Elsevier

Biomedicine & Pharmacotherapy

Volume 105, September 2018, Pages 1232-1239
Biomedicine & Pharmacotherapy

LncRNA MEG3 enhances 131I sensitivity in thyroid carcinoma via sponging miR-182

https://doi.org/10.1016/j.biopha.2018.06.087Get rights and content

Highlights

  • MEG3 expression was down-regulated in 131I-resistant TC cells.

  • MEG3 overexpression inhibited proliferation and induced apoptosis in 131I-resistant TC cells.

  • MEG3 suppressed miR-182 expression as a molecular sponge.

  • MEG3 exerted functions in 131I-resistant TC cells by regulating miR-182.

Abstract

Background

Long non-coding RNA (LncRNA) MEG3 has been demonstrated as a tumor suppressor in various cancers, including thyroid carcinoma (TC). However, the detail functions and possible mechanisms of MEG3 in 131I resistance of TC remain to be uncovered.

Methods

qRT-PCR was performed for the detection of MEG3 and miR-182 levels. 131I-resistant TC cells were constructed by continuous exposure to stepwise increased concentrations of 131I. Western blot assay was used to measure the protein expressions of γ-H2 AX and H2 AX. CCK-8 and flow cytometry assays were carried out for the evaluation of cell viability and apoptosis, respectively. Bioinformatics and dual-luciferse assays were conducted to prove the interaction of MEG3 and miR-182.

Results

MEG3 expression was down-regulated in TC tumor tissues, and the cumulative survival rate was decreased in low MEG3 expression group in TC patients under 131I treatment. MEG3 expression appeared a decline and miR-182 expression displayed an increase in 131I-resistant FTC-133 (res-FTC-133) and TPC-1 (res-TPC-1) cells. Moreover, MEG3 overexpression suppressed 131I-resistant cell viability, promoted apoptosis and induced DNA damage. MEG3 was verified as a molecular sponge for miR-182, and inhibition of miR-182 exerted similar functions as MEG3 overexpression. Furthermore, MEG3 knockdown substantially abrogated the anti-cancer functions of anti-miR-182.

Conclusions

MEG3 enhanced the radiosensitivity of 131I in TC cells via sponging miR-182, indicating that MEG3 may act as a potential biomarker and therapeutic target for TC patients with 131I resistance.

Graphical abstract

The flow chart schematic model. 131I-resistant TC cells induce down-regulation of MEG3. Then, decreased MEG3 resulted in an increase of miR-182 expression. Finally, up-regulated miR-182 enhanced 131I resistance of TC cells by facilitating proliferation, as well as suppressing apoptosis and DNA damage.

  1. Download : Download high-res image (70KB)
  2. Download : Download full-size image

Introduction

Thyroid cancer (TC) is a prevalent malignant tumor in endocrine system with a steadily increasing morbidity and mortality worldwide in the past decades [1,2]. TC has increased by 3% annually from 1974 to 2013 in United States [3], and a similar trend is also observed in China, especially in East China [4]. Specific genetic abnormalities and environmental exposures associated with immunologic functions represent a series of possible risk factors for the occurrence of TC [5]. Radioiodine (131I) has been considered as a common strategy for the treatment of TC, with the potential of destroying the occult lesions in remnant tissues after total or near total thyroidectomy [6]. However, several patients fail to respond to radioiodine ablation therapy due to the deficiency of radioiodine aggregate ability of thyroid follicular cells. Thus, it is urgent to develop novel methods for improving the 131I therapeutic effect.

Long non-coding RNAs (LncRNAs), a novel subgroup of non-protein transcripts with larger than 200 nucleotides in length, are closely involved in the modulation of cell apoptosis, proliferation and invasion through different biological and pathological pathways [7,8]. Mounting evidence has revealed the central participation of lncRNAs in various cancers, including TC [9,10]. Maternally expression gene 3 (MEG3), a lncRNA located on chromosome 14q32, is generally expressed in human normal tissues, while it is lost or decreased in many malignancies [11]. MEG3 have been demonstrated as a tumor-suppressor to be implicated in the etiology and progression of multiple cancers [12]. For instance, MEG3 expression was down-regulated in hepatocellular carcinoma, and MEG3 overexpression blocked cell proliferation at least partly through affecting miR-664-mediated regulation of ADH4 [13]. In TC, MEG3 appeared a decreased expression, and up-regulation of MEG3 strongly weakened cell migration and invasion through negatively regulating Rac1 [14]. However, there is still short of reliable researches on the effects and mechanisms of MEG3 in 131I-resistant TC.

MicroRNAs (miRNA), a type of endogenous and conserved non-coding RNAs, have been widely reported to be associated with the occurance and development of cancers via regulation of special target genes. MiR-182, located in the region of chromosome 7q32.2 of human genome, is revealed as an oncogene in a variety of tumors, such as non-small cell lung cancer [15], prostate cancer [16], breast cancer [17], sarcomas [18], and ovarian cancer [19]. In papillary thyroid cancer (PTC), increased expression of miR-182 was observed in tumor tissues, and knockdown of miR-182 repressed cell proliferation and invasion by targeting close homolog of LI (CHL1) [20]. However, the exact roles of miR-182 in 131I-resistant TC have not been elucidated.

It has been shown that lncRNAs could act as competing endogenous RNAs or molecular sponges of miRNAs to modulate the survival and development of multiple cancer cells [21]. In the present study, we observed that MEG3 expression was significantly reduced TC tumor tissues, and low MEG3 expression was associated with an unfavorable prognosis in TC patients. Then, 131I-resistant TC cells were established successfully, and further functional researches and mechanical assays suggested that MEG3 up-regulation promoted the sensitivity of 131I treatment in TC cells through acting as a sponge for miR-182, as reflected by the depressed viability, increased apoptotic rate, and enhanced DNA damage. In all, our study contributes to a better understanding of the involvement of MEG3 in TC progression, providing a potential target for TC patients with 131I resistance.

Section snippets

Patients and tissue specimens

A total of 20 fresh tumor tissues and adjacent noncancerous tissues were obtained from TC patients, who were undergoing surgical resection at the First People’s Hospital of Shangqiu from June 2016 to July 2017. Following surgery, these specimens were stored in liquid nitrogen at −80 °C for the detection of MEG3 expression.

A total of 48 patients diagnosed with TC were equally divided into low MEG3 expression group and high MEG3 expression group, and all of them received 131I treatment. Finally,

MEG3 level was decreased in TC tumor tissues and low MEG3 expression exhibited a worse prognosis in clinic

To explore the roles of MEG3 in TC, MEG3 levels were firstly detected by qRT-PCR. As illustrated in Fig. 1A, MEG3 displayed a significantly lowered expression in TC tumor tissues compared with that in contiguous normal tissues (n = 20). Additionally, 48 patients who underwent 131I treatment were followed up for 2000-day. The results showed that the rate of cumulative survival was higher in patients with high MEG3 expression than that with low MEG3 expression (Fig. 1B), indicating that MEG3 may

Discussion

It is well known that 131I exposure is a common strategy for TC therapy through inducing various types of DNA damage [22]. Thus, enhancing the sensitivity of TC cells to 131I can greatly improve the therapeutic effects of TC clinically. In recent years, lncRNAs have drawn extensive attention owing to their involvement in a wide range of malignancies [23]. Moreover, increasing lncRNAs have been discovered as important players in thyroid carcinogenesis through modulating different cellular genes [

Conclusion

This study revealed that MEG3 suppressed proliferation, induced apoptosis, and enhanced DNA damage in 131I-resistant TC cells by the negative modulation of miR-182. Therefore, our results imply that MEG3 function as a stabilizer for 131I sensitivity and may serve as a potential therapy target for TC patients with 131I treatment failure.

Conflicts of interest

The authors declare that they have no conflicts of interest.

References (34)

  • V.C. La et al.

    Thyroid cancer mortality and incidence: a global overview

    Int. J. Cancer

    (2015)
  • C.M. Kitahara et al.

    Increases in thyroid cancer incidence and mortality

    JAMA

    (2017)
  • H. Lim et al.

    Trends in thyroid cancer incidence and mortality in the United States, 1974-2013

    JAMA

    (2017)
  • Z.R. Li et al.

    Knockdown of lncRNA-PANDAR suppresses the proliferation, cell cycle and promotes apoptosis in thyroid cancer cells

    EXCLI J.

    (2017)
  • T. Liao et al.

    BRAF-activated LncRNA functions as a tumor suppressor in papillary thyroid cancer

    Oncotarget

    (2016)
  • J.K. Huang et al.

    LncRNA-MALAT1 promotes angiogenesis of thyroid cancer by modulating tumor-associated macrophage FGF2 protein secretion

    J. Cell. Biochem.

    (2017)
  • M. Sun et al.

    Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cellproliferation in gastric cancer

    Tumour Biol.

    (2014)
  • Cited by (40)

    • Genetic predisposition to papillary thyroid carcinoma is mediated by a long non-coding RNA TINCR enhancer polymorphism

      2022, International Immunopharmacology
      Citation Excerpt :

      These findings suggested that AP-2α enriched the enhancer region of TINCR containing the rs8101923, and promoted cell proliferation in PTC. Previous studies have demonstrated that various lncRNAs were differentially expressed in PTC, such as PTCSC3, PTCSC2, maternally expressed gene 3 (MEG3), and BRAF-activated non-protein coding RNA (BANCR), serving as tumor suppressors or oncogenic lncRNAs [8,35–38]. However, there is no report investigating the role of TINCR in the occurrence of PTC.

    • A four-enhancer RNA-based prognostic signature for thyroid cancer

      2022, Experimental Cell Research
      Citation Excerpt :

      Previous Kaplan-Meier curve analysis results have shown that gastric cancer patients with overexpression of NBDY show poor prognosis and silencing of NBDY can repress the proliferation, migration and invasion of gastric cancer cells [25]. Increasing evidence has identified that MEG3 can be used as a potential tool for the early diagnosis of thyroid cancer since the cumulative survival rate is decreased in thyroid cancer patients with low MEG3 expression [26,27]. Therefore, MEG3 might be a diagnostic biomarker for thyroid cancer.

    • Molecular mechanisms of thyroid cancer: A competing endogenous RNA (ceRNA) point of view

      2022, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      MEG3 has a low expression level in TC tissues. In FTC and PTC patients receiving radioiodine treatment, the survival rate rates are much lower in the decreased MEG3 expression class [159]. Besides, MEG3 increased radioiodine sensitivity in FTC and PTC cells through sponging miR-182, implying that MEG3 could be used as a biomarker and treatment option in TC individuals with radioiodine-resistance [159].

    • The contributory role of long non-coding RNAs (lncRNAs) in head and neck cancers: Possible biomarkers and therapeutic targets?

      2021, European Journal of Pharmacology
      Citation Excerpt :

      Down-regulation of the tumor suppressor lncRNA MEG3 was shown in patients with radioiodine-resistant 131I-resistant FTC-133 (res-FTC-133) and TPC-1 (res-TPC-1) TC cells. Liu et al. demonstrated that MEG3 overexpression could enhance radioactive iodine (RAI) therapy in TC cells via the suppression of cell proliferation, as well as the stimulation of apoptosis and DNA damage (Liu et al., 2018). RAI resistance in PTC has also been associated with NEAT1.

    • The cross-talk between signaling pathways, noncoding RNAs and DNA damage response: Emerging players in cancer progression

      2021, DNA Repair
      Citation Excerpt :

      One of the standard therapies for thyroid carcinoma (TC) is tumor cells' exposure to 131I, which results in apoptosis and DNA damage in affected TC cells. A recent study revealed that up-regulated lncRNA MEG3 acts as a ceRNAs (competing endogenous RNAs), and through sponging miR-182, the expression level of DNA damage protein γ-H2AX increases, consequently enhances DNA damage, apoptotic rate, and improvement of 131I sensitivity in TC [35]. MiRNAs are short and single-stranded RNA molecules with 20–22 nucleotides in length.

    View all citing articles on Scopus
    View full text