Endocrinol Metab.  2021 Dec;36(6):1243-1253. 10.3803/EnM.2021.1100.

The Effects of PPAR Agonists on Atherosclerosis and Nonalcoholic Fatty Liver Disease in ApoE−/−FXR−/− Mice

Affiliations
  • 1Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
  • 2Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
  • 3Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
  • 4Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea

Abstract

Background
Farnesoid X receptor (FXR), a bile acid–activated nuclear receptor, is a potent regulator of glucose and lipid metabolism as well as of bile acid metabolism. Previous studies have demonstrated that FXR deficiency is associated with metabolic derangements, including atherosclerosis and nonalcoholic fatty liver disease (NAFLD), but its mechanism remains unclear. In this study, we investigated the role of FXR in atherosclerosis and NAFLD and the effect of peroxisome proliferator-activated receptor (PPAR) agonists in mouse models with FXR deficiency.
Methods
En face lipid accumulation analysis, liver histology, serum levels of glucose and lipids, and mRNA expression of genes related to lipid metabolism were compared between apolipoprotein E (ApoE)−/− and ApoE−/−FXR−/− mice. The effects of PPARα and PPARγ agonists were also compared in both groups of mice.
Results
Compared with ApoE−/− mice, ApoE−/−FXR−/− mice showed more severe atherosclerosis, hepatic steatosis, and higher levels of serum cholesterol, low-density lipoprotein cholesterol, and triglycerides, accompanied by increased mRNA expression of FAS, ApoC2, TNFα, IL-6 (liver), ATGL, TGH, HSL, and MGL (adipocytes), and decreased mRNA expressions of CPT2 (liver) and Tfam (skeletal muscle). Treatment with a PPARα agonist, but not with a PPARγ agonist, partly reversed atherosclerosis and hepatic steatosis, and decreased plasma triglyceride levels in the ApoE−/−FXR−/− mice, in association with increased mRNA expression of CD36 and FATP and decreased expression of ApoC2 and ApoC3 (liver).
Conclusion
Loss of FXR is associated with aggravation of atherosclerosis and hepatic steatosis in ApoE-deficient mice, which could be reversed by a PPARα agonist through induction of fatty acid uptake, β-oxidation, and triglyceride hydrolysis.

Keyword

FXR; Peroxisome proliferator-activated receptors; Apolipoproteins E; Atherosclerosis; Non-alcoholic fatty liver disease

Figure

  • Fig. 1. Atherosclerotic lesions of the aorta stained with Oil Red O in mice fed a Western diet (WD) with or without pioglitazone/fenofibrate treatment. (A) Representative photographs of aorta prepared using the en face method (a, apolipoprotein E [ApoE]−/− WD; b, ApoE−/− farnesoid X receptor [FXR]−/− WD; c, ApoE−/−FXR−/− WD+pioglitazone; d, ApoE−/−FXR−/− WD+fenofibrate). (B) Comparison of percentage of atherosclerotic lesions in the aorta in ApoE−/− (n=7) and ApoE−/−FXR−/− (n=9) mice fed a WD. The horizontal bars represent the mean percentage of atherosclerotic lesions. (C) Comparison of percentage of atherosclerotic lesions in the aorta in ApoE−/−FXR−/− mice fed a WD with or without pioglitazone (n=8) or fenofibrate (n=9) treatment. The horizontal bars represent the mean percentage of atherosclerotic lesions. Error bars show standard deviations. aA significant difference between ApoE−/− and ApoE−/−FXR−/− mice; bA significant difference between ApoE−/−FXR−/− mice with no treatment and ApoE−/−FXR−/− mice treated with fenofibrate.

  • Fig. 2. Comparison of liver histology and nonalcoholic fatty liver disease (NAFLD) activity scores (NAS) in apolipoprotein E (ApoE)−/− and ApoE−/− farnesoid X receptor (FXR)−/− mice fed a Western diet (WD) with or without pioglitazone or fenofibrate treatment. (A) Microscopic findings of the liver (H&E stain, ×200) (a, ApoE−/− [n=7]; b, ApoE−/−FXR−/− [n=9]; c, ApoE−/−FXR−/−+pioglitazone [n=8]; d, ApoE−/−FXR−/−+fenofibrate [n=9]). (B) Comparison of NAS between ApoE−/− (n=7) and ApoE−/−FXR−/− (n=9) mice. (C) Comparison of NAS among treatment groups in ApoE−/−FXR−/− mice (n=8 for control, n=8 for pioglitazone treatment, and n=9 for fenofibrate treatment). Error bars show standard deviations. aA significant difference between ApoE−/− and ApoE−/−FXR−/− mice.

  • Fig. 3. Comparison of gene expression related to lipid metabolism analyzed by quantitative polymerase chain reaction in liver, related to fatty acid synthesis (A), fatty acid uptake and catabolism (B), triglyceride hydrolysis (C), cholesterol metabolism (D), inflammation and hepatic fibrosis in the liver (E), lipolysis in adipose tissue (F), and mitochondrial activation in skeletal muscle (G) between apolipoprotein E (ApoE)−/− and ApoE−/− farnesoid X receptor (FXR)−/− mice. Error bars show standard deviations. WD, Western diet; SREBP1c, sterol regulatory element-binding protein 1c; FAS, fatty acid synthase; ACC1, acetyl-CoA carboxylase 1; SCD, stearoyl CoA desaturase; ACLY, ATP citrate lyase; FAT, fatty acid translocase; FATP1, fatty acid transport protein 1; CPT, carnitine palmitoyltranferase; ACS, acyl-CoA synthase; ACO, acyl-CoA oxidase; ACAA1A, acetyl-coenzyme A acyltransferease 1A; ApoC, apolipoprotein C; ANGPTL3, angiopoietin-like 3; PPAR, peroxisome proliferator-activated receptor; ApoA, apolipoprotein A; PLTP, phospholipid transfer protein; ABCG1, ATP-binding cassette sub-family G member 1; ABCA1, ATP-binding cassette transporter sub-family A member 1; SRB1, scavenger receptor class B type 1; CLA1, CD36 and LIMPII analogous-1; LXR, liver X receptor; MTP, microsomal triglyceride transfer protein; ApoB100, apolipoprotein B100; LDLR, low density lipoprotein receptor; TNFα, tumor necrosis factor-α; IL-6, interleukin-6; TGFβ, transforming growth factor β1; Col1α1, α1-collagen; TIMP, tissue inhibitor of metalloproteinase; αSMA, α smooth muscle actin; ATGL, adipocyte triglyceride lipase; TGH, triglycerol hydrolase; HSL, hormone sensitive lipase; MGL, monoglyceride lipase; Nrf-1, nuclear respiratory factor 1; PGC1, peroxisome proliferator-activated receptor gamma coactivator 1. aP<0.05.

  • Fig. 4. Comparison of gene expression related to lipid metabolism analyzed by quantitative polymerase chain reaction in the liver, related to fatty acid synthesis (A), fatty acid uptake and catabolism (B), triglyceride hydrolysis (C), cholesterol metabolism (D), inflammation and hepatic fibrosis in the liver (E), lipolysis in adipose tissue (F), and mitochondrial activation in skeletal muscle (G) among apolipoprotein E (ApoE)−/− farnesoid X receptor (FXR)−/− mice with or without fenofibrate or pioglitazone treatment. Error bars show standard deviations. WD, Western diet; SREBP1c, sterol regulatory element-binding protein 1c; FAS, fatty acid synthase; ACC1, acetyl-CoA carboxylase 1; SCD, stearoyl CoA desaturase; ACLY, ATP citrate lyase; FAT, fatty acid translocase; FATP1, fatty acid transport protein 1; CPT, carnitine palmitoyltranferase; ACS, acyl-CoA synthase; ACO, acyl-CoA oxidase; ACAA1A, acetyl-coenzyme A acyltransferease 1A; ApoC, apolipoprotein C; ANGPTL3, angiopoietin-like 3; PPAR, peroxisome proliferator-activated receptor; ApoA, apolipoprotein A; PLTP, phospholipid transfer protein; ABCG1, ATP-binding cassette sub-family G member 1; ABCA1, ATP-binding cassette transporter sub-family A member 1; SRB1, scavenger receptor class B type 1; CLA1, CD36 and LIMPII analogous-1; LXR, liver X receptor; MTP, microsomal triglyceride transfer protein; ApoB100, apolipoprotein B100; LDLR, low density lipoprotein receptor; SOD, superoxide dismutase; TNFα, tumor necrosis factor-α; IL-6, interleukin-6; TGFβ, transforming growth factor β1; Col1α1, α1-collagen; TIMP, tissue inhibitor of metalloproteinase; αSMA, α smooth muscle actin; ATGL, adipocyte triglyceride lipase; TGH, triglycerol hydrolase; HSL, hormone sensitive lipase; MGL, monoglyceride lipase; Nrf-1, nuclear respiratory factor 1; PGC1, peroxisome proliferator-activated receptor gamma coactivator 1. aP<0.05.


Cited by  1 articles

Lipoprotein Lipase: Is It a Magic Target for the Treatment of Hypertriglyceridemia
Joon Ho Moon, Kyuho Kim, Sung Hee Choi
Endocrinol Metab. 2022;37(4):575-586.    doi: 10.3803/EnM.2022.402.


Reference

1. Gautier T, de Haan W, Grober J, Ye D, Bahr MJ, Claudel T, et al. Farnesoid X receptor activation increases cholesteryl ester transfer protein expression in humans and transgenic mice. J Lipid Res. 2013; 54:2195–205.
Article
2. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005; 352:1685–95.
Article
3. Makishima M, Okamoto AY, Repa JJ, Tu H, Learned RM, Luk A, et al. Identification of a nuclear receptor for bile acids. Science. 1999; 284:1362–5.
Article
4. Parks DJ, Blanchard SG, Bledsoe RK, Chandra G, Consler TG, Kliewer SA, et al. Bile acids: natural ligands for an orphan nuclear receptor. Science. 1999; 284:1365–8.
Article
5. Maloney PR, Parks DJ, Haffner CD, Fivush AM, Chandra G, Plunket KD, et al. Identification of a chemical tool for the orphan nuclear receptor FXR. J Med Chem. 2000; 43:2971–4.
Article
6. Cariou B, van Harmelen K, Duran-Sandoval D, van Dijk TH, Grefhorst A, Abdelkarim M, et al. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J Biol Chem. 2006; 281:11039–49.
Article
7. Lo Sasso G, Schlage WK, Boue S, Veljkovic E, Peitsch MC, Hoeng J. The Apoe(-/-) mouse model: a suitable model to study cardiovascular and respiratory diseases in the context of cigarette smoke exposure and harm reduction. J Transl Med. 2016; 14:146.
Article
8. Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, Gonzalez FJ. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell. 2000; 102:731–44.
Article
9. Hanniman EA, Lambert G, McCarthy TC, Sinal CJ. Loss of functional farnesoid X receptor increases atherosclerotic lesions in apolipoprotein E-deficient mice. J Lipid Res. 2005; 46:2595–604.
Article
10. Hartman HB, Gardell SJ, Petucci CJ, Wang S, Krueger JA, Evans MJ. Activation of farnesoid X receptor prevents atherosclerotic lesion formation in LDLR-/- and apoE-/- mice. J Lipid Res. 2009; 50:1090–100.
Article
11. Miyazaki-Anzai S, Masuda M, Levi M, Keenan AL, Miyazaki M. Dual activation of the bile acid nuclear receptor FXR and G-protein-coupled receptor TGR5 protects mice against atherosclerosis. PLoS One. 2014; 9:e108270.
Article
12. Armstrong LE, Guo GL. Role of FXR in liver inflammation during nonalcoholic steatohepatitis. Curr Pharmacol Rep. 2017; 3:92–100.
Article
13. Sanyal AJ. Use of farnesoid X receptor agonists to treat nonalcoholic fatty liver disease. Dig Dis. 2015; 33:426–32.
Article
14. Xu P, Zhai Y, Wang J. The role of PPAR and its cross-talk with CAR and LXR in obesity and atherosclerosis. Int J Mol Sci. 2018; 19:1260.
Article
15. Kubota N, Terauchi Y, Kubota T, Kumagai H, Itoh S, Satoh H, et al. Pioglitazone ameliorates insulin resistance and diabetes by both adiponectin-dependent and -independent pathways. J Biol Chem. 2006; 281:8748–55.
Article
16. Dasu MR, Park S, Devaraj S, Jialal I. Pioglitazone inhibits Toll-like receptor expression and activity in human monocytes and db/db mice. Endocrinology. 2009; 150:3457–64.
Article
17. Ahn HY, Kim HH, Hwang JY, Park C, Cho BY, Park YJ. Effects of pioglitazone on nonalcoholic fatty liver disease in the absence of constitutive androstane receptor expression. PPAR Res. 2018; 2018:9568269.
Article
18. Zhao W, Thacker SG, Hodgin JB, Zhang H, Wang JH, Park JL, et al. The peroxisome proliferator-activated receptor gamma agonist pioglitazone improves cardiometabolic risk and renal inflammation in murine lupus. J Immunol. 2009; 183:2729–40.
Article
19. Gong Y, Shao Z, Fu Z, Edin ML, Sun Y, Liegl RG, et al. Fenofibrate inhibits cytochrome P450 epoxygenase 2C activity to suppress pathological ocular angiogenesis. EBioMedicine. 2016; 13:201–11.
Article
20. Olivier P, Plancke MO, Marzin D, Clavey V, Sauzieres J, Fruchart JC. Effects of fenofibrate, gemfibrozil and nicotinic acid on plasma lipoprotein levels in normal and hyperlipidemic mice: a proposed model for drug screening. Atherosclerosis. 1988; 70:107–14.
Article
21. Paraskevas KI, Pantopoulou A, Vlachos IS, Agrogiannis G, Iliopoulos DG, Karatzas G, et al. Comparison of fibrate, ezetimibe, low- and high-dose statin therapy for the dyslipidemia of the metabolic syndrome in a mouse model. Angiology. 2011; 62:144–54.
Article
22. Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology. 2005; 41:1313–21.
Article
23. Watanabe M, Houten SM, Wang L, Moschetta A, Mangelsdorf DJ, Heyman RA, et al. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. J Clin Invest. 2004; 113:1408–18.
Article
24. Shen LL, Liu H, Peng J, Gan L, Lu L, Zhang Q, et al. Effects of farnesoid X receptor on the expression of the fatty acid synthetase and hepatic lipase. Mol Biol Rep. 2011; 38:553–9.
Article
25. Bugianesi E, McCullough AJ, Marchesini G. Insulin resistance: a metabolic pathway to chronic liver disease. Hepatology. 2005; 42:987–1000.
Article
26. Andersson Y, Majd Z, Lefebvre AM, Martin G, Sechkin AV, Kosykh V, et al. Developmental and pharmacological regulation of apolipoprotein C-II gene expression: comparison with apo C-I and apo C-III gene regulation. Arterioscler Thromb Vasc Biol. 1999; 19:115–21.
27. Carlson LA, Ballantyne D. Changing relative proportions of apolipoproteins CII and CIII of very low density lipoproteins in hypertriglyceridaemia. Atherosclerosis. 1976; 23:563–8.
Article
28. Erkelens DW, Brunzell JD, Bierman EL. Availability of apolipoprotein CII in relation to the maximal removal capacity for an infused triglyceride emulsion in man. Metabolism. 1979; 28:495–501.
Article
29. Wang CS, McConathy WJ, Kloer HU, Alaupovic P. Modulation of lipoprotein lipase activity by apolipoproteins: effect of apolipoprotein C-III. J Clin Invest. 1985; 75:384–90.
Article
30. Bishop-Bailey D. FXR as a novel therapeutic target for vascular disease. Drug News Perspect. 2004; 17:499–504.
Article
31. Li J, Wilson A, Kuruba R, Zhang Q, Gao X, He F, et al. FXR-mediated regulation of eNOS expression in vascular endothelial cells. Cardiovasc Res. 2008; 77:169–77.
Article
32. Mencarelli A, Cipriani S, Renga B, Francisci D, Palladino G, Distrutti E, et al. The bile acid sensor FXR protects against dyslipidemia and aortic plaques development induced by the HIV protease inhibitor ritonavir in mice. PLoS One. 2010; 5:e13238.
Article
33. Miyazaki-Anzai S, Levi M, Kratzer A, Ting TC, Lewis LB, Miyazaki M. Farnesoid X receptor activation prevents the development of vascular calcification in ApoE-/- mice with chronic kidney disease. Circ Res. 2010; 106:1807–17.
34. Hambruch E, Miyazaki-Anzai S, Hahn U, Matysik S, Boettcher A, Perovic-Ottstadt S, et al. Synthetic farnesoid X receptor agonists induce high-density lipoprotein-mediated transhepatic cholesterol efflux in mice and monkeys and prevent atherosclerosis in cholesteryl ester transfer protein transgenic low-density lipoprotein receptor (-/-) mice. J Pharmacol Exp Ther. 2012; 343:556–67.
Article
35. Liu HM, Lee TY, Liao JF. GW4064 attenuates lipopolysaccharide-induced hepatic inflammation and apoptosis through inhibition of the Toll-like receptor 4-mediated p38 mitogen-activated protein kinase signaling pathway in mice. Int J Mol Med. 2018; 41:1455–62.
Article
36. Nevens F, Andreone P, Mazzella G, Strasser SI, Bowlus C, Invernizzi P, et al. A placebo-controlled trial of obeticholic acid in primary biliary cholangitis. N Engl J Med. 2016; 375:631–43.
Article
37. Claudel T, Inoue Y, Barbier O, Duran-Sandoval D, Kosykh V, Fruchart J, et al. Farnesoid X receptor agonists suppress hepatic apolipoprotein CIII expression. Gastroenterology. 2003; 125:544–55.
Article
38. Mencarelli A, Renga B, Distrutti E, Fiorucci S. Antiatherosclerotic effect of farnesoid X receptor. Am J Physiol Heart Circ Physiol. 2009; 296:H272–81.
Article
39. Zhang Y, Wang X, Vales C, Lee FY, Lee H, Lusis AJ, et al. FXR deficiency causes reduced atherosclerosis in Ldlr-/- mice. Arterioscler Thromb Vasc Biol. 2006; 26:2316–21.
40. Pineda Torra I, Claudel T, Duval C, Kosykh V, Fruchart JC, Staels B. Bile acids induce the expression of the human peroxisome proliferator-activated receptor alpha gene via activation of the farnesoid X receptor. Mol Endocrinol. 2003; 17:259–72.
41. Kliewer SA, Mangelsdorf DJ. Bile acids as hormones: the FXR-FGF15/19 pathway. Dig Dis. 2015; 33:327–31.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr