Network


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

Hotspot


Dive into the research topics where Yuji Chikashige is active.

Publication


Featured researches published by Yuji Chikashige.


Nature | 2006

Selective elimination of messenger RNA prevents an incidence of untimely meiosis.

Yuriko Harigaya; Hirotsugu Tanaka; Soichiro Yamanaka; Kayoko Tanaka; Yoshinori Watanabe; Chihiro Tsutsumi; Yuji Chikashige; Yasushi Hiraoka; Akira Yamashita; Masayuki Yamamoto

Much remains unknown about the molecular regulation of meiosis. Here we show that meiosis-specific transcripts are selectively removed if expressed during vegetative growth in fission yeast. These messenger RNAs contain a cis-acting region—which we call the DSR—that confers this removal via binding to a YTH-family protein Mmi1. Loss of Mmi1 function severely impairs cell growth owing to the untimely expression of meiotic transcripts. Microarray analysis reveals that at least a dozen such meiosis-specific transcripts are eliminated by the DSR–Mmi1 system. Mmi1 remains in the form of multiple nuclear foci during vegetative growth. At meiotic prophase these foci precipitate to a single focus, which coincides with the dot formed by the master meiosis-regulator Mei2. A meiotic arrest due to the loss of the Mei2 dot is released by a reduction in Mmi1 activity. We propose that Mei2 turns off the DSR–Mmi1 system by sequestering Mmi1 to the dot and thereby secures stable expression of meiosis-specific transcripts.


Cell | 1989

Composite motifs and repeat symmetry in S. pombe centromeres: Direct analysis by integration of Notl restriction sites

Yuji Chikashige; Noriyuki Kinoshita; Yukinobu Nakaseko; Tomohiro Matsumoto; Shin Murakami; Osami Niwa; Mitsuhiro Yanagida

S. pombe centromeres are large and complex. We introduced a method that enables us to characterize directly centromere DNAs. Genomic DNA fragments containing cen1, cen2, or cen3, respectively, are made by cleaving NotI sites integrated on target sites and are partially restricted for long-range mapping in PFG electrophoresis. The 40 kb long cen1 consists of two inverted approximately 10 kb motifs, each containing centromeric elements dg and dh, flanked by a central region. In cen2, three motifs are arranged in inverted and direct orientations with flanking domains, making up the approximately 70 kb long repetitious region. In cen3, approximately 15 copies of dg-dh constitute a region longer than 100 kb. A set of inverted motifs with an approximately 15 kb central region might be a prototype for the S. pombe centromeres. The motifs appear to play a role in chromosome stability and segregation. Their action may be additive, and the mutual directions of dg and dh inside a motif may not be essential for function.


The EMBO Journal | 1997

Meiotic nuclear reorganization: switching the position of centromeres and telomeres in the fission yeast Schizosaccharomyces pombe

Yuji Chikashige; Da-Qiao Ding; Yoshiyuki Imai; Masayuki Yamamoto; Tokuko Haraguchi; Yasushi Hiraoka

In fission yeast meiotic prophase, telomeres are clustered near the spindle pole body (SPB; a centrosome‐equivalent structure in fungi) and take the leading position in chromosome movement, while centromeres are separated from the SPB. This telomere position contrasts with mitotic nuclear organization, in which centromeres remain clustered near the SPB and lead chromosome movement. Thus, nuclear reorganization switching the position of centromeres and telomeres must take place upon entering meiosis. In this report, we analyze the nuclear location of centromeres and telomeres in genetically well‐characterized meiotic mutant strains. An intermediate structure for telomere‐centromere switching was observed in haploid cells induced to undergo meiosis by synthetic mating pheromone; fluorescence in situ hybridization revealed that in these cells, both telomeres and centromeres were clustered near the SPB. Further analyses in a series of mutants showed that telomere‐centromere switching takes place in two steps; first, association of telomeres with the SPB and, second, dissociation of centromeres from the SPB. The first step can take place in the haploid state in response to mating pheromone, but the second step does not take place in haploid cells and probably depends on conjugation‐related events. In addition, a linear minichromosome was also co‐localized with authentic telomeres instead of centromeres, suggesting that telomere clustering plays a role in organizing chromosomes within a meiotic prophase nucleus.


Current Biology | 2001

Telomere binding of the Rap1 protein is required for meiosis in fission yeast

Yuji Chikashige; Yasushi Hiraoka

Telomeres are essential for chromosome integrity, protecting the ends of eukaryotic linear chromosomes during cell proliferation. Telomeres also function in meiosis; a characteristic clustering of telomeres beneath the nuclear membrane is observed during meiotic prophase in many organisms from yeasts to plants and humans, and the role of the telomeres in meiotic pairing and the recombination of homologous chromosomes has been demonstrated in the fission yeast Schizosaccharomyces pombe and in the budding yeast Saccharomyces cerevisiae. Here we report that S. pombe Rap1 is a telomeric protein essential for meiosis. While Rap1 is conserved in budding yeast and humans, schemes for telomere binding vary among species: human RAP1 binds to the telomere through interaction with the telomere binding protein TRF2; S. cerevisiae Rap1, however, binds telomeric DNA directly, and no orthologs of TRF proteins have been identified in this organism. In S. pombe, unlike in S. cerevisiae, an ortholog of human TRF has been identified. This ortholog, Taz1, binds directly to telomere repeats [18] and is necessary for telomere clustering in meiotic prophase. Our results demonstrate that S. pombe Rap1 binds to telomeres through interaction with Taz1, similar to human Rap1-TRF2, and that Taz1-mediated telomere localization of Rap1 is necessary for telomere clustering and for the successful completion of meiosis. Moreover, in taz1-disrupted cells, molecular fusion of Rap1 with the Taz1 DNA binding domain recovers telomere clustering and largely complements defects in meiosis, indicating that telomere localization of Rap1 is a key requirement for meiosis.


Genes to Cells | 2000

Large-scale screening of intracellular protein localization in living fission yeast cells by the use of a GFP-fusion genomic DNA library

Da-Qiao Ding; Yuki Tomita; Ayumu Yamamoto; Yuji Chikashige; Tokuko Haraguchi; Yasushi Hiraoka

Intracellular localization is an important part of the characterization of a gene product. In an attempt to search for genes based on the intracellular localization of their products, we constructed a green fluorescent protein (GFP)‐fusion genomic DNA library of S. pombe.


Science | 2008

Heterochromatin integrity affects chromosome reorganization after centromere dysfunction.

Kojiro Ishii; Yuki Ogiyama; Yuji Chikashige; Saeko Soejima; Fumie Masuda; Tatsuyuki Kakuma; Yasushi Hiraoka; Kohta Takahashi

The centromere is essential for the inheritance of genetic information on eukaryotic chromosomes. Epigenetic regulation of centromere identity has been implicated in genome stability, karyotype evolution, and speciation. However, little is known regarding the manner in which centromere dysfunction affects the chromosomal architectures. Here we show that in the fission yeast Schizosaccharomyces pombe, the conditional deletion of the centromere produces survivors that carry either a neocentromere-acquired chromosome at the subtelomeric region or an acentric chromosome rescued by intertelomere fusion with either of the remaining chromosomes. The ratio of neocentromere formation to telomere fusion is considerably decreased by the inactivation of genes involved in RNA interference–dependent heterochromatin formation. By affecting the modes of chromosomal reorganization, the genomic distribution of heterochromatin may influence the fate of karyotype evolution.


The EMBO Journal | 1989

Characterization of Schizosaccharomyces pombe minichromosome deletion derivatives and a functional allocation of their centromere.

Osami Niwa; Tomohiro Matsumoto; Yuji Chikashige; Mitsuhiro Yanagida

A 530 kb long Schizosaccharomyces pombe linear minichromosome, Ch16, containing a centric region of chromosome III, has previously been made. In the present study, we constructed a number of deletions in the right and/or left arms of Ch16, and compared their structure and behaviour with Ch16. The functional centromere, cen3, is allocated within a 120 kb long region which is covered by the shortest derivative, Ch10, and is comprised mostly of centromeric repeating sequences. The shortest minichromosome is stable in mitosis and the copy number control is apparently precise. In monosomic meiosis it segregates normally. In disomic meioses, however, the frequency of non‐disjunction is very high, suggesting that it may not form a pair. The mitotic loss rate of one of the left‐arm deletions, ChR32, which lacks a part of the centromeric repeating sequence, is the highest of all the deletions. This deletion also exhibits the highest precocious sister chromatid separation in meiosis I, suggesting that sister chromatid association might become weakened in ChR32. Our results indicate that the proper meiotic segregation of S.pombe minichromosomes is dependent upon the formation of a bivalent. S.pombe may not have the ‘distributive segregation’ found with Saccharomyces cerevisiae minichromosomes.


Molecular Genetics and Genomics | 1997

A NOVEL FISSION YEAST GENE, KMS1+, IS REQUIRED FOR THE FORMATION OF MEIOTIC PROPHASE-SPECIFIC NUCLEAR ARCHITECTURE

Mizuki Shimanuki; Futaba Miki; Da-Qiao Ding; Yuji Chikashige; Yasushi Hiraoka; Tetsuya Horio; Osami Niwa

Abstract In the meiotic prophase nucleus of the fission yeast Schizosaccharomyces pombe, chromosomes are arranged in an oriented manner: telomeres cluster in close proximity to the spindle pole body (SPB), while centromeres form another cluster at some distance from the SPB. We have isolated a mutant, kms1, in which the structure of the meiotic prophase nucleus appears to be distorted. Using specific probes to localize the SPB and telomeres, multiple signals were observed in the mutant nuclei, in contrast to the case in wild-type. Genetic analysis showed that in the mutant, meiotic recombination frequency was reduced to about one-quarter of the wild-type level and meiotic segregation was impaired. This phenotype strongly suggests that the telomere-led rearrangement of chromosomal distribution that normally occurs in the fission yeast meiotic nucleus is an important prerequisite for the efficient pairing of homologous chromosomes. The kms1 mutant was also impaired in karyogamy, suggesting that the kms1+ gene is involved in SPB function. However, the kms1+ gene is dispensable for mitotic growth. The predicted amino acid sequence of the gene product shows no significant similarity to known proteins.


Chromosoma | 2007

Another way to move chromosomes

Yuji Chikashige; Tokuko Haraguchi; Yasushi Hiraoka

A typical way of moving chromosomes is exemplified by mitotic segregation, in which the centromere is directly captured by spindle microtubules. In this study, we highlight another way of moving chromosomes remotely from outside the nucleus, which involves SUN and KASH domain nuclear envelope proteins. SUN and KASH domain protein families are known to connect the nucleus to cytoskeletal networks and play a role in migration and positioning of the nucleus. Recent studies in the fission yeast Schizossacharomyces pombe demonstrated an additional role for the SUN–KASH protein complex in chromosome movements. During meiotic prophase, telomeres are moved to rearrange chromosomes within the nucleus. The SUN–KASH protein complex located in the nuclear envelope is involved in this process. Telomeres are connected to the SUN protein on the nucleoplasmic side, and the dynein motor complex binds to the KASH protein on the cytoplasmic side. Telomeres are then moved along the nuclear envelope using cytoplasmic microtubules. These findings illustrate a general mechanism for transmitting a cytoskeletal driving force to chromosomes across the nuclear envelope.


Journal of Cell Biology | 2009

Membrane proteins Bqt3 and -4 anchor telomeres to the nuclear envelope to ensure chromosomal bouquet formation

Yuji Chikashige; Miho Yamane; Kasumi Okamasa; Chihiro Tsutsumi; Tomoko Kojidani; Mamiko Sato; Tokuko Haraguchi; Yasushi Hiraoka

A screen identifies two more bouquet proteins required for meiotic telomere clustering: Bqt4 anchors the telomeres, whereas Bqt3 protects Bqt4 from degradation.

Collaboration


Dive into the Yuji Chikashige's collaboration.

Top Co-Authors

Avatar

Yasushi Hiraoka

National Institute of Information and Communications Technology

View shared research outputs
Top Co-Authors

Avatar

Tokuko Haraguchi

National Institute of Information and Communications Technology

View shared research outputs
Top Co-Authors

Avatar

Chihiro Tsutsumi

National Institute of Information and Communications Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mitsuhiro Yanagida

Okinawa Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Da-Qiao Ding

National Institute of Information and Communications Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kasumi Okamasa

National Institute of Information and Communications Technology

View shared research outputs
Top Co-Authors

Avatar

Miho Yamane

National Institute of Information and Communications Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge