Network


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

Hotspot


Dive into the research topics where Masamichi Hirayama is active.

Publication


Featured researches published by Masamichi Hirayama.


Drug Metabolism and Disposition | 2005

INVOLVEMENT OF URIC ACID TRANSPORTER IN INCREASED RENAL CLEARANCE OF THE XANTHINE OXIDASE INHIBITOR OXYPURINOL INDUCED BY A URICOSURIC AGENT, BENZBROMARONE

Takashi Iwanaga; Daisuke Kobayashi; Masamichi Hirayama; Tomoji Maeda; Ikumi Tamai

Benzbromarone has been reported to increase the renal clearance of oxypurinol, an active metabolite of allopurinol. We examined the renal transport of oxypurinol to determine whether such a change in renal clearance could be explained by altered transporter-mediated reabsorption. Since the first step of reabsorption takes place at the renal epithelial apical membrane, we focused on membrane transporters. Benzbromarone is an inhibitor of reabsorption of uric acid mediated by the uric acid transporter (URAT) URAT1 (SLC22A12), which is expressed at the apical membrane of proximal tubular cells in humans. Uptake of oxypurinol by Xenopus oocytes injected with complementary RNA of URAT1 was significantly higher than that by water-injected oocytes, and the uptake was saturable, with a Km of about 800 μM. Moreover, benzbromarone inhibited the oxypurinol uptake by URAT1 at concentrations as low as 0.01 μM. The uptake of oxypurinol by another organic anion transporter (OAT), OAT4 (SLC22A11), which is also expressed at the apical membrane of proximal tubular epithelial cells, was negligible, whereas the uptake of [3H]estrone-3-sulfate by OAT4 was significantly inhibited by oxypurinol. Furthermore, neither the transport activity of organic cation/carnitine transporter (OCTN) 1 nor OCTN2 was affected by oxypurinol or benzbromarone. These results indicate that URAT1 is involved in renal reabsorption of oxypurinol, and the increment of renal clearance of oxypurinol upon concomitant administration of benzbromarone could be due to drug interaction at URAT1.


Drug Metabolism and Disposition | 2006

Mechanism of the Regulation of Organic Cation/Carnitine Transporter 1 (SLC22A4) by Rheumatoid Arthritis-Associated Transcriptional Factor RUNX1 and Inflammatory Cytokines

Tomoji Maeda; Masamichi Hirayama; Daisuke Kobayashi; Keiji Miyazawa; Ikumi Tamai

Recently, it was reported that the organic cation/carnitine transporter 1 (OCTN1, SLC22A4) is associated with chronic inflammatory diseases, such as rheumatoid arthritis (RA) and Crohns disease. OCTN1 in humans is expressed in synovial tissues of individuals with rheumatoid arthritis. Furthermore octn1 in mice is expressed in inflamed joints with collagen-induced arthritis, a model of human arthritis, but not in the joints of normal mice. OCTN1 should be involved in the inflammatory disease and in the present study, the regulatory mechanism of OCTN1 expression was characterized using the human fibroblast-like synoviocyte cell line MH7A, derived from RA patients. A luciferase-reporter gene assay and gel shift assay demonstrated that RUNX1, which is an essential hematopoietic transcription factor associated with acute myeloid leukemia and is related to RA and Sp1, is involved in the regulation of OCTN1 promoter activity. Inflammatory cytokines such as interleukin-1β and tumor necrosis factor-α increased the expression of OCTN1 mRNA. Furthermore, overexpression of nuclear factor-κB (NF-κB) activated promoter activity of OCTN1. These results clearly demonstrate that expression of OCTN1 is regulated by various factors, including RUNX1, inflammatory cytokines, and NF-κB, all of which are also related to the pathogenesis of RA. Further studies on the physiological substrate(s) of OCTN1 should be done to clarify the roles of OCTN1 in these diseases.


Pharmaceutical Research | 2006

Characterization of Human OATP2B1 (SLCO2B1) Gene Promoter Regulation

Tomoji Maeda; Masamichi Hirayama; Ryunosuke Higashi; Masanobu Sato; Ikumi Tamai

PurposeWe investigated transcriptional regulation of organic anion transporter OATP2B1 (SLCO2B1) that is expressed in multiple tissues such as liver, small intestine, and others and compared it with that of liver-specific OATPs.MethodsThe promoter activity was examined by luciferase assay. Specific bindings of transcription factors to the promoter region were examined by gel mobility shift assay using native and mutated nucleotides of the promoter region of OATP2B1.ResultsDeletion–mutation study of the promoter region of OATP2B1 showed that the −59 region that included the Sp1 binding site had basal promoter activity, whereas promoter activities of the further upper region were different between intestine-derived Caco-2 cells and liver-derived HepG2 cells. The association of Sp1 to the promoter region was confirmed by gel shift assay and overexpression of Sp1 in cultured cells. Although the promoter of OATP2B1 has a putative HNF1α binding site, overexpression of HNF1α did not induce the expression of OATP2B1.ConclusionSp1, a transcription factor, was required for constitutive expression of OATP2B1 in liver and small intestine, whereas HNF1α, which is involved in the expression of liver-specific OATPs, did not seem to play a role in OATP2B1 expression. Accordingly, it was suggested that the tissue expression profile of OATP2B1 was different from that of other liver-specific OATPs.


The Journal of Clinical Pharmacology | 2018

Pharmacokinetics and Pharmacodynamics of Azeloprazole Sodium, a Novel Proton Pump Inhibitor, in Healthy Japanese Volunteers

Ryoko Toda; Masanari Shiramoto; Emi Komai; Kazuyoshi Yoshii; Masamichi Hirayama; Yoshihiro Kawabata

The pharmacokinetics (PK) and pharmacodynamics (PD) of proton pump inhibitors differ among cytochrome P450 (CYP) 2C19 genotypes. Therefore, we developed azeloprazole sodium (Z‐215), a novel proton pump inhibitor, whose metabolism is not affected by CYP2C19 activity in vitro. However, the PK and PD of azeloprazole sodium have not been evaluated in Japanese subjects. We conducted an open‐label, crossover study in healthy Japanese male volunteers to evaluate the plasma concentration and intragastric pH with respect to CYP2C19 genotype after repeated administration of 10, 20, and 40 mg azeloprazole sodium and 10 and 20 mg rabeprazole sodium (rabeprazole). The plasma concentration profile of azeloprazole sodium was similar among genotypes, whereas that of rabeprazole differed. The 24‐hour intragastric pH ≥ 4 holding time ratio (pH ≥ 4 HTR) of azeloprazole sodium was similar among genotypes. The pH ≥ 4 HTR was 52.5%–60.3%, 55.1%–65.8%, and 69.4%–77.1% after administration of 10, 20, and 40 mg azeloprazole sodium, respectively, and 59.2%–72.3% and 64.4%–91.2% after administration of 10 and 20 mg rabeprazole, respectively, on the fifth day of dosing. The maximum plasma concentration (Cmax), area under the plasma concentration‐time curve (AUC), and pH ≥ 4 HTR of azeloprazole sodium were proportional to dose. The Cmax, AUC, and pH ≥ 4 HTR on day 5 were slightly higher following administration of 20 mg azeloprazole sodium before comparison with after a meal. No serious adverse events were observed. These results suggest that azeloprazole sodium is useful for treating gastroesophageal reflux disease in all CYP2C19 genotypes.


Xenobiotica | 2018

Mass balance and metabolism of Z-215, a novel proton pump inhibitor, in healthy volunteers

Ryoko Toda; Tomoharu Miyagawa; Yuka Masuda; Yusuke Hoshino; Kazuyoshi Yoshii; Masamichi Hirayama; Minaka Shibuya; Yoshihiro Kawabata

Abstract The human mass balance of [14 C]Z-215, a novel proton pump inhibitor, was characterised in six healthy male volunteers following single oral administration of [14 C]Z-215 (20 mg, 3.7 MBq) to determine the elimination pathway of Z-215 and the distribution of its metabolites in plasma, urine, and faeces (NCT02618629). [14 C]Z-215 was rapidly absorbed, with a Cmax of 434 ng/mL at 0.38 h for Z-215 and 732 ng eq./mL at 0.5 h for total radioactivity. Means of 59.61% and 31.36% of the administered radioactive dose were excreted in urine and faeces, respectively, within 168 h post-dose. The majority of the dose was recovered within 24 h in urine and 96 h in faeces. Unchanged Z-215 was excreted in urine at trace levels but was not detected in faeces. The main components in plasma were Z-215 and Z-215 sulphone, accounting for 29.8% and 13.3% of the total circulating radioactivity, respectively. Additionally, Z-215 was metabolised through oxidation, reduction and conjugation. Our in vitro Z-215 metabolism study showed that the major isozyme contributing to the oxidation of Z-215, including the formation of Z-215 sulphone, was CYP3A4. In conclusion, Z-215 is well absorbed in humans and primarily eliminated via metabolism, where CYP3A4 plays an important role.


Life Sciences | 2016

Distribution of acotiamide, an orally active acetylcholinesterase inhibitor, into the myenteric plexus of rat and dog stomachs

Kazuyoshi Yoshii; Takashi Yamaguchi; Masamichi Hirayama; Ryoko Toda; Toshiko Kinomoto; Yoshihiro Kawabata; Kan Chiba

AIMS Acotiamide is the first-in-class drug for the treatment of functional dyspepsia. Although pharmacological and therapeutic actions of acotiamide are thought to be derived from its inhibitory effects on acetylcholinesterase (AChE), whether the concentration of acotiamide at the site of action is sufficient to inhibit AChE remains unclear. Since major site of acotiamide action is thought to be the cholinergic nerve terminals in gastric myenteric plexus, we studied the distribution of [(14)C]acotiamide into gastric myenteric plexus. MAIN METHODS Distribution of [(14)C]acotiamide was evaluated using macro- and micro-autoradiography in rats and dogs. KEY FINDINGS The results of macro-autoradiography showed the concentration of radioactivity was 27.9μM in rat stomach, which was 12 times higher than IC50 of acotiamide for rat AChE. Being different from rats, the distribution of radioactivity in the muscular layer was distinguishable from that in the mucosal layer in dog stomach. The concentration of radioactivity in the muscular layer of dog stomach (1.41μM) was approximately two-times lower than those in the mucosal layer, however, it was approximately 1.2 times higher than IC50 of acotiamide for dog AChE. The results of micro-autoradiography also showed the radioactivity distributed homogenously in the muscular layer of rat stomach, suggesting the concentration of radioactivity around the ganglion of myenteric plexus is similar to that in the muscular layer of stomach. SIGNIFICANCE These findings suggest acotiamide distributes to the myenteric plexus of stomach, a putative site of acotiamide action, with adequate concentrations to inhibit AChE, in both of rat and dog stomachs.


Biochemical Pharmacology | 2005

Regulation of testis-specific carnitine transporter (octn3) gene by proximal cis-acting elements Sp1 in mice

Tomoji Maeda; Masamichi Hirayama; Daisuke Kobayashi; Ikumi Tamai


Pharmaceutical Research | 2016

Physiologically-Based Pharmacokinetic and Pharmacodynamic Modeling for the Inhibition of Acetylcholinesterase by Acotiamide, A Novel Gastroprokinetic Agent for the Treatment of Functional Dyspepsia, in Rat Stomach

Kazuyoshi Yoshii; Minami Iikura; Masamichi Hirayama; Ryoko Toda; Yoshihiro Kawabata


Abstracts of JSSX meeting | 2008

MECHANISM OF STOMACH DISTRIBUTION OF A NOVEL GASTROPROKINETIC AGENT, ACOTIAMIDE HYDROCHLORIDE (Z-338/YM443) IN RATS

Kazuyoshi Yoshii; Yoshihiro Kawabata; Masamichi Hirayama; Toshifumi Nakamura; Ryoko Toda; Junko Hasegawa; Mineo Takei; Yukinori Mera


Yakugaku Zasshi-journal of The Pharmaceutical Society of Japan | 2005

Involvement of uric acid transporter URAT1 in increased renal clearance of xanthine oxidase inhibitor oxypurinol by uricosuric agents

Takashi Iwanaga; Daisuke Kobayashi; Masamichi Hirayama; Tomoji Maeda; Ikumi Tamai

Collaboration


Dive into the Masamichi Hirayama's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomoji Maeda

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ryunosuke Higashi

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar

Takashi Iwanaga

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge