RESULTS Human CES1 is one of the most highly expressed drug metabolizing enzymes in the liver, while human intestine only expresses CES2. CES enzymes have moderate to high inter-individual variability and exhibit low to no expression in the fetus, but increase substantially during the first few months of life. The CES genes are highly polymorphic and some CES genetic variants show significant influence on metabolism and clinical outcome of certain drugs. Monkeys appear to be more predictive of human pharmacokinetics for CES substrates than other species. Low risk of clinical drug-drug interaction is anticipated for CES, although they should not be overlooked, particularly interaction with alcohols. CES enzymes are moderately inducible through a number of transcription factors and can be repressed by inflammatory cytokines. CONCLUSION Although significant advances have been made in our understanding of CESs, in vitro - in vivo extrapolation of clearance is still in its infancy and further exploration is needed. In vitro and in vivo tools are continuously being developed to characterize CES substrates and inhibitors.
Collapse
Affiliation(s)
- Li Di
- Pfizer Inc., Eastern Point Road, Groton, Connecticut, CT 06354, United States
| |
Collapse
|
27
|
Tian Z, Ding L, Li K, Song Y, Dou T, Hou J, Tian X, Feng L, Ge G, Cui J. Rational Design of a Long-Wavelength Fluorescent Probe for Highly Selective Sensing of Carboxylesterase 1 in Living Systems. Anal Chem 2019; 91:5638-5645. [PMID: 30968686 DOI: 10.1021/acs.analchem.8b05417] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Rational design of practical probes with excellent specificity and improved optical properties for a particular enzyme is always a big challenge. Herein, a practical and highly specific fluorescent probe for carboxylesterase 1 (CES1) was rationally designed using meso-carboxyl-BODIPY as the basic fluorophore based on the substrate preference and catalytic properties of CES1. Following molecular docking-based virtual screening combined with reaction phenotyping-based experimental screening, we found that MMB (probe 7) exhibited the optimal combination of sensitivity and specificity toward human CES1 in contrast to other ester derivatives. Under physiological conditions, MMB could be readily hydrolyzed by CES1 and release MCB; such biotransformation brought great changes in the electronic properties at the meso position of the fluorophore and triggered a dramatic increase in fluorescence emission around 595 nm. Moreover, MMB was cell membrane permeable and was successfully applied to monitor the real activities of CES1 in various biological samples including living cells, tissue slices, organs, and zebrafish. In summary, this study showed a good example for constructing specific fluorescent probe(s) for a target enzyme and also provided a practical and sensitive tool for real-time sensing of CES1 activities in complicated biological samples. All these findings would strongly facilitate high-throughput screening of CES1 modulators and the studies on CES1-associated physiological and pathological processes.
Collapse
Affiliation(s)
- Zhenhao Tian
- State Key Laboratory of Fine Chemicals , Dalian University of Technology , Dalian , 116024 , China
| | - Lele Ding
- State Key Laboratory of Fine Chemicals , Dalian University of Technology , Dalian , 116024 , China
| | - Kun Li
- School of Life Science and Medicine , Dalian University of Technology , Panjin , 124221 , China
| | - Yunqing Song
- Institute of Interdisciplinary Integrative Medicine Research , Shanghai University of Traditional Chinese Medicine , Shanghai , 201203 , China
| | - Tongyi Dou
- School of Life Science and Medicine , Dalian University of Technology , Panjin , 124221 , China
| | - Jie Hou
- Dalian Medical University , Dalian , 116044 , China
| | - Xiangge Tian
- Dalian Medical University , Dalian , 116044 , China
| | - Lei Feng
- Dalian Medical University , Dalian , 116044 , China
| | - Guangbo Ge
- Institute of Interdisciplinary Integrative Medicine Research , Shanghai University of Traditional Chinese Medicine , Shanghai , 201203 , China
| | - Jingnan Cui
- State Key Laboratory of Fine Chemicals , Dalian University of Technology , Dalian , 116024 , China
| |
Collapse
|
28
|
Label-free absolute protein quantification with data-independent acquisition. J Proteomics 2019; 200:51-59. [PMID: 30880166 DOI: 10.1016/j.jprot.2019.03.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/19/2019] [Accepted: 03/06/2019] [Indexed: 02/05/2023]
Abstract
Despite data-independent acquisition (DIA) has been increasingly used for relative protein quantification, DIA-based label-free absolute quantification method has not been fully established. Here we present a novel DIA method using the TPA algorithm (DIA-TPA) for the absolute quantification of protein expressions in human liver microsomal and S9 samples. To validate this method, both data-dependent acquisition (DDA) and DIA experiments were conducted on 36 individual human liver microsome and S9 samples. The MS2-based DIA-TPA was able to quantify approximately twice as many proteins as the MS1-based DDA-TPA method, whereas protein concentrations determined by the two approaches were comparable. To evaluate the accuracy of the DIA-TPA method, we absolutely quantified carboxylesterase 1 concentrations in human liver S9 fractions using an established SILAC internal standard-based proteomic assay; the SILAC results were consistent with those obtained from DIA-TPA analysis. Finally, we employed a unique algorithm in DIA-TPA to distribute the MS signals from shared peptides to individual proteins or isoforms and successfully applied the method to the absolute quantification of several drug-metabolizing enzymes in human liver microsomes. In sum, the DIA-TPA method not only can absolutely quantify entire proteomes and specific proteins, but also has the capability quantifying proteins with shared peptides. SIGNIFICANCE: Data independent acquisition (DIA) has emerged as a powerful approach for relative protein quantification at the whole proteome level. However, DIA-based label-free absolute protein quantification (APQ) method has not been fully established. In the present study, we present a novel DIA-based label-free APQ approach, named DIA-TPA, with the capability absolutely quantifying proteins with shared peptides. The method was validated by comparing the quantification results of DIA-TPA with that obtained from stable isotope-labeled internal standard-based proteomic assays.
Collapse
|
29
|
Ding L, Tian Z, Hou J, Dou T, Jin Q, Wang D, Zou L, Zhu Y, Song Y, Cui J, Ge G. Sensing carboxylesterase 1 in living systems by a practical and isoform-specific fluorescent probe. CHINESE CHEM LETT 2019. [DOI: 10.1016/j.cclet.2018.12.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Li M, Zhai C, Wang S, Huang W, Liu Y, Li Z. Detection of carboxylesterase by a novel hydrosoluble near-infrared fluorescence probe. RSC Adv 2019; 9:40689-40693. [PMID: 35542681 PMCID: PMC9076276 DOI: 10.1039/c9ra08150j] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/03/2019] [Indexed: 01/02/2023] Open
Abstract
A novel hydrosoluble near-infrared fluorescence off–on probe has been developed for detecting carboxylesterase activity.
Collapse
Affiliation(s)
- Mengyao Li
- Nutrition & Health Research Institute
- COFCO Corporation
- Beijing Key Laboratory of Nutrition & Health and Food Safety
- Beijing 102209
- China
| | - Chen Zhai
- Nutrition & Health Research Institute
- COFCO Corporation
- Beijing Key Laboratory of Nutrition & Health and Food Safety
- Beijing 102209
- China
| | - Shuya Wang
- Nutrition & Health Research Institute
- COFCO Corporation
- Beijing Key Laboratory of Nutrition & Health and Food Safety
- Beijing 102209
- China
| | - Weixia Huang
- Nutrition & Health Research Institute
- COFCO Corporation
- Beijing Key Laboratory of Nutrition & Health and Food Safety
- Beijing 102209
- China
| | - Yunguo Liu
- College of Life Science and Technology
- Xinjiang University
- Urumqi 830002
- China
| | - Zhao Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710062
- China
| |
Collapse
|
31
|
Human carboxylesterases: a comprehensive review. Acta Pharm Sin B 2018; 8:699-712. [PMID: 30245959 PMCID: PMC6146386 DOI: 10.1016/j.apsb.2018.05.005] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Mammalian carboxylesterases (CEs) are key enzymes from the serine hydrolase superfamily. In the human body, two predominant carboxylesterases (CES1 and CES2) have been identified and extensively studied over the past decade. These two enzymes play crucial roles in the metabolism of a wide variety of endogenous esters, ester-containing drugs and environmental toxicants. The key roles of CES in both human health and xenobiotic metabolism arouse great interest in the discovery of potent CES modulators to regulate endobiotic metabolism or to improve the efficacy of ester drugs. This review covers the structural and catalytic features of CES, tissue distributions, biological functions, genetic polymorphisms, substrate specificities and inhibitor properties of CES1 and CES2, as well as the significance and recent progress on the discovery of CES modulators. The information presented here will help pharmacologists explore the relevance of CES to human diseases or to assign the contribution of certain CES in xenobiotic metabolism. It will also facilitate medicinal chemistry efforts to design prodrugs activated by a given CES isoform, or to develop potent and selective modulators of CES for potential biomedical applications.
Collapse
|
32
|
Carboxylesterase-2-Selective Two-Photon Ratiometric Probe Reveals Decreased Carboxylesterase-2 Activity in Breast Cancer Cells. Anal Chem 2018; 90:9465-9471. [DOI: 10.1021/acs.analchem.8b02101] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
33
|
Hedges L, Brown S, MacLeod AK, Vardy A, Doyle E, Song G, Moreau M, Yoon M, Osimitz TG, Lake BG. Metabolism of deltamethrin and cis- and trans-permethrin by human expressed cytochrome P450 and carboxylesterase enzymes. Xenobiotica 2018; 49:521-527. [DOI: 10.1080/00498254.2018.1474283] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Laura Hedges
- Concept Life Sciences (formerly CXR Biosciences Ltd.), Dundee, UK
| | - Susan Brown
- Concept Life Sciences (formerly CXR Biosciences Ltd.), Dundee, UK
| | | | - Audrey Vardy
- Concept Life Sciences (formerly CXR Biosciences Ltd.), Dundee, UK
| | - Edward Doyle
- Concept Life Sciences (formerly CXR Biosciences Ltd.), Dundee, UK
| | - Gina Song
- ScitoVation LLC Research Triangle Park, NC, USA
| | | | - Miyoung Yoon
- ToxStrategies LLC Research Triangle Park, NC, USA
| | | | - Brian G. Lake
- Concept Life Sciences (formerly CXR Biosciences Ltd.), Dundee, UK
- Centre for Toxicology University of Surrey, Surrey, UK
| |
Collapse
|
34
|
Wang J, Chen Q, Tian N, Zhu W, Zou H, Wang X, Li X, Fan X, Jiang G, Tang BZ. A fast responsive, highly selective and light-up fluorescent probe for the two-photon imaging of carboxylesterase in living cells. J Mater Chem B 2018; 6:1595-1599. [DOI: 10.1039/c8tb00147b] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A fast responsive and two photon fluorescent probe (HCyNAc) for carboxylesterase (CaE) has been designed and used for the two-photon imaging of the endogenous CaE level in living HeLa cells under 800 nm NIR excitation.
Collapse
Affiliation(s)
- Jianguo Wang
- Key Laboratory of Organo-Pharmaceutical Chemistry
- Gannan Normal University
- Ganzhou 341000
- P. R. China
| | - Qingqing Chen
- Key Laboratory of Organo-Pharmaceutical Chemistry
- Gannan Normal University
- Ganzhou 341000
- P. R. China
| | - Na Tian
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials
- Technical Institute of Physics and Chemistry
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Wenping Zhu
- Key Laboratory of Organo-Pharmaceutical Chemistry
- Gannan Normal University
- Ganzhou 341000
- P. R. China
| | - Hang Zou
- Department of Chemistry
- Institute for Advanced Study
- Institute of Molecular Functional Materials, and State Key Laboratory of Molecular Neuroscience
- The Hong Kong University of Science & Technology
- Kowloon
| | - Xuesong Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials
- Technical Institute of Physics and Chemistry
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Xiaokang Li
- Key Laboratory of Organo-Pharmaceutical Chemistry
- Gannan Normal University
- Ganzhou 341000
- P. R. China
| | - Xiaolin Fan
- Key Laboratory of Organo-Pharmaceutical Chemistry
- Gannan Normal University
- Ganzhou 341000
- P. R. China
| | - Guoyu Jiang
- Key Laboratory of Organo-Pharmaceutical Chemistry
- Gannan Normal University
- Ganzhou 341000
- P. R. China
| | - Ben Zhong Tang
- Department of Chemistry
- Institute for Advanced Study
- Institute of Molecular Functional Materials, and State Key Laboratory of Molecular Neuroscience
- The Hong Kong University of Science & Technology
- Kowloon
| |
Collapse
|
35
|
Jiang J, Chen X, Zhong D. Predominant contributions of carboxylesterase 1 and 2 in hydrolysis of anordrin in humans. Xenobiotica 2017; 48:533-540. [DOI: 10.1080/00498254.2017.1333658] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Jinfang Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China and
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China and
- University of Chinese Academy of Sciences, Beijing, China
| | - Dafang Zhong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China and
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
36
|
In Vitro – In Vivo Extrapolation of Intestinal Availability for Carboxylesterase Substrates Using Portal Vein–Cannulated Monkey. J Pharm Sci 2017; 106:898-905. [DOI: 10.1016/j.xphs.2016.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/04/2016] [Accepted: 12/02/2016] [Indexed: 10/20/2022]
|
37
|
Lei W, Wang DD, Dou TY, Hou J, Feng L, Yin H, Luo Q, Sun J, Ge GB, Yang L. Assessment of the inhibitory effects of pyrethroids against human carboxylesterases. Toxicol Appl Pharmacol 2017; 321:48-56. [PMID: 28242322 DOI: 10.1016/j.taap.2017.02.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/12/2017] [Accepted: 02/22/2017] [Indexed: 12/12/2022]
Abstract
Pyrethroids are broad-spectrum insecticides that widely used in many countries, while humans may be exposed to these toxins by drinking or eating pesticide-contaminated foods. This study aimed to investigate the inhibitory effects of six commonly used pyrethroids against two major human carboxylesterases (CES) including CES1 and CES2. Three optical probe substrates for CES1 (DME, BMBT and DMCB) and a fluorescent probe substrate for CES2 (DDAB) were used to characterize the inhibitory effects of these pyrethroids. The results demonstrated that most of the tested pyrethroids showed moderate to weak inhibitory effects against both CES1 and CES2, but deltamethrin displayed strong inhibition towards CES1. The IC50 values of deltamethrin against CES1-mediated BMBT, DME, and DMCB hydrolysis were determined as 1.58μM, 2.39μM, and 3.3μM, respectively. Moreover, deltamethrin was cell membrane permeable and capable of inhibition endogenous CES1 in living cells. Further investigation revealed that deltamethrin inhibited CES1-mediated BMBT hydrolysis via competitive manner but noncompetitively inhibited DME or DMCB hydrolysis. The inhibition behaviors of deltamethrin against CES1 were also studied by molecular docking simulation. The results demonstrated that CES1 had at least two different ligand-binding sites, one was the DME site and another was the BMBT site which was identical to the binding site of deltamethrin. In summary, deltamethrin was a strong reversible inhibitor against CES1 and it could tightly bind on CES1 at the same ligand-binding site as BMBT. These findings are helpful for the deep understanding of the interactions between xenobiotics and CES1.
Collapse
Affiliation(s)
- Wei Lei
- The Second Affiliated Hospital of Dalian Medical University, Dalian 110623, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Dan-Dan Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Tong-Yi Dou
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jie Hou
- Dalian Medical University, Dalian 116044, China
| | - Liang Feng
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Heng Yin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Qun Luo
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100080, China
| | - Jie Sun
- The Second Affiliated Hospital of Dalian Medical University, Dalian 110623, China
| | - Guang-Bo Ge
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Ling Yang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| |
Collapse
|
38
|
Boberg M, Vrana M, Mehrotra A, Pearce RE, Gaedigk A, Bhatt DK, Leeder JS, Prasad B. Age-Dependent Absolute Abundance of Hepatic Carboxylesterases (CES1 and CES2) by LC-MS/MS Proteomics: Application to PBPK Modeling of Oseltamivir In Vivo Pharmacokinetics in Infants. Drug Metab Dispos 2017; 45:216-223. [PMID: 27895113 PMCID: PMC5267516 DOI: 10.1124/dmd.116.072652] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/22/2016] [Indexed: 12/17/2022] Open
Abstract
The age-dependent absolute protein abundance of carboxylesterase (CES) 1 and CES2 in human liver was investigated and applied to predict infant pharmacokinetics (PK) of oseltamivir. The CES absolute protein abundance was determined by liquid chromatography-tandem mass spectrometry proteomics in human liver microsomal and cytosolic fractions prepared from tissue samples obtained from 136 pediatric donors and 35 adult donors. Two surrogate peptides per protein were selected for the quantification of CES1 and CES2 protein abundance. Purified CES1 and CES2 protein standards were used as calibrators, and the heavy labeled peptides were used as the internal standards. In hepatic microsomes, CES1 and CES2 abundance (in picomoles per milligram total protein) increased approximately 5-fold (315.2 vs. 1664.4) and approximately 3-fold (59.8 vs. 174.1) from neonates to adults, respectively. CES1 protein abundance in liver cytosol also showed age-dependent maturation. Oseltamivir carboxylase activity was correlated with protein abundance in pediatric and adult liver microsomes. The protein abundance data were then used to model in vivo PK of oseltamivir in infants using pediatric physiologically based PK modeling and incorporating the protein abundance-based ontogeny function into the existing pediatric Simcyp model. The predicted pediatric area under the curve, maximal plasma concentration, and time for maximal plasma concentration values were below 2.1-fold of the clinically observed values, respectively.
Collapse
Affiliation(s)
- Mikael Boberg
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.B., M.V., A.M., D.K.B., B.P.); Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.B.); Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.); and School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.)
| | - Marc Vrana
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.B., M.V., A.M., D.K.B., B.P.); Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.B.); Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.); and School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.)
| | - Aanchal Mehrotra
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.B., M.V., A.M., D.K.B., B.P.); Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.B.); Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.); and School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.)
| | - Robin E Pearce
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.B., M.V., A.M., D.K.B., B.P.); Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.B.); Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.); and School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.)
| | - Andrea Gaedigk
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.B., M.V., A.M., D.K.B., B.P.); Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.B.); Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.); and School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.)
| | - Deepak Kumar Bhatt
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.B., M.V., A.M., D.K.B., B.P.); Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.B.); Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.); and School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.)
| | - J Steven Leeder
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.B., M.V., A.M., D.K.B., B.P.); Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.B.); Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.); and School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.)
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.B., M.V., A.M., D.K.B., B.P.); Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.B.); Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.); and School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri (R.E.P., A.G., J.S.L.)
| |
Collapse
|
39
|
Samant S, Jiang XL, Peletier LA, Shuldiner AR, Horenstein RB, Lewis JP, Lesko LJ, Schmidt S. Identifying clinically relevant sources of variability: The clopidogrel challenge. Clin Pharmacol Ther 2016; 101:264-273. [DOI: 10.1002/cpt.459] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/10/2016] [Accepted: 08/12/2016] [Indexed: 12/14/2022]
Affiliation(s)
- S Samant
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology; University of Florida at Lake Nona; Orlando Florida USA
| | - XL Jiang
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology; University of Florida at Lake Nona; Orlando Florida USA
| | - LA Peletier
- Mathematical Institute; Leiden University; PB 9512 2300 RA Leiden The Netherlands
| | - AR Shuldiner
- Division of Endocrinology, Diabetes and Nutrition; University of Maryland School of Medicine; Baltimore Maryland USA
| | - RB Horenstein
- Division of Endocrinology, Diabetes and Nutrition; University of Maryland School of Medicine; Baltimore Maryland USA
| | - JP Lewis
- Division of Endocrinology, Diabetes and Nutrition; University of Maryland School of Medicine; Baltimore Maryland USA
| | - LJ Lesko
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology; University of Florida at Lake Nona; Orlando Florida USA
| | - S Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology; University of Florida at Lake Nona; Orlando Florida USA
| |
Collapse
|
40
|
Nelveg-Kristensen KE, Bie P, Ferrero L, Bjerre D, Bruun NE, Egfjord M, Rasmussen HB, Hansen PR. Pharmacodynamic Impact of Carboxylesterase 1 Gene Variants in Patients with Congestive Heart Failure Treated with Angiotensin-Converting Enzyme Inhibitors. PLoS One 2016; 11:e0163341. [PMID: 27662362 PMCID: PMC5035013 DOI: 10.1371/journal.pone.0163341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 09/07/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Variation in the carboxylesterase 1 gene (CES1) may contribute to the efficacy of ACEIs. Accordingly, we examined the impact of CES1 variants on plasma angiotensin II (ATII)/angiotensin I (ATI) ratio in patients with congestive heart failure (CHF) that underwent ACEI dose titrations. Five of these variants have previously been associated with drug response or increased CES1 expression, i.e., CES1 copy number variation, the variant of the duplicated CES1 gene with high transcriptional activity, rs71647871, rs2244613, and rs3815583. Additionally, nine variants, representatives of CES1Var, and three other CES1 variants were examined. METHODS Patients with CHF, and clinical indication for ACEIs were categorized according to their CES1 genotype. Differences in mean plasma ATII/ATI ratios between genotype groups after ACEI dose titration, expressed as the least square mean (LSM) with 95% confidence intervals (CIs), were assessed by analysis of variance. RESULTS A total of 200 patients were recruited and 127 patients (63.5%) completed the study. The mean duration of the CHF drug dose titration was 6.2 (SD 3.6) months. After ACEI dose titration, there was no difference in mean plasma ATII/ATI ratios between subjects with the investigated CES1 variants, and only one previously unexplored variation (rs2302722) qualified for further assessment. In the fully adjusted analysis of effects of rs2302722 on plasma ATII/ATI ratios, the difference in mean ATII/ATI ratio between the GG genotype and the minor allele carriers (GT and TT) was not significant, with a relative difference in LSMs of 0.67 (95% CI 0.43-1.07; P = 0.10). Results of analyses that only included enalapril-treated patients remained non-significant after Bonferroni correction for multiple parallel comparisons (difference in LSM 0.60 [95% CI 0.37-0.98], P = 0.045). CONCLUSION These findings indicate that the included single variants of CES1 do not significantly influence plasma ATII/ATI ratios in CHF patients treated with ACEIs and are unlikely to be primary determinants of ACEI efficacy.
Collapse
Affiliation(s)
| | - Peter Bie
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Laura Ferrero
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - Ditte Bjerre
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - Niels E. Bruun
- Department of Cardiology, Gentofte University Hospital, Gentofte, Denmark
- Clinical Institute, Aalborg University, Aalborg, Denmark
| | - Martin Egfjord
- Department of Nephrology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Henrik B. Rasmussen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - Peter R. Hansen
- Department of Cardiology, Gentofte University Hospital, Gentofte, Denmark
| | | |
Collapse
|
41
|
Yap CG, Zaini A, Othman I. Targeted CYP2E1 quantification and its correlation to currently acceptable clinical biochemical indices. ACTA ACUST UNITED AC 2016; 23:15. [PMID: 27376033 PMCID: PMC4929739 DOI: 10.1186/s40709-016-0052-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 06/13/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND The Cytochrome P450 enzymes are commonly known for their major role in metabolism. Besides its metabolic role, CYP2E1 gene expression has been associated with the onset of diabetic nephropathy. CYP2E1 protein elevation has also been reported to be responsible for the production of reactive oxygen species. The aims of this study were (i) to optimize and validate a targeted proteomic approach for quantitating CYP2E1 and validating it as a suitable clinical test, (ii) to investigate the concurrency between ESI-LCMS-MS quantitated circulating CYP2E1 and gold standard indices in the context of outpatient point-of-care clinical settings involving various groups of diabetic patients and (iii) to investigate the concurrency profile of circulating CYP2E1 protein, CYP2E1 gene expression and reactive oxygen species (ROS). This is a cross sectional study involving three groups of subjects (n = 166): control, pre-diabetes, and diabetes. We optimized a targeted proteomic approach for absolute quantification of CYP2E1. "YPEIEEK" and "GTVVVPTLYDNQEFPDPEK" were the representative peptides of CYP2E1 for our analytical method. Deuterated forms of "YPEIEEK" and "GTVVVPTLYDNQEFPDPEK" were used as internal standards. Lymphocytes were isolated from whole blood, microsomes were prepared, followed by in-solution digestion for production of tryptic peptides. Amounts of "YPEIEEK" and "GTVVVPTLYDNQEFPDPEK" from patients' samples were calculated from a calibration curve. RESULTS "YPEIEEK" is a unique and reliable representative peptide for CYP2E1 quantification. "GTVVVPTLYDNQEFPDPEK" showed poor reproducibility and sensitivity. Incremental amounts of CYP2E1 protein in the peripheral circulation clearly showed concurrency with CYP2E1 gene expression and ROS levels in our study population. Elevations of CYP2E1 were observed even when gold standard clinical indicator for glycemic control (HbA1c) was within normal reference limits. Quantitated amounts of CYP2E1 protein in the pre-diabetes and diabetes groups showed significant difference relative to control group (p < 0.001). No significant differences were observed between the medians of pre-diabetes and diabetes groups (p = 0.870). CONCLUSIONS CYP2E1 protein in peripheral blood can be reliably quantitated by the validated targeted proteomic approach method. Quantifiable amounts of CYP2E1 preceded abnormal HbA1C levels which indicates quantitation of CYP2E1 could be useful as an additional tool for early indication of diabetic risks and it complications.
Collapse
Affiliation(s)
- Christina Gertrude Yap
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Pesiaran Lagoon Selatan, 46150 Petaling Jaya, Selangor Malaysia
| | - Anuar Zaini
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Pesiaran Lagoon Selatan, 46150 Petaling Jaya, Selangor Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Pesiaran Lagoon Selatan, 46150 Petaling Jaya, Selangor Malaysia
| |
Collapse
|
42
|
Prognostic impact of carboxylesterase 1 gene variants in patients with congestive heart failure treated with angiotensin-converting enzyme inhibitors. Pharmacogenet Genomics 2016; 26:169-177. [PMID: 26761119 DOI: 10.1097/fpc.0000000000000203] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Most angiotensin-converting enzyme inhibitors (ACEIs) are prodrugs activated by carboxylesterase 1 (CES1). We investigated the prognostic importance of CES1 gene (CES1) copy number variation and the rs3815583 single-nucleotide polymorphism in CES1 among ACEI-treated patients with congestive heart failure (CHF). METHODS Danish patients with chronic CHF enrolled in the previously reported Echocardiography and Heart Outcome Study were categorized according to their CES1 variants and followed up for up to 10 years. Risk for cardiovascular death and all-cause death was modeled by Cox proportional hazard analyses. RESULTS A total of 491 ACEI-treated patients were included in the analyses. After a mean follow-up of 5.5 years, we found no difference in the risk for cardiovascular death and all-cause death between patients having three [hazard ratios (HRs) 1.06 (95% confidence interval (CI) 0.77-1.45) and 1.16 (95% CI 0.88-1.52)] or four [HRs 0.88 (95% CI 0.39-2.01) and 1.37 (95% CI 0.74-2.54)] CES1 copies and those with two copies, respectively. Similarly, no difference in the risk for cardiovascular and all-cause death was found for patients heterozygous [HRs 0.91 (95% CI 0.70-1.19) and 0.88 (95% CI 0.69-1.12)] or homozygous [HRs 0.58 (95% CI 0.30-1.15) and 0.82 (95% CI 0.48-1.39)] for the rs3815583 minor allele versus patients homozygous for the major allele. The active promoter of CES1A2 and the rs71647871 single-nucleotide polymorphism minor allele were detected at very low frequencies. CONCLUSION This study did not support the use of CES1 copy number variation or rs3815583 as a predictor of fatal outcomes in ACEI-treated patients with CHF.
Collapse
|
43
|
Wang DD, Jin Q, Hou J, Feng L, Li N, Li SY, Zhou Q, Zou LW, Ge GB, Wang JG, Yang L. Highly sensitive and selective detection of human carboxylesterase 1 activity by liquid chromatography with fluorescence detection. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1008:212-218. [DOI: 10.1016/j.jchromb.2015.11.046] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 11/29/2022]
|
44
|
Wang DD, Jin Q, Zou LW, Hou J, Lv X, Lei W, Cheng HL, Ge GB, Yang L. A bioluminescent sensor for highly selective and sensitive detection of human carboxylesterase 1 in complex biological samples. Chem Commun (Camb) 2016; 52:3183-6. [DOI: 10.1039/c5cc09874b] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A highly selective and sensitive bioluminescent sensor (DME) for real-time monitoring of human carboxylesterase 1 (hCE1) activities in complex biological samples and bio-imaging of endogenous hCE1 in living cells.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Qiang Jin
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Li-Wei Zou
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Jie Hou
- Dalian Medical University
- Dalian
- China
| | - Xia Lv
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Wei Lei
- Dalian Medical University
- Dalian
- China
| | - Hai-Ling Cheng
- Cancer Institute
- The Second Hospital of Dalian Medical University
- Dalian
- China
| | - Guang-Bo Ge
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Ling Yang
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| |
Collapse
|
45
|
Lv X, Wang DD, Feng L, Wang P, Zou LW, Hao DC, Hou J, Cui JN, Ge GB, Yang L. A highly selective marker reaction for measuring the activity of human carboxylesterase 1 in complex biological samples. RSC Adv 2016. [DOI: 10.1039/c5ra23614b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
NMHN hydrolysis was found to be a highly selective marker reaction for sensing the activity of human carboxylesterase 1 (hCE1).
Collapse
Affiliation(s)
- Xia Lv
- Laboratory of Pharmaceutical Resource Discovery
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| | - Dan-Dan Wang
- Laboratory of Pharmaceutical Resource Discovery
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| | - Lei Feng
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- Dalian 116012
- China
| | - Ping Wang
- Laboratory of Pharmaceutical Resource Discovery
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| | - Li-Wei Zou
- Laboratory of Pharmaceutical Resource Discovery
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| | | | - Jie Hou
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- Dalian 116012
- China
- Dalian Medical University
| | - Jing-Nan Cui
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- Dalian 116012
- China
| | - Guang-Bo Ge
- Laboratory of Pharmaceutical Resource Discovery
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| | - Ling Yang
- Laboratory of Pharmaceutical Resource Discovery
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| |
Collapse
|
46
|
Jin Q, Feng L, Wang DD, Dai ZR, Wang P, Zou LW, Liu ZH, Wang JY, Yu Y, Ge GB, Cui JN, Yang L. A Two-Photon Ratiometric Fluorescent Probe for Imaging Carboxylesterase 2 in Living Cells and Tissues. ACS APPLIED MATERIALS & INTERFACES 2015; 7:28474-28481. [PMID: 26641926 DOI: 10.1021/acsami.5b09573] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In this study, a two-photon ratiometric fluorescent probe NCEN has been designed and developed for highly selective and sensitive sensing of human carboxylesterase 2 (hCE2) based on the catalytic properties and substrate preference of hCE2. Upon addition of hCE2, the probe could be readily hydrolyzed to release 4-amino-1,8-naphthalimide (NAH), which brings remarkable red-shift in fluorescence (90 nm) spectrum. The newly developed probe exhibits good specificity, ultrahigh sensitivity, and has been successfully applied to determine the real activities of hCE2 in complex biological samples such as cell and tissue preparations. NCEN has also been used for two-photon imaging of intracellular hCE2 in living cells as well as in deep-tissues for the first time, and the results showed that the probe exhibited high ratiometric imaging resolution and deep-tissue imaging depth. All these findings suggested that this probe holds great promise for applications in bioimaging of endogenous hCE2 in living cells and in exploring the biological functions of hCE2 in complex biological systems.
Collapse
Affiliation(s)
- Qiang Jin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023, China
| | - Lei Feng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology , Dalian 116012, China
| | - Dan-Dan Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023, China
| | - Zi-Ru Dai
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023, China
| | - Ping Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023, China
| | - Li-Wei Zou
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023, China
| | - Zhi-Hong Liu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University , Wuhan 430072, China
| | - Jia-Yue Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023, China
| | - Yang Yu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023, China
| | - Guang-Bo Ge
- 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
| | - Jing-Nan Cui
- State Key Laboratory of Fine Chemicals, Dalian University of Technology , Dalian 116012, China
| | - Ling Yang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023, China
| |
Collapse
|
47
|
Ohura K, Nakada Y, Kotani S, Imai T. Design of Fexofenadine Prodrugs Based on Tissue-Specific Esterase Activity and Their Dissimilar Recognition by P-Glycoprotein. J Pharm Sci 2015; 104:3076-83. [DOI: 10.1002/jps.24467] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 04/07/2015] [Accepted: 04/07/2015] [Indexed: 11/11/2022]
|
48
|
Rasmussen HB, Bjerre D, Linnet K, Jürgens G, Dalhoff K, Stefansson H, Hankemeier T, Kaddurah-Daouk R, Taboureau O, Brunak S, Houmann T, Jeppesen P, Pagsberg AK, Plessen K, Dyrborg J, Hansen PR, Hansen PE, Hughes T, Werge T. Individualization of treatments with drugs metabolized by CES1: combining genetics and metabolomics. Pharmacogenomics 2015; 16:649-65. [PMID: 25896426 DOI: 10.2217/pgs.15.7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
CES1 is involved in the hydrolysis of ester group-containing xenobiotic and endobiotic compounds including several essential and commonly used drugs. The individual variation in the efficacy and tolerability of many drugs metabolized by CES1 is considerable. Hence, there is a large interest in individualizing the treatment with these drugs. The present review addresses the issue of individualized treatment with drugs metabolized by CES1. It describes the composition of the gene encoding CES1, reports variants of this gene with focus upon those with a potential effect on drug metabolism and provides an overview of the protein structure of this enzyme bringing notice to mechanisms involved in the regulation of enzyme activity. Subsequently, the review highlights drugs metabolized by CES1 and argues that individual differences in the pharmacokinetics of these drugs play an important role in determining drug response and tolerability suggesting prospects for individualized drug therapies. Our review also discusses endogenous substrates of CES1 and assesses the potential of using metabolomic profiling of blood to identify proxies for the hepatic activity of CES1 that predict the rate of drug metabolism. Finally, the combination of genetics and metabolomics to obtain an accurate prediction of the individual response to CES1-dependent drugs is discussed.
Collapse
Affiliation(s)
- Henrik Berg Rasmussen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, 2 Boserupvej, DK-4000 Roskilde, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Oda S, Fukami T, Yokoi T, Nakajima M. A comprehensive review of UDP-glucuronosyltransferase and esterases for drug development. Drug Metab Pharmacokinet 2015; 30:30-51. [DOI: 10.1016/j.dmpk.2014.12.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/24/2014] [Accepted: 12/02/2014] [Indexed: 01/24/2023]
|
50
|
Nishimuta H, Houston JB, Galetin A. Hepatic, intestinal, renal, and plasma hydrolysis of prodrugs in human, cynomolgus monkey, dog, and rat: implications for in vitro-in vivo extrapolation of clearance of prodrugs. Drug Metab Dispos 2014; 42:1522-31. [PMID: 24994071 DOI: 10.1124/dmd.114.057372] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hydrolysis plays an important role in metabolic activation of prodrugs. In the current study, species and in vitro system differences in hepatic and extrahepatic hydrolysis were investigated for 11 prodrugs. Ten prodrugs in the data set are predominantly hydrolyzed by carboxylesterases (CES), whereas olmesartan medoxomil is also metabolized by carboxymethylenebutenolidase (CMBL) and paraoxonase. Metabolic stabilities were assessed in cryopreserved hepatocytes, liver S9 (LS9), intestinal S9 (IS9), kidney S9 (KS9), and plasma from human, monkey, dog, and rat. Of all the preclinical species investigated, monkey intrinsic hydrolysis clearance obtained in hepatocytes (CLint,hepatocytes) were the most comparable to human hepatocyte data. Perindopril and candesartan cilexetil showed the lowest and highest CLint,hepatocytes, respectively, regardless of the species investigated. Scaled intrinsic hydrolysis clearance obtained in LS9 were generally higher than CLint,hepatocytes in all species investigated, with the exception of dog. In the case of human and dog intestinal S9, hydrolysis intrinsic clearance could not be obtained for CES1 substrates, but hydrolysis for CES2 and CMBL substrates was detected in IS9 and KS9 from all species. Pronounced species differences were observed in plasma; hydrolysis of CES substrates was only evident in rat. Predictability of human hepatic intrinsic clearance (CLint,h) was assessed for eight CES1 substrates using hepatocytes and LS9; extrahepatic hydrolysis was not considered due to high stability of these prodrugs in intestinal and kidney S9. On average, predicted oral CLint,h from hepatocyte data represented 20% of the observed value; the underprediction was pronounced for high-clearance prodrugs, consistent with the predictability of cytochrome P450/conjugation clearance from this system. Prediction bias was less apparent with LS9, in particular for high-clearance prodrugs, highlighting the application of this in vitro system for investigation of prodrugs.
Collapse
Affiliation(s)
- Haruka Nishimuta
- Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, University of Manchester, Manchester, United Kingdom (H.N., J.B.H., A.G.); and Preclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (H.N.)
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, University of Manchester, Manchester, United Kingdom (H.N., J.B.H., A.G.); and Preclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (H.N.)
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, University of Manchester, Manchester, United Kingdom (H.N., J.B.H., A.G.); and Preclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (H.N.)
| |
Collapse
|