Abstract

Metformin is one of the most widely used anti-diabetic drugs in type-II diabetes treatment. The mechanism of decreasing blood glucose is believed to suppress hepatic gluconeogenesis by increasing muscular glucose uptake and insulin sensitivity. Recent studies suggest that metformin may reduce cancer risk; however, its anticancer mechanism in gastric cancers remains unclear. Here, we aim to evaluate the anticancer effects of metformin on human gastric adenocarcinoma (AGS) cells. Our results showed that metformin inhibited AGS cell proliferation in a dose-dependent manner. Using small-scale quantitative proteomics, we identified 177 differentially expressed proteins upon metformin treatment; among these, nine proteins such as 26S proteasome non-ATPase regulatory subunit 2 (PSMD2), stress-induced phosphoprotein 1 (STIP1), and adenylyl cyclase-associated protein 1 (CAP1) were significantly altered. We found that metformin induced cell cycle arrest at the G0/G1 phase, suppressed cell migration, and affected cytoskeleton distribution. Additionally, patients with highly expressed PSMD2, STIP1, and CAP1 have a poor clinical outcome. Our study provides a novel view of developing therapies for gastric cancer.
Introduction
Gastric cancer is the third common cancer in the world, accounting for 8% of total cancer and 10% of total deaths.1 Gastric cancer is asymptomatic in early stages. Most of the patients are diagnosed with gastric cancer at the terminal stage of cancer. Therefore, it is one of the reasons why the 5 yr survival rate of gastric cancer patients is less than 30%.2 Surgical resection is still the major curative treatment for gastric cancer; however, the survival rate remains low due to rapid metastasis and high recurrence rates. The recurrence rate of gastric cancer is 40–70%,3 and it is diagnosed within 20–28 months.4 Additional perioperative chemotherapy or adjuvant chemoradiation have shown a poor survival rate in gastric cancer;5 therefore, new therapies are urgently required.
Metformin is a biguanide derivative that originates from goat’s rue. Now, it serves as the first line and widely used oral anti-diabetic drug in the treatment of type-II diabetes. A previous study suggests that metformin can reduce metabolic risk factors and sensitize insulin receptors, especially in overweight youths with type-I diabetes.6,7 Metformin decreases the blood glucose level through increasing muscular glucose uptake, reducing glucose production in the liver, and enhancing insulin receptor sensitivity.8−11 Thus, it regulates hepatic gluconeogenesis and glycogenolysis. In recent years, the relationship between metformin and cancer has been discussed in several studies. Evans et al. found that metformin may reduce the risk of cancer in a case-control study.12 Several epidemiological studies also suggest that diabetic patients who use metformin have a lower cancer incidence than those who use other anti-diabetic treatments.7,13 Recent studies show that metformin inhibits cancer cell proliferation in the breast,14 lung,15 and colon16 cancer cell lines. Some studies indicate that metformin mediates the activation of 5’ adenosine monophosphate-activated protein kinase (AMPK) and results in the inhibition of breast cancer cell and intestinal polyp growth with decreased mTOR and S6K activity, which play an essential role in protein synthesis.17,18 Metformin has the ability to inhibit cancer viability through the inhibition of mTOR in an AMPK-independent manner.19 Therefore, metformin treatment in cancer research has become a suitable and interesting topic due to its safety, low cost, and the property of modulating energy metabolism.20
Most of the biological functions, regulatory switches, and signal transfections are controlled by multiple proteins rather than a single one.21 Proteomics can provide an integrative view of protein characterization and quantification in organisms under defined conditions.22 Therefore, proteomics is applied in many fields such as drug discovery,23−25 cancer therapy,26,27 and uncovering chemical-induced biological mechanisms.28−30 One of the proteomic techniques is called isobaric tagging for relative and absolute quantitation (iTRAQ), which was developed in 2004.31 iTRAQ is a technique that applied a multiplexed isobaric chemical tag, which allows labeling four groups of protein samples at a time. Relative quantification is achieved by calculating the peak areas for either four MS/MS reporter ions (ion range from 114 to 117 Da).32 In this study, we applied iTRAQ to identify and quantify metformin-regulated proteins. Furthermore, we performed several functional assays to show that metformin-regulated proteins were involved in those biological functions (Figure 1).
Figure 1.

Experimental design of this study. AGS cells were treated with metformin, proteins were labeled with isobaric tags for absolute quantitation, analyzed by mass spectrometry, and then validated.
Results and Discussion
Metformin Inhibits the Proliferation of Human Gastric Cancer AGS Cells
To determine whether metformin affects human gastric cancer cell growth, we analyzed the effect of metformin on cell proliferation using a human gastric cancer cell line, AGS. Cells were treated with different concentrations of metformin for 24, 48, and 72 h. As shown in Figure 2A, metformin inhibits the cell proliferation in a dose-dependent manner. We also observed similar results in the colony formation assay (Figure 2B). To further examine whether metformin inhibition of cell proliferation is reversible, we changed the medium after metformin treatment for 24 h (Figure 2C). We observed that cell proliferation was still suppressed when metformin had been removed (Figure 2D). The results suggest that metformin might irreversibly inhibit AGS cell proliferation. Together, metformin inhibits the AGS cell proliferation, and the effect might be irreversible.
Figure 2.
Metformin dose-dependently inhibits AGS cell proliferation and anchorage-dependent growth. (A) MTT assay results in which AGS cells were treated with different concentrations of metformin. (B) Colony formation assay results in which AGS cells were treated with 10 mM metformin or control for 7 days. The number of colonies was counted in triplicate experiments. Data are presented as mean ± S.D. (n = 3). * and ** indicate that p value <0.05 and <0.001, respectively. (C) Experimental design to check whether metformin-induced effect is reversible. Cells were seeded for 24 h and then the medium was changed with complete medium and medium containing metformin. For the control group and one of the Met-pretreated groups, the medium was changed with fresh complete medium after 48 h. For the Met-pretreated + Met group, the medium was changed with a medium that contained metformin after 48 h. (D) After 48 h of metformin treatment, AGS cells were incubated with or without metformin and analyzed for the cell proliferation using RTCA with an E-plate for another 72 h. Control: cells without any metformin treatment; Met-pretreated cells: cells pretreated with metformin for 48 h and then removed from metformin exposure; Met-pretreated cells + Met: cells pre-treated with metformin for 48 h and incubated in metformin. The x-axis indicates the normalized cell index and y-axis indicates the time in hour. Data are presented as mean ± S.D. (n = 3).
iTRAQ-Based Proteomics Reveals Metformin-Regulated Proteins
To investigate the differential proteins induced by metformin, we performed the iTRAQ-based quantitative proteomic approach, and the workflow is illustrated in Figure 3A. Cells were treated with metformin or control for 72 h. Proteins extracted from metformin-treated and control AGS cells were digested to peptides and labeled with iTRAQ. Duplicates of control and treatment showed a high correlation in iTRAQ analysis. The correlation coefficient R2 was 0.9206 between the control duplicates and 0.9461 between the treatment duplicates (Figure 3B,C).
Figure 3.
iTRAQ-based quantitative proteome in control and metformin-treated AGS cells. (A) Schematic representation of the experimental strategy for quantitative global proteomic profiling in response to metformin in AGS cells. Protein extracts obtained from the transfected cells were digested, iTRAQ-labeled, SCX-fractionated, and analyzed by mass spectrometry. (B,C) iTRAQ analysis showing high reproducibility in the duplicate sample. The correlation of iTRAQ intensity between duplicate samples (control: iTRAQ114 and iTRAQ115; metformin: iTRAQ116 and iTRAQ117). (D) Distribution of protein ratio in the control and metformin-treated samples. Protein ratios of total 177 proteins were log2-transformed and compared to the cutoff value to identify differentially expressed proteins. There were five downregulated proteins and four upregulated proteins in metformin treatment group compared to control. (E) Protein expression level of cancer-associated proteins identified by iTRAQ. Cells were treated with 10 mM metformin for 48 h, then harvested, and proteins extracted. The protein levels of PSMD2, STIP1, and CAP1 identified by iTRAQ were verified by western blotting. (F) Overall survival of the low expression (blue) and high expression (res) of (A) PSMD2, (B) STIP1, and (C) CAP1. Patients (n = 192) were classified according to low expression and high expression of specific genes. Data were obtained from Gene Expression Omnibus accession number GSE15459, and the P-values were obtained from the log-rank (Mantel–Haenszel) test.
A total of 177 proteins were identified. Nine differential proteins included five downregulated proteins and four upregulated proteins between the control and treatment (Figure 3D and Table 1). Five downregulated proteins include t-complex protein 1 subunit epsilon (CCT5), 26S proteasome non-ATPase regulatory subunit 2 (PSMD2), stress-induced phosphoprotein 1 (STIP1), polypyrimidine tract-binding protein 1 (PTBP1), and adenylyl cyclase-associated protein 1 (CAP1). The western blotting showed that PSMD2 (0.76-fold), STIP1 (0.22-fold) and CAP1 (0.61-fold) were downregulated in the protein sample of metformin treatment cells, which confirmed the results in iTRAQ analysis (Figure 3E).
Table 1. Total Protein Identified in Control and Metformin-Treated AGS Cells.
| accession no | gene symbol | protein name | mass (Da) | protein score | no. of peptidesa | Log2 (protein ratio) ± S.D. (treatment/control) |
|---|---|---|---|---|---|---|
| H15_HUMAN | HIST1H1B | histone H1.5 | 22566 | 77 | 2 | –1.05 ± 0.35 |
| CNTRL_HUMAN | CNTRL | centriolin | 268720 | 42 | 1 | –1.03 ± 0.28 |
| TCPH_HUMAN | CCT7 | T-complex protein 1 subunit eta | 59329 | 81 | 1 | –0.98 ± 0.29 |
| TIF1B_HUMAN | TRIM28 | transcription intermediary factor 1-beta | 88493 | 73 | 3 | –0.96 ± 0.36 |
| LA_HUMAN | SSB | Lupus La protein | 46808 | 125 | 4 | –0.95 ± 0.89 |
| LMNB1_HUMAN | LMNB1 | Lamin-B1 | 66368 | 43 | 1 | –0.89 ± 0.36 |
| U2AF2_HUMAN | U2AF2 | splicing factor U2AF 65 kDa subunit | 53467 | 52 | 2 | –0.88 ± 0.61 |
| MDHM_HUMAN | MDH2 | malate dehydrogenase, mitochondrial | 35481 | 162 | 3 | –0.86 ± 0.48 |
| 2AAA_HUMAN | PPP2R1A | serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform | 65267 | 156 | 4 | –0.83 ± 1.27 |
| SERPH_HUMAN | SERPINH1 | serpin H1 | 46411 | 153 | 3 | –0.71 ± 0.22 |
| G6PI_HUMAN | GPI | glucose-6-phosphate isomerase | 63107 | 310 | 7 | –0.68 ± 0.7 |
| MBB1A_HUMAN | MYBBP1A | Myb-binding protein 1A | 148762 | 70 | 2 | –0.64 ± 0.43 |
| RPN2_HUMAN | RPN2 | dolichyl-diphosphooligosaccharide--protein glycosyltransferase 2 | 69241 | 81 | 2 | –0.63 ± 1.03 |
| 1433E_HUMAN | YWHAE | 14-3-3 protein epsilon | 29155 | 327 | 4 | 0.36 ± 0.21 |
| RL27A_HUMAN | RPL27A | 60S ribosomal protein L27a | 16551 | 39 | 1 | 0.37 ± 0.42 |
| IPYR_HUMAN | PPA1 | inorganic pyrophosphatase | 32639 | 47 | 1 | –0.57 ± 0.07 |
| RL9_HUMAN | RPL9 | 60S ribosomal protein L9 | 21850 | 39 | 1 | –0.57 ± 1.17 |
| DX39A_HUMAN | DDX39A | ATP-dependent RNA helicase DDX39A | 49098 | 42 | 1 | –0.52 ± 0.4 |
| HNRPF_HUMAN | HNRNPF | heterogeneous nuclear ribonucleoprotein F | 45643 | 144 | 4 | –0.51 ± 0.11 |
| H2A1B_HUMAN | HIST1H2AB | histone H2A type-1-B/E | 14127 | 185 | 2 | –0.5 ± 0.22 |
| RS16_HUMAN | RPS16 | 40S ribosomal protein S16 | 16435 | 119 | 2 | –0.49 ± 0.19 |
| KBTB3_HUMAN | KBTBD3 | Kelch repeat and BTB domain-containing protein 3 | 69350 | 42 | 1 | –0.48 ± 0.14 |
| PGK1_HUMAN | PGK1 | phosphoglycerate kinase 1 | 44586 | 106 | 3 | –0.46 ± 0.31 |
| RS28_HUMAN | RPS28 | 40S ribosomal protein S28 | 7836 | 42 | 1 | –0.45 ± 0.25 |
| 6PGD_HUMAN | PGD | 6-phosphogluconate dehydrogenase, decarboxylating | 53106 | 158 | 5 | –0.44 ± 0.31 |
| K2C1_HUMAN | KRT1 | keratin, type-II cytoskeletal 1 | 65999 | 602 | 16 | –0.42 ± 0.17 |
| IMB1_HUMAN | KPNB1 | importin subunit beta-1 | 97108 | 237 | 5 | –0.41 ± 0.65 |
| FUS_HUMAN | FUS | RNA-binding protein FUS | 53394 | 53 | 1 | –0.39 ± 0.45 |
| IF4A1_HUMAN | EIF4A1 | eukaryotic initiation factor 4A-I | 46125 | 204 | 9 | –0.39 ± 0.17 |
| PCBP2_HUMAN | PCBP2 | poly(rC)-binding protein 2 | 38556 | 99 | 3 | –0.38 ± 0.77 |
| S10AB_HUMAN | S100A11 | protein S100-A11 | 11733 | 136 | 2 | –0.36 ± 0.54 |
| 1433B_HUMAN | YWHAB | 14-3-3 protein beta/alpha | 28065 | 377 | 3 | –0.36 ± 0.15 |
| ROA2_HUMAN | HNRNPA2B1 | heterogeneous nuclear ribonucleoproteins A2/B1 | 37407 | 431 | 8 | –0.22 ± 0.09 |
| H12_HUMAN | HIST1H1C | histone H1.2 | 21352 | 125 | 3 | –0.21 ± 0.36 |
| PRDX6_HUMAN | PRDX6 | peroxiredoxin-6 | 25019 | 143 | 4 | –0.21 ± 0.11 |
| GRP75_HUMAN | HSPA9 | stress-70 protein, mitochondrial | 73635 | 209 | 4 | –0.21 ± 0.24 |
| TBB4B_HUMAN | TUBB4B | tubulin beta-4B chain | 49799 | 396 | 2 | 0.44 ± 0.21 |
| CLIC1_HUMAN | CLIC1 | chloride intracellular channel protein 1 | 26906 | 167 | 3 | 0.44 ± 0.12 |
| RS25_HUMAN | RPS25 | 40S ribosomal protein S25 | 13734 | 68 | 3 | 0.44 ± 0.25 |
| RL4_HUMAN | RPL4 | 60S ribosomal protein L4 | 47667 | 102 | 3 | 0.46 ± 0.16 |
| TBB5_HUMAN | TUBB | tubulin beta chain | 49639 | 688 | 7 | –0.34 ± 0.3 |
| RS9_HUMAN | RPS9 | 40S ribosomal protein S9 | 22578 | 77 | 2 | 0.54 ± 0.29 |
| IF5A1_HUMAN | EIF5A | eukaryotic translation initiation factor 5A-1 | 16821 | 49 | 1 | 0.54 ± 0.55 |
| DDX3X_HUMAN | DDX3X | ATP-dependent RNA helicase DDX3X | 73198 | 209 | 5 | 0.48 ± 0.41 |
| RS10_HUMAN | RPS10 | 40S ribosomal protein S10 | 18886 | 102 | 2 | –0.1 ± 0.11 |
| HNRPU_HUMAN | HNRNPU | heterogeneous nuclear ribonucleoprotein U | 90528 | 54 | 2 | –0.09 ± 0.23 |
| TCPQ_HUMAN | CCT8 | T-complex protein 1 subunit theta | 59583 | 109 | 3 | 0.55 ± 0.35 |
| XRCC5_HUMAN | XRCC5 | X-ray repair cross-complementing protein 5 | 82652 | 80 | 1 | 0.62 ± 0.65 |
| SMD3_HUMAN | SNRPD3 | small nuclear ribonucleoprotein Sm D3 | 13907 | 68 | 2 | 0.9 ± 1.06 |
| GRP78_HUMAN | HSPA5 | 78 kDa glucose-regulated protein | 72288 | 392 | 8 | –0.21 ± 0.11 |
| 4F2_HUMAN | SLC3A2 | 4F2 cell–surface antigen heavy chain | 67952 | 88 | 2 | –0.21 ± 0.8 |
| TALDO_HUMAN | TALDO1 | transaldolase | 37516 | 65 | 1 | –0.2 ± 1.13 |
| EF2_HUMAN | EEF2 | elongation factor 2 | 95277 | 555 | 16 | –0.19 ± 0.16 |
| HMGB1_HUMAN | HMGB1 | high mobility group protein B1 | 24878 | 40 | 1 | –0.19 ± 0.04 |
| RLA0L_HUMAN | RPLP0P6 | 60S acidic ribosomal protein P0-like | 34343 | 53 | 1 | –0.16 ± 0.51 |
| HSPB1_HUMAN | HSPB1 | heat shock protein beta-1 | 22768 | 90 | 3 | –0.13 ± 0.11 |
| TCPZ_HUMAN | CCT6A | T-complex protein 1 subunit zeta | 57988 | 221 | 6 | –0.13 ± 0.26 |
| HSP7C_HUMAN | HSPA8 | heat shock cognate 71 kDa protein | 70854 | 748 | 14 | –0.13 ± 0.05 |
| SET_HUMAN | SET | protein SET | 33469 | 72 | 1 | –0.12 ± 0.42 |
| KPYM_HUMAN | PKM | pyruvate kinase isozymes M1/M2 | 57900 | 1101 | 31 | –0.12 ± 0.11 |
| RSSA_HUMAN | RPSA | 40S ribosomal protein SA | 32833 | 57 | 1 | –0.12 ± 0.27 |
| VDAC1_HUMAN | VDAC1 | voltage-dependent anion-selective channel protein 1 | 30754 | 142 | 2 | –0.11 ± 0.61 |
| RS27A_HUMAN | RPS27A | Ubiquitin-40S ribosomal protein S27a | 17953 | 160 | 6 | –0.11 ± 0.08 |
| CALR_HUMAN | CALR | calreticulin | 48112 | 52 | 1 | –0.1 ± 0.49 |
| H31T_HUMAN | HIST3H3 | histone H3.1t | 15499 | 80 | 6 | –0.1 ± 0.07 |
| TKT_HUMAN | TKT | transketolase | 67835 | 239 | 4 | –0.1 ± 0.46 |
| RS3_HUMAN | RPS3 | 40S ribosomal protein S3 | 26671 | 60 | 3 | –0.09 ± 0.08 |
| PAIRB_HUMAN | SERBP1 | plasminogen activator inhibitor 1 RNA-binding protein | 44938 | 74 | 2 | –0.08 ± 0.96 |
| HNRDL_HUMAN | HNRPDL | heterogeneous nuclear ribonucleoprotein D-like | 46409 | 76 | 2 | –0.08 ± 0.18 |
| GBLP_HUMAN | GNB2L1 | guanine nucleotide-binding protein subunit beta-2-like 1 | 35055 | 66 | 2 | –0.08 ± 0.26 |
| H4_HUMAN | HIST1H4A | histone H4 | 11360 | 367 | 12 | –0.08 ± 0.03 |
| RS26_HUMAN | RPS26 | 40S ribosomal protein S26 | 13007 | 55 | 1 | –0.08 ± 0.36 |
| RS13_HUMAN | RPS13 | 40S ribosomal protein S13 | 17212 | 127 | 3 | –0.07 ± 0.13 |
| K1C19_HUMAN | KRT19 | keratin,type-I cytoskeletal 19 | 44079 | 264 | 6 | –0.07 ± 0.08 |
| TBA1B_HUMAN | TUBA1B | tubulin alpha-1B chain | 50120 | 1224 | 26 | –0.07 ± 0.05 |
| HNRPK_HUMAN | HNRNPK | heterogeneous nuclear ribonucleoprotein K | 50944 | 219 | 7 | –0.06 ± 0.11 |
| RS3A_HUMAN | RPS3A | 40S ribosomal protein S3a | 29926 | 76 | 2 | –0.05 ± 0.26 |
| DDX5_HUMAN | DDX5 | probable ATP-dependent RNA helicase DDX5 | 69105 | 85 | 3 | –0.04 ± 0.16 |
| H2B1D_HUMAN | HIST1H2BD | histone H2B type-1-D | 13928 | 617 | 15 | –0.04 ± 0.04 |
| SAHH_HUMAN | AHCY | adenosylhomocysteinase | 47685 | 101 | 4 | –0.03 ± 0.19 |
| LDHA_HUMAN | LDHA | l-lactate dehydrogenase A chain | 36665 | 532 | 14 | –0.02 ± 0.21 |
| PDIA3_HUMAN | PDIA3 | protein disulfide-isomerase A3 | 56747 | 247 | 4 | –0.02 ± 0.23 |
| HS90B_HUMAN | HSP90AB1 | heat shock protein HSP 90-beta | 83212 | 957 | 10 | –0.01 ± 0.18 |
| G3P_HUMAN | GAPDH | glyceraldehyde-3-phosphate dehydrogenase | 36030 | 1036 | 24 | 0 ± 0.02 |
| ENOA_HUMAN | ENO1 | alpha-enolase | 47139 | 711 | 20 | 0.01 ± 0.04 |
| EF1G_HUMAN | EEF1G | elongation factor 1-gamma | 50087 | 280 | 10 | 0.02 ± 0.14 |
| CALX_HUMAN | CANX | calnexin | 67526 | 70 | 2 | 0.02 ± 0.23 |
| LDHB_HUMAN | LDHB | l-lactate dehydrogenase B chain | 36615 | 293 | 10 | 0.02 ± 0.05 |
| PPIA_HUMAN | PPIA | peptidyl-prolyl cis–trans isomerase A | 18001 | 269 | 5 | 0.03 ± 0.1 |
| EF1A1_HUMAN | EEF1A1 | elongation factor 1-alpha 1 | 50109 | 328 | 14 | 0.05 ± 0.09 |
| CH60_HUMAN | HSPD1 | 60 kDa heat shock protein, mitochondrial | 61016 | 1137 | 26 | 0.05 ± 0.09 |
| ROA3_HUMAN | HNRNPA3 | heterogeneous nuclear ribonucleoprotein A3 | 39571 | 105 | 2 | 0.06 ± 0.13 |
| LMNA_HUMAN | LMNA | prelamin-A/C | 74095 | 147 | 8 | 0.06 ± 0.05 |
| ACTG_HUMAN | ACTG1 | actin, cytoplasmic 2 | 41766 | 1188 | 31 | 0.07 ± 0.02 |
| TERA_HUMAN | VCP | transitional endoplasmic reticulum ATPase | 89266 | 220 | 4 | 0.08 ± 0.52 |
| HNRPM_HUMAN | HNRNPM | heterogeneous nuclear ribonucleoprotein M | 77464 | 66 | 2 | 0.08 ± 1.67 |
| ACTN4_HUMAN | ACTN4 | alpha-actinin-4 | 104788 | 397 | 2 | 0.08 ± 0.28 |
| GDIB_HUMAN | GDI2 | Rab GDP dissociation inhibitor beta | 50631 | 263 | 4 | 0.08 ± 0.68 |
| K1C10_HUMAN | KRT10 | keratin, type-I cytoskeletal 10 | 58792 | 741 | 13 | 0.1 ± 0.1 |
| RAB10_HUMAN | RAB10 | Ras-related protein Rab-10 | 22527 | 108 | 2 | 0.12 ± 0.85 |
| NPM_HUMAN | NPM1 | nucleophosmin | 32555 | 427 | 16 | 0.13 ± 0.07 |
| EZRI_HUMAN | EZR | ezrin | 69370 | 184 | 9 | 0.13 ± 0.16 |
| PSB3_HUMAN | PSMB3 | proteasome subunit beta type-3 | 22933 | 198 | 4 | 0.13 ± 0.08 |
| XPO2_HUMAN | CSE1L | exportin-2 | 110346 | 169 | 5 | 0.13 ± 0.1 |
| ANXA1_HUMAN | ANXA1 | annexin A1 | 38690 | 44 | 1 | 0.13 ± 0.37 |
| PUR6_HUMAN | PAICS | multifunctional protein ADE2 | 47049 | 45 | 2 | 0.14 ± 0.18 |
| RAB1A_HUMAN | RAB1A | ras-related protein Rab-1A | 22663 | 72 | 1 | 0.14 ± 1.3 |
| RSMB_HUMAN | SNRPB | small nuclear ribonucleoprotein-associated proteins B and B′ | 24594 | 39 | 1 | 0.15 ± 0.13 |
| RS5_HUMAN | RPS5 | 40S ribosomal protein S5 | 22862 | 134 | 2 | 0.16 ± 0.7 |
| RS23_HUMAN | RPS23 | 40S ribosomal protein S23 | 15798 | 75 | 2 | 0.16 ± 0.18 |
| TAGL2_HUMAN | TAGLN2 | transgelin-2 | 22377 | 202 | 5 | 0.17 ± 0.15 |
| NP1L1_HUMAN | NAP1L1 | nucleosome assembly protein 1-like 1 | 45346 | 262 | 5 | 0.17 ± 0.49 |
| NUCL_HUMAN | NCL | nucleolin | 76568 | 428 | 14 | 0.18 ± 0.18 |
| K1C18_HUMAN | KRT18 | keratin, type-I cytoskeletal 18 | 48029 | 1018 | 22 | 0.19 ± 0.01 |
| DYHC1_HUMAN | DYNC1H1 | cytoplasmic dynein 1 heavy chain 1 | 532072 | 48 | 1 | 0.21 ± 0.94 |
| PROF1_HUMAN | PFN1 | profilin-1 | 15045 | 164 | 4 | 0.22 ± 0.27 |
| COF1_HUMAN | CFL1 | cofilin-1 | 18491 | 269 | 5 | 0.22 ± 0.18 |
| FLNA_HUMAN | FLNA | filamin-A | 280564 | 197 | 5 | 0.22 ± 0.21 |
| RS11_HUMAN | RPS11 | 40S ribosomal protein S11 | 18419 | 88 | 2 | 0.22 ± 0.12 |
| ANXA2_HUMAN | ANXA2 | annexin A2 | 38580 | 820 | 24 | 0.23 ± 0.15 |
| 1433Z_HUMAN | YWHAZ | 14-3-3 protein zeta/delta | 27728 | 473 | 6 | 0.24 ± 0.38 |
| TCPG_HUMAN | CCT3 | T-complex protein 1 subunit gamma | 60495 | 95 | 3 | 0.26 ± 0.02 |
| FAS_HUMAN | FASN | fatty acid synthase | 273254 | 369 | 9 | 0.26 ± 0.6 |
| K1C9_HUMAN | KRT9 | keratin, type-I cytoskeletal 9 | 62027 | 86 | 1 | 0.28 ± 0.27 |
| RL10A_HUMAN | RPL10A | 60S ribosomal protein L10a | 24816 | 71 | 2 | 0.29 ± 0.04 |
| TPIS_HUMAN | TPI1 | triosephosphate isomerase | 30772 | 80 | 3 | 0.31 ± 0.25 |
| PHB_HUMAN | PHB | prohibitin | 29786 | 46 | 1 | 0.32 ± 1.46 |
| PDIA1_HUMAN | P4HB | protein disulfide-isomerase | 57081 | 195 | 7 | 0.34 ± 0.21 |
| K2C8_HUMAN | KRT8 | keratin, type-II cytoskeletal 8 | 53671 | 1343 | 33 | 0.34 ± 0.01 |
| IQGA1_HUMAN | IQGAP1 | ras GTPase-activating-like protein IQGAP1 | 189134 | 97 | 2 | 0.35 ± 0.65 |
| RS8_HUMAN | RPS8 | 40S ribosomal protein S8 | 24190 | 66 | 2 | 0.35 ± 0.46 |
| GTR1_HUMAN | SLC2A1 | solute carrier family 2, facilitated glucose transporter member 1 | 54049 | 59 | 2 | 0.38 ± 0.16 |
| RL6_HUMAN | RPL6 | 60S ribosomal protein L6 | 32708 | 142 | 3 | 0.38 ± 0.72 |
| RS4X_HUMAN | RPS4X | 40S ribosomal protein S4, X isoform | 29579 | 134 | 4 | 0.38 ± 0.22 |
| RL13A_HUMAN | RPL13A | 60S ribosomal protein L13a | 23562 | 54 | 1 | 0.43 ± 0.2 |
| PGAM1_HUMAN | PGAM1 | phosphoglycerate mutase 1 | 28786 | 133 | 4 | –0.6 ± 0.5 |
| MYH9_HUMAN | MYH9 | myosin-9 | 226392 | 176 | 4 | –0.59 ± 0.53 |
| RL15_HUMAN | RPL15 | 60S ribosomal protein L15 | 24131 | 51 | 1 | –0.59 ± 0.85 |
| TRY1_HUMAN | PRSS1 | trypsin-1 | 26541 | 54 | 1 | –0.58 ± 1.18 |
| DHX9_HUMAN | DHX9 | ATP-dependent RNA helicase A | 140869 | 447 | 12 | –0.58 ± 1.34 |
| NDKA_HUMAN | NME1 | nucleoside diphosphate kinase A | 17138 | 117 | 4 | –0.02 ± 0.29 |
| HS90A_HUMAN | HSP90AA1 | heat shock protein HSP 90-alpha | 84607 | 973 | 16 | –0.02 ± 0.07 |
| RL17_HUMAN | RPL17 | 60S ribosomal protein L17 | 21383 | 60 | 1 | 0.79 ± 0.21 |
| LPPRC_HUMAN | LRPPRC | leucine-rich PPR motif-containing protein, mitochondrial | 157805 | 141 | 4 | 0.82 ± 0.52 |
| RL1D1_HUMAN | RSL1D1 | ribosomal L1 domain-containing protein 1 | 54939 | 59 | 1 | 0.82 ± 0.79 |
| ENPL_HUMAN | HSP90B1 | endoplasmin | 92411 | 146 | 3 | 0.08 ± 0.1 |
| PRDX1_HUMAN | PRDX1 | peroxiredoxin-1 | 22096 | 188 | 8 | 0.09 ± 0.21 |
| ADT2_HUMAN | SLC25A5 | ADP/ATP translocase 2 | 32831 | 79 | 4 | 0.09 ± 0.12 |
| PP1A_HUMAN | PPP1CA | serine/threonine-protein phosphatase PP1-alpha catalytic subunit | 37488 | 148 | 2 | 0.19 ± 1.04 |
| MOT1_HUMAN | SLC16A1 | monocarboxylate transporter 1 | 53909 | 42 | 1 | 0.2 ± 0.32 |
| IPO7_HUMAN | IPO7 | importin-7 | 119440 | 80 | 3 | 0.62 ± 0.43 |
| HSP71_HUMAN | HSPA1A | heat shock 70 kDa protein 1A/1B | 70009 | 207 | 1 | 0.64 ± 0.75 |
| K22E_HUMAN | KRT2 | keratin, type-II cytoskeletal 2 epidermal | 65393 | 103 | 1 | 0.65 ± 0.35 |
| RS20_HUMAN | RPS20 | 40S ribosomal protein S20 | 13364 | 52 | 1 | 0.74 ± 0.79 |
| PSMD6_HUMAN | PSMD6 | 26S proteasome non-ATPase regulatory subunit 6 | 45502 | 65 | 2 | 0.79 ± 0.95 |
The number of nondegenerate (unique) peptides used for quantitation in two technical replicates.
To understand whether the expressions of PSMD2, STIP1, and CAP1 are associated with patient survival, the Kaplan–Meier survival analysis was performed. We classified the patients into low expression and high expression. We observed that patients with highly expressed PSMD2, STIP1, and CAP1 had a poorer survival rate with statistical significance (Figure 3F).
Human CAP1 plays a crucial role in the actin turnover.33 CAP1 knockdown shows an abnormal cell morphology and reduces cell migration in mammalian cells.34 These studies and our results suggest that metformin induces inhibition of cell migration and abnormal cell morphology by reducing the CAP1 level. PSMD2 is a subunit of the 19S regulator of 26S proteasome, which regulates the proteasomal activity and cell growth by arresting the G1 phase of cell cycle in the lung cancer cell line. It is a signature of poor prognosis and metastasis phenotype in lung cancer patients.35,36 siPSMD2 results in cell cycle arrest in breast and liver cancers and the inhibition of tumorigenesis.37,38 STIP1 is an oncoprotein that stimulates ovarian cancer cell proliferation by activating the SMAD-ID3 signaling pathways.39 A recent study suggests that siSTIP1 disrupts cellular migration and cell growth in lung cancer cells.40 These results suggest that metformin might reduce protein levels of STIP1 and PSMD2 to inhibit AGS cell proliferation.
Metformin Blocks the Cell Cycle in the G0/G1 Phase and Inhibits Migration
According to our iTRAQ-based proteome, several differential proteins, including PSMD2, CAP1, and STIP1, are responsible for cell proliferation and cell cycle. We have found that metformin decreased cell viability previously (Figure 2A). To determine the underlying mechanism of cell growth inhibition, metformin-treated AGS cells were analyzed with flow cytometry. Metformin-treated cells showed an increasing G0/G1 phase and failed to enter the S phase (Figure 4A). To determine whether metformin affects cell cycle regulatory proteins, we examined the expression level of the cell cycle regulatory proteins of AGS cells after the treatment of metformin. CDK4 and CDK6 are the members of the cyclin-dependent protein kinase (CDK) family, which are essential for cell cycle G1 phase progression and G1–S phase transition.41,42 CDK4 and CDK6 proteins’ expression level was significantly reduced in metformin-treated AGS cells (Figure 4B). These data suggested that metformin arrested the cell cycle in the G0/G1 phase by reducing CDK4 and CDK6. Many studies had been reported that the inhibition of cell cycle G1 regulatory protein inhibited cell proliferation in various cancers.43,44 Therefore, these results suggest that metformin inhibits AGS cell proliferation and block cell cycle at the G1 phase through decreasing the protein levels of CDK4 and CDK6.
Figure 4.
Metformin blocks the AGS cell cycle at the G0/G1 phase and affects the expression level of cell cycle regulatory proteins. (A) Cells were starved for 24 h, then treated with 10 mM metformin for 48 h and harvested for flow cytometry analysis or protein extraction. The percentage of each cell cycle phase was analyzed by FCS Express. (B) Expression of cell cycle-related proteins CDK4 and CDK6. (C) After 48 h of metformin treatment, AGS cells were analyzed for cell migration using the transwell assay for another 9 h. (D) Quantification of the transwell assay. Data are presented as mean ± S.D. (n = 3). ** indicates that the p value <0.001. (E) After 48 h of metformin treatment, AGS cells were incubated with or without metformin treatment and analyzed for the cell migration using RTCA with a CIM plate for another 9 h. Data are presented as mean ± S.D. (n = 3) (F) Metformin induces reorganization of cytoskeletons in AGS cells. Cells were seeded on the coverslips in the six-well plate, incubated for 24 h, and then treated with 10 mM metformin for 48 h. After treatment, cells were fixed, stained, and visualized with a fluorescence microscope.
To compare our results with previous studies, we have surveyed whether the proteomic results of metformin-treated gastric cancer have been published. We only found the secretome research of metformin-treated gastric tumor-associated fibroblasts (TAFs). Chen et al. suggested that TAFs are essential for the gastric cancer microenvironment.45 Factors involved in the functional enrichment of differential proteins of the secretome were cytoskeleton organization and cell cycle arrest. A previous study showed the enrichment of metformin-regulated proteins in cell cycle arrest and antiproliferation of breast cancer cell lines using deep proteomic techniques.17 The publications mentioned above found a common biological function that metformin induced cell cycle arrest and antiproliferation, resulting in cell death.
To evaluate whether metformin induces human gastric cancer cell apoptosis, we applied flow cytometry analysis. AGS cells were incubated with or without metformin for 72 h. We found that the percentages of the late apoptotic cells showed no significant difference between the control (8.84%) and metformin (9.16%)-treated cells (Figure S1A). Similarly, cells stained with 4′,6-diamidino-2-phenylindole (DAPI) did not show the difference in DNA condensation, one of the most important criteria to identify apoptotic cells,46 between the control and metformin-treated cells (Figure S1B). These results showed that metformin did not induce apoptosis in AGS cells.
The migration and invasion of cancer cells is an initial step in tumor metastasis which is the most frequent cause of death for patients with cancer,47 Therefore, we would like to determine if metformin affects cell migration ability in human gastric cancer cells. In the transwell assay, the cells that migrated across to the transwell filter were stained and counted. There was a remarkably reduced number of migratory cells in metformin treatment group compared with control (Figure 4C,D). Similarly, metformin inhibited the migration of AGS cells as assessed by real-time cell analysis (RTCA) (Figure 4E). These results indicate that metformin inhibits the migration ability of AGS cells.
Metformin Affects the Reorganization of the Actin Filament and the Distribution of Vinculin
Cell migration requires various cell-shape changes, which implicate the reorganization of the actin filament and distribution of focal adhesion proteins, such as vinculin.48 The interaction between vinculin and membrane-bound β-catenin directly affects colorectal cancer metastasis.49 Here, we examined the formation and distribution of the actin filament and vinculin in AGS cells. In control cells, the actin filament formed stress fiber formation, and the vinculin co-localized with F-actin at a distal end of the stress fiber. In contrast, metformin abolished the stress fiber formation of actin, and most of the vinculins were distributed around the cells (Figure 4F). Besides, the cell shape became more spindle-like and bigger in metformin treatment than in control. These data suggested that metformin affects the reorganization of the actin filament and distribution of vinculin, resulting in cell migration inhibition. Similar effects were found in hepatocellular carcinoma and cervical cancer cells through regulating the AMPK signaling pathway.50,51 AMPK is known for the upstream regulation of vinculin and other cytoskeletons.52 Previous research has shown that metformin-induced differential proteins were enriched in the mTOR signaling pathway, which regulated migration.53 We have found that metformin abolished actin stress fiber formation and changed the distribution of vinculin in AGS cells. These results suggest that metformin can inhibit cancer cell migration and enlarge the cell shape by affecting the distribution of focal adhesion and arrangement of actin.
Conclusions
In this study, we proposed the biological effects that are induced by metformin in Figure S2. Metformin-treated AGS cells had a slower growth rate and were arrested at the G0 phase by the suppression of PSMD2. On the other hand, metformin decreased the expression of CAP1 and STIP1, which regulated the cell motility, maintained the cell shape, and reduced the migration of AGS cells. Therefore, we consider that metformin has high potential in new anticancer therapy.
Experimental Section
Chemicals
Metformin (1,1-dimethylbiguanide hydrochloride), triethylammonium bicarbonate buffer (TEABC), thiazolyl blue tetrazolium bromide (MTT), tris(2-carboxyethyl) phosphine hydrochloride (TCEP), S-methylmethanethiosulfonate, Tween 20, and Triton X-100 were purchased from Sigma-Aldrich (St. Louis, MO, USA). Acetonitrile (ACN) was purchased from Lab-Scan. Sequencing grade modified trypsin was purchased from Promega (Madison, WI, USA). Dimethyl sulfoxide was purchased from Scharlau Chemie (Barcelona, Spain).
Cell Culture
AGS cell line was purchased from ATCC (CRL-1739). Cells were cultured in a complete medium (RPMI-1640 medium supplemented with 10% (v/v) FBS) at 37 °C with 5% CO2. The growing cells were sub-cultured every few days when it reached 80–90% confluency.
MTT Assay
MTT solution was prepared in phosphate-buffered saline (PBS) at a concentration of 5 mg/mL and sterilized by a 0.22 μm filter. 3,000 cells were seeded into a 96-well plate and incubated for 24 h before treated with 1, 5, 10, 20, and 50 mM metformin for 24, 48, and 72 h. Steps were described in our previous paper.54
Colony Formation
250 cells were seeded into each well of six-well plate and incubated for 24 h. After 24 h, the medium was changed to a complete medium or a complete medium with 10 mM metformin for treatment and incubated for 7 days at 37 °C. Steps were described in our previous paper.54
Cell Cycle and Apoptosis Analysis by Flow Cytometry
1 × 105 cells were seeded into a 10 cm dish and incubated for 24 h, followed by 24 h of starvation in serum-free medium (RPMI-1640 medium without FBS). 10 mM metformin was added for 24, 48, or 72 h. Steps were described in our previous paper.54 For apoptosis analysis, 1 × 105 cells were seeded for 24 h and then treated with 10 mM metformin. After being treated with 10 mM metformin for 72 h, the cells were trypsinized and stained with 100 μL binding buffer with annexin V-FITC and PI at room temperature for 15 min. The samples were analyzed by BD FACSCanto II.
Transwell Migration Assay
1 × 105 cells were seeded onto a 10 cm dish and incubated for 24 h and then treated with 10 mM metformin. Cells were trypsinized after 48 h of metformin treatment and seeded 5 × 104 of cells into 8 μm Hanging cell culture inserts (Millipore, Billerica, USA) of the transwell. 10 mM metformin in serum-free medium and a complete medium with 10 mM metformin were added to the bottom of the transwell. The migratory cells were fixed in methanol and stained by crystal violet after 8 h.
Immunofluorescence Staining
1 × 104 cells were seeded on the coverslips in the six-well plate and incubated for 24 h and then treated with 10 mM metformin for 48 h. The coverslip was fixed in 4% (v/v) paraformaldehyde for 15 min at room temperature and permeabilized by 0.25% (v/v) Triton X-100 for 5 min. The coverslip was blocked for 30 min in a blocking solution [1% (w/v) bovine serum albumin (Bioshop) in PBS] at room temperature. Primary mouse anti-human vinculin monoclonal antibody (1:200) (Millipore) and rabbit anti-human ki67 polyclonal antibody (1:100) (Abcam, Cambridge, MA, USA) were diluted in the blocking solution and incubated overnight at 4 °C. After washing twice with PBST (PBS with 0.05% (v/v) Tween-20), samples labeled with vinculin and ki67 were incubated in the secondary anti-mouse IgG FITC antibody (1:100 in PBS; Invitrogen, Carlsbad, CA) and anti-rabbit IgG FITC antibody (1:100 in PBS; Invitrogen, Carlsbad, CA), respectively, with TRITC–phalloidin (1:1250 in PBS; Millipore) for 30 min at room temperature, followed by washing three times with PBST. Subsequently, coverslips were mounted with a ProLong Gold antifade reagent with DAPI (Invitrogen) on the slide. Steps were described in our previous paper.54
xCELLigence RTCA Dual-Plate System
To determine whether the inhibition of cell proliferation by metformin is irreversible or reversible, 3,000 cells were seeded into each well of E-plate after 48 h of metformin treatment. Cells were incubated with or without 10 mM metformin and the cell growth was recorded once an hour for 72 h. A detailed protocol was published previously.54 To evaluate the cell migration ability after metformin treatment, we use the transwell migration assay and the RTCA DP system with cell invasion/migration (CIM)-plate.
iTRAQ Technique
10 mM metformin-treated cells or control were trypsinized after 72 h and lysis buffer containing 1% (v/v) SDS, 50 mM Tris-HCl, 10% (v/v) glycerol, and protease inhibitor (Bioman, Taipei, Taiwan) were added. The samples were homogenized by a LABSONIC M ultrasonic homogenizer (Sartorius AG, Goettingen, Germany) for 1.5 min on ice and then centrifuged at 17,000×g for 30 min at 4 °C. The proteins in the supernatant were collected with new tubes and quantified with a BCA Protein Assay Reagent kit (Pierce, Rockford, IL, USA). Samples were reduced by TCEP (Sigma-Aldrich) at 37 °C for 30 min and alkylated by iodoacetamide at room temperature in the dark for 30 min.
60 μg of proteins of each sample were adjusted to an equal volume. Proteins were dissolved in 50 mM TEABC to adjust the pH to about 8.5. Subsequently, to the protein solutions were added 5 mM TECP for 30 min at 37 °C and then added 2 mM MTTS at room temperature in the dark for 30 min to reduce and alkylate the cysteines, which might interfere with the iTRAQ signal. For gel-assisted digestion, the protein solution was mixed with acrylamide/bisacrylamide (40%, 37.5:1; Bioshop), APS, and TEMED in the ratio 14:5:0.3:0.3 (v/v) at room temperature for about 10 min of solidifying. The gels were cut into small pieces and washed with 25 mM TEABC and 25 mM TEABC containing 50% (v/v) ACN until no bubble was observed. The gels were dehydrated with 100% ACN and dried completely with a Centrifugal Evaporator CVE-2000 with Uni Trap UT-1000 (Eyela, Japan) for 20 min. The gels were digested with trypsin in 25 mM TEABC (protein amount/trypsin amount = 10:1) in a 37°C water bath overnight. After trypsin digestion, peptides were extracted with 0.1% (v/v) trifluoroacetyl (TFA), 50% (v/v) ACN containing 0.1% (v/v) TFA, and 100% ACN, and peptide solutions were combined and dried with the Centrifugal Evaporator CVE-2000 with Uni Trap UT-1000.
The iTRAQ Reagent kit was purchased from Applied Biosystems (Forster City, CA, USA). The dried peptides were dissolved in iTRAQ dissolution buffer and quantified with a BCA Protein Assay Reagent kit. 30 μg of peptides of each sample were labeled with the isobaric chemical tag. For duplication, the control samples were labeled with iTRAQ114 and iTRAQ115, and the metformin-treated samples were labeled with iTRAQ116 and iTRAQ117 for 1 h at room temperature. All labeled peptides were pooled and dried with the Centrifugal Evaporator CVE-2000 with Uni Trap UT-1000.
Peptides were desalted using ZipTip Pipette Tips (Millipore) before LC–MS/MS spectrometry. The peptides were dissolved in 0.1% (v/v) TFA and adjusted pH to 2–3 with 10% (v/v) TFA. The ZipTip was rinsed with 50% (v/v) ACN containing 0.1% (v/v) TFA by aspirating and dispensing the solution several times. The ZipTip was equilibrated in 0.1% TFA and bound the peptides by aspirating and dispensing the peptide solution 20 times. The peptides in ZipTip were washed with 0.1% TFA and ultimately eluted with 50% ACN containing 0.1% TFA at least 10 times. The desalted peptide solution was dried with the Centrifugal Evaporator CVE-2000 with Uni Trap UT-1000. After desalting, samples were analyzed by LC-ESI-Q-TOF mass spectrometry (Waters SYNAPT G2 HDMS; Waters Corp., Milford, MA, USA) equipped with a nanoACQUITY UPLC system (Waters Corp., Milford, MA, USA). Peptide samples were loaded onto a 2 cm × 180 μm capillary trap column and separated in a 75 μm × 25 cm nanoACQUITY 1.7 μm BEH C18 column. The mobile phases consisted of buffer A (0.1% formic acid) and buffer B (0.1% formic acid in acetonitrile). Peptides were eluted with a linear gradient of 6–50% buffer B. The flow rate was 300 nL/min for 100 min. A NanoLockSpray source was used for accurate mass measurement, and the lock mass channel was sampled every 30 s. A mass spectrometer was calibrated with a synthetic human [Glu1]–fibrinopeptide B solution (1 pmol/μL; Sigma-Aldrich) delivered through the NanoLockSpray source. Data acquisition was performed using the data-directed analysis (DDA). The DDA method included one full MS scan (m/z 350–1700, 1 s) and three MS/MS scans (m/z 100–1990, 1.5 s for each scan) sequentially on the three most intense ions present in the full-scan mass spectrum. Each sample was analyzed in technical replicates. Mass spectral data were deposited at 109 ProteomeXChange (http://www.proteomexchange.org/), project accession number PXD024996. Details for the protocols were published previously.32
Protein Identification
Mass spectral data were converted to mgf and mzXML file formats using Mascot Distiller (version 2.3.2; Matrix Science, London, United Kingdom) and massWolf (version 4.3.1; Institute for Systems Biology, Seattle, WA, USA), with default settings, respectively. The mgf files were submitted to Mascot Daemon (version 2.3.2; Matrix Science) for peptide identification. For peptide identification, the MS/MS spectral data were searched against the Homo sapiens of SwissProt database with the following parameters: trypsin defined as an enzyme with two missed cleavages allowed, a tolerance of 0.1 Da on the peptide ion, and an MS/MS fragment ion and variable modification settings such as iTRAQ4plex (N-term), iTRAQ4plex (K), deamidated (NQ), oxidation (M), and methylthio (C). Mascot search results were exported to the XML file format after being filtered by the significance threshold p < 0.05 and the ion score cutoff of 0.05. The XML and mzXML files were submitted to Multi-Q software (version 1.6.5.4)55 for selecting qualified peptides. The peptides that satisfied the following criteria were selected for further analysis: the peptide is labeled with iTRAQ; the peptide is nondegenerate; the ion score of the peptide is higher than the Mascot identity score (P < 0.05); and the average of all iTRAQ intensities higher or equal to 30.
Survival Analysis
The Kaplan–Meier overall survival analysis was performed for the gastric cancer patient with Gene Expression Omnibus accession number GSE15459.56
Statistical Analysis
The log-rank (Mantel–Haenszel) test was performed by R2: Genomics Analysis and Visualization Platform (http://r2.amc.nl). P-value <0.05 means two groups have statistical significance.
Acknowledgments
We thank Technology Commons in College of Life Science, National Taiwan University for technical assistance with the flow cytometer. We would like to thank Dr. Cheung, Hoi Yin Chantal for editing and proofreading this manuscript.
Supporting Information Available
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsomega.1c00894.
Metformin does not result in apoptosis but affects the cell cycle and migration in AGS cells (PDF)
This research was funded by the Ministry of Science and Technology (MOST 109-2221-E-010-012-MY3, MOST 109-2320-B-002-017-MY3, and MOST 109-2221-E-010-011-MY3), the Higher Education Sprout Project (109L8837A), and the National Health Research Institutes (NHRI-EX109-10709BI) in Taiwan. The APC was funded by MOST 109-2320-B-002-017-MY3.
The authors declare no competing financial interest.
Supplementary Material
References
- Rawla P.; Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and prevention. Przegl. Gastroenterol. 2019, 14, 26–38. 10.5114/pg.2018.80001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siegel R.; Naishadham D.; Jemal A. Cancer statistics, 2013. Ca-Cancer J. Clin. 2013, 63, 11–30. 10.3322/caac.21166. [DOI] [PubMed] [Google Scholar]
- Noh S. H.; Park S. R.; Yang H.-K.; Chung H. C.; Chung I.-J.; Kim S.-W.; Kim H.-H.; Choi J.-H.; Kim H.-K.; Yu W.; Lee J. I.; Shin D. B.; Ji J.; Chen J.-S.; Lim Y.; Ha S.; Bang Y.-J. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014, 15, 1389–1396. 10.1016/s1470-2045(14)70473-5. [DOI] [PubMed] [Google Scholar]
- Wu B.; Wu D.; Wang M.; Wang G. Recurrence in patients following curative resection of early gastric carcinoma. J. Surg. Oncol. 2008, 98, 411–414. 10.1002/jso.21133. [DOI] [PubMed] [Google Scholar]
- Cunningham D.; Allum W. H.; Stenning S. P.; Thompson J. N.; Van de Velde C. J. H.; Nicolson M.; Scarffe J. H.; Lofts F. J.; Falk S. J.; Iveson T. J.; Smith D. B.; Langley R. E.; Verma M.; Weeden S.; Chua Y. J.; Participants M. T. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N. Engl. J. Med. 2006, 355, 11–20. 10.1056/nejmoa055531. [DOI] [PubMed] [Google Scholar]
- Giugliano D.; Quatraro A.; Consoli G.; Minei A.; Ceriello A.; De Rosa N.; D’Onofrio F. Metformin for obese, insulin-treated diabetic patients: improvement in glycaemic control and reduction of metabolic risk factors. Eur. J. Clin. Pharmacol. 1993, 44, 107–112. 10.1007/bf00315466. [DOI] [PubMed] [Google Scholar]
- Cree-Green M.; Bergman B. C.; Cengiz E.; Fox L. A.; Hannon T. S.; Miller K.; Nathan B.; Pyle L.; Kahn D.; Tansey M.; Tichy E.; Tsalikian E.; Libman I.; Nadeau K. J. Metformin Improves Peripheral Insulin Sensitivity in Youth With Type 1 Diabetes. J. Clin. Endocrinol. Metab. 2019, 104, 3265–3278. 10.1210/jc.2019-00129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galuska D.; Zierath J.; Thörne A.; Sonnenfeld T.; Wallberg-Henriksson H. Metformin increases insulin-stimulated glucose transport in insulin-resistant human skeletal muscle. Diabete Metab. 1991, 17, 159–163. [PubMed] [Google Scholar]
- Winder W. W.; Hardie D. G. AMP-activated protein kinase, a metabolic master switch: possible roles in type 2 diabetes. Am. J. Physiol. 1999, 277, E1–E10. 10.1152/ajpendo.1999.277.1.e1. [DOI] [PubMed] [Google Scholar]
- Foretz M.; Guigas B.; Viollet B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2019, 15, 569–589. 10.1038/s41574-019-0242-2. [DOI] [PubMed] [Google Scholar]
- Hunter R. W.; Hughey C. C.; Lantier L.; Sundelin E. I.; Peggie M.; Zeqiraj E.; Sicheri F.; Jessen N.; Wasserman D. H.; Sakamoto K. Metformin reduces liver glucose production by inhibition of fructose-1-6-bisphosphatase. Nat. Med. 2018, 24, 1395–1406. 10.1038/s41591-018-0159-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evans J. M. M.; Donnelly L. A.; Emslie-Smith A. M.; Alessi D. R.; Morris A. D. Metformin and reduced risk of cancer in diabetic patients. BMJ 2005, 330, 1304. 10.1136/bmj.38415.708634.f7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Libby G.; Donnelly L. A.; Donnan P. T.; Alessi D. R.; Morris A. D.; Evans J. M. M. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care 2009, 32, 1620. 10.2337/dc08-2175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Menendez J. A. The anti-diabetic drug metformin suppresses the metastasis-associated protein CD24 in MDA-MB-468 triple-negative breast cancer cells. Oncol. Rep. 2011, 25, 135. 10.3892/or_00001052. [DOI] [PubMed] [Google Scholar]
- Storozhuk Y.; Hopmans S. N.; Sanli T.; Barron C.; Tsiani E.; Cutz J.-C.; Pond G.; Wright J.; Singh G.; Tsakiridis T. Metformin inhibits growth and enhances radiation response of non-small cell lung cancer (NSCLC) through ATM and AMPK. Br. J. Canc. 2013, 108, 2021–2032. 10.1038/bjc.2013.187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou X. Z.; Xue Y. M.; Zhu B.; Sha J. P. [Effects of metformin on proliferation of human colon carcinoma cell line SW-480]. Nan Fang Yi Ke Da Xue Xue Bao 2010, 30, 1935. [PubMed] [Google Scholar]
- Zakikhani M.; Dowling R.; Fantus I. G.; Sonenberg N.; Pollak M. Metformin Is an AMP Kinase-Dependent Growth Inhibitor for Breast Cancer Cells. Cancer Res. 2006, 66, 10269. 10.1158/0008-5472.can-06-1500. [DOI] [PubMed] [Google Scholar]
- Zhou X. Z.; Xue Y. M.; Zhu B.; Sha J. P. [Effects of metformin on proliferation of human colon carcinoma cell line SW-480]. Nan Fang Yi Ke Da Xue Xue Bao 2010, 30, 1935–1942. [PubMed] [Google Scholar]
- Ben Sahra I.; Regazzetti C.; Robert G.; Laurent K.; Le Marchand-Brustel Y.; Auberger P.; Tanti J.-F.; Giorgetti-Peraldi S.; Bost F. Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res. 2011, 71, 4366–4372. 10.1158/0008-5472.can-10-1769. [DOI] [PubMed] [Google Scholar]
- Pollak M. N. Investigating metformin for cancer prevention and treatment: the end of the beginning. Canc. Discov. 2012, 2, 778–790. 10.1158/2159-8290.cd-12-0263. [DOI] [PubMed] [Google Scholar]
- Cox J.; Mann M. Quantitative, high-resolution proteomics for data-driven systems biology. Annu. Rev. Biochem. 2011, 80, 273–299. 10.1146/annurev-biochem-061308-093216. [DOI] [PubMed] [Google Scholar]
- Persidis A. Proteomics. Nat. Biotechnol. 1998, 16, 393. 10.1038/nbt0498-393. [DOI] [PubMed] [Google Scholar]
- Chen X.; Wang Y.; Ma N.; Tian J.; Shao Y.; Zhu B.; Wong Y. K.; Liang Z.; Zou C.; Wang J. Target identification of natural medicine with chemical proteomics approach: probe synthesis, target fishing and protein identification. Signal Transduction Targeted Ther. 2020, 5, 72. 10.1038/s41392-020-0186-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruprecht B.; Di Bernardo J.; Wang Z.; Mo X.; Ursu O.; Christopher M.; Fernandez R. B.; Zheng L.; Dill B. D.; Wang H.; Xu Y.; Liaw A.; Mortison J. D.; Siriwardana N.; Andresen B.; Glick M.; Tata J. R.; Kutilek V.; Cornella-Taracido I.; Chi A. A mass spectrometry-based proteome map of drug action in lung cancer cell lines. Nat. Chem. Biol. 2020, 16, 1111–1119. 10.1038/s41589-020-0572-3. [DOI] [PubMed] [Google Scholar]
- Hsieh C.-H.; Cheung C. H. Y.; Liu Y.-L.; Hou C.-L.; Hsu C.-L.; Huang C.-T.; Yang T.-S.; Chen S.-F.; Chen C.-N.; Hsu W.-M.; Huang H.-C.; Juan H.-F. Quantitative Proteomics of Th-MYCN Transgenic Mice Reveals Aurora Kinase Inhibitor Altered Metabolic Pathways and Enhanced ACADM To Suppress Neuroblastoma Progression. J. Proteome Res. 2019, 18, 3850–3866. 10.1021/acs.jproteome.9b00245. [DOI] [PubMed] [Google Scholar]
- Lee J.-M.; Kohn E. C. Proteomics as a guiding tool for more effective personalized therapy. Ann. Oncol. 2010, 21, vii205–vii210. 10.1093/annonc/mdq375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu C.; Zheng L. Proteomics promises a new era of precision cancer medicine. Signal Transduction Targeted Ther. 2019, 4, 13. 10.1038/s41392-019-0046-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang Y.-W.; Hsu C.-L.; Tang C.-W.; Chen X.-J.; Huang H.-C.; Juan H.-F. Multiomics Reveals Ectopic ATP Synthase Blockade Induces Cancer Cell Death via a lncRNA-mediated Phospho-signaling Network. Mol. Cell. Proteomics 2020, 19, 1805–1825. 10.1074/mcp.ra120.002219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheung C. H. Y.; Hsu C. L.; Lin T. Y.; Chen W. T.; Wang Y. C.; Huang H. C.; Juan H. F. ZNF322A-mediated protein phosphorylation induces autophagosome formation through modulation of IRS1-AKT glucose uptake and HSP-elicited UPR in lung cancer. J. Biomed. Sci. 2020, 27, 75. 10.1186/s12929-020-00668-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang W.-H.; Hsu C.-L.; Huang H.-C.; Juan H.-F. Quantitative Phosphoproteomics Reveals Cell Alignment and Mitochondrial Length Change under Cyclic Stretching in Lung Cells. Int. J. Mol. Sci. 2020, 21, 4074. 10.3390/ijms21114074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ross P. L.; Huang Y. N.; Marchese J. N.; Williamson B.; Parker K.; Hattan S.; Khainovski N.; Pillai S.; Dey S.; Daniels S.; Purkayastha S.; Juhasz P.; Martin S.; Bartlet-Jones M.; He F.; Jacobson A.; Pappin D. J. Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol. Cell. Proteomics 2004, 3, 1154–1169. 10.1074/mcp.m400129-mcp200. [DOI] [PubMed] [Google Scholar]
- Chang H.-Y.; Li M.-H.; Huang T.-C.; Hsu C.-L.; Tsai S.-R.; Lee S.-C.; Huang H.-C.; Juan H.-F. Quantitative proteomics reveals middle infrared radiation-interfered networks in breast cancer cells. J. Proteome Res. 2015, 14, 1250–1262. 10.1021/pr5011873. [DOI] [PubMed] [Google Scholar]
- Moriyama K.; Yahara I. Human CAP1 is a key factor in the recycling of cofilin and actin for rapid actin turnover. J. Cell Sci. 2002, 115, 1591–1601. 10.1242/jcs.115.8.1591. [DOI] [PubMed] [Google Scholar]
- Bertling E.; Hotulainen P.; Mattila P. K.; Matilainen T.; Salminen M.; Lappalainen P. Cyclase-associated protein 1 (CAP1) promotes cofilin-induced actin dynamics in mammalian nonmuscle cells. Mol. Biol. Cell 2004, 15, 2324–2334. 10.1091/mbc.e04-01-0048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tomida S.; Yanagisawa K.; Koshikawa K.; Yatabe Y.; Mitsudomi T.; Osada H.; Takahashi T. Identification of a metastasis signature and the DLX4 homeobox protein as a regulator of metastasis by combined transcriptome approach. Oncogene 2007, 26, 4600–4608. 10.1038/sj.onc.1210242. [DOI] [PubMed] [Google Scholar]
- Matsuyama Y.; Suzuki M.; Arima C.; Huang Q. M.; Tomida S.; Takeuchi T.; Sugiyama R.; Itoh Y.; Yatabe Y.; Goto H.; Takahashi T. Proteasomal non-catalytic subunit PSMD2 as a potential therapeutic target in association with various clinicopathologic features in lung adenocarcinomas. Mol. Carcinog. 2011, 50, 301–309. 10.1002/mc.20632. [DOI] [PubMed] [Google Scholar]
- Li Y.; Huang J.; Zeng B.; Yang D.; Sun J.; Yin X.; Lu M.; Qiu Z.; Peng W.; Xiang T.; Li H.; Ren G. PSMD2 regulates breast cancer cell proliferation and cell cycle progression by modulating p21 and p27 proteasomal degradation. Canc. Lett. 2018, 430, 109–122. 10.1016/j.canlet.2018.05.018. [DOI] [PubMed] [Google Scholar]
- Tan Y.; Jin Y.; Wu X.; Ren Z. PSMD1 and PSMD2 regulate HepG2 cell proliferation and apoptosis via modulating cellular lipid droplet metabolism. BMC Mol. Biol. 2019, 20, 24. 10.1186/s12867-019-0141-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai C.-L.; Tsai C.-N.; Lin C.-Y.; Chen H.-W.; Lee Y.-S.; Chao A.; Wang T.-H.; Wang H.-S.; Lai C.-H. Secreted stress-induced phosphoprotein 1 activates the ALK2-SMAD signaling pathways and promotes cell proliferation of ovarian cancer cells. Cell Rep. 2012, 2, 283–293. 10.1016/j.celrep.2012.07.002. [DOI] [PubMed] [Google Scholar]
- Guo X.; Yan Z.; Zhang G.; Wang X.; Pan Y.; Huang M. STIP1 Regulates Proliferation and Migration of Lung Adenocarcinoma Through JAK2/STAT3 Signaling Pathway. Canc. Manag. Res. 2019, 11, 10061–10072. 10.2147/cmar.s233758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malumbres M.; Barbacid M. Mammalian cyclin-dependent kinases. Trends Biochem. Sci. 2005, 30, 630–641. 10.1016/j.tibs.2005.09.005. [DOI] [PubMed] [Google Scholar]
- Sherr C. J. Mammalian G1 cyclins. Cell 1993, 73, 1059–1065. 10.1016/0092-8674(93)90636-5. [DOI] [PubMed] [Google Scholar]
- Shao Y.; Qu Y.; Dang S.; Yao B.; Ji M. MiR-145 inhibits oral squamous cell carcinoma (OSCC) cell growth by targeting c-Myc and Cdk6. Canc. Cell Int. 2013, 13, 51. 10.1186/1475-2867-13-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing Z.; Zhang Y.; Zhang X.; Yang Y.; Ma Y.; Pang D. Fangchinoline induces G1 arrest in breast cancer cells through cell-cycle regulation. Phytother Res. 2013, 27, 1790–1794. 10.1002/ptr.4936. [DOI] [PubMed] [Google Scholar]
- Chen G.; Yu C.; Tang Z.; Liu S.; An F.; Zhu J.; Wu Q.; Cao J.; Zhan Q.; Zhang S. Metformin suppresses gastric cancer progression through calmodulinlike protein 3 secreted frzom tumorassociated fibroblasts. Oncol. Rep. 2019, 41, 405–414. 10.3892/or.2018.6783. [DOI] [PubMed] [Google Scholar]
- Oberhammer F. A.; Hochegger K.; Fröschl G.; Tiefenbacher R.; Pavelka M. Chromatin condensation during apoptosis is accompanied by degradation of lamin A+B, without enhanced activation of cdc2 kinase. J. Cell Biol. 1994, 126, 827–837. 10.1083/jcb.126.4.827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lambert A. W.; Pattabiraman D. R.; Weinberg R. A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. 10.1016/j.cell.2016.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pollard T. D.; Cooper J. A. Actin, a central player in cell shape and movement. Science 2009, 326, 1208–1212. 10.1126/science.1175862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li T.; Guo H.; Song Y.; Zhao X.; Shi Y.; Lu Y.; Hu S.; Nie Y.; Fan D.; Wu K. Loss of vinculin and membrane-bound β-catenin promotes metastasis and predicts poor prognosis in colorectal cancer. Mol. Canc. 2014, 13, 263. 10.1186/1476-4598-13-263. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Ferretti A. C.; Hidalgo F.; Tonucci F. M.; Almada E.; Pariani A.; Larocca M. C.; Favre C. Metformin and glucose starvation decrease the migratory ability of hepatocellular carcinoma cells: targeting AMPK activation to control migration. Sci. Rep. 2019, 9, 2815. 10.1038/s41598-019-39556-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hakimee H.; Hutamekalin P.; Tanasawet S.; Chonpathompikunlert P.; Tipmanee V.; Sukketsiri W. Metformin Inhibit Cervical Cancer Migration by Suppressing the FAK/Akt Signaling Pathway. Asian Pac. J. Cancer Prev. APJCP 2019, 20, 3539–3545. 10.31557/apjcp.2019.20.12.3539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collins M. P.; Stransky L. A.; Forgac M. AKT Ser/Thr kinase increases V-ATPase-dependent lysosomal acidification in response to amino acid starvation in mammalian cells. J. Biol. Chem. 2020, 295, 9433–9444. 10.1074/jbc.ra120.013223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sacco F.; Silvestri A.; Posca D.; Pirrò S.; Gherardini P. F.; Castagnoli L.; Mann M.; Cesareni G. Deep Proteomics of Breast Cancer Cells Reveals that Metformin Rewires Signaling Networks Away from a Pro-growth State. Cell Syst. 2016, 2, 159–171. 10.1016/j.cels.2016.02.005. [DOI] [PubMed] [Google Scholar]
- Chang H.-Y.; Huang T.-C.; Chen N.-N.; Huang H.-C.; Juan H.-F. Combination therapy targeting ectopic ATP synthase and 26S proteasome induces ER stress in breast cancer cells. Cell Death Dis. 2014, 5, e1540 10.1038/cddis.2014.504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin W.-T.; Hung W.-N.; Yian Y.-H.; Wu K.-P.; Han C.-L.; Chen Y.-R.; Chen Y.-J.; Sung T.-Y.; Hsu W.-L. Multi-Q: a fully automated tool for multiplexed protein quantitation. J. Proteome Res. 2006, 5, 2328–2338. 10.1021/pr060132c. [DOI] [PubMed] [Google Scholar]
- Subhash V. V.; Yeo M. S.; Wang L.; Tan S. H.; Wong F. Y.; Thuya W. L.; Tan W. L.; Peethala P. C.; Soe M. Y.; Tan D. S. P.; Padmanabhan N.; Baloglu E.; Shacham S.; Tan P.; Koeffler H. P.; Yong W. P. Anti-tumor efficacy of Selinexor (KPT-330) in gastric cancer is dependent on nuclear accumulation of p53 tumor suppressor. Sci. Rep. 2018, 8, 12248. 10.1038/s41598-018-30686-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



