基本情報

写真a

仁科 博史(ニシナ ヒロシ)

NISHINA Hiroshi


職名

教授

研究室住所

〒113-8510 東京都文京区湯島1-5-45 M&Dタワー21階

研究室電話番号

03-5803-4659

研究室FAX番号

03-5803-5829

メールアドレス

nishina.dbio@mri.tmd.ac.jp

ホームページ

http://www.tmd.ac.jp/mri/dbio/index.html#

研究分野・キーワード

ストレス, シグナル伝達, JNK, Hippo, 肝臓, 発生, 再生, モデル生物, メダカ

出身学校 【 表示 / 非表示

  • 東京大学  理学部  生物化学  1985年  卒業

出身大学院 【 表示 / 非表示

  • 東京大学  理学研究科  理学研究科  博士課程  1990年  修了

取得学位 【 表示 / 非表示

  • 理学博士  東京大学

経歴(学内) 【 表示 / 非表示

  • 2005年01月
    -
    2022年04月
    東京医科歯科大学 難治疾患研究所 難治病態研究部門 発生再生生物学分野 教授
  • 2005年01月
    -
    現在
    東京医科歯科大学 大学院医歯学総合研究科 生命理工学系専攻 生命理工学講座 発生再生生物学 教授
  • 2020年04月
    -
    現在
    東京医科歯科大学 難治疾患研究所 研究所長
  • 2022年05月
    -
    現在
    東京医科歯科大学 難治疾患研究所 未来生命科学研究部門 発生再生生物学分野 教授

経歴(学外) 【 表示 / 非表示

  • 1989年04月
    -
    1990年03月
    東京大学 日本学術振興会特別研究員
  • 1990年04月
    -
    1997年02月
    東京工業大学生命理工学部生命理学科 助手 生命理工学部 助手
  • 1995年04月
    -
    1997年02月
    カナダ国トロント大学/オンタリオ癌研究所 博士研究員
  • 1997年03月
    -
    1998年04月
    東京大学 薬学部 助手
  • 1998年05月
    -
    2004年12月
    東京大学 薬学部 助教授
  • 2005年01月
    -
    現在
    東京医科歯科大学 難治疾患研究所 教授

▼全件表示

所属学協会 【 表示 / 非表示

  • 日本生化学会、日本分子生物学会、日本薬学会、肝細胞研究会、日本肝臓学会、日本癌学会、日本再生医療学会、日本Cell Death学会

研究分野 【 表示 / 非表示

  • 薬系衛生、生物化学

資格、免許 【 表示 / 非表示

  • 放射線取扱主任者(第1~2種)

 

競争的資金等の研究課題 【 表示 / 非表示

  • フードテックを応用した細胞培養食品の先駆的な調査検討による食品衛生上のハザードやリスクに係る研究-リスクプロファイルの作成とモデル細胞実験系による検証・還元-

    厚生労働省 : 2022年04月 - 2025年03月

  • ドライバー遺伝子の痕跡が消失するがん発症機構の解明

    文部科学省/日本学術振興会 : 2018年 - 2019年

  • 細胞の張力を制御するHippo-YAPシグナル伝達系の肝形成における役割の解明

    文部科学省/日本学術振興会 : 2017年 - 2019年

  • 細胞外の多様な環境硬度に応じた細胞分化を制御するHippo-YAPシグナルの解析

    文部科学省/日本学術振興会 : 2017年 - 2018年

  • 肝臓組織構築制御における細胞品質管理機構の解明

    文部科学省/日本学術振興会 : 2014年 - 2016年

  • エクソソームを介したHBV感染及び発がんメカニズム解明と治療戦略

    厚生労働省 : 2013年04月 - 2016年03月

  • 器官サイズ制御因子YAP標的転写産物の網羅的発現解析

    文部科学省/日本学術振興会 : 2013年

  • 肝硬変に対する細胞治療法の臨床的確立とそのメカニズムの解明

    厚生労働省 : 2012年04月 - 2015年03月

  • 器官サイズ制御因子YAP依存的肝癌誘発系の開発とマイクロRNAの網羅的発現解析

    文部科学省/日本学術振興会 : 2012年

  • マウスやメダカを用いた肝発生・再生および肝病態シグナルネットワークの解明

    文部科学省/日本学術振興会 : 2011年 - 2013年

  • MAPキナーゼ・Hippoシグナル系による細胞運命決定制御の解明

    文部科学省/日本学術振興会 : 2011年 - 2012年

  • Hippoシグナル経路による細胞競合機構とその破綻病態

    文部科学省/日本学術振興会

  • 細胞張力を制御するHippo-YAPシグナル経路の肝発生・再生における役割の解明

    文部科学省/日本学術振興会

  • 臓器・細胞選択的in vivoゲノム編集による難治癌の遺伝子治療法開発

    文部科学省/日本学術振興会

▼全件表示

論文・総説 【 表示 / 非表示

  1. Hirotoshi Soyama, Miki Nishio, Junji Otani, Toshiko Sakuma, Shintaro Takao, Shigeo Hara, Takaaki Masuda, Koshi Mimori, Shinya Toyokuni, John P Lydon, Kazuwa Nakao, Hiroshi Nishina, Takumi Fukumoto, Tomohiko Maehama, Akira Suzuki. Hippo-TAZ signaling is the master regulator of the onset of triple-negative basal-like breast cancers. Proc Natl Acad Sci U S A. 2022.07; 119 (29): e2123134119. ( PubMed, DOI )

  2. Joshua Agbemefa Kuleape, Shakhawoat Hossain, Caleb Kwame Sinclear, Takanobu Shimizu, Hiroaki Iwasa, Junichi Maruyama, Kyoko Arimoto-Matsuzaki, Hiroshi Nishina, Yutaka Hata. DNA Damage Triggers the Nuclear Accumulation of RASSF6 Tumor Suppressor Protein via CDK9 and BAF53 To Regulate p53 Target Gene Transcription. Mol Cell Biol. 2022.02; 42 (2): e0031021. ( PubMed, DOI )

  3. Mayu Morishita, Kyoko Arimoto-Matsuzaki, Masami Kitamura, Kyohei Niimura, Hiroaki Iwasa, Junichi Maruyama, Yuichi Hiraoka, Kohei Yamamoto, Masanobu Kitagawa, Norio Miyamura, Hiroshi Nishina, Yutaka Hata. Characterization of mouse embryonic fibroblasts derived from Rassf6 knockout mice shows the implication of Rassf6 in the regulation of NF-κB signaling. Genes Cells. 2021.12; 26 (12): 999-1013. ( PubMed, DOI )

  4. Keisuke Nakatani, Tomohiko Maehama, Miki Nishio, Junji Otani, Keiko Yamaguchi, Miki Fukumoto, Hiroki Hikasa, Shinji Hagiwara, Hiroshi Nishina, Tak Wah Mak, Teruki Honma, Yasumitsu Kondoh, Hiroyuki Osada, Minoru Yoshida, Akira Suzuki. Alantolactone is a natural product that potently inhibits YAP1/TAZ through promotion of reactive oxygen species accumulation. Cancer Sci. 2021.10; 112 (10): 4303-4316. ( PubMed, DOI )

  5. Sachi Sunaga, Satoshi Kofuji, Hiroshi Nishina. YAP drives cell competition by activating choline metabolism. Biochem Biophys Res Commun. 2021.10; 572 178-184. ( PubMed, DOI )

  6. Shunta Nagashima, Junichi Maruyama, Kaori Honda, Yasumitsu Kondoh, Hiroyuki Osada, Makiko Nawa, Ken-Ichi Nakahama, Mari Ishigami-Yuasa, Hiroyuki Kagechika, Haruhiko Sugimura, Hiroaki Iwasa, Kyoko Arimoto-Matsuzaki, Hiroshi Nishina, Yutaka Hata. CSE1L promotes nuclear accumulation of transcriptional coactivator TAZ and enhances invasiveness of human cancer cells. J Biol Chem. 2021.07; 297 (1): 100803. ( PubMed, DOI )

  7. Takako Ooshio, Masahiro Yamamoto, Kiyonaga Fujii, Bing Xin, Kenji Watanabe, Masanori Goto, Yoko Okada, Akira Suzuki, Josef M Penninger, Hiroshi Nishina, Yuji Nishikawa. Hepatocyte Mitogen-Activated Protein Kinase Kinase 7 Contributes to Restoration of the Liver Parenchyma Following Injury in Mice. Hepatology. 2021.06; 73 (6): 2510-2526. ( PubMed, DOI )

  8. Xinliang Jiang, Junichi Maruyama, Hiroaki Iwasa, Kyoko Arimoto-Matsuzaki, Hiroshi Nishina, Yutaka Hata. Heat shock induces the nuclear accumulation of YAP1 via SRC. Exp Cell Res. 2021.02; 399 (1): 112439. ( PubMed, DOI )

  9. Tadashi Shin, Yuichi Hiraoka, Tokiwa Yamasaki, Jamey D Marth, Josef M Penninger, Masami Kanai-Azuma, Kohichi Tanaka, Satoshi Kofuji, Hiroshi Nishina. MKK7 deficiency in mature neurons impairs parental behavior in mice. Genes Cells. 2021.01; 26 (1): 5-17. ( PubMed, DOI )

  10. Yikelamu Alifu, Satoshi Kofuji, Sachi Sunaga, Mizuki Kusaba, Jun Hirayama, Hiroshi Nishina. The Light-Inducible Genes Per2, Cry1a, and Cry2a Regulate Oxidative Status in Zebrafish. Biol Pharm Bull. 2021; 44 (8): 1160-1165. ( PubMed, DOI )

  11. Erika Ishihara, Yuya Nagaoka, Toshiaki Okuno, Satoshi Kofuji, Mari Ishigami-Yuasa, Hiroyuki Kagechika, Kenya Kamimura, Shuji Terai, Takehiko Yokomizo, Yukihiko Sugimoto, Yasuyuki Fujita, Akira Suzuki, Hiroshi Nishina. Prostaglandin E2 and its receptor EP2 trigger signaling that contributes to YAP-mediated cell competition. Genes Cells. 2020.03; 25 (3): 197-214. ( PubMed, DOI )

  12. Caleb Kwame Sinclear, Junichi Maruyama, Shunta Nagashima, Kyoko Arimoto-Matsuzaki, Joshua Agbemefa Kuleape, Hiroaki Iwasa, Hiroshi Nishina, Yutaka Hata. Protein kinase Cα activation switches YAP1 from TEAD-mediated signaling to p73-mediated signaling. Cancer Sci. 2022.04; 113 (4): 1305-1320. ( PubMed, DOI )

  13. Hirofumi Omori, Miki Nishio, Muneyuki Masuda, Yosuke Miyachi, Fumihito Ueda, Takafumi Nakano, Kuniaki Sato, Koshi Mimori, Kenichi Taguchi, Hiroki Hikasa, Hiroshi Nishina, Hironori Tashiro, Tohru Kiyono, Tak Wah Mak, Kazuwa Nakao, Takashi Nakagawa, Tomohiko Maehama, Akira Suzuki. YAP1 is a potent driver of the onset and progression of oral squamous cell carcinoma. Sci Adv. 2020; 6 (12): eaay3324. ( PubMed, DOI )

  14. Takanobu Shimizu, Takeshi Nakamura, Hironori Inaba, Hiroaki Iwasa, Junichi Maruyama, Kyoko Arimoto-Matsuzaki, Takao Nakata, Hiroshi Nishina, Yutaka Hata. The RAS-interacting chaperone UNC119 drives the RASSF6-MDM2-p53 axis and antagonizes RAS-mediated malignant transformation. J. Biol. Chem.. 2020.08; 295 (32): 11214-11230. ( PubMed, DOI )

  15. Miki Nishio, Yoko To, Tomohiko Maehama, Yukari Aono, Junji Otani, Hiroki Hikasa, Akihiro Kitagawa, Koshi Mimori, Takehiko Sasaki, Hiroshi Nishina, Shinya Toyokuni, John P Lydon, Kazuwa Nakao, Tak Wah Mak, Tohru Kiyono, Hidetaka Katabuchi, Hironori Tashiro, Akira Suzuki. Endogenous YAP1 activation drives immediate onset of cervical carcinoma in situ in mice. Cancer Sci.. 2020.07; 111 (10): 3576-3587. ( PubMed, DOI )

  16. Kenya Kamimura, Takeshi Yokoo, Hiroyuki Abe, Norihiro Sakai, Takuro Nagoya, Yuji Kobayashi, Masato Ohtsuka, Hiromi Miura, Akira Sakamaki, Hiroteru Kamimura, Norio Miyamura, Hiroshi Nishina, Shuji Terai. Effect of Diphtheria Toxin-Based Gene Therapy for Hepatocellular Carcinoma. Cancers (Basel). 2020.02; 12 (2): ( PubMed, DOI )

  17. Nozomi Hanzawa, Koshi Hashimoto, Xunmei Yuan, Kenichi Kawahori, Kazutaka Tsujimoto, Miho Hamaguchi, Toshiya Tanaka, Yuya Nagaoka, Hiroshi Nishina, Sumiyo Morita, Izuho Hatada, Tetsuya Yamada, Yoshihiro Ogawa. Targeted DNA demethylation of the Fgf21 promoter by CRISPR/dCas9-mediated epigenome editing. Sci Rep. 2020; 10 (1): 5181. ( PubMed, DOI )

  18. Manami Kodaka, Fengju Mao, Kyoko Arimoto-Matsuzaki, Masami Kitamura, Xiaoyin Xu, Zeyu Yang, Kentaro Nakagawa, Junichi Maruyama, Kana Ishii, Chihiro Akazawa, Takuya Oyaizu, Naoki Yamamoto, Mari Ishigami-Yuasa, Nozomi Tsuemoto, Shigeru Ito, Hiroyuki Kagechika, Hiroshi Nishina, Yutaka Hata. Characterization of a novel compound that promotes myogenesis via Akt and transcriptional co-activator with PDZ-binding motif (TAZ) in mouse C2C12 cells. PLoS ONE. 2020; 15 (4): e0231265. ( PubMed, DOI )

  19. Takuro Nagoya, Kenya Kamimura, Ryo Goto, Yoko Shinagawa-Kobayashi, Yusuke Niwa, Atsushi Kimura, Norihiro Sakai, Masayoshi Ko, Hiroshi Nishina, Shuji Terai. Inhibition of sodium-glucose cotransporter 2 ameliorates renal injury in a novel medaka model of nonalcoholic steatohepatitis-related kidney disease. FEBS Open Bio. 2019.12; 9 (12): 2016-2024. ( PubMed, DOI )

  20. Jun Hirayama, Yikelamu Alifu, Rin Hamabe, Sho Yamaguchi, Jun Tomita, Yusuke Maruyama, Yoichi Asaoka, Ken-Ichi Nakahama, Teruya Tamaru, Ken Takamatsu, Nobuhiko Takamatsu, Atsuhiko Hattori, Sachiko Nishina, Noriyuki Azuma, Atsuo Kawahara, Kazuhiko Kume, Hiroshi Nishina. The clock components Period2, Cryptochrome1a, and Cryptochrome2a function in establishing light-dependent behavioral rhythms and/or total activity levels in zebrafish. Sci Rep. 2019.01; 9 (1): 196. ( PubMed, DOI )

  21. Miki Nishio, Yousuke Miyachi, Junji Otani, Shoji Tane, Hirofumi Omori, Fumihito Ueda, Hideru Togashi, Takehiko Sasaki, Tak Wah Mak, Kazuwa Nakao, Yasuyuki Fujita, Hiroshi Nishina, Tomohiko Maehama, Akira Suzuki. Hippo pathway controls cell adhesion and context-dependent cell competition to influence skin engraftment efficiency. FASEB J.. 2019.01; 33 (4): fj201802005R. ( PubMed, DOI )

  22. Ryo Goto, Kenya Kamimura, Yoko Shinagawa-Kobayashi, Norihiro Sakai, Takuro Nagoya, Yusuke Niwa, Masayoshi Ko, Kohei Ogawa, Ryosuke Inoue, Takeshi Yokoo, Akira Sakamaki, Hiroteru Kamimura, Satoshi Abe, Hiroshi Nishina, Shuji Terai. Inhibition of sodium glucose cotransporter 2 (SGLT2) delays liver fibrosis in a medaka model of nonalcoholic steatohepatitis (NASH). FEBS Open Bio. 2019.12; 9 (4): 643-652. ( PubMed, DOI )

  23. Asami Kawasaki, Masayasu Okada, Atsushi Tamada, Shujiro Okuda, Motohiro Nozumi, Yasuyuki Ito, Daiki Kobayashi, Tokiwa Yamasaki, Ryo Yokoyama, Takeshi Shibata, Hiroshi Nishina, Yutaka Yoshida, Yukihiko Fujii, Kosei Takeuchi, Michihiro Igarashi. Growth Cone Phosphoproteomics Reveals that GAP-43 Phosphorylated by JNK Is a Marker of Axon Growth and Regeneration. iScience. 2018.06; 4 190-203. ( PubMed, DOI )

  24. K Otsubo, H Goto, M Nishio, K Kawamura, S Yanagi, W Nishie, T Sasaki, T Maehama, H Nishina, K Mimori, T Nakano, H Shimizu, T W Mak, K Nakao, Y Nakanishi, A Suzuki. MOB1-YAP1/TAZ-NKX2.1 axis controls bronchioalveolar cell differentiation, adhesion and tumour formation. Oncogene. 2017.07; 36 (29): 4201-4211. ( PubMed, DOI )

  25. Norio Miyamura, Shoji Hata, Tohru Itoh, Minoru Tanaka, Miki Nishio, Michiko Itoh, Yoshihiro Ogawa, Shuji Terai, Isao Sakaida, Akira Suzuki, Atsushi Miyajima, Hiroshi Nishina. YAP determines the cell fate of injured mouse hepatocytes in vivo. Nat Commun. 2017.07; 8 16017. ( PubMed, DOI )

  26. Erika Ishihara, Hiroshi Nishina. The Hippo-YAP Pathway Regulates 3D Organ Formation and Homeostasis. Cancers (Basel). 2018.04; 10 (4): ( PubMed, DOI )

  27. Tokiwa Yamasaki, Norie Deki-Arima, Asahito Kaneko, Norio Miyamura, Mamiko Iwatsuki, Masato Matsuoka, Noriko Fujimori-Tonou, Yoshimi Okamoto-Uchida, Jun Hirayama, Jamey D Marth, Yuji Yamanashi, Hiroshi Kawasaki, Koji Yamanaka, Josef M Penninger, Shigenobu Shibata, Hiroshi Nishina. Age-dependent motor dysfunction due to neuron-specific disruption of stress-activated protein kinase MKK7. Sci Rep. 2017.08; 7 (1): 7348. ( PubMed, DOI )

  28. Seiichiro Mori, Takamasa Takeuchi, Yoshiyuki Ishii, Takashi Yugawa, Tohru Kiyono, Hiroshi Nishina, Iwao Kukimoto. Human Papillomavirus 16 E6 Upregulates APOBEC3B via the TEAD Transcription Factor. J. Virol.. 2017.03; 91 (6): ( PubMed, DOI )

  29. Tatsuyuki Matsudaira, Kojiro Mukai, Taishin Noguchi, Junya Hasegawa, Tomohisa Hatta, Shun-Ichiro Iemura, Tohru Natsume, Norio Miyamura, Hiroshi Nishina, Jun Nakayama, Kentaro Semba, Takuya Tomita, Shigeo Murata, Hiroyuki Arai, Tomohiko Taguchi. Endosomal phosphatidylserine is critical for the YAP signalling pathway in proliferating cells. Nat Commun. 2017.11; 8 (1): 1246. ( PubMed, DOI )

  30. Jun Negishi, Yuka Omori, Mami Shindo, Hayate Takanashi, Shiori Musha, Suminori Nagayama, Jun Hirayama, Hiroshi Nishina, Takashi Nakakura, Chihiro Mogi, Koichi Sato, Fumikazu Okajima, Yuta Mochimaru, Hideaki Tomura. Manganese and cobalt activate zebrafish ovarian cancer G-protein-coupled receptor 1 but not GPR4. J. Recept. Signal Transduct. Res.. 2017.08; 37 (4): 401-408. ( PubMed, DOI )

  31. Yoichi Asaoka, Hiroshi Nishina, Makoto Furutani-Seiki. YAP is essential for 3D organogenesis withstanding gravity. Dev. Growth Differ.. 2017.01; 59 (1): 52-58. ( PubMed, DOI )

  32. Yoshimi Okamoto-Uchida, Ruoxing Yu, Norio Miyamura, Norie Arima, Mari Ishigami-Yuasa, Hiroyuki Kagechika, Suguru Yoshida, Takamitsu Hosoya, Makiko Nawa, Takeshi Kasama, Yoichi Asaoka, Reiner Wimmer Alois, Ulrich Elling, Josef M Penninger, Sachiko Nishina, Noriyuki Azuma, Hiroshi Nishina. The mevalonate pathway regulates primitive streak formation via protein farnesylation. Sci Rep. 2016.11; 6 37697. ( PubMed, DOI )

  33. Takanori Chiba, Erika Ishihara, Norio Miyamura, Rika Narumi, Mihoko Kajita, Yasuyuki Fujita, Akira Suzuki, Yoshihiro Ogawa, Hiroshi Nishina. MDCK cells expressing constitutively active Yes-associated protein (YAP) undergo apical extrusion depending on neighboring cell status. Sci Rep. 2016.06; 6 28383. ( PubMed, DOI )

  34. Yoichi Asaoka, Yoko Nagai, Misako Namae, Makoto Furutani-Seiki, Hiroshi Nishina. SLC7 family transporters control the establishment of left-right asymmetry during organogenesis in medaka by activating mTOR signaling. Biochem. Biophys. Res. Commun.. 2016.05; 474 (1): 146-153. ( PubMed, DOI )

  35. Shunta Nagashima, Junichi Maruyama, Shodai Kawano, Hiroaki Iwasa, Kentaro Nakagawa, Mari Ishigami-Yuasa, Hiroyuki Kagechika, Hiroshi Nishina, Yutaka Hata. Validation of chemical compound library screening for transcriptional co-activator with PDZ-binding motif inhibitors using GFP-fused transcriptional co-activator with PDZ-binding motif. Cancer Sci.. 2016.06; 107 (6): 791-802. ( PubMed, DOI )

  36. Koichi Fujisawa, Shuji Terai, Taro Takami, Naoki Yamamoto, Takahiro Yamasaki, Toshihiko Matsumoto, Kazuhito Yamaguchi, Yuji Owada, Hiroshi Nishina, Takafumi Noma, Isao Sakaida. Modulation of anti-cancer drug sensitivity through the regulation of mitochondrial activity by adenylate kinase 4. J. Exp. Clin. Cancer Res.. 2016.03; 35 48. ( PubMed, DOI )

  37. Yusuke Nasu, Yoichi Asaoka, Misako Namae, Hiroshi Nishina, Hideaki Yoshimura, Takeaki Ozawa. Genetically Encoded Fluorescent Probe for Imaging Apoptosis in Vivo with Spontaneous GFP Complementation. Anal. Chem.. 2016.01; 88 (1): 838-844. ( PubMed, DOI )

  38. Miki Nishio, Keishi Sugimachi, Hiroki Goto, Jia Wang, Takumi Morikawa, Yosuke Miyachi, Yusuke Takano, Hiroki Hikasa, Tohru Itoh, Satoshi O Suzuki, Hiroki Kurihara, Shinichi Aishima, Andrew Leask, Takehiko Sasaki, Toru Nakano, Hiroshi Nishina, Yuji Nishikawa, Yoshitaka Sekido, Kazuwa Nakao, Kazuo Shin-Ya, Koshi Mimori, Akira Suzuki. Dysregulated YAP1/TAZ and TGF-β signaling mediate hepatocarcinogenesis in Mob1a/1b-deficient mice. Proc. Natl. Acad. Sci. U.S.A.. 2016.01; 113 (1): E71-E80. ( PubMed, DOI )

  39. Sean Porazinski, Huijia Wang, Yoichi Asaoka, Martin Behrndt, Tatsuo Miyamoto, Hitoshi Morita, Shoji Hata, Takashi Sasaki, S.F. Gabby Krens, Yumi Osada, Satoshi Asaka, Akihiro Momoi, Sarah Linton, Joel B. Miesfeld, Brian A. Link, Takeshi Senga, Atahualpa Castillo-Morales, Araxi O. Urrutia, Nobuyoshi Shimizu, Hideaki Nagase, Shinya Matsuura, Stefan Bagby, Hisato Kondoh, Hiroshi Nishina, Carl-Philipp Heisenberg and Makoto Furutani-Seiki. YAP is essential for tissue tension to ensure vertebrate 3D body shape Nature. 2015.04;

  40. Ruoxing Yu, Norio Miyamura, Yoshimi Okamoto-Uchida, Norie Arima, Mari Ishigami-Yuasa, Hiroyuki Kagechika, Hiroshi Nishina. A Modified Murine Embryonic Stem Cell Test for Evaluating the Teratogenic Effects of Drugs on Early Embryogenesis. PLoS ONE. 2015.12; 10 (12): e0145286. ( PubMed, DOI )

  41. Koichi Fujisawa, Shuji Terai, Toshihiko Matsumoto, Taro Takami, Naoki Yamamoto, Hiroshi Nishina, Makoto Furutani-Seiki, Isao Sakaida. Evidence for a Role of the Transcriptional Regulator Maid in Tumorigenesis and Aging. PLoS ONE. 2015.02; 10 (6): e0129950. ( PubMed, DOI )

  42. Shodai Kawano, Junichi Maruyama, Shunta Nagashima, Kazutoshi Inami, Wenzhe Qiu, Hiroaki Iwasa, Kentaro Nakagawa, Mari Ishigami-Yuasa, Hiroyuki Kagechika, Hiroshi Nishina, Yutaka Hata. A cell-based screening for TAZ activators identifies ethacridine, a widely used antiseptic and abortifacient, as a compound that promotes dephosphorylation of TAZ and inhibits adipogenesis in C3H10T1/2 cells. J. Biochem.. 2015.11; 158 (5): 413-423. ( PubMed, DOI )

  43. Yuta Mochimaru, Morio Azuma, Natsuki Oshima, Yuta Ichijo, Kazuhiro Satou, Kouhei Matsuda, Yoichi Asaoka, Hiroshi Nishina, Takashi Nakakura, Chihiro Mogi, Koichi Sato, Fumikazu Okajima, Hideaki Tomura. Extracellular acidification activates ovarian cancer G-protein-coupled receptor 1 and GPR4 homologs of zebra fish. Biochem. Biophys. Res. Commun.. 2015.02; 457 (4): 493-499. ( PubMed, DOI )

  44. Hajime Tajima Sakurai, Hidefumi Iwashita, Satoko Arakawa, Alifu Yikelamu, Mizuki Kusaba, Satoshi Kofuji, Hiroshi Nishina, Munetaka Ishiyama, Yuichiro Ueno, Shigeomi Shimizu. Development of small fluorescent probes for the analysis of autophagy kinetics. iScience. 2023.07; 26 (7): 107218. ( PubMed, DOI )

  45. Noriyuki Azuma, Tadashi Yokoi, Taku Tanaka, Emiko Matsuzaka, Yuki Saida, Sachiko Nishina, Miho Terao, Shuji Takada, Maki Fukami, Kohji Okamura, Kayoko Maehara, Tokiwa Yamasaki, Jun Hirayama, Hiroshi Nishina, Hiroshi Handa, Yuki Yamaguchi. Integrator complex subunit 15 controls mRNA splicing and is critical for eye development. Hum Mol Genet. 2023.06; 32 (12): 2032-2045. ( PubMed, DOI )

  46. Chiyumi Oda, Kenya Kamimura, Osamu Shibata, Shinichi Morita, Yuto Tanaka, Toru Setsu, Hiroyuki Abe, Takeshi Yokoo, Akira Sakamaki, Hiroteru Kamimura, Satoshi Kofuji, Toshifumi Wakai, Hiroshi Nishina, Shuji Terai. HBx and YAP expression could promote tumor development and progression in HBV-related hepatocellular carcinoma. Biochem Biophys Rep. 2022.12; 32 101352. ( PubMed, DOI )

  47. Sakurako Kobayashi, Nobuhiko Ogasawara, Satoshi Watanabe, Yosuke Yoneyama, Sakura Kirino, Yui Hiraguri, Masami Inoue, Sayaka Nagata, Yoshimi Okamoto-Uchida, Satoshi Kofuji, Hiromichi Shimizu, Go Ito, Tomohiro Mizutani, Shinichi Yamauchi, Yusuke Kinugasa, Yoshihito Kano, Yasuhiro Nemoto, Mamoru Watanabe, Kiichiro Tsuchiya, Hiroshi Nishina, Ryuichi Okamoto, Shiro Yui. Collagen type I-mediated mechanotransduction controls epithelial cell fate conversion during intestinal inflammation. Inflamm Regen. 2022.11; 42 (1): 49. ( PubMed, DOI )

  48. Hiroshi Nishina. Physiological and pathological roles of the Hippo-YAP/TAZ signaling pathway in liver formation, homeostasis, and tumorigenesis. Cancer Sci. 2022.06; 113 (6): 1900-1908. ( PubMed, DOI )

  49. Takeaki Shibata, Hiroki Kawana, Yuri Nishino, Yoshiko Ito, Hiroyasu Sato, Hirofumi Onishi, Kuniyuki Kano, Asuka Inoue, Yoshitaka Taketomi, Makoto Murakami, Satoshi Kofuji, Hiroshi Nishina, Atsuo Miyazawa, Nozomu Kono, Junken Aoki. Abnormal male reproduction and embryonic development induced by downregulation of a phospholipid fatty acid-introducing enzyme Lpgat1 in zebrafish. Sci Rep. 2022.05; 12 (1): 7312. ( PubMed, DOI )

  50. Sinclear Caleb Kwame, Maruyama Junichi, Nagashima Shunta, Arimoto-Matsuzaki Kyoko, Kuleape Joshua Agbemefa, Iwasa Hiroaki, Nishina Hiroshi, Hata Yutaka. Protein kinase Cα activation switches YAP1 from TEAD-mediated signaling to p73-mediated signaling(和訳中) Cancer Science. 2022.04; 113 (4): 1305-1320. ( 医中誌 )

  51. Shoko Miyata, Noriaki Saku, Saeko Akiyama, Palaksha Kanive Javaregowda, Kenta Ite, Nagisa Takashima, Masashi Toyoda, Kei Yura, Tohru Kimura, Hiroshi Nishina, Atsuko Nakazawa, Mureo Kasahara, Hidenori Nonaka, Tohru Kiyono, Akihiro Umezawa. Puromycin-based purification of cells with high expression of the cytochrome P450 CYP3A4 gene from a patient with drug-induced liver injury (DILI). Stem Cell Res Ther. 2022.01; 13 (1): 6. ( PubMed, DOI )

  52. Nakatani Keisuke, Maehama Tomohiko, Nishio Miki, Otani Junji, Yamaguchi Keiko, Fukumoto Miki, Hikasa Hiroki, Hagiwara Shinji, Nishina Hiroshi, Mak Tak Wah, Honma Teruki, Kondoh Yasumitsu, Osada Hiroyuki, Yoshida Minoru, Suzuki Akira. アラントラクトンは活性酸素種の蓄積を促進することによりYAP1/TAZを強力に阻害する天然物である(Alantolactone is a natural product that potently inhibits YAP1/TAZ through promotion of reactive oxygen species accumulation) Cancer Science. 2021.10; 112 (10): 4303-4316. ( 医中誌 )

  53. Alifu Yikelamu, Kofuji Satoshi, Sunaga Sachi, Kusaba Mizuki, Hirayama Jun, Nishina Hiroshi. ゼブラフィッシュにおいて光作動性遺伝子Per2、Cry1aおよびCry2aは酸化状態を調節する(The Light-Inducible Genes Per2, Cry1a, and Cry2a Regulate Oxidative Status in Zebrafish) Biological & Pharmaceutical Bulletin. 2021.08; 44 (8): 1160-1165. ( 医中誌 )

  54. Nishio Miki, To Yoko, Maehama Tomohiko, Aono Yukari, Otani Junji, Hikasa Hiroki, Kitagawa Akihiro, Mimori Koshi, Sasaki Takehiko, Nishina Hiroshi, Toyokuni Shinya, Lydon John P., Nakao Kazuwa, Mak Tak Wah, Kiyono Tohru, Katabuchi Hidetaka, Tashiro Hironori, Suzuki Akira. 内因性YAP1の活性化はマウスにおいて子宮頸部上皮内癌の即時発症を惹起させる(Endogenous YAP1 activation drives immediate onset of cervical carcinoma in situ in mice) Cancer Science. 2020.10; 111 (10): 3576-3587. ( 医中誌 )

  55. Hiroki Goto, Miki Nishio, Yoko To, Tatsuya Oishi, Yosuke Miyachi, Tomohiko Maehama, Hiroshi Nishina, Haruhiko Akiyama, Tak Wah Mak, Yuma Makii, Taku Saito, Akihiro Yasoda, Noriyuki Tsumaki, Akira Suzuki. in mice results in chondrodysplasia due to YAP1/TAZ-TEAD-dependent repression of SOX9. Development. 2018; 145 (6): ( PubMed, DOI )

  56. Asaoka Yoichi, Nishina Hiroshi, Furutani-Seiki Makoto. 【発生時のサイズ】YAPは重力に抵抗性の3D器官形成にとって必須である(【Size in development】YAP is essential for 3D organogenesis withstanding gravity) Development, Growth & Differentiation. 2017.01; 59 (1): 52-58. ( 医中誌 )

  57. Shuji Terai, Taro Takami, Naoki Yamamoto, Koichi Fujisawa, Tsuyoshi Ishikawa, Yohei Urata, Haruko Tanimoto, Takuya Iwamoto, Yuko Mizunaga, Takashi Matsuda, Takashi Oono, Miho Marumoto, Guzel Burganova, Luiz Fernando Quintanilha, Isao Hidaka, Yoshio Marumoto, Issei Saeki, Koichi Uchida, Takahiro Yamasaki, Kenji Tani, Yasuho Taura, Yasuhiko Fujii, Hiroshi Nishina, Kiwamu Okita, Isao Sakaida. Status and prospects of liver cirrhosis treatment by using bone marrow-derived cells and mesenchymal cells. Tissue Eng Part B Rev. 2014.06; 20 (3): 206-210. ( PubMed, DOI )

  58. Zeyu Yang, Kentaro Nakagawa, Aradhan Sarkar, Junichi Maruyama, Hiroaki Iwasa, Yijun Bao, Mari Ishigami-Yuasa, Shigeru Ito, Hiroyuki Kagechika, Shoji Hata, Hiroshi Nishina, Shinya Abe, Masanobu Kitagawa, Yutaka Hata. Screening with a novel cell-based assay for TAZ activators identifies a compound that enhances myogenesis in C2C12 cells and facilitates muscle repair in a muscle injury model. Mol. Cell. Biol.. 2014.05; 34 (9): 1607-1621. ( PubMed, DOI )

  59. Keita Nakanaga, Kotaro Hama, Kuniyuki Kano, Takanao Sato, Hiroshi Yukiura, Asuka Inoue, Daisuke Saigusa, Hidetoshi Tokuyama, Yoshihisa Tomioka, Hiroshi Nishina, Atsuo Kawahara, Junken Aoki. Overexpression of autotaxin, a lysophosphatidic acid-producing enzyme, enhances cardia bifida induced by hypo-sphingosine-1-phosphate signaling in zebrafish embryo. J. Biochem.. 2014.04; 155 (4): 235-241. ( PubMed, DOI )

  60. Tadanori Shimomura, Norio Miyamura, Shoji Hata, Ryota Miura, Jun Hirayama, Hiroshi Nishina. The PDZ-binding motif of Yes-associated protein is required for its co-activation of TEAD-mediated CTGF transcription and oncogenic cell transforming activity. Biochem. Biophys. Res. Commun.. 2014.01; 443 (3): 917-923. ( PubMed, DOI )

  61. Yoichi Asaoka, Shoji Hata, Misako Namae, Makoto Furutani-Seiki, Hiroshi Nishina. The Hippo pathway controls a switch between retinal progenitor cell proliferation and photoreceptor cell differentiation in zebrafish. PLoS ONE. 2014; 9 (5): e97365. ( PubMed, DOI )

  62. Menno J Oudhoff, Spencer A Freeman, Amber L Couzens, Frann Antignano, Ekaterina Kuznetsova, Paul H Min, Jeffrey P Northrop, Bernhard Lehnertz, Dalia Barsyte-Lovejoy, Masoud Vedadi, Cheryl H Arrowsmith, Hiroshi Nishina, Michael R Gold, Fabio M V Rossi, Anne-Claude Gingras, Colby Zaph. Control of the hippo pathway by Set7-dependent methylation of Yap. Dev. Cell. 2013.07; 26 (2): 188-194. ( PubMed, DOI )

  63. Yoichi Asaoka, Shuji Terai, Isao Sakaida, Hiroshi Nishina. The expanding role of fish models in understanding non-alcoholic fatty liver disease. Dis Model Mech. 2013.07; 6 (4): 905-914. ( PubMed, DOI )

  64. Norie Arima, Yoshimi Uchida, Ruoxing Yu, Koh Nakayama, Hiroshi Nishina. Acetylcholine receptors regulate gene expression that is essential for primitive streak formation in murine embryoid bodies. Biochem. Biophys. Res. Commun.. 2013.06; 435 (3): 447-453. ( PubMed, DOI )

  65. Miki Nishio, Koichi Hamada, Kohichi Kawahara, Masato Sasaki, Fumihito Noguchi, Shuhei Chiba, Kensaku Mizuno, Satoshi O Suzuki, Youyi Dong, Masaaki Tokuda, Takumi Morikawa, Hiroki Hikasa, Jonathan Eggenschwiler, Norikazu Yabuta, Hiroshi Nojima, Kentaro Nakagawa, Yutaka Hata, Hiroshi Nishina, Koshi Mimori, Masaki Mori, Takehiko Sasaki, Tak W Mak, Toru Nakano, Satoshi Itami, Akira Suzuki. Cancer susceptibility and embryonic lethality in Mob1a/1b double-mutant mice. J. Clin. Invest.. 2012.12; 122 (12): 4505-4518. ( PubMed, DOI )

  66. Hiroshi Nishina. hDlk-1: a cell surface marker common to normal hepatic stem/progenitor cells and carcinomas. J. Biochem.. 2012.08; 152 (2): 121-123. ( PubMed, DOI )

  67. Shoji Hata, Jun Hirayama, Hiroaki Kajiho, Kentaro Nakagawa, Yutaka Hata, Toshiaki Katada, Makoto Furutani-Seiki, Hiroshi Nishina. A novel acetylation cycle of transcription co-activator Yes-associated protein that is downstream of Hippo pathway is triggered in response to SN2 alkylating agents. J. Biol. Chem.. 2012.06; 287 (26): 22089-22098. ( PubMed, DOI )

  68. Ken Okada, Akihide Kamiya, Keiichi Ito, Ayaka Yanagida, Hidenori Ito, Hiroki Kondou, Hiroshi Nishina, Hiromitsu Nakauchi. Prospective isolation and characterization of bipotent progenitor cells in early mouse liver development. Stem Cells Dev.. 2012.05; 21 (7): 1124-1133. ( PubMed, DOI )

  69. Toshiyuki Oishi, Shuji Terai, Shinya Kuwashiro, Koichi Fujisawa, Toshihiko Matsumoto, Hiroshi Nishina, Isao Sakaida. Ezetimibe reduces fatty acid quantity in liver and decreased inflammatory cell infiltration and improved NASH in medaka model. Biochem. Biophys. Res. Commun.. 2012.05; 422 (1): 22-27. ( PubMed, DOI )

  70. Yoshimi Uchida, Tomomi Osaki, Tokiwa Yamasaki, Tadanori Shimomura, Shoji Hata, Kazumasa Horikawa, Shigenobu Shibata, Takeshi Todo, Jun Hirayama, Hiroshi Nishina. Involvement of stress kinase mitogen-activated protein kinase kinase 7 in regulation of mammalian circadian clock. J. Biol. Chem.. 2012.03; 287 (11): 8318-8326. ( PubMed, DOI )

  71. Takuya Iwamoto, Shuji Terai, Yuko Mizunaga, Naoki Yamamoto, Kaoru Omori, Koichi Uchida, Takahiro Yamasaki, Yasuhiko Fujii, Hiroshi Nishina, Isao Sakaida. Splenectomy enhances the anti-fibrotic effect of bone marrow cell infusion and improves liver function in cirrhotic mice and patients. J. Gastroenterol.. 2012.03; 47 (3): 300-312. ( PubMed, DOI )

  72. Tadashi Yokoi, Yuko Seko, Tae Yokoi, Hatsune Makino, Shin Hatou, Masakazu Yamada, Tohru Kiyono, Akihiro Umezawa, Hiroshi Nishina, Noriyuki Azuma. Establishment of functioning human corneal endothelial cell line with high growth potential. PLoS ONE. 2012; 7 (1): e29677. ( PubMed, DOI )

  73. Yuko Mizunaga, Shuji Terai, Naoki Yamamoto, Koichi Uchida, Takahiro Yamasaki, Hiroshi Nishina, Yusuke Fujita, Koh Shinoda, Yoshihiko Hamamoto, Isao Sakaida. Granulocyte colony-stimulating factor and interleukin-1β are important cytokines in repair of the cirrhotic liver after bone marrow cell infusion: comparison of humans and model mice. Cell Transplant. 2012; 21 (11): 2363-2375. ( PubMed, DOI )

  74. Tokiwa Yamasaki, Hiroshi Kawasaki, Hiroshi Nishina. Diverse Roles of JNK and MKK Pathways in the Brain. J Signal Transduct. 2012; 2012 459265. ( PubMed, DOI )

  75. Hiroshi Yukiura, Kotaro Hama, Keita Nakanaga, Masayuki Tanaka, Yoichi Asaoka, Shinichi Okudaira, Naoaki Arima, Asuka Inoue, Takafumi Hashimoto, Hiroyuki Arai, Atsuo Kawahara, Hiroshi Nishina, Junken Aoki. Autotaxin regulates vascular development via multiple lysophosphatidic acid (LPA) receptors in zebrafish. J. Biol. Chem.. 2011.12; 286 (51): 43972-43983. ( PubMed, DOI )

  76. Tokiwa Yamasaki, Hiroshi Kawasaki, Satoko Arakawa, Kimiko Shimizu, Shigeomi Shimizu, Orly Reiner, Hideyuki Okano, Sachiko Nishina, Noriyuki Azuma, Josef M Penninger, Toshiaki Katada, Hiroshi Nishina. Stress-activated protein kinase MKK7 regulates axon elongation in the developing cerebral cortex. J. Neurosci.. 2011.11; 31 (46): 16872-16883. ( PubMed, DOI )

  77. Takuro Hisanaga, Shuji Terai, Takuya Iwamoto, Taro Takami, Naoki Yamamoto, Tomoaki Murata, Toshifumi Matsuyama, Hiroshi Nishina, Isao Sakaida. TNFR1-mediated signaling is important to induce the improvement of liver fibrosis by bone marrow cell infusion. Cell Tissue Res.. 2011.10; 346 (1): 79-88. ( PubMed, DOI )

  78. Yijun Bao, Kentaro Nakagawa, Zeyu Yang, Mitsunobu Ikeda, Kanchanamala Withanage, Mari Ishigami-Yuasa, Yukiko Okuno, Shoji Hata, Hiroshi Nishina, Yutaka Hata. A cell-based assay to screen stimulators of the Hippo pathway reveals the inhibitory effect of dobutamine on the YAP-dependent gene transcription. J. Biochem.. 2011.08; 150 (2): 199-208. ( PubMed, DOI )

  79. S Takahashi, A Ebihara, H Kajiho, K Kontani, H Nishina, T Katada. RASSF7 negatively regulates pro-apoptotic JNK signaling by inhibiting the activity of phosphorylated-MKK7. Cell Death Differ.. 2011.04; 18 (4): 645-655. ( PubMed, DOI )

  80. Norio Miyamura, Takashi Nakamura, Naoko Goto-Inoue, Nobuhiro Zaima, Takahiro Hayasaka, Tokiwa Yamasaki, Shuji Terai, Isao Sakaida, Mitsutoshi Setou, Hiroshi Nishina. Imaging mass spectrometry reveals characteristic changes in triglyceride and phospholipid species in regenerating mouse liver. Biochem. Biophys. Res. Commun.. 2011.04; 408 (1): 120-125. ( PubMed, DOI )

  81. Shinya Kuwashiro, Shuji Terai, Toshiyuki Oishi, Koichi Fujisawa, Toshihiko Matsumoto, Hiroshi Nishina, Isao Sakaida. Telmisartan improves nonalcoholic steatohepatitis in medaka (Oryzias latipes) by reducing macrophage infiltration and fat accumulation. Cell Tissue Res.. 2011.04; 344 (1): 125-134. ( PubMed, DOI )

  82. Shinya Takahashi, Kyoko Sakurai, Arisa Ebihara, Hiroaki Kajiho, Kota Saito, Kenji Kontani, Hiroshi Nishina, Toshiaki Katada. RhoA activation participates in rearrangement of processing bodies and release of nucleated AU-rich mRNAs. Nucleic Acids Res.. 2011.04; 39 (8): 3446-3457. ( PubMed, DOI )

  83. Tomomi Osaki, Yoshimi Uchida, Jun Hirayama, Hiroshi Nishina. Diphenyleneiodonium chloride, an inhibitor of reduced nicotinamide adenine dinucleotide phosphate oxidase, suppresses light-dependent induction of clock and DNA repair genes in zebrafish. Biol. Pharm. Bull.. 2011; 34 (8): 1343-1347. ( PubMed )

  84. Jinzhan Wu, Junko Kubota, Jun Hirayama, Yoko Nagai, Sachiko Nishina, Tadashi Yokoi, Yoichi Asaoka, Jungwon Seo, Nao Shimizu, Hiroaki Kajiho, Takashi Watanabe, Noriyuki Azuma, Toshiaki Katada, Hiroshi Nishina. p38 Mitogen-activated protein kinase controls a switch between cardiomyocyte and neuronal commitment of murine embryonic stem cells by activating myocyte enhancer factor 2C-dependent bone morphogenetic protein 2 transcription. Stem Cells Dev.. 2010.11; 19 (11): 1723-1734. ( PubMed, DOI )

  85. Yoichi Asaoka, Hiroshi Nishina. Diverse physiological functions of MKK4 and MKK7 during early embryogenesis. J. Biochem.. 2010.10; 148 (4): 393-401. ( PubMed, DOI )

  86. Ryuichi Mashima, Kazuho Honda, Yi Yang, Yohei Morita, Akane Inoue, Sumimasa Arimura, Hiroshi Nishina, Hideo Ema, Hiromitsu Nakauchi, Brian Seed, Hideaki Oda, Yuji Yamanashi. Mice lacking Dok-1, Dok-2, and Dok-3 succumb to aggressive histiocytic sarcoma. Lab. Invest.. 2010.09; 90 (9): 1357-1364. ( PubMed, DOI )

  87. Toshihiko Matsumoto, Shuji Terai, Toshiyuki Oishi, Shinya Kuwashiro, Koichi Fujisawa, Naoki Yamamoto, Yusuke Fujita, Yoshihiko Hamamoto, Makoto Furutani-Seiki, Hiroshi Nishina, Isao Sakaida. Medaka as a model for human nonalcoholic steatohepatitis. Dis Model Mech. 2010.07; 3 (7-8): 431-440. ( PubMed, DOI )

  88. Jungwon Seo, Yoichi Asaoka, Yoko Nagai, Jun Hirayama, Tokiwa Yamasaki, Misako Namae, Shinya Ohata, Nao Shimizu, Takahiro Negishi, Daiju Kitagawa, Hisato Kondoh, Makoto Furutani-Seiki, Josef M Penninger, Toshiaki Katada, Hiroshi Nishina. Negative regulation of wnt11 expression by Jnk signaling during zebrafish gastrulation. J. Cell. Biochem.. 2010.07; 110 (4): 1022-1037. ( PubMed, DOI )

  89. Masahiko Tanaka, Hideyuki Hara, Hiroshi Nishina, Kentaro Hanada, Ken'ichi Hagiwara, Tomohiko Maehama. An improved method for cell-to-cell transmission of infectious prion. Biochem. Biophys. Res. Commun.. 2010.07; 397 (3): 505-508. ( PubMed, DOI )

  90. Yoko Nagai, Yoichi Asaoka, Misako Namae, Kota Saito, Haruka Momose, Hiroshi Mitani, Makoto Furutani-Seiki, Toshiaki Katada, Hiroshi Nishina. The LIM protein Ajuba is required for ciliogenesis and left-right axis determination in medaka. Biochem. Biophys. Res. Commun.. 2010.06; 396 (4): 887-893. ( PubMed, DOI )

  91. G Gregory Neely, Keiji Kuba, Anthony Cammarato, Kazuya Isobe, Sabine Amann, Liyong Zhang, Mitsushige Murata, Lisa Elmén, Vaijayanti Gupta, Suchir Arora, Rinku Sarangi, Debasis Dan, Susumu Fujisawa, Takako Usami, Cui-ping Xia, Alex C Keene, Nakissa N Alayari, Hiroyuki Yamakawa, Ulrich Elling, Christian Berger, Maria Novatchkova, Rubina Koglgruber, Keiichi Fukuda, Hiroshi Nishina, Mitsuaki Isobe, J Andrew Pospisilik, Yumiko Imai, Arne Pfeufer, Andrew A Hicks, Peter P Pramstaller, Sai Subramaniam, Akinori Kimura, Karen Ocorr, Rolf Bodmer, Josef M Penninger. A global in vivo Drosophila RNAi screen identifies NOT3 as a conserved regulator of heart function. Cell. 2010.04; 141 (1): 142-153. ( PubMed, DOI )

  92. Kentaro Nakagawa, Misato Sugahara, Tokiwa Yamasaki, Hiroaki Kajiho, Shinya Takahashi, Jun Hirayama, Yasuhiro Minami, Yasutaka Ohta, Toshio Watanabe, Yutaka Hata, Toshiaki Katada, Hiroshi Nishina. Filamin associates with stress signalling kinases MKK7 and MKK4 and regulates JNK activation. Biochem. J.. 2010.04; 427 (2): 237-245. ( PubMed, DOI )

  93. S Shimizu, A Konishi, Y Nishida, T Mizuta, H Nishina, A Yamamoto, Y Tsujimoto. Involvement of JNK in the regulation of autophagic cell death. Oncogene. 2010.04; 29 (14): 2070-2082. ( PubMed, DOI )

  94. Takahiro Negishi, Yoko Nagai, Yoichi Asaoka, Mami Ohno, Misako Namae, Hiroshi Mitani, Takashi Sasaki, Nobuyoshi Shimizu, Shuji Terai, Isao Sakaida, Hisato Kondoh, Toshiaki Katada, Makoto Furutani-Seiki, Hiroshi Nishina. Retinoic acid signaling positively regulates liver specification by inducing wnt2bb gene expression in medaka. Hepatology. 2010.03; 51 (3): 1037-1045. ( PubMed, DOI )

  95. Yoshimi Uchida, Jun Hirayama, Hiroshi Nishina. A common origin: signaling similarities in the regulation of the circadian clock and DNA damage responses. Biol. Pharm. Bull.. 2010; 33 (4): 535-544. ( PubMed )

  96. Ryota Saito, Tokiwa Yamasaki, Yoko Nagai, Jinzhan Wu, Hiroaki Kajiho, Tadashi Yokoi, Eiichiro Noda, Sachiko Nishina, Hitoshi Niwa, Noriyuki Azuma, Toshiaki Katada, Hiroshi Nishina. CrxOS maintains the self-renewal capacity of murine embryonic stem cells. Biochem. Biophys. Res. Commun.. 2009.12; 390 (4): 1129-1135. ( PubMed, DOI )

  97. Jun Hirayama, Norio Miyamura, Yoshimi Uchida, Yoichi Asaoka, Reiko Honda, Kenji Sawanobori, Takeshi Todo, Takuro Yamamoto, Paolo Sassone-Corsi, Hiroshi Nishina. Common light signaling pathways controlling DNA repair and circadian clock entrainment in zebrafish. Cell Cycle. 2009.09; 8 (17): 2794-2801. ( PubMed )

  98. Takashi Nakamura, Hiroshi Nishina. Liver development: lessons from knockout mice and mutant fish. Hepatol. Res.. 2009.07; 39 (7): 633-644. ( PubMed, DOI )

  99. Norio Miyamura, Jun Hirayama, Kenji Sawanobori, Teruya Tamaru, Yoichi Asaoka, Reiko Honda, Takuro Yamamoto, Hatsume Uno, Ken Takamatsu, Hiroshi Nishina. CLOCK:BMAL-independent circadian oscillation of zebrafish Cryptochrome1a gene. Biol. Pharm. Bull.. 2009.07; 32 (7): 1183-1187. ( PubMed )

  100. Nao Shimizu, Hajime Watanabe, Junko Kubota, Jinzhan Wu, Ryota Saito, Tadashi Yokoi, Takumi Era, Takeshi Iwatsubo, Takashi Watanabe, Sachiko Nishina, Noriyuki Azuma, Toshiaki Katada, Hiroshi Nishina. Pax6-5a promotes neuronal differentiation of murine embryonic stem cells. Biol. Pharm. Bull.. 2009.06; 32 (6): 999-1003. ( PubMed )

  101. Shuhei Tanemura, Haruka Momose, Nao Shimizu, Daiju Kitagawa, Jungwon Seo, Tokiwa Yamasaki, Kentaro Nakagawa, Hiroaki Kajiho, Josef M Penninger, Toshiaki Katada, Hiroshi Nishina. Blockage by SP600125 of Fcepsilon receptor-induced degranulation and cytokine gene expression in mast cells is mediated through inhibition of phosphatidylinositol 3-kinase signalling pathway. J. Biochem.. 2009.03; 145 (3): 345-354. ( PubMed, DOI )

  102. Shinya Ohata, Makiko Nawa, Takeshi Kasama, Tokiwa Yamasaki, Kenji Sawanobori, Shoji Hata, Takashi Nakamura, Yoichi Asaoka, Toshio Watanabe, Hitoshi Okamoto, Takahiko Hara, Shuji Terai, Isao Sakaida, Toshiaki Katada, Hiroshi Nishina. Hematopoiesis-dependent expression of CD44 in murine hepatic progenitor cells. Biochem. Biophys. Res. Commun.. 2009.02; 379 (4): 817-823. ( PubMed, DOI )

  103. Yoshifumi Matsumoto, Hiroki Oota, Yoichi Asaoka, Hiroshi Nishina, Koji Watanabe, Janusz M Bujnicki, Shoji Oda, Shoji Kawamura, Hiroshi Mitani. Medaka: a promising model animal for comparative population genomics. BMC Res Notes. 2009; 2 88. ( PubMed, DOI )

  104. Tomohiko Maehama, Masahiko Tanaka, Hiroshi Nishina, Makoto Murakami, Yasunori Kanaho, Kentaro Hanada. RalA functions as an indispensable signal mediator for the nutrient-sensing system. J. Biol. Chem.. 2008.12; 283 (50): 35053-35059. ( PubMed, DOI )

  105. Shinya Takahashi, Yasuhiro Araki, Yuriko Ohya, Takeshi Sakuno, Shin-Ichi Hoshino, Kenji Kontani, Hiroshi Nishina, Toshiaki Katada. Upf1 potentially serves as a RING-related E3 ubiquitin ligase via its association with Upf3 in yeast. RNA. 2008.09; 14 (9): 1950-1958. ( PubMed, DOI )

  106. Shizue Ohsawa, Tomomi Watanabe, Toshiaki Katada, Hiroshi Nishina, Masayuki Miura. Novel antibody to human BASP1 labels apoptotic cells post-caspase activation. Biochem. Biophys. Res. Commun.. 2008.07; 371 (4): 639-643. ( PubMed, DOI )

  107. Yoshio Marumoto, Shuji Terai, Yohei Urata, Toshihiko Matsumoto, Yuko Mizunaga, Naoki Yamamoto, Haiyan Jin, Koichi Fujisawa, Tomoaki Murata, Koh Shinoda, Hiroshi Nishina, Isao Sakaida. Continuous high expression of XBP1 and GRP78 is important for the survival of bone marrow cells in CCl4-treated cirrhotic liver. Biochem. Biophys. Res. Commun.. 2008.03; 367 (3): 546-552. ( PubMed, DOI )

  108. T Wada, E Stepniak, L Hui, A Leibbrandt, T Katada, H Nishina, E F Wagner, J M Penninger. Antagonistic control of cell fates by JNK and p38-MAPK signaling. Cell Death Differ.. 2008.01; 15 (1): 89-93. ( PubMed, DOI )

  109. Takashi Nakamura, Yuuki Imai, Takahiro Matsumoto, Shingo Sato, Kazusane Takeuchi, Katsuhide Igarashi, Yoshifumi Harada, Yoshiaki Azuma, Andree Krust, Yoko Yamamoto, Hiroshi Nishina, Shu Takeda, Hiroshi Takayanagi, Daniel Metzger, Jun Kanno, Kunio Takaoka, T John Martin, Pierre Chambon, Shigeaki Kato. Estrogen prevents bone loss via estrogen receptor alpha and induction of Fas ligand in osteoclasts. Cell. 2007.09; 130 (5): 811-823. ( PubMed, DOI )

  110. Seiji Ura, Hiroshi Nishina, Yukiko Gotoh, Toshiaki Katada. Activation of the c-Jun N-terminal kinase pathway by MST1 is essential and sufficient for the induction of chromatin condensation during apoptosis. Mol. Cell. Biol.. 2007.08; 27 (15): 5514-5522. ( PubMed, DOI )

  111. Tsuyoshi Ishikawa, Shuji Terai, Yohei Urata, Yoshio Marumoto, Koji Aoyama, Tomoaki Murata, Yuko Mizunaga, Naoki Yamamoto, Hiroshi Nishina, Koh Shinoda, Isao Sakaida. Administration of fibroblast growth factor 2 in combination with bone marrow transplantation synergistically improves carbon-tetrachloride-induced liver fibrosis in mice. Cell Tissue Res.. 2007.03; 327 (3): 463-470. ( PubMed, DOI )

  112. Shoji Hata, Misako Namae, Hiroshi Nishina. Liver development and regeneration: from laboratory study to clinical therapy. Dev. Growth Differ.. 2007.02; 49 (2): 163-170. ( PubMed, DOI )

  113. Daiju Kitagawa, Hiroaki Kajiho, Takahiro Negishi, Seiji Ura, Tomomi Watanabe, Teiji Wada, Hidenori Ichijo, Toshiaki Katada, Hiroshi Nishina. Release of RASSF1C from the nucleus by Daxx degradation links DNA damage and SAPK/JNK activation. EMBO J.. 2006.07; 25 (14): 3286-3297. ( PubMed, DOI )

  114. Yuichiro Yokoyama, Shuji Terai, Tsuyoshi Ishikawa, Koji Aoyama, Yohei Urata, Yoshio Marumoto, Hiroshi Nishina, Kazuyuki Nakamura, Kiwamu Okita, Isao Sakaida. Proteomic analysis of serum marker proteins in recipient mice with liver cirrhosis after bone marrow cell transplantation. Proteomics. 2006.04; 6 (8): 2564-2570. ( PubMed, DOI )

  115. Tsuyoshi Ishikawa, Shuji Terai, Yohei Urata, Yoshio Marumoto, Koji Aoyama, Isao Sakaida, Tomoaki Murata, Hiroshi Nishina, Koh Shinoda, Shunji Uchimura, Yoshihiko Hamamoto, Kiwamu Okita. Fibroblast growth factor 2 facilitates the differentiation of transplanted bone marrow cells into hepatocytes. Cell Tissue Res.. 2006.02; 323 (2): 221-231. ( PubMed, DOI )

  116. Teiji Wada, Tomoki Nakashima, Nishina Hiroshi, Josef M Penninger. RANKL-RANK signaling in osteoclastogenesis and bone disease. Trends Mol Med. 2006.01; 12 (1): 17-25. ( PubMed, DOI )

  117. Isao Sakaida, Shuji Terai, Hiroshi Nishina, Kiwamu Okita. Development of cell therapy using autologous bone marrow cells for liver cirrhosis. Med Mol Morphol. 2005.12; 38 (4): 197-202. ( PubMed, DOI )

  118. Masataka Asagiri, Kojiro Sato, Takako Usami, Sae Ochi, Hiroshi Nishina, Hiroki Yoshida, Ikuo Morita, Erwin F Wagner, Tak W Mak, Edgar Serfling, Hiroshi Takayanagi. Autoamplification of NFATc1 expression determines its essential role in bone homeostasis. J. Exp. Med.. 2005.11; 202 (9): 1261-1269. ( PubMed, DOI )

  119. Taro Takami, Shuji Terai, Yuichiro Yokoyama, Haruko Tanimoto, Kunihiko Tajima, Koichi Uchida, Takahiro Yamasaki, Isao Sakaida, Hiroshi Nishina, Snorri S Thorgeirsson, Kiwamu Okita. Human homologue of maid is a useful marker protein in hepatocarcinogenesis. Gastroenterology. 2005.05; 128 (5): 1369-1380. ( PubMed )

  120. Kota Saito, Yasuhiro Araki, Kenji Kontani, Hiroshi Nishina, Toshiaki Katada. Novel role of the small GTPase Rheb: its implication in endocytic pathway independent of the activation of mammalian target of rapamycin. J. Biochem.. 2005.03; 137 (3): 423-430. ( PubMed, DOI )

  121. 和田悌司, 中島友樹, 仁科博史, Josef M Penninger. アダプター分子Gab2は骨密度を制御する 細胞工学. 2005; (24): 704-705.

  122. Shuji Terai, Isao Sakaida, Hiroshi Nishina, Kiwamu Okita. Lesson from the GFP/CCl4 model--translational research project: the development of cell therapy using autologous bone marrow cells in patients with liver cirrhosis. J Hepatobiliary Pancreat Surg. 2005; 12 (3): 203-207. ( PubMed, DOI )

  123. Kota Saito, Hiroaki Kajiho, Yasuhiro Araki, Hiroshi Kurosu, Kenji Kontani, Hiroshi Nishina, Toshiaki Katada. Purification and analysis of RIN family-novel Rab5 GEFs. Meth. Enzymol.. 2005; 403 276-283. ( PubMed, DOI )

  124. 仁科 博史. 動物における臓器サイズ制御機構 再生医療. 2022.05; 21 (2): 74-79. ( 医中誌 )

  125. Kuleape Joshua Agbemefa, Hossain Shakhawoat, Sinclear Caleb Kwame, Shimizu Takanobu, Iwasa Hiroaki, Maruyama Junichi, Arimoto-Matsuzaki Kyoko, Nishina Hiroshi, Hata Yutaka. DNA Damage Triggers the Nuclear Accumulation of RASSF6 Tumor Suppressor Protein via CDK9 and BAF53 To Regulate p53 Target Gene Transcription MOLECULAR AND CELLULAR BIOLOGY. 2022.02; 42 (2):

  126. Kamimura Kenya, Yokoo Takeshi, Abe Hiroyuki, Ohtsuka Masato, Miura Hiromi, Nishina Hiroshi, Terai Shuji. Development of Hydrodynamics-Based Gene Therapy for Liver Cancer MOLECULAR THERAPY. 2021.04; 29 (4): 55-56.

  127. Jiang Xinliang, Maruyama Junichi, Iwasa Hiroaki, Arimoto-Matsuzaki Kyoko, Nishina Hiroshi, Hata Yutaka. Heat shock induces the nuclear accumulation of YAP1 via SRC (vol 399, 112439, 2021) EXPERIMENTAL CELL RESEARCH. 2021.04; 401 (2): 112555. ( PubMed, DOI )

  128. 仁科 博史. 【細胞競合による生体制御とがん】細胞競合とがん Hippo-YAPシグナル経路を介した異常細胞の排除 医学のあゆみ. 2020.08; 274 (5): 451-455. ( 医中誌 )

  129. 宮村 憲央, 仁科 博史. Hippo-YAPシグナル伝達経路による異常細胞の排除 生化学. 2018.12; 90 (6): 804-809. ( 医中誌 )

  130. Inoue Ryosuke, Kamimura Kenya, Nagoya Takuro, Sakai Norihiro, Yokoo Takeshi, Goto Ryo, Ogawa Kohei, Shinagawa-Kobayashi Yoko, Watanabe-Mori Yukari, Sakamaki Akira, Abe Satoshi, Kamimura Hiroteru, Miyamura Norio, Nishina Hiroshi, Terai Shuji. Effect of a neural relay on liver regeneration in mice: activation of serotonin release from the gastrointestinal tract FEBS OPEN BIO. 2018.03; 8 (3): 449-460. ( PubMed, DOI )

  131. Shinagawa-Kobayashi Yoko, Kamimura Kenya, Goto Ryo, Ogawa Kohei, Inoue Ryosuke, Yokoo Takeshi, Sakai Norihiro, Nagoya Takuro, Sakamaki Akira, Abe Satoshi, Sugitani Soichi, Yanagi Masahiko, Fujisawa Koichi, Nozawa Yoshizu, Koyama Naoto, Nishina Hiroshi, Furutani-Seiki Makoto, Sakaida Isao, Terai Shuji. Effect of histidine on sorafenib-induced vascular damage: Analysis using novel medaka fish model BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS. 2018.02; 496 (2): 556-561. ( PubMed, DOI )

  132. Maruyama Junichi, Inami Kazutoshi, Michishita Fumiyoshi, Jiang Xinliang, Iwasa Hiroaki, Nakagawa Kentaro, Ishigami-Yuasa Mari, Kagechika Hiroyuki, Miyamura Norio, Hirayama Jun, Nishina Hiroshi, Nogawa Daichi, Yamamoto Kouhei, Hata Yutaka. Novel YAP1 Activator, Identified by Transcription-Based Functional Screen, Limits Multiple Myeloma Growth MOLECULAR CANCER RESEARCH. 2018.02; 16 (2): 197-211. ( PubMed, DOI )

  133. Miyamura Norio, Nishina Hiroshi. YAP regulates liver size and function CELL CYCLE. 2018; 17 (3): 267-268. ( PubMed, DOI )

  134. 岡本 好海[内田], Yu Ruoxing, 仁科 博史. 【日本のイノベーションのアクセラレーター】ES細胞分化系を用いた簡便・高解像度な初期胚発生に対する医薬品リスク評価 ファルマシア. 2017.12; 53 (12): 1182-1186. ( 医中誌 )

  135. 仁科 博史. 細胞張力可視化プローブを用いた3D組織構築機構の解明 上原記念生命科学財団研究報告集. 2017.12; 31 1-5. ( 医中誌 )

  136. 宮村 憲央, 仁科 博史. モデル生物を用いた肝再生研究 臨床免疫・アレルギー科. 2017.11; 68 (5): 550-555. ( 医中誌 )

  137. 仁科 博史. メダカを用いた肝臓研究 お茶の水医学雑誌. 2017.01; 65 (1): 87-95. ( 医中誌 )

  138. 千葉恭敬, 仁科博史. 肝臓形成および肝がんにおけるHippo-YAPシグナル経路の役割 医学のあゆみ . 2014; (251): 405-409.

  139. 畠 星治, 堅田利明, 仁科博史. 器官サイズを調節する転写共役因子YAPの活性制御 生化学. 2014; (86): 464-468.

  140. 山崎世和, 仁科博史. 脳におけるSAPK/JNKシグナルの役割 CLINICAL NEUROSCIENCE. 2013; (31): 654-656.

  141. 平山 順, 仁科博史. 活性酸素シグナルと概日リズム 実験医学. 2012.11; 36-41.

  142. 宮村憲央, 仁科博史. モデル生物を用いた肝発生および肝サイズ制御機構の解明 肝胆膵. 2012; (65): 21-28.

  143. 宮村憲央, 瀬藤光利, 仁科博史. 質量顕微鏡法を用いたマウス再生肝の解析 生化学. 2012; (84): 680-684.

  144. 畠 星治, 仁科博史. Hippoシグナリング シグナル伝達キーワード事典. 2012; 58-60.

  145. 畑 裕, 仁科博史. 癌・細胞死・再生の新たな鍵を握る器官サイズ制御シグナル 細胞工学. 2011.09; 30 (9):

  146. 浅岡洋一, 仁科博史. メダカとゼブラフィッシュを用いた肝研究 実験医学. 2011.08; 29 (13): 2090-2095.

  147. 仁科博史. モデル生物‐マウスと小型魚類 トランスポートソームの世界-膜輸送研究の源流から未来へ. 2011; 434-439.

  148. 仁科博史. 小型魚類メダカを用いた肝形成および肝疾患研究 幹細胞研究会ホームページ. 2010;

  149. 中村 貴, 早坂孝宏, 井上菜穂子, 仁科博史, 瀬藤光利. メタボロームの分布可視化法について メタボロミクス:その解析技術と臨床・創薬応用研究の最前線, メディカルドゥ. 2010; (16): 131-135.

  150. 平山 順, 内田好海、宮村憲央、沢登健治、仁科博史. 概日リズムによる生理機能の制御機構 自律神経. 2010; (47): 297-300.

  151. 寺井崇二, 浦田洋平, 丸本芳雄, 石川 剛, 青山浩司, 大森 薫, 山本直樹, 坂井田功, 仁科博史, 奥本和夫, 斉藤貴史, 河田純男, 沖田 極,  . 他施設臨床研究: 肝硬変症に対するAMBI療法の開発 再生医療. 2006; (5): 79-87.

  152. 仁科博史. 一枚の写真館 肝臓に生じたブラックホール? 細胞工学. 2006; (25):

  153. 北川大樹, 堅田利明, 仁科博史. 新規核内複合体Daxx-RASSF1CはDNA損傷センターとして機能してシグナルを核から細胞質へと伝達する 細胞工学. 2006; (25): 1178-1179.

  154. 仁科博史, 堅田利明 . MAPキナーゼ 生物薬科学実験講座 . 2005; 424-433.

  155. 浦 誠司, 仁科博史. MAPキナーゼとストレス応答 シグナル伝達 集中マスター. 2005; 58-67.

  156. 仁科博史. 細胞を利用した治療 医薬品の開発と生産. 2005; 108-112.

▼全件表示

書籍等出版物 【 表示 / 非表示

  1. 分子細胞生物学 第9版 (Lodish et al.). 東京化学同人, 2023.07 (ISBN : 9784807920518)

  2. 分子細胞生物学 第8版 (Lodish et al.). 東京化学同人, 2019.12

  3. 薬学用語辞典. 東京化学同人, 2012.03

  4. 生物学事典. 東京化学同人, 2010.12

  5. 仁科博史. シグナル伝達の分子機構: ベーシックマスター生化学. オーム社, 2008.11

講演・口頭発表等 【 表示 / 非表示

  1. 仁科 博史. Hippoシグナル経路の新展開 器官サイズと恒常性を制御するHippo-YAPシグナル伝達経路(The Hippo-YAP pathway regulates organ size and homeostasis). 日本細胞生物学会大会講演要旨集 2021.06.01

  2. 仁科 博史. 肝細胞の生物学Up to Date 細胞の競合と肝臓の恒常性(Cell competition and liver homeostasis). 糖尿病 2020.08.01

  3. 丸山 順一, 影近 弘之, 仁科 博史, 畑 裕. 新規に同定したYAP1活性化化合物は多発性骨髄腫細胞においてYAP1-p73経路依存的な細胞死を誘導する. 日本癌学会総会記事 2017.09.01

  4. 虞 若星, 岡本 好海[内田], 仁科 博史. タンパク質ファルネシル化を介したメバロン酸経路による原始線条形成の制御機構の解明. 日本薬学会年会要旨集 2017.03.01

  5. Hiroshi Nishina. Mouse Embryonic Stem Cell-Based Drug Screen for Novel Modulators of Cell Differentiation in Early Mammalian Embryogenesis . 6th Asian Oceania Zebrafish PI’s Meeting (AOZM), Hong Kong, USA 2013.07.19

  6. 仁科博史. Mouse Embryonic Stem Cell-Based Drug Screen for Novel Modulators of Cell Differentiation in Early Mammalian Embryogenesis. 熊本大学HIGO Program 2012.02.12

  7. 辻本 和峰, 橋本 貢士, 榛澤 望, 川堀 健一, 袁 勲梅, 長岡 勇也, 仁科 博史, 畑田 出穂, 山田 哲也, 小川 佳宏. エピゲノム改変動物を用いたFibroblast growth factor 21(FGF21)遺伝子DNAメチル化の機能的意義解明. 糖尿病 2021.05.01

  8. 西川 祐司, 大塩 貴子, 山本 雅大, 藤井 清永, 辛 氷, 後藤 正憲, 岡田 陽子, 鈴木 聡, ペニンジャー・ジョゼフ , 仁科 博史. 肝傷害後の組織修復におけるストレスキナーゼMKK7の役割 uPA活性化の関与. 日本病理学会会誌 2021.03.01

  9. 小藤 智史, 進 匡, 仁科 博史. 育仔行動におけるMKK7-JNKシグナル伝達経路の役割の解明. 日本薬学会年会要旨集 2021.03.01

  10. 榛澤 望, 橋本 貢士, 袁 勲梅, 辻本 和峰, 川堀 健一, 長岡 勇也, 仁科 博史, 畑田 出穂, 山田 哲也, 小川 佳宏. Fibroblast growth factor(FGF)21遺伝子特異的DNA脱メチル化によるエピゲノム改変細胞および動物の樹立. 日本内分泌学会雑誌 2020.08.01

  11. 長岡 勇也, 石原 えりか, 仁科 博史. PGE2とその受容体EP2誘導性シグナルはYAP依存的細胞競合に必須である. 日本薬学会年会要旨集 2020.03.01

  12. 河嵜 麻実, 高杉 俊之, 玉田 篤史, 岡田 正康, 阿部 学, 崎村 建司, 仁科 博史, 五十嵐 道弘. 細胞の生・老・病・死を司るJNKの分子機構と役割 成長円錐のリン酸化プロテオミクスにより同定されたJNK基質の軸索伸長における役割. 日本生化学会大会プログラム・講演要旨集 2019.09.01

  13. 仁科 博史. 細胞の生・老・病・死を司るJNKの分子機構と役割 高次神経活動を制御するJNKシグナル. 日本生化学会大会プログラム・講演要旨集 2019.09.01

  14. 河嵜 麻実, 玉田 篤史, 奥田 修二郎, 崎村 建司, 仁科 博史, 五十嵐 道弘. 成長円錐リン酸化プロテオミクスによる神経成長のシグナル伝達解析. 日本生化学会大会プログラム・講演要旨集 2018.09.01

  15. 仁科 博史. 異分野連携が拓くシグナル伝達と疾患研究のフロンティア 神経系の恒常性維持を担うJNKシグナル経路. 日本生化学会大会プログラム・講演要旨集 2018.09.01

  16. 長岡 勇也, 仁科 博史. 転写共役因子YAP誘導性肝がん形成機構の解明. 日本薬学会年会要旨集 2018.03.01

  17. 長岡 勇也, 宮村 憲央, 仁科 博史. がん原遺伝子産物YAP依存的な肝がん誘導系の確立. 生命科学系学会合同年次大会 2017.12.01

  18. 宮村 憲央, 仁科 博史. 細胞競合の分子機構と生理的意義:どこまでわかって何がわからないのか YAP活性化によって誘導される肝細胞排除現象の発見. 生命科学系学会合同年次大会 2017.12.01

  19. 松平 竜之, 向井 康治朗, 野口 大心, 長谷川 純矢, 八田 知久, 家村 俊一郎, 夏目 徹, 宮村 憲央, 仁科 博史, 中山 淳, 仙波 憲太郎, 冨田 拓哉, 村田 茂穂, 新井 洋由, 田口 友彦. エンドソーム膜のホスファチジルセリンを介したYAPの活性化機構の解明. 生命科学系学会合同年次大会 2017.12.01

  20. 仁科博史. 細胞内シグナル伝達系. 秋田大学医学部セミナー 2014.12.12

  21. 仁科博史. がん原遺伝子産物YAP依存的肝細胞消失を誘導する新規マウスモデルの確立. 第37回日本分子生物学会 2014.11.25

  22. 浅岡洋一, 仁科博史. 網膜光受容細胞の分化におけるHippo-Yapシグナル伝達系の役割. 第37回日本分子生物学会 2014.11.25

  23. 浅岡洋一, 仁科博史. Hippo-Yap シグナル伝達系による網膜視細胞の分化制御機構. 第7回RRM 2014.11.22

  24. 平山 順, 他. ゼブラフィッシュ概日リズムの光同調を担うシグナル経路の解析. 第21回日本時間生物学会学術大会 2014.11.08

  25. 仁科博史. 成体マウス肝臓においてモザイク状のHippoシグナル伝達系破綻は細胞排除を誘導する. 第87回日本生化学会大会 2014.10.15

  26. 宮村憲央, 仁科博史. Hippoシグナル系破綻によって誘導されるマウス肝細胞の動態の解析. 第21回日本肝臓医生物学研究会 2014.10.04

  27. 仁科博史. スタチン催奇性誘導機構の解析. 第21回日本肝臓医生物学研究会 2014.10.04

  28. 濱部凛, 他. ノックアウトゼブラフィッシュを用いた時計遺伝子zPer2およびzCry1aの解析. 第3回修飾シグナル病若手ワークショップ 2014.09.30

  29. 濱部凛, 他. ノックアウトゼブラフィッシュを用いた時計遺伝子Per2とCry1aの解析. 第13回次世代を担う若手ファーマ・バイオフォーラム 2014.09.20

  30. 平山 順, 他. Study on a light signaling pathway for circadian entrainment in zebrafish. . 第20回小型魚類研究会 2014.09.20

  31. 浅岡洋一, 仁科博史. Hippo-Yap signaling acts as a molecular switch between retinal progenitor cell proliferation and photoreceptor cell differentiation.. 第20回小型魚類研究会 2014.09.20

  32. Yoichi Asaoka, 他. Hippo signaling regulates a switch between retinal progenitor cell proliferation and photoreceptor cell differentiation in zebrafish. . 日本比較生理生化学学会第36回大会 2014.07.28

  33. 仁科博史. 器官サイズを制御する転写共役因子YAPの役割. 第23回日本Cell Death学会 2014.07.18

  34. 浅岡洋一, 仁科博史. 器官サイズを制御するHippo-Yapシグナル伝達系の網膜分化における機能解析. 第23回日本Cell Death学会 2014.07.18

  35. 宮村憲央, 仁科博史. 成体マウス肝臓においてモザイク状のHippoシグナル伝達系破綻は細胞排除を誘導する. 第21回肝細胞研究会 2014.06.27

  36. 内田好海, 仁科博史. スタチン催奇性の分子機構の解明. 第21回肝細胞研究会 2014.06.27

  37. 内田好海, 仁科博史. スタチンの初期胚発生に対する作用機構の解明. 第13回生命科学研究会 2014.06.20

  38. 浅岡洋一, 仁科博史. 器官サイズ制御因子Yapの網膜視細胞分化における機能解析. 第13回生命科学研究会 2014.06.20

  39. 仁科博史. 器官サイズ制御シグナルHippo系による組織形成機構とその破綻病態. 東京女子医科大学セミナー 2014.05.29

  40. 有馬誉恵, 仁科博史. マウス初期胚におけるムスカリン性アセチルコリン受容体の機能解析. 第134回日本薬学会 2014.03.27

  41. Ruoxing Yu, Hiroshi Nishina. Assessment of teratogenic mechanisms of FDA pregnancy category D and X drugs using murine ES cell-derived 3D culture system. 第134回日本薬学会 2014.03.27

  42. 内田好海, 仁科博史. マウス胚性幹細胞を用いた薬剤スクリーニングによる三胚葉分化制御シグナルの同定とスタチン催奇性発症機構の解明. 第20回日本肝臓医生物学研究会 2014.02.15

  43. 仁科博史. 器官サイズ制御シグナルHippo系による組織形成機構とその破綻病態. 神戸大学医学部セミナー 2014.01.10

▼全件表示

その他業績 【 表示 / 非表示

  • 「原始線条形成制御因子としてセラミドを同定」 ― ノックアウトマウスデータベースとin vitro細胞分化誘導系の活用 ―,2023年10月

    Stem Cells

  • 新たな眼の難治疾患を発見 ― mRNA形成の障害によっておこる多彩な眼の先天形成異常 ―,2023年03月

    Human Molecular Genetics

  • 「遺伝子改変ゼブラフィッシュを用いて光刺激が体内時計を形成する仕組みを解明」― 体内時計の障害が行動量を低下させ、疾患の要因となる可能性が明らかに ―,2019年01月

    Scientific Reports

  • 「概日リズムや運動を制御する神経細胞内で働く遺伝子を同定」― 概日リズム障害と加齢依存的な運動異常を示す変異マウスの作出 ―,2017年08月

    Scientific Reports

  • 「損傷した肝細胞を排除する仕組みを発見」― 肝臓を構成する細胞の品質管理による恒常性維持機構 ―,2017年07月

    Nature Communications

  • 「発生初期の器官形成の鍵となる組織の分化に必要な代謝経路の解明」- 妊娠初期の安全な薬物治療設計への応用が期待 -,2016年11月

    Scientific Reports

  • 異常な細胞の除去を誘導する新たな仕組みの解明に成功,2016年06月

    Scientific Reports

  • 3D臓器形成遺伝子の同定に成功,2015年03月

    Nature

▼全件表示

 

担当授業科目(学内) 【 表示 / 非表示

  • 発生・再生科学,2014年 - 現在

  • 生命科学持論Ⅱ,2013年 - 現在

  • 発生学研究法

  • 細胞生物学持論

社会貢献活動 【 表示 / 非表示

  • 文藝春秋 連載「同級生交歓」,文藝春秋社,月刊誌「文藝春秋」,2020年12月29日