Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yorihisa Tanaka is active.

Publication


Featured researches published by Yorihisa Tanaka.


Toxicology Letters | 2000

Roles of selenium in endotoxin-induced lipid peroxidation in the rats liver and in nitric oxide production in J774A.1 cells

Shuhei Sakaguchi; Yukisumi Iizuka; Shinobu Furusawa; Yorihisa Tanaka; Motoaki Takayanagi; Yoshio Takayanagi

We examined the role of selenium (Se) in the mechanism of oxidative stress caused by endotoxin by feeding rats deficient a diet in this element. In rats fed the Se-deficient diet (concentration of Se, less than 0.027 microg g(-1)) for 10 weeks, Se level and glutathione peroxidase (GSH-Px) activity in the liver were about 47 and 43% lower, respectively, than those in rats fed a Se-adequate diet (Se, 0.2 microg g(-1)). Rat fed the Se-deficient diet and given endotoxin (6 mg kg(-1), i.p.) showed a mortality rates of about 43% at 18 h. Nevertheless, no lethality was observed with endotoxin (4 mg kg(-1), i.p.) challenge. Levels of serum lactate dehydrogenase and acid phosphatase leakage were significantly higher in Se-deficient rats than those in Se-adequate diet 18 h after endotoxin (4 mg kg(-1), i. p.) challenge. Superoxide anion generation and lipid peroxide formation in the liver of Se-deficient rat were markedly increased 18 h after endotoxin (4 mg kg(-1), i.p.) injection compared with those in the endotoxin/Se-adequate diet group, whereas non-protein sulfhydryl level in the liver after administration of endotoxin to Se-deficient rats was lower than that in Se-adequate rats treated with endotoxin. We investigated whether Se can suppress nitric oxide (NO) generation and cytotoxicity in endotoxin-treated J774A.1 cells. Treatment with Se (10(-6) M) markedly inhibited endotoxin (0.1 microg ml(-1))-induced NO production in J774A.1 cells. Se induced an increased activity of GSH-Px in cells after 24 h of incubation, suggesting that the preventive effect of Se on NO production in endotoxemia is due to the induction of Se-GSH-Px activity. However, Se did not affect endotoxin-induced cytotoxicity in J774A.1 cells. These findings suggested that the oxidative stress caused by endotoxin may be due, at least in part, to changes in Se regulation during endotoxemia.


Xenobiotica | 2007

Lack of dimer formation ability in rat strains with low aldehyde oxidase activity

Kunio Itoh; H. Maruyama; Mayuko Adachi; Kouichi Hoshino; Nobuaki Watanabe; Yorihisa Tanaka

Aldehyde oxidase (AO) is a homodimer with a molecular weight of 300 kDa. To clarify the reasons for the well-known differences in rat strains, we set out to study the relationship between AO activity and the expression levels of its dimer. AO-catalyzed 2-oxidation activity of (S)-RS-8359 was measured in liver cytosols from ten rat strains. The expression levels of AO dimeric protein were evaluated by the native-PAGE/Western blot. Rat strains with low AO activity showed only a monomer, whereas strains with high activity overwhelmingly exhibited a dimer. Exceptionally, one strain in the high AO activity group displayed complex mixed expression patterns of low and high AO activity groups. However, there was a good relationship between AO activity and the expression levels of a dimer, but not of a monomer. The results suggest that rat strains with low AO activity lack the ability to produce a dimer necessary for catalytic activity, and AO differences in rat strains should be discussed in terms of the expression levels of the dimer itself.


Inflammation Research | 2000

L-Histidine decarboxylase protein and activity in rat brain microvascular endothelial cells.

J. Yamakami; Eiichi Sakurai; Atsuo Kuramasu; Kazuhiko Yanai; Takehiko Watanabe; Yorihisa Tanaka

Abstract.Objective and Design: L-Histidine decarboxylase (HDC) is the primary enzyme regulating histamine biosynthesis. This study was carried out to examine whether the cultured rat brain microvascular endothelial cells (BMEC), which constitute the blood-brain barrier (BBB), have the ability to form histamine, and whether HDC mRNA is expressed in rat BMEC.¶Material: Male, 3-week-old Wistar rats were used. For in vitro studies, rat BMEC were isolated from rat brains, and subculture cells were grown on collagen-coated culture flask and slide.¶Methods: HDC assay, immunofluorescence analysis and expression of HDC mRNA by RT-PCR were performed in rat BMEC.¶Results: The HDC activity of the BMEC was estimated to be 0.14 ± 0.05 p mol/min/mg protein. This activity was completely inhibited by (S)-α-fluoromethylhistidine, a specific inhibitor of HDC. Using a polyclonal anti HDC antibody and immunofluorescence microscopy, we confirmed the presence of HDC protein in rat BMEC. RT-PCR also showed the expression of HDC mRNA in rat BMEC.¶Conculsions: L-Histidine uptaken by rat BMEC was shown to be converted to histamine, suggesting that HDC plays an important role in BBB.


Xenobiotica | 2005

Stereospecific oxidation of the (S)-enantiomer of RS-8359, a selective and reversible monoamine oxidase A (MAO-A) inhibitor, by aldehyde oxidase

Kunio Itoh; M. Yamamura; S. Muramatsu; Kouichi Hoshino; Akiko Masubuchi; Takamitsu Sasaki; Yorihisa Tanaka

In a previous paper by the authors on RS-8359, a new selective and reversible monoamine oxidase A (MAO-A) inhibitor, it was reported that the (S)-enantiomer of RS-8359 is rapidly eliminated from rats, monkeys and humans as a result of the formation of a 2-oxidative metabolite. The present study investigates the properties of the enzyme responsible for the 2-oxidation of RS-8359. Subcellular localization, cofactor requirement and the inhibitory effects of typical compounds were studied using rat liver preparations. In addition, the enzyme was purified from rat liver cytosol for further characterization. The enzyme activity was localized in the cytosolic fraction without the need for any cofactor and was extensively inhibited by menadione, chlorpromazine and quinacrine. The purified enzyme was also a homodimer with a monomeric molecular weight of 140 kDa and it had an A280/A450 ratio of 5.1 in the absorption spectrum. The results suggest that the enzyme responsible for the biotransformation of RS-8359 to give the 2-keto derivative is aldehyde oxidase (EC 1.2.3.1). The reaction of aldehyde oxidase is highly stereoselective for the (S)-configuration of RS-8359 and the (9R)-configuration of cinchona alkaloids.


Journal of Pharmacy and Pharmacology | 2006

Metabolism of nicotine in rat lung microvascular endothelial cells

Yoshinori Ochiai; Kunio Itoh; Yorihisa Tanaka; Eiichi Sakurai; Akio Nomura

The aim of this study was to examine whether cultured rat lung microvascular endothelial cells (LMECs), which constitute the gas‐blood barrier, have the ability to metabolize nicotine. Nicotine was biotransformed to cotinine and nicotine N′‐oxide by cytochrome 450 (CYP) and flavin‐containing monooxyganase (FMO), respectively, in rat LMECs. The intrinsic clearance (Vmax1/Km1) for the cotinine formation was about 20 times as high as that for the trans‐nicotine N′‐oxide formation in the low‐Km phase, indicating that oxidation by CYP was much higher than that by FMO. On the other hand, as shown in Eadie‐Hofstee plots, the formation of cis‐nicotine N′‐oxide was monophasic, whereas the plot for the trans‐nicotine N′‐oxide formation was clearly biphasic. These results suggest that nicotine N′‐oxide was stereoselectively metabolized to cis and trans forms. However, in the high‐Km phase there was no significant difference in N′‐oxidation between the cis and trans forms. Moreover, we suggest that CYP2C11 and CYP3A2 are key players in the metabolism to cotinine of nicotine in rat LMECs using the respective enzyme inhibitors (tranylcypromine and troleandomycine). On the other hand, methimazole (5 μm) caused 73 and 45% decreases in the formation of N′‐oxides of cis‐ and trans‐ enantiomers, respectively, demonstrating the presence of FMO in rat LMECs. These results suggest that rat LMEC enzymes can convert substrates of exogenous origin such as nicotine for detoxication, indicating LMECs are an important barrier for metabolic products, besides hepatic cells.


Drug Metabolism and Disposition | 2007

Genetic polymorphism of aldehyde oxidase in donryu rats

Kunio Itoh; Akiko Masubuchi; Takamitsu Sasaki; Mayuko Adachi; Nobuaki Watanabe; Kiyoshi Nagata; Yasushi Yamazoe; Masahiro Hiratsuka; Michinao Mizugaki; Yorihisa Tanaka

One of major metabolic pathways of [(±)-4-(4-cyanoanilino)-5,6-dihydro-7-hydroxy-7H-cyclopenta[d]-pyrimidine] (RS-8359), a selective and reversible monoamine oxidase type A inhibitor, is the aldehyde oxidase-catalyzed 2-hydroxylation at the pyrimidine ring. Donryu rats showed a dimorphic pattern for the 2-oxidation activity with about 20- to 40-fold variations in the Vmax/Km values between a low and a high activity group. The rats were classified as extensive metabolizers (EM) and poor metabolizers (PM) of RS-8359, of which ratios were approximately 1:1. One rat among the EM rats of each sex showed extremely high activity, and they were referred to as ultrarapid metabolizers. There was no significant difference in the expression levels of mRNA of aldehyde oxidase between the EM and PM rats. Analysis of nucleotide sequences showed four substitutions, of which the substitutions at 377G>A and 2604C>T caused 110Gly-Ser and 852Ala-Val amino acid changes, respectively. Amino acid residue 110 is located very near the second Fe-S center of aldehyde oxidase. Its change from nonchiral Gly to chiral Ser may result in a conformational change of aldehyde oxidase protein with the shift of isoelectric point value from 5.0 in the EM rats to 6.2 in the PM rats. The 110Gly-Ser amino acid substitution (377G>A) may be primarily responsible for the variations of aldehyde oxidase activity observed in Donryu rats, in addition to the difference of expression levels of aldehyde oxidase protein. If a new drug candidate is primarily metabolized by aldehyde oxidase, attention should be given to using a rat strain with high aldehyde oxidase activity and small individual variation.


Drug Metabolism and Disposition | 2007

Lack of Formation of Aldehyde Oxidase Dimer Possibly Due to 377G>A Nucleotide Substitution

Kunio Itoh; Hiroaki Maruyama; Mayuko Adachi; Kouichi Hoshino; Nobuaki Watanabe; Yorihisa Tanaka

In addition to the many articles reporting on the marked differences in species and large differences in rat strains in response to aldehyde oxidase (AO), individual differences in some rat strains have also been reported. However, little has been clarified about any related molecular biological mechanisms. We previously revealed that nucleotide substitutions of 377G>A and 2604C>T in the AO gene might be responsible for individual differences in AO activity in Donryu strain rats. By using native polyacrylamide gel electrophoresis/Western blotting in this study, the lack of formation of the AO dimer protein, which is essential for catalytic activity, was shown in poor metabolizer Donryu rats, and this could be a major reason for the individual differences. Rat strain differences were also verified from the same perspectives of nucleotide substitutions and expression levels of a dimer protein. Rat strains with high AO activity showed nucleotide sequences of (377G, 2604C) and a dimer protein. In the case of those with low AO activity, the nucleotide at position 2604 was fixed at T, but varied at position 377, such as G, G/A, and A. An AO dimer was detected in the liver cytosols of rat strains with (377G, 2604T), whereas a monomer was observed in those with (377A, 2604T). These results suggest that the lack of formation of a dimer protein leading to loss of catalytic activity might be due to 377G>A nucleotide substitution. Individual and strain differences in AO activity in rats could be explained by this 377G>A substitution, at least in the rat strains used in this study.


Journal of Chromatography B: Biomedical Sciences and Applications | 1993

Stereoselective determination of the active metabolites of a new anti-inflammatory agent (CS-670) in human and rat plasma using antibody-mediated extraction and high-performance liquid chromatography

Wataru Takasaki; Masato Asami; Shigeki Muramatsu; Ryozo Hayashi; Yorihisa Tanaka; Kiyoshi Kawabata; Kazuko Hoshiyama

The main metabolites of (+-)-2-[4-(2-oxocyclohexylidenemethyl)phenyl]propionic acid (CS-670), a new pro-drug anti-inflammatory agent of the 2-arylpropionic acid type, have one or two chiral centres arising from reduction of the oxocyclohexylidene moiety in addition to an original chiral centre in the propionic acid moiety. To determine these metabolites stereoselectively, antibody-mediated extraction was investigated as a stereoselective clean-up method prior to chiral HPLC. Immunoglobulin G, which recognizes each stereoisomeric cyclohexanol moiety, was coupled to cyanogen bromide-activated Sepharose 4B to prepare re-usable immobilized antibody, and its specificity was improved by examination of a washing process after charging of samples. Plasma extracted with the immobilized antibody column was derivatized with a chiral reagent to separate the enantiomers of the propionic acid moiety by HPLC. This newly developed analytical method clarified the stereoselective biotransformation of the pro-drug to pharmacologically active forms in humans and rats related to reduction of the oxocyclohexylidene moiety and chiral inversion in the propionic acid moiety.


Toxicology Letters | 2008

Involvement of cytochrome P450-mediated metabolism in tienilic acid hepatotoxicity in rats.

Takayoshi Nishiya; Michiyuki Kato; Takami Suzuki; Chikako Maru; Hiroko Kataoka; Chiharu Hattori; Kazuhiko Mori; Toshimasa Jindo; Yorihisa Tanaka; Sunao Manabe

Tienilic acid is reported to be converted into electrophilic metabolites by cytochrome P450 (CYP) in vitro. In vivo, however, the metabolites have not been detected and their effect on liver function is unknown. We previously demonstrated that tienilic acid decreased the GSH level and upregulated genes responsive to oxidative/electrophilic stresses, such as heme oxygenase-1 (Ho-1), glutamate-cysteine ligase modifier subunit (Gclm) and NAD(P)H dehydrogenase quinone 1 (Nqo1), in rat liver, as well as inducing hepatotoxicity by co-treatment with the glutathione biosynthesis inhibitor l-buthionine-(S,R)-sulfoximine (BSO). In this study, for the first time, we identified a glutathione-tienilic acid adduct, a stable conjugate of putative electrophilic metabolites with glutathione (GSH), in the bile of rats given a single oral dose of tienilic acid (300mg/kg). Furthermore, a tienilic acid-induced decrease in the GSH level and upregulation of Ho-1, Gclm and Nqo1 were completely blocked by pretreatment with the CYP inhibitor 1-aminobenzotriazole (ABT, 66mg/kg, i.p.). The increase in the serum ALT level and hepatocyte necrosis resulting from the combined dosing of BSO and tienilic acid was prevented by ABT, despite a low hepatic GSH level. These findings suggest that the electrophilic metabolites of tienilic acid produced by CYP induce electrophilic/oxidative stresses in the rat liver and this contributes to the hepatotoxicity of tienilic acid under impaired GSH biosynthesis.


Drug Metabolism and Disposition | 2010

A Single Amino Acid Substitution Confers High Cinchonidine Oxidation Activity Comparable with That of Rabbit to Monkey Aldehyde Oxidase 1

Kensuke Fukiya; Kunio Itoh; Satoshi Yamaguchi; Akiko Kishiba; Mayuko Adachi; Nobuaki Watanabe; Yorihisa Tanaka

Aldehyde oxidase 1 (AOX1) is a major member of the xanthine oxidase family belonging to the class of complex molybdo-flavoenzymes and plays an important role in the nucleophilic oxidation of N-heterocyclic aromatic compounds and various aldehydes. The enzyme has been well known to show remarkable species differences. Comparing the rabbit and monkey enzymes, the former showed extremely high activity toward cinchonidine and methotrexate, but the latter exhibited only marginal activities. In contrast, monkey had several times greater activity than did rabbit toward zonisamide and (+)-4-(4-cyanoanilino)-5,6-dihydro-7-hydroxy-7H-cyclopenta[d]-pyrimidine [(S)-RS-8359]. In this report, we tried to confer high cinchonidine oxidation activity comparable with that of rabbit AOX1 to monkey AOX1. The chimera proteins prepared by restriction enzyme digestion and recombination methods between monkey and rabbit AOX1s indicated that the sequences from Asn993 to Ala1088 of rabbit AOX1 are essential for the activity. The kinetic parameters were then measured using monkey AOX1 mutants prepared by site-directed mutagenesis. The monkey V1085A mutant acquired the high cinchonidine oxidation activity. Inversely, the reciprocal rabbit A1081V mutant lost the activity entirely: amino acid 1081 of rabbit AOX1 corresponding to amino acid 1085 of monkey AOX1. Thus, cinchonidine oxidation activity was drastically changed by mutation of a single residue in AOX1. However, this might be true for bulky substrates such as cinchonidine but not for small substrates. The mechanism of substrate-dependent species differences in AOX1 activity toward bulky substrates is discussed.

Collaboration


Dive into the Yorihisa Tanaka's collaboration.

Top Co-Authors

Avatar

Kunio Itoh

Tohoku Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mayuko Adachi

Tohoku Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eiichi Sakurai

Tohoku Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Kouichi Hoshino

Tohoku Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Akiko Masubuchi

Tohoku Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tasuku Asakawa

Tohoku Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge