Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
PLoS One ; 16(5): e0250408, 2021.
Article in English | MEDLINE | ID: mdl-33945567

ABSTRACT

Gyps species have been previously shown to be highly sensitive to the toxic effects of diclofenac, when present in their food sources as drug residues following use as a veterinary medicine. Vultures exposed to diclofenac soon become depressed and die with signs of severe visceral gout and renal damage on necropsy. The molecular mechanism behind toxicity and renal excretion of uric acid is still poorly understood. With the clinical pictures suggesting renal uric acid excretion as the target site for toxicity, as a first step the following study was undertaken to determine the uric acid excretory pathways present in the African white-backed vulture (Gyps africanus) (AWB), one of the species susceptible to toxicity. Using transcriptome analysis, immunohistochemistry and functional predictions, we demonstrated that AWB makes use of the organic anion transporter 2 (OAT2) for their uric acid excretion. RT-qPCR analysis subsequently demonstrated relatively similar expression of the OAT2 transporter in the vulture and chicken. Lastly docking analysis, predicted that the non-steroidal drugs induce their toxicity through an allosteric binding.


Subject(s)
Avian Proteins/metabolism , Kidney/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Birds , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Transcriptome , Uric Acid/metabolism
2.
Int J Mol Sci ; 21(8)2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32326111

ABSTRACT

The liver plays a pivotal role in drug handling due to its contribution to the processes of detoxification (phases 0 to 3). In addition, the liver is also an essential organ for the mechanism of action of many families of drugs, such as cholesterol-lowering, antidiabetic, antiviral, anticoagulant, and anticancer agents. Accordingly, the presence of genetic variants affecting a high number of genes expressed in hepatocytes has a critical clinical impact. The present review is not an exhaustive list but a general overview of the most relevant variants of genes involved in detoxification phases. The available information highlights the importance of defining the genomic profile responsible for the hepatic handling of drugs in many ways, such as (i) impaired uptake, (ii) enhanced export, (iii) altered metabolism due to decreased activation of prodrugs or enhanced inactivation of active compounds, and (iv) altered molecular targets located in the liver due to genetic changes or activation/downregulation of alternative/compensatory pathways. In conclusion, the advance in this field of modern pharmacology, which allows one to predict the outcome of the treatments and to develop more effective and selective agents able to overcome the lack of effect associated with the existence of some genetic variants, is required to step forward toward a more personalized medicine.


Subject(s)
Genetic Variation , Inactivation, Metabolic/genetics , Liver/metabolism , Pharmacogenomic Variants , Alleles , Animals , Humans , Metabolic Detoxication, Phase I/genetics , Metabolic Detoxication, Phase II/genetics , Mutation , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Oxidation-Reduction , Polymorphism, Single Nucleotide
3.
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
4.
PLoS One ; 12(6): e0180257, 2017.
Article in English | MEDLINE | ID: mdl-28644885

ABSTRACT

OATP1B3 is a 12 transmembrane domain protein expressed at the basolateral membrane of human hepatocytes where it mediates the uptake of numerous drugs and endogenous compounds. Previous western blot results suggest the formation of OATP1B3 multimers. In order to better understand the function of OATP1B3 under normal physiological conditions, we investigated its oligomerization status. We transiently transfected OATP1B3 with a C-terminal His-, FLAG- or HA-tag in HEK293 cells and used co-immunoprecipitation and a Proximity Ligation Assay to detect interactions between the different constructs. All three constructs retained similar transport rates as wild-type OATP1B3. Immunofluorescence experiments indicated that in contrast to wild-type, His- and FLAG-tagged OATP1B3, where the C-terminal end is on the cytoplasmic side of the membrane, the C-terminal end of HA-tagged OATP1B3 is extracellular. After cross-linking, anti-FLAG antibodies were able to pull down FLAG-tagged OATP1B3 (positive control) and co-transfected His- or HA-tagged OATP1B3, demonstrating the formation of homo-oligomers and suggesting that the C-terminal part is not involved in oligomer formation. We confirmed co-localization of His- and FLAG-tagged OATP1B3 in transfected HEK293 cells with the Proximity Ligation Assay. Transport studies with a non-functional OATP1B3 mutant suggest that the individual subunits and not the whole oligomer are the functional units in the homo-oligomers. In addition, we also detected OATP1B3-FLAG co-localization with OATP1B1-His or NTCP-His, suggesting that OATP1B3 also hetero-oligomerizes with other transport proteins. Using the Proximity Ligation Assay with transporter specific antibodies, we demonstrate close association of OATP1B3 with NTCP in frozen human liver tissue. These findings demonstrate that OATP1B3 can form homo- and hetero-oligomers and suggest a potential co-regulation of the involved transporters.


Subject(s)
Organic Anion Transporters, Sodium-Independent/metabolism , Blotting, Western , Cytoplasm/metabolism , Extracellular Space/metabolism , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoprecipitation , Liver-Specific Organic Anion Transporter 1/metabolism , Mutation , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Protein Domains , Protein Multimerization , Solute Carrier Organic Anion Transporter Family Member 1B3 , Symporters/metabolism , Transfection
5.
Biochem Pharmacol ; 131: 98-105, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28216016

ABSTRACT

Organic anion transporting polypeptide 1B3 (OATP1B3) is a major influx transporter mediating the hepatic uptake of various endogenous substrates as well as clinically important drugs such as statins and anticancer drugs. However, molecular mechanisms controlling the membrane trafficking of OATP1B3 have been largely unknown. Several reports recently indicated the presence of a distinct, cancer-type OATP1B3 variant lacking the N-terminal 28 amino acids compared to OATP1B3 expressed in non-malignant hepatocytes. Interestingly, the cancer-type OATP1B3 variant is located predominantly in the cytoplasm, implicating the involvement of the N-terminal region of OATP1B3 in its membrane trafficking. In the current study, we set out to experimentally validate the importance of the N-terminal region of OATP1B3 and to identify responsible sequence motif(s) in that region. A number of truncation or point mutants of OATP1B3 were transiently expressed in HEK293T, HCT-8 or MDCK II cells and their expression in cytoplasmic and surface membrane fractions were analyzed by immunoblotting. Our results indicated that the N-terminal sequence of OATP1B3, in particular, at the amino acid positions between 12 and 28, may be indispensable in its membrane trafficking. Moreover, our results using a fusion construct indicated that the first 50 amino acids of OATP1B3 are sufficient for its membrane localization. The importance of the N-terminal region in membranous localization was shared among the other OATP1B subfamily members, OATP1B1 and rat Oatp1b2. Our efforts to identify the responsible amino acid(s) or structure motif(s) in the N-terminal region did not pinpoint individual amino acids or motifs with putative secondary structures. Our current findings however demonstrate that the N-terminal region is important for the membrane localization of the OATP1B subfamily members and should facilitate future investigations of the mechanisms involved in the regulation and membrane trafficking of these important transporter proteins.


Subject(s)
Organic Anion Transporters, Sodium-Independent/physiology , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Humans , Mice , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Phosphorylation , Point Mutation , Protein Transport , Rats , Sequence Homology, Amino Acid , Solute Carrier Organic Anion Transporter Family Member 1B3 , Subcellular Fractions/metabolism
6.
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
7.
Anticancer Res ; 35(11): 5857-65, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26504008

ABSTRACT

Microcystin-LR (MC-LR) is a cyanobacterial cyclopeptide, known for its unique ability to cause acute liver injury. Its cellular uptake is facilitated by specific transmembrane organic anion-transporting polypeptides (OATPs) specifically OATP1B1 and 1B3. The objective of the present study was to investigate the expression of OATPs 1A2, 1B1 and 1B3 in pancreatic cancer cell lines BxPC-3 and MIA PACA-2 and assess their role in MC-LR-mediated cytotoxicity by using the novel xCELLigence system and flow cytometry. OATP1B1 and 1B3 were found to be expressed in both cell lines at both the mRNA and protein levels. The cytotoxic effects of MC-LR were proportionally related to the expression of these transporters. Moreover the cytotoxic potency of MC-LR was found superior to gemcitabine. Based on the expression of the organic anion transporting polypeptides 1B1 and 1B3 in pancreatic carcinoma tissue and cell lines and the potent cytotoxicity induced by MC-LR in vitro, we propose that this molecule could be held as structural basis for the development of novel targeted-compounds against pancreatic cancer.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Microcystins/pharmacology , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/metabolism , Pancreatic Neoplasms/pathology , Amino Acid Sequence , Biological Transport , Blotting, Western , Cell Proliferation/drug effects , Flow Cytometry , Humans , Immunoenzyme Techniques , Liver-Specific Organic Anion Transporter 1 , Marine Toxins , Molecular Docking Simulation , Molecular Sequence Data , Organic Anion Transporters/chemistry , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Conformation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Solute Carrier Organic Anion Transporter Family Member 1B3 , Tumor Cells, Cultured
8.
Eur J Med Chem ; 92: 723-31, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25618019

ABSTRACT

Antineoplastic compounds are used in the treatment of a variety of cancers. The effectiveness of an antineoplastic compound to exert its activity is largely dependent on transport proteins involved in the entry of the compound into the cells, and those which drive it out of the cell. Organic anion transporting polypeptide 1B1 (OATP1B1) and organic anion transporting polypeptide 1B3 (OATP1B3), belonging to the SLCO family of proteins, are specifically expressed in the sinusoidal membranes of the liver, and are known to interact with a variety of drugs. The present study deals with the interaction of these proteins with antineoplastic compounds routinely used in cancer chemotherapy. The proteins OATP1B1 and OATP1B3 were functionally characterized in stably transfected human embryonic kidney cells using [(3)H] labeled estrone 3-sulfate and [(3)H] labeled cholecystokinin octapeptide (CCK-8) as substrates, respectively. Substrate uptake experiments performed in the presence of antineoplastic compounds showed that vinblastine and paclitaxel strongly interacted with the OATP1B1 with Ki values of 10.2 µM and 0.84 µM, respectively. OATP1B3 showed highly significant interactions with a variety of antineoplastic compounds including chlorambucil, mitoxantrone, vinblastine, vincristine, paclitaxel and etoposide, with Ki values of 40.6 µM, 3.2 µM, 15.9 µM, 30.6 µM, 1.8 µM and 13.5 µM, respectively. We report several novel interactions of the transporter proteins OATP1B1 and OATP1B3 highlighting the need to investigate their role in drug-drug interactions and cancer chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/metabolism , Peptides/metabolism , Antineoplastic Combined Chemotherapy Protocols/chemical synthesis , Antineoplastic Combined Chemotherapy Protocols/chemistry , Cell Line , Humans , Liver-Specific Organic Anion Transporter 1 , Molecular Structure , Organic Anion Transporters/chemistry , Organic Anion Transporters, Sodium-Independent/chemistry , Peptides/chemistry , Solute Carrier Organic Anion Transporter Family Member 1B3
9.
PLoS One ; 9(4): e94926, 2014.
Article in English | MEDLINE | ID: mdl-24740327

ABSTRACT

Analogs of the cyclic nucleotides cAMP and cGMP have been extensively used to mimic or modulate cellular events mediated by protein kinase A (PKA), Exchange protein directly activated by cAMP (Epac), or protein kinase G (PKG). We report here that some of the most commonly used cyclic nucleotide analogs inhibit transmembrane transport mediated by the liver specific organic anion transporter peptides OATP1B1 and OATP1B3, unrelated to actions on Epac, PKA or PKG. Several cAMP analogs, particularly with 8-pCPT-substitution, inhibited nodularin (Nod) induced primary rat hepatocyte apoptosis. Inhibition was not mediated by PKA or Epac, since increased endogenous cAMP, and some strong PKA- or Epac-activating analogs failed to protect cells against Nod induced apoptosis. The cAMP analogs inhibiting Nod induced hepatocyte apoptosis also reduced accumulation of radiolabeled Nod or cholic acid in primary rat hepatocytes. They also inhibited Nod induced apoptosis in HEK293 cells with enforced expression of OATP1B1 or 1B3, responsible for Nod transport into cells. Similar results were found with adenosine analogs, disconnecting the inhibitory effect of certain cAMP analogs from PKA or Epac. The most potent inhibitors were 8-pCPT-6-Phe-cAMP and 8-pCPT-2'-O-Me-cAMP, whereas analogs like 6-MB-cAMP or 8-Br-cAMP did not inhibit Nod uptake. This suggests that the addition of aromatic ring-containing substituents like the chloro-phenyl-thio group to the purines of cyclic nucleotides increases their ability to inhibit the OATP-mediated transport. Taken together, our data show that aromatic ring substituents can add unwanted effects to cyclic nucleotides, and that such nucleotide analogs must be used with care, particularly when working with cells expressing OATP1B1/1B3, like hepatocytes, or intact animals where hepatic metabolism can be an issue, as well as certain cancer cells. On the other hand, cAMP analogs with substituents like bromo, monobutyryl were non-inhibitory, and could be considered an alternative when working with cells expressing OATP1 family members.


Subject(s)
Bacterial Toxins/metabolism , Hepatocytes/drug effects , Nucleotides, Cyclic/pharmacology , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/metabolism , Animals , Apoptosis/drug effects , Bacterial Toxins/pharmacokinetics , Bacterial Toxins/pharmacology , Biological Transport/drug effects , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Glycocholic Acid/metabolism , Glycocholic Acid/pharmacokinetics , Glycocholic Acid/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver-Specific Organic Anion Transporter 1 , Male , Microscopy, Confocal , Models, Molecular , Nucleotides, Cyclic/chemistry , Organic Anion Transporters/chemistry , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/pharmacology , Protein Structure, Tertiary , Rats, Wistar , Solute Carrier Organic Anion Transporter Family Member 1B3
10.
Mol Pharm ; 9(4): 986-95, 2012 Apr 02.
Article in English | MEDLINE | ID: mdl-22352740

ABSTRACT

The human organic anion-transporting polypeptides OATP1B1 (SLCO1B1) and OATP1B3 (SLCO1B3) are liver-enriched membrane transporters of major importance to hepatic uptake of numerous endogenous compounds, including bile acids, steroid conjugates, hormones, and drugs, including the 3-hydroxy-3-methylglutaryl Co-A reductase inhibitor (statin) family of cholesterol-lowering compounds. Despite their remarkable substrate overlap, there are notable exceptions: in particular, the gastrointestinal peptide hormone cholecystokinin-8 (CCK-8) is a high affinity substrate for OATP1B3 but not OATP1B1. We utilized homologous recombination of linear DNA by E. coli to generate a library of cDNA containing monomer size chimeric OATP1B1-1B3 and OATP1B3-1B1 transporters with randomly distributed chimeric junctions to identify three discrete regions of the transporter involved in conferring CCK-8 transport activity. Site-directed mutagenesis of three key residues in OATP1B1 transmembrane helices 1 and 10, and extracellular loop 6, to the corresponding residues in OATP1B3, resulted in a gain of CCK-8 transport by OATP1B1. The residues appear specific to CCK-8, as the mutations did not affect transport of the shared OATP1B substrate atorvastatin or the OATP1B1-specific substrate estrone sulfate. Regions involved in gain of CCK-8 transport by OATP1B1, when mapped to the crystal structures of bacterial transporters from the major facilitator superfamily, are positioned to suggest these regions could readily interact with drug substrates. Accordingly, our data provide new insight into the molecular determinants of the substrate specificity of these hepatic uptake transporters with relevance to targeted drug design and prediction of drug-drug interactions.


Subject(s)
Amino Acids/metabolism , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/chemistry , Organic Anion Transporters/metabolism , Amino Acids/chemistry , Amino Acids/genetics , Cholecystokinin/metabolism , Liver-Specific Organic Anion Transporter 1 , Mutagenesis, Site-Directed , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Peptide Fragments/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3 , Substrate Specificity
11.
Mol Pharmacol ; 80(3): 400-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21642393

ABSTRACT

The human organic anion transporting polypeptide 1B3 (OATP1B3), located in the basolateral membrane of hepatocytes, mediates the uptake of endogenous substrates such as taurocholate and drugs from blood into hepatocytes. The transport activity of OATP1B3 is influenced by positively charged amino acids, which are facing the central pore. Molecular modeling was performed to select conserved positively charged amino acids, which may influence transport activity and anchoring of OATP1B3 in the plasma membrane. The modeling revealed that Lys361 faces the pore, and Lys399 is oriented to the plasma membrane. Therefore, the mutants L361>A, L361>R, L399>A, and L399>R were generated using site-directed mutagenesis to investigate the impact of the positive charges on transport activity and anchoring in the membrane. Transport kinetic analyses for the substrates sulfobromophthalein and taurocholate showed a loss of function for the L361>A mutant, whereas the transport activity was maintained by the L361>R mutant, indicating that the positive charge at position 361 is important for transport activity of OATP1B3. Comparative modeling with OATP1A2 and OATP2B1 revealed that the pore size around this lysine residue is larger in OATP1A2 and smaller in OATP2B1 compared with OATP1B3, which could be related to the respective substrate spectra. Cell surface expression of L399>A and L399>R was decreased to 16 and 72% compared with wild-type OATP1B3 (p < 0.001), respectively, indicating that the positive charge of lysine at position 399 is necessary for an unimpaired cell surface expression. Furthermore, we provide a summary of amino acids, which influence the transport activity of OATP1B3.


Subject(s)
Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/physiology , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis , Organic Anion Transporters, Sodium-Independent/genetics , Structure-Activity Relationship
12.
Pharmacogenet Genomics ; 21(3): 103-14, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21278621

ABSTRACT

OBJECTIVE: The uptake carrier organic anion-transporting polypeptide 1B3 (OATP1B3, gene SLCO1B3) is involved in the hepatic clearance of xenobiotics including statins, taxanes, and mycophenolic acid. We thought to assess the SLCO1B3 coding region for yet unidentified polymorphisms and to analyze their functional relevance. METHODS: We used DNA of ethnically diverse individuals for polymerase chain reaction, and determined polymorphisms by sequencing or temperature-dependent capillary electrophoresis. We then created variant OATP1B3 expression plasmids by site-directed mutagenesis, which were transiently expressed and functionally characterized in human cervical carcinoma (HeLa) cells using radiolabeled substrates. RESULTS: We identified six nonsynonymous polymorphisms including novel variants such as 439A>G (Thr147Ala), 767G>C (Gly256Ala), 1559A>C (His520Pro), and 1679T>C (Val560Ala). Allelic frequencies occurred to be ethnicity-dependent, with the latter observed only in African-Americans (3.6%). After expression in HeLa cells, His520Pro, Val560Ala, and Met233Ile or Met233Ile_Ser112Ala haplotype variants showed decreased uptake activity compared with wild type for cholecystokinin-8 and rosuvastatin, but not for atorvastatin. Kinetic cholecystokinin-8 analysis showed reduced Vmax without altering Km. His520Pro and Val560Ala exhibited decreased total and plasma membrane protein expressions. Val560 mapped onto a structural model of OATP1B3 showed that this is a key region for substrate-transporter interaction. His520 resides in a predicted extracellular region thought to be critical to the pH-dependent component of OATP1B3 activity. Loss of activity at pH 7.4 and 8.0 relative to pH 6.5 was significantly greater for the Pro520 variant. CONCLUSION: OATP1B3 polymorphisms that result in altered expression, substrate specificity, and pH-dependent activity may be of potential relevance to hepatic clearance of substrate drugs in vivo.


Subject(s)
Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Atorvastatin , Fluorobenzenes/metabolism , Gene Expression , HeLa Cells , Heptanoic Acids/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Liver/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Organic Anion Transporters, Sodium-Independent/chemistry , Polymorphism, Genetic , Protein Conformation , Pyrimidines/metabolism , Pyrroles/metabolism , Rosuvastatin Calcium , Sincalide/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3 , Structure-Activity Relationship , Substrate Specificity/genetics , Sulfonamides/metabolism
13.
Yao Xue Xue Bao ; 46(11): 1279-85, 2011 Nov.
Article in Chinese | MEDLINE | ID: mdl-22260016

ABSTRACT

OATP1B3, a member of SLC superfamily, is specifically expressed on the sinusoidal membrane of hepatocytes and is considered to be important in hepatic drug elimination. The overexpression of OATP1B3 was found recently in tumor tissues such as prostate, colon, and pancreatic tumors. Sequence variations in SLCO1B3 gene, such as SNPs, have been described and a common haplotype consisting of 334T>G and 699G>A SNPs is related to altered transport characteristics of OATP1B3. OATP1B3 is of relevance to drug metabolism through affecting alteration of hepatic concentration of endo- and xenobiotic compounds that interact with nuclear receptors such as PXR and CAR, and thereby directly alter the extent of target gene transcription, including major CYP isoenzymes such as CYP3A4. This review will provide an overview of substrates and inhibitors of OATP1B3 and subsequently to assess the effect of genetic mutation on transport activity. The studies linking OATP1B3 with cancer clinical outcomes are also discussed in this review.


Subject(s)
Biological Transport , Hepatocytes/metabolism , Neoplasms/metabolism , Organic Anion Transporters, Sodium-Independent , Polymorphism, Single Nucleotide , Animals , Constitutive Androstane Receptor , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Gene Expression Regulation, Neoplastic , Gene Frequency , Humans , Liver/metabolism , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Pregnane X Receptor , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3
14.
Protein Sci ; 18(11): 2298-306, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19760661

ABSTRACT

The liver-specific organic anion transporting polypeptides OATP1B1 and OATP1B3 are highly homologous and share numerous substrates. However, at low concentrations OATP1B1 shows substrate selectivity for estrone-3-sulfate. In this study, we investigated the molecular mechanism for this substrate selectivity of OATP1B1 by constructing OATP1B1/1B3 chimeric transporters and by site-directed mutagenesis. Functional studies of chimeras showed that transmembrane domain 10 is critical for the function of OATP1B1. We further identified four amino acid residues, namely L545, F546, L550, and S554 in TM10, whose simultaneous mutation caused almost complete loss of OATP1B1-mediated estrone-3-sulfate transport. Comparison of the kinetics of estrone-3-sulfate transport confirmed a biphasic pattern for OATP1B1, but showed a monophasic pattern for the quadruple mutant L545S/F546L/L550T/S554T. This mutant also showed reduced transport for other OATP1B1 substrates such as bromosulfophthalein and [D-penicillamine(2,5)]enkephalin. Helical wheel analysis and molecular modeling suggest that L545 is facing the substrate translocation pathway, whereas F546, L550, and S554 are located inside the protein. These results indicate that L545 might contribute to OATP1B1 function by interacting with substrates, whereas F546, L550, and S554 seem important for protein structure. In conclusion, our results show that TM10 is critical for the function of OATP1B1.


Subject(s)
Organic Anion Transporters , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins , Substrate Specificity/genetics , Amino Acid Sequence , Analysis of Variance , Cell Line , Estrone/analogs & derivatives , Estrone/metabolism , Humans , Kinetics , Liver-Specific Organic Anion Transporter 1 , Models, Molecular , Molecular Sequence Data , Mutation , Organic Anion Transporters/chemistry , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Solute Carrier Organic Anion Transporter Family Member 1B3
15.
J Pharmacol Exp Ther ; 329(2): 551-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19244099

ABSTRACT

Organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 are responsible for the hepatic uptake of organic anions. They share similar sequences and structures with 12 putative transmembrane domains (TMs). Their substrate specificities are very broad and overlap each other, whereas each transporter specifically recognizes certain substrates. Because the homology of the amino acid sequence in the latter part of OATP1B1 and OATP1B3 is relatively low, to determine which TMs in the latter part of OATP1B1 are important for its substrate recognition, we constructed several cell lines expressing chimeric transporters in which some TMs of OATP1B1 were substituted with those of OATP1B3, and we investigated the transport kinetics of estrone-3-sulfate (E-sul; a substrate preferentially accepted by OATP1B1) and estradiol-17beta-D-glucuronide (EG; a substrate accepted by both transporters). As the number of substituted TMs at the N terminus with those of OATP1B3 increased, the K(m) value of E-sul greatly increased and its uptake clearance decreased. The substitution of TM7 or TM9 of OATP1B1 with that of OATP1B3 (named 1B1-TM7 or 1B1-TM9) did not change the transport kinetics of EG, whereas the K(m) value of E-sul in 1B1-TM9 increased 7.4-fold. Conversely, the substitution of TM8 resulted in an 18-fold increase in the K(m) value of E-sul and abolished the transporter-mediated uptake of EG. These results suggest that TM8 in OATP1B1 is critical for the substrate recognition of both E-sul and EG and that TM9 is important for the recognition of E-sul, whereas it is interchangeable with that of OATP1B3 for EG transport.


Subject(s)
Estradiol/analogs & derivatives , Estrone/analogs & derivatives , Organic Anion Transporters/physiology , Biological Transport , Cell Line , Estradiol/metabolism , Estrone/metabolism , Humans , Kinetics , Liver-Specific Organic Anion Transporter 1 , Organic Anion Transporters/chemistry , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters, Sodium-Independent/physiology , Protein Structure, Tertiary , Solute Carrier Organic Anion Transporter Family Member 1B3 , Substrate Specificity
16.
Biochemistry ; 47(35): 9090-7, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18690707

ABSTRACT

Human organic anion transporting polypeptides (OATP) 1B1 and 1B3 are multispecific transporters that mediate uptake of amphipathic organic compounds into hepatocytes. The two OATPs contain 12 transmembrane domains (TMs) and share 80% amino acid sequence identity. Besides common substrates with OATP1B1, OATP1B3 specifically transports cholecystokinin octapeptide (CCK-8). To determine which structural domains and/or residues are important for the substrate selectivity of OATP1B3, we constructed a series of chimeric proteins between OATP1B3 and 1B1, expressed them in HEK293 cells, and determined rates of uptake of CCK-8 along with surface expression of the proteins. Replacing TM10 in OATP1B3 with TM10 of OATP1B1 resulted in a dramatically reduced degree of CCK-8 transport, indicating that TM10 is crucial for recognition and/or translocation of CCK-8. Using site-directed mutagenesis, we identified three key residues within TM10, namely, Y537, S545, and T550. When we replaced these residues with the corresponding amino acid residues found in OATP1B1, the level of CCK-8 transport was similarly low as for the replacement of the whole TM10. Kinetic experiments showed that the K m values for CCK-8 transport in the TM10 replacement and triple mutant were only 1.3 and 1.1 microM, respectively, as compared to 16.3 microM for wild-type OATP1B3. Similarly, the V max values dropped from 495.5 pmol (normalized mg) (-1) min (-1) for wild-type OATP1B3 to 13.3 and 19.0 pmol (normalized mg) (-1) min (-1) for the TM10 replacement and triple mutant, respectively. Molecular modeling indicated that two of the three identified residues might form hydrogen bonds with CCK-8. In conclusion, we have identified three amino acid residues (Y537, S545, and T550) in TM10 of OATP1B3 that are important for CCK-8 transport.


Subject(s)
Organic Anion Transporters, Sodium-Independent/chemistry , Sincalide/metabolism , Amino Acid Sequence , Cell Line , Humans , Kinetics , Liver-Specific Organic Anion Transporter 1 , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Organic Anion Transporters/chemistry , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Solute Carrier Organic Anion Transporter Family Member 1B3
17.
Am J Physiol Gastrointest Liver Physiol ; 294(4): G1052-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18308854

ABSTRACT

Organic anion transport protein 1a1 (oatp1a1), a prototypical member of the oatp family of highly homologous transport proteins, is expressed on the basolateral (sinusoidal) surface of rat hepatocytes. The organization of oatp1a1 within the plasma membrane has not been well defined, and computer-based models have predicted possible 12- as well as 10-transmembrane domain structures. Which of oatp1a1's four potential N-linked glycosylation sites are actually glycosylated and their influence on transport function have not been investigated in a mammalian system. In the present study, topology of oatp1a1 in the rat hepatocyte plasma membrane was examined by immunofluorescence analysis using an epitope-specific antibody designed to differentiate a 10- from a 12-transmembrane domain model. To map glycosylation sites, the asparagines at the each of the four N-linked glycosylation consensus sites were mutagenized to glutamines. Mutagenized oatp1a1 constructs were expressed in HeLa cells, and effects on protein expression and transport activity were assessed. These studies revealed that oatp1a1 is a 12-transmembrane-domain protein in which the second and fifth extracellular loops are glycosylated at asparagines 124, 135, and 492, whereas the potential glycosylation site at asparagine 62 is not utilized, consistent with its position in a transmembrane domain. Constructs in which more than one glycosylation site were eliminated had reduced transport activity but not necessarily reduced transporter expression. This was in accord with the finding that fully unglycosylated oatp1a1 was well expressed but located intracellularly with limited transport ability as a consequence of its reduced cell surface expression.


Subject(s)
Cell Membrane/metabolism , Hepatocytes/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Asparagine/metabolism , Fluorescent Antibody Technique , Glycosylation , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Protein Conformation , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , Rats , Sulfobromophthalein/metabolism , Sulfur Radioisotopes/metabolism
18.
Mol Pharmacol ; 73(4): 1151-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18216183

ABSTRACT

The second messenger, cGMP, mediates a host of cellular responses to various stimuli, resulting in the regulation of many critical physiologic functions. The existence of specific cGMP transporters on the plasma membrane that participate in the regulation of cGMP levels has been suggested in a large number of studies. In this study, we identified a novel plasma membrane transporter for cGMP. In particular, we showed that hOAT2 (SLC22A7), a member of the solute carrier (SLC) superfamily, was a facilitative transporter for cGMP and other guanine nucleotides. hOAT2, which is ubiquitously expressed at high levels in many cell types, was previously thought to primarily transport organic anions. Among purine and pyrimidine nucleobases, nucleosides, and nucleotides, hOAT2 showed the greatest preference for cGMP, which transported cGMP with a K(m) value of 88 +/- 11 muM and exhibited between 50- and 100-fold enhanced uptake over control cells. Our data revealed that hOAT2 is a bidirectional facilitative transporter that can control both intracellular and extracellular levels of cGMP. In addition, we observed that a common alternatively spliced variant of hOAT2 demonstrated a complete loss of transport function as a result of a low expression level on the plasma membrane. We conclude that hOAT2 is a highly efficient, facilitative transporter of cGMP and may be involved in cGMP signaling in many tissues. Our study suggests that hOAT2 represents a potential new drug target for regulating cGMP levels.


Subject(s)
Cyclic GMP/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Alternative Splicing/genetics , Amino Acids , Biological Transport , Cell Line , Deoxyguanosine/metabolism , Extracellular Space/metabolism , Gene Expression Profiling , Humans , Intracellular Space/metabolism , Nucleosides/metabolism , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity , Transfection
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...