DOI QR코드

DOI QR Code

Ginsenoside compound K protects human umbilical vein endothelial cells against oxidized low-density lipoprotein-induced injury via inhibition of nuclear factor-κB, p38, and JNK MAPK pathways

  • Lu, Shan (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Luo, Yun (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Zhou, Ping (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Yang, Ke (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Sun, Guibo (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Sun, Xiaobo (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences)
  • Received : 2017.03.15
  • Accepted : 2017.09.04
  • Published : 2019.01.15

Abstract

Background: Oxidized low-density lipoprotein (ox-LDL) causes vascular endothelial cell inflammatory response and apoptosis and plays an important role in the development and progression of atherosclerosis. Ginsenoside compound K (CK), a metabolite produced by the hydrolysis of ginsenoside Rb1, possesses strong anti-inflammatory effects. However, whether or not CK protects ox-LDL-damaged endothelial cells and the potential mechanisms have not been elucidated. Methods: In our study, cell viability was tested using a 3-(4, 5-dimethylthiazol-2yl-)-2,5-diphenyl tetrazolium bromide (MTT) assay. Expression levels of interleukin-6, monocyte chemoattractant protein-1, tumor necrosis factor-${\alpha}$, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 were determined by enzyme-linked immunosorbent assay and Western blotting. Mitochondrial membrane potential (${\Delta}{\Psi}m$) was detected using JC-1. The cell apoptotic percentage was measured by the Annexin V/ propidium iodide (PI) assay, lactate dehydrogenase, and caspase-3 expression. Apoptosis-related proteins, nuclear factor $(NF)-{\kappa}B$, and mitogen-activated protein kinases (MAPK) signaling pathways protein expression were quantified by Western blotting. Results: Our results demonstrated that CK could ameliorate ox-LDL-induced human umbilical vein endothelial cells (HUVECs) inflammation and apoptosis, $NF-{\kappa}B$ nuclear translocation, and the phosphorylation of p38 and c-Jun N-terminal kinase (JNK). Moreover, anisomycin, an activator of p38 and JNK, significantly abolished the anti-apoptotic effects of CK. Conclusion: These results demonstrate that CK prevents ox-LDL-induced HUVECs inflammation and apoptosis through inhibiting the $NF-{\kappa}B$, p38, and JNK MAPK signaling pathways. Thus, CK is a candidate drug for atherosclerosis treatment.

Keywords

References

  1. Siegel D, Devaraj S, Mitra A, Raychaudhuri SP, Raychaudhuri SK, Jialal I. Inflammation, atherosclerosis, and psoriasis. Clin Rev Allergy Immunol 2013;44:194-204. https://doi.org/10.1007/s12016-012-8308-0
  2. Gutierrez E, Flammer AJ, Lerman LO, Elizaga J, Lerman A, Fernandez-Aviles F. Endothelial dysfunction over the course of coronary artery disease. Eur Heart J 2013;34:3175-81. https://doi.org/10.1093/eurheartj/eht351
  3. Matsuura E, Atzeni F, Sarzi-Puttini P, Turiel M, Lopez LR, Nurmohamed MT. Is atherosclerosis an autoimmune disease? BMC Med 2014;12:47. https://doi.org/10.1186/1741-7015-12-47
  4. Chistiakov DA, Orekhov AN, Bobryshev YV. LOX-1-mediated effects on vascular cells in atherosclerosis. Cell Physiol Biochem 2016;38:1851-9. https://doi.org/10.1159/000443123
  5. Ogura S, Kakino A, Sato Y, Fujita Y, Iwamoto S, Otsui K, Yoshimoto R, Sawamura T. Lox-1: the multifunctional receptor underlying cardiovascular dysfunction. Circ J 2009;73:1993-9. https://doi.org/10.1253/circj.CJ-09-0587
  6. Pirillo A, Norata GD, Catapano AL. LOX-1, OxLDL, and atherosclerosis. Mediators Inflamm 2013;2013:152786.
  7. Chae S, Kang KA, Chang WY, Kim MJ, Lee SJ, Lee YS, Kim HS, Kim DH, Hyun JW. Effect of compound K, a metabolite of ginseng saponin, combined with gamma-ray radiation in human lung cancer cells in vitro and in vivo. J Agric Food Chem 2009;57:5777-82. https://doi.org/10.1021/jf900331g
  8. Shin YW, Kim DH. Antipruritic effect of ginsenoside rb1 and compound k in scratching behavior mouse models. J Pharmacol Sci 2005;99:83-8. https://doi.org/10.1254/jphs.FP0050260
  9. Kim K, Park M, Lee YM, Rhyu MR, Kim HY. Ginsenoside metabolite compound K stimulates glucagon-like peptide-1 secretion in NCI-H716 cells via bile acid receptor activation. Arch Pharm Res 2014;37:1193-200. https://doi.org/10.1007/s12272-014-0362-0
  10. Zhang Z, Du GJ, Wang CZ, Wen XD, Calway T, Li Z, He TC, Du W, Bissonnette M, Musch MW, et al. Compound K, a ginsenoside metabolite, inhibits colon cancer growth via multiple pathways including p53-p21 interactions. Int J Mol Sci 2013;14:2980-95. https://doi.org/10.3390/ijms14022980
  11. Joh EH, Lee IA, Jung IH, Kim DH. Ginsenoside Rb1 and its metabolite compound K inhibit IRAK-1 activationethe key step of inflammation. Biochem Pharmacol 2011;82:278-86. https://doi.org/10.1016/j.bcp.2011.05.003
  12. Gao YL, Liu ZF, Li CM, Shen JY, Yin HX, Li GS. Subchronic toxicity studies with ginsenoside compound K delivered to dogs via intravenous administration. Food Chem Toxicol 2011;49:1857-62. https://doi.org/10.1016/j.fct.2011.05.003
  13. Tsutsumi YM, Tsutsumi R, Mawatari K, Nakaya Y, Kinoshita M, Tanaka K, Oshita S. Compound K, a metabolite of ginsenosides, induces cardiac protection mediated nitric oxide via Akt/PI3K pathway. Life Sci 2011;88:725-9. https://doi.org/10.1016/j.lfs.2011.02.011
  14. Lee HU, Bae EA, Han MJ, Kim NJ, Kim DH. Hepatoprotective effect of ginsenoside Rb1 and compound K on tert-butyl hydroperoxide-induced liver injury. Liver Int 2005;25:1069-73. https://doi.org/10.1111/j.1478-3231.2005.01068.x
  15. Lee E, Choi J, Kim MS, You HJ, Ji GE, Kang Y. Ginsenoside metabolite compound K differentially antagonizing tumor necrosis factor-a-induced monocytee endothelial trafficking. Chem-Biol Interact 2011;194:13-22. https://doi.org/10.1016/j.cbi.2011.08.008
  16. Qin M, Luo Y, Meng XB, Wang M, Wang HW, Song SY, Ye JX, Yao F, Wu P, Sun GB, et al. Myricitrin attenuates endothelial cell apoptosis to prevent atherosclerosis: an insight into PI3K/Akt activation and STAT3 signaling pathways. Vascul Pharmacol 2015;70:23-34. https://doi.org/10.1016/j.vph.2015.03.002
  17. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation 2002;105:1135-43. https://doi.org/10.1161/hc0902.104353
  18. Fan J, Watanabe T. Inflammatory reactions in the pathogenesis of atherosclerosis. J Atheroscler Thromb 2003;10:63-71. https://doi.org/10.5551/jat.10.63
  19. Li D, Chen H, Romeo F, Sawamura T, Saldeen T, Mehta JL. Statins modulate oxidized low-density lipoprotein-mediated adhesion molecule expression in human coronary artery endothelial cells: role of LOX-1. J Pharmacol Exp Ther 2002;302:601-5. https://doi.org/10.1124/jpet.102.034959
  20. Zheng J, Liu B, Lun Q, Gu X, Pan B, Zhao Y, Xiao W, Li J, Tu P. Longxuetongluo capsule inhibits atherosclerosis progression in high-fat dietinduced ApoE-/- mice by improving endothelial dysfunction. Atherosclerosis 2016;255:156-63. https://doi.org/10.1016/j.atherosclerosis.2016.08.022
  21. Mehta JL, Chen J, Hermonat PL, Romeo F, Novelli G. Lectin-like, oxidized lowdensity lipoprotein receptor-1 (LOX-1): a critical player in the development of atherosclerosis and related disorders. Cardiovasc Res 2006;69:36-45. https://doi.org/10.1016/j.cardiores.2005.09.006
  22. Hansson GK. Inflammatory mechanisms in atherosclerosis. J Thromb Haemost 2009;7:328-31. https://doi.org/10.1111/j.1538-7836.2009.03416.x
  23. Bao MH, Zhang YW, Zhou HH. Paeonol suppresses oxidized low-density lipoprotein induced endothelial cell apoptosis via activation of LOX-1/p38MAPK/NF-kappaB pathway. J Ethnopharmacol 2013;146:543-51. https://doi.org/10.1016/j.jep.2013.01.019
  24. Jiang JX, Zhang SJ, Liu YN, Lin XX, Sun YH, Shen HJ, Yan XF, Xie QM. EETs alleviate ox-LDL-induced inflammation by inhibiting LOX-1 receptor expression in rat pulmonary arterial endothelial cells. Eur J Pharmacol 2014;727:43-51. https://doi.org/10.1016/j.ejphar.2014.01.045
  25. Zhang H, Ma G, Yao Y, Qian H, Li W, Chen X, Jiang W, Zheng R. Olmesartan attenuates the impairment of endothelial cells induced by oxidized low density lipoprotein through downregulating expression of LOX-1. Int J Mol Sci 2012;13:1512-23. https://doi.org/10.3390/ijms13021512
  26. Zhang Y, Mu Q, Zhou Z, Song H, Zhang Y, Wu F, Jiang M, Wang F, Zhang W, Li L, et al. Protective effect of irisin on atherosclerosis via suppressing oxidized low density lipoprotein induced vascular inflammation and endothelial dysfunction. PLoS One 2016;11, e158038.
  27. Qiu L, Xu R, Wang S, Li S, Sheng H, Wu J, Qu Y. Honokiol ameliorates endothelial dysfunction through suppression of PTX3 expression, a key mediator of IKK/IkappaB/NF-kappaB, in atherosclerotic cell model. Exp Mol Med 2015;47, e171. https://doi.org/10.1038/emm.2015.37
  28. Chen J, Mehta JL, Haider N, Zhang X, Narula J, Li D. Role of caspases in Ox-LDLinduced apoptotic cascade in human coronary artery endothelial cells. Circ Res 2004;94:370-6. https://doi.org/10.1161/01.RES.0000113782.07824.BE
  29. Stoneman VE, Bennett MR. Role of apoptosis in atherosclerosis and its therapeutic implications. Clin Sci (Lond) 2004;107:343-54. https://doi.org/10.1042/CS20040086
  30. Li D, Mehta JL. Upregulation of endothelial receptor for oxidized LDL (LOX-1) by oxidized LDL and implications in apoptosis of human coronary artery endothelial cells: evidence from use of antisense LOX-1 mRNA and chemical inhibitors. Arterioscler Thromb Vasc Biol 2000;20:1116-22. https://doi.org/10.1161/01.ATV.20.4.1116
  31. Su X, Ao L, Shi Y, Johnson TR, Fullerton DA, Meng X. Oxidized low density lipoprotein induces bone morphogenetic protein-2 in coronary artery endothelial cells via Toll-like receptors 2 and 4. J Biol Chem 2011;286:12213-20. https://doi.org/10.1074/jbc.M110.214619
  32. Choi JS, Choi YJ, Shin SY, Li J, Kang SW, Bae JY, Kim DS, Ji GE, Kang JS, Kang YH. Dietary flavonoids differentially reduce oxidized LDL-induced apoptosis in human endothelial cells: role of MAPK- and JAK/STAT-signaling. J Nutr 2008;138:983-90. https://doi.org/10.1093/jn/138.6.983
  33. Yin Y, Liu W, Ji G, Dai Y. The essential role of p38 MAPK in mediating the interplay of oxLDL and IL-10 in regulating endothelial cell apoptosis. Eur J Cell Biol 2013;92:150-9. https://doi.org/10.1016/j.ejcb.2013.01.001

Cited by

  1. Exosome-encapsulated miR-505 from ox-LDL-treated vascular endothelial cells aggravates atherosclerosis by inducing NET formation vol.51, pp.12, 2019, https://doi.org/10.1093/abbs/gmz123
  2. Genome-scale CRISPR screening for potential targets of ginsenoside compound K vol.11, pp.1, 2020, https://doi.org/10.1038/s41419-020-2234-5
  3. Intracellular synthesis of gold nanoparticles by Gluconacetobacter liquefaciens for delivery of peptide CopA3 and ginsenoside and anti-inflammatory effect on lipopolysaccharide-activated macrophages vol.48, pp.1, 2020, https://doi.org/10.1080/21691401.2020.1748639
  4. Correlation between Mitochondrial Dysfunction, Cardiovascular Diseases, and Traditional Chinese Medicine vol.2020, 2020, https://doi.org/10.1155/2020/2902136
  5. Ginsenoside Compound K Attenuates Ox-LDL-Mediated Macrophage Inflammation and Foam Cell Formation via Autophagy Induction and Modulating NF-κB, p38, and JNK MAPK Signaling vol.11, 2019, https://doi.org/10.3389/fphar.2020.567238
  6. MicroRNAs mediate the anti-tumor and protective effects of ginsenosides vol.72, pp.8, 2019, https://doi.org/10.1080/01635581.2019.1675722
  7. FGF20 Protected Against BBB Disruption After Traumatic Brain Injury by Upregulating Junction Protein Expression and Inhibiting the Inflammatory Response vol.11, 2019, https://doi.org/10.3389/fphar.2020.590669
  8. Ginsenosides in vascular remodeling: Cellular and molecular mechanisms of their therapeutic action vol.169, 2021, https://doi.org/10.1016/j.phrs.2021.105647
  9. The promising therapeutic potentials of ginsenosides mediated through p38 MAPK signaling inhibition vol.7, pp.11, 2019, https://doi.org/10.1016/j.heliyon.2021.e08354
  10. Characterization of hyperglycemia due to sub-chronic administration of red ginseng extract via comparative global proteomic analysis vol.11, pp.1, 2019, https://doi.org/10.1038/s41598-021-91664-8