Network


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

Hotspot


Dive into the research topics where Yasuhiro Nemoto is active.

Publication


Featured researches published by Yasuhiro Nemoto.


Nature Medicine | 2012

Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5+ stem cell

Shiro Yui; Tetsuya Nakamura; Toshiro Sato; Yasuhiro Nemoto; Tomohiro Mizutani; Xiu Zheng; Shizuko Ichinose; Takashi Nagaishi; Ryuichi Okamoto; Kiichiro Tsuchiya; Hans Clevers; Mamoru Watanabe

Adult stem-cell therapy holds promise for the treatment of gastrointestinal diseases. Here we describe methods for long-term expansion of colonic stem cells positive for leucine-rich repeat containing G protein-coupled receptor 5 (Lgr5+ cells) in culture. To test the transplantability of these cells, we reintroduced cultured GFP+ colon organoids into superficially damaged mouse colon. The transplanted donor cells readily integrated into the mouse colon, covering the area that lacked epithelium as a result of the introduced damage in recipient mice. At 4 weeks after transplantation, the donor-derived cells constituted a single-layered epithelium, which formed self-renewing crypts that were functionally and histologically normal. Moreover, we observed long-term (>6 months) engraftment with transplantation of organoids derived from a single Lgr5+ colon stem cell after extensive in vitro expansion. These data show the feasibility of colon stem-cell therapy based on the in vitro expansion of a single adult colonic stem cell.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2009

Requirement of Notch activation during regeneration of the intestinal epithelia

Ryuichi Okamoto; Kiichiro Tsuchiya; Yasuhiro Nemoto; Junko Akiyama; Tetsuya Nakamura; Takanori Kanai; Mamoru Watanabe

Notch signaling regulates cell differentiation and proliferation, contributing to the maintenance of diverse tissues including the intestinal epithelia. However, its role in tissue regeneration is less understood. Here, we show that Notch signaling is activated in a greater number of intestinal epithelial cells in the inflamed mucosa of colitis. Inhibition of Notch activation in vivo using a gamma-secretase inhibitor resulted in a severe exacerbation of the colitis attributable to the loss of the regenerative response within the epithelial layer. Activation of Notch supported epithelial regeneration by suppressing goblet cell differentiation, but it also promoted cell proliferation, as shown in in vivo and in vitro studies. By utilizing tetracycline-dependent gene expression and microarray analysis, we identified a novel group of genes that are regulated downstream of Notch1 within intestinal epithelial cells, including PLA2G2A, an antimicrobial peptide secreted by Paneth cells. Finally, we show that these functions of activated Notch1 are present in the mucosa of ulcerative colitis, mediating cell proliferation, goblet cell depletion, and ectopic expression of PLA2G2A, thereby contributing to the regeneration of the damaged epithelia. This study showed the critical involvement of Notch signaling during intestinal tissue regeneration, regulating differentiation, proliferation, and antimicrobial response of the epithelial cells. Thus Notch signaling is a key intracellular molecular pathway for the proper reconstruction of the intestinal epithelia.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2009

Signaling pathway via TNF-α/NF-κB in intestinal epithelial cells may be directly involved in colitis-associated carcinogenesis

Michio Onizawa; Takashi Nagaishi; Takanori Kanai; Ken Ichi Nagano; Shigeru Oshima; Yasuhiro Nemoto; Atsushi Yoshioka; Teruji Totsuka; Ryuichi Okamoto; Tetsuya Nakamura; Naoya Sakamoto; Kiichiro Tsuchiya; Kazuhiro Aoki; Keiichi Ohya; Hideo Yagita; Mamoru Watanabe

Treatment with anti-TNF-alpha MAb has been accepted as a successful maintenance therapy for patients with inflammatory bowel diseases (IBD). Moreover, it has been recently reported that blockade of TNF receptor (TNFR) 1 signaling in infiltrating hematopoietic cells may prevent the development of colitis-associated cancer (CAC). However, it remains unclear whether the TNF-alpha signaling in epithelial cells is involved in the development of CAC. To investigate this, we studied the effects of anti-TNF-alpha MAb in an animal model of CAC by administration of azoxymethane (AOM) followed by sequential dextran sodium sulfate (DSS) ingestion. We observed that the NF-kappaB pathway is activated in colonic epithelia from DSS-administered mice in association with upregulation of TNFR2 rather than TNFR1. Immunoblot analysis also revealed that the TNFR2 upregulation accompanied by the NF-kappaB activation is further complicated in CAC tissues induced in AOM/DSS-administered mice compared with the nontumor area. Such NF-kappaB activity in the epithelial cells is significantly suppressed by the treatment of MP6-XT22, an anti-TNF-alpha MAb. Despite inability to reduce the severity of colitis, sequential administration of MP6-XT22 reduced the numbers and size of tumors in association with the NF-kappaB inactivation. Taken together, present studies suggest that the TNFR2 signaling in intestinal epithelial cells may be directly involved in the development of CAC with persistent colitis and imply that the maintenance therapy with anti-TNF-alpha MAb may prevent the development of CAC in patients with long-standing IBD.


Journal of Immunology | 2007

Intestinal Lamina Propria Retaining CD4+CD25+ Regulatory T Cells Is A Suppressive Site of Intestinal Inflammation

Shin Makita; Takanori Kanai; Yasuhiro Nemoto; Teruji Totsuka; Ryuichi Okamoto; Kiichiro Tsuchiya; Masafumi Yamamoto; Hiroshi Kiyono; Mamoru Watanabe

It is well known that immune responses in the intestine remain in a state of controlled inflammation, suggesting that not only does active suppression by regulatory T (TREG) cells play an important role in the normal intestinal homeostasis, but also that its dysregulation of immune response leads to the development of inflammatory bowel disease. In this study, we demonstrate that murine CD4+CD25+ T cells residing in the intestinal lamina propria (LP) constitutively express CTLA-4, glucocorticoid-induced TNFR, and Foxp3 and suppress proliferation of responder CD4+ T cells in vitro. Furthermore, cotransfer of intestinal LP CD4+CD25+ T cells prevents the development of chronic colitis induced by adoptive transfer of CD4+CD45RBhigh T cells into SCID mice. When lymphotoxin (LT)α-deficient intercrossed Rag2 double knockout mice (LTα−/− × Rag2−/−), which lack mesenteric lymph nodes and Peyer’s patches, are transferred with CD4+CD45RBhigh T cells, they develop severe wasting disease and chronic colitis despite the delayed kinetics as compared with the control LTα+/+ × Rag2−/− mice transferred with CD4+CD45RBhigh T cells. Of note, when a mixture of splenic CD4+CD25+ TREG cells and CD4+CD45RBhigh T cells are transferred into LTα−/− × Rag2−/− recipients, CD4+CD25+ TREG cells migrate into the colon and prevent the development of colitis in LTα−/− × Rag2−/− recipients as well as in the control LTα+/+ × Rag2−/− recipients. These results suggest that the intestinal LP harboring CD4+CD25+ TREG cells contributes to the intestinal immune suppression.


Journal of Immunology | 2007

IL-7 Is Essential for the Development and the Persistence of Chronic Colitis

Teruji Totsuka; Takanori Kanai; Yasuhiro Nemoto; Shin Makita; Ryuichi Okamoto; Kiichiro Tsuchiya; Mamoru Watanabe

Although IL-7 has recently emerged as a key cytokine involved in controlling the homeostatic turnover and the survival of peripheral resting memory CD4+ T cells, its potential to be sustained pathogenic CD4+ T cells in chronic immune diseases, such as inflammatory bowel diseases, still remains unclear. In this study, we demonstrate that IL-7 is essential for the development and the persistence of chronic colitis induced by adoptive transfer of normal CD4+CD45RBhigh T cells or colitogenic lamina propria (LP) CD4+ memory T cells into immunodeficient IL-7+/+ × RAG-1−/− and IL-7−/− × RAG-1−/− mice. Although IL-7+/+ × RAG-1−/− recipients transferred with CD4+CD45RBhigh splenocytes developed massive inflammation of the large intestinal mucosa concurrent with massive expansion of Th1 cells, IL-7−/− × RAG-1−/− recipients did not. Furthermore, IL-7−/− × RAG-1−/−, but not IL-7+/+ × RAG-1−/−, mice transferred with LP CD4+CD44highCD62L−IL-7Rαhigh effector-memory T cells (TEM) isolated from colitic CD4+CD45RBhigh-transferred mice did not develop colitis. Although rapid proliferation of transferred colitogenic LP CD4+ TEM cells was observed in the in IL-7−/− × RAG-1−/− mice to a similar extent of those in IL-7+/+ × RAG-1−/− mice, Bcl-2 expression was significantly down-modulated in the transferred CD4+ T cells in IL-7−/− × RAG-1−/− mice compared with those in IL-7+/+ × RAG-1−/− mice. Taken together, IL-7 is essential for the development and the persistence of chronic colitis as a critical survival factor for colitogenic CD4+ TEM cells, suggesting that therapeutic approaches targeting IL-7/IL-7R signaling pathway may be feasible in the treatment of inflammatory bowel diseases.


Journal of Immunology | 2008

MyD88-Dependent Pathway in T Cells Directly Modulates the Expansion of Colitogenic CD4+ T Cells in Chronic Colitis

Takayuki Tomita; Takanori Kanai; Toshimitsu Fujii; Yasuhiro Nemoto; Ryuichi Okamoto; Kiichiro Tsuchiya; Teruji Totsuka; Naoya Sakamoto; Shizuo Akira; Mamoru Watanabe

TLRs that mediate the recognition of pathogen-associated molecular patterns are widely expressed on/in cells of the innate immune system. However, recent findings demonstrate that certain TLRs are also expressed in conventional TCRαβ+ T cells that are critically involved in the acquired immune system, suggesting that TLR ligands can directly modulate T cell function in addition to various innate immune cells. In this study, we report that in a murine model of chronic colitis induced in RAG-2−/− mice by adoptive transfer of CD4+CD45RBhigh T cells, both CD4+CD45RBhigh donor cells and the expanding colitogenic lamina propria CD4+CD44high memory cells expresses a wide variety of TLRs along with MyD88, a key adaptor molecule required for signal transduction through TLRs. Although RAG-2−/− mice transferred with MyD88−/−CD4+CD45RBhigh cells developed colitis, the severity was reduced with the delayed kinetics of clinical course, and the expansion of colitogenic CD4+ T cells was significantly impaired as compared with control mice transferred with MyD88+/+CD4+CD45RBhigh cells. When RAG-2−/− mice were transferred with the same number of MyD88+/+ (Ly5.1+) and MyD88−/− (Ly5.2+) CD4+CD45RBhigh cells, MyD88−/−CD4+ T cells showed significantly lower proliferative responses assessed by in vivo CFSE division assay, and also lower expression of antiapoptotic Bcl-2/Bcl-xL molecules and less production of IFN-γ and IL-17, compared with the paired MyD88+/+CD4+ T cells. Collectively, the MyD88-dependent pathway that controls TLR signaling in T cells may directly promote the proliferation and survival of colitogenic CD4+ T cells to sustain chronic colitis.


Journal of Immunology | 2009

Rank-rankl signaling pathway is critically involved in the function of CD4+CD25+ regulatory T cells in chronic colitis

Teruji Totsuka; Takanori Kanai; Yasuhiro Nemoto; Takayuki Tomita; Ryuichi Okamoto; Kiichiro Tsuchiya; Tetsuya Nakamura; Naoya Sakamoto; Hisaya Akiba; Ko Okumura; Hideo Yagita; Mamoru Watanabe

It is now clear that functional CD4+CD25+ regulatory T (TR) cells exist as part of the normal immune population and prevent the development of intestinal inflammation. We have recently shown that CD4+CD25+ TR cells reside in the intestine and control intestinal homeostasis in humans and mice. In this study, we demonstrate that the TNF family molecule RANKL and its receptor RANK are critically involved in controlling the function of CD4+CD25+ TR cells in the intestine. We first found that RANKL was preferentially expressed on both CD4+CD25+ TR cells and colitogenic CD4+ T cells, whereas RANK was expressed on dendritic cells. Although neutralizing anti-RANKL mAb did not affect TR activity of CD4+CD25+ TR cells to suppress the proliferation of CD4+ responder cells in vitro, in vivo administration of anti-RANKL mAb abrogated CD4+CD25+ TR cell-mediated suppression of colitis induced by adoptive transfer of CD4+CD45RBhigh T cells into SCID mice. Interestingly, an adoptive transfer experiment using Ly5.1+CD4+CD45RBhigh cells and Ly5.2+CD4+CD25+ TR cells revealed that the ratio of CD4+CD25+ TR cells in total CD4+ T cells in inflamed mucosa was significantly decreased by anti-RANKL mAb treatment. Consistent with this, the expression of RANK on lamina propria CD11c+ cells from colitic mice was significantly increased as compared with that from normal mice, and in vitro treatment with anti-RANKL mAb suppressed the expansion of CD4+Foxp3+ TR cells in culture with colitic lamina propria CD11c+ cells. Together, these results suggest that the RANK-RANKL signaling pathway is critically involved in regulating the function of CD4+CD25+ TR cells in colitis.


Gut | 2013

Bone marrow-mesenchymal stem cells are a major source of interleukin-7 and sustain colitis by forming the niche for colitogenic CD4 memory T cells

Yasuhiro Nemoto; Takanori Kanai; Masahiro Takahara; Shigeru Oshima; Tetsuya Nakamura; Ryuichi Okamoto; Kiichiro Tsuchiya; Mamoru Watanabe

Objective Interleukin (IL)-7 is mainly produced in bone marrow (BM) that forms the niche for B cells. We previously demonstrated that BM also retains pathogenic memory CD4 T cells in murine models of inflammatory bowel disease (IBD). However, it remains unknown whether BM-derived IL-7 is sufficient for the development of IBD and which cells form the niche for colitogenic memory CD4 T cells in BM. Design To address these questions, we developed mice in which IL-7 expression was specific for BM, and identified colitis-associated IL-7-expressing mesenchymal stem cells (MSC) in the BM. Results IL-7–/–×RAG-1–/– mice injected with BM cells from IL-7+/+×RAG-1–/– mice, but not from IL-7–/–×RAG-1–/– mice, expressed IL-7 in BM, but not in their colon, and developed colitis when injected with CD4+CD45RBhigh T cells. Cultured BM MSC stably expressed a higher level of IL-7 than that of primary BM cells. IL-7-sufficient, but not IL-7-deficient, BM MSC supported upregulation of Bcl-2 in, and homeostatic proliferation of, colitogenic memory CD4 T cells in vitro. Notably, IL-7–/–×RAG-1–/– mice transplanted with IL-7-sufficient, but not IL-7-deficient, BM MSC expressed IL-7 in BM, but not in their colon, and developed colitis when transplanted with CD4+CD45RBhigh T cells. Conclusions We demonstrate for the first time that BM MSC are a major source of IL-7 and play a pathological role in IBD by forming the niche for colitogenic CD4 memory T cells in BM.


European Journal of Immunology | 2005

Regulation of murine chronic colitis by CD4+CD25– programmed death‐1+ T cells

Teruji Totsuka; Takanori Kanai; Shin Makita; Rei Fujii; Yasuhiro Nemoto; Shigeru Oshima; Ryuichi Okamoto; Akemi Koyanagi; Hisaya Akiba; Ko Okumura; Hideo Yagita; Mamoru Watanabe

Naturally arising CD4+CD25+ regulatory T (TR) cells are engaged in the maintenance of self tolerance and prevention of autoimmune diseases. However, accumulating evidence suggests that a fraction of peripheral CD4+CD25– T cells also possesses regulatory activity. Programmed death‐1 (PD‐1) is a new member of the CD28/CTLA‐4 family, which has been implicated in the maintenance of peripheral self tolerance. Here, we identified a subpopulation of CD4+CD25–PD‐1+ T cells in the spleen of naive mice that constitutively expressed CTLA‐4 and FoxP3 and was hypoproliferative in response to anti‐CD3 antibody stimulation in vitro. However, the CD4+CD25–PD‐1+ T cells uniquely produced large amounts of IL‐4 and IL‐10 in response to anti‐CD3 and anti‐CD28 mAb stimulation, unlike the CD4+CD25+ TR cells. The CD4+CD25–PD‐1+ T cells exhibited a suppressor activity against the proliferation of anti‐CD3 antibody‐stimulated CD4+CD25–PD‐1– T cells in vitro, which was partially abrogated by anti‐CTLA‐4 mAb, but not by anti‐IL‐10 or anti‐PD‐1 mAb. Remarkably, the CD4+CD25–PD‐1+ T cells inhibited the development of colitis induced by adoptive transfer of CD4+CD45RBhigh T cells into C.B17‐scid/scid mice, albeit to a lesser extent than CD4+CD25+ TR cells, in a CTLA‐4‐dependent manner. These results indicate that the CD4+CD25–PD‐1+ T cells contain substantial amounts of TR cells that are involved in the maintenance of peripheral tolerance.


Autophagy | 2015

TNFAIP3 promotes survival of CD4 T cells by restricting MTOR and promoting autophagy

Yu Matsuzawa; Shigeru Oshima; Masahiro Takahara; Chiaki Maeyashiki; Yasuhiro Nemoto; Masanori Kobayashi; Yoichi Nibe; Kengo Nozaki; Takashi Nagaishi; Ryuichi Okamoto; Kiichiro Tsuchiya; Tetsuya Nakamura; Averil Ma; Mamoru Watanabe

Autophagy plays important roles in metabolism, differentiation, and survival in T cells. TNFAIP3/A20 is a ubiquitin-editing enzyme that is thought to be a negative regulator of autophagy in cell lines. However, the role of TNFAIP3 in autophagy remains unclear. To determine whether TNFAIP3 regulates autophagy in CD4 T cells, we first analyzed Tnfaip3-deficient naïve CD4 T cells in vitro. We demonstrated that Tnfaip3-deficient CD4 T cells exhibited reduced MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3) puncta formation, increased mitochondrial content, and exaggerated reactive oxygen species (ROS) production. These results indicate that TNFAIP3 promotes autophagy after T cell receptor (TCR) stimulation in CD4 T cells. We then investigated the mechanism by which TNFAIP3 promotes autophagy signaling. We found that TNFAIP3 bound to the MTOR (mechanistic target of rapamycin) complex and that Tnfaip3-deficient cells displayed enhanced ubiquitination of the MTOR complex and MTOR activity. To confirm the effects of enhanced MTOR activity in Tnfaip3-deficient cells, we analyzed cell survival following treatment with Torin1, an MTOR inhibitor. Tnfaip3-deficient CD4 T cells exhibited fewer cell numbers than the control cells in vitro and in vivo. In addition, the impaired survival of Tnfaip3-deficient cells was ameliorated with Torin1 treatment in vitro and in vivo. The effect of Torin1 was abolished by Atg5 deficiency. Thus, enhanced MTOR activity regulates the survival of Tnfaip3-deficient CD4 T cells. Taken together, our findings illustrate that TNFAIP3 restricts MTOR signaling and promotes autophagy, providing new insight into the manner in which MTOR and autophagy regulate survival in CD4 T cells.

Collaboration


Dive into the Yasuhiro Nemoto's collaboration.

Top Co-Authors

Avatar

Mamoru Watanabe

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ryuichi Okamoto

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kiichiro Tsuchiya

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Teruji Totsuka

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Tetsuya Nakamura

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Takayuki Tomita

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Shigeru Oshima

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Takashi Nagaishi

Tokyo Medical and Dental University

View shared research outputs
Researchain Logo
Decentralizing Knowledge