2025/06/02 更新

所属以外の情報はresearchmapへの登録情報を転載しています。

写真a

センゴク トオル
仙石 徹
Toru Sengoku
所属
医学研究科 医科学専攻 生化学 准教授
医学部 医学科
職名
准教授
外部リンク

学位

  • 理学博士 ( 東京大学 )

研究キーワード

  • RNA

  • 薬剤設計

  • 転写

  • 環状ペプチド

  • 構造生物学

  • エピジェネティクス

研究分野

  • ライフサイエンス / 構造生物化学

経歴

  • 独立行政法人理化学研究所   研究員

    2011年

      詳細を見る

論文

  • Structure of the human Bre1 complex bound to the nucleosome 査読

    Shuhei Onishi, Kotone Uchiyama, Ko Sato, Chikako Okada, Shunsuke Kobayashi, Keisuke Hamada, Tomohiro Nishizawa, Osamu Nureki, Kazuhiro Ogata, Toru Sengoku

    Nature Communications   15 ( 1 )   2024年3月

     詳細を見る

    担当区分:最終著者, 責任著者   掲載種別:研究論文(学術雑誌)   出版者・発行元:Springer Science and Business Media LLC  

    Abstract

    Histone H2B monoubiquitination (at Lys120 in humans) regulates transcription elongation and DNA repair. In humans, H2B monoubiquitination is catalyzed by the heterodimeric Bre1 complex composed of Bre1A/RNF20 and Bre1B/RNF40. The Bre1 proteins generally function as tumor suppressors, while in certain cancers, they facilitate cancer cell proliferation. To obtain structural insights of H2BK120 ubiquitination and its regulation, we report the cryo-electron microscopy structure of the human Bre1 complex bound to the nucleosome. The two RING domains of Bre1A and Bre1B recognize the acidic patch and the nucleosomal DNA phosphates around SHL 6.0–6.5, which are ideally located to recruit the E2 enzyme and ubiquitin for H2BK120-specific ubiquitination. Mutational experiments suggest that the two RING domains bind in two orientations and that ubiquitination occurs when Bre1A binds to the acidic patch. Our results provide insights into the H2BK120-specific ubiquitination by the Bre1 proteins and suggest that H2B monoubiquitination can be regulated by nuclesomal DNA flexibility.

    DOI: 10.1038/s41467-024-46910-8

    PubMed

    researchmap

    その他リンク: https://www.nature.com/articles/s41467-024-46910-8

  • Structural basis of transcription regulation by CNC family transcription factor, Nrf2 査読

    Toru Sengoku, Masaaki Shiina, Kae Suzuki, Keisuke Hamada, Ko Sato, Akiko Uchiyama, Shunsuke Kobayashi, Asako Oguni, Hayato Itaya, Kota Kasahara, Hirotomo Moriwaki, Chiduru Watanabe, Teruki Honma, Chikako Okada, Shiho Baba, Tsutomu Ohta, Hozumi Motohashi, Masayuki Yamamoto, Kazuhiro Ogata

    Nucleic Acids Research   50 ( 21 )   12543 - 12557   2022年12月

     詳細を見る

    担当区分:筆頭著者, 責任著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Oxford University Press (OUP)  

    Abstract

    Several basic leucine zipper (bZIP) transcription factors have accessory motifs in their DNA-binding domains, such as the CNC motif of CNC family or the EHR motif of small Maf (sMaf) proteins. CNC family proteins heterodimerize with sMaf proteins to recognize CNC–sMaf binding DNA elements (CsMBEs) in competition with sMaf homodimers, but the functional role of the CNC motif remains elusive. In this study, we report the crystal structures of Nrf2/NFE2L2, a CNC family protein regulating anti-stress transcriptional responses, in a complex with MafG and CsMBE. The CNC motif restricts the conformations of crucial Arg residues in the basic region, which form extensive contact with the DNA backbone phosphates. Accordingly, the Nrf2–MafG heterodimer has approximately a 200-fold stronger affinity for CsMBE than canonical bZIP proteins, such as AP-1 proteins. The high DNA affinity of the CNC–sMaf heterodimer may allow it to compete with the sMaf homodimer on target genes without being perturbed by other low-affinity bZIP proteins with similar sequence specificity.

    DOI: 10.1093/nar/gkac1102

    researchmap

  • De Novo Discovery of Thiopeptide Pseudo-natural Products Acting as Potent and Selective TNIK Kinase Inhibitors 査読

    Alexander A. Vinogradov, Yue Zhang, Keisuke Hamada, Jun Shi Chang, Chikako Okada, Hirotaka Nishimura, Naohiro Terasaka, Yuki Goto, Kazuhiro Ogata, Toru Sengoku, Hiroyasu Onaka, Hiroaki Suga

    Journal of the American Chemical Society   144 ( 44 )   20332 - 20341   2022年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:American Chemical Society (ACS)  

    DOI: 10.1021/jacs.2c07937

    researchmap

  • LimF is a versatile prenyltransferase for histidine-C-geranylation on diverse non-natural substrates 査読

    Yuchen Zhang, Keisuke Hamada, Dinh Thanh Nguyen, Sumika Inoue, Masayuki Satake, Shunsuke Kobayashi, Chikako Okada, Kazuhiro Ogata, Masahiro Okada, Toru Sengoku, Yuki Goto, Hiroaki Suga

    Nature Catalysis   5 ( 8 )   682 - 693   2022年8月

     詳細を見る

    担当区分:責任著者   掲載種別:研究論文(学術雑誌)   出版者・発行元:Springer Science and Business Media LLC  

    DOI: 10.1038/s41929-022-00822-2

    researchmap

    その他リンク: https://www.nature.com/articles/s41929-022-00822-2

  • Structural basis of the regulation of the normal and oncogenic methylation of nucleosomal histone H3 Lys36 by NSD2 査読 国際誌

    Ko Sato, Amarjeet Kumar, Keisuke Hamada, Chikako Okada, Asako Oguni, Ayumi Machiyama, Shun Sakuraba, Tomohiro Nishizawa, Osamu Nureki, Hidetoshi Kono, Kazuhiro Ogata, Toru Sengoku

    Nature Communications   12 ( 1 )   6605 - 6605   2021年12月

     詳細を見る

    担当区分:筆頭著者, 最終著者, 責任著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Springer Science and Business Media {LLC}  

    <jats:title>Abstract</jats:title><jats:p>Dimethylated histone H3 Lys36 (H3K36me2) regulates gene expression, and aberrant H3K36me2 upregulation, resulting from either the overexpression or point mutation of the dimethyltransferase NSD2, is found in various cancers. Here we report the cryo-electron microscopy structure of NSD2 bound to the nucleosome. Nucleosomal DNA is partially unwrapped, facilitating NSD2 access to H3K36. NSD2 interacts with DNA and H2A along with H3. The NSD2 autoinhibitory loop changes its conformation upon nucleosome binding to accommodate H3 in its substrate-binding cleft. Kinetic analysis revealed that two oncogenic mutations, E1099K and T1150A, increase NSD2 catalytic turnover. Molecular dynamics simulations suggested that in both mutants, the autoinhibitory loop adopts an open state that can accommodate H3 more often than the wild-type. We propose that E1099K and T1150A destabilize the interactions that keep the autoinhibitory loop closed, thereby enhancing catalytic turnover. Our analyses guide the development of specific inhibitors of NSD2.</jats:p>

    DOI: 10.1038/s41467-021-26913-5

    PubMed

    researchmap

  • Ribosomal synthesis and de novo discovery of bioactive foldamer peptides containing cyclic β-amino acids 査読

    Takayuki Katoh, Toru Sengoku, Kunio Hirata, Kazuhiro Ogata, Hiroaki Suga

    Nature Chemistry   12 ( 11 )   1081 - 1088   2020年8月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Springer Science and Business Media LLC  

    DOI: 10.1038/s41557-020-0525-1

    researchmap

    その他リンク: https://www.nature.com/articles/s41557-020-0525-1

  • Structural basis of protein arginine rhamnosylation by glycosyltransferase EarP article 査読

    Toru Sengoku, Takehiro Suzuki, Naoshi Dohmae, Chiduru Watanabe, Teruki Honma, Yasushi Hikida, Yoshiki Yamaguchi, Hideyuki Takahashi, Shigeyuki Yokoyama, Tatsuo Yanagisawa

    Nature Chemical Biology   14 ( 4 )   368 - 374   2018年4月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Nature Publishing Group  

    Protein glycosylation regulates many cellular processes. Numerous glycosyltransferases with broad substrate specificities have been structurally characterized. A novel inverting glycosyltransferase, EarP, specifically transfers rhamnose from dTDP-β-l-rhamnose to Arg32 of bacterial translation elongation factor P (EF-P) to activate its function. Here we report a crystallographic study of Neisseria meningitidis EarP. The EarP structure contains two tandem Rossmann-fold domains, which classifies EarP in glycosyltransferase superfamily B. In contrast to other structurally characterized protein glycosyltransferases, EarP binds the entire β-sheet structure of EF-P domain I through numerous interactions that specifically recognize its conserved residues. Thus Arg32 is properly located at the active site, and causes structural change in a conserved dTDP-β-l-rhamnose-binding loop of EarP. Rhamnosylation by EarP should occur via an SN2 reaction, with Asp20 as the general base. The Arg32 binding and accompanying structural change of EarP may induce a change in the rhamnose-ring conformation suitable for the reaction.

    DOI: 10.1038/s41589-018-0002-y

    Scopus

    PubMed

    researchmap

    その他リンク: http://orcid.org/0000-0001-9461-8714

  • Structural basis for histone H3 Lys 27 demethylation by UTX/KDM6A 査読

    Toru Sengoku, Shigeyuki Yokoyama

    GENES & DEVELOPMENT   25 ( 21 )   2266 - 2277   2011年11月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:COLD SPRING HARBOR LAB PRESS, PUBLICATIONS DEPT  

    Tri- and dimethylations of histone H3K9 (H3K9me3/2) and H3K27 (H3K27me3/2), both situated in the "A-R-Kme-S&apos;&apos; sequence motif, mediate transcriptional repression of distinct genomic regions. H3K9me3/2 mainly governs constitutive heterochromatin formation, while H3K27me3/2 represses key developmental genes. The mechanisms by which histone-modifying enzymes selectively regulate the methylation states of H3K9 and H3K27 are poorly understood. Here we report the crystal structures of the catalytic fragment of UTX/KDM6A, an H3K27me3/2-specific demethylase, in the free and H3 peptide-bound forms. The catalytic jumonji domain binds H3 residues 25-33, recognizing H3R26, H3A29, and H3P30 in a sequence-specific manner, in addition to H3K27me3 in the catalytic pocket. A novel zinc-binding domain, conserved within the KDM6 family, binds residues 17-21 of H3. The zinc-binding domain changes its conformation upon H3 binding, and thereby recognizes the H3L20 side chain via a hydrophobic patch on its surface, which is inaccessible in the H3-free form. Mutational analyses showed that H3R17, H3L20, H3R26, H3A29, H3P30, and H3T32 are each important for demethylation. No other methyllysines in the histone tails have the same set of residues at the corresponding positions. Thus, we clarified how UTX discriminates H3K27me3/2 from the other methyllysines with distinct roles, including the near-cognate H3K9me3/2, in histones.

    DOI: 10.1101/gad.172296.111

    Web of Science

    PubMed

    J-GLOBAL

    researchmap

  • Crystal structure of the bifunctional tRNA modification enzyme MnmC from Escherichia coli 査読

    Aya Kitamura, Toru Sengoku, Madoka Nishimoto, Shigeyuki Yokoyama, Yoshitaka Bessho

    PROTEIN SCIENCE   20 ( 7 )   1105 - 1113   2011年7月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:WILEY-BLACKWELL  

    Post-transcriptional modifications of bases within the transfer RNAs (tRNA) anticodon significantly affect the decoding system. In bacteria and eukaryotes, uridines at the wobble position (U34) of some tRNAs are modified to 5-methyluridine derivatives (xm(5)U). These xm(5)U34-containing tRNAs read codons ending with A or G, whereas tRNAs with the unmodified U34 are able to read all four synonymous codons of a family box. In Escherichia coli (E. coli), the bifunctional enzyme MnmC catalyzes the two consecutive reactions that convert 5-carboxymethylaminomethyl uridine (cmnm(5)U) to 5-methylaminomethyl uridine (mnm(5)U). The C-terminal domain of MnmC (MnmC1) is responsible for the flavin adenine dinucleotide (FAD)-dependent deacetylation of cmnm(5)U to 5-aminomethyl uridine (nm(5)U), whereas the N-terminal domain (MnmC2) catalyzes the subsequent S-adenosyl-L-methionine-dependent methylation of nm(5)U, leading to the final product, mnm(5)U34. Here, we determined the crystal structure of E. coli MnmC containing FAD, at 3.0 angstrom resolution. The structure of the MnmC1 domain can be classified in the FAD-dependent glutathione reductase 2 structural family, including the glycine oxidase ThiO, whereas the MnmC2 domain adopts the canonical class I methyltransferase fold. A structural comparison with ThiO revealed the residues that may be involved in cmnm(5)U recognition, supporting previous mutational analyses. The catalytic sites of the two reactions are both surrounded by conserved basic residues for possible anticodon binding, and are located far away from each other, on opposite sides of the protein. These results suggest that, although the MnmC1 and MnmC2 domains are physically linked, they could catalyze the two consecutive reactions in a rather independent manner.

    DOI: 10.1002/pro.659

    Web of Science

    PubMed

    J-GLOBAL

    researchmap

  • Crystal structure of histone demethylase LSD1 and tranylcypromine at 2.25 angstrom 査読

    Shinya Mimasu, Toru Sengoku, Seketsu Fukuzawa, Takashi Umehara, Shigeyuki Yokoyama

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   366 ( 1 )   15 - 22   2008年2月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Transcriptional activity and chromatin structure accessibility are correlated with the methylation of specific histone residues. Lysine-specific demethylase 1 (LSD1) is the first discovered histone demethylase, which demethylates Lys4 or Lys9 of histone H3, using FAD. Among the known monoamine oxidase inhibitors, tranylcypromine (Parnate) showed the most potent inhibitory effect on LSD1. Recently, the crystal structure of LSD1 and tranylcypromine was solved at 2.75 angstrom, revealing a five-membered ring fused to the flavin of LSD1. In this study, we refined the crystal structure of the LSD1-tranylcypromine complex to 2.25 angstrom. The five-membered ring model did not fit completely with the electron density, giving R-work/R-free values of 0.226/0.254. On the other hand, the N(5) adduct gave the lowest R-work/R-free values of 0.218/0.248, among the tested models. These results imply that the LSD1-tranylcypromine complex is not completely composed of the five-membered adduct, but partially contains an intermediate, such as the N(5) adduct. (C) 2007 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2007.11.066

    Web of Science

    PubMed

    J-GLOBAL

    researchmap

  • Structural Basis for RNA Unwinding by the DEAD-Box Protein Drosophila Vasa 査読

    Toru Sengoku, Osamu Nureki, Akira Nakamura, Satoru Kobayashi, Shigeyuki Yokoyama

    Cell   125 ( 2 )   287 - 300   2006年4月

     詳細を見る

    担当区分:筆頭著者   掲載種別:研究論文(学術雑誌)   出版者・発行元:Elsevier BV  

    DOI: 10.1016/j.cell.2006.01.054

    Web of Science

    PubMed

    J-GLOBAL

    researchmap

  • Crystallization and preliminary X-ray analysis of the helicase domains of Vasa complexed with RNA and an ATP analogue 査読

    T Sengoku, O Nureki, N Dohmae, A Nakamura, S Yokoyama

    ACTA CRYSTALLOGRAPHICA SECTION D-BIOLOGICAL CRYSTALLOGRAPHY   60   320 - 322   2004年2月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:BLACKWELL MUNKSGAARD  

    The helicase fragment of Vasa was purified and its RNA-binding activity was examined by a UV cross-linking assay. The fragment was crystallized in complex with poly(U) RNA (U-10) and a nonhydrolyzable analogue of ATP The crystal belonged to space group P2(1), with unit-cell parameters a = 71.06, b = 142.35, c = 130.47 Angstrom, beta = 90.86degrees. The cryocooled crystal diffracted to about 2.2 Angstrom using synchrotron radiation from station BL41XU at SPring-8.

    DOI: 10.1107/S0907444903025897

    Web of Science

    PubMed

    researchmap

  • Germline-derived GNAS-Gsα variants associated with both gain-of-function and loss-of-function phenotypes

    Atilano Carcavilla, Arrate Pereda, Mami Miyado, Maki Fukami, Fumiko Kato, Toru Sengoku, Kazuhiro Ogata, María Clemente, Irene Valenzuela, Giovanna Mantovani, Marco Cappa, Paolo Cavarzere, Yerai Vado, Isabel González-Casado, Tsutomu Ogata, Guiomar Perez de Nanclares

    European Journal of Endocrinology   2025年3月

     詳細を見る

    掲載種別:研究論文(学術雑誌)  

    DOI: 10.1093/ejendo/lvaf006

    researchmap

  • Clinical and genetic spectrum of patients with IRF2BPL syndrome 査読

    Kazuhiro Iwama, Mitsuhiro Kato, Yuri Uchiyama, Masamune Sakamoto, Ryosuke Miyamoto, Yuishin Izumi, Kei Ohashi, Ayako Hattori, Noboru Yoshida, Yoshiteru Azuma, Akito Watanabe, Chizuru Ikeda, Yuko Shimizu-Motohashi, Shohei Kusabiraki, Eiji Nakagawa, Masayuki Sasaki, Kenji Sugai, Sachiko Ohori, Naomi Tsuchida, Kohei Hamanaka, Eriko Koshimizu, Atsushi Fujita, Mitsuko Nakashima, Satoko Miyatake, Toru Sengoku, Kazuhiro Ogata, Shinji Saitoh, Hirotomo Saitsu, Shuichi Ito, Takeshi Mizuguchi, Naomichi Matsumoto

    Journal of Human Genetics   70 ( 4 )   181 - 188   2025年1月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Springer Science and Business Media LLC  

    DOI: 10.1038/s10038-025-01316-2

    researchmap

    その他リンク: https://www.nature.com/articles/s10038-025-01316-2

  • De Novo Discovery of Pseudo‐Natural Prenylated Macrocyclic Peptide Ligands 査読

    Sumika Inoue, Dinh Thanh Nguyen, Keisuke Hamada, Rika Okuma, Chikako Okada, Masahiro Okada, Ikuro Abe, Toru Sengoku, Yuki Goto, Hiroaki Suga

    Angewandte Chemie International Edition   2024年6月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Wiley  

    Prenylation of peptides is widely observed in the secondary metabolites of diverse organisms, granting peptides unique chemical properties distinct from proteinogenic amino acids. Discovery of prenylated peptide agents has largely relied on isolation or genome mining of naturally occurring molecules. To devise a platform technology for de novo discovery of artificial prenylated peptides targeting a protein of choice, here we have integrated the thioether‐macrocyclic peptide (teMP) library construction/selection technology, so‐called RaPID (Random nonstandard Peptides Integrated Discovery) system, with a Trp‐C3‐prenyltransferase KgpF involved in the biosynthesis of a prenylated natural product. This unique enzyme exhibited remarkably broad substrate tolerance, capable of modifying various Trp‐containing teMPs to install a prenylated residue with tricyclic constrained structure. We constructed a vast library of prenylated teMPs and subjected it to in vitro selection against a phosphoglycerate mutase. This selection platform has led to the identification of a pseudo‐natural prenylated teMP inhibiting the target enzyme with an IC50 of 30 nM. Importantly, the prenylation was essential for the inhibitory activity, enhanced serum stability, and cellular uptake of the peptide, highlighting the benefits of peptide prenylation. This work showcases the de novo discovery platform for pseudo‐natural prenylated peptides, which is readily applicable to other drug targets.

    DOI: 10.1002/anie.202409973

    researchmap

  • Genetic and functional diversity of β-N-acetylgalactosamine-targeting glycosidases expanded by deep-sea metagenome analysis 査読

    Tomomi Sumida, Satoshi Hiraoka, Keiko Usui, Akihiro Ishiwata, Toru Sengoku, Keith A. Stubbs, Katsunori Tanaka, Shigeru Deguchi, Shinya Fushinobu, Takuro Nunoura

    Nature Communications   15 ( 1 )   2024年5月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Springer Science and Business Media LLC  

    Abstract

    β-N-Acetylgalactosamine-containing glycans play essential roles in several biological processes, including cell adhesion, signal transduction, and immune responses. β-N-Acetylgalactosaminidases hydrolyze β-N-acetylgalactosamine linkages of various glycoconjugates. However, their biological significance remains ambiguous, primarily because only one type of enzyme, exo-β-N-acetylgalactosaminidases that specifically act on β-N-acetylgalactosamine residues, has been documented to date. In this study, we identify four groups distributed among all three domains of life and characterize eight β-N-acetylgalactosaminidases and β-N-acetylhexosaminidase through sequence-based screening of deep-sea metagenomes and subsequent searching of public protein databases. Despite low sequence similarity, the crystal structures of these enzymes demonstrate that all enzymes share a prototype structure and have diversified their substrate specificities (oligosaccharide-releasing, oligosaccharide/monosaccharide-releasing, and monosaccharide-releasing) through the accumulation of mutations and insertional amino acid sequences. The diverse β-N-acetylgalactosaminidases reported in this study could facilitate the comprehension of their structures and functions and present evolutionary pathways for expanding their substrate specificity.

    DOI: 10.1038/s41467-024-47653-2

    researchmap

    その他リンク: https://www.nature.com/articles/s41467-024-47653-2

  • A Compact Reprogrammed Genetic Code for De Novo Discovery of Proteolytically Stable Thiopeptides. 査読 国際誌

    Alexander A Vinogradov, Yue Zhang, Keisuke Hamada, Shunsuke Kobayashi, Kazuhiro Ogata, Toru Sengoku, Yuki Goto, Hiroaki Suga

    Journal of the American Chemical Society   146 ( 12 )   8058 - 8070   2024年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Thiopeptides make up a group of structurally complex peptidic natural products holding promise in bioengineering applications. The previously established thiopeptide/mRNA display platform enables de novo discovery of natural product-like thiopeptides with designed bioactivities. However, in contrast to natural thiopeptides, the discovered structures are composed predominantly of proteinogenic amino acids, which results in low metabolic stability in many cases. Here, we redevelop the platform and demonstrate that the utilization of compact reprogrammed genetic codes in mRNA display libraries can lead to the discovery of thiopeptides predominantly composed of nonproteinogenic structural elements. We demonstrate the feasibility of our designs by conducting affinity selections against Traf2- and NCK-interacting kinase (TNIK). The experiment identified a series of thiopeptides with high affinity to the target protein (the best KD = 2.1 nM) and kinase inhibitory activity (the best IC50 = 0.15 μM). The discovered compounds, which bore as many as 15 nonproteinogenic amino acids in an 18-residue macrocycle, demonstrated high metabolic stability in human serum with a half-life of up to 99 h. An X-ray cocrystal structure of TNIK in complex with a discovered thiopeptide revealed how nonproteinogenic building blocks facilitate the target engagement and orchestrate the folding of the thiopeptide into a noncanonical conformation. Altogether, the established platform takes a step toward the discovery of thiopeptides with high metabolic stability for early drug discovery applications.

    DOI: 10.1021/jacs.3c12037

    PubMed

    researchmap

  • Switching Prenyl Donor Specificities of Cyanobactin Prenyltransferases 査読

    Yuchen Zhang, Keisuke Hamada, Masayuki Satake, Toru Sengoku, Yuki Goto, Hiroaki Suga

    Journal of the American Chemical Society   145 ( 44 )   23893 - 23898   2023年10月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:American Chemical Society (ACS)  

    DOI: 10.1021/jacs.3c07373

    researchmap

  • Biallelic structural variations withinFGF12detected by long-read sequencing in epilepsy 査読

    Sachiko Ohori, Akihiko Miyauchi, Hitoshi Osaka, Charles Marques Lourenco, Naohiro Arakaki, Toru Sengoku, Kazuhiro Ogata, Rachel Sayuri Honjo, Chong Ae Kim, Satomi Mitsuhashi, Martin C Frith, Rie Seyama, Naomi Tsuchida, Yuri Uchiyama, Eriko Koshimizu, Kohei Hamanaka, Kazuharu Misawa, Satoko Miyatake, Takeshi Mizuguchi, Kuniaki Saito, Atsushi Fujita, Naomichi Matsumoto

    Life Science Alliance   6 ( 8 )   e202302025 - e202302025   2023年8月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Life Science Alliance, LLC  

    We discovered biallelic intragenic structural variations (SVs) inFGF12by applying long-read whole genome sequencing to an exome-negative patient with developmental and epileptic encephalopathy (DEE). We also found another DEE patient carrying a biallelic (homozygous) single-nucleotide variant (SNV) inFGF12that was detected by exome sequencing.FGF12heterozygous recurrent missense variants with gain-of-function or heterozygous entire duplication ofFGF12are known causes of epilepsy, but biallelic SNVs/SVs have never been described.FGF12encodes intracellular proteins interacting with the C-terminal domain of the alpha subunit of voltage-gated sodium channels 1.2, 1.5, and 1.6, promoting excitability by delaying fast inactivation of the channels. To validate the molecular pathomechanisms of these biallelicFGF12SVs/SNV, highly sensitive gene expression analyses using lymphoblastoid cells from the patient with biallelic SVs, structural considerations, andDrosophilain vivo functional analysis of the SNV were performed, confirming loss-of-function. Our study highlights the importance of small SVs in Mendelian disorders, which may be overlooked by exome sequencing but can be detected efficiently by long-read whole genome sequencing, providing new insights into the pathomechanisms of human diseases.

    DOI: 10.26508/lsa.202302025

    researchmap

  • Brain monoamine vesicular transport disease caused by homozygous SLC18A2 variants: A study in 42 affected individuals 査読 国際誌

    Ken Saida, Reza Maroofian, Toru Sengoku, Tadahiro Mitani, Alistair T. Pagnamenta, Dana Marafi, Maha S. Zaki, Thomas J. O’Brien, Ehsan Ghayoor Karimiani, Rauan Kaiyrzhanov, Marina Takizawa, Sachiko Ohori, Huey Yin Leong, Gulsen Akay, Hamid Galehdari, Mina Zamani, Ratna Romy, Christopher J. Carroll, Mehran Beiraghi Toosi, Farah Ashrafzadeh, Shima Imannezhad, Hadis Malek, Najmeh Ahangari, Hoda Tomoum, Vykuntaraju K. Gowda, Varunvenkat M. Srinivasan, David Murphy, Natalia Dominik, Hasnaa M. Elbendary, Karima Rafat, Sanem Yilmaz, Seda Kanmaz, Mine Serin, Deepa Krishnakumar, Alice Gardham, Anna Maw, Tekki Sreenivasa Rao, Sarah Alsubhi, Myriam Srour, Daniela Buhas, Tamison Jewett, Rachel E. Goldberg, Hanan Shamseldin, Eirik Frengen, Doriana Misceo, Petter Strømme, José Ricardo Magliocco Ceroni, Chong Ae Kim, Gozde Yesil, Esma Sengenc, Serhat Guler, Mariam Hull, Mered Parnes, Dilek Aktas, Banu Anlar, Yavuz Bayram, Davut Pehlivan, Jennifer E. Posey, Shahryar Alavi, Seyed Ali Madani Manshadi, Hamad Alzaidan, Mohammad Al-Owain, Lama Alabdi, Ferdous Abdulwahab, Futoshi Sekiguchi, Kohei Hamanaka, Atsushi Fujita, Yuri Uchiyama, Takeshi Mizuguchi, Satoko Miyatake, Noriko Miyake, Reem M. Elshafie, Kamran Salayev, Ulviyya Guliyeva, Fowzan S. Alkuraya, Joseph G. Gleeson, Kristin G. Monaghan, Katherine G. Langley, Hui Yang, Mahsa Motavaf, Saeid Safari, Mozhgan Alipour, Kazuhiro Ogata, André E.X. Brown, James R. Lupski, Henry Houlden, Naomichi Matsumoto

    Genetics in Medicine   25 ( 1 )   90 - 102   2022年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Elsevier BV  

    PURPOSE: Brain monoamine vesicular transport disease is an infantile-onset movement disorder that mimics cerebral palsy. In 2013, the homozygous SLC18A2 variant, p.Pro387Leu, was first reported as a cause of this rare disorder, and dopamine agonists were efficient for treating affected individuals from a single large family. To date, only 6 variants have been reported. In this study, we evaluated genotype-phenotype correlations in individuals with biallelic SLC18A2 variants. METHODS: A total of 42 affected individuals with homozygous SLC18A2 variant alleles were identified. We evaluated genotype-phenotype correlations and the missense variants in the affected individuals based on the structural modeling of rat VMAT2 encoded by Slc18a2, with cytoplasm- and lumen-facing conformations. A Caenorhabditis elegans model was created for functional studies. RESULTS: A total of 19 homozygous SLC18A2 variants, including 3 recurrent variants, were identified using exome sequencing. The affected individuals typically showed global developmental delay, hypotonia, dystonia, oculogyric crisis, and autonomic nervous system involvement (temperature dysregulation/sweating, hypersalivation, and gastrointestinal dysmotility). Among the 58 affected individuals described to date, 16 (28%) died before the age of 13 years. Of the 17 patients with p.Pro237His, 9 died, whereas all 14 patients with p.Pro387Leu survived. Although a dopamine agonist mildly improved the disease symptoms in 18 of 21 patients (86%), some affected individuals with p.Ile43Phe and p.Pro387Leu showed milder phenotypes and presented prolonged survival even without treatment. The C. elegans model showed behavioral abnormalities. CONCLUSION: These data expand the phenotypic and genotypic spectra of SLC18A2-related disorders.

    DOI: 10.1016/j.gim.2022.09.010

    PubMed

    researchmap

  • Systematic analysis of exonic germline and postzygotic de novo mutations in bipolar disorder 査読 国際誌

    Masaki Nishioka, An-a Kazuno, Takumi Nakamura, Naomi Sakai, Takashi Hayama, Kumiko Fujii, Koji Matsuo, Atsuko Komori, Mizuho Ishiwata, Yoshinori Watanabe, Takashi Oka, Nana Matoba, Muneko Kataoka, Ahmed N. Alkanaq, Kohei Hamanaka, Takashi Tsuboi, Toru Sengoku, Kazuhiro Ogata, Nakao Iwata, Masashi Ikeda, Naomichi Matsumoto, Tadafumi Kato, Atsushi Takata

    Nature Communications   12 ( 1 )   3750 - 3750   2021年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Springer Science and Business Media LLC  

    <title>Abstract</title>Bipolar disorder is a severe mental illness characterized by recurrent manic and depressive episodes. To better understand its genetic architecture, we analyze ultra-rare de novo mutations in 354 trios with bipolar disorder. For germline de novo mutations, we find significant enrichment of loss-of-function mutations in constrained genes (corrected-<italic>P</italic> = 0.0410) and deleterious mutations in presynaptic active zone genes (FDR = 0.0415). An analysis integrating single-cell RNA-sequencing data identifies a subset of excitatory neurons preferentially expressing the genes hit by deleterious mutations, which are also characterized by high expression of developmental disorder genes. In the analysis of postzygotic mutations, we observe significant enrichment of deleterious ones in developmental disorder genes (<italic>P</italic> = 0.00135), including the <italic>SRCAP</italic> gene mutated in two unrelated probands. These data collectively indicate the contributions of both germline and postzygotic mutations to the risk of bipolar disorder, supporting the hypothesis that postzygotic mutations of developmental disorder genes may contribute to bipolar disorder.

    DOI: 10.1038/s41467-021-23453-w

    PubMed

    researchmap

    その他リンク: http://www.nature.com/articles/s41467-021-23453-w

  • An Ultrapotent and Selective Cyclic Peptide Inhibitor of Human β-Factor XIIa in a Cyclotide Scaffold 査読

    Wenyu Liu, Simon J. de Veer, Yen-Hua Huang, Toru Sengoku, Chikako Okada, Kazuhiro Ogata, Christina N. Zdenek, Bryan G. Fry, Joakim E. Swedberg, Toby Passioura, David J. Craik, Hiroaki Suga

    Journal of the American Chemical Society   143 ( 44 )   18481 - 18489   2021年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:American Chemical Society ({ACS})  

    DOI: 10.1021/jacs.1c07574

    researchmap

  • Pathogenic variants in the SMN complex gene GEMIN5 cause cerebellar atrophy. 査読 国際誌

    Ken Saida, Junya Tamaoki, Masayuki Sasaki, Muzhirah Haniffa, Eriko Koshimizu, Toru Sengoku, Hiroki Maeda, Masahiro Kikuchi, Haruna Yokoyama, Masamune Sakamoto, Kazuhiro Iwama, Futoshi Sekiguchi, Kohei Hamanaka, Atsushi Fujita, Takeshi Mizuguchi, Kazuhiro Ogata, Noriko Miyake, Satoko Miyatake, Makoto Kobayashi, Naomichi Matsumoto

    Clinical genetics   100 ( 6 )   722 - 730   2021年9月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Cerebellar ataxia is a genetically heterogeneous disorder. GEMIN5, encoding an RNA-binding protein of the survival of motor neuron complex, is essential for small nuclear ribonucleoprotein biogenesis, and it was recently reported that biallelic loss-of-function variants cause neurodevelopmental delay, hypotonia and cerebellar ataxia. Here, whole-exome analysis revealed compound heterozygous GEMIN5 variants in two individuals from our cohort of 162 patients with cerebellar atrophy. Three novel truncating variants and one previously reported missense variant were identified: c.2196dupA, p.(Arg733Thrfs*6) and c.1831G>A, p.(Val611Met) in individual 1, and c.3913delG, p.(Ala1305Leufs*14) and c.4496dupA, p.(Tyr1499*) in individual 2. Western blotting analysis using lymphoblastoid cell lines derived from both individuals showed significantly reduced levels of GEMIN5 protein. Zebrafish model for p.(Arg733Thrfs*6) and p.(Ala1305Leufs*14) exhibited complete lethality at 2 weeks and recapitulated a distinct dysplastic phenotype. The phenotypes of affected individuals and the zebrafish mutant model strongly suggest that biallelic loss-of-function variants in GEMIN5 cause cerebellar atrophy.

    DOI: 10.1111/cge.14066

    PubMed

    researchmap

  • De novo ARF3 variants cause neurodevelopmental disorder with brain abnormality. 査読 国際誌

    Masamune Sakamoto, Kazunori Sasaki, Atsushi Sugie, Yohei Nitta, Tetsuaki Kimura, Semra Gürsoy, Tayfun Cinleti, Mizue Iai, Toru Sengoku, Kazuhiro Ogata, Atsushi Suzuki, Nobuhiko Okamoto, Kazuhiro Iwama, Naomi Tsuchida, Yuri Uchiyama, Eriko Koshimizu, Atsushi Fujita, Kohei Hamanaka, Satoko Miyatake, Takeshi Mizuguchi, Masataka Taguri, Shuuichi Ito, Hidehisa Takahashi, Noriko Miyake, Naomichi Matsumoto

    Human molecular genetics   31 ( 1 )   69 - 81   2021年8月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    An optimal Golgi transport system is important for mammalian cells. The adenosine diphosphate (ADP) ribosylation factors (ARF) are key proteins for regulating cargo sorting at the Golgi network. In this family, ARF3 mainly works at the trans-Golgi network (TGN), and no ARF3-related phenotypes have yet been described in humans. We here report the clinical and genetic evaluations of two unrelated children with de novo pathogenic variants in the ARF3 gene: c.200A > T (p.Asp67Val) and c.296G > T (p.Arg99Leu). Although the affected individuals presented commonly with developmental delay, epilepsy, and brain abnormalities, there were differences in severity, clinical course, and brain lesions. In vitro subcellular localization assays revealed that the p.Arg99Leu mutant localized to Golgi apparatus, similar to the wild-type, whereas the p.Asp67Val mutant tended to show a disperse cytosolic pattern together with abnormally dispersed Golgi localization, similar to that observed in a known dominant negative variant (p.Thr31Asn). Pull-down assays revealed that the p.Asp67Val had a loss-of-function effect and the p.Arg99Leu variant had increased binding of the adaptor protein, Golgi-localized, γ-adaptin ear-containing, ARF-binding protein 1 (GGA1), supporting the gain of function. Furthermore, in vivo studies revealed that p.Asp67Val transfection led to lethality in flies. In contrast, flies expressing p.Arg99Leu had abnormal rough eye, as observed in the gain-of-function variant p.Gln71Leu. These data indicate that two ARF3 variants, the possibly loss-of-function p.Asp67Val and the gain-of-function p.Arg99Leu, both impair the Golgi transport system. Therefore, it may not be unreasonable that they showed different clinical features like diffuse brain atrophy (p.Asp67Val) and cerebellar hypoplasia (p.Arg99Leu).

    DOI: 10.1093/hmg/ddab224

    PubMed

    researchmap

  • OTUD5 Variants Associated With X-Linked Intellectual Disability and Congenital Malformation 査読 国際誌

    Ken Saida, Tokiko Fukuda, Daryl A. Scott, Toru Sengoku, Kazuhiro Ogata, Annarita Nicosia, Andres Hernandez-Garcia, Seema R. Lalani, Mahshid S. Azamian, Haley Streff, Pengfei Liu, Hongzheng Dai, Takeshi Mizuguchi, Satoko Miyatake, Miki Asahina, Tsutomu Ogata, Noriko Miyake, Naomichi Matsumoto

    Frontiers in Cell and Developmental Biology   9   631428 - 631428   2021年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Frontiers Media SA  

    <sec><title>Background</title>X-linked intellectual disability (XLID), which occurs predominantly in males, is a relatively common and genetically heterogeneous disorder in which over 100 mutated genes have been reported. The <italic>OTUD5</italic> gene at Xp11.23 encodes ovarian tumor deubiquitinase 5 protein, which is a deubiquitinating enzyme member of the ovarian tumor family. LINKage-specific-deubiquitylation-deficiency-induced embryonic defects (LINKED) syndrome, arising from pathogenic <italic>OTUD5</italic> variants, was recently reported as a new XLID with additional congenital anomalies.

    </sec><sec><title>Methods</title>We investigated three affected males (49- and 47-year-old brothers [Individuals 1 and 2] and a 2-year-old boy [Individual 3]) from two families who showed developmental delay. Their common clinical features included developmental delay, hypotonia, short stature, and distinctive facial features, such as telecanthus and a depressed nasal bridge. Individuals 1 and 2 showed epilepsy and brain magnetic resonance imaging showed a thin corpus callosum and mild ventriculomegaly. Individual 3 showed congenital malformations, including tetralogy of Fallot, hypospadias, and bilateral cryptorchidism. To identify the genetic cause of these features, we performed whole-exome sequencing.

    </sec><sec><title>Results</title>A hemizygous <italic>OTUD5</italic> missense variant, c.878A&amp;gt;T, p.Asn293Ile [NM_017602.4], was identified in one family with Individuals 1 and 2, and another missense variant, c.1210 C&amp;gt;T, p.Arg404Trp, in the other family with Individual 3, respectively. The former variant has not been registered in public databases and was predicted to be pathogenic by multiple <italic>in silico</italic> prediction tools. The latter variant p.Arg404Trp was previously reported as a pathogenic <italic>OTUD5</italic> variant, and Individual 3 showed a typical LINKED syndrome phenotype. However, Individuals 1 and 2, with the novel variant (p.Asn293Ile), showed no cardiac or genitourinary malformations.

    </sec><sec><title>Conclusions</title>Unlike previous reports of LINKED syndrome, which described early lethality with congenital cardiac anomalies, our three cases are still alive. Notably, the adult brothers with the novel missense <italic>OTUD5</italic> variant have lived into their forties. This may be indicative of a milder phenotype as a possible genotype-phenotype correlation. These findings imply a possible long-term prognosis for individuals with this new XLID syndrome, and a wider phenotypic variation than initially thought.

    </sec>

    DOI: 10.3389/fcell.2021.631428

    PubMed

    researchmap

  • Development and Structural Evaluation of N-Alkylated trans-2-Phenylcyclopropylamine-Based LSD1 Inhibitors. 査読 国際誌

    Hideaki Niwa, Shin Sato, Noriko Handa, Toru Sengoku, Takashi Umehara, Shigeyuki Yokoyama

    ChemMedChem   15 ( 9 )   787 - 793   2020年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Lysine-specific demethylase 1 (LSD1) is a flavin adenine dinucleotide (FAD)-dependent enzyme that catalyzes the demethylation of histone H3 and regulates gene expression. Because it is implicated in the regulation of diseases such as acute myeloid leukemia, potent LSD1-specific inhibitors have been pursued. Trans-2-phenylcyclopropylamine (2-PCPA)-based inhibitors featuring substitutions on the amino group have emerged, with sub-micromolar affinities toward LSD1 and high selectivities over monoamine oxidases (MAOs). We synthesized two N-alkylated 2-PCPA-based LSD1 inhibitors, S2116 and S2157, based on the previously developed S2101. S2116 and S2157 exhibited enhanced potency for LSD1 by 2.0- to 2.6-fold, as compared with S2101. In addition, they exhibited improved selectivity over MAOs. Structural analyses of LSD1 co-crystallized with S2101, S2116, S2157, or another N-alkylated inhibitor (FCPA-MPE) confirmed that the N-substituents enhance the potency of a 2-PCPA-based inhibitor of LSD1, without constituting the adduct formed with FAD.

    DOI: 10.1002/cmdc.202000014

    PubMed

    researchmap

  • A novel PAK1 variant causative of neurodevelopmental disorder with postnatal macrocephaly. 査読 国際誌

    Sachiko Ohori, Satomi Mitsuhashi, Revital Ben-Haim, Eli Heyman, Toru Sengoku, Kazuhiro Ogata, Naomichi Matsumoto

    Journal of human genetics   65 ( 5 )   481 - 485   2020年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    p21-activated kinases (PAKs) are protein serine/threonine kinases stimulated by Rho-family p21 GTPases such as CDC42 and RAC. PAKs have been implicated in several human disorders, with pathogenic variants in PAK3 associated with intellectual disability and several PAK members, especially PAK1 and PAK4, overexpressed in human cancer. Recently, de novo PAK1 variants were reported to be causative of neurodevelopmental disorder (ND) with secondary macrocephaly in three patients. We herein report a fourth patient with ND, epilepsy, and macrocephaly caused by a de novo PAK1 missense variant. Two previously reported missense PAK1 variants functioned as activating alleles by reducing PAK1 homodimerization. To examine the pathogenicity of the identified novel p.Ser110Thr variant, we carried out in silico structural analysis. Our findings suggest that this variant also prevents PAK1 homodimerization, leading to constitutive PAK1 activation.

    DOI: 10.1038/s10038-020-0728-8

    PubMed

    researchmap

  • The recurrent postzygotic pathogenic variant p.Glu47Lys in RHOA causes a novel recognizable neuroectodermal phenotype. 査読 国際誌

    Gökhan Yigit, Ken Saida, Danielle DeMarzo, Noriko Miyake, Atsushi Fujita, Tiong Yang Tan, Susan M White, Alexandrea Wadley, Mohammad R Toliat, Susanne Motameny, Marek Franitza, Chloe A Stutterd, Pin F Chong, Ryutaro Kira, Toru Sengoku, Kazuhiro Ogata, Maria J Guillen Sacoto, Christine Fresen, Bodo B Beck, Peter Nürnberg, Christoph Dieterich, Bernd Wollnik, Naomichi Matsumoto, Janine Altmüller

    Human mutation   41 ( 3 )   591 - 599   2020年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    RHOA is a member of the Rho family of GTPases that are involved in fundamental cellular processes including cell adhesion, migration, and proliferation. RHOA can stimulate the formation of stress fibers and focal adhesions and is a key regulator of actomyosin dynamics in various tissues. In a Genematcher-facilitated collaboration, we were able to identify four unrelated individuals with a specific phenotype characterized by hypopigmented areas of the skin, dental anomalies, body asymmetry, and limb length discrepancy due to hemihypotrophy of one half of the body, as well as brain magnetic resonance imaging (MRI) anomalies. Using whole-exome and ultra-deep amplicon sequencing and comparing genomic data of affected and unaffected areas of the skin, we discovered that all four individuals carried the identical RHOA missense variant, c.139G>A; p.Glu47Lys, in a postzygotic state. Molecular modeling and in silico analysis of the affected p.Glu47Lys residue in RHOA indicated that this exchange is predicted to specifically alter the interaction of RHOA with its downstream effectors containing a PKN-type binding domain and thereby disrupts its ability to activate signaling. Our findings indicate that the recurrent postzygotic RHOA missense variant p.Glu47Lys causes a specific mosaic disorder in humans.

    DOI: 10.1002/humu.23964

    PubMed

    researchmap

  • Phenotype-genotype correlations in patients with GNB1 gene variants, including the first three reported Japanese patients to exhibit spastic diplegia, dyskinetic quadriplegia, and infantile spasms. 査読 国際誌

    Wakaba Endo, Satoru Ikemoto, Noriko Togashi, Takuya Miyabayashi, Erika Nakajima, Shin-Ichiro Hamano, Moriei Shibuya, Ryo Sato, Yusuke Takezawa, Yukimune Okubo, Takehiko Inui, Mitsuhiro Kato, Toru Sengoku, Kazuhiro Ogata, Kohei Hamanaka, Takeshi Mizuguchi, Satoko Miyatake, Mitsuko Nakashima, Naomichi Matsumoto, Kazuhiro Haginoya

    Brain & development   42 ( 2 )   199 - 204   2020年2月

     詳細を見る

    記述言語:英語  

    We report the first three Japanese patients with missense variants in the GNB1 gene. Patients exhibited severe dyskinetic quadriplegia with cortical blindness and epileptic spasms, West syndrome (but with good outcomes), and hypotonic quadriplegia that later developed into spastic diplegia. Whole-exome sequencing revealed two recurrent GNB1 variants (p.Leu95Pro and p.Ile80Thr) and one novel variant (p.Ser74Leu). A recent investigation revealed large numbers of patients with GNB1 variants. Functional studies of such variants and genotype-phenotype correlation are required to enable future precision medicine.

    DOI: 10.1016/j.braindev.2019.10.006

    PubMed

    researchmap

  • Genetic landscape of Rett syndrome-like phenotypes revealed by whole exome sequencing. 査読 国際誌

    Kazuhiro Iwama, Takeshi Mizuguchi, Eri Takeshita, Eiji Nakagawa, Tetsuya Okazaki, Yoshiko Nomura, Yoshitaka Iijima, Ichiro Kajiura, Kenji Sugai, Takashi Saito, Masayuki Sasaki, Kotaro Yuge, Tomoko Saikusa, Nobuhiko Okamoto, Satoru Takahashi, Masano Amamoto, Ichiro Tomita, Satoko Kumada, Yuki Anzai, Kyoko Hoshino, Aviva Fattal-Valevski, Naohide Shiroma, Masaharu Ohfu, Masaharu Moroto, Koichi Tanda, Tomoko Nakagawa, Takafumi Sakakibara, Shin Nabatame, Muneaki Matsuo, Akiko Yamamoto, Shoko Yukishita, Ken Inoue, Chikako Waga, Yoko Nakamura, Shoko Watanabe, Chihiro Ohba, Toru Sengoku, Atsushi Fujita, Satomi Mitsuhashi, Satoko Miyatake, Atsushi Takata, Noriko Miyake, Kazuhiro Ogata, Shuichi Ito, Hirotomo Saitsu, Toyojiro Matsuishi, Yu-Ichi Goto, Naomichi Matsumoto

    Journal of medical genetics   56 ( 6 )   396 - 407   2019年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    BACKGROUND: Rett syndrome (RTT) is a characteristic neurological disease presenting with regressive loss of neurodevelopmental milestones. Typical RTT is generally caused by abnormality of methyl-CpG binding protein 2 (MECP2). Our objective to investigate the genetic landscape of MECP2-negative typical/atypical RTT and RTT-like phenotypes using whole exome sequencing (WES). METHODS: We performed WES on 77 MECP2-negative patients either with typical RTT (n=11), atypical RTT (n=22) or RTT-like phenotypes (n=44) incompatible with the RTT criteria. RESULTS: Pathogenic or likely pathogenic single-nucleotide variants in 28 known genes were found in 39 of 77 (50.6%) patients. WES-based CNV analysis revealed pathogenic deletions involving six known genes (including MECP2) in 8 of 77 (10.4%) patients. Overall, diagnostic yield was 47 of 77 (61.0 %). Furthermore, strong candidate variants were found in four novel genes: a de novo variant in each of ATPase H+ transporting V0 subunit A1 (ATP6V0A1), ubiquitin-specific peptidase 8 (USP8) and microtubule-associated serine/threonine kinase 3 (MAST3), as well as biallelic variants in nuclear receptor corepressor 2 (NCOR2). CONCLUSIONS: Our study provides a new landscape including additional genetic variants contributing to RTT-like phenotypes, highlighting the importance of comprehensive genetic analysis.

    DOI: 10.1136/jmedgenet-2018-105775

    PubMed

    researchmap

  • Thioether Macrocyclic Peptides Selected against TET1 Compact Catalytic Domain Inhibit TET1 Catalytic Activity. 査読 国際誌

    Kosuke Nishio, Roman Belle, Takayuki Katoh, Akane Kawamura, Toru Sengoku, Kazuharu Hanada, Noboru Ohsawa, Mikako Shirouzu, Shigeyuki Yokoyama, Hiroaki Suga

    Chembiochem : a European journal of chemical biology   19 ( 9 )   979 - 985   2018年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    The ten-eleven translocation (TET) protein family, consisting of three isoforms (TET1/2/3), have been found in mammalian cells and have a crucial role in 5-methylcytosine demethylation in genomic DNA through the catalysis of oxidation reactions assisted by 2-oxoglutarate (2OG). DNA methylation/demethylation contributes to the regulation of gene expression at the transcriptional level, and recent studies have revealed that TET1 is highly elevated in malignant cells of various diseases and related to malignant alteration. TET1 inhibitors based on a scaffold of thioether macrocyclic peptides, which have been discovered by the random nonstandard peptide integrated discovery (RaPID) system, are reported. The affinity-based selection was performed against the TET1 compact catalytic domain (TET1CCD) to yield thioether macrocyclic peptides. These peptides exhibited inhibitory activity of the TET1 catalytic domain (TET1CD), with an IC50 value as low as 1.1 μm. One of the peptides, TiP1, was also able to inhibit TET1CD over TET2CD with tenfold selectivity, although it was likely to target the 2OG binding site; this provides a good starting point to develop more selective inhibitors.

    DOI: 10.1002/cbic.201800047

    PubMed

    researchmap

  • A SelB/EF-Tu/aIF2γ-like protein from Methanosarcina mazei in the GTP-bound form binds cysteinyl-tRNACys 査読

    Yanagisawa Tatsuo, Ishii Ryohei, Hikida Yasushi, Fukunaga Ryuya, Sengoku Toru, Sekine Shun-ichi, Yokoyama Shigeyuki

    Journal of Structural and Functional Genomics   16 ( 1 )   25 - 41   2015年

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    DOI: 10.1007/s10969-015-9193-6

    Scopus

    PubMed

    researchmap

  • SET1 and p300 Act Synergistically, through Coupled Histone Modifications, in Transcriptional Activation by p53 査読

    Zhanyun Tang, Wei-Yi Chen, Miho Shimada, Uyen T. T. Nguyen, Jaehoon Kim, Xiao-Jian Sun, Toru Sengoku, Robert K. McGinty, Joseph P. Fernandez, Tom W. Muir, Robert G. Roeder

    CELL   154 ( 2 )   297 - 310   2013年7月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:CELL PRESS  

    The H3K4me3 mark in chromatin is closely correlated with actively transcribed genes, although the mechanisms involved in its generation and function are not fully understood. In vitro studies with recombinant chromatin and purified human factors demonstrate a robust SET1 complex (SET1C)-mediated H3K4 trimethylation that is dependent upon p53- and p300-mediated H3 acetylation, a corresponding SET1C-mediated enhancement of p53- and p300-dependent transcription that reflects a primary effect of SET1C through H3K4 trimethylation, and direct SET1C-p53 and SET1C-p300 interactions indicative of a targeted recruitment mechanism. Complementary cell-based assays demonstrate a DNA-damage-induced p53- SET1C interaction, a corresponding enrichment of SET1C and H3K4me3 on a p53 target gene (p21/WAF1), and a corresponding codependency of H3K4 trimethylation and transcription upon p300 and SET1C. These results establish a mechanism in which SET1C and p300 act cooperatively, through direct interactions and coupled histone modifications, to facilitate the function of p53.

    DOI: 10.1016/j.cell.2013.06.027

    Web of Science

    J-GLOBAL

    researchmap

  • Characterization and Structure of the Aquifex aeolicus Protein DUF752 A BACTERIAL tRNA-METHYLTRANSFERASE (MnmC2) FUNCTIONING WITHOUT THE USUALLY FUSED OXIDASE DOMAIN (MnmC1) 査読

    Aya Kitamura, Madoka Nishimoto, Toru Sengoku, Rie Shibata, Gunilla Jager, Glenn R. Bjork, Henri Grosjean, Shigeyuki Yokoyama, Yoshitaka Bessho

    JOURNAL OF BIOLOGICAL CHEMISTRY   287 ( 52 )   43950 - 43960   2012年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Post-transcriptional modifications of the wobble uridine (U34) of tRNAs play a critical role in reading NNA/G codons belonging to split codon boxes. In a subset of Escherichia coli tRNA, this wobble uridine is modified to 5-methylaminomethyluridine (mnm(5)U34) through sequential enzymatic reactions. Uridine 34 is first converted to 5-carboxymethylaminomethyluridine (cmnm(5)U34) by the MnmE-Mnm Genzyme complex. The cmnm(5)U34 is further modified to mnm(5)U by the bifunctional MnmC protein. In the first reaction, the FAD-dependent oxidase domain (MnmC1) converts cmnm(5)U into 5-aminomethyluridine (nm(5)U34), and this reaction is immediately followed by the methylation of the free amino group into mnm(5)U34 by the S-adenosylmethionine-dependent domain (MnmC2). Aquifex aeolicus lacks a bifunctional MnmC protein fusion and instead encodes the Rossmann-fold protein DUF752, which is homologous to the methyltransferase MnmC2 domain of Escherichia coli MnmC (26% identity). Here, we determined the crystal structure of the A. aeolicus DUF752 protein at 2.5 angstrom resolution, which revealed that it catalyzes the S-adenosylmethionine-dependent methylation of nm(5)U in vitro, to form mnm(5)U34 in tRNA. We also showed that naturally occurring tRNA from A. aeolicus contains the 5-mnm group attached to the C5 atom of U34. Taken together, these results support the recent proposal of an alternative MnmC1-independent shortcut pathway for producing mnm(5)U34 in tRNAs.

    DOI: 10.1074/jbc.M112.409300

    Web of Science

    PubMed

    J-GLOBAL

    researchmap

  • UTX and UTY Demonstrate Histone Demethylase-Independent Function in Mouse Embryonic Development 査読

    Karl B. Shpargel, Toru Sengoku, Shigeyuki Yokoyama, Terry Magnuson

    PLOS GENETICS   8 ( 9 )   2012年9月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:PUBLIC LIBRARY SCIENCE  

    UTX (KDM6A) and UTY are homologous X and Y chromosome members of the Histone H3 Lysine 27 (H3K27) demethylase gene family. UTX can demethylate H3K27; however, in vitro assays suggest that human UTY has lost enzymatic activity due to sequence divergence. We produced mouse mutations in both Utx and Uty. Homozygous Utx mutant female embryos are mid-gestational lethal with defects in neural tube, yolk sac, and cardiac development. We demonstrate that mouse UTY is devoid of in vivo demethylase activity, so hemizygous XUtx- Y+ mutant male embryos should phenocopy homozygous XUtx- XUtx- females. However, XUtx- Y+ mutant male embryos develop to term; although runted, approximately 25% survive postnatally reaching adulthood. Hemizygous X+ YUty- mutant males are viable. In contrast, compound hemizygous X Utx- YUty- males phenocopy homozygous XUtx- XUtx- females. Therefore, despite divergence of UTX and UTY in catalyzing H3K27 demethylation, they maintain functional redundancy during embryonic development. Our data suggest that UTX and UTY are able to regulate gene activity through demethylase independent mechanisms. We conclude that UTX H3K27 demethylation is non-essential for embryonic viability.

    DOI: 10.1371/journal.pgen.1002964

    Web of Science

    PubMed

    J-GLOBAL

    researchmap

  • Substrate tRNA Recognition Mechanism of a Multisite-specific tRNA Methyltransferase, Aquifex aeolicus Trm1, Based on the X-ray Crystal Structure 査読

    Takako Awai, Anna Ochi, Ihsanawati, Toru Sengoku, Akira Hirata, Yoshitaka Bessho, Shigeyuki Yokoyama, Hiroyuki Hori

    JOURNAL OF BIOLOGICAL CHEMISTRY   286 ( 40 )   35236 - 35246   2011年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Archaeal and eukaryotic tRNA (N-2,N-2-guanine)-dimethyltransferase (Trm1) produces N-2,N-2-dimethylguanine at position 26 in tRNA. In contrast, Trm1 from Aquifex aeolicus, a hyper-thermophilic eubacterium, modifies G27 as well as G26. Here, a gel mobility shift assay revealed that the T-arm in tRNA is the binding site of A. aeolicus Trm1. To address the multisite specificity, we performed an x-ray crystal structure study. The overall structure of A. aeolicus Trm1 is similar to that of archaeal Trm1, although there is a zinc-cysteine cluster in the C-terminal domain of A. aeolicus Trm1. The N-terminal domain is a typical catalytic domain of S-adenosyl-L-methionine-dependent methyltransferases. On the basis of the crystal structure and amino acid sequence alignment, we prepared 30 mutant Trm1 proteins. These mutant proteins clarified residues important for S-adenosyl-L-methionine binding and enabled us to propose a hypothetical reaction mechanism. Furthermore, the tRNA-binding site was also elucidated by methyl transfer assay and gel mobility shift assay. The electrostatic potential surface models of A. aeolicus and archaeal Trm1 proteins demonstrated that the distribution of positive charges differs between the two proteins. We constructed a tRNA-docking model, in which the T-arm structure was placed onto the large area of positive charge, which is the expected tRNA-binding site, of A. aeolicus Trm1. In this model, the target G26 base can be placed near the catalytic pocket; however, the nucleotide at position 27 gains closer access to the pocket. Thus, this docking model introduces a rational explanation of the multisite specificity of A. aeolicus Trm1.

    DOI: 10.1074/jbc.M111.253641

    Web of Science

    PubMed

    J-GLOBAL

    researchmap

  • Structural and mutational studies of the amino acid-editing domain from archaeal/eukaryal phenylalanyl-tRNA synthetase 査読

    Hiroshi M. Sasaki, Shun-ichi Sekine, Toru Sengoku, Ryuya Fukunaga, Motoyuki Hattori, Yukiko Utsunomiya, Chizu Kuroishi, Seiki Kuramitsu, Mikako Shirouzu, Shigeyuki Yokoyama

    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA   103 ( 40 )   14744 - 14749   2006年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:NATL ACAD SCIENCES  

    To achieve accurate aminoacylation of tRNAs with their cognate amino acids, errors in aminoacylation are corrected by the "editing" mechanism in several aminoacyl-tRNA synthetases. Phenylalanyl-tRNA synthetase (PheRS) hydrolyzes, or edits, misformed tyrosyl-tRNA with its editing domain in the beta subunit. We report the crystal structure of an N-terminal fragment of the PheRS beta subunit (PheRS-beta(N)) from the archaeon, Pyrococcus horikoshii, at 1.94-angstrom resolution. PheRS-beta(N) includes the editing domain B3/4, which has archaea/eukarya-specific insertions/deletions and adopts a different orientation relative to other domains, as compared with that of bacterial PheRS. Surprisingly, most residues constituting the editing active-site pocket were substituted between the archaeal/eukaryal and bacterial PheRSs. We prepared Ala-substituted mutants of A horikoshii PheRS for 16 editing pocket residues, of which 12 are archaea/eukarya-specific and four are more widely conserved. On the basis of their activities, Tyr-adenosine was modeled on the B3/4-domain structure. First, the mutations of Leu-202, Ser-211, Asp-234, and Thr-236 made the PheRS incorrectly hydrolyze the cognate Phe-tRNA(Phe), indicating that these residues participate in the Tyr hydroxy group recognition and are responsible for discrimination against Phe. Second, the mutations of Leu-168 and Arg-223, which could interact with the tRNA 3'-terminal adenosine, reduced Tyr-tRNA(Phe) deacylation activity. Third, the mutations of archaea/eukarya-specific Gln-126, Glu-127, Arg-137, and Asn-217, which are proximal to the ester bond to be cleaved, also reduced Tyr-tRNAPhe deacylation activity. In particular, the replacement of Asn-217 abolished the activity, revealing its absolute requirement for the catalysis.

    DOI: 10.1073/pnas.0603182103

    Web of Science

    PubMed

    J-GLOBAL

    researchmap

  • Crystal structure of tRNA adenosine deaminase (TadA) from Aquifex aeolicus 査読

    M Kuratani, R Ishii, Y Bessho, R Fukunaga, T Sengoku, M Shirouzu, S Sekine, S Yokoyama

    JOURNAL OF BIOLOGICAL CHEMISTRY   280 ( 16 )   16002 - 16008   2005年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    The bacterial tRNA adenosine deaminase ( TadA) generates inosine by deaminating the adenosine residue at the wobble position of tRNA(Arg-2). This modification is essential for the decoding system. In this study, we determined the crystal structure of Aquifex aeolicus TadA at a 1.8-angstrom resolution. This is the first structure of a deaminase acting on tRNA. A. aeolicus TadA has an alpha/beta/alpha three-layered fold and forms a homodimer. The A. aeolicus TadA dimeric structure is completely different from the tetrameric structure of yeast CDD1, which deaminates mRNA and cytidine, but is similar to the dimeric structure of yeast cytosine deaminase. However, in the A. aeolicus TadA structure, the shapes of the C-terminal helix and the regions between the beta 4 and beta 5 strands are quite distinct from those of yeast cytosine deaminase and a large cavity is produced. This cavity contains many conserved amino acid residues that are likely to be involved in either catalysis or tRNA binding. We made a docking model of TadA with the tRNA anticodon stem loop.

    DOI: 10.1074/jbc.M414541200

    Web of Science

    PubMed

    researchmap

  • A short peptide insertion crucial for angiostatic activity of human tryptophanyl-tRNA synthetase 査読

    Y Kise, SW Lee, SG Park, S Fukai, T Sengoku, R Ishii, S Yokoyama, S Kim, O Nureki

    NATURE STRUCTURAL & MOLECULAR BIOLOGY   11 ( 2 )   149 - 156   2004年2月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:NATURE PUBLISHING GROUP  

    Human tryptophanyl-tRNA synthetase (TrpRS) is secreted into the extracellular region of vascular endothelial cells. The splice variant form (mini TrpRS) functions in vascular endothelial cell apoptosis as an angiostatic cytokine. In contrast, the closely related human tyrosyl-tRNA synthetase (TyrRS) functions as an angiogenic cytokine in its truncated form (mini TyrRS). Here, we determined the crystal structure of human mini TrpRS at a resolution of 2.3 Angstrom and compared the structure with those of prokaryotic TrpRS and human mini TyrRS. Deletion of the tRNA anticodon-binding (TAB) domain insertion, consisting of eight residues in the human TrpRS, abolished the enzyme's apoptotic activity for endothelial cells, whereas its translational catalysis and cell-binding activities remained unchanged. Thus, we have identified the inserted peptide motif that activates the angiostatic signaling.

    DOI: 10.1038/nsmb722

    Web of Science

    researchmap

  • RNAヘリケースによるRNA translocationの構造的基盤

    仙石 徹, 濡木 理, 横山 茂之

    生物物理   43   S98   2003年

     詳細を見る

    記述言語:日本語   出版者・発行元:一般社団法人 日本生物物理学会  

    DOI: 10.2142/biophys.43.S98_2

    researchmap

▼全件表示

MISC

  • Cajal bodyにおけるLEC構成因子ZC3H8によるsnRNA遺伝子の発現制御機構の解明

    野口慶介, 鬼澤理紗, 阿部竜太, 池陽子, 仙石徹, 緒方一博, 井野洋子, 木村弥生, 鈴木秀文, 高橋秀尚

    日本薬学会年会要旨集(Web)   144th   2024年

     詳細を見る

  • Nrf2-小Mafヘテロ二量体による標的DNA認識機構のFMO法(フラグメント分子軌道法)による解析

    仙石徹, 森脇寛智, 渡邉千鶴, 本間光貴, 緒方一博

    日本分子生物学会年会プログラム・要旨集(Web)   42nd   2019年

     詳細を見る

  • 糖転移酵素EarP・翻訳因子EF-P複合体の構造から明らかになったEF-P(Arg32)ラムノシル化の反応メカニズム

    仙石徹, 鈴木健裕, 堂前直, 渡邊千鶴, 本間光貴, 疋田泰士, 山口芳樹, 高橋英之, 横山茂之, 柳沢達男

    日本RNA学会年会要旨集   20th   2018年

     詳細を見る

  • 反転型糖転移酵素によるタンパク質アルギニン・ラムノシル化の構造基盤

    仙石徹, 鈴木健裕, 堂前直, 渡邉千鶴, 本間光貴, 疋田泰士, 山口芳樹, 高橋英之, 柳沢達男, 横山茂之

    日本生化学会大会(Web)   90th   ROMBUNNO.2AW27‐6 (WEB ONLY)   2017年

     詳細を見る

    記述言語:日本語  

    J-GLOBAL

    researchmap

  • 髄膜炎菌由来の翻訳因子EF‐Pのラムノース修飾による活性化の構造的基盤

    仙石徹, 鈴木健裕, 堂前直, 渡邉千鶴, 本間光貴, 疋田泰士, 山口芳樹, 高橋英之, 横山茂之, 柳沢達男

    日本生化学会大会(Web)   90th   ROMBUNNO.4LT25‐09(3P‐0099) (WEB ONLY)   2017年

     詳細を見る

    記述言語:日本語  

    J-GLOBAL

    researchmap

  • 髄膜炎菌由来の翻訳因子EF‐Pのラムノース修飾による活性化の構造的基盤

    仙石徹, 柳沢達男, 鈴木健裕, 堂前直, 渡邉千鶴, 本間光貴, 疋田泰士, 高橋英之, 横山茂之

    日本分子生物学会年会プログラム・要旨集(Web)   39th   ROMBUNNO.3P‐0247 (WEB ONLY)   2016年

     詳細を見る

    記述言語:日本語  

    J-GLOBAL

    researchmap

  • アセチル化ヒストンH4K16を特異的に認識する抗体の結晶構造解析

    仙石徹, 森田鋭, 佐藤優子, KUMAREVEL Thirumananseri, 沖山佳生, 渡邉千鶴, 梅原崇史, 本間光貴, 横山茂之, 木村宏

    日本生化学会大会(Web)   88th   3P0620 (WEB ONLY)   2015年

     詳細を見る

    記述言語:日本語  

    J-GLOBAL

    researchmap

  • Catalytic fragment of UTX/KDM6A bound with histone H3K27me3 peptide, N-oxyalylglycine, and Ni(II)

    T. Sengoku, S. Yokoyama

    2011年10月

     詳細を見る

    担当区分:筆頭著者   出版者・発行元:Worldwide Protein Data Bank  

    DOI: 10.2210/pdb3avr/pdb

    researchmap

  • Tryptophanyl-tRNA synthetase for the Mycoplasma deviant genetic code

    KITAMURA Aya, SASAKI Hiroshi, HIGASHIJIMA Kyoko, NISHIMOTO Madoka, SENGOKU Toru, YOKOBORI Shin‐ichi, YOKOYAMA Shigeyuki, BESSHO Yoshitaka

    日本RNA学会年会要旨集   13th   372   2011年

     詳細を見る

    記述言語:英語  

    J-GLOBAL

    researchmap

  • Crystal structure of human LSD1 at 2.3 A resolution

    T. Sengoku, S. Yokoyama

    2007年9月

     詳細を見る

    担当区分:筆頭著者   出版者・発行元:Worldwide Protein Data Bank  

    DOI: 10.2210/pdb2dw4/pdb

    researchmap

  • Structural basis for RNA unwinding by the DEAD-box protein Drosophila Vasa

    T. Sengoku, O. Nureki, A. Nakamura, S. Kobayashi, S. Yokoyama

    2006年5月

     詳細を見る

    担当区分:筆頭著者   出版者・発行元:Worldwide Protein Data Bank  

    DOI: 10.2210/pdb2db3/pdb

    researchmap

  • Crystal structure of tRNA adenosine deaminase TadA from Aquifex aeolicus

    M. Kuratani, R. Ishii, Y. Bessho, R. Fukunaga, T. Sengoku, S. Sekine, S. Yokoyama

    2005年2月

     詳細を見る

    出版者・発行元:Worldwide Protein Data Bank  

    DOI: 10.2210/pdb1wwr/pdb

    researchmap

  • A short peptide insertion crucial for angiostatic activity of human tryptophanyl-tRNA synthetase

    Yoshiaki Kise, Sang Won Lee, Sang Gyu Park, Shuya Fukai, Toru Sengoku, Ryohei Ishii, Shigeyuki Yokoyama, Sunghoon Kim, Osamu Nureki

    Nature Structural and Molecular Biology   11 ( 2 )   149 - 156   2004年2月

     詳細を見る

    記述言語:英語   出版者・発行元:Worldwide Protein Data Bank  

    Human tryptophanyl-tRNA synthetase (TrpRS) is secreted into the extracellular region of vascular endothelial cells. The splice variant form (mini TrpRS) functions in vascular endothelial cell apoptosis as an angiostatic cytokine. In contrast, the closely related human tyrosyl-tRNA synthetase (TyrRS) functions as an angiogenic cytokine in its truncated form (mini TyrRS). Here, we determined the crystal structure of human mini TrpRS at a resolution of 2.3 Å and compared the structure with those of prokaryotic TrpRS and human mini TyrRS. Deletion of the tRNA anticodon-binding (TAB) domain insertion, consisting of eight residues in the human TrpRS, abolished the enzyme's apoptotic activity for endothelial cells, whereas its translational catalysis and cell-binding activities remained unchanged. Thus, we have identified the inserted peptide motif that activates the angiostatic signaling.

    DOI: 10.1038/nsmb722

    Scopus

    PubMed

    researchmap

▼全件表示

Works(作品等)

▼全件表示

共同研究・競争的資金等の研究課題

  • 時空間的な多因子間相互作用の理解による転写ユニティー機構の解明の総括

    研究課題/領域番号:21H05158  2021年8月 - 2024年3月

    日本学術振興会  科学研究費助成事業  学術変革領域研究(B)

    二村 圭祐, 高橋 秀尚, 仙石 徹

      詳細を見る

    配分額:1560000円 ( 直接経費:1200000円 、 間接経費:360000円 )

    「時空間的な多因子間相互作用の理解による転写ユニティー機構の解明」領域を発展させるためには、領域内で共同研究を推進すること、技術交流を積極的に進めることが重要である。また外部に向けて、領域の活動を積極的に広報することも必要である。現在までに、二村と高橋は共同研究を行い、スプライシング制御因子が転写を直接制御することを見出し、その成果は現在論文投稿中である。また、二村は開発中の新規タンパク質を構造学的視点から深めるために仙石とディスカッションを行っている。また、高橋は仙石と転写ユニティーを構築する因子に関して共同研究を行っている。新たな技術を開発するために、領域内で積極的に議論を行い、現在の技術でアプローチできない課題や、その課題を解決するために開発するべき技術の方向性などに関するブレインストーミングを行っている。その中から、転写ユニティー機構をさらに詳細に解析する新技術を考案し、現在その開発に向けて準備を行っている。また、構造生物学においてAlpha fold2のような構造を予測・解析する新規手法についても積極的に取り入れ、領域内の研究の進捗を加速するために利用している。
    広報のために転写ユニティー領域のホームページを作成し、公開した。今後、研究業績など、領域の活動を広く国民に対して公開していく予定である。また、領域内での研究交流セミナーを行うことで、さらに共同研究を増やし、迅速に研究成果を論文化できるように進めていく。

    researchmap

  • 転写ユニティー機構を構築する多因子間相互作用の構造的解明

    研究課題/領域番号:21H05161  2021年8月 - 2024年3月

    日本学術振興会  科学研究費助成事業  学術変革領域研究(B)

    仙石 徹

      詳細を見る

    配分額:27300000円 ( 直接経費:21000000円 、 間接経費:6300000円 )

    researchmap

  • トリプトファンプレニル基転移酵素のペプチド基質認識と反応機構の構造学的解明

    研究課題/領域番号:21K06051  2021年4月 - 2024年3月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    浜田 恵輔, 仙石 徹, 緒方 一博

      詳細を見る

    配分額:4030000円 ( 直接経費:3100000円 、 間接経費:930000円 )

    ペプチド創薬においてペプチドの膜透過性やプロテアーゼ耐性をいかに付与するかが課題となっている。プレニル基転移酵素によるペプチドのプレニル化はその解決法の一つに挙げられる。しかし、酵素の基質認識機構や反応機構の理解が十分進んでいないため、リード化合物検索に用いるプレニル化ペプチドライブラリーの構築に対する妨げになっている。
    微生物由来の酵素KgpFや酵素X(未発表のため仮名とする)はペプチド中のトリプトファンを基質とし、膜透過性向上が期待されるプレニル化ピロロインドリンを生成する。興味深いことに、これらの酵素は互いに異なる基質選択性をもち、立体化学的に異なる生成物を作り分ける。本研究では、分子構造解析により両酵素の基質特異性、反応機構を解明する。これらの知見は効率的なライブラリーのデザインを可能にし、また酵素の合理的改変を通じて多様性に富んだ次世代ライブラリーを構築するための基盤を与える。
    本研究では、トリプトファンプレニル基転移酵素であるKgpF (および酵素X)による基質認識の構造基盤を明らかにすることを目的として、酵素-プレニル基供与体アナログ(ジメチルアリルチオ二リン酸、DMASPP)-基質ペプチド複合体のX線結晶構造解析を推進した。
    これまでに酵素Xにおいては、5残基からなる活性上最適な基質ペプチドを用いて、酵素X-DMASPP-基質ペプチド複合体の結晶化に成功し、X線結晶構造解析を1.5Å分解能で行った。その結果、基質ペプチドのトリプトファンインドール環のNH基は類縁酵素間でよく保存され触媒に必須なグルタミン酸残基と水素結合を形成し認識されていた。また、このインドール環は、類縁酵素間で保存性が低い複数の残基とのファンデルワールス接触により認識されることが明らかになった。

    researchmap

  • 特殊ペプチド・擬天然物創薬の命題への挑戦

    研究課題/領域番号:20H05618  2020年7月 - 2025年3月

    日本学術振興会  科学研究費助成事業  特別推進研究

    菅 裕明, 仙石 徹

      詳細を見る

    配分額:631540000円 ( 直接経費:485800000円 、 間接経費:145740000円 )

    本特別推進研究計画の申請者は、一貫してCuriosity-Driven-ResearchとTechnology-Driven-Researchの両輪で研究を推進し、非タンパク質性アミノ酸を望みのtRNAにアシル化することを可能にしたリボザイム(フレキシザイム)の開発、フレキシザイムと再構成無細胞翻訳系(FITシステム)を組み合わせた遺伝暗号リプログラミング技術の革新とそれを活用した特殊ペプチドの翻訳合成、特殊ペプチドの翻訳合成と試験管内(mRNA)ディスプレイを組み合わせたRaPIDシステムにより生理活性中分子化合物の超迅速探索、を達成してきた。これまで様々な細胞内標的に対して特殊ペプチドを獲得してきたが、最近になりいくつかの特殊ペプチドに細胞膜透過性があることがわかった。しかし、その構造膜透過性相関に関しては十分な理解が進んでいない。また、ペプチド鎖への導入すら困難と言われてきたβアミノ酸やγアミノ酸に関しても、この数年で創出した人工tRNAを用いることにより、ようやく連続導入も可能であることを示した。しかし、これらの特殊アミノ酸を含む特殊ペプチドのライブラリー化や活性種探索はまだ初期段階にあり、そのポテンシャルの検討は十分できていない。また、天然物には遺伝暗号リプログラミングでは直接導入ができない化学構造を持っている分子も多く、それらを人工的に導入しライブラリーを構築する技術や探索の実証には十分至っていない。さらに、中分子薬剤に最も期待されている細胞膜透過性、さらには経口性につながる小腸吸収性をもつ化合物を確実に獲得する基盤技術としては未完成といえる。これらの残された課題を解決し突破することが、本研究の主題である。

    researchmap

  • 転写因子の天然変性領域におけるリン酸化ラッチ機構の普遍性の検証

    研究課題/領域番号:20K12069  2020年4月 - 2022年3月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    笠原 浩太, 仙石 徹

