Network


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

Hotspot


Dive into the research topics where Takami Yurugi-Kobayashi is active.

Publication


Featured researches published by Takami Yurugi-Kobayashi.


Nature | 2012

Structure of the human M2 muscarinic acetylcholine receptor bound to an antagonist.

Kazuko Haga; Andrew C. Kruse; Hidetsugu Asada; Takami Yurugi-Kobayashi; Mitsunori Shiroishi; Cheng Zhang; William I. Weis; Tetsuji Okada; Brian K. Kobilka; Tatsuya Haga; Takuya Kobayashi

The parasympathetic branch of the autonomic nervous system regulates the activity of multiple organ systems. Muscarinic receptors are G-protein-coupled receptors that mediate the response to acetylcholine released from parasympathetic nerves. Their role in the unconscious regulation of organ and central nervous system function makes them potential therapeutic targets for a broad spectrum of diseases. The M2 muscarinic acetylcholine receptor (M2 receptor) is essential for the physiological control of cardiovascular function through activation of G-protein-coupled inwardly rectifying potassium channels, and is of particular interest because of its extensive pharmacological characterization with both orthosteric and allosteric ligands. Here we report the structure of the antagonist-bound human M2 receptor, the first human acetylcholine receptor to be characterized structurally, to our knowledge. The antagonist 3-quinuclidinyl-benzilate binds in the middle of a long aqueous channel extending approximately two-thirds through the membrane. The orthosteric binding pocket is formed by amino acids that are identical in all five muscarinic receptor subtypes, and shares structural homology with other functionally unrelated acetylcholine binding proteins from different species. A layer of tyrosine residues forms an aromatic cap restricting dissociation of the bound ligand. A binding site for allosteric ligands has been mapped to residues at the entrance to the binding pocket near this aromatic cap. The structure of the M2 receptor provides insights into the challenges of developing subtype-selective ligands for muscarinic receptors and their propensity for allosteric regulation.


Nature | 2012

G-protein-coupled receptor inactivation by an allosteric inverse-agonist antibody

Tomoya Hino; Takatoshi Arakawa; Hiroko Iwanari; Takami Yurugi-Kobayashi; Chiyo Ikeda-Suno; Yoshiko Nakada-Nakura; Osamu Kusano-Arai; Simone Weyand; Tatsuro Shimamura; Norimichi Nomura; Alexander D. Cameron; Takuya Kobayashi; Takao Hamakubo; So Iwata; Takeshi Murata

G-protein-coupled receptors are the largest class of cell-surface receptors, and these membrane proteins exist in equilibrium between inactive and active states. Conformational changes induced by extracellular ligands binding to G-protein-coupled receptors result in a cellular response through the activation of G proteins. The A2A adenosine receptor (A2AAR) is responsible for regulating blood flow to the cardiac muscle and is important in the regulation of glutamate and dopamine release in the brain. Here we report the raising of a mouse monoclonal antibody against human A2AAR that prevents agonist but not antagonist binding to the extracellular ligand-binding pocket, and describe the structure of A2AAR in complex with the antibody Fab fragment (Fab2838). This structure reveals that Fab2838 recognizes the intracellular surface of A2AAR and that its complementarity-determining region, CDR-H3, penetrates into the receptor. CDR-H3 is located in a similar position to the G-protein carboxy-terminal fragment in the active opsin structure and to CDR-3 of the nanobody in the active β2-adrenergic receptor structure, but locks A2AAR in an inactive conformation. These results suggest a new strategy to modulate the activity of G-protein-coupled receptors.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Significance and therapeutic potential of the natriuretic peptides/cGMP/cGMP-dependent protein kinase pathway in vascular regeneration

Kenichi Yamahara; Hiroshi Itoh; Tae Hwa Chun; Yoshihiro Ogawa; Jun Yamashita; Naoki Sawada; Yasutomo Fukunaga; Masakatsu Sone; Takami Yurugi-Kobayashi; Kazutoshi Miyashita; Hirokazu Tsujimoto; Hyun Kook; Robert Feil; David L. Garbers; Franz Hofmann; Kazuwa Nakao

Natriuretic peptides (NPs), which consist of atrial, brain, and C-type natriuretic peptides (ANP, BNP, and CNP, respectively), are characterized as cardiac or vascular hormones that elicit their biological effects by activation of the cGMP/cGMP-dependent protein kinase (cGK) pathway. We recently reported that adenoviral gene transfer of CNP into rabbit blood vessels not only suppressed neointimal formation but also accelerated reendothelialization, a required step for endothelium-dependent vasorelaxation and antithrombogenicity. Accordingly, we investigated the therapeutic potential of the NPs/cGMP/cGK pathway for vascular regeneration. In transgenic (Tg) mice that overexpress BNP in response to hindlimb ischemia, neovascularization with appropriate mural cell coating was accelerated without edema or bleeding, and impaired angiogenesis by the suppression of nitric oxide production was effectively rescued. Furthermore, in BNP-Tg mice, inflammatory cell infiltration in ischemic tissue and vascular superoxide production were suppressed compared with control mice. Ischemia-induced angiogenesis was also significantly potentiated in cGK type I Tg mice, but attenuated in cGK type I knockout mice. NPs significantly stimulated capillary network formation of cultured endothelial cells by cGK stimulation and subsequent Erk1/2 activation. Furthermore, gene transfer of CNP into ischemic muscles effectively accelerated angiogenesis. These findings reveal an action of the NPs/cGMP/cGK pathway to exert multiple vasculoprotective and regenerative actions in the absence of apparent adverse effects, and therefore suggest that NPs as the endogenous cardiovascular hormone can be used as a strategy of therapeutic angiogenesis in patients with tissue ischemia.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2007

Pathway for differentiation of human embryonic stem cells to vascular cell components and their potential for vascular regeneration.

Masakatsu Sone; Hiroshi Itoh; Kenichi Yamahara; Jun Yamashita; Takami Yurugi-Kobayashi; Akane Nonoguchi; Yutaka Suzuki; Ting-Hsing Chao; Naoki Sawada; Yasutomo Fukunaga; Kazutoshi Miyashita; Kwijun Park; Naofumi Oyamada; Naoya Sawada; Daisuke Taura; Naohisa Tamura; Yasushi Kondo; Shinji Nito; Hirofumi Suemori; Norio Nakatsuji; Shin-Ichi Nishikawa; Kazuwa Nakao

Objective— We demonstrated previously that mouse embryonic stem (ES) cell–derived vascular endothelial growth factor receptor-2 (VEGF-R2)–positive cells can differentiate into both vascular endothelial cells and mural cells. This time, we investigated kinetics of differentiation of human ES cells to vascular cells and examined their potential as a source for vascular regeneration. Methods and Results— Unlike mouse ES cells, undifferentiated human ES cells already expressed VEGF-R2, but after differentiation, a VEGF-R2-positive but tumor rejection antigen 1-60 (TRA1-60)–negative population emerged. These VEGF-R2-positive but tumor rejection antigen 1-60–negative cells were also positive for platelet-derived growth factor receptor &agr; and &bgr; chains and could be effectively differentiated into both VE-cadherin+ endothelial cell and &agr;-smooth muscle actin+ mural cell. VE-cadherin+ cells, which were also CD34+ and VEGF-R2+ and thought to be endothelial cells in the early differentiation stage, could be expanded while maintaining their maturity. Their transplantation to the hindlimb ischemia model of immunodeficient mice contributed to the construction of new blood vessels and improved blood flow. Conclusions— We could identify the differentiation process from human ES cells to vascular cell components and demonstrate that expansion and transplantation of vascular cells at the appropriate differentiation stage may constitute a novel strategy for vascular regenerative medicine.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2006

Adrenomedullin/Cyclic AMP Pathway Induces Notch Activation and Differentiation of Arterial Endothelial Cells From Vascular Progenitors

Takami Yurugi-Kobayashi; Hiroshi Itoh; Timm Schroeder; Akiko Nakano; Genta Narazaki; Fumiyo Kita; Kentoku Yanagi; Mina Hiraoka-Kanie; Emi Inoue; Toshiaki Ara; Takashi Nagasawa; Ursula Just; Kazuwa Nakao; Shin-Ichi Nishikawa; Jun Yamashita

Objective—The acquisition of arterial or venous identity is highlighted in vascular development. Previously, we have reported an embryonic stem (ES) cell differentiation system that exhibits early vascular development using vascular endothelial growth factor (VEGF) receptor-2 (VEGFR2)-positive cells as common vascular progenitors. In this study, we constructively induced differentiation of arterial and venous endothelial cells (ECs) in vitro to elucidate molecular mechanisms of arterial-venous specification. Methods and Results—ECs were induced from VEGFR2+ progenitor cells with various conditions. VEGF was essential to induce ECs. Addition of 8bromo-cAMP or adrenomedullin (AM), an endogenous ligand-elevating cAMP, enhanced VEGF-induced EC differentiation. Whereas VEGF alone mainly induced venous ECs, 8bromo-cAMP (or AM) with VEGF supported substantial induction of arterial ECs. Stimulation of cAMP pathway induced Notch signal activation in ECs. The arterializing effect of VEGF and cAMP was abolished in recombination recognition sequence binding protein at the J&kgr; site deficient ES cells lacking Notch signal activation or in ES cells treated with &ggr;-secretase inhibitor. Nevertheless, forced Notch activation by the constitutively active Notch1 alone did not induce arterial ECs. Conclusions—Adrenomedullin/cAMP is a novel signaling pathway to activate Notch signaling in differentiating ECs. Coordinated signaling of VEGF, Notch, and cAMP is required to induce arterial ECs from vascular progenitors.


PLOS ONE | 2008

Augmentation of Neovascularizaiton in Hindlimb Ischemia by Combined Transplantation of Human Embryonic Stem Cells-Derived Endothelial and Mural Cells

Kenichi Yamahara; Masakatsu Sone; Hiroshi Itoh; Jun Yamashita; Takami Yurugi-Kobayashi; Koichiro Homma; Ting-Hsing Chao; Kazutoshi Miyashita; Kwijun Park; Naofumi Oyamada; Naoya Sawada; Daisuke Taura; Yasutomo Fukunaga; Naohisa Tamura; Kazuwa Nakao

Background We demonstrated that mouse embryonic stem (ES) cells-derived vascular endothelial growth factor receptor-2 (VEGF-R2) positive cells could differentiate into both endothelial cells (EC) and mural cells (MC), and termed them as vascular progenitor cells (VPC). Recently, we have established a method to expand monkey and human ES cells-derived VPC with the proper differentiation stage in a large quantity. Here we investigated the therapeutic potential of human VPC-derived EC and MC for vascular regeneration. Methods and Results After the expansion of human VPC-derived vascular cells, we transplanted these cells to nude mice with hindlimb ischemia. The blood flow recovery and capillary density in ischemic hindlimbs were significantly improved in human VPC-derived EC-transplanted mice, compared to human peripheral and umbilical cord blood-derived endothelial progenitor cells (pEPC and uEPC) transplanted mice. The combined transplantation of human VPC-derived EC and MC synergistically improved blood flow of ischemic hindlimbs remarkably, compared to the single cell transplantations. Transplanted VPC-derived vascular cells were effectively incorporated into host circulating vessels as EC and MC to maintain long-term vascular integrity. Conclusions Our findings suggest that the combined transplantation of human ES cells-derived EC and MC can be used as a new promising strategy for therapeutic vascular regeneration in patients with tissue ischemia.


Circulation | 2003

Different Differentiation Kinetics of Vascular Progenitor Cells in Primate and Mouse Embryonic Stem Cells

Masakatsu Sone; Hiroshi Itoh; Jun Yamashita; Takami Yurugi-Kobayashi; Yutaka Suzuki; Yasushi Kondo; Akane Nonoguchi; Naoki Sawada; Kenichi Yamahara; Kazutoshi Miyashita; Kwijun Park; Masabumi Shibuya; Shinji Nito; Shin-Ichi Nishikawa; Kazuwa Nakao

Background—We demonstrated that vascular endothelial growth factor receptor 2 (VEGF-R2)-positive cells derived from mouse embryonic stem (ES) cells can differentiate into both endothelial cells and mural cells to suffice as vascular progenitor cells (VPCs). Here we examined whether VPCs occur in primate ES cells and investigated the differences in VPC differentiation kinetics between primate and mouse ES cells. Methods and Results—In contrast to mouse ES cells, undifferentiated monkey ES cells expressed VEGF-R2. By culturing these undifferentiated ES cells for 4 days on OP9 feeder layer, VEGF-R2 expression disappeared, and then reappeared after 8 days of differentiation. We then isolated these VEGF-R2–positive and vascular endothelial cadherin (VEcadherin)-negative cells by flow cytometry sorting. Additional 5-day reculture of these VEGF-R2+ VEcadherin− cells on OP9 feeder layer resulted in the appearance of platelet endothelial cell adhesion molecule-1 (PECAM1)-positive, VEcadherin-positive, endothelial nitric oxide synthase (eNOS)-positive endothelial cells. On a collagen IV-coated dish in the presence of serum, these cells differentiated into smooth muscle actin (SMA)-positive and calponin-positive mural cells (pericytes or vascular smooth muscle cells). Addition of 50ng/mL VEGF to the culture on a collagen IV-coated dish resulted in the appearance of PECAM1+ cells surrounded by SMA+ cells. In addition, these differentiated VEGF-R2+ cells can form tube-like structures in a 3-dimensional culture. Conclusion—Our findings indicate that differentiation kinetics of VPCs derived from primate and mouse ES cells were different. Differentiated VEGF-R2+ VEcadherin− cells can act as VPCs in primates. To seek the clinical potential of VPCs for vascular regeneration, investigations of primate ES cells are indispensable.


FEBS Letters | 2003

Adrenomedullin provokes endothelial Akt activation and promotes vascular regeneration both in vitro and in vivo

Kazutoshi Miyashita; Hiroshi Itoh; Naoki Sawada; Yasutomo Fukunaga; Masakatsu Sone; Kenichi Yamahara; Takami Yurugi-Kobayashi; Kwijun Park; Kazuwa Nakao

We previously reported that adrenomedullin (AM), a vasodilating hormone secreted from blood vessels, promotes proliferation and migration of human umbilical vein endothelial cells (HUVECs). In this study, we examined the ability of AM to promote vascular regeneration. AM increased the phosphorylation of Akt in HUVECs and the effect was inhibited by the AM antagonists and the inhibitors for protein kinase A (PKA) or phosphatidylinositol 3‐kinase (PI3K). AM promoted re‐endothelialization in vitro of wounded monolayer of HUVECs and neo‐vascularization in vivo in murine gel plugs. These effects were also inhibited by the AM antagonists and the inhibitors for PKA or PI3K. The findings suggest that AM plays significant roles in vascular regeneration, associated with PKA‐ and PI3K‐dependent activation of Akt in endothelial cells, and possesses therapeutic potential for vascular injury and tissue ischemia.


Biochemical and Biophysical Research Communications | 2009

Comparison of functional non-glycosylated GPCRs expression in Pichia pastoris

Takami Yurugi-Kobayashi; Hidetsugu Asada; Mitsunori Shiroishi; Tatsuro Shimamura; Saeko Funamoto; Naoko Katsuta; Keisuke Ito; Taishi Sugawara; Natsuko Tokuda; Hirokazu Tsujimoto; Takeshi Murata; Norimichi Nomura; Kazuko Haga; Tatsuya Haga; So Iwata; Takuya Kobayashi

N-linked glycosylation is the most common post-translational modification of G-protein-coupled receptors (GPCRs) and is correlated to the localization and function of the receptors depending on each receptor. However, heterogeneity of glycosylation can interfere with protein crystallization. The removal of N-linked glycosylation from membrane proteins improves the ability to crystallize these proteins. We screened 25 non-glycosylated GPCRs for functional receptor production in the methylotrophic yeast Pichia pastoris using specific ligand-receptor binding assays. We found that five clones were expressed at greater than 10 pmol/mg, 9 clones at 1-10 pmol/mg and 11 clones at less than 1 pmol/mg of membrane protein. Further optimization of culture parameters including culture scale, induction time, pH and temperature enabled us to achieve expression of a functional human muscarinic acetylcholine receptor subtype 2 (CHRM2) with a B(max) value of 51.2 pmol/mg of membrane protein. Approximately 1.9 mg of the human CHRM2 was produced from a 1-L culture.


Microbial Cell Factories | 2012

Platform for the rapid construction and evaluation of GPCRs for crystallography in Saccharomyces cerevisiae

Mitsunori Shiroishi; Hirokazu Tsujimoto; Hisayoshi Makyio; Hidetsugu Asada; Takami Yurugi-Kobayashi; Tatsuro Shimamura; Takeshi Murata; Norimichi Nomura; Tatsuya Haga; So Iwata; Takuya Kobayashi

BackgroundRecent successes in the determination of G-protein coupled receptor (GPCR) structures have relied on the ability of receptor variants to overcome difficulties in expression and purification. Therefore, the quick screening of functionally expressed stable receptor variants is vital.ResultsWe developed a platform using Saccharomyces cerevisiae for the rapid construction and evaluation of functional GPCR variants for structural studies. This platform enables us to perform a screening cycle from construction to evaluation of variants within 6–7 days. We firstly confirmed the functional expression of 25 full-length class A GPCRs in this platform. Then, in order to improve the expression level and stability, we generated and evaluated the variants of the four GPCRs (hADRB2, hCHRM2, hHRH1 and hNTSR1). These stabilized receptor variants improved both functional activity and monodispersity. Finally, the expression level of the stabilized hHRH1 in Pichia pastoris was improved up to 65 pmol/mg from negligible expression of the functional full-length receptor in S. cerevisiae at first screening. The stabilized hHRH1 was able to be purified for use in crystallization trials.ConclusionsWe demonstrated that the S. cerevisiae system should serve as an easy-to-handle and rapid platform for the construction and evaluation of GPCR variants. This platform can be a powerful prescreening method to identify a suitable GPCR variant for crystallography.

Collaboration


Dive into the Takami Yurugi-Kobayashi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hidetsugu Asada

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge