Network


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

Hotspot


Dive into the research topics where Kengo Kitadokoro is active.

Publication


Featured researches published by Kengo Kitadokoro.


The EMBO Journal | 2001

CD81 extracellular domain 3D structure: insight into the tetraspanin superfamily structural motifs

Kengo Kitadokoro; Domenico Bordo; Giuliano Galli; Roberto Petracca; Fabiana Falugi; Sergio Abrignani; Guido Grandi; Martino Bolognesi

Human CD81, a known receptor for hepatitis C virus envelope E2 glycoprotein, is a transmembrane protein belonging to the tetraspanin family. The crystal structure of human CD81 large extracellular domain is reported here at 1.6 Å resolution. Each subunit within the homodimeric protein displays a mushroom‐like structure, composed of five α‐helices arranged in ‘stalk’ and ‘head’ subdomains. Residues known to be involved in virus binding can be mapped onto the head subdomain, providing a basis for the design of antiviral drugs and vaccines. Sequence analysis of 160 tetraspanins indicates that key structural features and the new protein fold observed in the CD81 large extracellular domain are conserved within the family. On these bases, it is proposed that tetraspanins may assemble at the cell surface into homo‐ and/or hetero‐dimers through a conserved hydrophobic interface located in the stalk subdomain, while interacting with other liganding proteins, including hepatitis C virus E2, through the head subdomain. The topology of such interactions provides a rationale for the assembly of the so‐called tetraspan‐web.


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

Crystal structures reveal a thiol protease-like catalytic triad in the C-terminal region of Pasteurella multocida toxin

Kengo Kitadokoro; Shigeki Kamitani; Masayuki Miyazawa; Miyuki Hanajima-Ozawa; Aya Fukui; Masami Miyake; Yasuhiko Horiguchi

Pasteurella multocida toxin (PMT), one of the virulence factors produced by the bacteria, exerts its toxicity by up-regulating various signaling cascades downstream of the heterotrimeric GTPases Gq and G12/13 in an unknown fashion. Here, we present the crystal structure of the C-terminal region (residues 575–1,285) of PMT, which carries an intracellularly active moiety. The overall structure of C-terminal region of PMT displays a Trojan horse-like shape, composed of three domains with a “feet”-,“body”-, and “head”-type arrangement, which were designated C1, C2, and C3 from the N to the C terminus, respectively. The C1 domain, showing marked similarity in steric structure to the N-terminal domain of Clostridium difficile toxin B, was found to lead the toxin molecule to the plasma membrane. The C3 domain possesses the Cys–His–Asp catalytic triad that is organized only when the Cys is released from a disulfide bond. The steric alignment of the triad corresponded well to that of papain or other enzymes carrying Cys–His–Asp. PMT toxicities on target cells were completely abrogated when one of the amino acids constituting the triad was mutated. Our results indicate that PMT is an enzyme toxin carrying the cysteine protease-like catalytic triad dependent on the redox state and functions on the cytoplasmic face of the plasma membrane of target cells.


Journal of Biological Chemistry | 2011

Crystal Structure of Clostridium perfringens Enterotoxin Displays Features of β-Pore-forming Toxins

Kengo Kitadokoro; Kousuke Nishimura; Shigeki Kamitani; Aya Fukui-Miyazaki; Hirono Toshima; Hiroyuki Abe; Yoichi Kamata; Yoshiko Sugita-Konishi; Shigeki Yamamoto; Hajime Karatani; Yasuhiko Horiguchi

Clostridium perfringens enterotoxin (CPE) is a cause of food poisoning and is considered a pore-forming toxin, which damages target cells by disrupting the selective permeability of the plasma membrane. However, the pore-forming mechanism and the structural characteristics of the pores are not well documented. Here, we present the structure of CPE determined by x-ray crystallography at 2.0 Å. The overall structure of CPE displays an elongated shape, composed of three distinct domains, I, II, and III. Domain I corresponds to the region that was formerly referred to as C-CPE, which is responsible for binding to the specific receptor claudin. Domains II and III comprise a characteristic module, which resembles those of β-pore-forming toxins such as aerolysin, C. perfringens ϵ-toxin, and Laetiporus sulfureus hemolytic pore-forming lectin. The module is mainly made up of β-strands, two of which span its entire length. Domain II and domain III have three short β-strands each, by which they are distinguished. In addition, domain II has an α-helix lying on the β-strands. The sequence of amino acids composing the α-helix and preceding β-strand demonstrates an alternating pattern of hydrophobic residues that is characteristic of transmembrane domains forming β-barrel-made pores. These structural features imply that CPE is a β-pore-forming toxin. We also hypothesize that the transmembrane domain is inserted into the membrane upon the buckling of the two long β-strands spanning the module, a mechanism analogous to that of the cholesterol-dependent cytolysins.


Genes to Cells | 2009

Human Nei-like protein NEIL3 has AP lyase activity specific for single-stranded DNA and confers oxidative stress resistance in Escherichia coli mutant

Masashi Takao; Yoshitsugu Oohata; Kengo Kitadokoro; Kumiko Kobayashi; Shigenori Iwai; Akira Yasui; Shuji Yonei; Qiu-Mei Zhang

Oxidative base damage leads to alteration of genomic information and is implicated as a cause of aging and carcinogenesis. To combat oxidative damage to DNA, cells contain several DNA glycosylases including OGG1, NTH1 and the Nei‐like proteins, NEIL1 and NEIL2. A third Nei‐like protein, NEIL3, is composed of an amino‐terminal Nei‐like domain and an unknown carboxy‐terminal domain. In contrast to the other well‐described DNA glycosylases, the DNA glycosylase activity and in vivo repair function of NEIL3 remains unclear. We show here that the structural modeling of the putative NEIL3 glycosylase domain (1–290) fits well to the known Escherichia coli Fpg crystal structure. In spite of the structural similarity, the recombinant NEIL3 and NEIL3(1–290) proteins do not cleave any of several test oligonucleotides containing a single modified base. Within the substrates, we detected AP lyase activity for single‐stranded (ss) DNA but double‐stranded (ds) DNA. The activity is abrogated completely in mutants with an amino‐terminal deletion and at the zinc‐finger motif. Surprisingly, NEIL3 partially rescues an E. coli nth nei mutant from hydrogen peroxide sensitivity. Taken together, repair of certain base damage including base loss in ssDNA may be mediated by NEIL3.


Biological Chemistry | 2002

Subunit Association and Conformational Flexibility in the Head-subdomain of Human CD81 Large Extracellular Loop.

Kengo Kitadokoro; Marco Ponassi; Giuliano Galli; Roberto Petracca; Fabiana Falugi; Guido Grandi; Martino Bolognesi

Abstract The large extracellular loop of human CD81, a tetraspanin mediating hepatitis C virus envelope protein E2 binding to human cells, has been crystallized in a hexagonal form. The threedimensional structure, solved and refined at 2.6 å resolution (Rfactor = 22.8%), shows that the protein adopts a dimeric assembly, based on an association interface built up by tetraspaninconserved residues. Structural comparisons with the tertiary structure of human CD81 large extracellular loop, previously determined in a different crystal form, show marked conformational fluctuations in the molecular regions thought to be involved in binding to the viral protein, suggesting rules for recognition and assembly within the tetraspan web.


Journal of Biological Chemistry | 2010

Characterization of the Membrane-targeting C1 Domain in Pasteurella multocida Toxin

Shigeki Kamitani; Kengo Kitadokoro; Masayuki Miyazawa; Hirono Toshima; Aya Fukui; Hiroyuki Abe; Masami Miyake; Yasuhiko Horiguchi

Pasteurella multocida toxin (PMT) is a virulence factor responsible for the pathogenesis of some forms of pasteurellosis. The toxin activates Gq- and G12/13-dependent pathways through the deamidation of a glutamine residue in the α-subunit of heterotrimeric GTPases. We recently reported the crystal structure of the C terminus (residues 575–1285) of PMT (C-PMT), which is composed of three domains (C1, C2, and C3), and that the C1 domain is involved in the localization of C-PMT to the plasma membrane, and the C3 domain possesses a cysteine protease-like catalytic triad. In this study, we analyzed the membrane-targeting function of the C1 domain in detail. The C1 domain consists of seven helices of which the first four (residues 590–670), showing structural similarity to the N terminus of Clostridium difficile toxin B, were found to be involved in the recruitment of C-PMT to the plasma membrane. C-PMT lacking these helices (C-PMT ΔC1(4H)) neither localized to the plasma membrane nor stimulated the Gq/12/13-dependent signaling pathways. When the membrane-targeting property was complemented by a peptide tag with an N-myristoylation motif, C-PMT ΔC1(4H) recovered the PMT activity. Direct binding between the C1 domain and liposomes containing phospholipids was evidenced by surface plasmon resonance analyses. These results indicate that the C1 domain of C-PMT functions as a targeting signal for the plasma membrane.


Acta Crystallographica Section D-biological Crystallography | 2001

Crystallization and preliminary crystallographic studies on the large extracellular domain of human CD81, a tetraspanin receptor for hepatitis C virus.

Kengo Kitadokoro; Giuliano Galli; Roberto Petracca; Fabiana Falugi; Guido Grandi; Martino Bolognesi

The large extracellular domain of CD81, a member of the tetraspanin family and a receptor protein for hepatitis C virus envelope E2 glycoprotein, has been expressed, purified and subsequently crystallized using the sitting-drop vapour-diffusion technique. Native diffraction data to 1.6 A resolution were obtained at the ID14 beamline of the European Synchrotron Radiation Facility from a flash-frozen crystal at 100 K. The crystals belong to space group P2(1), with unit-cell parameters a = 31.5, b = 77.2, c = 38.5 A, beta = 107.4 degrees, and are likely to contain two extracellular domains (2 x 99 residues) per asymmetric unit.


Acta Crystallographica Section F-structural Biology and Crystallization Communications | 2006

Crystallization and preliminary crystallographic studies of the Pasteurella multocida toxin catalytic domain

Masayuki Miyazawa; Kengo Kitadokoro; Shigeki Kamitani; Hiroaki Shime; Yasuhiko Horiguchi

The C-terminal catalytic domain of Pasteurella multocida toxin, which is the virulence factor of the organism in P. multocida, has been expressed, purified and subsequently crystallized using the sitting-drop vapour-diffusion technique. Native diffraction data to 1.9 A resolution were obtained at the BL44XU beamline of SPring-8 from a flash-frozen crystal at 100 K. The crystals belong to space group C2, with unit-cell parameters a = 111.0, b = 150.4, c = 77.1 A, beta = 105.5 degrees, and are likely to contain one C-PMT (726 residues) per asymmetric unit.


Advances in Bioscience and Biotechnology | 2018

Expression, Purification and Crystallization of Thermostable Mutant of Cutinase Est1 from Thermobifida alba

Kengo Kitadokoro; Shingo Matsui; Ryouhei Osokoshi; Kensuke Nakata; Shigeki Kamitani

A double mutantEst1, which is a plastic degrading cutinase-type esterase in Thermobifida alba, has been over-expressed in Escherichia coli. The recombinant protein was purified by a two-step protocol involving immobilized metal affinity chromatography and cation-exchange chromatography, yielding 120 mg of protein per liter of bacterial culture. Crystals have been obtained by using the sitting-drop vapor-diffusion technique. Native diffraction data to 1.37 A resolution were obtained at the BL44XU beam line of SPring-8 from a flash-frozen crystal at 100 K. The crystals belong to space group C2, with unit-cell parameters a = 127.2 A, b = 42.1 A, c = 63.2 A, β = 114.7°, likely containing one Est1 double mutant (296 residues) per asymmetric unit.


Journal of Biochemistry | 1998

Crystal Structure of Human Secretory Phospholipase A2-IIA Complex with the Potent Indolizine Inhibitor 120–1032

Kengo Kitadokoro; Sanji Hagishita; Tomohiro Sato; Mitsuaki Ohtani; Kunio Miki

Collaboration


Dive into the Kengo Kitadokoro's collaboration.

Top Co-Authors

Avatar

Shigeki Kamitani

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge