MicroRNA‑212 inhibits colorectal cancer cell viability and invasion by directly targeting PIK3R3

Retraction in: /10.3892/mmr.2023.13025

  • Authors:
    • Jian Zhang
    • Yongkang Zhang
    • Xiaoyun Li
    • Hongbo Wang
    • Quan Li
    • Xiaofeng Liao
  • View Affiliations

  • Published online on: September 20, 2017     https://doi.org/10.3892/mmr.2017.7552
  • Pages: 7864-7872
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) is the third most common cancer and the fourth most common cause of cancer‑associated mortalities worldwide. Emerging evidence has shown that abnormal microRNA (miRNA) expression contributes to CRC carcinogenesis and progression by inhibiting the expression of their target genes. Therefore, investigating the expression patterns and roles of miRNAs specifically involved in CRC formation, and progression would help expand our knowledge on CRC and provide novel therapeutic targets for CRC treatment. Previous studies suggest that miR‑212 is involved in the carcinogenesis and progression of multiple human cancer types. In this study, miR‑212 was significantly downregulated in CRC tissues and cell lines. Functional experiments demonstrated that miR‑212 overexpression inhibited the in vitro viability and invasion of CRC cells. In addition, phosphoinositide‑3‑kinase regulatory subunit 3 (PIK3R3) was confirmed as the direct target of miR‑212 in CRC. Furthermore, PIK3R3 was highly expressed in CRC tissues and inversely correlated with miR‑212 expression. Increased PIK3R3 expression effectively rescued the tumor‑suppressing roles of miR‑212 on CRC cell viability and invasion. Moreover, miR‑212 upregulation blocked the protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) signalling pathway in CRC cells. These findings suggest that miR‑212 acts as a tumor suppressor in CRC by directly targeting PIK3R3 and regulating the AKT/mTOR signaling pathway. Thus, miR‑212 may serve as an effective therapeutic target for the treatment of patients with CRC.

Introduction

Colorectal cancer (CRC) is the third most common cancer and the fourth most common cause of cancer-related deaths worldwide (1), with almost 1.23 million new cases and 0.6 million deaths per year (2). The initiation and progression of CRC are influenced by several risk factors, such as colon polyps, ulcerative colitis, age, smoking, obesity, environment, lifestyle, diet and genetic and epigenetic factors (3,4). Current standard treatments for CRC patients include surgical resection, chemotherapy, radiotherapy or a combination of these strategies (5). Despite remarkable progresses in the diagnosis and therapy of CRC patients, the prognosis of these patients remains dismal (6). Recurrence and metastasis are the major causes of death in CRC patients (7). Therefore, elucidating the mechanisms involved in the formation and progression of CRC and identifying novel therapeutic methods for CRC patients are urgently needed.

MicroRNAs (miRNAs) are a recently discovered large group of endogenous, non-coding, single-stranded and short RNA molecules with approximately 19–23 nucleotides in length (8). MiRNAs act as an endogenous regulator of gene expression by binding to the 3′-untranslated regions (3′-UTRs) of their target genes, leading to translational inhibition or mRNA degradation (9). Increasing evidence has indicated that >30% of genes are regulated by miRNAs and play important roles in various cellular biological processes, including proliferation, apoptosis, differentiation, movement, migration and survival (1012). Dysregulated miRNAs have been closely correlated with tumorigenesis, promotion and development by acting on many oncogenes and tumor suppressors (1315). Highly expressed miRNAs serve as oncogenes by negatively regulating tumor suppressor genes (16). By contrast, miRNAs expressed at low levels may act as tumor suppressors by clocking oncogenes (17). Therefore, exploring the expression and biological functions of miRNAs in CRC may provide potential diagnostic and therapeutic targets for the treatment of CRC patients.

Previous studies reported that aberrant miR-212 expression contributes to tumor progression in various types of human cancers (1820). In this study, we further investigate the expression, effects and related molecular mechanisms of miR-212 in CRC.

Materials and methods

Tissue specimens

Human CRC tissues and corresponding adjacent non-neoplastic tissues were collected between October 2014 and March 2016 from surgical specimens from 28 patients with CRC at Xiangyang Central Hospital (Xiangyang, China). None of these patients were treated with chemotherapy or radiotherapy before surgery. These specimens were frozen immediately after resection and stored at −80°C until further use. The study was approved by the Ethical Review Committees of Xiangyang Central Hospital, and all patients gave informed consent prior to specimen collection according to institutional guidelines.

Cell culture and transfection

Five human CRC cell lines (HCT116, HT29, LoVo, SW480, SW620) and normal human colon epithelium cell line FHC were purchased from American Type Culture Collection (Manassas, VA, USA). Cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% heat-inactivated foetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) 100 U/ml streptomycin and 100 U/ml penicillin and grown at 37°C in a humidified incubator with 5% CO2.

MiR-212 mimics and miRNA mimics negative control (miR-NC) were obtained from RiboBio Co., Ltd. (Guangzhou, China). PIK3R3 overexpression plasmid without the 3′-UTR of PIK3R3 (pcDNA3.1-PIK3R3) and blank plasmid pcDNA3.1 were acquired from GeneCopoeia (Guangzhou, China). Cells were seeded into 6-well plates with a density of 60–70% confluence each well. Cells were transfected with miR-212 mimics (100 pmol), miR-NC (100 pmol), or cotransfected with miR-212 mimics (100 pmol) and pcDNA3.1-PIK3R3 (1 µg) using Lipofectamine 2000 transfection reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from tissue samples or cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. The quantity and the quality of total RNA was assessed using a Nanodrop spectrophotometer (Thermo Fisher Scientific, Wilmington, DE, USA). To analysis miR-212 expression, cDNA of miRNA was synthesized from total RNA using TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems, Carlsbad, CA, USA). Real-time quantitative PCR was performed using a TaqMan MicroRNA PCR kit (Applied Biosystems). U6 was used as an internal control for miR-212. To quantify PIK3R3 mRNA expression, total RNA was reversed transcription into cDNA using M-MLV Reverse Transcriptase (Promega, Madison, WI, USA) and qPCR was conducted using SYBR Premix Ex Taq™ (Takara Biotechnology Co., Ltd., Dalian, China). β-actin served as an internal control for PIK3R3 mRNA expression. The primers were designed as follows: miR-212, 5′-CGCTAACAGTCTCCAGTC-3′ (forward) and 5′-GTGCAGGGTCCGAGGT-3′ (reverse); U6, 5′-CTCGCTTCGGCAGCACATATACT-3′ (forward) and 5′-ACGCTTCACGAATTTGCGTGTC-3′ (reverse); PIK3R3, 5′-CTTGCTCTGTGGTGGCCGAT-3′ (forward) and 5′-GACGTTGAGGGAGTCGTTGT-3′ (reverse); and β-actin, 5′-TGGCACCCAGCACAATGAA-3′ (forward) and 5′-TAAGTCATAGTCCGCCTAGAAGCA-3′ (reverse). The relative expression of miR-212 and PIK3R3 mRNA were calculated using the 2−ΔΔCt method (21).

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

MTT assay was utilized to determine cell viability. Briefly, cells were seeded into 96-well plates at a density of 3×103 cells per well. After incubation overnight at 37°C, cell transfection was performed and grown for 0, 24, 48, and 72 h. At indicated time point, 20 ul MTT solution (5 mg/ml; Sigma, St. Louis, MO, USA) was added into each well and incubated at 37°C for additional 4 h. Subsequently, culture medium was removed and 200 µl DMSO (Sigma) was added to each well. After incubation at 37°C incubator for 5 min to dissolve crystals, the optical density (OD) was examined at a wavelength of 490 nm using a microplate reader (Bio-Rad, Richmond, CA, USA). All experiments were performed in triplicate and repeated three times.

Transwell invasion assay

Cell invasive ability was evaluated with Transwell chambers (8-µm pores; Corning Costar Corp, Cambridge, MA, USA) coated with Matrigel (BD Biosciences, San Jose, CA, USA). Briefly, transfected cells were collected at 48 h post-transfection and suspended in FBS-free DMEM culture medium. 5×104 transfected cells were seeded into the upper chamber. DMEM containing 10% FBS was placed in the lower compartment as a chemoattractant. After 24 h incubation at 37°C with 5% CO2, non-invasive cells were wiped out carefully with cotton swab. The invasive cells on the underside of the filter membrane were fixed with 70% ethanol for 20 min and stained with 0.1% crystal violet for 10 min. Invasive cells were photographed and counted under a IX71 inverted microscope (Olympus, Tokyo, Japan) at magnification, ×200 in 5 randomly selected microscopic fields.

Bioinformatics analysis

The bioinformatics software PICTA (http://pictar.mdc-berlin.de/) and TargetScan (http://www.targetscan.org/) were adopted to predict candidate targets of miR-212.

Luciferase reporter assay

Luciferase reporter plasmid containing predicted miR-212 seed-matching sites in the 3′-UTR of PIK3R3 and corresponding mutant sites were constructed and confirmed by RiboBio. The constructed vectors were named as pMIR-PIK3R3-3′-UTR Wild-type (Wt) and pMIR-PIK3R3-3′-UTR mutant (Mut), respectively. For luciferase reporter assay, cells were seeded into 24-well plates and transfected with miR-212 mimics or miR-NC, and together with luciferase reporter plasmid using Lipofectamine 2000, according to the manufacturer's protocol. At 48 h after transfection, cells were harvested and luciferase activities were measured using the Dual-Luciferase Reporter Assay System (Promega), following the protocol provided by the manufacturer. Firefly luciferase activities were used to normalize Renilla luciferase activity. All experiments were performed in triplicate and repeated at least three times.

Western blot analysis

Total protein was extracted from cells using RIPA lysis buffer (Beyotime, Shanghai, People's Republic of China) supplemented with a protease inhibitor cocktail (Sigma), according to the manufacturer's instruction. Bicinchoninic Acid Protein Assay kit (Beyotime, Shanghai, People's Republic of China) was performed to detect concentration of total protein. Equivalent amounts of proteins were separated on 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and subsequently transferred to polyvinylidene fluoride (PVDF) membranes (Millipore, Billerica, MA, USA). After that, the membranes were blocked with 5% fat-free milk for 2 h at room temperature and incubated with primary antibodies at 4°C overnight. The primary antibodies used in this study include mouse anti-human monoclonal PIK3R3 antibody (sc-376615; 1:1,000 dilution), mouse anti-human monoclonal p-AKT antibody (sc-271966; 1:1,000 dilution), mouse anti-human monoclonal AKT antibody (sc-56878; 1:1,000 dilution), mouse anti-human monoclonal p-mTOR ser 2481 antibody (sc-293132; 1:1,000 dilution), mouse anti-human monoclonal mTOR antibody (sc-293089; 1:1,000 dilution) and mouse anti-human monoclonal GAPDH antibody (sc-32233; 1:1,000 dilution; all Santa Cruz Biotechnology, CA, USA). Then, the membranes were washed three times with Tris-buffered saline containing Tween-20 (TBST) and further incubated with goat anti-mouse horseradish peroxidase-conjugated secondary antibody (sc-2005; 1:5,000 dilution; Santa Cruz Biotechnology) at room temperature for 1 h. ECL Protein Detection kit (Millipore) was used to visualize the proteins. Optical densities were analyzed with ImageJ software (NIH, Bethesda, MD, USA).

Statistical analysis

Data are presented as the mean ± standard error and analyzed with SPSS 17.0 (SPSS, Inc., Chicago, IL, USA) using Student's t-test or one way ANOVA test. Student-Newman-Keuls (SNK) was used to compare between two groups in multiple groups. P-value <0.05 was considered to indicate a statistically significant difference.

Results

MiR-212 is downregulated in CRC tissue specimens and cell lines

To investigate the status of miR-212 in CRC, RT-qPCR was used to analyse the expression levels of miR-212 in 28 pairs of CRC tissues and corresponding adjacent non-neoplastic tissues. MiR-212 was obviously downregulated in CRC tissues compared with adjacent non-neoplastic tissues (Fig. 1A, P<0.05). Then, the expression levels of miR-212 in CRC cell lines (HCT116, HT29, LoVo, SW480 and SW620) and normal human colon epithelium cell line (FHC) were examined. The expression levels of miR-212 significantly reduced in all CRC cell lines compared with that in the FHC cell line (Fig. 1B, P<0.05). Among the five CRC cell lines, HCT116 and SW620 showed the lowest miR-212 expression. Thus, HCT116 and SW620 cells were selected for further investigation.

MiR-212 inhibits the viability and invasion of CRC cells

To observe the functional role of miR-212 in CRC cells, HCT116 and SW620 cells were transfected with miR-212 mimics or miR-NC. After transfection, RT-qPCR analysis demonstrated that miR-212 was remarkably upregulated in the HCT116 and SW620 cells transfected with miR-212 mimics compared with the cells transfected with miR-NC (Fig. 2A, P<0.05). The role of miR-212 in CRC cell viability was then evaluated using MTT assay. As shown in Fig. 2B, miR-212 overexpression decreased the viability of HCT116 and SW620 cells compared with the miR-NC groups (P<0.05). In addition, Transwell invasion assay showed that miR-212 upregulation inhibited the invasion capacities of HCT116 and SW620 cells (Fig. 2C, P<0.05). Overall, these data indicate the anti-viability and anti-metastasis roles of miR-212 in CRC.

PIK3R3 is a novel target of miR-212 in CRC

MiRNAs exert functional roles mainly by base-pairing with the complementary sequence of their target genes (9). Hence, bioinformatics analysis was used to predict the potential target genes of miR-212. PIK3R3, which is associated with CRC tumorigenesis and development (22), was chosen for further confirmation (Fig. 3A). Then, PIk3R3 mRNA expression in CRC tissues and corresponding adjacent non-neoplastic tissues was detected using RT-qPCR. Results showed that the mRNA expression of PIK3R3 was dramatically upregulated in CRC tissues compared with adjacent non-neoplastic tissues (Fig. 3B, P<0.05). In addition, Spearman's correlation analysis revealed an inverse correlation between miR-212 and PIK3R3 mRNA expression (Fig. 3C, r=−0.6185, P<0.001).

The mRNA and protein expression levels of PIK3R3 in HCT116 and SW620 cells were measured after transfection with miR-212 mimics or miR-NC via RT-qPCR and Western blot analyses to investigate the negative regulation effects of miR-212 on endogenous PIK3R3 expression. Remarkable inhibition of PIK3R3 expression at both mRNA and protein levels was observed in the HCT116 and SW620 cells transfected with miR-212 mimics compared with those transfected with miR-NC (Fig. 3D and E, P<0.05). Moreover, luciferase reporter assay was conducted to identify the relationship between miR-212 and the 3′-UTR of PIK3R3. Results revealed that the restoration expression of miR-212 reduced the luciferase activities of pMIR-PIK3R3-3′-UTR Wt (Fig. 3F, P<0.05) but exerted no effect on the luciferase activities of pMIR-PIK3R3-3′-UTR Mut. Overall, these findings suggest that PIK3R3 is a novel target of miR-212 in CRC.

Upregulation of PIK3R3 reverses the tumor-suppressing effects of miR-212 on CRC cells

Rescue experiments were performed to investigate whether the tumor suppressive role of miR-212 in CRC is mediated by inhibiting the expression of PIK3R3. HCT116 and SW620 cells were transfected with miR-212 mimics in the presence or absence of pcDNA3.1-PIK3R3. Western blot analysis demonstrated that miR-212-induced PIK3R3 downregulation was rescued following co-transfection with pcDNA3.1-PIK3R3 (Fig. 4A, P<0.05). Moreover, PIK3R3 upregulation rescued the suppressive effects of miR-212 overexpression on the viability (Fig. 4B, P<0.05) and invasion (Fig. 4C, P<0.05) of HCT116 and SW620 cells. These results clearly show that miR-212 exerts tumor-suppressive roles in CRC, at least in part, by suppressing PIK3R3.

MiR-212 inactivates the AKT/mTOR signalling pathway

Previous studies demonstrated that PIK3R3 plays important roles in tumor formation and progression by regulating the AKT/mTOR signalling pathway (2325). Hence, Western blot analysis was performed to determine p-AKT, AKT, p-mTOR and mTOR expression in the HCT116 and SW620 cells transfected with miR-212 mimics or miR-NC. MiR-212 overexpression decreased p-AKT and p-mTOR expression, whereas transfection with miR-212 mimics did not affect total AKT and mTOR protein levels (Fig. 5). These results indicate that miR-212 inhibits CRC progression by directly targeting PIK3R3 and regulating the AKT/mTOR signalling pathway.

Discussion

Emerging evidence has shown that abnormal miRNA expression contributes to CRC carcinogenesis and progression by inhibiting the expression of their target genes (2628). Therefore, investigating the expression patterns and roles of miRNAs specifically involved in CRC formation and progression would greatly help expand our knowledge on CRC and provide novel therapeutic targets for CRC treatment. In the present study, miR-212 expression was significantly downregulated in CRC tissues and cell lines. In addition, increased miR-212 expression inhibited the viability and invasion of CRC cells in vitro. Furthermore, PIK3R3 was identified as a novel direct target of miR-212 in CRC. Importantly, miR-212 upregulation blocked the AKT/mTOR signalling pathway. Overall, the present study demonstrated that miR-212 expression was downregulated in CRC and suggested that miR-212 can inhibit CRC viability and invasion by directly targeting PIK3R3 and regulating the AKT/mTOR signalling pathway.

The dysregulation of miR-212 expression has occurred in several human cancers. For example, in gastric cancer, miR-212 is downregulated in tumor tissues and associated with UICC staging, lymph node invasion, live metastasis and peritoneal dissemination of gastric cancer patients. Kaplan-Meier analysis showed that patients with low miR-212 expression have significantly poor prognosis compared with patients with high miR-212 expression (29). In CRC, previous study found that expression level of miR-212 is lower in tumor tissues and significantly associated with a more aggressive tumor phenotype and short disease-free survival times (30). In breast cancer, miR-212 is lowly expressed in tumor tissues. Low miR-212 expression is correlated with the tumor grade of breast cancer (31). Downregulation of miR-212 is also observed in cervical cancer (18), intrahepatic cholangiocarcinoma (19), hepatocellular carcinoma (20), ovarian cancer (32), glioblastoma (33) and osteosarcoma (34). However, miR-212 is aberrantly highly expressed in pancreatic ductal adenocarcinoma. High expression levels of miR-212 are obviously correlated with the tumor size and stage of pancreatic ductal adenocarcinoma. Moreover, univariant analysis demonstrated that patients with high miR-212 expression exhibit poorer overall survival than patients with low miR-212 expression (35). Furthermore, miR-212 is upregulated in acute myeloid leukaemia and significantly correlated with a prolonged overall survival, event-free and relapse-free survival (36). These conflicting studies indicate that the expression levels of miR-212 in human cancers show tissue specificity and suggest that miR-212 could serve as a useful prognosis marker in human cancers.

MiR-212 is reportedly involved in the origination and progression of certain varieties of cancer. For instance, miR-212 upregulation inhibits the cell proliferation, invasion and epithelial-to-mesenchymal transition and decreases the G1/S phase transition of the cell cycle in cervical cancer (18,37). Meng et al (30) found that resumption expression of miR-212 attenuates CRC cell migration and invasion in vitro and formation of intrahepatic and pulmonary metastasis in vivo. Hanieh (38) demonstrated that miR-212 overexpression suppresses the migration and invasion of breast cancer cells. Dou et al (20) reported that increased miR-212 expression reduces cell viability and proliferation and induces apoptosis in hepatocellular carcinoma. Wei et al (32) showed that miR-212 serves tumor-suppressing roles in the growth and metastasis of ovarian cancer cells. Li et al (29) found that the ectopic expression of miR-212 attenuates the proliferation, invasion and metastasis of gastric cancer cells (39). Luo et al (34) revealed that the resumption expression of miR-212 represses the proliferation, invasion in vitro and growth in vivo of osteosarcoma cells. These findings indicate that miR-212 acts as a tumor suppressor in these human cancers. However, miR-212 plays oncogenic roles in pancreatic ductal adenocarcinoma by promoting cell growth and motility (40). These conflicting findings suggest that the roles of miR-212 in tumor initiation and development are tissue specific. This phenomenon could be explained by the ‘imperfect complementarity’ of the interactions between miRNAs and their target genes (41).

Multiple targets of miR-212 have been validated, including SMAD2 (37) and TCF7L2 (18) in cervical cancer, FOXA1 (19,20) in intrahepatic cholangiocarcinoma and hepatocellular carcinoma, SOX4 (38) in breast cancer, RFXAP (42) in pancreatic cancer, SKOV3 (32) in ovarian cancer, PTCH1 (40) in pancreatic ductal adenocarcinoma, SGK3 (33) in glioblastoma and SOX4 (34) in osteosarcoma. In the present study, PIK3R3 was validated as a novel target of miR-212 in CRC. PIK3R3, which is a member of the phosphatidylinositol 3-kinase (PI3K) family, is upregulated in ovarian cancer (43), gastric cancer (44), lung cancer (45) and breast cancer (46). Accumulating evidence suggested that PIK3R3 plays key regulatory roles in various cellular processes, including cell proliferation, cell differentiation, angiogenesis and metastasis (4648). In CRC, the expression levels of PIK3R3 were evaluated in clinical specimens and cell lines. In addition, low PIK3R3 expression correlates with CRC metastasis. Moreover, PIK3R3 upregulation promotes CRC cell metastasis both in vitro and in vivo (22). These findings indicate that targeting PIK3R3 in CRC may provide a novel strategy for the treatment of patients with this disease.

In conclusion, miR-212 acts as a tumor suppressor in CRC cell viability and invasion by directly targeting PIK3R3 and regulating the AKT/mTOR signalling pathway. These findings may provide new insights into the mechanisms underlying CRC formation and progression, as well as promising therapeutic strategies for CRC. In our following experiments, we will focus on the PIK3R3 expression in CRC using immunohistochemical and the effects of miR-212 on CRC in vivo.

References

1 

Jemal A, Siegel R, Xu J and Ward E: Cancer statistics, 2010. CA Cancer J Clin. 60:277–300. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Vargas AJ and Thompson PA: Diet and nutrient factors in colorectal cancer risk. Nutr Clin Pract. 27:613–623. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Meyerhardt JA and Mayer RJ: Systemic therapy for colorectal cancer. N Engl J Med. 352:476–487. 2005. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Juez I, Rubio C and Figueras J: Multidisciplinary approach of colorectal liver metastases. Clin Transl Oncol. 13:721–727. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993. View Article : Google Scholar : PubMed/NCBI

9 

Bushati N and Cohen SM: microRNA functions. Annu Rev Cell Dev Biol. 23:175–205. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Stahlhut C and Slack FJ: MicroRNAs and the cancer phenotype: Profiling, signatures and clinical implications. Genome Med. 5:1112013. View Article : Google Scholar : PubMed/NCBI

11 

Bueno MJ, Pérez de Castro I and Malumbres M: Control of cell proliferation pathways by microRNAs. Cell Cycle. 7:3143–3148. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Babashah S and Soleimani M: The oncogenic and tumour suppressive roles of microRNAs in cancer and apoptosis. Eur J Cancer. 47:1127–1137. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Fu Q, Zhang J, Xu X, Qian F, Feng K and Ma J: miR-203 is a predictive biomarker for colorectal cancer and its expression is associated with BIRC5. Tumour Biol. Oct 6–2016.(Epub ahead of print). View Article : Google Scholar :

15 

Shi C and Zhang Z: MicroRNA-362 is downregulated in cervical cancer and inhibits cell proliferation, migration and invasion by directly targeting SIX1. Oncol Rep. 37:501–509. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Wei W, Yang Y, Cai J, Cui K, Li RX, Wang H, Shang X and Wei D: MiR-30a-5p suppresses tumor metastasis of human colorectal cancer by targeting ITGB3. Cell Physiol Biochem. 39:1165–1176. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Zheng YB, Xiao K, Xiao GC, Tong SL, Ding Y, Wang QS, Li SB and Hao ZN: MicroRNA-103 promotes tumor growth and metastasis in colorectal cancer by directly targeting LATS2. Oncol Lett. 12:2194–2200. 2016.PubMed/NCBI

18 

Zhou C, Tan DM, Chen L, Xu XY, Sun CC, Zong LJ, Han S and Zhang YZ: Effect of miR-212 targeting TCF7L2 on the proliferation and metastasis of cervical cancer. Eur Rev Med Pharmacol Sci. 21:219–226. 2017.PubMed/NCBI

19 

Zhu L, Huang F, Deng G, Nie W, Huang W, Xu H, Zheng S, Yi Z and Wan T: MicroRNA-212 targets FOXA1 and suppresses the proliferation and invasion of intrahepatic cholangiocarcinoma cells. Exp Ther Med. 12:3790–3796. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Dou C, Wang Y, Li C, Liu Z, Jia Y, Li Q, Yang W, Yao Y, Liu Q and Tu K: MicroRNA-212 suppresses tumor growth of human hepatocellular carcinoma by targeting FOXA1. Oncotarget. 6:13216–13228. 2015. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Wang G, Yang X, Li C, Cao X, Luo X and Hu J: PIK3R3 induces epithelial-to-mesenchymal transition and promotes metastasis in colorectal cancer. Mol Cancer Ther. 13:1837–1847. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Yu T, Li J, Yan M, Liu L, Lin H, Zhao F, Sun L, Zhang Y, Cui Y, Zhang F, et al: MicroRNA-193a-3p and −5p suppress the metastasis of human non-small-cell lung cancer by downregulating the ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Oncogene. 34:413–423. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Cao G, Dong W, Meng X, Liu H, Liao H and Liu S: MiR-511 inhibits growth and metastasis of human hepatocellular carcinoma cells by targeting PIK3R3. Tumour Biol. 36:4453–4459. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Liu K, Li X, Cao Y, Ge Y, Wang J and Shi B: MiR-132 inhibits cell proliferation, invasion and migration of hepatocellular carcinoma by targeting PIK3R3. Int J Oncol. 47:1585–1593. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Li J, Du L, Yang Y, Wang C, Liu H, Wang L, Zhang X, Li W, Zheng G and Dong Z: MiR-429 is an independent prognostic factor in colorectal cancer and exerts its anti-apoptotic function by targeting SOX2. Cancer Lett. 329:84–90. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Zheng GX, Qu AL, Yang YM, Zhang X, Zhang SC and Wang CX: miR-422a is an independent prognostic factor and functions as a potential tumor suppressor in colorectal cancer. World J Gastroenterol. 22:5589–5597. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Li P, Xue WJ, Feng Y and Mao QS: MicroRNA-205 functions as a tumor suppressor in colorectal cancer by targeting cAMP responsive element binding protein 1 (CREB1). Am J Transl Res. 7:2053–2059. 2015.PubMed/NCBI

29 

Li D, Li Z, Xiong J, Gong B, Zhang G, Cao C, Jie Z, Liu Y, Cao Y, Yan Y, et al: MicroRNA-212 functions as an epigenetic-silenced tumor suppressor involving in tumor metastasis and invasion of gastric cancer through down-regulating PXN expression. Am J Cancer Res. 5:2980–2997. 2015.PubMed/NCBI

30 

Meng X, Wu J, Pan C, Wang H, Ying X, Zhou Y, Yu H, Zuo Y, Pan Z, Liu RY and Huang W: Genetic and epigenetic down-regulation of microRNA-212 promotes colorectal tumor metastasis via dysregulation of MnSOD. Gastroenterology. 145:426–436. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Damavandi Z, Torkashvand S, Vasei M, Soltani BM, Tavallaei M and Mowla SJ: Aberrant expression of breast development-related MicroRNAs, miR-22, miR-132, and miR-212, in breast tumor tissues. J Breast Cancer. 19:148–155. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Wei LQ, Liang HT, Qin DC, Jin HF, Zhao Y and She MC: MiR-212 exerts suppressive effect on SKOV3 ovarian cancer cells through targeting HBEGF. Tumour Biol. 35:12427–12434. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Liu H, Li C, Shen C, Yin F, Wang K, Liu Y, Zheng B, Zhang W, Hou X, Chen X, et al: MiR-212-3p inhibits glioblastoma cell proliferation by targeting SGK3. J Neurooncol. 122:431–439. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Luo XJ, Tang DG, Gao TL, Zhang YL, Wang M, Quan ZX and Chen J: MicroRNA-212 inhibits osteosarcoma cells proliferation and invasion by down-regulation of Sox4. Cell Physiol Biochem. 34:2180–2188. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Wu Z, Zhou L, Ding G and Cao L: Overexpressions of miR-212 are associated with poor prognosis of patients with pancreatic ductal adenocarcinoma. Cancer Biomark. 18:35–39. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Sun SM, Rockova V, Bullinger L, Dijkstra MK, Döhner H, Löwenberg B and Jongen-Lavrencic M: The prognostic relevance of miR-212 expression with survival in cytogenetically and molecularly heterogeneous AML. Leukemia. 27:100–106. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Zhao JL, Zhang L, Guo X, Wang JH, Zhou W, Liu M, Li X and Tang H: miR-212/132 downregulates SMAD2 expression to suppress the G1/S phase transition of the cell cycle and the epithelial to mesenchymal transition in cervical cancer cells. IUBMB Life. 67:380–394. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Hanieh H: Aryl hydrocarbon receptor-microRNA-212/132 axis in human breast cancer suppresses metastasis by targeting SOX4. Mol Cancer. 14:1722015. View Article : Google Scholar : PubMed/NCBI

39 

Jiping Z, Ming F, Lixiang W, Xiuming L, Yuqun S, Han Y, Zhifang L, Yundong S, Shili L, Chunyan C and Jihui J: MicroRNA-212 inhibits proliferation of gastric cancer by directly repressing retinoblastoma binding protein 2. J Cell Biochem. 114:2666–2672. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Ma C, Nong K, Wu B, Dong B, Bai Y, Zhu H, Wang W, Huang X, Yuan Z and Ai K: miR-212 promotes pancreatic cancer cell growth and invasion by targeting the hedgehog signaling pathway receptor patched-1. J Exp Clin Cancer Res. 33:542014. View Article : Google Scholar : PubMed/NCBI

41 

Jackson RJ and Standart N: How do microRNAs regulate gene expression? Sci STKE 2007. re12007.

42 

Ding G, Zhou L, Qian Y, Fu M, Chen J, Chen J, Xiang J, Wu Z, Jiang G and Cao L: Pancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3p. Oncotarget. 6:29877–29888. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Zhang L, Huang J, Yang N, Greshock J, Liang S, Hasegawa K, Giannakakis A, Poulos N, O'Brien-Jenkins A, Katsaros D, et al: Integrative genomic analysis of phosphatidylinositol 3′-kinase family identifies PIK3R3 as a potential therapeutic target in epithelial ovarian cancer. Clin Cancer Res. 13:5314–5321. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Zhou J, Chen GB, Tang YC, Sinha RA, Wu Y, Yap CS, Wang G, Hu J, Xia X, Tan P, et al: Genetic and bioinformatic analyses of the expression and function of PI3K regulatory subunit PIK3R3 in an Asian patient gastric cancer library. BMC Med Genomics. 5:342012. View Article : Google Scholar : PubMed/NCBI

45 

Xu L, Wen Z, Zhou Y, Liu Z, Li Q, Fei G, Luo J and Ren T: MicroRNA-7-regulated TLR9 signaling-enhanced growth and metastatic potential of human lung cancer cells by altering the phosphoinositide-3-kinase, regulatory subunit 3/Akt pathway. Mol Biol Cell. 24:42–55. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Klahan S, Wu MS, Hsi E, Huang CC, Hou MF and Chang WC: Computational analysis of mRNA expression profiles identifies the ITG family and PIK3R3 as crucial genes for regulating triple negative breast cancer cell migration. Biomed Res Int. 2014:5365912014. View Article : Google Scholar : PubMed/NCBI

47 

Xia X, Cheng A, Akinmade D and Hamburger AW: The N-terminal 24 amino acids of the p55 gamma regulatory subunit of phosphoinositide 3-kinase binds Rb and induces cell cycle arrest. Mol Cell Biol. 23:1717–1725. 2003. View Article : Google Scholar : PubMed/NCBI

48 

Soroceanu L, Kharbanda S, Chen R, Soriano RH, Aldape K, Misra A, Zha J, Forrest WF, Nigro JM, Modrusan Z, et al: Identification of IGF2 signaling through phosphoinositide-3-kinase regulatory subunit 3 as a growth-promoting axis in glioblastoma. Proc Natl Acad Sci USA. 104:3466–3471. 2007; View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang J, Zhang Y, Li X, Wang H, Li Q and Liao X: MicroRNA‑212 inhibits colorectal cancer cell viability and invasion by directly targeting PIK3R3 Retraction in /10.3892/mmr.2023.13025. Mol Med Rep 16: 7864-7872, 2017
APA
Zhang, J., Zhang, Y., Li, X., Wang, H., Li, Q., & Liao, X. (2017). MicroRNA‑212 inhibits colorectal cancer cell viability and invasion by directly targeting PIK3R3 Retraction in /10.3892/mmr.2023.13025. Molecular Medicine Reports, 16, 7864-7872. https://doi.org/10.3892/mmr.2017.7552
MLA
Zhang, J., Zhang, Y., Li, X., Wang, H., Li, Q., Liao, X."MicroRNA‑212 inhibits colorectal cancer cell viability and invasion by directly targeting PIK3R3 Retraction in /10.3892/mmr.2023.13025". Molecular Medicine Reports 16.5 (2017): 7864-7872.
Chicago
Zhang, J., Zhang, Y., Li, X., Wang, H., Li, Q., Liao, X."MicroRNA‑212 inhibits colorectal cancer cell viability and invasion by directly targeting PIK3R3 Retraction in /10.3892/mmr.2023.13025". Molecular Medicine Reports 16, no. 5 (2017): 7864-7872. https://doi.org/10.3892/mmr.2017.7552