研究者業績

矢作 直也

ヤハギ ナオヤ  (Yahagi Naoya)

基本情報

所属
自治医科大学 内科学講座 内分泌代謝学部門 教授
学位
博士(医学)(東京大学)

連絡先
nyahagi-tkyumin.ac.jp
研究者番号
60420246
J-GLOBAL ID
200901002727554646
researchmap会員ID
6000006682

外部リンク

論文

 150
  • Yoshinori Takeuchi, Naoya Yahagi, Yuichi Aita, Yuki Murayama, Yoshikazu Sawada, Xiaoying Piao, Naoki Toya, Yukari Oya, Akito Shikama, Ayako Takarada, Yukari Masuda, Makiko Nishi, Midori Kubota, Yoshihiko Izumida, Takashi Yamamoto, Motohiro Sekiya, Takashi Matsuzaka, Yoshimi Nakagawa, Osamu Urayama, Yasushi Kawakami, Yoko Iizuka, Takanari Gotoda, Keiji Itaka, Kazunori Kataoka, Ryozo Nagai, Takashi Kadowaki, Nobuhiro Yamada, Yuan Lu, Mukesh K. Jain, Hitoshi Shimano
    CELL REPORTS 16(9) 2373-2386 2016年8月  査読有り責任著者
    Hepatic lipogenesis is nutritionally regulated (i.e., downregulated during fasting and upregulated during the postprandial state) as an adaptation to the nutritional environment. While alterations in the expression level of the transcription factor SREBP1c are known to be critical for nutritionally regulated lipogenesis, upstream mechanisms governing Srebf1 expression remain unclear. Here, we show that the fasting-induced transcription factor KLF15, a key regulator of gluconeogenesis, forms a complex with LXR/RXR, specifically on the Srebf1 promoter. This complex recruits the corepressor RIP140 instead of the coactivator SRC1, resulting in reduced Srebf1 and thus downstream lipogenic enzyme expression during the early and euglycemic period of fasting prior to hypoglycemia and PKA activation. Through this mechanism, KLF15 overexpression specifically ameliorates hypertriglyceridemia without affecting LXR-mediated cholesterol metabolism. These findings reveal a key molecular link between glucose and lipid metabolism and have therapeutic implications for the treatment of hyperlipidemia.
  • H. Suzuki, S. Furukawa, K. Fujihara, K. Kobayashi, H. Iwasaki, S. Yatoh, Y. Sugano, M. Sekiya, N. Yahagi, H. Yagyu, H. Shimano
    DIABETOLOGIA 59(1) S538-S538 2016年8月  
  • Yoshimi Nakagawa, Fusaka Oikawa, Seiya Mizuno, Hiroshi Ohno, Yuka Yagishita, Aoi Satoh, Yoshinori Osaki, Kenta Takei, Takuya Kikuchi, Song-iee Han, Takashi Matsuzaka, Hitoshi Iwasaki, Kazuto Kobayashi, Shigeru Yatoh, Naoya Yahagi, Masaaki Isaka, Hiroaki Suzuki, Hirohito Sone, Satoru Takahashi, Nobuhiro Yamada, Hitoshi Shimano
    SCIENTIFIC REPORTS 6 27857 2016年6月  査読有り
    cAMP responsive element binding protein 3-like 3 (CREB3L3), a transcription factor expressed in the liver and small intestine, governs fasting-response energy homeostasis. Tissue-specific CREB3L3 knockout mice have not been generated till date. To our knowledge, this is the first study using the one-step CRISPR/Cas9 system to generate CREB3L3 floxed mice and subsequently obtain liver-and small intestine-specific Creb3l3 knockout (LKO and IKO, respectively) mice. While LKO mice as well as global KO mice developed hypertriglyceridemia, LKO mice exhibited hypercholesterolemia in contrast to hypocholesterolemia in global KO mice. LKO mice demonstrated up-regulation of hepatic Srebf2 and its corresponding target genes. No phenotypic differences were observed between IKO and floxed mice. Severe liver injury was observed in LKO mice fed a methionine-choline deficient diet, a model for non-alcoholic steatohepatitis. These results provide new evidence regarding the hepatic CREB3L3 role in plasma triglyceride metabolism and hepatic and intestinal CREB3L3 contributions to cholesterol metabolism.
  • Noriko Suzuki-Kemuriyama, Takashi Matsuzaka, Motoko Kuba, Hiroshi Ohno, Song-iee Han, Yoshinori Takeuchi, Masaaki Isaka, Kazuto Kobayashi, Hitoshi Iwasaki, Shigeru Yatoh, Hiroaki Suzuki, Katsuhiro Miyajima, Dai Nakae, Naoya Yahagi, Yoshimi Nakagawa, Hirohito Sone, Nobuhiro Yamada, Hitoshi Shimano
    PLOS ONE 11(6) e0157580 2016年6月  査読有り
    Non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of metabolic syndrome, can progress to steatohepatitis (NASH) and advanced liver damage, such as that from liver cirrhosis and cancer. Recent studies have shown the benefits of consuming n-3 polyunsaturated fatty acids (PUFAs) for the treatment of NAFLD. In the present study, we investigated and compared the effects of the major n-3 PUFAs-eicosapentaenoic acid (EPA, C20:5) and docosahexaenoic acid (DHA, C22:6)-in preventing atherogenic high-fat (AHF) diet-induced NAFLD. Mice were fed the AHF diet supplemented with or without EPA or DHA for four weeks. Both EPA and DHA reduced the pathological features of AHF dietinduced NASH pathologies such as hepatic lobular inflammation and elevated serum transaminase activity. Intriguingly, EPA had a greater hepatic triacylglycerol (TG)-reducing effect than DHA. In contrast, DHA had a greater suppressive effect than EPA on AHF diet-induced hepatic inflammation and ROS generation, but no difference in fibrosis. Both EPA and DHA could be effective for treatment of NAFLD and NASH. Meanwhile, the two major n-3 polyunsaturated fatty acids might differ in a relative contribution to pathological intermediate steps towards liver fibrosis.
  • Motoko Kuba, Takashi Matsuzaka, Rie Matsumori, Ryo Saito, Naoko Kaga, Hikari Taka, Kei Ikehata, Naduki Okada, Takuya Kikuchi, Hiroshi Ohno, Song-Iee Han, Yoshinori Takeuchi, Kazuto Kobayashi, Hitoshi Iwasaki, Shigeru Yatoh, Hiroaki Suzuki, Hirohito Sone, Naoya Yahagi, Yoji Arakawa, Tsutomu Fujimura, Yoshimi Nakagawa, Nobuhiro Yamada, Hitoshi Shimano
    SCIENTIFIC REPORTS 5 17604 2015年12月  査読有り
    Nonalcoholic steatohepatitis (NASH) is a progressive form of nonalcoholic fatty liver disease (NAFLD) that can develop into liver cirrhosis and cancer. Elongation of very long chain fatty acids (ELOVL) family member 6 (Elovl6) is a microsomal enzyme that regulates the elongation of C12-16 saturated and monounsaturated fatty acids (FAs). We have previously shown that Elovl6 plays an important role in the development of hepatic insulin resistance and NASH by modifying FA composition. Recent studies have linked altered hepatic cholesterol homeostasis and cholesterol accumulation to the pathogenesis of NASH. In the present study, we further investigated the role of Elovl6 in the progression of lithogenic diet (LD)-induced steatohepatitis. We showed that the absence of Elovl6 suppresses hepatic lipid accumulation, plasma total cholesterol and total bile acid (BA) levels in LDL receptor-deficient (Ldlr(-/-)) mice challenged with a LD. The absence of Elovl6 also decreases hepatic inflammation, oxidative stress and liver injury, but increases the formation of cholesterol crystals in the less dilated gallbladder. These findings suggest that Elovl6-mediated changes in hepatic FA composition, especially oleic acid (C18:1n-9), control handling of hepatic cholesterol and BA, which protects against hepatotoxicity and steatohepatitis, but promotes gallstone formation in LD-fed Ldlr(-/-) mice.
  • Akito Shikama, Haruna Shinozaki, Yoshinori Takeuchi, Takashi Matsuzaka, Yuichi Aita, Tomoki Murayama, Yoshikazu Sawada, Xiaoying Piao, Naoki Toya, Yukari Oya, Ayako Takarada, Yukari Masuda, Makiko Nishi, Midori Kubota, Yoshihiko Izumida, Yoshimi Nakagawa, Hitoshi Iwasaki, Kazuto Kobayashi, Shigeru Yatoh, Hiroaki Suzuki, Hiroaki Yagyu, Yasushi Kawakami, Nobuhiro Yamada, Hitoshi Shimano, Naoya Yahagi
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 465(4) 857-863 2015年10月  査読有り最終著者責任著者
    Fatty acid elongase 5 (ELOVL5) is an enzyme involved in the synthesis of polyunsaturated fatty acids. Sterol Regulatory Element-binding Protein (SREBP)-1 activates ELOVL5 and increases polyunsaturated fatty acid synthesis, which in turn negatively affects SREBP-1 expression. Thus, ELOVL5 has been established as an SREBP-1 target gene and an important component of the negative feedback loop of de novo lipogenesis. However, the human ELOVL5 promoter/enhancer has not been fully analyzed and the location of SREBP biding sites around the ELOVL5 gene has yet to be defined. Here we performed a detailed promoter/enhancer analysis of human ELOVL5 gene, and identified two new SREBP binding sites, one in the 10 kb upstream region and one in the exon 1. These two SRE motifs are conserved among mammals and the mechanism found in the present study by which SREBP activates ELOVL5 is considered to be common in mammals. Through these findings, we clarified the molecular mechanism how SREBP activates ELOVL5, an important regulator of de novo lipogenesis. (C) 2015 Elsevier Inc. All rights reserved.
  • Yoshinori Osaki, Yoshimi Nakagawa, Shoko Miyahara, Hitoshi Iwasaki, Akiko Ishii, Takashi Matsuzaka, Kazuto Kobayashi, Shigeru Yatoh, Akimitsu Takahashi, Naoya Yahagi, Hiroaki Suzuki, Hirohito Sone, Ken Ohashi, Shun Ishibashi, Nobuhiro Yamada, Hitoshi Shimano
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 466(3) 536-540 2015年10月  査読有り
    HMG-CoA reductase (HMGCR) catalyzes the conversion of HMG-CoA to mevalonic acid (MVA); this is the rate-limiting enzyme of the mevalonate pathway that synthesizes cholesterol. Statins, HMGCR inhibitors, are widely used as cholesterol-reducing drugs. However, statin-induced myopathy is the most adverse side effect of statins. To eludicate the mechanisms underlying statin the myotoxicity and HMGCR function in the skeletal muscle, we developed the skeletal muscle-specific HMGCR knockout mice. Knockout mice exhibited postnatal myopathy with elevated serum creatine kinase levels and necrosis. Myopathy in knockout mice was completely rescued by the oral administration of MVA. These results suggest that skeletal muscle toxicity caused by statins is dependent on the deficiencies of HMGCR enzyme activity and downstream metabolites of the mevalonate pathway in skeletal muscles rather than the liver or other organs. (C) 2015 Elsevier Inc. All rights reserved.
  • Kazuya Fujihara, Hiroaki Suzuki, Akira Sato, Satoru Kodama, Yoriko Heianza, Kazumi Saito, Hitoshi Iwasaki, Kazuto Kobayashi, Shigeru Yatoh, Akimitsu Takahashi, Naoya Yahagi, Hiroaki Yagyu, Hirohito Sone, Hitoshi Shimano
    JOURNAL OF DIABETES RESEARCH 2015 507245 2015年  査読有り
    Aims. To elucidate the levels of malondialdehyde-modified LDL (MDA-LDL)-related variables for predicting coronary artery stenosis (CAS) by coronary CT angiography (CCTA) in asymptomatic patients with type 2 diabetes (T2DM). Methods. Enrolled were 36 Japanese patients with T2DM who underwent CCTA and in whom MDA-LDL levels were measured. Definition of CAS was luminal narrowing of >= 50%. Trends through tertiles of each MDA-LDL-related variable were analyzed with a general linear model. The ability of each MDA-LDL-related variable to predict CAS was compared to areas under the curve (AUCs) in receiver operating characteristic curve (ROC) analysis. Results. Seventeen patients had CAS. Each MDA-IDL-related variable was an independent predictor of CAS (P = 0.039 for MDALDL, P = 0.013 for MDA-LDL/LDL-C, P = 0.047 for MDA-LDL/HDL-C, and P = 0.013 for (MDA-LDULDL-C)/HDL-C). AUCs of MDA-LDL, MDA-LDL/LDL-C, MDA-LDL/HDL-C, and (MDA-LDULDLC)/HDL-C were 0.675 (95% CI 0.496-0.854), 0.765 (0.602-0.927), 0.752 (0.592-0.913), and 0.799 (0.643-0.955), respectively, for predicting CAS. Trends throughout the tertiles showed significant associations between N4DA-LDL/LDL-C, N4DA-LDL/41DL-C, or (MDALDL/LDL-C)/41DL-C and CAS (P 0.003 for MDA-LDL/LDL-C, P=0.042 for MDA-LDULIDL-C, and P 0.001 for (MDA-LDULDL-C)/HDL-C). Conclusions. Data suggest that measurements of MDA-LDL/LDL-C, MDA-LDL/HDLC, and (MakLDULDL-C)/HDL-C are useful for predicting CAS.
  • Yoshimi Nakagawa, Aoi Satoh, Sachiko Yabe, Mika Furusawa, Naoko Tokushige, Hitomi Tezuka, Motoki Mikami, Wakiko Iwata, Akiko Shingyouchi, Takashi Matsuzaka, Shiori Kiwata, Yuri Fujimoto, Hidehisa Shimizu, Hirosuke Danno, Takashi Yamamoto, Kiyoaki Ishii, Tadayoshi Karasawa, Yoshinori Takeuchi, Hitoshi Iwasaki, Masako Shimada, Yasushi Kawakami, Osamu Urayama, Hirohito Sone, Kazuhiro Takekoshi, Kazuto Kobayashi, Shigeru Yatoh, Akimitsu Takahashi, Naoya Yahagi, Hiroaki Suzuki, Nobuhiro Yamada, Hitoshi Shimano
    ENDOCRINOLOGY 155(12) 4706-4719 2014年12月  査読有り
    Transcriptional regulation of metabolic genes in the liver is the key to maintaining systemic energy homeostasis during starvation. The membrane-bound transcription factor cAMP-responsive element-binding protein 3-like 3 (CREB3L3) has been reported to be activated during fasting and to regulate triglyceride metabolism. Here, we show that CREB3L3 confers a wide spectrum of metabolic responses to starvation in vivo. Adenoviral and transgenic overexpression of nuclear CREB3L3 induced systemic lipolysis, hepatic ketogenesis, and insulin sensitivity with increased energy expenditure, leading to marked reduction in body weight, plasma lipid levels, and glucose levels. CREB3L3 overexpression activated gene expression levels and plasma levels of antidiabetic hormones, including fibroblast growth factor 21 and IGF-binding protein 2. Amelioration of diabetes by hepatic activation of CREB3L3 was also observed in several types of diabetic obese mice. Nuclear CREB3L3 mutually activates the peroxisome proliferator-activated receptor(PPAR) alpha promoter in an autoloop fashion and is crucial for the ligand transactivation of PPAR alpha by interacting with its transcriptional regulator, peroxisome proliferator-activated receptor gamma coactivator-1 alpha. CREB3L3 directly and indirectly controls fibroblast growth factor 21 expression and its plasma level, which contributes at least partially to the catabolic effects of CREB3L3 on systemic energy homeostasis in the entire body. Therefore, CREB3L3 is a therapeutic target for obesity and diabetes.
  • Nie Tang, Takashi Matsuzaka, Marii Suzuki, Yuta Nakano, Hui Zao, Tomotaka Yokoo, Noriko Suzuki-Kemuriyama, Motoko Kuba, Yuka Okajima, Yoshinori Takeuchi, Kazuto Kobayashi, Hitoshi Iwasaki, Shigeru Yatoh, Akimitsu Takahashi, Hiroaki Suzuki, Hirohito Sone, Masako Shimada, Yoshimi Nakagawa, Naoya Yahagi, Nobuhiro Yamada, Hitoshi Shimano
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 450(1) 318-323 2014年7月  査読有り
    ELOVL family member 6, elongation of very long-chain fatty acids (Elovl6) is a microsomal enzyme that regulates the elongation of C12-16 saturated and monounsaturated fatty acids and is related to the development of obesity-induced insulin resistance via the modification of the fatty acid composition. In this study, we investigated the role of systemic Elovl6 in the pancreatic islet and beta-cell function. Elovl6 is expressed in both islets and beta-cell lines. In mice fed with chow, islets of the Elovl6(-/-) mice displayed normal architecture and beta-cell mass compared with those of the wild-type mice. However, when fed a high-fat, high-sucrose (HFHS) diet, the islet hypertrophy in response to insulin resistance observed in normal mice was attenuated and glucose-stimulated insulin secretion (GSIS) increased in the islets of Elovl6(-/-) mice compared with those of the wild-type mice. Enhanced GSIS in the HFHS Elovl6-/- islets was associated with an increased ATP/ADP ratio and the suppression of ATF-3 expression. Our findings suggest that Elovl6 could be involved in insulin secretory capacity per beta-cell and diabetes. (C) 2014 Elsevier Inc. All rights reserved.
  • Takahiro Horie, Tomohiro Nishino, Osamu Baba, Yasuhide Kuwabara, Tetsushi Nakao, Masataka Nishiga, Shunsuke Usami, Masayasu Izuhara, Fumiko Nakazeki, Yuya Ide, Satoshi Koyama, Naoya Sowa, Naoya Yahagi, Hitoshi Shimano, Tomoyuki Nakamura, Koji Hasegawa, Noriaki Kume, Masayuki Yokode, Toru Kita, Takeshi Kimura, Koh Ono
    SCIENTIFIC REPORTS 4 5312 2014年6月  査読有り
    MicroRNAs (miRs) are small non-protein-coding RNAs that bind to specific mRNAs and inhibit translation or promote mRNA degradation. Recent reports, including ours, indicated that miR-33a located within the intron of sterol regulatory element-binding protein (SREBP) 2 controls cholesterol homeostasis and can be a possible therapeutic target for treating atherosclerosis. Primates, but not rodents, express miR-33b from an intron of SREBF1. Therefore, humanized mice, in which a miR-33b transgene is inserted within a Srebf1 intron, are required to address its function in vivo. We successfully established miR-33b knock-in (KI) mice and found that protein levels of known miR-33a target genes, such as ABCA1, ABCG1, and SREBP-1, were reduced compared with those in wild-type mice. As a consequence, macrophages from the miR-33b KI mice had a reduced cholesterol efflux capacity via apoA-I and HDL-C. Moreover, HDL-C levels were reduced by almost 35% even in miR-33b KI hetero mice compared with the control mice. These results indicate that miR-33b may account for lower HDL-C levels in humans than those in mice and that miR-33b is possibly utilized for a feedback mechanism to regulate its host gene SREBF1. Our mice will also aid in elucidating the roles of miR-33a/b in different genetic disease models.
  • Kazuya Fujihara, Hiroaki Suzuki, Akira Sato, Tomoko Ishizu, Satoru Kodama, Yoriko Heianza, Kazumi Saito, Hitoshi Iwasaki, Kazuto Kobayashi, Shigeru Yatoh, Akimitsu Takahashi, Naoya Yahagi, Hirohito Sone, Hitoshi Shimano
    JOURNAL OF ATHEROSCLEROSIS AND THROMBOSIS 21(8) 799-815 2014年  査読有り
    Aims: To compare the efficacy of Framingham Risk Score (FRS), UK Prospective Diabetes Study (UKPDS) risk engine, a risk score based on the Japanese Atherosclerosis Longitudinal Study-Existing Cohorts Combine (JALS-ECC), the maximum intima-media thickness (max-IMT) determined on coronary computed tomography angiography (CCTA) and their combination in asymptomatic patients with type 2 diabetes. Methods: A total of 116 Japanese patients with type 2 diabetes underwent CCTA. The risk of coronary heart disease was calculated according to the FRS, UKPDS and JALS-ECC. We evaluated the reclassification of coronary artery stenosis (CAS) based on the risk score categories after adding each IMT related variable. Results: Sixty-eight patients had CAS. The areas under the curves (AUCs) in the receiver operating characteristic curve analyses of FRS, UKPDS and JALS-ECC were 0.763 (95% confidence interval [CI]: 0.674-0.853), 0.785 (95% CI: 0.703-0.868) and 0.767 (95% CI: 0.681-0.853), respectively. The AUCs for FRS, UKPDS and JALS-ECC combined with the max-IMT were 0.788 (95% CI: 0.705-0.872), 0.800 (95% CI: 0.720-0.879) and 0.786 (95% CI: 0.703-0.869), respectively. Combining the max-IMT with the risk scores improved the identification of subjects with stenotic lesions, in particular, those in the first, second and third tertiles of the FRS, first and second tertiles of the UKPDS and first and second tertiles of the JALS-ECC (P=0.054, P=0.056, P=0.015, P=0.082, P=0.060, P=0.007, and P=0.080, respectively). The net reclassification improvement increased following the addition of a max-IMT of >= 1.9 mm (32.4% in FRS, 19.9% in UKPDS and 51.7% in JALS-ECC). Conclusions: These data suggest that combining a risk score with the max-IMT improves the prediction of CAS in comparison with the risk score alone.
  • Yoshihiko Izumida, Naoya Yahagi, Yoshinori Takeuchi, Makiko Nishi, Akito Shikama, Ayako Takarada, Yukari Masuda, Midori Kubota, Takashi Matsuzaka, Yoshimi Nakagawa, Yoko Iizuka, Keiji Itaka, Kazunori Kataoka, Seiji Shioda, Akira Niijima, Tetsuya Yamada, Hideki Katagiri, Ryozo Nagai, Nobuhiro Yamada, Takashi Kadowaki, Hitoshi Shimano
    NATURE COMMUNICATIONS 4 2316 2013年12月  査読有り責任著者
  • Takahiro Horie, Tomohiro Nishino, Osamu Baba, Yasuhide Kuwabara, Tetsushi Nakao, Masataka Nishiga, Shunsuke Usami, Masayasu Izuhara, Naoya Sowa, Naoya Yahagi, Hitoshi Shimano, Shigenobu Matsumura, Kazuo Inoue, Hiroyuki Marusawa, Tomoyuki Nakamura, Koji Hasegawa, Noriaki Kume, Masayuki Yokode, Toru Kita, Takeshi Kimura, Koh Ono
    NATURE COMMUNICATIONS 4 2883 2013年12月  査読有り
    MicroRNAs (miRs) are small non-protein-coding RNAs that bind to specific mRNAs and inhibit translation or promote mRNA degradation. Recent reports have indicated that miR-33, which is located within the intron of sterol regulatory element-binding protein (SREBP) 2, controls cholesterol homoeostasis and may be a potential therapeutic target for the treatment of atherosclerosis. Here we show that deletion of miR-33 results in marked worsening of high-fat diet-induced obesity and liver steatosis. Using miR-33(-/-) Srebf1(+/-) mice, we demonstrate that SREBP-1 is a target of miR-33 and that the mechanisms leading to obesity and liver steatosis in miR-33(-/-) mice involve enhanced expression of SREBP-1. These results elucidate a novel interaction between SREBP-1 and SREBP-2 mediated by miR-33 in vivo.
  • Yuri Fujimoto, Yoshimi Nakagawa, Aoi Satoh, Kanako Okuda, Akiko Shingyouchi, Ayano Naka, Takashi Matsuzaka, Hitoshi Iwasaki, Kazuto Kobayashi, Naoya Yahagi, Masako Shimada, Shigeru Yatoh, Hiroaki Suzuki, Satomi Yogosawa, Tetsuro Izumi, Hirohito Sone, Osamu Urayama, Nobuhiro Yamada, Hitoshi Shimano
    ENDOCRINOLOGY 154(10) 3577-3588 2013年10月  査読有り
    Transcription factor E3 (TFE3) is a transcription factor that binds to E-box motifs and promotes energy metabolism-related genes. We previously reported that TFE3 directly binds to the insulin receptor substrate-2 promoter in the liver, resulting in increased insulin response. However, the role of TFE3 in other tissues remains unclear. In this study, we generated adipose-specific TFE3 transgenic (aP2-TFE3 Tg) mice. These mice had a higher weight of white adipose tissue (WAT) and brown adipose tissue than wild-type (WT) mice under fasting conditions. Lipase activity in the WAT in these mice was lower than that in the WT mice. Them RNA level of adipose triglyceride lipase (ATGL), the rate-limiting enzyme for adipocyte lipolysis, was significantly decreased in aP2-TFE3 Tg mice. The expression of Foxo1, which directly activates ATG Lexpression, was also suppressed in transgenic mice. Promoter analysis confirmed that TFE3 suppressed promoter activities of the ATGL gene. In contrast, G0S2 and Perilipin1, which attenuate ATGL activity, were higher in transgenic mice than in WT mice. These results indicated that the decrease in lipase activity in adipose tissues was due to a decrease in ATGL expression and suppression of ATGL activity. We also showed that thermogenesis was suppressed in aP2-TFE3 Tg mice. The decrease in lipolysis in WAT of aP2-TFE3 Tg mice inhibited the supply of fatty acids to brown adipose tissue, resulting in the inhibition of the expression of thermogenesis-related genes such as UCP1. Our data provide new evidence that TFE3 regulates lipid metabolism by controlling the gene expression related to lipolysis and thermogenesis in adipose tissue.
  • 矢藤 繁, 志鎌 明人, 高木 聡, 岩崎 仁, 小林 和人, 矢作 直也, 鈴木 浩明, 須藤 玲子, 高木 彩, 木村 かよ, 杉本 佳奈美, 伊藤 由実子, 高橋 弘文, 岩部 博子, 鈴木 康裕, 横井 麻加, 大畑 瞳, 及川 仁, 島野 仁
    糖尿病 56(Suppl.1) S-204 2013年4月  
  • Ayano Naka, Kaoruko Tada Iida, Yoshimi Nakagawa, Hitoshi Iwasaki, Yoshinori Takeuchi, Aoi Satoh, Takashi Matsuzaka, Kiyo-aki Ishii, Kazuto Kobayashi, Shigeru Yatoh, Masako Shimada, Naoya Yahagi, Hiroaki Suzuki, Hirohito Sone, Nobuhiro Yamada, Hitoshi Shimano
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 430(2) 664-669 2013年1月  査読有り
    Transcription factor E3 (TFE3) belongs to a basic-helix-loop-helix family, and is involved in the biology of osteoclasts, melanocytes and their malignancies. We previously reported the metabolic effects of TFE3 on insulin in the liver and skeletal muscles in animal models. In the present study, we explored a novel role for TFE3 in a skeletal muscle cell line. When TFE3 was overexpressed in C2C12 myoblasts by adenovirus before induction of differentiation, myogenic differentiation of C2C12 cells was significantly inhibited. Adenovirus-mediated TFE3 overexpression also suppressed the gene expression of muscle regulatory factors (MRFs), such as MyoD and myogenin, during C2C12 differentiation. In contrast, knockdown of TFE3 using adenovirus encoding short-hairpin RNAi specific for TFE3 dramatically promoted myoblast differentiation associated with significantly increased expression of MRFs. Consistent with these findings, promoter analyses via luciferase reporter assay and electrophoretic mobility shift assay suggestid that TFE3 negatively regulated myogenin promoter activity by direct binding to an E-box, E2, in the myogenin promoter. These findings indicated that TFE3 has a regulatory role in myoblast differentiation, and that transcriptional suppression of myogenin expression may be part of the mechanism of action. (C) 2012 Elsevier Inc. All rights reserved.
  • Satoru Takase, Jun-Ichi Osuga, Hayato Fujita, Kazuo Hara, Motohiro Sekiya, Masaki Igarashi, Mikio Takanashi, Yoshinori Takeuchi, Yoshihiko Izumida, Keisuke Ohta, Masayoshi Kumagai, Makiko Nishi, Midori Kubota, Yukari Masuda, Yoshino Taira, Sachiko Okazaki, Yoko Iizuka, Naoya Yahagi, Ken Ohashi, Hiroshi Yoshida, Hidekatsu Yanai, Norio Tada, Takanari Gotoda, Shun Ishibashi, Takashi Kadowaki, Hiroaki Okazaki
    Journal of Atherosclerosis and Thrombosis 20(5) 481-493 2013年  査読有り
    marked hypertriglyceridemia resulting from impaired activation of lipoprotein lipase. In most cases of apoC-II deficiency, causative mutations have been found in the protein-coding region of APOC2 however, several atypical cases of apoC-II deficiency were reported to have markedly reduced, but detectable levels of plasma apoC-II protein (hereafter referred to as hypoapoC-II), which resulted from decreased promoter activity or improper splicing of apoC-II mRNA due to homozygous mutations in APOC2. Here we aim to dissect the molecular bases of a new case of hypoapoC-II. Methods: We performed detailed biochemical/genetic analyses of our new case of hypoapoC-II, manifesting severe hypertriglyceridemia (plasma triglycerides, 3235 mg·dL-1) with markedly reduced levels of plasma apoC-II (0.6 mg·dL-1). Results: We took advantage of a monocyte/macrophage culture system to prove that transcription of apoC-II mRNA was decreased in the patient's cells, which is compatible with the reported features of hypoapoC-II. Concomitantly, transcriptional activity of the minigene reporter construct of the patient's APOC2 gene was decreased however, no rare variant was detected in the patient's APOC2 gene. Fifty single nucleotide variants were detected in the patient's APOC2, but all were common variants (allele frequencies ≥35%) that are supposedly not causative. Conclusions: A case of apoC-II deficiency was found that is phenotypically identical to hypoapoC-II but with no causative mutations in APOC2, implying that other genes regulate apoC-II levels. The clinical entity of hypoapoC-II is discussed.
  • Takashi Matsuzaka, Ayaka Atsumi, Rie Matsumori, Tang Nie, Haruna Shinozaki, Noriko Suzuki-Kemuriyama, Motoko Kuba, Yoshimi Nakagawa, Kiyoaki Ishii, Masako Shimada, Kazuto Kobayashi, Shigeru Yatoh, Akimitsu Takahashi, Kazuhiro Takekoshi, Hirohito Sone, Naoya Yahagi, Hiroaki Suzuki, Soichiro Murata, Makoto Nakamuta, Nobuhiro Yamada, Hitoshi Shimano
    HEPATOLOGY 56(6) 2199-2208 2012年12月  査読有り
    Nonalcoholic steatohepatitis (NASH) is associated with obesity and type 2 diabetes, and an increased risk for liver cirrhosis and cancer. ELOVL family member 6, elongation of very long chain fatty acids (Elovl6), is a microsomal enzyme that regulates the elongation of C12-16 saturated and monounsaturated fatty acids (FAs). We have shown previously that Elovl6 is a major target for sterol regulatory element binding proteins in the liver and that it plays a critical role in the development of obesity-induced insulin resistance by modifying FA composition. To further investigate the role of Elovl6 in the development of NASH and its underlying mechanism, we used three independent mouse models with loss or gain of function of Elovl6, and human liver samples isolated from patients with NASH. Our results demonstrate that (1) Elovl6 is a critical modulator for atherogenic high-fat diet-induced inflammation, oxidative stress, and fibrosis in the liver; (2) Elovl6 expression is positively correlated with severity of hepatosteatosis and liver injury in NASH patients; and (3) deletion of Elovl6 reduces palmitate-induced activation of the NLR family pyrin domain-containing 3 inflammasome; this could be at least one of the underlying mechanisms by which Elovl6 modulates the progress of NASH. Conclusion: Hepatic long-chain fatty acid composition is a novel determinant in NASH development, and Elovl6 could be a potential therapeutic target for the prevention and treatment of NASH. (HEPATOLOGY 2012;56:2199-2208)
  • Yoriko Heianza, Yasuji Arase, Kazuya Fujhara, Shiun Dung Hsieh, Kazumi Saito, Hiroshi Tsuji, Satoru Kodama, Naoya Yahagi, Hitosh Shimano, Nobuhiro Yamada, Shigeko Hara, Hirohito Sone
    DIABETES CARE 35(5) 1050-1052 2012年5月  査読有り
    OBJECTIVE-To describe the trajectory of HbA(1c), and glucose concentrations before the diagnosis of diabetes. RESEARCH DESIGN AND METHODS-The study comprised 1,722 nondiabetic Japanese individuals aged 26-80 years. Fasting plasma glucose (FPG) and HbA(1c) were measured annually for a mean of 9.5 (SD 1.8) years. RESULTS-Diabetes occurred in 193 individuals (FPG >= 7.0 mmol/L, self-reported clinician-diagnosed diabetes, or HbA(1c) >= 6.5%). Mean HbA(1c) values were >5.6% each year before diagnosis in diabetes cases. Mean HbA(1c) (5.69% [95% CI 5.50-5.88]) was higher in the 21 individuals who developed diabetes 10 years after the baseline examination than in nondiabetic individuals after 10 years (5.27% [5.25-5.28]). From 3 years to 1 year prediagriosis, HbA(1c) increased 0.09% (SE 0.01)/year, reaching 5.90% (5.84-5.96) 1 year prediagnosis. In the entire group, marked increases in HbA(1c) of 0.3% (SE 0.05%)/year and FPG of 0.63 (0.07) mmol/L/year predicted diabetes. CONCLUSIONS HbA(1c) trajectory increased sharply after gradual long-term increases in diabetic individuals.
  • Hitoshi Iwasaki, Ayano Naka, Kaoruko Tada Iida, Yoshimi Nakagawa, Takashi Matsuzaka, Kiyo-aki Ishii, Kazuto Kobayashi, Akimitsu Takahashi, Shigeru Yatoh, Naoya Yahagi, Hirohito Sone, Hiroaki Suzuki, Nobuhiro Yamada, Hitoshi Shimano
    AMERICAN JOURNAL OF PHYSIOLOGY-ENDOCRINOLOGY AND METABOLISM 302(7) E896-E902 2012年4月  査読有り
    Iwasaki H, Naka A, Iida KT, Nakagawa Y, Matsuzaka T, Ishii K, Kobayashi K, Takahashi A, Yatoh S, Yahagi N, Sone H, Suzuki H, Yamada N, Shimano H. TFE3 regulates muscle metabolic gene expression, increases glycogen stores, and enhances insulin sensitivity in mice. Am J Physiol Endocrinol Metab 302: E896-E902, 2012. First published January 31, 2012; doi: 10.1152/ajpendo.00204.2011.-The role of transcription factor E3 (TFE3), a bHLH transcription factor, in immunology and cancer has been well characterized. Recently, we reported that TFE3 activates hepatic IRS-2 and hexokinase, participates in insulin signaling, and ameliorates diabetes. However, the effects of TFE3 in other organs are poorly understood. Herein, we examined the effects of TFE3 on skeletal muscle, an important organ involved in glucose metabolism. We generated transgenic mice that selectively express TFE3 in skeletal muscles. These mice exhibit a slight acceleration in growth prior to adulthood as well as a progressive increase in muscle mass. In TFE3 transgenic muscle, glycogen stores were more than twofold than in wild-type mice, and this was associated with an upregulation of genes involved in glucose metabolism, specifically glucose transporter 4, hexokinase II, and glycogen synthase. Consequently, exercise endurance capacity was enhanced in this transgenic model. Furthermore, insulin sensitivity was enhanced in transgenic mice and exhibited better improvement after 4 wk of exercise training, which was associated with increased IRS-2 expression. The effects of TFE3 on glucose metabolism in skeletal muscle were different from that in the liver, although they did, in part, overlap. The potential role of TFE3 in regulating metabolic genes and glucose metabolism within skeletal muscle suggests that it may be used for treating metabolic diseases as well as increasing endurance in sport.
  • Yuri Fujimoto, Yoshimi Nakagawa, Akiko Shingyouchi, Naoko Tokushige, Noriko Nakanishi, Aoi Satoh, Takashi Matsuzaka, Kiyo-aki Ishii, Hitoshi Iwasaki, Kazuto Kobayashi, Shigeru Yatoh, Hiroaki Suzuki, Naoya Yahagi, Osamu Urayama, Nobuhiro Yamada, Hitoshi Shimano
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 420(4) 931-936 2012年4月  査読有り
    Dicer is a rate-limiting enzyme for microRNA (miRNA) synthesis. To determine the effects of Dicer on adipogenesis, we performed stage-specific knockdown of Dicer using adenovirus encoding short-hairpin RNAi against Dicer in 3T3-L1 cells. When cells were infected with the adenovirus before induction of adipocyte differentiation, Dicer RNAi suppressed the gene expression of inducers of adipocyte differentiation such as PPAR gamma, C/EBP alpha, and FAS in 3T3-L1 cells during adipocyte differentiation. Concurrently, both adipocyte differentiation and cellular lipid accumulation were cancelled by Dicer RNAi when compared with control RNAi. Meanwhile, we addressed the roles of Dicer in lipid synthesis and accumulation in the final stages of differentiation. When the differentiated cells at day 4 after induction of differentiation were infected with adenovirus Dicer RNAi, cellular lipid accumulation was unchanged. Consistent with this, Dicer RNAi had no effects on the expression of genes related to cellular lipid accumulation, including PPAR gamma and FAS. Thus, Dicer controls proadipogenic genes such as C/EBP alpha and PPAR gamma in the early, but not in the late, stage of adipogenesis via regulation of miRNA synthesis. (C) 2012 Elsevier Inc. All rights reserved.
  • 岩崎 仁, 矢作 直也, 岩部 博子, 戸塚 久美子, 大畑 瞳, 須藤 玲子, 吉田 光歩, 伊藤 由実子, 鈴木 康裕, 松田 ひとみ, 高木 聡, 渋谷 正俊, 小林 和人, 矢藤 繁, 高橋 昭光, 鈴木 浩明, 山田 信博, 島野 仁
    糖尿病 55(Suppl.1) S-316 2012年4月  
  • Kumadaki S, Karasawa T, Matsuzaka T, Ema M, Nakagawa Y, Nakakuki M, Saito R, Yahagi N, Iwasaki H, Sone H, Takekoshi K, Yatoh S, Kobayashi K, Takahashi A, Suzuki H, Takahashi S, Yamada N, Shimano H
    The Journal of biological chemistry 286(47) 40835-40846 2011年11月  査読有り
  • Ryo Saito, Takashi Matsuzaka, Tadayoshi Karasawa, Motohiro Sekiya, Nazuki Okada, Masaki Igarashi, Rie Matsumori, Kiyoaki Ishii, Yoshimi Nakagawa, Hitoshi Iwasaki, Kazuto Kobayashi, Shigeru Yatoh, Akimitsu Takahashi, Hirohito Sone, Hiroaki Suzuki, Naoya Yahagi, Nobuhiro Yamada, Hitoshi Shimano
    ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY 31(9) 1973-U143 2011年9月  査読有り
    Objective-Elovl6, a long-chain fatty acid elongase, is a rate-limiting enzyme that elongates saturated and monounsaturated fatty acids and has been shown to be related to obesity-induced insulin resistance via modification of fatty acid composition. In this study, we investigated the roles of Elovl6 in foam cell formation in macrophages and atherosclerosis in mice. Methods and Results-To investigate the roles of Elovl6 in macrophages in the progression of atherosclerosis, we transplanted bone marrow cells of wild-type or Elovl6(-/-) mice into irradiated LDL-R(-/-) mice that were fed a western diet. Aortic atherosclerotic lesion areas and infiltration of macrophages were significantly smaller in Elovl6(-/-) bone marrow cells-transplanted LDL-R(-/-) mice than in wild-type. Accumulation of esterified cholesterol on exposure to acetylated-LDL was less severe in peritoneal macrophages from Elovl6(-/-) mice than those from wild-type. Cholesterol efflux and expression of cholesterol efflux transporters were increased in Elovl6(-/-) macrophages, although no difference in uptake of acetylated-LDL was found between the two groups. On analysis of fatty acid composition of the esterified cholesterol fraction in macrophages, n-6 polyunsaturated fatty acids were decreased by absence of Elovl6. Conclusion-These findings suggest that Elovl6 in macrophages may contribute to foam cell formation and progression of atherosclerosis. (Arterioscler Thromb Vasc Biol. 2011;31:1973-1979.)
  • Tadayoshi Karasawa, Akimitsu Takahashi, Ryo Saito, Motohiro Sekiya, Masaki Igarashi, Hitoshi Iwasaki, Shoko Miyahara, Saori Koyasu, Yoshimi Nakagawa, Kiyoaki Ishii, Takashi Matsuzaka, Kazuto Kobayashi, Naoya Yahagi, Kazuhiro Takekoshi, Hirohito Sone, Shigeru Yatoh, Hiroaki Suzuki, Nobuhiro Yamada, Hitoshi Shimano
    ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY 31(8) 1788-U201 2011年8月  査読有り
    Objective-Sterol regulatory element-binding protein-1 (SREBP-1) is nutritionally regulated and is known to be a key transcription factor regulating lipogenic enzymes. The goal of this study was to evaluate the roles of SREBP-1 in dyslipidemia and atherosclerosis. Methods and Results-Transgenic mice that overexpress SREBP-1c in the liver and SREBP-1-deficient mice were crossed with low-density lipoprotein receptor (LDLR)-deficient mice, and the plasma lipids and atherosclerosis were analyzed. Hepatic SREBP-1c overexpression in LDLR-deficient mice caused postprandial hypertriglyceridemia, increased very-low-density lipoprotein (VLDL) cholesterol, and decreased high-density lipoprotein cholesterol in plasma, which resulted in accelerated aortic atheroma formation. Conversely, absence of SREBP-1 suppressed Western diet-induced hyperlipidemia in LDLR-deficient mice and ameliorated atherosclerosis. In contrast, bone marrow-specific SREBP-1 deficiency did not alter the development of atherosclerosis. The size of nascent VLDL particles secreted from the liver was increased in SREBP-1c transgenic mice and reduced in SREBP-1-deficient mice, accompanied by upregulation and downregulation of phospholipid transfer protein expression, respectively. Conclusion-Hepatic SREBP-1c determines plasma triglycerides and remnant cholesterol and contributes to atherosclerosis in hyperlipidemic states. Hepatic SREBP-1c also regulates the size of nascent VLDL particles. (Arterioscler Thromb Vasc Biol. 2011;31:1788-1795.)
  • Michiyo Amemiya-Kudo, Junko Oka, Yoshinori Takeuchi, Hiroaki Okazaki, Takashi Yamamoto, Naoya Yahagi, Kaori Matsuzaka, Sachiko Okazaki, Jun-ichi Osuga, Nobuhiro Yamada, Toshio Murase, Hitoshi Shimano
    JOURNAL OF BIOLOGICAL CHEMISTRY 286(32) 27902-27914 2011年8月  査読有り
    Overexpression of sterol regulatory element-binding protein-1c (SREBP-1c) in beta cells causes impaired insulin secretion and beta cell dysfunction associated with diminished pancreatic duodenal homeodomain transcription factor-1 (PDX-1) expression in vitro and in vivo. To identify the molecular mechanism responsible for this effect, the mouse Pdx-1 gene promoter (2.7 kb) was analyzed in beta cell and non-beta cell lines. Despite no apparent sterol regulatory element-binding protein-binding sites, the Pdx-1 promoter was suppressed by SREBP-1c in beta cells in a dose-dependent manner. PDX-1 activated its own promoter. The E-box (-104/-99 bp) in the proximal region, occupied by ubiquitously expressed upstream stimulatory factors (USFs), was crucial for the PDX-1-positive autoregulatory loop through direct PDX-1.USF binding. This positive feedback activation was a prerequisite for SREBP-1c suppression of the promoter in non-beta cells. SREBP-1c and PDX-1 directly interact through basic helix-loop-helix and homeobox domains, respectively. This robust SREBP-1c.PDX-1 complex interferes with PDX-1.USF formation and inhibits the recruitment of PDX-1 coactivators. SREBP-1c also inhibits PDX-1 binding to the previously described PDX-1-binding site (-2721/-2646 bp) in the distal enhancer region of the Pdx-1 promoter. Endogenous up-regulation of SREBP-1c in INS-1 cells through the activation of liver X receptor and retinoid X receptor by 9-cis-retinoic acid and 22-hydroxycholesterol inhibited PDX-1 mRNA and protein expression. Conversely, SREBP-1c RNAi restored Pdx-1 mRNA and protein levels. Through these multiple mechanisms, SREBP-1c, when induced in a lipotoxic state, repressed PDX-1 expression contributing to the inhibition of insulin expression and beta cell dysfunction.
  • Keisuke Ohta, Motohiro Sekiya, Hiroshi Uozaki, Masaki Igarashi, Satoru Takase, Masayoshi Kumagai, Mikio Takanashi, Yoshinori Takeuchi, Yoshihiko Izumida, Midori Kubota, Makiko Nishi, Hiroaki Okazaki, Yoko Iizuka, Naoya Yahagi, Hiroaki Yagyu, Masashi Fukayama, Takashi Kadowaki, Ken Ohashi, Shun Ishibashi, Jun-ichi Osuga
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 404(1) 254-260 2011年1月  査読有り
    We have previously demonstrated that neutral cholesterol ester hydrolase 1 (Nceh1) regulates foam cell formation and atherogenesis through the catalytic activity of cholesterol ester hydrolysis, and that Nceh1 and hormone-sensitive lipase (Lipe) are responsible for the majority of neutral cholesterol ester hydrolase activity in macrophages. There are several cholesterol ester-metabolizing tissues and cells other than macrophages, among which adrenocortical cells are also known to utilize the intracellular cholesterol for steroidogenesis. It has been believed that the mobilization of intracellular cholesterol ester in adrenal glands was facilitated solely by Lipe. We herein demonstrate that Nceh1 is also involved in cholesterol ester hydrolysis in adrenal glands. While Lipe deficiency remarkably reduced the neutral cholesterol ester hydrolase activity in adrenal glands as previously reported, additional inactivation of Nceh1 gene completely abrogated the activity. Adrenal glands were enlarged in proportion to the degree of reduced neutral cholesterol ester hydrolase activity, and the enlargement of adrenal glands and the accumulation of cholesterol esters were most pronounced in the Nceh1/Lipe double-deficient mice. Thus Nceh1 is involved in the adrenal cholesterol metabolism, and the cholesterol ester hydrolytic activity in adrenal glands is associated with the organ enlargement. (C) 2010 Elsevier Inc. All rights reserved.
  • Takashi Yamamoto, Kazuhisa Watanabe, Noriyuki Inoue, Yoshimi Nakagawa, Naomi Ishigaki, Takashi Matsuzaka, Yoshinori Takeuchi, Kazuto Kobayashi, Shigeru Yatoh, Akimitsu Takahashi, Hiroaki Suzuki, Naoya Yahagi, Takanari Gotoda, Nobuhiro Yamada, Hitoshi Shimano
    JOURNAL OF LIPID RESEARCH 51(7) 1859-1870 2010年7月  査読有り
    Sterol-regulatory element binding protein-1c (SREBP-1c) is a transcription factor that controls lipogenesis in the liver. Hepatic SREBP-1c is nutritionally regulated, and its sustained activation causes hepatic steatosis and insulin resistance. Although regulation of SREBP-1c is known to occur at the transcriptional level, the precise mechanism by which insulin signaling activates SREBP-1c promoter remains to be elucidated. Here we show that protein kinase C beta (PKCbeta) is a key mediator of insulin-mediated activation of hepatic SREBP-1c and its target lipogenic genes. Activation of SREBP-1c in the liver of refed mice was suppressed by either adenoviral RNAi-mediated knockdown or dietary administration of a specific inhibitor of protein kinase C beta. The effect of PKCbeta inhibition was cancelled in insulin depletion by streptozotocin (STZ) treatment of mice. Promoter analysis indicated that PKCbeta activates SREBP-1c promoter through replacement of Sp3 by Sp1 for binding to the GC box in the sterol regulatory element (SRE) complex, a key cis-element of SREBP-1c promoter. Knockdown of Sp proteins demonstrated that Sp3 and Sp1 play reciprocally negative and positive roles in nutritional regulation of SREBP-1c, respectively. This new understanding of PKCbeta involvement in nutritional regulation of SREBP-1c activation provides a new aspect of PKCbeta inhibition as a potential therapeutic target for diabetic complications.-Yamamoto, T., K. Watanabe, N. Inoue, Y. Nakagawa, N. Ishigaki, T. Matsuzaka, Y. Takeuchi, K. Kobayashi, S. Yatoh, A. Takahashi, H. Suzuki, N. Yahagi, T. Gotoda, N. Yamada, and H. Shimano. Protein kinase Cbeta mediates hepatic induction of sterol-regulatory element binding protein-1c by insulin. J Lipid Res. 2010. 51: 1859-1870.
  • Yoshinori Takeuchi, Naoya Yahagi, Yoshihiko Izumida, Makiko Nishi, Midori Kubota, Yuji Teraoka, Takashi Yamamoto, Takashi Matsuzaka, Yoshimi Nakagawa, Motohiro Sekiya, Yoko Iizuka, Ken Ohashi, Jun-ichi Osuga, Takanari Gotoda, Shun Ishibashi, Keiji Itaka, Kazunori Kataoka, Ryozo Nagai, Nobuhiro Yamada, Takashi Kadowaki, Hitoshi Shimano
    JOURNAL OF BIOLOGICAL CHEMISTRY 285(15) 11681-11691 2010年4月  査読有り責任著者
    Sterol regulatory element-binding protein (SREBP)-1 is a key transcription factor for the regulation of lipogenic enzyme genes in the liver. Polyunsaturated fatty acids (PUFA) selectively suppress hepatic SREBP-1, but molecular mechanisms remain largely unknown. To gain insight into this regulation, we established in vivo reporter assays to assess the activities of Srebf1c transcription and proteolytic processing. Using these in vivo reporter assays, we showed that the primary mechanism for PUFA suppression of SREBP-1 is at the proteolytic processing level and that this suppression in turn decreases the mRNA transcription through lowering SREBP-1 binding to the SREBP-binding element on the promoter ("autoloop regulatory circuit"), although liver X receptor, an activator for Srebf1c transcription, is not involved in this regulation by PUFA. The mechanisms for PUFA suppression of SREBP-1 confirm that the autoloop regulation for transcription is crucial for the nutritional regulation of triglyceride synthesis.
  • Motohiro Sekiya, Naoya Yahagi, Yoshiaki Tamura, Hiroaki Okazaki, Masaki Igarashi, Keisuke Ohta, Mikio Takanashi, Masayoshi Kumagai, Satoru Takase, Makiko Nishi, Yoshinori Takeuchi, Yoshihiko Izumida, Midori Kubota, Ken Ohashi, Yoko Iizuka, Hiroaki Yagyu, Takanari Gotoda, Ryozo Nagai, Hitoshi Shimano, Nobuhiro Yamada, Takashi Kadowaki, Shun Ishibashi, Jun-ichi Osuga
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 387(3) 511-515 2009年9月  査読有り責任著者
    It has long been a matter of debate whether the hormone-sensitive lipase (HSL)-mediated lipolysis in pancreatic beta-cells can affect insulin secretion through the alteration of lipotoxicity. We generated mice lacking both leptin and HSL (Lep(ob/ob)/HSL(-/-)) and explored the role of HSL in pancreatic beta-cells in the setting of obesity. Lep(ob/ob)/HSL(-/-) developed elevated blood glucose levels and reduced plasma insulin levels compared with Lep(ob/ob)/HSL(+/+) in a fed state, while the deficiency of HSL did not affect glucose homeostasis in Lep(+/+) background. The deficiency of HSL exacerbated the accumulation of triglycerides in Lep(ob/ob) islets, leading to reduced glucose-stimulated insulin secretion. The deficiency of HSL also diminished the islet mass in Lep(ob/ob) mice due to decreased cell proliferation. In conclusion, HSL affects insulin secretary capacity especially in the setting of obesity. (C) 2009 Elsevier Inc. All rights reserved.
  • Motohiro Sekiya, Jun-ichi Osuga, Shuichi Nagashima, Taichi Ohshiro, Masaki Igarashi, Hiroaki Okazaki, Manabu Takahashi, Fumiko Tazoe, Taeko Wada, Keisuke Ohta, Mikio Takanashi, Masayoshi Kumagai, Makiko Nishi, Satoru Takase, Naoya Yahagi, Hiroaki Yagyu, Ken Ohashi, Ryozo Nagai, Takashi Kadowaki, Yusuke Furukawa, Shun Ishibashi
    CELL METABOLISM 10(3) 219-228 2009年9月  査読有り
    Cholesterol ester (CE)-laden macrophage foam cells are the hallmark of atherosclerosis, and the hydrolysis of intracellular CE is one of the key steps in foam cell formation. Although hormone-sensitive lipase (LIPE) and cholesterol ester hydrolase (CEH), which is identical to carboxylsterase 1 (CES1, hCE1), were proposed to mediate the neutral CE hydrolase (nCEH) activity in macrophages, recent evidences have suggested the involvement of other enzymes. We have recently reported the identification of a candidate, neutral cholesterol ester hydrolase 1 (Nceh1). Here we demonstrate that genetic ablation of Nceh1 promotes foam cell formation and the development of atherosclerosis in mice. We further demonstrate that Nceh1 and Lipe mediate a comparable degree of nCEH activity in macrophages; and together account for most of the activity. Mice lacking both Nceh1 and Lipe aggravated atherosclerosis in an additive manner. Thus, Nceh1 is a promising target for the treatment of atherosclerosis.
  • Noriko Nakanishi, Yoshimi Nakagawa, Naoko Tokushige, Naohito Aoki, Takashi Matsuzaka, Kiyoaki Ishii, Naoya Yahagi, Kazuto Kobayashi, Shigeru Yatoh, Akimitsu Takahashi, Hiroaki Suzuki, Osamu Urayama, Nobuhiro Yamada, Hitoshi Shimano
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 385(4) 492-496 2009年8月  査読有り
    MicroRNAs (miRNAs) are short non-coding RNA that post-transcriptionally regulates gene expression. Some miRNAs have been proposed to be associated with obesity. However, miRNAs, which are related to the development of obesity in vivo remains unknown. Here in, we found the up-regulation of miR-335 in obesity using microarray analysis for miRNA. The expressions of miR-335 in liver and white adipose tissue (WAT) were up-regulated in obese mice including ob/ob, db/db, and KKAy mice. Increased miR-335 expressions were associated with an elevated body, liver and WAT weight, and hepatic triglyceride and cholesterol. Furthermore, miR-335 levels were closely correlated with expression levels of adipocyte differentiation markers such as PPAR gamma, aP2, and FAS in 3T3-L1 adipocyte. These findings provide the first evidence that the up-regulated expressions of miR-335 in liver and WAT of obese mice might contribute to the pathophysiology of obesity. (c) 2009 Published by Elsevier Inc.
  • Makiko Hongo, Nobukazu Ishizaka, Kyoko Furuta, Naoya Yahagi, Kan Saito, Ryota Sakurai, Gen Matsuzaki, Kazuhiko Koike, Ryozo Nagai
    EUROPEAN JOURNAL OF PHARMACOLOGY 604(1-3) 87-92 2009年2月  査読有り
    Accumulation of lipids in the heart may cause cardiac dysfunction in various disorders, such as obesity and diabetes. In the current study, we have investigated whether administration of angiotensin II or norepinephrine induces accumulation of lipids and/or changes in the expression of genes related to lipid metabolism in the rat heart. Lipid deposition was found in myocardial, vascular wall, and perivascular cells of the angiotensin II-infused rat heart, and superoxide generation was increased in these lipid-positive cells. By contrast, intracardiac lipid deposition was not found in the heart of norepinephrine-induced hypertensive rats. Triglyceride content in the heart tissue of angiotensin II-infused rats increased more than 3-fold as compared with untreated controls. Losartan completely, but hydralazine only partially, suppressed the angiotensin II-induced intracardiac lipid deposition and increase in tissue triglyceride content. Administration of angiotensin II upregulated the mRNA expression of sterol regulatory element-binding protein-1c and fatty acid synthase, but downregulated that of uncoupling protein 2 and 3, in a manner dependent on the angiotensin AT, receptor. Collectively, these results suggest that angiotensin H may be involved in modulating both intracardiac lipid content and lipid metabolism-related gene expression, in part via an angiotensin AT, receptor-dependent and pressor-independent mechanism. (C) 2009 Elsevier B.V. All rights reserved.
  • Yuko Iwasaki, Hitoshi Iwasaki, Shigeru Yatoh, Mayumi Ishikawa, Toyonori Kato, Takashi Matsuzaka, Yoshimi Nakagawa, Naoya Yahagi, Kazuto Kobayashi, Akimitsu Takahashi, Hiroaki Suzuki, Nobuhiro Yamada, Hitoshi Shimano
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 378(3) 545-550 2009年1月  査読有り
    Transgenic mice expressing nuclear sterol regulatory element-binding protein-1a I a under the control of the insulin promoter were generated to determine the role of SREBP-la in pancreatic beta-cells. Only low expressors could be established, which exhibited mild hyperglycemia, impaired glucose tolerance, and reduced plasma insulin levels compared to C57BL/G controls. The islets isolated from the transgenic mice were fewer and smaller, and had decreased insulin content and unaltered glucagon staining. Both glucose- and potassium-stimulated insulin secretions were decreased. The transgenic islets consistently expressed genes for fatty acids and cholesterol synthesis, resulting in accumulation of triglycerides but not cholesterol. PDX-1, BETA2, MafA, and IRS-2 were suppressed, partially explaining the loss and dysfunction of beta-cell.mass. The transgenic mice on a high fat/high Sucrose diet still exhibited impaired insulin secretion and continuous P-cell growth defect. Therefore, nuclear SREBP-la, even at a low level, strongly disrupts p-cell mass and function. (C) 2008 Elsevier Inc. All rights reserved.
  • Mayumi Ishikawa, Yuko Iwasaki, Shigeru Yatoh, Toyonori Kato, Shin Kumadaki, Noriyuki Inoue, Takashi Yamamoto, Takashi Matsuzaka, Yoshimi Nakagawa, Naoya Yahagi, Kazuto Kobayashi, Akimitsu Takahashi, Nobuhiro Yamada, Hitoshi Shimano
    JOURNAL OF LIPID RESEARCH 49(12) 2524-2534 2008年12月  査読有り
    To determine the role of cholesterol synthesis in pancreatic beta-cells, a transgenic model of in vivo activation of sterol-regulatory element binding protein 2 (SREBP-2) specifically in beta-cells (TgRIP-SREBP-2) was developed and analyzed. Expression of nuclear human SREBP-2 in beta-cells resulted in severe diabetes as evidenced by greater than 5-fold elevations in glycohemoglobin compared with C57BL/6 controls. Diabetes in TgRIP-SREBP-2 mice was primarily due to defects in glucose-and potassium-stimulated insulin secretion as determined by glucose tolerance test. Isolated islets of TgSREBP-2 mice were fewer in number, smaller, deformed, and had decreased insulin content. SREBP-2-expressing islets also contained increased esterified cholesterol and unchanged triglycerides with reduced ATP levels. Consistently, these islets exhibited elevated expression of HMG-CoA synthase and reductase and LDL receptor, with suppression of endogenous SREBPs. Genes involved in beta-cell differentiation, such as PDX1 and BETA2, were suppressed, explaining loss of beta-cell mass, whereas IRS2 expression was not affected. These phenotypes were dependent on the transgene expression. Taken together, these results indicate that activation of SREBP-2 in beta-cells caused severe diabetes by loss of beta-cell mass with accumulation of cholesterol, providing a new lipotoxic model and a potential link of disturbed cholesterol metabolism to impairment of beta-cell function.-Ishikawa, M., Y. Iwasaki, S. Yatoh, T. Kato, S. Kumadaki, N. Inoue, T. Yamamoto, T. Matsuzaka, Y. Nakagawa, N. Yahagi, K. Kobayashi, A. Takahashi, N. Yamada, and H. Shimano. Cholesterol accumulation and diabetes in pancreatic beta-cell-specific SREBP-2 transgenic mice: a new model for lipotoxicity. J. Lipid Res. 2008. 49: 2524-2534.
  • Hiroaki Okazaki, Masaki Igarashi, Makiko Nishi, Motohiro Sekiya, Makiko Tajima, Satoru Takase, Mikio Takanashi, Keisuke Ohta, Yoshiaki Tamura, Sachiko Okazaki, Naoya Yahagi, Ken Ohashi, Michiyo Amemiya-Kudo, Yoshimi Nakagawa, Ryozo Nagai, Takashi Kadowaki, Jun-ichi Osuga, Shun Ishibashi
    JOURNAL OF BIOLOGICAL CHEMISTRY 283(48) 33357-33364 2008年11月  査読有り
    Unstable lipid-rich plaques in atherosclerosis are characterized by the accumulation of macrophage foam cells loaded with cholesterol ester (CE). Although hormone-sensitive lipase and cholesteryl ester hydrolase (CEH) have been proposed to mediate the hydrolysis of CE in macrophages, circumstantial evidence suggests the presence of other enzymes with neutral cholesterol ester hydrolase (nCEH) activity. Here we show that the murine orthologue of KIAA1363, designated as neutral cholesterol ester hydrolase (NCEH), is a microsomal nCEH with high expression in murine and human macrophages. The effect of various concentrations of NaCl on its nCEH activity resembles that on endogenous nCEH activity of macrophages. RNA silencing of NCEH decreases nCEH activity at least by 50%; conversely, its overexpression inhibits the CE formation in macrophages. Immunohistochemistry reveals that NCEH is expressed in macrophage foam cells in atherosclerotic lesions. These data indicate that NCEH is responsible for a major part of nCEH activity in macrophages and may be a potential therapeutic target for the prevention of atherosclerosis.
  • Toyonori Kato, Hitoshi Shimano, Takashi Yamamoto, Mayumi Ishikawa, Shin Kumadaki, Takashi Matsuzaka, Yoshimi Nakagawa, Naoya Yahagi, Masanori Nakakuki, Alyssa H. Hasty, Yoshinori Takeuchi, Kazuto Kobayashi, Akimitsu Takahashi, Shigeru Yatoh, Hiroaki Suzuki, Hirohito Sone, Nobuhiro Yamada
    DIABETES 57(9) 2382-2392 2008年9月  査読有り
    OBJECTIVE-Chronic exposure to fatty acids causes beta-cell failure, often referred to as lipotoxicity. We investigated its mechanisms, focusing on contribution of SREBP-1c, a key transcription factor for Lipogenesis. RESEARCH DESIGN AND METHODS-We studied in vitro and in vivo effects of saturated and polyunsaturated acids on insulin secretion, insulin signaling, and expression of genes involved in beta-cell functions. Pancreatic islets isolated from C57BL/6 control and SREBP-1-null mice and adenoviral gene delivery or knockdown systems of related genes were used. RESULTS-Incubation of C57BL/6 islets with palmitate caused inhibition of both glucose- and potassium-stimidated insulin secretion, but addition of eicosapentaenoate (EPA) restored both inhibitions. Concomitantly, palmitate activated and EPA abolished both mRNA and nuclear protein of SREBP-1c, accompanied by reciprocal changes of SREBP-1c target genes such as insulin receptor substrate-2 (IRS-2) and granuphilin. These palmitate-EPA effects on insulin secretion were abolished in SREBP-1-null islets. Suppression of IRS-2/Akt pathway could be a part of the downstream mechanism for the SREBP-1c-mediated insulin secretion defect because adenoviral constitutively active Akt compensated it. Uncoupling protein-2 (UCP-2) also plays a crucial role in the palmitate inhibition of insulin secretion, as confirmed by knockdown experiments, but SREBP-1c contribution to UCP-2 regulation was partial. The palmitate-EPA regulation of insulin secretion was similarly observed in islets from C57BL/6 mice pretreated with dietary manipulations. Furthermore, administration of EPA to diabetic KK-Ay mice ameliorated impairment of insulin secretion in their islets. CONCLUSIONS-SREBP-1c plays a dominant role in palmitate-mediated insulin secretion defect, and EPA prevents it through SREBP-1c inhibition, implicating a therapeutic potential for treating diabetes related to lipotoxicity.
  • Motohiro Sekiya, Jun-ichi Osuga, Naoya Yahagi, Hiroaki Okazaki, Yoshiaki Tamura, Masaki Igarashi, Satoru Takase, Kenji Harada, Sachiko Okazaki, Yoko Iizuka, Ken Ohashi, Hiroaki Yagyu, Mitsuyo Okazaki, Takanari Gotoda, Ryozo Nagai, Takashi Kadowaki, Hitoshi Shimano, Nobuhiro Yamada, Shun Ishibashi
    JOURNAL OF LIPID RESEARCH 49(8) 1829-1838 2008年8月  査読有り
    Hormone-sensitive lipase (HSL) regulates the hydrolysis of acylglycerol and cholesteryl ester (CE) in various organs, including adipose tissues. However, the hepatic expression level of HSL has been reported to be almost negligible. In the present study, we found that mice lacking both leptin and HSL (Lep(ob/ob)/HSL-/-) showed massive accumulation of CE in the liver compared with Lep(ob/ob)/HSL+/+ mice, while triacylglycerol (TG) accumulation was modest. Similarly, feeding with a high-cholesterol diet induced hepatic CE accumulation in HSL-/- mice. Supporting these observations, we detected significant expression of protein as well as mRNA of HSL in the liver. HSL-/- mice showed reduced activity of CE hydrolase, but not of TG lipase, in the liver compared with wild-type mice. Furthermore, we confirmed the expression of HSL in viable parenchymal cells isolated from wild-type mice. The hepatocytes from HSL-/- mice showed reduced activity of CE hydrolase and contained more CE than those from HSL-/- mice even without the incubation with lipoproteins. Incubation with LDL further augmented the accumulation of CE in the HSL-deficient hepatocytes. From these results, we conclude that HSL is involved in the hydrolysis of CE in hepatocyes.
  • Noriyuki Inoue, Naoya Yahagi, Takashi Yamamoto, Mayumi Ishikawa, Kazuhisa Watanabe, Takashi Matsuzaka, Yoshimi Nakagawa, Yoshinori Takeuchi, Kazuto Kobayashi, Akimitsu Takahashi, Hiroaki Suzuki, Alyssa H. Hasty, Hideo Toyoshima, Nobuhiro Yamada, Hitoshi Shimano
    JOURNAL OF BIOLOGICAL CHEMISTRY 283(30) 21220-21229 2008年7月  査読有り
    Both adipocyte hyperplasia and hypertrophy are determinant factors for adipocyte differentiation during the development of obesity. p21(WAF1/CIP1), a cyclin-dependent kinase inhibitor, is induced during adipocyte differentiation; however, its precise contribution to this process is unknown. Using both in vitro and in vivo systems, we show that p21 is crucial for maintaining adipocyte hypertrophy and obesity-induced insulin resistance. The absence of p21 in 3T3-L1 fibroblasts by RNA-mediated interference knockdown or in embryonic fibroblasts from p21(-/-) mice impaired adipocyte differentiation, resulting in smaller adipocytes. Despite normal adipose tissue mass on a normal diet, p21(-/-) mice fed high energy diets had reduced adipose tissue mass and adipocyte size accompanied by a marked improvement in insulin sensitivity. Knockdown of p21 in enlarged epididymal fat of diet-induced obese mice and also in fully differentiated 3T3-L1 adipocytes caused vigorous apoptosis by activating p53. Thus, p21 is involved in both adipocyte differentiation and in protecting hypertrophied adipocytes against apoptosis. Via both of these mechanisms, p21 promotes adipose tissue expansion during high fat diet feeding, leading to increased downstream pathophysiological consequences such as insulin resistance.
  • Shin Kumadaki, Takashi Matsuzaka, Toyonori Kato, Naoya Yahagi, Takashi Yamamoto, Sumiyo Okada, Kazuto Kobayashi, Akimitsu Takahashi, Shigeru Yatoh, Hiroaki Suzuki, Nobuhiro Yamada, Hitoshi Shimano
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 368(2) 261-266 2008年4月  査読有り
    Elovl-6, a long fatty acid elongase, contributes to de novo synthesis of fatty acids and regulates hepatic insulin sensitivity. Hepatic regulation of Elovl-6 gene expression in various nutritional conditions suggested that, like other lipogenic enzyme genes, Elovl-6 is a target of SREBP-1, a transcription factor governing fatty acid synthesis. Supportively, adenoviral RNAi knockdown of SREBP-1 in mouse liver suppressed Elovl-6 mRNA and fatty acid synthase levels. Therefore, we analyzed mouse Elovl-6 gene promoter to determine its role as an SREBP-1 target. Luciferase reporter assays of 1.4-kb 5' flanking region of mouse Elovl-6 gene in HepG2 cells demonstrated that nuclear SREBPs activated the Elovl-6 promoter, highlighting two SREBP binding sites: proximal SRE-1 and distal SRE-2. EMSA indicated that SRE-1 had higher affinity than SRE-2 for SREBP. ChIP assays confirmed in vivo binding of hepatic nuclear SREBP-1c protein. These data demonstrated that Elovl-6 is regulated directly and primarily by SREBP-1c. (C) 2008 Elsevier Inc. All rights reserved.
  • Ishikawa, M, Iwasaki, Y, Yatoh, S, Kato, T, Kumadaki, S, Inoue, N, Yamamoto, T, Matsuzaka, T, Nakagawa, Y, Yahagi, N, Kobayashi, K, Takahashi, A, Yamada, N, Shimano, H
    J Lipid Res. 49(12) 2524-34 2008年1月  査読有り
  • Yoshinori Takeuchi, Naoya Yahagi, Yoshimi Nakagawa, Takashi Matsuzaka, Ritsuko Shimizu, Motohiro Sekiya, Yoko Iizuka, Ken Ohashi, Takanari Gotoda, Masayuki Yamamoto, Ryozo Nagai, Takashi Kadowaki, Nobuhiro Yamada, Jun-ichi Osuga, Hitoshi Shimano
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 363(2) 329-335 2007年11月  査読有り
    Sterol regulatory element-binding protein (SREBP)-1c is the master regulator of lipogenic gene expression in liver. The mRNA abundance of SREBP- 1c is markedly induced when animals are refed after starvation, although the regulatory mechanism is so far unknown. To investigate the mechanism of refeeding response of SREBP-1c gene expression in vivo, we generated a transgenic mouse model that carries 2.2 kb promoter region fused to the luciferase reporter gene. These transgenic mice exhibited refeeding responses of the reporter in liver and adipose tissues with extents essentially identical to those of endogenous SREBP-1c mRNA. The same results were obtained from experiments using adenovirus-mediated SREBP-1c-promoter-luciferase fusion gene transduction to liver. These data demonstrate that the regulation of SREBP-1c gene expression is at the transcription level, and that the 2.2 kb 5'-flanking region is sufficient for this regulation. Moreover, when these transgenic or adenovirus-infected mice were placed on insulin-depleted state by streptozotocin treatment, the reporter expression was upregulated as strongly as in control mice, demonstrating that this regulation is not dominated by serum insulin level. These mice are the first models to provide the mechanistic insight into the transcriptional regulation of SREBP1c gene in vivo. (C) 2007 Elsevier Inc. All rights reserved.
  • Takashi Matsuzaka, Hitoshi Shimano, Naoya Yahagi, Toyonori Kato, Ayaka Atsumi, Takashi Yamamoto, Noriyuki Inoue, Mayumi Ishikawa, Sumiyo Okada, Naomi Ishigaki, Hitoshi Iwasaki, Yuko Iwasaki, Tadayoshi Karasawa, Shin Kumadaki, Toshiyuki Matsui, Motohiro Sekiya, Ken Ohashi, Alyssa H. Hasty, Yoshimi Nakagawa, Akimitsu Takahashi, Hiroaki Suzuki, Sigeru Yatoh, Hirohito Sone, Hideo Toyoshima, Jun-ichi Osuga, Nobuhiro Yamada
    NATURE MEDICINE 13(10) 1193-1202 2007年10月  査読有り
    Insulin resistance is often associated with obesity and can precipitate type 2 diabetes. To date, most known approaches that improve insulin resistance must be preceded by the amelioration of obesity and hepatosteatosis. Here, we show that this provision is not mandatory; insulin resistance and hyperglycemia are improved by the modification of hepatic fatty acid composition, even in the presence of persistent obesity and hepatosteatosis. Mice deficient for Elovl6, the gene encoding the elongase that catalyzes the conversion of palmitate to stearate, were generated and shown to become obese and develop hepatosteatosis when fed a high- fat diet or mated to leptin- deficient ob/ ob mice. However, they showed marked protection from hyperinsulinemia, hyperglycemia and hyperleptinemia. Amelioration of insulin resistance was associated with restoration of hepatic insulin receptor substrate- 2 and suppression of hepatic protein kinase C e activity resulting in restoration of Akt phosphorylation. Collectively, these data show that hepatic fatty acid composition is a new determinant for insulin sensitivity that acts independently of cellular energy balance and stress. Inhibition of this elongase could be a new therapeutic approach for ameliorating insulin resistance, diabetes and cardiovascular risks, even in the presence of a continuing state of obesity.
  • Masanori Nakakuki, Hitoshi Shimano, Noriyuki Inoue, Mariko Tamura, Takashi Matsuzaka, Yoshimi Nakagawa, Naoya Yahagi, Hideo Toyoshima, Ryuichiro Sato, Nobuhiro Yamada
    FEBS JOURNAL 274(17) 4440-4452 2007年9月  査読有り
    Sterol regulatory element-binding protein (SREBP)-1a is a unique membrane-bound transcription factor highly expressed in actively growing cells and involved in the biosynthesis of cholesterol, fatty acids, and phospholipids. Because mammalian cells need to synthesize membrane lipids for cell replication, the functional relevance of SREBP-1a in cell proliferation has been considered a biological adaptation. However, the effect of this potent lipid-synthesis activator on cell growth has never been explored. Here, we show that induction of nuclear SREBP-1a, but not SREBP-2, completely inhibited cell growth in inducible Chinese hamster ovary (CHO) cell lines. Growth inhibition occurred through G(1) cell-cycle arrest, which is observed in various cell types with transient expression of nuclear SREBP-1a. SREBP-1a caused the accumulation of cyclin-dependent kinase (cdk) inhibitors such as p27, p21, and p16, leading to reduced cdk2 and cdk4 activities and hypophosphorylation of Rb protein. In contrast to transactivation of p21, SREBP-1a activated p27 by enhancing stabilization of the protein through inhibition of SKP2 and KPC1. In vivo, SREBP-1a-expressing livers of transgenic mice exhibited impaired regeneration after partial hepatectomy. SREBP-1-null mouse embryonic fibroblasts had a higher cell proliferation rate than wild-type cells. The unexpected cell growth-inhibitory role of SREBP-1a provides a new paradigm to link lipid synthesis and cell growth.
  • Motohiro Sekiya, Naoya Yahagi, Takashi Matsuzaka, Yoshinori Takeuchi, Yoshimi Nakagawa, Haruka Takahashi, Hiroaki Okazaki, Yoko Iizuka, Ken Ohashi, Takanari Gotoda, Shun Ishibashi, Ryozo Nagai, Tsutomu Yamazaki, Takashi Kadowaki, Nobuhiro Yamada, Jun-ichi Osuga, Hitoshi Shimano
    JOURNAL OF LIPID RESEARCH 48(7) 1581-1591 2007年7月  査読有り
    Sterol regulatory element-binding protein (SREBP)-1c is now well established as a key transcription factor for the regulation of lipogenic enzyme genes such as FAS in hepatocytes. Meanwhile, the mechanisms of lipogenic gene regulation in adipocytes remain unclear. Here, we demonstrate that those in adipocytes are independent of SREBP-1c. In adipocytes, unlike in hepatocytes, the stimulation of SREBP-1c expression by liver X receptor agonist does not accompany lipogenic gene upregulation, although nuclear SREBP-1c protein is concomitantly increased, indicating that the activation process of SREBP-1c by the cleavage system is intact in adipocytes. Supportively, transcriptional activity of the mature form of SREBP-1c for the FAS promoter was negligible when measured by reporter analysis. As an underlying mechanism, accessibility of SREBP-1c to the functional elements was involved, because chromatin immunoprecipitation assays revealed that SREBP-1c does not bind to the functional SRE/E-box site on the FAS promoter in adipocytes. Moreover, genetic disruption of SREBP-1 did not cause any changes in lipogenic gene expression in adipose tissue. In summary, in adipocytes, unlike in hepatocytes, increments in nuclear SREBP-1c are not accompanied by transactivation of lipogenic genes; thus, SREBP-1c is not committed to the regulation of lipogenesis.
  • Takashi Yamamoto, Hitoshi Shimano, Noriyuki Inoue, Yoshimi Nakagawa, Takashi Matsuzaka, Akimitsu Takahashi, Naoya Yahagi, Hirohito Sone, Hiroaki Suzuki, Hideo Toyoshima, Nobuhiro Yamada
    JOURNAL OF BIOLOGICAL CHEMISTRY 282(16) 11687-11695 2007年4月  査読有り
    Sterol regulatory element-binding protein ( SREBP)-1c is a transcription factor that controls synthesis of fatty acids and triglycerides in the liver and is highly regulated by nutrition and hormones. In the current studies we show that protein kinase A ( PKA), a mediator of glucagon/cAMP, a fasting signaling, suppresses SREBP-1c by modulating the activity of liver X receptor alpha( LXR alpha), a dominant activator of SREBP-1c expression. Activation of PKA repressed LXR-induced SREBP-1c expression both in rat primary hepatocytes and mouse livers. Promoter analyses revealed that the LXR alpha-binding site in the SREBP-1c promoter is responsible for PKA inhibitory effect on SREBP-1c transcription. In vitro and in vivo PKA directly phosphorylated LXR alpha, and the two consensus PKA target sites ( 195, 196 serines and 290, 291 serines) in its ligand binding/heterodimerization domain were crucial for the inhibition of LXR signaling. PKA phosphorylation of LXR alpha caused impaired DNA binding activity by preventing LXR alpha/RXR dimerization and decreased its transcription activity by inhibiting recruitment of coactivator SCR-1 and enhancing recruitment of corepressor NcoR1. These results indicate that LXR alpha is regulated not only by oxysterol derivatives but also by PKA-mediated phosphorylation, which suggests that nutritional regulation of SREBP-1c and lipogenesis could be regulated at least partially through modulation of LXR.
  • Hiroaki Okazaki, Fumiko Tazoe, Sachiko Okazaki, Naoyuki Isoo, Kazuhisa Tsukamoto, Motohiro Sekiya, Naoya Yahagi, Yoko Iizuka, Ken Ohashi, Tetsuya Kitamine, Ryu-ichi Tozawa, Toshihiro Inaba, Hiroaki Yagyu, Mitsuyo Okazaki, Hitoshi Shimano, Norihito Shibata, Hiroyuki Arai, Ryo-zo Nagai, Takashi Kadowaki, Jun-ichi Osuga, Shun Ishibashi
    JOURNAL OF LIPID RESEARCH 47(9) 1950-1958 2006年9月  査読有り
    Squalene synthase (SS) is the first committed enzyme for cholesterol biosynthesis, located at a branch point in the mevalonate pathway. To examine the role of SS in the overall cholesterol metabolism, we transiently over-expressed mouse SS in the livers of mice using adenovirus-mediated gene transfer. Overexpression of SS increased de novo cholesterol biosynthesis with increased 3-hydroxy-3-methyglutaryl-CoA (HMG-CoA) reductase activity, in spite of the downregulation of its own mRNA expression. Furthermore, overexpression of SS increased plasma concentrations of LDL, irrespective of the presence of functional LDL receptor (LDLR). Thus, the hypercholesterolemia is primarily caused by increased hepatic production of cholesterol-rich VLDL, as demonstrated by the increases in plasma cholesterol levels after intravenous injection of Triton WR1339. mRNA expression of LDLR was decreased, suggesting that defective LDL clearance contributed to the development of hypercholesterolemia. Curiously, the liver was enlarged, with a larger number of Ki-67-positive cells. These results demonstrate that transient upregulation of SS stimulates cholesterol biosynthesis as well as lipoprotein production, providing the first in vivo evidence that SS plays a regulatory role in cholesterol metabolism through modulation of HMG-CoA reductase activity and cholesterol biosynthesis.
  • Toyonori Kato, Hitoshi Shimano, Takashi Yamamoto, Tomotaka Yokoo, Yuko Endo, Mayumi Ishikawa, Takashi Matsuzaka, Yoshimi Nakagawa, Shin Kumadaki, Naoya Yahagi, Akimitsu Takahashi, Hirohito Sone, Hiroaki Suzuki, Hideo Toyoshima, Alyssa H. Hasty, Satoru Takahashi, Hiroshi Gomi, Tetsuro Izumi, Nobuhiro Yamada
    CELL METABOLISM 4(2) 143-154 2006年8月  査読有り
    Granuphilin is a crucial component of the docking machinery of insulin-containing vesicles to the plasma membrane. Here, we show that the granuphilin promoter is a target of SREBP-1c, a transcription factor that controls fatty acid synthesis, and MafA, a beta cell differentiation factor. Potassium-stimulated insulin secretion (KSIS) was suppressed in islets with adenoviral-mediated overexpression of granuphilin and enhanced in islets with knockdown of granuphilin (in which granuphilin had been knocked down). SREBP-1c and granuphilin were activated in islets from beta cell-specific SREBP-1c transgenic mice, as well as in several diabetic mouse models and normal islets treated with palmitate, accompanied by a corresponding reduction in insulin secretion. Knockdown- or knockout-mediated ablation of granuphilin or SREBP-1c restored KSIS in these islets. Collectively, our data provide evidence that activation of the SREBP-1c/granuphilin pathway is a potential mechanism for impaired insulin secretion in diabetes, contributing to beta cell lipotoxicity.
  • H Okazaki, M Igarashi, M Nishi, M Tajima, M Sekiya, S Okazaki, N Yahagi, K Ohashi, K Tsukamoto, M Amemiya-Kudo, T Matsuzaka, H Shimano, N Yamada, J Aoki, R Morikawa, Y Takanezawa, H Arai, R Nagai, T Kadowaki, J Osuga, S Ishibashi
    DIABETES 55(7) 2091-2097 2006年7月  査読有り
    Molecular mechanisms underlying lipolysis, as defined by mobilization of fatty acids from adipose tissue, are not fully understood. A database search for enzymes with alpha/beta hydrolase folds, the GXSXG motif for serine esterase and the His-Gly dipeptide motif, has provided a previously unannotated gene that is induced during 3T3-L1 adipocytic differentiation. Because of its remarkable structural resemblance to triacylglycerol hydrolase (TGH) with 70.4% identity, we have tentatively designated this enzyme as TGH-2 and the original TGH as TGH-1. TGH-2 is also similar to TGH-1 in terms of tissue distribution, subcellular localization, substrate specificity, and regulation. Both enzymes are predominantly expressed in liver, adipose tissue, and kidney. In adipocytes, they are localized in microsome and fatcake. Both enzymes hydrolyzed p-nitophenyl butyrate, triolein, and monoolein but not diolein, cholesteryl oleate, or phospholipids; hydrolysis of short-chain fatty acid ester was 30,000-fold more efficient than that of long-chain fatty acid triacylglycerol. Fasting increased the expression of both genes in white adipose tissue, whereas refeeding suppressed their expression. RNA silencing of TGH-2 reduced isoproterenol-stimulated glycerol release by 10% in 3T3-L1 adipocytes, while its overexpression increased the glycerol release by 20%. Thus, TGH-2 may make a contribution to adipocyte lipoly-sis during period of increased energy demand.

MISC

 133

主要な書籍等出版物

 66

主要な講演・口頭発表等

 37

共同研究・競争的資金等の研究課題

 17