MicroRNA-511 inhibits malignant behaviors of breast cancer by directly targeting SOX9 and regulating the PI3K/Akt pathway

Retraction in: /10.3892/ijo.2022.5413

  • Authors:
    • Yuying Zhao
    • Weifeng Pang
    • Ning Yang
    • Ling Hao
    • Lei Wang
  • View Affiliations

  • Published online on: September 27, 2018     https://doi.org/10.3892/ijo.2018.4576
  • Pages: 2715-2726
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Numerous studies have revealed that a subset of microRNAs (miRNAs) is aberrantly expressed in breast cancer. The dysregulation of miRNAs is involved in the tumorigenesis and progression of breast cancer due to their negative regulation of downstream target genes. Therefore, the identification of deregulated miRNAs in breast cancer may provide important insights into the diagnosis and treatment of patients with this disease. miRNA‑511 (miR‑511) has been identified to be deregulated in diverse human cancer types; however, neither the expression status nor the detailed roles of miR‑511 in breast cancer have been clarified. Thus, it was aimed to determine the expression of miR‑511 in breast cancer, examine the role in malignant progression and explore its downstream targets. The results of the present study revealed that the expression of miR‑511 was downregulated in breast cancer tissues and cell lines. Decreased expression of miR‑511 was significantly associated with lymph node metastasis and tumor stage in patients with breast cancer. Functional analyses revealed that restoring miR‑511 expression suppressed breast cancer cell proliferation and colony formation, promoted apoptosis and reduced metastasis in vitro, while it attenuated tumor growth in vivo. Additionally, it was revealed that SRY‑box 9 (SOX9) was a direct target gene of miR‑511 in breast cancer cells. SOX9 was upregulated in breast cancer tissues and its expression was inversely correlated with that of miR‑511. Furthermore, SOX9 inhibition simulated the tumor‑suppressive roles of miR‑511 overexpression in breast cancer cells, while SOX9 reintroduction partially rescued these effects of miR‑511. Notably, the upregulation of miR‑511 targeted SOX9 to deactivate the PI3K/Akt signaling in breast cancer in vitro and in vivo. In conclusion, miR‑511 was downregulated in breast cancer, and impeded its malignant progression by directly targeting SOX9 and regulating the PI3K/Akt pathway. Thus, miR‑511 is a potential therapeutic target in breast cancer.

Introduction

Breast cancer is the most common malignant tumor and the second leading cause of cancer-associated death among women worldwide (1). In 2012, ~1.7 million diagnoses and 521,900 mortalities accounting for 25% of cancer cases and 15% of cancer-associated mortalities, respectively, were reported (2). Multiple factors, including obesity, a high-fat diet, hormone replacement therapy, early menarche and late menopause, have been identified to be involved in breast cancer onset and development (3); however, the detailed molecular mechanisms remain unclear. At present, the major treatments for breast cancer patients are surgery in combination with hormonotherapy, chemoradiotherapy and/ or biological therapy (4). Despite significant progress in the early detection and therapy of breast cancer, the therapeutic outcomes of patients diagnosed at an advanced stage remain unsatisfactory (5,6). Recurrence and metastasis are the primary causes of poor prognosis for patients with breast cancer (7). Therefore, characterization of the molecular footprint of breast cancer is urgently required to identify promising therapeutic targets for the management of patients with this disease.

MicroRNAs (miRNAs/miRs) are a group of endogenous, non-coding, short RNA molecules that are 19-24 nucleotides in length (8). Currently, >2,000 mature miRNAs have been identified in the human genome, and these miRNAs regulate gene expression by directly binding to a ‘seed sequence’ within the 3′-untranslated region (UTR) of each target gene, resulting in translational suppression and/or mRNA destabilization (9). It has been demonstrated that a single miRNA can modulate numerous target genes, and >50% of known miRNAs are located at cancer-associated chromosomal regions, suggesting that miRNAs may have crucial roles in tumorigenesis and tumor development (10). Recent studies have revealed the aberrant expression of various miRNAs in breast cancer, and their deregulation has been implicated in breast cancer genesis and development (11-13). Deregulated miRNAs can have oncogenic or tumor-suppressive roles in breast cancer and control biological behaviors, including cell proliferation, cell cycle, apoptosis, invasion, metastasis, epithelial-mesenchymal transition and angiogenesis (14-16). Therefore, further investigations of the biological functions of miRNAs may help us to better understand the mechanisms underlying breast cancer formation/progression and facilitate the development of attractive therapeutic approaches.

miR-511 has been identified to be deregulated in diverse human cancer types, including colorectal cancer (17), gastric cancer (18), lung adenocarcinoma (19) and hepatocellular carcinoma (20). miR-511 has been revealed as an important gene expression regulator in breast cancer (21). However, neither the expression status nor the detailed roles of miR-511 in breast cancer have been clarified. In this study, it was aimed to determine the expression of miR-511 in breast cancer, test its role in malignant progression and explore downstream targets. SRY-box 9 (SOX9) was predicted as a potential target of miR-511 and was chosen for further investigation as this gene was previously reported to be upregulated in breast cancer and implicated in breast carcinogenesis and development (22-28). The findings indicate that miR-511 may be a molecular target for the treatment of patients with breast cancer.

Materials and methods

Tissue specimens

This study was approved by the Ethics Committee of The Fourth Affiliated Hospital of Harbin Medical University (Harbin, China) and each patient enrolled provided written informed consent. Pairs of breast cancer tissues and adjacent normal breast tissues (n=51) were obtained at The Fourth Affiliated Hospital of Harbin Medical University between May 2014 and March 2017. Patients had been treated with preoperative adjuvant chemotherapy, radiotherapy or other therapy were excluded from this research. All tissue specimens were snap-frozen in liquid nitrogen and stored at −80°C prior to RNA isolation.

Cell lines

A human breast epithelial cell line (MCF-10A) and four human breast cancer cell lines (MCF-7, BT-474, MDA-MB-231 and SKBR3) were purchased from Shanghai Institute of Biochemistry and Cell Biology (Shanghai, China). All the cell lines were cultured at 37°C in a 5% CO2 incubator using Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin mixture (all from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA).

Oligonucleotide, small interfering RNA (siRNA) and plasmid transfection

miR-511 mimics and negative control miRNA mimics (miR-NC) were obtained from Shanghai GenePharma Co., Ltd. (Shanghai, China). The miR-511 mimics sequence was 5′-GUGUCUUUUGCUCUGCAGUCA-3′ and the miR-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. An siRNA targeting SOX9 and negative control siRNA (NC siRNA) were purchased from Guangzhou RiboBio (Guangzhou, China). The SOX9 siRNA sequence was 5′-AGCAAGTCCGCGAGCC AGTAC-3′ and the NC siRNA sequence was 5′-UUCUCCGAA CGUGUCACGUTT-3′. The full-length SOX9 lacking the 3′-UTR sequence was chemically synthesized by the Chinese Academy of Sciences (Changchun, China) and inserted into the pcDNA3.1 vector to produce pcDNA3.1-SOX9 plasmid.

Cells were inoculated into 6-well plates at 60-70% confluence. Following overnight incubation, cells were transfected with miR-511 mimics (100 pmol), miR-NC (100 pmol), SOX9 siRNA (100 pmol), NC siRNA (100 pmol), pcDNA3.1-SOX9 (4 µg) or empty pcDNA3.1 vector (4 µg) using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) in accordance with the manufacturer’s protocol. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was utilized to determine the transfection efficiency of miR-511 mimics. The transfection efficiencies of SOX9 siRNA and pcDNA3.1-SOX9 were evaluated by western blot analysis after 72 h of incubation.

RNA extraction and RT-qPCR

TRIzol® reagent (Thermo Fisher Scientific, Inc.) was used to isolate total RNA from tissue specimens or cultured cells. For the quantification of miR-511 expression, first-strand cDNA synthesis was performed using the miScript Reverse Transcription kit (Qiagen GmbH, Hilden, Germany) following the manufacturer’s protocol. Subsequently, qPCR was performed using the miScript SYBR®-Green PCR kit (Qiagen GmbH). The thermocycling conditions for qPCR were as follows: 95°C for 15 min, followed by 45 cycles of 94°C for 15 sec, 55°C for 30 sec and 70°C for 30 sec. miR-511 expression was normalized to that of U6 small nuclear RNA. To analyze the mRNA expression of SOX9, RT was conducted using the PrimeScript™ RT reagent kit, and the synthesized cDNA was then subjected to qPCR using a SYBR® Premix Ex Taq™ kit (both from Takara Biotechnology Co., Ltd., Dalian, China). qPCR for mRNA was performed with the following cycling conditions: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. GAPDH was used as an internal reference for the mRNA expression of SOX9. The primers were designed as follows: miR-511, 5′-GTGTCTTTTGCTCTGCAGTC-3′ (forward) and 5′-AACATGTACAGTCCATGGATG-3′ (reverse); U6, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ (forward) and 5′-CGCTTCACGAATTTGCGTGTCAT-3′ (reverse); SOX9, 5′-CTGGGAACAACCCGTCTA-3′ (forward) and 5′-GGGTAA TGCGCTTGGATA-3′ (reverse); and GAPDH, 5′-CGGAGTCA ACGGATTTGGTCGTAT-3′ (forward) and 5′-AGCCTTCTCC ATGGTGGTGAAGAC-3′ (reverse). Relative gene expression was analyzed using the 2−ΔΔCq method (29).

Cell counting kit 8 (CCK-8) assay

Transfected cells were collected at 24 h post-transfection and seeded into 96-well plates with an initial density of 3×103 cells per well. Cells were then incubated at 37°C under 5% CO2 for 0, 24, 48 or 72 h. At each time point, the CCK-8 assay was performed by adding 10 µl CCK-8 solution (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) into each well. After another 2 h incubation with CCK-8 reagent at 37°C, the optical density was detected at 450 nm wavelength using a SpectraMax® microplate spectrophotometer (Molecular Devices, LLC, Sunnyvale, CA, USA).

Colony formation assay

The colony formation assay was applied to assess the ability of breast cancer cells to form colonies. Following 24 h incubation, transfected cells were harvested, and single cells were plated into 6-well plates with a density of 1,000 cells per well. Cells were maintained at 37°C supplied with 5% CO2 for 2 weeks. On day 15, cells were fixed with 4% paraformaldehyde, stained with methyl violet (Beyotime Institute of Biotechnology, Haimen, China) and washed with PBS. The number of colonies was counted under an inverted microscope (IX83; Olympus, Tokyo, Japan).

Flow cytometry analysis of cell apoptosis

An Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit (Biolegend, Inc., San Diego, CA, USA) was used to measure the apoptosis rate of breast cancer cells. Cells were collected at 48 h after transfection, washed three times with cold PBS and resuspended in 100 µl binding buffer. Subsequently, cells were double-stained at 37°C with 5 µl Annexin V-FITC and 5 µl propidium iodide for 30 min in the dark. The percentage of apoptotic cells was measured using a flow cytometer (FACScan™; BD Biosciences, Franklin Lakes, NJ, USA). The data were processed using CellQuest™ version 5.1 software (BD Biosciences).

Migration and invasion assays

The migratory and invasive abilities of breast cancer cells were investigated using 24-well Transwell® inserts (pore size, 8 µm) coated without or with Matrigel® (both from BD Biosciences). In brief, a single-cell suspensions of the transfected cells was prepared and 1×105 cells in 200 µl FBS-free DMEM were added to the upper compartment of Transwell® inserts. DMEM supplemented with 10% FBS was added into the lower compartment to serve as a chemo-attractant. After 24 h of incubation at 37°C with 5% CO2, the cells were fixed with 4% paraformaldehyde at room temperature for 20 min and stained with 0.05% crystal violet (Beyotime Institute of Biotechnology) at room temperature for 20 min. The non-migrated or non-invading cells were carefully removed using a cotton swab, while the number of migrated or invaded cells was counted in five randomly chosen visual fields under an inverted microscope.

Xenograft tumor experiment

All animal experiments were performed in accordance with the Guide for the Care and Use of Laboratory Animals and approved by the Ethics Review Committee of The Fourth Affiliated Hospital of Harbin Medical University. Cells were transfected with miR-511 mimics or miR-NC and incubated at 37°C under 5% CO2 for 24 h. Transfected cells were harvested after 24 h of incubation and then subcutaneously administered into the hind flanks of 4-week-old BALB/c nude mice (1×106 cells per mouse). Tumor width and length were detected every 4 days for 4 weeks. All nude mice were sacrificed 4 weeks after tumor implantation and the tumor xenografts were excised and weighed. The tumor volumes were calculated using the equation: Tumor volume = 0.5 × tumor length × tumor width.

Bioinformatics analysis

TargetScan 7.1 (http://www.targetscan.org/) and MiRanda (http://www.microrna.org/) were used to predict the potential targets of miR-511. SOX9 was identified as a candidate target gene of miR-511.

Plasmid construction and luciferase reporter assay

The 3′-UTR of SOX9 containing the wild-type or mutant miR-511 binding site was amplified by Shanghai GenePharma and inserted into the pMIR-REPORT plasmid (Promega Corporation, Madison, WI, USA). Cells were plated into 24-well plates with a density of 1×105 cells/well and were co-transfected with the luciferase reporter plasmid (0.2 µg) together with miR-511 mimics (50 pmol) or miR-NC (50 pmol) using Lipofectamine® 2000. Luciferase activity was determined 48 h after transfection using the dual-luciferase reporter assay system (Promega Corporation). Renilla luciferase activity served as an internal reference for Firefly luciferase activity.

Western blot analysis

Cells or tissues were lysed in ice-cold radioimmunoprecipitation buffer (Cell Signaling Technology, Inc., Danvers, MA, USA) to extract total protein. The concentration of the isolated total protein was detected using a bicinchoninic acid assay kit (Beyotime Institute of Biotechnology). Equal amounts (30 µg) of protein were separated by SDS-PAGE on a 10% polyacrylamide gel. The separated proteins were then electronically transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). Following blocking at room temperature in 5% fat-free milk powder for 2 h, the membranes were incubated overnight at 4°C with primary antibodies against SOX9 (cat. no. ab185966; 1:1,000 dilution), phosphor (p)-phosphoinositide 3-kinase (PI3K; cat. no. ab182651; 1:1,000 dilution), PI3K (cat. no. ab191606; 1:1,000 dilution) (all from Abcam, Cambridge, UK), p-Akt (cat. no. sc-81433; 1:1,000 dilution), Akt (cat. no. sc-56878; 1:1,000 dilution) (both from Santa Cruz Biotechnology, Inc., Dallas, TX, USA) or GAPDH (cat. no. ab181603; 1:1,000 dilution; Abcam). Following three washes with Tris-buffered saline containing 0.1% Tween®-20, the membranes were further incubated with the goat anti-rabbit (cat. no. ab6721; Abcam; used for SOX9, p-PI3K, PI3K and GAPDH primary antibodies) or goat anti-mouse (cat. no. ab205719; Abcam; used for p-Akt and Akt primary antibodies) corresponding horseradish peroxidase-conjugated secondary antibody (both 1:5,000 dilution) at room temperature for 2 h and visualized using an enhanced chemiluminescence kit (EMD Millipore). GAPDH was used as a loading control. Protein expression was quantified using Quantity One software version 4.62 (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Statistical analysis

SPSS software (version 19.0; IBM Corp., Armonk, NY, USA) was used to perform all statistical analyses. Data are presented as the mean ± standard deviation from three independent experiments. The statistical differences between groups were analyzed using Student’s t-test or one-way analysis of variance combined with multiple comparisons using the Student-Newman-Keuls post-hoc test. The χ2 test was used to assess the association between miR-511 expression and the clinicopathological features of patients with breast cancer. Spearman’s correlation analysis was employed to identify the association between the expression levels of miR-511 and SOX9 mRNA in breast cancer tissues. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-511 is downregulated in breast cancer tissues and cell lines

To determine the expression of miR-511 in breast cancer, miR-511 expression was detected using RT-qPCR in 51 pairs of breast cancer tissue and adjacent normal breast tissue. miR-511 was significantly reduced in breast cancer tissues compared with adjacent normal breast tissues (P<0.05; Fig. 1A). To further confirm this observation, miR-511 expression was analyzed in four human breast cancer cell lines (MCF-7, BT-474, MDA-MB-231 and SKBR3) and a human breast epithelial cell line (MCF-10A). The expression of miR-511 was significantly lower in all four breast cancer cell lines compared with MCF-10A cells (P<0.05; Fig. 1B).

miR-511 expression is associated with the clinical characteristics of patients with breast cancer

To characterize the clinical characteristics of miR-511, all patients with breast cancer were divided into either the miR-511 low expression group (n=26) or miR-511 high expression group (n=25) based on the median value of miR-511. The results of the statistical analysis indicated that decreased miR-511 expression was associated with lymph node metastasis (P=0.001) and tumor stage (P=0.017; Table I). However, no significant associations were observed between miR-511 expression and other clinicopathological characteristics, including age, tumor diameter and histology grade (all P>0.05). The results suggest that the downregulation of miR-511 may be involved in breast cancer initiation and progression.

Table I

Association between miR-511 expression in tumor tissue and clinical characteristics of patients with breast cancer.

Table I

Association between miR-511 expression in tumor tissue and clinical characteristics of patients with breast cancer.

Clinical characteristicsmiR-511 expression
P-value
LowHigh
Age (years)0.447
 <50118
 ≥501517
Tumor diameter (cm)0.329
 <2811
 ≥21814
Histology grade0.461
 I-II129
 III1416
Lymph node metastasis0.001a
 Negative920
 Positive175
Tumor stage0.017a
 I-II715
 III1910

a P<0.05. The median value of miR-511 in tumor tissues was used to divide the samples in high and low expression groups. miR, microRNA.

miR-511 upregulation inhibits proliferation, promotes apoptosis, and decreases metastasis in breast cancer cells in vitro

To gain insight into the detailed roles of miR-511 in breast cancer, miR-511 mimics were transfected into MCF-7 and MDA-MB-231 cells to increase endogenous miR-511 expression. The data obtained by RT-qPCR analysis revealed that miR-511 was overexpressed in miR-511 mimics-transfected MCF-7 and MDA-MB-231 cells relative to that in cells transfected with miR-NC (P<0.05; Fig. 2A). The CCK-8 assay was performed to evaluate the effect of miR-511 upregulation on the proliferative ability of breast cancer cells. Ectopic miR-511 expression caused an obvious reduction in proliferation in MCF-7 and MDA-MB-231 cells (P<0.05; Fig. 2B). Colony formation assay was also used to confirm the regulatory effect of miR-511 on breast cancer cell proliferation. The results indicated that miR-511 overexpression significantly decreased the colony formation abilities of MCF-7 and MDA-MB-231 cells (P<0.05; Fig. 2C). Apoptosis is associated with the alterations in cell proliferation, suggesting that a high apoptosis rate mat cause suppression of cell proliferation. Thus, the apoptosis rate of MCF-7 and MDA-MB-231 cells transfected with miR-511 mimics or miR-NC was determined. As displayed in Fig. 2D, enforced miR-511 expression increased the rate of apoptosis in MCF-7 and MDA-MB-231 cells in comparison with the miR-NC groups (P<0.05). Furthermore, migration and invasion assays were performed to determine whether miR-511 was involved in the regulation of breast cancer cell metastasis. miR-511-overexpressing MCF-7 and MDA-MB-231 cells exhibited decreased migratory (P<0.05; Fig. 2E) and invasive (P<0.05: Fig. 2F) capacities as compared with the cells transfected with miR-NC. The findings suggest that miR-511 upregulation may inhibit the aggressiveness of breast cancer cells.

SOX9 is a direct target of miR-511 in breast cancer cells

To elucidate the mechanism by which miR-511 may act as a tumor suppressor in breast cancer, bioinformatics analysis was performed to search for putative targets of miR-511. SOX9 was predicted to be a major target of miR-511, as two complementary binding sites of miR-511 were identified in the 3′-UTR of the SOX9 gene (Fig. 3A). SOX9 was chosen for further experimental confirmation as this gene was previously reported to be involved in the pathogenesis of breast cancer. A luciferase reporter assay was performed to determine whether miR-511 directly recognized and bound to the 3′-UTR of SOX9. The data revealed that miR-511 upregulation decreased the luciferase activity of the plasmid harboring the wild-type (1 and 2) 3′-UTR of SOX9 in MCF-7 and MDA-MB-231 cells (P<0.05; Fig. 3B), but did not affect the activity of the mutant (1 and 2) 3′-UTR plasmid. To further confirm that SOX9 is a target of miR-511, RT-qPCR and western blot analysis were performed to detect SOX9 mRNA and protein expression in MCF-7 and MDA-MB-231 cells with overexpressed miR-511. The mRNA (P<0.05; Fig. 3C) and protein (P<0.05; Fig. 3D) levels of SOX9 were lowered in MCF-7 and MDA-MB-231 cells transfected with miR-511 mimics. Collectively, these findings suggest that SOX9 is a direct target gene of miR-511 in breast cancer cells.

SOX9 expression is negatively correlated with miR-511 expression in breast cancer tissues

To further illustrate the relationship between miR-511 and SOX9 in breast cancer, RT-qPCR was performed to measure SOX9 expression in breast cancer tissues and adjacent normal breast tissues. Compared with adjacent normal breast tissues, the level of SOX9 mRNA (P<0.05; Fig. 4A) was strikingly upregulated in breast cancer tissues. Then, Spearman’s correlation analysis was performed to analyze the association between miR-511 and SOX9 expression in breast cancer tissues, which revealed a statistically significant negative correlation between the miR-511 and SOX9 mRNA levels in these breast cancer tissues (r=−0.5683, P<0.0001; Fig. 4B). These results further supported the conclusion that SOX9 is a downstream target of miR-511 in breast cancer.

SOX9 knockdown imitates the tumor-suppressor activity of miR-511 upregulation in breast cancer cells

SOX9 was validated as a direct target gene of miR-511 in breast cancer cells; thus, it was hypothesized that SOX9 knockdown may mimic the tumor-suppressor role of miR-511 overexpression in breast cancer cells. To test this hypothesis, SOX9 siRNA was applied to knock down endogenous SOX9 expression in MCF-7 and MDA-MB-231 cells (P<0.05; Fig. 5A). Subsequent functional assays demonstrated that SOX9 inhibition impeded proliferation (P<0.05; Fig. 5B) and colony formation (P<0.05; Fig. 5C), increased apoptosis (P<0.05; Fig. 5D) and prohibited migration (P<0.05; Fig. 5E) and invasion (P<0.05; Fig. 5F) of MCF-7 and MDA-MB-231 cells. Thus, the downregulation of SOX9 had functional effects that were similar to those of miR-511 upregulation in breast cancer cells, suggesting that SOX9 is a functional target of miR-511 in breast cancer cells.

Restored SOX9 expression partially relieves the miR-511-mediated suppression of breast cancer progression in vitro

To further demonstrate whether SOX9 inhibition was essential for the tumor-suppressor activity of miR-511 in breast cancer, a SOX9 overexpression plasmid lacking the 3′-UTR (pcDNA3.1-SOX9) was transfected into miR-511-overexpressing MCF-7 and MDA-MB-231 cells. Western blot analysis indicated that the downregulation of SOX9 protein caused by miR-511 upregulation was re-established in MCF-7 and MDA-MB-231 cells following co-transfection with pcDNA3.1-SOX9 (P<0.05; Fig. 6A). CCK-8 and colony formation assays revealed that SOX9 reintroduction re-established the miR-511-induced inhibition of MCF-7 and MDA-MB-231 cell proliferation (P<0.05; Fig. 6B) and colony formation (P<0.05; Fig. 6C). Additionally, the miR-511 mimics-mediated promotion of cell apoptosis was partially reversed by SOX9 restoration in MCF-7 and MDA-MB-231 cells (P<0.05; Fig. 6D). Furthermore, the restoration of SOX9 overexpression blunted the inhibitory effects of miR-511 overexpression on the migratory (P<0.05; Fig. 6E) and invasive (P<0.05; Fig. 6F) abilities of MCF-7 and MDA-MB-231 cells. Together, these results provided sufficient evidence to demonstrate that miR-511 may inhibit the development of breast cancer cells, at least partially by suppressing SOX9 expression.

miR-511 inhibits the activation of the PI3K/Akt signaling pathway in breast cancer cells by targeting SOX9

SOX9 participates in the regulation of the PI3K/Akt signaling pathway (30,31); thus, whether miR-511 inactivates PI3K/Akt signaling in breast cancer cells was examined. To test this hypothesis, western blot analysis was performed to detect the molecules associated with the PI3K/Akt pathway in MCF-7 and MDA-MB-231 cells following co-transfection with miR-511 mimics and pcDNA3.1 or pcDNA3.1-SOX9. It was demonstrated that enforced miR-511 expression suppressed the levels of the p-PI3K and p-Akt phosphoproteins in MCF-7 and MDA-MB-231 cells; however, miR-511 overexpression did not affect the total levels of the PI3K and Akt proteins. Additionally, co-transfection with pcDNA3.1-SOX9 restored the miR-511 overexpression-inhibited levels of p-PI3K and p-Akt in these cell lines (Fig. 7). These findings suggest that miR-511 targets SOX9 to inactivate the PI3K/Akt pathway in breast cancer cells.

miR-511 hinders breast cancer tumor growth in vivo

A xenograft tumor experiment was performed to assess whether miR-511 contributes to the tumorigenesis of breast cancer cells in vivo. MDA-MB-231 cells were transfected with miR-511 mimics or miR-NC and were subcutaneously administered into the hind flanks of nude mice. In order to meet ethical requirements, all the nude mice were sacrificed before the tumor xenografts reached 2 cm. The maximum tumor burden was 1.8 cm. Consistent with the in vitro results, miR-511-overexpressing xenografts exhibited a lower tumor volume (P<0.05; Fig. 8A and B) and tumor weight (P<0.05; Fig. 8C) compared with miR-NC expressing tumors. In addition, the data obtained by RT-qPCR analysis revealed that miR-511 was specifically upregulated in the xenografts of the miR-511 mimics-overexpressing group compared the miR-NC-transfected group (P<0.05; Fig. 8D). Western blot analysis demonstrated that the xenografts of the miR-511-overexpressing group expressed lower levels of SOX9, p-PI3K, and p-Akt than the xenografts of the miR-NC-transfected group (Fig. 8E). Together, these results suggest that miR-511 targets SOX9 to inhibit breast cancer tumor growth in vivo via deactivation of the PI3K/Akt pathway.

Discussion

Numerous studies have revealed that an miRNA subset is aberrantly expressed in nearly all human cancer types, including breast cancer (32-34). The dysregulation of miRNAs is involved in the tumorigenesis and progression of breast cancer due to their negative regulation of downstream target genes (35-37). It is generally accepted that miRNAs may have tumor-suppressive or oncogenic roles in breast cancer and participate in the regulation of numerous cellular biological processes (16,38,39). Notably, development of effective computational models for the prediction of potential breast cancer-associated miRNAs has been a research hot spot in recent years (40-45). Therefore, the identification of deregulated miRNAs in breast cancer may provide important insights into the diagnosis and treatment of patients with this disease. miR-511 has been identified as an important gene expression regulator in breast cancer (21). In this study, it was aimed to measure miR-511 expression in breast cancer, analyze the clinical characteristics of miR-511 in breast cancer, examine the detailed roles of miR-511 in breast cancer progression and determine the underlying mechanisms. To the best of knowledge, this is the first report to demonstrate that miR-511 is downregulated in breast cancer and that it inhibits malignant behaviors by directly targeting SOX9 and regulating the PI3K/Akt pathway.

To elucidate the clinical value of miR-511, RT-qPCR was performed to detect expression in breast cancer tissues and cell lines. It was observed that miR-511 was significantly downregulated in breast cancer tissues and cell lines, and the downregulation of miR-511 was associated with lymph node metastasis and tumor stage. This finding is in agreement with previous studies (17-19) on the expression pattern of miR-511 in several human cancer subtypes. For example, the expression level of miR-511 was lower in colorectal cancer tissues and cell lines than in adjacent non-neoplastic tissues and normal human colon epithelium cell lines, respectively (17). miR-511 expression was also reported to be decreased in gastric cancer (18), and lung adenocarcinoma (19). By contrast, miR-511 was upregulated in hepatocellular carcinoma (20). These inconsistent findings indicate that the expression status of miR-511 exhibits tissue specificity. Thus, this miRNA could potentially be a biomarker for the diagnosis of patients with specific cancer subtypes.

The data from functional assays further demonstrated that miR-511 upregulation inhibits breast cancer cell proliferation and colony formation, promotes apoptosis, impedes cell migration and invasion and decreases in vivo tumor growth. Aberrant expression of miR-511 has previously been reported to have a role in multiple types of human cancer. For instance, miR-511 overexpression prohibited the proliferative and invasive abilities of colorectal cancer cells in vitro (17). In gastric cancer, the restoration of miR-511 expression suppressed cell growth and induced cell cycle arrest (18). miR-511 was also validated as a tumor suppressor affecting cell proliferation, apoptosis, cell cycle and radioresistance in lung adenocarcinoma (19,46). However, miR-511 was identified as an oncogene in hepatocellular carcinoma (20). These conflicting findings imply that the functional roles of miR-511 in malignancy have tissue specificity, suggesting that miR-511 may be developed as a valuable therapeutic target for treating patients with these specific cancer types.

Previous studies have validated multiple target genes of miR-511, including heparin binding growth factor (17) in colorectal cancer, tripartite motif containing 24 in gastric cancer (18), tribbles pseudokinase 2 (19) and BCL2 associated X apoptosis regulator (46) in lung adenocarcinoma, and BTG anti-proliferation factor 1 in hepatocellular carcinoma (20). Validation of the direct targets of miR-511 is important for understanding its roles in the development of breast cancer and may assist the development of effective therapeutic techniques. Thus, the molecular mechanisms underlying the tumor-suppressive activity of miR-511 in breast cancer were examined. SOX9, a member of the sex-determining region Y box family, was demonstrated to be a direct target of miR-511 in breast cancer. SOX9 has been reported to be upregulated in a number of human cancers, including prostate cancer (47), non-small cell lung cancer (48) and colorectal cancer (49). SOX9 was also highly expressed in breast cancer and reported to be significantly associated with estrogen receptor expression and tumor grade (22). Breast cancer patients with high expression of SOX9 exhibited shorter overall survival than those with low expression (22,23). Numerous other studies have supported the importance of SOX9 in breast cancer occurrence and development through the regulation of multiple biological behaviors (24-28). In the current study, it was demonstrated that miR-511 targets SOX9 to inhibit the malignant progression of breast cancer via the PI3K/ Akt pathway. Thus, SOX9 knockdown using miR-511-based targeted therapy may be a potential therapeutic strategy for patients with breast cancer in the future.

In summary, the current study demonstrated that miR-511 was downregulated in breast cancer tissues and cell lines. Functional experiments revealed that miR-511 overexpression inhibited the malignant progression of breast cancer in vitro and in vivo. Mechanistically, SOX9 was identified as a direct target of miR-511 in breast cancer cells. The present study provides novel insights into the mechanisms underlying breast cancer genesis and progression, and highlights a potential therapeutic target for breast cancer. However, rescue experiments were not performed to explore whether SOX9 expression can rescue the in vivo tumor growth inhibition induced by miR-511. In addition, a PI3K inhibitor was not used to illustrate whether the SOX9-induced rescue of miR-511 growth and metastasis inhibition could be blocked. Furthermore, the effect of miR-511 on the ERK pathway in breast cancer was not investigated. These are limitations of the study and these limitations will be resolved in the near future.

Funding

Not applicable.

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

YZ designed this research. WP and NY performed RT-qPCR, western blot analysis, CCK-8 assay, colony formation assay, flow cytometry analysis, and migration and invasion assays. LH and LW performed xenograft tumor experiment and luciferase reporter assay. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Research Ethics Committee of the Fourth Affiliated Hospital of Harbin Medical University, and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of the Fourth Affiliated Hospital of Harbin Medical University. Written informed consent was obtained from all patients for the use of their clinical tissues.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

DeSantis CE, Ma J, Goding Sauer A, Newman LA and Jemal A: Breast cancer statistics, 2017, racial disparity in mortality by state. CA Cancer J Clin. 67:439–448. 2017. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Veronesi U, Boyle P, Goldhirsch A, Orecchia R and Viale G: Breast cancer. Lancet. 365:1727–1741. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Nakamura S, Yagata H, Ohno S, Yamaguchi H, Iwata H, Tsunoda N, Ito Y, Tokudome N, Toi M, Kuroi K, et al: Multi-center study evaluating circulating tumor cells as a surrogate for response to treatment and overall survival in metastatic breast cancer. Breast Cancer. 17:199–204. 2010. View Article : Google Scholar

5 

Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature. 490:61–70. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Weigelt B, Peterse JL and van ‘t Veer LJ: Breast cancer metastasis: Markers and models. Nat Rev Cancer. 5:591–602. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Cardoso F, Costa A, Senkus E, Aapro M, André F, Barrios CH, Bergh J, Bhattacharyya G, Biganzoli L, Cardoso MJ, et al: 3rd ESO-ESMO International Consensus Guidelines for Advanced Breast Cancer (ABC 3). Ann Oncol. 28:16–33. 2017. View Article : Google Scholar : PubMed/NCBI

8 

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 :

9 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Ma XP, Zhang T, Peng B, Yu L and Jiang K: Association between microRNA polymorphisms and cancer risk based on the findings of 66 case-control studies. PLoS One. 8:e795842013. View Article : Google Scholar : PubMed/NCBI

11 

Singh T and Adams BD: The regulatory role of miRNAs on VDR in breast cancer. Transcription. 8:232–241. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Lü L, Mao X, Shi P, He B, Xu K, Zhang S and Wang J: MicroRNAs in the prognosis of triple-negative breast cancer: A systematic review and meta-analysis. Medicine (Baltimore). 96:e70852017. View Article : Google Scholar

13 

Evans-Knowell A, LaRue AC and Findlay VJ: MicroRNAs and Their Impact on Breast Cancer, the Tumor Microenvironment, and Disparities. Adv Cancer Res. 133:51–76. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Cao MQ, You AB, Zhu XD, Zhang W, Zhang YY, Zhang SZ, Zhang KW, Cai H, Shi WK, Li XL, et al: miR-182-5p promotes hepatocellular carcinoma progression by repressing FOXO3a. J Hematol Oncol. 11:122018. View Article : Google Scholar : PubMed/NCBI

15 

Hu TB, Chen HS, Cao MQ, Guo FD, Cheng XY, Han ZB and Li MQ: MicroRNA-421 inhibits caspase-10 expression and promotes breast cancer progression. Neoplasma. 65:49–54. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Lv ZD, Yang DX, Liu XP, Jin LY, Wang XG, Yang ZC, Liu D, Zhao JJ, Kong B, Li FN, et al: MiR-212-5p Suppresses the epithelial-mesenchymal transition in triple-negative breast cancer by targeting Prrx2. Cell Physiol Biochem. 44:1785–1795. 2017. View Article : Google Scholar : PubMed/NCBI

17 

He S, Wang G, Ni J, Zhuang J, Zhuang S, Wang G, Ye Y and Xia W: MicroRNA-511 inhibits cellular proliferation and invasion in colorectal cancer by directly targeting hepatoma-derived growth factor. Oncol Res, 2018 Oncol Res. Jan 10–2018.Epub ahead of print. View Article : Google Scholar

18 

Fang Z, Zhang L, Liao Q, Wang Y, Yu F, Feng M, Xiang X and Xiong J: Regulation of TRIM24 by miR-511 modulates cell proliferation in gastric cancer. J Exp Clin Cancer Res. 36:172017. View Article : Google Scholar : PubMed/NCBI

19 

Zhang C, Chi YL, Wang PY, Wang YQ, Zhang YX, Deng J, Lv CJ and Xie SY: miR-511 and miR-1297 inhibit human lung adenocarcinoma cell proliferation by targeting oncogene TRIB2. PLoS One. 7:e460902012. View Article : Google Scholar : PubMed/NCBI

20 

Zhang SQ, Yang Z, Cai XL, Zhao M, Sun MM, Li J, Feng GX, Feng JY, Ye LH, Niu JQ, et al: miR-511 promotes the proliferation of human hepatoma cells by targeting the 3′UTR of B cell translocation gene 1 (BTG1) mRNA. Acta Pharmacol Sin. 38:1161–1170. 2017. View Article : Google Scholar : PubMed/NCBI

21 

de Anda-Jáuregui G, Espinal-Enríquez J, Drago-García D and Hernández-Lemus E: Nonredundant, highly connected MicroRNAs control functionality in breast cancer networks. Int J Genomics. 2018:95853832018. View Article : Google Scholar : PubMed/NCBI

22 

Chakravarty G, Moroz K, Makridakis NM, Lloyd SA, Galvez SE, Canavello PR, Lacey MR, Agrawal K and Mondal D: Prognostic significance of cytoplasmic SOX9 in invasive ductal carcinoma and metastatic breast cancer. Exp Biol Med (Maywood). 236:145–155. 2011. View Article : Google Scholar

23 

Riemenschnitter C, Teleki I, Tischler V, Guo W and Varga Z: Stability and prognostic value of Slug, Sox9 and Sox10 expression in breast cancers treated with neoadjuvant chemotherapy. Springerplus. 2:6952013. View Article : Google Scholar

24 

Müller P, Crofts JD, Newman BS, Bridgewater LC, Lin CY, Gustafsson JA and Ström A: SOX9 mediates the retinoic acid-induced HES-1 gene expression in human breast cancer cells. Breast Cancer Res Treat. 120:317–326. 2010. View Article : Google Scholar

25 

Chakravarty G, Rider B and Mondal D: Cytoplasmic compartmentalization of SOX9 abrogates the growth arrest response of breast cancer cells that can be rescued by trichostatin A treatment. Cancer Biol Ther. 11:71–83. 2011. View Article : Google Scholar

26 

Wang H, He L, Ma F, Regan MM, Balk SP, Richardson AL and Yuan X: SOX9 regulates low density lipoprotein receptor-related protein 6 (LRP6) and T-cell factor 4 (TCF4) expression and Wnt/β-catenin activation in breast cancer. J Biol Chem. 288:6478–6487. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Fazilaty H, Gardaneh M, Akbari P, Zekri A and Behnam B: SLUG and SOX9 cooperatively regulate tumor initiating niche factors in breast cancer. Cancer Microenviron. 9:71–74. 2016. View Article : Google Scholar :

28 

Jeselsohn R, Cornwell M, Pun M, Buchwalter G, Nguyen M, Bango C, Huang Y, Kuang Y, Paweletz C, Fu X, et al: Embryonic transcription factor SOX9 drives breast cancer endocrine resistance. Proc Natl Acad Sci USA. 114:E4482–E4491. 2017. View Article : Google Scholar : PubMed/NCBI

29 

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

30 

Liu Z, Yan Y, Cao S and Chen Y: Long non-coding RNA SNHG14 contributes to gastric cancer development through targeting miR-145/SOX9 axis. J Cell Biochem. 119:6905–6913. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Xia S, Feng Z, Qi X, Yin Y, Jin J, Wu Y, Wu H, Feng Y and Tao M: Clinical implication of Sox9 and activated Akt expression in pancreatic ductal adenocarcinoma. Med Oncol. 32:3582015. View Article : Google Scholar

32 

Drusco A and Croce CM: MicroRNAs and cancer: A long story for short RNAs. Adv Cancer Res. 135:1–24. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Kanwal R, Plaga AR, Liu X, Shukla GC and Gupta S: MicroRNAs in prostate cancer: Functional role as biomarkers. Cancer Lett. 407:9–20. 2017. View Article : Google Scholar : PubMed/NCBI

34 

O’Bryan S, Dong S, Mathis JM and Alahari SK: The roles of oncogenic miRNAs and their therapeutic importance in breast cancer. Eur J Cancer. 72:1–11. 2017. View Article : Google Scholar

35 

Zheng Q, Cui X, Zhang D, Yang Y, Yan X, Liu M, Niang B, Aziz F, Liu S, Yan Q, et al: miR-200b inhibits proliferation and metastasis of breast cancer by targeting fucosyltransferase IV and α1,3-fucosylated glycans. Oncogenesis. 6:e3582017. View Article : Google Scholar

36 

Maimaitiming A, Wusiman A, Aimudula A, Tudahong T and Aisimutula D: WITHDRAWN: Downregulation of MicroRNA-152 and Inhibition of Cell Proliferation, Migration, and Invasion in Breast Cancer. Oncol Res, Oncol Res. Jun 15–2017.Epub ahead of print. View Article : Google Scholar : 032421

37 

Wang J, Song C, 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

38 

Sui Y, Zhang X, Yang H, Wei W and Wang M: MicroRNA-133a acts as a tumour suppressor in breast cancer through targeting LASP1. Oncol Rep. 39:473–482. 2018.

39 

Degli Esposti D, Aushev VN, Lee E, Cros MP, Zhu J, Herceg Z, Chen J and Hernandez-Vargas H: miR-500a-5p regulates oxidative stress response genes in breast cancer and predicts cancer survival. Sci Rep. 7:159662017. View Article : Google Scholar : PubMed/NCBI

40 

Chen X, Wang L, Qu J, Guan NN and Li JQ: Predicting miRNA-disease association based on inductive matrix completion. Bioinformatics. Jun 22–2018.Epub ahead of print. View Article : Google Scholar

41 

Chen X, Xie D, Wang L, Zhao Q, You ZH and Liu H: BNPMDA: Bipartite Network Projection for MiRNA-disease association prediction. Bioinformatics. 34:3178–3186. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Chen X and Huang L: LRSSLMDA: Laplacian regularized sparse subspace learning for MiRNA-disease association prediction. PLOS Comput Biol. 13:e10059122017. View Article : Google Scholar : PubMed/NCBI

43 

You ZH, Huang ZA, Zhu Z, Yan GY, Li ZW, Wen Z and Chen X: PBMDA: A novel and effective path-based computational model for miRNA-disease association prediction. PLOS Comput Biol. 13:e10054552017. View Article : Google Scholar : PubMed/NCBI

44 

Chen X, Xie D, Zhao Q and You ZH: MicroRNAs and complex diseases: From experimental results to computational models. Brief Bioinform. Oct 17–2017.Epub ahead of print. View Article : Google Scholar

45 

Chen X, Huang L, Xie D and Zhao Q: EGBMMDA: Extreme gradient boosting machine for MiRNA-disease association prediction. Cell Death Dis. 9:32018. View Article : Google Scholar : PubMed/NCBI

46 

Zhang HH, Pang M, Dong W, Xin JX, Li YJ, Zhang ZC, Yu L, Wang PY, Li BS and Xie SY: miR-511 induces the apoptosis of radioresistant lung adenocarcinoma cells by triggering BAX. Oncol Rep. 31:1473–1479. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Wang H, Leav I, Ibaragi S, Wegner M, Hu GF, Lu ML, Balk SP and Yuan X: SOX9 is expressed in human fetal prostate epithelium and enhances prostate cancer invasion. Cancer Res. 68:1625–1630. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Zhou CH, Ye LP, Ye SX, Li Y, Zhang XY, Xu XY and Gong LY: Clinical significance of SOX9 in human non-small cell lung cancer progression and overall patient survival. J Exp Clin Cancer Res. 31:182012. View Article : Google Scholar : PubMed/NCBI

49 

Bruun J, Kolberg M, Nesland JM, Svindland A, Nesbakken A and Lothe RA: Prognostic significance of β-catenin, E-cadherin, and SOX9 in colorectal cancer: Results from a large population-representative series. Front Oncol. 4:1182014. View Article : Google Scholar

Related Articles

Journal Cover

December-2018
Volume 53 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao Y, Pang W, Yang N, Hao L and Wang L: MicroRNA-511 inhibits malignant behaviors of breast cancer by directly targeting SOX9 and regulating the PI3K/Akt pathway Retraction in /10.3892/ijo.2022.5413. Int J Oncol 53: 2715-2726, 2018
APA
Zhao, Y., Pang, W., Yang, N., Hao, L., & Wang, L. (2018). MicroRNA-511 inhibits malignant behaviors of breast cancer by directly targeting SOX9 and regulating the PI3K/Akt pathway Retraction in /10.3892/ijo.2022.5413. International Journal of Oncology, 53, 2715-2726. https://doi.org/10.3892/ijo.2018.4576
MLA
Zhao, Y., Pang, W., Yang, N., Hao, L., Wang, L."MicroRNA-511 inhibits malignant behaviors of breast cancer by directly targeting SOX9 and regulating the PI3K/Akt pathway Retraction in /10.3892/ijo.2022.5413". International Journal of Oncology 53.6 (2018): 2715-2726.
Chicago
Zhao, Y., Pang, W., Yang, N., Hao, L., Wang, L."MicroRNA-511 inhibits malignant behaviors of breast cancer by directly targeting SOX9 and regulating the PI3K/Akt pathway Retraction in /10.3892/ijo.2022.5413". International Journal of Oncology 53, no. 6 (2018): 2715-2726. https://doi.org/10.3892/ijo.2018.4576