Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 49(10): 929-937, 2021 10.
Article in English | MEDLINE | ID: mdl-34315779

ABSTRACT

Pregnancy can significantly change the pharmacokinetics of drugs, including those renally secreted by organic anion transporters (OATs). Quantifying these changes in pregnant women is logistically and ethically challenging. Hence, predicting the in vivo plasma renal secretory clearance (CLsec) and renal CL (CLrenal) of OAT drugs in pregnancy is important to design correct dosing regimens of OAT drugs. Here, we first quantified the fold-change in renal OAT activity in pregnant versus nonpregnant individual using available selective OAT probe drug CLrenal data (training dataset; OAT1: tenofovir, OAT2: acyclovir, OAT3: oseltamivir carboxylate). The fold-change in OAT1 activity during the 2nd and 3rd trimester was 2.9 and 1.0 compared with nonpregnant individual, respectively. OAT2 activity increased 3.1-fold during the 3rd trimester. OAT3 activity increased 2.2, 1.7 and 1.3-fold during the 1st, 2nd, and 3rd trimester, respectively. Based on these data, we predicted the CLsec, CLrenal and total clearance ((CLtotal) of drugs in pregnancy, which are secreted by multiple OATs (verification dataset; amoxicillin, pravastatin, cefazolin and ketorolac, R-ketorolac, S-ketorolac). Then, the predicted clearances (CLs) were compared with the observed values. The predicted/observed CLsec, CLrenal, and CLtotal of drugs in pregnancy of all verification drugs were within 0.80-1.25 fold except for CLsec of amoxicillin in the 3rd trimester (0.76-fold) and cefazolin in the 2nd trimester (1.27-fold). Overall, we successfully predicted the CLsec, CLrenal, and CLtotal of drugs in pregnancy that are renally secreted by multiple OATs. This approach could be used in the future to adjust dosing regimens of renally secreted OAT drugs which are administered to pregnant women. SIGNIFICANCE STATEMENT: To the authors' knowledge, this is the first report to successfully predict renal secretory clearance and renal clearance of multiple OAT substrate drugs during pregnancy. The data presented here could be used in the future to adjust dosing regimens of renally secreted OAT drugs in pregnancy. In addition, the mechanistic approach used here could be extended to drugs transported by other renal transporters.


Subject(s)
Biological Transport, Active/physiology , Dose-Response Relationship, Drug , Organic Anion Transporters , Pharmacokinetics , Renal Elimination/physiology , Biotransformation/physiology , Drug Dosage Calculations , Female , HEK293 Cells , Humans , Metabolic Clearance Rate , Organic Anion Transporters/classification , Organic Anion Transporters/metabolism , Pharmaceutical Preparations/classification , Pharmaceutical Preparations/metabolism , Pregnancy , Pregnancy Trimesters/drug effects , Pregnancy Trimesters/metabolism , Reproducibility of Results
2.
Int J Mol Sci ; 21(5)2020 Mar 05.
Article in English | MEDLINE | ID: mdl-32150922

ABSTRACT

The SLC22 family of OATs, OCTs, and OCTNs is emerging as a central hub of endogenous physiology. Despite often being referred to as "drug" transporters, they facilitate the movement of metabolites and key signaling molecules. An in-depth reanalysis supports a reassignment of these proteins into eight functional subgroups, with four new subgroups arising from the previously defined OAT subclade: OATS1 (SLC22A6, SLC22A8, and SLC22A20), OATS2 (SLC22A7), OATS3 (SLC22A11, SLC22A12, and Slc22a22), and OATS4 (SLC22A9, SLC22A10, SLC22A24, and SLC22A25). We propose merging the OCTN (SLC22A4, SLC22A5, and Slc22a21) and OCT-related (SLC22A15 and SLC22A16) subclades into the OCTN/OCTN-related subgroup. Using data from GWAS, in vivo models, and in vitro assays, we developed an SLC22 transporter-metabolite network and similar subgroup networks, which suggest how multiple SLC22 transporters with mono-, oligo-, and multi-specific substrate specificity interact to regulate metabolites. Subgroup associations include: OATS1 with signaling molecules, uremic toxins, and odorants, OATS2 with cyclic nucleotides, OATS3 with uric acid, OATS4 with conjugated sex hormones, particularly etiocholanolone glucuronide, OCT with neurotransmitters, and OCTN/OCTN-related with ergothioneine and carnitine derivatives. Our data suggest that the SLC22 family can work among itself, as well as with other ADME genes, to optimize levels of numerous metabolites and signaling molecules, involved in organ crosstalk and inter-organismal communication, as proposed by the remote sensing and signaling theory.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Mutation , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Systems Biology/methods , Animals , Biological Transport , Humans , Multigene Family , Organic Anion Transporters/classification , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/classification , Organic Cation Transport Proteins/genetics , Signal Transduction , Substrate Specificity
3.
Clin J Am Soc Nephrol ; 10(11): 2039-49, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26490509

ABSTRACT

The proximal tubule of the kidney plays a crucial role in the renal handling of drugs (e.g., diuretics), uremic toxins (e.g., indoxyl sulfate), environmental toxins (e.g., mercury, aristolochic acid), metabolites (e.g., uric acid), dietary compounds, and signaling molecules. This process is dependent on many multispecific transporters of the solute carrier (SLC) superfamily, including organic anion transporter (OAT) and organic cation transporter (OCT) subfamilies, and the ATP-binding cassette (ABC) superfamily. We review the basic physiology of these SLC and ABC transporters, many of which are often called drug transporters. With an emphasis on OAT1 (SLC22A6), the closely related OAT3 (SLC22A8), and OCT2 (SLC22A2), we explore the implications of recent in vitro, in vivo, and clinical data pertinent to the kidney. The analysis of murine knockouts has revealed a key role for these transporters in the renal handling not only of drugs and toxins but also of gut microbiome products, as well as liver-derived phase 1 and phase 2 metabolites, including putative uremic toxins (among other molecules of metabolic and clinical importance). Functional activity of these transporters (and polymorphisms affecting it) plays a key role in drug handling and nephrotoxicity. These transporters may also play a role in remote sensing and signaling, as part of a versatile small molecule communication network operative throughout the body in normal and diseased states, such as AKI and CKD.


Subject(s)
Kidney Tubules, Proximal/metabolism , Pharmaceutical Preparations/metabolism , Biological Transport , Humans , Organic Anion Transporters/classification , Organic Anion Transporters/physiology , Signal Transduction , Uremia
4.
Drug Metab Pharmacokinet ; 30(5): 347-51, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26293543

ABSTRACT

Hepatic transporters, a major determinant of pharmacokinetics, have been used to profile drug properties like efficacy. Among hepatic transporters, importers alter the concentration of the drug by facilitating the transport of a drug into a cell. Despite vast pharmacokinetic studies, the interacting mechanisms of the importers with its substrates or inhibitors are not well understood. Hence, we developed compound binary classification models of whether a compound is binder or nonbinder to a hepatic transporter with experimental data of 284 compounds for four representative hepatic importers, OATP1B1, OATP1B3, OAT2, and OCT1. Support Vector Machine (SVM) along with Genetic Algorithm (GA) was used to construct the classification models of binder versus nonbinder for each target importer. To construct the models, we prepared two data sets, a training data set from Fujitsu database (284 compounds) and an external validation data set from ChEMBL database (1738 compounds). Since an experimental classification criterion between binder and nonbinder has some ambiguity, there is an intrinsic limitation to expect high predictability of the binary classification models developed with the experimental data. The predictability of the classification models calculated with external validation sets were obtained as 77.72%, 84.31%, 84.21%, and 76.38 for OATP1B1, OATP1B3, OAT2, and OCT1, respectively.


Subject(s)
Organic Anion Transporters/metabolism , Computer Simulation , Databases, Chemical , HEK293 Cells , Hepatocytes/metabolism , Humans , Models, Biological , Models, Molecular , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/classification , Pharmacokinetics , Predictive Value of Tests , Reproducibility of Results , Support Vector Machine
5.
Biochem Pharmacol ; 84(8): 1088-95, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22877817

ABSTRACT

Phenolic acids exert beneficial health effects such as anti-oxidant, anti-carcinogenic, and anti-inflammatory activities and show systemic exposure after consumption of common fruits, vegetables, and beverages. However, knowledge regarding which components convey therapeutic benefits and the mechanism(s) by which they cross cell membranes is extremely limited. Therefore, we determined the inhibitory effects of nine food-derived phenolic acids, p-coumaric acid, ferulic acid, gallic acid, gentisic acid, 4-hydroxybenzoic acid, protocatechuic acid, sinapinic acid, syringic acid, and vanillic acid, on human organic anion transporter 1 (hOAT1), hOAT3, and hOAT4. In the present study, inhibition of OAT-mediated transport of prototypical substrates (1 µM) by phenolic acids (100 µM) was examined in stably expressing cell lines. All compounds significantly inhibited hOAT3 transport, while just ferulic, gallic, protocatechuic, sinapinic, and vanillic acid significantly blocked hOAT1 activity. Only sinapinic acid inhibited hOAT4 (~35%). For compounds exhibiting inhibition > ~60%, known clinical plasma concentration levels and plasma protein binding in humans were examined to select compounds to evaluate further with dose-response curves (IC(50) values) and drug-drug interaction (DDI) index determinations. IC(50) values ranged from 1.24 to 18.08 µM for hOAT1 and from 7.35 to 87.36 µM for hOAT3. Maximum DDI indices for gallic and gentisic acid (≫0.1) indicated a very strong potential for DDIs on hOAT1 and/or hOAT3. This study indicates that gallic acid from foods or supplements, or gentisic acid from salicylate-based drug metabolism, may significantly alter the pharmacokinetics (efficacy and toxicity) of concomitant therapeutics that are hOAT1 and/or hOAT3 substrates.


Subject(s)
Food-Drug Interactions , Hydroxybenzoates/pharmacology , Organic Anion Transporters/drug effects , Humans , Inhibitory Concentration 50 , Organic Anion Transporters/classification
6.
J Clin Neurosci ; 17(6): 679-84, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20356750

ABSTRACT

The organic anion-transporting polypeptides (rodents: Oatps; human: OATPs) belong to the growing family of organic anion/prostaglandin transporters and are important components of the active efflux transport system at the choroid plexus epithelial cells. OATPs facilitate the elimination of xenobiotics and endogenous waste from the cerebrospinal fluid and prevent waste accumulation in the central nervous system (CNS). This review summarizes the structures, regulations and roles of Oatps/OATPs at the choroid plexus in drug delivery to the CNS.


Subject(s)
Choroid Plexus/metabolism , Drug Delivery Systems , Organic Anion Transporters/metabolism , Animals , Drug Delivery Systems/methods , Humans , Organic Anion Transporters/classification , Organic Anion Transporters/genetics , Protein Transport , Tissue Distribution
7.
Cell Physiol Biochem ; 25(4-5): 511-22, 2010.
Article in English | MEDLINE | ID: mdl-20332632

ABSTRACT

This study identifies a novel organic anion transporter Oat9 expressed in mouse liver and kidney. Two variants were detected by screening a mouse liver cDNA library; these varients consist of 1815 (designated Oat9S) and 2165 (Oat9L) base pairs which encode 443 and 551 amino acid proteins, respectively. Oat9S has a predicted structure containing eight transmembrane domains (TMD); whereas, Oat9L possesses twelve TMD. Oat9 mRNA expression was detected in kidney and liver. This transporter was located at the apical side of the late portion of proximal tubules and at the sinusoidal side of hepatocytes. When expressed in Xenopus oocytes, Oat9S mediated the transport of L-carnitine (Km = 2.9 microM), a representative zwitterion, as well as cimetidine (Km = 16.1 microM) and salicylic acid (Km = 175.5 microM), while Oat9L did not show any transport activity. Oat9S-mediated L-carnitine uptake was inhibited by D-carnitine, acetylcarnitine, octanoylcarnitine, betaine, and other organic compounds, suggesting that quaternary ammonium cation bulkiness and relative hydrophobicity are important factors for Oat9S-substrate interactions. Among OATs, Oat9S appears to be the first member to mediate the transport of carnitine and possesses eight TMD. Overall, these new results provide added insight into the structure-activity relationship comprising the organic ion-permeation pathway.


Subject(s)
Carnitine/metabolism , Kidney/metabolism , Liver/metabolism , Organic Anion Transporters/metabolism , Amino Acid Sequence , Animals , Biological Transport , Mice , Molecular Sequence Data , Oocytes/metabolism , Organic Anion Transporters/analysis , Organic Anion Transporters/classification , Organic Anion Transporters/genetics , Sequence Alignment , Xenopus laevis
8.
Article in English | MEDLINE | ID: mdl-20079461

ABSTRACT

The human liver-specific organic anion transporting polypeptides (OATPs) 1B1 and 1B3 are involved in the elimination of numerous xenobiotics and drugs. Although dogs are frequently used for toxicologic and pharmacokinetic characterization of novel drugs, nothing is known about their OATP1B1/1B3 ortholog. Therefore, we cloned and characterized the first canine organic anion transporting polypeptide from dog liver, termed Oatp1b4. The isolated Oatp1b4 cDNA comprises 3661 base pairs (bp) with an open reading frame of 2076bp, encoding a 692-amino acid protein with a molecular mass of approximately 85kDa. The Oatp1b4 gene is approximately 61kb long and has a similar organization as the human OATP1B1 and OATP1B3 with 13 exons identical in length. Northern blot analysis shows that Oatp1b4 is predominantly expressed in the liver. Oatp1b4 mediates sodium-independent transport of typical organic anions including bromosulfophthalein (BSP), [D-penicillamine(2,5)]enkephalin (DPDPE), estradiol-17beta-glucuronide (E17betaG), estrone-3-sulfate and taurocholate. In addition, Oatp1b4 transports the OATP1B3-specific substrate cholecystokinin octapeptide (CCK-8). Kinetic studies showed that Oatp1b4-mediated E17betaG and estrone-3-sulfate transports were monophasic with K(m) values of 5+/-1microM and 33+/-4microM, respectively. In conclusion, the cloned canine Oatp1b4 will provide additional molecular basis to further characterize the species difference of the OATP1B family members.


Subject(s)
Organic Anion Transporters/chemistry , Organic Anion Transporters/classification , Organic Anion Transporters/metabolism , Amino Acid Sequence , Animals , Base Pairing , Base Sequence , Biological Transport , Cell Line , Cloning, Molecular , DNA, Complementary/chemistry , Dogs , Enkephalin, D-Penicillamine (2,5)-/metabolism , Estradiol/analogs & derivatives , Estradiol/metabolism , Estrone/analogs & derivatives , Estrone/metabolism , Exons , Genes , Humans , Introns , Kidney/cytology , Kinetics , Liver/metabolism , Molecular Sequence Data , Molecular Weight , Open Reading Frames , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Sincalide/metabolism , Substrate Specificity , Sulfobromophthalein/metabolism , Taurocholic Acid/metabolism
9.
Drug Metab Pharmacokinet ; 19(3): 171-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15499184

ABSTRACT

In the last decade, many organic anion transporters have been isolated, characterized their distribution and substrates. The recently identified organic anion transporter family OATP (organic anion transporting polypeptide)/LST (liver-specific transporter) family, transport bile acids, hormones as well as eicosanoids, various compounds (BSP, HMG-CoA reductase inhibitor, angiotensin converting enzyme inhibitor, etc.). The isolation of the family revealed that not only hydrophilic compounds, drugs and hormones of lipophilic nature need a membrane transport system to penetrate cell membrane. In this family, the nomenclature becomes very complicated and the physiological role of this family is still unclear except about few organs such as the brain, liver and kidney. Even in such organs, the co-existence of the OATP/LST family and similar substrate specificity hamper the progress and clear characterization identifying the real role of the transporter family. Here, recent progress and an insight of this field are reviewed.


Subject(s)
Organic Anion Transporters/metabolism , Organic Anion Transporters/physiology , Animals , Humans , Organic Anion Transporters/classification , Organic Anion Transporters/genetics , Pharmaceutical Preparations/metabolism , Phylogeny
10.
Med Res Rev ; 24(6): 762-74, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15250040

ABSTRACT

Organic anion transporters (OAT) play essential roles in the body disposition of clinically important anionic drugs, including anti-viral drugs, anti-tumor drugs, antibiotics, anti-hypertensives, and anti-inflammatories. The activities of OATs are directly linked to drug toxicity and drug-drug interactions. So far, four members of the OAT family have been identified: OAT1, OAT2, OAT3, and OAT4. These transporters share several common structural features including 12 transmembrane domains, multiple glycosylation sites localized in the first extracellular loop between transmembrane domains 1 and 2, and multiple phosphorylation sites present in the intracellular loop between transmembrane domains 6 and 7, and in the carboxyl terminus. The impact of these structural features on the function of these transporters has just begun to be explored. In the present review, the author will summarize recent progress made from her laboratory as well as from others, on the molecular characterization of the structure-function relationships of OATs, including particular amino acid residues/regions of the transporter protein ("molecular domains") that potentially determine transport characteristics.


Subject(s)
Organic Anion Transporters/chemistry , Organic Anion Transporters/metabolism , Animals , Glycosylation , Humans , Inactivation, Metabolic/physiology , Organic Anion Transporters/classification , Pharmacology , Protein Conformation , Structure-Activity Relationship , Substrate Specificity
11.
Physiol Genomics ; 18(1): 12-24, 2004 Jun 17.
Article in English | MEDLINE | ID: mdl-15054140

ABSTRACT

Slc22 family organic anion and cation transporters (OATs, OCTs, and OCTNs) are transmembrane proteins expressed predominantly in kidney and liver. These proteins mediate the uptake or excretion of numerous physiologically (and pharmacologically) important compounds, and accordingly have been the focus of intensive study. Here we investigate the molecular phylogeny of the slc22 transporters, identifying homologs in Drosophila and C. elegans, several of which are developmentally regulated, as well as reporting the cloning of a novel human family member, UST6, expressed exclusively in liver in both embryo and adult. The latter helps define a subfamily within the OATs, which appears to have human- and rodent-specific members, raising potential issues with respect to the use of rodents as models for the transport of organic anions (which include many pharmaceuticals) in humans. Although this phylogenetic inference could not be made on the basis of sequence alignment, analysis of intron phasing suggests that the OAT, OCT, and OCTN lineages of the slc22 family formed after the divergence of vertebrates and invertebrates. Subsequently, these lineages expanded through independent tandem duplications to produce multiple gene pairs. After analyzing over 200 other transporter genes, we find such pairing to be relatively specific to vertebrate organic anion and cation transporters, suggesting selection for gene pairing operating within this family in particular. This might reflect a requirement for redundancy or broader substrate specificity in vertebrates (compared to invertebrates), due to their greater physiological complexity and thus potentially broader exposure to organic ions.


Subject(s)
Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Multigene Family , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Amino Acid Sequence , Animals , Caenorhabditis elegans/growth & development , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Evolution, Molecular , Gene Expression Regulation, Developmental , Genes , Humans , Ion Transport/genetics , Molecular Sequence Data , Organ Specificity , Organic Anion Transporters/classification , Organic Cation Transport Proteins/classification , Organic Cation Transport Proteins/isolation & purification , Phylogeny , Rats , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Solute Carrier Proteins , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...