中文English
ISSN 1001-5256 (Print)
ISSN 2097-3497 (Online)
CN 22-1108/R
Issue 5
May  2018
Turn off MathJax
Article Contents

Effect of ghrelin O-acyltransferase inhibition by small interfering RNA on hepatocyte fatty degeneration and related mechanism of action

DOI: 10.3969/j.issn.1001-5256.2018.05.027
Research funding:

 

  • Received Date: 2017-11-17
  • Published Date: 2018-05-20
  • Objective To investigate the effect of inhibition of ghrelin O-acyltransferase ( GOAT) by small interfering RNA ( siRNA) on hepatocyte fatty degeneration and related mechanism of action. Methods Human LO2 hepatocytes were treated with free fatty acid ( FFA) to induce hepatocyte fatty degeneration. LO2 hepatocytes were treated with FFA and siRNA-GOAT alone or in combination and then divided into normal control ( NC) group ( treated with phosphate buffered saline alone) , siRNA-GOAT group ( treated with siRNA-GOAT at a final concentration of 10 nm) , FFA group ( treated with FFA at a final concentration of 1 mm) , and FFA + siRNA-GOAT group ( treated with FFA at a final concentration of 1 mm and siRNA-GOAT at a final concentration of 10 nm) . Oil red O staining was performed for hepatocytes to identify lipid droplets; the triglyceride ( TG) test kit was used to measure the lipid level in LO2 hepatocytes; Western blot, qRT-PCR, immunofluorescent staining, and electron microscopy were used to measure autophagy; ELISA and RT-PCR were used to measure the levels of tumor necrosis factor-α ( TNFα) and interleukin-6 ( IL-6) ; ELISA was used to measure the changes in the levels of mammalian target of rapamycin ( m TOR) , phosphorylated m TOR ( p-m TOR) , AMP-activated protein kinase ( AMPK) , and phosphorylated AMPK. A one-way analysis of variance was used for comparison between multiple groups, and the least significant difference t-test was used for further comparison between any two groups. Results Compared with the FFA group, the FFA + siRNA-GOAT group had a significant reduction in the formation of lipid droplets and a significantly lower TG level ( P < 0. 001) . Compared with the FFA group, the FFA +siRNA-GOAT group had significant reductions in the protein and mRNA expression of TNFα and IL-6 ( all P < 0. 005) . The siRNA +GOAT group had significantly higher mRNA expression of LC3-II and Beclin-1 than the NC group ( all P < 0. 001) . The FFA + siRNA-GOAT group had significantly higher mRNA expression of LC3-II and Beclin-1 than the FFA group ( all P < 0. 001) . The siRNA + GOAT group had significantly higher protein expression of LC3-II and Beclin-1 than the NC group ( all P < 0. 05) . The FFA + siRNA-GOAT group had significantly higher protein expression of LC3-II and Beclin-1 than the FFA group ( all P < 0. 05) . Immunofluorescent staining showed that compared with the FFA group and the siRNA-GOAT group, the FFA + siRNA-GOAT group had a significant increase in the expression of endogenous LC3-II in LO2 hepatocytes. Electron microscopy showed that compared with the FFA group, the FFA + siRNA-GOAT group had a significant increase in the expression of autophagosome. After the LO2 hepatocytes were treated by autophagy inhibitors siRNA-ATG5 and 3-MA or an autophagy stimulant, rapamycin, there was a significant difference in TG level between the FFA + siRNA-ATG5 group and the FFA + siRNA-GOAT group ( P < 0. 001) , as well as between the FFA + 3-MA group and the FFA + rapamycin group ( P < 0. 001) . The FFA + siRNA-GOAT group had a significantly higher level of LC3-I/II than the FFA + siRNA-ATG5 group ( P <0. 05) , and the FFA + rapamycin group had a significantly higher level of LC3-I/II than the FFA + 3-MA group ( P < 0. 05) . Compared with the FFA group, the FFA + siRNA-GOAT group had significantly higher protein expression of p-AMPK ( P < 0. 05) and significantly lower protein expression of p-m TOR ( P < 0. 05) . Conclusion GOAT inhibition by siRNA can upregulate autophagy and alleviate hepatocyte fatty degeneration, possibly by regulating the AMPK/m TOR pathway.

     

  • loading
  • [1]ADAMS LA, LYMP JF, ST SAUVER J, et al.The natural history of nonalcoholic fatty liver disease:a population-based cohort study[J].Gastroenterology, 2005, 129 (1) :113-121.
    [2]BERLANGA A, GUIU-JURADO E, PORRAS JA, et al.Molecular pathways in non-alcoholic fatty liver disease[J].Clin Exp Gastroenterol, 2014, 7:221-239.
    [3]FERRE P, FOUFELLE F.Hepatic steatosis:a role for de novo lipogenesis and the transcription factor SREBP-1c[J].Diabetes Obes Metab, 2010, 12 (Suppl 2) :83-92.
    [4]INUI A, ASAKAWA A, BOWERS CY, et al.Ghrelin, appetite, and gastric motility:the emerging role of the stomach as an endocrine organ[J].FASEB J, 2004, 18 (3) :439-456.
    [5]BARNETT BP, HWANG Y, TAYLOR MS, et al.Glucose and weight control in mice with a designed ghrelin O-acyltransferase inhibitor[J].Science, 2010, 330 (6011) :1689-1692.
    [6]TEUFFEL P, WANG L, PRINZ P, et al.Treatment with the ghrelin-O-acyltransferase (GOAT) inhibitor GO-Co A-Tat reduces food intake by reducing meal frequency in rats[J].J Physiol Pharmacol, 2015, 66 (4) :493-503.
    [7]YANG J, BROWN MS, LIANG G, et al.Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone[J].Cell, 2008, 132 (3) :387-396.
    [8]GUALILLO O, LAGO F, DIEGUEZ C.Introducing GOAT:a target for obesity and anti-diabetic drugs?[J].Trends Pharmacol Sci, 2008, 29 (8) :398-401.
    [9]AMIR M, CZAJA MJ.Autophagy in nonalcoholic steatohepatitis[J].Expert Rev Gastroenterol Hepatol, 2011, 5 (2) :159-166.
    [10]NOUREDDIN M, YATES KP, VAUGHN IA, et al.Clinical and histological determinants of nonalcoholic steatohepatitis and advanced fibrosis in elderly patients[J].Hepatology, 2013, 58 (5) :1644-1654.
    [11]KIM D, CHOI SY, PARK EH, et al.Nonalcoholic fatty liver disease is associated with coronary artery calcification[J].Hepatology, 2012, 56 (2) :605-613.
    [12]DING ZY, PU L, LU HY, et al.Correlation of liver fat content with serum vitamin A level and insulin resistace in patients with nonalcoholic fatty liver disease[J].J Clin Hepatol, 2017, 33 (12) :2361-2365. (in Chinese) 丁智勇, 卜乐, 鲁鸿燕, 等.非酒精性脂肪性肝病患者血清维生素A水平、肝脏脂肪含量与胰岛素抵抗的相关性分析[J].临床肝胆病杂志, 2017, 33 (12) :2361-2365.
    [13]NI MM, ZHANG YQ, CHEN YX, et al.Prevalence of nonalcoholic fatty liver disease and its prognostic factors[J].J Clin Hepatol, 2016, 32 (3) :453-458. (in Chinese) 倪曼曼, 张颖秋, 陈岳祥, 等.非酒精性脂肪性肝病患者发病和预后的影响因素分析[J].临床肝胆病杂志, 2016, 32 (3) :453-458.
    [14]ALKHOURI N, CARTER-KENT C, FELDSTEIN AE.Apoptosis in nonalcoholic fatty liver disease:diagnostic and therapeutic implications[J].Expert Rev Gastroenterol Hepatol, 2011, 5 (2) :201-212.
    [15]KOEK GH, LIEDORP PR, BAST A.The role of oxidative stress in non-alcoholic steatohepatitis[J].Clin Chim Acta, 2011, 412 (15-16) :1297-1305.
    [16]YUAN RS, LI H, SUN JH, et al.Lipid-lowering effect and antioxidant activity of polysaccharide from Schisandra Chinensis in rats with non-alcoholic fatty liver disease induced by high-fat diet[J].J Jilin Univ:Med Edit, 2017, 43 (6) :1103-1108. (in Chinese) 苑荣爽, 李贺, 孙靖辉, 等.北五味子多糖对高脂诱导非酒精性脂肪性肝病大鼠的降血脂作用及其抗氧化活性[J].吉林大学学报:医学版, 2017, 43 (6) :1103-1108.
    [17]JORNAYVAZ FR, SHULMAN GI.Diacylglycerol activation of protein kinase C and hepatic insulin resistance[J].Cell Metabolism, 2012, 15 (5) :574-584.
    [18]OZCELIK F, YUKSEL C, ARSLAN E, et al.Relationship between visceral adipose tissue and adiponectin, inflammatory markers and thyroid hormones in obese males with hepatosteatosis and insulin resistance[J].Arch Med Res, 2013, 44 (4) :273-280.
    [19]MUSSO G, GAMBINO R, BO S, et al.Should non-alcoholic fatty liver disease be included in the definition of metabolic syndrome?A cross-sectional comparison with adult treatment Panel III criteria in nonobese nondiabetic subjects[J].Diabetes Care, 2008, 31 (3) :562-568.
    [20]RAVIKUMAR B, BERGER Z, RUBINSZTEIN DC, et al.Rapamycin pretreatment protects against apoptosis[J].Hum Mol Genet, 2006, 15 (7) :1209-1216.
    [21]YU L, Mc PHEE CK, ZHENG L, et al.Termination of autophagy and reformation of lysosomes regulated by Mtor[J].Nature, 2010, 465 (7300) :942-946.
    [22]LIU HY, HAN J, CAO SY, et al.Hepatic autophagy is suppressed in the presence of insulin resistance and hyperinsulinemia:inhibition of Fox O1-dependent expression of key autophagy genes by insulin[J].J Biol Chem, 2009, 284 (45) :31484-31492.
    [23]SINHA RA, FARAH BL, SINGH BK, et al.Caffeine stimulates hepatic lipid metabolism by the autophagy-lysosomal pathway in mice[J].Hepatology, 2014, 59 (4) :1366-1380.
    [24]SINGH R, KAUSHIK S, WANG Y, et al.Autophagy regulates lipid metabolism[J].Nature, 2009, 458 (7242) :1131-1135.
    [25]DING WX, LI M, CHEN X, et al.Autophagy reduces acute ethanol-induced hepatotoxicity and steatosis in mice[J].Gastroenterology, 2010, 139 (5) :1740-1752.
    [26]LIU K, CZAJA MJ.Regulation of lipid stores and metabolism by lipophagy[J].Cell Death Differ, 2013, 20 (1) :3-11.
    [27]LI Y, HAI J, LI L, et al.Administration of ghrelin improves inflammation, oxidative stress, and apoptosis during and after nonalcoholic fatty liver disease development[J].Endocrine, 2013, 43 (2) :376-386.
    [28]WINDER WW, HARDIE DG.AMP-activated protein kinase, a metabolic master switch:possible roles in type 2 diabetes[J].Am J Physiol, 1999, 277 (1 Pt 1) :e1-e10.
    [29]YIN XM, DING WX, GAO W.Autophagy in the liver[J].Hepatology, 2008, 47 (5) :1773-1785.
    [30]BARAZZONI R, SEMOLIC A, CATTIN MR, et al.Acylated ghrelin limits fat accumulation and improves redox state and inflammation markers in the liver of high-fat-fed rats[J].Obesity (Silver Spring) , 2014, 22 (1) :170-177.
    [31]SHOJI-KAWATA S, SUMPTER R, LEVENO M, et al.Identification of a candidate therapeutic autophagy-inducing peptide[J].Nature, 2013, 494 (7436) :201-206.
    [32]LIN CW, ZHANG H, LI M, et al.Pharmacological promotion of autophagy alleviates steatosis and injury in alcoholic and non-alcoholic fatty liver conditions in Mice[J].J Hepatol, 2013, 58 (5) :993-999.
    [33]SINHA RA, YOU SH, ZHOU J, et al.Thyroid hormone stimulates hepatic lipid catabolism via activation of autophagy[J].J Clin Invest, 2012, 122 (7) :2428-2438.
    [34]NI HM, WILLIAMS JA, YANG H, et al.Targeting autophagy for the treatment of liver diseases[J].Pharmacol Res, 2012, 66 (6) :463-474.
    [35]CODOGNO P, MEIJER AJ.Autophagy in the liver[J].J Hepatol, 2013, 59 (2) :389-391.
    [36]DING WX.Induction of autophagy, a promising approach for treating liver injury[J].Hepatology, 2014, 59 (1) :340-343.
    [37]LOMONACO R, SUNNY NE, BRIL F, et al.Nonalcoholic fatty liver disease:current issues and novel treatment approaches[J].Drugs, 2013, 73 (1) :1-14.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Article Metrics

    Article views (2029) PDF downloads(393) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return