Next Article in Journal
YAP/TAZ Signalling in Colorectal Cancer: Lessons from Consensus Molecular Subtypes
Next Article in Special Issue
LncRNAs in Cancer: From garbage to Junk
Previous Article in Journal
The Innate Immune Signalling Pathways: Turning RIG-I Sensor Activation against Cancer
Previous Article in Special Issue
Prognostic Stratification of Bladder Cancer Patients with a MicroRNA-Based Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Roles of Non-Coding RNAs on Anaplastic Thyroid Carcinomas

1
Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh
2
School of Medicine, Griffith University, Gold Coast, QLD 4222, Australia
3
Department of Endocrinology and Diabetes Mellitus, Fukuoka University Chikushi Hospital, Chikushino, Fukuoka 818-8502, Japan
4
Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, QLD 4222, Australia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 28 September 2020 / Revised: 23 October 2020 / Accepted: 24 October 2020 / Published: 28 October 2020
(This article belongs to the Collection Regulatory and Non-Coding RNAs in Cancer Epigenetic Mechanisms)

Abstract

:

Simple Summary

Anaplastic thyroid carcinoma (ATC) is a biological aggressive human carcinoma and causes most of the thyroid cancer related deaths. Non-protein producing RNAs, such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), play crucial roles in formation and therapy resistance of ATC by regulating the production of key proteins. These RNAs alter the proteome of cells, which in turn controls cancer cells’ growth, division, invasion, migration, metastasis, and recurrence of ATC. Thus, the exogenous manipulation of these RNAs could interfere cells growth and development of ATC. Considering this, we discuss the roles of various miRNAs and lncRNAs in the pathogenesis of ATC and their potential therapeutic applications.

Abstract

Anaplastic thyroid cancer (ATC) remains as one of the most aggressive human carcinomas with poor survival rates in patients with the cancer despite therapeutic interventions. Novel targeted and personalized therapies could solve the puzzle of poor survival rates of patients with ATC. In this review, we discuss the role of non-coding RNAs in the regulation of gene expression in ATC as well as how the changes in their expression could potentially reshape the characteristics of ATCs. A broad range of miRNA, such as miR-205, miR-19a, miR-17-3p and miR-17-5p, miR-618, miR-20a, miR-155, etc., have abnormal expressions in ATC tissues and cells when compared to those of non-neoplastic thyroid tissues and cells. Moreover, lncRNAs, such as H19, Human leukocyte antigen (HLA) complex P5 (HCP5), Urothelial carcinoma-associated 1 (UCA1), Nuclear paraspeckle assembly transcript 1 (NEAT1), etc., participate in transcription and post-transcriptional regulation of gene expression in ATC cells. Dysregulations of these non-coding RNAs were associated with development and progression of ATC by modulating the functions of oncogenes during tumour progression. Thus, restoration of the abnormal expression of these miRNAs and lncRNAs may serve as promising ways to treat the patients with ATC. In addition, siRNA mediated inhibition of several oncogenes may act as a potential option against ATC. Thus, non-coding RNAs can be useful as prognostic biomarkers and potential therapeutic targets for the better management of patients with ATC.

1. Introduction

Anaplastic thyroid carcinoma (ATC) accounts for 1–2% of all thyroid cancers, however, it is responsible for the most cancer deaths caused by thyroid cancer despite therapeutic intervention [1,2]. ATC is one of the most aggressive solid tumours, which contributes to 3.6% of all human cancers. Many patients with ATC die within a year of diagnosis [3,4,5,6]. Several cellular, genetic, and epigenetic aberrations have been reported to orchestrate the tumourigenesis of ATC [7,8]. These aberrations lead to the dysregulation of cellular signalling which as a result enhances cellular proliferation and accelerates tumourigenesis. In contrast to differentiated thyroid carcinoma such as papillary thyroid carcinoma and follicular thyroid carcinoma cells, cells in ATC do not normally retain biological features or functions of follicular thyroid cells [9,10]. ATC is characterized by aggressive local progression as well as high rates of distant metastasis, which lead to rapidly fatal clinical outcomes [1,2,9,10]. The treatment option of the patients with ATC includes surgery, radiotherapy, and/or chemotherapy (multimodal therapy), targeted therapy, and immunotherapy [1]. Among them, molecular targeted therapies are the promising emerging treatment modalities [7]. Hence, the management of ATC is always difficult; therefore, exploring new therapeutic modalities may provide favourable prediction as well as increasing the overall survival rate of patients [11]. Recent studies also revealed more information about genetic profiles on ATC and these genomic data are useful for the future advances of targeted therapy [12,13,14].
MicroRNAs (miRNA) regulate gene expression and therefore protein production by binding to their target mRNAs. miRNAs play crucial role in the regulation of many cellular processes such as growth, development, metabolism, proliferation and apoptosis etc. [15]. Many miRNAs are abnormally expressed in thyroid carcinomas. These dysregulations in miRNA expression may contribute to the pathophysiology of tumourigenesis and disease progression in ATC [15]. Similarly, long non-coding RNAs (lncRNAs) also play critical role in ATC by regulating transcription and epigenetic or post-transcriptional regulation of gene expression [16]. They have been found to be implicated in epithelial to mesenchymal transition (EMT) in ATC, which enhances the metastatic potential of ATC. For example, suppressing the expression of lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in ATC cells halted cell proliferation, migration and invasion. In addition, inhibition of MALAT1 caused increased apoptosis and autophagy formation [16].
In this review, we describe how non-coding RNAs (ncRNAs) function in epigenetic regulation of normal cells and ATC cells. Furthermore, the roles of specific miRNAs, lncRNA, and siRNA molecules in ATC as well as the potential use of ncRNAs in designing targeted therapy against ATC are also illustrated.

2. Non-Coding RNAs as the Important Players in the Regulation of Gene Expression

MicroRNAs (miRNAs/miRs), one of the classes of small non-coding RNAs comprising 19–25 nucleotides, mediate post-transcriptional gene silencing by partial complementarity with the 3′untranslated region (3ʹ UTR) of the target mRNAs (Figure 1) [17]. miRNAs are one of the most vigorously studied subclasses of non-coding RNAs as they play a regulatory role in epigenetic control of cells [18,19]. miRNA mediated gene silencing occurs through endonucleolytic cleavage or by translational repression [20,21]. Small interfering RNAs (siRNAs), another member of non-coding RNA family, have some similarities with miRNAs as far as the length and the pathways of their biogenesis are concerned. miRNAs are excised from stem-loop structures, whereas siRNAs are derived from long entirely complementary double-stranded RNA precursors primarily of exogenous origins, such as retrotransposons and viral sequences [21,22]. Both siRNAs and miRNAs are associated with translational repression and mRNA cleavage, which mainly depends on their homology to their mRNA targets (Figure 1).
Long non-coding RNAs (lncRNAs) are another type of non-coding RNAs that control transcription, translation, and mRNA stability in the processes of cell differentiation and development [23,24,25]. The tissue specific expression of lncRNAs is lower than the protein coding genes. They are generated via pathways similar to that of protein coding genes, with similar histone modification and splicing pattern. However, lncRNAs are typically without having an open reading frame (ORF) and found in the chromatin inside the nucleus [26]. PIWI-interacting RNAs (piRNAs), like other non-coding RNAs, have also been found to contribute in epigenetic programming [27,28]. A miRNA-like post-transcriptional gene silencing is mediated by piRNA through transposon sequences overlapping with the 3′ or 5′UTRs of mRNAs [29,30]. Though piRNAs show their expressions in somatic tissue, they are mainly transcribed in germline cells. In somatic tissue, the majority of piRNAs are transcribed in the known transcript, whereas they are encoded as piRNA clusters in germline cells [31]. In contrast to lncRNAs and miRNAs, piRNAs direct transposon cleavage, in order to protect the genome against transposon-induced insertional mutagenesis to maintain genome integrity [32]. In this review, we will focus only on miRNAs, lncRNAs, and siRNAs as they are mainly involved in pathogenesis in ATC.

3. Roles of miRNAs in Anaplastic Thyroid Carcinoma

MiRNA-mediated gene silencing in tumourigenesis has been a prominent topic over the last few decades due to a variety of reasons. First, various cellular processes including cell differentiation, division, proliferation, and apoptosis are controlled by miRNA expressions. Second, most of the miRNAs are in or near the fragile tumour-related genomic regions, and at the amplification or broken point region. The locations of these genes at these fragile regions could attribute to their aberrant expression by inducing genomic instability in various cancers [33]. Third, abnormal expressions of miRNAs have been noted in a variety of tumour tissues and cancer cells. Finally, alterations in the sequence of the miRNA precursors affect the processing of miRNA maturing, which in turn results in cancer susceptibility [34]. This information suggest that miRNAs are closely related to tumourigenesis in human cancers. Therefore, selective manipulation of dysregulated miRNAs expression could inhibit the formation of tumour [35]. In ATC, miRNAs are associated with the development, metastasis, and prognosis of patients with the cancer. Thus, altering the expression of these miRNA could be a potential therapeutic option against patients with ATC [36,37,38].
Several miRNAs have been found to be involved in identifying clinical aggressiveness of ATC and prognosis of patients with ATC (Table 1) (Figure 2). For example, one of the very commonly dysregulated miRNAs in ATC is miRNA-205-5p, which plays critical roles in angiogenesis and EMT [37]. Notably, miRNA-205 (miR-205) contributes to tumour vascularization and tumour invasion through regulating the expression of VEGF-A and ZEB1 genes in melanoma, glioblastoma, ovarian carcinoma, and breast carcinoma. Two ATC cell lines (MB-1, BHT-101) were transfected with the miR-205 expression vector and significant inhibition of angiogenesis and EMT was detected through suppression of VEGF-A, ZEB1. In addition, miR-205 overexpression impeded the migration, invasion, and tube formation of endothelial cells. Furthermore, in vivo miR-205 expression resulted in inhibition of tumour growth, vascularization and invasion [37]. Thus, miR-205 expression could be used as potential option in order to halt or inhibit both angiogenesis and EMT in ATC.
miR-19a is involved in the progression of ATC [38]. A significant reduction of cell proliferation was noticed in miR-19a downregulated ATC cells (8505c). Robust reduction in cell growth and viability after miR-19a inhibition was reported due to the induction of apoptosis by activation of caspases (caspase 9). PTEN, a tumour suppressor and is one of the known targets of miR-19a. Exogenous inhibition of miR-19a could activate PTEN in ATC cells which subsequently leads to reduced proliferation and decreased cell growth. Moreover, PTEN inactivation could correlate with the progression and aggressiveness of ATC [39]. Thus, the activation of PTEN by modulating miR-19a could provide better management of patients with ATC.
A study determining the role of miR-17-92 cluster suggested that miR-17-3p and miR-17-5p, two of the RNAs in miR-17-92 cluster, were overexpressed in ATCs when compared to adjacent non-cancerous tissues [40]. To determine the role of miR-17-3p and miR-17-5p, ATC cells (ARO and FRO) were transfected with antisense oligonucleotides containing locked nucleic acids, inhibitors of these miRNAs. Inhibition of miR-17-3p expression resulted in complete growth arrest followed by apoptosis of ATC cells. Whereas, miR-17-5p inhibitor induced substantial growth reduction and led to cellular senescence in ATC cells. However, inhibition of miR-18a, another member of the cluster, moderately attenuated the cell growth which indicated functional differences among the members of the cluster in ATC cells [40]. Inhibition of miR-17-3p expression resulted in increased apoptosis by caspase activation. However, transfection with other inhibitors of miR-17-3p cluster did not cause caspase activation. Thus, miR-17-3p could have a distinct function from other members of the cluster [40]. Thus, further studies are required to identify target mRNA for miR-17-3p in ATC.
X-linked inhibitor of apoptosis protein (XIAP) has been found to contribute to cancer growth and progression [41]. Inhibition of XIAP induced apoptosis in ATC. A study comparing miRNA mediated regulation of XIAP in non-neoplastic thyroid cells (Nthy-ori 3-1) and ATC cells (8305C) found that miR-618 targets XIAP expression, which results in inhibition of tumour growth [41]. In addition, ectopic miR-618 expression was associated with the prevention of invasion and migration of ATC (8305C) cells by silencing XIAP expression [41]. Therefore, the over-expression of miR-618 could inhibit ATC cells by targeting XIAP gene.
miR-20a is another example of miRNA inhibiting the growth of ATC cells by regulating the LIMK1 expression [42]. LIMK1 plays critical role in tumour cell invasion and metastasis in various cancers [43,44,45,46]. LIMK1 overexpression could increase the invasiveness of breast and prostate cancer cells both in vitro and in vivo, and knockdown of LIMK1 could diminish invasion of breast carcinoma and prostate carcinoma cells [46,47]. Rho signalling regulates the activity of LIMK1, which modulates actin dynamics by controlling the activity of the cofilin family proteins [48,49]. miR-20a expression was significantly higher in ATC tissues when compared with differentiated thyroid carcinoma, benign thyroid lesions, and non-cancerous thyroid tissues. In addition, the overexpression of miR-20a in ATC cells (C643) caused reduced proliferation and tumour growth both in vitro and in vivo by inducing silencing of LIMK1 expression [42]. Also, knockdown of LIMK1 resulted in inhibition of cellular invasion and migration of ATC cells [42].
Contrary to miR-20a, miR-155 could stimulate the growth of ATC cells by regulating the expression of suppressor of cytokine signalling 1 (SOCS1) [50]. miR-155 inhibits apoptosis as well as promotes proliferation, invasion and migration of ATC (8305c cells and FRO cells) [50]. SOCS1 is a member of the suppressor of cytokine signalling (SOCS) family, which negatively regulates cytokine signal transduction [51]. Aberrant expressions of miR-155 and SOCS1 showed inverse correlations in ATC. It was found that miR-155 lowers SOCS1 levels by binding to SOCS1 3′-UTR, therefore promoting its degradation [50]. In addition, miR-155 expression was associated with cervical metastasis and extra-thyroidal invasion in ATC. miR-155 inhibition caused a significant increase in the expression level of SOCS1. Thus, the expression of SOCS1 reversed the effects of miR-155 suppression on cell proliferation, apoptosis as well as invasion. Furthermore, miR-155 in association with downregulation of SOCS1 has been implicated in the progression of lung carcinoma and anaplastic large cell lymphoma [52,53]. Therefore, miR-155 could be a potential target for therapeutics against patients with various cancers, including ATC.
miR-125b is downregulated and identified as a tumour suppressor in various cancers including thyroid cancer [54]. miR-125b expression was downregulated in ATC tissues and cells when compared to that of adjacent non-neoplastic thyroid tissues and non-neoplastic thyroid follicular cells (Nthy-ori 3-1), respectively. There was an inverse correlation between phosphoinositide 3-kinase catalytic subunit delta (PIK3CD) and miR-125b expression noted in ATC tissues and cells (SW1736 and 8305C) [54]. PIK3CD contributes in the PI3K signalling pathway, which plays an important role in the development of various cancers [55,56]. A negative relationship between miR-125b and PIK3CD was noted, as miR-125b targets and inhibits the expression level of PIK3CD in ATC tissues and cells. Ectopic miR-125b expression inhibited the migration and invasion of ATC cells whereas PIK3CD overexpression reversed this effect. Furthermore, exogenous miR-125b expression in ATC cells (SW1736 and 8305C) suppressed PI3K, phospho-Akt, and phospho-mTOR expression [54]. Thus, suppression of cell migration and invasion by miR-125b expression through inhibiting PIK3CD suggested a potential therapeutic option for the treatment of patients with ATC.
Expression of miR-483 was increased, while the expression of partitioning-defective 3 (Pard3) was decreased in ATC cells and tissues [57]. A member of Pard family namely Pard 3, controls cellular polarity, migration, and cell division [58]. The study also investigated the role of miR-483 and Pard3 on the TGF-β 1-mediated progression of ATC. TGF-β1 was found to regulate miRNA driven migration and tumourigenesis in various cancers [59,60]. In ATC, activated TGF-β1 recruits Pard3 in order to control cell polarity and promote cell migration [57]. Moreover, inhibition of miR-483 expression increased Pard3 expression, thereby promoting TGF-β1-induced cell migration and invasion. It was shown that after binding to Pard 3, miR-483 targets and binds to Pard 3 to inhibit its expression in ATC cells which as a result induces TGF-β1-mediated cell proliferation, migration, and invasion, both in vitro and in vivo [57]. Clinically, poor disease-free survival rates were observed upon downregulation of miR-483 and upregulation of Pard3 in patients with ATC [57]. Furthermore, it was proven that overexpression of Pard3 inhibits TGF-β1 mediated EMT and Tiam1-Rac signalling in ATC cells [57].
In comparison to non-neoplastic thyroid tissues and differentiated thyroid carcinomas, the expression of miR-25 and miR-30d was significantly downregulated in ATC tissues [61]. Importantly, ectopic expression of miR-25 and miR-30d inhibited proliferation and colony formation of ATC cells by arresting cells at G2/M phase. Mechanistically, miR-25 and miR-30d reduced cancer cells growth and proliferation by targeting the expression of oncogene, polycomb protein enhancer of zeste 2 (EZH2) [62]. EZH2 is one of the oncogenes upregulated in ATCs when compared to differentiated thyroid carcinomas. This inverse relationship between miRNAs and oncogene EZH2 indicates an active role of these miRNAs in the pathogenesis of ATC.
MiR-34b was downregulated in cells derived from metastatic ATC (BHT-101) compared to the cells derived from primary ATC (8505C) [63]. Exogenous overexpression of miR-34b in ATC cells showed decreased cell proliferation, decreased wound healing, reduced cell cycle progression and increased apoptosis [63]. Besides, liposome-mediated delivery of miR-34 resulted in downregulation of vascular endothelial growth factor-A (VEGF-A) in ATC cells. VEGF-A is a principle factor for angiogenesis in cancer [63]. In vivo experiments suggested that intravenous administration of liposome-loaded miR-34b reduced the size of tumour formation when compared to control [63]. A promising tumour-suppressing role of miR-34b via VEGF-A regulation in ATC cells was demonstrated using both in vitro and in vivo model by the study [63]. Thus, miR-34b could be a useful therapeutic strategy, in addition to the VEGF-A, for design of targeting therapy in patients with ATC.
The epidermal growth factor receptor (EGFR) is overexpressed in ATC cells, which dictates its crucial role in tumourigenesis of ATC [64]. Epidermal growth factor (EGF) starts a signalling cascade after binding to EGFR, and this signalling may enhance migration and invasiveness of ATC cells. Importantly, there is an interaction identified among miR-200 family members, EGF/EGFR and EMT process in ATC cells. Induction of EGF in ATC cells leads to the loss of miR-200 expression and increases Rho/ROCK activity, resulting in increased EMT and subsequent cancer invasion [64]. Increased vimentin expression was noted in non-neoplastic human thyroid cells (Nthy-ori 3-1) followed by EGF treatment. On the other hand, EGFR silencing results in vimentin downregulation in ATC cells (SW1736), which indicates that EGF induces EMT in non-neoplastic thyroid cells, while EGFR silencing in ATC cells alters EMT. Moreover, EGF treatment downregulates miR-200s in non-neoplastic thyroid cells, whereas EGFR silencing increases miR-200s expression in ATC cells, indicating miR-200 family and EGF/EGFR mediated regulation of EMT [64]. Furthermore, Rho/ROCK signalling is crucial in TGFβ-induced EMT as inhibition of the Rho effector (ROCK) results in inhibition of TGFβ-induced EMT [65,66]. The study found an increase of Rho/ROCK activity in EGF-treated non-neoplastic thyroid cells and the reverse was noted in EGFR-silenced ATC cells [64]. Therefore, a model was proposed for EMT regulation by EGF/EGFR, miR-200s, Rho/ROCK and EMT markers suggesting that miR-200 upregulation could serve as a potential therapeutic option for ATC. Other microRNAs like miR-146a/b was found to be upregulated in ATC, whereas microRNA-30 family and let-7 family were downregulated [67,68,69]. Alterations of these microRNA expressions caused a significant change in cell proliferation, cell growth, and cell differentiation in ATC cells. It was found that aforementioned miRNAs modulate their actions via affecting various molecules, such as PTEN (microRNA-146a/b), p53 (microRNA-146a/b), Beclin1 (autophagy-promoting protein; microRNA-30 family) and RAS (let-7 family) [67,68,69]. On the other hand, upregulated microRNA-195 inhibited progression of ATC by affecting the expression of VEGF and p53 [70]. Moreover, another upregulated miRNA miR-126 also reported to be reduced tumour growth by suppressing angiogenesis via reduction of VEGF expression and increasing apoptosis in ATC [71]. Hence, studies of the roles of microRNA in anaplastic thyroid carcinoma could provide potential useful information for target therapeutic approaches on anaplastic thyroid carcinoma.

4. Roles of lncRNAs in Anaplastic Thyroid Carcinoma

Abnormal expressions of lncRNAs have been implicated in the tumuorigenesis, progression, and chemoresistance of different human cancers, including ATCs (Table 2) (Figure 2) [72,73]. For example, H19 gene encodes 2.3 kb long lncRNA, higher expression of this lncRNA stimulates initiation and progression and therapy resistance of ATC [73,74]. Another study reported H19 lncRNA mediated cancer initiation, progression and metastasis [75], indicating that H19 lncRNA could be used as a potential tumour marker for ATC. Moreover, inhibition of H19 expression by siRNA resulted in significantly decreased proliferation, invasion, as well as increased apoptosis of ATC cancer cells (8505C) [74]. Distant lung metastasis represents one of the most lethal threats of thyroid carcinomas and H19 has been proven to be engaged with lung metastasis in other tumour types [75]. An experimental metastasis model using a murine model of ATC unveils the functions of H19 lncRNA mediated tumour growth and metastasis [74]. This model used bioluminescence imaging, which has been a well-established method to investigate in vivo metastasis and tumour growth [76,77]. The study suggested that their bioluminescence measurements was consistent with tumour volumes during necropsy, despite of the fact that tumour bioluminescence could be affected by targeted agent mediated tumour necrosis and alterations in tumour vascularity. Targeting H19 lncRNA using shRNA reduces H19 expression level in vivo, which subsequently resulted in a strong reduction of tumour burden and inhibition of metastases in an orthotopic ATC mouse model [74]. Furthermore, targeting H19 inhibited tumour metastases by approximately eight-fold in comparison to that of control [74]. Thus, H19 might be a potential target for molecular therapy for the patients with ATC.
HLA complex P5 (HCP5) another lncRNA has been implicated in the pathogenesis of follicular thyroid carcinoma and anaplastic thyroid carcinoma by regulating the expression of tumour suppressor miR-128-3p [78]. HCP5 expression was significantly increased in ATC cells and tissue samples when compared to that of non-neoplastic human thyroid follicular cells (Nthy-ori 3-1) and adjacent non-neoplastic thyroid tissues, respectively [78]. Moreover, knockdown of HCP5 caused reduced cell viability, while elevated apoptotic rate and caspase -3/7 activity in ATC cells (ARO and SW1736). It was demonstrated that miR-128-3p was the target gene of HCP5. Thus, the higher expression of HCP5 lncRNA in ATC cells and tissues caused a significant reduction of miR-128-3p in ATC cells and tissues. Overexpression of miR-128-3p by suppressing HCP5 resulted in significant reduction of ATC cell viability and induction of apoptosis [78]. However, these effects of miR-128-3p on ATC cell viability and apoptosis were reversed when the inhibitor of miR-128-3p was applied [78]. This result could lead to a potential approach via regulating miR-128-3p for the treatment of patients with ATC.
The underlying molecular mechanism of lncRNA Urothelial carcinoma-associated 1 (UCA1) mediated progression of ATC has been described [79]. The expression levels of UCA1 were significantly upregulated in ATC cells (SW1736 and KAT-18) and tissues in comparison to that of non-neoplastic human thyroid cells (Nthy-ori3-1) and adjacent non-neoplastic tissues, respectively [79]. Suppression of UCA1 induced a significant reduction of ATC cells viability, proliferation, migration and invasion in vitro and inhibited tumour growth in vivo by repressing the expression of c-myc proto-oncogene [79]. miR-135a could directly bind to the 3’UTR of c-myc and UCA1 may bind to miR-135a. Thus, UCA1 represses the binding of c-myc with miR-135a, which inhibits miR-135a expression and increases the expression of c-myc. c-myc oncogene is overexpressed in many human cancers, including ATC and plays crucial role in the regulation of several cellular processes, including protein synthesis, cell growth and proliferation in various cell types [80,81]. Therefore, due to its oncogenic role in promoting ATC cell proliferation, UCA1 could be used as a potential target for treatment of ATC.
Nuclear paraspeckle assembly transcript 1 (NEAT1) is a lncRNA with a length of 4-kb and localized to the nucleus [82]. NEAT1 acts as an oncogene and involved in the pathogenesis of various human cancers [83,84,85,86]. It plays critical roles in cancer chemoresistance several cancer types such as in osteosarcoma, ovarian carcinoma, leukaemia, and ATC [82,87,88,89]. In ATC, NEAT1 was upregulated in tissues and cells (SW1736 and 8505C) when compared to that of non-neoplastic human thyroid tissues and non-neoplastic thyroid cells (Nthy-ori 3-1), respectively. Silencing of NEAT1 resulted in decreased resistance to the chemotherapeutic reagent, cisplatin of ATC cells (SW1736 and 8505C). In addition, NEAT1 binds to miR-9-5p to suppress its expression, while it was also suggested that miR-9-5p directly target sperm-associated antigen 9 (SPAG9). Thus, NEAT1 silencing lead the overexpression of miR-9-5p, which in turn sensitized ATC cells to cisplatin by the suppression of SPAG9 expression [82]. Therefore, NEAT1 should be considered as a potential therapeutic option as silencing of NEAT1 decreased cisplatin -resistance by reducing miR-9-5p sponging and regulating SPAG9 expression.
To add to the roles of lncRNAs in ATC, another lncRNA Prader Willi/AngelmanRegion RNA5 (PAR5), which is downregulated in ATC, acts as a tumour suppressor by repressing Enhancer of Zeste Homolog 2 (EZH2) activity [90]. PAR5 overexpression in ATC cells (FRO and 8505c) resulted in reduced cell proliferation and cell migration ability, which indicated the anti-oncogenic role played by this lncRNA and its downregulation could lead to development of ATC. PAR5 interacts with EZH2 and reduces its protein level in thyroid cancer cells [90]. Moreover, PAR5 escapes E-cadherin from the negative regulation of EZH2 by binding to EZH2 promoter and inhibiting its expression [90]. Recent studies suggested EZH2 mediated inhibition of ATC cell differentiation, which paved the way to investigate the molecular mechanism regarding the effects of PAR5 on EZH2 expression [91]. EZH2 was over expressed in ATCs and it inhibited thyroid cell differentiation [91]. Moreover, it was demonstrated that PAR5 directly binds EZH2 and restoration of PAR5 reduced EZH2 protein and H3K27me3 (DNA packaging protein Histone 3) levels in the absence of changes in EZH2 mRNA level, which suggested a consistency with some previous reports about PAR5 mediated effect on EZH2 stability [92,93]. Consistently, the study further found a negative correlation between EZH2 protein and PAR5 RNA levels, strongly supporting the effects of PAR5 on EZH2 [91]. As lack of E-cadherin expression is a hallmark of EMT [94,95,96,97], EZH2 could repress E-cadherin expression through H3K27 tri-methylation at its promoter [98]. ATC-PAR5 overexpressing cells were seen to be high in E-cadherin protein levels, as PAR5 restoration inhibited the binding of EZH2 to E-cadherin promoter, which subsequently reduces EZH2 mediated inhibitory activity on E-cadherin expression [90]. Therefore, it was cleared that PAR5 exhibits its anti-tumour activity by impairing the oncogenic roles of EZH2.
LncRNA PVT1 modulates thyroid cancer cells’ proliferation by recruiting EZH2 and regulating thyroid-stimulating hormone receptor [99]. The gene that encodes lncRNA PVT1 resides in the well-known cancer-risk region of chromosome 8q24 [100]. Accordingly, lncRNA PVT1 plays pivotal roles in a variety of human cancers, such as ovarian carcinoma, pancreatic carcinoma, and non-small cell lung carcinoma [101,102]. Moreover, LncRNA PVT1 was significantly upregulated in ATC tissues, as well as in ATC cells (8505C) when compared to that of non-neoplastic controls [86]. However, silencing of PVT1 resulted in a significant reduction of proliferation of ATC cells (8505C) followed by arresting cell cycle at G0/G1 stage. Cell cycle kinetics in ATC cells was changed due to PVT1 mediated decrease in cyclin D1 and thyroid-stimulating hormone receptor (TSHR) expressions [99]. Moreover, silencing of PVT1 halted recruitment of EZH2, which as a result ceased its enrichment. Furthermore, analysing the interaction between PVT1 and EZH2 by RNA immunoprecipitation assay in ATC cells indicated that PVT1 directly interacts with EZH2 to alter its expression in cells. Furthermore, the relative mRNA and protein levels of thyroid-stimulating hormone receptor in ATC cells (8505C) were significantly decreased after silencing of PVT1 when compared to the controls [99]. Interaction of thyroid-stimulating hormone with its receptor plays a crucial role in the development and cell proliferation [103]. Thus, lncRNA PVT1 may contribute to tumourigenesis of ATC through recruiting EZH2 and regulating thyroid-stimulating hormone receptor expression.
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a commonly overexpressed lncRNAs in human carcinomas [104,105]. MALAT1 is associated with tumourigenesis, apoptosis, and cell cycle progression in various carcinomas, including ATC [106,107,108]. It acts as miRNA sponge by inhibiting the functions of tumour suppressor miR-363-3p, which in turn leads to Mcl1 expression, a target of miR-363-3p and allows Mcl1 mediated oncogenic role in ATC. Also, enhanced MALAT 1 expression is closely associated with MEK/ERK signalling pathway [109,110,111]. MEK/ERK signalling reported to regulate cell cycle and apoptosis [109,110,111]. A study used MEK and Aurora kinase family inhibitor BI-847325 to determine its role in the expression of genes involved in cell cycle and apoptosis in ATC [108]. BI-847325 treatment significantly changed the expressions of MALAT1 and its downstream target genes such as miR-363-3p, Mcl1, and cyclin D1 in ATC cells (C643 and SW1736). miR-363-3p expression was significantly upregulated in both cells, whereas Mcl1 and MALAT1 expression were significantly down-regulated in C643 cells upon BI-847325 treatment [108]. Though Mcl1 is a target gene of MEK/ERK signalling pathway, however, the expression of Mcl1 was not significantly downregulated after BI-847325 treatment in the ATC (SW1736) cells [108]. Moreover, cyclin D1 is another oncogene affected by BI-847325 treatment in ATC cells as its expression was decreased followed by BI-847325 treatment [108]. Cyclin D1 is a prominent regulator of cell cycle and causes cell cycle arrest in G1 phase [112]. Thus, lncRNA MALAT1 mediated regulation of cell cycle and apoptosis related genes in ATC unveil potential target, which could be used as an effective option against ATC.
Another lncRNA known as papillary thyroid carcinoma susceptibility candidate 3 (PTCSC3) associated with ATC progression and doxorubicin resistance by targeting signal transducer and activator of transcription 3 (STAT3) protein [113,114]. STAT3 promotes tumourigenesis by enhancing inflammation, proliferation, invasion, and immunosuppression in cancer [113]. PTCSC3 was first identified in thyroid and plays tumour suppressive role in thyroid cancer [115,116]. STAT3 stimulates the expression of chromatin remodelling complex INO80, a member of INO80 subfamily, plays important functions in DNA replication, repair and transcriptional regulation [117]. PTCSC3 impedes the expression of INO80 through negatively regulating STAT3, which subsequently decreases drug resistance of ATC cells to chemotherapy drug doxorubicin [114]. In addition, PTCSC3 inhibits stem cell properties of ATC (8050C) cells, as the expression rate of CD133 and MDR-1 (cancer stem cell markers) was decreased [114]. Also, the expression of CD133 associated with poor disease outcome of patients with the cancer [114]. Furthermore, CD133 expressing ATC cells were involved in tumour progression, invasion and therapy resistance [118]. Thus, the lncRNA PTCSC3 regulates the pathogenesis of ATC by modulating STAT3 signalling and CSCs phenotype of cancer cells.
Additionally, non-coding pseudogenes such as high mobility group AT-hook 1pseudogene 6 and 7 (HMGA1P6 and HMGA1P7) could regulate nuclear proteins such as high Mobility Group A (HMGA), which in turn regulate chromatin structure, replication and gene transcription, making them an important player in cellular processes [119,120]. Altered expression of HMGA has been associated with many pathophysiological diseases, including human carcinomas [119,120]. For example, higher HMGA1 protein levels are observed in ATC when compared to normal thyroid tissues [121]. Most importantly, HMGA1P6 and HMGA1P7, two HMGA1 non-coding pseudogenes, are abnormally expressed in ATC [121]. These non-coding pseudogenes bind with miRNAs such as miR-16 and miR-17, thereby release their inhibitory effects on target proteins such as HMGA1, which are involved in cancer progression [121]. Moreover, overexpression of HMGA1P6 or HMGA1P7 reduced the effects exerted by miRNA on the levels of both the HMGA1 transcript and protein dose-dependently. Also, knockdown of HMGA1P6 and HMGA1P7 resulted in decreased HMGA1 mRNA and protein levels, indicating that HMGA1Ps may compete for the endogenous miRNA-binding sites [121]. Furthermore, overexpression of HMGA1Ps lead to increase of HMGA2, and other cancer related proteins such as VEGF and EZH2 [121]. Thus, HMGA1P6 and HMGA1P7 are important proto-oncogenic competitive endogenous RNAs and could be used as a potential tool in designing anti-cancer therapy.

5. Non-coding RNA (siRNA) Mediated Targeted Therapy for Anaplastic Thyroid Carcinoma

Small interfering RNA (siRNA) or short interfering RNA, is a class of double-stranded non-coding RNA molecules operates within the RNA interference (RNAi) pathway (Figure 1) [122,123]. Exogenous siRNA mediates transcriptional gene silencing (TGS) in cells through DNA methylation and histone modification [124,125,126]. siRNA could silence EZH2 and then reverses the therapeutic drug (cisplatin)-resistance of non-small cell lung carcinoma and gastric carcinoma cells [127]. In ATC, gene silencing with siRNA has been implemented in cells proliferation, invasion, migration, metastasis, and apoptosis [99,101]. For example, siRNA mediated suppression of MADD (MAPK-activating death domain activating protein) expression inhibits growth and spread of ATC cells (8505C, C643 and HTH7) [128]. MADD could play a survival-promoting role by suppressing TNFα mediated apoptosis. MADD also activates MAPKs through Grb2 and Sos1/2 recruitment, which subsequently results in activation of ERK pathway without affecting the function of p38, Jun, and NFκB [129]. Accordingly, treatment with MADD siRNA inhibited in vitro proliferation of ATC cells (8505C, C643 and HTH7). Furthermore, it decreased in vivo tumour growth of ATC cell (8505C)-derived-orthotopic tumour model when compared to untreated control and scramble siRNA treated groups [128]. Furthermore, a significant reduction in cellular migration and invasion potential, as well as clonogenic capacity, were seen upon MADD inhibition. In addition, inhibition of (EMT) and Wnt signalling was observed after MADD inhibition [128]. Moreover, knockdown of MADD could exhibit its anti-migratory/invasive effects by blocking TNFα/ERK/GSK3β axis as MADD siRNA inhibited TNFα induced activation of pERK, pGSK3β and β-catenin [128]. Likewise, in vivo MADD siRNA treatment also decreased lung metastases along with tumour regression [128]. In addition, a reduction in proliferative index (Ki-67) and N-cadherin expression, and an increase in E-cadherin expression were observed in MADD siRNA-treated xenotransplanted tumour tissues [128]. Thus, MADD siRNA resulted in a broad range of effects in ATC cells such as inhibiting β-catenin nuclear translocation and consequently, the expression of its target genes.
siRNA mediated silencing of target genes associated with the induction of apoptosis of ATC cells [130]. For example, Nijmegen breakage syndrome 1 (NBS1) plays an important role in the repair of radiation-induced DNA double-strand breaks (DSBs) and suppression of NBS1 could induce apoptosis of ATC cells (8305C cells) [130]. The frequency of apoptosis through the caspase pathway and heat sensitivity was increased by the transfection of NBS1-siRNA in these ATC cells. Furthermore, NBS1-siRNA transfected cells were more frequent with histone H2AX phosphorylated at serine 139 (H2AX) foci than control cells transfected with scrambled siRNA [130]. H2AX foci are a kind of indicator for the presence of DNA double-strand breaks [131]. Specifically, one H2AX focus correlates with one DNA double-strand break [132]. DSBs have been found to be associated with heat shock-induced cell death [131]. Thus, NBS1-siRNA mediated heat sensitization could result from the suppression of heat-induced DNA double-strand breaks repair, which suggestedNBS1-siRNA could potentially function as a pro-apoptotic strategy for patients with ATC.
Inhibition of zinc finger transcription factor Slug by siRNA may induce apoptosis and sensitizes ATC cells (SW1736) to chemotherapeutic drug, doxorubicin [133]. Slug is a member of the Snail family of zinc-finger transcription factors, which could protect cells against apoptosis induced by radiation [134,135]. It mediates its action via regulation of Bcl-2 and Bax expression and transactivation of PUMA (p53 upregulated modulator of apoptosis) [136]. In addition, strong nuclear immunostaining of Slug was noted in ATC tissues and higher Slug mRNA expression was found in ATC cells (THJ-16T and THJ-21T) when compared to that of non-neoplastic cells [137]. Importantly, siRNA mediated inhibition of Slug inhibits the growth of ATC cells (SW1736) and sensitizes these cells to doxorubicin both in vitro and in vivo [133]. Moreover, siRNA mediated inhibition of Slug combined with doxorubicin resulted in reduced cell viability in ATC cells compared to the treatment with doxorubicin alone [133]. In addition, it was noted that silencing of Slug sensitizes ATC cells to doxorubicin via a PUMA dependent pathway. As inhibiting PUMA caused chemoresistance of ATC (SW1736)/Slug siRNA cells to doxorubicin [133]. On the other hand, the suppression of Slug in combination with doxorubicin significantly inhibited tumour growth and induced apoptosis in mice xenograft model. siRNA mediated suppression of PUMA reduced the threshold of apoptosis induction by DOX confirm the involvement of PUMA in apoptosis in ATC cells [133]. Therefore, knockdown of Slug has the potential for the development of new therapeutic strategies in order to improve chemotherapy for patients with ATC.

6. Conclusions and Future Perspectives

The patterns of expression of non-coding RNAs in ATC could be a useful tool for predicting prognosis of the patients with the cancer. Moreover, since non-coding RNAs are involved in the acquisition and enhancement of malignant properties by cancer cells, they could be potential targets for RNA-based therapy. Also, overexpression or inhibition of non-coding RNAs can lead to the reduction of the chemoresistance in ATC cells against some known anticancer agents. However, the difficulties in the analysis of this non-coding RNAs mediated regulatory network limit the investigation of their potential clinical applications. Thus, the improvements in gene and genome scanning technologies are needed to maximize the functions of non-coding RNAs in ATC. The utmost goal is to unveil their interactions in normal and cancer cells and finally to use this knowledge in designing anticancer therapies.

Author Contributions

P.K.D. and S.Y.A. analysed the literature, synthesized the information and drafted the manuscript. I.A. edited the manuscript. F.I. designed the project and edited the manuscript. A.K.L. supervised the project and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding and the APC was funded by School of Medicine, Griffith University, Gold coast campus, Queensland, Australia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Deeken-Draisey, A.; Yang, G.Y.; Gao, J.; Alexiev, B.A. Anaplastic thyroid carcinoma: An epidemiologic, histologic, immunohistochemical, and molecular single-institution study. Hum. Pathol. 2018, 82, 140–148. [Google Scholar] [CrossRef] [PubMed]
  2. Lam, A.K. Update on Adrenal Tumours in 2017 World Health Organization (WHO) of Endocrine Tumours. Endocr. Pathol. 2017, 28, 213–227. [Google Scholar] [CrossRef] [PubMed]
  3. Rashid, M.; Agarwal, A.; Pradhan, R.; George, N.; Kumari, N.; Sabaretnam, M.; Chand, G.; Mishra, A.; Agarwal, G.; Mishra, S.K. Genetic Alterations in Anaplastic Thyroid Carcinoma. Indian J. Endocrinol. Metab. 2019, 23, 480–485. [Google Scholar] [PubMed]
  4. Tiedje, V.; Stuschke, M.; Weber, F.; Dralle, H.; Moss, L.; Führer, D. Anaplastic thyroid carcinoma: Review of treatment protocols. Endocr. Relat. Cancer 2018, 25, R153–R161. [Google Scholar] [CrossRef] [Green Version]
  5. García Carretero, R.; Peña-Arce, C.; Martinez-Quesada, G.; Garcia-Alvarez, J. Fatal outcome of an anaplastic thyroid tumour. BMJ. Case. Rep. 2017, 2017, bcr2017221672. [Google Scholar] [CrossRef] [PubMed]
  6. Bozorg-Ghalati, F.; Hedayati, M. Molecular biomarkers of anaplastic thyroid carcinoma. Curr. Mol. Med. 2017, 17, 181–188. [Google Scholar] [CrossRef] [PubMed]
  7. Abe, I.; Lam, A.K. Anaplastic thyroid carcinoma: Current issues in genomics and therapeutics. Curr. Oncol. Rep. 2020, in press. [Google Scholar]
  8. Wang, Y.; Hardin, H.; Chu, Y.H.; Esbona, K.; Zhang, R.; Lloyd, R.V. Long Non-coding RNA expression in anaplastic thyroid carcinomas. Endocr. Pathol. 2019, 30, 262–269. [Google Scholar] [CrossRef]
  9. Molinaro, E.; Romei, C.; Biagini, A.; Sabini, E.; Agate, L.; Mazzeo, S.; Materazzi, G.; Sellari-Franceschini, S.; Ribechini, A.; Torregrossa, L.; et al. Anaplastic thyroid carcinoma: From linicopathology to genetics and advancedtherapies. Nat. Rev. Endocrinol. 2017, 13, 644–660. [Google Scholar] [CrossRef]
  10. Acquaviva, G.; Visani, M.; Repaci, A.; Rhoden, K.J.; de Biase, D.; Pession, A.; Giovanni, T. Molecular pathology of thyroid tumours of follicular cells: A review of genetic alterations and their clinicopathological relevance. Histopathology 2018, 72, 6–31. [Google Scholar] [CrossRef]
  11. Saini, S.; Tulla, K.; Maker, A.V.; Burman, K.D.; Prabhakar, B.S. Therapeutic advances in anaplastic thyroid cancer: A current perspective. Mol. Cancer 2018, 17, 154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Javed, Z.; Ahmed Shah, F.; Rajabi, S.; Raza, Q.; Iqbal, Z.; Ullah, M.; Ahmad, T.; Salehi, B.; Sharifi-Rad, M.; Pezzani, R.; et al. LncRNAs as Potential Therapeutic Targets in Thyroid Cancer. Asian. Pac. J. Cancer Prev. 2020, 21, 281–287. [Google Scholar] [CrossRef] [PubMed]
  13. Landa, I.; Ibrahimpasic, T.; Boucai, L.; Sinha, R.; Knauf, J.A.; Shah, R.H.; Dogan, S.; Ricarte-Filho, J.C.; Krishnamoorthy, G.P.; Xu, B.; et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J. Clin. Investig. 2016, 126, 1052–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Xu, B.; Ghossein, R. Genomic landscape of poorly differentiated and anaplastic thyroid carcinoma. Endocr. Pathol. 2016, 27, 205–212. [Google Scholar] [CrossRef]
  15. Fuziwara, C.S.; Kimura, E.T. MicroRNAs in thyroid development, function and tumorigenesis. Mol. Cell. Endocrinol. 2017, 456, 44–50. [Google Scholar] [CrossRef]
  16. Gou, L.; Zou, H.; Li, B. Long noncoding RNA MALAT1 knockdown inhibits progression of anaplastic thyroid carcinoma by regulating miR-200a-3p/FOXA. Cancer Biol. Ther. 2019, 20, 1355–1365. [Google Scholar] [CrossRef] [PubMed]
  17. Das, P.K.; Siddika, M.A.; Asha, S.Y.; Aktar, S.; Rakib, M.A.; Khanam, J.A.; Pillai, S.; Islam, F. MicroRNAs, a promising target for breast cancer stem cells. Mol. Diagn. Ther. 2020, 24, 69–83. [Google Scholar] [CrossRef]
  18. Mohr, A.M.; Mott, J.L. Overview of microRNA biology. Semin. Liver. Dis. 2015, 35, 3–11. [Google Scholar] [CrossRef] [Green Version]
  19. Liu, B.; Li, J.; Cairns, M.J. Identifying miRNAs, targets and functions. Brief. Bioinform. 2014, 15, 1–19. [Google Scholar] [CrossRef]
  20. Kontomanolis, E.N.; Koukouli, A.; Liberis, G.; Stanulov, H.; Achouhan, A.; Pagkalos, A. MiRNAs: Regulators of human disease. Eur. J. Gynaecol. Oncol. 2016, 37, 759–765. [Google Scholar]
  21. Bertoli, G.; Cava, C.; Castiglioni, I. MicroRNAs: New Biomarkers for Diagnosis, Prognosis, Therapy Prediction and Therapeutic Tools for Breast Cancer. Theranostics 2015, 5, 1122–1143. [Google Scholar] [CrossRef] [PubMed]
  22. Tafrihi, M.; Hasheminasab, E. MiRNAs: Biology, Biogenesis, their Web-based Tools, and Databases. Microrna 2019, 8, 4–27. [Google Scholar] [CrossRef] [PubMed]
  23. Morris, K.V.; Mattick, J.S. The rise of regulatory RNA. Nat. Rev. Genet. 2014, 15, 423–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Fatica, A.; Bozzoni, I. Long non-coding RNAs: New players in cell differentiation and development. Nat. Rev. Genet. 2014, 15, 7–21. [Google Scholar] [CrossRef]
  25. Hu, W.; Alvarez-Dominguez, J.R.; Lodish, H.F. Regulation of mammalian cell differentiation by long noncoding RNAs. EMBO Rep. 2012, 13, 971–983. [Google Scholar] [CrossRef] [Green Version]
  26. Derrien, T.; Johnson, R.; Bussotti, G.; Tanzer, A.; Djebali, S.; Tilgner, H.; Guernec, G.; Martin, D.; Merkel, A.; Knowles, D.G.; et al. The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome. Res. 2012, 22, 1775–1789. [Google Scholar] [CrossRef] [Green Version]
  27. Huang, X.A.; Yin, H.; Sweeney, S.; Raha, D.; Snyder, M.; Lin, H. A major epigenetic programming mechanism guided by piRNAs. Dev. Cell. 2013, 24, 502–516. [Google Scholar] [CrossRef] [Green Version]
  28. Iwasaki, Y.W.; Siomi, M.C.; Siomi, H. PIWI-Interacting RNA: Its Biogenesis and Functions. Annu. Rev. Biochem. 2015, 84, 405–433. [Google Scholar] [CrossRef]
  29. Liu, Y.; Dou, M.; Song, X.; Dong, Y.; Liu, S.; Liu, H.; Tao, J.; Li, W.; Yin, X.; Xu, W. The emerging role of the piRNA/piwi complex in Cancer. Mol. Cancer 2019, 18, 123. [Google Scholar] [CrossRef] [Green Version]
  30. Lim, A.K.; Lorthongpanich, C.; Chew, T.G.; Tan, C.W.; Shue, Y.T.; Balu, S.; Gounko, N.; Kuramochi-Miyagawa, S.; Matzuk, M.M.; Chuma, S.; et al. The nuage mediates retrotransposon silencing in mouse primordial ovarian follicles. Development 2013, 140, 3819–3825. [Google Scholar] [CrossRef] [Green Version]
  31. Martinez, V.D.; Vucic, E.A.; Thu, K.L.; Hubaux, R.; Enfield, K.S.; Pikor, L.A.; Becker-Santos, D.D.; Brown, C.J.; Lam, S.; Lam, W.L. Unique somatic and malignant expression patterns implicate PIWI-interacting RNAs in cancer-type specific biology. Sci. Rep. 2015, 5, 10423. [Google Scholar] [CrossRef] [PubMed]
  32. Shi, X.; Sun, M.; Liu, H.; Yao, Y.; Song, Y. Long non-coding RNAs: A new frontier in the study of human diseases. Cancer Lett. 2013, 339, 159–166. [Google Scholar] [CrossRef]
  33. Ferdin, J.; Kunej, T.; Calin, G.A. MicroRNA: Genomic Association with Cancer Predisposition. J. Assoc. Genet. Technol. 2011, 37, 11–19. [Google Scholar] [PubMed]
  34. Di Leva, G.; Garofalo, M.; Croce, C.M. MicroRNAs in Cancer. Annu. Rev. Pathol. 2014, 9, 287–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Van Kouwenhove, M.; Kedde, M.; Agami, R. MicroRNA regulation by RNA-binding proteins and its implications for Cancer. Nat. Rev. Cancer 2011, 11, 644–656. [Google Scholar] [CrossRef] [PubMed]
  36. Chruścik, A.; Gopalan, V.; Lam, A.K. The clinical and biological roles oftransforming growth factor beta in colon cancer stem cells: A systematic review. Eur. J. Cell. Biol. 2018, 97, 15–22. [Google Scholar] [CrossRef]
  37. Salajegheh, A.; Vosgha, H.; Md Rahman, A.; Amin, M.; Smith, R.A.; Lam, A.K. Modulatory role of miR-205 in angiogenesis and progression of thyroid Cancer. J. Mol. Endocrinol. 2015, 55, 183–196. [Google Scholar] [CrossRef] [Green Version]
  38. Calabrese, G.; Dolcimascolo, A.; Caruso, G.; Forte, S. miR-19a is involved in progression and malignancy of anaplastic thyroid cancer cells. Onco. Targets. Ther. 2019, 12, 9571–9583. [Google Scholar] [CrossRef] [Green Version]
  39. Frisk, T.; Foukakis, T.; Dwight, T.; Lundberg, J.; Höög, A.; Wallin, G.; Eng, C.; Zedenius, J.; Larsson, C. Silencing of the PTEN tumor-suppressor gene in anaplastic thyroid Cancer. Genes. Chromosomes Cancer 2002, 35, 74–80. [Google Scholar] [CrossRef]
  40. Takakura, S.; Mitsutake, N.; Nakashima, M.; Namba, H.; Saenko, V.A.; Rogounovitch, T.I.; Nakazawa, Y.; Hayashi, T.; Ohtsuru, A.; Yamashita, S. Oncogenic role of miR-17-92 cluster in anaplastic thyroid cancer cells. Cancer Sci. 2008, 99, 1147–1154. [Google Scholar] [CrossRef] [Green Version]
  41. Cheng, Q.; Zhang, X.; Xu, X.; Lu, X. MiR-618 inhibits anaplastic thyroid cancer by repressing XIAP in one ATC cell line. Ann. Endocrinol. 2014, 75, 187–193. [Google Scholar] [CrossRef] [PubMed]
  42. Xiong, Y.; Zhang, L.; Kebebew, E. MiR-20a is upregulated in anaplastic thyroid cancer and targets LIMK1. PLoS ONE 2014, 9, e96103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kim, B.M.; Yoon, W.; Shim, J.H.; Jung, H.; Lim, J.H.; Choi, H.J.; Seo, M.; Lee, T.H.; Min, S.H. Roleof Protein Kinases and Their Inhibitors in Radiation Response of Tumor Cells. Curr. Pharm. Des. 2017, 23, 4259–4280. [Google Scholar] [CrossRef]
  44. Wei, W.; Zeng, G.; Liu, C.; Zou, B.; Li, L. Associations of LIM kinase1 (LIMK1) gene single nucleotide polymorphisms with prostate cancer susceptibility in Chinese population. Int. J. Clin. Exp. Pathol. 2017, 10, 9725–9734. [Google Scholar]
  45. Chen, J.; Ananthanarayanan, B.; Springer, K.S.; Wolf, K.J.; Sheyman, S.M.; Tran, V.D.; Kumar, S. Suppression of LIM Kinase 1 and LIM Kinase 2 Limits Glioblastoma Invasion. Cancer Res. 2020, 80, 69–78. [Google Scholar] [CrossRef] [Green Version]
  46. Davila, M.; Jhala, D.; Ghosh, D.; Grizzle, W.E.; Chakrabarti, R. Expression of LIM kinase 1 is associated with reversible G1/S phase arrest, chromosomal instability and prostate Cancer. Mol. Cancer 2007, 6, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Escuin, S.; Vernay, B.; Savery, D.; Gurniak, C.B.; Witke, W.; Greene, N.D.; Copp, A.J. Rho-kinase-dependent actin turnover and actomyosin disassembly are necessary for mouse spinal neural tube closure. J. Cell. Sci. 2015, 128, 2468–2481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Aggelou, H.; Chadla, P.; Nikou, S.; Karteri, S.; Maroulis, I.; Kalofonos, H.P.; Papadaki, H.; Bravou, V. LIMK/cofilin pathway and Slingshot are implicated in humancolorectal cancer progression and chemoresistance. Virchows. Arch. 2018, 472, 727–737. [Google Scholar] [CrossRef]
  49. George, J.; Soares, C.; Montersino, A.; Beique, J.C.; Thomas, G.M. Palmitoylation of LIM Kinase-1 ensures spine-specific actin polymerization and morphologicalplasticity. eLife 2015, 4, e06327. [Google Scholar] [CrossRef]
  50. Zhang, W.; Ji, W.; Zhao, X. MiR-155 promotes anaplastic thyroid cancer progression by directly targeting SOCS. BMC Cancer 2019, 19, 1093. [Google Scholar] [CrossRef]
  51. Oh, J.; Kim, S.H.; Ahn, S.; Lee, C.E. Suppressors of cytokine signaling promote Fas-induced apoptosis through downregulation of NF-kappaB and mitochondrial Bfl-1 in leukemic T cells. J. Immunol. 2012, 189, 5561–5571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Xue, X.; Liu, Y.; Wang, Y.; Meng, M.; Wang, K.; Zang, X.; Zhao, S.; Sun, X.; Cui, L.; Pan, L.; et al. MiR-21 and MiR-155 promote non-small cell lung cancer progression by downregulating SOCS1, SOCS6, and PTEN. Oncotarget 2016, 7, 84508–84519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Merkel, O.; Hamacher, F.; Griessl, R.; Grabner, L.; Schiefer, A.I.; Prutsch, N.; Baer, C.; Egger, G.; Schlederer, M.; Krenn, P.W.; et al. Oncogenic role of miR-155 in anaplastic large cell lymphoma lacking the t(2; 5) translocation. J. Pathol. 2015, 236, 445–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bu, Q.; You, F.; Pan, G.; Yuan, Q.; Cui, T.; Hao, L.; Zhang, J. MiR-125b inhibits anaplastic thyroid cancer cell migration and invasion by targeting PIK3CD. Biomed. Pharmacother. 2017, 88, 443–448. [Google Scholar] [CrossRef] [PubMed]
  55. Dong, T.; Liu, Z.; Zhao, S.; Hu, C.; Liu, Y.; Ma, W.; Zhang, Q. The Expression of CD9 and PIK3CD is associated with prognosis of follicular lymphoma. J. Cancer 2015, 6, 1222–1229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Liao, W.T.; Ye, Y.P.; Zhang, N.J.; Li, T.T.; Wang, S.Y.; Cui, Y.M.; Qi, L.; Wu, P.; Jiao, H.L.; Xie, Y.J.; et al. MicroRNA-30b functions as a tumour suppressor in human colorectal cancer by targeting KRAS, PIK3CD and BCLJ. Pathology 2014, 232, 415–427. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, X.; Liu, L.; Deng, X.; Li, D.; Cai, H.; Ma, Y.; Jia, C.; Wu, B.; Fan, Y.; Lv, Z. MicroRNA 483-3p targets Pard3 to potentiate TGF-β1-induced cell migration, invasion, and epithelial-mesenchymal transition in anaplastic thyroid cancer cells. Oncogene 2019, 38, 699–715. [Google Scholar] [CrossRef] [PubMed]
  58. Hayashi, K.; Suzuki, A.; Ohno, S. PAR-1/MARK: A kinase essential for maintaining the dynamic state of microtubules. Cell. Struct. Funct. 2012, 37, 21–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Hardin, H.; Guo, Z.; Shan, W.; Montemayor-Garcia, C.; Asioli, S.; Yu, X.M.; Harrison, A.D.; Chen, H.; Lloyd, R.V. The roles of the epithelial-mesenchymal transition marker PRRX1 and miR-146b-5p in papillary thyroid carcinoma progression. Am. J. Pathol. 2014, 184, 2342–2354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Zhang, J.; Wang, Y.; Li, D.; Jing, S. Notch and TGF-beta/Smad3 pathways are involved in the interaction between cancer cells and cancer-associated fibroblasts in papillary thyroid carcinoma. Tumour. Biol. 2014, 35, 379–385. [Google Scholar] [CrossRef]
  61. Visone, R.; Pallante, P.; Vecchione, A.; Cirombella, R.; Ferracin, M.; Ferraro, A.; Volinia, S.; Coluzzi, S.; Leone, V.; Borbone, E.; et al. Specific microRNAs are downregulated in human thyroid anaplastic carcinomas. Oncogene 2007, 26, 7590–7595. [Google Scholar] [CrossRef] [PubMed]
  62. Esposito, F.; Tornincasa, M.; Pallante, P.; Federico, A.; Borbone, E.; Pierantoni, G.M.; Fusco, A. Down-regulation of the miR-25 and miR-30d contributes to the development of anaplastic thyroid carcinoma targeting the polycomb protein EZH. J. Clin. Endocrinol. Metab. 2012, 97, E710–E718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Maroof, H.; Islam, F.; Dong, L.; Ajjikuttira, P.; Gopalan, V.; McMillan, N.A.J.; Lam, A.K. Liposomal Delivery of miR-34b-5p Induced Cancer Cell Death in Thyroid Carcinoma. Cells 2018, 7, 265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhang, Z.; Liu, Z.B.; Ren, W.M.; Ye, X.G.; Zhang, Y.Y. The miR-200 family regulates the epithelial-mesenchymal transition induced by EGF/EGFR in anaplastic thyroid cancer cells. Int. J. Mol. Med. 2012, 30, 856–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zhang, K.; Zhang, H.; Xiang, H.; Liu, J.; Liu, Y.; Zhang, X.; Wang, J.; Tang, Y. TGF-β1 induces the dissolution of tight junctions in human renal proximal tubularcells: Role of the RhoA/ROCK signaling pathway. Int. J. Mol. Med. 2013, 32, 464–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Markowski, M.C.; Brown, A.C.; Barker, T.H. Directing epithelial to mesenchymal transition through engineered microenvironments displaying orthogonal adhesive and mechanical cues. J. Biomed. Mater. Res. A. 2012, 100, 2119–2127. [Google Scholar] [CrossRef] [PubMed]
  67. Ramírez-Moya, J.; Wert-Lamas, L.; Santisteban, P. MicroRNA-146b promotes PI3K/AKT pathway hyperactivation and thyroid cancer progression by targeting PTEN. Oncogene 2018, 37, 3369–3383. [Google Scholar] [CrossRef] [PubMed]
  68. Zhu, H.; Wu, H.; Liu, X.; Li, B.; Chen, Y.; Ren, X.; Liu, C.G.; Yang, J.M. Regulation of autophagy by a beclin 1-targeted microRNA, miR-30a, in cancer cells. Autophagy 2009, 5, 816–823. [Google Scholar] [CrossRef] [Green Version]
  69. Johnson, S.M.; Grosshans, H.; Shingara, J.; Byrom, M.; Jarvis, R.; Cheng, A.; Labourier, E.; Reinert, K.L.; Brown, D.; Slack, F.J. RAS is regulated by the let-7 microRNA family. Cell 2005, 120, 635–647. [Google Scholar] [CrossRef] [Green Version]
  70. Maroof, H.; Irani, S.; Arianna, A.; Vider, J.; Gopalan, V.; Lam, A.K. Interactions of Vascular Endothelial Growth Factor and p53 with miR-195 in Thyroid Carcinoma: Possible Therapeutic Targets in Aggressive Thyroid Cancers. Current. Cancer Drug Targets 2019, 19, 561–570. [Google Scholar] [CrossRef]
  71. Salajegheh, A.; Vosgha, H.; Rahman, M.A.; Amin, M.; Smith, R.A.; Lam, A.K. Interactive role of miR-126 on VEGF-A and progression of papillary and undifferentiated thyroid carcinoma. Human. Pathol. 2016, 51, 75–85. [Google Scholar] [CrossRef] [PubMed]
  72. Murugan, A.K.; Munirajan, A.K.; Alzahrani, A.S. Long noncoding RNAs: Emerging players in thyroid cancer pathogenesis. Endocr. Relat. Cancer 2018, 25, R59–R82. [Google Scholar] [CrossRef]
  73. Samimi, H.; Sajjadi-Jazi, S.M.; Seifirad, S.; Atlasi, R.; Mahmoodzadeh, H.; Faghihi, M.A.; Haghpanah, V. Molecular mechanisms of long non-coding RNAs in anaplastic thyroid cancer: A systematic review. Cancer Cell. Int. 2020, 20, 352. [Google Scholar] [CrossRef]
  74. Zhang, H.; Yu, Y.; Zhang, K.; Liu, X.; Dai, Y.; Jiao, X. Targeted inhibition of long non-coding RNA H19 blocks anaplastic thyroid carcinoma growth and metastasis. Bioengineered 2019, 10, 306–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Raveh, E.; Matouk, I.J.; Gilon, M.; Hochberg, A. The H19 Long non-coding RNA in cancer initiation, progression and metastasis-a proposed unifying theory. Mol. Cancer 2015, 14, 184. [Google Scholar] [CrossRef] [Green Version]
  76. Jantscheff, P.; Esser, N.; Geipel, A.; Woias, P.; Ziroli, V.; Goldschmidtboing, F.; Massing, U. Metastasizing, Luciferase Transduced MAT-Lu Rat Prostate Cancer Models: Follow up of Bolus and Metronomic Therapy with Doxorubicin as Model Drug. Cancers 2011, 3, 2679–2695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Surguladze, D.; Steiner, P.; Prewett, M.; Tonra, J.R. Methods for evaluating effects of an irinotecan + 5-fluorouracil/leucovorin (IFL) regimen in an orthotopic metastatic colorectal cancer model utilizing in vivo bioluminescence imaging. Methods. Mol. Biol. 2010, 602, 235–252. [Google Scholar] [PubMed]
  78. Chen, J.; Zhao, D.; Meng, Q. Knockdown of HCP5 exerts tumor-suppressive functions by up-regulating tumor suppressor miR-128-3p in anaplastic thyroid Cancer. Biomed. Pharmacother. 2019, 116, 108966. [Google Scholar] [CrossRef]
  79. Wang, Y.; Hou, Z.; Li, D. Long noncoding RNA UCA1 promotes anaplastic thyroid cancer cell proliferation via miR-135a-mediated c-myc activation. Mol. Med. Rep. 2018, 18, 3068–3076. [Google Scholar] [CrossRef] [Green Version]
  80. Enomoto, K.; Zhu, X.; Park, S.; Zhao, L.; Zhu, Y.J.; Willingham, M.C.; Qi, J.; Copland, J.A.; Meltzer, P.; Cheng, S.Y. Targeting MYC as a Therapeutic Intervention for Anaplastic Thyroid Cancer. J. Clin. Endocrinol. Metab. 2017, 102, 2268–2280. [Google Scholar] [CrossRef] [Green Version]
  81. Dang, C.V. MYC on the path to Cancer. Cell 2012, 149, 22–35. [Google Scholar] [CrossRef] [Green Version]
  82. Yan, P.; Su, Z.; Zhang, Z.; Gao, T. LncRNA NEAT1 enhances the resistance of anaplastic thyroid carcinoma cells to cisplatin by sponging miR-9-5p and regulating SPAG9 expression. Int. J. Oncol. 2019, 55, 988–1002. [Google Scholar] [CrossRef] [Green Version]
  83. Yu, H.M.; Wang, C.; Yuan, Z.; Chen, G.L.; Ye, T.; Yang, B.W. LncRNA NEAT1 promotes thetumorigenesis of colorectal cancer by sponging miR-193a-3p. Cell. Prolif. 2019, 52, e12526. [Google Scholar] [CrossRef]
  84. Chen, Z.J.; Zhang, Z.; Xie, B.B.; Zhang, H.Y. Clinical significance of up-regulated lncRNA NEAT1 in prognosis of ovarian Cancer. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 3373–3377. [Google Scholar]
  85. Choudhry, H.; Albukhari, A.; Morotti, M.; Haider, S.; Moralli, D.; Smythies, J.; Schödel, J.; Green, C.M.; Camps, C.; Buffa, F.; et al. Tumor hypoxia induces nuclear paraspeckle formation through HIF-2α dependent transcriptional activation of NEAT1 leading to cancer cell survival. Oncogene 2015, 34, 4482–4490. [Google Scholar] [CrossRef] [Green Version]
  86. Chen, X.; Kong, J.; Ma, Z.; Gao, S.; Feng, X. Up regulation of the long non-coding RNA NEAT1 promotes esophageal squamous cell carcinoma cell progression and correlates with poor prognosis. Am. J. Cancer Res. 2015, 5, 2808–2815. [Google Scholar]
  87. Hu, Y.; Yang, Q.; Wang, L.; Wang, S.; Sun, F.; Xu, D.; Jiang, J. Knockdown of the oncogene lncRNA NEAT1 restores the availability of miR-34c and improves the sensitivity to cisplatin in osteosarcoma. Biosci. Rep. 2018, 38, BSR20180375. [Google Scholar] [CrossRef] [Green Version]
  88. An, J.; Lv, W.; Zhang, Y. LncRNA NEAT1 contributes to paclitaxel resistance of ovarian cancer cells by regulating ZEB1 expression via miR-194. Onco Targets. Ther. 2017, 10, 5377–5390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Gao, C.; Zhang, J.; Wang, Q.; Ren, C. Overexpression of lncRNA NEAT1 mitigates multidrug resistance by inhibiting ABCG2 in leukemia. Oncol. Lett. 2016, 12, 1051–1057. [Google Scholar] [CrossRef] [Green Version]
  90. Pellecchia, S.; Sepe, R.; Decaussin-Petrucci, M.; Ivan, C.; Shimizu, M.; Coppola, C.; Testa, D.; Calin, G.A.; Fusco, A.; Pallante, P. The Long Non-Coding RNA Prader Willi/Angelman Region RNA5 (PAR5) is downregulated in anaplastic thyroid carcinomas where it acts as a tumor suppressor by reducing EZH2 activity. Cancers 2020, 12, 235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Borbone, E.; Troncone, G.; Ferraro, A.; Jasencakova, Z.; Stojic, L.; Esposito, F.; Hornig, N.; Fusco, A.; Orlando, V. Enhancer of zeste homolog 2 overexpression has a role in the development of anaplastic thyroid carcinomas. J. Clin. Endocrinol. Metab. 2011, 96, 1029–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Li, Z.; Hou, P.; Fan, D.; Dong, M.; Ma, M.; Li, H.; Yao, R.; Li, Y.; Wang, G.; Geng, P.; et al. The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast Cancer. Cell Death Differ. 2017, 24, 59–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Jin, L.; Cai, Q.; Wang, S.; Mondal, T.; Wang, J.; Quan, Z. Long noncoding RNA MEG3 regulates LATS2 by promoting the ubiquitination of EZH2 and inhibits proliferation and invasion in gallbladder Cancer. Cell. Death. Dis. 2018, 9, 1017. [Google Scholar] [CrossRef] [Green Version]
  94. Serrano-Gomez, S.J.; Maziveyi, M.; Alahari, S.K. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol. Cancer 2016, 15, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Petrova, Y.I.; Schecterson, L.; Gumbiner, B.M. Roles for E-cadherin cell surface regulation in Cancer. Mol. Biol. Cell. 2016, 27, 3233–3244. [Google Scholar] [CrossRef]
  96. Jolly, M.K.; Ware, K.E.; Xu, S.; Gilja, S.; Shetler, S.; Yang, Y.; Wang, X.; Austin, R.G.; Runyambo, D.; Hish, A.J.; et al. E-Cadherin Represses Anchorage-Independent Growth in Sarcomas through Both Signaling and Mechanical Mechanisms. Mol. Cancer Res. 2019, 17, 1391–1402. [Google Scholar] [CrossRef] [Green Version]
  97. Loh, C.Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [Green Version]
  98. Chen, W.M.; Chen, W.D.; Jiang, X.M.; Jia, X.F.; Wang, H.M.; Zhang, Q.J.; Shu, Y.Q.; Zhao, H.B. HOXtranscript antisense intergenic RNA represses E-cadherin expression by binding to EZH2 in gastric Cancer. World J. Gastroenterol. 2017, 23, 6100–6110. [Google Scholar] [CrossRef]
  99. Zhou, Q.; Chen, J.; Feng, J.; Wang, J. Long noncoding RNA PVT1 modulates thyroid cancer cell proliferation by recruiting EZH2 and regulating thyroid-stimulating hormone receptor (TSHR). Tumour. Biol. 2016, 37, 3105–3113. [Google Scholar] [CrossRef]
  100. Colombo, T.; Farina, L.; Macino, G.; Paci, P. PVT1: A rising star among oncogenic long noncoding RNAs. Biomed. Res. Int. 2015, 2015, 304208. [Google Scholar] [CrossRef]
  101. Huang, C.; Yu, W.; Wang, Q.; Cui, H.; Wang, Y.; Zhang, L.; Han, F.; Huang, T. Increased expression of the lncRNA PVT1 is associated with poor prognosis in pancreatic cancer patients. Minerva. Med. 2015, 106, 143–149. [Google Scholar] [PubMed]
  102. Yang, Y.R.; Zang, S.Z.; Zhong, C.L.; Li, Y.X.; Zhao, S.S.; Feng, X.J. Increased expression of the lncRNA PVT1 promotes tumorigenesis in non-small cell lung Cancer. Int. J. Clin. Exp. Pathol. 2014, 7, 6929–6935. [Google Scholar] [PubMed]
  103. D’Agostino, M.; Sponziello, M.; Puppin, C.; Celano, M.; Maggisano, V.; Baldan, F.; Biffoni, M.; Bulotta, S.; Durante, C.; Filetti, S.; et al. Differentexpression of TSH receptor and NIS genes in thyroid cancer: Role of epigenetics. J. Mol. Endocrinol. 2014, 52, 121–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Schmidt, L.H.; Spieker, T.; Koschmieder, S.; Humberg, J.; Jungen, D.; Bulk, E.; Hascher, A.; Wittmer, D.; Marra, A.; Hillejan, L.; 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. [Google Scholar] [CrossRef] [Green Version]
  105. Gutschner, T.; Hämmerle, M.; Eißmann, M.; Hsu, J.; Kim, Y.; Hung, G.; Revenko, A.; Arun, G.; Stentrup, M.; Groß, M.; et al. The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 2013, 73, 31180–31189. [Google Scholar] [CrossRef] [Green Version]
  106. Cheetham, S.; Gruhl, F.; Mattick, J.; Dinger, M. Long noncoding RNAs and the genetics of Cancer. Br. J. Cancer 2013, 108, 2419–2425. [Google Scholar] [CrossRef] [Green Version]
  107. Zhang, R.; Xia, L.Q.; Lu, W.W.; Zhang, J.; Zhu, J.S. LncRNAs and Cancer. Oncol. Lett. 2016, 12, 1233–1239. [Google Scholar] [CrossRef] [Green Version]
  108. Samimi, H.; Haghpanah, V.; Irani, S.; Arefian, E.; Sohi, A.N.; Fallah, P.; Soleimani, M. Transcript-level regulation of MALAT1-mediated cell cycle and apoptosis genes using dual MEK/Aurora kinase inhibitor "BI-847325" on anaplastic thyroidcarcinoma. Daru 2019, 27, 1–7. [Google Scholar] [CrossRef]
  109. Wu, X.S.; Wang, X.A.; Wu, W.G.; Hu, Y.P.; Li, M.L.; Ding, Q.; Weng, H.; Shu, Y.-J.; Liu, T.-Y.; Jiang, L.; et al. MALAT1 promotes the proliferation and metastasis of gallbladder cancer cells by activating the ERK/MAPK pathway. Cancer Biol. Ther. 2014, 15, 806–814. [Google Scholar] [CrossRef] [Green Version]
  110. Dong, Y.; Liang, G.; Yuan, B.; Yang, C.; Gao, R.; Zhou, X. MALAT1 promotes the proliferation and metastasis of osteosarcoma cells by activating the PI3K/Akt pathway. Tumour. Biol. 2015, 36, 1477–1486. [Google Scholar] [CrossRef]
  111. Zhao, J.; Li, L.; Peng, L. MAPK1 up-regulates the expression of MALAT1 to promote the proliferation of cardiomyocytes through PI3K/AKT signaling pathway. Int. J. Clin. Exp. Pathol. 2015, 8, 15947. [Google Scholar]
  112. Li, C.; Chang, L.; Chen, Z.; Liu, Z.; Wang, Y.; Ye, Q. The role of lncRNA MALAT1 in the regulation of hepatocyte proliferation during liver regeneration. Int. J. Mol. Med. 2017, 39, 347–356. [Google Scholar] [CrossRef] [Green Version]
  113. Yu, H.; Lee, H.; Herrmann, A.; Buettner, R.; Jove, R. Revisiting STAT3 signalling in cancer: New and unexpected biological functions. Nat. Rev. Cancer 2014, 14, 736–746. [Google Scholar] [CrossRef]
  114. Wang, X.M.; Liu, Y.; Fan, Y.X.; Liu, Z.; Yuan, Q.L.; Jia, M.; Geng, Z.; Gu, L.; Lu, X. LncRNA PTCSC3 affects drug resistance of anaplastic thyroid cancer through STAT3/INO80 pathway. Cancer Biol. Ther. 2018, 19, 590–597. [Google Scholar] [CrossRef] [Green Version]
  115. Jendrzejewski, J.; He, H.; Radomska, H.S.; Li, W.; Tomsic, J.; Liyanarachchi, S.; Davuluri, R.V.; Nagy, R.; de la Chapelle, A. The polymorphismrs944289 predisposes to papillary thyroid carcinoma through a large intergenic noncoding RNA gene of tumor suppressor type. Proc. Natl. Acad. Sci. USA 2012, 109, 8646–8651. [Google Scholar] [CrossRef] [Green Version]
  116. Jendrzejewski, J.; Thomas, A.; Liyanarachchi, S.; Eiterman, A.; Tomsic, J.; He, H.; Radomska, H.S.; Li, W.; Nagy, R.; Sworczak, K.; et al. PTCSC3 Is Involved in Papillary Thyroid Carcinoma Development by Modulating S100A4 Gene Expression. J. Clin. Endocrinol. Metab. 2015, 100, E1370–E1377. [Google Scholar] [CrossRef] [Green Version]
  117. Conaway, R.C.; Conaway, J.W. The INO80 chromatin remodeling complex in transcription, replication and repair. Trends. Biochem. Sci. 2009, 34, 71–77. [Google Scholar] [CrossRef]
  118. Decaussin-Petrucci, M.; Delado€ey, J.; Hafdi-Nejjari, Z.; Sassolas, G.; Borson-Chazot, F.; Abu-Khudir, R.; Fusco, A.; Descotes, F.; Cournoyer, S.; Sartelet, H.; et al. Expression of CD133 in differentiated thyroid cancer of young patients. J. Clin. Pathol. 2015, 68, 434–440. [Google Scholar] [CrossRef] [Green Version]
  119. Berlingieri, M.T.; Manfiletti, G.; Santoro, M.; Bandiera, A.; Visconti, R.; Giancotti, V.; Fusco, A. Inhibition of HMGI-C protein synthesis suppresses retrovirally induced neoplastic transformation of rat thyroid cells. Mol. Cell. Biol. 1995, 15, 1545–1553. [Google Scholar] [CrossRef] [Green Version]
  120. Berlingieri, M.T.; Pierantoni, G.M.; Giancotti, V.; Santoro, M.; Fusco, A. Thyroid cell transformation requires the expression of the HMG1 proteins. Oncogene 2002, 21, 2971–2980. [Google Scholar] [CrossRef] [Green Version]
  121. Esposito, F.; De Martino, M.; Petti, M.G.; Forzati, F.; Tornincasa, M.; Federico, A.; Arra, C.; Pierantoni, G.M.; Fusco, A. HMGA1 pseudogenes as candidate proto-oncogenic competitive endogenous RNAs. Oncotarget 2014, 5, 8341–8354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Nambudiri, V.E.; Widlund, H.R. Small interfering RNA. J. Investig. Dermatol. 2013, 133, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Nikam, R.R.; Gore, K.R. Journey of siRNA: Clinical Developments and Targeted Delivery. Nucleic. Acid. Ther. 2018, 28, 209–224. [Google Scholar] [CrossRef] [PubMed]
  124. Singh, A.; Trivedi, P.; Jain, N.K. Advances in siRNA delivery in cancer therapy. Artif. Cells. Nanomed. Biotechnol. 2018, 46, 274–283. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, X.; Mangala, L.S.; Rodriguez-Aguayo, C.; Kong, X.; Lopez-Berestein, G.; Sood, A.K. RNA interference-based therapy and its delivery systems. Cancer Metastasis Rev. 2018, 37, 107–124. [Google Scholar] [CrossRef] [PubMed]
  126. Hu, B.; Weng, Y.; Xia, X.H.; Liang, X.J.; Huang, Y. Clinical advances of siRNA therapeutics. J. Gene. Med. 2019, 21, e3097. [Google Scholar] [CrossRef] [PubMed]
  127. Zhou, W.; Wang, J.; Man, W.Y.; Zhang, Q.W.; Xu, W.G. siRNA silencing EZH2 reverses cisplatin-resistance of human non-small cell lung and gastric cancer cells. Asian. Pac. J. Cancer Prev. 2015, 16, 2425–2430. [Google Scholar] [CrossRef] [Green Version]
  128. Saini, S.; Sripada, L.; Tulla, K.; Kumar, P.; Yue, F.; Kunda, N.; Maker, A.V.; Prabhakar, B.S. Loss of MADD expression inhibits cellular growth and metastasis in anaplastic thyroid Cancer. Cell. Death. Dis. 2019, 10, 145. [Google Scholar] [CrossRef]
  129. Kurada, B.R.; Li, L.C.; Mulherkar, N.; Subramanian, M.; Prasad, K.V.; Prabhakar, B.S. MADD, a splice variant of IG20, is indispensable for MAPK activation and protection against apoptosis upon tumor necrosis factor-alpha treatment. J. Biol. Chem. 2009, 284, 13533–13541. [Google Scholar] [CrossRef] [Green Version]
  130. Okamoto, N.; Takahashi, A.; Ota, I.; Ohnishi, K.; Mori, E.; Kondo, N.; Noda, T.; Nakagawa, Y.; Uemura, H.; Yane, K.; et al. siRNA targeted for NBS1 enhances heat sensitivity in human anaplastic thyroid carcinoma cells. Int. J. Hyperthermia. 2011, 27, 297–304. [Google Scholar] [CrossRef]
  131. Takahashi, A.; Matsumoto, H.; Nagayama, K.; Kitano, M.; Hirose, S.; Tanaka, H.; Mori, E.; Yamakawa, N.; Yasumoto, J.; Yuki, K.; et al. Evidence for the involvement of double-strandbreaks in heat-induced cell killing. Cancer Res. 2004, 64, 8839–8845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Sedelnikova, O.A.; Rogakou, E.P.; Panyutin, I.G.; Bonner, W.M. Quantitative detection of125IdU-induced DNA double-strandbreaks with gamma-H2AX antibody. Radiat. Res. 2002, 158, 486–492. [Google Scholar] [CrossRef]
  133. Pan, Y.; Liu, P.; Chen, D.; Dou, L. Small interfering RNA (siRNA) against Slug induces apoptosis and sensitizes human anaplastic thyroid carcinoma cells to doxorubicin. Cancer Biomark. 2017, 18, 357–366. [Google Scholar] [CrossRef] [PubMed]
  134. Wei, C.R.; Liu, J.; Yu, X.J. Targeting SLUG sensitizes leukemia cells to ADR-induced apoptosis. Int. J. Clin. Exp. Med. 2015, 8, 22139–22148. [Google Scholar]
  135. Torreggiani, E.; Lisignoli, G.; Manferdini, C.; Lambertini, E.; Penolazzi, L.; Vecchiatini, R.; Gabusi, E.; Chieco, P.; Facchini, A.; Gambari, R.; et al. Role of Slug transcription factor in human mesenchymal stem cells. J. Cell. Mol. Med. 2012, 16, 740–751. [Google Scholar] [CrossRef]
  136. Xu, T.; Fan, B.; Lv, C.; Xiao, D. Slug mediates nasopharyngeal carcinoma radioresistance via downregulation of PUMA in a p53-dependent and -independent manner. Oncol. Rep. 2015, 33, 2631–2638. [Google Scholar] [CrossRef] [Green Version]
  137. Buehler, D.; Hardin, H.; Shan, W.; Montemayor-Garcia, C.; Rush, P.S.; Asioli, S.; Chen, H.; Lloyd, R.V. Expression of epithelial-mesenchymal transition regulators SNAI2 and TWIST1 in thyroid carcinomas. Mod. Pathol. 2013, 26, 54–61. [Google Scholar] [CrossRef]
Figure 1. Regulation of non-coding RNA mediated gene expression in anaplastic thyroid carcinoma (ATC). (a) In normal cells, miR-205can target and inhibit the expression of vascular endothelial growth factor A (VEGF-A) and Zinc Finger E-Box Binding Homeobox 1 (ZEB1). In cancer cells, the expression of miR-205 is downregulated, which subsequently results into angiogenesis, epithelial-mesenchymal transition (EMT) and invasion of ATC cells. (b) siRNA mediates gene silencing in ATC cells. siRNA interact with DICER and facilitate RNA-induced silencing complex (RISC) mediated mRNA degradation. (c) Nuclear enriched abundant transcript 1 (NEAT1) is upregulated in ATC cells where it inhibits the expression of miR-9-5p and Sperm associated antigen 9 (SPAG9), which increases chemoresistance of ATC. On the other hand, in normal cells, NEAT1 is downregulated, which frees miR-9-5p and SPAG9 to mediate their normal function.
Figure 1. Regulation of non-coding RNA mediated gene expression in anaplastic thyroid carcinoma (ATC). (a) In normal cells, miR-205can target and inhibit the expression of vascular endothelial growth factor A (VEGF-A) and Zinc Finger E-Box Binding Homeobox 1 (ZEB1). In cancer cells, the expression of miR-205 is downregulated, which subsequently results into angiogenesis, epithelial-mesenchymal transition (EMT) and invasion of ATC cells. (b) siRNA mediates gene silencing in ATC cells. siRNA interact with DICER and facilitate RNA-induced silencing complex (RISC) mediated mRNA degradation. (c) Nuclear enriched abundant transcript 1 (NEAT1) is upregulated in ATC cells where it inhibits the expression of miR-9-5p and Sperm associated antigen 9 (SPAG9), which increases chemoresistance of ATC. On the other hand, in normal cells, NEAT1 is downregulated, which frees miR-9-5p and SPAG9 to mediate their normal function.
Cancers 12 03159 g001
Figure 2. Role of miRNAs in the regulation of gene expression in anaplastic thyroid carcinoma (ATC). miRNAs upregulate or downregulate the genes related to development, progression, invasion migration, and therapy resistance in ATC. PTEN: Phosphatase and tensin homolog; XIAP: X-linked inhibitor of apoptosis protein; SOCS1: Suppressor of Cytokine Signaling 1; PIK3CD: Phosphoinositide 3-kinase catalytic subunit delta.
Figure 2. Role of miRNAs in the regulation of gene expression in anaplastic thyroid carcinoma (ATC). miRNAs upregulate or downregulate the genes related to development, progression, invasion migration, and therapy resistance in ATC. PTEN: Phosphatase and tensin homolog; XIAP: X-linked inhibitor of apoptosis protein; SOCS1: Suppressor of Cytokine Signaling 1; PIK3CD: Phosphoinositide 3-kinase catalytic subunit delta.
Cancers 12 03159 g002
Table 1. Potential functions of miRNAs in anaplastic thyroid carcinoma (ATC).
Table 1. Potential functions of miRNAs in anaplastic thyroid carcinoma (ATC).
MiRNAsExpression Pattern in ATCPotential Target (s)Potential FunctionsReference
miR-205DownregulatedVEGF-A, ZEB1Causes significant inhibition of angiogenesis and EMT
Inhibits tumour growth, vascularization, migration and invasion.
[37]
miR-19aUpregulatedPTENIncreases cellular proliferation and progression by suppressing the expression of tumour suppressor PTEN[38,39]
miR-17-3p and miR-17-5pUpregulatedUnknownInhibition of these miRNA expression results in growth arrest and induction of apoptosis[40]
miR-618DownregulatedXIAPInhibits cell growth and induces apoptosis[41]
miR-20aDownregulatedLIMK1Inhibits proliferation, migration and invasion[42]
miR-155UpregulatedSOCS1Inhibits apoptosis, promotes proliferation, invasion and migration of ATC cells[51]
miR-125bDownregulatedPIK3CDDecreases the PI3K, phospho-Akt, and phospho-mTOR expression in ATC cells
Inhibits migration and invasion of ATC cells
[54]
miR-483UpregulatedPard3Causes significant increase in cell growth, migration, and invasion.[57]
miR-25 and miR-30dDownregulatedEZH2Inhibits proliferation and colony formation of ATC cells by inducing G2/M-phase cell-cycle arrest[62]
MiR-34bDownregulatedVEGF-ADecreases cell proliferation, and wound healing, reduces cell cycle progression and increases apoptosis[63]
miR-200 familyDownregulatedZEB1 and ZEB2Inhibits TGF-mediated EMT switch and decreases aggressiveness of ATC[64]
miR-146bUpregulatedPTENInhibits the expression of tumour suppressor PTEN
Decreases apoptosis, increases migration and invasion potential by regulating genes related to EMT
Activates PI3K/AKT signalling to drive oncogenic proliferation
[67]
Table 2. Implications of lncRNAs in anaplastic thyroid carcinoma (ATC).
Table 2. Implications of lncRNAs in anaplastic thyroid carcinoma (ATC).
LncRNAsExpression Pattern in ATCPotential Target (s)Potential Functions in ATCReference
H19UpregulatedUnknownIncreases proliferation, migration and decreases apoptosis in ATC cells[74]
HLA complex P5 (HCP5)UpregulatedmiR-128-3pIncreases cell viability, and decreases apoptosis[78]
Urothelial carcinoma-associated 1 (UCA1)UpregulatedmiR-135aPromotes ATC cell proliferation by acting as a competing endogenous RNA by binding with miR-135a
Increases cell viability, proliferation, migration and invasion in ATC cells
[79]
Nuclear paraspeckle assembly transcript 1 (NEAT1)UpregulatedmiR-9-5pIncreases chemoresistance to cisplatin via miR-9-5p/SPAG9 axis in vitro and in vivo[82]
Prader Willi/Angelman
Region RNA5 (PAR5)
DownregulatedEnhancer of Zeste Homolog 2 (EZH2)
E-cadherin
Reduces cell proliferation and cell migration ability
Increases E-cadherin expression
Mediates its action by impairing EZH2 oncogenic activity
[90]
PVT1UpregulatedEZH2Increases cell proliferation by enhancing cell cycle progression, cyclin D and TSHR expression[103]
MALAT1UpregulatedmiR-363-3pInhibits the expression of miR-363-3p by binding to it, thereby releases inhibitory effect of miR-363-3p to Mcl1 oncogene
Also decreases the expression of cell cycle and apoptosis related gene cyclin D1
[108]
PTCSC3DownregulatedSTAT3Inhibits the expression of INO80 through negatively regulating STAT3, which subsequently decreases drug resistance of ATC to doxorubicin.
PTCSC3 also inhibits stem cell properties of ATC 8050C cells
[114]
HMGA1PsUpregulatedmiR-16 and miR-17 etc.Leads to increase of HMGA2, and other cancer related proteins like VEGF and EZH2 levels[121]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Das, P.K.; Asha, S.Y.; Abe, I.; Islam, F.; Lam, A.K. Roles of Non-Coding RNAs on Anaplastic Thyroid Carcinomas. Cancers 2020, 12, 3159. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12113159

AMA Style

Das PK, Asha SY, Abe I, Islam F, Lam AK. Roles of Non-Coding RNAs on Anaplastic Thyroid Carcinomas. Cancers. 2020; 12(11):3159. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12113159

Chicago/Turabian Style

Das, Plabon Kumar, Saharia Yeasmin Asha, Ichiro Abe, Farhadul Islam, and Alfred K. Lam. 2020. "Roles of Non-Coding RNAs on Anaplastic Thyroid Carcinomas" Cancers 12, no. 11: 3159. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12113159

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop