Inhibitory roles of miR‑9 on papillary thyroid cancer through targeting BRAF

  • Authors:
    • Yi Gu
    • Nan Yang
    • Leping Yin
    • Chao Feng
    • Tong Liu
  • View Affiliations

  • Published online on: May 11, 2018     https://doi.org/10.3892/mmr.2018.9010
  • Pages: 965-972
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA‑9 (miR‑9) is reported to be underexpressed in papillary thyroid carcinoma (PTC) tissues; however, the molecular mechanisms underlying the implication of miR‑9 in PTC have yet to be elucidated. The present study aimed to explore the potential roles of miR‑9 in PTC. PTC tissue samples and paired non‑cancerous adjacent tissues were collected from 60 patients with PTC. The human TPC‑1 thyroid gland papillary carcinoma cell line was used to investigate the molecular mechanisms underlying the roles of miR‑9 in PTC. The levels of miR‑9 and its downstream target gene BRAF were detected through reverse transcription‑quantitative polymerase chain reaction. MTT assay and flow cytometry were performed to evaluate cell viability and apoptosis, respectively. A mouse xenograft tumor model was established to observe the effects of miR‑9 on thyroid gland tumorigenesis in vivo. The present study revealed that the expression of miR‑9 was significantly reduced in PTC tissues compared with paired normal tissues. In addition, miR‑9 upregulation suppressed the expression of BRAF in TPC‑1 cells in vitro. Luciferase reporter assay demonstrated that BRAF may be a direct target gene of miR‑9 in TPC‑1 cells. In addition, following transfection with miR‑9 mimics, the viability of TPC‑1 cells was suppressed and their apoptosis was enhanced; conversely, transfection with miR‑9 inhibitor exerted the opposite effects in vitro. miR‑9 overexpression or downregulation also affected in vivo PTC tumorigenesis in athymic mice. The present findings suggested that miR‑9 may suppress the viability of PTC cells and inhibit tumor growth through directly targeting the expression of BRAF in PTC.

Introduction

Thyroid cancer is one of the most frequent types of endocrine neoplasms worldwide, with ~300,000 new cases occurring every year (1,2). Due to several environmental and socioeconomic factors, the occurrence rate of thyroid cancer is more increased among rural populations (3). Thyroid cancer has been identified as one of the most common types of malignancy among Asian population in 2015 (4). Based on the degree of cell differentiation, thyroid cancer can be classified into several types, including papillary thyroid carcinoma (PTC), medullary thyroid carcinoma (MTC) and follicular thyroid carcinoma (5). PTC accounts for the majority of thyroid carcinomas, and is responsible for ~80% of global thyroid cancer cases (6,7).

Although the 5-year survival rate of PTC exceeds 90%, the occurrence of lymph node metastasis is frequent and the recurrence risk remains high (8,9). The most common therapeutic strategy used for PTC treatment is thyroidectomy, which is featured by a high success rate of ~85% (10,11). However, previous studies have reported that ~20% of patients with PTC who underwent thyroidectomy exhibited regional recurrence during a mean follow-up of <5 years (10,12). In addition, thyroidectomy is characterized by various limitations, and the application of surgery for lymph node excision remains controversial (13,14). Therefore, investigation of the cellular and molecular mechanisms involved in PTC is essential for the development of more effective therapeutic strategies to prevent the recurrence in patients.

MicroRNAs (miRNAs) are a class of small non-coding RNA molecules with a length of 20–22 nucleotides, which regulate gene expression at the post-transcriptional stage and participate in numerous cellular processes, including cell proliferation, differentiation and apoptosis (15,16). miRNAs have been suggested to serve an important role in the development of PTC (17). For example, let-7 is associated with PTC, through the regulation of high-mobility group AT-hook 2 (HMGA2) and solute carrier family 5 member 5 (SLC5A5) expression (18). miR-146 has been reported to suppress the expression of the retinoic acid receptor β in PTC, thus attenuating the efficiency of retinoic acid and radioactive iodine treatment, which suggests that the deregulation of miRNA may also influence the therapeutic outcome of PTC treatment (19). Previous studies have reported that the metastasis and proliferation of thyroid carcinoma cells could be regulated by several miRNAs (20), including miR-146b (21), miR-451a (22) and miR-205 (23). Sondermann et al (24) suggested that miR-9 may have potential prognostic value for predicting the recurrence of PTC, as miR-9 expression in patients with non-recurrent PTC was significantly higher compared with patients with recurrent PTC. Gundara et al (25) reported that miR-9 directly targeted autophagy protein 5 (Atg5) and could suppress the viability of MTC cells in vitro, thus indicating an important role for miR-9 in the pathogenesis of thyroid cancer. Furthermore, the aberrant expression of the proto-oncogene BRAF has been associated with the development of PTC, and the BRAFV600E mutation has been considered to be implicated in the progression and lymph node metastasis of PTC (2628). Therefore, the present study aimed to explore the putative relationship between miR-9 and BRAF expression in PTC.

In the present study, the roles of miR-9 in the development and progression of PTC were investigated in vitro and in vivo. The present findings will help in the understanding of the molecular processes involved in thyroid cancer and in the development of novel therapeutic approaches for the treatment of patients with PTC.

Materials and methods

Subjects

A total of 60 pairs of fresh frozen PTC tissue samples and paired adjacent non-cancerous tissues were collected at Sichuan Provincial People's Hospital (Chengdu, China) between March 2014 and November 2015. All samples were collected from patients with PTC who had not received any previous adjuvant treatments, including chemotherapy, radiotherapy or hormone therapy. All patients were diagnosed with PTC based on histopathological evaluation. The samples were immediately stored in liquid nitrogen until further use. The present study was approved by the Ethics Committee of Sichuan Provincial People's Hospital. Written informed consent was obtained from all patients prior to enrollment in the present study. The clinicopathological characteristics of the patients are presented in Table I.

Table I.

Association between miR-9 expression levels in PTC tissues and clinicopathological characteristics of patients with PTC.

Table I.

Association between miR-9 expression levels in PTC tissues and clinicopathological characteristics of patients with PTC.

miR-9 expression

CharacteristicTotal numberLow (n=28)High (n=32)P-value
Age (years)
  <502413110.342
  ≥50361521
Sex
  Male4723240.503
  Female13  5  8
Cervical lymph
node metastasis
  Positive2314  90.082
  Negative371423
TNM stage
  I/II3511240.005
  III/IV2517  8

[i] PTC, papillary thyroid carcinoma; TNM, tumor-lymph node-metastasis.

Cell culture

The human TPC-1 thyroid gland papillary carcinoma cell line was purchased from Bena Culture Collection (Beijing, China) and cultured in Dulbecco's modified Eagle's medium (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) supplemented with 10% fetal bovine serum, 100 U/ml penicillin and 100 µg/ml streptomycin (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Cells were maintained in a humidified incubator in a 5% CO2 atmosphere at 37°C.

Cell transfection

The miR-9 mimics, miR-9 inhibitor and scramble oligonucleotides were purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China). The sequence of the miR-9 mimics was 5′-UCUUUGGUUAUCUAGCUGUAUGA-3′; the sequence of the miR-9 inhibitor was 5′-UCAUACAGCUAGAUAACCAAAGA-3′; the sequence of the scramble oligonucleotides was 5′-CAGUACUUUUGUGUAGUACAA-3′. The overexpression vector for BRAF (pcDNA3.1-BRAF) was constructed by cloning the BRAF open reading frame sequence (range, 226–2,529, without 3′-UTR) into the multiple cloning site of the pcDNA3.1 vector (Invitrogen; Thermo Fisher Scientific, Inc.).

TPC-1 cells (1×106) were cultured in 6-well culture plates, and transfection was performed using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol until confluence reached 50–60%. Untransfected cells served as a control. Following 48 h of transfection, the cells were harvested and used for further experiments.

RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA from tissues and cells was isolated using TRIzol reagent (Invitrogen) according to the manufacturer's protocol. For miR-9 detection, cDNA was synthesized from total RNA using a One Step Prime script miRNA cDNA Synthesis kit (Qiagen, Inc., Valencia, CA, USA). The relative expression levels of miR-9 were detected using the mirVana™ qRT-PCR miRNA Detection kit (Invitrogen) on a preheated 7500 Real Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). For BRAF mRNA detection, cDNA was synthesized from total RNA using a PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). qPCR was performed for BRAF mRNA detection using the SYBR Premix Ex TaqII PCR kit (Takara Biotechnology Co., Ltd.). The relative expression levels of miR-9 were normalized to U6, whereas BRAF mRNA expression was normalized to β-actin. The data were analyzed according to the 2−ΔΔCq method (29). The PCR cycling parameters were as follows: 5 min denaturation at 95°C; then 30 cycles of 95°C for 15 sec, 55°C for 20 sec, 70°C for 30 sec; 5 min extension at 70°C. The primers for miR-9 (30) were designed by Applied Biosystems (Thermo Fisher Scientific, Inc.). The remaining primers were as follows: BRAF forward, 5′-ACCACCCAATACACAGGAA-3′ and reverse, 5′-CATTGGGAGCTGATGAGGAT-3′; U6 forward, 5′-CGCTTCGGCAGCACATATACTAAAATTGGAAC-3′ and reverse, 5′-GCTTCACGAATTTGCGTGTCATCCTTGC-3′; and β-actin forward, 5′-AAACTGGAACGGTGAAGGTG-3′ and reverse, 5′-AGAGAAGTGGGGTGGCTTTT-3′.

Protein isolation and western blot analysis

Cells were lysed using radioimmunoprecipitation assay lysis buffer containing protease inhibitor cocktail (Beyotime Institute of Biotechnology, Haimen, China). Protein concentration was determined using a bicinchoninic acid protein assay kit (Thermo Fisher Scientific, Inc.). Extracted protein samples (~30 µg) were separated by 10% SDS-PAGE and transferred onto nitrocellulose membranes (Bio-Rad Laboratories, Hercules, CA, USA). The membranes were blocked in TBS containing 0.1% Tween-20 (TBST) with 5% skimmed milk for 1 h at room temperature, and were then incubated with primary antibodies against BRAF (TA890103; 1:800; OriGene Technologies, Inc., Beijing, China) at 4°C overnight. Following washing, the membranes were incubated with horseradish peroxidase-conjugated secondary antibodies (TA130023; 1:2,000; OriGene Technologies, Inc.). β-actin was used as the endogenous control (4970; 1:1,000 dilution; Cell Signaling Technology, Inc., Danvers, MA, USA). Protein bands were visualized using the enhanced chemiluminescence (ECL) kit (Thermo Fisher Scientific, Inc.), and blots were semi-quantified using ImageJ software version 1.46 (National Institutes of Health, Bethesda, MD, USA).

Luciferase reporter assay

Bioinformatics analysis was performed to predict potential target genes for miR-9 using Targetscan (31) and miRanda (32). The results indicated that miR-9 may directly interact with the 3′-untranslated region (UTR) of the BRAF mRNA. To investigate the interaction in vitro, the 3′-UTR of BRAF containing the putative binding site for miR-9 was cloned downstream of the Renilla luciferase gene on a psiCHECK-2 reporter plasmid (Promega Corporation, Madison, WI, USA). BRAF 3′-UTR mutants were generated using the GeneTailor Site-Directed Mutagenesis system (Invitrogen; Thermo Fisher Scientific, Inc.) and were also ligated with the psiCHECK-2 vector. The constructed luciferase vectors (0.2 µg) were co-transfected with miR-9 mimics, miR-9 inhibitor or scramble oligonucleotides (100 nM) into TPC-1 cells using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.). The luciferase activities following 48 h of transfection were measured by the Dual Luciferase Reporter assay system (Promega Corporation).

Cellular apoptosis analysis

To assess cellular apoptosis, TPC-1 cells were seeded into 6-well plates at a density of 1×105 cells/well. After 48 h, the cells were harvested and the apoptotic rates were determined using an Annexin V-fluorescein isothiocyanate/propidium iodide Apoptosis Detection kit (BD Biosciences, San Jose, CA, USA) according to the manufacturer's protocol. Cells were analyzed and the apoptotic rate was quantified using a FACScan flow cytometer (BD Biosciences).

Cellular viability analysis

Cell viability was measured using an MTT assay. TPC-1 cells were seeded in 96 well plates at a density of 5×103 cells/well. Following incubation for 24 h at 37°C, the MTT Cell Proliferation and Cytotoxicity Assay kit (Beyotime Institute of Biotechnology) was used, and 40 µl MTT stock solution was added to each well for 4 h. The supernatants were discarded and 200 µl DMSO (Sigma-Aldrich; Merck KGaA) was added to dissolve the formazan crystals. The absorbance of each sample was measured at 570 nm using a microplate reader.

In vivo tumorigenesis

Male BALB/c athymic mice (n=30; age, 4–6 weeks; weight, 20–22 g), purchased from Shanghai SLAC Laboratory Animal Co., Ltd. (Shanghai, China), were housed in micro-isolator cages (22–25°C, 50–60% humidity, 12-h light/dark cycle) with free access to food and water. TPC-1 cells (2×106 cells in 100 µl PBS) were subcutaneously inoculated into the right flank of the mice. Then the mice were randomized into 5 groups (n=6 mice/group), and miR-9 mimics, miR-9 inhibitor, pcDNA3.1-BRAF, or scramble oligonucleotides (100 nM in 100 µl PBS) were injected into the tumors directly twice a week. The mice in the control group received vehicle (DMSO) injections. Tumor volumes were measured every 4 days using a caliper according to the following formula: Volume = length × width2/2. Mice were sacrificed 4 weeks after the initial injection, and tumors were dissected. All protocols of animal experiments were approved by the Animal Ethics Committee of Sichuan Provincial People's Hospital (Sichuan, China).

Statistical analysis

All statistical analyses were performed using SPSS software version 17.0 (SPSS, Inc., Chicago, IL, USA) and GraphPad Prism software 6.0 (GraphPad Software, Inc., La Jolla, CA, USA). The statistical significance of the differences between two groups was assessed using two-tailed unpaired Student's t-test. Categorical data was compared using the Chi-square test. Data were presented as the mean ± standard deviation from at least three independent experiments. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-9 expression is downregulated in PTC tissues

The relative expression levels of miR-9 and BRAF mRNA in PTC tissues and paired adjacent non-cancerous tissues were assessed using RT-qPCR. The relative expression levels of miR-9 in PTC tissues were significantly downregulated compared with in paired non-cancerous tissues (Fig. 1A). In addition, the mRNA expression levels of BRAF were significantly upregulated in PTC tissues compared with normal tissue samples (Fig. 1B). As presented in Table I, the downregulation of miR-9 expression was significantly associated with the advanced TNM stage (P=0.005), whereas no significant correlation was found between miR-9 expression and age (P=0.342), sex (P=0.503) or cervical lymph node metastasis (P=0.082).

BRAF expression is suppressed by miR-9 in TPC-1 cells

In the present study, human TPC-1 thyroid gland papillary carcinoma cells were transfected with miR-9 mimics or miR-9 inhibitor and the mRNA and protein expression levels of BRAF were then detected. The results of RT-qPCR demonstrated that overexpression of miR-9 significantly suppressed the mRNA expression of BRAF, whereas inhibition of miR-9 resulted in a significant upregulation in BRAF mRNA levels (Fig. 2A). Western blot analysis also revealed that the protein expression levels of BRAF were significantly decreased following miR-9 overexpression, whereas they were increased following miR-9 inhibition (Fig. 2B).

BRAF is a direct target gene of miR-9 in TSC-1 cells

Bioinformatics analysis identified a potential binding site for miR-9 in the 3′-UTR of the BRAF mRNA (Fig. 3A). A luciferase reporter assay was performed to confirm the putative interaction in vitro. As presented in Fig. 3B, the luciferase activity in TSC-1 cells transfected with a luciferase vector encoding the wild-type BRAF 3′-UTR was significantly suppressed following transfection with miR-9 mimics. Conversely, TSC-1 cells transfected with the mutated BRAF 3′-UTR sequence were not markedly affected by miR-9 mimics or inhibitor, indicating that miR-9 may directly target BRAF expression in TSC-1 cells (Fig. 3B).

miR-9 inhibits the proliferation and promotes the apoptosis of TSC-1 cells

Flow cytometric analysis was performed to evaluate the effects of miR-9 on cancer cell apoptosis. The apoptotic rates of TSC-1 cells were significantly elevated following transfection with miR-9 mimics. By contrast, co-transfection with pcDNA3.1-BRAF suppressed the increase of apoptotic rates. These findings suggested that miR-9 may be involved in the regulation of PTC cell apoptosis, through the modulation of BRAF expression (Fig. 4).

MTT assay demonstrated that overexpression of miR-9 resulted in the significant suppression of TPC-1 cell proliferation, whereas co-transfection with pcDNA3.1-BRAF restored the miR-9-induced inhibition of cell viability (Fig. 5).

miR-9 suppresses in vivo PTC tumorigenesis in xenografted mice

As shown in Fig. 6, tumor growth in mice treated with the miR-9 inhibitor was significantly increased compared with the control or scramble-treated mice. Notably, miR-9 overexpression significantly suppressed tumor growth in vivo, whereas this effect was restored following the co-administration of pcDNA3.1-BRAF (Fig. 6). These results indicated that miR-9/BRAF may be implicated in the regulation of in vivo PTC tumorigenesis.

Discussion

PTC is one of the most common types of thyroid cancer, and its incidence rate has increased by >240%, with ~62,980 expected new cases in 2014, thus making PTC a major issue for human health worldwide (33). Currently, miRNAs have been suggested as potential prognostic and diagnostic biomarkers in various types of human cancer. Previous studies have demonstrated that dysregulation of the expression of several miRNAs was involved in the pathogenesis of thyroid cancer (3436). The recurrence of PTC still remains high following thyroidectomy, thus stressing the need for the elucidation of the exact molecular mechanisms implicated in PTC pathogenesis.

The results of the present study demonstrated that miR-9 was significantly downregulated in PTC tissues compared with adjacent non-cancerous tissue samples. In addition, luciferase activity assay revealed that miR-9 directly targeted BRAF and suppressed its expression, thereby suppressing the viability and enhancing the apoptosis of human TSC-1 thyroid gland papillary carcinoma cells. Furthermore, the results of a mouse thyroid tumor xenograft model suggested that miR-9 may suppress tumor growth in vivo.

The present results demonstrating the suppression of miR-9 expression in PTC tissues were consistent with previous studies (20,24) investigating the roles of miR-9 in thyroid cancer. Gundara et al (25) reported that miR-9 inhibited the autophagic flux and increased the apoptosis of cancer cells in MTC; autophagy has been suggested as one of the survival mechanisms in MTC cells. However, the association between miR-9 and autophagy has yet to be further explored.

In recent years, studies of signal transduction pathways have made a great contribution to the understanding of the molecular mechanisms implicated in tumorigenesis. At present, four mitogen-activated protein kinase (MAPK) signal transduction pathways have been identified in eukaryotic cells, including the extracellular signal-regulated kinase (ERK) pathway, the c-Jun N-terminal kinase/stress activated protein kinase pathway, the p38 pathway and the ERK5 pathway. The MAPK pathway, which includes the kinases Ras, Raf, MAPK/ERK kinase (MEK) and ERK, is implicated in the regulation of several cellular processes, including proliferation, differentiation, apoptosis and survival (37). In patients with PTC, ~80% of gain-of-function mutations have been identified in genes encoding signaling molecules participating in the MAPK pathways, thus suggesting that MAPK pathways may serve a critical role in the pathogenesis of PTC (38). Zawistowski et al (39) reported that miR-9 inhibited the MEK/ERK signaling pathway via targeting integrinβ-1 in breast cancer. However, the exact molecular mechanisms underlying the involvement of miR-9 in the development of PTC have yet to be elucidated. To the best of our knowledge, the present study is the first to demonstrate that miR-9 could directly target BRAF to suppress the proliferation and growth of thyroid cancer cells in vitro.

BRAF is implicated in the activation of the MAPK pathway and belongs to the Raf family of kinases. BRAF is a critical factor participating in the pathogenesis of thyroid cancer, and a high frequency of BRAF mutations has been detected among patients with PTC (40). Accumulating evidence suggests that a mutation in BRAF at the amino acid residue 600 (V600E) may be associated with the prognosis of patients with PTC (4143). BRAF may be implicated in PTC progression through several signaling pathways, including the MEK/ERK and the phosphatidylinositol-4,5-bisphosphate 3-kinase/Akt pathway (4446). Notably, Zhang et al (47) demonstrated that inhibition of miR-9 reversed the effects of BRAF-activated non-coding RNA on gastric cancer cell growth and apoptosis, via targeting nuclear factor-κB1.

The results of the present study suggested that miR-9 may suppress the activation of the MEK/ERK signaling pathway via binding to the 3′-UTR of BRAF and suppressing its expression; low expression levels of BRAF may inhibit the activation of MEK/ERK signaling. Inhibition of the MEK/ERK pathway using synthetic pharmacological agents has been demonstrated to inhibit the progression of various types of cancer (48). In addition, BRAF has been associated with cancer development and it may serve an important role in signal transduction through the phosphorylation of MEK1/2 and ERK1/2 (49).

In conclusion, the present study revealed that miR-9 expression was downregulated in PTC tissues, whereas miR-9 overexpression inhibited BRAF expression, suppressed PTC cell proliferation and promoted PTC cell apoptosis. Further studies are required to investigate the molecular mechanisms underlying the pathogenesis of PTC, and identify novel therapeutic approaches for the treatment of patients with PTC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YG designed the research, analyzed data and wrote the manuscript. NY, LY, CF and TL performed the research and analyzed data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Sichuan Provincial People's Hospital (Sichuan, China). Written informed consent was obtained from all patients prior to enrollment in the present study. All protocols of animal experiments were approved by the Animal Ethics Committee of Sichuan Provincial People's Hospital.

Consent for publication

Written informed consent was obtained from all patients prior to enrollment in the present study.

Competing interests

The authors declare that they have no competing interests.

References

1 

Pacini F: Thyroid microcarcinoma. Best Pract Res Clin Endocrinol Metab. 26:381–389. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Pellegriti G, Frasca F, Regalbuto C, Squatrito S and Vigneri R: Worldwide increasing incidence of thyroid cancer: Update on epidemiology and risk factors. J Cancer Epidemiol. 2013:9652122013. View Article : Google Scholar : PubMed/NCBI

3 

Hanley JP, Jackson E, Morrissey LA, Rizzo DM, Sprague BL, Sarkar IN and Carr FE: Geospatial and temporal analysis of thyroid cancer incidence in a rural population. Thyroid. 25:812–822. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Magreni A, Bann DV, Schubart JR and Goldenberg D: The effects of race and ethnicity on thyroid cancer incidence. JAMA Otolaryngol Head Neck Surg. 141:319–323. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Zolotov S: Genetic testing in differentiated thyroid carcinoma: Indications and clinical implications. Rambam Maimonides Med J. 7:28–Jan;2016.doi: 10.5041/RMMJ.10236. View Article : Google Scholar : PubMed/NCBI

6 

Griniatsos J, Tsigris C, Kanakis M, Kaltsas G, Michail O, Dimitriou N, Argyrakopoulou G, Delladetsima I, Kyriakou V, Syriou V, et al: Increased incidence of papillary thyroid cancer detection among thyroidectomies in Greece between 1991 and 2006. Anticancer Res. 29:5163–5169. 2009.PubMed/NCBI

7 

Hakala T, Kellokumpu-Lehtinen P, Kholová I, Holli K, Huhtala H and Sand J: Rising incidence of small size papillary thyroid cancers with no change in disease-specific survival in finnish thyroid cancer patients. Scand J Surg. 101:301–306. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Kuo EJ, Goffredo P, Sosa JA and Roman SA: Aggressive variants of papillary thyroid microcarcinoma are associated with extrathyroidal spread and lymph-node metastases: A population-level analysis. Thyroid. 23:1305–1311. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Ghossein R, Ganly I, Biagini A, Robenshtok E, Rivera M and Tuttle RM: Prognostic factors in papillary microcarcinoma with emphasis on histologic subtyping: A clinicopathologic study of 148 cases. Thyroid. 24:245–253. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Grant CS: Recurrence of papillary thyroid cancer after optimized surgery. Gland Surg. 4:52–62. 2015.PubMed/NCBI

11 

Byeon HK, Ban MJ, Lee JM, Ha JG, Kim ES, Koh YW and Choi EC: Robot-assisted Sistrunk's operation, total thyroidectomy, and neck dissection via a transaxillary and retroauricular (TARA) approach in papillary carcinoma arising in thyroglossal duct cyst and thyroid gland. Ann Surg Oncol. 19:4259–4261. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Lang BH, Ng SH, Lau LL, Cowling BJ, Wong KP and Wan KY: A systematic review and meta-analysis of prophylactic central neck dissection on short-term locoregional recurrence in papillary thyroid carcinoma after total thyroidectomy. Thyroid. 23:1087–1098. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Macedo FI and Mittal VK: Total thyroidectomy versus lobectomy as initial operation for small unilateral papillary thyroid carcinoma: A meta-analysis. Surg Oncol. 24:117–122. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Matsuzu K, Sugino K, Masudo K, Nagahama M, Kitagawa W, Shibuya H, Ohkuwa K, Uruno T, Suzuki A, Magoshi S, et al: Thyroid lobectomy for papillary thyroid cancer: Long-term follow-up study of 1,088 cases. World J Surg. 38:68–79. 2014. View Article : Google Scholar : PubMed/NCBI

15 

He Y, Lin J, Kong D, Huang M, Xu C, Kim TK, Etheridge A, Luo Y, Ding Y and Wang K: Current state of circulating MicroRNAs as cancer biomarkers. Clin Chem. 61:1138–1155. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Zhou X, Du YL, Jin P and Ma F: Bioinformatic analysis of cancer-related microRNAs and their target genes. Yi Chuan. 37:855–864. 2015.PubMed/NCBI

17 

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

18 

Damanakis AI, Eckhardt S, Wunderlich A, Roth S, Wissniowski TT, Bartsch DK and Di Fazio P: MicroRNAs let7 expression in thyroid cancer: Correlation with their deputed targets HMGA2 and SLC5A5. J Cancer Res Clin Oncol. 142:1213–1220. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Czajka AA, Wójcicka A, Kubiak A, Kotlarek M, Bakuła-Zalewska E, Koperski Ł, Wiechno W and Jażdżewski K: Family of microRNA-146 regulates RARβ in papillary thyroid carcinoma. PLoS One. 11:e01519682016. View Article : Google Scholar : PubMed/NCBI

20 

Cong D, He M, Chen S, Liu X and Sun H: Expression profiles of pivotal microRNAs and targets in thyroid papillary carcinoma: An analysis of the cancer genome atlas. Onco Targets Ther. 8:2271–2277. 2015.PubMed/NCBI

21 

Lima CR, Geraldo MV, Fuziwara CS, Kimura ET and Santos MF: MiRNA-146b-5p upregulates migration and invasion of different papillary thyroid carcinoma cells. BMC Cancer. 16:1082016. View Article : Google Scholar : PubMed/NCBI

22 

Minna E, Romeo P, Dugo M, De Cecco L, Todoerti K, Pilotti S, Perrone F, Seregni E, Agnelli L, Neri A, et al: miR-451a is underexpressed and targets AKT/mTOR pathway in papillary thyroid carcinoma. Oncotarget. 7:12731–12747. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Salajegheh A, Vosgha H, Rahman Md A, Amin M, Smith RA and Lam AK: Modulatory role of miR-205 in angiogenesis and progression of thyroid cancer. J Mol Endocrinol. 55:183–196. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Sondermann A, Andreghetto FM, Moulatlet AC, da Silva Victor E, de Castro MG, Nunes FD, Brandão LG and Severino P: MiR-9 and miR-21 as prognostic biomarkers for recurrence in papillary thyroid cancer. Clin Exp Metastasis. 32:521–530. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Gundara JS, Zhao J, Gill AJ, Lee JC, Delbridge L, Robinson BG, McLean C, Serpell J and Sidhu SB: Noncoding RNA blockade of autophagy is therapeutic in medullary thyroid cancer. Cancer Med. 4:174–182. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Ma B, Shi R, Yang S, Zhou L, Qu N, Liao T, Wang Y, Wang Y and Ji Q: DUSP4/MKP2 overexpression is associated with BRAF(V600E) mutation and aggressive behavior of papillary thyroid cancer. Onco Targets Ther. 9:2255–2263. 2016.PubMed/NCBI

27 

Gao Q, Zhang W, Wang N, Duan H, Zhou Y, Zhang W and Zhao D: Study on the correlation between BRAF(V600E) mutation and lymphatic metastases in papillary thyroid cancer staged preoperativelv as N0. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 29:2048–2052. 2015.(In Chinese). PubMed/NCBI

28 

Cordioli MI, Moraes L, Carvalheira G, Sisdelli L, Alves MT, Delcelo R, Monte O, Longui CA, Cury AN and Cerutti JM: AGK-BRAF gene fusion is a recurrent event in sporadic pediatric thyroid carcinoma. Cancer Med. 5:1535–1541. 2016. View Article : Google Scholar : PubMed/NCBI

29 

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

30 

Zhuang G, Wu X, Jiang Z, Kasman I, Yao J, Guan Y, Oeh J, Modrusan Z, Bais C, Sampath D and Ferrara N: Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 31:3513–3523. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Agarwal V, Bell GW, Nam JW and Bartel DP: Predicting effective microRNA target sites in mammalian mRNAs. Elife. 4:12–Aug;2015.doi: 10.7554/eLife.05005. View Article : Google Scholar

32 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2:e3632004. View Article : Google Scholar : PubMed/NCBI

33 

Conzo G, Tartaglia E, Avenia N, Calò PG, de Bellis A, Esposito K, Gambardella C, Iorio S, Pasquali D, Santini L, et al: Role of prophylactic central compartment lymph node dissection in clinically N0 differentiated thyroid cancer patients: Analysis of risk factors and review of modern trends. World J Surg Oncol. 14:1492016. View Article : Google Scholar : PubMed/NCBI

34 

Chruścik A and Lam AK: Clinical pathological impacts of microRNAs in papillary thyroid carcinoma: A crucial review. Exp Mol Pathol. 99:393–398. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Min XS, Huang P, Liu X, Dong C, Jiang XL, Yuan ZT, Mao LF and Chang S: Bioinformatics analyses of significant prognostic risk markers for thyroid papillary carcinoma. Tumour Biol. 36:7457–7463. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Forte S, La Rosa C, Pecce V, Rosignolo F and Memeo L: The role of microRNAs in thyroid carcinomas. Anticancer Res. 35:2037–2047. 2015.PubMed/NCBI

37 

Roberts PJ and Der CJ: Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 26:3291–3310. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Schlumberger M and Sherman SI: Approach to the patient with advanced differentiated thyroid cancer. Eur J Endocrinol. 166:5–11. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Zawistowski JS, Nakamura K, Parker JS, Granger DA, Golitz BT and Johnson GL: MicroRNA 9-3p targets β1 integrin to sensitize claudin-low breast cancer cells to MEK inhibition. Mol Cell Biol. 33:2260–2274. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Ito T, Seyama T, Hayashi Y, Dohi K and Akiyama M: Unique association of p53 mutations with undifferentiated carcinoma of the thyroid. Nihon Rinsho. 52:1069–1074. 1994.(In Japanese). PubMed/NCBI

41 

Falchook GS, Millward M, Hong D, Naing A, Piha-Paul S, Waguespack SG, Cabanillas ME, Sherman SI, Ma B, Curtis M, et al: BRAF inhibitor dabrafenib in patients with metastatic BRAF-mutant thyroid cancer. Thyroid. 25:71–77. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Rothenberg SM, McFadden DG, Palmer EL, Daniels GH and Wirth LJ: Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib. Clin Cancer Res. 21:1028–1035. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Fnais N, Soobiah C, Al-Qahtani K, Hamid JS, Perrier L, Straus SE and Tricco AC: Diagnostic value of fine needle aspiration BRAF(V600E) mutation analysis in papillary thyroid cancer: A systematic review and meta-analysis. Hum Pathol. 46:1443–1454. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Yi W, Zhong D and Zou Q: Expression of BRAF and its extracellular signal-regulated kinase 1/2 signal pathway in papillary thyroid cancer. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 37:889–894. 2012.(In Chinese). PubMed/NCBI

45 

Kandil E, Tsumagari K, Ma J, Elmageed Abd ZY, Li X, Slakey D, Mondal D and Abdel-Mageed AB: Synergistic inhibition of thyroid cancer by suppressing MAPK/PI3K/AKT pathways. J Surg Res. 184:898–906. 2013. View Article : Google Scholar : PubMed/NCBI

46 

McCarty SK, Saji M, Zhang X, Knippler CM, Kirschner LS, Fernandez S and Ringel MD: BRAF activates and physically interacts with PAK to regulate cell motility. Endocr Relat Cancer. 21:865–877. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Zhang ZX, Liu ZQ, Jiang B, Lu XY, Ning XF, Yuan CT and Wang AL: BRAF activated non-coding RNA (BANCR) promoting gastric cancer cells proliferation via regulation of NF-κB1. Biochem Biophys Res Commun. 465:225–231. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Hu Y, Mintz A, Shah SR, Quinones-Hinojosa A and Hsu W: The FGFR/MEK/ERK/brachyury pathway is critical for chordoma cell growth and survival. Carcinogenesis. 35:1491–1499. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Wellbrock C and Arozarena I: The Complexity of the ERK/MAP-kinase pathway and the treatment of melanoma skin cancer. Front Cell Dev Biol. 4:332016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 18 Issue 1

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
Gu Y, Yang N, Yin L, Feng C and Liu T: Inhibitory roles of miR‑9 on papillary thyroid cancer through targeting BRAF. Mol Med Rep 18: 965-972, 2018
APA
Gu, Y., Yang, N., Yin, L., Feng, C., & Liu, T. (2018). Inhibitory roles of miR‑9 on papillary thyroid cancer through targeting BRAF. Molecular Medicine Reports, 18, 965-972. https://doi.org/10.3892/mmr.2018.9010
MLA
Gu, Y., Yang, N., Yin, L., Feng, C., Liu, T."Inhibitory roles of miR‑9 on papillary thyroid cancer through targeting BRAF". Molecular Medicine Reports 18.1 (2018): 965-972.
Chicago
Gu, Y., Yang, N., Yin, L., Feng, C., Liu, T."Inhibitory roles of miR‑9 on papillary thyroid cancer through targeting BRAF". Molecular Medicine Reports 18, no. 1 (2018): 965-972. https://doi.org/10.3892/mmr.2018.9010