Nutr Res Pract.  2024 Dec;18(6):774-792. 10.4162/nrp.2024.18.6.774.

p-Coumaric acid modulates cholesterol efflux and lipid accumulation and inflammation in foam cells

Affiliations
  • 1Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea

Abstract

BACKGROUND/OBJECTIVES
Atherosclerosis is a primary cause of cardiovascular disease associated with inflammation and lipid metabolism disorders. The accumulation of cholesterol-containing macrophage foam cells characterizes the early stages. The p-coumaric acid (p-CA) contained in vegetables may have various physiological activities. The inhibitory effect of p-CA on foam cell creation in THP-1 macrophages needs clarification. In this study, we explored the impact of p-CA on foam cells by co-treatment with oxidized lowdensity lipoprotein (ox-LDL) and lipopolysaccharides (LPS), mimicking the development of atherosclerosis in vitro and studied the regulation of its underlying mechanisms.
MATERIALS/METHODS
THP-1 cells differentiated by phorbol 12-myristate 13-acetate (1 μM) for 48 h and treated in the absence or presence of p-CA for 48 h. THP-1 macrophages were treated with combined ox-LDL (20 μg/mL) and LPS (500 ng/mL) for 24 h. The 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assays detected cell viability. Oil red O staining allowed us to observe lipid accumulation. Western blotting and quantitative polymerase chain reactions quantified corresponding proteins and mRNA.
RESULTS
Ox-LDL and LPS for 24 h enhanced the lipid accumulation using Oil red O in treated foam cells. By contrast, p-CA treatment inhibited lipid accumulation. p-CA significantly upregulated cholesterol efflux-related genes such as ATP binding cassette transporter A1, liver-X-receptor α and peroxisome proliferator-activated receptor gamma expression. Moreover, p-CA decreased lipid accumulation-related gene such as lectin-like oxidized low-density lipoprotein receptor-1, cluster of differentiation 36 and scavenger receptor class A1 expression. Combined ox-LDL and LPS increased nuclear factor-κB (NFκB), cyclooxygenase-2 (COX-2) and pro-inflammatory (tumor necrosis factor-α [TNF-α] and interleukin [IL]-6) activation and expression compared with untreated. p-CA suppressed this increased expression of NF-κB and COX-2, TNF-α and IL-6.
CONCLUSION
p-CA may play a vital role in atherosclerosis inhibition and protective effects by suppressing lipid accumulation and foam cell creation by increasing cholesterol efflux and can be potential agents for preventing atherosclerosis.

Keyword

Atherosclerosis; foam cells; inflammation; oxidized low density lipoprotein; p-coumaric acid

Figure

  • Fig. 1 Effect of p-CA on cell viability of LPS-treated THP-1 cells and upregulated expression of the inflammatory factor. (A) THP-1 monocytes were exposed to 1 μM of phorbol 12-myristate 13-acetate for 48 h, pretreated with p-CA at several concentrations and then induced with or without 500 ng/mL LPS for 24 h. Cell viability was measured using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay. Experiments were performed in triplicate, and results are presented as the mean ± SD. The protein expression levels of NF-κB and SIRT1 were determined using immunoblotting (B).LPS, lipopolysaccharides; p-CA, p-coumaric acid; NF-κB, nuclear factor-κB; SIRT1, sirtuin 1.Different letters indicate significant differences (P < 0.05) as determined by Duncan’s multiple range test.

  • Fig. 2 Downregulation of lipid accumulation by p-CA treatment in THP-1 foam cells. THP-1 differentiated macrophages were cultured in the absence or presence of p-CA (0–20 μM) before 24 h. Then, the THP-1 cell was cultured in LPS (500 ng/mL) containing ox-LDL (20 μg/mL) for 24 h. (A) Cells were stained with Oil Red O; microphotographs were obtained using an optical microscope, magnification 400×. (B) Stained cells were dissolved in an isopropanol solution, and the staining intensity was measured at 520 nm. Experiments were performed in triplicate, and results are presented as the mean ± SD.LPS, lipopolysaccharides; ox-LDL, oxidized low-density lipoprotein; p-CA, p-coumaric acid.Different letters indicate significant differences (P < 0.05), as determined by Duncan’s multiple range test.

  • Fig. 3 Inhibition of SR-A1, CD36, and LOX-1 expression by p-CA treatment in THP-1 foam cells. The protein expression levels of SR-A1, CD36, and LOX-1 were determined using (A) immunoblotting. (B, C) The relative mRNA expression levels are shown after normalization against β-actin mRNA expression. (B) CD36 and (C) LOX-1 levels. The data are expressed relative to the mRNA levels found in untreated cells, which was arbitrarily defined as 1. (D) SR-A1 levels densities were normalized to β-actin using ImageJ software. Experiments were performed at least in triplicate, and the results are presented as mean ± SD. Data were analyzed by applying the 2−ΔΔCT method.LOX-1, lectin-like oxidized low-density lipoprotein receptor-1; SR-A1, scavenger receptor class A1; LPS, lipopolysaccharides; ox-LDL, oxidized low-density lipoprotein; p-CA, p-coumaric acid; CD36, cluster of differentiation 36.Different letters indicate significant differences (P < 0.05) as determined by Duncan’s multiple range test.

  • Fig. 4 Upregulation of ABCA1, LXRα, and PPARγ expression by p-CA treatment in THP-1 foam cells. The protein expression levels of ABCA1 were determined using (A) immunoblotting. Cells were harvested, and the expression of the (B) ABCA1 mRNA in ox-LDL and LPS-induced foam cells was evaluated. The protein expression levels of LXRα and PPARγ were determined using (C) immunoblotting. Cells were harvested, and the expression of the (D, E) LXRα and PPARγ mRNA in ox-LDL and LPS-induced foam cells was evaluated. (D) LXRα and (E) PPARγ levels. Data are presented as the means ± SD. Data were analyzed by applying the 2−ΔΔCT method.LPS, lipopolysaccharides; ox-LDL, oxidized low-density lipoprotein; p-CA, p-coumaric acid; ABCA1, ATP binding cassette transporter A1; LXRα, liver X receptor α; PPARγ, Peroxisome proliferator-activated receptor γ.Different letters indicate significant differences (P < 0.05) as determined by Duncan’s multiple range test.

  • Fig. 5 Inhibition of inflammatory cytokine secretion by p-CA treatment in co-treated with ox-LDL and LPS-induced foam cells. THP-1 foam cells were pretreated with different concentrations of p-CA (0–20 μM) for 48 h. (A, B) The secretion of IL-6 and TNF-α was measured using an enzyme-linked immunosorbent assay kit. The expression levels of TNF-α and COX-2 were measured using (C) immunoblotting. Cells were harvested, and the expression of the (D) COX-2 mRNA in ox-LDL and LPS-induced foam cells was evaluated. (E) TNF-α levels densities were normalized to β-actin using ImageJ software. Data are presented as the means ± SD.LPS, lipopolysaccharides; ox-LDL, oxidized low-density lipoprotein; p-CA, p-coumaric acid; TNF-α, tumor necrosis factor-α; IL-6, interleukin 6; COX-2, cyclooxygenase-2.Different letters indicate significant differences (P < 0.05) as determined by Duncan’s multiple range test.

  • Fig. 6 Inhibition of NF-κB p65 activation by p-CA treatment in co-treated with ox-LDL and LPS-induced foam cells. The levels of the NF-κB and SIRT1 proteins were measured using (A) immunoblotting. Cells were harvested, and the expression of the (B) NF-κB mRNA in ox-LDL and LPS-induced foam cells was evaluated. (C) NF-κB levels densities were normalized to β-actin using ImageJ software. Data are presented as the means ± SD. (D) THP-1 foam cells were treated with p-CA (0–20 μM) and fixed with 4% paraformaldehyde. After blocking with an appropriate buffer, cells were incubated with antibodies. Next, DAPI staining was performed to confirm the nuclei in the cells. Signals were quantified using fluorescence microscopy at 400× magnification.LPS, lipopolysaccharides; ox-LDL, oxidized low-density lipoprotein; p-CA, p-coumaric acid; NF-κB, nuclear factor-κB; SIRT1, sirtuin 1; DAPI, 4′,6-diamidino-2-phenylindole.Different letters indicate significant differences (P < 0.05) as determined by Duncan’s multiple range test.


Reference

1. Levenson JW, Skerrett PJ, Gaziano JM. Reducing the global burden of cardiovascular disease: the role of risk factors. Prev Cardiol. 2002; 5:188–199. PMID: 12417828.
Article
2. Joshi R, Jan S, Wu Y, MacMahon S. Global inequalities in access to cardiovascular health care: our greatest challenge. J Am Coll Cardiol. 2008; 52:1817–1825. PMID: 19038678.
3. Björkegren JLM, Lusis AJ. Atherosclerosis: recent developments. Cell. 2022; 185:1630–1645. PMID: 35504280.
Article
4. Paulson KE, Zhu SN, Chen M, Nurmohamed S, Jongstra-Bilen J, Cybulsky MI. Resident intimal dendritic cells accumulate lipid and contribute to the initiation of atherosclerosis. Circ Res. 2010; 106:383–390. PMID: 19893012.
Article
5. Ley K, Miller YI, Hedrick CC. Monocyte and macrophage dynamics during atherogenesis. Arterioscler Thromb Vasc Biol. 2011; 31:1506–1516. PMID: 21677293.
Article
6. Rafieian-Kopaei M, Setorki M, Doudi M, Baradaran A, Nasri H. Atherosclerosis: process, indicators, risk factors and new hopes. Int J Prev Med. 2014; 5:927–946. PMID: 25489440.
7. Su Y, Gao J, Kaur P, Wang Z. Neutrophils and macrophages as targets for development of nanotherapeutics in inflammatory diseases. Pharmaceutics. 2020; 12:1222. PMID: 33348630.
Article
8. Zhu WW, Wang SR, Liu ZH, Cao YJ, Wang F, Wang J, Liu CF, Xie Y, Xie Y, Zhang YL. Gly[14]-humanin inhibits ox-LDL uptake and stimulates cholesterol efflux in macrophage-derived foam cells. Biochem Biophys Res Commun. 2017; 482:93–99. PMID: 27815075.
Article
9. Guo C, Ma R, Liu X, Chen T, Li Y, Yu Y, Duan J, Zhou X, Li Y, Sun Z. Silica nanoparticles promote oxLDL-induced macrophage lipid accumulation and apoptosis via endoplasmic reticulum stress signaling. Sci Total Environ. 2018; 631-632:570–579. PMID: 29533793.
Article
10. Ait-Oufella H, Taleb S, Mallat Z, Tedgui A. Recent advances on the role of cytokines in atherosclerosis. Arterioscler Thromb Vasc Biol. 2011; 31:969–979. PMID: 21508343.
Article
11. Yi BG, Park OK, Jeong MS, Kwon SH, Jung JI, Lee S, Ryoo S, Kim SE, Kim JW, Moon WJ, et al. In vitro photodynamic effects of scavenger receptor targeted-photoactivatable nanoagents on activated macrophages. Int J Biol Macromol. 2017; 97:181–189. PMID: 28082222.
Article
12. Wang G, Groman E, Simberg D. Discrepancies in the in vitro and in vivo role of scavenger receptors in clearance of nanoparticles by Kupffer cells. Precis Nanomed. 2018; 1:76–84.
Article
13. Moore KJ, Freeman MW. Scavenger receptors in atherosclerosis: beyond lipid uptake. Arterioscler Thromb Vasc Biol. 2006; 26:1702–1711. PMID: 16728653.
14. Ghosh S, Zhao B, Bie J, Song J. Macrophage cholesteryl ester mobilization and atherosclerosis. Vascul Pharmacol. 2010; 52:1–10. PMID: 19878739.
Article
15. Wang M, Xiang Q, Sun W, Zhang H, Shi R, Guo J, Tong H, Fan M, Ding Y, Shi H, et al. Qihuang zhuyu formula attenuates atherosclerosis via targeting PPARγ to regulate cholesterol efflux and endothelial cell inflammation. Oxid Med Cell Longev. 2022; 2022:2226168. PMID: 36518993.
16. Wu C, Luan H, Zhang X, Wang S, Zhang X, Sun X, Guo P. Chlorogenic acid protects against atherosclerosis in ApoE-/- mice and promotes cholesterol efflux from RAW264.7 macrophages. PLoS One. 2014; 9:e95452. PMID: 25187964.
17. Badimón JJ, Ibáñez B. Increasing high-density lipoprotein as a therapeutic target in atherothrombotic disease. Rev Esp Cardiol. 2010; 63:323–333. PMID: 20196993.
Article
18. Chen W, Wang N, Tall AR. A PEST deletion mutant of ABCA1 shows impaired internalization and defective cholesterol efflux from late endosomes. J Biol Chem. 2005; 280:29277–29281. PMID: 15951431.
Article
19. Wang J, Xiao Q, Wang L, Wang Y, Wang D, Ding H. Role of ABCA1 in cardiovascular disease. J Pers Med. 2022; 12:1010. PMID: 35743794.
20. Costet P, Luo Y, Wang N, Tall AR. Sterol-dependent transactivation of the ABC1 promoter by the liver X receptor/retinoid X receptor. J Biol Chem. 2000; 275:28240–28245. PMID: 10858438.
Article
21. Repa JJ, Turley SD, Lobaccaro JA, Medina J, Li L, Lustig K, Shan B, Heyman RA, Dietschy JM, Mangelsdorf DJ. Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers. Science. 2000; 289:1524–1529. PMID: 10968783.
Article
22. Chinetti G, Lestavel S, Bocher V, Remaley AT, Neve B, Torra IP, Teissier E, Minnich A, Jaye M, Duverger N, et al. PPAR-α and PPAR-γ activators induce cholesterol removal from human macrophage foam cells through stimulation of the ABCA1 pathway. Nat Med. 2001; 7:53–58. PMID: 11135616.
Article
23. Spagnoli LG, Bonanno E, Sangiorgi G, Mauriello A. Role of inflammation in atherosclerosis. J Nucl Med. 2007; 48:1800–1815. PMID: 17942804.
Article
24. Tipping PG, Hancock WW. Production of tumor necrosis factor and interleukin-1 by macrophages from human atheromatous plaques. Am J Pathol. 1993; 142:1721–1728. PMID: 8506944.
25. Barath P, Fishbein MC, Cao J, Berenson J, Helfant RH, Forrester JS. Detection and localization of tumor necrosis factor in human atheroma. Am J Cardiol. 1990; 65:297–302. PMID: 2405620.
Article
26. Yurdagul A Jr, Sulzmaier FJ, Chen XL, Pattillo CB, Schlaepfer DD, Orr AW. Oxidized LDL induces FAK-dependent RSK signaling to drive NF-κB activation and VCAM-1 expression. J Cell Sci. 2016; 129:1580–1591. PMID: 26906414.
Article
27. de Winther MP, Kanters E, Kraal G, Hofker MH. Nuclear factor κB signaling in atherogenesis. Arterioscler Thromb Vasc Biol. 2005; 25:904–914. PMID: 15731497.
Article
28. Cheng ZJ, Vapaatalo H, Mervaala E. Angiotensin II and vascular inflammation. Med Sci Monit. 2005; 11:RA194–RA205. PMID: 15917731.
29. Benigni A, Cassis P, Remuzzi G. Angiotensin II revisited: new roles in inflammation, immunology and aging. EMBO Mol Med. 2010; 2:247–257. PMID: 20597104.
Article
30. Salazar G. NADPH oxidases and mitochondria in vascular senescence. Int J Mol Sci. 2018; 19:1327. PMID: 29710840.
Article
31. Zeng HT, Fu YC, Yu W, Lin JM, Zhou L, Liu L, Wang W. SIRT1 prevents atherosclerosis via liver-X-receptor and NF-κB signaling in a U937 cell model. Mol Med Rep. 2013; 8:23–28. PMID: 23652462.
Article
32. Li Y, Wang P, Yang X, Wang W, Zhang J, He Y, Zhang W, Jing T, Wang B, Lin R. SIRT1 inhibits inflammatory response partly through regulation of NLRP3 inflammasome in vascular endothelial cells. Mol Immunol. 2016; 77:148–156. PMID: 27505710.
Article
33. Yoshizaki T, Schenk S, Imamura T, Babendure JL, Sonoda N, Bae EJ, Oh DY, Lu M, Milne JC, Westphal C, et al. SIRT1 inhibits inflammatory pathways in macrophages and modulates insulin sensitivity. Am J Physiol Endocrinol Metab. 2010; 298:E419–E428. PMID: 19996381.
Article
34. Pfluger PT, Herranz D, Velasco-Miguel S, Serrano M, Tschöp MH. Sirt1 protects against high-fat diet-induced metabolic damage. Proc Natl Acad Sci U S A. 2008; 105:9793–9798. PMID: 18599449.
Article
35. Tabansi D, Dahiru D, Patrick AT, Jahng WJ. Anti-atherosclerosis and anti-hyperlipidemia functions of Terminalia catappa fruit. ACS Omega. 2023; 8:35571–35579. PMID: 37810701.
Article
36. Hu R, Wu S, Li B, Tan J, Yan J, Wang Y, Tang Z, Liu M, Fu C, Zhang H, et al. Dietary ferulic acid and vanillic acid on inflammation, gut barrier function and growth performance in lipopolysaccharide-challenged piglets. Anim Nutr. 2022; 8:144–152. PMID: 34977384.
Article
37. Joo HK, Choi S, Lee YR, Lee EO, Park MS, Park KB, Kim CS, Lim YP, Park JT, Jeon BH. Anthocyanin-rich extract from red chinese cabbage alleviates vascular inflammation in endothelial cells and Apo E−/− mice. Int J Mol Sci. 2018; 19:816. PMID: 29534512.
Article
38. Wang P, Ma Y, Wang D, Zhao W, Hu X, Chen F, Zhao X. Protective effects of dietary resveratrol against chronic low-grade inflammation mediated through the gut microbiota in high-fat diet mice. Nutrients. 2022; 14:1994. PMID: 35631150.
Article
39. Jiang T, Zhang W, Wang Z. Laquinimod protects against TNF-α-induced attachment of monocytes to human aortic endothelial cells (HAECs) by increasing the expression of KLF2. Drug Des Devel Ther. 2020; 14:1683–1691.
40. Li J, Li Y, Yuan X, Yao D, Gao Z, Niu Z, Wang Z, Zhang Y. The effective constituent puerarin, from Pueraria lobata, inhibits the proliferation and inflammation of vascular smooth muscle in atherosclerosis through the miR-29b-3p/IGF1 pathway. Pharm Biol. 2023; 61:1–11. PMID: 36537316.
Article
41. Socha R, Gałkowska D, Bugaj M, Juszczak L. Phenolic composition and antioxidant activity of propolis from various regions of Poland. Nat Prod Res. 2015; 29:416–422. PMID: 25185953.
Article
42. Li H, Lee HS, Kim SH, Moon B, Lee C. Antioxidant and anti-inflammatory activities of methanol extracts of Tremella fuciformis and its major phenolic acids. J Food Sci. 2014; 79:C460–C468. PMID: 24547933.
43. Kong CS, Jeong CH, Choi JS, Kim KJ, Jeong JW. Antiangiogenic effects of p-coumaric acid in human endothelial cells. Phytother Res. 2013; 27:317–323. PMID: 22585412.
44. Yoon JH, Youn K, Ho CT, Karwe MV, Jeong WS, Jun M. p-Coumaric acid and ursolic acid from Corni fructus attenuated β-amyloid25-35-induced toxicity through regulation of the NF-κB signaling pathway in PC12 cells. J Agric Food Chem. 2014; 62:4911–4916. PMID: 24815946.
Article
45. Luceri C, Giannini L, Lodovici M, Antonucci E, Abbate R, Masini E, Dolara P. p-Coumaric acid, a common dietary phenol, inhibits platelet activity in vitro and in vivo . Br J Nutr. 2007; 97:458–463. PMID: 17313706.
Article
46. Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011; 21:103–115. PMID: 21187859.
Article
47. Ha KH, Kwon HS, Kim DJ. Epidemiologic characteristics of dyslipidemia in Korea. J Lipid Atheroscler. 2015; 4:93–99.
Article
48. WHO CVD Risk Chart Working Group. World Health Organization cardiovascular disease risk charts: revised models to estimate risk in 21 global regions. Lancet Glob Health. 2019; 7:e1332–e1345. PMID: 31488387.
49. Oh K, Kim Y, Kweon S, Kim S, Yun S, Park S, Lee YK, Kim Y, Park O, Jeong EK. Korea National Health and Nutrition Examination Survey, 20th anniversary: accomplishments and future directions. Epidemiol Health. 2021; 43:e2021025. PMID: 33872484.
Article
50. Shin HY, Kim J, Lee S, Park MS, Park S, Huh S. Cause-of-death statistics in 2018 in the Republic of Korea. J Korean Med Assoc. 2020; 63:286–297.
Article
51. Yuan Y, Li P, Ye J. Lipid homeostasis and the formation of macrophage-derived foam cells in atherosclerosis. Protein Cell. 2012; 3:173–181. PMID: 22447659.
Article
52. Volobueva A, Zhang D, Grechko AV, Orekhov AN. Foam cell formation and cholesterol trafficking and metabolism disturbances in atherosclerosis. Cor Vasa. 2019; 61:48–54.
Article
53. Pennings M, Meurs I, Ye D, Out R, Hoekstra M, Van Berkel TJ, Van Eck M. Regulation of cholesterol homeostasis in macrophages and consequences for atherosclerotic lesion development. FEBS Lett. 2006; 580:5588–5596. PMID: 16935283.
Article
54. Wang H, Li Y, Zhang X, Xu Z, Zhou J, Shang W. DPP-4 inhibitor linagliptin ameliorates oxidized LDL-induced THP-1 macrophage foam cell formation and inflammation. Drug Des Devel Ther. 2020; 14:3929–3940.
55. Yu XH, Fu YC, Zhang DW, Yin K, Tang CK. Foam cells in atherosclerosis. Clin Chim Acta. 2013; 424:245–252. PMID: 23782937.
Article
56. Wu X, Hu Z, Zhou J, Liu J, Ren P, Huang X. Ferulic acid alleviates atherosclerotic plaques by inhibiting VSMC proliferation through the NO/p21 signaling pathway. J Cardiovasc Transl Res. 2022; 15:865–875. PMID: 34993756.
Article
57. Ji W, Sun J, Hu Z, Sun B. Resveratrol protects against atherosclerosis by downregulating the PI3K/AKT/mTOR signaling pathway in atherosclerosis model mice. Exp Ther Med. 2022; 23:414. PMID: 35601067.
Article
58. Singh L, Sharma S, Xu S, Tewari D, Fang J. Curcumin as a natural remedy for atherosclerosis: a pharmacological review. Molecules. 2021; 26:4036. PMID: 34279384.
Article
59. Stojković D, Petrović J, Soković M, Glamočlija J, Kukić-Marković J, Petrović S. In situ antioxidant and antimicrobial activities of naturally occurring caffeic acid, p-coumaric acid and rutin, using food systems. J Sci Food Agric. 2013; 93:3205–3208. PMID: 23553578.
Article
60. Pragasam SJ, Venkatesan V, Rasool M. Immunomodulatory and anti-inflammatory effect of p-coumaric acid, a common dietary polyphenol on experimental inflammation in rats. Inflammation. 2013; 36:169–176. PMID: 22923003.
Article
61. Stanciulescu LA, Scafa-Udriste A, Dorobantu M. Exploring the association between low-density lipoprotein subfractions and major adverse cardiovascular outcomes-a comprehensive review. Int J Mol Sci. 2023; 24:6669. PMID: 37047642.
Article
62. Kannel WB, McGee D, Gordon T. A general cardiovascular risk profile: the Framingham Study. Am J Cardiol. 1976; 38:46–51. PMID: 132862.
Article
63. Kannel WB, Castelli WP, Gordon T. Cholesterol in the prediction of atherosclerotic disease. New perspectives based on the Framingham study. Ann Intern Med. 1979; 90:85–91. PMID: 217290.
Article
64. Khatana C, Saini NK, Chakrabarti S, Saini V, Sharma A, Saini RV, Saini AK. Mechanistic insights into the oxidized low-density lipoprotein-induced atherosclerosis. Oxid Med Cell Longev. 2020; 2020:5245308. PMID: 33014272.
Article
65. Detmers PA, Hernandez M, Mudgett J, Hassing H, Burton C, Mundt S, Chun S, Fletcher D, Card DJ, Lisnock J, et al. Deficiency in inducible nitric oxide synthase results in reduced atherosclerosis in apolipoprotein E-deficient mice. J Immunol. 2000; 165:3430–3435. PMID: 10975863.
Article
66. Glass CK, Witztum JL. Atherosclerosis: the road ahead. Cell. 2001; 104:503–516. PMID: 11239408.
67. Farahi L, Sinha SK, Lusis AJ. Roles of macrophages in atherogenesis. Front Pharmacol. 2021; 12:785220. PMID: 34899348.
Article
68. Meng Z, Yan C, Deng Q, Dong X, Duan ZM, Gao DF, Niu XL. Oxidized low-density lipoprotein induces inflammatory responses in cultured human mast cells via Toll-like receptor 4. Cell Physiol Biochem. 2013; 31:842–853. PMID: 23816956.
Article
69. Plotkin JD, Elias MG, Dellinger AL, Kepley CL. NF-κB inhibitors that prevent foam cell formation and atherosclerotic plaque accumulation. Nanomedicine. 2017; 13:2037–2048. PMID: 28457935.
Article
70. Kim AT, Kim DO. Anti-inflammatory effects of vanadium-binding protein from Halocynthia roretzi in LPS-stimulated RAW264.7 macrophages through NF-κB and MAPK pathways. Int J Biol Macromol. 2019; 133:732–738. PMID: 31002910.
Article
71. Adam GO, Kim GB, Lee SJ, Lee H, Kang HS, Kim SJ. Red ginseng reduces inflammatory response via suppression MAPK/P38 signaling and p65 nuclear proteins translocation in rats and raw 264.7 macrophage. Am J Chin Med. 2019; 47:1589–1609. PMID: 31645122.
Article
72. Zhang Y, Zhang XY, Shi SR, Ma CN, Lin YP, Song WG, Guo SD. Natural products in atherosclerosis therapy by targeting PPARs: a review focusing on lipid metabolism and inflammation. Front Cardiovasc Med. 2024; 11:1372055. PMID: 38699583.
Article
73. Im YS, Gwon MH, Yun JM. Protective effects of phenethyl isothiocyanate on foam cell formation by combined treatment of oxidized low-density lipoprotein and lipopolysaccharide in THP-1 macrophage. Food Sci Nutr. 2021; 9:3269–3279. PMID: 34136191.
Article
74. Wang D, Yu X, Gao K, Li F, Li X, Pu H, Zhang P, Guo S, Wang W. Sweroside alleviates pressure overload-induced heart failure through targeting CaMKIIδ to inhibit ROS-mediated NF-κB/NLRP3 in cardiomyocytes. Redox Biol. 2024; 74:103223. PMID: 38851078.
Article
75. He JH, Li XJ, Wang SP, Guo X, Chu HX, Xu HC, Wang YS. Eugenol inhibits ox-LDL-induced proliferation and migration of human vascular smooth muscle cells by inhibiting the Ang II/MFG-E8/MCP-1 signaling cascade. J Inflamm Res. 2024; 17:641–653. PMID: 38328560.
Article
76. Xue F, Nie X, Shi J, Liu Q, Wang Z, Li X, Zhou J, Su J, Xue M, Chen WD, et al. Quercetin inhibits LPS-induced inflammation and ox-LDL-induced lipid deposition. Front Pharmacol. 2017; 8:40. PMID: 28217098.
Article
77. Liu Y, Wang K, Yang S, Xue G, Lu L. Mulberry extract upregulates cholesterol efflux and inhibits p38 MAPK-NLRP3-mediated inflammation in foam cells. Food Sci Nutr. 2023; 11:3141–3153. PMID: 37324843.
Article
78. Levi M, van der Poll T, Schultz M. Infection and inflammation as risk factors for thrombosis and atherosclerosis. Semin Thromb Hemost. 2012; 38:506–514. PMID: 22399308.
Article
79. Nelken NA, Coughlin SR, Gordon D, Wilcox JN. Monocyte chemoattractant protein-1 in human atheromatous plaques. J Clin Invest. 1991; 88:1121–1127. PMID: 1843454.
Article
80. Boyle JJ, Weissberg PL, Bennett MR. Tumor necrosis factor-α promotes macrophage-induced vascular smooth muscle cell apoptosis by direct and autocrine mechanisms. Arterioscler Thromb Vasc Biol. 2003; 23:1553–1558. PMID: 12869351.
Article
81. Mussbacher M, Derler M, Basílio J, Schmid JA. NF-κB in monocytes and macrophages - an inflammatory master regulator in multitalented immune cells. Front Immunol. 2023; 14:1134661. PMID: 36911661.
Article
82. Hashizume M, Mihara M. Atherogenic effects of TNF-α and IL-6 via up-regulation of scavenger receptors. Cytokine. 2012; 58:424–430. PMID: 22436638.
Article
83. Shen W, Anwaier G, Cao Y, Lian G, Chen C, Liu S, Tuerdi N, Qi R. Atheroprotective mechanisms of tilianin by inhibiting inflammation through down-regulating NF-κB pathway and foam cells formation. Front Physiol. 2019; 10:825. PMID: 31333487.
Article
84. Lin CC, Ou HC, Pi I, You YC. Baicalein attenuates oxLDL-induced oxidative functional damages in endothelial cells. FASEB J. 2010; 24:1038.2.
Article
85. Kuo MY, Ou HC, Lee WJ, Kuo WW, Hwang LL, Song TY, Huang CY, Chiu TH, Tsai KL, Tsai CS, et al. Ellagic acid inhibits oxidized low-density lipoprotein (OxLDL)-induced metalloproteinase (MMP) expression by modulating the protein kinase C-α/extracellular signal-regulated kinase/peroxisome proliferator-activated receptor γ/nuclear factor-κB (PKC-α/ERK/PPAR-γ/NF-κB) signaling pathway in endothelial cells. J Agric Food Chem. 2011; 59:5100–5108. PMID: 21480623.
Article
86. Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV, Orekhov AN. Mechanisms of foam cell formation in atherosclerosis. J Mol Med (Berl). 2017; 95:1153–1165. PMID: 28785870.
Article
87. Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011; 145:341–355. PMID: 21529710.
Article
88. Javadifar A, Rastgoo S, Banach M, Jamialahmadi T, Johnston TP, Sahebkar A. Foam cells as therapeutic targets in atherosclerosis with a focus on the regulatory roles of non-coding RNAs. Int J Mol Sci. 2021; 22:2529. PMID: 33802600.
Article
89. Wang N, Westerterp M. ABC transporters, cholesterol efflux, and implications for cardiovascular diseases. Adv Exp Med Biol. 2020; 1276:67–83. PMID: 32705595.
Article
90. Tang CK, Yi GH, Yang JH, Liu LS, Wang Z, Ruan CG, Yang YZ. Oxidized LDL upregulated ATP binding cassette transporter-1 in THP-1 macrophages. Acta Pharmacol Sin. 2004; 25:581–586. PMID: 15132822.
91. Zhu X, Lee JY, Timmins JM, Brown JM, Boudyguina E, Mulya A, Gebre AK, Willingham MC, Hiltbold EM, Mishra N, et al. Increased cellular free cholesterol in macrophage-specific Abca1 knock-out mice enhances pro-inflammatory response of macrophages. J Biol Chem. 2008; 283:22930–22941. PMID: 18552351.
Article
92. Singaraja RR, Brunham LR, Visscher H, Kastelein JJ, Hayden MR. Efflux and atherosclerosis: the clinical and biochemical impact of variations in the ABCA1 gene. Arterioscler Thromb Vasc Biol. 2003; 23:1322–1332. PMID: 12763760.
93. Stamatikos A, Dronadula N, Ng P, Palmer D, Knight E, Wacker BK, Tang C, Kim F, Dichek DA. ABCA1 overexpression in endothelial cells in vitro enhances ApoAI-mediated cholesterol efflux and decreases inflammation. Hum Gene Ther. 2019; 30:236–248. PMID: 30079772.
Article
94. Ma Z, Deng C, Hu W, Zhou J, Fan C, Di S, Liu D, Yang Y, Wang D. Liver X receptors and their agonists: targeting for cholesterol homeostasis and cardiovascular diseases. Curr Issues Mol Biol. 2017; 22:41–64. PMID: 27669666.
Article
95. Zhao C, Dahlman-Wright K. Liver X receptor in cholesterol metabolism. J Endocrinol. 2010; 204:233–240. PMID: 19837721.
Article
96. Murthy S, Born E, Mathur SN, Field FJ. LXR/RXR activation enhances basolateral efflux of cholesterol in CaCo-2 cells. J Lipid Res. 2002; 43:1054–1064. PMID: 12091489.
Article
97. Shin SK, Ha TY, McGregor RA, Choi MS. Long-term curcumin administration protects against atherosclerosis via hepatic regulation of lipoprotein cholesterol metabolism. Mol Nutr Food Res. 2011; 55:1829–1840. PMID: 22058071.
Article
98. Oppi S, Nusser-Stein S, Blyszczuk P, Wang X, Jomard A, Marzolla V, Yang K, Velagapudi S, Ward LJ, Yuan XM, et al. Macrophage NCOR1 protects from atherosclerosis by repressing a pro-atherogenic PPARγ signature. Eur Heart J. 2020; 41:995–1005. PMID: 31529020.
Article
99. Han L, Shen WJ, Bittner S, Kraemer FB, Azhar S. PPARs: regulators of metabolism and as therapeutic targets in cardiovascular disease. Part II: PPAR-β/δ and PPAR-γ. Future Cardiol. 2017; 13:279–296. PMID: 28581362.
Article
100. Chawla A, Boisvert WA, Lee CH, Laffitte BA, Barak Y, Joseph SB, Liao D, Nagy L, Edwards PA, Curtiss LK, et al. A PPAR gamma-LXR-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol Cell. 2001; 7:161–171. PMID: 11172721.
Article
101. Franceschelli S, De Cecco F, Pesce M, Ripari P, Guagnano MT, Nuevo AB, Grilli A, Sancilio S, Speranza L. Hydroxytyrosol reduces foam cell formation and endothelial inflammation regulating the PPARγ/LXRα/ABCA1 pathway. Int J Mol Sci. 2023; 24:2057. PMID: 36768382.
Article
102. Zhao GJ, Tang SL, Lv YC, Ouyang XP, He PP, Yao F, Chen WJ, Lu Q, Tang YY, Zhang M, et al. Antagonism of betulinic acid on LPS-mediated inhibition of ABCA1 and cholesterol efflux through inhibiting nuclear factor-kappaB signaling pathway and miR-33 expression. PLoS One. 2013; 8:e74782. PMID: 24086374.
Article
103. Yan S, Zhou M, Zheng X, Xing Y, Dong J, Yan M, Li R. Anti-inflammatory effect of curcumin on the mouse model of myocardial infarction through regulating macrophage polarization. Mediators Inflamm. 2021; 2021:9976912. PMID: 34462629.
Article
104. Kunjathoor VV, Febbraio M, Podrez EA, Moore KJ, Andersson L, Koehn S, Rhee JS, Silverstein R, Hoff HF, Freeman MW. Scavenger receptors class A-I/II and CD36 are the principal receptors responsible for the uptake of modified low density lipoprotein leading to lipid loading in macrophages. J Biol Chem. 2002; 277:49982–49988. PMID: 12376530.
Article
105. Pirillo A, Norata GD, Catapano AL. LOX-1, OxLDL, and atherosclerosis. Mediators Inflamm. 2013; 2013:152786. PMID: 23935243.
Article
106. Sawamura T, Kume N, Aoyama T, Moriwaki H, Hoshikawa H, Aiba Y, Tanaka T, Miwa S, Katsura Y, Kita T, et al. An endothelial receptor for oxidized low-density lipoprotein. Nature. 1997; 386:73–77. PMID: 9052782.
Article
107. Hofnagel O, Luechtenborg B, Stolle K, Lorkowski S, Eschert H, Plenz G, Robenek H. Proinflammatory cytokines regulate LOX-1 expression in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2004; 24:1789–1795. PMID: 15271788.
Article
108. Mehta JL, Sanada N, Hu CP, Chen J, Dandapat A, Sugawara F, Satoh H, Inoue K, Kawase Y, Jishage K, et al. Deletion of LOX-1 reduces atherogenesis in LDLR knockout mice fed high cholesterol diet. Circ Res. 2007; 100:1634–1642. PMID: 17478727.
Article
109. Manning-Tobin JJ, Moore KJ, Seimon TA, Bell SA, Sharuk M, Alvarez-Leite JI, de Winther MP, Tabas I, Freeman MW. Loss of SR-A and CD36 activity reduces atherosclerotic lesion complexity without abrogating foam cell formation in hyperlipidemic mice. Arterioscler Thromb Vasc Biol. 2009; 29:19–26. PMID: 18948635.
Article
110. Janabi M, Yamashita S, Hirano K, Sakai N, Hiraoka H, Matsumoto K, Zhang Z, Nozaki S, Matsuzawa Y. Oxidized LDL-induced NF-kappa B activation and subsequent expression of proinflammatory genes are defective in monocyte-derived macrophages from CD36-deficient patients. Arterioscler Thromb Vasc Biol. 2000; 20:1953–1960. PMID: 10938017.
Article
111. Febbraio M, Podrez EA, Smith JD, Hajjar DP, Hazen SL, Hoff HF, Sharma K, Silverstein RL. Targeted disruption of the class B scavenger receptor CD36 protects against atherosclerotic lesion development in mice. J Clin Invest. 2000; 105:1049–1056. PMID: 10772649.
Article
112. Li X, Zhou Y, Yu C, Yang H, Zhang C, Ye Y, Xiao S. Paeonol suppresses lipid accumulation in macrophages via upregulation of the ATP-binding cassette transporter A1 and downregulation of the cluster of differentiation 36. Int J Oncol. 2015; 46:764–774. PMID: 25405950.
Article
113. Wang T, Lu H. Ganoderic acid A inhibits ox-LDL-induced THP-1-derived macrophage inflammation and lipid deposition via Notch1/PPARγ/CD36 signaling. Adv Clin Exp Med. 2021; 30:1031–1041. PMID: 34329545.
Article
114. Shen CY, Wang TX, Jiang JG, Huang CL, Zhu W. Bergaptol from blossoms of Citrus aurantium L. var. amara Engl inhibits LPS-induced inflammatory responses and ox-LDL-induced lipid deposition. Food Funct. 2020; 11:4915–4926. PMID: 32432251.
Article
Full Text Links
  • NRP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2025 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr