Open Access

Non‑coding RNA: A promising diagnostic biomarker and therapeutic target for esophageal squamous cell carcinoma (Review)

  • Authors:
    • Longze Zhang
    • Yanyang Wang
    • Jianmei Gao
    • Xue Zhou
    • Minglei Huang
    • Xianyao Wang
    • Zhixu He
  • View Affiliations

  • Published online on: April 9, 2024     https://doi.org/10.3892/ol.2024.14388
  • Article Number: 255
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Esophageal cancer (EC) is a common form of malignant tumor in the digestive system that is classified into two types: Esophageal squamous cell carcinomas (ESCC) and esophageal adenocarcinoma. ESCC is known for its early onset of symptoms, which can be difficult to identify, as well as its rapid progression and tendency to develop drug resistance to chemotherapy and radiotherapy. These factors contribute to the high incidence of disease and low cure rate. Therefore, a diagnostic biomarker and therapeutic target need to be identified for ESCC. Non‑coding RNAs (ncRNAs) are a class of molecules that are transcribed from DNA but do not encode proteins. Initially, ncRNAs were considered to be non‑functional segments generated during transcription. However, with advancements in high‑throughput sequencing technologies in recent years, ncRNAs have been associated with poor prognosis, drug resistance and progression of ESCC. The present study provides a comprehensive overview of the biogenesis, characteristics and functions of ncRNAs, particularly focusing on microRNA, long ncRNAs and circular RNAs. Furthermore, the ncRNAs that could potentially be used as diagnostic biomarkers and therapeutic targets for ESCC are summarized to highlight their application value and prospects in ESCC.

Introduction

Esophageal cancer (EC), which can be divided into esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC), is a common malignancy of the digestive tract (1). In 2020, ~604,000 new cases were reported and ~544,000 EC-associated mortalities were reported worldwide (2). EAC occurs mostly in North America and Western Europe, while ESCC occurs mostly in China (3,4). ESCC is usually caused by smoking, alcohol intake, poor diet, viral infection or genetic factors (5). Surgical resection, chemotherapy and radiotherapy are the primary therapeutic strategies of ESCC. Since ESCC is mostly asymptomatic in the early stages, patients often miss the optimal therapeutic opportunity (6). In addition, the therapy for ESCC is often ineffective due to metastasis and medication resistance (7). Therefore, the identification of promising diagnostic biomarkers and the development of effective therapeutic strategies are crucial in combatting ESCC.

Non-coding RNAs (ncRNAs) encompass a range of types of RNA, with microRNAs (miRNAs/miRs), long ncRNAs (lncRNAs) and circular RNAs (circRNAs) being the most well-known. They are transcribed from DNA but lack the ability to encode proteins and were previously considered to be ‘transcriptional waste’ (810). However, with the advent of high-throughput sequencing technology, recent research has revealed that ncRNAs constitute >90% of the human genome (11). By contrast, protein-coding genes make up <2% of the genome (1214). NcRNAs are widely involved in various cellular processes, such as cell proliferation, differentiation, aging, apoptosis and immune responses (1518). Furthermore, extensive research has highlighted the significant dysregulation of expression of numerous ncRNAs in ESCC cells and tissues. This dysregulation is closely linked to ESCC progression, drug resistance and unfavorable patient prognosis (1922). The present review provides a comprehensive overview of the functions of miRNAs, lncRNAs and circRNAs in ESCC, and investigates their roles as diagnostic biomarkers and potential therapeutic targets for ESCC treatment. This information highlights novel ideas and strategies for the early diagnosis and potential therapeutic targets for ESCC.

Characteristics of ncRNAs

The generation of ncRNAs is a multifaceted and intricate process that involves several sequential stages. In the subsequent sections, we provide a comprehensive and detailed explanation of the formation mechanisms that underlie miRNAs, lncRNAs and circRNAs (Fig. 1).

Characteristics of miRNAs

MiRNAs are a category of endogenous and highly conserved small ncRNAs that consist of ~22 nucleotides (23,24). Processing involves two stages. In the first stage, RNA polymerase II catalyzes DNA fragments in the nucleus to produce primary miRNA (pri-miRNA) (25); subsequently, a microprocessor complex, consisting of Drosha, an RNase type III protein and DiGeorge syndrome critical region gene 8, recognizes and cleaves the stem strand on the pri-miRNA hairpin to form pre-miRNA (26). Thereafter, Ran guanosine triphosphate-Exportin 5 facilitates pre-miRNA transfers from the nucleus to the cytoplasm (27,28). In the second stage, a mature double-stranded miRNA is generated after pre-miRNA is identified by Dicer with trans-activation response RNA-binding protein/protein activator of the interferon-induced protein kinase (28,29). The stem-loop structure is then cleaved and modified in the cytoplasm (30,31). The guide strand attaches to the RNA-induced silencing complex and targets the 3′-untranslated Region (3′-UTR) of the mRNA via base complementary pairing when the mature miRNA unwinds (32). The passenger strand is degraded in the cytoplasm (33). Complete base-pair complementation between miRNA and mRNA then causes cleavage of mRNA. An incomplete match can inhibit protein translation of mRNA, which is the most common mode of action of miRNA (3437).

Characteristics of lncRNAs

LncRNAs are a particular subtype of ncRNAs that are longer than 200 nucleotides and are produced by RNA polymerase II or RNA polymerase III (38,39). They are composed of multiple exons and have a structure similar to mRNA. However, lncRNA transcripts have fewer exons, lower overall expression levels and poorer sequence conservation compared with mRNA (40,41). LncRNAs can be divided into six types: i + ii) Bidirectional and intergenic lncRNAs; iii) sense and iv) antisense exonic lncRNAs; and v) sense and vi) antisense intronic lncRNAs, and they are able to regulate different stages of transcription (42,43).

In pre-transcription, lncRNAs participate in the regulation of histone acetylation and DNA methylation (44,45). They also play a regulatory role during transcription (46). In addition, lncRNAs are directly involved in post-transcriptional regulation of mRNA. Some lncRNAs that contain miRNA complementary sites can change the effect of miRNA by binding to the mRNA via a competing endogenous RNA (ceRNA) mechanism (47). LncRNAs can also regulate RNA editing (48). It is clear that lncRNAs have multiple functions and are crucial for the control of transcription.

Characteristics of circRNAs

CircRNAs are covalently closed, single-stranded ncRNAs formed by back-splicing of precursor mRNA (pre-mRNA) (49). CircRNAs lack 5′ caps and 3′ poly (A) tails and are classified into three types: i) Exonic circRNAs (EciRNA); ii) intronic circRNAs (ciRNA); and iii) exon-intron circRNAs (EIciRNA) (50,51). They are highly conserved, diverse and widely expressed in different cells and tissues at various stages of development (5254). The closed-loop structure and the absence of a 5′ cap or 3′ poly (A) tail in circRNAs also make them less susceptible to exonuclease R compared with linear RNA (55).

CircRNAs can competitively bind to proteins, serving as miRNA decoys or protein scaffolds (56). EciRNAs can control the levels of targeted genes by acting as miRNA sponges, due to their numerous miRNA binding sites in the cytoplasm (57,58). CiRNAs and EIciRNAs have few binding sites with miRNA in the nucleus; however, they can regulate gene transcription in the nucleus (5961). Moreover, some circRNAs contain extensive N6-methyladenosine (m6A) modification sites and can translate proteins after m6A modification (62). Studies have shown that a single m6A site is sufficient to drive the initiation of translation (63,64). This m6A-driven translation requires the involvement of eukaryotic translation initiation factor 4 gamma 2 (eIF4G2) and m6A reader YTH m6A RNA binding protein (YTHDF)3. It is also enhanced by methyltransferase like 3/14 methyltransferase, inhibited by fat mass and obesity-associated protein (FTO) demethylase and upregulated upon heat shock (65,66). Insulin-like growth factor 2 mRNA-binding protein 1 facilitates circMAP3K4 peptide translation and then promotes hepatocellular carcinoma progression, which is driven by m6A modification (67). In addition, accumulating evidence suggests that several circRNAs are related to polysomes, and they contain an AUG initiation codon and putative open reading frames of optimal length (65,68). This indicates an unexpected potential for protein coding by these circRNAs.

Role of ncRNAs in ESCC

The pathogenesis of ESCC involves dysregulation, activation, inactivation and mutation of multiple genes, with the exact mechanisms still not fully understood (69). Recent studies emphasize the critical role of ncRNAs in ESCC development (7072) (Fig. 2). Cui et al (73) revealed that m6A-mediated epigenetic modification of lncRNA is closely associated with ESCC tumorigenesis. The FTO m6A demethylase can mediate the demethylation of lncRNA LINC00022 in a YTHDF2-dependent manner, promoting its accumulation in cells. Elevated levels of LINC00022 can bind specifically to the p21 protein and promote its ubiquitination-mediated degradation, thereby upregulating the levels of cyclin dependent kinase 2 and cyclin E1 proteins (73). This leads to a shortened G0/G1 phase and prolonged G2/M phase in the cells, thus driving ESCC development (73). In addition, a study found that the lncRNA HOX transcript antisense RNA can promote ESCC tumorigenesis by binding to miR-1 and upregulating cyclin D1 expression, thereby regulating changes in the cell cycle (74). These findings emphasize the critical role of ncRNAs in ESCC pathogenesis. Current research predominantly explores how ncRNAs promote ESCC progression by regulating cell proliferation, metastasis and therapy resistance (21,75,76). Therefore, this article focuses on elucidating the specific mechanisms and current research status of ncRNAs in regulating ESCC cell proliferation, metastasis, and therapy resistance, alongside their potential as diagnostic markers for ESCC.

NcRNAs regulate proliferation and apoptosis of ESCC cells

Tumor cells exhibit unlimited proliferation and anti-apoptotic capabilities (77). The main challenge in treating tumors lies in controlling the unbridled proliferation of tumor cells in vivo. Prior research has demonstrated that miRNAs, lncRNAs and circRNAs have the ability to regulate the proliferation and death of tumor cells, hence limiting or promoting the development and growth of tumors (78,79).

The miR-106b-5p oncogene is commonly upregulated in various malignancies, such as ESCC (80,81). In patients with ESCC, elevated miR-106b-5p levels are associated with decreased expression of the 15-hydroxyprostaglandin dehydrogenase (HPGD) tumor suppressor gene (82). Dual luciferase reporter assays have confirmed HPGD as a direct target of miR-106b-5p, and miR-106b-5p expression is inversely correlated with HPGD levels (82). Overexpression of miR-106b-5p promotes tumor cell proliferation and inhibits apoptosis, while upregulation of HPGD reverses these effects (82).

The levels of miR-497 have been found to be lower in ESCC tissues and cells when compared with normal tissues (83). An increase in miR-497 expression suppresses ESCC cell growth, while a reduction in miR-497 expression promotes ESCC cell proliferation (83). Furthermore, overexpression of miR-497 in ESCC cells injected into BALB/c nude mice results in a decreased level of proliferating cell nuclear antigen, increased cleaved caspase-3 expression and inhibited tumor growth (83). Mechanistically, it has been revealed that the YY1 protein can inhibit HIF2α expression by binding to the HIF2α promoter region, and miR-497 upregulates YY1 protein expression (83).

Furthermore, He et al (84) showed that miR-377 may control the chromobox protein homolog 3 (CBX3)/P53/P21 axis, which impacts the ability of ESCC stem cells to proliferate. Overexpression of miR-377 downregulates the level of CBX3 but increases P21 protein levels and phosphorylation of P53 protein. Consequently, expression of the CD133 and CD13 ESCC stem cell markers, plus the Nanog transcription factor, Y-Box 2 sex determining region and octamer-binding transcription factor 4, is reduced (84). In addition, the proliferation, sphere formation and tumorigenicity of ESCC stem cells are significantly inhibited. Conversely, miR-377 inhibitor treatment results in the opposite trend (84).

The regulatory impacts of lncRNAs on the progression of ESCC have been shown in several studies. For example, the lncRNA small nucleolar RNA host gene 16 (SNHG16) oncogene is upregulated in the early stages of ESCC (85). It can enhance the stability of Ras homologue family member U and promote the proliferation of ESCC cells by recruiting the EIF4A3 RNA binding protein-eukaryotic translation initiation factor (85). When cells that overexpress SNHG16 are injected into mice, tumor growth is accelerated (85). Similarly, lncRNA GK intronic transcript 1 can competitively bind to mitogen-activated protein kinase 1 (MAPK1). This prevents its interaction with dual specificity phosphatase 6 and inhibits transmission of the extracellular signal-regulated kinase/MAPK1 signaling pathway, which is mediated by dual specificity phosphatase 6 (86). This facilitates growth and suppresses apoptosis of ESCC cells (86).

Xue et al (87) revealed that elevated lncRNA LINC00680 levels in ESCC may promote ESCC cell proliferation and it regulates a ceRNA interaction network known as the LINC00680/miR-423-5p/p21-activated kinase 6 (PAK6) axis. Knockdown of LINC00680 suppresses ESCC cell growth by increasing miR-423-5p expression and decreasing PAK6 expression (87). These effects can be reversed by a miR-423-5p inhibitor or upregulation of PAK6 expression. This suggests that LINC00680 promotes ESCC progression by upregulating PAK6 levels by acting as a sponge for miR-423-5p (87).

CircRNAs that are aberrantly expressed have been found to facilitate the proliferative ability of ESCC cells. Xiong et al (88) demonstrated that circNELL2 can activate the translation of cell division cyclin-6 by acting as a sponge for miR-127-5p, which in turn encourages the growth of ESCC cells. MiR-124 has been demonstrated to be an anti-oncogene and shows markedly declined levels in ESCC (89,90). The levels of circ2646 are elevated and inversely correlated with miR-124 expression in ESCC (91). Increased circ2646 reduces miR-124 levels and promotes tumor cell proliferation (91). Yao et al (92) report elevated circHIPK3 levels in ESCC tissues when compared with normal tissues, which correlates with lymph node metastasis (LNM) and poor tumor differentiation. Knockdown of circHIPK3 inhibits ESCC cell proliferation by acting as a sponge for miR-124 (92). These findings suggest that circRNAs can boost ESCC cell proliferation and tumor formation. Moreover, some circRNAs have been shown to increase apoptosis of ESCC cells (93,94). Jiang et al (95) found that low circ0086414 expression correlates with poor prognosis in patients with ESCC. They observed that circ0086414 inhibits tumor cell proliferation, suppresses glycolysis and increases apoptosis in ESCC cells. These findings highlight the importance of circRNAs in ESCC development and suggest potential therapeutic strategies that target circRNAs.

The aforementioned studies highlight the involvement of ncRNAs in the regulation of tumor cell proliferation via intricate mechanisms. Future research should aim for a deeper understanding of these mechanisms, to distinguish between ncRNAs that promote or inhibit cancer progression. Such insights can guide clinical approaches to treating ESCC.

NcRNAs regulate invasion and migration of ESCC cells

The two main factors that contribute to therapy failure in individuals with advanced-stage ESCC are local invasion and distant metastases (96). It is crucial to understand the molecular processes behind the spread of ESCC and to investigate prospective treatment targets. The present review provides a summary of current research on the connection between ncRNAs and ESCC metastasis.

The levels of miR-17-5p and miR-4443 are elevated in ESCC cells and correlate with the tumor node metastasis (TNM) stage. Both miR-17-5p and miR-4443 promote ESCC cell invasion and migration by targeting metallopeptidase-2 (97). Exosomes, which transport proteins, lipids and nucleic acids, are vital mediators of intercellular communication (98). Exosomes secreted by tumor cells are crucial for the development and spread of malignancies (99). Liu et al (100) revealed a positive correlation between the LNM of patients with ESCC and miR-320b levels in ESCC cell-derived exosomes. MiR-320b is transferred to human lymphatic endothelial cells via exosomes, inhibiting programmed cell death 4, activating protein kinase B (AKT) phosphorylation and promoting peritumoral lymphangiogenesis (100). It also enhances ESCC cell invasion and migration through these pathways (100). In addition, certain miRNAs have been shown to prevent ESCC cell invasion and migration. For instance, miR-338-5p targets epidermal growth factor receptor (EGFR) and the hepatocyte growth factor receptor (cMET) (101). Increasing miR-338-5p levels reduces cMET and EGFR expression and blocks their signaling pathways (101). Consequently, activation of growth factor receptor-bound protein 2-associated binding protein 1, AKT and extracellular signal-regulated kinase are inhibited (101). This prevents the extracellular matrix from remodeling and impedes ESCC cell invasion and migration (101). Moreover, it has been discovered that miR-485-5p significantly inhibits the mRNA and protein expression levels of Flotillin-1 by binding to its 3′-UTR, resulting in increased E-cadherin expression in ESCC cells, whereas the expression levels of Vimentin, N-cadherin, and zinc finger E-box-binding homeobox 1 were decreased (102). This impedes the invasion and migration of ESCC cells by inhibiting the epithelial-mesenchymal transition (EMT) (102). These results underline the critical role that miRNAs play in controlling ESCC cell invasion and migration.

Recently, several lncRNAs have been revealed to act as oncogenic genes and promote ESCC invasion and migration (103,104). The lncRNA LINC02820 correlates with poor prognosis in patients with ESCC. While its overexpression does not impact ESCC cell proliferation, it enhances cell invasion and migration (105). It also causes an increase in levels of the cortactin and phosphorylated Neural Wiskott-Aldrich syndrome protein invadopodia markers (105). A number of malignancies show high levels of expression of the GTPase-activating protein (SH3 domain)-binding protein 2 (G3BP2) oncogene (106,107). Zheng et al (108) found that G3BP2 can promote ESCC invasion and migration by upregulating the levels of hepatoma-derived growth factor mRNA, and the poor prognosis of patients with ESCC has a positive relationship with G3BP2 expression. The lncRNA LINC01554 binds to the RNA recognition motif of G3BP2 and prolongs its half-life by reducing the ubiquitination level of G3BP2. This indicates that LINC01554 is essential for the oncogenic effect of G3BP2 (108). The lncRNA BAALC-AS1 can also directly interact with G3BP2 to block G3BP2-mediated c-Myc degradation, and to enable ESCC invasion and migration (109). A study has revealed a correlation between the lncRNA PTPRG-AS1 and pyruvate dehydrogenase kinase 1 (PDK1) expression in tumor cells (110). Notably, PTPRG-AS1 has been revealed to regulate PDK1 expression by acting as a sponge for miR-599. Silencing PTPRG-AS1 has been shown to inhibit ESCC glycolysis, invasion and migration (110).

CircRNAs have been shown to regulate ESCC invasion and migration through their ability to act as a sponge for various types of miRNAs. For example, Shi et al (111) revealed that circLPAR3 upregulates MET gene expression and activates the RAS protein/MAPK and phosphoinositide 3-kinase/AKT pathways by acting as a sponge for miR-198. This promotes ESCC cell migration, invasion and metastasis. Meanwhile, Xu et al (112) demonstrated that circ0000654 promotes interleukin-6 secretion by acting as a sponge for miR-149-5p, which enhances ESCC cell migration by activating signal transducer and activator of transcription 3 (STAT3) signaling. This leads to malignant progression of the disease. Wang et al (113) demonstrated that circ0087378 correlates with poor overall survival (OS) in patients with ESCC and is highly expressed in ESCC cells. The authors observed a negative correlation between circ0087378 expression and miR-140-3p levels. Through binding to miR-140-3p, circ0087378 alleviates the suppressive effect of miR-140-3p on its target gene, E2F transcription factor 3 (E2F3). This circ0087378/miR-140-3p/E2F3 axis is crucial for promoting ESCC cell invasion and migration (114). The EMT facilitates tumor progression by altering cell-cell adhesion, polarity and morphology, which enhances tumor cell invasion and metastasis (114,115).

Numerous studies have demonstrated that, through affecting the EMT process, circRNAs may regulate the capacity of ESCC to invade and migrate. For instance, circLONP2 can promote EMT and lead to ESCC metastasis by regulating the miR-27b-3p/zinc finger E-box binding homeobox 1 axis (116). Chen et al (117) revealed that by reducing circ0004370, EMT-related protein expression can be decreased and ESCC cell invasion and migration can be prevented. Mechanically, collagen type I α 1 expression is upregulated by circ0004370 acting as a sponge for miR-1301-3p (117). Moreover, circARAP2 knockdown elevates E-Cadherin expression, while suppressing N-Cadherin expression. However, these effects are reversed when miR-761 is reduced or Forkhead Box M1 is upregulated (118). The downregulation of circARAP2 results in decreased expression of ESCC tumor stem cell-related genes and inhibits ESCC progression (118). Thus, targeting circRNAs in ESCC may represent a novel strategy to inhibit metastasis (118).

In summary, ncRNAs are pivotal regulators of ESCC metastasis, offering potential as therapeutic targets to curb tumor migration and recurrence. Further research is warranted to elucidate their precise mechanisms and optimize their utilization in controlling metastasis.

NcRNAs are associated with ESCC drug resistance

While chemotherapy and radiotherapy remain the primary treatment modalities for ESCC, frequent drug resistance is a major contributing factor to poor treatment outcomes. In recent years, several reports have indicated a significant association between ncRNAs and resistance to chemotherapy and radiotherapy in ESCC (119121).

Recent studies have shown that miRNAs are involved in regulating drug resistance in ESCC. For example, Zuo et al (122) demonstrated that overexpression of miR-153-3p inhibits the growth of ESCC cells and increases the level of cleaved caspase-3 after treatment with cisplatin. This indicates that upregulation of miR-153-3p may improve cisplatin resistance in ESCC cells. Previous studies have demonstrated that cancer-associated fibroblasts (CAFs) contribute to cisplatin resistance in ESCC (123,124). Another recent study further revealed the critical role of miRNAs in CAFs-induced cisplatin resistance (125,126). Exosomes secreted by CAFs, enriched with miR-21, inhibit phosphatase and tensin homolog (PTEN) activation in monocytes, which leads to STAT3 activation. This promotes monocyte differentiation into myeloid-derived suppressor cells, ultimately inducing cisplatin resistance (127). Gou et al (128) revealed that patients with ESCC with low miR-29b expression demonstrate increased resistance to radiotherapy. The authors identified a miR-29b binding site in the 3′-UTR of BTG anti-proliferation factor 2 (BTG2) mRNA. Downregulation of miR-29b increases BTG2 expression, decreases cyclin D1 expression and leads to cell cycle arrest in the G0/G1 phase of radiotherapy resistant ESCC cells (128). These findings suggest that targeting miRNAs could be a viable strategy to mitigate resistance to therapy in ESCC (128).

LncRNAs play a crucial role in drug resistance in ESCC. The ALKBH5-mediated m6A demethylation process upregulates lnc-cancer susceptibility 8 (lncCASC8) expression, which induces drug resistance in ESCC cells. LncCASC8 targets heterogeneous nuclear ribonucleoprotein L, preventing its ubiquitination and stabilizing its expression. This leads to increased Bcl2 production and decreased cleaved caspase-3 levels, ultimately resulting in ESCC drug resistance (129). The lnc-non-coding RNA activated by DNA damage (NORAD) is associated with cisplatin resistance and poor prognosis in patients with ESCC (130). Upregulation of lncNORAD increases the IC50 value of cisplatin and reduces apoptosis in ESCC cells treated with cisplatin. Mechanistically, lncNORAD acts as a sponge for miR-224-3p and represses miR-224-3p expression (130). Downregulation of miR-224-3p decreases the expression of its target gene, metadherin, and inhibits nuclear accumulation of β-catenin, which results in cisplatin resistance in ESCC (130). The lncPOU3F3 from tumor cell exosomes induces cisplatin resistance in ESCC by promoting the conversion of normal fibroblasts to CAFs (131). In addition, Wang et al (132) demonstrated that lnc-Taurine upregulated 1 (lncTUG1) knockdown, combined with radiotherapy, significantly inhibits ESCC cell growth, when compared to radiotherapy alone. Further investigation revealed that lncTUG1 acts as a sponge for miR-144-3p, promoting MET expression. Decreased lncTUG1 levels are associated with reduced MET and AKT phosphorylation, enhancing the efficacy of radiotherapy (132). These findings highlight the regulatory role of the lncTUG1/miR-144-3p/MET axis in ESCC radiotherapy resistance (132).

CircRNAs are also closely associated with ESCC drug resistance. The circ0014879 increases in radiotherapy resistant ESCC cells and controls the miR-519-3p/cell division cycle 25A axis to cause drug resistance (133). The upregulation of circ0000277 in cisplatin-resistant ESCC enhances drug resistance by modulating miR-873-5p and activating the SRY-box transcription factor 4/Wnt/β-catenin axis (134). Knockdown of circ0000277 promotes ESCC cell apoptosis and reduces cisplatin resistance, as evidenced by increased proapoptotic protein levels (PARP and caspase-3) and decreased IC50 values, following cisplatin treatment (134). Zhou et al (135) revealed that circ-glutamic-oxaloacetic transaminase 1 (circGOT1) is highly expressed in cisplatin-resistant ESCC cells, while miR-606 expression is reduced. This circRNA upregulates the expression of its host gene, GOT1, by acting as a sponge for miR-606. Inhibition of circGOT1 decreases ESCC cell glycolysis and enhances cisplatin sensitivity (135). These effects are reversed by miR-606 knockdown or GOT1 overexpression. It is hypothesized that circGOT1 promotes ESCC resistance via the circGOT1/miR-606/GOT1 axis (135).

Certain circRNAs, such as circDOPEY2, inhibit ESCC resistance and circDOPEY2 is expressed at low levels in cisplatin-resistant ESCC cells (136). It acts as a protein scaffold to enhance the binding between cytoplasmic polyadenylation element binding protein (CPEB4) and TRIM25 E3 ligase. This promotes the formation of the CPEB4/circDOPEY2/TRIM25 complex, accelerating CPEB4 degradation by promoting its ubiquitination and blocking CPEB4-mediated Mcl-1 translation (136). An in vivo study demonstrated that combining circDOPEY2 with cisplatin significantly suppresses cisplatin resistance (136). Zhu et al (137) indicated that circPSMC3 is downregulated in gefitinib-resistant ESCC cells. Increasing circPSMC3 expression significantly reduces ESCC resistance to gefitinib, inhibits cell proliferation and promotes apoptosis (137). A negative correlation has been found between circPSMC3 and miR-10a-5p expression, but circPSMC3 is positively correlated with PTEN expression, downstream of miR-10a-5p (137). Modulating miR-10a-5p or PTEN expression alters the effect of circPSMC3 on gefitinib resistance in ESCC. These findings suggest that circPSMC3 inhibits gefitinib resistance in ESCC by modulating the miR-10a-5p/PTEN axis (137). Programmed cell death receptor 1 (PD-1)/programmed cell death-ligand (PD-L1) inhibitors can act to slow tumor development, according to recent research (138). However, due to the emergence of resistance, only a small percentage of individuals benefit from this medication (139).

It has been demonstrated that circRNAs regulate drug resistance in PD-1/PD-L1 therapy (140,141). A study has reported elevated levels of circ-vimentin (circVIM) in ESCC cells and showed its ability to suppress miR-124 expression (142). Knockdown of circVIM in ESCC cells results in upregulation of miR-124 expression, a reduction in PD-L1 expression and a mitigation of tumor escape (142). Furthermore, silencing circVIM can be synergized with sevoflurane to inhibit multiple oncogenic activities and counter immune escape in ESCC by targeting the miR-124/PD-L1 axis (142). These findings suggest that circRNAs are an important regulator of drug resistance in ESCC therapy. If the efficacy and application plan can be further clarified, circRNA could have a high impact on the treatment of numerous patients and could reduce the recurrence rate of ESCC.

The aforementioned studies reveal a strong link between ncRNAs and tumor drug resistance, which highlights their potential in overcoming this challenge and guiding future therapeutic strategies.

NcRNAs as biomarkers in ESCC

Due to the subtle onset and rapid progression of ESCC, early detection and diagnosis are crucial for the treatment of this disease. Several studies have validated the use of ncRNAs as reliable biomarkers for the diagnosis of ESCC.

Researchers have shown that patients with ESCC with low miR-378 expression have a short OS and a poor prognosis. Low expression of miR-378 is correlated with TNM stage and LNM of patients (143). These findings indicate that miR-378 serves as an independent prognostic factor for the prediction of OS in patients diagnosed with ESCC (143). MiR-205-5-5p and miR-429 show substantial increases in plasma exosomes among patients with ESCC, while miR-375-3p exhibits a marked decrease (144).

Researchers have evaluated whether these miRNAs in plasma exosomes can be potential biomarkers for ESCC diagnosis using receiver operating characteristic (ROC) curves (144). It has been revealed that the area under the curve (AUC) of miR-205-5p, miR-429 and miR-375-3p are 0.770, 0.699 and 0.741, respectively (144). The sensitivities of miR-205-5-5p, miR-429 and miR-375-3p in diagnosing ESCC were 72.5, 60.0 and 65.0%, respectively, while the diagnostic specificities were 70.0, 60.0 and 65.0%, respectively (144). Wen et al (145) has identified miR-135b-5p, miR-139-5p, miR-29c-5p and miR-338-3p as potential predictors of OS in patients with ESCC with local metastasis, post surgical resection. These four miRNAs exhibit predictive capabilities that are comparable to established clinical prognostic indicators, such as pathological tumor-stage, pathological node-stage and lymph node examination count. Notably, the four-miRNA-based classifier has achieved an AUC of 0.597, underscoring its significance as a highly valuable diagnostic tool.

In addition, miRNAs in urine can be used for the diagnosis of ESCC. A study has revealed that the urine of patients contains elevated levels of certain miRNAs, such as miR-1273f, miR-619-5p, miR-150-3p, miR-4327 and miR-3135b. Urinary miR-1273f and miR-619-5p sensitivity and specificity for diagnosing all stages of ESCC were 82.5 and 59.0%, and 80.0 and 65.3%, respectively. These findings suggest that miRNAs have potential as biomarkers for ESCC (146).

LncRNAs have been shown to play a critical role in the diagnosis and prognosis of ESCC. Research suggests that the prognosis of patients with ESCC is inversely related to the risk score associated with an 8-lncRNA signature. This comprises ADAMTS9-AS1, DLX6-AS1, LINC00470, LINC00520, LINC01497, LINC01749, MAMDC2-AS1 and SSTR5-AS1 (147). In addition, six differentially expressed lncRNAs (6-DElncRNAs), namely AP000696.2, LINC01711, RP11-70C1.3, AP000487.5, AC011997.1 and RP11-225N10.1, have been identified as potential prognostic indicators for patients with ESCC. A 6-DElncRNA model has also been developed to classify patients with ESCC into high-risk and low-risk groups, with the former exhibiting lower OS rates. A ROC curve analysis showed that the AUCs for 6-DElncRNAs for predicting 1-, 3- and 5-year survival rates of patients with ESCC are 0.768, 0.825 and 0.822, respectively. As the high-risk group has higher gefitinib and lapatinib IC50 values, 6-DElncRNAs may also be able to predict drug susceptibility (148). Liu et al (149) demonstrated that a 6-lncRNA signature (AC005091.1, SNHG6, AC091544.4, DNAJB5-DT, HTT-AS and ANKRD10-IT1) has a high predictive value for ESCC. The ROC curve analysis gave AUCs of 0.785 and 0.786 for the 6-lncRNA signature for predicting 3- and 5-year survival rates, respectively. The AUCs of the 6-lncRNA signature for predicting 3- and 5-year recurrence-free survival are 0.795 and 0.783, respectively. These findings provide additional perspectives for the development and research of ESCC biomarkers.

Moreover, exosomal lncRNAs may also be used as ESCC biomarkers. Xie et al (150) demonstrated high expression of AC098818.2, LINC00958 and RASSF8-AS1 in ESCC cells, patient serum and exosomes. The diagnostic ability of these three lncRNAs for ESCC is 0.69, 0.72 and 0.78, respectively, in accordance with the ROC curve analysis. However, the diagnostic value ranges from 0.51 to 0.53 for common tumor biomarkers (CEA125, CA199, CA153, SCC, NSE and CYFRA21-1), which indicates that AC098818.2, LINC00958 and RASSF8-AS1 have certain advantages for the diagnosis of ESCC. The levels of these three lncRNAs also decrease in postoperative patients, which suggests that they have potential as dynamic monitoring indicators for tumor development.

There is growing evidence to suggest that circRNAs can serve as biomarkers for ESCC. Huang et al (151) revealed a substantial correlation between the level of circ0004771, vascular invasion and a worse OS in patients with ESCC. Furthermore, the authors observed that circ0004771 levels increase in an extracellular medium in a time-dependent manner and can be downregulated by the GW4869 exosome blocker, without affecting its expression within the cells. A ROC curve analysis provided an AUC for circ0004771 of 0.816, which indicates that circ0004771 has potential as a diagnostic biomarker for ESCC (151). Wang et al (152) identified 1,202 circRNAs related to ESCC prognosis through bioinformatics. The authors also revealed four circRNAs (circ0000005, circ0007541, circ0008199 and circ0077536) that are primarily found in the nucleus and can be used to divide patients with ESCC into two groups with noticeably different survival rates. This four-circRNA signature (AUC=0.839) performs better for ESCC prediction compared with using the TNM stage (AUC=0.657). When the four-circRNA signature and TNM are combined, the predictive value is higher (AUC=0.874).

Hu et al (153) showed that in individuals with ESCC, circGSK3β in plasma is positively linked with higher LNM and a poorer prognosis. The authors also demonstrated that the level of circGSK3β in the plasma may be a potential biomarker to detect ESCC and early stage ESCC, as indicated by the AUC values of 0.782 and 0.793, respectively. Moreover, diagnostic sensitivity is markedly increased by combining circGSK3β and carcinoembryonic antigen, which indicates that circGSK3β has potential as an early diagnostic marker for ESCC (153). Another study discovered that circADAMTS6 is positively associated with poor pathology grade, large tumors and LNM in patients (154). Patients who express higher levels of circADAMTS6 have a shorter OS compared with patients who express lower levels, which indicates that circADAMTS6 may be a potential prognostic factor for ESCC (154). Meanwhile, circSLC7A5 (155), circBMII (156) and circ0071106 (157) have also been shown to be potential biomarkers for ESCC. These results indicate that circRNAs may be employed as biomarkers for the early identification and prognosis of ESCC. CircRNAs are found in a range of tissues and cells and can withstand RNase R destruction, which makes them easy and cost-effective to detect. Thus, the use of circRNAs as biomarkers for ESCC could have significant clinical and economic benefits.

The aforementioned studies show the significant dysregulation of specific ncRNAs in ESCC, which facilitates early diagnosis. Given their widespread expression and easy detection, ncRNAs serve as promising and cost-effective biomarkers for ESCC.

Conclusions and future directions

Since the late 1990s to the present day, an increasing number of ncRNAs have been discovered. Initially, these non-coding fragments of RNA were considered to be useless byproducts (158). However, with the advent of RNA-sequencing technology, ncRNAs have gained growing attention and have emerged as a novel research paradigm within RNA biology (159). Furthermore, an expanding body of research has indicated that ncRNAs play pivotal roles in the onset and progression of various diseases, such as cancer, cardiovascular diseases and neurological disorders. This field of study has evolved into a crucial component of life sciences (160,161).

The present review provides an overview of the biogenesis, characteristics and functions of ncRNA family members, which include miRNAs, lncRNAs and circRNAs, along with the latest developments in the treatment of ESCC. The present study also summarizes, in Table I, studies on the regulation of ESCC progression by ncRNAs that are not detailed in this article. Aberrant expression of ncRNAs has been observed in ESCC cells, tissues and peripheral blood of patients with ESCC, which suggests their potential involvement in the disease and their utility as biomarkers (162). Furthermore, ncRNAs have been found to impact the progression of ESCC through multiple pathways, such as acting as molecular sponges for miRNA, participating in transcriptional regulation and influencing protein coding (163165). These findings underscore the potential of ncRNAs as biomarkers and therapeutic targets for ESCC, with implications for early diagnosis, treatment and prognosis assessment (162,166).

Table I.

Role of ncRNAs in esophageal squamous cell carcinomas.

Table I.

Role of ncRNAs in esophageal squamous cell carcinomas.

ncRNAsLocationExpressionTargetFunction(Refs.)
miR-3656--ACAP2Oncogene(181)
miR-154-5p-KIF14Anti-oncogene(182)
miR-149-RNF2Anti-oncogene(183)
miR-624-ARRDC3Oncogene(184)
miR-335-5p-TTKAnti-oncogene(185)
miR-942-5p--CST1Anti-oncogene(186)
miR-132-3p-KCNK2Oncogene(187)
miR-196b-SOCS2Oncogene(188)
miR-193a-3p-ALKBH5Oncogene(189)
miR-200a-KEAP1Oncogene(190)
TMPO-AS1-FUSOncogene(191)
PART1-miR-18a-5pAnti-oncogene(192)
LOC146880-miR-328-5pOncogene(193)
GATA2-AS1-miR-940Anti-oncogene(194)
KTN1-AS1Cytoplasm and nucleusRBBP4Oncogene(195)
LINC00886-ELF3Anti-oncogene(196)
BBOX1-AS1-miR-513a-3pOncogene(197)
RPL34-AS1CytoplasmmiR-575Anti-oncogene(198)
LINC00941-ILF2 and YBX1Oncogene(199)
NCK1-AS1-TGF-β1Oncogene(200)
CircRUNX1CytoplasmmiR-449b-5pOncogene(201)
CircFAM120BCytoplasmmiR-661Anti-oncogene(202)
Circ0003340CytoplasmmiR-940Oncogene(203)
Circ0023984CytoplasmmiR-1294Oncogene(204)
Circ0001093-miR-579-3pOncogene(205)
Circ0006948-miR-3612Oncogene(206)
CircFNDC3B-miR-214-3pOncogene(207)
Circ0005231CytoplasmmiR-383-5pOncogene(208)
Circ0003823CytoplasmmiR-607Oncogene(209)
Circ0021727CytoplasmmiR-23b-5pOncogene(210)

[i] ACAP2, arfGAP with coiled-coil, ankyrin repeat and PH domains 2; KIF14, kinesin family member 14; RNF2, ring finger protein 2; ARRDC3, arrestin domain-containing 3; TTK, Tyrosine threonine kinase; CST1, cysteine protease inhibitor 1; KCNK2, potassium channel subfamily K member 2; SOCS2, cytokine-signaling 2; ALKBH5, alkB Homolog 5, RNA demethylase; KEAP1, kelch-like ECH associated protein 1 FUS, fused in sarcoma; RBBP4, retinoblastoma-binding protein 4; ELF3, E74-like ETS transcription factor 3; ILF2, interleukin enhancer-binding factor 2; YBX1, Y-box-binding protein 1; TGF-β1, transforming growth factor beta 1; ↑, indicates upregulation; ↓, indicates downregulation; ncRNA, non-coding RNA; miR, microRNA.

Although the critical regulatory role of ncRNAs in the occurrence and progression of ESCC has received increasing attention, further in-depth research is still required. It is well known that ncRNAs exhibit rich diversity in terms of quantity and types. Future investigations should focus more on the interactions among ncRNAs, to elucidate their regulatory roles in ESCC progression by constructing and depicting ncRNA regulatory networks. In addition, the subcellular localization of ncRNAs is closely associated with their specific functions (167). Therefore, future studies should explore whether changes in the localization of ncRNAs during disease progression result in alterations to their structure, function and downstream target molecules. Most current studies focus primarily on the functional aspects of aberrantly expressed ncRNAs, in vivo, but lack investigation into the causes of their aberrant expression (168,169). Consequently, future research should further elucidate the specific mechanisms that underlie the aberrant expression of ncRNAs in cells or peripheral blood. Moreover, the occurrence and progression of various malignant tumors, such as ESCC, are closely related to the status of the tumor microenvironment. Therefore, subsequent research should not only focus on the role of ncRNAs in tumor cells but should also clarify their effects on the tumor microenvironment. For instance, it is crucial to elucidate the specific mechanisms by which ncRNAs, mediated by exosomes, enter other cells, as this leads to cellular dysfunction and disruption of the tumor microenvironment homeostasis.

Targeted therapies for ESCC have shown promising efficacy, with targeted drugs against key genes and signaling pathways implicated in ESCC now available. Given that ncRNAs can regulate the expression of downstream target genes through multiple mechanisms, future considerations may involve combining ncRNA-targeted therapy with existing targeted drugs for ESCC. This approach could involve the synergistic regulation of key genes in ESCC progression, such as Notch1, EGFR and human epidermal growth factor receptor 2 (170172). In addition, the regulatory effects of ncRNAs on multiple immune checkpoints have been confirmed and a potential combination of ncRNA-targeted therapy with immunotherapy could be trialed for ESCC in future studies (173,174). Increasing attention has been paid to the protein-coding ability of circRNAs. Peptide or protein-based drugs exhibit high specificity and efficiency in the clinical treatment of ESCC, which suggests that proteins encoded by circRNAs may serve as potential sources for screening therapeutic drugs for ESCC (175177). Indeed, studies have examined the potential of ncRNAs as target in the treatment of ESCC, such as miR-877-3p, miRNA-20b-5p and circFoxo3 (178180). However, the therapeutic efficacy of these ncRNA for ESCC in the clinic still requires in-depth study.

In summary, ncRNAs have potential as non-invasive diagnostic markers and specific therapeutic targets in clinical diagnosis and treatment. However, there is still a long way to go from basic research to clinical application. Extensive clinical studies are needed to demonstrate the safety of ncRNAs and gradually reduce the risks associated with their use as therapeutic targets, as well as to minimize treatment-related side effects. Moreover, continuous improvement and refinement of high-throughput sequencing technologies are necessary to ensure the specificity and integrity of ncRNA detection.

Acknowledgements

Not applicable.

Funding

This research was funded by the National Natural Science Foundation of China (grant nos. 32270848 and 82360044), the Collaborative Innovation Center of Chinese Ministry of Education (grant no. 2020-39), the Guizhou Provincial Natural Science Foundation [grant nos. ZK(2022)-607 and ZK(2024)-269], the Science and Technology Plan Project of Guizhou Province (grant no. 2020-032) and the Guizhou Provincial Traditional Chinese Medicine Project (grant no. QZYY-2022-027).

Availability of data and materials

Not applicable.

Authors' contributions

LZ, XW and ZH designed and organized this manuscript. LZ, YW and XZ contributed to the first draft of the manuscript. LZ, YW and MH designed the tables and figures. XW, JG and ZH reviewed the manuscript critically for important intellectual content. JG, MH and XZ revised the manuscript. YW, XZ and MH investigated and resolved any parts of the work appropriately. XW and ZH approved the final version to be published. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Rogers JE, Sewastjanow-Silva M, Waters RE and Ajani JA: Esophageal cancer: Emerging therapeutics. Expert Opin Ther Targets. 26:107–117. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Morgan E, Soerjomataram I, Rumgay H, Coleman HG, Thrift AP, Vignat J, Laversanne M, Ferlay J and Arnold M: The global landscape of esophageal squamous cell carcinoma and esophageal adenocarcinoma incidence and mortality in 2020 and projections to 2040: New estimates from GLOBOCAN 2020. Gastroenterology. 163:649–658.e2. 2022. View Article : Google Scholar : PubMed/NCBI

3 

He S, Xu J, Liu X and Zhen Y: Advances and challenges in the treatment of esophageal cancer. Acta Pharm Sin B. 11:3379–3392. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Rumgay H, Arnold M, Laversanne M, Whiteman DC, Thrift AP, Wei W, Lemmens VEPP and Soerjomataram I: International trends in esophageal squamous cell carcinoma and adenocarcinoma incidence. Am J Gastroenterol. 116:1072–1076. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Li J, Xu J, Zheng Y, Gao Y, He S, Li H, Zou K, Li N, Tian J, Chen W and He J: Esophageal cancer: Epidemiology, risk factors and screening. Chin J Cancer Res. 33:535–547. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Bhat AA, Nisar S, Maacha S, Carneiro-Lobo TC, Akhtar S, Siveen KS, Wani NA, Rizwan A, Bagga P, Singh M, et al: Cytokine-chemokine network driven metastasis in esophageal cancer; promising avenue for targeted therapy. Mol Cancer. 20:22021. View Article : Google Scholar : PubMed/NCBI

7 

Huang FL and Yu SJ: Esophageal cancer: Risk factors, genetic association, and treatment. Asian J Surg. 41:210–215. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Mattick JS and Makunin IV: Non-coding RNA. Hum Mol Genet. 15:R17–R29. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Liu Y, Liu X, Lin C, Jia X, Zhu H, Song J and Zhang Y: Noncoding RNAs regulate alternative splicing in cancer. J Exp Clin Cancer Res. 40:112021. View Article : Google Scholar : PubMed/NCBI

10 

Mohapatra S, Pioppini C, Ozpolat B and Calin GA: Non-coding RNAs regulation of macrophage polarization in cancer. Mol Cancer. 20:242021. View Article : Google Scholar : PubMed/NCBI

11 

Poller W, Sahoo S, Hajjar R, Landmesser U and Krichevsky AM: Exploration of the noncoding genome for human-specific therapeutic targets-recent insights at molecular and cellular level. Cells. 12:26602023. View Article : Google Scholar : PubMed/NCBI

12 

Beermann J, Piccoli MT, Viereck J and Thum T: Non-coding RNAs in development and disease: Background, mechanisms, and therapeutic approaches. Physiol Rev. 96:1297–1325. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Zhou B, Yang H, Yang C, Bao YL, Yang SM, Liu J and Xiao YF: Translation of noncoding RNAs and cancer. Cancer Lett. 497:89–99. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Slack FJ and Chinnaiyan AM: The role of non-coding RNAs in oncology. Cell. 179:1033–1055. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Ran H, Yang Y, Luo M, Liu X, Yue B, Chai Z, Zhong J and Wang H: Molecular regulation of yak preadipocyte differentiation and proliferation by LncFAM200B and ceRNA regulatory network analysis. Cells. 11:23662022. View Article : Google Scholar : PubMed/NCBI

16 

Min X, Cai MY, Shao T, Xu ZY, Liao Z, Liu DL, Zhou MY, Wu WP, Zhou YL, Mo MH, et al: A circular intronic RNA ciPVT1 delays endothelial cell senescence by regulating the miR-24-3p/CDK4/pRb axis. Aging Cell. 21:e135292022. View Article : Google Scholar : PubMed/NCBI

17 

Qu J, Xiong X, Hujie G, Ren J, Yan L and Ma L: MicroRNA-132-3p alleviates neuron apoptosis and impairments of learning and memory abilities in Alzheimer's disease by downregulation of HNRNPU stabilized BACE1. Cell Cycle. 20:2309–2320. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Liu CX, Guo SK, Nan F, Xu YF, Yang L and Chen LL: RNA circles with minimized immunogenicity as potent PKR inhibitors. Mol Cell. 82:420–434.e6. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Feng Q, Zhang H, Yao D, Chen WD and Wang YD: Emerging role of non-coding RNAs in esophageal squamous cell carcinoma. Int J Mol Sci. 21:2582019. View Article : Google Scholar : PubMed/NCBI

20 

Miyoshi J, Zhu Z, Luo A, Toden S, Zhou X, Izumi D, Kanda M, Takayama T, Parker IM, Wang M, et al: A microRNA-based liquid biopsy signature for the early detection of esophageal squamous cell carcinoma: A retrospective, prospective and multicenter study. Mol Cancer. 21:442022. View Article : Google Scholar : PubMed/NCBI

21 

Sharma U, Murmu M, Barwal TS, Tuli HS, Jain M, Prakash H, Kaceli T, Jain A and 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. 29:2326–2349. 2022. View Article : Google Scholar : PubMed/NCBI

22 

Wang C, Liu WR, Tan S, Zhou JK, Xu X, Ming Y, Cheng J, Li J, Zeng Z, Zuo Y, et al: Characterization of distinct circular RNA signatures in solid tumors. Mol Cancer. 21:632022. View Article : Google Scholar : PubMed/NCBI

23 

Jansson MD and Lund AH: MicroRNA and cancer. Mol Oncol. 6:590–610. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Acunzo M and Croce CM: MicroRNA in cancer and cachexia-a mini-review. J Infect Dis. 212 (Suppl 1):S74–S77. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Mengistu AA and Tenkegna TA: The role of miRNA in plant-virus interaction: A review. Mol Biol Rep. 48:2853–2861. 2021. View Article : Google Scholar : PubMed/NCBI

26 

Peng Y and Croce CM: The role of MicroRNAs in human cancer. Signal Transduct Target Ther. 1:150042016. View Article : Google Scholar : PubMed/NCBI

27 

Wahid F, Shehzad A, Khan T and Kim YY: MicroRNAs: Synthesis, mechanism, function, and recent clinical trials. Biochim Biophys Acta. 1803:1231–1243. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Budakoti M, Panwar AS, Molpa D, Singh RK, Büsselberg D, Mishra AP, Coutinho HDM and Nigam M: Micro-RNA: The darkhorse of cancer. Cell Signal. 83:1099952021. View Article : Google Scholar : PubMed/NCBI

29 

Krol J, Loedige I and Filipowicz W: The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet. 11:597–610. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Wilson RC, Tambe A, Kidwell MA, Noland CL, Schneider CP and Doudna JA: Dicer-TRBP complex formation ensures accurate mammalian microRNA biogenesis. Mol Cell. 57:397–407. 2015. View Article : Google Scholar : PubMed/NCBI

31 

He B, Zhao Z, Cai Q, Zhang Y, Zhang P, Shi S, Xie H, Peng X, Yin W, Tao Y and Wang X: miRNA-based biomarkers, therapies, and resistance in cancer. Int J Biol Sci. 16:2628–2647. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Ha M and Kim VN: Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 15:509–524. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Ho PTB, Clark IM and Le LTT: MicroRNA-based diagnosis and therapy. Int J Mol Sci. 23:71972022. View Article : Google Scholar

34 

Iqbal MA, Arora S, Prakasam G, Calin GA and Syed MA: MicroRNA in lung cancer: Role, mechanisms, pathways and therapeutic relevance. Mol Aspects Med. 70:3–20. 2019. View Article : Google Scholar : PubMed/NCBI

35 

O'Brien J, Hayder H, Zayed Y and Peng C: Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol (Lausanne). 9:4022018. View Article : Google Scholar : PubMed/NCBI

36 

Medley JC, Panzade G and Zinovyeva AY: microRNA strand selection: Unwinding the rules. Wiley Interdiscip Rev RNA. 12:e16272021. View Article : Google Scholar : PubMed/NCBI

37 

Bartel DP: Metazoan MicroRNAs. Cell. 173:20–51. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Sun Q, Hao Q and Prasanth KV: Nuclear long noncoding RNAs: Key regulators of gene expression. Trends Genet. 34:142–157. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Cao Q, Guo Z, Yan Y, Wu J and Song C: Exosomal long noncoding RNAs in aging and age-related diseases. IUBMB Life. 71:1846–1856. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Kour S and Rath PC: Long noncoding RNAs in aging and age-related diseases. Ageing Res Rev. 26:1–21. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Statello L, Guo CJ, Chen LL and Huarte M: Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 22:96–118. 2021. View Article : Google Scholar : PubMed/NCBI

42 

He Z, Yang D, Fan X, Zhang M, Li Y, Gu X and Yang M: The roles and mechanisms of lncRNAs in liver fibrosis. Int J Mol Sci. 21:14822020. View Article : Google Scholar : PubMed/NCBI

43 

Aboudehen K: Regulation of mTOR signaling by long non-coding RNA. Biochim Biophys Acta Gene Regul Mech. 1863:1944492020. View Article : Google Scholar : PubMed/NCBI

44 

Jain AK, Xi Y, McCarthy R, Allton K, Akdemir KC, Patel LR, Aronow B, Lin C, Li W, Yang L, et al: LncPRESS1 Is a p53-regulated LncRNA that safeguards pluripotency by disrupting SIRT6-mediated de-acetylation of histone H3K56. Mol Cell. 64:967–981. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Du Z, Wen X, Wang Y, Jia L, Zhang S, Liu Y, Zhou L, Li H, Yang W, Wang C, et al: Chromatin lncRNA Platr10 controls stem cell pluripotency by coordinating an intrachromosomal regulatory network. Genome Biol. 22:2332021. View Article : Google Scholar : PubMed/NCBI

46 

Zhang Y, Huang YX, Wang DL, Yang B, Yan HY, Lin LH, Li Y, Chen J, Xie LM, Huang YS, et al: LncRNA DSCAM-AS1 interacts with YBX1 to promote cancer progression by forming a positive feedback loop that activates FOXA1 transcription network. Theranostics. 10:10823–10837. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Su K, Wang N, Shao Q, Liu H, Zhao B and Ma S: The role of a ceRNA regulatory network based on lncRNA MALAT1 site in cancer progression. Biomed Pharmacother. 137:1113892021. View Article : Google Scholar : PubMed/NCBI

48 

Huang W, Sun YM, Pan Q, Fang K, Chen XT, Zeng ZC, Chen TQ, Zhu SX, Huang LB, Luo XQ, et al: The snoRNA-like lncRNA LNC-SNO49AB drives leukemia by activating the RNA-editing enzyme ADAR1. Cell Discov. 8:1172022. View Article : Google Scholar : PubMed/NCBI

49 

Yang X, Ye T, Liu H, Lv P, Duan C, Wu X, Jiang K, Lu H, Xia D, Peng E, et al: Expression profiles, biological functions and clinical significance of circRNAs in bladder cancer. Mol Cancer. 20:42021. View Article : Google Scholar : PubMed/NCBI

50 

Hu Q and Zhou T: EIciRNA-mediated gene expression: Tunability and bimodality. FEBS Lett. 592:3460–3471. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Zhou X and Du J: CircRNAs: Novel therapeutic targets in multiple myeloma. Mol Biol Rep. 49:10667–10676. 2022. View Article : Google Scholar : PubMed/NCBI

52 

Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al: Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 495:333–338. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Vahabi A, Rezaie J, Hassanpour M, Panahi Y and Nemati M, Rasmi Y and Nemati M: Tumor cells-derived exosomal CircRNAs: Novel cancer drivers, molecular mechanisms, and clinical opportunities. Biochem Pharmacol. 200:1150382022. View Article : Google Scholar : PubMed/NCBI

54 

Amicone L, Marchetti A and Cicchini C: Exosome-associated circRNAs as key regulators of EMT in cancer. Cells. 11:17162022. View Article : Google Scholar : PubMed/NCBI

55 

Guo X, Chang X, Wang Z, Jiang C and Wei Z: CircRNAs: Promising factors for regulating angiogenesis in colorectal cancer. Clin Transl Oncol. 24:1673–1681. 2022. View Article : Google Scholar : PubMed/NCBI

56 

Li X, Yang L and Chen LL: The biogenesis, functions, and challenges of circular RNAs. Mol Cell. 71:428–442. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK and Kjems J: Natural RNA circles function as efficient microRNA sponges. Nature. 495:384–388. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Zhang LX, Gao J, Long X, Zhang PF, Yang X, Zhu SQ, Pei X, Qiu BQ, Chen SW, Lu F, et al: The circular RNA circHMGB2 drives immunosuppression and anti-PD-1 resistance in lung adenocarcinomas and squamous cell carcinomas via the miR-181a-5p/CARM1 axis. Mol Cancer. 21:1102022. View Article : Google Scholar : PubMed/NCBI

59 

Hollensen AK, Thomsen HS, Lloret-Llinares M, Kamstrup AB, Jensen JM, Luckmann M, Birkmose N, Palmfeldt J, Jensen TH, Hansen TB and Damgaard CK: circZNF827 nucleates a transcription inhibitory complex to balance neuronal differentiation. Elife. 9:e584782020. View Article : Google Scholar : PubMed/NCBI

60 

Misir S, Wu N and Yang BB: Specific expression and functions of circular RNAs. Cell Death Differ. 29:481–491. 2022. View Article : Google Scholar : PubMed/NCBI

61 

Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, et al: Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 22:256–264. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Wang X, Ma R, Zhang X, Cui L, Ding Y, Shi W, Guo C and Shi Y: Crosstalk between N6-methyladenosine modification and circular RNAs: Current understanding and future directions. Mol Cancer. 20:1212021. View Article : Google Scholar : PubMed/NCBI

63 

Lin H, Wang Y, Wang P, Long F and Wang T: Mutual regulation between N6-methyladenosine (m6A) modification and circular RNAs in cancer: Impacts on therapeutic resistance. Mol Cancer. 21:1482022. View Article : Google Scholar : PubMed/NCBI

64 

Meyer KD, Patil DP, Zhou J, Zinoviev A, Skabkin MA, Elemento O, Pestova TV, Qian SB and Jaffrey SR: 5′ UTR m(6)A promotes cap-independent translation. Cell. 163:999–1010. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Li R, Jiang J, Shi H, Qian H, Zhang X and Xu W: CircRNA: A rising star in gastric cancer. Cell Mol Life Sci. 77:1661–1680. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Yang Y, Fan X, Mao M, Song X, Wu P, Zhang Y, Jin Y, Yang Y, Chen LL, Wang Y, et al: Extensive translation of circular RNAs driven by N6-methyladenosine. Cell Res. 27:626–641. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Duan JL, Chen W, Xie JJ, Zhang ML, Nie RC, Liang H, Mei J, Han K, Xiang ZC, Wang FW, et al: A novel peptide encoded by N6-methyladenosine modified circMAP3K4 prevents apoptosis in hepatocellular carcinoma. Mol Cancer. 21:932022. View Article : Google Scholar : PubMed/NCBI

68 

Lei M, Zheng G, Ning Q, Zheng J and Dong D: Translation and functional roles of circular RNAs in human cancer. Mol Cancer. 19:302020. View Article : Google Scholar : PubMed/NCBI

69 

Song Y, Li L, Ou Y, Gao Z, Li E, Li X, Zhang W, Wang J, Xu L, Zhou Y, et al: Identification of genomic alterations in oesophageal squamous cell cancer. Nature. 509:91–95. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Yu XM, Li SJ, Yao ZT, Xu JJ, Zheng CC, Liu ZC, Ding PB, Jiang ZL, Wei X, Zhao LP, et al: N4-acetylcytidine modification of lncRNA CTC-490G23.2 promotes cancer metastasis through interacting with PTBP1 to increase CD44 alternative splicing. Oncogene. 42:1101–1116. 2023. View Article : Google Scholar : PubMed/NCBI

71 

Huang X, Liu C, Li H, Dai T, Luo G, Zhang C, Li T and Lü M: Hypoxia-responsive lncRNA G077640 promotes ESCC tumorigenesis via the H2AX-HIF1α-glycolysis axis. Carcinogenesis. 44:383–393. 2023. View Article : Google Scholar : PubMed/NCBI

72 

Xiao Z, Feng X, Zhou Y, Li P, Luo J, Zhang W, Zhou J, Zhao J, Wang D, Wang Y, et al: Exosomal miR-10527-5p inhibits migration, invasion, lymphangiogenesis and lymphatic metastasis by affecting Wnt/β-catenin signaling via Rab10 in esophageal squamous cell carcinoma. Int J Nanomedicine. 18:95–114. 2023. View Article : Google Scholar : PubMed/NCBI

73 

Cui Y, Zhang C, Ma S, Li Z, Wang W, Li Y, Ma Y, Fang J, Wang Y, Cao W and Guan F: RNA m6A demethylase FTO-mediated epigenetic up-regulation of LINC00022 promotes tumorigenesis in esophageal squamous cell carcinoma. J Exp Clin Cancer Res. 40:2942021. View Article : Google Scholar : PubMed/NCBI

74 

Ren K, Li Y, Lu H, Li Z, Li Z, Wu K, Li Z and Han X: Long noncoding RNA HOTAIR controls cell cycle by functioning as a competing endogenous RNA in esophageal squamous cell carcinoma. Transl Oncol. 9:489–497. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Xu T, Hu Y, Zhao Y, Qi Y, Zhang S and Li P: Hsa_circ_0046534 accelerates esophageal squamous cell carcinoma proliferation and metastasis via regulating MMP2 expression by sponging miR-339-5p. Cell Signal. 112:1109062023. View Article : Google Scholar : PubMed/NCBI

76 

Xian D, Yang S, Liu Y, Liu Q, Huang D and Wu Y: MicroRNA-196a-5p facilitates the onset and progression via targeting ITM2B in esophageal squamous cell carcinoma. Pathol Int. 74:129–138. 2024. View Article : Google Scholar : PubMed/NCBI

77 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Zhang Y, Dong X, Guo X, Li C, Fan Y, Liu P, Yuan D, Ma X, Wang J, Zheng J, et al: LncRNA-BC069792 suppresses tumor progression by targeting KCNQ4 in breast cancer. Mol Cancer. 22:412023. View Article : Google Scholar : PubMed/NCBI

79 

Liu QL, Zhang Z, Wei X and Zhou ZG: Noncoding RNAs in tumor metastasis: Molecular and clinical perspectives. Cell Mol Life Sci. 78:6823–6850. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Enkhnaran B, Zhang GC, Zhang NP, Liu HN, Wu H, Xuan S, Yu XN, Song GQ, Shen XZ, Zhu JM, et al: microRNA-106b-5p promotes cell growth and sensitizes chemosensitivity to sorafenib by targeting the BTG3/Bcl-xL/p27 signaling pathway in hepatocellular carcinoma. J Oncol. 2022:19715592022. View Article : Google Scholar : PubMed/NCBI

81 

Wang H, Peng D, Gan M, He Z and Kuang Y: CPEB3 overexpression caused by miR-106b-5p inhibition inhibits esophageal carcinoma in-vitro progression and metastasis. Anticancer Drugs. 33:335–351. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Yang F, Sun Z, Wang D and Du T: MiR-106b-5p regulates esophageal squamous cell carcinoma progression by binding to HPGD. BMC Cancer. 22:3082022. View Article : Google Scholar : PubMed/NCBI

83 

Deng W, Fan W, Li P, Yao J, Qi J, Chi H, Ji G and Zhao J: microRNA-497-mediated Smurf2/YY1/HIF2α axis in tumor growth and metastasis of esophageal squamous cell carcinoma. J Biochem Mol Toxicol. 36:e231822022. View Article : Google Scholar : PubMed/NCBI

84 

He Z, Chen J, Chen X, Wang H, Tang L and Han C: microRNA-377 acts as a suppressor in esophageal squamous cell carcinoma through CBX3-dependent P53/P21 pathway. J Cell Physiol. 236:107–120. 2021. View Article : Google Scholar : PubMed/NCBI

85 

Ren L, Fang X, Shrestha SM, Ji Q, Ye H, Liang Y, Liu Y, Feng Y, Dong J and Shi R: LncRNA SNHG16 promotes development of oesophageal squamous cell carcinoma by interacting with EIF4A3 and modulating RhoU mRNA stability. Cell Mol Biol Lett. 27:892022. View Article : Google Scholar : PubMed/NCBI

86 

Yang X, Zeng T, Liu Z, He W, Hu M, Tang T, Chen L and Xing L: Long noncoding RNA GK-IT1 promotes esophageal squamous cell carcinoma by regulating MAPK1 phosphorylation. Cancer Med. 11:4555–4574. 2022. View Article : Google Scholar : PubMed/NCBI

87 

Xue ST, Zheng B, Cao SQ, Ding JC, Hu GS, Liu W and Chen C: Long non-coding RNA LINC00680 functions as a ceRNA to promote esophageal squamous cell carcinoma progression through the miR-423-5p/PAK6 axis. Mol Cancer. 21:692022. View Article : Google Scholar : PubMed/NCBI

88 

Xiong G, Diao D, Lu D, Liu X, Liu Z, Mai S, Feng S, Dong X and Cai K: Circular RNA circNELL2 acts as the sponge of miR-127-5p to promote esophageal squamous cell carcinoma progression. Onco Targets Ther. 13:9245–9255. 2020. View Article : Google Scholar : PubMed/NCBI

89 

Li Z, Qin X, Bian W, Li Y, Shan B, Yao Z and Li S: Exosomal lncRNA ZFAS1 regulates esophageal squamous cell carcinoma cell proliferation, invasion, migration and apoptosis via microRNA-124/STAT3 axis. J Exp Clin Cancer Res. 38:4772019. View Article : Google Scholar : PubMed/NCBI

90 

Tian Z, Li Z, Zhu Y, Meng L, Liu F, Sang M and Wang G: Hypermethylation-mediated inactivation of miR-124 predicts poor prognosis and promotes tumor growth at least partially through targeting EZH2/H3K27me3 in ESCC. Clin Exp Metastasis. 36:381–391. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Zeng B, Liu Z, Zhu H, Zhang X, Yang W, Li X and Cheng C: CircRNA_2646 functions as a ceRNA to promote progression of esophageal squamous cell carcinoma via inhibiting miR-124/PLP2 signaling pathway. Cell Death Discov. 7:992021. View Article : Google Scholar : PubMed/NCBI

92 

Yao D, Lin S, Chen S and Wang Z: circHIPK3 regulates cell proliferation and migration by sponging microRNA-124 and regulating serine/threonine kinase 3 expression in esophageal squamous cell carcinoma. Bioengineered. 13:9767–9780. 2022. View Article : Google Scholar : PubMed/NCBI

93 

Cheng J, Ma H, Yan M, Zhang Z and Xing W: Circ_0007624 suppresses the development of esophageal squamous cell carcinoma via targeting miR-224-5p/CPEB3 to inactivate the EGFR/PI3K/AKT signaling. Cell Signal. 99:1104482022. View Article : Google Scholar : PubMed/NCBI

94 

Chen Z, Yao N, Gu H, Song Y, Ye Z, Li L, Lu P and Shao Q: Circular RNA_LARP4 sponges miR-1323 and hampers progression of esophageal squamous cell carcinoma through modulating PTEN/PI3K/AKT pathway. Dig Dis Sci. 65:2272–2283. 2020. View Article : Google Scholar : PubMed/NCBI

95 

Jiang Q, Wang H, Yuan D, Qian X, Ma X, Yan M and Xing W: Circular_0086414 induces SPARC like 1 (SPARCL1) production to inhibit esophageal cancer cell proliferation, invasion and glycolysis and induce cell apoptosis by sponging miR-1290. Bioengineered. 13:12099–12114. 2022. View Article : Google Scholar : PubMed/NCBI

96 

Zheng S, Liu B and Guan X: The role of tumor microenvironment in invasion and metastasis of esophageal squamous cell carcinoma. Front Oncol. 12:9112852022. View Article : Google Scholar : PubMed/NCBI

97 

Wang X, Han J, Liu Y, Hu J, Li M, Chen X and Xu L: miR-17-5p and miR-4443 promote esophageal squamous cell carcinoma development by targeting TIMP2. Front Oncol. 11:6058942021. View Article : Google Scholar : PubMed/NCBI

98 

Liang Y, Duan L, Lu J and Xia J: Engineering exosomes for targeted drug delivery. Theranostics. 11:3183–3195. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Paskeh MDA, Entezari M, Mirzaei S, Zabolian A, Saleki H, Naghdi MJ, Sabet S, Khoshbakht MA, Hashemi M, Hushmandi K, et al: Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol. 15:832022. View Article : Google Scholar : PubMed/NCBI

100 

Liu T, Li P, Li J, Qi Q, Sun Z, Shi S, Xie Y, Liu S, Wang Y, Du L and Wang C: Exosomal and intracellular miR-320b promotes lymphatic metastasis in esophageal squamous cell carcinoma. Mol Ther Oncolytics. 23:163–180. 2021. View Article : Google Scholar : PubMed/NCBI

101 

Cui D, Zhu Y, Yan D, Lee NPY, Han L, Law S, Tsao GSW and Cheung ALM: Dual inhibition of cMET and EGFR by microRNA-338-5p suppresses metastasis of esophageal squamous cell carcinoma. Carcinogenesis. 42:995–1007. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Zhao R, Shan Y, Zhou X, Zhang C, Zhao R, Zhao L and Shan B: MicroRNA-485-5p suppresses the progression of esophageal squamous cell carcinoma by targeting flotillin-1 and inhibits the epithelial-mesenchymal transition. Oncol Rep. 45:932021. View Article : Google Scholar : PubMed/NCBI

103 

Cheng W, Yang F and Ma Y: lncRNA TPT1-AS1 promotes cell migration and invasion in esophageal squamous-cell carcinomas by regulating the miR-26a/HMGA1 axis. Open Med (Wars). 18:202205332023. View Article : Google Scholar : PubMed/NCBI

104 

Yang R, Wan J, Ma L, Zhou F, Yang Z, Li Z, Zhang M and Ming L: TMEM44-AS1 promotes esophageal squamous cell carcinoma progression by regulating the IGF2BP2-GPX4 axis in modulating ferroptosis. Cell Death Discov. 9:4312023. View Article : Google Scholar : PubMed/NCBI

105 

Wang J, Huang TJ, Mei Y, Luo FF, Xie DH, Peng LX, Liu BQ, Fan ML, Zhang JB, Zheng ST, et al: Novel long noncoding RNA LINC02820 augments TNF signaling pathway to remodel cytoskeleton and potentiate metastasis in esophageal squamous cell carcinoma. Cancer Gene Ther. 30:375–387. 2023. View Article : Google Scholar : PubMed/NCBI

106 

Li H, Lin PH, Gupta P, Li X, Zhao SL, Zhou X, Li Z, Wei S, Xu L, Han R, et al: MG53 suppresses tumor progression and stress granule formation by modulating G3BP2 activity in non-small cell lung cancer. Mol Cancer. 20:1182021. View Article : Google Scholar : PubMed/NCBI

107 

Zhang Y, Yue C, Krichevsky AM and Garkavtsev I: Repression of the stress granule protein G3BP2 inhibits immune checkpoint molecule PD-L1. Mol Oncol. Feb 1–2021.(Epub ahead of print).

108 

Zheng Y, Wu J, Deng R, Lin C, Huang Y, Yang X, Wang C, Yang M, He Y, Lu J, et al: G3BP2 regulated by the lncRNA LINC01554 facilitates esophageal squamous cell carcinoma metastasis through stabilizing HDGF transcript. Oncogene. 41:515–526. 2022. View Article : Google Scholar : PubMed/NCBI

109 

Zhang H, Wang Y, Zhang W, Wu Q, Fan J and Zhan Q: BAALC-AS1/G3BP2/c-Myc feedback loop promotes cell proliferation in esophageal squamous cell carcinoma. Cancer Commun (Lond). 41:240–257. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Wu K, Wang Z, Huang Y, Yao L, Kang N, Ge W, Zhang R and He W: LncRNA PTPRG-AS1 facilitates glycolysis and stemness properties of esophageal squamous cell carcinoma cells through miR-599/PDK1 axis. J Gastroenterol Hepatol. 37:507–517. 2022. View Article : Google Scholar : PubMed/NCBI

111 

Shi Y, Fang N, Li Y, Guo Z, Jiang W, He Y, Ma Z and Chen Y: Circular RNA LPAR3 sponges microRNA-198 to facilitate esophageal cancer migration, invasion, and metastasis. Cancer Sci. 111:2824–2836. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Xu Z, Tie X, Li N, Yi Z, Shen F and Zhang Y: Circular RNA hsa_circ_0000654 promotes esophageal squamous cell carcinoma progression by regulating the miR-149-5p/IL-6/STAT3 pathway. IUBMB Life. 72:426–439. 2020. View Article : Google Scholar : PubMed/NCBI

113 

Wang J, Wang Q, Gong Y, Hu Q, Zhang H, Ke S and Chen Y: Knockdown of circRNA circ_0087378 represses the tumorigenesis and progression of esophageal squamous cell carcinoma through modulating the miR-140-3p/E2F3 axis. Front Oncol. 10:6072312021. View Article : Google Scholar : PubMed/NCBI

114 

Brown MS, Muller KE and Pattabiraman DR: Quantifying the epithelial-to-mesenchymal transition (EMT) from bench to bedside. Cancers (Basel). 14:11382022. View Article : Google Scholar : PubMed/NCBI

115 

Huang Y, Hong W and Wei X: The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 15:1292022. View Article : Google Scholar : PubMed/NCBI

116 

Zhu C, Bi W, Li H and Wang W: CircLONP2 accelerates esophageal squamous cell carcinoma progression via direct MiR-27b-3p-ZEB1 axis. Front Oncol. 12:8228392022. View Article : Google Scholar : PubMed/NCBI

117 

Chen X, Sun H, Zhao Y, Zhang J, Xiong G, Cui Y and Lei C: CircRNA circ_0004370 promotes cell proliferation, migration, and invasion and inhibits cell apoptosis of esophageal cancer via miR-1301-3p/COL1A1 axis. Open Med (Wars). 16:104–116. 2021. View Article : Google Scholar : PubMed/NCBI

118 

Xu P, Wang L, Liu Q, Gao P, Hu F, Xie X, Jiang L, Bi R, Ding F, Yang Q and Xiao H: The abnormal expression of circ-ARAP2 promotes ESCC progression through regulating miR-761/FOXM1 axis-mediated stemness and the endothelial-mesenchymal transition. J Transl Med. 20:3182022. View Article : Google Scholar : PubMed/NCBI

119 

Song B, Liu X, Dong H and Roy R: miR-140-3P induces chemotherapy resistance in esophageal carcinoma by targeting the NFYA-MDR1 axis. Appl Biochem Biotechnol. 195:973–991. 2023. View Article : Google Scholar : PubMed/NCBI

120 

Zhao F, Tian H, Wang Y, Zhang J, Liu F and Fu L: LINC01004-SPI1 axis-activated SIGLEC9 in tumor-associated macrophages induces radioresistance and the formation of immunosuppressive tumor microenvironment in esophageal squamous cell carcinoma. Cancer Immunol Immunother. 72:1835–1851. 2023. View Article : Google Scholar : PubMed/NCBI

121 

Wu J, Liu Y, Huang X, Cheng Y, Qian Z, Ni X, Chen S, Lin M and Luo J: LncRNA DGCR5 silencing enhances the radio-sensitivity of human esophageal squamous cell carcinoma via negatively regulating the Warburg effect. Radiat Res. 199:264–272. 2023. View Article : Google Scholar : PubMed/NCBI

122 

Zuo J, Zhao M, Fan Z, Liu B, Wang Y, Li Y, Lv P, Xing L, Zhang X and Shen H: MicroRNA-153-3p regulates cell proliferation and cisplatin resistance via Nrf-2 in esophageal squamous cell carcinoma. Thorac Cancer. 11:738–747. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Qiao Y, Zhang C, Li A, Wang D, Luo Z, Ping Y, Zhou B, Liu S, Li H, Yue D, et al: IL6 derived from cancer-associated fibroblasts promotes chemoresistance via CXCR7 in esophageal squamous cell carcinoma. Oncogene. 37:873–883. 2018. View Article : Google Scholar : PubMed/NCBI

124 

Che Y, Wang J, Li Y, Lu Z, Huang J, Sun S, Mao S, Lei Y, Zang R, Sun N and He L: Cisplatin-activated PAI-1 secretion in the cancer-associated fibroblasts with paracrine effects promoting esophageal squamous cell carcinoma progression and causing chemoresistance. Cell Death Dis. 9:7592018. View Article : Google Scholar : PubMed/NCBI

125 

Zhuang J, Shen L, Li M, Sun J, Hao J, Li J, Zhu Z, Ge S, Zhang D, Guo H, et al: Cancer-associated fibroblast-derived miR-146a-5p generates a niche that promotes bladder cancer stemness and chemoresistance. Cancer Res. 83:1611–1627. 2023. View Article : Google Scholar : PubMed/NCBI

126 

Kang SH, Oh SY, Lee KY, Lee HJ, Kim MS, Kwon TG, Kim JW, Lee ST, Choi SY and Hong SH: Differential effect of cancer-associated fibroblast-derived extracellular vesicles on cisplatin resistance in oral squamous cell carcinoma via miR-876-3p. Theranostics. 14:460–479. 2024. View Article : Google Scholar : PubMed/NCBI

127 

Zhao Q, Huang L, Qin G, Qiao Y, Ren F, Shen C, Wang S, Liu S, Lian J, Wang D, et al: Cancer-associated fibroblasts induce monocytic myeloid-derived suppressor cell generation via IL-6/exosomal miR-21-activated STAT3 signaling to promote cisplatin resistance in esophageal squamous cell carcinoma. Cancer Lett. 518:35–48. 2021. View Article : Google Scholar : PubMed/NCBI

128 

Guo D, Jin J, Liu J, Dong X, Li D and He Y: MicroRNA-29b regulates the radiosensitivity of esophageal squamous cell carcinoma by regulating the BTG2-mediated cell cycle. Strahlenther Onkol. 197:829–835. 2021. View Article : Google Scholar : PubMed/NCBI

129 

Wu Q, Zhang H, Yang D, Min Q, Wang Y, Zhang W and Zhan Q: The m6A-induced lncRNA CASC8 promotes proliferation and chemoresistance via upregulation of hnRNPL in esophageal squamous cell carcinoma. Int J Biol Sci. 18:4824–4836. 2022. View Article : Google Scholar : PubMed/NCBI

130 

Jia Y, Tian C, Wang H, Yu F, Lv W, Duan Y, Cheng Z, Wang X, Wang Y, Liu T, et al: Long non-coding RNA NORAD/miR-224-3p/MTDH axis contributes to CDDP resistance of esophageal squamous cell carcinoma by promoting nuclear accumulation of β-catenin. Mol Cancer. 20:1622021. View Article : Google Scholar : PubMed/NCBI

131 

Tong Y, Yang L, Yu C, Zhu W, Zhou X, Xiong Y, Wang W, Ji F, He D and Cao X: Tumor-secreted exosomal lncRNA POU3F3 promotes cisplatin resistance in ESCC by inducing fibroblast differentiation into CAFs. Mol Ther Oncolytics. 18:1–13. 2020. View Article : Google Scholar : PubMed/NCBI

132 

Wang P, Yang Z, Ye T, Shao F, Li J, Sun N and He J: lncTUG1/miR-144-3p affect the radiosensitivity of esophageal squamous cell carcinoma by competitively regulating c-MET. J Exp Clin Cancer Res. 39:72020. View Article : Google Scholar : PubMed/NCBI

133 

Liu Z, Lu X, Wen L, You C, Jin X and Liu J: Hsa_circ_0014879 regulates the radiosensitivity of esophageal squamous cell carcinoma through miR-519-3p/CDC25A axis. Anticancer Drugs. 33:e349–e361. 2022. View Article : Google Scholar : PubMed/NCBI

134 

Cheng J, Zhang R, Yan M and Li Y: Circular RNA hsa_circ_0000277 promotes tumor progression and DDP resistance in esophageal squamous cell carcinoma. BMC Cancer. 22:2382022. View Article : Google Scholar : PubMed/NCBI

135 

Zhou S, Guo Z, Lv X and Zhang X: CircGOT1 promotes cell proliferation, mobility, and glycolysis-mediated cisplatin resistance via inhibiting its host gene GOT1 in esophageal squamous cell cancer. Cell Cycle. 21:247–260. 2022. View Article : Google Scholar : PubMed/NCBI

136 

Liu Z, Gu S, Wu K, Li L, Dong C, Wang W and Zhou Y: CircRNA-DOPEY2 enhances the chemosensitivity of esophageal cancer cells by inhibiting CPEB4-mediated Mcl-1 translation. J Exp Clin Cancer Res. 40:3612021. View Article : Google Scholar : PubMed/NCBI

137 

Zhu H, Du F and Cao C: Restoration of circPSMC3 sensitizes gefitinib-resistant esophageal squamous cell carcinoma cells to gefitinib by regulating miR-10a-5p/PTEN axis. Cell Biol Int. 45:107–116. 2021. View Article : Google Scholar : PubMed/NCBI

138 

Tang Q, Zhao S, Zhou N, He J, Zu L, Liu T, Song Z, Chen J, Peng L and Xu S: PD-1/PD-L1 immune checkpoint inhibitors in neoadjuvant therapy for solid tumors (review). Int J Oncol. 62:492023. View Article : Google Scholar : PubMed/NCBI

139 

Yi M, Zheng X, Niu M, Zhu S, Ge H and Wu K: Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Mol Cancer. 21:282022. View Article : Google Scholar : PubMed/NCBI

140 

Jiang W, Pan S, Chen X, Wang ZW and Zhu X: The role of lncRNAs and circRNAs in the PD-1/PD-L1 pathway in cancer immunotherapy. Mol Cancer. 20:1162021. View Article : Google Scholar : PubMed/NCBI

141 

Fang Z, Jiang C and Li S: The potential regulatory roles of circular RNAs in tumor immunology and immunotherapy. Front Immunol. 11:6175832021. View Article : Google Scholar : PubMed/NCBI

142 

Gao C, Xu YJ, Qi L, Bao YF, Zhang L and Zheng L: CircRNA VIM silence synergizes with sevoflurane to inhibit immune escape and multiple oncogenic activities of esophageal cancer by simultaneously regulating miR-124/PD-L1 axis. Cell Biol Toxicol. 38:825–845. 2022. View Article : Google Scholar : PubMed/NCBI

143 

Jin W, Wang L, Cheng S and Lv H: Prognostic value of microRNA-378 in esophageal cancer and its regulatory effect on tumor progression. Exp Ther Med. 22:7042021. View Article : Google Scholar : PubMed/NCBI

144 

Kim S, Kim GH, Park SJ, Kwon CH, I H, Lee MW and Lee BE: Exosomal MicroRNA analyses in esophageal squamous cell carcinoma cell lines. J Clin Med. 11:44262022. View Article : Google Scholar : PubMed/NCBI

145 

Wen J, Wang G, Xie X, Lin G, Yang H, Luo K, Liu Q, Ling Y, Xie X, Lin P, et al: Prognostic value of a four-miRNA signature in patients with lymph node positive locoregional esophageal squamous cell carcinoma undergoing complete surgical resection. Ann Surg. 273:523–531. 2021. View Article : Google Scholar : PubMed/NCBI

146 

Okuda Y, Shimura T, Iwasaki H, Fukusada S, Nishigaki R, Kitagawa M, Katano T, Okamoto Y, Yamada T, Horike SI and Kataoka H: Urinary microRNA biomarkers for detecting the presence of esophageal cancer. Sci Rep. 11:85082021. View Article : Google Scholar : PubMed/NCBI

147 

Zhang J, Ling X, Fang C and Ma J: Identification and validation of an eight-lncRNA signature that predicts prognosis in patients with esophageal squamous cell carcinoma. Cell Mol Biol Lett. 27:392022. View Article : Google Scholar : PubMed/NCBI

148 

Cao S, Wang X, Liu X, Li J, Duan L, Gao Z, Lun S, Zhu Y, Yang H, Zhang H and Zhou F: Integrative analysis of angiogenesis-related long non-coding RNA and identification of a six-DEARlncRNA signature associated with prognosis and therapeutic response in esophageal squamous cell carcinoma. Cancers (Basel). 14:41952022. View Article : Google Scholar : PubMed/NCBI

149 

Liu Y, Wang L, Liu H, Li C and He J: The prognostic significance of metabolic syndrome and a related Six-lncRNA signature in esophageal squamous cell carcinoma. Front Oncol. 10:612020. View Article : Google Scholar : PubMed/NCBI

150 

Xie K, Zheng C, Gu W, Jiang Z, Luo C, Luo J, Diao Y, Wang G, Cong Z, Yao X, et al: A RASSF8-AS1 based exosomal lncRNAs panel used for diagnostic and prognostic biomarkers for esophageal squamous cell carcinoma. Thorac Cancer. 13:3341–3352. 2022. View Article : Google Scholar : PubMed/NCBI

151 

Huang E, Fu J, Yu Q, Xie P, Yang Z, Ji H, Wang L, Luo G, Zhang Y and Li K: CircRNA hsa_circ_0004771 promotes esophageal squamous cell cancer progression via miR-339-5p/CDC25A axis. Epigenomics. 12:587–603. 2020. View Article : Google Scholar : PubMed/NCBI

152 

Wang W, Zhu D, Zhao Z, Sun M, Wang F, Li W, Zhang J and Jiang G: RNA sequencing reveals the expression profiles of circRNA and identifies a four-circRNA signature acts as a prognostic marker in esophageal squamous cell carcinoma. Cancer Cell Int. 21:1512021. View Article : Google Scholar : PubMed/NCBI

153 

Hu X, Wu D, He X, Zhao H, He Z, Lin J, Wang K, Wang W, Pan Z, Lin H and Wang M: circGSK3β promotes metastasis in esophageal squamous cell carcinoma by augmenting β-catenin signaling. Mol Cancer. 18:1602019. View Article : Google Scholar : PubMed/NCBI

154 

Bu J, Gu L, Liu X, Nan X, Zhang X, Meng L, Zheng Y, Liu F, Li J, Li Z, et al: The circRNA circADAMTS6 promotes progression of ESCC and correlates with prognosis. Sci Rep. 12:137572022. View Article : Google Scholar : PubMed/NCBI

155 

Wang Q, Liu H, Liu Z, Yang L, Zhou J, Cao X and Sun H: Circ-SLC7A5, a potential prognostic circulating biomarker for detection of ESCC. Cancer Genet. 240:33–39. 2020. View Article : Google Scholar : PubMed/NCBI

156 

Zhao Q, Zhu X, Ke JM, Su XY, Yi J, Wu DL, Lin J and Deng ZQ: Circular RNA BMI1 serves as a potential target for diagnosis and treatment in esophageal cancer. Technol Cancer Res Treat. 20:153303382110330752021. View Article : Google Scholar : PubMed/NCBI

157 

Tang Y, Gao Z and Liu R: Identification and function of circular RNA hsa_circ_0071106: A novel biomarker for differentiation degree of esophageal squamous cell carcinoma. Pathol Res Pract. 233:1538752022. View Article : Google Scholar : PubMed/NCBI

158 

Chen B, Dragomir MP, Yang C, Li Q, Horst D and Calin GA: Targeting non-coding RNAs to overcome cancer therapy resistance. Signal Transduct Target Ther. 7:1212022. View Article : Google Scholar : PubMed/NCBI

159 

Veneziano D, Di Bella S, Nigita G, Laganà A, Ferro A and Croce CM: Noncoding RNA: Current deep sequencing data analysis approaches and challenges. Hum Mutat. 37:1283–1298. 2016. View Article : Google Scholar : PubMed/NCBI

160 

Anastasiadou E, Faggioni A, Trivedi P and Slack F: The nefarious nexus of noncoding RNAs in cancer. Int J Mol Sci. 19:20722018. View Article : Google Scholar : PubMed/NCBI

161 

Nemeth K, Bayraktar R, Ferracin M and Calin GA: Non-coding RNAs in disease: From mechanisms to therapeutics. Nat Rev Genet. 25:211–232. 2024. View Article : Google Scholar : PubMed/NCBI

162 

Sugihara H, Ishimoto T, Miyake K, Izumi D, Baba Y, Yoshida N, Watanabe M and Baba H: Noncoding RNA expression aberration is associated with cancer progression and is a potential biomarker in esophageal squamous cell carcinoma. Int J Mol Sci. 16:27824–27834. 2015. View Article : Google Scholar : PubMed/NCBI

163 

Li R, Chai L, Lei L, Guo R and Wen X: MiR-671-5p sponging activity of circMMP1 promotes esophageal squamous cancer progression. Thorac Cancer. 14:2924–2933. 2023. View Article : Google Scholar : PubMed/NCBI

164 

Nan Y, Luo Q, Wu X, Chang W, Zhao P, Liu S and Liu Z: HCP5 prevents ubiquitination-mediated UTP3 degradation to inhibit apoptosis by activating c-Myc transcriptional activity. Mol Ther. 31:552–568. 2023. View Article : Google Scholar : PubMed/NCBI

165 

Qiao G, Zhang W and Dong K: Regulation of ferroptosis by noncoding RNAs: A novel promise treatment in esophageal squamous cell carcinoma. Mol Cell Biochem. 477:2193–2202. 2022. View Article : Google Scholar : PubMed/NCBI

166 

Song W and Zou SB: Prognostic role of lncRNA HOTAIR in esophageal squamous cell carcinoma. Clin Chim Acta. 463:169–173. 2016. View Article : Google Scholar : PubMed/NCBI

167 

Sang L, Yang L, Ge Q, Xie S, Zhou T and Lin A: Subcellular distribution, localization, and function of noncoding RNAs. Wiley Interdiscip Rev RNA. 13:e17292022. View Article : Google Scholar : PubMed/NCBI

168 

Shi X, Liu X, Pan S, Ke Y, Li Y, Guo W, Wang Y, Ruan Q, Zhang X and Ma H: A novel autophagy-related long non-coding RNA signature to predict prognosis and therapeutic response in esophageal squamous cell carcinoma. Int J Gen Med. 14:8325–8339. 2021. View Article : Google Scholar : PubMed/NCBI

169 

Zhu T, Ma Z, Wang H, Wei D, Wang B, Zhang C, Fu L, Li Z and Yu G: Immune-related long non-coding RNA signature and clinical nomogram to evaluate survival of patients suffering esophageal squamous cell carcinoma. Front Cell Dev Biol. 9:6419602021. View Article : Google Scholar : PubMed/NCBI

170 

Liao G, Tang J and Bai J: Early development of esophageal squamous cell cancer: Stem cells, cellular origins and early clone evolution. Cancer Lett. 555:2160472023. View Article : Google Scholar : PubMed/NCBI

171 

Hu XY, Wang R, Jin J, Liu XJ, Cui AL, Sun LQ, Li YP, Li Y, Wang YC, Zhen YS, et al: An EGFR-targeting antibody-drug conjugate LR004-VC-MMAE: potential in esophageal squamous cell carcinoma and other malignancies. Mol Oncol. 13:246–263. 2019. View Article : Google Scholar : PubMed/NCBI

172 

Mimura K, Kono K, Maruyama T, Watanabe M, Izawa S, Shiba S, Mizukami Y, Kawaguchi Y, Inoue M, Kono T, et al: Lapatinib inhibits receptor phosphorylation and cell growth and enhances antibody-dependent cellular cytotoxicity of EGFR- and HER2-overexpressing esophageal cancer cell lines. Int J Cancer. 129:2408–2416. 2011. View Article : Google Scholar : PubMed/NCBI

173 

Meng L, Wu H, Wu J, Ding Pa, He J, Sang M and Liu L: Mechanisms of immune checkpoint inhibitors: Insights into the regulation of circular RNAS involved in cancer hallmarks. Cell Death Dis. 15:32024. View Article : Google Scholar : PubMed/NCBI

174 

Ding L, Lu S and Li Y: Regulation of PD-1/PD-L1 pathway in cancer by noncoding RNAs. Pathol Oncol Res. 26:651–663. 2020. View Article : Google Scholar : PubMed/NCBI

175 

Peng Y, Xu Y, Zhang X, Deng S, Yuan Y, Luo X, Hossain MT, Zhu X, Du K, Hu F, et al: A novel protein AXIN1-295aa encoded by circAXIN1 activates the Wnt/β-catenin signaling pathway to promote gastric cancer progression. Mol Cancer. 20:1582021. View Article : Google Scholar : PubMed/NCBI

176 

Zheng X, Chen L, Zhou Y, Wang Q, Zheng Z, Xu B, Wu C, Zhou Q, Hu W, Wu C and Jiang J: A novel protein encoded by a circular RNA circPPP1R12A promotes tumor pathogenesis and metastasis of colon cancer via Hippo-YAP signaling. Mol Cancer. 18:472019. View Article : Google Scholar : PubMed/NCBI

177 

Imazeki H and Kato K: Development of chemotherapeutics for unresectable advanced esophageal cancer. Expert Rev Anticancer Ther. 20:1083–1092. 2020. View Article : Google Scholar : PubMed/NCBI

178 

Fukuda T, Baba H, Okumura T, Kanda M, Akashi T, Tanaka H, Miwa T, Watanabe T, Hirano K, Sekine S, et al: miR-877-3p as a potential tumour suppressor of oesophageal squamous cell carcinoma. Anticancer Res. 43:35–43. 2023. View Article : Google Scholar : PubMed/NCBI

179 

Yu J, Chen S, Niu Y, Liu M, Zhang J, Yang Z, Gao P, Wang W, Han X and Sun G: Functional significance and therapeutic potential of miRNA-20b-5p in esophageal squamous cell carcinoma. Mol Ther Nucleic Acids. 21:315–331. 2020. View Article : Google Scholar : PubMed/NCBI

180 

Xing Y, Zha WJ, Li XM, Li H, Gao F, Ye T, Du WQ and Liu YC: Circular RNA circ-Foxo3 inhibits esophageal squamous cell cancer progression via the miR-23a/PTEN axis. J Cell Biochem. 121:2595–2605. 2020. View Article : Google Scholar : PubMed/NCBI

181 

Jin Y, Meng Q, Zhang B, Xie C, Chen X, Tian B, Wang J, Shih TC, Zhang Y, Cao J, et al: Cancer-associated fibroblasts-derived exosomal miR-3656 promotes the development and progression of esophageal squamous cell carcinoma via the ACAP2/PI3K-AKT signaling pathway. Int J Biol Sci. 17:3689–3701. 2021. View Article : Google Scholar : PubMed/NCBI

182 

Shou Y, Wang X, Liang Y, Liu X and Chen K: Exosomes-derived miR-154-5p attenuates esophageal squamous cell carcinoma progression and angiogenesis by targeting kinesin family member 14. Bioengineered. 13:4610–4620. 2022. View Article : Google Scholar : PubMed/NCBI

183 

Yang J, Zhang Q, Zhao P, Qiao T, Cao Z, Gao F, Liu M and Wu S: DNA methyltransferase 3 beta regulates promoter methylation of microRNA-149 to augment esophageal squamous cell carcinoma development through the ring finger protein 2/Wnt/β-catenin axis. Bioengineered. 13:4010–4027. 2022. View Article : Google Scholar : PubMed/NCBI

184 

Yan J, Shi L, Lin S and Li Y: MicroRNA-624-mediated ARRDC3/YAP/HIF1alpha axis enhances esophageal squamous cell carcinoma cell resistance to cisplatin and paclitaxel. Bioengineered. 12:5334–5347. 2021. View Article : Google Scholar : PubMed/NCBI

185 

Suyal G, Pandey P, Saraya A and Sharma R: Tumour suppressor role of microRNA-335-5p in esophageal squamous cell carcinoma by targeting TTK (Mps1). Exp Mol Pathol. 124:1047382022. View Article : Google Scholar : PubMed/NCBI

186 

Zhang L, Yu S, Yin X, Tu M, Cai L, Zhang Y, Yu L, Zhang S, Pan X and Huang Y: MiR-942-5p inhibits tumor migration and invasion through targeting CST1 in esophageal squamous cell carcinoma. PLoS One. 18:e02770062023. View Article : Google Scholar : PubMed/NCBI

187 

Wang J, Li J, Cheng D, Zhang K, Liu W, Xue Q, Du R, Zhou L, Yeung YT, Bai R, et al: miR-132-3p promotes heat stimulation-induced esophageal squamous cell carcinoma tumorigenesis by targeting KCNK2. Mol Carcinog. 62:583–597. 2023. View Article : Google Scholar : PubMed/NCBI

188 

Xu J, Wang J, Liu L, Chen L, Hu S and Liu F: MicroRNA-196b is related to the overall survival of patients with esophageal squamous cell carcinoma and facilitates tumor progression by regulating SOCS2 (Suppressor Of Cytokine Signaling 2). Bioengineered. 12:7737–7746. 2021. View Article : Google Scholar : PubMed/NCBI

189 

Xue J, Xiao P, Yu X and Zhang X: A positive feedback loop between AlkB homolog 5 and miR-193a-3p promotes growth and metastasis in esophageal squamous cell carcinoma. Hum Cell. 34:502–514. 2021. View Article : Google Scholar : PubMed/NCBI

190 

Li P, Liu X, Xing W, Qiu H, Li R, Liu S and Sun H: Exosome-derived miR-200a promotes esophageal cancer cell proliferation and migration via the mediating Keap1 expression. Mol Cell Biochem. 477:1295–1308. 2022. View Article : Google Scholar : PubMed/NCBI

191 

Luo XJ, He MM, Liu J, Zheng JB, Wu QN, Chen YX, Meng Q, Luo KJ, Chen DL, Xu RH, et al: LncRNA TMPO-AS1 promotes esophageal squamous cell carcinoma progression by forming biomolecular condensates with FUS and p300 to regulate TMPO transcription. Exp Mol Med. 54:834–847. 2022. View Article : Google Scholar : PubMed/NCBI

192 

Zhao Y, Zhang Q, Liu H, Wang N, Zhang X and Yang S: lncRNA PART1, manipulated by transcriptional factor FOXP2, suppresses proliferation and invasion in ESCC by regulating the miR-18a-5p/SOX6 signaling axis. Oncol Rep. 45:1118–1132. 2021. View Article : Google Scholar : PubMed/NCBI

193 

Tang J, Xu H, Liu Q, Zheng J, Pan C, Li Z, Wen W, Wang J, Zhu Q, Wang Z and Chen L: LncRNA LOC146880 promotes esophageal squamous cell carcinoma progression via miR-328-5p/FSCN1/MAPK axis. Aging (Albany NY). 13:14198–14218. 2021. View Article : Google Scholar : PubMed/NCBI

194 

Niu Y, Guo Y, Li Y, Shen S, Liang J, Guo W and Dong Z: LncRNA GATA2-AS1 suppresses esophageal squamous cell carcinoma progression via the mir-940/PTPN12 axis. Exp Cell Res. 416:1131302022. View Article : Google Scholar : PubMed/NCBI

195 

Chen L, Lu J, Xu T, Yan Z, Guo Y, Dong Z and Guo W: KTN1-AS1, a SOX2-mediated lncRNA, activates epithelial-mesenchymal transition process in esophageal squamous cell carcinoma. Sci Rep. 12:201862022. View Article : Google Scholar : PubMed/NCBI

196 

Dong Z, Yang L, Lu J, Guo Y, Shen S, Liang J and Guo W: Downregulation of LINC00886 facilitates epithelial-mesenchymal transition through SIRT7/ELF3/miR-144 pathway in esophageal squamous cell carcinoma. Clin Exp Metastasis. 39:661–677. 2022. View Article : Google Scholar : PubMed/NCBI

197 

Pan C, Chen G, Zhao X, Xu X and Liu J: lncRNA BBOX1-AS1 silencing inhibits esophageal squamous cell cancer progression by promoting ferroptosis via miR-513a-3p/SLC7A11 axis. Eur J Pharmacol. 934:1753172022. View Article : Google Scholar : PubMed/NCBI

198 

Zhang H, Pan E, Zhang Y, Zhao C, Liu Q, Pu Y and Yin L: LncRNA RPL34-AS1 suppresses the proliferation, migration and invasion of esophageal squamous cell carcinoma via targeting miR-575/ACAA2 axis. BMC Cancer. 22:10172022. View Article : Google Scholar : PubMed/NCBI

199 

Lu JT, Yan ZY, Xu TX, Zhao F, Liu L, Li F and Guo W: Reciprocal regulation of LINC00941 and SOX2 promotes progression of esophageal squamous cell carcinoma. Cell Death Dis. 14:722023. View Article : Google Scholar : PubMed/NCBI

200 

Fu X, Chen X, Si Y, Yao Y, Jiang Z and Chen K: Long non-coding RNA NCK1-AS1 is overexpressed in esophageal squamous cell carcinoma and predicts survival. Bioengineered. 13:8302–8310. 2022. View Article : Google Scholar : PubMed/NCBI

201 

Wang C, Zhou M, Zhu P, Ju C, Sheng J, Du D, Wan J, Yin H, Xing Y, Li H, et al: IGF2BP2-induced circRUNX1 facilitates the growth and metastasis of esophageal squamous cell carcinoma through miR-449b-5p/FOXP3 axis. J Exp Clin Cancer Res. 41:3472022. View Article : Google Scholar : PubMed/NCBI

202 

Song H, Tian D, Sun J, Mao X, Kong W, Xu D, Ji Y, Qiu B, Zhan M and Wang J: circFAM120B functions as a tumor suppressor in esophageal squamous cell carcinoma via the miR-661/PPM1L axis and the PKR/p38 MAPK/EMT pathway. Cell Death Dis. 13:3612022. View Article : Google Scholar : PubMed/NCBI

203 

Guan X, Guan X, Wang Y, Lan T, Cheng T, Cui Y and Xu H: Circ_0003340 downregulation mitigates esophageal squamous cell carcinoma progression by targeting miR-940/PRKAA1 axis. Thorac Cancer. 13:1164–1175. 2022. View Article : Google Scholar : PubMed/NCBI

204 

Liang W, Wang C, Wang J and Zhang M: Hsa_circ_0023984 regulates cell proliferation, migration, and invasion in esophageal squamous cancer via regulating miR-1294/PI3K/Akt/c-Myc pathway. Appl Biochem Biotechnol. 194:1–16. 2022. View Article : Google Scholar : PubMed/NCBI

205 

Qian CJ, Tong YY, Wang YC, Teng XS and Yao J: Circ_0001093 promotes glutamine metabolism and cancer progression of esophageal squamous cell carcinoma by targeting miR-579-3p/glutaminase axis. J Bioenerg Biomembr. 54:119–134. 2022. View Article : Google Scholar : PubMed/NCBI

206 

Tang R, Zhou Q, Xu Q, Lu L and Zhou Y: Circular RNA circ_0006948 promotes esophageal squamous cell carcinoma progression by regulating microRNA-3612/LASP1 axis. Dig Dis Sci. 67:2158–2172. 2022. View Article : Google Scholar : PubMed/NCBI

207 

Wang J, Li X, Duan C and Jia Y: CircFNDC3B knockdown restrains the progression of oesophageal squamous cell carcinoma through miR-214-3p/CDC25A axis. Clin Exp Pharmacol Physiol. 49:1209–1220. 2022. View Article : Google Scholar : PubMed/NCBI

208 

Sun Z, Zhang S, Zhang N, Wang J, Wang J and Liu J: Circ_0005231 promotes the progression of esophageal squamous cell carcinoma via sponging miR-383-5p and regulating KIAA0101. Thorac Cancer. 13:1751–1762. 2022. View Article : Google Scholar : PubMed/NCBI

209 

Wang YM, Zhao QW, Sun ZY, Lin HP, Xu X, Cao M, Fu YJ, Zhao XJ, Ma XM and Ye Q: Circular RNA hsa_circ_0003823 promotes the tumor progression, metastasis and apatinib resistance of esophageal squamous cell carcinoma by miR-607/CRISP3 axis. Int J Biol Sci. 18:5787–5808. 2022. View Article : Google Scholar : PubMed/NCBI

210 

Meng F, Zhang X, Wang Y, Lin J, Tang Y, Zhang G, Qiu B, Zeng X, Liu W and He X: Hsa_circ_0021727 (circ-CD44) promotes ESCC progression by targeting miR-23b-5p to activate the TAB1/NFκB pathway. Cell Death Dis. 14:92023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2024
Volume 27 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang L, Wang Y, Gao J, Zhou X, Huang M, Wang X and He Z: Non‑coding RNA: A promising diagnostic biomarker and therapeutic target for esophageal squamous cell carcinoma (Review). Oncol Lett 27: 255, 2024
APA
Zhang, L., Wang, Y., Gao, J., Zhou, X., Huang, M., Wang, X., & He, Z. (2024). Non‑coding RNA: A promising diagnostic biomarker and therapeutic target for esophageal squamous cell carcinoma (Review). Oncology Letters, 27, 255. https://doi.org/10.3892/ol.2024.14388
MLA
Zhang, L., Wang, Y., Gao, J., Zhou, X., Huang, M., Wang, X., He, Z."Non‑coding RNA: A promising diagnostic biomarker and therapeutic target for esophageal squamous cell carcinoma (Review)". Oncology Letters 27.6 (2024): 255.
Chicago
Zhang, L., Wang, Y., Gao, J., Zhou, X., Huang, M., Wang, X., He, Z."Non‑coding RNA: A promising diagnostic biomarker and therapeutic target for esophageal squamous cell carcinoma (Review)". Oncology Letters 27, no. 6 (2024): 255. https://doi.org/10.3892/ol.2024.14388