Network


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

Hotspot


Dive into the research topics where Hideyuki Iwai is active.

Publication


Featured researches published by Hideyuki Iwai.


Journal of Immunology | 2002

Expression of Programmed Death 1 Ligands by Murine T Cells and APC

Tomohide Yamazaki; Hisaya Akiba; Hideyuki Iwai; Hironori Matsuda; Mami Aoki; Yuka Tanno; Tahiro Shin; Haruo Tsuchiya; Drew M. Pardoll; Ko Okumura; Miyuki Azuma; Hideo Yagita

Programmed death 1 (PD-1) is a new member of the CD28/CTLA-4 family, which has been implicated in the maintenance of peripheral tolerance. Two ligands for PD-1, namely, B7-H1 (PD-L1) and B7-DC (PD-L2), have recently been identified as new members of the B7 family but their expression at the protein level remains largely unknown. To characterize the expression of B7-H1 and B7-DC, we newly generated an anti-mouse B7-H1 mAb (MIH6) and an anti-mouse B7-DC mAb (TY25). MIH6 and TY25 immunoprecipitated a single molecule of 43 and 42 kDa from the lysate of B7-H1 and B7-DC transfectants, respectively. Flow cytometric analysis revealed that B7-H1 was broadly expressed on the surface of mouse tumor cell lines while the expression of B7-DC was rather restricted. PD-1 was expressed on anti-CD3-stimulated T cells and anti-IgM plus anti-CD40-stimulated B cells at high levels but was undetectable on activated macrophages or DCs. B7-H1 was constitutively expressed on freshly isolated splenic T cells, B cells, macrophages, and dendritic cells (DCs), and up-regulated on T cells by anti-CD3 stimulation on macrophages by LPS, IFN-γ, GM-CSF, or IL-4, and on DCs by IFN-γ, GM-CSF, or IL-4. In contrast, B7-DC expression was only inducible on macrophages and DCs upon stimulation with IFN-γ, GM-CSF, or IL-4. The inducible expression of PD-1 ligands on both T cells and APCs may suggest new paradigms of PD-1-mediated immune regulation.


Journal of Experimental Medicine | 2003

The Programmed Death-1 (PD-1) Pathway Regulates Autoimmune Diabetes in Nonobese Diabetic (NOD) Mice

Mohammed Javeed Ansari; Alan D. Salama; Tanuja Chitnis; R. Neal Smith; Hideo Yagita; Hisaya Akiba; Tomohide Yamazaki; Miyuki Azuma; Hideyuki Iwai; Samia J. Khoury; Hugh Auchincloss; Mohamed H. Sayegh

Programmed death-1 (PD-1) receptor, an inhibitory costimulatory molecule found on activated T cells, has been demonstrated to play a role in the regulation of immune responses and peripheral tolerance. We investigated the role of this pathway in the development of autoimmune diabetes. PD-1 or PD-L1 but not PD-L2 blockade rapidly precipitated diabetes in prediabetic female nonobese diabetic (NOD) mice regardless of age (from 1 to 10-wk-old), although it was most pronounced in the older mice. By contrast, cytotoxic T lymphocyte–associated antigen 4 (CTLA-4) blockade induced disease only in neonates. Male NOD mice also developed diabetes after PD-1–PD-L1 pathway blockade, but NOR mice, congenic to NOD but resistant to the development of diabetes, did not. Insulitis scores were significantly higher and frequency of interferon γ–producing GAD-reactive splenocytes was increased after PD-1–PD-L1 pathway blockade compared with controls. Interestingly, PD-L1 but not PD-L2 was found to be expressed on inflamed islets of NOD mice. These data demonstrate a central role for PD-1–PD-L1 interaction in the regulation of induction and progression of autoimmune diabetes in the NOD mouse and provide the rationale to develop new therapies to target this costimulatory pathway in this disease.


Journal of Immunology | 2003

Blockade of B7-H1 Suppresses the Development of Chronic Intestinal Inflammation

Takanori Kanai; Teruji Totsuka; Koji Uraushihara; Shin Makita; Tetsuya Nakamura; Kazutaka Koganei; Tsuneo Fukushima; Hisaya Akiba; Hideo Yagita; Ko Okumura; Utako Machida; Hideyuki Iwai; Miyuki Azuma; Lieping Chen; Mamoru Watanabe

A newly identified costimulatory molecule, programmed death-1 (PD-1), provides a negative signal that is essential for immune homeostasis. However, it has been suggested that its ligands, B7-H1 (PD-L1) and B7-dendritic cells (B7-DC; PD-L2), could also costimulate T cell proliferation and cytokine secretion. Here we demonstrate the involvement of PD-1/B7-H1 and B7-DC interaction in the development of colitis. We first examined the expression profiles of PD-1 and its ligands in both human inflammatory bowel disease and a murine chronic colitis model induced by adoptive transfer of CD4+CD45RBhigh T cells to SCID mice. Second, we assessed the therapeutic potential of neutralizing anti-B7-H1 and/or B7-DC mAbs using this colitis model. We found significantly increased expression of PD-1 on T cells and of B7-H1 on T, B, and macrophage/DCs in inflamed colon from both inflammatory bowel disease patients and colitic mice. Unexpectedly, the administration of anti-B7-H1, but not anti-B7-DC, mAb after transfer of CD4+CD45RBhigh T cells suppressed wasting disease with colitis, abrogated leukocyte infiltration, and reduced the production of IFN-γ, IL-2, and TNF-α, but not IL-4 or IL-10, by lamina propria CD4+ T cells. These data suggest that the interaction of PD-1/B7-H1, but not PD-1/B7-DC, might be involved in intestinal mucosal inflammation and also show a possible role of interaction between B7-H1 and an as yet unidentified receptor for B7-H1 in inducing T cell activation.


Journal of Immunology | 2002

Amelioration of Collagen-Induced Arthritis by Blockade of Inducible Costimulator-B7 Homologous Protein Costimulation

Hideyuki Iwai; Yuko Kozono; Sachiko Hirose; Hisaya Akiba; Hideo Yagita; Ko Okumura; Hitoshi Kohsaka; Nobuyuki Miyasaka; Miyuki Azuma

B7 homologous protein (B7h)/B7-related protein 1 (B7RP-1) is a new member of the B7 family of costimulatory molecules that specifically interacts with inducible costimulator (ICOS) expressed on activated T cells. Collagen type II (CII)-induced arthritis (CIA) is an experimental model of arthritis that has been used to dissect the pathogenesis of human rheumatoid arthritis. In this study, we have investigated the effect of neutralizing anti-B7h mAb on the development and disease progression of CIA. Administration of anti-B7h mAb significantly ameliorated the disease as assessed by clinical arthritis score and histology in the joints, and a beneficial effect was also obtained by a delayed treatment after the onset of disease. Expression of ICOS and B7h was observed in the inflamed synovial tissue as well as in the draining lymph nodes (LNs) and expansion of ICOS+ T cells in the LN was reduced by the anti-B7h mAb treatment. Expression of mRNA for proinflammatory cytokines such as TNF-α, IL-1β, and IL-6 in the joints was inhibited by the treatment. Proliferative responses and production of IFN-γ and IL-10 upon restimulation with CII in vitro were significantly inhibited in LN cells from the anti-B7h mAb-treated mice. Serum anti-CII IgG1, IgG2a, and IgG2b levels were also reduced. Our present results showed a beneficial effect of the B7h blockade on CIA through anti-inflammatory actions and inhibition of both Th1- and Th2-mediated immune responses, suggesting that the ICOS-B7h interaction plays an important role in the pathogenesis of CIA and thus the blockade of this pathway may be beneficial for the treatment of human rheumatoid arthritis.


Biochemical and Biophysical Research Communications | 2003

Differential binding properties of B7-H1 and B7-DC to programmed death-1.

Pornpan Youngnak; Yuko Kozono; Haruo Kozono; Hideyuki Iwai; Noriko Otsuki; Hisayo Jin; Ken Omura; Hideo Yagita; Drew M. Pardoll; Lieping Chen; Miyuki Azuma

Programmed death-1 (PD-1) is a negative regulatory receptor expressed on activated T and B cells. Two ligands for PD-1, B7-H1 (PD-L1) and B7-DC (PD-L2), have been identified, but their binding properties have not been characterized yet. In this study, we generated soluble Ig fusion proteins of these molecules and examined the kinetics and relative affinities of the interactions between B7-H1 or B7-DC and PD-1 by flow cytometry and surface plasmon resonance. The interaction of B7-DC/PD-1 exhibited a 2-6-fold higher affinity and had different association/dissociation kinetics compared with the interaction of B7-H1/PD-1. Our results suggest that the differential binding properties of B7-H1 and B7-DC may be responsible for differential contributions of these two PD-1 ligands to immune responses.


Journal of Immunology | 2003

Involvement of inducible costimulator-B7 homologous protein costimulatory pathway in murine lupus nephritis.

Hideyuki Iwai; Masaaki Abe; Sachiko Hirose; Fumihiko Tsushima; Katsunari Tezuka; Hisaya Akiba; Hideo Yagita; Ko Okumura; Hitoshi Kohsaka; Nobuyuki Miyasaka; Miyuki Azuma

Inducible costimulator (ICOS)-B7 homologous protein (B7h) is a new member of the CD28-B7 family of costimulatory molecules that regulates T cell-dependent humoral immune responses. In this study, we examined the involvement of this costimulatory pathway in the development and progression of lupus in NZB/W F1 mice. Expression of ICOS on T cells was enhanced with disease progression, whereas B7h expression on B cells was down-regulated. Administration of anti-B7h mAb before the onset of renal disease significantly delayed the onset of proteinuria and prolonged survival. Blockade of B7h effectively inhibited all subclasses of IgG autoantibody production and accumulation of both Th1 and Th2 cells. Hypercellularity and deposition of IgG and C3 in glomeruli were significantly reduced. B7h blockade after the onset of proteinuria prevented the disease progression and improved the renal pathology. Our results demonstrated the involvement of the ICOS-B7h costimulatory pathway in the pathogenesis of lupus nephritis, and the blockade of this pathway may be beneficial for the treatment of human systemic lupus erythematosus.


European Journal of Immunology | 2003

Preferential contribution of B7‐H1 to programmed death‐1‐mediated regulation of hapten‐specific allergic inflammatory responses

Fumihiko Tsushima; Hideyuki Iwai; Noriko Otsuki; Masaaki Abe; Sachiko Hirose; Tomohide Yamazaki; Hisaya Akiba; Hideo Yagita; Yuzo Takahashi; Ken Omura; Ko Okumura; Miyuki Azuma

Programmed death‐1 (PD‐1) and its ligands, B7‐H1/PD‐L1 and B7‐DC/PD‐L2, are new CD28‐B7 family members that may be involved in the regulation of immune responses. We examined the roles of these molecules in mouse hapten‐induced contact hypersensitivity (CH). Administration of anti‐PD‐1 mAb at sensitization significantly enhanced and prolonged ear swelling. Treatment with anti‐B7‐H1 mAb, but not anti‐B7‐DC mAb, also enhanced CH reactions. The anti‐PD‐1 mAb treatment at sensitization significantly increased the T cell number of draining lymph nodes (DLN). B7‐H1 was induced on activated T cells and antigen‐presenting cells (APC) in the skin and the DLN, whereas B7‐DC expression was restricted to dendritic cells (DC) in the dermis and the DLN. A particular subset of DC, B7‐H1+B7‐DC–CD86low, was found in sensitized DLN. The blockade of B7‐H1, but not B7‐DC, dramatically enhanced the initial T cell proliferative responses against hapten‐pulsed DLN APC, suggesting the preferential contribution of B7‐H1 to the T cell‐APC interaction. Our results demonstrate the regulatory role of PD‐1 and the differential roles of B7‐H1 and B7‐DC in hapten‐induced immune responses. The PD‐1‐B7‐H1 pathway may play a unique role in regulating inflammatory responses.


Biochemical and Biophysical Research Communications | 2008

GITR ligand-costimulation activates effector and regulatory functions of CD4+ T cells

Hanna Igarashi; Yujia Cao; Hideyuki Iwai; Jinhua Piao; Yosuke Kamimura; Masaaki Hashiguchi; Teruo Amagasa; Miyuki Azuma

Engagement of glucocorticoid-induced TNFR-related protein (GITR) enables the costimulation of both CD25(-)CD4(+) effector (Teff) and CD25(+)CD4(+) regulatory (Treg) cells; however, the effects of GITR-costimulation on Treg function remain controversial. In this study, we examined the effects of GITR ligand (GITRL) binding on the respective functions of CD4(+) T cells. GITRL-P815 transfectants efficiently augmented anti-CD3-induced proliferation and cytokine production by Teff cells. Proliferation and IL-10 production in Treg were also enhanced by GITRL transfectants when exogenous IL-2 and stronger CD3 stimulation was provided. Concomitant GITRL-costimulation of Teff and Treg converted the anergic state of Treg into a proliferating state, maintaining and augmenting their function. Thus, GITRL-costimulation augments both effector and regulatory functions of CD4(+) T cells. Our results suggest that highly activated and increased ratios of Treg reverse the immune-enhancing effects of GITRL-costimulation in Teff, which may be problematic for therapeutic applications using strong GITR agonists.


Immunology | 2009

Enhancement of T-cell-mediated anti-tumour immunity via the ectopically expressed glucocorticoid-induced tumour necrosis factor receptor-related receptor ligand (GITRL) on tumours

Jinhua Piao; Yosuke Kamimura; Hideyuki Iwai; Yujia Cao; Keisuke Kikuchi; Masaaki Hashiguchi; Taro Masunaga; Hongsi Jiang M.D.; Kouichi Tamura; Shimon Sakaguchi; Miyuki Azuma

Glucocorticoid‐induced tumour necrosis factor receptor‐related receptor (GITR) costimulates functions of both effector and regulatory T cells. The administration of agonistic anti‐GITR monoclonal antibodies efficiently enhances various T‐cell‐mediated immune responses; however, it is unknown to what extent the ligand of GITR (GITRL) contributes to T‐cell responses. We investigated the involvement of endogenously expressed GITRL on dendritic cells and ectopically expressed GITRL on tumours in T‐cell‐mediated immunity. Expression of GITRL on dendritic cells in secondary lymphoid organs was limited, and treatment with anti‐GITRL monoclonal antibodies did not substantially affect T‐cell‐mediated immunity to alloantigens, a specific protein antigen (ovalbumin), or tumour antigens. The introduction of GITRL promoted anti‐tumour immunity in four tumour models. Tumour‐associated GITRL greatly augmented the effector function of CD8+ T cells and enhanced the contribution of CD8+ T cells. These events reduced the crucial contribution of CD25+ CD4+ regulatory T cells, which were found to inhibit immunity against tumours lacking GITRL. Peritumoral injection of GITRL tumour vaccine efficiently inhibited the growth of established tumours. Our results suggest that the ectopic expression of GITRL in tumour cells enhances anti‐tumour immunity at peripheral tumour sites. Consequently, the combined use of a GITRL tumour vaccine with methods aimed at enhancing the activation of host antigen‐presenting cells in secondary lymphoid tissues may be a promising strategy for tumour immunotherapy.


Journal of Immunology | 2009

The Transmembrane E3 Ligase GRAIL Ubiquitinates and Degrades CD83 on CD4 T Cells

Leon Su; Hideyuki Iwai; Jack Lin; C. Garrison Fathman

Ubiquitination of eukaryotic proteins regulates a broad range of cellular processes, including T cell activation and tolerance. We have previously demonstrated that GRAIL (gene related to anergy in lymphocytes), a transmembrane RING finger ubiquitin E3 ligase, initially described as induced during the induction of CD4 T cell anergy, is also expressed in resting CD4 T cells. In this study, we show that GRAIL can down-modulate the expression of CD83 (previously described as a cell surface marker for mature dendritic cells) on CD4 T cells. GRAIL-mediated down-modulation of CD83 is dependent on an intact GRAIL extracellular protease-associated domain and an enzymatically active cytosolic RING domain, and proceeds via the ubiquitin-dependent 26S proteosome pathway. Ubiquitin modification of lysine residues K168 and K183, but not K192, in the cytoplasmic domain of CD83 was shown to be necessary for GRAIL-mediated degradation of CD83. Reduced CD83 surface expression levels were seen both on anergized CD4 T cells and following GRAIL expression by retroviral transduction, whereas GRAIL knock-down by RNA interference in CD4 T cells resulted in elevated CD83 levels. Furthermore, CD83 expression on CD4 T cells contributes to T cell activation as a costimulatory molecule. This study supports the novel mechanism of ubiquitination by GRAIL, identifies CD83 as a substrate of GRAIL, and ascribes a role for CD83 in CD4 T cell activation.

Collaboration


Dive into the Hideyuki Iwai's collaboration.

Top Co-Authors

Avatar

Miyuki Azuma

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Nobuyuki Miyasaka

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hitoshi Kohsaka

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fumihiko Tsushima

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Jinhua Piao

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Ken Omura

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Noriko Otsuki

Tokyo Medical and Dental University

View shared research outputs
Researchain Logo
Decentralizing Knowledge