1
|
Conchon Costa AC, Medeiros JIM, Kang W, Yamamoto PA, de Gaitani CM, Vasconcelos MED, Da Silva RM, Kemp R, Sankarankutty AK, Salgado W, Santos JS, Schmidt S, De Moraes NV. Redefining Statin Dosage Post-Gastric Bypass: Insights from a Population Pharmacokinetics-Pharmacodynamics Link Approach. J Clin Pharmacol 2024. [PMID: 39101567 DOI: 10.1002/jcph.6110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/19/2024] [Indexed: 08/06/2024]
Abstract
Roux-en-Y gastric bypass (RYGB) involves creating a small stomach pouch, bypassing part of the small intestine, and rerouting the digestive tract. These alterations can potentially change the drug exposure and response. Our primary aim was to assess the impact of RYGB on the pharmacokinetics of simvastatin lactone (SV) and its active metabolite, simvastatin hydroxy acid (SVA). Ultimately, we aimed to optimize dosing for this understudied population by employing a population pharmacokinetic-pharmacodynamic link approach. The study comprised patients who had undergone RYGB surgery and individuals without a previous history of RYGB. All participants received a single oral dose of simvastatin. Plasma concentration data were analyzed with a nonlinear mixed-effect modeling approach. A parent-metabolite model with first-order absorption, 2-compartments for SV and 1-compartment for SVA, linear elimination, and enterohepatic circulation best described the data. The model was linked to the turnover pharmacodynamic model to describe the SVA inhibition on LDL-cholesterol production. Our simulations indicated that following RYGB surgery, the exposure to SV and SVA decreased by 40%. Consequently, for low-intensity statin patients, we recommend increasing the dose from 10 to 20 mg in post-RYGB patients to maintain a comparable response to that of non-operated subjects. Moderate-intensity statin patients should require increasing doses to 40 or 60 mg or the addition of a non-statin medication to achieve similar therapeutic outcomes. In conclusion, individuals post-RYGB exhibit diminished exposure to SV and may benefit from increasing the dose or adjunctive therapy with non-statin drugs to attain equivalent responses and mitigate potential adverse events.
Collapse
Affiliation(s)
- Ana Carolina Conchon Costa
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | - Wonho Kang
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Priscila A Yamamoto
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Cristiane M de Gaitani
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Mayrla E D Vasconcelos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Moreira Da Silva
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Rafael Kemp
- School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Ajith K Sankarankutty
- School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Wilson Salgado
- School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Jose Sebastiao Santos
- School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Natalia Valadares De Moraes
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| |
Collapse
|
2
|
Wenzel C, Lapczuk-Romanska J, Malinowski D, Ostrowski M, Drozdzik M, Oswald S. Comparative Intra-Subject Analysis of Gene Expression and Protein Abundance of Major and Minor Drug Metabolizing Enzymes in Healthy Human Jejunum and Liver. Clin Pharmacol Ther 2024; 115:221-230. [PMID: 37739780 DOI: 10.1002/cpt.3055] [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: 06/27/2023] [Accepted: 09/10/2023] [Indexed: 09/24/2023]
Abstract
First pass metabolism by phase I and phase II enzymes in the intestines and liver is a major determinant of the oral bioavailability of many drugs. Several studies analyzed expressions of major drug-metabolizing enzymes (DMEs), such as CYP3A4 and UGT1A1 in the human gut and liver. However, there is still a lack of knowledge regarding other DMEs (i.e., "minor" DMEs), although several clinically relevant drugs are affected by those enzymes. Moreover, there is very limited intra-subject data on hepatic and intestinal expression levels of minor DMEs. To fill this gap of knowledge, we analyzed gene expression (quantitative real-time polymerase chain reaction) and protein abundance (targeted proteomics) of 24 clinically relevant DMEs, that is, carboxylesterases (CES), UDP-glucuronosyltransferases (UGT), and cytochrome P450 (CYP)-enzymes. We performed our analysis using jejunum and liver tissue specimens from the same 11 healthy organ donors (8 men and 3 women, aged 19-60 years). Protein amounts of all investigated DMEs, with the exception of CYP4A11, were detected in human liver samples. CES2, CYP2C18, CYP3A4, and UGT2B17 protein abundance was similar or even higher in the jejunum, and all other DMEs were found in higher amounts in the liver. Significant correlations between gene expression and protein levels were observed only for 2 of 15 jejunal, but 13 of 23 hepatic DMEs. Intestinal and hepatic protein amounts only significantly correlated for CYP3A4 and UGT1A3. Our results demonstrated a notable variability between the individuals, which was even higher in the intestines than in the liver. Our intrasubject analysis of DMEs in the jejunum and liver from healthy donors, may be useful for physiologically-based pharmacokinetic-based modeling and prediction in order to improve efficacy and safety of oral drug therapy.
Collapse
Affiliation(s)
- Christoph Wenzel
- Department of Pharmacology, University Medicine Greifswald, Greifswald, Germany
| | - Joanna Lapczuk-Romanska
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Damian Malinowski
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Szczecin, Poland
| | - Marek Ostrowski
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Szczecin, Poland
- Department of General and Transplantation Surgery, Pomeranian Medical University, Szczecin, Poland
| | - Marek Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
3
|
Gouju J, Legeay S. Pharmacokinetics of obese adults: Not only an increase in weight. Biomed Pharmacother 2023; 166:115281. [PMID: 37573660 DOI: 10.1016/j.biopha.2023.115281] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/04/2023] [Indexed: 08/15/2023] Open
Abstract
Obesity is a pathophysiological state defined by a body mass index > 30 kg/m2 and characterized by an adipose tissue accumulation leading to an important weight increased. Several pathologies named comorbidities such as cardiovascular disease, type 2 diabetes and cancer make obesity the fifth cause of death in the world. Physiological changes impact the four main phases of pharmacokinetics of some drugs and leads to an inappropriate drug-dose. For absorption, the gastrointestinal transit is accelerated, and the gastric empty time is shortened, that can reduce the solubilization and absorption of some oral drugs. The drug distribution is probably the most impacted by the obesity-related changes because the fat mass (FM) increases at the expense of the lean body weight (LBW), leading to an important increase of the volume of distribution for lipophilic drugs and a low or moderately increase of this parameter for hydrophilic drugs. This modification of the distribution may require drug-dose adjustments. By various mechanisms, the metabolism and elimination of drugs are impacted by obesity and should be considered as similar or lower than that non-obese patients. To better understand the necessary drug-dose adjustments in obese patients, a narrative review of the literature was conducted to highlight the main elements to consider in the therapeutic management of adult obese patients.
Collapse
Affiliation(s)
- Julien Gouju
- MINT, INSERM U1066, CNRS 6021, UNIV Angers, SFR-ICAT 4208, IBS-CHU Angers, 4 rue Larrey, Angers 49933 Cedex 9, France; CHU Angers, 4 rue Larrey, Angers 49933 Cedex 9, France.
| | - Samuel Legeay
- MINT, INSERM U1066, CNRS 6021, UNIV Angers, SFR-ICAT 4208, IBS-CHU Angers, 4 rue Larrey, Angers 49933 Cedex 9, France
| |
Collapse
|
4
|
Obesity-related genomic instability and altered xenobiotic metabolism: possible consequences for cancer risk and chemotherapy. Expert Rev Mol Med 2022; 24:e28. [PMID: 35899852 PMCID: PMC9884759 DOI: 10.1017/erm.2022.22] [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] [Indexed: 11/06/2022]
Abstract
The increase in the prevalence of obesity has led to an elevated risk for several associated diseases including cancer. Several studies have investigated the DNA damage in human blood samples and showed a clear trend towards increased DNA damage in obesity. Reduced genomic stability is thus one of the consequences of obesity, which may contribute to the related cancer risk. Whether this is influenced by compromised DNA repair has not been elucidated sufficiently yet. On the other hand, obesity has also been linked to reduced therapy survival and increased adverse effects during chemotherapy, although the available data are controversial. Despite some indications that obesity might alter hepatic metabolism, current literature in humans is insufficient, and results from animal studies are inconclusive. Here we have summarised published data on hepatic drug metabolism to understand the impact of obesity on cancer therapy better. Furthermore, we highlight knowledge gaps in the interrelationship between obesity and drug metabolism from a toxicological perspective.
Collapse
|
5
|
Gabel F, Hovhannisyan V, Berkati AK, Goumon Y. Morphine-3-Glucuronide, Physiology and Behavior. Front Mol Neurosci 2022; 15:882443. [PMID: 35645730 PMCID: PMC9134088 DOI: 10.3389/fnmol.2022.882443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Morphine remains the gold standard painkiller available to date to relieve severe pain. Morphine metabolism leads to the production of two predominant metabolites, morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G). This metabolism involves uridine 5'-diphospho-glucuronosyltransferases (UGTs), which catalyze the addition of a glucuronide moiety onto the C3 or C6 position of morphine. Interestingly, M3G and M6G have been shown to be biologically active. On the one hand, M6G produces potent analgesia in rodents and humans. On the other hand, M3G provokes a state of strong excitation in rodents, characterized by thermal hyperalgesia and tactile allodynia. Its coadministration with morphine or M6G also reduces the resulting analgesia. Although these behavioral effects show quite consistency in rodents, M3G effects are much more debated in humans and the identity of the receptor(s) on which M3G acts remains unclear. Indeed, M3G has little affinity for mu opioid receptor (MOR) (on which morphine binds) and its effects are retained in the presence of naloxone or naltrexone, two non-selective MOR antagonists. Paradoxically, MOR seems to be essential to M3G effects. In contrast, several studies proposed that TLR4 could mediate M3G effects since this receptor also appears to be essential to M3G-induced hyperalgesia. This review summarizes M3G's behavioral effects and potential targets in the central nervous system, as well as the mechanisms by which it might oppose analgesia.
Collapse
Affiliation(s)
- Florian Gabel
- CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France
| | - Volodya Hovhannisyan
- CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France
| | - Abdel-Karim Berkati
- CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France
| | - Yannick Goumon
- CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France
- SMPMS, Mass Spectrometry Facilities of the CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| |
Collapse
|
6
|
Li J, Zhang Y, Yu M, Wang A, Qiu Y, Fan W, Hovgaard L, Yang M, Li Y, Wang R, Li X, Gan Y. The upregulated intestinal folate transporters direct the uptake of ligand-modified nanoparticles for enhanced oral insulin delivery. Acta Pharm Sin B 2022; 12:1460-1472. [PMID: 35530154 PMCID: PMC9072239 DOI: 10.1016/j.apsb.2021.07.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 11/29/2022] Open
Abstract
Transporters are traditionally considered to transport small molecules rather than large-sized nanoparticles due to their small pores. In this study, we demonstrate that the upregulated intestinal transporter (PCFT), which reaches a maximum of 12.3-fold expression in the intestinal epithelial cells of diabetic rats, mediates the uptake of the folic acid-grafted nanoparticles (FNP). Specifically, the upregulated PCFT could exert its function to mediate the endocytosis of FNP and efficiently stimulate the traverse of FNP across enterocytes by the lysosome-evading pathway, Golgi-targeting pathway and basolateral exocytosis, featuring a high oral insulin bioavailability of 14.4% in the diabetic rats. Conversely, in cells with relatively low PCFT expression, the positive surface charge contributes to the cellular uptake of FNP, and FNP are mainly degraded in the lysosomes. Overall, we emphasize that the upregulated intestinal transporters could direct the uptake of ligand-modified nanoparticles by mediating the endocytosis and intracellular trafficking of ligand-modified nanoparticles via the transporter-mediated pathway. This study may also theoretically provide insightful guidelines for the rational design of transporter-targeted nanoparticles to achieve efficient drug delivery in diverse diseases.
Collapse
Affiliation(s)
- Jingyi Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaqi Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miaorong Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Aohua Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Qiu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiwei Fan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lars Hovgaard
- Oral Formulation Development, Novo Nordisk A/S, Maalov 2760, Denmark
| | - Mingshi Yang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rui Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Corresponding authors. Tel.: +86 021 51322181, fax: +86 021 51322193 (Rui Wang); Tel.: +01 972 883 4480, fax: +01 972 883 4440 (Xiuying Li); Tel.: +86 021 20231975, fax: +86 021 20231000 1425 (Yong Gan).
| | - Xiuying Li
- University of Texas at Dallas, Richardson, TX 75080, USA
- Corresponding authors. Tel.: +86 021 51322181, fax: +86 021 51322193 (Rui Wang); Tel.: +01 972 883 4480, fax: +01 972 883 4440 (Xiuying Li); Tel.: +86 021 20231975, fax: +86 021 20231000 1425 (Yong Gan).
| | - Yong Gan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing 100050, China
- Corresponding authors. Tel.: +86 021 51322181, fax: +86 021 51322193 (Rui Wang); Tel.: +01 972 883 4480, fax: +01 972 883 4440 (Xiuying Li); Tel.: +86 021 20231975, fax: +86 021 20231000 1425 (Yong Gan).
| |
Collapse
|
7
|
Lappas NT, Lappas CM. Morphine. Forensic Toxicol 2022. [DOI: 10.1016/b978-0-12-819286-3.00023-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
8
|
Joshi H, Vastrad B, Joshi N, Vastrad C. Integrated bioinformatics analysis reveals novel key biomarkers in diabetic nephropathy. SAGE Open Med 2022; 10:20503121221137005. [PMID: 36385790 PMCID: PMC9661593 DOI: 10.1177/20503121221137005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives: The underlying molecular mechanisms of diabetic nephropathy have yet not been investigated clearly. In this investigation, we aimed to identify key genes involved in the pathogenesis and prognosis of diabetic nephropathy. Methods: We downloaded next-generation sequencing data set GSE142025 from Gene Expression Omnibus database having 28 diabetic nephropathy samples and nine normal control samples. The differentially expressed genes between diabetic nephropathy and normal control samples were analyzed. Biological function analysis of the differentially expressed genes was enriched by Gene Ontology and REACTOME pathways. Then, we established the protein–protein interaction network, modules, miRNA-differentially expressed gene regulatory network and transcription factor-differentially expressed gene regulatory network. Hub genes were validated by using receiver operating characteristic curve analysis. Results: A total of 549 differentially expressed genes were detected including 275 upregulated and 274 downregulated genes. The biological process analysis of functional enrichment showed that these differentially expressed genes were mainly enriched in cell activation, integral component of plasma membrane, lipid binding, and biological oxidations. Analyzing the protein–protein interaction network, miRNA-differentially expressed gene regulatory network and transcription factor-differentially expressed gene regulatory network, we screened hub genes MDFI, LCK, BTK, IRF4, PRKCB, EGR1, JUN, FOS, ALB, and NR4A1 by the Cytoscape software. The receiver operating characteristic curve analysis confirmed that hub genes were of diagnostic value. Conclusions: Taken above, using integrated bioinformatics analysis, we have identified key genes and pathways in diabetic nephropathy, which could improve our understanding of the cause and underlying molecular events, and these key genes and pathways might be therapeutic targets for diabetic nephropathy.
Collapse
Affiliation(s)
- Harish Joshi
- Endocrine and Diabetes Care Center, Hubbali, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, KLE Society’s College of Pharmacy, Gadag, India
| | - Nidhi Joshi
- Dr. D. Y. Patil Medical College, Kolhapur, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Dharwad, India
- Chanabasayya Vastrad, Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, India.
| |
Collapse
|
9
|
Chen W, Yang L, Yan C, Yao B, Lu J, Xu J, Liu G. Surface-Confined Building of Au@Pt-Centered and Multi-G-Quadruplex/Hemin Wire-Surrounded Electroactive Super-nanostructures for Ultrasensitive Monitoring of Morphine. ACS Sens 2020; 5:2644-2651. [PMID: 32633121 DOI: 10.1021/acssensors.0c01230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Overuse and abuse of morphine (MOP), one of the main components of pericarpium papaveris, have attracted increasing attention in the medical field owing to its pharmacological and toxicological activity. Herein, we proposed a new electrochemical nano-biosensor for MOP detection based on surface-confined building of Au@Pt-centered and multi-G-quadruplex/hemin wire-surrounded electroactive super-nanostructures. The center Au@Pt was flower-shaped and irregularly protruded, allowing substantial loading of multiple G-quadruplex wire/hemin complexes on its surface to accomplish the assembly of electroactive super-nanostructures. Interestingly, as the super-nanostructures were closely confined on the electrode surface, a significantly amplified electrochemical signal was thus obtained in the absence of MOP. In contrast, the introduction of target MOP can induce an intense competitive effect and strongly destroy the assembly process, resulting in the reduction of the electrochemical response that is correlated with the logarithmic concentration of MOP. Under optimal conditions, the electrochemical nano-biosensor is capable of highly sensitive detection of MOP in a dynamic concentration range from 1 ppt to 500 ppb. The limit of detection is achieved as low as 0.69 ppt, and the practical application was confirmed by examining MOP from chafing dish condiments. We expect the electrochemical platform utilizing this unique nanoarchitecture to provide rational guidelines to design high-performance analytical tools.
Collapse
Affiliation(s)
- Wei Chen
- Engineering Research Center of Bioprocess, MOE, School of Food & Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Lijun Yang
- Engineering Research Center of Bioprocess, MOE, School of Food & Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Chao Yan
- Engineering Research Center of Bioprocess, MOE, School of Food & Biological Engineering, Hefei University of Technology, Hefei 230009, China
- Research Center for Biomedical and Health Science, School of Life and Health, Anhui Science & Technology University, Fengyang 233100, China
| | - Bangben Yao
- Engineering Research Center of Bioprocess, MOE, School of Food & Biological Engineering, Hefei University of Technology, Hefei 230009, China
- Anhui Province Institute of Product Quality Supervision & Inspection, Hefei 230051, P. R. China
| | - Jianfeng Lu
- Engineering Research Center of Bioprocess, MOE, School of Food & Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Jianguo Xu
- Engineering Research Center of Bioprocess, MOE, School of Food & Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Guodong Liu
- Research Center for Biomedical and Health Science, School of Life and Health, Anhui Science & Technology University, Fengyang 233100, China
| |
Collapse
|
10
|
Drozdzik M, Czekawy I, Oswald S, Drozdzik A. Intestinal drug transporters in pathological states: an overview. Pharmacol Rep 2020; 72:1173-1194. [PMID: 32715435 PMCID: PMC7550293 DOI: 10.1007/s43440-020-00139-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Marek Drozdzik
- Department of Pharmacology, Pomeranian Medical University, Powstancow Wlkp 72, 70-111, Szczecin, Poland.
| | - Izabela Czekawy
- Department of Pharmacology, Pomeranian Medical University, Powstancow Wlkp 72, 70-111, Szczecin, Poland
| | - Stefan Oswald
- Department of Pharmacology, Medicine University Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489, Greifswald, Germany.,Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18051, Rostock, Germany
| | - Agnieszka Drozdzik
- Department of Integrated Dentistry, Pomeranian Medical University, Powstancow Wlkp 72, 70-111, Szczecin, Poland
| |
Collapse
|
11
|
Murakami T, Bodor E, Bodor N. Modulation of expression/function of intestinal P-glycoprotein under disease states. Expert Opin Drug Metab Toxicol 2019; 16:59-78. [DOI: 10.1080/17425255.2020.1701653] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Nicholas Bodor
- Bodor Laboratories, Miami, FL, USA
- College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
12
|
Gómez-Silva M, Piñeyro-Garza E, Vargas-Zapata R, Gamino-Peña ME, León-García A, de León MB, Llerena A, León-Cachón RBR. Pharmacogenetics of amfepramone in healthy Mexican subjects reveals potential markers for tailoring pharmacotherapy of obesity: results of a randomised trial. Sci Rep 2019; 9:17833. [PMID: 31780765 PMCID: PMC6882847 DOI: 10.1038/s41598-019-54436-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022] Open
Abstract
Amfepramone (AFP) is an appetite-suppressant drug used in the treatment of obesity. Nonetheless, studies on interindividual pharmacokinetic variability and its association with genetic variants are limited. We employed a pharmacokinetic and pharmacogenetic approach to determine possible metabolic phenotypes of AFP and identify genetic markers that could affect the pharmacokinetic variability in a Mexican population. A controlled, randomized, crossover, single-blind, two-treatment, two-period, and two sequence clinical study of AFP (a single 75 mg dose) was conducted in 36 healthy Mexican volunteers who fulfilled the study requirements. Amfepramone plasma levels were measured using high-performance liquid chromatography mass spectrometry. Genotyping was performed using real-time PCR with TaqMan probes. Four AFP metabolizer phenotypes were found in our population: slow, normal, intermediate, and fast. Additionally, two gene polymorphisms, ABCB1-rs1045642 and CYP3A4-rs2242480, had a significant effect on AFP pharmacokinetics (P < 0.05) and were the predictor factors in a log-linear regression model. The ABCB1 and CYP3A4 gene polymorphisms were associated with a fast metabolizer phenotype. These results suggest that metabolism of AFP in the Mexican population is variable. In addition, the genetic variants ABCB1-rs1045642 and CYP3A4-rs2242480 may partially explain the AFP pharmacokinetic variability.
Collapse
Affiliation(s)
- Magdalena Gómez-Silva
- Forensic Medicine Service, School of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo Leon, Mexico.,Analytical Department of the Research Institute for Clinical and Experimental Pharmacology, Ipharma S.A, Monterrey, Nuevo Leon, Mexico
| | - Everardo Piñeyro-Garza
- Clinical Department of the Research Institute for Clinical and Experimental Pharmacology, Ipharma S.A, Monterrey, Nuevo Leon, Mexico
| | - Rigoberto Vargas-Zapata
- Quality Assurance Department of the Research Institute for Clinical and Experimental Pharmacology, Ipharma S.A, Monterrey, Nuevo Leon, Mexico
| | - María Elena Gamino-Peña
- Statistical Department of the Research Institute for Clinical and Experimental Pharmacology, Ipharma S.A, Monterrey, Nuevo Leon, Mexico
| | | | - Mario Bermúdez de León
- Department of Molecular Biology, Center for Biomedical Research of the Northeast, Mexican Institute of Social Security, Monterrey, Nuevo Leon, Mexico
| | - Adrián Llerena
- Clinical Research Center of Health Area, Hospital and Medical School of Extremadura University, Badajoz, Spain
| | - Rafael B R León-Cachón
- Center of Molecular Diagnostics and Personalized Medicine, Department of Basic Sciences, Division of Health Sciences, University of Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico.
| |
Collapse
|
13
|
Al-Ali AAA, Nielsen RB, Steffansen B, Holm R, Nielsen CU. Nonionic surfactants modulate the transport activity of ATP-binding cassette (ABC) transporters and solute carriers (SLC): Relevance to oral drug absorption. Int J Pharm 2019; 566:410-433. [DOI: 10.1016/j.ijpharm.2019.05.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/10/2019] [Accepted: 05/11/2019] [Indexed: 01/11/2023]
|
14
|
de Hoogd S, Välitalo PAJ, Dahan A, van Kralingen S, Coughtrie MMW, van Dongen EPA, van Ramshorst B, Knibbe CAJ. Influence of Morbid Obesity on the Pharmacokinetics of Morphine, Morphine-3-Glucuronide, and Morphine-6-Glucuronide. Clin Pharmacokinet 2018; 56:1577-1587. [PMID: 28510797 PMCID: PMC5694499 DOI: 10.1007/s40262-017-0544-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction Obesity is associated with many pathophysiological changes that may result in altered drug metabolism. The aim of this study is to investigate the influence of obesity on the pharmacokinetics of morphine, morphine-3-glucuronide (M3G), and morphine-6-glucuronide (M6G) through a combined analysis in morbidly obese patients and non-obese healthy volunteers. Methods In this analysis, data from 20 morbidly obese patients [mean body mass index 49.9 kg/m2 (range 37.6–78.6 kg/m2) and weight 151.3 kg (range 112–251.9 kg)] and 20 healthy volunteers [mean weight 70.6 kg (range 58–85 kg)] were included. Morbidly obese patients received 10 mg of intravenous (I.V.) morphine after gastric bypass surgery, with additional morphine I.V. doses as needed. Healthy volunteers received an I.V. bolus of morphine of 0.1 mg/kg followed by an infusion of 0.030 mg kg−1 h−1 for 1 h. Population pharmacokinetic modeling was performed using NONMEM 7.2. Results In morbidly obese patients, elimination clearance of M3G and M6G was decreased substantially compared with healthy volunteers (p < 0.001). Regarding glucuronidation, only a slight decrease in the formation of M6G and a delay in the formation of M3G was found (both p < 0.001). Obesity was also identified as a covariate for the peripheral volume of distribution of morphine (p < 0.001). Conclusion Metabolism of morphine is not altered in morbidly obese patients. However, decreased elimination of both M3G and M6G is evident, resulting in a substantial increase in exposure to these two metabolites. A rational explanation of this finding is that it results from alterations in membrane transporter function and/or expression in the liver. ClinicalTrials.gov identifier: NCT01097148. Electronic supplementary material The online version of this article (doi:10.1007/s40262-017-0544-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sjoerd de Hoogd
- Department of Clinical Pharmacy, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands
| | - Pyry A. J. Välitalo
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Albert Dahan
- Department of Anaesthesiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Simone van Kralingen
- Department of Anaesthesiology and Intensive Care, St. Antonius Hospital, Nieuwegein, The Netherlands
- Department of Anaesthesiology, OLVG, Amsterdam, The Netherlands
| | | | - Eric P. A. van Dongen
- Department of Anaesthesiology and Intensive Care, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Bert van Ramshorst
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Catherijne A. J. Knibbe
- Department of Clinical Pharmacy, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
15
|
Huang CS, Chen HW, Lin TY, Lin AH, Lii CK. Shikonin upregulates the expression of drug-metabolizing enzymes and drug transporters in primary rat hepatocytes. JOURNAL OF ETHNOPHARMACOLOGY 2018; 216:18-25. [PMID: 29414119 DOI: 10.1016/j.jep.2018.01.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shikonin, a naphthoquinone pigment abundant in the root of the Chinese herb Lithospermum erythrorhizon, has been widely used to treat inflammatory diseases for thousands of years. Whether shikonin changes drug metabolism remains unclear. AIM OF THE STUDY We investigated whether shikonin modulates the expression of hepatic drug-metabolizing enzymes and transporters as well as the possible mechanisms of this action. MATERIALS AND METHODS Primary hepatocytes isolated from Sprague-Dawley rats were treated with 0-2 μM shikonin and the protein and mRNA levels of drug-metabolizing enzymes and transporters as well as the activation of aryl hydrocarbon receptor (AhR) and NF-E2-related factor 2 (Nrf2) were determined. RESULTS Shikonin dose-dependently increased the protein and RNA expression of phase I enzymes, i.e., cytochrome P450 (CYP) 1A1/2, CYP3A2, CYP2D1, and CYP2C6; phase II enzymes, i.e., glutathione S-transferase (GST), NADP(H) quinone oxidoreductase 1 (NQO1), and UDP glucuronosyltransferase 1A1; and phase III drug transporters, i.e., P-glycoprotein, multidrug resistance-associated protein 2/3, organic anion transporting polypeptide (OATP) 1B1, and OATP2B1. Immunoblot analysis and EMSA revealed that shikonin increased AhR and Nrf2 nuclear contents and DNA binding activity. AhR and Nrf2 knockdown by siRNA attenuated the ability of shikonin to induce drug-metabolizing enzyme expression. In addition, shikonin increased p38, JNK, and ERK1/2 phosphorylation, and inhibitors of the respective kinases inhibited shikonin-induced Nrf2 nuclear translocation. CONCLUSIONS Shikonin effectively upregulates the transcription of CYP isozymes, phase II detoxification enzymes, and phase III membrane transporters and this function is at least partially through activation of AhR and Nrf2. Moreover, Nrf2 activation is dependent on mitogen-activated protein kinases.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Basic Helix-Loop-Helix Transcription Factors/agonists
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Biotransformation
- Cells, Cultured
- Dose-Response Relationship, Drug
- Drugs, Chinese Herbal/pharmacology
- Extracellular Signal-Regulated MAP Kinases
- Gene Expression Regulation, Enzymologic/drug effects
- Hepatocytes/drug effects
- Hepatocytes/enzymology
- JNK Mitogen-Activated Protein Kinases
- Male
- Membrane Transport Proteins/drug effects
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/metabolism
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/metabolism
- Naphthoquinones/pharmacology
- Phosphorylation
- Primary Cell Culture
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptors, Aryl Hydrocarbon/agonists
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Transcriptional Activation/drug effects
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Chin-Shiu Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, No. 91, Hsueh-Shih Road, Taichung 404, Taiwan
| | - Tzu-Yu Lin
- Department of Nutrition, China Medical University, No. 91, Hsueh-Shih Road, Taichung 404, Taiwan
| | - Ai-Hsuan Lin
- Department of Nutrition, China Medical University, No. 91, Hsueh-Shih Road, Taichung 404, Taiwan
| | - Chong-Kuei Lii
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan; Department of Nutrition, China Medical University, No. 91, Hsueh-Shih Road, Taichung 404, Taiwan.
| |
Collapse
|
16
|
Akazawa T, Uchida Y, Miyauchi E, Tachikawa M, Ohtsuki S, Terasaki T. High Expression of UGT1A1/1A6 in Monkey Small Intestine: Comparison of Protein Expression Levels of Cytochromes P450, UDP-Glucuronosyltransferases, and Transporters in Small Intestine of Cynomolgus Monkey and Human. Mol Pharm 2017; 15:127-140. [DOI: 10.1021/acs.molpharmaceut.7b00772] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Takanori Akazawa
- Division
of Membrane Transport and Drug Targeting, Graduate School
of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Yasuo Uchida
- Division
of Membrane Transport and Drug Targeting, Graduate School
of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Eisuke Miyauchi
- Division
of Membrane Transport and Drug Targeting, Graduate School
of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Masanori Tachikawa
- Division
of Membrane Transport and Drug Targeting, Graduate School
of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Sumio Ohtsuki
- Department
of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Tetsuya Terasaki
- Division
of Membrane Transport and Drug Targeting, Graduate School
of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
17
|
Hachon L, Reis R, Labat L, Poitou C, Jacob A, Declèves X, Lloret-Linares C. Morphine and metabolites plasma levels after administration of sustained release morphine in Roux-en-Y gastric bypass subjects versus matched control subjects. Surg Obes Relat Dis 2017; 13:1869-1874. [PMID: 28864105 DOI: 10.1016/j.soard.2017.07.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/18/2017] [Accepted: 07/25/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Better knowledge of opioid pharmacology after Roux-en-Y gastric bypass (RYGB) is required for optimizing their use in this growing population. OBJECTIVE The aim of this case-controlled pharmacokinetic (PK) study was to compare morphine and its glucuronidated metabolites (morphine-3-glucuronide and morphine-6-glucuronide) plasma PKs between patients with RYGB and their controls. SETTINGS University hospital, Lariboisière Hospital, Paris. METHODS Thirty milligrams of morphine as a sustained-release formulation was orally administered in 12 women who had undergone RYGB for at least 2 years (RYGB group) and in their nonsurgical controls matched for sex, body mass index (±2 points), and age (±5 yr). Morphine, morphine-3-glucuronide, and morphine-6-glucuronide plasma concentrations over a 12-hour period were determined by a validated method using liquid chromatography mass spectrometry in tandem. Drowsiness, respiratory rate, and oxygen saturation were monitored during the PK visit. RESULTS Morphine oral area under the curve (for time 0-12 hr; 115.8 ± 108.0 nmol.hr/L and 86.9 ± 38.8 nmol.hr/L for RYGB group and control group, respectively, P = .71), morphine at maximal concentration, metabolites oral area under the curve (for time 0-12 hr), and other PK parameters were similar between groups. After drug administration, mean drowsiness was superior in RYGB group. Mean respiratory rate and oxygen saturation were similar in both groups. CONCLUSION No dose adjustment seems to be needed for sustained release morphine when prescribed to RYGB patients.
Collapse
Affiliation(s)
- Lorry Hachon
- Inserm U1144, Paris, France; Therapeutic Research Unit, Department of Internal Medicine, Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Paris, France
| | - Rafael Reis
- Inserm U1144, Paris, France; Biologie du médicament et Toxicologie, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Laurence Labat
- Inserm U1144, Paris, France; Biologie du médicament et Toxicologie, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Paris, France; Université Paris Descartes, UMR-S 1144, Paris, France
| | - Christine Poitou
- Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Service de Nutrition, Université Pierre et Marie Curie, Institut cardiométabolisme et nutrition (ICAN), Paris, France
| | - Aude Jacob
- Inserm U1144, Paris, France; Université Paris Descartes, UMR-S 1144, Paris, France
| | - Xavier Declèves
- Inserm U1144, Paris, France; Biologie du médicament et Toxicologie, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Paris, France; Université Paris Descartes, UMR-S 1144, Paris, France
| | - Celia Lloret-Linares
- Inserm U1144, Paris, France; Therapeutic Research Unit, Department of Internal Medicine, Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Paris, France; Université Paris Diderot, UMR-S 1144, Paris, France.
| |
Collapse
|
18
|
Lloret-Linares C, Luo H, Rouquette A, Labat L, Poitou C, Tordjman J, Bouillot JL, Mouly S, Scherrmann JM, Bergmann JF, Declèves X. The effect of morbid obesity on morphine glucuronidation. Pharmacol Res 2017; 118:64-70. [DOI: 10.1016/j.phrs.2016.08.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/29/2016] [Accepted: 08/29/2016] [Indexed: 01/28/2023]
|
19
|
Lloret-Linares C. Pharmacokinetic considerations for patients with a history of bariatric surgery. Expert Opin Drug Metab Toxicol 2017; 13:493-496. [PMID: 28151695 DOI: 10.1080/17425255.2017.1290796] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Célia Lloret-Linares
- a Inserm, U1144 , Paris , F-75006 , France.,b Université Paris Diderot, UMR-S 1144 , Paris , F-75013 , France.,c Internal Medicine Department , Lariboisière Hospital , Paris , France
| |
Collapse
|
20
|
Zhang J, Qi L, Zheng WT, Tian YL, Chi AP, Zhang ZQ. Novel functionalized poly(glycidyl methacrylate-co-ethylene dimethacrylate) microspheres for the solid-phase extraction of glycopeptides/glycoproteins. J Sep Sci 2017; 40:1107-1114. [DOI: 10.1002/jssc.201600780] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/14/2016] [Accepted: 12/06/2016] [Indexed: 01/14/2023]
Affiliation(s)
- Jing Zhang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province; School of Chemistry and Chemical engineering; Shaanxi Normal University; Xi'an China
- Institute of Sports Biology; School of Physical Education; Shaanxi Normal University; Xi'an China
| | - Liang Qi
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province; School of Chemistry and Chemical engineering; Shaanxi Normal University; Xi'an China
| | - Wei-Ting Zheng
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province; School of Chemistry and Chemical engineering; Shaanxi Normal University; Xi'an China
| | - Yong-Le Tian
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province; School of Chemistry and Chemical engineering; Shaanxi Normal University; Xi'an China
| | - Ai-Ping Chi
- Institute of Sports Biology; School of Physical Education; Shaanxi Normal University; Xi'an China
| | - Zhi-Qi Zhang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province; School of Chemistry and Chemical engineering; Shaanxi Normal University; Xi'an China
| |
Collapse
|
21
|
Miyauchi E, Tachikawa M, Declèves X, Uchida Y, Bouillot JL, Poitou C, Oppert JM, Mouly S, Bergmann JF, Terasaki T, Scherrmann JM, Lloret-Linares C. Quantitative Atlas of Cytochrome P450, UDP-Glucuronosyltransferase, and Transporter Proteins in Jejunum of Morbidly Obese Subjects. Mol Pharm 2016; 13:2631-40. [DOI: 10.1021/acs.molpharmaceut.6b00085] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Eisuke Miyauchi
- Membrane Transport and Drug Targeting Laboratory,
Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masanori Tachikawa
- Membrane Transport and Drug Targeting Laboratory,
Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Xavier Declèves
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
- Pharmacokinetics and Pharmacochemistry Unit, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris F-75014, France
| | - Yasuo Uchida
- Membrane Transport and Drug Targeting Laboratory,
Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Jean-Luc Bouillot
- Department of Surgery, Université
Versailles Saint Quentin, Hôpital Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Boulogne 92100, France
| | - Christine Poitou
- Institut cardiométabolisme et nutrition
(ICAN), Université Pierre et Marie Curie, Service de Nutrition,
Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris F-75013, France
| | - Jean-Michel Oppert
- Institut cardiométabolisme et nutrition
(ICAN), Université Pierre et Marie Curie, Service de Nutrition,
Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris F-75013, France
| | - Stéphane Mouly
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
- Department of Internal Medicine, Therapeutic Research
Unit, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris F-75010, France
| | - Jean-François Bergmann
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
- Department of Internal Medicine, Therapeutic Research
Unit, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris F-75010, France
| | - Tetsuya Terasaki
- Membrane Transport and Drug Targeting Laboratory,
Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Jean-Michel Scherrmann
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
| | - Célia Lloret-Linares
- Inserm, UMR-S 1144 Université Paris Descartes-Paris Diderot, Variabilité de réponse aux psychotropes, Paris F-75010, France
- Department of Internal Medicine, Therapeutic Research
Unit, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris F-75010, France
| |
Collapse
|