Updated on 2024/04/10

写真a

 
Daisuke Yamada
 
Organization
Faculty of Medicine, Dentistry and Pharmaceutical Sciences Assistant Professor
Position
Assistant Professor
External link

Degree

  • 博士(医学) ( 2014.3   金沢大学 )

  • 修士(薬学) ( 2010.3   金沢大学 )

  • 学士(薬学) ( 2008.3   金沢大学 )

Research Interests

  • Tumor biology

  • Regenerative science

  • Tumor initiating cells

  • Regenerative medicine

  • pluripotent stem cells

Research Areas

  • Life Science / Tumor biology

  • Life Science / Cell biology

  • Life Science / Molecular biology

  • Life Science / Tumor diagnostics and therapeutics

  • Life Science / Developmental biology

Education

  • Kanazawa University   大学院医学系研究科   がん医科学専攻

    2010.4 - 2014.3

      More details

    Country: Japan

    researchmap

  • Kanazawa University   大学院自然科学研究科   生命薬学専攻

    2008.4 - 2010.3

      More details

    Country: Japan

    researchmap

  • Kanazawa University   薬学部   総合薬学科

    2004.4 - 2008.3

      More details

    Country: Japan

    researchmap

  • 石川県立金沢錦丘高等学校   普通科  

    2001.4 - 2004.3

      More details

    Country: Japan

    researchmap

Research History

  • 岡山大学 学術研究院 医歯薬学域(医学系)   研究准教授

    2023.2 - 2025.3

      More details

    Country:Japan

    researchmap

  • 岡山大学 学術研究院 医歯薬学域(医学系)   助教

    2021.4

      More details

  • 岡山大学大学院医歯薬学総合研究科   助教

    2017.4 - 2021.3

      More details

  • Niigata University of Pharmacy and Applied Life Sciences   Faculty of Pharmaceutical Sciences   Research Assistant

    2014.4 - 2017.3

      More details

Professional Memberships

▼display all

Committee Memberships

  • 岡山大学   医学部共同実験室運営委員会委員  

    2019.5   

      More details

  • 岡山大学   基礎・社会医学系教育企画委員  

    2017.4   

      More details

 

Papers

  • Identification of Surface Antigens That Define Human Pluripotent Stem Cell-Derived PRRX1+Limb-Bud-like Mesenchymal Cells. Reviewed International journal

    Daisuke Yamada, Tomoka Takao, Masahiro Nakamura, Toki Kitano, Eiji Nakata, Takeshi Takarada

    International journal of molecular sciences   23 ( 5 )   2022.2

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)  

    Stem cell-based therapies and experimental methods rely on efficient induction of human pluripotent stem cells (hPSCs). During limb development, the lateral plate mesoderm (LPM) produces limb-bud mesenchymal (LBM) cells that differentiate into osteochondroprogenitor cells and form cartilage tissues in the appendicular skeleton. Previously, we generated PRRX1-tdTomato reporter hPSCs to establish the protocol for inducing the hPSC-derived PRRX1+ LBM-like cells. However, surface antigens that assess the induction efficiency of hPSC-derived PRRX1+ LBM-like cells from LPM have not been identified. Here, we used PRRX1-tdTomato reporter hPSCs and found that high pluripotent cell density suppressed the expression of PRRX1 mRNA and tdTomato after LBM-like induction. RNA sequencing and flow cytometry suggested that PRRX1-tdTomato+ LBM-like cells are defined as CD44high CD140Bhigh CD49f-. Importantly, other hPSC lines, including four human induced pluripotent stem cell lines (414C2, 1383D2, HPS1042, HPS1043) and two human embryonic stem cell lines (SEES4, SEES7), showed the same results. Thus, an appropriate cell density of hPSCs before differentiation is a prerequisite for inducing the CD44high CD140Bhigh CD49f- PRRX1+ LBM-like cells.

    DOI: 10.3390/ijms23052661

    PubMed

    researchmap

  • Induction and expansion of human PRRX1+ limb-bud-like mesenchymal cells from pluripotent stem cells. Reviewed International journal

    Daisuke Yamada, Masahiro Nakamura, Tomoka Takao, Shota Takihira, Aki Yoshida, Shunsuke Kawai, Akihiro Miura, Lu Ming, Hiroyuki Yoshitomi, Mai Gozu, Kumi Okamoto, Hironori Hojo, Naoyuki Kusaka, Ryosuke Iwai, Eiji Nakata, Toshifumi Ozaki, Junya Toguchida, Takeshi Takarada

    Nature biomedical engineering   5 ( 8 )   926 - 940   2021.8

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)  

    Current protocols for the differentiation of human pluripotent stem cells (hPSCs) into chondrocytes do not allow for the expansion of intermediate progenitors so as to prospectively assess their chondrogenic potential. Here we report a protocol that leverages PRRX1-tdTomato reporter hPSCs for the selective induction of expandable and ontogenetically defined PRRX1+ limb-bud-like mesenchymal cells under defined xeno-free conditions, and the prospective assessment of the cells' chondrogenic potential via the cell-surface markers CD90, CD140B and CD82. The cells, which proliferated stably and exhibited the potential to undergo chondrogenic differentiation, formed hyaline cartilaginous-like tissue commensurate to their PRRX1-expression levels. Moreover, we show that limb-bud-like mesenchymal cells derived from patient-derived induced hPSCs can be used to identify therapeutic candidates for type II collagenopathy and we developed a method to generate uniformly sized hyaline cartilaginous-like particles by plating the cells on culture dishes coated with spots of a zwitterionic polymer. PRRX1+ limb-bud-like mesenchymal cells could facilitate the mass production of chondrocytes and cartilaginous tissues for applications in drug screening and tissue engineering.

    DOI: 10.1038/s41551-021-00778-x

    PubMed

    researchmap

  • PRRX1-TOP2A interaction is a malignancy-promoting factor in human malignant peripheral nerve sheath tumours. Reviewed International journal

    Shota Takihira, Daisuke Yamada, Tatsunori Osone, Tomoka Takao, Masakiyo Sakaguchi, Michiyuki Hakozaki, Takuto Itano, Eiji Nakata, Tomohiro Fujiwara, Toshiyuki Kunisada, Toshifumi Ozaki, Takeshi Takarada

    British journal of cancer   2024.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    BACKGROUND: Paired related-homeobox 1 (PRRX1) is a transcription factor in the regulation of developmental morphogenetic processes. There is growing evidence that PRRX1 is highly expressed in certain cancers and is critically involved in human survival prognosis. However, the molecular mechanism of PRRX1 in cancer malignancy remains to be elucidated. METHODS: PRRX1 expression in human Malignant peripheral nerve sheath tumours (MPNSTs) samples was detected immunohistochemically to evaluate survival prognosis. MPNST models with PRRX1 gene knockdown or overexpression were constructed in vitro and the phenotype of MPNST cells was evaluated. Bioinformatics analysis combined with co-immunoprecipitation, mass spectrometry, RNA-seq and structural prediction were used to identify proteins interacting with PRRX1. RESULTS: High expression of PRRX1 was associated with a poor prognosis for MPNST. PRRX1 knockdown suppressed the tumorigenic potential. PRRX1 overexpressed in MPNSTs directly interacts with topoisomerase 2 A (TOP2A) to cooperatively promote epithelial-mesenchymal transition and increase expression of tumour malignancy-related gene sets including mTORC1, KRAS and SRC signalling pathways. Etoposide, a TOP2A inhibitor used in the treatment of MPNST, may exhibit one of its anticancer effects by inhibiting the PRRX1-TOP2A interaction. CONCLUSION: Targeting the PRRX1-TOP2A interaction in malignant tumours with high PRRX1 expression might provide a novel tumour-selective therapeutic strategy.

    DOI: 10.1038/s41416-024-02632-8

    PubMed

    researchmap

  • Antitumor activity of α-pinene in T-cell tumors. Reviewed International journal

    Masaya Abe, Noboru Asada, Maiko Kimura, Chie Fukui, Daisuke Yamada, Ziyi Wang, Masayuki Miyake, Takeshi Takarada, Mitsuaki Ono, Michinori Aoe, Wataru Kitamura, Masayuki Matsuda, Takashi Moriyama, Akifumi Matsumura, Yoshinobu Maeda

    Cancer science   2024.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    T-cell acute leukemia and lymphoma have a poor prognosis. Although new therapeutic agents have been developed, their therapeutic effects are suboptimal. α-Pinene, a monoterpene compound, has an antitumor effect on solid tumors; however, few comprehensive investigations have been conducted on its impact on hematologic malignancies. This report provides a comprehensive analysis of the potential benefits of using α-pinene as an antitumor agent for the treatment of T-cell tumors. We found that α-pinene inhibited the proliferation of hematologic malignancies, especially in T-cell tumor cell lines EL-4 and Molt-4, induced mitochondrial dysfunction and reactive oxygen species accumulation, and inhibited NF-κB p65 translocation into the nucleus, leading to robust apoptosis in EL-4 cells. Collectively, these findings suggest that α-pinene has potential as a therapeutic agent for T-cell malignancies, and further investigation is warranted.

    DOI: 10.1111/cas.16086

    PubMed

    researchmap

  • Alpha-Pinene As a Novel Therapeutic Agent for T-Cell Tumors

    Masaya Abe, Noboru Asada, Maiko Kimura, Chie Fukui, Daisuke Yamada, Ziyi Wang, Masayuki Miyake, Takeshi Takarada, Mitsuaki Ono, Michinori Aoe, Wataru Kitamura, Masayuki Matsuda, Takashi Moriyama, Akifumi Matsumura, Yoshinobu Maeda

    Blood   142 ( Supplement 1 )   1441 - 1441   2023.11

     More details

    Publishing type:Research paper (scientific journal)   Publisher:American Society of Hematology  

    T-cell acute leukemia and lymphoma have poor prognoses, necessitating the development of novel therapeutic agents. In this study, we investigated the antitumor activity of α-pinene, a monoterpene compound, against T-cell tumors. We evaluated the effects of α-pinene on cell growth inhibition in vitro using the murine T-cell tumor cell line EL-4 and the human-derived T-cell tumor cell line Molt-4. In addition, we assessed the effects of limonene, another monoterpene with reported tumor-suppressive effects, as a comparative agent.

    Both α-pinene and limonene demonstrated concentration- and time-dependent inhibition of cell growth in both the EL-4 and Molt-4 cell lines (Figure 1). Moreover, α-pinene exhibited greater potency than limonene in inhibiting the growth of various hematologic malignancies. Importantly, α-pinene and limonene had minimal effects on the growth of normal murine spleen T cells. Further investigation of the mechanisms of action of α-pinene revealed its ability to induce apoptosis and cell cycle arrest in EL-4 and Molt-4 cells. Transcriptomic profiling of α-pinene-treated EL-4 cells using RNA-seq identified differentially expressed genes associated with cellular damage and mitochondrial dysfunction. Increased intracellular ROS levels and mitochondrial impairment were observed in the T-cell tumors treated with α-pinene. The activation of intrinsic apoptotic pathways involving EGR1, p53, BCL-2, and BAX was found to contribute to α-pinene-induced apoptosis in T-cell tumors. Moreover, α-pinene inhibited the NF-κB signaling pathway, resulting in the reduced nuclear translocation of NF-κB p65 and decreased total intracellular NF-κB p65 levels in EL-4 cells. Additionally, we discovered that α-pinene induced ferroptosis, a newly identified programmed cell death process, in EL-4 cells through lipid peroxidation and iron accumulation. Activation of the system x c−/GSH/GPX4 axis and upregulation of iron transporters, such as Slc39a8 and TfR1, were observed in α-pinene-induced ferroptosis. Treatment with the ferroptosis inhibitor Fer-1 partially reversed the tumor-inhibiting effect of α-pinene in EL-4 cells. Finally, we administrated α-pinene to mice subcutaneously injected with luciferase-expressing EL-4 to evaluate the efficacy of α-pinene in vivo. The tumor growth was significantly inhibited by α-pinene treatment (vehicle: 1055 ± 101 mm 3; α-pinene: 701 ± 82 mm 3, p < 0.01, Figure 2) without adverse effects on body weight or behavior. Immunohistochemistry analyses revealed a significant increase of CD8 + T-cells (vehicle: 8.1 ± 2.7 cells / field of view; α-pinene: 43.1 ± 12.2 cells / field of view , p < 0.05) and NK1.1 + cells (vehicle: 5.1 ± 1.0 cells / field of view ; α-pinene: 41.1 ± 9.8 cells / field of view , p < 0.01) in the tumors, suggesting that α-pinene may have an indirect antitumor effect by recruiting immune cells into tumors.

    Overall, our findings demonstrate the antitumor activity of α-pinene against T-cell tumors through the induction of apoptosis, cell cycle arrest, and ferroptosis. α-Pinene shows promise as a potential therapeutic agent for T-cell tumors and warrants further investigation for its clinical application.

    DOI: 10.1182/blood-2023-181447

    researchmap

  • Development of cartilage tissue using a stirred bioreactor and human iPSC-derived limb bud mesenchymal cells. Reviewed International journal

    Yuki Fujisawa, Tomoka Takao, Daisuke Yamada, Takeshi Takarada

    Biochemical and biophysical research communications   687   149146 - 149146   2023.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Production of cartilaginous particles for regenerative medicine requires a large supply of chondrocytes and development of suitable production techniques. Previously, we successfully produced human induced pluripotent stem cell (hiPSC)-derived limb bud mesenchymal cells (ExpLBM cells) with a high chondrogenic differentiation potential that stably proliferate. It may be possible to use these cells in combination with a stirred bioreactor to develop a tissue-engineered cell culture technology with potential for scale-up to facilitate production of large amounts of cartilaginous particles. ExpLBM cells derived from 414C2 and Ff-I 14s04 (human leukocyte antigen homozygous) hiPSCs were seeded into a stirred bioreactor containing cartilage induction medium. To characterize the cartilaginous particles produced, we performed real-time quantitative reverse transcription-polymerase chain reaction and histological analyses. Additionally, we transplanted the cartilage tissue into osteochondral defects of immunocompromised rats to assess its functionality, and evaluated engraftment of the grafted tissue. We successfully produced large amounts of cartilaginous particles via cartilage induction culture in a stirred bioreactor. This tissue exhibited significantly increased expression levels of type II collagen (COL2), aggrecan (ACAN), and SRY-box transcription factor 9 (SOX9), as well as positive Safranin O and Toluidine blue staining, indicating that it possesses characteristics of hyaline cartilage. Furthermore, engrafted tissues in osteochondral knee defects of immunodeficient rats were positively stained for human vimentin, COL2, and ACAN as well as with Safranin O. In this study, we successfully generated large amounts of hiPSC-derived cartilaginous particles using a combination of tissue engineering techniques. This method is promising as a cartilage regeneration technology with potential for scale-up.

    DOI: 10.1016/j.bbrc.2023.149146

    PubMed

    researchmap

  • Fabrication of shape-designable cartilage from human induced pluripotent stem cell-derived chondroprogenitors using a cell self-aggregation technique. Reviewed International journal

    Tomoyuki Ota, Tomoka Takao, Ryosuke Iwai, Takeshi Moriwaki, Yohei Kitaguchi, Yuki Fujisawa, Daisuke Yamada, Yoshihiro Kimata, Takeshi Takarada

    Biomedical materials (Bristol, England)   2023.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    With the advancement of tissue engineering technologies, implantable materials have been developed for use in facial plastic surgery, including auriculoplasty and rhinoplasty. Tissue-engineered cartilage comprising only cells and cell-produced extracellular matrix is considered valuable as there is no need to consider problems associated with the absorption of scaffold materials and the generation of immune responses. However, it is exceedingly difficult to produce large-sized complex shapes of cartilage without the use of scaffolds. In this study, we describe the production of shape-designable cartilage using a novel cell self-aggregation technique (CAT) and chondroprogenitor cells derived from human induced pluripotent stem cells as the source. The method described does not require special equipment such as bio-3D printers, and the produced tissue can be induced into well-matured cartilage with abundant cartilage matrix in vitro. Using CAT, we were able to generate cartilage in the form of rings or tubes with adjustable inner diameter and curvature, over a range of several centimeters, without the use of scaffolds. The in vitro fabrication of shape-designable cartilage using CAT will undoubtedly contribute to developments in the field of facial plastic surgery.&#xD.

    DOI: 10.1088/1748-605X/ad02d1

    PubMed

    researchmap

  • A novel chondrocyte sheet fabrication using human-induced pluripotent stem cell-derived expandable limb-bud mesenchymal cells. Reviewed International journal

    Tomoka Takao, Masato Sato, Yuki Fujisawa, Eriko Toyoda, Daisuke Yamada, Yukio Hitsumoto, Eiji Nakata, Toshifumi Ozaki, Takeshi Takarada

    Stem cell research & therapy   14 ( 1 )   34 - 34   2023.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    BACKGROUND: Cell sheet fabrication for articular cartilage regenerative medicine necessitates a large number of chondrocytes of consistent quality as a cell source. Previously, we have developed human-induced pluripotent stem cell (iPSC)-derived expandable PRRX1+ limb-bud mesenchymal cells (ExpLBM) with stable expansion and high chondrogenic capacity, while in this study; our ExpLBM technology was combined with cell sheet engineering to assess its potential as a stable cell source for articular cartilage regeneration. METHODS: ExpLBM cells derived from human-induced pluripotent stem cells (hiPSCs), including 414C2 and Ff-KVs09 (HLA homozygous), were seeded onto a culture plate and two-dimensional chondrogenic induction (2-DCI) was initiated. After 2-DCI, ExpLBM-derived chondrocytes were stripped and transferred to temperature-responsive culture inserts and the chondrocyte sheets were histologically examined or transplanted into osteochondral knee defects of immunodeficient rats. RESULTS: Immunohistochemistry revealed that ExpLBM-derived cell sheets were positive for Safranin O, COL2, and ACAN but that they were negative for COL1 and RUNX2. Furthermore, the engrafted tissues in osteochondral knee defects in immunodeficient rats were stained with SafO, human VIMENTIN, ACAN, and COL2. CONCLUSIONS: The present study is the first to report the chondrocyte sheet fabrication with hiPSC-derived cell source. hiPSC-derived ExpLBM would be a promising cell source for cell sheet technology in articular cartilage regenerative medicine.

    DOI: 10.1186/s13287-023-03252-4

    PubMed

    researchmap

  • A protocol to induce expandable limb-bud mesenchymal cells from human pluripotent stem cells. Reviewed International journal

    Tomoka Takao, Daisuke Yamada, Takeshi Takarada

    STAR protocols   3 ( 4 )   101786 - 101786   2022.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Here, we present a protocol for the selective differentiation of human pluripotent stem cells mimicking human developmental processes into expandable PRRX1+ limb-bud mesenchymal (ExpLBM) cells. This approach enables expansion through serial passage while maintaining capacity for chondrogenic differentiation. For complete details on the use and execution of this protocol, please refer to Yamada et al. (2021, 2022).

    DOI: 10.1016/j.xpro.2022.101786

    PubMed

    researchmap

  • Establishment of a human pluripotent stem cell-derived MKX-tdTomato reporter system. Reviewed International journal

    Yuki Fujisawa, Lu Ming, Daisuke Yamada, Tomoka Takao, Takeshi Takarada

    Stem cell research & therapy   13 ( 1 )   515 - 515   2022.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Tendon regeneration is difficult because detailed knowledge about tendon progenitor cells (TPCs), which produce tenocytes to repair tendon tissue, has not been revealed. Mohawk homeobox (MKX) is a marker of TPCs or tenocytes, but a human pluripotent stem cell (hPSC)-based reporter system that visualizes MKX+ cells has not been developed. Here, we established an hPSC-derived MKX-tdTomato reporter cell line and tested the induction ratio of MKX-tdTomato+ cells using our stepwise/xeno-free differentiation protocol. MKX-tdTomato+ cells were generated with high efficiency and expressed tendon-specific markers, including MKX, SCX, TNMD, and COL1A1. Our MKX-tdTomato hPSC line would be a useful tool for studying the development or regeneration of tendon tissue.

    DOI: 10.1186/s13287-022-03203-5

    PubMed

    researchmap

  • Vestigial-Like 3 Plays an Important Role in Osteoblast Differentiation by Regulating the Expression of Osteogenic Transcription Factors and BMP Signaling. Reviewed International journal

    Haoze Yuan, Mika Ikegame, Yoko Fukuhara, Fumiko Takemoto, Yaqiong Yu, Jumpei Teramachi, Yao Weng, Jiajie Guo, Daisuke Yamada, Takeshi Takarada, Ying Li, Hirohiko Okamura, Bin Zhang

    Calcified tissue international   111 ( 3 )   331 - 344   2022.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Our previous gene profiling analysis showed that the transcription cofactor vestigial-like 3 (VGLL3) gene expression was upregulated by mechanical tension in the mouse cranial suture, coinciding with accelerated osteoblast differentiation. Therefore, we hypothesized that VGLL3 plays a significant role in osteogenic differentiation. To clarify the function of VGLL3 in osteoblasts, we examined its expression characteristics in mouse bone tissue and the osteoblastic cell line MC3T3-E1. We further examined the effects of Vgll3 knockdown on osteoblast differentiation and bone morphogenetic protein (BMP) signaling. In the mouse cranial suture, where membranous ossification occurs, VGLL3 was immunohistochemically detected mostly in the nucleus of osteoblasts, preosteoblasts, and fibroblastic cells. VGLL3 expression in MC3T3-E1 cells was transient and peaked at a relatively early stage of differentiation. RNA sequencing revealed that downregulated genes in Vgll3-knockdown cells were enriched in gene ontology terms associated with osteoblast differentiation. Interestingly, most of the upregulated genes were related to cell division. Targeted Vgll3 knockdown markedly suppressed the expression of major osteogenic transcription factors (Runx2, Sp7/osterix, and Dlx5) and osteoblast differentiation. It also attenuated BMP signaling; moreover, exogenous BMP2 partially restore osteogenic transcription factors' expression in Vgll3-knockdown cells. Furthermore, overexpression of Vgll3 increased the expression of osteogenic transcription factors. These results suggest that VGLL3 plays a critical role in promoting osteoblast differentiation and that part of the process is mediated by BMP signaling. Further elucidation of VGLL3 function will increase our understanding of osteogenesis and skeletal disease etiology.

    DOI: 10.1007/s00223-022-00997-7

    PubMed

    researchmap

  • Mouse Model for Optogenetic Genome Engineering. Reviewed

    Tomoka Takao, Daisuke Yamada, Takeshi Takarada

    Acta medica Okayama   76 ( 1 )   1 - 5   2022.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Optogenetics, a technology to manipulate biological phenomena thorough light, has attracted much attention in neuroscience. Recently, the Magnet System, a photo-inducible protein dimerization system which can control the intracellular behavior of various biomolecules with high accuracy using light was developed. Furthermore, photoactivation systems for controlling biological phenomena are being developed by combining this technique with genome-editing technology (CRISPR/Cas9 System) or DNA recombination technology (Cre-loxP system). Herein, we review the history of optogenetics and the latest Magnet System technology and introduce our recently developed photoactivatable Cre knock-in mice with temporal-, spatial-, and cell-specific accuracy.

    DOI: 10.18926/AMO/63202

    PubMed

    researchmap

  • Oncogenic potential of human pluripotent stem cell-derived lung organoids with HER2 overexpression. Reviewed International journal

    Akihiro Miura, Daisuke Yamada, Masahiro Nakamura, Shuta Tomida, Dai Shimizu, Yan Jiang, Tomoka Takao, Hiromasa Yamamoto, Ken Suzawa, Kazuhiko Shien, Masaomi Yamane, Masakiyo Sakaguchi, Shinichi Toyooka, Takeshi Takarada

    International journal of cancer   149 ( 8 )   1593 - 1604   2021.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Lung adenocarcinoma (LUAD) is the most common types among lung cancers generally arising from terminal airway and understanding of multistep carcinogenesis is crucial to develop novel therapeutic strategy for LUAD. Here we used human induced pluripotent stem cells (hiPSCs) to establish iHER2-hiPSCs in which doxycycline induced the expression of the oncoprotein human epidermal growth factor receptor 2 (HER2)/ERBB2. Lung progenitors that differentiated from iHER2-hiPSCs, which expressed NKX2-1/TTF-1 known as a lung lineage maker, were cocultured with human fetal fibroblast and formed human lung organoids (HLOs) comprising alveolar type 2-like cells. HLOs that overexpressed HER2 transformed to tumor-like structures similar to atypical adenomatous hyperplasia, which is known for lung precancerous lesion and upregulated the activities of oncogenic signaling cascades such as RAS/RAF/MAPK and PI3K/AKT/mTOR. The degree of morphological irregularity and proliferation capacity were significantly higher in HLOs from iHER2-hiPSCs. Moreover, the transcriptome profile of the HLOs shifted from a normal lung tissue-like state to one characteristic of clinical LUAD with HER2 amplification. Our results suggest that hiPSC-derived HLOs may serve as a model to recapitulate the early tumorigenesis of LUAD and would provide new insights into the molecular basis of tumor initiation and progression.

    DOI: 10.1002/ijc.33713

    PubMed

    researchmap

  • O-GlcNAcylation drives calcium signaling toward osteoblast differentiation: A bioinformatics-oriented study. Reviewed International journal

    Yao Weng, Ziyi Wang, Yoko Fukuhara, Airi Tanai, Mika Ikegame, Daisuke Yamada, Takeshi Takarada, Takashi Izawa, Satoru Hayano, Kaya Yoshida, Hiroshi Kamioka, Hirohiko Okamura

    BioFactors (Oxford, England)   47 ( 6 )   992 - 1015   2021.8

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    This study aimed to reveal the possible mechanisms by which O-linked-N-acetylglucosaminylation (O-GlcNAcylation) regulates osteoblast differentiation using a series of bioinformatics-oriented experiments. To examine the influence of O-GlcNAcylation levels on osteoblast differentiation, osteoblastic MC3T3-E1 cells were treated with O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) inhibitors. Correlations between the levels of O-GlcNAcylation and the expression of osteogenic markers as well as OGT were evaluated by qPCR and western blotting. The O-GlcNAcylated proteins assumed to correlate with Runx2 expression were retrieved from several public databases and used for further bioinformatics analysis. Following the findings of the bioinformatics analysis, intracellular calcium ([Ca2+ ]i ) was monitored in the cells treated with OGT and OGA inhibitors using a confocal laser-scanning microscope (CLS). The interaction effect between O-GlcNAcylation and [Ca2+ ]i on osteogenic marker expression was determined using stable OGT knockdown MC3T3-E1 cells. O-GlcNAcylation was positively associated with osteoblast differentiation. The time-course profile of global O-GlcNAcylated proteins showed a distinctive pattern with different molecular weights during osteoblast differentiation. The expression pattern of several O-GlcNAcylated proteins was significantly similar to that of Runx2 expression. Bioinformatic analysis of the retrieved Runx2-related-O-GlcNAcylated-proteins revealed the importance of [Ca2+ ]i . CLS showed that alteration of O-GlcNAcylation rapidly changed [Ca2+ ]i in MC3T3-E1 cells. O-GlcNAcylation and [Ca2+ ]i showed an interaction effect on the expression of osteogenic markers. OGT knockdown disrupted the [Ca2+ ]i -induced expression changes of osteogenic markers. O-GlcNAcylation interacts with [Ca2+ ]i and elicits osteoblast differentiation by regulating the expression of osteogenic markers.

    DOI: 10.1002/biof.1774

    PubMed

    researchmap

  • PRRX1 promotes malignant properties in human osteosarcoma. Reviewed International journal

    Ryoji Joko, Daisuke Yamada, Masahiro Nakamura, Aki Yoshida, Shota Takihira, Tomoka Takao, Ming Lu, Kohei Sato, Tatsuo Ito, Toshiyuki Kunisada, Eiji Nakata, Toshifumi Ozaki, Takeshi Takarada

    Translational oncology   14 ( 1 )   100960 - 100960   2021.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Paired related homeobox 1 (PRRX1) is a marker of limb bud mesenchymal cells, and deficiency of p53 or Rb in Prrx1-positive cells induces osteosarcoma in several mouse models. However, the regulatory roles of PRRX1 in human osteosarcoma have not been defined. In this study, we performed PRRX1 immunostaining on 35 human osteosarcoma specimens to assess the correlation between PRRX1 level and overall survival. In patients with osteosarcoma, the expression level of PRRX1 positively correlated with poor prognosis or the ratio of lung metastasis. Additionally, we found PRRX1 expression on in 143B cells, a human osteosarcoma line with a high metastatic capacity. Downregulation of PRRX1 not only suppressed proliferation and invasion but also increased the sensitivity to cisplatin and doxorubicin. When 143B cells were subcutaneously transplanted into nude mice, PRRX1 knockdown decreased tumor sizes and rates of lung metastasis. Interestingly, forskolin, a chemical compound identified by Connectivity Map analysis using RNA expression signatures during PRRX1 knockdown, decreased tumor proliferation and cell migration to the same degree as PRRX1 knockdown. These results demonstrate that PRRX1 promotes tumor malignancy in human osteosarcoma.

    DOI: 10.1016/j.tranon.2020.100960

    PubMed

    researchmap

  • Establishment of a tTA-dependent photoactivatable Cre recombinase knock-in mouse model for optogenetic genome engineering. Reviewed International journal

    Tomoka Takao, Yuichi Hiraoka, Kenji Kawabe, Daisuke Yamada, Lu Ming, Kohichi Tanaka, Moritoshi Sato, Takeshi Takarada

    Biochemical and biophysical research communications   526 ( 1 )   213 - 217   2020.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    The Cre-loxP recombination system is widely used to generate genetically modified mice for biomedical research. Recently, a highly efficient photoactivatable Cre (PA-Cre) based on reassembly of split Cre fragments has been established. This technology enables efficient DNA recombination that is activated upon blue light illumination with spatiotemporal precision. In this study, we generated a tTA-dependent photoactivatable Cre-loxP recombinase knock-in mouse model (TRE-PA-Cre mice) using a CRISPR/Cas9 system. These mice were crossed with ROSA26-tdTomato mice (Cre reporter mouse) to visualize DNA recombination as marked by tdTomato expression. We demonstrated that external noninvasive LED blue light illumination allows efficient DNA recombination in the liver of TRE-PA-Cre:ROSA26-tdTomato mice transfected with tTA expression vectors using hydrodynamic tail vein injection. The TRE-PA-Cre mouse established here promises to be useful for optogenetic genome engineering in a noninvasive, spatiotemporal, and cell-type specific manner in vivo.

    DOI: 10.1016/j.bbrc.2020.03.015

    PubMed

    researchmap

  • Postnatal Runx2 deletion leads to low bone mass and adipocyte accumulation in mice bone tissues. Reviewed

    Tosa I, Yamada D, Yasumatsu M, Hinoi E, Ono M, Oohashi T, Kuboki T, Takarada T

    Biochemical and biophysical research communications   516 ( 4 )   1229 - 1233   2019.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Global gene deletion studies have established that Runt-related transcription factor-2 (Runx2) is essential during skeletogenesis for osteoblastic differentiation in both intramembranous and endochondral ossification processes. However, the postnatal significance of Runx2 in vivo is poorly understood because a global Runx2 deletion causes perinatal lethality. In this study, we generated tamoxifen-induced Runx2 global deficient mice by crossing Runx2(flox) mice with ROSA26-CreER(T2) mice (Rosa26-CreER(T2); Runx2(flox/flox)). Four-week-old mice were intraperitoneally treated with tamoxifen for five consecutive days, sacrificed, and analyzed six weeks after tamoxifen administration. Deletion of Runx2 led to low bone mass, which is associated with decreased bone formation and bone resorption as well as excessive bone marrow adiposity. Collectively, postnatal Runx2 absolutely plays an important role in maintaining the homeostasis of bone tissues not only in bone mass, but also in the bone marrow environment. (C) 2019 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2019.07.014

    Web of Science

    PubMed

    researchmap

  • Runx2 function in cells of neural crest origin during intramembranous ossification. Reviewed International journal

    Shirai Y, Kawabe K, Tosa I, Tsukamoto S, Yamada D, Takarada T

    Biochemical and biophysical research communications   509 ( 4 )   1028 - 1033   2019.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Runt-related transcription factor 2 (Runx2), also known as core binding factor 1 (Cbfa1), is a multifunctional transcription factor and an essential master gene controlling osteoblast differentiation. We previously demonstrated the in vivo functions of Runx2 in mesoderm-derived cells. However, no studies have been conducted on Runx2 function during the differentiation of neural crest (NC)-derived cells in vivo. Wingless-type MMTV integration site family member 1 (Wnt1) is expressed in the NC, and Wnt1-Cre efficiently targets craniofacial NC-derived cells. Runx2 deficiency in cells of the Wnt1 lineage (referred henceforth as Runx2wnt1-/- within mice) resulted in defective ossification in certain regions, primarily in the anterior half of the craniofacial bones, including the frontal bone, jugal bone, squamous temporal bone, mandible, maxilla, and nasal bone. The skeletal analysis also revealed that heterozygous Runx2wnt1+/- embryos had an impaired closure of the frontal bone at the metopic suture and lacked the secondary palate in spite of otherwise normal ossification. This result suggests that ossification at the central part of the frontal bone is more dependent on Runx2 expression in comparison to other areas. These results indicate that Runx2 is indispensable not only for mesoderm-derived cells but also for NC-derived cells to differentiate during intramembranous ossification after migration to their destination from the neural plate border. Moreover, this implies that there are different levels of dependency on Runx2 expression for successful ossification between NC-derived cells that have migrated to different locations.

    DOI: 10.1016/j.bbrc.2019.01.059

    PubMed

    researchmap

  • Design, synthesis, and biological evaluation of radioiodinated benzo[d]imidazole-quinoline derivatives for platelet-derived growth factor receptor β (PDGFRβ) imaging. Reviewed International journal

    Effendi N, Mishiro K, Takarada T, Yamada D, Nishii R, Shiba K, Kinuya S, Odani A, Ogawa K

    Bioorganic & medicinal chemistry   27 ( 2 )   383 - 393   2019.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Several malignant tumors and fibrotic diseases are associated with PDGFRβ overexpression and excessive signaling, making this receptor attractive for molecular targeting and imaging approaches. A series of benzo[d]imidazole-quinoline derivatives were designed and synthesized to develop radioiodinated compounds as PDGFRβ-specific imaging probes. The structure activity relationship (SAR) evaluation of the designed compounds was performed. Among them, 2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]-8-(piperazin-1-yl)quinoline (5a) and 4-{2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]quinolin-8-yl}morpholine (5d) exhibited a relatively high PDGFRβ-TK inhibitory potency, whereas iodinated 5a derivative 5-iodo-2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]-8-(piperazin-1-yl)quinoline (8) exhibited a superior inhibitory potency as PDGFRβ inhibitor than iodinated 5d derivative 4-{5-iodo-2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]quinolin-8-yl}morpholine (11). Furthermore, [125I]8 and [125I]11 were synthesized and evaluated for PDGFRβ radioligand ability, both in vitro and in vivo. Cellular uptake experiments showed that [125I]8 had a higher uptake in BxPC3-luc cells as PDGFRβ-positive cells than [125I]11. Incubation of [125I]8 after pretreatment of PDGFRβ ligands significantly reduced the uptake of [125I]8. In biodistribution experiments using tumor-bearing mice, [125I]8 accumulation in the tumor 1 h postinjection was higher than that of the benzo[d]imidazol-quinoline derivative [125I]IIQP, used in our previous research. These results indicate that [125I]8 could be a promising PDGFRβ imaging agent. Although its clinical application requires further structural modifications, the results obtained in this research may be useful for the development of PDGFRβ-specific radioligands.

    DOI: 10.1016/j.bmc.2018.12.016

    PubMed

    researchmap

  • Inhibition of the glutamine transporter SNAT1 confers neuroprotection in mice by modulating the mTOR-autophagy system. Reviewed International journal

    Yamada D, Kawabe K, Tosa I, Tsukamoto S, Nakazato R, Kou M, Fujikawa K, Nakamura S, Ono M, Oohashi T, Kaneko M, Go S, Hinoi E, Yoneda Y, Takarada T

    Communications biology   2   346 - 346   2019

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)  

    The pathophysiological role of mammalian target of rapamycin complex 1 (mTORC1) in neurodegenerative diseases is established, but possible therapeutic targets responsible for its activation in neurons must be explored. Here we identified solute carrier family 38a member 1 (SNAT1, Slc38a1) as a positive regulator of mTORC1 in neurons. Slc38a1 flox/flox and Synapsin I-Cre mice were crossed to generate mutant mice in which Slc38a1 was selectively deleted in neurons. Measurement of 2,3,5-triphenyltetrazolium chloride (TTC) or the MAP2-negative area in a mouse model of middle cerebral artery occlusion (MCAO) revealed that Slc38a1 deficiency decreased infarct size. We found a transient increase in the phosphorylation of p70S6k1 (pp70S6k1) and a suppressive effect of rapamycin on infarct size in MCAO mice. Autophagy inhibitors completely mitigated the suppressive effect of SNAT1 deficiency on neuronal cell death under in vitro stroke culture conditions. These results demonstrate that SNAT1 promoted ischemic brain damage via mTOR-autophagy system.

    DOI: 10.1038/s42003-019-0582-4

    PubMed

    researchmap

  • RUNX2 Promotes Malignant Progression in Glioma. Reviewed International journal

    Yamada D, Fujikawa K, Kawabe K, Furuta T, Nakada M, Takarada T

    Neurochemical research   43 ( 11 )   2047 - 2054   2018.11

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)  

    Glioblastoma (GBM) is the most aggressive and lethal form of brain tumor. However, therapeutic strategies against malignant gliomas have not been completely established. Runt-related transcription factor 2 (Runx2) is an essential gene for skeletal development but its regulatory role in the malignant progression of glioma remains unclear. Here we investigated expression levels of RUNX2 in glioma tissues and its regulatory effects on aberrant growth of glioma cells. RUNX2 mRNA levels were higher in GBM tissues than that of normal brains or low-grade gliomas. RUNX2 protein was detected in five out of seven human GBM cell lines and its level was positively correlated with proliferative capacity. Stable transduction of dominant-negative Runx2 in rat-derived C6 glioma cells not only inhibited the promoter activity containing Runx2 response element, but also decreased mRNA expression levels of Runx2 target genes, such as Mmp13 and Spp1, as well as the proliferative capacity. Furthermore, transient introduction of Runx2-targeted siRNAs into C6 glioma cells significantly decreased mRNA expression levels of Mmp13 and Spp1 and the proliferative capacity. Furthermore, Runx2 knockdown suppressed both Ccnd1 mRNA expression and activation of the Ccnd1 promoter by forskolin, a PKA-activating reagent, in C6 glioma cells. Our results demonstrate that cross-talk between cAMP/PKA signaling and RUNX2 promotes a malignant phenotype of glioma cells.

    DOI: 10.1007/s11064-018-2626-4

    PubMed

    researchmap

  • Radiobrominated benzimidazole-quinoline derivatives as Platelet-derived growth factor receptor beta (PDGFR beta) imaging probes Reviewed International journal

    Effendi Nurmaya, Mishiro Kenji, Takarada Takeshi, Makino Akira, Yamada Daisuke, Kitamura Yoji, Shiba Kazuhiro, Kiyono Yasushi, Odani Akira, Ogawa Kazuma

    SCIENTIFIC REPORTS   8 ( 1 )   10369 - 10369   2018.7

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Platelet-derived growth factor receptor beta (PDGFRβ) affects in numerous human cancers and has been recognized as a promising molecular target for cancer therapies. The overexpression of PDGFRβ could be a biomarker for cancer diagnosis. Radiolabeled ligands having high affinity for the molecular target could be useful tools for the imaging of overexpressed receptors in tumors. In this study, we aimed to develop radiobrominated PDGFRβ ligands and evaluate their effectiveness as PDGFRβ imaging probes. The radiolabeled ligands were designed by modification of 1-{2-[5-(2-methoxyethoxy)-1H- benzo[d]imidazol-1-yl]quinolin-8-yl}piperidin-4-amine (1), which shows selective inhibition profile toward PDGFRβ. The bromine atom was introduced directly into C-5 of the quinoline group of 1, or indirectly by the conjugation of 1 with the 3-bromo benzoyl group. [77Br]1-{5-Bromo-2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]quinoline-8-yl}piperidin-4-amine ([77Br]2) and [77Br]-N-3-bromobenzoyl-1-{2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]quinolin-8-yl}-piperidin-4-amine ([77Br]3) were prepared using a bromodestannylation reaction. In a cellular uptake study, [77Br]2 and [77Br]3 more highly accumulatd in BxPC3-luc cells (PDGFRβ-positive) than in MCF7 cells (PDGFRβ-negative), and their accumulation was significantly reduced by pretreatment with inhibitors. In biodistribution experiments, [77Br]2 accumulation was higher than [77Br]3 accumulation at 1 h postinjection. These findings suggest that [76Br]2 is more promising for positron emission tomography (PET) imaging of PDGFRβ than [76Br]3.

    DOI: 10.1038/s41598-018-28529-0

    Web of Science

    PubMed

    researchmap

  • Synthesis and evaluation of radioiodinated 1-{2-[5-(2-methoxyethoxy)-1H-benzo[d]imidazol-1-yl]quinolin-8-yl}piperidin-4-amine derivatives for platelet-derived growth factor receptor beta (PDGFR beta) imaging Reviewed

    Nurmaya Effendi, Kazuma Ogawa, Kenji Mishiro, Takeshi Takarada, Daisuke Yamada, Yoji Kitamura, Kazuhiro Shiba, Takehiko Maeda, Akira Odani

    BIOORGANIC & MEDICINAL CHEMISTRY   25 ( 20 )   5576 - 5585   2017.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:PERGAMON-ELSEVIER SCIENCE LTD  

    Platelet-derived growth factor receptor beta (PDGFR beta) is a transmembrane tyrosine kinase receptor and it is upregulated in various malignant tumors. Radiolabeled PDGFR beta inhibitors can be a convenient tool for the imaging of tumors overexpressing PDGFR beta. In this study, [I-125]-1-{5-iodo-2-[5-(2-methoxyethoxy)1H-benzo[d]imidazol-1-yl]quinoline-8-yl} piperidin-4-amine ([I-125]IIQP) and [I-125]-N-3-iodobenzoyl-1{2-[5-(2-methoxyethoxy)-1H-benzo[d] imidazol-1-yl] quinolin-8-yl}-piperidin-4-amine ([I-125]IB-IQP) were designed and synthesized, and their potential as PDGFR beta imaging agents was evaluated. In cellular uptake experiments, [I-125] IIQP and [I-125] IB-IQP showed higher uptake by PDGFR beta-positive cells than by PDGFR beta-negative cells, and the uptake in PDGFR beta-positive cells was inhibited by co-culture with PDGFR beta ligands. The biodistribution of both radiotracers in normal mice exhibited hepatobiliary excretion as the main route. In mice inoculated with BxPC3-luc (PDGFR beta-positive), the tumor uptake of radioactivity at 1 h after the injection of [I-125] IIQP was significantly higher than that after the injection of [I-125] IB-IQP. These results indicated that [I-125] IIQP can be a suitable PDGFR beta imaging agent. However, further modification of its structure will be required to obtain a more appropriate PDGFR beta-targeted imaging agent with a higher signal/noise ratio. (C) 2017 Published by Elsevier Ltd.

    DOI: 10.1016/j.bmc.2017.08.025

    Web of Science

    researchmap

  • Disruption of Bmal1 Impairs Blood-Brain Barrier Integrity via Pericyte Dysfunction Reviewed

    Ryota Nakazato, Kenji Kawabe, Daisuke Yamada, Shinsuke Ikeno, Michihiro Mieda, Shigeki Shimba, Eiichi Hinoi, Yukio Yoneda, Takeshi Takarada

    JOURNAL OF NEUROSCIENCE   37 ( 42 )   10052 - 10062   2017.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:SOC NEUROSCIENCE  

    Circadian rhythm disturbances are well established in neurological diseases. However, how these disruptions cause homeostatic imbalances remains poorly understood. Brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1) is a major circadian clock transcriptional activator, and Bmal1 deficiency in male Bmal1(nestin)(-/-) mice induced marked astroglial activation without affecting the number of astrocytes in the brain and spinal cord. Bmal1 deletion caused blood-brain barrier (BBB) hyperpermeability with an age-dependent loss of pericyte coverage of blood vessels in the brain. Using Nestin-green fluorescent protein (GFP) transgenic mice, we determined that pericytes are Nestin-GFP(+) in the adult brain. Bmal1 deletion caused Nestin-GFP(+) pericyte dysfunction, including the downregulation of platelet-derived growth factor receptor beta (PDGFR beta), a protein necessary for maintaining BBB integrity. Knockdown of Bmal1 downregulatedPDGFR beta transcription in the brain pericyte cell line. Thus, the circadian clock component Bmal1 maintains BBB integrity via regulating pericytes.

    DOI: 10.1523/JNEUROSCI.3639-16.2017

    Web of Science

    PubMed

    researchmap

  • Bone Resorption Is Regulated by Circadian Clock in Osteoblasts Reviewed

    Takeshi Takarada, Cheng Xu, Hiroki Ochi, Ryota Nakazato, Daisuke Yamada, Saki Nakamura, Ayumi Kodama, Shigeki Shimba, Michihiro Mieda, Kazuya Fukasawa, Kakeru Ozaki, Takashi Iezaki, Koichi Fujikawa, Yukio Yoneda, Rika Numano, Akiko Hida, Hajime Tei, Shu Takeda, Eiichi Hinoi

    JOURNAL OF BONE AND MINERAL RESEARCH   32 ( 4 )   872 - 881   2017.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY  

    We have previously shown that endochondral ossification is finely regulated by the Clock system expressed in chondrocytes during postnatal skeletogenesis. Here we show a sophisticated modulation of bone resorption and bone mass by the Clock system through its expression in bone-forming osteoblasts. Brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1) and Period1 (Per1) were expressed with oscillatory rhythmicity in the bone in vivo, and circadian rhythm was also observed in cultured osteoblasts of Per1::luciferase transgenic mice. Global deletion of murine Bmal1, a core component of the Clock system, led to a low bone mass, associated with increased bone resorption. This phenotype was recapitulated by the deletion of Bmal1 in osteoblasts alone. Co-culture experiments revealed that Bmal1-deficient osteoblasts have a higher ability to support osteoclastogenesis. Moreover, 1,25-dihydroxyvitamin D-3 [1,25(OH)(2)D-3]-induced receptor activator of nuclear factor B ligand (Rankl) expression was more strongly enhanced in both Bmal1-deficient bone and cultured osteoblasts, whereas overexpression of Bmal1/Clock conversely inhibited it in osteoblasts. These results suggest that bone resorption and bone mass are regulated at a sophisticated level by osteoblastic Clock system through a mechanism relevant to the modulation of 1,25(OH)(2)D-3-induced Rankl expression in osteoblasts. (c) 2017 American Society for Bone and Mineral Research.

    DOI: 10.1002/jbmr.3053

    Web of Science

    PubMed

    researchmap

  • The Intrinsic Microglial Clock System Regulates Interleukin-6 Expression Reviewed

    Ryota Nakazato, Shogo Hotta, Daisuke Yamada, Miki Kou, Saki Nakamura, Yoshifumi Takahata, Hajime Tei, Rika Numano, Akiko Hida, Shigeki Shimba, Michihiro Mieda, Eiichi Hinoi, Yukio Yoneda, Takeshi Takarada

    GLIA   65 ( 1 )   198 - 208   2017.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Similar to neurons, microglia have an intrinsic molecular clock. The master clock oscillator Bmal1 modulates interleukin-6 upregulation in microglial cells exposed to lipopolysaccharide. Bmal1 can play a role in microglial inflammatory responses. We previously demonstrated that gliotransmitter ATP induces transient expression of the clock gene Period1 via P2X7 purinergic receptors in cultured microglia. In this study, we further investigated mechanisms underlying the regulation of pro-inflammatory cytokine production by clock molecules in microglial cells. Several clock gene transcripts exhibited oscillatory diurnal rhythmicity in microglial BV-2 cells. Real-time luciferase monitoring also showed diurnal oscillatory luciferase activity in cultured microglia from Per1::Luciferase transgenic mice. Lipopolysaccharide (LPS) strongly induced the expression of pro-inflammatory cytokines in BV-2 cells, whereas an siRNA targeting Brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1), a core positive component of the microglial molecular clock, selectively inhibited LPS-induced interleukin-6 (IL-6) expression. In addition, LPS-induced IL-6 expression was attenuated in microglia from Bmal1-deficient mice. This phenotype was recapitulated by pharmacological disruption of oscillatory diurnal rhythmicity using the synthetic Rev-Erb agonist SR9011. Promoter analysis of the Il6 gene revealed that Bmal1 is required for LPS-induced IL-6 expression in microglia. Mice conditionally Bmal1 deficient in cells expressing CD11b, including microglia, exhibited less potent upregulation of Il6 expression following middle cerebral artery occlusion compared with that in control mice, with a significant attenuation of neuronal damage. These results suggest that the intrinsic microglial clock modulates the inflammatory response, including the positive regulation of IL-6 expression in a particular pathological situation in the brain.

    DOI: 10.1002/glia.23087

    Web of Science

    PubMed

    researchmap

  • The Novel Monoclonal Antibody 9F5 Reveals Expression of a Fragment of GPNMB/Osteoactivin Processed by Furin-like Protease(s) in a Subpopulation of Microglia in Neonatal Rat Brain Reviewed

    Kohichi Kawahara, Hiroshi Hirata, Kengo Ohbuchi, Kentaro Nishi, Akira Maeda, Akihiko Kuniyasu, Daisuke Yamada, Takehiko Maeda, Akihiko Tsuji, Makoto Sawada, Hitoshi Nakayama

    GLIA   64 ( 11 )   1938 - 1961   2016.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    To differentiate subtypes of microglia (MG), we developed a novel monoclonal antibody, 9F5, against one subtype (type 1) of rat primary MG. The 9F5 showed high selectivity for this cell type in Western blot and immunocytochemical analyses and no cross-reaction with rat peritoneal macrophages (M phi). We identified the antigen molecule for 9F5: the 50- to 70-kDa fragments of rat glycoprotein nonmetastatic melanoma protein B (GPNMB)/osteoactivin, which started at Lys(170). In addition, 9F5 immunoreactivity with GPNMB depended on the activity of furin-like protease(s). More important, rat type 1 MG expressed the GPNMB fragments, but type 2 MG and M phi did not, although all these cells expressed mRNA and the full-length protein for GPNMB. These results suggest that 9F5 reactivity with MG depends greatly on cleavage of GPNMB and that type 1 MG, in contrast to type 2 MG and M phi, may have furin-like protease(s) for GPNMB cleavage. In neonatal rat brain, amoeboid 9F5+ MG were observed in specific brain areas including forebrain subventricular zone, corpus callosum, and retina. Double-immunostaining with 9F5 antibody and anti-Iba1 antibody, which reacts with MG throughout the CNS, revealed that 9F5+ MG were a portion of Iba1+ MG, suggesting that MG subtype(s) exist in vivo. We propose that 9F5 is a useful tool to discriminate between rat type 1 MG and other subtypes of MG/M phi and to reveal the role of the GPNMB fragments during developing brain.

    DOI: 10.1002/glia.23034

    Web of Science

    PubMed

    researchmap

  • Plexin A1 signaling confers malignant phenotypes in lung cancer cells Reviewed

    Daisuke Yamada, Satoshi Watanabe, Kohichi Kawahara, Takehiko Maeda

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   480 ( 1 )   75 - 80   2016.11

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Aberrant changes to several signaling pathways because of genetic mutations or increased cytokine production are critical for tumor cells to become malignant. Semaphorin 3A (SEMA3A) acts as a bivalent factor that suppresses or promotes tumor development in different pathological backgrounds. Previously, we showed that SEMA3A positively regulated the proliferative and glycolytic activities of mouse derived Lewis lung carcinoma (LLC) cells. Plexins A1-A4 (PLXNA1-PLXNA4) are SEMA3A receptors; however, it is not known which subtype is critical for oncogenic SEMA3A signaling. We used LLC cells to investigate the role of PLXNA1 in oncogenic SEMA3A signaling. Using short hairpin RNA-mediated knockdown, we investigated the effects of constitutive inhibition of Plxna1 on cell proliferation, metabolic dependency, and epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) sensitivity. We found that Plxna1 knockdown did not affect apoptosis but resulted in decreased cell proliferation and reductions in mRNA expression levels of proliferation-marker genes, such as Ccnd1, Pcna, and Myc. In addition, we found decreased mRNA expression levels of glycolysis-associated genes, such as Pkm2 and Ldha, and decreased lactate production. In contrast, we found no changes in the mRNA expression levels of oxidative phosphorylation-associated genes, such as Cycs, Cox5a, and Atp5g1. We found that Plxna1 knockdown conferred resistance to glucose starvation but increased cytotoxicity to oligomycin. Plxna1 or Sema3a knockdown caused an increased sensitivity to the EGFR-TKIs gefitinib and erlotinib, in Lewis lung carcinoma (LLC) cells. These findings demonstrate that PLXNA1 mediates the acquisition of malignant phenotypes induced by autocrine SEMA3A signaling. (C) 2016 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2016.10.006

    Web of Science

    PubMed

    researchmap

  • Autocrine Semaphorin3A signaling is essential for the maintenance of stem-like cells in lung cancer Reviewed

    Daisuke Yamada, Kensuke Takahashi, Kohichi Kawahara, Takehiko Maeda

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   480 ( 3 )   375 - 379   2016.11

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Cancer stem-like cells (CSCs) exist in tumor tissues composed of heterogeneous cell population and are characterized by their self-renewal capacity and tumorigenicity. Many studies demonstrate that eradication of CSCs prevents development and recurrences of tumor; yet, molecules critical for the maintenance of CSCs have not been completely understood. We previously reported that Semaphorin3A (Sema3a) knockdown suppressed the tumorigenicity and proliferative capacity of Lewis lung carcinoma (LLC) cells. Therefore, we identified Sema3a as an essential factor for the establishment or maintenance of CSCs derived from LLC (LLC-stem cell). shRNA against Sema3a was introduced into LLC cells to establish a LLC-stem cell line and its effects on tumorigenesis, sphere formation, and mTORC1 activity were tested. Sema3a knockdown completely abolished tumorigenicity and the sphere-formation and self-renewal ability of LLC-stem cells. The Sema3a knockdown was also associated with decreased expression of mRNA for stem cell markers. The self-renewal ability abolished by Sema3a knockdown could not be recovered by exogenous addition of recombinant SEMA3A. In addition, the activity of mammalian target of rapamycin complex 1 (mTORC1) and the expression of its substrate p70S6K1 were also decreased. These results demonstrate that Sema3a is a potential therapeutic target in eradication of CSCs. (C) 2016 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2016.10.057

    Web of Science

    PubMed

    researchmap

  • Cancer pain relief achieved by disrupting tumor-driven semaphorin 3A signaling in mice Reviewed

    Takehiko Maeda, Daisuke Yamada, Kohichi Kawahara

    NEUROSCIENCE LETTERS   632   147 - 151   2016.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ELSEVIER IRELAND LTD  

    Cancer-induced bone pain (CIBP) is the most common pain arising from cancer and is inadequately managed with current standard therapeutics. While the etiology of CIBP remains to be fully elucidated, increasing evidence suggests that CIBP is uniquely complex. We tested whether semaphorin 3A (Sema3A) signals were involved in the development of CIBP in mice. The mouse model employed for CIBP - mice inoculated with Lewis lung carcinoma (LLC) cells injected into the femur intramedullary space - showed progressive decline in the weight bearing of the ipsilateral hind limb. The LLC cell inoculation resulted in a progressive increase in Sema3A mRNA expression over time and an increase in the number of Sema3A-immunoreactive cells in the ipsilateral femur. To define the role of Sema3A in development of CIBP, we employed a lentiviral expression system to establish a stable LLC cell line expressing scrambled shRNA for the control group (LLC/scramble) and shRNAs directed against Sema3A mRNA for the loss-of-function group (LLC/shSema3A). Inoculation of LLC/shSema3A did not cause upregulation of Sema3A mRNA expression and proliferation of the inoculated cells in the femur compared to that in mice inoculated with LLC/scramble. Mice inoculated with LLC/shSema3A, but not LLC/scramble, showed an attenuation of the significant decline in the weight bearing of the ipsilateral hind paw. Our findings indicate that Sema3A serves as a potential therapeutic target for CIBP. (C) 2016 Elsevier Ireland Ltd. All rights reserved.

    DOI: 10.1016/j.neulet.2016.08.060

    Web of Science

    PubMed

    researchmap

  • Therapeutic Strategy for Targeting Aggressive Malignant Gliomas by Disrupting Their Energy Balance Reviewed

    Ahmed M. Hegazy, Daisuke Yamada, Masahiko Kobayashi, Susumu Kohno, Masaya Ueno, Mohamed A. E. Ali, Kumiko Ohta, Yuko Tadokoro, Yasushi Ino, Tomoki Todo, Tomoyoshi Soga, Chiaki Takahashi, Atsushi Hirao

    JOURNAL OF BIOLOGICAL CHEMISTRY   291 ( 41 )   21496 - 21509   2016.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Although abnormal metabolic regulation is a critical determinant of cancer cell behavior, it is still unclear how an altered balance between ATP production and consumption contributes to malignancy. Here we show that disruption of this energy balance efficiently suppresses aggressive malignant gliomas driven by mammalian target of rapamycin complex 1 (mTORC1) hyperactivation. In a mouse glioma model, mTORC1 hyperactivation induced by conditional Tsc1 deletion increased numbers of glioma-initiating cells (GICs) in vitro and in vivo. Metabolic analysis revealed that mTORC1 hyperactivation enhanced mitochondrial biogenesis, as evidenced by elevations in oxygen consumption rate and ATP production. Inhibition of mitochondrial ATP synthetase was more effective in repressing sphere formation by Tsc1-deficient glioma cells than that by Tsc1-competent glioma cells, indicating a crucial function for mitochondrial bioenergetic capacity in GIC expansion. To translate this observation into the development of novel therapeutics targeting malignant gliomas, we screened drug libraries for small molecule compounds showing greater efficacy in inhibiting the proliferation/survival of Tsc1-deficient cells compared with controls. We identified several compounds able to preferentially inhibit mitochondrial activity, dramatically reducing ATP levels and blocking glioma sphere formation. In human patient-derived glioma cells, nigericin, which reportedly suppresses cancer stem cell properties, induced AMPK phosphorylation that was associated with mTORC1 inactivation and induction of autophagy and led to a marked decrease in sphere formation with loss of GIC marker expression. Furthermore, malignant characteristics of human glioma cells were markedly suppressed by nigericin treatment in vivo. Thus, targeting mTORC1-driven processes, particularly those involved in maintaining a cancer cell's energy balance, may be an effective therapeutic strategy for glioma patients.

    DOI: 10.1074/jbc.M116.734756

    Web of Science

    PubMed

    researchmap

  • mTORC1 is a critical mediator of oncogenic Semaphorin3A signaling Reviewed

    Daisuke Yamada, Kohichi Kawahara, Takehiko Maeda

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   476 ( 4 )   475 - 480   2016.8

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Aberration of signaling pathways by genetic mutations or alterations in the surrounding tissue environments can result in tumor development or metastasis. However, signaling molecules responsible for these processes have not been completely elucidated. Here, we used mouse Lewis lung carcinoma cells (LLC) to explore the mechanism by which the oncogenic activity of Semaphorin3A (Sema3A) signaling is regulated. Sema3A knockdown by shRNA did not affect apoptosis, but decreased cell proliferation in LLCs; both the mammalian target of rapamycin complex 1 (mTORC1) level and glycolytic activity were also decreased. In addition, Sema3A knockdown sensitized cells to inhibition of oxidative phosphorylation by oligomycin, but conferred resistance to decreased cell viability induced by glucose starvation. Furthermore, recombinant SEMA3A rescued the attenuation of cell proliferation and glycolytic activity in LLCs after Sema3A knockdown, whereas mTORC1 inhibition by rapamycin completely counteracted this effect. These results demonstrate that Sema3A signaling exerts its oncogenic effect by promoting an mTORC1-mediated metabolic shift from oxidative phosphorylation to aerobic glycolysis. (C) 2016 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2016.05.147

    Web of Science

    PubMed

    researchmap

  • Tumor cell-derived secretory factor downregulates Semaphorin-3a in osteoblasts by activating mammalian target of rapamycin pathway Reviewed

    Daisuke Yamada, Kohichi Kawahara, Masanobu Ozaki, Takehiko Maeda

    BIOSCIENCE BIOTECHNOLOGY AND BIOCHEMISTRY   80 ( 5 )   942 - 944   2016.5

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:TAYLOR & FRANCIS LTD  

    We found that conditioned medium derived from Lewis Lung Carcinoma cells down-regulated Semaphorin3a (Sema3a) mRNA expression and increased the activity of mammalian target of rapamycin complex 1 (mTORC1) in osteoblast-like MC3T3-E1 cells. Furthermore, mTORC1 inhibition with rapamycin counteracted the effect of conditioned media on Sema3a mRNA expression. These results suggest that tumor cells decrease Sema3a mRNA expression in osteoblast in an mTORC1-dependent manner.

    DOI: 10.1080/09168451.2015.1136881

    Web of Science

    PubMed

    researchmap

  • Strong therapeutic potential of gamma-secretase inhibitor MRK003 for CD44-high and CD133-low glioblastoma initiating cells Reviewed

    Shingo Tanaka, Mitsutoshi Nakada, Daisuke Yamada, Ichiro Nakano, Tomoki Todo, Yasushi Ino, Takayuki Hoshii, Yuko Tadokoro, Kumiko Ohta, Mohamed A. E. Ali, Yutaka Hayashi, Jun-ichiro Hamada, Atsushi Hirao

    JOURNAL OF NEURO-ONCOLOGY   121 ( 2 )   239 - 250   2015.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:SPRINGER  

    The Notch signal regulates both cell viability and apoptosis, and maintains stemness of various cancers including glioblastoma (GBM). Although Notch signal inhibition may be an effective strategy in treating GBM initiating cells (GICs), its applicability to the different subtypes of GBM remains unclear. Here, we analyzed the effectiveness of MRK003, a preclinical gamma-secretase inhibitor, on GICs. Nine patient-derived GICs were treated by MRK003, and its efficacy on cell viability, apoptosis, sphere forming ability and Akt expression level which might be related to Notch downstream and be greatly important signals in GBM was evaluated. MRK003 suppressed viability and sphere-formation ability, and induced apoptosis in all GICs in varying doses of MRK003. Based on their sensitivities to MRK003, the nine GICs were divided into "relatively sensitive'' and "relatively resistant'' GICs. Sensitivity to MRK003 was associated with its inhibitory effect on Akt pathway. Transgenic expression of the myristoylated Akt vector in relatively sensitive GICs partially rescued the effect of MRK003, suggesting that the effect of MRK003 was, at least in part, mediated through inhibition of the Akt pathway. These GICs were differentiated by the expression of CD44 and CD133 with flow cytometric analysis. The relatively sensitive GICs are CD44-high and CD133-low. The IC50 of MRK003 in a set of GICs exhibited a negative correlation with CD44 and positive correlation with CD133. Collectively, MRK003 is partially mediated by the Akt pathway and has strong therapeutic potential for CD44-high and CD133-low GICs.

    DOI: 10.1007/s11060-014-1630-z

    Web of Science

    PubMed

    researchmap

  • Loss of Tsc1 accelerates malignant gliomagenesis when combined with oncogenic signals. International journal

    Daisuke Yamada, Takayuki Hoshii, Shingo Tanaka, Ahmed M Hegazy, Masahiko Kobayashi, Yuko Tadokoro, Kumiko Ohta, Masaya Ueno, Mohamed A E Ali, Atsushi Hirao

    Journal of biochemistry   155 ( 4 )   227 - 33   2014.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Glioblastomas frequently harbour genetic lesions that stimulate the activity of mammalian target of rapamycin complex 1 (mTORC1). Loss of heterozygosity of tuberous sclerosis complex 1 (TSC1) or TSC2, which together form a critical negative regulator of mTORC1, is also seen in glioblastoma; however, it is not known how loss of the TSC complex affects the development of malignant gliomas. Here we investigated the role of Tsc1 in gliomagenesis in mice. Tsc1 deficiency up-regulated mTORC1 activity and suppressed the proliferation of neural stem/progenitor cells (NSPCs) in a serial neurosphere-forming assay, suggesting that Tsc1-deficient NSPCs have defective self-renewal activity. The neurosphere-forming capacity of Tsc1-deficient NSPCs was restored by p16(Ink4a)p19(Arf) deficiency. Combined Tsc1 and p16(Ink4a)p19(Arf) deficiency in NSPCs did not cause gliomagenesis in vivo. However, in a glioma model driven by an active mutant of epidermal growth factor receptor (EGFR), EGFRvIII, loss of Tsc1 resulted in an earlier onset of glioma development. The mTORC1 hyperactivation by Tsc1 deletion accelerated malignant phenotypes, including increased tumour mass and enhanced microvascular formation, leading to intracranial haemorrhage. These data demonstrate that, although mTORC1 hyperactivation itself may not be sufficient for gliomagenesis, it is a potent modifier of glioma development when combined with oncogenic signals.

    DOI: 10.1093/jb/mvt112

    PubMed

    researchmap

  • Osteoclastogenesis is negatively regulated by D-serine produced by osteoblasts Reviewed

    Takeshi Takarada, Mika Takarada-Iemata, Yoshifumi Takahata, Daisuke Yamada, Tomomi Yamamoto, Yukari Nakamura, Eiichi Hinoi, Yukio Yoneda

    JOURNAL OF CELLULAR PHYSIOLOGY   227 ( 10 )   3477 - 3487   2012.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    We have shown the functional expression by chondrocytes of serine racemase (SR) which is responsible for the synthesis of D-serine (Ser) from L-Ser in cartilage. In this study, we evaluated the possible functional expression of SR by bone-forming osteoblasts and bone-resorbing osteoclasts. Expression of SR mRNA was seen in osteoblasts localized at the cancellous bone surface in neonatal rat tibial sections and in cultured rat calvarial osteoblasts endowed to release D-Ser into extracellular medium, but not in cultured osteoclasts differentiated from murine bone marrow progenitor cells. Sustained exposure to D-Ser failed to significantly affect alkaline phosphatase activity and Ca2+ accumulation in cultured osteoblasts, but significantly inhibited differentiation and maturation in a concentration-dependent manner at a concentration range of 0.11?mM without affecting cellular survival in cultured osteoclasts. By contrast, L-Ser promoted osteoclastic differentiation in a manner sensitive to the inhibition by D-Ser. Matured osteoclasts expressed mRNA for the amino acid transporter B0,+ (ATB0,+) and the system alanine, serine, and cysteine amino acid transporter-2 (ASCT2), which are individually capable of similarly incorporating extracellular L- and D-Ser. Knockdown of these transporters by siRNA prevented both the promotion by L-Ser and the inhibition by D-Ser of osteoclastic differentiation in pre-osteoclastic RAW264.7 cells. These results suggest that D-Ser may play a pivotal role in osteoclastogenesis through a mechanism related to the incorporation mediated by both ATB0,+ and ASCT2 of serine enantiomers in osteoclasts after the synthesis and subsequent release from adjacent osteoblasts. J. Cell. Physiol. 227: 34773487, 2012. (C) 2012 Wiley Periodicals, Inc.

    DOI: 10.1002/jcp.24048

    Web of Science

    PubMed

    researchmap

  • NKX2.2 Suppresses Self-Renewal of Glioma-Initiating Cells Reviewed

    Teruyuki Muraguchi, Shingo Tanaka, Daisuke Yamada, Akira Tamase, Mitsutoshi Nakada, Hideo Nakamura, Takayuki Hoshii, Takako Ooshio, Yuko Tadokoro, Kazuhito Naka, Yasushi Ino, Tomoki Todo, Jun-ichi Kuratsu, Hideyuki Saya, Jun-ichiro Hamada, Atsushi Hirao

    CANCER RESEARCH   71 ( 3 )   1135 - 1145   2011.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER ASSOC CANCER RESEARCH  

    Glioblastoma (GBM) is the most aggressive and destructive form of brain cancer. Animal models that can unravel the mechanisms underlying its progression are needed to develop rational and effective molecular therapeutic approaches. In this study, we report the development of mouse models for spontaneous gliomas representing distinct progressive stages of disease that are governed by defined genetic alterations. Neural stem/progenitor cell (NPC)-specific constitutive Ras activation in vivo plus p53 deficiency led to development of primarily anaplastic astrocytoma (grade III), whereas combined loss of p53 plus p16(Ink4a)/p19(Arf) led to development of GBM (grade IV) at 100% penetrance within 6 weeks. These glioma models showed enhanced stem cell properties (stemness) accompanied by malignant progression. Notably, we determined that, in our models and in human specimens, downregulation of the homeodomain transcription factor NKX2.2, which is essential for oligodendroglial differentiation, was correlated with increased tumor malignancy. NKX2.2 overexpression by GBM-derived glioma-initiating cells (GIC) induced oligodendroglial differentiation and suppressed self-renewal capacity. By contrast, Nkx2.2 downregulation in mouse NPCs accelerated GBM formation. Importantly, the inhibitory effects of NXK2.2 on GIC self-renewal were conserved in human cells. Thus, our mouse models offer pathobiologically significant advantages to investigate the nature of brain tumors, with improved opportunities to develop novel mechanism-based therapeutic approaches. Cancer Res; 71(3); 1135-45. (C) 2010 AACR.

    DOI: 10.1158/0008-5472.CAN-10-2304

    Web of Science

    PubMed

    researchmap

▼display all

MISC

  • ヒトiPS細胞由来肢芽間葉系細胞を用いた骨再建法の開発

    佐藤 浩平, 高尾 知佳, 中田 英二, 藤澤 佑樹, 山田 大祐, 上原 健敬, 藤原 智洋, 依光 正則, 国定 俊之, 尾崎 敏文, 宝田 剛志

    日本整形外科学会雑誌   97 ( 8 )   S1868 - S1868   2023.8

     More details

    Language:Japanese   Publisher:(公社)日本整形外科学会  

    researchmap

  • ヒトiPS細胞由来肢芽間葉系細胞の発生機序の検討

    高尾 知佳, 大曽根 達則, 山田 大祐, 中田 英二, 尾崎 敏文, 宝田 剛志

    日本整形外科学会雑誌   97 ( 8 )   S1599 - S1599   2023.8

     More details

    Language:Japanese   Publisher:(公社)日本整形外科学会  

    researchmap

  • 悪性末梢神経鞘腫瘍における治療標的PRRX1の同定と新規創薬開発の可能性

    たき平 将太, 山田 大祐, 岡本 真幸, 高尾 知佳, 中田 英二, 板野 拓人, 藤原 智洋, 国定 俊之, 尾崎 敏文, 宝田 剛志

    日本整形外科学会雑誌   97 ( 6 )   S1428 - S1428   2023.6

     More details

    Language:Japanese   Publisher:(公社)日本整形外科学会  

    researchmap

  • ヒトiPS細胞から誘導した肢芽間葉系細胞による軟骨シートの作製

    藤澤 佑樹, 高尾 知佳, 佐藤 正人, 豊田 恵利子, 山田 大祐, 中田 英二, 尾崎 敏文, 宝田 剛志

    移植   57 ( 4 )   362 - 362   2023.4

     More details

    Language:Japanese   Publisher:(一社)日本移植学会  

    researchmap

  • ヒト多能性幹細胞から誘導した肢芽間葉系細胞と、その拡大培養法の開発

    中田 英二, 山田 大祐, 高尾 知佳, たき平 将太, 藤原 智洋, 尾崎 敏文, 宝田 剛志

    移植   57 ( 4 )   363 - 363   2023.4

     More details

    Language:Japanese   Publisher:(一社)日本移植学会  

    researchmap

  • 悪性末梢神経鞘腫瘍におけるPRRX1の治療標的分子としての可能性

    たき平 将太, 中田 英二, 大曽根 達則, 山田 大祐, 高尾 知佳, 佐藤 浩平, 畑 利彰, 藤原 智洋, 国定 俊之, 宝田 剛志, 尾崎 敏文

    日本整形外科学会雑誌   97 ( 3 )   S1040 - S1040   2023.3

     More details

    Language:Japanese   Publisher:(公社)日本整形外科学会  

    J-GLOBAL

    researchmap

  • 悪性末梢神経鞘腫瘍におけるPRRX1の悪性化因子としての役割

    たき平 将太, 中田 英二, 山田 大祐, 片山 晴喜, 畑 利彰, 藤原 智洋, 高尾 知佳, 国定 俊之, 宝田 剛志, 尾崎 敏文

    日本レックリングハウゼン病学会学術大会プログラム・抄録集   14回   16 - 16   2023.2

     More details

    Language:Japanese   Publisher:日本レックリングハウゼン病学会  

    researchmap

  • ヒトiPS細胞から誘導した肢芽間葉系細胞による軟骨シートの開発

    高尾知佳, 佐藤正人, 豊田恵利子, 藤澤佑樹, 山田大祐, 中田英二, 尾崎敏文, 宝田剛志

    日本再生医療学会総会(Web)   22nd   2023

  • ヒト多能性幹細胞由来肢芽間葉系細胞を用いた軟骨分化誘導技術の開発

    山田大祐, 高尾知佳, 中村正裕, 戸口田淳也, 宝田剛志

    日本再生医療学会総会(Web)   22nd   2023

  • 細胞自己凝集化技術とヒトiPS細胞由来肢芽間葉系細胞を組み合わせた形状型軟骨組織体の作製

    藤澤佑樹, 太田智之, 太田智之, 高尾知佳, 北口陽平, 北口陽平, 山田大祐, 岩井良輔, 木股敬裕, 宝田剛志

    日本再生医療学会総会(Web)   22nd   2023

  • 無糖培地による培養軟骨移植時の内軟骨性骨化抑制法の開発

    北口陽平, 太田智之, 高尾知佳, 岩井良輔, 山田大祐, 大曽根達則, 木股敬裕, 宝田剛志

    日本形成外科学会総会・学術集会プログラム・抄録集   66th   2023

  • Development of an Endochondral Ossification Suppression Method using no glucose medium

    北口陽平, 北口陽平, 太田智之, 太田智之, 高尾知佳, 岩井良輔, 藤澤祐樹, 山田大祐, 大曽根達則, 木股敬裕, 宝田剛志

    日本軟骨代謝学会プログラム・抄録集   35th   2023

  • 関節軟骨再生を指向したヒトiPS細胞に由来する板状軟骨組織体の開発

    藤澤佑樹, たき平将太, たき平将太, 高尾知佳, 山田大祐, 太田智之, 太田智之, 北口陽平, 北口陽平, 岩井良輔, 中田英二, 木股敬裕, 尾崎敏文, 宝田剛志

    日本整形外科学会雑誌   97 ( 8 )   S1662 - S1662   2023

     More details

    Language:Japanese   Publisher:(公社)日本整形外科学会  

    J-GLOBAL

    researchmap

  • Disease modeling and high-throughput screening using disease-specific human iPSC-derived limb bud mesenchymal cells

    山田大祐, 高尾知佳, 藤澤祐樹, 戸口田淳也, 宝田剛志

    日本軟骨代謝学会プログラム・抄録集   35th   2023

  • Production of chondrocyte sheet using human induced pluripotent stem cell-derived limb bud mesenchymal cells

    高尾知佳, 佐藤正人, 豊田恵利子, 藤澤佑樹, 山田大祐, 中田英二, 尾崎敏文, 宝田剛志

    日本軟骨代謝学会プログラム・抄録集   35th   2023

  • ヒトiPS細胞来肢芽間葉系細胞を用いた硝子軟骨シートの作製

    高尾 知佳, 佐藤 正人, 豊田 恵利子, 山田 大祐, 中田 英二, 尾崎 敏文, 宝田 剛志

    日本整形外科学会雑誌   96 ( 8 )   S1557 - S1557   2022.9

     More details

    Language:Japanese   Publisher:(公社)日本整形外科学会  

    researchmap

  • 悪性末梢神経鞘腫瘍におけるPRRX1の悪性化因子としての役割

    たき平 将太, 山田 大祐, 高尾 知佳, 中田 英二, 近藤 宏也, 佐藤 浩平, 畑 利彰, 藤原 智洋, 国定 俊之, 尾崎 敏文, 宝田 剛志

    日本整形外科学会雑誌   96 ( 8 )   S1768 - S1768   2022.9

     More details

    Language:Japanese   Publisher:(公社)日本整形外科学会  

    researchmap

  • Induction and stable expansion of human pluripotent stem cell-derived limb bud-mesenchymal cells

    山田大祐, 高尾知佳, 中村正裕, MING Lu, 戸口田淳也, 宝田剛志

    日本軟骨代謝学会プログラム・抄録集   34th   2022

  • Dissecting the heterogeneity of Prrx1-positive cells during limb development by single-cell RNA sequencing analysis

    高尾知佳, 中村正裕, MING Lu, 山田大祐, 北條宏徳, 宝田剛志

    日本軟骨代謝学会プログラム・抄録集   34th   2022

  • Preparation of scaffold-free tissue-engineered 3D cartilage construct for application in facial plastic surgery

    太田智之, 高尾知佳, 岩井良輔, 山田大祐, 北口陽平, 北口陽平, 木股敬裕, 宝田剛志

    日本軟骨代謝学会プログラム・抄録集   34th   2022

  • 自己凝集化技術を応用した形状型スキャフォールドフリー三次元培養軟骨の開発

    太田智之, 太田智之, 高尾知佳, 岩井良輔, 山田大祐, 北口陽平, 北口陽平, 森脇健司, 中村正裕, 大曽根達則, 木股敬裕, 宝田剛志

    日本形成外科学会基礎学術集会プログラム・抄録集   31st   2022

  • 形成外科領域における細胞自己凝集化技術を用いたスキャフォールドフリー三次元軟骨培養法の開発

    北口陽平, 太田智之, 高尾知佳, 岩井良輔, 山田大祐, 藤澤祐樹, 大曽根達則, 森脇健司, 中村正裕, 木股敬裕, 宝田剛志

    日本形成外科学会基礎学術集会プログラム・抄録集   31st   2022

  • ヒトiPS細胞から誘導した肢芽間葉系細胞による軟骨シートの作製

    藤澤佑樹, 高尾知佳, 佐藤正人, 豊田恵利子, 山田大祐, 中田英二, 尾崎敏文, 宝田剛志

    移植(Web)   57 ( 4 )   2022

  • ヒト多能性幹細胞から誘導した肢芽間葉系細胞と,その拡大培養法の開発

    中田英二, 山田大祐, 高尾知佳, たき平将太, たき平将太, 藤原智洋, 尾崎敏文, 宝田剛志

    移植(Web)   57 ( 4 )   2022

  • マウス骨髄内LepR陽性細胞中の細胞不均一性の解析

    高尾知佳, 中村正裕, 山田大祐, 宝田剛志

    日本骨形態計測学会雑誌   32 ( 1 )   2022

  • ヒトiPS細胞由来肢芽間葉系細胞から作製した軟骨組織体を用いた骨再生研究

    佐藤浩平, 佐藤浩平, 高尾知佳, 藤澤祐樹, 山田大祐, 上原健敬, 依光正則, 中田英二, 尾崎敏文, 宝田剛志

    整形外科バイオマテリアル研究会プログラム・抄録集   41st   2022

  • ヒト多能性幹細胞由来軟骨前駆細胞を用いた硝子軟骨組織作製技術の開発

    明ろ, 山田大祐, 高尾知佳, 戸口田淳也, 宝田剛志

    日本骨代謝学会学術集会プログラム抄録集(CD-ROM)   39th   2021

  • マウス四肢骨格発生過程の一細胞遺伝子発現解析

    高尾知佳, 明ろ, 山田大祐, 北條宏徳, 宝田剛志

    日本骨代謝学会学術集会プログラム抄録集(CD-ROM)   39th   2021

  • Heterogeneity and its hierarchy of Prrx1-positive cells during limb development

    中井結希, 高尾知佳, 中村正祐, MING Lu, 山田大祐, 宝田剛志

    日本分子生物学会年会プログラム・要旨集(Web)   44th   2021

  • ヒト多能性幹細胞からの軟骨前駆細胞の誘導と,その拡大培養方法の開発

    山田大祐, 高尾知佳, 中村正裕, 戸口田淳也, 宝田剛志

    日本整形外科学会雑誌   95 ( 8 )   S1481 - S1481   2021

     More details

    Language:Japanese   Publisher:(公社)日本整形外科学会  

    J-GLOBAL

    researchmap

  • SNAT1は脳内のmTOR-オートファジー経路の制御因子である

    山田大祐, 土佐郁恵, 塚本俊平, 米田幸雄, 米田幸雄, 宝田剛志

    日本薬理学雑誌   155 ( Supplement )   2020

  • Postnatal Runx2 deletion causes age-related changes in bone.

    Ikue Tosa, Daisuke Yamada, Shunpei Tsukamoto, Kenji Kawabe, Takeshi Takarada, Mitsuaki Ono, Toshitaka Oohashi, Takuo Kuboki

    JOURNAL OF BONE AND MINERAL RESEARCH   34   219 - 219   2019.12

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:WILEY  

    Web of Science

    researchmap

  • 【脳の半分を占めるグリア細胞 脳と心と体をつなぐ"膠"】(第2章)グリア細胞と神経免疫・臓器連関 ペリサイト機能欠損による血液脳関門の破綻

    中里 亮太, 山田 大祐, 宝田 剛志

    実験医学   37 ( 17 )   2854 - 2860   2019.11

     More details

    Language:Japanese   Publisher:(株)羊土社  

    ペリサイト(周皮細胞)は、血管を構成する血管内皮細胞の周囲に接着し、血管の安定化や機能維持において重要な役割を担う。特に、中枢神経系においては、血液脳関門の形成・維持においてペリサイトの存在が不可欠であることが知られている。正常な神経機能を維持するうえで、血液脳関門は必須な保護機構であり、加齢などに伴う血液脳関門の機能低下はさまざまな神経変性疾患を引き起こす原因の1つであると考えられている。これらの理由から、近年、ペリサイトによる血液脳関門の機能維持メカニズムは大きな注目を集めている。(著者抄録)

    J-GLOBAL

    researchmap

  • SNAT1は神経細胞特異的なmTOR-オートファジー経路の制御因子である

    山田大祐, 土佐郁恵, 塚本俊平, 宝田剛志

    日本分子生物学会年会プログラム・要旨集(Web)   42nd   2019

  • 骨髄間葉系幹細胞におけるRunx2の発現低下は骨および骨髄の加齢様変化をもたらす

    土佐郁恵, 土佐郁恵, 山田大祐, 大野充昭, 大野充昭, 大橋俊孝, 窪木拓男, 宝田剛志

    日本補綴歯科学会誌(Web)   11   2019

  • 骨髄内加齢変化のRunx2コンディショナル欠損による模倣

    土佐郁恵, 土佐郁恵, 山田大祐, 塚本俊平, 河邊憲司, 大野充昭, 大橋俊孝, 窪木拓男, 宝田剛志

    日本分子生物学会年会プログラム・要旨集(Web)   41st   2018

  • セマフォリン6Bは肺がん幹細胞の維持に関与する

    山田 大祐, 前田 武彦

    日本癌学会総会記事   76回   P - 3271   2017.9

     More details

    Language:English   Publisher:(一社)日本癌学会  

    researchmap

  • セマフォリン3AはmTORC1を介して発がんを制御する

    山田 大祐, 前田 武彦

    日本癌学会総会記事   75回   P - 3162   2016.10

     More details

    Language:English   Publisher:(一社)日本癌学会  

    researchmap

  • mTOR活性化グリオーマモデルを用いたエネルギー代謝調節制御の解析と新規治療法の開発

    小林昌彦, HEGAZY Ahmed M., 山田大祐, 河野晋, 曽我朋義, 高橋智聡, 平尾敦

    がんと代謝研究会プログラム&抄録集   4th   2016

  • 癌性疼痛下のモルヒネ報酬効果減弱におけるミクログリアの機能的関与

    前田武彦, 山田大祐, 川原浩一, 池田崇志, 丸山光, 塚本敬, 五十嵐悠亮, 中沢美穂

    日本薬理学会北部会プログラム・抄録集   67th   2016

  • がん疼痛下のモルヒネ報酬効果減弱におけるミクログリアの関与

    前田武彦, 池田崇志, 丸山光, 塚本敬, 五十嵐悠亮, 山田大祐, 川原浩一

    鎮痛薬・オピオイドペプチドシンポジウムプログラム・抄録集   36th   2016

  • がん疼痛形成機構のセマフォリンファミリーによる制御

    前田武彦, 山田大祐, 川原浩一

    生体機能と創薬シンポジウム要旨集   2016   2016

  • 骨がん性疼痛モデルマウスの特性とオキシコドンの効果

    前田武彦, 丸山恵奈, 柳真美, 山岸明, 山田大祐, 川原浩一

    日本薬理学会北部会プログラム・抄録集   66th   2015

  • 骨転移腫瘍におけるセマフォリンの発現制御機構

    山田大祐, 川原浩一, 前田武彦

    応用薬理   89 ( 1-2 )   2015

  • 骨転移腫瘍におけるセマフォリンの発現制御機溝

    山田大祐, 川原浩一, 前田武彦

    生体機能と創薬シンポジウム要旨集   2015   2015

  • mTOR複合体1活性化グリオーマの悪性化形質に対するミトコンドリア活性阻害の効果

    M. Hegazy Ahmed, 小林昌彦, 山田大祐, 平尾敦

    がんと代謝研究会プログラム&抄録集   3rd   2015

  • 腫瘍細胞はmTORC1の活性化を介して骨芽細胞におけるSema3aの発現を制御する

    山田大祐, 川原浩一, 前田武彦

    日本生化学会大会(Web)   88th   [1P1100] - [1P1100]   2015

     More details

    Language:Japanese   Publisher:(公社)日本生化学会  

    J-GLOBAL

    researchmap

  • グリオーマイニシエーティング細胞のエネルギー代謝制御におけるmTOR複合体1の役割(Role of mTORC1 in the regulation of energy metabolism of glioma initiating cells)

    小林 昌彦, 山田 大祐, 平尾 敦

    日本癌学会総会記事   73回   P - 1198   2014.9

     More details

    Language:English   Publisher:(一社)日本癌学会  

    researchmap

  • Loss of Tsc1 accelerates malignant gliomagenesis when combined with oncogenic signals. International journal

    Daisuke Yamada, Takayuki Hoshii, Shingo Tanaka, Ahmed M Hegazy, Masahiko Kobayashi, Yuko Tadokoro, Kumiko Ohta, Masaya Ueno, Mohamed A E Ali, Atsushi Hirao

    Journal of biochemistry   155 ( 4 )   227 - 33   2014.4

     More details

    Language:English  

    Glioblastomas frequently harbour genetic lesions that stimulate the activity of mammalian target of rapamycin complex 1 (mTORC1). Loss of heterozygosity of tuberous sclerosis complex 1 (TSC1) or TSC2, which together form a critical negative regulator of mTORC1, is also seen in glioblastoma; however, it is not known how loss of the TSC complex affects the development of malignant gliomas. Here we investigated the role of Tsc1 in gliomagenesis in mice. Tsc1 deficiency up-regulated mTORC1 activity and suppressed the proliferation of neural stem/progenitor cells (NSPCs) in a serial neurosphere-forming assay, suggesting that Tsc1-deficient NSPCs have defective self-renewal activity. The neurosphere-forming capacity of Tsc1-deficient NSPCs was restored by p16(Ink4a)p19(Arf) deficiency. Combined Tsc1 and p16(Ink4a)p19(Arf) deficiency in NSPCs did not cause gliomagenesis in vivo. However, in a glioma model driven by an active mutant of epidermal growth factor receptor (EGFR), EGFRvIII, loss of Tsc1 resulted in an earlier onset of glioma development. The mTORC1 hyperactivation by Tsc1 deletion accelerated malignant phenotypes, including increased tumour mass and enhanced microvascular formation, leading to intracranial haemorrhage. These data demonstrate that, although mTORC1 hyperactivation itself may not be sufficient for gliomagenesis, it is a potent modifier of glioma development when combined with oncogenic signals.

    DOI: 10.1093/jb/mvt112

    PubMed

    J-GLOBAL

    researchmap

  • 骨がん性疼痛モデルマウスにおける疼痛及びモルヒネ精神依存形成の評価

    前田武彦, 佐野礼奈, 新飯田充, 藤井優子, 山田大祐, 尾崎昌宣

    日本薬理学会近畿部会プログラム・要旨集   126th   2014

  • 骨がんモデルにおける疼痛行動および骨代謝マーカー分子の発現解析

    山田大祐, 佐野礼奈, 新飯田充, 藤井優子, 前田武彦

    日本薬学会関東支部大会講演要旨集   58th   2014

  • mTOR複合体1による代謝調節を介したグリオーマ悪性化機構

    小林昌彦, 山田大祐, HEGAZY Ahmed M., 平尾敦

    がんと代謝研究会プログラム&抄録集   2nd   2014

  • Mesenchymal type GBMに対するNotch阻害剤の有効性

    田中慎吾, 中田光俊, 山田大祐, 中野伊知郎, 藤堂具紀, 稲生靖, 林裕, 濱田潤一郎, 平尾敦

    日本脳腫瘍学会プログラム・抄録集   31st   2013

  • グリオーマの悪性化におけるmTOR1シグナリングの役割

    山田大祐, 星居孝之, 田中慎吾, 平尾敦

    日本分子生物学会年会プログラム・要旨集(Web)   35th   2012

  • 時計遺伝子Bmal1による破骨細胞の分化制御

    山田大祐, 児玉歩美, 宝田剛志, 米田幸雄

    日本骨代謝学会学術集会プログラム抄録集   27th   2009

  • PRA1ドメインファミリーの機能解析

    山田大祐, 吉田祐美, 吉田祐美, 米田幸雄, 中村暢宏

    生化学   2008

▼display all

Presentations

  • ヒト多能性幹細胞由来肢芽間葉系細胞を用いた軟骨分化誘導技術の開発

    山田大祐, 髙尾知佳, 中村正裕, 戸口田淳也, 宝田剛志

    第22回日本再生医療学会  2023.3.24  日本再生医療学会

     More details

    Event date: 2023.3.23 - 2023.3.25

    Language:Japanese   Presentation type:Oral presentation (general)  

    Venue:京都  

    researchmap

  • 骨系統疾患特異的iPS由来ヒト肢芽間葉系細胞を用いたハイスループット薬剤スクリーニングシムテムの構築

    山田大祐, 髙尾知佳, 中村正裕, Ming Lu, 戸口田淳也, 宝田剛志

    第95回日本薬理学会年会  2022.3.8 

     More details

    Event date: 2022.3.7 - 2022.3.9

    Language:Japanese   Presentation type:Oral presentation (general)  

    researchmap

  • Induction and stable expansion of human pluripotent stem cell-derived limb bud-mesenchymal cells

    Daisuke Yamada, Tomoka Takao, Masayuki Nakamura, Ming Lu, Junya Toguchida, Takeshi Takarada

    2022.2.18 

     More details

    Event date: 2022.2.18 - 2022.2.19

    Language:English   Presentation type:Oral presentation (general)  

    researchmap

  • ヒト多能性幹細胞からの軟骨前駆細胞の誘導と、その拡大培養方法の開発

    山田大祐, 髙尾知佳, 中村正裕, 戸口田淳也, 宝田剛志

    第36回日本整形外科学会基礎学術集会  2021.10.14 

     More details

    Event date: 2021.10.14 - 2021.10.15

    Language:Japanese   Presentation type:Oral presentation (general)  

    researchmap

  • ヒト多能性幹細胞由来PRRX1陽性肢芽間葉系細胞の拡大培養技術と、その品質評価方法の開発 Invited

    山田大祐, 髙尾知佳, 戸口田淳也, 宝田剛志

    第35回日本軟骨代謝学会  2023.3.4  日本軟骨代謝学会

     More details

    Language:Japanese   Presentation type:Oral presentation (invited, special)  

    Venue:横浜  

    researchmap

  • 疾患特異的ヒトiPS細胞由来肢芽間葉系細胞を用いた病態再現と創薬スクリーニングシステムの構築

    山田大祐, 髙尾知佳, 藤澤祐樹, 戸口田淳也, 宝田剛志

    第35回日本軟骨代謝学会  2023.3.3  日本軟骨代謝学会

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:横浜  

    researchmap

  • 軟骨組織の安定供給と軟骨破壊疾患のモデリングを可能とするヒト多能性幹細胞由来肢芽間葉系細胞の樹立

    山田大祐, 中村正裕, 宝田剛志

    岡山大学次世代研究拠点 シンポジウム2020口腔器官の再構築から器官の発生・再生の統一原理の解明  2020.1.10 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    researchmap

  • Glutamine transporter SNAT1 is a neuron-specific regulator of mTOR-autophagy system

    山田大祐, 土佐郁恵, 塚本俊平, 宝田剛志

    第42回日本分子生物学会年会  2019.12 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:福岡  

    researchmap

  • SNAT1は脳内のmTOR-オートファジー経路の制御因子である

    山田大祐, 土佐郁恵, 塚本俊平, 米田幸雄, 宝田剛志

    第136回日本薬理学会近畿部会  2019.11 

     More details

    Language:Japanese   Presentation type:Oral presentation (general)  

    Venue:大阪  

    researchmap

  • セマフォリン6Bは肺がん幹細胞の維持に関与する

    山田大祐、前田武彦

    第76回日本癌学会学術総会  2017.7 

     More details

    Language:English   Presentation type:Poster presentation  

    Venue:横浜  

  • セマフォリン6Bは肺がん幹細胞の維持に関与する

    山田大祐, 川原浩一, 前田武彦

    第90回日本薬理学会年会  2017.3 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:横浜  

    researchmap

  • Sema3A/PlexinA1 signaling confers malignant phenotypes to lung cancer cell

    山田大祐, 川原浩一, 前田武彦

    第39回日本分子生物学会年会  2016.12 

     More details

    Language:Japanese  

    Venue:横浜  

    researchmap

  • セマフォリン3AはmTORC1を介して発がんを制御する

    山田大祐, 前田武彦

    第75回日本癌学会学術総会  2016.10 

     More details

    Language:Japanese  

    Venue:横浜  

    researchmap

  • Regulatory role of autocrine Semaphorin3a signaling in bone metastatic tumor development

    山田大祐, 川原浩一, 前田武彦

    第89回日本薬理学会年会  2016.3 

     More details

    Language:Japanese  

    Venue:横浜  

    researchmap

  • Regulatory role of autocrine Semaphorin3a signaling in bone metastatic tumor development

    山田大祐, 川原浩一, 前田武彦

    がん研究分野の特性等を踏まえた支援活動 公開シンポジウム  2016.2 

     More details

    Language:Japanese  

    Venue:東京  

    researchmap

  • 腫瘍細胞はmTORC1の活性化を介して骨芽細胞におけるSema3aの発現を制御する

    山田大祐, 川原浩一, 前田武彦

    第38回日本分子生物学会年会 / 第88回日本生化学会大会・合同大  2015.12 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:神戸  

    researchmap

  • 骨転移腫瘍におけるセマフォリンの発現制御機構

    山田大祐, 川原浩一, 前田武彦

    第17回応用薬理シンポジウム  2015.9 

     More details

    Language:Japanese   Presentation type:Oral presentation (general)  

    Venue:新潟  

    researchmap

  • 骨転移腫瘍におけるセマフォリンの発現制御機構

    山田大祐, 川原浩一, 前田武彦

    生体機能と創薬シンポジウム2015  2015.8 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:千葉  

    researchmap

  • Tumor Cell-Derived Factors Decrease Sema3a Expression in Osteoblast-like MC3T3-E1

    山田大祐, 川原浩一, 前田武彦

    第135回日本薬学会年会  2015.3 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:神戸  

    researchmap

  • Loss of Tsc1 accelerates malignant gliomagenesis when combined with oncogenic signals

    山田大祐, 星居孝之, 田中慎吾, 前田武彦, 平尾敦

    第126回日本薬理学会近畿部会  2014.10 

     More details

    Language:Japanese   Presentation type:Oral presentation (general)  

    Venue:和歌山  

    researchmap

  • 骨がんモデルにおける疼痛行動および骨代謝マーカー分子の発現解析

    山田大祐, 佐野礼奈, 新飯田充, 藤井優子, 前田武彦

    第58回日本薬学会関東支部大会  2014.10 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:東京  

    researchmap

  • Roles of mTORC1 signaling in the regulation of glioma malignancy

    第72回日本癌学会学術総会  2013.10 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:横浜  

    researchmap

  • グリオーマの悪性化におけるmTORC1シグナリングの役割

    山田大祐, 星居孝之, 田中信吾, 平尾敦

    第35回日本分子生物学会年会  2012.12 

     More details

    Language:Japanese   Presentation type:Oral presentation (general)  

    Venue:福岡  

    researchmap

  • 時計遺伝子Bmallによる破骨細胞の分化制御

    山田大祐, 児玉歩美, 宝田剛志, 米田幸雄

    第27回日本骨代謝学会学術集会  2009.7 

     More details

    Language:Japanese   Presentation type:Poster presentation  

    Venue:大阪  

    researchmap

▼display all

Industrial property rights

  • 骨又は軟骨関連疾患のスクリーニング方法

    宝田剛志, 山田大祐

     More details

    Applicant:国立大学法人 岡山大学

    Application no:特願JP2020/146904  Date applied:2020.9.1

    Announcement no:特開2022-041601  Date announced:2022.3.11

    J-GLOBAL

    researchmap

  • LBM、CPC、OPC、それらの調製方法及び品質管理方法、キット、移植材料並びに疾患モデル

    宝田剛志, 山田大祐, 髙尾知佳, 戸口田淳也, 吉富啓之

     More details

    Application no:特願PCT/JP2020/035517  Date applied:2019.9.18

    researchmap

Awards

  • 第28回日本軟骨代謝学会賞

    2023.3   日本軟骨代謝学会  

    山田大祐

     More details

  • 総合研究奨励賞(結城賞)

    2022.6   Induction and expansion of human PRRX1+ limb-bud-like mesenchymal cells from pluripotent stem cells

     More details

  • 優秀演題賞

    2020.1   岡山大学次世代研究拠点 シンポジウム2020口腔器官の再構築から器官の発生・再生の統一原理の解明   軟骨組織の安定供給と軟骨破壊疾患のモデリングを可能とするヒト多能性幹細胞由来肢芽間葉系細胞の樹立

    山田大祐, 中村正裕, 宝田剛志

     More details

  • JB論文賞

    2015.12   Loss of Tsc1 accelerates malignant gliomagenesis when combined with oncogenic signals

    D. Yamada, T. Hoshii, S. Tanaka, A. M. Hegazy, M. Kobayashi, Y. Tadokoro, K. Ohta, M. Ueno, M. A. E. Ali, A. Hirao

     More details

Research Projects

  • ヒト肉腫モデルを用いた新規治療標的 lncRNA の in vivo スクリーニング

    Grant number:2023-II-18  2024.01 - 2025.03

    公益財団法人 中外創薬科学財団  研究助成  研究助成金Ⅱ

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1500000

    researchmap

  • ヒト肢芽間葉系細胞由来肉腫モデルを用いた肉腫の発症機構の解明

    Grant number:A2023-19  2023.12 - 2024.12

    公益財団法人SGH財団  がん研究振興  第35回SGHがん研究助成

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1000000

    researchmap

  • dCas13 in vivoスクリーニングシステムを用いた新規肉腫治療標的lncRNAの同定

    Grant number:2300064  2023.11 - 2024.11

    公益財団法人 住友財団  研究助成  基礎科学研究助成

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\2000000

    researchmap

  • ヒト多能性幹細胞由来肉腫モデルを用いた増悪因子間相互作用を標的とする新規がん幹細胞根絶療薬の開発

    Grant number:2022ER001  2023.06 - 2024.05

    公益財団法人 小林がん学術振興会  先駆的研究(2)萌芽的研究 

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1000000

    researchmap

  • 増悪因子間の相互作用を標的とした革新的肉腫治療薬の開発

    Grant number:2022045851  2022.11 - 2025.05

    武田科学振興財団  医学系研究助成(がん領域(基礎)) 

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\2000000 ( Direct expense: \2000000 )

    researchmap

  • 増悪因子間の相互作用を標的とした新規肉腫治療薬コンセプトの検証

    Grant number:DNW-22022  2022.10 - 2023.09

    国立研究開発法人日本医療研究開発機構(AMED)  創薬総合支援事業 (創薬ブースター)  創薬ブースター

    山田大祐, 宝田剛志, 髙尾知佳, 尾﨑敏文, 中田英二

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\13959000 ( Direct expense: \12690000 、 Indirect expense:\1269000 )

    researchmap

  • ヒト肢芽間葉系細胞を用いた腫瘍モデルの構築

    Grant number:202120121  2022.01 - 2023.04

    公益財団法人 上原記念生命科学財団  研究助成  基礎医学

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\5000000 ( Direct expense: \5000000 )

    researchmap

  • ヒト肉腫自然発症モデルを利用した血中悪性化指標マーカーの探索

    Grant number:21K07192  2021.04 - 2024.03

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

    山田 大祐, 中田 英二, 宝田 剛志, 高尾 知佳

      More details

    Authorship:Principal investigator 

    Grant amount:\4290000 ( Direct expense: \3300000 、 Indirect expense:\990000 )

    申請者らは、ヒト骨肉種では転写制御因子PRRX1が高発現していることを見出しているだけでなく、PRRX1の発現量が高い患者は予後不良を示すことも明らかにしている。さらに、マウス骨肉腫モデルでもPRRX1が発現しているだけでなく、ヒト骨肉腫細胞株143Bにおいては、PRRX1のノックダウンによって増殖性や浸潤性が低下するだけでなく、ドキソルビシンとシスプラチンへの抵抗性が解除されることも見出している。これらの研究実績は、Transl Oncol 誌(2021 Vol. 14 Issue 1 Pages 100960)にて発表を行い、骨肉腫におけるPRRX1の増悪因子としての機能を明らかにした。その後、悪性神経鞘腫においてもPRRX1が増悪因子として機能することも見出しており(未発表データ)、肉腫におけるPRRX1の重要性が明らかになってきている。また、Nature Biomedical Engineering誌(2021 Vol. 5 Issue 8 Pages 926-940)にて、ヒト多能性幹細胞から発生過程を模倣した分化誘導方法を用いて、肢芽間葉系細胞を作成する技術に関しての発表を行い、本研究課題を遂行するための研究ツールの開発にも成功している。研究成果は、AMED及び申請者の所属機関である岡山大学にてプレスリリースを行い、一般向けの内容紹介を掲載して公開されている。さらに、ヒト多能性幹細胞から作成した肺オルガノイドを用いて、前がん病変を再現することにも成功している(Int J Cancer 2021 Vol. 149 Issue 8 Pages 1593-1604)。以上の結果から、ヒト多能性幹細胞を用いた腫瘍モデルを構築するための実験技術に関しては、十分整っていると考えられる。

    researchmap

  • 軟骨破壊の治療を目的とした機能的軟骨シートの開発(継続)

    Grant number:A081  2021.04 - 2022.03

    橋渡し研究戦略的推進プログラム(岡山大学拠点)  シーズA 

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\2500000 ( Direct expense: \2500000 )

    researchmap

  • 軟骨破壊の治療を目的とした機能的軟骨シートの開発

    Grant number:A040  2020.04 - 2021.03

    橋渡し研究戦略的推進プログラム(岡山大学拠点)  シーズA 

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1750000 ( Direct expense: \1750000 )

    researchmap

  • Novel therapeutic strategy for sarcomas by targeting PPRX1 positive cancer stem-like cells

    Grant number:18K15212  2018.04 - 2021.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Early-Career Scientists  Grant-in-Aid for Early-Career Scientists

    Yamada Daisuke

      More details

    Authorship:Principal investigator 

    Grant amount:\4030000 ( Direct expense: \3100000 、 Indirect expense:\930000 )

    We performed PRRX1 immunostaining on 35 human osteosarcoma specimens to assess the correlation between PRRX1 level and overall survival. In patients with osteosarcoma, the expression level of PRRX1 positively correlated with poor prognosis or the ratio of lung metastasis. Additionally, we found PRRX1 expression on in 143B cells, a human osteosarcoma line with a high metastatic capacity. Downregulation of PRRX1 not only suppressed proliferation and invasion but also increased the sensitivity to cisplatin and doxorubicin. When 143B cells were subcutaneously transplanted into nude mice, PRRX1 knockdown decreased tumor sizes and rates of lung metastasis. Interestingly, forskolin, a chemical compound identified by Connectivity Map analysis using RNA expression signatures during PRRX1 knockdown, decreased tumor proliferation and cell migration to the same degree as PRRX1 knockdown. These results demonstrate that PRRX1 promotes tumor malignancy in human osteosarcoma.

    researchmap

  • 骨微小環境を介した制御性B細胞の維持機構の解明

    2018.04 - 2019.03

    公益財団法人ウエスコ学術振興財団  研究助成 

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\300000

    researchmap

  • 間葉系幹細胞による免疫抑制効果のメカニズム解明

    2018.04 - 2019.03

    公益財団法人岡山医学振興会  医学研究助成 

    山田大祐

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1000000

    researchmap

  • Novel therapeutic strategy for sarcomas by targeting PPRX1 positive cancer stem-like cells

    Grant number:15K18449  2015.04 - 2018.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Young Scientists (B)  Grant-in-Aid for Young Scientists (B)

    Yamada Daisuke

      More details

    Authorship:Principal investigator 

    Grant amount:\4160000 ( Direct expense: \3200000 、 Indirect expense:\960000 )

    In this study, we studied the oncogenic role of semaphorin 3A to establish the novel therapeutic strategy against lung cancer. Inhibition of semaphorin 3A signaling by shRNA system in lung cancer cells decreased their proliferative capacity and increased the sensitivity to EGFR inhibitors. Furthermore, the self-renewal or tumorigenic capacity of cancer stem-like cells, the origin of cancer, was completely abolished by the inhibition of semaphorin 3A signaling. These results demonstrate that the inhibition of semaphorin 3A signaling not only releases the resistance to several drugs but also eradicates cancer stem-like cell in lung cancer.

    researchmap

  • Development of new treatment for cartilage regeneration for hip osteoarthritis

    Grant number:23K08590  2023.04 - 2026.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research  Grant-in-Aid for Scientific Research (C)

    山田 和希, 中田 英二, 宝田 剛志, 尾崎 敏文, 高尾 知佳, 山田 大祐

      More details

    Grant amount:\4680000 ( Direct expense: \3600000 、 Indirect expense:\1080000 )

    researchmap

  • Development of new molecular targeted therapy for osteosarcoma lung metastasis

    Grant number:22K09401  2022.04 - 2025.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    中田 英二, 宝田 剛志, 尾崎 敏文, 高尾 知佳, 山田 大祐

      More details

    Authorship:Coinvestigator(s)  Grant type:Competitive

    Grant amount:\4160000 ( Direct expense: \3200000 、 Indirect expense:\960000 )

    researchmap

  • 新規肉腫モデルを用いた肉腫発生メカニズムの解明と治療標的分子同定の試み

    Grant number:22K09378  2022.04 - 2025.03

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

    上原 健敬, 中田 英二, 宝田 剛志, 尾崎 敏文, 高尾 知佳, 山田 大祐

      More details

    Authorship:Coinvestigator(s)  Grant type:Competitive

    Grant amount:\4160000 ( Direct expense: \3200000 、 Indirect expense:\960000 )

    researchmap

  • 新しい軟骨移植素材を用いた軟骨再生の開発

    Grant number:22K09356  2022.04 - 2025.03

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

    宮澤 慎一, 中田 英二, 尾崎 敏文, 高尾 知佳, 山田 大祐

      More details

    Authorship:Coinvestigator(s)  Grant type:Competitive

    Grant amount:\4290000 ( Direct expense: \3300000 、 Indirect expense:\990000 )

    researchmap

  • 関節軟骨の光in vivoイメージング技術の開発

    Grant number:22K09332  2022.04 - 2025.03

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

    鉄永 智紀, 中田 英二, 宝田 剛志, 尾崎 敏文, 高尾 知佳, 山田 大祐

      More details

    Authorship:Coinvestigator(s)  Grant type:Competitive

    Grant amount:\4160000 ( Direct expense: \3200000 、 Indirect expense:\960000 )

    researchmap

  • ヒト関節軟骨オルガノイドを利用した革新的創薬スクリーニング技術の開発

    Grant number:21H02643  2021.04 - 2025.03

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

    宝田 剛志, 高尾 知佳, 山田 大祐, 戸口田 淳也

      More details

    Authorship:Coinvestigator(s)  Grant type:Competitive

    Grant amount:\17160000 ( Direct expense: \13200000 、 Indirect expense:\3960000 )

    高齢化が急速に進行する中で、膝関節軟骨の代表的疾患である変形性関節症(Osteoarthritis, OA)に対する薬物治療開発は遅々として進んでいない。ヒト生体環境/病態を模倣したハイスループット化合物スクリーニングシステムの開発は、薬剤開発の初期段階に極めて重要であるが、ヒト関節軟骨組織(硝子軟骨組織)を均一大量に調整することは従来不可能であった。申請者は、ヒト多能性幹細胞より、高い軟骨分化指向性を有し、拡大培養可能で、前向き品質管理が可能なヒト軟骨前駆細胞を大量に調整する技術を開発することに成功し、「ヒト」の硝子軟骨組織(=ヒト関節軟骨オルガノイド)を「均一・大量」に、安定的に調整する準備が整った。本研究では、開発したヒト軟骨前駆細胞を細胞源とし、均一大量に作製したOA病態ヒト関節軟骨オルガノイドによるハイスループット化合物スクリーニング系を開発することで、OA治療候補化合物を同定し、独自に開発する疾患モデル動物での治療効果を検証することを目指す。この点において本年度は、ヒト多能性幹細胞株にpiggybacでのDOX inducible RUNX2発現カセットを導入し (hPSC-iRUNX2株の樹立)、同株より申請者らの開発した方法を使用して、ヒト軟骨前駆細胞(hCPC-iRUNX2)を樹立し、継代培養により増やした。DOXを添加することでRUNX2の発現が認められ系の確立に成功した。

    researchmap

  • 悪性軟部腫瘍におけるPRRX1の機能解析とその新規薬物療法への応用

    Grant number:21K07178  2021.04 - 2024.03

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

    たき平 将太, 中田 英二, 宝田 剛志, 尾崎 敏文, 山田 大祐

      More details

    Authorship:Coinvestigator(s) 

    Grant amount:\4030000 ( Direct expense: \3100000 、 Indirect expense:\930000 )

    我々は転写制御因子Paired related homeobox 1(PRRX1)について研究を行ってきた。PRRX1は四肢骨格形成に強く関与しているが、腫瘍の悪性化に関与するとの報告がある。オープンデータベースの解析にて悪性軟部腫瘍の1つである悪性末梢神経鞘腫(MPNST)においてPRRX1が比較的高発現していることを見出した。ヒト腫瘍検体においてPRRX1の発現の多寡を免疫染色にて確認し高発現/低発現と群分けし予後と肺転移について相関を評価したところ、高発現グループにおいて5年生存率は低く、転移率も高い結果となった。次にレンチウイルスベクターを用いてPRRX1に対するshRNA(shPRRX1)をヒトMPNST細胞株に導入、PRRX1の発現を抑制した細胞株を作製し、対照群(空ベクター導入群)とshPRRX1導入群間で増殖能、遊走能、浸潤能を検討したところ、増殖能・遊走能・浸潤能いずれも低下する結果となった。次にPiggybac systemを用いてPRRX1をドキシサイクリン依存的に過剰発現させるヒトMPNST細胞株を樹立した。対照群(ドキシサイクリン未処理群)とPRRX1過剰発現群(ドキシサイクリン処理群)間で増殖能、遊走能、浸潤能を検討したところ、PRRX1過剰発現群では増殖能に変化はなく、遊走能、浸潤能は増加していた。次に、PRRX1の発現を抑制した細胞株と対照群、それぞれの細胞株をマウスに皮下移植を行ったところshPRRX1導入群では腫瘍径は有意に縮小していた。本研究によりPRRX1は腫瘍悪性化の原因の可能性が示唆され、その働きを阻害する薬剤を見いだすことで、本来治療が困難なことが多いとされる軟部肉腫に対する新規治療法となり得る可能性が考えられた。本研究では悪性腫瘍のメカニズムの一端を解明しうるだけでなく、新規創薬開発の点においても非常に重要性が高いと考える。

    researchmap

  • 光活性型Creシステムを利用した生体内遺伝子操作法の開発

    Grant number:20K21373  2020.07 - 2023.03

    日本学術振興会  科学研究費助成事業 挑戦的研究(萌芽)  挑戦的研究(萌芽)

    宝田 剛志, 高尾 知佳, 山田 大祐, 佐藤 守俊

      More details

    Authorship:Coinvestigator(s) 

    Grant amount:\6370000 ( Direct expense: \4900000 、 Indirect expense:\1470000 )

    生体内遺伝子操作の精度(時期特異性や、細胞種特異性)は、Cre recombinase (Cre)loxP 部位特異的 DNA組換え酵素反応の応用により格段に上昇した。青光照射でDNA組み換え反応をコントロールできる光活性型Cre(Photoactivatable(PA)-Cre)に着目し、このPA-Cre技術と、テトラサイクリン誘導発現系システム(TetON/OFF)のActb locusへのノックイン技術を組み合わせることで、in vivoでのlight/Dox-dependentなDNA組み換え反応を可能とする遺伝子改変マウス(TRE-PA-Creマウス)の開発に成功した。同マウスを使用することで、個体レベルでの光活性型Creシステムの有用性を実証し、免疫/幹細胞の細胞動態研究(例:どのタイミングで傷害部位へ遊走し、遊走後どれくらい滞在するのか?遊走後の細胞は分化/機能変化の点でどのような運命を辿るのか?)や、がん研究(例:遺伝子変異細胞の動態を極めて早期に生体内で観察)への応用を目指す。本年度は、昨年作成した各種tTAマウス(ROSA-tTA:全身性にtTAを発現するマウス、Foxp3-tTAマウス:制御性T細胞にてtTAを発現するマウス、LepR-tTAマウス:LepR陽性間葉系間質細胞にてtTAを発現するマウス)とTRE-PA-Creマウス、LSL-tdTomatoマウスを交配し、各種tTA;PA-Crel;tdTomatoマウスを作製した。qPCRにより、それぞれの細胞種にてtTAを発現することを確認した。

    researchmap

  • Development of new combined cancer immunotherapy for malignant soft tissue tumor

    Grant number:19K09551  2019.04 - 2022.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    Nakata Eiji

      More details

    Authorship:Coinvestigator(s) 

    Grant amount:\4420000 ( Direct expense: \3400000 、 Indirect expense:\1020000 )

    When PRRX1 was knocked down in an osteosarcoma cell line, which is a hyperlung metastatic strain, and the proliferative capacity was measured by the WST-8 assay, it was confirmed in vitro that the proliferation was strongly suppressed. It was also confirmed that when these cells were transplanted into mice, proliferation was suppressed as compared with the control group. In addition, when the osteosarcoma cells in which PRRX1 was knocked down were examined by wound healing assay and migration assay, the number of invading cells was reduced compared to the control group. Furthermore, osteosarcoma cells in which PRRX1 was knocked down were transplanted under the skin of mice, and 6 weeks later, the lungs of the mice were taken out and the number and size of lung metastases were compared with those of the control group. Then, it was found that the osteosarcoma cell group in which PRRX1 was knocked down had a smaller number of lung metastases.

    researchmap

  • Role of tumor-derived semaphorin 3A in the development of cancer pain

    Grant number:15K08682  2015.04 - 2018.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    Maeda Takehiko

      More details

    Authorship:Coinvestigator(s) 

    Grant amount:\4940000 ( Direct expense: \3800000 、 Indirect expense:\1140000 )

    Elucidation of potential therapeutic targets for bone cancer pain would contribute to relief of the intractable and unbearable pain. We used microarray analysis to reveal upregulation of semaphorin 3A (Sema3A) in the model mice for bone cancer pain. Knockdown of Sema3A and inhibition of Sema3A cellular signaling attenuated the tumor growth and the development of the pain. These results suggest that Sema3A is a potential therapeutic target molecule for the development of bone cancer pain.

    researchmap

▼display all

 

Class subject in charge

  • Practicals: Regenerative Science (2023academic year) special  - その他

  • Research Projects: Regenerative Science (2023academic year) special  - その他

  • Research Projects and Practicals: Regenerative Science I (2023academic year) special  - その他

  • Lecture and Research Projects: Regenerative Science I (2023academic year) special  - その他

  • Research Projects and Practicals: Regenerative Science II (2023academic year) special  - その他

  • Lecture and Research Projects: Regenerative Science II (2023academic year) special  - その他

  • Research Projects and Practicals: Regenerative Science I (2022academic year) special  - その他

  • Lecture and Research Projects: Regenerative Science I (2022academic year) special  - その他

  • Research Projects and Practicals: Regenerative Science II (2022academic year) special  - その他

  • Lecture and Research Projects: Regenerative Science II (2022academic year) special  - その他

▼display all

 

Media Coverage

  • ヒト多能性幹細胞から手足の元である肢芽間葉系細胞の誘導・拡大培養に成功―軟骨再生医療やiPS細胞を用いた創薬研究への応用に期待― Internet

    日本医療研究開発機構(AMED)  https://www.amed.go.jp/news/release_20210810.html  2021.8

     More details

  • ヒト多能性幹細胞から手足の元である肢芽間葉系細胞の誘導・拡大培養に成功―軟骨再生医療やiPS細胞を用いた創薬研究への応用に期待― Internet

    岡山大学  https://www.okayama-u.ac.jp/tp/release/release_id866.html  2021.8

     More details

    Author:Myself 

    researchmap

  • 神経細胞でのオートファジーを治療標的とするための候補分子を発見新しい神経細胞保護薬の開発へ Internet

    岡山大学  https://www.okayama-u.ac.jp/tp/release/release_id662.html  2019.9

     More details

    Author:Myself 

    researchmap