Nutr Res Pract.  2014 Oct;8(5):521-525. 10.4162/nrp.2014.8.5.521.

Artemisinin attenuates platelet-derived growth factor BB-induced migration of vascular smooth muscle cells

Affiliations
  • 1Department of Medical Science, School of Medicine Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea. hhong@kku.ac.kr
  • 2Department of Biomedical Laboratory Science, Kyungbok University, Gyeonggi-do 487-717, Korea.
  • 3Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongsangbuk-do 780-714, Korea.

Abstract

BACKGROUND/OBJECTIVES
Artemisinin (AT), an active compound in Arternisia annua, is well known as an anti-malaria drug. It is also known to have several effects including anti-oxidant, anti-inflammation, and anti-cancer activities. To date, the effect of AT on vascular disorders has not been studied. In this study, we investigated the effects of AT on the migration and proliferation of vascular smooth muscle cells (VSMC) stimulated by platelet-derived growth factor BB (PDGF-BB).
MATERIALS/METHODS
Aortic smooth muscle cells were isolated from Sprague-Dawley rats. PDGF-BB stimulated VSMC migration was measured by the scratch wound healing assay and the Boyden chamber assay. Cell viability was determined by using an EZ-Cytox Cell Viability Assay Kit. The production of reactive oxygen species (ROS) in PDGF-BB stimulated VSMC was measured through H2DCF-DA staining. We also determined the expression levels of signal proteins relevant to ROS, including measures of extracellular signal-regulated kinase (ERK) 1/2 measured by western blot analysis and matrix metalloproteinase (MMP) 9 measured by reverse transcription-polymerase chain reaction (RT-PCR).
RESULTS
AT (10 microM and 30 microM) significantly reduced the proliferation and migration of PDGF-BB stimulated VSMC in a dose-dependent manner. The production of ROS, normally induced by PDGF-BB, is reduced by treatment with AT at both concentrations. PDGF-BB stimulated VSMC treated with AT (10 microM and 30 microM) have reduced phosphorylation of ERK1/2 and inhibited MMP9 expression compared to untreated PDGF-BB stimulated VSMC.
CONCLUSIONS
We suggest, based on these results, that AT may exert an anti-atherosclerotic effect on PDGF-BB stimulated VSMCs by inhibiting their proliferation and migration through down-regulation of ERK1/2 and MMP9 phosphorylation.

Keyword

Artemisinin; vascular smooth muscle cells; platelet-derived growth factor-BB; ERK1/2; MMP9

MeSH Terms

Blotting, Western
Cell Survival
Down-Regulation
Muscle, Smooth, Vascular*
Myocytes, Smooth Muscle
Phosphorylation
Phosphotransferases
Platelet-Derived Growth Factor
Rats, Sprague-Dawley
Reactive Oxygen Species
Wound Healing
Phosphotransferases
Platelet-Derived Growth Factor
Reactive Oxygen Species

Figure

  • Fig. 1 The effect of artemisinin on proliferation in PDGF-BB-stimulated VSMC. (A) The chemical structure of artemisinin. (B) VSMC were seeded (2 × 103 cells/well) and incubated for 12 h. VSMC were incubated in serum-free media for 24 h and then cells were treated with the indicated concentrations of AT (10 µM and 30 µM) in the presence or absence of PDGF-BB for 24 h. The graph indicated the representative cell viability from three independent experiments. Data are mean ± standard error. Values with the same superscript letter are not significantly different by Duncan's multiple range test (P < 0.05).

  • Fig. 2 Effects of AT on scratch wound healing in PDGF-BB-stimulated VSMC. VSMC were treated with AT (10 µM and 30 µM) and PDGF-BB (10 ng/mL) for 24 h. (A) Dotted white lines indicate the wounded area from the initial scratches. Magnification, × 200. Bar = 500 µm. The number in the top right of each image is concentration of PDGF-BB/concentration of AT. (B) Amount of migration relative to the untreated control (%). Results are presented as mean ± standard error. Values with the same superscript letter are not significantly different by Duncan's multiple range test (P < 0.05).

  • Fig. 3 Effects of AT on migration in PDGF-BB-stimulated VSMC. VSMC were treated with AT (10 µM and 30 µM) and PDGF-BB (10 ng/mL) for 90 min. (A) Migrating VSMC on membranes, the spots are Diff quick-stained cells. The number at the top right corner of each image is the concentration of PDGF-BB/concentration of AT. Magnification, × 200. Bar = 500 µm. (B) Migrated cell counts indicated relative to the untreated control (%). Results are presented as mean ± standard error. Values with the same superscript letter are not significantly different by Duncan's multiple range test (P < 0.05).

  • Fig. 4 Effects of AT on migration in H2O2-stimulated VSMC. (A) ROS production was assessed using 1 µg/mL DAPI (upper panel) or H2DCFDA (lower panel) in PDGF-BB-induced proliferating VSMC, Magnification, × 400. Bar = 50 µm. (B) Migrating VSMC on membranes, the spots are Hoescht stained cells. VSMC were treated with AT (10 µM and 30 µM) and H2O2 (100 µM) for 90 min. The number at the top right corner of each image is the concentration of H2O2/concentration of AT. Magnification, 200 ×. Bar = 500 µm. (C) Migrating cell number relative to the untreated control (%). Results are presented as mean ± standard error. Values with the same superscript letters are not significantly different by Duncan's multiple range test (P < 0.05).

  • Fig. 5 Effect of AT on PDGF-BB-induced phosphorylation of ERK1/2 and MMP9 transcription in VSMC. (A) VSMC were pretreated with or without AT (10 µM and 30 µM) for 1 h and then stimulated with 10 ng/mL of PDGF-BB for 15 min. For statistical analysis, densitometry of the band representing the phosphorylated form of ERK1/2 normalized to the total expression of ERK is considered 100%. (B) The levels of MMP9 mRNA were determined by RT-PCR. VSMC were pretreated with or without AT (10 µM and 30 µM) for 1 h and then stimulated with 10 ng/mL of PDGF-BB for 6 h. cDNA was synthesized using 1 µg of total RNA each treatment group. After PCR and electrophoresis on a 1% agarose gel, the bands were measured using densitometry of the ethidium bromide stained DNA under UV light. The background intensity is considered 100%, and the expression of MMP9 is defined relative to the GAPDH control. Results are presented as the mean ± standard error of three independent experiments. Values with the same superscript letters are not significantly different by Duncan's multiple range test (P < 0.05).


Reference

1. Ministry of Health and Welfare, Korea Centers for Disease Control and Prevention. Korea Health Statistics 2012: Korea National Health and Nutrition Examination Survey (KNHANES V-3). Cheongwon: Korea Centers for Disease Control and Prevention;2013.
2. Buttari B, Profumo E, Businaro R, Saso L, Capoano R, Salvati B, Riganò R. Oxidized haemoglobin-driven endothelial dysfunction and immune cell activation: novel therapeutic targets for atherosclerosis. Curr Med Chem. 2013; 20:4806–4814.
Article
3. Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature. 1993; 362:801–809.
Article
4. Shah PK. Inflammation, neointimal hyperplasia, and restenosis: as the leukocytes roll, the arteries thicken. Circulation. 2003; 107:2175–2177.
5. Kondo T, Konishi F, Inui H, Inagami T. Differing signal transductions elicited by three isoforms of platelet-derived growth factor in vascular smooth muscle cells. J Biol Chem. 1993; 268:4458–4464.
Article
6. Ross R, Raines EW, Bowen-Pope DF. The biology of platelet-derived growth factor. Cell. 1986; 46:155–169.
Article
7. Park SS, Kim WJ, Moon SK. Role of decursin in TNF-α-stimulated migration, invasion, and matrix metalloproteinase-9 induction in vascular smooth muscle cells. Food Sci Biotechnol. 2011; 20:913–918.
Article
8. Moon CY, Ku CR, Cho YH, Lee EJ. Protocatechuic aldehyde inhibits migration and proliferation of vascular smooth muscle cells and intravascular thrombosis. Biochem Biophys Res Commun. 2012; 423:116–121.
Article
9. Lyle AN, Griendling KK. Modulation of vascular smooth muscle signaling by reactive oxygen species. Physiology (Bethesda). 2006; 21:269–280.
Article
10. Kyaw M, Yoshizumi M, Tsuchiya K, Kirima K, Tamaki T. Antioxidants inhibit JNK and p38 MAPK activation but not ERK 1/2 activation by angiotensin II in rat aortic smooth muscle cells. Hypertens Res. 2001; 24:251–261.
Article
11. Zhou Y, Yan H, Guo M, Zhu J, Xiao Q, Zhang L. Reactive oxygen species in vascular formation and development. Oxid Med Cell Longev. 2013; 2013:374963.
Article
12. Park J, Ha H, Seo J, Kim MS, Kim HJ, Huh KH, Park K, Kim YS. Mycophenolic acid inhibits platelet-derived growth factor-induced reactive oxygen species and mitogen-activated protein kinase activation in rat vascular smooth muscle cells. Am J Transplant. 2004; 4:1982–1990.
Article
13. Bornfeldt KE, Raines EW, Nakano T, Graves LM, Krebs EG, Ross R. Insulin-like growth factor-I and platelet-derived growth factor-BB induce directed migration of human arterial smooth muscle cells via signaling pathways that are distinct from those of proliferation. J Clin Invest. 1994; 93:1266–1274.
Article
14. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res. 2003; 92:827–839.
15. Kohler M, Haerdi W, Christen P, Veuthey JL. Extraction of artemisinin and artemisinic acid from Artemisia annua L. using supercritical carbon dioxide. J Chromatogr A. 1997; 785:353–360.
Article
16. Ho WE, Peh HY, Chan TK, Wong WS. Artemisinins: pharmacological actions beyond anti-malarial. Pharmacol Ther. 2014; 142:126–139.
Article
17. Won KJ, Lee SC, Lee CK, Lee HM, Lee SH, Fang Z, Choi OB, Jin M, Kim J, Park T, Choi WS, Kim SK, Kim B. Cordycepin attenuates neointimal formation by inhibiting reactive oxygen species-mediated responses in vascular smooth muscle cells in rats. J Pharmacol Sci. 2009; 109:403–412.
Article
18. Singh NP, Lai HC. Artemisinin induces apoptosis in human cancer cells. Anticancer Res. 2004; 24:2277–2280.
19. Choi OB, Park JH, Lee YJ, Lee CK, Won KJ, Kim J, Lee HM, Kim B. Olibanum extract inhibits vascular smooth muscle cell migration and proliferation in response to platelet-derived growth factor. Korean J Physiol Pharmacol. 2009; 13:107–113.
Article
20. Jiang D, Li D, Wu W. Inhibitory effects and mechanisms of luteolin on proliferation and migration of vascular smooth muscle cells. Nutrients. 2013; 5:1648–1659.
Article
21. Konneh MK, Rutherford C, Li SR, Anggård EE, Ferns GA. Vitamin E inhibits the intimal response to balloon catheter injury in the carotid artery of the cholesterol-fed rat. Atherosclerosis. 1995; 113:29–39.
Article
Full Text Links
  • NRP
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