The incidence of oral squamous cell carcinoma (OSCC) is rising rapidly in developed countries, posing a growing challenge due to the poor management of this type of malignancy at present. In this study, we profiled tumor suppressive microRNAs (miRNAs) that are silenced by DNA hypermethylation in OSCC using a function-based screening approach. This approach employed a cell proliferation assay for 327 synthetic miRNAs in two OSCC cell lines. Among the 110 miRNAs identified in this set that exhibited inhibitory properties, we compared DNA methylation and expression status in a wider panel of OSCC cell lines and primary tumor tissues, resulting in the identification of miR-218 and miR-585 as functionally significant miRNA genes that are frequently silenced in OSCC by DNA hypermethylation. Ectopic expression of miR-218 and miR-585 in OSCC cells lacking endogenous expression reduced cell growth in part through caspase-mediated apoptosis. Notably, miR-218 reduced levels of the rapamycin-insensitive component of mTOR, Rictor, in a manner associated with a suppression of Akt S473 phosphorylation. Together our findings define miR-585 as a tumor suppressive function that is often epigenetically silenced in OSCC, and they identify Rictor as a novel target of miR-218, suggesting that activation of the mTOR-Akt signaling pathway induced by Rictor contributes centrally to oral carcinogenesis. Cancer Res; 71(17); 5765–78. ©2011 AACR.

MicroRNAs (miRNAs) are an abundant class of endogenous, small, non-coding RNAs, the products of which are small single-stranded RNAs of 19 to 22 nucleotides with a primary role in posttranscriptional silencing generally through binding to the 3′-UTR of protein-coding transcripts, in turn triggering mRNA degradation or translational repression (1). In human cancers, the expression of miRNAs is generally downregulated in malignant tissues compared with the corresponding nonmalignant tissues (2, 3), suggesting the deregulation of miRNA expression and the contribution of miRNAs to the multistep processes of carcinogenesis either as oncogenes or as tumor-suppressor genes (TSG; refs. 4, 5). Among various epigenetic mechanisms of cancer-related gene-silencing, DNA hypermethylation of CpG sites within CpG-islands is known to lead to the inactivation of many TSGs (6) and several tumor-suppressive miRNAs (TS-miRNAs; ref. 7). In fact, DNA methylation-mediated downregulation of miRNAs by proximal CpG-islands has been described by a number of groups including ours (8–10).

Oral cancer, predominantly oral squamous cell carcinoma (OSCC), is the most common head and neck neoplasm, affecting approximately 270,000 people worldwide in 2002 (11). In Japan, OSCC is relatively common, accounting for more than 5,500 deaths in 2003 (12). The carcinogenesis, including OSCC (13), is considered to arise through the progressive accumulation of multiple genetic abnormalities, which may impair the functions of oncogenes or TSGs that play a crucial role in the development of this disease. In addition, evidence has emerged that epigenetic mechanisms, such as altered DNA methylation patterns, play a significant role in the silencing of TSGs and contribute to malignant transformation during oral carcinogenesis (14).

In this study, we identified and characterized TS-miRNAs frequently silenced by DNA hypermethylation in OSCC and their targets using function-based screening with a cell proliferation assay for 327 synthetic miRNAs and a series of sequential analyses of DNA methylation and expression in OSCC cell lines and primary tumors. The function-based screening presented in this paper is a powerful tool for exploring a large number of double-stranded RNAs (dsRNAs) having tumor-suppressive effects, including TS-miRNAs and siRNAs, as therapeutic agents for several types of cancer cells directly (15, 16). Through this approach, 2 epigenetically silenced TS-miRNAs in OSCC, miR-218 and miR-585, which are located within the intron of SLIT2 and SLIT3, respectively, and a novel target of miR-218, Rictor, were identified in our study. This study is the first to show clearly that the tumor-suppressive activity of miR-585 is epigenetically silenced in OSCC and that miR-218 targets Rictor inducing the activation of a TOR-Akt signaling pathway.

Cell lines and primary tumor samples

Derivations of and culture conditions for cell lines were reported previously (17). OSCC cell lines were authenticated in our previous studies of array-CGH analyses (17). RT7, human oral keratinocytes immortalized by TERT and human papillomavirus (HPV) type 16 E6/E7 oncogenes, was kindly provided by Dr N. Kamata (Hiroshima University Faculty of Dentistry, Japan). To analyze the restoration of genes, cells were cultured with or without 10 μmol/L of 5-aza 2′-deoxycytidine (5-aza-dCyd) for 5 days. A total of 15 frozen primary samples were obtained from OSCC patients treated at Tokyo Medical and Dental University with written consent from each patient in the formal style and after approval by the local ethics committee.

Transfection with synthetic miRNAs and siRNAs

A total of 10 nmol/L of dsRNA mimicking human mature miRNAs or control nonspecific miRNA (Ambion; Thermo Scientific Dharmacon), and 20 nmol/L of target-specific siRNAs for PIK3CA (Dharmacon) and Rictor (Invitrogen) or their control nonspecific siRNA were transfected individually into OSCC cells using Lipofectamine RNAiMAX (Invitrogen). The numbers of viable cells were assessed by the colorimetric water-soluble tetrazolium salt (WST) assay (Cell counting kit-8; Dojindo Laboratories). The cell cycle was evaluated by a fluorescence-activated cell sorting (FACS) analysis as described elsewhere (17).

Methylation analysis

Each of gene and CpG-island was searched miRBase database (release April 17, 2011; ref. 18), UCSC Genome Browser on Human February 2009 Assembly (hg19; ref. 19) and PubMed (20). The combined bisulfite restriction analysis (COBRA) and the bisulfite-sequencing analysis, using primer sets designed to amplify regions of interest (Supplementary Table S1), were done as described elsewhere (8).

Real-time reverse transcription-PCR

Real-time reverse transcription PCR (RT-PCR) was done as described elsewhere (8).

miRNA target predictions, Western blotting, and luciferase activity assay

Predicted targets for miRNAs and their target sites were analyzed using Microcosm Targets (21), miRanda (22), and TargetScan (23). Anti-caspase-3, anti-cleaved caspase-3, anti-cleaved PARP, anti-Akt, anti-phospho-Akt (Ser-473), anti-Rictor (Cell Signaling Technology) and anti-PIK3CA (Upstate Biotechnology) rabbit polyclonal antibodies were used in Western blotting. Luciferase constructs were made by ligating oligonucleotides containing the 3′-UTR target sites downstream of the luciferase gene in the pMIR-REPORT luciferase vector (Ambion). Luciferase activity was measured as described elsewhere (8).

Statistical analysis

Differences between subgroups were tested with the Mann–Whitney U test.

Function-based screening of TS-miRNA in OSCC cell lines

To identify TS-miRNAs in OSCC, we first examined 327 synthetic miRNAs mimicking human mature miRNAs by function-based screening in 2 OSCC cell lines, NA and SKN3. The strategy used and partial results obtained are shown in Figure 1A. Since the proliferation-inhibitory effect was made an index for the rating of tumor-suppressive activity in our function-based screening, relative cell growth ratios in Figure 1B and Supplementary Table S2 indicate effects of each synthetic miRNA on cell growth 5 days after transfection. In this first screening, 110 miRNAs, including known TS-miRNAs, such as miR-34 (24), miR-124 (9, 25), miR-193a (8), and miR-491 (15), showed remarkable inhibitory effects on cell growth (relative growth ratio < 0.5). Then we selected novel miRNAs with CpG-islands in the region 5′ upstream or around these genes and excluded miRNAs previously reported to correlate with DNA hypermethylation and their tumor-suppressive activity among 110 miRNAs by means of database analyses. From the first screening, 25 mature sequences of miRNAs emerged as candidates for new TS-miRNAs silenced by DNA hypermethylation in OSCC cell lines (Table 1).

Figure 1.

Strategy employed and results of a function-based screening of TS-miRNAs in OSCC cell lines. A, strategy used for the identification of epigenetically silenced TS-miRNAs in OSCC. B, result of function-based screening of TS-miRNAs in NA and SKN3 cells using Pre-miR miRNA Precursor Library-Human V2 (Ambion). Cells (1.0 × 104/well) were inoculated onto 24-well plates. Following 24 hours of incubation, 10 nmol/L of 327 dsRNA mimicking mature miRNA or control nonspecific miRNA was transfected individually into cells. The numbers of viable cells 5 days after transfection were evaluated by the WST assay in duplicate. Relative cell growth ratios were then calculated by normalization of each result to the cell numbers in control cells transfected with nonspecific miRNA (see Supplementary Table S2). The lower solid arrow indicates the 327 miRNAs examined. The top and middle closed arrows indicate 98 miRNAs and 53 miRNAs which showed marked growth inhibitory effects in NA and SKN3 cells, respectively (growth ratio < 0.5). C, summary of the DNA methylation status of CpG-islands around 25 mature sequences of miRNAs located at 26 loci including 35 CpG-islands in 18 OSCC cell lines and RT7 as a control determined by COBRA. PCR products used for COBRA were digested with BstUI, TaqI, or HhaI, and electrophoresed (see Figure 2A and B, Tables 1, 2, Supplementary Fig. S2, Supplementary Table S1, and data not shown). Black, gray, white, and striped boxes indicate complete, partial, and no digestion by restriction enzymes, and not determined, respectively.

Figure 1.

Strategy employed and results of a function-based screening of TS-miRNAs in OSCC cell lines. A, strategy used for the identification of epigenetically silenced TS-miRNAs in OSCC. B, result of function-based screening of TS-miRNAs in NA and SKN3 cells using Pre-miR miRNA Precursor Library-Human V2 (Ambion). Cells (1.0 × 104/well) were inoculated onto 24-well plates. Following 24 hours of incubation, 10 nmol/L of 327 dsRNA mimicking mature miRNA or control nonspecific miRNA was transfected individually into cells. The numbers of viable cells 5 days after transfection were evaluated by the WST assay in duplicate. Relative cell growth ratios were then calculated by normalization of each result to the cell numbers in control cells transfected with nonspecific miRNA (see Supplementary Table S2). The lower solid arrow indicates the 327 miRNAs examined. The top and middle closed arrows indicate 98 miRNAs and 53 miRNAs which showed marked growth inhibitory effects in NA and SKN3 cells, respectively (growth ratio < 0.5). C, summary of the DNA methylation status of CpG-islands around 25 mature sequences of miRNAs located at 26 loci including 35 CpG-islands in 18 OSCC cell lines and RT7 as a control determined by COBRA. PCR products used for COBRA were digested with BstUI, TaqI, or HhaI, and electrophoresed (see Figure 2A and B, Tables 1, 2, Supplementary Fig. S2, Supplementary Table S1, and data not shown). Black, gray, white, and striped boxes indicate complete, partial, and no digestion by restriction enzymes, and not determined, respectively.

Close modal
Table 1.

Summary of 25 miRNAs (35 CpG islands, 26 loci) selected as candidates for novel TS-miRNA silenced by DNA hypermethylation in function-based screening using OSCC cell lines and Pre-miR miRNA Precursor Library - Human V2 (Ambion)

Number of candidate miRNAsmiRNAsmiRNA genesLocusHost genesClustered miRNAsNumber of CpG island in this analysisDistance of CpG island from the most 5′ mature miRNA sequenceMethylation frequency (total %)
hsa-let-7i hsa-let-7i 12q14.1 3.7 
hsa-miR-345 hsa-miR-345 14q32.2 523 16.7 
hsa-miR-375 hsa-miR-375 2q35 68.9 
hsa-miR-365 hsa-miR-365-1 16p13.12 miR-193b 6067 22.2 
  hsa-miR-365-2 17q11.2 miR-193a 15,053  
hsa-miR-432* hsa-miR-432 14q32.31 miR-337/665/431/433/127/432/136 960 ND 
hsa-miR-409-3p hsa-miR-409 14q32.31 miR-409-5p(miR-485/453/154/496/377/541/412/369/410/656) 7a ND 
hsa-miR-409-5p hsa-miR-409 14q32.31 miR-409-3p(miR-485/453/154/496/377/541/412/369/410/656) 7a ND 
hsa-miR-497 hsa-miR-497 17p13.1 miR-195 4,060 78.7 
hsa-miR-22 hsa-miR-22 17p13.3 C17orf92 1,924 0.0 
10 hsa-miR-28 hsa-miR-28 3q28 LPP 10 53,4218 22.2 
11 hsa-miR-139 hsa-miR-139 11q13.4 PDE2A 11 27,049 8.3 
12 hsa-miR-149 hsa-miR-149 2q37.3 GPC1 12 ND 
13 hsa-miR-208a hsa-miR-208a 14q11.2 MYH6 13 1,482 ND 
14 hsa-miR-326 hsa-miR-326 11q13.4 ARRB1 14 15,995 27.8 
15 hsa-miR-383 hsa-miR-383 8p22 SGCZ 15 61,6781 63.9 
16 hsa-miR-423 hsa-miR-423 17q11.2 CCDC55 16 87 0.0 
17 hsa-miR-455 hsa-miR-455 9q32 COL27A1 17 54,692 18.1 
      18 53,696  
      19 53,192  
      20 41,346  
18 hsa-miR-483 hsa-miR-483 11p15.5 IGF2 21 3,513 75.3 
19 hsa-miR-486-5p hsa-miR-486 8p11.21 ANK1 22 235,315 45.8 
      23 215,510  
      24 167,723  
      25 136,850  
      26 106,530  
      27 65,110  
      28 41,230  
20 hsa-miR-491 hsa-miR-491 9p21.3 KIAA1797 29 31,468 0.0 
24 hsa-miR-24 hsa-miR-24-1 9q22.32 C9orf3 miR-23b/27b 30 ND 
  hsa-miR-24-2 19p13.12 miR-23a/27a 31 6,050  
21 hsa-miR-218 hsa-miR-218-1 4p15.31 SLIT2 32 273,030 57.9 
  hsa-miR-218-2 5q35.1 SLIT3 miR-585 33 532,170  
22 hsa-miR-191hsa-miR-191 3q21.31 DALRD3 miR-425-3p 34a 184 0.0 
23 hsa-miR-425-3p hsa-miR-425 3p21.31 DALRD3 miR-191* 34a 659 0.0 
25 hsa-miR-489 hsa-miR-489 7q21.3 CALCR miR-653 35 90,755 77.8 
Number of candidate miRNAsmiRNAsmiRNA genesLocusHost genesClustered miRNAsNumber of CpG island in this analysisDistance of CpG island from the most 5′ mature miRNA sequenceMethylation frequency (total %)
hsa-let-7i hsa-let-7i 12q14.1 3.7 
hsa-miR-345 hsa-miR-345 14q32.2 523 16.7 
hsa-miR-375 hsa-miR-375 2q35 68.9 
hsa-miR-365 hsa-miR-365-1 16p13.12 miR-193b 6067 22.2 
  hsa-miR-365-2 17q11.2 miR-193a 15,053  
hsa-miR-432* hsa-miR-432 14q32.31 miR-337/665/431/433/127/432/136 960 ND 
hsa-miR-409-3p hsa-miR-409 14q32.31 miR-409-5p(miR-485/453/154/496/377/541/412/369/410/656) 7a ND 
hsa-miR-409-5p hsa-miR-409 14q32.31 miR-409-3p(miR-485/453/154/496/377/541/412/369/410/656) 7a ND 
hsa-miR-497 hsa-miR-497 17p13.1 miR-195 4,060 78.7 
hsa-miR-22 hsa-miR-22 17p13.3 C17orf92 1,924 0.0 
10 hsa-miR-28 hsa-miR-28 3q28 LPP 10 53,4218 22.2 
11 hsa-miR-139 hsa-miR-139 11q13.4 PDE2A 11 27,049 8.3 
12 hsa-miR-149 hsa-miR-149 2q37.3 GPC1 12 ND 
13 hsa-miR-208a hsa-miR-208a 14q11.2 MYH6 13 1,482 ND 
14 hsa-miR-326 hsa-miR-326 11q13.4 ARRB1 14 15,995 27.8 
15 hsa-miR-383 hsa-miR-383 8p22 SGCZ 15 61,6781 63.9 
16 hsa-miR-423 hsa-miR-423 17q11.2 CCDC55 16 87 0.0 
17 hsa-miR-455 hsa-miR-455 9q32 COL27A1 17 54,692 18.1 
      18 53,696  
      19 53,192  
      20 41,346  
18 hsa-miR-483 hsa-miR-483 11p15.5 IGF2 21 3,513 75.3 
19 hsa-miR-486-5p hsa-miR-486 8p11.21 ANK1 22 235,315 45.8 
      23 215,510  
      24 167,723  
      25 136,850  
      26 106,530  
      27 65,110  
      28 41,230  
20 hsa-miR-491 hsa-miR-491 9p21.3 KIAA1797 29 31,468 0.0 
24 hsa-miR-24 hsa-miR-24-1 9q22.32 C9orf3 miR-23b/27b 30 ND 
  hsa-miR-24-2 19p13.12 miR-23a/27a 31 6,050  
21 hsa-miR-218 hsa-miR-218-1 4p15.31 SLIT2 32 273,030 57.9 
  hsa-miR-218-2 5q35.1 SLIT3 miR-585 33 532,170  
22 hsa-miR-191hsa-miR-191 3q21.31 DALRD3 miR-425-3p 34a 184 0.0 
23 hsa-miR-425-3p hsa-miR-425 3p21.31 DALRD3 miR-191* 34a 659 0.0 
25 hsa-miR-489 hsa-miR-489 7q21.3 CALCR miR-653 35 90,755 77.8 

aSame CpG islands.

NOTE: These data were searched miRBase database (release April 17, 2011), UCSC Genome Browser on Human February 2009 Assembly (hg19) and PubMed.

Methylation and expression analyses of candidates in OSCC cell lines

Next, we explored the DNA methylation status of 35 CpG-islands around 25 miRNAs located at 26 loci in a panel of 18 OSCC cell lines and a normal counterpart, RT7, an immortalized human oral keratinocyte line, by COBRA (Fig. 1C). Since multiple copies of some mature miRNAs, as listed in Table 1, are transcribed from different loci, the number of mature forms of miRNAs is smaller than the number of genomic loci. In COBRA, on the basis of a comparison of mean values of methylation frequencies in each region examined, frequent DNA hypermethylation (>50% of OSCC lines) was found in only 6 of 25 miRNAs, that is, miR-218, miR-375, miR-383, miR-483, miR-489, and miR-497, although CpG-island hypermethylation on/around these miRNAs was also detected in RT7. We next investigated the consistency in the correlation between DNA methylation status in each region of these 6 miRNA genes by COBRA and their expression patterns in 18 OSCC cell lines and RT7 by TaqMan real-time RT-PCR (Table 2), and found that values were higher in some regions of miR-218-1 (50.0%–77.8%) and miR-375 (100%) than other miRNA genes (0%–56.3%; Fig. 1A–C and Supplementary Fig. S1A–C). miR-218-1 and miR-218-2 are located at 4p15.31 and 5q35.1 within the intron of SLIT2 and SLIT3, respectively (Fig. 2A and B). Interestingly, miR-585, together with miR-218-2, is also located within the intron of SLIT3, and the expression patterns of miR-218 and miR-585 were similar to those of SLIT2 and SLIT3, respectively (Fig. 2B and C); note that the percentage of OSCC cell lines with significant downregulation of miR-218, miR-585, SLIT2, SLIT3, and miR-375 expression (<0.5-fold) was 44.4% (8/18), 55.6% (10/18), 61.1% (11/18), 77.8% (14/18), and 100% (18/18), respectively (Fig. 2C and Supplementary Fig. 1C). To determine the relationship between DNA hypermethylation in CpG-islands on/around miR-218-1, miR-218-2, and miR-375 and the downregulation of gene expression, we treated OSCC cell lines with 5-aza-dCyd, observing a remarkable restoration of the expression levels of these miRNAs and their host genes in 55.6% (10/18), 44.4% (8/18), 33.3% (6/18), 61.1% (11/18), and 100% (18/18) of OSCC cell lines, respectively (Fig. 2C and Supplementary Fig. 1D). Consistent with these results, CpG-island hypermethylation was also confirmed by bisulfite-sequencing in the OSCC lines lacking the expression of those candidate miRNAs, but not in RT7 and the cell lines expressing these miRNAs (Fig. 2A and B and Supplementary Fig. 1E). These results strongly suggest that DNA hypermethylation of CpG sites within CpG-islands on/around these genes deregulates their expressions in OSCC cell lines.

Figure 2.

Correlation between methylation and expression of miR-218 and the host genes SLIT2 and SLIT3 in OSCC cell lines. A and B, methylation analysis for miR-218-1/SLIT2 (A) and miR-218-2/SLIT3 (B). Top, these maps show intronic miRNAs, host genes, CpG-islands, CpG sites, and PCR products used for COBRA and bisulfite sequencing. White boxes, exons of SLIT2 and SLIT3; gray box, CpG-island; closed arrows, PCR products (primers, Supplementary Table S1); vertical tick marks, CpG sites; vertical arrows, restriction enzyme sites. Middle, results of COBRA in 18 OSCC cell lines and RT7. Arrows, unmethylated alleles; arrowheads, methylated alleles; stars, samples with significant restricted fragments from methylated alleles. The presence of restriction enzyme processing is indicated with a plus or minus sign above the results of COBRA. Bottom, bisulfite sequencing of RT7 and representative OSCC cell lines with (+) or without (−) miR-218 expression in PCR products examined by COBRA. Horizontal bars with arrowheads, PCR product; vertical arrows, restriction enzyme sites. Open and filled squares represent unmethylated and methylated CpG sites respectively, and each row represents a single clone. C, TaqMan real-time RT-PCR analysis for miR-218, miR-585, SLIT2, and SLIT3 in 18 OSCC cell lines and RT7. Top, expression levels of intronic miRNAs and host genes were based on the amount of target message relative to RNU6B and GAPDH, respectively, to normalize the initial input of total RNA. Bar graphs show the ratio of the expression level in each cell line to that in RT7. Bottom, restoration of the expression of these genes after treatment with 10 μmol/L 5-aza-dCyd for 5 days in 18 OSCC cell lines. Bar graphs show the ratio of the expression level in treated cells to that in untreated cells. D, confirmation of tumor-suppressive activities of 3 candidate miRNAs in HSC-2 and NA cells using dsRNAs purchased from different companies (Ambion and Dharmacon). The numbers of viable cells 5 days after transfection with 10 nmol/L of dsRNAs were assessed by WST assay. Bar graphs show the growth ratio of cell numbers in miR-218-, -375-, and -585-transfectants relative to those in control-transfectants. Each point represents the mean of triplicate determinations (bars, SD).

Figure 2.

Correlation between methylation and expression of miR-218 and the host genes SLIT2 and SLIT3 in OSCC cell lines. A and B, methylation analysis for miR-218-1/SLIT2 (A) and miR-218-2/SLIT3 (B). Top, these maps show intronic miRNAs, host genes, CpG-islands, CpG sites, and PCR products used for COBRA and bisulfite sequencing. White boxes, exons of SLIT2 and SLIT3; gray box, CpG-island; closed arrows, PCR products (primers, Supplementary Table S1); vertical tick marks, CpG sites; vertical arrows, restriction enzyme sites. Middle, results of COBRA in 18 OSCC cell lines and RT7. Arrows, unmethylated alleles; arrowheads, methylated alleles; stars, samples with significant restricted fragments from methylated alleles. The presence of restriction enzyme processing is indicated with a plus or minus sign above the results of COBRA. Bottom, bisulfite sequencing of RT7 and representative OSCC cell lines with (+) or without (−) miR-218 expression in PCR products examined by COBRA. Horizontal bars with arrowheads, PCR product; vertical arrows, restriction enzyme sites. Open and filled squares represent unmethylated and methylated CpG sites respectively, and each row represents a single clone. C, TaqMan real-time RT-PCR analysis for miR-218, miR-585, SLIT2, and SLIT3 in 18 OSCC cell lines and RT7. Top, expression levels of intronic miRNAs and host genes were based on the amount of target message relative to RNU6B and GAPDH, respectively, to normalize the initial input of total RNA. Bar graphs show the ratio of the expression level in each cell line to that in RT7. Bottom, restoration of the expression of these genes after treatment with 10 μmol/L 5-aza-dCyd for 5 days in 18 OSCC cell lines. Bar graphs show the ratio of the expression level in treated cells to that in untreated cells. D, confirmation of tumor-suppressive activities of 3 candidate miRNAs in HSC-2 and NA cells using dsRNAs purchased from different companies (Ambion and Dharmacon). The numbers of viable cells 5 days after transfection with 10 nmol/L of dsRNAs were assessed by WST assay. Bar graphs show the growth ratio of cell numbers in miR-218-, -375-, and -585-transfectants relative to those in control-transfectants. Each point represents the mean of triplicate determinations (bars, SD).

Close modal
Table 2.

Correlation between DNA hypermethylation status in each region of these 6 miRNA genes by COBRA and their expression patterns in OSCC cell lines and primary cases by quantitative real-time RT-PCR analysis

OSCC cell linesPrimary OSCC cases
miRNAsmiRNA genesRegions in COBRAMethylation frequency (%)aConsistency of methylation with downregulation (%)bMethylation frequency (%)aConsistency of methylation with downregulation (%)b
hsa-miR-489 hsa-miR-489 77.78 14 0.00   
hsa-miR-483 hsa-miR-483 100.00 18 16.67   
  100.00 18 16.67   
  ND     
  100.00 18 16.67   
  94.44 17 17.65   
  66.67 12 16.67   
  72.22 13 15.38   
  ND     
  50.00 22.22   
  10 44.44    
  11 50.00 11.11   
hsa-miR-497 hsa-miR-195/497 ND     
  94.44 17 47.06   
  100.00 18 50.00   
  88.89 16 50.00   
  88.89 16 56.25   
  0.00   
hsa-miR-375 hsa-miR-375 88.89 16 100.00 16   
  83.33 15 100.00 15   
  72.22 13 100.00 13   
  11.11    
  83.33 15 100.00 15   
hsa-miR-383 hsa-miR-383 72.22 13 7.69   
  55.56 10 10.00   
  ND     
hsa-miR-218 hsa-miR-218-1 38.89  20.00 3/15 100.00 3/3 
  38.89  13.33 2/15 100.00 2/2 
  50.00 77.78 6.67 1/15 100.00 1/1 
  55.56 10 70.00 6.67 1/15 100.00 1/1 
  77.78 14 50.00 53.33 8/15 75.00 6/8 
  77.78 14 57.14 73.33 11/15 81.82 9/11 
  88.89 16 50.00 66.67 10/15 80.00 8/10 
  88.89 16 50.00 93.33 14/15 85.71 12/14 
  ND     
 hsa-miR-585/218-2 10 22.22  13.33 2/15 100.00 2/2 
  11 38.89  13.33 2/15 100.00 2/2 
  12 38.89  6.67 1/15 100.00 1/1 
  13 77.78 14 50.00 0/15 
hsa-miR-486-5p hsa-miR-486 88.89 16 31.25   
  ND     
  ND     
  ND     
  33.33    
  ND     
  ND     
  0.00 0.00   
  5.56    
  10 ND     
  11 88.89 16 31.25   
  12 22.22    
  13 27.78    
  14 100.00 18 27.78   
  15 ND     
hsa-miR-326 hsa-miR-326 ND     
  27.78    
hsa-miR-28 hsa-miR-28 5.56    
  0.00    
  44.44    
hsa-miR-365 hsa-miR-365-1 0.00    
  0.00    
  0.00    
 hsa-miR-365-2 0.00    
  61.11 11 54.55   
  72.22 13 53.85   
hsa-miR-455 hsa-miR-455 22.22    
  50.00 44.44   
  0.00 0.00   
  0.00 0.00   
  ND     
hsa-miR-345 hsa-miR-345 0.00 0.00   
  0.00 0.00   
  0.00 0.00   
  66.67 12 0.00   
hsa-miR-139 hsa-miR-139 0.00    
  0.00    
  0.00    
  0.00     
  33.33    
hsa-let-7i hsa-let-7i 11.11    
  0.00    
  0.00    
hsa-miR-191*/miR-425-3p hsa-miR-191/miR-425 0.00    
  0.00    
  0.00    
hsa-miR-22 hsa-miR-22 0.00    
  0.00    
hsa-miR-423 hsa-miR-423 0.00    
  0.00    
hsa-miR-491 hsa-miR-491 0.00    
  0.00    
hsa-miR-24 hsa-miR-24 ND     
  ND     
  ND     
hsa-miR-149 hsa-miR-149 ND     
  ND     
hsa-miR-208a hsa-miR-208a ND     
hsa-miR-409-3p/miR-409-5p hsa-miR-409 ND     
  ND     
hsa-miR-432hsa-miR-432 ND     
OSCC cell linesPrimary OSCC cases
miRNAsmiRNA genesRegions in COBRAMethylation frequency (%)aConsistency of methylation with downregulation (%)bMethylation frequency (%)aConsistency of methylation with downregulation (%)b
hsa-miR-489 hsa-miR-489 77.78 14 0.00   
hsa-miR-483 hsa-miR-483 100.00 18 16.67   
  100.00 18 16.67   
  ND     
  100.00 18 16.67   
  94.44 17 17.65   
  66.67 12 16.67   
  72.22 13 15.38   
  ND     
  50.00 22.22   
  10 44.44    
  11 50.00 11.11   
hsa-miR-497 hsa-miR-195/497 ND     
  94.44 17 47.06   
  100.00 18 50.00   
  88.89 16 50.00   
  88.89 16 56.25   
  0.00   
hsa-miR-375 hsa-miR-375 88.89 16 100.00 16   
  83.33 15 100.00 15   
  72.22 13 100.00 13   
  11.11    
  83.33 15 100.00 15   
hsa-miR-383 hsa-miR-383 72.22 13 7.69   
  55.56 10 10.00   
  ND     
hsa-miR-218 hsa-miR-218-1 38.89  20.00 3/15 100.00 3/3 
  38.89  13.33 2/15 100.00 2/2 
  50.00 77.78 6.67 1/15 100.00 1/1 
  55.56 10 70.00 6.67 1/15 100.00 1/1 
  77.78 14 50.00 53.33 8/15 75.00 6/8 
  77.78 14 57.14 73.33 11/15 81.82 9/11 
  88.89 16 50.00 66.67 10/15 80.00 8/10 
  88.89 16 50.00 93.33 14/15 85.71 12/14 
  ND     
 hsa-miR-585/218-2 10 22.22  13.33 2/15 100.00 2/2 
  11 38.89  13.33 2/15 100.00 2/2 
  12 38.89  6.67 1/15 100.00 1/1 
  13 77.78 14 50.00 0/15 
hsa-miR-486-5p hsa-miR-486 88.89 16 31.25   
  ND     
  ND     
  ND     
  33.33    
  ND     
  ND     
  0.00 0.00   
  5.56    
  10 ND     
  11 88.89 16 31.25   
  12 22.22    
  13 27.78    
  14 100.00 18 27.78   
  15 ND     
hsa-miR-326 hsa-miR-326 ND     
  27.78    
hsa-miR-28 hsa-miR-28 5.56    
  0.00    
  44.44    
hsa-miR-365 hsa-miR-365-1 0.00    
  0.00    
  0.00    
 hsa-miR-365-2 0.00    
  61.11 11 54.55   
  72.22 13 53.85   
hsa-miR-455 hsa-miR-455 22.22    
  50.00 44.44   
  0.00 0.00   
  0.00 0.00   
  ND     
hsa-miR-345 hsa-miR-345 0.00 0.00   
  0.00 0.00   
  0.00 0.00   
  66.67 12 0.00   
hsa-miR-139 hsa-miR-139 0.00    
  0.00    
  0.00    
  0.00     
  33.33    
hsa-let-7i hsa-let-7i 11.11    
  0.00    
  0.00    
hsa-miR-191*/miR-425-3p hsa-miR-191/miR-425 0.00    
  0.00    
  0.00    
hsa-miR-22 hsa-miR-22 0.00    
  0.00    
hsa-miR-423 hsa-miR-423 0.00    
  0.00    
hsa-miR-491 hsa-miR-491 0.00    
  0.00    
hsa-miR-24 hsa-miR-24 ND     
  ND     
  ND     
hsa-miR-149 hsa-miR-149 ND     
  ND     
hsa-miR-208a hsa-miR-208a ND     
hsa-miR-409-3p/miR-409-5p hsa-miR-409 ND     
  ND     
hsa-miR-432hsa-miR-432 ND     

aFrequency of cell lines or primary cases, in which DNA hypermethylation were detected by COBRA.

bFrequency of cell lines or primary cases, in which downregulation was consistent with DNA hypermethylation.

ND, not done.

Confirmation of dsRNA-induced growth inhibition in OSCC cell lines

To confirm the growth inhibitory effects of miR-218, miR-585, and miR-375 in the function-based screening, we used 2 synthetic miRNAs purchased from Ambion and Thermo Scientific Dharmacon to take into consideration the off-target effects of dsRNAs. The ectopic expression of these miRNAs in OSCC cell lines lacking their expressions by the transient transfection of dsRNA was evaluated by TaqMan real-time RT-PCR analysis (data not shown). Consistent with the results of the function-based screening (Supplementary Table S2), restoration of miR-218 expression significantly reduced cell growth in the OSCC cell lines tested (Fig. 2D). In addition, a growth inhibitory effect of miR-585 was clearly detected in Dharmacon product, whereas Ambion product of dsRNA mimicking miR-585 was not on the market and could not be examined. In our in vitro analysis, tumor-suppressive functions of miR-218 and miR-585 were confirmed. Ambion product of dsRNA mimicking miR-375 induced growth inhibition whereas Dharmacon product facilitated cell proliferation in both OSCC cell lines, and so miR-375 was excluded from candidates in this study.

Methylation and expression analyses of miR-218 and miR-585 in primary OSCCs

To determine whether the CpG-island hypermethylation of miR-218-1/SLIT2 and miR-218-2/miR-585/SLIT3 also occurs in primary OSCCs in a tumor-specific manner, the correlation between DNA methylation status and the expression patterns of these genes in 15 primary OSCCs and corresponding noncancerous oral mucosae was examined using COBRA and TaqMan real-time RT-PCR assay, respectively. In COBRA, the frequency of tumor-specific DNA hypermethylation in regions of miR-218-1 and miR-218-2/miR-585 was 6.7% to 93.4% and 6.7% to 13.3%, respectively, in primary OSCCs (Table 2, Supplementary Fig. 2A). Although the frequency of aberrant methylation at these CpG-islands was relatively low in primary tumors, expression levels of miR-218 and miR-585 in tumors as compared with paired normal oral mucosae were markedly reduced in 73.3% (11/15) and 66.7% (10/15) of primary OSCCs, respectively (< 0.5-fold expression, Supplementary Fig. 2B), and the consistency was comparatively high in these regions of miR-218-1 (75.0%–100%) and miR-218-2/miR-585 (100%). Although miR-218 expression had been reported to be specifically reduced in HPV-positive cell lines and tissues (26), all of OSCC cell lines and primary samples examined in this study were confirmed the negative HPV-status by genomic PCR (Supplementary Fig. S3). Thus, our results suggest that miR-218 and miR-585 are most likely TS-miRNAs frequently silenced through tumor-specific DNA hypermethylation in OSCC.

Tumor-suppressive effects of ectopic miR-218 and miR-585 expression on the growth of OSCC cell lines

We examined tumor-suppressive effects of miR-218 and miR-585 on HSC-2 and NA (Fig. 3A and B). Restoration of the expression of these miRNAs significantly reduced cell growth in both cell lines tested. Since a large number of transfectants were rounded and floating compared with the control counterpart at 5 days after transfection, we conducted a FACS analysis and a Western blotting for caspase-mediated apoptosis using both cell lines 72 hours after the transfection. In the FACS analysis, the effect of miR-218 on the cell cycle in HSC-2 cells was weak while the accumulation of NA cells in the G2/M phase was observed in miR-218-transfectants. Overexpression of miR-585 induced the accumulation of cells in the G2/M phase and sub-G1 phase in the HSC-2 and NA cell lines, respectively. However, the Western blotting showed that ectopic expression of miR-218 and miR-585 remarkably increased protein levels of caspase3, cleaved caspase3 and cleaved PARP in both cell lines (Fig. 3C), miR-218- and miR-585-induced reductions of cell growth in these OSCC cell lines were not inhibited by caspase inhibitors (Supplementary Fig. S4A). In contrast, the G2/M cell-cycle arrest was not confirmed by Western blotting for the phosphorylation status of Cdc2 and Chk1 in these transfectants (Supplementary Fig. S4B). These results, consistent with the previous report of miR-218-inducing apoptosis in vitro (27), suggest that miR-218 and miR-585 induce a reduction in cell growth at least in part through caspase-mediated apoptosis, whereas the cause of the cell death except the apoptosis in these transfectants remains unclear.

Figure 3.

Tumor-suppressive effects of miR-218 and miR-585 on OSCC cell lines, HSC-2 and NA, lacking their expression. A and B, growth curves, phase-contrast micrographs, and results of FACS analysis in HSC-2 (A) and NA (B) cells in which 10 nmol/L of dsRNA mimicking miR-218 or miR-585, or control nonspecific miRNA (ds-NC) was transfected. The numbers of viable cells after transfection were assessed by WST assay. Each data point represents the mean of triplicate determinations (bars, SD) in these experiments. Phase-contrast micrographs show OSCC cell lines cultured for 5 days after transfection. The size of the population in each phase of the cell cycle as assessed by FACS using OSCC cell lines 72 hours after transfection. C, the results of Western blotting of caspase3, cleaved caspase3, and cleaved PARP in HSC-2 and NA cells 48 hours after the transfection of each dsRNA.

Figure 3.

Tumor-suppressive effects of miR-218 and miR-585 on OSCC cell lines, HSC-2 and NA, lacking their expression. A and B, growth curves, phase-contrast micrographs, and results of FACS analysis in HSC-2 (A) and NA (B) cells in which 10 nmol/L of dsRNA mimicking miR-218 or miR-585, or control nonspecific miRNA (ds-NC) was transfected. The numbers of viable cells after transfection were assessed by WST assay. Each data point represents the mean of triplicate determinations (bars, SD) in these experiments. Phase-contrast micrographs show OSCC cell lines cultured for 5 days after transfection. The size of the population in each phase of the cell cycle as assessed by FACS using OSCC cell lines 72 hours after transfection. C, the results of Western blotting of caspase3, cleaved caspase3, and cleaved PARP in HSC-2 and NA cells 48 hours after the transfection of each dsRNA.

Close modal

Screening of predicted targets for miR-218 and miR-585 in OSCC cell lines

While 5 genes of EGFR-coamplified and overexpressed protein (ECOP), IκBs kinase (IKK-β), LIM and SH3 protein 1 (LASP1), paxillin (PXN), and Robo1 have also been reported as direct targets of miR-218 (28–32), the percentage of OSCC cell lines with notable upregulation of these genes (>2-fold expression) was 50.0% (9/18), 38.9% (7/18), 44.4% (8/18), 16.7% (3/18), and 11.1% (2/18), respectively (Fig. 4A). In addition, we also confirmed that their protein levels were decreased in miR-218-transfectants compared with their control counterparts (Fig. 4B).

Figure 4.

Identification and characterization of Rictor as a novel target of miR-218. A, expression analyses of 5 reported targets and one predicted target, Rictor, in 18 OSCC cell lines and RT7 using TaqMan real-time RT-PCR (top), and Western blotting (bottom). Expression levels of transcripts of these targets were based on the amount of target message relative to GAPDH to normalize the initial input of total RNA. Bar graphs show the ratio of the expression level in each cell line to that in RT7. B, identification of Rictor as a novel target of miR-218. Top, the results of Western blotting of 5 reported targets and Rictor in HSC-2 and NA cells 48 hours after the transfection of dsRNA mimicking miR-218 or control nonspecific miRNA (ds-NC). Middle, putative binding sites of miR-218 in the 3′-UTR region of Rictor. These sites were analyzed using Microcosm Targets (21), miRanda (22), and TargetScan (23). Bottom, luciferase assays of HSC-2 and NA cells 48 hours after cotransfection of pMIR-REPORT luciferase vectors containing a 3′-UTR target site of Rictor for miR-218, dsRNA mimicking miR-218, or control nonspecific miRNA, and pRL-CMV internal control vector. C, effects of miR-218 on Akt activation in OSCC cell lines with/without mutant forms of PIK3CA by Western blotting. Both cell lines, HSC-2 with PIK3CA mutation and NA without a genetic alteration of PIK3CA, EGFR, or PTEN, had been shown to be activated their PI3K–Akt signaling pathway (see Figure 4A; ref. 34). Top, the results of Western blotting in HSC-2 and NA cells 48 hours after transfection of dsRNA mimicking miR-218, control nonspecific miRNA, PIK3CA-specific siRNA, Rictor-specific siRNA, or control nonspecific siRNA (si-NC). To determine the increased phosphorylation of Akt protein at Ser-473, levels of total Akt protein in same samples were evaluated. Bottom, relative Akt activities in these transfectants. The quantification of each protein band in the result of Western blotting was done using LAS-3000 with MultiGauge software (Fuji film). The amount of phosphorylated Akt was normalized versus that of total Akt. Bar graphs show the ratio of the Akt activities in ds-miR-218-, si-PIK3CA-, and si-Rictor-transfectants relative to those in control transfectants. D, schema of the negative regulation of the mTORC2-dependent Akt signaling pathway by miR-218 in OSCC.

Figure 4.

Identification and characterization of Rictor as a novel target of miR-218. A, expression analyses of 5 reported targets and one predicted target, Rictor, in 18 OSCC cell lines and RT7 using TaqMan real-time RT-PCR (top), and Western blotting (bottom). Expression levels of transcripts of these targets were based on the amount of target message relative to GAPDH to normalize the initial input of total RNA. Bar graphs show the ratio of the expression level in each cell line to that in RT7. B, identification of Rictor as a novel target of miR-218. Top, the results of Western blotting of 5 reported targets and Rictor in HSC-2 and NA cells 48 hours after the transfection of dsRNA mimicking miR-218 or control nonspecific miRNA (ds-NC). Middle, putative binding sites of miR-218 in the 3′-UTR region of Rictor. These sites were analyzed using Microcosm Targets (21), miRanda (22), and TargetScan (23). Bottom, luciferase assays of HSC-2 and NA cells 48 hours after cotransfection of pMIR-REPORT luciferase vectors containing a 3′-UTR target site of Rictor for miR-218, dsRNA mimicking miR-218, or control nonspecific miRNA, and pRL-CMV internal control vector. C, effects of miR-218 on Akt activation in OSCC cell lines with/without mutant forms of PIK3CA by Western blotting. Both cell lines, HSC-2 with PIK3CA mutation and NA without a genetic alteration of PIK3CA, EGFR, or PTEN, had been shown to be activated their PI3K–Akt signaling pathway (see Figure 4A; ref. 34). Top, the results of Western blotting in HSC-2 and NA cells 48 hours after transfection of dsRNA mimicking miR-218, control nonspecific miRNA, PIK3CA-specific siRNA, Rictor-specific siRNA, or control nonspecific siRNA (si-NC). To determine the increased phosphorylation of Akt protein at Ser-473, levels of total Akt protein in same samples were evaluated. Bottom, relative Akt activities in these transfectants. The quantification of each protein band in the result of Western blotting was done using LAS-3000 with MultiGauge software (Fuji film). The amount of phosphorylated Akt was normalized versus that of total Akt. Bar graphs show the ratio of the Akt activities in ds-miR-218-, si-PIK3CA-, and si-Rictor-transfectants relative to those in control transfectants. D, schema of the negative regulation of the mTORC2-dependent Akt signaling pathway by miR-218 in OSCC.

Close modal

To explore novel oncogenic targets of miR-218 and miR-585 in OSCC cell lines, we used the algorithms, MicrocosmTargets, miRanda, and TargetScan, and selected rapamycin-insensitive companion of mTOR (Rictor) as miR-218-targets and jun B proto-oncogene (JunB) as miR-585-targets. The expression of Rictor or JunB was frequently upregulated in 38.9% (7/18) and 44.4% (8/18) of OSCC cell lines, respectively, compared with RT7 (>2-fold expression; Fig. 4A and Supplementary Fig. S5A). In Western blotting of Rictor and JunB, their protein levels were markedly reduced in miR-218- and miR-585-transfectants, respectively, compared with their control counterparts (Fig. 4B and Supplementary Fig. S5B). To further determine whether the predicted target sites of miR-218 and miR-585 in the 3′-UTR of mRNAs of Rictor and JunB, respectively (Fig. 4B and Supplementary Fig. S5C), were responsible for the translational regulation by dsRNA mimicking these miRNAs, we next conducted luciferase assays with vectors containing these 3′-UTR target sites downstream of the luciferase reporter gene. In this analysis, we observed a statistically significant reduction of luciferase activity in a vector containing the target site of Rictor, but not JunB (Fig. 4B and Supplementary Fig. S5D). These findings, together with the results of Western blotting, suggest Rictor and JunB to be a novel direct target of miR-218 and an indirect target of miR-585, respectively. Therefore, we focused on Rictor as the most likely target of miR-218, and conducted further analyses to explore the underlying molecular mechanisms of oral carcinogenesis.

Rictor, together with the mammalian target of rapamycin (mTOR) kinase, forms mTOR complex 2 (mTORC2), and the Rictor-mTOR complex directly regulates the phosphorylation of Akt at Ser-473, resulting in cell growth (33). We have previously reported that the phosphorylation of Akt at Ser-473 and Thr-308 was markedly increased in a HSC-2 cell line harboring a missense mutation in the PIK3CA gene, A3140G in exon 20, corresponding to the amino acid change H1047R (34). Interestingly, although NA cells without a genetic alteration of PIK3CA, EGFR, or PTEN also showed phosphorylated Akt, it has been unknown why the phosphorylation increased in this cell line. The phosphorylation status of Akt at Ser-473 in 18 OSCC cell lines and RT7 were shown in Figure 4A. To investigate whether Rictor might be associated with growth inhibitory effects of miR-218 in OSCC cells, we analyzed the phosphorylation of Akt and cell proliferation in HSC-2 and NA cells transfected with or without dsRNAs mimicking miR-218 or specific siRNAs for Rictor and PIK3CA (Fig. 4C). The treatment with PIK3CA- or Rictor-specific siRNA significantly inhibited cell growth in HSC-2 and NA cells whereas we found no noticeable differences in cell growth ratio of each transfectant in both cell lines (Supplementary Fig. S6). In Western blotting, the marked inhibition of the phosphorylation of Akt at Ser-473 in HSC-2 cells was observed in the treatment of RNA interference of PIK3CA, but not in restoration of miR-218 expression and knockdown of Rictor expression, suggesting that phosphatidylinositol 3-kinase (PI3K) seems mainly to regulate the phospho-Akt activation in HSC-2 cells with a mutant form of PIK3CA. On the other hand, a notable reduction in the phosphorylated Akt was detected in NA cells at 48 hours after the transient transfection of dsRNAs mimicking miR-218 or specific siRNA for Rictor compared with specific siRNA for PIK3CA, suggesting Akt to be significantly activated and phosphorylated by the TOR-Akt signaling pathway, not PI3K-Akt signaling pathway in NA cells without a mutant form of PIK3CA, EGFR, or PTEN. Our results clearly showed that miR-218 acts as a suppressor of the TOR-Akt signaling pathway, independently of the PI3K-Akt signaling pathway, in OSCC (Fig. 4D).

Here, we clearly identified miR-218 and miR-585 as TS-miRNAs silenced through tumor-specific DNA hypermethylation in oral cancer and characterized miR-218 targeting Rictor and inhibiting the phosphorylation of Akt at Ser-473 in a OSCC cell line without a mutant form of PIK3CA, EGFR, or PTEN. In this study, we carried out function-based screening using 327 dsRNAs mimicking mature miRNAs to identify TS-miRNAs having remarkable inhibitory effects on the growth of OSCC cell lines, although expression-based and DNA methylation-based screening had been successfully done previously in OSCC (8) and hepatocellular carcinoma (9), respectively. Since several known TS-miRNAs, such as miR-34 (24), miR-124 (9, 25), miR-193a (8), and miR-491 (15), were actually identified through this approach, function-based screening would be suitable for exploring dsRNAs, including miRNAs and siRNAs, as therapeutic agents for several types of cancer cells. The tumor-suppressive function of candidate miRNAs eventually identified in this screening was reevaluated using 2 kinds of dsRNA purchased independently to take account of off-target effects, known to complicate the interpretation of phenotypic effects in gene-silencing experiments using siRNAs (35).

We consider that the hypermethylation of CpG-islands on/around miRNA genes a good marker to explore novel epigenetically silenced TS-miRNAs, similar to classic TSGs in several types of cancer, and have already reported miR-137, miR-193a, miR-124, and miR-203 as epigenetically silenced TS-miRNAs (8, 9). In this study, a second screening, combining DNA methylation and expression analyses in a panel of OSCC cell lines, resulted in the identification of miR-218 and miR-585 as prime candidates for TS-miRNAs silenced by DNA hypermethylation in OSCC. Our study is the first to show that miR-218 and miR-585 were frequently silenced by DNA hypermethylation in OSCC. This miRNA gene is located at 4p15.31 (miR-218-1) and 5q35.1 (miR-218-2) in introns of 2 host genes, SLIT2 and SLIT3, respectively, and miR-585, together with miR-218-2, is also located within the intron of SLIT3 at 5q35.1. Copy number losses at these loci and the downregulated expressions of miR-218, SLIT2, and SLIT3 were reported in some types of cancer (36) although we found no homozygous loss in these regions in our previous studies of genomewide copy-number aberrations in 39 OSCC cell lines by array-CGH analyses (17, 37). Frequent epigenetic inactivation of SLIT2 and SLIT3 was also described in several cancers (38, 39). Although, in our study, the expression levels of these miRNAs and their host genes were remarkably restored in OSCC cell lines treated with 5-aza-dCyd, it was reported that the treatment of this inhibitor of DNA methylation did not reactivate the downregulated expression of SLITs in cervical cancer (40), suggesting a complex mechanism of inactivation in Slits in some types of cancers. Very recently, the downreguration of miR-218 and its inhibitory effects on cell proliferation and invasion have been shown in several types of cancer, including HPV-positive cell lines and tissues (26–29, 31, 32, 36, 41), and in the present study, we confirmed the tumor-suppressive functions and the mechanism of action of this miRNA in OSCC. Regarding miR-585, however, this is the first report that this is a novel TS-miRNA frequently silenced by tumor-specific DNA hypermethylation in OSCC, suggesting a crucial role, similar to miR-218, in human cancers.

In this study, we successfully identified a possible direct target of miR-218, Rictor, although ECOP, IKK-β, LASP1, PXN, and Robo1 have also been reported as direct targets of this miRNA. Since ECOP and IKK-β are components of the NF-κB pathway, our findings of caspase-mediated apoptosis in cells overexpressing miR-218 strongly support early reports of a correlation between miR-218 and targets (28, 29). LASP1, PXN, and Robo1 are known to be associated with cell adhesion, mobility, and migration, and their overexpression induced by miR-218 suppression seem to enhance tumor invasion and metastasis (30–32). Interestingly, although Slit-Robo signaling has been described to facilitate tumor cell migration, Robo1 targeted by miR-218 is known to be a receptor for Slit which is the host gene of miR-218 and shown to form a negative feedback loop involving Slit, miR-218, and Robo1 (27, 32). In our expression analysis, the percentage of OSCC cell lines with remarkable downregulation of miR-218, miR-585, SLIT2, and SLIT3 (< 0.5-fold), expression was 44.4% (8/18), 55.6% (10/18), 61.1% (11/18), and 77.8% (14/18), respectively, whereas that with notable upregulation of these genes (>2-fold expression) was 38.9% (7/18), 11.1% (2/18), 33.3% (6/18), and 0% (0/18), respectively. Since the biological and functional significances of Slits-miR-218-Robo signaling in cancer remains largely unknown, further study is needed for the characterization of this signaling.

Rictor is a component of the mTOR-containing complex mTORC2, which directly regulates the phosphorylation of Akt at Ser-473 (33). Knockdown of Rictor or mTOR in colon cancer was shown to inhibit cancer cell proliferation in vitro and tumor formation in vivo (42), and the selective requirement of Rictor for the development of prostate cancer induced by a loss of Pten in mice was shown (43). These results including our findings strongly support the notion that the silencing of miR-218 through the hypermethylation of CpG-islands around this miRNA is likely to be an important mechanism of carcinogenesis and cancer progression at least partly involving the activation of mTORC2-Akt signaling in OSCC. In this study, we analyzed the effects of miR-218 on the phosphorylation of Akt using 2 representative OSCC cell lines; HSC-2, with a missense mutation in the PIK3CA gene and activated PI3K-Akt signaling pathway, and NA, without genetic alterations of PIK3CA, EGFR, or PTEN (34). Although it has been unknown why the phosphorylation of Akt is increased in NA cells, our findings clearly showed miR-218 to act as a suppressor of the TOR-Akt signaling pathway, independently of the PI3K-Akt signaling pathway, in OSCC, and that the activation of this signaling pathway through methylation-mediated silencing of miR-218 may contribute to the pathogenesis of OSCC.

In conclusion, we described here the identification of 2 TS-miRNAs, miR-218 and miR-585, frequently silenced by DNA hypermethylation in OSCC, using function-based screening and a series of sequential analyses. Moreover, we identified Rictor as a potential target of miR-218, suggesting that the epigenetic silencing of miR-218 and consequent activation of the TOR-Akt signaling pathway induced by the overexpression of Rictor contribute to oral carcinogenesis, and that targeting miR-218 may provide a novel strategy for the treatment of OSCC, although further studies in vivo will be needed to confirm that dsRNA mimicking miR-218 can act as a TS-miRNA.

No potential conflicts of interest were disclosed.

We thank Ayako Takahashi and Rumi Mori for technical assistance.

This study was supported in part by Grant-in-Aid for Scientific Research (A), (B), and (C), and Scientific Research on Priority Areas and Innovative Areas, and a Global Center of Excellence (GCOE) Program for International Research Center for Molecular Science in Tooth and Bone Diseases from the Ministry of Education, Culture, Sports, Science, and Technology, Japan; a Health and Labour Sciences Research Grant by the Ministry of Health, Labour and Welfare, Japan; and a grant from the New Energy and Industrial Technology Development Organization (NEDO).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ambros
V
. 
The functions of animal microRNAs
.
Nature
2004
;
431
:
350
5
.
2.
Miska
EA
. 
How microRNAs control cell division, differentiation and death
.
Curr Opin Genet Dev
2005
;
15
:
563
8
.
3.
Lu
J
,
Getz
G
,
Miska
EA
,
Alvarez-Saavedra
E
,
Lamb
J
,
Peck
D
, et al
MicroRNA expression profiles classify human cancers
.
Nature
2005
;
435
:
834
8
.
4.
Esquela-Kerscher
A
,
Slack
FJ
. 
Oncomirs - microRNAs with a role in cancer
.
Nat Rev Cancer
2006
;
6
:
259
69
.
5.
Osada
H
,
Takahashi
T
. 
MicroRNAs in biological processes and carcinogenesis
.
Carcinogenesis
2007
;
28
:
2
12
.
6.
Herman
JG
,
Baylin
SB
. 
Gene silencing in cancer in association with promoter hypermethylation
.
N Engl J Med
2003
;
349
:
2042
54
.
7.
Chuang
JC
,
Jones
PA
. 
Epigenetics and microRNAs
.
Pediatr Res
2007
;
61
:
24R
9R
.
8.
Kozaki
K
,
Imoto
I
,
Mogi
S
,
Omura
K
,
Inazawa
J
. 
Exploration of tumor-suppressive microRNAs silenced by DNA hypermethylation in oral cancer
.
Cancer Res
2008
;
68
:
2094
105
.
9.
Furuta
M
,
Kozaki
K
,
Tanaka
S
,
Arii
S
,
Imoto
I
,
Inazawa
J
. 
miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma
.
Carcinogenesis
2009
;
31
:
766
76
.
10.
Iorio
MV
,
Piovan
C
,
Croce
CM
. 
Interplay between microRNAs and the epigenetic machinery: An intricate network
.
Biochim Biophys Acta
2010
;
1799
:
10
2
.
11.
Parkin
DM
,
Bray
F
,
Ferlay
J
,
Pisani
P
. 
Global cancer statistics, 2002
.
CA Cancer J Clin
2005
;
55
:
74
108
.
12.
Nomura
K
,
Sobue
T
,
Nakatani
H
,
Maehara
M
,
Kiryu
Y
,
Tsukuma
H
, et al
The Editorial Board of Cancer Statistics in Japan. Number of deaths and proportional mortality rates from malignant neoplasms by site in Japan (2003)
.
Cancer Statistics in Japan
2005
;
36
9
.
Available from:
http://www.ncc.go.jp/en/statistics/index.html.
13.
Scully
C
,
Field
JK
,
Tanzawa
H
. 
Genetic aberrations in oral or head and neck squamous cell carcinoma (SCCHN): 1. Carcinogen metabolism, DNA repair and cell cycle control
.
Oral Oncol
2000
;
36
:
256
63
.
14.
Ha
PK
,
Califano
JA
. 
Promoter methylation and inactivation of tumour-suppressor genes in oral squamous-cell carcinoma
.
Lancet Oncol
2006
;
7
:
77
82
.
15.
Nakano
H
,
Miyazawa
T
,
Kinoshita
K
,
Yamada
Y
,
Yoshida
T
. 
Functional screening identifies a microRNA, miR-491 that induces apoptosis by targeting Bcl-X(L) in colorectal cancer cells
.
Int J Cancer
2010
;
127
:
1072
80
.
16.
Levy
C
,
Khaled
M
,
Iliopoulos
D
,
Janas
MM
,
Schubert
S
,
Pinner
S
, et al
Intronic miR-211 assumes the tumor suppressive function of its host gene in melanoma
.
Mol Cell
2010
;
40
:
841
9
.
17.
Suzuki
E
,
Imoto
I
,
Pimkhaokham
A
,
Nakagawa
T
,
Kamata
N
,
Kozaki
K
, et al
PRTFDC1, a possible tumor-suppressor gene, is frequently silenced in oral squamous-cell carcinomas by aberrant promoter hypermethylation
.
Oncogene
2007
;
26
:
7921
32
.
18.
Available from miRBase database (Release 17 April 2011)
: http://www.mirbase.org/index.shtml.
19.
Available from UCSC Genome Browser on Human February 2009 Assembly (hg19): http://genome.ucsc.edu/cgi-bin/hgGateway
.
20.
Available from PubMed
: http://www.ncbi.nlm.nih.gov/pubmed.
23.
Available from TargetScan
http://www.targetscan.org/.
24.
He
L
,
He
X
,
Lowe
SW
,
Hannon
GJ
. 
microRNAs join the p53 network–another piece in the tumour-suppression puzzle
.
Nat Rev Cancer
2007
;
7
:
819
22
.
25.
Pierson
J
,
Hostager
B
,
Fan
R
,
Vibhakar
R
. 
Regulation of cyclin dependent kinase 6 by microRNA 124 in medulloblastoma
.
J Neurooncol
2008
;
90
:
1
7
.
26.
Martinez
I
,
Gardiner
AS
,
Board
KF
,
Monzon
FA
,
Edwards
RP
,
Khan
SA
. 
Human papillomavirus type 16 reduces the expression of microRNA-218 in cervical carcinoma cells
.
Oncogene
2008
;
27
:
2575
82
.
27.
Alajez
NM
,
Lenarduzzi
M
,
Ito
E
,
Hui
AB
,
Shi
W
,
Bruce
J
, et al
MiR-218 suppresses nasopharyngeal cancer progression through downregulation of survivin and the SLIT2-ROBO1 pathway
.
Cancer Res
2011
;
71
:
2381
91
.
28.
Gao
C
,
Zhang
Z
,
Liu
W
,
Xiao
S
,
Gu
W
,
Lu
H
. 
Reduced microRNA-218 expression is associated with high nuclear factor kappa B activation in gastric cancer
.
Cancer
2010
;
116
:
41
9
.
29.
Song
L
,
Huang
Q
,
Chen
K
,
Liu
L
,
Lin
C
,
Dai
T
, et al
miR-218 inhibits the invasive ability of glioma cells by direct downregulation of IKK-beta
.
Biochem Biophys Res Commun
2010
;
402
:
135
40
.
30.
Chiyomaru
T
,
Enokida
H
,
Kawakami
K
,
Tatarano
S
,
Uchida
Y
,
Kawahara
K
, et al
Functional role of LASP1 in cell viability and its regulation by microRNAs in bladder cancer
.
Urol Oncol
2010
[Epub ahead of print]
.
31.
Wu
DW
,
Cheng
YW
,
Wang
J
,
Chen
CY
,
Lee
H
. 
Paxillin predicts survival and relapse in non-small cell lung cancer by microRNA-218 targeting
.
Cancer Res
2010
;
70
:
10392
401
.
32.
Tie
J
,
Pan
Y
,
Zhao
L
,
Wu
K
,
Liu
J
,
Sun
S
, et al
MiR-218 inhibits invasion and metastasis of gastric cancer by targeting the Robo1 receptor
.
PLoS Genet
2010
;
6
:
e1000879
.
33.
Hresko
RC
,
Mueckler
M
. 
mTOR.RICTOR is the Ser473 kinase for Akt/protein kinase B in 3T3-L1 adipocytes
.
J Biol Chem
2005
;
280
:
40406
16
.
34.
Kozaki
K
,
Imoto
I
,
Pimkhaokham
A
,
Hasegawa
S
,
Tsuda
H
,
Omura
K
, et al
PIK3CA mutation is an oncogenic aberration at advanced stages of oral squamous cell carcinoma
.
Cancer Sci
2006
;
97
:
1351
8
.
35.
Svoboda
P
. 
Off-targeting and other non-specific effects of RNAi experiments in mammalian cells
.
Curr Opin Mol Ther
2007
;
9
:
248
57
.
36.
Davidson
MR
,
Larsen
JE
,
Yang
IA
,
Hayward
NK
,
Clarke
BE
,
Duhig
EE
, et al
MicroRNA-218 is deleted and downregulated in lung squamous cell carcinoma
.
PLoS One
2010
;
5
:
e12560
.
37.
Nakamura
E
,
Kozaki
K
,
Tsuda
H
,
Suzuki
E
,
Pimkhaokham
A
,
Yamamoto
G
, et al
Frequent silencing of a putative tumor suppressor gene melatonin receptor 1 A (MTNR1A) in oral squamous-cell carcinoma
.
Cancer Sci
2008
;
99
:
1390
400
.
38.
Dallol
A
,
Da Silva
NF
,
Viacava
P
,
Minna
JD
,
Bieche
I
,
Maher
ER
, et al
SLIT2, a human homologue of the Drosophila Slit2 gene, has tumor suppressor activity and is frequently inactivated in lung and breast cancers
.
Cancer Res
2002
;
62
:
5874
80
.
39.
Dickinson
RE
,
Dallol
A
,
Bieche
I
,
Krex
D
,
Morton
D
,
Maher
ER
, et al
Epigenetic inactivation of SLIT3 and SLIT1 genes in human cancers
.
Br J Cancer
2004
;
91
:
2071
8
.
40.
Narayan
G
,
Goparaju
C
,
Arias-Pulido
H
,
Kaufmann
AM
,
Schneider
A
,
Durst
M
, et al
Promoter hypermethylation-mediated inactivation of multiple Slit-Robo pathway genes in cervical cancer progression
.
Mol Cancer
2006
;
5
:
16
.
41.
Li
X
,
Shen
Y
,
Ichikawa
H
,
Antes
T
,
Goldberg
GS
. 
Regulation of miRNA expression by Src and contact normalization: effects on nonanchored cell growth and migration
.
Oncogene
2009
;
28
:
4272
83
.
42.
Roulin
D
,
Cerantola
Y
,
Dormond-Meuwly
A
,
Demartines
N
,
Dormond
O
. 
Targeting mTORC2 inhibits colon cancer cell proliferation in vitro and tumor formation in vivo
.
Mol Cancer
2010
;
9
:
57
.
43.
Guertin
DA
,
Stevens
DM
,
Saitoh
M
,
Kinkel
S
,
Crosby
K
,
Sheen
JH
, et al
mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice
.
Cancer Cell
2009
;
15
:
148
59
.