Network


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

Hotspot


Dive into the research topics where Tomohide Yamazaki is active.

Publication


Featured researches published by Tomohide Yamazaki.


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.


Immunity | 2009

T helper 17 cells promote cytotoxic T cell activation in tumor immunity.

Pawel Muranski; Yeonseok Chung; Xuexian O. Yang; Tomohide Yamazaki; Sijie Lu; Patrick Hwu; Nicholas P. Restifo; Willem W. Overwijk; Chen Dong

Although T helper 17 (Th17) cells have been found in tumor tissues, their function in cancer immunity is unclear. We found that interleukin-17A (IL-17A)-deficient mice were more susceptible to developing lung melanoma. Conversely, adoptive T cell therapy with tumor-specific Th17 cells prevented tumor development. Importantly, the Th17 cells retained their cytokine signature and exhibited stronger therapeutic efficacy than Th1 cells. Unexpectedly, therapy using Th17 cells elicited a remarkable activation of tumor-specific CD8(+) T cells, which were necessary for the antitumor effect. Th17 cells promoted dendritic cell recruitment into the tumor tissues and in draining lymph nodes increased CD8 alpha(+) dendritic cells containing tumor material. Moreover, Th17 cells promoted CCL20 chemokine production by tumor tissues, and tumor-bearing CCR6-deficient mice did not respond to Th17 cell therapy. Thus, Th17 cells elicited a protective inflammation that promotes the activation of tumor-specific CD8(+) T cells. These findings have important implications in antitumor immunotherapies.


Journal of Immunology | 2008

CCR6 Regulates the Migration of Inflammatory and Regulatory T Cells

Tomohide Yamazaki; Xuexian O. Yang; Yeonseok Chung; Atsushi Fukunaga; Roza Nurieva; Bhanu P. Pappu; Hong Soon Kang; Li Ma; Athanasia D. Panopoulos; Suzanne Craig; Stephanie S. Watowich; Anton M. Jetten; Qiang Tian; Chen Dong

Th17 and regulatory T (Treg) cells play opposite roles in autoimmune diseases. However, the mechanisms underlying their proper migration to inflammatory tissues are unclear. In this study, we report that these two T cell subsets both express CCR6. CCR6 expression in Th17 cells is regulated by TGF-β and requires two nuclear receptors, RORα and RORγ. Th17 cells also express the CCR6 ligand CCL20, which is induced synergistically by TGF-β and IL-6, which requires STAT3, RORγ and IL-21. Th17 cells, by producing CCL20, promote migration of Th17 and Treg cells in vitro in a CCR6-dependent manner. Lack of CCR6 in Th17 cells reduces the severity of experimental autoimmune encephalomyelitis and Th17 and Treg recruitment into inflammatory tissues. Similarly, CCR6 on Treg cells is also important for their recruitment into inflammatory tissues. Our data indicate an important role of CCR6 in Treg and Th17 cell migration.


Journal of Immunology | 2005

The Role of ICOS in the CXCR5+ Follicular B Helper T Cell Maintenance In Vivo

Hisaya Akiba; Kazuyoshi Takeda; Yuko Kojima; Yoshihiko Usui; Norihiro Harada; Tomohide Yamazaki; Juan Ma; Katsunari Tezuka; Hideo Yagita; Ko Okumura

ICOS is a new member of the CD28 family of costimulatory molecules that is expressed on activated T cells. Its ligand B7RP-1 is constitutively expressed on B cells. Although the blockade of ICOS/B7RP-1 interaction inhibits T cell-dependent Ab production and germinal center formation, the mechanism remains unclear. We examined the contribution of ICOS/B7RP-1 to the generation of CXCR5+ follicular B helper T (TFH) cells in vivo, which preferentially migrate to the B cell zone where they provide cognate help to B cells. In the spleen, anti-B7RP-1 mAb-treated or ICOS-deficient mice showed substantially impaired development of CXCR5+ TFH cells and peanut agglutinin+ germinal center B cells in response to primary or secondary immunization with SRBC. Expression of CXCR5 on CD4+ T cells was associated with ICOS expression. Adoptive transfer experiments showed that the development of CXCR5+ TFH cells was enhanced by interaction with B cells, which was abrogated by anti-B7RP-1 mAb treatment. The development of CXCR5+ TFH cells in the lymph nodes was also inhibited by the anti-B7RP-1 mAb treatment. These results indicated that the ICOS/B7RP-1 interaction plays an essential role in the development of CXCR5+ TFH cells in vivo.


European Journal of Immunology | 2009

B7-H3 is a potent inhibitor of human T-cell activation: No evidence for B7-H3 and TREML2 interaction

Judith Leitner; Christoph Klauser; Winfried F. Pickl; Johannes Stöckl; Otto Majdic; Anaïs F. Bardet; David P. Kreil; Chen Dong; Tomohide Yamazaki; Gerhard J. Zlabinger; Katharina Pfistershammer; Peter Steinberger

B7‐H3 belongs to the B7 superfamily, a group of molecules that costimulate or down‐modulate T‐cell responses. Although it was shown that B7‐H3 could inhibit T‐cell responses, several studies – most of them performed in murine systems – found B7‐H3 to act in a costimulatory manner. In this study, we have specifically addressed a potential functional dualism of human B7‐H3 by assessing the effect of this molecule under varying experimental conditions as well as on different T‐cell subsets. We show that B7‐H3 does not costimulate human T cells. In the presence of strong activating signals, B7‐H3 potently and consistently down‐modulated human T‐cell responses. This inhibitory effect was evident when analysing proliferation and cytokine production and affected naïve as well as pre‐activated T cells. Furthermore, we demonstrate that B7‐H3–T‐cell interaction is characterised by an early suppression of IL‐2 and that T‐cell inhibition can be reverted by exogenous IL‐2. Since the triggering receptor expressed on myeloid cells like transcript 2 (TREML2/TLT‐2) has been recently described as costimulatory receptor of murine B7‐H3 we have extensively analysed interaction of human B7‐H3 with TREML2/TLT‐2. In these experiments we found no evidence for such an interaction. Furthermore, our data do not point to a role for murine TREML2 as a receptor for murine B7‐H3.


