Network


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

Hotspot


Dive into the research topics where Hidetoshi Komatsu is active.

Publication


Featured researches published by Hidetoshi Komatsu.


Nature | 2003

Free fatty acids regulate insulin secretion from pancreatic beta cells through GPR40.

Yasuaki Itoh; Yuji Kawamata; Masataka Harada; Makoto Kobayashi; Ryo Fujii; Shoji Fukusumi; Kazuhiro Ogi; Masaki Hosoya; Yasuhiro Tanaka; Hiroshi Uejima; Minoru Maruyama; Rie Satoh; Shoichi Okubo; Hideki Kizawa; Hidetoshi Komatsu; Fumika Matsumura; Yuko Noguchi; Tokuyuki Shinohara; Shuji Hinuma; Yukio Fujisawa; Masahiko Fujino

Diabetes, a disease in which carbohydrate and lipid metabolism are regulated improperly by insulin, is a serious worldwide health issue. Insulin is secreted from pancreatic β cells in response to elevated plasma glucose, with various factors modifying its secretion. Free fatty acids (FFAs) provide an important energy source as nutrients, and they also act as signalling molecules in various cellular processes, including insulin secretion. Although FFAs are thought to promote insulin secretion in an acute phase, this mechanism is not clearly understood. Here we show that a G-protein-coupled receptor, GPR40, which is abundantly expressed in the pancreas, functions as a receptor for long-chain FFAs. Furthermore, we show that long-chain FFAs amplify glucose-stimulated insulin secretion from pancreatic β cells by activating GPR40. Our results indicate that GPR40 agonists and/or antagonists show potential for the development of new anti-diabetic drugs.


PLOS ONE | 2013

A Novel Antidiabetic Drug, Fasiglifam/TAK-875, Acts as an Ago-Allosteric Modulator of FFAR1

Chiori Yabuki; Hidetoshi Komatsu; Yoshiyuki Tsujihata; Risa Maeda; Ryo Ito; Kae Matsuda-Nagasumi; Kensuke Sakuma; Kazumasa Miyawaki; Naoya Kikuchi; Koji Takeuchi; Yugo Habata; Masaaki Mori

Selective free fatty acid receptor 1 (FFAR1)/GPR40 agonist fasiglifam (TAK-875), an antidiabetic drug under phase 3 development, potentiates insulin secretion in a glucose-dependent manner by activating FFAR1 expressed in pancreatic β cells. Although fasiglifam significantly improved glycemic control in type 2 diabetes patients with a minimum risk of hypoglycemia in a phase 2 study, the precise mechanisms of its potent pharmacological effects are not fully understood. Here we demonstrate that fasiglifam acts as an ago-allosteric modulator with a partial agonistic activity for FFAR1. In both Ca2+ influx and insulin secretion assays using cell lines and mouse islets, fasiglifam showed positive cooperativity with the FFAR1 ligand γ-linolenic acid (γ-LA). Augmentation of glucose-induced insulin secretion by fasiglifam, γ-LA, or their combination was completely abolished in pancreatic islets of FFAR1-knockout mice. In diabetic rats, the insulinotropic effect of fasiglifam was suppressed by pharmacological reduction of plasma free fatty acid (FFA) levels using a lipolysis inhibitor, suggesting that fasiglifam potentiates insulin release in conjunction with plasma FFAs in vivo. Point mutations of FFAR1 differentially affected Ca2+ influx activities of fasiglifam and γ-LA, further indicating that these agonists may bind to distinct binding sites. Our results strongly suggest that fasiglifam is an ago-allosteric modulator of FFAR1 that exerts its effects by acting cooperatively with endogenous plasma FFAs in human patients as well as diabetic animals. These findings contribute to our understanding of fasiglifam as an attractive antidiabetic drug with a novel mechanism of action.


PLOS ONE | 2014

Anatomical Transcriptome of G Protein-Coupled Receptors Leads to the Identification of a Novel Therapeutic Candidate GPR52 for Psychiatric Disorders

Hidetoshi Komatsu; Minoru Maruyama; Shuuhei Yao; Tokuyuki Shinohara; Kensuke Sakuma; Sachiko Imaichi; Tomoko Chikatsu; Kanako Kuniyeda; Foo Kok Siu; Lam Sock Peng; Katherine Zhuo; Lay Sock Mun; Tan Min Han; Yoshio Matsumoto; Tadatoshi Hashimoto; Nobuyuki Miyajima; Yasuaki Itoh; Kazuhiro Ogi; Yugo Habata; Masaaki Mori

Many drugs of abuse and most neuropharmacological agents regulate G protein-coupled receptors (GPCRs) in the central nervous system (CNS)_ENREF_1. The striatum, in which dopamine D1 and D2 receptors are enriched, is strongly innervated by the ventral tegmental area (VTA), which is the origin of dopaminergic cell bodies of the mesocorticolimbic dopamine system_ENREF_3 and plays a central role in the development of psychiatric disorders_ENREF_4. Here we report the comprehensive and anatomical transcript profiling of 322 non-odorant GPCRs in mouse tissue by quantitative real-time PCR (qPCR), leading to the identification of neurotherapeutic receptors exclusively expressed in the CNS, especially in the striatum. Among them, GPR6, GPR52, and GPR88, known as orphan GPCRs, were shown to co-localize either with a D2 receptor alone or with both D1 and D2 receptors in neurons of the basal ganglia. Intriguingly, we found that GPR52 was well conserved among vertebrates, is Gs-coupled and responsive to the antipsychotic drug, reserpine. We used three types of transgenic (Tg) mice employing a Cre-lox system under the control of the GPR52 promoter, namely, GPR52-LacZ Tg, human GPR52 (hGPR52) Tg, and hGPR52-GFP Tg mice. Detailed histological investigation suggests that GPR52 may modulate dopaminergic and glutamatergic transmission in neuronal circuits responsible for cognitive function and emotion. In support of our prediction, GPR52 knockout and transgenic mice exhibited psychosis-related and antipsychotic-like behaviors, respectively. Therefore, we propose that GPR52 has the potential of being a therapeutic psychiatric receptor. This approach may help identify potential therapeutic targets for CNS diseases.


The Journal of Neuroscience | 2015

Neuromodulatory Effect of Gαs- or Gαq-Coupled G-Protein-Coupled Receptor on NMDA Receptor Selectively Activates the NMDA Receptor/Ca2+/Calcineurin/cAMP Response Element-Binding Protein-Regulated Transcriptional Coactivator 1 Pathway to Effectively Induce Brain-Derived Neurotrophic Factor Expression in Neurons

Mamoru Fukuchi; Akiko Tabuchi; Yuki Kuwana; Shinjiro Watanabe; Minami Inoue; Ichiro Takasaki; Hironori Izumi; Ayumi Tanaka; Ran Inoue; Hisashi Mori; Hidetoshi Komatsu; Hiroshi Takemori; Hiroyuki Okuno; Haruhiko Bito; Masaaki Tsuda

Although coordinated molecular signaling through excitatory and modulatory neurotransmissions is critical for the induction of immediate early genes (IEGs), which lead to effective changes in synaptic plasticity, the intracellular mechanisms responsible remain obscure. Here we measured the expression of IEGs and used bioluminescence imaging to visualize the expression of Bdnf when GPCRs, major neuromodulator receptors, were stimulated. Stimulation of pituitary adenylate cyclase-activating polypeptide (PACAP)-specific receptor (PAC1), a Gαs/q-protein-coupled GPCR, with PACAP selectively activated the calcineurin (CN) pathway that is controlled by calcium signals evoked via NMDAR. This signaling pathway then induced the expression of Bdnf and CN-dependent IEGs through the nuclear translocation of CREB-regulated transcriptional coactivator 1 (CRTC1). Intracerebroventricular injection of PACAP and intraperitoneal administration of MK801 in mice demonstrated that functional interactions between PAC1 and NMDAR induced the expression of Bdnf in the brain. Coactivation of NMDAR and PAC1 synergistically induced the expression of Bdnf attributable to selective activation of the CN pathway. This CN pathway-controlled expression of Bdnf was also induced by stimulating other Gαs- or Gαq-coupled GPCRs, such as dopamine D1, adrenaline β, CRF, and neurotensin receptors, either with their cognate agonists or by direct stimulation of the protein kinase A (PKA)/PKC pathway with chemical activators. Thus, the GPCR-induced expression of IEGs in coordination with NMDAR might occur via the selective activation of the CN/CRTC1/CREB pathway under simultaneous excitatory and modulatory synaptic transmissions in neurons if either the Gαs/adenylate cyclase/PKA or Gαq/PLC/PKC-mediated pathway is activated.


PLOS ONE | 2015

Temporal and Spatial Transcriptional Fingerprints by Antipsychotic or Propsychotic Drugs in Mouse Brain

Kensuke Sakuma; Hidetoshi Komatsu; Minoru Maruyama; Sachiko Imaichi; Yugo Habata; Masaaki Mori

Various types of antipsychotics have been developed for the treatment of schizophrenia since the accidental discovery of the antipsychotic activity of chlorpromazine. Although all clinically effective antipsychotic agents have common properties to interact with the dopamine D2 receptor (D2R) activation, their precise mechanisms of action remain elusive. Antipsychotics are well known to induce transcriptional changes of immediate early genes (IEGs), raising the possibility that gene expressions play an essential role to improve psychiatric symptoms. Here, we report that while different classes of antipsychotics have complex pharmacological profiles against D2R, they share common transcriptome fingerprint (TFP) profile of IEGs in the murine brain in vivo by quantitative real-time PCR (qPCR). Our data showed that various types of antipsychotics with a profound interaction of D2R including haloperidol (antagonist), olanzapine (antagonist), and aripiprazole (partial agonist) all share common spatial TFPs closely homologous to those of D2R antagonist sulpiride, and elicited greater transcriptional responses in the striatum than in the nucleus accumbens. Meanwhile, D2R agonist quinpirole and propsychotic NMDA antagonists such as MK-801 and phencyclidine (PCP) exhibited the contrasting TFP profiles. Clozapine and propsychotic drug methamphetamine (MAP) displayed peculiar TFPs that reflect their unique pharmacological property. Our results suggest that transcriptional responses are conserved across various types of antipsychotics clinically effective in positive symptoms of schizophrenia and also show that temporal and spatial TFPs may reflect the pharmacological features of the drugs. Thus, we propose that a TFP approach is beneficial to evaluate novel drug candidates for antipsychotic development.


Pharmacology Research & Perspectives | 2016

Fasiglifam (TAK‐875) has dual potentiating mechanisms via Gαq‐GPR40/FFAR1 signaling branches on glucose‐dependent insulin secretion

Kensuke Sakuma; Chiori Yabuki; Minoru Maruyama; Akiko Abiru; Hidetoshi Komatsu; Nobuyuki Negoro; Yoshiyuki Tsujihata; Koji Takeuchi; Yugo Habata; Masaaki Mori

Fasiglifam (TAK‐875) is a free fatty acid receptor 1 (FFAR1)/G‐protein–coupled receptor 40 (GPR40) agonist that improves glycemic control in type 2 diabetes with minimum risk of hypoglycemia. Fasiglifam potentiates glucose‐stimulated insulin secretion (GSIS) from pancreatic β‐cells glucose dependently, although the precise mechanism underlying the glucose dependency still remains unknown. Here, we investigated key cross‐talk between the GSIS pathway and FFAR1 signaling, and Ca2+ dynamics using mouse insulinoma MIN6 cells. We demonstrated that the glucose‐dependent insulinotropic effect of fasiglifam required membrane depolarization and that fasiglifam induced a glucose‐dependent increase in intracellular Ca2+ level and amplification of Ca2+ oscillations. This differed from the sulfonylurea glimepiride that induced changes in Ca2+ dynamics glucose independently. Stimulation with cell‐permeable analogs of IP3 or diacylglycerol (DAG), downstream second messengers of Gαq‐FFAR1, augmented GSIS similar to fasiglifam, indicating their individual roles in the potentiation of GSIS pathway. Intriguingly, the IP3 analog triggered similar Ca2+ dynamics to fasiglifam, whereas the DAG analog had no effect. Despite the lack of an effect on Ca2+ dynamics, the DAG analog elicited synergistic effects on insulin secretion with Ca2+ influx evoked by an L‐type voltage‐dependent calcium channel opener that mimics glucose‐dependent Ca2+ dynamics. These results indicate that the Gαq signaling activated by fasiglifam enhances GSIS pathway via dual potentiating mechanisms in which IP3 amplifies glucose‐induced Ca2+ oscillations and DAG/protein kinase C (PKC) augments downstream secretory mechanisms independent of Ca2+ oscillations.


Journal of Pharmacology and Experimental Therapeutics | 2017

FTBMT, a Novel and Selective GPR52 Agonist, Demonstrates Antipsychotic-Like and Procognitive Effects in Rodents, Revealing a Potential Therapeutic Agent for Schizophrenia

Keiji Nishiyama; Hirobumi Suzuki; Toshiya Harasawa; Noriko Suzuki; Emi Kurimoto; Takayuki Kawai; Minoru Maruyama; Hidetoshi Komatsu; Kensuke Sakuma; Yuji Shimizu; Masato Shimojo

GPR52 is a Gs-coupled G protein–coupled receptor that is predominantly expressed in the striatum and nucleus accumbens (NAc) and was recently proposed as a potential therapeutic target for schizophrenia. In the current study, we investigated the in vitro and in vivo pharmacologic activities of a novel GPR52 agonist, 4-(3-(3-fluoro-5-(trifluoromethyl)benzyl)-5-methyl-1H-1,2,4-triazol-1-yl)-2-methylbenzamide (FTBMT). FTBMT functioned as a selective GPR52 agonist in vitro and in vivo, as demonstrated by the activation of Camp signaling in striatal neurons. FTBMT inhibited MK-801–induced hyperactivity, an animal model for acute psychosis, without causing catalepsy in mice. The c-fos expression also revealed that FTBMT preferentially induced neuronal activation in the shell of the Nac compared with the striatum, thereby supporting its antipsychotic-like activity with less catalepsy. Furthermore, FTBMT improved recognition memory in a novel object-recognition test and attenuated MK-801–induced working memory deficits in a radial arm maze test in rats. These recognitive effects were supported by the results of FTBMT-induced c-fos expression in the brain regions related to cognition, including the medial prefrontal cortex, entorhinal cortex, and hippocampus. Taken together, these findings suggest that FTBMT shows antipsychotic and recognitive properties without causing catalepsy in rodents. Given its unique pharmacologic profile, which differs from that of current antipsychotics, FTBMT may provide a new therapeutic option for the treatment of positive and cognitive symptoms of schizophrenia.


Journal of Biological Chemistry | 2003

A New Peptidic Ligand and Its Receptor Regulating Adrenal Function in Rats

Shoji Fukusumi; Hiromi Yoshida; Ryo Fujii; Minoru Maruyama; Hidetoshi Komatsu; Yugo Habata; Yasushi Shintani; Shuji Hinuma; Masahiko Fujino


Journal of Biological Chemistry | 2004

Identification of a G Protein-coupled Receptor Specifically Responsive to β-Alanine

Tokuyuki Shinohara; Masataka Harada; Kazuhiro Ogi; Minoru Maruyama; Ryo Fujii; Hideyuki Tanaka; Shoji Fukusumi; Hidetoshi Komatsu; Masaki Hosoya; Yuko Noguchi; Takuya Watanabe; Takeo Moriya; Yasuaki Itoh; Shuji Hinuma


Archive | 2003

Method of determining ligand

Shuji Hinuma; Ryo Fujii; Kazuhiro Ogi; Hidetoshi Komatsu; Yuji Kawamata; Masaki Hosoya

Collaboration


Dive into the Hidetoshi Komatsu's collaboration.

Top Co-Authors

Avatar

Yugo Habata

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Minoru Maruyama

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Ryo Fujii

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Shuji Hinuma

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masaki Hosoya

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Kensuke Sakuma

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masaaki Mori

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Shoji Fukusumi

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Kazuhiro Ogi

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masataka Harada

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge