Network


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

Hotspot


Dive into the research topics where Kosuke Aritake is active.

Publication


Featured researches published by Kosuke Aritake.


Journal of Immunology | 2002

Pronounced eosinophilic lung inflammation and Th2 cytokine release in human lipocalin-type prostaglandin D synthase transgenic mice

Yasushi Fujitani; Yoshihide Kanaoka; Kosuke Aritake; Nobuko Uodome; Kazue Okazaki-Hatake; Yoshihiro Urade

PGD2 is a major lipid mediator released from mast cells, but little is known about its role in the development of allergic reactions. We used transgenic (TG) mice overexpressing human lipocalin-type PGD synthase to examine the effect of overproduction of PGD2 in an OVA-induced murine asthma model. The sensitization of wild-type (WT) and TG mice was similar as judged by the content of OVA-specific IgE. After OVA challenge, PGD2, but not PGE2, substantially increased in the lungs of WT and TG mice with greater PGD2 increment in TG mice compared with WT mice. The numbers of eosinophils and lymphocytes in the bronchoalveolar lavage (BAL) fluid were significantly greater in TG mice than in WT mice on days 1 and 3 post-OVA challenge, whereas the numbers of macrophages and neutrophils were the same in both WT and TG mice. The levels of IL-4, IL-5, and eotaxin in BAL fluid were also significantly higher in TG mice than in WT mice, although the level of IFN-γ in the BAL fluid of TG mice was decreased compared with that in WT mice. Furthermore, lymphocytes isolated from the lungs of TG mice secreted less IFN-γ than those from WT mice, whereas IL-4 production was unchanged between WT and TG mice. Thus, overproduction of PGD2 caused an increase in the levels of Th2 cytokines and a chemokine, accompanied by the enhanced accumulation of eosinophils and lymphocytes in the lung. These results indicate that PGD2 plays an important role in late phase allergic reactions in the pathophysiology of bronchial asthma.


Journal of Immunology | 2006

Prostaglandin D2 Plays an Essential Role in Chronic Allergic Inflammation of the Skin via CRTH2 Receptor

Takahiro Satoh; Rie Moroi; Kosuke Aritake; Yoshihiro Urade; Yasumasa Kanai; Koji Sumi; Hiroo Yokozeki; Hiroyuki Hirai; Kinya Nagata; Toshifumi Hara; Masanori Utsuyama; Katsuiku Hirokawa; Kazuo Sugamura; Kiyoshi Nishioka; Masataka Nakamura

PGD2 plays roles in allergic inflammation via specific receptors, the PGD receptor designated DP and CRTH2 (chemoattractant receptor homologous molecule expressed on Th2 cells). We generated mutant mice carrying a targeted disruption of the CRTH2 gene to investigate the functional roles of CRTH2 in cutaneous inflammatory responses. CRTH2-deficent mice were fertile and grew normally. Ear-swelling responses induced by hapten-specific IgE were less pronounced in mutant mice, giving 35–55% of the responses of normal mice. Similar results were seen in mice treated with a hemopoietic PGD synthase inhibitor, HQL-79, or a CRTH2 antagonist, ramatroban. The reduction in cutaneous responses was associated with decreased infiltration of lymphocytes, eosinophils, and basophils and decreased production of macrophage-derived chemokine and RANTES at inflammatory sites. In models of chronic contact hypersensitivity induced by repeated hapten application, CRTH2 deficiency resulted in a reduction by approximately half of skin responses and low levels (63% of control) of serum IgE production, although in vivo migration of Langerhans cells and dendritic cells to regional lymph nodes was not impaired in CRTH2-deficient mice. In contrast, delayed-type hypersensitivity to SRBC and irritation dermatitis in mutant mice were the same as in wild-type mice. These findings indicate that the PGD2-CRTH2 system plays a significant role in chronic allergic skin inflammation. CRTH2 may represent a novel therapeutic target for treatment of human allergic disorders, including atopic dermatitis.


Immunity | 2011

Silica Crystals and Aluminum Salts Regulate the Production of Prostaglandin in Macrophages via NALP3 Inflammasome-Independent Mechanisms

Etsushi Kuroda; Ken J. Ishii; Satoshi Uematsu; Keiichi Ohata; Cevayir Coban; Shizuo Akira; Kosuke Aritake; Yoshihiro Urade; Yasuo Morimoto

Particulates such as silica crystal (silica) and aluminum salts (alum) activate the inflammasome and induce the secretion of proinflammatory cytokines in macrophages. These particulates also induce the production of immunoglobulin E via a T helper 2 (Th2) cell-associated mechanism. However, the mechanism involved in the induction of type 2 immunity has not been elucidated. Here, we showed that silica and alum induced lipopolysaccharide-primed macrophages to produce the lipid mediator prostaglandin E₂ (PGE₂) and interleukin-1β (IL-1β). Macrophages deficient in the inflammasome components caspase 1, NALP3, and ASC revealed that PGE₂ production was independent of the NALP3 inflammasome. PGE₂ expression was markedly reduced in PGE synthase-deficient (Ptges⁻/⁻) macrophages, and Ptges⁻/⁻ mice displayed reduced antigen-specific serum IgE concentrations after immunization with alum or silica. Our results indicate that silica and alum regulate the production of PGE₂ and that the induction of PGE₂ by particulates controls the immune response in vivo.


The Journal of Neuroscience | 2006

Prostaglandin D2-Mediated Microglia/Astrocyte Interaction Enhances Astrogliosis and Demyelination in twitcher

Ikuko Mohri; Masako Taniike; Hidetoshi Taniguchi; Takahisa Kanekiyo; Kosuke Aritake; Takashi Inui; Noriko Fukumoto; Naomi Eguchi; Atsuko Kushi; Hitoshi Sasai; Yoshihide Kanaoka; Keiichi Ozono; Shuh Narumiya; Kinuko Suzuki; Yoshihiro Urade

Prostaglandin (PG) D2 is well known as a mediator of inflammation. Hematopoietic PGD synthase (HPGDS) is responsible for the production of PGD2 involved in inflammatory responses. Microglial activation and astrogliosis are commonly observed during neuroinflammation, including that which occurs during demyelination. Using the genetic demyelination mouse twitcher, a model of human Krabbe’s disease, we discovered that activated microglia expressed HPGDS and activated astrocytes expressed the DP1 receptor for PGD2 in the brain of these mice. Cultured microglia actively produced PGD2 by the action of HPGDS. Cultured astrocytes expressed two types of PGD2 receptor, DP1 and DP2, and showed enhanced GFAP production after stimulation of either receptor with its respective agonist. These results suggest that PGD2 plays an important role in microglia/astrocyte interaction. We demonstrated that the blockade of the HPGDS/PGD2/DP signaling pathway using HPGDS- or DP1-null twitcher mice, and twitcher mice treated with an HPGDS inhibitor, HQL-79 (4-benzhydryloxy-1-[3-(1H-tetrazol-5-yl)-propyl]piperidine), resulted in remarkable suppression of astrogliosis and demyelination, as well as a reduction in twitching and spasticity. Furthermore, we found that the degree of oligodendroglial apoptosis was also reduced in HPGDS-null and HQL-79-treated twitcher mice. These results suggest that PGD2 is the key neuroinflammatory molecule that heightens the pathological response to demyelination in twitcher mice.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Lipocalin-type prostaglandin D synthase produces prostaglandin D2 involved in regulation of physiological sleep.

Wei-Min Qu; Zhi-Li Huang; Xin Hong Xu; Kosuke Aritake; Naomi Eguchi; Fumio Nambu; Shu Narumiya; Yoshihiro Urade; Osamu Hayaishi

Prostaglandin (PG) D2 has been proposed to be essential for the initiation and maintenance of the physiological sleep of rats because intracerebroventricular administration of selenium tetrachloride (SeCl4), a selective inhibitor of PGD synthase (PGDS), was shown to reduce promptly and effectively the amounts of sleep during the period of infusion. However, gene knockout (KO) mice of PGDS and prostaglandin D receptor (DP1R) showed essentially the same circadian profiles and daily amounts of sleep as wild-type (WT) mice, raising questions about the involvement of PGD2 in regulating physiological sleep. Here we examined the effect of SeCl4 on the sleep of WT and KO mice for PGDS and DP1R and that of a DP1R antagonist, ONO-4127Na, on the sleep of rats. The i.p. injection of SeCl4 into WT mice decreased the PGD2 content in the brain without affecting the amounts of PGE2 and PGF2α. It inhibited sleep dose-dependently and immediately after the administration during the light period when mice normally sleep, increasing the wake time; and the treatment with this compound resulted in a distinct sleep rebound during the following dark period. The SeCl4-induced insomnia was observed in hematopoietic PGDS KO mice but not at all in lipocalin-type PGDS KO, hematopoietic and lipocalin-type PGDS double KO or DP1R KO mice. Furthermore, the DP1R antagonist ONO-4127Na reduced sleep of rats by 30% during infusion into the subarachnoid space under the rostral basal forebrain at 200 pmol/min. These results clearly show that the lipocalin-type PGDS/PGD2/DP1R system plays pivotal roles in the regulation of physiological sleep.


Journal of Pharmacology and Experimental Therapeutics | 2008

Lipopolysaccharide induces macrophage migration via prostaglandin D(2) and prostaglandin E(2).

Tsuyoshi Tajima; Takahisa Murata; Kosuke Aritake; Yoshihiro Urade; Hiroyuki Hirai; Masataka Nakamura; Hiroshi Ozaki; Masatoshi Hori

Lipopolysaccharide (LPS) produces prostaglandins (PGs) concomitant to eliciting macrophage migration. We evaluated the role of PGs in initiating the migration of macrophages, especially focusing on PGD2 and PGE2. In RAW264.7 macrophages, cyclooxygenase (COX)-2 inhibitor, CAY10404 [3-(4-methylsulphonylphenyl)-4-phenyl-5-trifluoromethylisoxazole], completely inhibited LPS-mediated migration at 4 h (early phase) but only partially inhibited the migration at 8 h (late phase), suggesting the presence of PG-dependent and -independent pathways. In the early phase, LPS up-regulated mRNA expressions of COX-2, hematopoietic PGD synthase (H-PGDS), and microsomal-PGE synthase 1, increasing PGD2 and PGE2 substantially. The chemoattractant receptor-homologous molecule expressed on Th2 lymphocytes (CRTH2) agonist, DK-PGD2 (13–14-dihydro-15-keto-PGD2), and the EP4 agonist, ONO-AE1-329 (16-{3-methoxymethyl}phenyl-ω-tetranor-3,7-dithia-prostaglandin E1), but not selective agonists of D prostanoid receptor, E prostanoid receptor (EP) 2, or EP3, stimulated random migration (chemokinesis). In peritoneal macrophages from CRTH2-deficient and H-PGDS-deficient mice, LPS-mediated migration was significantly inhibited at either early or late phases of the migration. The H-PGDS inhibitor, HQL-79 [4-(diphenylmethoxy)-1-[3-(1H-tetrazol-5-yl)propyl-piperidine]], partially inhibited the migration of the RAW264.7 macrophage in both phases. These results suggest the importance of the PGD2/CRTH2 pathway in LPS-mediated migration of macrophages. In the late phase of migration, LPS up-regulated monocyte chemoattractant protein (MCP)-1 mRNA. The CC chemokine receptor (CCR2) antagonist, RS102895 [1′-[2-[4-(trifluoromethyl)phenyl]ethyl]-spiro[4H-3,1-benzoxazine-4,4′-piperidin]-2(1H)-one], inhibited LPS-mediated migration in the late phase without affecting the early phase. ONO-AE1-329, but not DK-PGD2, up-regulated MCP-1 mRNA. Taken together, LPS stimulation of chemokinesis or chemotaxis, or both, occurs in macrophages via PGD2 and PGE2 in tandem arrangement; i.e., 1) LPS stimulates prostaglandin signaling, initiating early migration through the PGD2/CRTH2 and PGE2/EP4 signaling pathways; and 2) LPS leads induction of MCP-1, which contributes to later phase migration of the macrophages through the PGE2/EP4 pathway.


Development | 2009

The PGD2 pathway, independently of FGF9, amplifies SOX9 activity in Sertoli cells during male sexual differentiation.

Brigitte Moniot; Faustine Declosmenil; Francisco Barrionuevo; Gerd Scherer; Kosuke Aritake; Safia Malki; Laetitia Marzi; Anne Cohen-Solal; Ina Georg; Jürgen Klattig; Christoph Englert; Yuna Kim; Blanche Capel; Naomi Eguchi; Yoshihiro Urade; Brigitte Boizet-Bonhoure; Francis Poulat

Activation by the Y-encoded testis determining factor SRY and maintenance of expression of the Sox9 gene encoding the central transcription factor of Sertoli cell differentiation are key events in the mammalian sexual differentiation program. In the mouse XY gonad, SOX9 upregulates Fgf9, which initiates a Sox9/Fgf9 feedforward loop, and Sox9 expression is stimulated by the prostaglandin D2 (PGD2) producing lipocalin prostaglandin D synthase (L-PGDS, or PTDGS) enzyme, which accelerates commitment to the male pathway. In an attempt to decipher the genetic relationships between Sox9 and the L-Pgds/PGD2 pathway during mouse testicular organogenesis, we found that ablation of Sox9 at the onset or during the time window of expression in embryonic Sertoli cells abolished L-Pgds transcription. By contrast, L-Pgds-/- XY embryonic gonads displayed a reduced level of Sox9 transcript and aberrant SOX9 protein subcellular localization. In this study, we demonstrated genetically that the L-Pgds/PGD2 pathway acts as a second amplification loop of Sox9 expression. Moreover, examination of Fgf9-/- and L-Pgds-/- XY embryonic gonads demonstrated that the two Sox9 gene activity amplifying pathways work independently. These data suggest that, once activated and maintained by SOX9, production of testicular L-PGDS leads to the accumulation of PGD2, which in turn activates Sox9 transcription and nuclear translocation of SOX9. This mechanism participates together with FGF9 as an amplification system of Sox9 gene expression and activity during mammalian testicular organogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Lipocalin-type prostaglandin D synthase/β-trace is a major amyloid β-chaperone in human cerebrospinal fluid

Takahisa Kanekiyo; Tadato Ban; Kosuke Aritake; Zhi-Li Huang; Wei-Min Qu; Issay Okazaki; Ikuko Mohri; Shigeo Murayama; Keiichi Ozono; Masako Taniike; Yuji Goto; Yoshihiro Urade

The conformational change in amyloid β (Aβ) peptide from its monomeric form to aggregates is crucial in the pathogenesis of Alzheimers disease (AD). In the healthy brain, some unidentified chaperones appear to prevent the aggregation of Aβ. Here we reported that lipocalin-type prostaglandin D synthase (L-PGDS)/β-trace, the most abundant cerebrospinal fluid (CSF) protein produced in the brain, was localized in amyloid plaques in both AD patients and AD-model Tg2576 mice. Surface plasmon resonance analysis revealed that L-PGDS/β-trace tightly bound to Aβ monomers and fibrils with high affinity (KD = 18–50 nM) and that L-PGDS/β-trace recognized residues 25–28 in Aβ, which is the key region for its conformational change to a β-sheet structure. The results of a thioflavin T fluorescence assay to monitor Aβ aggregation disclosed that L-PGDS/β-trace inhibited the spontaneous aggregation of Aβ (1–40) and Aβ (1–42) within its physiological range (1–5 μM) in CSF. L-PGDS/β-trace also prevented the seed-dependent aggregation of 50 μM Aβ with Ki of 0.75 μM. Moreover, the inhibitory activity toward Aβ (1–40) aggregation in human CSF was decreased by 60% when L-PGDS/β-trace was removed from the CSF by immunoaffinity chromatography. The deposition of Aβ after intraventricular infusion of Aβ (1–42) was 3.5-fold higher in L-PGDS-deficient mice and reduced to 23% in L-PGDS-overexpressing mice as compared with their wild-type levels. These data indicate that L-PGDS/β-trace is a major endogenous Aβ-chaperone in the brain and suggest that the disturbance of this function may be involved in the onset and progression of AD. Our findings may provide a diagnostic and therapeutic approach for AD.


Journal of Biological Chemistry | 2006

Structural and Functional Characterization of HQL-79, an Orally Selective Inhibitor of Human Hematopoietic Prostaglandin D Synthase.

Kosuke Aritake; Yuji Kado; Tsuyoshi Inoue; Masashi Miyano; Yoshihiro Urade

We determined the crystal structure of human hematopoietic prostaglandin (PG) D synthase (H-PGDS) as the quaternary complex with glutathione (GSH), Mg2+, and an inhibitor, HQL-79, having anti-inflammatory activities in vivo, at a 1.45-Å resolution. In the quaternary complex, HQL-79 was found to reside within the catalytic cleft between Trp104 and GSH. HQL-79 was stabilized by interaction of a phenyl ring of its diphenyl group with Trp104 and by its piperidine group with GSH and Arg14 through water molecules, which form a network with hydrogen bonding and salt bridges linked to Mg2+. HQL-79 inhibited human H-PGDS competitively against the substrate PGH2 and non-competitively against GSH with Ki of 5 and 3 μm, respectively. Surface plasmon resonance analysis revealed that HQL-79 bound to H-PGDS with an affinity that was 12-fold higher in the presence of GSH and Mg2+ (Kd, 0.8 μm) than in their absence. Mutational studies revealed that Arg14 was important for the Mg2+-mediated increase in the binding affinity of H-PGDS for HQL-79, and that Trp104, Lys112, and Lys198 were important for maintaining the HQL-binding pocket. HQL-79 selectively inhibited PGD2 production by H-PGDS-expressing human megakaryocytes and rat mastocytoma cells with an IC50 value of about 100 μm but only marginally affected the production of other prostanoids, suggesting the tight functional engagement between H-PGDS and cyclooxygenase. Orally administered HQL-79 (30 mg/kg body weight) inhibited antigen-induced production of PGD2, without affecting the production of PGE2 and PGF2α, and ameliorated airway inflammation in wild-type and human H-PGDS-overexpressing mice. Knowledge about this structure of quaternary complex is useful for understanding the inhibitory mechanism of HQL-79 and should accelerate the structure-based development of novel anti-inflammatory drugs that inhibit PGD2 production specifically.


Nature Immunology | 2013

Mast cell maturation is driven via a group III phospholipase A 2-prostaglandin D2-DP1 receptor paracrine axis

Yoshitaka Taketomi; Noriko Ueno; Takumi Kojima; Hiroyasu Sato; Remi Murase; Kei Yamamoto; Satoshi Tanaka; Mariko Sakanaka; Masanori Nakamura; Yasumasa Nishito; Momoko Kawana; Naotomo Kambe; Kazutaka Ikeda; Ryo Taguchi; Satoshi Nakamizo; Kenji Kabashima; Michael H. Gelb; Makoto Arita; Takehiko Yokomizo; Motonao Nakamura; Kikuko Watanabe; Hiroyuki Hirai; Masataka Nakamura; Yoshimichi Okayama; Chisei Ra; Kosuke Aritake; Yoshihiro Urade; Kazushi Morimoto; Yukihiko Sugimoto; Takao Shimizu

Microenvironment-based alterations in phenotypes of mast cells influence the susceptibility to anaphylaxis, yet the mechanisms underlying proper maturation of mast cells toward an anaphylaxis-sensitive phenotype are incompletely understood. Here we report that PLA2G3, a mammalian homolog of anaphylactic bee venom phospholipase A2, regulates this process. PLA2G3 secreted from mast cells is coupled with fibroblastic lipocalin-type PGD2 synthase (L-PGDS) to provide PGD2, which facilitates mast-cell maturation via PGD2 receptor DP1. Mice lacking PLA2G3, L-PGDS or DP1, mast cell–deficient mice reconstituted with PLA2G3-null or DP1-null mast cells, or mast cells cultured with L-PGDS–ablated fibroblasts exhibited impaired maturation and anaphylaxis of mast cells. Thus, we describe a lipid-driven PLA2G3–L-PGDS–DP1 loop that drives mast cell maturation.

Collaboration


Dive into the Kosuke Aritake's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ko Fujimori

Osaka University of Pharmaceutical Sciences

View shared research outputs
Top Co-Authors

Avatar

Masataka Nakamura

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Nanae Nagata

Osaka Bioscience Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Naomi Eguchi

Osaka Bioscience Institute

View shared research outputs
Top Co-Authors

Avatar

Tadayasu Ohkubo

Japan Advanced Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge