101
|
Schweizer U, Köhrle J. Function of thyroid hormone transporters in the central nervous system. Biochim Biophys Acta Gen Subj 2012; 1830:3965-73. [PMID: 22890106 DOI: 10.1016/j.bbagen.2012.07.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 07/12/2012] [Accepted: 07/30/2012] [Indexed: 01/24/2023]
Abstract
BACKGROUND Iodothyronines are charged amino acid derivatives that cannot passively cross a phospholipid bilayer. Transport of thyroid hormones across plasma membranes is mediated by integral membrane proteins belonging to several gene families. These transporters therefore allow or limit access of thyroid hormones into brain. Since thyroid hormones are essential for brain development and cell differentiation, it is expected that genetic deficiency of such transporters would result in neurodevelopmental derangements. SCOPE OF REVIEW We introduce concepts of thyroid hormone transport into the brain and into brain cells. Important thyroid hormone transmembrane transporters are presented along with their expression patterns in different brain cell types. A focus is placed on monocarboxylate transporter 8 (MCT8) which has been identified as an essential thyroid hormone transporter in humans. Mutations in MCT8 underlie one of the first described X-linked mental retardation syndromes, the Allan-Herndon-Dudley syndrome. MAJOR CONCLUSIONS Thyroid hormone transporter molecules are expressed in a developmental and cell type-specific pattern. Any thyroid hormone molecule has to cross consecutively the luminal and abluminal membranes of the capillary endothelium, enter astrocytic foot processes, and leave the astrocyte through the plasma membrane to finally cross another plasma membrane on its way towards its target nucleus. GENERAL SIGNIFICANCE We can expect more transporters being involved in or contributing to in neurodevelopmental or neuropsychiatric disease. Due to their expression in cellular components regulating the hypothalamus-pituitary-thyroid axis, mutations and polymorphisms are expected to impact on negative feedback regulation and hormonal setpoints. This article is part of a Special Issue entitled Thyroid hormone signalling.
Collapse
Affiliation(s)
- Ulrich Schweizer
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | |
Collapse
|
102
|
Kunze A, Huwyler J, Camenisch G, Gutmann H. Interaction of the antiviral drug telaprevir with renal and hepatic drug transporters. Biochem Pharmacol 2012; 84:1096-102. [PMID: 22902721 DOI: 10.1016/j.bcp.2012.07.032] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 02/06/2023]
Abstract
Telaprevir is a new, direct-acting antiviral drug that has been approved for the treatment of chronic hepatitis C viral infection. First data on drug-drug interactions with co-medications such as cyclosporine, tacrolimus and atorvastatin have been reported recently. Drug transporting proteins have been shown to play an important role in clinically observed drug-drug interactions. The aim of this study was therefore to systematically investigate the potential of telaprevir to inhibit drug transporting proteins. The effect of telaprevir on substrate uptake mediated by drug transporters located in human kidney and liver was investigated on a functional level in HEK293 cell lines that over-express single transporter. Telaprevir was shown to exhibit significant inhibition of the human renal drug transporters OCT2 and MATE1 with IC(50) values of 6.4 μM and 23.0 μM, respectively, whereas no inhibitory effect on OAT1 and OAT3 mediated transport by telaprevir was demonstrated. Liver drug transporters were inhibited with an IC(50) of 2.2 μM for OATP1B1, 6.8 μM for OATP1B3 and 20.7 μM for OCT1. Our data show that telaprevir exhibited significant potential to inhibit human drug transporters. In view of the inhibitory potential of telaprevir, clinical co-administration of telaprevir together with drugs that are substrates of renal or hepatic transporters should be carefully monitored.
Collapse
Affiliation(s)
- Annett Kunze
- Division of Drug Metabolism and Pharmacokinetics, Drug-Drug Interactions Section, Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | | | | | | |
Collapse
|
103
|
Wu LX, Guo CX, Chen WQ, Yu J, Qu Q, Chen Y, Tan ZR, Wang G, Fan L, Li Q, Zhang W, Zhou HH. Inhibition of the organic anion-transporting polypeptide 1B1 by quercetin: an in vitro and in vivo assessment. Br J Clin Pharmacol 2012; 73:750-7. [PMID: 22114872 DOI: 10.1111/j.1365-2125.2011.04150.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
AIM To investigate the effect of quercetin on organic anion transporting polypeptide 1B1 (OATP1B1) activities in vitro and on the pharmacokinetics of pravastatin, a typical substrate for OATP1B1 in healthy Chinese-Han male subjects. METHODS Using human embryonic kidney 293 (HEK293) cells stably expressing OATP1B1, we observed the effect of quercetin on OATP1B1-mediated uptake of estrone-3-sulphate (E3S) and pravastatin. The influence of quercetin on the pharmacokinetics of pravastatin was measured in 16 healthy Chinese-Han male volunteers receiving a single dose of pravastatin (40 mg orally) after co-administration of placebo or 500 mg quercetin capsules (once daily orally for 14 days). RESULTS Quercetin competitively inhibited OATP1B1-mediated E3S uptake with a K(i) value of 17.9 ± 4.6 µm and also inhibited OATP1B1-mediated pravastatin uptake in a concentration dependent manner (IC(50) , 15.9 ± 1.4 µm). In healthy Chinese-Han male subjects, quercetin increased the pravastatin area under the plasma concentration - time curve (AUC(0,10 h) and the peak plasma drug concentration (C(max)) to 24% (95% CI 15, 32%, P < 0.001) and 31% (95% CI 20, 42%, P < 0.001), respectively. After administration of quercetin, the elimination half-life (t(1/2) ) of pravastatin was prolonged by 14% (95% CI 4, 24%, P = 0.027), with no change in the time to reach C(max) (t(max) ). Moreover, quercetin decreased the apparent clearance (CL/F) of pravastatin by 18% (95% CI 75, 89%, P < 0.001). CONCLUSIONS These findings suggest that quercetin inhibits the OATP1B1-mediated transport of E3S and pravastatin in vitro and also has a modest inhibitory influence on the pharmacokinetics of pravastatin in healthy Chinese-Han male volunteers. The effects of quercetin on other OATP1B1 substrate drugs deserve further investigation.
Collapse
Affiliation(s)
- Lan-Xiang Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Cheng Y, Wang G, Zhang W, Fan L, Chen Y, Zhou HH. Effect of CYP2C9 and SLCO1B1 polymorphisms on the pharmacokinetics and pharmacodynamics of nateglinide in healthy Chinese male volunteers. Eur J Clin Pharmacol 2012; 69:407-13. [PMID: 22842957 DOI: 10.1007/s00228-012-1364-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 07/15/2012] [Indexed: 01/12/2023]
Abstract
PURPOSE Nateglinide is commonly used in the treatment of patients with type 2 diabetes mellitus. Our objective was to assess the association between CYP2C9 and SLCO1B1 polymorphisms and the metabolism of nateglinide in healthy Chinese male volunteers. METHODS A total of 35 healthy Chinese male volunteers with different CYP2C9 and SLCO1B1 genotypes were given a single oral dose of 120 mg nateglinide. Plasma concentrations of nateglinide and blood glucose level were measured up to 8 h. RESULTS In subjects with the CYP2C9*1/*3 & 521TT, CYP2C9*1/*1 & 521TC/CC and CYP2C9*1/*3 & 521TC genotype, AUC(0-∞) of nateglinide was 56 %, 34 % and 56 % higher (P = 0.002, P = 0.041 and P = 0.013, respectively), and the CL/F of nateglinide was 35 %, 11 % and 36 % lower (P = 0.000, P = 0.003 and P = 0.002, respectively) than that in the reference group. When only considering 521 T>C polymorphism, it had no significant association with the pharmacokinetics of nateglinide. CYP2C9*3 and 521 T>C polymorphisms were the significant predictors of the AUC(0-∞) and CL/F of nateglinide (adjusted multiple R(2) = 34 % and 43 %, respectively) according to multiple linear regression analyses, but they have no significant association with changes in the blood glucose-lowering effect of nateglinide. CONCLUSIONS Both SLCO1B1 521 T>C and the CYP2C9*3 polymorphisms can significantly affect the pharmacokinetics of nateglinide, but they could only partially explain the interindividual variability of plasma concentration of nateglinide. Moreover, 521 T>C and the CYP2C9*3 polymorphisms have no effect on pharmacodynamics of nateglinide in healthy Chinese male subjects.
Collapse
Affiliation(s)
- Yu Cheng
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Central South University, Changsha, Hunan 410078, China
| | | | | | | | | | | |
Collapse
|
105
|
Coviello AD, Haring R, Wellons M, Vaidya D, Lehtimäki T, Keildson S, Lunetta KL, He C, Fornage M, Lagou V, Mangino M, Onland-Moret NC, Chen B, Eriksson J, Garcia M, Liu YM, Koster A, Lohman K, Lyytikäinen LP, Petersen AK, Prescott J, Stolk L, Vandenput L, Wood AR, Zhuang WV, Ruokonen A, Hartikainen AL, Pouta A, Bandinelli S, Biffar R, Brabant G, Cox DG, Chen Y, Cummings S, Ferrucci L, Gunter MJ, Hankinson SE, Martikainen H, Hofman A, Homuth G, Illig T, Jansson JO, Johnson AD, Karasik D, Karlsson M, Kettunen J, Kiel DP, Kraft P, Liu J, Ljunggren Ö, Lorentzon M, Maggio M, Markus MRP, Mellström D, Miljkovic I, Mirel D, Nelson S, Morin Papunen L, Peeters PHM, Prokopenko I, Raffel L, Reincke M, Reiner AP, Rexrode K, Rivadeneira F, Schwartz SM, Siscovick D, Soranzo N, Stöckl D, Tworoger S, Uitterlinden AG, van Gils CH, Vasan RS, Wichmann HE, Zhai G, Bhasin S, Bidlingmaier M, Chanock SJ, De Vivo I, Harris TB, Hunter DJ, Kähönen M, Liu S, Ouyang P, Spector TD, van der Schouw YT, Viikari J, Wallaschofski H, McCarthy MI, Frayling TM, Murray A, Franks S, Järvelin MR, de Jong FH, Raitakari O, Teumer A, Ohlsson C, Murabito JM, Perry JRB. A genome-wide association meta-analysis of circulating sex hormone-binding globulin reveals multiple Loci implicated in sex steroid hormone regulation. PLoS Genet 2012; 8:e1002805. [PMID: 22829776 PMCID: PMC3400553 DOI: 10.1371/journal.pgen.1002805] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 05/19/2012] [Indexed: 01/28/2023] Open
Abstract
Sex hormone-binding globulin (SHBG) is a glycoprotein responsible for the transport and biologic availability of sex steroid hormones, primarily testosterone and estradiol. SHBG has been associated with chronic diseases including type 2 diabetes (T2D) and with hormone-sensitive cancers such as breast and prostate cancer. We performed a genome-wide association study (GWAS) meta-analysis of 21,791 individuals from 10 epidemiologic studies and validated these findings in 7,046 individuals in an additional six studies. We identified twelve genomic regions (SNPs) associated with circulating SHBG concentrations. Loci near the identified SNPs included SHBG (rs12150660, 17p13.1, p = 1.8 × 10(-106)), PRMT6 (rs17496332, 1p13.3, p = 1.4 × 10(-11)), GCKR (rs780093, 2p23.3, p = 2.2 × 10(-16)), ZBTB10 (rs440837, 8q21.13, p = 3.4 × 10(-09)), JMJD1C (rs7910927, 10q21.3, p = 6.1 × 10(-35)), SLCO1B1 (rs4149056, 12p12.1, p = 1.9 × 10(-08)), NR2F2 (rs8023580, 15q26.2, p = 8.3 × 10(-12)), ZNF652 (rs2411984, 17q21.32, p = 3.5 × 10(-14)), TDGF3 (rs1573036, Xq22.3, p = 4.1 × 10(-14)), LHCGR (rs10454142, 2p16.3, p = 1.3 × 10(-07)), BAIAP2L1 (rs3779195, 7q21.3, p = 2.7 × 10(-08)), and UGT2B15 (rs293428, 4q13.2, p = 5.5 × 10(-06)). These genes encompass multiple biologic pathways, including hepatic function, lipid metabolism, carbohydrate metabolism and T2D, androgen and estrogen receptor function, epigenetic effects, and the biology of sex steroid hormone-responsive cancers including breast and prostate cancer. We found evidence of sex-differentiated genetic influences on SHBG. In a sex-specific GWAS, the loci 4q13.2-UGT2B15 was significant in men only (men p = 2.5 × 10(-08), women p = 0.66, heterogeneity p = 0.003). Additionally, three loci showed strong sex-differentiated effects: 17p13.1-SHBG and Xq22.3-TDGF3 were stronger in men, whereas 8q21.12-ZBTB10 was stronger in women. Conditional analyses identified additional signals at the SHBG gene that together almost double the proportion of variance explained at the locus. Using an independent study of 1,129 individuals, all SNPs identified in the overall or sex-differentiated or conditional analyses explained ~15.6% and ~8.4% of the genetic variation of SHBG concentrations in men and women, respectively. The evidence for sex-differentiated effects and allelic heterogeneity highlight the importance of considering these features when estimating complex trait variance.
Collapse
Affiliation(s)
- Andrea D. Coviello
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Section of Endocrinology, Diabetes, and Nutrition, Boston University School of Medicine, Boston, Massachusetts, United States of America
- National Heart, Lung, and Blood Institute's The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Robin Haring
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine, Ernst-Moritz-Arndt University of Greifswald, Greifswald, Germany
| | - Melissa Wellons
- Department of Medicine and Department of Obstetrics and Gynecology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Dhananjay Vaidya
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Sarah Keildson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Chunyan He
- Department of Public Health, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana, United States of America
| | - Myriam Fornage
- University of Texas Health Sciences Center at Houston, Houston, Texas, United States of America
| | - Vasiliki Lagou
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - N. Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brian Chen
- Program on Genomics and Nutrition and the Center for Metabolic Disease Prevention, School of Public Health, University of California Los Angeles, Los Angeles, California, United States of America
| | - Joel Eriksson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Melissa Garcia
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Yong Mei Liu
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
| | - Annemarie Koster
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Kurt Lohman
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Ann-Kristin Petersen
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jennifer Prescott
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Consortium of Healthy Aging, Rotterdam, The Netherlands
| | - Liesbeth Vandenput
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andrew R. Wood
- Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, United Kingdom
| | - Wei Vivian Zhuang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Aimo Ruokonen
- Institute of Diagnostics, University of Oulu, Oulu, Finland
| | | | - Anneli Pouta
- National Institute for Health and Welfare and Institute of Health Sciences, University of Oulu, Oulu, Finland
| | | | - Reiner Biffar
- Department of Prosthetic Dentistry, Gerostomatology, and Dental Materials, University of Greifswald, Greifswald, Germany
| | - Georg Brabant
- Experimental and Clinical Endocrinology, University of Lübeck, Lübeck, Germany
| | - David G. Cox
- Cancer Research Center of Lyon, INSERM U1052, Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
| | - Yuhui Chen
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Steven Cummings
- California Pacific Medical Center, San Francisco, California, United States of America
| | - Luigi Ferrucci
- Longitudinal Studies Section, Clinical Research Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Marc J. Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
| | - Susan E. Hankinson
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Massachusetts, United States of America
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Hannu Martikainen
- Department of Obstetrics and Gynecology, University Hospital of Oulu, Oulu, Finland
| | - Albert Hofman
- Netherlands Consortium of Healthy Aging, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - John-Olov Jansson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andrew D. Johnson
- National Heart, Lung, and Blood Institute's The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - David Karasik
- Hebrew SeniorLife Institute for Aging Research and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Clinical Sciences and Department of Orthopaedics, Lund University, Malmö, Sweden
| | - Johannes Kettunen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Douglas P. Kiel
- Hebrew SeniorLife Institute for Aging Research and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Jingmin Liu
- Women's Health Initiative Clinical Coordinating Center, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Östen Ljunggren
- Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Mattias Lorentzon
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcello Maggio
- Department of Internal Medicine and Biomedical Sciences, Section of Geriatrics, University of Parma, Parma, Italy
| | | | - Dan Mellström
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Iva Miljkovic
- University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Daniel Mirel
- Gene Environment Initiative, Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, Massachusetts, United States of America
| | - Sarah Nelson
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Laure Morin Papunen
- Department of Obstetrics and Gynecology, University Hospital of Oulu, Oulu, Finland
| | - Petra H. M. Peeters
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Leslie Raffel
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Martin Reincke
- Medizinische Klinik and Poliklinik IV, Ludwig-Maximilians University, Munich, Germany
| | - Alex P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Kathryn Rexrode
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Consortium of Healthy Aging, Rotterdam, The Netherlands
| | - Stephen M. Schwartz
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - David Siscovick
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Nicole Soranzo
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Doris Stöckl
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Obstetrics and Gynaecology, Ludwig-Maximilians-University, Munich, Germany
| | - Shelley Tworoger
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Consortium of Healthy Aging, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Carla H. van Gils
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ramachandran S. Vasan
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- National Heart, Lung, and Blood Institute's The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - H.-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Klinikum Großhadern, Munich, Germany
| | - Guangju Zhai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Shalender Bhasin
- Section of Endocrinology, Diabetes, and Nutrition, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Martin Bidlingmaier
- Medizinische Klinik and Poliklinik IV, Ludwig-Maximilians University, Munich, Germany
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Immaculata De Vivo
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - David J. Hunter
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Simin Liu
- Program on Genomics and Nutrition, Department of Epidemiology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Pamela Ouyang
- Division of Cardiology, Johns Hopkins Bayview Medical Center, Baltimore, Maryland, United States of America
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Yvonne T. van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jorma Viikari
- Department of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Henri Wallaschofski
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine, Ernst-Moritz-Arndt University of Greifswald, Greifswald, Germany
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Timothy M. Frayling
- Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, United Kingdom
| | - Anna Murray
- Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, United Kingdom
| | - Steve Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Marjo-Riitta Järvelin
- Department of Biostatistics and Epidemiology, School of Public Health, MRC-HPA Centre for Environment and Health, Faculty of Medicine, Imperial College London, London, United Kingdom
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- National Institute of Health and Welfare, University of Oulu, Oulu, Finland
| | - Frank H. de Jong
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Olli Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital and Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joanne M. Murabito
- National Heart, Lung, and Blood Institute's The Framingham Heart Study, Framingham, Massachusetts, United States of America
- Section of General Internal Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - John R. B. Perry
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
106
|
Karlgren M, Vildhede A, Norinder U, Wisniewski JR, Kimoto E, Lai Y, Haglund U, Artursson P. Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions. J Med Chem 2012; 55:4740-63. [PMID: 22541068 PMCID: PMC3361267 DOI: 10.1021/jm300212s] [Citation(s) in RCA: 267] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
The hepatic organic anion transporting polypeptides (OATPs)
influence the pharmacokinetics of several drug classes and are involved
in many clinical drug–drug interactions. Predicting potential
interactions with OATPs is, therefore, of value. Here, we developed
in vitro and in silico models for identification and prediction of
specific and general inhibitors of OATP1B1, OATP1B3, and OATP2B1.
The maximal transport activity (MTA) of each OATP in human liver was
predicted from transport kinetics and protein quantification. We then
used MTA to predict the effects of a subset of inhibitors on atorvastatin
uptake in vivo. Using a data set of 225 drug-like compounds, 91 OATP
inhibitors were identified. In silico models indicated that lipophilicity
and polar surface area are key molecular features of OATP inhibition.
MTA predictions identified OATP1B1 and OATP1B3 as major determinants
of atorvastatin uptake in vivo. The relative contributions to overall
hepatic uptake varied with isoform specificities of the inhibitors.
Collapse
Affiliation(s)
- Maria Karlgren
- Department of Pharmacy, Uppsala University, 751 23 Uppsala, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
107
|
Tamai I. Oral drug delivery utilizing intestinal OATP transporters. Adv Drug Deliv Rev 2012; 64:508-14. [PMID: 21824501 DOI: 10.1016/j.addr.2011.07.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/23/2011] [Accepted: 07/25/2011] [Indexed: 12/16/2022]
Abstract
Transporters play important roles in tissue distribution and urinary- and biliary-excretion of drugs and transporter molecules involved in those processes have been elucidated well. Furthermore, an involvement of efflux transporters such as P-glycoproteins, multidrug resistance associated protein 2, and breast cancer resistance protein as the intestinal absorption barrier and/or intestinal luminal secretion mechanisms has been demonstrated. However, although there are many suggestions for the contribution of uptake/influx transporters in intestinal absorption of drugs, information on the transporter molecules responsible for the intestinal absorptive process is limited. Among them, most studied absorptive drug transporter is peptide transporter PEPT1. However, utilization of PEPT1 for oral delivery of drugs may not be high due to the chemical structural requirement of PEPT1 limited to peptide-mimetics. Recently, organic anion transporting polypeptide (OATP) family such as OATP1A2 and OATP2B1 has been suggested to mediate intestinal absorption of several drugs. Since OATPs exhibit species difference in expressed tissues and functional properties between human and animals, human studies are essential to clarify the intestinal absorption mechanisms of drugs via OATPs. Recent pharmacogenomic studies demonstrated that OATP2B1 is involved in the drug absorption in human. In addition, information of drug-juice interaction in the intestine also uncovered the contribution of OATP1A2 and OATP2B1 in drug absorption. Since OATP1A2 and OATP2B1 exhibit broader substrate selectivity compared with PEPT1, their potential to be applied for oral delivery should be high. In this review, current understanding of characteristics and contribution as the absorptive transporters of OATPs in small intestine in human is described. Now, it is getting clearer that OATPs have significant roles in intestinal absorption of drugs, therefore, there are higher possibility to utilize OATPs as the tools for oral delivery.
Collapse
Affiliation(s)
- Ikumi Tamai
- Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan.
| |
Collapse
|
108
|
Salyers KL, Xu Y. Animal Models for Studying Drug Metabolizing Enzymes and Transporters. ADME‐ENABLING TECHNOLOGIES IN DRUG DESIGN AND DEVELOPMENT 2012:253-276. [DOI: 10.1002/9781118180778.ch16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
109
|
Sharma P, Butters CJ, Smith V, Elsby R, Surry D. Prediction of the in vivo OATP1B1-mediated drug-drug interaction potential of an investigational drug against a range of statins. Eur J Pharm Sci 2012; 47:244-55. [PMID: 22538052 DOI: 10.1016/j.ejps.2012.04.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 02/23/2012] [Accepted: 04/03/2012] [Indexed: 01/17/2023]
Abstract
To support drug development, the drug-drug interaction potential (DDI) of an investigational drug (AZX) was assessed against the probe estradiol 17β-glucuronide as well as against simvastatin acid, atorvastatin, pravastatin, pitavastatin, fluvastatin, rosuvastatin and estrone 3-sulfate. The inhibitory potentials of the OATP1B1 inhibitors rifamycin SV and gemfibrozil were assessed in parallel. Monolayer cellular uptake assays were used to determine inhibition of human OATP1B1. Apparent K(m) values for the OATP1B1-mediated transport of [(3)H] substrates were determined prior to their use as probes in inhibition studies, and ranged from 0.6 to 29 μM for statins. The K(m) of lipophilic simvastatin acid could not be determined due to its high passive permeability that masked OATP1B1 transport, and therefore this statin could not be used as a probe. Estrone 3-sulfate exhibited biphasic kinetics, whereas estradiol 17β-glucuronide demonstrated simple Michaelis-Menton kinetics. AZX moderately inhibited OATP1B1-mediated transport of all statins (IC(50)=4.6-9.7 μM), except fluvastatin, of estradiol 17β-glucuronide (IC(50)=5.3 μM), and weakly inhibited estrone 3-sulfate (IC(50)=79 μM). Rifamycin SV strongly, and gemfibrozil weakly, inhibited the OATP1B1-mediated transport of substrates. Estradiol 17β-glucuronide was identified as a good surrogate probe for statins when assessing OATP1B1 inhibitory potential using this test system. Inhibition data was used to predict the likelihood of a clinical DDI, using current draft US FDA guidance and recommendations of the International Transporter Consortium. Predictions for AZX indicated the potential for an OATP1B1-mediated DDI in vivo and that a clinical interaction study is warranted to confirm whether AZX is an OATP1B1 inhibitor in the clinic.
Collapse
Affiliation(s)
- Pradeep Sharma
- Global DMPK, AstraZeneca R&D Alderley Park, Mereside, Macclesfield, Cheshire SK10 4TG, UK.
| | | | | | | | | |
Collapse
|
110
|
Sprowl JA, Mikkelsen TS, Giovinazzo H, Sparreboom A. Contribution of tumoral and host solute carriers to clinical drug response. Drug Resist Updat 2012; 15:5-20. [PMID: 22459901 DOI: 10.1016/j.drup.2012.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Members of the solute carrier family of transporters are responsible for the cellular uptake of a broad range of endogenous compounds and xenobiotics in multiple tissues. Several of these solute carriers are known to be expressed in cancer cells or cancer cell lines, and decreased cellular uptake of drugs potentially contributes to the development of resistance. As result, the expression levels of these proteins in humans have important consequences for an individual's susceptibility to certain drug-induced side effects, interactions, and treatment efficacy. In this review article, we provide an update of this rapidly emerging field, with specific emphasis on the direct contribution of solute carriers to anticancer drug uptake in tumors, the role of these carriers in regulation of anticancer drug disposition, and recent advances in attempts to evaluate these proteins as therapeutic targets.
Collapse
Affiliation(s)
- Jason A Sprowl
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | |
Collapse
|
111
|
DeGorter MK, Ho RH, Leake BF, Tirona RG, Kim RB. Interaction of three regiospecific amino acid residues is required for OATP1B1 gain of OATP1B3 substrate specificity. Mol Pharm 2012; 9:986-95. [PMID: 22352740 DOI: 10.1021/mp200629s] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The human organic anion-transporting polypeptides OATP1B1 (SLCO1B1) and OATP1B3 (SLCO1B3) are liver-enriched membrane transporters of major importance to hepatic uptake of numerous endogenous compounds, including bile acids, steroid conjugates, hormones, and drugs, including the 3-hydroxy-3-methylglutaryl Co-A reductase inhibitor (statin) family of cholesterol-lowering compounds. Despite their remarkable substrate overlap, there are notable exceptions: in particular, the gastrointestinal peptide hormone cholecystokinin-8 (CCK-8) is a high affinity substrate for OATP1B3 but not OATP1B1. We utilized homologous recombination of linear DNA by E. coli to generate a library of cDNA containing monomer size chimeric OATP1B1-1B3 and OATP1B3-1B1 transporters with randomly distributed chimeric junctions to identify three discrete regions of the transporter involved in conferring CCK-8 transport activity. Site-directed mutagenesis of three key residues in OATP1B1 transmembrane helices 1 and 10, and extracellular loop 6, to the corresponding residues in OATP1B3, resulted in a gain of CCK-8 transport by OATP1B1. The residues appear specific to CCK-8, as the mutations did not affect transport of the shared OATP1B substrate atorvastatin or the OATP1B1-specific substrate estrone sulfate. Regions involved in gain of CCK-8 transport by OATP1B1, when mapped to the crystal structures of bacterial transporters from the major facilitator superfamily, are positioned to suggest these regions could readily interact with drug substrates. Accordingly, our data provide new insight into the molecular determinants of the substrate specificity of these hepatic uptake transporters with relevance to targeted drug design and prediction of drug-drug interactions.
Collapse
Affiliation(s)
- Marianne K DeGorter
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
112
|
Ueno Y, Matsuda H, Mizutani H, Iwamoto T, Okuda M. Involvement of Specific Transport System on Uptake of Lactone Form of SN-38 in Human Intestinal Epithelial Cell Line Caco-2. Biol Pharm Bull 2012; 35:54-8. [DOI: 10.1248/bpb.35.54] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yusuke Ueno
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
| | - Hiroko Matsuda
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
| | - Hideki Mizutani
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
- College of Pharmacy, Kinjo Gakuin University
| | - Takuya Iwamoto
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
| | - Masahiro Okuda
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
| |
Collapse
|
113
|
Yoshie S, Ito J, Shirasawa S, Yokoyama T, Fujimura Y, Takeda K, Mizuguchi M, Matsumoto K, Tomotsune D, Sasaki K. Establishment of Novel Detection System for Embryonic Stem Cell-Derived Hepatocyte-Like Cells Based on Nongenetic Manipulation with Indocyanine Green. Tissue Eng Part C Methods 2012; 18:12-20. [DOI: 10.1089/ten.tec.2011.0179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Susumu Yoshie
- Department of Histology and Embryology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Jun Ito
- Department of Histology and Embryology, Shinshu University School of Medicine, Matsumoto, Japan
| | | | | | - Yuu Fujimura
- On-chip Biotechnologies Co., Ltd., Koganei, Japan
| | - Kazuo Takeda
- On-chip Biotechnologies Co., Ltd., Koganei, Japan
| | | | | | - Daihachiro Tomotsune
- Department of Histology and Embryology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Katsunori Sasaki
- Department of Histology and Embryology, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
114
|
Shitara Y, Takeuchi K, Nagamatsu Y, Wada S, Sugiyama Y, Horie T. Long-lasting Inhibitory Effects of Cyclosporin A, but Not Tacrolimus, on OATP1B1- and OATP1B3-mediated Uptake. Drug Metab Pharmacokinet 2012; 27:368-78. [DOI: 10.2133/dmpk.dmpk-11-rg-096] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
115
|
Nakanishi T, Tamai I. Genetic polymorphisms of OATP transporters and their impact on intestinal absorption and hepatic disposition of drugs. Drug Metab Pharmacokinet 2011; 27:106-21. [PMID: 22185815 DOI: 10.2133/dmpk.dmpk-11-rv-099] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There is convincing evidence that many organic anion transporting polypeptide (OATP) transporters influence the pharmacokinetics and pharmacological efficacy of their substrate drugs. Each OATP family member has a unique combination of tissue distribution, substrate specificity and mechanisms of gene expression. Among them, OATP1B1, OATP1B3 and OATP2B1 have been considered as critical molecular determinants of the pharmacokinetics of a variety of clinically important drugs. Liver-specific expression of OATP1B1 and OATP1B3 contributes to the hepatic uptake of drugs from the portal vein, and OATP2B1 may alter their intestinal absorption as well as hepatic extraction. Accordingly, changes in function and expression of these three OATPs owing to genetic polymorphisms may lead to altered pharmacological effects, including decreased drug efficacy and increased risk of adverse effects. Association of genetic polymorphisms in OATP genes with alterations in the pharmacokinetic properties of their substrate drugs has been reported; however, there still exists a degree of discordance between the reported outcomes in different clinical settings. For better understanding of the clinical relevance of genetic polymorphisms of OATP1B1, OATP1B3 and OATP2B1, the present review focuses on the association of the genotypes of these OATPs with in vitro activity changes and in vivo clinical outcomes of substrate drugs.
Collapse
Affiliation(s)
- Takeo Nakanishi
- Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Pharmacy, Kanazawa University, Kanazawa, Japan
| | | |
Collapse
|
116
|
Aquilante CL, Kiser JJ, Anderson PL, Christians U, Kosmiski LA, Daily EB, Hoffman KL, Hopley CW, Predhomme JA, Schniedewind B, Sidhom MS. Influence of SLCO1B1 polymorphisms on the drug-drug interaction between darunavir/ritonavir and pravastatin. J Clin Pharmacol 2011; 52:1725-38. [PMID: 22174437 DOI: 10.1177/0091270011427907] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The authors investigated whether SLCO1B1 polymorphisms contribute to variability in pravastatin pharmacokinetics when pravastatin is administered alone versus with darunavir/ritonavir. HIV-negative healthy participants were prospectively enrolled on the basis of SLCO1B1 diplotype: group 1 (*1A/*1A, n = 9); group 2 (*1A/*1B, n = 10; or *1B/*1B, n = 2); and group 3 (*1A/*15, n = 1; *1B/*15, n = 5; or *1B/*17, n = 1). Participants received pravastatin (40 mg) daily on days 1 through 4, washout on days 5 through 11, darunavir/ritonavir (600/100 mg) twice daily on days 12 through 18, with pravastatin 40 mg added back on days 15 through 18. Pharmacokinetic studies were conducted on day 4 (pravastatin alone) and day 18 (pravastatin + darunavir/ritonavir). Pravastatin area under the plasma concentration-time curve (AUC(tau)) was 21% higher during administration with darunavir/ritonavir compared with pravastatin alone; however, this difference was not statistically significant (P = .11). Group 3 variants had 96% higher pravastatin AUC(tau) on day 4 and 113% higher pravastatin AUC(tau) on day 18 compared with group 1. The relative change in pravastatin pharmacokinetics was largest in group 3 but did not differ significantly between diplotype groups. In sum, the influence of SLCO1B1*15 and *17 haplotypes on pravastatin pharmacokinetics was maintained in the presence of darunavir/ritonavir. Because OATP1B1 inhibition would be expected to be greater in carriers of normal or high-functioning SLCO1B1 haplotypes, these findings suggest that darunavir/ritonavir is not a potent inhibitor of OATP1B1-mediated pravastatin transport in vivo.
Collapse
Affiliation(s)
- Christina L Aquilante
- PharmD, Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, 12850 East Montview Blvd, Mail Stop C238, Room V20-4103, Aurora, CO 80045, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Chae YJ, Lee KR, Noh CK, Chong S, Kim DD, Shim CK, Chung SJ. Functional consequences of genetic variations in the human organic anion transporting polypeptide 1B3 (OATP1B3) in the Korean population. J Pharm Sci 2011; 101:1302-13. [PMID: 22147445 DOI: 10.1002/jps.23005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 10/18/2011] [Accepted: 11/11/2011] [Indexed: 01/09/2023]
Abstract
The objectives of this study were to investigate the allele frequencies and linkage disequilibrium (LD) in the organic anion transporting polypeptide 1B3 (OATP1B3) in the Korean population and to examine the functional consequences. Using samples from 48 Koreans, direct sequencing was carried out to determine the allele frequencies and LD of OATP1B3 in a representative Korean population. Thirty-six genetic variations in the transporter were found in Koreans; among them, five undocumented variations (i.e.,-6436G>C in the 5'-upstream region, 26A>C and 586A>G in the protein coding region, and IVS6-72A>T and IVS12-80A>T in intron regions) were identified. In the upstream region, -5035G>A was found to have lowered gene expression, as determined by a reporter gene assay, suggesting that this variation reduces the expression of OATP1B3 in humans. The functional relevance of the genetic variations in the protein coding region was determined by an uptake study involving representative substrates in human embryonic kidney 293 cells expressing wild type or variant forms. Variations involving 699G>A showed a reduced uptake activity for testosterone, but not for estradiol 17β-d-glucuronide or methotrexate, indicating that the functional impact of the variations is substrate specific. Considering the kinetic relevance of OATP1B3, the functionally affected variations may be therapeutically important.
Collapse
Affiliation(s)
- Yoon-Jee Chae
- Department of Pharmaceutics, College of Pharmacy, Seoul National University, Gwanak-gu, Seoul 151-742, Korea
| | | | | | | | | | | | | |
Collapse
|
118
|
Choi CI, Lee YJ, Lee HI, Kim BH, Kim MJ, Jang CG, Bae JW, Lee SY. Effects of theSLCO1B1*15allele on the pharmacokinetics of pitavastatin. Xenobiotica 2011; 42:496-501. [DOI: 10.3109/00498254.2011.632030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
119
|
Takanohashi T, Kubo S, Arisaka H, Shinkai K, Ubukata K. Contribution of organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 to hepatic uptake of nateglinide, and the prediction of drug–drug interactions via these transporters. J Pharm Pharmacol 2011; 64:199-206. [DOI: 10.1111/j.2042-7158.2011.01389.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Abstract
Objectives
We have investigated the contributions of organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 to the hepatic uptake of nateglinide, and the possibility of drug–drug interactions via these transporters.
Methods
Uptake studies using transporter-expressing HEK293 cells and cryopreserved human hepatocytes were performed to examine the contributions of each transporter. Inhibition studies using cryopreserved human hepatocytes were performed to examine the possibility of drug–drug interactions.
Key findings
The rate of saturable hepatic uptake of nateglinide using human hepatocytes was 47.6%. A certain increase in uptake was observed in the examination using transporter-expressing HEK293 cells, indicating contributions of OATP1B1 and OATP1B3 to hepatic nateglinide uptake. The 50% inhibitory concentration (IC50) values of nateglinide using cryopreserved human hepatocytes for uptake of estrone 3-sulfate (substrate of OATP1B1), and cholecystokinin octapeptide (substrate of OATP1B3) were 168 and 17.4 µmol/l, respectively. Moreover, ciclosporin inhibited saturable hepatic uptake of nateglinide with an IC50 value of 6.05 µmol/l. The calculated 1 + Iin,max,u/IC50 values for inhibition of OATP1B1 and OATP1B3 by nateglinide, and the inhibition of saturable uptake of nateglinide by ciclosporin, were all close to 1, indicating a low clinical risk of drug–drug interaction with nateglinide taken up via OATP1B1 and OATP1B3.
Conclusions
OATP1B1 and OATP1B3 may have contributed to the hepatic uptake of nateglinide, but the possibility of drug–drug interactions appeared to be low.
Collapse
Affiliation(s)
- Toshiyuki Takanohashi
- Drug Metabolism and Pharmacokinetics, Development Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Co., Ltd., Kawasaki, Japan
| | - Satoru Kubo
- Drug Metabolism and Pharmacokinetics, Development Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Co., Ltd., Kawasaki, Japan
| | - Harumi Arisaka
- Drug Metabolism and Pharmacokinetics, Development Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Co., Ltd., Kawasaki, Japan
| | - Kenji Shinkai
- Drug Metabolism and Pharmacokinetics, Development Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Co., Ltd., Kawasaki, Japan
| | - Kazuyuki Ubukata
- Drug Metabolism and Pharmacokinetics, Development Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Co., Ltd., Kawasaki, Japan
| |
Collapse
|
120
|
Wu LX, Guo CX, Qu Q, Yu J, Chen WQ, Wang G, Fan L, Li Q, Zhang W, Zhou HH. Effects of natural products on the function of human organic anion transporting polypeptide 1B1. Xenobiotica 2011; 42:339-48. [DOI: 10.3109/00498254.2011.623796] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
121
|
Yamane T, Hanaoka K, Muramatsu Y, Tamura K, Adachi Y, Miyashita Y, Hirata Y, Nagano T. Method for Enhancing Cell Penetration of Gd3+-based MRI Contrast Agents by Conjugation with Hydrophobic Fluorescent Dyes. Bioconjug Chem 2011; 22:2227-36. [DOI: 10.1021/bc200127t] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Takehiro Yamane
- Graduate School of
Pharmaceutical
Sciences, The University of Tokyo, 7-3-1,
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- CREST, Japan Science and Technology Agency, 5, Sanbancho, Chiyoda-ku, Tokyo,
102-0075, Japan
| | - Kenjiro Hanaoka
- Graduate School of
Pharmaceutical
Sciences, The University of Tokyo, 7-3-1,
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- CREST, Japan Science and Technology Agency, 5, Sanbancho, Chiyoda-ku, Tokyo,
102-0075, Japan
| | - Yasuaki Muramatsu
- Graduate School of
Pharmaceutical
Sciences, The University of Tokyo, 7-3-1,
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- CREST, Japan Science and Technology Agency, 5, Sanbancho, Chiyoda-ku, Tokyo,
102-0075, Japan
| | - Keita Tamura
- Department
of Physiology, The University of Tokyo School of Medicine, 7-3-1,
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yusuke Adachi
- Department
of Physiology, The University of Tokyo School of Medicine, 7-3-1,
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasushi Miyashita
- Department
of Physiology, The University of Tokyo School of Medicine, 7-3-1,
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasunobu Hirata
- Department of Cardiovascular
Medicine, The University of Tokyo Hospital, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tetsuo Nagano
- Graduate School of
Pharmaceutical
Sciences, The University of Tokyo, 7-3-1,
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- CREST, Japan Science and Technology Agency, 5, Sanbancho, Chiyoda-ku, Tokyo,
102-0075, Japan
| |
Collapse
|
122
|
OATP1B1/1B3 activity in plated primary human hepatocytes over time in culture. Biochem Pharmacol 2011; 82:1219-26. [DOI: 10.1016/j.bcp.2011.07.076] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 07/07/2011] [Accepted: 07/07/2011] [Indexed: 11/19/2022]
|
123
|
Mikkelsen TS, Thorn CF, Yang JJ, Ulrich CM, French D, Zaza G, Dunnenberger HM, Marsh S, McLeod HL, Giacomini K, Becker ML, Gaedigk R, Leeder JS, Kager L, Relling MV, Evans W, Klein TE, Altman RB. PharmGKB summary: methotrexate pathway. Pharmacogenet Genomics 2011; 21:679-86. [PMID: 21317831 PMCID: PMC3139712 DOI: 10.1097/fpc.0b013e328343dd93] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Torben S. Mikkelsen
- Department of Pediatric Oncology and Hematology, Aarhus University Hospital, Skejby, Denmark
- St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Caroline F. Thorn
- Department of Genetics, Stanford University Medical Center, Stanford
| | - Jun J. Yang
- St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Cornelia M. Ulrich
- German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Deborah French
- St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Gianluigi Zaza
- St Jude Children’s Research Hospital, Memphis, Tennessee
| | | | - Sharon Marsh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Howard L. McLeod
- Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, North Carolina
| | | | - Mara L. Becker
- Division of Clinical Pharmacology and Medical Toxicology, Children’s Mercy Hospitals and Clinics, Kansas City, Missouri, USA
| | - Roger Gaedigk
- Division of Clinical Pharmacology and Medical Toxicology, Children’s Mercy Hospitals and Clinics, Kansas City, Missouri, USA
| | - James Steven Leeder
- Division of Clinical Pharmacology and Medical Toxicology, Children’s Mercy Hospitals and Clinics, Kansas City, Missouri, USA
| | - Leo Kager
- St Anna Children’s Hospital, Vienna, Austria
| | | | - William Evans
- St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Teri E. Klein
- Department of Genetics, Stanford University Medical Center, Stanford
| | - Russ B. Altman
- Department of Genetics, Stanford University Medical Center, Stanford
- Department of Bioengineering, Stanford University Medical Center, Stanford
| |
Collapse
|
124
|
Nakanishi T, Tamai I. Solute Carrier Transporters as Targets for Drug Delivery and Pharmacological Intervention for Chemotherapy. J Pharm Sci 2011; 100:3731-50. [DOI: 10.1002/jps.22576] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 03/29/2011] [Accepted: 03/31/2011] [Indexed: 01/11/2023]
|
125
|
The impact of pharmacogenetics of metabolic enzymes and transporters on the pharmacokinetics of telmisartan in healthy volunteers. Pharmacogenet Genomics 2011; 21:523-30. [DOI: 10.1097/fpc.0b013e3283482502] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
126
|
Obaidat A, Roth M, Hagenbuch B. The expression and function of organic anion transporting polypeptides in normal tissues and in cancer. Annu Rev Pharmacol Toxicol 2011. [PMID: 21854228 DOI: 10.1146/annurev‐pharmtox‐010510‐100556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organic anion transporting polypeptides (OATPs) are members of the SLCO gene superfamily of proteins. The 11 human OATPs are classified into 6 families and subfamilies on the basis of their amino acid sequence similarities. OATPs are expressed in several epithelial tissues throughout the body and transport mainly amphipathic molecules with molecular weights of more than 300 kDa. Members of the OATP1 and OATP2 families are functionally the best-characterized OATPs. Among these are the multispecific OATP1A2, OATP1B1, OATP1B3, and OATP2B1. They transport various endo- and xenobiotics, including hormones and their conjugates as well as numerous drugs such as several anticancer agents. Recent reports demonstrate that some OATPs are up- or downregulated in several cancers and that OATP expression might affect cancer development. On the basis of the findings summarized in this review, we propose that OATPs could be valuable targets for anticancer therapy.
Collapse
Affiliation(s)
- Amanda Obaidat
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas 66160, USA.
| | | | | |
Collapse
|
127
|
Karlgren M, Ahlin G, Bergström CAS, Svensson R, Palm J, Artursson P. In vitro and in silico strategies to identify OATP1B1 inhibitors and predict clinical drug-drug interactions. Pharm Res 2011; 29:411-26. [PMID: 21861202 PMCID: PMC3264873 DOI: 10.1007/s11095-011-0564-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 08/08/2011] [Indexed: 12/02/2022]
Abstract
Purpose To establish in vitro and in silico models that predict clinical drug–drug interactions (DDIs) with the OATP1B1 (SLCO1B1) transporter. Methods The inhibitory effect of 146 drugs and drug-like compounds on OATP1B1-mediated transport was studied in HEK293 cells. A computational model was developed to predict OATP1B1 inhibition. Concentration-dependent effects were investigated for six compounds; clinical DDIs were predicted by calculating change in exposure (i.e. R-values) in eight different ways. Results Sixty-five compounds were identified as OATP1B1 inhibitors at 20 μM. The computational model predicted the test set with 80% accuracy for inhibitors and 91% for non-inhibitors. In vitro–in vivo comparisons underscored the importance of using drugs with known clinical effects as references. Thus, reference drugs, cyclosporin A, gemfibrozil, and fenofibrate, provided an inhibition interval to which three antiviral drugs, atazanavir, lopinavir, and amprenavir, could be compared and their clinical DDIs with OATP1B1 classified. Conclusions Twenty-two new OATP1B1 inhibitors were identified, a predictive OATP1B1 inhibition in silico model was developed, and successful predictions of clinical DDIs were obtained with OATP1B1.
Collapse
Affiliation(s)
- Maria Karlgren
- Uppsala University Drug Optimization and Pharmaceutical Profiling Platform, Department of Pharmacy, Uppsala University, The Biomedical Centre, P.O. Box 580, 751 23, Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
128
|
Obaidat A, Roth M, Hagenbuch B. The expression and function of organic anion transporting polypeptides in normal tissues and in cancer. Annu Rev Pharmacol Toxicol 2011; 52:135-51. [PMID: 21854228 DOI: 10.1146/annurev-pharmtox-010510-100556] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Organic anion transporting polypeptides (OATPs) are members of the SLCO gene superfamily of proteins. The 11 human OATPs are classified into 6 families and subfamilies on the basis of their amino acid sequence similarities. OATPs are expressed in several epithelial tissues throughout the body and transport mainly amphipathic molecules with molecular weights of more than 300 kDa. Members of the OATP1 and OATP2 families are functionally the best-characterized OATPs. Among these are the multispecific OATP1A2, OATP1B1, OATP1B3, and OATP2B1. They transport various endo- and xenobiotics, including hormones and their conjugates as well as numerous drugs such as several anticancer agents. Recent reports demonstrate that some OATPs are up- or downregulated in several cancers and that OATP expression might affect cancer development. On the basis of the findings summarized in this review, we propose that OATPs could be valuable targets for anticancer therapy.
Collapse
Affiliation(s)
- Amanda Obaidat
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas 66160, USA.
| | | | | |
Collapse
|
129
|
Meyer zu Schwabedissen HE, Ware JA, Finkelstein D, Chaudhry AS, Lemay S, Leon-Ponte M, Strom SC, Zaher H, Schwarz UI, Freeman DJ, Schuetz EG, Tirona RG, Kim RB. Hepatic organic anion transporting polypeptide transporter and thyroid hormone receptor interplay determines cholesterol and glucose homeostasis. Hepatology 2011; 54:644-54. [PMID: 21538436 PMCID: PMC3164486 DOI: 10.1002/hep.24391] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 04/15/2011] [Indexed: 12/07/2022]
Abstract
UNLABELLED The role of organic anion transporting polypeptides (OATPs), particularly the members of OATP1B subfamily, in hepatocellular handling of endogenous and exogenous compounds is an important and emerging area of research. Using a mouse model lacking Slco1b2, the murine ortholog of the OATP1B subfamily, we have demonstrated previously that genetic ablation causes reduced hepatic clearance capacity for substrates. In this study, we focused on the physiological function of the hepatic OATP1B transporters. First, we studied the influence of the Oatp1b2 deletion on bile acid (BA) metabolism, showing that lack of the transporter results in a significantly reduced expression of Cyp7a1, the key enzyme of BA synthesis, resulting in elevated cholesterol levels after high dietary fat challenge. Furthermore, Slco1b2-/- mice exhibited delayed clearance after oral glucose challenge resulting from reduced hepatic glucose uptake. In addition to increased hepatic glycogen content, Slco1b2-/- mice exhibited reduced glucose output after pyruvate challenge. This is in accordance with reduced hepatic expression of phosphoenolpyruvate carboxykinase (PEPCK) in knockout mice. We show that this phenotype is due to the loss of liver-specific Oatp1b2-mediated hepatocellular thyroid hormone entry, which then leads to reduced transcriptional activation of target genes of hepatic thyroid hormone receptor (TR), including Cyp7a1 and Pepck but also Dio1 and Glut2. Importantly, we assessed human relevance using a cohort of archived human livers in which OATP1B1 expression was noted to be highly associated with TR target genes, especially for glucose facilitating transporter 2 (GLUT2). Furthermore, GLUT2 expression was significantly decreased in livers harboring a common genetic polymorphism in SLCO1B1. CONCLUSION Our findings reveal that OATP1B-mediated hepatic thyroid hormone entry is a key determinant of cholesterol and glucose homeostasis.
Collapse
Affiliation(s)
| | - Joseph A. Ware
- Pfizer Global Research and Development, Ann Arbor, MI, USA
| | - David Finkelstein
- Information Sciences Department, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Amarjit S. Chaudhry
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sara Lemay
- Division of Clinical Pharmacology, Department of Medicine, The University of Western Ontario, London, ON, Canada
| | - Matilde Leon-Ponte
- Division of Clinical Pharmacology, Department of Medicine, The University of Western Ontario, London, ON, Canada
| | - Stephen C. Strom
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hani Zaher
- Pfizer Global Research and Development, Ann Arbor, MI, USA
| | - Ute I. Schwarz
- Division of Clinical Pharmacology, Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - David J. Freeman
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - Erin G. Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Rommel G. Tirona
- Division of Clinical Pharmacology, Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - Richard B. Kim
- Division of Clinical Pharmacology, Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
130
|
Kullak-Ublick GA, Ismair MG, Kubitz R, Schmitt M, Häussinger D, Stieger B, Hagenbuch B, Meier PJ, Beuers U, Paumgartner G. Stable expression and functional characterization of a Na+-taurocholate cotransporting green fluorescent protein in human hepatoblastoma HepG2 cells. Cytotechnology 2011; 34:1-9. [PMID: 19003375 DOI: 10.1023/a:1008152729133] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Sodium-dependent uptake of bile acids from blood is aliver-specific function which is mediated by theNa(+)-taurocholate cotransporting polypeptide(Ntcp). We report the stable expression of aNa(+)-taurocholate cotransporting green fluorescentfusion protein in the human hepatoblastoma cell lineHepG2, normally lacking Ntcp expression. Ntcp-EGFPassociated green fluorescence colocalized with Ntcpimmunofluorescence in the plasma membrane. Intransfected HepG2 cells, the fusion protein mediatedthe sodium-dependent uptake of the bile acidtaurocholate (K(m): 24.6 mumol/l) and of the anionicsteroids estrone-3-sulfate and dehydroepiandrosteronesulfate. We conclude that the Ntcp-EGFP fusion proteinfollows the sorting route of Ntcp, is functionallyidentical to Ntcp and could be used to monitor proteintrafficking in living HepG2 cells.
Collapse
Affiliation(s)
- G A Kullak-Ublick
- Divisions of Clinical Pharmacology/Toxicology, Department of Medicine, University Hospital, CH-8091, Zürich, Switzerland,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Determination of OATP-, NTCP- and OCT-mediated substrate uptake activities in individual and pooled batches of cryopreserved human hepatocytes. Eur J Pharm Sci 2011; 43:297-307. [DOI: 10.1016/j.ejps.2011.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 04/01/2011] [Accepted: 05/07/2011] [Indexed: 01/11/2023]
|
132
|
Kindla J, Müller F, Mieth M, Fromm MF, König J. Influence of non-steroidal anti-inflammatory drugs on organic anion transporting polypeptide (OATP) 1B1- and OATP1B3-mediated drug transport. Drug Metab Dispos 2011; 39:1047-53. [PMID: 21389119 DOI: 10.1124/dmd.110.037622] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The transporter-mediated uptake of drugs from blood into hepatocytes is a prerequisite for intrahepatic drug action or intracellular drug metabolism before excretion. Therefore, uptake transporters, e.g., members of the organic anion transporting polypeptide (OATP) family are important determinants of drug pharmacokinetics. Highly and almost exclusively expressed in hepatocytes are the OATP family members OATP1B1 (SLCO1B1) and OATP1B3 (SLCO1B3). Drug substrates of OATP1B1 and OATP1B3 include antibiotics and HMG-CoA reductase inhibitors (statins). It has been demonstrated that administration of two or more drugs that are substrates for these hepatic uptake transporters may lead to transporter-mediated drug-drug interactions, resulting in altered transport kinetics for drug substrates. In this study we investigated whether non-steroidal anti-inflammatory drugs (NSAIDs) and paracetamol interact with OATP1B1 and OATP1B3 using the standard substrate BSP and the drug substrate pravastatin. Using human embryonic kidney cells stably expressing OATP1B1 or OATP1B3, we demonstrated that bromosulfophthalein uptake was inhibited by diclofenac, ibuprofen. and lumiracoxib. Of interest, pravastatin uptake was stimulated by these NSAIDs, and for ibuprofen we determined activation constants (EC₅₀ values) of 64.0 and 93.1 μM for OATP1B1- and OATP1B3-mediated uptake, respectively. Furthermore, we investigated whether NSAIDs were also substrates for OATP1B1 and OATP1B3 and demonstrated that only diclofenac was significantly transported by OATP1B3, whereas all other NSAIDs investigated were not substrates for these uptake transporters. These results demonstrated that drugs may interact with transport proteins by allosteric mechanisms without being substrates and, therefore, not only uptake inhibition but also allosteric-induced modulation of transport function may be an important mechanism in transporter-mediated drug-drug interactions.
Collapse
Affiliation(s)
- Juergen Kindla
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | |
Collapse
|
133
|
Abstract
The aim of this review is to provide useful information not only for studying the effect of OATP1B1 and/or BCRP gene mutation on pharmacokinetics of novle statins of pitavastatin and rosuvastatin but also for studying drug-drug interactions (DDI) between the novle statins and other substrates of OATP1B1 and/or BCRP. Intra- and inter-ethnic differences in pharmacokinetic profiles of clinically relevant drugs are important issues reported in many papers not only for scenes of appropriate drug used in clinical settings but also for those of the drug development. Pharmacogenomics is extremely useful for understanding these racial differences. Recent pharmacogenetics study have disclosed important roles of drug transporters in the pharmacokinetic (PK) profiles of some clinically relevant drugs. In this presentation, we introduce single nucleotide polymorphisms (SNPs) of OATP1B1 and BCRP and review the contribution of genetic polymorphisms of the transporters to the pharmacokinetics of dual substrates as pitavastatin and rosuvastatin from recent study. At the same time, the DDIs between pitavastatin or rosuvastatin and other drug have been extensively concerned because of inhibiting OATP1B1-mediated hepatic uptake or BCRP-mediated hepatic efflux of pitavastatin and rosuvastatin. This review summarized the current studies about the role of OATP1B1 and BCRP in DDIs between pitavastatin or rosuvastatin and other clinically relevant drugs. The role of OATP1B1 and BCRP gene mutation can affect the PK profiles of pitavastatin and rosuvastatin. The DDIs between the novle statins and other substrates of OATP1B1 or BCRP may occur and cause change in the pharmacokinetic of the novle statins.
Collapse
Affiliation(s)
- Wen Jin Hua
- Department of Pharmacy, First Affiliated Hospital of Nanchang University, Nanchang, China.
| | | | | |
Collapse
|
134
|
Huppertz A, Breuer J, Fels LM, Schultze-Mosgau M, Sutter G, Klein S, Frericks B, Hamm B, Wagner M. Evaluation of possible drug-drug interaction between gadoxetic acid and erythromycin as an inhibitor of organic anion transporting peptides (OATP). J Magn Reson Imaging 2011; 33:409-16. [PMID: 21274983 DOI: 10.1002/jmri.22458] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To evaluate if erythromycin compromises liver-specific enhancement of gadoxetic acid; both compounds competing in organic anion transporting peptides (OATP) -mediated hepatocytic uptake. MATERIALS AND METHODS The study was approved by institutional review board. Twelve healthy subjects (nine men, three woman; mean age, 38.7 years) were examined twice by MR imaging with prior administration of NaCl solution (placebo) or 1000 mg of erythromycin following a randomized sequence. Gadoxetic acid (0.025 mmol/kg body weight) was administered 15 min after the end of infusions. Pre- and 20 min postcontrast two-dimensional gradient-recalled-echo sequences were acquired. Relative enhancements of liver parenchyma and ratio of means were calculated from signal intensity measurements. Plasma levels of gadoxetic acid and erythromycin were determined and given in geometric means and coefficients of variation (CV). RESULTS Concentration of erythromycin directly after end of infusion was 13.9 mg/L (CV 14.9%). Gadolinium plasma concentrations 5 min after gadoxetic acid administration were 138.7 μmol/L (CV 20.4%) after erythromycin infusion and 129.6 μmol/L (CV 22.8%) after placebo. Mean relative enhancements of liver parenchyma were 88.1 (SD 24.9%) after erythromycin infusion and 92.6 (SD 17.9%) after placebo. Ratio of relative enhancements was 0.951 (95% confidence interval, 0.833; 1.061; statistically not significant). CONCLUSION Coadministration of erythromycin has no effect on gadoxetic acid enhanced liver MR imaging.
Collapse
Affiliation(s)
- Alexander Huppertz
- Charité-University Hospital Berlin, Charité Campus Mitte, Department of Radiology, Berlin, Germany; Imaging Science Institute Charité, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
135
|
A possible mechanism for 2,2',4,4',5,5'-hexachlorobiphenyl-mediated decrease in serum thyroxine level in mice. Toxicol Appl Pharmacol 2011; 254:48-55. [PMID: 21569787 DOI: 10.1016/j.taap.2011.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 04/23/2011] [Accepted: 04/25/2011] [Indexed: 11/22/2022]
Abstract
Serum total thyroxine (T₄) level was markedly decreased, without significant increases in the levels of hepatic T₄-UDP-glucuronosyltransferase (T₄-UGT) and serum thyroid-stimulating hormone, 3 days after treatment with 2,2',4,4',5,5'-hexachlorobiphenyl (CB153) (100mg/kg, ip) in both 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-sensitive C57BL/6 and TCDD-resistant DBA/2 mice. Likewise, in either strain of mice, no CB153-mediated changes in the binding levels of [(125)I]T₄ to serum proteins, such as transthyretin, albumin, and thyroxine binding globulin, were observed, while in CB153-pretreated C57BL/6 mice, but not in CB153-pretreated DBA/2 mice, the levels of biliary [(125)I]₄T and [(125)I]T₄-glucuronide at 90-120 min after injection of [(125)I]T₄ slightly increased, as compared with those in the corresponding control mice. Concerning tissue distribution of [(125)I]T₄, liver-selective increases in the [(125)I]T₄ accumulation by CB153-pretreatment were observed in both C57BL/6 and DBA/2 mice, and the hepatic levels of [(125)I]T₄ in the C57BL/6 and DBA/2 mice became more than 44% and 34% of the [(125)I]T₄ dosed, respectively. The present findings indicated that the CB153-mediated decreases in the level of serum total T₄in C57BL/6 and DBA/2 mice occur mainly through an increase in the accumulation of T₄ in the liver.
Collapse
|
136
|
Roth M, Timmermann BN, Hagenbuch B. Interactions of green tea catechins with organic anion-transporting polypeptides. Drug Metab Dispos 2011; 39:920-6. [PMID: 21278283 PMCID: PMC3082372 DOI: 10.1124/dmd.110.036640] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 01/28/2011] [Indexed: 01/08/2023] Open
Abstract
Organic anion-transporting polypeptides (OATPs) are multispecific transporters that mediate the uptake of numerous drugs and xenobiotics into cells. Here, we examined the effect of green tea (Camellia sinensis) catechins on the function of the four OATPs expressed in human enterocytes and hepatocytes. Uptake of the model substrate estrone-3-sulfate by cells expressing OATP1A2, OATP1B1, OATP1B3, or OATP2B1 was measured in the absence and presence of the four most abundant flavonols found in green tea. Uptake by OATP1A2, OATP1B1, and OATP2B1 was inhibited by epicatechin gallate (ECG) and epigallocatechin gallate (EGCG) in a concentration-dependent way. In contrast, OATP1B3-mediated uptake of estrone-3-sulfate was strongly stimulated by EGCG at low substrate concentrations. The effect of EGCG on OATP1B3 was also studied with additional substrates: uptake of estradiol-17β-glucuronide was unchanged, whereas uptake of Fluo-3 was noncompetitively inhibited. Both ECG and EGCG were found to be substrates of OATP1A2 (K(m) values of 10.4 and 18.8 μM, respectively) and OATP1B3 (34.1 and 13.2 μM, respectively) but not of OATP1B1 or OATP2B1. These results indicate that two of the major flavonols found in green tea have a substantial effect on the function of OATPs expressed in enterocytes and hepatocytes and can potentially alter the pharmacokinetics of drugs and other OATP substrates. In addition, the diverse effects of EGCG on the transport of other OATP1B3 substrates suggest that different transport/binding sites are involved.
Collapse
Affiliation(s)
- Megan Roth
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | | | | |
Collapse
|
137
|
Kalliokoski A, Neuvonen PJ, Niemi M. SLCO1B1 polymorphism and oral antidiabetic drugs. Basic Clin Pharmacol Toxicol 2011; 107:775-81. [PMID: 20406215 DOI: 10.1111/j.1742-7843.2010.00581.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Organic anion-transporting polypeptide 1B1 (OATP1B1; gene: SLCO1B1) is an influx transporter expressed on the sinusoidal membrane of human hepatocytes, where it mediates the uptake of its substrates from blood into liver. In vitro, the SLCO1B1 c.521T>C (p.Val174Ala) single-nucleotide polymorphism (SNP) has been associated with reduced and the c.388A>G (p.Asn130Asp) SNP with both enhanced and reduced transport activity of OATP1B1. In vivo in humans, the c.521C allele (present in SLCO1B1*5 and *15 haplotypes) is associated with decreased hepatic uptake and increased plasma concentrations of several OATP1B1 substrates. The SLCO1B1*1B (c.388G-c.521T) haplotype is associated with enhanced hepatic uptake and decreased plasma concentrations of some OATP1B1 substrates. The SLCO1B1 c.521CC genotype has been associated with an about 60-190% increased, and the SLCO1B1*1B/*1B genotype with an about 30% decreased area under the plasma concentration-time curve of repaglinide. Moreover, SLCO1B1 polymorphism can affect the extent of interaction between OATP1B1 inhibitors and repaglinide. Accordingly, SLCO1B1 genotyping may help in choosing the optimal starting dose of repaglinide. In Chinese individuals, the SLCO1B1 c.521C allele has been associated with increased plasma concentrations of nateglinide, but the association could not be replicated in Caucasians. SLCO1B1 genotype has had no effect on the pharmacokinetics of rosiglitazone, pioglitazone or their metabolites. The hepatic uptake of metformin is mediated by organic cation transporters 1 and 3, and the liver is not important for the elimination or action of the dipeptidylpeptidase 4 inhibitors sitagliptin, vildagliptin and saxagliptin. Therefore, SLCO1B1 polymorphism unlikely affects the response to these antidiabetics. Possible effects of SLCO1B1 polymorphism on sulfonylureas remain to be investigated.
Collapse
|
138
|
Niemi M, Pasanen MK, Neuvonen PJ. Organic anion transporting polypeptide 1B1: a genetically polymorphic transporter of major importance for hepatic drug uptake. Pharmacol Rev 2011; 63:157-81. [PMID: 21245207 DOI: 10.1124/pr.110.002857] [Citation(s) in RCA: 481] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The importance of membrane transporters for drug pharmacokinetics has been increasingly recognized during the last decade. Organic anion transporting polypeptide 1B1 (OATP1B1) is a genetically polymorphic influx transporter expressed on the sinusoidal membrane of human hepatocytes, and it mediates the hepatic uptake of many endogenous compounds and xenobiotics. Recent studies have demonstrated that OATP1B1 plays a major, clinically important role in the hepatic uptake of many drugs. A common single-nucleotide variation (coding DNA c.521T>C, protein p.V174A, rs4149056) in the SLCO1B1 gene encoding OATP1B1 decreases the transporting activity of OATP1B1, resulting in markedly increased plasma concentrations of, for example, many statins, particularly of active simvastatin acid. The variant thereby enhances the risk of statin-induced myopathy and decreases the therapeutic indexes of statins. However, the effect of the SLCO1B1 c.521T>C variant is different on different statins. The same variant also markedly affects the pharmacokinetics of several other drugs. Furthermore, certain SLCO1B1 variants associated with an enhanced clearance of methotrexate increase the risk of gastrointestinal toxicity by methotrexate in the treatment of children with acute lymphoblastic leukemia. Certain drugs (e.g., cyclosporine) potently inhibit OATP1B1, causing clinically significant drug interactions. Thus, OATP1B1 plays a major role in the hepatic uptake of drugs, and genetic variants and drug interactions affecting OATP1B1 activity are important determinants of individual drug responses. In this article, we review the current knowledge about the expression, function, substrate characteristics, and pharmacogenetics of OATP1B1 as well as its role in drug interactions, in parts comparing with those of other hepatocyte-expressed organic anion transporting polypeptides, OATP1B3 and OATP2B1.
Collapse
Affiliation(s)
- Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, PO Box 20, Helsinki, FI-00014, Finland.
| | | | | |
Collapse
|
139
|
Yamaguchi H, Takeuchi T, Okada M, Kobayashi M, Unno M, Abe T, Goto J, Hishinuma T, Shimada M, Mano N. Screening of Antibiotics That Interact with Organic Anion-Transporting Polypeptides 1B1 and 1B3 Using Fluorescent Probes. Biol Pharm Bull 2011; 34:389-95. [DOI: 10.1248/bpb.34.389] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital
- Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Toshiko Takeuchi
- Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Masahiro Okada
- Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Minako Kobayashi
- Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Michiaki Unno
- Division of Gastroenterological Surgery, Department of Surgery, Tohoku University Graduate School of Medicine
| | - Takaaki Abe
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Biomedical Engineering
| | - Junichi Goto
- Department of Pharmaceutical Sciences, Tohoku University Hospital
| | - Takanori Hishinuma
- Division of Pharmacotherapy, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Miki Shimada
- Department of Pharmaceutical Sciences, Tohoku University Hospital
- Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital
- Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
140
|
Matsui O, Kitao A, Kobayashi S, Sanada J, Ryu Y, Minami T, Kozaka K, Shinnmura R, Nakamura K, Yoneda N, Koda W, Gabata T. Imaging of multi-step hepatocarcinogenesis: imaging, pathophysiologic and molecular correlation. ACTA ACUST UNITED AC 2011. [DOI: 10.2957/kanzo.52.415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
141
|
König J. Uptake transporters of the human OATP family: molecular characteristics, substrates, their role in drug-drug interactions, and functional consequences of polymorphisms. Handb Exp Pharmacol 2011:1-28. [PMID: 21103967 DOI: 10.1007/978-3-642-14541-4_1] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Organic anion transporting polypeptides (OATPs, gene family: SLC21/SLCO) mediate the uptake of a broad range of substrates including several widely prescribed drugs into cells. Drug substrates for members of the human OATP family include HMG-CoA-reductase inhibitors (statins), antibiotics, anticancer agents, and cardiac glycosides. OATPs are expressed in a variety of different tissues including brain, intestine, liver, and kidney, suggesting that these uptake transporters are important for drug absorption, distribution, and excretion. Because of their wide tissue distribution and broad substrate spectrum, altered transport kinetics, for example, due to drug-drug interactions or due to the functional consequences of genetic variations (polymorphisms), can contribute to the interindividual variability of drug effects. Therefore, the molecular characteristics of human OATP family members, the role of human OATPs in drug-drug interactions, and the in vitro analysis of the functional consequences of genetic variations in SLCO genes encoding OATP proteins are the focus of this chapter.
Collapse
Affiliation(s)
- Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
142
|
Lin R, Wang X, Zhou W, Fu W, Wang Y, Huang W, Jin L. Association of Polymorphisms in the Solute Carrier Organic Anion Transporter Family Member 1B1 Gene with Essential Hypertension in the Uyghur Population. Ann Hum Genet 2010; 75:305-11. [DOI: 10.1111/j.1469-1809.2010.00622.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
143
|
Sugiura T, Otake T, Shimizu T, Wakayama T, Silver DL, Utsumi R, Nishimura T, Iseki S, Nakamichi N, Kubo Y, Tsuji A, Kato Y. PDZK1 regulates organic anion transporting polypeptide Oatp1a in mouse small intestine. Drug Metab Pharmacokinet 2010; 25:588-98. [PMID: 21084765 DOI: 10.2133/dmpk.dmpk-10-rg-074] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recent studies indicate that various members of the organic anion transporting polypeptide (OATP) family are expressed on apical membranes of the small intestine. In the present study, we investigated possible interaction of Oatp with the PDZ protein PDZK1 in mouse small intestine, using [³H]estrone-3-sulfate (E3S) as a typical substrate. After intraduodenal administration, the level of [³H]E3S appearing in the portal vein of pdzk1 gene knockout (pdzk1(-/-)) mice was much lower than that in wild-type mice. Lower intestinal absorption of [³H]E3S in pdzk1(-/-) mice was confirmed in Ussing-type chamber experiments, which showed smaller uptake of [³H]E3S from the apical side in intestinal tissues of pdzk1(-/-) mice compared with wild-type mice. The kinetics and inhibition profile of [³H]E3S uptake in the Ussing-type chamber were similar to those in HEK293 cells stably expressing Oatp1a5, suggesting involvement of Oatp1a5 in [³H]E3S uptake. Immunoreactivity to anti-Oatp1a antibody was colocalized with PDZK1 in the small intestine of wild-type mice, whereas apical localization of Oatp1a protein was reduced in pdzk1(-/-) mice. An immunoprecipitation study revealed physical interaction of PDZK1 with Oatp1a. Thus, PDZK1 appears to act as an adaptor for Oatp1a. This is the first demonstration of a regulatory protein directly interacting with small-intestinal OATP.
Collapse
Affiliation(s)
- Tomoko Sugiura
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Meyer Zu Schwabedissen HE, Böttcher K, Chaudhry A, Kroemer HK, Schuetz EG, Kim RB. Liver X receptor α and farnesoid X receptor are major transcriptional regulators of OATP1B1. Hepatology 2010; 52:1797-807. [PMID: 20827719 DOI: 10.1002/hep.23876] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
UNLABELLED Organic anion transporting polypeptide 1B1 (OATP1B1) is a liver-enriched transporter involved in the hepatocellular uptake of many endogenous molecules and several structurally divergent drugs in clinical use. Although OATP1B1 coding region polymorphisms are known to make an impact on substrate drug disposition in humans, little is known regarding the mechanisms underlying the transcriptional regulation of this transporter. In this study, we note that messenger RNA (mRNA) expression of OATP1B1 in a large human liver bank exhibited marked interindividual variability that was not associated with coding region polymorphisms. Accordingly, we hypothesized that such variability in expression is reflective of nuclear receptor-mediated transcriptional regulation of this transporter. We tested prototypical ligands for the nuclear receptors pregnane X receptor (PXR), constitutive androstane receptor (CAR), liver X receptor (LXR) α, and farnesoid X receptor (FXR) in a human hepatoma-derived cell line and noted induction of OATP1B1 mRNA when the cells were treated with LXRα or FXR ligands. To confirm a direct role for LXRα and FXR to OATP1B1 expression, we performed detailed promoter analysis and cell-based reporter gene assays resulting in the identification of two functional FXR response elements and one LXRα response element. The direct interaction between nuclear receptors with the identified response elements was assessed using chromatin immunoprecipitation assays. Using isolated primary human hepatocytes, we show that LXRα or FXR agonists, but not PXR or CAR agonists, are capable of OATP1B1 induction. CONCLUSION We note that OATP1B1 transcriptional regulation is under dual nuclear receptor control through the oxysterol sensing LXRα and the bile acid sensor FXR. Accordingly, the interplay between OATP1B1 and nuclear receptors may play an important and heretofore unrecognized role during cholestasis, drug-induced liver injury, and OATP1B1 induction-related drug interactions.
Collapse
|
145
|
Abe M, Toyohara T, Ishii A, Suzuki T, Noguchi N, Akiyama Y, Shiwaku HO, Nakagomi-Hagihara R, Zheng G, Shibata E, Souma T, Shindo T, Shima H, Takeuchi Y, Mishima E, Tanemoto M, Terasaki T, Onogawa T, Unno M, Ito S, Takasawa S, Abe T. The HMG-CoA reductase inhibitor pravastatin stimulates insulin secretion through organic anion transporter polypeptides. Drug Metab Pharmacokinet 2010; 25:274-82. [PMID: 20610886 DOI: 10.2133/dmpk.25.274] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor pravastatin has been reported to have a beneficial effect on reducing the new onset of diabetes as well as lowering plasma lipids. Because pravastatin is a water-soluble organic anion, it cannot easily penetrate the lipid bilayer of the cell membrane. As the precise mechanisms of the effect of pravastatin on glucose metabolism and diabetes have not been clarified, we examined the roles of the organic anion transporter family on pravastatin-treated islet and adipocyte functions. Rat oatp1/slco1a1, oatp2/slco1a4 and oatp3/slco1a5 were expressed in the pancreas, and rat oatp3/slco1a5 was also detected in rat insulinoma cell line INS-1e. Pravastatin was transported not only by oatp1/slco1a1 and oatp2/slco1a4, but also by rat oatp3/slco1a5. Pravastatin uptake into INS-1e cells was detected and this transport was inhibited by sulfobromophthalein and rifampicin, both of which are known to inhibit oatp family-mediated uptake. In addition, pravastatin enhanced the glucose-stimulated insulin secretion from INS-1e cells. When fat-loaded db/db mice were treated with pravastatin, glucose intolerance and insulin resistance were prevented. In addition, insulin secretion from isolated islets was enhanced by pravastatin. These data suggest that pravastatin has pleiotropic effects on islets through membrane transport under high fat/glucose conditions.
Collapse
Affiliation(s)
- Michiaki Abe
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Fahrmayr C, Fromm MF, König J. Hepatic OATP and OCT uptake transporters: their role for drug-drug interactions and pharmacogenetic aspects. Drug Metab Rev 2010; 42:380-401. [PMID: 20100011 DOI: 10.3109/03602530903491683] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Uptake transporters in the basolateral membrane of hepatocytes are important for the hepatobiliary elimination of drugs. Further, since drug-metabolizing enzymes are located intracellularly, uptake into hepatocytes is a prerequisite for their subsequent metabolism. Therefore, alteration of uptake transporter function (e.g., by concomitantly administered drugs or due to functional consequences of genetic variations, leading to reduced transport function) may result in a change in drug pharmacokinetics. In this review, we focus on the hepatocellularly expressed members of the OATP and OCT family, their impact on transport-mediated drug-drug interactions, and on the functional consequences of variations in genes encoding these transporters.
Collapse
Affiliation(s)
- Christina Fahrmayr
- Department of Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | | | | |
Collapse
|
147
|
Suzuki T, Ohmuro A, Miyata M, Furuishi T, Hidaka S, Kugawa F, Fukami T, Tomono K. Involvement of an influx transporter in the blood-brain barrier transport of naloxone. Biopharm Drug Dispos 2010; 31:243-52. [PMID: 20437463 DOI: 10.1002/bdd.707] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Naloxone, a potent and specific opioid antagonist, has been shown in previous studies to have an influx clearance across the rat blood-brain barrier (BBB) two times greater than the efflux clearance. The purpose of the present study was to characterize the influx transport of naloxone across the rat BBB using the brain uptake index (BUI) method. The initial uptake rate of [(3)H]naloxone exhibited saturability in a concentration-dependent manner (concentration range 0.5 microM to 15 mM) in the presence of unlabeled naloxone. These results indicate that both passive diffusion and a carrier-mediated transport mechanism are operating. The in vivo kinetic parameters were estimated as follows: the Michaelis constant, K(t), was 2.99+/-0.71 mM; the maximum uptake rate, J(max), was 0.477+/-0.083 micromol/min/g brain; and the nonsaturable first-order rate constant, K(d), was 0.160+/-0.044 ml/min/g brain. The uptake of [(3)H]naloxone by the rat brain increased as the pH of the injected solution was increased from 5.5 to 8.5 and was strongly inhibited by cationic H(1)-antagonists such as pyrilamine and diphenhydramine and cationic drugs such as lidocaine and propranolol. In contrast, the BBB transport of [(3)H]naloxone was not affected by any typical substrates for organic cation transport systems such as tetraethylammonium, ergothioneine or L-carnitine or substrates for organic anion transport systems such as p-aminohippuric acid, benzylpenicillin or pravastatin. The present results suggest that a pH-dependent and saturable influx transport system that is a selective transporter for cationic H(1)-antagonists is involved in the BBB transport of naloxone in the rat.
Collapse
Affiliation(s)
- Toyofumi Suzuki
- Department of Pharmaceutics, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba, Japan
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Weaver YM, Hagenbuch B. Several conserved positively charged amino acids in OATP1B1 are involved in binding or translocation of different substrates. J Membr Biol 2010; 236:279-90. [PMID: 20821001 PMCID: PMC2989658 DOI: 10.1007/s00232-010-9300-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 08/16/2010] [Indexed: 12/20/2022]
Abstract
OATP1B1 and 1B3 are related transporters mediating uptake of numerous compounds into hepatocytes. A putative model of OATP1B3 with a "positive binding pocket" containing conserved positively charged amino acids was predicted (Meier-Abt et al. J Membr Biol 208:213-227, 2005). Based on this model, we tested the hypothesis that these positive amino acids are important for OATP1B1 function. We made mutants and measured surface expression and uptake of estradiol-17β-glucuronide, estrone-3-sulfate and bromosulfophthalein in HEK293 cells. Two of the mutants had low surface expression levels: R181K at 10% and R580A at 30% of wild-type OATP1B1. A lysine at position 580 (R580K) rescued the expression of R580A. Mutations of several amino acids resulted in substrate-dependent effects. The largest changes were seen for estradiol-17β-glucuronide, while estrone-3-sulfate and bromosulfophthalein transport were less affected. The wild-type OATP1B1 K (m) value for estradiol-17β-glucuronide of 5.35 ± 0.54 μM was increased by R57A to 30.5 ± 3.64 μM and decreased by R580K to 0.52 ± 0.18 μM. For estrone-3-sulfate the wild-type high-affinity K (m) value of 0.55 ± 0.12 μM was increased by K361R to 1.8 ± 0.47 μM and decreased by R580K to 0.1 ± 0.04 μM. In addition, R580K reduced the V (max) values for all three substrates to <25% of wild-type OATP1B1. Mutations at intracellular K90, H92 and R93 mainly affected V (max) values for estradiol-17β-glucuronide uptake. In conclusion, the conserved amino acids R57, K361 and R580 seem to be part of the substrate binding sites and/or translocation pathways in OATP1B1.
Collapse
Affiliation(s)
- Yi M. Weaver
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas 66160
- The University of Kansas Cancer Center, Kansas City, Kansas 66160
| |
Collapse
|
149
|
Kitao A, Zen Y, Matsui O, Gabata T, Kobayashi S, Koda W, Kozaka K, Yoneda N, Yamashita T, Kaneko S, Nakanuma Y. Hepatocellular carcinoma: signal intensity at gadoxetic acid-enhanced MR Imaging--correlation with molecular transporters and histopathologic features. Radiology 2010; 256:817-26. [PMID: 20663969 DOI: 10.1148/radiol.10092214] [Citation(s) in RCA: 277] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To analyze the correlation between signal intensity in the hepatobiliary phase of gadoxetic acid-enhanced magnetic resonance (MR) imaging and the expression of hepatocyte transporters with histopathologic features in hepatocellular carcinoma (HCC). MATERIALS AND METHODS Institutional ethics committee approval and informed consent were obtained. Forty surgically resected HCCs were classified as hypointense (n = 32) or iso- or hyperintense (n = 8) on the basis of findings in the hepatobiliary phase of gadoxetic acid-enhanced MR imaging. The following were compared between hypointense and iso- or hyperintense HCCs: the time-signal intensity curves at gadoxetic acid-enhanced MR imaging, the expression levels of seven transporters (four organic anion-transporting polypeptides [OATPs] and three multidrug-resistant proteins [MRPs]) at polymerase chain reaction (PCR) (for 22 nodules), results of immunostaining of OATP8, and histologic features. Statistical analysis (unpaired t test, Mann-Whitney test, chi(2) test, and Fisher exact test) was performed for each result. RESULTS On the time-signal intensity curves, hypointense HCCs showed a decreasing pattern, whereas iso- or hyperintense HCCs showed an increasing pattern after the dynamic phase. PCR revealed that expression of OATP8 (an uptake transporter) in hypointense HCCs was lower and that in iso- or hyperintense HCCs was higher than in background liver (P < .001). The expression level of MRP3 (a sinusoidal export transporter) showed a similar trend to that of OATP8 (P < .001). Immunostaining revealed that OATP8 expression was weak in hypointense HCCs, whereas it was sustained in iso- or hyperintense HCCs (P < .001). At histologic examination, a pseudoglandular proliferation pattern with bile plugs was more commonly observed in iso- or hyperintense HCCs than in hypointense HCCs (P = .01 for proliferation patterns and P = .006 for bile plugs). CONCLUSION The enhancement ratio of HCCs in the hepatobiliary phase of gadoxetic acid-enhanced MR imaging positively correlated with expression levels of OATP8 and MRP3, indicating that gadoxetic acid is taken up by OATP8 and excreted by MRP3.
Collapse
Affiliation(s)
- Azusa Kitao
- Department of Radiology, Human Pathology, and Gastroenterology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi, Kanazawa 920-8640, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Richardson TA, Klaassen CD. Disruption of thyroid hormone homeostasis in Ugt1a-deficient Gunn rats by microsomal enzyme inducers is not due to enhanced thyroxine glucuronidation. Toxicol Appl Pharmacol 2010; 248:38-44. [PMID: 20655938 DOI: 10.1016/j.taap.2010.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 07/10/2010] [Accepted: 07/14/2010] [Indexed: 11/25/2022]
Abstract
Microsomal enzyme inducers (MEI) that increase UDP-glucuronosyltransferases (UGTs) are thought to increase glucuronidation of thyroxine (T(4)), thus reducing serum T(4), and subsequently increasing thyroid stimulating hormone (TSH). Ugt1a1 and Ugt1a6 mediate T(4) glucuronidation. Therefore, this experiment determined the involvement of Ugt1a enzymes in increased T(4) glucuronidation, decreased serum T(4), and increased TSH after MEI treatment. Male Wistar and Ugt1a-deficient Wistar (Gunn) rats were fed a control diet or diet containing pregnenolone-16α-carbonitrile (PCN; 800 ppm), 3-methylcholanthrene (3-MC; 200 ppm), or Aroclor 1254 (PCB; 100 ppm) for 7 days. Serum T(4), triiodothyronine (T(3)), and TSH concentrations, hepatic T(4)/T(3) glucuronidation, and thyroid histology and follicular cell proliferation were investigated. PCN, 3-MC, and PCB treatments decreased serum T(4), whereas serum T(3) was maintained in both Gunn and Wistar rats (except for PCB treatment). TSH was increased in Wistar and Gunn rats after PCN (130 and 277%) or PCB treatment (72 and 60%). T(4) glucuronidation in Wistar rats was increased after PCN (298%), 3-MC (85%), and PCB (450%), but was extremely low in Gunn rats, and unchanged after MEI. T(3) glucuronidation was increased after PCN (121%) or PCB (58%) in Wistar rats, but only PCN increased T(3) glucuronidation in Gunn rats (43%). PCN treatment induced thyroid morphological changes and increased follicular cell proliferation in both strains. These data demonstrate that T(4) glucuronidation cannot be increased in Ugt1a-deficient Gunn rats. Thus, the decrease in serum T(4), increase in TSH, and increase in thyroid cell proliferation after MEI are not dependent on increased T(4) glucuronidation, and cannot be attributed to Ugt1a enzymes.
Collapse
Affiliation(s)
- Terrilyn A Richardson
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | |
Collapse
|