Allicin induces apoptosis through activation of both intrinsic and extrinsic pathways in glioma cells

  • Authors:
    • Chenlong Li
    • Hanguang Jing
    • Guangtao Ma
    • Peng Liang
  • View Affiliations

  • Published online on: February 2, 2018     https://doi.org/10.3892/mmr.2018.8552
  • Pages: 5976-5981
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Allicin is an extract purified from Allium sativum (garlic), and previous research has indicated that Allicin has an inhibitory effect on many kinds of tumor cells. The aim of the present study was to explore the anticancer activity of Allicin on human glioma cells and investigate the underlying mechanism. MTT and colony-formation assays were performed to detect glioma cell proliferation, and explore the effect of Allicin at various doses and time-points. The apoptosis of glioma cells was measured by fluorescence microscopy with Hoechst 33258 staining, and then flow cytometry was used to analyzed changes in glioma cell apoptosis. Reverse transcription‑quantitative polymerase chain reaction and western blot analysis were used to detect the effect of Allicin on the expression levels of Fas/Fas ligand (FasL), caspase‑3, B‑cell lymphoma 2 and Bcl‑2‑associated X protein. Allicin suppressed the proliferation and colony formation ability of U251 cells in a dose‑ and time‑dependent manner. A cytotoxic effect of Allicin was observed in glioma cells in a dose‑dependent manner. Changes in nuclear morphology were observed in U251 cells with Hoechst 33258 staining. The activity of caspases were significantly elevated and Fas/FasL expression levels were increased following treatment with Allicin, at both the mRNA and protein level. These results demonstrated that Allicin suppresses proliferation and induces glioma cell apoptosis in vitro. Both intrinsic mitochondrial and extrinsic Fas/FasL‑mediated pathways react in glioma cell after treating with Allicin, which then activate major apoptotic cascades. These results implicate Allicin as a novel antitumor agent in treating glioma.

Introduction

Glioblastoma (GBM) is the most aggressive subset of primary brain tumor in adults, and is responsible for ~50% of all cranial tumors. GBMs are highly infiltrative which results in difficulty for them to be resected completely (13). Comprehensive therapy including radiotherapy and chemotherapy is the main approach used for treatment; however, the overall survival of glioma patients is only 12–14 months post-diagnosis (4).

Allicin (Fig. 1A) is extracted from freshly crushed garlic (Allium sativum). The anti-bacterial and antiinflammatory effects of Allicin have been indicated, and previous studies have demonstrated the antitumor capacity which may inhibit tumor growth and induce apoptosis (57). Allicin has been demonstrated to have an inhibitory effect on different kinds of tumors including lung cancer, colorectal carcinoma, stomach cancer and liver cancer (8,9). Previous studies have further explored the effect of Allicin as an anticancer drug. However, the molecular mechanism underlying the apoptosis effect of Allicin in glioma remains to be clarified.

Activation of apoptosis signaling pathways may be involved in the treatment of malignant tumor. Two main apoptosis-associated signaling pathways have been addressed: The extrinsic death receptor pathway and the intrinsic mitochondrion pathway (10). The activation of Fas binding to its Fas ligand (FasL) could initiate the extrinsic pathway of apoptosis and serve a key role involved in death signaling in many cancer types (11). Both signaling pathways contain mitochondrial membrane and B-cell lymphoma 2 (Bcl-2) family proteins. Fas-associated protein with death domain (FADD) together with caspase-8 leads to autoproteolysis, and results in enzymatic activation of caspase-8 which in turn activates caspase-3, −6 and −7, resulting in the hydrolysis of cytosolic and substrates.

However, the roles of Allicin in human glioma remain largely unclear. The present study aimed to investigate the apoptotic activity of Allicin in human glioma cell lines, and explore the underlying mechanism. These results provide evidence that Allicin could inhibit proliferation and induce glioma cell apoptosis in vitro.

Materials and methods

Cell line and culture

The U251 human glioma cell line was obtained from the Chinese Academy of Sciences Cell Bank. Cells were cultured in Dulbecco's modified Eagle's medium (DMEM)/F12 (Hyclone; GE Healthcare Life Sciences, Chicago, IL, USA), supplemented with 10% (FBS) at 37°C with 5% CO2. Allicin (purity ≥90%) was purchased from Shanghai Harvest Pharmaceutical Co., Ltd. (Shanghai, China).

Cytotoxicity assay

Glioma cells were cultured at 5,000–8,000 cells/well in 96-well plates overnight. Cells were treated with 15, 30, 60 or 90 µg/ml of Allicin for 20 h. Subsequently, 5 mg/ml MTT (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was added for additional 4 h at 37°C. Optical density (OD) was assessed by using a TECAN microplate reader at 490 nm.

Cell proliferation assay and colony formation assay

Cell viability was measured by MTT assay. Cells were seeded at 5×103 cells/well into a 96-well plate and incubated with Allicin at various concentrations (15, 30, 60 and 90 µg/ml) for different periods (24, 48 and 72 h). The OD was assessed by using a TECAN microplate reader at 490 nm. For the colony formation assay, cells were cultured in a 6 cm dish (0.5×103 cells/well) and incubated at 37°C. Following this, different concentrations of Allicin were added for 24 h. The medium was then removed and cells were cultured for another 12 days. The colonies formed were fixed with 10% formalin for 10 min. Giemsa staining was used to stain the samples obtained for 30 min in room temperature and the number of colonies (>50 cells) was counted by using upright light microscope (Leica DM2000; Leica Microsystems, GmbH, Wetzlar, Germany).

Western blot analysis

U251 cells were treated with different concentrations Allicin (30 and 60 µg/ml) for 48 h, and then washed with ice-cold PBS. The cell lysates were centrifuged at 6,037 × g for 15 min at 4°C and the total protein was extracted using radioimmunoprecipitation buffer (Pierce; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with protease inhibitors, then subsequently separated via 4–10% Tris glycin/SDS-PAGE. A bicinchoninic acid protein assay kit was used to determine the concentration of protein. The total quantity of protein loaded onto each lane was 40 µg; separated proteins were electrotransferred to ECL nitrocellulose membranes (IPFL00010; EMD Millipore, Billerica, MA, USA). The membranes were blocked with 5% non-fat milk (Wuhan Boster Biological Technology, Ltd., Wuhan, China) at room temperature for 1 h and incubated separately with mouse anti-human Fas (1:1,000 dilution, cat. no. 8023S; Cell Signaling Technology, Inc., Danvers, MA, USA), mouse anti-human FasL (1:1,000 dilution, cat. no. 4273; Cell Signaling Technology, Inc.), mouse anti-human Bcl-2 (1:500 dilution, cat. no. sc-509; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), rabbit anti-human Bcl-2-associated X protein (Bax; 1:500 dilution, cat. no. sc-526; Santa Cruz Biotechnology, Inc.), rabbit anti-human caspase-3 (1:1,000 dilution, cat. no. 9665; Cell Signaling Technology, Inc.) and mouse anti-human β-actin (1:500 dilution, cat. no. 376421; Santa Cruz Biotechnology, Inc.) at 4°C overnight, and then incubated with a goat anti-rabbit horseradish peroxidase-conjugated secondary antibody (1:10,000 dilution, cat. no. ab150077; Abcam, Cambridge, MA, USA) for 1 h. The protein levels were visualized using an enhanced chemiluminescence kit (BeyoECL Plus, cat. no. P0018; Beyotime Institute of Biotechnology, Haimen, China) and determined by a Gel Doc 2000 imaging system (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total U251 cell RNA treated with different concentration of Allicin (0, 30 or 60 µg/ml) were isolated using TRIzol reagent (Thermo Fisher Scientific, Inc.). PCR was performed with ABI 7500 Real-Time PCR system (Applied Biosystems, Foster City, CA, USA). The thermocycling condition were as follows: 95°C for 30 sec, then followed by 40 cycles at 95°C for 5 sec, 60°C for 30 sec, and 72°C for 15 sec. The PCR primers were as follows: Fas sense, 5′-TTCTGCCATAAGCCCTGTC-3′ and antisense, 5′-TTGGTGTTGCTGGTGAGT-3′ (amplification fragment 320 bp); FasL sense, 5′-TTCAGCTCTTCCACCTACAG-3′ and antisense, 5′-ACATTCTCGGTGCCTGTAAC-3′ (amplification fragment 599 bp); caspase-3 sense, 5′-GACAGACAGTGGAAGCGACTGGAT-3′ and antisense, 5′-GCATGGCACAAAGCGACTGGAT-3′; Bcl-2 sense, 5′-CGCCCTGTGGATGACTGAGTA-3′ and antisense, 5′-GGGCCGTACAGTTCCACAAAG-3′; Bax sense, 5′-CCCTTTTGCTTCAGGGTTTCATCCA-3′ and antisense, 5′-CTTGAGACACTCGCTCAGCTTCTTG-3′; U6 sense, 5′-TGCGGGTGCTCGCTTCGGCAGC-3′ and antisense, 5′-CCAGTGCAGGGTCCGAGGT-3′. Fas/FasL, caspase-3, Bcl-2 and Bax mRNA expression levels were determined using a SYBR PrimeScript RT-PCR kit (Takara, Bio, Inc., Otsu, Japan) and normalized to U6 mRNA. The relative expression levels were analyzed using the 2−ΔΔCq method (12).

Detection of caspase activity

A caspase activity kit (Beyotime Institute of Biotechnology) was used to detected the activity of caspase-3, −8 and −9. Cells (1×106 cells/well) were added to Allicin (0, 30 and 60 µg/ml) for 24 h. Data was obtained by measuring the enzyme labeling meter with the wavelength of 490 nm via a microplate reader (Infinite F50 Tecan Group, Ltd., Männedorf, Switzerland). The U251 cells were isolated with caspase assay buffer (Nanjing Kaiji Materials, Co., Ltd., Nanjing, China) and then supernatant was collected and centrifuged at 7,546 × g at 4°C for 10 min. The relative activity of caspases was measuring by an enzyme-labeling meter with a microplate reader.

Hoechst 33258 staining

Cells were treated with Allicin for 24 h. The media was then removed and cells were fixed with 4% formaldehyde. Cells were then stained with 200 µM Hoechst 33258 for 10 min at room temperature, and the slides were examined under a fluorescence microscope (Olympus Corporation, Tokyo, Japan).

Cell apoptosis assay

U251 cells were seeded onto a 6-well plate at 5×105 cells/well for 24 h and treated with different amounts of Allicin (0, 30 and 60 µg/ml) for 48 h. The samples were collected at a concentration of 1×106 cells/ml with 4°C PBS and suspended in 400 µl binding buffer. Annexin V-fluorescein isothiocyanate (FITC) and propidium iodide (PI; BD Pharmingen; BD Biosciences, Franklin Lakes, NJ, USA) were added into the labeled tube and cells were incubated for 20 min. The samples were examined by using flow cytometry (BD Biosciences; Clontech, Palo Alto, CA, USA).

Statistical analysis

All results were summarized from three independent experiments. Results are expressed as the mean ± standard error. Comparisons of each treatment data with control were carried out for statistical difference by the paired t-test. One way analysis of variance with followed by a Turkey's post-hoc test was used to analyze statistical differences between groups by using the statistical software SPSS 17.0 (SPSS, Inc., Chicago, IL, USA); graphs were produced using by GraphPad Prism software (version 5.0; GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Cytotoxic effects of Allicin on U251 glioma cells

The cytotoxic effect of Allicin (P<0.05; Fig. 1B) on U251 cells was measured by MTT assay. The results demonstrated that Allicin decreased cell survival in a dose-dependent manner, indicating its cytotoxicity. The median IC50 was 41.97 µg/ml; therefore, 30 and 60 µg/ml doses of Allicin were chosen for the present study.

Allicin induces apoptosis of glioma cells

In the present study, Annexin V/PI staining was used to demonstrate the apoptosis-inducing effect of Allicin. The results demonstrated that the rate of apoptosis increased (30 µg/ml for 9.1±3.2% and 60 µg/ml for 51.4±3.8%) at higher Allicin concentrations compared with control cells (3.3±1.5%). Flow cytometry assay demonstrated that the rate of apoptosis increased in response to treatment with Allicin in a dose-dependent manner (P<0.05; Fig. 1C).

U251 cells were treated with Allicin for 24 h, then stained with Hoechst 33258. The morphological changes of apoptosis cells (such as condensation of chromatin and nuclear fragmentation) was detected under a fluorescence microscope (Fig. 2A). The level of apoptosis was increased with the dose of Allicin in U251 cells (Fig. 2B). These results demonstrated that Allicin induced apoptosis in a dose-dependent manner.

Allicin inhibits proliferation in U251 glioma cells

Colony formation was used to confirm the proliferation ability by treating with Allicin in U251 glioma cells, which indicated that the inhibition ability of Allicin was irreversible (Fig. 3A). MTT assay was used to detect the effect of Allicin on the growth of glioma cells. Allicin was demonstrated inhibit the viability of glioma cell in a dose- and time-dependent manner (Fig. 3B; P<0.05).

Allicin increases Fas and FasL expression in glioma cells

Fas (CD95/APO-1) induces apoptosis by binding to a member of the tumor necrosis factor receptor (TNFR) family (FasL), which is a cell surface receptor protein (13). RT-qPCR was used to measure the expression of Fas and FasL mRNA. The data demonstrated that after treating with different amounts of Alllicin, Fas, FasL and Bax mRNA expression levels were significantly increased, while Bcl-2 expression levels were decreased, in a dose-dependent manner (P<0.05, Fig. 4A). These results were reflected at the protein level (P<0.05; Fig. 4B and C). These results demonstrated that Allicin induces apoptosis a dose-dependent manner.

Allicin increases the activity of caspases

After treating cells with various amounts of Allicin for 24 h, caspase-3, −8 and −9 enzyme activities were upregulated (P<0.01; Fig. 4D). Therefore, apoptosis signaling pathways which were activated by caspases may be involved in the antitumor effects of Allicin.

Discussion

GBM is the most common and aggressive type of brain cancer, has an unfavorable prognosis and treatment still remains great challenge. The majority of chemotherapy options are not recommended because of various severe side effects, and the median survival time of the patients is extremely poor (14).

Garlic is the bulb of Allium plants, and Allicin (C6H10S3) is the main flavor compound. A previous study reported that Allicin supresses the growth of certain cancers, and may possess anticancer activity in vitro and in vivo (15). Allicin may induce cell apoptosis, cell cycle arrest and inhibit proliferation, but the underlying mechanisms remain unknown. A previous study demonstrated that Allicin can mediate the apoptosis of cancer cells by activating caspase-3, −8 and −9 (16). Allicin could activate autophagy of human liver cancer cells and induce cell death through apoptosis (7,17).

Apoptosis involves a series of biochemical processes of cell death controlled by several signaling pathways, such as the caspase and mitochondrial pathways. There are two main apoptotic pathways involved: The receptor-mediated pathway and the mitochondrial pathway, which is known as extrinsic and intrinsic. Allicin induces apoptosis by activating the extrinsic and intrinsic apoptosis pathways in gastric cancer cells (18).

The extrinsic pathway is initiated by stimulating the ligand of the death receptors on the cell surface, such as TNFR, Fas and FasL. Fas is a transmembrane protein binding to FasL, then trimerization and recruitment of FADD proteins occurs by activating caspase-8 and −10 (19). Caspase-8 regulates the expression of pro-caspase-3, −6, or −7. Caspase-3 serves an important role in cell death. In the present study, following Allicin treatment, the level of Fas and FasL increased in glioma cells, which has also been demonstrated in gastric cancer cells (20).

This leads to the activation of Bax, which belong to the Bcl-2 family, which leads to the release of cytochrome c (21). It has been reported that mitochondria are involved in inducing the intrinsic pathway, which is initiated via the release of signaling factors from the mitochondria. Cytochrome c is the first to be released into the cytosol, and interacts with pro-caspase-9, which could regulate caspase-3 or −7 (22).

The Bcl-2 family serves a pivotal a role in the intrinsic apoptosis pathway, which could maintain cell viability by inhibiting the capacity of Bax and impeding the release of cytochrome c (23). The results of the present study demonstrated that Allicin may inhibit the proliferation of U251 cells in vitro by suppressing Bcl-2 and inducing the expression of Bax.

In conclusion, the results of the present study indicated that Allicin can effectively inhibit proliferation and induce apoptosis in U251 glioma cells in vitro. It revealed that Allicin treatment increases the activation of both intrinsic and extrinsic apoptosis signaling pathways in U251 cells. Future studies should evaluate Allicin as a novel antitumor agent in treating glioma.

References

1 

Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P and Ellison DW: The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 131:803–820. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Bush NA, Chang SM and Berger MS: Current and future strategies for treatment of glioma. Neurosurg Rev. 14:1–14. 2017. View Article : Google Scholar

4 

Van Meir EG, Hadjipanayis CG, Norden AD, Shu HK, Wen PY and Olson JJ: Exciting new advances in neuro-oncology: The avenue to a cure for malignant glioma. CA Cancer J Clin. 60:166–193. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Zhou H, Qu Z, Mossine VV, Nknolise DL, Li J, Chen Z, Cheng J, Greenlief CM, Mawhinney TP, Brown PN, et al: Proteomic analysis of the effects of aged garlic extract and its FruArg component on lipopolysaccharide-induced neuroinflammatory response in microglial cells. PLoS One. 9:e1135312014. View Article : Google Scholar : PubMed/NCBI

6 

Lissiman E, Bhasale AL and Cohen M: Garlic for the common cold. Cochrane Database Syst Rev: CD006206. 2009. View Article : Google Scholar

7 

Chu YL, Ho CT, Chung JG, Rajasekaran R and Sheen LY: Allicin induces p53-mediated autophagy in HepG2 human liver cancer cells. J Agric Food Chem. 60:8363–8371. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Chu YL, Ho CT, Chung JG, Raghu R, Lo YC and Sheen LY: Allicin induces anti-human liver cancer cells through the p53 gene modulating apoptosis and autophagy. J Agric Food Chem. 61:9839–9848. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Zou X, Liang J, Sun J, Hu X, Lei L, Wu D and Liu L: Allicin sensitizes hepatocellular cancer cells to anti-tumor activity of 5-fluorouracil through ROS-mediated mitochondrial pathway. J Pharmacol Sci. 131:233–240. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Fuchs Y and Steller H: Programmed cell death in animal development and disease. Cell. 147:742–758. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Wang W, Zheng Z, Yu W, Lin H, Cui B and Cao F: Polymorphisms of the FAS and FASL genes and risk of breast cancer. Oncol Lett. 3:625–628. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Lapinski TW, Jaroszewicz J and Wiercinska-Drapalo A: Concentrations of soluble Fas and soluble Fas ligand as indicators of programmed cell death among patients coinfected with human immunodeficiency virus and hepatitis C virus. Viral Immunol. 19:570–575. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Shea A, Harish V, Afzal Z, Chijioke J, Kedir H, Dusmatova S, Roy A, Ramalinga M, Harris B, Blancato J, et al: MicroRNAs in glioblastoma multiforme pathogenesis and therapeutics. Cancer Med. 5:1917–1946. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Dvořáková M, Weingartová I, Nevoral J, Němeček D and Krejčová T: Garlic sulfur compounds suppress cancerogenesis and oxidative stress: A review. Scientia Agriculturae Bohemica. 46:65–72. 2015. View Article : Google Scholar

16 

Wang W, Du Z, Nimiya Y, Sukamtoh E, Kim D and Zhang G: Allicin inhibits lymphangiogenesis through suppressing activation of vascular endothelial growth factor (VEGF) receptor. J Nutr Biochem. 29:83–89. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Oommen S, Anto RJ, Srinivas G and Karunagaran D: Allicin (from garlic) induces caspase-mediated apoptosis in cancer cells. Eur J Pharmacol. 485:97–103. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Hebert DN, Lamriben L, Powers ET and Kelly JW: The intrinsic and extrinsic effects of N-linked glycans on glycoproteostasis. Nat Chem Biol. 10:902–910. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Li CL, Chang L, Guo L, Zhao D, Liu HB, Wang QS, Zhang P, Du WZ, Liu X, Zhang HT, et al: β-Elemene induces caspase-dependent apoptosis in human glioma cells in vitro through the upregulation of Bax and Fas/FasL and downregulation of Bcl-2. Asian Pac J Cancer Prev. 15:10407–10412. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Zhang W, Ha M, Gong Y, Xu Y, Dong N and Yuan Y: Allicin induces apoptosis in gastric cancer cells through activation of both extrinsic and intrinsic pathways. Oncol Rep. 24:1585–1592. 2010.PubMed/NCBI

21 

Liu Z, Ding Y, Ye N, Wild C, Chen H and Zhou J: Direct activation of Bax protein for cancer therapy. Med Res Rev. 36:313–341. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Jiang X and Wang X: Cytochrome c promotes caspase-9 activation by inducing nucleotide binding to Apaf-1. J Biol Chem. 275:31199–31203. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Du X, Zhao YP, Zhang TP, Zhou L, Chen G, Wang TX, You L and Shu H: Alteration of the intrinsic apoptosis pathway is involved in Notch-induced chemoresistance to gemcitabine in pancreatic cancer. Arch Med Res. 45:15–20. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 17 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li C, Jing H, Ma G and Liang P: Allicin induces apoptosis through activation of both intrinsic and extrinsic pathways in glioma cells. Mol Med Rep 17: 5976-5981, 2018
APA
Li, C., Jing, H., Ma, G., & Liang, P. (2018). Allicin induces apoptosis through activation of both intrinsic and extrinsic pathways in glioma cells. Molecular Medicine Reports, 17, 5976-5981. https://doi.org/10.3892/mmr.2018.8552
MLA
Li, C., Jing, H., Ma, G., Liang, P."Allicin induces apoptosis through activation of both intrinsic and extrinsic pathways in glioma cells". Molecular Medicine Reports 17.4 (2018): 5976-5981.
Chicago
Li, C., Jing, H., Ma, G., Liang, P."Allicin induces apoptosis through activation of both intrinsic and extrinsic pathways in glioma cells". Molecular Medicine Reports 17, no. 4 (2018): 5976-5981. https://doi.org/10.3892/mmr.2018.8552