Network


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

Hotspot


Dive into the research topics where Steven Kiyoshi Yoshinaga is active.

Publication


Featured researches published by Steven Kiyoshi Yoshinaga.


Nature | 1999

T-cell co-stimulation through B7RP-1 and ICOS.

Steven Kiyoshi Yoshinaga; John S. Whoriskey; Sanjay D. Khare; Ulla Sarmiento; Jane Guo; Tom Horan; Grace Shih; Ming Zhang; Marco A. Coccia; Tadahiko Kohno; Anna Tafuri-Bladt; David Brankow; Pauline Campbell; David Chang; Laura Chiu; Tianang Dai; Gordon S. Duncan; Gary Elliott; Ariela Hui; Susan McCabe; Sheila Scully; Arda Shahinian; Christine L. Shaklee; Gwyneth Van; Tak W. Mak; Giorgio Senaldi

T-cell activation requires co-stimulation through receptors such as CD28 (refs 1,2,3) and antigen-specific signalling through the T-cell antigen receptor. Here we describe a new murine co-stimulatory receptor–ligand pair. The receptor, which is related to CD28 and is the homologue of the human protein ICOS, is expressed on activated T cells and resting memory T cells. The ligand, which has homology to B7 molecules and is called B7-related protein-1 (B7RP-1), is expressed on B cells and macrophages. ICOS and B7RP-1 do not interact with proteins in the CD28–B7 pathway, and B7RP-1 co-stimulates T cells in vitro independently of CD28. Transgenic mice expressing a B7RP-1–Fc fusion protein show lymphoid hyperplasia in the spleen, lymph nodes and Peyers patches. Presensitized mice treated with B7RP-1–Fc during antigen challenge show enhanced hypersensitivity. Therefore, B7RP-1 exhibits co-stimulatory activities in vitro and in vivo. ICOS and B7RP-1 define a new and distinct receptor–ligand pair that is structurally related to CD28–B7 and is involved in the adaptive immune response.


Nature Immunology | 2003

The B7 family member B7-H3 preferentially down-regulates T helper type 1-mediated immune responses.

Woong-Kyung Suh; Beata U. Gajewska; Hitoshi Okada; Matthew A. Gronski; Edward M. Bertram; Wojciech Dawicki; Gordon S. Duncan; Jacob Bukczynski; Suzanne Plyte; Andrew Elia; Andrew Wakeham; Annick Itie; Stephen W. Chung; Joan da Costa; Sudha Arya; Tom Horan; Pauline Campbell; Kevin Gaida; Pamela S. Ohashi; Tania H. Watts; Steven Kiyoshi Yoshinaga; Mark R. Bray; Manel Jordana; Tak W. Mak

We investigated the in vivo function of the B7 family member B7-H3 (also known as B7RP-2) by gene targeting. B7-H3 inhibited T cell proliferation mediated by antibody to T cell receptor or allogeneic antigen-presenting cells. B7-H3-deficient mice developed more severe airway inflammation than did wild-type mice in conditions in which T helper cells differentiated toward type 1 (TH1) rather than type 2 (TH2). B7-H3 expression was consistently enhanced by interferon-γ but suppressed by interleukin 4 in dendritic cells. B7-H3-deficient mice developed experimental autoimmune encephalomyelitis several days earlier than their wild-type littermates, and accumulated higher concentrations of autoantibodies to DNA. Thus, B7-H3 is a negative regulator that preferentially affects TH1 responses.


Journal of Immunology | 2002

Inducible Costimulator Costimulates Cytotoxic Activity and IFN-γ Production in Activated Murine NK Cells

Kouetsu Ogasawara; Steven Kiyoshi Yoshinaga; Lewis L. Lanier

The functions of NK cells are regulated by the balance of activating and inhibitory signals. The inhibitory NK cell receptors are well understood; however, less is known about the activating signaling pathways. To explore whether a costimulatory receptor, inducible costimulator (ICOS), is involved in NK cell function, we assessed the role of ICOS in NK cell-mediated cytotoxicity and cytokine production. In addition, to determine whether ICOS contributes to the elimination of tumors in vivo, we examined the tumor growth survival of mice injected with a tumor expressing the ICOS ligand, B7RP-1. We found that ICOS was up-regulated by cytokine stimulation in murine NK cells. Consistent with ICOS expression on activated NK cells, ICOS-dependent cytotoxicity and IFN-γ production were observed, and appeared to require signaling through the phosphoinositide 3-kinase pathway. Interestingly, ICOS-mediated stimulation allowed activated NK cells to kill more efficiently tumor cells expressing MHC class I. Furthermore, fewer metastases appeared in the liver and spleen of mice injected with the ICOS ligand-expressing tumor compared with mice bearing the parental tumor. These results indicate that NK cell functions are regulated by ICOS.


Journal of Immunology | 2002

Essential Role of NF-κB-Inducing Kinase in T Cell Activation Through the TCR/CD3 Pathway

Mitsuru Matsumoto; Takuji Yamada; Steven Kiyoshi Yoshinaga; Tom Boone; Tom Horan; Shigeru Fujita; Yi Li; Tasuku Mitani

NF-κB-inducing kinase (NIK) is involved in lymphoid organogenesis in mice through lymphotoxin-β receptor signaling. To clarify the roles of NIK in T cell activation through TCR/CD3 and costimulation pathways, we have studied the function of T cells from aly mice, a strain with mutant NIK. NIK mutant T cells showed impaired proliferation and IL-2 production in response to anti-CD3 stimulation, and these effects were caused by impaired NF-κB activity in both mature and immature T cells; the impaired NF-κB activity in mature T cells was also associated with the failure of maintenance of activated NF-κB. In contrast, responses to costimulatory signals were largely retained in aly mice, suggesting that NIK is not uniquely coupled to the costimulatory pathways. When NIK mutant T cells were stimulated in the presence of a protein kinase C (PKC) inhibitor, proliferative responses were abrogated more severely than in control mice, suggesting that both NIK and PKC control T cell activation in a cooperative manner. We also demonstrated that NIK and PKC are involved in distinct NF-κB activation pathways downstream of TCR/CD3. These results suggest critical roles for NIK in setting the threshold for T cell activation, and partly account for the immunodeficiency in aly mice.


Journal of Immunology | 2001

Stimulatory Effects of B7-Related Protein-1 on Cellular and Humoral Immune Responses in Mice

Jane Guo; Marina Stolina; James Bready; Songmei Yin; Tom Horan; Steven Kiyoshi Yoshinaga; Giorgio Senaldi

Inducible costimulator (ICOS) and B7-related protein-1 (B7RP-1) constitute a receptor-ligand pair involved in T cell costimulation. In this study, the stimulatory effects of B7RP-1 on cellular and humoral immune responses were investigated giving mice a construct with the extracellular domain of murine B7RP-1 fused with human IgG1 Fc (B7RP-1-Fc). B7RP-1-Fc stimulated contact hypersensitivity (CH) given near either the time of sensitization or challenge with oxazolone. When given near challenge time, B7RP-1-Fc stimulated CH more than a construct containing the extracellular domain of murine B7.2 and Fc (B7.2-Fc). B7RP-1-Fc increased the number of cells in lymph nodes draining the skin sensitized with oxazolone, especially activated T cells. B7RP-1-Fc also increased the ability of the cells in these lymph nodes to induce CH when transfused into naive mice. B7RP-1-Fc stimulated the production of anti-keyhole limpet hemocyanin (KLH) Ab, increasing anti-KLH IgG, IgG2a, and IgE, whereas B7.2-Fc did not affect this production. B7RP-1-Fc also increased the number of cells in lymph nodes draining the skin immunized with KLH and their production of IFN-γ, IL-4, and IL-10 in response to KLH. Finally, B7RP-1-Fc increased the presence of eosinophils in the bronchoalveolar lavage and lungs of mice sensitized and challenged with OVA so to mount an asthmatic reaction. B7RP-1-Fc stimulates both cellular and humoral immune responses in vivo by increasing number and function of T and B cells reacting to Ag exposure.


Journal of Immunology | 2002

Inducible Costimulator Protein Controls the Protective T Cell Response Against Listeria monocytogenes

Hans-Willi Mittrücker; Mischo Kursar; Anne Köhler; Donna Yanagihara; Steven Kiyoshi Yoshinaga; Stefan H. E. Kaufmann

The inducible costimulator protein (ICOS) was recently identified as a costimulatory molecule for T cells. Here we analyze the role of ICOS for the acquired immune response of mice against the intracellular bacterium Listeria monocytogenes. During oral L. monocytogenes infection, low levels of ICOS expression were detected by extracellular and intracellular Ab staining of Listeria-specific CD4+ and CD8+ T cells. Blocking of ICOS signaling with a soluble ICOS-Ig fusion protein markedly impaired the Listeria-specific T cell responses. Compared with control mice, the ICOS-Ig treated mice generated significantly reduced numbers of Listeria-specific CD8+ T cells in spleen and liver, as determined by tetramer and intracellular cytokine staining. In contrast, the specific CD8+ T cell response in the intestinal mucosa did not appear to be impaired by the ICOS-Ig treatment. Analysis of the CD4+ T cell response revealed that ICOS-Ig treatment also affected the specific CD4+ T cell response. When restimulated with listerial Ag in vitro, reduced numbers of CD4+ T cells from infected and ICOS-Ig-treated mice responded with IFN-γ production. The impaired acquired immune response in ICOS-Ig treated mice was accompanied by their increased susceptibility to L. monocytogenes infection. ICOS-Ig treatment drastically enhanced bacterial titers, and a large fraction of mice succumbed to the otherwise sublethal dose of infection. Thus, ICOS costimulation is crucial for protective immunity against the intracellular bacterium L. monocytogenes.


International Journal of Cancer | 2003

Potent activity of soluble B7RP-1-Fc in therapy of murine tumors in syngeneic hosts.

Gulshan Ara; Angelo Baher; Neal Storm; Tom Horan; Claudia Baikalov; Emil Brisan; Reuben Camacho; Alison Moore; Hartt Goldman; Tadahiko Kohno; Russell C. Cattley; Gwyneth Van; Kevin Gaida; Ming Zhang; John S. Whoriskey; David Fong; Steven Kiyoshi Yoshinaga

We have characterized a receptor:ligand pair, ICOS:B7RP‐1, that is structurally and functionally related to CD28:B7.1/2. We reported previously that B7RP‐1 costimulates T cell proliferation and immune responses (Yoshinaga et al., Nature 1999;402:827–32; Guo et al., J Immunol 2001;166:5578–84; Yoshinaga et al., Int Immunol 2000;12:1439–47). We report that B7RP‐1‐Fc causes rejection or growth inhibition of Meth A, SA‐1 and EMT6 tumors in syngeneic mice. Established Meth A tumors were rejected effectively with a single dose of B7RP‐1‐Fc, however, the treatment was less effective on larger tumors. Mice that rejected Meth A tumors previously by Day 30, also rejected a subsequent Meth A challenge on Day 60, without additional B7RP‐1‐Fc treatment, indicating a long‐lived memory response. Tumor cells believed to be less immunogenic, such as P815 and EL‐4 cells, were less responsive to this treatment. The EL‐4 responsiveness to the B7RP‐1‐Fc treatment was enhanced, however, by pre‐treatment of the mice with cyclophosphamide. As expected, T cells appeared to be targeted by B7RP‐1‐Fc treatment. Thus, the administration of soluble B7RP‐1‐Fc may have therapeutic value in generating or enhancing anti‐tumor activity in a clinical setting.


Inflammation | 2003

Interaction of B7RP-1 with ICOS negatively regulates antigen presentation by B cells.

Patricia Wahl; Roland Schoop; Thomas P. Horan; Steven Kiyoshi Yoshinaga; Rudolf P. Wüthrich

Stimulation of T cells through the T cell receptor is insufficient for optimal T cell activation. A second activation signal is necessary, being usually provided by the costimulatory molecule CD28. Recently, additional costimulatory pathways have been identified, including inducible costimulator (ICOS) and its ligand B7RP-1. We have examined the role of the B7RP-1/ICOS costimulatory pathway on antigen presentation by B cells, using the I-Ak and I-Ek-positive CH27 B cell line and several different T cell lines. We found that CH27 expressed B7RP-1 and PD-L1 whereas the T cell lines expressed ICOS and PD-1. In the presence of HEL, the T cell hybridomas C10 and 3A9 released IL-2, which is indicative of antigen-specific T cell activation by the CH27 cells. Unexpectedly, blocking antibodies for B7RP-1 and ICOS enhanced the IL-2 response in both T cells. As expected, an increase in the production of IL-2 was seen when blocking antibodies for PD-1 were used. Blocking with antibodies for I-Ak, CD28, B7.1, and B7.2 lead to a decrease in IL-2 production. Additionally we tested a Th1 and a Th2 T cell clone. Blockade of B7RP-1/ICOS lead to an increased IFN-γ response in Th1 cells (A.E7) and an increased IL-4 response in Th2 cells (D10.G4.1). Intracellular staining also showed an increase in cytokine production when the B7RP-1/ICOS pathway was blocked. In conclusion, the B7RP-1/ICOS pathway is negatively regulating T cell activation by B cells and may play a role similar to that of the PD-L1/PD-1 pathway.


Nucleic Acids Research | 1995

Molecular cloning of LSIRF, a lymphoid-specific member of the interferon regulatory factor family that binds the interferon-stimulated response element (ISRE)

Toshifumi Matsuyama; Alex Grossman; Hans-Willi Mittrücker; David P. Siderovski; Friedemann Kiefer; Takatoshi Kawakami; Christopher D. Richardson; Tadatsugu Taniguchi; Steven Kiyoshi Yoshinaga; Tak W. Mak


Journal of Biological Chemistry | 1996

Transformation of NIH 3T3 Cells by HER3 or HER4 Receptors Requires the Presence of HER1 or HER2

Ke Zhang; Jilin Sun; Naili Liu; Duanzhi Wen; David Chang; Arlen R. Thomason; Steven Kiyoshi Yoshinaga

Collaboration


Dive into the Steven Kiyoshi Yoshinaga's collaboration.

Top Co-Authors

Avatar

Tak W. Mak

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Arda Shahinian

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge