Metformin inhibits TGF‑β1‑induced epithelial‑mesenchymal transition and liver metastasis of pancreatic cancer cells

  • Authors:
    • Juichiro Yoshida
    • Takeshi Ishikawa
    • Yuki Endo
    • Shinya Matsumura
    • Takayuki Ota
    • Katsura Mizushima
    • Yasuko Hirai
    • Kaname Oka
    • Tetsuya Okayama
    • Naoyuki Sakamoto
    • Ken Inoue
    • Kazuhiro Kamada
    • Kazuhiko Uchiyama
    • Tomohisa Takagi
    • Yuji Naito
    • Yoshito Itoh
  • View Affiliations

  • Published online on: April 23, 2020     https://doi.org/10.3892/or.2020.7595
  • Pages: 371-381
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epithelial‑mesenchymal transition (EMT) is considered a crucial event in the development of cancer metastasis. Metformin is a drug used in the treatment of type 2 diabetes. Recently, increasing evidence has indicated that metformin possesses anti‑tumor activities. However, the effects of metformin on EMT and metastases in pancreatic cancer remain unknown. Thus, the present study investigated whether metformin inhibits EMT of human pancreatic cancer cell lines. Pancreatic cancer cells were stimulated with transforming growth factor β1 (TGF‑β1), an activator of EMT signaling, with or without metformin. After 48 h, the levels of epithelial and mesenchymal markers were evaluated by western blot analysis, immunocytochemistry and RT‑qPCR. Cancer cell migration was evaluated by an in vitro wound healing assay. The cells stimulated with TGF‑β1 acquired an elongated and fusiform morphology, which was inhibited by metformin. The wound healing assay revealed that metformin significantly suppressed the TGF‑β1‑stimulated migration of pancreatic cancer cells. Following treatment with metformin, E‑cadherin expression (epithelial marker) was upregulated, and the levels of mesenchymal markers were downregulated, which had been increased by TGF‑β1 in these cells. Exposure of the cells to TGF‑β1 activated the Smad2/3 and Akt/mammalian target of rapamycin (mTOR) pathways, and this effect was inhibited by metformin, suggesting that metformin inhibits TGF‑β1‑induced‑EMT through the down‑regulation of the Smad pathway in PANC‑1 cells and the downregulation of the Akt/mTOR pathway in BxPC‑3 cells. In an animal model of surgical orthotopic implantation, metformin inhibited liver metastasis without a significant reduction in the size of the primary pancreatic tumor. On the whole, the findings of the present study suggest that metformin inhibits EMT and cancer metastasis through the Smad or Akt/mTOR pathway.

Introduction

Pancreatic cancer is the fourth most common cause of cancer-related mortality in Japan, and patients with pancreatic cancer have a poor prognosis with an overall 5-year survival rate of <5% (1). Factors contributing to the high mortality rates are late diagnosis due to the lack of early symptoms, a high resistance to treatment and a high invasive potential, which render the tumor surgically incurable. Due to resistance to chemotherapy, radiotherapy and immunotherapy, the radical resection of pancreatic cancer is the only available method that has the potential to cure the disease. Therefore, the inhibition of local recurrence and distant metastasis following surgical resection is crucial.

Longstanding type 2 diabetes is a risk factor for the development of pancreatic cancer due to insulin resistance and associated hyperinsulinemia, hyperglycemia and inflammation are related to the development of cancer (2). Metformin is widely used in the treatment of type 2 diabetes mellitus and has been investigated due to its anti-tumor effects (3). In various types of cancer, metformin has been suggested to exhibit anti-tumor activities and to prevent cancer development (46). Previous studies have also suggested that there is a positive association between metformin and overall survival in pancreatic cancer treatment (7,8). Wan et al reported in a meta-analysis of data from 36,791 patients with pancreatic cancer, that metformin treatment was significantly associated with a favorable overall survival. Subgroup analyses revealed a marked reduction in the mortality risk of patients with stage I–II disease treated with metformin and in patients receiving surgery following treatment with metformin (7). To date, a number of in vitro and in vivo studies have been performed to investigate the anti-tumor activity of metformin (3); however, there are limited studies available to date describing the effects of metformin on epithelial-mesenchymal transition (EMT) and metastasis.

EMT is one of the earliest and most crucial steps in cancer invasion and metastasis (912). The EMT phenotype is characterized by the loss of cell-cell adhesion and apical-basolateral polarity, and a phenotypic change in which cells shift from an epithelial morphology to an elongated fibroblast-like morphology with invasive properties. EMT involves the upregulation of mesenchymal markers, such as vimentin and α-smooth muscle actin (αSMA), and the downregulation of epithelial adhesion molecules, such as E-cadherin and cytokeratins. EMT is triggered by the interplay of extracellular signals (such as collagen) and a number of secreted soluble factors, such as transforming growth factor β1 (TGF-β1), fibroblast growth factor, epidermal growth factor and hepatocyte growth factor. Among these, TGF-β has been identified as the main factor involved in EMT in the tumor microenvironment. TGF-β1 has been shown to activate various downstream pathways, including Smads, Akt/mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK), thereby inducing EMT (9).

Recent studies have suggested that metformin inhibits EMT in several types of cancer (1319). However, the effects of metformin on EMT in pancreatic cancer remain unclear. Hence, the objective of the present study was to clarify whether metformin inhibits the EMT and liver metastasis of pancreatic cancer cells.

Materials and methods

Cell lines and culture

The human pancreatic cell lines, PANC-1 and MIAPaCa-2, were purchased from RIKEN Bioresources Center Cell Bank, BxPC-3 cells were from DS Pharma Biochemical Co., and the mouse pancreatic cancer cell line Panc02 was obtained from the National Cancer Institute. PANC-1, BxPC-3 and Panc02 cells were cultured in Roswell Park Memorial Institute (RPMI)-1640 medium supplemented with 10% fetal bovine serum (FBS), L-glutamine and penicillin (100 U/ml)/streptomycin (100 µg/ml). MIAPaCa-2 cells were cultured in Dulbecco's modified Eagle's medium (DMEM) with low glucose supplemented with 10% fetal bovine serum (FBS), L-glutamine and penicillin (100 U/ml)/streptomycin (100 µg/ml). All cell cultures were maintained at 37°C in a humidified atmosphere containing 5% CO2.

TGF-β1-induced EMT and metformin treatment

After the PANC-1, MIAPaCa-2 and BxPC-3 cells were grown to >80% confluency, they were transferred to a serum-free culture medium with 10 ng/ml TGF-β1 in a humidified 5% CO2 atmosphere at 37°C for the induction of EMT. For metformin treatment, the cells were incubated with 10 mM metformin at 37°C for 48 h (Wako Pure Chemical Industries, Ltd.) prior to stimulation with TGF-β1.

Evaluation of cell viability by WST-8 assay

The cells were seeded into 96-well plates at a density of 5×103 cells/well. The following day, the cells were treated with or without metformin and incubated at 37°C up to 48 h. Cell viability was examined by the addition of 10 µl of 5 mg/ml tetrazolium salt solution to the medium of each well and incubated at 37°C for 4 h, and the absorbance was read at 450 nm with a microplate reader (SpectraMax M2; Molecular Devices, LLC). The absolute values of the absorbance were converted to surviving fraction data and reported as the percentage of living cells relative to the control.

Evaluation of cell migration by wound healing assay

The wound-healing assay was performed as previously described with minor modifications (20,21). Briefly, cells were grown to 100% confluency in P60 culture dishes, and starved in serum-free culture medium for 24 h before scratching. A circle the cell layer of the monolayer approximately 500 µm in diameter was then made by scrapping with a 10-µl extra-long micro-pipette tip. The cells were washed twice with PBS and incubated with serum-free culture medium with or without TGF-β1 and metformin. Microphotographs were acquired with a digital camera at 0, 12 and 24 h after scratching, and the cell-free area was measured using ImageJ software (version 1.51). The migration area was evaluated as the cell-free area at 12 and 24 h, as a percentage of the scratch at 0 h.

Immunocytochemistry

After the PANC-1, MIAPaCA-2, and BxPC-3 cells were grown to >80% confluency in 35-mm µ-dishes (Ibidi), they were washed with PBS and fixed with 4% paraformaldehyde for 20 min at 25°C. The cells were then incubated with mouse anti-human-vimentin (Santa Cruz Biotechnology, Inc.) for 2 h at 25°C followed by 1 h of incubation with anti-rabbit IgG conjugated with Alexa Fluor 594 (Alexa Fluor 594, Life Technologies; Thermo Fisher Scientific, Inc.) at 25°C. The cells were observed using an epi-illumination on a laser scanning confocal microscope (Olympus Corp.).

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

RT-qPCR was performed to detect the mRNA expression levels of E-cadherin, vimentin and α-SMA. Total RNA was extracted from cells using the acid guanidinium phenol chloroform method with Isogen (Nippon Gene Co. Ltd.) from the cultured pancreatic cancer cells (PANC-1, MIAPaCa-2 and BxPC-3). The isolated RNA was stored at −80°C until use in RT-qPCR. Subsequently, complementary DNA (cDNA) was synthesized from reverse-transcribed 1 µg extracted RNA using the High Capacity cDNA Reverse Transcription kit (Applied Biosystems). The expression levels of E-cadherin, vimentin, Snail, ZEB-1 and GAPDH genes were analyzed by the 7300 Real-Time PCR system (Applied Biosystems) using the DNA-binding dye SYBR-Green to detect the PCR products. The relative quantification of gene expression with the RT-qPCR data was performed using the standard curve method, with GAPDH as a reference gene. The PCR conditions were as follows: Denaturation at 95°C for 15 sec, primer annealing and elongation at 60°C for 1 min, followed by melting curve analysis, in which the temperature was increased from 60 to 95°C. The sequences of primers used for RT-qPCR were as follows: E-cadherin sense, 5′-GAAGGTGACAGAGCCTCTGGAT-3′ and antisense, 5′-CATTCCCGTTGGATGACACA-3′; vimentin sense, 5′-ACACCCTGCAATCTTTCAGACA-3′ and antisense, 5′-GATTCCACTTTGCGTTCAAGGT-3′; α-SMA sense, 5′-GACCGAATGCAGAAGGAGAT-3′ and antisense, 5′-CCACCGATCCAGACAGAGTA-3′; GAPDH sense, 5′-ACCACAGTCCATGCCATCACT-3′ and antisense, 5′-CCATCACGCCACAGTTTCC-3′.

Western blot analysis

Cell debris were removed by washing with ice-cold PBS; the cells were lysed in Lysis Buffer (CelLytic M; Sigma-Aldrich; Merck KGaA), scraped and incubated on ice for 15 min. Tissues were frozen using liquid nitrogen, crushed, lysed in lysis buffer, scraped and incubated on ice for 15 min. The supernatants were collected, and total protein was mixed with an SDS sample buffer. The Bradford method was used to measure the protein content; 20 µg protein were used per lane. The protein lysates were then separated by electrophoresis on 10% SDS-PAGE and transblotted to a polyvinylidene fluoride membrane (Atto Corporation). The membrane was blocked with 10% EzBlock (Atto Corporation) in TBS-T [10 mM Tris-HCl (pH 8.0), 150 mM NaCl, 0.1% Tween-20 V/V] for 60 min at room temperature, washed with TBS-T 3 times, and incubated overnight at 4°C with goat anti-mouse-β-actin (ab8229, Abcam), mouse anti-human-E-cadherin (mab1838, R&D Systems, Inc.), mouse anti-mouse-α-SMA (a2547, Sigma-Aldrich; Merck KGaA), mouse anti-human-vimentin (sc32322, Santa Cruz Biotechnology, Inc.), mouse anti-human-Smad2/3 (sc8332, Santa Cruz Biotechnology, Inc.), rabbit anti-human-Snail (3879, Cell Signaling Technology, Inc.), rabbit anti-mouse-phospho-Akt (Ser473) (4058, Cell Signaling Technology, Inc.), rabbit anti-mouse-Akt (4691, Cell Signaling Technology, Inc.), rabbit anti-human-phospho-mTOR (2971, Cell Signaling Technology, Inc.), rabbit anti-human-mTOR (Cell Signaling Technology, Inc. 2983) and rabbit anti-human-phospho-Smad2/3 (8828, Cell Signaling Technology, Inc.) antibodies in TBS-T (diluted 1:500-1:1,000). The membrane was then washed with TBS-T 3 times and incubated with the secondary anti-rabbit (616520, GE Healthcare Japan Corporation), anti-goat (628420, GE Healthcare Japan Corporation) and anti-mouse (054220, GE Healthcare Japan Corporation) IgG antibodies in TBS-T (diluted 1:5,000-1:10,000) for 1 h at room temperature. Immuno-reactive proteins were detected using an ECL-kit (ECL plus, GE Healthcare Bio-Sciences K.K.). The blots were analyzed using ImageJ (version 1.51) software.

Animal models

A total of 33 of C57BL/6 immunocompetent female mice (aged 6–8 weeks; body weight, 15–17 g) were provided by Shimizu. Mice were kept at 18–24°C and 40–70% relative humidity, with a 12-h light/dark cycle. They were provided with free access to water and food (CE-2; CLEA Japan). Cultured Panc02 cells were collected and washed twice with PBS. Panc02 cells (3×106) were injected subcutaneously into both flanks of 3 mice within 30 min of collection. After 3 weeks, subcutaneous tumors growing to a maximum diameter of approximately 10 mm were collected as tumor fragments for subsequent surgical orthotopic implantation (SOI). SOI of tumor fragments was performed in 30 of the C57/BL6 mice, as previously described (22). For implantation, a small 6–10 mm transverse incision was created on the left flank of the mouse. The tail of the pancreas and spleen was carefully exposed through this incision, and a single tumor fragment (3 mm3), harvested from a subcutaneous tumor grown on a mouse, was sewn to the tail of the pancreas using 7-0 nylon surgical sutures (ELP). The pancreatic tail with the tumor fragment and spleen were stored in the abdomen, and the incision was sutured using 4-0 nylon surgical sutures (ELP). Mice were anesthetized with 5% isoflurane during SOI. A total of 15 mice each were used in the control and metformin groups. Mice were randomly assigned to receive either daily peritoneal injections of metformin (125 mg/kg) or normal saline for 28 days. The mice were monitored daily for any signs of toxicity or abnormalities, including their appetite and behavior. Body weight for each animal was measured once a week. Humane euthanasia was performed when mice reached an experimental endpoint or if their ascites increased and they gained >20% of their body weight, or if they were very weak and lost >20% of their body weight. Following SOI, it was difficult to monitor internal tumor growth exactly until sacrifice. Therefore, body weight was used as a humane endpoint index for internal tumor development. At the time of sacrifice, mice were sacrificed by an intraperitoneal injection of pentobarbital sodium overdose (120 mg/kg). All animal experiments were approved by the Institutional Animal Care and Use Committee of Kyoto Prefectural University of medicine under Assurance Number M30-618.

Tumor growth and metastatic pattern analysis

The animals were sacrificed 28 days following SOI. Tumor length, width, height and weight were measured and tumor volume was calculated as follows: Tumor volume = (length × width × height)/2. For the evaluation of metastasis, liver and lung surfaces were observed, and were further pathologically sectioned and metastases were observed under a microscope. The paraffin-embedded liver and lung tissues were sectioned at 4-µm thickness, then hematoxylin and eosin staining was and performed. Briefly, the sections were deparaffinized and rehydrated at 37°C with xylene 3 times for 5 min; then with 100% ethanol 2 times for 10 min each; and finally, in a series of 95, 80 and 70% ethanol for 5 min each. The sections were washed with deionized water for 5 min, and stained with hematoxylin for 5 min at room temperature. The sections were then rinsed with deionized water for 20 min and stained with eosin for 5 min at room temperature. Gradient dehydration was performed with 70, 90, 95 and 100% ethanol for 2, 2, 2 and 10 min, respectively, then with xylene 3 times for 5 min each at room temperature. The sections were sealed with neutral gum and placed in a ventilated room at room temperature overnight. Furthermore, western blot analysis, RT-qPCR and immunohistochemistry were performed for EMT-related markers of the primary tumors in the pancreas.

Immunohistochemistry of vimentin in the primary pancreatic tumors

The paraffin-embedded tumor tissues were sectioned at 4-µm thickness using a microtome cryostat and mounted on MAS-coated slides. Hemo-De was used to clear the sections 3 times for 5 min. The sections were incubated with 100% ethanol for 5 min 3 times, 95% ethanol for 5 min, 90% ethanol for 5 min and 70% ethanol for 5 min to block endogenous peroxidase activity, followed by rinsing with distilled water for 5 min. The slides were submerged in DAKO REAL Target Retrieval solution and then heated in a water bath for 20 min at 95°C for antigen retrieval. After cooling down to room temperature, they were rinsed with distilled water for 5 min; Dako Cytomation protein block (Dako, Tokyo, Japan) was used for 30 min at room temperature to block nonspecific background. The sections were then incubated overnight at 4°C with specific primary antibody against vimentin (sc32322, Santa Cruz Biotechnology, Inc.) diluted at 1:200 with antibody dilution (Dako; Agilent Technologies, Inc.). After washing the sections 3 times in PBS Tween-20 for 7 min, they were incubated with secondary antibody [Histofine Simple Stain mouse MAX PO (rabbit); Nichirei Biosciences, Inc.] for 30 min at room temperature. After removing unbound antibodies by washing 3 times in PBS for 7 min, diaminobenzidine, a chromogen substrate reagent, was used to visualize the bound antibodies. After counterstaining with Mayer's hematoxylin, the sections were washed for 30 min, dehydrated in graded ethanol (70% ethanol for 5 min, 90% ethanol for 5 min, 95% ethanol for 5 min and 100% ethanol for 5 min 3 times), cleared in Hemo-De (3 times for 5 min each) and cover-slipped.

Statistical analysis

All analyses were performed using the JMP ver13.0 (SAS Institute, Inc.). The differences between 2 groups were analyzed with the unpaired Student's t-test and a Chi-square test. Differences between >2 groups were determined by one-way ANOVA followed by Tukey's multiple comparison test to compare the mean values. All data are presented as the means ± standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

Metformin inhibits morphological changes consistent with EMT in the presence of TGF-β1

Cell was examined using the WST-8 assay. Cell viability was not significantly altered by treatment with 10 mM metformin compared to that in the untreated group (data not shown). Therefore, the concentration of 10 mM metformin was used for the subsequent experiments. Following the addition of 10 ng/ml TGF-β1 for 48 h, the PANC-1 and BxPC-3 pancreatic cancer cells acquired an elongated and fusiform morphology, suggesting that 10 ng/ml TGF-β1 was sufficient to induce EMT. Metformin treatment at 1 h prior to TGF-β1 exposure inhibited these morphological changes (Fig. 1). The MIAPaCA-2 cell line also an acquired elongated morphology after TGF-β1 exposure, and metformin treatment suppressed these morphological changes. However, the morphological changes of this cell line were smaller than other cell lines (data not shown).

Metformin suppresses the migratory potential of PANC-1 and MIAPaCa-2 cells

To examine the effect of metformin treatment on the migratory capability of PANC-1 and MIAPaCa-2 cells, wound healing assays were performed; at 24 h after scratching, cell migration into the wound was visualized using a light microscope and images were captured. The PANC-1 cells treated with TGF-β1 migrated significantly more rapidly than the control cells after 24 h. Metformin reduced wound healing in cells treated with TGF-β1 during the same period (Fig. 2A). Similar results were observed in the MIAPaCa-2 cells. However, the TGF-β1-induced migration of MIAPaCa-2 cells was weaker than in that of the PANC-1 cells. As a result, the difference in the migrated area between the TGF-β1-treated group and the metformin + TGF-β1-treated group in the MIAPaCa-2 cells was smaller than that for the PANC-1 cells; thus, the cell images for the MIAPaCa-2 are not shown (Fig. 2B). Wounds could not be made accurately in the BxPC-3 cells due to their strong adhesion to the plate, and therefore the wound healing assay in these cells could not be evaluated.

Effect of metformin on molecular markers of EMT

Western blot analysis and RT-qPCR for E-cadherin were performed to determine the effects of metformin on EMT. The results of western blot analysis revealed no marked changes in the expression of E-cadherin following exposure to TGF-β1, and an increase in E-cadherin expression was observed following metformin treatment in the BxPC3 cells (Fig. 3A). However, E-cadherin protein expression was not observed even in the absence of TGF-β1 in either the PANC-1 or MIAPaCa-2 cells (data not shown). RT-qPCR analysis revealed a decrease in the mRNA expression of E-cadherin following TGF-β1 exposure, which was blocked by metformin treatment in the PANC-1 cells (Fig. 3B). The mRNA expression of E-cadherin was not downregulated by TGF-β1 exposure in the BxPC3 cells, but was upregulated with a combination of TGF-β1 and metformin treatment (Fig. 3B).

Subsequently, immunocytochemistry, western blot analysis and RT-qPCR of mesenchymal markers (i.e., vimentin and α-SMA) and the EMT transcription factor, Snail, were performed. Immunofluorescence staining for vimentin revealed a strong expression following exposure to TGF-β1 for 48 h, and a stronger expression was observed at the tip of spindle-shaped cells, which was reversed by metformin treatment in the PANC-1 cells (Fig. 4A). Vimentin protein expression was observed only in PANC-1 cells using immunocytochemistry. As shown by western blot analysis, the expression of vimentin and Snail was significantly upregulated by TGF-β1 treatment, which was blocked by metformin treatment in the PANC-1 cells (Fig. 4B). In the BxPC-3 cells, vimentin expression was not observed (data not shown); however, the expression of α-SMA was upregulated by TGF-β1 treatment, which was blocked by metformin treatment. Furthermore, Snail expression was downregulated by the combination of TGF-β1 and metformin treatment (Fig. 4B). The mRNA expression of vimentin was significantly upregulated by TGF-β1 treatment, which was blocked by metformin treatment (Fig. 4C). Exposure to TGF-β1 slightly increased the expression of αSMA, although the change was not statistically significant, and this slight increase in expression was blocked by metformin treatment in BxPC-3 cells (Fig. 4C).

Effect of metformin on the Smad and Akt/mTOR pathways

To elucidate the mechanisms through which metformin inhibits TGF-β1-induced EMT, the levels of phosphorylation of Smad2/3, Akt, and mTOR were examined in the pancreatic cell lines. First, the role of metformin in the regulation of Smad2/3 expression and phosphorylation, which is involved in the downstream signaling pathway of TGF-β1, was investigated by western blot analysis. In the PANC-1 cells, exposure to TGF-β1 resulted in the phosphorylation of Smad2/3, which was blocked by metformin treatment (Fig. 5A). However, the exposure of BxPC-3 cells, which have a naturally low expression of Smad2/3, to TGF-β1 and metformin did not alter the phosphorylation of Smad2/3 (Fig. 5A). Subsequently, other potential pathways of TGF-β1-induced EMT were investigated. The role of metformin in regulating Akt and mTOR expression and phosphorylation is involved in the downstream signaling pathway of TGF-β1. The exposure of the PANC-1 cells to TGF-β1 did not result in the phosphorylation of Akt and mTOR, suggesting that this pathway is not involved in TGF-β1-induced EMT in these cells. On the other hand, in the BxPC-3 cells, exposure to TGF-β1 resulted in the phosphorylation of Akt and mTOR. Metformin treatment decreased the expression of Akt. Moreover, in the BxPC cells, metformin treatment decreased the phosphorylation of Akt primarily due to a reduction in total AKT protein levels. Metformin treatment also inhibited mTOR phosphorylation (Fig. 5B).

Metformin suppresses tumor growth and metastasis in vivo

Pancreatic tumors survived and grew in the pancreatic tails of all sacrificed mice in the present study. A total of 2 mice in the metformin group and 1 mouse in the control group were euthanized due to a >20% weight gain due to ascite retention. A total of 2 mice in the metformin group died within 3 days following SOI, presumably due to suture failure. Consequently, 14 mice in the control group and 11 mice in the metformin group were evaluated for metastasis. Primary pancreatic tumors tended to be smaller in both size and weight in the metformin group than in the control group, but this was not statistically significant (Table I) (Fig. 6A). For evaluation of metastasis, we observed liver and lung surfaces, and further sectioned to evaluate the metastasis microscopically (Fig. 6B). Liver metastasis was observed in 15 out of 25 mice. All mice with liver metastases had multiple tumors (range 3–20). In the control group, liver metastasis was observed in 78.6% (11/14) of the mice, whereas in the metformin group, metastasis was observed in only 36.4% (4/11). This suppression of metastasis by metformin treatment was statistically significant (P=0.049, Table I). Lung metastasis was observed in 5 out of 25 mice, and although the frequency of lung metastasis was slightly higher in the control group (28.6% vs. 9.1%), it did not reach statistical significance (Table I). Subsequently, the levels of EMT-related markers in the primary pancreatic tumors in 12 mice were measured. RT-qPCR analysis revealed that E-cadherin expression tended to increase and vimentin expression tended to decrease with metformin treatment (Fig. 6C), although neither reached statistical significance (P=0.07 and P=0.07, respectively). As shown by western blot analysis and immunohistochemical analysis, the expression of vimentin in the metformin group was significantly lower than that in the control group (Fig. 6D and E).

Table I.

Tumor growth and metastatic pattern in mice administered metformin and control mice.

Table I.

Tumor growth and metastatic pattern in mice administered metformin and control mice.

Control (n=14)Metformin (n=11)P-value
Body weight at SOI (g)19.9±1.220.0±0.90.835
Body weight at sacrifice (g)21.0±1.720.9±1.50.861
Tumor growth
  yes14111.000
  no00
Tumor volume (mm3)601.4±179.2492.1±262.30.249
Tumor weight (mg)456.5±182.9368.1±212.20.295
Liver metastasis
  yes1140.049
  no37
Lung metastasis
  yes410.341
  no1010

Discussion

EMT is a crucial biological process in cancer invasion and metastasis. However, the details of the mechanisms of EMT in pancreatic cancer and how this process can be suppressed remain to be explored. The present study focused on whether metformin, widely used in the treatment of type 2 diabetes mellitus, can inhibit the EMT of pancreatic cancer cells. The results indicated that metformin suppressed the TGF-β1-induced EMT of pancreatic cells and inhibited liver metastasis in vivo. Moreover, the results strongly suggested that metformin inhibited EMT by inhibiting the TGF-β signaling pathways (i.e., the Smad2/3 and Akt/mTOR pathways). To the best of our knowledge, this is the first study to demonstrate that metformin inhibits EMT and metastasis formation by suppressing the intracellular TGF-β signaling pathways in pancreatic cancer.

Recently, metformin has attracted attention due to its anti-tumor effects (3). Strong associations between metformin use and the reduced risk of several cancer types, including pancreatic cancer have been found in diabetic patients (4,23). Preclinical studies have suggested that metformin can inhibit the growth of pancreatic cancer both in vitro and in vivo (24,25). Metformin systemically ameliorates hyperinsulinemia, hyperglycemia and hyperlipidemia, which are involved in both cancer initiation and progression. An antitumor effect of metformin is triggered when several signals, such as phosphoinositide 3-kinase (PI3K) and MAPK are suppressed (2628). Metformin also inhibits the mTOR pathway and its downstream substrates (29,30), reduces cyclin D1 expression and cell cycle arrest in a concentration-dependent manner (31), suppresses angiogenesis through the reduction of vascular endothelial growth factor (VEGF) expression (32), and induces apoptosis by p53 activation (3,33).

Recent studies have suggested that metformin inhibits EMT in several types of cancer (1319). The results of these studies suggest that there are several mechanisms through which metformin inhibits EMT. Zhao et al reported that metformin inhibited interleukin (IL)-6-induced EMT in colorectal cancer by blocking signal transducer and activator of transcription 3 (STAT3) phosphorylation (17). Metformin has also been reported to inhibit EMT in prostate cancer through the COX2/PGE2/STAT3 axis (15). TGF-β1 acts as a potent driver of cancer progression through the induction of EMT in various types of cancer. In TGF-β-induced EMT, both the Smad and non-Smad pathways (i.e., Akt/mTOR and MAP kinase pathways) are activated, thereby increasing EMT-related gene expression (10,3438). Previous studies have demonstrated that metformin blocks the Smad signaling pathway and inhibits TGF-β-induced EMT (19,39,40). On the other hand, metformin has also been reported to block TGF-β1-induced EMT through non-Smad pathways, such as the Akt/mTOR/Zeb1 pathway (16). Thus, there are several mechanisms responsible for the inhibition of EMT by metformin, depending on the type of cell used, the EMT inducer, and others.

To the best of our knowledge, until recently, there were no studies available on the effect of metformin on EMT in pancreatic cancer. However, Duan et al demonstrated that metformin reduced TGF-β1 production in pancreatic cells, resulting in the inhibition of autocrine TGF-β1 signaling, thereby inhibiting EMT (41). However, the effect of metformin on TGF-β1 signaling pathways was not investigated in this previous study. In pancreatic cancer, there have been no studies published to date on the effect of metformin on the TGF-β-induced Smad and non-Smad signaling pathways involved in EMT, at least to the best of our knowledge. The results of the present study demonstrated that metformin inhibited TGF-β1-induced-EMT through the downregulation of the Smad pathway in PANC-1 cells and the downregulation of the Akt/mTOR pathway in BxPC-3 cells. These findings suggest that metformin inhibits TGF-β1-induced-EMT through the downregulation of different pathways, depending on the cell line, and to the best of our knowledge, this is the first report to demonstrate an inhibition of TGF-β1-induced-EMT by metformin through the downregulation of intracellular TGF-β1 signaling pathways in pancreatic cancer cells.

Of note, it was found that metformin inhibited EMT in vitro and liver metastasis in vivo at clinical doses. In the present study, metformin was administered to mice daily by intraperitoneal injection at 125 mg/kg, which is equivalent to the human dose of 600 mg/average size individual of 60 kg (42). Since the dose of metformin for patients with type 2 diabetes mellitus is up to 2,250 mg/individual in Japan (43), it is noteworthy that metformin has the potential to suppress cancer metastasis at relatively low doses used clinically. In the present study, an orthotopic tumor implantation model was used instead of a spontaneous metastasis model. Focusing on the process of metastasis formation, it was considered that this model would be more suitable for the purposes of the present study to assess the effect of metformin, which inhibits cancer cell proliferation, on cancer metastasis. Furthermore, it was confirmed that metformin suppressed liver metastasis without a significant decrease in the primary pancreatic lesion.

There are a few limitations to the present study. Although the assessment of motility is crucial for the assessment of EMT, for the BxPC cells, this assay was not technically feasible due to the potent adhesive force. Alternatively, other assays, such as invasion assays, could overcome this limitation. For proteins with a very low protein content, such as the phosphorylation of Smad, mTOR and Akt, multiple western blot analyses could not be performed as each assay required too many cells. As a result, statistical analysis could not be performed. In in vivo experiments, there were euthanized and dead mice in both groups prior to the analysis for metastasis. Although the present study demonstrated that metformin suppressed liver metastases, whether the long-term use of metformin in tumor-bearing mice improves survival needs to be confirmed in future studies. In addition, in the present study, the number of metastases could not be accurately compared due to the presence of a number of small metastatic foci. The use of Pan02 cells engineered to express luciferase may contribute to the accurate assessment of metastases.

In conclusion, the present study demonstrates that metformin suppresses TGF-β1-induced EMT in pancreatic cells and inhibits liver metastasis in vivo. Moreover, the results demonstrated that the process of inhibiting EMT is mediated through the Smad2/3 or Akt/mTOR pathway. Based on these findings, metformin is expected to be clinically useful for the suppression of cancer metastasis in patients with pancreatic cancer. To verify the inhibitory effects of metformin on cancer metastasis, prospective clinical studies, such as those in adjuvant settings are necessary.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Grant-in-Aid for Scientific Research (KAKENHI C) (17K09314) from the Japan Society for the Promotion of Science (JSPS). YI received a research fee from Takeda Pharmaceutical Co., Osaka, Japan, Sumitomo Dainippon Pharmaceutical Co., Osaka Japan, Daiichi Sankyo Co., Tokyo Japan, and Pfizer, Tokyo, Japan. YN received lecture fees from Takeda Pharmaceutical Co. Neither the funding agency nor any outside organization participated in the study design or have any competing interest.

Availability of data and materials

All data generated or analyzed during this study are included in this published article or are available from the corresponding author on reasonable request.

Authors' contributions

TI, YN and YI conceived and designed the study and modified the manuscript. JY collected and analyzed the data, designed and performed the experiments, and drafted the manuscript. JY, TI, TeO, NS, KI, KK, KU, and TT performed the cell culture and wound healing assay. JY, TI, YE, SM, TaO, KM and KO performed western blot analysis, RT-qPCR and in vivo experiments. JY and YH performed the immunocytochemistry and immunohistochemistry experiments. All authors participated in revising the manuscript, read and approved the final manuscript.

Ethics approval and consent to participate

All animal experiments were approved by the Institutional Animal Care and Use Committee of Kyoto Prefectural University of medicine under Assurance Number M30-618.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wolfgang CL, Herman JM, Laheru DA, Klein AP, Erdek MA, Fishman EK and Hruban RH: Recent progress in pancreatic cancer. CA Cancer J Clin. 63:318–348. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Andersen DK, Korc M, Petersen GM, Eibl G, Li D, Rickels MR, Chari ST and Abbruzzese J: Diabetes, pancreatogenic diabetes, and pancreatic cancer. Diabetes. 66:1103–1110. 2017. View Article : Google Scholar : PubMed/NCBI

3 

He H, Ke R, Lin H, Ying Y, Liu D and Luo Z: Metformin, an old drug, brings a new era to cancer therapy. Cancer J. 21:70–74. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Franciosi M, Lucisano G, Lapice E, Strippoli GF, Pellegrini F and Nicolucci A: Metformin therapy and risk of cancer in patients with type 2 diabetes: Systematic review. PLoS One. 8:e715832013. View Article : Google Scholar : PubMed/NCBI

5 

Zhang P, Li H, Tan X, Chen L and Wang S: Association of metformin use with cancer incidence and mortality: A meta-analysis. Cancer Epidemiol. 37:207–218. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Bayraktar S, Hernadez-Aya LF, Lei X, Meric-Bernstam F, Litton JK, Hsu L, Hortobagyi GN and Gonzalez-Angulo AM: Effect of metformin on survival outcomes in diabetic patients with triple receptor-negative breast cancer. Cancer. 118:1202–1211. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Wan G, Sun X, Li F, Wang X, Li C, Li H, Yu X and Cao F: Survival benefit of metformin adjuvant treatment for pancreatic cancer patients: A systematic review and meta-analysis. Cell Physiol Biochem. 49:837–847. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Broadhurst PJ and Hart AR: Metformin as an adjunctive therapy for pancreatic cancer: A review of the literature on its potential therapeutic use. Dig Dis Sci. 63:2840–2852. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Yang J and Weinberg RA: Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev Cell. 14:818–829. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Jie XX, Zhang XY and Xu CJ: Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: Mechanisms and clinical applications. Oncotarget. 8:81558–81571. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Okajima M, Kokura S, Ishikawa T, Mizushima K, Tsuchiya R, Matsuyama T, Adachi S, Okayama T, Sakamoto N, Kamada K, et al: Anoxia/reoxygenation induces epithelial-mesenchymal transition in human colon cancer cell lines. Oncol Rep. 29:2311–2317. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Wang Y, Wu Z and Hu L: The regulatory effects of metformin on the [SNAIL/miR-34]:[ZEB/miR-200] system in the epithelial-mesenchymal transition(EMT) for colorectal cancer(CRC). Eur J Pharmacol. 834:45–53. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Valaee S, Yaghoobi MM and Shamsara M: Metformin inhibits gastric cancer cells metastatic traits through suppression of epithelial-mesenchymal transition in a glucose-independent manner. PLoS One. 12:e01744862017. View Article : Google Scholar : PubMed/NCBI

15 

Tong D, Liu Q, Liu G, Xu J, Lan W, Jiang Y, Xiao H, Zhang D and Jiang J: Metformin inhibits castration-induced EMT in prostate cancer by repressing COX2/PGE2/STAT3 axis. Cancer Lett. 389:23–32. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Song Y, Chen Y, Li Y, Lyu X, Cui J, Cheng Y, Zhao L and Zhao G: Metformin inhibits TGF-beta1-induced epithelial-to-mesenchymal transition-like process and stem-like properties in GBM via AKT/mTOR/ZEB1 pathway. Oncotarget. 9:7023–7035. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Zhao Z, Cheng X, Wang Y, Han R, Li L, Xiang T, He L, Long H, Zhu B and He Y: Metformin inhibits the IL-6-induced epithelial-mesenchymal transition and lung adenocarcinoma growth and metastasis. PLoS One. 9:e958842014. View Article : Google Scholar : PubMed/NCBI

18 

Nakayama A, Ninomiya I, Harada S, Tsukada T, Okamoto K, Nakanuma S, Sakai S, Makino I, Kinoshita J, Hayashi H, et al: Metformin inhibits the radiation-induced invasive phenotype of esophageal squamous cell carcinoma. Int J Oncol. 49:1890–1898. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Li NS, Zou JR, Lin H, Ke R, He XL, Xiao L, Huang D, Luo L, Lv N and Luo Z: LKB1/AMPK inhibits TGF-β1 production and the TGF-beta signaling pathway in breast cancer cells. Tumour Biol. 37:8249–8258. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Uchiyama K, Naito Y, Takagi T, Mizushima K, Hayashi N, Harusato A, Hirata I, Omatsu T, Handa O, Ishikawa T, et al: Carbon monoxide enhance colonic epithelial restitution via FGF15 derived from colonic myofibroblasts. Biochem Biophys Res Commun. 391:1122–1126. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Kimura-Tsuchiya R, Ishikawa T, Kokura S, Mizushima K, Adachi S, Okajima M, Matsuyama T, Okayama T, Sakamoto N, Katada K, et al: The inhibitory effect of heat treatment against epithelial-mesenchymal transition (EMT) in human pancreatic adenocarcinoma cell lines. J Clin Biochem Nutr. 55:56–61. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Hwang HK, Murakami T, Kiyuna T, Kim SH, Lee SH, Kang CM, Hoffman RM and Bouvet M: Splenectomy is associated with an aggressive tumor growth pattern and altered host immunity in an orthotopic syngeneic murine pancreatic cancer model. Oncotarget. 8:88827–88834. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Li D, Yeung SC, Hassan MM, Konopleva M and Abbruzzese JL: Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology. 137:482–488. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Shi Y, He Z, Jia Z and Xu C: Inhibitory effect of metformin combined with gemcitabine on pancreatic cancer cells in vitro and in vivo. Mol Med Rep. 14:2921–2928. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Kisfalvi K, Moro A, Sinnett-Smith J, Eibl G and Rozengurt E: Metformin inhibits the growth of human pancreatic cancer xenografts. Pancreas. 42:781–785. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Algire C, Amrein L, Bazile M, David S, Zakikhani M and Pollak M: Diet and tumor LKB1 expression interact to determine sensitivity to anti-neoplastic effects of metformin in vivo. Oncogene. 30:1174–1182. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Jalving M, Gietema JA, Lefrandt JD, de Jong S, Reyners AK, Gans RO and de Vries EG: Metformin: Taking away the candy for cancer? Eur J Cancer. 46:2369–2380. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Schneider MB, Matsuzaki H, Haorah J, Ulrich A, Standop J, Ding XZ, Adrian TE and Pour PM: Prevention of pancreatic cancer induction in hamsters by metformin. Gastroenterology. 120:1263–1270. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Muniraj T and Chari ST: Diabetes and pancreatic cancer. Minerva Gastroenterol Dietol. 58:331–345. 2012.PubMed/NCBI

30 

Yue W, Yang CS, DiPaola RS and Tan XL: Repurposing of metformin and aspirin by targeting AMPK-mTOR and inflammation for pancreatic cancer prevention and treatment. Cancer Prev Res (Phila). 7:388–397. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Ben Sahra I, Laurent K, Loubat A, Giorgetti-Peraldi S, Colosetti P, Auberger P, Tanti JF, Le Marchand-Brustel Y and Bost F: The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene. 27:3576–3586. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Lund SS, Tarnow L, Stehouwer CD, Schalkwijk CG, Teerlink T, Gram J, Winther K, Frandsen M, Smidt UM, Pedersen O, et al: Impact of metformin versus repaglinide on non-glycaemic cardiovascular risk markers related to inflammation and endothelial dysfunction in non-obese patients with type 2 diabetes. Eur J Endocrinol. 158:631–641. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Buzzai M, Jones RG, Amaravadi RK, Lum JJ, DeBerardinis RJ, Zhao F, Viollet B and Thompson CB: Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res. 67:6745–6752. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Zhang YE: Non-Smad pathways in TGF-beta signaling. Cell Res. 19:128–139. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Hoot KE, Lighthall J, Han G, Lu SL, Li A, Ju W, Kulesz-Martin M, Bottinger E and Wang XJ: Keratinocyte-specific Smad2 ablation results in increased epithelial-mesenchymal transition during skin cancer formation and progression. J Clin Invest. 118:2722–2732. 2008.PubMed/NCBI

36 

Lamouille S and Derynck R: Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 178:437–451. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Lamouille S, Connolly E, Smyth JW, Akhurst RJ and Derynck R: TGF-β-induced activation of mTOR complex 2 drives epithelial-mesenchymal transition and cell invasion. J Cell Sci. 125:1259–1273. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Julien S, Puig I, Caretti E, Bonaventure J, Nelles L, van Roy F, Dargemont C, de Herreros AG, Bellacosa A and Larue L: Activation of NF-kappaB by Akt upregulates Snail expression and induces epithelium mesenchyme transition. Oncogene. 26:7445–7456. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Shin HS, Ko J, Kim DA, Ryu ES, Ryu HM, Park SH, Kim YL, Oh ES and Kang DH: Metformin ameliorates the phenotype transition of peritoneal mesothelial cells and peritoneal fibrosis via a modulation of oxidative stress. Sci Rep. 7:56902017. View Article : Google Scholar : PubMed/NCBI

40 

Thakur S, Viswanadhapalli S, Kopp JB, Shi Q, Barnes JL, Block K, Gorin Y and Abboud HE: Activation of AMP-activated protein kinase prevents TGF-β1-induced epithelial-mesenchymal transition and myofibroblast activation. Am J Pathol. 185:2168–2180. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Duan W, Qian W, Zhou C, Cao J, Qin T, Xiao Y, Cheng L, Li J, Chen K, Li X, et al: Metformin suppresses the invasive ability of pancreatic cancer cells by blocking autocrine TGF-β1 signaling. Oncol Rep. 40:1495–1502. 2018.PubMed/NCBI

42 

Tan XL, Bhattacharyya KK, Dutta SK, Bamlet WR, Rabe KG, Wang E, Smyrk TC, Oberg AL, Petersen GM and Mukhopadhyay D: Metformin suppresses pancreatic tumor growth with inhibition of NFkappaB/STAT3 inflammatory signaling. Pancreas. 44:636–647. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Odawara M, Kawamori R, Tajima N, Iwamoto Y, Kageyama S, Yodo Y, Ueki F and Hotta N: Long-term treatment study of global standard dose metformin in Japanese patients with type 2 diabetes mellitus. Diabetol Int. 8:286–295. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 44 Issue 1

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
Yoshida J, Ishikawa T, Endo Y, Matsumura S, Ota T, Mizushima K, Hirai Y, Oka K, Okayama T, Sakamoto N, Sakamoto N, et al: Metformin inhibits TGF‑β1‑induced epithelial‑mesenchymal transition and liver metastasis of pancreatic cancer cells. Oncol Rep 44: 371-381, 2020
APA
Yoshida, J., Ishikawa, T., Endo, Y., Matsumura, S., Ota, T., Mizushima, K. ... Itoh, Y. (2020). Metformin inhibits TGF‑β1‑induced epithelial‑mesenchymal transition and liver metastasis of pancreatic cancer cells. Oncology Reports, 44, 371-381. https://doi.org/10.3892/or.2020.7595
MLA
Yoshida, J., Ishikawa, T., Endo, Y., Matsumura, S., Ota, T., Mizushima, K., Hirai, Y., Oka, K., Okayama, T., Sakamoto, N., Inoue, K., Kamada, K., Uchiyama, K., Takagi, T., Naito, Y., Itoh, Y."Metformin inhibits TGF‑β1‑induced epithelial‑mesenchymal transition and liver metastasis of pancreatic cancer cells". Oncology Reports 44.1 (2020): 371-381.
Chicago
Yoshida, J., Ishikawa, T., Endo, Y., Matsumura, S., Ota, T., Mizushima, K., Hirai, Y., Oka, K., Okayama, T., Sakamoto, N., Inoue, K., Kamada, K., Uchiyama, K., Takagi, T., Naito, Y., Itoh, Y."Metformin inhibits TGF‑β1‑induced epithelial‑mesenchymal transition and liver metastasis of pancreatic cancer cells". Oncology Reports 44, no. 1 (2020): 371-381. https://doi.org/10.3892/or.2020.7595