Network


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

Hotspot


Dive into the research topics where Satoshi Matsumoto is active.

Publication


Featured researches published by Satoshi Matsumoto.


Gut | 1998

Inflammatory bowel disease-like enteritis and caecitis in a senescence accelerated mouse P1/Yit strain

Satoshi Matsumoto; Y Okabe; H Setoyama; K Takayama; J Ohtsuka; H Funahashi; A Imaoka; Y Okada; Y Umesaki

Background—A new subline of the senescence accelerated mouse (SAM) P1/Yit strain has been established which shows spontaneous enteric inflammation under specific pathogen free (SPF) conditions. Aims—To elucidate the pathogenesis of enteric inflammation in this new subline. Methods—The SPF and germ free (GF) SAMP1/Yit strains were used. Histological, immunological, and microbiological characterisation of the mice with enteric inflammation was performed. Results—Histologically, enteritic inflammation developed as a discontinuous lesion in the terminal ileum and caecum with the infiltration of many inflammatory cells after 10 weeks of age. The activity of myeloperoxidase, and both immunolocalisation and mRNA expression of inducible nitric oxide synthase increased in the lesion. CD3-ε positive T cells, neutrophils, and macrophages were more numerous in the inflamed mucosa of the SAMP1/Yit strain. The GF SAMP1/Yit strain did not show any inflammation in the intestinal wall, by the age of 30 weeks, and the enteritis and caecitis developed 10 weeks after the conventionalisation of the GF SAMP1/Yit strain. Conclusion—Enteric inflammation in the ileum and caecum developed in the SAMP1/Yit strain. The pathophysiological characteristics of the disease in this mouse have some similarities to those of human inflammatory bowel disease (IBD). This mouse strain should be a useful model system for elucidating the interaction between the pathogenesis of IBD and the gut microflora.


Journal of Immunology | 2010

Essential Roles of IL-6 Trans-Signaling in Colonic Epithelial Cells, Induced by the IL-6/Soluble–IL-6 Receptor Derived from Lamina Propria Macrophages, on the Development of Colitis-Associated Premalignant Cancer in a Murine Model

Satoshi Matsumoto; Taeko Hara; Keiichi Mitsuyama; Mayuko Yamamoto; Osamu Tsuruta; Michio Sata; Jürgen Scheller; Stefan Rose-John; Shoichi Kado; Toshihiko Takada

Activation of the IL-6/Stat3 via IL-6 trans-signaling plays an important role in the pathogenesis of inflammatory bowel disease. Colitis-associated cancer (CAC) is a large bowel cancer and occurs with long-standing inflammatory bowel disease. The role of the IL-6/Stat3 in the development of CAC has not been fully understood. We investigate whether IL-6 trans-signaling contributes to the development of CAC using a mouse colitis-associated premalignant cancer (CApC) model. Chronic colitis (CC) was induced in BALB/c mice using dextran sodium sulfate. CApC was induced by dextran sodium sulfate treatment to CC-affected mice. IL-6 expression was determined by quantitative RT-PCR and immunofluorescence staining in colon. Phospho-Stat3 expression was examined by Western blotting and immunofluorescence analysis. The expression of IL-6 receptors (i.e., the IL-6R α-chain and gp130) and tumor necrosis factor-α converting enzyme in the colon was examined by laser-capture microdissection and immunofluorescence staining. Soluble IL-6Rα (sIL-6Rα) was examined by Western blotting of epithelial cell-depleted colonic tissues. We also investigated whether a soluble gp130-Fc fusion protein could prevent CApC. IL-6 expression was increased in the colon of CC- and CApC-affected mice and was restricted to lamina propria-macrophages. The expression of IL-6Rα and tumor necrosis factor-α converting enzyme was increased in the lamina propria CD11b-macrophages of CC-affected mice. sIL-6Rα expression was also increased in these tissues. Reduced levels of IL-6Rα generation were observed in the colonic epithelial cells of CC- and CApC-affected mice and were associated with the increased expression of gp130 and phospho-Stat3. Treatment with soluble gp130Fc significantly reduced the CApC. IL-6 trans-signaling in epithelial cells induced by macrophage-derived IL-6/sIL-6Rα plays a crucial role in the development of CAC.


Gut | 2006

STAT3 activation via interleukin 6 trans-signalling contributes to ileitis in SAMP1/Yit mice

Keiichi Mitsuyama; Satoshi Matsumoto; Stefan Rose-John; Asuka Suzuki; Taeko Hara; Nobuo Tomiyasu; Kazuyo Handa; Osamu Tsuruta; Hideyuki Funabashi; Jürgen Scheller; Atsushi Toyonaga; Michio Sata

Background and aim: SAMP1/Yit mice spontaneously develops intestinal inflammation. Previously, we demonstrated that the signal transducer and activator of transcription (STAT)-3/suppressor of cytokine signalling (SOCS)-3 pathway is pivotal in human inflammatory bowel disease. In our studies in SAMP1/Yit mice, the aim was to investigate whether STAT3 activation contributes to ileitis and to examine the therapeutic effects of this signal blockade. Methods: Intestinal expression of phospho-STAT3 in SAMP1/Yit mice and control AKR/J mice was examined by western blotting and immunohistochemistry. SOCS3 and interleukin 6 (IL-6) mRNA were determined by northern blotting and reverse transcription-polymerase chain reaction, respectively. We also examined the effects of intravenously injected hyper-IL-6, an IL-6/soluble IL-6 receptor fusion protein, and of soluble gp130-Fc, a specific inhibitor of soluble IL-6 receptor signalling, on STAT3 phosphorylation and disease severity in SAMP1/Yit mice. Results: Phospho-STAT3 was expressed strongly during the disease course in SAMP1/Yit mice but only transiently in AKR/J mice. Phospho-STAT3 was localised to epithelial and mononuclear cells in the diseased intestine of SAMP1/Yit mice. SOCS3 as well as IL-6 mRNAs were expressed in affected intestine. Administration of hyper-IL-6 caused disease exacerbation and enhancement of STAT3 phosphorylation. In contrast, soluble gp130-Fc administration ameliorated the disease and suppressed STAT3 phosphorylation. Conclusion: STAT3 signalling is critical in the development of intestinal inflammation in SAMP1/Yit mice. Blockade of this signalling pathway by soluble gp130-Fc may have therapeutic effects in inflammatory bowel disease.


Current Pharmaceutical Design | 2009

Interleukin-6 trans-signaling and colonic cancer associated with inflammatory bowel disease.

Stefan Rose-John; Keiichi Mitsuyama; Satoshi Matsumoto; Wolfgang Thaiss; Jürgen Scheller

The complex pathogenesis of inflammatory bowel disease (IBD) and inflammation induced colon cancer involves a wide range of mediators including cytokines. Recent investigations underline the fundamental role of interleukin-6 (IL-6) signaling in the development and maintenance of IBD and for the progression of this inflammation to colon cancer. The molecular mechanisms of this pathway, the source of the cytokine and the identity of target cells in the intestine are incompletely understood. It is known that the circulating and intestinal levels of IL-6 as well as the soluble IL-6 receptor (sIL-6R) are increased in patients with IBD. Remarkably, mucosal T cells of IBD patients are extremely resistant to apoptosis and a large fraction of these cells express membrane-bound gp130 but not the IL-6R. Increasing evidence suggests that the development and perpetuation of IBD relies on IL-6 synthesized by T-cells and myeloid cells and on increased formation of IL-6/sIL-6R complexes interacting with membrane-bound gp130 on T-cells, a process called IL-6 trans-signaling. Recent investigations suggested a protective role of IL-6 mediated STAT3 signaling in intestinal epithelial cells. Here we review these pro- and anti-inflammatory properties of IL-6 in IBD and inflammation induced colon cancer and we will summarize the consequences of these new results for the prospects of IL-6 targeted therapeutic strategies for the treatment of chronic inflammatory diseases such as IBD.


Nature Communications | 2016

A key genetic factor for fucosyllactose utilization affects infant gut microbiota development

Takahiro Matsuki; Kana Yahagi; Hiroshi Mori; Hoshitaka Matsumoto; Taeko Hara; Saya Tajima; Eishin Ogawa; Hiroko Kodama; Kazuya Yamamoto; Takuji Yamada; Satoshi Matsumoto; Ken Kurokawa

Recent studies have demonstrated that gut microbiota development influences infants health and subsequent host physiology. However, the factors shaping the development of the microbiota remain poorly understood, and the mechanisms through which these factors affect gut metabolite profiles have not been extensively investigated. Here we analyse gut microbiota development of 27 infants during the first month of life. We find three distinct clusters that transition towards Bifidobacteriaceae-dominant microbiota. We observe considerable differences in human milk oligosaccharide utilization among infant bifidobacteria. Colonization of fucosyllactose (FL)-utilizing bifidobacteria is associated with altered metabolite profiles and microbiota compositions, which have been previously shown to affect infant health. Genome analysis of infants bifidobacteria reveals an ABC transporter as a key genetic factor for FL utilization. Thus, the ability of bifidobacteria to utilize FL and the presence of FL in breast milk may affect the development of the gut microbiota in infants, and might ultimately have therapeutic implications.


Gastroenterology | 1992

Differential induction of major histocompatibility complex molecules on mouse intestine by bacterial colonization

Satoshi Matsumoto; Hiromi Setoyama; Yoshimori Umesaki

The aim of this study was to determine what factors induce major histocompatibility complex (MHC) molecules on the mouse small intestinal epithelium by using immunohistochemical methods. In germ-free mice, although MHC class I molecules such as H-2K and thymus leukemia antigen (TLa) were expressed on the small intestinal epithelium, class II molecules were absent. The introduction of microorganisms into germ-free mice induced characteristic MHC molecules on the small intestinal epithelial cells. The I-A molecule was induced on the villus tip and crypt epithelial cells 7 days after conventionalization, and the I-E molecule was induced on the mid villus and crypt epithelial cells 14 days after conventionalization. The staining intensity of the H-2K molecules was increased 4 days after conventionalization. In contrast, TLa did not change during conventionalization of germ-free mice. These results suggest that the expression of MHC molecules, except for the TLa, is greatly dependent on the presence of intestinal microorganisms.


Journal of Immunology | 2009

Long-Lived Colitogenic CD4+ Memory T Cells Residing Outside the Intestine Participate in the Perpetuation of Chronic Colitis

Yasuhiro Nemoto; Takanori Kanai; Kaori Kameyama; Tamako Shinohara; Naoya Sakamoto; Teruji Totsuka; Ryuichi Okamoto; Kiichiro Tsuchiya; Tetsuya Nakamura; Tetsuo Sudo; Satoshi Matsumoto; Mamoru Watanabe

To understand the perpetuation of inflammatory bowel disease (IBD), it is important to clarify whether the colitogenic CD4+ T cells are self-limited effector or long-lived memory T cells. We here investigate the latency of colitogenic CD4+ T cells in the remission stage of colitis under germfree (GF) conditions. We isolated splenic (SP) CD4+ T cells from colitic CD4+CD45RBhigh T cell-injected SCID mice maintained under specific pathogen-free (SPF) conditions and transferred them into SPF or GF SCID mice. Donor colitic SP CD4+ T cells have a characteristic CD44highCD62L−IL-7Rαhigh effector-memory T-type phenotype. Six weeks after transfer of cells to GF SCID mice, one group of mice was continued in GF conditions (GF→GF), and the other was transferred into SPF conditions (GF→SPF). GF→SPF but not GF→GF SCID mice developed colitis with elevated production of Th1 and Th17 cytokines at 4 wk after transfer. Surprisingly, a large number of CD4+ effector-memory T cells and a small but substantial number of central-memory T cells remained resident in SP and bone marrow, but not in lamina propria, of the GF→GF SCID recipients. Consistent with this, GF→SPF but not GF→GF SCID mice rapidly developed colitis. Taken together, these findings suggest that long-lived colitogenic memory CD4+ cells can be established even in the presence of commensal Ags, reside outside the intestine in the absence of commensal bacteria, and participate in the perpetuation of colitis. Thus, blocking a stimulus of colitogenic memory CD4+ cells such as IL-7 may have therapeutic benefit for treatment of inflammatory bowel disease.


Nature microbiology | 2016

High-affinity monoclonal IgA regulates gut microbiota and prevents colitis in mice.

Shinsaku Okai; Fumihito Usui; Shuhei Yokota; Yusaku Hori-i; Makoto Hasegawa; Toshinobu Nakamura; Manabu Kurosawa; Seiji Okada; Kazuya Yamamoto; Eri Nishiyama; Hiroshi Mori; Takuji Yamada; Ken Kurokawa; Satoshi Matsumoto; Masanobu Nanno; Tomoaki Naito; Yohei Watanabe; Tamotsu Kato; Eiji Miyauchi; Hiroshi Ohno; Reiko Shinkura

Immunoglobulin A (IgA) is the main antibody isotype secreted into the intestinal lumen. IgA plays a critical role in the defence against pathogens and in the maintenance of intestinal homeostasis. However, how secreted IgA regulates gut microbiota is not completely understood. In this study, we isolated monoclonal IgA antibodies from the small intestine of healthy mouse. As a candidate for an efficient gut microbiota modulator, we selected a W27 IgA, which binds to multiple bacteria, but not beneficial ones such as Lactobacillus casei. W27 could suppress the cell growth of Escherichia coli but not L. casei in vitro, indicating an ability to improve the intestinal environment. Indeed W27 oral treatment could modulate gut microbiota composition and have a therapeutic effect on both lymphoproliferative disease and colitis models in mice. Thus, W27 IgA oral treatment is a potential remedy for inflammatory bowel disease, acting through restoration of host–microbial symbiosis.


Journal of Immunology | 2013

Th1/Th17-Mediated Interstitial Pneumonia in Chronic Colitis Mice Independent of Intestinal Microbiota

Yasuhiro Nemoto; Takanori Kanai; Masahiro Takahara; Shigeru Oshima; Ryuichi Okamoto; Kiichiro Tsuchiya; Satoshi Matsumoto; Mamoru Watanabe

Although intestinal microbiota are essential for the development of T cell–mediated colitis, it remains undetermined whether they enhance or suppress the chronic extraintestinal inflammation that often complicates inflammatory bowel diseases. In this study, we investigate the role of intestinal microbiota in the development of colitis and extraintestinal manifestations in a mouse model in which colitis was induced in SCID mice by adoptive transfer of CD4+CD45RBhigh T cells. Under specific pathogen-free conditions, these mice developed both colitis and extraintestinal interstitial pneumonia, whereas mice given a mixture of antibiotics did not develop colitis, but, surprisingly, developed Th1/Th17-mediated IP. Irrespective of antibiotic treatment, cotransfer of CD4+CD25+ regulatory T cells suppressed the development of pneumonitis and colitis, with all local CD4+CD45RBhigh T cell–derived cells converted to CD44highCD62L−IL-7Rαhigh effector–memory T cells. Retransfer of CD4+ effector–memory T cells from the lungs of antibiotic-treated mice with IP not only induced IP in both antibiotic-treated and -untreated recipients but also induced colitis in the untreated recipients. In summary, we have established a unique model of Th1/Th17-mediated IP in microbiota-free and antibiotic-treated mice. This model may be valuable in investigating the immunological mechanisms underlying extraintestinal disorders in patients with inflammatory bowel disease.


Gut microbes | 2017

Intestinal IgA as a modulator of the gut microbiota

Shinsaku Okai; Fumihito Usui; Misa Ohta; Hiroshi Mori; Ken Kurokawa; Satoshi Matsumoto; Tamotsu Kato; Eiji Miyauchi; Hiroshi Ohno; Reiko Shinkura

ABSTRACT Accumulating evidence suggests that dysbiosis plays a role in the pathogenesis of intestinal diseases including inflammatory bowel disease (IBD) as well as extra-intestinal disorders. As a modulator of the intestinal microbiota, we isolated a mouse monoclonal IgA antibody (clone W27) with high affinities for multiple commensal bacteria, but not for beneficial bacteria such as Lactobacillus casei (L. casei). Via specific recognition of an epitope in serine hydroxymethyltransferase (SHMT), a bacterial metabolic enzyme, W27 IgA selectively inhibited the in vitro growth of bound bacteria, including Escherichia coli (E. coli), while having no effect on unbound beneficial bacteria such as L. casei. By modulating the gut microbiota in vivo, oral administration of W27 IgA effectively prevented development of colitis in several mouse models. Here we discuss how intestinal IgA modulates the gut microbiota through recognition of SHMT.

Collaboration


Dive into the Satoshi Matsumoto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroshi Mori

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Ken Kurokawa

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fumihito Usui

Nagahama Institute of Bio-Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge