Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Low Urin Tract Symptoms ; 10(2): 116-121, 2018 May.
Article in English | MEDLINE | ID: mdl-29664239

ABSTRACT

OBJECTIVES: In recent years, some patients have been unresponsive to anticholinergics used in the treatment of pollakisuria/urinary incontinence. It has been suggested that propiverine hydrochloride, which has not only anticholinergic activity, but also calcium antagonistic activity, may be useful in poor responders to other anticholinergics. In this study, a specific drug use-results survey was conducted in poor responders to other anticholinergics to evaluate the usefulness of propiverine hydrochloride. METHODS: In this survey, propiverine hydrochloride was administered for 12 weeks to poor responders to previous anticholinergics, and its usefulness was evaluated by the overactive bladder symptom score (OABSS). RESULTS: A total of 3851 subjects at 680 institutions were enrolled in the survey. Of the 3624 subjects included in the safety evaluation (male 1899, female 1725, mean age 73.4 years), 2932 were included in the efficacy evaluation (male 1610, female 1322, mean age 73.8 years). Propiverine hydrochloride significantly improved the OABSS without any safety concerns in poor responders to previous anticholinergics (OABSS, 8.22 at baseline, 6.50 at Week 4, 5.87 at Week 8, and 5.57 at Week 12, P < 0.001). CONCLUSIONS: The present findings indicate that propiverine hydrochloride may be a useful therapeutic option for poor responders to previous anticholinergics.


Subject(s)
Benzilates/administration & dosage , Muscarinic Antagonists/administration & dosage , Urinary Bladder, Overactive/drug therapy , Urological Agents/administration & dosage , Aged , Benzilates/adverse effects , Drug Administration Schedule , Drug Substitution , Female , Humans , Male , Medication Adherence , Muscarinic Antagonists/adverse effects , Treatment Outcome , Urinary Bladder, Neurogenic/drug therapy , Urinary Incontinence/drug therapy , Urological Agents/adverse effects
2.
Yakugaku Zasshi ; 136(9): 1319-23, 2016.
Article in Japanese | MEDLINE | ID: mdl-27592835

ABSTRACT

We set students' learning goal of basic life support (BLS) education at "being able to describe all the steps of BLS in an appropriate order", and objectively analyzed the appropriateness of the learning goal we set and educational effects of lecture contents. Before delivering a lecture, we provided students with an assignment which asked them to "Describe the steps of BLS in an appropriate order", and investigated students' levels of acquiring knowledge on BLS. As the results, the majority of students failed to perform this assignment. Since many students did not understand the process of BLS correctly, the learning goal was considered appropriate in the sense of promoting students' understanding of BLS. We also investigated whether the contents of BLS education was effective to achieve the learning goal. We provided students with the same assignment after the lecture, and the results showed that most students successfully performed the assignment. Furthermore, the time required for students to recall the whole process of BLS was significantly reduced after receiving the lecture, showing that the BLS lecture was effective in improving students' "ability to act to save lives".


Subject(s)
Education, Pharmacy , Health Knowledge, Attitudes, Practice , Life Support Care , Students, Pharmacy/psychology , Education, Pharmacy/methods , Humans
3.
Drug Metab Dispos ; 38(2): 302-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19910515

ABSTRACT

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.


Subject(s)
Aldehyde Oxidase/genetics , Aldehyde Oxidase/metabolism , Amino Acid Substitution , Cinchona Alkaloids/metabolism , Aldehyde Oxidase/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Macaca fascicularis , Models, Structural , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nitriles/metabolism , Oxidation-Reduction , Plasmids , Pyrimidines/metabolism , Rabbits , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Species Specificity , Substrate Specificity
4.
Yakugaku Zasshi ; 129(12): 1487-93, 2009 Dec.
Article in Japanese | MEDLINE | ID: mdl-19952527

ABSTRACT

Together with xanthine oxidase, aldehyde oxidase (AO) is a major member of a relatively small family of molybdenum hydroxylases. Both enzymes are homodimers with a subunit molecular weight of about 150 kDa and exhibit catalytic activity only as a dimer. An AO subunit contains a molybdopterin cofactor, an FAD and two different 2Fe-2S redox centers. The enzyme catalyzes oxidation of a wide range of endogenous and exogenous aldehydes and N-heterocyclic aromatic compounds. N-heterocycle-containing drugs such as methotrexate, 6-mercaptopurine, cinchona alkaloids and famciclovir are oxidized by this enzyme. Marked species differences have been well documented for the AO-catalyzed metabolism of drugs including methotrexate and famciclovir. In addition, a large rat strain variation has also been demonstrated in the oxidation activity of benzaldehyde and methotrexate. Marked differences in species, large differences in rat strains and individual differences in AO activities in some rat strains have been reported. However, little has been elucidated about any related molecular biological mechanisms. We examined the mechanism of individual variations and strain difference of rat AO using the technology of molecular biology. Our recent studies regarding the inter- and intra-difference of AO activities in rats are described.


Subject(s)
Aldehyde Oxidase/genetics , Aldehyde Oxidase/metabolism , Polymorphism, Genetic , Rats, Inbred Strains/genetics , Aldehyde Oxidase/physiology , Animals , Cytosol/enzymology , DNA, Complementary , Gene Frequency , Liver/enzymology , Rats
5.
Biol Pharm Bull ; 32(2): 190-4, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19182374

ABSTRACT

Selenium deficiency has been reported to result in an extraordinary decrease of glutathione peroxidase (GSH-Px) and, reversely, an increase of detoxifying enzymes such as glutathione-S-transferase (GST), uridine-5'-diphosphate glucuronosyltransferase (UGT), nicotinamide-dependent quinine oxidoreductase (NQO1; DT-diaphorase), and epoxide hydrolase without significantly affecting cytochrome P450 activity. However, little is known about the effects on aldehyde oxidase 1 (AOX1) activity towards various kinds of aldehydes and N-heterocyclic aromatic compounds. The aim of this study is to clarify the effects of selenium deficiency on AOX1 in rats. As expected, selenium deficiency was confirmed by the extremely low activity of GSH-Px along with the increased activities of GST and DT-diaphorase. AOX1 activity towards vanillin and (S)-RS-8359 was increased by selenium deficiency, and that corresponded to an increase of AOX1 protein level but not to a decreased AOX1 mRNA level. It has been documented that the assembly of the catalytically active holoenzyme forms of the molybdo-flavoenzyme family is very complex and is controlled through transcriptional and translational events by many gene products. In addition, selenium deficiency has been known to cause oxidative stress that leads to an increase of AOX1 activity. Furthermore, aldehyde oxidase homolog 1 (AOH1) with properties similar to AOX1 is present in rodent liver. All the reports suggest that the mechanisms by which selenium deficiency increases AOX1 activity are highly complicated and investigated from different points of view.


Subject(s)
Aldehyde Oxidase/metabolism , Antioxidants/metabolism , Selenium/deficiency , Animals , Antioxidants/pharmacology , Benzaldehydes/pharmacology , Blotting, Western , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Glucuronosyltransferase/metabolism , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Indicators and Reagents , Liver/cytology , Liver/drug effects , Liver/metabolism , Male , Nitriles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Selenium/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
6.
Biol Pharm Bull ; 32(1): 31-5, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19122276

ABSTRACT

Aldehyde oxidase (AO) is a homodimer with a subunit molecular mass of approximately 150 kDa. Each subunit consists of about 20 kDa 2Fe-2S cluster domain storing reducing equivalents, about 40 kDa flavine adenine dinucleotide (FAD) domain and about 85 kDa molybdenum cofactor (MoCo) domain containing a substrate binding site. In order to clarify the properties of each domain, especially substrate binding domain, chimeric cDNAs were constructed by mutual exchange of 2Fe-2S/FAD and MoCo domains between monkey and rat. Chimeric monkey/rat AO was referred to one with monkey type 2Fe-2S/FAD domains and a rat type MoCo domain. Rat/monkey AO was vice versa. AO-catalyzed 2-oxidation activities of (S)-RS-8359 were measured using the expressed enzyme in Escherichia coli. Substrate inhibition was seen in rat AO and chimeric monkey/rat AO, but not in monkey AO and chimeric rat/monkey AO, suggesting that the phenomenon might be dependent on the natures of MoCo domain of rat. A biphasic Eadie-Hofstee profile was observed in monkey AO and chimeric rat/monkey AO, but not rat AO and chimeric monkey/rat AO, indicating that the biphasic profile might be related to the properties of MoCo domain of monkey. Two-fold greater V(max) values were observed in monkey AO than in chimeric rat/monkey AO, and in chimeric monkey/rat AO than in rat AO, suggesting that monkey has the more effective electron transfer system than rat. Thus, the use of chimeric enzymes revealed that 2Fe-2S/FAD and MoCo domains affect the velocity and the quantitative profiles of AO-catalyzed (S)-RS-8359 2-oxidation, respectively.


Subject(s)
Aldehyde Oxidase , Flavin-Adenine Dinucleotide , Gene Expression , Aldehyde Oxidase/chemistry , Aldehyde Oxidase/genetics , Aldehyde Oxidase/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Biochemical Phenomena , Cloning, Molecular , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/genetics , Flavin-Adenine Dinucleotide/metabolism , Haplorhini , Molecular Sequence Data , Rats , Recombinant Proteins
7.
Biol Pharm Bull ; 31(3): 380-5, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18310896

ABSTRACT

We previously demonstrated the existence of a minor 130 kDa subunit in the sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE)/Western blot analysis of monkey liver cytosol and expressed monkey aldehyde oxidase (AO) in Escherichia coli. In contrast, the 130 kDa subunit was not observed in rat AO. In the current study, the properties of the 130 kDa subunit were investigated from the viewpoint of species differences in the presence of the subunit and AO activity. Monkey AO with His-tag at the N- and C-terminus were expressed, and were immunoanalyzed with anti-AO and anti-His-tag antisera. The results revealed that the minor 130 kDa subunit was produced by cleavage at the N-terminal side of the 150 kDa subunit. The cleavage point was shown to be located between 188Leu and 189Pro of 150 kDa AO subunit by the Edman degradation method. The two amino acids related to the cleavage are contained in the linkage between the 2Fe-2S and FAD domains in AO of human and monkey, but not in AO of rat and mouse. As a fact, the 130 kDa subunit was observed in AO of human and monkey, but not in AO of rat and mouse, suggesting the two amino acids might be one reason of a species difference in the formation of the 130 kDa subunit. However, the existence of the 130 kDa subunit is not associated with the species differences in AO activity, because the cleavage results in the loss of 2Fe-2S cluster domain essential for exertion of AO activity.


Subject(s)
Aldehyde Oxidase , Aldehyde Oxidase/chemistry , Aldehyde Oxidase/genetics , Aldehyde Oxidase/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cloning, Molecular , Cytosol/enzymology , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Haplorhini , Humans , Liver/enzymology , Molecular Sequence Data , Protein Conformation , Protein Subunits , Species Specificity
8.
J Biochem Mol Biol ; 40(6): 1021-7, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-18047800

ABSTRACT

We demonstrated the genetic polymorphism of aldehyde oxidase (AO) in Donryu strain rats: the ultrarapid metabolizer (UM) with nucleotide mutation of (377G, 2604C) coding for amino acid substitution of (110Gly, 852Val), extensive metabolizer (EM) with (377G/A, 2604C/T) coding for (110Gly/Ser, 852Val/Ala), and poor metabolizer (PM) with (377A, 2604T) coding for (110Ser, 852Ala), respectively. The results suggested that 377G > A and/or 2604C > T should be responsible for the genetic polymorphism. In this study, we constructed an E. coli expression system of four types of AO cDNA including Mut-1 with (377G, 2604T) and Mut-2 with (377A, 2604C) as well as naturally existing nucleotide sequences of UM and PM in order to clarify which one is responsible for the polymorphism. Mut-1 and Mut-2 showed almost the same high and low activity as that of the UM and PM groups, respectively. Thus, the expression study of mutant AO cDNA directly revealed that the nucleotide substitution of 377G > A, but not that of 2604C > T, will play a critical role in the genetic polymorphism of AO in Donryu strain rats. The reason amino acid substitution will cause genetic polymorphism in AO activity was discussed.


Subject(s)
Aldehyde Oxidase/genetics , DNA, Complementary/genetics , Polymorphism, Single Nucleotide , Aldehyde Oxidase/metabolism , Amino Acid Substitution , Animals , Base Sequence , Escherichia coli/genetics , Gene Expression , Kinetics , Mutation , Plasmids/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity
9.
Drug Metab Dispos ; 35(10): 1860-4, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17639027

ABSTRACT

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.


Subject(s)
Aldehyde Oxidase/genetics , Liver/enzymology , Aldehyde Oxidase/chemistry , Aldehyde Oxidase/metabolism , Animals , Base Sequence , Cytosol/enzymology , Dimerization , Male , Nucleotides/genetics , Polymorphism, Genetic , Rats , Rats, Inbred Strains , Sequence Analysis, DNA , Species Specificity
10.
Biol Pharm Bull ; 30(7): 1191-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17603152

ABSTRACT

In this study, we investigated the properties of monkey liver aldehyde oxidase directed toward the clarification of species differences. The aldehyde oxidase preparation purified from male cynomolgus monkey liver cytosol showed a major 150 kDa Coomassie brilliant blue (CBB)-stained band together with a minor 130 kDa band using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Both bands were identified as being aldehyde oxidase by a database search of the MS data obtained with nano-liquid chromatography, quardrupole time of flight, mass spectrometry (nano-LC Q/TOF MS). Based on the sequence coverage, the 130 kDa protein was presumed to be deficient in 20-30 kDa mass from the N-terminus. Full male cynomolgus monkey aldehyde oxidase cDNA was cloned and sequenced with the four degenerate primers designed by considering the peptide sequences containing the amino acids specific for monkey aldehyde oxidase. The deduced amino acid sequences had 96% amino acid identity with those of human enzyme. The aldehyde oxidase expressed in Escherichia coli also exhibited two immunoreactive bands on SDS-PAGE/Western blot analysis. Further, the biphasic pattern was observed for Eadie-Hofstee plots of the (S)-enantiospecific 2-oxidation activity of RS-8359 with the expressed and cytosolic monkey liver aldehyde oxidase. The results suggested that two forms of aldehyde oxidase in monkey were the expression products by a single gene. In contrast, the similarly expressed rat aldehyde oxidase showed only one immunoreactive protein and monophasic pattern. The biphasic phenomenon could be caused by the existence of two aldehyde oxidase isoforms or two active sites in a single enzyme or some other reasons. Further studies on the problems of the biphasic pattern and species differences in aldehyde oxidase are needed.


Subject(s)
Aldehyde Oxidase/genetics , Liver/enzymology , Aldehyde Oxidase/chemistry , Aldehyde Oxidase/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Macaca fascicularis , Male , Molecular Sequence Data , Rats , Species Specificity
11.
Drug Metab Dispos ; 35(5): 734-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17293383

ABSTRACT

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.


Subject(s)
Aldehyde Oxidase/genetics , Aldehyde Oxidase/metabolism , Polymorphism, Single Nucleotide , Aldehyde Oxidase/antagonists & inhibitors , Amino Acid Substitution , Animals , Blotting, Western , Cytosol/enzymology , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Genotype , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Isoelectric Focusing/methods , Kinetics , Liver/cytology , Liver/enzymology , Luminescent Measurements/methods , Male , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Nitriles/chemistry , Nitriles/metabolism , Nitriles/pharmacology , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
12.
Biol Pharm Bull ; 29(12): 2362-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17142964

ABSTRACT

The substrate selectivity of monoamine oxidase A (MAO-A), monoamine oxidase B (MAO-B), diamine oxidase (DAO), and semicarbazide-sensitive amine oxidase (SSAO) was investigated in the absence of chemical inhibitors using the COS-1 cells expressed with respective amine oxidase. Serotonin (5-hydroxytryptamine), 1-methylhistamine, and histamine were preferentially oxidized by MAO-A, SSAO, and DAO, respectively, at a low substrate concentration. In contrast, benzylamine, tyramine, and beta-phenylethylamine served as substrates for all of MAO-A, MAO-B, and SSAO. Each amine oxidase showed broad substrate selectivity at a high substrate concentration. The cross-inhibition was remarkable in MAO-A and MAO-B, especially in MAO-A, but not in SSAO and DAO. A study of the substrate selectivity of amine oxidases should include consideration of the effects of substrate concentration and specific chemical inhibitors.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Monoamine Oxidase/metabolism , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , DNA Primers , Substrate Specificity
13.
Chirality ; 18(9): 698-706, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16823812

ABSTRACT

RS-8359, (+/-)-4-(4-cyanoanilino)-5,6-dihydro-7-hydroxy-7H-cyclopenta[d]-pyrimidine is a selective and reversible MAO-A inhibitor. The (S)-enantiomer of RS-8359 has been demonstrated to be inverted to the (R)-enantiomer after oral administration to rats. In the current study, we investigated the chiral inversion mechanism and the properties of involved enzymes using rat liver subcellular fractions. The 7-hydroxy function of RS-8359 was oxidized at least by the two different enzymes. The cytosolic enzyme oxidized enantiospecifically the (S)-enantiomer with NADP as a cofactor. On the other hand, the microsomal enzyme catalyzed more preferentially the oxidation of the (S)-enantiomer than the (R)-enantiomer with NAD as a cofactor. With to product enantioselectivity of reduction of the 7-keto derivative, it was found that only the alcohol bearing (R)-configuration was formed by the cytosolic enzyme with NADPH and the microsomal enzyme with NADH at almost equal rate. The reduction rate was much larger than the oxidation rate of 7-hydroxy group. The results suggest that the chiral inversion might occur via an enantioselectivity of consecutive two opposing reactions, oxidation and reduction of keto-alcohol group. In this case, the direction of chiral inversion from the (S)-enantiomer to the (R)-enantiomer is governed by the enantiospecific reduction of intermediate 7-keto group to the alcohol with (R)-configuration. The enzyme responsible for the enantiospecific reduction of the 7-keto group was purified from rat liver cytosolic fractions and identified as 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD) via database search of peptide mass data obtained by nano-LC/MS/MS.


Subject(s)
Alcohols/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Nitriles/pharmacology , Pyrimidines/pharmacology , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/chemistry , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/metabolism , Administration, Oral , Animals , Chromatography, Liquid , Cytosol/enzymology , Electrophoresis, Polyacrylamide Gel , Liver/enzymology , Male , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Molecular Structure , Monoamine Oxidase Inhibitors/administration & dosage , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/metabolism , NAD/metabolism , NADP/metabolism , Nanotechnology , Nitriles/administration & dosage , Nitriles/chemistry , Nitriles/metabolism , Oxidation-Reduction , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Pyrimidines/metabolism , Rats , Rats, Wistar , Stereoisomerism , Subcellular Fractions/enzymology , Substrate Specificity , Tandem Mass Spectrometry
14.
Biopharm Drug Dispos ; 27(5): 247-55, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16586463

ABSTRACT

Aldehyde oxidase catalysed 2-oxidation activity of the (S)-enantiomer of RS-8359, a selective and reversible monoamine oxidase A (MAO-A) inhibitor, was investigated in liver cytosolic fractions from ten rat strains. Remarkably large strain differences were observed with approximately a 230 variation between the highest activity in the Wistar-Imamichi strain and the lowest activity in the Slc:Wistar strain. The activities of Crj:SD and Slc:SD strain rats were considerably low, and that of the F344/DuCrj strain was very low. Among six Wistar strains, Crj:Wistar, Slc:Wistar, WKY/Izm, WKAH/Hkm, Jcl:Wistar and Wistar-Imamichi, the Slc:Wistar strain rats showed exceptionally low 2-oxidation activity that was comparable to that of the F344/DuCrj strain. The rat strain differences in the catalytic activity of aldehyde oxidase could correlate in part with the expressed levels of protein based on the mRNA of aldehyde oxidase. However, no small discrepancy existed in the almost negligible catalytic activity and the fairly high expression levels of protein and mRNA in the F344/DuCrj and Slc:Wistar strain rats. Some genetic factors might possibly be one of reasons for the discrepancy.


Subject(s)
Aldehyde Oxidase/metabolism , Liver/enzymology , Monoamine Oxidase Inhibitors/metabolism , Nitriles/metabolism , Pyrimidines/metabolism , Aldehyde Oxidase/analysis , Aldehyde Oxidase/genetics , Animals , Blotting, Northern , Catalysis , Cytosol/enzymology , Male , Mice , Oxidation-Reduction , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Stereoisomerism
15.
Chirality ; 18(8): 592-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16642492

ABSTRACT

RS-8359, (+/-)-4-(4-cyanoanilino)-5,6-dihydro-7-hydroxy-7H-cyclopenta[d]-pyrimidine is a racemic compound with a selective and reversible monoamine oxidase A (MAO-A) inhibition activity. The substrate and product enantioselectivity with respect to 2-hydroxylation of RS-8359 enantiomers was studied using mouse and rat liver microsomes. In mice, the (S)-enantiomer was transformed to the cis-diol metabolite, whereas the (R)-enantiomer to the trans-diol metabolite. The Vmax/Km value for the formation of the cis-diol metabolite from the (S)-enantiomer was sevenfold greater than that for the formation of the trans-diol metabolite from the (R)-enantiomer. The greater Vmax/Km value for the (S)-enantiomer was due to the tenfold smaller Km value compared to that for the (R)-enantiomer. The results were in fair agreement with the previously reported low plasma concentrations of the (S)-enantiomer and the high recovery of the cis-diol metabolite derived from the (S)-enantiomer in urine after oral administration of RS-8359 to mice. Similarly to mice, in rats the (R)-enantiomer was transformed to the trans-diol metabolite, whereas the (S)-enantiomer yielded the cis-diol and trans-diol metabolites. The Vmax/Km value for the (R)-enantiomer was larger than that for the (S)-enantiomer in rats, indicating that the low plasma concentration of the (S)-enantiomer in rats might be caused by a metabolic reaction other than P450-dependent hydroxylation. CYP3A was shown to be responsible for the trans-diol formation from the (R)-enantiomer.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Monoamine Oxidase Inhibitors/pharmacokinetics , Nitriles/pharmacokinetics , Pyrimidines/pharmacokinetics , Animals , Biotransformation , Hydroxylation , Male , Mice , Mice, Mutant Strains , Molecular Structure , Monoamine Oxidase Inhibitors/blood , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/metabolism , Nitriles/blood , Nitriles/chemistry , Nitriles/metabolism , Pyrimidines/blood , Pyrimidines/chemistry , Pyrimidines/metabolism , Rats , Rats, Wistar , Species Specificity , Stereoisomerism
16.
J Pharm Pharmacol ; 58(3): 403-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16536909

ABSTRACT

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 microM) 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.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Endothelial Cells/metabolism , Lung/blood supply , Nicotine/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Cotinine/metabolism , Cyclic N-Oxides/metabolism , Cytochrome P-450 CYP3A , Cytochrome P450 Family 2 , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Male , Membrane Proteins/metabolism , Microcirculation/metabolism , Nicotine/analogs & derivatives , Oxygenases/metabolism , Rats , Rats, Wistar , Steroid 16-alpha-Hydroxylase/metabolism
17.
Biopharm Drug Dispos ; 27(3): 133-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16400710

ABSTRACT

The 2-oxidation activity on the pyrimidine ring of RS-8359, a MAO-A inhibitor, is the major metabolic pathway catalysed by aldehyde oxidase. This study investigated the species differences in the 2-oxidation activity by using liver cytosolic fractions from rats, mice, guinea-pigs, rabbits, dogs, monkeys and humans. The Vmax/Km value for the (S)-enantiomer of RS-8359 was extremely high in monkeys and humans, moderate in guinea-pigs, and low in rats and mice. Dogs were deficient in 2-oxidation activity. The (R)-enantiomer was only oxidized at a very low rate in guinea-pigs, monkeys and humans, and not oxidized in rats, mice and rabbits. Thus, marked species differences and enantioselectivity were obvious for the 2-oxidation of the (S)-enantiomer of RS-8359. The in vitro results were in good accordance with previously reported in vivo excretion data of the 2-keto metabolite and the non-detectable plasma concentrations of the (S)-enantiomer in monkeys and humans after administration of racemic RS-8359. Enantioselectivity was also observed for the oxidation of cinchona alkaloids catalysed by aldehyde oxidase. Among the four cinchona alkaloids studied, the oxidation activity of cinchonidine, which has no substituents at the 6-hydroxy group but bears (8S,9R)-configurations, was highest. As opposed to the (S)-enantiomer, an extremely high catalytic activity of cinchonidine was confirmed in rabbits, but not in monkeys or humans. Rabbit liver aldehyde oxidase was suggested to have characteristic properties around the active site.


Subject(s)
Aldehyde Oxidase/metabolism , Cinchona Alkaloids/metabolism , Monoamine Oxidase Inhibitors/metabolism , Nitriles/metabolism , Pyrimidines/metabolism , Animals , Cytosol/metabolism , Dogs , Guinea Pigs , Humans , Keto Acids/metabolism , Liver/cytology , Liver/metabolism , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Monoamine Oxidase Inhibitors/chemistry , Nitriles/chemistry , Oxidation-Reduction , Pyrimidines/chemistry , Rabbits , Rats , Rats, Wistar , Species Specificity , Stereoisomerism
18.
Biol Pharm Bull ; 28(5): 882-5, 2005 May.
Article in English | MEDLINE | ID: mdl-15863898

ABSTRACT

To anticipate drug-drug interactions by nicardipine in vivo, cytochrome P450 (CYP) forms responsible for the metabolism of nicardipine and inhibition of CYP-dependent drug metabolism by nicardipine were investigated. Microsomes of human B-lymphoblastoid cells expressing each human CYP form were used for the metabolism of nicardipine. Inhibitory effects of nicardipine on drug metabolism were studied using human liver microsomes. CYP2C8, CYP2D6 and CYP3A4 were identified as major CYP forms for the metabolism of nicardipine in human liver microsomes. Nicardipine strongly inhibited two-pathways of triazolam hydroxylation both catalyzed by CYP3A4. Comparison of three Ca(2+) antagonists, nicardipine, nifedipine, and diltiazem revealed that only nicardipine showed such a strong inhibitory potency on the typical CYP2D6-catalyzed drug metabolism. Furthermore, nicardipine inhibited other reactions catalyzed by CYP1A, CYP2A6, CYP2C8, CYP2C9 and CYP2C19 with K(i) values ranging from 1.1 to 29.4 microM. In conclusion, nicardipine was a relatively potent inhibitor of human CYP2D6, CYP3A4 and CYP2C (especially for CYP2C8 and CYP2C19) in vitro, suggesting that drug-drug interactions between nicardipine and other drugs metabolized mainly by these CYP forms appear to occur in vivo.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Nicardipine/pharmacology , Cell Line , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans
19.
Biol Pharm Bull ; 28(3): 413-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15744061

ABSTRACT

Semicarbazide-sensitive amine oxidase (SSAO) (EC 1.4.3.6) is widely distributed in nature and catalyzes the oxidative deamination of primary amines. Although SSAO full-length cDNA sequences have been reported for some mammalian species, only a partial 5'-terminal sequence has been confirmed in the rat. In this study we isolated full-length SSAO cDNA from rat aorta and examined its mRNA expression in various rat tissues by real-time PCR, as well as the subcellular and tissue distributions of SSAO activity. The deduced amino acid sequence showed 91% and 80% identity with mouse and human SSAO, respectively. The mRNA was expressed in many rat tissues. Those findings were supported by the broad distribution of SSAO in the body. Thus, a high level of SSAO was shown in adipocytes by both mRNA expression and enzyme activity measurement. The results suggest that SSAO may play an important role in the degradation of biologically active amines in adipocytes.


Subject(s)
Amine Oxidase (Copper-Containing)/chemistry , Amine Oxidase (Copper-Containing)/genetics , Cloning, Molecular/methods , Amine Oxidase (Copper-Containing)/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Male , Molecular Sequence Data , Organ Specificity/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods
20.
Chirality ; 17(3): 135-41, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15704197

ABSTRACT

RS-8359, (+/-)-4-(4-cyanoanilino)-5,6-dihydro-7-hydroxy-7H-cyclopenta[d]pyrimidine selectively and reversibly inhibits monoamine oxidase A (MAO-A). After oral administration of rac-RS-8359 to rats, mice, dogs, monkeys, and humans, plasma concentrations of the (R)-enantiomer were greatly higher than were those of the (S)-enantiomer in all species studied. The AUC((R)) to AUC((S)) ratios were 2.6 in rats, 3.8 in mice, 31 in dogs, and 238 in monkeys, and the (S)-enantiomer was almost negligible in human plasma. After intravenous administration of RS-8359 enantiomers to rats, the pharmacokinetic parameters showed that the (S)-enantiomer had a 2.7-fold greater total clearance (CL(t)) and a 70% shorter half-life (t(1/2)) than those for the (R)-enantiomer but had no difference in distribution volume (V(d)). No significant difference in the intestinal absorption rate was observed. The principal metabolites were the 2-keto form, possibly produced by aldehyde oxidase, the cis-diol form, and the 2-keto-cis-diol form produced by cytochrome P450 in rats, the cis-diol form in mice, RS-8359 glucuronide in dogs, and the 2-keto form in monkeys and humans. Thus, the rapid disappearance of the (S)-enantiomer from the plasma was thought to be due to the rapid metabolism of the (S)-enantiomer by different drug-metabolizing enzymes, depending on species.


Subject(s)
Monoamine Oxidase Inhibitors/pharmacokinetics , Nitriles/pharmacokinetics , Pyrimidines/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Dogs , Humans , Injections, Intravenous , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Monoamine Oxidase Inhibitors/blood , Monoamine Oxidase Inhibitors/urine , Nitriles/blood , Nitriles/urine , Pyrimidines/blood , Pyrimidines/urine , Rats , Rats, Wistar , Species Specificity , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...