2024/04/13 更新

写真a

クボタ ショウ
久保田 翔
Sho Kubota
所属
医歯薬学域 助教
職名
助教
外部リンク

学位

  • 医薬学博士 ( 千葉大学 )

研究キーワード

  • 造血幹細胞

  • バイオインフォマティクス

  • NO

  • BPDCN

  • 細胞内シグナル伝達

  • エピジェネティクス

  • 環境化学物質

  • 白血病

  • チロシンキナーゼ

  • 転写因子

研究分野

  • ライフサイエンス / 薬理学

  • ライフサイエンス / 血液、腫瘍内科学

  • ライフサイエンス / 薬系衛生、生物化学

学歴

  • 千葉大学・薬学研究院・先端生命科学専攻・博士課程    

    2011年4月 - 2014年3月

      詳細を見る

  • 千葉大学・薬学研究院・修士課程    

    2009年4月 - 2011年3月

      詳細を見る

  • 千葉大学・薬学部・総合薬品科学科    

    2005年4月 - 2009年3月

      詳細を見る

経歴

  • 岡山大学・学術研究院医歯薬学域(薬学)・薬効解析学   助教

    2023年7月 - 現在

      詳細を見る

  • 熊本大学・国際先端医学研究機構・白血病転写制御   特任助教

    2022年4月 - 2023年6月

      詳細を見る

  • 熊本大学・国際先端医学研究機構・白血病転写制御   研究員・日本学術振興会特別研究員

    2015年3月 - 2022年3月

      詳細を見る

  • 千葉大学・大学院薬学研究院・分子細胞生物学研究室   日本学術振興会特別研究員

    2014年4月 - 2015年3月

      詳細を見る

所属学協会

委員歴

  • 幹細胞シンポジウム・若手の会・幹事  

    2019年4月 - 現在   

      詳細を見る

 

論文

  • The acidic domain of Hmga2 and the domain's linker region are critical for driving self-renewal of hematopoietic stem cell. 査読 国際共著 国際誌

    Yuqi Sun, Sho Kubota, Mihoko Iimori, Ai Hamashima, Haruka Murakami, Jie Bai, Mariko Morii, Takako Yokomizo-Nakano, Motomi Osato, Kimi Araki, Goro Sashida

    International journal of hematology   115 ( 4 )   553 - 562   2022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)  

    High mobility group AT-hook 2 (Hmga2) is a chromatin modifier protein that plays a critical role in fetal development and leukemia propagation by binding to chromatin and DNA via its AT-hook domains. However, the molecular mechanisms by which Hmga2 activates the expression of target genes to drive the self-renewal of hematopoietic stem cells (HSCs) remain unclear. We generated Rosa26 locus Hmga2 conditional knock-in mice and found that overexpression of Hmga2 promoted self-renewal of normal HSCs, but maintained their fitness in bone marrow, and consequently was not sufficient to initiate malignancy. This result is consistent with previous findings showing that Hmga2 is a proto-oncogene. We also assessed the cellular functions of Hmga2 mutants lacking functional domains and demonstrated that the C-terminus acidic domain of Hmga2 and the domain's linker region were critical for activating genes involved in stem cell signatures, such as the Igf2bp2 gene, to drive proliferation of HSCs. In contrast, overexpression of Hmga1, a member of the Hmga family with a different linker region, did not drive proliferation of HSCs. Our results reveal a critical role for the acidic domain of Hmga2 and the domain's linker region in modulating the transcription and self-renewal functions of HSCs.

    DOI: 10.1007/s12185-021-03274-9

    PubMed

    researchmap

  • Src-mediated tyrosine phosphorylation of PRC1 and kinastrin/SKAP on the mitotic spindle. 査読 国際誌

    Mariko Morii, Sho Kubota, Chizu Hasegawa, Yumi Takeda, Shiori Kometani, Kyoko Enomoto, Takayuki Suzuki, Sayuri Yanase, Rika Sato, Aki Akatsu, Kensuke Hirata, Takuya Honda, Takahisa Kuga, Takeshi Tomonaga, Yuji Nakayama, Noritaka Yamaguchi, Naoto Yamaguchi

    Scientific reports   11 ( 1 )   2616 - 2616   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)  

    Src-family tyrosine kinases (SFKs) play important roles in a number of signal transduction events during mitosis, such as spindle formation. A relationship has been reported between SFKs and the mitotic spindle; however, the underlying mechanisms remain unclear. We herein demonstrated that SFKs accumulated in the centrosome region at the onset of mitosis. Centrosomal Fyn increased in the G2 phase in a microtubule polymerization-dependent manner. A mass spectrometry analysis using mitotic spindle preparations was performed to identify tyrosine-phosphorylated substrates. Protein regulator of cytokinesis 1 (PRC1) and kinastrin/small kinetochore-associated protein (kinastrin/SKAP) were identified as SFK substrates. SFKs mainly phosphorylated PRC1 at Tyr-464 and kinastrin at Tyr-87. Although wild-type PRC1 is associated with microtubules, phosphomimetic PRC1 impaired the ability to bind microtubules. Phosphomimetic kinastrin at Tyr-87 also impaired binding with microtubules. Collectively, these results suggest that tyrosine phosphorylation of PRC1 and kinastrin plays a role in their delocalization from microtubules during mitosis.

    DOI: 10.1038/s41598-021-82189-1

    PubMed

    researchmap

  • Overexpression of RUNX3 represses RUNX1 to drive transformation of myelodysplastic syndrome. 査読 国際誌

    Takako Yokomizo-Nakano, Sho Kubota, Jie Bai, Ai Hamashima, Mariko Morii, Yuqi Sun, Seiichiro Katagiri, Mihoko Iimori, Akinori Kanai, Daiki Tanaka, Motohiko Oshima, Yuka Harada, Kazuma Ohyashiki, Atsushi Iwama, Hironori Harada, Motomi Osato, Goro Sashida

    Cancer research   80 ( 12 )   2523 - 2536   2020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)  

    RUNX3, a RUNX family transcription factor, regulates normal hematopoiesis and functions as a tumor suppressor in various tumors in humans and mice. However, emerging studies have documented increased expression of RUNX3 in hematopoietic stem/progenitor cells (HSPC) of a subset of patients with myelodysplastic syndrome (MDS) showing a worse outcome, suggesting an oncogenic function for RUNX3 in the pathogenesis of hematological malignancies. To elucidate the oncogenic function of RUNX3 in the pathogenesis of MDS in vivo, we generated a RUNX3-expressing, Tet2-deficient mouse model with the pan-cytopenia and dysplastic blood cells characteristic of MDS in patients. RUNX3-expressing cells markedly suppressed the expression levels of Runx1, a critical regulator of hemaotpoiesis in normal and malignant cells, as well as its target genes, which included crucial tumor suppressors such as Cebpa and Csf1r. RUNX3 bound these genes and remodeled their Runx1 binding regions in Tet2-deficient cells. Overexpression of RUNX3 inhibited the transcriptional function of Runx1 and compromised hematopoiesis to facilitate the development of MDS in the absence of Tet2, indicating that RUNX3 is an oncogene. Furthermore, overexpression of RUNX3 activated the transcription of Myc target genes and rendered cells sensitive to inhibition of Myc-Max heterodimerization. Collectively, these results reveal the mechanism by which RUNX3 overexpression exerts oncogenic effects on the cellular function of and transcriptional program in Tet2-deficient stem cells to drive the transformation of MDS.

    DOI: 10.1158/0008-5472.CAN-19-3167

    PubMed

    researchmap

  • Lineage-specific RUNX2 super-enhancer activates MYC and promotes the development of blastic plasmacytoid dendritic cell neoplasm. 査読 国際誌

    Sho Kubota, Kenji Tokunaga, Tomohiro Umezu, Takako Yokomizo-Nakano, Yuqi Sun, Motohiko Oshima, Kar Tong Tan, Henry Yang, Akinori Kanai, Eisaku Iwanaga, Norio Asou, Takahiro Maeda, Naomi Nakagata, Atsushi Iwama, Kazuma Ohyashiki, Motomi Osato, Goro Sashida

    Nature communications   10 ( 1 )   3943 - 3943   2019年8月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語  

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

    DOI: 10.1038/s41467-019-11919-x

    PubMed

    researchmap

  • Src Acts as an Effector for Ku70-dependent Suppression of Apoptosis through Phosphorylation of Ku70 at Tyr-530 査読

    Mariko Morii, Sho Kubota, Takuya Honda, Ryuzaburo Yuki, Takao Morinaga, Takahisa Kuga, Takeshi Tomonaga, Noritaka Yamaguchi, Naoto Yamaguchi

    JOURNAL OF BIOLOGICAL CHEMISTRY   292 ( 5 )   1648 - 1665   2017年2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  

    Src-family tyrosine kinases are widely expressed in many cell types and participate in a variety of signal transduction pathways. Despite the significance of Src in suppression of apoptosis, its mechanism remains poorly understood. Here we show that Src acts as an effector for Ku70-dependent suppression of apoptosis. Inhibition of endogenous Src activity promotes UV-induced apoptosis, which is impaired by Ku70 knockdown. Src phosphorylates Ku70 at Tyr-530, being close to the possible acetylation sites involved in promotion of apoptosis. Src-mediated phosphorylation of Ku70 at Tyr-530 decreases acetylation of Ku70, whereas Src inhibition augments acetylation of Ku70. Importantly, knockdown-rescue experiments with stable Ku70 knockdown cells show that the nonphosphorylatable Y530F mutant of Ku70 reduces the ability of Ku70 to suppress apoptosis accompanied by augmentation of Ku70 acetylation. Our results reveal that Src plays a protective role against hyperactive apoptotic cell death by reducing apoptotic susceptibility through phosphorylation of Ku70 at Tyr-530.

    DOI: 10.1074/jbc.M116.753202

    Web of Science

    PubMed

    researchmap

  • Role for Tyrosine Phosphorylation of A-kinase Anchoring Protein 8 (AKAP8) in its Dissociation from Chromatin and the Nuclear Matrix. 査読

    Sho Kubota, Mariko Morii, Ryuzaburo Yuki, Noritaka Yamaguchi, Hiromi Yamaguchi, Kazumasa Aoyama, Takahisa Kuga, Takeshi Tomonaga, Naoto Yamaguchi

    The Journal of biological chemistry   290 ( 17 )   10891 - 10904   2015年4月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)  

    DOI: 10.1074/jbc.M115.643882

    Web of Science

    PubMed

    researchmap

  • Phosphorylation of KRAB-associated Protein 1 (KAP1) at Tyr-449, Tyr-458, and Tyr-517 by Nuclear Tyrosine Kinases Inhibits the Association of KAP1 and Heterochromatin Protein 1α (HP1α) with Heterochromatin 査読

    Sho Kubota, Yasunori Fukumoto, Kazumasa Aoyama, Kenichi Ishibashi, Ryuzaburo Yuki, Takao Morinaga, Takuya Honda, Noritaka Yamaguchi, Takahisa Kuga, Takeshi Tomonaga, Naoto Yamaguchi

    Journal of Biological Chemistry   288 ( 24 )   17871 - 17883   2013年6月

     詳細を見る

    担当区分:筆頭著者   掲載種別:研究論文(学術雑誌)   出版者・発行元:Elsevier BV  

    DOI: 10.1074/jbc.m112.437756

    Web of Science

    PubMed

    researchmap

  • Multiple myeloma-associated DIS3 gene is essential for hematopoiesis but loss of DIS3 is insufficient for myelomagenesis 査読

    Hiroto Ohguchi, Yasuyo Ohguchi, Sho Kubota, Kan Etoh, Ai Hamashima, Shingo Usuki, Takako Yokomizo-Nakano, Jie Bai, Takeshi Masuda, Yawara Kawano, Takeshi Harada, Mitsuyoshi Nakao, Takashi Minami, Teru Hideshima, Kimi Araki, Goro Sashida

    Blood Neoplasia   100005 - 100005   2024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:Elsevier BV  

    DOI: 10.1016/j.bneo.2024.100005

    researchmap

  • Methyl vinyl ketone and its analogs covalently modify PI3K and alter physiological functions by inhibiting PI3K signaling.

    Morimoto A, NOBUMASA TAKASUGI, Pan Y, Kubota S, Dohmae N, Abiko Y, Uchida K, Kumagai Y, Uehara T

    The Journal of biological chemistry   2024年1月

     詳細を見る

    掲載種別:研究論文(学術雑誌)  

    Reactive carbonyl species (RCS), which are abundant in the environment and are produced in vivo under stress, covalently bind to nucleophilic residues such as Cys in proteins. Disruption of protein function by RCS exposure is predicted to play a role in the development of various diseases such as cancer and metabolic disorders, but most studies on RCS have been limited to simple cytotoxicity validation, leaving their target proteins and resulting physiological changes unknown. In this study, we focused on methyl vinyl ketone (MVK), which is one of the main RCS found in cigarette smoke and exhaust gas. We found that MVK suppressed PI3K-Akt signaling, which regulates processes involved in cellular homeostasis, including cell proliferation, autophagy, and glucose metabolism. Interestingly, MVK inhibits the interaction between the epidermal growth factor receptor and PI3K. Cys656 in the SH2 domain of the PI3K p85 subunit, which is the covalently binding site of MVK, is important for this interaction. Suppression of PI3K-Akt signaling by MVK reversed epidermal growth factor-induced negative regulation of autophagy and attenuated glucose uptake. Furthermore, we analyzed the effects of the 23 RCS compounds with structures similar to MVK and showed that their analogs also suppressed PI3K-Akt signaling in a manner that correlated with their similarities to MVK. Our study demonstrates the mechanism of MVK and its analogs in suppressing PI3K-Akt signaling and modulating physiological functions, providing a model for future studies analyzing environmental reactive species.

    DOI: 10.1016/j.jbc.2024.105679

    PubMed

    researchmap

  • Attenuation of protein arginine dimethylation via S-nitrosylation of protein arginine methyltransferase 1.

    Taniguchi R, Moriya Y, Dohmae N, Suzuki T, Nakahara K, Kubota S, Takasugi N, Uehara T

    Journal of pharmacological sciences   2023年12月

     詳細を見る

    掲載種別:研究論文(学術雑誌)  

    Upregulation of nitric oxide (NO) production contributes to the pathogenesis of numerous diseases via S-nitrosylation, a post-translational modification of proteins. This process occurs due to the oxidative reaction between NO and a cysteine thiol group; however, the extent of this reaction remains unknown. S-Nitrosylation of PRMT1, a major asymmetric arginine methyltransferase of histones and numerous RNA metabolic proteins, was induced by NO donor treatment. We found that nitrosative stress leads to S-nitrosylation of cysteine 119, located near the active site, and attenuates the enzymatic activity of PRMT1. Interestingly, RNA sequencing analysis revealed similarities in the changes in expression elicited by NO and PRMT1 inhibitors or knockdown. A comprehensive search for PRMT1 substrates using the proximity-dependent biotin identification method highlighted many known and new substrates, including RNA-metabolizing enzymes. To validate this result, we selected the RNA helicase DDX3 and demonstrated that arginine methylation of DDX3 is induced by PRMT1 and attenuated by NO treatment. Our results suggest the existence of a novel regulatory system associated with transcription and RNA metabolism via protein S-nitrosylation.

    DOI: 10.1016/j.jphs.2023.12.012

    PubMed

    researchmap

  • Exposure to microbial products followed by loss of Tet2 promotes myelodysplastic syndrome via remodeling HSCs. 査読 国際誌

    Takako Yokomizo-Nakano, Ai Hamashima, Sho Kubota, Jie Bai, Supannika Sorin, Yuqi Sun, Kenta Kikuchi, Mihoko Iimori, Mariko Morii, Akinori Kanai, Atsushi Iwama, Gang Huang, Daisuke Kurotaki, Hitoshi Takizawa, Hirotaka Matsui, Goro Sashida

    The Journal of experimental medicine   220 ( 7 )   2023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)  

    Aberrant innate immune signaling in myelodysplastic syndrome (MDS) hematopoietic stem/progenitor cells (HSPCs) has been implicated as a driver of the development of MDS. We herein demonstrated that a prior stimulation with bacterial and viral products followed by loss of the Tet2 gene facilitated the development of MDS via up-regulating the target genes of the Elf1 transcription factor and remodeling the epigenome in hematopoietic stem cells (HSCs) in a manner that was dependent on Polo-like kinases (Plk) downstream of Tlr3/4-Trif signaling but did not increase genomic mutations. The pharmacological inhibition of Plk function or the knockdown of Elf1 expression was sufficient to prevent the epigenetic remodeling in HSCs and diminish the enhanced clonogenicity and the impaired erythropoiesis. Moreover, this Elf1-target signature was significantly enriched in MDS HSPCs in humans. Therefore, prior infection stress and the acquisition of a driver mutation remodeled the transcriptional and epigenetic landscapes and cellular functions in HSCs via the Trif-Plk-Elf1 axis, which promoted the development of MDS.

    DOI: 10.1084/jem.20220962

    PubMed

    researchmap

  • CARD11 mutation and HBZ expression induce lymphoproliferative disease and adult T-cell leukemia/lymphoma 査読

    Takuro Kameda, Kotaro Shide, Ayako Kamiunten, Yasunori Kogure, Daisuke Morishita, Junji Koya, Yuki Tahira, Keiichi Akizuki, Takako Yokomizo-Nakano, Sho Kubota, Kosuke Marutsuka, Masaaki Sekine, Tomonori Hidaka, Yoko Kubuki, Yuichi Kitai, Tadashi Matsuda, Akinori Yoda, Takayuki Ohshima, Midori Sugiyama, Goro Sashida, Keisuke Kataoka, Seishi Ogawa, Kazuya Shimoda

    5 ( 1 )   2022年11月

     詳細を見る

    掲載種別:研究論文(学術雑誌)  

    DOI: 10.1038/s42003-022-04284-x

    researchmap

    その他リンク: https://www.nature.com/articles/s42003-022-04284-x

  • HMGN3 represses transcription of epithelial regulators to promote migration of cholangiocarcinoma in a SNAI2-dependent manner. 査読 国際誌

    Supannika Sorin, Sho Kubota, Sofiane Hamidi, Takako Yokomizo-Nakano, Kulthida Vaeteewoottacharn, Sopit Wongkham, Sakda Waraasawapati, Chawalit Pairojkul, Jie Bai, Mariko Morii, Guojun Sheng, Kanlayanee Sawanyawisuth, Goro Sashida

    FASEB journal : official publication of the Federation of American Societies for Experimental Biology   36 ( 7 )   e22345   2022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)  

    High mobility group nucleosome-binding protein 3 (HMGN3), a member of the HMGN family, modulates the structure of chromatin and regulates transcription through transcription factors. HMGN3 has been implicated in the development of various cancers; however, the underlying mechanisms remain unclear. We herein demonstrated that the high expression of HMGN3 correlated with the metastasis of liver fluke infection-induced cholangiocarcinoma (CCA) in patients in northeastern Thailand. The knockdown of HMGN3 in CCA cells significantly impaired the oncogenic properties of colony formation, migration, and invasion. HMGN3 inhibited the expression of and blocked the intracellular polarities of epithelial regulator genes, such as the CDH1/E-cadherin and TJAP1 genes in CCA cells. A chromatin immunoprecipitation sequencing analysis revealed that HMGN3 required the transcription factor SNAI2 to bind to and repress the expression of epithelial regulator genes, at least in part, due to histone deacetylases (HDACs), the pharmacological inhibition of which reactivated these epithelial regulators in CCA, leading to impairing the cell migration capacity. Therefore, the overexpression of HMGN3 represses the transcription of and blocks the polarities of epithelial regulators in CCA cells in a manner that is dependent on the SNAI2 gene and HDACs.

    DOI: 10.1096/fj.202200386R

    PubMed

    researchmap

  • ATP citrate lyase controls hematopoietic stem cell fate and supports bone marrow regeneration. 査読 国際誌

    Terumasa Umemoto, Alban Johansson, Shah Adil Ishtiyaq Ahmad, Michihiro Hashimoto, Sho Kubota, Kenta Kikuchi, Haruki Odaka, Takumi Era, Daisuke Kurotaki, Goro Sashida, Toshio Suda

    The EMBO journal   41 ( 8 )   e109463   2022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)  

    In order to support bone marrow regeneration after myeloablation, hematopoietic stem cells (HSCs) actively divide to provide both stem and progenitor cells. However, the mechanisms regulating HSC function and cell fate choice during hematopoietic recovery remain unclear. We herein provide novel insights into HSC regulation during regeneration by focusing on mitochondrial metabolism and ATP citrate lyase (ACLY). After 5-fluorouracil-induced myeloablation, HSCs highly expressing endothelial protein C receptor (EPCRhigh ) were enriched within the stem cell fraction at the expense of more proliferative EPCRLow HSCs. These EPCRHigh HSCs were initially more primitive than EPCRLow HSCs and enabled stem cell expansion by enhancing histone acetylation, due to increased activity of ACLY in the early phase of hematopoietic regeneration. In the late phase of recovery, HSCs enhanced differentiation potential by increasing the accessibility of cis-regulatory elements in progenitor cell-related genes, such as CD48. In conditions of reduced mitochondrial metabolism and ACLY activity, these HSCs maintained stem cell phenotypes, while ACLY-dependent histone acetylation promoted differentiation into CD48+ progenitor cells. Collectively, these results indicate that the dynamic control of ACLY-dependent metabolism and epigenetic alterations is essential for HSC regulation during hematopoietic regeneration.

    DOI: 10.15252/embj.2021109463

    PubMed

    researchmap

  • RUNX1-ETO (RUNX1-RUNX1T1) induces myeloid leukemia in mice in an age-dependent manner. 査読 国際誌

    Mohamed Gaber Abdallah, Akiko Niibori-Nambu, Mariko Morii, Takako Yokomizo, Tomomasa Yokomizo, Takako Ideue, Sho Kubota, Vania Swee Imm Teoh, Michelle Meng Huang Mok, Chelsia Qiuxia Wang, Abdellah Ali Omar, Kenji Tokunaga, Eisaku Iwanaga, Masao Matsuoka, Norio Asou, Naomi Nakagata, Kimi Araki, Mabrouk AboElenin, Sayed Hamada Madboly, Goro Sashida, Motomi Osato

    Leukemia   35 ( 10 )   2983 - 2988   2021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)  

    DOI: 10.1038/s41375-021-01268-4

    PubMed

    researchmap

  • Inflammation-driven senescence-associated secretory phenotype in cancer-associated fibroblasts enhances peritoneal dissemination. 査読 国際誌

    Tadahito Yasuda, Mayu Koiwa, Atsuko Yonemura, Keisuke Miyake, Ryusho Kariya, Sho Kubota, Takako Yokomizo-Nakano, Noriko Yasuda-Yoshihara, Tomoyuki Uchihara, Rumi Itoyama, Luke Bu, Lingfeng Fu, Kota Arima, Daisuke Izumi, Shiro Iwagami, Kojiro Eto, Masaaki Iwatsuki, Yoshifumi Baba, Naoya Yoshida, Hiroto Ohguchi, Seiji Okada, Keisuke Matsusaki, Goro Sashida, Akiko Takahashi, Patrick Tan, Hideo Baba, Takatsugu Ishimoto

    Cell reports   34 ( 8 )   108779 - 108779   2021年2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 the tumor microenvironment, senescent non-malignant cells, including cancer-associated fibroblasts (CAFs), exhibit a secretory profile under stress conditions; this senescence-associated secretory phenotype (SASP) leads to cancer progression and chemoresistance. However, the role of senescent CAFs in metastatic lesions and the molecular mechanism of inflammation-related SASP induction are not well understood. We show that pro-inflammatory cytokine-driven EZH2 downregulation maintains the SASP by demethylating H3K27me3 marks in CAFs and enhances peritoneal tumor formation of gastric cancer (GC) through JAK/STAT3 signaling in a mouse model. A JAK/STAT3 inhibitor blocks the increase in GC cell viability induced by senescent CAFs and peritoneal tumor formation. Single-cell mass cytometry revealed that fibroblasts exist in the ascites of GC patients with peritoneal dissemination, and the fibroblast population shows p16 expression and SASP factors at high levels. These findings provide insights into the inflammation-related SASP maintenance by histone modification and the role of senescent CAFs in GC peritoneal dissemination.

    DOI: 10.1016/j.celrep.2021.108779

    PubMed

    researchmap

  • Overexpression of Hmga2 activates Igf2bp2 and remodels transcriptional program of Tet2-deficient stem cells in myeloid transformation. 査読 国際誌

    Jie Bai, Takako Yokomizo-Nakano, Sho Kubota, Yuqi Sun, Akinori Kanai, Mihoko Iimori, Hironori Harada, Atsushi Iwama, Goro Sashida

    Oncogene   40 ( 8 )   1531 - 1541   2021年2月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    High Mobility Group AT-hook 2 (HMGA2) is a chromatin modifier and its overexpression has been found in patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Level of Hmga2 expression is fine-tuned by Lin28b-Let-7 axis and Polycomb Repressive Complex 2, in which deletion of Ezh2 leads to activation of Hmga2 expression in hematopoietic stem cells. To elucidate the mechanisms by which the overexpression of HMGA2 helps transformation of stem cells harboring a driver mutation of TET2, we generated an Hmga2-expressing Tet2-deficient mouse model showing the progressive phenotypes of MDS and AML. The overexpression of Hmga2 remodeled the transcriptional program of Tet2-deficient stem and progenitor cells, leading to the impaired differentiation of myeloid cells. Furthermore, Hmga2 was bound to a proximal region of Igf2bp2 oncogene, and activated its transcription, leading to enhancing self-renewal of Tet2-deficient stem cells that was suppressed by inhibition of the DNA binding of Hmga2. These combinatory effects on the transcriptional program and cellular function were not redundant to those in Tet2-deficient cells. The present results elucidate that Hmga2 targets key oncogenic pathways during the transformation and highlight the Hmga2-Igf2bp2 axis as a potential target for therapeutic intervention.

    DOI: 10.1038/s41388-020-01629-w

    PubMed

    researchmap

  • Calreticulin haploinsufficiency augments stem cell activity and is required for onset of myeloproliferative neoplasms in mice. 査読 国際誌

    Kotaro Shide, Takuro Kameda, Ayako Kamiunten, Yoshinori Ozono, Yuki Tahira, Takako Yokomizo-Nakano, Sho Kubota, Masaya Ono, Kazuhiko Ikeda, Masaaki Sekine, Keiichi Akizuki, Kenichi Nakamura, Tomonori Hidaka, Yoko Kubuki, Hisayoshi Iwakiri, Satoru Hasuike, Kenji Nagata, Goro Sashida, Kazuya Shimoda

    Blood   136 ( 1 )   106 - 118   2020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)  

    Mutations in JAK2, myeloproliferative leukemia virus (MPL), or calreticulin (CALR) occur in hematopoietic stem cells (HSCs) and are detected in more than 80% of patients with myeloproliferative neoplasms (MPNs). They are thought to play a driver role in MPN pathogenesis via autosomal activation of the JAK-STAT signaling cascade. Mutant CALR binds to MPL, activates downstream MPL signaling cascades, and induces essential thrombocythemia in mice. However, embryonic lethality of Calr-deficient mice precludes determination of a role for CALR in hematopoiesis. To clarify the role of CALR in normal hematopoiesis and MPN pathogenesis, we generated hematopoietic cell-specific Calr-deficient mice. CALR deficiency had little effect on the leukocyte count, hemoglobin levels, or platelet count in peripheral blood. However, Calr-deficient mice showed some hematopoietic properties of MPN, including decreased erythropoiesis and increased myeloid progenitor cells in the bone marrow and extramedullary hematopoiesis in the spleen. Transplantation experiments revealed that Calr haploinsufficiency promoted the self-renewal capacity of HSCs. We generated CALRdel52 mutant transgenic mice with Calr haploinsufficiency as a model that mimics human MPN patients and found that Calr haploinsufficiency restored the self-renewal capacity of HSCs damaged by CALR mutations. Only recipient mice transplanted with Lineage-Sca1+c-kit+ cells harboring both CALR mutation and Calr haploinsufficiency developed MPN in competitive conditions, showing that CALR haploinsufficiency was necessary for the onset of CALR-mutated MPNs.

    DOI: 10.1182/blood.2019003358

    PubMed

    researchmap

  • Low prevalence of the BCR-ABL1 fusion gene in a normal population in southern Sarawak. 査読

    Jew Win Kuan, Anselm Ting Su, Siow Phing Tay, Isabel Lim Fong, Sho Kubota, Lela Su'ut, Motomi Osato, Goro Sashida

    International journal of hematology   111 ( 2 )   217 - 224   2020年2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 BCR-ABL1 fusion gene is the driver mutation of Philadelphia chromosome-positive chronic myeloid leukemia (CML). Its expression level in CML patients is monitored by a real-time quantitative polymerase chain reaction defined by the International Scale (qPCRIS). BCR-ABL1 has also been found in asymptomatic normal individuals using a non-qPCRIS method. In the present study, we examined the prevalence of BCR-ABL1 in a normal population in southern Sarawak by performing qPCRIS for BCR-ABL1 with ABL1 as an internal control on total white blood cells, using an unbiased sampling method. While 146 of 190 (76.8%) or 102 of 190 (53.7%) samples showed sufficient amplification of the ABL1 gene at > 20,000 or > 100,000 copy numbers, respectively, in qPCRIS, one of the 190 samples showed amplification of BCR-ABL1 with positive qPCRIS of 0.0023% and 0.0032% in two independent experiments, the sequence of which was the BCR-ABL1 e13a2 transcript. Thus, we herein demonstrated that the BCR-ABL1 fusion gene is expected to be present in approximately 0.5-1% of normal individuals in southern Sarawak.

    DOI: 10.1007/s12185-019-02768-x

    PubMed

    researchmap

  • [Enhancer hijacking via disease-specific chromosomal translocation in blastic plasmacytoid dendritic cell neoplasm].

    Sho Kubota

    臨床血液   2020年

     詳細を見る

    記述言語:日本語  

    DOI: 10.11406/rinketsu.61.47

    researchmap

  • Ascl1-induced Wnt11 regulates neuroendocrine differentiation, cell proliferation, and E-cadherin expression in small-cell lung cancer and Wnt11 regulates small-cell lung cancer biology. 査読 国際誌

    Yuki Tenjin, Shinji Kudoh, Sho Kubota, Tatsuya Yamada, Akira Matsuo, Younosuke Sato, Takaya Ichimura, Hirotsugu Kohrogi, Goro Sashida, Takuro Sakagami, Takaaki Ito

    Laboratory investigation; a journal of technical methods and pathology   99 ( 11 )   1622 - 1635   2019年11月

     詳細を見る

    記述言語:英語  

    The involvement of Wnt signaling in human lung cancer remains unclear. This study investigated the role of Wnt11 in neuroendocrine (NE) differentiation, cell proliferation, and epithelial-to-mesenchymal transition (EMT) in human small-cell lung cancer (SCLC). Immunohistochemical staining of resected specimens showed that Wnt11 was expressed at higher levels in SCLCs than in non-SCLCs; 58.8% of SCLC, 5.2% of adenocarcinoma (ADC), and 23.5% of squamous cell carcinoma tissues stained positive for Wnt11. A positive relationship was observed between Achaete-scute complex homolog 1 (Ascl1) and Wnt11 expression in SCLC cell lines, and this was supported by transcriptome data from SCLC tissue. The expression of Wnt11 and some NE markers increased after the transfection of ASCL1 into the A549 ADC cell line. Knockdown of Ascl1 downregulated Wnt11 expression in SCLC cell lines. Ascl1 regulated Wnt11 expression via lysine H3K27 acetylation at the enhancer region of the WNT11 gene. Wnt11 controlled NE differentiation, cell proliferation, and E-cadherin expression under the regulation of Ascl1 in SCLC cell lines. The phosphorylation of AKT and p38 mitogen-activated protein kinase markedly increased after transfection of WNT11 into the SBC3 SCLC cell line, which suggests that Wnt11 promotes cell proliferation in SCLC cell lines. Ascl1 plays an important role in regulating the Wnt signaling pathway and is one of the driver molecules of Wnt11 in human SCLC. Ascl1 and Wnt11 may employ a cooperative mechanism to control the biology of SCLC. The present results indicate the therapeutic potential of targeting the Ascl1-Wnt11 signaling axis and support the clinical utility of Wnt11 as a biological marker in SCLC.

    DOI: 10.1038/s41374-019-0277-y

    PubMed

    researchmap

  • Role for tyrosine phosphorylation of SUV39H1 histone methyltransferase in enhanced trimethylation of histone H3K9 via neuregulin-1/ErbB4 nuclear signaling 査読

    Nakajo, H., Ishibashi, K., Aoyama, K., Kubota, S., Hasegawa, H., Yamaguchi, N., Yamaguchi, N.

    Biochemical and Biophysical Research Communications   511 ( 4 )   765 - 771   2019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)  

    Owner : NLM<br />
    Status : Publisher<br />
    PubModel : Print-Electronic<br />
    Language : eng<br />
    Pagination :

    DOI: 10.1016/j.bbrc.2019.02.130

    Scopus

    PubMed

    researchmap

  • Desuppression of TGF-β signaling via nuclear c-Abl-mediated phosphorylation of TIF1γ/TRIM33 at Tyr-524, -610, and -1048 査読

    Ryuzaburo Yuki, Takashi Tatewaki, Noritaka Yamaguchi, Kazumasa Aoyama, Takuya Honda, Sho Kubota, Mariko Morii, Ichiro Manabe, Takahisa Kuga, Takeshi Tomonaga, Naoto Yamaguchi

    Oncogene   38 ( 5 )   637 - 655   2018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:Springer Science and Business Media LLC  

    DOI: 10.1038/s41388-018-0481-z

    PubMed

    researchmap

    その他リンク: http://www.nature.com/articles/s41388-018-0481-z.pdf

  • Ezh2 loss propagates hypermethylation at T cell differentiation-regulating genes to promote leukemic transformation. 査読 国際誌

    Changshan Wang, Motohiko Oshima, Daisuke Sato, Hirotaka Matsui, Sho Kubota, Kazumasa Aoyama, Yaeko Nakajima-Takagi, Shuhei Koide, Jun Matsubayashi, Makiko Mochizuki-Kashio, Takako Nakano-Yokomizo, Jie Bai, Toshitaka Nagao, Akinori Kanai, Atsushi Iwama, Goro Sashida

    The Journal of clinical investigation   128 ( 9 )   3872 - 3886   2018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)  

    Early T cell precursor acute lymphoblastic leukemia (ETP-ALL) is a new pathological entity with poor outcomes in T cell ALL (T-ALL) that is characterized by a high incidence of loss-of-function mutations in polycomb repressive complex 2 (PRC2) genes. We generated a mouse model of ETP-ALL by deleting Ezh2, one of the PRC2 genes, in p53-null hematopoietic cells. The loss of Ezh2 in p53-null hematopoietic cells impeded the differentiation of ETPs and eventually induced ETP-ALL-like disease in mice, indicating that PRC2 functions as a bona fide tumor suppressor in ETPs. A large portion of PRC2 target genes acquired DNA hypermethylation of their promoters following reductions in H3K27me3 levels upon the loss of Ezh2, which included pivotal T cell differentiation-regulating genes. The reactivation of a set of regulators by a DNA-demethylating agent, but not the transduction of single regulator genes, effectively induced the differentiation of ETP-ALL cells. Thus, PRC2 protects key T cell developmental regulators from DNA hypermethylation in order to keep them primed for activation upon subsequent differentiation phases, while its insufficiency predisposes ETPs to leukemic transformation. These results revealed a previously unrecognized epigenetic switch in response to PRC2 dysfunction and provide the basis for specific rational epigenetic therapy for ETP-ALL with PRC2 insufficiency.

    DOI: 10.1172/JCI94645

    PubMed

    researchmap

  • c-Abl-mediated Tyrosine Phosphorylation of JunB is Required for Adriamycin-induced Expression of p21. 査読

    Noritaka Yamaguchi, Ryuzaburo Yuki, Sho Kubota, Kazumasa Aoyama, Takahisa Kuga, Yuuki Hashimoto, Takeshi Tomonaga, Naoto Yamaguchi

    The Biochemical journal   471 ( 1 )   67 - 77   2015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)  

    DOI: 10.1042/BJ20150372

    Web of Science

    PubMed

    researchmap

  • Src family kinases maintain the balance between replication stress and the replication checkpoint 査読

    Takahito Miura, Yasunori Fukumoto, Mariko Morii, Takuya Honda, Noritaka Yamaguchi, Yuji Nakayama, Naoto Yamaguchi

    Cell Biology International   40 ( 1 )   16 - 26   2015年8月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Wiley  

    DOI: 10.1002/cbin.10517

    PubMed

    researchmap

  • Imatinib inhibits inactivation of the ATM/ATR signaling pathway and recovery from adriamycin/doxorubicin-induced DNA damage checkpoint arrest 査読

    Mariko Morii, Yasunori Fukumoto, Sho Kubota, Noritaka Yamaguchi, Yuji Nakayama, Naoto Yamaguchi

    CELL BIOLOGY INTERNATIONAL   39 ( 8 )   923 - 932   2015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:WILEY-BLACKWELL  

    The DNA damage checkpoint arrests cell cycle progression to allow time for DNA repair. After completion of DNA repair, checkpoint activation is terminated, and cell cycle progression is resumed in a process called checkpoint recovery. The activation of the checkpoint has been studied in depth, but little is known about recovery from the DNA damage checkpoint. Recently we showed that Src family kinases promote recovery from the G2 DNA damage checkpoint. Here we show that imatinib inhibits inactivation of ATM/ATR signaling pathway to suppress recovery from Adriamycin/doxorubicin-induced DNA damage checkpoint arrest. Imatinib and pazopanib, two distinct inhibitors of PDGFR/c-Kit family kinases, delayed recovery from checkpoint arrest and inhibited the subsequent S-G2-M transition after Adriamycin exposure. By contrast, imatinib and pazopanib did not delay the recovery from checkpoint arrest in the presence of an ATM/ATR inhibitor caffeine. Consistently, imatinib induced a persistent activation of ATR-Chk1 signaling. By the way, the maintenance of G2 checkpoint arrest is largely dependent on ATR-Chk1 signaling. However, unlike Src inhibition, imatinib did not delay the recovery from checkpoint arrest in the presence of an ATM inhibitor KU-55933. Furthermore, imatinib induced a persistent activation of ATM-KAP1 signaling, and a possible involvement of imatinib in an ATM-dependent DNA damage response is suggested. These results reveal that imatinib inhibits recovery from Adriamycin-induced DNA damage checkpoint arrest in an ATM/ATR-dependent manner and raise the possibility that imatinib may inhibit resumption of tumor proliferation after chemo- and radiotherapy.

    DOI: 10.1002/cbin.10460

    Web of Science

    PubMed

    researchmap

  • Overexpression of Zinc-Finger Protein 777 (ZNF777) Inhibits Proliferation at Low Cell Density Through Down-Regulation of FAM129A. 査読

    Ryuzaburo Yuki, Kazumasa Aoyama, Sho Kubota, Noritaka Yamaguchi, Shoichi Kubota, Hitomi Hasegawa, Mariko Morii, Xiayu Huang, Kang Liu, Roy Williams, Michiko N. Fukuda, Naoto Yamaguchi

    Journal of cellular biochemistry   116 ( 6 )   954 - 968   2015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)  

    DOI: 10.1002/jcb.25046

    Web of Science

    PubMed

    researchmap

  • c-Abl induces stabilization of histone deacetylase 1 (HDAC1) in a kinase activity-dependent manner 査読

    Kazumasa Aoyama, Noritaka Yamaguchi, Ryuzaburo Yuki, Mariko Morii, Sho Kubota, Kensuke Hirata, Kohei Abe, Takuya Honda, Takahisa Kuga, Yuuki Hashimoto, Takeshi Tomonaga, Naoto Yamaguchi

    39 ( 4 )   446 - 456   2015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)  

    DOI: 10.1002/cbin.10413

    Web of Science

    PubMed

    researchmap

  • Lyn tyrosine kinase promotes silencing of ATM-dependent checkpoint signaling during recovery from DNA double-strand breaks 査読

    Yasunori Fukumoto, Kazumasa Kuki, Mariko Morii, Takahito Miura, Takuya Honda, Kenichi Ishibashi, Hitomi Hasegawa, Sho Kubota, Yudai Ide, Noritaka Yamaguchi, Yuji Nakayama, Naoto Yamaguchi

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   452 ( 3 )   542 - 547   2014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)   出版者・発行元:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    DNA damage activates the DNA damage checkpoint and the DNA repair machinery. After initial activation of DNA damage responses, cells recover to their original states through completion of DNA repair and termination of checkpoint signaling. Currently, little is known about the process by which cells recover from the DNA damage checkpoint, a process called checkpoint recovery. Here, we show that Src family kinases promote inactivation of ataxia telangiectasia mutated (ATM)-dependent checkpoint signaling during recovery from DNA double-strand breaks. Inhibition of Src activity increased ATM-dependent phosphorylation of Chk2 and Kap1. Src inhibition increased ATM signaling both in G2 phase and during asynchronous growth. shRNA knockdown of Lyn increased ATM signaling. Src-dependent nuclear tyrosine phosphorylation suppressed ATM-mediated Kap1 phosphorylation. These results suggest that Src family kinases are involved in upstream signaling that leads to inactivation of the ATM-dependent DNA damage checkpoint. (C)2014 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2014.08.113

    Web of Science

    PubMed

    researchmap

  • v-Src inhibits the interaction between Rad17 and Rad9 and induces replication fork collapse 査読

    Yasunori Fukumoto, Takahito Miura, Mariko Morii, Sho Kubota, Takuya Honda, Shoichi Kubota, Takao Morinaga, Noritaka Yamaguchi, Yuji Nakayama, Naoto Yamaguchi

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   450 ( 1 )   885 - 890   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:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    ATR-dependent DNA damage checkpoint is crucial to maintain genomic stability. Recently, we showed that Src family kinases suppress ATR-dependent checkpoint signaling in termination of DNA damage checkpoint. However, the precise molecular mechanism is unclear. Therefore, we examined the role of oncogenic v-Src on ATR-Chk1 signaling. We show that v-Src suppresses thymidine-induced Chk1 phosphorylation and induces replication fork collapse. v-Src inhibits interaction between Rad17 and Rad9 in chromatin fraction. By contrast, v-Src does not inhibit RPA32 phosphorylation, ATR autophosphorylation, or TopBP1-Rad9 interaction. These data suggest that v-Src attenuates ATR-Chk1 signaling through the inhibition of Radl7-Rad9 interaction. (C) 2014 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2014.06.078

    Web of Science

    PubMed

    researchmap

  • Src Family Kinases Promote Silencing of ATR-Chk1 Signaling in Termination of DNA Damage Checkpoint 査読

    Yasunori Fukumoto, Mariko Morii, Takahito Miura, Sho Kubota, Kenichi Ishibashi, Takuya Honda, Aya Okamoto, Noritaka Yamaguchi, Atsushi Iwama, Yuji Nakayama, Naoto Yamaguchi

    JOURNAL OF BIOLOGICAL CHEMISTRY   289 ( 18 )   12313 - 12329   2014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  

    Background: Once DNA repair is completed, the DNA damage checkpoint is terminated, and the cell cycle is resumed. Results: Src inhibition induced a delay in G(2) checkpoint recovery and persistent ATR-Chk1 activation. Conclusion: Src inhibits ATR signaling to promote recovery from G(2) checkpoint arrest. Significance: Src sends a termination signal between the completion of DNA repair and the initiation of checkpoint termination.
    The DNA damage checkpoint arrests cell cycle progression to allow time for repair. Once DNA repair is completed, checkpoint signaling is terminated. Currently little is known about the mechanism by which checkpoint signaling is terminated, and the disappearance of DNA lesions is considered to induce the end of checkpoint signaling; however, here we show that the termination of checkpoint signaling is an active process promoted by Src family tyrosine kinases. Inhibition of Src activity delays recovery from the G(2) phase DNA damage checkpoint following DNA repair. Src activity is required for the termination of checkpoint signaling, and inhibition of Src activity induces persistent activation of ataxia telangiectasia mutated (ATM)- and Rad3-related (ATR) and Chk1 kinases. Src-dependent nuclear protein tyrosine phosphorylation and v-Src expression suppress the ATR-mediated Chk1 and Rad17 phosphorylation induced by DNA double strand breaks or DNA replication stress. Thus, Src family kinases promote checkpoint recovery through termination of ATR- and Chk1-dependent G(2) DNA damage checkpoint. These results suggest a model according to which Src family kinases send a termination signal between the completion of DNA repair and the initiation of checkpoint termination.

    DOI: 10.1074/jbc.M113.533752

    Web of Science

    PubMed

    researchmap

  • Activation of the Prereplication Complex Is Blocked by Mimosine through Reactive Oxygen Species-activated Ataxia Telangiectasia Mutated (ATM) Protein without DNA Damage 査読

    Shoichi Kubota, Yasunori Fukumoto, Kenichi Ishibashi, Shuhei Soeda, Sho Kubota, Ryuzaburo Yuki, Yuji Nakayama, Kazumasa Aoyama, Noritaka Yamaguchi, Naoto Yamaguchi

    JOURNAL OF BIOLOGICAL CHEMISTRY   289 ( 9 )   5730 - 5746   2014年2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: Mimosine is a cell synchronization reagent used for arresting cells in late G(1) and S phases. Results: Replication fork assembly is reversibly blocked by ATM activation through mimosine-generated reactive oxygen species. Conclusion: Mimosine induces cell cycle arrest strictly at the G(1)-S phase boundary, which prevents replication fork stalling-induced DNA damage. Significance: These findings provide a novel mechanism of the mimosine-induced G(1) checkpoint.
    Mimosine is an effective cell synchronization reagent used for arresting cells in late G(1) phase. However, the mechanism underlying mimosine-induced G(1) cell cycle arrest remains unclear. Using highly synchronous cell populations, we show here that mimosine blocks S phase entry through ATM activation. HeLa S3 cells are exposed to thymidine for 15 h, released for 9 h by washing out the thymidine, and subsequently treated with 1 mm mimosine for a further 15 h (thymidine mimosine). In contrast to thymidine-induced S phase arrest, mimosine treatment synchronizes &gt;90% of cells at the G(1)-S phase boundary by inhibiting the transition of the prereplication complex to the preinitiation complex. Mimosine treatment activates ataxia telangiectasia mutated (ATM)/ataxia telangiectasia and Rad3-related (ATR)-mediated checkpoint signaling without inducing DNA damage. Inhibition of ATM activity is found to induce mimosine-arrested cells to enter S phase. In addition, ATM activation by mimosine treatment is mediated by reactive oxygen species (ROS). These results suggest that, upon mimosine treatment, ATM blocks S phase entry in response to ROS, which prevents replication fork stalling-induced DNA damage.

    DOI: 10.1074/jbc.M113.546655

    Web of Science

    PubMed

    researchmap

  • Formation of long and winding nuclear F-actin bundles by nuclear c-Abl tyrosine kinase 査読

    Kazumasa Aoyama, Ryuzaburo Yuki, Yasuyoshi Horiike, Sho Kubota, Noritaka Yamaguchi, Mariko Morii, Kenichi Ishibashi, Yuji Nakayama, Takahisa Kuga, Yuuki Hashimoto, Takeshi Tomonaga, Naoto Yamaguchi

    Experimental Cell Research   319 ( 20 )   3251 - 3268   2013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:Elsevier BV  

    The non-receptor-type tyrosine kinase c-Abl is involved in actin dynamics in the cytoplasm. Having three nuclear localization signals (NLSs) and one nuclear export signal, c-Abl shuttles between the nucleus and the cytoplasm. Although monomeric actin and filamentous actin (F-actin) are present in the nucleus, little is known about the relationship between c-Abl and nuclear actin dynamics. Here, we show that nuclear-localized c-Abl induces nuclear F-actin formation. Adriamycin-induced DNA damage together with leptomycin B treatment accumulates c-Abl into the nucleus and increases the levels of nuclear F-actin. Treatment of c-Abl-knockdown cells with Adriamycin and leptomycin B barely increases the nuclear F-actin levels. Expression of nuclear-targeted c-Abl (NLS-c-Abl) increases the levels of nuclear F-actin even without Adriamycin, and the increased levels of nuclear F-actin are not inhibited by inactivation of Abl kinase activity. Intriguingly, expression of NLS-c-Abl induces the formation of long and winding bundles of F-actin within the nucleus in a c-Abl kinase activity-dependent manner. Furthermore, NLS-c-AblδC, which lacks the actin-binding domain but has the full tyrosine kinase activity, is incapable of forming nuclear F-actin and in particular long and winding nuclear F-actin bundles. These results suggest that nuclear c-Abl plays critical roles in actin dynamics within the nucleus. © 2013 Elsevier Inc.

    DOI: 10.1016/j.yexcr.2013.09.003

    Scopus

    PubMed

    researchmap

  • Nuclear ErbB4 signaling through H3K9me3 is antagonized by EGFR-activated c-Src 査読

    Kenichi Ishibashi, Yasunori Fukumoto, Hitomi Hasegawa, Kohei Abe, Shoichi Kubota, Kazumasa Aoyama, Sho Kubota, Yuji Nakayama, Naoto Yamaguchi

    JOURNAL OF CELL SCIENCE   126 ( 2 )   625 - 637   2013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:COMPANY OF BIOLOGISTS LTD  

    The ErbB family of receptor tyrosine kinases comprises four members: epidermal growth factor receptor (EGFR)/ErbB1, HER2/ErbB2, ErbB3 and ErbB4, and plays roles in signal transduction at the plasma membrane upon ligand stimulation. Stimulation with neuregulin-1 (NRG-1) cleaves ErbB4 and releases the ErbB4 intracellular domain (4ICD) that translocates into the nucleus to control gene expression. However, little is known about the regulation of 4ICD nuclear signaling through tyrosine phosphorylation. We show here that 4ICD nuclear signaling is antagonized by EGF-induced c-Src activation through EGFR. Generation of 4ICD by NRG-1 leads to increased levels of trimethylated histone H3 on lysine 9 (H3K9me3) in a manner dependent on the nuclear accumulation of 4ICD and its tyrosine kinase activity. Once EGF activates c-Src downstream of EGFR concomitantly with NRG-1-induced ErbB4 activation, c-Src associates with phospho-Tyr950 and phospho-Tyr1056 on 4ICD, thereby decreasing nuclear accumulation of 4ICD and inhibiting an increase of H3K9me3 levels. Moreover, 4ICD-induced transcriptional repression of the human telomerase reverse transcriptase (hTERT) is inhibited by EGF-EGFR-Src signaling. Thus, our findings reveal c-Src-mediated inhibitory regulation of ErbB4 nuclear signaling upon EGFR activation.

    DOI: 10.1242/jcs.116277

    Web of Science

    PubMed

    researchmap

  • Nuclear c-Abl-mediated tyrosine phosphorylation induces chromatin structural changes through histone modifications that include H4K16 hypoacetylation 査読

    Kazumasa Aoyama, Yasunori Fukumoto, Kenichi Ishibashi, Sho Kubota, Takao Morinaga, Yasuyoshi Horiike, Ryuzaburo Yuki, Akinori Takahashi, Yuji Nakayama, Naoto Yamaguchi

    EXPERIMENTAL CELL RESEARCH   317 ( 20 )   2874 - 2903   2011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:ELSEVIER INC  

    c-Abl tyrosine kinase, which is ubiquitously expressed, has three nuclear localization signals and one nuclear export signal and can shuttle between the nucleus and the cytoplasm. c-Abl plays important roles in cell proliferation, adhesion, migration, and apoptosis. Recently, we developed a pixel imaging method for quantitating the level of chromatin structural changes and showed that nuclear Src-family tyrosine kinases are involved in chromatin structural changes upon growth factor stimulation. Using this method, we show here that nuclear c-Abl induces chromatin structural changes in a manner dependent on the tyrosine kinase activity. Expression of nuclear-targeted c-Abl drastically increases the levels of chromatin structural changes, compared with that of c-Abl. Intriguingly, nuclear-targeted c-Abl induces heterochromatic profiles of histone methylation and acetylation, including hypoacetylation of histone H4 acetylated on lysine 16 (H4K16Ac). The level of heterochromatic histone modifications correlates with that of chromatin structural changes. Adriamycin-induced DNA damage stimulates translocation of c-Abl into the nucleus and induces chromatin structural changes together with H4K16 hypoacetylation. Treatment with trichostatin A, a histone deacetylase inhibitor, blocks chromatin structural changes but not nuclear tyrosine phosphorylation by c-Abl. These results suggest that nuclear c-Abl plays an important role in chromatin dynamics through nuclear tyrosine phosphorylation-induced heterochromatic histone modifications. (C) 2011 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.yexcr.2011.09.013

    Web of Science

    PubMed

    researchmap

▼全件表示

MISC

  • Tet2欠損によるHmga2機能制御と骨髄異形成症候群発症の分子基盤(Tet2 loss reshapes Hmga2 binding regions to promote the development of myelodysplastic syndrome)

    指田 吾郎, 横溝 貴子, 久保田 翔, 原田 浩徳, 大里 元美, 岩間 厚志

    日本癌学会総会記事   78回   E - 2077   2019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

  • RUNX3発現による骨髄異形成症候群発症の分子基盤解明(RUNX3 over-expression impedes RUNX1 function and promotes the development of myelodysplastic syndrome)

    横溝 貴子, 久保田 翔, 岩間 厚志, 原田 浩徳, 大里 元美, 指田 吾郎

    日本癌学会総会記事   78回   P - 2224   2019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

  • Lineage-Specific RUNX2 Super-Enhancer Activates MYC Via Translocation (6;8) to Promote the Development of Blastic Plasmacytoid Dendritic Cell Neoplasm

    Sho Kubota, Kenji Tokunaga, Tomohiro Umezu, Takako Yokomizo, Motohiko Oshima, Eisaku Iwanaga, Norio Asou, Atsushi Iwama, Kazuma Ohyashiki, Motomi Osato, Goro Sashida

    BLOOD (International Conference)   132   2018年11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:AMER SOC HEMATOLOGY  

    0

    DOI: 10.1182/blood-2018-99-110333

    Web of Science

    researchmap

  • RUNX2スーパーエンハンサーによるMYCの活性化を介したBPDCNの発症メカニズムの解析(Lineage-specific RUNX2 super-enhancer activates MYC via a chromosomal translocation and promotes the BPDCN)

    久保田 翔, 梅津 知宏, 岩間 厚志, 大屋敷 一馬, 大里 元美, 指田 吾郎

    日本癌学会総会記事   77回   1966 - 1966   2018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

受賞

  • 熊本大学研究業績表彰

    2020年  

     詳細を見る

  • THE 9th JSH International Symposium 最優秀ポスター賞

    2018年  

     詳細を見る

  • 日本血液学会奨励賞

    2018年  

     詳細を見る

  • 第134年会日本薬学会 優秀発表賞

    2014年  

     詳細を見る

  • 第54回日本薬学会関東支部大会 優秀発表賞

    2010年  

     詳細を見る

共同研究・競争的資金等の研究

  • MLL融合白血病発症を受容する胎児造血幹細胞形質の解析

    研究課題/領域番号:21K08421  2021年04月 - 2024年03月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    久保田 翔

      詳細を見る

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

    MLL-AF9を発達時期依存的に誘導する為に、Rosa-LSL-MLL-AF9-IRES-GFPノックインマウスを作製した。eR1-CreERT2マウスと交配し、造血幹前駆細胞特異的にタモキシフェンによって、MLL-AF9を誘導可能なマウスモデル(MLL-AF9-KI)を作製した。胎児期、小児期、成人期、老年期にMLL-AF9を誘導発現し、生存曲線、末梢血の状態、白血病細胞の増殖などを全血球算定機、フローサイトメトリーを用いて解析を行った。
    既にMLL-AF9を小児期、成人期に誘導した実験はすでに結果を得ている。MLL-AF9を小児期、成人期両方の場合において、マウスはAMLを発症した。しかし、発症速度に大きな違いがあり、成人期に誘導した場合に比較して小児期では早く白血病が発症しており、有意な差がみられた。発症前に2か月の時点で、最終的に異常増殖を行う顆粒球細胞の割合をフローサイトメトリーで比較すると、成人期に比較して小児期で末梢血中の顆粒球の割合が高いことが分かった。胎児期および老年期のマウスについても準備及び白血病の発症への影響の解析を行っていく。また、造血幹前駆細胞のトランスクリプトーム・エピゲノム解析について現在サンプルを準備中である。
    造血幹細胞ストレスによる白血病発症への寄与を検証するため、上記のMLL-AF9-KIマウスに加え、コントロール用のRosa-LSL-YFP-KIマウスおよび小児のMLL融合がん遺伝子による白血病で二次変異として知られているNRasの活性化変異を導入したMLL-AF9-KI;NRasG12Dを準備した。これらのマウスの骨髄を移植したマウスと2回連続移植を行ってから移植したマウスとで白血病の発症速度を比較した。その結果、初回移植のマウスに比較して連続移植したマウスではAMLの発症速度が遅れた。今後この結果の再現性を取得していく。

    researchmap

  • 染色体転座由来スーパーエンハンサー活性化による白血病発症機構解明

    研究課題/領域番号:18K16090  2018年04月 - 2021年03月

    日本学術振興会  科学研究費助成事業  若手研究

    久保田 翔

      詳細を見る

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

    芽球形質細胞様樹状細胞腫瘍(BPDCN)は、pDCに類似した表現形・遺伝子発現様式を持つ急性骨髄性白血病の一亜形である。従来の抗がん剤治療には抵抗性で生命予後は不良であり、その病態は未だ明らかでない。申請者は、BPDCNに特異的に認める染色体転座t(6;8)によって、強力ながん遺伝子であるc-MYCとpDC分化に重要なRUNX2のエンハンサー領域が相互転座していることを見出した。申請者が新たに同定した転座由来がん特異的スーパーエンハンサー活性化によるBPDCNの発症機構を解明した。

    researchmap

  • 白血病幹細胞におけるエピゲノム制御異常の分子基盤解明と治療標的の検索

    研究課題/領域番号:17J02389  2017年04月 - 2020年03月

    日本学術振興会  科学研究費助成事業  特別研究員奨励費

    久保田 翔

      詳細を見る

    配分額:4420000円 ( 直接経費:3400000円 、 間接経費:1020000円 )

    近年、がんは加齢など環境因子に伴うエピゲノム変異に加えて、遺伝子変異や染色体異常などのゲノム変異が付加蓄積した病態としても新たに認識されつつある。ただし、こうした変異の協調作用によるがん発症のメカニズムは未だ明らかでない。こうした状況の中、申請者は“BCR-ABLチロシンキナーゼによるTIF1betaリン酸化を介したエピゲノム制御異常がCML幹細胞を発生・進展させる”仮説を得た。本研究ではこれらのエピゲノムとCMLとの関わりについて明らかにすることを目的とする。
    細胞株を用いた実験では内在性TIF1betaをノックダウンした後に、TIF1betaまたはTIF1beta-3YFリン酸化欠失変異体を導入したヒト白血病細胞株K562を樹立した。この細胞株を用いてFLAG-TIF1betaまたはFLAG-TIF1beta-3YF結合蛋白を精製した後にLC/MS解析によって、TIF1betaチロシンリン酸化特異的なクロマチン結合複合体のタンパク質を同定している。同定した因子の中から新たな治療標的因子としてタンパク質Xを見出し、阻害剤によってK562細胞の増殖が抑えられることを明らかにしている。
    BCR-ABL/TIFbetaKOマウスでは著しくCMLの進展を阻害し、一部のマウスでは白血病幹細胞を含めて白血病が消失した。これらのメカニズムについて明らかにするために、白血病幹細胞画分においてRNA-sequence解析を行った。その結果、BCR-ABLによって転写が変化する白血病遺伝子群の半分がTIF1betaが欠損した状態では変化しなかったことから、TIF1betaの機能変化はBCR-ABLによって誘導される白血病幹細胞の発生に重要であることが明らかになった。これらのことから、TIF1betaが重要な治療ターゲットの一つとなりうることが示唆された。

    researchmap

  • 造血器腫瘍におけるKAP1チロシンリン酸化の機能解析

    研究課題/領域番号:15K19545  2015年04月 - 2018年03月

    日本学術振興会  科学研究費助成事業  若手研究(B)

    久保田 翔, 指田 吾郎

      詳細を見る

    配分額:4030000円 ( 直接経費:3100000円 、 間接経費:930000円 )

    白血病原因遺伝子であるBCR-ABL チロシンキナーゼの活性型変異体は主に細胞質に局在し、過剰なシグナル伝達を介して造血器腫瘍の発症を促進する。BCR-ABL を含むチロシンキナーゼは実際には核内にも移行することが知られているが、核内におけるチロシンリン酸化シグナル伝達の役割は明らかでない。BCR-ABLトランスジェニックマウスとエピジェネティクスレギュレーターのKAP1をノックアウトマウスと交配した。これらのマウスを用いてKAP1のチロシンリン酸化のin vivoにおける正常造血および白血病発症での機能解析を行なった。分子メカニズムの解析ではK562細胞を用いて解析を行なった。

    researchmap

  • 核内チロシンリン酸化シグナルによるクロマチン構造制御メカニズムの解明

    研究課題/領域番号:14J04367  2014年04月 - 2016年03月

    日本学術振興会  科学研究費助成事業  特別研究員奨励費

    久保田 翔

      詳細を見る

    配分額:2170000円 ( 直接経費:1900000円 、 間接経費:270000円 )

    本年度はin vivo, in vitroの両方向からの解析を行なった。in vivoの解析では私が以前、核内チロシンリン酸化基質として同定したKAP1のノックアウトマウスを用いた解析を行なった。具体的には、造血幹細胞の分化を解析する系を利用して調べた。造血幹細胞の増殖分化においてKAP1はおよびチロシンリン酸化シグナルは重要な役割をもっている。KAP1ノックアウトマウスの造血幹細胞をセルソーターを用いて分取し、野生型のKAP1とチロシンリン酸化されない変異体をレトロウィルスベクターのインフェクションによって発現させ、レシピエントマウスに移植し、その分化増殖への影響を解析した。In vitroの解析では、核内チロシン基質群の中でヒストン修飾に関わる分子として A-kinase anchoring protein 8 (AKAP8) に着目した。AKAP8はクロマチンや核マトリクスに結合し、ヒストン脱アセチル化酵素であるhistone deacetylase 3 (HDAC3) などの分子のリクルートに関わっていることが知られている。昨年度の解析からAKAP8はチロシンキナーゼによってチロシンリン酸化されることで、クロマチンや核マトリクスとの結合が弱められることが示唆された。AKAP8のチロシンリン酸化による機能変化を解析するために、私はAKAP8の中でチロシンリン酸化されるチロシン残基をチロシンリン酸化されないフェニルアラニンに置換した変異体の作製を行なった。複数の部位のチロシンを置換した変異体では結合の変化がキャンセル出来た。このことから、AKAP8の結合の変化は一か所のチロシン残基のリン酸化によって制御されているのではなく、複数の部位のチロシンリン酸化によって制御されていることがわかった。

    researchmap

  • 芽球形質細胞様樹状細胞腫瘍モデルを用いた病態基盤解明と新規標的治療の 開発

    新日本先進医療研究財団 研究助成 

      詳細を見る

  • 染色体転座由来スーパーエンハンサー活性化による白血病発症の分子基盤解明

    武田科学振興財団 医学系研究助成 

      詳細を見る

  • HMGA2 によるストレス造血制御と幹細胞運命決定機構の解析

    持田記念医学薬学振興財団 研究助成 

      詳細を見る

  • 染色体転座由来スーパーエンハンサー活性化による白血病発症の分子基盤解明

    武田科学振興財団 医学系継続研究助成 

      詳細を見る

  • 白血病幹細胞におけるクロマチン制御破綻のメカニズム解明と治療標的検索.

    白血病基金 研究助成 

      詳細を見る

▼全件表示