Browse > Article
http://dx.doi.org/10.4014/jmb.1506.06073

Anticancer Effect of Thymol on AGS Human Gastric Carcinoma Cells  

Kang, Seo-Hee (Department of Biotechnology, Konkuk University)
Kim, Yon-Suk (Department of Biotechnology, Konkuk University)
Kim, Eun-Kyung (Division of Food Bioscience, Konkuk University)
Hwang, Jin-Woo (Department of Biotechnology, Konkuk University)
Jeong, Jae-Hyun (Department of Food Science and Technology, Korea National University of Transportation)
Dong, Xin (Department of Biotechnology, Konkuk University)
Lee, Jae-Woong (Department of Biotechnology, Konkuk University)
Moon, Sang-Ho (Nokyong Research Center, Konkuk University)
Jeon, Byong-Tae (Nokyong Research Center, Konkuk University)
Park, Pyo-Jam (Department of Biotechnology, Konkuk University)
Publication Information
Journal of Microbiology and Biotechnology / v.26, no.1, 2016 , pp. 28-37 More about this Journal
Abstract
Numerous plants have been documented to contain phenolic compounds. Thymol is one among these phenolic compounds that possess a repertoire of pharmacological activities, including anti-inflammatory, anticancer, antioxidant, antibacterial, and antimicrobial effects. Despite of the plethora of affects elicited by thymol, its activity profile on gastric cancer cells is not explored. In this study, we discovered that thymol exerts anticancer effects by suppressing cell growth, inducing apoptosis, producing intracellular reactive oxygen species, depolarizing mitochondrial membrane potential, and activating the proapoptotic mitochondrial proteins Bax, cysteine aspartases (caspases), and poly ADP ribose polymerase in human gastric AGS cells. The outcomes of this study displayed that thymol, via an intrinsic mitochondrial pathway, was responsible for inducing apoptosis in gastric AGS cells. Hence, thymol might serve as a tentative agent in the future to treat cancer.
Keywords
AGS human gastric carcinoma cells; anticancer; apoptosis; thymol;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Ali I, Wani WA, Saleem K, Haque A. 2013. Platinum compounds: a hope for future cancer chemotherapy. Anticancer Agents Med. Chem. 13: 296-306.   DOI
2 Al-Bandak G, Oreopoulou V. 2007. Antioxidant properties and composition of Majorana syriaca extracts. Eur. J. Lipid Sci. Technol. 109: 247-255.   DOI
3 Bellamy COC, Malcomson RDG, Harrison DJ, Wyllie AH. 1995. Cell death in health and disease: the biology and regulation of apoptosis. Semin. Cancer Biol. 6: 3-16.   DOI
4 Bhatia M. 2004. Apoptosis versus necrosis in acute pancreatitis. Am. J. Physiol. Gastrointest. 286:189-196.   DOI
5 Brana C, Benham C, Sundstrom L. 2002. A method for characterising cell death in vitro by combining propidium iodide staining with immunohistochemistry. Brain Res. Protoc. 10: 109-114.   DOI
6 Deb DD, Parimala G, Devi SS, Chakraborty T. 2011. Effect of thymol on peripheral blood mononuclear cell PBMC and acute promyelotic cancer cell line HL-60. Chem. Biol. Interact. 193: 97-106.   DOI
7 Devincenzi M, Stammati A, Silano M. 2004. Constituents of aromatic plants: carvacrol. Ritoterapia 75: 801-804.   DOI
8 Eivaz-Mohammadi S, Gonzalez-Ibarra F, Abdul W, Tarar O, Malik K, Syed AK. 2014. Her2+ and b-HCG producing undifferentiated gastric adenocarcinoma. Case Rep. Med. 2014: 268919-268923.
9 Jiang X, Wang X. 2004. Cytochrome c-mediated apoptosis. Annu. Rev. Biochem. 73: 87-106   DOI
10 Enari M, Sakahira H, Yokoyama H, Okawa K, Iwamatsu A, Nagata S. 1998. A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 391: 43-50.   DOI
11 Gerdes J, Lemke H, Baisch H, Wacker HH, Schwab U, Stein H. 1984. Cell cycle analysis of a cell proliferation-associated human nuclear antigen defined by the monoclonal antibody Ki-67. J. Immunol. 133: 1710-1715.
12 Jacobson MD. 1996. Reactive oxygen species and programmed cell death. Trends Biochem. Sci. 21: 83-86.   DOI
13 Kajstura M, Halicka HD, Pryjma J, Darzynkiewicz Z. 2007. Discontinuous fragmentation of nuclear DNA during apoptosis revealed by discrete “sub-G1” peaks on DNA content histograms. Int. Soc. Anal. Cytol. 71A: 125-131.   DOI
14 Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K, Tavtigian SV, et al. 1994. A cell cycle regulator potentially involved in genesis of many tumor types. Science 264: 436-440.   DOI
15 Kim YS, Hwang JW, Kang SH, Kim SH, Jeon YJ, Jeong JH, et al. 2014. Thymol from Thymus quinquecotatus Celak. protects against tert-butyl hydroperoxide-induced oxidative stress in Chang cells. J. Nat. Med. 68: 154-162.   DOI
16 Korsmeyer SJ, Wie MC, Saito M, Weiler S, Oh KJ, Schlesinger PH. 2000. Pro-apoptotic cascade activates BID, which oligomerizes BAK or BAX into pores that result in the release of cytochrome c. Cell Death Differ. 7: 1166-1173.   DOI
17 Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X. 1997. Cytochrome c and dATPdependent formation of Apaf-1/Caspase-9 complex initiates an apoptotic protease cascade. Cell 91: 479-489.   DOI
18 Leite de Oliveira R, Deschoemaeker S, Henze AT, Debackere K, Finisquerra V, Takeda Y, et al. 2012. Gene-targeting of Phd2 improves tumor response to chemotherapy and prevents side-toxicity. Cancer Cell 22: 263-277.   DOI
19 Lüpertz R, Wätjen W, Kahl R, Chovolou Y. 2010. Dose- and time-dependent effects of doxorubicin on cytotoxicity, cell cycle and apoptotic cell death in human colon cancer cells. Toxicology 271: 115-121   DOI
20 Li H, Zhy H, Xu CJ, Yuan J. 1998. Cleavage of BID by Caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 94: 491-501.   DOI
21 Llana-Ruiz-Cabello M, Maisanaba S, Puerto M, Prietom AI, Pichardo S, Jos A, Cameán AM. 2014. Evaluation of the mutagenicity and genotoxic potential of carvacrol and thymol using the Ames Salmonella test and alkaline, Endo III- and FPG-modified comet assays with the human cell line Caco-2. Food Chem. Toxicol. 72: 122-128.   DOI
22 Ly JD, Grubb DR, Lawen A. 2003. The mitochondrial membrane potential (øm) in apoptosis; an update. Apoptosis 8: 115-128.   DOI
23 Pelicano H, Carney D, Huang P. 2004. ROS stress in cancer cells and therapeutic implications. Drug Resist. Updat. 7: 97-110.   DOI
24 Rao RV, Ellerby HM, Bredesen DE. 2004. Coupling endoplasmic reticulum stress to the cell death program. Cell Death Differ. 11: 372-380.   DOI
25 Silke J, Vaux DI. 1998. Cell death: shadow boxing. Curr. Biol. 8: 528-531.   DOI
26 Rello S, Stockert JC, Moreno V, Gámez A, Pacheco M, Juarranz A, et al. 2005. Morphological criteria to distinguish cell death induced by apoptotic and necrotic treatments. Apoptosis 10: 201-208.   DOI
27 Schwartzman RA, Cidlowski JA. 1993. Apoptosis: the biochemistry and molecular biology of programmed cell death. Endocr. Rev. 14: 133-151.
28 Sun ZX, Zhang YH, Cheng S, Ma QW, Guo SL, Zhang JB. 2005. Anti-tumor effect of ethanol extracts from Thymus quinquecostatus Celak on human leukemia cell line. Zhong Xi Yi Jie He Xue Bao 3: 382-385.   DOI
29 Sobczak M, Kalemba D, Ferenc B, Zylinska L. 2014. Limited protective properties of thymol and thyme oil on differentiated PC12 cells with downregulated Mgst1. J. Appl. Biomed. 12: 235-243.   DOI
30 Su CC, Chen JYF, Din ZH, Su JH, Yang ZY, Chen YJ, et al. 2014. 13-Acetoxysarcocrassolide induces apoptosis on human gastric carcinoma cells through mitochondria-related apoptotic pathways: p38/JNK activat ion and PI3K/AKT suppression. Mar. Drugs 12: 5295-5315.   DOI
31 Trachootham D, Alexandre J, Huang P. 2009. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat. Rev. Drug Discov. 8: 579-591.   DOI
32 Xiong B, Ma L, Cheng Y, Zhang C. 2014. Clinical effectiveness of neoadjuvant chemotherapy in advanced gastric cancer: an updated meta-analysis of randomized controlled trials. Eur. J. Surg. Oncol. 40: 1321-1330.   DOI
33 Yin XM. 2000. Signal transduction mediated by Bid, a prodeath Bcl-2 family proteins, connects the death receptor and mitochondria apoptosis pathways. Cell Res. 10: 161-167.   DOI
34 Zou H, Li Y, Liu X, Wang X. 1999. An APAF-1·cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. J. Biol. Chem. 274: 11549-11556.   DOI
35 Zhang J, Jin HC, Zhu AK, Ying RC, Wei C, Zhang FJ. 2014. Prognostic significance of plasma chemerin levels in patients with gastric cancer. Peptides 61: 7-11.   DOI