Network


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

Hotspot


Dive into the research topics where Yoko Hayashi-Takanaka is active.

Publication


Featured researches published by Yoko Hayashi-Takanaka.


Nature | 2011

Crystal structure of the human centromeric nucleosome containing CENP-A

Hiroaki Tachiwana; Wataru Kagawa; Tatsuya Shiga; Akihisa Osakabe; Yuta Miya; Kengo Saito; Yoko Hayashi-Takanaka; Takashi Oda; Mamoru Sato; Sam-Yong Park; Hiroshi Kimura; Hitoshi Kurumizaka

In eukaryotes, accurate chromosome segregation during mitosis and meiosis is coordinated by kinetochores, which are unique chromosomal sites for microtubule attachment. Centromeres specify the kinetochore formation sites on individual chromosomes, and are epigenetically marked by the assembly of nucleosomes containing the centromere-specific histone H3 variant, CENP-A. Although the underlying mechanism is unclear, centromere inheritance is probably dictated by the architecture of the centromeric nucleosome. Here we report the crystal structure of the human centromeric nucleosome containing CENP-A and its cognate α-satellite DNA derivative (147 base pairs). In the human CENP-A nucleosome, the DNA is wrapped around the histone octamer, consisting of two each of histones H2A, H2B, H4 and CENP-A, in a left-handed orientation. However, unlike the canonical H3 nucleosome, only the central 121 base pairs of the DNA are visible. The thirteen base pairs from both ends of the DNA are invisible in the crystal structure, and the αN helix of CENP-A is shorter than that of H3, which is known to be important for the orientation of the DNA ends in the canonical H3 nucleosome. A structural comparison of the CENP-A and H3 nucleosomes revealed that CENP-A contains two extra amino acid residues (Arg 80 and Gly 81) in the loop 1 region, which is completely exposed to the solvent. Mutations of the CENP-A loop 1 residues reduced CENP-A retention at the centromeres in human cells. Therefore, the CENP-A loop 1 may function in stabilizing the centromeric chromatin containing CENP-A, possibly by providing a binding site for trans-acting factors. The structure provides the first atomic-resolution picture of the centromere-specific nucleosome.


Molecular Cell | 2012

Independence of Repressive Histone Marks and Chromatin Compaction during Senescent Heterochromatic Layer Formation

Tamir Chandra; Kristina Kirschner; Jean Yves Thuret; Benjamin D. Pope; Tyrone Ryba; Scott Newman; Kashif Ahmed; Shamith Samarajiwa; Rafik Salama; Thomas Carroll; Rory Stark; Rekin’s Janky; Masako Narita; Lixiang Xue; Agustin Chicas; Sabrina Nũnez; Ralf Janknecht; Yoko Hayashi-Takanaka; Michael D. Wilson; Aileen Marshall; Duncan T. Odom; M. Madan Babu; David P. Bazett-Jones; Simon Tavaré; Paul A.W. Edwards; Scott W. Lowe; Hiroshi Kimura; David M. Gilbert; Masashi Narita

The expansion of repressive epigenetic marks has been implicated in heterochromatin formation during embryonic development, but the general applicability of this mechanism is unclear. Here we show that nuclear rearrangement of repressive histone marks H3K9me3 and H3K27me3 into nonoverlapping structural layers characterizes senescence-associated heterochromatic foci (SAHF) formation in human fibroblasts. However, the global landscape of these repressive marks remains unchanged upon SAHF formation, suggesting that in somatic cells, heterochromatin can be formed through the spatial repositioning of pre-existing repressively marked histones. This model is reinforced by the correlation of presenescent replication timing with both the subsequent layered structure of SAHFs and the global landscape of the repressive marks, allowing us to integrate microscopic and genomic information. Furthermore, modulation of SAHF structure does not affect the occupancy of these repressive marks, nor vice versa. These experiments reveal that high-order heterochromatin formation and epigenetic remodeling of the genome can be discrete events.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Structural basis of instability of the nucleosome containing a testis-specific histone variant, human H3T

Hiroaki Tachiwana; Wataru Kagawa; Akihisa Osakabe; Koichiro Kawaguchi; Tatsuya Shiga; Yoko Hayashi-Takanaka; Hiroshi Kimura; Hitoshi Kurumizaka

A histone H3 variant, H3T, is highly expressed in the testis, suggesting that it may play an important role in the chromatin reorganization required for meiosis and/or spermatogenesis. In the present study, we found that the nucleosome containing human H3T is significantly unstable both in vitro and in vivo, as compared to the conventional nucleosome containing H3.1. The crystal structure of the H3T nucleosome revealed structural differences in the H3T regions on both ends of the central α2 helix, as compared to those of H3.1. The H3T-specific residues (Met71 and Val111) are the source of the structural differences observed between H3T and H3.1. A mutational analysis revealed that these residues are responsible for the reduced stability of the H3T-containing nucleosome. These physical and structural properties of the H3T-containing nucleosome may provide the basis of chromatin reorganization during spermatogenesis.


Nucleic Acids Research | 2011

Tracking epigenetic histone modifications in single cells using Fab-based live endogenous modification labeling

Yoko Hayashi-Takanaka; Kazuo Yamagata; Teruhiko Wakayama; Timothy J. Stasevich; Takashi Kainuma; Toshiki Tsurimoto; Makoto Tachibana; Yoichi Shinkai; Hitoshi Kurumizaka; Naohito Nozaki; Hiroshi Kimura

Histone modifications play an important role in epigenetic gene regulation and genome integrity. It remains largely unknown, however, how these modifications dynamically change in individual cells. By using fluorescently labeled specific antigen binding fragments (Fabs), we have developed a general method to monitor the distribution and global level of endogenous histone H3 lysine modifications in living cells without disturbing cell growth and embryo development. Fabs produce distinct nuclear patterns that are characteristic of their target modifications. H3K27 trimethylation-specific Fabs, for example, are concentrated on inactive X chromosomes. As Fabs bind their targets transiently, the ratio of bound and free molecules depends on the target concentration, allowing us to measure changes in global modification levels. High-affinity Fabs are suitable for mouse embryo imaging, so we have used them to monitor H3K9 and H3K27 acetylation levels in mouse preimplantation embryos produced by in vitro fertilization and somatic cell nuclear transfer. The data suggest that a high level of H3K27 acetylation is important for normal embryo development. As Fab-based live endogenous modification labeling (FabLEM) is broadly useful for visualizing any modification, it should be a powerful tool for studying cell signaling and diagnosis in the future.


Nature | 2014

Regulation of RNA polymerase II activation by histone acetylation in single living cells

Timothy J. Stasevich; Yoko Hayashi-Takanaka; Yuko Sato; Kazumitsu Maehara; Yasuyuki Ohkawa; Kumiko Sakata-Sogawa; Makio Tokunaga; Takahiro Nagase; Naohito Nozaki; James G. McNally; Hiroshi Kimura

In eukaryotic cells, post-translational histone modifications have an important role in gene regulation. Starting with early work on histone acetylation, a variety of residue-specific modifications have now been linked to RNA polymerase II (RNAP2) activity, but it remains unclear if these markers are active regulators of transcription or just passive byproducts. This is because studies have traditionally relied on fixed cell populations, meaning temporal resolution is limited to minutes at best, and correlated factors may not actually be present in the same cell at the same time. Complementary approaches are therefore needed to probe the dynamic interplay of histone modifications and RNAP2 with higher temporal resolution in single living cells. Here we address this problem by developing a system to track residue-specific histone modifications and RNAP2 phosphorylation in living cells by fluorescence microscopy. This increases temporal resolution to the tens-of-seconds range. Our single-cell analysis reveals histone H3 lysine-27 acetylation at a gene locus can alter downstream transcription kinetics by as much as 50%, affecting two temporally separate events. First acetylation enhances the search kinetics of transcriptional activators, and later the acetylation accelerates the transition of RNAP2 from initiation to elongation. Signatures of the latter can be found genome-wide using chromatin immunoprecipitation followed by sequencing. We argue that this regulation leads to a robust and potentially tunable transcriptional response.


Journal of Cell Biology | 2009

Visualizing histone modifications in living cells: spatiotemporal dynamics of H3 phosphorylation during interphase.

Yoko Hayashi-Takanaka; Kazuo Yamagata; Naohito Nozaki; Hiroshi Kimura

A new technique illuminates differential H3S10 phosphorylation dynamics in normal and cancer cells; spatial and temporal regulation of this process by aurora B kinase is required for accurate chromosome segregation.


Scientific Reports | 2013

Genetically encoded system to track histone modification in vivo

Yuko Sato; Masanori Mukai; Jun Ueda; Michiko Muraki; Timothy J. Stasevich; Naoki Horikoshi; Tomoya Kujirai; Hiroaki Kita; Taisuke Kimura; Seiji Hira; Yasushi Okada; Yoko Hayashi-Takanaka; Chikashi Obuse; Hitoshi Kurumizaka; Atsuo Kawahara; Kazuo Yamagata; Naohito Nozaki; Hiroshi Kimura

Post-translational histone modifications play key roles in gene regulation, development, and differentiation, but their dynamics in living organisms remain almost completely unknown. To address this problem, we developed a genetically encoded system for tracking histone modifications by generating fluorescent modification-specific intracellular antibodies (mintbodies) that can be expressed in vivo. To demonstrate, an H3 lysine 9 acetylation specific mintbody (H3K9ac-mintbody) was engineered and stably expressed in human cells. In good agreement with the localization of its target acetylation, H3K9ac-mintbody was enriched in euchromatin, and its kinetics measurably changed upon treatment with a histone deacetylase inhibitor. We also generated transgenic fruit fly and zebrafish stably expressing H3K9ac-mintbody for in vivo tracking. Dramatic changes in H3K9ac-mintbody localization during Drosophila embryogenesis could highlight enhanced acetylation at the start of zygotic transcription around mitotic cycle 7. Together, this work demonstrates the broad potential of mintbody and lays the foundation for epigenetic analysis in vivo.


Acta Crystallographica Section D-biological Crystallography | 2013

Structural polymorphism in the L1 loop regions of human H2A.Z.1 and H2A.Z.2

Naoki Horikoshi; Koichi Sato; Keisuke Shimada; Yasuhiro Arimura; Akihisa Osakabe; Hiroaki Tachiwana; Yoko Hayashi-Takanaka; Wakana Iwasaki; Wataru Kagawa; Masahiko Harata; Hiroshi Kimura; Hitoshi Kurumizaka

The crystal structures of human nucleosomes containing H2A.Z.1 and H2A.Z.2 have been determined. Structural polymorphisms were found in the L1 loop regions of H2A.Z.1 and H2A.Z.2 in the nucleosomes that are likely to be caused by their flexible nature.


Histochemistry and Cell Biology | 2015

Visualizing posttranslational and epigenetic modifications of endogenous proteins in vivo

Hiroshi Kimura; Yoko Hayashi-Takanaka; Timothy J. Stasevich; Yuko Sato

Protein localization and dynamics can now be visualized in living cells using the fluorescent protein fusion technique, but it is still difficult to selectively detect molecules with a specific function. As a posttranslational protein modification is often associated with a specific function, marking specifically modified protein molecules in living cells is a way to track an important fraction of protein. In the nucleus, histones are subjected to a variety of modifications such as acetylation and methylation that are associated with epigenetic gene regulation. RNA polymerase II, an enzyme that transcribes genes, is also differentially phosphorylated during the initiation and elongation of transcription. To understand the mechanism of gene regulation in vivo, we have developed methods to track histone and RNA polymerase II modifications using probes derived from modification-specific monoclonal antibodies. In Fab-based live endogenous modification labeling (FabLEM), fluorescently labeled antigen-binding fragments (Fabs) are loaded into cells. Fabs bind to target modifications in the nucleus with a binding time of a second to tens of seconds, and so the modification can be tracked without disturbing cell function. For tracking over longer periods of time or in living animals, we have also developed a genetically encoded system to express a modification-specific intracellular antibody (mintbody). Transgenic fruit fly and zebrafish that express histone H3 Lys9 acetylation-specific mintbody developed normally and remain fertile, suggesting that visualizing histone modifications in any tissue in live animals has become possible. These live cell modification tracking techniques will facilitate future studies on epigenetic regulation related to development, differentiation, and disease. Moreover, these techniques can be applied to any other protein modification, opening up new avenues in broad areas in biology and medicine.


Current Opinion in Cell Biology | 2010

Visualization of DNA methylation and histone modifications in living cells.

Hiroshi Kimura; Yoko Hayashi-Takanaka; Kazuo Yamagata

DNA methylation and histone modifications play important roles in genome function, including epigenetic gene regulation. These modifications undergo drastic changes when nuclei are reprogrammed during development and differentiation. Recent studies have enabled the detection of the dynamics of modifications in living cultured cells and mouse preimplantation embryos. DNA methylation was visualized using the methyl-CpG-binding domain of the human MBD1 protein. The level and distribution of histone modifications can be monitored by two different methods. One approach uses fluorescence/Förster resonance energy transfer (FRET)-based sensors and another uses fluorescently labeled antigen binding fragments of specific antibodies. These visualization techniques will facilitate future studies on epigenetic regulation related to development, differentiation, and disease.

Collaboration


Dive into the Yoko Hayashi-Takanaka's collaboration.

Top Co-Authors

Avatar

Hiroshi Kimura

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yuko Sato

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge