Network


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

Hotspot


Dive into the research topics where Toru Atsumi is active.

Publication


Featured researches published by Toru Atsumi.


Journal of Immunology | 2004

IL-6 Regulates In Vivo Dendritic Cell Differentiation through STAT3 Activation

Sung-Joo Park; Takayuki Nakagawa; Hidemitsu Kitamura; Toru Atsumi; Hokuto Kamon; Shinichiro Sawa; Daisuke Kamimura; Naoko Ueda; Yoichiro Iwakura; Katsuhiko Ishihara; Masaaki Murakami; Toshio Hirano

Dendritic cells (DCs) orchestrate immune responses according to their state of maturation. In response to infection, DCs differentiate into mature cells that initiate immune responses, while in the absence of infection, most of them remain in an immature form that induces tolerance to self Ags. Understanding what controls these opposing effects is an important goal for vaccine development and prevention of unwanted immune responses. A crucial question is what cytokine(s) regulates DC maturation in the absence of infection. In this study, we show that IL-6 plays a major role in maintaining immature DCs. IL-6 knockout (KO) mice had increased numbers of mature DCs, indicating that IL-6 blocks DC maturation in vivo. We examined this effect further in knockin mice expressing mutant versions of the IL-6 signal transducer gp130, with defective signaling through either Src homology region 2 domain-containing phosphatase 2/Gab/MAPK (gp130F759/F759) or STAT3 (gp130FxxQ/FxxQ), and combined gp130 and IL-6 defects (gp130F759/F759/IL-6 KO mice). Importantly, we found STAT3 activation by IL-6 was required for the suppression of LPS-induced DC maturation. In addition, STAT3 phosphorylation in DCs was regulated by IL-6 in vivo, and STAT3 was necessary for the IL-6 suppression of bone marrow-derived DC activation/maturation. DC-mediated T cell activation was enhanced in IL-6 KO mice and suppressed in gp130F759/F759 mice. IL-6 is thus a potent regulator of DC differentiation in vivo, and IL-6-gp130-STAT3 signaling in DCs may represent a critical target for controlling T cell-mediated immune responses in vivo.


Journal of Experimental Medicine | 2002

A Point Mutation of Tyr-759 in Interleukin 6 Family Cytokine Receptor Subunit gp130 Causes Autoimmune Arthritis

Toru Atsumi; Katsuhiko Ishihara; Daisuke Kamimura; Hideto Ikushima; Takuya Ohtani; Seiichi Hirota; Hideyuki Kobayashi; Sung-Joo Park; Yukihiko Saeki; Yukihiko Kitamura; Toshio Hirano

We generated a mouse line in which the src homology 2 domain–bearing protein tyrosine phosphatase (SHP)-2 binding site of gp130, tyrosine 759, was mutated to phenylalanine (gp130 F759/F759). The gp130 F759/F759 mice developed rheumatoid arthritis (RA)-like joint disease. The disease was accompanied by autoantibody production and accumulated memory/activated T cells and myeloid cells. Before the disease onset, the T cells were hyperresponsive and thymic selection and peripheral clonal deletion were impaired. The inhibitory effect of IL-6 on Fas ligand expression during activation-induced cell death (AICD) was augmented in gp130 F759/F759 T cells in a manner dependent on the tyrosine residues of gp130 required for signal transducer and activator of transcription 3 activation. Finally, we showed that disease development was dependent on lymphocytes. These results provide evidence that a point mutation of a cytokine receptor has the potential to induce autoimmune disease.


Journal of Immunology | 2009

Comment on “Gene Disruption Study Reveals a Nonredundant Role for TRIM21/Ro52 in NF-κB-Dependent Cytokine Expression in Fibroblasts”

Ryusuke Yoshimi; Tsung-Hsien Chang; Hongsheng Wang; Toru Atsumi; Herbert C. Morse; Keiko Ozato

The tripartite motif (TRIM) family member, TRIM21, is an E3 ubiquitin ligase for IFN regulatory factor (IRF)3 and IRF8 that functions in both innate and acquired immunity. It is also an autoantigen known as Ro52/SS-A. The function of TRIM21 in vivo, however, has remained elusive. We generated Trim21−/− mice with the Trim21 gene replaced by an enhanced GFP (EGFP) reporter. EGFP expression analyses showed that Trim21 was widely expressed in many tissues, with the highest levels in immune cells. Studies of Trim21−/− embryonic fibroblasts demonstrated that TLR-mediated induction of proinflammatory cytokines, including IL-1β, IL-6, TNF-α, and CXCL10, was consistently up-regulated relative to wild-type cells. Reporter analyses demonstrated that TLR-mediated NF-κB activation was higher in Trim21−/− cells than in wild-type cells, most likely accounting for their enhanced cytokine expression. In contrast, functional analyses of immune cells from Trim21−/− mice revealed no abnormalities in their composition or function, even though ubiquitylation of IRF3 and IRF8 was impaired. These results suggested possible redundancies in activities mediated by TRIM21. In keeping with this concept, we found that a number of TRIM family members were up-regulated in Trim21−/− cells. Taken together, these findings demonstrate that TRIM21 plays a previously unrecognized role in the negative regulation of NF-κB-dependent proinflammatory cytokine responses, and suggest that multiple TRIM proteins contribute to the maintenance of functional equilibrium in inflammatory responses, in part through functional redundancy.


Cancer Research | 2014

Inflammation Amplifier, a New Paradigm in Cancer Biology

Toru Atsumi; Rajeev Singh; Lavannya Sabharwal; Hidenori Bando; Jie Meng; Yasunobu Arima; Moe Yamada; Masaya Harada; Jing-Jing Jiang; Daisuke Kamimura; Hideki Ogura; Toshio Hirano; Masaaki Murakami

Tumor-associated inflammation can induce various molecules expressed from the tumors themselves or surrounding cells to create a microenvironment that potentially promotes cancer development. Inflammation, particularly chronic inflammation, is often linked to cancer development, even though its evolutionary role should impair nonself objects including tumors. The inflammation amplifier, a hyperinducer of chemokines in nonimmune cells, is the principal machinery for inflammation and is activated by the simultaneous stimulation of NF-κB and STAT3. We have redefined inflammation as local activation of the inflammation amplifier, which causes an accumulation of various immune cells followed by dysregulation of local homeostasis. Genes related to the inflammation amplifier have been genetically associated with various human inflammatory diseases. Here, we describe how cancer-associated genes, including interleukin (IL)-6, Ptgs2, ErbB1, Gas1, Serpine1, cMyc, and Vegf-α, are strongly enriched in genes related to the amplifier. The inflammation amplifier is activated by the stimulation of cytokines, such as TNF-α, IL-17, and IL-6, resulting in the subsequent expression of various target genes for chemokines and tumor-related genes like BCL2L11, CPNE7, FAS, HIF1-α, IL-1RAP, and SOD2. Thus, we conclude that inflammation does indeed associate with the development of cancer. The identified genes associated with the inflammation amplifier may thus make potential therapeutic targets of cancers.


International Immunology | 2010

Zinc suppresses Th17 development via inhibition of STAT3 activation

Chika Kitabayashi; Toshiyuki Fukada; Minoru Kanamoto; Wakana Ohashi; Shintaro Hojyo; Toru Atsumi; Naoko Ueda; Ichiro Azuma; Hiroshi Hirota; Masaaki Murakami; Toshio Hirano

Zinc (Zn) is an essential trace metal required by many enzymes and transcription factors for their activity or the maintenance of their structure. Zn has a variety of effects in the immune responses and inflammation, although it has not been well known how Zn affects these reactions on the molecular basis. We here showed that Zn suppresses T(h)17-mediated autoimmune diseases at lest in part by inhibiting the development of T(h)17 cells via attenuating STAT3 activation. In mice injected with type II collagen to induce arthritis, Zn treatment inhibited T(h)17 cell development. IL-6-mediated activation of STAT3 and in vitro T(h)17 cell development were all suppressed by Zn. Importantly, Zn binding changed the alpha-helical secondary structure of STAT3, disrupting the association of STAT3 with JAK2 kinase and with a phospho-peptide that included a STAT3-binding motif from the IL-6 signal transducer gp130. Thus, we conclude that Zn suppresses STAT3 activation, which is a critical step for T(h)17 development.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Zinc transporter SLC39A10/ZIP10 controls humoral immunity by modulating B-cell receptor signal strength.

Shintaro Hojyo; Tomohiro Miyai; Hitomi Fujishiro; Masami Kawamura; Takuwa Yasuda; Atsushi Hijikata; Bum-Ho Bin; Tarou Irie; Junichi Tanaka; Toru Atsumi; Masaaki Murakami; Manabu Nakayama; Osamu Ohara; Seiichiro Himeno; Hisahiro Yoshida; Haruhiko Koseki; Tomokatsu Ikawa; Kenji Mishima; Toshiyuki Fukada

Significance The essential micronutrient zinc is known to modulate adaptive immune responses and dysregulated zinc homeostasis leads to immunodeficiency. However, the molecular mechanisms underlying this zinc-mediated modulation are unknown. We show that the zinc transporter ZIP10 plays an important role in B-cell receptor (BCR) signaling. Zip10-deficiency in mature B cells attenuated both T-cell–dependent and –independent immune responses. Zip10-deficient mature B cells proliferated poorly in response to BCR cross-linking, as a result of dysregulated BCR signaling. Our data establish that ZIP10 functions as a cellular regulator to modulate BCR signaling in humoral immune responses. The humoral immune response, also called the antibody-mediated immune response, is one of the main adaptive immune systems. The essential micronutrient zinc (Zn) is known to modulate adaptive immune responses, and dysregulated Zn homeostasis leads to immunodeficiency. However, the molecular mechanisms underlying this Zn-mediated modulation are largely unknown. Here, we show that the Zn transporter SLC39A10/ZIP10 plays an important role in B-cell antigen receptor (BCR) signal transduction. Zip10-deficiency in mature B cells attenuated both T-cell–dependent and –independent immune responses in vivo. The Zip10-deficient mature B cells proliferated poorly in response to BCR cross-linking, as a result of dysregulated BCR signaling. The perturbed signaling was found to be triggered by a reduction in CD45R phosphatase activity and consequent hyperactivation of LYN, an essential protein kinase in BCR signaling. Our data suggest that ZIP10 functions as a positive regulator of CD45R to modulate the BCR signal strength, thereby setting a threshold for BCR signaling in humoral immune responses.


Cell Reports | 2013

Disease-Association Analysis of an Inflammation-Related Feedback Loop

Masaaki Murakami; Masaya Harada; Daisuke Kamimura; Hideki Ogura; Yuko Okuyama; Noriko Kumai; Azusa Okuyama; Rajeev Singh; Jing-Jing Jiang; Toru Atsumi; Sayaka Shiraya; Yuji Nakatsuji; Makoto Kinoshita; Hitoshi Kohsaka; Makoto Nishida; Saburo Sakoda; Nobuyuki Miyasaka; Keiko Yamauchi-Takihara; Toshio Hirano

The IL-6-triggered positive feedback loop for NFκB signaling (or the IL-6 amplifier/Inflammation amplifier) was originally discovered as a synergistic-activation signal that follows IL-17/IL-6 stimulation in nonimmune cells. Subsequent results from animal models have shown that the amplifier is activated by stimulation of NFκB and STAT3 and induces chemokines and inflammation via an NFκB loop. However, its role in human diseases is unclear. Here, we combined two genome-wide mouse screens with SNP-based disease association studies, revealing 1,700 genes related to the IL-6 amplifier, 202 of which showed 492 indications of association with ailments beyond autoimmune diseases. We followed up on ErbB1 from our list. Blocking ErbB1 signaling suppressed the IL-6 amplifier, whereas the expression of epiregulin, an ErbB1 ligand, was higher in patients with inflammatory diseases. These results indicate that the IL-6 amplifier is indeed associated with human diseases and disorders and that the identified genes may make for potential therapeutic targets.


Journal of Immunology | 2004

Evidence of a Novel IL-2/15Rβ-Targeted Cytokine Involved in Homeostatic Proliferation of Memory CD8+ T Cells

Daisuke Kamimura; Naoko Ueda; Yukihisa Sawa; Shinji Hachida; Toru Atsumi; Takayuki Nakagawa; Shinichiro Sawa; Gui-Hua Jin; Haruhiko Suzuki; Katsuhiko Ishihara; Masaaki Murakami; Toshio Hirano

The homeostasis of memory CD8+ T cells is regulated by cytokines. IL-15 is shown to promote the proliferation of memory CD8+ T cells, while IL-2 suppresses their division in vivo. This inhibitory effect of IL-2 appears to occur indirectly, through other cell populations including CD25+CD4+ T cells; however, the details of this mechanism remain unclear. In this study, we show that 1) both Ag-experienced and memory phenotype CD8+ T cells divided after the depletion of IL-2 in vivo; 2) this division occurred normally and CD44highIL-2/15Rβhigh CD8+ T cells generated after IL-2 depletion in IL-15 knockout (KO) and in IL-7-depleted IL-15 KO mice; 3) surprisingly, the blockade of IL-2/15Rβ signaling in IL-2-depleted IL-15 KO mice completely abolished the division of memory CD8+ T cells, although the only cytokines known to act through IL-2/15Rβ are IL-2 and IL-15; and 4) the expression of IL-2/15Rβ molecules on memory CD8+ T cells was required for their division induced by IL-2 depletion. These results demonstrate that the depletion of IL-2 in vivo induced memory CD8+ T cell division by an IL-15-independent but by an IL-2/15Rβ-dependent mechanism, suggesting the existence of a novel IL-2/15Rβ-utilizing cytokine that acts directly on memory CD8+ T cells to promote cell division.


eLife | 2015

A pain-mediated neural signal induces relapse in murine autoimmune encephalomyelitis, a multiple sclerosis model

Yasunobu Arima; Daisuke Kamimura; Toru Atsumi; Masaya Harada; Tadafumi Kawamoto; Naoki Nishikawa; Andrea Stofkova; Takuto Ohki; Kotaro Higuchi; Yuji Morimoto; Peter Wieghofer; Yuka Okada; Yuki Mori; Saburo Sakoda; Shizuya Saika; Yoshichika Yoshioka; Issei Komuro; Toshihide Yamashita; Toshio Hirano; Marco Prinz; Masaaki Murakami

Although pain is a common symptom of various diseases and disorders, its contribution to disease pathogenesis is not well understood. Here we show using murine experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis (MS), that pain induces EAE relapse. Mechanistic analysis showed that pain induction activates a sensory-sympathetic signal followed by a chemokine-mediated accumulation of MHC class II+CD11b+ cells that showed antigen-presentation activity at specific ventral vessels in the fifth lumbar cord of EAE-recovered mice. Following this accumulation, various immune cells including pathogenic CD4+ T cells recruited in the spinal cord in a manner dependent on a local chemokine inducer in endothelial cells, resulting in EAE relapse. Our results demonstrate that a pain-mediated neural signal can be transformed into an inflammation reaction at specific vessels to induce disease relapse, thus making this signal a potential therapeutic target. DOI: http://dx.doi.org/10.7554/eLife.08733.001


Journal of Immunology | 2015

Temporal Expression of Growth Factors Triggered by Epiregulin Regulates Inflammation Development

Masaya Harada; Daisuke Kamimura; Yasunobu Arima; Hitoshi Kohsaka; Yuji Nakatsuji; Makoto Nishida; Toru Atsumi; Jie Meng; Hidenori Bando; Rajeev Singh; Lavannya Sabharwal; Jing-Jing Jiang; Noriko Kumai; Nobuyuki Miyasaka; Saburo Sakoda; Keiko Yamauchi-Takihara; Hideki Ogura; Toshio Hirano; Masaaki Murakami

In this study, we investigated the relationship between several growth factors and inflammation development. Serum concentrations of epiregulin, amphiregulin, betacellulin, TGF-α, fibroblast growth factor 2, placental growth factor (PLGF), and tenascin C were increased in rheumatoid arthritis patients. Furthermore, local blockades of these growth factors suppressed the development of cytokine-induced arthritis in mice by inhibiting chemokine and IL-6 expressions. We found that epiregulin expression was early and followed by the induction of other growth factors at different sites of the joints. The same growth factors then regulated the expression of epiregulin at later time points of the arthritis. These growth factors were increased in patients suffering from multiple sclerosis (MS) and also played a role in the development of an MS model, experimental autoimmune encephalomyelitis. The results suggest that the temporal expression of growth factors is involved in the inflammation development seen in several diseases, including rheumatoid arthritis and MS. Therefore, various growth factor pathways might be good therapeutic targets for various inflammatory diseases.

Collaboration


Dive into the Toru Atsumi'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge