Apigenin inhibits HeLa sphere‑forming cells through inactivation of casein kinase 2α

  • Authors:
    • Jie Liu
    • Xiao‑Cheng Cao
    • Qiao Xiao
    • Mei‑Fang Quan
  • View Affiliations

  • Published online on: October 21, 2014     https://doi.org/10.3892/mmr.2014.2720
  • Pages: 665-669
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The protein kinase casein kinase 2 (CK2) has been implicated in stem cell maintenance and its aberrant activation has been demonstrated in several types of cancer, including cervical cancer. In the present study, it was demonstrated that the sphere‑forming cells (SFCs) of HeLa cell lines exhibited self‑renewal capacity, indicating that they possessed the properties of cervical cancer stem‑like cells. HeLa‑derived SFCs exhibited a higher level of CK2α protein, compared with the parental cells. Apigenin, a dietary flavonoid, led to a dose‑dependent inhibition of the self‑renewal capacity and the protein expression of CK2α in HeLa‑derived SFCs. Furthermore, forced overexpression of CK2α resulted in a decrease in the inhibition of CK2α expression and the self‑renewal capacity induced by apigenin in HeLa‑derived SFCs. These results suggested that apigenin inhibits the self‑renewal capacity of HeLa‑derived SFCs through downregulation of CK2α expression.

Introduction

Several studies have demonstrated that flavonoids are preventive in cancer (1,2). The most common nonmutagenic flavonoid, 4′,5,7-trihydroxyflavone (apigenin), has demonstrated marked effects in inhibiting cancer cell growth in vitro and in vivo (24). Apigenin has also been found to possess anti-inflammatory and antioxidant properties (5,6) and inhibit tumor cell invasion, metastasis (7), mitogen-activated protein kinases and downstream oncogenes (8). Our previous study demonstrated that apigenin was able to affect the number of glioma stem-like cells (GSLCs) derived from U251 cells (9) However, the effect of apigenin on the self-renewal capacity of cervical cancer stem-like cells (CCSLCs) remains to be elucidated.

The protein kinase casein kinase 2 (CK2) is a highly conserved serine/threonine kinase with a broad spectrum of substrates. CK2 is a multifunctional protein kinase that has been demonstrated to be involved in almost every aspect of cell proliferation and survival (1013). The expression and activity of CK2 are frequently elevated in cancer cells, including cervical cancer (14,15). Previous studies by Zhang et al demonstrated that the function of CK2α is involved in the activation of Hedgehog (Hh) and Notch pathways and in the maintenance of cancer stem cell properties (16,17). Whether or not targeting CK2 by the selective CK2 kinase inhibitor apigenin leads to self-renewal inhibition of cervical cancer stem-like cells remains to be elucidated.

The present study was performed to examine whether apigenin inhinited the self-reneweal capactiy of sphere-forming cells (SFCs) of the cervical cancer HeLA cell line and its underlying mechanisms, which aimed to assess the possibility for its use in the treatment of human cervical cancer by targeting cancer stem cells.

Materials and methods

Reagents

Apigenin was obtained from Sigma (St. Louis, MO, USA) and was dissolved in dimethyl sulfoxide to a final concentration of 0.1% in media without causing cytotoxicity. Anti-β-actin and CK2α antibodies were obtained from Calbiochem (La Jolla, CA, USA).

Cell culture

The HeLa human cervical cell line was maintained in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Carslbad, CA, USA) supplemented with 10% fetal bovine serum (Gibco), 100 units/ml penicillin (Gibco) and 100 μg/ml streptomycin (Gibco). In all experiments, the cells were maintained at 37°C, in a 5% CO2 and 95% air atmosphere. All the experiments were performed on cultures that were 70% confluent. The present study was approved by the ethics committee of Hunan Normal University (Changsha, China).

Tumorsphere culture

Single cell suspensions were suspended at a density of 5,000 cells/ml in serum-free DMEM/F12 supplemented with 100 IU/ml penicillin, 100 μg/ml streptomycin, 20 ng/ml human recombinant epidermal growth factor, 10 ng/ml human recombinant basic fibroblast growth factor, 2% B27 supplement without vitamin A and 1% N2 supplement (Invitrogen Life Technologies, Carlsbad, CA, USA) and seeded into ultra low attachment 6-well plates (Corning Inc., Corning, NY, USA). Suspension cultures were continued for 6 days until tumorspheres were formed. In order to propagate spheres in vitro, the sphere cells were collected by centrifugation (1,000 g for 5 min), dissociated into single-cell suspensions and cultured to permit the regeneration of spheres. Third-generation spheres were used for all subsequent experiments.

To investigate the percentage of single cells capable of regenerating new spheres, cells were plated at a density of 1,000 cells/ml in a 6-well plate in order to obtain new spheres. The total number of tumor spheres was counted after 6 days of culture. Sphere formation efficiency was calculated using the following formula: (Total number of spheres formed / total number of live cells seeded) × 100.

Limiting dilution analysis

The third-generation spheres were dissociated, as described above, and 100 cells were plated in 150 μl of growth medium in a 96-well culture plate to obtain a single cell per well. Growth medium (20 μl) was added to each well every 3 days. The number of clonal tumor spheres in each 96-well culture plate was evaluated after 6 days of culture.

MTT assay

SFCs from the HeLa cell line and the parental cells were seeded in 96-well plates precoated with Matrigel (Gibco-BRL) at a density of 5,000 cells per well. Cells were exposed to increasing concentrations (10, 20 and 40 μmol/l) of apigenin. After 48 h, MTT reagent (Sigma) was added to each well according to the manufacturer’s instructions. Absorbance was measured at 570 nm using an automated microplate reader (Bio-RAD 550; Bio-Rad, Hercules, CA, USA).

Overexpression of the CK2α protein

The pcDNA3.1-CK2α or control pcDNA3.1-LacZ plasmid vectors were transfected into HeLa cells or the SFCs of HeLa cells (0.5 μg/ml in a 24-well plate) using Lipofectamine 2000 transfection reagent (Invitrogen Life Technologies), according to the manufacturer’s instructions. The cells were resuspended in complete medium (DMEM supplemented with 10% fetal bovine serum; Gibo-BRL) for 48 h. The cells were harvested and western blotting was performed using mouse monoclonal antibodies against βactin and CK2α.

Western blot analysis

Western blot analysis was performed, as previously described (18). Cells were lysed by incubating in lysis buffer for 20 min at 4°C. The protein concentration was determined using the Bio-Rad assay system (Bio-Rad, Hercules, CA, USA). Total proteins were fractionated using SDS-PAGE and transferred onto a polyvinylidene fluoride membrane (Millipore, Billerica, MA, USA). Signals were detected using an ECL Advance western blot analysis system (Amersham Pharmacia Biotech, Inc., Piscataway, NJ, USA).

Statistical analysis

Statistical analysis and database management was performed using SPSS (version 15.0) software (SPSS, Inc., Chicago, IL, USA). Data are expressed as the mean ± standard deviation. Multiple group comparisons were made using one-way analysis of variance and pairwise comparison was performed using the least squares difference method. P<0.05 was considered to indicate a statistically significant difference.

Results

Sphere formation and self-renewal in the HeLa cell line

HeLa cells were plated in stem cell-conditioned culture medium in 6-well plates at a density of 10,000 cells/well, which enabled the formation of discrete colonies. Under these conditions, cells grew as non-adherent, three-dimensional sphere clusters. The HeLa cella maintained in DMEM medium supplemented with 10% fetal bovine serum (monolayer adhere growth cells) and anchorage-independent spheres formed in the HeLa cell line are shown in Fig. 1A and Fig 1B, respectively. The spheres were passaged after 6 days once they had reached ~50 μm in diameter. The HeLa sphere-derived cells were serially passaged for >12 generations, indicating that HeLa sphere-derived cells were fully capable of self-renewal in vitro.

To determine the percentage of CCSLCs in HeLa third-generation spheres, a limiting dilution assay was used to examine the ability of single cells from third-generation spheres to produce new spheres. After 6 days of culture, 31% of the single cells had generated new spheres (Fig. 1C). By contrast, a lower percentage of single cells derived from the HeLa cells could regenerate spheres when compared with the single cells derived from third-generation spheres (Fig. 1D). These results demonstrated that a considerable percentage of single cells derived from third-generation spheres were self-renewing cells that were able to be expanded and maintained in culture as tumor spheres.

Apigenin inhibits the proliferation and self-renewal capacity of the HeLa sphere-derived cells

It has been reported that cancer stem cells have the characteristics of extensive proliferation and apigenin has been demonstrated to inhibit the proliferative activity of glioma cancer stem-like cells (9). In the present study, the MTT results demonstrated that apigenin (10, 20 and 40 μmol/l) preferentially inhibited the proliferation of SFCs derived from HeLa cells (Fig. 2A), suggesting that apigenin is able to preferentially suppress the proliferative ability of CCSLCs.

Apigenin (10, 20 and 40 μmol/l) reduced the number of spheroids formed in the SFCs of HeLa cells in a concentration-dependent manner (Fig. 2B). These results suggest that apigenin can suppress the self-renewal capacity of CCSLCs.

Apigenin downregulates the protein expression of CK2α in HeLa sphere-derived cells

Previous studies have reported that CK2α is involved in the activation of the Hh and Notch pathways and is associated with the maintenance of cancer stem cell properties (16,17). The present study aimed to compare the status of CK2α protein expression in parental cells and in SFCs. The results demonstrated that the expression of CK2α was higher in the SFCs than in the parental cells (Fig. 3A). In addition, CK2α expression in SFCs was downregulated by apigenin (Fig. 3B).

Overexpression of CK2α attenuates the inhibitory effects of apigenin on the self-renewal capacity of HeLa sphere-derived cells

Western blot analysis demonstrated that upregulation of CK2α by pcDNA3.1-CK2α transfection resulted in overexpression of the CK2α protein in the parental HeLa cells (Fig. 4A). The results from the tumorsphere formation assay revealed that overexpression of CK2α attenuated apigenin-induced downregulation of CK2α protein expression in the HeLa SFCs (Fig. 4B) and also revealed that overexpression of the CK2α protein increased the self-renewal capacity of HeLa cells (Fig. 4C). In addition, . It also partially reduced the inhibition of self-renewal of SFCs by apigenin (Fig. 4D). These results provided mechanistic evidence that apigenin-inhibited self-renewal was, in part, due to inactivation of CK2α in SFCs derived from HeLa cells.

Discussion

The present study demonstrated that a natural dietary flavonoid, apigenin, inhibited the proliferation and self-renewal capacity of HeLa sphere-derived cells and inhibited the protein expression of CK2α. The inhibition of the CK2-mediated self-renewal capacity in HeLa sphere-derived cells by apigenin may be the primary mechanism mediating the anticancer activities of apigenin.

Previous studies have reported that the function of CK2α is involved in the activation of Hh and Notch pathways and in the maintenance of cancer stem cell properties (16,17). In order to investigate the potential mechanism through which CK2 positively regulates the self-renewal capacity of CCSLSCs, the present study performed a tumorsphere formation assay in the parental HeLa cells following transfection with pcDNA3.1-CK2α. The results indicated that forced overexpression of CK2α resulted in an increase in sphere formation rate in HeLa cells. Furthermore, apigenin, a small molecule CK2α inhibitor, significantly inhibited sphere formation of HeLa sphere-derived cells. Taken together, these results suggest that CK2α is important in facilitating the self-renewal capacity of CCSLCs. Further investigation is required to elucidate the precise mechanisms.

Zhao et al (19) found that apigenin inhibits proliferation and induces apoptosis in human multiple myeloma cells through the regulation of CK2, Cdc37 and Hsp90 expression. Feliciano et al (20) indicated that miR-125b is inversely correlated with glutamyl aminopeptidase and CK2α expression in breast tumorigenesis and that CK2α overexpression is associated with the presence and number of lymph node metastases. Our previous study demonstrated that apigenin preferentially inhibited the proliferation of GSLCs derived from U251 cells (9). The results of the present study demonstrated that apigenin significantly inhibited the proliferation and tumorsphere formation of HeLa sphere-derived cells. Furthermore, forced overexpression of CK2α effectively attenuated the apigenin inhibited tumorsphere formation of HeLa sphere-derived cells. These results suggested that apigenin-inhibited self-renewal is partly due to the inactivation of CK2α in SFCs derived from HeLa cells.

In conclusion, the present study demonstrated that CK2 is a positive regulator in the self-renewal of CCSLCs and apigenin, inhibiting the self-renewal capability, is involved in the downregulation of CK2α protein expression. The findings of the present study provide important evidence for the potential benefits of CK2 inhibitors in the treatment of human cervical cancer by targeting cancer stem cells.

Acknowledgements

The authors would like to thank Professor Jian-Guo Cao and Dr Xi-Yun Deng, Medical College, Hunan Normal University, (Changsha, Hunan, China) for their input into the scientific content of this study. The present study was supported by the Construct Program of the Key Discipline of Basic Medicine in Hunan.

References

1 

Birt DF, Hendrich S and Wang W: Dietary agents in cancer prevention: flavonoids and isoflavonoids. Pharmacol Ther. 90:157–177. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Benavente-García O and Castillo J: Update on uses and properties of citrus flavonoids: new findings in anticancer, cardiovascular, and anti-inflammatory activity. J Agric Food Chem. 56:6185–6205. 2008.PubMed/NCBI

3 

Arango D, Morohashi K, Yilmaz A, Kuramochi K, Parihar A, Brahimaj B, Grotewold E and Doseff AI: Molecular basis for the action of a dietary flavonoid revealed by the comprehensive identification of apigenin human targets. Proc Natl Acad Sci USA. 110:e2153–e2162. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Oishi M, Iizumi Y, Taniguchi T, Goi W, Miki T and Sakai T: Apigenin sensitizes prostate cancer cells to Apo2L/TRAIL by targeting adenine nucleotide translocase-2. PLoS One. 8:e559222013. View Article : Google Scholar : PubMed/NCBI

5 

Pham H, Chen M, Takahashi H, King J, Reber HA, Hines OJ, Pandol S and Eibl G: Apigenin inhibits NNK-induced focal adhesion kinase activation in pancreatic cancer cells. Pancreas. 41:1306–1315. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Kim HK, Cheon BS, Kim YH, Kim SY and Kim HP: Effects of naturally occurring flavonoids on nitric oxide production in the macrophage cell line RAW 264.7 and their structure-activity relationships. Biochem Pharmacol. 58:759–765. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Raso GM, Meli R, Di Carlo G, Pacilio M and Di Carlo R: Inhibition of inducible nitric oxide synthase and cyclooxygenase-2 expression by flavonoids in macrophage J774A.1. Life Sci. 68:921–931. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Lindenmeyer F, Li H, Menashi S, Soria C and Lu H: Apigenin acts on the tumor cell invasion process and regulates protease production. Nutr Cancer. 39:139–147. 2001. View Article : Google Scholar : PubMed/NCBI

9 

He J, Xu Q, Wang M, Li C, Qian X, Shi Z, Liu LZ and Jiang BH: Oral administration of apigenin inhibits metastasis through AKT/P70S6K1/MMP-9 pathway in orthotopic ovarian tumor model. Int J Mol Sci. 13:7271–7282. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Feng X, Zhou Q, Liu C and Tao ML: Drug screening study using glioma stem-like cells. Mol Med Rep. 6:1117–1120. 2012.PubMed/NCBI

11 

Litchfield DW: Protein kinase CK2: structure, regulation and role in cellular decisions of life and death. Biochem J. 369:1–15. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Trembley JH, Wang G, Unger G, Slaton J and Ahmed K: Protein kinase CK2 in health and disease: CK2: a key player in cancer biology. Cell Mol Life Sci. 66:1858–1867. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Ahmad KA, Wang G, Unger G, Slaton J and Ahmed K: Protein kinase CK2 - a key suppressor of apoptosis. Adv Enzyme Regul. 48:179–187. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Piazza FA, Ruzzene M, Gurrieri C, Montini B, Bonanni L, Chioetto G, Di Maira G, Barbon F, Cabrelle A, Zambello R, Adami F, Trentin L, Pinna LA and Semenzato G: Multiple myeloma cell survival relies on high activity of protein kinase CK2. Blood. 108:1698–1707. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Solares AM, Santana A, Baladrón I, Valenzuela C, González CA, Díaz A, Castillo D, Ramos T, Gómez R, Alonso DF, Herrera L, Sigman H, Perea SE, Acevedo BE and López-Saura P: Safety and preliminary efficacy data of a novel casein kinase 2 (CK2) peptide inhibitor administered intralesionally at four dose levels in patients with cervical malignancies. BMC Cancer. 9:1462009. View Article : Google Scholar

16 

Perea SE, Baladron I, Garcia Y, Perera Y, Lopez A, Soriano JL, Batista N, Palau A, Hernández I, Farina H, Garcia I, Gonzalez L, Gil J, Rodriguez A, Solares M, Santana A, Cruz M, Lopez M, Valenzuela C, Reyes O, López-Saura PA, González CA, Diaz A, Castellanos L, Sanchez A, Betancourt L, Besada V, González LJ, Garay H, Gómez R, Gómez DE, Alonso DF, Perrin P, Renualt JY, Sigman H, Herrera L and Acevedo B: CIGB-300, a synthetic peptide-based drug that targets the CK2 phosphoaceptor domain. Translational and clinical research. Mol Cell Biochem. 356:45–50. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Zhang S, Long H, Yang YL, Wang Y, Hsieh D, Li W, Au A, Stoppler HJ, Xu Z, Jablons DM and You L: Inhibition of CK2α down-regulates Notch1 signalling in lung cancer cells. J Cell Mol Med. 17:854–862. 2013.

18 

Zhang S, Wang Y, Mao JH, Hsieh D, Kim IJ, Hu LM, Xu Z, Long H, Jablons DM and You L: Inhibition of CK2α down-regulates Hedgehog/Gli signaling leading to a reduction of a stem-like side population in human lung cancer cells. PLoS One. 7:e389962012.

19 

Ren KQ, Cao XZ, Liu ZH, Guo H, Quan MF, Liu F, Jiang L, Xiang HL, Deng XY and Cao JG: 8-bromo-5-hydroxy-7-methoxychrysin targeting for inhibition of the properties of liver cancer stem cells by modulation of Twist signaling. Int J Oncol. 43:1719–1729. 2013.PubMed/NCBI

20 

Zhao M, Ma J, Zhu HY, Zhang XH, Du ZY, Xu YJ and Yu XD: Apigenin inhibits proliferation and induces apoptosis in human multiple myeloma cells through targeting the trinity of CK2, Cdc37 and Hsp90. Mol Cancer. 10:1042011. View Article : Google Scholar : PubMed/NCBI

21 

Feliciano A, Castellvi J, Artero-Castro A, Leal JA, Romagosa C, Hernández-Losa J, Peg V, Fabra A, Vidal F, Kondoh HY, Cajal S and Lleonart ME: miR-125b acts as a tumor suppressor in breast tumorigenesis via its novel direct targets ENPEP, CK2-α, CCNJ and MEGF9. PLoS One. 8:e762472013.PubMed/NCBI

Related Articles

Journal Cover

January-2015
Volume 11 Issue 1

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
Liu J, Cao XC, Xiao Q and Quan MF: Apigenin inhibits HeLa sphere‑forming cells through inactivation of casein kinase 2α. Mol Med Rep 11: 665-669, 2015
APA
Liu, J., Cao, X., Xiao, Q., & Quan, M. (2015). Apigenin inhibits HeLa sphere‑forming cells through inactivation of casein kinase 2α. Molecular Medicine Reports, 11, 665-669. https://doi.org/10.3892/mmr.2014.2720
MLA
Liu, J., Cao, X., Xiao, Q., Quan, M."Apigenin inhibits HeLa sphere‑forming cells through inactivation of casein kinase 2α". Molecular Medicine Reports 11.1 (2015): 665-669.
Chicago
Liu, J., Cao, X., Xiao, Q., Quan, M."Apigenin inhibits HeLa sphere‑forming cells through inactivation of casein kinase 2α". Molecular Medicine Reports 11, no. 1 (2015): 665-669. https://doi.org/10.3892/mmr.2014.2720