Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
Xenobiotica ; 40(3): 184-94, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20102295

ABSTRACT

Human sulfotransferase 2A1 (SULT2A1) is a member of the hydroxysteroid sulfotransferase (SULT2) family that mediates sulfo-conjugation of a variety of endogenous molecules including dehydroepiandrosterone (DHEA) and bile acids. In this study, we have constructed a stable cell line expressing SULT2A1 by transfection into HEK293 cells. The expression system was used to characterize and compare the sulfation kinetics of DHEA and 15 human bile acids by SULT2A1. Formation of DHEA sulfate demonstrated Michaelis-Menten kinetics with apparent K(m) and V(max) values of 3.8 muM and 130.8 pmol min(-1) mg(-1) protein, respectively. Sulfation kinetics of bile acids also demonstrated Michaelis-Menten kinetics with a marked variation in apparent K(m) and V(max) values between individual bile acids. Sulfation affinity was inversely proportional to the number of hydroxyl groups of bile acids. The monohydroxy- and most toxic bile acid (lithocholic acid) had the highest affinity, whereas the trihydroxy- and least toxic bile acid (cholic acid) had the lowest affinity to sulfation by SULT2A1. Intrinsic clearance (CL(int)) of ursodeoxycholic acid (UDCA) was approximately 1.5- and 9.0-fold higher than that of deoxycholic acid (DCA) and chenodeoxycholic acid (CDCA), respectively, despite the fact that all three are dihydroxy bile acids.


Subject(s)
Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Sulfur/metabolism , Cell Line , Coenzymes , Dehydroepiandrosterone/metabolism , Gene Expression , Humans , Kinetics
2.
Clin Pharmacol Ther ; 87(1): 39-47, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19924123

ABSTRACT

Drugs and their metabolites are eliminated mainly by excretion into urine and bile. Studies in whole animals, isolated organs, cells, and membrane vesicles led to the conclusion that different transport systems are responsible for the transport of different classes of organic compounds (small, large, anionic, and cationic). In the early 1990s, functional expression cloning resulted in the identification of the first transporters for organic anions and cations. Eventually, all the major transport systems involved in the uptake of these organic compounds were cloned and characterized, and we now know that they belong to the organic anion transporters (OATs) and organic cation transporters (OCTs) of the SLC22A superfamily and the organic anion-transporting polypeptides (OATPs) of the SLCO superfamily of polyspecific drug transporters. Today we can explain, at the molecular level, why small and hydrophilic organic compounds are excreted predominantly through urine whereas large and amphipathic compounds are excreted mainly through bile, and we can start to predict drug-drug interactions in the case of new compounds.


Subject(s)
Kidney/metabolism , Liver/metabolism , Organic Anion Transporters/history , Organic Cation Transport Proteins/history , Pharmaceutical Preparations/history , Animals , Biological Transport, Active/genetics , Biological Transport, Active/physiology , Drug Interactions/genetics , Drug Interactions/physiology , History, 20th Century , History, 21st Century , Humans , Organic Anion Transporters/genetics , Organic Anion Transporters/pharmacokinetics , Organic Cation Transport Proteins/pharmacokinetics , Pharmaceutical Preparations/metabolism
3.
Xenobiotica ; 38(7-8): 778-801, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18668430

ABSTRACT

1. The organic anion transporting polypeptides (humans OATP; other species Oatp) belong to the SLCO gene superfamily of transporters and are twelve transmembrane domain glycoproteins expressed in various epithelial cells. Some OATPs/Oatps are expressed in a single organ, while others are expressed ubiquitously. 2. The functionally characterized members mediate sodium-independent transport of a variety of structurally independent, mainly amphipathic organic compounds, including bile salts, hormones and their conjugates, toxins, and various drugs. 3. This review summarizes the general features and the substrates of the eleven human OATPs. Furthermore, it reviews what is known about the mechanism of their multispecificity, their predicted structure, their role in drug-food interactions, and their role in cancer. 4. Finally, some open questions are raised that need to be addressed to advance OATP research in the near future.


Subject(s)
Epithelial Cells/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Organic Anion Transporters/metabolism , Animals , Bile Acids and Salts/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hormones/metabolism , Humans , Ion Transport/drug effects , Neoplasm Proteins/chemistry , Neoplasms/drug therapy , Organ Specificity/drug effects , Organic Anion Transporters/chemistry , Protein Structure, Tertiary , Toxins, Biological/metabolism , Xenobiotics/pharmacokinetics
4.
Toxicol Appl Pharmacol ; 203(3): 257-63, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15737679

ABSTRACT

Microcystins are toxins produced by freshwater cyanobacteria. They are cyclic heptapeptides that exhibit hepato- and neurotoxicity. However, the transport systems that mediate uptake of microcystins into hepatocytes and across the blood-brain barrier have not yet been identified. Using the Xenopus laevis oocyte expression system we tested whether members of the organic anion transporting polypeptide superfamily (rodent: Oatps; human: OATPs) are involved in transport of the most common microcystin variant microcystin-LR by measuring uptake of a radiolabeled derivative dihydromicrocystin-LR. Among the tested Oatps/OATPs, rat Oatp1b2, human OATP1B1, human OATP1B3, and human OATP1A2 transported microcystin-LR 2- to 5-fold above water-injected control oocytes. This microcystin-LR transport was inhibited by co-incubation with the known Oatp/OATP substrates taurocholate (TC) and bromosulfophthalein (BSP). Microcystin-LR transport mediated by the human OATPs was further characterized and showed saturability with increasing microcystin-LR concentrations. The apparent K(m) values amounted to 7 +/- 3 microM for OATP1B1, 9 +/- 3 microM for OATP1B3, and 20 +/- 8 microM for OATP1A2. No microcystin-LR transport was observed in oocytes expressing Oatp1a1, Oatp1a4, and OATP2B1. These results may explain some of the observed organ-specific toxicity of microcystin-LR. Oatp1b2, OATP1B1, and OATP1B3 are responsible for microcystin transport into hepatocytes, whereas OATP1A2 mediates microcystin-LR transport across the blood-brain barrier.


Subject(s)
Brain/metabolism , Liver/metabolism , Organic Anion Transporters/biosynthesis , Organic Anion Transporters/physiology , Peptides, Cyclic/metabolism , Animals , Brain/microbiology , Female , Humans , Liver/microbiology , Microcystins , Rats , Xenopus laevis
5.
Curr Drug Metab ; 4(3): 185-211, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12769665

ABSTRACT

The hepatobiliary system and the kidneys are the main routes by which drugs and their metabolites leave the body. Compounds that are mainly excreted into bile in general have relatively high molecular weights, are amphipathic and highly bound to plasma proteins. In contrast, compounds that are predominantly excreted into urine have relatively low molecular weights, are more hydrophilic and generally less protein bound. The first step in drug elimination in liver and kidney is uptake into hepatocytes or into proximal tubular cells. The substrate specificity and affinity of the uptake carriers expressed at the basolateral membranes of hepatocytes and proximal tubular cells could therefore play an important role for the determination of the main elimination route of a compound. This review discusses the tissue distribution, substrate specificity, transport mechanism, and regulation of the members of the organic anion transporting polypeptide (Oatp/OATP) superfamily (solute carrier family SLC21A) and the SLC22A family containing transporters for organic cations (OCTs) and organic anions (OATs). The Oatps/OATPs are mainly important for the hepatic uptake of large amphipathic organic anions, organic cations and uncharged substrates, whereas OCTs and OATs mediate uptake of predominantly small organic cations and anions in liver and kidney.


Subject(s)
Kidney/metabolism , Liver/metabolism , Pharmaceutical Preparations/metabolism , Animals , Humans , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism
6.
Biochim Biophys Acta ; 1609(1): 1-18, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12507753

ABSTRACT

Organic anion transporting polypeptides (Oatps/OATPs) form a growing gene superfamily and mediate transport of a wide spectrum of amphipathic organic solutes. Different Oatps/OATPs have partially overlapping and partially distinct substrate preferences for organic solutes such as bile salts, steroid conjugates, thyroid hormones, anionic oligopeptides, drugs, toxins and other xenobiotics. While some Oatps/OATPs are preferentially or even selectively expressed in one tissue such as the liver, others are expressed in multiple organs including the blood-brain barrier (BBB), choroid plexus, lung, heart, intestine, kidney, placenta and testis. This review summarizes the actual state of the rapidly expanding OATP superfamily and covers the structural properties, the genomic classification, the phylogenetic relationships and the functional transport characteristics. In addition, we propose a new species independent and open ended nomenclature and classification system, which is based on divergent evolution and agrees with the guidelines of the Human Genome Nomenclature Committee.


Subject(s)
Organic Anion Transporters/chemistry , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Phylogeny , Protein Conformation , Sequence Homology, Amino Acid , Terminology as Topic
7.
Mol Endocrinol ; 16(10): 2283-96, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12351693

ABSTRACT

Transport of various amphipathic organic compounds is mediated by organic anion transporting polypeptides (OATPs in humans, Oatps in rodents), which belong to the solute carrier family 21A (SLC21A/Slc21a). Several of these transporters exhibit a broad and overlapping substrate specificity and are expressed in a variety of different tissues. We have isolated and functionally characterized OATP-F (SLC21A14), a novel member of the OATP family. The cDNA (3059 bp) contains an open reading frame of 2136 bp encoding a protein of 712 amino acids. Its gene containing 15 exons is located on chromosome 12p12. OATP-F exhibits 47-48% amino acid identity with OATP-A, OATP-C, and OATP8, the genes of which are clustered on chromosome 12p12. OATP-F is predominantly expressed in multiple brain regions and Leydig cells of the testis. OATP-F mediates high affinity transport of T(4) and reverse T(3) with apparent K(m) values of approximately 90 nM and 128 nM, respectively. Substrates less well transported by OATP-F include T(3), bromosulfophthalein, estrone-3-sulfate, and estradiol-17beta-glucuronide. Furthermore, OATP-F-mediated T(4) uptake could be cis-inhibited by L-T(4) and D-T(4), but not by 3,5-diiodothyronine, indicating that T(4) transport is not stereospecific, but that 3',5'-iodination is important for efficient transport by OATP-F. Thus, in contrast to most other family members, OATP-F has a more selective substrate preference and may play an important role in the disposition of thyroid hormones in brain and testis.


Subject(s)
Brain/metabolism , Estradiol/analogs & derivatives , Estrone/analogs & derivatives , Organic Anion Transporters/metabolism , Testis/metabolism , Thyroxine/metabolism , Amino Acid Sequence , Animals , CHO Cells/metabolism , Chromosomes, Human, Pair 12 , Cloning, Molecular , Cricetinae , Diiodothyronines/pharmacology , Estradiol/metabolism , Estrone/metabolism , Female , Humans , Leydig Cells/metabolism , Male , Membrane Proteins , Molecular Sequence Data , Oocytes/metabolism , Organ Specificity , Organic Anion Transporters/genetics , Sequence Homology, Amino Acid , Sulfobromophthalein/metabolism , Triiodothyronine/metabolism , Xenopus
8.
J Clin Endocrinol Metab ; 87(4): 1856-63, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11932330

ABSTRACT

Organic anion-transporting polypeptides (OATPs) are a family of multispecific carriers that mediate the sodium-independent transport of steroid hormone and conjugates, drugs, and numerous anionic endogenous substrates. We investigated whether members of the OATP gene family could mediate fetal-maternal transfer of anionic steroid conjugates in the human placenta. OATP-B (gene symbol SLC21A9) was isolated from a placenta cDNA library. An antiserum to OATP-B detected an 85-kDa protein in basal but not apical syncytiotrophoblast membranes. Immunohistochemistry of first-, second-, and third-trimester placenta showed staining in the cytotrophoblast membranes and at the basal surface of the syncytiotrophoblast. Trophoblasts that reacted with an antibody to Ki-67, a proliferation-associated antigen, expressed lower levels of OATP-B. OATP-B mRNA levels were measured in isolated trophoblasts under culture conditions that promoted syncytia formation. Real-time quantitative PCR estimated an 8-fold increase in OATP-B expression on differentiation to syncytia. The uptake of [(3)H]estrone-3-sulfate, a substrate for OATP-B, was measured in basal syncytiotrophoblast membrane vesicles. Transport was saturable and partially inhibited by pregnenolone sulfate, a progesterone precursor. Pregnenolone sulfate also partially inhibited OATP-B-mediated transport of estrone-3-sulfate in an oocyte expression system. These findings suggest a physiological role for OATP-B in the placental uptake of fetal-derived sulfated steroids.


Subject(s)
Estrone/analogs & derivatives , Organic Anion Transporters/metabolism , Placenta/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Cells, Cultured , Estrone/antagonists & inhibitors , Estrone/pharmacokinetics , Female , Giant Cells/physiology , Humans , Oocytes/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/physiology , Pregnancy , Pregnenolone/pharmacology , RNA, Messenger/metabolism , Trophoblasts/metabolism
9.
Pflugers Arch ; 443(2): 188-95, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11713643

ABSTRACT

Organic anion transporting polypeptides (rodents: Oatps; human: OATPs) are involved in the absorption and elimination of a wide variety of structurally unrelated amphipathic organic compounds. Several members of this protein family mediate the uptake of substrates across the basolateral membrane of hepatocytes as the first step in hepatic elimination. In contrast to the well-characterized Oatp1 and Oatp2, the localization and substrate specificity of the recently cloned Oatp4 have not been investigated in detail. Therefore, we raised an antibody against the C-terminal end of Oatp4 and localized this 85-kDa protein to the basolateral membrane of rat hepatocytes. Similar to Oatp1 and Oatp2, Oatp4 is a multispecific transporter with high affinities for bromosulfophthalein, dehydroepiandrosterone sulfate, leukotriene C4, and anionic peptides. In addition, we compared the substrate specificity of Oatp4 to that of Oatp3, which so far has mainly been shown to mediate intestinal bile acid transport. Oatp3 had a similar broad substrate specificity, but in general much lower affinities than Oatp4. Thus, while Oatp4 seems to work in concert with Oatp1 and Oatp2 in the basolateral membrane of rat hepatocytes, Oatp3 is a multispecific transport system in the small intestine.


Subject(s)
Organic Anion Transporters, Sodium-Independent/metabolism , Animals , Biological Transport , Female , Liver/cytology , Liver/metabolism , Oocytes/metabolism , Rats , Solute Carrier Organic Anion Transporter Family Member 1B3 , Subcellular Fractions/metabolism , Substrate Specificity , Tissue Distribution , Xenopus laevis
10.
Gastroenterology ; 121(5): 1185-90, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11677211

ABSTRACT

BACKGROUND & AIMS: Cholecystokinin (CCK) is a major gastrointestinal peptide hormone that is released postprandially from the small intestine and exerts marked effects on gallbladder and gastrointestinal motility. The smaller isoforms CCK-8 and CCK-4 are rapidly taken up into hepatocytes, metabolized, and excreted into bile. Our aim was to identify and characterize the hepatocellular CCK-8 uptake system. METHODS: CCK-8 uptake was measured in Xenopus laevis oocytes expressing the organic anion-transporting polypeptides of rat liver (Oatp1, Oatp2, Oatp3, or Oatp4) and of human liver (OATP-A, OATP-B, OATP-C, or OATP8) and in primary cultured rat hepatocytes. RESULTS: Rat Oatp4 and human OATP8 efficiently mediated CCK-8 uptake in oocytes, with Michaelis constant (Km) values of 14.9 +/- 2.9 micromol/L and 11.1 +/- 2.9 micromol/L, respectively. CCK-8 uptake by hepatocytes was also saturable, with a Km of 6.7 +/- 2.1 micromol/L. The Km value in rat hepatocytes is consistent with Oatp4-mediated transport. CONCLUSIONS: CCK-8 is selectively transported by rat Oatp4 and human OATP8, both of which are exclusively expressed at the basolateral membrane of hepatocytes. These 2 transporters are the first and probably the predominant hepatic uptake systems for CCK-8 and may be critical for the rapid clearance of this hormone from the circulation.


Subject(s)
Liver/metabolism , Organic Anion Transporters, Sodium-Independent/physiology , Organic Anion Transporters/physiology , Sincalide/pharmacokinetics , Animals , Cells, Cultured , Hepatocytes/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley , Solute Carrier Organic Anion Transporter Family Member 1B3 , Xenopus laevis
11.
J Biol Chem ; 276(40): 37206-14, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11483603

ABSTRACT

OATP-C (SLC21A6) is the predominant Na(+)-independent uptake system for bile salts and bilirubin of human liver and is expressed exclusively at the basolateral (sinusoidal) hepatocyte membrane. To investigate the basis of liver-specific expression of OATP-C, we studied promoter function in the two hepatocyte-derived cell lines HepG2 and Huh7 and in nonhepatic HeLa cells. OATP-C promoter constructs containing from 66 to 950 nucleotides of 5'-regulatory sequence were active in HepG2 and Huh7 but not HeLa cells, indicating that determinants of hepatocyte-specific expression reside within the minimal promoter. Deoxyribonuclease I footprint analysis revealed a single region that was protected by HepG2 and Huh7 but not HeLa cell nuclear extracts. The liver-enriched transcription factor hepatocyte nuclear factor 1 alpha (HNF1 alpha) was shown by mobility shift assays to bind within this footprint. Coexpression of HNF1 alpha stimulated OATP-C promoter activity 30-fold in HepG2 and 49-fold in HeLa cells. Mutation of the HNF1 site abolished promoter function, indicating that HNF1 alpha is critical for hepatocyte-specific OATP-C gene expression. The human OATP8 (SLC21A8) and mouse Oatp4 (Slc21a6) promoters were also responsive to HNF1 alpha coexpression in HepG2 cells. These data support a role for HNF1 alpha as a global regulator of liver-specific bile salt and organic anion transporter genes.


Subject(s)
Anion Transport Proteins/genetics , DNA-Binding Proteins , Gene Expression Regulation , Liver/physiology , Nuclear Proteins , Promoter Regions, Genetic/genetics , Transcription Factors/physiology , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , DNA/analysis , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Hepatocyte Nuclear Factor 1-beta , Humans , Mice , Molecular Sequence Data , Mutagenesis , Organ Specificity , Organic Anion Transporters, Sodium-Independent/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3 , Transcription, Genetic , Transcriptional Activation
12.
Hepatology ; 33(5): 1223-31, 2001 May.
Article in English | MEDLINE | ID: mdl-11343252

ABSTRACT

The bile salt export pump (Bsep), a member of the ATP-binding cassette superfamily of transporters, mediates the ATP-dependent canalicular secretion of bile salts. We have cloned and expressed the mouse Bsep (mBsep) protein in Sf9 insect cells, and characterized its transport and ATPase properties. Because its deduced amino acid sequence predicts multiple phosphorylation sites for protein kinase A, protein kinase C (PKC) and Ca(2+)-calmodulin dependent kinase II, we have also tested whether mBsep undergoes phosphorylation. MBsep transports both glycine and taurine conjugated bile salts. Sf9 cell membranes that express mBsep exhibit higher basal ATPase activity than control membranes, and this is further stimulated by bile salts and inhibited by vanadate. Taurochenodeoxycholate is transported with the highest affinity and is the most potent inducer of ATPase activity. Cyclosporin A, glibenclamide and rifamycin SV, all competitive inhibitors of Bsep transport, also reduced the bile salt-stimulated ATPase activity. MBsep exists as a phospho-protein when expressed in Sf9 cells and the immunoprecipitated mBsep complex is a substrate for the catalytic subunit of PKC. When mBsep and the alpha-isoform of mouse PKC are co-expressed in Sf9 cells, a ninefold stimulation of phosphorylation occurs. This is further increased to 18-fold after activation by phorbol ester. Given that bile salts activate selected PKC isoforms in hepatocytes, including the alpha isoform, the phosphorylation of mBsep by PKCalpha may represent a point of regulation for this transporter that is mediated by its own substrate.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Baculoviridae/genetics , Gene Expression , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/metabolism , Amino Acid Sequence/genetics , Animals , Biological Transport/physiology , Blotting, Southern , Cell Line , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Insecta/cytology , Mice , Molecular Sequence Data , Phosphorylation , Precipitin Tests
13.
J Exp Biol ; 204(Pt 10): 1673-86, 2001 May.
Article in English | MEDLINE | ID: mdl-11316487

ABSTRACT

Bile acids are steroidal amphipathic molecules derived from the catabolism of cholesterol. They modulate bile flow and lipid secretion, are essential for the absorption of dietary fats and vitamins, and have been implicated in the regulation of all the key enzymes involved in cholesterol homeostasis. Bile acids recirculate through the liver, bile ducts, small intestine and portal vein to form an enterohepatic circuit. They exist as anions at physiological pH and, consequently, require a carrier for transport across the membranes of the enterohepatic tissues. Individual bile acid carriers have now been cloned from several species. Na+-dependent transporters that mediate uptake into hepatocytes and reabsorption from the intestine and biliary epithelium and an ATP-dependent transporter that pumps bile acids into bile comprise the classes of transporter that are specific for bile acids. In addition, at least four human and five rat genes that code for Na+-independent organic anion carriers with broad multi-substrate specificities that include bile acids have been discovered. Studies concerning the regulation of these carriers have permitted identification of molecular signals that dictate eventual changes in the uptake or excretion of bile acids, which in turn have profound physiological implications. This overview summarizes and compares all known bile acid transporters and highlights findings that have identified diseases linked to molecular defects in these carriers. Recent advances that have fostered a more complete appreciation for the elaborate disposition of bile acids in humans are emphasized.


Subject(s)
Bile Acids and Salts/metabolism , Carrier Proteins/metabolism , Hydroxysteroid Dehydrogenases , Liver/metabolism , Membrane Glycoproteins , Animals , Biological Transport, Active , Cell Polarity , Female , Hepatocytes/metabolism , Humans , Intestinal Absorption , Kidney Tubules , Liver/cytology , Molecular Structure , Placenta/metabolism , Pregnancy
14.
Hepatology ; 33(4): 776-82, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283839

ABSTRACT

To maintain ongoing vectorial bile secretion, hepatocytes localize distinct transport systems at their basolateral and canalicular membrane domains. Here we compare the expression of the basolateral Na(+)-taurocholate cotransporter (Ntcp) and organic anion transporting polypeptides 1 and 2 (Oatp1, Oatp2) and the canalicular bile salt export pump (Bsep) and multidrug resistance-associated protein 2 (Mrp2) in primary cultured rat hepatocytes. During 72 hours of culturing time the messenger RNA (mRNA) and protein levels of Ntcp and Oatp1 decreased in parallel to 2% to 7% of initial values at 3 hours. Although Oatp2 mRNA exhibited a similar down-regulation to 4%, Oatp2 protein and function were maintained at 25% to 47% of initial values. Furthermore, Bsep and Mrp2 protein levels were maintained at about 50%, while the Mrp2 mRNA showed a transient up-regulation to 154% at 24 and 48 hours. Also, induction of Mrp1 mRNA and protein was observed starting after 24 hours. These results indicate transcriptional down-regulation or decreased mRNA stability of Ntcp and Oatp1, transcriptional and posttranslational regulation of Oatp2, Bsep, and Mrp2 and transcriptional up-regulation of Mrp1 in primary cultured hepatocytes. Furthermore, and most importantly, the observed changes in transporter expression closely resemble the altered transporter phenotypes of cholestatic and proliferating hepatocytes in vivo, thus indicating that primary cultured hepatocytes acquire a cholestatic phenotype, and that the transporter expression might be a suitable differentiation marker for maintenance of hepatocytes in vitro.


Subject(s)
Bile Canaliculi/metabolism , Carrier Proteins/metabolism , Cholestasis/metabolism , Hepatocytes/metabolism , Liver/metabolism , Membrane Transport Proteins , Mitochondrial Proteins , Saccharomyces cerevisiae Proteins , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/metabolism , Animals , Anion Transport Proteins , Cells, Cultured , Liver/cytology , Male , Organic Anion Transporters, Sodium-Dependent , Rats , Rats, Sprague-Dawley , Ribosomal Proteins/metabolism , Symporters
15.
Biochim Biophys Acta ; 1518(1-2): 73-8, 2001 Mar 19.
Article in English | MEDLINE | ID: mdl-11267661

ABSTRACT

Mouse Oatp1 was recently identified as a new murine member of the organic anion transporting polypeptide (Oatp) family and suggested to represent the counterpart of rat Oatp1. Northern blot analysis detected expression of several mouse Oatp-transcripts predominantly in liver and kidney. In the present study we describe the strict androgen-dependent expression of mouse Oatp1 mRNA in kidney and obtained further information about its substrate specificity using Xenopus oocytes. In addition to the previously reported estrone-3-sulfate, we demonstrate that mouse Oatp1 mediates sodium-independent uptake of the anionic steroid conjugates dehydroepiandrosterone sulfate (K(m) approximately 8 microM) and estradiol-17-glucuronide (K(m) approximately 5 microM) and also of the prostaglandin PGE(2).


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Testosterone/analogs & derivatives , Testosterone/metabolism , 3' Untranslated Regions , Animals , Anion Transport Proteins , Base Sequence , Carrier Proteins/metabolism , DNA, Complementary , Dehydroepiandrosterone Sulfate/pharmacokinetics , Dinoprostone/pharmacokinetics , Estradiol/analogs & derivatives , Estradiol/pharmacokinetics , Female , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Testosterone/pharmacology , Xenopus laevis
16.
Gastroenterology ; 120(2): 525-33, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11159893

ABSTRACT

BACKGROUND & AIMS: Hepatic uptake of cholephilic organic compounds is mediated by members of the organic anion-transporting polypeptide (OATP) family. We aimed to characterize the novel OATP-B with respect to tissue distribution and hepatocellular localization and to compare its substrate specificity with those of OATP-A, OATP-C, and OATP8. METHODS: Tissue distribution and hepatocellular localization of OATP-B were analyzed by Northern blotting and immunofluorescence, respectively. Transport of 16 substrates was measured for each individual human OATP in complementary RNA-injected Xenopus laevis oocytes. RESULTS: Expression of OATP-B was most abundant in human liver, where it is localized at the basolateral membrane of hepatocytes. OATP-B, OATP-C, and OATP8 mediated high-affinity uptake of bromosulphophthalein (K(m), approximately 0.7, 0.3, and 0.4 micromol/L, respectively). OATP-B also transported estrone-3-sulfate but not bile salts. Although OATP-A, OATP-C, and OATP8 exhibit broad overlapping substrate specificities, OATP8 was unique in transporting digoxin and exhibited especially high transport activities for the anionic cyclic peptides [D-penicillamine(2,5)]enkephalin (DPDPE; opioid-receptor agonist) and BQ-123 (endothelin-receptor antagonist). CONCLUSIONS: OATP-B is the third bromosulphophthalein uptake system localized at the basolateral membrane of human hepatocytes. OATP-B, OATP-C, and OATP8 account for the major part of sodium-independent bile salt, organic anion, and drug clearance of human liver.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Estrone/analogs & derivatives , Liver/metabolism , Animals , Anion Transport Proteins , Anions/pharmacokinetics , Antibodies , Arylsulfatases/pharmacokinetics , Biological Transport/physiology , Blotting, Northern , Carrier Proteins/immunology , Coloring Agents/pharmacokinetics , DNA, Complementary , Estrone/pharmacokinetics , Gene Expression/physiology , Humans , Liver/chemistry , Molecular Weight , Oocytes/physiology , RNA, Messenger/analysis , Rabbits , Steryl-Sulfatase , Sulfobromophthalein/pharmacokinetics , Xenopus laevis
17.
J Pharmacol Exp Ther ; 296(1): 91-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11123367

ABSTRACT

Chlorogenic acid derivatives were recently identified as novel, potent, and specific inhibitors of the hepatic glucose 6-phosphate translocase. Inhibition of the glucose 6-phosphate translocase leads to a decrease in hepatic glucose production, rendering chlorogenic acid derivatives as potential novel therapeutics in patients with type 2 diabetes. The present study examines the hepatic uptake mechanism of the radiolabeled chlorogenic acid derivative S 1743 into freshly isolated rat hepatocytes. Initial uptake rates were Na(+)-independent and followed saturation kinetics with no superimposition of facilitated diffusion. Inhibition studies demonstrated that other chlorogenic acid derivatives inhibited uptake of the radiolabeled compound S 1743 into rat hepatocytes in the range of 1.1 to 11 microM, whereas the natural chlorogenic acid (up to 100 microM) had no effect at all. In addition, inhibition of S 1743 uptake into rat hepatocytes was found in the presence of sulfobromophthalein, sulfolithocholyltaurine, estrone-3-sulfate, cholyltaurine, verapamil, bumetanide, probenecide, phenol red, digoxin, and ouabain (in decreasing order) but not with N-methylnicotinamide, alpha-ketoglutarate, p-aminohippurate, geneticin sulfate, and 5-sulfosalicylate. The observed inhibition pattern suggested that members of the family of the organic anion transporting polypeptides (Oatps) could be involved in hepatic uptake of chlorogenic acid derivatives. Indeed, S 1743 uptake could be demonstrated in Oatp1- and Oatp2-expressing Xenopus laevis oocytes as well as in Oatp1-expressing Chinese hamster ovary cells. A comparison of the inhibition pattern obtained in hepatocytes compared with that obtained in Oatp1-expressing Chinese hamster ovary cells suggests that facilitated uptake by Oatp1 is a major contributor in total hepatic uptake of chlorogenic acid derivatives.


Subject(s)
Carrier Proteins/metabolism , Chlorogenic Acid/analogs & derivatives , Hepatocytes/metabolism , Liver/metabolism , Sulfonamides/metabolism , Animals , Anion Transport Proteins , CHO Cells , Chlorogenic Acid/metabolism , Cricetinae , Hepatocytes/drug effects , In Vitro Techniques , Kinetics , Male , Microsomes, Liver/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Oocytes/metabolism , Rats , Rats, Sprague-Dawley , Xenopus laevis
18.
Semin Liver Dis ; 20(3): 273-92, 2000.
Article in English | MEDLINE | ID: mdl-11076396

ABSTRACT

The vectorial secretion of bile salts from blood into bile is a major driving force for bile formation. The basolateral hepatocyte membrane extracts bile salts from sinusoidal blood via Na(+)-dependent and Na(+)-independent membrane transporters. Na(+)-dependent uptake of bile salts is mediated by the Na(+)-taurocholate co-transporting polypeptide, a 51-kDa protein that is exclusively expressed in hepatocytes. Na(+)-independent uptake of bile salts is mediated by the organic anion transporting polypeptides, a superfamily of multispecific bile salt and amphipathic substrate transporters. Within the hepatocyte, bile salts are bound to cytosolic proteins and traverse the cell mainly by diffusion. Transport across the canalicular membrane is the rate-limiting step in overall hepatocellular bile salt excretion and is mediated by the bile salt export pump (BSEP), a homologue of the P-glycoproteins or multidrug resistance gene products. BSEP is a vulnerable target for inhibition by estrogen metabolites, drugs such as cyclosporine A, and abnormal bile salt metabolites, all of which can cause retention of bile salts and consequently intrahepatic cholestasis. Canalicular efflux of divalent sulfated or glucuronidated bile salts is mediated by the multidrug resistance protein 2 (MRP2), which is strongly decreased in cholestasis. Decreased MRP2 expression leads to compensatory increases in the basolateral expression of MRP1 and MRP3, which mediate the sinusoidal efflux of divalent bile salt conjugates and other organic anions. Thus, the hepatocyte can regulate expression levels of individual bile salt transporters during cholestasis to evade hepatotoxic injury.


Subject(s)
Bile Acids and Salts/metabolism , Liver/metabolism , Membrane Transport Proteins , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/physiology , Animals , Anion Transport Proteins , Biological Transport, Active , Carrier Proteins/physiology , Humans , Organic Anion Transporters, Sodium-Dependent , Rats , Symporters
19.
FEBS Lett ; 474(2-3): 242-5, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10838093

ABSTRACT

A novel organic anion transporting polypeptide (Oatp)4(1) was isolated from rat liver that is 35 amino acids longer than the reported rat liver specific organic anion transporter (rlst)-1 and exhibits a 64% amino acid sequence identity with the human OATP-C (LST-1/OATP2; gene symbol SLC21A6). When expressed in Xenopus laevis oocytes, Oatp4 (Slc21a10) mediated polyspecific uptake of a variety of organic anions including taurocholate (K(m) approximately 27 microM), bromosulfophthalein (K(m) approximately 1.1 microM) and steroid conjugates. Based on nuclease protection analysis Oatp4 appears to be the predominant transcript in rat liver indicating that rlst-1 plays a minor role in overall hepatic organic anion uptake.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Liver/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Anion Transport Proteins , Anions/metabolism , Biological Transport , Carrier Proteins/chemistry , Cloning, Molecular , Kinetics , Molecular Sequence Data , Oocytes/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Homology, Amino Acid , Substrate Specificity , Xenopus laevis
20.
J Pharmacol Exp Ther ; 294(1): 73-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10871297

ABSTRACT

Organic anion-transporting polypeptides (Oatps) are a rapidly growing gene family of polyspecific membrane transporters. In rat brain, Oatp1 (gene symbol Slc21a1) and Oatp2 (Slc21a5) are localized at the apical and basolateral domains, respectively, of the choroid plexus epithelium. Furthermore, Oatp2 is strongly expressed at the rat blood-brain barrier (BBB). This study localizes the human OATP (now called OATP-A; SLC21A3) at the BBB in humans. Furthermore, with the Xenopus laevis oocyte system the delta-opioid receptor agonists [D-penicillamine(2,5)]enkephalin (DPDPE) and deltorphin II were identified as new transport substrates of OATP-A. This OATP-A-mediated DPDPE and deltorphin II transport exhibited apparent K(m) values of approximately 202 and 330 microM, respectively, and OATP-A-mediated deltorphin II transport was inhibited by the mu-opioid receptor agonist Tyr-D-Ala-Gly-N-methyl-Phe-glycinol, the endogenous peptide Leu-enkephalin, and the opiate antagonists naloxone and naltrindole. DPDPE also was transported by rat Oatp1 (K(m) approximately 48 microM) and Oatp2 (K(m) approximately 19 microM), whereas deltorphin II was only transported by Oatp1 (K(m) approximately 137 microM). These results demonstrate that OATP-A can mediate transport of the analgesic opioid peptides DPDPE and deltorphin II across the human BBB. Furthermore, because rat Oatp1 and Oatp2 exhibit similar but not identical transport activities as OATP-A, the results generally indicate that members of the Oatp/OATP gene family of membrane transporters play an important role in carrier-mediated transport of opioid peptides across the BBB and blood-cerebrospinal fluid barrier of the mammalian brain.


Subject(s)
Blood-Brain Barrier , Carrier Proteins/physiology , Opioid Peptides/pharmacokinetics , Aged , Animals , Anion Transport Proteins , Biological Transport , Carrier Proteins/analysis , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Humans , Male , Molecular Weight , Oligopeptides/pharmacokinetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...