Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter June 1, 2021

In vitro chemotherapeutic and antiangiogenic properties of cardenolides from Acokanthera oblongifolia (Hochst.) Codd

  • Maha M. Soltan ORCID logo EMAIL logo , Howaida I. Abd-Alla ORCID logo , Amal Z. Hassan and Atef G. Hanna

Abstract

Acovenoside A (Acov-A) and acobioside A (Acob-A) were isolated from Acokanthera oblongifolia. Their anticancer properties were explored regarding, antiproliferative and antiangiogenic activities. The study included screening phase against six cancer cell lines followed by mechanistic investigation against HepG2 cancer cell line. The sulforhodamine-B (SRB) was used to determine their growth inhibitory power. In the other hand, flow cytometry techniques were recorded the cell death type and cell cycle analysis. The clonogenic (colony formation) and wound healing assays, enzyme-linked immunosorbent assay (ELISA) and molecular docking, were performed to evaluate the antiangiogenesis capability. Both compounds were strongly, inhibited four cancer cell lines at GI50 less than 100 nM. The in vitro mechanistic investigation against HepG2 resulted in cell accumulations at G2M phase and induction of apoptosis upon treating cells separately, with 400 nM Acov-A and 200 nM Acob-A. Interestingly, the same concentrations were able to activate caspase-3 by 7.2 and 4.8-fold, respectively. Suppressing the clonogenic capacity of HepG2 cells (20 and 40 nM) and inhibiting the migration of the colon Caco-2 cancer cells were provoke the results of vascular endothelial growth factor receptor2 (VEGFR2) kinase enzyme inactivation. The docked study was highly supportive, to the antiangiogenic approach of both cardenolides. The isolated cardenolides could orchestrate pivotal events in fighting cancer.


Corresponding author: Maha M. Soltan, Biology Unit, Central Laboratory for Pharmaceutical and Drug Industries Research Division, Chemistry of Medicinal Plants Department, National Research Centre, El Buhouth St. 33Dokki-Giza12622, Egypt, E-mail:

Funding source: National Research Centre

Acknowledgement

We would like to thank our institute; National Research Centre, Egypt for the laboratory facilities and partial financial support.

  1. Author contribution: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: The study received partial financial support from the National Research Centre.

  3. Conflict of interest statement: The authors declare no conflict of interest.

References

1. Shoemaker, RH. The NCI60 human tumour cell line anti-cancer drug screen. Nat Rev Canc 2006;6:813–23. https://doi.org/10.1038/nrc1951.Search in Google Scholar

2. Hosseini, A, Ghorbani, A. Cancer therapy with phytochemicals: evidence from clinical studies. Avicenna J Phytomed 2015;5:84–97.Search in Google Scholar

3. Bethesda, MD. Biological sciences curriculum study. Understanding Cancer. In: NIH Curriculum Supplement Series; 2007. https://www.ncbi.nlm.nih.gov/books/NBK20362/.Search in Google Scholar

4. Adair, TH, Montani, JP. Overview of angiogenesis. San Rafael (CA): Morgan and Claypool Life Sciences; 2010. https://www.ncbi.nlm.nih.gov/books/NBK53242/.Search in Google Scholar

5. Hock, SW, Fan, Z, Buchfelder, M, Eyüpoglu, IY, Savaskan, NE. Brain tumor-induced angiogenesis: approaches and bioassays. In: Lichtor, T, editor Evolution of the molecular biology of brain tumors and the therapeutic implications. InTech; 2013. https://doi.org/10.5772/53182.Search in Google Scholar

6. Ou, JM, Yu, ZY, Qiu, MK, Dai, YX, Dong, Q, Shen, J, et al.. Knockdown of VEGFR2 inhibits proliferation and induces apoptosis in hemangioma-derived endothelial cells. Eur J Histochem 2014;58:2263. https://doi.org/10.4081/ejh.2014.2263.Search in Google Scholar

7. Ucuzian, AA, Gassman, AA, East, AT, Greisler, HP. Molecular mediators of angiogenesis. J Burn Care Res 2010;31:158–75. https://doi.org/10.1097/bcr.0b013e3181c7ed82.Search in Google Scholar

8. Fallah, A, Sadeghinia, A, Kahroba, H, Samadi, A, Heidari, HR, Bradaran, B, et al.. Therapeutic targeting of angiogenesis molecular pathways in angiogenesis-dependent diseases. Biomed Pharmacother 2019;110:775–85. https://doi.org/10.1016/j.biopha.2018.12.022.Search in Google Scholar

9. Shaaban, M, Yassin, FY, Soltan, MM. Calamusins J-K: new anti-angiogenic sesquiterpenes from Sarcophyton glaucum. Nat Prod Res 2020. in press. https://doi.org/10.1080/14786419.2020.1828404.Search in Google Scholar

10. Abuelizz, HA, Marzouk, M, Bakheit, AH, Awad, HM, Soltan, MM, Naglah, AM, et al.. Anti-proliferative and anti-angiogenic properties of new VEGFR-2-targeting 2-thioxo-benzo[g]quinazoline derivatives. Molecules 2020;25:5944. https://doi.org/10.3390/molecules25245944.Search in Google Scholar

11. Hassan, AZ, Mekhael, MKG, Hanna, AG, Simon, A, Toth, G, Duddeck, H. Phytochemical investigation of Corchorus olitorius and Corchorus capsularis (family Tiliaceae) that grow in Egypt. Egypt Pharm J 2019;18:123–34.10.4103/epj.epj_51_18Search in Google Scholar

12. Schneider, N, Cerella, C, Simões, CMO, Diederich, M. Anticancer and immunogenic properties of cardiac glycosides. Molecules 2017;22:1932. https://doi.org/10.3390/molecules22111932.Search in Google Scholar

13. Cerella, C, Dicato, M, Diederich, M. Assembling the puzzle of anti-cancer mechanisms triggered by cardiac glycosides. Mitochondrion 2013;13:225–34. https://doi.org/10.1016/j.mito.2012.06.003.Search in Google Scholar

14. López-Lázaro, M. Digitoxin as an anti-cancer agent with selectivity for cancer cells: possible mechanisms involved. Expert Opin Ther Targets 2007;11:1043–53. https://doi.org/10.1517/14728222.11.8.1043.Search in Google Scholar

15. Haux, J. Digitoxin is a potential anti-cancer agent for several types of cancer. Med Hypotheses 1999;53:543–8. https://doi.org/10.1054/mehy.1999.0985.Search in Google Scholar

16. Newman, RA, Yang, P, Pawlus, AD, Block, KI. Cardiac glycosides as novel cancer therapeutic agents. Mol Interv 2008;8:36–49. https://doi.org/10.1124/mi.8.1.8.Search in Google Scholar

17. Prassas, I, Diamandis, EP. Novel therapeutic applications of cardiac glycosides. Nat Rev Drug Discov 2008;7:926–35. https://doi.org/10.1038/nrd2682.Search in Google Scholar

18. Mijatovic, T, Dufrasne, F, Kiss, R. Cardiotonic steroids-mediated targeting of the Na+/K+-ATPase to combat chemoresistant cancers. Curr Med Chem 2012;19:627–46. https://doi.org/10.2174/092986712798992075.Search in Google Scholar

19. Sabry, OMM, El Sayed, AM, Ezzat, SM, Yousef, Z. Bioactive compounds from Acokanthera oblongifolia. Adv Life Sci Technol 2016;46:86–93.Search in Google Scholar

20. Pecio, L, Hassan, EM, Omer, EA, Gajek, G, Kontek, R, Sobieraj, A, et al.. Cytotoxic cardenolides from the leaves of Acokanthera oblongifolia. Planta Med 2019;85:965–72. https://doi.org/10.1055/a-0958-2566.Search in Google Scholar

21. Pieri, F, Arnould-Guerin, ML, Sefraoni, EH. Cardiotonic glycosides from Acokanthera spectabilis. Fitoterapia 1992;63:333–6.Search in Google Scholar

22. El-Gaafary, M, Ezzat, SM, El Sayed, AM, Sabry, OM, Hafner, S, Lang, S, et al.. Acovenoside A induces mitotic catastrophe followed by apoptosis in non-small-cell lung cancer cells. J Nat Prod 2017;80:3203–10. https://doi.org/10.1021/acs.jnatprod.7b00546.Search in Google Scholar

23. Abdelaziz, AM, Yu, M, Li, P, Zhong, L, Singab, ANB, Hanna, AG, et al.. Synthesis and evaluation of 5-chloro-2-methoxy-N-(4-sulphamoylphenyl) benzamide derivatives as anti-cancer agents. Med Chem 2015;5:253–60.10.4172/2161-0444.1000272Search in Google Scholar

24. Galal, AMF, Soltan, MM, Esam, AR, Hanna, AG. Synthesis and biological evaluation of novel 5-chloro-N-(4-sulfamoylbenzyl) salicylamide derivatives as tubulin polymerization inhibitors. MedChemComm 2018;9:1511–28. https://doi.org/10.1039/c8md00214b.Search in Google Scholar

25. Ediriweera, MK, Tennekoon, KH, Samarakoon, SR. In vitro assays and techniques utilized in anticancer drug discovery. J Appl Toxicol 2019;39:38–71. https://doi.org/10.1002/jat.3658.Search in Google Scholar

26. Iyer, AKV, Zhou, M, Azad, N, Elbaz, H, Wang, L, Rogalsky, DK, et al.. A direct comparison of the anticancer activities of digitoxin MeON-Neoglycosides and O-glycosides: oligosaccharide chain length-dependent induction of caspase-9-mediated apoptosis. ACS Med Chem Lett 2010;1:326–30. https://doi.org/10.1021/ml1000933.Search in Google Scholar

27. Al-Abd, AM, Alamoudi, AJ, Abdel-Naim, AB, Neamatallah, TA, Ashour, OM. Anti-angiogenic agents for the treatment of solid tumors: potential pathways, therapy and current strategies - a review. J Adv Res 2017;8:591–605. https://doi.org/10.1016/j.jare.2017.06.006.Search in Google Scholar

28. Folkman, JN. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971;285:1182–6. https://doi.org/10.1056/nejm197108122850711.Search in Google Scholar

29. Eke, I, Leonhardt, F, Storch, K, Hehlgans, S, Cordes, N. The small molecule inhibitor QLT0267 Radiosensitizes squamous cell carcinoma cells of the head and neck. PloS One 2009;4: e6434. https://doi.org/10.1371/journal.pone.0006434.Search in Google Scholar

30. Rafehi, H, Orlowski, C, Georgiadis, GT, Ververis, K, El-Osta, A, Karagiannis, TC. Clonogenic assay: adherent cells. JoVE 2011;49:2573–5. https://doi.org/10.3791/2573.Search in Google Scholar

31. Abd-Alla, HI, Soltan, MM, Hassan, AZ, Taie, HAA, Abo-Salem, HM, Karam, EA, et al.. Cardenolides and pentacyclic triterpenes isolated from Acokanthera oblongifolia leaves: their biological activities with molecular docking study. Z Naturforsch 2020;76c. in press. https://doi.org/10.1515/znc-2020-0198.Search in Google Scholar

32. Ezzat, SM, El-Gaafary, M, El Sayed, AM, Sabry, OM, Ali, ZY, Hafner, S, et al.. The cardenolide glycoside acovenoside A affords protective activity in doxorubicin-induced cardiotoxicity in mice. J Pharmacol Exp Therapeut 2016;358:262–70. https://doi.org/10.1124/jpet.116.232652.Search in Google Scholar

33. Shibuya, M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes Cancer 2011;2:1097–105. https://doi.org/10.1177/1947601911423031.Search in Google Scholar

34. Trott, O, Olson, AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J Comput Chem 2010;31:455–61. https://doi.org/10.1002/jcc.21334.Search in Google Scholar

35. Skehan, P, Storeng, R, Scudiero, D, Monks, A, McMahon, J, Vistica, D, et al.. New colorimetric cytotoxicity assay for anti-cancer-drug screening. J Natl Cancer Inst 1990;82:1107–12. https://doi.org/10.1093/jnci/82.13.1107.Search in Google Scholar

36. Vichai, V, Kirtikara, K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat Protoc 2006;1:1112–6. https://doi.org/10.1038/nprot.2006.179.Search in Google Scholar

37. Zhang, W, Wang, Y, Du, F, He, M, Gu, Y, Bai, L, et al.. Evaluation of anti-cancer effect in vitro and in vivo of iridium(III) complexes on gastric carcinoma SGC-7901 cells. Eur J Med Chem 2019;178:401–16. https://doi.org/10.1016/j.ejmech.2019.06.003.Search in Google Scholar

38. Liang, C, Park, A, Guan, J. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc 2007;2:329–33. https://doi.org/10.1038/nprot.2007.30.Search in Google Scholar

39. Barua, A, Yellapa, A, Bahr, JM, Machado, SA, Bitterman, P, Basu, S, et al.. VEGFR2-targeted ultrasound imaging agent enhances the detection of ovarian tumors at early stage in laying hens, a preclinical model of spontaneous ovarian cancer. Ultrason Imag 2015;37:224–37. https://doi.org/10.1177/0161734614553603.Search in Google Scholar

Received: 2020-12-19
Accepted: 2021-05-08
Published Online: 2021-06-01
Published in Print: 2021-07-27

© 2021 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 12.5.2024 from https://www.degruyter.com/document/doi/10.1515/znc-2020-0302/html
Scroll to top button