Thyroid cancer is a rare malignancy and accounts for less than 1% of malignant neoplasms in humans; however, it is the most common cancer of the endocrine system and responsible for most deaths from endocrine cancer. Long non-coding (Lnc)RNAs are defined as non-coding transcripts that are more than 200 nucleotides in length. Their expression deregulation plays an important role in the progress of cancer. These molecules are involved in physiologic cellular processes, genomic imprinting, inactivation of chromosome X, maintenance of pluripotency, and the formation of different organs via changes in chromatin, transcription, and translation. LncRNAs can act as a tumor suppressor genes or oncogenes. Several studies have shown that these molecules can interact with microRNAs and prevent their binding to messenger RNAs. Research has shown that these molecules play an important role in tumorigenicity, angiogenesis, proliferation, migration, apoptosis, and differentiation. In thyroid cancer, several lncRNAs (MALAT1, H19, BANCR, HOTAIR) have been identified as contributing factors to cancer development, and can be used as novel biomarkers for early diagnosis or even treatment. In this article, we study the newest lncRNAs and their role in thyroid cancer.

Two identifiable cell types are observed in the normal parenchymal histology of the thyroid gland. The first type comprises follicular cells that surround the colloid follicles and uptake iodine to synthesize thyroid hormones; anaplastic thyroid and well-differentiated tumors such as papillary and follicular can develop in these cells. The second type comprises parafollicular cells whose main function is to secrete calcitonin; parafollicular cells have attracted interest primarily as the origin of medullary thyroid cancer (MTC) [1]. Thyroid cancer (TC) is the 9th most common malignancy and the most common endocrine cancer [2]. The most common treatment for TC without metastasis is surgery. Chemotherapy and radioactive iodine treatment are also medical treatment options in addition to surgery [3]. However, the prognosis of recurrent TC is not good with the above treatments [4]. Histologic grade and age at onset are 2 major prognostic factors [5]. Ultrasound and fine-needle aspiration (FNA) biopsy of suspicious nodules are routinely employed to detect TC. Identification of novel biomarkers is necessary to improve the management of papillary thyroid cancer (PTC) and the decision-making regarding the therapeutic approach. Long non-coding RNAs (lncRNA) are an important part of the newly discovered non-coding RNAs (ncRNAs) [6]. Increasing evidence suggests that lncRNA play a key regulatory role in the genome and important functional roles in different fundamental biologic processes in transcription and translation. The challenge in TC diagnosis and treatment is to identify patients that have a high risk of developing radioiodine-refractory disease on the one hand and to prevent overtreatment on the other hand. While the clinical risk stratifications proposed by the American Thyroid Association are very helpful in guiding decision-making, it would be beneficial to have additional biomarkers such as potentially lncRNAs. The novel TC biomarkers give insight into how the disease begins and progresses while improving the success rate of early diagnosis and treatment. In TC, gene mutations, proteins, and messenger RNA (mRNA) markers such as BRAF, RAS, RET/PTC, and PAX8/PPARγ have been used to increase the diagnostic accuracy of patients with thyroid nodules [4]. Several microRNAs (miRNAs) are associated with PTC [7]. According to American Thyroid Association guidelines, the use of molecular biomarkers for TC is recommended for FNA samples with an unclear cytological diagnosis [8]. Several lncRNAs, such as NAMA, BANCR, and PTCSC3, have been identified as contributing factors to PTC [9, 10, 11] and may therefore be used as biomarker in this setting (table 1).

Table 1

Detailed characteristics based on the findings of included studies

Detailed characteristics based on the findings of included studies
Detailed characteristics based on the findings of included studies

LncRNAs are small classes of transcribed molecules that range from 200 nucleotides to 100 kb in length [11]. LncRNAs account for about 80% of ncRNAs and play an important role in genomic organization. In cancerous tissues, there is a clear relationship between the expression and epigenetic state of these molecules. These molecules can interact with chromatin-modifying enzymes and histones, including DNA methyl transferase 3 and PRC2 [12]. LncRNAs can act as tumor suppressors or oncogenes in cancer [13]. Research has shown that these molecules play an important role in tumorigenicity, angiogenesis, proliferation, migration, apoptosis, and differentiation [14]. LncRNAs induce epithelial-mesenchymal transition (EMT) through the PI3K-AKT and Wnt/β-catenin pathways, thereby promoting metastasis [15]. A number of studies have shown that lncRNAs can interact with miRNAs by preventing the binding of miRNAs to mRNA targets [16]. LncRNAs have different expression levels at different stages of differentiation and can be used as biomarkers to identify the disease according to their specific expression in the tissue [17].

Nuclease-based genome editing technology is a powerful tool for clarifying the function of lncRNA in both in vitro and in vivo conditions [18]. The CRISPR-CAS9 system rapidly and efficiently removes relative or complete lncRNA [19]. On the other hand, inhibition of lncRNA expression has also been accomplished via inter-promoter polyadenylation and lncRNA sequences [20]. CRISPR-CAS9 also has the ability to increase lncRNA expression by introducing a strong promoter to the upstream gene or through targeting the transcriptional activation complex [21]. These systems play a significant role in increasing and reducing the expression of lncRNA in cancer, leading to new therapeutic approaches.

BRAF-activated non-protein coding RNA (BANCR) is a 693 bp lncRNA primarily identified using vastly parallel cDNA sequencing in melanoma cells. It is highly expressed in melanoma and increases cell migration [22]. Studies have shown that BANCR functions as both an oncogene and a tumor suppressor. Wang et al. [23]showed that BANCR was overexpressed in 6 pairs of PTC and matching normal tissues and that it increased PTC cell proliferation by stimulating autophagy. In the study by Liao et al. [10]BANCR expression was downregulated in PTC tissues compared with normal tissues. The significance of BANCR in TC is still unclear. BANCR is ordinarily activated by BRAF. The BRAF V600E mutation is very frequent in PTCs but the association with poorer prognosis is very controversial. However, studies have shown that BRAFV600E in combination with TERT promoter mutations is linked to poorer prognosis in PTC patients [24]. BRAF-activated BANCR has been shown to increase cell migration and proliferation in melanoma [25] and lung carcinoma [26] via the MAPK signaling pathway. It was believed that BANCR can progress EMT in PTC. In TC, there are 2 classical cell signaling pathways: ERK/MAPK and phosphoinositide 3-kinase/protein kinase B. A number of studies have shown that the V600E mutation of BRAF stimulates the MAPK signaling pathway [27]. Overexpression of BANCR might upregulate the expression of p-c-Raf, p-MEK1/2, and p-ERK1/2. Therefore, BANCR might activate the Raf/MEK/ERK signaling pathway. This effect was reversed by U0126 treatment [28]. Downregulated BANCR expression was correlated with tumor size, multifocal lesions, and an advanced pathologic stage. This change might stem from tumor heterogeneity. Low BANCR levels are associated with poor results in PTC patients. Liao et al. [10]indicated that overexpression of BANCR elevates apoptosis in TPC1 and K1 cell lines.

H19 is a typical lncRNA that is associated with various cancer types. H19 was originally thought to possess tumor suppressive properties based on its capacity to prevent tumorigenesis, but current studies have shown that it has both tumor promoter and suppressive functions [29, 30, 31]. There is a high level of expression of H19 in extraembryonic tissues, the embryo itself, and most fetal tissues, but postnatal expression of H19 is highly reduced [32]. In bladder cancer, H19 is a tumor recurrence marker and its expression is correlated with tumor grade, and in hepatocellular carcinoma, ectopic expression of H19 stimulates tumorigenesis, suggesting that H19 acts as an oncogene [33]. A recent study indicated that H19 expression was higher in tumor tissues and TC cell lines than in normal thyroid cells. In addition, it was stated that overexpression of H19 elevated proliferation, migration, and invasion of TC cells, whereas low expression of H19 reduced cell viability, migration, and invasion and caused stimulated growth arrest in vitro and in vivo [34].

Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1) is an lncRNA marker of metastasis and poor prognosis in lung and other cancers [35]. Prior studies have shown that MALAT1-deficient lung cancer cells showed inferior motility and decreased tumor formation in inoculated mice [36]. Moreover, the pro-oncogenic possibility of MALAT1 was supported by its regulation of cell cycle-related transcription factors B-MYB and p53 [37]. Since its characterization in lung cancer, MALAT1 upregulation has also been described in several tumor types including follicular cell-derived thyroid tumors [38], although MALAT1 has remained unknown in MTC. MALAT1, an lncRNA that is involved in the regulation of cell cycle and migration, has been identified to be deregulated in malignancies of multiple organs, including lung, cervix, uterus, colon, breast, stomach, pancreas, kidney, bone, and bladder [39]. The recent study by Huang et al. [40] on thyroid carcinoma cell lines suggested that the follicular thyroid carcinoma (FTC) cell line FTC133 expressed higher levels of MALAT1 than the SW1736 anaplastic thyroid carcinoma (ATC) cell line. Studies have indicated that PTC had higher levels of MALAT1 than FTC and ATC [38]. MALAT1 expression was higher in MTC compared to normal thyroid, and its inhibition produced in vitro anti-oncogenic effects including decreased tumor cell proliferation and invasion [41]. It is worth noting that formerly, in murine mammary and lung cancer models, MALAT1 knockdown by genetic knockout or systemic administration of antisense oligonucleotides was shown to repress tumor growth and metastasis [42], validating the role of MALAT1 as a pro-oncogenic lncRNA in several cancer types. In addition, MALAT1 was demonstrated to promote EMT by epigenetically silencing E-cadherin expression [43]. MALAT1 was remarkably upregulated through transforming growth factor beta (TGFβ)-induced EMT in TC cells [38]. MALAT1 upregulation was found in the primary stage and could affect cell invasion [41].

Homeobox transcript antisense RNA (HOTAIR) is a 2,000 bp lncRNA that encodes antisense to the HOXC locus [44]. HOTAIR acts as an oncogene in tumorigenesis and stimulates invasion and metastases in tumor progression [45]. Dysregulation of HOTAIR is correlated with metastasis and poor prognosis in patients with TC. Through loss-of-function analysis, the biologic function of HOTAIR has been validated in human TC cell lines. The expression of HOTAIR is upregulated in human TC cells compared with normal human thyroid cells. Moreover, knockdown of HOTAIR significantly inhibited cell growth and invasion in TPC-1 and SW579 human TC [46].

Papillary thyroid carcinoma susceptibility candidate 3 (PTCSC3) is a recently recognized ncRNA, which is highly thyroid-specific. PTCSC3 as a tumor suppressor was considered a competing endogenous RNA for miR-574-5p [47]. PTCSC3 expression is strictly thyroid-specific and is downregulated in thyroid tumor tissues and thyroid cell lines.

Maternally expressed gene 3 (MEG3) is a lncRNA gene expressed in several normal tissues. Loss of MEG3 expression has been found in several types of human tumors. The regulation of MEG3 and its exact mechanisms of action in TC are unknown. MEG3 is downregulated in PTC; downregulated MEG3 was significantly associated with lymph node metastasis. MEG3 is an important tumor suppressor in TC which suppresses migration and invasion by targeting Rac1 [48].

Antisense non-coding RNA in the INK4 locus (ANRIL) has been proven to bind to chromobox 7 (CBX7) and to SUZ12, and is involved in transcriptional suppression through these interactions. Silencing ANRIL, as a key lncRNA, inhibits the invasion and metastasis of TPC1 cells. Moreover, ANRIL may prevent the expression of the TGFβ/Smad signaling pathway to decrease p15INK4B expression and stimulate invasion and metastasis of TC cells, which provides a novel perception of the practical role of lncRNA ANRIL-driven tumorigenesis [49]. LncRNAs (ENST00000426615 and ENST00000537266) may be significant regulators of PTC cell proliferation and motility, which could provide new insights into PTC pathogenesis [50]. Downregulation of lncRNA PANDAR suppresses cell growth/cell cycle and produces apoptosis in TC [51].

Many studies have shown that lncRNAs have wide biologic activity, especially in cell growth, differentiation, and tumorigenicity. LncRNAs may be a worthy biomarker for the diagnosis and prognosis of cancer. These molecules are expressed naturally in different cell types and can act as oncogenes or tumor suppressors, and play an important role in controlling cell growth through regulation of the cell cycle and apoptosis. Increases or decreases in their expression can lead to various types of cancer, such as TC. These molecules can be used as novel biomarkers in diagnosis or as prognostic factors for various types of cancer, without the need for invasive methods. However, to elucidate the regulatory role of these molecules, more research is needed in the future. TC is a disease that develops due to genetic and epigenetic abnormalities. Several classical disorders that are involved in tumorigenesis and progression of TC include changes in point mutations, copy number variations, and DNA methylation. In addition, many studies have shown that lncRNAs also play a role in regulating all types of biochemical activity in TC cells, including cell growth and survival. This study gives a summary of disorders affecting regulation and of recent developments regarding lncRNAs in TC. Given that lncRNAs have advantages such as noninvasiveness and easy access, they have a high potential to be utilized in the diagnosis and treatment evaluation of TC. Moreover, there are other aspects of lncRNAs in TC that will be investigated in the near future. For example, FNA biopsy and cytological analysis are a primary diagnostic measure that is widely used in the evaluation of thyroid nodules. However, at least 20% of biopsies show unclear cytological findings that cannot distinguish malignant from benign nodules, leading to poor management for these patients. Detection of cancer-related lncRNAs in FNA biopsies can be a useful strategy for the detection of malignant thyroid tumors. The study of lncRNAs in TC is becoming an attractive and useful field. The role and mechanism of lncRNAs in TC should be investigated in the future, and some lncRNAs may serve as biomarkers for diagnosis, prognosis, and therapeutic purposes.

The authors declare that they have no competing interests.

This article does not contain any studies with human participants or animals performed by any of the authors.

1.
Carling T, Udelsman R: Thyroid cancer. Annu Rev Med 2014;65:125-137.
[PubMed]
2.
Pellegriti G, Frasca F, Regalbuto C, Squatrito S, Vigneri R: Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. J Cancer Epidemiol 2013;2013:965212.
[PubMed]
3.
Grewal RK, Ho A, Schoder H: Novel approaches to thyroid cancer treatment and response assessment. Semin Nucl Med 2016;46:109-118.
[PubMed]
4.
Schneider DF, Chen H: New developments in the diagnosis and treatment of thyroid cancer. CA Cancer J Clin 2013;63:374-394.
[PubMed]
5.
Shah JP: Thyroid carcinoma: epidemiology, histology, and diagnosis. Clin Adv Hematol Oncol 2015;13(4 suppl 4):3-6.
[PubMed]
6.
Kung JT, Colognori D, Lee JT: Long noncoding RNAs: past, present, and future. Genetics 2013;193:651-669.
[PubMed]
7.
Mahmoudian-Sani MR, Mehri-Ghahfarrokhi A, Asadi-Samani M, Mobini GR: Serum miRNAs as biomarkers for the diagnosis and prognosis of thyroid cancer: a comprehensive review of the literature. Eur Thyroid J 2017;6:171-177.
[PubMed]
8.
Ferris RL, Baloch Z, Bernet V, et al.: American Thyroid Association Statement on Surgical Application of Molecular Profiling for Thyroid Nodules: current impact on perioperative decision making. Thyroid 2015;25:760-768.
[PubMed]
9.
Goedert L, Placa JR, Fuziwara CS: Identification of long noncoding RNAs deregulated in papillary thyroid cancer and correlated with BRAF (V600E) mutation by bioinformatics integrative analysis. Sci Rep 2017;7:1662.
[PubMed]
10.
Liao T, Qu N, Shi RL, et al.: BRAF-activated LncRNA functions as a tumor suppressor in papillary thyroid cancer. Oncotarget 2017;8:238-247.
[PubMed]
11.
Wang XM, Liu Y, Fan YX, et al.: LncRNA PTCSC3 affects drug resistance of anaplastic thyroid cancer through STAT3/INO80 pathway. Cancer Biol Ther 2018;19:590-597.
[PubMed]
12.
Nagano T, Mitchell JA, Sanz LA, et al.: The Air noncoding RNA epigenetically silences transcription by targeting G9a to chromatin. Science 2008;322:1717-1720.
[PubMed]
13.
Rossi S, Sevignani C, Nnadi SC, Siracusa LD, Calin GA: Cancer-associated genomic regions (CAGRs) and noncoding RNAs: bioinformatics and therapeutic implications. Mamm Genome 2008;19:526-540.
[PubMed]
14.
Di Gesualdo F, Capaccioli S, Lulli M: A pathophysiological view of the long non-coding RNA world. Oncotarget 2014;5:10976-10996.
[PubMed]
15.
Xu S, Sui S, Zhang J, et al.: Downregulation of long noncoding RNA MALAT1 induces epithelial-to-mesenchymal transition via the PI3K-AKT pathway in breast cancer. Int J Clin Exp Pathol 2015;8:4881-4891.
[PubMed]
16.
Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP: A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 2010;465:1033-1038.
[PubMed]
17.
Yarmishyn AA, Kurochkin IV: Long noncoding RNAs: a potential novel class of cancer biomarkers. Front Genet 2015;6:145.
[PubMed]
18.
Wiedenheft B, Sternberg SH, Doudna JA: RNA-guided genetic silencing systems in bacteria and archaea. Nature 2012;482:331-338.
[PubMed]
19.
Han J, Zhang J, Chen L, et al.: Efficient in vivo deletion of a large imprinted lncRNA by CRISPR/Cas9. RNA Biology 2014;11:829-835.
[PubMed]
20.
Eissmann M, Gutschner T, Hammerle M, et al.: Loss of the abundant nuclear non-coding RNA MALAT1 is compatible with life and development. RNA Biol 2012;9:1076-1087.
[PubMed]
21.
Konermann S, Brigham MD, Trevino AE, et al.: Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 2015;517:583.
[PubMed]
22.
Flockhart RJ, Webster DE, Qu K, et al.: BRAFV600E remodels the melanocyte transcriptome and induces BANCR to regulate melanoma cell migration. Genome Res 2012;22:1006-1014.
[PubMed]
23.
Wang Y, Guo Q, Zhao Y, et al.: BRAF-activated long non-coding RNA contributes to cell proliferation and activates autophagy in papillary thyroid carcinoma. Oncol Lett 2014;8:1947-1952.
[PubMed]
24.
Xing M, Liu R, Liu X, et al.: BRAF V600E and TERT promoter mutations cooperatively identify the most aggressive papillary thyroid cancer with highest recurrence. J Clin Oncol 2014;32:2718.
[PubMed]
25.
Li R, Zhang L, Jia L, et al.: Long non-coding RNA BANCR promotes proliferation in malignant melanoma by regulating MAPK pathway activation. PloS One 2014;9:e100893.
[PubMed]
26.
Jiang W, Zhang D, Xu B, et al.: Long non-coding RNA BANCR promotes proliferation and migration of lung carcinoma via MAPK pathways. Biomed Pharmacother 2015;69:90-95.
[PubMed]
27.
Fraser S, Go C, Aniss A, et al.: BRAFV600E mutation is associated with decreased disease-free survival in papillary thyroid cancer. World J Surg 2016;40:1618-1624.
[PubMed]
28.
Wang Y, Gu J, Lin X, Yan W, Yang W, Wu G: lncRNA BANCR promotes EMT in PTC via the Raf/MEK/ERK signaling pathway. Oncol Lett 2018;15:5865-5870.
[PubMed]
29.
Hao Y, Crenshaw T, Moulton T, Newcomb E, Tycko B: Tumour-suppressor activity of H19 RNA. Nature 1993;365:764-767.
[PubMed]
30.
Luo M, Li Z, Wang W, Zeng Y, Liu Z, Qiu J: Upregulated H19 contributes to bladder cancer cell proliferation by regulating ID2 expression. FEBS J 2013;280:1709-1716.
[PubMed]
31.
Lv J, Ma L, Chen XL, Huang XH, Wang Q: Downregulation of LncRNAH19 and MiR-675 promotes migration and invasion of human hepatocellular carcinoma cells through AKT/GSK-3beta/Cdc25A signaling pathway. J Huazhong Univ Sci Technolog Med Sci 2014;34:363-369.
[PubMed]
32.
Tabano S, Colapietro P, Cetin I, et al.: Epigenetic modulation of the IGF2/H19 imprinted domain in human embryonic and extra-embryonic compartments and its possible role in fetal growth restriction. Epigenetics 2010;5:313-324.
[PubMed]
33.
Matouk IJ, DeGroot N, Mezan S, et al.: The H19 non-coding RNA is essential for human tumor growth. PloS One 2007;2:e845.
[PubMed]
34.
Liu L, Yang J, Zhu X, Li D, Lv Z, Zhang X: Long noncoding RNA H19 competitively binds miR-17-5p to regulate YES1 expression in thyroid cancer. FEBS J 2016;283:2326-2339.
[PubMed]
35.
Schmidt LH, Spieker T, Koschmieder S, et al.: The long noncoding MALAT-1 RNA indicates a poor prognosis in non-small cell lung cancer and induces migration and tumor growth. J Thorac Oncol 2011;6:1984-1992.
[PubMed]
36.
Tano K, Mizuno R, Okada T, et al.: MALAT-1 enhances cell motility of lung adenocarcinoma cells by influencing the expression of motility-related genes. FEBS Lett 2010;584:4575-4580.
[PubMed]
37.
Tripathi V, Shen Z, Chakraborty A, et al.: Long noncoding RNA MALAT1 controls cell cycle progression by regulating the expression of oncogenic transcription factor B-MYB. PLoS Genet 2013;9:e1003368.
[PubMed]
38.
Zhang R, Hardin H, Huang W, et al.: MALAT1 long non-coding RNA expression in thyroid tissues: analysis by in situ hybridization and real-time PCR. Endocr Pathol 2017;28:7-12.
[PubMed]
39.
Ma XY, Wang JH, Wang JL, Ma CX, Wang XC, Liu FS: Malat1 as an evolutionarily conserved lncRNA, plays a positive role in regulating proliferation and maintaining undifferentiated status of early-stage hematopoietic cells. BMC Genomics 2015;16:676.
[PubMed]
40.
Huang JK, Ma L, Song WH, et al.: MALAT1 promotes the proliferation and invasion of thyroid cancer cells via regulating the expression of IQGAP1. Biomed Pharmacother 2016;83:1-7.
[PubMed]
41.
Chu Y-H, Hardin H, Schneider DF, Chen H, Lloyd RV: MicroRNA-21 and long non-coding RNA MALAT1 are overexpressed markers in medullary thyroid carcinoma. Exp Mol Pathol 2017;103:229-236.
[PubMed]
42.
Arun G, Diermeier S, Akerman M, et al.: Differentiation of mammary tumors and reduction in metastasis upon Malat1 lncRNA loss. Genes Dev 2016;30:34-51.
[PubMed]
43.
Hirata H, Hinoda Y, Shahryari V, et al.: Long noncoding RNA MALAT1 promotes aggressive renal cell carcinoma through Ezh2 and interacts with miR-205. Cancer Res 2015;75:1322-1331.
[PubMed]
44.
Gupta RA, Shah N, Wang KC, et al.: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010;464:1071-1076.
[PubMed]
45.
Yu X, Li Z: Long non-coding RNA HOTAIR: a novel oncogene (review). Mol Med Rep 2015;12:5611-5618.
[PubMed]
46.
Zhang Y, Yu S, Jiang L, Wang X, Song X: HOTAIR is a promising novel biomarker in patients with thyroid cancer. Exp Ther Med 2017;13:2274-2278.
[PubMed]
47.
Fan M, Li X, Jiang W, Huang Y, Li J, Wang Z: A long non-coding RNA, PTCSC3, as a tumor suppressor and a target of miRNAs in thyroid cancer cells. Exp Ther Med 2013;5:1143-1146.
[PubMed]
48.
Wang C, Yan G, Zhang Y, Jia X, Bu P: Long non-coding RNA MEG3 suppresses migration and invasion of thyroid carcinoma by targeting of Rac1. Neoplasma 2015;62:541-549.
[PubMed]
49.
Zhao JJ, Hao S, Wang LL, et al.: Long non-coding RNA ANRIL promotes the invasion and metastasis of thyroid cancer cells through TGF-beta/Smad signaling pathway. Oncotarget 2016;7:57903-57918.
[PubMed]
50.
Xu B, Shao Q, Xie K, et al.: The long non-coding RNA ENST00000537266 and ENST00000426615 influence papillary thyroid cancer cell proliferation and motility. Cell Physiol Biochem 2016;38:368-378.
[PubMed]
51.
Li Z, Gao B, Hao S, et al.: Knockdown of lncRNA-PANDAR suppresses the proliferation, cell cycle and promotes apoptosis in thyroid cancer cells. EXCLI J 2017;16:354-362.
[PubMed]
52.
Zhang J, Du Y, Zhang X, Li M, Li X: Downregulation of BANCR promotes aggressiveness in papillary thyroid cancer via the MAPK and PI3K pathways. J Cancer 2018;9:1318-1328.
[PubMed]
53.
Chen B, Li J, Yi C, Jiao Y, Gu X, Feng: Long non-coding RNA POLR2E rs3787016 is associated with the risk of papillary thyroid carcinoma in Chinese population. Pathol Res Pract 2018;214:1040-1044.
[PubMed]
54.
Ding S, Qu W, Jiao Y, Zhang J, Zhang C, Dang S: LncRNA SNHG12 promotes the proliferation and metastasis of papillary thyroid carcinoma cells through regulating wnt/beta-catenin signaling pathway. Cancer Biomark 2018;22:217-226.
[PubMed]
55.
Liao D, Lv G, Wang T, Min J, Wang Y, Liu S: Prognostic value of long non-coding RNA BLACAT1 in patients with papillary thyroid carcinoma. Cancer Cell Int 2018;18:47.
[PubMed]
56.
Sun W, Lan X, Zhang H, et al.: NEAT1_2 functions as a competing endogenous RNA to regulate ATAD2 expression by sponging microRNA-106b-5p in papillary thyroid cancer. Cell Death Dis 2018;9:380.
[PubMed]
57.
Liang L, Xu J, Wang M, et al.: LncRNA HCP5 promotes follicular thyroid carcinoma progression via miRNAs sponge. Cell Death Dis 2018;9:372.
[PubMed]
58.
Yuan Q, Liu Y, Fan Y, et al.: LncRNA HOTTIP promotes papillary thyroid carcinoma cell proliferation, invasion and migration by regulating miR-637. Int J Biochem Cell Biol 2018;98:1-9.
[PubMed]
59.
Ye Y, Song Y, Zhuang J, He S, Ni J, Xia W: Long non-coding RNA CCAL promotes papillary thyroid cancer progression by activation of NOTCH1 pathway. Oncol Res 2018;Epub ahead of print.
[PubMed]
60.
Xia E, Bhandari A, Shen Y, et al.: LncRNA CCND2-AS1 promotes proliferation, migration, and invasion in papillary thyroid carcinoma. Biochem Biophys Res Commun 2018;496:628-632.
[PubMed]
61.
Dai W, Tian Y, Jiang B, Chen W: Down-regulation of long non-coding RNA AFAP1-AS1 inhibits tumor growth, promotes apoptosis and decreases metastasis in thyroid cancer. Biomed Pharmacother 2018;99:191-197.
[PubMed]
62.
Zhao Y, Wang H, Wu C, et al.: Construction and investigation of lncRNA-associated ceRNA regulatory network in papillary thyroid cancer. Oncol Rep 2018;39:1197-1206.
[PubMed]
63.
Qin Y, Sun W, Zhang H, et al.: LncRNA GAS8-AS1 inhibits cell proliferation through ATG5-mediated autophagy in papillary thyroid cancer. Endocrine 2018;59:555-564.
[PubMed]
64.
Lan X, Sun W, Dong W, et al.: Downregulation of long noncoding RNA H19 contributes to the proliferation and migration of papillary thyroid carcinoma. Gene 2018;646:98-105.
[PubMed]
65.
Liu N, Zhou Q, Qi YH, Wang H, Yang L, Fan QY: Effects of long non-coding RNA H19 and microRNA let7a expression on thyroid cancer prognosis. Exp Mol Pathol 2017;103:71-77.
[PubMed]
66.
Zhang R, Hardin H, Huang W, Buehler D, Lloyd RV: Long non-coding RNA Linc-ROR is upregulated in papillary thyroid carcinoma. Endocr Pathol 2018;29:1-8.
[PubMed]
67.
Zhou H, Sun Z, Li S, Wang X, Zhou X: LncRNA SPRY4-IT was concerned with the poor prognosis and contributed to the progression of thyroid cancer. Cancer Gene Ther 2018;25:39-46.
68.
Huang JK, Ma L: LncRNA-MALAT1 promotes angiogenesis of thyroid cancer by modulating tumor-associated macrophage FGF2 protein secretion. J Cell Biochem 2017;118:4821-4830.
[PubMed]
69.
Di W, Li Q, Shen W, Guo H, 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 2017;7:1298-1309.
[PubMed]
70.
Li HM, Yang H, Wen DY, et al.: Overexpression of LncRNA HOTAIR is associated with poor prognosis in thyroid carcinoma: a study based on TCGA and GEO data. Horm Metab Res 2017;49:388-399.
[PubMed]
71.
Liu K, Huang W, Yan DQ, Luo Q, Min X: Overexpression of long intergenic noncoding RNA LINC00312 inhibits the invasion and migration of thyroid cancer cells by down-regulating microRNA-197-3p. Biosci Rep 2017;37.
[PubMed]
72.
Guo LJ, Zhang S, Gao B, et al.: Low expression of long non-coding RNA GAS5 is associated with poor prognosis of patients with thyroid cancer. Exp Mol Pathol 2017;102:500-504.
[PubMed]
73.
Ma B, Liao T, Wen D, et al.: Long intergenic non-coding RNA 271 is predictive of a poorer prognosis of papillary thyroid cancer. Sci Rep 2016;6:36973.
[PubMed]
74.
Xiang C, Zhang ML, Zhao QZ, et al.: LncRNA-SLC6A9-5:2: a potent sensitizer in 131I-resistant papillary thyroid carcinoma with PARP-1 induction. Oncotarget 2017;8:22954-22967.
[PubMed]
75.
Sun W, Lan X, Wang Z, et al.: Overexpression of long non-coding RNA NR_036575.1 contributes to the proliferation and migration of papillary thyroid cancer. Med Oncol 2016;33:102.
[PubMed]
76.
Fan M, Li X, Jiang W, Huang Y, Li J, Wang Z: A long non-coding RNA, PTCSC3, as a tumor suppressor and a target of miRNAs in thyroid cancer cells. Exp Ther Med 2013;5:1143-1146.
[PubMed]