Network


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

Hotspot


Dive into the research topics where Chie Kurihara is active.

Publication


Featured researches published by Chie Kurihara.


Hepatology | 2014

Free cholesterol accumulation in hepatic stellate cells: Mechanism of liver fibrosis aggravation in nonalcoholic steatohepatitis in mice

Kengo Tomita; Toshiaki Teratani; Takahiro Suzuki; Motonori Shimizu; Hirokazu Sato; Kazuyuki Narimatsu; Yoshikiyo Okada; Chie Kurihara; Rie Irie; Hirokazu Yokoyama; Katsuyoshi Shimamura; Shingo Usui; Hirotoshi Ebinuma; Hidetsugu Saito; Chikako Watanabe; Shunsuke Komoto; Atsushi Kawaguchi; Shigeaki Nagao; Kazuo Sugiyama; Ryota Hokari; Takanori Kanai; Soichiro Miura; Toshifumi Hibi

Although nonalcoholic steatohepatitis (NASH) is associated with hypercholesterolemia, the underlying mechanisms of this association have not been clarified. We aimed to elucidate the precise role of cholesterol in the pathophysiology of NASH. C57BL/6 mice were fed a control, high‐cholesterol (HC), methionine‐choline‐deficient (MCD), or MCD+HC diet for 12 weeks or a control, HC, high‐fat (HF), or HF+HC diet for 24 weeks. Increased cholesterol intake accelerated liver fibrosis in both the mouse models without affecting the degree of hepatocellular injury or Kupffer cell activation. The major causes of the accelerated liver fibrosis involved free cholesterol (FC) accumulation in hepatic stellate cells (HSCs), which increased Toll‐like receptor 4 protein (TLR4) levels through suppression of the endosomal‐lysosomal degradation pathway of TLR4, and thereby sensitized the cells to transforming growth factor (TGF)β‐induced activation by down‐regulating the expression of bone morphogenetic protein and activin membrane‐bound inhibitor. Mammalian‐cell cholesterol levels are regulated by way of a feedback mechanism mediated by sterol regulatory element‐binding protein 2 (SREBP2), maintaining cellular cholesterol homeostasis. Nevertheless, HSCs were sensitive to FC accumulation because the high intracellular expression ratio of SREBP cleavage‐activating protein (Scap) to insulin‐induced gene (Insig) disrupted the SREBP2‐mediated feedback regulation of cholesterol homeostasis in these cells. HSC activation subsequently enhanced the disruption of the feedback system by Insig‐1 down‐regulation. In addition, the suppression of peroxisome proliferator‐activated receptor γ signaling accompanying HSC activation enhanced both SREBP2 and microRNA‐33a signaling. Consequently, FC accumulation in HSCs increased and further sensitized these cells to TGFβ‐induced activation in a vicious cycle, leading to exaggerated liver fibrosis in NASH. Conclusion: These characteristic mechanisms of FC accumulation in HSCs are potential targets to treat liver fibrosis in liver diseases including NASH. (Hepatology 2014;58:154–169)


Gastroenterology | 2012

A High-Cholesterol Diet Exacerbates Liver Fibrosis in Mice via Accumulation of Free Cholesterol in Hepatic Stellate Cells

Toshiaki Teratani; Kengo Tomita; Takahiro Suzuki; Tetsuya Oshikawa; Hirokazu Yokoyama; Katsuyoshi Shimamura; Susumu Tominaga; Sadayuki Hiroi; Rie Irie; Yoshikiyo Okada; Chie Kurihara; Hirotoshi Ebinuma; Hidetsugu Saito; Ryota Hokari; Kazuo Sugiyama; Takanori Kanai; Soichiro Miura; Toshifumi Hibi

BACKGROUND & AIMS Some studies have indicated that dietary cholesterol has a role in the progression of liver fibrosis. We investigated the mechanisms by which dietary cholesterol might contribute to hepatic fibrogenesis. METHODS C57BL/6 mice were fed a high-cholesterol diet or a control diet for 4 weeks; liver fibrosis then was induced by bile-duct ligation or carbon tetrachloride administration. Hepatic stellate cells (HSCs) were isolated from mice fed high-cholesterol diets or from Niemann-Pick type C1-deficient mice, which accumulate intracellular free cholesterol. RESULTS After bile-duct ligation or carbon tetrachloride administration, mice fed high-cholesterol diets had significant increases in liver fibrosis and activation of HSCs compared with mice fed control diets. There were no significant differences in the degree of hepatocellular injury or liver inflammation, including hepatocyte apoptosis or Kupffer cell activation, between mice fed high-cholesterol or control diets. Levels of free cholesterol were much higher in HSCs from mice fed high-cholesterol diets than those fed control diets. In cultured HSCs, accumulation of free cholesterol in HSCs increased levels of Toll-like receptor 4 (TLR4), leading to down-regulation of bone morphogenetic protein and activin membrane-bound inhibitor (a pseudoreceptor for transforming growth factor [TGF]β); the HSCs became sensitized to TGFβ-induced activation. Liver fibrosis was not aggravated by the high-cholesterol diet in C3H/HeJ mice, which express a mutant form of TLR4; HSCs that express mutant TLR4 were not activated by accumulation of free cholesterol. CONCLUSIONS Dietary cholesterol aggravates liver fibrosis because free cholesterol accumulates in HSCs, leading to increased TLR4 signaling, down-regulation of bone morphogenetic protein and activin membrane-bound inhibitor, and sensitization of HSC to TGFβ. This pathway might be targeted by antifibrotic therapies.


Inflammatory Bowel Diseases | 2008

Omega‐3 fatty acids exacerbate DSS‐induced colitis through decreased adiponectin in colonic subepithelial myofibroblasts

Hisayuki Matsunaga; Ryota Hokari; Chie Kurihara; Yoshikiyo Okada; Koichi Takebayashi; Keisuke Okudaira; Chikako Watanabe; S. Komoto; Mitsuyasu Nakamura; Yoshikazu Tsuzuki; A. Kawaguchi; Shigeaki Nagao; Kazuro Itoh; Soichiro Miura

Background: Although the immunoregulatory effects of &ohgr;‐3 fatty acid and adiponectin have been postulated, their role in intestinal inflammation is controversial. The aim of this study was to determine whether dietary fat intake influences activity of colonic inflammation through modulating this system. Methods: C57BL/6 mice received dextran sulfate sodium for induction of colitis. Mice were fed a control diet, &ohgr;‐3 fat‐rich diet, &ohgr;‐6 fat‐rich diet, or saturated fat‐rich diet. Some mice were administered a peroxisome proliferator activated receptor‐gamma; agonist, pioglitazone. Messenger RNA expression of adiponectin and its receptors were analyzed. Adiponectin expression in colonic mucosa of ulcerative colitis patients was also analyzed. Results: The receptors for adiponectin were found to be ubiquitously expressed in epithelial cells, intraepithelial lymphocytes, lamina proprial mononuclear cells, and subepithelial myofibroblasts from colonic tissue, but adiponectin was only expressed in myofibroblasts. Induction of colitis significantly decreased the expression of adiponectin in colonic mucosa. The &ohgr;‐3 fat diet group, but not the other fat diet groups, showed exacerbated colitis with a further decrease of adiponectin expression. Pioglitazone treatment ameliorated the level of decrease in adiponectin expression and improved colonic inflammation induced by the &ohgr;‐3 fat‐rich diet. In patients with ulcerative colitis, the expression level of adiponectin in colonic mucosa was also decreased compared with that in control mucosa. Conclusions: Adiponectin was found to be expressed in myofibroblasts. Adiponectin expression was significantly suppressed by induction of colitis, and aggravation of colitis after exposure to &ohgr;‐3 fat may be due to a further decrease in the expression level of adiponectin.


The American Journal of Gastroenterology | 2007

Expression of PD-1, PD-L1, and PD-L2 in the liver in autoimmune liver diseases

Kentaro Kikuchi; Toshihiro Kawai; Masaaki Higashiyama; Yoshikiyo Okada; Chie Kurihara; Ryota Hokari; Atsushi Kawaguchi; Shigeaki Nagao; Toshiro Kondo; Kazuro Itoh; Hiroshi Miyakawa; Soichiro Miura

OBJECTIVES:PD-L1 (also B7-H1) and PD-L2 (also B7-DC) are ligands for programmed death-1 (PD-1), which is a member of the CD28/B7 superfamily of costimulatory molecules and plays an inhibitory role on the periphery. Impaired regulation of this system may cause disruption to self-tolerance leading to autoimmunity; however, the role of these molecules in the liver is unknown. Therefore, we examined the expression of PD-1, PD-L1, and PD-L2 in the liver in autoimmune liver diseases.METHODS:We examined the liver expression of these molecules in autoimmune hepatitis (AIH) and primary biliary cirrhosis (PBC) with no previous medical treatment using immunohistochemical staining and real-time PCR, and compared with chronic hepatitis type C (CHC) as a control.RESULTS:Although PD-1, PD-L1, and PD-L2 were expressed in the liver in AIH, PBC, as well as CHC, the expressions were relatively lower in PBC. In AIH, despite more severe inflammation than in CHC, the expression of these molecules was not greater than in CHC, and when compared with the relative expression of PD-L1, PD-L2 was lower in AIH. PD-L1 and PD-L2 expressions were well correlated with the level of IFN-γ; however, relatively decreased induction for PD-L1 and PD-L2 by IFN-γ was observed in AIH or PBC than in CHC.CONCLUSION:Modulation of PD-1/PD-L1 and PD-L2 systems may play a role in the development of autoimmune liver diseases.


Journal of Hepatology | 2012

p53/p66Shc-mediated signaling contributes to the progression of non-alcoholic steatohepatitis in humans and mice

Kengo Tomita; Toshiaki Teratani; Takahiro Suzuki; Tetsuya Oshikawa; Hirokazu Yokoyama; Katsuyoshi Shimamura; Kiyoshi Nishiyama; Rie Irie; Tohru Minamino; Yoshikiyo Okada; Chie Kurihara; Hirotoshi Ebinuma; Hidetsugu Saito; Ippei Shimizu; Yohko Yoshida; Ryota Hokari; Kazuo Sugiyama; Kazuo Hatsuse; Junji Yamamoto; Takanori Kanai; Soichiro Miura; Toshifumi Hibi

BACKGROUND & AIMS The tumor suppressor p53 is a primary sensor of stressful stimuli, controlling a number of biologic processes. The aim of our study was to examine the roles of p53 in non-alcoholic steatohepatitis (NASH). METHODS Male wild type and p53-deficient mice were fed a methionine- and choline-deficient diet for 8 weeks to induce nutritional steatohepatitis. mRNA expression profiles in normal liver samples and liver samples from patients with non-alcoholic liver disease (NAFLD) were also evaluated. RESULTS Hepatic p53 and p66Shc signaling was enhanced in the mouse NASH model. p53 deficiency suppressed the enhanced p66Shc signaling, decreased hepatic lipid peroxidation and the number of apoptotic hepatocytes, and ameliorated progression of nutritional steatohepatitis. In primary cultured hepatocytes, transforming growth factor (TGF)-β treatment increased p53 and p66Shc signaling, leading to exaggerated reactive oxygen species (ROS) accumulation and apoptosis. Deficient p53 signaling inhibited TGF-β-induced p66Shc signaling, ROS accumulation, and hepatocyte apoptosis. Furthermore, expression levels of p53, p21, and p66Shc were significantly elevated in human NAFLD liver samples, compared with results obtained with normal liver samples. Among NAFLD patients, those with NASH had significantly higher hepatic expression levels of p53, p21, and p66Shc compared with the group with simple steatosis. A significant correlation between expression levels of p53 and p66Shc was observed. CONCLUSIONS p53 in hepatocytes regulates steatohepatitis progression by controlling p66Shc signaling, ROS levels, and apoptosis, all of which may be regulated by TGF-β. Moreover, p53/p66Shc signaling in the liver appears to be a promising target for the treatment of NASH.


Archives of Virology | 1998

Full-length GBV-C/HGV genomes from nine Japanese isolates: characterization by comparative analyses

Kazuhiko Katayama; Tsutomu Kageyama; Shuetsu Fukushi; F. B. Hoshino; Chie Kurihara; Narihiro Ishiyama; Hirofumi Okamura; A. Oya

SummaryThe genomes of nine GBV-C/HGV isolates from Japanese chronic hepatitis patients were fully sequenced and characterized. They shared 85% nucleotide sequence homology with previously characterized isolates from the US and West Africa. Homology studies and phylogenetic analyses showed that the Japanese isolates formed a third group distinct from the established groups 1 and 2. The genetic distances between the three groups of GBV-C/HGV were very similar to the distances between the two classical swine fever virus (CSFV) serotypes, which suggested that they might belong to a separate GBV-C/HGV serotype. Plot similarity analysis comparing the three groups exposed relatively conserved terminal non-coding regions. Hairpin structures predicted in the Japanese isolates are probably involved in viral replication. The region coding E1-E2-NS-2 showed the least similarity (80%); in HCV the similarity here is only 50% due to its hypervariablity. NS-3 and NS-5b that respectivity encode the helicase/protease and RNA-dependent RNA polymerase, had a high degree of amino acid homo- logy, suggesting a high degree of functional constraint in this region. The NS-5b nucleotide sequence was highly conserved perhaps because of constraints from RNA secondary structure and/or an open reading frame in the negative strand.


Journal of Gastroenterology and Hepatology | 2008

Changes in regulatory molecules for lymphangiogenesis in intestinal lymphangiectasia with enteric protein loss

Ryota Hokari; Noritake Kitagawa; Chikako Watanabe; Shunsuke Komoto; Chie Kurihara; Yoshikiyo Okada; Atsushi Kawaguchi; Shigeaki Nagao; Toshifumi Hibi; Soichiro Miura

Background and Aim:  Vascular endothelial growth factor receptor 3 (VEGFR3) and LYVE‐1 are specifically expressed in the endothelium of the lymphatic systems. VEGF‐C, D, FOXC2, Prox 1, and SOX18 are known to play central roles in lymphatic development. We investigated the expression of regulatory molecules for lymphangiogenesis in the duodenal mucosa of idiopathic intestinal lymphangiectasia.


Journal of Hepatology | 2014

Acyl-CoA:cholesterol acyltransferase 1 mediates liver fibrosis by regulating free cholesterol accumulation in hepatic stellate cells

Kengo Tomita; Toshiaki Teratani; Takahiro Suzuki; Motonori Shimizu; Hirokazu Sato; Kazuyuki Narimatsu; Shingo Usui; Hirotaka Furuhashi; Akifumi Kimura; Kiyoshi Nishiyama; Tadashi Maejima; Yoshikiyo Okada; Chie Kurihara; Katsuyoshi Shimamura; Hirotoshi Ebinuma; Hidetsugu Saito; Hirokazu Yokoyama; Chikako Watanabe; Shunsuke Komoto; Shigeaki Nagao; Kazuo Sugiyama; Suefumi Aosasa; Kazuo Hatsuse; Junji Yamamoto; Toshifumi Hibi; Soichiro Miura; Ryota Hokari; Takanori Kanai

BACKGROUND & AIMS Acyl-coenzyme A: cholesterol acyltransferase (ACAT) catalyzes the conversion of free cholesterol (FC) to cholesterol ester, which prevents excess accumulation of FC. We recently found that FC accumulation in hepatic stellate cells (HSCs) plays a role in progression of liver fibrosis, but the effect of ACAT1 on liver fibrosis has not been clarified. In this study, we aimed to define the role of ACAT1 in the pathogenesis of liver fibrosis. METHODS ACAT1-deficient and wild-type mice, or Toll-like receptor 4 (TLR4)(-/-)ACAT1(+/+) and TLR4(-/-)ACAT1(-/-) mice were subjected to bile duct ligation (BDL) for 3 weeks or were given carbon tetrachloride (CCl4) for 4 weeks to induce liver fibrosis. RESULTS ACAT1 was the major isozyme in mice and human primary HSCs, and ACAT2 was the major isozyme in mouse primary hepatocytes and Kupffer cells. ACAT1 deficiency significantly exaggerated liver fibrosis in the mouse models of liver fibrosis, without affecting the degree of hepatocellular injury or liver inflammation, including hepatocyte apoptosis or Kupffer cell activation. ACAT1 deficiency significantly increased FC levels in HSCs, augmenting TLR4 protein and downregulating expression of transforming growth factor-β (TGFβ) pseudoreceptor Bambi (bone morphogenetic protein and activin membrane-bound inhibitor), leading to sensitization of HSCs to TGFβ activation. Exacerbation of liver fibrosis by ACAT1 deficiency was dependent on FC accumulation-induced enhancement of TLR4 signaling. CONCLUSIONS ACAT1 deficiency exaggerates liver fibrosis mainly through enhanced FC accumulation in HSCs. Regulation of ACAT1 activities in HSCs could be a target for treatment of liver fibrosis.


Journal of Leukocyte Biology | 2012

HIF-1 in T cells ameliorated dextran sodium sulfate-induced murine colitis

Masaaki Higashiyama; Ryota Hokari; Hideaki Hozumi; Chie Kurihara; Toshihide Ueda; Chikako Watanabe; Kengo Tomita; Mitsuyasu Nakamura; Shunsuke Komoto; Yoshikiyo Okada; Atsushi Kawaguchi; Shigeaki Nagao; Makoto Suematsu; Nobuhito Goda; Soichiro Miura

HIF‐1 is active in hypoxia, such as inflamed mucosa, and HIF‐1 in epithelium has been reported to control inflamed mucosa in IBD models. Although T cells play an important role for pathogenesis of IBD, the function of HIF‐1 in T cells remains to be elucidated. We aimed to clarify the function of HIF‐1 in T cells in IBD with focus on the balance between Treg and Teff. Double immunohistochemistry of colonic mucosa in IBD patients showed that HIF‐1 was expressed in T cells infiltrating the inflamed mucosa, suggesting that HIF‐1 in T cells is involved in the pathogenesis. DSS administration to T cell‐specific HIF‐1α KO mice showed more severe colonic inflammation than control mice with the up‐regulation of Th1 and Th17. Hypoxic stimulation in vitro increased Treg activation in WT T cells but not in HIF‐1‐deleted T cells. In contrast, hypoxic stimulation increased Th17 activation, and the degree was higher in HIF‐1‐deleted cells than in control cells. These results show that hypoxia controls intestinal inflammation by regulating cytokine balance in a HIF‐1‐dependent manner, suggesting that strengthening HIF‐1 induction in T cells at the sites of inflammation might be a therapeutic strategy for IBD regulation.


PLOS ONE | 2013

Norovirus binding to intestinal epithelial cells is independent of histo-blood group antigens.

Kosuke Murakami; Chie Kurihara; Tomoichiro Oka; Takashi Shimoike; Yoshiki Fujii; Reiko Takai-Todaka; YoungBin Park; Takaji Wakita; Tsukasa Matsuda; Ryota Hokari; Soichiro Miura; Kazuhiko Katayama

Human noroviruses (NoVs) are a major cause of non-bacterial gastroenteritis. Although histo-blood group antigens (HBGAs) have been implicated in the initial binding of NoV, the mechanism of that binding before internalization is not clear. To determine the involvement of NoVs and HBGAs in cell binding, we examined the localization of NoV virus-like particles (VLPs) and HBGAs in a human intestinal cell line and the human ileum biopsy specimens by immunofluorescence microscopy. The localizations of Ueno 7k VLPs (genogroup II.6) and each HBGA (type H1-, H2- and Leb-HBGAs) on the human intestinal cell line, Caco-2, were examined by confocal laser-scanning microscopy. To explore any interactions of NoVs and HBGAs in vivo, fresh biopsy specimens from human ileum were directly incubated with NoV VLPs and examined by immunofluorescence microscopy. We found that VLP binding depended on the state of cell differentiation, but not on the presence of HBGAs. In differentiated Caco-2 cells, we detected no type H1 HBGAs, but VLPs bound to the cells anyway. We incubated fresh biopsies of human ileum directly with VLPs, a model that better replicates the in vivo environment. VLPs mainly bound epithelial cells and goblet cells. Although the incubations were performed at 4°C to hinder internalization, VLPs were still detected inside cells. Our results suggest that VLPs utilize molecule(s) other than HBGAs during binding and internalization into cells.

Collaboration


Dive into the Chie Kurihara's collaboration.

Top Co-Authors

Avatar

Ryota Hokari

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoshikiyo Okada

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Shigeaki Nagao

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Chikako Watanabe

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Shunsuke Komoto

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Kengo Tomita

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Masaaki Higashiyama

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hideaki Hozumi

National Defense Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge