2024/02/02 更新

写真a

ムラタ ヒトシ
村田 等
MURATA Hitoshi
所属
医歯薬学域 講師
職名
講師
外部リンク

学位

  • 工学(修士) ( 岡山大学 )

  • 工学(博士) ( 岡山大学 )

研究キーワード

  • SARM1

  • タンパク質工学

  • 細胞生物学

  • PINK1

  • ミトコンドリア

  • Protein Engneering

  • Cell Biology

  • Mitochondria

  • 神経変性疾患

  • パーキンソン病

  • NAD

研究分野

  • ライフサイエンス / 分子生物学

  • ライフサイエンス / 機能生物化学

学歴

  • 岡山大学   The Graduate School of Natural Science and Technology   (Doctor Course) Division of Chemistry and Biochemistry

    - 2008年

      詳細を見る

    国名: 日本国

    researchmap

  • 岡山大学   The Graduate School of Natural Science and Technology  

    - 2005年

      詳細を見る

    国名: 日本国

    researchmap

  • 岡山大学   Faculty of Engineering   Department of Bioscience and Biotechnology

    - 2003年

      詳細を見る

    国名: 日本国

    researchmap

経歴

  • 岡山大学 大学院医歯薬学総合研究科   講師

    2016年 - 現在

      詳細を見る

  • 岡山大学 大学院医歯薬学総合研究科   助教

    2010年 - 2016年

      詳細を見る

  • 岡山大学 大学院医歯薬学総合研究科   特任助教

    2008年 - 2010年

      詳細を見る

所属学協会

  • 米国細胞生物学会

    2015年 - 現在

  • 日本神経科学学会

      詳細を見る

  • 日本分子生物学会

      詳細を見る

  • 日本生化学会

      詳細を見る

  • 日本組織培養学会

      詳細を見る

 

論文

  • Phosphorylated SARM1 is involved in the pathological process of rotenone-induced neurodegeneration. 国際誌

    Hitoshi Murata, May Tha Zin Phoo, Toshiki Ochi, Nahoko Tomonobu, Ken-Ichi Yamamoto, Rie Kinoshita, Ikuko Miyazaki, Masahiro Nishibori, Masato Asanuma, Masakiyo Sakaguchi

    Journal of biochemistry   2023年9月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Sterile alpha and Toll/interleukin receptor motif-containing protein 1 (SARM1) is a NAD+ hydrolase that plays a key role in axonal degeneration and neuronal cell death. We reported that c-Jun N-terminal kinase (JNK) activates SARM1 through phosphorylation at Ser-548. The importance of SARM1 phosphorylation in the pathological process of Parkinson's disease (PD) has not been determined. We thus conducted the present study by using rotenone (an inducer of PD-like pathology) and neurons derived from induced pluripotent stem cells (iPSCs) from healthy donors and a patient with familial PD PARK2 (FPD2). The results showed that compared to the healthy neurons, FPD2 neurons were more vulnerable to rotenone-induced stress and had higher levels of SARM1 phosphorylation. Similar cellular events were obtained when we used PARK2-knockdown neurons derived from healthy donor iPSCs. These events in both types of PD-model neurons were suppressed in neurons treated with JNK inhibitors, Ca2+-signal inhibitors, or by a SARM1-knockdown procedure. The degenerative events were enhanced in neurons overexpressing wild-type SARM1 and conversely suppressed in neurons overexpressing the SARM1-S548A mutant. We also detected elevated SARM1 phosphorylation in the midbrain of PD-model mice. The results indicate that phosphorylated SARM1 plays an important role in the pathological process of rotenone-induced neurodegeneration.

    DOI: 10.1093/jb/mvad068

    PubMed

    researchmap

  • STAT1/3 signaling suppresses axon degeneration and neuronal cell death through regulation of NAD+-biosynthetic and consuming enzymes. 国際誌

    Hitoshi Murata, Yu Yasui, Kazuma Oiso, Toshiki Ochi, Nahoko Tomonobu, Ken-Ichi Yamamoto, Rie Kinoshita, Masakiyo Sakaguchi

    Cellular signalling   108   110717 - 110717   2023年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Nicotinamide adenine dinucleotide (NAD)+-biosynthetic and consuming enzymes are involved in various intracellular events through the regulation of NAD+ metabolism. Recently, it has become clear that alterations in the expression of NAD+-biosynthetic and consuming enzymes contribute to the axonal stability of neurons. We explored soluble bioactive factor(s) that alter the expression of NAD+-metabolizing enzymes and found that cytokine interferon (IFN)-γ increased the expression of nicotinamide nucleotide adenylyltransferase 2 (NMNAT2), an NAD+-biosynthetic enzyme. IFN-γ activated signal transducers and activators of transcription 1 and 3 (STAT1/3) followed by c-Jun N-terminal kinase (JNK) suppression. As a result, STAT1/3 increased the expression of NMNAT2 at both mRNA and protein levels in a dose- and time-dependent manner and, at the same time, suppressed activation of sterile alpha and Toll/interleukin receptor motif-containing 1 (SARM1), an NAD+-consuming enzyme, and increased intracellular NAD+ levels. We examined the protective effect of STAT1/3 signaling against vincristine-mediated cell injury as a model of chemotherapy-induced peripheral neuropathy (CIPN), in which axonal degeneration is involved in disease progression. We found that IFN-γ-mediated STAT1/3 activation inhibited vincristine-induced downregulation of NMNAT2 and upregulation of SARM1 phosphorylation, resulting in modest suppression of subsequent neurite degradation and cell death. These results indicate that STAT1/3 signaling induces NMNAT2 expression while simultaneously suppressing SARM1 phosphorylation, and that both these actions contribute to suppression of axonal degeneration and cell death.

    DOI: 10.1016/j.cellsig.2023.110717

    PubMed

    researchmap

  • Novel extracellular role of REIC/Dkk-3 protein in PD-L1 regulation in cancer cells. 国際誌

    Yuma Gohara, Nahoko Tomonobu, Rie Kinoshita, Junichiro Futami, Léna Audebert, Youyi Chen, Ni Luh Gede Yoni Komalasari, Fan Jiang, Chikako Yoshizawa, Hitoshi Murata, Ken-Ichi Yamamoto, Masami Watanabe, Hiromi Kumon, Masakiyo Sakaguchi

    Journal of molecular medicine (Berlin, Germany)   101 ( 4 )   431 - 447   2023年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    The adenovirus-REIC/Dkk-3 expression vector (Ad-REIC) has been the focus of numerous clinical studies due to its potential for the quenching of cancers. The cancer-suppressing mechanisms of the REIC/DKK-3 gene depend on multiple pathways that exert both direct and indirect effects on cancers. The direct effect is triggered by REIC/Dkk-3-mediated ER stress that causes cancer-selective apoptosis, and the indirect effect can be classified in two ways: (i) induction, by Ad-REIC-mis-infected cancer-associated fibroblasts, of the production of IL-7, an important activator of T cells and NK cells, and (ii) promotion, by the secretory REIC/Dkk-3 protein, of dendritic cell polarization from monocytes. These unique features allow Ad-REIC to exert effective and selective cancer-preventative effects in the manner of an anticancer vaccine. However, the question of how the REIC/Dkk-3 protein leverages anticancer immunity has remained to be answered. We herein report a novel function of the extracellular REIC/Dkk-3-namely, regulation of an immune checkpoint via modulation of PD-L1 on the cancer-cell surface. First, we identified novel interactions of REIC/Dkk-3 with the membrane proteins C5aR, CXCR2, CXCR6, and CMTM6. These proteins all functioned to stabilize PD-L1 on the cell surface. Due to the dominant expression of CMTM6 among the proteins in cancer cells, we next focused on CMTM6 and observed that REIC/Dkk-3 competed with CMTM6 for PD-L1, thereby liberating PD-L1 from its complexation with CMTM6. The released PD-L1 immediately underwent endocytosis-mediated degradation. These results will enhance our understanding of not only the physiological nature of the extracellular REIC/Dkk-3 protein but also the Ad-REIC-mediated anticancer effects. KEY MESSAGES: • REIC/Dkk-3 protein effectively suppresses breast cancer progression through an acceleration of PD-L1 degradation. • PD-L1 stability on the cancer cell membrane is kept high by binding with mainly CMTM6. • Competitive binding of REIC/Dkk-3 protein with CMTM6 liberates PD-L1, leading to PD-L1 degradation.

    DOI: 10.1007/s00109-023-02292-w

    PubMed

    researchmap

  • LOXL1 and LOXL4 are novel target genes of the Zn2+-bound form of ZEB1 and play a crucial role in the acceleration of invasive events in triple-negative breast cancer cells

    Daisuke Hirabayashi, Ken-ichi Yamamoto, Akihiro Maruyama, Nahoko Tomonobu, Rie Kinoshita, Youyi Chen, Ni Luh Gede Yoni Komalasari, Hitoshi Murata, Yuma Gohara, Fan Jiang, Jin Zhou, I Made Winarsa Ruma, I Wayan Sumardika, Akira Yamauchi, Futoshi Kuribayashi, Shinichi Toyooka, Yusuke Inoue, Masakiyo Sakaguchi

    Frontiers in Oncology   13   2023年2月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Frontiers Media SA  

    Background

    EMT has been proposed to be a crucial early event in cancer metastasis. EMT is rigidly regulated by the action of several EMT-core transcription factors, particularly ZEB1. We previously revealed an unusual role of ZEB1 in the S100A8/A9-mediated metastasis in breast cancer cells that expressed ZEB1 at a significant level and showed that the ZEB1 was activated on the MCAM-downstream pathway upon S100A8/A9 binding. ZEB1 is well known to require Zn2+ for its activation based on the presence of several Zn-finger motifs in the transcription factor. However, how Zn2+-binding works on the pleiotropic role of ZEB1 through cancer progression has not been fully elucidated.

    Methods

    We established the engineered cells, MDA-MB-231 MutZEB1 (MDA-MutZEB1), that stably express MutZEB1 (ΔZn). The cells were then evaluated in vitro for their invasion activities. Finally, an RNA-Seq analysis was performed to compare the gene alteration profiles of the established cells comprehensively.

    Results

    MDA-MutZEB1 showed a significant loss of the EMT, ultimately stalling the invasion. Inclusive analysis of the transcription changes after the expression of MutZEB1 (ΔZn) in MDA-MB-231 cells revealed the significant downregulation of LOX family genes, which are known to play a critical role in cancer metastasis. We found that LOXL1 and LOXL4 remarkably enhanced cancer invasiveness among the LOX family genes with altered expression.

    Conclusions

    These findings indicate that ZEB1 potentiates Zn2+-mediated transcription of plural EMT-relevant factors, including LOXL1 and LOXL4, whose upregulation plays a critical role in the invasive dissemination of breast cancer cells.

    DOI: 10.3389/fonc.2023.1142886

    researchmap

  • Lysyl oxidase-like 4 exerts an atypical role in breast cancer progression that is dependent on the enzymatic activity that targets the cell-surface annexin A2. 国際誌

    Ni Luh Gede Yoni Komalasari, Nahoko Tomonobu, Rie Kinoshita, Youyi Chen, Yoshihiko Sakaguchi, Yuma Gohara, Fan Jiang, Ken-Ich Yamamoto, Hitoshi Murata, I Made Winarsa Ruma, I Wayan Sumardika, Jin Zhou, Akira Yamauchi, Futoshi Kuribayashi, Yusuke Inoue, Shinichi Toyooka, Masakiyo Sakaguchi

    Frontiers in oncology   13   1142907 - 1142907   2023年

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    BACKGROUND: LOX family members are reported to play pivotal roles in cancer. Unlike their enzymatic activities in collagen cross-linking, their precise cancer functions are unclear. We revealed that LOXL4 is highly upregulated in breast cancer cells, and we thus sought to define an unidentified role of LOXL4 in breast cancer. METHODS: We established the MDA-MB-231 sublines MDA-MB-231-LOXL4 mutCA and -LOXL4 KO, which stably overexpress mutant LOXL4 that loses its catalytic activity and genetically ablates the intrinsic LOXL4 gene, respectively. In vitro and in vivo evaluations of these cells' activities of cancer outgrowth were conducted by cell-based assays in cultures and an orthotopic xenograft model, respectively. The new target (s) of LOXL4 were explored by the MS/MS analytic approach. RESULTS: Our in vitro results revealed that both the overexpression of mutCA and the KO of LOXL4 in cells resulted in a marked reduction of cell growth and invasion. Interestingly, the lowered cellular activities observed in the engineered cells were also reflected in the mouse model. We identified a novel binding partner of LOXL4, i.e., annexin A2. LOXL4 catalyzes cell surface annexin A2 to achieve a cross-linked multimerization of annexin A2, which in turn prevents the internalization of integrin β-1, resulting in the locking of integrin β-1 on the cell surface. These events enhance the promotion of cancer cell outgrowth. CONCLUSIONS: LOXL4 has a new role in breast cancer progression that occurs via an interaction with annexin A2 and integrin β-1 on the cell surface.

    DOI: 10.3389/fonc.2023.1142907

    PubMed

    researchmap

  • Toll-like receptor 4 promotes bladder cancer progression upon S100A8/A9 binding, which requires TIRAP-mediated TPL2 activation. 国際誌

    Acosta Gonzalez Herik Rodrigo, Nahoko Tomonobu, Haruka Yoneda, Rie Kinoshita, Yosuke Mitsui, Takuya Sadahira, Shin-Ichi Terawaki, Yuma Gohara, Ni Luh Gede Yoni Komalasari, Fan Jiang, Hitoshi Murata, Ken-Ichi Yamamoto, Junichiro Futami, Akira Yamauchi, Futoshi Kuribayashi, Yusuke Inoue, Eisaku Kondo, Shinichi Toyooka, Masahiro Nishibori, Masami Watanabe, Yasutomo Nasu, Masakiyo Sakaguchi

    Biochemical and biophysical research communications   634   83 - 91   2022年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Bladder cancer is an often widely disseminated and deadly cancer. To block the malignant outgrowth of bladder cancer, we must elucidate the molecular-level characteristics of not only bladder cancer cells but also their surrounding milieu. As part of this effort, we have long been studying extracellular S100A8/A9, which is elevated by the inflammation associated with certain cancers. Extracellularly enriched S100A8/A9 can hasten a shift to metastatic transition in multiple types of cancer cells. Intriguingly, high-level S100A8/A9 has been detected in the urine of bladder-cancer patients, and the level increases with the stage of malignancy. Nonetheless, S100A8/A9 has been investigated mainly as a potential biomarker of bladder cancers, and there have been no investigations of its role in bladder-cancer growth and metastasis. We herein report that extracellular S100A8/A9 induces upregulation of growth, migration and invasion in bladder cancer cells through its binding with cell-surface Toll-like receptor 4 (TLR4). Our molecular analysis revealed the TLR4 downstream signal that accelerates such cancer cell events. Tumor progression locus 2 (TPL2) was a key factor facilitating the aggressiveness of cancer cells. Upon binding of S100A8/A9 with TLR4, TPL2 activation was enhanced by an action with a TLR4 adaptor molecule, TIR domain-containing adaptor protein (TIRAP), which in turn led to activation of the mitogen-activated protein kinase (MAPK) cascade of TPL2. Finally, we showed that sustained inhibition of TLR4 in cancer cells effectively dampened cancer survival in vivo. Collectively, our results indicate that the S100A8/A9-TLR4-TPL2 axis influences the growth, survival, and invasive motility of bladder cancer cells.

    DOI: 10.1016/j.bbrc.2022.09.116

    PubMed

    researchmap

  • Histidine-Rich Glycoprotein Suppresses the S100A8/A9-Mediated Organotropic Metastasis of Melanoma Cells

    Nahoko Tomonobu, Rie Kinoshita, Hidenori Wake, Yusuke Inoue, I Made Winarsa Ruma, Ken Suzawa, Yuma Gohara, Ni Luh Gede Yoni Komalasari, Fan Jiang, Hitoshi Murata, Ken-ichi Yamamoto, I Wayan Sumardika, Youyi Chen, Junichiro Futami, Akira Yamauchi, Futoshi Kuribayashi, Eisaku Kondo, Shinichi Toyooka, Masahiro Nishibori, Masakiyo Sakaguchi

    International Journal of Molecular Sciences   23 ( 18 )   10300 - 10300   2022年9月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:MDPI AG  

    The dissection of the complex multistep process of metastasis exposes vulnerabilities that could be exploited to prevent metastasis. To search for possible factors that favor metastatic outgrowth, we have been focusing on secretory S100A8/A9. A heterodimer complex of the S100A8 and S100A9 proteins, S100A8/A9 functions as a strong chemoattractant, growth factor, and immune suppressor, both promoting the cancer milieu at the cancer-onset site and cultivating remote, premetastatic cancer sites. We previously reported that melanoma cells show lung-tropic metastasis owing to the abundant expression of S100A8/A9 in the lung. In the present study, we addressed the question of why melanoma cells are not metastasized into the brain at significant levels in mice despite the marked induction of S100A8/A9 in the brain. We discovered the presence of plasma histidine-rich glycoprotein (HRG), a brain-metastasis suppression factor against S100A8/A9. Using S100A8/A9 as an affinity ligand, we searched for and purified the binding plasma proteins of S100A8/A9 and identified HRG as the major protein on mass spectrometric analysis. HRG prevents the binding of S100A8/A9 to the B16-BL6 melanoma cell surface via the formation of the S100A8/A9 complex. HRG also inhibited the S100A8/A9-induced migration and invasion of A375 melanoma cells. When we knocked down HRG in mice bearing skin melanoma, metastasis to both the brain and lungs was significantly enhanced. The clinical examination of plasma S100A8/A9 and HRG levels showed that lung cancer patients with brain metastasis had higher S100A8/A9 and lower HRG levels than nonmetastatic patients. These results suggest that the plasma protein HRG strongly protects the brain and lungs from the threat of melanoma metastasis.

    DOI: 10.3390/ijms231810300

    researchmap

  • The heterodimer S100A8/A9 is a potent therapeutic target for idiopathic pulmonary fibrosis. 国際誌

    Kota Araki, Rie Kinoshita, Nahoko Tomonobu, Yuma Gohara, Shuta Tomida, Yuta Takahashi, Satoru Senoo, Akihiko Taniguchi, Junko Itano, Ken-Ichi Yamamoto, Hitoshi Murata, Ken Suzawa, Kazuhiko Shien, Hiromasa Yamamoto, Mikio Okazaki, Seiichiro Sugimoto, Kouichi Ichimura, Masahiro Nishibori, Nobuaki Miyahara, Shinichi Toyooka, Masakiyo Sakaguchi

    Journal of molecular medicine (Berlin, Germany)   99 ( 1 )   131 - 145   2021年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    In patients with interstitial pneumonia, pulmonary fibrosis is an irreversible condition that can cause respiratory failure. Novel treatments for pulmonary fibrosis are necessary. Inflammation is thought to activate lung fibroblasts, resulting in pulmonary fibrosis. Of the known inflammatory molecules, we have focused on S100A8/A9 from the onset of inflammation to the subsequent progression of inflammation. Our findings confirmed the high expression of S100A8/A9 in specimens from patients with pulmonary fibrosis. An active role of S100A8/A9 was demonstrated not only in the proliferation of fibroblasts but also in the fibroblasts' differentiation to myofibroblasts (the active form of fibroblasts). S100A8/A9 also forced fibroblasts to upregulate the production of collagen. These effects were induced via the receptor of S100A8/A9, i.e., the receptor for advanced glycation end products (RAGE), on fibroblasts. The anti-S100A8/A9 neutralizing antibody inhibited the effects of S100A8/A9 on fibroblasts and suppressed the progression of fibrosis in bleomycin (BLM)-induced pulmonary fibrosis mouse model. Our findings strongly suggest a crucial role of S100A8/A9 in pulmonary fibrosis and the usefulness of S100A8/A9-targeting therapy for fibrosis interstitial pneumonia. HIGHLIGHTS: S100A8/A9 level is highly upregulated in the IPF patients' lungs as well as the blood. S100A8/A9 promotes not only the growth of fibroblasts but also differentiation to myofibroblasts. The cell surface RAGE acts as a crucial receptor to the extracellular S100A8/A9 in fibroblasts. The anti-S100A8/A9 antibody effectively suppresses the progression of IPF in a mouse model. In idiopathic pulmonary fibrosis (IPF), S100A8/A9, a heterodimer composed of S100A8 and S100A9 proteins, plays a crucial role in the onset of inflammation and the subsequent formation of a feed-forward inflammatory loop that promotes fibrosis. (1) The local, pronounced increase in S100A8/A9 in the injured inflammatory lung region-which is provided mainly by the activated neutrophils and macrophages-exerts strong inflammatory signals accompanied by dozens of inflammatory soluble factors including cytokines, chemokines, and growth factors that further act to produce and secrete S100A8/A9, eventually making a sustainable inflammatory circuit that supplies an indefinite presence of S100A8/A9 in the extracellular space with a mal-increased level. (2) The elevated S100A8/A9 compels fibroblasts to activate through receptor for advanced glycation end products (RAGE), one of the major S100A8/A9 receptors, resulting in the activation of NFκB, leading to fibroblast mal-events (e.g., elevated cell proliferation and transdifferentiation to myofibroblasts) that actively produce not only inflammatory cytokines but also collagen matrices. (3) Finally, the S100A8/A9-derived activation of lung fibroblasts under a chronic inflammation state leads to fibrosis events and constantly worsens fibrosis in the lung. Taken together, these findings suggest that the extracellular S100A8/A9 heterodimer protein is a novel mainstay soluble factor for IPF that exerts many functions as described above (1-3). Against this background, we herein applied the developed S100A8/A9 neutralizing antibody to prevent IPF. The IPF imitating lung fibrosis in an IPF mouse model was effectively blocked by treatment with the antibody, leading to enhanced survival. The developed S100A8/A9 antibody, as an innovative novel biologic, may help shed light on the difficulties encountered with IPF therapy in clinical settings.

    DOI: 10.1007/s00109-020-02001-x

    PubMed

    researchmap

  • Neuroplastinβ-mediated upregulation of solute carrier family 22 member 18 antisense (SLC22A18AS) plays a crucial role in the epithelial-mesenchymal transition, leading to lung cancer cells' enhanced motility 査読

    Karolina Bajkowska, I. Wayan Sumardika, Nahoko Tomonobu, Youyi Chen, Ken-ichi Yamamoto, Rie Kinoshita, Hitoshi Murata, Ni Luh Gede Yoni Komalasari, Fan Jiang, Akira Yamauchi, I. Made Winarsa Ruma, Carlos Ichiro Kasano-Camones, Yusuke Inoue, Masakiyo Sakaguchi

    Biochemistry and Biophysics Reports   22   100768 - 100768   2020年7月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Elsevier BV  

    DOI: 10.1016/j.bbrep.2020.100768

    researchmap

  • Xylitol acts as an anticancer monosaccharide to induce selective cancer death via regulation of the glutathione level. 国際誌

    Nahoko Tomonobu, Ni Luh Gede Yoni Komalasari, I Wayan Sumardika, Fan Jiang, Youyi Chen, Ken-Ichi Yamamoto, Rie Kinoshita, Hitoshi Murata, Yusuke Inoue, Masakiyo Sakaguchi

    Chemico-biological interactions   324   109085 - 109085   2020年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Herbal medicines and their bioactive compounds are increasingly being recognized as useful drugs for cancer treatments. The parasitic fungus Cordyceps militaris is an attractive anticancer herbal since it shows very powerful anticancer activity due to its phytocompound cordycepin. We previously discovered and reported that a high amount of xylitol is present in Cordyceps militaris extract, and that xylitol unexpectedly showed anticancer activity in a cancer-selective manner. We thus hypothesized that xylitol could become a useful supplement to help prevent various cancers, if we can clarify the specific machinery by which xylitol induces cancer cell death. It is also unclear whether xylitol acts on cancer suppression in vivo as well as in vitro. Here we show for the first time that induction of the glutathione-degrading enzyme CHAC1 is the main cause of xylitol-induced apoptotic cell death in cancer cells. The induction of CHAC1 is required for the endoplasmic reticulum (ER) stress that is triggered by xylitol in cancer cells, and is linked to a second induction of oxidative stress in the treated cells, and eventually leads to apoptotic cell death. Our in vivo approach also demonstrated that an intravenous injection of xylitol had a tumor-suppressing effect in mice, to which the xylitol-triggered ER stress also greatly contributed. We also observed that xylitol efficiently sensitized cancer cells to chemotherapeutic drugs. Based on our findings, a chemotherapeutic strategy combined with xylitol might improve the outcomes of patients facing cancer.

    DOI: 10.1016/j.cbi.2020.109085

    PubMed

    researchmap

  • 転移がんに有効なS100A8/A9阻害薬の開発(Novel therapeutic approach based on S100A8/A9-mediated organ tropic cancer metastasis)

    木下 理恵, 友信 奈保子, 山内 明, 枝園 和彦, 冨田 秀太, 村田 等, 二見 淳一郎, 近藤 英作, 豊岡 伸一, 阪口 政清

    日本癌学会総会記事   78回   P - 2252   2019年9月

     詳細を見る

    記述言語:英語   出版者・発行元:(一社)日本癌学会  

    researchmap

  • Upregulation of Mobility in Pancreatic Cancer Cells by Secreted S100A11 Through Activation of Surrounding Fibroblasts. 査読 国際誌

    Yosuke Mitsui, Nahoko Tomonobu, Masami Watanabe, Rie Kinoshita, I Wayan Sumardika, Chen Youyi, Hitoshi Murata, Ken-Ichi Yamamoto, Takuya Sadahira, Acosta Gonzalez Herik Rodrigo, Hitoshi Takamatsu, Kota Araki, Akira Yamauchi, Masahiro Yamamura, Hideyo Fujiwara, Yusuke Inoue, Junichiro Futami, Ken Saito, Hidekazu Iioka, Eisaku Kondo, Masahiro Nishibori, Shinichi Toyooka, Yasuhiko Yamamoto, Yasutomo Nasu, Masakiyo Sakaguchi

    Oncology research   27 ( 8 )   945 - 956   2019年8月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    S100A11, a member of the S100 family of proteins, is actively secreted from pancreatic ductal adenocarcinoma (PDAC) cells. However, the role of the extracellular S100A11 in PDAC progression remains unclear. In the present study, we investigated the extracellular role of S100A11 in crosstalking between PDAC cells and surrounding fibroblasts in PDAC progression. An abundant S100A11 secreted from pancreatic cancer cells stimulated neighboring fibroblasts through receptor for advanced glycation end products (RAGE) upon S100A11 binding and was followed by not only an enhanced cancer cell motility in vitro but also an increased number of the PDAC-derived circulating tumor cells (CTCs) in vivo. Mechanistic investigation of RAGE downstream in fibroblasts revealed a novel contribution of a mitogen-activated protein kinase kinase kinase (MAPKKK), tumor progression locus 2 (TPL2), which is required for positive regulation of PDAC cell motility through induction of cyclooxygenase 2 (COX2) and its catalyzed production of prostaglandin E2 (PGE2), a strong chemoattractive fatty acid. The extracellularly released PGE2 from fibroblasts was required for the rise in cellular migration as well as infiltration of their adjacent PDAC cells in a coculture setting. Taken together, our data reveal a novel role of the secretory S100A11 in PDAC disseminative progression through activation of surrounding fibroblasts triggered by the S100A11-RAGE-TPL2-COX2 pathway. The findings of this study will contribute to the establishment of a novel therapeutic antidote to PDACs that are difficult to treat by regulating cancer-associated fibroblasts (CAFs) through targeting the identified pathway.

    DOI: 10.3727/096504019X15555408784978

    PubMed

    researchmap

  • Critical role of the MCAM-ETV4 axis triggered by extracellular S100A8/A9 in breast cancer aggressiveness. 査読 国際誌

    Youyi Chen, I Wayan Sumardika, Nahoko Tomonobu, Rie Kinoshita, Yusuke Inoue, Hidekazu Iioka, Yosuke Mitsui, Ken Saito, I Made Winarsa Ruma, Hiroki Sato, Akira Yamauchi, Hitoshi Murata, Ken-Ichi Yamamoto, Shuta Tomida, Kazuhiko Shien, Hiromasa Yamamoto, Junichi Soh, Junichiro Futami, Miyoko Kubo, Endy Widya Putranto, Takashi Murakami, Ming Liu, Toshihiko Hibino, Masahiro Nishibori, Eisaku Kondo, Shinichi Toyooka, Masakiyo Sakaguchi

    Neoplasia (New York, N.Y.)   21 ( 7 )   627 - 640   2019年7月

     詳細を見る

    記述言語:英語  

    Metastatic breast cancer is the leading cause of cancer-associated death in women. The progression of this fatal disease is associated with inflammatory responses that promote cancer cell growth and dissemination, eventually leading to a reduction of overall survival. However, the mechanism(s) of the inflammation-boosted cancer progression remains unclear. In this study, we found for the first time that an extracellular cytokine, S100A8/A9, accelerates breast cancer growth and metastasis upon binding to a cell surface receptor, melanoma cell adhesion molecule (MCAM). Our molecular analyses revealed an important role of ETS translocation variant 4 (ETV4), which is significantly activated in the region downstream of MCAM upon S100A8/A9 stimulation, in breast cancer progression in vitro as well as in vivo. The MCAM-mediated activation of ETV4 induced a mobile phenotype called epithelial-mesenchymal transition (EMT) in cells, since we found that ETV4 transcriptionally upregulates ZEB1, a strong EMT inducer, at a very high level. In contrast, downregulation of either MCAM or ETV4 repressed EMT, resulting in greatly weakened tumor growth and lung metastasis. Overall, our results revealed that ETV4 is a novel transcription factor regulated by the S100A8/A9-MCAM axis, which leads to EMT through ZEB1 and thereby to metastasis in breast cancer cells. Thus, therapeutic strategies based on our findings might improve patient outcomes.

    DOI: 10.1016/j.neo.2019.04.006

    PubMed

    researchmap

  • Melanoma cell adhesion molecule is the driving force behind the dissemination of melanoma upon S100A8/A9 binding in the original skin lesion. 査読 国際誌

    Youyi Chen, I Wayan Sumardika, Nahoko Tomonobu, I Made Winarsa Ruma, Rie Kinoshita, Eisaku Kondo, Yusuke Inoue, Hiroki Sato, Akira Yamauchi, Hitoshi Murata, Ken-Ichi Yamamoto, Shuta Tomida, Kazuhiko Shien, Hiromasa Yamamoto, Junichi Soh, Ming Liu, Junichiro Futami, Kaori Sasai, Hiroshi Katayama, Miyoko Kubo, Endy Widya Putranto, Toshihiko Hibino, Bei Sun, Masahiro Nishibori, Shinichi Toyooka, Masakiyo Sakaguchi

    Cancer letters   452   178 - 190   2019年6月

     詳細を見る

    記述言語:英語  

    Since metastasis accounts for the majority of cancer-associated deaths, studies on the mechanisms of metastasis are needed to establish innovative strategies for cancer treatment. We previously reported that melanoma cell adhesion molecule (MCAM) functions as a critical receptor for S100A8/A9, and binding of S100A8/A9 to MCAM results in the migration of melanoma cells to lung tissue. However, the critical role of MCAM in the original melanoma skin lesion is still not clear. In this study, we aimed to determine the importance of the S100A8/A9-MCAM axis in melanoma dissemination in a skin lesion as a critical early step for metastasis. Mechanistic studies revealed the downstream signaling of MCAM that signaled the induction of metastasis. S100A8/A9-MCAM binding activates mitogen-activated protein kinase kinase kinase 8 (MAP3K8), also termed TPL2, leading to strong activation of the transcription factor ETV4 and subsequent induction of matrix metalloproteinase-25 (MMP25), and finally to induction of melanoma lung tropic metastasis. Collectively, our results demonstrate a crucial role of the S100A8/A9-MCAM signaling axis in metastatic onset of melanoma cells and indicate that strategies targeting the identified pathway may be useful for the establishment of innovative anti-cancer therapies.

    DOI: 10.1016/j.canlet.2019.03.023

    PubMed

    researchmap

  • Extracellular S100A11 Plays a Critical Role in Spread of the Fibroblast Population in Pancreatic Cancers. 査読 国際誌

    Hitoshi Takamatsu, Ken-Ichi Yamamoto, Nahoko Tomonobu, Hitoshi Murata, Yusuke Inoue, Akira Yamauchi, I Wayan Sumardika, Youyi Chen, Rie Kinoshita, Masahiro Yamamura, Hideyo Fujiwara, Yosuke Mitsui, Kota Araki, Junichiro Futami, Ken Saito, Hidekazu Iioka, I Made Winarsa Ruma, Endy Widya Putranto, Masahiro Nishibori, Eisaku Kondo, Yasuhiko Yamamoto, Shinichi Toyooka, Masakiyo Sakaguchi

    Oncology research   27 ( 6 )   713 - 727   2019年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    The fertile stroma in pancreatic ductal adenocarcinomas (PDACs) has been suspected to greatly contribute to PDAC progression. Since the main cell constituents of the stroma are fibroblasts, there is crosstalking(s) between PDAC cells and surrounding fibroblasts in the stroma, which induces a fibroblast proliferation burst. We have reported that several malignant cancer cells including PDAC cells secrete a pronounced level of S100A11, which in turn stimulates proliferation of cancer cells via the receptor for advanced glycation end products (RAGE) in an autocrine manner. Owing to the RAGE+ expression in fibroblasts, the extracellular abundant S100A11 will affect adjacent fibroblasts. In this study, we investigated the significance of the paracrine axis of S100A11-RAGE in fibroblasts for their proliferation activity. In in vitro settings, extracellular S100A11 induced upregulation of fibroblast proliferation. Our mechanistic studies revealed that the induction is through RAGE-MyD88-mTOR-p70 S6 kinase upon S100A11 stimulation. The paracrine effect on fibroblasts is linked mainly to triggering growth but not cellular motility. Thus, the identified pathway might become a potential therapeutic target to suppress PDAC progression through preventing PDAC-associated fibroblast proliferation.

    DOI: 10.3727/096504018X15433161908259

    PubMed

    researchmap

  • exSSSRs (extracellular S100 soil sensor receptors)-Fc fusion proteins work as prominent decoys to S100A8/A9-induced lung tropic cancer metastasis. 査読 国際誌

    Rie Kinoshita, Hiroki Sato, Akira Yamauchi, Yuta Takahashi, Yusuke Inoue, I Wayan Sumardika, Youyi Chen, Nahoko Tomonobu, Kota Araki, Kazuhiko Shien, Shuta Tomida, Hidejiro Torigoe, Kei Namba, Eisuke Kurihara, Yusuke Ogoshi, Hitoshi Murata, Ken-Ichi Yamamoto, Junichiro Futami, Endy Widya Putranto, I Made Winarsa Ruma, Hiromasa Yamamoto, Junichi Soh, Toshihiko Hibino, Masahiro Nishibori, Eisaku Kondo, Shinichi Toyooka, Masakiyo Sakaguchi

    International journal of cancer   144 ( 12 )   3138 - 3145   2019年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Within the "seed and soil" theory of organ tropic cancer metastasis is a growing compilation of evidence that S100A8/A9 functions as a soil signal that attracts cancer cells to certain organs, which prove beneficial to their growth. S100A8/A9-sensing receptors including Toll-like receptor 4 (TLR4), advanced glycation end products (RAGE), and also important receptors we recently succeeded in identifying (EMMPRIN, NPTNβ, MCAM, and ALCAM) have the potential to become promising therapeutic targets. In our study, we prepared extracellular regions of these novel molecules and fused them to human IgG2-Fc to extend half-life expectancy, and we evaluated the anti-metastatic effects of the purified decoy proteins on metastatic cancer cells. The purified proteins markedly suppressed S100A8/A9-mediated lung tropic cancer metastasis. We hence expect that our novel biologics may become a prominent medicine to prevent cancer metastasis in clinical settings through cutting the linkage between "seed and soil".

    DOI: 10.1002/ijc.31945

    PubMed

    researchmap

  • NEUROPLASTIN-β MEDIATES S100A8/A9-INDUCED LUNG CANCER DISSEMINATIVE PROGRESSION. 査読 国際誌

    Molecular Carcinogenesis   58 ( 6 )   980 - 995   2019年6月

     詳細を見る

    記述言語:英語  

    DOI: 10.1002/mc.22987

    PubMed

    researchmap

  • Convenient methodology for extraction and subsequent selective propagation of mouse melanocytes in culture from adult mouse skin tissue. 査読 国際誌

    Biochem Biophys Rep.   18   100619 - 100619   2019年

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    DOI: 10.1016/j.bbrep.2019.100619

    PubMed

    researchmap

  • Newly developed anti-S100A8/A9 monoclonal antibody efficiently prevents lung tropic cancer metastasis. 査読

    Intarnational Journal of Cancer   2018年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    researchmap

  • JNK-mediated phosphorylation of SARM1 regulates NAD+ cleavage activity to inhibit mitochondrial respiration. 査読

    Journal of Biological Chemistry   2018年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    researchmap

  • Embigin Promotes Prostate Cancer Progression by S100A4-Dependent and-Independent Mechanisms. 国際誌

    I Made Winarsa Ruma, Rie Kinoshita, Nahoko Tomonobu, Yusuke Inoue, Eisaku Kondo, Akira Yamauchi, Hiroki Sato, I Wayan Sumardika, Youyi Chen, Ken-Ichi Yamamoto, Hitoshi Murata, Shinichi Toyooka, Masahiro Nishibori, Masakiyo Sakaguchi

    Cancers   10 ( 7 )   2018年7月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Embigin, a transmembrane glycoprotein belonging to the immunoglobulin superfamily, is involved in prostate and mammary gland development. As embigin's roles in cancer remain elusive, we studied its biological functions and interaction with extracellular S100A4 in prostate cancer progression. We found by a pull-down assay that embigin is a novel receptor for S100A4, which is one of the vital cancer microenvironment milleu. Binding of extracellular S100A4 to embigin mediates prostate cancer progression by inhibition of AMPK activity, activation of NF-κB, MMP9 and mTORC1 signaling, and inhibition of autophagy, which increase prostate cancer cell motility. We also found that embigin promotes prostate cancer growth, spheroid- and colony-forming ability, and survival upon chemotherapy independently of S100A4. An in vivo growth mouse model confirmed the importance of embigin and its cytoplasmic tail in mediating prostate tumor growth. Moreover, embigin and p21WAF1 can be used to predict survival of prostate cancer patients. Our results demonstrated for the first time that the S100A4-embigin/AMPK/mTORC1/p21WAF1 and NF-κB/MMP9 axis is a vital oncogenic molecular cascade for prostate cancer progression. We proposed that embigin and p21WAF1 could be used as prognostic biomarkers and a strategy to inhibit S100A4-embigin binding could be a therapeutic approach for prostate cancer patients.

    DOI: 10.3390/cancers10070239

    PubMed

    researchmap

  • 膵がん進展に導く膵がん細胞 間質線維芽細胞クロストークを介在する分泌性S100A11 受容体RAGE連携の役割

    光井 洋介, 山本 健一, Sumardika I Wayan, 木下 理恵, 村田 等, 二見 淳一郎, 高松 仁, 山本 靖彦, 西堀 正洋, 豊岡 伸一, 渡部 昌実, 那須 保友, 阪口 政清

    日本がん免疫学会総会プログラム・抄録集   22回   156 - 156   2018年7月

     詳細を見る

    記述言語:日本語   出版者・発行元:日本がん免疫学会  

    researchmap

  • 分泌性S100A11-受容体RAGEシグナルに着眼した膵がん間質増大のメカニズムの解明

    山本 健一, 高松 仁, 光井 洋介, 木下 理恵, 村田 等, 二見 淳一郎, 山本 靖彦, 西堀 正洋, 豊岡 伸一, 阪口 政清

    日本がん免疫学会総会プログラム・抄録集   22回   117 - 117   2018年7月

     詳細を見る

    記述言語:日本語   出版者・発行元:日本がん免疫学会  

    researchmap

  • Tumor necrosis factor-α downregulates the REIC/Dkk-3 tumor suppressor gene in normal human skin keratinocytes. 国際誌

    Ken Kataoka, Natsumi Maehara, Yuki Ayabe, Hitoshi Murata, Nam-Ho Huh, Masakiyo Sakaguchi

    Molecular medicine reports   17 ( 5 )   6661 - 6666   2018年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Our previous studies revealed that REIC/Dkk-3 was expressed various tissues, including skin keratinocytes. The aim of the present study was to identify the factors that regulate the expression of the dickkopf Wnt signaling pathway inhibitor 3 (REIC/Dkk‑3) tumor suppressor gene in normal human skin keratinocytes (NHKs). Several growth factors and cytokines that have previously been reported to be involved in the growth and differentiation of keratinocytes were screened as potential regulators. Western blot analysis was performed using protein from NHKs cultured with/without various factors including the epidermal growth factor, tumor necrosis factor‑α, transforming growth factor‑β, interleukin (IL)‑1F9, IL‑6, IL‑8 and Ca2+. The results indicated that only TNF‑α downregulated REIC/Dkk‑3 expression in NHKs. Subsequently, TNF‑α was confirmed to reduce the expression levels of REIC/Dkk‑3 in mouse skin tissue and hair culture models. TNF‑α‑mediated downregulation of REIC/Dkk‑3 expression in NHKs was abrogated by the addition of a TNF‑α‑specific antibody. In conclusion, the results indicate that TNF‑α downregulates REIC/Dkk‑3 expression in normal skin keratinocytes.

    DOI: 10.3892/mmr.2018.8676

    PubMed

    researchmap

  • β-1,3-Galactosyl-O-Glycosyl-Glycoprotein β-1,6-N-Acetylglucosaminyltransferase 3 Increases MCAM Stability, Which Enhances S100A8/A9-Mediated Cancer Motility. 査読 国際誌

    Sumardika IW, Youyi C, Kondo E, Inoue Y, Ruma IMW, Murata H, Kinoshita R, Yamamoto KI, Tomida S, Shien K, Sato H, Yamauchi A, Futami J, Putranto EW, Hibino T, Toyooka S, Nishibori M, Sakaguchi M

    Oncology research   26 ( 3 )   431 - 444   2018年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    DOI: 10.3727/096504017X15031557924123

    PubMed

    researchmap

  • Tumor necrosis factor-α downregulates the REIC/Dkk-3 tumor suppressor gene in normal human skin keratinocytes.

    Molecular Medicine Reports   2018年

     詳細を見る

  • Signal Diversity of Receptor for Advanced Glycation End Products

    Masakiyo Sakaguchi, Rie Kinoshita, Endy Widya Putranto, I. Made Winarsa Ruma, I. Wayan Sumardika, Chen Youyi, Naoko Tomonobu, Ken-ichi Yamamoto, Hitoshi Murata

    ACTA MEDICA OKAYAMA   71 ( 6 )   459 - 465   2017年12月

     詳細を見る

    記述言語:英語   出版者・発行元:OKAYAMA UNIV MED SCHOOL  

    The receptor for advanced glycation end products (RAGE) is involved in inflammatory pathogenesis. It functions as a receptor to multiple ligands such as AGEs, HMGB1 and S100 proteins, activating multiple intracellular signaling pathways with each ligand binding. The molecular events by which ligand-activated RAGE controls diverse signaling are not well understood, but some progress was made recently. Accumulating evidence revealed that RAGE has multiple binding partners within the cytoplasm and on the plasma membrane. It was first pointed out in 2008 that RAGE's cytoplasmic tail is able to recruit Diaphanous-1 (Dia-1), resulting in the acquisition of increased cellular motility through Rac1/Cdc42 activation. We also observed that within the cytosol, RAGE's cytoplasmic tail behaves similarly to a Toll-like receptor (TLR4)-TIR domain, interacting with TIRAP and MyD88 adaptor molecules that in turn activate multiple downstream signals. Subsequent studies demonstrated the presence of an alternative adaptor molecule, DAP10, on the plasma membrane. The coupling of RAGE with DAP10 is critical for enhancing the RAGE-mediated survival signal. Interestingly, RAGE interaction on the membrane was not restricted to DAP10 alone. The chemotactic G-protein-coupled receptors (GPCRs) formyl peptide receptors1 and 2 (FPR1 and FPR2) also interacted with RAGE on the plasma membrane. Binding interaction between leukotriene B4 receptor 1 (BLT1) and RAGE was also demonstrated. All of the interactions affected the RAGE signal polarity. These findings indicate that functional interactions between RAGE and various molecules within the cytoplasmic area or on the membrane area coordinately regulate multiple ligand-mediated RAGE responses, leading to typical cellular phenotypes in several pathological settings. Here we review RAGE's signaling diversity, to contribute to the understanding of the elaborate functions of RAGE in physiological and pathological contexts.

    Web of Science

    researchmap

  • Robust cancer-specific gene expression by a novel cassette with hTERT and CMV promoter elements

    Masakiyo Sakaguchi, Takuya Sadahira, Hideo Ueki, Rie Kinoshita, Hitoshi Murata, Ken-Ichi Yamamoto, Junichiro Futami, Yasutomo Nasu, Kazuhiko Ochiai, Hiromi Kumon, Nam-Ho Huh, Masami Watanabe

    ONCOLOGY REPORTS   38 ( 2 )   1108 - 1114   2017年8月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SPANDIDOS PUBL LTD  

    We developed and validated a novel hTERT/CMV promoter element-driven gene expression cassette that can robustly enhance cancer-specific gene expression. The following gene expressional elements were located in tandem within the plasmid construct: [hTERT core promoter, cytomegalovirus (CMV) minimized promoter, RU5' sequence, an inserted gene, BGH polyA, hTERT enhancer]; this is hereafter referred to as the hT/Cm-R-hT construct. Using various human cancer cell lines and normal cells, the cancer-specific transcription of the green fluorescent protein (GFP) gene was examined by western blotting and fluorescence microscopy. Cancer-specific gene expression was robustly achieved in the hT/Cm-R-hT plasmid in comparison to the other control hT/Cm-driven construct. Notably, the expression level of GFP observed in the hT/Cm-RhT-driven construct was superior to that of the control plasmid with the conventional CMV promoter in HEK293 cells, which are known to possess higher hTERT activity than normal cells. We next examined the availability of hT/Cm-R-hT in detecting the target GFP expressing cancer cells from human peripheral blood mononuclear cells (PBMCs). The hT/Cm-R-hT plasmid successfully induced cancer-specific gene expression; the robust expression of GFP was observed in target HeLa cancer cells, whereas GFP was not visibly expressed in normal PBMCs. The plasmid allowed for the selective visualization of viable HeLa cancer cells in mixed cell cultures containing up to 10000-fold more PBMCs. These findings indicate that the hT/Cm-R-hT expressional system is a valuable tool for detecting viable cancer cells mixed with normal cells. The current system can therefore be applied to the in vitro detection of cancer cells that are disseminated in the blood and other types of body fluid in vivo. Since the current system can also be applied to other types of vectors, including virus vectors, this approach using the hTERT promoter-based construct is expected to become a valuable tool for enhancing cancer specific gene expression.

    DOI: 10.3892/or.2017.5710

    Web of Science

    researchmap

  • Expression of tumor suppressor REIC/Dkk-3 by a newly improved adenovirus vector with insertion of a hTERT promoter at the 3'-side of the transgene

    Endy Widya Putranto, Rie Kinoshita, Masami Watanabe, Takuya Sadahira, Hitoshi Murata, Ken-Ichi Yamamoto, Junichiro Futami, Ken Kataoka, Yusuke Inoue, I. Made Winarsa Ruma, I. Wayan Sumardika, Chen Youyi, Miyoko Kubo, Yoshihiko Sakaguchi, Kenji Saito, Yasutomo Nasu, Hiromi Kumon, Nam-Ho Huh, Masakiyo Sakaguchi

    ONCOLOGY LETTERS   14 ( 1 )   1041 - 1048   2017年7月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SPANDIDOS PUBL LTD  

    Reduced expression in immortalized cells (REIC)/Dickkopf-3 (Dkk-3) overexpression, induced using an adenovirus (Ad)-REIC, has been revealed to have a dramatic therapeutic effect on multiple types of cancer. To achieve an improved therapeutic effect from Ad-REIC on cancer, our group previously developed an enhanced gene expression system, the C-TSC cassette [cytomegalovirus (CMV)-RU5' located upstream (C); another promoter unit composed of triple tandem promoters, human telomerase reverse transcriptase (hTERT), simian virus 40 and CMV, located downstream of the cDNA (TSC); plus a polyadenylation (polyA) signal]. When applied to the conventional Ad-REIC, this novel system induced the development of an enhanced product, Ad-C-TSC-REIC, which exhibited a noticeable anticancer effect. However, there were difficulties in terms of Ad-C-TSC-REIC productivity in HEK293 cells, which are a widely used donor cell line for viral production. Productivity of Ad-C-TSC-REIC was significantly reduced compared with the conventional Ad-REIC, as the Ad-C-TSC-REIC had a significantly higher ability to induce apoptotic cell death of not only various types of cancer cell, but also HEK293 cells. The present study aimed to overcome this problem by modifying the C-TSC structure, resulting in an improved candidate: A C-T cassette (C: CMV-RU5' located upstream; T: another promoter unit composed of a single hTERT promoter, located downstream of the cDNA plus a polyA signal), which demonstrated gene expression comparable to that of the C-TSC system. The improved adenovirus REIC/Dkk-3 product with the C-T cassette, named Ad-C-T-REIC, exhibited a higher expression level of REIC/Dkk3, similar to that of Ad-C-TSC-REIC. Notably, the vector mitigated the cell death of donor HEK293 cells, resulting in a higher rate of production of its adenovirus. These results indicated that Ad-C-T-REIC has the potential to be a useful tool for application in cancer gene therapy.

    DOI: 10.3892/ol.2017.6201

    Web of Science

    researchmap

  • ß-1,3-galactosyl-O-glycosyl-glycoprotein ß-1,6-N-acetylglucosaminyltransferase 3 Increases MCAM Stability, Which Enhances S100A8/A9-Mediated Cancer Motility.

    Oncology research   2017年

     詳細を見る

  • Active Secretion of Dimerized S100A11 Induced by the Peroxisome in Mesothelioma Cells. 国際誌

    Satomi Saho, Hiroki Satoh, Eisaku Kondo, Yusuke Inoue, Akira Yamauchi, Hitoshi Murata, Rie Kinoshita, Ken-Ichi Yamamoto, Junichiro Futami, Endy Widya Putranto, I Made Winarsa Ruma, I Wayan Sumardika, Chen Youyi, Ken Suzawa, Hiromasa Yamamoto, Junichi Soh, Shuta Tomida, Yoshihiko Sakaguchi, Ken Saito, Hidekazu Iioka, Nam-Ho Huh, Shinichi Toyooka, Masakiyo Sakaguchi

    Cancer microenvironment : official journal of the International Cancer Microenvironment Society   9 ( 2-3 )   93 - 105   2016年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    S100A11, a small Ca2+ binding protein, acts extracellularly as a mediator of cancer progression. That raises the question of how a protein that lacks the classical secretory signal is able to be secreted outside cells without being damaged. Some insights into this question have been obtained, and there has been accumulating evidence indicating a pivotal role of a non-classical vesicle-mediated pathway using lysosomes or peroxisomes for the protein secretion. To obtain a more precise insight into the secretory mechanism of S100A11, we first screened representative cancer cells exhibiting significantly active secretion of S100A11. From the results of profiling, we turned our attention to aggressive cancer mesothelioma cells. In mesothelioma cells, we found that abundant dimeric S100A11 was produced selectively in the peroxisome after transportation of monomeric S100A11 through an interaction with PEX14, a peroxisome membrane protein, resulting in peroxisomal secretion of dimerized S100A11. In an extracellular environment in vitro, dimerized S100A11 promoted mesothelial cell invasion indirectly with the help of fibroblast cells. Overall, the results indicate that the peroxisome functions as an essential vesicle for the production of dimerized S100A11 and the subsequent secretion of the protein from mesothelioma cells and that peroxisome-mediated secretion of dimerized S100A11 might play a critical role in mesothelioma progression in a tumor microenvironment.

    DOI: 10.1007/s12307-016-0185-2

    PubMed

    researchmap

  • Identification of an S100A8 Receptor Neuroplastin-β and its Heterodimer Formation with EMMPRIN

    Masakiyo Sakaguchi, Mami Yamamoto, Masashi Miyai, Tatsuo Maeda, Junichiro Hiruma, Hitoshi Murata, Rie Kinoshita, I. Made, Winarsa Ruma, ndy Widya Putranto, Yusuke Inoue, Shin Morizane, Nam Ho Huh, Ryoji Tsuboi, Toshihiko Hibino

    Journal of Investigative Dermatology, Advances in biology of skin   136 ( 11 )   2240 - 2250   2016年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    DOI: 10.1016/j.jid.2016.06.617

    Web of Science

    researchmap

  • MCAM, as a novel receptor for S100A8/A9, mediates progression of malignant melanoma through prominent activation of NF-κB and ROS formation upon ligand binding

    I. Made, Winarsa Ruma, ndy Widya Putranto, Eisaku Kondo, Hitoshi Murata, Masami Watanabe, Peng Huang, Rie Kinoshita, Junichiro Futami, Yusuke Inoue, Akira Yamauchi, I. Wayan, Sumardika,Chen Youyi, Ken Ichi Yamamoto, Yasutomo Nasu, Masahiro Nishibori, Toshihiko H

    Clinical and Experimental Metastasis   33 ( 6 )   609 - 627   2016年8月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    DOI: 10.1007/s10585-016-9801-2

    Web of Science

    researchmap

  • An efficient method for the preparation of preferentially heterodimerized recombinant S100A8/A9 coexpressed in Escherichia coli

    Junichiro Futami, Yuki Atago, Akari Azuma, Endy Widya Putranto, Rie Kinoshita, Hitoshi Murata, Masakiyo Sakaguchi

    Biochemistry and Biophysics Reports   6   94 - 100   2016年7月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Elsevier B.V.  

    It is now known that multicomponent protein assemblies strictly regulate many protein functions. The S100 protein family is known to play various physiological roles, which are associated with alternative complex formations. To prepare sufficient amounts of heterodimeric S100A8 and S100A9 proteins, we developed a method for bicistronic coexpression from a single-vector system using Escherichia coli cells as a host. The complex formation between S100A8 and S100A9 appears to be dependent on the thermodynamic stability of the protein during expression. The stable S100A8/A9 heterodimer complex spontaneously formed during coexpression, and biologically active samples were purified by cation-exchange chromatography. Semi-stable homodimers of S100A8 and S100A9 were also formed when expressed individually. These results suggest that the assembly of S100 protein complexes might be regulated by expression levels of partner proteins in vivo. Because protein assembly occurs rapidly after protein synthesis, coexpression of relevant proteins is crucial for the design of multicomponent recombinant protein expression systems.

    DOI: 10.1016/j.bbrep.2016.03.009

    Scopus

    researchmap

  • PINK1 Regulation of Mitochondrial Homeostasis and Cell Survival. 査読

    Murata Hitoshi, Yamamoto Ken-ichi, Kinoshita Rie, Kataoka Ken, Huh Nam-ho, Sakaguchi Masakiyo

    IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY-ANIMAL   52   S8   2016年6月

  • NRF2 Regulates PINK1 Expression under Oxidative Stress Conditions

    Hitoshi Murata, Hitoshi Takamatsu, Sulai Lie, Ken Kataoka, Nam-ho Huh, Masakiyo Sakaguchi

    PLoS One   10 ( 11 )   2015年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:PUBLIC LIBRARY SCIENCE  

    Mutations of the PTEN-induced putative kinase 1 (PINK1) gene are a cause of autosomal recessive forms of Parkinson's disease. Recent studies have revealed that PINK1 is an essential factor for controlling mitochondrial quality, and that it protects cells from oxidative stresses. Although there has been considerable progress in the elucidation of various aspects of PINK1 protein regulation such as activation, stability and degradation, the transcriptional regulation of PINK1 mRNA under stress conditions remains unclear. In this study, we found that nuclear factor (erythroid-derived 2)-like 2 (NRF2), an antioxidant transcription factor, regulates PINK1 expression under oxidative stress conditions. Damaged mitochondria arising from stress conditions induced NRF2-dependent transcription of the PINK1 gene through production of reactive oxygen species (ROS). Either an ROS scavenger or forced expression of KEAP1, a potent inhibitory partner to NRF2, restricted PINK1 expression induced by activated NRF2. Transcriptionally up-regulated PINK1 diminished oxidative stress-associated cell death. The results indicate that PINK1 expression is positively regulated by NRF2 and that the NRF2-PINK1 signaling axis is deeply involved in cell survival.

    DOI: 10.1371/journal.pone.0142438

    Web of Science

    researchmap

  • DNAX-activating Protein 10 (DAP10) Membrane Adaptor Associates with Receptor for Advanced Glycation End Products (RAGE) and Modulates the RAGE-triggered Signaling Pathway in Human Keratinocytes

    Masakiyo Sakaguchi, Hitoshi Murata, Yumi Aoyama, Toshihiko Hibino, Endy Widya Putranto, I. Made Winarsa Ruma, Yusuke Inoue, Yoshihiko Sakaguchi, Ken-ichi Yamamoto, Rie Kinoshita, Junichiro Futami, Ken Kataoka, Keiji Iwatsuki, Nam-ho Huh

    JOURNAL OF BIOLOGICAL CHEMISTRY   289 ( 34 )   23389 - 23402   2014年8月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Background: RAGE receptor plays a critical role in many inflammatory disorders. Results: Functional interaction between RAGE and DAP10 coordinately regulates S100A8/A9-mediated cell survival. Conclusion: DAP10 membrane adaptor is critically involved in RAGE-mediated survival signaling upon S100A8/A9 binding. Significance: This is the first report demonstrating that RAGE-mediated survival signaling is critically regulated by DAP10 interaction.
    The receptor for advanced glycation end products (RAGE) is involved in the pathogenesis of many inflammatory, degenerative, and hyperproliferative diseases, including cancer. Previously, we revealed mechanisms of downstream signaling from ligand-activated RAGE, which recruits TIRAP/MyD88. Here, we showed that DNAX-activating protein 10 (DAP10), a transmembrane adaptor protein, also binds to RAGE. By artificial oligomerization of RAGE alone or RAGE-DAP10, we found that RAGE-DAP10 heterodimer formation resulted in a marked enhancement of Akt activation, whereas homomultimeric interaction of RAGE led to activation of caspase 8. Normal human epidermal keratinocytes exposed to S100A8/A9, a ligand for RAGE, at a nanomolar concentration mimicked the pro-survival response of RAGE-DAP10 interaction, although at a micromolar concentration, the cells mimicked the pro-apoptotic response of RAGE-RAGE. In transformed epithelial cell lines, A431 and HaCaT, in which endogenous DAP10 was overexpressed, and S100A8/A9, even at a micromolar concentration, led to cell growth and survival due to RAGE-DAP10 interaction. Functional blocking of DAP10 in the cell lines abrogated the Akt phosphorylation from S100A8/A9-activated RAGE, eventually leading to an increase in apoptosis. Finally, S100A8/A9, RAGE, and DAP10 were overexpressed in the psoriatic epidermis. Our findings indicate that the functional interaction between RAGE and DAP10 coordinately regulates S100A8/A9-mediated survival and/or apoptotic response of keratinocytes.

    DOI: 10.1074/jbc.M114.573071

    Web of Science

    researchmap

  • Extract of Cordyceps militaris inhibits angiogenesis and suppresses tumor growth of human malignant melanoma cells

    I. Made Winarsa Ruma, Endy Widya Putranto, Eisaku Kondo, Risayo Watanabe, Ken Saito, Yusuke Inoue, Ken-Ichi Yamamoto, Susumu Nakata, Masaji Kaihata, Hitoshi Murata, Masakiyo Sakaguchi

    INTERNATIONAL JOURNAL OF ONCOLOGY   45 ( 1 )   209 - 218   2014年7月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SPANDIDOS PUBL LTD  

    Angiogenesis is essential for tumor development and metastasis. Among several angiogenic factors, vascular endothelial growth factor receptor (VEGF) is important for tumor-derived angiogenesis and commonly overexpressed in solid tumors. Thus, many antitumor strategies targeting VEGF have been developed to inhibit cancer angiogenesis, offering insights into the successful treatment of solid cancers. However, there are a number of issues such as harmful effects on normal vascularity in clinical trials. Taking this into consideration, we employed Cordyceps militaris as an antitumor approach due to its biological safety in vivo. The herbal medicinal mushroom Cordyceps militaris has been reported to show potential anticancer properties including anti-angiogenic capacity; however, its concrete properties have yet to be fully demonstrated. In this study, we aimed to elucidate the biological role of Cordyceps militaris extract in tumor cells, especially in regulating angiogenesis and tumor growth of a human malignant melanoma cell line. We demonstrated that Cordyceps militaris extract remarkably suppressed tumor growth via induction of apoptotic cell death in culture that links to the abrogation of VEGF production in melanoma cells. This was followed by mitigation of Akt1 and GSK-3 beta activation, while p38 alpha phosphorylation levels were increased. Extract treatment in mouse model xenografted with human melanoma cells resulted in a dramatic antitumor effect with downregulation of VEGF expression. The results suggest that suppression of tumor growth by Cordyceps militaris extract is, at least, mediated by its anti-angiogenicity and apoptosis induction capacities. Cordyceps militaris extract may be a potent antitumor herbal drug for solid tumors.

    DOI: 10.3892/ijo.2014.2397

    Web of Science

    researchmap

  • Dramatic Increase in Expression of a Transgene by Insertion of Promoters Downstream of the Cargo Gene

    Masakiyo Sakaguchi, Masami Watanabe, Rie Kinoshita, Haruki Kaku, Hideo Ueki, Junichiro Futami, Hitoshi Murata, Yusuke Inoue, Shun-Ai Li, Peng Huang, Endy Widya Putranto, I. Made Winarsa Ruma, Yasutomo Nasu, Hiromi Kumon, Nam-ho Huh

    MOLECULAR BIOTECHNOLOGY   56 ( 7 )   621 - 630   2014年7月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:HUMANA PRESS INC  

    For expression of genes in mammalian cells, various vectors have been developed using promoters including CMV, EF-1 alpha, and CAG promoters and have been widely used. However, such expression vectors sometimes fail to attain sufficient expression levels depending on the nature of cargo genes and/or on host cell types. In the present study, we aimed to develop a potent promoter system that enables high expression levels of cargo genes ubiquitously in many different cell types. We found that insertion of an additional promoter downstream of a cargo gene greatly enhanced the expression levels. Among the constructs we tested, C-TSC cassette (C: CMV-RU5' located upstream; TSC: another promoter unit composed of triple tandem promoters, hTERT, SV40, and CMV, located downstream of the cDNA plus a polyadenylation signal) had the most potent capability, showing far higher efficiency than that of potent conventional vector systems. The results indicate that the new expression system is useful for production of recombinant proteins in mammalian cells and for application as a gene therapeutic measure.

    DOI: 10.1007/s12033-014-9738-0

    Web of Science

    researchmap

  • DOCK7 is a critical regulator of the RAGE-Cdc42 signaling axis that induces formation of dendritic pseudopodia in human cancer cells

    Ken-Ichi Yamamoto, Hitoshi Murata, Endy Widya Putranto, Ken Kataoka, Akira Motoyama, Toshihiko Hibino, Yusuke Inoue, Masakiyo Sakaguchi, Nam-Ho Huh

    Oncology Reports   29 ( 3 )   1073 - 1079   2013年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Cellular migration is a fundamental process linked to cancer metastasis. Growing evidence indicates that the receptor for advanced glycation end products (RAGE) plays a pivotal role in this process. With regard to downstream signal transducers of RAGE, diaphanous-1 and activated small guanine nucleotide triphosphatases, Rac1 and Cdc42, have been identified. To obtain precise insight into the direct downstream signaling mechanism of RAGE, we screened for proteins interacting with the cytoplasmic domain of RAGE employing an immunoprecipitation-liquid chromatography coupled with an electrospray tandem mass spectrometry system. In the present study, we found that the cytoplasmic domain of RAGE interacted with an atypical DOCK180-related guanine nucleotide exchange factor, dedicator of cytokinesis protein 7 (DOCK7). DOCK7 bound to the RAGE cytoplasmic domain and transduced a signal to Cdc42, resulting in the formation of abundant highly branched filopodia-like protrusions, dendritic pseudopodia. Blocking of the function of DOCK7 greatly abrogated the formation of dendritic pseudopodia and suppressed cellular migration. These results indicate that DOCK7 functions as an essential and downstream regulator of RAGE-mediated cellular migration through the formation of dendritic pseudopodia.

    DOI: 10.3892/or.2012.2191

    Scopus

    PubMed

    researchmap

  • SARM1 and TRAF6 bind to and stabilize PINK1 on depolarized mitochondria

    Murata H, Sakaguchi M, Kataoka K, Huh NH

    Molecular Biology of the Cell   2013年

  • Inhibition of RAGE signaling by intracellularly delivered inhibitor peptides using PEI-cationization

    Putranto EW, Murata H, Yamamoto KI, Kataoka K, Yamada H, Futami J, Sakaguchi M, Huh NH

    International Journal of Molecular Medicine   32 ( 4 )   938 - 944   2013年

     詳細を見る

    記述言語:英語   出版者・発行元:Spandidos Publications Ltd.  

    The receptor for advanced glycation end products (RAGE) is a multi-ligand cell surface receptor and a member of the immunoglobulin superfamily. RAGE is involved in a wide range of inflammatory, degenerative and hyper-proliferative disorders which span over different organs by engaging diverse ligands, including advanced glycation end products, S100 family proteins, high-mobility group protein B1 (HMGB1) and amyloid beta. We previously demonstrated that the cytoplasmic domain of RAGE is phosphorylated upon the binding of ligands, enabling the recruitment of two distinct pairs of adaptor proteins, Toll-interleukin 1 receptor domain-containing adaptor protein (TIRAP) and myeloid differentiation protein 88 (MyD88). This engagement allows the activation of downstream effector molecules, and thereby mediates a wide variety of cellular processes, such as inflammatory responses, apoptotic cell death, migration and cell growth. Therefore, inhibition of the binding of TIRAP to RAGE may abrogate intracellular signaling from ligand-activated RAGE. In the present study, we developed inhibitor peptides for RAGE signaling (RAGE-I) by mimicking the phosphorylatable cytosolic domain of RAGE. RAGE-I was efficiently delivered into the cells by polyethylenimine (PEI) cationization. We demonstrated that RAGE-I specifically bound to TIRAP and abrogated the activation of Cdc42 induced by ligand-activated RAGE. Furthermore, we were able to reduce neuronal cell death induced by an excess amount of S100B and to inhibit the migration and invasion of glioma cells in vitro. Our results indicate that RAGE-I provides a powerful tool for therapeutics to block RAGE-mediated multiple signaling.

    DOI: 10.3892/ijmm.2013.1467

    CiNii Article

    CiNii Books

    researchmap

  • Uniformly Cationized Protein Efficiently Reaches the Cytosol of Mammalian Cells

    Midori Futami, Yasuyoshi Watanabe, Takashi Asama, Hitoshi Murata, Hiroko Tada, Megumi Kosaka, Hidenori Yamada, Junichiro Futami

    BIOCONJUGATE CHEMISTRY   23 ( 10 )   2025 - 2031   2012年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER CHEMICAL SOC  

    Protein cationization techniques are powerful protein transduction methods for mammalian cells. As we demonstrated previously, cationized proteins with limited conjugation to polyethylenimine have excellent ability to enter into cells by adsorption-mediated endocytosis [Futami, J., et al. (2005) J. Biosci. Bioeng. 99, 95-103]. In this study, we show that proteins with extensive and uniform cationization covering the protein surface reach the cytoplasm and nucleus more effectively than proteins with limited cationic polymers or proteins that are fused to cationic peptides. Although extensive modification of carboxylates results in loss of protein function, chicken avidin retains biotin-binding ability even after extensive amidation of carboxylates. Using this cationized avidin carrier system, the protein transduction ability of variously cationized avidins was investigated using biotinylated protein as a probe. The results revealed that cationized avidins bind rapidly to the cell surface followed by endocytotic uptake. Small amounts of uniformly cationized avidin showed direct penetration into the cytoplasm within a 15 min incubation. This penetration route seemed to be energy dependent and functioned under cellular physiological conditions. A biotinylated exogenous transcription factor protein that penetrated cells was demonstrated to induce target gene expression in living cells.

    DOI: 10.1021/bc300030d

    Web of Science

    researchmap

  • Expression pattern of REIC/Dkk-3 in mouse squamous epithelia

    K. Kataoka, G. Du, N. Maehara, H. Murata, M. Sakaguchi, N. Huh

    CLINICAL AND EXPERIMENTAL DERMATOLOGY   37 ( 4 )   428 - 431   2012年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:WILEY-BLACKWELL  

    The REIC/Dkk (reduced expression in immortalized cells/Dickkopf-3) gene was originally identified as a tumour-suppressor gene with reduced expression in immortalized cells, cancer-cell lines and tumour tissues. Of the four members of the Dkk family, the REIC/Dkk-3 protein is unique in terms of DNA sequence, expression profiles and biological functions. In this study, we investigated and compared the expression patterns of the REIC/Dkk-3 protein in mouse squamous epithelia. Expression of REIC/Dkk-3 in the back skin was localized to the upper layer of the interfollicular epidermis, and the inner root sheath of hair follicles. Expression of REIC/Dkk-3 was detected in the ear skin, oral mucosa, tongue, oesophagus, uterine cervix, footpad and tail skin, but not in the cornea. Interestingly, expression was localized to the upper layers of these epithelial tissues. The physiological function of REIC/Dkk-3 is still unclear, but our detailed observation highlight its unique expression pattern in squamous epithelia.

    DOI: 10.1111/j.1365-2230.2011.04301.x

    Web of Science

    researchmap

  • S100A7 promotes the migration and invasion of osteosarcoma cells via the receptor for advanced glycation end products

    Ken Kataoka, Tomoyuki Ono, Hitoshi Murata, Mika Morishita, Ken-Ichi Yamamoto, Masakiyo Sakaguchi, Nam-Ho Huh

    ONCOLOGY LETTERS   3 ( 5 )   1149 - 1153   2012年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SPANDIDOS PUBL LTD  

    Osteosarcoma is the most common malignant tumor of bone in childhood and adolescence. Despite intensive research for new therapies, the outcome in patients with metastasis remains extremely poor. S100 proteins are involved in the proliferation, cell cycle progression and metastasis of numerous malignant tumors, including osteosarcoma. In the present study, we identified S100A7 as a candidate to promote the migration of osteosarcoma cells. S100A7 promoted the migration and invasion of osteosarcoma cells as assayed in vitro. An in vitro pull-down assay revealed the binding of the recombinant S100A7 protein with its putative receptor, the receptor for advanced glycation end products (RAGE). The downregulation of RAGE by a specific siRNA markedly suppressed the migration and invasion of osteosarcoma cells. Furthermore, the matrix metalloproteinase activity of osteosarcoma cells was enhanced by S100A7 and suppressed by the downregulation of RAGE. These results indicate that S100A7 promotes the migration and invasion of osteosarcoma cells through RAGE. The S100A7-RAGE axis may thus be a new target for preventing the invasion and/or metastasis of osteosarcoma.

    DOI: 10.3892/ol.2012.612

    Web of Science

    researchmap

  • Partial sensitization of human bladder cancer cells to a gene-therapeutic adenovirus carrying REIC/Dkk-3 by downregulation of BRPK/PINK1

    Yu Jin, Hitoshi Murata, Masakiyo Sakaguchi, Ken Kataoka, Masami Watanabe, Yasutomo Nasu, Hiromi Kumon, Nam-Ho Huh

    ONCOLOGY REPORTS   27 ( 3 )   695 - 699   2012年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SPANDIDOS PUBL LTD  

    REIC/Dkk-3 is a tumor suppressor gene that was first identified as a gene downregulated in association with immortalization of normal human fibroblasts. We have demonstrated that an adenovirus carrying REIC/Dkk-3 (Ad-REIC) showed a tumor-specific killing effect on a wide range of cancers. However, some human cancers, bladder cancers in particular, are resistant to Ad-REIC. In this study, we investigated the combination effect of downregulation of BRPK/PINK1 (PINK1) and Ad-REIC on bladder cancer cells. Five bladder cancer cell lines among six cell lines examined were resistant to Ad-REIC. Among the cell lines, the resistance of two cell lines was probably due to low infection efficiency of the adenovirus. PINK1-specific siRNA remarkably downregulated Bcl-x(L) and TRAP1 proteins and upregulated BAX protein expression. Finally, downregulation of PINK1 partially sensitized the other three cell lines that were resistant to Ad-REIC. This sensitization was associated with increasing production of reactive oxygen species (ROS). These results indicate that PINK1 is one of the key molecules for the mitochondrial protection system and that PINK1 can be a new target molecule to sensitize bladder cancer cells that are resistant to Ad-REIC.

    DOI: 10.3892/or.2011.1543

    Web of Science

    researchmap

  • TIRAP, an Adaptor Protein for TLR2/4, Transduces a Signal from RAGE Phosphorylated upon Ligand Binding

    Masakiyo Sakaguchi, Hitoshi Murata, Ken-ichi Yamamoto, Tomoyuki Ono, Yoshihiko Sakaguchi, Akira Motoyama, Toshihiko Hibino, Ken Kataoka, Nam-ho Huh

    PLOS ONE   6 ( 8 )   2011年8月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:PUBLIC LIBRARY SCIENCE  

    The receptor for advanced glycation end products (RAGE) is thought to be involved in the pathogenesis of a broad range of inflammatory, degenerative and hyperproliferative diseases. It binds to diverse ligands and activates multiple intracellular signaling pathways. Despite these pivotal functions, molecular events just downstream of ligand-activated RAGE have been surprisingly unknown. Here we show that the cytoplasmic domain of RAGE is phosphorylated at Ser391 by PKCf upon binding of ligands. TIRAP and MyD88, which are known to be adaptor proteins for Toll-like receptor-2 and -4 (TLR2/4), bound to the phosphorylated RAGE and transduced a signal to downstream molecules. Blocking of the function of TIRAP and MyD88 largely abrogated intracellular signaling from ligand-activated RAGE. Our findings indicate that functional interaction between RAGE and TLRs coordinately regulates inflammation, immune response and other cellular functions.

    DOI: 10.1371/journal.pone.0023132

    Web of Science

    researchmap

  • Multiple functions of PINK1 at different intracellular locations Beyond neurodegenerative diseases

    Hitoshi Murata, Masakiyo Sakaguchi, Ken Kataoka, Nam-ho Huh

    CELL CYCLE   10 ( 10 )   1518 - 1519   2011年5月

     詳細を見る

    記述言語:英語   出版者・発行元:LANDES BIOSCIENCE  

    DOI: 10.4161/cc.10.10.15445

    Web of Science

    researchmap

  • Intraperitoneal administration of an adenovirus vector carrying REIC/Dkk-3 suppresses peritoneal dissemination of scirrhous gastric carcinoma

    Swe Swe Than, Ken Kataoka, Masakiyo Sakaguchi, Hitoshi Murata, Fernando Abarzua, Chika Taketa, Gang Du, Masakazu Yashiro, Kazuyoshi Yanagihara, Yasutomo Nasu, Hiromi Kumon, Nam-Ho Huh

    ONCOLOGY REPORTS   25 ( 4 )   989 - 995   2011年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SPANDIDOS PUBL LTD  

    Expression levels of the novel tumor suppressor gene REIC/Dkk-3 are reduced in many human cancers. We have previously showed that an adenovirus vector carrying REIC/Dkk-3 (Ad-REIC) induced apoptosis of cancer cells selectively and exerted bystander antitumor effects via ER stress. We examined possible effects of Ad-REIC in a peritoneal dissemination model of scirrhous gastric carcinoma (SGC). Among various types of gastric cancer, SGC continues to be associated with the worst prognosis due to a high incidence of metastases in the peritoneal cavity. We found that a single intraperitoneal injection of Ad-REIC suppressed tumor dissemination and disease progression. Immunomodulation by Ad-REIC led to recruitment of natural killer cells inside tumor nodules. We conclude that Ad-REIC gene therapy may be a potential tool in combinatorial approaches to achieve curative effects in SGC.

    DOI: 10.3892/or.2011.1149

    Web of Science

    researchmap

  • A new cytosolic pathway from a Parkinson’s disease-associated kinase, BRPK/PINK1: activation of AKT via MTORC2.

    Murata H, Sakaguchi M, Jin Y, Sakaguchi Y, Futami JI, Kataoka K, Huh NH

    Journal of biological chemistry   286 ( 9 )   7182 - 7189   2011年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    DOI: 10.1074/jbc.M110.179390

    Web of Science

    researchmap

  • Internalization of REIC/Dkk-3 protein by induced pluripotent stem cell-derived embryoid bodies and extra-embryonic tissues

    Ken Kataoka, Masakiyo Sakaguchi, Kun Peng Li, Chika Taketa, Ken-Ichi Yamamoto, Gang Du, Hiroaki Funahashi, Hitoshi Murata, Nam-Ho Huh

    INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE   26 ( 6 )   853 - 859   2010年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SPANDIDOS PUBL LTD  

    REIC/Dkk-3 was first identified as a down-regulated gene in a number of human immortalized cells and human tumor-derived cell lines. Overexpression of the REIC/Dkk-3 gene using an adenovirus vector (Ad-REIC) has showed a potent selective therapeutic effect on various human cancers through induction of ER stress. Furthermore, we recently showed that Ad-REIC has an indirect host-mediated anti-tumor activity by induction of IL-7. However, the physiological function of REIC/Dkk-3 is still unclear. As a first step to study the possible receptor(s) for secreted REIC/Dkk-3, we analyzed the internalization of Cy3-labeled recombinant REIC/Dkk-3 protein. Among the cell lines screened, mouse induced pluripotent stem (iPS) cells showed a unique pattern of internalization. The internalization was observed in peripheral cells of spherical colonies formed spontaneously, but not in undifferentiated iPS cells. When we analyzed embryoid bodies (EBs) derived from iPS cells, REIC/Dkk-3 protein was internalized specifically by differentiated cells located at the periphery of EBs. Interestingly, Dkk-1 was internalized by undifferentiated cells at the center of the EBs. When developmental tissue was analyzed, internalization of REIC/Dkk-3 protein was strictly limited to extra-embryonic tissue, such as the trophectoderm layer of 4.5 days post-coitus (dpc) blastocysts and the chorionic membrane at 16.5 dpc. The mechanism of the internalization was confirmed to be endocytosis. These findings will contribute to knowledge on the interaction of REIC/Dkk-3 with a possible receptor(s).

    DOI: 10.3892/ijmm_00000534

    Web of Science

    researchmap

  • Down-regulation of BiP/GRP78 sensitizes resistant prostate cancer cells to gene-therapeutic overexpression of REIC/Dkk-3

    Ryuta Tanimoto, Masakiyo Sakaguchi, Fernando Abarzua, Ken Kataoka, Kaoru Kurose, Hitoshi Murata, Yasutomo Nasu, Hiromi Kumon, Nam-Ho Huh

    INTERNATIONAL JOURNAL OF CANCER   126 ( 7 )   1562 - 1569   2010年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:JOHN WILEY & SONS INC  

    We have recently shown that an adenovirus carrying REIC/Dkk-3 (Ad-REIC) exhibits a potent tumor-specific cell-killing function for various human cancers. It has also become evident that some human cancers are resistant to Ad-REIC-induced apoptosis. The aim of the present study was to determine the molecular mechanisms of resistance to Ad-REIC. First, we isolated resistant clones from a human prostate cancer cell line, PC3, after repeated exposure to Ad-REIC. Infection efficiency of the adenovirus vector and expression level of REIC/Dkk-3 in the resistant clones were similar to those in the parental PC3 cells. By screening for alteration in levels and functional status of proteins involved in Ad-REIC-induced apoptosis, we found that BiP/GRP78, an ER-residing chaperone protein, was expressed at higher levels consistently among resistant cells. Expression levels of BiP and rates of apoptosis induced by Ad-REIC were inversely correlated. Down-regulation of BiP with siRNA sensitized the resistant cells to Ad-REIC in vivo as well as in culture. These results indicate that BiP is a major determinant of resistance to Ad-REIC-induced apoptosis. Thus BiP is useful for diagnosis of inherent and acquired resistance of cancers and also as a target molecule to overcome resistance to the gene therapeutic Ad-REIC.

    DOI: 10.1002/ijc.24764

    Web of Science

    researchmap

  • Overexpression of REIC/Dkk-3 in Normal Fibroblasts Suppresses Tumor Growth via Induction of Interleukin-7

    Masakiyo Sakaguchi, Ken Kataoka, Fernando Abarzua, Ryuta Tanimoto, Masami Watanabe, Hitoshi Murata, Swe Swe Than, Kaoru Kurose, Yuji Kashiwakura, Kazuhiko Ochiai, Yasutomo Nasu, Hiromi Kumon, Nam-ho Huh

    JOURNAL OF BIOLOGICAL CHEMISTRY   284 ( 21 )   14236 - 14244   2009年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    We previously showed that the tumor suppressor gene REIC/Dkk-3, when overexpressed by an adenovirus (Ad-REIC), exhibited a dramatic therapeutic effect on human cancers through a mechanism triggered by endoplasmic reticulum stress. Adenovirus vectors show no target cell specificity and thus may elicit unfavorable side effects through infection of normal cells even upon intra-tumoral injection. In this study, we examined possible effects of Ad-REIC on normal cells. We found that infection of normal human fibroblasts (NHF) did not cause apoptosis but induced production of interleukin (IL)-7. The induction was triggered by endoplasmic reticulum stress and mediated through IRE1 alpha, ASK1, p38, and IRF-1. When Ad-REIC-infected NHF were transplanted in a mixture with untreated human prostate cancer cells, the growth of the cancer cells was significantly suppressed. Injection of an IL-7 antibody partially abrogated the suppressive effect of Ad-REIC-infected NHF. These results indicate that Ad-REIC has another arm against human cancer, an indirect host-mediated effect because of overproduction of IL-7 by mis-targeted NHF, in addition to its direct effect on cancer cells.

    DOI: 10.1074/jbc.M808002200

    Web of Science

    researchmap

  • Intracellular delivery of glutathione S-transferase-fused proteins into mammalian cells by polyethylenimine-glutathione conjugates

    Hitoshi Murata, Junichiro Futami, Midori Kitazoe, Takayuki Yonehara, Hidetaka Nakanishi, Megumi Kosaka, Hiroko Tada, Masakiyo Sakaguchi, Yasuyuki Yagi, Masaharu Seno, Nam-ho Huh, Hidenori Yamada

    JOURNAL OF BIOCHEMISTRY   144 ( 4 )   447 - 455   2008年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:OXFORD UNIV PRESS  

    The glutathione S-transferase (GST)-fused protein expression system has been extensively used to generate a large quantity of proteins and has served for functional analysis in vitro. In this study, we developed a novel approach for the efficient intracellular delivery of GST-fused proteins into living cells to expand their usefulness up to in vivo use. Since protein cationization techniques are powerful strategies for efficient intracellular uptake by adsorptive-mediated endocytosis, GST-fused proteins were cationized by forming a complex with a polycationic polyethylenimine (PEI)-glutathione conjugate. On screening of protein transduction, optimized PEI-glutathione conjugate for protein transduction was characterized by a partly oligomerized mixture of PEI with average molecular masses of 600 (PEI600) modified with multiple glutathiones, which could have sufficient avidity for GST. Furthermore, enhanced endosomal escape of transduced GST-fused proteins was observed when they were delivered with a glutathione-conjugated PEI600 derivative possessing a hydroxybutenyl moiety. These results were confirmed by both intracellular confocal imaging of GST-fused green fluorescent protein and activation of an endogenous growth signal transduction pathway by a GST-fused constitutively active mutant of a kinase protein. These PEI-glutathione conjugates seem to be convenient molecular tools for protein transduction of widely used GST-fused proteins.

    DOI: 10.1093/jb/mvn087

    Web of Science

    researchmap

  • S100A11, an dual mediator for growth regulation of human keratinocytes

    Masakiyo Sakaguchi, Hiroyuki Sonegawa, Hitoshi Murata, Midori Kitazoe, Jun-ichiro Futami, Ken Kataoka, Hidenori Yamada, Nam-ho Huh

    MOLECULAR BIOLOGY OF THE CELL   19 ( 1 )   78 - 85   2008年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC CELL BIOLOGY  

    We previously revealed a novel signal pathway involving S100A11 for inhibition of the growth of normal human keratinocytes (NHK) caused by high Ca++ or transforming growth factor beta. Exposure to either agent resulted in transfer of S100A11 to nuclei, where it induced p21(WAF1). In contrast, S100A11 has been shown to be overexpressed in many human cancers. To address this apparent discrepancy, we analyzed possible new functions of S100A11, and we provide herein evidence that 1) S100A11 is actively secreted by NHK; 2) extracellular S100A11 acts on NHK to enhance the production of epidermal growth factor family proteins, resulting in growth stimulation; 3) receptor for advanced glycation end products, nuclear factor-kappa B, Akt, and cAMP response element-binding protein are involved in the S100A11-triggered signal transduction; and 4) production and secretion of S100A11 are markedly enhanced in human squamous cancer cells. These findings indicate that S100A11 plays a dual role in growth regulation of epithelial cells.

    DOI: 10.1091/mbc.E07-07-0682

    Web of Science

    researchmap

  • Transient cell proliferation with polyethylenimine-cationized N-terminal domain of simian virus 40 large T-antigen

    Hitoshi Murata, Junichiro Futami, Midori Kitazoe, Megumi Kosaka, Hiroko Tada, Masaharu Seno, Hidenori Yamada

    JOURNAL OF BIOSCIENCE AND BIOENGINEERING   105 ( 1 )   34 - 38   2008年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SOC BIOSCIENCE BIOENGINEERING JAPAN  

    Polyethylenimine (PEI) cationization is a powerful strategy for protein transduction into cells. In this study, we attempted the artificial regulation of cell proliferation by protein transduction of the N-terminal domain (1-132 amino acids) of the simian virus 40 large T-antigen (SVLT-N), which inactivates retinoblastoma family proteins but not p53. To deliver SVLT-N into cells, we employed an indirect cationization method by forming a complex of biotynylated SVLT-N through disulfide bonds (biotin-SS-SVLT-N) and PEI-cationized avidin (PEI600-avidin). Using this complex, SVLT-N was transduced into the nucleus of confluent and quiescent Balb/c 3T3 cells and was found to be complexed with a cellular target protein, pRb. Furthermore, SVLT-N transduction induced cell proliferation in spite of confluent conditions. Because SVLT-N thus transduced into cells gradually degraded and was not detectable after a 4-d incubation, transiently transformed cells were obtained by this method. These results suggest that oncogene protein transduction technology has great potential for in vitro regulation of cell proliferation.

    DOI: 10.1263/jbb.105.34

    Web of Science

    researchmap

  • Truncation of annexin A1 is a regulatory lever for linking epidermal growth factor signaling with cytosolic phospholipase A2 in normal and malignant squamous epithelial cells

    Masakiyo Sakaguchi, Hitoshi Murata, Hiroyuki Sonegawa, Yoshihiko Sakaguchi, Jun-ichiro Futami, Midori Kitazoe, Hidenori Yamada, Nam-ho Huh

    JOURNAL OF BIOLOGICAL CHEMISTRY   282 ( 49 )   35679 - 35686   2007年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Regulation of cell growth and apoptosis is one of the pleiotropic functions of annexin A1 (ANXA1). Although previous reports on the overexpression of ANXA1 in many human cancers and on growth suppression and/or induction of apoptosis by ANXA1 may indicate the tumor-suppressive nature of ANXA1, molecular mechanisms of the function of ANXA1 remain largely unknown. Here we provide evidence that ANXA1 mechanistically links the epidermal growth factor-triggered growth signal pathway with cytosolic phospholipase A(2) (cPLA(2)), an initiator enzyme of the arachidonic acid cascade, through interaction with S100A11 in normal human keratinocytes (NHK). Ca2+ -dependent binding of S100A11 to ANXA1 facilitated the binding of the latter to cPLA2, resulting in inhibition of cPLA2 activity, which is essential for the growth of NHK. On exposure of NHK to epidermal growth factor, ANXA1 was cleaved solely at Trp(12), and this cleavage was executed by cathepsin D. In squamous cancer cells, this pathway was shown to be constitutively activated. The newly found mechanistic intersection may be a promising target for establishing new measures against human cancer and other cell growth disorders.

    DOI: 10.1074/jbc.M707538200

    Web of Science

    researchmap

  • Exploiting protein cationization techniques in future drug development

    Junichiro Futami, Midori Kitazoe, Hiroshi Murata, Hidenori Yamada

    EXPERT OPINION ON DRUG DISCOVERY   2 ( 2 )   261 - 269   2007年2月

     詳細を見る

    記述言語:英語   出版者・発行元:INFORMA HEALTHCARE  

    The development of a method for the efficient intracellular delivery of inherently non-permeable proteins is needed for manipulation of cellular phenotypes or the discovery of protein-based drugs. It has been demonstrated that proteins artificially cationized by chemical conjugation show efficient intracellular delivery via adsorptive-mediated endocytosis and then can exert their biological activity in cells. Studies have also revealed that cationic peptides known as cell-penetrating peptides (CPPs) provide a means to deliver molecules into mammalian cells. Although the internalization mechanisms remain controversial, it is now becoming clear that the main port of entry into cells by CPPs also involves adsorptive-mediated endocytosis rather than the direct penetration of the plasma membrane. As the mammalian cell membrane possesses an abundance of negatively charged glycoproteins and glycosphingolipids, cationization of proteins is a reasonable choice to endow them with the ability for intracellular delivery. Cationization of proteins is usually accompanied by drastic changes in protein properties, structure and biological activities. Recently developed sophisticated protein chemistry can minimize these side effects. Therefore, protein cationization techniques will hopefully prove to be powerful tools for innovative research and drug discovery. In this review, techniques for cationization of proteins and their intracellular delivery, as well as some of their potential therapeutic applications, are discussed.

    DOI: 10.1517/17460441.2.2.261

    Web of Science

    researchmap

  • Denatured and reversibly cationized p53 readily enters cells and simultaneously folds to the functional protein in the cells

    H Murata, M Sakaguchi, J Futami, M Kitazoe, T Maeda, H Doura, M Kosaka, H Tada, M Seno, N Huh, H Yamada

    BIOCHEMISTRY   45 ( 19 )   6124 - 6132   2006年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER CHEMICAL SOC  

    Cationization is a powerful strategy for internalizing a protein into living cells. On the other hand, a reversibly cationized denatured protein through disulfide bonds is not only soluble in water but also able to fold to the native conformation in vitro. When these advantages in cationization were combined, we developed a novel method to deliver a denatured protein into cells and simultaneously let it fold to express its function within cells. This "in-cell folding" method enhances the utility of recombinant proteins expressed in Escherichia coli as inclusion bodies; that is, the recombinant proteins in inclusion bodies are solubilized by reversible cationization through cysteine residues by disulfide bonds with aminopropyl methanethiosulfonate or pyridyldithiopropionylpolyethylenimine and then incubated with cells without an in vitro folding procedure. As a model protein, we investigated human tumor-suppressor p53. Treatment of p53-null Saos-2 cells with reversibly cationized p53 revealed that all events examined as indications of the activation of p53 in cells, such as reduction of disulfide bonds followed by tetramer formation, localization into the nucleus, induction of p53 target genes, and induction of apoptosis of cells, occurred. These results suggest that reversible cationization of a denatured protein through cysteine residues is an alternative method for delivery of a functional protein into cells. This method would be very useful when a native folded protein is not readily available.

    DOI: 10.1021/bi052642a

    Web of Science

    researchmap

  • Protein transduction assisted by polyethylenimine-cationized carrier proteins

    M Kitazoe, H Murata, J Futami, T Maeda, M Sakaguchi, M Miyazaki, M Kosaka, H Tada, M Seno, N Huh, M Namba, M Nishikawa, Y Maeda, H Yamada

    JOURNAL OF BIOCHEMISTRY   137 ( 6 )   693 - 701   2005年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:JAPANESE BIOCHEMICAL SOC  

    Previously, we have reported that cationized-proteins covalently modified with polyethylenimine (PEI) (direct PEI-cationization) efficiently enter cells and function in the cytosol [Futami et al. (2005) J. Biosci. Bioeng. 99,95-1031. However, it may be more convenient if a protein could be delivered into cells just by mixing the protein with a PEI-cationized carrier protein having a specific affinity (indirect PEI-cationization). Thus, we prepared PEI-cationized avidin (PEI-avidin), streptavidin (PEI-streptavidin), and protein G (PEI-protein G), and examined whether they could deliver biotinylated proteins and antibodies into living cells. PEI-avidin (and/or PEI-streptavidin) carried biotinylated GFPs into various mammalian cells very efficiently. A GFP variant containing a nuclear localization signal was found to arrive even in the nucleus. The addition of a biotinylated RNase A derivative mixed with PEI-streptavidin to a culture medium of 3T3-SV-40 cells resulted in remarkable cell growth inhibition, suggesting that the biotinylated RNase A derivative entered cells and digested intracellular RNA molecules. Furthermore, the addition of a fluorescein-labeled antiS100C (beta-actin binding protein) antibody mixed with PEI-protein G to human fibroblasts resulted in the appearance of a fluorescence image of actin-like filamentous structures in the cells. These results indicate that indirect PEI-cationization using non-covalent interaction is as effective as the direct PEI-cationization for the transduction of proteins into living cells and for expression of their functions in the cytosol. Thus, PEI-cationized proteins having a specific affinity for certain molecules such as PEI-streptavidin, PEI-avidin and PEI-protein G are concluded to be widely applicable protein transduction carrier molecules.

    DOI: 10.1093/jb/mvi081

    Web of Science

    researchmap

  • Targeted disruption of transcriptional regulatory function of p53 by a novel efficient method for introducing a decoy oligonucleotide into nuclei

    M Sakaguchi, T Nukui, H Sonegawa, H Murata, J Futami, H Yamada, N Huh

    NUCLEIC ACIDS RESEARCH   33 ( 9 )   2005年

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:OXFORD UNIV PRESS  

    Decoy oligonucleotides have been used for functional sequestering of transcription factors. Efficient introduction into cells is a prerequisite for the oligonucleotides to exert their blocking function. Lipofection is the most widely used technique for that purpose because of its convenience and relatively high efficiency. However, the transduction efficiency of lipofection largely depends on cell types and experimental conditions and the introduced nucleotides are not specifically directed to nuclei where they exert their major function. In the present study, we designed a new system for transporting oligonucleotides into cell nuclei. The vehicle is composed of glutathione-S-transferase, 7 arginine residues, the DNA- binding domain of GAL4 and a nuclear localization signal, which are linked with flexible glycine stretches. The p53-responsive element linked to the GAL4 upstream activating sequence was efficiently transferred by the vehicle protein into nuclei of primary cultures of neuronal cells, embryonic stem cells and various human normal cells. Transcriptional activation of p21(WAF1/CIP1) and Bax by p53 on exposure to cisplatin was completely blocked by introducing the p53 decoy oligonucleotide. Thus, the system developed in the present study can be a convenient and powerful tool for specifically disrupting the function of DNA- binding proteins in culture.

    DOI: 10.1093/nar/gni088

    Web of Science

    researchmap

▼全件表示

MISC

  • 分泌性S100A11は膵臓癌の細胞運動性を高める腫瘍周囲の線維芽細胞を活性化する

    合原勇馬, 光井洋介, 友信奈保子, 木下理恵, 山本健一, 山内明, 山村真弘, 近藤英作, 豊岡伸一, 那須保友, 村田等, 阪口政清

    日本分子生物学会年会プログラム・要旨集(Web)   43rd   2020年

     詳細を見る

  • 分泌性S100A11-受容体RAGEシグナルに着眼した膵がん間質増大のメカニズムの解明

    山本 健一, 高松 仁, 光井 洋介, 木下 理恵, 村田 等, 二見 淳一郎, 山本 靖彦, 西堀 正洋, 豊岡 伸一, 阪口 政清

    日本がん免疫学会総会プログラム・抄録集   22回   117 - 117   2018年7月

     詳細を見る

    記述言語:日本語   出版者・発行元:日本がん免疫学会  

    researchmap

  • 膵がん進展に導く膵がん細胞 間質線維芽細胞クロストークを介在する分泌性S100A11 受容体RAGE連携の役割

    光井 洋介, 山本 健一, Sumardika I Wayan, 木下 理恵, 村田 等, 二見 淳一郎, 高松 仁, 山本 靖彦, 西堀 正洋, 豊岡 伸一, 渡部 昌実, 那須 保友, 阪口 政清

    日本がん免疫学会総会プログラム・抄録集   22回   156 - 156   2018年7月

     詳細を見る

    記述言語:日本語   出版者・発行元:日本がん免疫学会  

    researchmap

  • Topics from special edition 転移先臓器を感知する受容体群の分子制御機構の解明とその応用

    阪口 政清, 木下 理恵, 村田 等, 山本 健一, 許 南浩, 日比野 利彦

    細胞   49 ( 7 )   359 - 362   2017年6月

     詳細を見る

    記述言語:日本語   出版者・発行元:ニューサイエンス社  

    CiNii Article

    CiNii Books

    researchmap

    その他リンク: http://search.jamas.or.jp/link/ui/2017264472

  • Topics from special edition 転移先臓器を感知する受容体群

    阪口 政清, 木下 理恵, 村田 等, 山本 健一, 許 南浩, 日比野 利彦

    細胞   49 ( 3 )   143 - 146   2017年3月

     詳細を見る

    記述言語:日本語   出版者・発行元:ニューサイエンス社  

    CiNii Article

    CiNii Books

    researchmap

    その他リンク: http://search.jamas.or.jp/link/ui/2017181594

  • 転移先臓器を感知する受容体群の分子制御機構の解明とその応用. Advanced approach to prevent cancer metastasis based on S100A8/A9-mediated seed and soil theory Innovative approach to prevent cancer metastasis based on S100A8/A9-mediated seed and soil theory.

    阪口政清, 木下理恵, 村田 等, 山本健一, 許 南浩, 日比野 利彦

    月刊「細胞」6月号   49 ( 7 )   43 - 46   2017年

     詳細を見る

  • 転移先臓器を感知する受容体群Innovative approach to prevent cancer metastasis based on S100A8/A9-mediated seed and soil theory.

    阪口政清, 木下理恵, 村田 等, 山本健一, 許 南浩, 日比野 利彦

    月刊「細胞」3月号   49 ( 3 )   39 - 42   2017年

     詳細を見る

  • Neuroplastinβとextracellular matrix metalloprotease inducerはS100A8/A9に対する機能的な受容体であり,ケラチノサイトの増殖とアトピーの皮膚炎症の増強に関与している. Critical role of novel receptors to S100A8/A9, EMMPRIN and NPTNβ, on keratinocyte growth and inflammation in atopic dermatitis.

    阪口政清, 山本真実, 宮井雅史, 松元 有羽子, 木下理恵, 村田 等, 山本健一, 森実 真, 岩月啓氏, 許 南浩, 坪井良治, 日比野 利彦

    臨床免疫   49 ( 6 )   594 - 598   2017年

     詳細を見る

    記述言語:日本語   出版者・発行元:科学評論社  

    CiNii Article

    CiNii Books

    researchmap

    その他リンク: http://search.jamas.or.jp/link/ui/2017265401

  • 【アトピー性皮膚炎の病態研究】 Neuroplastin βとextracellular matrix metallo-protease inducerはS100A8/A9に対する機能的な受容体であり、ケラチノサイトの増殖とアトピーの皮膚炎症の増強に関与している

    阪口 政清, 山本, 真実, 宮井 雅史, 木下 理恵, 村田 等, 山本 健一, 森実 真, 岩月 啓氏, 許 南浩, 坪井 良治, 日比野 利彦

    臨床免疫・アレルギー科   67 ( 6 )   594 - 598   2017年

     詳細を見る

  • Tumor Necrosis Factor-alpha Down-regulates a Tumor Suppressor Gene, REIC/Dkk-3, in Normal Skin Keratinocytes

    K. Kataoka, M. Sakaguchi, H. Murata, N. Hashikawa, N. Huh

    IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY-ANIMAL   49   S43 - S43   2013年6月

     詳細を見る

    記述言語:英語   掲載種別:研究発表ペーパー・要旨(国際会議)   出版者・発行元:SPRINGER  

    Web of Science

    researchmap

  • TRAF6 ubiquitinates and stabilizes PINK1 on the outer membrane of depolarized mitochondria through interaction with SARM1

    H. Murata, M. Sakaguchi, K. Kataoka, N-H. Huh

    MOLECULAR BIOLOGY OF THE CELL   24   2013年

     詳細を見る

    記述言語:英語   掲載種別:研究発表ペーパー・要旨(国際会議)   出版者・発行元:AMER SOC CELL BIOLOGY  

    Web of Science

    researchmap

  • Identification of RAGE-coupled membran adaptor that modulates RAGE-triggered signaling pathway

    M. Sakaguchi, H. Murata, T. Hibino, Y. Aoyama, K. Iwatsuki, N-H. Huh

    MOLECULAR BIOLOGY OF THE CELL   24   2013年

     詳細を見る

    記述言語:英語   掲載種別:研究発表ペーパー・要旨(国際会議)   出版者・発行元:AMER SOC CELL BIOLOGY  

    Web of Science

    researchmap

  • S100A8およびS100A9タンパク質の新規受容体の探索とそのがん進展における役割(Identification of novel receptors for pro-inflammatory S100A8/A9 proteins and their potential roles in tumor progression)

    阪口 政清, 日比野 利彦, 村田 等, 井上 裕介, 山本 健一, 片岡 健, 許 南浩

    日本癌学会総会記事   71回   59 - 59   2012年8月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • A Novel Tumor Suppressor, REIC/Dkk-3 Gene Identified by Our In Vitro Transformation Model of Normal Human Fibroblasts Works as a Potent Therapeutic Anti-tumor Agent

    Agent. M. Sakaguchi, K. Kataoka, H. Murata, M. Namba, N. H. Huh

    IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY-ANIMAL   48   17 - 17   2012年7月

     詳細を見る

    記述言語:英語   掲載種別:研究発表ペーパー・要旨(国際会議)   出版者・発行元:SPRINGER  

    Web of Science

    researchmap

  • TRAF6とSARM1によるPINK1のユビキチン化はマイトファジーの誘導に必須である

    村田等, 阪口政清, 片岡健, HUH Nam‐ho

    日本分子生物学会年会プログラム・要旨集(Web)   35th   4P-0506 (WEB ONLY)   2012年

     詳細を見る

    記述言語:日本語  

    J-GLOBAL

    researchmap

  • 4Ca14 カチオン化アビジンを介したビオチン化タンパク質細胞導入法における導入効率の最適化(酵素学,酵素工学/タンパク質工学,一般講演)

    二見 翠, 渡邉 泰宜, 村田 等, 多田 宏子, 山田 秀徳, 二見 淳一郎

    日本生物工学会大会講演要旨集   64   194 - 194   2012年

     詳細を見る

    記述言語:日本語   出版者・発行元:日本生物工学会  

    CiNii Article

    CiNii Books

    researchmap

    その他リンク: http://search.jamas.or.jp/link/ui/2013125670

  • S100A7による骨肉腫細胞の遊走・浸潤の多機能受容体RAGEを介した制御機構(Promotion of migration and invasion of osteosarcoma cells by S100A7 through RAGE)

    片岡 健, 阪口 政清, 小野 智之, 森下 美加, 山本 健一, 村田 等, 許 南浩

    日本癌学会総会記事   70回   79 - 79   2011年9月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • 多機能受容体RAGEの下流信号伝達機構の解明(TIRAP is a critical transducer of RAGE-mediated inflammatory signaling)

    阪口 政清, 村田 等, 山本 健一, 阪口 義彦, 本山 晃, 日比野 利彦, 片岡 健, 許 南浩

    日本癌学会総会記事   70回   225 - 225   2011年9月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • PINK1発現抑制によるミトコンドリア機能不全はREIC/Dkk-3のアポトーシス誘導効果を増強する(Mitochondrial malfunction by PINK1 knockdown augments apoptosis induced by adenovirus carrying REIC/Dkk-3)

    村田 等, 金 玉, 阪口 政清, 片岡 健, 許 南浩

    日本癌学会総会記事   70回   51 - 51   2011年9月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • REIC/Dkk-3の扁平上皮における発現とその制御因子の探索

    片岡 健, 阪口 政清, 杜 剛, 前原 奈都美, 村田 等, 許 南浩

    組織培養研究   30 ( 1 )   99 - 99   2011年3月

     詳細を見る

    記述言語:日本語   出版者・発行元:日本組織培養学会  

    researchmap

  • 正常皮膚におけるREIC/Dkk‐3の発現制御因子の検索

    前原奈都美, 片岡健, DU Gang, 村田等, 山本健一, 阪口政清, HUH Nam‐Ho

    日本分子生物学会年会プログラム・要旨集(Web)   34th   1P-0254 (WEB ONLY)   2011年

     詳細を見る

    記述言語:日本語  

    J-GLOBAL

    researchmap

  • S100タンパク質受容体の結合解析

    森下美加, 村田等, 山本健一, 片岡健, 阪口政清, HUH Nam‐Ho

    日本分子生物学会年会プログラム・要旨集(Web)   34th   4P-0293 (WEB ONLY)   2011年

     詳細を見る

    記述言語:日本語  

    J-GLOBAL

    researchmap

  • BRPK/PINK1のmTORC2経路活性化を介したがん進展への寄与(BRPK/PINK1 promotes tumor progression through activation of mTORC2 pathway)

    村田 等, 阪口 政清, 金 玉, 片岡 健, 許 南浩

    日本生化学会大会・日本分子生物学会年会合同大会講演要旨集   83回・33回   4P - 0934   2010年12月

     詳細を見る

    記述言語:英語   出版者・発行元:(公社)日本生化学会  

    researchmap

  • スキルス胃がん腹膜播種へのREIC/Dkk-3遺伝子治療の効果(REIC/Dkk-3 gene therapy suppresses peritoneal dissemination of scirrhous gastric carcinoma)

    片岡 健, タン・スエスエ, 阪口 政清, アバルスァ・フェルナンド, 村田 等, 八代 正和, 那須 保友, 公文 裕巳, 許 南浩

    日本癌学会総会記事   69回   252 - 253   2010年8月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • 分化誘導したマウスES細胞及びiPS細胞におけるCy3標識REIC/Dkk‐3タンパク質のエンドサートーシスによる取り込み

    片岡健, 阪口政清, LI Kun Peng, DU Gang, 武田千佳, 舟橋弘晃, 村田等, HUH Nam‐Ho

    組織培養研究   29 ( 1 )   74 - 74   2010年3月

     詳細を見る

    記述言語:日本語   出版者・発行元:日本組織培養学会  

    J-GLOBAL

    researchmap

  • BRPK/PINK1のmTORC2経路活性化を介したがん浸潤への寄与(BRPK/PINK1 enhances invasiveness of cancer cells trough activation of mTORC2 pathway)

    村田 等, 阪口 政清, 片岡 健, 許 南浩

    日本癌学会総会記事   68回   205 - 205   2009年8月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • BRPK/PINK1のmTORC2活性化を介したがん進展への寄与

    村田 等, 阪口 政清, 片岡 健, 許 南浩

    組織培養研究   28 ( 1 )   66 - 66   2009年3月

     詳細を見る

    記述言語:日本語   出版者・発行元:日本組織培養学会  

    researchmap

  • REIC/Dkk-3の強制発現はERストレスを介してIL-7を誘導する(Over-expression of REIC/Dkk-3 induces IL-7 in normal human fibroblasts via ER stress)

    阪口 政清, 片岡 健, アバルスア・フェルナンド, 谷本 竜太, 渡辺 昌実, 村田 等, 那須 保友, 公文 裕己, 許 南浩

    日本癌学会総会記事   67回   238 - 238   2008年9月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • BRPK/PINK1はAktのリン酸化によってがん細胞のアポトーシスを抑制する(BRPK/PINK1 blocks apoptotic cell death of cancer cells by phosphotylation of Akt)

    村田 等, 阪口 政清, 片岡 健, 許 南浩

    日本癌学会総会記事   67回   64 - 64   2008年9月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • REIC/Dkk-3遺伝子治療の正常細胞を介する抗腫瘍効果(Transduction of gene therapeutic REIC/Dkk-3 into normal cells provides another arm for tumor suppression in vivo)

    片岡 健, 阪口 政清, Fernando Abarzua, 谷本 竜太, 渡部 昌実, 村田 等, 那須 保友, 公文 裕巳, 許 南浩

    日本癌学会総会記事   67回   93 - 93   2008年9月

     詳細を見る

    記述言語:英語   出版者・発行元:日本癌学会  

    researchmap

  • S100A11 functions as a dual mediator for growth regulation in normal human keratinocytes

    M. Sakaguchi, H. Murata, N. Huh

    JOURNAL OF INVESTIGATIVE DERMATOLOGY   128   S138 - S138   2008年4月

     詳細を見る

    記述言語:英語   掲載種別:研究発表ペーパー・要旨(国際会議)   出版者・発行元:NATURE PUBLISHING GROUP  

    Web of Science

    researchmap

  • 2B11-4 ポリエチレンイミン(PEI)-グルタチオンキャリアーを用いたGST-融合タンパク質の細胞導入(酵素学・酵素工学・タンパク質工学,一般講演)

    村田 等, 二見 淳一郎, 北添 翠, 小坂 恵, 多田 宏子, 甲斐 敬, 妹尾 昌治, 山田 秀徳

    日本生物工学会大会講演要旨集   19   63 - 63   2007年

     詳細を見る

    記述言語:日本語   出版者・発行元:日本生物工学会  

    CiNii Article

    CiNii Books

    researchmap

  • 1C16-5 蛋白質カチオン化による細胞内導入技術開発と機構解明(酵素学・酵素工学・タンパク質工学,一般講演)

    二見 淳一郎, 北添 翠, 村田 等, 渡邉 泰宣, 八木 康行, 多田 宏子, 妹尾 昌治, 甲斐 敬, 山田 秀徳

    日本生物工学会大会講演要旨集   17   103 - 103   2005年

     詳細を見る

    記述言語:日本語   出版者・発行元:日本生物工学会  

    CiNii Article

    CiNii Books

    researchmap

▼全件表示

講演・口頭発表等

  • 軸索変性誘導分子SARM1の阻害剤開発

    村田 等, 安藤 隆幸, 大磯 和真, 友信 奈保子, 山本 健一, 木下 理恵, 阪口 政清

    NEURO2022  2022年6月30日 

     詳細を見る

    開催年月日: 2022年6月30日 - 2022年7月3日

    記述言語:英語   会議種別:ポスター発表  

    researchmap

  • REIC/Dkk-3の受容体への結合はトリプルネガティブ乳がんのPD-L1の発現を抑制する

    吉澤 智香子, 合原 勇馬, 友信 奈保子, 木下 理恵, 二見 淳一郎, 村田 等, 山本 健一, 阪口 政清

    第45回 日本分子生物学会年会  2022年12月1日 

     詳細を見る

  • 神経軸索変性メカニズムの解明と創薬への応用 招待

    村田 等

    第37回 創薬・薬理フォーラム岡山  2022年7月23日 

     詳細を見る

    会議種別:口頭発表(招待・特別)  

    researchmap

  • 炎症性疾患に対する抗S100A8/A9抗体療法におけるマクロファージの役割

    木下理恵, 小林和子, 荒木恒太, 佐藤博紀, 友信奈保子, 村田等, 許南浩, 豊岡伸一, 阪口政清

    日本組織培養学会 第94回大会  2022年7月8日 

     詳細を見る

  • Lysyl oxidase-like 4 is prominent in breast cancer progression through its en]ymatic activities

    Ni Luh Gede, Yoni Komalasari, Nahoko Tomonobu, Fan Jiang, Acosta Gonzalez, Herik Rodrigo, Yuma Gohara, Ken-ichi Yamamoto, Rie Kinoshita, Hitoshi Murata, Yusuke Inoue, Masakiyo Sakaguchi

    日本組織培養学会 第94回大会  2022年7月8日 

     詳細を見る

  • REIC protein suppresses tumor progression through PD-L1 regulation in cancer cells

    Yuma Gohara, Nahoko Tomonobu, Rie Kinoshita, Hitoshi Murata, Ken-ichi Yamamoto, Masayoshi Namba, Nam-ho Huh, Hiromi Kumon, Masakiyo Sakaguchi

    日本組織培養学会 第94回大会  2022年7月8日 

     詳細を見る

  • 疾患特異的iPS細胞と動物モデルを用いたパーキンソン病における軸索変性誘導分子SARM1のリン酸化制御解析

    村田 等, 越智 俊樹, 友信 奈保子, 山本 健一, 木下 理恵, 阪口 政清

    日本組織培養学会 第93回大会  2021年9月3日 

     詳細を見る

  • S100A8/A9を標的とした炎症性疾患に対する治療法の開発

    木下 理恵, 荒木 恒太, 佐藤 博紀, 友信 奈保子, 村田 等, 山本 健一, 許 南浩, 豊岡 伸一, 阪 口 政清

    日本組織培養学会 第93回大会  2021年9月3日 

     詳細を見る

  • キシリトールの細胞内グルタチオン調節によるがん選択的細胞死誘導機序の解明

    友信 奈保子, 木下 理恵, 山本 健一, 村田 等, 阪口 政清

    日本組織培養学会 第93回大会  2021年9月2日 

     詳細を見る

  • ロテノン誘発性パーキンソニズムの病態形成におけるSARM1リン酸化制御の意義

    村田 等, 越智 俊樹, 山本 健一, 木下 理恵, 阪口 政清

    第43回 日本分子生物学会年会  2020年12月2日 

     詳細を見る

  • 分泌性S100A11は膵臓癌の細胞運動性を高める腫瘍周囲の線維芽細胞を活性化する

    合原 勇馬, 光井 洋介, 友信 奈保子, 木下 理恵, 山本 健一, 山内 明, 山村 真弘, 近藤 英作, 豊岡 伸一, 那須 保友, 村田 等, 阪口 政清

    第43回 日本分子生物学会年会  2020年12月2日 

     詳細を見る

  • JNKによるリン酸化を介したSARM1のNAD+代謝と軸索変性への寄与 招待

    村田等, 阪口政清

    第92回日本生化学会大会  2019年9月18日 

     詳細を見る

    記述言語:日本語   会議種別:シンポジウム・ワークショップ パネル(指名)  

    researchmap

  • Regulation of SARM1-NAD+ cleavage activity and mitochondrial function through phosphorylation by JNK

    村田等, 山本健一, 木下理恵, 阪口政清

    Neuro2019  2019年7月25日 

     詳細を見る

    記述言語:英語   会議種別:ポスター発表  

    researchmap

  • 分泌性S100A11-受容体RAGEシグナルを介した膵臓がん周辺微小環境における間質繊維芽細胞の増殖誘導の解明

    山本健一, 高松仁志, 友信奈保子, 光井洋介, 二見淳一郎, 木下理恵, 村田等, 阪口政清

    第41回日本分子生物学会年会  2018年11月30日 

     詳細を見る

    記述言語:日本語   会議種別:ポスター発表  

    researchmap

  • JNKによるリン酸化はSARM1のNAD分解活性を制御し、ミトコンドリア呼吸阻害を誘導する

    村田等, 山本健一, 木下理恵, 阪口政清

    第41回日本分子生物学会年会  2018年11月27日 

     詳細を見る

    記述言語:日本語  

    researchmap

  • ミトコンドリア呼吸鎖複合体への作用を介したSARM1の神経細胞死誘導

    第90回日本組織培養学会大会  2017年 

     詳細を見る

  • S100A8を標的としたがん転移制御法の開発

    日本組織培養学会第90回大会  2017年 

     詳細を見る

  • 健常者及びパーキンソン病患者iPS細胞由来の神経細胞を用いたミトコンドリア機能解析

    2017年度生命科学系学会合同年次大会  2017年 

     詳細を見る

  • 転移先臓器を感知する受容体

    第90回日本組織培養学会大会  2017年 

     詳細を見る

  • Development of a novel biologics for suppression of S100A8/A9-induced cancer metastasis

    第75回日本癌学会学術総会  2016年 

     詳細を見る

  • 神経細胞死・軸索変性に関与するSARM1のリン酸化制御

    第39回日本分子生物学会年会  2016年 

     詳細を見る

  • Regulation of PINK1 expression under oxidative stress conditions

    第89回日本組織培養学会大会  2016年 

     詳細を見る

  • Extract of Cordyceps militaris inhibits angiogenesis and suppresses tumor growth of human malignant melanoma cells

    第89回日本組織培養学会大会  2016年 

     詳細を見る

  • PINK1 regulation of mitochondrial homeostasis and cell survival

    World congress on in vitro biology 2016  2016年 

     詳細を見る

  • NRF2 pathway activation as a target to counteract mitochondrial dysfunction in Parkinson's disease

    ICME international conference on complex medical engineering 2015  2015年 

     詳細を見る

  • Regulation of PINK1 expression through the NRF2 transcription factor under mitochondrial stress conditions

    Biochemical Society PINK1-Parkin signaling in Parkinson’s disease and beyond  2014年 

     詳細を見る

  • Transcriptional regulation of PINK1 by NRF2 under mitochondrial stress conditions

    第37回日本分子生物学会年会  2014年 

     詳細を見る

  • PINK1のSARM1、TRAF6との複合体形成及びユビキチン化によるミトコンドリア局在制御とマイトファジー誘導

    第36回日本分子生物学会年会  2013年 

     詳細を見る

  • Identification of RAGE-coupled membran adaptor that modulates RAGE-triggered signaling pathway

    2013年アメリカ細胞生物学会総会  2013年 

     詳細を見る

  • TRAF6 ubiquitinates and stabilizes PINK1 on the outer membrane of depolarized mitochondria through interaction with SARM1

    The American Society for Cell Biology 53th Annual Meeting  2013年 

     詳細を見る

  • カチオン化を利用したタンパク質細胞内導入技術の開発

    JST推薦シーズ新技術説明会第2回ライフイノベーション分野  2013年 

     詳細を見る

  • Regulation of Cellular Stress Response By Mitochondrial Kinase PINK1

    日本組織培養学会第86回大会  2013年 

     詳細を見る

  • S100A8/A9 targets oncogenic survival pathway via RAGE that critically enhanced by a RAGE co-receptor. 多機能受容体RAGE の共役受容体の発見とその意義

    第72回日本癌学会学術総会  2013年 

     詳細を見る

  • TRAF6 and SARM1 regulate PINK1 ubiquitination and stabilization on depolarized mitochondria for mitophagy

    International symposium on mitochondria 2013  2013年 

     詳細を見る

  • Identification of novel receptors for pro-inflammatory S100A8/A9 proteins and their roles inmelanoma metastasis

    IEIIS2012. Homeostatic Inflammation Symposium  2012年 

     詳細を見る

  • Development of RAGE-inhibitor delivered into cells.

    第85回日本組織培養学会大会  2012年 

     詳細を見る

  • A Novel Tumor Suppressor, REIC/Dkk-3 Gene Identified by our in Vitro Transformation Model of Normal Human Fibroblasts Works as a Potent Therapeutic Anti-tumor Agent

    2012 World Congress on In Vitro Biology  2012年 

     詳細を見る

  • 細胞内導入型RAGE阻害ペプチドの開発

    第2回日本生物工学会西日本支部講演会  2012年 

     詳細を見る

  • TRAF6 ubiquitinates and stabilizes PINK1 on damaged mitochondria through interaction with SARM1

    第35回日本分子生物学会年会  2012年 

     詳細を見る

  • DOCK7 is a critical regulator of the RAGE-Cdc42 signaling axis that induces formation of dendritic pseudopodia in human cancer cells

    第35回日本分子生物学会年会  2012年 

     詳細を見る

  • S100A8およびS100A9タンパク質の新規受容体の探索とそのがん進展における役割

    第71回日本癌学会学術総会  2012年 

     詳細を見る

  • カチオン化を利用したタンパク質DDS

    イノベーション・ジャパン2012  2012年 

     詳細を見る

  • S100タンパク質受容体の結合解析

    第34回日本分子生物学会年会  2011年 

     詳細を見る

  • BRPK/PINK1 inhibits apoptotic cell death and enhances cellular invasiveness through an activation of mTORC2 pathway

    平成23年度がん若手研究者ワークショップ  2011年 

     詳細を見る

  • TIRAP is a critical transducer of RAGE-mediated inflammatory signaling

    2011年 

     詳細を見る

  • PINK1 knockdown increases the apoptosis induced by adenovirus-mediated REIC/Dkk-3 overexpression on bladder cancer cells

    第84回日本組織培養学会大会  2011年 

     詳細を見る

  • REIC/Dkk-3 の扁平上皮における発現とその制御因子の探索

    第84回日本組織培養学会大会  2011年 

     詳細を見る

  • S100A7による骨肉腫細胞の遊走・浸潤の多機能受容体RAGEを介した制御機構

    第70回日本癌学会学術総会  2011年 

     詳細を見る

  • 多機能受容体RAGEの下流信号伝達機構の解明

    第70回日本癌学会学術総会  2011年 

     詳細を見る

  • Mitochondrial dysfunction by PINK1 knockdown augments apoptosis induced by gene-therapeutic adenovirus carrying REIC/Dkk-3.

    第34回日本分子生物学会年会  2011年 

     詳細を見る

  • 正常皮膚におけるREIC/Dkk-3の発現制御因子の検索

    第34回日本分子生物学会年会  2011年 

     詳細を見る

  • Mitochondrial malfunction by PINK1 knockdown augments apoptosis induced by adenovirus carrying REIC/Dkk-3.

    第70回日本癌学会学術総会  2011年 

     詳細を見る

  • 多機能受容体RAGEの下流信号伝達機構の解明

    第70回日本癌学会学術総会  2011年 

     詳細を見る

  • BRPK/PINK1 promotes tumor progression trough activation of mTORC2 pathway.

    第33回日本分子生物学会年会第83回日本生化学大会合同大会  2010年 

     詳細を見る

  • Effect of Adenovirus-mediated Overexpression of REIC/Dkk-3 on Scirrhous Gastric Carcinoma Cells

    2010年 

     詳細を見る

  • タンパク質カチオン化技術を活用した細胞機能制御

    日本組織培養学会 第83回大会  2010年 

     詳細を見る

  • Endocytotic internalization of Cy3-labeled REIC/Dkk-3 protein by differentiated ES cells and iPS cells

    2010年 

     詳細を見る

  • Effect of Adenovirus-mediated Overexpression of REIC/Dkk-3 on Scirrhous Gastric Carcinoma Cells

    日本組織培養学会 第83回大会  2010年 

     詳細を見る

  • 神経変性疾患とがんをつなぐ分子 ~BRPK/PINK1~

    第13回生物工学若手研究者セミナー  2010年 

     詳細を見る

  • REIC/Dkk-3 gene therapy suppresses peritoneal dissemination of scirrhous gastric carcinoma

    第69回日本癌学会学術総会  2010年 

     詳細を見る

  • PINK1/BRPK inhibits apoptotic cell death and enhances cellular invasiveness through an activation of mTORC2 pathway.

    16th International Charles Heidelberger Symposium on Cancer Research,  2010年 

     詳細を見る

  • 分化誘導したマウスES細胞及びiPS細胞におけるCy3標識REIC/Dkk-3タンパク質のエンドサートーシスによる取り込み

    日本組織培養学会 第83回大会  2010年 

     詳細を見る

  • BRPK/PINK1のmTORC2活性化を介したがん進展への寄与

    第82回日本組織培養学会大会  2009年 

     詳細を見る

  • Overexpression of REIC/Dkk-3 in normal human fibroblasts suppresses tumor growth via induction of IL-7.

    第10回文部科学省特定領域研究「がん」5領域若手研究者ワークショップ  2009年 

     詳細を見る

  • BRPK/PINK1 enhances invasiveness of cancer cells trough activation of mTORC2 pathway.

    第68回日本癌学会学術総会  2009年 

     詳細を見る

  • BRPK/PINK1 blocks apoptotic cell death of cancer cells by phosphorylation of Akt.

    第67回日本癌学会学術総会  2008年 

     詳細を見る

  • ポリエチレンイミン-グルタチオンキャリアーを用いたGST融合タンパク質細胞導入技術の開発

    第31回蛋白質と酵素の構造と機能に関する九州シンポジウム  2007年 

     詳細を見る

  • ポリエチレンイミン(PEI)グルタチオンキャリアーを用いたGST-融合タンパク質の細胞導入

    第59回日本生物工学会大会  2007年 

     詳細を見る

  • Truncation of annexin A1 is a regulatory lever for linking epidermal growth factor signaling with cytosolic phospholipase A2 in normal and malignant squamous epithelial cells.

    The American Society for Cell Biology 47th Annual Meeting  2007年 

     詳細を見る

  • Artificial regulation of cell proliferation by protein transduction of N-terminal domain of simian virus 40 large T antigen using PEI-cationization method.

    20th IUBMB international congress of biochemistry and molecular biology.  2006年 

     詳細を見る

  • Applications of intracellular delivery of a protein by cationization. From in vitro folding to in cell folding of denatured proteins.

    A Satellite Meeting of International Conference of 43rd Japanese Peptide Symposium and 4th Peptide Engineering Meeting  2006年 

     詳細を見る

  • PEIカチオン化法を利用したSV40ラージT抗原N末端ドメインによる細胞増殖の人工制御

    第29回蛋白質と酵素の構造と機能に関する九州シンポジウム  2005年 

     詳細を見る

  • Artificial Control of Cell Proliferation Using an N-terminal Domain of Simian Virus 40 Large T Antigen by Means of PEI-cationization.

    The American Society for Cell Biology 45th Annual Meeting  2005年 

     詳細を見る

  • カチオン化SV40T抗原N末端ドメインによる細胞増殖の人工制御

    第27回分子生物学会年会  2004年 

     詳細を見る

  • Reversible cationization, a new method for delivery of a denatured protein into living cells followed by intracellular folding. Application to p53.

    International symposium on nano-biotechnology.  2004年 

     詳細を見る

  • タンパク質導入におけるクロロキンの効果

    第28回蛋白質と酵素の構造と機能に関する九州シンポジウム  2004年 

     詳細を見る

  • Reversible cationization, a new method for delivery of a denatured protein into living cells. Application to p53.

    第76回日本生化学会大会  2003年 

     詳細を見る

  • 可逆的カチオン化による変性タンパク質の細胞導入とそれに伴う機能発現

    第27回蛋白質と酵素の構造と機能に関する九州シンポジウム  2003年 

     詳細を見る

▼全件表示

産業財産権

  • リン酸化SARM1、抗体、SARM1リン酸化阻害剤、神経変性疾患の予防又は治療薬、スクリーニング方法、SARM1改変体及び使用

    村田 等, 阪口 政清, 木下 理恵, 山本 健一

     詳細を見る

    出願人:国立大学法人 岡山大学

    出願番号:特願2018-507366  出願日:2017年3月22日

    公表番号:WO2017-164230  公表日:2017年9月28日

    特許番号/登録番号:特許第7198489号 

    J-GLOBAL

    researchmap

  • 遺伝子発現用カセット及びその産生物

    阪口 政清, 西堀 正洋, 村田 等, 山本 健一, 木下 理恵

     詳細を見る

    出願人:国立大学法人 岡山大学

    出願番号:特願2016-059297  出願日:2016年3月23日

    公開番号:特開2017-169486  公開日:2017年9月28日

    特許番号/登録番号:特許第6871679号  登録日:2021年4月20日 

    J-GLOBAL

    researchmap

  • PINK1のユビキチン化アッセイ及びスクリーニングへの利用

    許 南浩, 村田 等, 阪口 政清

     詳細を見る

    出願人:国立大学法人 岡山大学

    出願番号:特願2012-165160  出願日:2012年7月25日

    公開番号:特開2014-025764  公開日:2014年2月6日

    特許番号/登録番号:特許第6024953号  登録日:2016年10月21日 

    特許第6024953号

    J-GLOBAL

    researchmap

  • タンパク質の細胞内導入剤

    山田 秀徳, 村田 等, 二見 淳一郎, 阪口 政清, 許 南浩, 八木 康行, 甲斐 敬

     詳細を見る

    出願人:株式会社日本触媒

    出願番号:特願2007-046025  出願日:2007年2月26日

    公開番号:特開2008-115150  公開日:2008年5月22日

    特許番号/登録番号:特許第5097412号  登録日:2012年9月28日 

    5097412

    J-GLOBAL

    researchmap

  • 細胞増殖剤及び細胞の増殖方法

    山田 秀徳, 二見 淳一郎, 村田 等, 阪口 政清, 許 南浩, 八木 康行, 甲斐 敬

     詳細を見る

    出願人:株式会社日本触媒

    出願番号:特願2005-356631  出願日:2005年12月9日

    公開番号:特開2007-159429  公開日:2007年6月28日

    2007-159429

    J-GLOBAL

    researchmap

受賞

  • 日本分子生物学会年会優秀ポスター賞

    2016年  

     詳細を見る

    受賞国:日本国

    researchmap

  • 岡山医学会賞(結城賞)

    2013年  

     詳細を見る

    受賞国:日本国

    researchmap

  • 日本組織培養学会奨励賞

    2009年  

     詳細を見る

    受賞国:日本国

    researchmap

  • 岡山大学大学院自然科学研究科長賞

    2007年  

     詳細を見る

    受賞国:日本国

    researchmap

  • 岡山大学ベンチャー・ビジネス・ラボラトリー研究企画賞

    2005年  

     詳細を見る

    受賞国:日本国

    researchmap

  • 第27回 蛋白質と酵素の構造と機能に関する九州シンポジウムポスター大賞

    2003年  

     詳細を見る

    受賞国:日本国

    researchmap

▼全件表示

共同研究・競争的資金等の研究

  • S100A8/A9-向転移とHRG-抗転移の細胞間・分子間クロストークの解明

    研究課題/領域番号:23H02748  2023年04月 - 2026年03月

    日本学術振興会  科学研究費助成事業  基盤研究(B)

    阪口 政清, 山本 健一, 近藤 英作, 豊岡 伸一, 木下 理恵, 西堀 正洋, 山内 明, 友信 奈保子, 村田 等

      詳細を見る

    配分額:18720000円 ( 直接経費:14400000円 、 間接経費:4320000円 )

    researchmap

  • 軸索変性誘導分子SARM1の活性・分解制御によるパーキンソン病治療法の開発

    研究課題/領域番号:22K06884  2022年04月 - 2025年03月

    日本学術振興会  科学研究費助成事業 基盤研究(C)  基盤研究(C)

    村田 等, 阪口 政清

      詳細を見る

    配分額:4160000円 ( 直接経費:3200000円 、 間接経費:960000円 )

    researchmap

  • 新規S100A8/A9阻害分子の発見に基づいたがん脳指向転移の機構解明とその制御

    研究課題/領域番号:20H03516  2020年04月 - 2023年03月

    日本学術振興会  科学研究費助成事業 基盤研究(B)  基盤研究(B)

    阪口 政清, 山本 健一, 近藤 英作, 豊岡 伸一, 木下 理恵, 西堀 正洋, 村田 等

      詳細を見る

    配分額:17420000円 ( 直接経費:13400000円 、 間接経費:4020000円 )

    S100A8/A9誘導性脳指向性転移をHRGがどのように阻害するかの分子原理の解明を目指した。HRGがS100A8/A9吸着能を示す発見し、HRGの受容体の発見を切り口に原理解明を進めている。本年度の判明した事象を示す。①HRGはS100A8/A9を吸着し、がん細胞へのS100A8/A9の浸潤、走化性亢進を抑制するが、HRGはS100A9側に親和性を有する。②HRGはがん細胞の血清誘導性浸潤、走化性亢進をも抑制することから、S100A8/A9吸着性によらない抑制効能をも有する。③がん細胞にHRG受容体が存在する仮説を立て、その候補受容体を独自スクリーニングから2つ見出した。一つはCLEC1A(1回膜貫通型)で好中球と血管内皮細胞に発現し、HRGにより活性が制御される。④好中球のネットーシスより引き起こされたS100A8/A9は、がん細胞を刺激し、増殖ならびに浸潤、走化性を亢進するが、これをHRGが抑制する。これにはHRGの好中球CLEC1Aへの作用(ネットーシス抑制)とS100A8/A9吸着作用が大きく関わる。⑤見出したもう一つの受容体候補は7開幕貫通型で、これはがん細胞側に発現している。現在この受容体を糸口にS100A8/A9吸着性によらないがん細胞抑制機構の詳細を検討している。さらに計画にはないが、HRGには未知の受容体があるものと考え、全てのCLEC family、IgG family、他複数回幕貫通型受容体群もクローニングしそれら発現vectorを追加作成中にあり、HRGの新たな受容体となりうるか独自のassay系にて検討中である。HRGのがん転移抑制機能は多岐に渡ると予想されることから、がん細胞のみならず好中球をはじめとする炎症性免疫細胞、血管内皮細胞への作用の未解明な分子機構を解く糸口になるものと考えている。

    researchmap

  • ミトコンドリア呼吸阻害による軸索変性誘導機構の解明とパーキンソン病への展開

    2019年04月 - 2022年03月

    日本学術振興会  基盤研究C 

    村田 等

      詳細を見る

    担当区分:研究代表者  資金種別:競争的資金

    researchmap

  • 新規S100受容体による原発巣転移前がん微小環境構築とがん転移動力獲得の分子機構

    研究課題/領域番号:17H03577  2017年04月 - 2020年03月

    日本学術振興会  科学研究費助成事業 基盤研究(B)  基盤研究(B)

    阪口 政清, 山本 健一, 冨田 秀太, 豊岡 伸一, 木下 理恵, 村田 等, 枝園 和彦

      詳細を見る

    配分額:17810000円 ( 直接経費:13700000円 、 間接経費:4110000円 )

    臓器指向性転移を誘導する新規S100A8/A9受容体群の作動原理を解明することを目指した。SSSRsの内、MCAMとNPTNβに関する転移動力供給のシグナル伝達メカニズムが不明であったが、研究からMCAMとNPTNβについて転移動力を引き出す各々の下流信号伝達を解明するに至った。解析の結果、メラノーマと乳がんでは、MCAM→TPL2(MAPKKK)→ETV4(転写因子)が、肺がんでは、NPTNβ→RAS & TRAF2(アダプター因子)→NFIA/NFIB(転写因子)→SPDEF(転写因子)が転移を促す重要信号伝達経路として新規に同定することができた。

    researchmap

  • ミトコンドリア呼吸制御機構の解析と神経変性疾患におけるその破綻

    2016年04月 - 2019年03月

    日本学術振興会  若手研究B 

    村田 等

      詳細を見る

    担当区分:研究代表者  資金種別:競争的資金

    researchmap

  • がんの転移先臓器親和性を制御する新規生物製剤の開発

    研究課題/領域番号:15K14382  2015年04月 - 2017年03月

    日本学術振興会  科学研究費助成事業 挑戦的萌芽研究  挑戦的萌芽研究

    阪口 政清, 豊岡 伸一, 村田 等

      詳細を見る

    配分額:3770000円 ( 直接経費:2900000円 、 間接経費:870000円 )

    S100A8/A9を吸着し、がん細胞に作用させないデコイタンパク質製剤{受容体細胞外領域(exReceptors)とIgGのFc領域との融合キメラタンパク質}( exEMMPRIN-Fc, exNPTNβ-Fc, exMCAM-Fc)のCHO細胞による大量調製系を確立した。これらデコイタンパク質の効能を動物転移モデルで検討したところ、メラノーマの肺転移性の顕著な減少が認められた。

    researchmap

  • 転移先臓器を感知する受容体群の分子制御機構の解明とその応用

    研究課題/領域番号:26290039  2014年04月 - 2017年03月

    日本学術振興会  科学研究費助成事業 基盤研究(B)  基盤研究(B)

    阪口 政清, 渡部 昌実, 村田 等

      詳細を見る

    配分額:16250000円 ( 直接経費:12500000円 、 間接経費:3750000円 )

    本計画では、同定できた新規S100A8/A9受容体群EMMPRIN, NPTNα, NPTNβ, MCAM, ALCAMについて、メラノーマの肺転移における重要性について検討した。結果、EMMPRIN, NPTNβ, MCAMが、肺から分泌されるS100A8/A9に応答して、メラノーマの肺転移を強く誘導する事が判明した。EMMPRIN, NPTNβ, MCAMが治療の有効な分子標的となる可能性が示されたのだ。

    researchmap

  • PINK1キナーゼを中心としたミトコンドリア恒常性維持機構の解析

    2013年04月 - 2016年03月

    日本学術振興会  若手研究B 

    村田 等

      詳細を見る

    担当区分:研究代表者  資金種別:競争的資金

    researchmap

  • がん幹細胞をも標的とするREIC遺伝子治療の消化器がんへの応用

    研究課題/領域番号:24591943  2012年04月 - 2015年03月

    日本学術振興会  科学研究費助成事業 基盤研究(C)  基盤研究(C)

    片岡 健, 阪口 政清, 村田 等

      詳細を見る

    配分額:5330000円 ( 直接経費:4100000円 、 間接経費:1230000円 )

    マウス組織を用いてREIC/Dkk-3の発現局在の詳細なスクリーニングを行ったところ、皮膚と腸上皮組織において幹細胞のニッチとREIC/Dkk-3の局在が一致していた。また腸管上皮細胞株Caco-2を細胞外マトリクスタンパク質に包埋して3次元培養を行ったスフェロイドにおいてもREIC/Dkk-3の発現を認めた。またREIC/Dkk-3の発現制御因子を探索したところ、TNF-αが表皮ケラチノサイトのREIC/Dkk-3発現を低下させることがわかった。REIC/Dkk-3とTNF-αのバランスが発生過程や炎症における表皮ケラチノサイトと血管内皮細胞による組織構築を調整していることが示唆された。

    researchmap

  • 新世代REICアデノウィルスによる純国産がん遺伝子治療の統合新戦略

    研究課題/領域番号:23650625  2011年 - 2012年

    日本学術振興会  科学研究費助成事業 挑戦的萌芽研究  挑戦的萌芽研究

    許 南浩, 阪口 政清, 片岡 健, 村田 等

      詳細を見る

    配分額:3640000円 ( 直接経費:2800000円 、 間接経費:840000円 )

    抗がんウイルス製剤 REIC アデノウイルスに、独自に開発した「がん特異的新規プロモーターシステム」と「新規アデノウイルスアダプター」を適用することで、REIC アデノウィルスを高機能化させることに成功した。これは、従来に比較して遥かに強力ながん選択的なアデノウイルス感染とREIC 遺伝子発現を可能にし、抗腫瘍効果を大幅に上昇させた。

    researchmap

  • がん幹細胞特異的殺傷抗体作成技術の創出

    研究課題/領域番号:23650608  2011年 - 2012年

    日本学術振興会  科学研究費助成事業 挑戦的萌芽研究  挑戦的萌芽研究

    阪口 政清, 片岡 健, 村田 等, 許 南浩

      詳細を見る

    配分額:3640000円 ( 直接経費:2800000円 、 間接経費:840000円 )

    がん幹細胞クローニング用プラスミドベクターを完成させたこと、このベクターを使用して、がん幹細胞の特性を備えたクローン化がん幹細胞株を樹立できたこと、があげられる。クローン化の高効率化を目指し、独自のプラスミドベクターを完成させるまでに多大な時間を費やした。現在、膜タンパク質を調製し、免疫のステップに移行している。

    researchmap

▼全件表示

 

担当授業科目

  • 生化学 (2023年度) 集中  - その他

  • 細胞生物学実習 (2023年度) 特別  - その他

  • 細胞生物学演習 (2023年度) 特別  - その他

  • 細胞生物学I(演習・実習) (2023年度) 特別  - その他

  • 細胞生物学I(講義・演習) (2023年度) 特別  - その他

  • 細胞生物学II(演習・実習) (2023年度) 特別  - その他

  • 細胞生物学II(講義・演習) (2023年度) 特別  - その他

  • 生化学 (2022年度) 集中  - その他

  • 細胞生物学I(演習・実習) (2022年度) 特別  - その他

  • 細胞生物学I(講義・演習) (2022年度) 特別  - その他

  • 細胞生物学II(演習・実習) (2022年度) 特別  - その他

  • 細胞生物学II(講義・演習) (2022年度) 特別  - その他

  • 生化学 (2021年度) 集中  - その他

  • 細胞生物学I(演習・実習) (2021年度) 特別  - その他

  • 細胞生物学I(講義・演習) (2021年度) 特別  - その他

  • 細胞生物学II(演習・実習) (2021年度) 特別  - その他

  • 細胞生物学II(講義・演習) (2021年度) 特別  - その他

  • 生化学 (2020年度) 集中  - その他

  • 細胞生物学I(演習・実習) (2020年度) 特別  - その他

  • 細胞生物学I(講義・演習) (2020年度) 特別  - その他

  • 細胞生物学II(演習・実習) (2020年度) 特別  - その他

  • 細胞生物学II(講義・演習) (2020年度) 特別  - その他

▼全件表示

 

社会貢献活動

  • パーキンソン病の病気のメカニズムと予防・治療について

    役割:講師, 情報提供

    2021年3月23日