Journal of Immunology | 2005

Blockade of B7-H1 on Macrophages Suppresses CD4+ T Cell Proliferation by Augmenting IFN-γ-Induced Nitric Oxide Production

Tomohide Yamazaki; Hisaya Akiba; Akemi Koyanagi; Miyuki Azuma; Hideo Yagita; Ko Okumura

PD-1 is an immunoinhibitory receptor that belongs to the CD28/CTLA-4 family. B7-H1 (PD-L1) and B7-DC (PD-L2), which belong to the B7 family, have been identified as ligands for PD-1. Paradoxically, it has been reported that both B7-H1 and B7-DC costimulate or inhibit T cell proliferation and cytokine production. To determine the role of B7-H1 and B7-DC in T cell-APC interactions, we examined the contribution of B7-H1 and B7-DC to CD4+ T cell activation by B cells, dendritic cells, and macrophages using anti-B7-H1, anti-B7-DC, and anti-PD-1 blocking mAbs. Anti-B7-H1 mAb and its Fab markedly inhibited the proliferation of anti-CD3-stimulated naive CD4+ T cells, but enhanced IL-2 and IFN-γ production in the presence of macrophages. The inhibition of T cell proliferation by anti-B7-H1 mAb was abolished by neutralizing anti-IFN-γ mAb. Coculture of CD4+ T cells and macrophages from IFN-γ-deficient or wild-type mice showed that CD4+ T cell-derived IFN-γ was mainly responsible for the inhibition of CD4+ T cell proliferation. Anti-B7-H1 mAb induced IFN-γ-mediated production of NO by macrophages, and inducible NO synthase inhibitors abrogated the inhibition of CD4+ T cell proliferation by anti-B7-H1 mAb. These results indicated that the inhibition of T cell proliferation by anti-B7-H1 mAb was due to enhanced IFN-γ production, which augmented NO production by macrophages, suggesting a critical role for B7-H1 on macrophages in regulating IFN-γ production by naive CD4+ T cells and, hence, NO production by macrophages.


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.


Blood | 2009

Abundant c-Fas–associated death domain–like interleukin-1–converting enzyme inhibitory protein expression determines resistance of T helper 17 cells to activation-induced cell death

Yu Yu; Cristina Iclozan; Tomohide Yamazaki; Xuexian O. Yang; Claudio Anasetti; Chen Dong; Xue-Zhong Yu

Activation-induced cell death (AICD) plays an important role in peripheral T-cell tolerance. AICD in CD4 T helper (Th) cells, including Th1 and Th2 effectors, has been extensively studied. Recently, interleukin-17-producing CD4(+) T cells (Th17 cells) have been identified as a unique Th subset, but their susceptibility to AICD and the underlying molecular mechanisms have not been defined. In this study, we found that Th17 cells were significantly less susceptible to AICD than Th1 cells, and Th17 cell resistance to AICD is due to the high levels of c-Fas-associated death domain-like interleukin-1-converting enzyme inhibitory protein preventing Fas-mediated apoptosis. The resistance of Th17 cells to AICD reveals a novel mechanism to explain the high pathogenicity of Th17 cells in autoimmune diseases, and may also provide a rationale to generate tumor-specific Th17 cells for adoptive immunotherapy.


Journal of Immunology | 2010

A Butyrophilin Family Member Critically Inhibits T Cell Activation

Tomohide Yamazaki; Iñigo Goya; Daniel Graf; Suzanne Craig; Chen Dong

The costimulatory molecules in the B7-CD28 families are important in the regulation of T cell activation and tolerance. The butyrophilin family of proteins shares sequence and structure homology with B7 family molecules; however, the function of the butyrophilin family in the immune system has not been defined. In this study, we performed an analysis on multiple butyrophilin molecules and found that butyrophilin-like (BTNL)1 molecule functions to dampen T cell activation. BTNL1 mRNA was broadly expressed, but its protein was only found in APCs and not T cells. The putative receptor for BTNL1 was found on activated T cells and APCs. Also, recombinant BTNL1 molecule inhibited T cell proliferation by arresting cell cycle progression. The administration of neutralizing Abs against BTNL1 provoked enhanced T cell activation and exacerbated disease in autoimmune and asthma mouse models. Therefore, BTNL1 is a critical inhibitory molecule for T cell activation and immune diseases.

Collaboration


Dive into the Tomohide Yamazaki's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yeonseok Chung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Miyuki Azuma

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Xuexian O. Yang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hideyuki Iwai

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Yoshihiko Usui

Tokyo Medical University

View shared research outputs
Top Co-Authors

Avatar

Gustavo J. Martinez

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge