Network


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

Hotspot


Dive into the research topics where Yoshiyuki Mishima is active.

Publication


Featured researches published by Yoshiyuki Mishima.


Journal of Immunology | 2009

MFG-E8 Attenuates Intestinal Inflammation in Murine Experimental Colitis by Modulating Osteopontin-Dependent αvβ3 Integrin Signaling

Md. M. Aziz; Shunji Ishihara; Yoshiyuki Mishima; Naoki Oshima; Ichiro Moriyama; Takafumi Yuki; Yasunori Kadowaki; Mohammad Azharul Karim Rumi; Yuji Amano; Yoshikazu Kinoshita

MFG-E8 (milk fat globule-epidermal growth factor 8) deficiency is strongly associated with acquisition of immune-mediated disorders due to the loss of tissue homeostasis. However, comparatively little is known regarding its functions in gastrointestinal tract disorders, in which immune homeostasis is a major concern. Herein, we report altered MFG-E8 expression in inflamed colons during the acute phase of murine experimental colitis and found that treatment with recombinant MFG-E8, but not its arginine-glycine-aspartate mutant counterpart, ameliorated colitis by reducing inflammation and improving disease parameters. To reveal the MFG-E8-mediated antiinflammatory mechanism, we employed an in vitro system, which showed the down-regulation of NF-κB in an LPS-dependent manner. Additionally, MFG-E8 altered αvβ3 integrin-mediated focal adhesion kinase phosphorylation by impeding the binding of one of its potent ligands osteopontin, which becomes activated during colitis. Taken together, our results indicated that MFG-E8 has a novel therapeutic potential for treatment of colitis.


Clinical and Experimental Immunology | 2010

A20 is an early responding negative regulator of Toll-like receptor 5 signalling in intestinal epithelial cells during inflammation.

Naoki Oshima; Shunji Ishihara; Mohammad Rumi; Monowar Aziz; Yoshiyuki Mishima; Chikara Kadota; Ichiro Moriyama; Norihisa Ishimura; Yuji Amano; Yoshikazu Kinoshita

Several negative regulatory mechanisms control Toll‐like receptor (TLR)‐mediated inflammatory responses and restore immune system balance, including the zinc‐finger protein A20, a negative regulator of TLR signalling that inhibits nuclear factor kappa B (NF‐κB) activity. In the present study, we investigated TLR‐5‐mediated A20 expression and its role in intestinal epithelial cells (IECs) during inflammation. HCT‐15 and HT‐29 cells were stimulated with flagellin, then the expressions of A20, interleukin‐1 receptor‐associated kinase (IRAK‐M) and Tollip were evaluated using RNase protection assay. Furthermore, experimental colitis was induced in tlr4‐deficient CH3/HeJ mice by administration of dextran sodium sulphate (DSS), then flagellin was injected anally, and the colonic expression of A20 was examined by real‐time polymerase chain reaction (PCR) and immunohistochemistry. To confirm flagellin‐induced expression of A20, we employed an organ culture system. The role of A20 in flagellin‐induced tolerance induction was evaluated in vitro, using a gene knock‐down method targeting A20. A20 expression increased rapidly and peaked at 1 h after flagellin stimulation in cultured IECs, then declined gradually to the basal level. In vivo, anal injection of flagellin induced epithelial expression of A20 in injured colonic tissue, whereas flagellin did not cause a significant increase in A20 expression in non‐injured normal tissue, which was also confirmed in vitro using the organ culture system. Gene knock‐down using A20 siRNA did not influence tolerance induced by restimulation with flagellin. A20 is an early response negative regulator of TLR‐5 signalling in IECs that functions during intestinal inflammation. Our results provide new insights into the negative feedback regulation of TLR‐5 signalling that maintains the innate immune system in the gut.


Clinical and Vaccine Immunology | 2006

Epithelial toll-like receptor 5 is constitutively localized in the mouse cecum and exhibits distinctive down-regulation during experimental colitis.

Cesar F. Ortega-Cava; Shunji Ishihara; Mohammad Azharul Karim Rumi; Monowar Aziz; Hideaki Kazumori; Takafumi Yuki; Yoshiyuki Mishima; Ichiro Moriyama; Chikara Kadota; Naoki Oshima; Yuji Amano; Yasunori Kadowaki; Norihisa Ishimura; Yoshikazu Kinoshita

ABSTRACT We recently demonstrated that the pattern recognition receptors (PRRs) toll-like receptor 2 (TLR2), TLR4, and CD14 are expressed in mouse colonic epithelium in a compartmentalized manner. Here we report the localization of TLR5, the receptor for bacterial flagellin, and its distinctive down-regulation during experimental colitis. Guts from normal BALB/c mice and those with dextran sodium sulfate (DSS)-induced colitis were compared. Each gut was divided into seven segments (stomach, small intestine [three parts], and colon [three parts]), and epithelial cells and crypt units were collected by scraping and EDTA treatment, respectively. Northern blotting showed that TLR5 mRNA was preferentially expressed in the epithelium of the proximal colon in normal mice. Laser capture microdissection coupled to reverse transcriptase PCR confirmed this localization. TLR5 protein expression reflected mRNA expression, as evidenced by Western blotting. In mice with acute colitis, inflammation occurred mainly in the distal colon. Interestingly, while TLR2, TLR4, and CD14 were up-regulated in the inflamed colon, TLR5 was down-regulated at both the mRNA and protein levels. Decreased TLR5 expression was more evident during chronic colitis. Additional in vitro studies using a mouse cell line, Colon-26, showed that gamma interferon (IFN-γ) time- and dose-dependently down-regulates TLR5. In conclusion, epithelial cells, mainly in the proximal colon, constitutively express TLR5. TLR5 expression is down-regulated in vivo during acute and chronic DSS-induced colitis, in contrast to the expression of TLR2, TLR4, and CD14. The mechanism governing TLR5 regulation may therefore differ from that controlling other PRRs. Finally, IFN-γ may be involved in down-regulating TLR5 expression.


Infection and Immunity | 2014

Induction of Bacterial Antigen-Specific Colitis by a Simplified Human Microbiota Consortium in Gnotobiotic Interleukin-10−/− Mice

Chang Soo Eun; Yoshiyuki Mishima; Steffen Wohlgemuth; Bo Liu; Maureen Bower; Ian M. Carroll; R. Balfour Sartor

ABSTRACT We evaluated whether a simplified human microbiota consortium (SIHUMI) induces colitis in germfree (GF) 129S6/SvEv (129) and C57BL/6 (B6) interleukin-10-deficient (IL-10−/−) mice, determined mouse strain effects on colitis and the microbiota, examined the effects of inflammation on relative bacterial composition, and identified immunodominant bacterial species in “humanized” IL-10−/− mice. GF wild-type (WT) and IL-10−/− 129 and B6 mice were colonized with 7 human-derived inflammatory bowel disease (IBD)-related intestinal bacteria and maintained under gnotobiotic conditions. Quantification of bacteria in feces, ileal and colonic contents, and tissues was performed using 16S rRNA gene selective quantitative PCR. Colonic segments were scored histologically, and gamma interferon (IFN-γ), IL-12p40, and IL-17 levels were measured in supernatants of unstimulated colonic tissue explants and of mesenteric lymph node (MLN) cells stimulated by lysates of individual or aggregate bacterial strains. Relative bacterial species abundances changed over time and differed between 129 and B6 mice, WT and IL-10−/− mice, luminal and mucosal samples, and ileal and colonic or fecal samples. SIHUMI induced colitis in all IL-10−/− mice, with more aggressive colitis and MLN cell activation in 129 mice. Escherichia coli LF82 and Ruminococcus gnavus lysates induced dominant effector ex vivo MLN TH1 and TH17 responses, although the bacterial mucosal concentrations were low. In summary, this study shows that a simplified human bacterial consortium induces colitis in ex-GF 129 and B6 IL-10−/− mice. Relative concentrations of individual SIHUMI species are determined by host genotype, the presence of inflammation, and anatomical location. A subset of IBD-relevant human enteric bacterial species preferentially stimulates bacterial antigen-specific TH1 and TH17 immune responses in this model, independent of luminal and mucosal bacterial concentrations.


Journal of Immunology | 2012

IL-10 Regulates Il12b Expression via Histone Deacetylation: Implications for Intestinal Macrophage Homeostasis

Taku Kobayashi; Katsuyoshi Matsuoka; Shehzad Z. Sheikh; Steven Russo; Yoshiyuki Mishima; Colm B. Collins; Christopher L. Karp; Jenny P.-Y. Ting; R. Balfour Sartor; Scott E. Plevy

To prevent excessive inflammatory responses to commensal microbes, intestinal macrophages, unlike their systemic counterparts, do not produce inflammatory cytokines in response to enteric bacteria. Consequently, loss of macrophage tolerance to the enteric microbiota plays a central role in the pathogenesis of inflammatory bowel diseases. Therefore, we examined whether the hyporesponsive phenotype of intestinal macrophages is programmed by prior exposure to the microbiota. IL-10, but not in vivo exposure to the microbiota, programs intestinal macrophage tolerance, because wild-type (WT) colonic macrophages from germ-free and specific pathogen-free (SPF)-derived mice produce IL-10, but not IL-12 p40, when activated with enteric bacteria. Basal and activated IL-10 expression is mediated through a MyD88-dependent pathway. Conversely, colonic macrophages from germ-free and SPF-derived colitis-prone Il10−/− mice demonstrated robust production of IL-12 p40. Next, mechanisms through which IL-10 inhibits Il12b expression were investigated. Although Il12b mRNA was transiently induced in LPS-activated WT bone marrow-derived macrophages (BMDMs), expression persisted in Il10−/− BMDMs. There were no differences in nucleosome remodeling, mRNA stability, NF-κB activation, or MAPK signaling to explain prolonged transcription of Il12b in Il10−/− BMDMs. However, acetylated histone H4 transiently associated with the Il12b promoter in WT BMDMs, whereas association of these factors was prolonged in Il10−/− BMDMs. Experiments using histone deacetylase (HDAC) inhibitors and HDAC3 short hairpin RNA indicate that HDAC3 is involved in histone deacetylation of the Il12b promoter by IL-10. These results suggest that histone deacetylation on the Il12b promoter by HDAC3 mediates homeostatic effects of IL-10 in macrophages.


Clinical and Experimental Immunology | 2010

Down-regulation of single immunoglobulin interleukin-1R-related molecule (SIGIRR)/TIR8 expression in intestinal epithelial cells during inflammation

Chikara Kadota; Shunji Ishihara; Monowar Aziz; Mohammad Rumi; Naoki Oshima; Yoshiyuki Mishima; Ichiro Moriyama; Takafumi Yuki; Yuji Amano; Yoshikazu Kinoshita

Single immunoglobulin (Ig) interleukin‐1R‐related molecule (SIGIRR) is an Ig‐like membrane protein critical for negative regulation of Toll‐like receptor (TLR)‐4‐mediated signalling. We investigated SIGIRR expression and its regulation mechanism in intestinal epithelial cells (IECs) during inflammation. Endoscopic biopsy specimens were obtained from active and inactive colonic mucosa of ulcerative colitis (UC) patients, then SIGIRR expression was examined using real‐time polymerase chain reaction (PCR) and immunohistochemistry (IH). Mice experimental colitis models were established by administrations of sulphonic acid (TNBS) and dextran sodium sulphate (DSS), and epithelial expression of SIGIRR was examined using real‐time PCR, IH and flow cytometry. The effects of lipopolysaccharide (LPS) and tumour necrosis factor (TNF)‐α on SIGIRR expression were evaluated in vitro using cultured IECs. To elucidate SIGIRR expression regulation in IECs, binding ability of the transcription factor SP1 at the responsive element of the SIGIRR promoter was examined using gel‐shift and chromatin immunoprecipitation (ChIP) assays. In human colonic samples, SIGIRR was expressed mainly in IECs at levels significantly higher in inactive compared to active mucosa. In the mice, SIGIRR colonic expression decreased rapidly after colitis development and returned gradually to basal levels. Experimental colitis‐mediated down‐regulation of SIGIRR in IECs was also confirmed by IH and flow cytometry results. Further, inflammatory conditions induced by TLR ligands and TNF‐α caused significant down‐regulation of SIGIRR expression in IECs, which was dependent upon decreased SP1 binding at the responsive element of the SIGIRR promoter. We found that SIGIRR is expressed in IECs and serves as a negative regulator to maintain gut innate immunity, which is down‐regulated during inflammation by inhibition of an SP1‐mediated pathway.


Digestion | 2013

Pathogenesis of Irritable Bowel Syndrome - Review Regarding Associated Infection and Immune Activation

Shunji Ishihara; Yasumasa Tada; Nobuhiko Fukuba; Akihiko Oka; Ryusaku Kusunoki; Yoshiyuki Mishima; Naoki Oshima; Ichiro Moriyama; Takafumi Yuki; Kousaku Kawashima; Yoshikazu Kinoshita

There is increasing evidence regarding the role of immune activation in the etiology of irritable bowel syndrome (IBS), which has been mainly been shown in studies investigating mechanisms of postinfectious IBS (PI-IBS). Exposure to intestinal infection induces persistent low-grade systemic and mucosal inflammation, which is characterized by an altered population of circulating cells, mucosal infiltration of immune cells and increased production of various cytokines in IBS patients. Recent studies have also indicated an increased innate immune response in these patients by evaluating expression and activation of Toll-like receptors (TLRs). These findings suggest that immune activation may play a crucial role in the pathogenesis of IBS. In addition, psychological stress has been reported to be one of the factors that induces immune activation. However, it remains unknown whether immune activation in IBS patients is largely dependent on infectious gastroenteritis and/or psychological stress. Additional studies are necessary to understand the precise mechanism of immune activation and its relationship to the development of IBS.


Laboratory Investigation | 2008

Decoy oligodeoxynucleotide targeting activator protein-1 (AP-1) attenuates intestinal inflammation in murine experimental colitis

Ichiro Moriyama; Shunji Ishihara; M Azharul Karim Rumi; Monowar Aziz; Yoshiyuki Mishima; Naoki Oshima; Chikara Kadota; Yasunori Kadowaki; Yuji Amano; Yoshikazu Kinoshita

Various therapies are used for inflammatory bowel diseases (IBD), though none seem to be extremely effective. AP-1 is a major transcription factor that upregulates genes involved in immune and proinflammatory responses. We investigated decoy oligodeoxynucleotide (ODN) targeting AP-1 to prevent dextran sulfate sodium (DSS)-induced colitis in mice. Functional efficacies of synthetic decoy and scrambled ODNs were evaluated in vitro by a reporter gene luciferase assay and measuring flagellin-induced IL-8 expression by HCT-15 cells transfected with ODNs. Experimental colitis was induced in mice with a 2.5% DSS solution in drinking water for 7 days, and decoy or scrambled ODNs were intraperitoneally injected from days 2 to 5. Colitis was assessed by weight loss, colon length, histopathology, and detection of myeloperoxidase (MPO), IL-1β, and TNF-α in colon tissue. Therapeutic effects of AP-1 and NF-κB decoy ODNs were compared. Transfection of AP-1 decoy ODN inhibited AP-1 transcriptional activity in reporter assays and flagellin-induced IL-8 production in vitro. In mice, AP-1 decoy ODN, but not scrambled ODN, significantly inhibited weight loss, colon shortening, and histological inflammation induced by DSS. Further, AP-1 decoy ODN decreased MPO, IL-1β, and TNF-α in colonic tissue of mice with DSS-induced colitis. The AP-1 decoy therapeutic effect was comparable to that of NF-κB decoy ODN, which also significantly decreased intestinal inflammation. Double-strand decoy ODN targeting AP-1 effectively attenuated intestinal inflammation associated with experimental colitis in mice, indicating the potential of targeting proinflammatory transcription factors in new therapies for IBD.


Apoptosis | 2008

Prolactin induces MFG-E8 production in macrophages via transcription factor C/EBPβ-dependent pathway

Md. M. Aziz; Shunji Ishihara; Mohammad Rumi; Yoshiyuki Mishima; Naoki Oshima; Chikara Kadota; Ichiro Moriyama; Yong-Yu Li; Farzana Binte Rahman; Aya Otani; Akihiko Oka; Norihisa Ishimura; Yasunori Kadowaki; Yuji Amano; Yoshikazu Kinoshita

The lactogenic hormone prolactin (PRL) regulates milk protein gene expression in mammary glands. To maintain homeostatic balance in the body, milk fat globule epidermal growth factor 8 (MFG-E8) is vital for phagocytic clearance of apoptotic cells. We investigated the effects of PRL on MFG-E8 expression in macrophages by evaluating its promoter function. Macrophages were stimulated with PRL, and the expression of MFG-E8 was determined using real-time PCR and Western blotting. The role of MFG-E8 on phagocytosis of apoptotic cells in PRL-treated macrophages was assessed using microscopy, while the response of PRL to MFG-E8 expression was evaluated using luciferase assay. Following treatment with PRL, significant up-regulations of the PRL receptor and MFG-E8 were observed in macrophages, though PRL-treated macrophages more efficiently engulfed apoptotic cells. The results of MFG-E8 promoter analysis showed considerable up-regulation of promoter activity in macrophages following PRL treatment and results from mutation analysis of the MFG-E8 promoter suggested that the C/EBPβ binding site was responsible for PRL-induced activation of the MFG-E8 promoter. C/EBPβ activity was found to be up-regulated in PRL-treated cells as revealed by an electrophoretic mobility shift assay (EMSA). In conclusion, PRL is a potent inducer of MFG-E8 expression in macrophages, while its effect is mediated by the presence of a responsive element in the MFG-E8 promoter.


Inflammatory Bowel Diseases | 2014

Role of regulatory B cells in chronic intestinal inflammation: association with pathogenesis of Crohn's disease.

Akihiko Oka; Shunji Ishihara; Yoshiyuki Mishima; Yasumasa Tada; Ryusaku Kusunoki; Nobuhiko Fukuba; Takafumi Yuki; Kousaku Kawashima; Satoshi Matsumoto; Yoshikazu Kinoshita

Abstract:The role of regulatory B cells (Bregs) producing interleukin (IL)-10 in the pathogenesis of inflammatory bowel diseases remains unknown. We investigated IL-10 production in B cells from patients with inflammatory bowel diseases and immunoregulatory functions of Bregs in experimental colitis mouse models. CpG DNA-induced IL-10 production in peripheral blood B cells isolated from patients with inflammatory bowel diseases and control subjects was examined. CD19 and CD1d were used for evaluating possible cell surface markers of Bregs. Colitis models of severe combined immunodeficiency mice were established by adoptive transfer of whole CD4+ T cells or regulatory T cell (Treg)-depleted T cells (CD4+CD25−) isolated from SAMP1/Yit mice and the function of Bregs in intestinal inflammation was elucidated by evaluating the effects of cotransfer of whole or Breg-depleted B cells. CpG DNA-induced IL-10 production was significantly decreased in B cells from patients with Crohns disease (CD), as compared with those from healthy controls, whereas Bregs were found to be enriched in a population of CD19hi and CD1dhi B cells isolated from both human and mouse samples. The severity of intestinal inflammation was significantly increased in the Breg-depleted mice, with similar results also found in adoptive transfer colitis model mice even after Treg depletion. Our findings show that Bregs, characterized by the cell surface markers CD19hi and CD1dhi, significantly reduced experimental colitis regardless of the presence or absence of Tregs. These results suggest that a deficiency or decrease of Bregs function exacerbates intestinal inflammation, which may be associated with the pathogenesis of CD.

Collaboration


Dive into the Yoshiyuki Mishima'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

Bo Liu

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge