Network


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

Hotspot


Dive into the research topics where Yoshikiyo Okada is active.

Publication


Featured researches published by Yoshikiyo Okada.


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.


Gut | 2006

Propionibacterium freudenreichii component 1.4‐dihydroxy‐2‐naphthoic acid (DHNA) attenuates dextran sodium sulphate induced colitis by modulation of bacterial flora and lymphocyte homing

Yoshikiyo Okada; Yoshikazu Tsuzuki; Jun-ichi Miyazaki; Koji Matsuzaki; Ryota Hokari; Shunsuke Komoto; Shingo Kato; Atsushi Kawaguchi; Shigeaki Nagao; Kazuro Itoh; Tadao Watanabe; Soichiro Miura

Background and aim: 1.4-Dihydroxy-2-naphthoic acid (DHNA), a bifidogenic growth stimulator from Propionibacterium freudenreichii, is thought to have a beneficial effect as a prebiotic; however, its in vivo effect on intestinal inflammation remains unknown. The aim of this study was to determine whether oral administration of DHNA can ameliorate dextran sodium sulphate (DSS) induced colitis and to determine the possible underlying mechanisms. Method: Colitis was induced in mice by treatment with 2.0% DSS for seven days. DHNA (0.6 or 2.0 mg/kg) was given in drinking water prior to (preventive study) or after (therapeutic study) DSS administration. Colonic damage was histologically scored, and mucosal addressin cell adhesion molecule 1 (MAdCAM-1) expression and β7 positive cell infiltration were determined by immunohistochemistry. mRNA levels of proinflammatory cytokines (interleukin (IL)-1β, IL-6 and tumour necrosis factor α (TNF-α)) were determined by quantitative real time polymerase chain reaction. In addition, bacterial flora in the caecum, concentrations of short chain acids, and luminal pH were examined. Results: DHNA improved survival rate and histological damage score in mice administered DSS in both the preventive and therapeutic studies. DHNA significantly attenuated the enhanced expression of MAdCAM-1, the increased β7 positive cell number, and the increased mRNA levels of IL-1β, IL-6, and TNF-α in DSS treated colon. In addition, the decreased number of Lactobacillus and Enterobacteriaceae induced by DSS was recovered by DHNA. Preventive effects on decrease in butyrate concentration and decrease in pH level in mice administered DSS were also observed in the DHNA preventive study. Conclusion: DHNA, a novel type of prebiotic, attenuates colonic inflammation not only by balancing intestinal bacterial flora but also by suppressing lymphocyte infiltration through reduction of MAdCAM-1.


Journal of Leukocyte Biology | 2005

Blockade of PSGL-1 attenuates CD14+ monocytic cell recruitment in intestinal mucosa and ameliorates ileitis in SAMP1/Yit mice.

Takuya Inoue; Yoshikazu Tsuzuki; Koji Matsuzaki; Hisayuki Matsunaga; Junichi Miyazaki; Ryota Hokari; Yoshikiyo Okada; Atsushi Kawaguchi; Shigeaki Nagao; Kazuro Itoh; Satoshi Matsumoto; Soichiro Miura

The pathogenesis of Crohn’s disease (CD) is not known. However, monocytes and macrophages are thought to play important roles in the development of mucosal inflammation. Therefore, in this study, we examined the role of monocyte‐endothelial cell interactions in senescence‐accelerated mouse P1 (SAMP1)/Yit mice, a murine model of spontaneous ileitis. Fluorescence‐labeled CD14+ monocytic cells isolated from the spleen and mesenteric lymph nodes of AKR/J (control) mice were injected into the tail veins of recipient (AKR/J and SAMP1/Yit) mice, and migration in the postcapillary venules (PCV) of Peyer’s patches, submucosal venules, and villus microvessels of the terminal ileum was monitored by using an intravital microscope. Rolling and adhesion of CD14+ monocytic cells in the PCV of Peyer’s patches and microvessels of the terminal ileum were increased in SAMP1/Yit mice. An imunohistochemical study showed increased expression of P‐selectin glycoprotein‐1 (PSGL‐1), P‐selectin, and vascular cell adhesion molecule‐1 in the terminal ileum of SAMP1/Yit mice. Antibodies against these three adhesion molecules significantly inhibited adhesion of CD14+ monocytic cells to the PCV of Peyer’s patches and microvessels of the terminal ileum, treatment with an anti‐PSGL‐1 monoclonal antibody (mAb) showing the strongest suppressive effect. Anti‐PSGL‐1 mAb also attenuated T cell adhesion in microvessels of intestinal mucosa. In addition, periodical administration of an anti‐PSGL‐1 mAb for 7 weeks significantly ameliorated ileitis of SAMP1/Yit mice. The results suggest that PSGL‐1‐P‐selectin interaction plays an important role in monocyte‐endothelial cell interactions and the development of ileitis in a murine model of CD and that the blockade of this adhesion molecule may be a novel strategy for treating CD.


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.


Clinical and Experimental Immunology | 2005

In vivo demonstration of T lymphocyte migration and amelioration of ileitis in intestinal mucosa of SAMP1/Yit mice by the inhibition of MAdCAM-1

Kouji Matsuzaki; Yoshikazu Tsuzuki; Hisayuki Matsunaga; Takuya Inoue; Jyunichi Miyazaki; Ryota Hokari; Yoshikiyo Okada; A. Kawaguchi; Shigeaki Nagao; Kazurou Itoh; S. Matsumoto; Souichirou Miura

The aetiology of Crohns disease (CD) remains unknown. Since SAMP1/Yit mice have been reported to develop CD‐like spontaneous enteric inflammation, such mice have been studied as an animal model of CD. In this study, using this model we examined T lymphocyte migration in microvessels of intestinal mucosa in vivo and the expression of adhesion molecules by immunohistochemistry. Fluorescence‐labelled T lymphocytes isolated from AKR/J (control) mice were injected into the tail veins of recipient mice, and T lymphocyte migration in the postcapillary venules of Peyers patches, submucosal microvessels, and villus capillaries of the terminal ileum was monitored using an intravital microscope. Adhesion of T lymphocytes was significantly increased in 35 week old SAMP1/Yit mice compared with that in AKR/J or 15 week old SAMP1/Yit mice. Immunohistochemical study showed increased infiltration of CD4, CD8 and β7‐integrin‐positive cells and increased expression of MAdCAM‐1 and VCAM‐1 in the terminal ileum of SAMP1/Yit mice. Antibodies against MAdCAM‐1 and VCAM‐1 significantly inhibited adhesion of T lymphocytes to microvessels of the terminal ileum, and anti‐MAdCAM‐1 antibody showed stronger suppressive effect than the anti‐VCAM‐1 antibody. Periodical administration of anti‐MAdCAM‐1 antibody twice a week for 7 weeks significantly ameliorated ileitis of SAMP1/Yit mice, but submucosal hypertrophy was not significantly suppressed. Anti‐VCAM‐1 antibody treatment failed to show significant resolution of ileitis. In addition, anti‐MAdCAM‐1 antibody treatment also attenuated established ileitis. The results demonstrate that, although MAdCAM‐1 and VCAM‐1 play an important role in T lymphocyte–endothelial cell interactions in SAMP1/Yit mice, MAdCAM‐1 may be a more appropriate target for therapeutic modulation of chronic ileitis.


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.


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.

Collaboration


Dive into the Yoshikiyo Okada's collaboration.

Top Co-Authors

Avatar

Ryota Hokari

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shigeaki Nagao

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Chie Kurihara

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
Top Co-Authors

Avatar

Masaaki Higashiyama

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Yoshikazu Tsuzuki

National Defense Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge