
Functions and mechanisms of long non‑coding RNA in esophageal squamous cell carcinoma (Review)
- Authors:
- Published online on: July 1, 2025 https://doi.org/10.3892/ol.2025.15164
- Article Number: 418
-
Copyright: © Lin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Esophageal squamous cell carcinoma (ESCC) is a leading cause of cancer-associated mortality worldwide. In 2022, there were ~510,000 new cases of esophageal cancer (EC) and 440,000 mortalities worldwide, with 75% of the cases and mortalities occurring in Asia, this may be related to Asians' preference for hot food (1,2). The risk factors for EC include smoking, drinking (3), eating salted vegetables (4) and low fruit intake (5). Although conventional endoscopy is still the gold standard for diagnosing ESCC, with a pooled sensitivity of 94% and specificity of 92% (6), its high cost and the need for specially trained personnel are significant barriers to widespread implementation. Novel EC markers include tissue biomarkers (7), exhaled gas markers (8), saliva markers (9) and circulating biomarkers (10). Previous studies have highlighted the key role of long non-coding RNAs (lncRNAs) as master regulators of ESCC pathogenesis, orchestrating oncogenic processes through intricate molecular networks (11,12). Clinically, lncRNAs have been associated with various aspects of ESCC, including lymph node metastasis, tumorigenesis (13), tumor nodes metastasis stage (14) and prognosis (15). In ESCC cell lines, lncRNAs have been implicated in processes such as cell proliferation (16), migration (17), invasion (18) and epithelial-to-mesenchymal transition (19). In both cell-derived xenograft models and patient-derived tumor xenografts, lncRNAs have been linked to tumor growth (20), lymph node metastasis (21), liver metastasis (19) and bone metastasis (16). These lncRNAs transcripts modulate proliferation, invasion and metastasis by targeting key signaling pathways. For example, The oncogenic effect of lncRNA BBOX1-AS1 in esophageal squamous cell carcinoma was found to activate the Hedgehog signaling pathway through the miR-506-5p/EIF5A/PTCH1 axis, promoting cancer cell proliferation and stemness (22), while cleavage and polyadenylation specific factor 3 (CPSF3) disrupts miR-125a-5p-mediated suppression of cornichon family AMPA receptor auxiliary protein 2 (CNIH2) to enhance proliferation and migration (23). Conversely, membrane associated guanylate kinase, WW and PDZ domain containing 2 (MAGI2)-AS3 acts as a tumor suppressor by binding enhancer of zeste homolog 2 (EZH2) to downregulate homeobox B7 (HOXB7), thereby inhibiting proliferation and improving radiosensitivity (24). Such dual regulatory roles underscore the versatility of lncRNAs in ESCC progression, mirroring the competing endogenous RNA (ceRNA) network complexity observed in nasopharyngeal carcinoma where lncRNA/circular RNA-miRNA-mRNA interactions govern therapeutic resistance (25).
Epithelial-mesenchymal transition (EMT), a cornerstone of metastasis, is controlled by lncRNAs. While hepatocyte nuclear factor 1 homeobox A (HNF1A)-AS1 suppresses EMT by sponging miR-298 to inhibit transcription factor 4 (TCF4) and mesenchymal markers (26), kinectin 1 (KTN1)-AS1 promotes EMT via the Hippo/YAP1 pathway (27). Notably, LINC00858 exacerbates invasion by inducing EMT markers (such as N-cadherin) through miR-425-5p/ABL proto-oncogene 2, non-receptor tyrosine kinase (ABL2) signaling (28), whereas small nucleolar RNA host gene 12 (SNHG12) dual-regulates EMT and stemness via β-catenin stabilization (29), revealing the complexity of lncRNA-mediated EMT networks. This regulatory intricacy parallels findings in other types of cancer, such as colorectal cancer (30) and breast cancer (31), where miRNA-mediated EMT modulation (such as CD44-STAT3 axis inhibition) enhances radiosensitivity (32), suggesting potential cross-cancer therapeutic strategies.
Cell cycle dysregulation, a hallmark of ESCC, is governed by lncRNAs such as cancer susceptibility 9 (CASC9), which recruits EZH2 to suppress programmed cell death 4 (PDCD4) and accelerate G1/S transition (33) and LINC00022, which promotes p21 degradation via m6A demethylation (34). Conversely, LINC01980 induces G1 arrest through growth arrest and DNA damage inducible a (GADD45A) upregulation (35), illustrating their context-dependent roles in cell cycle control.
Therapeutic resistance, a notable barrier in ESCC management, is associated with lncRNAs. Colon cancer associated transcript 2 (CCAT2) enhances radioresistance by inhibiting miR-145/p53 pathways (36), while CASC8 stabilizes heterogeneous nuclear ribonucleoprotein L (hnRNPL) to activate anti-apoptotic Bcl2/caspase3 signaling (37). Hypoxia-driven LINC01116 and exosomal myocardial infarction associated transcript (MIAT) mediate resistance via hypoxia inducible factor 1α (HIF-1α)/miR-3612 and TATA-box binding protein associated factor 1 (TAF1)/sterol regulatory element binding transcription factor 1 (SREBF1) axes (38,39). These findings align with studies demonstrating that lncRNAs such as ectodermal-neural cortex 1 regulate tumor microenvironments and immune evasion in cancers (39,40), highlighting their systemic role in therapy resistance. Cancer stemness, a driver of recurrence, is amplified by BBOX1-AS1 via Hedgehog activation (22) and LINC00941 via SOX2/OCT4/NANOG feedback loops (41). Apoptosis evasion, which is key for tumor survival, is regulated by deleted in lymphocytic leukemia 1 (DLEU1; via BCL2 stabilization) (42) and reversed by interferon regulatory factor 1 (IRF1)-AS through interferon signaling (43), highlighting lncRNAs as dual modulators of cell death.
Collectively, lncRNAs are central players in ESCC malignancy, governing proliferation, metastasis, therapy resistance and stemness through multitarget mechanisms (Fig. 1). Their interactions with super-enhancers (a type of DNA regulatory element with strong transcriptional regulation ability), revealed by tools such as SEanalysis 2.0 (44), may further elucidate context-specific transcriptional networks. Their regulatory complexity positions them as promising diagnostic biomarkers and therapeutic targets. Integrating multi-omics approaches, such as ceRNA networks (25) and phosphorylation proteomics (32), may accelerate biomarker discovery and combination therapy design. The present review aimed to summarize the involvement of lncRNAs in ESCC, providing insight into the clinical diagnosis, treatment and prevention of ESCC.
History of lncRNAs
A class of RNA molecule >200 nucleotides, lncRNAs were first discovered in the 1970s. Scientists mainly concentrated on mRNA that encodes proteins, while ncRNAs were regarded as ‘noise’ or ‘by-products’. Nevertheless, the advancement of technologies such as high-throughput sequencing (45) for detecting non-coding RNA and research (46) demonstrated that ncRNAs serve vital roles in gene regulation, epigenetics and the occurrence of disease. In 2003, researchers first found an lncRNA associated with gene silencing on the X chromosome (47). Guttman et al (48) discovered the lncRNA-HOTAIR, which is notable in gene locus regulation. In 2007, Rinn et al (49) identified an lncRNA located in the HOX gene cluster and found its crucial participation in gene locus regulation. lncRNAs serve important roles in embryonic development (50). Studies have also demonstrated the role of lncRNA in the initiation and progression of tumors, which has triggered research into the role of lncRNA in cancer (51–53).
Classification of lncRNAs
According to genomic annotations in Ensembl Release 96 (April 2019) (54), human lncRNAs are categorized into distinct functional groups. These include 3′overlapping nc transcripts, antisense-oriented lncRNAs, genomic-spanning interspersed RNAs, intronic sequence-retained transcripts, intra-exonic sense RNAs, coding sequence-overlapping molecules and macro-scale regulatory RNAs. Intronic lncRNAs reside within the nc segments (introns) of protein-coding genetic loci. Although transcribed from these intronic regions, they are functionally independent from protein synthesis pathways (55). Antisense lncRNAs exhibit sequence complementarity to protein-coding gene strands, potentially modulating gene activity through RNA duplex formation via Watson-Crick base pairing mechanisms (56,57). Positioned between coding genetic elements, intergenic lncRNAs may exert cis-regulatory effects on neighboring genes (48). Sense strand-aligned lncRNAs demonstrate exonic overlap with protein-coding transcripts through co-directional transcription (58). Messenger-like lncRNAs serve as epigenetic modulators capable of fine-tuning target gene expression (50). Structure-encoded lncRNAs contribute to cell organization by maintaining nuclear compartmentalization and chromatin topology (59).
lncRNA localization and associated research techniques
lncRNAs are found in the cytoplasm (60), nucleus (61), nucleolus (62), as well as other subcellular regions and vesicles (such as nucleolus and exosomes). Their localization is associated with molecular functions, including nuclear lncRNA target-specific genomic regions by binding to chromatin modification complexes, while cytoplasmic lncRNAs act by binding to mRNA or miRNA (60,63). Specific sequence motifs in their primary sequences are associated with subcellular localization (64). Investigating the localization of lncRNAs is key for understanding their roles in gene regulation, disease development and cell function. The techniques used to study the localization of lncRNAs include in situ hybridization (65), RNA immunoprecipitation (66), RNA (67) and single-cell RNA sequencing (68) and fluorescence in situ hybridization-flow cytometry (69).
Conservation of lncRNAs
While lncRNAs serve key biological roles, the majority of these molecules demonstrate poor evolutionary preservation in nucleotide alignment across organisms. This sequence divergence implies potential challenges in cross-species identification through conventional homology searches. This low degree of conservation is considered to reflect the diversity and specificity of lncRNA functions, as well as their rapid evolution (70). Notwithstanding this sequence variability, certain lncRNAs exhibit conserved architectural features or regulatory capacity in divergent species. These molecules may retain comparable spatial configurations or participate in equivalent gene expression networks despite sequence dissimilarity (59,71). Numerous lncRNAs display pronounced phyletic restriction, being uniquely present in particular evolutionary lineages. This taxonomic exclusivity implies specialized biological contributions during species-specific developmental or adaptive mechanisms (70,72). Notably, the regulatory regions controlling lncRNA transcription show evolutionary stability similar to those governing protein-coding genes, suggesting shared regulatory principles (73,74).
lncRNAs as diagnostic or prognostic biomarkers for ESCC in blood
For ESCC diagnostic biomarkers, ease of acquisition and detectability are key. In early-stage cases, invasive methods such as endoscopic biopsy are often declined by patients due to discomfort and complexity, making blood or saliva-based biomarkers key for improving early screening compliance (75–85). LncRAs show promise as non-invasive tools, given their stability in the circulation (86). Emerging evidence highlights their potential to address the need for accessible screening in high-prevalence regions. LncRNAs can circulate in bodily fluids such as blood and urine due to their ability to traverse cell membranes (87). This enables their identification through non-invasive diagnostic approaches (88). LncRNAs in bodily fluids directly reflect the expression patterns of specific genes and distinguish patients with cancer from healthy individuals (89). Furthermore, circulating lncRNAs resist enzymatic degradation by RNases (86,90). Apoptotic bodies, microvesicles and exosomes represent phospholipid bilayer-enclosed vesicles carrying DNA, RNA, lipid, proteins, polysaccharides and metabolites. These structures are released into the circulatory system for systemic distribution, mediating intercellular material transfer (91–93). Reverse transcription-quantitative PCR (RT-qPCR) is utilized for detecting circulating lncRNAs due to its high sensitivity and specificity (94). Multiple lncRNAs are stably present in the blood of patients with ESCC through plasma, serum, whole blood or exosomes, demonstrating non-invasive diagnostic potential; for example, upregulated lncRNAs such as Linc00152 (plasma, AUC=0.698) (95) and MALAT1 (serum exosomes, AUC=0.755) (96), and downregulated lncRNAs, such as LINC-PINT (plasma, AUC=0.862) (97), were detected in body fluids to distinguish tumors from healthy controls. And as detected in the whole blood, LINC00324 (AUC=0.627) (98), EWSAT1 (AUC=0.717) (99) and NEF (AUC=0.904) (100) were also of a certain diagnostic value. It is worth noting that the high diagnostic efficacy of 5 plasma exosome lncRNA combinations (AUC=0.999) (101) and 4 serum exosome lncRNA panels (AUC=0.853) (102) confirms a synergistic detection advantage of circulating lncRNAs. In this chapter, the research on lncRNAs as diagnostic or prognostic biomarkers for ESCC in blood is discussed (Table I).
Role and regulatory axes of lncRNAs in ESCC
lncRNAs serve a key role in the onset and development of ESCC. lncRNAs serve multifaceted roles in ESCC, influencing biological processes including cell cycle control, cell proliferation, EMT, migration, invasion, drug resistance, apoptosis and cell stemness. lncRNAs within ESCC contribute to both the development and progression of the disease (Table II). Potential lncRNA candidate markers in EC are presented in Fig. 2.
lncRNAs regulate proliferation, invasion and migration in ESCC
Multiple studies demonstrate that lncRNAs markedly influence ESCC proliferation, invasion and migration by regulating key signaling pathways (103–115). For example, BBOX1-AS1 promotes ESCC cell proliferation and stemness by sponging miR-506-5p to upregulate EIF5A, activating the Hedgehog signaling pathway (22). CPSF3 enhances ESCC proliferation and migration by altering CNIH2 mRNA 3′untranslated region polyadenylation, thereby abolishing miR-125a-5p suppression (23). Forkhead box D2 (FOXD2)-AS1 drives EC cell proliferation and invasion via the miR-145-5p/cyclin-dependent kinase (CDK) 6 axis (116), while HLA complex P5 (HCP5) activates the PI3K/AKT/mTOR pathway through the miR-139-5p/phosphodiesterase 4A axis to promote malignancy (117). Potassium calcium-activated channel subfamily M regulatory β subunit 2-AS1 accelerates EC growth and migration via the miR-3194-3p/glycogen phosphorylase L axis (118), and LINC00626 enhances esophagogastric junction adenocarcinoma malignancy by activating the JAK1/STAT3/KH-type splicing regulatory protein axis (119). LINC00941 forms a positive feedback loop with SOX2 to promote transcriptional reprogramming via ILF2/Y-box binding protein 1 recruitment (41). LINC00858 upregulates ABL2 by sponging miR-425-5p, activating EMT and metastasis pathways (28). Conversely, MAGI2-AS3 suppresses proliferation and enhances radiosensitivity by binding EZH2 to downregulate HOXB7 (24). MIAT promotes migration and invasion via MMP-2/9 modulation, with knockdown inducing G1-phase arrest (120). Motor neuron and pancreas homeobox 1 (MNX1)-AS1 upregulates sirtuin 1 (SIRT1) via miR-34a sponging to drive proliferation and metastasis (121), while NOP2/Sun RNA methyltransferase 2 methylated lncRNA activates MMP3/MMP10 via ERK1/2 signaling to enhance metastasis (122). These findings highlight the multitarget regulatory roles of lncRNAs in ESCC malignancy. The above findings reveal the essence of the role of lncRNAs in ESCC through a ‘multi-target multi pathway’ network: They can act as microRNA sponges [such as BBOX1-AS1 (22) and FOXD2-AS1 (116)], intervene in mRNA processing [such as CPSF3 (23)] or chromatin remodeling [such as MAGI2-AS3 (24)], and ultimately manifest in key biological processes affecting cell proliferation (22,117) and invasion (122).
lncRNAs regulate EMT in ESCC
EMT serves a key role in ESCC progression, with lncRNAs regulating transcription factors or pathways to induce EMT. HNF1A-AS1 inhibits EMT and stemness by sponging miR-298 to suppress TCF4 expression, thereby downregulating mesenchymal markers such as N-cadherin (26). KTN1-AS1 activates the Hippo pathway via the miR-885-5p/striatin 3/YAP1 axis to promote EMT and metastasis (27). LINC00152 drives EMT and oxaliplatin resistance by enhancing zinc finger E-box binding homeobox 1 (ZEB1) histone modification through EZH2 binding (123), while LINC-ubiquitin C suppresses E-cadherin and upregulates EZH2 to facilitate metastasis (124). The LINC00858/miR-425-5p/ABL2 axis exacerbates invasion by inducing EMT markers (such as N-cadherin and vimentin) (28). Maternally expressed 3 attenuates EMT in EC109 cells by inhibiting phosphoserine aminotransferase 1-dependent GSK3β/Snail signaling (125), whereas SNHG12 dual-drives EMT and stemness via the miR-6835-3p/BMI1 proto-oncogene, polycomb ring finger (BMI1) axis and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2)-mediated β-catenin stabilization (29). X-inactive specific transcript accelerates invasion through the miR-34a/ZEB1/E-cadherin axis (126), illustrating the complexity of lncRNA-mediated EMT networks. The regulation of EMT by lncRNA has duality: There are both KTN1-AS1 (27) and LINC00152 (123), which promote EMT, while HNF1A-AS1 (26) inhibits EMT. Such regulatory complexity indicates that targeting a single lncRNA may fail due to compensatory mechanisms, necessitating the development of combined intervention strategies integrating ‘lncRNA-transcription factor-pathway’ axes. For instance, simultaneously inhibiting SNHG12 and activating GSK3β may be more effective in blocking EMT and stemness (29).
lncRNAs regulate cell cycle progression in ESCC
Cell cycle dysregulation is a hallmark of tumor proliferation (127–129). CASC9 promotes G1/S transition by recruiting EZH2 to the PDCD4 promoter, increasing H3K27me3 modification to suppress PDCD4 (33). LINC00022 accelerates cell cycle progression via FTO α-ketoglutarate-dependent dioxygenase-regulated m6A demethylation, promoting p21 ubiquitination and degradation (34). LINC01980 induces G1 arrest and apoptosis by upregulating GADD45A (35), while LIPH-4 activates cyclin D1/CDK4 via the miR-216b/IGF2BP2 pathway to drive S-phase entry (127). Similarly, MIAT knockdown induces G1-phase arrest (120), MNX1-AS1 modulates the cell cycle via miR-34a/SIRT1 (121) and nicotinamide phosphoribosyltransferase (NLIPMT) triggers G0/G1 arrest by suppressing survivin via miR-320 (128). These mechanisms position lncRNAs as molecular switches for cell cycle checkpoints. LncRNAs also have dual roles as cell cycle ‘molecular switches’: Some like CASC9 (33) and LINC00022 (34) promote proliferation, while others like LINC01980 (35) and NLIPMT (128) suppress it. Their regulatory networks involve multiple mechanisms, including epigenetic modifications [m6A (34), H3K27me3 (33)], protein ubiquitination (34) and signaling pathway activation (127).
lncRNAs regulate radiosensitivity or chemoresistance in ESCC
Radiotherapy and chemotherapy resistance are challenges in ESCC treatment (130–135). CCAT2 enhances radioresistance by suppressing miR-145/p70S6K1 and p53 pathways (36), while diGeorge syndrome critical region gene 5 decreases radiosensitivity via the miR-195/hexokinase 2 axis-mediated Warburg effect inhibition (136). Family with sequence similarity 201 member A mediates radioresistance via the miR-101/ATM/mTOR pathway (137), whereas growth arrest specific 5 improves radiosensitivity by downregulating miR-21 to upregulate reversion inducing cysteine rich protein with Kazal motifs (138). Chemoresistance mechanisms include CASC8 stabilizing hnRNPL to activate Bcl2/caspase 3 (37); CCAT1 promoting cisplatin resistance via the miR-143/polo like kinase 1/BUB1 mitotic checkpoint serine/threonine kinase B pathway (139) and HCP5 suppressing apoptosis via the UTP3 small subunit processome component/c-Myc/vesicle-associated membrane protein 3 pathway (140). LINC00473 reduces radiosensitivity by upregulating spindling 1 via miR-374a-5p sponging or inhibiting DNA repair via the miR-497-5p/cell division cycle 25A pathway (141,142). Hypoxia-induced LINC01116 activates anti-apoptotic pathways via HIF-1α/miR-3612 (38), while LINC00261 reverses cisplatin resistance via miR-545-3p/metallothionein 1M (143). LINC01270 induces 5-fluorouracil (5-FU) resistance by recruiting DNA methyltransferases to methylate the glutathione S-transferase P promoter (144). Exosome-mediated resistance is exemplified by MIAT, which activates the TAF1/SREBF1 axis to induce paclitaxel resistance (39). ncRNA activated by DNA damage dual-regulates cisplatin resistance and radioresistance via miR-224-3p/metadherin/β-catenin and primary-miR-199a1/endonuclease/exonuclease/phosphatase family domain containing 1 (145,146). Metabolic reprogramming via Pvt1 oncogene/miR-181a-5p/glutaminase underscores the role of lncRNAs in therapy resistance (147). In ESCC drug resistance, lncRNAs act via a ‘multi-pathway-multi-node’ network. They regulate DNA repair [e.g., LINC00473 (141,142)], apoptosis resistance [e.g., CASC8 (37)], metabolic reprogramming [e.g., Pvt1 (147)] and stress response [e.g., LINC01116 (38)]. Future research combining single-cell sequencing to analyze the heterogeneous lncRNA expression in drug-resistant tumor subgroups may better facilitate the clinical translation of lncRNA-based drug resistance reversal therapy.
lncRNAs regulate stemness in ESCC
Cancer stemness is associated with recurrence and drug resistance (112). BBOX1-AS1 enhances stemness via Hedgehog pathway activation (22), while HNF1A-AS1 suppresses stemness markers via miR-298/TCF4 (26). LINC-regulator of reprogramming maintains stemness by sponging miR-145 and miR-15b to target SOX9 (148,149). LINC00941 forms a feedback loop with SOX2 to upregulate OCT4 and NANOG (41) and prostate cancer-associated transcript 6 activates JAK/STAT signaling to amplify stemness markers (104). Protein disulfide isomerase family A member 3 pseudogene 1 sustains stemness via the WW domain containing E3 ubiquitin protein ligase 2/OCT4 axis (105), and LINC-POU class 3 homeobox 3 promotes radioresistant stemness by upregulating CD44/CD133/CD90 (150). SNHG12 and SOX2-overlapping transcript regulate pluripotency through BMI1/β-catenin and SOX2/OCT4 pathways, respectively (29,151). Targeting these lncRNAs may mitigate tumor recurrence. LncRNAs regulate pluripotency signaling pathways [e.g., Hedgehog (22) and JAK/STAT (104)] during embryonic development, forming a core regulatory hub in ESCC stemness maintenance, providing a precise therapeutic solution for clinical use [such as Hedgehog pathway combined intervention (22)].
lncRNAs regulate apoptosis in ESCC
Apoptosis evasion is key for tumor survival (113,152–154). DLEU1 stabilizes dynein light chain LC8-type 1 to upregulate BCL2, inhibiting cisplatin-induced apoptosis (42). FAM136A suppresses apoptosis via CDK5RAP1 mitochondrial tRNA methylthiotransferase regulation (155), whereas IRF1-AS promotes apoptosis through interferon response and IRF1 activation (43). LINC00261 enhances radiation-induced apoptosis via the DIRAS family GTPase 1/Bcl-2 pathway (156), while LINC00337 suppresses apoptosis by upregulating TPX2 microtubule nucleation factor-mediated autophagy (157). LINC01980 inhibits apoptosis through the GADD45A-dependent DNA damage response (35). Key mechanisms include MAGI2-AS3 promoting radiotherapy-induced apoptosis via HOXB7 downregulation (24); NLIPMT activating caspase pathways via miR-320/survivin (128) and SNHG1 suppressing endoplasmic reticulum stress-related apoptosis via the miR-216a-3p/transmembrane BAX inhibitor motif containing 6 axis (158). Zinc finger protein 582 (ZNF582)-AS1 induces apoptosis by demethylating ZNF582 (159), highlighting lncRNAs as potential therapeutic targets. LncRNAs play a crucial role in tumorigenesis, therapeutic resistance and prognosis by finely regulating apoptosis in ESCC cells. These discoveries deepen our understanding of ESCC developmental mechanisms, such as the molecular basis of apoptosis evasion (160). Furthermore, they offer new insights for developing diagnostic biomarkers such as DLEU1 (42) and IRF1-AS (43), and for innovating therapeutic strategies such as lncRNA-targeted chemoradiotherapy sensitization.
Conclusion
lncRNAs are pivotal orchestrators of ESCC pathogenesis, efficiently regulating hallmark oncogenic processes, including unrestrained proliferation, metastatic dissemination, treatment resistance and cancer stemness, through their ability to engage in multifaceted regulatory networks (22,26,37). Their capacity to modulate epigenetic reprogramming, post-transcriptional modulation and intercellular communication underscores their dual role as molecular linchpins in tumor evolution and promising candidates for precision oncology. Crosstalk between lncRNAs and key signaling cascades (such as Hedgehog, PI3K/AKT/mTOR and JAK/STAT) (104,117) not only demonstrates their mechanistic versatility but also positions them as novel diagnostic biomarkers and druggable targets for overcoming therapeutic bottlenecks in ESCC management.
LncRNAs have diverse modes of action, complex mechanisms and unclear functions. lncRNAs affect gene expression through various mechanisms such as epigenetic modifications (161), RNA-RNA (162) and RNA-protein (163) interactions and regulation of miRNA activity (164); however, the specific mechanisms are often difficult to clarify. The function of lncRNA is associated with its location within the cell (such as the nucleus or cytoplasm), but the technology for dynamically tracking its localization changes is still immature (61,64). Technical bottlenecks limit research; current detection methods have low sensitivity and lncRNA expression levels are typically low, and traditional sequencing techniques may miss key molecules (165). Evolutionary conservation is low, model construction is difficult and species specificity is strong; >80% of lncRNAs only exist in humans or primates, and model animals such as mice cannot fully simulate their function. In addition their function is difficult to extrapolate. The lncRNA mechanisms discovered in animal experiments may not be directly applicable to the treatment of human disease. Clinical applications face multiple obstacles such as low biomarker specificity as numerous lncRNAs (such as HOTAIR) are abnormally expressed in various types of cancer, making it difficult to serve as a single diagnostic biomarker. There are difficulties in developing therapeutic targets as there are high off-target risks; targeted intervention may affect other non-target RNAs or genes. Finally, the detection methods for circulating lncRNA, such as exosome extraction and RT-qPCR primer design, lack a unified standard, which affects the comparability of results. Although this review comprehensively summarizes the latest research on lncRNAs in EC, it does not delve into the complex ceRNA network formed by lncRNAs, circRNAs, miRNAs and other factors; the application of cutting-edge RNA detection technology in EC is also rarely mentioned, and these contents will be supplemented in future research.
Future investigations should deconstruct the spatiotemporal dynamics of lncRNA interactions within tumor microenvironments, identifying their isoform-specific functions and translating these insights into clinically actionable strategies, ranging from liquid biopsy-based early detection platforms to RNA-targeted therapeutics. Meanwhile, future research should also clarify their dynamic interactions and transformation potential to overcome clinical challenges in ESCC. By summarizing lncRNA-mediated oncogenic pathways, the present review provides a multidimensional framework for understanding ESCC biology, while proposing innovative avenues to reimagine clinical paradigms in diagnosis (such as leveraging exosomal lncRNA signatures) and therapeutic intervention (such as the mechanism of drug resistance regulated by lncRNA). Such efforts hold transformative potential to mitigate the global burden of ESCC, a malignancy notable for its late-stage detection and poor survival outcomes.
Acknowledgements
Not applicable.
Funding
The present study was supported by Xiamen Natural Science Foundation (grant no. 3502Z20227389).
Availability of data and materials
Not applicable.
Authors' contributions
YL and HJ conceived the study, designed the methodology, performed the literature review and wrote and edited the manuscript. ZL conceived the study, designed the methodology, performed the literature review and wrote the manuscript. HX performed the literature review and wrote the manuscript. WZ and RP drafted the initial manuscript, retrieved papers and collected the required data, organised ESCC-related papers retrieved from databases. WZ also used software such as EndNote and Word, provided resources, supervised and performed visualization. WZ and RP performed the literature review. ZZ wrote and edited the manuscript. Data authentication is not applicable. All authors have read and approved the final 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
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI | |
Teng Y, Xia C, Cao M, Yang F, Yan X, He S, Cao M, Zhang S, Li Q, Tan N, et al: Esophageal cancer global burden profiles, trends, and contributors. Cancer Biol Med. 21:656–666. 2024.PubMed/NCBI | |
Prabhu A, Obi KO and Rubenstein JH: The synergistic effects of alcohol and tobacco consumption on the risk of esophageal squamous cell carcinoma: A meta-analysis. Am J Gastroenterol. 109:822–827. 2014. View Article : Google Scholar : PubMed/NCBI | |
Islami F, Ren JS, Taylor PR and Kamangar F: Pickled vegetables and the risk of oesophageal cancer: A meta-analysis. Br J Cancer. 101:641–1647. 2009. View Article : Google Scholar : PubMed/NCBI | |
Li B, Jiang G, Zhang G, Xue Q, Zhang H, Wang C and Zhao T: Intake of vegetables and fruit and risk of esophageal adenocarcinoma: A meta-analysis of observational studies. Eur J Nutr. 53:1511–1521. 2014. View Article : Google Scholar : PubMed/NCBI | |
Wong MCS, Deng Y, Huang J, Bai Y, Wang HHX, Yuan J, Zhang L, Yip HC and Chiu PWY: Performance of screening tests for esophageal squamous cell carcinoma: A systematic review and meta-analysis. Gastrointest Endosc. 96:197–207.e34. 2022. View Article : Google Scholar : PubMed/NCBI | |
Min Q, Zhang M, Lin D, Zhang W, Li X, Zhao L, Teng H, He T, Sun W, Fan J, et al: Genomic characterization and risk stratification of esophageal squamous dysplasia. Med Rev. 4:244–256. 2024. View Article : Google Scholar | |
Sasaki F, Kanmura S, Oda K, Maeda H, Kabayama M, Iwaya H, Komaki Y, Arima S, Tanoue S, Hashimoto S, et al: Acetaldehyde breath test as a cancer risk marker in patients with esophageal and hypopharyngeal squamous cell carcinoma. PLoS One. 16:e02514572021. View Article : Google Scholar : PubMed/NCBI | |
Li K, Lin Y, Zhou Y, Xiong X, Wang L, Li J, Zhou F, Guo Y, Chen S, Chen Y, et al: Salivary extracellular MicroRNAs for early detection and prognostication of esophageal cancer: A clinical study. Gastroenterology. 165:932–945.e9. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Yu N, Cheng G, Zhang T, Wang J, Deng L, Li J, Zhao X, Xu Y, Yang P, et al: Prognostic value of circulating tumour DNA during post-radiotherapy surveillance in locally advanced esophageal squamous cell carcinoma. Clin Transl Med. 12:e11162022. View Article : Google Scholar : PubMed/NCBI | |
Su M, Xiao Y, Ma J, Cao D, Zhou Y, Wang H, Liao Q and Wang W: Long non-coding RNAs in esophageal cancer: Molecular mechanisms, functions, and potential applications. J Hematol Oncol. 11:1182018. View Article : Google Scholar : PubMed/NCBI | |
Yan Q, Wong W, Gong L, Yang J, Liang D, Chin KY, Dai S and Wang J: Roles of long non-coding RNAs in esophageal cell squamous carcinoma (Review). Int J Mol Med. 54:722024. View Article : Google Scholar : PubMed/NCBI | |
Li H, Jia J, Yang L, Chu J, Sheng J, Wang C, Meng W, Jia Z, Yin H, Wan J and He F: LncRNA MIR205HG drives esophageal squamous cell carcinoma progression by regulating miR-214/SOX4 axis. Onco Targets Ther. 13:13097–13109. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jia J, Li H, Chu J, Sheng J, Wang C, Jia Z, Meng W, Yin H, Wa J and He F: LncRNA FAM83A-AS1 promotes ESCC progression by regulating miR-214/CDC25B axis. J Cancer. 12:1200–1211. 2021. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
Wang L, Ren X, Ma X, Yin L, Niu X and Xing S: LncRNA OIP5-AS1 promotes the development of esophageal squamous cell carcinoma by binding to miR-1297. Panminerva Med. 64:589–590. 2022. View Article : Google Scholar : PubMed/NCBI | |
Lin P, Li Q, Lv X, Qu J, Wang D, Li A and Jiang G: HMGA1 promotes the development of esophageal squamous cell carcinoma by mediating miR-671-5p/lncRNA DLEU1. Panminerva Med. 65:264–266. 2023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Xu ML, Liu TC, Dong FX, Meng LX, Ling AX and Liu S: Exosomal lncRNA LINC01711 facilitates metastasis of esophageal squamous cell carcinoma via the miR-326/FSCN1 axis. Aging (Albany NY). 13:19776–19788. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Zhang W, Liu W, Huang L, Wang Y, Li D, Wang G, Zhao Z, Chi X, Xue Y, et al: Long noncoding RNA VESTAR regulates lymphangiogenesis and lymph node metastasis of esophageal squamous cell carcinoma by enhancing VEGFC mRNA stability. Cancer Res. 81:3187–3199. 2021. View Article : Google Scholar : PubMed/NCBI | |
Xu ML, Liu TC, Dong FX, Meng LX, Ling AX and Liu S: BBOX1-AS1 activates hedgehog signaling pathway to facilitate the proliferation and stemness of esophageal squamous cell carcinoma cells via miR-506-5p/EIF5A/PTCH1 axis. Curr Mol Pharmacol. 16:894–904. 2023.PubMed/NCBI | |
Zhang Y, Liu D, Guo D, Lin W, Lu W, Hu L, Chen S and Chen C: CPSF3 regulates alternative polyadenylation of CNIH2 to promote esophageal squamous cell carcinoma progression. Cancer Lett. 593:2169252024. View Article : Google Scholar : PubMed/NCBI | |
Cheng W, Shi X, Lin M, Yao Q, Ma J and Li J: LncRNA MAGI2-AS3 overexpression sensitizes esophageal cancer cells to irradiation through Down-regulation of HOXB7 via EZH2. Front Cell Dev Biol. 8:5528222020. View Article : Google Scholar : PubMed/NCBI | |
Liu S, Li X, Xie Q, Zhang S, Liang X, Li S and Zhang P: Identification of a lncRNA/circRNA-miRNA-mRNA network in Nasopharyngeal Carcinoma by deep sequencing and bioinformatics analysis. J Cancer. 15:1916–1928. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wang Z, Huang YF, Yu L and Jiao Y: sh-HNF1A-AS1 reduces the epithelial-mesenchymal transition and stemness of esophageal cancer cells. Neoplasma. 69:560–570. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chen L, Lu J, Li X, Wang X, Qiao R, Guo W and Ren Q: LncRNA KTN1-AS1 facilitates esophageal squamous cell carcinoma progression via miR-885-5p/STRN3 axis. Genes Genomics. 46:241–252. 2024. View Article : Google Scholar : PubMed/NCBI | |
Li P, Ding H, Han S, Ding S and Yang Y: Long noncoding RNA LINC00858 aggravates the progression of esophageal squamous cell carcinoma via regulating the miR-425-5p/ABL2 axis. Heliyon. 10:e273372024. View Article : Google Scholar : PubMed/NCBI | |
Wu D, He X, Wang W, Hu X, Wang K and Wang M: Long noncoding RNA SNHG12 induces proliferation, migration, epithelial-mesenchymal transition, and stemness of esophageal squamous cell carcinoma cells via post-transcriptional regulation of BMI1 and CTNNB1. Mol Oncol. 14:2332–2351. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li S, Lv C, Li J, Xie T, Liu X, Zheng Z, Qin Z, Hui X and Yu Y: LncRNA LINC00473 promoted colorectal cancer cell proliferation and invasion by targeting miR-195 expression. Am J Transl Res. 13:6066–6075. 2021.PubMed/NCBI | |
Song B, Wei F, Peng J, Wei X, Liu M, Nie Z, Ma Y and Peng T: Icariin regulates EMT and stem Cell-like character in breast cancer through modulating lncRNA NEAT1/TGFβ/SMAD2 signaling pathway. Biol Pharm Bull. 47:399–410. 2024. View Article : Google Scholar : PubMed/NCBI | |
Huang H, Huang F, Liang X, Fu Y, Cheng Z, Huang Y, Chen Z, Duan Y and Chen Y: Afatinib reverses EMT via inhibiting CD44-Stat3 axis to promote radiosensitivity in nasopharyngeal carcinoma. Pharmaceuticals (Basel). 16:372022. View Article : Google Scholar : PubMed/NCBI | |
Wu Y, Hu L, Liang Y, Li J, Wang K, Chen X, Meng H, Guan X, Yang K and Bai Y: Up-regulation of lncRNA CASC9 promotes esophageal squamous cell carcinoma growth by negatively regulating PDCD4 expression through EZH2. Mol Cancer. 16:1502017. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Zhang S, Liang Y, Wu Y, Chen X, Wang K, Li J, Guan X, Xiong G, Yang K, Bai Y, et al: Upregulation of a novel lncRNA LINC01980 promotes tumor growth of esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 513:73–80. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang M, Wang L, He X, Zhang J, Zhu Z, Zhang M and Li X: lncRNA CCAT2 promotes radiotherapy resistance for human esophageal carcinoma cells via the miR-145/p70S6K1 and p53 pathway. Int J Oncol. 56:327–336. 2020.PubMed/NCBI | |
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 | |
Zhang M, Wang Z, Wu Y, Chen M, Li J and Liu G: Hypoxia-induced factor-1α promotes radioresistance of esophageal cancer cells by transcriptionally activating LINC01116 and suppressing miR-3612 under hypoxia. J Biochem Mol Toxicol. 38:e235512024. View Article : Google Scholar : PubMed/NCBI | |
Zhang S, Zhong J, Guo D, Zhang S, Huang G, Chen Y, Xu C, Chen W, Zhang Q, Zhao C, et al: MIAT shuttled by tumor-secreted exosomes promotes paclitaxel resistance in esophageal cancer cells by activating the TAF1/SREBF1 axis. J Biochem Mol Toxicol. 37:e233802023. View Article : Google Scholar : PubMed/NCBI | |
Cao Z, Zhu J, Wang Z, Peng Y and Zeng L: Comprehensive pan-cancer analysis reveals ENC1 as a promising prognostic biomarker for tumor microenvironment and therapeutic responses. Sci Rep. 14:253312024. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Li Q, Zhang Z, Jiang H, Hou J, Chai Y, Nan H, Li F and Wang L: DLEU1 promotes cell survival by preventing DYNLL1 degradation in esophageal squamous cell carcinoma. J Transl Med. 20:2452022. View Article : Google Scholar : PubMed/NCBI | |
Huang J, Li J, Li Y, Lu Z, Che Y, Mao S, Lei Y, Zang R, Zheng S, Liu C, et al: Interferon-inducible lncRNA IRF1-AS represses esophageal squamous cell carcinoma by promoting interferon response. Cancer Lett. 459:86–99. 2019. View Article : Google Scholar : PubMed/NCBI | |
Qian FC, Zhou LW, Li YY, Yu ZM, Li LD, Wang YZ, Xu MC, Wang QY and Li CQ: SEanalysis 2.0: A comprehensive Super-enhancer regulatory network analysis tool for human and mouse. Nucleic Acids Res. 51:W520–W527. 2023. View Article : Google Scholar : PubMed/NCBI | |
Saville L, Wu L, Habtewold J, Cheng Y, Gollen B, Mitchell L, Stuart-Edwards M, Haight T, Mohajerani M and Zovoilis A: NERD-seq: A novel approach of Nanopore direct RNA sequencing that expands representation of Non-coding RNAs. Genome Biol. 25:2332024. View Article : Google Scholar : PubMed/NCBI | |
Liu Z, Gao L, Cheng L, Lv G, Sun B, Wang G and Tang Q: The roles of N6-methyladenosine and its target regulatory noncoding RNAs in tumors: Classification, mechanisms, and potential therapeutic implications. Exp Mol Med. 55:487–501. 2023. View Article : Google Scholar : PubMed/NCBI | |
Plath K, Fang J, Mlynarczyk-Evans SK, Cao R, Worringer KA, Wang H, de la Cruz CC, Otte AP, Panning B and Zhang Y: Role of histone H3 lysine 27 methylation in X inactivation. Science. 300:131–135. 2003. View Article : Google Scholar : PubMed/NCBI | |
Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP, et al: Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 458:223–227. 2009. View Article : Google Scholar : PubMed/NCBI | |
Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI | |
Rinn JL and Chang HY: Genome regulation by long noncoding RNAs. Annu Rev Biochem. 81:145–166. 2012. View Article : Google Scholar : PubMed/NCBI | |
Huarte M: The emerging role of lncRNAs in cancer. Nat Med. 21:1253–1261. 2015. View Article : Google Scholar : PubMed/NCBI | |
Schmitt AM and Chang HY: Long Noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI | |
Bhan A and Mandal SS: Long noncoding RNAs: Emerging stars in gene regulation, epigenetics and human disease. ChemMedChem. 9:1932–1956. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yates AD, Achuthan P, Akanni W, Allen J, Allen J, Alvarez-Jarreta J, Amode MR, Armean IM, Azov AG, Bennett R, et al: Ensembl 2020. Nucleic Acids Res. 48:D682–D688. 2020.PubMed/NCBI | |
Mattick JS and Makunin IV: Non-coding RNA. Hum Mol Genet. 15:R17–R29. 2006. View Article : Google Scholar : PubMed/NCBI | |
Katayama S, Tomaru Y, Kasukawa T, Waki K, Nakanishi M, Nakamura M, Nishida H, Yap CC, Suzuki M, Kawai J, et al: Antisense transcription in the mammalian transcriptome. Science. 309:1564–1566. 2005. View Article : Google Scholar : PubMed/NCBI | |
Pelechano V and Steinmetz LM: Gene regulation by antisense transcription. Nat Rev Genet. 14:880–893. 2013. View Article : Google Scholar : PubMed/NCBI | |
Ma L, Bajic VB and Zhang Z: On the classification of long non-coding RNAs. RNA Biol. 10:925–934. 2013. View Article : Google Scholar : PubMed/NCBI | |
Quinn JJ and Chang HY: Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 17:47–62. 2016. View Article : Google Scholar : PubMed/NCBI | |
Cheng S, Jia Y, Wu J, Li J and Cao Y: Helicobacter pylori infection induces gastric cancer cell malignancy by targeting HOXA-AS2/miR-509-3p/MMD2 axis. Genes Genomics. 46:647–657. 2024. View Article : Google Scholar : PubMed/NCBI | |
Yan S, Teng L, Du J, Ji L, Xu P, Zhao W and Tao W: Long non-coding RNA DANCR aggravates breast cancer through the miR-34c/E2F1 feedback loop. Mol Med Rep. 29:932024. View Article : Google Scholar : PubMed/NCBI | |
Wang C, Chen R, Zhu X, Zhang X and Lian N: Long noncoding RNA small nucleolar RNA host gene 5 facilitates neuropathic pain in spinal nerve injury by promoting SCN9A expression via CDK9. Hum Cell. 37:451–464. 2024. View Article : Google Scholar : PubMed/NCBI | |
Geisler S and Coller J: RNA in unexpected places: Long non-coding RNA functions in diverse cellular contexts. Nat Rev Mol Cell Biol. 14:699–712. 2013. View Article : Google Scholar : PubMed/NCBI | |
Gudenas BL and Wang L: Prediction of LncRNA subcellular localization with deep learning from sequence features. Sci Rep. 8:163852018. View Article : Google Scholar : PubMed/NCBI | |
Gozzetti A and Le Beau MM: Fluorescence in situ hybridization: Uses and limitations. Semin Hematol. 37:320–333. 2000. View Article : Google Scholar : PubMed/NCBI | |
Keene JD, Komisarow JM and Friedersdorf MB: RIP-Chip: The isolation and identification of mRNAs, microRNAs and protein components of ribonucleoprotein complexes from cell extracts. Nat Protoc. 1:302–307. 2006. View Article : Google Scholar : PubMed/NCBI | |
Wang Z, Gerstein M and Snyder M: RNA-Seq: A revolutionary tool for transcriptomics. Nat Rev Genet. 10:57–63. 2009. View Article : Google Scholar : PubMed/NCBI | |
Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, et al: Highly parallel Genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 161:1202–1214. 2015. View Article : Google Scholar : PubMed/NCBI | |
Shaffer SM, Dunagin MC, Torborg SR, Torre EA, Emert B, Krepler C, Beqiri M, Sproesser K, Brafford PA, Xiao M, et al: Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature. 546:431–435. 2017. View Article : Google Scholar : PubMed/NCBI | |
Hezroni H, Koppstein D, Schwartz MG, Avrutin A, Bartel DP and Ulitsky I: Principles of long noncoding RNA evolution derived from direct comparison of transcriptomes in 17 species. Cell Rep. 11:1110–1122. 2015. View Article : Google Scholar : PubMed/NCBI | |
Necsulea A, Soumillon M, Warnefors M, Liechti A, Daish T, Zeller U, Baker JC, Grützner F and Kaessmann H: The evolution of lncRNA repertoires and expression patterns in tetrapods. Nature. 505:635–640. 2014. View Article : Google Scholar : PubMed/NCBI | |
Pang KC, Frith MC and Mattick JS: Rapid evolution of noncoding RNAs: Lack of conservation does not mean lack of function. Trends Genet. 22:1–5. 2006. View Article : Google Scholar : PubMed/NCBI | |
Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, Oyama R, Ravasi T, Lenhard B, Wells C, et al: The transcriptional landscape of the mammalian genome. Science. 309:1559–1563. 2005. View Article : Google Scholar : PubMed/NCBI | |
Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al: The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 22:1775–1789. 2012. View Article : Google Scholar : PubMed/NCBI | |
Tong YS, Wang XW, Zhou XL, Liu ZH, Yang TX, Shi WH, Xie HW, Lv J, Wu QQ and Cao XF: Identification of the long non-coding RNA POU3F3 in plasma as a novel biomarker for diagnosis of esophageal squamous cell carcinoma. Mol Cancer. 14:32015. View Article : Google Scholar : PubMed/NCBI | |
Wang W, He X, Zheng Z, Ma X, Hu X, Wu D and Wang M: Serum HOTAIR as a novel diagnostic biomarker for esophageal squamous cell carcinoma. Mol Cancer. 16:752017. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Pang D and Zhang X: The function of lncRNA LINC00997 as a diagnostic marker in the progression of esophageal squamous cell carcinoma. Ann Clin Lab Sci. 53:230–237. 2023.PubMed/NCBI | |
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 | |
Yan Y, Li S, Wang S, Rubegni P, Tognetti L, Zhang J and Yan L: Long noncoding RNA HAND2-AS1 inhibits cancer cell proliferation, migration, and invasion in esophagus squamous cell carcinoma by regulating microRNA-21. J Cell Biochem. 120:9564–9571. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhihua Z, Weiwei W, Lihua N, Jianying Z and Jiang G: p53-induced long non-coding RNA PGM5-AS1 inhibits the progression of esophageal squamous cell carcinoma through regulating miR-466/PTEN axis. IUBMB Life. 71:1492–1502. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhou M, Bao S, Gong T, Wang Q, Sun J, Li J, Lu M, Sun W, Su J, Chen H and Liu Z: The transcriptional landscape and diagnostic potential of long non-coding RNAs in esophageal squamous cell carcinoma. Nat Commun. 14:37992023. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Li T, Peng C, Mao Q, Shen B, Shi M, Lu H, Xiao T, Yang A and Cheng C: Knockdown of long noncoding RNA LINC00240 inhibits esophageal cancer progression by regulating miR-26a-5p. Contrast Media Mol Imaging. 2022:10716272022. View Article : Google Scholar : PubMed/NCBI | |
Luo D, Huang Z, Lv H, Wang Y, Sun W and Sun X: Up-regulation of MicroRNA-21 indicates poor prognosis and promotes cell proliferation in esophageal squamous cell carcinoma via upregulation of lncRNA SNHG1. Cancer Manag Res. 12:1–14. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ma L, Yan W, Sun X and Chen P: Chen, long noncoding RNA VPS9D1-AS1 promotes esophageal squamous cell carcinoma progression via the Wnt/β-catenin signaling pathway. J Cancer. 12:6894–6904. 2021. View Article : Google Scholar : PubMed/NCBI | |
Sun K, Zhao X, Wan J, Yang L, Chu J, Dong S, Yin H, Ming L and He F: The diagnostic value of long non-coding RNA MIR31HG and its role in esophageal squamous cell carcinoma. Life Sci. 202:124–130. 2018. View Article : Google Scholar : PubMed/NCBI | |
Shi T, Gao G and Cao Y: Long noncoding RNAs as novel biomarkers have a promising future in cancer diagnostics. Dis Markers. 2016:90851952016. View Article : Google Scholar : PubMed/NCBI | |
Shi K, Liu T, Fu H, Li W and Zheng X: Zheng, Genome-wide analysis of lncRNA stability in human. PLoS Comput Biol. 17:e10089182021. View Article : Google Scholar : PubMed/NCBI | |
Sole C, Arnaiz E, Manterola L, Otaegui D and Lawrie CH: The circulating transcriptome as a source of cancer liquid biopsy biomarkers. Semin Cancer Biol. 58:100–108. 2019. View Article : Google Scholar : PubMed/NCBI | |
Lo YM: Circulating nucleic acids in plasma and serum: An overview. Ann N Y Acad Sci. 945:1–7. 2001. View Article : Google Scholar : PubMed/NCBI | |
Qi P, Zhou XY and Du X: Circulating long non-coding RNAs in cancer: Current status and future perspectives. Mol Cancer. 15:392016. View Article : Google Scholar : PubMed/NCBI | |
Xie H, Ma H and Zhou D: Plasma HULC as a promising novel biomarker for the detection of hepatocellular carcinoma. Biomed Res Int. 2013:1361062013. View Article : Google Scholar : PubMed/NCBI | |
Revenfeld AL, Bæk R, Nielsen MH, Stensballe A, Varming K and Jørgensen M: Diagnostic and prognostic potential of extracellular vesicles in peripheral blood. Clin Ther. 36:830–846. 2014. View Article : Google Scholar : PubMed/NCBI | |
Mathieu M, Martin-Jaular L, Lavieu G and Théry C: Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 21:9–17. 2019. View Article : Google Scholar : PubMed/NCBI | |
Benes V and Castoldi M: Expression profiling of microRNA using real-time quantitative PCR, how to use it and what is available. Methods. 50:244–249. 2010. View Article : Google Scholar : PubMed/NCBI | |
Hu HB, Jie HY and Zheng XX: Three circulating LncRNA predict early progress of esophageal squamous cell carcinoma. Cell Physiol Biochem. 40:117–125. 2016. View Article : Google Scholar : PubMed/NCBI | |
Hu L, Xie K, Zheng C, Qiu B, Jiang Z, Luo C, Diao Y, Luo J, Yao X and Shen Y: Exosomal MALAT1 promotes the proliferation of esophageal squamous cell carcinoma through glyoxalase 1-dependent methylglyoxal removal. Noncoding RNA Res. 9:330–340. 2024. View Article : Google Scholar : PubMed/NCBI | |
Rong H, Chen B, Ma K, Wei X, Peng J and Zhu J: Downregulation of lncRNA LINC-PINT participates in the recurrence of esophageal squamous cell carcinoma possibly by interacting miRNA-21. Cancer Biother Radiopharm. 36:273–279. 2021.PubMed/NCBI | |
Sharma U, Barwal TS, Khandelwal A, Rana MK, Rana APS, Singh K and Jain A: Circulating long Non-Coding RNAs LINC00324 and LOC100507053 as potential liquid biopsy markers for esophageal squamous cell carcinoma: A pilot study. Front Oncol. 12:8239532022. View Article : Google Scholar : PubMed/NCBI | |
Uttam V, Rana MK, Sharma U, Singh K and Jain A: Circulating long non-coding RNA EWSAT1 acts as a liquid biopsy marker for esophageal squamous cell carcinoma: A pilot study. Noncoding RNA Res. 9:1–11. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Hu SL, Qiao CH, Ye JF, Li M, Ma HM, Wang JH, Xin SY and Yuan ZL: LncRNA-NEF inhibits proliferation, migration and invasion of esophageal squamous-cell carcinoma cells by inactivating wnt/β-catenin pathway. Eur Rev Med Pharmacol Sci. 22:6824–6831. 2018.PubMed/NCBI | |
Jiao Z, Yu A, Rong W, He X, Zen K, Shi M and Wang T: Five-lncRNA signature in plasma exosomes serves as diagnostic biomarker for esophageal squamous cell carcinoma. Aging (Albany NY). 12:15002–15010. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yan S, Du L, Jiang X, Duan W, Li J, Xie Y, Zhan Y, Zhang S, Wang L, Li S and Wang C: Evaluation of serum exosomal lncRNAs as diagnostic and prognostic biomarkers for esophageal squamous cell carcinoma. Cancer Manag Res. 12:9753–9763. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yao W, Liu J, Hou Z, Jia X, Yang D, Feng M, Wu S and Wei L: Recombinant human protein TCFL5-activated NRSN2-AS1 promotes esophageal cancer progression via the microRNA-874-5p/RELT regulatory axis. Int J Biol Macromol. 277:1338142024. View Article : Google Scholar : PubMed/NCBI | |
Luo Q, Kuang Y, Tao X, Zhu X, Zhao X, Lu G, Zhang W, Sun E and Chen B: Pan-cancer analysis of PCAT6 and its effect on oesophageal squamous cell carcinoma cell proliferation and migration. Med Oncol. 40:1252023. View Article : Google Scholar : PubMed/NCBI | |
Huang T, You Q, Huang D, Zhang Y, He Z, Shen X, Li F, Shen Q, Onyebuchi IC, Wu C, et al: A positive feedback between PDIA3P1 and OCT4 promotes the cancer stem cell properties of esophageal squamous cell carcinoma. Cell Commun Signal. 22:602024. View Article : Google Scholar : PubMed/NCBI | |
Zhang S, Jiang H, Xu Z, Jiang Y, She Y, Huang X, Feng S, Chen W, Chen S, Chen Y, et al: The resistance of esophageal cancer cells to paclitaxel can be reduced by the knockdown of long noncoding RNA DDX11-AS1 through TAF1/TOP2A inhibition. Am J Cancer Res. 9:2233–2248. 2019.PubMed/NCBI | |
Liu H, Zhang J, Luo X, Zeng M and Xu L, Zhang Q, Liu H, Guo J and Xu L: Overexpression of the long noncoding RNA FOXD2-AS1 promotes cisplatin resistance in esophageal squamous cell carcinoma through the miR-195/Akt/mTOR axis. Oncol Res. 28:65–73. 2020. View Article : Google Scholar : PubMed/NCBI | |
Shen GY, Zhang Y, Huang RZ, Huang ZY, Yang LY, Chen DZ and Yang SB: FOXP4-AS1 promotes CD8+ T cell exhaustion and esophageal cancer immune escape through USP10-stabilized PD-L1. Immunol Res. 72:766–775. 2024. View Article : Google Scholar : PubMed/NCBI | |
Guo Y, Wang L, Yang H and Ding N: Knockdown long non-coding RNA HCP5 enhances the radiosensitivity of esophageal carcinoma by modulating AKT signaling activation. Bioengineered. 13:884–893. 2022. View Article : Google Scholar : PubMed/NCBI | |
Lin K, Jiang H, Zhuang SS, Qin YS, Qiu GD, She YQ, Zheng JT, Chen C, Fang L and Zhang SY: Long noncoding RNA LINC00261 induces chemosensitization to 5-fluorouracil by mediating methylation-dependent repression of DPYD in human esophageal cancer. FASEB J. 33:1972–1988. 2019. View Article : Google Scholar : PubMed/NCBI | |
Sun Y, Wang J, Pan S, Yang T, Sun X, Wang Y, Shi X, Zhao X, Guo J and Zhang X: LINC00657 played oncogenic roles in esophageal squamous cell carcinoma by targeting miR-615-3p and JunB. Biomed Pharmacother. 108:316–324. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liang F, Luo Q, Han H, Zhang J, Yang Y and Chen J: Long noncoding RNA LINC01088 inhibits esophageal squamous cell carcinoma progression by targeting the NPM1-HDM2-p53 axis. Acta Biochim Biophys Sin (Shanghai). 55:367–381. 2023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Tan R, Liu J, Wang J, Zhang W, He M and Zhang Y: Long noncoding RNA SNHG6 silencing sensitized esophageal cancer cells to 5-FU via EZH2/STAT pathway. Sci Rep. 13:53632023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Shi W, Gao Z, Song J and Wang W: Silence of FOXD2-AS1 inhibited the proliferation and invasion of esophagus cells by regulating miR-145-5p/CDK6 axis. Histol Histopathol. 35:1013–1021. 2020.PubMed/NCBI | |
Xu J, Ma J, Guan B, Li J, Wang Y and Hu S: LncRNA HCP5 promotes malignant cell behaviors in esophageal squamous cell carcinoma via the PI3K/AKT/mTOR signaling. Cell Cycle. 20:1374–1388. 2021. View Article : Google Scholar : PubMed/NCBI | |
Xu J, Liu X, Liu X and Zhi Y: Long noncoding RNA KCNMB2-AS1 promotes the development of esophageal cancer by modulating the miR-3194-3p/PYGL axis. Bioengineered. 12:6687–6702. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang F, Fan L, Kang X, Wei H and Li L: High LINC00626 expression promotes esophagogastric junction adenocarcinoma metastasis: The mediating role of the JAK1/STAT3/KHSRP axis. Nan Fang Yi Ke Da Xue Xue Bao. 44:541–552. 2024.PubMed/NCBI | |
Zhang W, Chen Q and Lei C: lncRNA MIAT promotes cell invasion and migration in esophageal cancer. Exp Ther Med. 19:3267–3274. 2020.PubMed/NCBI | |
Chu J, Li H, Xing Y, Jia J, Sheng J, Yang L, Sun K, Qu Y, Zhang Y, Yin H, et al: LncRNA MNX1-AS1 promotes progression of esophageal squamous cell carcinoma by regulating miR-34a/SIRT1 axis. Biomed Pharmacother. 116:1090292019. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Li J, Luo M, Zhou C, Shi X, Yang W, Lu Z, Chen Z, Sun N and He J: Novel long noncoding RNA NMR promotes tumor progression via NSUN2 and BPTF in esophageal squamous cell carcinoma. Cancer Lett. 430:57–66. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang S, Liao W, Wu Q, Huang X, Pan Z, Chen W, Gu S, Huang Z, Wang Y, Tang X, et al: LINC00152 upregulates ZEB1 expression and enhances epithelial-mesenchymal transition and oxaliplatin resistance in esophageal cancer by interacting with EZH2. Cancer Cell Int. 20:5692020. View Article : Google Scholar : PubMed/NCBI | |
Niu G, Zhuang H, Li B and Cao G: Long noncoding RNA linc-UBC1 promotes tumor invasion and metastasis by regulating EZH2 and repressing E-cadherin in esophageal squamous cell carcinoma. J BUON. 23:157–162. 2018.PubMed/NCBI | |
Li MK, Liu LX, Zhang WY, Zhan HL, Chen RP, Feng JL and Wu LF: Long non-coding RNA MEG3 suppresses epithelial-to-mesenchymal transition by inhibiting the PSAT1-dependent GSK-3β/Snail signaling pathway in esophageal squamous cell carcinoma. Oncol Rep. 44:2130–2142. 2020.PubMed/NCBI | |
Guo B, He M, Ma M, Tian Z, Jin J and Tian G: Long Non-coding RNA X-inactive specific transcript promotes esophageal squamous cell carcinoma progression via the MicroRNA 34a/Zinc finger E-box-Binding homeobox 1 pathway. Dig Dis Sci. 69:1169–1181. 2024. View Article : Google Scholar : PubMed/NCBI | |
Xiao Y, Tang J, Yang D, Zhang B, Wu J, Wu Z, Liao Q, Wang H, Wang W and Su M: Long noncoding RNA LIPH-4 promotes esophageal squamous cell carcinoma progression by regulating the miR-216b/IGF2BP2 axis. Biomark Res. 10:602022. View Article : Google Scholar : PubMed/NCBI | |
Li D, Li D, Meng L, Liu J, Huang C and Sun H: LncRNA NLIPMT inhibits tumorigenesis in esophageal Squamous-cell carcinomas by regulating miR-320/Survivin axis. Cancer Manag Res. 12:12603–12612. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Li R, Ding X, He M and Zhang R: Long noncoding RNA SNHG6 promotes oesophageal squamous cell carcinoma by downregulating the miR-101-3p/EZH2 pathway. J Biochem Mol Toxicol. 36:e229592022. View Article : Google Scholar : PubMed/NCBI | |
Hu D, Ma A, Lu H, Gao Z, Yu Y, Fan J, Liu S, Wang Y and Zhang M: LINC00963 promotes cisplatin resistance in esophageal squamous cell carcinoma by interacting with miR-10a to Upregulate SKA1 expression. Appl Biochem Biotechnol. 196:7219–7232. 2024. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Liu G, Guo W, Chen G, Li W, Cui Y, Qin J and Peng J: Lnc-MCEI mediated the chemosensitivity of esophageal squamous cell carcinoma via miR-6759-5p to competitively regulate IGF2. Int J Biol Sci. 16:2938–2950. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhu ZJ, Pang Y, Jin G, Zhang HY, Wang WH, Liu JW, Tuo GX, Wu P, Yang Y, Wang ZQ and Wang K: Hypoxia induces chemoresistance of esophageal cancer cells to cisplatin through regulating the lncRNA-EMS/miR-758-3p/WTAP axis. Aging (Albany NY). 13:17155–17176. 2021. View Article : Google Scholar : PubMed/NCBI | |
Xu C, Guo Y, Liu H, Chen G, Yan Y and Liu T: TUG1 confers cisplatin resistance in esophageal squamous cell carcinoma by epigenetically suppressing PDCD4 expression via EZH2. Cell Biosci. 8:612018. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Hua Y, Jiang Z, Yue J, Shi M, Zhen X, Zhang X, Yang L, Zhou R and Wu S: Cancer-associated Fibroblast-promoted LncRNA DNM3OS confers radioresistance by regulating DNA damage response in esophageal squamous cell carcinoma. Clin Cancer Res. 25:1989–2000. 2019. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Chen M, Liu P, Chen Y, Chen Z, Shen M, Liu X, Li X, Li A, Lin Y, Yang R, et al: Long noncoding RNA FAM201A mediates the radiosensitivity of esophageal squamous cell cancer by regulating ATM and mTOR expression via miR-101. Front Genet. 9:6112018. View Article : Google Scholar : PubMed/NCBI | |
Lin J, Liu Z, Liao S, Li E, Wu X and Zeng W: Elevation of long non-coding RNA GAS5 and knockdown of microRNA-21 up-regulate RECK expression to enhance esophageal squamous cell carcinoma cell radio-sensitivity after radiotherapy. Genomics. 112:2173–2185. 2020. View Article : Google Scholar : PubMed/NCBI | |
Hu M, Zhang Q, Tian XH, Wang JL, Niu YX and Li G: lncRNA CCAT1 is a biomarker for the proliferation and drug resistance of esophageal cancer via the miR-143/PLK1/BUBR1 axis. Mol Carcinog. 58:2207–2217. 2019. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Chen W, Zhang Y, Wang H, Pan T, Zhang Y and Li C: LINC00473/miR-374a-5p regulates esophageal squamous cell carcinoma via targeting SPIN1 to weaken the effect of radiotherapy. J Cell Biochem. 120:14562–14572. 2019. View Article : Google Scholar : PubMed/NCBI | |
Liu WH, Qiao HY, Xu J, Wang WQ, Wu YL and Wu X: LINC00473 contributes to the radioresistance of esophageal squamous cell carcinoma by regulating microRNA-497-5p and cell division cycle 25A. Int J Mol Med. 46:571–582. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Wang X, Yan P, Liu Y and Jiang X: LINC00261 suppresses cisplatin resistance of esophageal squamous cell carcinoma through miR-545-3p/MT1M Axis. Front Cell Dev Biol. 9:6877882021. View Article : Google Scholar : PubMed/NCBI | |
Li N, Zhao Z, Miao F, Cai S, Liu P, Yu Y and Wang B: Silencing of long non-coding RNA LINC01270 inhibits esophageal cancer progression and enhances chemosensitivity to 5-fluorouracil by mediating GSTP1methylation. Cancer Gene Ther. 28:471–485. 2021. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Sun Y, Wang J, Ma Y, Li J, Sun X, Zhao X, Shi X, Hu Y, Qu F and Zhang X: Radiation induces NORAD expression to promote ESCC radiotherapy resistance via EEPD1/ATR/Chk1 signalling and by inhibiting pri-miR-199a1 processing and the exosomal transfer of miR-199a-5p. J Exp Clin Cancer Res. 40:3062021. View Article : Google Scholar : PubMed/NCBI | |
Chen Z, Wang Q, Huang L, Xu G and Hu J: LncRNA PVT1 confers cisplatin resistance of esophageal cancer cells through modulating the miR-181a-5p-glutaminase (GLS) axis. Nutr Cancer. 75:1646–1657. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Yu X, Zhang Z, Pang L, Xu J, Jiang J, Liang W, Chai Y, Hou J and Li F: Linc-ROR promotes esophageal squamous cell carcinoma progression through the derepression of SOX9. J Exp Clin Cancer Res. 36:1822017. View Article : Google Scholar : PubMed/NCBI | |
Sahebi R, Malakootian M, Balalaee B, Shahryari A, Khoshnia M, Abbaszadegan MR, Moradi A and Javad Mowla S: Linc-ROR and its spliced variants 2 and 4 are significantly up-regulated in esophageal squamous cell carcinoma. Iran J Basic Med Sci. 19:1131–1135. 2016.PubMed/NCBI | |
Chen Y, Tang J, Li L and Lu T: Effect of Linc-POU3F3 on radiotherapy resistance and cancer stem cell markers of esophageal cancer cells. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 46:583–590. 2021.(In English, Chinese). PubMed/NCBI | |
Shahryari A, Rafiee MR, Fouani Y, Oliae NA, Samaei NM, Shafiee M, Semnani S, Vasei M and Mowla SJ: Two novel splice variants of SOX2OT, SOX2OT-S1, and SOX2OT-S2 are coupregulated with SOX2 and OCT4 in esophageal squamous cell carcinoma. Stem Cells. 32:126–134. 2014. View Article : Google Scholar : PubMed/NCBI | |
Xue ST, Cao SQ, Ding JC, Li WJ, Hu GS, Zheng JC, Lin X, Chen C, Liu W and Zheng B: LncRNA LUESCC promotes esophageal squamous cell carcinoma by targeting the miR-6785-5p/NRSN2 axis. Cell Mol Life Sci. 81:1212024. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Wang Y, Zhang J, Shi Z and Liu J: LncRNA NONHSAT227443.1 confers esophageal squamous cell carcinoma chemotherapy resistance by activating PI3K/AKT signaling via targeting MRTFB. Technol Cancer Res Treat. 23:153303382412743692024. View Article : Google Scholar : PubMed/NCBI | |
Li HM, Yu YK, Liu Q, Wei XF, Zhang J, Zhang RX, Sun HB, Wang ZF, Xing WQ and Li Y: LncRNA SNHG1 regulates the progression of esophageal squamous cell cancer by the miR-204/HOXC8 axis. Onco Targets Ther. 13:757–767. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sun S, Huang C, Fan W, Wang Z, Li K, Liu X, Wang Z, Zhao T, Zhang G and Li X: FAM136A as a diagnostic biomarker in esophageal cancer: Insights into immune infiltration, m6A modification, alternative splicing, cuproptosis, and the ceRNA network. Adv Biol (Weinh). 8:e24001572024. View Article : Google Scholar : PubMed/NCBI | |
Yang B, Ma H and Bian Y: LINC00261 inhibits esophageal cancer radioresistance by Down-regulating microRNA-552-3p and promoting DIRAS1. Cancer Manag Res. 13:8559–8573. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yang C, Shen S, Zheng X, Ye K, Ge H, Sun Y and Lu Y: Long non-coding RNA LINC00337 induces autophagy and chemoresistance to cisplatin in esophageal squamous cell carcinoma cells via upregulation of TPX2 by recruiting E2F4. FASEB J. 34:6055–6069. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kong N, Chi Y, Ma H and Luo D: LncRNA SNHG1 acts as a ceRNA for miR-216a-3p to regulate TMBIM6 expression in esophageal squamous cell carcinoma. J Cancer. 15:3128–3139. 2024. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Yan H, Wang B, Wang H, Chen A, Zhu T, Liu J, Yu G and Kang M: High methylation of the same promoter of lncRNA ZNF582-AS1/ZNF582 promotes malignant progression of esophageal cancer. Epigenomics. 16:733–752. 2024. View Article : Google Scholar : PubMed/NCBI | |
Sharma A, Boise LH and Shanmugam M: Cancer metabolism and the evasion of apoptotic cell death. Cancers (Basel). 11:11442019. View Article : Google Scholar : PubMed/NCBI | |
Neve B, Jonckheere N, Vincent A and Van Seuningen I: Epigenetic regulation by lncRNAs: An overview focused on UCA1 in colorectal cancer. Cancers (Basel). 10:4402018. View Article : Google Scholar : PubMed/NCBI | |
Lou W, Ding B and Fu P: Pseudogene-derived lncRNAs and their miRNA sponging mechanism in human cancer. Front Cell Dev Biol. 8:852020. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Nasrolahi A, Khojasteh Pour F, Mousavi Salehi A, Kempisty B, Hajizadeh M, Feghhi M, Azizidoost S and Farzaneh M: Potential roles of lncRNA MALAT1-miRNA interactions in ocular diseases. J Cell Commun Signal. 17:1203–1217. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sarropoulos I, Marin R, Cardoso-Moreira M and Kaessmann H: Developmental dynamics of lncRNAs across mammalian organs and species. Nature. 571:510–514. 2019. View Article : Google Scholar : PubMed/NCBI |