Network


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

Hotspot


Dive into the research topics where Ryosuke Tsunematsu is active.

Publication


Featured researches published by Ryosuke Tsunematsu.


The EMBO Journal | 2000

Targeted disruption of Skp2 results in accumulation of cyclin E and p27Kip1, polyploidy and centrosome overduplication

Keiko Nakayama; Hiroyasu Nagahama; Yohji A. Minamishima; Masaki Matsumoto; Ikuo Nakamichi; Kyoko Kitagawa; Michiko Shirane; Ryosuke Tsunematsu; Tadasuke Tsukiyama; Noriko Ishida; Masatoshi Kitagawa; Keiichi I. Nakayama; Shigetsugu Hatakeyama

The ubiquitin–proteasome pathway plays an important role in control of the abundance of cell cycle regulators. Mice lacking Skp2, an F‐box protein and substrate recognition component of an Skp1–Cullin–F‐box protein (SCF) ubiquitin ligase, were generated. Although Skp2−/− animals are viable, cells in the mutant mice contain markedly enlarged nuclei with polyploidy and multiple centrosomes, and show a reduced growth rate and increased apoptosis. Skp2−/− cells also exhibit increased accumulation of both cyclin E and p27Kip1. The elimination of cyclin E during S and G2 phases is impaired in Skp2−/− cells, resulting in loss of cyclin E periodicity. Biochemical studies showed that Skp2 interacts specifically with cyclin E and thereby promotes its ubiquitylation and degradation both in vivo and in vitro. These results suggest that specific degradation of cyclin E and p27Kip1 is mediated by the SCFSkp2 ubiquitin ligase complex, and that Skp2 may control chromosome replication and centrosome duplication by determining the abundance of cell cycle regulators.


The EMBO Journal | 2004

Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7

Masayoshi Yada; Shigetsugu Hatakeyama; Takumi Kamura; Masaaki Nishiyama; Ryosuke Tsunematsu; Hiroyuki Imaki; Noriko Ishida; Fumihiko Okumura; Keiko Nakayama; Keiichi I. Nakayama

The F‐box protein Skp2 mediates c‐Myc ubiquitylation by binding to the MB2 domain. However, the turnover of c‐Myc is largely dependent on phosphorylation of threonine‐58 and serine‐62 in MB1, residues that are often mutated in cancer. We now show that the F‐box protein Fbw7 interacts with and thereby destabilizes c‐Myc in a manner dependent on phosphorylation of MB1. Whereas wild‐type Fbw7 promoted c‐Myc turnover in cells, an Fbw7 mutant lacking the F‐box domain delayed it. Furthermore, depletion of Fbw7 by RNA interference increased both the abundance and transactivation activity of c‐Myc. Accumulation of c‐Myc was also apparent in mouse Fbw7−/− embryonic stem cells. These observations suggest that two F‐box proteins, Fbw7 and Skp2, differentially regulate c‐Myc stability by targeting MB1 and MB2, respectively.


Nature | 2004

Fbxw7 / Cdc4 is a p53-dependent, haploinsufficient tumour suppressor gene

Jian-Hua Mao; Jesus Perez-Losada; Di Wu; Reyno DelRosario; Ryosuke Tsunematsu; Keiichi I. Nakayama; Kenneth A. Brown; Sheila Bryson; Allan Balmain

The FBXW7/hCDC4 gene encodes a ubiquitin ligase implicated in the control of chromosome stability. Here we identify the mouse Fbxw7 gene as a p53-dependent tumour suppressor gene by using a mammalian genetic screen for p53-dependent genes involved in tumorigenesis. Radiation-induced lymphomas from p53+/- mice, but not those from p53-/- mice, show frequent loss of heterozygosity and a 10% mutation rate of the Fbxw7 gene. Fbxw7+/- mice have greater susceptibility to radiation-induced tumorigenesis, but most tumours retain and express the wild-type allele, indicating that Fbxw7 is a haploinsufficient tumour suppressor gene. Loss of Fbxw7 alters the spectrum of tumours that develop in p53 deficient mice to include a range of tumours in epithelial tissues such as the lung, liver and ovary. Mouse embryo fibroblasts from Fbxw7-deficient mice, or wild-type mouse cells expressing Fbxw7 small interfering RNA, have higher levels of Aurora-A kinase, c-Jun and Notch4, but not of cyclin E. We propose that p53-dependent loss of Fbxw7 leads to genetic instability by mechanisms that might involve the activation of Aurora-A, providing a rationale for the early occurrence of these mutations in human cancers.


Journal of Biological Chemistry | 2004

Mouse Fbw7/Sel-10/Cdc4 Is Required for Notch Degradation during Vascular Development

Ryosuke Tsunematsu; Keiko Nakayama; Yuichi Oike; Masaaki Nishiyama; Noriko Ishida; Shigetsugu Hatakeyama; Yasumasa Bessho; Ryoichiro Kageyama; Toshio Suda; Keiichi I. Nakayama

Mammalian Fbw7 (also known as Sel-10, hCdc4, or hAgo) is the F-box protein component of an SCF (Skp1-Cul1-F-box protein-Rbx1)-type ubiquitin ligase, and the mouse Fbw7 is expressed prominently in the endothelial cell lineage of embryos. We generated mice deficient in Fbw7 and found that the embryos died in utero at embryonic day 10.5-11.5, manifesting marked abnormalities in vascular development. Vascular remodeling was impaired in the brain and yolk sac, and the major trunk veins were not formed. In vitro para-aortic splanchnopleural explant cultures from Fbw7-/- embryos also manifested an impairment of vascular network formation. Notch4, which is the product of the proto-oncogene Int3 and an endothelial cell-specific mammalian isoform of Notch, accumulated in Fbw7-/- embryos, resulting in an increased expression of Hey1, which encodes a transcriptional repressor that acts downstream of Notch signaling and is implicated in vascular development. Expression of Notch1, -2, or -3 or of cyclin E was unaffected in Fbw7-/- embryos. Mammalian Fbw7 thus appears to play an indispensable role in negative regulation of the Notch4-Hey1 pathway and is required for vascular development.


Cell | 2013

FBXL21 Regulates Oscillation of the Circadian Clock through Ubiquitination and Stabilization of Cryptochromes

Arisa Hirano; Kanae Yumimoto; Ryosuke Tsunematsu; Masaki Matsumoto; Masaaki Oyama; Hiroko Kozuka-Hata; Tomoki Nakagawa; Darin Lanjakornsiripan; Keiichi I. Nakayama; Yoshitaka Fukada

In the mammalian circadian clockwork, CRY1 and CRY2 repressor proteins are regulated by posttranslational modifications for temporally coordinated transcription of clock genes. Previous studies revealed that FBXL3, an F-box-type E3 ligase, ubiquitinates CRYs and mediates their degradation. Here, we found that FBXL21 also ubiquitinates CRYs but counteracts FBXL3. Fbxl21(-/-) mice exhibited normal periodicity of wheel-running rhythms with compromised organization of daily activities, while an extremely long-period phenotype of Fbxl3(-/-) mice was attenuated in Fbxl3/Fbxl21 double-knockout mice. The double knockout destabilized the behavioral rhythms progressively and sometimes elicited arrhythmicity. Surprisingly, FBXL21 stabilized CRYs and antagonized the destabilizing action by FBXL3. Predominantly cytosolic distribution of FBXL21 contrasts with nuclear localization of FBXL3. These results emphasize the physiological importance of antagonizing actions between FBXL21 and FBXL3 on CRYs, and their combined actions at different subcellular locations stabilize oscillation of the circadian clock.


PLOS ONE | 2011

Isolation and Characterization of Human Trophoblast Side-Population (SP) Cells in Primary Villous Cytotrophoblasts and HTR-8/SVneo Cell Line

Tomoka Takao; Kazuo Asanoma; Kiyoko Kato; Kotaro Fukushima; Ryosuke Tsunematsu; Toshio Hirakawa; Sueo Matsumura; Hiroyuki Seki; Satoru Takeda; Norio Wake

Recently, numerous studies have identified that immature cell populations including stem cells and progenitor cells can be found among “side-population” (SP) cells. Although SP cells isolated from some adult tissues have been reported elsewhere, isolation and characterization of human trophoblast SP remained to be reported. In this study, HTR-8/SVneo cells and human primary villous cytotrophoblasts (vCTBs) were stained with Hoechst 33342 and SP and non-SP (NSP) fractions were isolated using a cell sorter. A small population of SP cells was identified in HTR-8/SVneo cells and in vCTBs. SP cells expressed several vCTB-specific markers and failed to express syncytiotrophoblast (STB) or extravillous cytotrophopblast (EVT)-specific differentiation markers. SP cells formed colonies and proliferated on mouse embryonic fibroblast (MEF) feeder cells or in MEF conditioned medium supplemented with heparin/FGF2, and they also showed long-term repopulating property. SP cells could differentiate into both STB and EVT cell lineages and expressed several differentiation markers. Microarray analysis revealed that IL7R and IL1R2 were exclusively expressed in SP cells and not in NSP cells. vCTB cells sorted as positive for both IL7R and IL1R2 failed to express trophoblast differentiation markers and spontaneously differentiated into both STB and EVT in basal medium. These features shown by the SP cells suggested that IL7R and IL1R2 are available as markers to detect the SP cells and that vCTB progenitor cells and trophoblast stem cells were involved in the SP cell population.


Cancer Science | 2006

Fbxw7 contributes to tumor suppression by targeting multiple proteins for ubiquitin-dependent degradation

Yo Fujii; Masayoshi Yada; Masaaki Nishiyama; Takumi Kamura; Hidehisa Takahashi; Ryosuke Tsunematsu; Etsuo Susaki; Tadashi Nakagawa; Akinobu Matsumoto; Keiichi I. Nakayama

Fbxw7 (also known as Sel‐10, hCdc4 or hAgo) is the F‐box protein component of a Skp1–Cul1–F‐box protein (SCF) ubiquitin ligase. Fbxw7 contributes to the ubiquitin‐mediated degradation of cyclin E, c‐Myc, Aurora‐A, Notch and c‐Jun, all of which appear to function as cell‐cycle promoters and oncogenic proteins. Loss of Fbxw7 results in elevated expression of its substrates, which may lead to oncogenesis. However, it remains largely unclear which accumulating substrate is most related to cancer development in Fbxw7‐mutant cancer cells. In the present study, we examined the abundance of cyclin E, c‐Myc and Aurora‐A in seven cancer cell lines, which harbor wild‐type (three lines) or mutant (four lines) Fbxw7. Although these three substrates accumulated in the Fbxw7‐mutant cells, the extent of increase in the expression of these proteins varied in each line. Forced expression of Fbxw7 reduced the levels of cyclin E, c‐Myc and Aurora‐A in the Fbxw7‐mutant cells. In contrast, a decrease in the expression of cyclin E, c‐Myc or Aurora‐A by RNA interference significantly suppressed the rate of proliferation and anchorage‐independent growth of the Fbxw7‐mutant cells. These findings thus suggest that the loss of Fbxw7 results in accumulation of cyclin E, c‐Myc and Aurora‐A, all of which appear to be required for growth promotion of cancer cells. Fbxw7 seems to regulate the levels of multiple targets to suppress cancer development. (Cancer Sci 2006; 97: 729–736)


Molecular and Cellular Biology | 2012

Genetic Reevaluation of the Role of F-Box Proteins in Cyclin D1 Degradation

Tomoharu Kanie; Ichiro Onoyama; Akinobu Matsumoto; Masanori Yamada; Hirokazu Nakatsumi; Yuki Tateishi; So Yamamura; Ryosuke Tsunematsu; Masaki Matsumoto; Keiichi I. Nakayama

ABSTRACT D-type cyclins play a pivotal role in G1-S progression of the cell cycle, and their expression is frequently deregulated in cancer. Cyclin D1 has a half-life of only ∼30 min as a result of its ubiquitylation and proteasomal degradation, with various F-box proteins, including Fbxo4, Fbxw8, Skp2, and Fbxo31, having been found to contribute to its ubiquitylation. We have now generated Fbxo4-deficient mice and found no abnormalities in these animals. Cyclin D1 accumulation was thus not observed in Fbxo4−/− mouse tissues. The half-life of cyclin D1 in mouse embryonic fibroblasts (MEFs) prepared from Fbxo4−/−, Fbxw8−/−, and Fbxo4−/−; Fbxw8−/− mice also did not differ from that in wild-type MEFs. Additional depletion of Skp2 and Fbxo31 in Fbxo4−/−; Fbxw8−/− MEFs by RNA interference did not affect cyclin D1 stability. Although Fbxo31 depletion in MEFs increased cyclin D1 abundance, this effect appeared attributable to upregulation of cyclin D1 mRNA. Furthermore, abrogation of the function of the Skp1–Cul1–F-box protein (SCF) complex or the anaphase-promoting complex/cyclosome (APC/C) complexes did not alter the half-life of cyclin D1, whereas cyclin D1 degradation was dependent largely on proteasome activity. Our genetic analyses thus do not support a role for any of the four F-box proteins examined in cyclin D1 degradation during normal cell cycle progression. They suggest the existence of other ubiquitin ligases that target cyclin D1 for proteolysis.


Molecular and Cellular Biology | 2004

Early Embryonic Death in Mice Lacking the β-Catenin-Binding Protein Duplin

Masaaki Nishiyama; Keiko Nakayama; Ryosuke Tsunematsu; Tadasuke Tsukiyama; Akira Kikuchi; Keiichi I. Nakayama

ABSTRACT The Wnt signaling pathway plays a pivotal role in vertebrate early development and morphogenesis. Duplin (axis duplication inhibitor) interacts with β-catenin and prevents its binding to Tcf, thereby inhibiting downstream Wnt signaling. Here we show that Duplin is expressed predominantly from early- to mid-stage mouse embryogenesis, and we describe the generation of mice deficient in Duplin. Duplin−/− embryos manifest growth retardation from embryonic day 5.5 (E5.5) and developmental arrest accompanied by massive apoptosis at E7.5. The mutant embryos develop into an egg cylinder but do not form a primitive streak or mesoderm. Expression of β-catenin target genes, including those for T (brachyury), Axin2, and cyclin D1, was not increased in Duplin −/− embryos, suggesting that the developmental defect is not simply attributable to upregulation of Wnt signaling caused by the lack of this inhibitor. These results suggest that Duplin plays an indispensable role, likely by a mechanism independent of inhibition of Wnt signaling, in mouse embryonic growth and differentiation at an early developmental stage.


Molecular and Cellular Biology | 2006

Fbxw8 Is Essential for Cul1-Cul7 Complex Formation and for Placental Development

Ryosuke Tsunematsu; Masaaki Nishiyama; Shuhei Kotoshiba; Toru Saiga; Takumi Kamura; Keiichi I. Nakayama

ABSTRACT Cullin-based ubiquitin ligases (E3s) constitute one of the largest E3 families. Fbxw8 (also known as Fbw6 or Fbx29) is an F-box protein that is assembled with Cul7 in an SCF-like E3 complex. Here we show that Cul7 forms a heterodimeric complex with Cul1 in a manner dependent on Fbxw8. We generated mice deficient in Fbxw8 and found that Cul7 did not associate with Cul1 in cells of these mice. Two-thirds of Fbxw8−/− embryos die in utero, whereas the remaining one-third are born alive and grow to adulthood. Fbxw8−/− embryos show intrauterine growth retardation and abnormal development of the placenta, characterized by both a reduced thickness of the spongiotrophoblast layer and abnormal vessel structure in the labyrinth layer. Although the placental phenotype of Fbxw8−/− mice resembles that of Cul7−/− mice, other abnormalities of Cul7−/− mice are not apparent in Fbxw8−/− mice. These results suggest that the Cul7-based SCF-like E3 complex has both Fbxw8-dependent and Fbxw8-independent functions.

Collaboration


Dive into the Ryosuke Tsunematsu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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