Network


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

Hotspot


Dive into the research topics where Norio Miyamura is active.

Publication


Featured researches published by Norio Miyamura.


Cell Cycle | 2009

Common light signaling pathways controlling DNA repair and circadian clock entrainment in zebrafish.

Jun Hirayama; Norio Miyamura; Yoshimi Uchida; Yoichi Asaoka; Reiko Honda; Kenji Sawanobori; Takeshi Todo; Takuro Yamamoto; Paolo Sassone-Corsi; Hiroshi Nishina

UV radiation causes a number of harmful events including growth delay, cell death and ultimately cancer. The reversal of such effects by concomitant exposure to visible light is a conserved mechanism which has been uncovered in many multi-cellular organisms. Here we show that light-dependent UV-tolerance is a cell autonomous phenomenon in zebrafish. In addition, we provide several lines of evidence indicating that light induction of 64PHR, a DNA repair enzyme, and the subsequent light-dependent DNA repair mediated by this enzyme are prerequisites for light-mediated UV tolerance. 64PHR is evolutionary related to and has a high degree of structural similarity to animal CRY, an essential circadian regulator. The zebrafish circadian clock is controlled by a cell-autonomous and light-dependent oscillator, where zCRY1a functions as an important mediator of light entrainment of the circadian clock. In this study, we show that light directly activates MAPK signaling cascades in zebrafish cells and we provide evidence that light-induced activation of these pathways controls the expression of two evolutionary-related genes, z64Phr and zCry1a, revealing that light-dependent DNA repair and the entrainment of circadian clock share common regulatory pathways.


Biochemical and Biophysical Research Communications | 2014

The PDZ-binding motif of Yes-associated protein is required for its co-activation of TEAD-mediated CTGF transcription and oncogenic cell transforming activity

Tadanori Shimomura; Norio Miyamura; Shoji Hata; Ryota Miura; Jun Hirayama; Hiroshi Nishina

YAP is a transcriptional co-activator that acts downstream of the Hippo signaling pathway and regulates multiple cellular processes, including proliferation. Hippo pathway-dependent phosphorylation of YAP negatively regulates its function. Conversely, attenuation of Hippo-mediated phosphorylation of YAP increases its ability to stimulate proliferation and eventually induces oncogenic transformation. The C-terminus of YAP contains a highly conserved PDZ-binding motif that regulates YAPs functions in multiple ways. However, to date, the importance of the PDZ-binding motif to the oncogenic cell transforming activity of YAP has not been determined. In this study, we disrupted the PDZ-binding motif in the YAP (5SA) protein, in which the sites normally targeted by Hippo pathway-dependent phosphorylation are mutated. We found that loss of the PDZ-binding motif significantly inhibited the oncogenic transformation of cultured cells induced by YAP (5SA). In addition, the increased nuclear localization of YAP (5SA) and its enhanced activation of TEAD-dependent transcription of the cell proliferation gene CTGF were strongly reduced when the PDZ-binding motif was deleted. Similarly, in mouse liver, deletion of the PDZ-binding motif suppressed nuclear localization of YAP (5SA) and YAP (5SA)-induced CTGF expression. Taken together, our results indicate that the PDZ-binding motif of YAP is critical for YAP-mediated oncogenesis, and that this effect is mediated by YAPs co-activation of TEAD-mediated CTGF transcription.


Biochemical and Biophysical Research Communications | 2011

Imaging mass spectrometry reveals characteristic changes in triglyceride and phospholipid species in regenerating mouse liver

Norio Miyamura; Takashi Nakamura; Naoko Goto-Inoue; Nobuhiro Zaima; Takahiro Hayasaka; Tokiwa Yamasaki; Shuji Terai; Isao Sakaida; Mitsutoshi Setou; Hiroshi Nishina

After partial hepatectomy (PH), regenerating liver accumulates unknown lipid species. Here, we analyzed lipids in murine liver and adipose tissues following PH by thin-layer chromatography (TLC), imaging mass spectrometry (IMS), and real-time RT-PCR. In liver, IMS revealed that a single TLC band comprised major 19 TG species. Similarly, IMS showed a single phospholipid TLC band to be major 13 species. In adipose tissues, PH induced changes to expression of genes regulating lipid metabolism. Finally, IMS of phosphatidylcholine species demonstrated distribution gradients in lobules that resembled hepatic zonation. IMS is thus a novel and power tool for analyzing lipid species with high resolution.


Scientific Reports | 2016

MDCK cells expressing constitutively active Yes-associated protein (YAP) undergo apical extrusion depending on neighboring cell status

Takanori Chiba; Erika Ishihara; Norio Miyamura; Rika Narumi; Mihoko Kajita; Yasuyuki Fujita; Akira Suzuki; Yoshihiro Ogawa; Hiroshi Nishina

Cell competition is a cell-cell interaction by which a cell compares its fitness to that of neighboring cells. The cell with the relatively lower fitness level is the “loser” and actively eliminated, while the cell with the relatively higher fitness level is the “winner” and survives. Recent studies have shown that cells with high Yes-associated protein (YAP) activity win cell competitions but the mechanism is unknown. Here, we report the unexpected finding that cells overexpressing constitutively active YAP undergo apical extrusion and are losers, rather than winners, in competitions with normal mammalian epithelial cells. Inhibitors of metabolism-related proteins such as phosphoinositide-3-kinase (PI3K), mammalian target of rapamycin (mTOR), or p70S6 kinase (p70S6K) suppressed this apical extrusion, as did knockdown of vimentin or filamin in neighboring cells. Interestingly, YAP-overexpressing cells switched from losers to winners when co-cultured with cells expressing K-Ras (G12V) or v-Src. Thus, the role of YAP in deciding cell competitions depends on metabolic factors and the status of neighboring cells.


Nature Communications | 2017

YAP determines the cell fate of injured mouse hepatocytes in vivo

Norio Miyamura; Shoji Hata; Tohru Itoh; Minoru Tanaka; Miki Nishio; Michiko Itoh; Yoshihiro Ogawa; Shuji Terai; Isao Sakaida; Akira Suzuki; Atsushi Miyajima; Hiroshi Nishina

The presence of senescent, transformed or damaged cells can impair tissue function or lead to tumorigenesis; therefore, organisms have evolved quality control mechanisms to eliminate them. Here, we show that YAP activation induced by inactivation of the Hippo pathway specifically in damaged hepatocytes promotes their selective elimination by using in vivo mosaic analysis in mouse liver. These damaged hepatocytes migrate into the hepatic sinusoids, undergo apoptosis and are engulfed by Kupffer cells. In contrast, YAP activation in undamaged hepatocytes leads to proliferation. Cellular stresses such as ethanol that damage both liver sinusoidal endothelial cells and hepatocytes switch cell fate from proliferation to migration/apoptosis in the presence of activated YAP. This involves the activation of CDC42 and Rac that regulate cell migration. Thus, we suggest that YAP acts as a stress sensor that induces elimination of injured cells to maintain tissue and organ homeostasis.


PLOS ONE | 2015

A Modified Murine Embryonic Stem Cell Test for Evaluating the Teratogenic Effects of Drugs on Early Embryogenesis.

Ruoxing Yu; Norio Miyamura; Yoshimi Okamoto-Uchida; Norie Arima; Mari Ishigami-Yuasa; Hiroyuki Kagechika; Hiroshi Nishina

Mammalian fetal development is easily disrupted by exogenous agents, making it essential to test new drug candidates for embryotoxicity and teratogenicity. To standardize the testing of drugs that might be used to treat pregnant women, the U.S. Food and Drug Administration (FDA) formulated special grade categories, labeled A, B, C, D and X, that define the level of risk associated with the use of a specific drug during pregnancy. Drugs in categories (Cat.) D and X are those with embryotoxic and/or teratogenic effects on humans and animals. However, which stages of pregnancy are affected by these agents and their molecular mechanisms are unknown. We describe here an embryonic stem cell test (EST) that classifies FDA pregnancy Cat.D and Cat.X drugs into 4 classes based on their differing effects on primitive streak formation. We show that ~84% of Cat.D and Cat.X drugs target this period of embryogenesis. Our results demonstrate that our modified EST can identify how a drug affects early embryogenesis, when it acts, and its molecular mechanism. Our test may thus be a useful addition to the drug safety testing armamentarium.


Scientific Reports | 2016

The mevalonate pathway regulates primitive streak formation via protein farnesylation

Yoshimi Okamoto-Uchida; Ruoxing Yu; Norio Miyamura; Norie Arima; Mari Ishigami-Yuasa; Hiroyuki Kagechika; Suguru Yoshida; Takamitsu Hosoya; Makiko Nawa; Takeshi Kasama; Yoichi Asaoka; Reiner Wimmer Alois; Ulrich Elling; Josef M. Penninger; Sachiko Nishina; Noriyuki Azuma; Hiroshi Nishina

The primitive streak in peri-implantation embryos forms the mesoderm and endoderm and controls cell differentiation. The metabolic cues regulating primitive streak formation remain largely unknown. Here we utilised a mouse embryonic stem (ES) cell differentiation system and a library of well-characterised drugs to identify these metabolic factors. We found that statins, which inhibit the mevalonate metabolic pathway, suppressed primitive streak formation in vitro and in vivo. Using metabolomics and pharmacologic approaches we identified the downstream signalling pathway of mevalonate and revealed that primitive streak formation requires protein farnesylation but not cholesterol synthesis. A tagging-via-substrate approach revealed that nuclear lamin B1 and small G proteins were farnesylated in embryoid bodies and important for primitive streak gene expression. In conclusion, protein farnesylation driven by the mevalonate pathway is a metabolic cue essential for primitive streak formation.


Scientific Reports | 2017

Age-dependent motor dysfunction due to neuron-specific disruption of stress-activated protein kinase MKK7

Tokiwa Yamasaki; Norie Deki-Arima; Asahito Kaneko; Norio Miyamura; Mamiko Iwatsuki; Masato Matsuoka; Noriko Fujimori-Tonou; Yoshimi Okamoto-Uchida; Jun Hirayama; Jamey D. Marth; Yuji Yamanashi; Hiroshi Kawasaki; Koji Yamanaka; Josef M. Penninger; Shigenobu Shibata; Hiroshi Nishina

Abstractc-Jun N-terminal kinase (JNK) is a member of the mitogen-activated protein kinase family and controls various physiological processes including apoptosis. A specific upstream activator of JNKs is the mitogen-activated protein kinase kinase 7 (MKK7). It has been reported that MKK7-JNK signaling plays an important regulatory role in neural development, however, post-developmental functions in the nervous system have not been elucidated. In this study, we generated neuron-specific Mkk7 knockout mice (MKK7 cKO), which impaired constitutive activation of JNK in the nervous system. MKK7 cKO mice displayed impaired circadian behavioral rhythms and decreased locomotor activity. MKK7 cKO mice at 8 months showed motor dysfunctions such as weakness of hind-limb and gait abnormality in an age-dependent manner. Axonal degeneration in the spinal cord and muscle atrophy were also observed, along with accumulation of the axonal transport proteins JNK-interacting protein 1 and amyloid beta precursor protein in the brains and spinal cords of MKK7 cKO mice. Thus, the MKK7-JNK signaling pathway plays important roles in regulating circadian rhythms and neuronal maintenance in the adult nervous system.


FEBS Open Bio | 2018

Effect of a neural relay on liver regeneration in mice: activation of serotonin release from the gastrointestinal tract

Ryosuke Inoue; Kenya Kamimura; Takuro Nagoya; Norihiro Sakai; Takeshi Yokoo; Ryo Goto; Kohei Ogawa; Yoko Shinagawa-Kobayashi; Yukari Watanabe‐Mori; Akira Sakamaki; Satoshi Abe; Hiroteru Kamimura; Norio Miyamura; Hiroshi Nishina; Shuji Terai

The development of therapeutic options to promote hepatic regeneration following severe liver injury is essential. While humoral factors have been reported as mechanisms of liver regeneration, the contributions of interorgan communication to liver regeneration have not been reported. In this study, we examined the effect of a neural relay on liver regeneration via activation of serotonin release from the gastrointestinal (GI) tract. Our results demonstrated that the afferent visceral nerve from the liver activates the efferent vagus nerve from the brain, leading to activation of serotonin release from the GI tract and contributing to liver regeneration. While it is difficult to apply these results directly to human health, we believe that this study may represent a step toward developing essential therapeutics to promote liver regeneration.


Molecular Cancer Research | 2017

Novel YAP1 Activator, Identified by Transcription-Based Functional Screen, Limits Multiple Myeloma Growth

Junichi Maruyama; Kazutoshi Inami; Fumiyoshi Michishita; Xinliang Jiang; Hiroaki Iwasa; Kentaro Nakagawa; Mari Ishigami-Yuasa; Hiroyuki Kagechika; Norio Miyamura; Jun Hirayama; Hiroshi Nishina; Daichi Nogawa; Kouhei Yamamoto; Yutaka Hata

Yes-associated protein 1 (YAP1) interacts with numerous transcription factors, including TEA-domain family proteins (TEAD) and p73. YAP1 is negatively regulated by the tumor suppressor Hippo pathway. In human cancers, the deregulation of the Hippo pathway and YAP1 gene amplification lead to the activation of YAP1, which induces epithelial–mesenchymal transition (EMT) and drug resistance. YAP1 inhibitors are expected to be useful in cancer therapy. On the other hand, in certain cancers, YAP1 upregulates p73-dependent gene transcription and behaves as a tumor suppressor. Moreover, as YAP1 regulates self-renewal and differentiation of tissue stem cells and plays an important role in tissue homeostasis, YAP1 activators may contribute to the regenerative medicine. With this in our mind, we screened for YAP1 activators by using human retinal pigment epithelial ARPE-19 cells expressing the TEAD-responsive fluorescence reporter under the coexpression of YAP1. From an extensive chemical compound library (n = 18,606) 47 candidate YAP1 activators were identified. These compounds were characterized to determine whether this assay provides bona fide YAP1 activators. Importantly, one YAP1 activator was effective against the human multiple myeloma IM-9 cells and chronic myeloid leukemia K562 cells. Implications: YAP1 activation limits growth, induces apoptosis, and may be useful at suppressing hematological cancers. Mol Cancer Res; 16(2); 197–211. ©2017 AACR.

Collaboration


Dive into the Norio Miyamura's collaboration.

Top Co-Authors

Avatar

Hiroshi Nishina

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Jun Hirayama

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroyuki Kagechika

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mari Ishigami-Yuasa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Shoji Hata

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Yoichi Asaoka

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge