2024/12/23 更新

お知らせ

 

写真a

ソノダ ソウイチロウ
園田 聡一朗
SONODA SOICHIRO
所属
歯学研究院 歯学部門 助教
歯学部 歯学科(併任)
歯学府 口腔科学専攻(併任)
歯学府 歯学専攻(併任)
職名
助教
連絡先
メールアドレス
電話番号
0926426302
プロフィール
1. 再生医療におけるヒト間葉系幹細胞を応用したトランスレーショナルリサーチ 2. 間葉系幹細胞の起源と特性、動態の解析 3. 歯学部生を対象とした解剖学実習、歯牙解剖実習 4. 学部学生および大学院生の研究指導
外部リンク

学位

  • 博士(歯学)

研究テーマ・研究キーワード

  • 研究テーマ: 歯髄・口腔組織幹細胞研究

    研究キーワード: 幹細胞、再生治療、病因解析、 トランスレーショナルリサーチ

    研究期間: 2018年6月

受賞

  • 日本歯科保存学会奨励賞

    2017年6月   日本歯科保存学会  

     詳細を見る

    Soichiro Sonoda, Haruyoshi Yamaza, Lan Ma, Yosuke Tanaka, Erika Tomoda, Reona Aijima, Kazuaki Nonaka, Toshio Kukita, Songtao Shi, Fusanori Nishimura, Takayoshi Yamaza. Interferon-gamma improves impaired dentinogenic and immunosuppressive functions of irreversible pulpitis-derived human dental pulp stem cells. Scientific Reports. Nature publishing group. 6, Article number: 19286. 2016.

論文

  • NaV1.1 contributes to the cell cycle of human mesenchymal stem cells by regulating AKT and CDK2. 査読

    Zakaria MF, Kato H, Sonoda S, Kato K, Uehara N, Kyumoto-Nakamura Y, Sharifa MM, Yu L, Dai L, Yamaza H, Kajioka S, Nishimura F, Yamaza T

    Journal of cell science   137 ( 19 )   2024年9月   ISSN:0021-9533 eISSN:1477-9137

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Journal of cell science  

    Non-excitable cells express sodium voltage-gated channel alpha subunit 1 gene and protein (known as SCN1A and NaV1.1, respectively); however, the functions of NaV1.1 are unclear. In this study, we investigated the role of SCN1A and NaV1.1 in human mesenchymal stem cells (MSCs). We found that SCN1A was expressed in MSCs, and abundant expression of NaV1.1 was observed in the endoplasmic reticulum; however, this expression was not found to be related to Na+ currents. SCN1A-silencing reduced MSC proliferation and delayed the cell cycle in the S phase. SCN1A silencing also suppressed the protein levels of CDK2 and AKT (herein referring to total AKT), despite similar mRNA expression, and inhibited AKT phosphorylation in MSCs. A cycloheximide-chase assay showed that SCN1A-silencing induced CDK2 but not AKT protein degradation in MSCs. A proteolysis inhibition assay using epoxomicin, bafilomycin A1 and NH4Cl revealed that both the ubiquitin-proteasome system and the autophagy and endo-lysosome system were irrelevant to CDK2 and AKT protein reduction in SCN1A-silenced MSCs. The AKT inhibitor LY294002 did not affect the degradation and nuclear localization of CDK2 in MSCs. Likewise, the AKT activator SC79 did not attenuate the SCN1A-silencing effects on CDK2 in MSCs. These results suggest that NaV1.1 contributes to the cell cycle of MSCs by regulating the post-translational control of AKT and CDK2.

    DOI: 10.1242/jcs.261732

    Web of Science

    Scopus

    PubMed

    researchmap

  • Erythropoietin receptor signal is crucial for periodontal ligament stem cell-based tissue reconstruction in periodontal disease 査読

    加藤 大樹, 上原 範久, 久本 由香里, ヤマグチ エリカ, 合島 怜央奈, 山座 治義, 西村 英紀, 山座 孝義

    Scientific Reports   14   6719   2024年3月   eISSN:20452322

     詳細を見る

    記述言語:英語   出版者・発行元:Springer  

    Alveolar bone loss caused by periodontal disease eventually leads to tooth loss. Periodontal ligament stem cells (PDLSCs) are the tissue-specific cells for maintaining and repairing the periodontal ligament, cementum, and alveolar bone. Here, we investigated the role of erythropoietin receptor (EPOR), which regulates the microenvironment-modulating function of mesenchymal stem cells, in PDLSC-based periodontal therapy. We isolated PDLSCs from patients with chronic periodontal disease and healthy donors, referred to as PD-PDLSCs and Cont-PDLSCs, respectively. PD-PDLSCs exhibited reduced potency of periodontal tissue regeneration and lower expression of EPOR compared to Cont-PDLSCs. EPOR-silencing suppressed the potency of Cont-PDLSCs mimicking PD-PDLSCs, whereas EPO-mediated EPOR activation rejuvenated the reduced potency of PD-PDLSCs. Furthermore, we locally transplanted EPOR-silenced and EPOR-activated PDLSCs into the gingiva around the teeth of ligament-induced periodontitis model mice and demonstrated that EPOR in PDLSCs participated in the regeneration of the periodontal ligament, cementum, and alveolar bone in the ligated teeth. The EPOR-mediated paracrine function of PDLSCs maintains periodontal immune suppression and bone metabolic balance via osteoclasts and osteoblasts in the periodontitis model mice. Taken together, these results suggest that EPOR signaling is crucial for PDLSC-based periodontal regeneration and paves the way for the development of novel options for periodontal therapy.

    CiNii Research

  • Bone metastatic mammary tumor cell-derived extracellular vesicles inhibit osteoblast maturation via JNK signaling 査読

    Uehara, N; Shibusawa, N; Mikami, Y; Kyumoto-Nakamura, Y; Sonoda, S; Kato, H; Yamaza, T; Kukita, T

    ARCHIVES OF BIOCHEMISTRY AND BIOPHYSICS   750   109821   2023年11月   ISSN:0003-9861 eISSN:1096-0384

     詳細を見る

    記述言語:その他   掲載種別:研究論文(学術雑誌)   出版者・発行元:Archives of Biochemistry and Biophysics  

    The metastases of breast cancer to bone often cause osteolytic lesions not only by stimulating osteoclasts to resorb the bone but also by inhibiting osteoblasts from bone formation. Although tumor cell-derived extracellular vesicles (EVs) promote osteoclast differentiation and bone resorption, their roles in osteoblast differentiation and functions have not been elucidated. In this study, we investigated the effects of breast cancer cell-derived EVs on osteoblast differentiation and functions in vitro. We found that upon osteogenic induction, 4T1 bone metastatic mouse mammary tumor cell-derived EVs (4T1-EVs) were inhibited matrix mineralization of ST2 mouse bone marrow stromal cells. Temporal expression analysis of osteoblast marker genes, including runt-related transcription factor 2 (Runx2), osterix (Osx), alkaline phosphatase (Alp), collagen type I (Col1a1), bone sialoprotein (Bsp), and osteocalcin (Bglap) revealed that 4T1-EVs decreased their expression during the late stage of osteoblast differentiation. Elevated levels of c-Jun N-terminal kinase (JNK) phosphorylation, upon osteogenic induction, were diminished by 4T1-EVs, significantly. In contrast, the nullification of reduced JNK phosphorylation by anisomycin, a potent JNK activator, increased the expression levels of osteoblast differentiation markers. Overall, our data indicated that 4T1-EVs affect osteoblast maturation, at least partially, through the regulation of JNK activity, which provides novel insights into the pathological impact of osteolytic bone metastasis and the role of EVs in osteoblast differentiation.

    DOI: 10.1016/j.abb.2023.109821

    Web of Science

    Scopus

    PubMed

    researchmap

  • Cutting-edge regenerative therapy for Hirschsprung disease and its allied disorders 査読

    Yoshimaru, K; Matsuura, T; Uchida, Y; Sonoda, S; Maeda, S; Kajihara, K; Kawano, Y; Shirai, T; Toriigahara, Y; Kalim, AS; Zhang, XY; Takahashi, Y; Kawakubo, N; Nagata, K; Yamaza, H; Yamaza, T; Taguchi, T; Tajiri, T

    SURGERY TODAY   54 ( 9 )   977 - 994   2023年9月   ISSN:0941-1291 eISSN:1436-2813

     詳細を見る

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

    Hirschsprung disease (HSCR) and its associated disorders (AD-HSCR) often result in severe hypoperistalsis caused by enteric neuropathy, mesenchymopathy, and myopathy. Notably, HSCR involving the small intestine, isolated hypoganglionosis, chronic idiopathic intestinal pseudo-obstruction, and megacystis-microcolon-intestinal hypoperistalsis syndrome carry a poor prognosis. Ultimately, small-bowel transplantation (SBTx) is necessary for refractory cases, but it is highly invasive and outcomes are less than optimal, despite advances in surgical techniques and management. Thus, regenerative therapy has come to light as a potential form of treatment involving regeneration of the enteric nervous system, mesenchyme, and smooth muscle in affected areas. We review the cutting-edge regenerative therapeutic approaches for managing HSCR and AD-HSCR, including the use of enteric nervous system progenitor cells, embryonic stem cells, induced pluripotent stem cells, and mesenchymal stem cells as cell sources, the recipient intestine's microenvironment, and transplantation methods. Perspectives on the future of these treatments are also discussed.

    DOI: 10.1007/s00595-023-02741-6

    Web of Science

    Scopus

    PubMed

    researchmap

  • Extracellular vesicles rejuvenate the microenvironmental modulating function of recipient tissue-specific mesenchymal stem cells in osteopenia treatment 査読 国際誌

    Sonoda, S; Yamaza, T

    FRONTIERS IN ENDOCRINOLOGY   14   1151429 - 1151429   2023年3月   ISSN:1664-2392

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Frontiers in Endocrinology  

    Systemic transplantation of mesenchymal stem cells (MSCs), such as bone marrow MSCs (BMMSCs) and stem cells from human exfoliated deciduous teeth (SHED), is considered a prominent treatment for osteopenia. However, the mechanism of action of the transplanted MSCs has been poorly elucidated. In the recipient target tissue, including bone and bone marrow, only a few donor MSCs can be detected, suggesting that the direct contribution of donor MSCs may not be expected for osteopenia treatment. Meanwhile, secretomes, especially contents within extracellular vesicles (EVs) released from donor MSCs (MSC-EVs), play key roles in the treatment of several diseases. In this context, administrated donor MSC-EVs may affect bone-forming function of recipient cells. In this review, we discuss how MSC-EVs contribute to bone recovery recipient tissue in osteopenia. We also summarize a novel mechanism of action of systemic administration of SHED-derived EVs (SHED-EVs) in osteopenia. We found that reduced telomerase activity in recipient BMMSCs caused the deficiency of microenvironmental modulating function, including bone and bone marrow-like niche formation and immunomodulation in estrogen-deficient osteopenia model mice. Systemic administration of SHED-EVs could exert therapeutic effects on bone reduction via recovering the telomerase activity, leading to the rejuvenation of the microenvironmental modulating function in recipient BMMSCs, as seen in systemic transplantation of SHED. RNase-preconditioned donor SHED-EVs diminished the therapeutic benefits of administrated SHED-EVs in the recipient osteopenia model mice. These facts suggest that MSC-EV therapy targets the recipient BMMSCs to rejuvenate the microenvironmental modulating function via telomerase activity, recovering bone density. We then introduce future challenges to develop the reproducible MSC-EV therapy in osteopenia.

    DOI: 10.3389/fendo.2023.1151429

    Web of Science

    Scopus

    PubMed

    researchmap

  • miR-92a-3p encapsulated in bone metastatic mammary tumor cell–derived extracellular vesicles modulates mature osteoclast longevity 査読

    Uehara, N; Kyumoto-Nakamura, Y; Mikami, Y; Hayatsu, M; Sonoda, S; Yamaza, T; Kukita, A; Kukita, T

    CANCER SCIENCE   113 ( 12 )   4219 - 4229   2022年12月   ISSN:1347-9032 eISSN:1349-7006

     詳細を見る

    記述言語:その他   掲載種別:研究論文(学術雑誌)   出版者・発行元:Cancer Science  

    Aberrant osteoclast formation and activation are the hallmarks of osteolytic metastasis. Extracellular vesicles (EVs), released from bone metastatic tumor cells, play a pivotal role in the progression of osteolytic lesions. However, the mechanisms through which tumor cell–derived EVs regulate osteoclast differentiation and function have not been fully elucidated. In this study, we found that 4T1 bone metastatic mouse mammary tumor cell–derived EVs (4T1-EVs) are taken up by mouse bone marrow macrophages to facilitate osteoclastogenesis. Furthermore, treatment of mature osteoclasts with 4T1-EVs promoted bone resorption, which was accompanied by enhanced survival of mature osteoclasts through the negative regulation of caspase-3. By comparing the miRNA content in 4T1-EVs with that in 67NR nonmetastatic mouse mammary tumor cell–derived EVs (67NR-EVs), miR-92a-3p was identified as one of the most enriched miRNAs in 4T1-EVs, and its transfer into mature osteoclasts significantly reduced apoptosis. Bioinformatic and Western blot analyses revealed that miR-92a-3p directly targeted phosphatase and tensin homolog (PTEN) in mature osteoclasts, resulting in increased levels of phospho-Akt. Our findings provide novel insights into the EV-mediated regulation of osteoclast survival through the transfer of miR-92a-3p, which enhances mature osteoclast survival via the Akt survival signaling pathway, thus promoting bone resorption.

    DOI: 10.1111/cas.15557

    Web of Science

    Scopus

    PubMed

    researchmap

  • Targeting hepatic oxidative stress rescues bone loss in liver fibrosis 査読

    Yamaza, T; Sonoda, S; Murata, S; Yamaza, H; Yuniartha, R; Fujiyoshi, J; Yoshimaru, K; Matsuura, T; Oda, Y; Ohga, S; Tajiri, T; Taguchi, T

    MOLECULAR METABOLISM   66   101599   2022年12月   ISSN:2212-8778 eISSN:2212-8778

     詳細を見る

    記述言語:その他   掲載種別:研究論文(学術雑誌)   出版者・発行元:Molecular Metabolism  

    Objective: Chronic liver diseases often involve metabolic damage to the skeletal system. The underlying mechanism of bone loss in chronic liver diseases remains unclear, and appropriate therapeutic options, except for orthotopic liver transplantation, have proved insufficient for these patients. This study aimed to investigate the efficacy and mechanism of transplantation of immature hepatocyte-like cells converted from stem cells from human exfoliated deciduous teeth (SHED-Heps) in bone loss of chronic liver fibrosis. Methods: Mice that were chronically treated with CCl4 received SHED-Heps, and trabecular bone density, reactive oxygen species (ROS), and osteoclast activity were subsequently analyzed in vivo and in vitro. The effects of stanniocalcin 1 (STC1) knockdown in SHED-Heps were also evaluated in chronically CCl4 treated mice. Results: SHED-Hep transplantation (SHED-HepTx) improved trabecular bone loss and liver fibrosis in chronic CCl4-treated mice. SHED-HepTx reduced hepatic ROS production and interleukin 17 (Il-17) expression under chronic CCl4 damage. SHED-HepTx reduced the expression of both Il-17 and tumor necrosis factor receptor superfamily 11A (Tnfrsf11a) and ameliorated the imbalance of osteoclast and osteoblast activities in the bone marrow of CCl4-treated mice. Functional knockdown of STC1 in SHED-Heps attenuated the benefit of SHED-HepTx including anti-bone loss effect by suppressing osteoclast differentiation through TNFSF11–TNFRSF11A signaling and enhancing osteoblast differentiation in the bone marrow, as well as anti-fibrotic and anti-ROS effects in the CCl4-injured livers. Conclusions: These findings suggest that targeting hepatic ROS provides a novel approach to treat bone loss resulting from chronic liver diseases.

    DOI: 10.1016/j.molmet.2022.101599

    Web of Science

    Scopus

    PubMed

    researchmap

  • 骨転移乳癌細胞由来細胞外小胞に封入されたmiR-92a-3pは成熟破骨細胞の寿命を調節する(miR-92a-3p encapsulated in bone metastatic mammary tumor cell-derived extracellular vesicles modulates mature osteoclast longevity) 査読

    Uehara Norihisa, Kyumoto-Nakamura Yukari, Mikami Yoshikazu, Hayatsu Manabu, Sonoda Soichiro, Yamaza Takayoshi, Kukita Akiko, Kukita Toshio

    Cancer Science   113 ( 12 )   4219 - 4229   2022年12月   ISSN:1347-9032

     詳細を見る

    記述言語:英語   出版者・発行元:John Wiley & Sons Australia, Ltd  

    骨転移乳癌細胞由来細胞外小胞(EV)が破骨細胞の分化や機能に及ぼす影響とそのメカニズムについて検討した。マウス乳癌細胞株4T1由来EV(4T1-EV)がマウス骨髄マクロファージに取り込まれ、破骨細胞形成を促進することを見出した。成熟破骨細胞を4T1-EVで処理すると、骨吸収が促進され、カスパーゼ3を負に制御して成熟破骨細胞の生存が促進された。4T1-EVと非転移性マウス乳癌細胞株67NR由来EV(67NR-EV)におけるmiRNAの内容を比較した。その結果、miR-92a-3pが4T1-EVに最も濃縮されたmiRNAの一つとして特定され、miR-92a-3pの成熟破骨細胞への移入によりアポトーシスが著しく抑制された。さらに、miR-92a-3pは成熟破骨細胞においてphosphatase and tensin homolog(PTEN)を直接標的とし、リン酸化Aktのレベルを増加させた。以上より、マウス乳癌細胞株4T1由来EVに存在するmiR-92a-3pは、Akt生存シグナル経路を介して成熟破骨細胞の生存を高め、骨吸収を促進させることが示された。

  • Protocol to generate xenogeneic-free/serum-free human dental pulp stem cells 査読

    Sonoda, S; Yamaza, H; Yoshimaru, K; Taguchi, T; Yamaza, T

    STAR PROTOCOLS   3 ( 2 )   101386   2022年6月   ISSN:2666-1667 eISSN:2666-1667

     詳細を見る

    記述言語:その他   掲載種別:研究論文(学術雑誌)   出版者・発行元:STAR Protocols  

    Human dental pulp stem cell (hDPSCs)-based therapy is a feasible option for regenerative medicine, such as dental pulp regeneration. Here, we show the steps needed to colony-forming unit-fibroblasts (CFU-F)-based isolation, expansion, and cryopreservation of hDPSCs for manufacturing clinical-grade products under a xenogeneic-free/serum-free condition. We also demonstrate the characterization of hDPSCs by CFU-F, flow cytometric, and in vitro multipotent assays. For complete details on the use and execution of this protocol, please refer to Iwanaka et al. (2020).

    DOI: 10.1016/j.xpro.2022.101386

    Web of Science

    Scopus

    PubMed

    researchmap

  • Dental pulp stem cells as a therapy for congenital entero-neuropathy 査読

    Yoshimaru, K; Yamaza, T; Kajioka, S; Sonoda, S; Yanagi, Y; Matsuura, T; Yoshizumi, J; Oda, Y; Iwata, N; Takai, C; Nakayama, S; Taguchi, T

    SCIENTIFIC REPORTS   12 ( 1 )   6990   2022年4月   ISSN:2045-2322 eISSN:2045-2322

     詳細を見る

    記述言語:その他   掲載種別:研究論文(学術雑誌)   出版者・発行元:Scientific Reports  

    Hirschsprung’s disease is a congenital entero-neuropathy that causes chronic constipation and intestinal obstruction. New treatments for entero-neuropathy are needed because current surgical strategies have limitations5. Entero-neuropathy results from enteric nervous system dysfunction due to incomplete colonization of the distal intestine by neural crest-derived cells. Impaired cooperation between the enteric nervous system and intestinal pacemaker cells may also contribute to entero-neuropathy. Stem cell therapy to repair these multiple defects represents a novel treatment approach. Dental pulp stem cells derived from deciduous teeth (dDPSCs) are multipotent cranial neural crest-derived cells, but it remains unknown whether dDPSCs have potential as a new therapy for entero-neuropathy. Here we show that intravenous transplantation of dDPSCs into the Japanese Fancy-1 mouse, an established model of hypoganglionosis and entero-neuropathy, improves large intestinal structure and function and prolongs survival. Intravenously injected dDPSCs migrate to affected regions of the intestine through interactions between stromal cell-derived factor-1α and C-X-C chemokine receptor type-4. Transplanted dDPSCs differentiate into both pacemaker cells and enteric neurons in the proximal colon to improve electrical and peristaltic activity, in addition to their paracrine effects. Our findings indicate that transplanted dDPSCs can differentiate into different cell types to correct entero-neuropathy-associated defects.

    DOI: 10.1038/s41598-022-10077-3

    Web of Science

    Scopus

    PubMed

    researchmap

  • A New Target of Dental Pulp-Derived Stem Cell-Based Therapy on Recipient Bone Marrow Niche in Systemic Lupus Erythematosus 査読

    Sonoda, S; Yamaza, T

    INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES   23 ( 7 )   2022年4月   ISSN:1661-6596 eISSN:1422-0067

     詳細を見る

    記述言語:英語   出版者・発行元:International Journal of Molecular Sciences  

    Recent advances in mesenchymal stem/stromal cell (MSC) research have led us to con-sider the feasibility of MSC-based therapy for various diseases. Human dental pulp-derived MSCs (hDPSCs) have been identified in the dental pulp tissue of deciduous and permanent teeth, and they exhibit properties with self-renewal and in vitro multipotency. Interestingly, hDPSCs exhibit superior immunosuppressive functions toward immune cells, especially T lymphocytes, both in vitro and in vivo. Recently, hDPSCs have been shown to have potent immunomodulatory functions in treating systemic lupus erythematosus (SLE) in the SLE MRL/lpr mouse model. However, the mechanisms underlying the immunosuppressive efficacy of hDPSCs remain unknown. This review aims to introduce a new target of hDPSC-based therapy on the recipient niche function in SLE.

    DOI: 10.3390/ijms23073479

    Web of Science

    Scopus

    PubMed

    researchmap

  • Biliary atresia-specific deciduous pulp stem cells feature biliary deficiency. 査読 国際誌

    Soichiro Sonoda, Koichiro Yoshimaru, Haruyoshi Yamaza, Ratih Yuniartha, Toshiharu Matsuura, Erika Yamauchi-Tomoda, Sara Murata, Kento Nishida, Yoshinao Oda, Shouichi Ohga, Tasturo Tajiri, Tomoaki Taguchi, Takayoshi Yamaza

    Stem cell research & therapy   12 ( 1 )   582 - 582   2021年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    BACKGROUND: Biliary atresia (BA) is a severe hepatobiliary disease in infants that ultimately results in hepatic failure; however, its pathological mechanism is poorly elucidated. Current surgical options, including Kasai hepatoportoenterostomy and orthotopic liver organ transplantations, are palliative; thus, innovation in BA therapy is urgent. METHODS: To examine whether BA-specific post-natal stem cells are feasible for autologous cell source for BA treatment, we isolated from human exfoliated deciduous teeth, namely BA-SHED, using a standard colony-forming unit fibroblast (CFU-F) method and compared characteristics as mesenchymal stem cells (MSCs) to healthy donor-derived control SHED, Cont-SHED. BA-SHED and Cont-SHED were intrasplenically transplanted into chronic carbon tetrachloride (CCl4)-induced liver fibrosis model mice, followed by the analysis of bile drainage function and donor integration in vivo. Immunohistochemical assay was examined for the regeneration of intrahepatic bile ducts in the recipient's liver using anti-human specific keratin 19 (KRT19) antibody. RESULTS: BA-SHED formed CFU-F, expressed MSC surface markers, and exhibited in vitro mesenchymal multipotency similar to Cont-SHED. BA-SHED showed less in vitro hepatogenic potency than Cont-SHED. Cont-SHED represented in vivo bile drainage function and KRT19-positive biliary regeneration in chronic carbon tetrachloride-induced liver fibrosis model mice. BA-SHED failed to show in vivo biliary potency and bile drainage function compared to Cont-SHED. CONCLUSION: These findings indicate that BA-SHED are not feasible source for BA treatment, because BA-SHED may epigenetically modify the underlying prenatal and perinatal BA environments. In conclusion, these findings suggest that BA-SHED-based studies may provide a platform for understanding the underlying molecular mechanisms of BA development and innovative novel modalities in BA research and treatment.

    DOI: 10.1186/s13287-021-02652-8

  • Modulation of osteoclastogenesis through adrenomedullin receptors on osteoclast precursors: initiation of differentiation by asymmetric cell division 査読

    Toshio Kukita, Hidenobu Hiura, Jiong-Yan Gu, Jing-Qi Zhang, Yukari Kyumoto-Nakamura, Norihisa Uehara, Sara Murata, Soichiro Sonoda, Takayoshi Yamaza, Ichiro Takahashi, Akiko Kukita

    Laboratory Investigation   101 ( 11 )   1449 - 1457   2021年10月

     詳細を見る

    記述言語:その他   掲載種別:研究論文(学術雑誌)  

    Adrenomedullin (ADM), a member of the calcitonin family of peptides, is a potent vasodilator and was shown to have the ability to modulate bone metabolism. We have previously found a unique cell surface antigen (Kat1 antigen) expressed in rat osteoclasts, which is involved in the functional regulation of the calcitonin receptor (CTR). Cross-linking of cell surface Kat1 antigen with anti-Kat1 antigen monoclonal antibody (mAbKat1) stimulated osteoclast formation only under conditions suppressed by calcitonin. Here, we found that ADM provoked a significant stimulation in osteoclastogenesis only in the presence of calcitonin; a similar biological effect was seen with mAbKat1 in the bone marrow culture system. This stimulatory effect on osteoclastogenesis mediated by ADM was abolished by the addition of mAbKat1. 125I-labeled rat ADM (125I-ADM)-binding experiments involving micro-autoradiographic studies demonstrated that mononuclear precursors of osteoclasts abundantly expressed ADM receptors, and the specific binding of 125I-ADM was markedly inhibited by the addition of mAbKat1, suggesting a close relationship between the Kat1 antigen and the functional ADM receptors expressed on cells in the osteoclast lineage. ADM receptors were also detected in the osteoclast progenitor cells in the late mitotic phase, in which only one daughter cell of the dividing cell express ADM receptors, suggesting the semiconservative cell division of the osteoclast progenitors in the initiation of osteoclastogenesis. Messenger RNAs for the receptor activity-modifying-protein 1 (RAMP1) and calcitonin receptor-like receptor (CRLR) were expressed in cells in the osteoclast lineage; however, the expression of RAMP2 or RAMP3 was not detected in these cells. It is suggested that the Kat1 antigen is involved in the functional ADM receptor distinct from the general ADM receptor, consisting of CRLR and RAMP2 or RAMP3. Modulation of osteoclastogenesis through functional ADM receptors abundantly expressed on mononuclear osteoclast precursors is supposed to be important in the fine regulation of osteoclast differentiation in a specific osteotrophic hormonal condition with a high level of calcitonin in blood.

    DOI: 10.1038/s41374-021-00633-2

  • Targeting of deciduous tooth pulp stem cell-derived extracellular vesicles on telomerase-mediated stem cell niche and immune regulation in systemic lupus erythematosus 査読

    Soichiro Sonoda, Sara Murata, Hiroki Kato, Fouad Zakaria, Yukari Kyumoto-Nakamura, Norihisa Uehara, Haruyoshi Yamaza, Toshio Kukita, Takayoshi Yamaza

    Journal of Immunology   206 ( 12 )   3053 - 3063   2021年6月

     詳細を見る

    記述言語:その他   掲載種別:研究論文(学術雑誌)  

    Systemic transplantation of stem cells from human exfoliated deciduous teeth (SHED) is used to treat systemic lupus erythematosus (SLE)-like disorders in MRL/lpr mice. However, the mechanisms underlying the SHED-based therapy remain unclear. In this study, we hypothesized that trophic factors within SHED-releasing extracellular vesicles (SHED-EVs) ameliorate the SLE-like phenotypes in MRL/lpr mice. SHED-EVs were isolated from the culture supernatant of SHED. SHED-EVs were treated with or without RNase and systemically administered to MRL/lpr mice. Subsequently, recipient bone marrow mesenchymal stem cells (BMMSCs) isolated from SHED-EV-administered MRL/lpr mice were examined for the in vitro and in vivo activity of hematopoietic niche formation and immunoregulation. Furthermore, the recipient BMMSCs were secondarily transplanted into MRL/lpr mice. The systemic SHED-EV infusion ameliorated the SLE-like phenotypes in MRL/lpr mice and improved the functions of recipient BMMSCs by rescuing Tert mRNA-associated telomerase activity, hematopoietic niche formation, and immunoregulation. The secondary transplantation of recipient BMMSCs recovered the immune condition and renal functions of MRL/lpr mice. The RNase treatment depleted RNAs, such as microRNAs, within SHED-EVs, and the RNA-depleted SHED-EVs attenuated the benefits of SHED-EVs in MRL/lpr mice. Collectively, our findings suggest that SHED-secreted RNAs, such as microRNAs, play a crucial role in treating SLE by targeting the telomerase activity of recipient BMMSCs.

    DOI: 10.4049/jimmunol.2001312

  • Cholangiogenic potential of human deciduous pulp stem cell-converted hepatocyte-like cells 査読 国際誌

    Ratih Yuniartha, Takayoshi Yamaza, Soichiro Sonoda, Koichiro Yoshimaru, Toshiharu Matsuura, Haruyoshi Yamaza, Yoshinao Oda, Shouichi Ohga, Tomoaki Taguchi

    Stem Cell Research & Therapy   12 ( 1 )   57 - 57   2021年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    <title>Abstract</title><sec>
    <title>Background</title>
    Stem cells from human exfoliated deciduous teeth (SHED) have been reported to show the in vivo and in vitro hepatic differentiation, SHED-Heps; however, the cholangiogenic potency of SHED-Heps remains unclear. Here, we hypothesized that SHED-Heps contribute to the regeneration of intrahepatic bile duct system in chronic fibrotic liver.


    </sec><sec>
    <title>Methods</title>
    SHED were induced into SHED-Heps under cytokine stimulation. SHED-Heps were intrasplenically transplanted into chronically CCl4-treated liver fibrosis model mice, followed by the analysis of donor integration and hepatobiliary metabolism in vivo. Immunohistochemical assay was examined for the regeneration of intrahepatic bile duct system in the recipient liver. Furthermore, SHED-Heps were induced under the stimulation of tumor necrosis factor alpha (TNFA).


    </sec><sec>
    <title>Results</title>
    The intrasplenic transplantation of SHED-Heps into CCl4-treated mice showed that donor SHED-Heps behaved as human hepatocyte paraffin 1- and human albumin-expressing hepatocyte-like cells in situ and ameliorated CCl4-induced liver fibrosis. Of interest, the integrated SHED-Heps not only expressed biliary canaliculi ATP-binding cassette transporters including ABCB1, ABCB11, and ABCC2, but also recruited human keratin 19- (KRT19-) and KRT17-positive cells, which are considered donor-derived cholangiocytes, regenerating the intrahepatic bile duct system in the recipient liver. Furthermore, the stimulation of TNFA induced SHED-Heps into KRT7- and SRY-box 9-positive cells.


    </sec><sec>
    <title>Conclusions</title>
    Collectively, our findings demonstrate that infused SHED-Heps showed cholangiogenic ability under the stimulation of TNFA in CCl4-damaged livers, resulting in the regeneration of biliary canaliculi and interlobular bile ducts in chronic fibrotic liver. Thus, the present findings suggest that SHED-Heps may be a novel source for the treatment of cholangiopathy.


    </sec>

    DOI: 10.1186/s13287-020-02113-8

  • Extracellular vesicles from deciduous pulp stem cells recover bone loss by regulating telomerase activity in an osteoporosis mouse model 査読

    Soichiro Sonoda, Sara Murata, Kento Nishida, Hiroki Kato, Norihisa Uehara, Yukari N. Kyumoto, Haruyoshi Yamaza, Ichiro Takahashi, Toshio Kukita, Takayoshi Yamaza

    Stem Cell Research and Therapy   11 ( 1 )   2020年7月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Background: Systemic transplantation of stem cells from human exfoliated deciduous teeth (SHED) recovers bone loss in animal models of osteoporosis; however, the mechanisms underlying this remain unclear. Here, we hypothesized that trophic factors within SHED-releasing extracellular vesicles (SHED-EVs) rescue osteoporotic phenotype. Methods: EVs were isolated from culture supernatant of SHED. SHED-EVs were treated with or without ribonuclease and systemically administrated into ovariectomized mice, followed by the function of recipient bone marrow mesenchymal stem cells (BMMSCs) including telomerase activity, osteoblast differentiation, and sepmaphorine-3A (SEMA3A) secretion. Subsequently, human BMMSCs were stimulated by SHED-EVs with or without ribonuclease treatment, and then human BMMSCs were examined regarding the function of telomerase activity, osteoblast differentiation, and SEMA3A secretion. Furthermore, SHED-EV-treated human BMMSCs were subcutaneously transplanted into the dorsal skin of immunocompromised mice with hydroxyapatite tricalcium phosphate (HA/TCP) careers and analyzed the de novo bone-forming ability. Results: We revealed that systemic SHED-EV-infusion recovered bone volume in ovariectomized mice and improved the function of recipient BMMSCs by rescuing the mRNA levels of Tert and telomerase activity, osteoblast differentiation, and SEMA3A secretion. Ribonuclease treatment depleted RNAs, including microRNAs, within SHED-EVs, and these RNA-depleted SHED-EVs attenuated SHED-EV-rescued function of recipient BMMSCs in the ovariectomized mice. These findings were supported by in vitro assays using human BMMSCs incubated with SHED-EVs. Conclusion: Collectively, our findings suggest that SHED-secreted RNAs, such as microRNAs, play a crucial role in treating postmenopausal osteoporosis by targeting the telomerase activity of recipient BMMSCs.

    DOI: 10.1186/s13287-020-01818-0

  • A model study for the manufacture and validation of clinical-grade deciduous dental pulp stem cells for chronic liver fibrosis treatment 査読

    Tsuyoshi Iwanaka, Takayoshi Yamaza, Soichiro Sonoda, Koichiro Yoshimaru, Toshiharu Matsuura, Haruyoshi Yamaza, Shouichi Ohga, Yoshinao Oda, Tomoaki Taguchi

    Stem Cell Research and Therapy   11 ( 1 )   2020年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Background: Human deciduous pulp stem cells (hDPSCs) have remarkable stem cell potency associated with cell proliferation, mesenchymal multipotency, and immunosuppressive function and have shown beneficial effects in a variety of animal disease models. Recent studies demonstrated that hDPSCs exhibited in vivo anti-fibrotic and anti-inflammatory action and in vivo hepatogenic-associated liver regeneration, suggesting that hDPSCs may offer a promising source with great clinical demand for treating liver diseases. However, how to manufacture ex vivo large-scale clinical-grade hDPSCs with the appropriate quality, safety, and preclinical efficacy assurances remains unclear. Methods: We isolated hDPSCs from human deciduous dental pulp tissues formed by the colony-forming unit-fibroblast (CFU-F) method and expanded them under a xenogeneic-free and serum-free (XF/SF) condition; hDPSC products were subsequently stored by two-step banking including a master cell bank (MCB) and a working cell bank (WCB). The final products were directly thawed hDPSCs from the WCB. We tested the safety and quality check, stem cell properties, and preclinical potentials of final hDPSC products and hDPSC products in the MCB and WCB. Results: We optimized manufacturing procedures to isolate and expand hDPSC products under a XF/SF culture condition and established the MCB and the WCB. The final hDPSC products and hDPSC products in the MCB and WCB were validated the safety and quality including population doubling ability, chromosome stability, microorganism safety, and stem cell properties including morphology, cell surface marker expression, and multipotency. We also evaluated the in vivo immunogenicity and tumorigenicity and validated in vivo therapeutic efficacy for liver regeneration in a CCl4-induced chronic liver fibrosis mouse model in the final hDPSC products and hDPSC products in the WCB. Conclusion: The manufacture and quality control results indicated that the present procedure could produce sufficient numbers of clinical-grade hDPSC products from a tiny deciduous dental pulp tissue to enhance clinical application of hDPSC products in chronic liver fibrosis.

    DOI: 10.1186/s13287-020-01630-w

  • Therapeutic potential of hepatocyte-like-cells converted from stem cells from human exfoliated deciduous teeth in fulminant Wilson’s disease 査読

    Junko Fujiyoshi, Haruyoshi Yamaza, Soichiro Sonoda, Ratih Yuniartha, Kenji Ihara, Kazuaki Nonaka, Tomoaki Taguchi, Shouichi Ohga, Takayoshi Yamaza

    Scientific Reports   9 ( 1 )   2019年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Wilson’s disease (WD) is an inherited metabolic disease arising from ATPase copper transporting beta gene (ATP7B) mutation. Orthotoropic liver transplantation is the only radical treatment of fulminant WD, although appropriate donors are lacking at the onset of emergency. Given the hepatogenic capacity and tissue-integration/reconstruction ability in the liver of stem cells from human exfoliated deciduous teeth (SHED), SHED have been proposed as a source for curing liver diseases. We hypothesized the therapeutic potential of SHED and SHED-converted hepatocyte-like- cells (SHED-Heps) for fulminant WD. SHED and SHED-Heps were transplanted into WD model Atp7b-mutated Long-Evans Cinnamon (LEC) rats received copper overloading to induce a lethal fulminant liver failure. Due to the superior copper tolerance via ATP7B, SHED-Hep transplantation gave more prolonged life-span of fulminant LEC rats than SHED transplantation. The integrated ATP7B-expressing SHED-Heps showed more therapeutic effects on to restoring the hepatic dysfunction and tissue damages in the recipient liver than the integrated naïve SHED without ATP7B expression. Moreover, SHED-Heps could reduce copper-induced oxidative stress via ATP7B- independent stanniocalcin 1 secretion in the fulminant LEC rats, suggesting a possible role for paracrine effect of the integrated SHED-Heps. Taken together, SHED-Heps offer a potential of functional restoring, bridging, and preventive approaches for treating fulminant WD.

    DOI: 10.1038/s41598-018-38275-y

  • Therapeutic potential of spheroids of stem cells from human exfoliated deciduous teeth for chronic liver fibrosis and hemophilia A 査読

    Yoshiaki Takahashi, Ratih Yuniartha, Takayoshi Yamaza, Soichiro Sonoda, Haruyoshi Yamaza, Kosuke Kirino, Koichiro Yoshimaru, Toshiharu Matsuura, Tomoaki Taguchi

    Pediatric Surgery International   35 ( 12 )   1379 - 1388   2019年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Purpose: Mesenchymal stem cell (MSC)-based cell therapies have emerged as a promising treatment option for various diseases. Due to the superior survival and higher differentiation efficiency, three-dimensional spheroid culture systems have been an important topic of MSC research. Stem cells from human exfoliated deciduous teeth (SHED) have been considered an ideal source of MSCs for regenerative medicine. Thus, in the present study, we introduce our newly developed method for fabricating SHED-based micro-hepatic tissues, and demonstrate the therapeutic effects of SHED-based micro-hepatic tissues in mouse disease models. Methods: SHED-converted hepatocyte-like cells (SHED-HLCs) were used for fabricating spherical micro-hepatic tissues. The SHED-HLC-based spheroids were then transplanted both into the liver of mice with CCl4-induced chronic liver fibrosis and the kidney of factor VIII (F8)-knock-out mice. At 4 weeks after transplantation, the therapeutic efficacy was investigated. Results: Intrahepatic transplantation of SHED-HLC-spheroids improved the liver dysfunction in association with anti-fibrosis effects in CCl4-treated mice. Transplanted SHED-converted cells were successfully engrafted in the recipient liver. Meanwhile, renal capsular transplantation of the SHED-HLC-spheroids significantly extended the bleeding time in F8-knock-out mice. Conclusions: These findings suggest that SHED-HLC-based micro-hepatic tissues might be a promising source for treating pediatric refractory diseases, including chronic liver fibrosis and hemophilia A.

    DOI: 10.1007/s00383-019-04564-4

  • Osteoblast lineage-specific cell-surface antigen (A7) regulates osteoclast recruitment and calcification during bone remodeling 査読

    Tamer Badawy, Yukari Kyumoto-Nakamura, Norihisa Uehara, Jingqi Zhang, Soichiro Sonoda, Hidenobu Hiura, Takayoshi Yamaza, Akiko Kukita, Toshio Kukita

    Laboratory Investigation   99 ( 6 )   866 - 884   2019年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Bone remodeling is a continuous process characterized by highly coordinated cell-cell interactions in distinct multi-cellular units. Osteoclasts, which are specialized bone resorbing cells, play a central role in bone remodeling. Although the RANKL/RANK axis determines the gross number of osteoclasts present in bone tissue, detailed molecular events regulating bone remodeling related to osteoclast recruitment, initiation of bone remodeling, and coupling of bone resorption and bone formation are still ambiguous. We hypothesized that osteoblast-specific cell-surface molecules contribute to the molecular modulation of bone remodeling. Therefore, we searched for regulatory cell-surface molecules expressed on osteoblasts by use of B-cell hybridoma technology. We obtained a monoclonal antibody A7 (A7 MAb) highly specific to cells of osteoblast-lineage. Here we describe the expression pattern and possible role of A7 antigen specifically recognized by A7 MAb. In vitro, A7 antigen was expressed on cell-surface of osteoblasts and osteoblast-like bone marrow stromal cells. In vivo, A7 antigen was detected in a subset of bone surface osteoblasts and in osteocytes, with a typical cell membrane expression pattern. Tissue array analysis showed only a limited expression of A7 antigen in osteocytes close to the bone surface. Immunoblotting and immunoprecipitation analysis showed that A7 antigen is a lineage-specific cell-surface protein with an approximate molecular weight of 45 KDa. Cross-linking of cell-surface A7 antigen in cultures of osteoclastogenesis showed stimulation of osteoclast formation. Marked suppression of calcification in primary osteoblast cultures was observed when A7 antigen was cross-linked with anti-A7 antigen MAb, A7 MAb. These data suggest that A7 antigen regulates recruitment of osteoclasts and triggering of calcification. A7 antigen may be an important molecule involved in the precise regulation of bone remodeling.

    DOI: 10.1038/s41374-018-0179-4

  • Acetylsalicylic Acid Treatment and Suppressive Regulation of AKT Accelerate Odontogenic Differentiation of Stem Cells from the Apical Papilla 査読

    Yosuke Tanaka, Soichiro Sonoda, Haruyoshi Yamaza, Sara Murata, Kento Nishida, Yukari Kyumoto-Nakamura, Norihisa Uehara, Kazuaki Nonaka, Toshio Kukita, Takayoshi Yamaza

    Journal of Endodontics   45 ( 5 )   591 - 598.e6   2019年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Introduction: Stem cells isolated from the root apical papilla of human teeth (stem cells from the apical papilla [SCAPs])are capable of forming tooth root dentin and are a feasible source for bioengineered tooth root regeneration. In this study, we examined the effect of acetylsalicylic acid (ASA)on odontogenic differentiation of SCAPs in vitro and in vivo. Methods: SCAPs were cultured under odontogenic conditions supplemented with or without ASA. ASA-treated SCAPs were also subcutaneously transplanted into immunocompromised mice. Results: ASA accelerates in vitro and in vivo odontogenic differentiation of SCAPs associated with down-regulation of runt-related nuclear factor 2 and up-regulation of specificity protein 7, nuclear factor I C, and dentin phosphoprotein. ASA up-regulated the phosphorylation of AKT in the odontogenic SCAPs. Of interest, pretreatments with phosphoinositide 3-kinase inhibitor LY294402 and small interfering RNA for AKT promoted ASA-induced in vitro and in vivo odontogenic differentiation of SCAPs. LY294402 and small interfering RNA for AKT also suppressed the ASA-induced expression of runt-related nuclear factor 2 and enhanced ASA-induced expression of specificity protein 7, nuclear factor I C, and dentin phosphoprotein in SCAPs. Conclusions: These findings suggest that a combination of ASA treatment and suppressive regulation of the phosphoinositide 3-kinase–AKT signaling pathway is a novel approach for SCAP-based tooth root regeneration.

    DOI: 10.1016/j.joen.2019.01.016

  • Exogenous nitric oxide stimulates the odontogenic differentiation of rat dental pulp stem cells 査読

    Soichiro Sonoda, Yu Feng Mei, Ikiru Atsuta, Atsushi Danjo, Haruyoshi Yamaza, Shion Hama, Kento Nishida, Ronghao Tang, Yukari Kyumoto-Nakamura, Norihisa Uehara, Toshio Kukita, Fusanori Nishimura, Takayoshi Yamaza

    Scientific Reports   8 ( 1 )   2018年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Nitric oxide (NO) is thought to play a pivotal regulatory role in dental pulp tissues under both physiological and pathological conditions. However, little is known about the NO functions in dental pulp stem cells (DPSCs). We examined the direct actions of a spontaneous NO gas-releasing donor, NOC-18, on the odontogenic capacity of rat DPSCs (rDPSCs). In the presence of NOC-18, rDPSCs were transformed into odontoblast-like cells with long cytoplasmic processes and a polarized nucleus. NOC-18 treatment increased alkaline phosphatase activity and enhanced dentin-like mineralized tissue formation and the expression levels of several odontoblast-specific genes, such as runt related factor 2, dentin matrix protein 1 and dentin sialophosphoprotein, in rDPSCs. In contrast, carboxy-PTIO, a NO scavenger, completely suppressed the odontogenic capacity of rDPSCs. This NO-promoted odontogenic differentiation was activated by tumor necrosis factor-NF-κB axis in rDPSCs. Further in vivo study demonstrated that NOC-18-application in a tooth cavity accelerated tertiary dentin formation, which was associated with early nitrotyrosine expression in the dental pulp tissues beneath the cavity. Taken together, the present findings indicate that exogenous NO directly induces the odontogenic capacity of rDPSCs, suggesting that NO donors might offer a novel host DPSC-targeting alternative to current pulp capping agents in endodontics.

    DOI: 10.1038/s41598-018-21183-6

  • Suppression of AKT-mTOR signal pathway enhances osteogenic/dentinogenic capacity of stem cells from apical papilla 査読

    Yosuke Tanaka, Soichiro Sonoda, Haruyoshi Yamaza, Sara Murata, Kento Nishida, Shion Hama, Yukari Kyumoto-Nakamura, Norihisa Uehara, Kazuaki Nonaka, Toshio Kukita, Takayoshi Yamaza

    Stem Cell Research and Therapy   9 ( 1 )   2018年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Background: Stem cells from apical papilla (SCAP) are a subpopulation of mesenchymal stem cells (MSCs) isolated from the apical papilla of the developing tooth root apex of human teeth. Because of their osteogenic/dentinogenic capacity, SCAP are considered as a source for bone and dentin regeneration. However, little is understood about the molecular mechanism of osteogenic/dentinogenic differentiation of SCAP. Phosphoinositide 3 kinase (PI3K)-AKT-mammalian target of rapamycin (mTOR) signal pathway participates in regulating the differentiation of various cell types, such as MSCs. In this study, we examined the role of the PI3K-AKT-mTOR signal pathway in the osteogenic/dentinogenic differentiation of SCAP. Moreover, we challenge to fabricate scaffold-free SCAP-based spheroidal calcified constructs. Methods: SCAP were pretreated with or without small interfering RNA for AKT (AKT siRNA), PI3K inhibitor LY294402, and mTOR inhibitor rapamycin and were cultured under osteogenic/dentinogenic differentiation to examine in vitro and in vivo calcified tissue formation. Moreover, SCAP-based cell aggregates were pretreated with or without LY294402 and rapamycin. The cell aggregates were cultured under osteogenic/dentinogenic condition and were analyzed the calcification of the aggregates. Results: Pretreatment with AKT siRNA, LY294402, and rapamycin enhances the in vitro and in vivo calcified tissue-forming capacity of SCAP. SCAP were fabricated as scaffold-free spheroids and were induced into forming calcified 3D constructs. The calcified density of the spheroidal constructs was enhanced when the spheroids were pretreated with LY294402 and rapamycin. Conclusions: Our findings indicate that the suppression of PI3K-AKT-mTOR signal pathway plays a role in not only enhancing the in vivo and in vitro osteogenic/dentinogenic differentiation of SCAP, but also promoting the calcification of scaffold-free SCAP-based calcified constructs. These findings suggest that a suppressive regulation of PI3K-AKT-mTOR signal pathway is a novel approach for SCAP-based bone and dentin regeneration.

    DOI: 10.1186/s13287-018-1077-9

  • Pamidronate decreases bilirubin-impaired cell death and improves dentinogenic dysfunction of stem cells from human deciduous teeth 査読

    Haruyoshi Yamaza, Soichiro Sonoda, Kazuaki Nonaka, Toshio Kukita, Takayoshi Yamaza

    Stem Cell Research and Therapy   9 ( 1 )   2018年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Background: Hyperbilirubinemia that occurs in pediatric liver diseases such as biliary atresia can result in the development of not only jaundice in the brain, eyes, and skin, but also tooth abnormalities including green pigmentation and dentin hypoplasia in the developing teeth. However, hyperbilirubinemia-induced tooth impairments remain after liver transplantation. No effective dental management to prevent hyperbilirubinemia-induced tooth impairments has been established. Methods: In this study, we focused on pamidronate, which is used to treat pediatric osteopenia, and investigated its effects on hyperbilirubinemia-induced tooth impairments. We cultured stem cells from human exfoliated deciduous teeth (SHED) under high and low concentrations of unconjugated bilirubin in the presence or absence of pamidronate. We then analyzed the effects of pamidronate on the cell death, associated signal pathways, and dentinogenic function in SHED. Results: We demonstrated that a high concentration of unconjugated bilirubin induced cell death in SHED via the mitochondrial pathway, and this was associated with the suppression of AKT and extracellular signal-related kinase 1 and 2 (ERK1/2) signal pathways and activation of the nuclear factor kappa B (NF-κB) signal pathway. The high concentration of unconjugated bilirubin impaired the in vitro and in vivo dentinogenic capacity of SHED, but not the low concentration. We then demonstrated that pamidronate decreased the bilirubin-induced cell death in SHED via the altered AKT, ERK1/2, and NF-κB signal pathways and recovered the bilirubin-impaired dentinogenic function of SHED. Conclusions: Our findings suggest that pamidronate may prevent tooth abnormalities in pediatric patients with hyperbilirubinemia.

    DOI: 10.1186/s13287-018-1042-7

  • Bilirubin reversibly affects cell death and odontogenic capacity in stem cells from human exfoliated deciduous teeth 査読

    H. Yamaza, E. Tomoda, S. Sonoda, K. Nonaka, T. Kukita, T. Yamaza

    Oral Diseases   24 ( 5 )   809 - 819   2018年7月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Objective: Hyperbilirubinemia in patients with biliary atresia causes deciduous tooth injuries such as green pigmentation and dentin hypoplasia. In patients with biliary atresia who received liver transplantation, tooth structure appears to be recovered radiographically. Nevertheless, little is known about cellular mechanisms underlying bilirubin-induced damage and suppression of deciduous tooth formation. In this study, we examined the effects of bilirubin in stem cells from human exfoliated deciduous teeth (SHED) in vitro. Materials and Methods: SHED were cultured under exposure to excess of bilirubin and then interruption of bilirubin stimulation. Results: Bilirubin induced cell death and inhibited the odontogenic capacity of SHED by suppressing AKT and extracellular signal-regulated kinase 1 and 2 (ERK1/2) pathways and enhancing nuclear factor kappa B p65 (NF-κB p65) pathway. The interruption of bilirubin stimulation reduced cell death and recovered the inhibited odontogenic capacity of bilirubin-damaged SHED. The bilirubin interruption also normalized the impaired AKT, ERK1/2, and NF-κB p65 signaling pathways. Conclusion: These findings suggest that tooth hypodontia in patients with hyperbilirubinemia might be due to bilirubin-induced cell death and dentinogenic dysfunction of odontogenic stem cells via AKT, ERK1/2, and NF-κB pathways and also suggested that bilirubin-induced impairments in odontogenic stem cells were reversible when bilirubin stimulation is interrupted.

    DOI: 10.1111/odi.12827

  • Unique Osteoblast-Specific Cell-Surface Antigen Useful for Odontoblast Ontology and Dentin Regeneration 査読

    Tamer Badawy, Yukari Kyumoto Nakamura, Norihisa Uehara, Jingqi Zhang, Hidenobu Hiura, Soichiro Sonoda, Takayoshi Yamaza, Akiko Kukita, Toshio Kukita

    Oral Health & Dental Science   2 ( 2 )   1 - 7   2018年6月

     詳細を見る

    記述言語:その他   掲載種別:研究論文(学術雑誌)  

    DOI: 10.33425/2639-9490.1018

  • Interferon-gamma improves impaired dentinogenic and immunosuppressive functions of irreversible pulpitis-derived human dental pulp stem cells 査読

    Soichiro Sonoda, Haruyoshi Yamaza, Lan Ma, Yosuke Tanaka, Erika Tomoda, Reona Aijima, Kazuaki Nonaka, Toshio Kukita, Songtao Shi, Fusanori Nishimura, Takayoshi Yamaza

    Scientific Reports   6   2016年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Clinically, irreversible pulpitis is treated by the complete removal of pulp tissue followed by replacement with artificial materials. There is considered to be a high potential for autologous transplantation of human dental pulp stem cells (DPSCs) in endodontic treatment. The usefulness of DPSCs isolated from healthy teeth is limited. However, DPSCs isolated from diseased teeth with irreversible pulpitis (IP-DPSCs) are considered to be suitable for dentin/pulp regeneration. In this study, we examined the stem cell potency of IP-DPSCs. In comparison with healthy DPSCs, IP-DPSCs expressed lower colony-forming capacity, population-doubling rate, cell proliferation, multipotency, in vivo dentin regeneration, and immunosuppressive activity, suggesting that intact IP-DPSCs may be inadequate for dentin/pulp regeneration. Therefore, we attempted to improve the impaired in vivo dentin regeneration and in vitro immunosuppressive functions of IP-DPSCs to enable dentin/pulp regeneration. Interferon gamma (IFN-γ) treatment enhanced in vivo dentin regeneration and in vitro T cell suppression of IP-DPSCs, whereas treatment with tumor necrosis factor alpha did not. Therefore, these findings suggest that IFN-γ may be a feasible modulator to improve the functions of impaired IP-DPSCs, suggesting that autologous transplantation of IFN-γ-accelerated IP-DPSCs might be a promising new therapeutic strategy for dentin/pulp tissue engineering in future endodontic treatment.

    DOI: 10.1038/srep19286

  • Transplantation of mesenchymal stem cells ameliorates secondary osteoporosis through interleukin-17-impaired functions of recipient bone marrow mesenchymal stem cells in MRL/lpr mice 査読

    Lan Ma, Reona Aijima, Yoshihiro Hoshino, Haruyoshi Yamaza, Erika Tomoda, Yosuke Tanaka, Soichiro Sonoda, Guangtai Song, Wei Zhao, Kazuaki Nonaka, Songtao Shi, Takayoshi Yamaza

    Stem Cell Research and Therapy   6 ( 1 )   2015年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Introduction: Secondary osteoporosis is common in systemic lupus erythematosus and leads to a reduction in quality of life due to fragility fractures, even in patients with improvement of the primary disorder. Systemic transplantation of mesenchymal stem cells could ameliorate bone loss and autoimmune disorders in a MRL/lpr mouse systemic lupus erythematosus model, but the detailed therapeutic mechanism of bone regeneration is not fully understood. In this study, we transplanted human bone marrow mesenchymal stem cells (BMMSCs) and stem cells from exfoliated deciduous teeth (SHED) into MRL/lpr mice and explored their therapeutic mechanisms in secondary osteoporotic disorders of the systemic lupus erythematosus model mice. Methods: The effects of systemic human mesenchymal stem cell transplantation on bone loss of MRL/lpr mice were analyzed in vivo and ex vivo. After systemic human mesenchymal stem cell transplantation, recipient BMMSC functions of MRL/lpr mice were assessed for aspects of stemness, osteogenesis and osteoclastogenesis, and a series of co-culture experiments under osteogenic or osteoclastogenic inductions were performed to examine the efficacy of interleukin (IL)-17-impaired recipient BMMSCs in the bone marrow of MRL/lpr mice. Results: Systemic transplantation of human BMMSCs and SHED recovered the reduction in bone density and structure in MRL/lpr mice. To explore the mechanism, we found that impaired recipient BMMSCs mediated the negative bone metabolic turnover by enhanced osteoclastogenesis and suppressed osteoblastogenesis in secondary osteoporosis of MRL/lpr mice. Moreover, IL-17-dependent hyperimmune conditions in the recipient bone marrow of MRL/lpr mice damaged recipient BMMSCs to suppress osteoblast capacity and accelerate osteoclast induction. To overcome the abnormal bone metabolism, systemic transplantation of human BMMSCs and SHED into MRL/lpr mice improved the functionally impaired recipient BMMSCs through IL-17 suppression in the recipient bone marrow and then maintained a regular positive bone metabolism via the balance of osteoblasts and osteoclasts. Conclusions: These findings indicate that IL-17 and recipient BMMSCs might be a therapeutic target for secondary osteoporosis in systemic lupus erythematosus.

    DOI: 10.1186/s13287-015-0091-4

▼全件表示

講演・口頭発表等

  • 胸腺間葉系ストロマ細胞による内在性制御性T細胞の産生メカニズムの解明

    園田 聡一朗、久本 由香里、加藤 大樹、上 原 範久、山座 孝義

    第65回歯科基礎医学会学術大会  2023年9月 

     詳細を見る

    開催年月日: 2023年9月

    記述言語:日本語   会議種別:口頭発表(一般)  

    開催地:日本大学歯学部(東京都千代田区)   国名:日本国  

  • 乳歯幹細胞の細胞外小胞による宿主間葉系幹細胞を標的とした全身性エリテマトーデス治療メカニズムの解明

    園田 聡一朗, 山座 孝義

    第21回日本再生医療学会総会  2022年3月 

     詳細を見る

    開催年月日: 2022年3月

    記述言語:日本語  

    国名:その他  

  • 乳歯幹細胞の細胞外小胞による宿主間葉系幹細胞を標的とした全身性エリテマトーデス治療メカニズムの解明

    園田 聡一朗, 山座 孝義

    第21回日本再生医療学会総会  2022年3月 

     詳細を見る

    開催年月日: 2022年3月

    記述言語:日本語   会議種別:口頭発表(一般)  

    researchmap

  • Extracellular vesicles-mediated novel therapeutic mechanism of deciduous toot pulp stem cell-based therapy for systemic lupus erythematosus

    園田 聡一朗, 山座 孝義

    第69回国際歯科研究学会日本部会総会  2021年10月 

     詳細を見る

    開催年月日: 2021年10月

    記述言語:英語  

    国名:その他  

    Extracellular vesicles-mediated novel therapeutic mechanism of deciduous toot pulp stem cell-based therapy for systemic lupus erythematosus

  • 乳歯幹細胞の細胞外小胞を介した全身性エリテマトーデスの新規治療メカニズムの解明

    園田 聡一朗, 山座 孝義

    第63回歯科基礎医学会学術大会  2021年10月 

     詳細を見る

    開催年月日: 2021年10月

    記述言語:英語  

    国名:その他  

    Novel therapeutic mechanism of extracellular vesicles-mediated deciduous toot pulp stem cell-based therapy for systemic lupus erythematosus

  • 胆道閉鎖症患児由来乳歯幹細胞の細胞移植治療効果低下をもたらす機構の解析

    園田 聡一朗, 村田 早羅, 加藤 大樹, 久本 由香里, 上原 範久, 久木田 敏夫, 山座 孝義

    第62回歯科基礎医学会学術大会  2020年9月 

     詳細を見る

    開催年月日: 2020年10月

    記述言語:その他  

    国名:その他  

  • 胆道閉鎖症患児由来乳歯幹細胞の細胞移植治療効果低下をもたらす機構の解析

    園田 聡一朗, 村田 早羅, 加藤 大樹, 久本 由香里, 上原 範久, 久木田 敏夫, 山座 孝義

    第62回歯科基礎医学会学術大会  2020年9月 

     詳細を見る

    開催年月日: 2020年10月

    researchmap

  • 胆道閉鎖症由来乳歯幹細胞の特性とその肝再生能力

    園田 聡一朗, 村田 早羅, 山座 孝義

    日本再生医療学会  2020年5月 

     詳細を見る

    開催年月日: 2020年5月

    記述言語:日本語   会議種別:口頭発表(一般)  

    開催地:オンライン   国名:日本国  

  • 胆道閉鎖症患児に対する自家乳歯幹細胞移植治療を目指した基礎研究

    園田 聡一朗, 村田 早羅, 久本 由香里, 上原 範久, 久木田 敏夫, 山座 孝義

    歯科基礎医学会  2019年10月 

     詳細を見る

    開催年月日: 2019年10月

    記述言語:日本語   会議種別:口頭発表(一般)  

    開催地:東京医科歯科大学   国名:日本国  

  • Improvement of dentinogenesis of Irreversible pulpitis-derived DPSCs. 国際会議

    Soichiro Sonoda, Takayoshi Yamaza, Yusuke Makino, La Ma, Toshio Kukita.

    The 59th Annual Meeting of Japanese Association for Dental Research.  2011年6月 

     詳細を見る

    開催年月日: 2019年6月

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

    開催地:Hiroshima   国名:日本国  

  • 非可逆性炎歯髄由来幹細胞は歯髄再生治療に応用可能か?

    園田聡一朗, 山座孝義, 久木田敏夫.

    第57回歯科基礎医学会学術大会・総会.  2015年9月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:日本語  

    国名:日本国  

  • Interferon-gamma improves impaired dentinogenic function and immunosuppressive properties in irreversible pulpitis-derived human dental pulp stem cells. 国際会議

    Soichiro Sonoda, Erika Tomoda, Yosuke Tanaka, Fusanori Nishimura, Takayoshi Yamaza.

    he 63rd Annual Meeting of Japanese Association for Dental Research.  2015年10月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:英語  

    開催地:Fukuoka   国名:日本国  

  • 炎症性サイトカイン刺激による不可逆性歯髄炎組織由来歯髄幹細胞の象牙質形成能ならびに免疫抑制能の改善.

    園田聡一朗, 山座孝義, 西村英紀.

    日本歯周病学会九州五大学・日本臨床歯周病学会九州支部 合同研修会.  2015年11月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:日本語  

    国名:日本国  

  • Patient-derived dental pulp stem cells based regeneration of dentin/pulp-complex. 国際会議

    Soichiro Sonoda, Takayoshi Yamaza, Fusanori Nishimura.

    Kyudai Oral Bioscience  2016年2月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:英語  

    国名:日本国  

  • 不可逆性歯髄炎組織由来歯髄幹細胞を応用した象牙質/歯髄複合体の再生.

    園田聡一朗, 山座孝義, 西村英紀.

    第144回日本歯科保存学会春季学術大会.  2016年6月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:日本語  

    国名:日本国  

  • 一酸化窒素によるラット歯髄幹細胞の象牙芽細胞分化促進.

    園田聡一朗, 山座孝義, 友田恵利佳, 田中陽介, 久木田敏夫.

    第58回歯科基礎医学会学術大会・総会.  2016年8月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:日本語  

    開催地:札幌コンベンションセンター. 北海道.   国名:日本国  

  • 一酸化窒素によるラット歯髄幹細胞の象牙質形成促進.

    園田聡一朗, 山座孝義, 久木田敏夫.

    第73回日本解剖学会九州支部学術集会.  2017年10月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:日本語  

    開催地:都久志会館. 福岡.   国名:日本国  

  • 外因性の一酸化窒素刺激は歯髄幹細胞の象牙芽細胞分化を促進する

    園田聡一朗, 山座孝義, 西村英紀

    歯科保存学会2018春季大会  2018年6月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:日本語  

    国名:日本国  

  • 乳歯幹細胞の単離時に血清濃度が与える影響について

    園田 聡一朗, 大谷 憲司, 山座 孝義

    第60回日本歯科基礎医学会総会  2018年9月 

     詳細を見る

    開催年月日: 2019年6月

    記述言語:日本語  

    国名:日本国  

  • 骨転移性乳癌細胞由来細胞外小胞に内包されるmiR-92a-3pは成熟破骨細胞の生存を促進する(Bone metastatic mammary tumor cell-derived extracellular vesicles promote mature osteoclast longevity)

    上原 範久, 久本 由香里, 三上 剛和, 園田 聡一朗, 山座 孝義, 久木田 敏夫

    日本癌学会総会記事  2023年9月  (一社)日本癌学会

     詳細を見る

    記述言語:英語  

  • 胸腺間葉系ストロマ細胞による内在性制御性T細胞の産生メカニズムの解明

    園田 聡一朗, 久本 由香里, 加藤 大樹, 上原 範久, 山座 孝義

    Journal of Oral Biosciences Supplement  2023年9月  (一社)歯科基礎医学会

     詳細を見る

    記述言語:日本語  

  • 根尖歯乳頭組織由来幹細胞に発現する転写因子PITX2の機能解析

    久本 由香里, 園田 聡一朗, 加藤 大樹, 上原 範久, 山座 孝義

    Journal of Oral Biosciences Supplement  2023年9月  (一社)歯科基礎医学会

     詳細を見る

    記述言語:日本語  

  • 小児外科における最先端医療の現状と展望【International】ヒト脱落乳歯歯髄幹細胞移植によるhypoganglionosisに対する新規治療法開発

    吉丸 耕一朗, 園田 聡一朗, 山内 恵利佳, 河野 淳, 松浦 俊治, 山座 孝義, 小田 義直, 田尻 達郎, 田口 智章

    日本外科学会定期学術集会抄録集  2022年4月  (一社)日本外科学会

     詳細を見る

    記述言語:日本語  

▼全件表示

MISC

  • 疾患特異的乳歯幹細胞モデルによる胆管症の病因解明

    園田 聡一朗, 吉丸 耕一朗, 田口 智章, 山座 孝義

    細胞   56 ( 10 )   747 - 749   2024年9月   ISSN:1346-7557

     詳細を見る

    記述言語:日本語   出版者・発行元:(株)ニュー・サイエンス社  

    胆道閉鎖症Biliary Atresia(BA)は,肝外胆管の閉塞によって胆汁がうっ滞する難病であるが,その病因は不明である。乳歯歯髄の体性幹細胞である乳歯幹細胞Stem Cells from Exfoliated Deciduous Teeth(SHED)は,その発生学的起源ゆえ,胎児組織幹細胞でもある。そのため,胎児期の母体内で生じる大きなエピジェネティック変化を記憶している可能性が推測される。四塩化炭素誘導性肝障害モデルマウスを用いたSHED注入実験において,SHEDは,レシピエント肝組織で肝細胞様細胞と胆管様構造を呈する胆管上皮細胞様細胞に分化した。しかし,BA特異的SHEDは,肝細胞様細胞に分化したが,胆管上皮細胞様細胞には分化しなかった。また,ドナー由来の肝芽細胞もレシピエント肝組織で確認できた。したがって,BAの発症機序として,母体内の誘発因子が,肝芽細胞から胆管形成を抑制するのではないかと仮説した。現在,筆者らは,疾患特異的SHEDを用いた研究モデルで,BAなどの胆管症発症分子機序の解明を試みている。(著者抄録)

  • Cutting-edge regenerative therapy for Hirschsprung disease and its allied disorders(タイトル和訳中)

    Yoshimaru Koichiro, Matsuura Toshiharu, Uchida Yasuyuki, Sonoda Soichiro, Maeda Shohei, Kajihara Keisuke, Kawano Yuki, Shirai Takeshi, Toriigahara Yukihiro, Kalim Alvin Santoso, Zhang Xiu-Ying, Takahashi Yoshiaki, Kawakubo Naonori, Nagata Kouji, Yamaza Haruyoshi, Yamaza Takayoshi, Taguchi Tomoaki, Tajiri Tatsuro

    Surgery Today   54 ( 9 )   977 - 994   2024年9月   ISSN:0941-1291

     詳細を見る

    記述言語:英語   出版者・発行元:シュプリンガー・ジャパン(株)  

  • Extracellular vesicles rejuvenate the microenvironmental modulating function of recipient tissue-specific mesenchymal stem cells in osteopenia treatment. 査読

    Soichiro Sonoda, Takayoshi Yamaza

    Frontiers in endocrinology   2023年3月

     詳細を見る

    記述言語:英語   掲載種別:記事・総説・解説・論説等(学術雑誌)  

    Systemic transplantation of mesenchymal stem cells (MSCs), such as bone marrow MSCs (BMMSCs) and stem cells from human exfoliated deciduous teeth (SHED), is considered a prominent treatment for osteopenia. However, the mechanism of action of the transplanted MSCs has been poorly elucidated. In the recipient target tissue, including bone and bone marrow, only a few donor MSCs can be detected, suggesting that the direct contribution of donor MSCs may not be expected for osteopenia treatment. Meanwhile, secretomes, especially contents within extracellular vesicles (EVs) released from donor MSCs (MSC-EVs), play key roles in the treatment of several diseases. In this context, administrated donor MSC-EVs may affect bone-forming function of recipient cells. In this review, we discuss how MSC-EVs contribute to bone recovery recipient tissue in osteopenia. We also summarize a novel mechanism of action of systemic administration of SHED-derived EVs (SHED-EVs) in osteopenia. We found that reduced telomerase activity in recipient BMMSCs caused the deficiency of microenvironmental modulating function, including bone and bone marrow-like niche formation and immunomodulation in estrogen-deficient osteopenia model mice. Systemic administration of SHED-EVs could exert therapeutic effects on bone reduction via recovering the telomerase activity, leading to the rejuvenation of the microenvironmental modulating function in recipient BMMSCs, as seen in systemic transplantation of SHED. RNase-preconditioned donor SHED-EVs diminished the therapeutic benefits of administrated SHED-EVs in the recipient osteopenia model mice. These facts suggest that MSC-EV therapy targets the recipient BMMSCs to rejuvenate the microenvironmental modulating function via telomerase activity, recovering bone density. We then introduce future challenges to develop the reproducible MSC-EV therapy in osteopenia.

    DOI: 10.3389/fendo.2023.1151429

  • 【疾患と免疫の新たな関係】歯髄幹細胞を用いたSLEの治療戦略

    山座 孝義, 園田 聡一朗

    臨床免疫・アレルギー科   77 ( 5 )   575 - 583   2022年5月   ISSN:1881-1930

     詳細を見る

    記述言語:日本語   出版者・発行元:(有)科学評論社  

  • A New Target of Dental Pulp-Derived Stem Cell-Based Therapy on Recipient Bone Marrow Niche in Systemic Lupus Erythematosus

    Soichiro Sonoda, Takayoshi Yamaza

    International Journal of Molecular Sciences   2022年3月

     詳細を見る

    記述言語:その他  

    Recent advances in mesenchymal stem/stromal cell (MSC) research have led us to con-sider the feasibility of MSC-based therapy for various diseases. Human dental pulp-derived MSCs (hDPSCs) have been identified in the dental pulp tissue of deciduous and permanent teeth, and they exhibit properties with self-renewal and in vitro multipotency. Interestingly, hDPSCs exhibit superior immunosuppressive functions toward immune cells, especially T lymphocytes, both in vitro and in vivo. Recently, hDPSCs have been shown to have potent immunomodulatory functions in treating systemic lupus erythematosus (SLE) in the SLE MRL/lpr mouse model. However, the mechanisms underlying the immunosuppressive efficacy of hDPSCs remain unknown. This review aims to introduce a new target of hDPSC-based therapy on the recipient niche function in SLE.

    DOI: 10.3390/ijms23073479

  • Properties and Possibilities of Human Dental Pulp-Derived Stem Cells.

    Soichiro Sonoda, Erika Tomoda, Yosuke Tanaka, Takayoshi Yamaza.

    2015年2月

     詳細を見る

    記述言語:英語  

▼全件表示

所属学協会

  • 日本歯周病学会

  • 日本歯科保存学会

  • 歯科基礎医学会

  • Japanese Association of Denal Research

  • International Association of Dental research

  • 日本再生医療学会

  • 歯科基礎医学会

      詳細を見る

  • 日本歯科保存学会

      詳細を見る

  • 日本歯周病学会

      詳細を見る

  • 日本再生医療学会

      詳細を見る

  • Japanese Association of Denal Research

      詳細を見る

  • International Association of Dental research

      詳細を見る

▼全件表示

委員歴

  • 九州大学   OSCE委員  

    2021年4月 - 2022年3月   

      詳細を見る

共同研究・競争的資金等の研究課題

  • 胸腺髄質線維芽細胞による制御性T細胞の生成制御因子の探究

    研究課題/領域番号:24K12873  2024年4月 - 2027年3月

    科学研究費助成事業  基盤研究(C)

    園田 聡一朗, 山座 孝義

      詳細を見る

    資金種別:科研費

    制御性T細胞(Treg)は「免疫寛容」を実装した司令塔である。胸腺で生成される内在性Treg(nTreg)には自己抗体産生の制御に関わるサブセットが報告されており、nTregが自己免疫疾患の発症に重要な働きをもつことが示唆されている。しかし、nTregの生成メカニズムについては不明な点が多く残されている。髄質線維芽細胞(mFB)が間葉系ストロマ細胞として同定され、mFBがnTregの生成に関わる微小環境を形成する細胞として注目されている。本研究では、mFBによるnTreg生成の分子メカニズムの解明を試み、mFBを標的とした自己免疫疾患の新たな治療法の開発に向けた学術的基盤の構築を目指す。

    CiNii Research

  • 口腔がんの転移をもたらすリンパ節線維芽細胞様細網細胞の変異メカニズムの解明

    2023年4月 - 2027年3月

      詳細を見る

    担当区分:研究分担者 

  • 口腔がんの転移をもたらすリンパ節線維芽細胞様細網細胞の変異メカニズムの解明

    研究課題/領域番号:23K27762  2023年4月 - 2027年3月

    科学研究費助成事業  基盤研究(B)

    山座 孝義, 園田 聡一朗, 久本 由香里

      詳細を見る

    資金種別:科研費

    口腔がんのリンパ節転移は不顕性に進行し、転移後に発見されるケースが多い。患者のQOL向上のために、低侵襲的な口腔がんのリンパ節転移に対する新規治療法が必要である。センチネルリンパ節は、がん遠隔転移の防波堤である。がん転移センチネルリンパ節では、線維芽細胞様細網細胞による物質輸送系の線維化が起こり、T細胞による抗腫瘍免疫機能が低下すると考えられている。本研究では、口腔がん細胞が線維芽細胞様細網細胞を介して抗腫瘍免疫機能を抑制するメカニズムおよびセンチネルリンパ節への転移機序を解明し、センチネルリンパ節転移に対する非外科的治療法の学術的基盤の確立を目指す。

    CiNii Research

  • 口腔がんの転移をもたらすリンパ節線維芽細胞様細網細胞の変異メカニズムの解明

    研究課題/領域番号:23H03071  2023年 - 2026年

    日本学術振興会  科学研究費助成事業  基盤研究(B)

      詳細を見る

    担当区分:研究分担者  資金種別:科研費

  • 疾患特異的乳歯幹細胞モデルによる胆管障害症の病態解明

    研究課題/領域番号:22K19565  2022年 - 2024年

    日本学術振興会  科学研究費助成事業  挑戦的研究(萌芽)

    山座 孝義, 園田 聡一朗

      詳細を見る

    担当区分:研究分担者  資金種別:科研費

    本研究の目的は「乳歯幹細胞を用いた胆道上皮細胞分化誘導法を開発し、胆管障害患児由来の疾患特異的な乳歯幹細胞を用いて新規ex vivo解析モデルを構築すること」である。胆管障害とは胆汁の排泄障害を呈する進行性の慢性肝疾患であるが、原因・発症機構が未解明である。肝移植以外の根治治療がなく、代替となる有効な治療法の開発が責務である。申請者らは乳歯幹細胞の肝細胞分化について多数の研究成果を報告してきた。本研究ではその知見を生かし、乳歯幹細胞による胆道上皮細胞分化誘導法を開発する。さらに胆管障害の患児より乳歯幹細胞を単離し、胆管上皮細胞への分化能や機能を比較検討することで胆管障害発症機序の解明を目指す。

    CiNii Research

  • 歯髄幹細胞移植による胸腺機能の改善を介した免疫調節機序の研究

    2021年5月 - 2024年3月

      詳細を見る

    担当区分:研究代表者 

  • 歯髄幹細胞移植による胸腺機能の改善を介した免疫調節機序の研究

    研究課題/領域番号:21K16932  2021年 - 2023年

    日本学術振興会  科学研究費助成事業  若手研究

      詳細を見る

    担当区分:研究代表者  資金種別:科研費

  • 歯髄幹細胞移植による胸腺機能の改善を介した免疫調節機序の研究

    研究課題/領域番号:21K16932  2021年 - 2023年

    日本学術振興会  科学研究費助成事業  若手研究

      詳細を見る

    資金種別:科研費

  • 胆道閉鎖症患児由来の乳歯幹細胞を用いた病態解析および疾患治療のための基礎研究

    2019年4月 - 2021年3月

      詳細を見る

    担当区分:研究代表者 

    成体幹細胞が生体の恒常性の維持と組織修復に働く。一方で、成体幹細胞に生じた変異 が疾患を誘発することも知られている。幹細胞は、外因性に受けた障害をエピジェネティ ックな変異として遺伝情報に記憶し、その組織発生や再生・修復の能力が障害され、病態へ 発展することが報告されている。従って、その成体幹細胞の障害記憶を解析することが病 因解明へ繋がると考えられる。本研究では、疾患由来の乳歯幹細胞 stem cells from exfoliated deciduous teeth(SHED)の間葉系幹細胞 mesenchymal stem cells(MSCs)としての障害記憶 力を利用して、胆道閉鎖症の病因解明ならびに自家幹細胞移植治療を目指す研究を行う。

  • 胆道閉鎖症患児由来の乳歯幹細胞を用いた病態解析および疾患治療のための基礎研究

    研究課題/領域番号:19K18945  2019年 - 2020年

    日本学術振興会  科学研究費助成事業  若手研究

      詳細を見る

    担当区分:研究代表者  資金種別:科研費

  • 患者由来乳歯幹細胞を用いた肝疾患病因解析と幹細胞治療

    研究課題/領域番号:17J03382  2017年 - 2018年

    日本学術振興会  科学研究費助成事業  基盤研究(C)

      詳細を見る

    資金種別:科研費

  • 患者由来乳歯幹細胞を用いた肝疾患病因解析と幹細胞治療

    研究課題/領域番号:17J0382  2017年 - 2018年

    日本学術振興会  科学研究費助成事業  特別研究員奨励費

      詳細を見る

    担当区分:研究代表者  資金種別:科研費

▼全件表示

教育活動概要

  • 1. 学部学生(2年生)に対する解剖学実習、口腔解剖学実習、口腔組織学演習
    2. 学部学生(4年生)に対するアーリーイクスポージャー
    3. 学部学生(5年生)に対するリサーチイクスポージャー
    4. 大学院学生に対する研究指導
    5. 学部学生に対する研究指導

担当授業科目

  • 分子口腔解剖学(高年次) D

    2023年12月 - 2024年2月   冬学期

  • 解剖学実習2

    2023年12月 - 2024年2月   冬学期

  • 口腔解剖学4

    2023年12月 - 2024年2月   冬学期

  • 口腔組織学3

    2023年12月 - 2024年2月   冬学期

  • 分子口腔解剖学(低年次) D

    2023年12月 - 2024年2月   冬学期

  • 分子口腔解剖学(コア) D

    2023年12月 - 2024年2月   冬学期

  • 分子口腔解剖学(高年次) C

    2023年10月 - 2023年12月   秋学期

  • 解剖学実習1

    2023年10月 - 2023年12月   秋学期

  • 口腔解剖学3

    2023年10月 - 2023年12月   秋学期

  • 口腔組織学2

    2023年10月 - 2023年12月   秋学期

  • 分子口腔解剖学(低年次) C

    2023年10月 - 2023年12月   秋学期

  • 分子口腔解剖学(コア) C

    2023年10月 - 2023年12月   秋学期

  • 分子口腔解剖学(高年次) B

    2023年6月 - 2023年8月   夏学期

  • 口腔組織学1

    2023年6月 - 2023年8月   夏学期

  • 解剖学2

    2023年6月 - 2023年8月   夏学期

  • 口腔解剖学2

    2023年6月 - 2023年8月   夏学期

  • 分子口腔解剖学(低年次) B

    2023年6月 - 2023年8月   夏学期

  • 分子口腔解剖学(コア) B

    2023年6月 - 2023年8月   夏学期

  • 分子口腔解剖学(高年次) A

    2023年4月 - 2023年6月   春学期

  • 発生学

    2023年4月 - 2023年6月   春学期

  • 解剖学1

    2023年4月 - 2023年6月   春学期

  • 口腔解剖学1

    2023年4月 - 2023年6月   春学期

  • 分子口腔解剖学(低年次) A

    2023年4月 - 2023年6月   春学期

  • 分子口腔解剖学(コア) A

    2023年4月 - 2023年6月   春学期

  • 解剖学実習2

    2022年12月 - 2023年2月   冬学期

  • 分子口腔解剖学(高年次) D

    2022年12月 - 2023年2月   冬学期

  • 分子口腔解剖学(コア) D

    2022年12月 - 2023年2月   冬学期

  • 分子口腔解剖学(低年次) D

    2022年12月 - 2023年2月   冬学期

  • 口腔組織学3

    2022年12月 - 2023年2月   冬学期

  • 口腔解剖学4

    2022年12月 - 2023年2月   冬学期

  • 解剖学実習1

    2022年10月 - 2022年12月   秋学期

  • 分子口腔解剖学(高年次) C

    2022年10月 - 2022年12月   秋学期

  • 分子口腔解剖学(コア) C

    2022年10月 - 2022年12月   秋学期

  • 分子口腔解剖学(低年次) C

    2022年10月 - 2022年12月   秋学期

  • 口腔組織学2

    2022年10月 - 2022年12月   秋学期

  • 口腔解剖学3

    2022年10月 - 2022年12月   秋学期

  • 口腔組織学1

    2022年6月 - 2022年8月   夏学期

  • 分子口腔解剖学(高年次) B

    2022年6月 - 2022年8月   夏学期

  • 分子口腔解剖学(コア) B

    2022年6月 - 2022年8月   夏学期

  • 分子口腔解剖学(低年次) B

    2022年6月 - 2022年8月   夏学期

  • 口腔解剖学2

    2022年6月 - 2022年8月   夏学期

  • 解剖学2

    2022年6月 - 2022年8月   夏学期

  • 発生学

    2022年4月 - 2022年6月   春学期

  • 分子口腔解剖学(高年次) A

    2022年4月 - 2022年6月   春学期

  • 分子口腔解剖学(コア) A

    2022年4月 - 2022年6月   春学期

  • 分子口腔解剖学(低年次) A

    2022年4月 - 2022年6月   春学期

  • 口腔解剖学1

    2022年4月 - 2022年6月   春学期

  • 解剖学1

    2022年4月 - 2022年6月   春学期

  • 分子口腔解剖学(高年次) D

    2021年12月 - 2022年2月   冬学期

  • 分子口腔解剖学(低年次) D

    2021年12月 - 2022年2月   冬学期

  • 分子口腔解剖学(コア) D

    2021年12月 - 2022年2月   冬学期

  • 口腔組織学2

    2021年10月 - 2022年3月   後期

  • 分子口腔解剖学(高年次) C

    2021年10月 - 2021年12月   秋学期

  • 分子口腔解剖学(低年次) C

    2021年10月 - 2021年12月   秋学期

  • 分子口腔解剖学(コア) C

    2021年10月 - 2021年12月   秋学期

  • 分子口腔解剖学(高年次) B

    2021年6月 - 2021年8月   夏学期

  • 分子口腔解剖学(低年次) B

    2021年6月 - 2021年8月   夏学期

  • 分子口腔解剖学(コア) B

    2021年6月 - 2021年8月   夏学期

  • 口腔解剖学

    2021年4月 - 2022年3月   通年

  • 解剖学

    2021年4月 - 2022年3月   通年

  • 口腔組織学1

    2021年4月 - 2021年9月   前期

  • 発生学

    2021年4月 - 2021年9月   前期

  • 分子口腔解剖学(高年次) A

    2021年4月 - 2021年6月   春学期

  • 分子口腔解剖学(低年次) A

    2021年4月 - 2021年6月   春学期

  • 分子口腔解剖学(コア) A

    2021年4月 - 2021年6月   春学期

  • 口腔組織学II

    2020年10月 - 2021年3月   後期

  • 口腔解剖学

    2020年4月 - 2021年3月   通年

  • 解剖学

    2020年4月 - 2021年3月   通年

  • 発生学

    2020年4月 - 2020年9月   前期

  • 口腔組織学1

    2020年4月 - 2020年9月   前期

▼全件表示

FD参加状況

  • 2019年4月   役割:参加   名称:平成31年度 第1回全学FD(新任教員の研修)

    主催組織:全学

その他教育活動及び特記事項

  • 2022年  その他特記事項  歯学府学生への研究指導

     詳細を見る

    歯学府学生への研究指導

  • 2021年  その他特記事項  歯学部学生への研究指導

     詳細を見る

    歯学部学生への研究指導

  • 2020年  その他特記事項  歯学部学生への研究指導

     詳細を見る

    歯学部学生への研究指導

  • 2019年  その他特記事項  歯学部学生への研究指導

     詳細を見る

    歯学部学生への研究指導

学内運営に関わる各種委員・役職等

  • 2024年4月 - 2025年3月   その他 OSCE委員

  • 2023年4月 - 2024年3月   その他 OSCE委員

  • 2021年4月 - 2022年3月   その他 OSCE委員

  • 2020年4月 - 2021年3月   その他 OSCE委員