中文English
ISSN 1001-5256 (Print)
ISSN 2097-3497 (Online)
CN 22-1108/R

留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

猪去氧胆酸对脂肪变性肝细胞活性的影响及其机制

汪远远 邹艳 刘朝霞 阳学风

引用本文:
Citation:

猪去氧胆酸对脂肪变性肝细胞活性的影响及其机制

DOI: 10.12449/JCH240212
基金项目: 

湖南省教育厅一般项目 (20C1586);

湖南省科技创新计划项目 (2020SK51910);

湖南省科技创新计划项目 (2021SK51902)

利益冲突声明:本文不存在任何利益冲突。
作者贡献声明:汪远远负责实验设计和实施,撰写论文;邹艳负责数据收集与分析;刘朝霞负责图表制作;阳学风指导撰写文章并最后定稿。
详细信息
    通信作者:

    阳学风, yxf009988@sina.com (ORCID: 0000-0002-3470-0350)

Effect of hyodeoxycholic acid on the activity of steatosis hepatocytes and its mechanism

Research funding: 

General Project of Hunan Provincial Education Department (20C1586);

Hunan Provincial Science and Technology Innovation Program Project (2020SK51910);

Hunan Provincial Science and Technology Innovation Program Project (2021SK51902)

More Information
    Corresponding author: YANG Xuefeng, yxf009988@sina.com (ORCID: 0000-0002-3470-0350)
  • 摘要:   目的  探讨猪去氧胆酸(HDCA)在代谢相关脂肪性肝病(MAFLD)发展中的作用及机制,为进一步阐明MAFLD的发病机制提供新的理论依据。  方法  L02肝细胞作为实验细胞,利用棕榈酸诱导L02细胞发生脂肪变性。采用FXR siRNA干扰链技术,构建FXR低表达的肝细胞株。CCK8实验检测HDCA在不同浓度(0、100、200、300、400 μmol/L)和时间(12、24、36、48 h)对L02脂肪变性肝细胞的影响。通过qRT-PCR检测法尼醇X受体(FXR)、增殖细胞核抗原(PCNA)、周期蛋白D1(Cyclin D1)、磷脂酰肌醇-3-激酶(PI3K)和蛋白激酶B(AKT)mRNA表达;Western Blot检测FXR、Cyclin D1、PCNA、PI3K、p-PI3K、AKT和p-AKT蛋白表达。计量资料服从正态分布且方差齐时多组间比较采用单因素方差分析,进一步两两比较采用Tukey HSD检验;服从正态分布但方差不齐时采用Welch方差分析,进一步两两比较采用Games-Howell检验。两组间比较采用成组t检验。  结果  CCK8检测结果显示,300 μmol/L HDCA处理的L02细胞和脂肪变性肝细胞活性明显下降(P值均<0.05);qRT-PCR检测结果显示,FXR mRNA表达增强,PCNA、Cyclin D1、PI3K、AKT的mRNA表达下降,差异均有统计学意义(P值均<0.05)。Western Blot检测结果显示,FXR蛋白表达明显上升(P<0.05);干扰L02细胞FXR的表达后,PCNA、PI3K、p-PI3K、AKT和p-AKT的蛋白表达均明显增加(P值均<0.05)。  结论  HDCA通过上调FXR表达抑制PI3K/AKT信号通路,从而造成脂肪变性肝细胞活性下降。

     

  • 图  1  油红O染色结果(×20)

    注: a,PA诱导前油红O染色;b,PA诱导后24 h油红O染色。

    Figure  1.  Results of the oil red O staining

    图  2  PA诱导L02细胞后甘油三酯水平随时间的变化

    Figure  2.  Changes in triglyceride levels over time after PA induction in L02 cells

    图  3  HDCA对正常肝细胞及脂肪变性肝细胞活性的影响

    注: a,不同浓度HDCA对L02细胞活性的影响;b,不同浓度HDCA对PA诱导的脂肪变性肝细胞活性的影响;c,HDCA(300 μmol/L)不同时间对PA诱导的脂肪变性肝细胞活性的影响。

    Figure  3.  Effect of HDCA on the activity of normal hepatocytes and steatosis hepatocytes

    图  4  HDCA对脂肪变性肝细胞FXR表达的影响

    注: a,FXR蛋白印迹图;b,FXR蛋白相对表达量。

    Figure  4.  Effect of HDCA on the expression of FXR in steatotic hepatocytes

    图  5  FXR siRNA干扰效果及HDCA对脂肪变性肝细胞FXR表达的影响

    注: a,3条FXR siRNA干扰链对脂肪变性肝细胞中FXR mRNA表达的干扰效果;b,FXR siRNA 3对HDCA刺激的干扰效果。

    Figure  5.  Effect of FXR siRNA interference and the effect of HDCA on FXR expression in lipid-denatured hepatocytes

    图  6  抑制FXR表达后脂肪变性肝细胞FXR-PI3K/AKT通路关键分子及PCNA和Cyclin D1蛋白表达的变化

    注: a,FXR、PCNA、Cyclin D1、PI3K、p-PI3K、AKT和p-AKT的蛋白印迹图;b,FXR蛋白的相对表达水平;c,PCNA蛋白的相对表达水平;d,Cyclin D1蛋白的相对表达水平;e,PI3K蛋白的相对表达水平;f,p-PI3K蛋白的相对表达水平;g,AKT蛋白的相对表达水平;h,p-AKT蛋白的相对表达水平。

    Figure  6.  Changes in the expression of key molecules of FXR-PI3K/AKT pathway and PCNA and Cyclin D1 proteins in steatosis hepatocytes after inhibition of FXR expression

    表  1  qRT-PCR引物序列

    Table  1.   qRT-PCR primer sequences

    引物 序列(5'-3')
    FXR 上游:AACCATACTCGCAATACAGCAA
    下游:ACAGCTCATCCCCTTTGATCC
    PCNA 上游:CCTGCTGGGATATTAGCTCCA
    下游:CAGCGGTAGGTGTCGAAGC
    Cyclin D1 上游:GCTGCGAAGTGGAAACCATC
    下游:CCTCCTTCTGCACACATTTGAA
    PI3K 上游:TATTTGGACTTTGCGACAAGACT
    下游:TCGAACGTACTGGTCTGGATAG
    AKT-F 上游:AGCGACGTGGCTATTGTGAAG
    下游:GCCATCATTCTTGAGGAGGAAGT
    下载: 导出CSV

    表  2  HDCA对脂肪变性肝细胞FXR、PI3K、AKT、PCNA和Cyclin D1 mRNA表达的影响

    Table  2.   Effect of HDCA on the mRNA expression of FXR, PI3K, AKT, PCNA, and Cyclin D1 in steatotic hepatocytes

    指标 M组 M+HDCA组 t P
    FXR mRNA 0.485±0.162 1.010±0.013 5.576 0.005
    PCNA mRNA 1.710±0.052 1.034±0.054 -15.679 <0.001
    Cyclin D1 mRNA 1.435±0.124 1.041±0.053 -5.083 0.007
    PI3K mRNA 1.951±0.500 1.008±0.009 -3.266 0.031
    AKT mRNA 2.373±0.316 1.030±0.050 -7.266 0.002
    下载: 导出CSV
  • [1] ESLAM M, SANYAL AJ, GEORGE J. MAFLD: A consensus-driven proposed nomenclature for metabolic associated fatty liver disease[J]. Gastroenterology, 2020, 158( 7): 1999- 2014. DOI: 10.1053/j.gastro.2019.11.312.
    [2] COBBINA E, AKHLAGHI F. Non-alcoholic fatty liver disease(NAFLD)-pathogenesis, classification, and effect on drug metabolizing enzymes and transporters[J]. Drug Metab Rev, 2017, 49( 2): 197- 211. DOI: 10.1080/03602532.2017.1293683.
    [3] DEPRINCE A, HAAS JT, STAELS B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease[J]. Mol Metab, 2020, 42: 101092. DOI: 10.1016/j.molmet.2020.101092.
    [4] ALVES-BEZERRA M, COHEN DE. Triglyceride metabolism in the liver[J]. Compr Physiol, 2017, 8( 1): 1- 8. DOI: 10.1002/cphy.c170012.
    [5] CHÁVEZ-TALAVERA O, TAILLEUX A, LEFEBVRE P, et al. Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease[J]. Gastroenterology, 2017, 152( 7): 1679- 1694. DOI: 10.1053/j.gastro.2017.01.055.
    [6] WANG XW, SEED B. A PCR primer bank for quantitative gene expression analysis[J]. Nucleic Acids Res, 2003, 31( 24): e154. DOI: 10.1093/nar/gng154.
    [7] O'LEARY NA, WRIGHT MW, BRISTER JR, et al. Reference sequence(RefSeq) database at NCBI: Current status, taxonomic expansion, and functional annotation[J]. Nucleic Acids Res, 2016, 44( D1): D733- D745. DOI: 10.1093/nar/gkv1189.
    [8] YOUNOSSI ZM, RINELLA ME, SANYAL AJ, et al. From NAFLD to MAFLD: Implications of a premature change in terminology[J]. Hepatology, 2021, 73( 3): 1194- 1198. DOI: 10.1002/hep.31420.
    [9] Italian Association for the Study of the Liver(AISF). AISF position paper on nonalcoholic fatty liver disease(NAFLD): Updates and future directions[J]. Dig Liver Dis, 2017, 49( 5): 471- 483. DOI: 10.1016/j.dld.2017.01.147.
    [10] JIANG TT, SUN FF, ZENG Z, et al. Progress on metabolic associated fatty liver disease related liver cancer[J/CD]. Chin J Liver Dis(Electronic Version), 2022, 14( 3): 14- 17. DOI: 10.3969/j.issn.1674-7380.2022.03.004.

    蒋婷婷, 孙芳芳, 曾湛, 等. 代谢相关脂肪性肝病相关肝癌研究进展[J/CD]. 中国肝脏病杂志(电子版), 2022, 14( 3): 14- 17. DOI: 10.3969/j.issn.1674-7380.2022.03.004.
    [11] RIZZOLO D, BUCKLEY K, KONG B, et al. Bile acid homeostasis in a cholesterol 7α-hydroxylase and sterol 27-hydroxylase double knockout mouse model[J]. Hepatology, 2019, 70( 1): 389- 402. DOI: 10.1002/hep.30612.
    [12] WATANABE S, FUJITA K. Dietary hyodeoxycholic acid exerts hypolipidemic effects by reducing farnesoid X receptor antagonist bile acids in mouse enterohepatic tissues[J]. Lipids, 2014, 49( 10): 963- 973. DOI: 10.1007/s11745-014-3947-y.
    [13] SONG M, MA XY, ZHANG FL, et al. Effects of hyodeoxycholic acid on growth performance, energy metabolism and fat digestion and absorption of mice[J]. Chin J Anim Nutr, 2022, 34( 6): 3983- 3990. DOI: 10.3969/j.issn.1006-267x.2022.06.057.

    宋敏, 马现永, 张枫琳, 等. 猪去氧胆酸对小鼠生长性能、能量代谢及脂肪消化吸收的影响[J]. 动物营养学报, 2022, 34( 6): 3983- 3990. DOI: 10.3969/j.issn.1006-267x.2022.06.057.
    [14] SEHAYEK E, ONO JG, DUNCAN EM, et al. Hyodeoxycholic acid efficiently suppresses atherosclerosis formation and plasma cholesterol levels in mice[J]. J Lipid Res, 2001, 42( 8): 1250- 1256.
    [15] SHIH DM, SHAPOSHNIK Z, MENG YH, et al. Hyodeoxycholic acid improves HDL function and inhibits atherosclerotic lesion formation in LDLR-knockout mice[J]. FASEB J, 2013, 27( 9): 3805- 3817. DOI: 10.1096/fj.12-223008.
    [16] FORMAN BM, GOODE E, CHEN J, et al. Identification of a nuclear receptor that is activated by farnesol metabolites[J]. Cell, 1995, 81( 5): 687- 693. DOI: 10.1016/0092-8674(95)90530-8.
    [17] PELLICCIARI R, COSTANTINO G, FIORUCCI S. Farnesoid X receptor: From structure to potential clinical applications[J]. J Med Chem, 2005, 48( 17): 5383- 5403. DOI: 10.1021/jm0582221.
    [18] PARKS DJ, BLANCHARD SG, BLEDSOE RK, et al. Bile acids: Natural ligands for an orphan nuclear receptor[J]. Science, 1999, 284( 5418): 1365- 1368. DOI: 10.1126/science.284.5418.1365.
    [19] DOWNES M, VERDECIA MA, ROECKER AJ, et al. A chemical, genetic, and structural analysis of the nuclear bile acid receptor FXR[J]. Mol Cell, 2003, 11( 4): 1079- 1092. DOI: 10.1016/s1097-2765(03)00104-7.
    [20] PELLICCIARI R, FIORUCCI S, CAMAIONI E, et al. 6alpha-ethyl-chenodeoxycholic acid(6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity[J]. J Med Chem, 2002, 45( 17): 3569- 3572. DOI: 10.1021/jm025529g.
    [21] SAYIN S, WAHLSTRÖM A, FELIN J, et al. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist[J]. Cell Metab, 2013, 17( 2): 225- 235. DOI: 10.1016/j.cmet.2013.01.003.
    [22] SUN LL, XIE C, WANG G, et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin[J]. Nat Med, 2018, 24( 12): 1919- 1929. DOI: 10.1038/s41591-018-0222-4.
    [23] CHIANG JYL, FERRELL JM. Bile acids as metabolic regulators and nutrient sensors[J]. Annu Rev Nutr, 2019, 39: 175- 200. DOI: 10.1146/annurev-nutr-082018-124344.
    [24] MUELLER M, THORELL A, CLAUDEL T, et al. Ursodeoxycholic acid exerts farnesoid X receptor-antagonistic effects on bile acid and lipid metabolism in morbid obesity[J]. J Hepatol, 2015, 62( 6): 1398- 1404. DOI: 10.1016/j.jhep.2014.12.034.
    [25] HARRISON SA, BASHIR MR, LEE KJ, et al. A structurally optimized FXR agonist, MET409, reduced liver fat content over 12 weeks in patients with non-alcoholic steatohepatitis[J]. J Hepatol, 2021, 75( 1): 25- 33. DOI: 10.1016/j.jhep.2021.01.047.
    [26] HAN CY, RHO HS, KIM A, et al. FXR inhibits endoplasmic reticulum stress-induced NLRP3 inflammasome in hepatocytes and ameliorates liver injury[J]. Cell Rep, 2018, 24( 11): 2985- 2999. DOI: 10.1016/j.celrep.2018.07.068.
    [27] JUNG K, KIM M, SO J, et al. Farnesoid X receptor activation impairs liver progenitor cell-mediated liver regeneration via the PTEN-PI3K-AKT-mTOR axis in zebrafish[J]. Hepatology, 2021, 74( 1): 397- 410. DOI: 10.1002/hep.31679.
    [28] FRIEDMAN ES, LI Y, SHEN TC D, et al. FXR-dependent modulation of the human small intestinal microbiome by the bile acid derivative obeticholic acid[J]. Gastroenterology, 2018, 155( 6): 1741- 1752. DOI: 10.1053/j.gastro.2018.08.022.
    [29] MAKRI E, CHOLONGITAS E, TZIOMALOS K. Emerging role of obeticholic acid in the management of nonalcoholic fatty liver disease[J]. World J Gastroenterol, 2016, 22( 41): 9039- 9043. DOI: 10.3748/wjg.v22.i41.9039.
    [30] XU J, YAO X, LI X, et al. Farnesoid X receptor regulates PI3K/AKT/mTOR signaling pathway, lipid metabolism, and immune response in hybrid grouper[J]. Fish Physiol Biochem, 2022, 48( 6): 1521- 1538. DOI: 10.1007/s10695-022-01130-z.
  • 加载中
图(6) / 表(2)
计量
  • 文章访问数:  156
  • HTML全文浏览量:  88
  • PDF下载量:  33
  • 被引次数: 0
出版历程
  • 收稿日期:  2023-05-08
  • 录用日期:  2023-06-09
  • 出版日期:  2024-02-19
  • 分享
  • 用微信扫码二维码

    分享至好友和朋友圈

目录

    /

    返回文章
    返回