Network


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

Hotspot


Dive into the research topics where Kengo Tsuda is active.

Publication


Featured researches published by Kengo Tsuda.


Nucleic Acids Research | 2009

Structural basis for the sequence-specific RNA-recognition mechanism of human CUG-BP1 RRM3

Kengo Tsuda; Kanako Kuwasako; Mari Takahashi; Tatsuhiko Someya; Makoto Inoue; Takaho Terada; Naohiro Kobayashi; Mikako Shirouzu; Takanori Kigawa; Akiko Tanaka; Sumio Sugano; Peter Güntert; Yutaka Muto; Shigeyuki Yokoyama

The CUG-binding protein 1 (CUG-BP1) is a member of the CUG-BP1 and ETR-like factors (CELF) family or the Bruno-like family and is involved in the control of splicing, translation and mRNA degradation. Several target RNA sequences of CUG-BP1 have been predicted, such as the CUG triplet repeat, the GU-rich sequences and the AU-rich element of nuclear pre-mRNAs and/or cytoplasmic mRNA. CUG-BP1 has three RNA-recognition motifs (RRMs), among which the third RRM (RRM3) can bind to the target RNAs on its own. In this study, we solved the solution structure of the CUG-BP1 RRM3 by hetero-nuclear NMR spectroscopy. The CUG-BP1 RRM3 exhibited a noncanonical RRM fold, with the four-stranded β-sheet surface tightly associated with the N-terminal extension. Furthermore, we determined the solution structure of the CUG-BP1 RRM3 in the complex with (UG)3 RNA, and discovered that the UGU trinucleotide is specifically recognized through extensive stacking interactions and hydrogen bonds within the pocket formed by the β-sheet surface and the N-terminal extension. This study revealed the unique mechanism that enables the CUG-BP1 RRM3 to discriminate the short RNA segment from other sequences, thus providing the molecular basis for the comprehension of the role of the RRM3s in the CELF/Bruno-like family.


Nucleic Acids Research | 2011

Structural basis for the dual RNA-recognition modes of human Tra2-β RRM

Kengo Tsuda; Tatsuhiko Someya; Kanako Kuwasako; Mari Takahashi; Fahu He; Satoru Unzai; Makoto Inoue; Takushi Harada; Satoru Watanabe; Takaho Terada; Naohiro Kobayashi; Mikako Shirouzu; Takanori Kigawa; Akiko Tanaka; Sumio Sugano; Peter Güntert; Shigeyuki Yokoyama; Yutaka Muto

Human Transformer2-β (hTra2-β) is an important member of the serine/arginine-rich protein family, and contains one RNA recognition motif (RRM). It controls the alternative splicing of several pre-mRNAs, including those of the calcitonin/calcitonin gene-related peptide (CGRP), the survival motor neuron 1 (SMN1) protein and the tau protein. Accordingly, the RRM of hTra2-β specifically binds to two types of RNA sequences [the CAA and (GAA)2 sequences]. We determined the solution structure of the hTra2-β RRM (spanning residues Asn110–Thr201), which not only has a canonical RRM fold, but also an unusual alignment of the aromatic amino acids on the β-sheet surface. We then solved the complex structure of the hTra2-β RRM with the (GAA)2 sequence, and found that the AGAA tetra-nucleotide was specifically recognized through hydrogen-bond formation with several amino acids on the N- and C-terminal extensions, as well as stacking interactions mediated by the unusually aligned aromatic rings on the β-sheet surface. Further NMR experiments revealed that the hTra2-β RRM recognizes the CAA sequence when it is integrated in the stem-loop structure. This study indicates that the hTra2-β RRM recognizes two types of RNA sequences in different RNA binding modes.


Nucleic Acids Research | 2012

Structure of Musashi1 in a complex with target RNA: the role of aromatic stacking interactions

Takako Ohyama; Takashi Nagata; Kengo Tsuda; Naohiro Kobayashi; Takao Imai; Hideyuki Okano; Toshio Yamazaki; Masato Katahira

Mammalian Musashi1 (Msi1) is an RNA-binding protein that regulates the translation of target mRNAs, and participates in the maintenance of cell ‘stemness’ and tumorigenesis. Msi1 reportedly binds to the 3′-untranslated region of mRNA of Numb, which encodes Notch inhibitor, and impedes initiation of its translation by competing with eIF4G for PABP binding, resulting in triggering of Notch signaling. Here, the mechanism by which Msi1 recognizes the target RNA sequence using its Ribonucleoprotein (RNP)-type RNA-binding domains (RBDs), RBD1 and RBD2 has been revealed on identification of the minimal binding RNA for each RBD and determination of the three-dimensional structure of the RBD1:RNA complex. Unique interactions were found for the recognition of the target sequence by Msi1 RBD1: adenine is sandwiched by two phenylalanines and guanine is stacked on the tryptophan in the loop between β1 and α1. The minimal recognition sequences that we have defined for Msi1 RBD1 and RBD2 have actually been found in many Msi1 target mRNAs reported to date. The present study provides molecular clues for understanding the biology involving Musashi family proteins.


FEBS Letters | 2012

Structural insight into the interaction of ADP-ribose with the PARP WWE domains

Fahu He; Kengo Tsuda; Mari Takahashi; Kanako Kuwasako; Takaho Terada; Mikako Shirouzu; Satoru Watanabe; Takanori Kigawa; Naohiro Kobayashi; Peter Güntert; Shigeyuki Yokoyama; Yutaka Muto

The WWE domain is often identified in proteins associated with ubiquitination or poly‐ADP‐ribosylation. Structural information about WWE domains has been obtained for the ubiquitination‐related proteins, such as Deltex and RNF146, but not yet for the poly‐ADP‐ribose polymerases (PARPs). Here we determined the solution structures of the WWE domains from PARP11 and PARP14, and compared them with that of the RNF146 WWE domain. NMR perturbation experiments revealed the specific differences in their ADP‐ribose recognition modes that correlated with their individual biological activities. The present structural information sheds light on the ADP‐ribose recognition modes by the PARP WWE domains.


Protein Science | 2008

Solution structure of the RNA binding domain in the human muscleblind-like protein 2.

Fahu He; Weirong Dang; Chikage Abe; Kengo Tsuda; Makoto Inoue; Satoru Watanabe; Naohiro Kobayashi; Takanori Kigawa; Takayoshi Matsuda; Takashi Yabuki; Masaaki Aoki; Eiko Seki; Takushi Harada; Yuri Tomabechi; Takaho Terada; Mikako Shirouzu; Akiko Tanaka; Peter Güntert; Yutaka Muto; Shigeyuki Yokoyama

The muscleblind‐like (MBNL) proteins 1, 2, and 3, which contain four CCCH zinc finger motifs (ZF1–4), are involved in the differentiation of muscle inclusion by controlling the splicing patterns of several pre‐mRNAs. Especially, MBNL1 plays a crucial role in myotonic dystrophy. The CCCH zinc finger is a sequence motif found in many RNA binding proteins and is suggested to play an important role in the recognition of RNA molecules. Here, we solved the solution structures of both tandem zinc finger (TZF) motifs, TZF12 (comprising ZF1 and ZF2) and TZF34 (ZF3 and ZF4), in MBNL2 from Homo sapiens. In TZF12 of MBNL2, ZF1 and ZF2 adopt a similar fold, as reported previously for the CCCH‐type zinc fingers in the TIS11d protein. The linker between ZF1 and ZF2 in MBNL2 forms an antiparallel β‐sheet with the N‐terminal extension of ZF1. Furthermore, ZF1 and ZF2 in MBNL2 interact with each other through hydrophobic interactions. Consequently, TZF12 forms a single, compact global fold, where ZF1 and ZF2 are approximately symmetrical about the C2 axis. The structure of the second tandem zinc finger (TZF34) in MBNL2 is similar to that of TZF12. This novel three‐dimensional structure of the TZF domains in MBNL2 provides a basis for functional studies of the CCCH‐type zinc finger motifs in the MBNL protein family.


Journal of Molecular Biology | 2010

Solution Structure of Histone Chaperone ANP32B: Interaction with Core Histones H3–H4 through Its Acidic Concave Domain

Naoya Tochio; Takashi Umehara; Yoshiko Munemasa; Toru Suzuki; Shin Sato; Kengo Tsuda; Seizo Koshiba; Takanori Kigawa; Ryozo Nagai; Shigeyuki Yokoyama

Eukaryotic gene expression is regulated by histone deposition onto and eviction from nucleosomes, which are mediated by several chromatin-modulating factors. Among them, histone chaperones are key factors that facilitate nucleosome assembly. Acidic nuclear phosphoprotein 32B (ANP32B) belongs to the ANP32 family, which shares N-terminal leucine-rich repeats (LRRs) and a C-terminal variable anionic region. The C-terminal region functions as an inhibitor of histone acetylation, but the functional roles of the LRR domain in chromatin regulation have remained elusive. Here, we report that the LRR domain of ANP32B possesses histone chaperone activity and forms a curved structure with a parallel beta-sheet on the concave side and mostly helical elements on the convex side. Our analyses revealed that the interaction of ANP32B with the core histones H3-H4 occurs on its concave side, and both the acidic and hydrophobic residues that compose the concave surface are critical for histone binding. These results provide a structural framework for understanding the functional mechanisms of acidic histone chaperones.


ChemBioChem | 2015

Controlling the Fluorescence of Benzofuran‐Modified Uracil Residues in Oligonucleotides by Triple‐Helix Formation

Takashi Kanamori; Hiroki Ohzeki; Yoshiaki Masaki; Akihiro Ohkubo; Mari Takahashi; Kengo Tsuda; Takuhiro Ito; Mikako Shirouzu; Kanako Kuwasako; Yutaka Muto; Mitsuo Sekine; Kohji Seio

We developed fluorescent turn‐on probes containing a fluorescent nucleoside, 5‐(benzofuran‐2‐yl)deoxyuridine (dUBF) or 5‐(3‐methylbenzofuran‐2‐yl)deoxyuridine (dUMBF), for the detection of single‐stranded DNA or RNA by utilizing DNA triplex formation. Fluorescence measurements revealed that the probe containing dUMBF achieved superior fluorescence enhancement than that containing dUBF. NMR and fluorescence analyses indicated that the fluorescence intensity increased upon triplex formation partly as a consequence of a conformational change at the bond between the 3‐methylbenzofuran and uracil rings. In addition, it is suggested that the microenvironment around the 3‐methylbenzofuran ring contributed to the fluorescence enhancement. Further, we developed a method for detecting RNA by rolling circular amplification in combination with triplex‐induced fluorescence enhancement of the oligonucleotide probe containing dUMBF.


Protein Science | 2007

Solution structure of the zinc finger HIT domain in protein FON

Fahu He; Takashi Umehara; Kengo Tsuda; Makoto Inoue; Takanori Kigawa; Takayoshi Matsuda; Takashi Yabuki; Masaaki Aoki; Eiko Seki; Takaho Terada; Mikako Shirouzu; Akiko Tanaka; Sumio Sugano; Yutaka Muto; Shigeyuki Yokoyama

The zinc finger HIT domain is a sequence motif found in many proteins, including thyroid hormone receptor interacting protein 3 (TRIP‐3), which is possibly involved in maturity‐onset diabetes of the young (MODY). Novel zinc finger motifs are suggested to play important roles in gene regulation and chromatin remodeling. Here, we determined the high‐resolution solution structure of the zinc finger HIT domain in ZNHIT2 (protein FON) from Homo sapiens, by an NMR method based on 567 upper distance limits derived from NOE intensities measured in three‐dimensional NOESY spectra. The structure yielded a backbone RMSD to the mean coordinates of 0.19 Å for the structured residues 12–48. The fold consists of two consecutive antiparallel β‐sheets and two short C‐terminal helices packed against the second β‐sheet, and binds two zinc ions. Both zinc ions are coordinated tetrahedrally via a CCCC‐CCHC motif to the ligand residues of the zf‐HIT domain in an interleaved manner. The tertiary structure of the zinc finger HIT domain closely resembles the folds of the B‐box, RING finger, and PHD domains with a cross‐brace zinc coordination mode, but is distinct from them. The unique three‐dimensional structure of the zinc finger HIT domain revealed a novel zinc‐binding fold, as a new member of the treble clef domain family. On the basis of the structural data, we discuss the possible functional roles of the zinc finger HIT domain.


Nature Structural & Molecular Biology | 2014

RBFOX and SUP-12 sandwich a G base to cooperatively regulate tissue-specific splicing

Kanako Kuwasako; Mari Takahashi; Satoru Unzai; Kengo Tsuda; Seiko Yoshikawa; Fahu He; Naohiro Kobayashi; Peter Güntert; Mikako Shirouzu; Takuhiro Ito; Akiko Tanaka; Shigeyuki Yokoyama; Masatoshi Hagiwara; Hidehito Kuroyanagi; Yutaka Muto

Tissue-specific alternative pre-mRNA splicing is often cooperatively regulated by multiple splicing factors, but the structural basis of cooperative RNA recognition is poorly understood. In Caenorhabditis elegans, ligand binding specificity of fibroblast growth factor receptors (FGFRs) is determined by mutually exclusive alternative splicing of the sole FGFR gene, egl-15. Here we determined the solution structure of a ternary complex of the RNA-recognition motif (RRM) domains from the RBFOX protein ASD-1, SUP-12 and their target RNA from egl-15. The two RRM domains cooperatively interact with the RNA by sandwiching a G base to form the stable complex. Multichromatic fluorescence splicing reporters confirmed the requirement of the G and the juxtaposition of the respective cis elements for effective splicing regulation in vivo. Moreover, we identified a new target for the heterologous complex through an element search, confirming the functional significance of the intermolecular coordination.


FEBS Open Bio | 2014

The zinc-binding region (ZBR) fragment of Emi2 can inhibit APC/C by targeting its association with the coactivator Cdc20 and UBE2C-mediated ubiquitylation

Shisako Shoji; Yutaka Muto; Mariko Ikeda; Fahu He; Kengo Tsuda; Noboru Ohsawa; Ryogo Akasaka; Takaho Terada; Motoaki Wakiyama; Mikako Shirouzu; Shigeyuki Yokoyama

Anaphase‐promoting complex or cyclosome (APC/C) is a multisubunit ubiquitin ligase E3 that targets cell‐cycle regulators. Cdc20 is required for full activation of APC/C in M phase, and mediates substrate recognition. In vertebrates, Emi2/Erp1/FBXO43 inhibits APC/C‐Cdc20, and functions as a cytostatic factor that causes long‐term M phase arrest of mature oocytes. In this study, we found that a fragment corresponding to the zinc‐binding region (ZBR) domain of Emi2 inhibits cell‐cycle progression, and impairs the association of Cdc20 with the APC/C core complex in HEK293T cells. Furthermore, we revealed that the ZBR fragment of Emi2 inhibits in vitro ubiquitin chain elongation catalyzed by the APC/C cullin‐RING ligase module, the ANAPC2–ANAPC11 subcomplex, in combination with the ubiquitin chain‐initiating E2, E2C/UBE2C/UbcH10. Structural analyses revealed that the Emi2 ZBR domain uses different faces for the two mechanisms. Thus, the double‐faced ZBR domain of Emi2 antagonizes the APC/C function by inhibiting both the binding with the coactivator Cdc20 and ubiquitylation mediated by the cullin‐RING ligase module and E2C. In addition, the tail region between the ZBR domain and the C‐terminal RL residues [the post‐ZBR (PZ) region] interacts with the cullin subunit, ANAPC2. In the case of the ZBR fragment of the somatic paralogue of Emi2, Emi1/FBXO5, these inhibitory activities against cell division and ubiquitylation were not observed. Finally, we identified two sets of key residues in the Emi2 ZBR domain that selectively exert each of the dual Emi2‐specific modes of APC/C inhibition, by their mutation in the Emi2 ZBR domain and their transplantation into the Emi1 ZBR domain.

Collaboration


Dive into the Kengo Tsuda's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Takanori Kigawa

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Peter Güntert

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mari Takahashi

Frankfurt Institute for Advanced Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fahu He

Frankfurt Institute for Advanced Studies

View shared research outputs
Researchain Logo
Decentralizing Knowledge