Network


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

Hotspot


Dive into the research topics where Ayako Wakabayashi is active.

Publication


Featured researches published by Ayako Wakabayashi.


Immunobiology | 2012

Disruption of maternal immune balance maintained by innate DC subsets results in spontaneous pregnancy loss in mice.

Yasuyuki Negishi; Ayako Wakabayashi; Masumi Shimizu; Tomoko Ichikawa; Yoshihiro Kumagai; Toshiyuki Takeshita; Hidemi Takahashi

Dendritic cells (DCs) play an important role in providing an appropriate fetal/maternal balance between Th1 and Th2 during pregnancy. The Th1/Th2 balance seems to be regulated mainly by two distinct DC subsets, DEC-205(+) DCs having the capacity to establish Th1 polarization and 33D1(+) DCs to induce Th2 dominance. Pregnancy is established and maintained by maternal hormones, such as progesterone and estrogen, and the balance of DC subtypes was affected mainly by progesterone, which induced a dose-dependent reduction of the DEC-205/33D1 ratio together with/without a stable amount of estrogen. The DEC-205/33D1 ratio decreased gradually with the progress of pregnancy and rapid augmentation of the ratio was seen around delivery in vivo. Here, we demonstrate that depletion of 33D1(+) DCs during the perinatal period caused substantial fetal loss probably mediated through Th1 up-regulation via transient IL-12 secretion, and pre-administration of progesterone could rescue the fetal loss. Similar miscarriages were also observed when pregnant mice were intraperitoneally (i.p.) injected twice with IL-12 on Gd 9.5 and 10.5. Moreover, prior inoculation of progesterone suppressed the enhanced serum IL-12 production in mice treated with 33D1 antibody, indicating that progesterone might inhibit temporal IL-12 secretion around Gd 10.5 and miscarriage was avoided. These findings suggest the importance of balancing DC subsets during pregnancy and reveal that we can avoid miscarriage by manipulating the activity of the DC subpopulation of pregnant individuals with maternal hormones.


Journal of Immunology | 2008

Suppression of an Already Established Tumor Growing through Activated Mucosal CTLs Induced by Oral Administration of Tumor Antigen with Cholera Toxin

Ayako Wakabayashi; Yohko Nakagawa; Masumi Shimizu; Keiichi Moriya; Yasuhiro Nishiyama; Hidemi Takahashi

Priming of CTLs at mucosal sites, where various tumors are originated, seems critical for controlling tumors. In the present study, the effect of the oral administration of OVA plus adjuvant cholera toxin (CT) on the induction of Ag-specific mucosal CTLs as well as their effect on tumor regression was investigated. Although OVA-specific TCRs expressing lymphocytes requiring in vitro restimulation to gain specific cytotoxicity could be detected by OVA peptide-bearing tetramers in both freshly isolated intraepithelial lymphocytes and spleen cells when OVA was orally administered CT, those showing direct cytotoxic activity without requiring in vitro restimulation were dominantly observed in intraepithelial lymphocytes. The magnitude of such direct cytotoxicity at mucosal sites was drastically enhanced after the second oral administration of OVA with intact whole CT but not with its subcomponent, an A subunit (CTA) or a B subunit (CTB). When OVA plus CT were orally administrated to C57BL/6 mice bearing OVA-expressing syngeneic tumor cells, E.G7-OVA, in either gastric tissue or the dermis, tumor growth was significantly suppressed after the second oral treatment; however, s.c. or i.p. injection of OVA plus CT did not show any remarkable suppression. Those mucosal OVA-specific CTLs having direct cytotoxicity expressed CD8αβ but not CD8αα, suggesting that they originated from thymus-educated cells. Moreover, the infiltration of such OVA-specific CD8+ CTLs was observed in suppressed tumor tissues. These results indicate that the growth of ongoing tumor cells can be suppressed by activated CD8αβ CTLs with tumor-specific cytotoxicity via an orally administered tumor Ag with a suitable mucosal adjuvant.


Immunology | 2006

Importance of gastrointestinal ingestion and macromolecular antigens in the vein for oral tolerance induction

Ayako Wakabayashi; Yoshihiro Kumagai; Eiji Watari; Masumi Shimizu; Masanori Utsuyama; Katsuiku Hirokawa; Hidemi Takahashi

Oral administration of a certain dose of antigen can generally induce immunological tolerance against the same antigen. In this study, we showed the temporal appearance of ovalbumin (OVA) antigens in both portal and peripheral blood of mice after the oral administration of OVA. Furthermore, we detected 45 000 MW OVA in mouse serum 30 min after the oral administration of OVA. Based on this observation, we examined whether the injection of intact OVA into the portal or peripheral vein induces immunological tolerance against OVA. We found that the intravenous injection of intact OVA did not induce immunological tolerance but rather enhanced OVA‐specific antibody production in some subclasses, suggesting that OVA antigens via the gastrointestinal tract but not intact OVA may contribute to establish immunological tolerance against OVA. Therefore, we examined the effects of digesting intact OVA in the gastrointestinal tract on the induction of oral tolerance. When mice were orally administered or injected into various gastrointestinal organs, such as the stomach, duodenum, ileum, or colon and boosted with intact OVA, OVA‐specific antibody production and delayed‐type hypersensitivity (DTH) response were significantly enhanced in mice injected into the ileum or colon, compared with orally administered mice. These results suggest that although macromolecular OVA antigens are detected after oral administration of OVA in tolerant‐mouse serum, injection of intact OVA cannot contribute to tolerance induction. Therefore, some modification of macromolecular OVA in the gastrointestinal tract and ingestion may be essential for oral tolerance induction.


International Archives of Allergy and Immunology | 2015

Effects of Dendritic Cell Subset Manipulation on Airway Allergy in a Mouse Model.

Ryosuke Murakami; Yohko Nakagawa; Masumi Shimizu; Ayako Wakabayashi; Yasuyuki Negishi; Takachika Hiroi; Kimihiro Okubo; Hidemi Takahashi

Background: Two major distinct subsets of dendritic cells (DCs) are arranged to regulate immune responses: DEC-205+ DCs drive Th1 polarization and 33D1+ DCs establish Th2 dominancy. Th1 polarization can be achieved either by depletion of 33D1+ DCs with a 33D1-specific monoclonal antibody (mAb) or by activation of DEC-205+ DCs via intraperitoneal injection of α-galactosylceramide (α-GalCer). We studied the effect of 33D1+ DC depletion or DEC-205+ DC activation in vivo using an established mouse model of allergic rhinitis (AR). Methods: Mice were injected intraperitoneally with OVA plus alum and challenged 4 times with daily intranasal administration of OVA. Immediately after the last challenge, allergic symptoms such as sneezing and nasal rubbing as well as the number of cells in the bronchoalveolar lavage fluid (BALF) and nasal lavage fluid (NALF) were counted. The levels of serum OVA-specific IgG1, IgG2a, and IgE were also determined by ELISA. Results: The allergic symptom scores were significantly decreased in 33D1+ DC-depleted or DEC-205+ DC-activated AR mice. The levels of OVA-specific IgG1, IgG2a, and IgE, and the number of NALF cells, but not BALF cells, were reduced in 33D1+ DC-depleted but not in DEC-205+ DC-activated AR mice. Moreover, the activated DEC-205+ DCs suppressed histamine release from IgE-sensitized mast cells, probably through IL-12 secretion. Conclusions: The manipulation of innate DC subsets may provide a new therapeutic strategy for controlling various allergic diseases by reducing histamine release from IgE-sensitized mast cells by driving the immune response towards Th1 dominancy via activation of DEC-205+ DCs in vivo.


Cell Death and Disease | 2018

HMGB1 released from intestinal epithelia damaged by cholera toxin adjuvant contributes to activation of mucosal dendritic cells and induction of intestinal cytotoxic T lymphocytes and IgA

Ayako Wakabayashi; Masumi Shimizu; Eiji Shinya; Hidemi Takahashi

Cholera toxin (CT) is a potent mucosal adjuvant and oral administration of ovalbumin (OVA) antigens plus CT induces OVA-specific CD8+ cytotoxic T lymphocytes (CTLs) and IgA production in intestinal mucosa. However, the mechanisms of induction of these immune responses remain unknown. Intestinal OVA-specific CD8+ CTLs were not induced by oral administration of the CT active (CTA) or CT binding (CTB) subunit as an adjuvant and CD11c+ DCs were involved in cross-priming of intestinal CTLs. CD8+CD103+CD11c+CD11b−DCs and DCIR2+CD103+CD11c+CD11b+ DCs were distributed in the intestinal lamina propria and mesenteric lymph nodes, both DC subsets expressed DEC-205, and the expression of co-stimulatory molecules such as CD80 and CD86 was enhanced in both DC subsets after oral administration of intact CT but not the CTA or CTB subunit. Intestinal DCs activated by the oral administration of OVA plus CT cross-presented OVA antigens and DCs that captured OVA antigen through DEC-205, but not DCIR2, could cross-present antigen. We found that oral administration of intact CT, but not the CTA or CTB subunit, enhanced cell death, cytoplasmic expression of high-mobility group box 1 protein (HMGB1) in epithelial cell adhesion molecule (EpCAM)+CD45− intestinal epithelial cells (IECs), and HMGB1 levels in fecal extracts. HMGB1 dose-dependently enhanced the expression of CD80 and CD86 on DCs in vitro, and intravenous or oral administration of glycyrrhizin, an HMGB1 inhibitor, significantly suppressed activation of mucosal DCs and induction of intestinal OVA-specific CTLs and IgA by oral CT administration. These results showed that oral administration of intact CT triggers epithelial cell death in the gut and the release of HMGB1 from damaged IECs, and that the released HMGB1 may mediate activation of mucosal DCs and induction of CTLs and IgA in the intestine.


International Archives of Allergy and Immunology | 2015

Contents Vol. 168, 2015

Maria Tzardi; Kimihiro Okubo; Ryosuke Murakami; Yohko Nakagawa; Masumi Shimizu; Ayako Wakabayashi; Yasuyuki Negishi; Takachika Hiroi; Hidemi Takahashi; Venu Gangur; Sebastian Heck; Dd Le; Robert Bals; Juliane Nguyen; Quoc Thai Dinh; Claas Gloistein; Anatoli Astvatsatourov; Silke Allekotte; Ralph Mösges; Natalia Blanca-López; Gabriela Canto; Per Stahl-Skov; Miguel Blanca; Maria J. Torres; Fernando Pineda; Adriana Ariza; Cristobalina Mayorga; Francisca Arribas; Rosario González-Mendiola; Galicia Davila

Founded 1950 by D. Harley, P. Kallós, W. Löffler and F.W. Wittich Continued by E.A. Brown (1952–1954), W. Kaufman (1955–1967), F. Hahn (1961–1972), H.C. Goodman (1963–1975), R.R.A. Coombs (1955–1984), Z. Trnka (1972–1987), P. Kallós (1950–1988), G.B. West (1959–1991), J. Bienenstock (1990–1991), L.Å. Hanson (1981–1991), K. Kano (1982–1991), F. Milgrom (1965–1991), K. Rother (1982–1991), G. Wick (1991–1997), B. Grubeck-Loebenstein (1991–1997), D. Kraft (1998–2002) Official Journal of the


Immunology and Cell Biology | 2013

Inactivation of tumor-specific CD8+ CTLs by tumor-infiltrating tolerogenic dendritic cells

Hirotomo Harimoto; Masumi Shimizu; Yohko Nakagawa; Katsuhisa Nakatsuka; Ayako Wakabayashi; Choitsu Sakamoto; Hidemi Takahashi


Cancer Immunology, Immunotherapy | 2010

Induction of tumor-specific acquired immunity against already established tumors by selective stimulation of innate DEC-205+ dendritic cells

Keiichi Moriya; Ayako Wakabayashi; Masumi Shimizu; Hideto Tamura; Kazuo Dan; Hidemi Takahashi


Journal of Nutrition Health & Aging | 2006

Induction of immunological tolerance by oral, but not intravenous and intraportal, administration of ovalbumin and the difference between young and old mice.

Ayako Wakabayashi; Masanori Utsuyama; Hosoda T; Kazuto Sato; Hidemi Takahashi; Katsuiku Hirokawa


Biochemical and Biophysical Research Communications | 2004

Resistance to viral infection by intraepithelial lymphocytes in HIV-1 P18-I10-specific T-cell receptor transgenic mice

Hideki Kuribayashi; Ayako Wakabayashi; Masumi Shimizu; Hiroshi Kaneko; Yoshihiko Norose; Yohko Nakagawa; Jian Wang; Yoshihiro Kumagai; David H. Margulies; Hidemi Takahashi

Collaboration


Dive into the Ayako Wakabayashi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katsuiku Hirokawa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masanori Utsuyama

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge