Network


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

Hotspot


Dive into the research topics where Yuji Mukai is active.

Publication


Featured researches published by Yuji Mukai.


Biological & Pharmaceutical Bulletin | 2015

Effects of Angiotensin II Receptor Blockers on Metabolism of Arachidonic Acid via CYP2C8

Asuna Senda; Yuji Mukai; Takaki Toda; Toru Hayakawa; Miki Yamashita; Erik Eliasson; Anders Rane; Nobuo Inotsume

Arachidonic acid (AA) is metabolized to epoxyeicosatrienoic acids (EETs) via cytochrome enzymes such as CYP 2C9, 2C8 and 2J2. EETs play a role in cardioprotection and regulation of blood pressure. Recently, adverse reactions such as sudden heart attack and fatal myocardial infarction were reported among patients taking angiotensin II receptor blockers (ARBs). As some ARBs have affinity for these CYP enzymes, metabolic inhibition of AA by ARBs is a possible cause for the increase in cardiovascular events. In this study, we quantitatively investigated the inhibitory effects of ARBs on the formation of EETs and further metabolites, dihydroxyeicosatrienoic acids (DHETs), from AA via CYP2C8. In incubations with recombinant CYP2C8 in vitro, the inhibitory effects were compared by measuring EETs and DHETs by HPLC-MS/MS. Inhibition of AA metabolism by ARBs was detected in a concentration-dependent manner with IC50 values of losartan (42.7 µM), telmisartan (49.5 µM), irbesartan (55.6 µM), olmesartan (66.2 µM), candesartan (108 µM), and valsartan (279 µM). Losartan, telmisartan and irbesartan, which reportedly accumulate in the liver and kidneys, have stronger inhibitory effects than other ARBs. The lower concentration of EETs leads to less protective action on the cardiovascular system and a higher incidence of adverse effects such as sudden heart attack and myocardial infarction in patients taking ARBs.


Basic & Clinical Pharmacology & Toxicology | 2017

Angiotensin II Receptor Blockers Inhibit the Generation of Epoxyeicosatrienoic Acid from Arachidonic Acid in Recombinant CYP2C9, CYP2J2 and Human Liver Microsomes

Asuna Senda; Yuji Mukai; Toru Hayakawa; Yuka Kato; Erik Eliasson; Anders Rane; Takaki Toda; Nobuo Inotsume

Cytochrome P450 (CYP) 2C9, CYP2C8 and CYP2J2 enzymes, which metabolize arachidonic acid (AA) to epoxyeicosatrienoic acids, have cardioprotective effects including anti‐inflammation and vasodilation. We have recently shown that some angiotensin II receptor blockers (ARBs) may inhibit AA metabolism via CYP2C8. Using recombinant CYP2C9, CYP2J2 and human liver microsomes (HLMs), the aim was now to compare the ability of six different clinically used ARBs to inhibit AA metabolism in vitro. The rank order of the ARBs for the 50% inhibitory concentration (IC50) of AA metabolism was losartan


Biological & Pharmaceutical Bulletin | 2015

Simultaneous Determination Method of Epoxyeicosatrienoic Acids and Dihydroxyeicosatrienoic Acids by LC-MS/MS System

Yuji Mukai; Takaki Toda; Satoya Takeuchi; Asuna Senda; Miki Yamashita; Erik Eliasson; Anders Rane; Nobuo Inotsume

Epoxyeicosatrienoic acids (EETs) are produced primarily by CYPs from arachidonic acid (AA) and then further metabolized to the corresponding dihydroxyeicosatrienoic acids (DHETs). EETs play important roles in physiological processes such as regulating vasodilation and inflammation. Thus, the drug inhibition of CYP-mediated AA metabolism could reduce production of EETs, potentially resulting in adverse cardiovascular events. The aim of this study was to develop a simple method to simultaneously determine the concentrations of both EETs and DHETs using a conventional LC-MS/MS system to evaluate drug-endogenous substance interactions, including eicosanoids. Eight eicosanoids (5,6-EET, 8,9-EET, 11,12-EET, 14,15-EET, 5,6-DHET, 8,9-DHET, 11,12-DHET, and 14,15-DHET) were detected with their corresponding deuterium-labeled eicosanoids as internal standards. The samples were purified by solid-phase extraction columns. Liquid chromatographic separation was achieved on a C18 column. DHETs and EETs were eluted at 4-7 and 18-26 min, respectively. The weighted (1/y(2)) calibration curves were linear over a range of 5-2000 nmol/L for EETs and 2-2000 nmol/L for DHETs. In quality control (QC) samples, the recoveries of eicosanoids were 95.2-118%. The intra-day precisions were within 6% in all three QC samples, and the inter-day precisions were <16.7% at 50 nmol/L, <8.6% at 200 nmol/L, and <9.8% at 1000 nmol/L. We have applied this method for the determination of the eicosanoid levels in samples from incubation studies of AA by using human recombinant CYP enzyme (rCYP), and confirmed that the method has sensitivity sufficient for assessment of rCYP incubation study.


Biological & Pharmaceutical Bulletin | 2017

Co-administration of Fluvastatin and CYP3A4 and CYP2C8 Inhibitors May Increase the Exposure to Fluvastatin in Carriers of CYP2C9 Genetic Variants

Yuji Mukai; Masayuki Narita; Erika Akiyama; Kanami Ohashi; Yasutaka Horiuchi; Yuka Kato; Takaki Toda; Anders Rane; Nobuo Inotsume

Fluvastatin, which is one of the hydroxymethylglutaryl-CoA (HMG-CoA) reductase inhibitors (statins), is primarily metabolized by CYP2C9 and to a lesser extent by CYP3A4 and CYP2C8. Predictions of drug-drug interactions (DDI) are important for the safety of combination therapies with statins, in particular drugs that are metabolized by CYP3A4. Little information is available regarding drug interactions with fluvastatin. Since CYP2C9 is a polymorphic enzyme, we investigated the effect of DDI via CYP2C9, CYP3A4, and CYP2C8 on fluvastatin pharmacokinetics by using a validated prediction method in relation to CYP2C9 variants. The predicted increases in the area under the concentration-time curve (AUC) ratios of fluvastatin in carriers with CYP2C9*1/*2, CYP2C9*1/*3, CYP2C9*2/*2, CYP2C9*2/*3, and CYP2C9*3/*3 versus that found in carriers with CYP2C9*1/*1 were 1.16, 1.35, 1.37, 1.65, and 2.06, respectively. Our in silico model predicted that administration of fluvastatin in conjunction with the potent inhibitors that completely inhibited CYP3A4 and CYP2C8 in carriers with the CYP2C9*3/*3 variant would cause a 3.23- and 2.60-fold increase in the AUC ratios, respectively, when compared to that for the carriers with the CYP2C9*1/*1 taking fluvastatin alone. We also predicted the effect of telmisartan when coadministered with fluvastatin. Our prediction results showed that the interaction between telmisartan and fluvastatin via CYP enzymes were negligible in clinical situations.


Biological & Pharmaceutical Bulletin | 2017

The Inhibitory Effect of Telmisartan on the Metabolism of Arachidonic Acid by CYP2C9 and CYP2C8: An in Vitro Study

Yuka Kato; Yuji Mukai; Anders Rane; Nobuo Inotsume; Takaki Toda

Epoxyeicosatorienoic acids (EETs) are generated from arachidonic acid (AA) by CYPs. EETs comprise four regioisomers (14,15-, 11,12-, 8,9-, and 5,6-EET). EETs show potent physiological effects, including vasodilation, anti-inflammation, myocardial preconditioning, and anti-platelet aggregation effects. We recently demonstrated that telmisartan, one of angiotensin II receptor blockers, inhibits AA metabolism by CYP enzymes, including CYP2C8, CYP2C9, and CYP2J2. We conducted studies of AA metabolism using recombinant CYP enzymes to estimate the inhibition constant and the type of inhibition by telmisartan of CYP2C9 and CYP2C8. The contribution ratio (CR) of each CYP enzyme was investigated using human liver microsomes. Dixon and Lineweaver-Burk plots indicated that telmisartan is a mixed inhibitor of both CYP2C9 and CYP2C8; telmisartan did not show a time-dependent inhibition toward these CYP enzymes. Based on the CRs, both CYP2C9 and CYP2C8 are the key enzymes in the metabolism of AA in the human liver. Uptake of telmisartan in the liver by organic anion transporting polypeptide (OATP) 1B3 and the non-linear metabolism in gastrointestinal tract augment the potential of the drug to inhibit the CYP enzymes in the liver.


Xenobiotica | 2018

Combined effect of telmisartan and fluvastatin on arachidonic acid metabolism in human liver microsomes

Yuka Kato; Yuji Mukai; Anders Rane; Nobuo Inotsume; Takaki Toda

Abstract 1. Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid (AA) via cytochrome P450s, have a protective effect on the cardiovascular system involving vasodilation. We have previously demonstrated that telmisartan (TEL) inhibits EETs production from AA in vitro. 2. The objectives of the study were to examine the inhibitory effect of fluvastatin (FLU), an inhibitor of CYP2C9, and the combined effect of TEL and FLU on the production of EETs using human liver microsomes. The combined effect of TEL and FLU was evaluated using two methods, the fixed concentration method and the fixed ratio method. 3. FLU significantly reduced total eicosanoids (sum of EETs and their subsequent metabolites dihydroxyeicosatrienoic acids) production at > 0.25 µM. The results of the fixed concentration method indicated that the addition of the other inhibitor resulted in significant reduction of the production of total eicosanoids in a concentration-dependent manner. In the fixed ratio method, the combination of TEL and FLU over all concentration ratios tested did not produce a horizontal shift in the dose response curves. 4. Our results showing an additive combined effect of TEL and FLU on AA metabolism, suggest that concomitant treatment with TEL and FLU would theoretically affect the vascular tone mediated by EETs from AA.


Biological & Pharmaceutical Bulletin | 2017

Concentration of Sulfate and Glucuronide Conjugates of Ritodrine in Twin Pregnancy

Mayuko Soma; Ainari Konda; Satoko Fujieda; Yoichi Sasaki; Mitsuaki Keira; Hiroshi Yoshida; Yuji Mukai; Takaki Toda; Nobuo Inotsume

Ritodrine, a drug for the treatment of threatened premature labor, is a highly selective beta-2 agonist with the major metabolites of sulfate and glucuronide conjugates. This study investigated the continuous evaluation of the concentration of ritodrine conjugates in relation to the clinical course in twin pregnancy. The subjects were 9 twin-pregnancy mothers who delivered after receiving ritodrine treatment between April 2012 and December 2013. Serum ritodrine sulfate and glucuronide conjugates were deconjugated using their specific enzymes. Ritodrine concentration was measured by liquid chromatography-tandem mass spectrometry. The continuous infusion rate of ritodrine was 2.66±0.67 (0.8-3.54) µg/min/kg, and the average concentration of unchanged ritodrine was 118.8±33.2 (63.8-194.0) ng/mL. During the study period between week 32 and week 36 of gestation, the average ratio of unchanged ritodrine concentration and sulfate ritodrine conjugate concentration for weeks 32, 33, 34, 35, and 36 were 1.7, 1.9, 1.5, 1.7, and 1.7 not significant (N.S.), respectively. The average ratio of unchanged ritodrine concentration and glucuronide ritodrine conjugate concentration were 1.8, 2.2, 1.9, 1.8, and 2.1 (N.S.), respectively. No statistical difference was identified in the ratios of unchanged ritodrine concentration and sulfate or glucuronide ritodrine conjugate concentrations. Large individual differences were shown in the concentration of sulfate and glucuronide during the gestational period. No change in the ratio of the formation of ritodrine metabolites was identified as the gestational age progressed.


Basic & Clinical Pharmacology & Toxicology | 2017

Reply to ‘Multiple and Opposite Effects of Angiotensin II Receptor Blockers on the Bioavailability of Epoxyeicosatrienoic Acids’

Asuna Senda; Yuji Mukai; Toru Hayakawa; Yuka Kato; Erik Eliasson; Anders Rane; Takaki Toda; Nobuo Inotsume

Dear Editor, We have read the letter against our recent research of the effects of angiotensin II receptor blockers (ARBs) on the production of arachidonic acid metabolites published in BCPT, from Bellien et al. with great interest, although some concerns were raised about their interpretations. They expanded results of some studies using only one agent of ARBs to the effect of all ARBs. The inhibitory effects of ARBs on the production of epoxyeicosatrienoic acids (EETs) were different among ARBs as shown in our reports [1,2]. Koh et al. [3] have reported that losartan exerted the different effect on plasma level of plasminogen activator I antigen, a thrombolytic marker, from irbesartan and candesartan in patients with hypertension. Thus, the effect of each ARB on the cardiovascular system might be inherent to the drugs. They stated that blockage of angiotensin II type I (AT1) receptor by ARB resulted in down-regulation of soluble epoxide hydrolase (sEH) and is expected to elevate EET levels. However, only high dosage of losartan (25 mg/kg/day) was used in the original report [4]. It is uncertain whether the same phenomenon will be observed by the administration of losartan in clinical settings. They described as ‘it was demonstrated that angiotensin II stimulates the CYP450-dependent production of arachidonic acid metabolites in cultured endothelial cells [5], and further ex vivo experiments confirmed that EETs are in fact produced in response to angiotensin II type II (AT2) receptors stimulation and this effect is potentiated by AT1 receptors blockage [6,7]’. However, none of concentrations of EETs and dihydroxyeicosatrienoic acids (DHETs) were included in these articles [5–7]. The in vitro study [5] was designed to examine the response to the stimulation induced by angiotensin II in the absence or presence of each EET regioisomer without monitoring any production of arachidonic acid metabolites. The results of the report [6] showed the interesting results using CV11974 (candesartan). Concomitant treatment with candesartan (10 nM) and miconazole, one of the typical cytochrome P450 (CYP) inhibitors, suppressed the vasodilation effect that was observed in the presence of candesartan, suggesting that EETs are associated with vasodilation mediated by AT2 receptor. As shown in our studies [1,2], candesartan was a weak inhibitor of the production of EETs among ARBs. Thus, we should expand our study to examine the effect of telmisartan, which showed the most potent inhibitory effects on the production of EETs. AT2 receptor was not easy to find in healthy organs in the adults, and its expression is induced under pathophysiological conditions such as mechanical injury or ischaemia [8,9]. Recently, Romero-Nava et al. [10] reported that the hypertensive rats had a higher mRNA and protein expression of AT1 receptor than normotensive rats, while the AT2 expression in aorta remained unchanged. Thus, the effect through the activation of AT2 receptor of each ARB in patients with hypertension should be further evaluated. They stated that the activation of Mas receptors associated with ARB administration contributed to an increase in EET production. However, this evidence was limited to azilsartan (3 mg/kg/day), which showed a weak inhibition of the production of EETs (unpublished data). It should be noted that levels of EETs/DHETs did not alter before and after treatment with azilsartan in the original article [11]. We are now conducting a clinical trial to evaluate the impact of ARBs on the circulating EET/DHET concentrations in patients with hypertension. We would like to report the results in near future.


Basic & Clinical Pharmacology & Toxicology | 2016

The role of CYP2C8 and CYP2C9 Genotypes in Losartan‐Dependent Inhibition of Paclitaxel Metabolism in Human Liver Microsomes

Yuji Mukai; Asuna Senda; Takaki Toda; Erik Eliasson; Anders Rane; Nobuo Inotsume

The aim of the present study was to further investigate a previously identified metabolic interaction between losartan and paclitaxel, which is one of the marker substrates of CYP2C8, by using human liver microsomes (HLMs) from donors with different CYP2C8 and CYP2C9 genotypes. Although CYP2C8 and CYP2C9 exhibit genetic linkage, previous studies have yet to determine whether losartan or its active metabolite, EXP‐3174 which is specifically generated by CYP2C9, is responsible for CYP2C8 inhibition. Concentrations of 6α‐hydroxypaclitaxel and EXP‐3174 were measured by high‐performance liquid chromatography after incubations with paclitaxel, losartan or EXP‐3174 in HLMs from seven donors with different CYP2C8 and CYP2C9 genotypes. The half maximal inhibitory concentration (IC50) values were not fully dependent on CYP2C8 genotypes. Although the degree of inhibition was small, losartan significantly inhibited the production of 6α‐hydroxypaclitaxel at a concentration of 1 μmol/L in only HL20 with the CYP2C8*3/*3 genotype. HLMs with either CYP2C9*2/*2 or CYP2C9*1/*3 exhibited a lower losartan intrinsic clearance (Vmax/Km) than other HLMs including those with CYP2C9*1/*1 and CYP2C9*1/*2. Significant inhibition of 6α‐hydroxypaclitaxel formation by EXP‐3174 could only be found at levels that were 50 times higher (100 μmol/L) than the maximum concentration generated in the inhibition study using losartan. These results suggest that the metabolic interaction between losartan and paclitaxel is dependent on losartan itself rather than its metabolite and that the CYP2C8 inhibition by losartan is not affected by the CYP2C9 genotype. Further study is needed to define the effect of CYP2C8 genotypes on losartan–paclitaxel interaction.


Rinsho Yakuri\/japanese Journal of Clinical Pharmacology and Therapeutics | 2017

Elimination of Ritodrine in Mothers Pregnant with Twins and Their Neonatal Twins

Satoko Fujieda; Mitsuaki Keira; Mayuko Soma; Yoichi Sasaki; Ainari Konda; Yuji Mukai; Takaki Toda; Nobuo Inotsume; Hiroshi Yoshida

Collaboration


Dive into the Yuji Mukai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anders Rane

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Erik Eliasson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Toru Hayakawa

Hokkaido College of Pharmacy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge