Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Asian Nat Prod Res ; 21(8): 754-771, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30606060

ABSTRACT

Andrographolide, a major bioactive compound isolated from Andrographis paniculata (Burm. F.) Nees, was evaluated for its effects on the hOAT1 membrane transporter. Substrate determination and inhibition of hOAT1-mediated uptake transport assay was carried out using recombinant CHO-hOAT1 cells. The results showed that the uptake ratio of andrographolide was less than 2.0 at all concentrations tested, indicating that andrographolide is not a hOAT1 substrate. Andrographolide has no significant effects on the p-aminohippuric acid uptake and on the mRNA and protein expression of hOAT1. In conclusion, andrographolide may not pose a drug-herb interaction risk related to hOAT1.


Subject(s)
Diterpenes/pharmacology , Organic Anion Transport Protein 1/antagonists & inhibitors , Animals , CHO Cells , Cell Proliferation/drug effects , Cricetulus , Diterpenes/pharmacokinetics , Herb-Drug Interactions , Humans , Molecular Docking Simulation , Organic Anion Transport Protein 1/analysis , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/genetics , Probenecid/chemistry , Probenecid/pharmacology , p-Aminohippuric Acid/pharmacokinetics
2.
Toxicol Sci ; 161(2): 321-334, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29045746

ABSTRACT

Mercury accumulates in kidneys and produces acute kidney injury. Semen cassiae (SC), a widely consumed tea and herbal medicine in Eastern Asia, has been reported to have protective effects on kidneys. In this study, SC extract was shown to almost abolish the histological alterations induced by mercuric chloride in rat kidneys. A total of 22 compounds were isolated from SC, and 1,7,8-methoxyl-2-hydroxyl-3-methyl-anthraquinone was detected in SC for the first time. Among the eight compounds identified in the blood of rats after SC treatment, six were strong inhibitors of human organic anion transporter 1 and 3 (OAT1 and OAT3). Inhibitory studies revealed that OAT1 and OAT3 were inhibited by SC constituents, in both a competitive and noncompetitive manner. Both OAT1- and OAT3-overexpressing cells were susceptible to the cytotoxicity of the cysteine-mercury conjugate, but only OAT1-overexpressing cells could be protected by 200 µM probenecid or 10 µM of the eight inhibitors in SC, suggesting that OAT1 is the major determinant in the cellular uptake of mercury. To facilitate the identification of inhibitors of OAT1 and OAT3, models of OAT1 and OAT3 were constructed using recently determined protein templates. By combining in silico and in vitro methods, inhibitors of OAT1 and OAT3 were predicted and validated from SC constituents. Collectively, the present study suggests that additional inhibitors of OAT1 and OAT3 can be predicted and validated from natural products by combining docking and in vitro screening, and could be a source of pharmaceutical compounds for developing treatments for mercury-induced kidney injury.


Subject(s)
Acute Kidney Injury/prevention & control , Drugs, Chinese Herbal/therapeutic use , Mercuric Chloride/toxicity , Organic Anion Transport Protein 1/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Protective Agents/therapeutic use , Senna Plant/chemistry , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Amino Acid Sequence , Animals , Cell Survival/drug effects , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/pharmacokinetics , HEK293 Cells , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Function Tests , Male , Molecular Docking Simulation , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Protective Agents/isolation & purification , Protective Agents/pharmacokinetics , Rats, Sprague-Dawley , Structural Homology, Protein
3.
Sci Rep ; 6: 34995, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713539

ABSTRACT

Gout is caused by elevated serum urate levels, which can be treated using inhibitors of the uric acid transporter, URAT1. We exploited affinity differences between the human and rat transporters to map inhibitor binding sites in URAT1. Human-rat transporter chimeras revealed that human URAT1 serine-35, phenylalanine-365 and isoleucine-481 are necessary and sufficient to provide up to a 100-fold increase in affinity for inhibitors. Moreover, serine-35 and phenylalanine-365 are important for high-affinity interaction with the substrate urate. A novel URAT1 binding assay provides support for direct interaction with these amino acids; thus, current clinically important URAT1 inhibitors likely bind the same site in URAT1. A structural model suggests that these three URAT1 residues are in close proximity potentially projecting within the channel. Our results indicate that amino acids from several transmembrane segments functionally cooperate to form a high-affinity URAT1 inhibitor binding site that, when occupied, prevents substrate interactions.


Subject(s)
Organic Anion Transporters/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Amino Acid Substitution , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/chemistry , Anion Transport Proteins/genetics , Binding Sites/genetics , HEK293 Cells , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Organic Anion Transport Protein 1/chemistry , Organic Anion Transporters/chemistry , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/chemistry , Organic Cation Transport Proteins/genetics , Protein Interaction Domains and Motifs , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Uric Acid/metabolism
4.
Bone ; 93: 97-103, 2016 12.
Article in English | MEDLINE | ID: mdl-27664568

ABSTRACT

CONTEXT: Adefovir dipivoxil (ADV) was an important cause of adult-onset hypophosphatemic osteomalacia. However, its clinical characteristics and mechanisms have not been well defined. OBJECTIVE: The objective of the study was to summarize the clinical characteristics of ADV-induced osteomalacia and to explore the association between ADV-associated tubulopathy and polymorphisms in genes encoding drug transporters. DESIGN, SETTING, PATIENTS, AND MAIN OUTCOME MEASURE: Seventy-six affected patients were clinically studied. The SLC22A6 and ABCC2 genes were screened and compared with healthy people from the HapMap. RESULTS: Hypophosphatemia, high serum alkaline phosphatase (ALP) levels, hypouricemia, nondiabetic glycosuria, proteinuria, metabolic acidosis and high bone turnover markers were the main metabolic characteristics. Fractures and pseudofractures occurred in 39 patients. Stopping ADV administration, supplementing calcitriol and calcium was effective during the follow-up period. Single SNP analysis revealed a higher percentage of the G/A genotype at c.2934 in exon 22 of the ABCC2 gene (rs3740070) in patients than in healthy people (12% [7 of 58 patients] vs. 0% [0 of 45 patients]; P=0.017), while there was no subject with homozygosity for the A allele at c.2934. CONCLUSIONS: ADV can be nephrotoxic at a conventional dosage. The G/A genotype at c.2934 of the ABCC2 gene may be a predictor of patients at greater risk for developing ADV-associated tubulopathy. Larger case-control studies are needed to further verify this finding.


Subject(s)
Adenine/analogs & derivatives , Genetic Predisposition to Disease , Organophosphonates/adverse effects , Osteomalacia/chemically induced , Osteomalacia/genetics , Adenine/adverse effects , Amino Acid Sequence , Case-Control Studies , DNA Mutational Analysis , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Gene Frequency/genetics , Genetic Association Studies , Humans , Male , Middle Aged , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/genetics , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/genetics , Osteomalacia/diagnostic imaging , Osteomalacia/therapy , Polymorphism, Single Nucleotide/genetics , Risk Factors , Sequence Homology, Amino Acid
5.
J Pharmacol Exp Ther ; 359(1): 215-29, 2016 10.
Article in English | MEDLINE | ID: mdl-27488918

ABSTRACT

Statistical analysis was performed on physicochemical descriptors of ∼250 drugs known to interact with one or more SLC22 "drug" transporters (i.e., SLC22A6 or OAT1, SLC22A8 or OAT3, SLC22A1 or OCT1, and SLC22A2 or OCT2), followed by application of machine-learning methods and wet laboratory testing of novel predictions. In addition to molecular charge, organic anion transporters (OATs) were found to prefer interacting with planar structures, whereas organic cation transporters (OCTs) interact with more three-dimensional structures (i.e., greater SP3 character). Moreover, compared with OAT1 ligands, OAT3 ligands possess more acyclic tetravalent bonds and have a more zwitterionic/cationic character. In contrast, OCT1 and OCT2 ligands were not clearly distinquishable form one another by the methods employed. Multiple pharmacophore models were generated on the basis of the drugs and, consistent with the machine-learning analyses, one unique pharmacophore created from ligands of OAT3 possessed cationic properties similar to OCT ligands; this was confirmed by quantitative atomic property field analysis. Virtual screening with this pharmacophore, followed by transport assays, identified several cationic drugs that selectively interact with OAT3 but not OAT1. Although the present analysis may be somewhat limited by the need to rely largely on inhibition data for modeling, wet laboratory/in vitro transport studies, as well as analysis of drug/metabolite handling in Oat and Oct knockout animals, support the general validity of the approach-which can also be applied to other SLC and ATP binding cassette drug transporters. This may make it possible to predict the molecular properties of a drug or metabolite necessary for interaction with the transporter(s), thereby enabling better prediction of drug-drug interactions and drug-metabolite interactions. Furthermore, understanding the overlapping specificities of OATs and OCTs in the context of dynamic transporter tissue expression patterns should help predict net flux in a particular tissue of anionic, cationic, and zwitterionic molecules in normal and pathophysiological states.


Subject(s)
Computational Biology/methods , Machine Learning , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations/metabolism , Hydrogen-Ion Concentration , Membrane Transport Proteins/chemistry , Models, Molecular , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Cation Transporter 1/chemistry , Organic Cation Transporter 1/metabolism , Pharmaceutical Preparations/chemistry , Protein Binding , Protein Conformation , Substrate Specificity
6.
J Biol Chem ; 286(43): 37874-86, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21896487

ABSTRACT

Polyspecific organic anion transporters (OATs) and organic cation transporters (OCTs) of the SLC22 transporter family play a pivotal role in absorption, distribution, and excretion of drugs. Polymorphisms in these transporters influence therapeutic effects. On the basis of functional characterizations, homology modeling, and mutagenesis, hypotheses for how OCTs bind and translocate structurally different cations were raised, assuming functionally competent monomers. However, homo-oligomerization has been described for OATs and OCTs. In the present study, evidence is provided that the large extracellular loops (EL) of rat Oct1 (rOct1) and rat Oat1 (rOat1) mediate homo- but not hetero-oligomerization. Replacement of the cysteine residues in the EL of rOct1 by serine residues (rOct1(6ΔC-l)) or breaking disulfide bonds with dithiothreitol prevented oligomerization. rOct1 chimera containing the EL of rOat1 (rOct1(rOat1-l)) showed oligomerization but reduced transporter amount in the plasma membrane. For rOct1(6ΔC-l) and rOct1(rOat1-l), similar K(m) values for 1-methyl-4-phenylpyridinium(+) (MPP(+)) and tetraethylammonium(+) (TEA(+)) were obtained that were higher compared with rOct1 wild type. The increased K(m) of rOct1(rOat1-l) indicates an allosteric effect of EL on the cation binding region. The similar substrate affinity of the oligomerizing and non-oligomerizing loop mutants suggests that oligomerization does not influence transport function. Independent transport function of rOct1 monomers was also demonstrated by showing that K(m) values for MPP(+) and TEA(+) were not changed after treatment with dithiothreitol and that a tandem protein with two rOct1 monomers showed about 50% activity with unchanged K(m) values for MPP(+) and TEA(+) when one monomer was blocked. The data help to understand how OCTs work and how mutations in patients may affect their functions.


Subject(s)
Catecholamine Plasma Membrane Transport Proteins/metabolism , Protein Multimerization/physiology , Animals , Catecholamine Plasma Membrane Transport Proteins/chemistry , Catecholamine Plasma Membrane Transport Proteins/genetics , Dithiothreitol/chemistry , Dithiothreitol/pharmacology , HEK293 Cells , Humans , Ion Transport/drug effects , Ion Transport/physiology , Mutation , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Protein Multimerization/drug effects , Protein Structure, Quaternary , Protein Structure, Secondary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Xenopus laevis
7.
Cell Biochem Biophys ; 61(2): 251-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21499753

ABSTRACT

The solute carrier (SLC) family of transporters play key roles in the movement of charged organic ions across the blood-urine, blood-cerebrospinal fluid, and blood-brain barriers and thus mediate the absorption, disposition, and elimination of many common pharmaceuticals (i.e., nonsteroidal anti-inflammatory drug (NSAIDs), antibiotics, and diuretics). They have also been proposed to participate in a remote sensing and signaling network involving small molecules. Nevertheless, other than possessing a 12-transmembrane α-helical topology comprised of two six-helix hemidomains interacting through a long loop, the structural and mechanistic details for these transporters remains unclear. Recent crystallographic studies of bacterial homologs support the idea of a "switching" mechanism, which allows for periodic changes in the overall transporter configuration and cyclic opening of the transporter to the extracellular or cytoplasmic sides of the membrane. To investigate this, computational modeling based on our recent study of glycerol-3-phosphate transporter (GlpT) (Tsigelny et al. J Bioinform Comput Biol 6:885-904, 2008) was performed for organic anion transporter 1 (OAT1/SLC22A6, originally identified as NKT), the prototypical member of this family. OAT1 was inserted into an artificial phospholipid bilayer and the positional change of the six-helix hemidomains relative to each other was followed for 100 ns. The hemidomains were found to tilt relative to each other while their configuration is mostly inflexible. Since the modeling was performed for about 100 ns, the data suggest that this tilting mechanism might explain the early steps in the transport of organic anionic metabolites, drugs, and toxins by this clinically important transporter.


Subject(s)
Molecular Dynamics Simulation , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/metabolism , Pharmaceutical Preparations/metabolism , Amino Acid Sequence , Biological Transport , Cell Membrane/metabolism , Extracellular Space/metabolism , Lipid Bilayers/metabolism , Molecular Sequence Data , Phospholipids/metabolism , Protein Structure, Tertiary
8.
Mol Pharmacol ; 79(3): 569-74, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21159999

ABSTRACT

Human organic anion transporter 1 (hOAT1) plays a critical role in the body disposition of environmental toxins and clinically important drugs, including anti-HIV therapeutics, antitumor drugs, antibiotics, antihypertensives, and anti-inflammatories. We have demonstrated previously that hOAT1 forms homo-oligomers in cultured cells and in rat kidney. However, the functional consequence of such oligomerization has never been elucidated. In the current study, we used a novel approach by examining the effects of short hydrophobic peptides corresponding to transmembrane domains (TMDs) 1 to 12 of hOAT1 on the oligomerization and function of the transporter. We constructed expression vectors encoding short fusion peptides corresponding to TMDs 1 to 12 of hOAT1. These peptides were transfected into hOAT1-expressing COS-7 cells. Our results showed that among all 12 peptides examined, only the peptide corresponding to TMD 6 of hOAT1 significantly disrupted hOAT1 oligomerization demonstrated by cross-linking and coimmunoprecipitation experiments. The same peptide also caused a reduced expression of hOAT1 at the cell surface. As a result, hOAT1-mediated transport activity was compromised. Our data suggest that the peptide corresponding to TMD 6 of hOAT1 is a potent inhibitor of hOAT1 oligomerization and that oligomerization of hOAT1 is critical for the expression of the transporter at the cell surface and consequently for the proper function of the transporter.


Subject(s)
Organic Anion Transport Protein 1/antagonists & inhibitors , Protein Multimerization , Animals , Biotinylation , COS Cells , Chlorocebus aethiops , Cross-Linking Reagents/pharmacology , Electrophoresis, Polyacrylamide Gel , Humans , Immunoblotting , Immunoprecipitation , Membrane Proteins/metabolism , Organic Anion Transport Protein 1/chemistry , Peptides/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry
9.
J Pharmacol Exp Ther ; 332(2): 650-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19892921

ABSTRACT

Human organic anion transporter hOAT1 plays a critical role in the body disposition of clinically important drugs. In transmembrane segment (TM) 12, residues Tyr-490 and dileucine Leu-503/Leu-504 were identified to be critical for hOAT1 function. Substitution of Tyr-490 with alanine led to a dramatic reduction in protein expression of hOAT1 and its transport activity. The contribution of the side chain of Tyr-490 to transport activity was then evaluated by replacing this residue with Trp or Phe. Substitution of Tyr-490 with Trp or Phe partially or fully recovered the protein expression of hOAT1 and its transport activity, respectively, that were lost by substitution of Tyr-490 with alanine, suggesting that the aromatic ring and the size of the side chain of Tyr-490 are critical for hOAT1 expression and function. Studies with protease inhibitors and pulse-chase labeling further showed that the loss of expression of hOAT1 and its transport activity by replacing Tyr-490 with alanine resulted from accelerated degradation of the transporter, whereas its maturation efficiency was not affected. In contrast to Tyr-490, substitution of Leu-503/Leu-504 with alanine also resulted in complete loss of protein expression of hOAT1 and its transport activity. However, such loss of protein expression could not be prevented by treating mutant-expressing cells with protease inhibitors. Pulse-chase experiments showed that the mutant transporter (L503/L504A) was trapped in the endoplasmic reticulum without conversion into mature form of the transporter. Our results are the first to highlight the central role of TM 12 in maintaining the stability and in promoting the maturation efficiency of hOAT1.


Subject(s)
Organic Anion Transport Protein 1/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , COS Cells , Chlorocebus aethiops , Humans , LLC-PK1 Cells , Mutagenesis, Site-Directed , Mutant Proteins , Organic Anion Transport Protein 1/biosynthesis , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/physiology , Swine , Transfection
10.
J Biol Chem ; 284(45): 31422-30, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19737926

ABSTRACT

Studies of the organic anion transporters (Oats) have focused mainly on their interactions with organic anionic substrates. However, as suggested when Oat1 was originally identified as NKT (Lopez-Nieto, C. E., You, G., Bush, K. T., Barros, E. J., Beier, D. R., and Nigam, S. K. (1997) J. Biol. Chem. 272, 6471-6478), since the Oats share close homology with organic cation transporters (Octs), it is possible that Oats interact with cations as well. We now show that mouse Oat1 (mOat1) and mOat3 and, to a lesser degree, mOat6 bind a number of "prototypical" Oct substrates, including 1-methyl-4-phenylpyridinium. In addition to oocyte expression assays, we have tested binding of organic cations to Oat1 and Oat3 in ex vivo assays by analyzing interactions in kidney organ cultures deficient in Oat1 and Oat3. We also demonstrate that mOat3 transports organic cations such as 1-methyl-4-phenylpyridinium and cimetidine. A pharmacophore based on the binding affinities of the tested organic cations for Oat3 was generated. Using this pharmacophore, we screened a chemical library and were able to identify novel cationic compounds that bound to Oat1 and Oat3. These compounds bound Oat3 with an affinity higher than the highest affinity compounds in the original set of prototypical Oct substrates. Thus, whereas Oat1, Oat3, and Oat6 appear to function largely in organic anion transport, they also bind and transport some organic cations. These findings could be of clinical significance, since drugs and metabolites that under normal physiological conditions do not bind to the Oats may undergo changes in charge and become Oat substrates during pathologic conditions wherein significant variations in body fluid pH occur.


Subject(s)
Cations/metabolism , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters/metabolism , Animals , Anions/metabolism , Biological Transport , Kidney/chemistry , Kidney/metabolism , Kinetics , Mice , Mice, Knockout , Models, Molecular , Oocytes/chemistry , Oocytes/metabolism , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters/chemistry , Organic Anion Transporters/genetics , Protein Binding , Xenopus laevis
11.
Article in English | MEDLINE | ID: mdl-17977807

ABSTRACT

This paper reports the development of liquid chromatographic columns containing immobilized organic anion transporters (hOAT1 and hOAT2). Cellular membrane fragments from MDCK cells expressing hOAT1 and S2 cells expressing hOAT2 were immobilized on the surface of the immobilized artificial membrane (IAM) liquid chromatographic stationary phase. The resulting stationary phases were characterized by frontal affinity chromatography, using the marker ligand [3H]-adefovir for the hOAT1 and [14C]-p-aminohippurate for the hOAT2 in the presence of multiple displacers. The determined binding affinities (Kd) for eight OAT1 ligands and eight OAT2 ligands were correlated with literature values and a statistically significant correlation was obtained for both the hOAT1 and hOAT2 columns: r2=0.688 (p<0.05) and r2=0.9967 (p<0.0001), respectively. The results indicate that the OAT1 and OAT2 have been successfully immobilized with retention of their binding activity. The use of these columns to identify ligands to the respective transporters will be presented.


Subject(s)
Chromatography, Liquid/methods , Organic Anion Transport Protein 1/chemistry , Organic Anion Transporters, Sodium-Independent/chemistry , Animals , Cell Line , Chromatography, Affinity , Chromatography, Liquid/instrumentation , Protein Binding
12.
Biochem J ; 401(2): 515-20, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17014423

ABSTRACT

The OAT (organic anion transporter) family mediates the absorption, distribution and excretion of a diverse array of environmental toxins and clinically important drugs. OAT dysfunction significantly contributes to renal, hepatic, neurological and fetal toxicity and disease. As a first step to establish the topological model of hOAT1 (human OAT1), we investigated the external loops and the cellular orientation of the N- and the C-termini of this transporter. Combined approaches of immunofluorescence studies and site-directed chemical labelling were used for such purpose. Immunofluorescence microscopy of Myc-tagged hOAT1 expressed in cultured cells identified that both the N- and the C-termini of the transporter were located in the cytoplasm. Replacement of Lys59 in the predicted extracellular loop I with arginine resulted in a mutant (K59R), which was largely inaccessible for labelling by membrane-impermeable NHS (N-hydroxysuccinimido)-SS (dithio)-biotin present in the extracellular medium. This result suggests that loop I faces outside of the cell membrane. A single lysine residue introduced into putative extracellular loops III, V and VI of mutant K59R, which is devoid of extracellular lysine, reacted readily with membrane-impermeable NHS-SS-biotin, suggesting that these putative extracellular loops are in the extracellular domains of the protein. These studies provided the first experimental evidence on the extracellular loops and the cellular orientation of the N- and the C-termini of hOAT1.


Subject(s)
Organic Anion Transport Protein 1/chemistry , p-Aminohippuric Acid/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Epitopes/genetics , Fluorescent Antibody Technique , Humans , Lysine/chemistry , Organic Anion Transport Protein 1/genetics , Proto-Oncogene Proteins c-myc/genetics , Transfection
13.
J Biol Chem ; 281(49): 38071-9, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17038320

ABSTRACT

Organic anion transporters (OATs) play a critical role in the handling of endogenous and exogenous organic anions by excretory and barrier tissues. Little is known about the OAT three-dimensional structure or substrate/protein interactions involved in transport. In this investigation, a theoretical three-dimensional model was generated for human OAT1 (hOAT1) based on fold recognition to the crystal structure of the glycerol 3-phosphate transporter (GlpT) from Escherichia coli. GlpT and hOAT1 share several sequence motifs as major facilitator superfamily members. The structural hOAT1 model shows that helices 5, 7, 8, 10, and 11 surround an electronegative putative active site ( approximately 830A(3)). The site opens to the cytoplasm and is surrounded by three residues not previously examined for function (Tyr(230) (domain 5) and Lys(431) and Phe(438) (domain 10)). Effects of these residues on p-aminohippurate (PAH) and cidofovir transport were assessed by point mutations in a Xenopus oocyte expression system. Membrane protein expression was severely limited for the Y230A mutant. For the K431A and F438A mutants, [(3)H]PAH uptake was less than 30% of wild-type hOAT1 uptake after protein expression correction. Reduced V(max) values for the F438A mutant confirmed lower protein expression. In addition, the F438A mutant exhibited an increased affinity for cidofovir but was not significantly different for PAH. Differences in handling of PAH and cidofovir were also observed for the Y230F mutant. Little uptake was determined for cidofovir, whereas PAH uptake was similar to wild-type hOAT1. Therefore, the hOAT1 structural model has identified two new residues, Tyr(230) and Phe(438), which are important for substrate/protein interactions.


Subject(s)
Organic Anion Transport Protein 1/chemistry , Amino Acid Sequence , Amino Acid Substitution , Amino Acids, Aromatic/chemistry , Animals , Base Sequence , Binding Sites/genetics , DNA Primers/genetics , Female , Humans , In Vitro Techniques , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/metabolism , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Static Electricity , Xenopus laevis
14.
J Biol Chem ; 281(42): 31178-83, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16920720

ABSTRACT

Human organic anion transporter hOAT1 plays critical roles in the body disposition of environmental toxins and clinically important drugs. In the present study, we examined the role of the C terminus of hOAT1 in its function. Combined approaches of cell surface biotinylation and transport analysis were employed for such purposes. It was found that deletion of the last 15 amino acids (residues 536-550) or the last 30 amino acids (residues 521-550) had no significant effect on transport activity. However, deletion of the entire C terminus (residues 506-550) completely abolished transport activity. Alanine scanning mutagenesis within the region of amino acids 506-520 led to the discovery of two critical amino acids: Glu-506 and Leu-512. Substitution of negatively charged Glu-506 with neutral amino acids alanine or glutamine resulted in complete loss of transport activity. However, such loss of transport activity could be rescued by substitution of Glu-506 with another negatively charged amino acid aspartic acid, suggesting the importance of negative charge at this position for maintaining the correct tertiary structure of the transporter, possibly by forming a salt bridge with a positively charged amino acid. Substitution of Leu-512 with amino acids carrying progressively smaller side chains including isoleucine, valine, and alanine resulted in mutants (L512I, L512V, and L512A) with increasingly impaired transport activity. However, the cell surface expression of these mutants was not affected. Kinetic analysis of mutant L512V revealed that the reduced transport activity of this mutant resulted mainly from a reduced maximum transport velocity Vmax without affecting the binding affinity (1/Km) of the transporter for its substrates, suggesting that the size of the side chain at position 512 critically affects transporter turnover number. Together, our results are the first to highlight the central role of the C terminus of hOAT1 in the function of this transporter.


Subject(s)
Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/physiology , Amino Acid Sequence , Amino Acids/chemistry , Animals , Biological Transport , Biotinylation , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Humans , Molecular Sequence Data , Mutagenesis , Protein Conformation , Protein Structure, Tertiary
15.
Am J Physiol Regul Integr Comp Physiol ; 291(6): R1773-80, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16857889

ABSTRACT

The flounder renal organic anion transporter (fOat) has substantial sequence homology to mammalian basolateral organic anion transporter orthologs (OAT1/Oat1 and OAT3/Oat3), suggesting that fOat may have functional properties of both mammalian forms. We therefore compared uptake of various substrates by rat Oat1 and Oat3 and human OAT1 and OAT3 with the fOat clone expressed in Xenopus oocytes. These data confirm that estrone sulfate is an excellent substrate for mammalian OAT3/Oat3 transporters but not for OAT1/Oat1 transporters. In contrast, 2,4-dichlorophenoxyacetic acid and adefovir are better transported by mammalian OAT1/Oat1 than by the OAT3/Oat3 clones. All three substrates were well transported by fOat-expressing Xenopus oocytes. fOat K(m) values were comparable to those obtained for mammalian OAT/Oat1/3 clones. We also characterized the ability of these substrates to inhibit uptake of the fluorescent substrate fluorescein in intact teleost proximal tubules isolated from the winter flounder (Pseudopleuronectes americanus) and killifish (Fundulus heteroclitus). The rank order of the IC(50) values for inhibition of cellular fluorescein accumulation was similar to that for the K(m) values obtained in fOat-expressing oocytes, suggesting that fOat may be the primary teleost renal basolateral Oat. Assessment of the zebrafish (Danio rerio) genome indicated the presence of a single Oat (zfOat) with similarity to both mammalian OAT1/Oat1 and OAT3/Oat3. The puffer fish (Takifugu rubripes) also has an Oat (pfOat) similar to mammalian OAT1/Oat1 and OAT3/Oat3 members. Furthermore, phylogenetic analyses argue that the teleost Oat1/3-like genes diverged from a common ancestral gene in advance of the divergence of the mammalian OAT1/Oat1, OAT3/Oat3, and, possibly, Oat6 genes.


Subject(s)
Flounder/genetics , Flounder/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Amino Acid Sequence , Animals , Evolution, Molecular , Humans , Molecular Sequence Data , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/chemistry , Sequence Homology, Amino Acid , Species Specificity , Substrate Specificity
16.
J Biol Chem ; 280(37): 32285-90, 2005 Sep 16.
Article in English | MEDLINE | ID: mdl-16046403

ABSTRACT

Human organic anion transporter hOAT1 belongs to a superfamily of organic anion transporters, which play critical roles in the body disposition of clinically important drugs, including anti-human immunodeficiency virus therapeutics, anti-tumor drugs, antibiotics, anti-hypertensives, and anti-inflammatories. To gain insight into the regulation of hOAT1, detailed information on its structural assembly is essential. In the present study, we investigate the quaternary structure of hOAT1 using combined approaches of chemical cross-linking, gel filtration chromatography, co-immunoprecipitation, cell surface biotinylation, and metabolic labeling. Chemical cross-linking of intact membrane proteins from LLC-PK1 cells stably expressing hOAT1 converted quantitatively hOAT1 monomer to putative trimer and higher order of oligomer, indicating that hOAT1 is present in the membrane as multimeric complexes. When co-expressed in LLC-PK1 cells, FLAG-tagged hOAT1 co-immunoprecipitated with myc-tagged hOAT1. The hOAT1 oligomer was also detected in gel filtration chromatography of total membranes from hOAT1-expressing LLC-PK1 cells. Cell surface biotinylation with membrane-impermeable reagents and metabolic labeling with [(35)S]methionine followed by immunoprecipitation showed that the oligomeric hOAT1 did not contain any other proteins. Taken together, this is the first study demonstrating that hOAT1 exists in the plasma membrane as a homooligomer, possibly trimer, and higher order of oligomer.


Subject(s)
Cell Membrane/metabolism , Organic Anion Transport Protein 1/physiology , Animals , Biological Transport , Biotinylation , Cell Line , Cell Line, Transformed , Chromatography , Chromatography, Gel , Cross-Linking Reagents/pharmacology , Dimerization , Electrophoresis, Polyacrylamide Gel , Epitopes/chemistry , Humans , Immunoblotting , Immunoprecipitation , Kidney/metabolism , Membrane Proteins/chemistry , Organic Anion Transport Protein 1/chemistry , Protein Binding , Protein Structure, Quaternary , Proto-Oncogene Proteins c-myc/metabolism , Rats , Swine
17.
Mol Pharmacol ; 67(5): 1600-11, 2005 May.
Article in English | MEDLINE | ID: mdl-15662044

ABSTRACT

To identify functionally relevant amino acids in the rat organic cation transporter 1 (rOCT1), 18 consecutive amino acids in the presumed fourth transmembrane alpha helix (TMH) were mutated and functionally characterized after expression in Xenopus laevis oocytes. After mutation of three amino acids on successive turns of the alpha helix, K(m) values for tetraethylammonium (TEA) and/or 1-methyl-4-phenylpyridinium (MPP) were decreased. After replacement of Trp218 by tyrosine (W218Y) and Tyr222 by leucine (Y222L), the K(m) values for both TEA and MPP were decreased. In mutants Y222F and T226A, only the K(m) values for TEA and MPP were decreased, respectively. The data suggest that amino acids Trp218 and Tyr222 participate in the binding of both TEA and MPP, whereas Thr226 is only involved in the binding of MPP. Using the crystal structure of the lactose permease LacY from Escherichia coli that belongs to the same major facilitator superfamily as rOCT1, we modeled the tertiary structure of the presumed 12 transmembrane alpha helices. The validity of the model was suggested because seven amino acids that have been shown to participate in the binding of cations by mutagenesis experiments [fourth TMH Trp218, Tyr222, and Thr226 (this work); 10th TMH Ala443, Leu447, and Gln448 (companion work in this issue of Molecular Pharmacology); 11th TMH Asp475 (previous report)] are located in one region surrounding a large cleft that opens to the intracellular side. The dimensions of TEA in comparison with the interacting amino acids in the modeled cleft suggest that more than one TEA molecule can bind in parallel to the modeled conformation of the transporter.


Subject(s)
Amino Acids/metabolism , Membrane Transport Proteins/metabolism , Models, Molecular , Organic Anion Transport Protein 1/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Animals , Female , Membrane Transport Proteins/chemistry , Molecular Sequence Data , Organic Anion Transport Protein 1/chemistry , Protein Structure, Tertiary/physiology , Substrate Specificity/physiology , Xenopus laevis
18.
J Biol Chem ; 279(30): 31478-82, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15145940

ABSTRACT

Human organic anion transporter 1 (hOAT1) belongs to a superfamily of organic anion transporters, which play critical roles in the body disposition of clinically important drugs, including anti-human immunodeficiency virus therapeutics, anti-tumor drugs, antibiotics, anti-hypertensives, and anti-inflammatories. Previously we suggested that the predicted transmembrane domain 1 (TM1) of hOAT1 might be important for its function. In the present study, we examined the role of each residue within TM1 of hOAT1 in substrate recognition and transport. Alanine scanning was used to construct mutants of hOAT1, and the uptake of model substrate para-aminohippurate was studied in COS-7 cells expressing the mutant transporters. This approach led to the discovery of two critical amino acid residues, Leu-30 and Thr-36. A substitution of Leu-30 or Thr-36 with alanine resulted in a complete loss of transport activities. We then further characterized Leu-30 and Thr-36 by mutagenizing these residues to amino acids with different physicochemical properties. Leu-30 was replaced with amino acids with varying sizes of side chains, including glycine, valine, and isoleucine. We showed that progressively smaller side chains at position 30 increasingly impaired hOAT1 function mainly because of the impaired surface expression of the transporter. Thr-36, another critical amino acid in TM1, was replaced by serine and cysteine. Similar to the substitution of Thr-36 by alanine, substitution by serine and cysteine at this position abolished transport activity without affecting the surface expression of the transporter. The fact that Thr-36 cannot be substituted with serine and that the side chains of alanine, serine, and cysteine are smaller than that of threonine by a methyl group indicate that both the methyl group and the hydroxyl group of Thr-36 could be critical for hOAT1 activity. Together we conclude that Leu-30 and Thr-36 play distinct roles in hOAT1 function. Leu-30 is important in targeting the transporter to the plasma membrane. In contrast, Thr-36 is critical for substrate recognition. The present study provided the first molecular evidence that transmembrane domain 1 is a critical determinant of hOAT1 function and may provide important insights into the structure-function relationships of the organic anion transporter family.


Subject(s)
Organic Anion Transport Protein 1/chemistry , Amino Acid Substitution , Animals , Binding Sites/genetics , COS Cells , Cell Membrane/metabolism , Humans , In Vitro Techniques , Leucine/chemistry , Mutagenesis, Site-Directed , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Threonine/chemistry
19.
Drug Metab Dispos ; 32(4): 424-30, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15039295

ABSTRACT

The human organic anion transporter 1 (hOAT1) facilitates the basolateral entry of organic anions such as endogenous metabolites, xenobiotics, and drugs into the proximal tubule cells. In the present study we investigated the general occurrence of hOAT1 isoforms in the kidneys and performed functional characterizations. Kidney specimens of 10 patients were analyzed by reverse transcription-polymerase chain reaction. We detected hOAT1-2 as the main transcript in almost all patients, and weak transcripts of hOAT1-1, hOAT1-3, and hOAT1-4 in many of them. An evaluation of the renal distribution showed all four mRNAs mostly restricted to the cortex. Western blot analysis of membrane fractions from two kidney specimens yielded two bands corresponding to the observed mRNA expression, suggesting hOAT1-3 and hOAT1-4 to be expressed on the protein level in vivo. This observation is further supported by immunofluorescence analyses of all four cloned hOAT1 isoforms transiently transfected in COS 7 cells. Functional characterizations did not show any transport activity of hOAT1-3 and hOAT1-4 for the tested substrates. Cotransfection studies of each of them with hOAT1-1 did not alter fluorescein uptake indicating no regulatory impact of these isoforms. Further functional comparisons of hOAT1-1 and hOAT1-2 in fluorescein uptake studies exhibited almost identical affinities for fluorescein with Michaelis constants of 11.6 +/- 3.7 microM (hOAT1-1) and 11.9 +/- 6.4 microM (hOAT1-2), and similar sensitivities to inhibition by p-aminohippurate [IC(50): 16 microM (hOAT1-1), 10 microM (hOAT1-2)], urate [IC(50): 440 microM (hOAT1-1), 385 microM (hOAT1-2)], and furosemide (IC(50): 14 microM (hOAT1-1), 20 microM (hOAT1-2)], implying functional equivalence.


Subject(s)
Kidney Cortex/cytology , Organic Anion Transport Protein 1/genetics , Protein Isoforms/genetics , Up-Regulation/genetics , Animals , Blotting, Western/methods , COS Cells , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/genetics , Chlorocebus aethiops , Cloning, Organism/methods , Fluorescein/metabolism , Fluorescent Antibody Technique/methods , Furosemide/pharmacokinetics , Humans , Kidney Cortex/drug effects , Kidney Cortex/physiology , Models, Molecular , Organic Anion Transport Protein 1/chemistry , Organic Anion Transport Protein 1/drug effects , Protein Isoforms/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Subcellular Fractions/chemistry , Subcellular Fractions/drug effects , Transfection/methods , Uric Acid/chemistry , Uric Acid/pharmacokinetics , p-Aminohippuric Acid/pharmacokinetics
20.
J Biol Chem ; 279(15): 14961-6, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-14749323

ABSTRACT

Organic anion transporters (OAT) play essential roles in the body disposition of clinically important anionic drugs, including antiviral drugs, antitumor drugs, antibiotics, antihypertensives, and anti-inflammatories. We reported previously (Kuze, K., Graves, P., Leahy, A., Wilson, P., Stuhlmann, H., and You, G. (1999) J. Biol. Chem. 274, 1519-1524) that tunicamycin, an inhibitor of asparagine-linked glycosylation, significantly inhibited organic anion transport in COS-7 cells expressing a mouse organic anion transporter (mOAT1), suggesting an important role of glycosylation in mOAT1 function. In the present study, we investigated the effect of disrupting putative glycosylation sites in mOAT1 as well as its human counterpart, hOAT1, by mutating asparagine to glutamine and assessing mutant transporters in HeLa cells. We showed that the putative glycosylation site Asp-39 in mOAT1 was not glycosylated but the corresponding site (Asp-39) in hOAT1 was glycosylated. Disrupting Asp-39 resulted in a complete loss of transport activity in both mOAT1 and hOAT1 without affecting their cell surface expression, suggesting that the loss of function is not because of deglycosylation of Asp-39 per se but rather is likely because of the change of this important amino acid critically involved in the substrate binding. Single replacement of asparagines at other sites had no effect on transport activity indicating that glycosylation at individual sites is not essential for OAT function. In contrast, a simultaneous replacement of all asparagines in both mOAT1 and hOAT1 impaired the trafficking of the transporters to the plasma membrane. In summary, we provided the evidence that 1) Asp-39 is crucially involved in substrate recognition of OAT1, 2) glycosylation at individual sites is not required for OAT1 function, and 3) glycosylation plays an important role in the targeting of OAT1 onto the plasma membrane. This study is the first molecular identification and characterization of glycosylation of OAT1 and may provide important insights into the structure-function relationships of the organic anion transporter family.


Subject(s)
Organic Anion Transport Protein 1/chemistry , Animals , Anions , Asparagine/chemistry , Binding Sites , Biological Transport , Biotinylation , Blotting, Western , COS Cells , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Glutamine/chemistry , Glycosylation , HeLa Cells , Humans , Immunoblotting , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Mutagenesis, Site-Directed , Mutation , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...