1
|
Wang YG, Gan CP, Beukers-Korver J, Rosing H, Li WL, Wagenaar E, Lebre MC, Song JY, Pritchard C, Bin Ali R, Huijbers I, Beijnen JH, Schinkel AH. Intestinal human carboxylesterase 2 (CES2) expression rescues drug metabolism and most metabolic syndrome phenotypes in global Ces2 cluster knockout mice. Acta Pharmacol Sin 2024:10.1038/s41401-024-01407-4. [PMID: 39496863 DOI: 10.1038/s41401-024-01407-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024] Open
Abstract
Carboxylesterase 2 (CES2) is expressed mainly in liver and intestine, but most abundantly in intestine. It hydrolyzes carboxylester, thioester, and amide bonds in many exogenous and endogenous compounds, including lipids. CES2 therefore not only plays an important role in the metabolism of many (pro-)drugs, toxins and pesticides, directly influencing pharmacology and toxicology in humans, but it is also involved in energy homeostasis, affecting lipid and glucose metabolism. In this study we investigated the pharmacological and physiological functions of CES2. We constructed Ces2 cluster knockout mice lacking all eight Ces2 genes (Ces2-/- strain) as well as humanized hepatic or intestinal CES2 transgenic strains in this Ces2-/- background. We showed that oral availability and tissue disposition of capecitabine were drastically increased in Ces2-/- mice, and tissue-specifically decreased by intestinal and hepatic human CES2 (hCES2) activity. The metabolism of the chemotherapeutic agent vinorelbine was strongly reduced in Ces2-/- mice, but only marginally rescued by hCES2 expression. On the other hand, Ces2-/- mice exhibited fatty liver, adipositis, hypercholesterolemia and diminished glucose tolerance and insulin sensitivity, but without body mass changes. Paradoxically, hepatic hCES2 expression rescued these metabolic phenotypes but increased liver size, adipose tissue mass and overall body weight, suggesting a "healthy" obesity phenotype. In contrast, intestinal hCES2 expression efficiently rescued all phenotypes, and even improved some parameters, including body weight, relative to the wild-type baseline values. Our results suggest that the induction of intestinal hCES2 may combat most, if not all, of the adverse effects of metabolic syndrome. These CES2 mouse models will provide powerful preclinical tools to enhance drug development, increase physiological insights, and explore potential solutions for metabolic syndrome-associated disorders.
Collapse
Affiliation(s)
- Yao-Geng Wang
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Chang-Pei Gan
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Joke Beukers-Korver
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Wen-Long Li
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Els Wagenaar
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Maria C Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, the Netherlands
| | - Colin Pritchard
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Rahmen Bin Ali
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Ivo Huijbers
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Cavallero A, Donadel G, Puccini P, Gervasi PG, Gabisonia K, Longo V, Gabriele M. New insight on porcine carboxylesterases expression and activity in lung tissues. Res Vet Sci 2024; 175:105314. [PMID: 38823354 DOI: 10.1016/j.rvsc.2024.105314] [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: 01/12/2024] [Revised: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Over the course of the last twenty years, there has been a growing recognition of the pig's potential as a valuable model for studying human drug metabolism. This study aimed to investigate the expression, enzymatic activity, inhibitory susceptibility, and cellular localization of carboxylesterases (CES) in porcine lung tissue not yet explored. Our results showed that CESs hydrolysis activity followed Michaelis-Menten kinetics in both cytosolic and microsomal fractions of porcine lung tissues (N = 8), with comparable hydrolysis rates for tested substrates, namely 4-nitrophenyl acetate (pNPA), 4-methylumbelliferyl acetate (4-MUA), and fluorescein diacetate (FD). We also determined the CESs hydrolysis activity in a representative sample of the porcine liver that, as expected, displayed higher activity than the lung ones. The study demonstrated variable levels of enzyme activities and interindividual variability in both porcine lung fractions. Inhibition studies used to assess the CESs' involvement in the hydrolysis of pNPA, 4-MUA, and FD suggested that CESs may be the enzymes primarily involved in the metabolism of ester compounds in the pig lung tissue. Overall, this study provides insight into the distribution and diversity of CES isoforms involved in substrate hydrolysis across different cellular fractions (cytosol and microsomes) in porcine lungs.
Collapse
Affiliation(s)
- Andrea Cavallero
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Giorgia Donadel
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Paola Puccini
- Chiesi Farmaceutici S.P.A., via Palermo 26/A, Parma, Italy
| | - Pier Giovanni Gervasi
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Khatia Gabisonia
- Interdisciplinary Center "Health Science", Scuola Superiore Sant'Anna, c/o Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Vincenzo Longo
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Morena Gabriele
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
3
|
Liu Y, Li J, Zhu HJ. Regulation of carboxylesterases and its impact on pharmacokinetics and pharmacodynamics: an up-to-date review. Expert Opin Drug Metab Toxicol 2024; 20:377-397. [PMID: 38706437 PMCID: PMC11151177 DOI: 10.1080/17425255.2024.2348491] [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: 02/28/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
INTRODUCTION Carboxylesterase 1 (CES1) and carboxylesterase 2 (CES2) are among the most abundant hydrolases in humans, catalyzing the metabolism of numerous clinically important medications, such as methylphenidate and clopidogrel. The large interindividual variability in the expression and activity of CES1 and CES2 affects the pharmacokinetics (PK) and pharmacodynamics (PD) of substrate drugs. AREAS COVERED This review provides an up-to-date overview of CES expression and activity regulations and examines their impact on the PK and PD of CES substrate drugs. The literature search was conducted on PubMed from inception to January 2024. EXPERT OPINION Current research revealed modest associations of CES genetic polymorphisms with drug exposure and response. Beyond genomic polymorphisms, transcriptional and posttranslational regulations can also significantly affect CES expression and activity and consequently alter PK and PD. Recent advances in plasma biomarkers of drug-metabolizing enzymes encourage the research of plasma protein and metabolite biomarkers for CES1 and CES2, which could lead to the establishment of precision pharmacotherapy regimens for drugs metabolized by CESs. Moreover, our understanding of tissue-specific expression and substrate selectivity of CES1 and CES2 has shed light on improving the design of CES1- and CES2-activated prodrugs.
Collapse
Affiliation(s)
- Yaping Liu
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan
| | - Jiapeng Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
4
|
Kailass K, Casalena D, Jenane L, McEdwards G, Auld DS, Sadovski O, Kaye EG, Hudson E, Nettleton D, Currie MA, Beharry AA. Tight-Binding Small-Molecule Carboxylesterase 2 Inhibitors Reduce Intracellular Irinotecan Activation. J Med Chem 2024; 67:2019-2030. [PMID: 38265364 DOI: 10.1021/acs.jmedchem.3c01850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
As the primary enzyme responsible for the activatable conversion of Irinotecan (CPT-11) to SN-38, carboxylesterase 2 (CES2) is a significant predictive biomarker toward CPT-11-based treatments for pancreatic ductal adenocarcinoma (PDAC). High SN-38 levels from high CES2 activity lead to harmful effects, including life-threatening diarrhea. While alternate strategies have been explored, CES2 inhibition presents an effective strategy to directly alter the pharmacokinetics of CPT-11 conversion, ultimately controlling the amount of SN-38 produced. To address this, we conducted a high-throughput screening to discover 18 small-molecule CES2 inhibitors. The inhibitors are validated by dose-response and counter-screening and 16 of these inhibitors demonstrate selectivity for CES2. These 16 inhibitors inhibit CES2 in cells, indicating cell permeability, and they show inhibition of CPT-11 conversion with the purified enzyme. The top five inhibitors prohibited cell death mediated by CPT-11 when preincubated in PDAC cells. Three of these inhibitors displayed a tight-binding mechanism of action with a strong binding affinity.
Collapse
Affiliation(s)
- Karishma Kailass
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - Dominick Casalena
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Lina Jenane
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - Gregor McEdwards
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada, L5L 1C6
| | - Douglas S Auld
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Oleg Sadovski
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - Esther G Kaye
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - Elyse Hudson
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - David Nettleton
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Mark A Currie
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada, L5L 1C6
| | - Andrew A Beharry
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| |
Collapse
|
5
|
Garcia ALH, de Souza MR, Picinini J, Soares S, Rohr P, Linden R, Schneider A, Freitas MPM, Ely HC, Bobermin LD, Dos Santos AQ, Dalberto D, da Silva J. Unraveling gene expression and genetic instability in dental fluorosis: Investigating the impact of chronic fluoride exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 906:167393. [PMID: 37769727 DOI: 10.1016/j.scitotenv.2023.167393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 08/31/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Chronic fluoride exposure, even in small quantities, when continuously ingested by the human population, can lead to a significant public health concern known as fluorosis. Our understanding of the effects of fluoride on human health, as well as its potential to impact DNA, is limited. The present study aimed to assess genetic instability in 20 individuals diagnosed with dental fluorosis and 20 individuals without the condition from the state of Rio Grande do Sul, Brazil. The participants' dental fluorosis was evaluated using the Thylstrup-Fejerskov index (TF). To further evaluate genetic instability, several assays were conducted, including the alkaline and modified (+FPG) comet assay (using a visual score, VS), the buccal micronucleus (MN) cytome (BMCyt) assay, the cytokinesis-block MN (CBMN-Cyt) assay, and the measurement of telomere length (TL). In addition, the study utilized tools from Systems Biology to gain insights into the effects of fluoride exposure on humans, which aided in the selection and evaluation of mRNA expression levels of specific genes, namely PPA1 (inorganic pyrophosphatase 1), AQP5 (Aquaporin 5), and MT-ATP6 (Mitochondrially Encoded Adenosine Triphosphate Synthase Membrane Subunit 6). Furthermore, fluoride levels in the blood and urine were assessed using an ion-selective electrode, along with the evaluation of the inflammatory response in serum. The group with dental fluorosis exhibited 2.18 times higher MN frequencies specifically when assessed using the CBMN-Cyt assay, in comparison with individuals without fluorosis. Findings from the enzyme-modified comet assay indicated oxidative damage to purines in DNA. Furthermore, a decrease in TL was observed, along with elevated expression patterns of the PPA1 and AQP5 genes, and significant alterations in cytokine release. Significant correlations were identified between the TF and age, as well as the levels of necrotic cells. Additionally, noteworthy correlations were established between fluoride levels and the levels of MN, VS, and MT-ATP6. Although dental fluorosis results from fluoride exposure, our research highlights the potential influence of this condition on genomic instability and gene expression. Consequently, our findings stress the importance of continuously monitoring populations with a high incidence of dental fluorosis to enhance our comprehension of how genomic instability might correlate with the origins and consequences of health problems in these individuals.
Collapse
Affiliation(s)
- Ana Leticia Hilario Garcia
- Laboratory of Genetic Toxicology, PPGBioSaúde (Postgraduate Program in Cellular and Molecular Biology Applied to Health), Lutheran University of Brazil (ULBRA), 92425-900 Canoas, Rio Grande do Sul, Brazil; Laboratory of Genetic Toxicology, La Salle University (UniLaSalle), Canoas, Rio Grande do Sul, Brazil.
| | - Melissa Rosa de Souza
- Laboratory of Genetic Toxicology, PPGBioSaúde (Postgraduate Program in Cellular and Molecular Biology Applied to Health), Lutheran University of Brazil (ULBRA), 92425-900 Canoas, Rio Grande do Sul, Brazil
| | - Juliana Picinini
- Laboratory of Genetic Toxicology, PPGBioSaúde (Postgraduate Program in Cellular and Molecular Biology Applied to Health), Lutheran University of Brazil (ULBRA), 92425-900 Canoas, Rio Grande do Sul, Brazil
| | - Solange Soares
- Laboratory of Genetic Toxicology, PPGBioSaúde (Postgraduate Program in Cellular and Molecular Biology Applied to Health), Lutheran University of Brazil (ULBRA), 92425-900 Canoas, Rio Grande do Sul, Brazil
| | - Paula Rohr
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Rafael Linden
- Laboratory of Analytical Toxicology, Institute of Health Sciences, Graduate Program on Toxicology and Analytical Toxicology, Feevale University, Novo Hamburgo, RS, Brazil
| | - Anelise Schneider
- Laboratory of Analytical Toxicology, Institute of Health Sciences, Graduate Program on Toxicology and Analytical Toxicology, Feevale University, Novo Hamburgo, RS, Brazil
| | - Maria Perpétua Mota Freitas
- Lutheran University of Brazil (ULBRA), Graduate Program in Dentistry, 92425-900 Canoas, Rio Grande do Sul, Brazil
| | | | - Larissa Daniele Bobermin
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Anexo, Bairro Santa Cecília, Porto Alegre, RS 90035-003, Brazil
| | - André Quincozes Dos Santos
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Anexo, Bairro Santa Cecília, Porto Alegre, RS 90035-003, Brazil
| | - Daiana Dalberto
- Laboratory of Genetic Toxicology, PPGBioSaúde (Postgraduate Program in Cellular and Molecular Biology Applied to Health), Lutheran University of Brazil (ULBRA), 92425-900 Canoas, Rio Grande do Sul, Brazil
| | - Juliana da Silva
- Laboratory of Genetic Toxicology, PPGBioSaúde (Postgraduate Program in Cellular and Molecular Biology Applied to Health), Lutheran University of Brazil (ULBRA), 92425-900 Canoas, Rio Grande do Sul, Brazil; Laboratory of Genetic Toxicology, La Salle University (UniLaSalle), Canoas, Rio Grande do Sul, Brazil.
| |
Collapse
|
6
|
Yamada N, Negoro R, Watanabe K, Fujita T. Generation of Caco-2 cells with predictable metabolism by CYP3A4, UGT1A1 and CES using the PITCh system. Drug Metab Pharmacokinet 2023; 50:100497. [PMID: 37037169 DOI: 10.1016/j.dmpk.2023.100497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Caco-2 cells are widely used as an in vitro intestinal model. However, the expression levels of the drug-metabolizing enzymes CYP3A4 and UGT1A1 are lower in these cells than in intestinal cells. Furthermore, the majority of prodrugs in use today are ester-containing, and carboxylesterase (CES) 1 and CES2 are among the enzymes that process the prodrugs into drugs. In the human small intestine, CES1 is hardly expressed while CES2 is highly expressed, but the CES expression pattern in Caco-2 cells is the opposite. In this study, we generated CYP3A4-POR-UGT1A1-CES2 knock-in (KI) and CES1 knock-out (KO) Caco-2 (genome-edited Caco-2) cells using a PITCh system. Genome-edited Caco-2 cells were shown to express functional CYP3A4, POR, UGT1A1 and CES2 while the expression of the CES1 protein was completely knocked out. We performed transport assays using temocapril. The Papp value of temocapril in genome-edited Caco-2 cells was higher than that in WT Caco-2 cells. Interestingly, the amount of temocaprilat on the apical side in genome-edited Caco-2 cells was lower than that in WT Caco-2 cells. These results suggest that genome-edited Caco-2 cells are more suitable than WT Caco-2 cells as a model for predicting intestinal drug absorption and metabolism.
Collapse
Affiliation(s)
- Naoki Yamada
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan
| | - Ryosuke Negoro
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan.
| | - Keita Watanabe
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan
| | - Takuya Fujita
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan; Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan; Research Center for Drug Discovery and Development, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan
| |
Collapse
|
7
|
Canlas J, Myers AL. Interactions of Betel Quid Constituents with Drug Disposition Pathways: An Overview. Curr Drug Metab 2023; 24:92-105. [PMID: 36852799 PMCID: PMC11271041 DOI: 10.2174/1389200224666230228142052] [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: 07/18/2022] [Revised: 01/10/2023] [Accepted: 01/25/2023] [Indexed: 03/01/2023]
Abstract
Global estimates indicate that over 600 million individuals worldwide consume the areca (betel) nut in some form. Nonetheless, its consumption is associated with a myriad of oral and systemic ailments, such as precancerous oral lesions, oropharyngeal cancers, liver toxicity and hepatic carcinoma, cardiovascular distress, and addiction. Users commonly chew slivers of areca nut in a complex consumable preparation called betel quid (BQ). Consequently, the user is exposed to a wide array of chemicals with diverse pharmacokinetic behavior in the body. However, a comprehensive understanding of the metabolic pathways significant to BQ chemicals is lacking. Henceforth, we performed a literature search to identify prominent BQ constituents and examine each chemical's interplay with drug disposition proteins. In total, we uncovered over 20 major chemicals (e.g., arecoline, nicotine, menthol, quercetin, tannic acid) present in the BQ mixture that were substrates, inhibitors, and/or inducers of various phase I (e.g., CYP, FMO, hydrolases) and phase II (e.g., GST, UGT, SULT) drug metabolizing enzymes, along with several transporters (e.g., P-gp, BCRP, MRP). Altogether, over 80 potential interactivities were found. Utilizing this new information, we generated theoretical predictions of drug interactions precipitated by BQ consumption. Data suggests that BQ consumers are at risk for drug interactions (and possible adverse effects) when co-ingesting other substances (multiple therapeutic classes) with overlapping elimination mechanisms. Until now, prediction about interactions is not widely known among BQ consumers and their clinicians. Further research is necessary based on our speculations to elucidate the biological ramifications of specific BQ-induced interactions and to take measures that improve the health of BQ consumers.
Collapse
Affiliation(s)
- Jasmine Canlas
- Department of Pharmaceutical & Biomedical Sciences, The University of Georgia, Athens, GA 30602, United States
| | - Alan L. Myers
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77030, United States
| |
Collapse
|
8
|
Characterisation of AMB-FUBINACA metabolism and CB 1-mediated activity of its acid metabolite. Forensic Toxicol 2023; 41:114-125. [PMID: 36652070 PMCID: PMC9849163 DOI: 10.1007/s11419-022-00649-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/17/2022] [Indexed: 01/22/2023]
Abstract
PURPOSE AMB-FUBINACA is a synthetic cannabinoid receptor agonist (SCRA) which is primarily metabolised by hepatic enzymes producing AMB-FUBINACA carboxylic acid. The metabolising enzymes associated with this biotransformation remain unknown. This study aimed to determine if AMB-FUBINACA metabolism could be reduced in the presence of carboxylesterase (CES) inhibitors and recreational drugs commonly consumed with it. The affinity and activity of the AMB-FUBINACA acid metabolite at the cannabinoid type-1 receptor (CB1) was investigated to determine the activity of the metabolite. METHODS The effect of CES1 and CES2 inhibitors, and delta-9-tetrahydrocannabinol (Δ9-THC) on AMB-FUBINACA metabolism were determined using both human liver microsomes (HLM) and recombinant carboxylesterases. Radioligand binding and cAMP assays comparing AMB-FUBINACA and AMB-FUBINACA acid were carried out in HEK293 cells expressing human CB1. RESULTS AMB-FUBINACA was rapidly metabolised by HLM in the presence and absence of NADPH. Additionally, CES1 and CES2 inhibitors both significantly reduced AMB-FUBINACA metabolism. Furthermore, digitonin (100 µM) significantly inhibited CES1-mediated metabolism of AMB-FUBINACA by ~ 56%, while the effects elicited by Δ9-THC were not statistically significant. AMB-FUBINACA acid produced only 26% radioligand displacement consistent with low affinity binding. In cAMP assays, the potency of AMB-FUBINACA was ~ 3000-fold greater at CB1 as compared to the acid metabolite. CONCLUSIONS CES1A1 was identified as the main hepatic enzyme responsible for the metabolism of AMB-FUBINACA to its less potent carboxylic acid metabolite. This biotransformation was significantly inhibited by digitonin. Since other xenobiotics may also inhibit similar SCRA metabolic pathways, understanding these interactions may elucidate why some users experience high levels of harm following SCRA use.
Collapse
|
9
|
Zhu T, Wu Y, Li XM, Jia YM, Zhou H, Jiang LP, Tai T, Mi QY, Ji JZ, Xie HG. Vicagrel is hydrolyzed by Raf kinase inhibitor protein in human intestine. Biopharm Drug Dispos 2022; 43:247-254. [PMID: 36519186 DOI: 10.1002/bdd.2340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/12/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022]
Abstract
As an analog of clopidogrel and prasugrel, vicagrel is completely hydrolyzed to intermediate thiolactone metabolite 2-oxo-clopidogrel (also the precursor of active thiol metabolite H4) in human intestine, predominantly by AADAC and CES2; however, other unknown vicagrel hydrolases remain to be identified. In this study, recombinant human Raf kinase inhibitor protein (rhRKIP) and pooled human intestinal S9 (HIS9) fractions and microsome (HIM) preparations were used as the different enzyme sources; prasugrel as a probe drug for RKIP (a positive control), vicagrel as a substrate drug of interest, and the rate of the formation of thiolactone metabolites 2-oxo-clopidogrel and R95913 as metrics of hydrolase activity examined, respectively. In addition, an IC50 value of inhibition of rhRKIP-catalyzed vicagrel hydrolysis by locostatin was measured, and five classical esterase inhibitors with distinct esterase selectivity were used to dissect the involvement of multiple hydrolases in vicagrel hydrolysis. The results showed that rhRKIP hydrolyzed vicagrel in vitro, with the values of Km , Vmax , and CLint measured as 20.04 ± 1.99 μM, 434.60 ± 12.46 nM/min/mg protein, and 21.69 ± 0.28 ml/min/mg protein, respectively, and that an IC50 value of locostatin was estimated as 1.24 ± 0.04 mM for rhRKIP. In addition to locostatin, eserine and vinblastine strongly suppressed vicagrel hydrolysis in HIM. It is concluded that RKIP can catalyze the hydrolysis of vicagrel in the human intestine, and that vicagrel can be hydrolyzed by multiple hydrolases, such as RKIP, AADAC, and CES2, concomitantly.
Collapse
Affiliation(s)
- Ting Zhu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yu Wu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xue-Mei Li
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Meng Jia
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Huan Zhou
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li-Ping Jiang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ting Tai
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qiong-Yu Mi
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin-Zi Ji
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hong-Guang Xie
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Clinical Pharmacy, Nanjing Medical University School of Pharmacy, Nanjing, 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
|
Solé M, Figueres E, Mañanós E, Rojo-Solís C, García-Párraga D. Characterisation of plasmatic B-esterases in bottlenose dolphins (Tursiops truncatus) and their potential as biomarkers of xenobiotic chemical exposures. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 313:120149. [PMID: 36115493 DOI: 10.1016/j.envpol.2022.120149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/23/2022] [Accepted: 09/06/2022] [Indexed: 06/15/2023]
Abstract
A total of 164 blood samples from 16 clinically healthy bottlenose dolphins (Tursiops truncatus), were obtained from an aquarium in Spain between 2019 and 2020, as part of their preventive medicine protocol. In addition to conventional haematological and biochemical analyses, plasmatic B-esterase activities were characterised to determine the potential application of such analyses in wild counterparts. The hydrolysis rates for the substrates of acetylcholinesterase (AChE), butyrylcholinesterase (BuChE) and carboxylesterase (CE) activity in plasma were measured, the last using two commercial substrates, p-nitrophenyl acetate (pNPA) and p-nitrophenyl butyrate (pNPB). Activity rates (mean ± SEM in nmol/min/mL plasma) were (in descending order): AChE (125.6 ± 3.8), pNPB-CE (65.0 ± 2.2), pNPA-CE (49.7 ± 1.1) and BuChE (12.8 ± 1.3). These values for dolphins are reported in here for the first time in this species. Additionally, the in vitro sensitivity of two B-esterases (AChE and pNPB-CE) to chemicals of environmental concern was determined, and the protective role of plasmatic albumin assessed. Out of the B-esterases measured in plasma of dolphin, AChE activity was more responsive in vitro to pesticides, while CEs had a low response to plastic additives, likely due to the protective presence of albumin. However, the clear in vitro interaction of these environmental chemicals with purified AChE from electric eels and recombinant human hCEs (hCE1 and hCE2) and albumin, predicts their impact in other tissues that require in vivo validation. A relationship between esterase-like activities and health parameters in terrestrial mammals has already been established. Thus, B-esterase measures could be easily included in marine mammal health assessment protocols for dolphins as well, once the relationship between these measures and the animal's fitness has been established.
Collapse
Affiliation(s)
- M Solé
- Institut de Ciències del Mar, CSIC, Psg. Marítim de La Barceloneta 37-49, 08003, Barcelona, Spain.
| | - E Figueres
- Institut de Ciències del Mar, CSIC, Psg. Marítim de La Barceloneta 37-49, 08003, Barcelona, Spain
| | - E Mañanós
- Institute of Aquaculture Torre La Sal (IATS),-CSIC, 12595, Cabanes, Castellón, Spain
| | - C Rojo-Solís
- Veterinary Services, Oceanogràfic, Ciudad de Las Artes y Las Ciencias, C/Eduardo Primo Yúfera (Científic) 1B, 46013, València, Spain
| | - D García-Párraga
- Veterinary Services, Oceanogràfic, Ciudad de Las Artes y Las Ciencias, C/Eduardo Primo Yúfera (Científic) 1B, 46013, València, Spain
| |
Collapse
|
12
|
Sole M, Bassols A, Labrada-Martagón V. Plasmatic B-esterases as potential biomarkers of exposure to marine plastics in loggerhead turtles. ENVIRONMENTAL RESEARCH 2022; 213:113639. [PMID: 35688215 DOI: 10.1016/j.envres.2022.113639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/30/2022] [Accepted: 06/05/2022] [Indexed: 06/15/2023]
Abstract
Sea turtles are particularly vulnerable to plastic exposures, and the associated chemical additives, due to their feeding strategies. The species Caretta caretta is a proposed sentinel of plastic pollution worldwide. Thus, there is a need to find adequate biomarkers of plastic exposure through non-invasive protocols for this IUCN protected species. Plasmatic acetylcholinesterase (AChE), butyrylcholinesterase (BuChE) and carboxylesterase (CE) which participate in xenobiotic and endogenous metabolic reactions could all serve as biomarkers, as they are responsive to plasticizers and have already proved adequate for identifying organophosphorus esters exposures. Here we measured plasmatic B-esterases in wild specimens captured as accidental by-catch. Measurements were taken in each individual either at entry into the rehabilitation program or immediately before release after a recovery period. For CE measurements, 4 commercial substrates were used as potentially indicative of distinct enzyme isoforms. Increased activity was seen with the butyrate-derived substrates. Plasmatic CE activities were over one order of magnitude higher than AChE and BuChE substrates. Moreover, an in vitro protocol with the inclusion of plastic additives such as tetrabromobisphenol A (TBBPA), bisphenol A and some of its analogues was considered a proxy of enzymatic interactions. A clear inhibition by TBBPA was found when using commercially purified AChE and recombinant CE proteins. Overall, from in vitro and in vivo evidences, CEs in plasma are sensitive and easily measurable and have been shown to significantly increase after turtles have been rehabilitated in rescue centres. Nevertheless, the inclusion of plastic (or plasticizers) characterisation would help to confirm its association with plasmatic enzyme modifications before they can be adopted as biomarkers of plastic contamination.
Collapse
Affiliation(s)
- M Sole
- Institut de Ciències del Mar, CSIC, Psg. Marítim de la Barceloneta 37-49, 08003, Barcelona, Spain.
| | - A Bassols
- Fundació per a la Conservació i Recuperació d'Animals Marins-CRAM, Psg. de la Platja 28-30, 08820, El Prat de Llobregat, Spain
| | - V Labrada-Martagón
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, UASLP, Av. Chapultepec #1570, Col. Privadas del Pedregal, CP 78295, San Luis Potosí, S.L.P., Mexico
| |
Collapse
|
13
|
Involvement of esterases in the pulmonary metabolism of beclomethasone dipropionate and the potential influence of cannabis use. Chem Biol Interact 2022; 368:110228. [DOI: 10.1016/j.cbi.2022.110228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/07/2022] [Accepted: 10/19/2022] [Indexed: 11/23/2022]
|
14
|
Ozsan C, Kailass K, Digby EM, Almammadov T, Beharry AA, Kolemen S. Selective detection of carboxylesterase 2 activity in cancer cells using an activity-based chemiluminescent probe. Chem Commun (Camb) 2022; 58:10929-10932. [PMID: 36065979 DOI: 10.1039/d2cc03309g] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Carboxylesterase 2 (CES2) has crucial roles in both xenobiotic metabolism and formation of pathogenic states including cancer. Thus, it is highly critical to monitor intracellular CES2 activity in living cancer cells. Here, we report a CES2 activatable phenoxy 1,2-dioxetane based chemiluminescent agent (CL-CES2). The probe exhibited a selective turn-on response in the presence of CES2 enzyme and enabled detection of CES2 activity in three different cancer cells that possess varying enzyme concentrations with high signal to noise ratios. In contrast no signal was obtained with CES1, an isoform of CES2 enzyme. CL-CES2 marks the first ever example of a CES2-responsive chemiluminescent luminophore and holds a great potential in further understanding the roles of CES2 activity in tumorogenesis.
Collapse
Affiliation(s)
- Cagri Ozsan
- Department of Chemistry, Koç University, 34450 Istanbul, Turkey.
| | - Karishma Kailass
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada.
| | - Elyse M Digby
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada.
| | | | - Andrew A Beharry
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada.
| | - Safacan Kolemen
- Department of Chemistry, Koç University, 34450 Istanbul, Turkey. .,Surface Science and Technology Center (KUYTAM), Koç University, 34450 Istanbul, Turkey.,Boron and Advanced Materials Application and Research Center, Koç University, 34450 Istanbul, Turkey
| |
Collapse
|
15
|
Omedes S, Andrade M, Escolar O, Villanueva R, Freitas R, Solé M. B-esterases characterisation in the digestive tract of the common octopus and the European cuttlefish and their in vitro responses to contaminants of environmental concern. ENVIRONMENTAL RESEARCH 2022; 210:112961. [PMID: 35181305 DOI: 10.1016/j.envres.2022.112961] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 06/14/2023]
Abstract
Cephalopods are a group of marine invertebrates that have received little attention as sentinel species in comparison to other molluscs, such as bivalves. Consequently, their physiological and biochemical xenobiotic metabolism responses are poorly understood. Here we undertake a comparative analysis of the enzymatic activities involved in detoxification reactions and neural transmission in the digestive tract of two commercial cephalopods: the Common octopus, Octopus vulgaris, and the European cuttlefish, Sepia officinalis. For methodological purposes, several common B-esterases (five carboxylesterase (CE) substrates and three cholinesterase (ChE) determinations) were assayed as a proxy of metabolic and neuronal activities, respectively. Four components of the digestive tract in each species were considered: salivary glands, the stomach, the digestive gland and the caecum. The in vitro responses of digestive gland homogenates to model chemicals and contaminants of environmental concern were contrasted between both cephalopod species. The baseline biochemical activities in the four digestive tract components were also determined. Moreover, in order to validate the protocol, purified proteins, recombinant human CE (CE1 and CE2) and purified eel acetylcholinesterase (AChE) were included in the analysis. Overall, carboxylesterase activities were higher in octopus than in cuttlefish, with the activity quantified in the digestive tract components in the following order: digestive gland ≈ caecum > stomach ≈ salivary glands, with higher hydrolysis rates reached with naphthyl-derived substrates. In contrast, cuttlefish hydrolysis rates with ChE substrates were higher than in octopus. This trend was also reflected in a higher sensitivity to CE inhibitors in octopus and to AChE inhibitors in cuttlefish. Given the detoxification character of CEs and its protective role preventing AChE inhibition, octopus could be regarded as more efficiently protected than cuttlefish from neurotoxic exposures. A full characterisation of B-esterases in the digestive tract of the two common cephalopods is also provided.
Collapse
Affiliation(s)
- S Omedes
- Institut de Ciències del Mar ICM-CSIC, E-08003, Barcelona, Spain
| | - M Andrade
- Departamento de Biologia & CESAM, Universidade de Aveiro, 3810-193, Aveiro, Portugal
| | - O Escolar
- Institut de Ciències del Mar ICM-CSIC, E-08003, Barcelona, Spain
| | - R Villanueva
- Institut de Ciències del Mar ICM-CSIC, E-08003, Barcelona, Spain
| | - R Freitas
- Departamento de Biologia & CESAM, Universidade de Aveiro, 3810-193, Aveiro, Portugal
| | - M Solé
- Institut de Ciències del Mar ICM-CSIC, E-08003, Barcelona, Spain.
| |
Collapse
|
16
|
Lai Y, Chu X, Di L, Gao W, Guo Y, Liu X, Lu C, Mao J, Shen H, Tang H, Xia CQ, Zhang L, Ding X. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development. Acta Pharm Sin B 2022; 12:2751-2777. [PMID: 35755285 PMCID: PMC9214059 DOI: 10.1016/j.apsb.2022.03.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Drug metabolism and pharmacokinetics (DMPK) is an important branch of pharmaceutical sciences. The nature of ADME (absorption, distribution, metabolism, excretion) and PK (pharmacokinetics) inquiries during drug discovery and development has evolved in recent years from being largely descriptive to seeking a more quantitative and mechanistic understanding of the fate of drug candidates in biological systems. Tremendous progress has been made in the past decade, not only in the characterization of physiochemical properties of drugs that influence their ADME, target organ exposure, and toxicity, but also in the identification of design principles that can minimize drug-drug interaction (DDI) potentials and reduce the attritions. The importance of membrane transporters in drug disposition, efficacy, and safety, as well as the interplay with metabolic processes, has been increasingly recognized. Dramatic increases in investments on new modalities beyond traditional small and large molecule drugs, such as peptides, oligonucleotides, and antibody-drug conjugates, necessitated further innovations in bioanalytical and experimental tools for the characterization of their ADME properties. In this review, we highlight some of the most notable advances in the last decade, and provide future perspectives on potential major breakthroughs and innovations in the translation of DMPK science in various stages of drug discovery and development.
Collapse
Affiliation(s)
- Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA 94404, USA
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Wei Gao
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Yingying Guo
- Eli Lilly and Company, Indianapolis, IN 46221, USA
| | - Xingrong Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, MA 02142, USA
| | - Chuang Lu
- Drug Metabolism and Pharmacokinetics, Accent Therapeutics, Inc. Lexington, MA 02421, USA
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, A Member of the Roche Group, South San Francisco, CA 94080, USA
| | - Hong Shen
- Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, NJ 08540, USA
| | - Huaping Tang
- Bioanalysis and Biomarkers, Glaxo Smith Kline, King of the Prussia, PA 19406, USA
| | - Cindy Q. Xia
- Department of Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, MA 02139, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, CDER, FDA, Silver Spring, MD 20993, USA
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
17
|
Makino C, Watanabe A, Kato M, Shiozawa H, Takakusa H, Nakai D, Honda T, Watanabe N. Species differences between rats and primates (humans and monkeys) in complex cleavage pathways of DS-8500a characterized by 14C-ADME studies in humans and monkeys after administration of two radiolabeled compounds and in vitro studies. Drug Metab Pharmacokinet 2022; 45:100459. [DOI: 10.1016/j.dmpk.2022.100459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/21/2022] [Accepted: 03/15/2022] [Indexed: 11/03/2022]
|
18
|
Marzolini C, Gibbons S, van Oosterhout JJ, Khoo S. Drug-Drug Interaction Potential with Once-Weekly Isoniazid/Rifapentine (3HP) for the Treatment of Latent Tuberculosis Infection. Clin Pharmacokinet 2022; 61:339-346. [PMID: 34905153 DOI: 10.1007/s40262-021-01098-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Catia Marzolini
- Division of Infectious Diseases and Hospital Epidemiology, Departments of Medicine and Clinical Research, University Hospital of Basel, University of Basel, Petersgraben 4, 4031, Basel, Switzerland.
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Sara Gibbons
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Joep J van Oosterhout
- Partners in Hope, Lilongwe, Malawi
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Saye Khoo
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
19
|
Zhong Y, Yang L, Zhou Y, Peng J. Biomarker-responsive Fluorescent Probes for In Vivo Imaging of Liver Injury. Chem Asian J 2022; 17:e202200038. [PMID: 35182452 DOI: 10.1002/asia.202200038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/16/2022] [Indexed: 11/08/2022]
Abstract
Liver injury-related diseases have aroused widespread concern due to its extreme unpredictability, acute onset, and severe consequences. Nowadays, the clinical prediction and assessment of liver injury mainly focus on histopathological and serological approaches, which undergoes a tedious process and sometimes requires invasive biopsy. Over the past decades, fluorescence imaging technique have emerged as a rising star for the diagnosis of diseases owing to its noninvasiveness, high fidelity and ease of operation. On regard to liver injury, the fluorescent probes have been delicately designed to response a variety of endogenous biomolecules to precisely offer comprehensive information about the lesion site. Herein, we make a brief summary and discussion about the design strategies and applications of the recently reported fluorescent biosensors responsive to a series of biomarkers involved in the liver injury. The potential prospects and remaining challenges are also discussed to promote the progression in this field.
Collapse
Affiliation(s)
- Yang Zhong
- China Pharmaceutical University, State Key Laboratory of Natural Medicines, CHINA
| | - Lulu Yang
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, CHINA
| | - Yunyun Zhou
- China Pharmaceutical University, State Key Laboratory of Natural Medicines, CHINA
| | - Juanjuan Peng
- China Pharmaceutical University, #24 Tong Jia Xiang, Nanjing, CHINA
| |
Collapse
|
20
|
Johnson JL, Huang J, Rooney M, Gu C. Optimal pH 8.5 to 9 for the Hydrolysis of Vixotrigine and Other Basic Substrates of Carboxylesterase-1 in Human Liver Microsomes. Xenobiotica 2021; 52:105-112. [PMID: 34904522 DOI: 10.1080/00498254.2021.2018629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Vixotrigine is a voltage- and use-dependent sodium channel blocker under investigation for the potential treatment of neuropathic pain. One of the major in vivo metabolic pathways of vixotrigine in humans is the hydrolysis of the carboxamide to form the carboxylic acid metabolite M14.The in vitro formation of M14 in human hepatocytes was inhibited by the carboxylesterase (CES) inhibitor Bis(4-nitrophenyl) phosphate in a concentration-dependent manner. The hydrolysis reaction was identified to be catalyzed by recombinant human CES1b.Initial observation of only trace level formation of M14 in human liver microsomes at pH 7.4 caused us to doubt the involvement of CES1, an enzyme localized at the endoplasmic reticulum and the dominant carboxylesterase in human liver. Further investigation has revealed that optimal pH for the hydrolysis of vixotrigine and two other basic substrates of CES1, methylphenidate and oseltamivir, in human liver microsomes was pH 8.5 to 9 which is higher than their respective pKa(base), suggesting that neutral form of basic substrates is probably preferred for CES1 catalysis in liver microsomes.
Collapse
Affiliation(s)
- Joshua L Johnson
- Drug Metabolism and Pharmacokinetics.,Current affiliation of JLJ: Drug Metabolism and Pharmacokinetics, Takeda, San Diego, CA, USA
| | | | - Michael Rooney
- Clinical Pharmacology and Pharmacometrics, Biogen, Cambridge, MA, USA
| | | |
Collapse
|
21
|
Sakai Y, Fukami T, Nagaoka M, Hirosawa K, Ichida H, Sato R, Suzuki K, Nakano M, Nakajima M. Arylacetamide deacetylase as a determinant of the hydrolysis and activation of abiraterone acetate in mice and humans. Life Sci 2021; 284:119896. [PMID: 34450168 DOI: 10.1016/j.lfs.2021.119896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/30/2021] [Accepted: 08/07/2021] [Indexed: 11/26/2022]
Abstract
AIM Abiraterone acetate for metastatic castration-resistant prostate cancer is an acetylated prodrug to be hydrolyzed to abiraterone. Abiraterone acetate is known to be hydrolyzed by pancreatic cholesterol esterase secreted into the intestinal lumen. This study aimed to investigate the possibility that arylacetamide deacetylase (AADAC) expressed in enterocytes contributes to the hydrolysis of abiraterone acetate based on its substrate preference. MATERIALS AND METHODS Abiraterone acetate hydrolase activity was measured using human intestinal (HIM) and liver microsomes (HLM) as well as recombinant AADAC. Correlation analysis between activity and AADAC expression was performed in 14 individual HIMs. The in vivo pharmacokinetics of abiraterone acetate was examined using wild-type and Aadac knockout mice administered abiraterone acetate with or without orlistat, a pancreatic cholesterol esterase inhibitor. KEY FINDINGS Recombinant AADAC showed abiraterone acetate hydrolase activity with similar Km value to HIM and HLM. The positive correlation between activity and AADAC levels in individual HIMs supported the responsibility of AADAC for abiraterone acetate hydrolysis. The area under the plasma concentration-time curve (AUC) of abiraterone after oral administration of abiraterone acetate in Aadac knockout mice was 38% lower than that in wild-type mice. The involvement of pancreatic cholesterol esterase in abiraterone formation was revealed by the decreased AUC of abiraterone by coadministration of orlistat. Orlistat potently inhibited AADAC, implying its potential as a perpetrator of drug-drug interactions. SIGNIFICANCE AADAC is responsible for the hydrolysis of abiraterone acetate in the intestine and liver, suggesting that concomitant use of abiraterone acetate and drugs potently inhibiting AADAC should be avoided.
Collapse
Affiliation(s)
- Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute, Kakuma-machi, Kanazawa 920-1192, Japan.
| | - Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Keiya Hirosawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Hiroyuki Ichida
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Rei Sato
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kohei Suzuki
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute, Kakuma-machi, Kanazawa 920-1192, Japan
| |
Collapse
|
22
|
Solé M, Montemurro N, Pérez S. Biomarker responses and metabolism in Lumbricus terrestris exposed to drugs of environmental concern, an in vivo and in vitro approach. CHEMOSPHERE 2021; 277:130283. [PMID: 33774234 DOI: 10.1016/j.chemosphere.2021.130283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 06/12/2023]
Abstract
The earthworm Lumbricus terrestris is an anecic species living in natural soils but it is also a sentinel in pollution monitoring. Specimens of L.terrestris were exposed for 48 h though the filter paper contact test at 1 mg/mL of the chemicals: Lamotrigine (LMG), Cocaine (COC), Fipronil (FIP) and the pesticide bis-4-nitrophenyl phosphate (BNPP). After that period, the activities of Acetylcholinesterase, Glutathione S-transferase, Carboxylesterase (CE) using different substrates, and lipid peroxidation levels were evaluated in the exposed whole tissue earthworms. The results revealed differences only in CE activity, with 4-nitrophenyl butyrate (4NPB) and 1-naphthyl butyrate (1NB) the most responsive substrates to COC. The kinetic parameters of CE were characterized, for the first time, in whole tissue of this species. The chemical analysis by LC-MS/MS, confirmed the exposure to the parent compounds, identified metabolites and evidenced biotransformation pathways in earthworms. Metabolic reactions included oxidation (LMG and FIP), hydrolysis (COC and FIP) as well as glycosylation (LMG, COC and FIP). A hitherto unknown metabolite of LMG due to the conjugation with phenylalanine glutamine was formed. The in vivo results on CE activity with the specific inhibitor, BNPP, were confirmed in vitro. Moreover, in the in vitro approach, the inclusion of other contaminants of environmental concern supports the potential of CE as biomarker. This study identifies the main metabolites formed by earthworms for further in vivo exposures under more realistic conditions and the potential use of CE measures as biomarker of emerging contaminants.
Collapse
Affiliation(s)
- M Solé
- Renewable Marine Resources Department, Institute of Marine Sciences (ICM-CSIC), Barcelona, Spain.
| | - N Montemurro
- ENFOCHEM, Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034, Barcelona, Spain
| | - S Pérez
- ENFOCHEM, Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034, Barcelona, Spain
| |
Collapse
|
23
|
Song YQ, Jin Q, Wang DD, Hou J, Zou LW, Ge GB. Carboxylesterase inhibitors from clinically available medicines and their impact on drug metabolism. Chem Biol Interact 2021; 345:109566. [PMID: 34174250 DOI: 10.1016/j.cbi.2021.109566] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/21/2021] [Accepted: 06/16/2021] [Indexed: 12/11/2022]
Abstract
Mammalian carboxylesterases (CES), the key members of the serine hydrolase superfamily, hydrolyze a wide range of endogenous substances and xenobiotics bearing ester or amide bond(s). In humans, most of identified CES are segregated into the CES1A and CES2A subfamilies. Strong inhibition on human CES (including hCES1A and hCES2A) may modulate pharmacokinetic profiles of CES-substrate drugs, thereby changing the pharmacological and toxicological responses of these drugs. This review covered recent advances in discovery of hCES inhibitors from clinically available medications, as well as their impact on CES-associated drug metabolism. Three comprehensive lists of hCES inhibitors deriving from clinically available medications including therapeutic drugs, pharmaceutical excipients and herbal medicines, alongside with their inhibition potentials and inhibition parameters, are summarized. Furthermore, the potential risks of hCES inhibitors to trigger drug/herb-drug interactions (DDIs/HDIs) and future concerns in this field are highlighted. Potent hCES inhibitors may trigger clinically relevant DDIs/HDIs, especially when these inhibitors are co-administrated with CES substrate-drugs with very narrow therapeutic windows. All data and knowledge presented here provide key information for the clinicians to assess the risks of clinically available hCES inhibitors on drug metabolism. In future, more practical and highly specific substrates for hCES1A/hCES2A should be developed and used for studies on CES-mediated DDIs/HDIs both in vitro and in vivo.
Collapse
Affiliation(s)
- Yun-Qing Song
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qiang Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan-Dan Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jie Hou
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Li-Wei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
24
|
Trivedi A, Wahlstrom J, Mackowski M, Dutta S, Lee E. Pharmacokinetics, Disposition, and Biotransformation of [ 14C]Omecamtiv Mecarbil in Healthy Male Subjects after a Single Intravenous or Oral Dose. Drug Metab Dispos 2021; 49:619-628. [PMID: 34011533 DOI: 10.1124/dmd.121.000444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022] Open
Abstract
Omecamtiv mecarbil (OM) is a novel cardiac myosin activator that is currently in clinical development for the treatment of heart failure. The absorption and disposition of [14C]OM (60 µCi) were studied after a single intravenous infusion (35 mg over 1 hour) or oral solution dose (35 mg) in 14 healthy male subjects. Mean recovery of the administered [14C]OM dose was 85.1% and 86.5% over 336 hours for the intravenous and oral routes, respectively. After intravenous dosing, 47.8% and 37.3% of the dose was recovered in urine and feces, respectively; after oral dosing, 48.6% and 38.0% was recovered in urine and feces, respectively. Unchanged OM accounted for a minor percentage of radioactivity in urine (mean 7.7% of dose) and feces (mean 4.1% of dose) across all subjects. The major metabolites recovered in urine and feces were M3 (decarbamoylation product) and sequential metabolite M4 (lactam of M3), which accounted for means of 26.5% and 11.6% of the administered dose, respectively. The CYP4 family of enzymes was primarily responsible for the formation of M3 based on in vitro studies. Other metabolic pathways accounted for 14.9% of the administered dose. In pooled plasma, OM, M3, and M4 accounted for 83.8%, 6.0%, and 3.3% of the total [14C]OM-related materials. No other plasma metabolites constituted more than 3% of the administered dose. The bioavailability for OM solution was 93.5% after rapid and extensive absorption. SIGNIFICANCE STATEMENT: This study characterized the absorption and disposition of OM, a novel small molecule being developed for the treatment of heart failure. OM was primarily cleared through metabolism by the CYP4 family through oxidative cleavage of a terminal carbamate moiety that resembles hydrolysis.
Collapse
|
25
|
Hu SX, Ernst K, Benner CP, Feenstra KL. Stability of Ketoprofen Methylester in Plasma of Different Species. Curr Drug Metab 2021; 22:215-223. [PMID: 33334282 DOI: 10.2174/1389200221666201217141025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/30/2020] [Accepted: 11/04/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Pharmacokinetic and pharmacodynamic assessment of ester-containing drugs can be impacted by hydrolysis of the drugs in plasma samples post blood collection. The impact is different in the plasma of different species. OBJECTIVE This study evaluated the stability of a prodrug, ketoprofen methylester (KME), in commercially purchased and freshly collected plasma of mouse, rat, dog, cat, pig, sheep, cattle and horse. METHODS KME hydrolysis was determined following its incubation in commercially purchased and freshly collected plasma of those species. Different esterase inhibitors were evaluated for prevention of the hydrolysis in rat, dog and pig plasma. RESULTS KME was rapidly hydrolyzed in both commercially purchased and freshly collected plasma of mouse, rat, and horse. The hydrolysis was initially quick and then limited in cat plasma. KME hydrolysis was minimum in commercially purchased plasma of dog, pig, sheep and cattle but substantial in freshly collected plasma of those species. Different esterase inhibitors showed different effects on the stability of KME in rat, dog and pig plasma. CONCLUSION These results indicate that plasma of different species has different hydrolytic activities to estercontaining drugs. The activities in commercially purchased and freshly collected plasma may be different and species-dependent. Esterase inhibitors have different effects on preventing hydrolysis of the ester-containing drugs in the plasma of different species.
Collapse
Affiliation(s)
- Steven X Hu
- Zoetis, Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49007, United States
| | - Kelsey Ernst
- Zoetis, Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49007, United States
| | - Charles P Benner
- Zoetis, Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49007, United States
| | - Kenneth L Feenstra
- Zoetis, Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49007, United States
| |
Collapse
|
26
|
Metabolism and Interspecies Variation of IMMH-010, a Programmed Cell Death Ligand 1 Inhibitor Prodrug. Pharmaceutics 2021; 13:pharmaceutics13050598. [PMID: 33919384 PMCID: PMC8143347 DOI: 10.3390/pharmaceutics13050598] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/07/2021] [Accepted: 04/19/2021] [Indexed: 11/17/2022] Open
Abstract
IMMH-010 is an ester prodrug of YPD-29B, a potent programmed cell death ligand 1 (PD-L1) inhibitor. The metabolism of IMMH-010 was investigated and compared in various species. Four metabolites of IMMH-010 were identified, and the major metabolite was the parent compound, YPD-29B, which was mainly catalyzed by carboxylesterase 1 (CES1). We observed IMMH-010 metabolism in the plasma of various species. IMMH-010 was rapidly metabolized to YPD-29B in rat and mouse plasma, whereas it remained stable in human and monkey plasma. In the liver S9 fractions of human, monkey, dog, and rat, IMMH-010 was quickly transformed to YPD-29B with no obvious differences among species. In addition, the transformation ratio of IMMH-010 to YPD-29B was low in rat and human intestines, which indicated that the intestine was not an important site for IMMH-010 hydrolysis. Moreover, we demonstrated the remarkable antitumor efficacy of IMMH-010 in B16F10 melanoma and MC38 colon carcinoma xenograft mouse models. We also compared the pharmacokinetic profiles of IMMH-010 in rodents and primates. After oral administration of IMMH-010, the general exposure of active metabolite YPD-29B was slightly lower in primates than in rodents, suggesting that data should be extrapolated cautiously from rodents to humans.
Collapse
|
27
|
Yasuda K, Watanabe K, Fukami T, Nakashima S, Ikushiro SI, Nakajima M, Sakaki T. Epicatechin gallate and epigallocatechin gallate are potent inhibitors of human arylacetamide deacetylase. Drug Metab Pharmacokinet 2021; 39:100397. [PMID: 34171773 DOI: 10.1016/j.dmpk.2021.100397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/21/2021] [Accepted: 04/06/2021] [Indexed: 11/29/2022]
Abstract
Recently, in addition to carboxylesterases (CESs), we found that arylacetamide deacetylase (AADAC) plays an important role in the metabolism of some clinical drugs. In this study, we screened for food-related natural compounds that could specifically inhibit human AADAC, CES1, or CES2. AADAC, CES1, and CES2 activities in human liver microsomes were measured using phenacetin, fenofibrate, and procaine as specific substrates, respectively. In total, 43 natural compounds were screened for their inhibitory effects on each of these enzymes. Curcumin and quercetin showed strong inhibitory effects against all three enzymes, whereas epicatechin, epicatechin gallate (ECg), and epigallocatechin gallate (EGCg) specifically inhibited AADAC. In particular, ECg and EGCg showed strong inhibitory effects on AADAC (IC50 values: 3.0 ± 0.5 and 2.2 ± 0.2 μM, respectively). ECg and EGCg also strongly inhibited AADAC-mediated rifampicin hydrolase activity in human liver microsomes with IC50 values of 2.2 ± 1.4 and 1.7 ± 0.4 μM, respectively, whereas it weakly inhibited p-nitrophenyl acetate hydrolase activity, which is catalyzed by AADAC, CES1, and CES2. Our results indicate that ECg and EGCg are potent inhibitors of AADAC.
Collapse
Affiliation(s)
- Kaori Yasuda
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan.
| | - Kazuki Watanabe
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama, 939-0398, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Shimon Nakashima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University Kakuma-machi, Kanazawa 920-1192, Japan
| | - Shin-Ichi Ikushiro
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama, 939-0398, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Toshiyuki Sakaki
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| |
Collapse
|
28
|
Gabriele M, Puccini P, Gervasi PG, Longo V. Carboxylesterases and arylacetamide deacetylase comparison in human A549, H460, and H727 pulmonary cells. Life Sci 2021; 277:119486. [PMID: 33864822 DOI: 10.1016/j.lfs.2021.119486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 11/27/2022]
Abstract
AIMS Human carboxylesterases (CESs) and arylacetamide deacetylase (AADAC) are serine-esterase enzymes catalyzing the hydrolysis of many compounds containing esters, amides, thioesters, or acetyl groups. This study aimed to investigate the presence, kinetic parameters, and inhibition of CES1, CES2, and AADAC in A549, H460, and H727 pulmonary cells in both living cells and S9 fractions. MATERIALS AND METHODS The p-nitrophenyl acetate (pNPA) and 4-methylumbelliferyl acetate (4-MUA) were used as non-selective esterase substrates, whereas phenacetin as selective AADAC substrate. CESs activities were also investigated in living cells by cellular bioimaging using selective fluorescent probes. KEY FINDINGS AADAC gene was detected in A549 and H460 cells; nevertheless, arylesterase activity was not found in relative S9 fractions. Besides, CES1 and CES2 were expressed to a different extent by all lung cells, and enzymatic activities were quite overlapping each other. All enzymes exhibited a typical Michaelis-Menten saturation curve and, regarding 4-MUA, similar Km values were found in both living cells and S9 fractions. Conversely, kinetic parameters relative to the pNPA hydrolysis by S9 fractions were significantly lower than those detected in living cells. Inhibition studies revealed that 4-MUA hydrolysis was inhibited by bis-p-nitrophenyl phosphate and phenylmethanesulfonyl fluoride more than loperamide; on the contrary, pNPA hydrolysis inhibition was limited with similar inhibition profiles being obtained in both living cells and S9 fractions. The presence of carboxylesterases was definitely confirmed by cellular bioimaging. SIGNIFICANCE These findings add information to esterase knowledge in pulmonary cells that could be used as in vitro models for toxicological and pharmacological studies.
Collapse
Affiliation(s)
- Morena Gabriele
- National Research Council, Institute of Agricultural Biology and Biotechnology, via Moruzzi 1, 56124 Pisa, Italy.
| | - Paola Puccini
- Chiesi Farmaceutici S.p.A., via Palermo 26/A, Parma, Italy
| | - Pier Giovanni Gervasi
- National Research Council, Institute of Agricultural Biology and Biotechnology, via Moruzzi 1, 56124 Pisa, Italy
| | - Vincenzo Longo
- National Research Council, Institute of Agricultural Biology and Biotechnology, via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
29
|
Pham Nguyen TP, Brensinger CM, Bilker WB, Hennessy S, Leonard CE. Evaluation of serious bleeding signals during concomitant use of clopidogrel and hypnotic drugs. Biomed Pharmacother 2021; 139:111559. [PMID: 33845372 DOI: 10.1016/j.biopha.2021.111559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND In a previous drug-drug interaction (DDI) screening study intended to generate hypotheses, clopidogrel + either eszopiclone or zolpidem (vs. clopidogrel alone) were associated with serious bleeding. OBJECTIVES To confirm or refute these DDI signals and examine associations with other hypnotics in an independent population of United States Medicaid beneficiaries METHODS: We employed a bi-directional self-controlled case series design in eligible individuals concomitantly exposed to one of 12 hypnotics (precipitants, exposures of interest) plus either clopidogrel (the object drug) or pravastatin (the negative control object drug). The outcome was hospital presentation with serious bleeding. Using conditional Poisson regression, we calculated confounder-adjusted rate ratios (RRs) and 95% confidence intervals for serious bleeding during clopidogrel + precipitant use (vs. clopidogrel alone). To distinguish a DDI from a precipitant's inherent effect on bleeding, we divided effect measures by the adjusted RR for the corresponding pravastatin + precipitant pair to obtain ratios of RR (RRRs). RESULTS Among 23,194 users of clopidogrel and 3824 of pravastatin who experienced serious bleeding during an active prescription for one of these agents, confounder-adjusted RRRs for serious bleeding were 6.63 (0.39-113.01) and 0.77 (0.53-1.11) with eszopiclone and zolpidem, respectively, whereas confounder-adjusted RRRs for other hypnotics ranged from 0.18 (0.04-0.85) for triazolam to 1.79 (0.16-20.44) for zaleplon. Statistical imprecision therefore precluded us from confirming or refuting these prior signals with eszopiclone and zolpidem. CONCLUSIONS While we could not confirm or refute previously identified DDI signals, numerically elevated RRRs for serious bleeding with several clopidogrel + hypnotic pairs warrant further examination.
Collapse
Affiliation(s)
- Thanh Phuong Pham Nguyen
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Translational Center for Excellence for Neuroepidemiology and Neurological Outcomes Research, Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Clinical Epidemiology and Biostatistics, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Colleen M Brensinger
- Center for Clinical Epidemiology and Biostatistics, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Warren B Bilker
- Center for Clinical Epidemiology and Biostatistics, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sean Hennessy
- Center for Clinical Epidemiology and Biostatistics, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Therapeutic Effectiveness Research, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Charles E Leonard
- Center for Clinical Epidemiology and Biostatistics, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Therapeutic Effectiveness Research, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
30
|
Hammid A, Fallon JK, Lassila T, Salluce G, Smith PC, Tolonen A, Sauer A, Urtti A, Honkakoski P. Carboxylesterase Activities and Protein Expression in Rabbit and Pig Ocular Tissues. Mol Pharm 2021; 18:1305-1316. [PMID: 33595329 PMCID: PMC8023712 DOI: 10.1021/acs.molpharmaceut.0c01154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022]
Abstract
Hydrolytic reactions constitute an important pathway of drug metabolism and a significant route of prodrug activation. Many ophthalmic drugs and prodrugs contain ester groups that greatly enhance their permeation across several hydrophobic barriers in the eye before the drugs are either metabolized or released, respectively, via hydrolysis. Thus, the development of ophthalmic drug therapy requires the thorough profiling of substrate specificities, activities, and expression levels of ocular esterases. However, such information is scant in the literature, especially for preclinical species often used in ophthalmology such as rabbits and pigs. Therefore, our aim was to generate systematic information on the activity and expression of carboxylesterases (CESs) and arylacetamide deacetylase (AADAC) in seven ocular tissue homogenates from these two species. The hydrolytic activities were measured using a generic esterase substrate (4-nitrophenyl acetate) and, in the absence of validated substrates for rabbit and pig enzymes, with selective substrates established for human CES1, CES2, and AADAC (d-luciferin methyl ester, fluorescein diacetate, procaine, and phenacetin). Kinetics and inhibition studies were conducted using these substrates and, again due to a lack of validated rabbit and pig CES inhibitors, with known inhibitors for the human enzymes. Protein expression levels were measured using quantitative targeted proteomics. Rabbit ocular tissues showed significant variability in the expression of CES1 (higher in cornea, lower in conjunctiva) and CES2 (higher in conjunctiva, lower in cornea) and a poor correlation of CES expression with hydrolytic activities. In contrast, pig tissues appear to express only CES1, and CES3 and AADAC seem to be either low or absent, respectively, in both species. The current study revealed remarkable species and tissue differences in ocular hydrolytic enzymes that can be taken into account in the design of esterase-dependent prodrugs and drug conjugates, the evaluation of ocular effects of systemic drugs, and in translational and toxicity studies.
Collapse
Affiliation(s)
- Anam Hammid
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70210 Kuopio, Finland
| | - John K. Fallon
- Division
of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School
of Pharmacy, University of North Carolina
at Chapel Hill, Campus Box 7355, Chapel Hill, North Carolina 27599-7355, United States
| | | | - Giulia Salluce
- Centro
Singular de Investigación en Química Biolóxica
e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago
de Compostela, Spain
| | - Philip C. Smith
- Division
of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School
of Pharmacy, University of North Carolina
at Chapel Hill, Campus Box 7355, Chapel Hill, North Carolina 27599-7355, United States
| | - Ari Tolonen
- Admescope
Ltd, Typpitie 1, 90620 Oulu, Finland
| | - Achim Sauer
- Department
of Drug Discovery Sciences, Boehringer Ingelheim
Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Arto Urtti
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70210 Kuopio, Finland
- Institute
of Chemistry, Saint Petersburg State University, Universitetskii pr. 26, 198584 Saint Petersburg, Russia
- Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland
| | - Paavo Honkakoski
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70210 Kuopio, Finland
- Division
of Pharmacotherapy and Experimental Therapeutics, Eshelman School
of Pharmacy, University of North Carolina
at Chapel Hill, Campus Box 7569, Chapel Hill, North Carolina 27599-7569, United States
| |
Collapse
|
31
|
Solé M, Freitas R, Rivera-Ingraham G. The use of an in vitro approach to assess marine invertebrate carboxylesterase responses to chemicals of environmental concern. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 82:103561. [PMID: 33307128 DOI: 10.1016/j.etap.2020.103561] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 06/12/2023]
Abstract
Carboxylesterases (CEs) are key enzymes which catalyse the hydrolysis reactions of multiple xenobiotics and endogenous ester moieties. Given their growing interest in the context of marine pollution and biomonitoring, this study focused on the in vitro sensitivity of marine invertebrate CEs to some pesticides, pharmaceuticals, personal care products and plastic additives to assess their potential interaction on this enzymatic system and its suitability as biomarkers. Three bivalves, one gastropod and two crustaceans were used and CEs were quantified following current protocols set for mammalian models. Four substrates were screened for CEs determination and to test their adequacy in the hepatic fraction measures of the selected invertebrates. Two commercial recombinant human isoforms (hCE1 and hCE2) were also included for methodological validation. Among the invertebrates, mussels were revealed as the most sensitive to xenobiotic exposures while gastropods were the least as well as with particular substrate-specific preferences. Among chemicals of environmental concern, the plastic additive tetrabromobisphenol A displayed the highest CE-inhibitory capacity in all species. Since plastic additives easily breakdown from the polymer and may accumulate and metabolise in marine biota, their interaction with the CE key metabolic/detoxification processes may have consequences in invertebrate's physiology, affect bioaccumulation and therefore trophic web transfer and, ultimately, human health as shellfish consumers.
Collapse
Affiliation(s)
- Montserrat Solé
- Institute of Marine Sciences (ICM-CSIC), Pg. Marítim de la Barceloneta 37-49, 08003, Barcelona, Spain.
| | - Rosa Freitas
- Department of Biology & CESAM, Universidade de Aveiro, 3810-193, Aveiro, Portugal
| | - Georgina Rivera-Ingraham
- Laboratorio de Fisiología y Genética Marina, Centro de Estudios Avanzados en Zonas Áridas, Universidad Católica del Norte, Larrondo, 1281, Coquimbo, Chile
| |
Collapse
|
32
|
Hirosawa K, Fukami T, Tashiro K, Sakai Y, Kisui F, Nakano M, Nakajima M. Role of Human Arylacetamide Deacetylase (AADAC) on Hydrolysis of Eslicarbazepine Acetate and Effects of AADAC Genetic Polymorphisms on Hydrolase Activity. Drug Metab Dispos 2021; 49:322-329. [DOI: 10.1124/dmd.120.000295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/07/2021] [Indexed: 12/31/2022] Open
|
33
|
Liu S, Wang Z, Tian X, Cai W. Predicting the Effects of CYP2C19 and Carboxylesterases on Vicagrel, a Novel P2Y12 Antagonist, by Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling Approach. Front Pharmacol 2021; 11:591854. [PMID: 33424602 PMCID: PMC7793822 DOI: 10.3389/fphar.2020.591854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/22/2020] [Indexed: 11/13/2022] Open
Abstract
Vicagrel, a novel acetate derivative of clopidogrel, exhibits a favorable safety profile and excellent antiplatelet activity. Studies aim at identifying genetic and non-genetic factors affecting vicagrel metabolic enzymes Cytochrome P450 2C19 (CYP2C19), Carboxylesterase (CES) 1 and 2 (CES1 and CES2), which may potentially lead to altered pharmacokinetics and pharmacodynamics, are warranted. A physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) model incorporating vicagrel and its metabolites was constructed, verified and validated in our study, which could simultaneously characterize its sequential two step metabolism and clinical response. Simulations were then performed to evaluate the effects of CYP2C19, CES1 and CES2 genetic polymorphisms as well as inhibitors of these enzymes on vicagrel pharmacokinetics and antiplatelet effects. Results suggested vicagrel was less influenced by CYP2C19 metabolic phenotypes and CES1 428 G > A variation, in comparison to clopidogrel. No pharmacokinetic difference in the active metabolite was also noted for volunteers carrying different CES2 genotypes. Omeprazole, a CYP2C19 inhibitor, and simvastatin, a CES1 and CES2 inhibitor, showed weak impact on the pharmacokinetics and pharmacodynamics of vicagrel. This is the first study proposing a dynamic PBPK/PD model of vicagrel able to capture its pharmacokinetic and pharmacodynamic profiles simultaneously. Simulations indicated that genetic polymorphisms and drug-drug interactions showed no clinical relevance for vicagrel, suggesting its potential advantages over clopidogrel for treatment of cardiovascular diseases. Our model can be utilized to support further clinical trial design aiming at exploring the effects of genetic polymorphisms and drug-drug interactions on PK and PD of this novel antiplatelet agent.
Collapse
Affiliation(s)
- Shuaibing Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ziteng Wang
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, China
| | - Xin Tian
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weimin Cai
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Jithavech P, Ratnatilaka Na Bhuket P, Supasena W, Qiu G, Ye S, Wu J, Wong TW, Rojsitthisak P. In Vitro Hepatic Metabolism of Curcumin Diethyl Disuccinate by Liver S9 from Different Animal Species. Front Pharmacol 2020; 11:577998. [PMID: 33312126 PMCID: PMC7703437 DOI: 10.3389/fphar.2020.577998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Liver S9 (LS9) is a nearly complete collection of all hepatic drug-metabolizing enzymes. It is a low-cost model for predicting drug metabolic activity. This study aimed to identify the suitability of using LS9 of different animal sources in drug metabolism profiling with respect to the possible translation of the in vitro outcomes to clinical studies. The in vitro hepatic metabolism of curcumin diethyl disuccinate (CDD) in LS9 of rats, dogs, monkeys, and humans was evaluated. The identity of CDD metabolites and the metabolism kinetic parameters, including degradation rate constant, in vitro/in vivo intrinsic clearance, and half-life, were determined. CDD was rapidly metabolized into monoethylsuccinyl curcumin and curcumin in LS9 of all tested species mainly by carboxylesterases (CESs), including CES1 and CES2, and butyrylcholinesterase. The in vitro intrinsic clearance of CDD was in the order of human > dog > monkey > rat, whereas that of monoethylsuccinyl curcumin in the order of dog > monkey > human > rat; this parameter was not correlated with their respective in vivo clearance, which followed the order of dog > monkey > rat > human. Therefore, in vitro drug metabolism data inferred from LS9 of nonhuman origin, especially from monkeys and dogs, cannot be used as preclinical data for human trials, as humans have a smaller liver-to-body weight ratio than monkeys, dogs, and rats. The in vivo drug metabolism is dictated by the anatomical factors of the test subject.
Collapse
Affiliation(s)
- Ponsiree Jithavech
- Pharmaceutical Chemistry and Natural Products Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Natural Products for Ageing and Chronic Diseases Research Unit, Chulalongkorn University, Bangkok, Thailand
| | | | - Wiwat Supasena
- Natural Products for Ageing and Chronic Diseases Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Guanyinsheng Qiu
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing, China
| | - Shengqing Ye
- School of Pharmaceutical and Materials Engineering & Institute for Advanced Studies, Taizhou University, Taizhou, China
| | - Jie Wu
- School of Pharmaceutical and Materials Engineering & Institute for Advanced Studies, Taizhou University, Taizhou, China
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA, Puncak Alam, Malaysia
| | - Pornchai Rojsitthisak
- Natural Products for Ageing and Chronic Diseases Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
35
|
Zhao YS, Qian XK, Guan XQ, Song PF, Song YQ, He RJ, Sun MR, Wang XY, Zou LW, Ge GB. Discovery of natural alkaloids as potent and selective inhibitors against human carboxylesterase 2. Bioorg Chem 2020; 105:104367. [PMID: 33080495 DOI: 10.1016/j.bioorg.2020.104367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/02/2020] [Accepted: 10/08/2020] [Indexed: 01/10/2023]
Abstract
Human Carboxylesterase 2A (hCES2A), one of the most important serine hydrolases, plays crucial roles in the hydrolysis and the metabolic activation of a wide range of esters and amides. Increasing evidence has indicated that potent inhibition on intestinal hCES2A may reduce the excessive accumulation of SN-38 (the hydrolytic metabolite of irinotecan with potent cytotoxicity) in the intestinal tract and thereby alleviate the intestinal toxicity triggered by irinotecan. In this study, more than sixty natural alkaloids have been collected and their inhibitory effects against hCES2A are assayed using a fluorescence-based biochemical assay. Following preliminary screening, seventeen alkaloids are found with strong to moderate hCES2A inhibition activity. Primary structure-activity relationships (SAR) analysis of natural isoquinoline alkaloids reveal that the benzo-1,3-dioxole group and the aromatic pyridine structure are beneficial for hCES2A inhibition. Further investigations demonstrate that a steroidal alkaloid reserpine exhibits strong hCES2A inhibition activity (IC50 = 0.94 μM) and high selectivity over other human serine hydrolases including hCES1A, dipeptidyl peptidase IV (DPP-IV), butyrylcholinesterase (BChE) and thrombin. Inhibition kinetic analyses demonstrated that reserpine acts as a non-competitive inhibitor against hCES2A-mediated FD hydrolysis. Molecular docking simulations demonstrated that the potent inhibition of hCES2A by reserpine could partially be attributed to its strong σ-π and S-π interactions between reserpine and hCES2A. Collectively, our findings suggest that reserpine is a potent and highly selective inhibitor of hCES2A, which can be served as a promising lead compound for the development of more efficacious and selective alkaloids-type hCES2A inhibitors for biomedical applications.
Collapse
Affiliation(s)
- Yi-Shu Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xing-Kai Qian
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao-Qing Guan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pei-Fang Song
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yun-Qing Song
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rong-Jing He
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meng-Ru Sun
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiu-Yang Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li-Wei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
36
|
Khojasteh SC, Driscoll JP, Jackson KD, Miller GP, Mitra K, Rietjens IMCM, Zhang D. Novel advances in biotransformation and bioactivation research-2019 year in review .. Drug Metab Rev 2020; 52:333-365. [PMID: 32645275 PMCID: PMC10805366 DOI: 10.1080/03602532.2020.1772281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/14/2020] [Indexed: 01/25/2023]
Abstract
Biotransformation is one of the main mechanisms used by the body to eliminate drugs. As drug molecules become more complicated, the involvement of drug metabolizing enzymes increases beyond those that are typically studied, such as the cytochrome P450 enzymes. In this review, we try to capture the many outstanding articles that were published in the past year in the field of biotransformation (see Table 1). We have divided the articles into two categories of (1) metabolites and drug metabolizing enzymes, and (2) bioactivation and safety. This annual review is the fifth of its kind since 2016 (Baillie et al. 2016; Khojasteh et al. 2017, 2018, 2019). This effort in itself also continues to evolve. We have followed the same format we used in previous years in terms of the selection of articles and the authoring of each section. I am pleased of the continued support of Rietjens, Miller, Zhang, Driscoll and Mitra to this review. We would like to welcome Klarissa D. Jackson as a new author for this year's issue. We strive to maintain a balance of authors from academic and industry settings. We would be pleased to hear your opinions of our commentary, and we extend an invitation to anyone who would like to contribute to a future edition of this review. Cyrus Khojasteh, on behalf of the authors.
Collapse
Affiliation(s)
- S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - James P Driscoll
- Department of Drug Metabolism and Pharmacokinetics, MyoKardia, Inc, South San Francisco, CA, USA
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Grover P Miller
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kaushik Mitra
- Department of Safety Assessment and Laboratory Animal Resources, Merck Research Laboratories (MRL), Merck & Co., Inc, West Point, PA, USA
| | | | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
37
|
Qian Y, Markowitz JS. Natural Products as Modulators of CES1 Activity. Drug Metab Dispos 2020; 48:993-1007. [PMID: 32591414 DOI: 10.1124/dmd.120.000065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/12/2020] [Indexed: 12/30/2022] Open
Abstract
Carboxylesterase (CES) 1 is the predominant esterase expressed in the human liver and is capable of catalyzing the hydrolysis of a wide range of therapeutic agents, toxins, and endogenous compounds. Accumulating studies have demonstrated associations between the expression and activity of CES1 and the pharmacokinetics and/or pharmacodynamics of CES1 substrate medications (e.g., methylphenidate, clopidogrel, oseltamivir). Therefore, any perturbation of CES1 by coingested xenobiotics could potentially compromise treatment. Natural products are known to alter drug disposition by modulating cytochrome P450 and UDP-glucuronosyltransferase enzymes, but this issue is less thoroughly explored with CES1. We report the results of a systematic literature search and discuss natural products as potential modulators of CES1 activity. The majority of research reports reviewed were in vitro investigations that require further confirmation through clinical study. Cannabis products (Δ 9-tetrahydrocannabinol, cannabidiol, cannabinol); supplements from various plant sources containing naringenin, quercetin, luteolin, oleanolic acid, and asiatic acid; and certain traditional medicines (danshen and zhizhuwan) appear to pose the highest inhibition potential. In addition, ursolic acid, gambogic acid, and glycyrrhetic acid, if delivered intravenously, may attain high enough systemic concentrations to significantly inhibit CES1. The provision of a translational interpretation of in vitro assessments of natural product actions and interactions is limited by the dearth of basic pharmacokinetic data of the natural compounds exhibiting potent in vitro influences on CES1 activity. This is a major impediment to assigning even potential clinical significance. The modulatory effects on CES1 expression after chronic exposure to natural products warrants further investigation. SIGNIFICANCE STATEMENT: Modulation of CES1 activity by natural products may alter the course of treatment and clinical outcome. In this review, we have summarized the natural products that can potentially interact with CES1 substrate medications. We have also noted the limitations of existing reports and outlined challenges and future directions in this field.
Collapse
Affiliation(s)
- Yuli Qian
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - John S Markowitz
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| |
Collapse
|
38
|
The influence of carboxylesterase 1 polymorphism and cannabidiol on the hepatic metabolism of heroin. Chem Biol Interact 2019; 316:108914. [PMID: 31837295 DOI: 10.1016/j.cbi.2019.108914] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/21/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023]
Abstract
Heroin (diamorphine) is a highly addictive opioid drug synthesized from morphine. The use of heroin and incidence of heroin associated overdose death has increased sharply in the US. Heroin is primarily metabolized via deacetylation (hydrolysis) forming the active metabolites 6-monoacetylmorphine (6-MAM) and morphine. A diminution in heroin hydrolysis is likely to cause higher drug effects and toxicities. In this study, we sought to determine the contribution of the major hepatic hydrolase carboxylesterase 1 (CES1) to heroin metabolism in the liver as well as the potential influence of one of its known genetic variants, G143E (rs71647871). Furthermore, given the potential therapeutic application of cannabidiol (CBD) for heroin addiction and the frequent co-abuse of cannabis and heroin, we also assessed the effects of CBD on heroin metabolism. In vitro systems containing human liver, wild-type CES1, and G143E CES1 S9 fractions were utilized in the assessment. The contribution of CES1 to the hydrolysis of heroin to 6-MAM was determined as 3.66%, and CES1 was unable to further catalyze 6-MAM under our assay conditions. The G143E variant showed a 3.2-fold lower intrinsic clearance of heroin as compared to the WT. CBD inhibited heroin and 6-MAM hydrolysis in a reversible manner, with IC50s of 14.7 and 12.1 μM, respectively. Our study results suggested only minor involvement of CES1 in heroin hydrolysis in the liver. Therefore, the G143E variant is unlikely to cause significant impact despite a much lower hydrolytic activity. CBD exhibited potent in vitro inhibition toward both heroin and 6-MAM hydrolysis, which may be of potential clinical relevance.
Collapse
|
39
|
Tian X, Yan F, Zheng J, Cui X, Feng L, Li S, Jin L, James TD, Ma X. Endoplasmic Reticulum Targeting Ratiometric Fluorescent Probe for Carboxylesterase 2 Detection in Drug-Induced Acute Liver Injury. Anal Chem 2019; 91:15840-15845. [PMID: 31713417 DOI: 10.1021/acs.analchem.9b04189] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Carboxylesterase 2 (CES2), an endoplasmic reticulum (ER) located phase I enzyme, plays a vital role in the metabolism of various endogenous and exogenous substances, and is regarded as an important target for the design of prodrugs. Unfortunately, superior highly selective ER targeting fluorescent probes for monitoring of CES2 are not currently available. Herein, we report an ER targeting CES2 selective and sensitive ratiometric fluorescent probe ERNB based on the ER localizing group p-toluenesulfonamide. ERNB possessed high specificity, sensitivity, and exhibited excellent subcellular localization when compared to commercial ER tracker, and was used to image CES2 in the ER of living cells. Additionally, using ERNB we evaluated the CES2 regulation under d,l-dithiothreitol and tunicamycin-induced ER stress. Furthermore, we determined the down regulation of CES2 activity and expression in the acetaminophen-induced acute liver injury model. On the basis of these results, we conclude that ERNB is a promising tool for highlighting the role of CES2 in the ER and in exploring the role of CES2 in the development of diseases associated with ER stress.
Collapse
Affiliation(s)
- Xiangge Tian
- Academy of Integrative Medicine, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, College of Pharmacy , Dalian Medical University , Lvshun South Road No. 9 , Dalian 116044 , China
| | - Fei Yan
- Academy of Integrative Medicine, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, College of Pharmacy , Dalian Medical University , Lvshun South Road No. 9 , Dalian 116044 , China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , 209 Tongshan Road , Xuzhou , Jiangsu 221004 , China
| | - Jingyuan Zheng
- Academy of Integrative Medicine, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, College of Pharmacy , Dalian Medical University , Lvshun South Road No. 9 , Dalian 116044 , China
| | - Xiaolin Cui
- Academy of Integrative Medicine, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, College of Pharmacy , Dalian Medical University , Lvshun South Road No. 9 , Dalian 116044 , China
| | - Lei Feng
- Academy of Integrative Medicine, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, College of Pharmacy , Dalian Medical University , Lvshun South Road No. 9 , Dalian 116044 , China
| | - Sheng Li
- Academy of Integrative Medicine, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, College of Pharmacy , Dalian Medical University , Lvshun South Road No. 9 , Dalian 116044 , China
| | - Lingling Jin
- Academy of Integrative Medicine, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, College of Pharmacy , Dalian Medical University , Lvshun South Road No. 9 , Dalian 116044 , China
| | - Tony D James
- Department of Chemistry , University of Bath , Bath BA2 7AY , United Kingdom
| | - Xiaochi Ma
- Academy of Integrative Medicine, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, College of Pharmacy , Dalian Medical University , Lvshun South Road No. 9 , Dalian 116044 , China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , 209 Tongshan Road , Xuzhou , Jiangsu 221004 , China
| |
Collapse
|
40
|
Wang YQ, Shang XF, Wang L, Zhang P, Zou LW, Song YQ, Hao DC, Fang SQ, Ge GB, Tang H. Interspecies variation of clopidogrel hydrolysis in liver microsomes from various mammals. Chem Biol Interact 2019; 315:108871. [PMID: 31669218 DOI: 10.1016/j.cbi.2019.108871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/09/2019] [Accepted: 10/21/2019] [Indexed: 02/08/2023]
Abstract
Clopidogrel, a clinically used antiplatelet agent, can be readily hydrolyzed by human carboxylesterase 1A (CES1A) to release an inactive metabolite clopidogrel carboxylic acid (CCA). In this study, clopidogrel was used as a tool substrate to investigate the interspecies variation of clopidogrel hydrolysis in hepatic microsomes from various mammals including human and six laboratory animals (such as mouse, rat, rabbit, beagle dog, minipig and cynomolgus monkey). The results demonstrated that clopidogrel could be hydrolyzed into CCA by all tested hepatic microsomes from human or other mammals, but the hydrolytic rates greatly varied among species. Inhibition assays demonstrated that BNPP (an inactivator of mammalian CES) strongly inactivated clopidogrel hydrolytic activity in all tested hepatic microsomes, suggested that mammalian CES were major contributor(s) responsible for clopidogrel hydrolysis in hepatic preparations from all above-mentioned species. By contrast, the response of a reversible inhibitor of human CES1A on clopidogrel hydrolysis in these liver preparations varied significantly among different species. Moreover, the enzymatic kinetics and the apparent kinetic parameters of clopidogrel hydrolysis in hepatic microsomes from various animal species were evaluated and compared to each other. These findings provide crucial information for deeply understanding the differences in catalytic behaviors of mammalian CES, which will be very helpful for choosing suitable laboratory animal(s) for whole tests of CES1A substrate-drugs.
Collapse
Affiliation(s)
- Ya-Qiao Wang
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, 832000, China
| | - Xiao-Feng Shang
- Zhangye People's Hospital affiliated to Hexi University, Zhangye, Gansu, 734000, China
| | - Lu Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, 832000, China
| | - Ping Zhang
- Department of Cardiology, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Li-Wei Zou
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China
| | - Yun-Qing Song
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China
| | - Da-Cheng Hao
- Dalian Jiaotong University, Dalian, 116028, China
| | - Sheng-Quan Fang
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China
| | - Guang-Bo Ge
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China.
| | - Hui Tang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, 832000, China.
| |
Collapse
|
41
|
Kato Y, Kawai M, Kawai S, Okano Y, Rokkaku N, Ishisaka A, Murota K, Nakamura T, Nakamura Y, Ikushiro S. Dynamics of the Cellular Metabolism of Leptosperin Found in Manuka Honey. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:10853-10862. [PMID: 31496237 DOI: 10.1021/acs.jafc.9b03894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Leptosperin (methyl syringate β-d-gentiobioside) is abundantly found in manuka honey, which is widely used because of its antibacterial and possible anti-inflammatory activities. The aim of this study was to examine the molecular mechanism underlying the metabolism of leptosperin. Five phytochemicals (leptosperin, methyl syringate (MSYR), glucuronate conjugate of MSYR (MSYR-GA), sulfonate conjugate of MSYR (MSYR-S), and syringic acid (SYR)) were separately incubated with HepG2 and Caco-2 cells. After incubation, we found that the concentration of MSYR decreased, whereas the concentrations of SYR, MSYR-GA, and MSYR-S increased. By profiling with inhibitors and carboxylesterases (CES1, 2), we found that the conversion from MSYR to SYR was mediated by CES1. Lipopolysaccharide-stimulated RAW264.7 cells restored MSYR-GA to MSYR possibly by the secreted β-glucuronidase. All of the mice administered with leptosperin, MSYR, or manuka honey showed higher MSYR (13.84 ± 11.51, 14.29 ± 9.19, or 6.66 ± 2.30 nM) and SYR (1.85 ± 0.66, 6.01 ± 1.20, or 8.16 ± 3.10 nM) levels in the plasma compared with that of the vehicle controls (3.33 ± 1.45 (MSYR) and 1.85 ± 0.66 (SYR) nM). The findings of our study indicate that the unique metabolic pathways of these compounds may account for possible functionalities of manuka honey.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kaeko Murota
- Faculty of Life and Environmental Science , Shimane University , Matsue , Shimane 690-8504 , Japan
| | - Toshiyuki Nakamura
- Graduate School of Environmental and Life Science , Okayama University , Okayama 700-0082 , Japan
| | - Yoshimasa Nakamura
- Graduate School of Environmental and Life Science , Okayama University , Okayama 700-0082 , Japan
| | - Shinichi Ikushiro
- Department of Biotechnology , Toyama Prefectural University , Imizu , Toyama 939-0398 , Japan
| |
Collapse
|
42
|
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: 1.8] [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]
|
43
|
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: 114] [Impact Index Per Article: 22.8] [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
|
44
|
Briand E, Thomsen R, Linnet K, Rasmussen HB, Brunak S, Taboureau O. Combined Ensemble Docking and Machine Learning in Identification of Therapeutic Agents with Potential Inhibitory Effect on Human CES1. Molecules 2019; 24:molecules24152747. [PMID: 31362390 PMCID: PMC6696021 DOI: 10.3390/molecules24152747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/11/2019] [Accepted: 07/24/2019] [Indexed: 01/08/2023] Open
Abstract
The human carboxylesterase 1 (CES1), responsible for the biotransformation of many diverse therapeutic agents, may contribute to the occurrence of adverse drug reactions and therapeutic failure through drug interactions. The present study is designed to address the issue of potential drug interactions resulting from the inhibition of CES1. Based on an ensemble of 10 crystal structures complexed with different ligands and a set of 294 known CES1 ligands, we used docking (Autodock Vina) and machine learning methodologies (LDA, QDA and multilayer perceptron), considering the different energy terms from the scoring function to assess the best combination to enable the identification of CES1 inhibitors. The protocol was then applied on a library of 1114 FDA-approved drugs and eight drugs were selected for in vitro CES1 inhibition. An inhibition effect was observed for diltiazem (IC50 = 13.9 µM). Three others drugs (benztropine, iloprost and treprostinil), exhibited a weak CES1 inhibitory effects with IC50 values of 298.2 µM, 366.8 µM and 391.6 µM respectively. In conclusion, the binding site of CES1 is relatively flexible and can adapt its conformation to different types of ligands. Combining ensemble docking and machine learning approaches improves the prediction of CES1 inhibitors compared to a docking study using only one crystal structure.
Collapse
Affiliation(s)
- Eliane Briand
- INSERM U1133, CNRS UMR 8251, Unit of functional and adaptive biology, Université de Paris, Paris 75013, France
| | - Ragnar Thomsen
- Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Kristian Linnet
- Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Henrik Berg Rasmussen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, 4000 Roskilde, Denmark
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Olivier Taboureau
- INSERM U1133, CNRS UMR 8251, Unit of functional and adaptive biology, Université de Paris, Paris 75013, France.
| |
Collapse
|
45
|
Gabriele M, Puccini P, Lucchi M, Aprile V, Gervasi PG, Longo V. Arylacetamide Deacetylase Enzyme: Presence and Interindividual Variability in Human Lungs. Drug Metab Dispos 2019; 47:961-965. [PMID: 31235486 DOI: 10.1124/dmd.119.087031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Human arylacetamide deacetylase (AADAC) is a single microsomal serine esterase involved in the hydrolysis of many acetyl-containing drugs. To date, the presence and activity of the AADAC enzyme in human lungs has been scarcely examined. We investigated its gene and protein expression as well as interindividual variations in AADAC activities in a large number of human lungs (n = 25) using phenacetin as a selective substrate. The kinetic parameters K m and V max were determined. Our findings highlighted a high interindividual variability in both AADAC mRNA levels and hydrolysis activities. Furthermore, for the first time we demonstrated the presence of the AADAC protein in various lung samples by means of immunoblot analysis. As a comparison, phenacetin hydrolysis was detected in pooled human liver microsomes. Lung activities were much lower than those found in the liver. However, similar K m values were found, which suggests that this hydrolysis could be due to the same enzyme. Pulmonary phenacetin hydrolysis proved to be positively correlated with AADAC mRNA (*P < 0.05) and protein (*P < 0.05) levels. Moreover, the average values of AADAC activity in smokers was significantly higher than in nonsmoker subjects (*P < 0.05), and this might have an important role in the administration of some drugs. These findings add more information to our knowledge of pulmonary enzymes and could be particularly useful in the design and preclinical development of inhaled drugs. SIGNIFICANCE STATEMENT: This study investigated the presence and activity of the AADAC enzyme in several human lungs. Our results highlight high interindividual variability in both AADAC gene and protein expression as well as in phenacetin hydrolysis activity. These findings add more information to our knowledge of pulmonary enzymes and could be particularly useful in the design and preclinical development of inhaled drugs.
Collapse
Affiliation(s)
- Morena Gabriele
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Paola Puccini
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Marco Lucchi
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Vittorio Aprile
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Pier Giovanni Gervasi
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Vincenzo Longo
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| |
Collapse
|
46
|
Zhao YS, Ruan HL, Wang XY, Chen C, Song PF, Lü CW, Zou LW. Catalyst-free visible-light-induced condensation to synthesize bis(indolyl)methanes and biological activity evaluation of them as potent human carboxylesterase 2 inhibitors. RSC Adv 2019; 9:40168-40175. [PMID: 35541371 PMCID: PMC9076199 DOI: 10.1039/c9ra08593a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 11/25/2019] [Indexed: 11/21/2022] Open
Abstract
A mild strategy for visible-light-induced synthesis of bis(indolyl)methanes was developed using aromatic aldehydes and indole as substrates. This reaction could be performed at room temperature under catalyst- and additive-free conditions to synthesize a series of bis(indolyl)methanes in good to excellent yields. In addition, all synthesized bis(indolyl)methanes together with β-substituted indole derivatives synthesized according to our previous work, were evaluated for their inhibitory effect against human carboxylesterase (CES1 and CES2). Primary structure–activity relationship analysis of all tested compounds showed that the modifications of β-substituted indole at the β-site with another indolyl group led to a significant enhancement of the inhibitory effect on CES2, and the bisindolyl structure is essential for CES2 inhibition. These results demonstrated that these bis(indolyl)methanes are potent and selective CES2 inhibitors, which might be helpful for medicinal chemists to design and develop more potent and selective CES2 inhibitors for biomedical applications. Bis(indolyl)methanes were synthesized by a green protocol. Primary structure–activity relationship analysis showed that the bisindolyl structure is essential for CES2 inhibition.![]()
Collapse
Affiliation(s)
- Yi-Shu Zhao
- Institute of Interdisciplinary Integrative Medicine Research
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- People's Republic of China
| | - Hong-Li Ruan
- School of Chemistry and Chemical Engineering
- Liaoning Normal University
- Dalian 116029
- People's Republic of China
| | - Xiu-Yang Wang
- Institute of Interdisciplinary Integrative Medicine Research
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- People's Republic of China
| | - Chen Chen
- School of Chemistry and Chemical Engineering
- Liaoning Normal University
- Dalian 116029
- People's Republic of China
| | - Pei-Fang Song
- Institute of Interdisciplinary Integrative Medicine Research
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- People's Republic of China
| | - Cheng-Wei Lü
- School of Chemistry and Chemical Engineering
- Liaoning Normal University
- Dalian 116029
- People's Republic of China
| | - Li-Wei Zou
- Institute of Interdisciplinary Integrative Medicine Research
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- People's Republic of China
| |
Collapse
|
47
|
Huttunen K. Identification of human, rat and mouse hydrolyzing enzymes bioconverting amino acid ester prodrug of ketoprofen. Bioorg Chem 2018; 81:494-503. [DOI: 10.1016/j.bioorg.2018.09.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 12/25/2022]
|
48
|
Wang YQ, Weng ZM, Dou TY, Hou J, Wang DD, Ding LL, Zou LW, Yu Y, Chen J, Tang H, Ge GB. Nevadensin is a naturally occurring selective inhibitor of human carboxylesterase 1. Int J Biol Macromol 2018; 120:1944-1954. [DOI: 10.1016/j.ijbiomac.2018.09.178] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/26/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
|
49
|
Konishi K, Fukami T, Ogiso T, Nakajima M. In vitro approach to elucidate the relevance of carboxylesterase 2 and N-acetyltransferase 2 to flupirtine-induced liver injury. Biochem Pharmacol 2018; 155:242-251. [PMID: 30028988 DOI: 10.1016/j.bcp.2018.07.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/14/2018] [Indexed: 12/30/2022]
Abstract
The use of flupirtine, an analgesic, has been restricted in European countries because it causes liver injury in rare cases. Flupirtine is primarily metabolized to D-13223, an acetylamino form. In the process of D-13223 formation, it has been hypothesized that a reactive metabolite is formed which may be involved in flupirtine hepatotoxicity. The purpose of this study was to identify the potential reactive metabolite and the responsible enzymes in the human liver to get a clue to the mechanism of hepatotoxicity. Using recombinant enzymes, we found that D-13223 was formed from flupirtine via hydrolysis by carboxylesterase 2 (CES2) and subsequent acetylation by N-acetyltransferase (NAT) 2. A conjugate of N-acetyl-l-cysteine (NAC), a nucleophile, was detected by incubation of flupirtine with CES2, and the conjugate formation in human liver microsomes was inhibited by CES2 inhibitors, indicating that a reactive metabolite, which may be a quinone diimine, was produced in the process of CES2-mediated hydrolysis of flupirtine. The formation of the NAC conjugate in liver S9 samples from NAT2 slow acetylators was significantly higher than that from NAT2 rapid/intermediate acetylators, indicating that NAT2 could function as a detoxification enzyme for flupirtine. CES2-overexpressing HepG2 cells showed remarkable lactate dehydrogenase leakage under flupirtine treatment, while no cytotoxicity was observed in control cells, suggesting that the reactive metabolite formed by CES2-mediated hydrolysis of flupirtine would be a trigger of hepatotoxicity. NAT2 slow acetylators with high CES2 activity could be highly susceptible to flupirtine-induced liver injury.
Collapse
Affiliation(s)
- Keigo Konishi
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI Nano-LSI), Kanazawa University, Kanazawa, Japan.
| | - Takuo Ogiso
- 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 Nano-LSI), Kanazawa University, Kanazawa, Japan
| |
Collapse
|
50
|
Quiroga AD, Lehner R. Pharmacological intervention of liver triacylglycerol lipolysis: The good, the bad and the ugly. Biochem Pharmacol 2018; 155:233-241. [PMID: 30006193 DOI: 10.1016/j.bcp.2018.07.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023]
Abstract
Excessive triacylglycerol (TG) accumulation is the distinctive feature of obesity. In the liver, sustained TG accretion leads to nonalcoholic fatty liver disease (NAFLD), eventually progressing to non-alcoholic steatohepatitis (NASH) and cirrhosis, which is associated with complications including hepatic failure, hepatocellular carcinoma and death. Pharmacological interventions are actively pursued to prevent lipid accumulation in hepatocytes and, therefore, to ameliorate the associated pathophysiological conditions. Here, we sought to provide an overview of the pharmacological approaches to up- or downregulate the expression and activities of the enzymes involved in hepatic TG hydrolysis. Fatty acids (FA) released by hydrolysis of hepatic TG can be used for β-oxidation, signaling, and for very low-density lipoprotein (VLDL)-TG synthesis. Originally, lipolysis was believed to be centered in the adipose and to be catalyzed by only two lipases, hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MAGL). However, genetic ablation of HSL expression in mice failed to erase TG hydrolysis in adipocytes leading to the identification of a third lipase termed adipose triglyceride lipase (ATGL). Although these three enzymes are considered to be the main players governing lipolysis in the adipocyte, other lipolytic enzymes have been described to contribute to hepatic TG metabolism. These include adiponutrin/patatin-like phospholipase domain containing 3 (PNPLA3), some members of the carboxylesterase family (CES/Ces), arylacetamide deacetylase (AADAC), lysosomal acid lipase (LAL) and hepatic lipase (HL). This review highlights the consequences of pharmacological interventions of liver lipases that degrade TG in cytosolic lipid droplets, in the endoplasmic reticulum, in the late endosomes/lysosomes and along the secretory route.
Collapse
Affiliation(s)
- Ariel D Quiroga
- Instituto de Fisiología Experimental (IFISE), Área Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, UNR, Rosario, Argentina.
| | - Richard Lehner
- Group on Molecular and Cell Biology of Lipids, Department of Pediatrics, Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|