Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Clin Transl Sci ; 10(5): 412-420, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28689374

ABSTRACT

Cisplatin is among the most widely used anticancer drugs and known to cause a dose-limiting nephrotoxicity, which is partially dependent on the renal uptake carrier OCT2. We here report a previously unrecognized, OCT2-independent pathway of cisplatin-induced renal injury that is mediated by the organic anion transporters OAT1 and OAT3. Using transporter-deficient mouse models, we found that this mechanism regulates renal uptake of a mercapturic acid metabolite of cisplatin that acts as a precursor of a potent nephrotoxin. The function of these two transport systems can be simultaneously inhibited by the tyrosine kinase inhibitor nilotinib through noncompetitive mechanisms, without compromising the anticancer properties of cisplatin. Collectively, our findings reveal a novel pathway that explains the fundamental basis of cisplatin-induced nephrotoxicity, with potential implications for its therapeutic management.


Subject(s)
Cisplatin/toxicity , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Animals , Biological Transport/drug effects , Cell Death/drug effects , Gene Expression Profiling , Kidney/drug effects , Kidney/metabolism , Male , Metabolome/drug effects , Mice, Inbred C57BL , Organic Anion Transport Protein 1/deficiency , Organic Anion Transporters, Sodium-Independent/deficiency , Phenotype , Pyrimidines/pharmacology
2.
Kidney Int ; 69(5): 837-45, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16518343

ABSTRACT

Organic anion and cation transporters (OATs, OCTs, and OCTNs) mediate the proximal tubular secretion of numerous clinically important compounds, including various commonly prescribed pharmaceuticals. Here, we report determination of the ontogeny of these transporters and of NaP(i)2 and SGLT1, using quantitative polymerase chain reaction (QPCR) to determine expression levels of transporter genes in rat embryonic kidneys on each day of gestation from embryonic day (ed) 13 to ed18, in cultures of induced and uninduced metanephric mesenchyme (MM), and on each day of 1 week of whole embryonic kidney (WEK) culture. We also examined ontogeny of Oat1 protein expression in rat embryonic kidney by immunohistochemistry. Finally, we used uptake of fluorescein (FL) as a novel in vitro functional assay of OAT expression in WEK and MM. Developmental induction of OAT and OCT genes does not occur uniformly: some genes are induced early (e.g., Oat1 and Oat3, potential early markers of proximal tubulogenesis), and others after kidney development is relatively advanced (e.g., Oct1, a potential marker of terminal differentiation). The ontogeny of transporter genes in WEK and MM is similar to that observed in vivo, indicating that these organ culture systems may represent convenient in vitro models to study the developmental induction of OATs, OCTs, and OCTNs. Functional transport was evidenced by accumulation of FL in the developing tubule in WEK and MM organ cultures. Our findings on the renal ontogeny of OATs and OCTs could carry implications both for the development of more rational therapeutics for premature infants, as well as for our understanding of proximal tubule differentiation.


Subject(s)
Kidney/embryology , Kidney/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Animals , Base Sequence , Female , Gene Expression Regulation, Developmental , Gestational Age , Mesoderm/metabolism , Organ Culture Techniques , Pregnancy , RNA/genetics , RNA/metabolism , Rats , Rats, Sprague-Dawley
3.
Mol Pharmacol ; 60(5): 1091-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11641438

ABSTRACT

Mercapturic acids are N-acetyl-L-cysteine S-conjugates that are formed from a range of endogenous and exogenous chemicals. Although the kidney is a major site for elimination of mercapturic acids, the transport mechanisms involved have not been identified. The present study examined whether mercapturic acids are substrates for the renal basolateral organic anion transporter-1 (Oat1) from rat kidney. This carrier mediates uptake of organic anions from the bloodstream in exchange for intracellular alpha-ketoglutarate. Uptake of [(3)H]p-aminohippuric acid (PAH) in Oat1-expressing Xenopus laevis oocytes was strongly inhibited by S-(2,4-dinitrophenyl)-N-acetyl-L-cysteine (DNP-NAC) and by all other mercapturic acids tested, including the endogenous mercapturic acid N-acetyl-leukotriene E(4). Inhibition by the mercapturic acids was competitive, which is consistent with the hypothesis that these compounds are substrates for Oat1. This conclusion was supported by the direct demonstration of saturable [(35)S]DNP-NAC uptake in Oat1-expressing oocytes. [(35)S]DNP-NAC uptake was inhibited by PAH and other mercapturic acids and was stimulated in oocytes preloaded with glutarate. The apparent K(m) value for DNP-NAC uptake was only 2 microM, indicating that this mercapturic acid is a high affinity substrate for Oat1. Together, these data indicate that clearance of endogenous mercapturic acids is an important function of the renal organic anion transporter.


Subject(s)
Acetylcysteine/metabolism , Kidney/metabolism , Organic Anion Transport Protein 1/metabolism , Acetylcysteine/analogs & derivatives , Animals , Humans , Oocytes/metabolism , Tritium , Xenopus laevis , p-Aminohippuric Acid/metabolism
4.
J Biol Chem ; 276(45): 41611-9, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11553644

ABSTRACT

To determine whether organic cation transporter (OCT) family members might mediate choline transport in choroid plexus (CP), the handling of choline by cloned transporters and by intact CP isolated from the adult rat was investigated. Expression of OCT1 and OCT2 in Xenopus oocytes increased hemicholinium-3-sensitive choline uptake. In contrast, OCT3 did not mediate choline transport. Estimated K(m) values for choline in rOCT1-, rOCT2-, and hOCT2-expressing oocytes were 346 +/- 50, 441 +/- 67, and 102 +/- 80 microm, respectively. Membrane potential was the major driving force for choline uptake in rat and human OCT2-expressing oocytes and in intact CP in vitro. Lowering of medium pH (6 versus 7.4) was equally effective at inhibiting choline uptake in CP, suggesting that there might be a non-OCT component of choline uptake that is responsive to an H(+) gradient. However, choline efflux from CP was not stimulated by a trans-applied H(+) gradient. Choline uptake by CP was Na(+)-independent with an estimated K(m) of 183 microm. Reverse transcriptase-polymerase chain reaction detected OCT2 and OCT3, but not OCT1, mRNA expression in CP. Transfection of intact CP with a rOCT2/green fluorescent protein fusion construct resulted in strong apical membrane fluorescence with no detectable signal in the basal and lateral plasma membranes. These data indicate that OCT2 mediates choline transport across the ventricular membrane of CP.


Subject(s)
Choline/metabolism , Choroid Plexus/metabolism , Organic Cation Transport Proteins/physiology , Animals , Biological Transport , Male , Organic Cation Transport Proteins/analysis , Organic Cation Transporter 2 , Rats , Rats, Sprague-Dawley , Xenopus laevis
5.
Am J Physiol Renal Physiol ; 281(2): F197-205, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11457711

ABSTRACT

The organic anion transporter (OAT) family handles a wide variety of clinically important compounds (antibiotics, nonsteriodal anti-inflammatory drugs, etc.) and toxins. However, little is known about their appearance during development despite documented differences in the handling of anionic drugs among neonates, children, and adults. A similar spatiotemporal pattern of mRNA expression of the OATs (OAT1-4) during kidney development suggests that OAT genes may be useful in understanding the mechanisms of proximal tubule maturation. Moreover, OAT expression in unexpected extrarenal sites (e.g., spinal cord, bone, skin) has also been detected during development, possibly indicating a role for these transporters in the formation or preservation of extrarenal tissues. The cloning of these transporters also paves the way for computer-based modeling of drug-transporter interactions at the molecular level, potentially aiding in the design and assessment of new drugs. Additionally, increased understanding of single nucleotide polymorphisms in OATs and other transporters may eventually allow the use of a patient's expression profile and polymorphisms to individualize drug therapy.


Subject(s)
Carrier Proteins/metabolism , Kidney/physiology , Animals , Anion Transport Proteins , Biological Transport , Carrier Proteins/genetics , Humans , Inactivation, Metabolic , Kidney/cytology , Kidney/growth & development , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/physiology , Models, Biological , Pharmacogenetics , Phylogeny , Protein Structure, Tertiary
6.
J Pharmacol Exp Ther ; 296(2): 450-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160630

ABSTRACT

Retinal pigment epithelial (RPE) cells transport a variety of solutes, but the capacity of human RPE cells to transport drugs and xenobiotics is not well understood. As an initial step to address this issue, we have examined human RPE transport of verapamil. Transport of [3H]verapamil was measured in two human RPE cell lines (RPE/Hu and ARPE-19) grown to confluence on 12-well culture plates. Verapamil uptake by RPE/Hu cells was highly concentrative, reaching cell-to-medium ratios as high as 42 by 1 h. Uptake was saturable, with an apparent K(m) of 7.2 microM. Verapamil uptake decreased in the presence of metabolic inhibitors, low temperature, and organic cations, including quinidine, pyrilamine, quinacrine, and diphenhydramine. However, other organic cations, including tetraethylammonium and cimetidine failed to inhibit. Verapamil uptake was also inhibited by the cationic antiglaucoma drugs diltiazem, timolol, and propranolol. Verapamil uptake was insensitive to changes in membrane potential. However, transport was markedly altered by changes in pH. Decreasing external pH inhibited uptake, whereas efflux was stimulated. Intracellular acidification via NH4Cl prepulse also stimulated uptake. Identical findings were obtained using the commercially available cell line ARPE-19. In view of its unique specificity, the RPE cell verapamil transporter described above is a novel, heretofore undescribed, organic cation transporter, distinct from the known members of the OCT family of organic cation transporters.


Subject(s)
Calcium Channel Blockers/metabolism , Carrier Proteins/metabolism , Pigment Epithelium of Eye/metabolism , Verapamil/metabolism , Algorithms , Cells, Cultured , Diphenhydramine/pharmacology , Histamine H1 Antagonists/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics
7.
Am J Physiol Renal Physiol ; 279(5): F826-34, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053042

ABSTRACT

The localization of organic cation transporter 2 (OCT2) within renal cells is the subject of considerable controversy, resulting in marked uncertainty as to its function. To resolve this issue, we made an OCT2/green fluorescent protein (GFP) fusion construct (rOCT2-GFP) and determined its localization within Xenopus laevis oocytes and renal cells using confocal microscopy. Oocytes expressing rOCT2-GFP exhibited plasma membrane fluorescence as well as greatly increased specific, potential-driven uptake of [(14)C]tetraethylammonium (TEA). Polarized monolayers of renal epithelial cell lines [LLC-PK(1) and Madin-Darby canine kidney (MDCK)] transiently transfected with pEGFP-C3, which codes for a cytoplasmic GFP, showed a diffuse, evenly distributed cytoplasmic signal with no plasma membrane fluorescence. In contrast, cells transiently transfected with pEGFP-C3/rOCT2 (the vector coding for rOCT2-GFP) showed predominantly plasma membrane fluorescence, which was most prominent in the lateral membrane. MDCK cells stably expressing rOCT2-GFP (MDCK/rOCT2-GFP) maintained in long-term culture showed a greatly increased basal and lateral membrane fluorescence. When grown on porous supports, MDCK/rOCT2-GFP monolayers showed specific, potential-driven TEA uptake from the basal side. Finally, expression and distribution of rOCT2-GFP were investigated in isolated killifish (Fundulus heteroclitus) renal proximal tubules. On expression of rOCT2-GFP, transfected tubules showed marked basal and lateral membrane fluorescence, with no detectable signal at the apical membrane. In contrast, tubules expressing a luminal sodium-dicarboxylate cotransporter (rbNaDC-1)-GFP construct showed apical membrane fluorescence, and tubules expressing cytoplasmic GFP had a diffuse cytoplasmic fluorescence. These results indicate that rOCT2 is basolateral in renal proximal tubule cells.


Subject(s)
Carrier Proteins/metabolism , Kidney Tubules, Proximal/metabolism , Organic Cation Transport Proteins , Animals , Biological Transport/genetics , Carrier Proteins/genetics , Cell Line , Dogs , Gene Expression , Green Fluorescent Proteins , In Vitro Techniques , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/cytology , Killifishes , Luminescent Proteins/genetics , Microscopy, Fluorescence , Oocytes/cytology , Oocytes/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Swine , Transfection , Xenopus laevis
8.
Am J Physiol ; 277(6): F890-8, 1999 12.
Article in English | MEDLINE | ID: mdl-10600936

ABSTRACT

The driving forces mediating tetraethylammonium (TEA) transport were systematically assessed in Xenopus oocytes and MDCK cells expressing organic cation transporter (OCT) 2 cloned from rat kidney (rOCT2). In rOCT2 cRNA-injected oocytes, uptake of [14C]TEA was saturable, with an estimated Michaelis constant (Km) of 393 microM, and was specifically inhibited by organic cations. Furthermore, TEA uptake demonstrated two distinct components, one that was potential sensitive and one that was pH sensitive. When membrane potential was intact, TEA uptake was largely unaffected by changes in medium pH; when the oocyte membrane was depolarized (K+ in = out = 102.5 mM, plus valinomycin), decreasing external medium pH significantly reduced TEA uptake. Consistent with the potential sensitivity of uptake, electrophysiological analysis of rOCT2-injected oocytes demonstrated movement of positive charge into the oocyte upon TEA addition. To further evaluate the nature of the pH effect and assess the properties of rOCT2 in a renal epithelium, rOCT2 was introduced into MDCK cells. A stably transfected single cell clone (MDCK-rOCT2) showed mediated, potential-sensitive, pH-sensitive TEA uptake (Km = 48 microM). TEA efflux from preloaded MDCK-rOCT2 cells was stimulated by externally applied (trans) tetramethylammonium but was trans-inhibited by H+ (external pH 5.4). The effect of external H+ was to modulate rOCT2-mediated transport. Thus rOCT2 is a potential-driven transporter, not an organic cation/H+ exchanger, consistent with a physiological role in the basolateral entry step in renal organic cation secretion.


Subject(s)
Carrier Proteins/physiology , Organic Cation Transport Proteins , Animals , Biological Transport , Carrier Proteins/genetics , Cell Membrane/physiology , Cells, Cultured , Cyclosporine/pharmacology , Dogs , Female , Gene Library , In Vitro Techniques , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Oocytes/physiology , Organic Cation Transporter 2 , Probenecid/pharmacology , Quinacrine/pharmacology , Quinine/pharmacology , Rats , Recombinant Proteins/metabolism , Tetraethylammonium/pharmacokinetics , Tetraethylammonium/pharmacology , Xenopus laevis
9.
J Biol Chem ; 274(47): 33382-7, 1999 Nov 19.
Article in English | MEDLINE | ID: mdl-10559217

ABSTRACT

The mechanism and membrane localization of choroid plexus (CP) organic anion transport were determined in apical (or brush border) membrane vesicles isolated from bovine choroid plexus and in intact CP tissue from cow and rat. Brush border membrane vesicles were enriched in Na(+),K(+)-ATPase (20-fold; an apical marker in CP) and demonstrated specific, sodium-coupled transport of proline, glucose, and glutarate. Vesicular uptake of the anionic herbicide 2, 4-dichlorophenoxyacetic acid (2,4-D) was markedly stimulated by an inward sodium gradient but only in the presence of glutarate, indicating the presence of apical dicarboxylate/organic anion exchange. Consistent with this interpretation, an imposed outward glutarate gradient stimulated 2,4-D uptake in the absence of sodium. Under both conditions, uptake was dramatically slowed and overshoot was abolished by probenecid. Likewise, apical accumulation of 2,4-D by intact bovine choroid plexus tissue in vitro was stimulated by external glutarate in the presence of sodium. Glutarate stimulation was abolished by 5 mM LiCl. Identical findings were obtained using rat CP tissue, which showed both sodium/glutarate-stimulated 2,4-D (tissue/medium (T/M) approximately 8) and p-aminohippurate (T/M = 2) transport. Finally, since the renal exchanger (rROAT1) has been cloned in rat kidney, a rROAT1-green fluorescent protein construct was used to analyze exchanger distribution directly in transiently transfected rat CP. As predicted by the functional studies, the fluorescently tagged transporter was seen in apical but not basolateral membranes of the CP.


Subject(s)
Choroid Plexus/metabolism , Glucose/metabolism , Glutarates/metabolism , Proline/metabolism , 2,4-Dichlorophenoxyacetic Acid/metabolism , Animals , Anion Transport Proteins , Anions , Carrier Proteins/metabolism , Cattle , Green Fluorescent Proteins , Herbicides/metabolism , Ion Transport , Luminescent Proteins/metabolism , Male , Microvilli/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism
11.
Mol Pharmacol ; 56(3): 570-80, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10462545

ABSTRACT

Nephrotoxicity is the dose-limiting clinical adverse effect of cidofovir and adefovir, two potent antiviral therapeutics. Because renal uptake likely plays a role in the etiology of cidofovir- and adefovir-associated nephrotoxicity, we attempted to identify a renal transporter capable of interacting with these therapeutics. A cDNA clone was isolated from a human renal library and designated human organic anion transporter 1 (hOAT1). Northern analysis detected a specific 2.5-kilobase pair hOAT1 transcript only in human kidney. However, reverse transcription-polymerase chain reaction revealed hOAT1 expression in human brain and skeletal muscle, as well. Immunoblot analysis of human kidney cortex demonstrated that hOAT1 is an 80- to 90-kilodalton heterogeneous protein modified by abundant N-glycosylation. Xenopus laevis oocytes expressing hOAT1 supported probenecid-sensitive uptake of [(3)H]p-aminohippurate (K(m) = 4 microM), which was trans-stimulated in oocytes preloaded with glutarate. Importantly, both hOAT1 and rat renal organic anion transporter 1 (rROAT1) mediated saturable, probenecid-sensitive uptake of cidofovir, adefovir, and other nucleoside phosphonate antivirals. The affinity of hOAT1 toward cidofovir and adefovir (K(m) = 46 and 30 microM, respectively) was 5- to 9-fold higher compared with rROAT1 (K(m) = 238 and 270 microM, respectively). These data indicate that hOAT1 may significantly contribute to the accumulation of cidofovir and adefovir in renal proximal tubules and, thus, play an active role in the mechanism of nephrotoxicity associated with these antiviral therapeutics.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/metabolism , Carrier Proteins/genetics , Cytosine/analogs & derivatives , Kidney/drug effects , Organophosphonates , Organophosphorus Compounds/metabolism , Adenine/metabolism , Adenine/pharmacology , Amino Acid Sequence , Animals , Anion Transport Proteins , Antiviral Agents/pharmacology , Base Sequence , Biological Transport , Carrier Proteins/metabolism , Cidofovir , Cloning, Molecular , Cytosine/metabolism , Cytosine/pharmacology , DNA, Complementary/analysis , Humans , Immunoblotting , Kidney/metabolism , Molecular Sequence Data , Nucleosides/metabolism , Organophosphorus Compounds/pharmacology , Rats
12.
Am J Physiol ; 276(6): F864-73, 1999 06.
Article in English | MEDLINE | ID: mdl-10362775

ABSTRACT

The organic anion transporter, rROAT1, is a dicarboxylate/organic anion exchanger, a function associated with the basolateral membrane in rat proximal tubule. To directly establish the subcellular localization of rROAT1 in renal epithelia, we made a rROAT1-green fluorescent protein (GFP) fusion construct (rROAT1-GFP). Plasma membrane-associated fluorescence was observed in rROAT1-GFP-expressing Xenopus oocytes examined by confocal microscopy. Uptake of 3H-labeled p-aminohippurate (PAH) increased 2. 5-fold in rROAT1-GFP-expressing Xenopus oocytes, and this increase was abolished by 1 mM probenecid. Thus the construct was capable of specific organic anion transport. Cultured renal epithelial cell lines (MDCK and LLC-PK1) transfected with the vector pEGFP-C3 showed a diffuse, evenly distributed cytoplasmic signal. However, when transfected with pEGFP-C3/rROAT1 (vector coding for rROAT1-GFP), both cell lines showed predominantly plasma membrane fluorescence. The expression and distribution of rROAT1-GFP in intact renal proximal tubules was also investigated. Isolated killifish (Fundulus heteroclitus) renal tubules transfected with pEGFP-C3/rROAT1 showed marked basal and lateral membrane-associated fluorescence, but no detectable signal in the nucleus or the apical pole of tubule cells. Tubules transfected with pEGFP-C3 showed diffuse cytoplasmic fluorescence. Function of the rROAT1-GFP construct was demonstrated in transfected killifish tubules by fluorescein transport assay. These results demonstrate the basolateral subcellular localization of rROAT1 in polarized renal epithelia and validate a new technique for localizing cloned transporters within intact renal tubules.


Subject(s)
Carrier Proteins/genetics , Indicators and Reagents , Kidney Tubules, Proximal/metabolism , Luminescent Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Animals , Anion Transport Proteins , Cell Line , Dogs , Green Fluorescent Proteins , Killifishes , LLC-PK1 Cells , Oocytes/metabolism , Rats , Swine , Tissue Distribution , Xenopus
13.
Toxicol Sci ; 47(2): 181-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10220855

ABSTRACT

A cDNA for the organic cation transporter (rOCT2) of the rat kidney was inserted into the retroviral plasmid pLXSN. This plasmid was used to stably transfect NIH3T3 cells. The transfected cell line exhibited an enhanced rate of tetraethylammonium (TEA) uptake and efflux compared to wild-type NIH3T3 cells. Uptake of TEA by the transfected cells was markedly reduced upon incubation at 4 degrees C. When the extracellular pH was lowered from 8.1 to 5.9, uptake was also reduced, suggesting inhibition of rOCT2 by extracellular protons. The apparent K(m) for TEA in the transfected cells was 141 microM. The classical organic cation transport inhibitors, cyanine 863 and cimetidine, produced noncompetitive inhibition with apparent Ki values of 0.81 and 198 microM, respectively. Daunomycin, vinblastine, and the deoxyadenosine analogs, 2'-deoxytubercidin and 2-chlorodeoxyadenosine, did not appear to be substrates for rOCT2. However, the anticancer drug, cisplatin, competitively inhibited TEA uptake by rOCT2 with an apparent Ki value of 925 microM, suggesting that rOCT2 may play a role in its renal secretion. In summary, transfected NIH3T3 cells provide a facile system by which this and other organic ion transporters can be studied.


Subject(s)
Carrier Proteins/genetics , DNA, Complementary/isolation & purification , Kidney/metabolism , Models, Genetic , Organic Cation Transport Proteins , Transfection , 3T3 Cells , Animals , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Daunorubicin/pharmacology , Deoxyadenosines/pharmacology , Kinetics , Mice , Organic Cation Transporter 2 , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tetraethylammonium Compounds/metabolism , Vinblastine/pharmacology
14.
J Biol Chem ; 272(48): 30088-95, 1997 Nov 28.
Article in English | MEDLINE | ID: mdl-9374486

ABSTRACT

Expression cloning in Xenopus laevis oocytes was used to isolate an organic anion transport protein from rat kidney. A cDNA library was constructed from size-fractionated poly(A)+ RNA and screened for probenecid-sensitive transport of p-aminohippurate (PAH). A 2, 227-base pair cDNA clone containing a 1,656-base pair open reading frame coding for a peptide 551 amino acids long was isolated and named ROAT1. ROAT1-mediated transport of 50 mu M [3H]PAH was independent of imposed changes in membrane potential. Transport was significantly inhibited at 4 degrees C, or upon incubation with other organic anions, but not by the organic cation tetraethylammonium, by the multidrug resistance ATPase inhibitor cyclosporin A, or by urate. External glutarate and alpha-ketoglutarate (1 mM), both counterions for basolateral PAH exchange, also inhibited transport, suggesting that ROAT1 is functionally similar to the basolateral PAH carrier. Consistent with this conclusion, PAH uptake was trans-stimulated in oocytes preloaded with glutarate, whereas the dicarboxylate methylsuccinate, which is not accepted by the basolateral exchanger, did not trans-stimulate. Finally, ROAT1-mediated PAH transport was saturable, with an estimated Km of 70 mu M. Each of these properties is identical to those previously described for the basolateral alpha-ketoglutarate/PAH exchanger in isolated membrane vesicles or intact renal tubules.


Subject(s)
Carrier Proteins/genetics , Kidney/chemistry , Organic Anion Transporters , Amino Acid Sequence , Animals , Anion Transport Proteins , Base Sequence , Biological Transport , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Polarity , Cloning, Molecular , Gene Expression , Kidney/metabolism , Membrane Potentials , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Molecular Weight , Oocytes , Organic Anion Transport Protein 1 , RNA, Messenger/genetics , Rats , Sequence Alignment , Solubility , Substrate Specificity , Tissue Distribution , Xenopus laevis , p-Aminohippuric Acid/metabolism
15.
Mol Cell Biol ; 17(11): 6223-35, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9343383

ABSTRACT

In Saccharomyces cerevisiae UV radiation and a variety of chemical DNA-damaging agents induce the transcription of specific genes, including several involved in DNA repair. One of the best characterized of these genes is PHR1, which encodes the apoenzyme for DNA photolyase. Basal-level and damage-induced expression of PHR1 require an upstream activation sequence, UAS(PHR1), which has homology with DRC elements found upstream of at least 19 other DNA repair and DNA metabolism genes in yeast. Here we report the identification of the UME6 gene of S. cerevisiae as a regulator of UAS(PHR1) activity. Multiple copies of UME6 stimulate expression from UAS(PHR1) and the intact PHR1 gene. Surprisingly, the effect of deletion of UME6 is growth phase dependent. In wild-type cells PHR1 is induced in late exponential phase, concomitant with the initiation of glycogen accumulation that precedes the diauxic shift. Deletion of UME6 abolishes this induction, decreases the steady-state concentration of photolyase molecules and PHR1 mRNA, and increases the UV sensitivity of a rad2 mutant. Despite the fact that UAS(PHR1) does not contain the URS1 sequence, which has been previously implicated in UME6-mediated transcriptional regulation, we find that Ume6p binds to UAS(PHR1) with an affinity and a specificity similar to those seen for a URS1 site. Similar binding is also seen for DRC elements from RAD2, RAD7, and RAD53, suggesting that UME6 contributes to the regulated expression of a subset of damage-responsive genes in yeast.


Subject(s)
Apoenzymes/genetics , Cell Cycle Proteins , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Deoxyribodipyrimidine Photo-Lyase/genetics , Endodeoxyribonucleases , Gene Expression Regulation, Fungal , Membrane Glycoproteins , Protein Serine-Threonine Kinases , Repressor Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Transcription Factors/metabolism , Apoenzymes/biosynthesis , Binding Sites , Checkpoint Kinase 2 , Cloning, Molecular , Deoxyribodipyrimidine Photo-Lyase/biosynthesis , Fungal Proteins/genetics , Gene Deletion , Gene Dosage , Genes, Fungal , Mutagenesis , Protein Binding , Protein Kinases/genetics , Pyrimidine Dimers/metabolism , Regulatory Sequences, Nucleic Acid , Saccharomyces cerevisiae/radiation effects , Transcription, Genetic , Ultraviolet Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...