Network


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

Hotspot


Dive into the research topics where Megumi Takehara is active.

Publication


Featured researches published by Megumi Takehara.


Transplantation | 2003

Long-term acceptance of rat cardiac allografts on the basis of adenovirus mediated CD40Ig plus CTLA4Ig gene therapies

Kenichiro Yamashita; Taro Masunaga; Naoyuki Yanagida; Megumi Takehara; Taku Hashimoto; Tokushi Kobayashi; Hayato Echizenya; Nan Hua; Miri Fujita; Masaaki Murakami; Hiroyuki Furukawa; T Uede; Satoru Todo

Background. We have previously demonstrated that blockade of either CD80/86-CD28 or CD40-CD154 costimulatory pathways by using adenovirus vector coding CTLA4Ig (AdCTLA4Ig) or CD40Ig (AdCD40Ig) genes induced donor-specific tolerance in rat liver transplantation. In this study, we asked whether these gene–therapy-based costimulation blockade would induce tolerance in cardiac transplantation. Methods. Heterotopic heart transplantation was performed in a full major histocompatibility complex (MHC) barrier combination of ACI (RT1avl) to Lewis (LEW, RT1l) rats. Vector (1x109 plaque forming unit [PFU]), AdLacZ, AdCTLA4Ig, or AdCD40Ig, was administered intravenously to recipient animals immediately after grafting, and graft survival, serum CTLA4Ig/CD40Ig levels, and graft histology were assessed. Tolerance was determined by secondary skin-graft challenging. Results. Allografts of both untreated and AdLacZ controls were promptly rejected within 7 days, whereas a single treatment with AdCTLA4Ig or AdCD40Ig significantly prolonged median graft survival to 55.5 and 28.5 days, respectively. In contrast, the combined AdCTLA4Ig and AdCD40Ig gene therapy maintained high CTLA4Ig and CD40Ig levels through the posttransplant period and allowed long-term cardiac allograft survival for more than 270 days. However, both donor and third-party skin grafts were rejected in the animals who harbored cardiac grafts over 150 days. Also, typical features of chronic rejection were evident in the long-term surviving grafts. Conclusion. Simultaneous blockade of CD28 and CD154 pathways by AdCTLA4Ig plus AdCD40Ig induces a strong immunosuppression that allows long-term acceptance of full MHC mismatched cardiac graft in rats. This strategy, however, was not enough to induce tolerance to skin grafts and to avoid chronic rejection, as shown in the liver-transplantation model.


Transplantation | 2002

Induction of donor-specific tolerance by adenovirus-mediated CD40Ig gene therapy in rat liver transplantation

M Nomura; Kenichiro Yamashita; Masaaki Murakami; Megumi Takehara; Hayato Echizenya; Masao Sunahara; Norihiko Kitagawa; Miri Fujita; Hiroyuki Furukawa; T Uede; Satoru Todo

Background. Blockade of CD40-CD40 ligand (CD154) costimulatory pathway with anti-CD154 antibody (Ab) prolongs allograft survival in experimental organ transplantations; however, repeated agent administration is needed to provide an adequate immunosuppression. Seeking for simple and effective approach to interfere this signaling, we applied adenovirus-mediated gene therapy by encoding CD40Ig gene (AdCD40Ig). Methods. Liver graft from ACI (RT1av1) rat was transplanted orthotopically into LEW (RT1l) rat, and AdCD40Ig was given to animals via the penile vein immediately after grafting (n=6). Results. A single treatment with AdCD40Ig at 1×109 plaque forming units induced specific expression of CD40Ig gene on allograft liver, produced substantial amount of the protein in the sera, and allowed indefinite graft survival. Whereas, LEW recipients given no treatment or control adenovirus vector (AdLacZ) promptly rejected ACI liver. In addition, AdCD40Ig-treated, long-term survivors accepted skin graft from the donor strain but not the third party graft. Histopathology revealed that liver structure of the long-term surviving animals was completely preserved in normal with no infiltration of mononuclear cells. Conclusion. Blockade of CD40-CD154 pathway by CD40Ig gene therapy is a potent alloantigen-specific immunosuppressive strategy to induce permanent acceptance of liver allograft and would be a new therapeutic candidate in a clinical liver transplantation.


Transplantation | 2000

Blocking the CD28-B7 T-cell costimulatory pathway abrogates the development of obliterative bronchiolitis in a murine heterotopic airway model.

Akira Yamada; Katsuhito Konishi; Georgina Lou E. Cruz; Megumi Takehara; Masayuki Morikawa; Izumi Nakagawa; Masaaki Murakami; Tomio Abe; Satoru Todo; Toshimitsu Uede

BACKGROUND CTLA4IgG that binds to B7 effectively inhibits the signaling of CD28/B7 pathway and induces antigen-specific T-cell unresponsiveness in vitro and in vivo. We examined whether the development of obliterative bronchiolitis in a murine heterotopic airway transplantation model is T cell dependent and whether CTLA4IgG abrogates the development of obliterative bronchiolitis. METHODS Tracheae with main bronchi from C3H/He (H2k), BALB/C (H2d), or C57BL/6 (H2b) mice were transplanted heterotopically into subcutaneous pockets on the backs of BALB/C or BALB/C nu/nu mice on day 0. Recipient mice were untreated or intraperitoneally treated with either CTLA4IgG or human IgG with different time and dose schedules. RESULTS The development of obliterative bronchiolitis, which leads to luminal obliteration by fibrous tissue in a murine heterotopic airway transplantation model, was T cell dependent and the development of obliterative bronchiolitis was significantly abrogated by the CTLA4IgG treatment. However, the normal ciliated columnar respiratory epithelial cells in allografts were lost and replaced by flattened attenuated epithelial cells even after the CTLA4IgG treatment. We further demonstrated that CTLA4IgG treatment did not result in the induction of donor-specific unresponsiveness. CONCLUSIONS We demonstrated that the development of obliterative bronchiolitis in a murine heterotopic airway model involves both CD28/B7-dependent and -independent processes. The luminal obliteration by fibrous tissue is clearly CD28/B7 dependent and can be inhibited by CTLA4IgG. The luminal obliteration of allografted trachea by fibrous tissues and the loss of ciliated columnar respiratory epithelial cells represent distinct disease processes.


American Journal of Transplantation | 2002

Prevention of Acute Lung Allograft Rejection in Rat by CTLA4Ig

Takeshi Shiraishi; Yohichi Yasunami; Megumi Takehara; Toshimitsu Uede; Katsunobu Kawahara; Takayuki Shirakusa

CTLA4 immunoglobulin (CTLA4Ig), which binds with a high affinity to B7‐1 and B7‐2, interrupts T‐cell activation by inhibiting costimulatory signal. CTLA4Ig has been used in hopes of achieving antigen‐specific tolerance induction in several solid organ transplants. In lung allograft rejection, however, its use has been controversial in terms of its effect on prevention of rejection. In the present study, the effect of murine CTLA4Ig on rat‐lung allograft rejection was investigated. Rat left‐lung transplantation was performed in an RT1 incompatible donor (Brown Norway; BN)–recipient (F344) combination. All allografts (n = 12) without any treatment were rejected within 7 days after transplantation. A single injection of murine form CTLA4Ig at a dose of 100 μg intraperitoneally (ip) or intravenously (iv) on day 1 post‐transplantation achieved long‐term graft survival (> 90 days) in 2/5 (40%) and 3/8 (38%), respectively. Moreover, 6/7 (86%) allografts in rats that received iv injection of 500 μg CTLA4Ig survived more than 90 days. Allograft survival in the CTLA4Ig 500 μg iv recipient group was significantly longer than that in the no‐treatment control or control immunoglobulin group (p < 0.01). Four out of seven recipients bearing functional allografts for more than 90 days with the CTLA4Ig treatment accepted donor‐specific skin grafts, whereas all third‐party skin grafts (n = 3) were rejected. Prevention of rat‐lung allograft rejection could be achieved by intravenous administration of CTLA4Ig, resulting in long‐term allograft survival with acceptance of donor‐specific skin grafts.


Human Gene Therapy | 2001

Long-Term Acceptance of Allografts by in Vivo Gene Transfer of Regulatable Adenovirus Vector Containing CTLA4IgG and loxP

Megumi Takehara; Masaaki Murakami; Manabu Inobe; Kumiko Tanaka; Shunsuke Chikuma; Izumu Saito; Yumi Kanegae; Yohichi Yasunami; Masahiko Nakano; Kenichiro Yamashita; Satoru Todo; Toshimitsu Uede

CTLA4IgG was shown to inhibit the costimulatory signal for T cell activation by interfering with the ligation of CD28 and B7-1 or B7-2. To inhibit various immune responses including acute cellular rejection of allografts, a certain level of serum CTLA4IgG should be maintained for an appropriate period. We previously reported on an adenovirus vector containing CTLA4IgG, which we designated Adex1CACTLA4IgG. Adex1CACTLA4IgG was able to maintain a significant level of serum CTLA4IgG for a long period on intravenous injection, which in turn inhibited various immune responses including protective immunity against infectious agents. To overcome the inhibitory effect, we constructed a new adenovirus vector, Adex1CALoxCTLA4IgGLox, by cloning CTLA4IgG cDNA between two loxP sequences under the control of the CAG promoter. We demonstrated that the administration of adenovirus vector containing Cre recombinase gene (Adex1CACre) at the desired time induced Cre-mediated recombination within a gene derived from Adex1CALoxCTLA4IgGLox vector, and the cDNA of CTLA4IgG was excised from the transduced gene and terminated the expression of CTLA4IgG in vitro and in vivo. More importantly, we also demonstrated that the long-term acceptance of allografts was achieved after the termination of CTLA4IgG expression, while the immune response against adenovirus was restored.


Pancreas | 2004

Amelioration of hyperglycemia in streptozotocin-induced diabetic mice with fetal pancreatic allografts: prevention of rejection by donor specific transfusion in conjunction with CTLA4Ig.

Eiji Hirakawa; Yohichi Yasunami; Masahiko Nakano; Masafumi Shiiba; Megumi Takehara; Toshimitsu Uede; Satoru Todo; Junko Ono; Seiyo Ikeda

Introduction Fetal pancreas has been considered as an alternative donor source for islet transplantation since it has potent capacity for &bgr; cell differentiation and proliferation. However, prevention of fetal pancreatic allograft rejection can be hardly achieved compared with adult islet allografts. Aims The aim of the study is to determine whether donor specific transfusion (DST) in conjunction with CTLA4Ig has any favorable effect on prevention of fetal pancreatic allograft rejection in mice. Methods BALB/c splenocytes (SPC, 1 × 107) were injected iv into C57BL/6 mice in conjunction with CTLA4Ig (ip, 50 &mgr;g, day 0, 2, and 4). Fourteen days later, the mice were made diabetic with streptozotocin (STZ, iv) and donor specific or third party pancreatic allografts were transplanted beneath the kidney capsule. Results Morphologically, it was found that rejection of fetal pancreatic allografts can be prevented at 30 days after transplantation only when donor specific allografts were grafted into the mice treated with DST in conjunction with CTLA4Ig. Functionally, 3 out of 9 diabetic mice became normoglycemic by 120 days after transplantation of fetal pancreatic allografts. Conclusion DST in conjunction with CTLA4Ig can have a favorable effect on prevention of fetal pancreatic allograft rejection resulting in amelioration of STZ-induced diabetes in mice.


Transplantation Proceedings | 2001

Tolerance induction by a single donor pretreatment with the adenovirus vector encoding CTLA4Ig gene in rat orthotopic liver transplantation

Naoyuki Yanagida; M Nomura; Kenichiro Yamashita; Megumi Takehara; Masaaki Murakami; Hayato Echizenya; Katsuhito Konishi; Norihiko Kitagawa; Hiroyuki Furukawa; T Uede; Satoru Todo


Transplantation Proceedings | 1999

Effects of a novel immunosuppressant, FTY720, on heart and liver transplantations in rats

Kenichiro Yamashita; M Nomura; T Omura; Megumi Takehara; Tomomi Suzuki; Tsuyoshi Shimamura; Akihiro Kishida; Hiroyuki Furukawa; Masaaki Murakami; Toshimitu Uede; Satoru Todo


Transplantation Proceedings | 2001

Adenovirus-mediated CTLA4-IgG gene therapy in orthotopic small intestinal transplantation in rats

Hayato Echizenya; Kenichiro Yamashita; Megumi Takehara; Katsuhito Konishi; M Nomura; Naoyuki Yanagida; Norihiko Kitagawa; T Kobayashi; Hiroyuki Furukawa; Manabu Inobe; T Uede; Satoru Todo


Cardiovascular Research | 2006

Cre/loxP-mediated CTLA4IgG gene transfer induces clinically relevant immunosuppression via on-off gene recombination in vivo

Atsushi Izawa; Kenji Sano; Megumi Takehara; Manabu Inobe; Jun-ichi Suzuki; Hiroshi Imamura; Masafumi Takahashi; Uichi Ikeda; Mitsuaki Isobe; Toshimitsu Uede

Collaboration


Dive into the Megumi Takehara's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroyuki Furukawa

Asahikawa Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

T Uede

Hokkaido University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge