Next Article in Journal
Characteristic of Uterine Rhabdomyosarcoma by Algorithm of Potential Biomarkers for Uterine Mesenchymal Tumor
Next Article in Special Issue
Hereditary Diffuse Gastric Cancer—Update Based on the Current Consort Recommendations
Previous Article in Journal
Comprehensive Treatment of Hematological Patients with SARS-CoV-2 Infection Including Anti-SARS-CoV-2 Monoclonal Antibodies: A Single-Center Experience Case Series
Previous Article in Special Issue
Impact of Postoperative Chemotherapy in Patients with Gastric/Gastroesophageal Adenocarcinoma Treated with Perioperative Chemotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Pleiotropic Role of Long Non-Coding RNAs in the Modulation of Wnt/β-Catenin and PI3K/Akt/mTOR Signaling Pathways in Esophageal Squamous Cell Carcinoma: Implication in Chemotherapeutic Drug Response

1
Department of Zoology, Central University of Punjab, Ghudda 151 401, Punjab, India
2
Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133 207, Haryana, India
3
Department of Biochemistry, Central University of Punjab, Ghudda 151 401, Punjab, India
4
Amity Institute of Virology and Immunology, Amity University, Noida 201 301, Uttar Pradesh, India
5
College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
*
Authors to whom correspondence should be addressed.
Curr. Oncol. 2022, 29(4), 2326-2349; https://doi.org/10.3390/curroncol29040189
Submission received: 4 February 2022 / Revised: 19 March 2022 / Accepted: 20 March 2022 / Published: 26 March 2022

Abstract

:
Despite the availability of modern techniques for the treatment of esophageal squamous cell carcinoma (ESCC), tumor recurrence and metastasis are significant challenges in clinical management. Thus, ESCC possesses a poor prognosis and low five-year overall survival rate. Notably, the origin and recurrence of the cancer phenotype are under the control of complex cancer-related signaling pathways. In this review, we provide comprehensive knowledge about long non-coding RNAs (lncRNAs) related to Wnt/β-catenin and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway in ESCC and its implications in hindering the efficacy of chemotherapeutic drugs. We observed that a pool of lncRNAs, such as HERES, TUG1, and UCA1, associated with ESCC, directly or indirectly targets various molecules of the Wnt/β-catenin pathway and facilitates the manifestation of multiple cancer phenotypes, including proliferation, metastasis, relapse, and resistance to anticancer treatment. Additionally, several lncRNAs, such as HCP5 and PTCSC1, modulate PI3K/Akt/mTOR pathways during the ESCC pathogenesis. Furthermore, a few lncRNAs, such as AFAP1-AS1 and LINC01014, block the efficiency of chemotherapeutic drugs, including cisplatin, 5-fluorouracil, paclitaxel, and gefitinib, used for ESCC treatment. Therefore, this review may help in designing a better therapeutic strategy for ESCC patients.

1. Introduction

Worldwide, esophageal cancer (EC) ranks eighth and sixth in terms of incidence and mortality among all cancers, respectively [1]. Despite the advancement in diagnostic and therapeutic applications, the overall survival of esophageal squamous cell carcinoma (ESCC) patients is still meager. For example, the five-year survival rate of ESCC patients in several less developed countries is very low (~10%), whereas in developed countries, such as the United States, the five-year survival rate is ~18% [2]. Although chemotherapy and radiotherapy can increase the disease-free and overall survival among ESCC patients, tumor cells adopt the tendency to resist the effect of chemotherapeutic drugs or radiation doses, suggesting the development of therapy resistance mechanisms in tumor cells [3]. According to a previous report, chemotherapeutic drug resistance leads to more than 90% of deaths in patients with ESCC [4]. This may be due to the cross-networking of the vital biological, molecular, and cellular signaling pathways, such as Wnt/β-catenin, phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathways, with the various chemotherapeutic drugs [4], thus limiting the efficacy of therapies, resulting in poor prognosis, tumor metastasis, and recurrence [5]. Therefore, understanding the underpinnings that regulate the vital signaling pathways and resist the efficacy of the chemotherapeutic drugs, is urgently required.
In recent years, improved knowledge of oncology research has led to the identification of non-coding RNAs (ncRNAs) that regulate tumor cell proliferation, differentiation, angiogenesis, metastasis, and invasion. ncRNAs include structural and regulatory RNAs, representing ~90% of the human genome. Structural long non-coding RNAs (lncRNAs) include ribosomal RNAs and transfer RNAs, whereas regulatory RNAs include small conditional RNAs, small nucleolar RNAs, microRNAs (miRNAs), and lncRNAs [6,7]. Among them, lncRNAs having size ≥200 nucleotides are involved in various biological, molecular, and cellular processes, such as transcription, splicing, translation, protein localization, epigenetics, cell structure integrity, cell cycle, cell fate determination, cell differentiation, cell migration, and cell proliferation [8]. Furthermore, lncRNAs have been implicated in modulating various cancer-related signaling pathways, such as Wnt/β-catenin [9,10,11,12,13,14,15,16,17], PI3K/Akt/mTOR [18,19,20], Janus kinase/signal transducers and activators of transcription (JAK/STAT3) [21,22], mitogen-activated protein kinase 1 (MAPK) [23,24], nuclear factor-κB (NF-κB) [25,26], and NOTCH [27,28,29], and display the cancer phenotypes [9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29]. Additionally, an explosion of research revealed that lncRNAs act as a mediator in regulating chemoresistance by altering the efflux of a drug, DNA damage repair, inhibition of apoptosis, and mutation of the drug targets [3,30]. Furthermore, lncRNAs play an important role in conferring radioresistance in ESCC, as documented previously by our group [3].
Current research advancements in clinical oncology revealed that lncRNAs play a vital role in cancer therapeutics, diagnosis, and prognosis. Interestingly, ESCC attracted the interest of oncologists due to its delayed diagnosis and vast number of annual deaths. Based on this idea, we searched in PubMed with a combination of keywords: lncRNA; long non-coding RNA; and esophageal squamous cell carcinoma. We obtained a pool of lncRNAs evolved from 2012–2021 in ESCC, evidenced by the increasing number of research papers appearing in PubMed (Figure 1 and Supplementary Table S1). As a result, we found that ESCC-associated lncRNAs, such as LINC01014 [18], HCP5 [19], and PTCSC1 [20], modulate the PI3K/Akt/mTOR pathway. In addition, lncRNAs, namely LOC146880 [23], BANCR [31], and LINC00324 [24], are involved in MAPK signaling pathways. Similarly, studies reported that dysregulation of other lncRNAs (shown in Supplementary Table S1) dysregulate hedgehog [32], p53 [33], NF-κB [25,26], NOTCH [27,28], TGFβ1 [34], STAT [21,22], and Wnt/β-catenin signaling [9,10,11,12,13,14,15,16,17] in ESCC. Among all the signaling pathways, we identified most of the lncRNAs modulating the Wnt/β-catenin and PI3K/Akt/mTOR signaling pathways in ESCC, suggesting the implication of these two pathways during ESCC pathogenesis.
Previous studies highlighted the role of lncRNAs with Hippo, transforming growth factor beta (TGFβ)/SMAD, and JAK/STAT signaling pathways [7,35,36] but did not summarize the detailed association with Wnt/β-catenin and PI3K/Akt/mTOR in ESCC. Therefore, the objective of this work was to analyze the regulatory role of Wnt/β-catenin and PI3K/Akt/mTOR pathways in association with lncRNAs in ESCC and their role in chemotherapeutics drug response. Additionally, we have presented the crosstalk between the Wnt/β-catenin and PI3K/Akt/mTOR signaling pathway in ESCC. Thus, our review provides comprehensive knowledge about the underpinnings that need to be targeted to better the treatment of ESCC patients.

2. Wnt/β-Catenin Signaling Pathway-Related lncRNAs in ESCC

The Wnt signaling pathway is a well-known, evolutionarily conserved pathway that regulates cell proliferation, migration, and invasion and thus controls tumor progression [37]. Genetic and epigenetic alterations, such as DNA hypermethylation in the promoter region of axis inhibition protein 2 (Axin2), adenomatous polyposis coli (APC), Wnt inhibitory factor 1 (WIF-1), and secreted frizzled-related protein (SFRPs), lead to the aberrant activation of Wnt⁄β-catenin signaling pathway in several types of tumors, including ESCC [14]. Based on the signal transduction mechanism, Wnt signaling is classified into canonical and non-canonical pathways. Canonical Wnt signaling translates the transcriptional activator β-catenin into the nucleus, and constitutive activation leads to cancer pathogenesis. In contrast, non-canonical Wnt pathways are independent of β-catenin transcriptional activity and hence regulated via Wnt polarity, Wnt-Ca2+, and Wnt-atypical protein kinase signaling (Figure 2).
In cancer cells, several lncRNAs, such as highly expressed lncRNA in esophageal squamous cell carcinoma (HERES), small nucleolar RNA host gene 16 (SNHG16), urothelial cancer associated 1 (UCA1), maternally expressed 3 (MEG3), LINC00675, HOX antisense intergenic RNA (HOTAIR), taurine upregulated gene 1 (TUG1), and growth-arrest-associated long non-coding R/NA (GASL1), target and affect the expression of β-catenin, a pivotal molecule of the Wnt signaling pathway, which regulates the expression of Wnt target genes and the function of cancer cells in Wnt/β-catenin signaling pathway. These lncRNAs have been observed to play a pivotal role in Wnt/β-catenin signaling modulation in various cancers, including ESCC (Figure 3). Interestingly, Wnt/β-catenin pathway-related lncRNAs can directly or indirectly stimulate various subunits of the Wnt/β-catenin pathway, thereby activating or inhibiting the pathway’s activity. Therefore, understanding Wnt signaling in the context of lncRNAs may be a valuable strategy for managing ESCC.
For instance, HERES showed upregulation in 66 ESCC tissues compared to adjacent non-cancerous tissue samples [9]. lncRNA HERES augments five Wnt signaling regulated genes viz, calcium voltage-gated channel auxiliary subunit alpha2 delta 3 (CACNA2D3), secreted frizzled related protein 2 (SFRP2), calcium voltage-gated channel subunit alpha1 E (CACNA1E), CXXC finger protein 4 (CXXC4), and secreted frizzled related protein 2 (SFRP4) [9]. CACNA2D3 encodes a Wnt/Ca2+ complex subunit by decreasing intracellular calcium levels and the expression of Nemo-like kinase (NLK). Furthermore, downregulation of HERES increases NLK protein expression and reduces the β-catenin levels in KYSE-70 and HCE-7 ESCC cell lines (Figure 4). Additionally, SFRP2 encodes a member of the SFRP family that regulates the Wnt signaling pathway [9]. Furthermore, SFRP2 and CXXC4 act as negative regulators of the canonical Wnt signaling pathways. Thus, taken together, HERES promotes the ESCC pathogenesis by targeting both canonical and non-canonical pathways.
Similarly, lncRNA SNHG16 levels were significantly upregulated in ESCC tissues compared to normal tissue samples [10]. lncRNA SNHG16 promotes cell proliferation and invasion by modulating the targets of the Wnt/β-catenin pathway. In line with this, the TOPFLASH and FOPFLASH (TOP/FOP) luciferase reporter system showed that knockdown of SNHG16 expression inhibited the activation of the Wnt/β-catenin signaling in ESCC cell line EC-1 and Eca-109 [10]. Subsequently, the expression of regulatory molecules of Wnt signaling, such as c-Myc, β-catenin, and cyclin D1, was markedly reduced in the SNHG16 knockdown cell line, suggesting SNHG16 could be one of the markers to detect the alternation of the Wnt/β-catenin signaling pathway [10] (Figure 4). In addition to SNHG16, FEZF1-AS1 was significantly upregulated in 45 pairs of ESCC tissues and cells compared to adjacent non-neoplastic tissues and Het-1A cells, respectively, using real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) [38]. lncRNA FEZF1-AS1 promoted the migration and invasion of ESCC cells but did not affect the cell proliferation and cell cycle of ESCC cells. This phenotype is ascertained by the overexpression of Wnt regulated protein β-catenin in ESCC tissues [38].
In addition to the above, lncRNA UCA1 acts as a tumor suppressor with low expression in 106 ESCC tumor samples compared to adjacent normal tissues [11]. Through bioinformatics analysis, the authors suggested that UCA1 regulates Wnt signaling downstream molecules, such as catenin β1 (CTNNB1), dishevelled associated activator of morphogenesis 2 (DAAM2), dickkopf wnt signaling pathway inhibitor 1 (DKK1), and Wnt family member 2B (WNT2B) (Figure 4) [11]. Further overexpression of UCA1 leads to a reduction in the expression of the c-Myc target gene of Wnt signaling and thus regulates the cell cycle, suggesting that overexpression of UCA1 inhibits the activity of the Wnt/β-catenin signaling pathway in EC109 cells (Figure 4) [11]. Interestingly, the tumor suppressor MEG3 promotes tumor progression through targeting miR-4261, modulating the expression of DKK2, β-catenin, Bcl-2, and c-Myc, thus activating the Wnt/β-catenin signaling pathway [12] (Figure 4). As a result, MEG3 enhances the proliferation, migration, and invasion of ESCC cells [12]. Not only MEG3, but lncRNA LINC00675 was also downregulated in ESCC tissue samples compared to matched normal tissues [13]. Ectopic expression of LINC00675 reduced cell proliferation, migration, and invasion by decreasing cell cycle proteins, such as cyclin D1 and c-Myc, and epithelial-mesenchymal transition (EMT) regulated proteins, such as N-cadherin and vimentin, in EC9706 and EC-1 cells [13] (Figure 4). The above mechanism was occurred by targeting the β-catenin, a vital molecule of the Wnt/β-catenin signaling pathway, which suggests that enhancing the expression of LINC00675 inhibits the activities of the Wnt/β-catenin signaling pathway in ESCC cells [13]. lncRNA HOTAIR is frequently detected as oncogenic in ESCC patients’ tissues and is associated with the Wnt/β-catenin signaling pathway subunits. The upregulated profile of HOTAIR in ESCC tissues and cell lines targets an essential regulatory molecule of the Wnt pathway, WNT5B, and WIF-1 [14]. WIF-1 acts as a key inhibitor of the Wnt/β-catenin signaling pathway. It facilitates the degradation of β-catenin via APC⁄Axin1 destruction complex and by preventing the interaction of extracellular Wnt ligands with their receptors [14] (Figure 4). Mechanistically, epigenetic silencing of WIF-1 causes altered activation of the Wnt/β-catenin pathway in ESCC. Notably, WIF-1 downregulation is a prominent marker of tumor progression. qRT-PCR data revealed that the HOTAIR overexpression decreases the protein levels of WIF-1 and thus alters the Wnt/β-catenin signaling [14] (Figure 4). Moreover, HOTAIR exerts its function via a PRC2-dependent mechanism. The depletion of PRC2 enhances the levels of WIF-1 mRNA [14]. Additionally, an immunoblot assay revealed HOTAIR overexpressed ESCC cells possess a higher concentration of β-catenin expression in the nucleus, which indicates the activation of the canonical Wnt/β-catenin pathway [14]. Taken together, PRC2-associated HOTAIR inhibits the expression of WIF-1 by increasing trimethylation at H3K27 in the WIF-1 promoter region and then activates the Wnt/β-catenin signaling pathway and manifests the cell proliferation, migration, and invasion of ESCC cells (Figure 4).
At the same time, lncRNA TUG1 contributes to tumor progression by overexpressing their levels in 40 ESCC patients’ tissues and cell lines EC9706 and OE19tumor-adjacent corresponding tissues and HEEC cells, respectively. Upregulated TUG1 exerts its potential effects on ESCC manifestation through enhancing Wnt/β-catenin pathway-associated protein markers, such as Wnt1, c-Myc, cyclinD1, and β-catenin [15] (Figure 4). Furthermore, the administration of an activator (SKL2001) or inhibitor (XAV939) of Wnt/β-catenin signaling pathway to the TUG1-knockout ESCC cell lines EC9706 and OE19 revealed that SKL2001 promoted the expression of N-cadherin, Vimentin, and Snail and abolished the expression of E-cadherin, and thus enhanced the migration and invasion of the ESCC cells [15]. In addition to EMT, SKL2001 accelerated cell viability and cell apoptosis. Moreover, reverse effects were observed in XAV939 administered TUG1-knockout ESCC cell line, which suggests that the upregulation profile of TUG1 activates the Wnt/β-catenin signaling pathway [15], thus enhancing the proliferation, migration, and invasion and diminishing the apoptosis of ESCC cells. Another set of tumor suppressor lncRNA GASL1 manifests the ESCC pathogenesis by regulating the subunits of Wnt/β-catenin signaling pathway subunits. Downregulated GASL1 levels increase the protein expression of Wnt3a, β-catenin, and c-Myc and decrease the protein expression of DKK1 [16] (Figure 4), which suggests the activation of the canonical Wnt/β-catenin signaling pathway and ultimately the enhancement of ESCC cell proliferation, migration, and invasion.
Overall, we can say that the above-mentioned Wnt/β-catenin signaling pathway-related lncRNAs contribute to developing a therapeutic target in treating ESCC. Therefore, inactivation of the Wnt/β-catenin signaling pathway through altering the levels of lncRNAs could be effective in treating ESCC patients. Moreover, intrinsic and acquired resistance may limit the therapeutic efficacy of Wnt/β-catenin signaling pathway inhibitors

3. PI3K/Akt/mTOR Pathway-Related lncRNAs in ESCC

PI3K, Akt, and mTOR are the three significant nodes in the PI3K/Akt/mTOR pathway [39,40]. Tyrosine kinases and other receptor molecules, such as growth factors, hormones, and mitogen stimuli, activate the PI3K, Akt, and mTOR [39]. The PI3K/Akt/mTOR signaling pathway is one of the most critical intracellular pathways regulating cell growth, proliferation, metabolism, motility, survival, and apoptosis [41]. Therefore, aberrant activation of the PI3K/Akt pathway contributes to the development of tumor PI3K. As a result, PI3K promotes the survival and proliferation of tumor cells in many human cancers [42,43,44,45]), including ESCC (Figure 5). Recently, it has been reported that lncRNAs, such as HLA Complex P5 (HCP5), Papillary Thyroid Carcinoma Susceptibility Candidate 1 (PTCSC1), and LINC01014, and the PI3K/Akt/mTOR pathway are in tight conjunction during ESCC pathogenesis. This emphasizes the need to target this pathway with associated lncRNAs in treating ESCC patients.
lncRNA HCP5 was upregulated in ESCC tissues compared to control tissues [19]. Additionally, lncRNA HCP5 promotes proliferation, migration, invasion ability, and stemness characteristics of ESCC cells. Furthermore, it suppresses ESCC cell apoptosis by sponging miR-139-5p, thus upregulating phosphodiesterase 4A (PDE4A), a downstream target gene of the PI3K/Akt/mTOR pathway [19] (Figure 6). Additionally, lncRNA PTCSC1 expression was elevated in ESCC tissues and cells compared to adjacent non-cancerous tissues [20]. As a result, PTCSC1 promotes cell proliferation, migration, and invasion by activating the Akt p85 subunit of the PI3K/Akt/mTOR pathway [20]. In line with this, phosphorylated Akt levels also increased in PTCSC1 overexpressed KYSE30 cells [20], suggesting that PTCSC1 activated Akt signaling in ESCC cells (Figure 6). Last but not least, LINC01014 is associated with the PI3K/Akt/mTOR pathway in relation to gefitinib drug resistance in ESCC [18]. However, a detailed study in the context of this topic has not been elucidated in detail yet.

4. Crosstalk between Wnt/β-Catenin and PI3K/Akt/mTOR Pathway in ESCC

In the previous section, we discussed the regulatory role of Wnt/β-catenin and PI3K/Akt/mTOR related lncRNAs during ESCC pathogenesis. Studies suggested that Wnt/β-catenin and PI3K/Akt/mTOR pathways regulate themselves via a feedback mechanism, thus representing the resistance potential to chemotherapeutic drugs in clinical settings [46]. Therefore, understanding the crosstalk between the two mentioned pathways in ESCC is of immense importance. These pathways are finely connected at multiple levels during the homeostasis and pathological condition. For instance, glycogen synthase kinase 3β (GSK3β) is identified as a common key element in both the signaling pathways and thus regulates different cellular processes (Figure 7) [47]. During the activation of both signaling pathways, GSK3β activity becomes inhibited via various upstream events. Furthermore, a fraction of AXIN-bound GSK3β targets β-catenin degradation through the phosphorylation of β-catenin. At the same time, activated PI3K phosphorylates Akt at Ser9 residue, which further inhibits the GSK3β activity (Figure 7) [46].
At the same time, Akt hyperactivation and active canonical Wnt signaling pathways inhibit the GSK3β activity, resulting in the accumulation of β-catenin, thus representing the ESCC cell proliferation, migration, and invasion [46]. Recent studies showed that Wnt pathway activation leads to phosphorylation of S6K and 4E binding protein 1 (4E-BP1) and may affect the protein synthesis, thus turning on the mTOR complex 1 cascade (Figure 7) [48]. Notably, the activation of the Wnt-driven mTORC1 signaling could be independent of β-catenin and mediated by APC-AXIN-GSK3β axis and tuberous sclerosis 2 protein (TSC2) [48]. Moreover, it has recently been proposed that nuclear translocation of GSK3β is facilitated by rapamycin (mTOR) via increased phosphorylation of the forkhead box O1 (FOXO1) and general transcription factor IIF subunit 1 (GTF2F1), decreasing cell proliferation (Figure 7) [49,50].

5. LncRNAs Regulate the Efficacy of Chemotherapeutic Drugs in ESCC

Worldwide, esophageal tumors develop resistance to chemotherapy during treatment, leading to multiple fatal complications implicated in treatment failure and tumor relapse [51]. Generally, chemotherapy represents the frontline treatment for both early and advanced staged tumors. However, chemotherapeutic drug resistance limits the efficacy of conventional chemotherapeutics and the United States Food and Drug Administration-approved biological agents, such as keytruda (pembrolizumab) nivolumab, opdivo (nivolumab), and pembrolizumab. Notably, multidrug resistance in ESCC patients can be due to the higher expression of transporters that eject drugs from cells [52]. It can be intrinsic (tumor insensitive to therapeutic agents before treatment) or extrinsic (tumor becomes resistant during the treatment) [52]. However, the mechanisms associated with drug resistance in ESCC patients, including resistance to apoptosis induced by drugs, decreased intracellular accumulation of therapeutics, increased repair of damaged DNA, and induction of mechanisms capable of drug detoxification, are still in their infancy [53]. Furthermore, due to the limitations associated with the drug potency in ESCC patients, scientists aim to develop/identify new biomarkers to assess and predict patients’ responses against the chemotherapeutic drugs. Recent studies showed that lncRNAs play important roles in regulating the chemo- and radio-resistance of ESCC by controlling several signaling pathways and modulating the mechanisms associated with the cell cycle, proliferation, apoptosis, and DNA damage repair [3].
With a view to the challenges faced by the ESCC patients during chemotherapy, we tried our best to sort the relevant drug molecules, which need to be studied in the context of lncRNA and ESCC for disease management. Firstly, we searched in PubMed all recent studies (published from 2012 onwards) investigating lncRNAs in chemoresistance during ESCC therapy. The primary screening result was manually curated to avoid and remove articles with generic statements and not direct links between lncRNAs and drugs in the context of ESCC. Thus, we have chosen the research paper showing the direct association of lncRNAs with chemoresistance in ESCC treatment for this section. Furthermore, we searched the reported drugs in the NoncoRNA database for collecting all information about lncRNA-target gene drugs used for ESCC therapy in association with lncRNAs.
Mounting evidence suggests that a pool of lncRNAs (LOC285194/ (tumor suppressor candidate 7 (TUSC7), taurine upregulated 1 (TUG1), AFAP1 antisense RNA 1 (AFAP1-AS1), prostate androgen-regulated transcript 1 (PART1), colon cancer-associated transcript 1 (CCAT1), long intergenic non-protein coding RNA 1419 (LINC01419), long intergenic non-protein coding RNA 337 (LINC00337), long intergenic non-protein coding RNA 1014 (Linc01014), MACC1 antisense RNA 1 (MACC1-AS1), FOXD2 adjacent opposite strand RNA 1 (FOXD2-AS1)) are involved in ESCC chemotherapy resistance.
It was observed that lncRNA LOC285194 or TUSC7 downregulated in ESCC tissues and cell lines compared to adjacent normal tissues [54]. Moreover, downregulated LOC285194 hinders the potential of cisplatin (20 mg/m2/day, for five days in combination with radiotherapy (40 Gy in 20 fractions of 2 Gy each, with five fractions per week for four weeks) (Table 1). Kaplan–Meier survival analysis revealed that low expressed LOC285194ESCC patients group showed decreased disease-free survival (DFS) and overall survival compared to high expressed LOC285194 group. The complete pathological response (pathCR) rate was 57% in the LOC285194-high group, while only 15% in the LOC285194-low group suggested that patients with low expression LOC285194 showed resistance to chemoradiotherapy treatment (Table 1) [54]. At the same time, lncRNA TUSC7 was downregulated in the chemotherapy resistance patients’ group compared to the chemotherapy responsive patients’ group, and thus the survival rate of the ESCC patients became very poor. The low expression of TUSC7 resists the potency of cisplatin (1, 2, 4, 8, and 16 μM for 48 h) or 5-FU (1, 4, 16, 32, and 64 μM for 48 h) in ESCC cell lines EC9706 and KYSE30 [55] (Table 1). In addition to that, TUG1 is significantly upregulated in ESCC tissues compared to paired adjacent normal tissues [56]. Furthermore, TUG1 expression is higher in TE-1 derived cisplatin (DDP)-resistant (TE-1/DDP) cells (1 μg/mL for 48 h) compared to TE-1 cells [57], suggesting that high TUG1 expression was significantly implicated with chemotherapy resistance and inversely correlated with overall survival of ESCC patients [56] (Table 1). Moreover, lncRNA AFAP1-AS1 showed upregulation (~14-fold) in paired cisplatin-resistant (KYSE30-R) and parental ESCC cell lines (KYSE30) [58]. Moreover, an upregulation profile of AFAP1-AS1 was observed in 162 pretreatment biopsy specimens of ESCC who underwent definitive chemoradiotherapy (dCRT). Notably, upregulated AFAP1-AS1 undergoes cross-resistance of cisplatin (0.3125, 0.625, 1.25, 2.5 5, 10, 25, and 50 μM for 24 h on days 1–4) along with two combinations of anticancer drugs viz, 5-fluorouracil (5-FU) (2, 4, 8, 16, 32, 64,128, and 256 μM for 24 h on days 1–4) and paclitaxel (0.03125, 0.0652. 0.125, 0.25, 0.5, 1, 2, 4, and 8 µM for 24 h on days 1–4), when administered to ESCC patients [58] (Table 1). Chemotherapeutics treated patients represent low overall survival and progression-free survival. Moreover, the pathological complete response rate was 19.8%, the partial response rate was 40.7%, no response rate was 37.7%, and progressive disease response was 1.8%, suggesting the strong hindrance property of lncRNA AFAP1-AS1 in conferring the chemotherapy in ESCC management [58] (Table 1). In line with this, high expression levels of AFAP1-AS1 serve as a potential biomarker to predict tumor response and survival.
Furthermore, higher expression of CCAT1 slightly decreases the viability of cisplatin-resistant ESCC cells (0.1, 0.2, 0.5, 1, 2, and 5 µg/mL for 48 h) compared with cisplatin-sensitive ESCC cells [59]. However, the half-maximal inhibitory concentration (IC50) value of cisplatin treatment (Table 1) showed that CCAT1 positively correlates with cisplatin resistance in ESCC cells. Importantly, LINC01419 overexpression contributes to the diminished effect of 5-FU (10 µg/mL for 48 h) in ESCC cells by promoting the methylation of the promoter region of the glutathione S-transferase Pi 1 (GSTP1) gene [60] (Table 1). In addition to the lncRNAs mentioned above, LINC00337 was overexpressed in ESCC patients’ tissues and cell lines, which hindered the effects of cisplatin dose (0.5, 1, 2, and 3 μg/mL for 48 h) via the upregulation of TPX2 by recruiting the E2F4 transcription factor [51] (Table 1). Furthermore, two lncRNAs, MACC1-AS1 and FOXD2-AS1, were upregulated in ESCC cells and tumor tissues [61,62]. As a result, both the lncRNAs hinder cisplatin’s efficacy (20, 40, 60, 80, and 100 µM) through the overexpression of NSD2 mRNA and protein in ESCC tissues compared to adjacent non-cancerous tissues [61] (Table 1). Moreover, FOXD2-AS1 increases the cisplatin resistance (6.25, 12.5, 25, 50, and 100 µg/mL) by promoting the Akt/mTOR axis stimulation in ESCC cells [62] (Table 1).
Besides resistance to cisplatin, 5-FU, and paclitaxel, altered expression of lncRNAs resist the potential of gefitinib in ESCC treatment. For example, lncRNA PART1 upregulated in gefitinib-resistant ESCC cells compared to parental ESCC cells. The resistance to gefitinib (0.01, 0.1, 1, 2, 3, 8, and 10 µM for 48 h) (Table 1) by lncRNA PART1 is facilitated by the transportation of extracellular PART1 into exosomes and incorporation into sensitive cells, which ultimately inhibits apoptotic proteins and cell apoptosis by regulating the Bcl-2/Bax pathway [63]. Moreover, upregulated LINC01014 confers gefitinib resistance (1, 10, 20, and 30 µM for 48 h) (Table 1) in ESCC cells by inhibiting ESCC cells’ apoptosis via PI3K/Akt/mTOR signaling pathway [18].

6. Conclusions and Future Aspects

In this review, we highlight the immense potential of oncogenic and tumor suppressive lncRNAs in regulating cancer-associated signaling pathways and their implication in drug resistance in ESCC patients. As mentioned in the previous sections, the Wnt/β-catenin and PI3K/Akt/mTOR pathway comprises multiple downstream signaling proteins, such as β-catenin, GSK3-β, Akt, PI3K, and mTORC1 complex, whose activation in association with dysregulated lncRNAs can manifest several hallmarks of cancer, including uncontrolled cell growth, inhibition of apoptosis, proliferation, increased metastasis, and invasion. We found that upregulated expression levels of HERES, TUG1, HCP5, and PTCSC1 and downregulated expression levels of UCA1 could be best suited for therapeutic application in clinical settings. These lncRNAs significantly target the key downstream molecules of cancer-related pathways, namely the Wnt/β-catenin and PI3K/Akt/mTOR pathway. Based on the ESCC cohort size and the detailed mechanism, UCA1, HCP5, and PTCSC1 possess great potential as a therapeutic target for ESCC in association with the Wnt/ β-catenin and PI3K/Akt/mTOR pathways, which signifies the clinical potential of lncRNAs for the treatment of ESCC patients. Thus, targeting lncRNAs and their associated pathways, i.e., Wnt/β-catenin and PI3K/Akt/mTOR, may provide novel approaches in the treatment and better management of ESCC patients.
We also presented the potential of lncRNAs as an effective regulator of chemotherapeutic drugs, such as paclitaxel, 5-FU, gefitinib, and cisplatin. In our opinion, upregulated levels of AFAP1-AS1 could be a selective prognostic and therapeutic marker for the ESCC patients treated with cisplatin, 5-FU, and paclitaxel. Additionally, we observed that AFAP1-AS1 lowers the efficacy of the maximum dose of cisplatin, 5-FU, and paclitaxel drugs at 24 h in 162 ESCC patients [58]. Therefore, the modulation of AFAP-AS1 expression levels is of utmost importance in the management of ESCC. Furthermore, it has been shown that LINC01014 resists the efficacy of gefitinib (30 µM at 48 h), which suggests that LINC01014 requires critical attention concerning prognostic and therapeutic aspects in the clinical settings [18]. The diverse functional repertoire of lncRNAs reveals various opportunities for their therapeutic targeting, including inhibition at transcriptional and post-transcriptional levels, steric hindrance on protein interaction and formation of secondary structures, and the modulation of genomic loci or lncRNA expression patterns using clustered regularly interspaced short palindromic repeats (CRISPR)-associated proteins (Cas) technology. However, the application of RNA based therapeutics in clinical settings has been hampered by the lack of specificity, delivery method, and tolerability.
Besides, many clinical trials have shown the development of RNA therapeutics, such as miRNA mimics or antimiRs, and several are in phase II or III. Still, no lncRNA-based therapeutic agent has entered the clinical setting. In future, we believe that lncRNAs and their related signaling pathways can be targeted using secondary plant metabolites in combination with chemotherapeutic drugs for the betterment of cancer treatment, as supported by recently published studies [64,65,66]. Experimental findings have indicated that bioactive phytochemicals, such as anacardic acid, baicalein, berberis, bharangin, genistein, calycosin, and silibinin, could be utilized to target the expression of lncRNAs in various cancers, including ESCC [64,65,66]. Additionally, it is likely that these bioactive phytochemicals may also modulate a diverse range of cell signaling pathways in cancer cells. Furthermore, novel formulations, such as nano-drug delivery systems, can be utilized to enhance the bioavailability of phytochemicals alone or in combination with chemotherapeutic drugs. In addition, synthetic chemistry tools may also be implemented to design new derivatives of existing drugs to analyze their potential to modulate lncRNAs in ESCC. Synergistic approaches may further enhance the activity of chemopreventive agents to optimize the levels of dysregulated lncRNAs.
Overall, our study provides comprehensive knowledge of the lncRNA regulated Wnt/β-catenin and PI3K/Akt signaling pathways and highlights the potential of lncRNAs hindering the therapeutic efficacy in ESCC. The techniques mentioned above can be employed to target desired lncRNAs clinically. However, future studies are required to translate the findings into clinical settings.

Supplementary Materials

Author Contributions

U.S. and A.J. conceived the original idea. U.S. wrote the manuscript and prepared the tables and figures. M.M. and H.S.T. wrote the conclusion section. T.S.B. formatted the references as per journal style. A.J., U.S., H.P., M.J., T.K. and A.B. contributed to the final editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Department of Science and Technology of India supported this work through the Indo-Russia grant (INT/RUS/RFBR/P-311) to AJ and DST-INSPIRE fellowship (IF180680) to U.S.

Conflicts of Interest

The authors declare no competing or conflicting interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, L.; Li, P.; Liu, E.; Xing, C.; Zhu, D.; Zhang, J.; Wang, W.; Jiang, G. Prognostic value of a five-lncRNA signature in esophageal squamous cell carcinoma. Cancer Cell Int. 2020, 20, 386. [Google Scholar] [CrossRef] [PubMed]
  3. Sharma, U.; Barwal, T.; Acharya, V.; Singh, K.; Rana, M.; Singh, S.; Prakash, H.; Bishayee, A.; Jain, A. Long Non-Coding RNAs as Strategic Molecules to Augment the Radiation Therapy in Esophageal Squamous Cell Carcinoma. Int. J. Mol. Sci. 2020, 21, 6787. [Google Scholar] [CrossRef] [PubMed]
  4. Si, W.; Shen, J.; Zheng, H.; Fan, W. The role and mechanisms of action of microRNAs in cancer drug resistance. Clin. Epigenet. 2019, 11, 25. [Google Scholar] [CrossRef]
  5. Wang, W.-T.; Han, C.; Sun, Y.-M.; Chen, T.-Q.; Chen, Y.-Q. Noncoding RNAs in cancer therapy resistance and targeted drug development. J. Hematol. Oncol. 2019, 12, 1–15. [Google Scholar] [CrossRef]
  6. Tamang, S.; Acharya, V.; Roy, D.; Sharma, R.; Aryaa, A.; Sharma, U.; Khandelwal, A.; Prakash, H.; Vasquez, K.M.; Jain, A. SNHG12: An LncRNA as a Potential Therapeutic Target and Biomarker for Human Cancer. Front. Oncol. 2019, 9, 901. [Google Scholar] [CrossRef]
  7. Farooqi, A.A.; Nayyab, S.; Martinelli, C.; Berardi, R.; Katifelis, H.; Gazouli, M.; Cho, W.C. Regulation of Hippo, TGFβ/SMAD, Wnt/β-Catenin, JAK/STAT, and NOTCH by Long Non-Coding RNAs in Pancreatic Cancer. Front. Oncol. 2021, 11, 657965. [Google Scholar] [CrossRef]
  8. Sharma, U.; Barwal, T.S.; Acharya, V.; Tamang, S.; Vasquez, K.M.; Jain, A. Cancer Susceptibility Candidate 9 (CASC9): A Novel Targetable Long Noncoding RNA in Cancer Treatment. Transl. Oncol. 2020, 13, 100774. [Google Scholar] [CrossRef]
  9. You, B.-H.; Yoon, J.-H.; Kang, H.; Lee, E.K.; Lee, S.K.; Nam, J.-W. HERES, a lncRNA that regulates canonical and noncanonical Wnt signaling pathways via interaction with EZH2. Proc. Natl. Acad. Sci. USA 2019, 116, 24620–24629. [Google Scholar] [CrossRef] [Green Version]
  10. Han, G.H.; Lu, K.J.; Wang, P.; Ye, J.; Ye, Y.Y.; Huang, J.X. LncRNA SNHG16 predicts poor prognosis in ESCC and promotes cell proliferation and invasion by regulating Wnt/beta-catenin signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3795–3803. [Google Scholar]
  11. Wang, X.; Gao, Z.; Liao, J.; Shang, M.; Li, X.; Yin, L.; Pu, Y.; Liu, R. lncRNA UCA1 inhibits esophageal squamous-cell carcinoma growth by regulating the Wnt signaling pathway. J. Toxicol. Environ. Health Part A 2016, 79, 407–418. [Google Scholar] [CrossRef] [PubMed]
  12. Ma, J.; Li, T.-F.; Han, X.-W.; Yuan, H.-F. Downregulated MEG3 contributes to tumour progression and poor prognosis in oesophagal squamous cell carcinoma by interacting with miR-4261, downregulating DKK2 and activating the Wnt/β-catenin signalling. Artif. Cells Nanomed. Biotechnol. 2019, 47, 1513–1523. [Google Scholar] [CrossRef] [PubMed]
  13. Zhong, Y.B.; Shan, A.J.; Lv, W.; Wang, J.; Xu, J.Z. Long non-coding RNA LINC00675 inhibits tumorigenesis and EMT via repressing Wnt/beta-catenin signaling in esophageal squamous cell carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 8288–8297. [Google Scholar] [PubMed]
  14. Ge, X.-S.; Ma, H.-J.; Zheng, X.-H.; Ruan, H.-L.; Liao, X.; Xue, W.-Q.; Chen, Y.-B.; Zhang, Y.; Jia, W.-H. HOTAIR, a prognostic factor in esophageal squamous cell carcinoma, inhibits WIF-1 expression and activates Wnt pathway. Cancer Sci. 2013, 104, 1675–1682. [Google Scholar] [CrossRef]
  15. Tang, Y.; Yang, P.; Zhu, Y.; Su, Y. LncRNA TUG1 contributes to ESCC progression via regulating miR-148a-3p/MCL-1/Wnt/beta-catenin axis in vitro. Thorac. Cancer 2020, 11, 82–94. [Google Scholar] [CrossRef]
  16. Ren, Y.; Guo, T.; Xu, J.; Liu, Y.; Huang, J. The novel target of esophageal squamous cell carcinoma: lncRNA GASL1 regulates cell migration, invasion and cell cycle stagnation by inactivating the Wnt3a/β-catenin signaling. Pathol.-Res. Pract. 2020, 217, 153289. [Google Scholar] [CrossRef]
  17. Zhong, B.; Wang, Q.; He, J.; Xiong, Y.; Cao, J. LncRNA LOC285194 modulates gastric carcinoma progression through activating Wnt/beta-catenin signaling pathway. Cancer Med. 2020, 9, 2181–2189. [Google Scholar] [CrossRef]
  18. Fu, X.; Cui, G.; Liu, S.; Zhao, S. Linc01014 regulates gefitinib resistance in oesophagus cancer via EGFR-PI3K-AKT-mTOR signalling pathway. J. Cell Mol. Med. 2020, 24, 1670–1675. [Google Scholar] [CrossRef] [Green Version]
  19. Xu, J.; Ma, J.; Guan, B.; Li, J.; Wang, Y.; Hu, S. LncRNA HCP5 promotes malignant cell behaviors in esophageal squamous cell carcinoma via the PI3K/AKT/mTOR signaling. Cell Cycle 2021, 20, 1374–1388. [Google Scholar] [CrossRef]
  20. Liu, T.; Liang, X.; Yang, S.; Sun, Y. Long noncoding RNA PTCSC1 drives esophageal squamous cell carcinoma progression through activating Akt signaling. Exp. Mol. Pathol. 2020, 117, 104543. [Google Scholar] [CrossRef]
  21. Fang, Y.; Zhang, S.; Yin, J.; Shen, Y.-X.; Wang, H.; Chen, X.-S.; Tang, H. LINC01535 promotes proliferation and inhibits apoptosis in esophageal squamous cell cancer by activating the JAK/STAT3 pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 3694–3700. [Google Scholar] [PubMed]
  22. Cao, Y.; Wang, X.; Chen, L. [Knockdown of Fez family zinc finger protein 1 antisense ribonucleic acid 1 (FEZF1-AS1) inhibits invasion and migration of esophageal squamous cell carcinoma cells by blocking JAK2/STAT3 pathway]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2020, 36, 317–324. [Google Scholar] [PubMed]
  23. Tang, J.; Xu, H.; Liu, Q.; Zheng, J.; Pan, C.; Li, Z.; Wen, W.; Wang, J.; Zhu, Q.; Wang, Z.; et al. LncRNA LOC146880 promotes esophageal squamous cell carcinoma progression via miR-328-5p/FSCN1/MAPK axis. Aging 2021, 13, 14198–14218. [Google Scholar] [CrossRef]
  24. Sharma, U.; Barwal, T.S.; Khandelwal, A.; Rana, M.K.; Rana, A.P.S.; Singh, K.; Jain, A. Circulating Long Non-Coding RNAs LINC00324 and LOC100507053 as Potential Liquid Biopsy Markers for Esophageal Squamous Cell Carcinoma: A Pilot Study. Front. Oncol. 2022, 12. [Google Scholar] [CrossRef] [PubMed]
  25. Lu, Z.; Chen, Z.; Li, Y.; Wang, J.; Zhang, Z.; Che, Y.; Huang, J.; Sun, S.; Mao, S.; Lei, Y.; et al. TGF-beta-induced NKILA inhibits ESCC cell migration and invasion through NF-kappaB/MMP14 signaling. J. Mol. Med. 2018, 96, 301–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Yang, L.; Sun, K.; Chu, J.; Qu, Y.; Zhao, X.; Yin, H.; Ming, L.; Wan, J.; He, F. Long non-coding RNA FTH1P3 regulated metastasis and invasion of esophageal squamous cell carcinoma through SP1/NF-kB pathway. Biomed. Pharmacother. 2018, 106, 1570–1577. [Google Scholar] [CrossRef]
  27. Zhang, Y.; Jin, X.; Wang, Z.; Zhang, X.; Liu, S.; Liu, G. Downregulation of SNHG1 suppresses cell proliferation and invasion by regulating Notch signaling pathway in esophageal squamous cell cancer. Cancer Biomark. 2017, 21, 89–96. [Google Scholar] [CrossRef]
  28. Chen, M.; Xia, Z.; Chen, C.; Hu, W.; Yuan, Y. LncRNA MALAT1 promotes epithelial-to-mesenchymal transition of esophageal cancer through Ezh2-Notch1 signaling pathway. Anti-Cancer Drugs 2018, 29, 767–773. [Google Scholar] [CrossRef]
  29. Aggarwal, V.; Tuli, H.S.; Varol, M.; Tuorkey, M.; Sak, K.; Parashar, N.C.; Barwal, T.S.; Sharma, U.; Iqubal, A.; Parashar, G.; et al. NOTCH signaling: Journey of an evolutionarily conserved pathway in driving tumor progression and its modulation as a therapeutic target. Crit. Rev. Oncol. 2021, 164, 103403. [Google Scholar] [CrossRef]
  30. Zhang, X.; Xie, K.; Zhou, H.; Wu, Y.; Li, C.; Liu, Y.; Liu, Z.; Xu, Q.; Liu, S.; Xiao, D.; et al. Role of non-coding RNAs and RNA modifiers in cancer therapy resistance. Mol. Cancer 2020, 19, 1–26. [Google Scholar] [CrossRef] [Green Version]
  31. Yu, X.; Huang, M.; Yang, G. Long non-coding RNA BANCR promotes proliferation, invasion and migration in esophageal squamous cell carcinoma cells via the Raf/MEK/ERK signaling pathway. Mol. Med. Rep. 2021, 23, 1–9. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, Y.; Zheng, A.; Xu, R.; Zhou, F.; Hao, A.; Yang, H.; Yang, P. NR2F1-induced NR2F1-AS1 promotes esophageal squamous cell carcinoma progression via activating Hedgehog signaling pathway. Biochem. Biophys. Res. Commun. 2019, 519, 497–504. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, Y.; Miao, Y.; Shang, M.; Liu, M.; Liu, R.; Pan, E.; Pu, Y.; Yin, L. LincRNA-p21 leads to G1 arrest by p53 pathway in esophageal squamous cell carcinoma. Cancer Manag. Res. 2019, 11, 6201–6214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Chen, D.; Zhang, Z.; Mao, C.; Zhou, Y.; Yu, L.; Yin, Y.; Wu, S.; Mou, X.; Zhu, Y. ANRIL inhibits p15(INK4b) through the TGFbeta1 signaling pathway in human esophageal squamous cell carcinoma. Cell Immunol. 2014, 289, 91–96. [Google Scholar] [CrossRef] [PubMed]
  35. Tu, C.; Yang, K.; Wan, L.; He, J.; Qi, L.; Wang, W.; Lu, Q.; Li, Z. The crosstalk between lncRNAs and the Hippo signalling pathway in cancer progression. Cell Prolif. 2020, 53. [Google Scholar] [CrossRef]
  36. Fu, P.-F.; Zheng, X.; Fan, X.; Lin, A.-F. Role of cytoplasmic lncRNAs in regulating cancer signaling pathways. J. Zhejiang Univ. Sci. B 2019, 20, 1–8. [Google Scholar] [CrossRef]
  37. Pai, S.G.; Carneiro, B.A.; Mota, J.M.; Costa, R.; Leite, C.A.; Barroso-Sousa, R.; Kaplan, J.B.; Chae, Y.K.; Giles, F.J. Wnt/beta-catenin pathway: Modulating anticancer immune response. J. Hematol. Oncol. 2017, 10, 1–12. [Google Scholar] [CrossRef] [Green Version]
  38. Yang, L.; Ye, Y.; Chu, J.; Jia, J.; Qu, Y.; Sun, T.; Yin, H.; Ming, L.; Wan, J.; He, F. Long noncoding RNA FEZF1-AS1 promotes the motility of esophageal squamous cell carcinoma through Wnt/beta-catenin pathway. Cancer Manag. Res. 2019, 11, 4425–4435. [Google Scholar] [CrossRef] [Green Version]
  39. Ruggero, D.; Sonenberg, N. The Akt of translational control. Oncogene 2005, 24, 7426–7434. [Google Scholar] [CrossRef] [Green Version]
  40. Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: Mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 1–28. [Google Scholar] [CrossRef] [Green Version]
  41. Porta, C.; Paglino, C.; Mosca, A. Targeting PI3K/Akt/mTOR Signaling in Cancer. Front. Oncol. 2014, 4, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Myers, A.P.; Cantley, L.C. Targeting a Common Collaborator in Cancer Development. Sci. Transl. Med. 2010, 2, 48ps45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Morgan, T.M.; Koreckij, T.D.; Corey, E. Targeted Therapy for Advanced Prostate Cancer: Inhibition of the PI3K/Akt/mTOR Pathway. Curr. Cancer Drug Targets 2009, 9, 237–249. [Google Scholar] [CrossRef] [Green Version]
  45. Sarris, E.G.; Saif, M.W.; Syrigos, K.N. The Biological Role of PI3K Pathway in Lung Cancer. Pharmaceuticals 2012, 5, 1236–1264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Prossomariti, A.; Piazzi, G.; Alquati, C.; Ricciardiello, L. Are Wnt/beta-Catenin and PI3K/AKT/mTORC1 Distinct Pathways in Colorectal Cancer? Cell Mol. Gastroenterol. Hepatol. 2020, 10, 491–506. [Google Scholar] [CrossRef]
  47. Kaidanovich-Beilin, O.; Woodgett, J.R. GSK-3: Functional Insights from Cell Biology and Animal Models. Front. Mol. Neurosci. 2011, 4, 40. [Google Scholar] [CrossRef] [Green Version]
  48. Inoki, K.; Ouyang, H.; Zhu, T.; Lindvall, C.; Wang, Y.; Zhang, X.; Yang, Q.; Bennett, C.; Harada, Y.; Stankunas, K.; et al. TSC2 Integrates Wnt and Energy Signals via a Coordinated Phosphorylation by AMPK and GSK3 to Regulate Cell Growth. Cell 2006, 126, 955–968. [Google Scholar] [CrossRef] [Green Version]
  49. He, L.; Gomes, A.P.; Wang, X.; Yoon, S.O.; Lee, G.; Nagiec, M.J.; Cho, S.; Chavez, A.; Islam, T.; Yu, Y.; et al. mTORC1 Promotes Metabolic Reprogramming by the Suppression of GSK3-Dependent Foxk1 Phosphorylation. Mol. Cell 2018, 70, 949–960.e4. [Google Scholar] [CrossRef] [Green Version]
  50. He, L.; Fei, D.L.; Nagiec, M.J.; Mutvei, A.P.; Lamprakis, A.; Kim, B.Y.; Blenis, J. Regulation of GSK3 cellular location by FRAT modulates mTORC1-dependent cell growth and sensitivity to rapamycin. Proc. Natl. Acad. Sci. USA 2019, 116, 19523–19529. [Google Scholar] [CrossRef] [Green Version]
  51. Yang, C.; Shen, S.; Zheng, X.; Ye, K.; Ge, H.; Sun, Y.; Lu, Y. Long non-coding RNA LINC00337 induces autophagy and chemoresistance to cisplatin in esophageal squamous cell carcinoma cells via upregulation of TPX2 by recruiting E2F4. FASEB J. 2020, 34, 6055–6069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Corrà, F.; Agnoletto, C.; Minotti, L.; Baldassari, F.; Volinia, S. The Network of Non-coding RNAs in Cancer Drug Resistance. Front. Oncol. 2018, 8, 327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tong, Y.-S.; Zhou, X.-L.; Wang, X.-W.; Wu, Q.-Q.; Yang, T.-X.; Lv, J.; Yang, J.-S.; Zhu, B.; Cao, X.-F. Association of decreased expression of long non-coding RNA LOC285194 with chemoradiotherapy resistance and poor prognosis in esophageal squamous cell carcinoma. J. Transl. Med. 2014, 12, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Chang, Z.W.; Jia, Y.X.; Zhang, W.J.; Song, L.J.; Gao, M.; Li, M.J.; Zhao, R.H.; Li, J.; Zhong, Y.L.; Sun, Q.Z.; et al. LncRNA-TUSC7/miR-224 affected chemotherapy resistance of esophageal squamous cell carcinoma by competitively regulating DESC1. J. Exp. Clin. Cancer Res. 2018, 37, 56. [Google Scholar] [CrossRef] [Green Version]
  56. Jiang, L.; Wang, W.; Li, G.; Sun, C.; Ren, Z.; Sheng, H.; Gao, H.; Wang, C.; Yu, H. High TUG1 expression is associated with chemotherapy resistance and poor prognosis in esophageal squamous cell carcinoma. Cancer Chemother. Pharmacol. 2016, 78, 333–339. [Google Scholar] [CrossRef]
  57. Zhang, Z.; Xiong, R.; Li, C.; Xu, M.; Guo, M. LncRNA TUG1 promotes cisplatin resistance in esophageal squamous cell carcinoma cells by regulating Nrf2. Acta Biochim. Biophys. Sin. 2019, 51, 826–833. [Google Scholar] [CrossRef]
  58. Zhou, X.; Wang, W.; Zhu, W.; Yu, C.; Tao, G.; Wu, Q.; Song, Y.; Pan, P.; Tong, Y. High expression of long non-coding RNA AFAP1-AS1 predicts chemoradioresistance and poor prognosis in patients with esophageal squamous cell carcinoma treated with definitive chemoradiotherapy. Mol. Carcinog. 2016, 55, 2095–2105. [Google Scholar] [CrossRef] [Green Version]
  59. Hu, M.; Zhang, Q.; Tian, X.H.; Wang, J.L.; Niu, Y.X.; Li, G. lncRNA CCAT1 is a biomarker for the proliferation and drug resistance of esophageal cancer via the miR-143/PLK1/BUBR1 axis. Mol. Carcinog. 2019, 58, 2207–2217. [Google Scholar] [CrossRef]
  60. Chen, J.-L.; Lin, Z.-X.; Qin, Y.-S.; She, Y.-Q.; Chen, Y.; Chen, C.; Qiu, G.-D.; Zheng, J.-T.; Chen, Z.-L.; Zhang, S.-Y. Overexpression of long noncoding RNA LINC01419 in esophageal squamous cell carcinoma and its relation to the sensitivity to 5-fluorouracil by mediating GSTP1 methylation. Ther. Adv. Med Oncol. 2019, 11, 1758835919838958. [Google Scholar] [CrossRef] [Green Version]
  61. Xue, W.; Shen, Z.; Li, L.; Zheng, Y.; Yan, D.; Kan, Q.; Zhao, J. Long non-coding RNAs MACC1-AS1 and FOXD2-AS1 mediate NSD2-induced cisplatin resistance in esophageal squamous cell carcinoma. Mol. Ther.-Nucleic Acids 2021, 23, 592–602. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, H.; Zhang, J.; Luo, X.; Zeng, M.; Xu, L.; Zhang, Q.; Liu, H.; Guo, J.; Xu, L. Overexpression of the Long Noncoding RNA FOXD2-AS1 Promotes Cisplatin Resistance in Esophageal Squamous Cell Carcinoma Through the miR-195/Akt/mTOR Axis. Oncol. Res. 2020, 28, 65–73. [Google Scholar] [CrossRef] [PubMed]
  63. Kang, M.; Ren, M.; Li, Y.; Fu, Y.; Deng, M.; Li, C. Exosome-mediated transfer of lncRNA PART1 induces gefitinib resistance in esophageal squamous cell carcinoma via functioning as a competing endogenous RNA. J. Exp. Clin. Cancer Res. 2018, 37, 171. [Google Scholar] [CrossRef] [PubMed]
  64. Kalhori, M.; Khodayari, H.; Khodayari, S.; Vesovic, M.; Jackson, G.; Farzaei, M.; Bishayee, A. Regulation of Long Non-Coding RNAs by Plant Secondary Metabolites: A Novel Anticancer Therapeutic Approach. Cancers 2021, 13, 1274. [Google Scholar] [CrossRef]
  65. Tuli, H.S.; Mittal, S.; Aggarwal, D.; Parashar, G.; Parashar, N.C.; Upadhyay, S.K.; Barwal, T.S.; Jain, A.; Kaur, G.; Savla, R.; et al. Path of Silibinin from diet to medicine: A dietary polyphenolic flavonoid having potential anti-cancer therapeutic significance. Semin. Cancer Biol. 2020, 73, 196–218. [Google Scholar] [CrossRef]
  66. Tuli, H.S.; Tuorkey, M.J.; Thakral, F.; Sak, K.; Kumar, M.; Sharma, A.K.; Sharma, U.; Jain, A.; Aggarwal, V.; Bishayee, A. Molecular Mechanisms of Action of Genistein in Cancer: Recent Advances. Front. Pharmacol. 2019, 10, 1336. [Google Scholar] [CrossRef] [Green Version]
  67. Chen, F.-J.; Sun, M.; Li, S.-Q.; Wu, Q.-Q.; Ji, L.; Liu, Z.-L.; Zhou, G.-Z.; Cao, G.; Jin, L.; Xie, H.-W.; et al. Upregulation of the long non-coding rna hotair promotes esophageal squamous cell carcinoma metastasis and poor prognosis. Mol. Carcinog. 2012, 52, 908–915. [Google Scholar] [CrossRef]
  68. Wu, H.; Zheng, J.; Deng, J.; Hu, M.; You, Y.; Li, N.; Li, W.; Lu, J.; Zhou, Y. A genetic polymorphism in lincRNA-uc003opf.1 is associated with susceptibility to esophageal squamous cell carcinoma in Chinese populations. Carcinogenesis 2013, 34, 2908–2917. [Google Scholar] [CrossRef] [Green Version]
  69. Pan, F.; Yao, J.; Chen, Y.; Zhou, C.; Geng, P.; Mao, H.; Fang, X. A novel long non-coding RNA FOXCUT and mRNA FOXC1 pair promote progression and predict poor prognosis in esophageal squamous cell carcinoma. Int. J. Clin. Exp. Pathol. 2014, 7, 2838–2849. [Google Scholar]
  70. Gao, T.; He, B.; Pan, Y.; Gu, L.; Chen, L.; Nie, Z.; Xu, Y.; Li, R.; Wang, S. H19 DMR methylation correlates to the progression of esophageal squamous cell carcinoma through IGF2 imprinting pathway. Clin. Transl. Oncol. 2013, 16, 410–417. [Google Scholar] [CrossRef]
  71. Li, W.; Zheng, J.; Deng, J.; You, Y.; Wu, H.; Li, N.; Lu, J.; Zhou, Y. Increased Levels of the Long Intergenic Non–Protein Coding RNA POU3F3 Promote DNA Methylation in Esophageal Squamous Cell Carcinoma Cells. Gastroenterology 2014, 146, 1714–1726.e5. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, C.-M.; Wu, Q.-Q.; Li, S.-Q.; Chen, F.-J.; Tuo, L.; Xie, H.-W.; Tong, Y.-S.; Ji, L.; Zhou, G.-Z.; Cao, G.; et al. Upregulation of the Long Non-coding RNA PlncRNA-1 Promotes Esophageal Squamous Carcinoma Cell Proliferation and Correlates with Advanced Clinical Stage. Am. J. Dig. Dis. 2013, 59, 591–597. [Google Scholar] [CrossRef] [PubMed]
  73. Shahryari, A.; Rafiee, M.R.; Fouani, Y.; Oliae, N.A.; Samaei, N.M.; Shafiee, M.; Semnani, S.; Vasei, M.; Mowla, S.J. Two Novel Splice Variants of SOX2OT, SOX2OT-S1, and SOX2OT-S2 are Coupregulated with SOX2 and OCT4 in Esophageal Squamous Cell Carcinoma. Stem Cells 2013, 32, 126–134. [Google Scholar] [CrossRef] [PubMed]
  74. Xie, H.-W.; Wu, Q.-Q.; Zhu, B.; Chen, F.-J.; Ji, L.; Li, S.-Q.; Wang, C.-M.; Tong, Y.-S.; Tuo, L.; Wu, M.; et al. Long noncoding RNA SPRY4-IT1 is upregulated in esophageal squamous cell carcinoma and associated with poor prognosis. Tumor Biol. 2014, 35, 7743–7754. [Google Scholar] [CrossRef]
  75. Li, J.-Y.; Ma, X.; Zhang, C.-B. Overexpression of long non-coding RNA UCA1 predicts a poor prognosis in patients with esophageal squamous cell carcinoma. Int. J. Clin. Exp. Pathol. 2014, 7, 7938–7944. [Google Scholar]
  76. Gao, T.; He, B.; Pan, Y.; Xu, Y.; Li, R.; Deng, Q.; Sun, H.; Wang, S. Long non-coding RNA 91H contributes to the occurrence and progression of esophageal squamous cell carcinoma by inhibiting IGF2 expression. Mol. Carcinog. 2014, 54, 359–367. [Google Scholar] [CrossRef]
  77. Zhang, H.; Luo, H.; Hu, Z.; Peng, J.; Jiang, Z.; Song, T.; Wu, B.; Yue, J.; Zhou, R.; Xie, R.; et al. Targeting WISP1 to sensitize esophageal squamous cell carcinoma to irradiation. Oncotarget 2015, 6, 6218–6234. [Google Scholar] [CrossRef]
  78. Zhang, X.; Xu, Y.; He, C.; Guo, X.; Zhang, J.; He, C.; Zhang, L.; Kong, M.; Chen, B.; Zhu, C. Elevated expression of CCAT2 is associated with poor prognosis in esophageal squamous cell carcinoma. J. Surg. Oncol. 2015, 111, 834–839. [Google Scholar] [CrossRef]
  79. Wu, H.; Zheng, J.; Deng, J.; Zhang, L.; Li, N.; Li, W.; Li, F.; Lu, J.; Zhou, Y. LincRNA-uc002yug.2 involves in alternative splicing of RUNX1 and serves as a predictor for esophageal cancer and prognosis. Oncogene 2014, 34, 4723–4734. [Google Scholar] [CrossRef] [Green Version]
  80. Kang, M.; Sang, Y.; Gu, H.; Zheng, L.; Wang, L.; Liu, C.; Shi, Y.; Shao, A.; Ding, G.; Chen, S.; et al. Long noncoding RNAs POLR2E rs3787016 C/T and HULC rs7763881 A/C polymorphisms are associated with decreased risk of esophageal cancer. Tumor Biol. 2015, 36, 6401–6408. [Google Scholar] [CrossRef]
  81. Hu, L.; Wu, Y.; Tan, D.; Meng, H.; Wang, K.; Bai, Y.; Yang, K. Up-regulation of long noncoding RNA MALAT1 contributes to proliferation and metastasis in esophageal squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2015, 34, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. 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] [PubMed]
  83. Shi, W.-H.; Wu, Q.-Q.; Li, S.-Q.; Yang, T.-X.; Liu, Z.-H.; Tong, Y.-S.; Tuo, L.; Wang, S.; Cao, X.-F. Upregulation of the long noncoding RNA PCAT-1 correlates with advanced clinical stage and poor prognosis in esophageal squamous carcinoma. Tumor Biol. 2015, 36, 2501–2507. [Google Scholar] [CrossRef]
  84. Tong-Xin, Y.; Wang, X.-W.; Zhou, X.-L.; Liu, Z.-H.; Yang, T.-X.; Shi, W.-H.; Xie, H.-W.; Lv, J.; Wu, Q.-Q.; Cao, X.-F. Identification of the long non-coding RNA POU3F3 in plasma as a novel biomarker for diagnosis of esophageal squamous cell carcinoma. Mol. Cancer 2015, 14, 1–13. [Google Scholar] [CrossRef] [Green Version]
  85. Wu, Y.; Liu, X.; Hu, L.; Tao, H.; Guan, X.; Zhang, K.; Bai, Y.; Yang, K. Copy number loss of variation_91720 in PIK3CA predicts risk of esophageal squamous cell carcinoma. Int. J. Clin. Exp. Pathol. 2015, 8, 14479–14485. [Google Scholar] [PubMed]
  86. Xu, Y.; Wang, J.; Qiu, M.; Xu, L.; Li, M.; Jiang, F.; Yin, R.; Xu, L. Upregulation of the long noncoding RNA TUG1 promotes proliferation and migration of esophageal squamous cell carcinoma. Tumor Biol. 2014, 36, 1643–1651. [Google Scholar] [CrossRef]
  87. Wang, Y.-L.; Bai, Y.; Yao, W.-J.; Guo, L.; Wang, Z.-M. Expression of long non-coding RNA ZEB1-AS1 in esophageal squamous cell carcinoma and its correlation with tumor progression and patient survival. Int. J. Clin. Exp. Pathol. 2015, 8, 11871–11876. [Google Scholar]
  88. Luo, H.; Huang, M.; Guo, J.; Fan, R.; Xia, X.; He, J.; Chen, X. AFAP1-AS1 is upregulated and promotes esophageal squamous cell carcinoma cell proliferation and inhibits cell apoptosis. Cancer Med. 2016, 5, 2879–2885. [Google Scholar] [CrossRef] [Green Version]
  89. Liu, Z.; Yang, T.; Xu, Z.; Cao, X. Upregulation of the long non-coding RNA BANCR correlates with tumor progression and poor prognosis in esophageal squamous cell carcinoma. Biomed. Pharmacother. 2016, 82, 406–412. [Google Scholar] [CrossRef]
  90. Lu, C.; Yang, L.; Chen, H.; Shan, Z. Upregulated long non-coding RNA BC032469 enhances carcinogenesis and metastasis of esophageal squamous cell carcinoma through regulating hTERT expression. Tumor Biol. 2016, 37, 16065–16075. [Google Scholar] [CrossRef]
  91. Cao, X.-G.; Zhao, R.-H.; Zhu, C.-H.; Li, X.-K.; Cao, W.; Zong, H.; Hu, H.-Y. BC200 LncRNA a potential predictive marker of poor prognosis in esophageal squamous cell carcinoma patients. Onco Targets Ther. 2016, 9, 2221–2226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Pan, Z.; Mao, W.; Bao, Y.; Zhang, M.; Su, X.; Xu, X. The long noncoding RNA CASC9 regulates migration and invasion in esophageal cancer. Cancer Med. 2016, 5, 2442–2447. [Google Scholar] [CrossRef] [PubMed]
  93. Chen, X.; Han, H.; Li, Y.; Zhang, Q.; Mo, K.; Chen, S. Upregulation of long noncoding RNA HOTTIP promotes metastasis of esophageal squamous cell carcinoma via induction of EMT. Oncotarget 2016, 7, 84480–84485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Sahebi, R.; Malakootian, M.; Balalaee, B.; Shahryari, A.; Khoshnia, M.; Abbaszadegan, M.R.; Moradi, A.; Mowla, S.J. Linc-ROR and its spliced variants 2 and 4 are significantly up-regulated in esophageal squamous cell carcinoma. Iran. J. Basic Med. Sci. 2016, 19, 1131–1135. [Google Scholar] [CrossRef]
  95. Guo, W.; Dong, Z.; Shi, Y.; Liu, S.; Liang, J.; Guo, Y.; Guo, X.; Shen, S.; Shan, B. Aberrant methylation-mediated downregulation of long noncoding RNA LOC100130476 correlates with malignant progression of esophageal squamous cell carcinoma. Dig. Liver Dis. 2016, 48, 961–969. [Google Scholar] [CrossRef]
  96. Lv, D.; Sun, R.; Yu, Q.; Zhang, X. The long non-coding RNA maternally expressed gene 3 activates p53 and is downregulated in esophageal squamous cell cancer. Tumor Biol. 2016, 37, 16259–16267. [Google Scholar] [CrossRef]
  97. Shafiee, M.; Aleyasin, S.A.; Vasei, M.; Semnani, S.S.; Mowla, S.J. Down-Regulatory Effects of miR-211 on Long Non-Coding RNA SOX2OT and SOX2 Genes in Esophageal Squamous Cell Carcinoma. Cell J. 2016, 17, 593–600. [Google Scholar] [CrossRef]
  98. Li, Z.; Wu, X.; Gu, L.; Shen, Q.; Luo, W.; Deng, C.; Zhou, Q.; Chen, X.; Li, Y.; Lim, Z.; et al. Long non-coding RNA ATB promotes malignancy of esophageal squamous cell carcinoma by regulating miR-200b/Kindlin-2 axis. Cell Death Dis. 2017, 8, e2888. [Google Scholar] [CrossRef]
  99. Zhang, E.; Han, L.; Yin, D.; He, X.; Hong, L.; Si, X.; Qiu, M.; Xu, T.; De, W.; Xu, L.; et al. H3K27 acetylation activated-long non-coding RNA CCAT1 affects cell proliferation and migration by regulating SPRY4 and HOXB13 expression in esophageal squamous cell carcinoma. Nucleic Acids Res. 2016, 45, 3086–3101. [Google Scholar] [CrossRef]
  100. Cui, Y.; Wu, W.; Lv, P.; Zhang, J.; Bai, B.; Cao, W. Down-regulation of long non-coding RNA ESCCAL_1 inhibits tumor growth of esophageal squamous cell carcinoma in a xenograft mouse model. Oncotarget 2017, 9, 783–790. [Google Scholar] [CrossRef]
  101. Liu, H.; Zhen, Q.; Fan, Y. LncRNA GHET1 Promotes Esophageal Squamous Cell Carcinoma Cells Proliferation and Invasion via Induction of EMT. Int. J. Biol. Markers 2017, 32, 403–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Wang, G.; Zhao, W.; Gao, X.; Zhang, D.; Li, Y. HNF1A?AS1 promotes growth and metastasis of esophageal squamous cell carcinoma by sponging miR?214 to upregulate the expression of SOX-4. Int. J. Oncol. 2017, 51, 657–667. [Google Scholar] [CrossRef] [PubMed]
  103. Da, C.; Zhan, Y.; Li, Y.; Tan, Y.; Li, R.; Wang, R. The expression and significance of HOX transcript antisense RNA and epithelial-mesenchymal transition-related factors in esophageal squamous cell carcinoma. Mol. Med. Rep. 2017, 15, 1853–1862. [Google Scholar] [CrossRef]
  104. Liang, Y.; Wu, Y.; Chen, X.; Zhang, S.; Wang, K.; Guan, X.; Yang, K.; Li, J.; Bai, Y. A novel long noncoding RNA linc00460 up-regulated by CBP/P300 promotes carcinogenesis in esophageal squamous cell carcinoma. Biosci. Rep. 2017, 37, BSR20171019. [Google Scholar] [CrossRef]
  105. Han, L.; Liu, S.; Liang, J.; Guo, Y.; Shen, S.; Guo, X.; Dong, Z.; Guo, W. A genetic polymorphism at miR-526b binding-site in the lincRNA-NR_024015 exon confers risk of esophageal squamous cell carcinoma in a population of North China. Mol. Carcinog. 2017, 56, 960–971. [Google Scholar] [CrossRef]
  106. Dong, Z.; Zhang, A.; Liu, S.; Lu, F.; Guo, Y.; Zhang, G.; Xu, F.; Shi, Y.; Shen, S.; Liang, J.; et al. Aberrant Methylation-Mediated Silencing of lncRNA MEG3 Functions as a ceRNA in Esophageal Cancer. Mol. Cancer Res. 2017, 15, 800–810. [Google Scholar] [CrossRef] [Green Version]
  107. Ren, Z.-P.; Chu, X.-Y.; Xue, Z.-Q.; Zhang, L.-B.; Wen, J.-X.; Deng, J.-Q.; Hou, X.-B. Down-regulation of lncRNA MIR31HG correlated with aggressive clinicopathological features and unfavorable prognosis in esophageal squamous cell carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 3866–3870. [Google Scholar]
  108. Wu, X.; Lim, Z.-F.; Li, Z.; Gu, L.; Ma, W.; Zhou, Q.; Su, H.; Wang, X.; Yang, X.; Zhang, Z. NORAD Expression Is Associated with Adverse Prognosis in Esophageal Squamous Cell Carcinoma. Oncol. Res. Treat. 2017, 40, 370–374. [Google Scholar] [CrossRef]
  109. Feng, F.; Qiu, B.; Zang, R.; Song, P.; Gao, S. Pseudogene PHBP1 promotes esophageal squamous cell carcinoma proliferation by increasing its cognate gene PHB expression. Oncotarget 2017, 8, 29091–29100. [Google Scholar] [CrossRef] [Green Version]
  110. Li, P.-D.; Hu, J.-L.; Ma, C.; Ma, H.; Yao, J.; Chen, L.-L.; Chen, J.; Cheng, T.-T.; Yang, K.-Y.; Wu, G.; et al. Upregulation of the long non-coding RNA PVT1 promotes esophageal squamous cell carcinoma progression by acting as a molecular sponge of miR-203 and LASP1. Oncotarget 2017, 8, 34164–34176. [Google Scholar] [CrossRef]
  111. Yao, G.-L.; Pan, C.-F.; Xu, H.; Wei, K.; Liu, B.; Zhai, R.; Chen, Y.-J. Long noncoding RNA RP11-766N7.4 functions as a tumor suppressor by regulating epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Biomed. Pharmacother. 2017, 88, 778–785. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, K.; Chen, J.; Song, H.; Chen, L.-B. SNHG16/miR-140-5p axis promotes esophagus cancer cell proliferation, migration and EMT formation through regulating ZEB1. Oncotarget 2017, 9, 1028–1040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Wu, X.; Dinglin, X.; Wang, X.; Luo, W.; Shen, Q.; Li, Y.; Gu, L.; Zhou, Q.; Zhu, H.; Li, Y.; et al. Long noncoding RNA XIST promotes malignancies of esophageal squamous cell carcinoma via regulation of miR-101/EZH2. Oncotarget 2017, 8, 76015–76028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Shi, H.; Liu, Z.; Pei, D.; Jiang, Y.; Zhu, H.; Chen, B. Development and validation of nomogram based on lncRNA ZFAS1 for predicting survival in lymph node-negative esophageal squamous cell carcinoma patients. Oncotarget 2017, 8, 59048–59057. [Google Scholar] [CrossRef] [Green Version]
  115. Liu, B.; Pan, C.-F.; Yao, G.-L.; Wei, K.; Xia, Y.; Chen, Y.-J. The long non-coding RNA AK001796 contributes to tumor growth via regulating expression of p53 in esophageal squamous cell carcinoma. Cancer Cell Int. 2018, 18, 38. [Google Scholar] [CrossRef] [Green Version]
  116. Shi, H.; Shi, J.; Zhang, Y.; Guan, C.; Zhu, J.; Wang, F.; Xu, M.; Ju, Q.; Fang, S.; Jiang, M. Long non-coding RNA DANCR promotes cell proliferation, migration, invasion and resistance to apoptosis in esophageal cancer. J. Thorac. Dis. 2018, 10, 2573–2582. [Google Scholar] [CrossRef]
  117. Wang, Z.; Ren, B.; Huang, J.; Yin, R.; Jiang, F.; Zhang, Q. LncRNA DUXAP10 modulates cell proliferation in esophageal squamous cell carcinoma through epigenetically silencing p21. Cancer Biol. Ther. 2018, 19, 998–1005. [Google Scholar] [CrossRef] [Green Version]
  118. Xu, L.-J.; Yu, X.-J.; Wei, B.; Hui, H.-X.; Sun, Y.; Dai, J.; Chen, X.-F. Long non-coding RNA DUXAP8 regulates proliferation and invasion of esophageal squamous cell cancer. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2646–2652. [Google Scholar]
  119. Yao, J.; Shen, X.; Li, H.; Xu, J.; Shao, S.; Huang, J.X.; Lin, M. LncRNA-ECM is overexpressed in esophageal squamous cell carcinoma and promotes tumor metastasis. Oncol. Lett. 2018, 16, 3935–3942. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, X.-D.; Huang, G.-W.; Xie, Y.-H.; He, J.-Z.; Guo, J.-C.; Xu, X.-E.; Liao, L.-D.; Xie, Y.-M.; Song, Y.-M.; Lian-Di, L.; et al. The interaction of lncRNA EZR-AS1 with SMYD3 maintains overexpression of EZR in ESCC cells. Nucleic Acids Res. 2017, 46, 1793–1809. [Google Scholar] [CrossRef] [Green Version]
  121. Chen, M.; Liu, P.; Chen, Y.; Chen, Z.; Shen, M.; Liu, X.; Li, X.; Li, A.; Lin, Y.; Yang, R.; et al. Long Noncoding RNA FAM201A Mediates the Radiosensitivity of Esophageal Squamous Cell Cancer by Regulating ATM and mTOR Expression via miR-101. Front. Genet. 2018, 9, 611. [Google Scholar] [CrossRef] [PubMed]
  122. Ma, W.; Zhang, C.-Q.; Li, H.-L.; Gu, J.; Miao, G.-Y.; Cai, H.-Y.; Wang, J.-K.; Zhang, L.-J.; Song, Y.-M.; Tian, Y.-H.; et al. LncRNA FER1L4 suppressed cancer cell growth and invasion in esophageal squamous cell carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2638–2645. [Google Scholar] [PubMed]
  123. Bao, J.; Zhou, C.; Zhang, J.; Mo, J.; Ye, Q.; He, J.; Diao, J. Upregulation of the long noncoding RNA FOXD2-AS1 predicts poor prognosis in esophageal squamous cell carcinoma. Cancer Biomark. 2018, 21, 527–533. [Google Scholar] [CrossRef] [PubMed]
  124. Ke, K.; Sun, Z.; Wang, Z. Downregulation of long non-coding RNA GAS5 promotes cell proliferation, migration and invasion in esophageal squamous cell carcinoma. Oncol. Lett. 2018, 16, 1801–1808. [Google Scholar] [CrossRef] [Green Version]
  125. Sun, X.Y.; Wang, X.F.; Cui, Y.B.; Cao, X.G.; Zhao, R.H.; Wei, H.Y.; Cao, W.; Wu, W. [Expression level and clinical significance of LncRNA HOXA11-AS in esophageal squamous cell carcinoma patients]. Zhonghua Zhong Liu Za Zhi Chin. J. Oncol. 2018, 40, 186–190. [Google Scholar]
  126. Chen, Z.; Lin, J.; Wu, S.; Xu, C.; Chen, F.; Huang, Z. Up-regulated miR-548k promotes esophageal squamous cell carcinoma progression via targeting long noncoding RNA-LET. Exp. Cell Res. 2018, 362, 90–101. [Google Scholar] [CrossRef]
  127. Sun, Y.; Wang, J.; Pan, S.; Yang, T.; Sun, X.; Wang, Y.; Shi, X.; Zhao, X.; Guo, J.; Zhang, X. LINC00657 played oncogenic roles in esophageal squamous cell carcinoma by targeting miR-615-3p and JunB. Biomed. Pharmacother. 2018, 108, 316–324. [Google Scholar] [CrossRef]
  128. Yang, X.-Z.; He, Q.-J.; Cheng, T.-T.; Chi, J.; Lei, Z.-Y.; Tang, Z.; Liao, Q.-X.; Zhang, H.; Zeng, L.-S.; Cui, S.-Z. Predictive Value of LINC01133 for Unfavorable Prognosis was Impacted by Alcohol in Esophageal Squamous Cell Carcinoma. Cell. Physiol. Biochem. 2018, 48, 251–262. [Google Scholar] [CrossRef]
  129. Wang, B.; Liang, T.; Li, J. Long noncoding RNA LINC01296 is associated with poor prognosis in ESCC and promotes ESCC cell proliferation, migration and invasion. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4524–4531. [Google Scholar]
  130. Xie, J.-J.; Jiang, Y.-Y.; Jiang, Y.; Li, C.; Lim, M.-C.; An, O.; Mayakonda, A.; Ding, L.-W.; Long, L.; Sun, C.; et al. Super-Enhancer-Driven Long Non-Coding RNA LINC01503, Regulated by TP63, Is Over-Expressed and Oncogenic in Squamous Cell Carcinoma. Gastroenterology 2018, 154, 2137–2151. [Google Scholar] [CrossRef] [Green Version]
  131. Niu, G.; Zhuang, H.; Li, B.; Cao, G. Long noncoding RNA linc-UBC1 promotes tumor invasion and metastasis by regulating EZH2 and repressing E-cadherin in esophageal squamous cell carcinoma. JBUON Off. J. Balk. Union Oncol. 2018, 23, 157–162. [Google Scholar]
  132. Guo, W.; Liu, S.; Dong, Z.; Guo, Y.; Ding, C.; Shen, S.; Liang, J.; Shan, B. Aberrant methylation-mediated silencing of lncRNA CTC-276P9.1 is associated with malignant progression of esophageal squamous cell carcinoma. Clin. Exp. Metastasis 2018, 35, 53–68. [Google Scholar] [CrossRef] [PubMed]
  133. Yoon, J.-H.; You, B.-H.; Park, C.H.; Kim, Y.J.; Nam, J.-W.; Kil Lee, S. The long noncoding RNA LUCAT1 promotes tumorigenesis by controlling ubiquitination and stability of DNA methyltransferase 1 in esophageal squamous cell carcinoma. Cancer Lett. 2018, 417, 47–57. [Google Scholar] [CrossRef] [PubMed]
  134. Li, Y.; Li, J.; Luo, M.; Zhou, C.; Shi, X.; Yang, W.; Lu, Z.; Chen, Z.; Sun, N.; He, J. Novel long noncoding RNA NMR promotes tumor progression via NSUN2 and BPTF in esophageal squamous cell carcinoma. Cancer Lett. 2018, 430, 57–66. [Google Scholar] [CrossRef]
  135. Chen, R.; Xia, W.; Wang, X.; Qiu, M.; Yin, R.; Wang, S.; Xi, X.; Wang, J.; Xu, Y.; Dong, G.; et al. Upregulated long non-coding RNA SBF2-AS1 promotes proliferation in esophageal squamous cell carcinoma. Oncol. Lett. 2018, 15, 5071–5080. [Google Scholar] [CrossRef] [PubMed]
  136. Fan, R.; Guo, J.; Yan, W.; Huang, M.; Zhu, C.; Yin, Y.; Chen, X. Small nucleolar host gene 6 promotes esophageal squamous cell carcinoma cell proliferation and inhibits cell apoptosis. Oncol. Lett. 2018, 15, 6497–6502. [Google Scholar] [CrossRef] [Green Version]
  137. Lin, C.; Zhang, S.; Wang, Y.; Wang, Y.; Nice, E.; Guo, C.; Zhang, E.; Chenyu, L.; Li, M.; Liu, C.; et al. Functional Role of a Novel Long Noncoding RNA TTN-AS1 in Esophageal Squamous Cell Carcinoma Progression and Metastasis. Clin. Cancer Res. 2017, 24, 486–498. [Google Scholar] [CrossRef] [Green Version]
  138. Kang, K.; Huang, Y.-H.; Li, H.-P.; Guo, S.-M. Expression of UCA1 and MALAT1 long-chain non-coding RNAs in esophageal squamous cell carcinoma tissues is predictive of patient prognosis. Arch. Med. Sci. 2018, 14, 752–759. [Google Scholar] [CrossRef]
  139. Sun, K.; Zhang, G. Long noncoding RNA CASC2 suppresses esophageal squamous cell carcinoma progression by increasing SOCS1 expression. Cell Biosci. 2019, 9, 1–14. [Google Scholar] [CrossRef]
  140. Zhang, C.; Wang, L.; Yang, J.; Fu, Y.; Li, H.; Xie, L.; Cui, Y. MicroRNA-33a-5p suppresses esophageal squamous cell carcinoma progression via regulation of lncRNA DANCR and ZEB1. Eur. J. Pharmacol. 2019, 861, 172590. [Google Scholar] [CrossRef]
  141. Wang, M.; Li, Y.; Yang, Y.; Liu, X.; Zang, M.; Li, Y.; Yang, K.; Yang, W.; Zhang, S. Long non-coding RNA DLX6-AS1 is associated with malignant progression and promotes proliferation and invasion in esophageal squamous cell carcinoma. Mol. Med. Rep. 2018, 19, 1942–1950. [Google Scholar] [CrossRef] [Green Version]
  142. Zhang, H.; Hua, Y.; Jiang, Z.; Yue, J.; Shi, M.; Zheng, X.; Zhang, X.; Yang, L.; Zhou, R.; Wu, S. Cancer-associated Fibroblast–promoted LncRNA DNM3OS Confers Radioresistance by Regulating DNA Damage Response in Esophageal Squamous Cell Carcinoma. Clin. Cancer Res. 2019, 25, 1989–2000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Zhang, Y.; Zhang, L.; Wang, R.; Wang, B.; Hua, P.; Li, J. LncRNA Erbb4-IR promotes esophageal squamous cell carcinoma (ESCC) by downregulating miR-145. J. Cell. Biochem. 2019, 120, 17566–17572. [Google Scholar] [CrossRef]
  144. Li, W.; Zhang, L.; Guo, B.; Deng, J.; Wu, S.; Li, F.; Wang, Y.; Lu, J.; Zhou, Y. Exosomal FMR1-AS1 facilitates maintaining cancer stem-like cell dynamic equilibrium via TLR7/NFkappaB/c-Myc signaling in female esophageal carcinoma. Mol. Cancer 2019, 18, 22. [Google Scholar] [CrossRef] [PubMed]
  145. Yan, Y.; Li, S.; Wang, S.; Rubegni, P.; Tognetti, L.; Zhang, J.; Yan, L. Long noncoding RNA HAND2-AS1 inhibits cancer cell proliferation, migration, and invasion in esophagus squamous cell carcinoma by regulating microRNA-21. J. Cell Biochem. 2019, 120, 9564–9571. [Google Scholar] [CrossRef]
  146. Huang, J.; Li, J.; Li, Y.; Lu, Z.; Che, Y.; Mao, S.; Lei, Y.; Zang, R.; Zheng, S.; Liu, C.; et al. Interferon-inducible lncRNA IRF1-AS represses esophageal squamous cell carcinoma by promoting interferon response. Cancer Lett. 2019, 459, 86–99. [Google Scholar] [CrossRef] [PubMed]
  147. Zhang, Y.; Chen, W.; Pan, T.; Wang, H.; Zhang, Y.; Li, C. LBX2-AS1 is activated by ZEB1 and promotes the development of esophageal squamous cell carcinoma by interacting with HNRNPC to enhance the stability of ZEB1 and ZEB2 mRNAs. Biochem. Biophys. Res. Commun. 2019, 511, 566–572. [Google Scholar] [CrossRef]
  148. Zong, M.-Z.; Feng, W.-T.; Du, N.; Yu, X.-J.; Yu, W.-Y. Upregulation of long noncoding RNA LEF1-AS1 predicts a poor prognosis in patients with esophageal squamous cell carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7929–7934. [Google Scholar]
  149. Yang, Y.; Sun, X.; Chi, C.; Liu, Y.; Lin, C.; Xie, D.; Shen, X.; Lin, X. Upregulation of long noncoding RNA LINC00152 promotes proliferation and metastasis of esophageal squamous cell carcinoma. Cancer Manag. Res 2019, 11, 4643–4654. [Google Scholar] [CrossRef] [Green Version]
  150. He, Z. LINC00473/miR-497-5p Regulates Esophageal Squamous Cell Carcinoma Progression Through Targeting PRKAA1. Cancer Biother. Radiopharm. 2019, 34, 650–659. [Google Scholar] [CrossRef]
  151. Zhang, D.; Zhang, H.; Wang, X.; Hu, B.; Zhang, F.; Wei, H.; Li, L. LINC01518 knockdown inhibits tumorigenicity by suppression of PIK3CA/Akt pathway in oesophageal squamous cell carcinoma. Artif. Cells Nanomed. Biotechnol. 2019, 47, 4284–4292. [Google Scholar] [CrossRef] [PubMed]
  152. Zhang, S.; Liang, Y.; Wu, Y.; Chen, X.; Wang, K.; Li, J.; Guan, X.; Xiong, G.; Yang, K.; Bai, Y. Upregulation of a novel lncRNA LINC01980 promotes tumor growth of esophageal squamous cell carcinoma. Biochem. Biophys. Res. Commun. 2019, 513, 73–80. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, S.-M.; Li, S.-Y.; Hao-Bin, Y.; Lin-Yan, X.; Sheng, X. IL-11 activated by lnc-ATB promotes cell proliferation and invasion in esophageal squamous cell cancer. Biomed. Pharmacother. 2019, 114, 108835. [Google Scholar] [CrossRef] [PubMed]
  154. Li, F.-Z.; Zang, W.-Q. Knockdown of lncRNAXLOC_001659 inhibits proliferation and invasion of esophageal squamous cell carcinoma cells. World J. Gastroenterol. 2019, 25, 6299–6310. [Google Scholar] [CrossRef] [PubMed]
  155. Jing, L.; Lin, J.; Zhao, Y.; Liu, G.-J.; Liu, Y.-B.; Feng, L.; Yang, H.-Y.; Cui, W.-X.; Zhang, X.-H. Long noncoding RNA LSINCT5 is upregulated and promotes the progression of esophageal squamous cell carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5195–5205. [Google Scholar]
  156. Chu, J.; Li, H.; Xing, Y.; Jia, J.; Sheng, J.; Yang, L.; Sun, K.; Qu, Y.; Zhang, Y.; Yin, H.; et al. LncRNA MNX1-AS1 promotes progression of esophageal squamous cell carcinoma by regulating miR-34a/SIRT1 axis. Biomed. Pharmacother. 2019, 116, 109029. [Google Scholar] [CrossRef]
  157. Shi, W.; Wang, Q.; Bian, Y.; Fan, Y.; Zhou, Y.; Feng, T.; Li, Z.; Cao, X. Long noncoding RNA PANDA promotes esophageal squamous carcinoma cell progress by dissociating from NF-YA but interact with SAFA. Pathol.-Res. Pr. 2019, 215, 152604. [Google Scholar] [CrossRef]
  158. Zang, B.; Zhao, J.; Chen, C. LncRNA PCAT-1 Promoted ESCC Progression via Regulating ANXA10 Expression by Sponging miR-508-3p. Cancer Manag. Res. 2019, 11, 10841–10849. [Google Scholar] [CrossRef]
  159. Zhihua, Z.; Weiwei, W.; Lihua, N.; Jianying, Z.; Jiang, G. p53-induced long non-coding RNA PGM5-AS1 inhibits the progression of esophageal squamous cell carcinoma through regulating miR-466/PTEN axis. IUBMB Life 2019, 71, 1492–1502. [Google Scholar] [CrossRef]
  160. Dong, Z.; Liang, X.; Wu, X.; Kang, X.; Guo, Y.; Shen, S.; Liang, J.; Guo, W. Promoter hypermethylation-mediated downregulation of tumor suppressor gene SEMA3B and lncRNA SEMA3B-AS1 correlates with progression and prognosis of esophageal squamous cell carcinoma. Clin. Exp. Metastasis 2019, 36, 225–241. [Google Scholar] [CrossRef]
  161. Zhang, C.; Jiang, F.; Su, C.; Xie, P.; Xu, L. Upregulation of long noncoding RNA SNHG20 promotes cell growth and metastasis in esophageal squamous cell carcinoma via modulating ATM-JAK-PD-L1 pathway. J. Cell. Biochem. 2019, 120, 11642–11650. [Google Scholar] [CrossRef] [PubMed]
  162. Zhang, Y.; Li, R.; Ding, X.; Zhang, K.; Qin, W. Upregulation of long non-coding RNA SNHG6 promote esophageal squamous cell carcinoma cell malignancy and its diagnostic value. Am. J. Transl. Res. 2019, 11, 1084–1091. [Google Scholar] [PubMed]
  163. Song, H.; Song, J.; Lu, L.; Li, S. SNHG8 is upregulated in esophageal squamous cell carcinoma and directly sponges microRNA-411 to increase oncogenicity by upregulating KPNA2. Onco Targets Ther. 2019, 12, 6991–7004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Shen, F.-F.; Pan, Y.; Yang, H.-J.; Li, J.-K.; Zhao, F.; Su, J.-F.; Li, Y.-Y.; Tian, L.-Q.; Yu, P.-T.; Cao, Y.-T.; et al. Decreased expression of SPINT1-AS1 and SPINT1 mRNA might be independent unfavorable prognostic indicators in esophageal squamous cell carcinoma. Onco Targets Ther. 2019, 12, 4755–4763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Chen, C.; Shu, L.; Zou, W. Role of long non-coding RNA TP73-AS1 in cancer. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef]
  166. Dong, Z.; Li, S.; Wu, X.; Niu, Y.; Liang, X.; Yang, L.; Guo, Y.; Shen, S.; Liang, J.; Guo, W. Aberrant hypermethylation-mediated downregulation of antisense lncRNA ZNF667-AS1 and its sense gene ZNF667 correlate with progression and prognosis of esophageal squamous cell carcinoma. Cell Death Dis. 2019, 10, 1–18. [Google Scholar] [CrossRef] [Green Version]
  167. Liu, J.; Liu, Z.-X.; Wu, Q.-N.; Lu, Y.-X.; Wong, C.-W.; Miao, L.; Wang, Y.; Wang, Z.; Jin, Y.; He, M.-M.; et al. Long noncoding RNA AGPG regulates PFKFB3-mediated tumor glycolytic reprogramming. Nat. Commun. 2020, 11, 1507–1516. [Google Scholar] [CrossRef]
  168. Sang, Y.; Gu, H.; Chen, Y.; Shi, Y.; Liu, C.; Lv, L.; Sun, Y.; Zhang, Y. Long non-coding RNA CASC8 polymorphisms are associated with the risk of esophageal cancer in a Chinese population. Thorac. Cancer 2020, 11, 2852–2857. [Google Scholar] [CrossRef]
  169. Feng, Z.; Li, X.; Qiu, M.; Luo, R.; Lin, J.; Liu, B. LncRNA EGFR-AS1 Upregulates ROCK1 by Sponging miR-145 to Promote Esophageal Squamous Cell Carcinoma Cell Invasion and Migration. Cancer Biother. Radiopharm. 2020, 35, 66–71. [Google Scholar] [CrossRef]
  170. Zhang, C.; Luo, Y.; Cao, J.; Wang, X.; Miao, Z.; Shao, G. Exosomal lncRNA FAM225A accelerates esophageal squamous cell carcinoma progression and angiogenesis via sponging miR-206 to upregulate NETO2 and FOXP1 expression. Cancer Med. 2020, 9, 8600–8611. [Google Scholar] [CrossRef]
  171. Feng, B.; Wang, G.; Liang, X.; Wu, Z.; Wang, X.; Dong, Z.; Guo, Y.; Shen, S.; Liang, J.; Guo, W. LncRNA FAM83H-AS1 promotes oesophageal squamous cell carcinoma progression via miR-10a-5p/Girdin axis. J Cell Mol Med. 2020, 24, 8962–8976. [Google Scholar]
  172. Gao, J.; Zhang, Z.; Su, H.; Zong, L.; Li, Y. Long Noncoding RNA FGD5-AS1 Acts as a Competing Endogenous RNA on microRNA-383 to Enhance the Malignant Characteristics of Esophageal Squamous Cell Carcinoma by Increasing SP1 Expression. Cancer Manag. Res. 2020, 12, 2265–2278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Li, Y.; Li, T.; Yang, Y.; Kang, W.; Dong, S.; Cheng, S. YY1-induced upregulation of FOXP4-AS1 and FOXP4 promote the proliferation of esophageal squamous cell carcinoma cells. Cell Biol. Int. 2020, 44, 1447–1457. [Google Scholar] [CrossRef] [PubMed]
  174. Li, X.; Xiao, X.; Chang, R.; Zhang, C. Comprehensive bioinformatics analysis identifies lncRNA HCG22 as a migration inhibitor in esophageal squamous cell carcinoma. J. Cell. Biochem. 2019, 121, 468–481. [Google Scholar] [CrossRef] [PubMed]
  175. Wang, D.; You, D.; Pan, Y.; Liu, P. Downregulation of lncRNA-HEIH curbs esophageal squamous cell carcinoma progression by modulating miR-4458/PBX3. Thorac. Cancer 2020, 11, 1963–1971. [Google Scholar] [CrossRef]
  176. Wang, B.; Hua, P.; Zhang, L.; Li, J.; Zhang, Y. LncRNA-IUR up-regulates PTEN by sponging miR-21 to regulate cancer cell proliferation and apoptosis in esophageal squamous cell carcinoma. Esophagus 2020, 17, 298–304. [Google Scholar] [CrossRef]
  177. Liu, J.-Q.; Deng, M.; Xue, N.-N.; Li, T.-X.; Guo, Y.-X.; Gao, L.; Zhao, D.; Fan, R.-T. lncRNA KLF3-AS1 Suppresses Cell Migration and Invasion in ESCC by Impairing miR-185-5p-Targeted KLF3 Inhibition. Mol. Ther.-Nucleic Acids 2020, 20, 231–241. [Google Scholar] [CrossRef]
  178. Wu, S.; Zhang, L.; Deng, J.; Guo, B.; Li, F.; Wang, Y.; Wu, R.; Zhang, S.; Lu, J.; Zhou, Y. A Novel Micropeptide Encoded by Y-Linked LINC00278 Links Cigarette Smoking and AR Signaling in Male Esophageal Squamous Cell Carcinoma. Cancer Res. 2020, 80, 2790–2803. [Google Scholar] [CrossRef] [Green Version]
  179. Zhang, Z.; Liang, X.; Ren, L.; Zhang, S.; Li, S.; Wan, T.; Xu, D.; Lv, S. LINC00662 promotes cell viability and metastasis in esophageal squamous cell carcinoma by sponging miR -340-5p and upregulating HOXB2. Thorac. Cancer 2020, 11, 2306–2315. [Google Scholar] [CrossRef]
  180. Zhou, M.; Mao, Y.; Yu, S.; Li, Y.; Yin, R.; Zhang, Q.; Lu, T.; Sun, R.; Lin, S.; Qian, Y.; et al. LINC00673 Represses CDKN2C and Promotes the Proliferation of Esophageal Squamous Cell Carcinoma Cells by EZH2-Mediated H3K27 Trimethylation. Front. Oncol. 2020, 10, 1546. [Google Scholar] [CrossRef]
  181. Liu, H.-F.; Zhen, Q.; Fan, Y.-K. LINC00963 predicts poor prognosis and promotes esophageal cancer cells invasion via targeting miR-214-5p/RAB14 axis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 164–173. [Google Scholar] [PubMed]
  182. Zhao, M.; Cui, H.; Zhao, B.; Li, M.; Man, H. Long intergenic non-coding RNA LINC01232 contributes to esophageal squamous cell carcinoma progression by sequestering microRNA-654-3p and consequently promoting hepatoma-derived growth factor expression. Int. J. Mol. Med. 2020, 46, 2007–2018. [Google Scholar] [CrossRef] [PubMed]
  183. Zheng, L.; Liu, Y.-T.; Wu, C.-P.; Jiang, J.-T.; Zhang, L.; Wang, Z.-L.; Wang, Q.-Y. Long non-coding RNA linc01433 promotes tumorigenesis and progression in esophageal squamous cell carcinoma by sponging miR-1301. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 4785–4792. [Google Scholar] [PubMed]
  184. Du, J.; Zhang, G.; Qiu, H.; Yu, H.; Yuan, W. A novel positive feedback loop of linc02042 and c-Myc mediated by YBX1 promotes tumorigenesis and metastasis in esophageal squamous cell carcinoma. Cancer Cell Int. 2020, 20, 1–10. [Google Scholar] [CrossRef]
  185. Lu, T.; Ma, K.; Zhan, C.; Yang, X.; Shi, Y.; Jiang, W.; Wang, H.; Wang, S.; Wang, Q.; Tan, L. Downregulation of long non-coding RNA LINP1 inhibits the malignant progression of esophageal squamous cell carcinoma. Ann. Transl. Med. 2020, 8, 675. [Google Scholar] [CrossRef]
  186. Ma, J.; Xiao, Y.; Tian, B.; Chen, S.; Zhang, B.; Wu, J.; Wu, Z.; Li, X.; Tang, J.; Yang, D.; et al. Long noncoding RNA lnc-ABCA12-3 promotes cell migration, invasion, and proliferation by regulating fibronectin 1 in esophageal squamous cell carcinoma. J. Cell. Biochem. 2019, 121, 1374–1387. [Google Scholar] [CrossRef]
  187. Liu, G.; Guo, W.; Chen, G.; Li, W.; Cui, Y.; Qin, J.; Peng, J. Lnc-MCEI mediated the chemosensitivity of esophageal squamous cell carcinoma via miR-6759-5p to competitively regulate IGF2. Int. J. Biol. Sci. 2020, 16, 2938–2950. [Google Scholar] [CrossRef]
  188. Wang, P.; Yang, Z.; Ye, T.; Shao, F.; Li, J.; Sun, N.; He, J. lncTUG1/miR-144-3p affect the radiosensitivity of esophageal squamous cell carcinoma by competitively regulating c-MET. J. Exp. Clin. Cancer Res. 2020, 39, 7. [Google Scholar] [CrossRef]
  189. Guan, Z.; Wang, Y.; Wang, Y.; Liu, X.; Wang, Y.; Zhang, W.; Chi, X.; Dong, Y.; Liu, X.; Shao, S.; et al. Long non-coding RNA LOC100133669 promotes cell proliferation in oesophageal squamous cell carcinoma. Cell Prolif. 2020, 53, e12750. [Google Scholar] [CrossRef] [Green Version]
  190. Wang, G.; Feng, B.; Niu, Y.; Wu, J.; Yang, Y.; Shen, S.; Guo, Y.; Liang, J.; Guo, W.; Dong, Z. A novel long noncoding RNA, LOC440173, promotes the progression of esophageal squamous cell carcinoma by modulating the miR-30d-5p/HDAC9 axis and the epithelial-mesenchymal transition. Mol. Carcinog. 2020, 59, 1392–1408. [Google Scholar] [CrossRef]
  191. Hu, W.; Chen, Z.; Chen, J.; Cai, D.; Chen, C.; Fang, T. LOC441178 Overexpression Inhibits the Proliferation and Migration of Esophageal Carcinoma Cells via Methylation of miR-182. Onco Targets Ther. 2020, 13, 11253–11263. [Google Scholar] [CrossRef] [PubMed]
  192. Qian, C.J.; Xu, Z.R.; Chen, L.Y.; Wang, Y.C.; Yao, J. LncRNA MAFG-AS1 Accelerates Cell Migration, Invasion and Aerobic Glycolysis of Esophageal Squamous Cell Carcinoma Cells via miR-765/PDX1 Axis. Cancer Manag. Res. 2020, 12, 6895–6908. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, C.; Xie, L.; Fu, Y.; Yang, J.; Cui, Y. lncRNA MIAT promotes esophageal squamous cell carcinoma progression by regulating miR-1301-3p/INCENP axis and interacting with SOX2. J. Cell. Physiol. 2020, 235, 7933–7944. [Google Scholar] [CrossRef] [PubMed]
  194. Li, H.; Jia, J.; Yang, L.; Chu, J.; Sheng, J.; Wang, C.; Meng, W.; Jia, Z.; Yin, H.; Wan, J.; et al. LncRNA MIR205HG Drives Esophageal Squamous Cell Carcinoma Progression by Regulating miR-214/SOX4 Axis. Onco Targets Ther. 2020, 13, 13097–13109. [Google Scholar] [CrossRef]
  195. Li, D.; Li, D.; Meng, L.; Liu, J.; Huang, C.; Sun, H. LncRNA NLIPMT Inhibits Tumorigenesis in Esophageal Squamous-Cell Carcinomas by Regulating miR-320/Survivin Axis. Cancer Manag. Res. 2020, 12, 12603–12612. [Google Scholar] [CrossRef]
  196. Qiu, B.Q.; Lin, X.H.; Ye, X.D.; Huang, W.; Pei, X.; Xiong, D.; Long, X.; Zhu, S.Q.; Lu, F.; Lin, K.; et al. Long non-coding RNA PSMA3-AS1 promotes malignant phenotypes of esophageal cancer by modulating the miR-101/EZH2 axis as a ceRNA. Aging (Albany NY) 2020, 12, 1843–1856. [Google Scholar] [CrossRef]
  197. Li, Z.W.; Zhang, T.Y.; Yue, G.J.; Tian, X.; Wu, J.Z.; Feng, G.Y.; Wang, Y.S. Small nucleolar RNA host gene 22 (SNHG22) promotes the progression of esophageal squamous cell carcinoma by miR-429/SESN3 axis. Ann. Transl. Med. 2020, 8, 1007. [Google Scholar] [CrossRef]
  198. Liang, M.; Pan, Z.; Yu, F.; Chen, C. Long noncoding RNA SNHG12 suppresses esophageal squamous cell carcinoma progression through competing endogenous RNA networks. Clin. Transl. Oncol. 2020, 22, 1786–1795. [Google Scholar] [CrossRef]
  199. Wang, W.; Yang, J. Long noncoding RNA TTTY15 promotes growth and metastasis of esophageal squamous cell carcinoma by sponging microRNA-337-3p to upregulate the expression of JAK2. Anti-Cancer Drugs 2020, 31, 1038–1045. [Google Scholar] [CrossRef]
  200. Zhao, K.; Guo, Y.; Huo, Z.; Ma, G.; Zhang, G.; Xing, Y.; Xu, Q. [Serum level of lncRNA TUSC7 in patients with esophageal squamous cell carcinoma and its role in promoting tumor cell migration and invasion]. Nan Fang Yi Ke Da Xue Xue Bao 2020, 40, 661–669. [Google Scholar]
  201. Yao, J.; Zhang, H.; Li, H.; Qian, R.; Liu, P.; Huang, J. P53-regulated lncRNA uc061hsf.1 inhibits cell proliferation and metastasis in human esophageal squamous cell cancer. IUBMB Life 2019, 72, 401–412. [Google Scholar] [CrossRef] [PubMed]
  202. Du, F.; Guo, T.; Cao, C. Restoration of UPK1A-AS1 Expression Suppresses Cell Proliferation, Migration, and Invasion in Esophageal Squamous Cell Carcinoma Cells Partially by Sponging microRNA-1248. Cancer Manag. Res. 2020, ume 12, 2653–2662. [Google Scholar] [CrossRef] [Green Version]
  203. Zhang, Q.; Guan, F.; Fan, T.; Li, S.; Ma, S.; Zhang, Y.; Guo, W.; Liu, H. LncRNA WDFY3-AS2 suppresses proliferation and invasion in oesophageal squamous cell carcinoma by regulating miR-2355-5p/SOCS2 axis. J. Cell. Mol. Med. 2020, 24, 8206–8220. [Google Scholar] [CrossRef] [PubMed]
  204. Xu, J.-H.; Chen, R.-Z.; Liu, L.-Y.; Li, X.-M.; Wu, C.-P.; Zhou, Y.-T.; Yan, J.-D.; Zhang, Z.-Y. LncRNA ZEB2-AS1 promotes the proliferation, migration and invasion of esophageal squamous cell carcinoma cell through miR-574-3p/HMGA2 axis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 5391–5403. [Google Scholar]
  205. Sun, G.; Wu, C. ZFPM2-AS1 facilitates cell growth in esophageal squamous cell carcinoma via up-regulating TRAF4. Biosci. Rep. 2020, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Zhang, H.; Wang, Y.; Zhang, W.; Wu, Q.; Fan, J.; Zhan, Q. BAALC-AS1/G3BP2/c-Myc feedback loop promotes cell proliferation in esophageal squamous cell carcinoma. Cancer Commun. 2021, 41, 240–257. [Google Scholar] [CrossRef]
  207. Yan, S.; Xu, J.; Liu, B.; Ma, L.; Feng, H.; Tan, H.; Fang, C. Long non-coding RNA BCAR4 aggravated proliferation and migration in esophageal squamous cell carcinoma by negatively regulating p53/p21 signaling pathway. Bioengineered 2021, 12, 682–696. [Google Scholar] [CrossRef] [PubMed]
  208. Qin, B.; Dong, M.; Wang, Z.; Wan, J.; Xie, Y.; Jiao, Y.; Yan, D. Long non-coding RNA CASC15 facilitates esophageal squamous cell carcinoma tumorigenesis via decreasing SIM2 stability via FTO-mediated demethylation. Oncol. Rep. 2020, 45, 1059–1071. [Google Scholar] [CrossRef] [PubMed]
  209. Duan, Y.; Jia, Y.; Wang, J.; Liu, T.; Cheng, Z.; Sang, M.; Lv, W.; Qin, J.; Liu, L. Long noncoding RNA DGCR5 involves in tumorigenesis of esophageal squamous cell carcinoma via SRSF1-mediated alternative splicing of Mcl-1. Cell Death Dis. 2021, 12, 1–14. [Google Scholar] [CrossRef]
  210. Liu, J.; Zhou, R.; Deng, M.; Xue, N.; Li, T.; Guo, Y.; Gao, L.; Fan, R.; Zhao, D. Long non-coding RNA DIO3OS binds to microRNA-130b to restore radiosensitivity in esophageal squamous cell carcinoma by upregulating PAX9. Cancer Gene Ther. 2021, 1–12. [Google Scholar] [CrossRef]
  211. Jia, J.; Li, H.; Chu, J.; Sheng, J.; Wang, C.; Jia, Z.; Meng, W.; Yin, H.; Wan, J.; He, F. LncRNA FAM83A-AS1 promotes ESCC progression by regulating miR-214/CDC25B axis. J. Cancer 2021, 12, 1200–1211. [Google Scholar] [CrossRef] [PubMed]
  212. Wang, P.; Yang, S.; Dai, S.; Ni, Q.; Liu, H.; Yu, L.; Lu, K.; Han, G.; Huang, J. Expression and Clinical Value of LncRNA GAPLINC in Esophageal Squamous Cell Carcinoma. Onco Targets Ther. 2021, 14, 4039–4045. [Google Scholar] [CrossRef] [PubMed]
  213. Xu, H.; Miao, J.; Liu, S.; Liu, H.; Zhang, L.; Zhang, Q. Long non-coding RNA KCNQ1 overlapping transcript 1 promotes the progression of esophageal squamous cell carcinoma by adsorbing microRNA-133b. Clinics 2021, 76, e2175. [Google Scholar] [CrossRef] [PubMed]
  214. Liang, X.; Lu, J.; Wu, Z.; Guo, Y.; Shen, S.; Liang, J.; Dong, Z.; Guo, W. LINC00239 Interacts with C-Myc Promoter-Binding Protein-1 (MBP-1) to Promote Expression of C-Myc in Esophageal Squamous Cell Carcinoma. Mol. Cancer Res. 2021, 19, 1465–1475. [Google Scholar] [CrossRef]
  215. Wang, L.; Wang, X.; Yan, P.; Liu, Y.; Jiang, X. LINC00261 Suppresses Cisplatin Resistance of Esophageal Squamous Cell Carcinoma Through miR-545-3p/MT1M Axis. Front. Cell Dev. Biol. 2021, 9, 1915. [Google Scholar] [CrossRef]
  216. Liu, Z.; Yang, S.; Chen, X.; Dong, S.; Zhou, S.; Xu, S. LncRNA LINC00467 acted as an oncogene in esophageal squamous cell carcinoma by accelerating cell proliferation and preventing cell apoptosis via the miR-485-5p / DPAGT1 axis. J. Gastroenterol. Hepatol. 2020, 36, 721–730. [Google Scholar] [CrossRef]
  217. Ye, H.; Shrestha, S.M.; Zhu, J.; Ding, Y.; Shi, R. Long non-coding RNA LINC00491 promotes proliferation and inhibits apoptosis in esophageal squamous cell carcinoma. Int. J. Mol. Med. 2021, 47, 1-1. [Google Scholar] [CrossRef]
  218. Peng, X.; Zhou, Y.; Chen, Y.; Tang, L.; Wang, G.; Jiang, H.; Wang, X.; Tao, Y.; Zhuang, W. Reduced LINC00551 expression promotes proliferation and invasion of esophageal squamous cancer by increase in HSP27 phosphorylation. J. Cell. Physiol. 2020, 236, 1418–1431. [Google Scholar] [CrossRef]
  219. Zhang, Y.; Zhu, H.; Sun, N.; Zhang, X.; Liang, G.; Zhu, J.; Xia, L.; Kou, Y.; Lu, J. Linc00941 regulates esophageal squamous cell carcinoma via functioning as a competing endogenous RNA for miR-877-3p to modulate PMEPA1 expression. Aging 2021, 13, 17830–17846. [Google Scholar] [CrossRef]
  220. Wang, B.; Tang, D.; Liu, Z.; Wang, Q.; Xue, S.; Zhao, Z.; Feng, D.; Sheng, C.; Li, J.; Zhou, Z. LINC00958 promotes proliferation, migration, invasion, and epithelial-mesenchymal transition of oesophageal squamous cell carcinoma cells. PLoS ONE 2021, 16, e0251797. [Google Scholar] [CrossRef]
  221. Huang, G.-W.; Chen, Q.-Q.; Ma, C.-C.; Xie, L.-H.; Gu, J. linc01305 promotes metastasis and proliferation of esophageal squamous cell carcinoma through interacting with IGF2BP2 and IGF2BP3 to stabilize HTR3A mRNA. Int. J. Biochem. Cell Biol. 2021, 136, 106015. [Google Scholar] [CrossRef] [PubMed]
  222. Tan, Z.; Zhou, P.; Zhu, Z.; Wang, Y.; Guo, Z.; Shen, M.; Xiao, Y.; Shen, W.; Wu, D. Upregulated long noncoding RNA LincIN promotes tumor progression via the regulation of nuclear factor 90/microRNA7/HOXB13 in esophageal squamous cell carcinoma. Int. J. Mol. Med. 2021, 47, 1–12. [Google Scholar] [CrossRef] [PubMed]
  223. Rong, H.; Chen, B.; Ma, K.; Wei, X.; Peng, J.; Zhu, J. Downregulation of lncRNA LINC-PINT Participates in the Recurrence of Esophageal Squamous Cell Carcinoma Possibly by Interacting miRNA-21. Cancer Biother. Radiopharm. 2021, 36, 273–279. [Google Scholar] [CrossRef] [PubMed]
  224. Kong, D.; Long, D.; Liu, B.; Pei, D.; Cao, N.; Zhang, G.; Xia, Z.; Luo, M. Downregulation of long non-coding RNA LOC101928477 correlates with tumor progression by regulating the epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Thorac. Cancer 2021, 12, 1303–1311. [Google Scholar] [CrossRef]
  225. Luo, J.; Xie, K.; Gao, X.; Yao, Y.; Wang, G.; Shao, C.; Li, X.; Xu, Y.; Ren, B.; Hu, L.; et al. Long Noncoding RNA Nuclear Paraspeckle Assembly Transcript 1 Promotes Progression and Angiogenesis of Esophageal Squamous Cell Carcinoma through miR-590-3p/MDM2 Axis. Front. Oncol. 2021, 10. [Google Scholar] [CrossRef]
  226. Wang, Y.; Bao, D.; Wan, L.; Zhang, C.; Hui, S.; Guo, H. Long non-coding RNA small nucleolar RNA host gene 7 facilitates the proliferation, migration, and invasion of esophageal cancer cells by regulating microRNA-625. J. Gastrointest. Oncol. 2021, 12, 423–432. [Google Scholar] [CrossRef]
  227. Li, H.; Chu, J.; Jia, J.; Sheng, J.; Zhao, X.; Xing, Y.; He, F. LncRNA LOXL1-AS1 promotes esophageal squamous cell carcinoma progression by targeting DESC1. J. Cancer 2021, 12, 530–538. [Google Scholar] [CrossRef]
  228. Li, J.; Han, X.; Gu, Y.; Wu, J.; Song, J.; Shi, Z.; Chang, H.; Liu, M.; Zhang, Y. LncRNA MTX2-6 Suppresses Cell Proliferation by Acting as ceRNA of miR-574-5p to Accumulate SMAD4 in Esophageal Squamous Cell Carcinoma. Front. Cell Dev. Biol. 2021, 9, 496. [Google Scholar] [CrossRef]
  229. Gu, S.; Qian, L.; Liu, Y.; Miao, J.; Shen, H.; Zhang, S.; Mao, G. Upregulation of long non-coding RNA MYU promotes proliferation, migration and invasion of esophageal squamous cell carcinoma cells. Exp. Ther. Med. 2021, 21, 1–9. [Google Scholar] [CrossRef]
  230. Wei, S.; Sun, S.; Zhou, X.; Zhang, C.; Li, X.; Dai, S.; Wang, Y.; Zhao, L.; Shan, B. SNHG5 inhibits the progression of EMT through the ubiquitin-degradation of MTA2 in oesophageal cancer. Carcinogenesis 2020, 42, 315–326. [Google Scholar] [CrossRef]
  231. Cheng, J.; Ma, H.; Yan, M.; Xing, W. THAP9-AS1/miR-133b/SOX4 positive feedback loop facilitates the progression of esophageal squamous cell carcinoma. Cell Death Dis. 2021, 12, 401. [Google Scholar] [CrossRef] [PubMed]
  232. Pan, Q.; Li, B.; Zhang, J.; Du, X.; Gu, D. LncRNA THAP9-AS1 accelerates cell growth of esophageal squamous cell carcinoma through sponging miR-335–5p to regulate SGMS2. Pathol.-Res. Pr. 2021, 224, 153526. [Google Scholar] [CrossRef] [PubMed]
  233. Shi, Z.; Li, G.; Li, Z.; Liu, J.; Tang, Y. TMEM161B-AS1 suppresses proliferation, invasion and glycolysis by targeting miR-23a-3p/HIF1AN signal axis in oesophageal squamous cell carcinoma. J. Cell Mol. Med. 2021, 25, 6535–6549. [Google Scholar] [CrossRef] [PubMed]
  234. Wang, Y.; Zhang, W.; Liu, W.; Huang, L.; Wang, Y.; Li, D.; Wang, G.; Zhao, Z.; Chi, X.; Xue, Y.; et al. Long Noncoding RNA VESTAR Regulates Lymphangiogenesis and Lymph Node Metastasis of Esophageal Squamous Cell Carcinoma by Enhancing VEGFC mRNA Stability. Cancer Res. 2021, 81, 3187–3199. [Google Scholar] [CrossRef]
Figure 1. The bar graph represents the trends of lncRNAs evolution with year.
Figure 1. The bar graph represents the trends of lncRNAs evolution with year.
Curroncol 29 00189 g001
Figure 2. Role of Wnt/β-catenin signaling pathway in cancer. In the activated canonical pathway. Wnt allows the connection between Frizzled receptor and lipoprotein receptor-related protein (LRP), which further activates the Dishevelled followed by inhibition of glycogen synthase kinase 3 (GSK-3β), axis inhibitor (AXIN), adenomatous polyposis coli (APC) and cyclin-dependent kinase inhibitor (CKIα) complex. This complex inhibits the phosphorylation of β-catenin, subsequently enters into the nucleus, and transcribes the cancer-related genes with the help of the TCF/LEF complex. The mechanism is vice-versa in the inhibited canonical Wnt pathway. The non-canonical Wnt pathway utilizes Wnt5a for the activation of the pathway and allows the gene transcription through calcium ions. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Figure 2. Role of Wnt/β-catenin signaling pathway in cancer. In the activated canonical pathway. Wnt allows the connection between Frizzled receptor and lipoprotein receptor-related protein (LRP), which further activates the Dishevelled followed by inhibition of glycogen synthase kinase 3 (GSK-3β), axis inhibitor (AXIN), adenomatous polyposis coli (APC) and cyclin-dependent kinase inhibitor (CKIα) complex. This complex inhibits the phosphorylation of β-catenin, subsequently enters into the nucleus, and transcribes the cancer-related genes with the help of the TCF/LEF complex. The mechanism is vice-versa in the inhibited canonical Wnt pathway. The non-canonical Wnt pathway utilizes Wnt5a for the activation of the pathway and allows the gene transcription through calcium ions. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Curroncol 29 00189 g002
Figure 3. Dysregulation profile of lncRNAs in various cancers including ESCC irrespective of signaling pathways. Each colored box represents the association of lncRNAs with various cancers.
Figure 3. Dysregulation profile of lncRNAs in various cancers including ESCC irrespective of signaling pathways. Each colored box represents the association of lncRNAs with various cancers.
Curroncol 29 00189 g003
Figure 4. Mechanism of Wnt/β-catenin related lncRNAs in normal and tumorigenic conditions. LncRNA HOTAIR in association with PRC2 complex inhibits the WIF-1 expression, and thus Wnt5B becomes free and allows the binding of the Frizzled receptor and lipoprotein receptor-related protein (LRP). As a result, β-catenin does get not phosphorylated and eventually enters the nucleus and transcribes the c-Myc, cyclin D1, Bcl-2, EMT genes. Furthermore, low expression of MEG3 inhibits the Dickkopf-2 (DKK2), whereas, GASL1 and UCA1 inhibit the Dickkopf-1 (DKK1). In addition, HERES inhibits the expression of Nemo Like Kinase (NLK). As a result, an enormous amount of β-catenin is generated and transcribe the c-Myc, cyclin D1, Bcl-2, EMT genes. At the same time, another set of lncRNAs Taurine Up-Regulated 1 (TUG1), LINC00675, Small Nucleolar RNA Host Gene 16 (SNHG16) localized in the nucleus and executes their action on cancer-related proteins. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Figure 4. Mechanism of Wnt/β-catenin related lncRNAs in normal and tumorigenic conditions. LncRNA HOTAIR in association with PRC2 complex inhibits the WIF-1 expression, and thus Wnt5B becomes free and allows the binding of the Frizzled receptor and lipoprotein receptor-related protein (LRP). As a result, β-catenin does get not phosphorylated and eventually enters the nucleus and transcribes the c-Myc, cyclin D1, Bcl-2, EMT genes. Furthermore, low expression of MEG3 inhibits the Dickkopf-2 (DKK2), whereas, GASL1 and UCA1 inhibit the Dickkopf-1 (DKK1). In addition, HERES inhibits the expression of Nemo Like Kinase (NLK). As a result, an enormous amount of β-catenin is generated and transcribe the c-Myc, cyclin D1, Bcl-2, EMT genes. At the same time, another set of lncRNAs Taurine Up-Regulated 1 (TUG1), LINC00675, Small Nucleolar RNA Host Gene 16 (SNHG16) localized in the nucleus and executes their action on cancer-related proteins. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Curroncol 29 00189 g004
Figure 5. Mechanism of PI3K/Akt/mTOR pathway in cancer. The tyrosine kinase (RTK) receptor becomes activated by growth factors, hormones, and cytokines, which activates the p85 subunit, Ras and phosphatase and tensin homolog (PTEN). As a result, GSK-3β becomes inhibited, which further activates the mTORC1 complex and thus manifests the hallmarks of cancer. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Figure 5. Mechanism of PI3K/Akt/mTOR pathway in cancer. The tyrosine kinase (RTK) receptor becomes activated by growth factors, hormones, and cytokines, which activates the p85 subunit, Ras and phosphatase and tensin homolog (PTEN). As a result, GSK-3β becomes inhibited, which further activates the mTORC1 complex and thus manifests the hallmarks of cancer. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Curroncol 29 00189 g005
Figure 6. Interaction of lncRNAs with PI3K/Akt/mTOR pathway in ESCC. Long non-coding RNA Papillary Thyroid Carcinoma Susceptibility Candidate 1 (PTCSC1) upregulates the p85 subunit and thus phosphorylate the Akt and increases the mechanistic target of rapamycin kinase (mTOR) levels followed by an increase in phosphodiesterase 4A (PDE4A) expression. At the same time, lncRNA HCP5 acts on the PDE4A and further increases its level, enhancing the cell proliferation, angiogenesis, migration, and invasion of ESCC cells. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Figure 6. Interaction of lncRNAs with PI3K/Akt/mTOR pathway in ESCC. Long non-coding RNA Papillary Thyroid Carcinoma Susceptibility Candidate 1 (PTCSC1) upregulates the p85 subunit and thus phosphorylate the Akt and increases the mechanistic target of rapamycin kinase (mTOR) levels followed by an increase in phosphodiesterase 4A (PDE4A) expression. At the same time, lncRNA HCP5 acts on the PDE4A and further increases its level, enhancing the cell proliferation, angiogenesis, migration, and invasion of ESCC cells. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Curroncol 29 00189 g006
Figure 7. Crosstalks between Wnt/ β-catenin and PI3K/Akt/mTOR pathway. The activation of both signaling pathways results in the inhibition of glycogen synthase kinase-3β (GSK-3β) activity via various upstream events. In the Wnt/β-catenin pathway, a fraction of AXIN-bound GSK3β has a vital role in controlling β-catenin degradation through the regulation of β-catenin phosphorylation. At the same time, activated PI3K phosphorylate Akt at Ser9 residue, which further inhibits the GSK3β activity. As a result, transcription of oncogenes was initiated. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Figure 7. Crosstalks between Wnt/ β-catenin and PI3K/Akt/mTOR pathway. The activation of both signaling pathways results in the inhibition of glycogen synthase kinase-3β (GSK-3β) activity via various upstream events. In the Wnt/β-catenin pathway, a fraction of AXIN-bound GSK3β has a vital role in controlling β-catenin degradation through the regulation of β-catenin phosphorylation. At the same time, activated PI3K phosphorylate Akt at Ser9 residue, which further inhibits the GSK3β activity. As a result, transcription of oncogenes was initiated. This illustration was created using resources available at www.biorender.com (accessed on 25 September 2021).
Curroncol 29 00189 g007
Table 1. Characteristics of lncRNAs in clinical studies.
Table 1. Characteristics of lncRNAs in clinical studies.
LncRNAExpression Pattern (Up/Down Regulation)DrugConc. of Drugs UsedTime Points of TreatmentPatient Tissue/Cell Line/In Vivo ModelClinical EndpointPathological ResponseCohort SizeReferences
LOC285194DownCisplatinNANATissue and cell linesDFS and OSCR = 15%Female = 48[54]
Male = 94
TUG1UpCisplatin1 μg/mL48 hTissue and cellsOSNAMale = 171
Female = 47
[56]
[57]
AFAP1-AS1Up5-Fluorouracial Cisplatin
Paclitaxel
2, 4, 8, 16, 32, 64, 128, 256 μM
0.3125, 0.625, 1.25, 2.5 5, 10, 25, 50 μM
0.03125, 0.0652, 0.125, 0.25, 0.5, 1, 2, 4, 8 μM
24 h on days 1–4Tissue and cellsOS and PFSCR = 19.8%
PR = 40.7%
NC = 37.7%
PD = 1.8%
Male = 123
Female = 39
[58]
PART1UpGefitinib0.01–10 μM48 hSerumNANA79[63]
TUSC7DownCisplatin
5-Fluorouracial
1, 2, 4, 8, 16 μM
1, 4, 16, 32,
64 μM
48 hTissue and cell linesOSNAMale = 43
Female= 19
[55]
CCAT1UpCisplatin0.1, 0.2, 0.5, 1, 2, 5 μM48 hCell linesNANANA[59]
LINC01419Up5-fluorouracil10 μg/mL48 hTissue and cell linesNANA76[60]
LINC00337UpCisplatin0.5, 1, 2, 3 μg/mL48 hTissue and cell linesNANAMale = 48
Female = 26
[51]
Linc01014UpGefitinib10 μM48 hCell linesNANANA [18]
MACC1-AS1 UpCisplatin20, 40, 60, 80, 100 µMNATissue and cell linesNANAMale = 62
Female = 8
[61]
FOXD2-AS1UpCisplatin20, 40, 60, 80, 100 µM
6.25, 12.5, 25, 50, 100 µg/mL
NATissue and cell linesNANAMale = 62
Female = 8
[61]
[62]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sharma, U.; Murmu, M.; Barwal, T.S.; Tuli, H.S.; Jain, M.; Prakash, H.; Kaceli, T.; Jain, A.; Bishayee, A. A Pleiotropic Role of Long Non-Coding RNAs in the Modulation of Wnt/β-Catenin and PI3K/Akt/mTOR Signaling Pathways in Esophageal Squamous Cell Carcinoma: Implication in Chemotherapeutic Drug Response. Curr. Oncol. 2022, 29, 2326-2349. https://doi.org/10.3390/curroncol29040189

AMA Style

Sharma U, Murmu M, Barwal TS, Tuli HS, Jain M, Prakash H, Kaceli T, Jain A, Bishayee A. A Pleiotropic Role of Long Non-Coding RNAs in the Modulation of Wnt/β-Catenin and PI3K/Akt/mTOR Signaling Pathways in Esophageal Squamous Cell Carcinoma: Implication in Chemotherapeutic Drug Response. Current Oncology. 2022; 29(4):2326-2349. https://doi.org/10.3390/curroncol29040189

Chicago/Turabian Style

Sharma, Uttam, Masang Murmu, Tushar Singh Barwal, Hardeep Singh Tuli, Manju Jain, Hridayesh Prakash, Tea Kaceli, Aklank Jain, and Anupam Bishayee. 2022. "A Pleiotropic Role of Long Non-Coding RNAs in the Modulation of Wnt/β-Catenin and PI3K/Akt/mTOR Signaling Pathways in Esophageal Squamous Cell Carcinoma: Implication in Chemotherapeutic Drug Response" Current Oncology 29, no. 4: 2326-2349. https://doi.org/10.3390/curroncol29040189

Article Metrics

Back to TopTop