Korean J Physiol Pharmacol.  2015 Jul;19(4):335-340. 10.4196/kjpp.2015.19.4.335.

Zerumbone, Sesquiterpene Photochemical from Ginger, Inhibits Angiogenesis

Affiliations
  • 1Department of Biomedical Science, Catholic University of Daegu, Gyeongsan 712-702, Korea. toto0818@cu.ac.kr

Abstract

Here, we investigated the role of zerumbone, a natural cyclic sesquiterpene of Zingiber zerumbet Smith, on angiogenesis using human umbilical vein endothelial cells (HUVECs). Zerumbone inhibited HUVECs proliferation, migration and tubule formation, as well as angiogenic activity by rat aorta explants. In particular, zerumbone inhibited phosphorylation of vascular endothelial growth factor receptor-2 and fibroblast growth factor receptor-1, which are key regulators of endothelial cell function and angiogenesis. In vivo matrigel plug assay in mice demonstrated significant decrease in vascularization and hemoglobin content in the plugs from zerumbone-treated mice, compared with control mice. Overall, these results suggest that zerumbone inhibits various attributes of angiogenesis, which might contribute to its reported antitumor effects.

Keyword

Angiogenesis; Zerumbone; Vascular endothelial growth factor receptor; Human umbilical vein endothelial cells

MeSH Terms

Animals
Aorta
Endothelial Cells
Fibroblast Growth Factors
Ginger*
Human Umbilical Vein Endothelial Cells
Mice
Phosphorylation
Rats
Receptors, Vascular Endothelial Growth Factor
Vascular Endothelial Growth Factor Receptor-2
Fibroblast Growth Factors
Receptors, Vascular Endothelial Growth Factor
Vascular Endothelial Growth Factor Receptor-2

Figure

  • Fig. 1 Structure of Zerumbone.

  • Fig. 2 Inhibition of endothelial cell proliferation, migration, and capillary-like tubule formation by zerumbone. HUVECs were plated in 96-well plates, allowed to attach overnight, and then cultured for 24 h with indicated concentration of zerumbone. (A) The proliferation and (B) LDH releases were determined as described in the Methods section. (C) For cell migration, a monolayer of inactivated HUVECs was wounded by scratching with a 0.1 ml pipette tip, and fresh medium containing indicated concentration of zerumbone was added. After 18 h, migration of HUVECs was measured as described in the Methods section. (D) For capillary-like tubule formation, HUVECs were seeded onto matrigel-coated 48-well plates and incubated with indicated concentration of zerumbone for 18 h. Endothelial tubules were photographed and quantitated. The results are means±SEM for at least three independent experiments. ***p<0.001 versus 0 µM zerumbone-treated cells.

  • Fig. 3 Effect of zerumbone on microvessel outgrowth arising from rat aortic rings. Aortic rings isolated from SD rats were embedded in matrigel in 48-well plates, and then fed with medium containing zerumbone for 7 days. Photographs are representative of three independent experiments.

  • Fig. 4 Effects of zerumbone on VEGFR2- and FGFR1-mediated signaling pathways. (A) HUVECs were treated with various concentrations of zerumbone and stimulated with human VEGF (10 ng/ml) for 15 min. Phosphorylation of VEGFR2 by VEGF was determined using Western blot. (B) HUVECs were treated with zerumbone and stimulated with human bFGF (50 ng/ml) for 15 min. Phosphorylation of of FGFR by bFGF were determined using Western blot. The bands were quantified using image analysis software and their relative intensity was expressed as fold against the image of the VEGF- or bFGF- stimulated cells. ***p<0.001 versus VEGF or bFGF+0 µM zerumbone-treated cells.

  • Fig. 5 Effects of zerumbone in vivo angiogenesis by matrigel plug assay. (A) Matrigel containing mouse VEGF (100 ng/ml), mouse bFGF (100 ng/ml), and zerumbone was subcutaneously injected into C57BL/6J mice. After 7 days, matrigels ware removed and photographed. (B) Paraffin sections of matrigel plugs were stained with hematoxylin and eosin. Original magnification, ×100; Scale bar 100 µm. (C) The quantification of neovascularization on the matrigel was performed by measuring the hemoglobin content. The results are reported as the mean±SEM of three independent experiments. Statistical significance is based on the difference when compared with non-treated animals, **p<0.01, ***p<0.001.


Reference

1. Liu RH. Health-promoting components of fruits and vegetables in the diet. Adv Nutr. 2013; 4:384S–392S. PMID: 23674808.
Article
2. Key TJ, Appleby PN, Crowe FL, Bradbury KE, Schmidt JA, Travis RC. Cancer in British vegetarians: updated analyses of 4998 incident cancers in a cohort of 32,491 meat eaters, 8612 fish eaters, 18,298 vegetarians, and 2246 vegans. Am J Clin Nutr. 2014; 100(Suppl 1):378S–385S. PMID: 24898235.
Article
3. Bhathena SJ, Velasquez MT. Beneficial role of dietary phytoestrogens in obesity and diabetes. Am J Clin Nutr. 2002; 76:1191–1201. PMID: 12450882.
Article
4. Canter PH, Thomas H, Ernst E. Bringing medicinal plants into cultivation: opportunities and challenges for biotechnology. Trends Biotechnol. 2005; 23:180–185. PMID: 15780709.
Article
5. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000; 407:249–257. PMID: 11001068.
Article
6. Montesano R, Vassalli JD, Baird A, Guillemin R, Orci L. Basic fibroblast growth factor induces angiogenesis in vitro. Proc Natl Acad Sci U S A. 1986; 83:7297–7301. PMID: 2429303.
Article
7. Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer. 2013; 13:871–882. PMID: 24263190.
Article
8. Carmeliet P. Angiogenesis in health and disease. Nat Med. 2003; 9:653–660. PMID: 12778163.
Article
9. Koch S, Tugues S, Li X, Gualandi L, Claesson-Welsh L. Signal transduction by vascular endothelial growth factor receptors. Biochem J. 2011; 437:169–183. PMID: 21711246.
Article
10. Spano D, Zollo M. Tumor microenvironment: a main actor in the metastasis process. Clin Exp Metastasis. 2012; 29:381–395. PMID: 22322279.
Article
11. Gao F, Liang B, Reddy ST, Farias-Eisner R, Su X. Role of inflammation-associated microenvironment in tumorigenesis and metastasis. Curr Cancer Drug Targets. 2014; 14:30–45. PMID: 24200082.
Article
12. Bellou S, Pentheroudakis G, Murphy C, Fotsis T. Anti-angiogenesis in cancer therapy: Hercules and hydra. Cancer Lett. 2013; 338:219–228. PMID: 23707856.
Article
13. Dev S. Studies in sesquiterpenes-XVI: zerumbone, a monocyclic sesquiterpene ketone. Tetrahedron. 1960; 8:171–180.
14. Murakami A, Takahashi M, Jiwajinda S, Koshimizu K, Ohigashi H. Identification of zerumbone in Zingiber zerumbet Smith as a potent inhibitor of 12-O-tetradecanoylphorbol-13-acetate-induced Epstein-Barr virus activation. Biosci Biotechnol Biochem. 1999; 63:1811–1812. PMID: 10586508.
15. Kirana C, McIntosh GH, Record IR, Jones GP. Antitumor activity of extract of Zingiber aromaticum and its bioactive sesquiterpenoid zerumbone. Nutr Cancer. 2003; 45:218–225. PMID: 12881017.
Article
16. Huang GC, Chien TY, Chen LG, Wang CC. Antitumor effects of zerumbone from Zingiber zerumbet in P-388D1 cells in vitro and in vivo. Planta Med. 2005; 71:219–224. PMID: 15770541.
17. Szabolcs A, Tiszlavicz L, Kaszaki J, Pósa A, Berkó A, Varga IS, Boros I, Szüts V, Lonovics J, Takács T. Zerumbone exerts a beneficial effect on inflammatory parameters of cholecystokinin octapeptide-induced experimental pancreatitis but fails to improve histology. Pancreas. 2007; 35:249–255. PMID: 17895846.
Article
18. Kitayama T. Attractive reactivity of a natural product, zerumbone. Biosci Biotechnol Biochem. 2011; 75:199–207. PMID: 21307568.
Article
19. Shamoto T, Matsuo Y, Shibata T, Tsuboi K, Nagasaki T, Takahashi H, Funahashi H, Okada Y, Takeyama H. Zerumbone inhibits angiogenesis by blocking NF-κB activity in pancreatic cancer. Pancreas. 2014; 43:396–404. PMID: 24622069.
Article
20. Tsuboi K, Matsuo Y, Shamoto T, Shibata T, Koide S, Morimoto M, Guha S, Sung B, Aggarwal BB, Takahashi H, Takeyama H. Zerumbone inhibits tumor angiogenesis via NF-κB in gastric cancer. Oncol Rep. 2014; 31:57–64. PMID: 24220661.
Article
21. Arnaoutova I, George J, Kleinman HK, Benton G. The endothelial cell tube formation assay on basement membrane turns 20: state of the science and the art. Angiogenesis. 2009; 12:267–274. PMID: 19399631.
Article
22. Go RS, Owen WG. The rat aortic ring assay for in vitro study of angiogenesis. Methods Mol Med. 2003; 85:59–64. PMID: 12710197.
23. Shibuya M, Claesson-Welsh L. Signal transduction by VEGF receptors in regulation of angiogenesis and lymphangiogenesis. Exp Cell Res. 2006; 312:549–560. PMID: 16336962.
Article
24. Beenken A, Mohammadi M. The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov. 2009; 8:235–253. PMID: 19247306.
Article
25. Woad KJ, Hunter MG, Mann GE, Laird M, Hammond AJ, Robinson RS. Fibroblast growth factor 2 is a key determinant of vascular sprouting during bovine luteal angiogenesis. Reproduction. 2012; 143:35–43. PMID: 21998077.
Article
26. Carmeliet P. Angiogenesis in life, disease and medicine. Nature. 2005; 438:932–936. PMID: 16355210.
Article
27. Mittal K, Ebos J, Rini B. Angiogenesis and the tumor microenvironment: vascular endothelial growth factor and beyond. Semin Oncol. 2014; 41:235–251. PMID: 24787295.
Article
28. Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell. 2011; 146:873–887. PMID: 21925313.
Article
29. Ayers M, Fargnoli J, Lewin A, Wu Q, Platero JS. Discovery and validation of biomarkers that respond to treatment with brivanib alaninate, a small-molecule VEGFR-2/FGFR-1 antagonist. Cancer Res. 2007; 67:6899–6906. PMID: 17638901.
Article
30. Renhowe PA, Pecchi S, Shafer CM, Machajewski TD, Jazan EM, Taylor C, Antonios-McCrea W, McBride CM, Frazier K, Wiesmann M, Lapointe GR, Feucht PH, Warne RL, Heise CC, Menezes D, Aardalen K, Ye H, He M, Le V, Vora J, Jansen JM, Wernette-Hammond ME, Harris AL. Design, structure-activity relationships and in vivo characterization of 4-amino-3-benzimidazol-2-ylhydroquinolin-2-ones: a novel class of receptor tyrosine kinase inhibitors. J Med Chem. 2009; 52:278–292. PMID: 19113866.
Article
31. Bello E, Colella G, Scarlato V, Oliva P, Berndt A, Valbusa G, Serra SC, D'Incalci M, Cavalletti E, Giavazzi R, Damia G, Camboni G. E-3810 is a potent dual inhibitor of VEGFR and FGFR that exerts antitumor activity in multiple preclinical models. Cancer Res. 2011; 71:1396–1405. PMID: 21212416.
Article
Full Text Links
  • KJPP
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