Metformin inhibits mTOR and c-Myc by decreasing YAP protein expression in OSCC cells

  • Authors:
    • Ying Wang
    • Yafei Zhang
    • Xiaomei Feng
    • Hui Tian
    • Xucheng Fu
    • Weiting Gu
    • Yong Wen
  • View Affiliations

  • Published online on: December 24, 2020     https://doi.org/10.3892/or.2020.7909
  • Pages: 1249-1260
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Metformin is an antidiabetic drug that has been reported to have an inhibitory effect on different types of cancers, including oral squamous cell carcinoma (OSCC). However, few studies have explored the role of Yes‑associated protein (YAP), a vital factor contributing to OSCC biology, in metformin‑induced anticancer activity in OSCC cells. Thus, the purpose of the present study was to investigate the molecular relationship between metformin and YAP in OSCC cells. CAL27 and SCC25 cell lines were treated with various concentrations of metformin for 24 h. Cell proliferation was detected by Cell Counting Kit‑8 (CCK‑8) and flow cytometric assays. Cell apoptosis was analyzed using flow cytometry. The intracellular protein levels of target genes were determined by western blotting. It was demonstrated that metformin affected the cell cycle and apoptosis of CAL27 and SCC25 cells. Alteration of YAP protein expression affected metformin‑mediated changes in the cell cycle and apoptosis of CAL27 and SCC25 cells. In addition, compared to the control treatment, metformin treatment decreased the protein levels of YAP, mTOR, p‑mTOR and c‑Myc. The overexpression of YAP alleviated the inhibitory effect of metformin on the protein expression of mTOR, p‑mTOR and c‑Myc. The combination of metformin and verteporfin markedly enhanced the effects of metformin on the protein expression of mTOR, p‑mTOR and c‑Myc. Therefore, the results of the present study suggest that metformin suppresses OSCC by inhibiting YAP protein expression and by suppressing the YAP‑mediated effects of metformin on the protein expression of mTOR and c‑Myc.

Introduction

Oral squamous cell carcinoma (OSCC) is a major burden to human health according to a 2019 epidemiological investigation of cancer (1). The major carcinogenic factors of OSCC are tobacco, alcohol and ultraviolet rays. The typical clinical features of OSCC are obscure boundaries, bleeding and ulcers that affect the chewing, speech and appearance of patients (2). The biological behavior of OSCC cells, including rapid proliferation, resistance to apoptosis, rapid invasion and epithelial-mesenchymal transition, is related to the OSCC clinical phenotype (3). With the rapid development of molecular biology, it has been revealed that the expression of some intracellular molecules is correlated with the biological behavior of OSCC (4,5), and it has been demonstrated that high expression of Yes-associated protein (YAP) results in a rapid cell proliferation rate and poor differentiation of OSCC (6,7). Our previous study also revealed that YAP could be transported into the nucleus, driving the transcription of the c-Myc and Bcl-2 genes, which impairs apoptosis and facilitates the proliferation of OSCC cells (8). In addition, it has also been reported that overexpression of YAP is related to the drug resistance and epithelial to mesenchymal properties of OSCC (9,10). Another study also indicated that cell growth and invasion could be inhibited by the suppression of YAP in OSCC (11,12). Therefore, YAP may be a tumor promoter and could be used as a potential anticancer drug target for OSCC (13).

Metformin, which is extracted from French lilac and easily produced, is an effective hypoglycemic drug with low toxicity (14). It has been revealed that metformin reduces the cancer risk and mortality of diabetic patients compared with that of nonusers via meta-analysis and epidemiology studies (15,16). Therefore, as metformin is widely used, researchers have revealed that metformin can effectively reduce the risk of cancer in people with diabetes, and a dose-response relationship was proposed (16). In addition, previous studies have also revealed that metformin improves the survival of patients who suffer from head and neck squamous cell carcinoma (including oral cancer) (17,18). Recently, it has been demonstrated that metformin suppresses the growth of OSCC cells by promoting cell apoptosis and decreasing the malignant potential of OSCC (19,20). Metformin can also increase the sensitivity of OSCC cells to cisplatin and disulfiram (21,22). Thus, metformin may become a promising anticancer drug for OSCC. However, it has been confirmed that metformin inhibits OSCC by inhibiting the AMPK/mTOR (19), NF-κB/HIF-1α (21) and LSF/Aurora-A (23) pathways in cells. The relationship between metformin and YAP in OSCC, according to our knowledge, remains uncertain.

Metformin exerts its anticancer effect by reducing mTOR and c-Myc protein expression in cancer cells (2427), and YAP expression is related to mTOR and c-Myc expression. YAP translocates from the cytoplasm to the nucleus once the Hippo signaling pathway is inactivated, which leads to increased expression of mTOR in tuberous sclerosis complex cells and colorectal cancer cells (28,29). Hansen et al also explained that YAP promotes the expression of mTOR protein via the high-affinity leucine transporter LAT1 (30). Moreover, it has been verified that the protein expression of c-Myc in gastric cancer cells and liver cancer cells is upregulated by the overexpression of YAP (3134). Our previous research also indicated that YAP translocates to the nucleus, driving c-Myc gene transcription in OSCC cells (8). Thus, we hypothesized that metformin can downregulate the protein expression of c-Myc and mTOR via a YAP-dependent mechanism in OSCC cells.

In the present study, the relationship between metformin and YAP in OSCC and the regulatory mechanism by which YAP regulates the protein expression of c-Myc and mTOR in metformin-treated cells were investigated. The present study thus revealed a novel molecular mechanism of metformin in OSCC.

Materials and methods

Cell lines

Human tongue squamous cell carcinoma CAL27 and SCC25 cell lines were purchased from ATCC. CAL27 cells were cultured in high-glucose culture medium (DMEM; HyClone; Cytiva) containing 10% fetal bovine serum (FBS; Biological Industries) and 1% penicillin-streptomycin (HyClone; Cytiva) . SCC25 cells were cultured in high-glucose culture medium (DMEM F12; HyClone; Cytiva) containing 10% FBS and 1% penicillin-streptomycin.

Reagents and antibodies

Metformin (item no. D9351) was purchased from Solarbio Life Sciences, and Verteporfin (CL 318952) was purchased from Sigma-Aldrich; Merck KGaA. Cell Counting Kit-8 (CCK-8) was purchased from Dojindo Molecular Technologies, Inc. Both cell cycle assay kit and Annexin V-PI cell apoptosis assay kit were purchased from Hangzhou Multi Sciences (Lianke) Biotech Co., Ltd. APC-7AAD cell apoptosis assay kit was purchased from Sungene Biotech Co., Ltd. Antibodies against CDK4 (D9G3E; product no. 12790), CDK6 (D4S8S; product no. 13331), p21 (12D1; product no. 2947), Bax (D2E11; product no. 5023), Bcl-2 (D55G8; product no. 4223), mammalian STE20-like kinase 1 (MST1) (D8B9Q; product no. 14946), phosphorylated (p)-MST (Thr183; E7UD1; product no. 49332), large tumor suppressor 1 (LATS1) (C66B5; product no. 3477), p-LATS1 (Thr1097; D57D3; product no. 8654), YAP (D8H1X; product no. 14074), p-YAP (Ser127; D9W2I; product no. 13008), mTOR (7C10; product no. 2983), p-mTOR (Ser2448; D9C2; product no. 5536), c-Myc (D84C12; product no. 5605), SAV1 (D6M6X; product no. 13301) and MOB1 (E1N9D; product no. 13730) were obtained from Cell Signaling Technologies, Inc.

Cell proliferation assay

A Cell Counting Kit-8 (CCK-8) assay was used to detect changes in cell proliferation. First, the cells were seeded at 2,500 cells/well in 96-well plates and cultured in a 37°C carbon dioxide incubator for 24 h. Next, 0, 5, 15 and 30 mmol/l metformin was added to 96-well plates, and the cells were cultured for 24, 48 and 72 h. Then, 10 µl of CCK-8 reagent was added to each well and incubated for 2.5 h. Subsequently, a microplate reader was used to detect the absorbance at 450 nm.

Cells treated without metformin or YAP overexpression lentiviral vector served as the control group. The experimental groups were as follows: Cells treated with metformin; cells treated with YAP overexpression lentiviral vector; and cells treated with metformin and YAP overexpression lentiviral vector. A 15-mmol/ml concentration of metformin was used in the relevant experimental groups. The cell viability of each group was detected by CCK-8 assay after 24 h of drug incubation, and a microplate reader was used to detect the absorbance at 450 nm.

Cell cycle assay

Firstly, cells (2.5×106) were treated with 0, 5, 15 and 30 mmol/l metformin for 24 h. Furthermore, cells treated without metformin and overexpression YAP gene lentiviral vector served as the control group; cells treated with metformin, overexpression YAP gene lentiviral vector as well as metformin and overexpression YAP gene lentiviral vector served as the experimental groups. In addition, cells treated without metformin and verteporfin served as the control group; cells treated with verteporfin, as well as metformin and verteporfin served as the experimental groups. A concentration of 15 mmol/l metformin or 1 µM verteporfin was used in the experimental groups. After 24-h drug incubation, 70% ethanol was used to fix cells for 12 h at 4°C, after washing the cells with PBS. The cells were treated with cell cycle detection kit (item no. CA1510; Solarbio Life Sciences) according to the manufacturer's instructions and the proportion of each cell cycle was analyzed using a flow cytometer (FACSCanto II; BD Biosciences).

Cell apoptosis assay

Cells treated with 0 mmol/l metformin served as the control group and cells respectively treated with 5, 15 and 30 mmol/l metformin served as the experimental groups. In addition, cells treated without metformin and overexpression YAP gene lentiviral vector served as the control group while cells treated with metformin, overexpression YAP gene lentiviral or cells treated with metformin combined with overexpression YAP gene lentiviral vector served as the experimental groups. Similarly, cells treated without metformin or verteporfin served as the control group while cells treated with verteporfin, metformin or metformin combined with verteporfin served as the experimental groups. The concentration of metformin was 15 mmol/l and the concentration of verteporfin was 1 µl in the experimental groups. After 24 h of drug incubation, cells were stained using a green fluorescent-labeled Annexin V-APC staining kit (Sungene Biotech Co., Ltd.) according to the manufacturer's instructions. Cells (2×105) were collected from each group of samples. Secondly, 500 µl of 1X binding buffer and 8 µl of Annexin V-fluorescein APC were used to suspend cells for 10 min. Finally, the cells were incubated with 5 µl of 7-AAD solution for 5 min. Similarly, Annexin V-fluorescein isothiocyanate/propidium iodide (FITC/PI) (Hangzhou Multi Sciences (Lianke) Biotech Co., Ltd.) was used to detect cells with no staining. Finally, the proportion of apoptotic cells was calculated by flow cytometry (FACSCanto II; BD Biosciences).

Western blotting

Cells (8×106) were cultured in a culture plate and were collected, and the total protein in cells was extracted with an ice-cold lysate containing a phosphatase inhibitor and a protease inhibitor (Solarbio Life Sciences). The protein concentration was determined by a bicinchoninic acid assay (Boster Biological Technology, Co., Ltd.). The proteins (20 µg total protein/lane) were separated using 10% SDS-PAGE. Electrophoresis and transfer were performed according to the standard protocol of western blotting. After blocking the PVDF membrane for 1 h at 4°C with 5% non-fat milk in Tris-buffered saline (TBST) containing 0.1% Tween-20, the membrane was probed with a primary antibody (1:1,000) overnight at 4°C. Then the membrane was incubated with HRP-conjugated secondary antibody (1:20,000; product no. 7074; Cell Signaling Technology, Inc.) for 40 min at 25°C. Finally, protein bands were detected using the Immobilon Western Chemiluminescent HRP Substrate Kit (Vazyme Biotech Co., Ltd.). Densitometric analysis was performed using ImageJ v1.52 (National Institutes of Health).

Construction of YAP-overexpressing cell lines

A YAP overexpression lentiviral vector (1×108 TU/ml) was constructed, and an empty lentiviral vector was used as a control. Both vectors were prepared by the Shanghai Genechem Co., Ltd. and transfected into the CAL27 and SCC25 cell lines for 72 h at 27°C. Subsequently, 24 h later the transfection efficiency was detected by western blotting and RT-qPCR.

Reverse transcription quantitative (RT-q)PCR analysis

Cells treated with lentiviral blank vector served as the control group while cells treated with overexpressed YAP vector served as the experimental group. Then, 72 h later, total RNA in cells was extracted by TRIzol total RNA isolation (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The purity of the obtained total RNA samples was detected by NanoDrop spectrophotometer (Thermo Fisher Scientific, Inc.). Total RNA (1 µg) was reverse transcribed into cDNA using PrimeScript™ RT II reagent kit (Takara Bio, Inc.) on Biometra PCR thermal cycler (Applied Biosystems; Thermo Fisher Scientific, Inc.). Then, 2 µl RNase-free water (Takara Bio, Inc.), 2 µl primers (Sangon Biotech Co., Ltd.), 1 µl obtained cDNA samples and 5 µl SYBR-Green fluorescent dye (Takara Bio, Inc.) were mixed into a 10-µl reaction system. The forward primer of YAP was 5′-TCTTACACCGTGCTGCCATT-3′ and the reverse primer was 5′-AGCACCTGTCCAGGTATCAC-3′. The forward primer of GAPDH was 5′-GCACCGTCAAGGCTGAGAAC-3′ and the reverse primer was 5′-TGGTGAAGACGCCAGTGGA-3′. Finally, the gene expression was detected by Light Cycler Roche 480 RT-qPCR instrument (Roche Diagnostics). The results of fold-changes in mRNA levels were calculated by using a 2−ΔΔCq method (35).

Statistical analysis

One-way analysis of variance (ANOVA) followed by Tukey's post hoc test were used to analyze the differences between several research groups, and the difference between two groups was analyzed by two-tailed t-test (P<0.05 and P<0.01 were considered to indicate statistically significant differences). All the statistical results were analyzed by SPSS software 20.0 (IBM Corp.).

Results

Metformin inhibits cell proliferation and promotes cell apoptosis in OSCC

To illustrate the effect of metformin on the growth of CAL27 and SCC25 cells, cells were first treated with 0, 5, 15 and 30 mmol/l metformin, and the changes in cell proliferation were assessed at 24, 48 and 72 h by CCK-8 assay. It was evident that cell proliferation decreased with increasing metformin concentration and increasing dosing time (Fig. 1). The results of flow cytometric assays revealed that the proportion of cells arrested in the G0/G1 phase was increased, while the proportion of cells in the S phase and G2 phase were reduced in a dose-dependent manner (Fig. 2A and B). Then, FITC/PI staining and flow cytometry were used to detect the cell apoptosis induced by metformin and it was revealed that the early apoptosis rate and late apoptosis rate were significantly increased with metformin treatment compared with the control treatment (Fig. 2C and D). In addition, biomarkers of proliferation and apoptosis were detected by western blotting. The protein expression levels of CDK4 and CDK6, which are involved in the G0/G1 phase (36), were markedly decreased in metformin-treated cells compared to control cells. p21 has the ability to inhibit the transition from the G1 to S phase in the cell cycle (37), and its expression was increased accordingly (Fig. 3A). The decreased protein expression of Bcl-2 and the increased expression of the proapoptotic protein Bax in the metformin group vs. the control group confirmed that metformin inhibited cell growth by promoting apoptosis in the CAL27 and SCC25 cell lines (Fig. 3A). The results revealed that metformin arrested the cell cycle in the G1/S phase and promoted cell apoptosis in vitro.

Metformin stimulates the Hippo signaling pathway in OSCC cells

YAP is one of the transcription coactivators inhibited by the Hippo pathway and is related to OSCC cell growth (6). The signaling molecules related to the Hippo pathway rely on a phosphorylation cascade. MST1 can be phosphorylated at Thr183 as a result of activation of the Hippo pathway. Then, phosphorylated MST1 can bind with SAV1 to phosphorylate LATS1 at T1097, which can bind MOB1. Next, upstream activation of the Hippo pathway can result in the phosphorylation of Yap at S127. Subsequently, YAP is susceptible to proteasomal degradation (38). In the present results, metformin increased the phosphorylation of MST1 (Thr183) and LATS1 (T1097), and the expression of total protein was not markedly altered as the dose of metformin increased. The protein expression of SAV1 and MOB1 also increased with metformin treatment (Fig. 3B).

Metformin inhibits OSCC cell growth by decreasing YAP

The YAP expression level was enhanced in CAL27 and SCC25 cells by using YAP overexpression lentiviral particles. YAP protein expression was confirmed to be higher in the overexpression group than in the control group (Fig. 4A). The mRNA expression of YAP in the YAP overexpression group was also higher than that in the control group (Fig. 4B). In contrast to transfection with empty lentiviral vector, transfection with YAP overexpression vector significantly weakened the effects of metformin on OSCC cells. The optical density (OD) value at 450 nm in the CCK-8 assay of YAP-overexpressing cells was higher than that of empty lentivirus-transfected cells after metformin treatment for 24 h (Fig. 4C). The proportion of cells arrested in G1 phase of the cell cycle in the YAP overexpression group was lower than that in the empty lentivirus group after metformin treatment for 24 h (Fig. 5A and B). The cell apoptosis rate in the YAP overexpression group was lower than that in the empty lentivirus group after metformin treatment for 24 h (Fig. 5C and D). To further identify the role of YAP in metformin treatment, metformin was combined with the YAP inhibitor verteporfin. The proportion of cells at the G1 phase (Fig. 6A and B) and the proportion of apoptotic cells in the metformin and verteporfin treated group were higher than those in the metformin-treated group (Fig. 6C and D). The YAP overexpression group exhibited higher expression of CDK4, CDK6 and Bcl-2 and lower expression of Bax than the empty lentivirus group after metformin treatment for 24 h (Fig. 7A). However, the CDK4, CDK6, and Bcl-2 protein expression was lower and the Bax expression was higher in the verteporfin and metformin combination treatment group than in the metformin treatment group (Fig. 7B). These results clearly indicated that metformin had an inhibitory effect on the proliferation of OSCC cells by decreasing YAP protein.

Metformin decreases mTOR and c-Myc through the downregulation of YAP

Other studies have revealed that YAP promotes the expression of mTOR in hepatocellular cells and colorectal cancer cells (39,40). Our previous study also revealed that YAP facilitated the protein expression of c-Myc (8). However, few studies have reported that metformin inhibits the expression of mTOR and c-Myc through a YAP-dependent mechanism in OSCC cells (2427). To further illustrate the molecular relationship between metformin and YAP in OSCC cells, western blotting was used to detect changes in mTOR and c-Myc expression in OSCC cells. As anticipated, a gradual reduction in mTOR expression, mTOR phosphorylation at Ser2448 and c-Myc expression was observed as the metformin concentration increased (Fig. 8A). Overexpression of YAP facilitated mTOR and c-Myc protein expression in CAL27 and SCC25 cells treated with metformin (Fig. 8B). The metformin and verteporfin combination group exhibited a decreased expression level of mTOR and c-Myc protein than the metformin treatment groups (Fig. 8C). These results indicated that YAP inhibition was associated with the metformin-induced decrease in mTOR and c-Myc expression in OSCC cells.

Discussion

Metformin, discovered from French lilac, has been widely studied for its role in cancer treatment since it has an antitumor ability with characteristics of low toxicity and low cost production (41). It has been revealed that metformin-treated head and neck squamous cell carcinoma patients had a higher survival rate than those not treated with metformin, according to a meta-analysis (17,18). Previous studies have indicated that metformin promotes the toxic effects of disulfiram, 5-fluorouracil and cisplatin on OSCC cells since metformin enhances the inhibitory effects of these traditional chemotherapeutics, even at low concentrations (21,22,42,43). In addition, it has also been demonstrated that metformin suppresses cell proliferation and increases cell apoptosis in OSCC in vivo and in vitro (19). Although some studies have elucidated the effects of metformin on OSCC, the molecular mechanism of metformin on OSCC cells remains undefined.

Prolonged proliferation and insensitivity to apoptosis are two main types of biological characteristics typically featured in human tumors (44). Therefore, controlling cancer cell proliferation and apoptosis has a certain significance for the clinical treatment of cancer. YAP, as a vital carcinogenic factor, is important in OSCC growth, and high expression of YAP protein may accelerate the progression of OSCC (6). YAP was demonstrated to enhance the proliferation rate of cells and reduce apoptosis by promoting c-Myc and Bcl-2 protein expression in OSCC (8). Moreover, cell growth and invasion were inhibited by the suppression of YAP in OSCC (11). Inhibition of YAP protein also decreased the drug resistance of cisplatin in OSCC cells (9). Thus, to determine whether metformin has an inhibitory effect on OSCC, the YAP protein was inhibited.

The protein binding between YAP and TEAD4 has been demonstrated to accelerate the cell proliferation rate by markedly promoting the transition of the cell cycle from the G1 to S phase and reducing cell apoptosis (8). In addition, the protein expression of CDK4/6 and p21 is involved in the transition of the cell cycle from the G1 to S phase. CDK4, CDK6 and other cyclin-dependent kinases can accelerate the transition of the cell cycle from the G1 to the S phase (36). p21 is an inhibitor of cyclin-dependent kinases, and it has a negative regulatory effect on the cell cycle transition from the G1 to the S phase (37). The Bcl-2 protein family can control the apoptosis process of cells by influencing the outer mitochondrial membrane potential. Bcl-2 protein and Bax protein belong to the Bcl-2 protein family. The former blocks the process of cell apoptosis, while the latter stimulates the processes of cell apoptosis (45). As anticipated, in the present study, metformin inhibited the transition of the cell cycle from the G1 to S phase and increased cell apoptosis in CAL27 and SCC25 cells. The protein expression of YAP in CAL27 and SCC25 cells was also decreased with metformin treatment compared to control treatment. The protein expression of CDK4, CDK6, p21, Bcl-2 and Bax also paralleled the phenotypic changes upon metformin exposure.

Next, YAP overexpression lentiviral particles or empty lentiviral vectors were transfected into CAL27 and SCC25 cells. The cells were divided into a YAP overexpression group and a negative control group. The protein and mRNA expression levels of YAP in the YAP overexpression group were higher than those of the empty vector group. In fact, metformin inhibited the transition of the cell cycle from the G1 to S phase and increased cell apoptosis in the control group. However, overexpression of YAP reversed the inhibitory effects of metformin on the cell cycle and attenuated metformin-induced cell apoptosis. The protein expression of CDK4, CDK6, p21, Bcl-2 and Bax also paralleled the phenotypic changes in the YAP overexpression group.

To further confirm that metformin affected the cell cycle and apoptosis by decreasing YAP protein, cells were treated with both metformin and verteporfin, which is an inhibitor of YAP protein. Verteporfin is a photosensitizer that can be used for photodynamic therapy to treat macular degeneration with limited toxicity (46). Recent studies have illustrated that verteporfin effectively inhibits the development of cancer by decreasing the expression of YAP protein in cancer cells (47,48). For example, verteporfin inhibits the translocation of YAP protein into the nucleus by inhibiting the binding of YAP and TEAD (49) or promotes the expression of 14-3-3 protein so that YAP remains in the cytoplasm (50). Our previous experiments also verified that the expression of YAP protein in OSCC cells decreased with increasing verteporfin drug concentration (8). Therefore, verteporfin was combined with metformin. As anticipated, the effects of the combined use of metformin and verteporfin on the cell cycle and apoptosis were greater than those of a single medication. The protein expression of CDK4, CDK6, Bcl-2 and Bax also paralleled the phenotypic changes in the metformin and verteporfin combination group. These data clarified that metformin has an inhibitory effect on OSCC by inhibiting the YAP protein.

YAP is a main downstream factor of the Hippo signaling pathway and can be inhibited by the Hippo signaling pathway (6). MST1, LATS1, SAV1 and MOB1 constitute the upstream regulatory signal transduction of the Hippo signaling pathway. It is known that MST1 can be phosphorylated at Thr183 as a result of activation of the Hippo pathway. Then, phosphorylated MST1 can bind with SAV1 to phosphorylate LATS1 at T1097, which can bind MOB1. Subsequently, the upstream Hippo pathway is activated, which could result in the phosphorylation of YAP at S127. Phosphorylated YAP is susceptible to proteasomal degradation (51). Phosphorylation of S127 results in YAP sequestration in the cytoplasm via interaction with 14-3-3 (11) or SCFβ-TrCP E3 ubiquitin ligase (52). The present results demonstrated that metformin increased the phosphorylation of MST1 (Thr183) and LATS1 (T1097), with virtually no change in the expression level of total protein as metformin concentration increased. The protein expression of SAV1 and MOB1 also increased with metformin treatment. A change in YAP phosphorylation (S127) and a progressive decrease in YAP expression followed the activation of the upstream signaling factor in the Hippo pathway. These results indicated that metformin may reduce the protein expression of YAP by activating the Hippo pathway.

The mTOR signaling pathway transmits information concerning synthesis or catabolism in cells and affects the metabolism of proteins, lipids and nucleotides in cells, thereby providing sufficient substances for cancer growth (53). Observation of patients with tongue squamous cell carcinoma revealed high expression levels of mTOR and p-mTOR proteins in patient tissue sections (54). It has been demonstrated that circular RNA hsa-circ-0007059 or activated C kinase 1 receptor promotes the proliferation of OSCC cells by increasing the expression of mTOR protein (55,56). YAP serves as a vital association between the Hippo pathway and the PI(3)K-mTOR pathway. It has been confirmed that SAV1 inhibits the protein expression of mTOR by inactivating the YAP protein in colorectal cancer (29). It has also been illustrated that mTOR blocks the inhibitory effect of AMOTL2 on YAP to promote cell growth and invasion in glioblastoma (57). In addition, YAP has been revealed to increase the expression level of miR-29, which indirectly increases the protein expression of mTOR in cells (40). In the present study, metformin decreased the protein expression of mTOR in a dose-dependent manner. YAP overexpression inhibited the ability of metformin to decrease the protein expression of mTOR. In contrast, the combination of metformin with verteporfin enhanced the ability of metformin to decrease the protein expression of mTOR.

The Myc genes are a family of proto-oncogenes. MYC family proteins are involved in regulating the proliferation, differentiation and apoptosis of cancer cells and are closely related to cell division activities (58). A study of OSCC patients revealed that the level of c-Myc protein expression was closely related to disease severity and survival rate (59). Our previous study revealed that YAP is transported into the nucleus, where it drives the transcription of the c-Myc gene to accelerate OSCC cell growth (8). Other studies also demonstrated by RNA sequencing that c-Myc serves as a definitive target of YAP and mediates the ability of YAP to promote cancer growth (31,33). Thus, it was investigated whether metformin has an inhibitory effect on c-Myc in OSCC cells by inhibiting the YAP protein. In the present study, metformin decreased the protein expression of c-Myc in a dose-dependent manner. YAP overexpression inhibited the ability of metformin to decrease the protein expression of c-Myc. Conversely, the combination of metformin with verteporfin enhanced the ability of metformin to decrease the protein expression of c-Myc.

In conclusion, the present experiments demonstrated that metformin prevented OSCC by reducing the expression of YAP protein. Furthermore, it was revealed that YAP-mediated metformin also had regulatory effects on c-Myc and mTOR proteins. This provides a new reference for the anticancer mechanism of metformin. However, this study still has some limitations. For example, this molecular mechanism has not been verified by in vivo experiments in the present study. If conditions permit, further experiments will be designed to explore and verify this related hypothesis in the future. The present study thus revealed that YAP may be recognized as a potential target for metformin treatment in OSCC.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the Shandong Provincial Natural Science Foundation (grant no. ZR2018MH018), the Young Scholars Program of Shandong University (grant no. 2015WLJH53), the China Postdoctoral Science Foundation (grant no. 2017M610432), the Shandong Postdoctoral Innovation Project (grant no. 201703071) and the Construction Engineering Special Fund of Taishan Scholars (grant no. ts201511106).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

YWa, YWe, WG and YZ designed and executed the study. YZ, XF, HT and XF analyzed the data. WY wrote the study. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Feller L and Lemmer J: Oral squamous cell carcinoma: Epidemiology, clinical presentation and treatment. J Cancer Ther. 3:263–268. 2012. View Article : Google Scholar

3 

Rodrigues MFSD, Miguita L, De Andrade NP, Heguedusch D, Rodini CO, Moyses RA, Toporcov TN, Gama RR, Tajara EE and Nunes FD: GLI3 knockdown decreases stemness, cell proliferation and invasion in oral squamous cell carcinoma. Int J Oncol. 53:2458–2472. 2018.PubMed/NCBI

4 

Zhang L, Meng X, Zhu XW, Yang DC, Chen R, Jiang Y and Xu T: Long non-coding RNAs in oral squamous cell carcinoma: Biologic function, mechanisms and clinical implications. Mol Cancer. 18:1022019. View Article : Google Scholar : PubMed/NCBI

5 

Sakata J, Hirosue A, Yoshida R, Kawahara K, Matsuoka Y, Yamamoto T, Nakamoto M, Hirayama M, Takahashi N, Nakamura T, et al: HMGA2 contributes to distant metastasis and poor prognosis by promoting angiogenesis in oral squamous cell carcinoma. Int J Mol Sci. 20:202019. View Article : Google Scholar

6 

Hiemer SE, Zhang L, Kartha VK, Packer TS, Almershed M, Noonan V, Kukuruzinska M, Bais MV, Monti S and Varelas X: A YAP/TAZ-regulated molecular signature is associated with oral squamous cell carcinoma. Mol Cancer Res. 13:957–968. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Li SY, Hu JA and Wang HM: Expression of Yes-associated protein 1 gene and protein in oral squamous cell carcinoma. Chin Med J (Engl). 126:655–658. 2013.PubMed/NCBI

8 

Chen X, Gu W, Wang Q, Fu X, Wang Y, Xu X and Wen Y: C-MYC and BCL-2 mediate YAP-regulated tumorigenesis in OSCC. Oncotarget. 9:668–679. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Yoshikawa K, Noguchi K, Nakano Y, Yamamura M, Takaoka K, Hashimoto-Tamaoki T and Kishimoto H: The Hippo pathway transcriptional co-activator, YAP, confers resistance to cisplatin in human oral squamous cell carcinoma. Int J Oncol. 46:2364–2370. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Zeng G, Xun W, Wei K, Yang Y and Shen H: MicroRNA-27a-3p regulates epithelial to mesenchymal transition via targeting YAP1 in oral squamous cell carcinoma cells. Oncol Rep. 36:1475–1482. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Dong C, Wei KJ, Zhang WB, Sun H, Pan HY and Zhang L: LATS2 induced by TNF-alpha and inhibited cell proliferation and invasion by phosphorylating YAP in oral squamous cell carcinoma. J Oral Pathol Med. 44:475–481. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Fan H, Tian H, Cheng X, Chen Y, Liang S, Zhang Z, Liao Y and Xu P: Aberrant Kank1 expression regulates YAP to promote apoptosis and inhibit proliferation in OSCC. J Cell Physiol. 235:1850–1865. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Nakatani K, Maehama T, Nishio M, Goto H, Kato W, Omori H, Miyachi Y, Togashi H, Shimono Y and Suzuki A: Targeting the Hippo signalling pathway for cancer treatment. J Biochem. 161:237–244. 2017.PubMed/NCBI

14 

Witters LA: The blooming of the French lilac. J Clin Invest. 108:1105–1107. 2001. View Article : Google Scholar : PubMed/NCBI

15 

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 : PubMed/NCBI

16 

Landman GW, Kleefstra N, van Hateren 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 : PubMed/NCBI

17 

Rêgo DF, Pavan LM, Elias ST, De Luca Canto G and Guerra EN: Effects of metformin on head and neck cancer: A systematic review. Oral Oncol. 51:416–422. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Chang PH, Yeh KY, Wang CH, Chen EY, Yang SW, Chou WC and Hsieh JC: Impact of metformin on patients with advanced head and neck cancer undergoing concurrent chemoradiotherapy. Head Neck. 39:1573–1577. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Luo Q, Hu D, Hu S, Yan M, Sun Z and Chen F: In vitro and in vivo anti-tumor effect of metformin as a novel therapeutic agent in human oral squamous cell carcinoma. BMC Cancer. 12:5172012. View Article : Google Scholar : PubMed/NCBI

20 

Chen CH, Tsai HT, Chuang HC, Shiu LY, Su LJ, Chiu TJ, Luo SD, Fang FM, Huang CC and Chien CY: Metformin disrupts malignant behavior of oral squamous cell carcinoma via a novel signaling involving Late SV40 factor/Aurora-A. Sci Rep. 7:13582017. View Article : Google Scholar : PubMed/NCBI

21 

Qi X, Xu W, Xie J, Wang Y, Han S, Wei Z, Ni Y, Dong Y and Han W: Metformin sensitizes the response of oral squamous cell carcinoma to cisplatin treatment through inhibition of NF-κB/HIF-1α signal axis. Sci Rep. 6:357882016. View Article : Google Scholar : PubMed/NCBI

22 

Jivan R, Peres J, Damelin LH, Wadee R, Veale RB, Prince S and Mavri-Damelin D: Disulfiram with or without metformin inhibits oesophageal squamous cell carcinoma in vivo. Cancer Lett. 417:1–10. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Harada K, Ferdous T, Harada T and Ueyama Y: Metformin in combination with 5-fluorouracil suppresses tumor growth by inhibiting the Warburg effect in human oral squamous cell carcinoma. Int J Oncol. 49:276–284. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Alalem M, Ray A and Ray BK: Metformin induces degradation of mTOR protein in breast cancer cells. Cancer Med. 5:3194–3204. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Ben Sahra I, Regazzetti C, Robert G, Laurent K, Le Marchand-Brustel Y, Auberger P, Tanti JF, Giorgetti-Peraldi S and Bost F: Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res. 71:4366–4372. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Akinyeke T, Matsumura S, Wang X, Wu Y, Schalfer ED, Saxena A, Yan W, Logan SK and Li X: Metformin targets c-MYC oncogene to prevent prostate cancer. Carcinogenesis. 34:2823–2832. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Javeshghani S, Zakikhani M, Austin S, Bazile M, Blouin MJ, Topisirovic I, St-Pierre J and Pollak MN: Carbon source and myc expression influence the antiproliferative actions of metformin. Cancer Res. 72:6257–6267. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Liang N and Pende M: YAP enters the mTOR pathway to promote tuberous sclerosis complex. Mol Cell Oncol. 2:e9981002015. View Article : Google Scholar : PubMed/NCBI

29 

Jiang J, Chang W, Fu Y, Gao Y, Zhao C, Zhang X and Zhang S: SAV1 represses the development of human colorectal cancer by regulating the Akt-mTOR pathway in a YAP-dependent manner. Cell Prolif. 50:502017. View Article : Google Scholar

30 

Hansen CG, Ng YL, Lam WL, Plouffe SW and Guan KL: The Hippo pathway effectors YAP and TAZ promote cell growth by modulating amino acid signaling to mTORC1. Cell Res. 25:1299–1313. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Choi W, Kim J, Park J, Lee DH, Hwang D, Kim JH, Ashktorab H, Smoot D, Kim SY, Choi C, et al: YAP/TAZ initiates gastric tumorigenesis via upregulation of MYC. Cancer Res. 78:3306–3320. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Cai J, Song X, Wang W, Watnick T, Pei Y, Qian F and Pan D: A RhoA-YAP-c-Myc signaling axis promotes the development of polycystic kidney disease. Genes Dev. 32:781–793. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Xiao W, Wang J, Ou C, Zhang Y, Ma L, Weng W, Pan Q and Sun F: Mutual interaction between YAP and c-Myc is critical for carcinogenesis in liver cancer. Biochem Biophys Res Commun. 439:167–172. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Turato C, Cannito S, Simonato D, Villano G, Morello E, Terrin L, Quarta S, Biasiolo A, Ruvoletto M, Martini A, et al: SerpinB3 and Yap Interplay Increases Myc Oncogenic Activity. Sci Rep. 5:177012015. View Article : Google Scholar : PubMed/NCBI

35 

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

36 

Lulla AR, Slifker MJ, Zhou Y, Lev A, Einarson MB, Dicker DT and El-Deiry WS: miR-6883 family miRNAs target CDK4/6 to induce G1 phase cell-cycle arrest in colon cancer cells. Cancer Res. 77:6902–6913. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Gulappa T, Reddy RS, Suman S, Nyakeriga AM and Damodaran C: Molecular interplay between cdk4 and p21 dictates G0/G1 cell cycle arrest in prostate cancer cells. Cancer Lett. 337:177–183. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Ren F, Zhang L and Jiang J: Hippo signaling regulates Yorkie nuclear localization and activity through 14-3-3 dependent and independent mechanisms. Dev Biol. 337:303–312. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Park YY, Sohn BH, Johnson RL, Kang MH, Kim SB, Shim JJ, Mangala LS, Kim JH, Yoo JE, Rodriguez-Aguayo C, et al: Yes-associated protein 1 and transcriptional coactivator with PDZ-binding motif activate the mammalian target of rapamycin complex 1 pathway by regulating amino acid transporters in hepatocellular carcinoma. Hepatology. 63:159–172. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Tumaneng K, Schlegelmilch K, Russell RC, Yimlamai D, Basnet H, Mahadevan N, Fitamant J, Bardeesy N, Camargo FD and Guan KL: YAP mediates crosstalk between the Hippo and PI(3)K-TOR pathways by suppressing PTEN via miR-29. Nat Cell Biol. 14:1322–1329. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Safe S, Nair V and Karki K: Metformin-induced anticancer activities: Recent insights. Biol Chem. 399:321–335. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Mynhardt C, Damelin LH, Jivan R, Peres J, Prince S, Veale RB and Mavri-Damelin D: Metformin-induced alterations in nucleotide metabolism cause 5-fluorouracil resistance but gemcitabine susceptibility in oesophageal squamous cell carcinoma. J Cell Biochem. 119:1193–1203. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Damelin LH, Jivan R, Veale RB, Rousseau AL and Mavri-Damelin D: Metformin induces an intracellular reductive state that protects oesophageal squamous cell carcinoma cells against cisplatin but not copper-bis(thiosemicarbazones). BMC Cancer. 14:3142014. View Article : Google Scholar : PubMed/NCBI

44 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Czabotar PE, Lessene G, Strasser A and Adams JM: Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat Rev Mol Cell Biol. 15:49–63. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Henney JE: From the Food and Drug Administration. JAMA. 283:27792000. View Article : Google Scholar : PubMed/NCBI

47 

Wang B, Shao W, Shi Y, Liao J, Chen X and Wang C: Verteporfin induced SUMOylation of YAP1 in endometrial cancer. Am J Cancer Res. 10:1207–1217. 2020.PubMed/NCBI

48 

Lui JW, Xiao S, Ogomori K, Hammarstedt JE, Little EC and Lang D: The efficiency of verteporfin as a therapeutic option in pre-clinical models of melanoma. J Cancer. 10:1–10. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Wei H, Wang F, Wang Y, Li T, Xiu P, Zhong J, Sun X and Li J: Verteporfin suppresses cell survival, angiogenesis and vasculogenic mimicry of pancreatic ductal adenocarcinoma via disrupting the YAP-TEAD complex. Cancer Sci. 108:478–487. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Wang C, Zhu X, Feng W, Yu Y, Jeong K, Guo W, Lu Y and Mills GB: Verteporfin inhibits YAP function through up-regulating 14-3-3σ sequestering YAP in the cytoplasm. Am J Cancer Res. 6:27–37. 2015.PubMed/NCBI

51 

Meng Z, Moroishi T and Guan KL: Mechanisms of Hippo pathway regulation. Genes Dev. 30:1–17. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Zhao B, Li L, Tumaneng K, Wang CY and Guan KL: A coordinated phosphorylation by Lats and CK1 regulates YAP stability through SCF(beta-TRCP). Genes Dev. 24:72–85. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Mossmann D, Park S and Hall MN: mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat Rev Cancer. 18:744–757. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Li SH, Chien CY, Huang WT, Luo SD, Su YY, Tien WY, Lan YC and Chen CH: Prognostic significance and function of mammalian target of rapamycin in tongue squamous cell carcinoma. Sci Rep. 7:81782017. View Article : Google Scholar : PubMed/NCBI

55 

Su W, Wang Y, Wang F, Zhang B, Zhang H, Shen Y and Yang H: Circular RNA hsa_circ_0007059 indicates prognosis and influences malignant behavior via AKT/mTOR in oral squamous cell carcinoma. J Cell Physiol. 234:15156–15166. 2019. View Article : Google Scholar

56 

Zhang X, Liu N, Ma D, Liu L, Jiang L, Zhou Y, Zeng X, Li J and Chen Q: Receptor for activated C kinase 1 (RACK1) promotes the progression of OSCC via the AKT/mTOR pathway. Int J Oncol. 49:539–548. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Artinian N, Cloninger C, Holmes B, Benavides-Serrato A, Bashir T and Gera J: Phosphorylation of the Hippo pathway component AMOTL2 by the mTORC2 kinase promotes YAP signaling, resulting in enhanced glioblastoma growth and invasiveness. J Biol Chem. 290:19387–19401. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Gallant P and Steiger D: Myc's secret life without Max. Cell Cycle. 8:3848–3853. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Pérez-Sayáns M, Suárez-Peñaranda JM, Padín-Iruegas E, Gayoso-Diz P, Reis-De Almeida M, Barros-Angueira F, Gándara-Vila P, Blanco-Carrión A and García-García A: Quantitative determination of c-myc facilitates the assessment of prognosis of OSCC patients. Oncol Rep. 31:1677–1682. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 45 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Zhang Y, Feng X, Tian H, Fu X, Gu W and Wen Y: Metformin inhibits mTOR and c-Myc by decreasing YAP protein expression in OSCC cells. Oncol Rep 45: 1249-1260, 2021
APA
Wang, Y., Zhang, Y., Feng, X., Tian, H., Fu, X., Gu, W., & Wen, Y. (2021). Metformin inhibits mTOR and c-Myc by decreasing YAP protein expression in OSCC cells. Oncology Reports, 45, 1249-1260. https://doi.org/10.3892/or.2020.7909
MLA
Wang, Y., Zhang, Y., Feng, X., Tian, H., Fu, X., Gu, W., Wen, Y."Metformin inhibits mTOR and c-Myc by decreasing YAP protein expression in OSCC cells". Oncology Reports 45.3 (2021): 1249-1260.
Chicago
Wang, Y., Zhang, Y., Feng, X., Tian, H., Fu, X., Gu, W., Wen, Y."Metformin inhibits mTOR and c-Myc by decreasing YAP protein expression in OSCC cells". Oncology Reports 45, no. 3 (2021): 1249-1260. https://doi.org/10.3892/or.2020.7909