Open Access

miR‑496, miR‑1185, miR‑654, miR‑3183 and miR‑495 are downregulated in colorectal cancer cells and have putative roles in the mTOR pathway

  • Authors:
    • Naif Alqurashi
    • Saeed M. Hashimi
    • Faisal Alowaidi
    • Saso Ivanovski
    • Amro Farag
    • Ming Q. Wei
  • View Affiliations

  • Published online on: June 21, 2019     https://doi.org/10.3892/ol.2019.10508
  • Pages: 1657-1668
  • Copyright: © Alqurashi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs) are small non‑coding RNAs that regulate gene expression by suppressing the target mRNA and inhibiting translation in order to regulate multiple biological processes. miRNAs play important roles as oncogenes or tumor suppressors in the development of various types of human cancer. The regulation of mammalian target of rapamycin (mTOR) by miRNAs has been studied in several types of cancer, including colorectal cancer (CRC). However, to the best of our knowledge, only limited information regarding the function of miRNAs in human CRC is available. In the present study, the expression of 22 miRNAs in CRC cell lines were investigated in regard to key genes in the mTOR pathway. Initially, it was revealed that mTOR, regulatory‑associated protein of mTOR complex I and rapamycin‑intensive companion of mTOR were overexpressed in CRC cell lines when compared with a normal colorectal cell line. Subsequently, putative miRNA‑mRNA associations were identified via multiple miRNA target prediction programs. The expression levels for the candidate miRNAs were validated using quantitative real‑time polymerase chain reaction. Expression analysis revealed that, among 20 miRNAs, five miRNAs (miR‑496, miR‑1185, miR‑654, miR‑3183 and miR‑495) exhibited significant downregulation in association with the mTOR signaling pathway. Taken together, the results from the present study suggest that several miRNAs that are associated with CRC, with possible roles in mTOR signaling, may have potential therapeutic or diagnostic benefits in CRC treatment.

Introduction

Colorectal cancer (CRC) accounted for ~1.4 million of the new cancer cases diagnosed in 2012 worldwide and is considered the third most common cancer (1). According to the International Agency for Research on Cancer, by 2035, the estimated number of CRC cases will reach 2.4 million cases diagnosed each year (1). Although a lot has been learned about CRC, novel therapeutic strategies are needed in order to tackle this cancer. Mammalian target of rapamycin (mTOR) is a catalytic subunit of two large signaling complexes; mTORC1 and mTORC2, along with key proteins, form these complexes. Regulatory-associated protein of mTOR (RPTOR) is the unique component of mTORC1, whereas rapamycin-insensitive companion of mTOR (RICTOR) is an exclusive component of mTORC2. The two complexes play a central role in tumorigenesis via phosphorylation of key proteins within the mTOR pathway. mTORC1 regulates mRNA translation and elongation by phosphorylating its downstream effectors, such as eukaryotic initiation factor 4E-binding protein 1 and the p70 ribosomal S6 kinase 1 (2). mTORC2 phosphorylates protein kinase B, promoting cell proliferation, apoptosis and survival (3).

MicroRNAs (miRNAs/miRs) are single-stranded, non-coding RNAs, ranging from 19–24 nucleotides in length. In order to perform their regulatory functions, mature miRNAs bind most often to the 3′untranslated region (UTR) of messenger RNAs, which inhibit the translation of mRNA and result in the downregulation of gene expression at the post-transcriptional level (4). Approximately 50% of human miRNA genes are located in genomic regions that have fragile sites, and these locations contain chromosomal abnormalities, such as deletions and amplifications. These genomic regions are vulnerable to genetic alteration in several types of human cancer (5,6). Furthermore, a single miRNA can interact and regulate more than one mRNA target (7).

In the last decade, a growing body of evidence has revealed that miRNAs are involved in tumorigenesis and cancer progression (8,9). Deregulation of miRNAs has been demonstrated in several types of cancer, including CRC (1016). In cancer cells, alteration of miRNA expression levels can be abnormally down- or upregulated, to function as either tumor suppressors or oncogenes (17). In multiple studies, miRNAs have been demonstrated to play an important role in CRC. For example, miR-21, miR-31 and miR-223 are upregulated in CRC, whereas miR-143, miR-145 and miR-126 are downregulated (1820).

The potential use of miRNA as diagnostic or prognostic markers in the clinical setting has been highlighted previously (21). Furthermore, miRNA mimics can be used as therapeutic agents to restore miRNA function, or miRNA inhibitors can be used to disrupt upregulated miRNA. Therefore, identifying novel cancer-associated miRNAs is important for the potential treatment of cancer.

Downregulated miRNAs are believed to act as tumor suppressors, which may greatly contribute to colorectal carcinogenesis (19,20,22,23). The aim of the present study was to identify miRNA-mRNA associations and evaluate miRNA expression in human CRC cells, by focusing on the mTOR signaling pathway.

Materials and methods

Cell lines

The human CRC cell lines, HT29, HCT116, SW620, SW480 and the FHC normal fetal human colon epithelial cell line were purchased from the American Type Culture Collection (ATCC). The CSC480 cell line was purchased from BioMedicure (San Diego, CA, USA) All cell lines were maintained in Dulbecco's modified Eagle's medium (DMEM; 4.5 g/l D-Glucose, L-Glutamine, 110 mg/l sodium pyruvate; Invitrogen; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (Invitrogen; Thermo Fisher Scientific, Inc.) and cultured at 37°C in a humidified incubator with 5% CO2. Cells were induced with insulin (200 nM) as previously described (24). The FHC cell line was cultured according to previously described culture conditions (25).

Bioinformatics analysis

mRNA targets were predicted for 22 miRNAs of interest using five miRNA target-prediction programs: DIANA-MICROT (diana.imis.athena-innovation.gr) (26), miRWalk (umm.uni-heidelberg.de) (27), TargetScan (targetscan.org) (7), PicTar (pictar.mdc-berlin.de) (28) and miRDB (mirdb.org) (29). These databases use a computational algorithm of miRNAs by searching for the presence of conserved sites on the 3′UTR of mRNA that match the seed region of each miRNA. The top target candidates according to the score in the database for each miRNA provided by each program were chosen for further molecular analysis.

RNA isolation and reverse transcription polymerase chain reaction (RT-PCR)

Total RNA, including miRNA, was extracted from 1×106 cultured cells using an miRNeasy Mini kit (Qiagen, Inc.), according to the manufacturer's protocol. Briefly, cells were trypsinized, collected from a 10-cm cell-culture dish and placed into a centrifuge tube, followed by the addition of 700 µl TRIzol® reagent (Qiagen, Inc., Valencia, CA, USA) then 140 µl chloroform. The aqueous phase was separated by centrifugation (5 min at room temperature) in a microfuge (Eppendorf 5430; Eppendorf, Hamburg, Germany) followed by washing sequentially with 100% ethanol. Samples were centrifuged in an RNeasy Mini spin column membrane (Qiagen, Inc.) and RNA was then eluted in RNase-free water. The concentration of RNA was determined via absorbance measurements at 260 nm using a NanoDrop spectrophotometer, and the RNA was checked by determining absorbance ratios, 260/280 and 260/230 nm, for any contamination. For mRNA expression, cDNAs were synthesized using a BluePrint 1st Strand cDNA Synthesis kit (Takara Bio, Inc.), according to the manufacturer's protocol. miRNAs were reverse-transcribed using an miScript II RT kit (Qiagen, Inc.), also according to the manufacturer's protocol. Briefly, single-stranded cDNA was synthesized from RNA in a 20 µl reaction volume. The reactions were incubated: first, at 37°C for 60 min, then the reverse transcriptase mix was inactivated by incubation at 95°C for 5 min. The cDNA generated was diluted 10-fold in RNase-free water.

RT-quantitative (q)PCR analysis of mRNA

Quantitative, RT-qPCR was used to measure mRNA expression levels for all three genes (mTOR, RPTOR and RICTOR). qPCR was performed using a LightCycler 480 thermal cycling system (Roche Diagnostics) with SYBR Premix Ex Taq II (Takara Bio, Inc.) in a 20 µl reaction mixture. The sequences of primers used for qPCR analysis are listed in Table I. Thermal cycling conditions were stage 1: Initial denaturation (1 cycle) 95°C for 5 min; stage 2: PCR (40 cycles) 95°C for 10 sec followed by 56°C for 10 sec and 72°C for 20 sec; and stage 3: Melting curve analysis 95°C for 5 sec, 65°C for 60 sec and 97°C. A non-template control (nuclease-free water) was included for each primer set and each sample was analyzed in triplicate. Relative expression was calculated using the 2−ΔΔCq method (30).

Table I.

mTOR, RPTOR and RICTOR primer sequences.

Table I.

mTOR, RPTOR and RICTOR primer sequences.

PrimerSequenceAmplicon size
mTOR 146 (bp)
  F 5′-AGCATCGGATGCTTAGGAGTGG-3′
  R 5′-CAGCCAGTCATCTTTGGAGACC-3′
RPTOR 148 (bp)
  F 5′-GATCGTCAACAGCTATCACACGG-3′
  R 5′-CGAGTCGAAGTTCTGCCAGATC-3′
RICTOR 125 (bp)
  F 5′-GCCAAACAGCTCACGGTTGTAG-3′
  R 5′-CCAGATGAAGCATTGAGCCACTG-3′
GAPDH 131 (bp)
  F GTCTCCTCTGACTTCAACAGCG
  R ACCACCCTGTTGCTGTAGCCAA

[i] mTOR, mammalian target of rapamycin; RPTOR, regulatory-associated protein of mTOR complex I; RICTOR, rapamycin-insensitive companion of mTOR; F, forward; R, reverse.

RT-qPCR analysis of miRNAs

RT-qPCR was performed on selected miRNAs using the LightCycler 480 thermal cycling system (Roche Diagnostics). Expression levels of mature miRNAs were quantified using an miScript SYBR Green PCR kit (Qiagen), according to the manufacturer's instructions. The small nucleolar RNA, C/D box 68 (SNORD68), and the U6 small nuclear 2 RNA (RNU6-2) (Qiagen, Inc.) were used as endogenous controls, due to their relatively stable expression. Forward primers (Table II) were designed by first converting the miRNA sequences to DNA and adjusting the melting temperature of the primer to 60°C, either by adding a thymine to the 3′ of the primer or removing a nucleotide from the 5′ end of the primer. Primers were purchased from Integrated DNA Technologies, Inc., while the universal reverse primer was included in the miScript SYBR Green kit. The amplification reaction (10 µl) included 2X SYBR Green PCR master mix (5 µl), universal primer (250 nM), miRNA-specific primer (250 nM), RNase-free water (1.75 µl) and cDNA (2 µl). Pipetting was performed using the epMotion 5075 automated pipetting system (Eppendorf). The reaction conditions were as follows: Initial activation step (1 cycle), 95°C for 15 min followed by 3-step cycling (40 cycles), (denaturation) 94°C for 15 sec, (annealing) 55°C for 30 sec and (extension) 70°C for 30 sec, with melting curve: 95°C for 5 sec, 65°C for 60 sec and 97°C. All reactions were performed in triplicate and the average values were used in subsequent analysis. Expression was normalized to SNORD68 and RNU6-2. The fold change was calculated using the 2−ΔΔCq method (30) to acquire relative expression levels.

Table II.

Designed miRNA forward sequences.

Table II.

Designed miRNA forward sequences.

miRNAForward sequence primer
hsa-miR-1271-5p 5′-TTGGCACCTAGCAAGCACTC-3′
hsa-miR-496 5′-GCTGAGTATTACATGGCCAATCTC-3′
hsa-miR-581 5′-GCGTCTTGTGTTCTCTAGATCAGTAAA-3′
hsa-miR-1185-3p 5′-GCATATACAGGGGGAGACTCTTATAAA-3′
hsa-miR-767-3pa 5′-CTGCTCATACCCCATGGTTTC-3′
hsa-miR-96-5p 5′-TTGGCACTAGCACATTTTTGC-3′
hsa-miR-335-3p 5′-GTTTTTCATTATTGCTCCTGACCA-3′
hsa-miR-3182 5′-GCCGCTTCTGTAGTGTAGTCAAA-3′
hsa-miR-1294 5′-TGTGAGGTTGGCATTGTTGTCT-3′
hsa-miR-2114-3p 5′-GAGCCTCAAGCAAGGGACTT-3′
hsa-miR-3121-3p 5′-GCTAAATAGAGTAGGCAAAGGACAAA-3′
hsa-miR-3183 5′-CTCTCTCGGAGTCGCTCG-3′
hsa-miR-340-3p 5′-GCTCCGTCTCAGTTACTTTATAGCAA-3′
hsa-miR-4802-3p 5′-TACATGGATGGAAACCTTCAAGC-3′
hsa-miR-548o-3p 5′-CCAAAACTGCAGTTACTTTTGCA-3′
hsa-miR-654-5p 5′-GTGGGCCGCAGAACATG-3′
hsa-miR-1323 5′-TCAAAACTGAGGGGCATTTTC-3′
hsa-miR-142-3p 5′-GCTGTAGTGTTTCCTACTTTATGGAAAA-3′
hsa-miR-194-5p 5′-TGTAACAGCAACTCCATGTGGA-3′
hsa-miR-495 5′-GAAACAAACATGGTGCACTTCTTAA-3′
hsa-miR-659-3p 5′-TTGGTTCAGGGAGGGTCC-3′
hsa-miR-98-5P 5′-GCCTGAGGTAGTAAGTTGTATTGTTAAAA-3′

[i] miRNA, microRNA; miR, microRNA.

Statistical analysis

Data analysis was conducted using Prism software (version 6.0; GraphPad Software, Inc.). One-way analysis of variance (ANOVA) was used to determine statistical significance, with the Dunnett's multiple comparisons test used as a post hoc test. P<0.05 was considered to indicate a statistically significant difference. In the ANOVA, each CRC line was compared to the control FHC cell line, and the multiple group graphs represent significance in regard to FHC. Data are presented as the mean of three replicates with standard error.

Results

miRNA target prediction analysis of mTOR pathway genes

In order to determine potential target genes and signaling pathways implicated in mTOR, RPTOR and RICTOR, five programs were used for miRNA target prediction analysis, namely DIANA-MICROT, miRWalk, TargetScan, PicTar and miRDB. Using the combination of five programs provided a more reliable model of target prediction, due to a different computer-aided algorithm for each target prediction program. From the miRNAs predicted for each gene, the highest miRNAs predicted by at least 2 prediction programs were chosen. A total of 22 miRNAs for all three genes were selected for further analysis (Tables IIIV).

Table III.

miRNA candidates targeting the mTOR gene predicted by bioinformatics analysis.

Table III.

miRNA candidates targeting the mTOR gene predicted by bioinformatics analysis.

Database algorithm

miRNA IDProposed target geneEnsembl gene IDmiRNA sequence DianamiRWalkTargetScanPicTarmiRDB
hsa-miR-581mTOR ENSG00000198793 UCUUGUGUUCUCUAGAUCAGUScore0.953.4263
P-value0.040.014Rank 20
Binding8mer8mer7mer
RegionUTR3UTR3UTR3UTR3
hsa-miR-96-5pmTOR ENSG00000198793 UUUGGCACUAGCACAUUUUUGCUScore0.859560
P-value0.040.79Rank 24
Binding7mer7mer-m87mer
RegionUTR3UTR3UTR3
hsa-miR-767-3pamTOR ENSG00000198793 UCUGCUCAUACCCCAUGGUUUCUScore0.9988
P-value0.0040.001Rank 4
Binding6mer10mer7mer
RegionUTR3UTR3UTR3
hsa-miR-1185-3pmTOR ENSG00000198793 AUAUACAGGGGGAGACUCUUAUScore0.8865
P-value0.04Rank 15
Binding8mer7mer
RegionUTR3UTR3
hsa-miR-1271-5pmTOR ENSG00000198793 CUUGGCACCUAGCAAGCACUCAScore0.849561
P-value0.030.79Rank 22
Binding7mer7mer7mer
RegionUTR3UTR3UTR3
hsa-miR-496mTOR ENSG00000198793 UGAGUAUUACAUGGCCAAUCUCScore0.8672
P-value0.030.014Rank 9
Binding8mer8mer7mer
RegionUTR3UTR3UTR3
hsa-miR-335-3pmTOR ENSG00000198793 UUUUUCAUUAUUGCUCCUGACCScore0.8567
P-value0.030.014Rank 13
Binding8mer8mer7mer
RegionUTR3UTR3UTR3
hsa-miR-3182mTOR ENSG00000198793 GCUUCUGUAGUGUAGUCScore0.8579
P-value0.028Rank 6
Binding7mer7mer
RegionUTR3UTR3

[i] miRNA, microRNA; miR, microRNA; UTR3, 3 untranslated region; mTOR, mammalian target of rapamycin.

Table V.

miRNA candidates targeting the RICTOR gene predicted by bioinformatics analysis.

Table V.

miRNA candidates targeting the RICTOR gene predicted by bioinformatics analysis.

Database algorithm
miRNA IDProposed target geneEnsembl gene IDmiRNA sequence DianamiRWalkTargetScanPicTarmiRDB
hsa-miR-495RICTOR ENSG00000164327 AAACAAACAUGGUGCACUUCUUScore0.871−0.17
P-value0.0020.0166N/A
Binding8mer9mer8mer
RegionUTR3UTR3UTR3
hsa-miR-194-5pRICTOR ENSG00000164327 UGUAACAGCAACUCCAUGUGGAScore0.7983
P-value0.003Rank 28
Binding6mer7mer
RegionUTR3UTR3
hsa-miR-659-3pRICTOR ENSG00000164327 CUUGUUCAGGGAGGGUCCCCAScore0.98675
P-value0.008Rank 48
Binding7mer7mer
RegionUTR3UTR3
hsa-miR-142-3pRICTOR ENSG00000164327 UGUAGUGUUUCCUACUUUAUGGAScore0.999100
P-value0.030.01660.99Rank 61
Binding8mer9mer8mer7mer
RegionUTR3UTR3UTR3UTR3
hsa-miR-98-5PRICTOR ENSG00000164327 UGAGGUAGUAAGUUGUAUUScore0.999−0.14
P-value0.0070.01660.98Rank
Binding8mer9mer8mer7mer
RegionUTR3UTR3UTR3UTR3
hsa-miR-1323RICTOR ENSG00000164327 UCAAAACUGAGGGGCAUUUUCUScore0.7580
P-value0.0060.004Rank 37
Binding8mer10mer7mer
RegionUTR3UTR3UTR3

[i] miRNA, microRNA; miR, microRNA; UTR3, 3 untranslated region; RICTOR; rapamycin-insensitive companion of mammalian target of rapamycin.

Gene expression of mTOR, RICTOR, RPTOR and validation of specific miRNAs

To assess alterations of the mTOR pathway in CRC cells, mTOR, RPTOR and RICTOR expression levels were investigated in five human CRC cell lines, HT29, HCT116, SW620, SW480 and CSC480, using RT-qPCR. The gene GAPDH was used as a reference. The comparison of results from mTOR, RPTOR and RICTOR expression revealed significantly (P<0.01) elevated mRNA expression levels in all cell lines compared with normal FHC cells (Fig. 1).

mTOR pathway-targets miRNAs differentially regulated

Next, the most significant candidates of miRNAs were selected, as predicted by the bioinformatics analysis, to target the genes of interest for further confirmatory studies. A selection of CRC cell lines, as well as the control (FHC), were used to examine whether miRNAs were associated with transcriptional expression of their relevant target gene and to identify miRNAs involved in CRC cells. Based on the results from the bioinformatics analysis, candidate miRNAs that were identified to be associated with each gene were assessed using qPCR.

mTOR

A statistically significant upregulation in the expression levels of miR-96 (P<0.01) and miR-335 (P<0.001), which was associated with mTOR, was observed in all cell lines (Fig. 2). miRNAs, such as miR-676a (HT29 and SW480, P<0.001) and miR-3182 (HCT116 P<0.05), were upregulated in certain cell lines, while miR-1272 and miR-581 expression remained unchanged. miR-496 and miR-1185 expression levels were significantly downregulated (P<0.001) in all cancer cell lines (Fig. 2).

RPTOR

In miRNAs associated with RPTOR, miR-340 exhibited high expression levels in all cell lines (P<0.01), whereas miR-4802 was upregulated (P<0.001) in HT29 cells only (Fig. 3). Furthermore, miR-548 was upregulated in all cell lines (P<0.05) except HCT116. HT29 and CSC480 were the only cell lines that exhibited upregulated miR-2114 (P<0.001). There was no statistically significant difference in the expression levels of miR-3121, except for the downregulation in HCT116 cells (P<0.05). However, miR-654 (P<0.01) and miR-3183 (P<0.001) expression levels were downregulated in all cell lines (Fig. 3).

RICTOR

In miRNAs associated with RICTOR, miR-142, miR-194, miR-98 and miR-659 demonstrated high expression levels in all cells (P<0.01), while miR-1323 was upregulated (P<0.001) in CSC480 cells only (Fig. 4). The expression levels of miR-495 were downregulated in all cell lines (P<0.001).

Discussion

Deregulation of the mTOR pathway occurs frequently in human cancer types (3,24,31,32). Regulation of mTOR by miRNAs has been investigated in a number of different types of cancer, including CRC (33,34). In the present study, it was revealed that 6 miRNAs (miR-96, miR-335, miR-340, miR-142, miR-194, miR-659 and miR-98) were upregulated in the 5 investigated human CRC cell lines when comparing with FHC. Furthermore, among 20 miRNAs, 5 (miR-496, miR-1185, miR-3183, miR-654 and miR-495) revealed significant downregulation in association with the 3 key mTOR signaling pathway genes, mTOR, RPTOR and RICTOR.

Several software programs are available to aid in identifying miRNA target prediction. The majority of these approaches use the seed region, which is approximately 6–8 nucleotides in length within the miRNA, as methods to target mRNA and bind at the 3′UTR of the target gene when searching for complementary strands (35). The present study used five algorithmic tools, Diana-MicroT, miRWalk, TargetScan, PicTar and miRDB, for miRNA target prediction analysis, which also considered seed-based methods, and miRDB analyses identified those genes affected by miRNAs, using a support vector machine trained on multiple microarray datasets (29). Despite the recent advances in miRNA target prediction databases (2629), the results of miRNA-mRNA association analyses were varied depending on the database. A possible explanation is that each program has its methodological variances with respect to detecting miRNA-binding regions. To minimize the differences in miRNA target predictions, miRNAs that were identified by at least three prediction programs were selected for analysis.

Consistent with the results from the previous study, miR-335 has been demonstrated to be overexpressed in gastric cancer and involved in the oncogenic mTOR signaling pathway (36). In addition, miR-335 has been revealed as upregulated in a number of different types of cancer, including CRC (3739). In numerous studies, the miRNAs miR-96 (4045), miR-340 (46), miR-142 (4749), miR-194 (5052) and miR-98 (5355), have demonstrated overexpression in a number of different types of cancer, including CRC.

Numerous studies have demonstrated downregulation of miRNAs in different types of cancer and their capacity to play a role in the mTOR signaling pathway (5659). In the present study, the expression analysis was focused on the most highly downregulated miRNAs that were associated with upregulated mTOR, RPTOR and RICTOR gene expression in CRC cell lines. Among all five downregulated miRNAs investigated in the present study, miR-495, miR-1185, miR-654 and miR-496 have previously been reported in various types of cancer (5659).

When regarding the role of miR-495 in cancer, several studies have revealed decreased miR-495 expression levels in a number of different types of cancer, such as CRC, prostate cancer, glioma, leukemia and lung cancer (58,6063); in addition, miR-495 has been identified as a tumor suppressor in these cancer types. However, in a study conducted on breast cancer stem cells, miR-495 overexpression promoted oncogenesis (64).

Xu et al (65) demonstrated downregulation of miR-1185 in stage IV colorectal carcinoma. Tan et al (66) revealed that miR-654 acts as a tumor suppressor in breast cancer, by modulation of its target EPSTI1. Furthermore, another study indicated that miR-654 has tumor suppressor properties in papillary thyroid cancer (67). miR-495 was revealed to be downregulated in malignant cells and tissues of the breast (68), while its overexpression acts as a critical tumor suppressor in CRC cells, through targeting FAM83D (69).

Previous in vitro findings have identified an inverse correlation between miR-496 and miR-1185 expression and mTOR (1), miR-654 and miR-3183 expression and RPTOR (2), miR-495 expression and RICTOR (3) in human CRC cells. These downregulated miRNAs highlight the importance of miRNAs for use as potential tumor suppressors via targeting the mTOR signaling pathway. Further studies using 3′ luciferase reporters are needed to confirm the targets of these miRNAs in the mTOR pathway. Increasing miRNA expression levels using mimics to examine mTOR signaling and cancer progression is an important approach in CRC research. Furthermore, the role of these miRNAs requires confirmation by applying more functional in vitro and in vivo studies, such as miRNA inhibitor studies.

Acknowledgements

Not applicable.

Funding

This present study was supported by a PhD scholarship awarded to Naif Alqurashi by the Imam Abdulrahman Bin Faisal University. Funding at Griffith University was provided to Ming Wei through the higher degree research office.

Availability of data and materials

The data and materials including within the present study are available from the corresponding author upon reasonable request.

Authors contributions

SMH conceptualized the design of the present study. NA curated the data, while SMH and AF analyzed the data. FA and MQW acquired funding for the present study. NA, SMH and FA performed the experiments. MQW and SI supervised and provided administrative support for the study. NA wrote the manuscript, and SMH, FA, SI, AF and MQW critically reviewed the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no conflicts of interest.

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Zoncu R, Efeyan A and Sabatini DM: mTOR: From growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 12:21–35. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Sarbassov DD, Guertin DA, Ali SM and Sabatini DM: Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 307:1098–1101. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Bagga S, Bracht J, Hunter S, Massirer K, Holtz J, Eachus R and Pasquinelli AE: Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell. 122:553–563. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M and Croce CM: Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 101:2999–3004. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Kitade Y and Akao Y: MicroRNAs and their therapeutic potential for human diseases: microRNAs, miR-143 and −145, function as anti-oncomirs and the application of chemically modified miR-143 as an anti-cancer drug. J Pharmacol Sci. 114:276–280. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M and Rajewsky N: Combinatorial microRNA target predictions. Nat Genet. 37:495–500. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Farazi TA, Spitzer JI, Morozov P and Tuschl T: miRNAs in human cancer. J Pathol. 223:102–115. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Ciafrè SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G, Negrini M, Maira G, Croce CM and Farace MG: Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun. 334:1351–1358. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Kim YK, Yu J, Han TS, Park SY, Namkoong B, Kim DH, Hur K, Yoo MW, Lee HJ, Yang HK and Kim VN: Functional links between clustered microRNAs: Suppression of cell-cycle inhibitors by microRNA clusters in gastric cancer. Nucleic Acids Res. 37:1672–1681. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, et al: MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 65:7065–7070. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Wu WK, Law PT, Lee CW, Cho CH, Fan D, Wu K, Yu J and Sung JJ: MicroRNA in colorectal cancer: From benchtop to bedside. Carcinogenesis. 32:247–253. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Michael MZ, SM OC, van Holst Pellekaan NG, Young GP and James RJ: Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol Cancer Res. 1:882–891. 2003.PubMed/NCBI

16 

Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H, Harano T, Yatabe Y, Nagino M, Nimura Y, et al: Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res. 64:3753–3756. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Pradhan AK, Emdad L, Das SK, Sarkar D and Fisher PB: The enigma of miRNA regulation in cancer. Adv Cancer Res. 135:25–52. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Slaby O, Svoboda M, Fabian P, Smerdova T, Knoflickova D, Bednarikova M, Nenutil R and Vyzula R: Altered expression of miR-21, miR-31, miR-143 and miR-145 is related to clinicopathologic features of colorectal cancer. Oncology. 72:397–402. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Guo C, Sah JF, Beard L, Willson JK, Markowitz SD and Guda K: The noncoding RNA, miR-126, suppresses the growth of neoplastic cells by targeting phosphatidylinositol 3-kinase signaling and is frequently lost in colon cancers. Genes Chromosomes Cancer. 47:939–946. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Gaedcke J, Grade M, Camps J, Søkilde R, Kaczkowski B, Schetter AJ, Difilippantonio MJ, Harris CC, Ghadimi BM, Møller S, et al: The rectal cancer microRNAome-microRNA expression in rectal cancer and matched normal mucosa. Clin Cancer Res. 18:4919–4930. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Shah MY, Ferrajoli A, Sood AK, Lopez-Berestein G and Calin GA: microRNA therapeutics in cancer-an emerging concept. EBioMedicine. 12:34–42. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Grady WM, Parkin RK, Mitchell PS, Lee JH, Kim YH, Tsuchiya KD, Washington MK, Paraskeva C, Willson JK, Kaz AM, et al: Epigenetic silencing of the intronic microRNA hsa-miR-342 and its host gene EVL in colorectal cancer. Oncogene. 27:3880–3888. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Tazawa H, Tsuchiya N, Izumiya M and Nakagama H: Tumor-suppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc Natl Acad Sci USA. 104:15472–15477. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Alqurashi N, Hashimi SM, Alowaidi F, Ivanovski S and Wei MQ: Dual mTOR/PI3K inhibitor NVP-BEZ235 arrests colorectal cancer cell growth and displays differential inhibition of 4E-BP1. Oncol Rep. 40:1083–1092. 2018.PubMed/NCBI

25 

Alowaidi F, Hashimi SM, Alqurashi N, Alhulais R, Ivanovski S, Bellette B, Meedenyia A, Lam A and Wood S: Assessing stemness and proliferation properties of the newly established colon cancer ‘stem’ cell line, CSC480 and novel approaches to identify dormant cancer cells. Oncol Rep. 39:2881–2891. 2018.PubMed/NCBI

26 

Kiriakidou M, Nelson PT, Kouranov A, Fitziev P, Bouyioukos C, Mourelatos Z and Hatzigeorgiou A: A combined computational-experimental approach predicts human microRNA targets. Genes Dev. 18:1165–1178. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Dweep H, Sticht C, Pandey P and Gretz N: miRWalk-database: Prediction of possible miRNA binding sites by ‘walking’ the genes of three genomes. J Biomed Inform. 44:839–847. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Friedman RC, Farh KK, Burge CB and Bartel DP: Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 19:92–105. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Wang X and El Naqa IM: Prediction of both conserved and nonconserved microRNA targets in animals. Bioinformatics. 24:325–332. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Dowling RJ, Topisirovic I, Alain T, Bidinosti M, Fonseca BD, Petroulakis E, Wang X, Larsson O, Selvaraj A, Liu Y, et al: mTORC1-mediated cell proliferation, but not cell growth, controlled by the 4E-BPs. Science. 328:1172–1176. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Laplante M and Sabatini DM: mTOR signaling in growth control and disease. Cell. 149:274–293. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Melo SA and Esteller M: Dysregulation of microRNAs in cancer: Playing with fire. FEBS Lett. 585:2087–2099. 2011. View Article : Google Scholar : PubMed/NCBI

34 

AlQurashi N, Hashimi S and Wei M: Chemical inhibitors and microRNAs (miRNA) targeting the mammalian target of rapamycin (mTOR) pathway: Potential for novel anticancer therapeutics. Int J Mol Sci. 14:3874–3900. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Oğul H: Computational prediction of microRNA function and activity. Methods Mol Biol. 1107:243–256. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Yan Z, Xiong Y, Xu W, Gao J, Cheng Y, Wang Z, Chen F and Zheng G: Identification of hsa-miR-335 as a prognostic signature in gastric cancer. PLoS One. 7:e400372012. View Article : Google Scholar : PubMed/NCBI

37 

Peng HH, Zhang YD, Gong LS, Liu WD and Zhang Y: Increased expression of microRNA-335 predicts a favorable prognosis in primary gallbladder carcinoma. Onco Targets Ther. 6:1625–1630. 2013.PubMed/NCBI

38 

Wang YX, Zhang XY, Zhang BF, Yang CQ, Chen XM and Gao HJ: Initial study of microRNA expression profiles of colonic cancer without lymph node metastasis. J Dig Dis. 11:50–54. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Vickers MM, Bar J, Gorn-Hondermann I, Yarom N, Daneshmand M, Hanson JE, Addison CL, Asmis TR, Jonker DJ, Maroun J, et al: Stage-dependent differential expression of microRNAs in colorectal cancer: Potential role as markers of metastatic disease. Clin Exp Metastasis. 29:123–132. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Sarver AL, French AJ, Borralho PM, Thayanithy V, Oberg AL, Silverstein KA, Morlan BW, Riska SM, Boardman LA, Cunningham JM, et al: Human colon cancer profiles show differential microRNA expression depending on mismatch repair status and are characteristic of undifferentiated proliferative states. BMC Cancer. 9:4012009. View Article : Google Scholar : PubMed/NCBI

41 

Bandrés E, Cubedo E, Agirre X, Malumbres R, Zárate R, Ramirez N, Abajo A, Navarro A, Moreno I, Monzó M and García-Foncillas J: Identification by Real-time PCR of 13 mature microRNAs differentially expressed in colorectal cancer and non-tumoral tissues. Mol Cancer. 5:292006. View Article : Google Scholar : PubMed/NCBI

42 

Zhang J, Kong X, Li J, Luo Q, Li X, Shen L, Chen L and Fang L: miR-96 promotes tumor proliferation and invasion by targeting RECK in breast cancer. Oncol Rep. 31:1357–1363. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, Terris B, Mazzaferro V, Lowe SW, Croce CM and Dejean A: miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci USA. 107:264–269. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Navon R, Wang H, Steinfeld I, Tsalenko A, Ben-Dor A and Yakhini Z: Novel rank-based statistical methods reveal microRNAs with differential expression in multiple cancer types. PLoS One. 4:e80032009. View Article : Google Scholar : PubMed/NCBI

45 

Wang L, Zhu MJ, Ren AM, Wu HF, Han WM, Tan RY and Tu RQ: A ten-microRNA signature identified from a genome-wide microRNA expression profiling in human epithelial ovarian cancer. PLoS One. 9:e964722014. View Article : Google Scholar : PubMed/NCBI

46 

Yao Y, Suo AL, Li ZF, Liu LY, Tian T, Ni L, Zhang WG, Nan KJ, Song TS and Huang C: MicroRNA profiling of human gastric cancer. Mol Med Rep. 2:963–970. 2009.PubMed/NCBI

47 

Wang XY, Wu MH, Liu F, Li Y, Li N, Li GY and Shen SR: Differential miRNA expression and their target genes between NGX6-positive and negative colon cancer cells. Mol Cell Biochem. 345:283–290. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Chen WC, Lin MS, Ye YL, Gao HJ, Song ZY and Shen XY: microRNA expression pattern and its alteration following celecoxib intervention in human colorectal cancer. Exp Ther Med. 3:1039–1048. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Wang B, Li J, Sun M, Sun L and Zhang X: miRNA expression in breast cancer varies with lymph node metastasis and other clinicopathologic features. IUBMB Life. 66:371–377. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Gu J, Wang Y and Wu X: MicroRNA in the pathogenesis and prognosis of esophageal cancer. Curr Pharm Des. 19:1292–1300. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Sundaram P, Hultine S, Smith LM, Dews M, Fox JL, Biyashev D, Schelter JM, Huang Q, Cleary MA, Volpert OV and Thomas-Tikhonenko A: p53-responsive miR-194 inhibits thrombospondin-1 and promotes angiogenesis in colon cancers. Cancer Res. 71:7490–7501. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Zhang J, Zhao CY, Zhang SH, Yu DH, Chen Y, Liu QH, Shi M, Ni CR and Zhu MH: Upregulation of miR-194 contributes to tumor growth and progression in pancreatic ductal adenocarcinoma. Oncol Rep. 31:1157–1164. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Sampson VB, Rong NH, Han J, Yang Q, Aris V, Soteropoulos P, Petrelli NJ, Dunn SP and Krueger LJ: MicroRNA let-7a down-regulates MYC and reverts MYC-induced growth in Burkitt lymphoma cells. Cancer Res. 67:9762–9770. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Du L, Schageman JJ, Subauste MC, Saber B, Hammond SM, Prudkin L, Wistuba II, Ji L, Roth JA, Minna JD and Pertsemlidis A: miR-93, miR-98, and miR-197 regulate expression of tumor suppressor gene FUS1. Mol Cancer Res. 7:1234–1243. 2009. View Article : Google Scholar : PubMed/NCBI

55 

Deng ZQ, Yin JY, Tang Q, Liu FQ, Qian J, Lin J, Shao R, Zhang M and He L: Over-expression of miR-98 in FFPE tissues might serve as a valuable source for biomarker discovery in breast cancer patients. Int J Clin Exp Pathol. 7:1166–1171. 2014.PubMed/NCBI

56 

Nagaraja AK, Creighton CJ, Yu Z, Zhu H, Gunaratne PH, Reid JG, Olokpa E, Itamochi H, Ueno NT, Hawkins SM, et al: A link between mir-100 and FRAP1/mTOR in clear cell ovarian cancer. Mol Endocrinol. 24:447–463. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Duan Z, Choy E, Harmon D, Liu X, Susa M, Mankin H and Hornicek F: MicroRNA-199a-3p is downregulated in human osteosarcoma and regulates cell proliferation and migration. Mol Cancer Ther. 10:1337–1345. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Chen SM, Chen HC, Chen SJ, Huang CY, Chen PY, Wu TW, Feng LY, Tsai HC, Lui TN, Hsueh C and Wei KC: MicroRNA-495 inhibits proliferation of glioblastoma multiforme cells by downregulating cyclin-dependent kinase 6. World J Surg Oncol. 11:872013. View Article : Google Scholar : PubMed/NCBI

59 

Fornari F, Milazzo M, Chieco P, Negrini M, Calin GA, Grazi GL, Pollutri D, Croce CM, Bolondi L and Gramantieri L: miR-199a-3p regulates mTOR and c-Met to influence the doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res. 70:5184–5193. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Formosa A, Markert EK, Lena AM, Italiano D, Finazzi-Agro' E, Levine AJ, Bernardini S, Garabadgiu AV, Melino G and Candi E: MicroRNAs, miR-154, miR-299-5p, miR-376a, miR-376c, miR-377, miR-381, miR-487b, miR-485-3p, miR-495 and miR-654-3p, mapped to the 14q32.31 locus, regulate proliferation, apoptosis, migration and invasion in metastatic prostate cancer cells. Oncogene. 33:5173–5182. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Jiang X, Huang H, Li Z, He C, Li Y, Chen P, Gurbuxani S, Arnovitz S, Hong GM, Price C, et al: miR-495 is a tumor-suppressor microRNA down-regulated in MLL-rearranged leukemia. Proc Natl Acad Sci USA. 109:19397–19402. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Chu H, Chen X, Wang H, Du Y, Wang Y, Zang W, Li P, Li J, Chang J, Zhao G and Zhang G: miR-495 regulates proliferation and migration in NSCLC by targeting MTA3. Tumour Biol. 35:3487–3494. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Chuang AY, Chuang JC, Zhai Z, Wu F and Kwon JH: NOD2 expression is regulated by microRNAs in colonic epithelial HCT116 cells. Inflamm Bowel Dis. 20:126–135. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Hwang-Verslues WW, Chang PH, Wei PC, Yang CY, Huang CK, Kuo WH, Shew JY, Chang KJ, Lee EY and Lee WH: miR-495 is upregulated by E12/E47 in breast cancer stem cells, and promotes oncogenesis and hypoxia resistance via downregulation of E-cadherin and REDD1. Oncogene. 30:2463–2474. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Xu XH, Wu XB, Wu SB, Liu HB, Chen R and Li Y: Identification of miRNAs differentially expressed in clinical stages of human colorectal carcinoma-an investigation in Guangzhou, China. PLoS One. 9:e940602014. View Article : Google Scholar : PubMed/NCBI

66 

Tan YY, Xu XY, Wang JF, Zhang CW and Zhang SC: miR-654-5p attenuates breast cancer progression by targeting EPSTI1. Am J Cancer Res. 6:522–532. 2016.PubMed/NCBI

67 

Geraldo MV, Nakaya HI and Kimura ET: Down-regulation of 14q32-encoded miRNAs and tumor suppressor role for miR-654-3p in papillary thyroid cancer. Oncotarget. 8:9597–9607. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Wang L, Liu JL, Yu L, Liu XX, Wu HM, Lei FY, Wu S and Wang X: Downregulated miR-495 [Corrected] inhibits the G1-S phase transition by targeting Bmi-1 in breast cancer. Medicine (Baltimore). 94:e7182015. View Article : Google Scholar : PubMed/NCBI

69 

Yan L, Yao J and Qiu J: miRNA-495 suppresses proliferation and migration of colorectal cancer cells by targeting FAM83D. Biomed Pharmacother. 96:974–981. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 18 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Alqurashi N, Hashimi SM, Alowaidi F, Ivanovski S, Farag A and Wei MQ: miR‑496, miR‑1185, miR‑654, miR‑3183 and miR‑495 are downregulated in colorectal cancer cells and have putative roles in the mTOR pathway. Oncol Lett 18: 1657-1668, 2019
APA
Alqurashi, N., Hashimi, S.M., Alowaidi, F., Ivanovski, S., Farag, A., & Wei, M.Q. (2019). miR‑496, miR‑1185, miR‑654, miR‑3183 and miR‑495 are downregulated in colorectal cancer cells and have putative roles in the mTOR pathway. Oncology Letters, 18, 1657-1668. https://doi.org/10.3892/ol.2019.10508
MLA
Alqurashi, N., Hashimi, S. M., Alowaidi, F., Ivanovski, S., Farag, A., Wei, M. Q."miR‑496, miR‑1185, miR‑654, miR‑3183 and miR‑495 are downregulated in colorectal cancer cells and have putative roles in the mTOR pathway". Oncology Letters 18.2 (2019): 1657-1668.
Chicago
Alqurashi, N., Hashimi, S. M., Alowaidi, F., Ivanovski, S., Farag, A., Wei, M. Q."miR‑496, miR‑1185, miR‑654, miR‑3183 and miR‑495 are downregulated in colorectal cancer cells and have putative roles in the mTOR pathway". Oncology Letters 18, no. 2 (2019): 1657-1668. https://doi.org/10.3892/ol.2019.10508