Network


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

Hotspot


Dive into the research topics where Kotaro Sakamoto is active.

Publication


Featured researches published by Kotaro Sakamoto.


Nanotechnology | 2008

Photoinduced antiviral carbon nanohorns

Eijiro Miyako; Hideya Nagata; Ken Hirano; Kotaro Sakamoto; Yoji Makita; Ken-ichi Nakayama; Takahiro Hirotsu

Nanocarbons, such as carbon nanohorns (CNH) and carbon nanotubes, are materials of interest in many fields of science and technology because of their remarkable physical properties. We report here a novel approach for using NIR laser-driven CNH as an antiviral agent. NIR laser-driven functional CNH complexes could open the way to a new range of antiviral materials.


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.


Journal of Medicinal Chemistry | 2017

Discovery of a B-Cell Lymphoma 6 Protein–Protein Interaction Inhibitor by a Biophysics-Driven Fragment-Based Approach

Yusuke Kamada; Nozomu Sakai; Satoshi Sogabe; Koh Ida; Hideyuki Oki; Kotaro Sakamoto; Weston Lane; Gyorgy Snell; Motoo Iida; Yasuhiro Imaeda; Junichi Sakamoto; Junji Matsui

B-cell lymphoma 6 (BCL6) is a transcriptional factor that expresses in lymphocytes and regulates the differentiation and proliferation of lymphocytes. Therefore, BCL6 is a therapeutic target for autoimmune diseases and cancer treatment. This report presents the discovery of BCL6-corepressor interaction inhibitors by using a biophysics-driven fragment-based approach. Using the surface plasmon resonance (SPR)-based fragment screening, we successfully identified fragment 1 (SPR KD = 1200 μM, ligand efficiency (LE) = 0.28), a competitive binder to the natural ligand BCoR peptide. Moreover, we elaborated 1 into the more potent compound 7 (SPR KD = 0.078 μM, LE = 0.37, cell-free protein-protein interaction (PPI) IC50 = 0.48 μM (ELISA), cellular PPI IC50 = 8.6 μM (M2H)) by a structure-based design and structural integration with a second high-throughput screening hit.


Peptides | 2017

PEGylated neuromedin U-8 shows long-lasting anorectic activity and anti-obesity effect in mice by peripheral administration

Yasushi Masuda; Satoshi Kumano; Jiro Noguchi; Kotaro Sakamoto; Hiroshi Inooka; Tetsuya Ohtaki

HIGHLIGHTSIntraperitoneal injection of neuromedin U‐23 and neuromedin S inhibited food intake.PEGylation gave neuromedin U‐8 a long‐lasting anorectic activity.Subcutaneous injection of PEGylated neuromedin U‐8 exerted an anti‐obesity effect. ABSTRACT Neuromedin U (NMU) is a neuropeptide found in the brain and gastrointestinal tract. The NMU system has been shown to regulate energy homeostasis by both a central and a peripheral mechanism. Peripheral administration of human NMU‐25 was recently shown to inhibit food intake in mice. We examined the possibility that other NMU‐related peptides exert an anorectic activity by intraperitoneal (i.p.) administration. We found that rat NMU‐23 and its structurally‐related peptide rat neuromedin S (NMS) significantly reduced food intake in lean mice, whereas NMU‐8, an active fragment of the octapeptide sequence conserved in porcine, human and mouse NMU, had no effect. When rat NMU‐23, NMU‐8, and rat NMS were covalently conjugated to polyethylene glycol (PEG) (PEGylation) at the N‐terminus of these peptides, PEGylated NMU‐8 showed the most long‐lasting and robust anorectic activity. The exploration of the linker between NMU‐8 and PEG using hetero‐bifunctional chemical cross‐linkers led to an identification of PEGylated NMU‐8 analogs with higher affinity for NMU receptors and with more potent anorectic activity in lean mice. The PEGylated NMU‐8 showed potent and robust anorectic activity and anti‐obesity effect in diet‐induced obesity (DIO) mice by once‐daily subcutaneous (s.c.) administration. These results suggest that PEGylated NMU‐8 has the therapeutic potential for treatment of obesity.


Biochemical and Biophysical Research Communications | 2017

Discovery of GPX4 inhibitory peptides from random peptide T7 phage display and subsequent structural analysis

Kotaro Sakamoto; Satoshi Sogabe; Yusuke Kamada; Shin-ichi Matsumoto; Akito Kadotani; Junichi Sakamoto; Akiyoshi Tani

The phospholipid hydroperoxidase glutathione peroxidase (GPX4) is an enzyme that reduces lipid hydroperoxides in lipid membranes. Recently, GPX4 has been investigated as a target molecule that induces iron-dependent cell death (ferroptosis) selectively in cancer cells that express mutant Ras. GPX4 inhibitors have the potential to become novel anti-cancer drugs. However, there are no druggable pockets for conventional small molecules on the molecular surface of GPX4. To generate GPX4 inhibitors, we examined the use of peptides as an alternative to small molecules. By screening peptide libraries displayed on T7 phages, and analyzing the X-ray crystal structures of the peptides, we successfully identified one peptide that binds to near Sec73 of catalytic site and two peptides that bind to another site on GPX4. To our knowledge, this is the first study reporting GPX4 inhibitory peptides and their structural information.


Bioorganic & Medicinal Chemistry Letters | 2017

Investigation of the structural requirements of K-Ras(G12D) selective inhibitory peptide KRpep-2d using alanine scans and cysteine bridging

Ayumu Niida; Shigekazu Sasaki; Kazuko Yonemori; Tomoya Sameshima; Masahiro Yaguchi; Taiji Asami; Kotaro Sakamoto; Masahiro Kamaura

A structure-activity relationship study of a K-Ras(G12D) selective inhibitory cyclic peptide, KRpep-2d was performed. Alanine scanning of KRpep-2d focusing on the cyclic moiety showed that Leu7, Ile9, and Asp12 are the key elements for K-Ras(G12D) selective inhibition of KRpep-2d. The cysteine bridging was also examined to identify the stable analog of KRpep-2d under reductive conditions. As a result, the KRpep-2d analog (12) including mono-methylene bridging showed potent K-Ras(G12D) selective inhibition in both the presence and the absence of dithiothreitol. This means that mono-methylene bridging is an effective strategy to obtain a reduction-resistance analog of parent disulfide cyclic peptides. Peptide 12 inhibited proliferation of K-Ras(G12D)-driven cancer cells significantly. These results gave valuable information for further optimization of KRpep-2d to provide novel anti-cancer drug candidates targeting the K-Ras(G12D) mutant.


Bioorganic & Medicinal Chemistry | 2017

Investigation on cellular uptake and pharmacodynamics of DOCK2-inhibitory peptides conjugated with cell-penetrating peptides

Yusuke Adachi; Kotaro Sakamoto; Tadashi Umemoto; Yasunori Fukuda; Akiyoshi Tani; Taiji Asami

Protein-protein interaction between dedicator of cytokinesis 2 (DOCK2) and Ras-related C3 botulinum toxin substrate 1 (Rac1) is an attractive intracellular target for transplant rejection and inflammatory diseases. Recently, DOCK2-selective inhibitory peptides have been discovered, and conjugation with oligoarginine cell-penetrating peptide (CPP) improved inhibitory activity in a cell migration assay. Although a number of CPPs have been reported, oligoarginine was only one example introduced to the inhibitory peptides. In this study, we aimed to confirm the feasibility of CPP-conjugation approach for DOCK2-inhibitory peptides, and select preferable sequences as CPP moiety. First, we evaluated cell permeability of thirteen known CPPs and partial sequences of influenza A viral protein PB1-F2 using an internalization assay system based on luciferin-luciferase reaction, and then selected four CPPs with efficient cellular uptake. Among four conjugates of these CPPs and a DOCK2-inhibitory peptide, the inhibitory activity of a novel CPP, PB1-F2 fragment 5 (PF5), conjugate was comparable to oligoarginine conjugate and higher than that of the non-conjugated peptide. Finally, internalization assay revealed that oligoarginine and PF5 increased the cellular uptake of inhibitory peptides to the same extent. Hence, we demonstrated that CPP-conjugation approach is applicable to the development of novel anti-inflammatory drugs based on DOCK2 inhibition by investigating both cellular uptake and bioactivity.


Bioorganic & Medicinal Chemistry | 2017

Discovery of a novel B-cell lymphoma 6 (BCL6)–corepressor interaction inhibitor by utilizing structure-based drug design

Takeshi Yasui; Takeshi Yamamoto; Nozomu Sakai; Kouhei Asano; Takafumi Takai; Yayoi Yoshitomi; Melinda Davis; Terufumi Takagi; Kotaro Sakamoto; Satoshi Sogabe; Yusuke Kamada; Weston Lane; Gyorgy Snell; Masashi Iwata; Masayuki Goto; Hiroshi Inooka; Junichi Sakamoto; Yoshihisa Nakada; Yasuhiro Imaeda

B-cell lymphoma 6 (BCL6) is a transcriptional repressor that can form complexes with corepressors via protein-protein interactions (PPIs). The complexes of BCL6 and corepressors play an important role in the formation of germinal centers (GCs), and differentiation and proliferation of lymphocytes. Therefore, BCL6-corepressor interaction inhibitors would be drug candidates for managing autoimmune diseases and cancer. Starting from high-throughput screening hits 1a and 2a, we identified a novel BCL6-corepressor interaction inhibitor 8c (cell-free enzyme-linked immunosorbent assay [ELISA] IC50=0.10µM, cell-based mammalian two-hybrid [M2H] assay IC50=0.72µM) by utilizing structure-based drug design (SBDD) based on an X-ray crystal structure of 1a bound to BCL6. Compound 8c also showed a good pharmacokinetic profile, which was acceptable for both in vitro and in vivo studies.


ACS Medicinal Chemistry Letters | 2017

Crystal Structure of a Human K-Ras G12D Mutant in Complex with GDP and the Cyclic Inhibitory Peptide KRpep-2d

Satoshi Sogabe; Yusuke Kamada; Masanori Miwa; Ayumu Niida; Tomoya Sameshima; Masahiro Kamaura; Kazuko Yonemori; Shigekazu Sasaki; Junichi Sakamoto; Kotaro Sakamoto

The Ras proteins play roles in cell differentiation, proliferation, and survival. Aberrant signaling through Ras-mediated pathways in tumor cells occurs as a result of several types of mutational damage, which most frequently affects the amino acids G12, G13, and Q61. Recently, KRpep-2d was identified as a K-Ras(G12D) selective inhibitory peptide against the G12D mutant of K-Ras, which is a key member of the Ras protein family and an attractive cancer therapeutic target. In this study, the crystal structure of the human K-Ras(G12D) mutant was determined in complex with GDP and KRpep-2d at 1.25 Å resolution. This structure revealed that the peptide binds near Switch II and allosterically blocks protein-protein interactions with the guanine nucleotide exchange factor. This discovery of a unique binding pocket provides valuable information that will facilitate the design of direct Ras inhibitors.


Biochemistry | 2018

Discovery of an irreversible and cell-active BCL6 inhibitor selectively targeting Cys53 located at the protein-protein interaction interface

Tomoya Sameshima; Takeshi Yamamoto; Osamu Sano; Satoshi Sogabe; Shigeru Igaki; Kotaro Sakamoto; Koh Ida; Mika Gotou; Yasuhiro Imaeda; Junichi Sakamoto; Ikuo Miyahisa

B-cell lymphoma 6 (BCL6) is the most frequently involved oncogene in diffuse large B-cell lymphomas (DLBCLs). BCL6 shows potent transcriptional repressor activity through interactions with its corepressors, such as BCL6 corepressor (BCOR). The inhibition of the protein-protein interaction (PPI) between BCL6 and its corepressors suppresses the growth of BCL6-dependent DLBCLs, thus making BCL6 an attractive drug target for lymphoma treatment. However, potent small-molecule PPI inhibitor identification remains challenging because of the lack of deep cavities at PPI interfaces. This article reports the discovery of a potent, cell-active small-molecule BCL6 inhibitor, BCL6-i (8), that operates through irreversible inhibition. First, we synthesized irreversible lead compound 4, which targets Cys53 in a cavity on the BCL6-BTB domain dimer by introducing an irreversible warhead to high-throughput screening hit compound 1. Further chemical optimization of 4 based on kinact/KI evaluation produced BCL6-i with a kinact/KI value of 1.9 × 104 M-1 s-1, corresponding to a 670-fold improvement in potency compared to that of 4. By exploiting the property of irreversible inhibition, engagement of BCL6-i to intracellular BCL6 was confirmed. BCL6-i showed intracellular PPI inhibitory activity between BCL6 and its corepressors, thus resulting in BCL6-dependent DLBCL cell growth inhibition. BCL6-i is a cell-active chemical probe with the most potent BCL6 inhibitory activity reported to date. The discovery process of BCL6-i illustrates the utility of irreversible inhibition for identifying potent chemical probes for intractable target proteins.

Collaboration


Dive into the Kotaro Sakamoto's collaboration.

Top Co-Authors

Avatar

Yusuke Kamada

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Junichi Sakamoto

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Taiji Asami

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Akiyoshi Tani

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Inooka

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Satoshi Sogabe

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tetsuya Ohtaki

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masanori Miwa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tadashi Umemoto

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tomoya Sameshima

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge