Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Mol Biosci ; 8: 689757, 2021.
Article in English | MEDLINE | ID: mdl-34079822

ABSTRACT

Three carriers of the solute carrier family SLC10 have been functionally characterized so far. Na+/taurocholate cotransporting polypeptide NTCP is a hepatic bile acid transporter and the cellular entry receptor for the hepatitis B and D viruses. Its intestinal counterpart, apical sodium-dependent bile acid transporter ASBT, is responsible for the reabsorption of bile acids from the intestinal lumen. In addition, sodium-dependent organic anion transporter SOAT specifically transports sulfated steroid hormones, but not bile acids. All three carriers show high sequence homology, but significant differences in substrate recognition that makes a systematic structure-activity comparison attractive in order to define the protein domains involved in substrate binding and transport. By using stably transfected NTCP-, ASBT-, and SOAT-HEK293 cells, systematic comparative transport and inhibition experiments were performed with more than 20 bile acid and steroid substrates as well as different inhibitors. Taurolithocholic acid (TLC) was identified as the first common substrate of NTCP, ASBT and SOAT with K m values of 18.4, 5.9, and 19.3 µM, respectively. In contrast, lithocholic acid was the only bile acid that was not transported by any of these carriers. Troglitazone, BSP and erythrosine B were identified as pan-SLC10 inhibitors, whereas cyclosporine A, irbesartan, ginkgolic acid 17:1, and betulinic acid only inhibited NTCP and SOAT, but not ASBT. The HBV/HDV-derived myr-preS1 peptide showed equipotent inhibition of the NTCP-mediated substrate transport of taurocholic acid (TC), dehydroepiandrosterone sulfate (DHEAS), and TLC with IC50 values of 182 nM, 167 nM, and 316 nM, respectively. In contrast, TLC was more potent to inhibit myr-preS1 peptide binding to NTCP with IC50 of 4.3 µM compared to TC (IC50 = 70.4 µM) and DHEAS (IC50 = 52.0 µM). Based on the data of the present study, we propose several overlapping, but differently active binding sites for substrates and inhibitors in the carriers NTCP, ASBT, SOAT.

2.
PLoS One ; 13(6): e0199200, 2018.
Article in English | MEDLINE | ID: mdl-29912972

ABSTRACT

The hepatic Na+/taurocholate co-transporting polypeptide (NTCP in man, Ntcp in animals) is the high-affinity receptor for the hepatitis B (HBV) and hepatitis D (HDV) viruses. Species barriers for human HBV/HDV within the order Primates were previously attributed to Ntcp sequence variations that disable virus-receptor interaction. However, only a limited number of primate Ntcps have been analysed so far. In the present study, a total of 11 Ntcps from apes, Old and New World monkeys were cloned and expressed in vitro to characterise their interaction with HBV and HDV. All Ntcps showed intact bile salt transport. Human NTCP as well as the Ntcps from the great apes chimpanzee and orangutan showed transport-competing binding of HBV derived myr-preS1-peptides. In contrast, all six Ntcps from the group of Old World monkeys were insensitive to HBV myr-preS1-peptide binding and HBV/HDV infection. This is basically predetermined by the amino acid arginine at position 158 of all studied Old World monkey Ntcps. An exchange from arginine to glycine (as present in humans and great apes) at this position (R158G) alone was sufficient to achieve full transport-competing HBV myr-preS1-peptide binding and susceptibility for HBV/HDV infection. New World monkey Ntcps showed higher sequence heterogeneity, but in two cases with 158G showed transport-competing HBV myr-preS1-peptide binding, and in one case (Saimiri sciureus) even susceptibility for HBV/HDV infection. In conclusion, amino acid position 158 of NTCP/Ntcp is sufficient to discriminate between the HBV/HDV susceptible group of humans and great apes (158G) and the non-susceptible group of Old World monkeys (158R). In the case of the phylogenetically more distant New World monkey Ntcps amino acid 158 plays a significant, but not exclusive role.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B/veterinary , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Animals , Callithrix/genetics , Chlorocebus aethiops/genetics , Cloning, Molecular , HEK293 Cells , Hep G2 Cells , Hepatitis B/transmission , Humans , Macaca/genetics , Macaca fascicularis/genetics , Macaca mulatta/genetics , Organic Anion Transporters, Sodium-Dependent/genetics , Pan troglodytes/genetics , Papio anubis/genetics , Papio hamadryas/genetics , Pongo abelii/genetics , Saguinus/genetics , Saimiri/genetics , Sequence Alignment , Symporters/genetics , Transfection
3.
J Steroid Biochem Mol Biol ; 179: 20-25, 2018 05.
Article in English | MEDLINE | ID: mdl-28951227

ABSTRACT

The sodium-dependent organic anion transporter SOAT/Soat shows highly specific transport activity for sulfated steroids. SOAT substrates identified so far include dehydroepiandrosterone sulfate, 16α-hydroxydehydroepiandrosterone sulfate, estrone-3-sulfate, pregnenolone sulfate, 17ß-estradiol-3-sulfate, and androstenediol sulfate. Apart from these compounds, many other sulfated steroids occur in mammals. Therefore, we aimed to expand the substrate spectrum of SOAT and analyzed the SOAT-mediated transport of eight different sulfated steroids by combining in vitro transport experiments in SOAT-transfected HEK293 cells with LC-MS/MS analytics of cell lysates. In addition, we aimed to better understand the structural requirements for SOAT substrates and so selected structural pairs varying only at specific positions: 3α/3ß-sulfate, 17α/17ß-sulfate, mono-sulfate/di-sulfate, and 17α-hydroxylation. We found significant and sodium-dependent SOAT-mediated transport of 17α-hydroxypregnenolone sulfate, 17ß-estradiol-17-sulfate, androsterone sulfate, epiandrosterone sulfate, testosterone sulfate, epitestosterone sulfate, and 5α-dihydrotestosterone sulfate. However, 17ß-estradiol-3,17-disulfate was not transported by SOAT. IN CONCLUSION: SOAT substrates from the group of sulfated steroids are characterized by a planar and lipophilic steroid backbone in trans-trans-trans conformation of the rings and a negatively charged mono-sulfate group at positions 3' or 17' with flexibility for α- or ß- orientation. Furthermore, 5α-reduction, 16α-hydroxylation, and 17α-hydroxylation are acceptable for SOAT substrate recognition, whereas addition of a second negatively charged sulfate group seems to abolish substrate binding to SOAT, and so 17ß-estradiol-3,17-disulfate is not transported by SOAT.


Subject(s)
Organic Anion Transporters/metabolism , Steroids/chemistry , Steroids/metabolism , Androsterone/analogs & derivatives , Androsterone/chemistry , Androsterone/metabolism , Biological Transport , Dihydrotestosterone/chemistry , Dihydrotestosterone/metabolism , Estradiol/analogs & derivatives , Estradiol/chemistry , Estradiol/metabolism , HEK293 Cells , Humans , Hydroxylation , Organic Anion Transporters/chemistry , Structure-Activity Relationship , Testosterone/chemistry , Testosterone/metabolism
4.
Mol Cell Endocrinol ; 428: 133-41, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27033324

ABSTRACT

The sodium-dependent organic anion transporter SOAT specifically transports sulfated steroid hormones and is supposed to play a role in testicular steroid regulation and male fertility. The present study aimed to identify novel specific SOAT inhibitors for further in vitro and in vivo studies on SOAT function. More than 100 compounds of different molecular structures were screened for inhibition of the SOAT-mediated transport of dehydroepiandrosterone sulfate in stably transfected SOAT-HEK293 cells. Twenty-five of these with IC50 values covering four orders of magnitude were selected as training set for 3D pharmacophore modelling. The SOAT pharmacophore features were calculated by CATALYST and consist of three hydrophobic sites and two hydrogen bond acceptors. By substrate database screening, compound T 0511-1698 was predicted as a novel SOAT inhibitor with an IC50 of 15 µM. This value was confirmed by cell-based transport assays. Therefore, the developed SOAT pharmacophore model demonstrated its suitability in predicting novel SOAT inhibitors.


Subject(s)
Dehydroepiandrosterone Sulfate/metabolism , Drug Evaluation, Preclinical , Models, Molecular , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/chemistry , Quantitative Structure-Activity Relationship , Bile Acids and Salts/chemistry , Bile Acids and Salts/pharmacology , HEK293 Cells , Humans , Inhibitory Concentration 50 , Reproducibility of Results
5.
Arch Toxicol ; 89(12): 2253-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25319728

ABSTRACT

Soy isoflavones (IF) are phytoestrogens, which interact with estrogen receptors. They are extensively metabolized by glucuronosyltransferases and sulfotransferases, leading to the modulation of their estrogenic activity. It can be assumed that this biotransformation also has a crucial impact on the uptake of IF by active or passive cellular transport mechanisms, but little is known about the transport of IF phase II metabolites into the cell. Therefore, transport assays for phase II metabolites of daidzein (DAI) were carried out using HEK293 cell lines transfected with five human candidate carriers, i.e., organic anion transporter OAT4, sodium-dependent organic anion transporter (SOAT), Na(+)-taurocholate cotransporting polypeptide (NTCP), apical sodium-dependent bile acid transporter ASBT, and organic anion transporting polypeptide OATP2B1. Cellular uptake was monitored by UHPLC-DAD. DAI monosulfates were transported by the carriers NTCP and SOAT in a sodium-dependent manner, while OAT4-HEK293 cells revealed a partly sodium-dependent transport for these compounds. In contrast, DAI-7,4'-disulfate was only taken up by NTCP-HEK293 cells. DAI-7-glucuronide, but not DAI-4'-glucuronide, was transported exclusively by OATP2B1 in a sodium-independent manner. DAI-7-glucuronide-4'-sulfate, DAI-7-glucoside, and DAI were no substrate of any of the tested carriers. In addition, the inhibitory potency of the DAI metabolites toward estrone-sulfate (E1S) uptake of the above-mentioned carriers was determined. In conclusion, human SOAT, NTCP, OATP2B1, and OAT4 were identified as carriers for the DAI metabolites. Several metabolites were able to inhibit carrier-dependent E1S uptake. These findings might contribute to a better understanding of the bioactivity of IF especially in case of hormone-related cancers.


Subject(s)
Isoflavones/pharmacokinetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Phytoestrogens/pharmacokinetics , Symporters/metabolism , Biological Transport , Chromatography, High Pressure Liquid/methods , HEK293 Cells , Humans , Isoflavones/metabolism , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Sterol O-Acyltransferase/metabolism
6.
J Hepatol ; 61(4): 867-75, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24845614

ABSTRACT

BACKGROUND & AIMS: The human liver bile acid transporter Na(+)/taurocholate cotransporting polypeptide (NTCP) has recently been identified as liver-specific receptor for infection of hepatitis B virus (HBV), which attaches via the myristoylated preS1 (myr-preS1) peptide domain of its large surface protein to NTCP. Since binding of the myr-preS1 peptide to NTCP is an initiating step of HBV infection, we investigated if this process interferes with the physiological bile acid transport function of NTCP. METHODS: HBV infection, myr-preS1 peptide binding, and bile acid transport assays were performed with primary Tupaia belangeri (PTH) and human (PHH) hepatocytes as well as NTCP-transfected human hepatoma HepG2 cells allowing regulated NTCP expression, in the presence of various bile acids, ezetimibe, and myr-preS1 peptides. RESULTS: The myr-preS1 peptide of HBV inhibited bile acid transport in PTH and PHH as well as in NTCP-expressing HEK293 and HepG2 cells. Inversely, HBV infection of PTH, PHH, and NTCP-transfected HepG2 cells was inhibited in a concentration-dependent manner by taurine and glycine conjugates of cholic acid and ursodeoxycholic acid as well as by ezetimibe. In NTCP-HepG2 cells and PTH, NTCP expression, NTCP transport function, myr-preS1 peptide binding, and HBV infection followed comparable kinetics. CONCLUSIONS: Myr-preS1 virus binding to NTCP, necessary for productive HBV infection, interferes with the physiological bile acid transport function of NTCP. Therefore, HBV infection via NTCP may be lockable by NTCP substrates and NTCP-inhibiting drugs. This opens a completely new way for an efficient management of HBV infection by the use of NTCP-directed drugs.


Subject(s)
Bile Acids and Salts/metabolism , Carrier Proteins/metabolism , Hepatitis B virus/physiology , Hepatitis B , Hepatocytes/physiology , Membrane Glycoproteins/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Animals , Biological Transport , Hep G2 Cells , Hepatitis B/metabolism , Hepatitis B/virology , Humans , Tupaia , Viral Proteins/metabolism , Virus Internalization
7.
Drug Metab Rev ; 46(3): 291-324, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24666398

ABSTRACT

In general, xenobiotic metabolizing enzymes (XMEs) are expressed in lower levels in the extrahepatic tissues than in the liver, making the former less relevant for the clearance of xenobiotics. Local metabolism, however, may lead to tissue-specific adverse responses, e.g. organ toxicities, allergies or cancer. This review summarizes the knowledge on the expression of phase I and phase II XMEs and transporters in extrahepatic tissues at the body's internal-external interfaces. In the lung, CYPs of families 1, 2, 3 and 4 and epoxide hydrolases are important phase I enzymes, while conjugation is less relevant. In skin, phase I-related enzymatic reactions are considered less relevant. Predominant skin XMEs are phase II enzymes, whereby glucuronosyltransferases (UGT) 1, glutathione-S-transferase (GST) and N-acetyltransferase (NAT) 1 are important for detoxification. The intestinal epithelium expresses many transporters and phase I XME with high levels of CYP3A4 and CYP3A5 and phase II metabolism is mainly related to UGT, NAT and Sulfotransferases (SULT). In the kidney, conjugation reactions and transporters play a major role for excretion processes. In the bladder, CYPs are relevant and among the phase II enzymes, NAT1 is involved in the activation of bladder carcinogens. Expression of XMEs is regulated by several mechanisms (nuclear receptors, epigenetic mechanisms, microRNAs). However, the understanding why XMEs are differently expressed in the various tissues is fragmentary. In contrast to the liver - where for most XMEs lower expression is demonstrated in early life - the XME ontogeny in the extrahepatic tissues remains to be investigated.


Subject(s)
Biological Transport/physiology , Xenobiotics/metabolism , Animals , Cytochrome P-450 Enzyme System/metabolism , Humans , Membrane Transport Proteins/metabolism
8.
Drug Metab Rev ; 46(3): 261-82, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24483608

ABSTRACT

The historical phasing concept of drug metabolism and elimination was introduced to comprise the two phases of metabolism: phase I metabolism for oxidations, reductions and hydrolyses, and phase II metabolism for synthesis. With this concept, biological membrane barriers obstructing the accessibility of metabolism sites in the cells for drugs were not considered. The concept of two phases was extended to a concept of four phases when drug transporters were detected that guided drugs and drug metabolites in and out of the cells. In particular, water soluble or charged drugs are virtually not able to overcome the phospholipid membrane barrier. Drug transporters belong to two main clusters of transporter families: the solute carrier (SLC) families and the ATP binding cassette (ABC) carriers. The ABC transporters comprise seven families with about 20 carriers involved in drug transport. All of them operate as pumps at the expense of ATP splitting. Embedded in the former phase concept, the term "phase III" was introduced by Ishikawa in 1992 for drug export by ABC efflux pumps. SLC comprise 52 families, from which many carriers are drug uptake transporters. Later on, this uptake process was referred to as the "phase 0 transport" of drugs. Transporters for xenobiotics in man and animal are most expressed in liver, but they are also present in extra-hepatic tissues such as in the kidney, the adrenal gland and lung. This review deals with the function of drug carriers in various organs and their impact on drug metabolism and elimination.


Subject(s)
Biological Transport/physiology , Inactivation, Metabolic/physiology , Membrane Transport Proteins/metabolism , Xenobiotics/metabolism , Animals , Drug Carriers/metabolism , Humans , Liver/metabolism
9.
Brain Pathol ; 24(3): 270-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24372704

ABSTRACT

Biotin-thiamine-responsive basal ganglia disease (BTBGD) is a potentially treatable disorder caused by mutations in the SLC19A3 gene, encoding the human thiamine transporter 2. Manifestation of BTBGD as acute encephalopathy triggered by a febrile infection has been frequently reported, but the underlying mechanisms are not clear. We investigated a family with two brothers being compound heterozygous for the SLC19A3 mutations p.W94R and p.Q393*fs. Post-mortem analysis of the brain of one brother showed a mixture of acute, subacute and chronic changes with cystic and necrotic lesions and hemorrhage in the putamen, and hemorrhagic lesions in the caudate nucleus and cortical layers. SLC19A3 expression was substantially reduced in the cortex, basal ganglia and cerebellum compared with an age-matched control. Importantly, exposure of fibroblasts to stress factors such as acidosis or hypoxia markedly upregulated SLC19A3 in control cells, but failed to elevate SLC19A3 expression in the patient's fibroblasts. These results demonstrate ubiquitously reduced thiamine transporter function in the cerebral gray matter, and neuropathological alterations similar to Wernicke's disease in BTBGD. They also suggest that episodes of encephalopathy are caused by a substantially reduced capacity of mutant neuronal cells to increase SLC19A3 expression, necessary to adapt to stress conditions.


Subject(s)
Basal Ganglia Diseases/genetics , Basal Ganglia Diseases/pathology , Brain/metabolism , Membrane Transport Proteins/genetics , Stress, Psychological/physiopathology , Up-Regulation/physiology , Acidosis/pathology , Brain/drug effects , Brain/pathology , Cell Line, Transformed , DNA Mutational Analysis , Female , Fibroblasts/pathology , Humans , Hypoxia/pathology , Magnetic Resonance Imaging , Male , Membrane Transport Proteins/metabolism , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism
10.
PLoS One ; 8(5): e62638, 2013.
Article in English | MEDLINE | ID: mdl-23667501

ABSTRACT

Sulfated steroid hormones are commonly considered to be biologically inactive metabolites, but may be reactivated by the steroid sulfatase into biologically active free steroids, thereby having regulatory function via nuclear androgen and estrogen receptors which are widespread in the testis. However, a prerequisite for this mode of action would be a carrier-mediated import of the hydrophilic steroid sulfate molecules into specific target cells in reproductive tissues such as the testis. In the present study we detected predominant expression of the Sodium-dependent Organic Anion Transporter (SOAT), the Organic Anion Transporting Polypeptide 6A1, and the Organic Solute Carrier Partner 1 in human testis biopsies. All of these showed significantly lower or even absent mRNA expression in severe disorders of spermatogenesis (arrest at the level of spermatocytes or spermatogonia, Sertoli cell only syndrome). Only SOAT was significantly lower expressed in biopsies showing hypospermatogenesis. By use of immunohistochemistry SOAT was localized to germ cells at various stages in human testis biopsies showing normal spermatogenesis. SOAT immunoreactivity was detected in zygotene primary spermatocytes of stage V, pachytene spermatocytes of all stages (I-V), secondary spermatocytes of stage VI, and round spermatids (step 1 and step 2) in stages I and II. Furthermore, SOAT transport function for steroid sulfates was analyzed with a novel liquid chromatography tandem mass spectrometry procedure capable of profiling steroid sulfate molecules from cell lysates. With this technique, the cellular inward-directed SOAT transport was verified for the established substrates dehydroepiandrosterone sulfate and estrone-3-sulfate. Additionally, ß-estradiol-3-sulfate and androstenediol-3-sulfate were identified as novel SOAT substrates.


Subject(s)
Membrane Transport Proteins/metabolism , Oligospermia/metabolism , Organic Anion Transporters/metabolism , Steroids/metabolism , Testis/metabolism , Blotting, Western , Chromatography, Liquid , DNA Primers/genetics , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunohistochemistry , In Situ Hybridization , Male , Real-Time Polymerase Chain Reaction , Sulfates/metabolism , Tandem Mass Spectrometry
11.
Curr Top Membr ; 70: 105-68, 2012.
Article in English | MEDLINE | ID: mdl-23177985

ABSTRACT

The SLC10 family represents seven genes containing 1-12 exons that encode proteins in humans with sequence lengths of 348-477 amino acids. Although termed solute carriers (SLCs), only three out of seven (i.e. SLC10A1, SLC10A2, and SLC10A6) show sodium-dependent uptake of organic substrates across the cell membrane. These include the uptake of bile salts, sulfated steroids, sulfated thyroidal hormones, and certain statin drugs by SLC10A1 (Na(+)-taurocholate cotransporting polypeptide (NTCP)), the uptake of bile salts by SLC10A2 (apical sodium-dependent bile acid transporter (ASBT)), and uptake of sulfated steroids and sulfated taurolithocholate by SLC10A6 (sodium-dependent organic anion transporter (SOAT)). The other members of the family are orphan carriers not all localized in the cell membrane. The name "bile acid transporter family" arose because the first two SLC10 members (NTCP and ASBT) are carriers for bile salts that establish their enterohepatic circulation. In recent years, information has been obtained on their 2D and 3D membrane topology, structure-transport relationships, and on the ligand and sodium-binding sites. For SLC10A2, the putative 3D morphology was deduced from the crystal structure of a bacterial SLC10A2 analog, ASBT(NM). This information was used in this chapter to calculate the putative 3D structure of NTCP. This review provides first an introduction to recent knowledge about bile acid synthesis and newly found bile acid hormonal functions, and then describes step-by-step each individual member of the family in terms of expression, localization, substrate pattern, as well as protein topology with emphasis on the three functional SLC10 carrier members.


Subject(s)
Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/metabolism , Sodium/metabolism , Symporters/chemistry , Symporters/metabolism , Animals , Bile Acids and Salts/metabolism , Biological Transport , Cell Membrane/metabolism , Humans , Taurolithocholic Acid/metabolism
12.
Vet J ; 189(1): 67-71, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20655253

ABSTRACT

A 4-bp deletion mutation associated with multiple drug sensitivity exists in the canine multidrug resistance (MDR1) gene. This mutation has been detected in more than 10 purebred dog breeds as well as in mixed breed dogs. To evaluate the breed distribution of this mutation in Germany, 7378 dogs were screened, including 6999 purebred and 379 mixed breed dogs. The study included dog breeds that show close genetic relationship or share breeding history with one of the predisposed breeds but in which the occurrence of the MDR1 mutation has not been reported. The breeds comprised Bearded Collies, Anatolian Shepherd Dog, Greyhound, Belgian Tervuren, Kelpie, Borzoi, Australian Cattle Dog and the Irish Wolfhound. The MDR1 mutation was not detected is any of these breeds, although it was found as expected in the Collie, Longhaired Whippet, Shetland Sheepdog, Miniature Australian Shepherd, Australian Shepherd, Wäller, White Swiss Shepherd, Old English Sheepdog and Border Collie with varying allelic frequencies for the mutant MDR1 allele of 59%, 45%, 30%, 24%, 22%, 17%, 14%, 4% and 1%, respectively. Allelic frequencies of 8% and 2% were determined in herding breed mixes and unclassified mixed breeds, respectively. Because of its widespread breed distribution and occurrence in many mixed breed dogs, it is difficult for veterinarians and dog owners to recognise whether MDR1-related drug sensitivity is relevant for an individual animal. This study provides a comprehensive overview of all affected dog breeds and many dog breeds that are probably unaffected on the basis of ∼15,000 worldwide MDR1 genotyping data.


Subject(s)
Dogs/genetics , Genes, MDR/genetics , Mutation , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Drug Resistance, Multiple/genetics , Gene Frequency , Genetic Testing , Genotype , Germany , Species Specificity
13.
Vet J ; 185(3): 272-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19733104

ABSTRACT

For detection of the nt230[del4] MDR1 mutation, a 4-bp deletion in the canine MDR1 (ABCB1) gene, a TaqMan allelic discrimination assay was designed that allows for MDR1 genotyping without post-PCR processing. Directly after completion of the PCR amplification, the MDR1 genotype can be assigned based on selective fluorescence measurement. For primer selection the locus of a potential 265A>G single nucleotide polymorphism was omitted; this locus is covered by the oligonucleotide PCR primers from most of the hitherto established MDR1 genotyping methods. Dogs homozygous for the nt230[del4] MDR1 mutation show highly increased susceptibility to many drugs commonly used in veterinary medicine including ivermectin. As more than 10 dog breeds are predisposed to this mutation, reliable genotyping methods are necessary to identify affected dogs before drug treatment. This study provides a new allelic discrimination method that detects the MDR1 mutation with high specificity and reliability and is useful for routine diagnostics.


Subject(s)
Alleles , Dogs/genetics , Genes, MDR/genetics , Polymerase Chain Reaction/veterinary , Sequence Deletion/genetics , Taq Polymerase/metabolism , Animals , DNA Mutational Analysis/veterinary , DNA Primers , Fluorescent Dyes/metabolism , Genotype , Polymerase Chain Reaction/methods , Reproducibility of Results , Sensitivity and Specificity , Species Specificity
14.
Biochem Biophys Res Commun ; 361(1): 26-32, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17632081

ABSTRACT

Here we report the identification, cloning, and characterization of SLC10A5, which is a new member of Solute Carrier Family 10 (SLC10), also known as the "sodium/bile acid cotransporter family". Expression of SLC10A5/Slc10a5 was examined by quantitative real-time PCR and revealed its highest expression levels in liver and kidney in humans, rat and mouse. In rat liver and kidney, Slc10a5 expression was localized by in situ hybridization to hepatocytes and proximal tubules, respectively. A SLC10A5-FLAG fusion protein was expressed in HEK293 cells and showed an apparent molecular weight of 42 kDa after immunoprecipitation. When expressed in Xenopus laevis oocytes, the SLC10A5-FLAG protein was detected in the oocyte's plasma membrane but showed no transport activity for taurocholate, cholate, estrone-3-sulfate, or dehydroepiandrosterone sulfate. As bile acid carriers are the most related carriers to SLC10A5 though, we strongly suppose that SLC10A5 is an orphan carrier with yet non-identified substrates.


Subject(s)
Kidney/metabolism , Liver/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Gene Expression , Humans , Mice , Molecular Sequence Data , Oocytes/metabolism , Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/genetics , Rats , Symporters/chemistry , Symporters/genetics , Xenopus laevis
15.
Eur J Cell Biol ; 86(8): 445-60, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17628207

ABSTRACT

The 'Solute Carrier Family SLC10' consists of six annotated members in humans, comprising two bile acid carriers (SLC10A1 and SLC10A2), one steroid sulfate transporter (SLC10A6), and three orphan carriers (SLC10A3 to SLC10A5). In this study we report molecular characterization and expression analysis of a novel member of the SLC10 family, SLC10A7, previously known as C4orf13. SLC10A7 proteins consist of 340-343 amino acids in humans, mice, rats, and frogs and show an overall amino acid sequence identity of >85%. SLC10A7 genes comprise 12 coding exons and show broad tissue expression pattern. When expressed in Xenopus laevis oocytes and HEK293 cells, SLC10A7 was detected in the plasma membrane but revealed no transport activity for bile acids and steroid sulfates. By immunofluorescence analysis of dual hemagglutinin (HA)- and FLAG-labeled SLC10A7 proteins in HEK293 cells, we established a topology of 10 transmembrane domains with an intracellular cis orientation of the N-terminal and C-terminal ends. This topology pattern is clearly different from the seven-transmembrane domain topology of the other SLC10 members but similar to hitherto uncharacterized non-vertebrate SLC10A7-related proteins. In contrast to the established SLC10 members, which are restricted to the taxonomic branch of vertebrates, SLC10A7-related proteins exist also in yeasts, plants, and bacteria, making SLC10A7 taxonomically the most widespread member of this carrier family. Vertebrate and bacterial SLC10A7 proteins exhibit >20% sequence identity, which is higher than the sequence identity of SLC10A7 to any other member of the SLC10 carrier family.


Subject(s)
Bacteria/genetics , Conserved Sequence , Organic Anion Transporters, Sodium-Dependent/genetics , Phylogeny , Vertebrates/genetics , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Biological Transport , Cell Membrane/metabolism , Cloning, Molecular , Exons/genetics , Genome , Humans , Immunoprecipitation , Mice , Molecular Sequence Data , Oocytes , Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Symporters/metabolism , Xenopus
16.
J Biol Chem ; 282(27): 19728-41, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17491011

ABSTRACT

We have cloned human sodium-dependent organic anion transporter (SOAT) cDNA, which consists of 1502 bp and encodes a 377-amino acid protein. SOAT shows 42% sequence identity to the ileal apical sodium-dependent bile acid transporter ASBT and 33% sequence identity to the hepatic Na(+)/taurocholate-cotransporting polypeptide NTCP. Immunoprecipitation of a SOAT-FLAG-tagged protein revealed a glycosylated form at 46 kDa that decreased to 42 kDa after PNGase F treatment. SOAT exhibits a seven-transmembrane domain topology with an outside-to-inside orientation of the N-terminal and C-terminal ends. SOAT mRNA is most highly expressed in testis. Relatively high SOAT expression was also detected in placenta and pancreas. We established a stable SOAT-HEK293 cell line that showed sodium-dependent transport of dehydroepiandrosterone sulfate, estrone-3-sulfate, and pregnenolone sulfate with apparent K(m) values of 28.7, 12.0, and 11.3 microm, respectively. Although bile acids, such as taurocholic acid, cholic acid, and chenodeoxycholic acid, were not substrates of SOAT, the sulfoconjugated bile acid taurolithocholic acid-3-sulfate was transported by SOAT-HEK293 cells in a sodium-dependent manner and showed competitive inhibition of SOAT transport with an apparent K(i) value of 0.24 mum. Several nonsteroidal organosulfates also strongly inhibited SOAT, including 1-(omega-sulfooxyethyl)pyrene, bromosulfophthalein, 2- and 4-sulfooxymethylpyrene, and alpha-naphthylsulfate. Among these inhibitors, 2- and 4-sulfooxymethylpyrene were competitive inhibitors of SOAT, with apparent K(i) values of 4.3 and 5.5 microm, respectively, and they were also transported by SOAT-HEK293 cells.


Subject(s)
Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters/metabolism , Protein Processing, Post-Translational/physiology , Amino Acid Sequence , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Female , Humans , Male , Membrane Transport Modulators/pharmacology , Molecular Sequence Data , Organ Specificity/physiology , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Dependent/genetics , Pancreas/metabolism , Placenta/metabolism , Pregnancy , Sequence Homology, Amino Acid , Symporters/genetics , Testis/metabolism
17.
Comp Biochem Physiol B Biochem Mol Biol ; 137(3): 317-29, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15050519

ABSTRACT

We describe the cloning, functional characterization and tissue localization of a novel membrane transporter of the OATP/Oatp-gene family obtained from liver and kidney of cattle (Bos taurus). The carrier protein exhibits highest sequence identity to the human OATP1A2 (previously called OATP-A) and is, therefore, named bovine Oatp1a2. Bovine Oatp1a2 received the gene symbol Slco1a2 that is identical to the SLC classification of human OATP1A2 (SLCO1A2, previously called SLC21A3) and is likely an orthologue of the human gene. Two different full-length bOatp1a2 cDNAs of 2316-bp and 3504-bp were obtained and encoded for a 666 amino acid membrane protein, which contains twelve putative transmembrane spanning domains. Bovine Oatp1a2 expression was detected in liver, kidney, brain and adrenal gland. Uptake studies in cRNA-injected oocytes demonstrated that bOatp1a2 transports estrone-3-sulfate and taurocholate, with K(m) values of 9.6 microM and 51 microM, respectively, and estradiol-17beta-glucuronide. However, the structurally-related heart glycosides ouabain (1 microM) and digoxin (1 microM) are neither transported by bovine Oatp1a2 nor by human OATP1A2. We conclude that based on the tested substrates bovine Oatp1a2 shows functional homology to human OATP1A2.


Subject(s)
Cloning, Molecular/methods , Estradiol/analogs & derivatives , Estrone/analogs & derivatives , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Animals , Base Sequence , Biological Transport , Cattle , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Estradiol/metabolism , Estrone/metabolism , Kinetics , Membrane Proteins , Molecular Sequence Data , Organic Anion Transporters/chemistry , Sequence Alignment , Taurocholic Acid/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...