Downregulation of microRNA‑629‑5p in colorectal cancer and prevention of the malignant phenotype by direct targeting of low‑density lipoprotein receptor‑related protein 6

Retraction in: /10.3892/ijmm.2021.5057

  • Authors:
    • Guoqiang Yan
    • Chenyao Li
    • Yuhang Zhao
    • Meng Yue
    • Lei Wang
  • View Affiliations

  • Published online on: June 18, 2019     https://doi.org/10.3892/ijmm.2019.4245
  • Pages: 1139-1150
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Aberrant expression of numerous microRNAs (miRNAs/miRs) in colorectal cancer (CRC) significantly affects disease progression. Recently, miR‑629‑5p (miR‑629) was identified as a tumor‑promoting miRNA in the malignant processes of a number of human cancers. However, few studies have been conducted regarding expression profiles and detailed roles of miR‑629 in CRC. In the present study, reverse transcription‑quantitative polymerase chain reaction was used to assess miR‑629 expression in CRC tissues and cell lines. Cell Counting Kit‑8 assay, flow cytometry and Transwell assays were performed to determine the in vitro effects of miR‑629 on CRC cell proliferation, apoptosis, and metastasis, respectively. Xenograft models were employed to determine the in vivo effects of miR‑629 on tumor growth in nude mice. Molecular mechanisms underlying the activity of miR‑629 in CRC cells were explored. miR‑629 expression decreased in CRC tissues and cell lines. The decreased aberrant miR‑629 expression was significantly associated with tumor size, lymphatic metastasis and tumor‑node‑metastasis stage of CRC, and was a predictor of poor prognosis. Restoring miR‑629 expression attenuated CRC cell proliferation, migration and invasion; promoted cell apoptosis in vitro; and inhibited tumor growth in vivo. Low‑density lipoprotein receptor‑related protein 6 (LRP6) was a direct target gene of miR‑629 in CRC cells. Furthermore, the effect of LRP6 knockdown was similar to that of miR‑629 overexpression in CRC cells. Restoration of LRP6 expression neutralized the effects of miR‑629 in CRC cells. miR‑629 suppressed the activation of the Wnt/β‑catenin pathway through LRP6 regulation both in vitro and in vivo. In conclusion, miR‑629 suppressed the development and progression of CRC by directly targeting LRP6 and inhibiting the Wnt/β‑catenin pathway both in vitro and in vivo. Therefore, miR‑629 may be a novel prognostic biomarker and therapeutic target in CRC.

Introduction

Colorectal cancer (CRC) is the third-most common human malignancy and the fourth-most common cause of cancer-related death worldwide (1). A total of ~1.2 million new cases of CRC and 600,000 fatalities due to CRC have been estimated to occur annually throughout the world (2). Early diagnosis of CRC is challenging owing to the lack of effective diagnostic approaches; therefore, the majority of CRC cases are diagnosed at advanced stages (3). Although the current multimodal treatments for CRC have advanced rapidly (4), their therapeutic effects have been unsatisfactory and long-term survival remains poor (5). Multiple risk factors, such as poor dietary habits, obesity, alcohol consumption and smoking, are implicated in the pathogenesis of CRC (6); however, detailed mechanisms underlying the genesis and development of CRC remain to be elucidated. Therefore, uncovering the molecular bases of crucial tumorigenic events is imperative to identify effective targets for the diagnosis and treatment of CRC.

In recent years, numerous studies have demonstrated that microRNAs (miRNAs/miRs) contribute to the genesis and development of tumors (7-9). miRNAs are a group of single-stranded, noncoding RNAs ranging from 19 to 23 nucleotides in length (10). miRNAs play important roles in the regulation of gene expression via direct interactions with the 3′ untranslated (UTRs) regions of their target genes (11). Imperfect base pairing with specific sequences promotes mRNA degradation and/or translational suppression (12). Over 1,500 mature miRNAs have been identified in the human genome, which are speculated to regulate ~30% of the human protein-coding genes (13). Studies exploring miRNA expression profiles in CRC have indicated that a number of miRNAs are aberrantly expressed and that this aberrant expression is closely associated with the development and progression of CRC (14-16). Therefore, miRNAs might be potential biomarkers for the diagnosis, prognosis and therapy of CRC.

Recent studies have indicated that miR-629-5p (miR-629) plays important roles in the malignant processes of a number of human cancers, such as breast cancer (17), hepatocellular carcinoma (18), nasopharyngeal carcinoma (19) and cervical cancer (20). However, few studies have examined expression profiles and specific roles of miR-629 in CRC. The present study assessed miR-629 expression in CRC and investigated its effects on the aggressive behavior of CRC cells in vitro and in vivo. Furthermore, the molecular mechanisms underlying the activity of miR-629 in CRC were comprehensively explored.

Materials and methods

Patients and tumor specimens

Human CRC tissues and paired adjacent normal colorectal tissues were obtained from 51 patients (17 males and 34 females; age range, 47-71 years; mean age, 58 years) with CRC who presented to the Department of Colorectal and Anal Surgery in The First Hospital of Jilin University (Changchun, China) between January 2012 and March 2018. Patients who were treated with preoperative radiotherapy or chemotherapy were excluded from the study. After tissue excision, all specimens were quickly frozen in liquid nitrogen and stored at −80°C. The study was approved by the Ethics Committee of The First Hospital of Jilin University and all patients provided written informed consent.

Cell lines

A total of four human CRC cell lines, namely HT29, HCT116, SW480 and SW620, as well as a normal human colon epithelium cell line (FHC) were purchased from the American Type Culture Collection. Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific Inc.) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific Inc.), 100 U/ml penicillin and 100 mg/ml streptomycin (Gibco; Thermo Fisher Scientific Inc.) were used for cell culture. The cultures were incubated at 37°C in a humidified incubator at 5% CO2.

Transfection experiment

The agomir-629 and agomir-negative control (NC) were purchased from Shanghai GenePharma Co., Ltd. The agomir-629 sequence was 5′-UGG GUU UAC GUU GGG AGA ACU-3′ and the agomir-NC sequence was 5′-UUC UCC GAA CGU GUC ACG UTT-3′. The low-density lipoprotein receptor-related protein 6 (LRP6) small interfering (si)RNA that silences endogenous LRP6 expression and the NC siRNA were synthesized by and purchased from Guangzhou RiboBio Co. Ltd. The LRP6 siRNA sequence was 5′-CCA CAA AUC CAU GUG GAA UTT-3′ and the NC siRNA sequence was 5′-UUC UCC GAA CGU GUC ACG UTT-3′. The LRP6 overexpression plasmid pcDNA3.1-LRP6 and the empty pcDNA3.1 plasmid were obtained from Wanleibio Co., Ltd. Cells in the logarithmic phase were harvested and resuspended in culture medium. Cell suspension (2 ml) containing 6×105 cells was inoculated into each well of the 6-well plates. After overnight incubation, cells were transfected with agomir-629 (50 nM), agomir-NC (50 nM), LRP6 siRNA (100 pmol), NC siRNA (100 pmol), pcDNA3.1-LRP6 (4 µg), or pcDNA3.1 (4 µg) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), flow cytometry and Transwell assay were performed 48 h post-transfection. Cell Counting Kit-8 assay and animal studies were conducted 24 h post-transfection.

RT-qPCR

Total RNA was isolated from the tissue samples and cells using the TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). To determine miR-629 expression, single-stranded complementary DNA was synthesized from total RNA using the miScript Reverse Transcription kit (Qiagen GmbH). The temperature protocol for RT was as follows: 37°C for 60 min, 95°C for 5 min and storage at 4°C. Thereafter, qPCR was performed using the miScript SYBR-Green PCR kit (Qiagen GmbH) on ABI 7900 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific Inc.). For LRP6 mRNA quantification, cDNA was synthesized was using the PrimeScript RT-Reagent kit (Takara Bio, Inc.) and subjected to qPCR using the SYBR Premix Ex Taq™ kit (Takara Bio, Inc.). The temperature protocol for qPCR was as follows: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec, and a final extension step at 72°C for 35 sec. Relative miR-629 and LRP6 expression were analyzed using the 2−ΔΔCq method (21) and normalized to U6 small nuclear RNA and GAPDH expression.

Sequences of designed primers were as follows: miR-629, 5′-CGT GGG TTT ACG TTG GG-3′ (forward) and 5′-CTC GCT TCG GCA GCA CA-3′ (reverse); U6, 5′-CTC GCT TCG GCA GCA CA-3′ (forward) and 5′-AAC GCT TCA CGA ATT TGC GT-3′ (reverse); LRP6, 5′-ACG ATT GTA GTT GGA GGC TTG-3′ (forward) and 5-ATG GCTT CTT CGC TGA CAT CA-3′ (reverse); and GAPDH, 5′-GGA GTC AAC GGA TTT GGT-3′ (forward) and 5′-GTG ATGG GAT TTC CAT TGA T-3′ (reverse).

Cell Counting Kit-8 assay

Transfected cells were collected and suspended in culture medium. Cell suspension (100 µl) containing 3×103 cells was seeded into 96-well plates. Cellular proliferation was detected after incubation for 0, 24, 48 and 72 h. Briefly, 20 µl Cell Counting Kit (CCK)-8 solution (Beyotime Institute of Biotechnology) was added to each well prior to being incubated at 37°C for another 2 h. Following incubation, absorbance was detected at a wavelength of 450 nm using the iMark microplate absorbance reader (Bio-Rad Laboratories, Inc.).

Flow cytometry

Transfected cells (1.0×106/well) in 6-well plates were collected after 48 h of incubation and the rate of apoptosis was measured using the Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit (Biolegend, Inc.). Briefly, cells were washed with ice-cold phosphate-buffered solution (Gibco; Thermo Fisher Scientific Inc.) and resuspended in 100 µl binding buffer. Thereafter, cells were double-labeled with 5 µl Annexin V-FITC and 5 µl propidium iodide by incubating at room temperature for 30 min in the dark prior to quantification using a flow cytometer (FACScan™; BD Biosciences; Becton, Dickinson and Company). Data was analyzed with CellQuest™ software version 5.1 (BD Biosciences; Becton, Dickinson and Company).

Transwell assay

Transwell chambers (BD Biosciences; Becton, Dickinson and Company) precoated with Matrigel (BD Biosciences; Becton, Dickinson and Company) were employed to determine the invasive ability of cells. The migratory capacity of cells was determined using non-Matrigel-coated Transwell chambers. Transfected cells were collected at 48 h post-transfection and resuspended in FBS-free DMEM. A cell suspension (200 µl) containing 1×105 cells was inoculated in the upper compartment and 500 µl DMEM medium with 20% FBS was seeded in the lower compartment. After 24 h of incubation at 37°C, the non-migrated and non-invaded cells were removed with a cotton swab. Cells on the lower chamber membrane were fixed with 70% ethanol at room temperature for 30 min and stained with 0.5% crystal violet at room temperature for 30 min. Their migratory and invasive abilities were quantified by counting the number of migrated and invaded cells in five randomly selected visual fields per chamber under an Olympus BX50 light microscope (magnification, ×200; Olympus Corporation).

Xenograft model in nude mice

A total of eight female 4-week-old BALB/c nude mice (20 g) were purchased from the Animal Center of Southern Medical University. The animals were maintained under specific pathogen-free conditions (25°C; 50% humidity; 10-h light/14-h dark cycle) and access to food/water ad libitum. For the tumorigenesis assays, 1×107 HCT116 cells transfected with agomir-629 or agomir-NC were subcutaneously injected into the flanks of each mice (n=4, each group). The tumor xenograft size was measured using a Vernier caliper and tumor volume was calculated using the following formula: 1/2× (tumor length x tumor width2). All animals were euthanized at 4 weeks after inoculation. Tumor xenografts were excised, weighed and stored for further use. During the assay, the nude mice were subjected to euthanasia when the tumor size reached 2 cm. The maximum tumor diameter and volume observed during the xenograft study was 1.3 cm and 2,460 mm3, respectively. All protocols involving animals were approved by the Ethics Committee of The First Hospital of Jilin University (201706-12).

Bioinformatics target prediction

A total of three miRNA target prediction tools, including miRDB (http://www.mirdb.org/), Target Scan (http://www.targetscan.org/) and miRanda (http://www.microrna.org), were used for predicting miR-629 targets.

Luciferase reporter assay

The wild-type (wt) LRP6 3′-UTR sequence carrying the miR-629-binding site and the mutant (mut) LRP6 3′-UTR were amplified by Shanghai GenePharma Co., Ltd. The 3′-UTR wt and mut fragments were then inserted into the pMIR-REPORT vector (Promega Corporation) to obtain pMIR-LRP6-3′-UTR wt and pMIR-LRP6-3′-UTR mut, respectively. For the reporter assay, cells were plated onto 24-well plates and incubated overnight prior to transfection. The recombinant luciferase reporter plasmids (0.8 µg) were co-transfected with agomir-629 (20 pmol) or agomir-NC (20 pmol) into cells using Lipofectamine® 2000. After 48 h of culture, the transfected cells were harvested and luciferase activity was measured using a dual-luciferase reporter assay system (Promega Corporation). Luciferase activity was normalized to firefly luciferase activity.

Western blotting

The tissue specimens, cultured cells and tumor xenografts were lysed in radioimmunoprecipitation assay buffer (Sigma-Aldrich; Merck KGaA). The bicinchoninic acid assay (Beyotime Institute of Biotechnology) was used to quantify total protein concentration. Equal amounts of protein (30 µg) were loaded, electrotransferred to polyvinylidene fluoride membranes (Beyotime Institute of Biotechnology) and blocked with 5% fat-free milk diluted in TBS containing 0.05% Tween-20 (TBST). Then, the membranes were incubated overnight at 4°C with the following primary antibodies: Mouse anti-human monoclonal LRP6 antibody (cat. no. sc-25317; 1:1,000; Santa Cruz Biotechnology, Inc.), mouse anti-human monoclonal β-catenin antibody (cat. no. sc-59737; 1:1,000; Santa Cruz Biotechnology, Inc.), mouse anti-human monoclonal phosphorylated (p)-β-catenin (Tyr 86 phosphorylated) antibody (cat. no. sc-57534; 1:1,000; Santa Cruz Biotechnology, Inc.), rabbit anti-human monoclonal cyclin D1 antibody (cat. no. ab134175; 1:1,000; Abcam) and rabbit anti-human GAPDH antibody (cat. no. ab128915; 1:1,000; Abcam). GAPDH was used as a loading control. After washing with TBST three times, the membranes were incubated with goat anti-rabbit (cat. no. ab97051; 1:5,000; Abcam) or goat anti-mouse (cat. no. ab6789; 1:5,000; Abcam) horseradish peroxidase-conjugated IgG secondary antibodies. Finally, protein signals were visualized using Pierce™ ECL Western Blotting Substrate (Pierce; Thermo Fisher Scientific, Inc.). Quantity One software version 4.62 (Bio-Rad Laboratories, Inc.) was used to analyze protein signals.

Statistical analysis

All results are presented as the mean and standard deviation from at least three independent experiments, and were analyzed using SPSS software (version 17; SPSS, Inc., Chicago, IL, USA). The association between miR-629 and clinicopathological characteristics of patients with CRC was examined using a chi-squared test. Kaplan-Meier survival curves were plotted to explore the prognostic value of miR-629. Differences between two groups were analyzed using a Student's t-test. One-way analysis of variance, followed by Bonferroni's post hoc test, was used to compare differences between multiple groups. Correlation between miR-629 and LRP6 expression in CRC tissues was investigated using Pearson's correlation analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-629 expression decreases in CRC

Whether miR-629 was aberrantly expressed in 51 CRC tissues and paired adjacent normal colorectal tissues was examined using RT-qPCR. miR-629 expression in CRC tissues was significantly downregulated compared with in adjacent normal colorectal tissues (P<0.05; Fig. 1A). miR-629 expression in all four tested CRC cell lines, including HT29, HCT116, SW480 and SW620, was significantly decreased compared with the FHC line (P<0.05; Fig. 1B).

To explore the prognostic significance of miR-629 in patients with CRC, all patients were divided into miR-629 high expression (n=25) or miR-629 low expression (n=26) using the median value of miR-629 expression in CRC tissues as a cutoff. Decreased miR-629 expression was significantly associated with tumor size (P= 0.012), lymphatic metastasis (P=0.009) and tumor-node-metastasis (TNM) stage (P= 0.040); however, there was no clear association with sex, age, or tumor location (Table I). In addition, patients in the low miR-629 expression group showed poorer overall survival compared with those in the high miR-629 expression group (P=0.0339; Fig. 1C). These results demonstrated that miR-629 downregulation may be closely associated with poor prognosis in patients with CRC.

Table I

Association between miR-629 expression and clinicopathological features in patients with CRC.

Table I

Association between miR-629 expression and clinicopathological features in patients with CRC.

Clinicopathological featuresmiR-629 low expression group (n=26)miR-629 high expression group (n=25)P-value
Sex0.237
 Male116
 Female1519
Age, years0.267
 <601611
 ≥601014
Tumor location0.565
 Rectum810
 Colon1815
Tumor size, cm0.012a
 <5918
 ≥5177
Lymphatic metastasis0.009a
 Absence1120
 Presence155
TNM stage0.040a
 I-II1320
 III-IV135

a P<0.05 vs. miR-629 high expression group. TNM, tumor-node-metastasis; miR, microRNA.

miR-629 inhibits CRC cell proliferation and metastasis and increases cell apoptosis in vitro

Of the four CRC cell lines tested, miR-629 expression was low in the HCT116 and SW620 cell lines; thus, these two cell lines were selected for the following experiments. To investigate the specific roles of miR-629 in the development of CRC, agomir-629 or agomir-NC was transfected into HCT116 and SW620 cells and miR-629 significant overexpression was confirmed via RT-qPCR (P<0.05; Fig. 2A). Exogenous miR-629 expression significantly suppressed proliferation (P<0.05; Fig. 2B) and significantly induced apoptosis (P<0.05; Fig. 2C) in the HCT116 and SW620 cell lines, as was evident from the CCK-8 assay and flow cytometry. Furthermore, a Transwell assay was used to determine the migratory and invasive capacities of HCT116 and SW620 cells upon miR-629 overexpression. The migration (P<0.05; Fig. 2D) and invasion (P<0.05; Fig. 2E) of HCT116 and SW620 cells was significantly suppressed after transfection with agomir-629 compared to that after transfection with agomir-NC. These results strongly suggest that miR-629 exerts suppressive effects on CRC growth and metastasis in vitro.

LRP6 is a direct target of miR-629 in CRC

To clarify the molecular mechanism through which miR-629 exerts its tumor-suppressing role in CRC, bioinformatics analysis was performed to determine the putative target of miR-629. A total of three miRNA target prediction tools identified a potential binding site of miR-629 in the 3′-UTR of the LRP6 gene (Fig. 3A). To confirm this prediction, RT-qPCR was performed to measure LRP6 expression in CRC tissues and paired adjacent normal colorectal tissues. LRP6 expression in CRC tissues was significantly upregulated compared with that in adjacent normal colorectal tissues (P<0.05; Fig. 3B). LRP6 mRNA level was inversely correlated with miR-629 level in CRC tissues (P<0.0001; Fig. 3C; R2=0.3873). Furthermore, mRNA (P<0.05; Fig. 3D) and protein (P<0.05; Fig. 3E) expression levels of LRP6 in the miR-629 high expression group were decreased compared with in the miR-629 low expression group. Moreover, RT-qPCR and western blotting revealed that ectopic miR-629 expression significantly decreased LRP6 expression in HCT116 and SW620 cells at the mRNA (P<0.05; Fig. 3F) and protein (P<0.05; Fig. 3G) levels. These results indicated that miR-629 negatively regulated LRP6 expression in both CRC tissues and cell lines.

Additionally, a luciferase reporter assay was performed to ascertain whether LRP6 was modulated by miR-629 in CRC via direct binding to its 3′-UTR region. miR-629 upregulation significantly decreased the luciferase activity of the reporter plasmid harboring the wt miR-629 binding site (P<0.05; Fig. 3H); however, mutations in the miR-629-binding sequences in the 3′-UTR of LRP6 reversed the suppressive effects of miR-629 overexpression in HCT116 and SW620 cells. Overall, these results suggest that LRP6 is a direct target gene of miR-629 in CRC cells.

LRP6 silencing mimics the tumor-suppressing roles of miR-629 in CRC cells

Since LRP6 was demonstrated to be a direct target gene of miR-629 in CRC cells, whether miR-629 inhibited CRC progression via LRP6 regulation was further explored. A gene-specific siRNA was employed to silence endogenous LRP6 expression in HCT116 and SW620 cells, and the transfection efficiency was confirmed by western blotting (P<0.05; Fig. 4A). Following LRP6 knockdown, cell proliferation was significantly inhibited (P<0.05; Fig. 4B) and cell apoptosis was significantly promoted (P<0.05; Fig. 4C) in both HCT116 and SW620 cells. Then, a Transwell assay was performed to determine effects of LRP6 knockdown in the regulation of CRC cell metastasis. LRP6 knockdown significantly attenuated the migration (P<0.05; Fig. 4D) and invasion (P<0.05; Fig. 4E) of HCT116 and SW620 cells. These results indicated that LRP6 knockdown conferred effects similar to miR-629 overexpression in CRC cells, suggesting that LRP6 is a functional target of miR-629 in CRC cells.

LRP6 restoration abrogates the inhibitory effects of miR-629 upregulation in CRC cells

Rescue experiments performed to confirm whether LRP6 downregulation was essential for the tumor-suppressive roles of miR-629 in CRC cells. First, RT-qPCR was performed to detect LRP6 mRNA expression in HCT116 and SW620 cells following LRP6 overexpression plasmid pcDNA3.1-LRP6 or empty pcDNA3.1 plasmid transfection. The empty pcDNA3.1 plasmid was used as a control for pcDNA3.1-LRP6 transfection. After transfection, LRP6 mRNA expression was significantly upregulated in pcDNA3.1-LRP6-transfected HCT116 and SW620 cells (P<0.05; Fig. 5A). Afterwards, LRP6 protein expression in agomir-629-trasnfected HCT116 and SW620 cells was significantly restored through co-transfection with the pcDNA3.1-LRP6 (P<0.05; Fig. 5B). Induced miR-629 overexpression significantly restricted proliferation (P<0.05; Fig. 5C and D), promoted apoptosis (P<0.05; Fig. 5E), and decreased migration (P<0.05; Fig. 5F) and invasion (P<0.05; Fig. 5G) of HCT116 and SW620 cells, whereas restoration of LRP6 expression abrogated all these effects. These results further confirmed that LRP6 downregulation was essential for tumor-suppressing roles of miR-629 in CRC cells.

miR-629 inhibits the Wnt/β-catenin signaling pathway in CRC cells

To further explore the mechanism underlying the anticancer roles of miR-629 in CRC cells, whether miR-629 was involved in the regulation of the Wnt/β-catenin signaling pathway, which is regulated by LRP6 was assessed (22-24). Western blotting revealed that miR-629 overexpression significantly downregulated the expression of p-β-catenin and cyclin D1 proteins in HCT116 and SW620 cells. However, this inhibitory effect was rescued in agomir-629-trans-fected HCT116 and SW620 cells via co-transfection with pcDNA3.1-LRP6 (P<0.05; Fig. 6). These results demonstrate that miR-629 inhibits the Wnt/β-catenin pathway in CRC cells via LRP6 regulation.

miR-629 plays an inhibitory role in CRC tumor growth in vivo

Effects of miR-629 on CRC tumor growth in vivo were explored using a xenograft model in nude mice. Time-dependent analysis indicated that the volume of tumor xenografts was significantly decreased in the agomir-629 group compared with in the agomir-NC group (P<0.05; Fig. 7A). Representative images of the tumor xenograft derived from agomir-NC- or agomir-629-transfected HCT116 cells are shown in Fig. 7B. In addition, the tumor xenografts from the agomir-629 group were lighter compared with in the agomir-NC group (P<0.05; Fig. 7C). Total RNA of the tumor xenografts was isolated and subjected to RT-qPCR for the measurement of miR-629 expression. miR-629 was significantly overexpressed in the tumor xenografts derived from agomir-629-transfected HCT116 cells (P<0.05; Fig. 7D) and this miR-629 upregulation inhibited CRC tumor growth in vivo. Furthermore, western blotting revealed that LRP6, p-β-catenin and cyclin D1 expression was notably downregulated in the tumor xenografts from the agomir-629 group (Fig. 7E). These results suggest that miR-629 inhibits CRC tumor growth in vivo via regulation of the LRP6/Wnt/β-catenin pathway.

Discussion

Emerging evidence in the past decades has demonstrated aberrant expression of numerous miRNAs in CRC (16,25,26). Moreover, miRNA dysregulation has been shown to play a significant role in the oncogenicity of CRC by regulating all aspects of aggressive cell behavior, such as proliferation, division, apoptosis, metastasis and angiogenesis (15,27,28). Therefore, in-depth exploration of cancer-associated miRNAs in CRC may provide novel insights into the mechanism underlying CRC development and progression and may facilitate the identification of promising diagnostic and therapeutic targets in CRC. The present study attempted to determine the expression profile of miR-629 and assessed its clinical value in CRC. The relevance of miR-629 expression to CRC cell behaviors such as proliferation, apoptosis, migration and invasion in vitro as well as tumor growth in vivo was examined. Furthermore, the molecular mechanisms underlying the role of miR-629 in CRC cells in vitro and in vivo were studied. To the best of our knowledge, this is the first study to highlight the link between miR-629 and malignant processes in CRC.

miR-629 expression is upregulated in breast cancer and high miR-629 expression is closely related to decreased overall and disease-free survival (17). miR-629 has been identified as an independent risk factor for lung metastasis in patients with breast cancer (17). Moreover, miR-629 is upregulated in multiple cancer types, including hepatocellular carcinoma (18), nasopharyngeal carcinoma (19), cervical cancer (20), ovarian cancer (29), clear cell renal cell carcinoma (30) and pancreatic cancer (31). However, the expression profile of miR-629 in CRC requires further investigation. In this study, the low expression of miR-629 in CRC tissues and cell lines was first demonstrated. Decreased miR-629 expression was associated with tumor size, lymphatic metastasis and TNM stage of CRC. Moreover, CRC patients with low miR-629 expression exhibited decreased overall survival. These findings suggest that miR-629 is can be a diagnostic and prognostic biomarker for CRC.

miR-629 plays oncogenic roles in the genesis and progression of cancer. For instance, miR-629 silencing inhibits breast cancer cell viability, reduces migratory ability in vitro and suppresses tumor growth in vivo (17). In contrast, miR-629 upregulation promotes cell proliferation, migration and invasion in nasopharyngeal carcinoma (19). Furthermore, miR-629 knockdown suppresses the migratory and invasive capacities of clear cell renal cell carcinoma cells (30). In addition, miR-629 downregulation decreases cell proliferation, induces cell apoptosis and improves chemosensitivity to 1′S-1′-acetoxychavicol acetate in cervical cancer (20). miR-629 has also been identified as a tumor-promoting miRNA in ovarian (29) and pancreatic cancers (31). However, to the best of our knowledge no studies have focused on detailed roles of miR-629 in the oncogenicity of CRC in vitro and in vivo. In this study, a series of functional assays revealed that exogenous miR-629 expression suppressed CRC cell proliferation, migration and invasion in vitro; promoted cell apoptosis in vitro; and suppressed tumor growth in vivo. These results suggest that miR-629 can be a potential therapeutic target in CRC.

Multiple genes, including LIFR (17), PDCD4 (19), RSU1 (20), TSPYL5 (29), RIM33 (30) and FOXO3 (31), have been validated as direct target genes of miR-629. Detailed investigation of mechanisms underlying the action of miR-629 in CRC may provide novel therapeutic approaches for patients with CRC. LRP6, a member of the Ras super-family of Rho GTPases, was confirmed as a novel target of miR-629 in CRC cells. LRP6 is upregulated in various human cancers, including breast cancer (32), osteosarcoma (24), oral squamous cell carcinoma (33), thyroid cancer (34) and hepatocellular carcinoma (35). Moreover, LRP6 activates the Wnt/β-catenin pathway, promoting the genesis and development of tumors (36,37). LRP6 is expressed at high levels in CRC (38) and is involved in the regulation of its malignant development in vitro and in vivo (23,39). In the present, it was demonstrated that miR-629 directly targeted LRP6 and suppressed the Wnt/β-catenin pathway, thereby controlling proliferation, apoptosis, migration, and invasion of CRC cells in vitro and tumor growth in vivo. As expected, restoration of miR-629 expression induced LRP6 silencing and Wnt/β-catenin signaling inhibition, which may be a novel therapeutic approach to CRC.

Nonetheless, there is a limitation of this study. Transcriptional activity of catenin was not detected, which will be addressed in future investigations.

In conclusion, decreased miR-629 expression and its inhibitory roles in the development of CRC was demonstrated. Notably, concrete evidence was provided that miR-629 inhibits the oncogenicity of CRC by directly targeting LRP6 and thereby inhibiting the downstream Wnt/β-catenin pathway. Finally, the results of the present study offer advanced understanding of the pathogenesis of CRC as well as shed light on novel prognostic indicators and therapeutic targets in CRC.

Acknowledgments

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

All authors have made a significant contribution to the study. The present study was designed by LW and GY. RT-qPCR, CCK-8 assay, flow cytometry and bioinformatics target prediction were performed by GY and CL. Transwell assay and xenograft model experiments in nude mice were conducted by YZ and MY. Luciferase reporter assay and western blotting were conducted by LW. All authors have read and approved the final draft of the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of The First Hospital of Jilin University (Changchun, China) and all patients provided written informed consent. All protocols involving animals were approved by the Ethics Committee of The First Hospital of Jilin University (201706-12).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Tenesa A and Dunlop MG: New insights into the aetiology of colorectal cancer from genome-wide association studies. Nat Rev Genet. 10:353–358. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Brenner H, Kloor M and Pox CP: Colorectal cancer. Lancet. 383:1490–1502. 2014. View Article : Google Scholar

3 

Labianca R and Merelli B: Screening and diagnosis for colorectal cancer: Present and future. Tumori. 96:889–901. 2010. View Article : Google Scholar

4 

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

5 

Haggar FA and Boushey RP: Colorectal cancer epidemiology: Incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 22:191–197. 2009. View Article : Google Scholar :

6 

Center MM, Jemal A, Smith RA and Ward E: Worldwide variations in colorectal cancer. CA Cancer J Clin. 59:366–378. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Lo Russo G, Tessari A, Capece M, Galli G, de Braud F, Garassino MC and Palmieri D: MicroRNAs for the diagnosis and management of malignant pleural mesothelioma: A literature review. Front Oncol. 8:6502018. View Article : Google Scholar

8 

Mollaei H, Safaralizadeh R and Rostami Z: MicroRNA replacement therapy in cancer. J Cell Physiol. 234:12369–12384. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Ors-Kumoglu G, Gulce-Iz S and Biray-Avci C: Therapeutic microRNAs in human cancer. Cytotechnology. 71:411–425. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Kala R, Peek GW, Hardy TM and Tollefsbol TO: MicroRNAs: An emerging science in cancer epigenetics. J Clin Bioinforma. 3:62013. View Article : Google Scholar : PubMed/NCBI

11 

Stark A, Brennecke J, Bushati N, Russell RB and Cohen SM: Animal MicroRNAs confer robustness to gene expression and have a significant impact on 3′UTR evolution. Cell. 123:1133–1146. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Hede K: Studies define role of microRNA in cancer. J Natl Cancer Inst. 97:1114–1115. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A and Enright AJ: miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 34(Database Issue): D140–D144. 2006. View Article : Google Scholar :

14 

Huang S, Tan X, Huang Z, Chen Z, Lin P and Fu SW: microRNA biomarkers in colorectal cancer liver metastasis. J Cancer. 9:3867–3873. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Ding L, Lan Z and Xiong X: The dual role of micrornas in colorectal cancer progression. Int J Mol Sci. 19:pii: E2791. 2018. View Article : Google Scholar

16 

Yang S, Sun Z, Zhou Q, Wang W, Wang G, Song J, Li Z, Zhang Z, Chang Y, Xia K, et al: MicroRNAs, long noncoding RNAs, and circular RNAs: Potential tumor biomarkers and targets for colorectal cancer. Cancer Manag Res. 10:2249–2257. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Wang J, Song C, Tang H, Tang H, Zhang C, Tang J, Li X, Chen B and Xie X: miR-629-3p may serve as a novel biomarker and potential therapeutic target for lung metastases of triple-negative breast cancer. Breast Cancer Res. 19:722017. View Article : Google Scholar : PubMed/NCBI

18 

Zhang X, Luo P, Jing W, Zhou H, Liang C and Tu J: circSMAD2 inhibits the epithelial-mesenchymal transition by targeting miR-629 in hepatocellular carcinoma. Onco Targets Ther. 11:2853–2863. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Zheng YQ, Bai YF, Yang S, Cui YR, Wang YP and Hu WL: MircoRNA-629 promotes proliferation, invasion and migration of nasopharyngeal carcinoma through targeting PDCD4. Eur Rev Med Pharmacol Sci. 23:207–216. 2019.PubMed/NCBI

20 

Phuah NH, Azmi MN, Awang K and Nagoor NH: Suppression of microRNA-629 enhances sensitivity of cervical cancer cells to 1′S-1′-acetoxychavicol acetate via regulating RSU1. Onco Targets Ther. 10:1695–1705. 2017. View Article : Google Scholar :

21 

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

22 

Peröbner I, Karow M, Jochum M and Neth P: LRP6 mediates Wnt/β-catenin signaling and regulates adipogenic differentiation in human mesenchymal stem cells. Int J Biochem Cell Biol. 44:1970–1982. 2012. View Article : Google Scholar

23 

Lemieux E, Cagnol S, Beaudry K, Carrier J and Rivard N: Oncogenic KRAS signalling promotes the Wnt/β-catenin pathway through LRP6 in colorectal cancer. Oncogene. 34:4914–4927. 2015. View Article : Google Scholar

24 

Yang X, Wang L, Wang Q, Li L, Fu Y and Sun J: MiR-183 inhibits osteosarcoma cell growth and invasion by regulating LRP6-Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun. 496:1197–1203. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Shirafkan N, Mansoori B, Mohammadi A, Shomali N, Ghasbi M and Baradaran B: MicroRNAs as novel biomarkers for colorectal cancer: New outlooks. Biomed Pharmacother. 97:1319–1330. 2018. View Article : Google Scholar

26 

Masuda T, Hayashi N, Kuroda Y, Ito S, Eguchi H and Mimori K: MicroRNAs as biomarkers in colorectal cancer. Cancers (Basel). 9:pii: E124. 2017. View Article : Google Scholar

27 

Sarvizadeh M, Malekshahi ZV, Razi E, Sharifi H, Moussavi N and Taghizadeh M: MicroRNA: A new player in response to therapy for colorectal cancer. J Cell Physiol. 234:8533–8540. 2019. View Article : Google Scholar

28 

Balacescu O, Sur D, Cainap C, Visan S, Cruceriu D, Manzat-Saplacan R, Muresan MS, Balacescu L, Lisencu C and Irimie A: The impact of miRNA in colorectal cancer progression and its liver metastases. Int J Mol Sci. 19:pii: E3711. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Shao L, Shen Z, Qian H, Zhou S and Chen Y: Knockdown of miR-629 inhibits ovarian cancer malignant behaviors by targeting testis-specific Y-like protein 5. DNA Cell Biol. 36:1108–1116. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Jingushi K, Ueda Y, Kitae K, Hase H, Egawa H, Ohshio I, Kawakami R, Kashiwagi Y, Tsukada Y, Kobayashi T, et al: miR-629 Targets TRIM33 to promote TGFβ/Smad signaling and metastatic phenotypes in ccRCC. Mol Cancer Res. 13:565–574. 2015. View Article : Google Scholar

31 

Yan H, Li Q, Wu J, Hu W, Jiang J, Shi L, Yang X, Zhu D, Ji M and Wu C: MiR-629 promotes human pancreatic cancer progression by targeting FOXO3. Cell Death Dis. 8:e31542017. View Article : Google Scholar : PubMed/NCBI

32 

Liu CC, Prior J, Piwnica-Worms D and Bu G: LRP6 overexpression defines a class of breast cancer subtype and is a target for therapy. Proc Natl Acad Sci USA. 107:5136–5141. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Yuan Y, Xie X, Jiang Y, Wei Z, Wang P, Chen F, Li X, Sun C, Zhao H, Zeng X, et al: LRP6 is identified as a potential prognostic marker for oral squamous cell carcinoma via MALDI-IMS. Cell Death Dis. 8:e30352017. View Article : Google Scholar : PubMed/NCBI

34 

Wen Q, Zhao J, Bai L, Wang T, Zhang H and Ma Q: miR-126 inhibits papillary thyroid carcinoma growth by targeting LRP6. Oncol Rep. 34:2202–2210. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Du C, Lv Z, Cao L, Ding C, Gyabaah OA, Xie H, Zhou L, Wu J and Zheng S: MiR-126-3p suppresses tumor metastasis and angiogenesis of hepatocellular carcinoma by targeting LRP6 and PIK3R2. J Transl Med. 12:2592014. View Article : Google Scholar : PubMed/NCBI

36 

Bhanot P, Brink M, Samos CH, Hsieh JC, Wang Y, Macke JP, Andrew D, Nathans J and Nusse R: A new member of the frizzled family from Drosophila functions as a Wingless receptor. Nature. 382:225–230. 1996. View Article : Google Scholar : PubMed/NCBI

37 

Yang C, Yao C, Tian R, Zhu Z, Zhao L, Li P, Chen H, Huang Y, Zhi E, Gong Y, et al: miR-202-3p regulates sertoli cell proliferation, synthesis function, and apoptosis by targeting LRP6 and Cyclin D1 of Wnt/β-catenin signaling. Mol Ther Nucleic Acids. 14:1–19. 2019. View Article : Google Scholar

38 

Rismani E, Fazeli MS, Mahmoodzadeh H, Movassagh A, Azami S, Karimipoor M and Teimoori-Toolabi L: Pattern of LRP6 gene expression in tumoral tissues of colorectal cancer. Cancer Biomark. 19:151–159. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Yao Q, An Y, Hou W, Cao YN, Yao MF, Ma NN, Hou L, Zhang H, Liu HJ and Zhang B: LRP6 promotes invasion and metastasis of colorectal cancer through cytoskeleton dynamics. Oncotarget. 8:109632–109645. 2017. View Article : Google Scholar

Related Articles

Journal Cover

September-2019
Volume 44 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yan G, Li C, Zhao Y, Yue M and Wang L: Downregulation of microRNA‑629‑5p in colorectal cancer and prevention of the malignant phenotype by direct targeting of low‑density lipoprotein receptor‑related protein 6 Retraction in /10.3892/ijmm.2021.5057. Int J Mol Med 44: 1139-1150, 2019
APA
Yan, G., Li, C., Zhao, Y., Yue, M., & Wang, L. (2019). Downregulation of microRNA‑629‑5p in colorectal cancer and prevention of the malignant phenotype by direct targeting of low‑density lipoprotein receptor‑related protein 6 Retraction in /10.3892/ijmm.2021.5057. International Journal of Molecular Medicine, 44, 1139-1150. https://doi.org/10.3892/ijmm.2019.4245
MLA
Yan, G., Li, C., Zhao, Y., Yue, M., Wang, L."Downregulation of microRNA‑629‑5p in colorectal cancer and prevention of the malignant phenotype by direct targeting of low‑density lipoprotein receptor‑related protein 6 Retraction in /10.3892/ijmm.2021.5057". International Journal of Molecular Medicine 44.3 (2019): 1139-1150.
Chicago
Yan, G., Li, C., Zhao, Y., Yue, M., Wang, L."Downregulation of microRNA‑629‑5p in colorectal cancer and prevention of the malignant phenotype by direct targeting of low‑density lipoprotein receptor‑related protein 6 Retraction in /10.3892/ijmm.2021.5057". International Journal of Molecular Medicine 44, no. 3 (2019): 1139-1150. https://doi.org/10.3892/ijmm.2019.4245