Network


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

Hotspot


Dive into the research topics where Kentaro Nakagawa is active.

Publication


Featured researches published by Kentaro Nakagawa.


Cell Stem Cell | 2012

NuRD Suppresses Pluripotency Gene Expression to Promote Transcriptional Heterogeneity and Lineage Commitment

Nicola Reynolds; Paulina Latos; Antony Hynes-Allen; Remco Loos; Donna Leaford; Aoife O'Shaughnessy; Olukunbi Mosaku; Jason Signolet; Philip Brennecke; Tuezer Kalkan; Ita Costello; Peter Humphreys; William Mansfield; Kentaro Nakagawa; John Strouboulis; Axel Behrens; Paul Bertone; Brian Hendrich

Summary Transcriptional heterogeneity within embryonic stem cell (ESC) populations has been suggested as a mechanism by which a seemingly homogeneous cell population can initiate differentiation into an array of different cell types. Chromatin remodeling proteins have been shown to control transcriptional variability in yeast and to be important for mammalian ESC lineage commitment. Here we show that the Nucleosome Remodeling and Deacetylation (NuRD) complex, which is required for ESC lineage commitment, modulates both transcriptional heterogeneity and the dynamic range of a set of pluripotency genes in ESCs. In self-renewing conditions, the influence of NuRD at these genes is balanced by the opposing action of self-renewal factors. Upon loss of self-renewal factors, the action of NuRD is sufficient to silence transcription of these pluripotency genes, allowing cells to exit self-renewal. We propose that modulation of transcription levels by NuRD is key to maintaining the differentiation responsiveness of pluripotent cells.


Journal of Biochemistry | 2011

A cell-based assay to screen stimulators of the Hippo pathway reveals the inhibitory effect of dobutamine on the YAP-dependent gene transcription

Yijun Bao; Kentaro Nakagawa; Zeyu Yang; Mitsunobu Ikeda; Kanchanamala Withanage; Mari Ishigami-Yuasa; Yukiko Okuno; Shoji Hata; Hiroshi Nishina; Yutaka Hata

The mammalian Hippo pathway is composed of mammalian Ste20-like (MST) kinases and large tumour suppressor (LATS) kinases. Upon the activation of the pathway, MST kinases phosphorylate and activate LATS kinases, which in turn phosphorylate transcriptional co-activators, yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), recruit them to the cytosol from the nucleus and turn off cell cycle-promoting and anti-apoptotic gene transcriptions. Thus, the pathway restricts cell overgrowth and prevents tumourigenesis. Although a high cell density and stress signallings are known to activate the pathway, no specific stimulators are so far reported. As the dysfunction of the pathway is frequent in human cancers and correlates with poor prognosis, it is important to find out reagents that stimulate the pathway for not only basic research but also clinical medicine. We here developed a cell-based method of screening reagents that induce the recruitment of YAP to the cytosol. Using this method, we found that dobutamine inhibits the YAP-dependent gene transcription. Contrary to our expectations, the effect of dobutamine is independent of the Hippo pathway but our method opens the possibility to discover Hippo pathway stimulators or Hippo-independent YAP inhibitors.


Science Signaling | 2009

Hippo Pathway–Dependent and –Independent Roles of RASSF6

Mitsunobu Ikeda; Akira Kawata; Misa Nishikawa; Yuko Tateishi; Masato Yamaguchi; Kentaro Nakagawa; Susumu Hirabayashi; Yijun Bao; Shiho Hidaka; Yukio Hirata; Yutaka Hata

RASSF6 is both an inhibitor and a promoter of apoptosis, and its proapoptotic activity is regulated by the mammalian kinase MST2, a Hippo homolog. Apoptosis, With or Without Hippo The Hippo signaling pathway, named after the kinase Hippo, limits organ size without affecting patterning, and the components of this pathway are conserved from Drosophila to mammals. Drosophila RASSF (Ras association domain family) not only inhibits Hippo signaling and thus counters Hippo-mediated apoptosis, but also exhibits tumor suppressor function. Of the 10 mammalian RASSF isoforms, the best-characterized isoform, RASSF1A, activates the mammalian homologs of Hippo (the MST kinases), which contrasts with the role of dRASSF. Ikeda et al. show that RASSF6 represents another twist in the RASSF protein family. RASSF6 and MST2 are mutual inhibitors: Activation of MST2 disrupted binding of RASSF6 to MST2, enabling MST2 to induce apoptosis through a process dependent on the Hippo pathway and allowing RASSF6 to mediate apoptosis through a Hippo-independent pathway. These results suggest that mammalian RASSF isoforms may have divergent functions and may link the Hippo pathway to other signal transduction cascades. The Hippo pathway restricts cell growth and proliferation and promotes apoptosis to control organ size. The Drosophila melanogaster isoform of RASSF (Ras association domain family; dRASSF) antagonizes proapoptotic Hippo signaling by inhibiting the binding of the adaptor protein Salvador to the kinase Hippo. Paradoxically, however, dRASSF also functions as a tumor suppressor. In mammals, RASSF1A induces apoptosis by stimulating the mammalian Ste20–like kinases (MSTs) 1 and 2, which are Hippo homologs. Here, we characterize the interaction between MST2 and another mammalian RASSF isoform, RASSF6. When bound to MST2, RASSF6 inhibited MST2 activity to antagonize Hippo signaling. However, RASSF6 caused apoptosis when released from activated MST2 in a manner dependent on WW45, the mammalian Salvador homolog. Thus, RASSF6 antagonizes Hippo signaling and mediates apoptosis through a pathway that is parallel to the canonical Hippo pathway. Our findings suggest that activation of MST2 causes apoptosis through the Hippo pathway, as well as through a RASSF6-mediated pathway.


Nature | 2011

c-Jun N-terminal phosphorylation antagonises recruitment of the Mbd3/NuRD repressor complex.

Cristina Aguilera; Kentaro Nakagawa; Rocio Sancho; Atanu Chakraborty; Brian Hendrich; Axel Behrens

AP-1 (activator protein 1) activity is strongly induced in response to numerous signals, including growth factors, cytokines and extracellular stresses. The proto-oncoprotein c-Jun belongs to the AP-1 group of transcription factors and it is a crucial regulator of intestinal progenitor proliferation and tumorigenesis. An important mechanism of AP-1 stimulation is phosphorylation of c-Jun by the Jun amino-terminal kinases (JNKs). N-terminal phosphorylation of the c-Jun transactivation domain increases target gene transcription, but a molecular explanation was elusive. Here we show that unphosphorylated, but not N-terminally phosphorylated c-Jun, interacts with Mbd3 and thereby recruits the nucleosome remodelling and histone deacetylation (NuRD) repressor complex. Mbd3 depletion in colon cancer cells increased histone acetylation at AP-1-dependent promoters, which resulted in increased target gene expression. The intestinal stem cell marker lgr5 was identified as a novel target gene controlled by c-Jun/Mbd3. Gut-specific conditional deletion of mbd3 (mbd3ΔG/ΔG mice) stimulated c-Jun activity and increased progenitor cell proliferation. In response to inflammation, mdb3 deficiency resulted in colonic hyperproliferation and mbd3ΔG/ΔG mice showed markedly increased susceptibility to colitis-induced tumorigenesis. Notably, concomitant inactivation of a single allele of c-jun reverted physiological and pathological hyperproliferation, as well as the increased tumorigenesis in mbd3ΔG/ΔG mice. Thus the transactivation domain of c-Jun recruits Mbd3/NuRD to AP-1 target genes to mediate gene repression, and this repression is relieved by JNK-mediated c-Jun N-terminal phosphorylation.


Journal of Clinical Investigation | 2012

Cancer susceptibility and embryonic lethality in Mob1a/1b double-mutant mice

Miki Nishio; Koichi Hamada; Kohichi Kawahara; Masato Sasaki; Fumihito Noguchi; Shuhei Chiba; Kensaku Mizuno; Satoshi Suzuki; Youyi Dong; Masaaki Tokuda; Takumi Morikawa; Hiroki Hikasa; Jonathan T. Eggenschwiler; Norikazu Yabuta; Hiroshi Nojima; Kentaro Nakagawa; Yutaka Hata; Hiroshi Nishina; Koshi Mimori; Masaki Mori; Takehiko Sasaki; Tak W. Mak; Toru Nakano; Satoshi Itami; Akira Suzuki

Mps one binder 1a (MOB1A) and MOB1B are key components of the Hippo signaling pathway and are mutated or inactivated in many human cancers. Here we show that intact Mob1a or Mob1b is essential for murine embryogenesis and that loss of the remaining WT Mob1 allele in Mob1a(Δ/Δ)1b(tr/+) or Mob1a(Δ/+)1b(tr/tr) mice results in tumor development. Because most of these cancers resembled trichilemmal carcinomas, we generated double-mutant mice bearing tamoxifen-inducible, keratinocyte-specific homozygous-null mutations of Mob1a and Mob1b (kDKO mice). kDKO mice showed hyperplastic keratinocyte progenitors and defective keratinocyte terminal differentiation and soon died of malnutrition. kDKO keratinocytes exhibited hyperproliferation, apoptotic resistance, impaired contact inhibition, enhanced progenitor self renewal, and increased centrosomes. Examination of Hippo pathway signaling in kDKO keratinocytes revealed that loss of Mob1a/b altered the activities of the downstream Hippo mediators LATS and YAP1. Similarly, YAP1 was activated in some human trichilemmal carcinomas, and some of these also exhibited MOB1A/1B inactivation. Our results clearly demonstrate that MOB1A and MOB1B have overlapping functions in skin homeostasis, and exert their roles as tumor suppressors by regulating downstream elements of the Hippo pathway.


Oncogene | 2008

Threonine 74 of MOB1 is a putative key phosphorylation site by MST2 to form the scaffold to activate nuclear Dbf2-related kinase 1.

Susumu Hirabayashi; Kentaro Nakagawa; Kazutaka Sumita; S Hidaka; T Kawai; Mitsunobu Ikeda; Akira Kawata; Kikuo Ohno; Yutaka Hata

Mammalian nuclear Dbf2-related (NDR) kinases (LATS1 and 2, NDR1 and 2) play a role in cell proliferation, apoptosis and morphological changes. These kinases are regulated by mammalian sterile 20-like kinases (MSTs) and Mps one binder (MOB) 1. Okadaic acid (OA), which activates MST2, facilitates the complex formation of MOB1, MST2 and NDR1 in HEK293FT cells. The in vitro biochemical study demonstrates the phosphorylation of MOB1 by MST2. The phosphorylated MOB1 alone is capable to partially activate NDR1 in vitro, but MST2 is also required for the full activation. The knockdown of MOB1 or MST2 abolishes the OA-induced NDR1 activation in HEK293FT cells. Among MOB1 mutants, in which each serine or threonine residue is replaced with alanine, MOB1 T74A and T181A mutants fail to activate NDR1. Thr74, but not Thr181, is phosphorylated by MST2 in vitro, although MOB1 is also phosphorylated by MST2 at other site(s). The interaction of MOB1 T74A with NDR1 is barely enhanced by OA treatment. These findings indicate that the phosphorylation of MOB1 at Thr74 by MST2 is essential to make a complex of MOB1, MST2 and NDR1, and to fully activate NDR1.


Journal of Biological Chemistry | 2012

A Novel Acetylation Cycle of Transcription Co-activator Yes-associated Protein That Is Downstream of Hippo Pathway Is Triggered in Response to SN2 Alkylating Agents

Shoji Hata; Jun Hirayama; Hiroaki Kajiho; Kentaro Nakagawa; Yutaka Hata; Toshiaki Katada; Makoto Furutani-Seiki; Hiroshi Nishina

Background: YAP is a target molecule of the Hippo pathway. Results: YAP acetylation and deacetylation were mediated by CBP/p300 acetyltransferase and SIRT1 deacetylase, respectively. Conclusion: A YAP acetylation/deacetylation cycle is located downstream of the Hippo pathway. Significance: The discovery that YAP undergoes an acetylation cycle advances our understanding of YAP functions. Yes-associated protein (YAP) is a transcriptional co-activator that acts downstream of the Hippo signaling pathway and regulates multiple cellular processes. Although cytoplasmic retention of YAP is known to be mediated by Hippo pathway-dependent phosphorylation, post-translational modifications that regulate YAP in the nucleus remain unclear. Here we report the discovery of a novel cycle of acetylation/deacetylation of nuclear YAP induced in response to SN2 alkylating agents. We show that after treatment of cells with the SN2 alkylating agent methyl methanesulfonate, YAP phosphorylation mediated by the Hippo pathway is markedly reduced, leading to nuclear translocation of YAP and its acetylation. This YAP acetylation occurs on specific and highly conserved C-terminal lysine residues and is mediated by the nuclear acetyltransferases CBP (CREB binding protein) and p300. Conversely, the nuclear deacetylase SIRT1 is responsible for YAP deacetylation. Intriguingly, we found that YAP acetylation is induced specifically by SN2 alkylating agents and not by other DNA-damaging stimuli. These results identify a novel YAP acetylation cycle that occurs in the nucleus downstream of the Hippo pathway. Intriguingly, our findings also indicate that YAP acetylation is involved in responses to a specific type of DNA damage.


Cancer Research | 2012

The RASSF3 Candidate Tumor Suppressor Induces Apoptosis and G1–S Cell-Cycle Arrest via p53

Takumi Kudo; Mitsunobu Ikeda; Misa Nishikawa; Zeyu Yang; Kikuo Ohno; Kentaro Nakagawa; Yutaka Hata

RASSF3 is the smallest member of the RASSF family of proteins that function as tumor suppressors. Unlike other members of this important family, the mechanisms through which RASSF3 suppresses tumor formation remain unknown. Here, we show that RASSF3 expression induces p53-dependent apoptosis and its depletion attenuates DNA damage-induced apoptosis. We found that RASSF3-induced apoptosis depended upon p53 expression. Exogenous expression of RASSF3 induced G(1)-S arrest, which was also p53 dependent. In contrast, loss of RASSF3 promoted cell-cycle progression, abrogated UVB- and VP-16-induced G(1)-S arrest, decreased p53 protein and target gene expression, and prevented DNA repair. RASSF3 was shown to directly interact with and facilitate the ubiquitination of MDM2, the E3 ligase that targets p53 for degradation, thereby increasing p53 stabilization. Together, our findings show the tumor suppressor activity of RASSF3, which occurs through p53 stabilization and regulation of apoptosis and the cell cycle.


Molecular and Cellular Biology | 2014

Screening with a Novel Cell-Based Assay for TAZ Activators Identifies a Compound That Enhances Myogenesis in C2C12 Cells and Facilitates Muscle Repair in a Muscle Injury Model

Zeyu Yang; Kentaro Nakagawa; Aradhan Sarkar; Junichi Maruyama; Hiroaki Iwasa; Yijun Bao; Mari Ishigami-Yuasa; Shigeru Ito; Hiroyuki Kagechika; Shoji Hata; Hiroshi Nishina; Shinya Abe; Masanobu Kitagawa; Yutaka Hata

ABSTRACT The transcriptional coactivator with a PDZ-binding motif (TAZ) cooperates with various transcriptional factors and plays various roles. Immortalized human mammalian epithelial MCF10A cells form spheres when TAZ is overexpressed and activated. We developed a cell-based assay using sphere formation by TAZ-expressing MCF10A cells as a readout to screen 18,458 chemical compounds for TAZ activators. Fifty compounds were obtained, and 47 were confirmed to activate the TAZ-dependent TEAD-responsive reporter activity in HEK293 cells. We used the derived subset of compounds as a TAZ activator candidate minilibrary and searched for compounds that promote myogenesis in mouse C2C12 myoblast cells. In this study, we focused on one compound, IBS008738. IBS008738 stabilizes TAZ, increases the unphosphorylated TAZ level, enhances the association of MyoD with the myogenin promoter, upregulates MyoD-dependent gene transcription, and competes with myostatin in C2C12 cells. TAZ knockdown verifies that the effect of IBS008738 depends on endogenous TAZ in C2C12 cells. IBS008738 facilitates muscle repair in cardiotoxin-induced muscle injury and prevents dexamethasone-induced muscle atrophy. Thus, this cell-based assay is useful to identify TAZ activators with a variety of cellular outputs. Our findings also support the idea that TAZ is a potential therapeutic target for muscle atrophy.


Journal of Biological Chemistry | 2013

The RASSF6 tumor suppressor protein regulates apoptosis and the cell cycle via MDM2 protein and p53 protein.

Hiroaki Iwasa; Takumi Kudo; Sainawaer Maimaiti; Mitsunobu Ikeda; Jun-ichi Maruyama; Kentaro Nakagawa; Yutaka Hata

Background: RASSF6 is a proapoptotic protein related to the Hippo pathway. Results: RASSF6 interacts with MDM2 and stabilizes p53. Conclusion: RASSF6 induces apoptosis and cell cycle arrest via p53. Significance: Our work supports the importance of the C-terminal RASSF-MDM2-p53 axis. Ras association domain family (RASSF) 6 is a member of the C-terminal RASSF proteins such as RASSF1A and RASSF3. RASSF6 is involved in apoptosis in various cells under miscellaneous conditions, but it remains to be clarified how RASSF6 exerts tumor-suppressive roles. We reported previously that RASSF3 facilitates the degradation of MDM2, a major E3 ligase of p53, and stabilizes p53 to function as a tumor suppressor. In this study, we demonstrate that RASSF6 overexpression induces G1/S arrest in p53-positive cells. Its depletion prevents UV- and VP-16-induced apoptosis and G1/S arrest in HCT116 and U2OS cells. RASSF6-induced apoptosis partially depends on p53. RASSF6 binds MDM2 and facilitates its ubiquitination. RASSF6 depletion blocks the increase of p53 in response to UV exposure and up-regulation of p53 target genes. RASSF6 depletion delays DNA repair in UV- and VP-16-treated cells and increases polyploid cells after VP-16 treatment. These findings indicate that RASSF6 stabilizes p53, regulates apoptosis and the cell cycle, and functions as a tumor suppressor. Together with the previous reports regarding RASSF1A and RASSF3, the stabilization of p53 may be the common function of the C-terminal RASSF proteins.

Collaboration


Dive into the Kentaro Nakagawa's collaboration.

Top Co-Authors

Avatar

Yutaka Hata

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Hiroaki Iwasa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Mitsunobu Ikeda

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Nishina

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Hiroyuki Kagechika

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Junichi Maruyama

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Mari Ishigami-Yuasa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Takumi Kudo

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kanchanamala Withanage

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Shigeru Ito

Tokyo Medical and Dental University

View shared research outputs
Researchain Logo
Decentralizing Knowledge