Network


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

Hotspot


Dive into the research topics where Hisashi Fujita is active.

Publication


Featured researches published by Hisashi Fujita.


Journal of Neurochemistry | 2011

A novel glycogen synthase kinase-3 inhibitor 2-methyl-5-(3-{4-[(S )-methylsulfinyl]phenyl}-1-benzofuran-5-yl)-1,3,4-oxadiazole decreases tau phosphorylation and ameliorates cognitive deficits in a transgenic model of Alzheimer’s disease

Tomohiro Onishi; Hiroki Iwashita; Yumiko Uno; Jun Kunitomo; Morihisa Saitoh; Eiji Kimura; Hisashi Fujita; Noriko Uchiyama; Masakuni Kori; Masayuki Takizawa

J. Neurochem. (2011) 10.1111/j.1471‐4159.2011.07532.x


Drug Metabolism and Disposition | 2007

Asymmetric Intestinal First-Pass Metabolism Causes Minimal Oral Bioavailability of Midazolam in Cynomolgus Monkey

Tomohiro Nishimura; Nobuyuki Amano; Yoshiyuki Kubo; Midori Ono; Yukio Kato; Hisashi Fujita; Yoshiaki Kimura; Akira Tsuji

Oral bioavailability of some drugs is substantially lower in cynomolgus monkeys than in various other species, including humans. In the present study, midazolam was used as a model drug to investigate the reason for the lower bioavailability in these monkeys. The bioavailability of midazolam after oral administration was minimal in monkeys and rats, being only 2.1 and 1.1%, respectively. In monkeys, this low bioavailability could not be explained simply in terms of a hepatic first-pass effect. To examine the roles of intestinal metabolism and transport, we evaluated apical-to-basal and basal-to-apical transport of midazolam, and the formation of metabolites in small intestinal tissues using an Ussing-type chamber. The values of mucosal extraction ratio were estimated to be 0.97, 0.93, and 0.89 during apical-to-basal transport in the upper, middle, and lower small intestine of monkeys, respectively, whereas the corresponding values for rats were close to zero, indicating that extensive metabolism of midazolam occurs, particularly in the upper region of the small intestine in monkeys, but not rats. Interestingly, formation of the metabolites was much greater during transport in the apical-to-basal direction than in the basal-to-apical direction, and this could be well explained by a mathematical model based on the assumption that extensive metabolism is associated with the uptake process of midazolam from the apical cell surface. Thus, we conclude that an asymmetric distribution of metabolic activity in the small intestine, leading to extensive metabolism during uptake from the apical cell surface, accounts for the minimal oral bioavailability of midazolam in cynomolgus monkeys.


Journal of Biological Chemistry | 2017

In Vivo Knockdown of Pathogenic Proteins via Specific and Nongenetic IAP-dependent Protein Erasers (SNIPERs)

Nobumichi Ohoka; Keiichiro Okuhira; Masahiro Ito; Katsunori Nagai; Norihito Shibata; Takayuki Hattori; Osamu Ujikawa; Kenichiro Shimokawa; Osamu Sano; Ryokichi Koyama; Hisashi Fujita; Mika Teratani; Hirokazu Matsumoto; Yasuhiro Imaeda; Hiroshi Nara; Nobuo Cho; Mikihiko Naito

Many diseases, especially cancers, result from aberrant or overexpression of pathogenic proteins. Specific inhibitors against these proteins have shown remarkable therapeutic effects, but these are limited mainly to enzymes. An alternative approach that may have utility in drug development relies on selective degradation of pathogenic proteins via small chimeric molecules linking an E3 ubiquitin ligase to the targeted protein for proteasomal degradation. To this end, we recently developed a protein knockdown system based on hybrid small molecule SNIPERs (Specific and Nongenetic IAP-dependent Protein Erasers) that recruit inhibitor of the apoptosis protein (IAP) ubiquitin ligases to specifically degrade targeted proteins. Here, we extend our previous study to show a proof of concept of the SNIPER technology in vivo. By incorporating a high affinity IAP ligand, we developed a novel SNIPER against estrogen receptor α (ERα), SNIPER(ER)-87, that has a potent protein knockdown activity. The SNIPER(ER) reduced ERα levels in tumor xenografts and suppressed the growth of ERα-positive breast tumors in mice. Mechanistically, it preferentially recruits X-linked IAP (XIAP) rather than cellular IAP1, to degrade ERα via the ubiquitin-proteasome pathway. With this IAP ligand, potent SNIPERs against other pathogenic proteins, BCR-ABL, bromodomain-containing protein 4 (BRD4), and phosphodiesterase-4 (PDE4) could also be developed. These results indicate that forced ubiquitylation by SNIPERs is a useful method to achieve efficient protein knockdown with potential therapeutic activities and could also be applied to study the role of ubiquitylation in many cellular processes.


Pharmaceutical Research | 2008

Species difference in intestinal absorption mechanism of etoposide and digoxin between cynomolgus monkey and rat

Tomohiro Nishimura; Yukio Kato; Nobuyuki Amano; Midori Ono; Yoshiyuki Kubo; Yoshiaki Kimura; Hisashi Fujita; Akira Tsuji

PurposeThe oral bioavailability of some therapeutic agents is markedly lower in cynomolgus monkeys than in humans. We investigated small-intestinal absorption of the P-glycoprotein (P-gp) substrates etoposide and digoxin in monkeys to clarify the influence of efflux transport on their intestinal permeability.MethodsThe pharmacokinetics of etoposide and digoxin was examined in monkeys and rats after oral and intravenous administration. Intestinal permeability and segmental differences in permeability were investigated with an Ussing-type chamber.ResultsThe bioavailability of etoposide was 12.9 and 13.9% in monkeys and rats, respectively. Total body clearance of etoposide in monkeys was much less than hepatic blood flow, suggesting that the bioavailability would be limited at intestinal absorption. Marked vectorial transport of etoposide in the secretory direction was observed in rats, especially in the lower small intestine, and segmental differences were consistent with the distribution of P-gp expression. Vectorial transport was minimal in monkey small intestine. Our kinetic analysis indicated that P-gp contributes little to the intestinal permeability of etoposide and digoxin in monkeys, and apical uptake is rate-limiting.ConclusionLow bioavailability of etoposide in monkeys is due to poor intestinal uptake resulting from low influx from the apical side, rather than secretion via P-gp.


Bioorganic & Medicinal Chemistry | 2010

Synthesis and structure-activity relationship of tetrahydropyrazolopyrimidine derivatives--a novel structural class of potent calcium-sensing receptor antagonists.

Masato Yoshida; Akira Mori; Atsuhiro Inaba; Masahiro Oka; Haruhiko Makino; Masashi Yamaguchi; Hisashi Fujita; Tomohiro Kawamoto; Mika Goto; Hiroyuki Kimura; Atsuo Baba; Tsuneo Yasuma

A series of novel tetrahydropyrazolopyrimidine derivatives containing an adamantyl group were synthesized and evaluated as potential calcium-sensing receptor (CaSR) antagonists. After chemical modification of 9a, which was identified as a hit compound in a random screening of CaSR antagonist assay, 7,7-dimethyl derivative 16c was found to be the most active compound of this new series (IC(50)=10nM). We report the synthesis of this series and their biological activities and structure-activity relationship.


Journal of Medicinal Chemistry | 2011

Discovery of novel and potent orally active calcium-sensing receptor antagonists that stimulate pulselike parathyroid hormone secretion: synthesis and structure-activity relationships of tetrahydropyrazolopyrimidine derivatives.

Masato Yoshida; Akira Mori; Etsuo Kotani; Masahiro Oka; Haruhiko Makino; Hisashi Fujita; Junko Ban; Yukihiro Ikeda; Tomohiro Kawamoto; Mika Goto; Hiroyuki Kimura; Atsuo Baba; Tsuneo Yasuma

As part of our research for novel calcium-sensing receptor (CaSR) antagonists that can function as oral bone anabolic agents, we recently reported the discovery of a tetrahydropyrazolopyrimidine derivative featuring adamantyl group 1b with potent CaSR antagonistic activity. To explore the potential of this calcilytic congener, we introduced the gem-dialkyl benzyl group at the 3-position of the tetrahydropyrazolopyrimidine ring, forming a bioisostere of the adamantyl group by mimicking the adamantyl groups lipophilicity and bulkiness. Optimization directed toward the improvement of solubility and metabolic stability led to the discovery of compound 9e, which stimulated transient PTH secretion when orally administered to normal rats. Further, compound 9e proved to be fully effective in an osteopenic ovariectomized rat model.


Bioorganic & Medicinal Chemistry | 2017

A PEGylated analog of short-length Neuromedin U with potent anorectic and anti-obesity effects

Hiroshi Inooka; Kotaro Sakamoto; Tokuyuki Shinohara; Yasushi Masuda; Michiko Terada; Satoshi Kumano; Kotaro Yokoyama; Jiro Noguchi; Naoki Nishizawa; Hidenori Kamiguchi; Hisashi Fujita; Taiji Asami; Shiro Takekawa; Tetsuya Ohtaki

Neuromedin U (NMU) is a neuropeptide known to regulate food intake and energy homeostasis that is widely distributed in the gastrointestinal tract, hypothalamus, and pituitary. A short form of NMU, porcine NMU-8 has potent agonist activity for the receptors NMUR1 and NMUR2; however, its short half-life precludes its effective use in vivo. To address this limitation, we designed and synthesized NMU-8 analogs modified by polyethylene glycol (PEG) with a molecular weight of 30kDa (PEG30k) via a variety of linkers (i.e., ω-amino- and ω-imino-carboxylic acid linker). Integrated evaluation of NMUR1 and NMUR2 binding affinities in vitro and anorectic activity in mice revealed that the introduction of a linker with a rigid ring group, e.g., 2-(piperazin-1-yl)acetic acid (PipAc), yielded a highly potent anorectic peptide, PEG30k-PipAc-NMU-8 (14), possessing improved receptor binding affinity. Subsequent optimization of the molecular weight of the PEG moiety led to the discovery of a PEG20k conjugate (15), which exhibited significant anti-obesity effect upon once-daily subcutaneous administration in diet-induced obese mice with 10% and 22% body weight loss at doses of 10 and 30nmol/kg, respectively. In addition, 15 reduced the weights of the liver and adipose tissue in a dose-dependent manner and improved the plasma biochemical parameters, e.g., insulin, glutamic pyruvic transaminase, glutamic oxaloacetic transaminase, and total cholesterol. Thus, our results suggest that 15 (NMU-0002), which showed potent and long-lasting biological profiles in vivo, represents a candidate peptide for investigating the central and peripheral actions of NMU and its potential for clinical use.


Bioorganic & Medicinal Chemistry | 2011

Novel and potent calcium-sensing receptor antagonists: Discovery of (5R)-N-[1-ethyl-1-(4-ethylphenyl)propyl]-2,7,7-trimethyl-5-phenyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide monotosylate (TAK-075) as an orally active bone anabolic agent

Masato Yoshida; Akira Mori; Shinji Morimoto; Etsuo Kotani; Masahiro Oka; Kohei Notoya; Haruhiko Makino; Midori Ono; Mikio Shirasaki; Norio Tada; Hisashi Fujita; Junko Ban; Yukihiro Ikeda; Tomohiro Kawamoto; Mika Goto; Hiroyuki Kimura; Atsuo Baba; Tsuneo Yasuma

The calcium-sensing receptor antagonist (CaSR) has been recognized as a promising target of anabolic agents for treating osteoporosis. In the course of developing a new drug candidate for osteoporosis, we found tetrahydropyrazolopyrimidine derivative 1 to be an orally active CaSR antagonist that stimulated transient PTH secretion in rats. However, compound 1 showed poor physical and chemical stability. In order to work out this compounds chemical stability and further understand its in vivo efficacy, we focused on modifying the 2-position of the tetrahydropyrazolopyrimidine. As a result of chemical modification, we discovered (5R)-N-[1-ethyl-1-(4-ethylphenyl)propyl]-2,7,7-trimethyl-5-phenyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide monotosylate 10m (TAK-075), which showed improved solubility, chemical stability, and in vivo efficacy. Furthermore, we describe that evaluating the active metabolite is important during repeated treatment with short-acting CaSR antagonists.


Journal of Medicinal Chemistry | 2017

Potent Body Weight-Lowering Effect of a Neuromedin U Receptor 2-selective PEGylated Peptide

Yoko Kanematsu-Yamaki; Naoki Nishizawa; Tomoko Kaisho; Hiroaki Nagai; Taisuke Mochida; Tomoko Asakawa; Hiroshi Inooka; Katsuko Dote; Hisashi Fujita; Kouta Matsumiya; Hideki Hirabayashi; Junichi Sakamoto; Tetsuya Ohtaki; Shiro Takekawa; Taiji Asami

Neuromedin U (NMU) is a neuropeptide that mediates a variety of physiological functions via its receptors, NMUR1 and NMUR2. Recently, there has been an increased focus on NMU as a promising treatment option for diabetes and obesity. A short form of NMU (NMU-8) has potent agonist activity for both receptors but is metabolically unstable. Therefore, we designed and synthesized NMU-8 analogues modified by polyethylene glycol (PEG; molecular weight, 20 kDa; PEG20k) via a linker. 3-(2-Naphthyl)alanine substitution at position 19 increased NMUR2 selectivity of NMU-8 analogues with retention of high agonist activity. Compound 37, an NMUR2-selective PEG20k analogue containing piperazin-1-ylacetyl linker, exhibited a potent body weight-lowering effect with concomitant inhibition of food intake in a dose-dependent manner (body weight loss of 12.4% at 30 nmol/kg) by once-daily repeated dosing for 2 weeks in mice with diet-induced obesity.


Journal of Medicinal Chemistry | 2016

Discovery of 5-Chloro-1-(5-chloro-2-(methylsulfonyl)benzyl)-2-imino-1,2-dihydropyridine-3-carboxamide (TAK-259) as a Novel, Selective, and Orally Active α1D Adrenoceptor Antagonist with Antiurinary Frequency Effects: Reducing Human Ether-a-go-go-Related Gene (hERG) Liabilities

Nobuki Sakauchi; Yasuhisa Kohara; Ayumu Sato; Tomohiko Suzaki; Yumi N. Imai; Yuichi Okabe; Shigemitsu Imai; Reiko Saikawa; Hiroshi Nagabukuro; Haruhiko Kuno; Hisashi Fujita; Izumi Kamo; Masato Yoshida

A novel structural class of iminopyridine derivative 1 was identified as a potent and selective human α1D adrenoceptor (α1D adrenergic receptor; α1D-AR) antagonist against α1A- and α1B-AR through screening of an in-house compound library. From initial structure-activity relationship studies, we found lead compound 9m with hERG K(+) channel liability. To develop analogues with reduced hERG K(+) channel inhibition, a combination of site-directed mutagenesis and docking studies was employed. Further optimization led to the discovery of (R)-9s and 9u, which showed antagonistic activity by a bladder strip test in rats with bladder outlet obstruction, as well as ameliorated cystitis-induced urinary frequency in rats. Ultimately, 9u was selected as a clinical candidate. This is the first study to show the utility of iminopyridine derivatives as selective α1D-AR antagonists and evaluate their effects in vivo.

Collaboration


Dive into the Hisashi Fujita's collaboration.

Top Co-Authors

Avatar

Naoki Nishizawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Shiro Takekawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Taiji Asami

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Junichi Sakamoto

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masahiro Oka

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masato Yoshida

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Yoko Kanematsu-Yamaki

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Akira Mori

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Atsuo Baba

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Haruhiko Makino

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge