Volume 40 Issue 5
May. 2023
Turn off MathJax
Article Contents
TANG Jing-hui, QIN Man-man, TANG Le, WEI Hua, QIU Liang, YU Jun. Lactobacillus plantarum ZDY04 reduces Trimethylamine N-Oxide-induced atherosclerosis by promoting reverse cholesterol transport in ApoE-/- mice[J]. JOURNAL OF MECHANICAL ENGINEERING, 2023, 1(5): 24-34. doi: 10.13381/j.cnki.cjm.00000-00
Citation: TANG Jing-hui, QIN Man-man, TANG Le, WEI Hua, QIU Liang, YU Jun. Lactobacillus plantarum ZDY04 reduces Trimethylamine N-Oxide-induced atherosclerosis by promoting reverse cholesterol transport in ApoE-/- mice[J]. JOURNAL OF MECHANICAL ENGINEERING, 2023, 1(5): 24-34. doi: 10.13381/j.cnki.cjm.00000-00

Lactobacillus plantarum ZDY04 reduces Trimethylamine N-Oxide-induced atherosclerosis by promoting reverse cholesterol transport in ApoE-/- mice

doi: 10.13381/j.cnki.cjm.00000-00
More Information
  • Corresponding author: QIU Liang, E-mail: liangqiu@jxutcm.edu.cn
  • Received Date: 22 Dec 2020
  • Rev Recd Date: 24 Mar 2021
  • Available Online: 19 Sep 2022
  • Issue Publish Date: 31 May 2023
  • Objective  To observe the effect of Lactobacillus plantarum ZDY04 in Trimethylamine N-Oxide (TMAO)-induced atherosclerosis in mice and its underlying mechanisms.
    Methods  Thirty female (six to eight-weeks old) apolipoprotein E-deficient (ApoE-/-) mice were randomly assigned into three groups as follows: Chow group, Choline+PBS group and Choline+ZDY04 group, and fed with 1.3% high-choline diet to establish atherosclerosis models, then treated with sterile PBS or ZDY04 containing 15% glycerol for 16 weeks. After treatment, serum lipids were measured using cholesterol kit. The serum TMAO and fecal TMA content were determined by using liquid chromatography-tandem mass spectrometry LC-MS. Atherosclerotic lesion formation in whole aorta was detected with Oil red O staining. Hematoxylin-eosin, Oil red O and Masson staining were used to detect the atherosclerotic area, lipid deposition and collagen content in the aortic roots. Immunofluorescent staining of macrophage specific antigen CD68 and smooth muscle a-actin (SMA) were used to examine the lesion composition in the aortic root. The expression of genes related to reverse cholesterol transport and Fmo3 in liver was detected with RT-qPCR and Western blot.
    Results  Compared with Choline+PBS group, the atherosclerotic lesions in the whole aortas significantly reduced in Choline+ZDY04 group. ZDY04 treatment significantly decreased atherosclerotic lesion size, lipid deposition and macrophage content, but not the contents of lesion smooth muscle cells or collagen, when compared to Choline+PBS fed mice. The content of cecum TMA and serum TMAO decreased significantly in Choline+ZDY04 group; serum high density lipoprotein-cholesterol significantly increased, but total cholesterol and low density lipoprotein-cholesterol only marginally decreased. ZDY04 significantly increased the mRNA expression of reverse cholesterol transport genes Sr-b1, Abcg5, Cyp7a1 but not Fmo3. Liver CYP7A1 protein level was also increased by ZDY04 compared to controls.
    Conclusion  Lactobacillus plantarum ZDY04 can inhibit TMAO induced thermogenesis by promoting reverse transport and decomposition of cholesterol in ApoE-/- mice.

     

  • loading
  • [1]
    Dagenais GR, Leong DP, Rangarajan S, et al. Variations in common diseases, hospital admissions, and deaths in middle-aged adults in 21 countries from five continents(PURE): a prospective cohort study[J]. Lancet, 2020, 395(10226): 785-794. doi: 10.1016/S0140-6736(19)32007-0
    [2]
    Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk[J]. N Engl J Med, 2013, 368(17): 1575-1584. doi: 10.1056/NEJMoa1109400
    [3]
    Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease[J]. Nature, 2011, 472(7341): 57-63. doi: 10.1038/nature09922
    [4]
    Bennett BJ, de Aguiar Vallim TQ, Wang Z, et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation[J]. Cell Metab, 2013, 17(1): 49-60. doi: 10.1016/j.cmet.2012.12.011
    [5]
    Rath S, Heidrich B, Pieper DH, et al. Uncovering the trimethylamine-producing bacteria of the human gut microbiota[J]. Microbiome, 2017, 5(1): 54. doi: 10.1186/s40168-017-0271-9
    [6]
    Seldin MM, Meng Y, Qi H, et al. Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-kappaB[J]. J Am Heart Assoc, 2016, 5(2): e002767. doi: 10.1161/JAHA.115.002767
    [7]
    Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis[J]. Nat Med, 2013, 19(5): 576-585. doi: 10.1038/nm.3145
    [8]
    Macpherson ME, Hov JR, Ueland T, et al. Gut microbiota-dependent trimethylamine N-oxide associates with inflammation in common variable immunodeficiency[J]. Front Immunol, 2020, 11: 574500. doi: 10.3389/fimmu.2020.574500
    [9]
    Zhang H, Meng J, Yu H. Trimethylamine N-oxide supplementation abolishes the cardioprotective effects of voluntary exercise in mice fed a western diet[J]. Front Physiol, 2017, 8: 944. doi: 10.3389/fphys.2017.00944
    [10]
    Hill C, Guarner F, Reid G, et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic[J]. Nat Rev Gastroenterol Hepatol, 2014, 11(8): 506-514. doi: 10.1038/nrgastro.2014.66
    [11]
    Din AU, Hassan A, Zhu Y, et al. Amelioration of TMAO through probiotics and its potential role in atherosclerosis[J]. Appl Microbiol Biotechnol, 2019, 103(23/24): 9217-9228.
    [12]
    Wu TR, Lin CS, Chang CJ, et al. Gut commensal Parabacteroides goldsteinii plays a predominant role in the anti-obesity effects of polysaccharides isolated from Hirsutella sinensis[J]. Gut, 2019, 68(2): 248-262. doi: 10.1136/gutjnl-2017-315458
    [13]
    Zhu Y, Li T, Din AU, et al. Beneficial effects of Enterococcus faecalis in hypercholesterolemic mice on cholesterol transportation and gut microbiota[J]. Appl Microbiol Biotechnol, 2019, 103(7): 3181-3191. doi: 10.1007/s00253-019-09681-7
    [14]
    Qiu L, Tao X, Xiong H, et al. Lactobacillus plantarum ZDY04 exhibits a strain-specific property of lowering TMAO via the modulation of gut microbiota in mice[J]. Food Funct, 2018, 9(8): 4299-4309. doi: 10.1039/C8FO00349A
    [15]
    Qiu L, Yang D, Tao X, et al. Enterobacter aerogenes ZDY01 attenuates choline-induced trimethylamine N-oxide levels by remodeling gut microbiota in mice[J]. J Microbiol Biotechnol, 2017, 27(8): 1491-1499. doi: 10.4014/jmb.1703.03039
    [16]
    Kuka J, Liepinsh E, Makrecka-Kuka M, et al. Suppression of intestinal microbiota-dependent production of pro-atherogenic trimethylamine N-oxide by shifting L-carnitine microbial degradation[J]. Life Sci, 2014, 117(2): 84-92. doi: 10.1016/j.lfs.2014.09.028
    [17]
    Wu WK, Panyod S, Ho CT, et al. Dietary allicin reduces transformation of L-carnitine to TMAO through impact on gut microbiota[J]. J Funct Foods, 2015, 15: 408-417. doi: 10.1016/j.jff.2015.04.001
    [18]
    Chen ML, Yi L, Zhang Y, et al. Resveratrol attenuates trimethylamine-N-oxide(TMAO)-induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota[J]. mBio, 2016, 7(2): e02210-02215.
    [19]
    Wang Z, Roberts AB, Buffa JA, et al. Non-lethal Inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis[J]. Cell, 2015, 163(7): 1585-1595. doi: 10.1016/j.cell.2015.11.055
    [20]
    Tripolt NJ, Leber B, Triebl A, et al. Effect of Lactobacillus casei shirota supplementation on trimethylamine-N-oxide levels in patients with metabolic syndrome: an open-label, randomized study[J]. Atherosclerosis, 2015, 242(1): 141-144. doi: 10.1016/j.atherosclerosis.2015.05.005
    [21]
    Canyelles M, Tondo M, Cedo L, et al. Trimethylamine N-oxide: a link among diet, gut microbiota, gene regulation of liver and intestine cholesterol homeostasis and HDL function[J]. Int J Mol Sci, 2018, 19(10): 3228. doi: 10.3390/ijms19103228
    [22]
    Zhu W, Gregory JC, Org E, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk[J]. Cell, 2016, 165(1): 111-124. doi: 10.1016/j.cell.2016.02.011
    [23]
    Linton MF, Tao H, Linton EF, et al. SR-BI: a multifunctional receptor in cholesterol homeostasis and atherosclerosis[J]. Trends Endocrinol Metab, 2017, 28(6): 461-472. doi: 10.1016/j.tem.2017.02.001
    [24]
    Yu L, Li-Hawkins J, Hammer RE, et al. Overexpression of ABCG5 and ABCG8 promotes biliary cholesterol secretion and reduces fractional absorption of dietary cholesterol[J]. J Clin Investig, 2002, 110(5): 671-680. doi: 10.1172/JCI0216001
    [25]
    Wahlstrom A, Sayin SI, Marschall HU, et al. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism[J]. Cell Metab, 2016, 24(1): 41-50. doi: 10.1016/j.cmet.2016.05.005
  • 加载中

Catalog

    Figures(6)  / Tables(1)

    Article Metrics

    Article views(133) PDF downloads(0) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return