      詳細を見る

    配分額:4420000円 ( 直接経費:3400000円 、 間接経費:1020000円 )

    本研究は大きく下記3ステップより成る(a) ゲノムワイドな候補転写因子の探索、(b) 独自の分子動力学法(MD)を用いたメカニズム解析、(c) 実験による検証。
    (a) 蛋白質のデータベースであるUniProtよりヒト転写因子をゲノムワイドに取得し、天然変性領域(IDR)予測を網羅的に行った。さらにリン酸化部位のデータベースであるPhosphoSitePlusの情報をマッピングし、DNA結合ドメイン近傍のIDRに3ないし4残基間隔で2つのリン酸化部位が存在する転写因子を絞り込んだ。さらに目視で検討を行い、FOXO4、TGIF1、PC4の転写因子をリン酸化ラッチ機構を持つ転写因子の候補として同定した。
    (b) これら転写因子についてMDによる検討を行った。IDRによる複雑な構造アンサンブルをより正確に解析するため、MDの技術開発や基礎的検討も並行して行った。特に、独自のMD手法としてVirutal-system coupled canonical MD法の開発を行い、またMD計算を実施するためのソフトウェアとしてmyPresto/omegageneを開発した。転写因子FOXO4については独自の手法であるvirtual-system coupled multi-canonical MD法を適用し、TGIF1およびPC4ではさらに発展させた手法であるvirtual-system coupeld canonical MD法による検討を行った。いずれについてもDNA結合ドメインとその近傍のIDRを含む分子モデルを構築し、IDRのダイナミクスを解析した。特にFOXO4とTGIF1ではリン酸化状態と非リン酸化状態両方について計算をおこない、IDRのリン酸化部位がDNA結合領域と相互作用する結果が得られた。これは進化的に類縁関係にない複数の転写因子において共通してリン酸化ラッチ機構が働いていることを示唆し、そのメカニズムの普遍性をサポートしている。その結果を元に、(c)仮説を検証するin vitro実験の設計および予備的検討を進めた。

    researchmap

  • ヒストンメチル化状態の確立・維持・破綻機構

    研究課題/領域番号:20H05394  2020年4月 - 2022年3月

    日本学術振興会  科学研究費助成事業  新学術領域研究(研究領域提案型)

    仙石 徹

      詳細を見る

    配分額:8320000円 ( 直接経費:6400000円 、 間接経費:1920000円 )

    ヒストンH3 Lys36のジメチル化(H3K36me2)は遺伝子発現を制御するエピジェネティック修飾であり、その異常は様々ながんを引き起こす。血液がんの一部ではH3K36メチル化酵素NSD2の点変異E1099KによるNSD2活性の異常亢進が見られる一方、リンカーヒストンH1はNSD2の活性を抑制し、一部のリンパ腫に見られるH1遺伝子のミスセンス変異はH3K36me2レベル上昇を引き起こす。
    本研究では、クライオ電子顕微鏡を用いた単粒子解析により、NSD2の触媒ドメインがヌクレオソームに結合した複合体の構造を2.8 Å分解能で決定した。NSD2が結合したヌクレオソームはSHL 5.5より外側のDNAが剥がれており、それによりNSD2のH3K36へのアクセスが可能になっていた。H1はヌクレオソームの両側のリンカーDNAと同時に相互作用することによってヌクレオソーム構造を安定化することが知られている。従って、H1はヌクレオソームDNAを剥がれにくくすることでNSD2によるH3K36認識に必要なヌクレオソーム構造変化を妨げ、NSD2の活性を抑制していると考えられる。
    発がん性変異E1099Kによる活性異常亢進メカニズムを調べるために生化学的解析を行ったところ、E1099K変異はNSD2の触媒回転数を増加させる一方でヌクレオソームへの親和性には大きな影響を与えないことが明らかになった。また、分子動力学シミュレーション解析から、E1099K変異体においては自己阻害ループが開いた構造をとりやすくなっていることが示唆された。これらの結果から、我々は「発がん性E1099Kは自己阻害ループの柔軟性に影響を与え、H3結合に適したコンフォメーションを取りやすくすることで活性を異常亢進させる」というメカニズムを提唱する。

    researchmap

  • ヌクレオソームーエンハンソソーム変換の機構の解明

    研究課題/領域番号:18K19305  2018年6月 - 2021年3月

    日本学術振興会  科学研究費助成事業  挑戦的研究(萌芽)

    緒方 一博, 仙石 徹, 浜田 恵輔

      詳細を見る

    配分額:6370000円 ( 直接経費:4900000円 、 間接経費:1470000円 )

    転写活性化に伴うヌクレオソームーエンハンソソーム構造変換にはヒストン修飾酵素が重要な役割を果たす。本研究では、クライオ電子顕微鏡を用いた単粒子解析法により、ヒストンメチル化酵素の一つであり、ヒストンH3のリジン36(H3K36)をジメチル化するNSD2と基質であるヌクレオソームとの複合体の立体構造を決定した。NSD2は単独では自己阻害ループにより不活性であるが、ヌクレオソームが存在すると、自己阻害ループの構造変化とDNAのヌクレオソームからの解けが協調してH3K36のメチル化が行われ、がん化型NSD2変異体では自己阻害ループの作用の損傷により、酵素が脱抑制状態になっていることが明らかになった。

    researchmap

  • がん関連転写因子Runx1/CBFβを標的とした活性制御薬物の研究

    研究課題/領域番号:16H03293  2016年4月 - 2019年3月

    日本学術振興会  科学研究費助成事業  基盤研究(B)

    緒方 一博, 浜田 恵輔, 仙石 徹, 椎名 政昭

      詳細を見る

    配分額:18850000円 ( 直接経費:14500000円 、 間接経費:4350000円 )

    がんの原因分子を標的とする薬剤を用いたがん分子標的治療は、様々ながんに対して延命効果を示している。しかし最も重要な原因分子の一種である転写因子の多くは、その対象となっていない。そこで我々は、急性白血病の発症・進展に関与する転写因子Runx1/CBFβを対象とし、低分子阻害薬の開発を進めた。
    本研究では、分子構造に基づきRunx1のDNA結合の制御部位に対して作用する化合物の探索を行い、複数種類の化合物がRunx1との結合を介してDNA結合を阻害することを確認した。さらにRunx1に変異を有する白血病細胞株において、これら化合物がRunx1の転写活性化能を抑制し、生存率を低下させることを示した。

    researchmap

  • ヒストン脱メチル化酵素の反応機構解析と阻害剤開発

    研究課題/領域番号:15K21626  2015年4月 - 2019年3月

    日本学術振興会  科学研究費助成事業  若手研究(B)

    仙石 徹

      詳細を見る

    配分額:3900000円 ( 直接経費:3000000円 、 間接経費:900000円 )

    タンパク質の翻訳後修飾を操作する低分子は、生物学研究ツールとして有用であると共に創薬リード化合物として豊かな可能性を持つ。本研究では、塩基性残基の修飾に関与する以下の2種類の酵素について、酵素の反応機構理解と阻害剤開発を目標とした。ヒストンH3K27脱メチル化酵素KDM6Aについて構造解析と阻害剤開発を行い、in silicoスクリーニングにより得られた化合物のうち一つがH3K27メチル化に異常を持つグリオーマ細胞の増殖を抑制することを見出した。また、アルギニンラムノシル化酵素EarPについて、その標的タンパク質である翻訳因子EF-Pとの複合体の構造を決定し、反応機構の構造基盤を明らかにした。

    researchmap

  • ヒストンメチル化酵素複合体の構造解析

    研究課題/領域番号:25121740  2013年4月 - 2015年3月

    日本学術振興会  科学研究費助成事業  新学術領域研究(研究領域提案型)

    仙石 徹

      詳細を見る

    配分額:4680000円 ( 直接経費:3600000円 、 間接経費:1080000円 )

    Polycomb Repressive Complex 2 (PRC2)はヒストンH3 Lys27のトリメチル化(H3K27me3)を触媒するタンパク質複合体であり、標的遺伝子の発現を抑制することにより他細胞生物の分化と癌化において重要な役割を果たす。本研究では、PRC2の構造機能解析を目的として研究を行った。HEK293細胞においてPRC2のコアサブユニット4種類を共発現させ、アフィニティークロマトグラフィーとゲルろ過クロマトグラフィーにより4者複合体の調製に成功した。この手法は、これまでに報告されていた昆虫細胞を利用する方法より収量が良く、構造解析のための資料調製として適した方法と考えられる。さらに、付加サブユニットAEBP2を大腸菌で単独発現・精製し、上記の4者複合体と混合してさらに精製することにより、5者複合体の調製に成功した。これらの複合体について、結晶化スクリーニングと低温電子顕微鏡による構造解析のための条件検討を行っている。
    MLL1複合体はヒストンH3 Lys4をメチル化するタンパク質複合体であり、標的遺伝子の発現を正に制御している。MLL1複合体によるクロマチンのメチル化には、Bre1複合体によるヒストンH2Bのユビキチン化が必要である。このヒストン修飾のクロストークの構造的基盤を得るため、MLL1複合体とBre1複合体の再構成を行った。MLL1複合体の5種類のコアサブユニットを別々に大腸菌で発現させ、菌体を混合しアフィニティークロマトグラフィーとゲルろ過クロマトグラフィーにより5者複合体の調製に成功した。また、Bre1AとBre1Bを昆虫細胞で共発現させ、アフィニティークロマトグラフィーとゲルろ過クロマトグラフィーにより2者複合体の調製に成功した。これらについて、結晶化スクリーニングが進行中である。

    researchmap

  • 細胞分化を制御するヒストン脱メチル化酵素の構造機能解析

    研究課題/領域番号:23770131  2011年 - 2012年

    日本学術振興会  科学研究費助成事業  若手研究(B)

    仙石 徹

      詳細を見る

    配分額:4550000円 ( 直接経費:3500000円 、 間接経費:1050000円 )

    H3K27特異的脱メチル化酵素UTXは、転写抑制マークのH3K27me3を脱メチル化することにより動物細胞の分化・運命決定に関与する。本研究では、UTXのH3K27特異性決定機構を解明するため、X線結晶構造解析によりUTXとH3ペプチドとの複合体の立体構造を決定した。その結果,UTXは触媒ドメイン(Jumonjiドメイン)の他に固有の亜鉛結合ドメインを持つことが分かった。JumonjiドメインはH3K27とその周辺の残基を、亜鉛結合ドメインはH3L20とその周辺の残基を認識しており、2つのドメインを介してH3の幅広い領域を認識することにより厳密な特異性を獲得していることが明らかになった.

    researchmap

▼全件表示