Network


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

Hotspot


Dive into the research topics where Shigetsugu Hatakeyama is active.

Publication


Featured researches published by Shigetsugu Hatakeyama.


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.


The EMBO Journal | 1999

An F-box protein, FWD1, mediates ubiquitin-dependent proteolysis of beta-catenin.

Masatoshi Kitagawa; Shigetsugu Hatakeyama; Michiko Shirane; Masaki Matsumoto; Noriko Ishida; Kimihiko Hattori; Ikuo Nakamichi; Akira Kikuchi; Keiichi I. Nakayama; Keiko Nakayama

β‐catenin plays an essential role in the Wingless/Wnt signaling cascade and is a component of the cadherin cell adhesion complex. Deregulation of β‐catenin accumulation as a result of mutations in adenomatous polyposis coli (APC) tumor suppressor protein is believed to initiate colorectal neoplasia. β‐catenin levels are regulated by the ubiquitin‐dependent proteolysis system and β‐catenin ubiquitination is preceded by phosphorylation of its N‐terminal region by the glycogen synthase kinase‐3β (GSK‐3β)/Axin kinase complex. Here we show that FWD1 (the mouse homologue of Slimb/βTrCP), an F‐box/WD40‐repeat protein, specifically formed a multi‐molecular complex with β‐catenin, Axin, GSK‐3β and APC. Mutations at the signal‐induced phosphorylation site of β‐catenin inhibited its association with FWD1. FWD1 facilitated ubiquitination and promoted degradation of β‐catenin, resulting in reduced cytoplasmic β‐catenin levels. In contrast, a dominant‐negative mutant form of FWD1 inhibited the ubiquitination process and stabilized β‐catenin. These results suggest that the Skp1/Cullin/F‐box protein FWD1 (SCFFWD1)–ubiquitin ligase complex is involved in β‐catenin ubiquitination and that FWD1 serves as an intracellular receptor for phosphorylated β‐catenin. FWD1 also links the phosphorylation machinery to the ubiquitin–proteasome pathway to ensure prompt and efficient proteolysis of β‐catenin in response to external signals. SCFFWD1 may be critical for tumor development and suppression through regulation of β‐catenin protein stability.


Molecular Cell | 2002

CHIP Is Associated with Parkin, a Gene Responsible for Familial Parkinson's Disease, and Enhances Its Ubiquitin Ligase Activity

Yuzuru Imai; Mariko Soda; Shigetsugu Hatakeyama; Takumi Akagi; Tsutomu Hashikawa; Keiichi I. Nakayama; Ryosuke Takahashi

Unfolded Pael receptor (Pael-R) is a substrate of the E3 ubiquitin ligase Parkin. Accumulation of Pael-R in the endoplasmic reticulum (ER) of dopaminergic neurons induces ER stress leading to neurodegeneration. Here, we show that CHIP, Hsp70, Parkin, and Pael-R formed a complex in vitro and in vivo. The amount of CHIP in the complex was increased during ER stress. CHIP promoted the dissociation of Hsp70 from Parkin and Pael-R, thus facilitating Parkin-mediated Pael-R ubiquitination. Moreover, CHIP enhanced Parkin-mediated in vitro ubiquitination of Pael-R in the absence of Hsp70. Furthermore, CHIP enhanced the ability of Parkin to inhibit cell death induced by Pael-R. Taken together, these results indicate that CHIP is a mammalian E4-like molecule that positively regulates Parkin E3 activity.


Nature | 2002

Increased proliferation of B cells and auto-immunity in mice lacking protein kinase Cδ

Akitomo Miyamoto; Keiko Nakayama; Hiroyuki Imaki; Sachiko Hirose; Yi Jiang; Masaaki Abe; Tadasuke Tsukiyama; Hiroyasu Nagahama; Shigeo Ohno; Shigetsugu Hatakeyama; Keiichi I. Nakayama

Protein kinase C (PKC), which comprises 11 closely related isoforms, has been implicated in a wide variety of cellular processes, such as growth, differentiation, secretion, apoptosis and tumour development. Among the PKC isotypes, PKC-δ is unique in that its overexpression results in inhibition of cell growth. Here we show that mice that lack PKC-δ exhibit expansion of the B-lymphocyte population with the formation of numerous germinal centres in the absence of stimulation. The rate of proliferation in response to stimulation was greater for B cells from PKC-δ-deficient mice than for those from wild-type mice. Adoptive transfer experiments suggested that the hyperproliferation phenotype is B-cell autonomous. Production of interleukin-6 was markedly increased in B cells of PKC-δ-null mice as a result of an increase in the DNA-binding activity of NF-IL6. Furthermore, the PKC-δ-deficient mice contain circulating autoreactive antibodies and display immune-complex-type glomerulonephritis, as well as lymphocyte infiltration in many organs. These results suggest that PKC-δ has an indispensable function in negative regulation of B-cell proliferation, and is particularly important for the establishment of B-cell tolerance.


Nature Cell Biology | 2004

Cytoplasmic ubiquitin ligase KPC regulates proteolysis of p27Kip1 at G1 phase

Takumi Kamura; Taichi Hara; Masaki Matsumoto; Noriko Ishida; Fumihiko Okumura; Shigetsugu Hatakeyama; Minoru Yoshida; Keiko Nakayama; Keiichi I. Nakayama

The cyclin-dependent kinase inhibitor p27Kip1 is degraded at the G0–G1 transition of the cell cycle by the ubiquitin–proteasome pathway. Although the nuclear ubiquitin ligase (E3) SCFSkp2 is implicated in p27Kip1 degradation, proteolysis of p27Kip1 at the G0–G1 transition proceeds normally in Skp2−/− cells. Moreover, p27Kip1 is exported from the nucleus to the cytoplasm at G0–G1 (refs 9–11). These data suggest the existence of a Skp2-independent pathway for the degradation of p27Kip1 at G1 phase. We now describe a previously unidentified E3 complex: KPC (Kip1 ubiquitination-promoting complex), consisting of KPC1 and KPC2. KPC1 contains a RING-finger domain, and KPC2 contains a ubiquitin-like domain and two ubiquitin-associated domains. KPC interacts with and ubiquitinates p27Kip1 and is localized to the cytoplasm. Overexpression of KPC promoted the degradation of p27Kip1, whereas a dominant-negative mutant of KPC1 delayed p27Kip1 degradation. The nuclear export of p27Kip1 by CRM1 seems to be necessary for KPC-mediated proteolysis. Depletion of KPC1 by RNA interference also inhibited p27Kip1 degradation. KPC thus probably controls degradation of p27Kip1 in G1 phase after export of the latter from the nucleus.


Developmental Cell | 2004

Skp2-Mediated Degradation of p27 Regulates Progression into Mitosis

Keiko Nakayama; Hiroyasu Nagahama; Yohji A. Minamishima; Satoshi Miyake; Noriko Ishida; Shigetsugu Hatakeyama; Masatoshi Kitagawa; Shun-ichiro Iemura; Tohru Natsume; Keiichi I. Nakayama

Although Skp2 has been thought to mediate the degradation of p27 at the G(1)-S transition, Skp2(-/-) cells exhibit accumulation of p27 in S-G(2) phase with overreplication. We demonstrate that Skp2(-/-)p27(-/-) mice do not exhibit the overreplication phenotype, suggesting that p27 accumulation is required for its development. Hepatocytes of Skp2(-/-) mice entered the endoduplication cycle after mitogenic stimulation, whereas this phenotype was not apparent in Skp2(-/-)p27(-/-) mice. Cdc2-associated kinase activity was lower in Skp2(-/-) cells than in wild-type cells, and a reduction in Cdc2 activity was sufficient to induce overreplication. The lack of p27 degradation in G(2) phase in Skp2(-/-) cells may thus result in suppression of Cdc2 activity and consequent inhibition of entry into M phase. These data suggest that p27 proteolysis is necessary for the activation of not only Cdk2 but also Cdc2, and that Skp2 contributes to regulation of G(2)-M progression by mediating the degradation of p27.


Nature Reviews Cancer | 2011

TRIM proteins and cancer

Shigetsugu Hatakeyama

Emerging clinical evidence shows that the deregulation of ubiquitin-mediated degradation of oncogene products or tumour suppressors is likely to be involved in the aetiology of carcinomas and leukaemias. Recent studies have indicated that some members of the tripartite motif (TRIM) proteins (one of the subfamilies of the RING type E3 ubiquitin ligases) function as important regulators for carcinogenesis. This Review focuses on TRIM proteins that are involved in tumour development and progression.


Journal of Biological Chemistry | 1999

Down-regulation of p27(Kip1) by two mechanisms, ubiquitin-mediated degradation and proteolytic processing.

Michiko Shirane; Yumiko Harumiya; Noriko Ishida; Aizan Hirai; Chikara Miyamoto; Shigetsugu Hatakeyama; Kei-ichi Nakayama; Masatoshi Kitagawa

The intracellular level of p27 Kip1 , a cyclin-dependent kinase (CDK) inhibitory protein, is rapidly reduced at the G1/S transition phase when the cell cycle pause ceases. In this study, we demonstrated that two posttranslational mechanisms were involved in p27 Kip1 breakdown: degradation via the ubiquitin (Ub)-proteasome pathway and proteolytic processing that rapidly eliminates the cyclin-binding domain. We confirmed that p27 Kip1 was ubiquitinated in vitroas well as in vivo. The p27 Kip1 -ubiquitination activity was higher at the G1/S boundary than during the G0/G1 phase, and p27 Kip1 ubiquitination was reduced significantly when the lysine residues at positions 134, 153, and 165 were replaced by arginine, suggesting that these lysine residues are the targets for Ub conjugation. In parallel with its Ub-dependent degradation, p27 Kip1 was processed rapidly at its N terminus, reducing its molecular mass from 27 to 22 kDa, by a ubiquitination-independent but adenosine triphosphate (ATP)-dependent mechanism with higher activity during the S than the G0/G1 phase. This 22-kDa intermediate had no cyclin-binding domain at its N terminus and virtually no CDK2 kinase inhibitory activity. These results suggest that p27 Kip1 is eliminated by two independent mechanisms, ubiquitin-mediated degradation and ubiquitin-independent processing, during progression from the G1 to S phase.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Degradation of p57Kip2 mediated by SCFSkp2-dependent ubiquitylation

Takumi Kamura; Taichi Hara; Shuhei Kotoshiba; Masayoshi Yada; Noriko Ishida; Hiroyuki Imaki; Shigetsugu Hatakeyama; Keiko Nakayama; Keiichi I. Nakayama

The abundance of the cyclin-dependent kinase (CDK) inhibitor p57Kip2, an important regulator of cell cycle progression, is thought to be controlled by the ubiquitin-proteasome pathway. The Skp1/Cul1/F-box (SCF)-type E3 ubiquitin ligase complex SCFSkp2 has now been shown to be responsible for regulating the cellular level of p57Kip2 by targeting it for ubiquitylation and proteolysis. The elimination of p57Kip2 was impaired in Skp2-/- cells, resulting in abnormal accumulation of the protein. Coimmunoprecipitation analysis also revealed that Skp2 interacts with p57Kip2 in vivo. Overexpression of WT Skp2 promoted degradation of p57Kip2, whereas expression of a dominant negative mutant of Skp2 prolonged the half-life of p57Kip2. Mutation of the threonine residue (Thr-310) of human p57Kip2 that is conserved between the COOH-terminal QT domains of p57Kip2 and p27Kip1 prevented the effect of Skp2 on the stability of p57Kip2, suggesting that phosphorylation at this site is required for SCFSkp2-mediated ubiquitylation. Finally, the purified recombinant SCFSkp2 complex mediated p57Kip2 ubiquitylation in vitro in a manner dependent on the presence of the cyclin E-CDK2 complex. These observations thus demonstrate that the SCFSkp2 complex plays an important role in cell-cycle progression by determining the abundance of p57Kip2 and that of the related CDK inhibitor p27Kip1.

Collaboration


Dive into the Shigetsugu Hatakeyama'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

Kazumasa Ogasawara

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge