Researcher Database

Researcher Profile and Settings

Master

Affiliation (Master)

  • Faculty of Medicine Physiological Science Biochemistry

Affiliation (Master)

  • Faculty of Medicine Physiological Science Biochemistry

researchmap

Profile and Settings

Profile and Settings

  • Name (Japanese)

    TSUKASA
  • Name (Kana)

    OIKAWA
  • Name

    200901075245308688

Achievement

Research Interests

  • 浸潤   破骨細胞   がん細胞   細胞融合   細胞極性   イノシトールリン脂質   がん   cell biology   

Research Areas

  • Life sciences / Cell biology
  • Life sciences / Pathobiochemistry
  • Life sciences / Medical biochemistry

Research Experience

  • 2014/10 Hokkaido University Assistant Professor
  • 2013/08 - 2014/09 Hokkaido University Graduate School of Medicine Department of Molecular Biology
  • 2009/01 - 2013/07 Keio University School of Medicine
  • 2007/04 - 2008/12 Kobe University Graduate School of Medicine Lipid Biochemistry

Education

  • 2002/04 - 2007/03  The University of Tokyo  Graduate School of Pharmaceutical Sciences
  • 1998/04 - 2002/03  The University of Tokyo  Faculty of Pharmaceutical Sciencees

Published Papers

  • Haruka Handa, Yasuhito Onodera, Tsukasa Oikawa, Shingo Takada, Koji Ueda, Daiki Setoyama, Takashi Yokota, Miwako Yamasaki, Masahiko Watanabe, Yoshizuki Fumoto, Ari Hashimoto, Soichiro Hata, Masaaki Murakami, Hisataka Sabe
    bioRxiv 2024/07/29 
    Mitochondrial functions range from catabolic to anabolic, which are tightly coordinated to meet cellular demands for proliferation and motility. MitoNEET is a mitochondrial outer membrane protein with a CDGSH domain and is involved in mitochondrial function. Epithelial-to-mesenchymal transition (EMT) is the process in which cells lose their epithelial characteristics and acquire mesenchymal traits, such as motility, which is a vital step for organism development and wound-healing. Cellular motility is associated with high ATP consumption owing to lamellipodia formation, which is supported by upregulated oxidative phosphorylation (OXPHOS) capacity. However, how mitoNEET is involved in the regulation of OXPHOS capacity and subsequent cellular motility remains unclear. Here we show that loss of mitoNEET regulation during EMT impairs both OXPHOS enhancement and cell motility in non-transformed NMuMG mouse mammary gland epithelial cells. We found that mitoNEET is downregulated during EMT, and that the aberrant expression of mitoNEET abolishes the upregulation of OXPHOS, leading to the inhibition of cell motility. Furthermore, we found that mitoNEET topology may be crucial for the regulation of the mitochondrial electron transfer chain, suggesting an additional regulatory pathway for OXPHOS capacity. Our results demonstrate that mitochondrial OXPHOS capacity during EMT is partly regulated by the dynamics of the outer membrane protein. We believe that our findings are the first step towards understanding the mechanisms by which mitochondrial outer membrane protein topology affects organelle functions
  • Tsukasa Oikawa, Junya Hasegawa, Haruka Handa, Naomi Ohnishi, Yasuhito Onodera, Ari Hashimoto, Junko Sasaki, Takehiko Sasaki, Koji Ueda, Hisataka Sabe
    Life Science Alliance 7 (9) e202402835  2024/06 [Refereed][Not invited]
  • Tsukasa Oikawa, Junya Hasegawa, Haruka Handa, Naomi Ohnishi, Yasuhito Onodera, Ari Hashimoto, Junko Sasaki, Takehiko Sasaki, Koji Ueda, Hisataka Sabe
    bioRxiv 2023/06/28 [Not refereed][Not invited]
     
    Abstract Histones are key molecules of epigenetic regulation and inheritance, and are thought to be chaperoned and transported into the nucleus appropriately prior to being integrated into nucleosomes. H3.1 histone is predominantly synthesized and enters the nucleus during the G1/S phase of the cell cycle, as a new component of duplicating nucleosomes. Here we found that p53 is necessary to secure the normal behavior and modification of H3.1 in the nucleus during the G1/S phase, in which p53 increases C-terminal domain nuclear envelope phosphatase 1 (CTDNEP1) levels and decreases enhancer of zeste homolog 2 (EZH2) levels in the H3.1 interactome. In the absence of p53, H3.1 molecules tended to be tethered at or near the nuclear envelope (NE), where they were predominantly trimethylated at lysine 27 (H3K27me3) by EZH2, without forming nucleosomes. This accumulation was likely caused by the high affinity of H3.1 towards phosphatidic acid (PA). p53 reduced nuclear PA levels by increasing levels of CTDNEP1, which activates lipin to convert PA into diacylglycerol. Induction of theTMEM255Agene by p53 linked p53 with CTDNEP1, in which TMEM255A stabilized CTDNEP1. We moreover found that the cytosolic H3 chaperone HSC70 attenuates the H3.1-PA interaction, and our molecular imaging analyses suggested that H3.1 molecules may be anchored around the NE after their nuclear entry. Our results expand our knowledge of p53 function in regulation of the nuclear behavior of H3.1 during the G1/S phase, in which p53 may primarily target nuclear PA and EZH2.
  • Hiroaki Kida, Jing-Jing Jiang, Yuichiro Matsui, Ikuko Takahashi, Rie Hasebe, Daisuke Kawamura, Takeshi Endo, Hiroki Shibayama, Makoto Kondoh, Yasuhiko Nishio, Kinya Nishida, Yoshihiro Matsuno, Tsukasa Oikawa, Shimpei Kubota, Shintaro Hojyo, Norimasa Iwasaki, Shigeru Hashimoto, Yuki Tanaka, Masaaki Murakami
    International immunology 2023/01/31 [Refereed][Not invited]
     
    Dupuytren's contracture (DC) is an inflammatory fibrosis characterized by fibroproliferative disorders of the palmar aponeurosis, for which there is no effective treatment. Although several genome-wide association studies have identified risk alleles associated with DC, the functional linkage between these alleles and the pathogenesis remains elusive. We here focused on two SNPs associated with DC, rs16879765 and rs17171229, in secreted frizzled related protein 4 (SFRP4). We investigated the association of SRFP4 with the IL-6 amplifier, which amplifies the production of IL-6, growth factors, and chemokines in non-immune cells and aggravates inflammatory diseases via NF-κB enhancement. Knockdown of SFRP4 suppressed activation of the IL-6 amplifier in vitro and in vivo, whereas the overexpression of SFRP4 induced the activation of NF-κB-mediated transcription activity. Mechanistically, SFRP4 induced NF-κB activation by directly binding to molecules of the ubiquitination SFC complex, such as IkBα and βTrCP, followed by IkBα degradation. Furthermore, SFRP4 expression was significantly increased in fibroblasts derived from DC patients bearing the risk alleles. Consistently, fibroblasts with the risk alleles enhanced activation of the IL-6 amplifier. These findings indicate that the IL-6 amplifier is involved in the pathogenesis of DC, particularly in patients harboring the SFRP4 risk alleles. Therefore, SFRP4 is a potential therapeutic target for various inflammatory diseases and disorders, including DC.
  • Mika Watanabe, Hideyuki Kosumi, Shin-Ichi Osada, Shota Takashima, Yunan Wang, Wataru Nishie, Tsukasa Oikawa, Tomonori Hirose, Hiroshi Shimizu, Ken Natsuga
    Experimental dermatology 30 (1) 62 - 67 2021/01 [Refereed]
     
    Type XVII collagen (COL17) is a transmembrane protein expressed in the basal epidermis. COL17 serves as a niche for epidermal stem cells, and although its reduction has been implicated in altering cell polarity and ageing of the epidermis, it is unknown how COL17 affects epidermal cell polarity. Here, we uncovered COL17 as a binding partner of the aPKC-PAR complex, which is a key regulating factor of cell polarity. Immunoprecipitation-immunoblot assay and protein-protein binding assay revealed that COL17 interacts with aPKC and PAR3. COL17 deficiency or epidermis-specific aPKCλ deletion destabilized PAR3 distribution in the epidermis, while aPKCζ knockout did not. Asymmetrical cell division was pronounced in COL17-null neonatal paw epidermis. These results show that COL17 is pivotal for maintaining epidermal cell polarity. Our study highlights the previously unrecognized role of COL17 in the basal keratinocytes.
  • Shigeru Hashimoto, Shotaro Furukawa, Ari Hashimoto, Akio Tsutaho, Akira Fukao, Yurika Sakamura, Gyanu Parajuli, Yasuhito Onodera, Yutaro Otsuka, Haruka Handa, Tsukasa Oikawa, Soichiro Hata, Yoshihiro Nishikawa, Yusuke Mizukami, Yuzo Kodama, Masaaki Murakami, Toshinobu Fujiwara, Satoshi Hirano, Hisataka Sabe
    Proceedings of the National Academy of Sciences of the United States of America 116 (35) 17450 - 17459 0027-8424 2019/08/27 [Refereed][Not invited]
     
    © 2019 National Academy of Sciences. All rights reserved. Although KRAS and TP53 mutations are major drivers of pancreatic ductal adenocarcinoma (PDAC), the incurable nature of this cancer still remains largely elusive. ARF6 and its effector AMAP1 are often overexpressed in different cancers and regulate the intracellular dynamics of integrins and E-cadherin, thus promoting tumor invasion and metastasis when ARF6 is activated. Here we show that the ARF6-AMAP1 pathway is a major target by which KRAS and TP53 cooperatively promote malignancy. KRAS was identified to promote eIF4A-dependent ARF6 mRNA translation, which contains a quadruplex structure at its 5′-untranslated region, by inducing TEAD3 and ETV4 to suppress PDCD4; and also eIF4E-dependent AMAP1 mRNA translation, which contains a 5′- terminal oligopyrimidine-like sequence, via up-regulating mTORC1. TP53 facilitated ARF6 activation by platelet-derived growth factor (PDGF), via its known function to promote the expression of PDGF receptor β (PDGFRβ) and enzymes of the mevalonate pathway (MVP). The ARF6-AMAP1 pathway was moreover essential for PDGF-driven recycling of PD-L1, in which KRAS, TP53, eIF4A/4Edependent translation, mTOR, and MVP were all integral. We moreover demonstrated that the mouse PDAC model KPC cells, bearing KRAS/TP53 mutations, express ARF6 and AMAP1 at high levels and that the ARF6-based pathway is closely associated with immune evasion of KPC cells. Expression of ARF6 pathway components statistically correlated with poor patient outcomes. Thus, the cooperation among eIF4A/4E-dependent mRNA translation and MVP has emerged as a link by which pancreatic driver mutations may promote tumor cell motility, PD-L1 dynamics, and immune evasion, via empowering the ARF6-based pathway and its activation by external ligands.
  • Handa H, Hashimoto A, Hashimoto S, Sugino H, Oikawa T, Sabe H
    Cell communication and signaling : CCS 16 (1) 94 - 94 2018/12 [Refereed][Not invited]
     
    BACKGROUND: TP53 mutations in cancer cells often evoke cell invasiveness, whereas fibroblasts show invasiveness in the presence of intact TP53. AMAP1 (also called DDEF1 or ASAP1) is a downstream effector of ARF6 and is essential for the ARF6-driven cell-invasive phenotype. We found that AMAP1 levels are under the control of p53 (TP53 gene product) in epithelial cells but not in fibroblasts, and here addressed that molecular basis of the epithelial-specific function of p53 in suppressing invasiveness via targeting AMAP1. METHODS: Using MDA-MB-231 cells expressing wild-type and p53 mutants, we identified miRNAs in which their expression is controlled by normal-p53. Among them, we identified miRNAs that target AMAP1 mRNA, and analyzed their expression levels and epigenetic statuses in epithelial cells and nonepithelial cells. RESULTS: We found that normal-p53 suppresses AMAP1 mRNA in cancer cells and normal epithelial cells, and that more than 30 miRNAs are induced by normal-p53. Among them, miR-96 and miR-182 were found to target the 3'-untranslated region of AMAP1 mRNA. Fibroblasts did not express these miRNAs at detectable levels. The ENCODE dataset demonstrated that the promoter region of the miR-183-96-182 cistron is enriched with H3K27 acetylation in epithelial cells, whereas this locus is enriched with H3K27 trimethylation in fibroblasts and other non-epithelial cells. miRNAs, such as miR-423, which are under the control of p53 but not associated with AMAP1 mRNA, demonstrated similar histone modifications at their gene loci in epithelial cells and fibroblasts, and were expressed in these cells. CONCLUSION: Histone modifications of certain miRNA loci, such as the miR-183-96-182 cistron, are different between epithelial cells and non-epithelial cells. Such epithelial-specific miRNA regulation appears to provide the molecular basis for the epithelial-specific function of p53 in suppressing ARF6-driven invasiveness.
  • Tsukasa Oikawa, Yutaro Otsuka, Hisataka Sabe
    Cancers 10 (6) E162  2072-6694 2018/06/01 [Refereed][Not invited]
     
    In addition to its classical roles as a tumor suppressor, p53 has also been shown to act as a guardian of epithelial integrity by inducing the microRNAs that target transcriptional factors driving epithelial–mesenchymal transition. On the other hand, the ENCODE project demonstrated an enrichment of putative motifs for the binding of p53 in epithelial-specific enhancers, such as CDH1 (encoding E-cadherin) enhancers although its biological significance remained unknown. Recently, we identified two novel modes of epithelial integrity (i.e., maintenance of CDH1 expression): one involves the binding of p53 to a CDH1 enhancer region and the other does not. In the former, the binding of p53 is necessary to maintain permissive histone modifications around the CDH1 transcription start site, whereas in the latter, p53 does not bind to this region nor affect histone modifications. Furthermore, these mechanisms likely coexisted within the same tissue. Thus, the mechanisms involved in epithelial integrity appear to be much more complex than previously thought. In this review, we describe our findings, which may instigate further experimental scrutiny towards understanding the whole picture of epithelial integrity as well as the related complex asymmetrical functions of p53. Such understanding will be important not only for cancer biology but also for the safety of regenerative medicine.
  • Yutaro Otsuka, Tsukasa Oikawa, Hinako Yoshino, Shigeru Hashimoto, Haruka Handa, Hiroki Yamamoto, Ari Hashimoto, Hisataka Sabe
    Cell Communication and Signaling 16 (1) 1  1478-811X 2018/01/05 [Refereed][Not invited]
     
    Background: The small GTPase Arf6 and its downstream effector AMAP1 (also called ASAP1/DDEF1) constitute a signaling pathway promoting cell invasion, in which AMAP1 interacts with several different proteins, including PRKD2, EPB41L5, paxillin, and cortactin. Components of this pathway are often overexpressed in human breast cancer cells, to be correlated with poor prognosis of the patients, whereas overexpression of the Arf6 pathway did not correlate with the four main molecular classes of human breast tumors. In this pathway, receptor tyrosine kinases, including EGFR and Her2, activate Arf6 via GEP100. MMTV-PyMT mice and MMTV-Neu mice are well-established models of human breast cancer, and exhibit the early dissemination and the lung metastasis, by utilizing protein tyrosine phosphorylation for oncogenesis. PyMT-tumors and Neu-tumors are known to have overlapping gene expression profiles, which primarily correspond to the luminal B-type of human mammary tumors, although they differ in the time necessary for tumor onset and metastasis. Given the common usage of protein tyrosine phosphorylation, as well as the frequent use of these animal models for studying breast cancer at the molecular level, we here investigated whether mammary tumors in these mouse models utilize the Arf6-based pathway for invasion. Methods: Expression levels of Arf6, AMAP1, and GEP100 were analyzed in PyMT-tumors and Neu-tumors by western blotting. Expression of Arf6 and AMAP1 was also analyzed by immunohistochemistry. The involvement of AMAP1 in invasion, and the possible correlation of its high expression levels with cancer mesenchymal properties were also investigated. Results: We found that PyMT-tumors, but not Neu-tumors, frequently overexpress AMAP1 and use it for invasion, whereas both types of tumors expressed Arf6 and GEP100 at different levels. High levels of the AMAP1 expression among PyMT-tumor cells were frequently correlated with loss of the epithelial marker CK8 and also with expression of the mesenchymal marker vimentin both at the primary sites and at sites of the lung metastases. Conclusions: PyMT-tumors appear to frequently utilize the Arf6-based invasive machinery, whereas Neu-tumors do not. Our results suggest that MMTV-PyMT mice, rather than MMTV-Neu mice, are useful to study the Arf6-based mammary tumor malignancies, as a representative model of human breast cancer.
  • Oikawa T, Otsuka Y, Onodera Y, Horikawa M, Handa H, Hashimoto S, Suzuki Y, Sabe H
    Scientific reports 8 (1) 1595 - 1595 2018/01 [Refereed][Not invited]
     
    TP53 mutation (i.e., loss of normal-p53) may evoke epithelial-mesenchymal transition (EMT), which was previously attributed to loss of certain miRNAs. However, not all epithelial cells undergo EMT upon TP53 mutation, and the p53-miRNA axis may not fully explain p53 function in epithelial integrity. We here show two modes of epithelial integrity: one involves p53-binding to a nucleotide region and the other does not. In the former, p53 binds to the CDH1 (encoding E-cadherin) locus to antagonize EZH2-mediated H3K27 trimethylation (H3K27me3) to maintain high levels of acetylation of H3K27 (H3K27ac). In the latter, the same locus is not highly acetylated at H3K27, and does not allow p53-binding, nor needs to antagonize EZH2. We moreover demonstrated that although the CDH1 locus in the p53-independent cells, but not in fibroblasts, becomes high-H3K27ac by butyrate and allows p53-biniding, their CDH1 expression does not become dependent on p53. Our results identified novel modes of the epithelial integrity, in which the same epithelial-specific gene locus exhibits different requirement for p53 with different histone modifications among different epithelial cells to warrant its expression.
  • Yuichi Mazaki, Yasuhito Onodera, Tsunehito Higashi, Takahiro Horinouchi, Tsukasa Oikawa, Hisataka Sabe
    CELL COMMUNICATION AND SIGNALING 15 (1) 36  1478-811X 2017/10 [Refereed][Not invited]
     
    Background: The small GTPase ARF1 mediates membrane trafficking mostly from the Golgi, and is essential for the G protein-coupled receptor (GPCR)-mediated chemotaxis of neutrophils. In this process, ARF1 is activated by the guanine nucleotide exchanger GBF1, and is inactivated by the GTPase-activating protein GIT2. Neutrophils generate the G beta gamma.-PAK1-aPIX-GIT2 linear complex during GPCR-induced chemotaxis, in which aPIX activates RAC1/CDC42, which then employs PAK1. However, it has remained unclear as to why GIT2 is included in this complex. Results: We investigated the association between ARF1 and RAC1/CDC42 during the fMLP-stimulated chemotaxis of HL60 cells. We found that the silencing of GBF1 significantly impaired the recruitment of RAC1 to the leading edges, but not PAK1, aPIX, RAC2, or CDC42. A significant population of RAC1 colocalized with ARF1 at the leading edges in stimulated cells, whereas fMLP activated both ARF1 and ARF5. Consistently, the silencing of ARF1, but not ARF5, impaired the recruitment of RAC1, whereas the silencing of RAC1 did not affect the recruitment of ARF1 to the leading edges. Conclusions: Our results indicated that the activation of ARF1 triggers the plasma membrane recruitment of RAC1 in GPCR-mediated chemotaxis, which is essential for cortical actin remodeling. Thus, membrane remodeling at the leading edges appears to precede actin remodeling in chemotaxis. Together with the fact that GIT2, which inactivates ARF1, is an integral component of the machinery activating RAC1, we proposed a model in which the ARF1-RAC1 linkage enables the regulation of ARF1 by repetitive on/off cycles during GPCR-mediated neutrophil chemotaxis.
  • Yutaro Otsuka, Hiroki Sato, Tsukasa Oikawa, Yasuhito Onodera, Jin-Min Nam, Ari Hashimoto, Kiyoshi Fukunaga, Kanako C. Hatanaka, Yutaka Hatanaka, Yoshihiro Matsuno, Satoshi Fukuda, Hisataka Sabe
    CELL COMMUNICATION AND SIGNALING 14 (1) 28  1478-811X 2016/11 [Refereed][Not invited]
     
    Background: Squamous cell carcinoma of the tongue (tongue SCC) is a major subtype of head and neck squamous cell carcinoma (HNSCC), which is an intractable cancer under current therapeutics. ARF6 and its effector AMAP1 are often overexpressed in different types of cancers, such as breast cancer and renal cancer, and in these cancers, AMAP1 binds to EPB41L5 to promote invasion, metastasis, and drug resistance. EPB41L5 is a mesenchymal-specific protein, normally induced during epithelial-mesenchymal transition (EMT) to promote focal adhesion dynamics. Similarly to breast cancer and renal cancer, the acquisition of mesenchymal phenotypes is the key process that drives the malignancy of HNSCC. We previously showed that the overexpression of AMAP1 in tongue SCC is statistically correlated with the poor outcome of patients. In this study, we examined whether tongue SCC also expresses EPB41L5 at high levels. Results: Immunohistochemical staining of clinical specimens of tongue SCC demonstrated that high expression levels of EPB41L5 statistically correlate with poor disease-free survival and poor overall survival rates of patients. The tongue SCC cell line SCC-9, which overexpress Arf6 and AMAP1, also expressed EPB41L5 at high levels to promote invasiveness, whereas the weakly invasive SCC-25 cells did not express EPB41L5 at notable levels. Among the different EMT-associated transcriptional factors, ZEB1 was previously found to be most crucial in inducing EPB41L5 in breast cancer and renal cancer. In contrast, expression levels of ZEB1 did not correlate with the expression levels of EPB41L5 in tongue SCC, whereas KLF8 and FOXO3 levels showed positive correlations with EPB41L5 levels. Moreover, silencing of EPB41L5 only marginally improved the drug resistance of SCC-9 cells, even when coupled with ionizing radiation. Conclusion: Our results indicate that activation of the cancer mesenchymal program in tongue SCC, which leads to EPB41L5 expression, closely correlates with the poor prognosis of patients. However, ZEB1 was not the major inducer of EPB41L5 in tongue SCC, unlike in breast cancer and renal cancer. Thus, processes that trigger the mesenchymal program of tongue SCC, which drives their malignancies, seem to be substantially different from those of other cancers.
  • A. Hashimoto, S. Hashimoto, H. Sugino, A. Yoshikawa, Y. Onodera, H. Handa, T. Oikawa, H. Sabe
    ONCOGENESIS 5 (9) e259  2157-9024 2016/09 [Refereed][Not invited]
     
    Onset of the cancer mesenchymal program is closely associated with cancer malignancy and drug resistance. Among the different epithelial-mesenchymal transition (EMT)-associated transcriptional factors, ZEB1 has a key role in inducing the mesenchymal phenotypes and stem cell-like properties of different breast cancer cells. ARF6 and its effector AMAP1 are frequently overexpressed in breast cancer cells, and promote invasion, metastasis and drug resistance. EPB41L5 is induced during EMT, and mediates the disruption of E-cadherin-based cell-cell adhesion and the promotion of focal adhesion dynamics. Here we show that EPB41L5 is an integral component of the ARF6-based pathway, which is induced by ZEB1. We found that EPB41L5 is expressed at high levels in malignant breast cancer cells and binds to AMAP1. ZEB1 induced EPB41L5 both in cancer cells and normal cells. This relationship was recaptured with The Cancer Genome Atlas RNASeq data set, and correlated with the poor outcome of the patients. In contrast, diversified events, such as tumor growth factor beta 1 stimulation, expression of SNAI1 and TP53 mutation, can each cause the induction of ZEB1 and EPB41L5, depending on the cellular context. Our results demonstrated that the ZEB1-EPB41L5 axis is at the core of the cancer mesenchymal program that drives ARF6-based invasion, metastasis and drug resistance of significant populations of primary breast cancers, and is tightly correlated with the poor outcomes of patients.
  • Sabe H, Hashimoto A, Hashimoto S, Oikawa T
    Molecular & cellular oncology 3 (4) e1185564  2016/07 [Refereed][Not invited]
     
    The mevalonate pathway results in the prenylation of small GTPases, which are pivotal for oncogenesis and cancer malignancies. However, inhibitors of this pathway, such as statins, have not necessarily produced favorable results in clinical trials. We recently identified properties of statin responders, together with the underlying molecular mechanisms and simple biomarkers to predict these responders.
  • Ari Hashimoto, Tsukasa Oikawa, Shigeru Hashimoto, Hirokazu Sugino, Ayumu Yoshikawa, Yutaro Otsuka, Haruka Handa, Yasuhito Onodera, Jin-Min Nam, Chitose Oneyama, Masato Okada, Mitsunori Fukuda, Hisataka Sabe
    JOURNAL OF CELL BIOLOGY 213 (1) 81 - 95 0021-9525 2016/04 [Refereed][Not invited]
     
    Drug resistance, metastasis, and a mesenchymal transcriptional program are central features of aggressive breast tumors. The GTPase Arf6, often overexpressed in tumors, is critical to promote epithelial mesenchymal transition and invasiveness. The metabolic mevalonate pathway (MVP) is associated with tumor invasiveness and known to prenylate proteins, but which prenylated proteins are critical for MVP-driven cancers is unknown. We show here that MVP requires the Arf6-dependent mesenchymal program. The MVP enzyme geranylgeranyl transferase II (GGT-II) and its substrate Rab11b are critical for Arf6 trafficking to the plasma membrane, where it is activated by receptor tyrosine kinases. Consistently, mutant p53, which is known to support tumorigenesis via MVP, promotes Arf6 activation via GGT-II and Rab11b. Inhibition of MVP and GGT-II blocked invasion and metastasis and reduced cancer cell resistance against chemotherapy agents, but only in cells overexpressing Arf6 and components of the mesenchymal program. Overexpression of Arf6 and mesenchymal proteins as well as enhanced MVP activity correlated with poor patient survival. These results provide insights into the molecular basis of MVP-driven malignancy.
  • Shigeru Hashimoto, Shuji Mikami, Hirokazu Sugino, Ayumu Yoshikawa, Ari Hashimoto, Yasuhito Onodera, Shotaro Furukawa, Haruka Handa, Tsukasa Oikawa, Yasunori Okada, Mototsugu Oya, Hisataka Sabe
    NATURE COMMUNICATIONS 7 10656  2041-1723 2016/02 [Refereed][Not invited]
     
    Acquisition of mesenchymal properties by cancer cells is critical for their malignant behaviour, but regulators of the mesenchymal molecular machinery and how it is activated remain elusive. Here we show that clear cell renal cell carcinomas (ccRCCs) frequently utilize the Arf6-based mesenchymal pathway to promote invasion and metastasis, similar to breast cancers. In breast cancer cells, ligand-activated receptor tyrosine kinases employ GEP100 to activate Arf6, which then recruits AMAP1; and AMAP1 then binds to the mesenchymal-specific protein EPB41L5, which promotes epithelial-mesenchymal transition and focal adhesion dynamics. In renal cancer cells, lysophosphatidic acid (LPA) activates Arf6 via its G-protein-coupled receptors, in which GTP-G alpha 12 binds to EFA6. The Arf6-based pathway may also contribute to drug resistance. Our results identify a specific mesenchymal molecular machinery of primary ccRCCs, which is triggered by a product of autotaxin and it is associated with poor outcome of patients.
  • Maiko Matsushita, Yoshie Ozaki, Yuka Hasegawa, Fukiko Terada, Noriko Tabata, Hirokazu Shiheido, Hiroshi Yanagawa, Tsukasa Oikawa, Koichi Matsuo, Wenlin Du, Taketo Yamada, Masashi Hozumi, Daiju Ichikawa, Yutaka Hattori
    PLOS ONE 10 (1) e0116135  1932-6203 2015/01 [Refereed][Not invited]
     
    Despite the recent advances in the treatment of multiple myeloma (MM), MM patients with high-risk cytogenetic changes such as t(4;14) translocation or deletion of chromosome 17 still have extremely poor prognoses. With the goal of helping these high-risk MM patients, we previously developed a novel phthalimide derivative, TC11. Here we report the further characterization of TC11 including anti-myeloma effects in vitro and in vivo, a pharmacokinetic study in mice, and anti-osteoclastogenic activity. Intraperitoneal injections of TC11 significantly delayed the growth of subcutaneous tumors in human myeloma-bearing SCID mice. Immunohistochemical analyses showed that TC11 induced apoptosis of MM cells in vivo. In the pharmacokinetic analyses, the C-max was 2.1 mu M at 1 h after the injection of TC11, with 1.2 h as the half-life. TC11 significantly inhibited the differentiation and function of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclasts in mouse osteoclast cultures using M-CSF and RANKL. We also revealed that TC11 induced the apoptosis of myeloma cells accompanied by alpha-tubulin fragmentation. In addition, TC11 and lenalidomide, another phthalimide derivative, directly bound to nucleophosmin 1 (NPM1), whose role in MM is unknown. Thus, through multiple molecular interactions, TC11 is a potentially effective drug for high-risk MM patients with bone lesions. The present results suggest the possibility of the further development of novel thalidomide derivatives by drug designing.
  • Tsukasa Oikawa, Yukiko Kuroda, Koichi Matsuo
    CELLULAR AND MOLECULAR LIFE SCIENCES 70 (18) 3341 - 3353 1420-682X 2013/09 [Refereed][Not invited]
     
    Osteoclasts are bone-resorbing cells of monocytic origin. An imbalance between bone formation and resorption can lead to osteoporosis or osteopetrosis. Osteoclastogenesis is triggered by RANKL- and IP3-induced Ca2+ influx followed by activation of NFATc1, a master transcription factor for osteoclastogenic gene regulation. During differentiation, osteoclasts undergo cytoskeletal remodeling to migrate and attach to the bone surface. Simultaneously, they fuse with each other to form multinucleated cells. These processes require PI3-kinase-dependent cytoskeletal protein activation to initiate cytoskeletal remodeling, resulting in the formation of circumferential podosomes and fusion-competent protrusions. In multinucleated osteoclasts, circumferential podosomes mature into stabilized actin rings, which enables the formation of a ruffled border where intensive membrane trafficking is executed. Membrane lipids, especially phosphoinositides, are key signaling molecules that regulate osteoclast morphology and act as second messengers and docking sites for multiple important effectors. We examine the critical roles of phosphoinositides in the signaling cascades that regulate osteoclast functions.
  • Tsukasa Oikawa, Atsuko Nakamura, Nobuyuki Onishi, Taketo Yamada, Koichi Matsuo, Hideyuki Saya
    Cancer Research 73 (16) 5100 - 5109 0008-5472 2013/08/15 [Refereed][Not invited]
     
    NFATc1 is a transcription factor that regulates T-cell development, osteoclastogenesis, and macrophage function. Given that T cells, osteoclasts, and macrophages in the tumor microenvironment are thought to modulate tumor progression, tumor cells may acquire NFATc1 expression through fusion with these NFATc1- expressing normal cells. We here revealed that a small proportion of tumor cells in human carcinoma specimens expressed NFATc1. To investigate the consequences of NFATc1 acquisition by tumor cells, we established A549 and MCF7 cell lines expressing a constitutively active form of NFATc1 (NFATc1CA) in an inducible manner. The expression of NFATc1CA promoted cancer cell invasion in association with changes in cell morphology. Analysis of gene expression andRNAinterference experiments revealed that NFATc1CA suppressed E-cadherin expression by upregulating the transcriptional repressors Snail and Zeb1 in a manner independent of TGF-b signaling. Induced expression of NFATc1CA also downregulated E-cadherin expression and increased invasive activity in tumor xenografts in vivo. Our results thus suggest that the acquisition of NFATc1 expression contributes to tumor progression. © 2013 American Association for Cancer Research.
  • Tsukasa Oikawa, Hitomi Okamura, Franziska Dietrich, Yosuke Senju, Tadaomi Takenawa, Shiro Suetsugu
    PLOS ONE 8 (3) e60528  1932-6203 2013/03 [Refereed][Not invited]
     
    Podosomes are cellular "`feet," characterized by F-actin-rich membrane protrusions, which drive cell migration and invasion into the extracellular matrix. Small GTPases that regulate the actin cytoskeleton, such as Cdc42 and Rac are central regulators of podosome formation. The adaptor protein IRSp53 contains an I-BAR domain that deforms membranes into protrusions and binds to Rac, a CRIB motif that interacts with Cdc42, an SH3 domain that binds to many actin cytoskeletal regulators with proline-rich peptides including VASP, and the C-terminal variable region by splicing. However, the role of IRSp53 and VASP in podosome formation had been unclear. Here we found that the knockdown of IRSp53 by RNAi attenuates podosome formation and migration in Src-transformed NIH3T3 ( NIH-Src) cells. Importantly, the differences in the IRSp53 C-terminal splicing isoforms did not affect podosome formation. Overexpression of IRSp53 deletion mutants suggested the importance of linking small GTPases to SH3 binding partners. Interestingly, VASP physically interacted with IRSp53 in NIH-Src cells and was essential for podosome formation. These data highlight the role of IRSp53 as a linker of small GTPases to VASP for podosome formation.
  • Tsukasa Oikawa, Koichi Matsuo
    Communicative and Integrative Biology 5 (5) 511 - 515 1942-0889 2012/09 [Refereed][Not invited]
     
    Podosomes and invadopodia seen in osteoclasts and cancer cells, respectively, are actin-rich membrane protrusions. We recently demonstrated that an adaptor protein, Tks5, which is an established regulator of invadopodia in cancer cells, drives osteoclast-osteoclast fusion as well as osteoclast-cancer cell fusion by generating circumferential podosomes/invadopodia. This finding revealed an unexpected potential of podosomes/invadopodia to act as fusion-competent protrusions. Fusion of biological membranes involves the intricate orchestration of various proteins and lipids. Recent literature suggests the importance of membrane curvature formation in lipid bilayer fusion. In this study, we investigated the expression of Bin-Amphiphysin-Rvs161/167 (BAR) domain superfamily proteins, which have membrane deforming activity, during osteoclastogenesis. We found that IRTKS was specifically induced during osteoclast fusion and interacted with Tks5, suggesting the role of IRTKS in the formation of fusion-competent protrusions via its BAR domain. © 2012 Landes Bioscience.
  • Tsukasa Oikawa, Masaaki Oyama, Hiroko Kozuka-Hata, Shunsuke Uehara, Nobuyuki Udagawa, Hideyuki Saya, Koichi Matsuo
    JOURNAL OF CELL BIOLOGY 197 (4) 553 - 568 0021-9525 2012/05 [Refereed][Not invited]
     
    Osteoclasts fuse to form multinucleated cells during osteoclastogenesis. This process is mediated by dynamic rearrangement of the plasma membrane and cytoskeleton, and it requires numerous factors, many of which have been identified. The underlying mechanism remains obscure, however. In this paper, we show that Tks5, a master regulator of invadopodia in cancer cells, is crucial for osteoclast fusion downstream of phosphoinositide 3-kinase and Src. Expression of Tks5 was induced during osteoclastogenesis, and prevention of this induction impaired both the formation of circumferential podosomes and osteoclast fusion without affecting cell differentiation. Tyrosine phosphorylation of Tks5 was attenuated in Src(-/-) osteoclasts, likely accounting for defects in podosome organization and multinucleation in these cells. Circumferential invadopodia formation in B16F0 melanoma cells was also accompanied by Tks5 phosphorylation. Co-culture of B16F0 cells with osteoclasts in an inflammatory milieu promoted the formation of melanoma-osteoclast hybrid cells. Our results thus reveal an unexpected link between circumferential podosome/invadopodium formation and cell-cell fusion in and beyond osteoclasts.
  • Hideki Yamaguchi, Tsukasa Oikawa
    ONCOTARGET 1 (5) 320 - 328 1949-2553 2010/09 [Refereed][Not invited]
     
    Invadopodia are extracellular matrix (ECM)-degrading protrusions formed by invasive cancer cells. Podosomes are structures functionally similar to invadopodia that are found in oncogene-transformed fibroblasts and monocyte-derived cells, including macrophages and osteoclasts. These structures are thought to play important roles in the pericellular remodeling of ECM during cancer invasion and metastasis. Much effort has been directed toward identification of the molecular components and regulators of invadopodia/podosomes, which could be therapeutic targets in the treatment of malignant cancers. However, it remains largely unknown how these components are assembled into invadopodia/podosomes and how the assembly process is spatially and temporally regulated. This review will summarize recent progress on the molecular mechanisms of invadopodia/podosome formation, with strong emphasis on the roles of lipid rafts and phosphoinositides.
  • Tsukasa Oikawa, Tadaomi Takenawa
    CELL ADHESION & MIGRATION 3 (2) 195 - 197 1933-6918 2009/04 [Refereed][Not invited]
     
    Cell-to-extracellular matrix (ECM) adhesion plays important roles in various biological events, such as proliferation, differentiation and migration. Distinct from other types of adhesion structures (focal complexes, focal adhesions and so on), podosomes and invadopodia are thought to have additional functions beyond attachment, possibly including invasion into the ECM. For podosomes and invadopodia to invade into the ECM, molecules involved in adhesion, actin polymerization and ECM degradation must be recruited to sites of action. Our recent study demonstrated that podosomes form near newly formed focal adhesions via the minimally expressed phosphoinositide PtdIns(3,4) P2-mediated recruitment of the Tks5-Grb2 scaffold, followed by the accumulation of N-WASP. Although this study demonstrated details of molecular interplay during the transformation of focal adhesion, its regulation in the in vivo invasion process remains to be clarified. Here, we discuss the molecular bases of the transformation of focal adhesions to podosomes/invadopodia based on current understanding.
  • Tsukasa Oikawa, Toshiki Itoh, Tadaomi Takenawa
    JOURNAL OF CELL BIOLOGY 182 (1) 157 - 169 0021-9525 2008/07 [Refereed][Not invited]
     
    Podosomes (also termed invadopodia in cancer cells) are actin-rich adhesion structures with matrix degradation activity that develop in various cell types. Despite their significant physiological importance, the molecular mechanism of podosome formation is largely unknown. In this study, we investigated the molecular mechanisms of podosome formation. The expression of various phosphoinositide-binding domains revealed that the podosomes in Src-transformed NIH3T3 (NIH-src) cells are enriched with PtdIns(3,4)P2, suggesting an important role of this phosphoinositide in podosome formation. Live-cell imaging analysis revealed that Src-expression stimulated podosome formation at focal adhesions of NIH3T3 cells after PtdIns(3,4)P2 accumulation. The adaptor protein Tks5/FISH, which is essential for podosome formation, was found to form a complex with Grb2 at adhesion sites in an Src-dependent manner. Further, it was found that N-WASP bound all SH3 domains of Tks5/FISH, which facilitated circular podosome formation. These results indicate that augmentation of the N-WASP-Arp2/3 signal was accomplished on the platform of Tks5/FISH-Grb2 complex at focal adhesions, which is stabilized by PtdIns(3,4) P2.
  • Daisuke Yamazaki, Tsukasa Oikawa, Tadaomi Takenawa
    JOURNAL OF CELL SCIENCE 120 (1) 86 - 100 0021-9533 2007/01 [Refereed][Not invited]
     
    During cadherin-dependent cell-cell adhesion, the actin cytoskeleton undergoes dynamic reorganization in epithelial cells. Rho-family small GTPases, which regulate actin dynamics, play pivotal roles in cadherin-dependent cell-cell adhesion; however, the precise molecular mechanisms that underlie cell-cell adhesion formation remain unclear. Here we show that Wiskott-Aldrich syndrome protein family verprolin-homologous protein (WAVE)-mediated reorganization of actin, downstream of Rac plays an important role in normal development of cadherin-dependent cell-cell adhesions in MDCK cells. Rac-induced development of cadherin-dependent adhesions required WAVE2-dependent actin reorganization. The process of cell-cell adhesion is divided into three steps: formation of new cell-cell contacts, stabilization of these new contacts and junction maturation. WAVE1 and WAVE2 were expressed in MDCK cells. The functions of WAVE1 and WAVE2 were redundant in this system but WAVE2 appeared to play a more significant role. During the first step, WAVE2-dependent lamellipodial protrusions facilitated formation of cell-cell contacts. During the second step, WAVE2 recruited actin filaments to new cell-cell contacts and stabilized newly formed cadherin clusters. During the third step, WAVE2-dependent actin reorganization was required for organization and maintenance of mature cell-cell adhesions. Thus, Rac-WAVE-dependent actin reorganization is not only involved in formation of cell-cell adhesions but is also required for their maintenance.
  • Shiro Suetsugu, Kazutaka Murayama, Ayako Sakamoto, Kyoko Hanawa-Suetsugu, Azusa Seto, Tsukasa Oikawa, Chiemi Mishima, Mikako Shirouzu, Tadaomi Takenawa, Shigeyuki Yokoyama
    JOURNAL OF BIOLOGICAL CHEMISTRY 281 (46) 35347 - 35358 0021-9258 2006/11 [Refereed][Not invited]
     
    The concave surface of the crescent-shaped Bin-amphiphysin-Rvs (BAR) domain is postulated to bind to the cell membrane to induce membrane deformation of a specific curvature. The Rac binding (RCB) domain/IRSp53-MIM homology domain (IMD) has a dimeric structure that is similar to the structure of the BAR domain; however, the RCB domain/IMD has a "zeppelin-shaped" dimer. Interestingly, the RCB domain/IMD of IRSp53 possesses Rac binding, membrane binding, and actin filament binding abilities. Here we report that the RCB domain/IMD of IRSp53 induces membrane deformation independent of the actin filaments in a Rac-dependent manner. In contrast to the BAR domain, the RCB domain/IMD did not cause long tubulation of the artificial liposomes; however, the Rac binding domain caused the formation of small buds on the liposomal surface. When expressed in cells, the Rac binding domain induced outward protrusion of the plasma membrane in a direction opposite to that induced by the BAR domain. Mapping of the amino acids responsible for membrane deformation suggests that the convex surface of the Rac binding domain binds to the membrane in a Rac-dependent manner, which may explain the mechanism of the membrane deformation induced by the RCB domain/IMD.
  • S Suetsugu, S Kurisu, T Oikawa, D Yamazaki, A Oda, T Takenawa
    JOURNAL OF CELL BIOLOGY 173 (4) 571 - 585 0021-9525 2006/05 [Refereed][Not invited]
     
    WAVE2 activates the actin-related protein (Arp) 2/3 complex for Rac-induced actin polymerization during lamellipodium formation and exists as a large WAVE2 protein complex with Sra1/PIR121, Nap1, Abi1, and HSPC300. IRSp53 binds to both Rac and Cdc42 and is proposed to link Rac to WAVE2. We found that the knockdown of IRSp53 by RNA interference decreased lamellipodium formation without a decrease in the amount of WAVE2 complex. Localization of WAVE2 at the cell periphery was retained in IRSp53 knockdown cells. Moreover, activated Cdc42 but not Rac weakened the association between WAVE2 and IRSp53. When we measured Arp2/3 activation in vitro, the WAVE2 complex isolated from the membrane fraction of cells was fully active in an IRSp53-dependent manner but WAVE2 isolated from the cytosol was not. Purified WAVE2 and purified WAVE2 complex were activated by IRSp53 in a Rac-dependent manner with PIP3-containing liposomes. Therefore, IRSp53 optimizes the activity of the WAVE2 complex in the presence of activated Rac and PIP3.
  • K Tsujita, S Suetsugu, N Sasaki, M Furutani, T Oikawa, T Takenawa
    JOURNAL OF CELL BIOLOGY 172 (2) 269 - 279 0021-9525 2006/01 [Refereed][Not invited]
     
    T he conserved FER-CIP4 homology (FCH) domain is found in the pombe Cdc15 homology (PCH) protein family members, including formin-binding protein 17 (FBP17). However, the amino acid sequence homology extends beyond the FCH domain. We have termed this region the extended FC (EFC) domain. We found that FBP17 coordinated membrane deformation with actin cytoskeleton reorganization during endocytosis. The EFC domains of FBP17, CIP4, and other PCH protein family members show weak homology to the Bin-amphiphysin-Rvs ( BAR) domain. The EFC domains bound strongly to phosphatidylserine and phosphatidylinositol 4,5-bisphosphate and deformed the plasma membrane and liposomes into narrow tubules. Most PCH proteins possess an SH3 domain that is known to bind to dynamin and that recruited and activated neural Wiskott-Aldrich syndrome protein (N-WASP) at the plasma membrane. FBP17 and/or CIP4 contributed to the formation of the protein complex, including N-WASP and dynamin-2, in the early stage of endocytosis. Furthermore, knockdown of endogenous FBP17 and CIP4 impaired endocytosis. Our data indicate that PCH protein family members couple membrane deformation to actin cytoskeleton reorganization in various cellular processes.
  • T Oikawa, H Yamaguchi, T Itoh, M Kato, T Ijuin, D Yamazaki, S Suetsugu, T Takenawa
    NATURE CELL BIOLOGY 6 (5) 420 - + 1465-7392 2004/05 [Refereed][Not invited]
     
    Polarized cell movement is triggered by the development of a PtdIns(3,4,5)P-3 gradient at the membrane, which is followed by rearrangement of the actin cytoskeleton. The WASP family verprolin homologous protein (WAVE) is essential for lamellipodium formation at the leading edge by activating the Arp2/3 complex downstream of Rac GTPase. Here, we report that WAVE2 binds to Ptdlns(3,4,5)P-3 through its basic domain. The amino-terminal portion of WAVE2, which includes the Ptdlns(3,4,5)P-3-binding sequence, was localized at the leading edge of lamellipodia induced by an active form of Rac (RacDA) or by treatment with platelet-derived growth factor (PDGF). Production of Ptdlns(3,4,5)P-3 at the cell membrane by myristoylated phosphatidylinositol-3-OH kinase (PION is sufficient to recruit WAVE2 in the presence of dominant-negative Rac and latrunculin, demonstrating that Ptdlns(3,4,5)P-3 alone is able to recruit WAVE2. Expression of a full-length mutant of WAVE2 that lacks the lipid-binding activity inhibited proper formation of lamellipodia induced by RacDA. These results suggest that one of the products of PI(3)K, Ptdlns(3,4,5)P-3, recruits WAVE2 to the polarized membrane and that this recruitment is essential for lamellipodium formation at the leading edge.

MISC

  • 麓佳月, 高田真吾, 小野寺康仁, 川口喬吾, 畠山鎮次, 佐邊壽孝, 及川司  日本分子生物学会年会プログラム・要旨集(Web)  45th-  2022
  • 及川司, 大西なおみ, 小野寺康仁, 橋本あり, 植田幸嗣, 佐邊壽孝  日本生化学会大会(Web)  93rd-  2020
  • 膵癌ドライバー変異はmRNA翻訳と蛋白質プレニル化を介しARF6が駆動する癌免疫回避を促進する(Pancreatic KRAS/TP53 mutations promote ARF6-based immune evasion via activating mRNA translation and protein prenylation)
    橋本 あり, 橋本 茂, 古川 聖太郎, 蔦保 暁生, 小野寺 康人, 半田 悠, 及川 司, 水上 裕輔, 西川 義浩, 児玉 裕三, 村上 正晃, 平野 聡, 佐邊 壽孝  日本癌学会総会記事  78回-  P  -3033  2019/09
  • 及川司, 大西なおみ, 小野寺康仁, 橋本あり, 植田幸嗣, 佐邊壽孝  日本生化学会大会(Web)  92nd-  2019
  • 膵癌ドライバー変異はARF6-AMAP1経路を活性化し悪性度と免疫回避能を促進する(Pancreatic KRAS and TP53 oncogenes cooperatively activate ARF6-AMAP1 pathway to drive malignancy and immune evasion)
    橋本 あり, 橋本 茂, 古川 聖太郎, 蔦保 暁生, 小野寺 康仁, 大塚 勇太郎, 半田 悠, 及川 司, 水上 裕輔, 村上 正晃, 平野 聡, 佐邊 壽孝  日本癌学会総会記事  77回-  2219  -2219  2018/09  [Not refereed][Not invited]
  • 上皮細胞においてp53はE-cadherin遺伝子発現制御部位に結合し、EZH2による発現抑制に拮抗する
    及川 司, 大塚 勇太郎, 小野寺 康仁, 堀川 芽衣, 橋本 あり, 橋本 茂, 鈴木 穣, 佐邊 壽孝  生命科学系学会合同年次大会  2017年度-  [3PT18  -06(3P  2017/12  [Not refereed][Not invited]
  • 橋本あり, 橋本茂, 古川聖太郎, 古川聖太郎, 蔦保暁生, 蔦保暁生, 大塚勇太郎, 半田悠, 小野寺康仁, 及川司, 平野聡, 佐邊壽孝  日本生化学会大会(Web)  90th-  ROMBUNNO.1P‐1028 (WEB ONLY)  -1028]  2017/12  [Not refereed][Not invited]
  • がんの浸潤・転移研究の新機軸 Arf6経路 難治性癌の悪性度進展・抗癌剤抵抗性に根幹的経路
    佐邊 壽孝, 橋本 あり, 小野寺 康仁, 及川 司, 橋本 茂  日本癌学会総会記事  76回-  S1  -2  2017/09  [Not refereed][Not invited]
  • 上皮形質安定性をp53に依存する上皮細胞と依存しない上皮細胞の差異に関する解析
    及川 司, 大塚 勇太郎, 小野寺 康仁, 堀川 芽衣, 橋本 あり, 橋本 茂, 鈴木 穣, 佐邊 壽孝  日本癌学会総会記事  76回-  P  -1107  2017/09  [Not refereed][Not invited]
  • 真崎雄一, 小野寺康仁, 東恒仁, 堀之内孝広, 及川司, 佐邊壽孝  日本細胞生物学会大会(Web)  69th-  ROMBUNNO.T8‐11(P1‐077) (WEB ONLY)  -63  2017/05  [Not refereed][Not invited]
  • p53はEZH2と機能的に競合することで上皮性維持に寄与する
    及川 司, 大塚 勇太郎, 小野寺 康仁, 半田 悠, 橋本 あり, 橋本 茂, 鈴木 穣, 佐邊 壽孝  日本癌学会総会記事  75回-  J  -3030  2016/10  [Not refereed][Not invited]
  • 橋本 あり, 橋本 茂, 及川 司, 大塚 勇太郎, 半田 悠, 小野寺 康仁, 佐邊 壽孝  日本生化学会大会プログラム・講演要旨集  89回-  [1P  -268]  2016/09  [Not refereed][Not invited]
  • 古川 聖太郎, 橋本 あり, 橋本 茂, 小野寺 康仁, 及川 司, 大塚 勇太郎, 佐邊 壽孝, 平野 聡  日本外科学会定期学術集会抄録集  116回-  PS  -002  2016/04  [Not refereed][Not invited]
  • p53はエピジェネティック制御を介して上皮性を維持する
    及川 司, 小野寺 康仁, 大塚 勇太郎, 半田 悠, 橋本 あり, 橋本 茂, 鈴木 穣, 佐邊 壽孝  日本生化学会大会・日本分子生物学会年会合同大会講演要旨集  88回・38回-  [2P1108]  -[2P1108]  2015/12  [Not refereed][Not invited]
  • Arf6-AMAP1経路によるROS制御は乳癌の放射線抵抗性に寄与する
    小野寺 康仁, 南 ジンミン, 及川 司, 白土 博樹, 佐邊 壽孝  日本癌学会総会記事  74回-  E  -1049  2015/10  [Not refereed][Not invited]
  • Arf6-AMAP1経路によるROS制御は乳癌の放射線抵抗性に寄与する
    小野寺 康仁, 南 ジンミン, 及川 司, 白土 博樹, 佐邊 壽孝  日本癌学会総会記事  74回-  E  -1049  2015/10  [Not refereed][Not invited]
  • Arf6-AMAP1経路による酸化還元状態の恒常性維持は乳癌の放射線抵抗性に寄与する(Robust redox homeostasis mediated by Arf6-AMAP1 pathway confers resistance to ionizing radiation in breast cancer)
    小野寺 康仁, 南 ジンミン, 及川 司, 白土 博樹, 佐邊 壽孝  日本癌学会総会記事  73回-  E  -3010  2014/09  [Not refereed][Not invited]
  • EZH2発現亢進により創出されるArf6を中心とした間葉浸潤に特化した分子装置は腎癌の予後不良に関与する(EZH2 generates Arf6-based mesenchymal invasion machinery that is central to poor prognosis of renal cancer)
    橋本 茂, 杉野 弘和, 橋本 あり, 吉河 歩, 及川 司, 半田 悠, 大家 基嗣, 三上 修治, 佐邊 壽孝  日本癌学会総会記事  73回-  J  -2075  2014/09  [Not refereed][Not invited]
  • 乳癌において変異p53がリガンド反応性の間葉型浸潤分子装置を創出する機序(TP53 alterations generate Arf6-based mesenchymal invasion pathway that is activated by RTKs and TGFβ1 in breast cancer)
    橋本 あり, 橋本 茂, 杉野 弘和, 吉河 歩, 及川 司, 小野寺 康仁, 半田 悠, 大塚 勇太郎, 岩見 昴亮, 小根山 千歳, 岡田 雅人, 福田 光則, 佐邊 壽孝  日本癌学会総会記事  73回-  J  -2077  2014/09  [Not refereed][Not invited]
  • p53は間葉系形質を持つ乳がん細胞に上皮系形質を再獲得させる(p53 recalls epithelial memory in mammary cancer cells with mesenchymal phenotypes)
    及川 司, 小野寺 康仁, 橋本 あり, 橋本 茂, 佐邊 壽孝  日本癌学会総会記事  73回-  P  -1159  2014/09  [Not refereed][Not invited]
  • 癌放射線治療への分子生物学的アプローチ 変異p53が放射線抵抗性に根幹的な間葉型浸潤経路を創出する機構(Toward the improvement of radiotherapy: Approaches from the molecular biological point of view Mechanisms by which oncogenic mutant-p53 generates mesenchymal invasive pathway pivotal to a radiation resis
    佐邊 壽孝, 橋本 あり, 橋本 茂, 小野寺 康仁, 及川 司, Nam Jin-Min, 小根山 千歳, 杉野 弘和, 吉河 歩, 大塚 勇太郎, 半田 悠, 芳野 正修, 岡田 雅人  日本癌学会総会記事  72回-  64  -64  2013/10  [Not refereed][Not invited]
  • アクチン細胞骨格による新たな細胞機能と生体機能の制御 アダプター分子Tks5依存的なポドソーム/インベードポディア形成と、これを介した破骨/がん細胞融合の解析
    及川 司, 尾山 大明, 秦 裕子, 中村 敦子, 大西 伸幸, 上原 俊介, 宇田川 信之, 山田 健人, 佐谷 秀行, 松尾 光一  日本生化学会大会プログラム・講演要旨集  86回-  1S16a  -3  2013/09  [Not refereed][Not invited]
  • 及川司, 尾山大明, 秦裕子, 中村敦子, 大西伸幸, 上原俊介, 宇田川信之, 山田健人, 佐谷秀行, 松尾光一  日本生化学会大会(Web)  86th-  1S16A-3 (WEB ONLY)  -3  2013  [Not refereed][Not invited]
  • ポドソーム形成と細胞融合におけるアダプター分子Tks5の機能解析(Tks5-dependent formation of circumferential podosomes mediates cell-cell fusion)
    及川 司  松本歯学  37-  (2-3)  186  -186  2011/12  [Not refereed][Not invited]
  • Tks5によるポドソーム形成と破骨細胞融合メカニズム
    及川 司, 尾山 大明, 秦 裕子, 佐谷 秀行, 松尾 光一  日本骨代謝学会学術集会プログラム抄録集  29回-  241  -241  2011/07  [Not refereed][Not invited]
  • ポドソーム形成と細胞融合におけるアダプター分子Tks5の機能解析(Tks5-dependent formation of circumferential podosome mediates cell-cell fusion)
    及川 司, 尾山 大明, 秦 裕子, 佐谷 秀行, 松尾 光一  日本細胞生物学会大会講演要旨集  63回-  144  -144  2011/05  [Not refereed][Not invited]
  • 破骨細胞融合におけるアダプター分子Tks5の機能解析
    及川 司, 松尾 光一  日本骨代謝学会学術集会プログラム抄録集  28回-  260  -260  2010/07  [Not refereed][Not invited]
  • 細胞接着・ECM・細胞間相互作用 破骨細胞融合におけるアダプター分子Tks5の機能解析(Cell adhesion, ECM and cell-cell interaction Tks5 is required for the cell-cell fusion process of osteoclastogenesis)
    及川 司, 佐谷 秀行, 松尾 光一  日本細胞生物学会大会講演要旨集  62回-  121  -121  2010/05  [Not refereed][Not invited]
  • 及川司  臨床整形外科  45-  (4)  330  -334  2010/04/25  [Not refereed][Not invited]
  • (あり方委員会企画)Bone Cell Topology 破骨細胞と骨芽細胞の極性決定
    及川 司, 松尾 光一  日本骨代謝学会学術集会プログラム抄録集  27回-  118  -118  2009/07  [Not refereed][Not invited]
  • 及川司, 松尾光一  日本骨代謝学会学術集会プログラム抄録集  27th-  118  2009  [Not refereed][Not invited]
  • 癌浸潤転移における細胞運動のメカニズム 浸潤突起形成の分子メカニズム(Molecular mechanisms of cell migration in cancer invasion and metastasis Sequential signals toward podosome formation in NIH-src cells)
    及川 司, 伊藤 俊樹, 竹縄 忠臣  日本細胞生物学会大会講演要旨集  60回-  94  -94  2008/06  [Not refereed][Not invited]
  • 浸潤突起形成の分子メカニズム(Sequential signals toward podosome formation in NIH-src cells)
    及川 司, 伊藤 俊樹, 竹縄 忠臣  日本細胞生物学会大会講演要旨集  60回-  166  -166  2008/06  [Not refereed][Not invited]
  • WASP/WAVE蛋白質はがん細胞の運動や浸潤に関与する(Cancer and Failures in Cell Polaritv Regulation WASP/WAVE proteins regulate the migration and invasion of cancer cells)
    竹縄 忠臣, 及川 司, 末次 志郎  日本癌学会総会記事  66回-  68  -68  2007/08  [Not refereed][Not invited]
  • PI3Kおよび関連シグナル伝達分子の機能 癌細胞におけるD3-ホスホイノシチドの定量化と視覚化のためのホスホイノシチド結合ドメインの利用(Use of phosphoinositide-binding domains for the quantification and visualization of D3-phosphoinositides in cancer cells)
    伊藤 俊樹, 及川 司, 辻田 和也, 竹縄 忠臣  日本発生生物学会・日本細胞生物学会合同大会要旨集  40回・59回-  15  -15  2007/05  [Not refereed][Not invited]
  • Tks5/FISHはPtdIns(3,4)P2とPtdIns(3,4,5)P3が豊富な局所接着部(focal adhesion)へ自己分子を局在化させることによりpodosome形成を開始させる(Tks5/FISH initiates podosome formation by localizing itself to PtdIns(3,4)P2- and PtdIns(3,4,5)P3-enriched focal adhesions)
    及川 司, 伊藤 俊樹, 竹縄 忠臣  日本発生生物学会・日本細胞生物学会合同大会要旨集  40回・59回-  131  -131  2007/05  [Not refereed][Not invited]
  • 分子標的としてのシグナル伝達系 N-WASP/WAVEのシグナルと癌浸潤、転移
    竹縄 忠臣, 末次 志郎, 山崎 大輔, 及川 司, 栗栖 修作  日本癌学会総会記事  65回-  433  -433  2006/09  [Not refereed][Not invited]
  • 竹縄忠臣, 末次志郎, 山崎大輔, 及川司, 栗栖修作  日本癌学会学術総会記事  65th-  433  2006/08/28  [Not refereed][Not invited]
  • 及川司  実験医学  24-  (13)  2063  -2068  2006/08/15  [Not refereed][Not invited]
  • 【形と運動を司る細胞のダイナミクス 最新技術による分子機構の解明から疾患治療への展開まで】 細胞と形態形成・疾患 癌の浸潤,転移とアクチン細胞骨格による浸潤突起形成
    及川 司  実験医学  24-  (13)  2063  -2068  2006/08  [Not refereed][Not invited]
     
    癌の浸潤,転移において細胞外基質(ECM)の分解は非常に重要なステップである.ECMを分解し浸潤する癌細胞は,浸潤突起とよばれる特別な伸展構造を形成し,そこでECMを分解し,浸潤していくと考えられる.浸潤突起はアクチンフィラメントに富み,アクチン調節分子,プロテアーゼなど多くの分子が集積する動的な構造であるが,形成機構や制御機構,生理的な意義など不明な点も多い.RNAiによるタンパク質の発現抑制を中心とした最近の研究から,WASP/N-WASPとArp2/3複合体を中心とした浸潤突起形成から,メタロプロテアーゼによるECMの分解に至る機構が少しずつ明らかになってきた(著者抄録)
  • 竹縄忠臣, 及川司, 伊集院壮  日本分子生物学会年会プログラム・講演要旨集  27th-  312  2004/11/25  [Not refereed][Not invited]
  • Takenawa T, Suetsugu S, Oikawa T  Biophysics  43-  (1)  S8  -S8  2003/08/25  [Not refereed][Not invited]
  • Racによるlamellipodia形成におけるWAVE2とPtdIns(3,4,5)P3の関わり
    及川 司, 末次 志郎, 伊藤 俊樹, 山口 英樹, 竹縄 忠臣  日本細胞生物学会大会講演要旨集  56回-  23  -23  2003/05  [Not refereed][Not invited]
  • 及川司, 末次志郎, 伊藤俊樹, 山口英樹, 竹縄忠臣  日本細胞生物学会大会講演要旨集  56th-  23  2003  [Not refereed][Not invited]

Research Projects

  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2022/04 -2025/03 
    Author : 伊藤 俊樹, 及川 司
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2020/04 -2023/03 
    Author : 及川 司
     
    本研究は、DNA複製期のヒストンH3と相互作用するタンパク質群(インタラクトーム)において、癌抑制遺伝子産物p53が、抑制性ヒストン修飾H3K27me3化を担う分子EZH2を排除する、という申請者らの新しい知見に焦点を当て、その分子的基盤とともに、細胞の上皮性維持への寄与を明らかにすることを目的としている。 本年度は、p53が存在する時にH3のインタラクトームに含まれる量が大きく増加する分子、CTDNEP1及び、その脱リン酸化基質分子の一つでありフォスファチジン酸(PA)からジアシルグリセロール(DAG)への変換を担うLipinの機能解析を行った。その結果、(1)CTDNEP1は核膜及び小胞体様の細胞内構造に局在すること、(2)CTDNEP1の発現はp53の存在によりタンパクレベルで安定化されること、(3)CTDNEP1の発現抑制は、p53の発現抑制と同様にH3K27me3の核膜近傍領域への異所性蓄積を引き起こすこと、(4)核内に存在するLipinタンパク量はp53やCTDNEP1に依存すること、(5)p53やCTDNEP1は核内PA量の調節に関与することを細胞核脂質の質量分析から明らかにした。これらのことから、p53が存在することでCTDNEP1が核膜において安定化し、Lipinは脱リン酸化されて活性化し、核膜の脂質組成がPAからDAGに富んだ状態になることが示唆された。ヒストンH3はDAGよりもPAに強く結合することを考え合わせると、上記のp53-CTDNEP1-Lipinによる核内PA量の低下が行われない状況では、DNA複製に伴い細胞質から核内へ一斉に輸送されるヒストンH3.1が核膜付近に蓄積し、これが異所性H3K27me3の原因となり得るのではないかと考えられた。
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2020/04 -2022/03 
    Author : 及川 司
     
    遺伝子発現に関わるヒストン修飾は、細胞内代謝産物によって動的に制御されているが、ヌクレオソーム構造の再編成を伴うDNA複製時、娘鎖においてヒストンの化学修飾状態が維持または変化する機構は良くわかっていない。申請者らは、転写抑制性メチル化ヒストン修飾(K27-trimethylated histone H3: H3K27me3)のゲノム上におけるパターンがDNA複製を経ても安定に維持されるにあたり、がん抑制遺伝子産物p53が重要な役割を果たすことを見出した。p53が存在しない時、DNA複製期に細胞質から核内へ輸送される新規未修飾ヒストンH3.1が、核膜に一時的に蓄積し、ここで異所性にH3K27me3化された。このH3K27me3化には、核膜近傍領域においてPRC2タンパク複合体の触媒サブユニットであるEZH2とH3との分子近接が観察された。従って、娘鎖において少なくとも一部のゲノム領域で、新規未修飾ヒストンH3が供給される代わりに、H3K27me3が供給されると考えられる。この系を用い、DNA複製部位において新規未修飾ヒストンH3の供給量が減少し、代わりにH3K27me3が供給されたとき、娘鎖においてメチル化ヒストンパターン(H3K27me3, H3K4me3)がどのように遷移するか、理論解析を進めた。その結果、DNA複製部位への未修飾新規ヒストンの流入量、あるいはメチル基ドナーであるSAMの供給量が過剰であるなど、平衡から遠く離れた時、メチル化ヒストンのわずかな初期濃度差(擾乱)が増幅し、ある複製回数以降から不可逆的に隔たるというシミュレーション結果を得た。これを上記のp53喪失の系で検証したところ、p53を喪失した上皮細胞においては、既にH3K27me3化されているゲノム領域(PRC2標的領域)でH3K27me3量の増加が観察された。
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2018/04 -2021/03 
    Author : Onodera Yasuhito
     
    Using cell-specific regulation of glucose metabolism, we co-cultured glucose-sufficient and glucose-deficient cancer cells and established "metabolic cooperation" between them to analyze viability, intracellular metabolism, phenotypic changes, and response to radiation. The above experimental system revealed that cancer cells in a glucose-depleted state maintain their viability by obtaining metabolites from glucose-sufficient cells, and that oxidative stress and ER stress in the former cells are markedly suppressed. Estimation of the mediating substances and mechanisms, as well as the various phenotypic changes that occur in the metabolic cooperative state, are currently under analysis.
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)
    Date (from‐to) : 2016/04 -2019/03 
    Author : Oikawa Tsukasa
     
    Analyses of various cancer cells have demonstrated a statistical correlation between TP53 mutations and the infringement of epithelial phenotypes, suggesting that some epithelial cells require TP53 to maintain their integrity. Likewise, the ENCODE project indicates the enrichment of putative p53 binding motifs within the regulatory regions of epithelial genes. However, the roles of p53 in epithelial integrity still largely remain elusive. We showed that epithelial genes may require normal-p53 to encounter EZH2. The loss of normal-p53 induced EZH2-mediated H3K27me3 deposition at histones regulating epithelial genes, such as CDH1. p53 can access this locus in epithelial cells but not in mesenchymal cells. Our results in vitro and from TCGA datasets indicated that H3K27me3 deposition by the loss of p53 is specific to epithelial genes. Our results identified an uncharted function of normal-p53 to protect epithelial genes from EZH2-mediated repression to maintain epithelial integrity.
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research Grant-in-Aid for Young Scientists (A)
    Date (from‐to) : 2011/04 -2014/03 
    Author : OIKAWA Tsukasa
     
    Cell-cell fusion requires dynamic rearrangement of the plasma membrane and cytoskeleton, and this process involves numerous previously characterized factors. However, the mechanisms and consequences of cell-cell fusion remains obscure. This study revealed that an adaptor protein Tks5 is essential for both formation of circumferential podosomes and osteoclast fusion without altering osteoclast differentiation. As Tks5 is known to promote the formation of invadopodia in cancer cells, I tested if these cells also have the potential to fuse with osteoclasts. Among the cells tested, B16F0 melanoma cells formed circumferential invadopodia with Tks5 accumulation. Co-culture of B16F0 melanoma cells with osteoclasts in an inflammatory milieu promoted increased formation of melanoma-osteoclast hybrid cells. These results revealed a previously unknown mechanism of regulation of both circumferential podosomes/invadopodia formation and cell-cell fusion by Tks5.
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research Grant-in-Aid for Challenging Exploratory Research
    Date (from‐to) : 2011 -2013 
    Author : OIKAWA Tsukasa
     
    Among various phosphatidylinositols (PI) on the plasma membrane, the most abundant product, PI(4,5)P2 and locally-produced PI(3,4,5)P3 and PI(3,4)P2 seem to be particularly important in polarity formation of the cells. The PH domain of PLC delta1 binds to PI(4,5)P2 while the PH domain of Akt binds to PI(3,4)P2 and PI(3,4,5)P3, which is locally produced depending on PI3-kinase activity. By using these domains, I succeeded in observing the polarity of fusing cells. To apply this system in vivo, KH2 ES cells which can express those domains fused to GFP or RFP in response to doxycyclin were generated. Now that genetic quality of the ES cells were confirmed, the mice who express the polarity probe is being generated.
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research Grant-in-Aid for Young Scientists (B)
    Date (from‐to) : 2008 -2009 
    Author : OIKAWA Tsukasa
     
    The great majority of cancers occur in epithelial tissues, yielding carcinomas. Given that epithelial cells are on the basement membrane underneath, they must degrade it to travel to distant sites. To initiate the invasion process, they must make contact with the extracellular matrices (ECM). Thus, the cell-matrix interactions must be one of the triggers that allow cancer cells to invade. To penetrate into the ECM, actin-rich adhesion structures called invadopodia or podosomes is thought to play pivotal roles for degrading it. I have studied the mechanism of invadopodium/podosome formation in Src-transformed NIH3T3 cells. In this study, based on the results of RNAi, identification of binding proteins by mass spectrometry, protein interaction and live-cell imaging analysis, I have established a stepwise mechanism for invadopodium formation. Invadopodia initiate from focal adhesions (FAs) due to changes in the phosphorylation status of proteins and in the composition of phosphoinositides on the plasma membrane. The onset of actin polymerization is triggered by PI(3,4)P2 production and Tks5 recruitment followed by N-WASP accumulation. This step involves intricate interactions of Tks5 with both PI(3,4)P2 and Grb2 at FAs
  • 日本学術振興会:科学研究費助成事業 特別研究員奨励費
    Date (from‐to) : 2005 -2006 
    Author : 及川 司
     
    近年、粘菌や好中球から繊維芽細胞にいたる様々な細胞の方向性を持った遊走が、走化性因子のわずかな勾配を感じて活性化されたPI3-キナーゼとその産物のひとつ、PIP3から始まるシグナルによって起こることが分かってきている。一方でGFPを融合させたAktのPHドメインを用いてPIP3の局在を可視化すると、この脂質は遊走細胞の先端に局所的に産生されていることが明らかになり、イノシトールリン脂質が細胞の極性や遊走に積極的に関与していることが明らかになっている。研究代表者はこれまで細胞の移動先端部で活性化され、アクチン重合を促進し細胞に駆動力を与えるWASPファミリータンパク質のひとつ、WAVE2がいかにして活性化されるのかということを解く過程で、イノシトールリン脂質との結合を見出した。本研究ではイノシトールリン脂質結合の性質を明らかにし、邸WASPによる浸潤突起形成やWA〜Eによる葉状仮足形成、及び方向性を持った細胞遊走へのイノシトールリン脂質の関与を解明することを目的としている。浸潤能の高いがん細胞に多く見られる浸潤斑と呼ばれる浸潤突起は、N-WASPがその骨格であるアクチン重合において主要な役割を果たしている。一方、様々なイノシトールリン脂質結合ドメインを用いて細胞内のイノシトールリン脂質の局在を可視化したところ、PI(3,4)P2が浸潤斑に強く極性を持って局在していた。リボソーム法やDot Blot法により、各種イノシトールリン脂質とN-WASPとの結合特異性を検証したところ、NLWASPは特定のイノシトールリン脂質とは強く結合しなかった。一方、N-WASPとともに浸潤斑に局在するアダプタータンパク質、Tks5/FISHはN-WASPやdynaminと結合し、さらにいくつかのイノシトールリン脂質とも結合することがわかった。これらのことから、N-WASPはその結合タンパク質であるTks5/FISHと協調してイノシトールリン脂質による制御を受けていることが示唆された。またRNAiによるTks5/FISHやN-WASPの発現抑制実験から、浸潤突起ができるメカニズムとして、(1)focal adhesionにおけるPI(3,4)P2やPI(3,4,5)P3の集積、(2)Tks5/FISHとN-WASPを含む複合体の集積、(3)N-WASPによるアクチン重合、という時間、空間制御が行われていることが示唆された。


Copyright © MEDIA FUSION Co.,Ltd. All rights reserved.