Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where M Hasegawa is active.

Publication


Featured researches published by M Hasegawa.


Gene Therapy | 2003

Simian lentiviral vector-mediated retinal gene transfer of pigment epithelium-derived factor protects retinal degeneration and electrical defect in Royal College of Surgeons rats

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.


Gene Therapy | 2007

Sendai virus-mediated CFTR gene transfer to the airway epithelium

Stefano Ferrari; U Griesenbach; Akihiro Iida; Raymond Farley; Angela Wright; Jie Zhu; Felix M. Munkonge; Stephen N Smith; Jun You; H Ban; Makoto Inoue; Mario Chan; Charanjit Singh; Bernard Verdon; Barry E. Argent; Brandon J. Wainwright; Peter K. Jeffery; Duncan M. Geddes; David J. Porteous; S C Hyde; Michael A. Gray; M Hasegawa; E W F W Alton

The potential for gene therapy to be an effective treatment for cystic fibrosis has been hampered by the limited gene transfer efficiency of current vectors. We have shown that recombinant Sendai virus (SeV) is highly efficient in mediating gene transfer to differentiated airway epithelial cells, because of its capacity to overcome the intra- and extracellular barriers known to limit gene delivery. Here, we have identified a novel method to allow the cystic fibrosis transmembrane conductance regulator (CFTR) cDNA sequence to be inserted within SeV (SeV-CFTR). Following in vitro transduction with SeV-CFTR, a chloride-selective current was observed using whole-cell and single-channel patch-clamp techniques. SeV-CFTR administration to the nasal epithelium of cystic fibrosis (CF) mice (CftrG551D and Cftrtm1UncTgN(FABPCFTR)#Jaw mice) led to partial correction of the CF chloride transport defect. In addition, when compared to a SeV control vector, a higher degree of inflammation and epithelial damage was found in the nasal epithelium of mice treated with SeV-CFTR. Second-generation transmission-incompetent F-deleted SeV-CFTR led to similar correction of the CF chloride transport defect in vivo as first-generation transmission-competent vectors. Further modifications to the vector or the host may make it easier to translate these studies into clinical trials of cystic fibrosis.


Gene Therapy | 2004

A defective nontransmissible recombinant Sendai virus mediates efficient gene transfer to airway epithelium in vivo.

Stefano Ferrari; U Griesenbach; T Shiraki-Iida; T Shu; Takashi Hironaka; X Hou; J Williams; Jie Zhu; Peter K. Jeffery; Duncan M. Geddes; M Hasegawa; Ewfw Alton

Recombinant Sendai virus (SeV)-mediated gene transfer to differentiated airway epithelial cells has shown to be very efficient, because of its ability to overcome the intra- and extracellular barriers known to limit gene delivery. However, this virus is transmission competent and therefore unlikely to be suitable for use in clinical trials. A nontransmissible, replication-competent recombinant SeV has recently been developed by deleting the envelope Fusion (F) protein gene (SeV/ΔF). Here we show that SeV/ΔF is able to mediate β-galactosidase reporter gene transfer to the respiratory tract of mice in vivo, as well as to human nasal epithelial cells in vitro. Further, in an ex vivo model of differentiated airway epithelium, SeV/ΔF gene transfer was not importantly inhibited by native mucus. When compared to the transmission-competent SeV in vivo, no difference in gene expression was observed at the time of peak expression. The development of an F-defective nontransmissible SeV, which can still efficiently mediate gene transfer to the airway epithelium, represents the first important step towards the use of a cytoplasmic RNA viral vector in clinical trials of gene therapy.


Gene Therapy | 2009

Generation of optimized and urokinase-targeted oncolytic Sendai virus vectors applicable for various human malignancies

Hiroaki Kinoh; Makoto Inoue; Atsushi Komaru; Yasuji Ueda; M Hasegawa; Yoshikazu Yonemitsu

We previously reported the development of a prototype ‘oncolytic Sendai virus (SeV) vector’ formed by introducing two major genomic modifications to the original SeV, namely deletion of the matrix (M) gene to avoid budding of secondary viral particles and manipulation of the trypsin-dependent cleavage site of the fusion (F) gene to generate protease-specific sequences. As a result, the ‘oncolytic SeV’ that was susceptible to matrix metalloproteinases (MMPs) was shown to selectively kill MMP-expressing tumors through syncytium formation in vitro and in vivo. However, its efficacy has been relatively limited because of the requirement of higher expression of MMPs and smaller populations of MMP-expressing tumors. To overcome these limitations, we have designed an optimized and dramatically powerful oncolytic SeV vector. Truncation of 14-amino acid residues of the cytoplasmic domain of F protein resulted in dramatic enhancement of cell-killing activities of oncolytic SeV, and the combination with replacement of the trypsin cleavage site with the new urokinase type plasminogen activator (uPA)-sensitive sequence (SGRS) led a variety of human tumors, including prostate (PC-3), renal (CAKI-I), pancreatic (BxPC3) and lung (PC14) cancers, to extensive death through massive cell-to-cell spreading without significant dissemination to the surrounding noncancerous tissue in vivo. These results indicate a dramatic improvement of antitumor activity; therefore, extensive utility of the newly designed uPA-targeted oncolytic SeV has significant potential for treating patients bearing urokinase-expressing cancers in clinical settings.


Gene Therapy | 2003

Simian immunodeficiency virus-based lentivirus vector for retinal gene transfer: a preclinical safety study in adult rats

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.


Gene Therapy | 2007

Expression and maturation of Sendai virus vector-derived CFTR protein : functional and biochemical evidence using a GFP-CFTR fusion protein

H Ban; Makoto Inoue; U Griesenbach; Felix M. Munkonge; Mario Chan; Akihiro Iida; E W F W Alton; M Hasegawa

Sendai virus (SeV) vector has been shown to efficiently transduce airway epithelial cells. As a precursor to the potential use of this vector for cystic fibrosis (CF) gene therapy, the correct maturation of the SeV vector-derived CF transmembrane conductance regulator (CFTR) protein was examined using biochemical and functional analyses. We constructed a recombinant SeV vector, based on the fusion (F) gene-deleted non-transmissible SeV vector, carrying the GFP-CFTR gene in which the N terminus of CFTR was fused to green fluorescence protein (GFP). This vector was recovered and propagated to high titers in the packaging cell line. Western blotting using an anti-GFP antibody detected both the fully glycosylated (mature) and the core-glycosylated (immature) proteins, indicating that SeV vector-derived GFP-CFTR was similar to endogenous CFTR. We also confirmed the functional channel activity of GFP-CFTR in an iodide efflux assay. The efficient expression of GFP-CFTR, and its apical surface localization, were observed in both MDCK cells in vitro, and in the nasal epithelium of mice in vivo. We concluded that recombinant SeV vector, a cytoplasmically maintained RNA vector, is able to direct production of a correctly localized, mature form of CFTR, suggesting the value of this vector for studies of CF gene therapy.


Gene Therapy | 2009

Complete elimination of established neuroblastoma by synergistic action of |[gamma]|-irradiation and DCs treated with rSeV expressing interferon-|[beta]| gene

K Tatsuta; Sakura Tanaka; T Tajiri; Satoko Shibata; Atsushi Komaru; Yasuji Ueda; Makoto Inoue; M Hasegawa; Sachiyo Suita; Katsuo Sueishi; Tomoaki Taguchi; Yoshikazu Yonemitsu

Dendritic cell (DC)-based immunotherapy has been investigated as a new therapeutic approach to intractable neuroblastomas; however, only limited clinical effect has been reported. To overcome the relatively low sensitivity of neuroblastomas against immunotherapy, we undertook a preclinical efficacy study to examine murine models to assess the combined effects of γ-irradiation pretreatment and recombinant Sendai virus (ts-rSeV/dF)-mediated murine interferon-β (mIFN-β) gene transfer to DCs using established c1300 neuroblastomas. Similar to intractable neuroblastomas in the clinic, established c1300 tumors were highly resistant to monotherapy with either γ-irradiation or DCs activated by ts-rSeV/dF without transgene (ts-rSeV/dF-null) that has been shown to be effective against other murine tumors, including B16F10 melanoma. In contrast, immunotherapy using DCs expressing mIFN-β through ts-rSeV/dF (ts-rSeV/dF-mIFNβ-DCs) effectively reduced tumor size, and its combination with γ-irradiation pretreatment dramatically enhanced its antitumor effect, resulting frequently in the complete elimination of established c1300 tumors 7–9u2009mm in diameter, in a high survival rate among mice, and in the development of protective immunity in the mice against rechallenge by the tumor cells. These results indicate that the combination of ts-rSeV/dF-mIFNβ-DCs with γ-irradiation is a hopeful strategy for the treatment of intractable neuroblastomas, warranting further investigation in the clinical setting.


Gene Therapy | 2006

Effect of tolerance induction to immunodominant T-cell epitopes of Sendai virus on gene expression following repeat administration to lung

U Griesenbach; Rosemary J. Boyton; L. Somerton; S E Garcia; Stefano Ferrari; T Owaki; Z Ya-Fen; Duncan M. Geddes; M Hasegawa; Daniel M. Altmann; E W F W Alton

Sendai virus (SeV) is able to transfect airway epithelial cells efficiently in vivo. However, as with other viral vectors, repeated administration leads to reduced gene expression. We have investigated the impact of inducing immunological tolerance to immunodominant T-cell epitopes on gene expression following repeated administration. Immunodominant CD4 and CD8 T-cell peptide epitopes of SeV were administered to C57BL/6 mice intranasally 10 days before the first virus administration with transmission-incompetent F-protein-deleted ΔF/SeV-GFP. At 21 days after the first virus administration, mice were again transfected with ΔF/SeV. To avoid interference of anti-GFP antibodies, the second transfection was carried out with ΔF/SeV-lacZ. At 2 days after the final transfection lung β-galactosidase expression, T-cell proliferation and antibody responses were measured. A state of ‘split tolerance’ was achieved with reduced T-cell proliferation, but no impact on antiviral antibody production. There was no enhancement of expression on repeat administration; instead, T-cell tolerance was, paradoxically, associated with a more profound extinction of viral expression. Multiple immune mechanisms operate to eradicate viruses from the lung, and these findings indicate that impeding the adaptive T-cell response to the immunodominant viral epitope is not sufficient to prevent the process.


Frontiers in Bioscience | 2008

Immunostimulatory virotherapy using recombinant sendai virus as a new cancer therapeutic regimen

Yoshikazu Yonemitsu; Yasuji Ueda; Hiroaki Kinoh; M Hasegawa

The utility of recombinant Sendai virus (rSeV) has been considerably examined over the last decade as a potent gene transfer candidate in a cytoplasmic gene expression system. Such risks as excessive immune responses associated with this virus administration in vivo however have limited its applicability in clinical settings as is the case with other viral vectors including adenoviruses. In consequence of extensive assessment on the mechanisms of immune responses against SeV, we found that ex vivo infection of immature dendritic cells (DCs) with SeV demonstrates their spontaneous maturation and activation. We applied this result to create a unique, representative, and powerful agent to activate DCs, namely rSeV-modified DCs (rSeV/DCs), for use in cancer immunotherapy. Use of this system in vivo resulted in the induction of efficient antitumor immunity against vascularized rodent tumors, including melanoma, hepatocellular carcinoma, neuroblastoma, squamous cell carcinoma, and prostatic cancer, and it even frequently associated with elimination of those tumors. These results indicate that rSeV could be a powerful immune booster for DC-based cancer immunotherapy that is worth investigating further. We propose a conceptual term immunostimulatory virotherapy to describe this new method of cancer therapy using the rSeV/DCs system.


Gene Therapy | 2007

Impact of deletion of envelope-related genes of recombinant Sendai viruses on immune responses following pulmonary gene transfer of neonatal mice

Sakura Tanaka; Yoshikazu Yonemitsu; Kumi Yoshida; Shinji Okano; Haruhiko Kondo; Makoto Inoue; M Hasegawa; Kouji Masumoto; Sachiyo Suita; Tomoaki Taguchi; Katsuo Sueishi

We demonstrated previously that the additive-type recombinant Sendai virus (rSeV) is highly efficient for use in pulmonary gene transfer; however, rSeV exhibits inflammatory responses. To overcome this problem, we tested newly developed non-transmissible constructs, namely, temperature-sensitive F-deleted vector, rSeV/dF (ts-rSeV/dF) and a rSeV with all the envelope-related genes deleted (rSeV/dFdMdHN), for pulmonary gene transfer in neonatal mice, by assessing their toxicity and immune responses. The gene expression in the lungs of neonatal ICR mice peaked on day 2, then gradually decreased until almost disappearing at 14 days after infection in all constructs. Loss of body weight and mortality rate, however, were dramatically improved in mice treated with SeV/dFdMdHN (mortality=0%, n=41) and ts-rSeV/dF (24.2%, n=33) compared with additive rSeV (70.7%, n=58). Although the deletion of envelope-related genes of SeV had a small impact on the production of antibody and cytotoxic T-lymphocyte activity in both adults and neonates, a dramatic reduction was found in the events related to innate responses, including the production of proinflammatory cytokines, particularly in the case of neonates. These results indicate that pulmonary gene transfer using SeV/dFdMdHN warrants further investigation for its possible use in developing safer therapeutics for neonatal lung diseases, including cystic fibrosis.

Collaboration


Dive into the M Hasegawa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

U Griesenbach

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mario Chan

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoshinobu Goto

International University of Health and Welfare

View shared research outputs
Researchain Logo
Decentralizing Knowledge