研究者業績

久米 晃啓

クメ アキヒロ  (Akihiro Kume)

基本情報

所属
自治医科大学 附属病院臨床研究センター企画開発部 教授
学位
博士(医学)(東北大学)

J-GLOBAL ID
200901072254016208
researchmap会員ID
1000193248

論文

 124
  • Yuji Kashiwakura, Kazuhiro Endo, Atsushi Ugajin, Tomohiro Kikuchi, Shuji Hishikawa, Hitoyasu Nakamura, Yuko Katakai, Nemekhbayar Baatartsogt, Takafumi Hiramoto, Morisada Hayakawa, Nobuhiko Kamoshita, Shoji Yamazaki, Akihiro Kume, Harushi Mori, Naohiro Sata, Yoichi Sakata, Shin-ichi Muramatsu, Tsukasa Ohmori
    Molecular Therapy - Methods & Clinical Development 2023年8月  
  • 柏倉 裕志, 遠藤 和洋, 宇賀神 敦, 菊地 智博, 菱川 修司, 中村 仁康, 片貝 祐子, Nemekhbayar Baatartsogt, 平本 貴史, 早川 盛禎, 鴨下 信彦, 山崎 晶司, 久米 晃啓, 森 墾, 佐田 尚宏, 坂田 洋一, 村松 慎一, 大森 司
    日本血栓止血学会誌 34(2) 240-240 2023年5月  
  • Teruhide Yamaguchi, Eriko Uchida, Takashi Okada, Keiya Ozawa, Masafumi Onodera, Akihiro Kume, Takashi Shimada, Satoru Takahashi, Kenzaburo Tani, Yasutomo Nasu, Tomoji Mashimo, Hiroyuki Mizuguchi, Kohnosuke Mitani, Kazushige Maki
    Human gene therapy 31(19-20) 1043-1053 2020年10月  
    The development of genome-editing technology could lead to breakthrough gene therapy. Genome editing has made it possible to easily knock out or modify a target gene, while current gene therapy using a virus vector or plasmid hampering modification with respect to gene replacement therapies. Clinical development using these genome-editing tools is progressing rapidly. However, it is also becoming clear that there is a possibility of unintended gene sequence modification or deletion, or the insertion of undesired genes, or the selection of cells with abnormalities in the cancer suppressor gene p53; these unwanted actions are not possible with current gene therapy. The Science Board of the Pharmaceuticals and Medical Devices Agency of Japan has compiled a report on the expected aspects of such genome-editing technology and the risks associated with it. This article summarizes the history of that discussion and compares the key concepts with information provided by other regulatory authorities.
  • Miyata S, Tominaga K, Sakashita E, Urabe M, Onuki Y, Gomi A, Yamaguchi T, Mieno M, Mizukami H, Kume A, Ozawa K, Watanabe E, Kawai K, Endo H
    Scientific reports 9(1) 9787 2019年7月  査読有り
  • Fumio Kurosaki, Ryosuke Uchibori, Yoshihide Sehara, Yasushi Saga, Masashi Urabe, Hiroaki Mizukami, Koichi Hagiwara, Akihiro Kume
    Human gene therapy 29(11) 1242-1251 2018年11月  査読有り
    Idiopathic pulmonary fibrosis (IPF) is a fibroproliferative disorder with limited therapeutic options. An aberrant wound healing process in response to repetitive lung injury has been suggested for its pathogenesis, and a number of cytokines including transforming growth factor β1 play pivotal roles in the induction and progression of fibrosis. Thus, the regulation of these pro-inflammatory conditions may reduce the progression of IPF and ameliorate its symptoms in patients. Interleukin-10 (IL-10), a pleiotropic cytokine, exerts anti-inflammatory and anti-fibrotic effects in numerous biological settings. In the present study, we investigated the preventive effects of IL-10 on bleomycin-induced pulmonary fibrosis in mice with the continuous expression of this cytokine via an adeno-associated virus serotype 6 vector. Mice were administered the adeno-associated virus serotype 6 vector encoding mouse IL-10 by intratracheal injection, and osmotic minipumps containing bleomycin were subcutaneously implanted seven days later. Lung histology and the expression levels of pro-inflammatory cytokines and fibrogenic cytokines were then analyzed. In mice exhibiting persistent IL-10 expression on day 35, the number of infiltrated inflammatory cells and the development of fibrosis in lung tissues were significantly reduced. Increases in transforming growth factor β1 and decreases in IFN-γ were also suppressed in treated animals, with changes in these cytokines playing important roles in the pathogenesis of pulmonary fibrosis. Furthermore, IL-10 significantly improved survival in bleomycin-induced mice. Our results provide insights into the potential benefit of the anti-fibrotic effects of IL-10 as a novel therapeutic approach for IPF.

MISC

 173
  • M Kametaka, A Kum, T Okada, H Mizukami, Y Hanazono, K Ozawa
    CANCER SCIENCE 94(7) 639-643 2003年7月  
    Allogeneic bone marrow transplantation and donor lymphocyte infusion are powerful treatments for chemotherapy-resistant leukemia. Tumor eradication is attributed to a graft-versus-leukemia reaction by the donor-derived cytotoxic T lymphocytes (CTLs), but the same cell population may cause severe graft-versus-host disease. One strategy to suppress harmful CTL activity is to incorporate a suicide gene into the donor lymphocytes prior to infusion, and to destroy these cells when they aggressively attack nonmalignant host tissues. In this study, we investigated the feasibility of using a Fas-estrogen receptor fusion protein (MfasER) to control T cell-mediated cytotoxicity, based on our previous finding that the chimera transmits a Fas-mediated death signal through activation by estrogen binding. A murine CTL line CTLL-2 was transfected with a vector encoding MfasER, and the growth, viability and cytotoxic activity of the transfected cells (CTLL/MfasER) were analyzed. The expression of apoptosis-related proteins such as Fas ligand and perforin was also investigated. In the absence of estrogen, CTLL/MfasER showed similar growth to parental CTLL-2, and the killing activity was preserved. Addition of 10(-7) M estrogen induced a rapid apoptosis of CTLL/MfasER, and the cytotoxicity was severely impaired. A decrease of Fas ligand and perforin in the estrogen-treated CTLL/MfasER was seen in an immunoblot analysis. These functional and biochemical analyses showed that the estrogen-inducible apoptosis in MfasER-expressing CTLs rapidly terminated their target cell killing. The feasibility of using the MfasER-estrogen system to control graft-versus-host disease was demonstrated.
  • H Mizukami, J Mimuro, T Ogura, F Ono, E Kobayashi, S Muramatsu, S Madoiwa, T Matsushita, T Okada, Y Hanazono, A Kume, K Terao, K Ozawa, Y Sakata
    MOLECULAR THERAPY 7(5) S81-S81 2003年5月  
  • T Yoshioka, T Okada, Y Maeda, M Shimpo, T Nomoto, K Takeuchi, T Matsushita, H Mizukami, Y Hanazono, A Kume, U Ikeda, S Ishibashi, K Shimada, K Ozawa
    MOLECULAR THERAPY 7(5) S23-S23 2003年5月  
  • T Hara, A Kume, Y Hanazono, H Mizukami, T Okada, H Tsurumi, H Moriwaki, M Hasegawa, K Ozawa
    MOLECULAR THERAPY 7(5) S407-S407 2003年5月  
  • T Okada, T Nomoto, K Shimazaki, M Iwata-Okada, T Yoshioka, M Takahashi, R Ajalli, YH Liu, T Matsushita, H Mizukami, M Urabe, Y Hanazono, A Kume, K Ozawa
    MOLECULAR THERAPY 7(5) S47-S47 2003年5月  
  • Y Takei, H Mizukami, Y Saga, S Kohno, T Matsushita, T Okada, Y Hanazono, A Kume, M Suzuki, K Ozawa
    MOLECULAR THERAPY 7(5) S136-S136 2003年5月  
  • A Kume, M Koremoto, RF Xu, T Okada, H Mizukami, Y Hanazono, M Hasegawa, K Ozawa
    JOURNAL OF GENE MEDICINE 5(3) 175-181 2003年3月  
    Background Hematopoietic stem-cell-directed gene transfer has achieved limited success in transducing clinically relevant levels of target cells. The expansion of gene-modified cells is one way to circumvent the problem of inefficient transduction with current vectors. To this end, we have developed 1 selective amplifier genes' (SAGS) that encode chimeric proteins that are a fusion of granulocyte colony-stimulating factor receptor and the steroid-binding domain. Prototype SAGS conferred estrogen-responsive growth on murine hematopoietic progenitors. Methods We constructed a retroviral vector coexpressing an SAG for 4-hydroxytamoxifen (Tm)-specific proliferation and the enhanced green fluorescent protein (EGFP). Murine bone marrow cells were transduced with this vector and transplanted into myeloablated mice. Subsequently, recipients were challenged with Tm, and EGFP(+) cells were enumerated. Results The challenge induced a significant increase in EGFP(+) leukocytes (21 +/- 4% to 27 +/- 5%), while EGFP(+) cells decreased in untreated animals (21 +/- 5% to 10 +/- 3%). Three months later, bone marrow cells were transplanted from the unchallenged mice to secondary hosts. Again the administration of Tm resulted in an increase of EGFP(+) cells (16 +/- 4% to 35 +/- 3%), contrasting to a decrease in controls (22 +/- 4% to 12 +/- 4%), and the difference was significant for more than 3 months. A detailed study of lineage showed a preferential expansion of EGFP(+) cells in granulocytes and monocytes following Tm administration. Conclusions Long-term repopulating cells were transduced with the SAG, and the transduced granulocyte/monocyte precursors were most likely to be expandable in vivo upon Tm stimulation. Copyright (C) 2002 John Wiley Sons, Ltd.
  • T Kanazawa, H Mizukami, T Okada, Y Hanazono, A Kume, H Nishino, K Takeuchi, K Kitamura, K Ichimura, K Ozawa
    GENE THERAPY 10(1) 51-58 2003年1月  
    The application of adeno-associated virus (AAV) vectors to cancers is limited by their low transduction efficiency. Previously, we reported that gamma-ray enhanced the second-strand synthesis, leading to the improvement of the transgene expression, and cytocidal effect of the herpes simplex virus type-1 thymidine kinase (HSVtk) and ganciclovir (GCV) system. In this study, we extended this in vitro findings to in vivo. First, the laryngeal cancer cell line (HEp-2) and HeLa were treated with AAVtk/GCV, the number of surviving cells was reduced as the concentration of GCV increased. Furthermore, the 4 Gy irradiation enhanced the killing effects of AAVtk/GCV by four-fold on HeLa cells and 15-fold on HEp-2 cells. Following the in vitro experiments, we evaluated the transgene expression and the antitumor activity of the AA V vectors in combination with gamma-ray in nude mice inoculated with HEp-2 subcutaneously. The LacZ expression was observed in the xenografted tumors and significantly increased by gamma-ray. The AAVtk/GCV system suppressed the tumors growth, and gamma-ray augmented the antitumor activity by five-fold. These findings suggest that the combination of AAVtk/GCV system with radiotherapy is significantly effective in the treatment of cancers and may lead to reduction of the potential toxicity of both AAVtk/GCV and gamma-ray.
  • J Mimuro, J Kikuchi, T Tabata, Y Ueda, A Kume, Y Hanazono, H Mizukami, K Ogata, S Madoiwa, T Sugo, M Hasegawa, K Ozawa, Y Sakata
    BLOOD 100(11) 441A-441A 2002年11月  
  • T Yoshioka, Y Maeda, T Okada, M Shimpo, T Nomoto, K Takeuchi, H Mizukami, Y Hanazono, A Kume, S Ookawara, K Ozawa, U Ikeda
    CIRCULATION 106(19) 30-30 2002年11月  
  • T Hara, A Kume, Y Hanazono, HI Mizukami, T Okada, H Tsurumi, H Moriwaki, M Hasegawa, K Ozawa
    BLOOD 100(11) 117A-117A 2002年11月  
  • A Itoh, T Okada, H Mizuguchi, T Hayakawa, H Mizukami, M Takatoku, N Komatsu, A Kume, Y Hanazono, K Ozawa
    BLOOD 100(11) 654A-654A 2002年11月  
  • A Kume, Y Hanazono, H Mizukami, T Okada, K Sugamura, K Ozawa
    BLOOD 100(11) 440A-440A 2002年11月  
  • F Mimuro, K Ogata, H Mizukami, J Kikuchi, S Madoiwa, T Sugo, Y Hanazono, A Kume, A Yoshioka, K Ozawa, Y Sakata
    BLOOD 100(11) 90B-90B 2002年11月  
  • Y Hanazono, T Nagashima, M Takatoku, H Shibata, N Ageyama, T Asano, Y Ueda, C E Dunbar, A Kume, K Terao, M Hasegawa, K Ozawa
    Gene therapy 9(16) 1055-64 2002年8月  
    A major problem limiting hematopoietic stem cell (HSC) gene therapy is the low efficiency of gene transfer into human HSCs using retroviral vectors. Strategies, which would allow in vivo expansion of gene-modified hematopoietic cells, could circumvent the problem. To this end, we developed a selective amplifier gene (SAG) consisting of a chimeric gene composed of the granulocyte colony-stimulating factor (G-CSF) receptor gene and the estrogen receptor gene hormone-binding domain. We have previously demonstrated that primary bone marrow progenitor cells transduced with the SAG could be expanded in response to estrogen in vitro. In the present study, we evaluated the efficacy of the SAG in the setting of a clinically applicable cynomolgus monkey transplantation protocol. Cynomolgus bone marrow CD34(+) cells were transduced with retroviral vectors encoding the SAG and reinfused into each myeloablated monkey. Three of the six monkeys that received SAG transduced HSCs showed an increase in the levels of circulating progeny containing the provirus in vivo following administration of estrogen or tamoxifen without any serious adverse effects. In one monkey examined in detail, transduced hematopoietic progenitor cells were increased by several-fold (from 5% to 30%). Retroviral integration site analysis revealed that this observed increase was polyclonal and no outgrowth of a dominant single clonal population was observed. These results demonstrate that the inclusion of our SAG in the retroviral construct allows selective in vivo expansion of genetically modified cells by a non-toxic hormone treatment.
  • A Kume, M Koremoto, H Mizukami, T Okada, Y Hanazono, K Sugamura, K Ozawa
    BONE MARROW TRANSPLANTATION 30(2) 113-118 2002年7月  
    The cytokine receptor common gamma chain (gammac) plays a pivotal role in multiple interleukin signaling, and gammac gene mutations cause an X-linked form of SCID (X-SCID). Recently, gammac gene transfer into the autologous X-SCID BM achieved appreciable lymphocyte reconstitution, contrasting with the limited success in previous gene therapy trials targeting hematopoietic stem cells. To understand the mechanisms underlying this success, we examined the repopulating potential of the wild-type (WT) BM cells using an X-SCID mouse model. Limited numbers of WT cells were infused into non-ablated WT and X-SCID hosts. Whereas no appreciable engraftment was observed in WT recipients, donor-derived lymphocytes repopulated well in X-SCID, reaching 37% (10(6) cells given) and 53% (10(7) cells given) of the normal control value 5 months post BMT. A lineage analysis showed a predominance of the donor-derived lymphocytes (CD4(+) T, CD8(+) T, B and NK cells) in X-SCID while the donor-derived granulocytes and monocytes engrafted poorly. These results showed a selective advantage of WT cells in X-SCID, and that the advantage was restricted to lymphocytes. In human gene therapy for X-SCID, an analogous growth advantage would greatly enhance the repopulation of lymphocytes derived from a very small number of gammac gene-supplemented precursors.
  • K Ozawa, Y Hanazono, A Kume, T Nagashima, Y Ueda, K Terao, M Hasegawa
    BLOOD CELLS MOLECULES AND DISEASES 28(3) 342-342 2002年5月  
  • SI Muramatsu, KI Fujimoto, K Ikeguchi, N Shizuma, K Kawasaki, F Ono, Y Shen, LJ Wang, H Mizukami, A Kume, M Matsumura, Nagatsu, I, F Urano, H Ichinose, T Nagatsu, K Terao, Nakano, I, K Ozawa
    HUMAN GENE THERAPY 13(3) 345-354 2002年2月  
    One potential strategy for gene therapy of Parkinson's disease (PD) is the local production of dopamine (DA) in the striatum induced by restoring DA-synthesizing enzymes. In addition to tyrosine hydroxylase (TH) and aromatic-L-amino-acid decarboxylase (AADC), GTP cyclohydrolase I (GCH) is necessary for efficient DA production. Using adeno-associated virus (AAV) vectors, we previously demonstrated that expression of these three enzymes in the striatum resulted in long-term behavioral recovery in rat models of PD. We here extend the preclinical exploration to primate models of PD. Mixtures of three separate AAV vectors expressing TH, AADC, and GCH, respectively, were stereotaxically injected into the unilateral putamen of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated monkeys. Coexpression of the enzymes in the unilateral putamen resulted in remarkable improvement in manual dexterity on the contralateral to the AAV-TH/-AADC/-GCH-injected side. Behavioral recovery persisted during the observation period (four monkeys: 48 days, 65 days, 50 days, and >10 months, each). TH-immunoreactive (TH-IR), AADC-IR, and GCH-IR cells were present in a large region of the putamen. Microdialysis demonstrated that concentrations of DA in the AAV-TH/-AADC/-GCH-injected putamen were increased compared with the control side. Our results show that AAV vectors efficiently introduce DA-synthesizing enzyme genes into the striatum of primates with restoration of motor functions. This triple transduction method may offer a potential therapeutic strategy for PD.
  • H Shibata, Y Hanazono, T Nagashima, N Ageyama, T Asano, Y Ueda, A Kume, Kato, I, M Hasegawa, K Ozawa, Y Yoshikawa, K Terao
    ANTHROPOLOGICAL SCIENCE 110(1) 87-87 2002年1月  
  • A Kume, M Koremoto, RF Xu, T Okada, H Mizukami, Y Hanazono, M Hasagawa, K Ozawa
    BLOOD 98(11) 212A-213A 2001年11月  
  • Y Ueda, T Nagashima, Y Hanazono, H Shibata, A Kume, K Terao, N Komatsu, K Ozawa, M Hasegawa
    BLOOD 98(11) 212A-212A 2001年11月  
  • Y Hanazono, T Asano, Y Ueda, SI Muramatsu, A Kume, H Suemori, Y Suzuki, Y Kondo, K Harii, M Hasegawa, N Nakatsuji, K Ozawa
    BLOOD 98(11) 746A-746A 2001年11月  
  • 金沢丈治, 水上浩明, 岡田尚巳, 花園豊, 久米晃啓, 西野宏, 市村恵一, 小沢敬也
    日本癌学会総会記事 60th 254 2001年9月26日  
  • Y Hakamata, K Tahara, H Uchida, Y Sakuma, M Nakamura, A Kume, T Murakami, M Takahashi, R Takahashi, M Hirabayashi, M Ueda, Miyoshi, I, N Kasai, E Kobayashi
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 286(4) 779-785 2001年8月  
    The purpose of this study is to evaluate green fluorescent protein (GFP) transgenic rats for use as a tool for organ transplantation research. The GFP gene construct was designed to express ubiquitously. By flow cytometry, the cells obtained from the bone marrow, spleen, and peripheral blood of the GFP transgenic rats consisted of 77, 91, and 75% GFP-positive cells, respectively. To examine cell migration of GFP-positive cells after organ transplantation, pancreas graft with or without spleen transplantation, heart graft with or without lung transplantation, auxiliary liver and small bowel transplantation were also performed from GFP transgenic rat to LEW (RT1(1)) rats under a 2-week course of 0.64 mg/kg tacrolimus administration. GFP-positive donor cells were detected in the fully allogenic LEW rats after organ transplantation. These results showed that GFP transgenic rat is a useful tool for organ transplantation research such as cell migration study after organ transplantation without donor cell staining. (C) 2001 Academic Press.
  • 佐久間 康成, 久米 晃啓, 永井 秀雄, 内田 広夫, 田原 和典, 袴田 陽二, 上田 正次, 高橋 将文, 小林 英司
    日本消化器外科学会雑誌 34(7) 889-889 2001年7月  
  • K Kogure, M Urabe, H Mizukami, A Kume, Y Sato, J Monahan, K Ozawa
    INTERNATIONAL JOURNAL OF HEMATOLOGY 73(4) 469-475 2001年6月  
    Targeted integration of foreign DNA is ideal for gene therapy particularly when target cells such as hematopoietic cells actively divide and proliferate. Adeno-associated virus (AAV) has been shown to integrate its genome into a defined locus, AAVS1 (19q13.3-qter). The inverted terminal repeat (ITR) and Rep proteins are responsible for this site-specific integration, and a system has been developed that delivers a gene preferentially into AAVS1 by using these components of AAV. We examined whether this system could be: applied to gene transfer into K562 cells, Two rep, expression plasmids were tested, 1 driven by the cytomegalovirus (CMV) promoter (pCMVR78) and the other under the translational control of an internal ribosome entry site (pMGiR78) with mouse mammary tumor virus promoter. K562 cells were cotransfected with a rep plasmid and a plasmid containing a neo gene flanked by the ITRs. G418-resistant clones were isolated and analyzed by Southern blot analysis and fluorescence in situ hybridization (FISH). Southern blot analysis suggested AAVS1-specific integration of the neo gene in 6 (35%) of 17 clones when K562 cells were transfected with pMGiR78 by lipofection. FISH located the neo gene on chromosome 19 in 5 of these 6 clones (29%). Eight (32%) of 25 clones obtained by electroporation with pCMVR78 had the neo gene at AAVS1, according to Southern blot analysis, and 4 of these 8 clones (16%) were positive according to FISH analysis, These results suggest that site-specific integration of foreign DNA can be achieved at a significantly high rate in human hematopoietic cells using the AAV components. Int J Hematol. 2001;73:469-475. (C) 2001 The Japanese Society of Hematology.
  • M Kametaka, A Kume, H Mizukami, Y Hanazono, K Ozawa
    EXPERIMENTAL HEMATOLOGY 28(12) 1495-1495 2000年12月  
  • Y Hanazono, T Nagashima, H Shibata, N Ageyama, T Asano, Y Ueda, A Kume, K Terao, M Hasegawa, K Ozawa
    BLOOD 96(11) 524A-524A 2000年11月  
  • M Kametaka, A Kume, H Mizukami, Y Hanazono, K Ozawa
    BLOOD 96(11) 311B-311B 2000年11月  
  • A Kume, M Koremoto, Y Hanazono, H Mizukami, T Okada, K Sugamura, K Ozawa
    BLOOD 96(11) 211A-211A 2000年11月  
  • 花園豊, 長島建之, 柴田宏昭, 揚山直英, 浅野隆之, 上田泰次, 久米晃啓, 加藤郁之進, 小沢敬也
    Japanese Journal of Cancer Research 91(Supplement (Sept)) 37 2000年9月1日  
  • 金沢丈治, 水上浩明, 岡田尚巳, 花園豊, 久米晃啓, 西野宏, 市村恵一, 小沢敬也, MONAHAM J
    Jpn J Cancer Res 91(Supplement (Sept)) 201 2000年9月1日  
  • Y Shen, SI Muramatsu, K Ikeguchi, KI Fujimoto, DS Fan, M Ogawa, H Mizukami, M Urabe, A Kume, Nagatsu, I, F Urano, T Suzuki, H Ichinose, T Nagatsu, J Monahan, Nakano, I, K Ozawa
    HUMAN GENE THERAPY 11(11) 1509-1519 2000年7月  
    Parkinson's disease (PD), a neurological disease suited to gene therapy, is biochemically characterized by a severe decrease in the dopamine content of the striatum, One current strategy for gene therapy of PD involves local production of dopamine in the striatum achieved by inducing the expression of enzymes involved in the biosynthetic pathway for dopamine. We previously showed that the coexpression of tyrosine hydroxylase (TH) and aromatic-L-amino-acid decarboxylase (AADC), using two separate adeno-associated virus (AAV) vectors, resulted in more effective dopamine production and more remarkable behavioral recovery in 6-hydroxydopamine-lesioned parkinsonian rats, compared with the expression of TH alone. Not only levels of TH and AADC but also levels of tetrahydrobiopterin (BH(4)), a cofactor of TH, and GTP cyclohydrolase I (GCH), a rate-limiting enzymes for BH4 biosynthesis, are reduced in parkinsonian striatum, In the present study, we investigated whether transduction with separate AAV vectors expressing TH, AADC, and GCH was effective for gene therapy of PD, In vitro experiments showed that triple transduction with AAV-TH, AAV-AADC, and AAV-GCH resulted in greater dopamine production than double transduction with AAV-TH and AAV-AADC in 293 cells. Furthermore, triple transduction enhanced BH4 and dopamine production in denervated striatum of parkinsonian rats and improved the rotational behavior of the rats more efficiently than did double transduction. Behavioral recovery persisted for at least 12 months after stereotaxic intrastriatal injection. These results suggest that GCH, in addition to TH and AADC, is important for effective gene therapy of PD.
  • A Kume, R Xu, Y Ueda, M Urabe, K Ozawa
    GENE THERAPY 7(14) 1193-1199 2000年7月  
    Hematopoietic stem cells (HSCs) are attractive targets for gene therapy, but current gene transfer methodologies are inadequate for efficient HSC transduction and perpetual transgene expression. To improve gene transfer vectors and transduction protocols, it is vital to establish a system to evaluate transgene expression and the long-term behavior of transduced cells in vivo. For this purpose, we constructed a bicistronic retrovirus encoding the human CD24 (as the first cistron) and the enhanced green fluorescent protein (EGFP; as the second cistron). Murine bone marrow cells were transduced with this vector and the transgene expression was monitored along with hematopoietic reconstitution. Stable expression of CD24 and EGFP was demonstrated in the long-term repopulating cells for at least 6 months, and multi-parameter flow cytometry illustrated expression of both markers in all the lymphohematopoietic lineages examined (B and T lymphoid, erythroid and myeloid). Sustained expression was also shown in the secondary transplants for 6 months, suggesting that self-renewing HSCs were transduced by this vector. Overall, EGFP-tagged bicistronic retroviruses would provide powerful tools for detailed in vivo analysis of transduced hematopoietic cells, such as transgene expression in conjunction with lineage differentiation.
  • 長島建之, 上田泰次, 花園豊, 久米晃啓, 小松則夫, 小沢敬也, 長谷川護, XU R
    International Journal of Hematology. Supplement 71(1) 175 2000年4月  
  • 久米 晃啓
    日本小児血液学会雑誌 14(1) 1-8 2000年2月29日  
  • Journal of Clinical and Laboratory Medicine 135(2) 122-128 2000年  
  • K Kogure, M Urabe, A Kume, Y Sato, J Monahan, K Ozawa
    BLOOD 94(10) 412B-412B 1999年11月  
  • Y Hanazono, N Ageyama, H Shibata, T Nagashima, Y Ueda, M Hasegawa, Kato, I, A Kume, K Ozawa, K Terao
    BLOOD 94(10) 357A-357A 1999年11月  
  • R Xu, A Kume, Y Hanazono, KM Matsuda, Y Ueda, M Urabe, H Mano, M Hasegawa, K Ozawa
    BLOOD 94(10) 356A-356A 1999年11月  
  • 金沢丈治, 卜部匡司, 久米晃啓, 西野宏, MONAHAN J, 市村恵一, 小沢敬也
    日本癌学会総会記事 58th 732 1999年8月30日  
  • K Ozawa, KM Matsuda, R Xu, Y Ueda, M Urabe, A Kume, Y Miura, M Hasegawa
    EXPERIMENTAL HEMATOLOGY 27(7) 63-63 1999年7月  
  • RF Xu, A Kume, KM Matsuda, Y Ueda, H Kodaira, Y Ogasawara, M Urabe, Kato, I, M Hasegawa, K Ozawa
    JOURNAL OF GENE MEDICINE 1(4) 236-244 1999年7月  
    Background We have developed a novel system for expansion of gene-modified hematopoietic stem/progenitor cells to overcome the low efficiency of current gene transfer methodology. This system involves 'selective amplifier genes', that encode fusion proteins between the granulocyte colony-stimulating factor receptor (GCR) and the hormone-binding domain of estrogen receptor (ER). Hematopoietic progenitors expressing the chimeras showed estrogen-responsive growth in a controllable manner. However, endogenous estrogen may activate the fusion proteins in vivo, depending on the hormonal status of the subjects. Methods We replaced ER with a mutant receptor (TmR) which specifically binds to 4-hydroxytamoxifen (Tm), to overcome limitations with wild-type ER. Interleukin-3 (IL-3)-dependent Ba/F3 cells and hematopoietic progenitor cells transduced with the resultant fusion proteins (GCRTmR and Delta GCRTmR) were examined for ligand-inducible grouch. Results GCRTmR- and Delta GCRTmR-expressing Ba/F3 showed IL-3-independent growth in response to Tm, while the cells were unresponsive to estrogen at concentrations up to 10(-7)-10(-6) M. Furthermore, murine bone marrow cells transduced with GCRTmR and Delta GCRTmR formed colonies in methyl-cellulose medium in response to Tm, while virtually no colonies appeared with 10(-7) M estrogen or without cytokines. Conclusions These results suggest that influences of the endogenous estrogen can be almost eliminated by using the GCRTmR/Tm or Delta GCRTmR/ Tm system to expand gene-modified hematopoietic stem/progenitor cells. Copyright (C) 1999 John Wiley & Sons, Ltd.
  • A Kume, Y Hanazono, H Mizukami, M Urabe, K Ozawa
    INTERNATIONAL JOURNAL OF HEMATOLOGY 69(4) 227-233 1999年6月  
    Retrovirus-mediated gene transfer into murine hematopoietic stem cells and reconstitution of syngeneic mice have demonstrated persistence and functioning of the transgenes over extended periods of time. In contrast. clinically relevant levels of gene transfer into large animal and human stem cells have not been widely achieved. Results of current clinical gene transfer studies have raised fundamental questions about the physiology of primitive human hematopoietic cells and gene therapy vectors. Efforts are being undertaken to answer these problems and to develop more efficient gene therapy strategies. (C) 1999 The Japanese Society of Hematology.
  • A Kume, K Ito, Y Ueda, M Hasegawa, M Urabe, H Mano, K Ozawa
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 260(1) 9-12 1999年6月  
    We have developed a novel system for expansion of transduced hematopoietic cells. This system involves "selective amplifier genes" encoding fusion proteins between the granulocyte colony-stimulating factor receptor (Gcr) and the estrogen receptor (Er). The GcrEr chimeric gene conferred estrogen-dependent growth ability on murine hematopoietic cells. Here, we constructed a modified "selective amplifier gene" to circumvent possible concerns with the Er/estrogen switching system. The bacterial gyrase B (Gyr) gene was fused to the Gcr gene, and the GcrGyr fusion construct was introduced into interleukin-3 (IL-3)-dependent Ba/F3 cells. The dimeric antibiotic coumermycin induced IL-3-independent growth in Ba/F3 cells expressing GcrGyr, This stimulatory effect was antagonized by an excess amount of novobiocin, a monomeric form of coumermycin. These results suggest the feasibility of using Gyr as a molecular switch to regulate a growth signal in hematopoietic cells. (C) 1999 Academic.
  • KM Matsuda, A Kume, Y Ueda, M Urabe, M Hasegawa, K Ozawa
    GENE THERAPY 6(6) 1038-1044 1999年6月  
    We have proposed a novel concept, ie selective expansion of transduced cells, to overcome the low efficiency of gene transfer into hematopoietic stem cells. Previously, a fusion gene encoding a chimeric receptor (Delta GCRER) between the mouse granulocyte colony-stimulating factor receptor (G-CSFR) and the hormone-binding domain of rat estrogen receptor was constructed as a 'selective amplifier gene'. Although the chimeric gene conferred estrogen-inducible proliferation on the transduced Ba/F3 cells, it also mediated differentiation of the retrovirally transduced 32D cells upon estrogen treatment. Since only a growth signal is required for our purpose, we further modified the Delta GCRER gene to attenuate its differentiation signal. Based on the observation that tyrosine-703 in wild-type G-CSFR plays a pivotal role in transmitting the differentiation signal, phenylalanine was substituted for this residue in Delta GCRER. When the resultant selective amplifier gene (Delta Y703F-GCRER gene) was expressed in 32D cells, sustained growth was supported by estrogen, while differentiation was suppressed. These cells ceased to grow upon estrogen withdrawal and differentiated with G-CSF treatment. The present findings suggested that Delta Y703F-GCRER may have desirable properties as a selective amplifier for hematopoietic stem cell expansion and gene therapy.
  • 金沢丈治, 斗部匡司, 久米晃啓, 西野宏, MONAHAN J, 小沢敬也, 喜多村健
    頭頚部腫よう 25(1) 148-152 1999年3月25日  
  • HANAZONO Yutaka, KUME Akihiro, DUNBAR Cynthia E, DONAHUE Robert E, KATO Ikunoshin, HASEGAWA Mamoru, TERAO Keiji, OZAWA Keiya
    Keio journal of medicine 48 A31 1999年1月1日  
  • K Ozawa, D Fan, M Ogawa, M Urabe, A Kume, J Monahan, Nakano, I
    BIOGENIC AMINES 15(1) 21-37 1999年  
    An adeno-associated virus (AAV) vector is a potentially useful gene transfer vehicle for neurologic gene therapy by virtue of its unique characteristics, including lack of any associated disease with wild-type virus, the ability to transduce non-dividing cells, and prolonged expression of the transgene. Parkinson's disease (PD), an appropriate candidate for gene therapy, is characterized by the progressive loss of the dopaminergic neurons in the substantia nigra and a severe decrease in dopamine in the striatum. Current strategies for gene therapy of PD include 1) local production of dopamine in the striatum, and 2) protection of dopaminergic neurons in the substantia nigra. Dopamine is biosynthesized from tyrosine, which is first hydroxylated to L-dopa by tyrosine hydroxylase (TH). L-Dopa is then decarboxylated to dopamine by aromatic L-amino acid decarboxylase (AADC). Therefore, dopamine would be efficiently produced by the combined use of these dopamine-biosynthesizing enzyme genes. In a rodent model of PD (6-OHDA-lesioned rats), the TH expression in the denervated striatum has been reported to induce behavioral recovery. Since a very limited number of striatal cells produce AADC, we constructed two AAV vectors, AAV-TH and AAV-AADC, and attempted to coexpress both genes in striatal cells to enhance the dopamine synthesis. Lesioned rats were stereotaxically injected with the AAV-TH and/or AAV-AADC vectors into the denervated striatum. As a result, cotransduction with these two AAV vectors caused better behavioral recovery compared with AAV-TH alone. However, this approach can not halt the progressive degeneration of nigral dopaminergic neurons. To prevent dopaminergic neuron death, we are focusing on the protective effects of glial cell line-derived neurotrophic factor (GDNF), a potent neurotrophic factor For dopaminergic neurons. When rat E14 mesencephalic cells were transduced with AAV-GDNF, a larger number of dopaminergic neurons survived in cultures as compared with mock transduction. Furthermore, the dopaminergic neurons in the AAV-GDNF-transduced cultures grew more prominent neurites. Taken together, joint use of these therapeutic strategies, dopamine supplement gene therapy and GDNF gene therapy for protection of dopaminergic neurons, would be a logical approach for the treatment of PD.

共同研究・競争的資金等の研究課題

 27