hosted by
publicationslist.org
    

Naohiko Anzai


anzai@ks.kyorin-u.ac.jp

Journal articles

2010
Hiroki Tsuchida, Naohiko Anzai, Ho J Shin, Michael F Wempe, Promsuk Jutabha, Atsushi Enomoto, Seok H Cha, Takeo Satoh, Masashi Ishida, Hiroyuki Sakurai, Hitoshi Endou (2010)  Identification of a novel organic anion transporter mediating carnitine transport in mouse liver and kidney.   Cell Physiol Biochem 25: 4-5. 511-522 03  
Abstract: This study identifies a novel organic anion transporter Oat9 expressed in mouse liver and kidney. Two variants were detected by screening a mouse liver cDNA library; these varients consist of 1815 (designated Oat9S) and 2165 (Oat9L) base pairs which encode 443 and 551 amino acid proteins, respectively. Oat9S has a predicted structure containing eight transmembrane domains (TMD); whereas, Oat9L possesses twelve TMD. Oat9 mRNA expression was detected in kidney and liver. This transporter was located at the apical side of the late portion of proximal tubules and at the sinusoidal side of hepatocytes. When expressed in Xenopus oocytes, Oat9S mediated the transport of L-carnitine (Km = 2.9 microM), a representative zwitterion, as well as cimetidine (Km = 16.1 microM) and salicylic acid (Km = 175.5 microM), while Oat9L did not show any transport activity. Oat9S-mediated L-carnitine uptake was inhibited by D-carnitine, acetylcarnitine, octanoylcarnitine, betaine, and other organic compounds, suggesting that quaternary ammonium cation bulkiness and relative hydrophobicity are important factors for Oat9S-substrate interactions. Among OATs, Oat9S appears to be the first member to mediate the transport of carnitine and possesses eight TMD. Overall, these new results provide added insight into the structure-activity relationship comprising the organic ion-permeation pathway.
Notes:
Masahiko Kaji, Maryam Kabir-Salmani, Naohiko Anzai, Chun Ji Jin, Yoshihiro Akimoto, Ayako Horita, Atsuhiko Sakamoto, Yoshikatsu Kanai, Hiroyuki Sakurai, Mitsutoshi Iwashita (2010)  Properties of L-type amino acid transporter 1 in epidermal ovarian cancer.   Int J Gynecol Cancer 20: 3. 329-336 Apr  
Abstract: HYPOTHESIS: To investigate the expression and the functional properties of L-type amino acid transporter 1 (LAT1) in human epithelial ovarian cancer to provide a basis for potential new therapies to control the growth and the metastasis of ovarian cancer. METHODS: The material used comprised 63 surgically resected specimens obtained from female patients undergoing gynecologic surgery at Kyorin University School of Medicine (Tokyo, Japan). The expression of LAT1 in 53 cases of ovarian cancers was determined by Western blot and immunohistochemical staining, and results were compared with those of normal ovarian tissues (5 cases) and benign ovarian tumors (5 cases). Furthermore, we examined the effect of 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH), the classic inhibitor of system L on the survival, the migration, and the uptake of l-leucine by human epithelial ovarian cancer cell line (OVCAR-3). RESULTS: The LAT1 was significantly up-regulated in various human epithelial ovarian cancers that was localized predominantly on their plasma membrane and in the plasma membrane of the ovarian cancer cell line in conjunction with 4F2hc via disulfide bonds. The BCH inhibited the proliferation and the migration of the OVCAR-3 cells and the uptake of [14C]l-leucine by these cells in a dose-dependent manner. The OVCAR-3 cells did not express LAT2, and the uptake of [14C]l-leucine by these cells was Na-independent and almost completely inhibited by BCH. Thus, our findings indicated that most l-leucine uptake in OVCAR-3 cells was mediated by LAT1. CONCLUSIONS: The LAT1 plays significant roles in nutrition, proliferation, and migration of ovarian cancer. Then, LAT1 inhibition would be useful for anticancer therapy in suppressing tumor growth without affecting normal tissues.
Notes:
Naohiko Anzai, Promsuk Jutabha, Hitoshi Endou (2010)  Renal Solute Transporters and Their Relevance to Serum Urate Disorder   Current Hypertension Reviews 6: 3. 148-154 Aug.  
Abstract: Since uric acid (urate), the final product of purine metabolism, exhibits antioxidative activity, its protective role against oxidative stress becomes attractive. Low serum urate levels have been associated with multiple sclerosis, Parkinson’s disease, and Alzheimer’s disease. Despite its beneficial role, hyperuricemia is associated with gout, hypertension, cardiovascular diseases such as myocardial infarction and stroke, and renal diseases such as acute urate nephropathy and nephrolithiasis. The urate transport system of the kidney is an important determinant of the serum urate level, but clarification of its molecular mechanism remains incomplete. In 2002, our group identified URAT1 (SLC22A12), a renal apical urate/anion exchanger, leading to the accumulation of information concerning individual molecules involved in urate transport in the kidney. In 2008, we functionally characterized facilitatory glucose transporter family member GLUT9 (SLC2A9) as a voltage-driven urate transporter URATv1 and analysis of a renal hypouricemia patient with a genetic defect in SLC2A9 have established the main route of the urate reabsorption pathway at the basolateral side of renal proximal tubules, where urate in the urinary lumen is taken up via apical URAT1 and intracellular urate exits from the cell to the interstitium/blood space via basolateral URATv1. In this review, recent findings concerning these molecules are presented.
Notes:
Masanobu Sato, Hideaki Mamada, Naohiko Anzai, Yoshiyuki Shirasaka, Takeo Nakanishi, Ikumi Tamai (2010)  Renal secretion of uric acid by organic anion transporter 2 (OAT2/SLC22A7) in human.   Biol Pharm Bull 33: 3. 498-503  
Abstract: The physiological function of organic anion transporter OAT2 (SLC22A7) remains unclear, but since OAT2 transports purine derivatives, it may be involved in renal handling of uric acid, the final metabolite of purine derivatives. In the present study, we studied uric acid transport in stably OAT2-expressing HEK293 cells (HEK293/OAT2). OAT2 mediated uptake, but not efflux, of [(14)C]uric acid. Uric acid transport was saturable with K(m) of 1168+/-335 muM (mean and S.E.M.) and V(max) of 2.57+/-0.350 nmol/min/mg protein. The [(14)C]uric acid uptake was sensitive to Cl(-) and was enhanced at acidic pH. In cis-inhibition assay, [(14)C]uric acid uptake was inhibited by several mono- or dicarboxylic acids, but it was not trans-stimulated by any of the compounds tested. The pattern of inhibition of OAT2-mediated uric acid transport by various drugs was different from that of OAT1- or OAT3-mediated transport. Furthermore, OAT2-mediated transport of uric acid was inhibited by an antiuricosuric drug, pyrazinecarboxylic acid. These results revealed distinct characteristics of uric acid transport via OAT2 compared with other uric acid transporters, suggesting that OAT2 plays a role in renal uric acid uptake from blood as a first step of tubular secretion. OAT2 may therefore be a potential target for regulating serum uric acid level.
Notes:
Koji Oda, Noriko Hosoda, Hiroshi Endo, Kunio Saito, Kenji Tsujihara, Michio Yamamura, Takeshi Sakata, Naohiko Anzai, Michael F Wempe, Yoshikatsu Kanai, Hitoshi Endou (2010)  L-type amino acid transporter 1 inhibitors inhibit tumor cell growth.   Cancer Sci 101: 1. 173-179 Jan  
Abstract: Most tumor cell membranes overexpress L-type amino acid transporter 1, while normal cell membranes contain l-type amino acid transporter 2; both are Na(+)-independent amino acid transporters. Therefore, compounds that selectively inhibit L-type amino acid transporter 1 offer researchers with a novel cancer molecular target. Synthetic chemistry efforts and in vitro screening have produced a variety of novel compounds possessing high in vitrol-type amino acid transporter 1 selectivity; KYT-0353 was one such compound. The present studies illustrate that KYT-0353 inhibited (14)C-leucine uptake and cell growth in human colon cancer-derived HT-29 cells; IC(50)s were 0.06 microm and 4.1 microm, respectively. KYT-0353 also inhibited (14)C-leucine uptake in mouse renal proximal tubule cells expressing l-type amino acid transporter 1, and inhibited cell growth; IC(50)s were 0.14 microm and 16.4 microm, respectively. Compared to control animals, intravenously administered KYT-0353 (12.5 mg/kg and 25.0 mg/kg) showed statistically significant growth inhibition against HT-29 tumors transplanted to nude mice with maximal inhibition ratios of 65.9% and 77.2%, respectively. Body weight increase with time--a safety indicator--was slightly depressed at 12.5 mg/kg and 25.0 mg/kg with maximal ratios of 3.7% (day 2) and 6.3% (day 11), respectively. Thus, KYT-0353 showed significant growth inhibitory effects on HT-29 cells both in vitro and in vivo, whereas it only caused a slight body weight depression. Therefore, KYT-0353 appears to have potential as a novel anti-tumor agent, presumably via selective in vivol-type amino acid transporter 1 inhibition.
Notes:
Wako Urano, Atsuo Taniguchi, Naohiko Anzai, Eisuke Inoue, Yoshikatsu Kanai, Mariko Yamanaka, Hitoshi Endou, Naoyuki Kamatani, Hisashi Yamanaka (2010)  Sodium-dependent phosphate cotransporter type 1 sequence polymorphisms in male patients with gout.   Ann Rheum Dis 69: 6. 1232-1234 Jun  
Abstract: OBJECTIVES: Molecular biological approaches have recently identified urate transporters in renal proximal tubular cells. Human sodium-dependent phosphate cotransporter type 1 encoded by SLC17A1 is a urate transporter localised to the renal proximal tubular cells and candidate molecule to secret urate from renal tubular cells to urine. This study investigated the roles of SLC17A1 in the development of gout. PATIENTS AND METHODS: Single nucleotide polymorphisms in the human SLC17A1 gene (rs1165176, rs1165151, rs1165153, rs1165196, rs1165209, rs1165215, rs1179086, rs3799344 and rs3757131) were selected, and an association study was conducted using male patients with gout (n=175) and male controls (n=595). RESULTS: There were significant differences between gout and control groups in the distribution of genotypes at rs1165196 (T806C; Ile269Thr, odds ratio (OR) 0.55, p=0.0035), rs1179086 (OR 0.57, p=0.0018) and rs3757131 (OR 0.54, p=0.0026). In controls, T806C alone had no effect on serum uric acid (sUA) levels. However, T806C showed significant interaction with a reduction of sUA in obese individuals (body mass index > or = 25) using multiple regression analysis. CONCLUSIONS: Our data suggest that SLC17A1 polymorphisms are associated with the development of gout.
Notes:
Promsuk Jutabha, Michael F Wempe, Naohiko Anzai, Jun Otomo, Toshihito Kadota, Hitoshi Endou (2010)  Xenopus laevis oocytes expressing human P-glycoprotein: probing trans- and cis-inhibitory effects on [3H]vinblastine and [3H]digoxin efflux.   Pharmacol Res 61: 1. 76-84 Jan  
Abstract: P-glycoprotein (P-gp; MDR1) recognizes and actively transports many structurally diverse compounds (hydrophobic neutral and cationic). We studied MDR1-mediated drug transport using a high-throughput (96-well) oocyte expression system. MDR1-expressing oocytes contained sufficient ATP levels to conduct fundamental efflux studies; the optimal experimental temperature was 25 degrees C. [(3)H]Vinblastine efflux by MDR1-expressing oocytes was detectable and afforded a K(m) of 145.5+/-25.4microM. [(3)H]Vinblastine (5.6+/-0.3microM) and [(3)H]digoxin (1.0+/-0.1microM) were individually injected into MDR1-expressing oocytes and their efflux monitored. Quinidine and verapamil, known MDR1 substrates/inhibitors, showed trans-inhibition on MDR1-mediated [(3)H]vinblastine and [(3)H]digoxin efflux. Conversely, doxorubicin demonstrated cis-inhibition without trans-inhibition on MDR1-mediated [(3)H]vinblastine efflux. The MDR1-expressing oocyte system offers researchers with an alternative in vitro method to screen compounds and may allow one to probe P-gp drug-drug and/or drug-inhibitor interactions.
Notes:
Naohiko Anzai, Promsuk Jutabha, Hitoshi Endou (2010)  Molecular Mechanism of Ochratoxin A Transport in the Kidney   Toxins 2: 6. 1381-1398 June  
Abstract: The mycotoxin, ochratoxin A (OTA), is thought to be responsible for Balkan endemic nephropathy. OTA accumulates in several tissues, especially in the kidneys and liver. The excretion of OTA into urine is thought to be mainly by tubular secretion, presumably via the organic anion transport system. Recently, several families of multispecific organic anion transporters have been identified: organic anion transporters (OATs), organic anion-transporting polypeptides (OATPs), oligopeptide transporters (PEPTs), and ATP-binding cassette (ABC) transporters, such as MRP2 and BCRP. These renal transporters mediate the transmembrane transport of OTA and play a pivotal role in the development of OTA-induced nephrotoxicity.
Notes:
Promsuk Jutabha, Naohiko Anzai, Kenichiro Kitamura, Atsuo Taniguchi, Shuji Kaneko, Kunimasa Yan, Hideomi Yamada, Hidetaka Shimada, Toru Kimura, Tomohisa Katada, Toshiyuki Fukutomi, Kimio Tomita, Wako Urano, Hisashi Yamanaka, George Seki, Toshiro Fujita, Yoshinori Moriyama, Akira Yamada, Shunya Uchida, Michael F Wempe, Hitoshi Endou, Hiroyuki Sakurai (2010)  Human sodium-phosphate transporter 4 (hNPT4/SLC17A3) as a common renal secretory pathway for drugs and urate.   J Biol Chem Sep  
Abstract: The evolutionary loss of hepatic urate oxidase (uricase) has resulted in humans with elevated serum uric acid (urate). Uricase loss may have been beneficial to early primate survival. However, elevated serum urate has predisposed man to hyperuricemia; a metabolic disturbance leading to gout, hypertension, and various cardiovascular diseases. Human serum urate levels are largely determined by urate reabsorption and secretion in the kidney. Renal urate reabsorption is controlled via two proximal tubular urate transporters: apical URAT1 (SLC22A12) and basolateral URATv1/GLUT9 (SLC2A9). In contrast, molecular mechanism(s) for renal urate secretion remain unknown. In this report, we demonstrate that an orphan transporter hNPT4 (human sodium-phosphate transporter 4; SLC17A3) was a multispecific organic anion efflux transporter expressed in the kidneys and liver. hNPT4 was localized at the apical side of renal tubules and functioned as a voltage-driven urate transporter. Furthermore, loop diuretics, such as furosemide and bumetanide, substantially interacted with hNPT4. Thus, this protein is likely to act as a common secretion route for both drugs and may play an important role in diuretics-induced hyperuricemia. In vivo role of hNPT4 was suggested by two hyperuricemia patients with missense mutations in SLC17A3. These mutated hNPT4 exhibited reduced urate efflux when they were expressed in Xenopus oocytes. Our findings will complete a model of urate secretion in the renal tubular cell, where intracellular urate taken up via OAT1 and/or OAT3 from the blood, exits from the cell into the lumen via hNPT4.
Notes:
Masafumi Iharada, Takaaki Miyaji, Takahiro Fujimoto, Miki Hiasa, Naohiko Anzai, Hiroshi Omote, Yoshinori Moriyama (2010)  Type 1 sodium-dependent phosphate transporter (SLC17A1 Protein) is a Cl(-)-dependent urate exporter.   J Biol Chem 285: 34. 26107-26113 Aug  
Abstract: SLC17A1 protein (NPT1) is the first identified member of the SLC17 phosphate transporter family and mediates the transmembrane cotransport of Na(+)/P(i) in oocytes. Although this protein is believed to be a renal polyspecific anion exporter, its transport properties are not well characterized. Here, we show that proteoliposomes containing purified SLC17A1 transport various organic anions such as p-aminohippuric acid and acetylsalicylic acid (aspirin) in an inside positive membrane potential (Deltapsi)-dependent manner. We found that NPT1 also transported urate. The uptake characteristics were similar to that of SLC17 members in its Cl(-) dependence and inhibitor sensitivity. When arginine 138, an essential amino acid residue for members of the SLC17 family such as the vesicular glutamate transporter, was specifically mutated to alanine, the resulting mutant protein was inactive in Deltapsi-dependent anion transport. Heterologously expressed and purified human NPT1 carrying the single nucleotide polymorphism mutation that is associated with increased risk of gout in humans exhibited 32% lower urate transport activity compared with the wild type protein. These results strongly suggested that NPT1 is a Cl(-)-dependent polyspecific anion exporter involved in urate excretion under physiological conditions.
Notes:
Anna D'Amico, Andrea Soragna, Eliana Di Cairano, Nicola Panzeri, Nahoiko Anzai, Franca Vellea Sacchi, Carla Perego (2010)  The surface density of the glutamate transporter EAAC1 is controlled by interactions with PDZK1 and AP2 adaptor complexes.   Traffic Aug  
Abstract: The glutamate transporter EAAC1/EAAT3 mediates the absorption of dicarboxylic amino acids in epithelial cells as well as the uptake of glutamate from the synaptic cleft. Its cell-surface density is regulated by interaction with accessory proteins which remain to be identified. We detected a consensus sequence for interaction with PDZ proteins (-SQF) and a tyrosine-based internalisation signal (-YVNG-) in the C-terminus of EAAC1, and investigated their role in the transporter localisation. We demonstrated that PDZ interactions are required for the efficient delivery to and the retention in the plasma membrane of EAAC1 and we identified PDZK1/NHERF3 as a novel EAAC1 interacting protein. Expression of PDZK1 in MDCK cells tethered EAAC1 to filopodia and increased its surface activity. Removal of PDZ-target motif promoted the EAAC1 binding to alpha-adaptin and clathrin and the transporter internalisation in endocytic/degradative compartments. This defect was largely prevented by hypertonic treatment or overexpression of the dominant-negative mu2-W421A-subunit of AP-2 clathrin-adaptor. The rate of transporter endocytosis was attenuated following tyrosine mutagenesis in the internalisation signal, thus indicating that this motif can regulate the transporter endocytosis. We suggest that EAAC1 density is controlled by balanced interactions with PDZK1 and AP2: the former promotes the transporter expression at the cell surface, and the latter mediates its constitutive endocytosis.
Notes:
Makiko Nakamura, Naohiko Anzai, Promsuk Jutabha, Hiroyuki Sato, Hiroyuki Sakurai, Kimiyoshi Ichida (2010)  Concentration-Dependent Inhibitory Effect of Irbesartan on Renal Uric Acid Transporters.   J Pharmacol Sci Aug  
Abstract: Hyperuricemia is currently recognized as a risk factor for cardiovascular diseases. It has been reported that the angiotensin II-receptor blocker (ARB) losartan decreases serum uric acid level. In this study, the effects of another ARB, irbesartan, on [(14)C]uric acid-transport activity of renal uric acid reabsorptive transporters URAT1 and URATv1 were examined with Xenopus oocytes expressing each transporter. The results showed that irbesartan (100 - 500 microM) inhibited the uptake of uric acid via both transporters. The inhibitory effects of irbesartan exceeded those of losartan and other ARBs, and the results suggest that irbesartan can reduce serum uric acid level.
Notes:
Takeshi Sakata, Naohiko Anzai, Toru Kimura, Daisaku Miura, Toshiyuki Fukutomi, Michio Takeda, Hiroyuki Sakurai, Hitoshi Endou (2010)  Functional analysis of human organic cation transporter OCT3 (SLC22A3) polymorphisms.   J Pharmacol Sci 113: 3. 263-266 06  
Abstract: We analyzed the functional properties of five single nucleotide polymorphisms (SNPs) in organic cation transporter OCT3 gene (SLC22A3) resulting in the amino acid changes with a transient expression system. Three SNPs (A116S, T400I, and A439V) exhibited reduced uptake of both [(3)H]histamine and [(3)H]MPP(+), although their protein expressions were detected in the plasma membrane of transfected cells. This study suggests that the OCT3 variants will contribute to inter-individual variations leading to the differences in cationic drug disposition as well as certain disease processes such as hypertension, allergic diseases, and neuropsychiatric diseases by the clearance of endogenous organic cations such as biogenic amines.
Notes:
Naoko Ohtsu, Naohiko Anzai, Toshiyuki Fukutomi, Toru Kimura, Hiroyuki Sakurai, Hitoshi Endou (2010)  Human renal urate transpoter URAT1 mediates the transport of salicylate   Nippon Jinzo Gakkai Shi 52: 4. 499-504  
Abstract: Salicylic acid derivatives are the most prescribed analgesic-antipyretic and anti-inflammatory agents. It is well known that salicylate has a paradoxical effect on renal urate excretion. At low doses (5 - 10 mg/dL serum), renal urate excretion is decreased, whereas at high doses (> 15 mg/dL serum), renal urate excretion is increased. Since the molecular identification of the renal apical urate/anion exchanger URAT1, it has been suggested that this protein is responsible for the paradoxical effect because of cis-inhibition of salicylate (1 mM) on urate uptake by URAT1-expressing oocytes. The purpose of this study was to examine whether or not URAT1 is responsible for the paradoxical effect of salicylate. In URAT1-stably expressing HEK293 (HEK293-URAT1) cells, salicylate inhibited [14C] urate uptake dose-dependently (IC50, 23.9 microM). URAT1 mediated the time-dependent uptake of [3H] salicylate in these cells. [3H] Salicylate uptake via URAT1 was inhibited by non-labelled urate and salicylate, and the uricosuric agent, benzbromarone. In the URAT1-expressing oocytes, we observed the time- and concentration-dependent transport of salicylate (Km : 25.3 microM). Moreover, non-labelled salicylate injected into the URATI-expressing oocytes stimulated [14C] urate uptake. These results suggest that the "paradoxical effect" of salicylate can be explained by two modes of salicylate interaction with URAT1 : (1) acting as an exchange substrate to facilitate urate reabsorption, and (2) acting as an inhibitor for urate reabsorption.
Notes:
Katsuko Shiraya, Taku Hirata, Ryo Hatano, Shushi Nagamori, Pattama Wiriyasermkul, Promsuk Jutabha, Mitsunobu Matsubara, Shigeaki Muto, Hidekazu Tanaka, Shinji Asano, Naohiko Anzai, Hitoshi Endou, Akira Yamada, Hiroyuki Sakurai, Yoshikatsu Kanai (2010)  A novel transporter of SLC22 family specifically transports prostaglandins and co-localizes with 15-hydroxyprostaglandin dehydrogenase in renal proximal tubules.   J Biol Chem 285: 29. 22141-22151 Jul  
Abstract: We identified a novel prostaglandin (PG)-specific organic anion transporter (OAT) in the OAT group of the SLC22 family. The transporter designated OAT-PG from mouse kidney exhibited Na(+)-independent and saturable transport of PGE(2) when expressed in a proximal tubule cell line (S(2)). Unusual for OAT members, OAT-PG showed narrow substrate selectivity and high affinity for a specific subset of PGs, including PGE(2), PGF(2alpha), and PGD(2). Similar to PGE(2) receptor and PGT, a structurally distinct PG transporter, OAT-PG requires for its substrates an alpha-carboxyl group, with a double bond between C13 and C14 as well as a (S)-hydroxyl group at C15. Unlike the PGE(2) receptor, however, the hydroxyl group at C11 in a cyclopentane ring is not essential for OAT-PG substrates. Addition of a hydroxyl group at C19 or C20 impairs the interaction with OAT-PG, whereas an ethyl group at C20 enhances the interaction, suggesting the importance of hydrophobicity around the omega-tail tip forming a "hydrophobic core" accompanied by a negative charge, which is essential for substrates of OAT members. OAT-PG-mediated transport is concentrative in nature, although OAT-PG mediates both facilitative and exchange transport. OAT-PG is kidney-specific and localized on the basolateral membrane of proximal tubules where a PG-inactivating enzyme, 15-hydroxyprostaglandin dehydrogenase, is expressed. Because of the fact that 15-keto-PGE(2), the metabolite of PGE(2) produced by 15-hydroxyprostaglandin dehydrogenase, is not a substrate of OAT-PG, the transport-metabolism coupling would make unidirectional PGE(2) transport more efficient. By removing extracellular PGE(2), OAT-PG is proposed to be involved in the local PGE(2) clearance and metabolism for the inactivation of PG signals in the kidney cortex.
Notes:
Ellappan Babu, Michio Takeda, Ritsuo Nishida, Rie Noshiro-Kofuji, Masaaki Yoshida, Shiro Ueda, Toshiyuki Fukutomi, Naohiko Anzai, Hitoshi Endou (2010)  Interactions of human organic anion transporters with aristolochic acids.   J Pharmacol Sci 113: 2. 192-196 Jun  
Abstract: Aristolochic acids (AAs), contained in Chinese herbal preparations, have been considered to induce nephropathy. In order to elucidate the molecular mechanisms of AA-induced nephrotoxicity, we have elucidated the interaction of human organic anion transporters (hOATs) with AAs using their stable cell lines. AA-I and AA-II inhibited organic anion uptake by hOAT1, hOAT3, and hOAT4 in dose-dependent manners. Treatment of hOAT3 with AA-I resulted in a significant reduction in viability compared with that of mock, which was rescued by the organic anion transport inhibitor probenecid. In conclusion, hOAT3-mediated AA-I uptake may be associated with the induction of nephrotoxicity.
Notes:
2009
Satoko Minami, Kousei Ito, Masashi Honma, Yuki Ikebuchi, Naohiko Anzai, Yoshikatsu Kanai, Tamotsu Nishida, Sachiko Tsukita, Shuichi Sekine, Toshiharu Horie, Hiroshi Suzuki (2009)  Posttranslational regulation of Abcc2 expression by SUMOylation system.   Am J Physiol Gastrointest Liver Physiol 296: 2. G406-G413 Feb  
Abstract: The ATP-binding cassette transporter family C 2 (Abcc2) is a member of efflux transporters involved in the biliary excretion of organic anions from hepatocytes. Posttranslational regulation of Abcc2 has been implicated, although the molecular mechanism is not fully understood. In the present study, we performed yeast two-hybrid screening to identify novel protein(s) that particularly interacts with the linker region of Abcc2 located between the NH(2)-terminal nucleotide binding domain and the last membrane-spanning domain. The screening resulted in the identification of a series of small ubiquitin-like modifier (SUMO)-related enzymes and their substrates. In yeast experiments, all of these interactions were abolished by substituting the putative SUMO consensus site in the linker region (IKKE) in Abcc2 to IRKE. In vitro SUMOylation experiments confirmed that the Abcc2 linker was a substrate of Ubc9-mediated SUMOylation. It was also found that the IKKE sequence is the target of SUMOylation, since a mutant with IKKE is substituted by IRKE was not SUMOylated. Furthermore, we demonstrated for the first time that Abcc2, endogenously expressed in rat hepatoma-derived McARH7777 cells, is SUMOylated. Suppression of endogenous Ubc9 by small interfering RNA resulted in a selective 30% reduction in Abcc2 protein expression in the postnuclear supernatant, whereas subcellular localization of Abcc2 confirmed by semiquantitative immunofluorescence analysis was minimally affected. This is the first demonstration showing the regulation of ABC transporter expression by SUMOylation.
Notes:
Gisela Di Giusto, Naohiko Anzai, Hitoshi Endou, Adriana M Torres (2009)  Oat5 and NaDC1 protein abundance in kidney and urine after renal ischemic reperfusion injury.   J Histochem Cytochem 57: 1. 17-27 Jan  
Abstract: The aim of this study was to evaluate the abundance of the organic anion transporter 5 (Oat5) and the sodium-dicarboxylate cotransporter 1 (NaDC1) in kidney and urine after renal ischemic reperfusion injury. Renal injury was induced in male Wistar rats by occlusion of both renal pedicles for 0 (Group Sham), 5 (Group I5R60), or 60 (Group I60R60) min. The studies were performed after 60 min of reperfusion. The expression of Oat5 and NaDC1 was evaluated by IHC and Western blotting. Oat5 and NaDC1 abundance and alkaline phosphatase activity (AP) were assayed in urine. A decreased expression in renal homogenates and apical membranes and an increase in urinary excretion of Oat5 and NaDC1 were observed in I60R60 rats, as well as alterations of other widely used parameters for renal dysfunction and injury (plasma creatinine, urinary AP activity, kidney weight, histological lesions). In contrast, in the I5R60 group, only an increase in urinary excretion of Oat5 and mild histopathological damage was detected. This is the first study on Oat5 and NaDC1 detection in urine. These results suggest that urinary excretion of Oat5 might be an early indicator of renal dysfunction, which is useful for detection of even minor alterations in renal structural and functional integrity.
Notes:
Gisela Di Giusto, Naohiko Anzai, María L Ruiz, Hitoshi Endou, Adriana M Torres (2009)  Expression and function of Oat1 and Oat3 in rat kidney exposed to mercuric chloride.   Arch Toxicol 83: 10. 887-897 Oct  
Abstract: This study was designed to evaluate the expression and function of the organic anion transporters, Oat1 and Oat3, in rats exposed to a nephrotoxic dose of HgCl(2). Oat1 protein expression increased in renal homogenates and decreased in renal basolateral membranes from HgCl(2) rats, while Oat3 protein abundance decreased in both kidney homogenates and basolateral membranes. The lower protein levels of Oat1 and Oat3 in basolateral membranes explain the lower uptake capacity for p-aminohippurate (in vitro assays) and the diminution of the systemic clearance of this organic anion (in vivo studies) observed in treated rats. Since both transporters mediate mercury access to the renal cells, their down-regulation in basolateral membranes might be a defensive mechanism developed by the cell to protect itself against mercury injury. The pharmacological modulation of the expression and/or the function of Oat1 and Oat3 might be an effective therapeutic strategy for reducing the nephrotoxicity of mercury.
