201
|
Prediction of drug-induced nephrotoxicity and injury mechanisms with human induced pluripotent stem cell-derived cells and machine learning methods. Sci Rep 2015. [PMID: 26212763 PMCID: PMC4515747 DOI: 10.1038/srep12337] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The renal proximal tubule is a main target for drug-induced toxicity. The prediction of proximal tubular toxicity during drug development remains difficult. Any in vitro methods based on induced pluripotent stem cell-derived renal cells had not been developed, so far. Here, we developed a rapid 1-step protocol for the differentiation of human induced pluripotent stem cells (hiPSC) into proximal tubular-like cells. These proximal tubular-like cells had a purity of >90% after 8 days of differentiation and could be directly applied for compound screening. The nephrotoxicity prediction performance of the cells was determined by evaluating their responses to 30 compounds. The results were automatically determined using a machine learning algorithm called random forest. In this way, proximal tubular toxicity in humans could be predicted with 99.8% training accuracy and 87.0% test accuracy. Further, we studied the underlying mechanisms of injury and drug-induced cellular pathways in these hiPSC-derived renal cells, and the results were in agreement with human and animal data. Our methods will enable the development of personalized or disease-specific hiPSC-based renal in vitro models for compound screening and nephrotoxicity prediction.
Collapse
|
202
|
Babelova A, Burckhardt BC, Salinas-Riester G, Pommerenke C, Burckhardt G, Henjakovic M. Next generation sequencing of sex-specific genes in the livers of obese ZSF1 rats. Genomics 2015. [PMID: 26200819 DOI: 10.1016/j.ygeno.2015.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Type 2 diabetes induces pathophysiological changes in the liver. The aim of this study was to identify differently expressed genes in the livers of male and female ZSF1 rats (ZDFxSHHF-hybrid, generation F1), a model for type 2 diabetes. Gene expression was investigated using next-generation sequencing (NGS). Selected candidate genes were verified by real-time PCR in the livers of obese and lean rats. 103 sex-different genes, associated to pathways "response to chemical stimulus", "lipid metabolism", and "response to organic substance", were identified. Male-specific genes were involved in hepatic metabolism, detoxification, and secretion, e.g. cytochrome P450 2c11 (Cyp2c11), Cyp4a2, glutathione S-transferases mu 2 (Gstm2), and Slc22a8 (organic anion transporter 3, Oat3). Most female-specific genes were associated to lipid metabolism (e.g. glycerol-3-phosphate acyltransferase 1, Gpam) or glycolysis (e.g. glucokinase, Gck). Our data suggest the necessity to pay attention to sex- and diabetes-dependent changes in pre-clinical testing of hepatic metabolized and secreted drugs.
Collapse
Affiliation(s)
- Andrea Babelova
- Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 83391 Bratislava, Slovak Republic
| | - Birgitta C Burckhardt
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Goettingen, 37073 Goettingen, Germany
| | - Gabriela Salinas-Riester
- Department of Developmental Biochemistry, DNA Microarray and Deep-Sequencing Facility, University Medical Center Goettingen, 37077 Goettingen, Germany
| | - Claudia Pommerenke
- Department of Developmental Biochemistry, DNA Microarray and Deep-Sequencing Facility, University Medical Center Goettingen, 37077 Goettingen, Germany
| | - Gerhard Burckhardt
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Goettingen, 37073 Goettingen, Germany
| | - Maja Henjakovic
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Goettingen, 37073 Goettingen, Germany.
| |
Collapse
|
203
|
Walsh DR, Nolin TD, Friedman PA. Drug Transporters and Na+/H+ Exchange Regulatory Factor PSD-95/Drosophila Discs Large/ZO-1 Proteins. Pharmacol Rev 2015; 67:656-80. [PMID: 26092975 PMCID: PMC4485015 DOI: 10.1124/pr.115.010728] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Drug transporters govern the absorption, distribution, and elimination of pharmacologically active compounds. Members of the solute carrier and ATP binding-cassette drug transporter family mediate cellular drug uptake and efflux processes, thereby coordinating the vectorial movement of drugs across epithelial barriers. To exert their physiologic and pharmacological function in polarized epithelia, drug transporters must be targeted and stabilized to appropriate regions of the cell membrane (i.e., apical versus basolateral). Despite the critical importance of drug transporter membrane targeting, the mechanisms that underlie these processes are largely unknown. Several clinically significant drug transporters possess a recognition sequence that binds to PSD-95/Drosophila discs large/ZO-1 (PDZ) proteins. PDZ proteins, such as the Na(+)/H(+) exchanger regulatory factor (NHERF) family, act to stabilize and organize membrane targeting of multiple transmembrane proteins, including many clinically relevant drug transporters. These PDZ proteins are normally abundant at apical membranes, where they tether membrane-delimited transporters. NHERF expression is particularly high at the apical membrane in polarized tissue such as intestinal, hepatic, and renal epithelia, tissues important to drug disposition. Several recent studies have highlighted NHERF proteins as determinants of drug transporter function secondary to their role in controlling membrane abundance and localization. Mounting evidence strongly suggests that NHERF proteins may have clinically significant roles in pharmacokinetics and pharmacodynamics of several pharmacologically active compounds and may affect drug action in cancer and chronic kidney disease. For these reasons, NHERF proteins represent a novel class of post-translational mediators of drug transport and novel targets for new drug development.
Collapse
Affiliation(s)
- Dustin R Walsh
- Laboratory for G Protein-Coupled Receptor Biology, Department of Pharmacology and Chemical Biology, and Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (P.A.F.); and Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (D.R.W., T.D.N.)
| | - Thomas D Nolin
- Laboratory for G Protein-Coupled Receptor Biology, Department of Pharmacology and Chemical Biology, and Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (P.A.F.); and Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (D.R.W., T.D.N.)
| | - Peter A Friedman
- Laboratory for G Protein-Coupled Receptor Biology, Department of Pharmacology and Chemical Biology, and Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (P.A.F.); and Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (D.R.W., T.D.N.)
| |
Collapse
|
204
|
Tomi M, Eguchi H, Ozaki M, Tawara T, Nishimura S, Higuchi K, Maruyama T, Nishimura T, Nakashima E. Role of OAT4 in Uptake of Estriol Precursor 16α-Hydroxydehydroepiandrosterone Sulfate Into Human Placental Syncytiotrophoblasts From Fetus. Endocrinology 2015; 156:2704-12. [PMID: 25919187 DOI: 10.1210/en.2015-1130] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estriol biosynthesis in human placenta requires the uptake of a fetal liver-derived estriol precursor, 16α-hydroxydehydroepiandrosterone sulfate (16α-OH DHEAS), by placental syncytiotrophoblasts at their basal plasma membrane (BM), which faces the fetal circulation. The aim of this work is to identify the transporter(s) mediating 16α-OH DHEAS uptake at the fetal side of syncytiotrophoblasts by using human placental BM-enriched vesicles and to examine the contribution of the putative transporter to estriol synthesis at the cellular level, using choriocarcinoma JEG-3 cells. Organic anion transporter (OAT)-4 and organic anion transporting polypeptide 2B1 proteins were enriched in human placental BM vesicles compared with crude membrane fraction. Uptake of [(3)H]16α-OH DHEAS by BM vesicles was partially inhibited in the absence of sodium but was significantly increased in the absence of chloride and after preloading glutarate. Uptake of [(3)H]16α-OH DHEAS by BM vesicles was significantly inhibited by OAT4 substrates such as dehydroepiandrosterone sulfate, estrone-3-sulfate, and bromosulfophthalein but not by cyclosporin A, tetraethylammonium, p-aminohippuric acid, or cimetidine. These characteristics of vesicular [(3)H]16α-OH DHEAS uptake are in good agreement with those of human OAT4-transfected COS-7 cells as well as forskolin-differentiated JEG-3 cells. Estriol secretion from differentiated JEG-3 cells was detected when the cells were incubated with 16α-OH DHEAS for 8 hours but was inhibited in the presence of 50 μM bromosulfophthalein. Our results indicate that OAT4 at the BM of human placental syncytiotrophoblasts plays a predominant role in the uptake of 16α-OH DHEAS for placental estriol synthesis.
Collapse
Affiliation(s)
- Masatoshi Tomi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Hiromi Eguchi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Mayuko Ozaki
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tomohiro Tawara
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Sachika Nishimura
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Kei Higuchi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tetsuo Maruyama
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tomohiro Nishimura
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Emi Nakashima
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| |
Collapse
|
205
|
Fardel O, Le Vee M, Jouan E, Denizot C, Parmentier Y. Nature and uses of fluorescent dyes for drug transporter studies. Expert Opin Drug Metab Toxicol 2015; 11:1233-51. [PMID: 26050735 DOI: 10.1517/17425255.2015.1053462] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Drug transporters are now recognized as major players involved in pharmacokinetics and toxicology. Methods for assessing their activity are important to consider, particularly owing to regulatory requirements with respect to inhibition of drug transporter activity and prediction of drug-drug interactions. In this context, the use of fluorescent-dye-based transport assays is likely to deserve attention. AREAS COVERED This review provides an overview of the nature of fluorescent dye substrates for ATP-binding cassette and solute carrier drug transporters. Their use for investigating drug transporter activity in cultured cells and clinical hematological samples, drug transporter inhibition, drug transporter imaging and drug transport at the organ level are summarized. EXPERT OPINION A wide range of fluorescent dyes is now available for use in various aspects of drug transporter studies. The use of these dyes for transporter analyses may, however, be hampered by classic pitfalls of fluorescence technology, such as quenching. Transporter-independent processes such as passive diffusion of dyes through plasma membrane or dye sequestration into subcellular compartments must also be considered, as well as the redundant handling by various distinct transporters of some fluorescent probes. Finally, standardization of dye-based transport assays remains an important on-going issue.
Collapse
Affiliation(s)
- Olivier Fardel
- Institut de Recherches en Santé, Environnement et Travail (IRSET) , UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes , France
| | | | | | | | | |
Collapse
|
206
|
Sassier M, Dugué AE, Clarisse B, Lesueur P, Avrillon V, Bizieux-Thaminy A, Auliac JB, Kaluzinski L, Tillon J, Robinet G, Le Caer H, Monnet I, Madroszyk A, Boza G, Falchero L, Fournel P, Egenod T, Toffart AC, Leiber N, Do P, Gervais R. Renal insufficiency is the leading cause of double maintenance (bevacizumab and pemetrexed) discontinuation for toxicity to advanced non-small cell lung cancer in real world setting. Lung Cancer 2015; 89:161-6. [PMID: 26037036 DOI: 10.1016/j.lungcan.2015.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/06/2015] [Accepted: 05/07/2015] [Indexed: 01/18/2023]
Abstract
OBJECTIVES In advanced non-small cell lung cancer (NSCLC), maintenance therapy has emerged as a novel therapeutic reference for patients with non-progressive disease after platinum-based induction chemotherapy. However, the use of double maintenance (DM) with pemetrexed and bevacizumab is still being evaluated in terms of its clinical benefits and safety profile. The objective of this retrospective study was to describe the reasons for DM discontinuation in a real-world setting. MATERIALS AND METHODS Patients with advanced non-squamous NSCLC were eligible if they had received at least 4 cycles of induction chemotherapy, followed by at least 1 cycle of DM. They were identified by using the oncology pharmacy database of 17 French centers. RESULTS Eighty-one patients who began a DM after induction chemotherapy were identified from September 2009 to April 2013. Among the 78 patients who had stopped DM at the time of the analysis, the main reasons for discontinuation were disease progression (42%), adverse events (33%), and personal preference (8%). The most frequent toxicity responsible for DM discontinuation was renal insufficiency (54%). CONCLUSION For patients with advanced NSCLC eligible for DM therapy, a particular attention should be paid to potential renal failure. Kidney function should be monitored carefully before and during DM to detect and manage early this adverse event.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Laure Kaluzinski
- Centre Hospitalier Public du Cotentin, Cherbourg-Octeville F-50100, France
| | | | - Gilles Robinet
- Centre Hospitalier Régional Universitaire, Brest F-29200, France
| | | | | | | | | | | | - Pierre Fournel
- Institut de Cancérologie Lucien Neuwirth, Saint-Priest en Jarez F-42270, France
| | - Thomas Egenod
- Centre Hospitalier Universitaire, Limoges F-87000, France
| | | | | | - Pascal Do
- Centre François Baclesse, Caen F-14000, France
| | | |
Collapse
|
207
|
Endocytotic uptake of zoledronic acid by tubular cells may explain its renal effects in cancer patients receiving high doses of the compound. PLoS One 2015; 10:e0121861. [PMID: 25756736 PMCID: PMC4355483 DOI: 10.1371/journal.pone.0121861] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 02/16/2015] [Indexed: 12/22/2022] Open
Abstract
Zoledronic acid, a highly potent nitrogen-containing bisphosphonate used for the treatment of pathological bone loss, is excreted unmetabolized via the kidney if not bound to the bone. In cancer patients receiving high doses of the compound renal excretion may be associated with acute tubular necrosis. The question of how zoledronic acid is internalized by renal tubular cells has not been answered until now. In the current work, using a primary human tubular cell culture system, the pathway of cellular uptake of zoledronic acid (fluorescently/radiolabeled) and its cytotoxicity were investigated. Previous studies in our laboratory have shown that this primary cell culture model consistently mimics the physiological characteristics of molecular uptake/transport of the epithelium in vivo. Zoledronic acid was found to be taken up by tubular cells via fluid-phase-endocytosis (from apical and basolateral side) as evidenced by its co-localization with dextran. Cellular uptake and the resulting intracellular level was twice as high from the apical side compared to the basolateral side. Furthermore, the intracellular zoledronic acid level was found to be dependent on the administered concentration and not saturable. Cytotoxic effects however, were only seen at higher administration doses and/or after longer incubation times. Although zoledronic acid is taken up by tubular cells, no net tubular transport could be measured. It is concluded that fluid-phase-endocytosis of zoledronic acid and cellular accumulation at high doses may be responsible for the acute tubular necrosis observed in some cancer patients receiving high doses of the compound.
Collapse
|
208
|
Probert PME, Meyer SK, Alsaeedi F, Axon AA, Fairhall EA, Wallace K, Charles M, Oakley F, Jowsey PA, Blain PG, Wright MC. An expandable donor-free supply of functional hepatocytes for toxicology. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00214h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Abstract
The B-13 cell is a readily expandable rat pancreatic acinar-like cell that differentiates on simple plastic culture substrata into replicatively-senescent hepatocyte-like (B-13/H) cells in response to glucocorticoid exposure. B-13/H cells express a variety of liver-enriched and liver-specific genes, many at levels similar to hepatocytes in vivo. Furthermore, the B-13/H phenotype is maintained for at least several weeks in vitro, in contrast to normal hepatocytes which rapidly de-differentiate under the same simple – or even under more complex – culture conditions. The origin of the B-13 cell line and the current state of knowledge regarding differentiation to B-13/H cells are presented, followed by a review of recent advances in the use of B-13/H cells in a variety of toxicity endpoints. B-13 cells therefore offer Toxicologists a cost-effective and easy to use system to study a range of toxicologically-related questions. Dissecting the mechanism(s) regulating the formation of B-13/H cell may also increase the likelihood of engineering a human equivalent, providing Toxicologists with an expandable donor-free supply of functional rat and human hepatocytes, invaluable additions to the tool kit of in vitro toxicity tests.
Collapse
Affiliation(s)
- Philip M. E. Probert
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Stephanie K. Meyer
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Fouzeyyah Alsaeedi
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Andrew A. Axon
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Emma A. Fairhall
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Karen Wallace
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Michelle Charles
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Fiona Oakley
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Paul A. Jowsey
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Peter G. Blain
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Matthew C. Wright
- Institute Cellular Medicine, Level 4 Leech Building; Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| |
Collapse
|
209
|
Posada MM, Bacon JA, Schneck KB, Tirona RG, Kim RB, Higgins JW, Pak YA, Hall SD, Hillgren KM. Prediction of renal transporter mediated drug-drug interactions for pemetrexed using physiologically based pharmacokinetic modeling. Drug Metab Dispos 2015; 43:325-34. [PMID: 25504564 DOI: 10.1124/dmd.114.059618] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Pemetrexed, an anionic anticancer drug with a narrow therapeutic index, is eliminated mainly by active renal tubular secretion. The in vitro to in vivo extrapolation approach used in this work was developed to predict possible drug-drug interactions (DDIs) that may occur after coadministration of pemetrexed and nonsteroidal anti-inflammatory drugs (NSAIDs), and it included in vitro assays, risk assessment models, and physiologically based pharmacokinetic (PBPK) models. The pemetrexed transport and its inhibition parameters by several NSAIDs were quantified using HEK-PEAK cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs were ranked according to their DDI index, calculated as the ratio of their maximum unbound concentration in plasma over the concentration inhibiting 50% (IC50) of active pemetrexed transport. A PBPK model for ibuprofen, the NSAID with the highest DDI index, was built incorporating active renal secretion in Simcyp Simulator. The bottom-up model for pemetrexed underpredicted the clearance by 2-fold. The model we built using a scaling factor of 5.3 for the maximal uptake rate (Vmax) of OAT3, which estimated using plasma concentration profiles from patients given a 10-minute infusion of 500 mg/m(2) of pemetrexed supplemented with folic acid and vitamin B12, recovered the clinical data adequately. The observed/predicted increases in Cmax and the area under the plasma-concentration time curve (AUC0-inf) of pemetrexed when ibuprofen was coadministered were 1.1 and 1.0, respectively. The coadministration of all other NSAIDs was predicted to have no significant impact on the AUC0-inf based on their DDI indexes. The PBPK model reasonably reproduced pemetrexed concentration time profiles in cancer patients and its interaction with ibuprofen.
Collapse
Affiliation(s)
- Maria M Posada
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| | - James A Bacon
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| | - Karen B Schneck
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| | - Rommel G Tirona
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| | - Richard B Kim
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| | - J William Higgins
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| | - Y Anne Pak
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| | - Stephen D Hall
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| | - Kathleen M Hillgren
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.)
| |
Collapse
|
210
|
Pastor-Anglada M, Pérez-Torras S. Nucleoside transporter proteins as biomarkers of drug responsiveness and drug targets. Front Pharmacol 2015; 6:13. [PMID: 25713533 PMCID: PMC4322540 DOI: 10.3389/fphar.2015.00013] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/13/2015] [Indexed: 12/13/2022] Open
Abstract
Nucleoside and nucleobase analogs are currently used in the treatment of solid tumors, lymphoproliferative diseases, viral infections such as hepatitis and AIDS, and some inflammatory diseases such as Crohn. Two gene families are implicated in the uptake of nucleosides and nucleoside analogs into cells, SCL28 and SLC29. The former encodes hCNT1, hCNT2, and hCNT3 proteins. They translocate nucleosides in a Na+ coupled manner with high affinity and some substrate selectivity, being hCNT1 and hCNT2 pyrimidine- and purine-preferring, respectively, and hCNT3 a broad selectivity transporter. SLC29 genes encode four members, being hENT1 and hENT2 the only two which are unequivocally implicated in the translocation of nucleosides and nucleobases (the latter mostly via hENT2) at the cell plasma membrane. Some nucleoside-derived drugs can also interact with and be translocated by members of the SLC22 gene family, particularly hOCT and hOAT proteins. Inter-individual differences in transporter function and perhaps, more importantly, altered expression associated with the disease itself might modulate the transporter profile of target cells, thereby determining drug bioavailability and action. Drug transporter pharmacology has been periodically reviewed. Thus, with this contribution we aim at providing a state-of-the-art overview of the clinical evidence generated so far supporting the concept that these membrane proteins can indeed be biomarkers suitable for diagnosis and/or prognosis. Last but not least, some of these transporter proteins can also be envisaged as drug targets, as long as they can show “transceptor” functions, in some cases related to their role as modulators of extracellular adenosine levels, thereby providing a functional link between P1 receptors and transporters.
Collapse
Affiliation(s)
- Marçal Pastor-Anglada
- Molecular Pharmacology and Experimental Therapeutics, Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University of Barcelona, Barcelona Spain ; Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona Spain
| | - Sandra Pérez-Torras
- Molecular Pharmacology and Experimental Therapeutics, Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University of Barcelona, Barcelona Spain ; Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona Spain
| |
Collapse
|
211
|
|
212
|
Babelova A, Burckhardt BC, Wegner W, Burckhardt G, Henjakovic M. Sex-differences in renal expression of selected transporters and transcription factors in lean and obese Zucker spontaneously hypertensive fatty rats. J Diabetes Res 2015; 2015:483238. [PMID: 25710042 PMCID: PMC4325971 DOI: 10.1155/2015/483238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 01/22/2023] Open
Abstract
The aim of this study was to identify sex-dependent expression of renal transporter mRNA in lean and obese Zucker spontaneously hypertensive fatty (ZSF1) rats and to investigate the interaction of the most altered transporter, organic anion transporter 2 (Oat2), with diabetes-relevant metabolites and drugs. Higher incidence of glomerulosclerosis, tubulointerstitial fibrosis, and protein casts in Bowman's space and tubular lumen was detected by PAS staining in obese male compared to female ZSF1 rats. Real-time PCR on RNA isolated from kidney cortex revealed that Sglt1-2, Oat1-3, and Oct1 were higher expressed in kidneys of lean females. Oct2 and Mrp2 were higher expressed in obese males. Renal mRNA levels of transporters were reduced with diabetic nephropathy in females and the expression of transcription factors Hnf1β and Hnf4α in both sexes. The highest difference between lean and obese ZSF1 rats was found for Oat2. Therefore, we have tested the interaction of human OAT2 with various substances using tritium-labeled cGMP. Human OAT2 showed no interaction with diabetes-related metabolites, diabetic drugs, and ACE-inhibitors. However, OAT2-dependent uptake of cGMP was inhibited by furosemide. The strongly decreased expression of Oat2 and other transporters in female diabetic ZSF1 rats could possibly impair renal drug excretion, for example, of furosemide.
Collapse
Affiliation(s)
- Andrea Babelova
- Institute for Cardiovascular Physiology (Physiology I), Faculty of Medicine, Goethe-University, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 83391 Bratislava, Slovakia
| | - Birgitta C. Burckhardt
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Waja Wegner
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Gerhard Burckhardt
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Maja Henjakovic
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
- *Maja Henjakovic:
| |
Collapse
|
213
|
Abstract
Measuring renal function in neonates and small infants is important to ensure that drugs are safely dosed and to detect acute kidney injuries early on. Serum creatinine (Cr) remains the most widely used marker, but its shortcomings are particularly important in neonates. For example, neonatal Cr largely depends on maternal renal function for at least the first 72 h of life. Novel approaches for assessing neonatal renal function include cystatin C and beta-trace protein. Another way to assess renal function is to measure renal volume by ultrasound. Although this approach may assess neonatal nephron endowment, it is insensitive to the postnatal adaptation of renal function in term and preterm neonates. The purpose of this review is to summarize what is known about measuring renal function in term and preterm newborns, and to summarize existing knowledge gaps, including a description of steps to take to close these gaps.
Collapse
|
214
|
Hazelhoff MH, Trebucobich MS, Stoyanoff TR, Chevalier AA, Torres AM. Amelioration of mercury nephrotoxicity after pharmacological manipulation of organic anion transporter 1 (Oat1) and multidrug resistance-associated protein 2 (Mrp2) with furosemide. Toxicol Res (Camb) 2015. [DOI: 10.1039/c5tx00100e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Furosemide improves HgCl2-induced tubule injury up-regulating Oat1 and Mrp2, thus increasing renal elimination of mercuric ions.
Collapse
Affiliation(s)
- María H. Hazelhoff
- Área Farmacología
- Facultad de Ciencias Bioquímicas y Farmacéuticas
- Universidad Nacional de Rosario
- Rosario
- Argentina
| | - Mara S. Trebucobich
- Área Farmacología
- Facultad de Ciencias Bioquímicas y Farmacéuticas
- Universidad Nacional de Rosario
- Rosario
- Argentina
| | - Tania R. Stoyanoff
- Departamento de Bioquímica
- Facultad de Medicina
- Universidad Nacional del Nordeste
- Corrientes
- Argentina
| | - Alberto A. Chevalier
- GIHON Laboratorios Químicos SRL
- Facultad de Ciencias Exactas
- Universidad Nacional de Mar del Plata
- Mar del Plata
- Argentina
| | - Adriana M. Torres
- Área Farmacología
- Facultad de Ciencias Bioquímicas y Farmacéuticas
- Universidad Nacional de Rosario
- Rosario
- Argentina
| |
Collapse
|
215
|
|
216
|
Nigam SK, Bush KT, Martovetsky G, Ahn SY, Liu HC, Richard E, Bhatnagar V, Wu W. The organic anion transporter (OAT) family: a systems biology perspective. Physiol Rev 2015; 95:83-123. [PMID: 25540139 PMCID: PMC4281586 DOI: 10.1152/physrev.00025.2013] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The organic anion transporter (OAT) subfamily, which constitutes roughly half of the SLC22 (solute carrier 22) transporter family, has received a great deal of attention because of its role in handling of common drugs (antibiotics, antivirals, diuretics, nonsteroidal anti-inflammatory drugs), toxins (mercury, aristolochic acid), and nutrients (vitamins, flavonoids). Oats are expressed in many tissues, including kidney, liver, choroid plexus, olfactory mucosa, brain, retina, and placenta. Recent metabolomics and microarray data from Oat1 [Slc22a6, originally identified as NKT (novel kidney transporter)] and Oat3 (Slc22a8) knockouts, as well as systems biology studies, indicate that this pathway plays a central role in the metabolism and handling of gut microbiome metabolites as well as putative uremic toxins of kidney disease. Nuclear receptors and other transcription factors, such as Hnf4α and Hnf1α, appear to regulate the expression of certain Oats in conjunction with phase I and phase II drug metabolizing enzymes. Some Oats have a strong selectivity for particular signaling molecules, including cyclic nucleotides, conjugated sex steroids, odorants, uric acid, and prostaglandins and/or their metabolites. According to the "Remote Sensing and Signaling Hypothesis," which is elaborated in detail here, Oats may function in remote interorgan communication by regulating levels of signaling molecules and key metabolites in tissues and body fluids. Oats may also play a major role in interorganismal communication (via movement of small molecules across the intestine, placental barrier, into breast milk, and volatile odorants into the urine). The role of various Oat isoforms in systems physiology appears quite complex, and their ramifications are discussed in the context of remote sensing and signaling.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Kevin T Bush
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Gleb Martovetsky
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Sun-Young Ahn
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Henry C Liu
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Erin Richard
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Vibha Bhatnagar
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Wei Wu
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
217
|
Donovan MD, O'Brien FE, Boylan GB, Cryan JF, Griffin BT. The effect of organic anion transporter 3 inhibitor probenecid on bumetanide levels in the brain: an integrated in vivo microdialysis study in the rat. J Pharm Pharmacol 2014; 67:501-10. [DOI: 10.1111/jphp.12341] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 09/28/2014] [Indexed: 01/29/2023]
Abstract
Abstract
Objectives
Recent data highlight the potential of bumetanide as a treatment for neonatal seizures and autism, as it facilitates the excitatory to inhibitory switch in gamma-aminobutyric acid signalling. This study examines the extent of blood-brain barrier (BBB) permeation of bumetanide, a key determinant of the efficacy of centrally acting drugs. Furthermore, the impact of efflux transporter organic anion transporter 3 (oat3) inhibition on bumetanide pharmacokinetics was investigated.
Methods
Bumetanide levels in extracellular fluid (ECF) and plasma in the presence and absence of oat3 inhibitor probenecid were monitored using integrated microdialysis.
Key findings
Following a bumetanide bolus/continuous infusion of 10 mg/kg and 6 mg/kg/h, bumetanide was detected in hippocampal ECF at the estimated concentration of 131 ± 55 ng/ml. Plasma bumetanide levels were ∼20 mg/l at steady state. Coadministration of probenecid resulted in an increase in bumetanide levels in both ECF and plasma, indicating that oat3 inhibition influences the pharmacokinetics of bumetanide primarily in the periphery.
Conclusion
Although bumetanide reached detectable levels in hippocampal ECF, bumetanide concentration in ECF was low relative to systemic concentration. Oat3 inhibition by probenecid resulted in increased bumetanide concentrations in brain and plasma. As an acute treatment in neonatal seizures, the bumetanide/probenecid combination may hold therapeutic potential.
Collapse
Affiliation(s)
- Maria D Donovan
- Pharmacodelivery Group, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Fionn E O'Brien
- Pharmacodelivery Group, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Geraldine B Boylan
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- Irish Centre for Fetal and Neonatal Translational Research, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | |
Collapse
|
218
|
Wegner W, Burckhardt G, Henjakovic M. Transcriptional regulation of human organic anion transporter 1 by B-cell CLL/lymphoma 6. Am J Physiol Renal Physiol 2014; 307:F1283-91. [DOI: 10.1152/ajprenal.00426.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The human organic anion transporter 1 (OAT1) is crucial for the excretion of organic anions in renal proximal tubular cells and has been classified as a clinically relevant transporter in the kidneys. Our previous study indicated that renal male-predominant expression of rat Oat1 and Oat3 appears to be regulated by transcription factor B-cell CLL/lymphoma 6 (BCL6). The aim of this study was to characterize the effect of BCL6 on human OAT1 promoter and on the transcription of OAT1 mediated by hepatocyte nuclear factor-1α (HNF-1α). Luciferase assays were carried out in opossum kidney (OK) cells transiently transfected with promoter constructs of OAT1, expression vectors for BCL6 and HNF-1α, and the empty control vectors. BCL6 and HNF-1α binding on OAT1 promoter was analyzed using electrophoretic mobility shift assay (EMSA). Protein expression of HNF-1α was investigated by Western blot analysis. Site-directed mutagenesis was used to introduce mutations into BCL6 and HNF-1α binding sites within the OAT1 promoter. BCL6 enhanced the promoter activity of OAT1 independently of predicted BCL6 binding sites but was dependent on HNF-1α response element and HNF-1α protein. Coexpression of BCL6 and HNF-1α induced an additive effect on OAT1 promoter activation compared with BCL6 or HNF-1α alone. BCL6 does not bind directly or indirectly to OAT1 promoter but increases the protein expression of HNF-1α and thereby indirectly enhances OAT1 gene transcription. BCL6 constitutes a promising candidate gene for the regulation of human OAT1 transcription and other renal and/or hepatic drug transporters that have been already shown to be activated by HNF-1α.
Collapse
Affiliation(s)
- Waja Wegner
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerhard Burckhardt
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Maja Henjakovic
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
219
|
Chioukh R, Noel-Hudson MS, Ribes S, Fournier N, Becquemont L, Verstuyft C. Proton pump inhibitors inhibit methotrexate transport by renal basolateral organic anion transporter hOAT3. Drug Metab Dispos 2014; 42:2041-8. [PMID: 25239859 DOI: 10.1124/dmd.114.058529] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The coadministration of methotrexate (MTX) and proton pump inhibitors (PPIs) can result in a pharmacokinetic interaction that delays MTX elimination and subsequently increases the MTX blood concentrations. Human organic anion transporters (hOATs) are responsible for the renal tubular secretion of MTX and are thought to be involved in this drug interaction. The aim of this study was to evaluate the inhibitory potencies of PPIs on hOAT1 and hOAT3, which are the two isoforms of OATs predominantly expressed in kidney proximal tubules. Using stably transfected cell systems that express the uptake transporters human embryonic kidney (HEK)-hOAT1 and HEK-hOAT3, we analyzed the inhibitory potencies of omeprazole, lansoprazole, and pantoprazole on OAT-mediated [(3)H]estrone sulfate (ES), [(3)H]p-aminohippuric acid (PAH), and [(3)H]MTX uptake in vitro. hOAT3 is a high affinity transporter for MTX (Km = 21.17 ± 5.65 µM). Omeprazole, lansoprazole, and pantoprazole inhibited [(3)H]MTX uptake in HEK-hOAT3 cells with an IC50 of 6.8 ± 1.16, 1.14 ± 0.26, and 4.45 ± 1.62 µM, respectively, and inhibited the [(3)H]ES uptake in HEK-hOAT3 cells with an IC50 of 20.59 ± 4.07, 3.96 ± 0.96, and 7.89 ± 2.31 µM, respectively. Furthermore, omeprazole, lansoprazole, and pantoprazole exhibited inhibited PAH uptake on hOAT1 in a concentration-dependent manner (IC50 = 4.32 ± 1.26, 7.58 ± 1.06, and 63.21 ± 4.74 µM, respectively). These in vitro results suggest that PPIs inhibit [(3)H]MTX transport via hOAT3 inhibition, which most likely explains the drug-drug interactions between MTX and PPIs and should be considered for other OATs substrates.
Collapse
Affiliation(s)
- Rym Chioukh
- EA 4123 Barrières Physiologiques et Réponses Thérapeutiques (R.C., M.-S.N.-H., S.R., L.B., C.V.) and EA 4529 Lipides Membranaires et Régulation Fonctionnelle du Cœur et des Vaisseaux (N.F.), Université Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France; Centre de Recherche Clinique Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (L.B.); and Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (C.V.)
| | - Marie-Sophie Noel-Hudson
- EA 4123 Barrières Physiologiques et Réponses Thérapeutiques (R.C., M.-S.N.-H., S.R., L.B., C.V.) and EA 4529 Lipides Membranaires et Régulation Fonctionnelle du Cœur et des Vaisseaux (N.F.), Université Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France; Centre de Recherche Clinique Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (L.B.); and Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (C.V.)
| | - Sandy Ribes
- EA 4123 Barrières Physiologiques et Réponses Thérapeutiques (R.C., M.-S.N.-H., S.R., L.B., C.V.) and EA 4529 Lipides Membranaires et Régulation Fonctionnelle du Cœur et des Vaisseaux (N.F.), Université Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France; Centre de Recherche Clinique Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (L.B.); and Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (C.V.)
| | - Natalie Fournier
- EA 4123 Barrières Physiologiques et Réponses Thérapeutiques (R.C., M.-S.N.-H., S.R., L.B., C.V.) and EA 4529 Lipides Membranaires et Régulation Fonctionnelle du Cœur et des Vaisseaux (N.F.), Université Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France; Centre de Recherche Clinique Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (L.B.); and Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (C.V.)
| | - Laurent Becquemont
- EA 4123 Barrières Physiologiques et Réponses Thérapeutiques (R.C., M.-S.N.-H., S.R., L.B., C.V.) and EA 4529 Lipides Membranaires et Régulation Fonctionnelle du Cœur et des Vaisseaux (N.F.), Université Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France; Centre de Recherche Clinique Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (L.B.); and Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (C.V.)
| | - Celine Verstuyft
- EA 4123 Barrières Physiologiques et Réponses Thérapeutiques (R.C., M.-S.N.-H., S.R., L.B., C.V.) and EA 4529 Lipides Membranaires et Régulation Fonctionnelle du Cœur et des Vaisseaux (N.F.), Université Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France; Centre de Recherche Clinique Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (L.B.); and Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France (C.V.)
| |
Collapse
|
220
|
Töllner K, Brandt C, Römermann K, Löscher W. The organic anion transport inhibitor probenecid increases brain concentrations of the NKCC1 inhibitor bumetanide. Eur J Pharmacol 2014; 746:167-73. [PMID: 25449033 DOI: 10.1016/j.ejphar.2014.11.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 10/22/2014] [Accepted: 11/09/2014] [Indexed: 12/14/2022]
Abstract
Bumetanide is increasingly being used for experimental treatment of brain disorders, including neonatal seizures, epilepsy, and autism, because the neuronal Na-K-Cl cotransporter NKCC1, which is inhibited by bumetanide, is implicated in the pathophysiology of such disorders. However, use of bumetanide for treatment of brain disorders is associated with problems, including poor brain penetration and systemic adverse effects such as diuresis, hypokalemic alkalosis, and hearing loss. The poor brain penetration is thought to be related to its high ionization rate and plasma protein binding, which restrict brain entry by passive diffusion, but more recently brain efflux transporters have been involved, too. Multidrug resistance protein 4 (MRP4), organic anion transporter 3 (OAT3) and organic anion transporting polypeptide 2 (OATP2) were suggested to mediate bumetanide brain efflux, but direct proof is lacking. Because MRP4, OAT3, and OATP2 can be inhibited by probenecid, we studied whether this drug alters brain levels of bumetanide in mice. Probenecid (50 mg/kg) significantly increased brain levels of bumetanide up to 3-fold; however, it also increased its plasma levels, so that the brain:plasma ratio (~0.015-0.02) was not altered. Probenecid markedly increased the plasma half-life of bumetanide, indicating reduced elimination of bumetanide most likely by inhibition of OAT-mediated transport of bumetanide in the kidney. However, the diuretic activity of bumetanide was not reduced by probenecid. In conclusion, our study demonstrates that the clinically available drug probenecid can be used to increase brain levels of bumetanide and decrease its elimination, which could have therapeutic potential in the treatment of brain disorders.
Collapse
Affiliation(s)
- Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
221
|
Chen X, Loryan I, Payan M, Keep RF, Smith DE, Hammarlund-Udenaes M. Effect of transporter inhibition on the distribution of cefadroxil in rat brain. Fluids Barriers CNS 2014; 11:25. [PMID: 25414790 PMCID: PMC4237734 DOI: 10.1186/2045-8118-11-25] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/18/2014] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Cefadroxil, a cephalosporin antibiotic, is a substrate for several membrane transporters including peptide transporter 2 (PEPT2), organic anion transporters (OATs), multidrug resistance-associated proteins (MRPs), and organic anion transporting polypeptides (OATPs). These transporters are expressed at the blood-brain barrier (BBB), blood-cerebrospinal fluid barrier (BCSFB), and/or brain cells. The effect of these transporters on cefadroxil distribution in brain is unknown, especially in the extracellular and intracellular fluids within brain. METHODS Intracerebral microdialysis was used to measure unbound concentrations of cefadroxil in rat blood, striatum extracellular fluid (ECF) and lateral ventricle cerebrospinal fluid (CSF). The distribution of cefadroxil in brain was compared in the absence and presence of probenecid, an inhibitor of OATs, MRPs and OATPs, where both drugs were administered intravenously. The effect of PEPT2 inhibition by intracerebroventricular (icv) infusion of Ala-Ala, a substrate of PEPT2, on cefadroxil levels in brain was also evaluated. In addition, using an in vitro brain slice method, the distribution of cefadroxil in brain intracellular fluid (ICF) was studied in the absence and presence of transport inhibitors (probenecid for OATs, MRPs and OATPs; Ala-Ala and glycylsarcosine for PEPT2). RESULTS The ratio of unbound cefadroxil AUC in brain ECF to blood (Kp,uu,ECF) was ~2.5-fold greater during probenecid treatment. In contrast, the ratio of cefadroxil AUC in CSF to blood (Kp,uu,CSF) did not change significantly during probenecid infusion. Icv infusion of Ala-Ala did not change cefadroxil levels in brain ECF, CSF or blood. In the brain slice study, Ala-Ala and glycylsarcosine decreased the unbound volume of distribution of cefadroxil in brain (Vu,brain), indicating a reduction in cefadroxil accumulation in brain cells. In contrast, probenecid increased cefadroxil accumulation in brain cells, as indicated by a greater value for Vu,brain. CONCLUSIONS Transporters (OATs, MRPs, and perhaps OATPs) that can be inhibited by probenecid play an important role in mediating the brain-to-blood efflux of cefadroxil at the BBB. The uptake of cefadroxil in brain cells involves both the influx transporter PEPT2 and efflux transporters (probenecid-inhibitable). These findings demonstrate that drug-drug interactions via relevant transporters may affect the distribution of cephalosporins in both brain ECF and ICF.
Collapse
Affiliation(s)
- Xiaomei Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Mi 48109 USA ; Department of Pharmaceutical Biosciences, Translational PKPD Research Group, Uppsala University, Box 591, SE-75124 Uppsala, Sweden
| | - Irena Loryan
- Department of Pharmaceutical Biosciences, Translational PKPD Research Group, Uppsala University, Box 591, SE-75124 Uppsala, Sweden
| | - Maryam Payan
- Department of Pharmaceutical Biosciences, Translational PKPD Research Group, Uppsala University, Box 591, SE-75124 Uppsala, Sweden ; Biopharmaceutics and Pharmacokinetic Division, Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Health System, Ann Arbor, MI 48109 USA
| | - David E Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Mi 48109 USA
| | - Margareta Hammarlund-Udenaes
- Department of Pharmaceutical Biosciences, Translational PKPD Research Group, Uppsala University, Box 591, SE-75124 Uppsala, Sweden
| |
Collapse
|
222
|
Ruiz ML, Mottino AD, Catania VA, Vore M. Hormonal regulation of hepatic drug biotransformation and transport systems. Compr Physiol 2014; 3:1721-40. [PMID: 24265243 DOI: 10.1002/cphy.c130018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The human body is constantly exposed to many xenobiotics including environmental pollutants, food additives, therapeutic drugs, etc. The liver is considered the primary site for drug metabolism and elimination pathways, consisting in uptake, phase I and II reactions, and efflux processes, usually acting in this same order. Modulation of biotransformation and disposition of drugs of clinical application has important therapeutic and toxicological implications. We here provide a compilation and analysis of relevant, more recent literature reporting hormonal regulation of hepatic drug biotransformation and transport systems. We provide additional information on the effect of hormones that tentatively explain differences between sexes. A brief discussion on discrepancies between experimental models and species, as well as a link between gender-related differences and the hormonal mechanism explaining such differences, is also presented. Finally, we include a comment on the pathophysiological, toxicological, and pharmacological relevance of these regulations.
Collapse
Affiliation(s)
- María L Ruiz
- Institute of Experimental Physiology, National University of Rosario, Rosario, Argentina
| | | | | | | |
Collapse
|
223
|
Schreuder MF, Bueters RRG, Allegaert K. The interplay between drugs and the kidney in premature neonates. Pediatr Nephrol 2014; 29:2083-91. [PMID: 24217783 DOI: 10.1007/s00467-013-2651-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 09/24/2013] [Accepted: 09/26/2013] [Indexed: 02/06/2023]
Abstract
The kidney plays a central role in the clearance of drugs. However, renal drug handling entails more than glomerular filtration and includes tubular excretion and reabsorption, and intracellular metabolization by cellular enzyme systems, such as the Cytochrome P450 isoenzymes. All these processes show maturation from birth onwards, which is one of the reasons why drug dosing in children is not simply similar to dosing in small adults. As kidney development normally finishes around the 36th week of gestation, being born prematurely will result in even more immature renal drug handling. Environmental effects, such as extra-uterine growth restriction, sepsis, asphyxia, or drug treatments like caffeine, aminoglycosides, or non-steroidal anti-inflammatory drugs, may further hamper drug handling in the kidney. Dosing in preterm neonates is therefore dependent on many factors that need to be taken into account. Drug treatment may significantly hamper postnatal kidney development in preterm neonates, just like renal immaturity has an impact on drug handling. The restricted kidney development results in a lower number of nephrons that may have several long-term sequelae, such as hypertension, albuminuria, and renal failure. This review focuses on the interplay between drugs and the kidney in premature neonates.
Collapse
Affiliation(s)
- Michiel F Schreuder
- Department of Pediatric Nephrology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands,
| | | | | |
Collapse
|
224
|
Schwob E, Hagos Y, Burckhardt G, Burckhardt BC. Transporters involved in renal excretion of N-carbamoylglutamate, an orphan drug to treat inborn n-acetylglutamate synthase deficiency. Am J Physiol Renal Physiol 2014; 307:F1373-9. [PMID: 25354943 DOI: 10.1152/ajprenal.00482.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Inborn defects in N-acetylglutamate (NAG) synthase (NAGS) cause a reduction of NAG, an essential cofactor for the initiation of the urea cycle. As a consequence, blood ammonium concentrations are elevated, leading to severe neurological disorders. The orphan drug N-carbamoylglutamate (NCG; Carbaglu), efficiently overcomes NAGS deficiency. However, not much is known about the transporters involved in the uptake, distribution, and elimination of the divalent organic anion NCG. Organic anion-transporting polypeptides (OATPs) as well as organic anion transporters (OATs) working in cooperation with sodium dicarboxylate cotransporter 3 (NaDC3) accept a wide variety of structurally unrelated drugs. To test for possible interactions with OATPs and OATs, the impact of NCG on these transporters in stably transfected human embryonic kidney-293 cells was measured. The two-electrode voltage-clamp technique was used to monitor NCG-mediated currents in Xenopus laevis oocytes that expressed NaDC3. Neither OATPs nor OAT2 and OAT3 interacted with NCG, but OAT1 transported NCG. In addition, NCG was identified as a high-affinity substrate of NaDC3. Preincubation of OAT4-transfected human embryonic kidney-293 cells with NCG showed an increased uptake of estrone sulfate, the reference substrate of OAT4, indicating efflux of NCG by OAT4. In summary, NaDC3 and, to a lesser extent, OAT1 are likely to be responsible for the uptake of NCG from the blood. Efflux of NCG across the luminal membrane into the tubular lumen probably occurs by OAT4 completing renal secretion of this drug.
Collapse
Affiliation(s)
- Elisabeth Schwob
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Yohannes Hagos
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerhard Burckhardt
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Birgitta C Burckhardt
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
225
|
Hagos Y, Wegner W, Kuehne A, Floerl S, Marada VV, Burckhardt G, Henjakovic M. HNF4α Induced Chemosensitivity to Oxaliplatin and 5-FU Mediated by OCT1 and CNT3 in Renal Cell Carcinoma. J Pharm Sci 2014; 103:3326-34. [DOI: 10.1002/jps.24128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/11/2014] [Accepted: 07/31/2014] [Indexed: 12/16/2022]
|
226
|
Abstract
PURPOSE OF REVIEW Although uricosuric agents provide the most time-honoured approach to the control of hyperuricemia, their place in the armamentarium has been eclipsed by that of xanthine oxidase inhibitors. This review considers the potential for uricosuric agents from the perspective of recent progress in the understanding of urate transport systems. RECENT FINDINGS No new agents have yet become available, but promising new drugs are under development. Better understanding of the transporters URAT1 and ABCG2 in particular would appear to provide opportunities for more selective, better tolerated agents to increase the renal clearance of uric acid and thereby control hyperuricemia. SUMMARY Conceptually, modest inhibition of renal tubular reabsorption should provide effective relief for the millions of individuals who are now hyperuricemic and who suffer from its principal consequence, gout.
Collapse
|
227
|
Kim YC, Kim IB, Noh CK, Quach HP, Yoon IS, Chow ECY, Kim M, Jin HE, Cho KH, Chung SJ, Pang KS, Maeng HJ. Effects of 1α,25-dihydroxyvitamin D3 , the natural vitamin D receptor ligand, on the pharmacokinetics of cefdinir and cefadroxil, organic anion transporter substrates, in rat. J Pharm Sci 2014; 103:3793-3805. [PMID: 25266751 DOI: 10.1002/jps.24195] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 08/21/2014] [Accepted: 09/11/2014] [Indexed: 01/31/2023]
Abstract
Evidence in the literature suggests that 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ], the vitamin D receptor ligand, down-regulated the expression of the rat renal organic anion (renal organic anion transporter, rOAT) and oligopeptide (rPEPT) transporters, but increased intestinal rPEPT1 expression. We investigated, in rats, the intravenous and oral pharmacokinetics of 2 mg/kg cefdinir and cefadroxil, two cephalosporins that are eliminated via renal OAT1/OAT3 and are substrates of PEPT1/PEPT2, with and without 1,25(OH)2 D3 treatment. The area under the plasma concentration-time curve (AUC) of cefdinir or cefadroxil after 1,25(OH)2 D3 treatment was increased significantly because of decreased clearance (CL). Both kidney uptake and cumulative urinary recovery were significantly decreased, whereas liver uptake and fecal recovery remained unchanged in 1,25(OH)2 D3 -treated rats. Similar changes in AUC and CL were observed for both drugs upon coadministration of probenecid, the OAT inhibitor. Oral availability of cefdinir and cefadroxil remained unchanged with 1,25(OH)2 D3 treatment, suggesting lack of a role for intestinal rPEPT1. Rather, reduction of rOAT1/rOAT3 mRNA expression in kidney with 1,25(OH)2 D3 -treatment was observed, confirmed by decreased function in MDCKII cells overexpressing human OAT1 and OAT3. These composite results suggest that 1,25(OH)2 D3 treatment reduces cefdinir and cefadroxil clearances by diminution of renal OAT1/OAT3 expression, implicating a role for 1,25(OH)2 D3 in eliciting transporter-based drug interactions.
Collapse
Affiliation(s)
- Yu Chul Kim
- C&C Research Laboratories, Suwon, Gyeonggi, Republic of Korea
| | - In-Bong Kim
- College of Pharmacy, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Chi-Kyoung Noh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Holly P Quach
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - In-Soo Yoon
- College of Pharmacy, and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Edwin C Y Chow
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Myungsoo Kim
- College of Pharmacy, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Hyo-Eon Jin
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Kwan Hyung Cho
- College of Pharmacy, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Suk-Jae Chung
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Han-Joo Maeng
- College of Pharmacy, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea.
| |
Collapse
|
228
|
An G, Wang X, Morris ME. Flavonoids are inhibitors of human organic anion transporter 1 (OAT1)-mediated transport. Drug Metab Dispos 2014; 42:1357-66. [PMID: 25002746 PMCID: PMC4152875 DOI: 10.1124/dmd.114.059337] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/07/2014] [Indexed: 11/22/2022] Open
Abstract
Organic anion transporter 1 (OAT1) has been reported to be involved in the nephrotoxicity of many anionic xenobiotics. As current clinically used OAT1 inhibitors are often associated with safety issues, identifying potent OAT1 inhibitors with little toxicity is of great value in reducing OAT1-mediated drug nephrotoxicity. Flavonoids are a class of polyphenolic compounds with exceptional safety records. Our objective was to evaluate the effects of 18 naturally occurring flavonoids, and some of their glycosides, on the uptake of para-aminohippuric acid (PAH) in both OAT1-expressing and OAT1-negative LLC-PK1 cells. Most flavonoid aglycones produced substantial decreases in PAH uptake in OAT1-expressing cells. Among the flavonoids screened, fisetin, luteolin, morin, and quercetin exhibited the strongest effect and produced complete inhibition of OAT1-mediated PAH uptake at a concentration of 50 μM. Further concentration-dependent studies revealed that both morin and luteolin are potent OAT1 inhibitors, with IC50 values of <0.3 and 0.47 μM, respectively. In contrast to the tested flavonoid aglycones, all flavonoid glycosides had negligible or small effects on OAT1. In addition, the role of OAT1 in the uptake of fisetin, luteolin, morin, and quercetin was investigated and fisetin was found to be a substrate of OAT1. Taken together, our results indicate that flavonoids are a novel class of OAT1 modulators. Considering the high consumption of flavonoids in the diet and in herbal products, OAT1-mediated flavonoid-drug interactions may be clinically relevant. Further investigation is warranted to evaluate the nephroprotective role of flavonoids in relation to drug-induced nephrotoxicity mediated by the OAT1 pathway.
Collapse
Affiliation(s)
- Guohua An
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York, University at Buffalo, Buffalo, New York
| | - Xiaodong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York, University at Buffalo, Buffalo, New York
| | - Marilyn E Morris
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York, University at Buffalo, Buffalo, New York
| |
Collapse
|
229
|
Bulacio RP, Torres AM. Time course of organic anion transporter 5 (Oat5) urinary excretion in rats treated with cisplatin: a novel urinary biomarker for early detection of drug-induced nephrotoxicity. Arch Toxicol 2014; 89:1359-69. [PMID: 25164828 DOI: 10.1007/s00204-014-1345-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/14/2014] [Indexed: 01/08/2023]
Abstract
Cisplatin is a widely used citostatic drug employed in the treatment of many solid tumors. Its principal side-effect is nephrotoxicity. The organic anion transporter 5 (Oat5) is exclusively expressed in the kidneys. The aim of this study was to evaluate the time course of Oat5 urinary excretion and changes in conventional biomarkers, such as creatinine and urea plasma levels (Urp and Crp), and protein and glucose urinary levels (Pu and Gluu), between others, and compared them to the onset and progression of histological changes after cisplatin treatment. Male Wistar rats were treated with cisplatin with 5 mg/kg b.w., i.p., and experiments were carried out after 2, 4, 7 and 14 days of treatment. Two days after cisplatin administration, only Oat5 urinary excretion was found markedly modified. On day 4, Urp, Crp, PU and GluU were increased. By the seventh day, a severe impairment in tubular architecture was observed, and from this point and thereon, Oat5 urinary excretion and PU showed a tendency to return to their basal values. Meanwhile, Urp, Crp and GluU tended to return to their basal values by the day 14 of treatment, when kidney morphology showed an important recovery. So Oat5 urinary abundance was elevated 2 days after cisplatin treatment, when no modifications of traditional markers of renal injury were still observed. Therefore, the results showed in this work, in addition to previous data obtained by our group, propose that Oat5 urinary excretion might potentially serve as a noninvasive early biomarker of cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Romina Paula Bulacio
- Area Farmacología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, CONICET, Suipacha 531, 2000, Rosario, Argentina
| | | |
Collapse
|
230
|
Li X, Sun X, Chen J, Lu Y, Zhang Y, Wang C, Li J, Zhang Q, Zhao D, Chen X. Investigation of the role of organic cation transporter 2 (OCT2) in the renal transport of guanfacine, a selective α2A-adrenoreceptor agonist. Xenobiotica 2014; 45:88-94. [DOI: 10.3109/00498254.2014.949904] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
231
|
Post-transcriptional regulation of the human reduced folate carrier as a novel adaptive mechanism in response to folate excess or deficiency. Biosci Rep 2014; 34:BSR20140065. [PMID: 24949876 PMCID: PMC4122975 DOI: 10.1042/bsr20140065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The RFC (reduced folate carrier) is the principal mechanism by which folates and clinically used antifolates are delivered to mammalian cells. hRFC (human RFC) is subject to complex transcriptional controls and exists as homo-oligomer. To explore the post-transcriptional regulation of hRFC by exogenous folates, hRFC-null HeLa cells were stably transfected with hRFC under control of a constitutive promoter. hRFC transcripts and the total membrane protein increased with increasing LCV [(6R,S)5-formyl tetrahydrofolate (leucovorin)] with a maximum at 20 nM LCV, attributable to reduced turnover of hRFC transcripts. hRFC homo-oligomerization was unaffected by increasing LCV. Cell surface hRFC paralleled [3H]methotrexate transport and increased from 0.5 to 2 nM LCV, and then decreased (~2-fold) with increasing LCV up to 20 nM. hRFC was localized to the cell surface at low LCV concentrations (0.5–1.5 nM). However, at higher LCV concentrations, significant intracellular hRFC was localized to the ER (endoplasmic reticulum), such that at 20 nM LCV, intracellular hRFC was predominated. Our results demonstrate a novel post-transcriptional regulation of hRFC involving: (i) increased hRFC transcripts and proteins, accompanying increased extracellular folates, attributable to differences in hRFC transcript stabilities; and (ii) increased retention of hRFC in the ER under conditions of folate excess, because of impaired intracellular trafficking and plasma membrane targeting. A novel regulation of the physiologically/pharmacologically important human reduced folate carrier was demonstrated in response to increasing extracellular folates, involving: (i) increased transcripts and total protein, reflecting increased transcript stabilities; and (ii) increased endoplasmic reticulum trapping, due to impaired intracellular trafficking.
Collapse
|
232
|
Ingraham L, Li M, Renfro JL, Parker S, Vapurcuyan A, Hanna I, Pelis RM. A plasma concentration of α-ketoglutarate influences the kinetic interaction of ligands with organic anion transporter 1. Mol Pharmacol 2014; 86:86-95. [PMID: 24770989 DOI: 10.1124/mol.114.091777] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
The purpose of the present study was to determine whether a physiologic plasma concentration of α-ketoglutarate (αKG) influences the kinetic interaction of ligands with organic anion transporter 1 (OAT1). The effect of extracellular αKG on the kinetics of para-aminohippurate (PAH) and cidofovir transport was examined along with its effect on the potency of 10 drugs in five different classes (uricosuric, nonsteroidal anti-inflammatories, loop diuretics, angiotensin II receptor antagonists, and β-lactam antibiotics) to inhibit OAT1 expressed in Chinese hamster ovary cells. Extracellular αKG competitively inhibited PAH and cidofovir transport with Ki values (∼5 μM) approximating its unbound plasma concentration (determined by equilibrium dialysis). When PAH was the substrate, extracellular αKG (5 μM) significantly increased IC50 values for some inhibitors (up to 4-fold), such as probenecid, but not for others (an inhibitor-dependent effect). For some inhibitors, a significant increase in IC50 value was observed when cidofovir was the substrate, but not PAH (a substrate-dependent effect). A significant increase in IC50 value was also observed for inhibition of PAH transport by probenecid in renal basolateral membrane vesicles (5.2-fold). The substrate- and inhibitor-dependent effect of extracellular αKG on ligand interactions with OAT1 highlights the complexity of the OAT1 ligand-binding surface. The effect of extracellular αKG on the potency of OAT1 inhibition should be considered when assessing drug-drug interaction potential at the transporter.
Collapse
Affiliation(s)
- Leslie Ingraham
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada (L.I., M.L., R.M.P.); Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (J.L.R., S.P.); and Department of Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (A.V., I.H.)
| | - Mansong Li
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada (L.I., M.L., R.M.P.); Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (J.L.R., S.P.); and Department of Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (A.V., I.H.)
| | - J Larry Renfro
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada (L.I., M.L., R.M.P.); Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (J.L.R., S.P.); and Department of Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (A.V., I.H.)
| | - Sonda Parker
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada (L.I., M.L., R.M.P.); Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (J.L.R., S.P.); and Department of Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (A.V., I.H.)
| | - Arpine Vapurcuyan
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada (L.I., M.L., R.M.P.); Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (J.L.R., S.P.); and Department of Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (A.V., I.H.)
| | - Imad Hanna
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada (L.I., M.L., R.M.P.); Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (J.L.R., S.P.); and Department of Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (A.V., I.H.)
| | - Ryan M Pelis
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada (L.I., M.L., R.M.P.); Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (J.L.R., S.P.); and Department of Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (A.V., I.H.)
| |
Collapse
|
233
|
Effects of renal impairment on the pharmacokinetics of morinidazole: uptake transporter-mediated renal clearance of the conjugated metabolites. Antimicrob Agents Chemother 2014; 58:4153-61. [PMID: 24820074 DOI: 10.1128/aac.02414-14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Morinidazole is a novel 5-nitroimidazole antimicrobial drug that undergoes extensive metabolism in humans via N(+)-glucuronidation (N(+)-glucuronide of S-morinidazole [M8-1] and N(+)-glucuronide of R-morinidazole [M8-2]) and sulfation (sulfate conjugate of morinidazole [M7]). Our objectives were to assess the effects of renal impairment on the pharmacokinetics (PK) of morinidazole and to elucidate the potential mechanisms. In this parallel-group study, healthy subjects and patients with severe renal impairment received an intravenous infusion of 500 mg of morinidazole. Plasma and urine samples were collected and analyzed. The areas under the plasma concentration-time curves (AUC) for M7, M8-1, and M8-2 were 15.1, 20.4, and 17.4 times higher, respectively, in patients with severe renal impairment than in healthy subjects, while the AUC for morinidazole was 1.5 times higher. The urinary recovery of the major metabolites was not significantly different between the two groups over 0 to 48 h, but the renal clearances of M7, M8-1, and M8-2 in patients were 85.3%, 92.5%, and 92.2% lower, respectively. In vitro transporter studies revealed that M7 is a substrate for organic anion transporter 1 (OAT1) and OAT3 (Km = 28.6 and 54.0 μM, respectively). Only OAT3 transported M8-1 and M8-2. Morinidazole was not a substrate for the transporter-transfected cells examined. These results revealed that the function or activity of renal uptake transporters might be impaired in patients with severe renal impairment, which accounted for dramatically increased plasma exposure and reduced renal clearance of the conjugated metabolites of morinidazole, the substrates of renal transporters in patients. It will help clinicians to adjust the dose in patients with severe renal impairment and to predict possible transporter-based drug-drug interactions.
Collapse
|
234
|
Klein DM, Cherrington NJ. Organic and inorganic transporters of the testis: A review. SPERMATOGENESIS 2014; 4:e979653. [PMID: 26413398 PMCID: PMC4581056 DOI: 10.4161/21565562.2014.979653] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/16/2014] [Indexed: 12/16/2022]
Abstract
Transporters have a huge impact on the toxicology and pharmacological effects of xenobiotics in addition to being implicated in several diseases. While these important proteins have been well studied in organs such as the kidney or liver, characterization of transporters in the testis is still in the early stages. Knowledge of transporter function may greatly advance the field's understanding of the physiological and toxicological processes that occur in the testis. Several foundational studies involving both organic and inorganic transporters have been critical in furthering our understanding of how the testis interacts with endogenous and xenobiotic compounds. This review provides an overview of how transporters function, their clinical significance, and highlights what is known for many of the important transporters in the testis.
Collapse
Affiliation(s)
- David M Klein
- University of Arizona; Department of Pharmacology and Toxicology; Tucson, AZ, US
| | - Nathan J Cherrington
- University of Arizona; Department of Pharmacology and Toxicology; Tucson, AZ, US
| |
Collapse
|
235
|
Maeda K, Tian Y, Fujita T, Ikeda Y, Kumagai Y, Kondo T, Tanabe K, Nakayama H, Horita S, Kusuhara H, Sugiyama Y. Inhibitory effects of p-aminohippurate and probenecid on the renal clearance of adefovir and benzylpenicillin as probe drugs for organic anion transporter (OAT) 1 and OAT3 in humans. Eur J Pharm Sci 2014; 59:94-103. [PMID: 24747579 DOI: 10.1016/j.ejps.2014.04.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 04/06/2014] [Accepted: 04/07/2014] [Indexed: 01/04/2023]
Abstract
Probe substrates for, and inhibitors of, specific transporters are desired to evaluate quantitatively the in vivo functions of transporters in humans. Based on published data, adefovir and benzylpenicillin were selected as organic anion transporter (OAT) 1- and OAT3-selective probe substrates, respectively. In human kidney slices, probenecid potently inhibited the uptake of both adefovir and benzylpenicillin with inhibition constant (Ki) values of 18.6±5.1 and 12.6±4.2μM, respectively, whereas p-aminohippurate (PAH) preferentially inhibited adefovir uptake. A clinical drug-interaction study involving healthy subjects was performed to investigate the dose-dependent inhibition potencies of probenecid and PAH on the renal clearance of the probe substrates. Adefovir or benzylpenicillin was coadministered with different oral doses of probenecid (500, 750, or 1500mg) or intravenous PAH infusion rates (70, 120, or 210mg/min/person) to the same subject using a crossover design. The renal clearance of adefovir was reduced by 45% and 46% in the subjects treated with the maximum dose of probenecid and PAH, respectively, which was in accordance with the results of in vitro inhibition study. On the other hand, renal clearance of benzylpenicillin was reduced by 78% in the subjects treated with the maximum dose of probenecid (1500mg), which could be explained by its in vitro Ki values. However, PAH unexpectedly increased the renal clearance of benzylpenicillin by 47%. These results suggest that adefovir and benzylpenicillin can be used as probe drugs for OAT1 and OAT3, respectively, and that PAH can be used to investigate the role of OAT1 in the urinary excretion of drugs in humans, whereas it may modulate other transport processes in the kidney.
Collapse
Affiliation(s)
- Kazuya Maeda
- Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ying Tian
- Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoe Fujita
- Clinical Trial Center, Kitasato University East Hospital, 2-1-1 Asamizodai, Minami-ku, Sagamihara City, Kanagawa 252-0380, Japan
| | - Yasuhiko Ikeda
- Clinical Trial Center, Kitasato University East Hospital, 2-1-1 Asamizodai, Minami-ku, Sagamihara City, Kanagawa 252-0380, Japan
| | - Yuji Kumagai
- Clinical Trial Center, Kitasato University East Hospital, 2-1-1 Asamizodai, Minami-ku, Sagamihara City, Kanagawa 252-0380, Japan
| | - Tsunenori Kondo
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Kazunari Tanabe
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Hideki Nakayama
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Shigeru Horita
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Hiroyuki Kusuhara
- Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Research Cluster for Innovation, RIKEN, 1-6 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan.
| |
Collapse
|
236
|
Imamura Y, Tsuruya Y, Damme K, Heer D, Kumagai Y, Maeda K, Murayama N, Okudaira N, Kurihara A, Izumi T, Sugiyama Y, Kusuhara H. 6β-Hydroxycortisol is an endogenous probe for evaluation of drug-drug interactions involving a multispecific renal organic anion transporter, OAT3/SLC22A8, in healthy subjects. Drug Metab Dispos 2014; 42:685-94. [PMID: 24487120 DOI: 10.1124/dmd.113.055475] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
6β-Hydroxycortisol (6β-OHF) is a substrate of the organic anion transporter 3 (OAT3) and the multidrug and toxin extrusion proteins MATE1 and MATE-2K in the corresponding cDNA-transfected cells. This study aimed to examine the contribution of OAT3 and MATEs to the urinary excretion of 6β-OHF in humans using the appropriate in vivo inhibitors, probenecid and pyrimethamine, for OAT3 and MATEs, respectively. Oat3(-/-) mice showed significantly reduced renal clearance of 6β-OHF (CL(renal, 6β-OHF)) compared with wild-type mice (18.1 ± 1.5 versus 7.60 ± 1.8 ml/min/kg). 6β-OHF uptake by human kidney slices was inhibited significantly by probenecid to 20-45% of the control values and partly by 1-methyl-4-phenylpyridinium. 6β-OHF plasma concentration and the amount of 6β-OHF excreted into the urine (X(6β-OHF)) were measured in healthy subjects enrolled in drug-drug interaction studies of benzylpenicillin alone or with probenecid (study 1), adefovir alone or with probenecid (study 2), and metformin alone or with pyrimethamine (study 3). Probenecid treatment caused a 57 and 76% increase in the area under the plasma concentration-time curve for 6β-OHF (AUC(6β-OHF)) in studies 1 and 2, respectively, but did not affect X(6β-OHF). Consequently, CL(renal, 6β-OHF) (milliliters per minute) decreased significantly from 231 ± 11 to 135 ± 9 and from 225 ± 26 to 141 ± 12 after probenecid administration in studies 1 and 2, respectively. By contrast, neither AUC(6β-OHF) nor CL(renal, 6β-OHF) was significantly altered by pyrimethamine administration. Taken together, these data suggest that OAT3 plays a significant role in the urinary excretion of 6β-OHF, and that 6β-OHF can be used to investigate the perpetrators of the pharmacokinetic drug interactions involving OAT3 in humans.
Collapse
Affiliation(s)
- Yuichiro Imamura
- Drug Metabolism & Pharmacokinetics Research Laboratories, R&D Division, Daiichi Sankyo Co., Ltd., Shinagawa-ku, Tokyo, Japan (Y.I., N.M., N.O., A.K., T.I.); Daiichi Sankyo Tissue and Cell Research Center Munich, Daiichi Sankyo Europe GmbH, Martinsried, Germany (K.D., D.H.); Clinical Trial Center, Kitasato East Hospital, Sagamihara, Kanagawa, Japan (Y.K.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Yokohama, Kanagawa, Japan (Y.S.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan (Y.T., K.M., H.K.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Matherly LH, Wilson MR, Hou Z. The major facilitative folate transporters solute carrier 19A1 and solute carrier 46A1: biology and role in antifolate chemotherapy of cancer. Drug Metab Dispos 2014; 42:632-49. [PMID: 24396145 PMCID: PMC3965896 DOI: 10.1124/dmd.113.055723] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/06/2014] [Indexed: 01/19/2023] Open
Abstract
This review summarizes the biology of the major facilitative membrane transporters, the reduced folate carrier (RFC) (Solute Carrier 19A1) and the proton-coupled folate transporter (PCFT) (Solute Carrier 46A1). Folates are essential vitamins, and folate deficiency contributes to a variety of health disorders. RFC is ubiquitously expressed and is the major folate transporter in mammalian cells and tissues. PCFT mediates the intestinal absorption of dietary folates and appears to be important for transport of folates into the central nervous system. Clinically relevant antifolates for cancer, such as methotrexate and pralatrexate, are transported by RFC, and loss of RFC transport is an important mechanism of methotrexate resistance in cancer cell lines and in patients. PCFT is expressed in human tumors, and is active at pH conditions associated with the tumor microenvironment. Pemetrexed is an excellent substrate for both RFC and PCFT. Novel tumor-targeted antifolates related to pemetrexed with selective membrane transport by PCFT over RFC are being developed. In recent years, there have been major advances in understanding the structural and functional properties and the regulation of RFC and PCFT. The molecular bases for methotrexate resistance associated with loss of RFC transport and for hereditary folate malabsorption, attributable to mutant PCFT, were determined. Future studies should continue to translate molecular insights from basic studies of RFC and PCFT biology into new therapeutic strategies for cancer and other diseases.
Collapse
Affiliation(s)
- Larry H Matherly
- Department of Oncology (L.H.M., M.R.W., Z.H.) and Department of Pharmacology (L.H.M.), Wayne State University School of Medicine, Detroit, Michigan; and Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan (L.H.M., Z.H.)
| | | | | |
Collapse
|
238
|
Pastor CM, Müllhaupt B, Stieger B. The role of organic anion transporters in diagnosing liver diseases by magnetic resonance imaging. Drug Metab Dispos 2014; 42:675-84. [PMID: 24398460 DOI: 10.1124/dmd.113.055707] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The expression and transport functions of organic anion transporters are modified in liver diseases, and therefore the vascular clearances of endogenous and exogenous organic anions that are taken up by these transporters have been used to assess liver diseases in patients. More recently, liver imaging with hepatobiliary contrast agents, tracers, and dyes that cross hepatocytes through the organic anion transporting polypeptides (OATPs)-multidrug resistance-associated proteins (MRPs) pathway were developed to detect and characterize focal lesions and to assess the severity of diffuse liver diseases. This review focuses mainly on magnetic resonance imaging and highlights the growing interest in imaging the OATPs-MRP2 pathway to better understand liver diseases. Imaging provides noninvasive measurements of tissue concentrations that result from the interplay between influx and efflux membrane transport systems in normal or injured hepatocytes. Imaging with magnetic resonance hepatobiliary contrast agents improves the detection and the characterization of hepatic focal lesions. New developments of imaging to assess liver function and understand the hepatocellular concentrations of contrast agents are discussed.
Collapse
Affiliation(s)
- Catherine M Pastor
- Laboratoire de Physiopathologie Hépatique et Imagerie Moléculaire, Hôpitaux Universitaires de Genève, Switzerland, and U1149 INSERM-Université Paris Diderot, Sorbonne Paris Cité, Paris, France (C.M.P.); Swiss HPB and Department of Gastroenterology and Hepatology, University Hospital, Zurich, Switzerland (B.M.); and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland (B.S.)
| | | | | |
Collapse
|
239
|
Trebucobich MS, Hazelhoff MH, Chevalier AA, Passamonti S, Brandoni A, Torres AM. Protein expression of kidney and liver bilitranslocase in rats exposed to mercuric chloride--a potential tissular biomarker of toxicity. Toxicol Lett 2014; 225:305-310. [PMID: 24374050 DOI: 10.1016/j.toxlet.2013.11.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 11/18/2013] [Accepted: 11/20/2013] [Indexed: 10/25/2022]
Abstract
Bilitranslocase (BTL) is a plasma membrane carrier that transports organic anions of physiological and pharmacological interest. It is expressed in basolateral plasma membrane of kidney and liver. BTL has been recently described as a marker of transition from normal tissue to its neoplastic transformation in human kidney. Inorganic mercury is a major environmental contaminant that produces many toxic effects. Previous reports have described an interaction between BTL and mercuric ions. This study was designed to evaluate the renal and hepatic expression of BTL in rats exposed to a nephrotoxic and hepatotoxic dose of HgCl2. Male rats were treated with a single injection of HgCl2 at a dose of 4mg/kg body wt, i.p. (HgCl2 group). Control rats received the vehicle alone (Control group). Studies were carried out 18h after injection. Afterwards, the kidneys and livers were excised and processed for histopathological studies or immunoblot (homogenates and crude membranes) techniques. In rats treated with HgCl2, immunoblotting showed a significant decrease in the abundance of BTL in homogenates and plasma membranes from kidney and liver. BTL decrease of expression might reflect the grade of damage in renal tubule cells and in hepatocytes. Thus, BTL might be postulated as a new biomarker of tissue toxicity induced by mercury.
Collapse
Affiliation(s)
- Mara Soledad Trebucobich
- Area Farmacología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, CONICET, Rosario, Argentina
| | - María Herminia Hazelhoff
- Area Farmacología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, CONICET, Rosario, Argentina
| | - Alberto A Chevalier
- GIHON Laboratorios Quimicos SRL/Facultad de Ciencias Exactas, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | | | - Anabel Brandoni
- Area Farmacología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, CONICET, Rosario, Argentina
| | - Adriana Mónica Torres
- Area Farmacología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, CONICET, Rosario, Argentina.
| |
Collapse
|
240
|
Meyer Zu Schwabedissen HE, Begunk R, Hussner J, Juhnke BO, Gliesche D, Böttcher K, Sternberg K, Schmitz KP, Kroemer HK. Cell-Specific Expression of Uptake Transporters—A Potential Approach for Cardiovascular Drug Delivery Devices. Mol Pharm 2014; 11:665-72. [DOI: 10.1021/mp400245g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
241
|
Franconi F, Campesi I. Pharmacogenomics, pharmacokinetics and pharmacodynamics: interaction with biological differences between men and women. Br J Pharmacol 2014; 171:580-94. [PMID: 23981051 PMCID: PMC3969074 DOI: 10.1111/bph.12362] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 08/05/2013] [Accepted: 08/16/2013] [Indexed: 12/16/2022] Open
Abstract
Pharmacological response depends on multiple factors and one of them is sex-gender. Data on the specific effects of sex-gender on pharmacokinetics, as well as the safety and efficacy of numerous medications, are beginning to emerge. Nevertheless, the recruitment of women for clinical research is inadequate, especially during the first phases. In general, pharmacokinetic differences between males and females are more numerous and consistent than disparities in pharmacodynamics. However, sex-gender pharmacodynamic differences are now increasingly being identified at the molecular level. It is now even becoming apparent that sex-gender influences pharmacogenomics and pharmacogenetics. Sex-related differences have been reported for several parameters, and it is consistently shown that women have a worse safety profile, with drug adverse reactions being more frequent and severe in women than in men. Overall, the pharmacological status of women is less well studied than that of men and deserves much more attention. The design of clinical and preclinical studies should have a sex-gender-based approach with the aim of tailoring therapies to an individual's needs and concerns.
Collapse
Affiliation(s)
- Flavia Franconi
- Department of Biomedical Sciences, University of SassariSassari, Italy
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and BiosystemsOsilo, Italy
| | - Ilaria Campesi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and BiosystemsOsilo, Italy
- Department of Surgical, Microsurgical and Medical Sciences, University of SassariSassari, Italy
| |
Collapse
|
242
|
Macha S, Koenen R, Sennewald R, Schöne K, Hummel N, Riedmaier S, Woerle HJ, Salsali A, Broedl UC. Effect of Gemfibrozil, Rifampicin, or Probenecid on the Pharmacokinetics of the SGLT2 Inhibitor Empagliflozin in Healthy Volunteers. Clin Ther 2014; 36:280-90.e1. [DOI: 10.1016/j.clinthera.2014.01.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/20/2013] [Accepted: 01/07/2014] [Indexed: 11/25/2022]
|
243
|
Xu Q, Wang C, Meng Q, Liu Q, Sun P, Sun H, Guo X, Liu K. The oligopeptide transporter 2-mediated reabsorption of entecavir in rat kidney. Eur J Pharm Sci 2014; 52:41-7. [DOI: 10.1016/j.ejps.2013.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/16/2013] [Accepted: 10/16/2013] [Indexed: 11/17/2022]
|
244
|
Kell DB, Goodacre R. Metabolomics and systems pharmacology: why and how to model the human metabolic network for drug discovery. Drug Discov Today 2014; 19:171-82. [PMID: 23892182 PMCID: PMC3989035 DOI: 10.1016/j.drudis.2013.07.014] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 07/03/2013] [Accepted: 07/16/2013] [Indexed: 02/06/2023]
Abstract
Metabolism represents the 'sharp end' of systems biology, because changes in metabolite concentrations are necessarily amplified relative to changes in the transcriptome, proteome and enzyme activities, which can be modulated by drugs. To understand such behaviour, we therefore need (and increasingly have) reliable consensus (community) models of the human metabolic network that include the important transporters. Small molecule 'drug' transporters are in fact metabolite transporters, because drugs bear structural similarities to metabolites known from the network reconstructions and from measurements of the metabolome. Recon2 represents the present state-of-the-art human metabolic network reconstruction; it can predict inter alia: (i) the effects of inborn errors of metabolism; (ii) which metabolites are exometabolites, and (iii) how metabolism varies between tissues and cellular compartments. However, even these qualitative network models are not yet complete. As our understanding improves so do we recognise more clearly the need for a systems (poly)pharmacology.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK.
| | - Royston Goodacre
- School of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| |
Collapse
|
245
|
Döring B, Petzinger E. Phase 0 and phase III transport in various organs: combined concept of phases in xenobiotic transport and metabolism. Drug Metab Rev 2014; 46:261-82. [PMID: 24483608 DOI: 10.3109/03602532.2014.882353] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The historical phasing concept of drug metabolism and elimination was introduced to comprise the two phases of metabolism: phase I metabolism for oxidations, reductions and hydrolyses, and phase II metabolism for synthesis. With this concept, biological membrane barriers obstructing the accessibility of metabolism sites in the cells for drugs were not considered. The concept of two phases was extended to a concept of four phases when drug transporters were detected that guided drugs and drug metabolites in and out of the cells. In particular, water soluble or charged drugs are virtually not able to overcome the phospholipid membrane barrier. Drug transporters belong to two main clusters of transporter families: the solute carrier (SLC) families and the ATP binding cassette (ABC) carriers. The ABC transporters comprise seven families with about 20 carriers involved in drug transport. All of them operate as pumps at the expense of ATP splitting. Embedded in the former phase concept, the term "phase III" was introduced by Ishikawa in 1992 for drug export by ABC efflux pumps. SLC comprise 52 families, from which many carriers are drug uptake transporters. Later on, this uptake process was referred to as the "phase 0 transport" of drugs. Transporters for xenobiotics in man and animal are most expressed in liver, but they are also present in extra-hepatic tissues such as in the kidney, the adrenal gland and lung. This review deals with the function of drug carriers in various organs and their impact on drug metabolism and elimination.
Collapse
Affiliation(s)
- Barbara Döring
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg, Justus-Liebig-University Giessen , Giessen , Germany
| | | |
Collapse
|
246
|
Soars MG, Barton P, Elkin LL, Mosure KW, Sproston JL, Riley RJ. Application of anin vitroOAT assay in drug design and optimization of renal clearance. Xenobiotica 2014; 44:657-65. [DOI: 10.3109/00498254.2013.879625] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
247
|
Fisel P, Renner O, Nies AT, Schwab M, Schaeffeler E. Solute carrier transporter and drug-related nephrotoxicity: the impact of proximal tubule cell models for preclinical research. Expert Opin Drug Metab Toxicol 2014; 10:395-408. [PMID: 24397389 DOI: 10.1517/17425255.2014.876990] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The final excretion step of several drugs is facilitated by membrane transporters of the Solute carrier (SLC) family expressed in the proximal tubules of the kidney. Membrane transporters contribute substantially to the pharmacokinetic profile of drugs and play important roles in drug-induced nephrotoxicity. Different cell models have been applied as tools for the assessment of nephrotoxic effects caused by drugs. AREAS COVERED This review gives an overview over clinically relevant SLC transporters involved in the renal elimination of drug agents and their specific role in drug-induced nephrotoxicity. Most widely applied cell models are described and their advantages and limitations are outlined. EXPERT OPINION In vitro cell culture models (e.g., continuous and primary renal cell lines, polarized cell monolayers) represent valuable tools for early assessment of the nephrotoxic potential of drugs. Since SLC transporters contribute to drug excretion in a large part, in vitro cell culture models might be very helpful to study transport pathways and/or potential drug-drug interactions at an early stage of the drug development process to predict nephrotoxic effects.
Collapse
Affiliation(s)
- Pascale Fisel
- Margarete Fischer-Bosch-Institute of Clinical Pharmacology , Auerbachstrasse 125, Stuttgart, 70376 , Germany
| | | | | | | | | |
Collapse
|
248
|
Volk C. OCTs, OATs, and OCTNs: structure and function of the polyspecific organic ion transporters of the SLC22 family. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/wmts.100] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Christopher Volk
- Department of Natural Sciences; Bonn-Rhein-Sieg University of Applied Sciences; Rheinbach Germany
| |
Collapse
|
249
|
Heussner AH, Dietrich DR. Primary porcine proximal tubular cells as an alternative to human primary renal cells in vitro: an initial characterization. BMC Cell Biol 2013; 14:55. [PMID: 24308307 PMCID: PMC4234457 DOI: 10.1186/1471-2121-14-55] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 11/28/2013] [Indexed: 02/07/2023] Open
Abstract
Background A good in vitro model should approximate an in vivo-like behavior as closely as possible in order to reflect most likely the in vivo situation. Regarding renal physiology of different species, humans are more closely related to pigs than to rodents, therefore primary porcine kidney cells (PKC) and their subsequent cell strain could be a valid alternative to primary human cells for renal in vitro toxicology. For this PKC must display inherent characteristics (e.g. structural organization) and functions (e.g. transepithelial transport) as observed under in vivo conditions within the respective part of the kidney. Results We carried out a comprehensive characterization of PKC and their subsequent cell strain, including morphology and growth as well as transporter expression and functionality. The data presented here demonstrate that PKC express various transporters including pMrp1 (abcc1), pMrp2 (abcc2), pOat1 (slc22a6) and pOat3 (slc22a8), whereas pMdr1 (abcb1) and pOatp1a2 (slco1a2) mRNA could not be detected in either the PKCs or in the porcine cortical tissue. Functionality of the transporters was demonstrated by determining the specific PAH transport kinetics. Conclusions On the basis of the presented results it can be concluded that PKC and to some extent their subsequent cell strain represent a valuable model for in vitro toxicology, which might be used as an alternative to human primary cells.
Collapse
Affiliation(s)
| | - Daniel R Dietrich
- Human and Environmental Toxicology, University of Konstanz, 78457 Konstanz, Germany.
| |
Collapse
|
250
|
Inoue K, Yuasa H. Molecular basis for pharmacokinetics and pharmacodynamics of methotrexate in rheumatoid arthritis therapy. Drug Metab Pharmacokinet 2013; 29:12-9. [PMID: 24284432 DOI: 10.2133/dmpk.dmpk-13-rv-119] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Methotrexate (MTX) is a derivative of folic acid (folate) and commonly used as an anchor drug for the treatment of rheumatoid arthritis (RA). The pharmacokinetics (PK) and pharmacodynamics (PD) of MTX entirely depends on the function of specific transporters that belong to the two major superfamilies, solute carrier transporters and ATP-binding cassette transporters. Several transporters have been identified as being able to mediate the transport of MTX, and suggested to be involved in the disposition in the body and in the regulation of intracellular metabolism in target cells, together with several enzymes involved in folate metabolism. Thus, drug-drug interactions through the transporters and their genetic polymorphisms may alter the PK and PD of MTX, resulting in an interpatient variability of efficacy. This review summarizes the PK and PD of MTX, particularly in relation to RA therapy and focuses on the roles of transporters involved in PK and PD with the aim of facilitating an understanding of the molecular basis of the mechanism of MTX action to achieve its effective use in RA therapy.
Collapse
Affiliation(s)
- Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | | |
Collapse
|