Network


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

Hotspot


Dive into the research topics where Noriko Yoneda-Kato is active.

Publication


Featured researches published by Noriko Yoneda-Kato.


Journal of Biological Chemistry | 2004

Multiple Functions of Jab1 Are Required for Early Embryonic Development and Growth Potential in Mice

Kiichiro Tomoda; Noriko Yoneda-Kato; Akihisa Fukumoto; Shinya Yamanaka; Jun-ya Kato

Jab1 interacts with a variety of signaling molecules and regulates their stability in mammalian cells. As the fifth component of the COP9 signalosome (CSN) complex, Jab1 (CSN5) plays a central role in the deneddylation of the cullin subunit of the Skp1-Cullin-F box protein ubiquitin ligase complex. In addition, a CSN-independent function of Jab1 is suggested but is less well characterized. To elucidate the function of Jab1, we targeted the Jab1 locus by homologous recombination in mouse embryonic stem cells. Jab1-null embryos died soon after implantation. Jab1-/- embryonic cells, which lacked other CSN components, expressed higher levels of p27, p53, and cyclin E, resulting in impaired proliferation and accelerated apoptosis. Jab1 heterozygous mice were healthy and fertile but smaller than their wild-type littermates. Jab1+/- mouse embryonic fibroblast cells, in which the amount of Jab1-containing small subcomplex, but not that of CSN, was selectively reduced, proliferated poorly, showed an inefficient down-regulation of p27 during G1, and was delayed in the progression from G0 to S phase by 3 h compared with the wild-type cells. Most interestingly, in Jab1+/- mouse embryonic fibroblasts, the levels of cyclin E and deneddylated Cul1 were unchanged, and p53 was not induced. Thus, Jab1 controls cell cycle progression and cell survival by regulating multiple cell cycle signaling pathways.


Genes to Cells | 2009

Mammalian COP9 signalosome.

Jun-ya Kato; Noriko Yoneda-Kato

The COP9 signalosome (CSN) complex is highly conserved from yeast to human. Although the plant CSN was first identified as a negative regulator of photomorphogenesis, the mammalian CSN is linked to different biological responses such as checkpoint control, signal transduction, development and the cell cycle. Frequent over‐expression of the CSN subunit in a variety of human cancers suggests its involvement in cell transformation and tumorigenesis. The best‐known biochemical function associated with the CSN is the control of protein stability via the ubiquitin–proteasome system through regulation of cullin‐RING‐E3 ubiquitin ligase activity by deneddylation, by controlling the activity of COP1 E3 ligase, or by counteracting ubiquitin‐mediated degradation through a CSN‐associated deubiquitinating enzyme. In addition to affecting the stability of transcription factors, the CSN may regulate gene transcription by directly associating with chromatin. This review summarizes recent findings and discusses the physiological role and the cellular function of the mammalian CSN in terms of the regulation of cell proliferation.


The EMBO Journal | 2005

Myeloid leukemia factor 1 regulates p53 by suppressing COP1 via COP9 signalosome subunit 3

Noriko Yoneda-Kato; Kiichiro Tomoda; Mari Umehara; Yukinobu Arata; Jun-ya Kato

Myeloid leukemia factor 1 (MLF1) was first identified as the leukemic fusion protein NPM‐MLF1 generated by the t(3;5)(q25.1;q34) chromosomal translocation. Although MLF1 expresses normally in a variety of tissues including hematopoietic stem cells and the overexpression of MLF1 correlates with malignant transformation in human cancer, little is known about how MLF1 is involved in the regulation of cell growth. Here we show that MLF1 is a negative regulator of cell cycle progression functioning upstream of the tumor suppressor p53. MLF1 induces p53‐dependent cell cycle arrest in murine embryonic fibroblasts. This action requires a novel binding partner, subunit 3 of the COP9 signalosome (CSN3). A reduction in the level of CSN3 protein with small interfering RNA abrogated MLF1‐induced G1 arrest and impaired the activation of p53 by genotoxic stress. Furthermore, ectopic MLF1 expression and CSN3 knockdown inversely affect the endogenous level of COP1, a ubiquitin ligase for p53. Exogenous expression of COP1 overcomes MLF1‐induced growth arrest. These results indicate that MLF1 is a critical regulator of p53 and suggest its involvement in leukemogenesis through a novel CSN3–COP1 pathway.


FEBS Letters | 2005

Small Jab1-containing subcomplex is regulated in an anchorage- and cell cycle-dependent manner, which is abrogated by ras transformation

Akihisa Fukumoto; Kiichiro Tomoda; Makoto Kubota; Jun-ya Kato; Noriko Yoneda-Kato

Jab1 interacts with a variety of cell cycle and signal transduction regulators to control cell proliferation, differentiation, and tumorigenesis. In this study, we employed a non‐denaturing gel electrophoresis method to separate different Jab1‐containing complexes, the COP9 signalosome complex and the small Jab1‐containing subcomplex. The formation of the small Jab1 complex was dependent on a low cell density and anchorage to a solid support, and enhanced during the early G1 phase of the cell cycle, which was abrogated in ras‐transformed cells. The small Jab1‐containing subcomplex may be a novel mediator of anchorage and cell–cell contact‐dependent signal transduction.


Molecular and Cellular Biology | 2008

Shuttling Imbalance of MLF1 Results in p53 Instability and Increases Susceptibility to Oncogenic Transformation

Noriko Yoneda-Kato; Jun-ya Kato

ABSTRACT Myeloid leukemia factor 1 (MLF1) stabilizes the activity of the tumor suppressor p53 by suppressing its E3 ubiquitin ligase, COP1, through a third component of the COP9 signalosome (CSN3). However, little is known about how MLF1 functions upstream of the CSN3-COP1-p53 pathway and how its deregulation by the formation of the fusion protein nucleophosmin (NPM)-MLF1, generated by t(3;5)(q25.1;q34) chromosomal translocation, leads to leukemogenesis. Here we show that MLF1 is a cytoplasmic-nuclear-shuttling protein and that its nucleolar localization on fusing with NPM prevents the full induction of p53 by both genotoxic and oncogenic cellular stress. The majority of MLF1 was located in the cytoplasm, but the treatment of cells with leptomycin B rapidly induced a nuclear accumulation of MLF1. A mutation of the nuclear export signal (NES) motif identified in the MLF1 sequence enhanced the antiproliferative activity of MLF1. The fusion of MLF1 with NPM translocated MLF1 to the nucleolus and abolished the growth-suppressing activity. The introduction of NPM-MLF1 into early-passage murine embryonic fibroblasts allowed the cells to escape from cellular senescence at a markedly earlier stage and induced neoplastic transformation in collaboration with the oncogenic form of Ras. Interestingly, disruption of the MLF1-derived NES sequence completely abolished the growth-promoting activity of NPM-MLF1 in murine fibroblasts and hematopoietic cells. Thus, our results provide important evidence that the shuttling of MLF1 is critical for the regulation of cell proliferation and a disturbance in the shuttling balance increases the cells susceptibility to oncogenic transformation.


Journal of Biological Chemistry | 2001

Direct Binding of the Signal-transducing Adaptor Grb2 Facilitates Down-regulation of the Cyclin-dependent Kinase Inhibitor p27Kip1

Yoriko Sugiyama; Kiichiro Tomoda; Toshiaki Tanaka; Yukinobu Arata; Noriko Yoneda-Kato; Jun-ya Kato

Ectopic expression of Jab1/CSN5 induces specific down-regulation of the cyclin-dependent kinase (Cdk) inhibitor p27 (p27Kip1) in a manner dependent upon transportation from the nucleus to the cytoplasm. Here we show that Grb2 and Grb3-3, the molecules functioning as an adaptor in the signal transduction pathway, specifically and directly bind to p27 in the cytoplasm and participate in the regulation of p27. The interaction requires the C-terminal SH3-domain of Grb2/3-3 and the proline-rich sequence contained in p27 immediately downstream of the Cdk binding domain. In living cells, enforcement of the cytoplasmic localization of p27, either by artificial manipulation of the nuclear/cytoplasmic transport signal sequence or by coexpression of ectopic Jab1/CSN5, markedly enhances the stable interaction between p27 and Grb2. Overexpression of Grb2 accelerates Jab1/CSN5-mediated degradation of p27, while Grb3-3 expression suppresses it. A p27 mutant unable to bind to Grb2 is transported into the cytoplasm in cells ectopically expressing Jab1/CSN5 but is refractory to the subsequent degradation. These findings indicate that Grb2 participates in a negative regulation of p27 and may directly link the signal transduction pathway with the cell cycle regulatory machinery.


FEBS Letters | 2006

Depletion of Jab1 inhibits proliferation of pancreatic cancer cell lines

Akihisa Fukumoto; Kiichiro Tomoda; Noriko Yoneda-Kato; Yoshiyuki Nakajima; Jun-ya Kato

Jab1 overexpression is observed in many human cancers, but its physiological significance remains to be investigated. We reduced the level of Jab1 expression in pancreatic cancer cell lines, MIA PaCa‐2 and PANC‐1 by the RNA interference and found that Jab1‐knockdown resulted in impaired cell proliferation and enhanced apoptosis regardless of the genotype of the tumor suppressor p53. This growth inhibition was rescued by the introduction of siRNA‐resistant mouse Jab1 cDNA. Jab1‐knocked‐down cells expressed a higher level of c‐myc, and additional depletion of c‐myc rescued cells from Jab1‐knockdown‐mediated growth suppression. Thus, Jab1 overexpression contributes to pancreatic cancer cell proliferation and survival. Jab1 could be a novel target in cancer therapy.


Blood | 2013

COP1 targets C/EBPα for degradation and induces acute myeloid leukemia via Trib1

Akihiro Yoshida; Jun-ya Kato; Ikuko Nakamae; Noriko Yoneda-Kato

The ubiquitin ligase constitutively photomorphogenic 1 (COP1) is involved in many biological responses in mammalian cells, but its role in tumorigenesis remains unclear. Here we show that COP1 is a ubiquitin ligase for the tumor suppressor CCAAT/enhancer-binding protein (C/EBPα) and promotes its degradation in vivo, thereby blocking myeloid differentiation of hematopoietic cells for tumorigenesis. In this process, mammalian homolog of Tribbles, Trib1, which contains a COP1-binding motif, is essential for down-regulation of C/EBPα expression. Murine bone marrow transplantation experiments showed that coexpression of COP1 accelerates development of acute myeloid leukemia induced by Trib1, which pathologically resembles that of p42C/EBPα-deficient mice. Interestingly, coexpression of ligase activity-deficient COP1 mutant abrogated Trib1-induced leukemogenesis. These results indicate that COP1 and Trib1 act as an oncoprotein complex functioning upstream of C/EBPα, and its ligase activity is crucial for leukemogenesis.


Scientific Reports | 2013

CSN5 specifically interacts with CDK2 and controls senescence in a cytoplasmic cyclin E-mediated manner.

Akihiro Yoshida; Noriko Yoneda-Kato; Jun-ya Kato

The fifth component (CSN5) of the mammalian COP9 signalosome complex plays an essential role in cell proliferation and senescence, but its molecular mediator remains to be determined. Here, we searched for interactors among various cell cycle regulators, and found that CSN5, but not the CSN holo-complex, bound to CDK2 in vivo and in vitro. Depletion of CSN5 enhanced phosphorylation of CDK2 by Akt, resulting in cytoplasmic accumulation of CDK2 together with cyclin E in a leptomycin B-resistant manner, and impaired phosphorylation of the retinoblastoma protein. Additional knockdown of CDK2, which reduced the expression of cyclin E to the normal level, did not restore cell proliferation, but significantly suppressed senescence in CSN5-depleted cells. Enforced expression of cytoplasmic cyclin E induced premature senescence in immortalized cell lines. These results show that CSN5 functions through CDK2 to control premature senescence in a novel way, depending on cyclin E in the cytoplasm.


Journal of Biological Chemistry | 2008

Stable Form of JAB1 Enhances Proliferation and Maintenance of Hematopoietic Progenitors

Masaaki Mori; Noriko Yoneda-Kato; Akihiro Yoshida; Jun-ya Kato

Overexpression of JAB1 is observed in a variety of human cancers, but how JAB1 is involved in tumor development remained to be investigated. Here we analyzed mice with modified Jab1 expression. Mice ectopically expressing a more stable form of JAB1 protein under the control of a constitutive promoter were rescued from the embryonic lethality caused by the Jab1–/– allele and developed a myeloproliferative disorder in a gene dosage-dependent manner. Hematopoietic cells from the bone marrow of Jab1 transgenic mice had a significantly larger stem cell population and exhibited higher and transplantable proliferative potential. In contrast, Jab1+/– mice, which express ∼70% as much JAB1 protein as their wild-type littermates, showed inefficient hematopoiesis. Expression of the tumor suppressor p16INK4a was inversely correlated with that of JAB1, and the oncoprotein SMYD3, a newly identified JAB1 interactor, suppressed transcription of p16 in cooperation with JAB1. Thus, the expression and function of JAB1 are critical for the proliferation and maintenance of hematopoietic progenitors.

Collaboration


Dive into the Noriko Yoneda-Kato's collaboration.

Top Co-Authors

Avatar

Jun-ya Kato

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Kiichiro Tomoda

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Akihiro Yoshida

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Ikuko Nakamae

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yukinobu Arata

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Takashi Yokoyama

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Toshiaki Tanaka

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge