Volume 9, Issue 3 (Vol.9 No.3 Oct 2020)                   rbmb.net 2020, 9(3): 338-347 | Back to browse issues page


XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Dashtaki A, Mahjoub S, Zabihi E, Pourbagher R. The Effects of Pre-Treatment and Post-Treatment of Thymol against tert-Butyl Hydroperoxide (t-BHP) Cytotoxicity in MCF-7 Cell Line and Fibroblast Derived Foreskin. rbmb.net 2020; 9 (3) :338-347
URL: http://rbmb.net/article-1-516-en.html
Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, I. R. Iran & Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, I. R. Iran.
Abstract:   (3202 Views)
Background: Some recent studies have reported anti-tumor activity for Thymol, but the findings are inconsistent. This study aimed to investigate and compare Thymol's effects on MCF-7 cancer cells and fibroblasts while treated with tert-Butyl hydroperoxide (t-BHP).

Methods: In the pre-treatment, MCF-7 and fibroblast cells were treated with various Thymol concentrations and incubated for 24 h. Then, t-BHP was added to a final concentration of 50 μM, and the cells were incubated for one h. In the post-treatment, cells were incubated first with 50 μM t-BHP for one h and then treated with Thymol. Cell viability was tested by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Thymol's antioxidant capacity was measured by DPPH and FRAP assays, and lipid peroxidation levels were determined by the TBARS method.

Results: The thymol effects were dose-dependent, and despite their antioxidant properties, at concentrations of 100 µg/ml or more, increased t-BHP toxicity and reduced cancer cell viability. MTT assay result showed that pre-treatment and post-treatment with Thymol for 24 hours effectively reduced MCF-7 and fibroblast cell viability compared with the untreated control group. Both pre- and post-treatment of Thymol, normal fibroblast cell viability was significantly greater than that of the MCF-7 cells.

Conclusions: Our finding showed that Thymol appears to be toxic to MCF-7 cells at lower concentrations than fibroblasts after 24 hours of incubation. Pre-treatment with Thymol neutralized the oxidative effect of t-BHP in fibroblasts but was toxic for MCF-7 cells.
Full-Text [PDF 269 kb]   (1611 Downloads)    
Type of Article: Original Article | Subject: Biochemistry
Received: 2020/06/18 | Accepted: 2020/06/21 | Published: 2020/12/1

References
1. Rojas K, Stuckey A. Breast cancer epidemiology and risk factors. Clin Obstet Gynecol. 2016;59(4):651-672. [DOI:10.1097/GRF.0000000000000239] [PMID]
2. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394-424. [DOI:10.3322/caac.21492] [PMID]
3. Nourazarian AR, Kangari P, Salmaninejad A. Roles of oxidative stress in the development and progression of breast cancer. Asian Pac J Cancer Prev. 2014;15(12):4745-51. [DOI:10.7314/APJCP.2014.15.12.4745] [PMID]
4. Gupta RK, Patel AK, Kumari R, Chugh S, Shrivastav C, Mehra S, et al. Interactions between oxidative stress, lipid profile and antioxidants in breast cancer: a case control study. Asian Pac J Cancer Prev. 2012;13(12):6295-8. [DOI:10.7314/APJCP.2012.13.12.6295] [PMID]
5. Vieira FGK, Pietro PFD, Boaventura BCB, Ambrosi C, Rockenbach G, Fausto MA, et al. Factors associated with oxidative stress in women with breast cancer. 2011.
6. Abazari O, Shafaei Z, Divsalar A, Eslami-Moghadam M, Ghalandari B, Saboury AA. Probing the biological evaluations of a new designed Pt (II) complex using spectroscopic and theoretical approaches: Human hemoglobin as a target. J Biomol Struct Dyn. 2016;34(5):1123-31. [DOI:10.1080/07391102.2015.1071280] [PMID]
7. Seresht HR, Albadry BJ, Al-mosawi AKM, Gholami O, Cheshomi H. The cytotoxic effects of Thymol as the major component of trachyspermum ammi on breast cancer (MCF-7) cells. Pharmaceutical Chemistry Journal. 2019;53(2):101-107. [DOI:10.1007/s11094-019-01961-w]
8. Deb DD, Parimala G, Devi SS, Chakraborty T. Effect of Thymol on peripheral blood mononuclear cell PBMC and acute promyelotic cancer cell line HL-60. Chem Biol Interact. 2011;193(1):97-106. [DOI:10.1016/j.cbi.2011.05.009] [PMID]
9. Islam MT, Khalipha AB, Bagchi R, Mondal M, Smrity SZ, Uddin SJ, et al. Anticancer activity of Thymol: A literature‐based review and docking study with Emphasis on its anticancer mechanisms. IUBMB life. 2019;71(1):9-19. [DOI:10.1002/iub.1935] [PMID]
10. Elbe H, Yigitturk G, Cavusoglu T, Uyanikgil Y, Ozturk F. Apoptotic effects of Thymol, a novel monoterpene phenol, on different types of cancer. Bratisl Lek Listy. 2020;121(2):122-128. [DOI:10.4149/BLL_2020_016] [PMID]
11. Abazari O, Divsalar A, Ghobadi R. Inhibitory effects of oxali-Platin as a chemotherapeutic drug on the function and structure of bovine liver catalase. J Biomol Struct Dyn. 2020;38(2):609-615. [DOI:10.1080/07391102.2019.1581088] [PMID]
12. Meeran MFN, Javed H, Al Taee H, Azimullah S, Ojha SK. Pharmacological properties and molecular mechanisms of Thymol: prospects for its therapeutic potential and pharmaceutical development. Front pharmacol. 2017;8:380. [DOI:10.3389/fphar.2017.00380] [PMID] [PMCID]
13. Li X, He T, Wang X, Shen M, Yan X, Fan S, et al. Traditional uses, chemical constituents and biological activities of plants from the genus Thymus. Chem Biodivers. 2019;16(9):e1900254. [DOI:10.1002/cbdv.201900254] [PMID]
14. Nabavi SM, Marchese A, Izadi M, Curti V, Daglia M, Nabavi SF. Plants belonging to the genus Thymus as antibacterial agents: From farm to pharmacy. Food chem. 2015;173:339-47. [DOI:10.1016/j.foodchem.2014.10.042] [PMID]
15. Asadi A, Nezhad DY, Javazm AR, Khanicheragh P, Mashouri L, Shakeri F, et al. In vitro Effects of Curcumin on Transforming Growth Factor-β-mediated Non-Smad Signaling Pathway, Oxidative Stress, and Pro‐inflammatory Cytokines Production with Human Vascular Smooth Muscle Cells. Iranian J Allergy Asthma and Immunol. 2019;19(1):84-93. [DOI:10.18502/ijaai.v19i1.2421] [PMID]
16. Kim Y-S, Hwang J-W, Kang S-H, Kim E-H, Jeon Y-J, Jeong J-H, et al. Thymol from Thymus quinquecostatus Celak. protects against tert-butyl hydroperoxide-induced oxidative stress in Chang cells. J Nat Med. 2014;68(1):154-62. [DOI:10.1007/s11418-013-0786-8] [PMID]
17. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65(1-2):55-63. [DOI:10.1016/0022-1759(83)90303-4]
18. Niehaus Jr W, Samuelsson B. Formation of malonaldehyde from phospholipid arachidonate during microsomal lipid peroxidation. Eur J Biochem. 1968;6(1):126-30. [DOI:10.1111/j.1432-1033.1968.tb00428.x] [PMID]
19. Adewoyin M, Ibrahim M, Roszaman R, Isa MLM, Alewi NAM, Rafa AAA, et al. Male infertility: the effect of natural antioxidants and phytocompounds on seminal oxidative stress. Diseases. 2017;5(1):9. [DOI:10.3390/diseases5010009] [PMID] [PMCID]
20. Ceriello A, Testa R, Genovese S. Clinical implications of oxidative stress and potential role of natural antioxidants in diabetic vascular complications. Nutr Metab Cardiovasc Dis. 2016;26(4):285-92. [DOI:10.1016/j.numecd.2016.01.006] [PMID]
21. Abazari O, Shafaei Z, Divsalar A, Eslami-Moghadam M, Ghalandari B, Saboury AA, et al. Interaction of the synthesized anticancer compound of the methyl-glycine 1, 10-phenanthroline platinum nitrate with human serum albumin and human hemoglobin proteins by spectroscopy methods and molecular docking. Journal of the Iranian Chemical Society. 2020:1601-1614. [DOI:10.1007/s13738-020-01879-1]
22. Wang X, Wu Q, Liu A, Anadón A, Rodríguez J-L, Martínez-Larrañaga M-R, et al. Paracetamol: overdose-induced oxidative stress toxicity, metabolism, and protective effects of in vivo various compounds and in vitro. Drug Metab Rev. 2017;49(4):395-437. [DOI:10.1080/03602532.2017.1354014] [PMID]
23. Wang N, Yi WJ, Tan L, Zhang JH, Xu J, Chen Y, et al. Apigenin attenuates streptozotocin-induced pancreatic β cell damage by its protective effects on cellular antioxidant defense. In Vitro Cell Dev Biol Anim. 2017;53(6):554-563. [DOI:10.1007/s11626-017-0135-4] [PMID]
24. Olson KR, Gao Y. Effects of inhibiting antioxidant pathways on cellular hydrogen sulfide and polysulfide metabolism. Free Radic Biol Med. 2019;135:1-14. [DOI:10.1016/j.freeradbiomed.2019.02.011] [PMID]
25. Aydın E, Turkez H, Tasdemir S, Hacımuftuoglu F. Anticancer, antioxidant and cytotoxic potential of Thymol in vitro brain tumor cell model. Cent Nerv Syst Agents in Med Chem. 2017;17(2):116-122. [DOI:10.2174/1871524916666160823121854] [PMID]
26. Li Y, Wen J-m, Du C-j, Hu S-m, Chen J-x, Zhang S-g, et al. Thymol inhibits bladder cancer cell proliferation via inducing cell cycle arrest and apoptosis. Biochem Biophys Res Commun. 2017;491(2):530-536. https://doi.org/10.1006/bbrc.1996.1541 https://doi.org/10.1016/j.bbrc.2017.04.009 [DOI:10.1016/j.bbrc.2018.09.124] [PMID]
27. Jamali T, Kavoosi G, Safavi M, Ardestani SK. In-vitro evaluation of apoptotic effect of OEO and Thymol in 2D and 3D cell cultures and the study of their interaction mode with DNA. Sci Rep. 2018;8(1):15787. [DOI:10.1038/s41598-018-34055-w] [PMID] [PMCID]
28. Abbasi M, Abazari OO. Probing the Biological evaluations of a new designed Palladium (II) complex using spectroscopic and theoretical approaches: Human Hemoglobin as a Target. Archives of Medical Laboratory Sciences. 2018;3(3).
29. Zheng C, An X, Yin T. New metal-free catalytic degradation systems with carbon dots for thymol blue. New Journal of Chemistry. 2017;41(22):13365-9. [DOI:10.1039/C7NJ02642K]
30. Qazi MA, Molvi KI. Free radicals and their management. American Journal of Pharmacy and Health Research. 2018;6(4):1-10. [DOI:10.46624/ajphr.2018.v6.i4.001]
31. Jafari A, Rasmi Y, Hajaghazadeh M, Karimipour M. Hepatoprotective effect of Thymol against subchronic toxicity of titanium dioxide nanoparticles: Biochemical and histological evidences. Environmental Toxicology and Pharmacology. 2018;58:29-36. [DOI:10.1016/j.etap.2017.12.010] [PMID]
32. Palabiyik S, Karakus E, Halici Z, Cadirci E, Bayir Y, Ayaz G, et al. The protective effects of carvacrol and Thymol against paracetamol-induced
33. toxicity on human hepatocellular carcinoma cell lines (HepG2). Hum Exp Toxicol. 2016;35(12):1252-1263. [DOI:10.1177/0960327115627688] [PMID]
34. Kang S-H, Kim Y-S, Kim E-K, Hwang J-W, Jeong J-H, Dong X, et al. Anticancer effect of Thymol on AGS human gastric carcinoma cells. J Microbiol Biotechnol. 2016;26(1):28-37. [DOI:10.4014/jmb.1506.06073] [PMID]
35. Günes-Bayir A, Kocyigit A, Kiziltan HS. Effects of Thymol, a natural phenolic compound, on human gastric adenocarcinoma cells In vitro. Alternative therapies in health and medicine. 2019;25(2):12-21.
36. Llana-Ruiz-Cabello M, Gutiérrez-Praena D, Pichardo S, Moreno FJ, Bermúdez JM, Aucejo S, et al. Cytotoxicity and morphological effects induced by carvacrol and thymol on the human cell line Caco-2. Food and Chemical Toxicology. 2014;64:281-290. [DOI:10.1016/j.fct.2013.12.005] [PMID]

Add your comments about this article : Your username or Email:
CAPTCHA

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2015 All Rights Reserved | Reports of Biochemistry and Molecular Biology

Designed & Developed by : Yektaweb