Network


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

Hotspot


Dive into the research topics where Toyo Suzuki is active.

Publication


Featured researches published by Toyo Suzuki.


Journal of Immunology | 2001

CD24 Induces Apoptosis in Human B Cells Via the Glycolipid-Enriched Membrane Domains/Rafts-Mediated Signaling System

Toyo Suzuki; Nobutaka Kiyokawa; Tomoko Taguchi; Takaomi Sekino; Yohko U. Katagiri; Junichiro Fujimoto

The glycosylphosphatidylinositol-anchored CD24 protein is a B cell differentiation Ag that is expressed on mature resting B cells but disappears upon Ag stimulation. We used Burkitt’s lymphoma (BL) cells, which are thought to be related to germinal center B cells, to examine the biological effect of Ab-mediated CD24 cross-linking on human B cells and observed 1) induction of apoptosis in BL cells mediated by cross-linking of CD24; and 2) synergism between the cross-linking of CD24 and that of the B cell receptor for Ag in the effect on apoptosis induction. We also observed activation of mitogen-activated protein kinases following CD24 cross-linking, suggesting that CD24 mediates the intracellular signaling that leads to apoptosis in BL cells. Although CD24 has no cytoplasmic portion to transduce signals intracellularly, analysis of biochemically separated glycolipid-enriched membrane (GEM) fractions indicated enhanced association of CD24 and Lyn protein tyrosine kinase in GEM as well as increased Lyn kinase activity after CD24 cross-linking, suggesting that CD24 mediates intracellular signaling via a GEM-dependent mechanism. Specific microscopic cocapping of CD24 and Lyn, but not of other kinases, following CD24 cross-linking supported this idea. We further observed that apoptosis induction by cross-linking is a common feature shared by GEM-associated molecules expressed on BL cells, including GPI-anchored proteins and glycosphingolipids. CD24-mediated apoptosis in BL cells may provide a model for the cell death mechanism initiated by GEM-associated molecules, which is closely related to B cell receptor for Ag-mediated apoptosis.


Journal of Biological Chemistry | 2001

Kinetic Analysis of Binding between Shiga Toxin and Receptor Glycolipid Gb3Cer by Surface Plasmon Resonance

Hideki Nakajima; Nobutaka Kiyokawa; Yohko U. Katagiri; Tomoko Taguchi; Toyo Suzuki; Takaomi Sekino; Kenichi Mimori; Tomohiko Ebata; Masahiro Saito; Hiroshi Nakao; Tae Takeda; Junichiro Fujimoto

Shiga toxin (Stx) binds to the receptor glycolipid Gb3Cer on the cell surface and is responsible for hemolytic uremic syndrome. Stx has two isoforms, Stx1 and Stx2, and in clinical settings Stx2 is known to cause more severe symptoms, although the differences between the mechanisms of action of Stx1 and Stx2 are as yet unknown. In this study, the binding modes of these two isoforms to the receptor were investigated with a surface plasmon resonance analyzer to compare differences by real time receptor binding analysis. A sensor chip having a lipophilically modified dextran matrix or quasicrystalline hydrophobic layer was used to immobilize an amphipathic lipid layer that mimics the plasma membrane surface. Dose responsiveness was observed with both isoforms when either the toxin concentration or the Gb3Cer concentration was increased. In addition, this assay was shown to be specific, because neither Stx1 nor Stx2 bound to GM3, but both bound weakly to Gb4Cer. It was also shown that a number of fitting models can be used to analyze the sensorgrams obtained with different concentrations of the toxins, and the “bivalent analyte” model was found to best fit the interaction between Stxs and Gb3Cer. This shows that the interaction between Stxs and Gb3Cer in the lipid bilayer has a multivalent effect. The presence of cholesterol in the lipid bilayer significantly enhanced the binding of Stxs to Gb3Cer, although kinetics were unaffected. The association and dissociation rate constants of Stx1 were larger than those of Stx2: Stx2 binds to the receptor more slowly than Stx1 but, once bound, is difficult to dissociate. The data described herein clearly demonstrate differences between the binding properties of Stx1 and Stx2 and may facilitate understanding of the differences in clinical manifestations caused by these toxins.


Experimental Hematology | 2000

Globotriaosyl ceramide (CD77/Gb3) in the glycolipid-enriched membrane domain participates in B-cell receptor–mediated apoptosis by regulating Lyn kinase activity in human B cells

Tetsuya Mori; Nobutaka Kiyokawa; Yohko U. Katagiri; Tomoko Taguchi; Toyo Suzuki; Takaomi Sekino; Norihide Sato; Kazuhiro Ohmi; Hideki Nakajima; Tae Takeda; Junichiro Fujimoto

The role of CD77 expressed on a fraction of germinal center B cells, also known as glycosphyngolipid Gb3, and as a functional receptor for Shiga toxins (Stx) in B-cell receptor (BCR)-mediated apoptosis was investigated. Using Stx1-sensitive Burkitts lymphoma Ramos cells as an in vitro model of CD77(+) germinal center B cells, intracellular signaling events mediated by either Stx1 or anti-CD77 antibody were examined immunobiochemically and immunocytologically. We observed prompt activation of Lyn and Syk kinases leading to increased binding of these proteins to surface IgM (sIgM) in Ramos cells after Stx1 treatment. We also observed microscopic colocalization of CD77 and sIgM after stimulation with Stx1. Along with the synergism between the cross-linking of CD77 and that of sIgM in their effect on apoptosis induction, it was highly probable that CD77 cross-linking induces activation of the BCR signaling cascade. Analysis using sucrose density gradient centrifugation suggested that Stx1 binding to CD77 induced recruitment and activation of Lyn in the glycolipid-enriched membrane (GEM) fractions. Once activated, however, Lyn seemed to acquire an increased detergent solubility and moved outside of the GEM fractions. This study describes the participation of the GEM domain in BCR-signaling cascade and suggests a possible role of CD77 as a regulator of BCR-induced apoptosis in human B cells.


Journal of Immunology | 2003

Pre-B Cell Antigen Receptor-Mediated Signal Inhibits CD24-Induced Apoptosis in Human Pre-B Cells

Tomoko Taguchi; Nobutaka Kiyokawa; Kenichi Mimori; Toyo Suzuki; Takaomi Sekino; Hideki Nakajima; Masahiro Saito; Yohko U. Katagiri; Nobutake Matsuo; Yoshinobu Matsuo; Hajime Karasuyama; Junichiro Fujimoto

We previously reported that the cross-linking of cluster of differentiation (CD)24 induces apoptosis in Burkitt’s lymphoma cells and that this phenomenon can be enhanced by a B cell Ag receptor (BCR)-mediated signal. In this study, we extend our previous observation and report that CD24 also mediated apoptosis in human precursor-B acute lymphoblastic leukemia cell lines in the pro-B and pre-B stages accompanying activation of multiple caspases. Interestingly, simultaneous cross-linking of pre-BCR clearly inhibited CD24-mediated apoptosis in pre-B cells. We also observed that mitogen-activated protein kinases (MAPKs) were involved in the regulation of this apoptotic process. Pre-BCR cross-linking induced prompt and strong activation of extracellular signal-regulated kinase 1, whereas CD24 cross-linking induced the sustained activation of p38 MAPK, following weak extracellular signal-regulated kinase 1 activation. SC68376, a specific inhibitor of p38 MAPK, inhibited apoptosis induction by CD24 cross-linking, whereas anisomycin, an activator of p38 MAPK, enhanced the apoptosis. In addition, PD98059, a specific inhibitor of MEK-1, enhanced apoptosis induction by CD24 cross-linking and reduced the antiapoptotic effects of pre-BCR cross-linking. Collectively, whether pre-B cells survive or die may be determined by the magnitude of MAPK activation, which is regulated by cell surface molecules. Our findings should be important to understanding the role of CD24-mediated cell signaling in early B cell development.


Experimental Hematology | 2002

Granulocyte colony-stimulating factor directly affects human monocytes and modulates cytokine secretion

Masahiro Saito; Nobutaka Kiyokawa; Tomoko Taguchi; Kyoko Suzuki; Takaomi Sekino; Kenichi Mimori; Toyo Suzuki; Hideki Nakajima; Yohko U. Katagiri; Junya Fujimura; Hiroo Fujita; Koichi Ishimoto; Yuichiro Yamashiro; Junichiro Fujimoto

OBJECTIVE Recent reports have indicated that monocytes express receptors for the granulocyte colony-stimulating factor (G-CSF). The direct effects of G-CSF on cytokine secretion in monocytes were examined. MATERIALS AND METHODS A monocytic cell line NOMO-1 that secretes multiple cytokines upon stimulation with lipopolysaccharide (LPS) was used. Normal human monocytes were purified by negative selection using magnetic beads. Cells pretreated with or without G-CSF were stimulated with LPS, and the subsequent concentrations of cytokines and chemokines in supernatants were determined by sandwich enzyme-linked immunosorbent assay. RESULTS NOMO-1 cells were found to express receptors for G-CSF. Although G-CSF stimulation did not induce cytokine secretion, pretreatment with G-CSF significantly attenuated LPS-stimulated secretion of the proinflammatory cytokines tumor necrosis factor-alpha and interleukin (IL)-12 in NOMO-1 cells. Simultaneously, however, G-CSF pretreatment apparently enhanced LPS-induced secretion of IL-10 and monocyte chemoattractant protein-1, whereas secretions of IL-1beta, IL-6, and IL-8 were unaffected. When normal human monocytes from healthy volunteers were similarly examined, marked individual variations in LPS-induced secretion of cytokines were observed. Although some exceptions exist, a similar tendency as to the effects of G-CSF treatment on cytokine secretions as that in NOMO-1 cells was observed in human monocytes. CONCLUSIONS Our data suggest that G-CSF directly affects monocytes and modulates their cytokine secretion. NOMO-1 cells can provide an alternate model for in vitro culture of monocytes to investigate the effects of G-CSF on cytokine secretion by these cells.


Journal of Cellular Biochemistry | 2001

Activation of the caspase cascade during Stx1‐induced apoptosis in Burkitt's lymphoma cells

Nobutaka Kiyokawa; Tetsuya Mori; Tomoko Taguchi; Masahiro Saito; Kenichi Mimori; Toyo Suzuki; Takaomi Sekino; Norihide Sato; Hideki Nakajima; Yohko U. Katagiri; Tae Takeda; Junichiro Fujimoto

Shiga toxin 1 (Stx1) produced by Escherichia coli has been reported to induce apoptosis in many different cell types, including Burkitts lymphoma (BL) cells. Since it has been established that the caspases play essential roles as the effector molecules in the apoptotic process in most cases, we examined the kinetics of caspase activation during the process of Stx1‐mediated apoptosis of BL cells. Using Ramos BL cells that are highly sensitive to Stx1‐mediated cytotoxicity, we observed that multiple caspases, including caspase‐3, ‐7, and ‐8 were promptly activated following Stx1 treatment, as indicated by both the procaspase cleavages and enhancement of cleavage of the tetrapeptide substrates of the caspases. In addition, the inhibition assay revealed that caspase‐8 is located upstream of both caspase‐3 and ‐7, suggesting that Stx1‐mediated apoptosis utilizes a similar caspase cascade to that involved in Fas‐mediated apoptosis. Neither anti‐Fas mAb nor TNF‐α, however, affected the Stx1‐mediated apoptosis of Ramos cells. Although the precise mechanism of Stx1‐mediated activation of caspase‐8 is still unclear, we have demonstrated that crosslinkage of CD77, a functional receptor for Stx1, with specific antibody is sufficient to induce activation of caspase‐8. Our findings should provide new insight into the understanding of the molecular basis of Stx1‐mediated cell injury. J. Cell. Biochem. 81:128–142, 2001.


Microbiology and Immunology | 2004

Characterization of a Shiga‐Toxin 1‐Resistant Stock of Vero Cells

Takaomi Sekino; Nobutaka Kiyokawa; Tomoko Taguchi; Hisami Takenouchi; Jun Matsui; Wei Ran Tang; Toyo Suzuki; Hideki Nakajima; Masahiro Saito; Kazuhiro Ohmi; Yohko U. Katagiri; Hajime Okita; Hiroshi Nakao; Tae Takeda; Junichiro Fujimoto

Shiga toxins (Stxs, also referred to as verotoxins) were first described as a novel cytotoxic activity against Vero cells. In this study, we report the characterization of an Stx1‐resistant (R‐) stock of Vero cells. (1) When the susceptibility of R‐Vero cells to Stx1 cytotoxicity was compared to that of Stx1‐sensitive (S‐) Vero cells by methylthiazolyldiphenyl‐tetrazolium bromide (MTT) assay, cell viability after 48‐hr exposure to 10 pg/ml of Stx1 was greater than 80% and less than 15%, respectively. (2) Although both a binding assay of fluorescence‐labeled Stx1 and lipid analysis indicated considerable expression of Gb3Cer, a functional receptor for Stxs, in both Vero cells, anti‐Gb3Cer monoclonal antibodies capable of binding to S‐Vero cells failed to effectively label R‐Vero cells, suggesting a conformational difference in the Gb3Cer expressed on R‐Vero cells. (3) The lipid analysis also showed that the R‐Vero cells contained significant amounts of Gb4Cer. In addition, introduction of exogenous Gb4Cer into S‐Vero cells slightly inhibited Stx1 cytotoxicity, suggesting some correlation between glycosphingolipid composition and Stx1 resistance. (4) Both butyrate treatment and serum depression eliminated the Stx1 resistance of R‐Vero cells. (5) The results of the analysis by confocal microscopy suggest a difference in intracellular transport of Stx1 between R‐Vero and S‐Vero cells. Further study of R‐Vero cells may provide a model of Stx1 resistance via distinct intracellular transport of Stx1.


Hybridoma | 2000

Characterization of monoclonal antibodies against mouse and rat platelet glycoprotein V (CD42d).

Norihide Sato; Nobutaka Kiyokawa; Koji Takada; Mitsuko Itagaki; Masahiro Saito; Takaomi Sekino; Toyo Suzuki; Tomoko Taguchi; Kenichi Mimori; François Lanza; Junichiro Fujimoto

The mouse- and rat-platelet-specific hamster monoclonal antibody (MAb) 1C2, previously found to react with a thrombin-sensitive 74-kD glycoprotein, was now shown to recognize platelet glycoprotein V (GPV, CD42d). 1C2 reacted with NIH-3T3 cells in which recombinant mouse or rat GPV was expressed. Both 1C2 and 4A5, another mouse-platelet-specific rat MAb, immunoprecipitated GVP, although they recognized different epitopes. Side-by-side comparison confirmed that 1C2 as well as RPM.9, a MAb against rat GPV, recognized the same rat platelet molecule. In a mouse bone marrow culture, 1C2+ megakaryocytes emerged from CD41 (GPIIb)+1C2- megakaryocytes. Because 1C2+ megakaryocytes exhibited higher DNA ploidy distribution than CD41+ cells, GPV likely appears in the late stage of megakaryocyte maturation. This study established 1C2 as a MAb against mouse and rat GPV, namely CD42d, and as useful tool to study rodent megakaryopoiesis.


The Journal of Infectious Diseases | 2002

Inhibition of Shiga Toxin Cytotoxicity in Human Renal Cortical Epithelial Cells by Nitrobenzylthioinosine

Takaomi Sekino; Nobutaka Kiyokawa; Tomoko Taguchi; Kazuhiro Ohmi; Hideki Nakajima; Toyo Suzuki; Susumu Furukawa; Hiroshi Nakao; Tae Takeda; Junichiro Fujimoto

Nitrobenzylthioinosine (NBTI), a nucleoside-transport inhibitor, has been found to possess the ability to prevent the cytotoxic action of Shiga toxin (Stx) 1 in human renal cortical epithelial cells (HRCECs), thereby protecting HRCECs from cell death. Further examination revealed that NBTI does not affect either the binding or the endocytosis of Stx1 but alters the intracellular transport of Stx1. Generally, endocytosed Stx1 is thought to be transported from endosomes to the endoplasmic reticulum. In NBTI-treated cells, however, the endocytosed Stx1 is delivered to an early endosome, but no further transportation occurs. Moreover, Stx1 is rapidly excreted from NBTI-treated HRCECs, preventing the accumulation of Stx1. Investigation of the NBTI-mediated protection mechanism against Stx cytotoxicity may provide insights into the analysis of Stx-mediated cell damage and lead to improvements in therapeutic approaches for diseases caused by Stx.


Protein Expression and Purification | 2001

Single-step method for purification of Shiga toxin-1 B subunit using receptor-mediated affinity chromatography by globotriaosylceramide-conjugated octyl sepharose CL-4B.

Hideki Nakajima; Yohko U. Katagiri; Nobutaka Kiyokawa; Tomoko Taguchi; Toyo Suzuki; Takaomi Sekino; Kenichi Mimori; Masahiro Saito; Hiroshi Nakao; Tae Takeda; Junichiro Fujimoto

Collaboration


Dive into the Toyo Suzuki's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomoko Taguchi

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hideki Nakajima

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masahiro Saito

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge