MicroRNA‑766 inhibits papillary thyroid cancer progression by directly targeting insulin receptor substrate 2 and regulating the PI3K/Akt pathway

Retraction in: /10.3892/ijo.2022.5376

  • Authors:
    • Jianjie Zhao
    • Zhirong Li
    • Yi Chen
    • Shu Zhang
    • Lingji Guo
    • Bo Gao
    • Yan Jiang
    • Wuguo Tian
    • Shuai Hao
    • Xiaohua Zhang
  • View Affiliations

  • Published online on: November 1, 2018     https://doi.org/10.3892/ijo.2018.4615
  • Pages: 315-325
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs/miRs) are widely dysregulated in papillary thyroid cancer (PTC). Dysregulated miRNAs, together with their target genes, comprise a complex network that has been implicated in the regulation of PTC pathogenesis. Further knowledge of the functional roles of aberrantly expressed miRNAs in PTC, and the underlying molecular mechanisms, may assist in the identification of novel therapeutic targets. miR‑766 has been well studied in human cancer; however, the expression status, specific roles and regulatory mechanisms of miR‑766 in PTC remain unclear. The present study aimed to detect miR‑766 expression in PTC tissues and cell lines, to explore the biological roles of miR‑766 in the malignant biological behaviors of PTC cells, and to determine the underlying mechanism of action of miR‑766 in PTC cells. The results revealed that miR‑766 was downregulated in PTC tissues and cell lines, and its downregulation was strongly associated with TNM stage and lymph node metastasis. Overexpression of miR‑766 inhibited PTC cell proliferation, colony formation, migration and invasion, promoted cell apoptosis and reduced tumor growth in vivo. Mechanistically, insulin receptor substrate 2 (IRS2) was identified as a direct target of miR‑766 in PTC cells. IRS2 was upregulated in PTC tissues, and this was inversely correlated with miR‑766 expression. Inhibition of IRS2 simulated the tumor suppressor activity of miR‑766 in PTC cells. Restoration of IRS2 expression negated the tumor‑suppressing effects of miR‑766 overexpression on PTC cells. Notably, miR‑766 directly targeted IRS2 to inhibit activation of the phosphoinositide 3‑kinase (PI3K)/protein kinase B (Akt) pathway in PTC cells in vitro and in vivo. Overall, these findings indicated that miR‑766 may inhibit the malignant biological behaviors of PTC cells by directly targeting IRS2 and regulating the PI3K/Akt pathway, thus suggesting that this miRNA may be a promising therapeutic target for PTC.

Introduction

Thyroid cancer, which is derived from follicular thyroid cells, is the most common endocrine malignancy, which accounts for ~90% of neuroendocrine tumors (1,2). The global incidence of thyroid cancer has markedly increased in recent decades (3), with an estimated 300,000 new cases diagnosed annually and 40,000 cases of thyroid cancer-associated morality occurring globally (4). Based on different pathological features, thyroid cancer can be divided into five subtypes: Papillary thyroid cancer (PTC), follicular thyroid cancer, poorly differentiated thyroid cancer, anaplastic thyroid cancer and thyroid squamous cell carcinoma (5). PTC, which accounts for 85-90% of all thyroid cancer cases, is the most prevalent histological subtype of thyroid cancer (6). Despite significant advances in diagnosis and therapy, recurrence and/or metastasis occurs in ~10% of patients with PTC, which heralds a poor prognosis (7). Therefore, an in-depth understanding of the molecular mechanism underlying the initiation and progression of PTC will aid in the development of novel diagnostic biomarkers and effective therapeutic strategies for patients with PTC.

MicroRNAs (miRNAs/miRs) are an abundant class of 17-24 nucleotide long, non-coding RNAs (8). miRNAs negatively modulate gene expression through direct binding to the 3-untranslated regions (3'-UTRs) of target genes, thus leading to translational inhibition and/or mRNA degradation (9). miRNAs not only serve crucial roles in regulating various fundamental cellular processes but are also closely associated with tumorigenesis and tumor development (10,11). Previous studies have reported that miRNAs are aberrantly expressed in almost all types of human cancer, including PTC (12), colorectal cancer (13), lung cancer (14), glioblastoma (15) and bladder cancer (16). Increasing evidence has revealed that various miRNAs are differentially expressed in PTC, and their dysregulation has been implicated in the regulation of PTC occurrence and development (17-19). Furthermore, miRNAs may have tumor suppressive or oncogenic roles in the progression of PTC and are able to regulate various cancer-associated biological processes, including cell proliferation, cell cycle progression, apoptosis, invasion, metastasis and epithelial-mesenchymal transition (20-22). Therefore, dysregulated miRNAs require further investigation, in order to identify potential therapeutic targets for the treatment of patients with PTC.

miR-766 has been well studied in numerous types of human cancer, including renal cell carcinoma (23), lung adenocarcinoma (24) and colorectal cancer (25). However, the expression status, specific roles and regulatory mechanisms of miR-766 in PTC remain largely unclear. Therefore, the aim of the present study was to detect miR-766 expression in PTC tissues and cell lines, to clarify the clinical significance of miR-766 in patients with PTC, and to explore the biological roles of miR-766 in the malignant biological behaviors of PTC cells. This study also aimed to determine the underlying mechanism of action of miR-766 in PTC cells.

Materials and methods

Clinical specimens

This study was approved by the Ethics Committee of Daping Hospital (Chongqing, China), in accordance with the principles expressed in the Declaration of Helsinki. Written informed consent was obtained from all participants prior to surgery. In total, 47 pairs of PTC tissues and matched adjacent normal tissues were collected from patients who had received surgical resection at Daping Hospital between June 2015 and May 2017. None of the patients had undergone chemotherapy or radiotherapy prior to surgery. All tissues were quickly frozen in liquid nitrogen after being excised and were stored at −80°C until further use.

Cell lines and culture conditions

A normal human thyroid cell line (HT-ori3), two human PTC cell lines (HTH83 and TPC-1) and a thyroid cancer cell line (BCPAP) were purchased from American Type Culture Collection (Manassas, VA, USA). Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% v/v heat-inactivated fetal bovine serum (FBS; both from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and 1% v/v penicillin-streptomycin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) were used to culture the cells. Cells were grown at 37°C in a humidified atmosphere containing 5% CO2.

Transfection

miR-766 mimics and a corresponding negative control (miR-NC) were purchased from Guangzhou RiboBio Co., Ltd. (Guangzhou, China). The sequences were as follows: miR-766 mimics, 5'-ACUCCAGCCCCACAGCCUCAGC-3'; miR-NC, 5'-UUCUCCGAACGUGUCACGUTT-3'. Small interfering RNA (siRNA) targeting insulin receptor substrate 2 (IRS2) expression (IRS2 siRNA) and a negative control siRNA (NC siRNA) were acquired from Shanghai GenePharma Co., Ltd. (Shanghai, China). The sequences were as follows: IRS2 siRNA, 5'-AAUAGCUGCAAGAGCGAUGAC-3'; NC siRNA, 5'-UUCUCCGAACGUGUCACGUTT-3'. An IRS2 overexpression plasmid was chemically synthesized using pcDNA3.1(+) basic vectors at the Chinese Academy of Sciences (Changchun, China). HTH83 and TPC-1 cells were seeded into 6-well plates at a density of 8×105 cells/well. Cells were transfected with mimics (100 pmol), siRNAs (100 pmol) or plasmids (4 µg) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Cells were incubated at 37°C in a humidified atmosphere containing 5% CO2. After 48 h, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), flow cytometric analysis, and cell migration and invasion assays were performed. MTT and colony formation assays were conducted at 24 h post-transfection. Western blot analysis was conducted 72 h post-transfection.

RT-qPCR analysis

Total RNA was isolated from cultured cells or homogenized tissues using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. For the detection of miR-766 expression, RT was performed using a TaqMan MicroRNA RT kit, according to the manufacturer's protocol, followed by qPCR with a TaqMan MicroRNA PCR kit (both from Applied Biosystems; Thermo Fisher Scientific, Inc.). The cycling conditions for qPCR were as follows: 50°C for 2 min and 95°C for 10 min, followed by 40 cycles of denaturation at 95°C for 15 sec and annealing/extension at 60°C for 60 sec, and a final extension step at 4°C for 5 min. U6 small nuclear RNA served as an internal control for miR-766 expression. For IRS2 mRNA quantification, first strand cDNA was synthesized using a PrimeScript RT Reagent kit (Takara Biotechnology Co., Ltd., Dalian, China), according to the manufacturer's protocol. qPCR was then performed using a SYBR Premix Ex Taq™ kit (Takara Biotechnology Co., Ltd.). The cycling conditions for qPCR were as follows: 5 min at 95°C, followed by 40 cycles at 95°C for 30 sec and 65°C for 45 sec, and a final extension step at 40°C for 30 sec. GAPDH was used for IRS2 mRNA normalization. The primers were designed as follows: miR-766, forward 5'-TCG AGTACTTGAGATGGAGTTTT-3', reverse 5'-GGCCGCGTTGCAGTGAGCCGAG-3'; U6, forward 5'-GCTTCGGCAGCACATATACTAAAAT-3', reverse 5'-CGCTTCACGAATTTGCGTGTCAT-3'; IRS2, forward, 5'-CACAATTCCAAGCGCCACAA-3', reverse 5'-ATCAAAGCTCCAGGCTGACC-3'; and GAPDH, forward 5'-ACCCACTCCTCCACCTTTG-3' and reverse 5'-CTCTTGTGCTCTTGCTGGG-3'. Relative gene expression was calculated using the 2−ΔΔCq method (26).

MTT assay

After 24 h of incubation, transfected cells in the exponential growth stage were collected and seeded into 96-well plates at a density of 3,000 cells/well. At 0, 24, 48 and 72 h following inoculation, the MTT assay was performed to determine cell proliferation. Briefly, 20 µl 5 mg/ml MTT solution (Sigma-Aldrich; Merck KGaA) was added to each well for 4 h at 37°C and 5% CO2. Subsequently, the culture medium was gently removed and formazan precipitates were dissolved in 100 µl dimethyl sulfoxide (Beyotime Institute of Biotechnology,, Inc., Shanghai, China). The absorbance was detected at a wavelength of 490 nm using a microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Colony formation assay

Logarithmically growing transfected cells were harvested after 24 h of incubation and were plated into 6-well plates at a density of 1,000 cells/well. Cells were then maintained at 37°C in a humidified atmosphere containing 5% CO2 for 2 weeks. Subsequently, cells were washed with PBS (Gibco; Thermo Fisher Scientific, Inc.), fixed with 100% methanol at room temperature for 20 min and stained with methyl violet (Beyotime Institute of Biotechnology) at room temperature for 20 min. The number of colonies (>50 cells/colony) was counted under an inverted light microscope (IX71; Olympus Corporation, Tokyo, Japan).

Flow cytometric analysis of cell apoptosis

An Annexin V-fluorescein isothiocyanate (FITC) Apoptosis Detection kit (Biolegend, Inc., San Diego, CA, USA) was used to evaluate the percentage of apoptotic cells. Briefly, transfected cells were seeded into 6-well plates (1×106 cells/well), incubated for 48 h at 37°C and 5% CO2, harvested, washed three times with PBS, and suspended in 100 µl binding buffer. Subsequently, 5 µl Annexin V-FITC and 5 µl propidium iodide were added and the transfected cells were incubated for 15 min at room temperature in the dark. Finally, flow cytometry (FACScan; BD Biosciences, Franklin Lakes, NJ, USA) was used to measure the rate of apoptosis. The data were analyzed with CellQuest version 5.1 (BD Biosciences).

Cell migration and invasion assays

The migratory ability of PTC cells was evaluated using Transwell chambers (BD Biosciences) with an 8-µm pore polycarbonate membrane. A total of 48 h post-transfection, 5×104 cells were suspended in FBS-free DMEM and were inoculated into the upper chamber. The lower chambers were filled with 500 µl DMEM supplemented with 20% FBS. After 24 h at 37°C, the non-migrated cells were removed using a cotton swab, whereas the migrated cells were fixed with 100% methanol and stained with 0.5% crystal violet (Beyotime Institute of Biotechnology). Images of the cells were captured and migratory ability was quantified by counting the number of migrated cells in five randomly selected visual fields from each chamber using an inverted light microscope. The cell invasion assay was conducted in a similar manner to the migration assay; however, the Transwell chambers were initially coated with Matrigel (BD Biosciences).

In vivo xenograft experiment

BALB/c nude mice (female; age, 4 weeks; weight, 20 g) were purchased from the Shanghai Laboratory Animal Center (Chinese Academy of Sciences, Shanghai, China) and were divided into two groups (n=4/group), which were subcutaneously injected with TPC-1 cells transfected with miR-766 mimics or miR-NC, respectively. The animals were maintained under specific pathogen-free conditions (25°C, 50% humidity, 10-h light/14-h dark cycle). The mice received free access to normal rodent food and water, which was autoclaved. Tumor length and width were measured every 2 days. BALB/c nude mice were sacrificed 30 days following implantation, and the tumor xenografts were excised and weighed. The tumor volumes were analyzed using the following formula: Tumor volume = 1/2 × tumor length × tumor width. Experimental procedures and protocols were approved by the Institutional Animal Care and Use Committee of Daping Hospital.

Bioinformatics analysis and luciferase reporter assay

The putative miR-766 target genes were predicted using TargetScan software (http://www.targetscan.org) and miRDB software (http://mirdb.org/). The wild-type (wt) and mutant (mut) 3'-UTRs of IRS2 were created by Shanghai GenePharma Co., Ltd., and were inserted downstream of the psiCHECK2 luciferase reporter vector (Promega Corporation, Madison, WI, USA), in order to generate psiCHECK2-IRS2-3'-UTR wt and psiCHECK2-IRS2-3'-UTR mut, respectively. Cells were plated into 24-well plates 12 h prior to transfection at a density of 1.5×105 cells/well. Co-transfection with the constructed luciferase reporter plasmids (0.2 µg) and miR-766 mimics (50 pmol) or miR-NC (50 pmol) was performed using Lipofectamine® 2000, according to the manufacturer's protocol. A total of 48 h post-transfection, luciferase activity was determined using a dual-luciferase reporter assay system (Promega Corporation), according to the manufacturer's protocol. The activity of firefly luciferase was normalized to that of Renilla luciferase.

Western blot analysis

Total protein was isolated from tissues or cells using radioimmunoprecipitation assay buffer (Sigma-Aldrich; Merck KGaA) and was quantified using a bicinchoninic acid protein assay kit (Beyotime Institute of Biotechnology). Equal amounts of protein (30 µg) were separated by 10% SDS-PAGE and were transferred to polyvinylidene fluoride membranes (Beyotime Institute of Biotechnology), followed by blocking at room temperature for 1 h with 5% w/v dried skimmed milk diluted in Tris-buffered saline with 0.1% Tween-20 (TBST). The membranes were then incubated overnight at 4°C with primary antibodies against IRS2 (cat. no. ab52606; 1:1,000 dilution; Abcam, Cambridge, UK), phosphorylated (p)-phosphoinositide 3-kinase (PI3K; cat. no. ab182651; 1:1,000 dilution; Abcam), PI3K (cat. no. ab191606; 1:1000 dilution; Abcam), p-protein kinase B (Akt; cat. no. sc-81433; 1:1,000 dilution; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), Akt (cat. no. sc-56878; 1:1,000 dilution; Santa Cruz Biotechnology, Inc.), or GAPDH (cat. no. ab128915; 1:1,000 dilution; Abcam). After three washes with TBST, the membranes were incubated with corresponding horseradish peroxidase-conjugated secondary antibodies (cat. nos. ab205719 or ab6721; 1:5,000 dilutions; Abcam) for 2 h at room temperature and were subjected to visualization using an enhanced chemiluminescence detection kit (Pierce; Thermo Fisher Scientific, Inc.). Protein expression was semi-quantified using Quantity One software version 4.62 (Bio-Rad Laboratories, Inc.).

Statistical analyses

SPSS (version 17.0; SPSS, Inc., Chicago, IL, USA) was used for statistical analysis. Data are presented as the means ± standard deviation from at least three independent experiments, and the differences between groups were compared using Student's t-test or one-way analysis of variance (ANOVA). Student-Newman-Keuls test was used as a post hoc test following ANOVA. The association between miR-766 and clinicopathological characteristics of patients with PTC was analyzed using the χ2 test. Spearman correlation analysis was employed to investigate the association between miR-766 and IRS2 mRNA expression in PTC tissues. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-766 expression is decreased in PTC tissues and cell lines

To uncover the expression pattern of miR-766 in PTC, RT-qPCR analysis was performed to detect miR-766 expression in 47 pairs of PTC tissues and matched adjacent normal tissues. PTC tissues exhibited significantly lower miR-766 expression compared with in the matched adjacent normal tissues (Fig. 1A; P<0.05). Subsequently, the clinical significance of miR-766 in PTC was investigated. Briefly, all patients with PTC were divided into miR-766 low (n=24) or miR-766 high (n=23) expression groups, based on the median value of miR-766 expression. Low miR-766 expression was significantly associated with TNM stage (P=0.008) and lymph node metastasis (P=0.039), whereas no obvious association was observed between miR-766 and age, sex or tumor size (all P>0.05; Table I). In addition, miR-766 expression was measured in two PTC cell lines and in a general thyroid cancer cell line. The data obtained from RT-qPCR indicated that miR-766 was downregulated in the two PTC cell lines (HTH83 and TPC-1) and the thyroid cancer cell line (BCPAP) compared with in the normal human thyroid cell line (HT-ori3) (Fig. 1B; P<0.05). These results indicated that downregulation of miR-766 may have a critical role in PTC tumorigenesis and development.

Table I

Association between miR-766 expression and clinico-pathological features in patients with papillary thyroid cancer.

Table I

Association between miR-766 expression and clinico-pathological features in patients with papillary thyroid cancer.

VariablemiR-766 expression
P-value
LowHigh
Age0.556
 <60 years810
 ≥60 years1613
Sex0.561
 Male127
 Female1216
Tumor size0.238
 <5 cm1512
 ≥5 cm911
Lymph node metastasis0.039a
 Negative613
 Positive1810
TNM stage0.008a
 I-II615
 III-IV188

a P<0.05. miR-766, microRNA-766.

miR-766 is involved in the regulation of cell proliferation, apoptosis, migration and invasion in PTC

To clarify the roles of miR-766 in PTC progression, HTH83 and TPC-1 cell lines, which exhibited lower miR-766 expression among the three thyroid cancer cell lines, were selected for functional assays and were transfected with miR-766 mimics or miR-NC. Post-transfection, miR-766 was markedly overexpressed in miR-766 mimics-transfected HTH83 and TPC-1 cells compared with in cells transfected with miR-NC (Fig. 2A; P<0.05). The role of miR-766 overexpression in the proliferation of PTC cells was determined by MTT and colony formation assays. Upregulation of miR-766 significantly inhibited the proliferation (Fig. 2B, P<0.05) and colony formation (Fig. 2C, P<0.05) of HTH83 and TPC-1 cells. Since an alteration in cell proliferation is often accompanied with changes in the rate of apoptosis, the effects of miR-766 on cell apoptosis were assessed using flow cytometry. Transfection of miR-766 mimics markedly promoted the apoptosis of HTH83 and TPC-1 cells (Fig. 2D; P<0.05). Cell migration and invasion assays were used to determine whether miR-766 was implicated in the regulation of PTC cell metastasis. Ectopic miR-766 expression markedly restricted the migration (Fig. 2E; P<0.05) and invasion (Fig. 2F; P<0.05) of HTH83 and TPC-1 cells. These findings suggested that miR-766 may exert tumor suppressor activity in PTC cells.

IRS2 is a direct target gene of miR-766 in PTC cells

Based on bioinformatics analysis, the present study revealed that the 3'-UTR of IRS2 matched the seed sequence of miR-766 (Fig. 3A). IRS2 was selected for further experimental identification because this gene has previously been implicated in the initiation and progression of PTC (27). A luciferase reporter assay was performed to test this hypothesis. The assay demonstrated that ectopic miR-766 expression significantly reduced the luciferase activity of psiCHECK2-IRS2-3'-UTR wt in HTH83 and TPC-1 cells (Fig. 3B, P<0.05), but not that of psiCHECK2-IRS2-3'-UTR mut. These findings indicated that the 3'-UTR of IRS2 may be directly targeted by miR-766 in PTC cells. To further clarify the association between miR-766 and IRS2 in PTC, IRS2 mRNA expression was detected in 47 pairs of PTC tissues and matched adjacent normal tissues using RT-qPCR. The mRNA expression levels of IRS2 were significantly increased in PTC tissues (Fig. 3C; P<0.05) and were inversely correlated with the expression levels of miR-766 (Fig. 3D; r=-0.5143, P=0.0002), as determined by Spearman correlation analysis. To investigate whether endogenous IRS2 expression could be regulated by miR-766, RT-qPCR and western blot analysis were carried out to detect IRS2 mRNA and protein expression in HTH83 and TPC-1 cells upon miR-766 overexpression. Compared with in the miR-NC group, HTH83 and TPC-1 cells transfected with miR-766 mimics exhibited significantly decreased levels of IRS2 mRNA (Fig. 3E; P<0.05) and protein (Fig. 3F, P<0.05). Taken together, these results supported the hypothesis that IRS2 is a direct target gene of miR-766 in PTC cells.

Knockdown of IRS2 simulates tumor suppressor activity of miR-766 in PTC cells

IRS2 was validated as a direct target gene of miR-766 in PTC cells. Therefore, it was hypothesized that the tumor-suppressing roles of miR-766 in PTC cells may be achieved by IRS2 knockdown. To evaluate this hypothesis, HTH83 and TPC-1 cells were transfected with IRS2 siRNA to knockdown endogenous IRS2 expression and examine the function of IRS2 in PTC cells. Western blot analysis indicated that IRS2 protein expression was significantly decreased in IRS2 siRNA-transfected HTH83 and TPC-1 cells (Fig. 4A; P<0.05). Inhibition of IRS2 significantly restricted HTH83 and TPC-1 cell proliferation (Fig. 4B; P<0.05) and colony formation (Fig. 4C; P<0.05), and markedly enhanced cell apoptosis (Fig. 4D; P<0.05). In addition, as shown in Fig. 4E and F, IRS2 knockdown significantly attenuated the migratory (P<0.05) and invasive (P<0.05) abilities of HTH83 and TPC-1 cells compared with in NC siRNA-transfected cells. These results demonstrated that the effects of IRS2 knockdown on PTC cells were similar to the effects of miR-766 overexpression, further suggesting that IRS2 is a functional downstream target of miR-766 in PTC cells.

Reintroduction of IRS2 rescues the effects of miR-766 on the malignant behaviors of PTC cells

Rescue experiments were performed to determine whether miR-766 exerts tumor-suppressing roles in PTC cells by inhibiting IRS2 expression. HTH83 and TPC-1 cells were co-transfected with miR-766 mimics and empty pcDNA3.1 plasmid or pcDNA3.1-IRS2 lacking the 3'-UTR. Western blot analysis was used to detect the protein expression levels of IRS2 in the rescue experiment. miR-766 overexpression-induced down-regulation of IRS2 was significantly recovered in HTH83 and TPC-1 cells following co-transfection with miR-766 mimics and pcDNA3.1-IRS2 (Fig. 5A; P<0.05). In addition, functional analyses indicated that restored IRS2 expression could reverse the tumor suppressive roles of miR-766 overexpression on the proliferation (Fig. 5B; P<0.05), colony formation (Fig. 5C; P<0.05), apoptosis (Fig. 5D; P<0.05), migration (Fig. 5E; P<0.05) and invasion (Fig. 5F; P<0.05) of HTH83 and TPC-1 cells. These results collectively indicated that miR-766 may inhibit the development of PTC cells, at least partially, through the downregulation of IRS2 expression.

miR-766 suppresses activation of the PI3K/Akt pathway in PTC cells by directly targeting IRS2

IRS2 has previously been reported to participate in regulation of the PI3K/Akt pathway (28-30). Therefore, it was hypothesized that miR-766 may target IRS2 to inhibit the activation of PI3K/Akt signaling in PTC cells. To confirm this hypothesis, HTH83 and TPC-1 cells were co-transfected with miR-766 mimics and pcDNA3.1 or pcDNA3.1-IRS2, and western blot analysis was performed 72 h post-transfection to determine the expression of molecules associated with the PI3K/Akt pathway. A marked decrease in p-PI3K and p-Akt was observed in HTH83 and TPC-1 cells upon miR-766 overexpression; however, this did not affect total PI3K and Akt expression (Fig. 6). Furthermore, recovered IRS2 expression rescued the cellular levels of p-PI3K and p-Akt, which were downregulated by miR-766 overexpression. These results suggested that miR-766 may inhibit the PI3K/Akt signaling in PTC cells via regulation of IRS2.

miR-766 hinders tumor growth in PTC in vivo

To explore the precise role of miR-766 in PTC in vivo, xenograft experiments were performed by subcutaneously injecting miR-766-overexpressing TPC-1 cells into nude mice. The miR-766-overexpressing tumor xenografts exhibited significant suppression of tumor volume compared with in the miR-NC group (Fig. 7A and B; P<0.05). Subsequently, tumor xenografts were weighed; upregulation of miR-766 decreased tumor weight in vivo (Fig. 7C; P<0.05). In addition, RT-qPCR analysis revealed that miR-766 was markedly upregulated in tumor xenografts that were generated using miR-766 mimics-transfected cells (Fig. 7D; P<0.05). Subsequently, the protein expression levels of IRS2 and molecules associated with the PI3K/Akt pathway were detected in tumor xenografts using western blot analysis. The protein expression levels of IRS2, p-PI3K and p-Akt were downregulated in the tumor xenografts from the miR-766 mimics groups compared with in those from the miR-NC group (Fig. 7E). Taken together, these results suggested that miR-766 may hinder PTC tumor growth in vivo by suppressing IRS2 and activation of the PI3K/Akt pathway.

Discussion

miRNAs are widely dysregulated in PTC, and dysregulated miRNAs, together with their target genes, comprise a complex network that has been implicated in the regulation of PTC pathogenesis (25,31,32). Notably, miRNAs have been proposed as novel diagnostic biomarkers and potential therapeutic targets for anticancer treatment (33). Therefore, further exploration of the functional roles of aberrantly expressed miRNAs in PTC and the underlying molecular mechanisms may aid in the identification of novel therapeutic targets. The present study is the first, to the best of our knowledge, to detect miR-766 expression in PTC, to clarify the clinical significance of miR-766 in PTC, and to examine the detailed roles of miR-766 in PTC progression. Notably, the molecular mechanisms underlying the activity of miR-766 in PTC cells were explored.

Expression of miR-766 is decreased in renal cell carcinoma, and low miR-766 expression is significantly correlated with the clinical stage. In addition, patients with renal cell carcinoma and low miR-766 expression exhibit poorer prognosis compared with those patients with high miR-766 expression (23). Conversely, miR-766 is overexpressed in lung adenocarcinoma (24) and colorectal cancer (25). miR-766 has been identified as an independent prognostic biomarker predicting the clinical outcomes of patients with lung adenocarcinoma (24). These inconsistent observations indicate that the expression pattern of miR-766 displays tissue specificity; however, the expression status of miR-766 in PTC remains unknown. The present study demonstrated that miR-766 was markedly downregulated in PTC tissues and cell lines. In addition, the expression levels of miR-766 in PTC tissues were associated with TNM stage and lymph node metastasis. These findings implicated miR-766 as an attractive diagnostic and prognosis biomarker for patients with PTC.

Dysregulation of miR-766 is involved in the modulation of tumorigenesis and tumor development of numerous types of human cancer. Restoration of miR-766 expression inhibits renal cell carcinoma cell growth and promotes cell cycle arrest in vitro, as well as decreasing tumor growth in vivo (23). In breast cancer, upregulation of miR-766 suppresses tumor sphere formation and invasion in vitro, and attenuates in vivo lung metastasis (34). Conversely, miR-766 acts as an oncogene in colorectal cancer and increases cell growth (35,36). Nevertheless, the biological roles of miR-766 in PTC remain largely unidentified. In the present study, functional assays revealed that miR-766 may have tumor suppressor activity in PTC by affecting cell proliferation, colony formation, apoptosis, migration and invasion in vitro, and tumor growth in vivo. Therefore, miR-766 may represent a valuable therapeutic target for the treatment of patients with PTC.

miRNAs regulate biological behaviors associated with carcinogenesis and cancer progression by directly binding to the 3'-UTRs of their target genes. Several genes, including splicing factor SF2 (23) in renal cell carcinoma, sex determining region Y-box 6 (35) and DNA methyl-transferase 3B (36) in colorectal cancer, have previously been demonstrated to be direct target genes of miR-766. To illustrate the mechanisms underlying the cellular response to miR-766, this study aimed to determine whether IRS2 is a direct target gene of miR-766 in PTC. Bioinformatics analysis predicted that the 3'-UTR of IRS2 matched the seed sequence of miR-766. Luciferase reporter assays, RT-qPCR and western blot analysis revealed that miR-766 could directly bind to the 3'-UTR of IRS2 and decrease endogenous IRS2 expression in PTC cells. IRS2 was upregulated in PTC tissues, and the IRS2 expression was inversely correlated with miR-766 expression. The effects of IRS2 inhibition on PTC cells were similar to the effects of miR-766 overexpression. Furthermore, IRS2 reintroduction could abrogate the effects of miR-766 on the malignant behaviors of PTC cells. These collective results provided sufficient evidence to indicate that IRS2 may be a direct and functional downstream target of miR-766 in PTC cells.

IRS2 is a member of the IRS family, which mainly interacts with SH2 domain-containing proteins to serve as adaptor proteins for additional surface receptors (37). IRS2 is overexpressed in several types of human cancer, including colorectal cancer (38), renal cell carcinoma (39), lung cancer (40) and breast cancer (41). It is a multifunctional oncogene that has been implicated in the regulation of numerous biological behaviors, such as cell proliferation, cell cycle, apoptosis, invasion and epithelial-mesenchymal transition (42-44). A previous study also demonstrated that IRS2 is upregulated in PTC and serves oncogenic roles in the progression of PTC (27). In this study, miR-766 directly targeted IRS2 to inhibit the malignancy of PTC cells. These prior data suggested that the newly identified miR-766/IRS2 axis may provide an effective therapeutic target for the management of patients with PTC.

In conclusion, miR-766 was revealed to possess antitumor properties and restrict the malignant biological behaviors of PTC cells, at least in part by inhibiting IRS2 and activation of the PI3K/Akt pathway. However, this study did not explore the association between miR-766 and prognosis of patients with PTC; this is a limitation, which will be resolved in future studies.

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

XZ and JZ designed the study. JZ, ZL and YC conducted RT-qPCR, MTT assay and colony formation assay. SZ, LG and BG performed flow cytometry, migration and invasion assays, and luciferase reporter assay. YJ, WT and SH carried out the in vivo xenograft experiment and western blot analysis. They have read and approved the final draft.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Daping Hospital, and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of Daping Hospital. 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 

Xiang D, Xie L, Xu Y, Li Z, Hong Y and Wang P: Papillary thyroid microcarcinomas located at the middle part of the middle third of the thyroid gland correlates with the presence of neck metastasis. Surgery. 157:526–533. 2015. View Article : Google Scholar

2 

Kim HY, Park WY, Lee KE, Park WS, Chung YS, Cho SJ and Youn YK: Comparative analysis of gene expression profiles of papillary thyroid microcarcinoma and papillary thyroid carcinoma. J Cancer Res Ther. 6:452–457. 2010. View Article : Google Scholar

3 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Ye Y, Zhuang J, Wang G, He S, Ni J and Xia W: MicroRNA-139 targets fibronectin 1 to inhibit papillary thyroid carcinoma progression. Oncol Lett. 14:7799–7806. 2017.PubMed/NCBI

5 

Catalano MG, Poli R, Pugliese M, Fortunati N and Boccuzzi G: Emerging molecular therapies of advanced thyroid cancer. Mol Aspects Med. 31:215–226. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Peng XG, Chen ZF, Zhang KJ, Wang PG, Liu ZM, Chen ZJ, Hou GY and Niu M: VEGF Trapon inhibits tumor growth in papillary thyroid carcinoma. Eur Rev Med Pharmacol Sci. 19:235–240. 2015.PubMed/NCBI

7 

Fagin JA and Wells SA Jr: Biologic and clinical perspectives on thyroid cancer. N Engl J Med. 375:23072016. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

He L and Hannon GJ: MicroRNAs: Small RNAs with a big role in gene regulation. Nat Rev Genet. 5:522–531. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Dragomir M, Mafra ACP, Dias SMG, Vasilescu C and Calin GA: Using microRNA Networks to understand cancer. Int J Mol Sci. 19:192018. View Article : Google Scholar

11 

Iorio MV, Casalini P, Tagliabue E, Ménard S and Croce CM: MicroRNA profiling as a tool to understand prognosis, therapy response and resistance in breast cancer. Eur J Cancer. 44:2753–2759. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Aragon Han P, Weng CH, Khawaja HT, Nagarajan N, Schneider EB, Umbricht CB, Witwer KW and Zeiger MA: MicroRNA expression and association with clinicopathologic features in papillary thyroid cancer: A systematic review. Thyroid. 25:1322–1329. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Yi R, Li Y, Wang FL, Miao G, Qi RM and Zhao YY: MicroRNAs as diagnostic and prognostic biomarkers in colorectal cancer. World J Gastrointest Oncol. 8:330–340. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Uddin A and Chakraborty S: Role of miRNAs in lung cancer. J Cell Physiol. Apr 20–2018.Epub ahead of print. View Article : Google Scholar

15 

Ahir BK, Ozer H, Engelhard HH and Lakka SS: MicroRNAs in glioblastoma pathogenesis and therapy: A comprehensive review. Crit Rev Oncol Hematol. 120:22–33. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Homami A and Ghazi F: MicroRNAs as biomarkers associated with bladder cancer. Med J Islam Repub Iran. 30:4752016.

17 

Mutalib NS, Yusof AM, Mokhtar NM, Harun R, Muhammad R and Jamal R: MicroRNAs and lymph node metastasis in papillary thyroid cancers. Asian Pac J Cancer Prev. 17:25–35. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Lee JC, Gundara JS, Glover A, Serpell J and Sidhu SB: MicroRNA expression profiles in the management of papillary thyroid cancer. Oncologist. 19:1141–1147. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Zhu G, Xie L and Miller D: Expression of microRNAs in thyroid carcinoma. Methods Mol Biol. 1617:261–280. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Fu YT, Zheng HB, Zhang DQ, Zhou L and Sun H: MicroRNA-1266 suppresses papillary thyroid carcinoma cell metastasis and growth via targeting FGFR2. Eur Rev Med Pharmacol Sci. 22:3430–3438. 2018.PubMed/NCBI

21 

Wang R, Ma Q, Ji L, Yao Y, Ma M and Wen Q: miR-622 suppresses tumor formation by directly targeting VEGFA in papillary thyroid carcinoma. OncoTargets Ther. 11:1501–1509. 2018. View Article : Google Scholar

22 

Ma S, Jia W and Ni S: miR-199a-5p inhibits the progression of papillary thyroid carcinoma by targeting SNAI1. Biochem Biophys Res Commun. 497:181–186. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Chen C, Xue S, Zhang J, Chen W, Gong D, Zheng J, Ma J, Xue W, Chen Y, Zhai W, et al: DNA-methylation-mediated repression of miR-766-3p promotes cell proliferation via targeting SF2 expression in renal cell carcinoma. Int J Cancer. 141:1867–1878. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Li X, Shi Y, Yin Z, Xue X and Zhou B: An eight-miRNA signature as a potential biomarker for predicting survival in lung adenocarcinoma. J Transl Med. 12:1592014. View Article : Google Scholar : PubMed/NCBI

25 

Suresh R, Sethi S, Ali S, Giorgadze T and Sarkar FH: Differential expression of microRNAs in papillary thyroid carcinoma and their Role in Racial Disparity. J Cancer Sci Ther. 7:145–154. 2015.PubMed/NCBI

26 

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

27 

Dong S, Meng X, Xue S, Yan Z, Ren P and Liu J: microRNA-141 inhibits thyroid cancer cell growth and metastasis by targeting insulin receptor substrate 2. Am J Transl Res. 8:1471–1481. 2016.PubMed/NCBI

28 

Landis J and Shaw LM: Insulin receptor substrate 2-mediated phosphatidylinositol 3-kinase signaling selectively inhibits glycogen synthase kinase 3β to regulate aerobic glycolysis. J Biol Chem. 289:18603–18613. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Mercado-Matos J, Clark JL, Piper AJ, Janusis J and Shaw LM: Differential involvement of the microtubule cytoskeleton in insulin receptor substrate 1 (IRS-1) and IRS-2 signaling to AKT determines the response to microtubule disruption in breast carcinoma cells. J Biol Chem. 292:7806–7816. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Jeong SH and Lim DS: Insulin receptor substrate 2: A bridge between Hippo and AKT pathways. BMB Rep. 51:209–210. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Hu J, Li C, Liu C, Zhao S, Wang Y and Fu Z: Expressions of miRNAs in papillary thyroid carcinoma and their associations with the clinical characteristics of PTC. Cancer Biomark. 18:87–94. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Yoruker EE, Terzioglu D, Teksoz S, Uslu FE, Gezer U and Dalay N: MicroRNA expression profiles in papillary thyroid carcinoma, benign thyroid nodules and healthy controls. J Cancer. 7:803–809. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Hu Y, Wang H, Chen E, Xu Z, Chen B and Lu G: Candidate microRNAs as biomarkers of thyroid carcinoma: A systematic review, meta-analysis, and experimental validation. Cancer Med. 5:2602–2614. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Oh K and Lee DS: In vivo validation of metastasis-regulating microRNA-766 in human triple-negative breast cancer cells. Lab Anim Res. 33:256–263. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Li YC, Li CF, Chen LB, Li DD, Yang L, Jin JP and Zhang B: MicroRNA-766 targeting regulation of SOX6 expression promoted cell proliferation of human colorectal cancer. OncoTargets Ther. 8:2981–2988. 2015. View Article : Google Scholar

36 

Afgar A, Fard-Esfahani P, Mehrtash A, Azadmanesh K, Khodarahmi F, Ghadir M and Teimoori-Toolabi L: miR-339 and especially miR-766 reactivate the expression of tumor suppressor genes in colorectal cancer cell lines through DNA methyltransferase 3B gene inhibition. Cancer Biol Ther. 17:1126–1138. 2016. View Article : Google Scholar : PubMed/NCBI

37 

White MF: IRS proteins and the common path to diabetes. Am J Physiol Endocrinol Metab. 283:E413–E422. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Day E, Poulogiannis G, McCaughan F, Mulholland S, Arends MJ, Ibrahim AE and Dear PH: IRS2 is a candidate driver oncogene on 13q34 in colorectal cancer. Int J Exp Pathol. 94:203–211. 2013.PubMed/NCBI

39 

Ma Y, Zhang H, He X, Song H, Qiang Y, Li Y, Gao J and Wang Z: miR-106a* inhibits the proliferation of renal carcinoma cells by targeting IRS-2. Tumour Biol. 36:8389–8398. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Xu H, Lee MS, Tsai PY, Adler AS, Curry NL, Challa S, Freinkman E, Hitchcock DS, Copps KD, White MF, et al: Ablation of insulin receptor substrates 1 and 2 suppresses Kras-driven lung tumorigenesis. Proc Natl Acad Sci USA. 115:4228–4233. 2018. View Article : Google Scholar

41 

Porter HA, Perry A, Kingsley C, Tran NL and Keegan AD: IRS1 is highly expressed in localized breast tumors and regulates the sensitivity of breast cancer cells to chemotherapy, while IRS2 is highly expressed in invasive breast tumors. Cancer Lett. 338:239–248. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Zhang P, Shao G, Lin X, Liu Y and Yang Z: miR-338-3p inhibits the growth and invasion of non-small cell lung cancer cells by targeting IRS2. Am J Cancer Res. 7:53–63. 2017.PubMed/NCBI

43 

de Melo Campos P, Machado-Neto JA, Eide CA, Savage SL, Scopim-Ribeiro R, da Silva Souza Duarte, Favaro PA, Lorand-Metze I, Costa FF, Tognon CE, et al: IRS2 silencing increases apoptosis and potentiates the effects of ruxolitinib in JAK2V617F-positive myeloproliferative neoplasms. Oncotarget. 7:6948–6959. 2016.PubMed/NCBI

44 

Liu H, Ren G, Zhu L, Liu X and He X: The upregulation of miRNA-146a inhibited biological behaviors of ESCC through inhibition of IRS2. Tumour Biol. 37:4641–4647. 2016. View Article : Google Scholar

Related Articles

Journal Cover

January-2019
Volume 54 Issue 1

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 J, Li Z, Chen Y, Zhang S, Guo L, Gao B, Jiang Y, Tian W, Hao S, Zhang X, Zhang X, et al: MicroRNA‑766 inhibits papillary thyroid cancer progression by directly targeting insulin receptor substrate 2 and regulating the PI3K/Akt pathway Retraction in /10.3892/ijo.2022.5376. Int J Oncol 54: 315-325, 2019
APA
Zhao, J., Li, Z., Chen, Y., Zhang, S., Guo, L., Gao, B. ... Zhang, X. (2019). MicroRNA‑766 inhibits papillary thyroid cancer progression by directly targeting insulin receptor substrate 2 and regulating the PI3K/Akt pathway Retraction in /10.3892/ijo.2022.5376. International Journal of Oncology, 54, 315-325. https://doi.org/10.3892/ijo.2018.4615
MLA
Zhao, J., Li, Z., Chen, Y., Zhang, S., Guo, L., Gao, B., Jiang, Y., Tian, W., Hao, S., Zhang, X."MicroRNA‑766 inhibits papillary thyroid cancer progression by directly targeting insulin receptor substrate 2 and regulating the PI3K/Akt pathway Retraction in /10.3892/ijo.2022.5376". International Journal of Oncology 54.1 (2019): 315-325.
Chicago
Zhao, J., Li, Z., Chen, Y., Zhang, S., Guo, L., Gao, B., Jiang, Y., Tian, W., Hao, S., Zhang, X."MicroRNA‑766 inhibits papillary thyroid cancer progression by directly targeting insulin receptor substrate 2 and regulating the PI3K/Akt pathway Retraction in /10.3892/ijo.2022.5376". International Journal of Oncology 54, no. 1 (2019): 315-325. https://doi.org/10.3892/ijo.2018.4615