Notes:
Hrvoje Brzica, Davorka Breljak, Marija Ljubojević, Daniela Balen, Vedran Micek, Naohiko Anzai, Ivan Sabolić (2009)  Optimal methods of antigen retrieval for organic anion transporters in cryosections of the rat kidney.   Arh Hig Rada Toksikol 60: 1. 7-17 Mar  
Abstract: To localise antigens by immunocytochemistry (IC), the samples of tissues or cells are usually denatured by fixation, and either frozen and cryosectioned, or embedded in paraffin before sectioning. p-Formaldehyde (PFA; formalin) is a common fixative, which preserves antigenicity of proteins, but damages the tissue/cell morphology and "masks" the antibody binding sites (epitopes). In order to "unmask" epitopes, some kind of antigen retrieval (AR) is used. The aim of this study was: a) to find an optimal AR method in cryosections of in vivo PFA-fixed kidneys for organic anion transporters (Oat) that reside in the basolateral (Oat1, Oat3) and brush-border membrane (Oat2, Oat5) of the rat renal proximal tubules, and b) using optimal method, to compare IC staining of Oats in kidneys that had been PFA-fixed in vivo or in vitro. IC staining in untreated cryosections was compared with that following detergent treatment or microwave heating in citrate buffer of pH 3, pH 6, or pH 8, with or without alcohol pre-treatment. The preferred AR method for Oat1, Oat2, and Oat5 was heating of cryosections at pH 6, and for Oat3 heating at pH 3, without alcohol pre-treatment. Compared with tissue fixed in vivo, tissue fixed in vitro exhibited damaged tubule morphology, similar staining intensity of Oat1 and Oat3, and higher staining intensity of Oat2 and Oat5. We conclude that for optimal IC presentation, each Oat in the rat kidney has to be treated individually, with different fixation and AR approach.
Notes:
Vilim Zlender, Davorka Breljak, Marija Ljubojević, Dubravka Flajs, Daniela Balen, Hrvoje Brzica, Ana-Marija Domijan, Maja Peraica, Radovan Fuchs, Naohiko Anzai, Ivan Sabolić (2009)  Low doses of ochratoxin A upregulate the protein expression of organic anion transporters Oat1, Oat2, Oat3 and Oat5 in rat kidney cortex.   Toxicol Appl Pharmacol 239: 3. 284-296 Sep  
Abstract: Mycotoxin ochratoxin A (OTA) is nephrotoxic in various animal species. In rodents, OTA intoxication impairs various proximal tubule (PT) functions, including secretion of p-aminohippurate (PAH), possibly via affecting the renal organic anion (OA) transporters (Oat). However, an effect of OTA on the activity/expression of specific Oats in the mammalian kidney has not been reported. In this work, male rats were gavaged various doses of OTA every 2nd day for 10 days, and in their kidneys we studied: tubule integrity by microscopy, abundance of basolateral (rOat1, rOat3) and brush-border (rOat2, rOat5) rOat proteins by immunochemical methods, and expression of rOats mRNA by RT-PCR. The OTA treatment caused: a) dose-dependent damage of the cells in S3 segments of medullary rays, b) dual effect upon rOats in PT: low doses (50-250 microg OTA/kg b.m.) upregulated the abundance of all rOats, while a high dose (500 microg OTA/kg b.m.) downregulated the abundance of rOat1, and c) unchanged mRNA expression for all rOats at low OTA doses, and its downregulation at high OTA dose. Changes in the expression of renal Oats were associated with enhanced OTA accumulation in tissue and excretion in urine, whereas the indicators of oxidative stress either remained unchanged (malondialdehyde, glutathione, 8-hydroxydeoxyguanosine) or became deranged (microtubules). While OTA accumulation and downregulation of rOats in the kidney are consistent with the previously reported impaired renal PAH secretion in rodents intoxicated with high OTA doses, the post-transcriptional upregulation of Oats at low OTA doses may contribute to OTA accumulation and development of nephrotoxicity.
Notes:
2008
Hirokazu Yokoyama, Naohiko Anzai, Marija Ljubojevic, Naoko Ohtsu, Takeshi Sakata, Hiroki Miyazaki, Hiroshi Nonoguchi, Rafiqul Islam, Maristella Onozato, Akihiro Tojo, Kimio Tomita, Yoshikatsu Kanai, Takashi Igarashi, Ivan Sabolic, Hitoshi Endou (2008)  Functional and immunochemical characterization of a novel organic anion transporter Oat8 (Slc22a9) in rat renal collecting duct.   Cell Physiol Biochem 21: 4. 269-278 04  
Abstract: In this study, we demonstrate that a putative membrane unknown solute transporter 1 of the rat kidney (UST1r; Slc22a9) is a multispecific transporter of organic anions (OAs). When expressed in Xenopus oocytes, UST1r mediated uptake of ochratoxin A (OTA; K(m) = 1.0 microM) and sulfate conjugates of steroids, such as estrone-3-sulfate (ES; K(m) = 3.1 microM) and dehydroepiandrosterone sulfate (DHEAS; K(m) = 2.1 microM) in a sodium-independent manner. We herein propose that UST1r be renamed OA transporter 8 (rOat8). rOat8 interacted with chemically heterogenous anionic compounds, such as nonsteroidal anti-inflammatory drugs, diuretics, probenecid, taurocholate, and methotrexate, but not with the organic cation tetraethylammonium. The rOat8-mediated ES transport was: a) cis-inhibited by 4-methylumbelliferyl sulfate and beta-estradiol sulfate, but not by glucuronide conjugates of these compounds, b) cis-inhibited by four- and five- carbon (C4/C5) dicarboxylates (succinate and glutarate (GA)), and c) trans-stimulated by GA, whereas the efflux of GA was significantly trans-stimulated by ES. By RT-PCR, rOat8 mRNA was expressed in proximal convoluted tubules and cortical and outer medullary collecting ducts, whereas in immunochemical studies, Oat8 was identified as the ñ58 kDa protein that in the collecting duct colocalized with the V-ATPase in plasma membranes and intracellular vesicles in various subtypes of intercalated cells. Molecular identification of Oat8 in these cells indicates a possible novel role of OAT family in the renal secretion/reabsorption of OA and acids and bases via affecting the V-ATPase-dependent functions.
Notes:
Hatsuki Nakagawa, Taku Hirata, Tomohiro Terada, Promsuk Jutabha, Daisaku Miura, Kouji H Harada, Kayoko Inoue, Naohiko Anzai, Hitoshi Endou, Ken-Ichi Inui, Yoshikatsu Kanai, Akio Koizumi (2008)  Roles of organic anion transporters in the renal excretion of perfluorooctanoic acid.   Basic Clin Pharmacol Toxicol 103: 1. 1-8 Jul  
Abstract: Perfluorooctanoic acid, an environmental contaminant, is found in both wild animals and human beings. There are large species and sex differences in the renal excretion of perfluorooctanoic acid. In the present study, we aimed to characterize organic anion transporters 1-3 (OAT1-3) in human beings and rats to investigate whether the species differences in the elimination kinetics of perfluorooctanoic acid from the kidneys can be attributed to differences in the affinities of these transporters for perfluorooctanoic acid. We used human (h) and rat (r) OAT transient expression cell systems and measured the [(14)C] perfluorooctanoic acid transport activities. Both human and rat OAT1 and OAT3 mediated perfluorooctanoic acid transport to similar degrees. Specifically, the kinetic parameters, K(m), were 48.0 +/- 6.4 microM for h OAT1; 51.0 +/- 12.0 microM for rOAT1; 49.1 +/- 21.4 microM for hOAT3 and 80.2 +/- 17.8 microM for rOAT3, respectively. These data indicate that both human and rat OAT1 and OAT3 have high affinities for perfluorooctanoic acid and that the species differences in its renal elimination are not attributable to affinity differences in these OATs between human beings and rats. In contrast, neither hOAT2 nor rOAT2 transported perfluorooctanoic acid. In conclusion, OAT1 and OAT3 mediated perfluorooctanoic acid transport in vitro, suggesting that these transporters also transport perfluorooctanoic acid through the basolateral membrane of proximal tubular cells in vivo in both human beings and rats. Neither human nor rat OAT2 mediated perfluorooctanoic acid transport. Collectively, the difference between the perfluorooctanoic acid half-lives in human beings and rats is not likely to be attributable to differences in the affinities of these transporters for perfluorooctanoic acid.
Notes:
Kittipong Tachampa, Michio Takeda, Suparat Khamdang, Rie Noshiro-Kofuji, Minoru Tsuda, Surawat Jariyawat, Toshiyuki Fukutomi, Samaisukh Sophasan, Naohiko Anzai, Hitoshi Endou (2008)  Interactions of organic anion transporters and organic cation transporters with mycotoxins.   J Pharmacol Sci 106: 3. 435-443 Mar  
Abstract: Mycotoxins are secondary metabolites of moulds that which exert adverse effects in humans and animals. It is known that direct cellular toxicity is often associated with increased cellular accumulation of toxic compounds, and membrane transport may be the first fundamental stage in the development of the cytotoxicity. To elucidate the entry pathway for mycotoxins into cells, we have investigated the interactions of human and rat organic anion transporters (hOATs/rOats) and human organic cation transporters (hOCTs) with mycotoxins using cells stably expressing hOATs/rOats/hOCTs. The mycotoxins tested were aflatoxin B1, alpha-zearalenol, citrinin, citrioveridine, cyclopiazonic acid, fumonisin B1, gliotoxin, patulin, penicillic acid, rubratoxin B, and zearalenone. These mycotoxins inhibited organic anion uptake mediated by hOAT1-4, and organic cation uptake mediated by hOCT1-2. By comparing the IC(50) values of mycotoxins for hOATs, it was found that hOAT1 and hOAT3 exhibited higher affinity interactions with mycotoxins than hOAT2 and hOAT4. There was no interspecies difference between humans and rats for the interactions of OATs with mycotoxins except that of OAT3 with rubratoxin B. Finally, we observed that hOAT1-4 and hOCT1-2 mediated the uptake of aflatoxin B1. In conclusion, hOATs and hOCTs interacted with various mycotoxins. Considering the localization of hOATs/rOats and hOCTs, it was suggested that these transporters were the possible entrance pathway for mycotoxins in kidney and liver, leading to the induction of adverse effects in humans and rats.
Notes:
Gisela Di Giusto, Naohiko Anzai, Hitoshi Endou, Adriana M Torres (2008)  Elimination of organic anions in response to an early stage of renal ischemia-reperfusion in the rat: role of basolateral plasma membrane transporters and cortical renal blood flow.   Pharmacology 81: 2. 127-136 10  
Abstract: BACKGROUND/AIMS: The knowledge of molecular mechanisms determining drug pharmacokinetics in pathological states is relevant for the development of new therapeutic approaches. This study was undertaken to evaluate the cortical renal blood flow (cRBF) and the renal protein expression of the organic anion transporters (OAT1 and OAT3) in association with the elimination of organic anions in an early stage of renal ischemia-reperfusion. METHODS: Ischemic acute renal failure (ARF) was induced in adult male Wistar rats by occlusion of both renal pedicles during 60 min, followed by 60 min of reperfusion (ARF group). Pair-fed sham-operated rats served as controls. The renal protein expression of OAT1 and OAT3 was evaluated by immunohistochemistry techniques and by Western blotting in renal cortex homogenates and in basolateral plasma membranes. A pharmacokinetic study of p-aminohippurate (PAH, a prototypical organic anion) was performed. cRBF was determined using fluorescent microspheres. RESULTS: ARF rats displayed a significant decrease in systemic clearance and in renal excretion of PAH. OAT1 and OAT3 protein abundance showed a statistically significant reduction both in homogenates and in basolateral plasma membranes from ARF rats. Immunohistochemical studies confirmed the changes in the cortical renal expression of these transporters. ARF animals also showed a decrease in cRBF. CONCLUSIONS: The decrease in PAH elimination observed in an early stage of renal ischemia-reperfusion in male Wistar rats might be explained by the sum of the lower OAT1 and OAT3 expression in renal basolateral plasma membranes plus the decrease in cRBF. These findings might have significant implications in the development of novel pharmacological strategies to be applied in the initial stages of ischemic ARF.
Notes:
Hitoshi Endou, Naohiko Anzai (2008)  Urate transport across the apical membrane of renal proximal tubules.   Nucleosides Nucleotides Nucleic Acids 27: 6. 578-584 Jun  
Abstract: Since the molecular cloning of the renal apical urate/anion exchanger URAT1 (SLC22A12), several membrane proteins relevant to urate transport have been identified. In addition, the identification of PDZ (PSD-95, DglA, and ZO-1) domain protein PDZK1 as a binding partner of URAT1, and the emerging role of PDZ scaffold for renal apical transporters have led to a new concept of renal urate transport: urate-transporting multimolecular complex, or "urate transportsome," that may form an ultimate functional unit at the apical membrane of renal proximal tubules. Elucidation of urate transportsome will lead to the new drug development for hyperuricemia.
Notes:
Rafiqul Islam, Naohiko Anzai, Nesar Ahmed, Ellappan Babu, Chun Ji Jin, Sunena Srivastava, Daisaku Miura, Toshiyuki Fukutomi, Yoshikatsu Kanai, Hitoshi Endou (2008)  Mouse organic anion transporter 2 (mOat2) mediates the transport of short chain fatty acid propionate.   J Pharmacol Sci 106: 3. 525-528 Mar  
Abstract: In this study, we have elucidated that propionate, one of the short chain fatty acids (SCFAs), is the transport substrate for murine organic anion transporter 2 (mOat2), which is expressed in the kidneys and the liver. When expressed in Xenopus oocytes, mOat2-mediated [(3)H]PGE(2) transport was inhibited by three- to five-carbon SCFAs (C3 to C5). Among the SCFAs tested, propionate (3-carbon SCFA) was transported by mOat2 in a time-dependent manner. Since propionate is a potent glucogenic compound, Oat2 may be involved in the regulation of cellular metabolism through the transport of these metabolites in the kidneys and the liver.
Notes:
Keisuke Tachibana, Naohiko Anzai, Chihiro Ueda, Tatsuya Katayama, Daisuke Yamasaki, Takayoshi Kirino, Rika Takahashi, Kenji Ishimoto, Hidenori Komori, Toshiya Tanaka, Takao Hamakubo, Yukihiko Ueda, Hiroyuki Arai, Juro Sakai, Tatsuhiko Kodama, Takefumi Doi (2008)  Regulation of the human PDZK1 expression by peroxisome proliferator-activated receptor alpha.   FEBS Lett 582: 28. 3884-3888 Nov  
Abstract: Although PDZK1 is a well-known adaptor protein, the mechanisms for its role in transcriptional regulation are largely unknown. The peroxisome proliferator-activated receptor alpha (PPARalpha) is a ligand-activated transcription factor that plays an important role in the regulation of lipid homeostasis. Previously, we established a tetracycline-regulated human cell line that can be induced to express PPARalpha and identified candidate target genes, one of which was PDZK1. In this study, we cloned and characterized the promoter region of the human pdzk1 gene and determined the PPAR response element. Finally, we demonstrate that endogenous PPARalpha regulates PDZK1 expression.
Notes:
Emiko Morimoto, Yoshikatsu Kanai, Do Kyung Kim, Arthit Chairoungdua, Hye Won Choi, Michael F Wempe, Naohiko Anzai, Hitoshi Endou (2008)  Establishment and characterization of mammalian cell lines stably expressing human L-type amino acid transporters.   J Pharmacol Sci 108: 4. 505-516 Dec  
Abstract: System L (SL), a basolateral amino acid transporter, transports large neutral amino acids (LNAAs) in a Na(+)-independent manner. Previously, we identified two isoforms of transporters: L-type amino acid transporter 1 (LAT1) and 2 (LAT2) and revealed their distinct substrate selectivity and transport properties. In this study, to establish more stable human LAT1 (hLAT1) and LAT2 (hLAT2) in vitro assay systems, we established mouse cell lines stably expressing hLAT1 (S2-LAT1) and hLAT2 (S2-LAT2). Real-time quantitative RT-PCR analysis revealed that S2-LAT1 and S2-LAT2 cells express hLAT1 and hLAT2 mRNAs at 20 - 1000-fold higher levels than those of endogenous mouse Lat1 and Lat2. S2-LAT1 and S2-LAT2 mediated [(14)C]L-leucine transport properties were measured and corresponded to results observed via Xenopus oocytes. Using these cells, the data demonstrate that hLAT1 and hLAT2 exhibit different characters in the acceptance of alpha-methyl amino acids and amino acid-related compounds with bulky side chains such as thyroid hormones and melphalan. S2-LAT1 and S2-LAT2 cells are expected to facilitate hLAT1 and hLAT2 substrate recognition research and contribute to drug development by providing an efficient assay system to screen for chemical compounds that interact with hLAT1 and hLAT2.
Notes:
Naohiko Anzai, Kimiyoshi Ichida, Promsuk Jutabha, Toru Kimura, Ellappan Babu, Chun Ji Jin, Sunena Srivastava, Kenichiro Kitamura, Ichiro Hisatome, Hitoshi Endou, Hiroyuki Sakurai (2008)  Plasma urate level is directly regulated by a voltage-driven urate efflux transporter URATv1 (SLC2A9) in humans.   J Biol Chem 283: 40. 26834-26838 Oct  
Abstract: Hyperuricemia is a significant factor in a variety of diseases, including gout and cardiovascular diseases. Although renal excretion largely determines plasma urate concentration, the molecular mechanism of renal urate handling remains elusive. Previously, we identified a major urate reabsorptive transporter, URAT1 (SLC22A12), on the apical side of the renal proximal tubular cells. However, it is not known how urate taken up by URAT1 exits from the tubular cell to the systemic circulation. Here, we report that a sugar transport facilitator family member protein GLUT9 (SLC2A9) functions as an efflux transporter of urate from the tubular cell. GLUT9-expressed Xenopus oocytes mediated saturable urate transport (K(m): 365+/-42 microm). The transport was Na(+)-independent and enhanced at high concentrations of extracellular potassium favoring negative to positive potential direction. Substrate specificity and pyrazinoate sensitivity of GLUT9 was distinct from those of URAT1. The in vivo role of GLUT9 is supported by the fact that a renal hypouricemia patient without any mutations in SLC22A12 was found to have a missense mutation in SLC2A9, which reduced urate transport activity in vitro. Based on these data, we propose a novel model of transcellular urate transport in the kidney; urate [corrected] is taken up via apically located URAT1 and exits the cell via basolaterally located GLUT9, which we suggest be renamed URATv1 (voltage-driven urate transporter 1).
Notes:
2007
Sirinun Nilwarangkoon, Naohiko Anzai, Katsuko Shiraya, Erkang Yu, Rafiqul Islam, Seok Ho Cha, Maristela Lika Onozato, Daisaku Miura, Promsuk Jutabha, Akihiro Tojo, Yoshikatsu Kanai, Hitoshi Endou (2007)  Role of mouse organic anion transporter 3 (mOat3) as a basolateral prostaglandin E2 transport pathway.   J Pharmacol Sci 103: 1. 48-55 Jan  
Abstract: Renal organic anion transporters play an important role in the handling of a number of endogenous and exogenous anionic substances in the kidney. In this study, we investigated prostaglandin E(2) (PGE(2)) transport properties and intrarenal localization of mouse organic anion transporter 3 (mOat3). When expressed in Xenopus oocytes, mOat3 mediated the time- and concentration-dependent transport of PGE(2) (K(m): 1.48 microM). PGE(2) transport mediated by mOat3 was trans-stimulated by intracellular glutarate injected into the oocytes. PGE(2) efflux via mOat3 was also trans-stimulated by extracellular glutarate. Thus, mOat3 was shown to mediate the bidirectional transport of PGE(2), partly coupled to the dicarboxylate exchange mechanism. Immunohistochemical study revealed that mOat3 protein was localized at the basolateral membrane of renal proximal and distal tubules. Furthermore, diffuse expression of mOat3, including expression in the basolateral membrane in macula densa (MD) cells, was observed. These results indicate that mOat3 plays an important role as a basolateral transport pathway of PGE(2) in the distal nephron including MD cells that may constitute one of the indispensable steps for renin release and regulation of the tubuloglomerular feedback mechanism.
Notes:
Rafiqul Islam, Naohiko Anzai, Sophapun Ekaratanawong, Sunena Kundu-Srivastava, Chun Ji Jin, Rie Noshiro-Kofuji, Kazuhiro Miyao, Ai Yamanishi, Toshiyuki Fukutomi, Yoshikatsu Kanai, Hitoshi Endou (2007)  Characterization of Human Orgnic Anion Transporter 4 (hOAT4) as a Low Affinity p-aminohippurate (PAH) Transporter   Journal of The Kyorin Medical Society 38: 2/3. 61-68 Sep  
Abstract: Organic anion transporters play an essential role in the elimination of numerous endogenous and exogenous organic anions from the body. In the present study, we investigated the transport property of human organic anion transporter 4 (hOAT4) as a p-aminohippurate (PAH) transporter. For this purpose, we established and utilized cells derived from the second segment of mice proximal tubule stably expressing hOAT4 (S2 hOAT4). RT-PCR result shows the expression of hOAT4 in S2 hOAT4. [3H] estrone sulfate (ES) and [14C] PAH uptake via hOAT4 were increased with time and concentration dependent manner. Km of ES and PAH were 9.9μM and 2.2 mM, respectively. ES uptake via hOAT4 was inhibited by PAH with competetive manner (Ki = 4.3 mM). hOAT4-mediated PAH uptake was strongly inhibited by sulfate conjugates, non-steroidal anti-inflammatory drugs, bile acids, penicillin G, BSP, probenecid, ouabain and quinidine and moderately inhibited by cAMP, cGMP and cimetidine. No inhibitory effect by tetraethylammonium and β-estradiol was observed. The present study may provide the molecular basis of drug efflux pathway in human kidneys.
Notes:
Marija Ljubojević, Daniela Balen, Davorka Breljak, Marija Kusan, Naohiko Anzai, Andrew Bahn, Gerhard Burckhardt, Ivan Sabolić (2007)  Renal expression of organic anion transporter OAT2 in rats and mice is regulated by sex hormones.   Am J Physiol Renal Physiol 292: 1. F361-F372 Jan  
Abstract: The renal reabsorption and/or excretion of various organic anions is mediated by specific organic anion transporters (OATs). OAT2 (Slc22a7) has been identified in rat kidney, where its mRNA expression exhibits gender differences [females (F) > males (M)]. The exact localization of OAT2 protein in the mammalian kidney has not been reported. Here we studied the expression of OAT2 mRNA by RT-PCR and its protein by Western blotting (WB) and immunocytochemistry (IC) in kidneys of adult intact and gonadectomized M and F, sex hormone-treated castrated M, and prepubertal M and F rats, and the protein in adult M and F mice. In adult rats, the expression of OAT2 mRNA was predominant in the outer stripe (OS) tissue, exhibiting 1) gender dependency (F > M), 2) upregulation by castration and downregulation by ovariectomy, and 3) strong downregulation by testosterone and weak upregulation by estradiol and progesterone treatment. A polyclonal antibody against rat OAT2 on WB of isolated renal membranes labeled a approximately 66-kDa protein band that was stronger in F. By IC, the antibody exclusively stained brush border (BB) of the proximal tubule S3 segment (S3) in the OS and medullary rays (F > M). In variously treated rats, the pattern of 66-kDa band density in the OS membranes and the staining intensity of BB in S3 matched the mRNA expression. The expression of OAT2 protein in prepubertal rats was low and gender independent. In mice, the expression pattern largely resembled that in rats. Therefore, OAT2 in rat (and mouse) kidney is localized to the BB of S3, exhibiting gender differences (F > M) that appear in puberty and are caused by strong androgen inhibition and weak estrogen and progesterone stimulation.
Notes:
Naohiko Anzai, Yoshikatsu Kanai, Hitoshi Endou (2007)  New insights into renal transport of urate.   Curr Opin Rheumatol 19: 2. 151-157 Mar  
Abstract: PURPOSE OF REVIEW: This review focuses on recent progress in the understanding of various aspects of renal transport of urate. RECENT FINDINGS: Since the molecular cloning of the renal apical urate/anion exchanger URAT1 (SLC22A12), several membrane proteins relevant to the transport of urate have been identified. The molecular identification of two sodium-coupled monocarboxylate transporters, SMCT1(SLC5A8) and SMCT2(SLC5A12), and the emerging role of PDZ (PSD-95, DglA, and ZO-1) scaffold for renal apical transporters have led to a new concept of renal urate transport: urate-transporting multimolecular complex, or 'urate transportsome', that may form an ultimate functional unit including the sodium-coupled urate transport system by linking URAT1 and sodium-coupled monocarboxylate transporters or the coordinated apical urate uptake system by balancing reabsorptive (URAT1) and efflux (NPT1/OATv1 and MRP4) transporters. In addition, genetic variations of the URAT1 gene are associated not only with idiopathic renal hypouricemia but also with reduced renal urate excretion. SUMMARY: Although our knowledge of renal urate handling has been increased by the molecular identification of urate transport proteins and by results of genetic studies on patients with serum urate disorders, current evidence is insufficient to fully understand the precise mechanism governing the bi-directional transport of urate. Further studies are still necessary.
Notes:
Rochelle Cunningham, Marc Brazie, Srilatha Kanumuru, E Xiaofei, Rajat Biswas, Fengying Wang, Deborah Steplock, James B Wade, Naohiko Anzai, Hitoshi Endou, Shirish Shenolikar, Edward J Weinman (2007)  Sodium-hydrogen exchanger regulatory factor-1 interacts with mouse urate transporter 1 to regulate renal proximal tubule uric acid transport.   J Am Soc Nephrol 18: 5. 1419-1425 May  
Abstract: Sodium-hydrogen exchanger regulatory factor-1-deficient (NHERF-1(-/-)) mice demonstrate increases in the urinary excretion of phosphate, calcium, and uric acid associated with interstitial deposition of calcium in the papilla of the kidney. These studies examine the role of NHERF-1 in the tubular reabsorption of uric acid and regulation of mouse urate transporter 1 (mURAT1), a newly described transporter that is responsible for the renal tubular reabsorption of uric acid. In primary cultures of mouse renal proximal tubule cells, uric acid uptake was significantly lower in NHERF-1(-/-) cells compared with wild-type cells over a large range of uric acid concentrations in the media. Western immunoblotting revealed a 56 +/- 6% decrease in the brush border membrane (BBM) expression of mURAT1 in NHERF-1(-/-) compared with wild-type control kidneys (P < 0.05). Confocal microscopy confirmed the reduced apical membrane expression of mURAT1 in NHERF-1(-/-) kidneys and demonstrated mislocalization of mURAT1 to intracellular vesicular structures. Para-aminohippurate significantly inhibited uric acid uptake in wild-type cells (41 +/- 2%) compared with NHERF-1(-/-) cells (8.2 +/- 3%). Infection of NHERF-1(-/-) cells with adenovirus-green fluorescence protein-NHERF-1 resulted in significantly higher rates of uric acid transport (15.4 +/- 1.1 pmol/microg protein per 30 min) compared with null cells that were infected with control adenovirus-green fluorescence protein (7.9 +/- 0.3) and restoration of the inhibitory effect of para-aminohippurate (% inhibition 34 +/- 4%). These findings indicate that NHERF-1 exerts a significant effect on the renal tubular reabsorption of uric acid in the mouse by modulating the BBM abundance of mURAT1 and possibly other BBM uric acid transporters.
Notes:
Naohiko Anzai, Hitoshi Endou (2007)  Drug discovery for hyperuricemia   Expert Opinion on Drug Discovery 2: 9. 1251-1261 Sep  
Abstract: Hyperuricemia is associated with an increased risk of developing gout. This increases with the degree and duration of hyperuricemia. Gout can be managed by dietary modification and pharmacologic urate-lowering therapies. The recent identification of the renal apical urate/anion exchanger URAT1 (SLC22A12) and several membrane proteins relevant to the transport of urate play an important role in gaining a better understanding of the mode of action of many drugs used to treat gout. As described in this review, therapeutics designed to modify URAT1 transport activities might be useful in treating pathologies associated with hyperuricemia such as gout and urolithiasis. Continuing studies into the urate transportsome hold promise for the development of new, more effective therapeutics for hyperuricemia.
Notes:
Ho Jung Shin, Naohiko Anzai, Atsushi Enomoto, Xin He, Do Kyung Kim, Hitoshi Endou, Yoshikatsu Kanai (2007)  Novel liver-specific organic anion transporter OAT7 that operates the exchange of sulfate conjugates for short chain fatty acid butyrate.   Hepatology 45: 4. 1046-1055 Apr  
Abstract: The liver plays an important role in the elimination of endogenous and exogenous lipophilic organic compounds from the body, which is mediated by various carrier proteins that differ in substrate specificity and kinetic properties. Here, we have characterized a novel member of the organic anion transporter family (SLC22) isolated from human liver. The transporter named organic anion transporter 7 (OAT7/ SLC22A9) showed 35% to 46% identities to those of other organic anion transporters of SLC22 family. When expressed in Xenopus oocytes, OAT7 mediated Na(+)-independent, high-affinity transport of sulfate-conjugated steroids, estrone sulfate (ES; K(m) = 8.7 microM), and dehydroepiandrosterone sulfate (K(m) = 2.2 microM). In addition, OAT7 interacted with negatively charged sulfobromophthalein, indocyanine green, and several sulfate-conjugated xenobiotics. In contrast, glucuronide and glutathione conjugates exhibited no inhibitory effects on OAT7-mediated [(3)H]ES transport. OAT7-mediated [(3)H]ES transport was trans-stimulated by three-carbon to five-carbon (C3 to C5) short-chain fatty acids. The efflux of [(14)C]butyrate (C4) via OAT7 was significantly trans-stimulated by extracellular ES. Furthermore, OAT7 mediated [(14)C]butyrate uptake and [(3)H]ES efflux in exchange for extracellular butyrate both in Xenopus oocytes and OAT7-stably expressing cells. OAT7 protein was localized in the sinusoidal membrane of hepatocytes by immunohistochemical analysis. CONCLUSION: OAT7 is the first liver-specific transporter among members of the organic anion transporters of SLC22 family. Our findings suggest a new class of substrates for organic anion transporters and provide evidence for the transport of anionic substances such as sulfate-conjugates in exchange for butyrate in hepatocytes.
Notes:
T Kimura, J Perry, N Anzai, J B Pritchard, R Moaddel (2007)  Development and characterization of immobilized human organic anion transporter-based liquid chromatographic stationary phase: hOAT1 and hOAT2.   J Chromatogr B Analyt Technol Biomed Life Sci 859: 2. 267-271 Nov  
Abstract: This paper reports the development of liquid chromatographic columns containing immobilized organic anion transporters (hOAT1 and hOAT2). Cellular membrane fragments from MDCK cells expressing hOAT1 and S2 cells expressing hOAT2 were immobilized on the surface of the immobilized artificial membrane (IAM) liquid chromatographic stationary phase. The resulting stationary phases were characterized by frontal affinity chromatography, using the marker ligand [3H]-adefovir for the hOAT1 and [14C]-p-aminohippurate for the hOAT2 in the presence of multiple displacers. The determined binding affinities (Kd) for eight OAT1 ligands and eight OAT2 ligands were correlated with literature values and a statistically significant correlation was obtained for both the hOAT1 and hOAT2 columns: r2=0.688 (p<0.05) and r2=0.9967 (p<0.0001), respectively. The results indicate that the OAT1 and OAT2 have been successfully immobilized with retention of their binding activity. The use of these columns to identify ligands to the respective transporters will be presented.
Notes:
2006
Anabel Brandoni, Silvina R Villar, Juan C Picena, Naohiko Anzai, Hitoshi Endou, Adriana M Torres (2006)  Expression of rat renal cortical OAT1 and OAT3 in response to acute biliary obstruction.   Hepatology 43: 5. 1092-1100 May  
Abstract: Renal function in the course of obstructive jaundice has been the subject of great interest; however, little is known about the expression of renal organic anion transporters. The objective of this work was to study, in rats with acute extrahepatic cholestasis, the cortical renal expression of the organic anion transporter 1 (OAT1) and the organic anion transporter 3 (OAT3), in association with the pharmacokinetics and renal excretion of furosemide (FS). Male Wistar rats underwent bile duct ligation (BDL rats). Pair-fed sham-operated rats served as controls. All studies were carried out 21 hours after surgery. Rats were anesthetized and the pharmacokinetic parameters of FS and the renal elimination of FS were determined. Afterwards, the kidneys were excised and processed for immunoblot (basolateral membrane and renal homogenates) or immunocytochemical (light microscopic and confocal immunofluorescence microscopic analysis) techniques. The systemic and renal clearance of FS as well as the excreted and secreted load of FS increased in BDL rats. In kidneys from BDL rats, immunoblotting showed a significant increase in the abundance of both OAT1 and OAT3 in homogenates from renal cortex. In basolateral membranes from kidney cortex of BDL rats, OATI abundance was also increased and OAT3 abundance was not modified. Immunocytochemical techniques confirmed these results. In conclusion, acute obstructive jaundice is associated with an upregulation of OAT1 and OAT3, which might explain, at least in part, the increased systemic and renal elimination of FS.
Notes:
R Noshiro, N Anzai, T Sakata, H Miyazaki, T Terada, H J Shin, X He, D Miura, K Inui, Y Kanai, H Endou (2006)  The PDZ domain protein PDZK1 interacts with human peptide transporter PEPT2 and enhances its transport activity.   Kidney Int 70: 2. 275-282 Jul  
Abstract: The proton-coupled peptide transporter PEPT2 (SLC15A2) mediates the high-affinity low-capacity transport of small peptides as well as various oral peptide-like drugs in the kidney. In contrast to its well-characterized transport properties, there is less information available on its regulatory mechanism, although the interaction of PEPT2 to the PDZ (PSD-95, DglA, and ZO-1)-domain protein PDZK1 has been preliminarily reported. To examine whether PDZK1 is a physiological partner of PEPT2 in kidneys, we started from a yeast two-hybrid screen of a human kidney cDNA library with the C-terminus of PEPT2 (PEPT2 C-terminus (PEPT2-CT)) as bait. We could identify PDZK1 as one of the positive clones. This interaction requires the PDZ motif of PEPT2-CT detected by a yeast two-hybrid assay, in vitro binding assay and co-immunoprecipitation. The binding affinities of second and third PDZ domains of PDZK1 to PEPT2-CT were measured by surface plasmon resonance. Co-immunoprecipitation using human kidney membrane fraction and localization of PEPT2 in renal apical proximal tubules revealed the physiological meaning of this interaction in kidneys. Furthermore, we clarified the mechanism of enhanced glycylsarcosine (Gly-Sar) transport activity in PEPT2-expressing HEK293 cells after the PDZK1 coexpression. This augmentation was accompanied by a significant increase in the V(max) of Gly-Sar transport via PEPT2 and it was also associated with the increased surface expression level of PEPT2. These results indicate that the PEPT2-PDZK1 interaction thus plays a physiologically important role in both oligopeptide handling as well as peptide-like drug transport in the human kidney.
Notes:
Y Shigeta, Y Kanai, A Chairoungdua, N Ahmed, S Sakamoto, H Matsuo, D K Kim, M Fujimura, N Anzai, K Mizoguchi, T Ueda, K Akakura, T Ichikawa, H Ito, H Endou (2006)  A novel missense mutation of SLC7A9 frequent in Japanese cystinuria cases affecting the C-terminus of the transporter.   Kidney Int 69: 7. 1198-1206 Apr  
Abstract: Cystinuria is caused by the inherited defect of apical membrane transport systems for cystine and dibasic amino acids in renal proximal tubules. Mutations in either SLC7A9 or SLC3A1 gene result in cystinuria. The mutations of SLC7A9 gene have been identified mainly from Italian, Libyan Jewish, North American, and Spanish patients. In the present study, we have analyzed cystinuria cases from oriental population (mostly Japanese). Mutation analyses of SLC7A9 and SLC3A1 genes were performed on 41 cystinuria patients. The uptake of 14C-labeled cystine in COS-7 cells was measured to determine the functional properties of mutants. The protein expression and localization were examined by Western blot and confocal laser-scanning microscopy. Among 41 patients analyzed, 35 were found to possess mutations in SLC7A9. The most frequent one was a novel missense mutation P482L that affects a residue near the C-terminus end of the protein and causes severe loss of function. In MDCK II and HEK293 cells, we found that P482L protein was expressed and sorted to the plasma membrane as well as wild type. The alteration of Pro482 with amino acids with bulky side chains reduced the transport function of b(0,+)AT/BAT1. Interestingly, the mutations of SLC7A9 for Japanese cystinuria patients are different from those reported for European and American population. The results of the present study contribute toward understanding the distribution and frequency of cystinuria-related mutations of SLC7A9.
Notes:
Keisuke Tachibana, Naohiko Anzai, Chihiro Ueda, Tatsuya Katayama, Takayoshi Kirino, Rika Takahashi, Daisuke Yamasaki, Kenji Ishimoto, Toshiya Tanaka, Takao Hamakubo, Yukihiko Ueda, Hiroyuki Arai, Juro Sakai, Tatsuhiko Kodama, Takefumi Doi (2006)  Analysis of PPAR alpha function in human kidney cell line using siRNA.   Nucleic Acids Symp Ser (Oxf) 50. 257-258  
Abstract: The peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that belong to the nuclear hormone receptor superfamily. PPARalpha is mainly expressed in the liver, kidney, heart and muscle. PPARalpha activates fatty acid catabolism, stimulates gluconeogenesis and ketone body synthesis and is involved in the control of lipoprotein assembly. Although PPARalpha is well characterized in the liver, its physiological function is unknown in the kidney. To investigate the intimate function of PPARalpha in the kidney, we analyzed the target gene expression in human metastatic renal cell carcinoma cell line, Caki-1, using small interfering RNA (siRNA) against PPARalpha and real-time RT-PCR methods. We found that some selected genes (long-chain fatty-acid-CoA ligase (FACL1), carnitine palmitoyltransferase 1A (CPT1A), adipose differentiation-related protein (ADRP) and aquaporin 3 (AQP3)) were down-regulated by PPARalpha siRNA. These results suggest that PPARalpha regulates fatty acid metabolism and body water homeostasis in this cell line.
Notes:
Naohiko Anzai, Yoshikatsu Kanai, Hitoshi Endou (2006)  Organic anion transporter family: current knowledge.   J Pharmacol Sci 100: 5. 411-426  
Abstract: Organic anion transporters (OATs) play an essential role in the elimination of numerous endogenous and exogenous organic anions from the body. The renal OATs contribute to the excretion of many drugs and their metabolites that are important in clinical medicine. Several families of multispecific organic anion and cation transporters, including OAT family transporters, have recently been identified by molecular cloning. The OAT family consists of six isoforms (OAT1 - 4, URAT1, and rodent Oat5) and they are all expressed in the kidney, while some are also expressed in the liver, brain, and placenta. The OAT family represents mainly the renal secretory and reabsorptive pathway for organic anions and is also involved in the distribution of organic anions in the body, drug-drug interactions, and toxicity of anionic substances such as nephrotoxic drugs and uremic toxins. In this review, current knowledge of and recent progress in the understanding of several aspects of OAT family members are discussed.
Notes:
Anabel Brandoni, Naohiko Anzai, Yoshikatsu Kanai, Hitoshi Endou, Adriana Mónica Torres (2006)  Renal elimination of p-aminohippurate (PAH) in response to three days of biliary obstruction in the rat. The role of OAT1 and OAT3.   Biochim Biophys Acta 1762: 7. 673-682 Jul  
Abstract: Pharmacokinetic studies of the drugs administered to subjects with mechanical cholestasis are scarce. The purpose of the present study was to examine the effects of bile duct ligation of 3 days (peak of elevation of serum bile acids and bilirubin) on the systemic and renal PAH clearance and on the expression of cortical renal OAT1 and OAT3 in a rat model. PAH is the prototypical substrate of the renal organic anion transport system. Male Wistar rats underwent a bile duct ligation (BDL rats). Pair-fed sham-operated rats served as controls. BDL rats displayed a significantly lower systemic PAH clearance. Renal studies revealed a reduction in the renal clearance and in the excreted and secreted load of PAH in BDL rats. The OAT1 protein expression in kidney homogenates was not modified, but it decreased in the basolateral membranes from BDL rats. In contrast, OAT3 abundance in both kidney cortex homogenates and in basolateral membranes increased by 3 days after the ligation. Immunocytochemical studies (light microscopic and confocal immunofluorescence microscopic analyses) confirmed the changes in the renal expression of these transport proteins. The present study demonstrates the key role of OAT1 expression in the impaired elimination of PAH after 3 days of obstructive cholestasis.
Notes:
Abdul R Asif, Marija Ljubojevic, Ivan Sabolic, Vladimir Shnitsar, Maria Metten, Naohiko Anzai, Gerhard A Müller, Gerhard Burckhardt, Yohannes Hagos (2006)  Regulation of steroid hormone biosynthesis enzymes and organic anion transporters by forskolin and DHEA-S treatment in adrenocortical cells.   Am J Physiol Endocrinol Metab 291: 6. E1351-E1359 Dec  
Abstract: Several important physiological functions are regulated by cortisol. Previously, we demonstrated the involvement of human organic anion transporter 3 (hOAT3) in cortisol release. In the present study, we investigated the influence of dehydroepiandrosterone sulfate (DHEA-S) and estrone sulfate on cortisol release in a human adrenocortical cell line (NCI-H295R) compared with forskolin stimulation. Additionally, we examined the impact of forskolin and DHEA-S on the expression of key enzymes in steroid biosynthesis and expression of hOAT3 and -4 in NCI-H295R cells. The cortisol release was increased 10-fold after 24-h incubation with DHEA-S, but incubation with estrone sulfate did not show any significant change in cortisol release. When cells were incubated with DHEA-S in the presence of forskolin, an additive influence of DHEA-S stimulation of cortisol was recorded over forskolin alone. The 24-h stimulation of NCI-H295R cells with forskolin increased the expression of steroidogenic acute regulatory protein (StAR), CYP17, CYP21A2, and CYP11A1, whereas only StAR mRNA expression was increased significantly by incubation with DHEA-S. Immunofluorescence analyses revealed strongly elevated expression of hOAT3 by forskolin as well as by DHEA-S stimulation. We conclude that the increased cortisol release of adrenocortical cells by DHEA-S and forskolin stimulation is probably due to high expression of the key enzymes of steroid biosynthesis and hOAT3.
Notes:
2005
Vladiana Crljen, Ivana Sabolić, Jelena Susac, Dorothea Appenroth, Carol M Herak-Kramberger, Marija Ljubojević, Naohiko Anzai, Roberto Antolović, Gerhard Burckhardt, Christian Fleck, Ivan Sabolić (2005)  Immunocytochemical characterization of the incubated rat renal cortical slices.   Pflugers Arch 450: 4. 269-279 Jul  
Abstract: The use of renal cortical slices in vitro and the data obtained in these studies have been subjects of controversy, largely due to uncertain viability, e.g., structural and functional integrity of the proximal and other tubules. However, detailed studies of tubule integrity have not been reported. To correlate functional and structural viability of the hand-cut rat renal cortical slices, incubated in optimally conditioned media for up to 25 h, we studied the time course of p-aminohippurate (PAH) uptake, the immunocytochemical distribution of several proteins that reside in the proximal tubule basolateral [Na/K-ATPase, organic anion transporters (OAT)1 and OAT3], or brush border [megalin, sodium-proton exchanger (NHE)3] membrane, as well as the general integrity of the tubule epithelium and its cytoskeleton (actin filaments, microtubules). PAH uptake in slices was proportional to time within 1 h of incubation and gradually declined thereafter. The immunostaining experiments indicated a fast, time-dependent loss of basolateral transporters, at a rate of OAT1 > Na/K-ATPase > OAT3. In the brush border membrane, the loss of megalin was faster than that of NHE3, and a partial redistribution of NHE3 into the basolateral domain indicated the loss of cell polarity. The loss of intracellular actin and tubulin cytoskeleton in the proximal tubule was already visible after 15 min of incubation and gradually increased with time, whereas a partial redistribution of actin to the basolateral domain indicated a compromised polarity of the cells. The data also revealed very early (after 15 min) necrotic events in the proximal tubule epithelium, with sloughing of brush border and cell debris into the tubule lumen, detachment of cells from the basal membrane, and opening and widening of the tubule lumen. We conclude that the loss of cellular structure, cytoskeleton, and cell membrane transporters in the nephron epithelium is a very early event in the incubated rat renal cortical slices.
Notes:
Adriana Mónica Torres, Myriam Mac Laughlin, Angélica Muller, Anabel Brandoni, Naohiko Anzai, Hitoshi Endou (2005)  Altered renal elimination of organic anions in rats with chronic renal failure.   Biochim Biophys Acta 1740: 1. 29-37 Apr  
Abstract: The progress of chronic renal failure (CRF) is characterized by the development of glomerular and tubular lesions. However, little is known about the expression of organic anions renal transporters. The objective of this work was to study, in rats with experimental CRF (5/6 nephrectomy), the expression of the organic anion transporter 1 (OAT1) and organic anion transporter 3 (OAT3) and their contribution to the pharmacokinetics and renal excretion of p-aminohippurate (PAH). Two groups of animals were used: Sham and CRF. Six months after surgery, systolic blood pressure and plasma creatinine concentrations were significantly higher in CRF groups. CRF rats showed a diminution in: the filtered, secreted and excreted load of PAH; the systemic clearance of PAH; the renal OAT1 expression; and the renal Na-K-ATPase activity. No remarkable modifications were observed in the OAT3 expression from CRF kidneys. The diminution in the systemic depuration and renal excretion of PAH may be explained by the decrease in its filtered and secreted load. The lower OAT1 expression in remnant renal mass of CRF rats or/and the lower activity of Na-K-ATPase might justify, at least in part, the diminished secreted load of this organic anion.
Notes:
Naohiko Anzai, Atsushi Enomoto, Hitoshi Endou (2005)  Renal urate handling: clinical relevance of recent advances.   Curr Rheumatol Rep 7: 3. 227-234 Jun  
Abstract: Urate is the major inert end product of purine degradation in higher primates in contrast to most other mammals because of the genetic silencing of hepatic oxidative enzyme uricase. The kidney plays a dominant role in urate elimination. The kidney excretes 70% of the daily urate production. Therefore, it is important to understand renal urate handling mechanism because the under excretion of urate has been implicated in the development of hyperuricemia that leads to gout. The urate transport systems exist in the proximal tubule but they are complicated because of their bidirectional transport and the species differences. Recently, we have identified the urate-anion exchanger URAT1 (SLC22A12) in the human kidney and found that defects in SLC22A12 lead to idiopathic renal hypouricemia. URAT1 is targeted by uricosuric and antiuricosuric agents that affect urate excretion. Molecular identification of urate transporting proteins will lead to the new drug development for hyperuricemia.
Notes:
Naohiko Anzai, Promsuk Jutabha, Yoshikatsu Kanai, Hitoshi Endou (2005)  Integrated physiology of proximal tubular organic anion transport.   Curr Opin Nephrol Hypertens 14: 5. 472-479 Sep  
Abstract: PURPOSE OF REVIEW: Renal organic anion transport proteins play important roles in the reabsorption and the secretion of endogenous and exogenous compounds. This review focuses on the interpretation of the physiological integration of identified transport molecules in the renal proximal tubules. RECENT FINDINGS: To date, molecular identification of organic anion transport proteins is still continuing: rodent organic anion transporter 5, organic anion-transporting polypeptide 4C1, voltage-driven organic anion transporter 1, multidrug resistance-associated protein 4, and sodium-coupled monocarboxylate transporter have yielded additional information in this field. In addition, particularly at the apical membrane of the proximal tubules, the importance of the PDZ (PSD-95, DglA, and ZO-1) binding domain proteins has emerged in the formation of the multimolecular complex as a functional unit of membrane transport. Finally, discovery of dicarboxylate receptors in the renal tubular cells raises the possibility that dicarboxylate anions function as intrarenal signaling molecules. This novel aspect of renal organic anion transport, the potential modulation of signaling via dicarboxylate receptors, may be of significant relevance to renovascular hypertension and other renal diseases. SUMMARY: Comprehensive understanding of the multimolecular complex, which is composed of transporters and their related signaling elements and is supported by the scaffold proteins underneath the plasma membrane, may be useful in clarifying complex transport phenomena such as renal apical organic anion handling. In addition to the recent proteomics approaches and conventional molecular physiology, it is necessary to develop novel methods to analyze the overall function of the multimolecular complex for the post-genomic era.
Notes:
Silvina R Villar, Anabel Brandoni, Naohiko Anzai, Hitoshi Endou, Adriana M Torres (2005)  Altered expression of rat renal cortical OAT1 and OAT3 in response to bilateral ureteral obstruction.   Kidney Int 68: 6. 2704-2713 Dec  
Abstract: BACKGROUND: Bilateral ureteral obstruction (BUO) is characterized by the development of hemodynamic and tubular lesions. However, little is known about the expression of organic anion renal transporters. The objective of this work was to study the renal excretion of p-aminohippurate (PAH) and the cortical renal expression of the organic anion transporter 1 (OAT1) and organic anion transporter 3 (OAT3) in BUO rats. METHODS: Male Wistar rats underwent bilateral obstruction of the proximal ureters for 24 hours (BUO) or sham operation. After 24 hours of ureteral releasing, the following studies were performed: PAH renal excretion employing conventional clearance techniques and OAT1 and OAT3 abundance (homogenates, intracellular and basolateral plasma membrane fractions from renal cortex) using immunoblotting and immunocytochemical techniques (light microscopic and confocal immunofluorescence microscopic analysis). RESULTS: BUO rats showed a lower renal excretion of PAH. In obstructed kidneys, immunoblotting revealed a significant decrease in the abundance of both OAT1 and OAT3 in basolateral plasma membranes from renal cortex. An increase of OAT1 expression was observed in homogenates and in intracellular membrane fractions in kidneys from BUO rats compared with sham-operated ones, indicating an internalization of this carrier. Immunocytochemical techniques confirmed these results. On the contrary, OAT3 expression was reduced both in homogenates and in intracellular membrane fractions in obstructed kidneys. CONCLUSION: BUO was associated with down-regulation of OAT1 and OAT3 in basolateral plasma membranes from proximal tubule cells, thus these carriers may play important roles in the impaired organic anion excretion displayed in the obstructed kidney.
Notes:
Hiroki Miyazaki, Naohiko Anzai, Sophapun Ekaratanawong, Takeshi Sakata, Ho Jung Shin, Promsuk Jutabha, Taku Hirata, Xin He, Hiroshi Nonoguchi, Kimio Tomita, Yoshikatsu Kanai, Hitoshi Endou (2005)  Modulation of renal apical organic anion transporter 4 function by two PDZ domain-containing proteins.   J Am Soc Nephrol 16: 12. 3498-3506 Dec  
Abstract: Human organic anion transporter 4 (OAT4) is an apical organic anion/dicarboxylate exchanger in the renal proximal tubules and mediates high-affinity transport of steroid sulfates such as estrone-3-sulfate (E1S) and dehydroepiandrosterone sulfate. Here, two multivalent PDZ (PSD-95/Discs Large/ZO-1) proteins PDZK1 and NHERF1 were examined as interactors of OAT4 by a yeast two-hybrid assay. These interactions require the extreme C-terminal region of OAT4 and the first and fourth PDZ domains of PDZK1 and the first PDZ domain of NHERF1. These interactions were confirmed by surface plasmon resonance assays (K(D): 36 nM, 1.2 microM, and 41.7 microM, respectively). In vitro binding assays and co-immunoprecipitation studies revealed that the OAT4 wild-type but not a mutant lacking the PDZ motif interacted directly with both PDZK1 and NHERF1. OAT4, PDZK1, and NHERF1 proteins were shown to be localized at the apical membrane of renal proximal tubules. The association with PDZK1 or NHERF1 enhanced OAT4-mediated E1S transport activities in HEK293 cells (1.2- to 1.4-fold), and the deletion of the OAT4 C-terminal PDZ motif abolished this effect. The augmentation of the transport activity was accompanied by alteration in V(max) of E(1)S transport via OAT4 and was associated with the increased surface expression level of OAT4 protein. This study indicates that the functional activity of OAT4 is modulated through the PDZ interaction with the network of PDZK1 and NHERF1 and suggests that OAT4 is involved in the regulated apical organic anion handling in the renal proximal tubules, provided by the PDZ scaffold.
Notes:
Naohiko Anzai, Promsuk Jutabha, Atsushi Enomoto, Hirokazu Yokoyama, Hiroshi Nonoguchi, Taku Hirata, Katsuko Shiraya, Xin He, Seok Ho Cha, Michio Takeda, Hiroki Miyazaki, Takeshi Sakata, Kimio Tomita, Takashi Igarashi, Yoshikatsu Kanai, Hitoshi Endou (2005)  Functional characterization of rat organic anion transporter 5 (Slc22a19) at the apical membrane of renal proximal tubules.   J Pharmacol Exp Ther 315: 2. 534-544 Nov  
Abstract: A novel member of the organic anion transporter (OAT) family, Oat5 (Slc22a19), has been reported to transport a naturally occurring mycotoxin, ochratoxin A (OTA). However, neither its endogenous substrate and driving force nor physiological functions have been determined. Herein, we report the functional characterization of rat Oat5 (rOat5), as well as its intrarenal distribution and membrane localization. When expressed in Xenopus laevis oocytes, rOat5 mediated the transport of sulfate conjugates of steroids such as estrone-3-sulfate (E(1)S; K(m) = 18.9 +/- 3.9 microM) and dehydroepiandrosterone sulfate (K(m) = 2.3 +/- 0.2 microM) in a sodium-independent manner, in addition to OTA. The rOat5-mediated E(1)S transport was strongly inhibited by four-carbon (C4) dicarboxylate succinate and longer dicarboxylates (C7-C9). The uptake of [(3)H]E(1)S via rOat5 was significantly trans-stimulated by succinate, and the efflux of [(14)C]succinate was significantly trans-stimulated by E(1)S. A similar trans-stimulatory effect of preloaded succinate on E(1)S uptake was also detected in cells stably expressing rOat5 (S(2) rOat5). rOat5 interacted with chemically heterogenous anionic compounds. The rOat5-mediated E(1)S transport was inhibited by several sulfate conjugates, such as 4-methylumbelliferyl sulfate and beta-estradiol sulfate, but not by glucuronide conjugates. An immunohistochemical study showed that rOat5 was localized at the apical membrane of renal proximal tubules in the corticomedullary region. rOat5 mRNA was expressed in the late segments (S(2) and S(3)) of proximal tubules. These results indicate that rOat5 is renal organic anion/dicarboxylates exchanger and, under physiological conditions, may function as an apical reabsorptive pathway for organic anions in proximal tubules driven by an outward gradient of dicarboxylates.
Notes:
2004
Takeshi Sakata, Naohiko Anzai, Ho Jung Shin, Rie Noshiro, Taku Hirata, Hirokazu Yokoyama, Yoshikatsu Kanai, Hitoshi Endou (2004)  Novel single nucleotide polymorphisms of organic cation transporter 1 (SLC22A1) affecting transport functions.   Biochem Biophys Res Commun 313: 3. 789-793 Jan  
Abstract: Organic cation transporter OCT1 (SLC22A1) plays an essential role in absorption, distribution, and excretion of various xenobiotics including therapeutically important drugs. In the present study, we analyzed the functional properties of the single nucleotide polymorphisms (SNPs) in SLC22A1 gene found in Japanese control individuals. Four mutations resulting in the amino acid changes (F160L, P283L, R287G, and P341L) were functionally characterized in Xenopus oocyte expression system. Two new SNPs, identified in Japanese population, P283L and R287G exhibited no uptake of both [14C]TEA and [3H]MPP+, although their protein expressions were detected in the plasma membrane of the oocytes injected with their cRNAs. Uptake of [14C]TEA by P341L was reduced to 65.1% compared to wild type, whereas F160L showed no significant change in its transport activity. This study suggests that the newly found OCT1 variants will contribute to inter-individual variations leading to the differences in cationic drug disposition and perhaps certain disease processes.
Notes:
Toshihisa Ishikawa, Akira Tsuji, Kenichi Inui, Yoshimichi Sai, Naohiko Anzai, Morimasa Wada, Hitoshi Endou, Yasuhiro Sumino (2004)  The genetic polymorphism of drug transporters: functional analysis approaches.   Pharmacogenomics 5: 1. 67-99 Jan  
Abstract: Evidence is accumulating to strongly suggest that drug transporters are one of the determining factors governing the pharmacokinetic profile of drugs. To date, a variety of drug transporters have been cloned and classified as solute carriers and ATP-binding cassette transporters. Such drug transporters are expressed in various tissues such as the intestine, brain, liver, and kidney, and play critical roles in the absorption, distribution and excretion of drugs. However, at the present time, information is limited regarding the genetic polymorphism of drug transporters and its impact on their function. In this context, we have undertaken the functional analyses of the polymorphisms identified in drug transporter genes. This article aims to provide an overview on the functional aspects of the non-synonymous polymorphisms of drug transporters and to present standard methods for the evaluation of the effect of polymorphisms on their function.
Notes:
Sophapun Ekaratanawong, Naohiko Anzai, Promsuk Jutabha, Hiroki Miyazaki, Rie Noshiro, Michio Takeda, Yoshikatsu Kanai, Samaisukh Sophasan, Hitoshi Endou (2004)  Human organic anion transporter 4 is a renal apical organic anion/dicarboxylate exchanger in the proximal tubules.   J Pharmacol Sci 94: 3. 297-304 Mar  
Abstract: Human organic anion transporter OAT4 is expressed in the kidney and placenta and mediates high-affinity transport of estrone-3-sulfate (E1S). Because a previous study demonstrated no trans-stimulatory effects by E1S, the mode of organic anion transport via OAT4 remains still unclear. In the present study, we examined the driving force of OAT4 using mouse proximal tubular cells stably expressing OAT4 (S2 OAT4). OAT4-mediated E1S uptake was inhibited by glutarate (GA) (IC50:1.25 mM) and [14C]GA uptake via S2 OAT4 was significantly trans-stimulated by unlabeled GA (5 mM) (P<0.001). [3H]E1S uptake via S2 OAT4 was significantly trans-stimulated by preloaded GA (P<0.001) and its [14C]GA efflux was significantly trans-stimulated by unlabeled E1S in the medium (P<0.05). In addition, both the uptake and efflux of [14C]p-aminohippuric acid (PAH) and [14C]GA via S2 OAT4 were significantly trans-stimulated by unlabeled GA or PAH. The immunoreactivities of OAT4 were observed in the apical membrane of proximal tubules along with those of basolateral organic anion/dicarboxylate exchangers such as hOAT1 and hOAT3 in the same tubular population. These results indicate that OAT4 is an apical organic anion/dicarboxylate exchanger and mainly functions as an apical pathway for the reabsorption of some organic anions in renal proximal tubules driven by an outwardly directed dicarboxylate gradient.
Notes:
Habib Hasannejad, Michio Takeda, Kentarou Taki, Ho Jung Shin, Ellappan Babu, Promsuk Jutabha, Suparat Khamdang, Mahmoud Aleboyeh, Maristela Lika Onozato, Akihiro Tojo, Atsushi Enomoto, Naohiko Anzai, Shinichi Narikawa, Xiu-Lin Huang, Toshimitsu Niwa, Hitoshi Endou (2004)  Interactions of human organic anion transporters with diuretics.   J Pharmacol Exp Ther 308: 3. 1021-1029 Mar  
Abstract: The tubular secretion of diuretics in the proximal tubule has been shown to be critical for the action of drugs. To elucidate the molecular mechanisms for the tubular excretion of diuretics, we have elucidated the interactions of human organic anion transporters (hOATs) with diuretics using cells stably expressing hOATs. Diuretics tested were thiazides, including chlorothiazide, cyclothiazide, hydrochlorothiazide, and trichlormethiazide; loop diuretics, including bumetanide, ethacrynic acid, and furosemide; and carbonic anhydrase inhibitors, including acetazolamide and methazolamide. These diuretics inhibited organic anion uptake mediated by hOAT1, hOAT2, hOAT3, and hOAT4 in a competitive manner. hOAT1 exhibited the highest affinity interactions for thiazides, whereas hOAT3 did those for loop diuretics. hOAT1, hOAT3, and hOAT4 but not hOAT2, mediated the uptake of bumetanide. hOAT3 and hOAT4, but not hOAT1 mediated the efflux of bumetanide. hOAT1 and hOAT3, but not hOAT2 and hOAT4 mediated the uptake of furosemide. In conclusion, it was suggested that hOAT1 may play an important role in the basolateral uptake of thiazides, and hOAT3 in the uptake of loop diuretics. In addition, it was also suggested that bumetanide taken up by hOAT3 and/or hOAT1 is excreted into the urine by hOAT4.
Notes:
Naohiko Anzai, Hiroki Miyazaki, Rie Noshiro, Suparat Khamdang, Arthit Chairoungdua, Ho-Jung Shin, Atsushi Enomoto, Shinichi Sakamoto, Taku Hirata, Kimio Tomita, Yoshikatsu Kanai, Hitoshi Endou (2004)  The multivalent PDZ domain-containing protein PDZK1 regulates transport activity of renal urate-anion exchanger URAT1 via its C terminus.   J Biol Chem 279: 44. 45942-45950 Oct  
Abstract: The urate-anion exchanger URAT1 is a member of the organic anion transporter (OAT) family that regulates blood urate level in humans and is targeted by uricosuric and antiuricosuric agents. URAT1 is expressed only in the kidney, where it is thought to participate in tubular urate reabsorption. We found that the multivalent PDZ (PSD-95, Drosophila discs-large protein, Zonula occludens protein 1) domain-containing protein, PDZK1 interacts with URAT1 in a yeast two-hybrid screen. Such an interaction requires the PDZ motif of URAT1 in its extreme intracellular C-terminal region and the first, second, and fourth PDZ domains of PDZK1 as identified by yeast two-hybrid assay, in vitro binding assay and surface plasmon resonance analysis (K(D) = 1.97-514 nM). Coimmunoprecipitation studies revealed that the wild-type URAT1, but not its mutant lacking the PDZ-motif, directly interacts with PDZK1. Colocalization of URAT1 and PDZK1 was observed at the apical membrane of renal proximal tubular cells. The association of URAT1 with PDZK1 enhanced urate transport activities in HEK293 cells (1.4-fold), and the deletion of the URAT1 C-terminal PDZ motif abolished this effect. The augmentation of the transport activity was accompanied by a significant increase in the V(max) of urate transport via URAT1 and was associated with the increased surface expression level of URAT1 protein from HEK293 cells stably expressing URAT1 transfected with PDZK1. Taken together, the present study indicates the novel role of PDZK1 in regulating the functional activity of URAT1-mediated urate transport in the apical membrane of renal proximal tubules.
Notes:
Robert Kleta, Elisa Romeo, Zorica Ristic, Toshihiro Ohura, Caroline Stuart, Mauricio Arcos-Burgos, Mital H Dave, Carsten A Wagner, Simone R M Camargo, Sumiko Inoue, Norio Matsuura, Amanda Helip-Wooley, Detlef Bockenhauer, Richard Warth, Isa Bernardini, Gepke Visser, Thomas Eggermann, Philip Lee, Arthit Chairoungdua, Promsuk Jutabha, Ellappan Babu, Sirinun Nilwarangkoon, Naohiko Anzai, Yoshikatsu Kanai, Francois Verrey, William A Gahl, Akio Koizumi (2004)  Mutations in SLC6A19, encoding B0AT1, cause Hartnup disorder.   Nat Genet 36: 9. 999-1002 Sep  
Abstract: Hartnup disorder, an autosomal recessive defect named after an English family described in 1956 (ref. 1), results from impaired transport of neutral amino acids across epithelial cells in renal proximal tubules and intestinal mucosa. Symptoms include transient manifestations of pellagra (rashes), cerebellar ataxia and psychosis. Using homozygosity mapping in the original family in whom Hartnup disorder was discovered, we confirmed that the critical region for one causative gene was located on chromosome 5p15 (ref. 3). This region is homologous to the area of mouse chromosome 13 that encodes the sodium-dependent amino acid transporter B(0)AT1 (ref. 4). We isolated the human homolog of B(0)AT1, called SLC6A19, and determined its size and molecular organization. We then identified mutations in SLC6A19 in members of the original family in whom Hartnup disorder was discovered and of three Japanese families. The protein product of SLC6A19, the Hartnup transporter, is expressed primarily in intestine and renal proximal tubule and functions as a neutral amino acid transporter.
Notes:
2003
Mahmoud Alebouyeh, Michio Takeda, Maristela Lika Onozato, Akihiro Tojo, Rie Noshiro, Habib Hasannejad, Jun Inatomi, Shinichi Narikawa, Xiu-Lin Huang, Suparat Khamdang, Naohiko Anzai, Hitoshi Endou (2003)  Expression of human organic anion transporters in the choroid plexus and their interactions with neurotransmitter metabolites.   J Pharmacol Sci 93: 4. 430-436 Dec  
Abstract: The purpose of the present study was to elucidate the expression of human organic anion transporter 1 (hOAT1) and hOAT3 in the choroid plexus of the human brain and their interactions with neurotransmitter metabolites using stable cell lines. Immunohistochemical analysis revealed that hOAT1 and hOAT3 are expressed in the cytoplasmic membrane and cytoplasm of human choroid plexus. Neurotransmitter metabolites, namely, 5-methoxyindole-3-acetic acid (5-MI-3-AA), homovanillic acid (HVA), vanilmandelic acid (VMA), 3,4-dihydroxyphenylacetic acid (DOPAC), 5-hydroxyindole-3-acetic acid (5-HI-3-AA), N-acetyl-5-hydroxytryptamine (NA-5-HTT), melatonin, 5-methoxytryptamine (5-MTT), 3,4-dihidroxymandelic acid (DHMA), 5-hydroxytryptophol, and 5-methoxytryptophol (5-MTP), but not methanephrine (MN), normethanephrine (NMN), and 3-methyltyramine (3-MT), at 2 mM, inhibited para-aminohippuric acid uptake mediated by hOAT1. On the other hand, melatonin, 5-MI-3-AA, NA-5-HTT, 5-MTT, 5-MTP, HVA, 5-HI-3-AA, VMA, DOPAC, 5-hydroxytryptophol, and MN, but not 3-MT, DHMA, and NMN, at 2 mM, inhibited estrone sulfate uptake mediated by hOAT3. Differences in the IC(50) values between hOAT1 and hOAT3 were observed for DHMA, DOPAC, HVA, 5-HI-3-AA, melatonin, 5-MI-3-AA, 5-MTP, 5-MTT, and VMA. HOAT1 and hOAT3 mediated the transport of VMA but not HVA and melatonin. These results suggest that hOAT1 and hOAT3 are involved in the efflux of various neurotransmitter metabolites from the cerebrospinal fluid to the blood across the choroid plexus.
Notes:
Suparat Khamdang, Michio Takeda, Ellappan Babu, Rie Noshiro, Maristela Lika Onozato, Akihiro Tojo, Atsushi Enomoto, Xiu-Lin Huang, Shinichi Narikawa, Naohiko Anzai, Pawinee Piyachaturawat, Hitoshi Endou (2003)  Interaction of human and rat organic anion transporter 2 with various cephalosporin antibiotics.   Eur J Pharmacol 465: 1-2. 1-7 Mar  
Abstract: Cephalosporin antibiotics are thought to be excreted into the urine via organic anion transporters (OATs) and OAT can mediate nephrotoxicity by cephalosporins, particularly by cephaloridine. The purpose of this study was to elucidate the interaction of human-OAT2 and rat-OAT2 with cephalosporin antibiotics using proximal tubule cells stably expressing human-OAT2 and rat-OAT2. Human-OAT2 is localized to the basolateral side of the proximal tubule, whereas rat-OAT2 is localized to the apical side of the proximal tubule. Cephalosporins tested were cephalothin, cefoperazone, cefazolin, ceftriaxone, cephaloridine, cefotaxime, cefadroxil and cefamandole. These cephalosporins dose-dependently inhibited organic anion uptake mediated by human-OAT2 and rat-OAT2. There was no species difference observed for the effects of OAT2 with cephalosporins between human and rat transporters. Kinetic analysis revealed that the inhibitory effects for human-OAT2 were competitive. Cephaloridine significantly decreased the viability of cells stably expressing human-OAT2, human-OAT1, human-OAT3 and human-OAT4. The decreased viability of cells stably expressing human-OAT1, human-OAT3 and human-OAT4 but not human-OAT2 was reversed by probenecid. In conclusion, human-OAT2 interacts with cephalosporins, and thus, human-OAT2 may mediate the uptake of cephalosporins on the basolateral side of the proximal tubule. The interaction of human-OAT2 with cephalosporins was the weakest among the basolateral human-OATs tested. In addition, it is suggested that human-OATs mediate cephaloridine-induced nephrotoxicity.
Notes:
Ellappan Babu, Yoshikatsu Kanai, Arthit Chairoungdua, Do Kyung Kim, Yuji Iribe, Sahatchai Tangtrongsup, Promsuk Jutabha, Yuewei Li, Nesar Ahmed, Shinichi Sakamoto, Naohiko Anzai, Seishi Nagamori, Hitoshi Endou (2003)  Identification of a novel system L amino acid transporter structurally distinct from heterodimeric amino acid transporters.   J Biol Chem 278: 44. 43838-43845 Oct  
Abstract: A cDNA that encodes a novel Na+-independent neutral amino acid transporter was isolated from FLC4 human hepatocarcinoma cells by expression cloning. When expressed in Xenopus oocytes, the encoded protein designated LAT3 (L-type amino acid transporter 3) transported neutral amino acids such as l-leucine, l-isoleucine, l-valine, and l-phenylalanine. The LAT3-mediated transport was Na+-independent and inhibited by 2-aminobicyclo[2.2.1]heptane-2-carboxylic acid, consistent with the properties of system L. Distinct from already known system L transporters LAT1 and LAT2, which form heterodimeric complex with 4F2 heavy chain, LAT3 was functional by itself in Xenopus oocytes. The deduced amino acid sequence of LAT3 was identical to the gene product of POV1 reported as a prostate cancer-up-regulated gene whose function was not determined, whereas it did not exhibit significant similarity to already identified transporters. The Eadie-Hofstee plots of LAT3-mediated transport were curvilinear, whereas the low affinity component is predominant at physiological plasma amino acid concentration. In addition to amino acid substrates, LAT3 recognized amino acid alcohols. The transport of l-leucine was electroneutral and mediated by a facilitated diffusion. In contrast, l-leucinol, l-valinol, and l-phenylalaninol, which have a net positive charge induced inward currents under voltage clamp, suggesting these compounds are transported by LAT3. LAT3-mediated transport was inhibited by the pretreatment with N-ethylmaleimide, consistent with the property of system L2 originally characterized in hepatocyte primary culture. Based on the substrate selectivity, affinity, and N-ethylmaleimide sensitivity, LAT3 is proposed to be a transporter subserving system L2. LAT3 should denote a new family of organic solute transporters.
Notes:
Promsuk Jutabha, Yoshikatsu Kanai, Makoto Hosoyamada, Arthit Chairoungdua, Do Kyung Kim, Yuji Iribe, Ellappan Babu, Ju Young Kim, Naohiko Anzai, Varanuj Chatsudthipong, Hitoshi Endou (2003)  Identification of a novel voltage-driven organic anion transporter present at apical membrane of renal proximal tubule.   J Biol Chem 278: 30. 27930-27938 Jul  
Abstract: A novel transport protein with the properties of voltage-driven organic anion transport was isolated from pig kidney cortex by expression cloning in Xenopus laevis oocytes. A cDNA library was constructed from size-fractionated poly(A)+ RNA and screened for p-aminohippurate (PAH) transport in high potassium medium. A 1856-base pair cDNA encoding a 467-amino acid peptide designated as OATV1 (voltage-driven organic anion transporter 1) was isolated. The predicted amino acid sequence of OATV1 exhibited 60-65% identity to those of human, rat, rabbit, and mouse sodium-dependent phosphate cotransporter type 1 (NPT1), although OATV1 did not transport phosphate. The homology of this transporter to known members of the organic anion transporter family (OAT family) was about 25-30%. OATV1-mediated PAH transport was affected by the changes in membrane potential. The transport was Na+-independent and enhanced at high concentrations of extracellular potassium and low concentrations of extracellular chloride. Under the voltage clamp condition, extracellularly applied PAH induced outward currents in oocytes expressing OATV1. The current showed steep voltage dependence, consistent with the voltage-driven transport of PAH by OATV1. The PAH transport was inhibited by various organic anions but not by organic cations, indicating the multispecific nature of OATV1 for anionic compounds. This transport protein is localized at the apical membrane of renal proximal tubule, consistent with the proposed localization of a voltage-driven organic anion transporter. Therefore, it is proposed that OATV1 plays an important role to excrete drugs, xenobiotics, and their metabolites driven by membrane voltage through the apical membrane of the tubular epithelial cells into the urine.
Notes:
Naohiko Anzai, Hiroki Miyazaki, Shinichi Sakamoto (2003)  Identification of intracellular regulatory protein for ion channel using yeast two-hybrid system   Nippon Yakurigaku Zasshi 122: 4. 331-337 Oct  
Abstract: Many proteins including ion channels mediate a variety of biological activities of cells through forming large molecular assemblies via protein-protein interactions. Hence, to understand the function of a particular protein, it is often useful to identify other proteins with which it associates. The two-hybrid system is a widely used yeast-based genetic approach for detecting protein-protein interactions and provides a powerful way of isolating ion channel-associated proteins in an in vivo setting without purifying both proteins. This system is thought to be one of the important techniques for "Proteome" analysis in the post-genomic era. In this article, we describe the yeast two-hybrid assay we have used to identify proteins binding to asic-sensing ion channel ASIC3 and urate transporter 1 (URAT1) and discuss some general and specific considerations about using ion channel proteins in the two-hybrid system.
Notes:
2002
Atsushi Enomoto, Michael F Wempe, Hiroki Tsuchida, Ho Jung Shin, Seok Ho Cha, Naohiko Anzai, Akiteru Goto, Atsuhiko Sakamoto, Toshimitsu Niwa, Yoshikatsu Kanai, M W Anders, Hitoshi Endou (2002)  Molecular identification of a novel carnitine transporter specific to human testis. Insights into the mechanism of carnitine recognition.   J Biol Chem 277: 39. 36262-36271 Sep  
Abstract: l-Carnitine is an essential component of mitochondrial fatty acid beta-oxidation and plays a pivotal role in the maturation of spermatozoa within the male reproductive tract. Epididymal plasma contains the highest levels of l-carnitine found in the human body, and initiation of sperm motility occurs in parallel to l-carnitine increase in the epididymal lumen. Using a specific carrier, epididymal epithelium secretes l-carnitine into the lumen by an active transport mechanism; however, the structure-activity relationship comprising the carnitine-permeation pathway is poorly understood. We discovered a novel carnitine transporter (CT2) specifically located in human testis. Analyzing the primary structure of CT2 revealed that it is phylogenetically located between the organic cation transporter (OCT/OCTN) and anion transporter (OAT) families. Hence, CT2 represents a novel transporter family. When expressed in Xenopus oocytes, CT2 mediates the high affinity transport of l-carnitine but does not accept mainstream OCT/OCTN cationic or OAT anionic substrates. We synthesized and tested various carnitine-related compounds and investigated the physicochemical properties of substrate recognition by semi-empirical computational chemistry. The data suggest that the quaternary ammonium cation bulkiness and relative hydrophobicity be the most important factors that trigger CT2-substrate interactions. Immunohistochemistry showed that the CT2 protein is located in the luminal membrane of epididymal epithelium and within the Sertoli cells of the testis. The identification of CT2 represents an interesting evolutionary link between OCT/OCTNs and OATs, as well as provides us with an important insight into the maturation of human spermatozoa.
Notes:
Naohiko Anzai, Emmanuel Deval, Lionel Schaefer, Valerie Friend, Michel Lazdunski, Eric Lingueglia (2002)  The multivalent PDZ domain-containing protein CIPP is a partner of acid-sensing ion channel 3 in sensory neurons.   J Biol Chem 277: 19. 16655-16661 May  
Abstract: Acid-sensing ion channels (ASICs) are cationic channels activated by extracellular pH. They are present in the brain, where they are thought to participate in signal transduction associated with local pH variations, and in sensory neurons, where they have been involved in pain perception associated with tissue acidosis and in mechanoperception. The ASIC3 subunit is mainly expressed in dorsal root ganglion neurons. Its expression is associated with a rapidly inactivating current followed by a slowly activating sustained current thought to be required for the tonic sensation of pain caused by acids. We report here the interaction of this channel subunit with the multivalent PDZ (PSD-95 Drosophila discs-large protein, Zonula occludens protein 1) domain-containing protein CIPP. This interaction requires the C-terminal region of ASIC3 and the fourth PDZ domain of CIPP. Co-expression of CIPP and ASIC3 in COS cells increases the maximal ASIC3 peak current density by a factor of 5 and slightly shifts the pH(0.5) for activation from pH 6.2 to pH 6.4. CIPP mRNA is found at a significant level in the same dorsal root ganglion neuronal cell population that expresses the ASIC3 subunit, i.e. mainly in the small nociceptive neurons. CIPP is thus a scaffolding protein that could both enhance the surface expression of ASIC3 and bring together ASIC3 and functionally related proteins in the membrane of sensory neurons.
Notes:
Do Kyung Kim, Yoshikatsu Kanai, Hye Won Choi, Sahatchai Tangtrongsup, Arthit Chairoungdua, Ellappan Babu, Kittipong Tachampa, Naohiko Anzai, Yuji Iribe, Hitoshi Endou (2002)  Characterization of the system L amino acid transporter in T24 human bladder carcinoma cells.   Biochim Biophys Acta 1565: 1. 112-121 Sep  
Abstract: System L is a major nutrient transport system responsible for the Na(+)-independent transport of large neutral amino acids including several essential amino acids. In malignant tumors, a system L transporter L-type amino acid transporter 1 (LAT1) is up-regulated to support tumor cell growth. LAT1 is also essential for the permeation of amino acids and amino acid-related drugs through the blood-brain barrier. To search for in vitro assay systems to examine the interaction of chemical compounds with LAT1, we have investigated the expression of system L transporters and the properties of [14C]L-leucine transport in T24 human bladder carcinoma cells. Northern blot, real-time quantitative PCR and immunofluorescence analyses have reveled that T24 cells express LAT1 in the plasma membrane together with its associating protein 4F2hc, whereas T24 cells do not express the other system L isoform LAT2. The uptake of [14C]L-leucine by T24 cells is Na(+)-independent and almost completely inhibited by system L selective inhibitor BCH. The profiles of the inhibition of [14C]L-leucine uptake by amino acids and amino acid-related compounds in T24 cells are comparable with those for the LAT1 expressed in Xenopus oocytes. The majority of [14C]L-leucine uptake is, therefore, mediated by LAT1 in T24 cells. Consistent with LAT1 in Xenopus oocytes, the efflux of preloaded [14C]L-leucine is induced by extracellularly applied substrates of LAT1 in T24 cells. This efflux measurement has been proven to be more sensitive than that in Xenopus oocytes, because triiodothyronine, thyroxine and melphalan were able to induce the efflux of preloaded [14C]L-leucine in T24 cells, which was not detected for Xenopus oocyte expression system. T24 cell is, therefore, proposed to be an excellent tool to examine the interaction of chemical compounds with LAT1.
Notes:
Suparat Khamdang, Michio Takeda, Rie Noshiro, Shinichi Narikawa, Atsushi Enomoto, Naohiko Anzai, Pawinee Piyachaturawat, Hitoshi Endou (2002)  Interactions of human organic anion transporters and human organic cation transporters with nonsteroidal anti-inflammatory drugs.   J Pharmacol Exp Ther 303: 2. 534-539 Nov  
Abstract: The purpose of this study was to elucidate the interactions of human organic anion transporters (hOATs) and human organic cation transporters (hOCTs) with nonsteroidal anti-inflammatory drugs (NSAIDs) using cells stably expressing hOATs and hOCTs. NSAIDs tested were acetaminophen, acetylsalicylate, salicylate, diclofenac, ibuprofen, indomethacin, ketoprofen, mefenamic acid, naproxen, piroxicam, phenacetin, and sulindac. These NSAIDs inhibited organic anion uptake mediated by hOAT1, hOAT2, hOAT3, and hOAT4. By comparing the IC(50) values of NSAIDs for hOATs, it was found that hOAT1 and hOAT3 exhibited higher affinity interactions with NSAIDs than did hOAT2 and hOAT4. HOAT1, hOAT2, hOAT3, and hOAT4 mediated the uptake of either ibuprofen, indomethacin, ketoprofen, or salicylate, but not acetylsalicylate. Although organic cation uptake mediated by hOCT1 and hOCT2 was also inhibited by some NSAIDs, hOCT1 and hOCT2 did not mediate the uptake of NSAIDs. In conclusion, hOATs and hOCTs interacted with various NSAIDs, whereas hOATs but not hOCTs mediated the transport of some of these NSAIDs. Considering the localization of hOATs, it was suggested that the interactions of hOATs with NSAIDs are associated with the pharmacokinetics and the induction of adverse reactions of NSAIDs.
Notes:
2001
N Anzai, Y Suzuki, M Nishikitani, I Izumida-Moriguchi, A Kokubo, K Kawahara (2001)  Development of renal potassium excretion capacity in the neonatal rat.   Jpn J Physiol 51: 6. 745-752 Dec  
Abstract: We investigated the capacity of newborn rats to excrete an acute potassium load to understand the development of a renal potassium excretion system. Three groups of the rats (7-14 d) were used to collect urine periodically over 6 h after oral infusion of potassium: control (no potassium loading) and low- and high-potassium-loaded rats. In the low-potassium-loaded group, infused with about 0.6 microEq of potassium chloride/g body wt., the rate of renal potassium excretion increased from 0.08 plus minus 0.02 (7 d) to 0.13 plus minus 0.02 (10 d) and 0.21 plus minus 0.03 (14 d) microEq/h/g body wt. The high-potassium-loaded rats (1.5-2.8 microEq/g body wt. potassium load) excreted potassium at a higher rate of 0.18 +/- 0.05 (7 d), 0.30 +/- 0.02 (10 d), and 0.45 +/- 0.10 (14 d) microEq/h/g body wt. They excreted 77% (7 d), 76% (10 d), and 95% (14 d) of the potassium load. These values were much larger than the rate of 0.026 microEq/h/g body wt. of the control rats and of 0.08 microEq/h/g body wt., a mean potassium excretion rate during development from 7 to 14 d calculated from the data in the previous study (Kanno T et al.: J. Pediatr. Gastr. Nutr. 24: 242-252, 1997). In the same period, serum potassium concentration in the newborn rats decreased significantly (p < 0.01) from 7.2 +/- 0.1 (7 d) to 6.7 +/- 0.1 mEq/l (14 d). All these results suggest that a renal potassium excretion system in the rat develops at least in the second week of life, and its capacity is high enough to excrete the daily potassium intake.
Notes:
1999
N Anzai, I Izumida, Y Kobayashi, K Kawahara (1999)  Roles of vasopressin and hypertonicity in basolateral Na/K/2Cl cotransporter expression in rat kidney inner medullary collecting duct cells.   Jpn J Physiol 49: 2. 201-206 Apr  
Abstract: The basolateral Na/K/2Cl cotransporter mRNA (rNKCC1) increased when the cultured kidney inner medullary collecting duct (IMCD) cells of rats were exposed to vasopressin (10(-8) M) and/or hypertonicity (500 mOsm/kgH2O). However, only hypertonicity was effective in increasing the expression of rNKCC1 protein.
Notes:
1998
N Anzai, K Kawahara (1998)  Renal compensation for body water loss during dehydration in neonatal rats.   Jpn J Physiol 48: 3. 181-187 Jun  
Abstract: The present study was designed to investigate whether the limited capacity for concentrating urine in neonatal rats is associated with an immature ability to regulate serum osmolality. During milk deprivation, the percent of reduction in body weight per 10 h (mean +/- SE) was 4.3 +/- 0.2, 3.7 +/- 0.1, 4.8 +/- 0.2, and 6.0 +/- 0.1% in 4-, 7-, 10-, and 14-d-old rats, respectively (n = 23-24, each age). The osmolality of urine increased to 718 +/- 12 (4 d), 741 +/- 28 (7 d), 792 +/- 20 (10 d), and 1,203 +/- 41 mosmol/kg H2O (14 d). Free-water absorption (TcH2O) promptly increased after deprivation of milk: It significantly increased from 2.3 +/- 0.3 (0-4 h) to 3.4 +/- 0.1 (4-7 h) (4 d), from 3.1 +/- 0.3 to 4.1 +/- 0.3 (7 d), from 3. 6 +/- 0.4 to 5.2 +/- 0.3 (10 d), and from 5.0 +/- 0.4 to 7.9 +/- 0.7 microliter/min/100 g (14 d). The raised values were maintained at the later period of dehydration. Thus serum osmolality was unchanged throughout dehydration: 287 +/- 1.0 (7 d) and 292 +/- 0.9 mosmol/kg H2O (14 d). On the other hand, the level of serum sodium concentration slightly but significantly increased (r = 0.61) when the body weight reduction was higher than 5% of the control (14-d-old rats). These results indicate that neonatal rats of 4-14 d control their serum osmolality by reabsorbing free water in the kidney during the 10 or 12 h of milk deprivation.
Notes:
1997
K Kawahara, N Anzai (1997)  Potassium transport and potassium channels in the kidney tubules.   Jpn J Physiol 47: 1. 1-10 Feb  
Abstract: A variety of K+ channels have been identified using electrophysiological techniques including the patch-clamp method. The types of channels include inwardly-rectifying, ATP-dependent, Ca(2+)-dependent, voltage-gated, and so on. Some of them have been cloned by expression and/or PCR cloning techniques, which give us their amino-acid sequences and molecular topology in the cell membrane. Immunohistochemical studies have shown the proteins (channels) to be localized to the luminal and/or basolateral membranes of a certain nephron segment. However, the relationships of these proteins with the ionic channels identified functionally must be examined by their reconstitution in cell-free systems (lipid bilayer membrane) and/or their expression in cells lacking native K+ channels. Much more care should also be taken to avoid artifacts due to channel-inducing factors (CHIF). Structure-function studies at the molecular level will advance our knowledge of the renal K+ channels and provide us with a further understanding of the role of the kidney in K+ homeostasis.
Notes:
1995
J Hori, U Shiro, N Anzai, K Itoh, M Ogawa, M Ohto, Y Wakashin, M Wakashin (1995)  Primary culture of proximal tubular cells (PTC) from normal mouse kidney as an in vitro model to study mechanisms of development of tubulointerstitial nephritis. Induction of ICAM-1 in PTC by antigen-primed lymphocytes   Nippon Jinzo Gakkai Shi 37: 3. 157-164 Mar  
Abstract: Cultured PTC clearly expressed intercellular adhesion molecule-1 (ICAM-1) under the influence of tubular basement membrane antigen (TBM)-primed lymphocytes. These TBM-primed lymphocytes also demonstrated a high cytotoxic activity against cultured PTC. A pure preparation of isolated PTC from BALB/c mouse kidney was brought into primary culture. PTC was prepared by the method of Boogaard PJ et al, and our modification. Briefly, kidney was perfused with buffer containing 0.08% (w/v) collagenase. The cortical tissue was then filtered through nylon-gauze. Viable PTC were separated from other materials by isopycnic centrifugation on a discontinuous Nycodenz gradient. The confluent monolayer of PTC showed a typical epithelial morphology with cobblestone-like cells in the center of the cell-islands. Typical dome formation was observed in PTC cultures. These cells also strongly expressed gamma-glutamyl transpeptitase activity. Coculture of PTC with syngeneic lymphocytes primed with TBM antigen induced ICAM-1 expression in PTC. The TBM-primed lymphocytes had a cytotoxic activity without complement. However, neither virgin lymphocytes nor liver antigen-primed lymphocytes had cytotoxic activity. This simple syngeneic experimental model may allow us further molecular biological examination of renal tubulointerstitial diseases.
Notes:
Powered by PublicationsList.org.