Function of miR-212 as a tumor suppressor in thyroid cancer by targeting SIRT1

Retraction in: /10.3892/or.2021.8173

  • Authors:
    • Dandan Li
    • Lin Bai
    • Tongtong Wang
    • Qian Xie
    • Minglong Chen
    • Yantao Fu
    • Qiang Wen
  • View Affiliations

  • Published online on: November 28, 2017     https://doi.org/10.3892/or.2017.6119
  • Pages: 695-702
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

miR-212 as a tumor suppressor has been reported to be downregulated in multiple cancer cells lines and tumor tissues. However, its role in thyroid cancer has nor been investigated. Therefore, the present study aimed to investigate the role of miR-212 in human thyroid cancer and the underlying mechanisms. In the present study, we demonstrated that miR-212 expression was significantly decreased in thyroid cancer specimens and cell lines compared with adjacent normal tissues and normal thyroid cell lines. In addition, we demonstrated that miR-212 downrwegulation in thyroid cancer tissues was negatively associated with lymph node metastasis and advanced clinical stage. Functionally, ectopic expression of miR-212 by transfection with miR-212 mimic significantly inhibited proliferation, colony formation, migration and invasion in TPC-1 cells. In addition, Sirtuin 1 (SIRT1) was identified as a direct target of miR-212 and its expression was inversely correlated with miR-212 expression in thyroid cancer tissues. Overexpression of SIRT1 could effectively rescue miR-212 mimic-induced suppression of cell proliferation, migration and invasion in TPC-1 cells. In vivo, miR-212 overexpression significantly inhibited tumor growth in a nude mice model. In light of these findings, miR-212 may function as a tumor suppressor in thyroid cancer by targeting SIRT1.

Introduction

Thyroid cancer is the most common endocrine malignancy worldwide with a rapidly increasing incidence and prevalence over the past 20 years (1). Despite the fact that thyroid cancer patients in early stages have a favorable prognosis with high overall 5-year survival rate, patients with advanced thyroid cancer have a 5-year survival rate of only ~59% (2,3). Therefore, elucidating the molecular mechanisms underlying thyroid cancer is required in order to develop novel therapeutic strategies for this disease.

MicroRNAs (miRNAs) are a class of endogenous small RNAs containing ~22 nucleotides that disturb the protein translation process by targeting the sequences on their 3′untranslated region (3′UTR) (46). By negatively regulating the protein expression levels of their target genes, miRNAs play central roles in tumor initiation, development and progression (7,8). Since protein expression is dysregulated in various human cancers, including thyroid cancers and some miRNAs act as oncogenes or tumor suppressors in thyroid cancer, miRNAs may serve as potential molecular targets or candidates for the treatment of thyroid cancer (9,10).

miR-212, a tumor associated miRNA, has been reported to play a suppressive role in many types of cancer, including gastric (11), hepatocellular carcinoma (12), bladder (13), non-small cell lung (14), ovarian (15) and cervical cancer (16). However, to the best of our knowledge, the role and molecular mechanism of miR-212 in thyroid cancer has not been determined. The present study aimed to investigate the expression levels of miR-212 in thyroid cancer tissues and cell lines, as well as its clinical significance. Furthermore, the present study examined the role of miR-212 in the regulation of thyroid cancer cell growth and invasion, as well as the underlying regulatory mechanism of its action by in vitro and in vivo experiments.

Materials and methods

Patients and tissue samples

Primary thyroid cancer samples and their matched non-cancerous tissues (normal) were obtained from 42 patients who underwent surgery of thyroid cancer at China-Japan Union Hospital of Jilin University (Changchun, China). Following surgery all samples were snap-frozen in liquid nitrogen and stored at −80°C until RNA extraction. Patients who underwent chemotherapy, radiotherapy or other treatment before the operation were excluded. The study was approved by the Research Ethics Committee of China-Japan Union Hospital of Jilin University and informed consents was obtained from all patients.

Cell lines and transfection

Three human thyroid cancer cell lines (TPC-1, BCPAP and SW1736) and human thyroid follicular epithelial cells (Nthy-ori3-1) were obtained from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) and were grown in Dulbeccos modified Eagles medium (DMEM; Gibco, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS; Gibco) at 37°C in a humidified chamber supplemented with 5% CO2.

miR-212 mimic or negative control mimic (miR-NC) were designed and synthesized by Shanghai GenePharma Co., Ltd. (Shanghai, China). Overexpression SIRT1 plasmid (pCDNA3.1-SIRT1) was granted by Dr Peng Li (Jilin University, Changchun, China). These molecules were transiently transfected into TPC-1 cell using Lipofectamine™ 2000 (Invitrogen, Waltham, MA, USA) according to the manufacturers protocol. After 24–72 h transfection, the cells were harvested for further analysis.

Real-time polymerase chain reaction analysis of miR-212 and SIRT1 mRNA expression

Total RNA including miRNAs was isolated from cultured cells or tissues, using miRNeasy Mini kit (Qiagen, Hilden, Germany). Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed in ABI PRISM 7000 Sequence Detection System (Applied Biosystems, Foster City, CA, USA) using QuantiTect SYBR-Green PCR kit (Qiagen). The primers of miR-212 and U6 were purchased from Qiagen. U6 RNA was used as an internal control.

For the detection of the SIRT1 expression, total RNA was extracted from cultured cells and tissues using TRIzol reagent (Life Technologies, Carlsbad, CA, USA). RNA was reverse-transcribed into cDNA using M-MLV reverse transcriptase kits (Promega, Madison, WI, USA), and then quantified using SYBR Premix Ex Taq (Takara, Dalian, China) under ABI PRISM 7000 Sequence Detection system. The primers of SIRT1 and GAPDH were used in this study as previously described (17). GAPDH was used as an internal control. The 2−∆∆Ct method was used to analyze and quantify the transcription levels.

Proliferation assay

The proliferation of thyroid cancer cell was assessed by the Cell Counting Kit-8 (CCK-8) assay kit (Dojindo Laboratories, Kumamoto, Japan). Briefly, ~1×104 transfected cells were seeded in each well of 96-well plates and cultured for 24–72 h. At indicated time-points (24, 48 and 72 h), 10 µl of CCK-8 was added to each well. After incubation at 37°C for 4 h, the absorbance was detected at 450 nm by a microplate reader (Bio-Tek Instruments, Winooski, VT, USA).

Colony formation assay

Transfected cells were digested with trypsin and suspended into a single cell status. A total of 1,000 cells from each group were seeded in 6-well plates and cultured in DMEM medium with 10% FBS for 14 days. The colonies were fixed with 4% paraformaldehyde for 20 min and stained with 0.1% crystal violet for 15 min. The total number of colonies was counted under a light microscope (Olympus, Tokyo, Japan).

Wound healing assay

Wound healing assays were performed to assess the motility of indicated cells. Briefly, the transfected cells were seeded in 6-well plates and allowed to reach to 90–95% confluency. The cells were scratched with a sterile plastic micropipette tip in the cell monolayer. After wounding, the cells were cultured in complete DMEM medium with 10% FBS for 24 h. Wound closure was observed at 0 and 24 h, and photographed under a light microscope (Olympus).

Transwell invasion assay

The invasive ability of indicated cells was analyzed by Transwell (Corning Costar Corp., Cambridge, MA, USA) assay. In brief, 2×105 transfected cells suspended in serum-free medium were added to each upper chamber pre-coated with Matrigel matrix and 500 µl of DMEM medium containing 10% FBS were added to the lower chamber as a chemoattractant. After 48-h incubation, the remaining cells in the upper chambers were removed with a cotton swab and the invasive cells on the lower membrane surface were fixed with 4% paraformaldehyde, stained with 0.1% crystal violet. Invasive cells were photographed and quantified by counting them in five random fields using a light microscope (Olympus).

Luciferase assay

The human SIRT1 3UTR oligonucleotides containing the wild-type (WT) or mutant-type (MT) miR-212 binding site were amplified by PCR and inserted downstream of the luciferase gene in the pGL3-luciferase reporter plasmid (Ambion, Austin, TX, USA). TPC-1 cells seeded in 96-well plates in triplicate were cotransfected with WT/MT-SIRT1-3′UTR reported plasmid and miR-212 mimic or miR-NC using Lipofectamine 2000 according to the manufacturers protocol. Forty-eight hours after transfection, the cells were harvested and luciferase activity was assessed using a Dual-Luciferase reporter assay kit (Promega).

Western blotting

The protein was extracted by lysing cells in ice-cold radioimmunoprecipitation assay buffer (Nanjing KeyGen Biotech, Co., Ltd., Nanjing, China). The protein concentration was quantified with the bicinchoninic acid (BCA) kit (Pierce, Rockford, IL, USA). Equivalent amounts of protein samples (20 µg each lane) were separated on 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto a polyvinylidene fluoride membrane (Millipore, Billerica, MA, USA), which was then blocked for 1 h with 5% non-fat milk in PBST. After incubation overnight with antibodies against SIRT1 and GAPDH (dilution 1:2,000; both antibodies from Santa Cruz Biotechnology, Inc., Santa Cruz, CΑ, USA) at 4°C, the membranes were incubated with horseradish peroxidase-conjugated (HRP) goat-anti-mouse secondary antibody at room temperature for 2 h. The protein bands were observed using chemiluminescence (Thermo Fisher Scientific, Waltham, MA, USA).

In vivo tumorigenesis assay

All animal studies were approved by the Institutional Animal Care and Use Committee of Jilin University (Changchun, China). Twenty female BALB/C nude mice (18–20 g, 6–7 weeks old) were obtained from the Experimental Animal Center of Jilin University (Changchun, China) and maintained under specific pathogen-free conditions at Jilin University.

For the in vivo tumorigenesis assay, ~2×106 TPC-1 cells stably expressing miR-212 or miR-NC were suspended in 100 µl of phosphate buffered saline (PBS), and then injected subcutaneously in the left posterior flank of the BALB/c-nude mice (10 mice in each group). After ten days, the tumor growth was assessed and recorded by measuring tumor length (L) and width (W) every five days until the nude mice were sacrificed. Tumor volume was calculated according to the formula: V = 1/2 × L × W2. The mice were sacrificed and photographed at 30 days post-implantation. Xenograft tumors were excised, photographed, weighed and stored at −80°C for further analysis.

Statistical analysis

All statistical analyses were performed using the SPSS 19.0 software (SPSS, Inc., Chicago, IL, USA). Experimental data are presented as the mean ± standard error (SE) at least from three independent experiments. The two-tailed Students t-test was adopted for comparison between the two groups and one-way ANOVA was used for comparisons of more than two groups. A P-value of 0.05 was considered to indicate a statistically significant difference.

Results

miR-212 is downregulated in thyroid cancer tissues and cell lines

To determine the expression of miR-212 in thyroid cancer, we first detected miR-212 expression level by qRT-PCR in human thyroid cancer tissues and adjacent normal thyroid tissues. As depicted in Fig. 1A, miR-212 expression was significantly downregulated in human thyroid cancer tissues compared with adjacent normal tissues. In addition, we found that the expression levels of miR-212 in advanced clinical stage (III–IV) were significantly downregulated compared with those in low clinical stage (TNM stage I and II) (Fig. 1B). Consistent with the above mentioned results, miR-212 levels in tissues with lymph node metastases were markedly decreased compared to the tissues without lymph node metastases (Fig. 1C). In addition, we investigated the expression of miR-212 in three thyroid cancer cell lines (TPC-1, BCPAP and SW1736), using the human thyroid epithelial cell line Nthy-ori3-1 as a control. We found that miR-212 was downregulated in thyroid cancer cell lines compared with the human thyroid epithelial cell line (Fig. 1D). In particular, TPC-1 cell line exhibited the lowest levels of miR-212 expression and was used for subsequent studies (Fig. 1D). These results indicated that low miR-212 may be associated with thyroid cancer progression.

miR-212 inhibits thyroid cancer cell proliferation, migration and invasion

To explore the possible biological functions of miR-212 in thyroid cancer cells, we transfected TPC-1 cells with miR-212 mimic or negative controls (miR-NC) to enhance miR-212 expression. As shown in Fig. 2A, cells transfected with miR-212 mimic significantly increased miR-212 expression levels compared to cells transfected with miR-NC. CCK-8 assays demonstrated that miR-212 overexpression significantly inhibited thyroid cancer cell proliferation (Fig. 2B). In addition, examined the colony formation capacity of TPC-1 cells and observed that miR-212 overexpression significantly inhibited thyroid cancer cell colony formation (Fig. 2C). To investigate the effect of miR-212 on cellular motility, the migration and invasion ability of TPC-1 cells after modification of miR-212 expression were determined by wound healing and Transwell invasion assays, respectively. It was observed that miR-212 overexpression significantly inhibited the migration and invasion of TPC-1 cells (Fig. 2D and E). Collectively, these results indicated that miR-212 may impede thyroid cancer cell proliferation, migration and invasion in vitro.

SIRT1 is a direct target of miR-212 in thyroid cancer cells

To fully understand the mechanism of miR-212 inhibition of human thyroid cancer progression, three bioinformatic databases (TargetScan, miRanda and PicTar) were used to predict the targets of miR-212. We identified the 3′-UTR of SIRT1 that were able to bind to the ‘seed region’ of miR-212 (Fig. 3A). To determine whether SIRT1 is a target of miR-212, the luciferase activity assay was performed. As excepted, miR-212 bound to SIRT1 3′-UTR, resulting in markedly reduced luciferase activities (Fig. 3B). In addition, we also found that miR-212 overexpression obviously decreased SIRT1 expression on mRNA expression and protein levels (Fig. 3C and D). These results indicated that miR-212 directly targets SIRT1 by binding its seed region of the 3′-UTR region in human thyroid cancer cells.

Inverse correlation between SIRT1 and miR-212 expression in thyroid cancer

Subsequently, we examined the SIRT1 mRNA in 42 pairs of thyroid cancer tissue specimens and adjacent normal tissues by qRT-PCR. The SIRT1 expression was higher in thyroid cancer specimens than that of adjacent normal thyroid tissues (Fig. 4A). The inverse correlation between miR-212 and SIRT1 mRNA levels was further confirmed by Pearson correlation analysis in 42 thyroid cancer tissues (r=−0.486, P=0.001; Fig. 4B). Furthermore, the SIRT1 expression on mRNA and protein levels was increased in thyroid cancer cell lines compared to the normal thyroid cells (Fig. 4C and D).

miR-212 exerts its suppressive function by targeting SIRT1 in thyroid cancer cells

To examine whether miR-212 exerted its suppressive function through its target gene SIRT1, we rescued the expression of SIRT1 in miR-212 mimic-transfected cells. qRT-PCR and western blot assays revealed that transfection of SIRT1 overexpression plasmid in miR-212 mimic-transfected cells restored the SIRT1 expression in TPC-1 cells (Fig. 5A and B). Furthermore, restoration of SIRT1 expression partially reversed the inhibition effect on cell proliferation, colony formation, migration and invasion in TPC-1 cells mediated by miR-212 (Fig. 5C-F). These results indicated that miR-212 impaired cell growth, migration and invasion of TPC-1 by targeting SIRT1.

miR-212 suppresses tumor growth in vivo

To determine the effects of miR-212 on tumorigenicity in vivo, TPC-1 cells transfected with miR-212 mimic or miR-NC were injected into the flanks of nude mice to form ectopic tumors. We found that tumor growth was slower in the TPC-1/miR-212 group than that in the TPC-1/miR-NC group (Fig. 6A). Consistent with the tumor growth curve, the tumor size and weight in the TPC-1/miR-212 group were significantly decreased compared to TPC-1/miR-NC group (Fig. 6B and C). We also analyzed the expression of miR-212 and SIRT1 in xenograft tumors. In the TPC-1/miR-212 group, miR-212 expression was upregulated (Fig. 6D), whereas SIRT1 expression was downregulated both on mRNA level (Fig. 6E) and protein level (Fig. 6F). These data indicated that miR-212 suppresses tumor growth in vivo by suppressing SIRT1.

Discussion

miRNAs have been found to play crucial roles in the carcinogenesis in various types of cancers (18). In line with this notion, miRNAs participation in thyroid cancer progression has been widely reported (9,10). In the present study, we found that miR-212 was downregulated in both thyroid cancer tissues and thyroid cancer cell lines and that decreased miR-212 was associated with lymph node metastasis and clinical stage. We also found that miR-212 overexpression by transfection with miR-212 mimic significantly inhibited thyroid cancer cell proliferation, colony formation, migration and invasion in vitro. In vivo, miR-212 overexpression inhibited tumor growth in nude mice model. These results may provide evidence for using miR-212 as a novel target for treating thyroid cancer.

miR-212 has been revealed to be downregulated and function as tumor suppressor in thr majority of types of cancers by regulating different oncogene (1115). For example, Fu et al (19) revealed that miR-212 may act as tumor suppressor in prostate cancer progression through disrupting epithelial to mesenchymal transition (EMT) process by directly targeting SOX4. Jiping et al (11) reported that miR-212 functions as a tumor suppressor involved in tumor metastasis and invasion of gastric cancer through suppressing paxillin (PXN) expression. Zhao et al (20) demonstrated that miR-212 delayed cell arrest in the G1/S phase transition and suppressed cell proliferation, as well as EMT migration and invasion in cervical cancer cell by targeting SMAD2. In contrast, recently a study revealed that miR-212 facilitated pancreatic cancer cell growth and invasion by targeting the hedgehog signaling pathway receptor patched-1 (21). Thus, the biological role of miR-212 in carcinogenesis seems to be complicated and highly tissue-specific. In the present study, we investigated miR-212 expression in thyroid cancer tissues and cell lines by qRT-PCR and found that miR-212 was downregulated in thyroid cancer tissues and cell lines. Furthemore, miR-212 expression was significantly downregulated in patients with advanced clinical stage and lymph node metastasis. Thus, we hypothesized that miR-212 overexpression may suppress the malignant phenotypes of thyroid cancer cells. As excepted, our further results revealed that ectopic miR-212 expression suppressed thyroid cancer cell growth, migration and invasion in vitro, as well as suppressed tumor growth in vivo. Altogether, both clinical and experimental data indicated a tumor suppressive role of miR-212 in thyroid cancer.

It is well known that miRNAs can act as tumor suppressors by targeting specific oncogenes (22). Thus, three bioinformatic databases (TargetScan, miRanda and PicTar) were used to predict targets of miR-212. Sirtuin 1 (SIRT1), an known oncogene, was selected as a target gene of miR-212. SIRT1 is a member of the sirtuin (SIRT) family that exerts multiple cellular functions and is conserved from bacteria to eukaryotes (23). SIRT1 expression has been reported to be higher in numerous human cancer cell lines and tissues including thyroid cancer (24). SIRT1 has been implicated in the cell cycle, as well as apoptosis and cancer metastasis by regulating its substrates such as Myc, p53, nuclear factor-κB, Ku70 and forkhead transcription factor (25,26). Recently, a study revealed that inhibition of SIRT1 expression impaired proliferation and induced cell apoptosis and cell cycle arrest in thyroid cancer cell lines (27), indicating SIRT1 as an oncogene in thyroid cancer. SIRT1 has been reported to be a target of miR-212 in prostate cancer (28), however, the interaction between miR-212 and SIRT1 has not been experimentally validated in thyroid cancer. In the present study, using luciferase reporter assays, qRT-PCR and western blot assays, we verified the SIRT1 gene as a direct target of miR-212 in thyroid cancer. In addition, SIRT1 expression was upregulated in thyroid cancer tissues and was negatively correlated with the expression level of miR-212. SIRT1 overexpression reversed the inhibition effect on cell proliferation, migration and invasion in thyroid cancer cells induced by miR-212 overexpression. In vivo, miR-212 also displayed an inhibitory role in thyroid cancer growth by suppressing SIRT1. These findings indicated that miR-212 impaired thyroid cancer development via repressing SIRT1.

In conclusion, this study first demonstrated that miR-212 is downregulated in thyroid cancer tissues and cell lines and functions as a tumor suppressor in thyroid cancer cell growth by downregulating SIRT1. Thus, miR-212/SIRT1 may provide a promising therapeutic strategy for the treatment of thyroid cancer.

Acknowledgements

The present study was supported by the Research Fund of Science and Technology Department of Jilin Province (20160101064JC) and the Jilin University Funding Project for Young Teacher Cultivation Plan (419080500365).

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Pemayun TG: Current diagnosis and management of thyroid nodules. Acta Med Indones. 48:247–257. 2016.PubMed/NCBI

3 

Liu S, Semenciw R, Ugnat AM and Mao Y: Increasing thyroid cancer incidence in Canada, 1970–1996: Time trends and age-period-cohort effects. Br J Cancer. 85:1335–1339. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Valinezhad Orang A, Safaralizadeh R and Kazemzadeh-Bavili M: Mechanisms of miRNA-mediated gene regulation from common downregulation to mRNA-specific upregulation. Int J Genomics. 2014:9706072014. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Hwang HW and Mendell JT: MicroRNAs in cell proliferation, cell death, and tumorigenesis. Br J Cancer. 96 Suppl:R40–R44. 2007.PubMed/NCBI

7 

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

8 

Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, et al: A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 103:pp. 2257–2261. 2006; View Article : Google Scholar : PubMed/NCBI

9 

de la Chapelle A and Jazdzewski K: MicroRNAs in thyroid cancer. J Clin Endocrinol Metab. 96:3326–3336. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Leonardi GC, Candido S, Carbone M, Colaianni V, Garozzo SF, Cinà D and Libra M: microRNAs and thyroid cancer: Biological and clinical significance (Review). Int J Mol Med. 30:991–999. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Jiping Z, Ming F, Lixiang W, Xiuming L, Yuqun S, Han Y, Zhifang L, Yundong S, Shili L, Chunyan C, et al: 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

12 

Tu H, Wei G, Cai Q, Chen X, Sun Z, Cheng C, Zhang L, Feng Y, Zhou H, Zhou B, et al: MicroRNA-212 inhibits hepatocellular carcinoma cell proliferation and induces apoptosis by targeting FOXA1. Onco Targets Ther. 8:2227–2235. 2015.PubMed/NCBI

13 

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

14 

Jiang X, Chen X, Chen L, Ma Y, Zhou L, Qi Q, Liu Y, Zhang S, Luo J and Zhou X: Upregulation of the miR-212/132 cluster suppresses proliferation of human lung cancer cells. Oncol Rep. 33:705–712. 2015. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

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

17 

Zhang S, Zhang D, Yi C, Wang Y, Wang H and Wang J: MicroRNA-22 functions as a tumor suppressor by targeting SIRT1 in renal cell carcinoma. Oncol Rep. 35:559–567. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Tomasetti M, Amati M, Santarelli L and Neuzil J: MicroRNA in metabolic re-programming and their role in tumorigenesis. Int J Mol Sci. 17:172016. View Article : Google Scholar

19 

Fu W, Tao T, Qi M, Wang L, Hu J, Li X, Xing N, Du R and Han B: MicroRNA-132/212 upregulation inhibits TGF-β-mediated epithelial-mesenchymal transition of prostate cancer cells by targeting SOX4. Prostate. 76:1560–1570. 2016. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

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

22 

Kinose Y, Sawada K, Nakamura K and Kimura T: The role of microRNAs in ovarian cancer. BioMed Res Int. 2014:2493932014. View Article : Google Scholar : PubMed/NCBI

23 

Correia M, Perestrelo T, Rodrigues AS, Ribeiro MF, Pereira SL, Sousa MI and Ramalho-Santos J: Sirtuins in metabolism, stemness and differentiation. Biochim Biophys Acta. 1861:3444–3455. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Herranz D, Maraver A, Cañamero M, Gómez-López G, Inglada-Pérez L, Robledo M, Castelblanco E, Matias-Guiu X and Serrano M: SIRT1 promotes thyroid carcinogenesis driven by PTEN deficiency. Oncogene. 32:4052–4056. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Deng CX: SIRT1, is it a tumor promoter or tumor suppressor? Int J Biol Sci. 5:147–152. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Palmirotta R, Cives M, Della-Morte D, Capuani B, Lauro D, Guadagni F and Silvestris F: Sirtuins and cancer: Role in the epithelial-mesenchymal transition. Oxid Med Cell Longev. 2016:30314592016. View Article : Google Scholar : PubMed/NCBI

27 

Wu W, Zhang L, Lin J, Huang H, Shi B, Lin X, Huang Z, Wang C, Qiu J and Wei X: Hypermethylation of the HIC1 promoter and aberrant expression of HIC1/SIRT1 contribute to the development of thyroid papillary carcinoma. Oncotarget. 7:84416–84427. 2016.PubMed/NCBI

28 

Ramalinga M, Roy A, Srivastava A, Bhattarai A, Harish V, Suy S, Collins S and Kumar D: MicroRNA-212 negatively regulates starvation induced autophagy in prostate cancer cells by inhibiting SIRT1 and is a modulator of angiogenesis and cellular senescence. Oncotarget. 6:34446–34457. 2015.PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 39 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li D, Bai L, Wang T, Xie Q, Chen M, Fu Y and Wen Q: Function of miR-212 as a tumor suppressor in thyroid cancer by targeting SIRT1 Retraction in /10.3892/or.2021.8173. Oncol Rep 39: 695-702, 2018
APA
Li, D., Bai, L., Wang, T., Xie, Q., Chen, M., Fu, Y., & Wen, Q. (2018). Function of miR-212 as a tumor suppressor in thyroid cancer by targeting SIRT1 Retraction in /10.3892/or.2021.8173. Oncology Reports, 39, 695-702. https://doi.org/10.3892/or.2017.6119
MLA
Li, D., Bai, L., Wang, T., Xie, Q., Chen, M., Fu, Y., Wen, Q."Function of miR-212 as a tumor suppressor in thyroid cancer by targeting SIRT1 Retraction in /10.3892/or.2021.8173". Oncology Reports 39.2 (2018): 695-702.
Chicago
Li, D., Bai, L., Wang, T., Xie, Q., Chen, M., Fu, Y., Wen, Q."Function of miR-212 as a tumor suppressor in thyroid cancer by targeting SIRT1 Retraction in /10.3892/or.2021.8173". Oncology Reports 39, no. 2 (2018): 695-702. https://doi.org/10.3892/or.2017.6119