Network


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

Hotspot


Dive into the research topics where Toshiya Nishimura is active.

Publication


Featured researches published by Toshiya Nishimura.


Science | 2010

Identification of Germline Stem Cells in the Ovary of the Teleost Medaka

Shuhei Nakamura; Kayo Kobayashi; Toshiya Nishimura; Shin-ichi Higashijima; Minoru Tanaka

Keeping Egg Production Going Whether oogenesis ceases around birth in vertebrate ovaries has been a topic of long-standing interest and considerable debate. Nakamura et al. (p. 1561, published online 20 May) now identify germline stem cells in the ovary of the teleost fish medaka. The stem cells are found as clusters of germ cells (termed germinal cradles) in a cord-like structure that expresses sox9, a gene critical for testis formation in mammals. The cords are buried within the ovary within the germinal epithelium. This work in fish shows that there can indeed be continuing egg production from vertebrate germline stem cells. Ongoing follicle production is maintained by stem cells in an adult fish ovary. Germline stem cells continually produce sperm in vertebrate testes, whereas there is no direct evidence showing that germline stem cells are present in adult vertebrate ovaries. By using transgenic methods and clonal analysis, we identified germline stem cells that supported oogenesis and the production of offspring in the ovaries of adult medaka fish. Early-stage germ cells were localized in clusters along interwoven threadlike cords of sox9b-expressing somatic cells (termed germinal cradles) where the germ cells developed. Germline stem cells gave rise to germ cells that divided to produce cysts, which then underwent cell death or separated to form follicles. Our results provide insight into the germline stem cell biology of medaka and provide a model system for studying vertebrate stem cell niches.


PLOS ONE | 2012

Analysis of Medaka sox9 Orthologue Reveals a Conserved Role in Germ Cell Maintenance

Shuhei Nakamura; Ikuko Watakabe; Toshiya Nishimura; Atsushi Toyoda; Yoshihito Taniguchi; Minoru Tanaka

The sex determining gene is divergent among different animal species. However, sox9 is up-regulated in the male gonads in a number of species in which it is the essential regulator of testis determination. It is therefore often discussed that the sex determining gene-sox9 axis functions in several vertebrates. In our current study, we show that sox9b in the medaka (Oryzias latipes) is one of the orthologues of mammalian Sox9 at syntenic and expression levels. Medaka sox9b affects the organization of extracellular matrices, which represents a conserved role of sox9, but does not directly regulate testis determination. We made this determination via gene expression and phenotype analyses of medaka with different copy numbers of sox9b. Sox9b is involved in promoting cellular associations and is indispensible for the proper proliferation and survival of germ cells in both female and male medaka gonads. Medaka mutants that lack sox9b function exhibit a seemingly paradoxical phenotype of sex reversal to male. This is explained by a reduction in the germ cell number associated with aberrant extracellular matrices. Together with its identified roles in other vertebrate gonads, a testis-determining role for Sox9 in mammals is likely to have been neofunctionalized and appended to its conserved role in germ cell maintenance.


Development | 2012

Hyperproliferation of mitotically active germ cells due to defective anti-Müllerian hormone signaling mediates sex reversal in medaka

Shuhei Nakamura; Ikuko Watakabe; Toshiya Nishimura; Jean-Yves Picard; Atsushi Toyoda; Yoshihito Taniguchi; Nathalie di Clemente; Minoru Tanaka

The function of AMH (Anti-Müllerian hormone), a phylogenetically ancient member of the TGFβ family of proteins, in lower vertebrates is largely unknown. Previously, we have shown that the gene encoding the type II anti-Müllerian hormone receptor, amhrII, is responsible for excessive germ cell proliferation and male-to-female sex reversal in the medaka hotei mutant. In this study, functional analyses in cultured cells and of other amhrII mutant alleles indicate that lack of AMH signaling causes the hotei phenotype. BrdU incorporation experiments identified the existence of both quiescent and mitotically active germ cells among the self-renewing, type I population of germ cells in the developing gonad. AMH signaling acts in supporting cells to promote the proliferation of mitotically active germ cells but does not trigger quiescent germ cells to proliferate in the developing gonad. Furthermore, we show that the male-to-female sex reversal phenotype in hotei mutants is not a direct consequence of AMH signaling in supporting cells, but is instead mediated by germ cells. Our data demonstrate that interfollicular AMH signaling regulates proliferation at a specific stage of germ cell development, and that this regulation is crucial for the proper manifestation of gonadal sex directed by sex determination genes.


Science | 2015

foxl3 is a germ cell–intrinsic factor involved in sperm-egg fate decision in medaka

Toshiya Nishimura; Tetsuya Sato; Yasuhiro Yamamoto; Ikuko Watakabe; Yasuyuki Ohkawa; Mikita Suyama; Satoru Kobayashi; Minoru Tanaka

How germ cells become sperm or egg During vertebrate development, germ cells switch from a sexually indifferent to a committed state for either egg or sperm. Signals from somatic gonadal cells are generally thought to influence the sexual differentiation of germ cells. However, Nishimura et al. demonstrate that germ cell–intrinsic sex determination cues are at play in the teleost fish medaka. The forkhead box transcriptional factor foxl3 represses the initiation of spermatogenesis. In the absence of foxl3 function, females develop ovaries filled with functional sperm. Thus, the male gonad environment is not required for spermatogenesis. Science, this issue p. 328 The switch mechanism involved in germ cell fate determination is identified in medaka fish. Sex determination is an essential step in the commitment of a germ cell to a sperm or egg. However, the intrinsic factors that determine the sexual fate of vertebrate germ cells are unknown. Here, we show that foxl3, which is expressed in germ cells but not somatic cells in the gonad, is involved in sperm-egg fate decision in medaka fish. Adult XX medaka with disrupted foxl3 developed functional sperm in the expanded germinal epithelium of a histologically functional ovary. In chimeric medaka, mutant germ cells initiated spermatogenesis in female wild-type gonad. These results indicate that a germ cell–intrinsic cue for the sperm-egg fate decision is present in medaka and that spermatogenesis can proceed in a female gonadal environment.


Sexual Development | 2014

Gonadal Development in Fish

Toshiya Nishimura; Minoru Tanaka

Vertebrate reproduction depends on the function of 2 distinct gametes, sperm and eggs, which develop in 2 different organs, the testis and the ovary. Testes and ovaries are composed of germ cells, supporting cells and interstitial cells. In this review, we describe the origin and the fate of these cell lineages and how they interact with each other to form sexually dimorphic reproductive organs in medaka. We delineate how the temporally different association and establishment of these lineages contribute to a variety of seemingly different sex differentiation processes among teleost fish. Thus, teleosts represent an intriguing group in which to study the fundamental processes of gonadal development through comparing conserved and unique mechanisms.


Nature Communications | 2017

Dynamic plasticity in phototransduction regulates seasonal changes in color perception

Tsuyoshi Shimmura; Tomoya Nakayama; Ai Shinomiya; Shoji Fukamachi; Masaki Yasugi; Eiji Watanabe; Takayuki Shimo; Takumi Senga; Toshiya Nishimura; Minoru Tanaka; Yasuhiro Kamei; Kiyoshi Naruse; Takashi Yoshimura

To cope with seasonal changes in the environment, organisms adapt their physiology and behavior. Although color perception varies among seasons, the underlying molecular basis and its physiological significance remain unclear. Here we show that dynamic plasticity in phototransduction regulates seasonal changes in color perception in medaka fish. Medaka are active and exhibit clear phototaxis in conditions simulating summer, but remain at the bottom of the tank and fail to exhibit phototaxis in conditions simulating winter. Mate preference tests using virtual fish created with computer graphics demonstrate that medaka are more attracted to orange-red-colored model fish in summer than in winter. Transcriptome analysis of the eye reveals dynamic seasonal changes in the expression of genes encoding photopigments and their downstream pathways. Behavioral analysis of photopigment-null fish shows significant differences from wild type, suggesting that plasticity in color perception is crucial for the emergence of seasonally regulated behaviors.Animal coloration and behavior can change seasonally, but it is unclear if visual sensitivity to color shifts as well. Here, Shimmura et al. show that medaka undergo seasonal behavioral change accompanied by altered expression of opsin genes, resulting in reduced visual sensitivity to mates during winter-like conditions.


Development | 2014

Analysis of a novel gene, Sdgc , reveals sex chromosome-dependent differences of medaka germ cells prior to gonad formation

Toshiya Nishimura; Amaury Herpin; Tetsuaki Kimura; Ikuyo Hara; Toshihiro Kawasaki; Shuhei Nakamura; Yasuhiro Yamamoto; Taro Saito; Jun Yoshimura; Shinichi Morishita; Tatsuya Tsukahara; Satoru Kobayashi; Kiyoshi Naruse; Shuji Shigenobu; Noriyoshi Sakai; Manfred Schartl; Minoru Tanaka

In vertebrates that have been examined to date, the sexual identity of germ cells is determined by the sex of gonadal somatic cells. In the teleost fish medaka, a sex-determination gene on the Y chromosome, DMY/dmrt1bY, is expressed in gonadal somatic cells and regulates the sexual identity of germ cells. Here, we report a novel mechanism by which sex chromosomes cell-autonomously confer sexually different characters upon germ cells prior to gonad formation in a genetically sex-determined species. We have identified a novel gene, Sdgc (sex chromosome-dependent differential expression in germ cells), whose transcripts are highly enriched in early XY germ cells. Chimeric analysis revealed that sexually different expression of Sdgc is controlled in a germ cell-autonomous manner by the number of Y chromosomes. Unexpectedly, DMY/dmrt1bY was expressed in germ cells prior to gonad formation, but knockdown and overexpression of DMY/dmrt1bY did not affect Sdgc expression. We also found that XX and XY germ cells isolated before the onset of DMY/dmrt1bY expression in gonadal somatic cells behaved differently in vitro and were affected by Sdgc. Sdgc maps close to the sex-determination locus, and recombination around the two loci appears to be repressed. Our results provide important insights into the acquisition and plasticity of sexual differences at the cellular level even prior to the developmental stage of sex determination.


Biology of Reproduction | 2016

The Mechanism of Germline Sex Determination in Vertebrates

Toshiya Nishimura; Minoru Tanaka

ABSTRACT Germ cells are the common cells of origin for the two different types of gametes, sperm and eggs. In vertebrates so far examined, the sex of germ cells is determined by gonadal somatic cells. However, influenced by the somatic cells, how germ cells adopt their sexual fates by intrinsic factors has long been unclear in vertebrates. We recently identified forkhead box L3 (FOXL3) as a germ cell-intrinsic factor involved in the sperm–egg fate decision in the teleost fish medaka (Oryzias latipes). On the basis of the results obtained by the analysis of foxl3/FOXL3 expression and loss-of-function mutants, we review when and how germ cell sex is regulated non-cell-autonomously and cell-autonomously. We then discuss the fact that the germline sex determination pathway is genetically distinct from other essential gametogenic pathways such as meiotic entry and the establishment of germline stem cells. Another extraordinary finding in the foxl3 mutant is that functional sperm can be produced in the ovary, which provides a new notion that gametogenesis can proceed regardless of the sex of the surrounding somatic cells once the sexual identity of germ cells is established in medaka.


Sexual Development | 2016

A Structurally and Functionally Common Unit in Testes and Ovaries of Medaka (Oryzias latipes), a Teleost Fish

Toshiya Nishimura; Shuhei Nakamura; Minoru Tanaka

Testes and ovaries are structurally and functionally different organs, but they originate from the same gonadal primordium. Here, we propose how a tissue composed of germ cells and sox9b-expressing cells underlain by a basement membrane develops a functionally common unit that reserves sexually indifferent or unfixed germline stem cells in both the ovaries and testes of adult medaka. During testicular development, the unit expands and gives rise to lobules where germline stem cells continuously produce a tremendous amount of sperm, while the unit in the female becomes germinal cradles that represent niche areas for neo-oogenesis. A recent finding in female foxl3 mutants demonstrates that the unit can also serve as the niche for germline stem cells continuously producing sperm in the ovary. We hypothesize that this unit in medaka might correspond to the medullary cords of the gonadal primordium in amniotes and that fragmentation of the medullary cords during ovarian development may be related to the absence of germline stem cells.


PLOS Genetics | 2018

Germ cells in the teleost fish medaka have an inherent feminizing effect

Toshiya Nishimura; Kazuki Yamada; Chika Fujimori; Mariko Kikuchi; Toshihiro Kawasaki; Kellee R. Siegfried; Noriyoshi Sakai; Minoru Tanaka

Germ cells give rise to eggs or sperm. However, recent analyses in medaka (Oryzias latipes) showed that germ cells are also important for feminization of gonads, although this novel role of germ cells has not been characterized in detail. Here, we show that the feminizing effect is inherent to germ cells and is not affected by gametogenic stages or the sexual fate of germ cells. Three medaka mutants were generated to demonstrate this effect: figlα mutants, in which follicle formation is disrupted; meioC mutants, in which germ cells are unable to commit to gametogenesis and meiosis; and dazl mutants, in which germ cells do not develop into gonocytes. All these different stages of germ cells in XX mutants have an ability to feminize the gonads, resulting in the formation of gonads with ovarian structures. In addition to normal ovarian development, we also suggest that the increased number of gonocytes is sufficient for male to female sex reversal in XY medaka. These results may genetically demonstrate that the mechanism underlying the feminizing effect of germ cells is activated before the sexual fate decision of germ cells and meiosis, probably by the time of gonocyte formation in medaka. Author summary Germ cells are the only cells that can transfer genetic materials to the next generation via the sperm or egg. However, recent analyses in teleosts revealed another essential role of germ cells: feminizing the gonads. In our study, medaka mutants in which gametogenesis was blocked at specific stages provides the novel view that the feminizing effect of germ cells occurs in parallel with other reproductive elements, such as meiosis, the sexual fate decision of germ cells, and gametogenesis. Germ cells in medaka may have a potential to feminize gonads at the moment they have developed.

Collaboration


Dive into the Toshiya Nishimura's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Atsushi Toyoda

National Institute of Genetics

View shared research outputs
Top Co-Authors

Avatar

Daisuke Saito

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Noriyoshi Sakai

National Institute of Genetics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toshihiro Kawasaki

National Institute of Genetics

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge