Metformin induces apoptosis by microRNA-26a-mediated downregulation of myeloid cell leukaemia-1 in human oral cancer cells

  • Authors:
    • Fang Wang
    • Jincheng Xu
    • Hao Liu
    • Zhe Liu
    • Fei Xia
  • View Affiliations

  • Published online on: April 15, 2016     https://doi.org/10.3892/mmr.2016.5143
  • Pages: 4671-4676
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In recent years, population-based studies and retrospective analyses of clinical studies have shown that metformin treatment is associated with reduced cancer incidence and a decrease in cancer‑associated mortality. However, its mechanism of action remains to be fully understood. The present study demonstrates the effects of metformin on KB human oral cancer cells and explores the role of myeloid cell leukaemia‑1 (Mcl‑1) in metformin‑induced mitochondria‑dependent cellular apoptosis. It was demonstrated that metformin exposure caused significant suppression of KB cell proliferation and induced cell death. Furthermore, metformin induced apoptosis through the downregulation of Mcl‑1 in KB human oral cancer cells, and the overexpression of Mcl‑1 in metformin‑treated KB cells significantly increased cell viability. Consistently, Bax and Bim were upregulated in metformin‑treated cells. The results also reveal that microRNA (miR)‑26a expression was markedly increased by metformin. Subsequent to enforced miR‑26a expression in KB cells using miR‑26a mimics, cell viability and the level of Mcl‑1 decreased. These results suggest that the anti‑proliferative effects of metformin in KB cells may result partly from induction of apoptosis by miR-26a-induced downregulation of Mcl-1.

Introduction

Human oral cancer is a frequently occurring subclass of head and neck tumours. Surgery, radiation and chemotherapy are the predominant clinical therapeutic strategies for oral cancer. However, despite advances in curative multimodal treatments, the overall survival rate in patients with oral cancer remains unsatisfactory (1,2). Therefore, it is essential to determine the most effective therapeutic agents to control this disease and enhance patient quality of life.

Apoptosis, or programmed cell death, is regulated in a complex manner by a multitude of factors (3). B-cell lymphoma/leukaemia-2 (Bcl-2) family proteins are important regulators of cell death, and the dysregulation of Bcl-2 family members is key in certain human diseases, including cancer. Myeloid cell leukaemia-1 (Mcl-1) is a pro-survival member of the Bcl-2 protein family that suppresses apoptosis by inhibiting the activity of pro-apoptotic proteins (4,5). Moreover, Mcl-1 expression is highly amplified in a variety of human cancers and is frequently associated with chemotherapeutic resistance and recurrence, suggesting that overexpression of Mcl-1 may contribute to apoptotic evasion and malignant tumour growth. Therefore, targeting Mcl-1 expression in these cancers, using genetic and pharmacological approaches, represents a potential means of developing novel efficacious cancer therapies (6).

Metformin (1,1-dimethylbiguanide hydrochloride) is a biguanide traditional oral hypoglycaemic agent that is widely used in the clinic for the treatment of type 2 diabetes mellitus. It reduces plasma glucose levels by increasing fatty acid oxidation and glucose utilisation, decreasing hepatic gluconeogenesis (7). Recently, metformin has received considerable attention for its antitumour efficacy against numerous types of malignancies. For example, retrospective studies in patients with type 2 diabetes showed reduced cancer incidence in patients who had been treated with metformin for several years (811). Furthermore, other studies have demonstrated that metformin can inhibit tumour growth in vitro and increase tumour sensitivity to chemotherapeutic drugs (1215). All of these studies strongly support the clinical development of metformin as a potentially useful therapeutic agent for cancer.

In the present study, experiments were conducted to examine the effects of metformin on KB human oral cancer cells and determine whether metformin induces apoptosis via Bcl-2 family proteins. Moreover, the present study aimed to further elucidate the mechanism underlying Mcl-1 regulation.

Materials and methods

Chemicals and reagents

Metformin was purchased from Sigma-Aldrich (St. Louis, MO, USA). Metformin solution was prepared by dissolving metformin in phosphate-buffered saline (PBS). Minimum essential medium (MEM), foetal bovine serum (FBS) and PBS were purchased from Gibco Life Technologies (Grand Island, NY, USA). An Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) apoptosis detection kit was purchased from Keygen Biotech (Nanjing, China). Anti-Mcl-1 (rabbit anti-human; 1:100), anti-Bax (rabbit anti-human; 1:1,000), anti-Bim (rabbit anti-human; 1:1,000), and anti-caspase-3 (rabbit anti-human; 1:1,000) monoclonal antibodies were obtained from Abcam (Cambridge, UK). Mouse anti-β-actin antibody (1:2,000) was obtained from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, USA). Cells were transfected with miR-26a mimics, negative control or miR-26a inhibitor purchased from Guangzhou RiboBio (Guangzhou, China).

Cell lines and cell culture

The KB human oral cancer cell line was obtained from the Shanghai Cell Bank at the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in MEM with 10% FBS, and supplemented with 100 U/ml penicillin and 100 mg/ml streptomycin in a humidified incubator at 37°C containing 5% CO2. Cells were passaged at 80% confluence using 0.25% trypsin (Gibco Life Technologies).

Cell proliferation assay

The effect of metformin on KB cell viability was evaluated using the 3-(4,5-dimethylthia-zolyl-2)-2-5 diphenyltetrazolium bromide (MTT) assay. KB cells in the exponential growth phase were plated in 96-well plates at 8×103/well in a final volume of 100 µl. KB cells were exposed to different concentrations of metformin for 24, 48 and 72 h in a humidified incubator at 37°C with 5% CO2. For the MTT assay, cells were then incubated with MTT (5 mg/ml in PBS) for 4 h. Subsequently, the MTT solution was removed and replaced with 150 µl dimethyl sulphoxide/well, and the absorbance was measured at 490 nm using an automated microplate reader (Synergy™ HT; Bio-Tek Instruments, Inc., Winooski, VT, USA). Experiments were repeated in triplicate, and three parallel samples were measured each time.

Annexin V-FITC/PI apoptosis assay

Apoptosis was assessed using double staining with Annexin V-FITC/PI. Briefly, KB cells were treated with metformin at different doses in a 12-well plate for 24 h. Cells were harvested, rinsed twice with PBS and re-suspended in 300 µl ice-cold binding buffer. They were then stained with Annexin V-FITC/PI for 15 min in the dark. Stained cells were analysed using a flow cytometer (Accuri C6; BD Biosciences, Franklin Lakes, NJ, USA).

Western blot analysis

KB cells grown in 6-well plates at a density of 3×105 cells/well were treated with metformin for various incubation periods. Cells were collected by centrifugation and lysed in radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Beijing, China) for 30 min on ice. Cell lysates were centrifuged at 12,000 × g for 30 min at 4°C. Total protein (50 µg) was separated by 15% sodium dodecyl sulphate-polyacrylamide gel (Beyotime Institute of Biotechnology) electrophoresis (70 V, 30 min; 120 V, 90 min) and electrophoretically transferred to polyvinylidene fluoride membranes (Bio-Rad, Hercules, CA, USA). The membranes were blocked with 5% non-fat dry milk for 4 h at room temperature and subsequently incubated with the appropriate antibody for 4 h at room temperature or overnight at 4°C. After washing three times with Tris-PBS, the membranes were incubated with the corresponding secondary antibody. β-actin was used as a control for protein loading.

Plasmid transfection

The pCMV-HA-Mcl-1 plasmid and control plasmid were obtained from GenePharma (Shanghai, China). KB cells were transfected using Lipofectamine 2000 reagent (Invitrogen Life Technologies, Carlsbad, CA, USA) according to the manufacturer's instructions. After cultivation for 48 h, total cell lysates were prepared for western blot analysis or an MTT assay.

Reverse transcription-quantitative polymerase chain reaction for miRNAs

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed for miRNA detection. Total RNA was extracted from KB cells treated with or without metformin using TRIzol reagent (Invitrogen Life Technologies) and an miRNeasy Mini kit (Qiagen, Shanghai, China) according to the manufacturer's instructions. For the detection of miR-26a, mature hsa-miR-26a and U6 primer from All-in-One™ miRNA qPCR Primer (GeneCopoeia, Guangzhou, China) was used. RT-qPCR was performed with the All-in-One™ miRNA qRT-PCR Detection kit (AOMD-Q050, GeneCopoeia) in an ABI StepOne™ Real-Time PCR System (Applied Biosystems Life Technologies, Foster City, CA, USA) at 95°C for 10 min, followed by 40 cycles of 95°C for 10 sec, 60°C for 30 sec and 72°C for 15 sec. PCR data were analysed using the 2−ΔΔCt method and were normalised against RNU6B expression in each sample. Paired Student's t-test was performed to appraise the difference in the level of miRNA expression.

Transfection of miRNA precursor miR-26a

KB cells were seeded in 6-well plates at a density of 3×105 cells/well. Cells at 60–80% confluency were transfected with microRNA (miR)-26a mimics, negative control, miR-26a inhibitor, or inhibitor control at a final concentration of 20 µM using Lipofectamine 2000 reagent (Invitrogen Life Technologies, Carlsbad, CA, USA) according to the manufacturer instructions. Cells were harvested 48 h later for western blot analysis and PI viability assays.

Plate clone formation assay

Cells were seeded at a density of 1×104 cells/well in 6-well plates with growth medium 48 h after transfection and incubated in MEM containing 10% FBS at 37°C with 5% CO2 for 7 days. The colonies were washed with PBS, fixed with 10% formaldehyde for 10 min on ice, and stained with 1.0% crystal violet for 30 min.

Propidium iodide staining

Prior to metformin treatment, cells were plated in 12-well plates and subsequently transfected with miR-26a mimics, negative control, miR-26a inhibitor, or inhibitor control for 48 h. Cells were then subjected to PI staining and evaluated using flow cytometry (Accuri C6).

Statistical analysis

All experiments were repeated at least three times and the values were expressed as the mean ± standard deviation. Statistical significance was determined by Student's t-test using SPSS software, version 13.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Metformin inhibits the proliferation and induces apoptosis of KB cells

To evaluate the growth inhibitory effect of metformin on human KB cells in vitro, cells were treated with different concentrations of metformin for 24, 48 and 72 h. The rate of cell proliferation was inversely related to the time of exposure to metformin and the metformin concentration (Fig. 1A).

Flow cytometric analysis revealed that metformin markedly increased the number of apoptotic KB cells (Fig. 1B). Furthermore, in KB cells, metformin decreased the expression of Mcl-1 and increased the expression of Bim and Bax in a time-dependent manner (Fig. 1C). Downstream of apoptosis signalling pathways, there was a significant activation of caspase-3 cleavage (Fig. 1C). These results confirmed the prediction that metformin induces apoptosis, and that this is predominantly through the mitochondria-mediated internal pathway.

Upregulation of Mcl-1 protects KB cells from apoptosis

To evaluate the association between Mcl-1 and metformin-induced apoptosis, an Mcl-1 expression plasmid was transfected into KB cells (Fig. 2A). Then, overexpression of Mcl-1 significantly increased cell viability in response to metformin treatment and decreased the expression of Bim and Bax compared with their respective controls (Fig. 2B and C). In addition, the cleaved products of caspase-3 were scarcely activated in the Mcl-1-overexpressing KB cells (Fig. 2D). These findings suggest that Mcl-1 may be important in metformin-induced apoptosis as an anti-apoptotic protein.

Metformin increases the expression of miR-26a

Expression of miR-26a was analysed by RT-qPCR and normalised against an endogenous control (RNU6B). Total RNA was extracted from KB cells treated with or without 10 mmol/l metformin. The results indicated that miR-26a was significantly upregulated in metformin-treated cells compared with non-treated cells (Fig. 3).

Overexpression of miR-26a reduces cell viability and regulated Mcl-1 expression in KB cells

In order to confirm the biological function of miR-26a, KB cells were transfected with miR-26a mimics or corresponding negative controls for 7 days, miR-26a inhibited KB cell growth compared with miR-Ctrl (Fig. 4A). As shown in Fig. 4B, the level of Mcl-1 decreased in cells transfected with miR-26a mimics compared with cells transfected with the corresponding negative control. By contrast, Mcl-1 was increased in KB cells transfected with anti-miR-26a inhibitor compared with cells transfected with the corresponding negative control. These experiments confirm that the expression of Mcl-1 can be regulated by miR-26a.

Discussion

Metformin is widely used for the treatment of diabetes mellitus, however, recent epidemiological and preclinical studies have demonstrated that metformin is also a promising anticancer agent. In this study, exposure of KB oral cancer cells to metformin led to a time- and concentration-dependent inhibition of proliferation. Moreover, metformin was found to induce apoptosis.

Apoptosis is an activated cellular death process that is induced by physiological or pathological factors to eliminate redundant and damaged cells. Furthermore, apoptosis is an important defence against cancer (16). When cells are exposed to various stress stimuli, Bim relays apoptotic signals to the mitochondria through the activation of Bax, resulting in an increase in mitochondrial outer membrane permeabilisation and the consequent release of cytochrome c into the cytosol. Cytochrome c then binds to Apaf-1 to ensure the formation of the apoptosome that leads to activation of caspase-9 and the induction of the apoptosis-promoting caspase cascade (17). By contrast, anti-apoptotic Bcl-2 proteins, such as Mcl-1 serve to inhibit apoptosis. Generally, promotion of cell survival by Mcl-1 is hypothesised to be due to sequestration of Bim, thereby inhibiting Bax/Bim interaction in the mitochondrial outer membrane. Alternatively, Mcl-1 may directly bind to Bax and maintain it in an inactive conformation. This competition leads to the suppression of cytochrome c release from the mitochondria and a reduction in Apaf-1-dependent activation of caspase-3. Therefore, Mcl-1 acts in a significant regulatory role that can modulate the expression of pro-apoptotic proteins and control cell fate decisions (18). Consistent with this theory, the results of the present study demonstrate that metformin downregulated the expression of Mcl-1 and upregulated the expression of Bim and Bax. Conversely, overexpression of Mcl-1 downregulated Bim and Bax expression, suggesting that Mcl-1 is involved in KB cell apoptosis, and predominantly through mitochondria-mediated pathways.

In vitro and in vivo studies have shown that the expression of various miRNAs was markedly altered by treatment with metformin (19). By binding to the 3′-untranslated region (UTR) of target mRNAs, miRNAs act as endogenous sequence-specific suppressors that regulate gene expression by eliciting mRNA degradation or inhibition of mRNA translation (20). Therefore, miRNAs are important regulators of tumourigenicity, proliferation, apoptosis, invasion and metastasis. Numerous studies have highlighted the role of miRNAs in Mcl-1 regulation (2123). Analysis of candidate target genes for miR-26a using miRBase previously revealed perfect complementarity between miR-26a and the 3′ UTR of Mcl-1 over the first 9 nucleotides (21,23). In the current study, it was demonstrated that miR-26a significantly downregulated Mcl-1, which led to the apoptosis of KB cells. Furthermore, miR-26a overexpression suppressed in vitro cell proliferation. Conversely, downregulation of miR-26a inhibited apoptosis and promoted proliferation. These results indicate that miR-26a may be a novel tumour suppressor that is important in the regulation of tumoural Mcl-1 expression.

In conclusion, the results of this study confirm that metformin restricted tumour growth and induced apoptosis predominantly by regulating the expression of Bcl-2 family members in the KB human oral cancer cell line. Furthermore, it was demonstrated that metformin increased expression of miR-26a in KB cells and miR-26a itself mediated inhibition of proliferation and induction of apoptosis. In combination, these findings suggest that metformin induces apoptosis in human oral cancer cells by downregulating Mcl-1 via miR-26a and these results provide in vitro evidence to support the use of metformin as a novel and efficient candidate for the treatment of human oral cancer.

Acknowledgments

This study was supported by the National Natural Science Foundation of China (grant nos. 81000992 and 81072207); the Natural Science Foundation of Anhui Province (grant no. KJ2012A201); and Graduate Scientific Research and Innovation Projects of Bengbu Medical College of the Anhui Province (grant no. Byycx1315).

References

1 

Cruz GD, Ostroff JS, Kumar JV and Gajendra S: Preventing and detecting oral cancer. Oral health care providers' readiness to provide health behavior counseling and oral cancer examinations. J Am Dent Assoc. 136:594–601. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Rogers SN, Brown JS, Woolgar JA, Lowe D, Magennis P, Shaw RJ and Vaughan D: Survival following primary surgery for oral cancer. Oral Oncol. 45:201–211. 2009. View Article : Google Scholar

3 

Plati J, Bucur O and Khosravi-Far R: Apoptotic cell signaling in cancer progression and therapy. Integr Biol (Camb). 3:279–296. 2011. View Article : Google Scholar

4 

Perciavalle RM and Opferman JT: Delving deeper: MCL-1′s contributions to normal and cancer biology. Trends Cell Biol. 23:22–29. 2013. View Article : Google Scholar

5 

Thomas LW, Lam C and Edwards SW: Mcl-1; the molecular regulation of protein function. FEBS Lett. 584:2981–2989. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Quinn BA, Dash R, Azab B, Sarkar S, Das SK, Kumar S, Oyesanya RA, Dasgupta S, Dent P, Grant S, et al: Targeting Mcl-1 for the therapy of cancer. Expert Opin Investig drugs. 20:1397–1411. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Foretz M and Viollet B: New promises for metformin: Advances in the understanding of its mechanisms of action. Med Sci (Paris). 30:82–92. 2014.In French. View Article : Google Scholar

8 

Evans JM, Donnelly LA, Emslie Smith AM, Alessi DR and Morris AD: Metformin and reduced risk of cancer in diabetic patients. BMJ. 330:1304–1305. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Noto H, Goto A, Tsujimoto T and Noda M: Cancer risk in diabetic patients treated with metformin: A systematic review and meta-analysis. PLoS One. 7:e334112012. View Article : Google Scholar : PubMed/NCBI

10 

Decensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B and Gandini S: Metformin and cancer risk in diabetic patients: A systematic review and meta-analysis. Cancer Prev Res (Phila). 3:1451–1461. 2010. View Article : Google Scholar

11 

Landman GW, Kleefstra N, van Haterenm KJ, Groenier KH, Gans RO and Bilo HJ: Metformin associated with lower cancer mortality in type 2 diabetes: ZODIAC-16. Diabetes Care. 33:322–326. 2010. View Article : Google Scholar :

12 

Wang F, Xu J, Xia F, Liu Z, Zhao S, Liu H and Jiang Z: Effects of metformin on human oral cancer KB cell proliferation and apoptosis in vitro. Nan Fang Yi Ke Da Xue Xue Bao. 34:159–163. 2014.In Chinese. PubMed/NCBI

13 

Marini C, Salani B, Massollo M, Amaro A, Esposito AI, Orengo AM, Capitanio S, Emionite L, Riondato M, Bottoni G, et al: Direct inhibition of hexokinase activity by metformin at least partially impairs glucose metabolism and tumor growth in experimental breast cancer. Cell Cycle. 12:3490–3499. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Mohammed A, Janakiram NB, Brewer M, Ritchie RL, Marya A, Lightfoot S, Steele VE and Rao CV: Antidiabetic drug metformin prevents progression of pancreatic cancer by targeting in part cancer stem cells and mTOR signaling. Transl Oncol. 6:649–659. 2013. View Article : Google Scholar

15 

Miyoshi H, Kato K, Iwama H, Maeda E, Sakamoto T, Fujita K, Toyota Y, Tani J, Nomura T, Mimura S, et al: Effect of the anti-diabetic drug metformin in hepatocellular carcinoma in vitro and in vivo. Int J Oncol. 45:322–332. 2014.PubMed/NCBI

16 

Melet A, Song K, Bucur O, Jagani Z, Grassian AR and Khosravi-Far R: Apoptotic pathways in tumor progression and therapy. Adv Exp Med Biol. 615:47–79. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Del Gaizo Moore V and Letai A: BH3 profiling-measuring integrated function of the mitochondrial apoptotic pathway to predict cell fate decisions. Cancer Lett. 332:202–205. 2013. View Article : Google Scholar

18 

Karnak D and Xu L: Chemosensitizaition of prostate cancer by modulating Bcl-2 family proteins. Curr Drug Targets. 11:699–707. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Li W, Yuan Y, Huang L, Qiao M and Zhang Y: Metformin alters the expression profiles of microRNAs in human pancreatic cancer cells. Diabetes Res Clin Pract. 96:187–195. 2012. View Article : Google Scholar : PubMed/NCBI

20 

O'Hara SP, Mott JL, Splinter PL, Gores GJ and LaRusso NF: MicroRNAs: Key modulators of posttranscriptional gene expression. Gastroenterology. 136:17–25. 2009. View Article : Google Scholar

21 

Yang X, Liang L, Zhang XF, Jia HL, Qin Y, Zhu XC, Gao XM, Qiao P, Zheng Y, Sheng YY, et al: MicroRNA-26a suppresses tumor growth and metastasis of human hepatocellular carcinoma by targeting interleukin-6-Stat3 pathway. Hepatology. 58:158–170. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Zhang YK, Wang H, Leng Y, Li ZL, Yang YF, Xiao FJ, Li QF, Chen XQ and Wang LS: Overexpression of microRNA-29b induces apoptosis of multiple myeloma cells through down regulating Mcl-1. Biochem Biophys Res Commun. 414:233–239. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Gao J, Li L, Wu M, Liu M and Xie X, Guo J, Tang H and Xie X: MiR-26a inhibits proliferation and migration of breast cancer through repression of MCL-1. PLoS One. 8:e651382013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 13 Issue 6

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
Wang F, Xu J, Liu H, Liu Z and Xia F: Metformin induces apoptosis by microRNA-26a-mediated downregulation of myeloid cell leukaemia-1 in human oral cancer cells. Mol Med Rep 13: 4671-4676, 2016
APA
Wang, F., Xu, J., Liu, H., Liu, Z., & Xia, F. (2016). Metformin induces apoptosis by microRNA-26a-mediated downregulation of myeloid cell leukaemia-1 in human oral cancer cells. Molecular Medicine Reports, 13, 4671-4676. https://doi.org/10.3892/mmr.2016.5143
MLA
Wang, F., Xu, J., Liu, H., Liu, Z., Xia, F."Metformin induces apoptosis by microRNA-26a-mediated downregulation of myeloid cell leukaemia-1 in human oral cancer cells". Molecular Medicine Reports 13.6 (2016): 4671-4676.
Chicago
Wang, F., Xu, J., Liu, H., Liu, Z., Xia, F."Metformin induces apoptosis by microRNA-26a-mediated downregulation of myeloid cell leukaemia-1 in human oral cancer cells". Molecular Medicine Reports 13, no. 6 (2016): 4671-4676. https://doi.org/10.3892/mmr.2016.5143