研究者業績

内堀 亮介

ウチボリ リヨウスケ  (ryosuke Uchibori)

基本情報

所属
自治医科大学 医学部難治性疾患遺伝子細胞治療開発講座 講師
学位
博士(医学)(自治医科大学)

J-GLOBAL ID
201401092855808609
researchmap会員ID
B000237426

外部リンク

論文

 46
  • Yoshihide Sehara, Yuki Hashimotodani, Ryota Watano, Kenji Ohba, Ryosuke Uchibori, Kuniko Shimazaki, Kensuke Kawai, Hiroaki Mizukami
    Molecular neurobiology 2024年4月27日  
    It is established that neurogenesis of dentate gyrus is increased after ischemic insult, although the regulatory mechanisms have not yet been elucidated. In this study, we focused on Ezh2 which suppresses gene expression through catalyzing trimethylation of lysine 27 of histone 3. Male gerbils were injected with adeno-associated virus (AAV) carrying shRNA targeting to Ezh2 into right dentate gyrus 2 weeks prior to forebrain ischemia. One week after ischemia, animals were injected with thymidine analogue to label proliferating cells. Three weeks after ischemia, animals were killed for histological analysis. AAV-mediated knockdown of Ezh2 significantly decreased the ischemia-induced increment of proliferating cells, and the proliferated cells after ischemia showed significantly longer migration from subgranular zone (SGZ), compared to the control group. Furthermore, the number of neural stem cells in SGZ significantly decreased after ischemia with Ezh2 knockdown group. Of note, Ezh2 knockdown did not affect the number of proliferating cells or the migration from SGZ in the non-ischemic condition. Our data showed that, specifically after ischemia, Ezh2 knockdown shifted the balance between self-renewal and differentiation toward differentiation in adult dentate gyrus.
  • Somsak Prasongtanakij, Sarinthip Preedagasamzin, Bunyada Jittorntrum, Usanarat Anurathapan, Teeraya Puavilai, Pimjai Niparuck, Pichika Chantrathammachart, Thanakrit Piyajaroenkij, Kitipong Uaesoontrachoon, Ryosuke Uchibori, Keiya Ozawa, Ken Ohmine, Suradej Hongeng
    European Journal of Haematology 2023年5月24日  査読有り
    Abstract Objectives Multiple myeloma (MM) accounts for 10% of hematologic malignancies. However, most of the patients suffered from relapsed/refractory disease. We would like to expand CAR T cell therapy to treat MM using our current platform. Methods BCMA CAR T lymphocytes were generated for volunteers or MM patients. The transduction efficiency was detected by the ddPCR technique. Immunophenotyping and exhaustion markers were monitored by flow cytometry. The efficacy of BCMA CAR T cells was tested using coculturing with BCMA CAR or mock, and the positive and negative targets, K562/hBCMA‐ECTM and K562, respectively. Results BCMA CAR T cells were generated from consented volunteers or MM patients and could be detected CAR BCMA expression at a mean of 4.07 ± 1.95 or 4.65 ± 1.21 copies/cell, respectively. Those modified T cells were primarily effector memory T cells. Our BCMA CAR T cells could explicitly eradicate the K562/hBCMA‐ECTM cell line while the K562 cell line survived. Interestingly, the BCMA CAR, mock T cells, and peripheral blood mononuclear cells from MM patients expressed similar levels of the exhaustion makers, TIM‐3, LAG‐3, and PD1. Conclusions Our BCMA CAR T cells, mainly effector/effector memory, could eliminate BCMA‐expressing cells in vitro and had similar levels of exhaustion markers among different populations.
  • 内堀 亮介, 大嶺 謙
    BIO Clinica 37(6) 525-529 2022年6月  
    医薬品の開発において、1970年代~80年代に次々と同定されたがん関連遺伝子を標的とするimatinibなどの低分子化合物が、薬剤としてがんに特異的に作用することが明確に示された。1980年代後半には、rituximabやtrastuzumabをはじめとする抗体医薬の開発が脚光を浴びてきた。近年、ヒト免疫メカニズムの理解の更なる深まりと遺伝子工学技術の発展により、患者自身のT細胞の働きを遺伝子操作で高めたCAR-T細胞を用いる治療法が、がんに対する革新的な細胞免疫療法として大きく期待されている。(著者抄録)
  • Ken Ohmine, Ryosuke Uchibori
    115(6) 799-810 2022年5月18日  査読有り
  • 内堀 亮介, 大嶺 謙
    血液内科 84(5) 730-736 2022年5月  
  • Yoshihide Sehara, Yuka Hayashi, Kenji Ohba, Ryosuke Uchibori, Masashi Urabe, Ayumu Inutsuka, Kuniko Shimazaki, Kensuke Kawai, Hiroaki Mizukami
    Human Gene Therapy 2021年8月5日  査読有り
  • 内堀 亮介, 大嶺 謙
    血液内科 82(1) 125-131 2021年1月  
  • 内堀 亮介
    臨床薬理 51(Suppl.) S195-S195 2020年10月  
  • 内堀 亮介, 小澤 敬也
    がん免疫療法 4(1) 4-8 2020年5月  
  • Taisuke Kondo, Makoto Ando, Nao Nagai, Wataru Tomisato, Tanakorn Srirat, Binbin Liu, Setsuko Mise-Omata, Mari Ikeda, Shunsuke Chikuma, Hiroshi Nishimasu, Osamu Nureki, Mitsuyo Ohmura, Noriyo Hayakawa, Takako Hishiki, Ryosuke Uchibori, Keiya Ozawa, Akihiko Yoshimura
    Cancer Research 80(3) 471-483 2020年2月1日  査読有り
    © 2019 American Association for Cancer Research. Recent studies have shown that stem cell memory T (TSCM) cell-like properties are important for successful adoptive immunotherapy by the chimeric antigen receptor–engineered-T (CART) cells. We previously reported that both human and murineactivated T cells are converted into stem cell memory-like T (iTSCM) cells by coculture with stromal OP9 cells expressing the NOTCH ligand. However, the mechanism of NOTCH-mediated iTSCM reprogramming remains to be elucidated. Here, we report that the NOTCH/OP9 system efficiently converted conventional human CAR-T cells into TSCM-like CAR-T, “CAR-iTSCM” cells, and that mitochondrial metabolic reprogramming played a key role in this conversion. NOTCH signaling promoted mitochondrial biogenesis and fatty acid synthesis during iTSCM formation, which are essential for the properties of iTSCM cells. Forkhead box M1 (FOXM1) was identified as a downstream target of NOTCH, which was responsible for these metabolic changes and the subsequent iTSCM differentiation. Like NOTCH-induced CAR-iTSCM cells, FOXM1-induced CAR-iTSCM cells possessed superior antitumor potential compared with conventional CAR-T cells. We propose that NOTCH-or FOXM1-driven CAR-iTSCM formation is an effective strategy for improving cancer immunotherapy.
  • 伊藤 祥子, 大嶺 謙, 内堀 亮介
    医学と薬学 77(2) 235-245 2020年1月  
  • 内堀 亮介, 大嶺 謙
    血液内科 79(4) 529-534 2019年10月  
  • Uchibori R, Teruya T, Ido H, Ohmine K, Sehara Y, Urabe M, Mizukami H, Mineno J, Ozawa K
    Molecular therapy oncolytics 12 16-25 2019年3月  査読有り
  • Yoshiba T, Saga Y, Urabe M, Uchibori R, Matsubara S, Fujiwara H, Mizukami H
    Oncology letters 17(2) 2197-2206 2019年2月  査読有り
  • 内堀 亮介
    Pharma Medica 36(11) 15-18 2018年11月  
  • Kurosaki F, Uchibori R, Sehara Y, Saga Y, Urabe M, Mizukami H, Hagiwara K, Kume A
    Human gene therapy 29(11) 1242-1251 2018年11月  査読有り
  • Sehara Y, Shimazaki K, Kurosaki F, Kaneko N, Uchibori R, Urabe M, Kawai K, Mizukami H
    Neuroscience letters 682 27-31 2018年8月  査読有り
  • Sato N, Saga Y, Uchibori R, Tsukahara T, Urabe M, Kume A, Fujiwara H, Suzuki M, Ozawa K, Mizukami H
    International journal of oncology 52(3) 687-696 2018年3月  査読有り
  • 内堀 亮介, 小澤 敬也
    医薬ジャーナル 54(1) 79-85 2018年1月  
    近年,B細胞性造血器腫瘍に対する治療で,CD19特異的キメラ抗原受容体(chimeric antigen receptor:CAR)を用いた養子免疫遺伝子療法が目覚ましい成果を上げている。がんに対する免疫療法の成否は,がん細胞特異的に免疫反応を示すT細胞をいかにして活性化するかにかかっている。CARを用いた遺伝子修飾によって,がん特異的な免疫応答を示すT細胞の作製が可能となった。本稿では養子免疫療法,特にキメラ抗原受容体を用いた細胞療法の現状と課題について概説する。(著者抄録)
  • Naoki Hosen, Yukiko Matsunaga, Kana Hasegawa, Hiroshi Matsuno, Yuki Nakamura, Mio Makita, Kouki Watanabe, Mikako Yoshida, Kei Satoh, Soyoko Morimoto, Fumihiro Fujiki, Hiroko Nakajima, Jun Nakata, Sumiyuki Nishida, Akihiro Tsuboi, Yoshihiro Oka, Masahiro Manabe, Hiroyoshi Ichihara, Yasutaka Aoyama, Atsuko Mugitani, Takafumi Nakao, Masayuki Hino, Ryosuke Uchibori, Keiya Ozawa, Yoshihiro Baba, Seitaro Terakura, Naoki Wada, Eiichi Morii, Junichi Nishimura, Kiyoshi Takeda, Yusuke Oji, Haruo Sugiyama, Junichi Takagi, Atsushi Kumanogoh
    Nature medicine 23(12) 1436-1443 2017年12月  査読有り
    Cancer-specific cell-surface antigens are ideal targets for monoclonal antibody (mAb)-based immunotherapy but are likely to have previously been identified in transcriptome or proteome analyses. Here, we show that the active conformer of an integrin can serve as a specific therapeutic target for multiple myeloma (MM). We screened >10,000 anti-MM mAb clones and identified MMG49 as an MM-specific mAb specifically recognizing a subset of integrin β7 molecules. The MMG49 epitope, in the N-terminal region of the β7 chain, is predicted to be inaccessible in the resting integrin conformer but exposed in the active conformation. Elevated expression and constitutive activation of integrin β7 conferred high MMG49 reactivity on MM cells, whereas MMG49 binding was scarcely detectable in other cell types including normal integrin β7+ lymphocytes. T cells transduced with MMG49-derived chimeric antigen receptor (CAR) exerted anti-MM effects without damaging normal hematopoietic cells. Thus, MMG49 CAR T cell therapy is promising for MM, and a receptor protein with a rare but physiologically relevant conformation can serve as a cancer immunotherapy target.
  • 内堀 亮介
    血液フロンティア 27(7) 969-977 2017年6月  
    多発性骨髄腫は白血病や悪性リンパ腫とともに3大血液がんの1つであり,社会の高齢化とともに患者数は増えていくとみられている。かつては治療法が限られ,さまざまな合併症により予後は好ましくなかったが,近年,プロテアソーム阻害薬であるボルテゾミブや,免疫調節薬サリドマイドやレナリドミドなどの登場により,治療が大きく進歩している。しかし,そうした治療法の進歩にもかかわらず,再発・難治症例に対する治療は依然として困難であり,免疫療法など新たな治療法の開発が望まれている。本稿では骨髄腫に対する免疫療法の現状と,キメラ抗原受容体を用いた治療法の今後の展開について展望したい。(著者抄録)
  • F. Kurosaki, R. Uchibori, N. Mato, Y. Sehara, Y. Saga, M. Urabe, H. Mizukami, Y. Sugiyama, A. Kume
    GENE THERAPY 24(5) 290-297 2017年5月  査読有り
    An efficient adeno-associated virus (AAV) vector was constructed for the treatment of respiratory diseases. AAV serotypes, promoters and routes of administration potentially influencing the efficiency of gene transfer to airway cells were examined in the present study. Among the nine AAV serotypes (AAV1-9) screened in vitro and four serotypes (AAV1, 2, 6, 9) evaluated in vivo, AAV6 showed the strongest transgene expression. As for promoters, the cytomegalovirus (CMV) early enhancer/chicken beta-actin (CAG) promoter resulted in more robust transduction than the CMV promoter. Regarding delivery routes, intratracheal administration resulted in strong transgene expression in the lung, whereas the intravenous and intranasal administration routes yielded negligible expression. The combination of the AAV6 capsid and CAG promoter resulted in sustained expression, and the intratracheally administered AAV6-CAG vector transduced bronchial cells and pericytes in the lung. These results suggest that AAV6-CAG vectors are more promising than the previously preferred AAV2 vectors for airway transduction, particularly when administered into the trachea. The present study offers an optimized strategy for AAV-mediated gene therapy for lung diseases, such as cystic fibrosis and pulmonary fibrosis.
  • 内堀 亮介
    血液内科 74(3) 392-397 2017年3月  
  • 内堀 亮介
    医学のあゆみ 257(3) 239-243 2016年4月  
    近年、B細胞性造血器腫瘍に対する治療で、CD19特異的キメラ抗原受容体(CAR)を用いた養子免疫遺伝子療法がめざましい成果をあげている。がんに対する免疫療法の成否は、がん細胞特異的に免疫反応を示すT細胞をいかにして活性化するかにかかっている。従来の免疫療法もT細胞を活性化するという点では同じであるが、活性化されたT細胞がかならずしもがん特異的に免疫反応を示すわけではなかったため、十分な臨床効果が得られなかった。一方、CARを用いて遺伝子修飾することで、がんに特異性の高い免疫応答を示すT細胞の作製が可能となった。本稿では、養子免疫遺伝子療法、とくにキメラ抗原受容体を用いた遺伝子治療の現状と課題について概説する。(著者抄録)
  • 内堀 亮介
    最新医学 70(12) 2559-2565 2015年12月  
    近年,CD19特異的キメラ抗原受容体を用いたB細胞性造血器腫瘍に対する養子免疫遺伝子療法の臨床試験結果が相次いで報告されている.従来の治療法に抵抗性の難治性造血器腫瘍に対しても優れた治療効果が認められる一方で,重篤な有害事象も報告されており,十分な安全管理や対策を講じたうえでの治療が望まれる.本稿では,CARを用いた治療戦略の概念と,これまでに報告された臨床試験について概説する.(著者抄録)
  • 内堀 亮介
    血液フロンティア 25(3) 368-372 2015年2月  
    HIVはヒトの免疫細胞に感染して免疫不全を引き起こす。現行の治療では抗ウイルス剤でHIVの増殖を抑える方法が採られているが、HIVは潜伏期間を経て再び活性化するため生涯に渡る投薬治療が必要になり、完全治癒も望めない。しかし、近年目覚ましい進歩を遂げている、いわゆるゲノム編集技術を応用することで、HIVを体内から完全に排除して感染症をコントロールする新しい治療法が開発されている。(著者抄録)
  • T. Tsukahara, N. Iwase, K. Kawakami, M. Iwasaki, C. Yamamoto, K. Ohmine, R. Uchibori, T. Teruya, H. Ido, Y. Saga, M. Urabe, H. Mizukami, A. Kume, M. Nakamura, R. Brentjens, K. Ozawa
    GENE THERAPY 22(2) 209-215 2015年2月  査読有り
    Engineered T-cell therapy using a CD19-specific chimeric antigen receptor ( CD19-CAR) is a promising strategy for the treatment of advanced B-cell malignancies. Gene transfer of CARs to T-cells has widely relied on retroviral vectors, but transposon-based gene transfer has recently emerged as a suitable nonviral method to mediate stable transgene expression. The advantages of transposon vectors compared with viral vectors include their simplicity and cost-effectiveness. We used the Tol2 transposon system to stably transfer CD19-CAR into human T-cells. Normal human peripheral blood lymphocytes were co-nucleofected with the Tol2 transposon donor plasmid carrying CD19-CAR and the transposase expression plasmid and were selectively propagated on NIH3T3 cells expressing human CD19. Expanded CD3+ T-cells with stable and high-level transgene expression (similar to 95%) produced interferon. gamma upon stimulation with CD19 and specifically lysed Raji cells, a CD19(+) human B-cell lymphoma cell line. Adoptive transfer of these T-cells suppressed tumor progression in Raji tumor-bearing Rag2(-/-)gamma c(-/-) immunodeficient mice compared with control mice. These results demonstrate that the Tol2 transposon system could be used to express CD19-CAR in genetically engineered T-cells for the treatment of refractory B-cell malignancies.
  • Ryosuke Uchibori, Tomonori Tsukahara, Ken Ohmine, Keiya Ozawa
    INTERNATIONAL JOURNAL OF HEMATOLOGY 99(4) 377-382 2014年4月  査読有り
    Cellular and gene therapies represent promising treatment strategies at the frontier of medicine. Hematopoietic stem cells, lymphocytes, and mesenchymal stem cells (MSCs) can all serve as sources of cells for use in such therapies. Strategies for gene therapy are often based on those of cell therapy, and it is anticipated that some examples will be put to practical use in the near future. Given their ability to support hematopoiesis, MSCs may be useful for the enhancement of stem cell engraftment, and the acceleration of hematopoietic reconstitution. Furthermore, MSCs may advance the treatment of severe graft-versus-host disease, based on their immunosuppressive ability. This application is also based on the homing behavior of MSCs to sites of injury and inflammation. Interestingly, MSCs possess tumor-homing ability, opening up the possibility of applications in the targeted delivery of anti-cancer genes to tumors. Many reports have indicated that MSCs can be utilized to target tumors and to deliver anti-cancer molecules locally, as tumors are recognized as non-healing wounds with inflammatory tissue. Here, we review both the potential of MSCs as cellular vehicles for targeted cancer therapy and the molecular mechanisms underlying MSC accumulation at tumor sites.
  • Takayuki Uehara, Takeharu Kanazawa, Hiroaki Mizukami, Ryosuke Uchibori, Tomonori Tsukahara, Masashi Urabe, Akihiro Kume, Kiyoshi Misawa, Thomas E. Carey, Mikio Suzuki, Keiichi Ichimura, Keiya Ozawa
    Cancer Science 105(1) 72-80 2014年1月  査読有り
    Galanin and its receptors, GALR1 and GALR2, are known tumor suppressors and potential therapeutic targets in head and neck squamous cell carcinoma (HNSCC). Previously, we demonstrated that, in GALR1-expressing HNSCC cells, the addition of galanin suppressed tumor proliferation via upregulation of ERK1/2 and cyclin-dependent kinase inhibitors, whereas, in GALR2-expressing cells, the addition of galanin not only suppressed proliferation, but also induced apoptosis. In this study, we first transduced HEp-2 and KB cell lines using a recombinant adeno-associated virus (rAAV)-green fluorescent protein (GFP) vector and confirmed a high GFP expression rate (&gt 90%) in both cell lines at the standard vector dose. Next, we demonstrated that GALR2 expression in the presence of galanin suppressed cell viability to 40-60% after 72 h in both cell lines. Additionally, the annexin V-positive rate and sub-G0/G1 phase population were significantly elevated in HEp-2 cells (mock vs GALR2: 12.3 vs 25.0% (P &lt 0.01) and 9.1 vs 32.0% (P &lt 0.05), respectively) after 48 h. These changes were also observed in KB cells, although to a lesser extent. Furthermore, in HEp-2 cells, GALR2-mediated apoptosis was caspase-independent, involving downregulation of ERK1/2, followed by induction of the pro-apoptotic Bcl-2 protein, Bim. These results illustrate that transient GALR2 expression in the presence of galanin induces apoptosis via diverse pathways and serves as a platform for suicide gene therapy against HNSCC. © 2013 The Authors.
  • Takayuki Uehara, Takeharu Kanazawa, Hiroaki Mizukami, Ryosuke Uchibori, Tomonori Tsukahara, Masashi Urabe, Akihiro Kume, Kiyoshi Misawa, Thomas E. Carey, Mikio Suzuki, Keiichi Ichimura, Keiya Ozawa
    CANCER SCIENCE 105(1) 72-80 2014年1月  査読有り
    Galanin and its receptors, GALR1 and GALR2, are known tumor suppressors and potential therapeutic targets in head and neck squamous cell carcinoma (HNSCC). Previously, we demonstrated that, in GALR1-expressing HNSCC cells, the addition of galanin suppressed tumor proliferation via upregulation of ERK1/2 and cyclin-dependent kinase inhibitors, whereas, in GALR2-expressing cells, the addition of galanin not only suppressed proliferation, but also induced apoptosis. In this study, we first transduced HEp-2 and KB cell lines using a recombinant adeno-associated virus (rAAV)-green fluorescent protein (GFP) vector and confirmed a high GFP expression rate (>90%) in both cell lines at the standard vector dose. Next, we demonstrated that GALR2 expression in the presence of galanin suppressed cell viability to 40-60% after 72h in both cell lines. Additionally, the annexin V-positive rate and sub-G0/G1 phase population were significantly elevated in HEp-2 cells (mock vs GALR2: 12.3 vs 25.0% (P < 0.01) and 9.1 vs 32.0% (P < 0.05), respectively) after 48h. These changes were also observed in KB cells, although to a lesser extent. Furthermore, in HEp-2 cells, GALR2-mediated apoptosis was caspase-independent, involving downregulation of ERK1/2, followed by induction of the pro-apoptotic Bcl-2 protein, Bim. These results illustrate that transient GALR2 expression in the presence of galanin induces apoptosis via diverse pathways and serves as a platform for suicide gene therapy against HNSCC.
  • Tomonori Tsukahara, Ken Ohmine, Chihiro Yamamoto, Ryosuke Uchibori, Hiroyuki Ido, Takeshi Teruya, Masashi Urabe, Hiroaki Mizukami, Akihiro Kume, Masataka Nakamura, Junichi Mineno, Kazutoh Takesako, Isabelle Riviere, Michel Sadelain, Renier Brentjens, Keiya Ozawa
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 438(1) 84-89 2013年8月  査読有り
    Adoptive T-cell therapy with CD19-specific chimeric antigen receptors (CARs) is promising for treatment of advanced B-cell malignancies. Tumor targeting of CAR-modified T-cells is likely to contribute therapeutic potency; therefore we examined the relationship between the ability of CD19-specific CAR (CD19-CAR)-transduced T-cells to accumulate at CD19(+) tumor lesions, and their ability to provide antitumor effects in xenograft mouse models. Normal human peripheral blood lymphocytes, activated with immobilized RetroNectin and anti-CD3 antibodies, were transduced with retroviral vectors that encode CD19-CAR. Expanded CD19-CAR T-cells with a high transgene expression level of about 75% produced IL-2 and IFN-gamma in response to CD19, and lysed both Raji and Daudi CD19(+) human B-cell lymphoma cell lines. Furthermore, these cells efficiently accumulated at Raji tumor lesions where they suppressed tumor progression and prolonged survival in tumor-bearing Rag2(-/-)gamma c(-/-) immunodeficient mice compared to control cohorts. These results show that the ability of CD19-CAR T-cells to home in on tumor lesions is pivotal for their anti-tumor effects in our xenograft models, and therefore may enhance the efficacy of adoptive T-cell therapy for refractory B-cell lymphoma. (C) 2013 Elsevier Inc. All rights reserved.
  • Uchibori R, Tsukahara T, Mizuguchi H, Saga Y, Urabe M, Mizukami H, Kume A, Ozawa K
    Cancer research 73(1) 364-372 2013年1月  査読有り
  • 塚原 智典, 大嶺 謙, 内堀 亮介, 卜部 匡司, 水上 浩明, 久米 晃啓, 小澤 敬也
    自治医科大学紀要 34 160-161 2012年3月  
  • 内堀 亮介
    自治医科大学紀要 33 199-199 2011年3月  
  • Uchibori R, Ozawa K
    [Rinsho ketsueki] The Japanese journal of clinical hematology 51(11) 1641-1646 2010年11月  査読有り
  • 内堀 亮介, 小澤 敬也
    臨床血液 51(11) 1641-1646 2010年11月  
  • Minoru Kobayashi, Takashi Murakami, Ryosuke Uchibori, Nicole A. L. Chun, Eiji Kobayashi, Tatsuo Morita, Keiya Ozawa
    JOURNAL OF UROLOGY 183(5) 2029-2035 2010年5月  査読有り
    Purpose: Since renal cell carcinoma is considered an immunogenic tumor, testing therapeutic strategies has been impeded by the lack of relevant tumor models in immunocompetent animals. Recent advances in bioluminescence imaging permit sensitive in vivo detection and quantification of cells emitting light. Thus, we established bioluminescent rat renal cell carcinoma cell lines for immunocompetent rats. Materials and Methods: The rat renal cell carcinoma cell line ACI-RCC stemming from chemically induced renal cell carcinoma in syngeneic ACI rats was stably transfected with a recombinant retroviral vector encoding luciferase genes derived from fireflies (ACI-RCC-ffLuc) or click beetles (ACI-RCC-cbLuc). Cell line growth patterns were characterized by bioluminescence imaging. Results: Linear correlations noted observed between cell number and photon counts in each cell type. ACI-RCC-cbLuc emitted light about 500-fold higher than ACI-RCC-ffLuc. When transplanted subcutaneously, only ACI-RCC-ffLuc grew, possibly because of less antigenicity. ACI-RCC-ffLuc photon emission correlated significantly with subcutaneous tumor size. Orthotopic tumor growth and subsequent metastatic spread were monitored with time by increased photon intensity on bioluminescence imaging. Based on ACI-RCC-cbLuc bioluminescent intensity the in vitro screening test allowed the identification of several anticancer agents, including molecules related to human renal cell carcinoma progression. Conclusions: The new in vivo rat renal cell carcinoma model with luciferase labeled tumor cells allowed us to monitor tumor growth noninvasively and semiquantitatively by bioluminescence imaging. This model system coupled with in vitro screening permits precise evaluation of tumor behavior in intact animals and determination of the therapeutic efficacy of anticancer agents for renal cell carcinoma.
  • Okada T, Nonaka-Sarukawa M, Uchibori R, Kinoshita K, Hayashita-Kinoh H, Nitahara-Kasahara Y, Takeda S, Ozawa K
    Human gene therapy 20 1013-1021 2009年9月  査読有り
  • Ryosuke Uchibori, Takashi Okada, Takayuki Ito, Masashi Urabe, Hiroaki Mizukami, Akihiro Kume, Keiya Ozawa
    JOURNAL OF GENE MEDICINE 11(5) 373-381 2009年5月  査読有り
    Background Mesenchymal stein cells (MSCs) are a promising vehicle for targeted cancer gene therapy because of their potential of tumor tropism. For efficient therapeutic application, we developed retroviral vector-producing MSCs that enhance tumor transduction via progeny vector production. Methods Rat bone marrow-derived MSCs were nucleofected with the proviral plasmids (vesicular stomatitis virus-G protein-pseudotyped retroviral vector components) (VP-MSCs) or pLTR plasmid alone (non-VP-MSCs). The luciferase-based in vivo imaging system was used to assess gene expression periodically. To evaluate the anticancer effects, we administered MSCs expressing herpes simplex virus-thymidine kinase (HSV-tk) into the left ventricular cavity of nude mice engrafted with 9L glioma cells subcutaneously. Results In vivo imaging revealed that administration of luciferase-expressing non-VP-MSCs enhanced the bioluminescence signal at the inoculation sites of 9L cells, whereas no accumulation was observed in juice at the site of the control Rat-1 fibroblasts. Compared to non-VP-MSCs, the administration of VP-MSCs resulted in significant augmentation of the signal with an increase in transgene copy number. Immunohistochemical analysis showed marked luciferase expression at the tumor periphery in mice injected with VP-MSCs, whereas little expression was detected in those in injected. with non-VP-MSCs. Under the continuous infusion of ganciclovir systemic, administration of VP-MSCs expressing HSV-tk Suppressed tumor growth more effectively than non-VP-MSC administration, whereas no anticancer effect was observed Without ganciclovir treatment. Furthermore, VP-MSC administration caused no transgene transduction in the normal tissues and organs. Conclusions VP-MSCs accumulated at the site of tumors after intravascular injection in tumor-bearing mice, followed by in situ gene transfer to tumors without transduction of normal organs. When applied to the HSV-tk/ganciclovir suicide gene therapy, more efficient tumor growth Suppression was observed using VP-MSCs compared to non-VP-MSCs. This VP-MSC-based system has great potential for improved cancer gene therapy. Copyright (C) 2009 John Wiley & Sons, Ltd.
  • Keiya Ozawa, Kazuya Sato, Iekuni Oh, Katsutoshi Ozaki, Ryosuke Uchibori, Yoko Obara, Yuji Kikuchi, Takayuki Ito, Takashi Okada, Masashi Urabe, Hiroaki Mizukami, Akihiro Kume
    JOURNAL OF AUTOIMMUNITY 30(3) 121-127 2008年5月  査読有り
    Mesenchymal stem cells (MSCs) are considered to be a promising platform for cell and gene therapy for a variety of diseases. First, in the field of hematopoietic stem cell transplantation, there are two applications of MSCs: 1) the improvement of stem cell engrafting and the acceleration of hematopoietic reconstitution based on the hematopoiesis-supporting ability; and 2) the treatment of severe graft-versus-host disease (GVHD) based on the immunomodulatory ability. Regarding the immunosuppressive ability, we found that nitric oxide (NO) is involved in the MSC-mediated suppression of T cell proliferation. Second, tumor-bearing nude mice were injected with luciferase-expressing MSCs. An in vivo imaging analysis showed the significant accumulation of the MSCs at the site of tumors. The findings suggest that MSCs can be utilized to target metastatic tumors and to deliver anti-cancer molecules locally. As the third application, MSCs may be utilized as a cellular vehicle for protein-supplement gene therapy. When long-term transgene expression is needed, a therapeutic gene should be introduced with a minimal risk of insertional mutagenesis. To this end, site-specific integration into the AAVS 1 locus on the chromosome 19 (19q13.4) by using the integration machinery of adeno-associated virus (AAV) would be particularly valuable. There will be wide-ranging applications of MSCs to frontier medical treatments in the near future. (C) 2008 Elsevier Ltd. All rights reserved.
  • Takayuki Ito, Takashi Okada, Jun Mimuro, Hiroshi Miyashita, Ryosuke Uchibori, Masashi Urabe, Hiroaki Mizukami, Akihiro Kume, Masafumi Takahashi, Uichi Ikeda, Yoichi Sakata, Kazuyuki Shimada, Keiya Ozawa
    HYPERTENSION 50(3) 531-536 2007年9月  査読有り
    Prostacyclin synthase (PGIS) is the final committed enzyme in the metabolic pathway of prostacyclin production. The therapeutic option of intravenous prostacyclin infusion in patients with pulmonary arterial hypertension is limited by the short half-life of the drug and life-threatening catheter-related complications. To develop a better delivery system for prostacyclin, we examined the feasibility of intramuscular injection of an adenoassociated virus (AAV) vector expressing PGIS for preventing monocrotaline-induced pulmonary arterial hypertension in rats. We developed an AAV serotype 1-based vector carrying a human PGIS gene (AAV-PGIS). AAV-PGIS or the control AAV vector expressing enhanced green fluorescent protein was injected into the anterior tibial muscles of 3-week-old male Wistar rats; this was followed by the monocrotaline administration at 7 weeks. Eight weeks after injecting the vector, the plasma levels of 6-keto-prostaglandin F-1 alpha increased in a vector dose-dependent manner. At this time point, the PGIS transduction (1x10(10) genome copies per body) significantly decreased mean pulmonary arterial pressure (33.9 +/- 2.4 versus 46.1 +/- 3.0 mm Hg; P < 0.05), pulmonary vascular resistance (0.26 +/- 0.03 versus 0.41 +/- 0.03 mm Hg . mL(-1) . min(-1) . kg(-1); P < 0.05), and medial thickness of the peripheral pulmonary artery (14.6 +/- 1.5% versus 23.5 +/- 0.5%; P < 0.01) as compared with the controls. Furthermore, the PGIS-transduced rats demonstrated significantly improved survival rates as compared with the controls (100% versus 50%; P < 0.05) at 8 weeks postmonocrotaline administration. An intramuscular injection of AAV-PGIS prevents monocrotaline-pulmonary arterial hypertension in rats and provides a new therapeutic alternative for preventing pulmonary arterial hypertension in humans.
  • Takayuki Ito, Takashi Okada, Hiroshi Miyashita, Tatsuya Nomoto, Mutsuko Nonaka-Sarukawa, Ryosuke Uchibori, Yoshikazu Maeda, Masashi Urabe, Hiroaki Mizukami, Akihiro Kume, Masafumi Takahashi, Uichi Ikeda, Kazuyuki Shimada, Keiya Ozawa
    CIRCULATION RESEARCH 101(7) 734-741 2007年9月  査読有り
    Pulmonary arterial hypertension (PAH) is a fatal disease associated with inflammation and pathological remodeling of the pulmonary artery (PA). Interleukin (IL)-10 is a pleiotropic antiinflammatory cytokine with vasculoprotective properties. Here, we report the preventive effects of IL-10 on monocrotaline-induced PAH. Three-week-old Wistar rats were intramuscularly injected with an adeno-associated virus serotype 1 vector expressing IL-10, followed by monocrotaline injection at 7 weeks old. IL-10 transduction significantly improved survival rates of the PAH rats 8 weeks after monocrotaline administration compared with control gene transduction (75% versus 0%, P < 0.01). IL-10 also significantly reduced mean PA pressure (22.8 +/- 1.5 versus 29.7 +/- 2.8 mm Hg, P < 0.05), a weight ratio of right ventricle to left ventricle plus septum (0.35 +/- 0.04 versus 0.42 +/- 0.05, P < 0.05), and percent medial thickness of the PA (12.9 +/- 0.3% versus 21.4 +/- 0.4%, P < 0.01) compared with controls. IL-10 significantly reduced macrophage infiltration and vascular cell proliferation in the remodeled PA in vivo. It also significantly decreased the lung levels of transforming growth factor-beta(1) and IL-6, which are indicative of PA remodeling. In addition, IL-10 increased the lung level of heme oxygenase-1, which strongly prevents PA remodeling. In vitro analysis revealed that IL-10 significantly inhibited excessive proliferation of cultured human PA smooth muscle cells treated with transforming growth factor-beta(1) or the heme oxygenase inhibitor tin protoporphyrin IX. Thus, IL-10 prevented the development of monocrotaline-induced PAH, and these results provide new insights into the molecular mechanisms of human PAH.
  • Kiwamu Akagi, Ryosuke Uchibori, Kensei Yamaguchi, Keiko Kurosawa, Yoichiro Tanaka, Tomoko Kozu
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 352(3) 728-732 2007年1月  査読有り
    Activating mutations of RAS are frequently observed in subsets of human cancers, indicating that RAS activation is involved in tumorigenesis. Here, we identified and characterized a novel G to T transversion mutation of the K-ras gene at the third position of codon 19 (TTG) which substituted phenylalanine for leucine in 3 primary colon carcinomas. Biological and biochemical activity was examined using transformed NIH3T3 cells expressing mutant or wild-type K-ras. Transformants harboring the K-ras mutation at codon 19 showed proliferative capacity under serum-starved conditions, less contact inhibition, anchorage-independent growth, tumorigenicity in nude mice and elevation of active Ras-GTP levels. These results indicated that this novel mutation possesses high oncogenic activity. (c) 2006 Elsevier Inc. All rights reserved.
  • T Okada, R Uchibori, M Iwata-Okada, M Takahashi, T Nomoto, M Nonaka-Sarukawa, T Ito, Y Liu, H Mizukami, A Kume, E Kobayashi, K Ozawa
    MOLECULAR THERAPY 13(4) 738-746 2006年4月  査読有り
    The transduction of cancer cells using recombinant adeno-associated virus (rAAV) occurs with low efficiency, which limits its utility in cancer gene therapy. We have previously sought to enhance rAAV-mediated transduction of cancer cells by applying DNA-damaging stresses. In this study, we examined the effects of the histone deacetylase inhibitor FR901228 on tumor transduction mediated by rAAV types 2 and 5. FR901228 treatment significantly improved the expression of the transgene in four cancer cell lines. The cell surface levels of alpha v integrin, FGF-R1, and PDGF-R were modestly enhanced by the presence of FR901228. These results suggest that the superior transduction induced by the HDAC inhibitor was due to an enhancement of transgene expression rather than increased viral entry. Furthermore, we characterized the association of the acetylated histone H3 in the episomal AAV vector genome by using the chromatin immunoprecipitation assay. The results suggest that the superior transduction may be related to the proposed histone-associated chromatin form of the rAAV concatemer in transduced cells. In the analysis with subcutaneous tumor models, strong enhancement of the transgene expression as well as therapeutic effect was confirmed in vivo. The use of this HDAC inhibitor may enhance the utility of rAAV-mediated transduction strategies for cancer gene therapy.
  • T Okada, T Nomoto, T Yoshioka, M Nonaka-Sarukawa, T Ito, T Ogura, M Iwata-Okada, R Uchibori, K Shimazaki, H Mizukami, A Kume, K Ozawa
    HUMAN GENE THERAPY 16(10) 1212-1218 2005年10月  査読有り
    Adenovirus and adeno-associated virus (AAV) vectors are increasingly used for gene transduction experiments. However, to produce a sufficient amount of these vectors for in vivo experiments requires large-capacity tissue culture facilities, which may not be practical in limited laboratory space. We describe here a large-scale method to produce adenovirus and AAV vectors with an active gassing system that uses large culture vessels to process labor-and cost-effective infection or transfection in a closed system. Development of this system was based on the infection or transfection of 293 cells on a large scale, using a large culture vessel with a surface area of 6320 cm(2). A minipump was connected to the gas inlet of the large vessel, which was placed inside the incubator, so that the incubator atmosphere was circulated through the vessel. When active gassing was employed, the productivity of the adenovirus and AAV vectors significantly increased. This vector production system was achieved by improved CO2 and air exchange and maintenance of pH in the culture medium. Viral production with active gassing is particularly promising, as it can be used with existing incubators and the large culture vessel can readily be converted for use with the active gassing system.

MISC

 30

書籍等出版物

 1

共同研究・競争的資金等の研究課題

 5