MicroRNAs (miRNA) are negative regulators of gene expression at the posttranscriptional level, which are involved in tumorigenesis. Two miRNAs, miR-15a and miR-16, which are located at chromosome 13q14, have been implicated in cell cycle control and apoptosis, but little information is available about their role in solid tumors. To address this question, we established a protocol to quantify miRNAs from laser capture microdissected tissues. Here, we show that miR-15a/miR-16 are frequently deleted or down-regulated in squamous cell carcinomas and adenocarcinomas of the lung. In these tumors, expression of miR-15a/miR-16 inversely correlates with the expression of cyclin D1. In non–small cell lung cancer (NSCLC) cell lines, cyclins D1, D2, and E1 are directly regulated by physiologic concentrations of miR-15a/miR-16. Consistent with these results, overexpression of these miRNAs induces cell cycle arrest in G1-G0. Interestingly, H2009 cells lacking Rb are resistant to miR-15a/miR-16–induced cell cycle arrest, whereas reintroduction of functional Rb resensitizes these cells to miRNA activity. In contrast, down-regulation of Rb in A549 cells by RNA interference confers resistance to these miRNAs. Thus, cell cycle arrest induced by these miRNAs depends on the expression of Rb, confirming that G1 cyclins are major targets of miR-15a/miR-16 in NSCLC. Our results indicate that miR-15a/miR-16 are implicated in cell cycle control and likely contribute to the tumorigenesis of NSCLC. [Cancer Res 2009;69(13):5553–9]

Lung cancer is the leading cause of cancer-associated death and is responsible for more deaths than the next three most common tumors combined (breast, prostate, and colon; ref. 1). The high mortality rate of this disease entity is primarily due to the fact that it is only diagnosed at an advanced stage. Lung cancer comprises several histologic types including small cell lung cancer and non–small cell lung cancer (NSCLC); the latter can be further subdivided into two major types, squamous cell carcinoma and adenocarcinoma (2). Squamous cell carcinomas usually arise from the major bronchi, whereas adenocarcinomas arise from distant airway bronchioles and alveoli.

Lung cancer is characterized by multiple genetic changes affecting different oncogenes or tumor suppressor genes involved in cell cycle control, DNA repair, and apoptosis (2). The most common alterations are overexpression of cyclin D1 (CCND1) and Bcl2, mutations of KRAS or members of the ERBB family, and mutations or inactivation of Rb, p16INK4a, and TP53 (2). Altered expression of marker genes in neoplastic tissues may be due to genetic alterations or promoter methylation, but in many cases the mechanism of dysregulation is unknown.

MicroRNAs (miRNA) constitute a novel class of regulatory molecules at the posttranscriptional level, which are involved in tumorigenesis (3). These short RNAs of 19 to 25 nucleotides play key roles in a wide variety of biological processes including cell fate specification, proliferation, cell death, and energy metabolism (4). Several miRNAs have been identified, which are directly involved in tumorigenesis of lung cancer and are considered as prognostic markers for overall survival or predictive markers for chemotherapy (5, 6).

miR-15a and miR-16-1 are located at chromosome 13q14, a region that is deleted in 68% of chronic lymphocytic leukemia (7). Cimmino and colleagues showed that miR-15a/miR-16 expression is inversely correlated to Bcl2 expression and that Bcl2 repression by these miRNAs induces spontaneous apoptosis in a leukemic cell line model (8). In a colon carcinoma cell line, however, the same miRNAs coordinately regulate different mRNA targets, including CDK6, CARD10, CDC27, and C10orf46, which act in concert to control cell cycle progression (9), suggesting that these miRNAs may have cell type-specific functions. The spectrum of potential targets of miR-16 was further expanded by the recent work of Chen and colleagues (10), Liu and colleagues (11), and Bonci and colleagues (12), who showed that G1 cyclins are also regulated by this miRNA.

To investigate if miR-15a/miR-16 are important for the tumorigenesis of lung cancer, we collected tissues from squamous cell carcinomas and adenocarcinomas and corresponding normal tissues by laser capture microdissection and compared these tissues for miRNA expression. We report that miR-15a/miR-16 are deleted or down-regulated in the majority of NSCLC. In addition, we investigated the molecular mechanism underlying miR-15a/miR-16 regulation in different NSCLC cell lines. We show that miR-15a/miR-16 is able to induce cell cycle arrest in G1-G0 in a Rb-dependent manner and that G1 cyclins are physiologic targets of these miRNAs. We propose two alternative pathways by which NSCLC cells escape miR-15a/miR-16–induced cell cycle arrest: (a) down-regulation of miR-15a/miR-16 and (b) inactivation of the Rb gene.

Cell lines and culture conditions. The NSCLC cell lines Calu-1, Calu-6, A549, H2009, H1299, and H358 were obtained from the American Type Culture Collection. All cell lines were cultured in Iscove's modified Dulbecco's medium supplemented with 2 mmol/L l-alanyl-l-glutamine, 1% penicillin/streptomycin, and 5% fetal bovine serum (Sigma) at 37°C and 5% CO2.

Constructs. Luciferase reporter constructs were generated containing a firefly luciferase gene cloned between HindIII and XbaI sites of pcDNA3.0 (Invitrogen). DNA fragments encompassing the 3′-untranslated region (UTR) of CCND1, cyclin D2 (CCND2), and cyclin E1 (CCNE1), respectively, were amplified from genomic DNA and cloned into the XbaI site of the luciferase construct. Luciferase plasmids containing one or three copies of a miR-15a/miR-16 target site from CCND1, CCND2, or CCNE1, respectively, were obtained by cloning double-stranded oligonucleotides into the XbaI site of the luciferase plasmid (Supplementary Table S1; Supplementary Fig. S1). pcDNA3 3xmiR-15a/miR-16 was constructed by cloning three copies of the miR-15a/miR-16 locus in tandem into pcDNA3.0. Rc/CMV cyclinD1 and Rc/CMV cyclin E expression constructs (13) were obtained from Addgene. The miR-15a/miR-16 target sites were deleted from Rc/CMV CCNE1 by PCR amplification, giving rise to Rc/CMV cyclin EΔ3′-UTR. Primers used for amplification are indicated in Supplementary Table S1.

Transfection. Cells were seeded in culture flasks 24 h before transfection. Cotransfections with plasmid DNA were done using Effectene reagent (Qiagen); all other transfections were done using HiPerFect (Qiagen). Transfection was done using 10 to 20 nmol/L of a mixture of equal amounts of hsa pre-miR-15a and hsa pre-miR-16 precursors or pre-miR miRNA precursor control 1 (Ambion), 60 nmol/L Rb-kd small interfering RNA (siGENOME SMARTpool; Dharmacon), 60 nmol/L siCONTROL nontargeting Pool 2 (Dharmacon), 100 nmol/L of a mixture of equal amounts of anti-miR-15a and anti-miR-16 inhibitors or anti-miRNA inhibitor control (Ambion), pCMV-Rb (14) or empty control plasmid, firefly luciferase constructs, and Renilla luciferase reporter plasmid pGl4.74 (Promega) or expression constructs as described above. Transfection efficiency of short RNAs and plasmid DNA was monitored using siGloGreen transfection indicator (Dharmacon) or a RFP expression plasmid, respectively.

Luciferase activity assay, cell cycle analysis, and apoptosis assay. Luciferase activity assays were done 48 h after transfection using a dual-luciferase reporter assay system (Promega) and an Infinite 200 reader (Tecan).

Cell cycle analysis was done essentially as described (9). Cells were analyzed using a LSR II flow cytometer (BD Biosciences) and FlowJo 8.8.4 software (Tree Star).

For apoptosis assays, floating and adherent cells were harvested 24 to 72 h after transfection, combined, and washed with PBS. Annexin V (1:100) either alone or in combination with 10 μg/mL propidium iodide (Sigma) was added to the cells and samples were analyzed within 30 min after staining. Quantification of fluorescence was done by flow cytometry as described above.

RNA isolation and real-time PCR. Total RNA was extracted from cultured cells using the miRVana RNA isolation kit according to the manufacturer's instructions (Ambion). TaqMan miRNA assays (Applied Biosystems) were used to quantify the expression of mature hsa miR-15a/miR-16. Reverse transcription was done using the TaqMan miRNA reverse transcription kit. Reverse transcription of mRNAs was done using TaqMan reverse transcription reagents (Applied Biosystems). Quantitative PCR of CCND2 was done using a TaqMan assay (Applied Biosystems); all other amplifications were done using Quantitect Primer assays (Qiagen). Quantitative PCR was done in a Real-time PCR System 7500 (Applied Biosystems). Gene expression of miRNAs was calculated relative to RNU48 or RNU6B; mRNA levels were normalized to the level obtained for GAPDH, hb2m, HPRT-1, or RPL13A, respectively. Changes in expression were calculated using the ΔΔCt method.

Western blot analysis and immunohistochemistry. Proteins were separated by SDS-PAGE and Western blotting was done using polyvinylidene difluoride membrane (Millipore). Unspecific sites were blocked in 5% nonfat dry milk or 5% bovine serum albumin at room temperature for 1 h. Antibodies directed against CCND1 (clone DCS-6; DAKO; diluted 1:100), CCNE1 (clone 13A3; Neomarkers; 1:200), Rb (clone 3C8; QED Bioscience; 1:1,000), phospho-Rb (Ser807/Ser811; Cell Signaling Technology; 1:1,000), and α-tubulin (clone B512; Sigma; 1:5,000) were used. Secondary antibody was goat anti-mouse horseradish peroxidase or goat anti-rabbit horseradish peroxidase (Bio-Rad) used at 1:5,000 or 1:7,000, respectively.

For immunohistochemistry, 4 μm formalin-fixed, paraffin-embedded sections were treated essentially as described (15). Anti-Bcl2 (clone 124; DAKO), anti-CCND1, anti-CCNE1, and anti-Rb antibodies were used at 1:30, 1:25, 1:25, and 1:100 dilutions, respectively. Mouse IgG1 (DAKO; 1:20) was used as a negative control. Sections were incubated with EnVision+ system (labeled polymer horseradish peroxidase anti-mouse; DAKO) for 30 min at room temperature, developed in 3,3′-diaminobenzidine (Sigma) for 8 min, and counterstained with hematoxylin.

Laser capture microdissection and loss of heterozygosity analysis. Formalin-fixed, paraffin-embedded sections were deparaffinized and stained with methyl green (Merck). Approximately 1,000 tumor cells were captured onto an adhesive cap using a PALM Microbeam (PALM; Zeiss). The dissected material was resuspended in digestion buffer (RecoverAll; Ambion). tRNA (120 ng) was added to the samples and incubated for 10 min at 90°C. All subsequent purification steps were done following the manufacturer's instructions, except that the DNase treatment step was omitted. Loss of heterozygosity (LOH) analysis was done as described (7) using a genetic analyzer (ABI Prism 3100 Avant; Applied Biosystems) and the GeneMapper software 4.0.

All experiments using human specimens were done according to the ethical guidelines of the Institute of Pathology at the University of Bern and were reviewed by the institutional review board.

Statistics. Statistical differences were calculated using unpaired two-tailed Student's t test. A probability of P ≤ 0.05 was considered statistically significant. Statistical significance of inverse correlation was calculated by the N-1 χ2 test (2 × 2 table Pearson's analysis; ref. 16).

miR-15a and miR-16 are frequently down-regulated in lung cancer. It has been shown previously that miR-15a/miR-16 can induce apoptosis or cell cycle arrest depending on the cell line (811). To address their role in NSCLC, we analyzed 11 adenocarcinomas and 12 squamous cell carcinomas of the lung from the archive of the Institute of Pathology at the University of Bern. Quantification of RNAs is challenged by extensive fragmentation and modification of nucleic acids during formalin fixation (17). To address this problem, nucleic acids were subjected to a heat treatment before RNA extraction, which reverses methylol groups introduced during formalin fixation. Under these conditions, fresh and formalin-fixed, paraffin-embedded tissues gave rise to similar miR-16 levels as indicated by quantitative PCR (Supplementary Fig. S2A). The quality of miRNAs may also depend on the postoperative period before fixation. Experimental tissue samples were left at room temperature for up to 5 h before fixation, which, however, did not affect miR-16 quantification (Supplementary Fig. S2A). In addition, detection of miR-16 was linear over a wide concentration range (Supplementary Fig. S2B), indicating that miR-16 can be quantified accurately from formalin-fixed laser capture microdissected material.

Tumor tissues from adenocarcinomas and squamous cell carcinomas were compared with matched normal tissue from alveolar or bronchial epithelium, respectively, which had been microdissected from the same slide. miR-16 was down-regulated in 82% (9 of 11) of adenocarcinomas (P < 0.001) and 67% (8 of 12) of squamous cell carcinomas (in 5 of which down-regulation was statistically significant; P < 0.05; Fig. 1A). Interestingly, miR-16 was significantly up-regulated in 26% of tumors. miR-15a gave rise to a similar expression pattern as miR-16 (data not shown) but was more difficult to detect due to its low abundance. Both miRNAs were also significantly down-regulated in the NSCLC cell lines Calu-1, Calu-6, A549, H2009, H1299, and H358 (Fig. 1B).

Figure 1.

miR-15a and miR-16 are frequently deleted or down-regulated in NSCLC. A, miR-16 levels in adenocarcinomas and squamous cell carcinomas relative to matched normal tissues (n = 3). LOH status for the presented samples: -, heterozygosity; +, LOH. Tumors with no informative microsatellite locus were excluded. B, miR-15a and miR-16 levels in NSCLC cell lines relative to normal lung tissue (n = 3). *, P < 0.05.

Figure 1.

miR-15a and miR-16 are frequently deleted or down-regulated in NSCLC. A, miR-16 levels in adenocarcinomas and squamous cell carcinomas relative to matched normal tissues (n = 3). LOH status for the presented samples: -, heterozygosity; +, LOH. Tumors with no informative microsatellite locus were excluded. B, miR-15a and miR-16 levels in NSCLC cell lines relative to normal lung tissue (n = 3). *, P < 0.05.

Close modal

In the majority of cases, down-regulation of miR-15a/miR-16 was associated with a deletion of one allele of the miR-15a/miR-16 locus. This was based on the finding that two microsatellites, D13S273 and D13S272, flanking the miR-15a/miR-16 locus revealed a LOH in 73% (8 of 11) of adenocarcinomas and 83% (10 of 12) squamous cell carcinomas (Fig. 1A).

miR-16 expression inversely correlates to CCND1 in NSCLC. To investigate if dysregulation of miR-16 may affect the expression of potential targets, CCND1, CCNE1, and Bcl2 were analyzed by immunohistochemistry. Analysis of the same tumor area on serial tissue sections revealed an inverse correlation in the expression of miR-16 and CCND1 in 19 of 23 samples (Table 1; Supplementary Fig. S3), suggesting that CCND1 is a physiologic target. In contrast, no inverse correlation was observed for CCNE1 or Bcl2. Based on these results, we cannot exclude that the latter genes are targets, because additional layers of gene regulation may exist, which contribute to CCNE1 and Bcl2 expression.

Table 1.

Expression of CCND1, CCNE1, and Bcl2 relative to miR-16 in adenocarcinomas and squamous cell carcinomas of the lung

Protein expressionmiR-16 expression*
nP
LowHigh
CCND1     
    − 23 0.02 
    + 16   
CCNE1     
    − 10 23 0.3 
    +   
Bcl2     
    − 12 23 0.4 
    +   
Protein expressionmiR-16 expression*
nP
LowHigh
CCND1     
    − 23 0.02 
    + 16   
CCNE1     
    − 10 23 0.3 
    +   
Bcl2     
    − 12 23 0.4 
    +   
*

Number of tumors with reduced or increased levels of miR-16 relative to normal tissue.

Cellular phenotypes triggered by miR-15a and miR-16 in NSCLC cell lines. To investigate the cellular phenotypes triggered by these miRNAs, NSCLC cell lines were cotransfected with miR-15a/miR-16 precursors and analyzed for apoptosis and cell cycle arrest. None of these cell lines underwent spontaneous apoptosis as indicated by propidium iodide staining (Fig. 2A), Annexin V staining, or morphologic changes (data not shown). However, in 5 of 6 cell lines, miR-15a/miR-16 induced cell cycle arrest in G1-G0 in a significant percentage of the cell population (P < 0.01; Fig. 2B). This phenotype was more pronounced when cells were treated with nocodazole 24 h after transfection with miRNA precursors (P < 0.001). In the case of A549, 33% of the cell population had undergone arrest in G1-G0, whereas only 8% of the control transfected with an unrelated RNA were in this phase of the cell cycle (Fig. 2B and C). Comparable results were obtained for the cell lines Calu-1, Calu-6, H1299, and H358 (Fig. 2B; Supplementary Fig. S4). In marked contrast, only 2% of H2009 cells were in the G1-G0 phase on cotransfection with miR-15a/miR-16 precursors (Fig. 2B and C). It can be excluded that the low percentage of cells in G1-G0 was due to a low transfection efficiency, because ∼90% of H2009 cells were transfected with siGloGreen (fluorescently labeled nonfunctional small interfering RNA). Thus, the cell cycle regulatory activity of miR-15a/miR-16 is cell line-specific.

Figure 2.

Phenotypic analysis of miR-15a/miR-16 overexpression. A, spontaneous apoptosis. A549 cells were stained with propidium iodide 72 h post-transfection. As a positive control, cells were treated with UV (200 mJ). B, G1-G0 arrest. Untreated cells or cells treated with nocodazole for 20 h beginning at 24 h post-transfection were analyzed by flow cytometry. The increase in G1-G0 of cells cotransfected with miR-15a/miR-16 precursors relative to cells transfected with precursor control is presented for each cell line (n = 3-4). C, DNA content distribution of A549 or H2009 cells treated with nocodazole as described above. 2N, cells having diploid DNA content; 4N, cells having tetraploid DNA content.

Figure 2.

Phenotypic analysis of miR-15a/miR-16 overexpression. A, spontaneous apoptosis. A549 cells were stained with propidium iodide 72 h post-transfection. As a positive control, cells were treated with UV (200 mJ). B, G1-G0 arrest. Untreated cells or cells treated with nocodazole for 20 h beginning at 24 h post-transfection were analyzed by flow cytometry. The increase in G1-G0 of cells cotransfected with miR-15a/miR-16 precursors relative to cells transfected with precursor control is presented for each cell line (n = 3-4). C, DNA content distribution of A549 or H2009 cells treated with nocodazole as described above. 2N, cells having diploid DNA content; 4N, cells having tetraploid DNA content.

Close modal

Down-regulation of endogenous G1 cyclin mRNAs by miR-15a and miR-16. A search of the TargetScan database allowed the identification of CCND1, CCND2, CCND3, and CCNE1 as predicted targets of miR-15a/miR-16 (Supplementary Fig. S5). To analyze if miR-15a/miR-16 are able to regulate the steady-state level of mRNA of these potential target genes, cells were cotransfected with miR-15a/miR-16 precursors and analyzed for the expression of G1 cyclin mRNAs by quantitative PCR. CCND2 did not yield any PCR product in most of the cell lines analyzed. The results of the other three G1 cyclins are shown in Fig. 3A. All cell lines transfected with miR-15a/miR-16 precursors gave rise to reduced levels of CCND1, CCND3, and CCNE1 mRNAs relative to the control transfected with an unrelated RNA.

Figure 3.

miR-15a and miR-16 induce down-regulation of G1 cyclin mRNAs and proteins. A, mRNA levels. Cells were cotransfected with miR-15a/miR-16 precursors and harvested 48 h post-transfection. The steady-state level of G1 cyclin mRNAs was normalized to the level obtained for GAPDH. Expression values are presented as mean ± SD (n = 3) relative to the expression values obtained for the control transfected with precursor control. Comparable results were obtained when mRNAs were normalized to hb2M, HPRT-1, or RPL13A mRNAs, respectively (data not shown). B, protein levels. Transfected cells were analyzed by Western blot using antibodies as indicated in the figure. Protein levels were normalized to α-tubulin and presented relative to the levels obtained for the control. C, overexpression of CCND1 and CCNE1 partially restores the effect of miR-15a/miR-16. A549 was cotransfected with miRNA precursors and expression constructs and treated for 20 h with nocodazole beginning 48 h post-transfection. The percentage of cells in G1-G0 was determined by flow cytometry (n = 3).

Figure 3.

miR-15a and miR-16 induce down-regulation of G1 cyclin mRNAs and proteins. A, mRNA levels. Cells were cotransfected with miR-15a/miR-16 precursors and harvested 48 h post-transfection. The steady-state level of G1 cyclin mRNAs was normalized to the level obtained for GAPDH. Expression values are presented as mean ± SD (n = 3) relative to the expression values obtained for the control transfected with precursor control. Comparable results were obtained when mRNAs were normalized to hb2M, HPRT-1, or RPL13A mRNAs, respectively (data not shown). B, protein levels. Transfected cells were analyzed by Western blot using antibodies as indicated in the figure. Protein levels were normalized to α-tubulin and presented relative to the levels obtained for the control. C, overexpression of CCND1 and CCNE1 partially restores the effect of miR-15a/miR-16. A549 was cotransfected with miRNA precursors and expression constructs and treated for 20 h with nocodazole beginning 48 h post-transfection. The percentage of cells in G1-G0 was determined by flow cytometry (n = 3).

Close modal

Cotransfection of A549 and H2009 cells with miR-15a/miR-16 precursors also resulted in 50% to 70% less CCND1 and CCNE1 proteins relative to the control (Fig. 3B). Thus, both cell lines are equally able to down-regulate G1 cyclins in response to miR-15a/miR-16. In agreement with these results, the level of phospho-Rb was significantly reduced (Fig. 3B).

To confirm that cell cycle arrest was due to down-regulation of G1 cyclins, A549 was cotransfected with miR-15a/miR-16–refractory expression constructs and miR-15a/miR-16 precursors. Overexpression of CCNE1 and CCND1 was confirmed by Western blotting (Supplementary Fig. S6). As shown in Fig. 3C, cotransfection with miR-15a/miR-16 precursors in combination with Rc/CMV cyclinD1 (P = 0.003) or Rc/CMV cyclin EΔ3′-UTR (P = 0.04) in each case leads to a significant decrease in G1-G0 arrest compared with cells cotransfected with miR-15a/miR-16 precursors in combination with a control construct. Thus, both CCND1 and CCNE1 can partially rescue miR-15a/miR-16–induced cell cycle arrest.

G1 cyclins are physiologic targets of miR-15a and miR-16 in NSCLC cell lines. To investigate if G1 cyclins are regulated directly by miR-15a/miR-16, a series of luciferase constructs were made containing predicted target sequences from the different G1 cyclins cloned downstream of the luciferase reporter gene (Fig. 4A; Supplementary Fig. S1) and cotransfected into H2009 cells together with miR-15a/miR-16 precursors. CCND1 contains two potential target sites for miR-15a/miR-16 (Supplementary Fig. S5). A construct containing the first target site within the 3′-UTR of CCND1 (Luc 1xTS) resulted in 35% luciferase activity relative to the parental Luc construct (Fig. 4B). The activity obtained with this construct was comparable with a construct containing a part of the 3′-UTR of CCND1 encompassing both target sites (Luc 3′-UTR). A third construct containing three copies of a single target site in tandem (Luc 3xTS) gave rise to 25% activity. To confirm the specificity of the assay, mutations were introduced into the target sequence of miR-15a/miR-16 giving rise to Luc mTS (Fig. 4A; Supplementary Fig. S1). The activity obtained with this construct was almost restored to the activity obtained with the Luc construct. CCND1 and CCNE1 constructs gave rise to comparable luciferase activities (Fig. 4B). In contrast, CCND2 seems to be less responsive to miR-15a/miR-16. Insertion of a single target site (TS-1 or TS-2) or three copies of one target site (TS-2) in tandem into the Luc construct did not significantly reduce luciferase activity (Fig. 4B). However, a region of the 3′-UTR encompassing two target sites conferred efficient down-regulation of luciferase activity. Thus, one target site may not be sufficient for down-regulation of luciferase activity. Again, comparable results were obtained in H2009 and A549 cells (Fig. 4B).

Figure 4.

G1 cyclins are directly regulated by endogenous miR-15a and miR-16. A, luciferase constructs containing miR-15a/miR-16 target sites (TS) from the different G1 cyclins. B, luciferase activity of H2009 and A549 cells cotransfected with miR-15a/miR-16 precursors and luciferase constructs. Luciferase activity is presented relative to the activity obtained with the Luc construct (n = 3). C, down-regulation of G1 cyclins by endogenous miR-15a/miR-16. Cells were transfected with luciferase constructs alone (open columns) or in combination with anti-miR-15a and anti-miR-16 (gray columns) in the absence of precursor miRNAs (n = 3-6).

Figure 4.

G1 cyclins are directly regulated by endogenous miR-15a and miR-16. A, luciferase constructs containing miR-15a/miR-16 target sites (TS) from the different G1 cyclins. B, luciferase activity of H2009 and A549 cells cotransfected with miR-15a/miR-16 precursors and luciferase constructs. Luciferase activity is presented relative to the activity obtained with the Luc construct (n = 3). C, down-regulation of G1 cyclins by endogenous miR-15a/miR-16. Cells were transfected with luciferase constructs alone (open columns) or in combination with anti-miR-15a and anti-miR-16 (gray columns) in the absence of precursor miRNAs (n = 3-6).

Close modal

We next investigated if G1 cyclins are regulated by endogenous miR-15a/miR-16. H2009 cells were transfected with the Luc 3xTS construct containing three target sites from the CCND1 or CCNE1 genes, respectively, or with the Luc 3′-UTR construct containing CCND2-specific sequences. All three constructs gave rise to luciferase activities, which were significantly lower than the activity obtained with the Luc construct (Fig. 4C). Relative activities were 47% (P < 0.00001) for the CCND1 Luc 3xTS construct, 33% (P = 0.003) for the CCNE1 Luc 3xTS construct, and 62% (P = 0.001) for the CCND2 Luc 3′-UTR construct. These results clearly indicate that endogenous levels of miR-15a/miR-16 are sufficient to down-regulate G1 cyclins. In agreement with these results, anti-miRs specific for miR-15a/miR-16 were able to completely restore the activity obtained with the Luc 3xTS constructs from CCND1 and CCNE1 (P < 0.002; Fig. 4C) to the level obtained with the Luc construct.

These results indicate that although miR-15a/miR-16 are significantly reduced (Fig. 1B), they are still able to control G1 cyclins in NSCLC cell lines. To investigate if down-regulation of miR-15a/miR-16 is a mechanism by which NSCLC cells induce overexpression of target genes, H2009 cells were cotransfected with an expression construct containing three copies of the miR-15a/miR-16 locus and CCND1 Luc 3xTS. In these cells, the level of miRNAs was restored almost to the level obtained in normal lung tissue. These cells exhibited two times less luciferase activity relative to the control (Supplementary Fig. S7), showing that a reduction in miR-15a/miR-16 activity as we observed in our lung cancer cell lines leads to overexpression of CCND1.

miR-15a– and miR-16–induced cell cycle arrest depends on Rb expression. Based on our results, we may conclude that miR-15a/miR-16–induced cell cycle arrest is due to down-regulation of G1 cyclins, but this may not apply for H2009, which is resistant. H2009 cells differ from the other NSCLC cell lines by the fact that they lack Rb. Cyclin D in complexes with CDK4 and CDK6 and cyclin E in a complex with CDK2 regulate progression through the G1-S boundary of the cell cycle. These complexes phosphorylate and thereby prevent Rb from binding to E2F, which, on release, drives cells from G1 into S phase (reviewed by ref. 18). Thus, Rb-deficient cells no longer depend on cyclin D (19, 20) and therefore may not respond to miR-15a/miR-16. To test this hypothesis, Rb was expressed in H2009 cells and analyzed for miR-15a/miR-16–induced cell cycle arrest. Cotransfection with miR-15a/miR-16 precursors and an empty control plasmid induced cell cycle arrest in 7.4 ± 1% of the population (Fig. 5A). The percentage of cells in the G1-G0 phase of the cell cycle increased to 23 ± 1% on cotransfection with a Rb expression plasmid and control precursor RNA (P = 0.001), indicating that Rb per se is able to induce cell cycle arrest in a relatively high number of cells. However, Rb in combination with miR-15a/miR-16 precursors induced cell cycle arrest in significantly more cells (30 ± 3%; P = 0.01) than Rb in combination with the control precursor (Fig. 5A). Thus, miR-15a/miR-16 depend on Rb to exert their phenotype. The expression of Rb protein was not affected by coexpression with miR-15a/miR-16, but phospho-Rb was reduced by 50% (Fig. 5B).

Figure 5.

miR-15a/miR-16-induced cell cycle arrest depends on Rb. A and D, H2009 and A549 cells were treated with nocodazole beginning 48 h posttransfection and analyzed by flow cytometry (n = 3). B and C, Western blot analysis of H2009 (B) and A549 cells (C) subjected to the same conditions as in A and D using antibodies directed against Rb or phospho-Rb. Protein levels were normalized to α-tubulin.

Figure 5.

miR-15a/miR-16-induced cell cycle arrest depends on Rb. A and D, H2009 and A549 cells were treated with nocodazole beginning 48 h posttransfection and analyzed by flow cytometry (n = 3). B and C, Western blot analysis of H2009 (B) and A549 cells (C) subjected to the same conditions as in A and D using antibodies directed against Rb or phospho-Rb. Protein levels were normalized to α-tubulin.

Close modal

To verify our results, the complementary experiment was done in A549 cells in which the Rb gene was knocked down by RNA interference. These cells expressed three times less Rb protein than the control (Fig. 5C). As expected, A549 cells transfected with small interfering RNAs against Rb were significantly more resistant to miR-15a/miR-16–induced cell cycle arrest (11 ± 1% in G1-G0) than cells transfected with control small interfering RNA (23 ± 2% in G1-G0; P < 0.001; Fig. 5D). In contrast, Rb small interfering RNA in combination with precursor control RNA had no effect (3 ± 1%). In conclusion, loss of Rb confers resistance to miR-15a/miR-16–induced cell cycle arrest.

Deregulated cell proliferation is a key mechanism for neoplastic progression (21). This study shows that miR-15a/miR-16 are negative regulators of cell cycle progression in NSCLC. We propose two mechanisms by which tumor cells can escape miR-15a/miR-16–induced cell cycle arrest either by down-regulation of miR-15a/miR-16 or by inactivation of Rb.

miR-15a/miR-16 are expressed at reduced levels in ∼70% of adenocarcinomas or squamous cell carcinomas and in 6 NSCLC cell lines; in the majority of cases, this is associated with a LOH of the miRNA locus. However, other mechanisms may contribute to miRNA down-regulation. For example, miRNA processing is compromised, owing to reduced expression of Dicer in a significant number of lung cancers (22). Transcription of the miR-15a/miR-16 locus may also be affected. Chung and colleagues showed that the promoter activity was reduced by PAX5 in B-cell neoplasm (23), and it is likely that a similar mechanism may exist in lung cancer. In addition, a point mutation in the miR-16 primary transcript has been linked to diminished expression of the mature miRNA (24). Our results suggest that down-regulation of miR-15a/miR-16 may contribute to tumor growth because this is directly coupled to increased levels of G1 cyclins.

That miR-15a/miR-16–induced cell cycle arrest depends on Rb was evidenced by our findings that H2009 cells lacking Rb are completely resistant to miR-15a/miR-16, whereas introduction of a functional copy of Rb into these cells renders them more sensitive. Consistent with these results, down-regulation of Rb in A549 by RNA interference confers resistance to these miRNAs. This mechanism may apply for up to 14% of squamous cell carcinomas and 33% of adenocarcinomas, which are Rb-negative (25). We found that Rb was down-regulated in 4 of 11 adenocarcinomas (tumors 5-7 and 9) and 3 of 12 squamous cell carcinomas (tumors 1, 4, and 12; see Fig. 1). It is unlikely that two Rb-related proteins, p107 and p130, can compensate for a loss of Rb, because their functions are distinct from that of Rb (26). In addition, these proteins are normally expressed in H2009 cells, which are refractory to miR-15a/miR-16 (27). In conclusion, 87% (20 of 23) of NSCLC had either down-regulated miR-15a/miR-16 or inactivated Rb.

Hemizygous or homozygous loss of 13q14 has been associated with numerous malignancies including chronic lymphocytic leukemia (7), mantle cell lymphoma (28), multiple myeloma (29), breast cancer (30), and high-grade carcinoma of the prostate (31), suggesting that these deletions are of pathogenetic significance. miR-15a/miR-16 are presumably the long-sought candidate tumor suppressors of this region and are proven to be important regulators of Bcl2 expression in chronic lymphocytic leukemia (7, 8). miR-15a/miR-16 are also frequently down-regulated in chronic lymphocytic leukemia and pituitary adenomas (32). Interestingly, both miRNAs are frequently up-regulated in cervical cancer (33). Because these cells normally express an inactive form of Rb, we may conclude that cell cycle progression of cervical cancer cells no longer depends on miR-15a/miR-16 activity. Because the level of miR-15a/miR-16 is normally high in these tumors, this may suggest a dual role of miR-15a/miR-16 as tumor-suppressing and oncogenic miRNAs. A similar mechanism may also exist in NSCLC, because both miRNAs were up-regulated in 28% (5 of 18) of the tumor samples, in 3 of which Rb was down-regulated.

What are the targets that are responsible for miR-15a/miR-16–induced cell cycle arrest in NSCLC? miRNAs including miR-15a/miR-16 can affect hundreds of mRNAs (9), which renders it difficult to identify the biologically relevant targets. Although it is believed that miR-15a/miR-16-induced cell cycle arrest is mediated by targeting G1 cyclins (10, 11), it cannot be excluded that this is induced by an indirect mechanism, for example, by targeting mRNAs from essential genes. Based on our findings, however, that miR-15a/miR-16-induced cell cycle arrest depends on Rb, we can conclude that this is due to down-regulation of proteins directly involved in cell cycle control.

It was shown recently that CCND1, CCND3, CCNE1, CDK4, and CDK6 are direct targets of miR-15a/miR-16 (912). We confirm that CCND1, CCND3, and CCNE1 are also targets in NSCLC cell lines and provide evidence that CCND2 is an additional target of these miRNAs. For most of these targets, it was unclear if they are also regulated by physiologic concentrations of miR-15a/miR-16. We show that CCND1, CCND2, and CCNE1 are directly regulated by miR-15a/miR-16 under physiologic conditions. Using luciferase reporter constructs containing target sequences from the different cyclin genes, we showed that endogenous miRNAs are able to down-regulate luciferase activity and that this effect is reversed by cotransfection with anti-miRs against miR-15a/miR-16. miR-16 inversely correlates with CCND1 protein in NSCLC, which is in line with the finding that this is a physiologic target. In addition, miR-15a/miR-16–induced cell cycle arrest can be partially restored by overexpression of CCND1 or CCNE1.

In conclusion, we show that miR-15a/miR-16 induce cell cycle arrest by down-regulation of G1 cyclins, whereas NSCLC cells escape these growth-inhibitory signals either by down-regulation or loss of function of Rb, by down-regulation of miR-15a/miR-16, or by other means. This may constitute a general mechanism implicated in tumorigenesis, because LOH 13q14 and Rb inactivation are frequent events in various tumors. Furthermore, our data suggest that miR-15a/miR-16 might be used as therapeutic agents in Rb-proficient NSCLC.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

Current address for L. Hasan: BÜHLMANN Laboratories, CH-4124 Schönenbuch, Switzerland.

Grant support: Swiss National Science Foundation (E. Vassella).

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.

We thank Mario Tschan for plasmids and cell lines, Bernadette Wyder for introduction into flow cytometry, and Sabine Jakob and Silvia Rihs for protocols and helpful discussions.

1
Jemal A, Siegel R, Ward E, Murray T, Xu J, Thun MJ. Cancer statistics, 2007.
CA Cancer J Clin
2007
;
57
:
43
–66.
2
Wistuba II, Gazdar AF. Lung cancer preneoplasia.
Annu Rev Pathol
2006
;
1
:
331
–48.
3
Mattie MD. MicroRNAs in cancer (Oncomirs). In: Clarke NJ, Sanseau P, editors. MicroRNAs: biology, function and expression. Eagleville: DNA Press; 2007. p. 251–80.
4
Hwang HW, Mendell JT. MicroRNAs in cell proliferation, cell death, and tumorigenesis.
Br J Cancer
2006
;
94
:
776
–80.
5
Johnson SM, Grosshans H, Shingara J, et al. RAS is regulated by the let-7 microRNA family.
Cell
2005
;
120
:
635
–47.
6
Takamizawa J, Konishi H, Yanagisawa K, et al. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival.
Cancer Res
2004
;
64
:
3753
–6.
7
Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia.
Proc Natl Acad Sci U S A
2002
;
99
:
15524
–9.
8
Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2.
Proc Natl Acad Sci U S A
2005
;
102
:
13944
–9.
9
Linsley PS, Schelter J, Burchard J, et al. Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression.
Mol Cell Biol
2007
;
27
:
2240
–52.
10
Chen RW, Bemis LT, Amato CM, et al. Truncation in CCND1 mRNA alters miR-16-1 regulation in mantle cell lymphoma.
Blood
2008
;
112
:
822
–9.
11
Liu Q, Fu H, Sun F, et al. miR-16 family induces cell cycle arrest by regulating multiple cell cycle genes.
Nucleic Acids Res
2008
;
36
:
5391
–404.
12
Bonci D, Coppola V, Musumeci M, et al. The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities.
Nat Med
2008
;
14
:
1271
–7.
13
Hinds P, Mittnacht S, Dulic V, Arnold A, Reed S, Weinberg R. Regulation of retinoblastoma protein functions by ectopic expression of human cyclins.
Cell
1992
;
70
:
993
–1007.
14
Tan X, Martin SJ, Green DR, Wang JYJ. Degradation of retinoblastoma protein in tumor necrosis factor- and CD95-induced cell death.
J Biol Chem
1997
;
272
:
9613
–6.
15
Gugger M, Kappleler A, Vonlanthen S, et al. Alterations of cell cycle regulators are less frequent in advanced non-small cell lung cancer than in resectable tumours.
Lung Cancer
2001
;
33
:
229
–39.
16
Campbell I. Chi-squared and Fisher-Irwin tests of two-by-two tables with small sample recommendations.
Stat Med
2007
;
26
:
3661
–75.
17
Masuda N, Ohnishi T, Kawamoto S, Monden M, Okubo K. Analysis of chemical modification of RNA from formalin-fixed samples and optimization of molecular biology applications for such samples.
Nucleic Acids Res
1999
;
27
:
4436
–43.
18
Chivukula RR, Mendell JT. Circular reasoning: microRNAs and cell-cycle control.
Trends Biochem Sci
2008
;
33
:
474
–81.
19
Lukas J, Bartkova J, Rohde M, Strauss M, Bartek J. Cyclin D1 is dispensable for G1 control in retinoblastoma gene-deficient cells independently of cdk4 activity.
Mol Cell Biol
1995
;
15
:
2600
–11.
20
McGahren-Murray M, Nicholas H, Keyomarsi K. The differential suaurosporine-mediated G1 arrest in normal versus tumor cells is dependent on the retinoblastoma protein.
Cancer Res
2006
;
66
:
9744
–53.
21
Evan GI, Vousden KH. Proliferation, cell cycle and apoptosis in cancer.
Nature
2001
;
411
:
342
–8.
22
Karube Y, Tanaka H, Osada H, et al. Reduced expression of Dicer associated with poor prognosis in lung cancer patients.
Cancer Sci
2005
;
96
:
111
–5.
23
Chung EY, Dews M, Cozma D, et al. c-Myb oncoprotein is an essential target of the dleu2 tumor suppressor microRNA cluster.
Cancer Biol Ther
2008
;
7
:
1758
–64.
24
Calin GA, Ferracin M, Cimmino A, et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia.
N Engl J Med
2005
;
353
:
1793
–801.
25
Leversha MA, Fielding P, Watson S, Gosney JR, Field JK. Expression of p53, pRB, and p16 in lung tumours: a validation study on tissue microarrays.
J Pathol
2003
;
200
:
610
–9.
26
Bruce J, Hurford RJ, Classon M, Koh J, Dyson N. Requirements for cell cycle arrest by p16INK4a.
Mol Cell
2000
;
6
:
737
–42.
27
Modi S, Kubo A, Oie H, Coxon AB, Rehmatulla A, Kaye FJ. Protein expression of the RB-related gene family and SV40 large T antigen in mesothelioma and lung cancer.
Oncogene
2000
;
19
:
4632
–9.
28
Stilgenbauer S, Nickolenko J, Wilhelm J, et al. Expressed sequences as candidates for a novel tumor suppressor gene at band 13q14 in B-cell chronic lymphocytic leukemia and mantle cell lymphoma.
Oncogene
1998
;
16
:
1891
–7.
29
Elnenaei MO, Hamoudi RA, Swansbury J, et al. Delineation of the minimal region of loss at 13q14 in multiple myeloma.
Genes Chromosome Cancer
2003
;
36
:
99
–106.
30
Bieche I, Lidereau R. Loss of heterozygosity at 13q14 correlates with RB1 gene underexpression in human breast cancer.
Mol Carcinog
2000
;
29
:
151
–8.
31
Dong JT, Boyd JC, Frierson HF, Jr. Loss of heterozygosity at 13q14 and 13q21 in high grade, high stage prostate cancer.
Prostate
2001
;
49
:
166
–71.
32
Bottoni A, Piccin D, Tagliati F, Luchin A, Zatelli MC, degli Uberti EC. miR-15a and miR-16-1 down-regulation in pituitary adenomas.
J Cell Physiol
2005
;
204
:
280
–5.
33
Wang X, Tang S, Le SY, et al. Aberrant expression of oncogenic and tumor-suppressive microRNAs in cervical cancer is required for cancer cell growth.
PLoS ONE
2008
;
3
:
e2557
.