1
|
Ryu S, Yamaguchi E, Sadegh Modaresi SM, Agudelo J, Costales C, West MA, Fischer F, Slitt AL. Evaluation of 14 PFAS for permeability and organic anion transporter interactions: Implications for renal clearance in humans. CHEMOSPHERE 2024; 361:142390. [PMID: 38801906 DOI: 10.1016/j.chemosphere.2024.142390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/26/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) encompass a diverse group of synthetic fluorinated chemicals known to elicit adverse health effects in animals and humans. However, only a few studies investigated the mechanisms underlying clearance of PFAS. Herein, the relevance of human renal transporters and permeability to clearance and bioaccumulation for 14 PFAS containing three to eleven perfluorinated carbon atoms (ηpfc = 3-11) and several functional head-groups was investigated. Apparent permeabilities and interactions with human transporters were measured using in vitro cell-based assays, including the MDCK-LE cell line, and HEK293 stable transfected cell lines expressing organic anion transporter (OAT) 1-4 and organic cation transporter (OCT) 2. The results generated align with the Extended Clearance Classification System (ECCS), affirming that permeability, molecular weight, and ionization serve as robust predictors of clearance and renal transporter engagement. Notably, PFAS with low permeability (ECCS 3A and 3B) exhibited substantial substrate activity for OAT1 and OAT3, indicative of active renal secretion. Furthermore, we highlight the potential contribution of OAT4-mediated reabsorption to the renal clearance of PFAS with short ηpfc, such as perfluorohexane sulfonate (PFHxS). Our data advance our mechanistic understanding of renal clearance of PFAS in humans, provide useful input parameters for toxicokinetic models, and have broad implications for toxicological evaluation and regulatory considerations.
Collapse
Affiliation(s)
- Sangwoo Ryu
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, United States; Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Pfizer Inc., Groton, CT, 06340, United States
| | - Emi Yamaguchi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Pfizer Inc., Groton, CT, 06340, United States
| | - Seyed Mohamad Sadegh Modaresi
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, United States
| | - Juliana Agudelo
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, United States
| | - Chester Costales
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Pfizer Inc., Groton, CT, 06340, United States
| | - Mark A West
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Pfizer Inc., Groton, CT, 06340, United States
| | - Fabian Fischer
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, United States; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, United States.
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, United States.
| |
Collapse
|
2
|
Vávra J, Pavelcová K, Mašínová J, Hasíková L, Bubeníková E, Urbanová A, Mančíková A, Stibůrková B. Examining the Association of Rare Allelic Variants in Urate Transporters SLC22A11, SLC22A13, and SLC17A1 with Hyperuricemia and Gout. DISEASE MARKERS 2024; 2024:5930566. [PMID: 38222853 PMCID: PMC10787658 DOI: 10.1155/2024/5930566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/28/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024]
Abstract
Genetic variations in urate transporters play a significant role in determining human urate levels and have been implicated in developing hyperuricemia or gout. Polymorphism in the key urate transporters, such as ABCG2, URAT1, or GLUT9 was well-documented in the literature. Therefore in this study, our objective was to determine the frequency and effect of rare nonsynonymous allelic variants of SLC22A11, SLC22A13, and SLC17A1 on urate transport. In a cohort of 150 Czech patients with primary hyperuricemia and gout, we examined all coding regions and exon-intron boundaries of SLC22A11, SLC22A13, and SLC17A1 using PCR amplification and Sanger sequencing. For comparison, we used a control group consisting of 115 normouricemic subjects. To examine the effects of the rare allelic nonsynonymous variants on the expression, intracellular processing, and urate transporter protein function, we performed a functional characterization using the HEK293A cell line, immunoblotting, fluorescent microscopy, and site directed mutagenesis for preparing variants in vitro. Variants p.V202M (rs201209258), p.R343L (rs75933978), and p.P519L (rs144573306) were identified in the SLC22A11 gene (OAT4 transporter); variants p.R16H (rs72542450), and p.R102H (rs113229654) in the SLC22A13 gene (OAT10 transporter); and the p.W75C variant in the SLC17A1 gene (NPT1 transporter). All variants minimally affected protein levels and cytoplasmic/plasma membrane localization. The functional in vitro assay revealed that contrary to the native proteins, variants p.P519L in OAT4 (p ≤ 0.05), p.R16H in OAT10 (p ≤ 0.05), and p.W75C in the NPT1 transporter (p ≤ 0.01) significantly limited urate transport activity. Our findings contribute to a better understanding of (1) the risk of urate transporter-related hyperuricemia/gout and (2) uric acid handling in the kidneys.
Collapse
Affiliation(s)
- Jiří Vávra
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | | | | | | | - Eliška Bubeníková
- Institute of Rheumatology, Prague, Czech Republic
- Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Aneta Urbanová
- 1st Department of Medicine, Department of Hematology; First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Andrea Mančíková
- Department of Staphylococcal and Food-Borne Bacterial Infections, The National Institute of Public Health, Prague, Czech Republic
| | - Blanka Stibůrková
- Institute of Rheumatology, Prague, Czech Republic
- Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
3
|
Jilek JL, Frost KL, Marie S, Myers CM, Goedken M, Wright SH, Cherrington NJ. Attenuated Ochratoxin A Transporter Expression in a Mouse Model of Nonalcoholic Steatohepatitis Protects against Proximal Convoluted Tubule Toxicity. Drug Metab Dispos 2022; 50:1389-1395. [PMID: 34921099 PMCID: PMC9513848 DOI: 10.1124/dmd.121.000451] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 12/16/2021] [Indexed: 12/16/2022] Open
Abstract
Ochratoxin A (OTA) is an abundant mycotoxin, yet the toxicological impact of its disposition is not well studied. OTA is an organic anion transporter (OAT) substrate primarily excreted in urine despite a long half-life and extensive protein binding. Altered renal transporter expression during disease, including nonalcoholic steatohepatitis (NASH), may influence response to OTA exposure, but the impact of NASH on OTA toxicokinetics, tissue distribution, and associated nephrotoxicity is unknown. By inducing NASH in fast food-dieted/thioacetamide-exposed mice, we evaluated the effect of NASH on a bolus OTA exposure (12.5 mg/kg by mouth) after 3 days. NASH mice presented with less gross toxicity (44% less body weight loss), and kidney and liver weights of NASH mice were 11% and 24% higher, respectively, than healthy mice. Organ and body weight changes coincided with reduced renal proximal tubule cells vacuolation, degeneration, and necrosis, though no OTA-induced hepatic lesions were found. OTA systemic exposure in NASH mice increased modestly from 5.65 ± 1.10 to 7.95 ± 0.61 mg*h/ml per kg BW, and renal excretion increased robustly from 5.55% ± 0.37% to 13.11% ± 3.10%, relative to healthy mice. Total urinary excretion of OTA increased from 24.41 ± 1.74 to 40.07 ± 9.19 µg in NASH mice, and kidney-bound OTA decreased by ∼30%. Renal OAT isoform expression (OAT1-5) in NASH mice decreased by ∼50% with reduced OTA uptake by proximal convoluted cells. These data suggest that NASH-induced OAT transporter reductions attenuate renal secretion and reabsorption of OTA, increasing OTA urinary excretion and reducing renal exposure, thereby reducing nephrotoxicity in NASH. SIGNIFICANCE STATEMENT: These data suggest a disease-mediated transporter mechanism of altered tissue-specific toxicity after mycotoxin exposure, despite minimal systemic changes to ochratoxin A (OTA) concentrations. Further studies are warranted to evaluate the clinical relevance of this functional model and the potential effect of human nonalcoholic steatohepatitis on OTA and other organic anion substrate toxicity.
Collapse
Affiliation(s)
- Joseph L Jilek
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson, Arizona (J.L.J., K.L.F., S.M., C.M.M., N.J.C.); Rutgers Translational Sciences, Rutgers University, Piscataway, New Jersey (M.G.); and Department of Physiology, University of Arizona, College of Medicine, Tucson, Arizona (S.H.W.)
| | - Kayla L Frost
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson, Arizona (J.L.J., K.L.F., S.M., C.M.M., N.J.C.); Rutgers Translational Sciences, Rutgers University, Piscataway, New Jersey (M.G.); and Department of Physiology, University of Arizona, College of Medicine, Tucson, Arizona (S.H.W.)
| | - Solène Marie
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson, Arizona (J.L.J., K.L.F., S.M., C.M.M., N.J.C.); Rutgers Translational Sciences, Rutgers University, Piscataway, New Jersey (M.G.); and Department of Physiology, University of Arizona, College of Medicine, Tucson, Arizona (S.H.W.)
| | - Cassandra M Myers
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson, Arizona (J.L.J., K.L.F., S.M., C.M.M., N.J.C.); Rutgers Translational Sciences, Rutgers University, Piscataway, New Jersey (M.G.); and Department of Physiology, University of Arizona, College of Medicine, Tucson, Arizona (S.H.W.)
| | - Michael Goedken
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson, Arizona (J.L.J., K.L.F., S.M., C.M.M., N.J.C.); Rutgers Translational Sciences, Rutgers University, Piscataway, New Jersey (M.G.); and Department of Physiology, University of Arizona, College of Medicine, Tucson, Arizona (S.H.W.)
| | - Stephen H Wright
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson, Arizona (J.L.J., K.L.F., S.M., C.M.M., N.J.C.); Rutgers Translational Sciences, Rutgers University, Piscataway, New Jersey (M.G.); and Department of Physiology, University of Arizona, College of Medicine, Tucson, Arizona (S.H.W.)
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson, Arizona (J.L.J., K.L.F., S.M., C.M.M., N.J.C.); Rutgers Translational Sciences, Rutgers University, Piscataway, New Jersey (M.G.); and Department of Physiology, University of Arizona, College of Medicine, Tucson, Arizona (S.H.W.)
| |
Collapse
|
4
|
Yee SW, Stecula A, Chien HC, Zou L, Feofanova EV, van Borselen M, Cheung KWK, Yousri NA, Suhre K, Kinchen JM, Boerwinkle E, Irannejad R, Yu B, Giacomini KM. Unraveling the functional role of the orphan solute carrier, SLC22A24 in the transport of steroid conjugates through metabolomic and genome-wide association studies. PLoS Genet 2019; 15:e1008208. [PMID: 31553721 PMCID: PMC6760779 DOI: 10.1371/journal.pgen.1008208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/17/2019] [Indexed: 12/16/2022] Open
Abstract
Variation in steroid hormone levels has wide implications for health and disease. The genes encoding the proteins involved in steroid disposition represent key determinants of interindividual variation in steroid levels and ultimately, their effects. Beginning with metabolomic data from genome-wide association studies (GWAS), we observed that genetic variants in the orphan transporter, SLC22A24 were significantly associated with levels of androsterone glucuronide and etiocholanolone glucuronide (sentinel SNPs p-value <1x10-30). In cells over-expressing human or various mammalian orthologs of SLC22A24, we showed that steroid conjugates and bile acids were substrates of the transporter. Phylogenetic, genomic, and transcriptomic analyses suggested that SLC22A24 has a specialized role in the kidney and appears to function in the reabsorption of organic anions, and in particular, anionic steroids. Phenome-wide analysis showed that functional variants of SLC22A24 are associated with human disease such as cardiovascular diseases and acne, which have been linked to dysregulated steroid metabolism. Collectively, these functional genomic studies reveal a previously uncharacterized protein involved in steroid homeostasis, opening up new possibilities for SLC22A24 as a pharmacological target for regulating steroid levels.
Collapse
Affiliation(s)
- Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Adrian Stecula
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Ling Zou
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Elena V. Feofanova
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Marjolein van Borselen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Kit Wun Kathy Cheung
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Noha A. Yousri
- Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
- Computer and Systems Engineering, Alexandria University, Alexandria, Egypt
| | - Karsten Suhre
- Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Roshanak Irannejad
- The Cardiovascular Research Institute, University of California, San Francisco, California, United States of America
| | - Bing Yu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Kathleen M. Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, California, United States of America
| |
Collapse
|
5
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Nebbia CS, Oswald IP, Petersen A, Rose M, Roudot AC, Vleminckx C, Vollmer G, Wallace H, Bodin L, Cravedi JP, Halldorsson TI, Haug LS, Johansson N, van Loveren H, Gergelova P, Mackay K, Levorato S, van Manen M, Schwerdtle T. Risk to human health related to the presence of perfluorooctane sulfonic acid and perfluorooctanoic acid in food. EFSA J 2018. [PMID: 32625773 DOI: 10.2903/j.efsa.2018.5194">10.2903/j.efsa.2018.5194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [10.2903/j.efsa.2018.5194','32625773', '10.1152/ajprenal.00312.2010')">Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
10.2903/j.efsa.2018.5194" />
Abstract
The European Commission asked EFSA for a scientific evaluation on the risks to human health related to the presence of perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) in food. Regarding PFOS and PFOA occurrence, the final data set available for dietary exposure assessment contained a total of 20,019 analytical results (PFOS n = 10,191 and PFOA n = 9,828). There were large differences between upper and lower bound exposure due to analytical methods with insufficient sensitivity. The CONTAM Panel considered the lower bound estimates to be closer to true exposure levels. Important contributors to the lower bound mean chronic exposure were 'Fish and other seafood', 'Meat and meat products' and 'Eggs and egg products', for PFOS, and 'Milk and dairy products', 'Drinking water' and 'Fish and other seafood' for PFOA. PFOS and PFOA are readily absorbed in the gastrointestinal tract, excreted in urine and faeces, and do not undergo metabolism. Estimated human half-lives for PFOS and PFOA are about 5 years and 2-4 years, respectively. The derivation of a health-based guidance value was based on human epidemiological studies. For PFOS, the increase in serum total cholesterol in adults, and the decrease in antibody response at vaccination in children were identified as the critical effects. For PFOA, the increase in serum total cholesterol was the critical effect. Also reduced birth weight (for both compounds) and increased prevalence of high serum levels of the liver enzyme alanine aminotransferase (ALT) (for PFOA) were considered. After benchmark modelling of serum levels of PFOS and PFOA, and estimating the corresponding daily intakes, the CONTAM Panel established a tolerable weekly intake (TWI) of 13 ng/kg body weight (bw) per week for PFOS and 6 ng/kg bw per week for PFOA. For both compounds, exposure of a considerable proportion of the population exceeds the proposed TWIs.
Collapse
|
6
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Nebbia CS, Oswald IP, Petersen A, Rose M, Roudot AC, Vleminckx C, Vollmer G, Wallace H, Bodin L, Cravedi JP, Halldorsson TI, Haug LS, Johansson N, van Loveren H, Gergelova P, Mackay K, Levorato S, van Manen M, Schwerdtle T. Risk to human health related to the presence of perfluorooctane sulfonic acid and perfluorooctanoic acid in food. EFSA J 2018; 16:e05194. [PMID: 32625773 PMCID: PMC7009575 DOI: 10.2903/j.efsa.2018.5194] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The European Commission asked EFSA for a scientific evaluation on the risks to human health related to the presence of perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) in food. Regarding PFOS and PFOA occurrence, the final data set available for dietary exposure assessment contained a total of 20,019 analytical results (PFOS n = 10,191 and PFOA n = 9,828). There were large differences between upper and lower bound exposure due to analytical methods with insufficient sensitivity. The CONTAM Panel considered the lower bound estimates to be closer to true exposure levels. Important contributors to the lower bound mean chronic exposure were 'Fish and other seafood', 'Meat and meat products' and 'Eggs and egg products', for PFOS, and 'Milk and dairy products', 'Drinking water' and 'Fish and other seafood' for PFOA. PFOS and PFOA are readily absorbed in the gastrointestinal tract, excreted in urine and faeces, and do not undergo metabolism. Estimated human half-lives for PFOS and PFOA are about 5 years and 2-4 years, respectively. The derivation of a health-based guidance value was based on human epidemiological studies. For PFOS, the increase in serum total cholesterol in adults, and the decrease in antibody response at vaccination in children were identified as the critical effects. For PFOA, the increase in serum total cholesterol was the critical effect. Also reduced birth weight (for both compounds) and increased prevalence of high serum levels of the liver enzyme alanine aminotransferase (ALT) (for PFOA) were considered. After benchmark modelling of serum levels of PFOS and PFOA, and estimating the corresponding daily intakes, the CONTAM Panel established a tolerable weekly intake (TWI) of 13 ng/kg body weight (bw) per week for PFOS and 6 ng/kg bw per week for PFOA. For both compounds, exposure of a considerable proportion of the population exceeds the proposed TWIs.
Collapse
|
7
|
Zou L, Stecula A, Gupta A, Prasad B, Chien HC, Yee SW, Wang L, Unadkat JD, Stahl SH, Fenner KS, Giacomini KM. Molecular Mechanisms for Species Differences in Organic Anion Transporter 1, OAT1: Implications for Renal Drug Toxicity. Mol Pharmacol 2018; 94:689-699. [PMID: 29720497 DOI: 10.1124/mol.117.111153] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/25/2018] [Indexed: 12/31/2022] Open
Abstract
Species differences in renal drug transporters continue to plague drug development with animal models failing to adequately predict renal drug toxicity. For example, adefovir, a renally excreted antiviral drug, failed clinical studies for human immunodeficiency virus due to pronounced nephrotoxicity in humans. In this study, we demonstrated that there are large species differences in the kinetics of interactions of a key class of antiviral drugs, acyclic nucleoside phosphonates (ANPs), with organic anion transporter 1 [(OAT1) SLC22A6] and identified a key amino acid residue responsible for these differences. In OAT1 stably transfected human embryonic kidney 293 cells, the Km value of tenofovir for human OAT1 (hOAT1) was significantly lower than for OAT1 orthologs from common preclinical animals, including cynomolgus monkey, mouse, rat, and dog. Chimeric and site-directed mutagenesis studies along with comparative structure modeling identified serine at position 203 (S203) in hOAT1 as a determinant of its lower Km value. Furthermore, S203 is conserved in apes, and in contrast alanine at the equivalent position is conserved in preclinical animals and Old World monkeys, the most related primates to apes. Intriguingly, transport efficiencies are significantly higher for OAT1 orthologs from apes with high serum uric acid (SUA) levels than for the orthologs from species with low serum uric acid levels. In conclusion, our data provide a molecular mechanism underlying species differences in renal accumulation of nephrotoxic ANPs and a novel insight into OAT1 transport function in primate evolution.
Collapse
Affiliation(s)
- Ling Zou
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Adrian Stecula
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Anshul Gupta
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Bhagwat Prasad
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Li Wang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Jashvant D Unadkat
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Simone H Stahl
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Katherine S Fenner
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (L.Z., A.S., H.-C.C., S.W.Y., K.M.G.); Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts (A.G.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., L.W., J.D.U.); and Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK (S.H.S., K.S.F.)
| |
Collapse
|
8
|
Barin-Le Guellec C, Largeau B, Bon D, Marquet P, Hauet T. Ischemia/reperfusion-associated tubular cells injury in renal transplantation: Can metabolomics inform about mechanisms and help identify new therapeutic targets? Pharmacol Res 2018; 129:34-43. [PMID: 29309901 DOI: 10.1016/j.phrs.2017.12.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/29/2017] [Accepted: 12/29/2017] [Indexed: 12/31/2022]
Abstract
Tubular cells are central targets of ischemia-reperfusion (I/R) injury in kidney transplantation. Inflammation and metabolic disturbances occurring within these cells are deleterious by themselves but also favor secondary events, such as activation of immune response. It is critical to have an in depth understanding of the mechanisms governing tubular cells response to I/R if one wants to define pertinent biomarkers or to elaborate targeted therapeutic interventions. As oxidative damage was shown to be central in the patho-physiological mechanisms, the impact of I/R on proximal tubular cells metabolism has been widely studied, contrary to its effects on expression and activity of membrane transporters of the proximal tubular cells. Yet, temporal modulation of transporters over ischemia and reperfusion periods appears to play a central role, not only in the induction of cells injury but also in graft function recovery. Metabolomics in cell models or diverse biofluids has the potential to provide large pictures of biochemical consequences of I/R. Metabolomic studies conducted in experimental models of I/R or in transplanted patients indeed retrieved metabolites belonging to the pathways known to be particularly affected. Interestingly, they also revealed that metabolic disturbances and transporters activities are in very close mutual interplay. As well as helping to select diagnostic biomarkers, such analyses could also contribute to identify new pharmacological targets and to set up innovative nephroprotective strategies for the future. Even if various therapeutic approaches have been evaluated for a long time to prevent or treat I/R injuries, metabolomics has helped identifying new ones, those related to membrane transporters seeming to be of particular interest. However, considering the very complex and multifactorial effects of I/R in the context of kidney transplantation, all tracks must be followed if one wants to prevent or limit its deleterious consequences.
Collapse
Affiliation(s)
- Chantal Barin-Le Guellec
- INSERM UMR 1248, IPPRITT, Limoges, France; CHU Tours, Laboratory of Biochemistry and Molecular Biology, Tours, France; FHU SUPORT, Limoges, Poitiers, Tours, France.
| | - Bérenger Largeau
- CHU Tours, Laboratory of Biochemistry and Molecular Biology, Tours, France
| | - Delphine Bon
- FHU SUPORT, Limoges, Poitiers, Tours, France; University of Poitiers, Poitiers, France; INSERM UMR 1082, IRTOMIT, Poitiers, France; CHU Poitiers, Laboratory of Biochemistry, Poitiers, France
| | - Pierre Marquet
- INSERM UMR 1248, IPPRITT, Limoges, France; FHU SUPORT, Limoges, Poitiers, Tours, France; University of Limoges, Faculty of Medicine, Limoges, France; CHU Limoges, Department of Pharmacology, Toxicology & Pharmacovigilance, Limoges, France
| | - Thierry Hauet
- FHU SUPORT, Limoges, Poitiers, Tours, France; University of Poitiers, Poitiers, France; INSERM UMR 1082, IRTOMIT, Poitiers, France; CHU Poitiers, Laboratory of Biochemistry, Poitiers, France
| |
Collapse
|
9
|
Skwara P, Schömig E, Gründemann D. A novel mode of operation of SLC22A11: Membrane insertion of estrone sulfate versus translocation of uric acid and glutamate. Biochem Pharmacol 2016; 128:74-82. [PMID: 28027879 DOI: 10.1016/j.bcp.2016.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/23/2016] [Indexed: 01/18/2023]
Abstract
Estrone sulfate alias estrone-3-sulfate (E3S) is considerably larger and much more hydrophobic than typical substrates of SLC22 transporters. It is puzzling that many otherwise unrelated transporters have been reported to transport E3S. Here we scrutinized the mechanism of transport of E3S by SLC22A11 (alias OAT4), by direct comparison with uric acid (UA), an important physiological substrate. Heterologous expression of SLC22A11 in human 293 cells gave rise to a huge unidirectional efflux of glutamate (Glu) and aspartate, as determined by LC-MS/MS. The uptake of E3S was 20-fold faster than the uptake of UA. Yet, the outward transport of Glu was inhibited by extracellular E3S, but not by UA. The release of E3S after preloading was trans-stimulated by extracellular dehydroepiandrosterone sulfate (DHEAS), but neither by UA nor 6-carboxyfluorescein (6CF). The equilibrium accumulation of E3S was enhanced 3-fold by replacement of chloride with gluconate, but the opposite effect was observed for UA. These results establish that SLC22A11 provides entirely different transport mechanisms for E3S and UA. Therefore, E3S must not be used as a substitute for UA to assay the function of SLC22A11. In equilibrium accumulation experiments, the transporter-mediated uptake was a linear function of the concentration of UA and 6CF. By contrast, in the same concentration range the graph for E3S was hyperbolic. This suggests that SLC22A11 inserts E3S into a small volume with limited capacity, the plasma membrane. Our data support the notion that the reverse process, extraction from the membrane, is also catalyzed by the carrier.
Collapse
Affiliation(s)
- Peter Skwara
- Department of Pharmacology, University of Cologne, Gleueler Straße 24, 50931 Cologne, Germany
| | - Edgar Schömig
- Department of Pharmacology, University of Cologne, Gleueler Straße 24, 50931 Cologne, Germany
| | - Dirk Gründemann
- Department of Pharmacology, University of Cologne, Gleueler Straße 24, 50931 Cologne, Germany.
| |
Collapse
|
10
|
Tomi M, Eguchi H, Ozaki M, Tawara T, Nishimura S, Higuchi K, Maruyama T, Nishimura T, Nakashima E. Role of OAT4 in Uptake of Estriol Precursor 16α-Hydroxydehydroepiandrosterone Sulfate Into Human Placental Syncytiotrophoblasts From Fetus. Endocrinology 2015; 156:2704-12. [PMID: 25919187 DOI: 10.1210/en.2015-1130] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estriol biosynthesis in human placenta requires the uptake of a fetal liver-derived estriol precursor, 16α-hydroxydehydroepiandrosterone sulfate (16α-OH DHEAS), by placental syncytiotrophoblasts at their basal plasma membrane (BM), which faces the fetal circulation. The aim of this work is to identify the transporter(s) mediating 16α-OH DHEAS uptake at the fetal side of syncytiotrophoblasts by using human placental BM-enriched vesicles and to examine the contribution of the putative transporter to estriol synthesis at the cellular level, using choriocarcinoma JEG-3 cells. Organic anion transporter (OAT)-4 and organic anion transporting polypeptide 2B1 proteins were enriched in human placental BM vesicles compared with crude membrane fraction. Uptake of [(3)H]16α-OH DHEAS by BM vesicles was partially inhibited in the absence of sodium but was significantly increased in the absence of chloride and after preloading glutarate. Uptake of [(3)H]16α-OH DHEAS by BM vesicles was significantly inhibited by OAT4 substrates such as dehydroepiandrosterone sulfate, estrone-3-sulfate, and bromosulfophthalein but not by cyclosporin A, tetraethylammonium, p-aminohippuric acid, or cimetidine. These characteristics of vesicular [(3)H]16α-OH DHEAS uptake are in good agreement with those of human OAT4-transfected COS-7 cells as well as forskolin-differentiated JEG-3 cells. Estriol secretion from differentiated JEG-3 cells was detected when the cells were incubated with 16α-OH DHEAS for 8 hours but was inhibited in the presence of 50 μM bromosulfophthalein. Our results indicate that OAT4 at the BM of human placental syncytiotrophoblasts plays a predominant role in the uptake of 16α-OH DHEAS for placental estriol synthesis.
Collapse
Affiliation(s)
- Masatoshi Tomi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Hiromi Eguchi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Mayuko Ozaki
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tomohiro Tawara
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Sachika Nishimura
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Kei Higuchi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tetsuo Maruyama
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tomohiro Nishimura
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Emi Nakashima
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| |
Collapse
|
11
|
Abstract
Organic anions and cations (OAs and OCs, respectively) comprise an extraordinarily diverse array of compounds of physiological, pharmacological, and toxicological importance. The kidney, primarily the renal proximal tubule, plays a critical role in regulating the plasma concentrations of these organic electrolytes and in clearing the body of potentially toxic xenobiotics agents, a process that involves active, transepithelial secretion. This transepithelial transport involves separate entry and exit steps at the basolateral and luminal aspects of renal tubular cells. Basolateral and luminal OA and OC transport reflects the concerted activity of a suite of separate proteins arranged in parallel in each pole of proximal tubule cells. The cloning of multiple members of several distinct transport families, the subsequent characterization of their activity, and their subcellular localization within distinct regions of the kidney, now allows the development of models describing the molecular basis of the renal secretion of OAs and OCs. New information on naturally occurring genetic variation of many of these processes provides insight into the basis of observed variability of drug efficacy and unwanted drug-drug interactions in human populations. The present review examines recent work on these issues.
Collapse
Affiliation(s)
- Ryan M Pelis
- Novartis Pharmaceuticals Corp., Translational Sciences, East Hanover, New Jersey, USA
| | | |
Collapse
|
12
|
Koepsell H. The SLC22 family with transporters of organic cations, anions and zwitterions. Mol Aspects Med 2013; 34:413-35. [PMID: 23506881 DOI: 10.1016/j.mam.2012.10.010] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 08/18/2012] [Indexed: 12/14/2022]
Abstract
The SLC22 family contains 13 functionally characterized human plasma membrane proteins each with 12 predicted α-helical transmembrane domains. The family comprises organic cation transporters (OCTs), organic zwitterion/cation transporters (OCTNs), and organic anion transporters (OATs). The transporters operate as (1) uniporters which mediate facilitated diffusion (OCTs, OCTNs), (2) anion exchangers (OATs), and (3) Na(+)/zwitterion cotransporters (OCTNs). They participate in small intestinal absorption and hepatic and renal excretion of drugs, xenobiotics and endogenous compounds and perform homeostatic functions in brain and heart. Important endogeneous substrates include monoamine neurotransmitters, l-carnitine, α-ketoglutarate, cAMP, cGMP, prostaglandins, and urate. It has been shown that mutations of the SLC22 genes encoding these transporters cause specific diseases like primary systemic carnitine deficiency and idiopathic renal hypouricemia and are correlated with diseases such as Crohn's disease and gout. Drug-drug interactions at individual transporters may change pharmacokinetics and toxicities of drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, 97070 Würzburg, Germany.
| |
Collapse
|
13
|
Yee SW, Nguyen AN, Brown C, Savic RM, Zhang Y, Castro RA, Cropp CD, Choi JH, Singh D, Tahara H, Stocker SL, Huang Y, Brett CM, Giacomini KM. Reduced renal clearance of cefotaxime in asians with a low-frequency polymorphism of OAT3 (SLC22A8). J Pharm Sci 2013; 102:3451-7. [PMID: 23649425 DOI: 10.1002/jps.23581] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 03/29/2013] [Accepted: 04/12/2013] [Indexed: 12/31/2022]
Abstract
Organic anion transporter 3 (OAT3, SLC22A8), a transporter expressed on the basolateral membrane of the proximal tubule, plays a critical role in the renal excretion of organic anions including many therapeutic drugs. The goal of this study was to evaluate the in vivo effects of the OAT3-Ile305Phe variant (rs11568482), present at 3.5% allele frequency in Asians, on drug disposition with a focus on cefotaxime, a cephalosporin antibiotic. In HEK293-Flp-In cells, the OAT3-Ile305Phe variant had a lower maximum cefotaxime transport activity, Vmax , [159 ± 3 nmol*(mg protein)(-1) /min (mean ± SD)] compared with the reference OAT3 [305 ± 28 nmol*(mg protein)(-1) /min, (mean ± SD), p < 0.01], whereas the Michaelis-Menten constant values (Km ) did not differ. In healthy volunteers, we found volunteers that were heterozygous for the Ile305Phe variant and had a significantly lower cefotaxime renal clearance (CLR ; mean ± SD: 84.8 ± 32.1 mL/min, n = 5) compared with volunteers that were homozygous for the reference allele (158 ± 44.1 mL/min, n = 10; p = 0.006). Furthermore, the net secretory component of cefotaxime renal clearance (CLsec ) was reduced in volunteers heterozygous for the variant allele [33.3 ± 31.8 mL/min (mean ± SD)] compared with volunteers homozygous for the OAT3 reference allele [97.0 ± 42.2 mL/min (mean ± SD), p = 0.01]. In summary, our study suggests that a low-frequency reduced-function polymorphism of OAT3 associates with reduced cefotaxime CLR and CL(sec) .
Collapse
Affiliation(s)
- Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Felmlee MA, Dave RA, Morris ME. Mechanistic models describing active renal reabsorption and secretion: a simulation-based study. AAPS JOURNAL 2012. [PMID: 23196805 DOI: 10.1208/s12248-012-9437-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The objective of the present study was to evaluate mechanistic pharmacokinetic models describing active renal secretion and reabsorption over a range of Michaelis-Menten parameter estimates and doses. Plasma concentration and urinary excretion profiles were simulated and renal clearance (CL(r)) was calculated for two pharmacokinetic models describing active renal reabsorption (R1/R2), two models describing active secretion (S1/S2), and a model containing both processes. A range of doses (1-1,000 mg/kg) was evaluated, and V (max) and K (m) parameter estimates were varied over a 100-fold range. Similar CL(r) values were predicted for reabsorption models (R1/R2) with variations in V (max) and K (m). Tubular secretion models (S1/S2) yielded similar relationships between Michaelis-Menten parameter perturbations and CL(r), but the predicted CL(r) values were threefold higher for model S1. For both reabsorption and secretion models, the greatest changes in CL(r) were observed with perturbations in V (max), suggesting the need for an accurate estimate of this parameter. When intrinsic clearance was substituted for Michaelis-Menten parameters, it failed to predict similar CL(r) values even within the linear range. For models S1 and S2, renal secretion was predominant at low doses, whereas renal clearance was driven by fraction unbound in plasma at high doses. Simulations demonstrated the importance of Michaelis-Menten parameter estimates (especially V (max)) for determining CL(r). K (m) estimates can easily be obtained directly from in vitro studies. However, additional scaling of in vitro V (max) estimates using in vitro/in vivo extrapolation methods are required to incorporate these parameters into pharmacokinetic models.
Collapse
Affiliation(s)
- Melanie A Felmlee
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | | | | |
Collapse
|
15
|
Wang L, Sweet DH. Renal organic anion transporters (SLC22 family): expression, regulation, roles in toxicity, and impact on injury and disease. AAPS JOURNAL 2012; 15:53-69. [PMID: 23054972 DOI: 10.1208/s12248-012-9413-y] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 09/12/2012] [Indexed: 01/25/2023]
Abstract
Organic solute flux across the basolateral and apical membranes of renal proximal tubule cells is a key process for maintaining systemic homeostasis. It represents an important route for the elimination of metabolic waste products and xenobiotics, as well as for the reclamation of essential compounds. Members of the organic anion transporter (OAT, SLC22) family expressed in proximal tubules comprise one pathway mediating the active renal secretion and reabsorption of organic anions. Many drugs, pesticides, hormones, heavy metal conjugates, components of phytomedicines, and toxins are OAT substrates. Thus, through transporter activity, the kidney can be a target organ for their beneficial or detrimental effects. Detailed knowledge of the OATs expressed in the kidney, their membrane targeting, substrate specificity, and mechanisms of action is essential to understanding organ function and dysfunction. The intracellular processes controlling OAT expression and function, and that can thus modulate kidney transport capacity, are also critical to this understanding. Such knowledge is also providing insight to new areas such as renal transplant research. This review will provide an overview of the OATs for which transport activity has been demonstrated and expression/function in the kidney observed. Examples establishing a role for renal OATs in drug clearance, food/drug-drug interactions, and renal injury and pathology are presented. An update of the current information regarding the regulation of OAT expression is also provided.
Collapse
Affiliation(s)
- Li Wang
- Department of Pharmaceutics, Virginia Commonwealth University, Medical College of Virginia Campus, 410 N 12th Street, PO Box 980533, Richmond, VA 23298, USA
| | | |
Collapse
|
16
|
Sakurai H. Transporter-centric view of urate metabolism: From genome-wide association study to pathophysiology. JOURNAL OF PHYSICAL FITNESS AND SPORTS MEDICINE 2012. [DOI: 10.7600/jpfsm.1.413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
17
|
A common 5'-UTR variant in MATE2-K is associated with poor response to metformin. Clin Pharmacol Ther 2011; 90:674-84. [PMID: 21956618 DOI: 10.1038/clpt.2011.165] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Multidrug and toxin extrusion 2 (MATE2-K (SLC47A2)), a polyspecific organic cation exporter, facilitates the renal elimination of the antidiabetes drug metformin. In this study, we characterized genetic variants of MATE2-K, determined their association with metformin response, and elucidated their impact by means of a comparative protein structure model. Four nonsynonymous variants and four variants in the MATE2-K basal promoter region were identified from ethnically diverse populations. Two nonsynonymous variants-c.485C>T and c.1177G>A-were shown to be associated with significantly lower metformin uptake and reduction in protein expression levels. MATE2-K basal promoter haplotypes containing the most common variant, g.-130G>A (>26% allele frequency), were associated with a significant increase in luciferase activities and reduced binding to the transcriptional repressor myeloid zinc finger 1 (MZF-1). Patients with diabetes who were homozygous for g.-130A had a significantly poorer response to metformin treatment, assessed as relative change in glycated hemoglobin (HbA1c) (-0.027 (-0.076, 0.033)), as compared with carriers of the reference allele, g.-130G (-0.15 (-0.17, -0.13)) (P=0.002). Our study showed that MATE2-K plays a role in the antidiabetes response to metformin.
Collapse
|
18
|
Cutler MJ, Urquhart BL, Velenosi TJ, Meyer Zu Schwabedissen HE, Dresser GK, Leake BF, Tirona RG, Kim RB, Freeman DJ. In vitro and in vivo assessment of renal drug transporters in the disposition of mesna and dimesna. J Clin Pharmacol 2011; 52:530-42. [PMID: 21505084 DOI: 10.1177/0091270011400414] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mesna and its dimer, dimesna, are coadministered for mitigation of ifosfamide- and cisplatin-induced toxicities, respectively. Dimesna is selectively reduced to mesna in the kidney, producing its protective effects. In vitro screens of uptake and efflux transporters revealed saturable uptake by renal organic anion transporters OAT1, OAT3, and OAT4. Efflux transporters breast cancer resistance protein; multidrug and toxin extrusion 1 (MATE1); multidrug resistance proteins MRP1, MRP2, MRP4, and MRP5; and P-glycoprotein (Pgp) significantly reduced dimesna accumulation. Further investigation demonstrated that renal apical efflux transporters MATE1, MRP2, and Pgp were also capable of mesna efflux. Administration of OAT inhibitor probenecid to healthy subjects significantly increased combined mesna and dimesna plasma exposure (91% ± 34%) while decreasing the renal clearance due to net secretion (67.0% ± 12.7%) and steady-state volume of distribution (45.2% ± 13.4%). Thus, the kidney represents a significant sink of total mesna, whereas function of renal drug transporters facilitates clearance in excess of glomerular filtration rate and likely the presence of active mesna in the urine. Loss of renal transporter function due to genetic variability or drug-drug interactions may decrease the efficacy of chemoprotectants, increasing the risk of ifosfamide- and cisplatin-induced toxicities.
Collapse
Affiliation(s)
- M J Cutler
- Department of Medicine, Division of Clinical Pharmacology, The University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|