Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Toshiaki Tabata.
Proceedings of the National Academy of Sciences of the United States of America | 2011
Hiroshi Ban; Naoki Nishishita; Noemi Fusaki; Toshiaki Tabata; Koichi Saeki; Masayuki Shikamura; Nozomi Takada; Makoto Inoue; Mamoru Hasegawa; Shin Kawamata; Shin-Ichi Nishikawa
After the first report of induced pluripotent stem cells (iPSCs), considerable efforts have been made to develop more efficient methods for generating iPSCs without foreign gene insertions. Here we show that Sendai virus vector, an RNA virus vector that carries no risk of integrating into the host genome, is a practical solution for the efficient generation of safer iPSCs. We improved the Sendai virus vectors by introducing temperature-sensitive mutations so that the vectors could be easily removed at nonpermissive temperatures. Using these vectors enabled the efficient production of viral/factor-free iPSCs from both human fibroblasts and CD34+ cord blood cells. Temperature-shift treatment was more effective in eliminating remaining viral vector-related genes. The resulting iPSCs expressed human embryonic stem cell markers and exhibited pluripotency. We suggest that generation of transgene-free iPSCs from cord blood cells should be an important step in providing allogeneic iPSC-derived therapy in the future.
Molecular Therapy | 2010
Katsuyuki Mitomo; U Griesenbach; Makoto Inoue; Lucinda Somerton; Cuixiang Meng; Eiji Akiba; Toshiaki Tabata; Yasuji Ueda; Gad Frankel; Raymond Farley; Charanjit Singh; Mario Chan; Felix M. Munkonge; Andrea Brum; Stefania Xenariou; Sara Escudero-Garcia; Mamoru Hasegawa; Eric W. F. W. Alton
Gene therapy for cystic fibrosis (CF) is making encouraging progress into clinical trials. However, further improvements in transduction efficiency are desired. To develop a novel gene transfer vector that is improved and truly effective for CF gene therapy, a simian immunodeficiency virus (SIV) was pseudotyped with envelope proteins from Sendai virus (SeV), which is known to efficiently transduce unconditioned airway epithelial cells from the apical side. This novel vector was evaluated in mice in vivo and in vitro directed toward CF gene therapy. Here, we show that (i) we can produce relevant titers of an SIV vector pseudotyped with SeV envelope proteins for in vivo use, (ii) this vector can transduce the respiratory epithelium of the murine nose in vivo at levels that may be relevant for clinical benefit in CF, (iii) this can be achieved in a single formulation, and without the need for preconditioning, (iv) expression can last for 15 months, (v) readministration is feasible, (vi) the vector can transduce human air-liquid interface (ALI) cultures, and (vii) functional CF transmembrane conductance regulator (CFTR) chloride channels can be generated in vitro. Our data suggest that this lentiviral vector may provide a step change in airway transduction efficiency relevant to a clinical programme of gene therapy for CF.
Gene Therapy | 2003
M. Miyazaki; Yasuhiro Ikeda; Yoshikazu Yonemitsu; Yoshinobu Goto; Taiji Sakamoto; Toshiaki Tabata; Yasuji Ueda; M Hasegawa; Shozo Tobimatsu; Tatsuro Ishibashi; Katsuo Sueishi
Retinitis pigmentosa (RP) is a heterogenous group of inherited retinal diseases resulting in adult blindness caused by mutations of various genes. Although it is difficult to cure the blindness that results from these diseases, delaying the disease progression may be of great benefit, since the majority of RP diseases are seen in middle age or later. To test a gene therapy strategy for RP using a neurotrophic factor gene, we assessed the effect of simian lentivirus (SIV)-mediated subretinal gene transfer of pigment epithelium-derived factor (PEDF), a potent neurotrophic factor, during the disease progression in Royal College of Surgeons (RCS) rats, a well-accepted animal model of RP. Regional gene transfer via SIV into the peripheral subretinal space at the nasal hemisphere was performed in all animals to monitor site-specific transgene expression as well as the therapeutic effect in each retina. Gene transfer of lacZ and PEDF was observed in the regional pigment epithelium corresponding to the regional gene transfer. Histologically, PEDF gene transfer significantly protected the loss of photoreceptor cells (PCs) corresponding to the regions of the gene transfer, compared to those of control groups during the course of the experiment. The antiapoptotic effect of PEDF on PCs is likely to be a related mechanism, because a significant reduction of terminal dUTP-nicked end labeling-positive PC numbers was found in PEDF-treated eyes compared to those of the control group (P<0.05). PEDF-treated eyes also retained a significant sensitivity to light flash during the experimental course. These findings clearly show that neuroprotective gene therapy using PEDF can protect retinal degeneration and functional defects in individuals with RP.
The FASEB Journal | 2006
Tsukasa Ohmori; Jun Mimuro; Katsuhiro Takano; Seiji Madoiwa; Yuji Kashiwakura; Akira Ishiwata; Masanori Niimura; Katsuyuki Mitomo; Toshiaki Tabata; Mamoru Hasegawa; Keiya Ozawa; Yoichi Sakata
Platelets release several mediators that modify vascular integrity and hemostasis. In the present study, we developed a technique for efficient transgene expression in platelets in vivo and examined whether this targeted‐gene‐product delivery system using a platelet release reaction could be exploited for clinical applications. Analysis of luciferase reporter gene constructs driven by platelet‐specific promoters (the GPIIb, GPIbα, and GPVI) revealed that the GPIbα promoter was the most potent in the megakaryoblastic cell line UT‐7/TPO and human CD34+‐derived megakaryocytes. Transduction of UT‐7/TPO;CD34+‐derived megakaryocytes; and c‐Kit+, ScaI+, and Lineage− (KSL) murine hematopoietic stem cells with a simian immunodeficiency virus (SIV)‐based lentiviral vector carrying eGFP resulted in efficient, dose‐dependent expression of eGFP, and the GPIbα promoter seemed to bestow megakaryocytic‐specific expression. Transplantation of KSL cells transduced with SIV vector containing eGFP into mice showed that there was preferable expression of eGFP in platelets driven by the GPIbα promoter [7–11% for the cytomeglovirus (CMV) promoter, 16–27% for the GPIbα promoter]. Furthermore, transplantation of ex vivo‐transduced KSL cells by SIV vector carrying human factorVIII (hFVIII) driven by the GPIbα promoter induced the production of detectable transcripts of the hFVIII gene and the hFVIII antigen in bone marrow and spleen for at least 90 days and partially corrected the hemophilia A phenotype. Platelet‐targeting gene therapy using SIV vectors appears to be promising for gene therapy approaches toward not only inherited platelet diseases but also other hemorrhagic disorders such as hemophilia A.—Ohmori, T., Mimuro, J., Takano, K., Madoiwa, S., Kashiwakura, Y., Ishiwata, A., Niimura, M., Mitomo, K., Tabata, T., Hasegawa, M., Ozawa, K., Sakata, Y. Efficient expression of a transgene in platelets using simian immunodeficiency virus‐based vector harboring glycoprotein Ibα promoter: in vivo model for platelet‐targeting gene therapy. FASEB J. 20, E769–E779 (2006)
Gene Therapy | 2003
Yasuhiro Ikeda; Yoshinobu Goto; Yoshikazu Yonemitsu; M. Miyazaki; Taiji Sakamoto; T. Ishibashi; Toshiaki Tabata; Yasuji Ueda; M Hasegawa; Shozo Tobimatsu; Katsuo Sueishi
Although lentivirus vectors hold promise for ocular gene therapy, they also have potential safety issues, particularly in the case of the current human immunodeficiency virus-based vectors. We recently developed a novel lentivirus vector derived from the nonpathogenic simian immunodeficiency virus from African green monkeys (SIVagm) to minimize these potentials. In this preclinical study, we evaluated whether SIV vector could be efficiently and safely applicable to retinal gene transfer by assessing the transgene expression, retinal function and histology over a 1-year period following subretinal injection in adult rats. The functional assessment via electroretinogram after both titers of SIV-lacZ (2.5 × 107 or 2.5 × 108 transducing units/ml) injection revealed both the dark and light adaptations to soon be impaired, in a dose-dependent manner, after a buffer injection as well, and all of them recovered to the control range by day 30. In both titers tested, the retinas demonstrated a frequent transgene expression mainly in the retinal pigment epithelium; however, the other retinal cells rarely expressed the transgene. Retinas exposed to a low titer virus showed no significant inflammatory reaction throughout the observation period, and also maintained the transgene expression over a 1-year period. In the retinas exposed to a high titer virus, however, mononuclear cell infiltration persisted in the subretinal area, and the retina that corresponded to the injected area finally underwent degeneration by around day 90. No retinal neoplastic lesions could be found in any animals over the 1-year period. We thus propose that SIV-mediated stable gene transfer might be useful for ocular gene transfer; however, more attention should be paid to avoiding complications when administering high titer lentivirus to the retina.
Journal of Gene Medicine | 2004
Jiro Kikuchi; Jun Mimuro; Kyoichi Ogata; Toshiaki Tabata; Yasuji Ueda; Akira Ishiwata; Konzoh Kimura; Katsuhiro Takano; Seiji Madoiwa; Hiroaki Mizukami; Yutaka Hanazono; Akihiro Kume; Mamoru Hasegawa; Keiya Ozawa; Yoichi Sakata
An Erratum has been published for this article in Journal of Gene Medicine 7(6), 2005, 836.
Human Gene Therapy | 2009
Yasuhiro Ikeda; Yoshikazu Yonemitsu; Masanori Miyazaki; Ri-ichiro Kohno; Yusuke Murakami; Toshinori Murata; Toshiaki Tabata; Yasuji Ueda; Fumiko Ono; Toshimichi Suzuki; Naohide Ageyama; Keiji Terao; Mamoru Hasegawa; Katsuo Sueishi; Tatsuro Ishibashi
Abstract Gene therapy may hold promise as a therapeutic approach for the treatment of intractable ocular diseases, including retinitis pigmentosa (RP). Gene transfer vectors that are able to show long-lasting transgene expression in vivo are highly desirable to treat RP; however, there is a dearth of information regarding long-term transgene expression in the eyes of large animals. We previously reported that the simian immunodeficiency virus from African green monkeys (SIVagm)-based lentiviral vector showed efficient, stable, and safe retinal gene transfer, resulting in significant prevention of retinal degeneration by gene transfer of a neurotrophic factor, human pigment epithelium-derived factor (hPEDF), in rodents. Before applying this strategy in a clinical setting, we here assessed the long-lasting transgene expression of our third-generation SIVagm-based lentiviral vectors in the retinal tissue of nonhuman primates. Approximately 20-50 mul of SIV-EGFP (enhanced green fluorescent protein) or SIV-hPEDF was injected into the subretinal space via a glass capillary tube. To detect EGFP expression in the retina, we used a fluorescence fundus camera at various time points after gene transfer. Human PEDF expression was assessed by immunohistochemical analysis, Western blot assay, and enzyme-linked immunosorbent assay. The retinas demonstrated frequent EGFP expression that was preserved for at least 4 years without significant decline. The expression of hPEDF was stable, and occurred mainly in the retinal pigment epithelium. The secreted protein was detected in vitreous and aqueous humor. We thus propose that SIVagm-mediated stable gene transfer might be significantly useful for ocular gene transfer in a clinical setting.
Gene Therapy | 2004
Kyoichi Ogata; Jun Mimuro; Jiro Kikuchi; Toshiaki Tabata; Yasuji Ueda; Masao Naito; Seiji Madoiwa; Katsuhiro Takano; Mamoru Hasegawa; Keiya Ozawa; Yoichi Sakata
We demonstrate that transduction of adipocytes with a simian immunodeficiency virus agm TYO1 (SIVagm)-based lentiviral vector carrying the human coagulation factor VIII gene (SIVhFVIII) resulted in expression of the human FVIII transgene in vitro and in db/db mice in vivo. Cultured human adipocytes were transduced with the SIVagm vector carrying the GFP gene in a dose-dependent manner and transduction of adipocytes with SIVhFVIII resulted in efficient expression of human coagulation factor VIII (hFVIII; 320±39.8 ng/106 adipocytes/24 h) in vitro. Based upon successful transduction of adipocytes by SIV vectors carrying the lacZ gene in vivo in mice, the adipose tissue of db/db mice was transduced with SIVhFVIII. There was a transient appearance of human FVIII in mouse plasma (maximum 1.8 ng/ml) on day 11 after the injection. Transcripts of human FVIII transgene and human FVIII antigen also were detected in the adipose tissue by RT-PCR and immunofluorescence, respectively, on day 14. Emergence of anti-human FVIII antibodies 14 days after the injection of SIVhFVIII may explain the disappearance of human FVIII from the circulation. These results suggest that transduction of the adipocytes with vectors carrying the human FVIII gene may be potentially applicable for gene therapy of hemophilia A.
Human Gene Therapy | 2009
Yasuhiro Ikeda; Yoshikazu Yonemitsu; Masanori Miyazaki; Ri Ichiro Kohno; Yusuke Murakami; Toshinori Murata; Yoshinobu Goto; Toshiaki Tabata; Yasuji Ueda; Fumiko Ono; Toshimichi Suzuki; Naohide Ageyama; Keiji Terao; Mamoru Hasegawa; Katsuo Sueishi; Tatsuro Ishibashi
A phase 1 clinical trial evaluating the safety of gene therapy for patients with wet age-related macular degeneration (AMD) or retinoblastoma has been completed without problems. The efficacy of gene therapy for Lebers congenital amaurosis (LCA) was reported by three groups. Gene therapy may thus hold promise as a therapeutic method for the treatment of intractable ocular diseases. However, it will first be important to precisely evaluate the efficiency and safety of alternative gene transfer vectors in a preclinical study using large animals. In the present study, we evaluated the acute local (ophthalmic) and systemic toxicity of our simian immunodeficiency virus from African green monkeys (SIVagm)-based lentiviral vectors carrying human pigment epithelium-derived factor (SIV-hPEDF) for transferring genes into nonhuman primate retinas. Transient inflammation and elevation of intraocular pressure were observed in some animals, but these effects were not dose dependent. Electroretinograms (ERGs), including multifocal ERGs, revealed no remarkable change in retinal function. Histopathologically, SIV-hPEDF administration resulted in a certain degree of inflammatory reaction and no apparent structural destruction in retinal tissue. Regarding systemic toxicity, none of the animals died, and none showed any serious side effects during the experimental course. No vector leakage was detected in serum or urine samples. We thus propose that SIVagm-mediated stable gene transfer might be useful and safe for ocular gene transfer in a clinical setting.
Gene Therapy and Regulation | 2004
Kyoji Ueda; Yutaka Hanazono; Naohide Ageyama; Hiroaki Shibata; Satoko Ogata; Yasuji Ueda; Toshiaki Tabata; Susumu Ikehara; Masafumi Taniwaki; Mamoru Hasegawa; Keiji Terao; Keiya Ozawa
It has recently been reported that bone marrow cells can efficiently engraft without marrow conditioning when implanted directly into the bone marrow cavity (intra-bone marrow transplantation, iBMT) in mice. We have successfully examined the efficacy of autologous iBMT in a cynomolgus monkey model in conjuction with an in vivo expansion of transplanted cells by a selective amplifier transgene (Ueda et al., 2004) and provide here the detailed parameters of our iBMT method. We injected retrovirally-marked autologous CD34+ cells directly into the non-conditioned marrow cavity of the femur and humerus after gently irrigating the cavity with saline. This transplant procedure was safely performed without pulmonary embolism. Gene-marked cells were not detectable in the peripheral blood at one hour and one day after iBMT as assessed by sensitive PCR, indicating that iBMT hardly generated a systemic delivery of transplanted cells. On the other hand, 2 to 30% of clonogenic hematopoietic colonies produced from the implanted marrow were gene-marked at 6–12 months after iBMT. Our iBMT method for non-human primates is thus discussed in terms of long-lived hematopoietic stem/progenitor cells, bone marrow niche and long-term engraftment after iBMT without myeloablative conditioning.