Open Access

MicroRNA‑30a suppresses papillary thyroid cancer cell proliferation, migration and invasion by directly targeting E2F7

  • Authors:
    • Haiyan Guo
    • Linyun Zhang
  • View Affiliations

  • Published online on: April 30, 2019     https://doi.org/10.3892/etm.2019.7532
  • Pages: 209-215
  • Copyright: © Guo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

microRNA (miRNA/miR)‑30a, a tumor‑associated miRNA, has been implicated in the tumorigenesis and progression of different types of human cancer. Thyroid cancer is a common endocrine malignancy, of which papillary thyroid cancer (PTC) accounts for ~80‑90% of all TC. However, the effect of miR‑30a in PTC is yet to be fully elucidated. The TPC‑1 human PTC cell line, as well as the normal human thyroid cell line (HT‑ori3), were utilized in the current study. The PTC cell line was transfected with a miR‑30a mimic. Subsequently, reverse transcription‑quantitative polymerase chain reaction was performed to detect the expression of miR‑30a and E2F transcription factor 7 (E2F7). Cell proliferation was assessed via a MTT assay and transwell migration and invasion assays were performed to detect the migration and invasion of PTC cells. A dual‑luciferase reporter assay was also utilized to clarify the association between miR‑30a and E2F7. The results of the current study revealed that miR‑30a was significantly downregulated in TPC‑1 cells compared with HT‑ori3 cells and that the expression of E2F7 was significantly upregulated in PTC cells. The upregulation of miR‑30a also inhibited the proliferation, migration and invasion of PTC cells. Furthermore, the luciferase assay revealed that miR‑30a binds to the 3'‑UTR of E2F7. Additionally, the overexpression of miR‑30a decreased E2F7 levels in TPC‑1 cells. These results indicate that miR‑30a functions as a tumor suppressor in PTC by direct targeting E2F7 and that miR‑30a may be a novel therapeutic target for patients with PTC.

Introduction

The incidence of thyroid cancer (TC) is the highest among endocrine malignancies (14), with ~62,450 new diagnoses and ~1,950 TC mortalities in 2015 alone (5). Furthermore, the incidence of TC has increased rapidly in recent years and as such has attracted more scientific attention; the age-adjusted incidence of TC is estimated to be 9.1 per 100,000 females and 2.9 per 100,000 males in developed countries (69). TC can be divided into four histologic groups, including papillary TC (PTC), poorly differentiated carcinoma, follicular TC and anaplastic TC (10). Among these, PTC accounts for ~80–90% of all patients with TC (10). PTC has a poor prognosis, so studies assessing the molecular mechanism of PTC development are urgently required (1114).

MicroRNAs (miRNAs) are small non-coding RNA molecules comprised of 20–22 nucleotides, which inhibit mRNA expression at the post-transcriptional level (15,16). Several studies have demonstrated that various miRNAs function as promoters or suppressors in many types of tumor, meaning that the identification of miRNAs may serve as a useful diagnostic and therapeutic approaches to cancer (10,17,18). Furthermore, previous studies have reported the use of miRNAs as biomarkers and their impact on the development of therapeutic strategies in various malignancies, including lung cancer (18), TC (19) and prostate cancer (20). Therefore, the identification of novel biomarkers and molecular targets may provide more effective treatment options for patients with PTC.

The results of the current study determined the expression of miR-30a in two TC cell lines and identified the effect of miR-30a on the viability, migration and invasion of PTC cells. These data indicated that miR-30a was downregulated in PTC cells, while its ectopic overexpression significantly inhibited the viability, migration and invasion of PTC cells. Therefore, miR-30a may have the potential to function as a diagnostic biomarker or a curative target in the future diagnosis and treatment of patients with PTC.

Materials and methods

Study sample

A total of 15 pairs of PTC tissues and matched adjacent non-tumor tissues were obtained from Fenyang Prison Hospital (Fenyang, China). All tissue samples were obtained following the receipt of written informed consent. The current study was approved by the Ethics Review Board of Fenyang College Shanxi Medical University (Shanxi, China). Seven tissue samples were from males and eight were from females. The average age of the study population was 58.4 years (range, 47–78 years). The data range of sample collection was between October 2017 and May 2018. The patients who were diagnosed with PTC were included in the study. The diagnoses were made by pathologists. Adjacent non-tumor tissues were isolated ≥2 cm away from the tumor border and were shown to be free of tumor cells via microscopy. The tissues were fixed with 10% formalin for 24 h at room temperature, processed in paraffin and sectioned using a microtome. The thickness of sections was 20 µm. Hematoxylin and eosin staining was used to confirm the diagnosis. Briefly, the tissue was stained in hematoxylin for 4 min at room temperature, washed under running tap water for 5 min, differentiated in 1% acid alcohol for 5 min at room temperature, and under running tap water for 5 min. The tissues were stained in 1% eosin Y for 10 min at room temperature, washed under running tap water for 5 min, and then dehydrated 95% ethanol and absolute ethanol. Subsequently, the tissues were cleared in xylene. The samples were observed under a light microscope at a magnification of ×100. Following tissue collection, samples were frozen in liquid nitrogen immediately, transported to the laboratory and stored at −80°C for RNA isolation. Following resection, tissues were washed with PBS, immediately frozen in liquid nitrogen and stored at −80°C. The expression of miR-30a in PTC tissue was then compared with adjacent non-tumor tissues.

Cell culture

The TPC-1 cell line and the normal PTC cell line (HT-ori3) were purchased from the American Type Culture Collection (Manassas, VA, USA). Cells were maintained in RPMI-1640 medium (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (Invitrogen; Thermo Fisher Scientific, Inc.), 100 IU/ml of penicillin and 100 µg/ml of streptomycin. All cells were cultured in a humidified incubator containing 5% CO2 at 37°C.

miRNA transfection

miR-30a mimics (20 nmol/l) and negative control (NC) miRNA (20 nmol/l) were acquired from Shanghai GenePharma Co., Ltd. (Shanghai, China). The miR-30a mimics sequence was 5′-UGUAAACAUCCUCGACUGGAAG-3′. The NC miRNA sequence was 5′-ACAUUUGUAGGAGCUGACCGGC-3′. Lipofectamine 2000® (Invitrogen; Thermo Fisher Scientific, Inc.) was subsequently utilized to perform transient transfection following incubation at 37°C for 6 h. Transfected cells were collected and purified after 48 h incubation.

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

RNA was extracted from cultured cells and tissues using the TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Subsequently, RNA was reverse transcribed using the RevertAid First Strand cDNA Synthesis kit (Thermo Fisher Scientific, Inc.). qPCR was performed to determine the expression of miR-30a and E2F transcription factor 7 (E2F7) using SYBRGreen (Qiagen, Inc., Valencia, CA, USA). RT-qPCR amplification was performed for 40 cycles using the following thermal profile: Denaturation for 15 sec at 95°C, annealing and extension for 1 min at 60°C. The following primers were utilized: miR-30a forward, 5′-CGCGATGTGTAAACATCCTCGAC-3′ and reverse, 5′-ATCCAGTGCAGGGTCCGAGG-3′; E2F7 forward, 5′-ACCCTCAGATTCCACAGACC-3′ and reverse, 5′-AGTTTGCTGTTGCCTTTCCT-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′. U6 served as an internal control. Furthermore, the 2-∆∆Cq method was utilized for RNA quantification and associated analysis (21).

MTT assay

Cell viability was assessed via an MTT assay. Cultured TPC-1 cells were first transfected with the miRNA mimics, then seeded into 96-well plates (~4,000 cells/well). Subsequently, MTT solution (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was added to each well and incubated at 37°C for 4 h. A solubilization solution, dimethyl sulfoxide, was added to dissolve the insoluble purple formazan. A microplate reader was used for detecting the viability of cells at an absorbance at 490 nm.

Invasion and migration assays

Transwell inserts that were precoated with Matrigel (BD Biosciences, San Jose, CA, USA) were utilized to identify the effect of miR-30a on the invasive capacity of PTC cells. PTC cells (1×105) were suspended in Dulbecco's Modified Eagle's medium (DMEM; Thermo Fisher Scientific, Inc.) without serum. Subsequently, these cells and medium were seeded into the upper chamber of the system and DMEM containing 20% FBS was added to the lower chamber. Cells were incubated at 37°C for 24 h and fixed using 4% polyoxymethylene at 4°C for 15 min. Following 15 min of incubation at 4°C, samples were stained using 0.1% crystal violet dye for 10 min at 37°C. The experimental procedure for the migration assay was the same as aforementioned, except the transwell inserts were not coated with Matrigel prior to experimentation. A light microscope was utilized to visualize the results at magnification, ×200. Each assay was performed in triplicate and repeated three times.

Western blotting

Protein from TPC-1 cells was extracted using modified radioimmunoprecipitation assay buffer containing 0.5% sodium dodecyl sulfate (SDS) in the presence of a proteinase inhibitor cocktail (all Roche Applied Science, Madison, WI, USA) at 96 h following transfection. A bicinchoninic acid assay was used for the detection of protein concentration. Protein (2 µg per lane) was then separated using 10% SDS-PAGE and transferred onto a polyvinylidene difluoride membrane. Membranes were blocked with 5% non-fat at room temperature for 1 h and incubated with the following primary antibodies at 4°C overnight: Anti-E2F7 (cat. no. sc-32574; 1:5,000; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) and anti-GAPDH (cat. no. 2118; 1:5,000; Cell Signaling Technology, Inc.). Samples were then incubated with horseradish peroxidase-conjugated goat anti-rabbit immunoglobulin G secondary antibodies (cat. no. 65-6120; 1:5,000; Invitrogen; Thermo Fisher Scientific, Inc.) at 4°C overnight. Immunoreactive bands were visualized on film by enhanced chemiluminescent substrate (Pierce; Thermo Fisher Scientific, Inc.). GAPDH was utilized as an internal control and data were analyzed using ImageJ software version 1.8 (National Institutes of Health, Bethesda, MD, USA).

Luciferase reporter assay

TargetScan (www.targetscan.org/) was used to predict the target gene of miR-30a. To determine the association between miRNA and target genes, PCR was used to amplify the 3′-untranslated region (UTR) of human E2F7, which was then cloned into a pmirGLO vector (Promega Corporation, Madison, WI, USA). The PCR method used is detailed in the previous subsection. Subsequently, Lipofectamine 2000® was utilized to transfect cells in the 24-well plates. The obtained wild type or mutant pmirGLO vector, pRL-SV40 Renilla luciferase construct (5 ng; Promega Corporation) and miR-30a mimic or the respective negative control were co-transfected to each well. After 48 h transfection, cells were extracted and the luciferase activity was determined using the Dual luciferase reporter assay system (DLR® Assay) following 48 h. Firefly luciferase activity in the vectors was normalized to Renilla luciferase activity.

Statistical analysis

Data were analyzed using GraphPad Prism 6.0 software (GraphPad Software, Inc., La Jolla, CA, USA) and presented as the mean ± standard deviation. One-way analysis of variance followed by a Tukey's post-hoc test was used for comparisons between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-30a is downregulated in human PTC tissue

To obtain a greater understanding of the clinical interrelation of the expression of miR-30a in PTC tissue, 15 pairs of PTC tissue samples and non-tumor tissues were assessed via RT-qPCR. The results revealed that the expression of miR-30a was significantly downregulated in PTC tissue compared with non-tumor tissue (Fig. 1).

miR-30a is downregulated and E2F7 is upregulated in human PTC cells

The current study aimed to determine the expression of miR-30a in a PTC cell line (TPC-1) and a normal thyroid cell line (HT-ori3). Compared with HT-ori3 cells, the expression of miR-30a and the levels of E2F7 in TCP-1 cells were significantly decreased and increased, respectively (Fig. 2A-B). Western blotting was subsequently performed to detect the protein expression of E2F7 in these cell lines. The results revealed that E2F7 levels in TPC-1 cells were markedly increased compared with HT-ori3 cells (Fig. 2C-D).

miR-30a decreases the proliferation of TPC-1 cells

To determine whether miR-30a regulates the proliferation of PTC cells, miR-30a mimics were utilized to increase the expression of miR-30a. Following transfection for 6 h, a significant increase in miR-30a expression was observed when compared with the NC mimic group (Fig. 3A). An MTT assay was subsequently performed to determine the proliferation of TPC-1 cells following miR-30a mimic or NC mimic transfection. The results revealed that transfection with miR-30a mimics significantly decreased cell proliferation compared with the NC mimics group. Furthermore, the proliferation of cells in the miR-30a mimic group achieved the minimal viability rate and become stable after 48 h (Fig. 3B). These results confirmed that the upregulation of miR-30a inhibits the proliferation of PTC cells.

miR-30a inhibits the migration and invaison of TPC-1 cells

To assess the roles of miR-30a in PTC cell migration and invasion, transwell migration and invasion assays were performed in TPC-1 cells transfected with miR-30a or NC mimics. The results indicated that the overexpression of miR-30a, induced via the transfection of a miR-30a mimic, significantly suppressed the migration (Fig. 4A) and invasion (Fig. 4B) of TPC-1 cells compared with the NC mimics group.

miR-30a targets the E2F7 3′-UTR directly

To identify the molecular mechanism of miR-30a in the regulation of cell survival, putative miR-30a targets were predicted via bioinformatics analysis (Fig. 5A). The results revealed that E2F7 is a direct target of miR-30a. A luciferase reporter assay was then performed to confirm whether miR-30a targets E2F7 directly (Fig. 5B). The results demonstrated that, compared with the NC mimics group, co-transfection with miR-30a mimics significantly decreased the luciferase activity of the wild-type E2F7-3′-UTR luciferase vector in TPC-1 cells. However, no significant effect on the luciferase activity of mutant E2F7-3′-UTR following transfection of miR-30a was observed. Furthermore, the protein levels of E2F7 were markedly decreased following miR-30a transfection (Fig. 5C). These data indicate that E2F7 may be a direct target of miR-30a in PTC cells.

Discussion

TC is a common endocrine malignancy that is primarily derived from follicular thyroid or parafollicular C cells (22). The overall 10-year survival rate of differentiated TC was >80%, with ~5–20% of TC patients developing local or regional recurrence and 10–15% developing distant metastases in a 2014 study (23). Furthermore, PTC accounts for ~80–90% of all TC patients. In recent years, previous studies have aimed to assess certain miRNAs as possible regulators of tumorigenesis and PTC development (2426). The current study aimed to assess the role of miR-30a in PTC via the regulation of E2F7. The results demonstrated that the overexpression of miR-30a suppressed the proliferation, migration and invasion of PTC cells by directly targeting E2F7.

Previous studies have demonstrated that miRNAs serve as tumor suppressors in various types of cancer (2730). A previous study revealed that miR-141 suppressed TC cell growth and metastasis by suppressing insulin receptor substrate 2, indicating that miR-141 may serve as a potential therapeutic target for the treatment of patients with TC (31). Furthermore, miR-497 is also considered to be a TC tumor suppressor that acts by repressing brain derived neurotrophic factor (32). A previous study also indicated that miR-7 may function as a tumor suppressor by directly targeting serine/threonine protein kinase 1, potentially serving as a novel therapeutic target for TC (30). Therefore, miRNAs have been demonstrated to be associated with tumor development and progression, highlighting their potential use as biomarkers for the diagnosis and prognosis of PTC. However, the prognostic relevance and functions of miR-30a in PTC remain largely unclear. The current study assessed the expression of miR-30a and their role in PTC cells. The results indicated that miR-30a was downregulated in PTC cell lines compared with normal cell lines. Furthermore, the ectopic expression of miR-30a inhibited PTC cell proliferation, migration and invasion.

It is well known that E2F7 serves predominantly as a transcriptional repressor, binding to miRNA promoters and protein-coding genes bearing E2F consensus motifs, thereby inhibiting their expression (33). The E2F family of transcription factors are important regulators of cellular proliferation (34). A previous study has revealed that the E2F7 protein serves an essential role in the regulation of cell cycle progression and may be a key component of a negative feedback loop required to turn off the transcription of E2F-driven G1/S target genes, thus allowing progression through the cell cycle (35). The p53-dependent transcriptional upregulation of E2F7 results in the repression of relevant gene expression and the E2F7-dependent mechanism contributes to p53-dependent cell cycle arrest in response to DNA damage (36). Furthermore, E2F7 may serve as an independent prognostic factor of glioma and may therefore constitute a potential therapeutic target for this disease (37). A recent study demonstrated that the inhibition of E2F7 inhibits gallbladder cancer cell proliferation, migration and metastasis (38). In the current study, miR-30a was confirmed to target E2F7 directly and the increased expression of miR-30a significantly suppressed E2F7 expression in human PTC cells. Furthermore, the results of transwell migration and invasion assays indicated that the overexpression of miR-30a, induced via transfection with miR-30a mimics, significantly suppressed the migration and invasion of PTC cells. In future experiments, studies will aim to perform other functional assays to determine the effect of miR-30a on the proliferation and metastasis of PTC cells in vitro and in vivo, including cell cycle analysis by flow cytometry and wound healing assay. Collectively, the current study revealed that miR-30a inhibited cell proliferation, migration and invasion, partially by targeting E2F7. Therefore, the current study provided evidence for the critical regulatory axis of miR-30a/E2F7 in the developmental process of PTC.

Overall, the current study demonstrated that the proliferation, migration and invasion of PTC cells are inhibited by miR-30a overexpression by targeting E2F7. All results obtained in the current study indicate that miR-30a may serve as a tumor suppressor gene in the tumorigenesis and progression of PTC. Therefore, miR-30a and E2F7 may serve as potential therapeutic targets for the treatment of patients with PTC.

Acknowledgements

Not applicable.

Funding

No funding 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

GY conceived the current study, wrote the paper and created the figures. GY and ZY performed the experiments, searched the literature, analyzed the data, created the tables and reviewed the manuscript.

Ethics approval and consent to participate

All tissue samples were obtained following the receipt of written informed consent. The current study was approved by the Ethics Review Board of Fenyang College Shanxi Medical University (Shanxi, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wang J, Yang H, Si Y, Hu D, Yu Y, Zhang Y, Gao M and Zhang H: Iodine promotes tumorigenesis of thyroid cancer by suppressing Mir-422a and up-regulating MAPK1. Cell Physiol Biochem. 43:1325–1336. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Kaptan E, Sancar-Bas S, Sancakli A, Bektas S and Bolkent S: The effect of plant lectins on the survival and malignant behaviors of thyroid cancer cells. J Cell Biochem. 119:6274–6287. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Sokić SI, Adanja BJ, Vlajinac HD, Janković RR, Marinković JP and Zivaljević VR: Risk factors for thyroid cancer. Neoplasma. 41:371–374. 1994.PubMed/NCBI

4 

Takacsova E, Kralik R, Waczulikova I, Zavodna K and Kausitz J: A different prognostic value of BRAFV600E mutation positivity in various age groups of patients with papillary thyroid cancer. Neoplasma. 64:156–164. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Di W, Li Q, Shen W, Guo H and Zhao S: The long non-coding RNA HOTAIR promotes thyroid cancer cell growth, invasion and migration through the miR-1-CCND2 axis. Am J Cancer Res. 7:1298–1309. 2017.PubMed/NCBI

6 

Xing M, Haugen BR and Schlumberger M: Progress in molecular-based management of differentiated thyroid cancer. Lancet. 381:1058–1069. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Ding ZY, Huang YJ, Tang JD, Li G, Jiang PQ and Wu HT: Silencing of hypoxia-inducible factor-1α promotes thyroid cancer cell apoptosis and inhibits invasion by downregulating WWP2, WWP9, VEGF and VEGFR2. Exp Ther Med. 12:3735–3741. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Lim ST, Jeon YW and Suh YJ: The prognostic values of preoperative tumor volume and tumor diameter in T1N0 papillary thyroid cancer. Cancer Res Treat. 49:890–897. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Rachinsky I, Rajaraman M, Leslie WD, Zahedi A, Jefford C, McGibbon A, Young JE, Pathak KA, Badreddine M, De Brabandere S, et al: Regional variation across Canadian centers in radioiodine administration for thyroid remnant ablation in well-differentiated thyroid cancer diagnosed in 2000–2010. J Thyroid Res. 2016:28679162016. View Article : Google Scholar : PubMed/NCBI

10 

Guan H, Liang W, Xie Z, Li H, Liu J, Liu L, Xiu L and Li Y: Down-regulation of miR-144 promotes thyroid cancer cell invasion by targeting ZEB1 and ZEB2. Endocrine. 48:566–574. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Vasko VV and Saji M: Molecular mechanisms involved in differentiated thyroid cancer invasion and metastasis. Curr Opin Oncol. 19:11–17. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Liao T, Wang YJ, Hu JQ, Wang Y, Han LT, Ma B, Shi RL, Qu N, Wei WJ, Guan Q, et al: Histone methyltransferase KMT5A gene modulates oncogenesis and lipid metabolism of papillary thyroid cancer in vitro. Oncol Rep. 39:2185–2192. 2018.PubMed/NCBI

13 

Lu ZL, Chen YJ, Jing XY, Wang NN, Zhang T and Hu CJ: Detection and identification of serum peptides biomarker in papillary thyroid cancer. Med Sci Monit. 24:1581–1587. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Zhao W, Chen S, Hou X, Chen G and Zhao Y: CHK2 promotes anoikis and is associated with the progression of papillary thyroid cancer. Cell Physiol Biochem. 45:1590–1602. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Shi Z, Zhou H, Lu L, Li X, Fu Z, Liu J, Kang Y, Wei Z, Pan B, Liu L, et al: The roles of microRNAs in spinal cord injury. Int J Neurosci. 127:1104–1115. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Ouyang Q, Xu L, Cui H, Xu M and Yi L: MicroRNAs and cell cycle of malignant glioma. Int J Neurosci. 126:1–9. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, et al: Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 18:997–1006. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Wang RJ, Zheng YH, Wang P and Zhang JZ: Serum miR-125a-5p, miR-145 and miR-146a as diagnostic biomarkers in non-small cell lung cancer. Int J Clin Exp Pathol. 8:765–771. 2015.PubMed/NCBI

19 

Fuziwara CS and Kimura ET: MicroRNA deregulation in anaplastic thyroid cancer biology. Int J Endocrinol. 2014:7434502014. View Article : Google Scholar : PubMed/NCBI

20 

Costa-Pinheiro P, Ramalho-Carvalho J, Vieira FQ, Torres-Ferreira J, Oliveira J, Gonçalves CS, Costa BM, Henrique R and Jerónimo C: MicroRNA-375 plays a dual role in prostate carcinogenesis. Clin Epigenetics. 7:422015. 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 

Xing M: Molecular pathogenesis and mechanisms of thyroid cancer. Nat Rev Cancer. 13:184–199. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Perros P, Boelaert K, Colley S, Evans C, Evans RM, Gerrard Ba G, Gilbert J, Harrison B, Johnson SJ, Giles TE, et al: Guidelines for the management of thyroid cancer. Clin Endocrinol (Oxf). 81 (Suppl 1):S1–S122. 2014. View Article : Google Scholar

24 

Bi CL, Zhang YQ, Li B, Guo M and Fu YL: MicroRNA-520a-3p suppresses epithelial-mesenchymal transition, invasion, and migration of papillary thyroid carcinoma cells via the JAK1-mediated JAK/STAT signaling pathway. J Cell Physiol. 234:4054–4067. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Liu H, Guo J, Chai H and Meng X: MicroRNA-744 suppresses cell proliferation and invasion of papillary thyroid cancer by directly targeting NOB1. Mol Med Rep. 19:1903–1910. 2019.PubMed/NCBI

26 

Heidari Z, Mohammadpour-Gharehbagh A, Eskandari M, Harati-Sadegh M and Salimi S: Genetic polymorphisms of miRNA let7a-2 and pri-mir-34b/c are associated with an increased risk of papillary thyroid carcinoma and clinical/pathological features. J Cell Biochem. 14–Dec;2018.(Epub ahead of print).

27 

Damodaran M, Paul SFD and Venkatesan V: Genetic polymorphisms in miR-146a, miR-196a2 and miR-125a genes and its association in prostate cancer. Pathol Oncol Res. 29–Mar;2018.(Epub ahead of print). View Article : Google Scholar

28 

Mo JS, Park YR and Chae SC: MicroRNA 196B regulates HOXA5, HOXB6 and GLTP expression levels in colorectal cancer cells. Pathol Oncol Res. 12–Mar;2018.(Epub ahead of print). View Article : Google Scholar

29 

Seclaman E, Narita D, Anghel A, Cireap N, Ilina R, Sirbu IO and Marian C: MicroRNA expression in laser micro-dissected breast cancer tissue samples-a pilot study. Pathol Oncol Res. 25:233–239. 2019. View Article : Google Scholar

30 

Yue K, Wang X, Wu Y, Zhou X, He Q and Duan Y: microRNA-7 regulates cell growth, migration and invasion via direct targeting of PAK1 in thyroid cancer. Mol Med Rep. 14:2127–2134. 2016. View Article : Google Scholar : PubMed/NCBI

31 

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

32 

Wang P, Meng X, Huang Y, Lv Z, Liu J, Wang G, Meng W, Xue S, Zhang Q, Zhang P and Chen G: MicroRNA-497 inhibits thyroid cancer tumor growth and invasion by suppressing BDNF. Oncotarget. 8:2825–2834. 2017.PubMed/NCBI

33 

Westendorp B, Mokry M, Groot Koerkamp MJ, Holstege FC, Cuppen E and de Bruin A: E2F7 represses a network of oscillating cell cycle genes to control S-phase progression. Nucleic Acids Res. 40:3511–3523. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Chen D, Pacal M, Wenzel P, Knoepfler PS, Leone G and Bremner R: Division and apoptosis of E2f-deficient retinal progenitors. Nature. 462:925–929. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Mitxelena J, Apraiz A, Vallejo-Rodríguez J, García-Santisteban I, Fullaondo A, Alvarez-Fernández M, Malumbres M and Zubiaga AM: An E2F7-dependent transcriptional program modulates DNA damage repair and genomic stability. Nucleic Acids Res. 46:4546–4559. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Carvajal LA, Hamard PJ, Tonnessen C and Manfredi JJ: E2F7, a novel target, is up-regulated by p53 and mediates DNA damage-dependent transcriptional repression. Genes Dev. 26:1533–1545. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Yin W, Wang B, Ding M, Huo Y, Hu H, Cai R, Zhou T, Gao Z, Wang Z and Chen D: Elevated E2F7 expression predicts poor prognosis in human patients with gliomas. J Clin Neurosci. 33:187–193. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Ye YY, Mei JW, Xiang SS, Li HF, Ma Q, Song XL, Wang Z, Zhang YC, Liu YC, Jin YP, et al: MicroRNA-30a-5p inhibits gallbladder cancer cell proliferation, migration and metastasis by targeting E2F7. Cell Death Dis. 9:4102018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2019
Volume 18 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo H and Guo H: MicroRNA‑30a suppresses papillary thyroid cancer cell proliferation, migration and invasion by directly targeting E2F7. Exp Ther Med 18: 209-215, 2019
APA
Guo, H., & Guo, H. (2019). MicroRNA‑30a suppresses papillary thyroid cancer cell proliferation, migration and invasion by directly targeting E2F7. Experimental and Therapeutic Medicine, 18, 209-215. https://doi.org/10.3892/etm.2019.7532
MLA
Guo, H., Zhang, L."MicroRNA‑30a suppresses papillary thyroid cancer cell proliferation, migration and invasion by directly targeting E2F7". Experimental and Therapeutic Medicine 18.1 (2019): 209-215.
Chicago
Guo, H., Zhang, L."MicroRNA‑30a suppresses papillary thyroid cancer cell proliferation, migration and invasion by directly targeting E2F7". Experimental and Therapeutic Medicine 18, no. 1 (2019): 209-215. https://doi.org/10.3892/etm.2019.7532