Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Oncology
Join Editorial Board Propose a Special Issue
Print ISSN: 1019-6439 Online ISSN: 1791-2423
Journal Cover
March-2024 Volume 64 Issue 3

Full Size Image

Cover Legend PDF

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
March-2024 Volume 64 Issue 3

Full Size Image

Cover Legend PDF

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review

Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review)

  • Authors:
    • Yifan Shao
    • Yuwei Dong
    • Jing Zhou
    • Zhihua Lu
    • Chen Chen
    • Xiaomin Yuan
    • Linhai He
    • Wenwen Tang
    • Zepeng Chen
    • Yuji Wang
    • Qiurong Li
    • Shuhui Zhan
    • Zhengxi Qiu
    • Kuiling Wang
    • Jiaze Ma
    • Yugen Chen
    • Yang Li
  • View Affiliations / Copyright

    Affiliations: The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China, Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
  • Article Number: 22
    |
    Published online on: January 9, 2024
       https://doi.org/10.3892/ijo.2024.5610
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Long‑stranded non‑coding RNAs (lncRNAs) are RNAs that consist of >200 nucleotides. The majority of lncRNAs do not encode proteins but have been revealed to mediate a variety of important physiological functions. Antisense‑lncRNAs (AS‑lncRNAs) are transcribed from the opposite strand of a protein or non‑protein coding gene as part of the antisense strand of the coding gene. AS‑lncRNAs can serve an important role in the tumorigenesis, prognosis, metastasis and drug resistance of a number of malignancies. This has been reported to be exerted through various mechanisms, such as endogenous competition, promoter interactions, direct interactions with mRNAs, acting as ‘scaffolds’ to regulate mRNA half‑life, interactions with 5‑untranslated regions and regulation of sense mRNAs. AS‑lncRNAs have been found to either inhibit or promote tumor aggressiveness by regulating cell proliferation, energy metabolism, inflammation, inflammatory‑carcinoma transformation, invasion, migration and angiogenesis. In addition, accumulating evidence has documented that AS‑lncRNAs can regulate tumor therapy resistance. Therefore, targeting aberrantly expressed AS‑lncRNAs for cancer treatment may prove to be a promising approach to reverse therapy resistance. In the present review, research advances on the role of AS‑lncRNAs in tumor occurrence and development were summarized, with the aim of providing novel ideas for further research in this field.

Introduction

Malignant neoplasms collectively are the second leading cause of mortality globally, imposing a significant economic burden on public healthcare systems around the world (1). By 2022, China had ~4.82 million new cancer cases, with 3.21 million cases of cancer-associated mortality (2). Compared with USA and UK, China has a lower cancer incidence rate but a higher cancer mortality rate (3). Although there had been a gradual decline in the incidence of gastric cancer (GC), hepatocellular carcinoma (HCC) and esophageal cancers, the morbidity and mortality rates of colorectal cancer (CRC), prostate and breast cancers have seen a progressive rise (4). Although notable advances in surgical, radiotherapeutic chemotherapeutic, targeted therapeutic, immunotherapeutic and multimodality therapeutic methods have all led to significant progress in enhancing treatment efficacy (5), the overall therapeutic effect remains poor in the long-term. Therefore, further studies are required to explore the mechanisms that regulate tumor development and responses to therapy.

Non-coding RNAs form a family that includes microRNAs (miRNA or miR), long non-coding RNAs (lncRNAs) and circular RNAs, which are classified according to their nucleotide length (6,7). In particular, lncRNAs are typically >200 nucleotides in length and can form conserved secondary structures, although the majority of which do not encode proteins (8). lncRNAs are structurally similar to mRNAs, since they also contain promoters and can undergo splicing, capping, polyadenylation and editing (9). In addition, their expression is regulated in a time- and space-specific manner (9). lncRNAs have been reported to regulate gene expression upstream of various biological processes, including cell proliferation and metabolism (10–13). lncRNAs can be classified into the following six categories based on their location on the genome: i) Intergenic lncRNAs, which are transcripts located between two protein-coding genes; ii) intronic lncRNAs, which are located within an intron of a coding gene; iii) bidirectional lncRNAs, which are located within 1 kb of promoters in the opposite direction from the protein-coding transcript; iv) enhancer lncRNAs, which are located in the enhancer regions; v) sense lncRNAs, located in a small segment of a coding gene, which is the overlapping region of one or several introns or exons; and vi) antisense-lncRNAs (AS-lncRNAs), which are transcribed from the antisense strand of protein-coding genes and overlap one or several introns and exons of the sense sequence (14). AS-lncRNAs are transcribed from the opposite strand of a protein or non-protein-coding gene (15). AS-lncRNAs have also been previously observed to serve an important role in the development and progression of malignancies (16–18). In the present review, the mechanism underlying the regulation of target gene expression by AS-lncRNAs was described, before its downstream consequences in tumor occurrence and development were summarized. Finally, the possible association between AS-lncRNA expression and therapy resistance in cancers was reported.

Mechanism underlying antisense-long-stranded non-coding RNA (AS-lncRNA)-mediated regulation of gene expression

AS-lncRNAs typically do not translate into proteins that can exert biological effects, but it can control gene expression through other means (8). AS-lncRNAs can mainly regulate the expression of target genes through eight methods (Fig. 1), which are summarized in this section.

Figure 1.

AS-lncRNAs can regulate tumor gene expression through endogenous competition mechanism, promoter interaction, direct interaction with mRNA, acting as ‘scaffold’ to change the half-life of mRNA, interaction with 5-UTR, regulation of meaningful mRNA, positive feedback loop of ‘AS-lncRNA-mediator protein-meaning gene’ model, transcriptional regulation in nucleus and so on. AS-lncRNAs can regulate tumor by regulating tumor cell death and proliferation, energy metabolism, inflammatory cancer transformation, invasion and migration, and regulation of tumor neovascularization. AS-lncRNA, antisense-long-stranded non-coding RNA; UTR, untranslated regions; ceRNA, competing endogenous RNA; EMT, epithelial-mesenchymal transition.

Through competing endogenous RNAs (ceRNAs)

A number of studies have previously reported that there is a regulatory relationship between AS-lncRNAs and miRNAs. Specifically, AS-lncRNAs can negatively regulate the expression of miRNAs upstream of the occurrence and development of tumors through the ceRNA mechanism (19), namely by serving as an endogenous miRNA sponge (20). Si et al (21) found that zinc finger protein (ZNF)561-AS1 expression was increased in tissues from patients with CRC and mediated carcinogenic effects in CRC, which predicted poor prognosis in patients. Serine and arginine rich splicing factor 6 (SRSF6) is a proto-oncogene that was also found to be overexpressed in CRC. In addition, ZNF561-AS1 was found to be an upstream post-transcriptional regulator of SRSF6. Using Starbase and Targetscan bioinformatics analyses, ZNF561-AS1 and SRSF6 were found to have potential binding sites for miR-26a-5p and miR-128-3p. The results of agonaute 2 (ago2)-RNA immunoprecipitation (RIP) assay and luciferase reporter assays revealed that ZNF561-AS1 was a ceRNA of miR-26a-5p and miR-128-3p. Further luciferase reporter assays later found that ZNF561-AS1 knockdown significantly inhibited SRSF6 wild-type plasmid luciferase activity. However, transfection with miR-26a-5p and/or miR-128-3p inhibitors was able to either partially or completely rescue such luciferase activity in CRC cells with ZNF561-AS1 expression knocked down. This led to the conclusion that ZNF561-AS1 can promote the expression of SRSF6 by functioning as a sponge for miR-26a-5p and miR-128-3p, thereby promoting the proliferation of CRC cells. In a different study, Shuai et al (22) found that lncRNA motor neuron and pancreas homeobox 1 (MNX1)-AS1 expression was increased in GC tissues, suggesting poor patient prognosis. Subsequent ago2-RIP assay results suggested that MNX1-AS1 may serve as a ceRNA. RNA-sequencing and reverse transcription-quantitative PCR assays found that Bcl-2 expression was significantly decreased in GC cells with MNX1-AS1 expression knocked down. MiRanda prediction then revealed that Bcl-2 was likely to be candidate target to bind to miR-6785-5p. Luciferase reporter gene assay then found miR-6785-5p to bind to the Bcl-2 3′untranslated region (3′UTR), suggesting that miR-6785-5p can target Bcl-2 mRNA. Results from both bioinformatics analyses and luciferase reporter assays suggested that lncRNA MNX1-AS1 can also serve as a ceRNA of miR-6785-5p to upregulate the expression of Bcl-2 in GC cells by sponging miR-6785-5p, thereby promoting GC progression.

Interacting with promoters

A number of pathways that can promote the co-expression of AS-lncRNAs and nearby protein-coding genes have been reported (23,24). On the transcriptional level, one possible mechanism is that AS-lncRNAs share a promoter with neighboring protein-coding genes (25), where they can regulate the H3K4me3 enrichment state in the promoter regions of genes of interest to in turn modulate their expression (26). Bartl et al (27) previously reported that the lncRNA hedgehog-interacting protein 1 (HHIP)-AS1 and HHIP share a promoter, whereby promoter activity was negatively regulated by DNA methylation. By contrast, Shuai et al (22) found that lncRNA MNX1-AS1 was overexpressed in GC tissues, which was not conducive to GC prognosis. Gene Ontology analysis revealed that the potential targets of MNX1-AS1 are possibly involved in various biological processes, such as signal transduction, cell proliferation and cell death. Chromatin immunoprecipitation (ChIP) experiments subsequently revealed that enhancer of zeste homolog 2 (EZH2) can bind to the promoter region of B-cell translocation gene anti-proliferation factor 2 (BTG2) to induce H3K27me3 modification in GC cells, which inhibits BTG2 transcription. Subsequent rescue experiments revealed that BTG2 can regulate the proliferation, migration and invasion of GC cells induced by MNX1-AS1. These results suggested that lncRNA MNX1-AS1 can suppress the expression of BTG2 in GC through EZH2-mediated H3K27me3 to promote GC progression.

Direct interactions alter the half-life of mRNAs

AS-lncRNAs can regulate the stability of sense RNAs by forming RNA duplexes with them (28). Pan and Xie (29) previously observed through subcellular fractionation and fluorescence in situ hybridization assays that the lncRNA forkhead box C2 (FOXC2)-AS1 is mainly located in the nucleus where its expression is increased in CRC tissues, which indicates poor patient prognosis. FOXC2 is the closest gene to FOXC2-AS1, where their sequences overlap. The expression of FOXC2 and FOXC2-AS1 in CRC tissues was also found to be positively correlated. FOXC2-AS1 was subsequently found to positively regulate the expression of FOXC2. Reverse transcription-quantitative PCR (RT-qPCR) assay results suggested that FOXC2-AS1 can enhance the stability of FOXC2 mRNA. According to the site and gene structure, exon 1 of FOXC2-AS1 was completely complementary to FOXC2. RNase protection assay then revealed that FOXC2-AS1 and FOXC2 formed a double-stranded RNA structure, which was confirmed by RNA pull-down assay. This resulted in the conclusion that FOXC2-AS1 and FOXC2, and hence AS-lncRNAs and mRNAs, can form double-stranded structures, which enhanced the stability of FOXC2 mRNA (27). In addition, Zhao et al (30) found that metastasis-associated in colon cancer protein 1 (MACC1)-AS1 expression was increased in GC tissues, indicating a poor prognosis. MACC1-AS1 was found to promote metabolic plasticity by regulating MACC1 expression, thereby promoting GC progression. Through bioinformatics analysis, it was found that MACC1-AS1 contained the binding site of MACC1 mRNA. MACC1-AS1 was predominantly found to localize in the cytoplasm, allowing for the regulation of mRNA stability. Subsequent RT-qPCR analysis found that MACC1-AS1 significantly reduced the degradation of MACC1 mRNA, whilst affinity pull-down assays confirmed the direct interaction between MACC1-AS1 and MACC1. These findings suggested that MACC1-AS1 promoted mRNA stability through direct binding, thereby promoting GC progression.

Functioning as a ‘scaffold’ to regulating the half-life of mRNA

AS-lncRNAs can serve as scaffolds to interact with various proteins to form RNA/protein complexes (31). Zhu et al (32) found that Rho GTPase-activating protein 5 (ARHGAP5)-AS1 expression was increased in GC cells. Clinical data from The Cancer Genome Atlas (TCGA) database found that higher ARHGAP5-AS1 expression levels were associated with shorter overall survival. m6A modifications were previously found to regulate mRNA stability through RNA-binding proteins (33). Zhu et al (32) used RMBase and SRAMP analyses to show that there may be m6A modification in ARHGAP5 mRNA. When ARHGAP5-AS1 was overexpressed, the binding of both m6A modification and methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit (METTL3) to ARHGAP5 mRNA were found to be elevated. Taken together, ARHGAP5-AS1 may recruit METTL3 to promote the m6A modification of ARHGAP5 mRNA, thereby stabilizing ARHGAP5 mRNA in the cytoplasm and promoting GC chemoresistance.

In summary, AS-lncRNAs can regulate gene expression through a variety of mechanisms, including ceRNA mechanisms, interaction with promoters and direct interaction or indirect interaction with mRNAs by serving as ‘scaffolds’ to alter its half-life.

Interacting with 5′-UTRs

Tran et al (34) reported an AS-lncRNA, AS-putative RNA-binding protein 15 (RBM15). The sequences of RBM15 and RBM15-AS1 both were first analyzed, which found that exon 1 of RBM15-AS1 coincided with the 5′-UTR of RBM15. The main functional region of lncRNA was then searched further, before full-length and multiple truncated versions were constructed. It was found that the 5-end of RBM15 was the main functional region, which could promote differentiation. The relationship between RBM15-AS1 and RBM15 was then verified experimentally, revealing that the shortest functional region exon 1 of RBM15-AS 1 could promote the translation of RBM15 without significantly affecting its mRNA levels. By constructing a dual-luciferase reporter system with the 5′-UTR of RBM15 and constructs encoding the different truncated versions of the lncRNAs, it was found that the region overlapping the 5′-UTR of AS-RBM15 and RBM15 was responsible for the upregulation of RBM15 protein translation.

Exerting regulatory effects on sense mRNAs

Alternative splicing is a common phenomenon during post-transcriptional regulation, which produces different mRNA variants and unique isoforms of the same protein (35,36). Yuan et al (37) previously identified an oncofetal splicing factor, muscleblind-like protein 3 (MBNL3), which promoted HCC tumorigenesis and suggested poor prognosis in patients. Transcriptomic analysis revealed that MBNL3 induced lncRNA PXN-AS1 exon 4 encapsulation. Transcripts of lncRNA PXN-AS1 with exon 4 deleted were found to bind to the coding sequence of PXN mRNA, leading to translation elongation factor separation from the PXN mRNA to inhibit PXN translation. By contrast, exon 4-containing transcripts preferentially bind to the 3′ UTRs of PXN mRNA to protect the PXN mRNA from miR-24-ago2 complex-induced degradation, which in turn increases PXN expression. Through this mechanism, MBNL3 was concluded to upregulate PXN expression by promoting exon 4 inclusion, where PXN mediated the pro-tumor effects of MBNL3 in HCC.

Positive feedback loop of ‘AS-lncRNA/protein/gene of interest’ model

AS-lncRNA/protein/gene of interest can form stable complexes to continuously promote the expression of AS-lncRNAs and sense genes (38). Luo et al (39) previously found that zinc finger E-box binding homeobox 1 (ZEB1)-AS1 and ZEB1 expression is increased in triple-negative breast cancer (TNBC), both of which demonstrated direct binding to embryonic lethal, abnormal vision, drosophila, homolog-like 1 (ELAVL1) according to pull-down silver staining and RIP experiments. Specifically, ZEB1-AS1 directed ELAVL1 to the ZEB1 mRNA to protect it from degradation. ChIP experiments then demonstrated that ZEB1 exhibited potent binding affinity to part 1 of the ZEB1-AS1 promoter. ZEB1 could bind to the promoter of ZEB1-AS1 to promote the activation of ZEB1-AS1 expression, which was also verified by luciferase reporter assays. This suggested that ZEB1-AS1 interacted with ZEB1, which then formed a positive feedback loop with ELAVL1 to amplify the promotion of TNBC.

Transcriptional regulation of AS-lncRNAs in the nucleus

AS-lncRNAs can regulate gene expression on a post-transcriptional level by interacting with RNA-binding proteins to regulate mRNA stability (25). Wang et al (40) previously found that EGFR-AS1 expression was significantly increased in renal cell carcinoma tissues. Based on literature search results and sequence complementarity between EGFR and EGFR-AS1, the aforementioned study then confirmed that EGFR-AS1 can directly bind to EGFR mRNA to inhibit its degradation. In addition, subsequent RNA pull-down experiments and mass spectrometry analysis revealed that EGFR-AS1 interacted with human antigen R (HuR), which was the reason for the stability of EGFR mRNA. In summary, EGFR-AS1 regulated gene expression on the post-transcriptional level in renal cell carcinoma by enhancing the stability of EGFR mRNA in a HuR dependent manner.

In conclusion, this section mainly summarized the mechanism by which AS-lncRNAs can regulate gene expression.

Effect of AS-lncRNAs on tumors and underlying mechanism

Accumulating evidence has revealed that AS-lncRNAs are involved in tumorigenesis and development, where they serve important roles as either tumor suppressors or oncogenes (Table I). They can mediate a variety of processes, including cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), apoptosis, drug resistance and immune escape (Fig. 1) (41). lncRNA MBNL1-AS1 and ZNF667-AS1 have been documented to be tumor suppressors (42,43), whilst lncRNA homeobox A11 (HOXA11)-AS, integrin subunit β1-AS1 were reported to be oncogenes (44,45).

Table I.

AS-lncRNAs play a role as tumor suppressor or oncogene.

Table I.

AS-lncRNAs play a role as tumor suppressor or oncogene.

TumorAS-lncRNAUpregulatedDownregulated
Colorectal cancerZNF561-AS1√
Colorectal cancerFOXC2-AS1√
Colorectal cancerSLCO4A1-AS1√
Colorectal cancerRAD51-AS1 √
Colorectal cancerDNAJC3-AS1√
Colorectal cancerSATB2-AS1 √
Colorectal cancerSTEAP3-AS1√
Colorectal cancerLDLRAD4-AS1√
Colorectal cancerZNF667-AS1 √
Colorectal cancerSLCO4A1-AS1√
Colorectal cancerZEB1-AS1√
Colorectal cancerLOXL1-AS1√
Colorectal cancerDLGAP1-AS1√
Colorectal cancerOIP5-AS1√
Colorectal cancerPGM5-AS1 √
Colorectal cancerELFN1-AS1√
Colorectal cancerTTN-AS1√
Colorectal cancerLBX2-AS1√
Colorectal cancerMFI2-AS1√
Colorectal cancerFOXD3-AS1√
Colorectal cancerFGD5-AS1√
Gastric cancerMNX1-AS1√
Gastric cancerMACC1-AS1√
Gastric cancerARHGAP5-AS1√
Gastric cancerOIP5-AS1√
Gastric cancerADAMTS9-AS2 √
Gastric cancerBDNF-AS√
Gastric cancerMACC1-AS1√
Gastric cancerMSC-AS1√
Gastric cancerELF3-AS1 √
Gastric cancerMBNL1-AS1 √
Gastric cancerNKX2-1-AS1√
Gastric cancerHNF1A-AS1√
Gastric cancerSAMD12-AS1√
Gastric adenocarcinomaHAND2-AS1 √
Gastric adenocarcinomaPGM5-AS1 √
Gastric cancerCCDC144NL-AS1√
Gastric cancerNUTM2A-AS1√
Gastric cancerVCAN-AS1√
Gastric cancerDLX6-AS1√
Gastric cancerNKX2-1-AS1√
Gastric cancerGATA6-AS √
Hepatocellular carcinomaPXN-AS1√
Hepatocellular carcinomaIGF2-AS √
Hepatocellular carcinomaGABPB1-AS1 √
Hepatocellular carcinomaRAB11B-AS1 √
Hepatocellular carcinomaHMMR-AS1√
Hepatocellular carcinomaKDM4A-AS1√
Hepatocellular carcinomaCCDC144NL-AS1√
Hepatocellular carcinomaMCM3AP-AS1√
Hepatocellular carcinomaCDKN2B-AS1√
Hepatocellular carcinomaBZRAP1-AS1 √
Hepatocellular carcinomaF11-AS1 √
Hepatocellular carcinomaZFPM2-AS1√
Hepatocellular carcinomaSBF2-AS1√
Hepatocellular carcinomaSLC2A1-AS1 √
Hepatocellular carcinomaGAS6-AS1√
Hepatocellular carcinomaUPK1A-AS1√
Hepatocellular carcinomaKTN1-AS1√
Hepatocellular carcinomaCACNA1G-AS1√
Hepatocellular carcinomaAGAP2-AS1√
Hepatocellular carcinomaFOXD2-AS1√
Hepatocellular carcinomaFEZF1-AS1√
Renal cell carcinomaEGFR-AS1√
Renal cell carcinomaENTPD3-AS1 √
Renal cell carcinomaLOXL1-AS1√
Renal cell carcinomaZFPM2-AS1√
Renal cell carcinomaFGD5-AS1√
Renal cell carcinomaZNF582-AS1 √
Renal cell carcinomaDARS-AS1√
Renal cell carcinomaNNT-AS1√
Renal cell carcinomaDNAJC3-AS1√
Renal cell carcinomaCDKN2B-AS1√
Renal cell carcinomaITGB2-AS1√
Renal cell carcinomaMSC-AS1√
Renal cell carcinomaARAP1-AS1√
Renal cell carcinomaTP73-AS1√
Renal cell carcinomaOTUD6B-AS1 √
Renal cell carcinomaDLX6-AS1√
Renal cell carcinomaADAMTS9-AS2 √
Renal cell carcinomaPCED1B-AS1√
Triple negative breast cancerZEB1-AS1√
Triple negative breast cancerHAND2-AS1 √
Triple negative breast cancerDLX6-AS1√
Triple negative breast cancerWEE2-AS1√
Triple negative breast cancerHLA-F-AS1√
Triple negative breast cancerFAM83H-AS1√
Triple negative breast cancerLRP11-AS1√
Triple negative breast cancerAFAP1-AS1√
Breast cancerVCAN-AS1√
Breast cancerA1BG-AS1√
Breast cancerCERS6-AS1√
Breast cancerZBED3-AS1 √
Breast cancerHNF1A-AS1√
Breast cancerMIF-AS1√
Breast cancerDSCAM-AS1√
Breast cancerADPGK-AS1√
Breast cancerMCM3AP-AS1√
Breast cancerMBNL1-AS1 √
Breast cancerOIP5-AS1√
Breast cancerHOXD-AS1√
Breast cancerFBXL19-AS1√
Breast cancerTFAP2A-AS1 √
Breast cancerMAFG-AS1√
Breast cancerCDKN2B-AS1√
LeukemiaAS-RBM15√
LeukemiaUSP30-AS1√
LeukemiaSATB1-AS1√
LeukemiaCD27-AS1√
LeukemiaSBF2-AS1√
LeukemiaFBXL19-AS1√
LeukemiaGAS6-AS1√
LeukemiaMBNL1-AS1 √
LeukemiaTP73-AS1√
LeukemiaPRR34-AS1√
LeukemiaITGB2-AS1√
LeukemiaLEF1-AS1 √
LeukemiaZEB2-AS1√
LeukemiaLAMP5-AS1√
LeukemiaGAS6-AS1√
LeukemiaDNAJC3-AS1√
GliomaGATA6-AS√
GliomaFAM181A-AS1√
GliomaPCED1B-AS1√
GliomaFOXD2-AS1√
GliomaDLX6-AS1√
GliomaFLVCR1-AS1√
GliomaGAS5-AS1 √
GliomaAGAP2-AS1√
GliomaTP73-AS1√
GliomaPSMB1-AS1√
GliomaOIP7-AS5√
GliomaMATN1-AS1√
GliomaHOXD-AS1√
GliomaHOXA11-AS√
GliomaSOX21-AS1√
GliomaZEB1-AS1√
GliomaTPT1-AS1√
GliomaEGFR-AS1√
GliomaFOXD1-AS1√
GlioblastomaWEE2-AS1√
GlioblastomaUBA6-AS1
GlioblastomaTP73-AS1√
Squamous cell carcinoma of the hypopharynxHOXA11-AS1√
Squamous cell carcinoma of the hypopharynxLEF1-AS1√
Squamous cell carcinoma of the hypopharynxMNX1-AS1√
Nasopharyngeal carcinomaDLX6-AS1√
Nasopharyngeal carcinomaSMAD5-AS1√
Nasopharyngeal carcinomaHOXC13-AS√
Nasopharyngeal carcinomaPTPRG-AS1√
Nasopharyngeal carcinomaAFAP1-AS1√
Nasopharyngeal carcinomaTP73-AS1√
Nasopharyngeal carcinomaDLX6-AS1√
Nasopharyngeal carcinomaOIP5-AS1√
Nasopharyngeal carcinomaHOXC-AS1√
Nasopharyngeal carcinomaFEZF1-AS1√
Nasopharyngeal carcinomaFOXP4-AS1√
Nasopharyngeal carcinomaFOXD2-AS1√
OsteosarcomaOIP5-AS1√
OsteosarcomaCCDC144NL-AS1√
OsteosarcomaPCED1B-AS1√
OsteosarcomaPGM5-AS1√
OsteosarcomaTTN-AS1√
OsteosarcomaRHPN1-AS1√
OsteosarcomaNR2F1-AS1√
OsteosarcomaDLX6-AS1√
OsteosarcomaC5ORF66-AS1√
OsteosarcomaASB16-AS1√
OsteosarcomaADPGK-AS1√
OsteosarcomaAFAP1-AS1√
OsteosarcomaHNF1A-AS1√
OsteosarcomaRUSC1-AS1√
OsteosarcomaFBXL19-AS1√
OsteosarcomaMSC-AS1√
OsteosarcomaLBX2-AS1√
OsteosarcomaFOXD2-AS1√
OsteosarcomaNNT-AS1√
OsteosarcomaDSCAM-AS1√

[i] AS-lncRNA, antisense-long-stranded non-coding RNA.

Regulation tumor cell death and proliferation

Programmed cell death mainly includes apoptosis, autophagy, pyroptosis and ferroptosis pathways (46,47). Results from several studies have shown that AS-lncRNAs can regulate tumor cell death and proliferation through the aforementioned pathways (48–51).

Apoptosis

AS-lncRNAs can reduce cell cycle arrest and apoptosis and promote cell proliferation (48). Opa-interacting protein 5 (OIP5)-AS1 overexpression was previously found to downregulate nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing (NLRP)6 by interacting with EZH2 to promote GC cell proliferation whilst suppressing apoptosis (52). Wang et al (53) also reported that OIP5-AS1 can promote cell proliferation by accelerating cell cycle progression in GC cells through sponging miR-641 to increase the expression of cyclin D1 and activation of AKT (53). In a different study, OIP5-AS1 was found to promote GC cell proliferation but inhibited apoptosis by regulating the miR-367-3p/high mobility group AT-hook 2 axis and activating the PI3K/AKT and Wnt/β-catenin pathways (54). In glioma, Liao et al (55) found that the expression of GATA6-AS was significantly upregulated, where it promoted proliferation and inhibited apoptosis through taurine upregulated 1.

Autophagy

Autophagy serves a dual role in the physiology of a wide variety of cancer types (56,57). In cancer cells, autophagy can inhibit tumorigenesis by preventing cancer cell survival and inducing cell death (58), but it can also promote tumorigenesis by promoting cancer cell proliferation and tumor growth (59). Wang and Jin (49) previously detected that solute carrier organic anion transporter family member 4A1 (SLCO4A1)-AS1 expression levels in patients with CRC were positively associated with Par-3 family cell polarity regulator (PARD3) expression. PARD3 protein is a key molecule that can trigger autophagy initiation. SLCO4A1-AS1 was also found to promote autophagy and the proliferation of CRC cells both in vitro and in vivo. Overexpression and knockdown experiments revealed that SLCO4A1-AS1 functioned as a ceRNA to induce autophagy and promote the proliferation of CRC cells by sponging endogenous miR-508-3p, which promoted PARD3 protein expression (47).

Pyroptosis

Pyroptosis is a relatively recently discovered form of programmed cell death mode that is mediated by the gasdermin protein family (60), which also serves a duplicitous role in tumor progression (61). Previous studies have shown that pyroptosis is involved in the growth, differentiation, invasion and late metastasis of cancer (62). Components of pyroptosis have been reported to serve an important carcinogenic role in GC progression (63). Previously, AS-lncRNAs have been found to be key regulators of apoptosis (50). Wang et al (64) found that heart and neural crest derivatives-expressed 2-AS1 and phosphoglucomutases (PGM5)-AS1 were independently associated with pyroptosis in GC, where they appeared to participate in the occurrence and development of GC according to co-expression analysis and Kaplan-Meier survival analysis (64). In addition, Ren et al (65) found that the overexpression of lncRNA A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)9-AS2 inhibited GC progression by regulating the miR-223-3p/NLRP3 axis-mediated pyroptosis.

Ferroptosis

Ferroptosis is another recently discovered form of cell death that is different from apoptosis, necrosis and autophagy mechanistically (66). In particular, epigenetic mechanisms, such as DNA methylation, histone modifications and AS-lncRNAs, have been reported to serve roles in ferroptosis (67,68). Huang et al (51) previously found that reactive oxygen species (ROS), Fe2+ and total iron concentrations were decreased after overexpressing brain-derived neurotrophic factor (BDNF)-AS following RT-PCR detection. By contrast, ROS, Fe2+ and total iron concentrations were revealed to be increased after BDNF-AS expression was knocked down. A ferroptosis model was then established using erastin and Ras-selective lethal 3 (RSL3), where it was found that the changes in the level of ferroptosis markers induced by erastin were more significant compared with those induced by RSL3. A different previous study also found that erastin can induce ferroptosis by binding to voltage-dependent anion-selective channel protein (VDAC)2/VDAC3 (68). The protein expression profile in GC tissues studied by Huang et al (51) revealed that VDAC2/VDAC3 was abnormally expressed in GC samples. However, VDAC3 protein expression was found to be significantly increased when BDNF-AS was overexpressed but was decreased when BDNF-AS expression was knocked down, compared with VDAC2 (49). These results suggested that BDNF-AS can regulate ferroptosis in GC cells through VDAC3. In conclusion, the aforementioned results from previous studies suggested that when BDNF-AS was overexpressed, the degree of ferroptosis in GC cells was reduced, which favored GC invasion and metastasis.

Regulation of energy metabolism reprogramming in tumor cells

In cancer cells, glucose metabolism is reprogrammed to adapt to the increased energy requirements. Unlike in normal cells, the main metabolic process in cancer cells is glycolysis (69). The predominance of aerobic glycolysis is a key metabolic feature of the Warburg phenotype, which is caused by the active metabolic reprogramming required to support the sustained cancer cell proliferation and malignant progression (70). Since the German scientist Otto Warburg put forward the ‘Warburg effect’ (71), the relationship between aerobic glycolysis and tumorigenesis has become a topic of intense scientific research (72). The mechanism of glucose metabolism in cancer cells can be summarized mainly as follows (70): i) Overexpression of glucose transporters and key glycolytic enzymes, which accelerates the glycolytic flux, followed by the accumulation and transfer of glycolytic intermediates for cancer biomass synthesis; ii) high-speed ATP production to meet the energy demand; and iii) accumulation of lactic acid, which promotes tumor progression, greatly contributing to tumor acidosis. Therefore, the Warburg effect is one of the core factors in underlying the mechanism of cancer progression. Zhao et al (30) previously found that in GC cells MACC1-AS1 significantly upregulated the expression of glycolysis components glucose transporter 1 (GLUT1), hexokinase 2 (HK2), glucose-6-phosphate dehydrogenase and lactate transporter monocarboxylate transporter 1 on the mRNA level, whilst significantly upregulating the expression of GLUT1, HK2 and lactate dehydrogenase (LDH) on the protein level. In addition, following 2-NBDG assays, MACC1-AS1 was found to accelerate glucose uptake in GC cells. Under either normal conditions or those of glucose deprivation, MACC1-AS1 also increased the production of ATP and lactic acid, presumably by increasing the activity of key glycolytic enzymes HK2 and LDHA. These results suggested that a potential role of MACC1-AS1 is to promote glycolysis by upregulating the activity and expression of glycolytic enzymes in GC cells. This metabolic transformation may serve to improve cell viability and resistance to apoptosis, which in turn promotes the progression of GC. In a previous study, Li et al (73) found that the overexpression of RAD51-AS1 could reduce glucose uptake by CRC cells, which led to a reduction in lactic acid concentrations in CRC cells. Subsequently, through western blotting, it was found that the RAD51-AS1 overexpression led to the reduction in HK2 and GLUT1 levels, suggesting that RAD51-AS1 can inhibit the glycolytic process in CRC cells. In conclusion, these results strongly suggested that RAD51-AS1 serves an important role in maintaining the glycolysis balance, which may affect the proliferation of CRC.

Lipid metabolism reprogramming has also been proposed to be a marker of cancer (74). In rapidly proliferating tumor cells, the re-synthesis of fatty acids is enhanced, such that the expression of key fatty acid synthases, including acetyl-CoA carboxylase (ACC) and fatty acid synthase (FASN), is increased. This subsequently results in the accumulation of a variety of fatty acids (75). Accumulating lipids can promote cell proliferation and tumor progression in a complex tumor microenvironment (76). Previous studies have demonstrated that fibroblasts in the tumor microenvironment express FASN highly (76), which mediates lipid metabolism reprogramming, resulting in the accumulation of various fatty acids to promote the migration of CRC cells (76). EGFR/PI3K/AKT signaling has been shown to activate sterol regulatory element-binding protein-1 cleavage and upregulate the expression of ACC1 and FASN, resulting in increased lipid metabolism (77,78). Tang et al (79) previously found that interferon-induced, double-stranded RNA-activated protein kinase inhibitor-AS1 regulated fatty acid synthase through the EGFR/PI3K/AKT/NF-κB signal pathway to promote the progression of CRC. Fatty acid is an important energy source through fatty acid oxidation (FAO), which has been shown to be necessary for the proliferation and survival of cancer cells (80). Inhibition of FAO can inhibit cell stemness and alleviate tumor growth (81). Mesenchymal stem cells (MSCs) have been revealed to promote the dryness of GC cells and predict the poor prognosis of GC. He et al (82) found that inhibition of FAO could reverse the MSC-mediated stemness of GC cells, suggesting that FAO is involved in the self-renewal of GC stem cells induced by MSCs, whilst having little effect on non-stem cells.

Regulation of inflammation in cancer

A large number of studies have shown that inflammatory immune cells can serve diverse but key roles in promoting tumorigenesis (83). Tumor-promoting inflammatory cells mainly include macrophages, mast cells, neutrophils, T and B lymphocytes (84). Nie et al (85) employed the Gene Set Enrichment Analysis (GSEA) pre-ranking of tumor characteristics to find that there was a decrease in memory B cells but an increase in naive and effector CD8T cells in the high-risk group, suggesting immune-related regulation. Tumor-infiltrating inflammatory cells have been demonstrated to induce and then maintain tumor angiogenesis (86), stimulate cancer cell proliferation, promote foreign tissue invasion and support cell migration (84,87). AS-lncRNAs have correspondingly been detected to regulate inflammation in tumors and cancer transformation, by regulating the expression of genes associated with the immune response. This was proposed to be exerted through the synthesis of inflammatory factors and inflammatory signaling pathways (88). In addition, AS-lncRNAs have been reported to alter the profile of immune cell infiltration in the tumor microenvironment, mediating a profound impact on tumor invasiveness, progression and prognosis (89). Xu et al (90) found that the expression of the CRC tissue-specific lncRNA special AT-rich sequence-binding protein 2 (SATB2)-AS1 was downregulated in CRC tissues, which predicted a favourable prognosis. Transwell and wound healing assays demonstrated that SATB2-AS1 knockdown significantly increased CRC cell migration and invasion ability. The results of GSEA also revealed that the expression of SATB2-AS1 was associated with various immune responses. In addition, it was found that when SATB2-AS1 was negatively associated with the expression of Th1 chemokines C-X-C motif chemokine ligand (CXCL)9 and CXCL10, these chemokines could be induced by IFN-γ and mediated effector T cell transport. In conclusion, these results suggested that SATB2-AS1 is a mediator of the CRC immune response by regulating immune cell density in CRC to inhibit CRC cell metastasis, slowing the progression of CRC.

Regulation of EMT, invasion and migration in tumor cells

One of the key mechanisms for cancer cells to enhance their invasive ability is by detaching from their intercellular adhesion, followed by the acquisition of a more dynamic mesenchymal phenotype as part of the EMT process (91). EMT has been previously considered to be a binary process with two distinct cell groups, namely epithelial cells and mesenchymal cells, characterized by the loss of the epithelial marker E-cadherin and the increased expression of the mesenchymal marker vimentin (92,93). During EMT, epithelial cells lose apical-basal polarity and E-cadherin expression, whilst upregulating the expression of mesenchymal biomarkers, including twist, slug, snail, vimentin and N-cadherin (94,95). EMT has been associated with a number of tumor functions, including tumor initiation, malignant progression, tumor stemness, cell migration and metastasis (92). In addition, EMT is considered to be a key factor on worsening patient prognosis by accelerating the invasion and migration of cancer cells (96).

Yu et al (97) previously observed that lncRNA SLCO4A1-AS1 expression was significantly increased in CRC tissues, which was in turn associated with poor patient prognosis and CRC tumor metastasis. By knocking down SLCO4A1-AS1 expression, it was then detected that SLCO4A1-AS1 inhibited the migration, invasion and EMT of CRC cells in vitro. In a different study, Zhou et al (98) found that SLCO4A1-AS1 activated Wnt/β-catenin signaling in CRC cells, which promoted cell migration and invasion. The authors explored TCGA database for antisense lncRNAs involved in CRC under hypoxic conditions. Among the candidate genes found, lncRNA six-transmembrane epithelial antigen of prostate 3 (STEAP3)-AS1 was reported to be aberrantly transcribed under hypoxic conditions in clinical CRC tissues, where it was positively associated with poor patient prognosis (96). Furthermore, it was revealed that lncRNA STEAP3-AS1 interacted with YTH N6-methyladenosine RNA-binding protein 2 to stabilize STEAP3 mRNA, which increased the expression of the STEAP3 protein. The STEAP3 protein then activated Wnt/β-catenin signaling in an iron-dependent manner to promote cell invasion and migration, in turn accelerating CRC progression (96).

A previous independent study of RNA-sequencing data found that in A549 and HeLa cells, downregulation of E74-like ETS transcription factor 3 (ELF3)-AS1 led to the upregulation of snail family transcriptional repressor (SNAI)2 mRNA expression, suggesting that the negative regulation of SNAI2 expression by ELF3-AS1 may be widespread in cancer (99). In addition, Li et al (26) identified 123 lncRNAs regulated by SNAI2 in GC by RNA sequencing, where both the ELF3 gene and ELF3-AS1 were found to be transcriptionally repressed by EMT-associated transcription factors SNAI2 or SNAI1. In addition, the gene expression profiles produced by SNAI2 overexpression and ELF3-AS1 knockdown were revealed to be highly very similar. These results suggested that knocking down ELF3-AS1 expression cannot only upregulate the expression of SNAI2, but can also activate the downstream signal pathways of SNAI2 in GC. Cell proliferation, Transwell and wound healing experiments revealed that downregulation of ELF3-AS1 promoted the proliferation, migration and invasion of GC cells, indicating poor patient GC prognosis. In conclusion, ELF3-AS1 was suggested to inhibit EMT mainly by inhibiting the SNAI2-mediated transduction of GC-associated transcription factors. Mo et al (100) previously found that the higher expression levels of lncRNA low density lipoprotein receptor class A domain-containing 4 (LDLRAD4)-AS1 promoted the progression of CRC, which is associated with poor patient prognosis. Subsequent western blotting and immunohistochemistry results revealed that lncRNA LDLRAD4-AS1 could promote EMT both in vitro and in vivo. lncRNA LDLRAD4-AS1 was then observed to promote EMT by reducing the expression of LDLRAD4, promoting the process of GC.

The TGF-β/EMT pathway is closely associated with cancer progression (101). In particular, Smad7 is activated by all members of the TGF-β superfamily and has a negative regulatory effect on the TGF-β signal pathway (102). Su et al (42) previously reported that MBNL1-AS1 knockdown can increase the proliferation, migration and invasion of GC cells according to in vitro experimental results and animal models. MBNL1-AS1 expression was demonstrated to be downregulated in GC tissues and cells, which played a positive role in inhibiting the growth of the GC tumor. Western blotting data demonstrated that MBNL1-AS1 affected the TGF-β/SMAD pathway by regulating the miR-424-5p/Smad7 axis, which inhibited the proliferation of GC.

Collectively, the aforementioned observations strongly suggested that AS-lncRNAs can regulate EMT through the Wnt, SNAI2 and TGF-β signal pathways, serving an important role in the occurrence and development of various tumors.

Regulation of tumor neovascularization

The plasminogen activator system, which includes urokinase type plasminogen activator (uPA), uPA cell receptor and its specific inhibitor plasminogen activator inhibitor-1 (PAI-1), has been reported to serve an important role in tumor progression and angiogenesis (103). The role of PAI-1 in tumor angiogenesis has been extensively studied, the expression of which is essential for tumor angiogenesis (104). Teng et al (105) found that the overexpression of NK2 homeobox 1 (NKX2)-1-AS1 and PAI-1 was associated with GC progression and prognosis. Specifically, NKX2-1-AS1 served as a ceRNA of miR-145-5p and promoted tumor progression and angiogenesis, by activating VEGFR2 signaling through PAI-1. Previous studies have shown that the PI3K/AKT signaling pathway is involved in cancer cell invasion and angiogenesis (106,107). Liu et al (108) previously detected that hepatic nuclear factor 1 α (HNF1A)-AS1 can promote GC invasion, metastasis and angiogenesis both in vitro and in vivo. In particular, the high degree of PI3K/AKT signaling pathway activation was found to promote the progression of GC. Results from Transwell migration and matrix-based capillary formation assays revealed that overexpression of HNF1A-AS1 can promote GC angiogenesis in vitro. ELISA and rescue experiments confirmed that HNF1A-AS1 exerted its biological function via the PI3K/AKT signaling pathway. The activation of PI3K/AKT signaling in tumor cells could increase the secretion of VEGFA, which serves a role in promoting angiogenesis in human cancers. In conclusion, these aforementioned findings suggested that lncRNA-HNF1A-AS1 can function as a ceRNA of miR-30b-3p to activate PI3K/AKT signaling to increase the secretion of VEGF-A, which promotes angiogenesis and therefore the progression of GC.

AS-lncRNAs in the diagnosis, prognosis and treatment of tumors

AS-lncRNAs have the potential to assist in tumor diagnosis and prognosis

A number of previous studies have demonstrated that AS-lncRNAs have significant potential as a biomarker for both cancer diagnosis and prognosis (10). Guo et al (109) previously found that overexpression of muskelin 1, intracellular mediator containing Kelch motifs-AS enhanced the stability of Yes-associated protein 1 mRNA, which promoted carcinogenic effects. This was proposed to facilitate the diagnosis and prognosis of HCC. In a different previous study, El-Ashmawy et al (110) revealed that family with sequence similarity 83-member H (FAM83H)-AS1 was involved in the progression of various malignant tumors. Coincidentally, it was found to be particularly dysregulated in cancers, potentially serving a crucial role in their diagnosis and prognosis (110). Supporting this, Da et al (111) demonstrated that FAM83H-AS1 may serve a prognostic and diagnostic role in GC.

AS-lncRNAs in the treatment of cancer

AS-lncRNAs have been documented to not only have the potential for cancer diagnosis and prognosis, but may also facilitate the design of treatment strategies for patients with cancer (41). AS-lncRNAs are increasingly becoming biomarkers for the treatment of tumors (45,64,112,113).

Activation of tumor suppressors

Tumor suppressors refer to a large number of molecules that can reliably control cell division, promote apoptosis and inhibit metastasis (114). The loss of tumor suppressor functions may lead to uncontrolled cell division malignant transformation (115). Tumor suppressors mainly exert their roles through the following four main mechanisms: i) Inhibition of cell division; ii) induction of apoptosis; iii) repair of DNA damage; and iv) inhibition of metastasis (116). Lu et al (113) previously observed that the expression of sterile α motif domain-containing 12 (SAMD12)-AS1 was increased in human GC tissues and cell lines. SAMD12-AS1 was then detected to exert its biological role in GC through direct interactions with DNA methyltransferase 1 (DNMT1), which inhibits p53 signaling to promote the malignant transformation of GC cells. Silencing SAMD12-AS1 expression restored p53 signaling to reverse the progression of GC, suggesting that SAMD12-AS1 may serve as a potential diagnostic and therapeutic target for GC. Liu et al (117) previously reported that the expression of HNF1A-AS1 was significantly upregulated in GC tissues. Similar to the function of HNF1A-AS1, the overexpression of early growth response 1 (EGR1) was reported to enhance cell proliferation and promote GC cell cycle progression. The RT-qPCR assay showed that EGR1 promotes the expression of HNF1A-AS1. EGR1 and HNF1A-AS also inhibited the expression of the antibiotic growth factor p21 by promoting cell division cycle 34-mediated ubiquitination and p21 degradation. It was then suggested that the inhibition of EGR1-activated HNF1A-AS1 and the subsequent upregulation of anti-growth factors could inhibit the development of GC. In another study, Zhuang et al (43) previously observed that the lower expression levels of ZNF667-AS1 predicted poorer prognosis and were associated with the progression of CRC. Upregulation of ankyrin 2 by the overexpression of ZNF667-AS1 was then found to inhibit the proliferation, migration and invasion of CRC cells, which hindered the development of CRC progression.

In conclusion, AS-lncRNA could regulate tumor development by activating tumor suppressor factors, which may be exploited as a target for tumor therapy.

Inhibition of oncogene expression

SLCO4A1-AS1 is highly expressed in CRC cells and tissues, which is associated with poor patient prognosis (118). Zhang et al (118) previously documented that SLCO4A1-AS1 increased the expression of CDK2 by enhancing the interaction between CDK2 and heat shock protein 90 (HSP90) and activating c-MYC signaling. This increase in CDK2 expression downstream of SLCO4A1-AS enhanced tumor growth by promoting the phosphorylation of c-Myc at the Ser6 site, activating this protein (115). Downregulation of SLCO4A1-AS1 inhibited the proliferation of CRC cells in vivo and in vitro. Ni et al (119) reported that the expression of ZEB1-AS1 in CRC was significantly upregulated, which was associated with poor prognosis in patients with colon adenocarcinoma. Specifically, ZEB1-AS1 promoted the expression of p21 Protein (Cdc42/Rac)-activated kinase 2 (PAK2) by sponging miR-455-3p, which advanced the proliferation and metastasis of colon adenocarcinoma cells. Knocking down ZEB1-AS expression was then observed to increase the expression of miR-455-3p and decreased the expression of PAK2, which suppressed the proliferation and metastasis of cancer cells. It would be of significance to identify novel therapeutic targets for patients with colon adenocarcinoma. Wu et al (120) previously found that the expression of lysyl oxidase homolog 1 (LOXL1)-AS1 was upregulated in CRC, which then increased the expression of target gene CD44 by sponging miR708-5p. This in turn promoted the expression of EGF to facilitate CRC progression. Knocking down LOXL1-AS1 expression was found to reverse this upregulated CD44/EGFR signaling pathway to inhibit the progression of CRC, providing another potential novel pathway to explore the treatment strategy of CRC.

In conclusion, these previous findings aforementioned suggested that knocking down AS-lncRNAs can regulate tumor development by inhibiting the expression of oncogenes, which may become a novel method for tumor therapy.

Activation of antitumor immunity

Possibly the biggest natural antagonist of cancer development is the host immune system, especially T cells, which can initiate an antitumor response by expressing specific receptors of tumor antigens (121). To prevent immune hyperactivation, effector lymphocytes express immune checkpoints, which are mainly receptor proteins expressed on the cell surface (122). Several tumors, including liver cancer, exploited this by expressing corresponding ligands to these checkpoints (123). After ligand binding, the activity of effector lymphocytes becomes impaired. Programmed cell death protein 1 (PD-1) was one of the main such co-inhibitory receptors (124), whereas programmed death ligand (PD-L)1 is one of the ligands for PD-1 (125). As a co-suppressor molecule during cancer development, PD-L1 is expressed on T lymphocytes and promotes immune escape (126). Fan et al (127) previously found that PC-esterase domain-containing 1B (PCED1B)-AS1 enhanced the expression and function of PD-L1 and PD-L2 by sponging miR-194-5p, which induced immunosuppression by HCC. Therefore, PCED1B-AS1 was a potential therapeutic target for HCC. In cancer, the PD-1/PD-L1 pathway is responsible for T cell activation, proliferation and cytotoxic secretion, leading to the degeneration of the antitumor immune response (128). Zhou et al (129) reported that in hypopharyngeal squamous cell carcinoma there was a positive regulatory relationship between HOXA11-AS1 and PD-L1. In particular, downregulation of HOXA11-AS1 inhibited PD-L1-mediated immune escape and metastasis by reducing the association between polypyrimidine tract binding protein 1 and FOS-like 1 in hypopharyngeal squamous cell carcinoma. Results from the aforementioned study provided a preliminary basis for applying HOXA11-AS1 as a potential target for the immunotherapy of hypopharyngeal squamous cell carcinoma. In addition, AS-lncRNAs can regulate the expression of genes associated with immune responses, changing the status of immune cell infiltration in the tumor microenvironment and profoundly dictate tumor invasiveness, progression and prognosis (88,130). Nie et al (85) performed an integrative analysis using publicly available gene expression datasets from TCGA project and the Genotype-Tissue Expression project from various GC patients' cohorts and a novel pairing algorithm to identify and validate 13 immune-related lncRNA pair signatures. This may provide novel insights into the role of AS-lncRNAs in tumor immunity and to facilitate the development of antitumor immunotherapeutic methods.

In conclusion, AS-lncRNAs can regulate gene expression related to immune response, thereby regulating antitumor immunity to regulate tumor development, and it may become a novel target for tumor therapy.

To sum up, tumor gene therapy is currently a topic of intense research, with main fields including the activation of tumor suppressor factors, inhibiting oncogene expression and inducing antitumor immune activation, which can all be regulated by AS-lncRNAs according to the aforementioned previous findings.

Enhancing chemoradiotherapy and chemotherapy by interfering with tumor drug resistance

Radiotherapy and chemotherapy form the core of all treatment strategies for cancer; however, the majority of patients will develop drug resistance, which is a major obstacle to the long-term efficacy of radiotherapy and chemotherapy (131,132). The mechanisms underlying drug resistance are complex, which can involve a wide array of factors, such as drug efflux, DNA damage repair, apoptosis and target mutations (133,134). Since AS-lncRNAs have been reported to be aberrantly expressed in tumors, they regulate chemoradiotherapy resistance (135–138).

5-Fluorouracil resistance

In a previous study, Zhou et al (135) found the lncRNA transmembrane protein 44 (TMEM44)-AS1 to be associated with resistance to 5-FU by GC cells. TMEM44-AS1 was observed to function as a ceRNA, which upregulated protein phosphatase 1 regulatory subunit 13-like expression and inhibited the p53 pathway, finally promoting resistance to 5-FU. To combat this, Zhou et al (129) developed the nano-carrier chitosan-gelatin epigallocatechin gallate to deliver small interfering RNA-TMEM44-AS1 to knock down TMEM44-AS1 expression, which effectively reversed 5-FU resistance, specifically by observing a significant enhancement in the therapeutic effects of 5-FU in xenograft mouse models of GC. Gui et al (137) recently observed that FEZ family zinc finger (FEZF1)-AS1 was upregulated in 5-FU chemically resistant GC tissues. RIP then revealed that FEZF1-AS1 increased the chemical resistance of GC cells through directly targeting autophagy-related 5 ganglion autophagy. By contrast, knocking down FEZF1-AS1 expression was documented to increase the sensitivity to 5-FU in GC cells in vivo. In a recent study, Qu et al (136) revealed that lncRNA discs large homolog associated protein 1 (DLGAP1)-AS1 promoted the progression of CRC. Subsequently, functional assays revealed that silencing DLGAP1-AS1 expression enhanced the chemosensitivity of cells to 5-FU, suggesting that silencing DLGAP1-AS1 expression may be a promising therapeutic target for CRC.

Cisplatin resistance

Ren et al (65) found that lncRNA ADAMTS9-AS2 overexpression inhibited the progression of GC and promoted cisplatin chemosensitivity by regulating the pyroptosis process mediated by the miR-223-3p/NLRP3 axis. This molecular mechanism underlying the drug resistance in GC to cisplatin provided a novel therapeutic target for the clinical treatment of GC. Song et al (139) previously reported that OIP5-AS1 was highly expressed in cisplatin-resistant (CR) osteosarcoma cells. Silencing OIP5-AS1 expression promoted the apoptosis in CR osteosarcoma cells, suggesting that the downregulation of OIP5-AS1 can significantly reverse cisplatin resistance in osteosarcoma cells. These results deepened the understanding into the role of OIP5-AS1 in cisplatin resistance in osteosarcoma, possibly providing a novel therapeutic target for patients with CR osteosarcoma. Wu et al (140) previously revealed that upregulation of FOXD1-AS1 increased the resistance of GC cells to cisplatin. FOXD1-AS1 promoted FOXD1 translocation through PIK3CA/PI3K/AKT/mTOR signaling, which increased the progression of GC and chemotherapy resistance. These findings provided a novel target for the treatment of patients with CR GC.

Oxaliplatin resistance

Recent studies have demonstrated that lncRNAs are involved in the regulation of oxaliplatin resistance (141–143). Liang et al (144) previously found that OIP5-AS1 expression was upregulated in CRC tissues, whilst that of miR-137 was downregulated. Therefore, there was a negative association between the expression of OIP5-AS1 and miR-137. OIP5-AS1 was subsequently found to directly target miR-137, such that silencing OIP5-AS1 expression reversed the resistance of CRC cells to oxaliplatin by promoting the expression of miR-137. Furthermore, Hui et al (145) reported that overexpression of PGM5-AS1 inhibited the proliferation of oxaliplatin-resistant colon cancer cells by using colony formation assays, Cell Counting Kit-8 analysis and EdU assays. Transwell analysis revealed that compared with those in the control group, the overexpression of PGM5-AS1 inhibited the invasion and migration of oxaliplatin-resistant colon cancer cells. These results suggested that PGM5-AS1 overexpression can render colon cancer cells more sensitive to oxaliplatin. Hui et al (136) also found that PGM5-AS1 and oxaliplatin could enter colon cancer cells through engineered exosomes to effectively reverse drug resistance. Li et al (143) previously observed that extracellular leucine rich repeat and fibronectin type III domain-containing 1 (ELFN1)-AS1 interacted with the promoter region of meis homeobox 1 (MEIS1) to inhibit the expression of MEIS1. By contrast, MEIS1 can enhance the sensitivity of CRC cells to oxaliplatin. This resulted in the conclusion that the combination of oxaliplatin and ELFN1-AS1 ASO can reverse oxaliplatin resistance in CRC, highlighting the potential of targeting ELFN1-AS1 as a treatment method oxaliplatin resistance.

Radiotherapy resistance

Zou et al (138) previously found that lncRNA OIP5-AS1 and dual specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) were downregulated in radiation-resistant CRC cell lines. Specially, overexpression of OIP5-AS1 can regulate DYRK1A expression through miR-369-3p, damage the survival of cell clones and promote apoptosis after radiotherapy, suggestive of the reversal of radiation resistance in CRC cells.

Conclusion

Malignant tumors are one of the main causes of mortality worldwide. As the second leading cause of mortality in China, cancer seriously threatens the lives of the population and restricts social and economic development. lncRNAs are RNAs that contain >200 nucleotides. Although the majority of lncRNAs do not encode proteins, they can mediate a variety of important physiological functions. AS-lncRNAs are part of the antisense chains of coding genes that are transcribed from the opposite strands of protein or non-protein coding genes. Accumulating evidence has shown that AS-lncRNAs can serve an important role in tumorigenesis, metastasis, prognosis and drug resistance. AS-lncRNAs can regulate gene expression in tumors through endogenous competition mechanisms, promoter interactions, direct interactions with mRNA, acting as ‘scaffolds’ to regulate the half-life of mRNA, interactions with 5′-UTR, regulation of mRNAs of interest, positive feedback loop of ‘AS-lncRNA/mediator protein/gene of interest’ model and transcriptional regulation in the nucleus. Over the past decade, AS-lncRNAs have been found to mediate a variety of molecular functions, such as the regulation of cell proliferation, invasion, migration and apoptosis. In addition, they have been shown to regulate tumor energy metabolism, inflammation and tumor neovascularization. Although the discovery of AS-lncRNAs has only been relatively recent, their possible application in tumor treatment has been widely studied. AS-lncRNAs can potentially serve as markers of tumor therapy, as they may activate tumor suppressor factors, inhibit oncogene expression and induce antitumor immune activation. In addition, there is evidence that AS-lncRNAs can regulate the mechanism underlying tumor treatment and drug resistance. To conclude, AS-lncRNAs are likely to serve a role in tumor gene expression, the mechanisms regulating tumors physiology and tumor response to treatment. Treatment of abnormally expressed AS-lncRNAs in tumors is a promising method. The study into AS-lncRNAs has opened up a new era for the exploration, diagnosis and treatment of tumors.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 82174379), the Special Project of Jiangsu Provincial Traditional Chinese Medicine Hospital Innovation and Development Fund (grant no. Y2020CX38) and the National Clinical Research Base of Traditional Chinese Medicine in Jiangsu (grant no. JD2023SZ04).

Availability of data and materials

Not applicable.

Authors' contributions

YS wrote the manuscript, searched the literature and prepared the table. YD, YL and YC was involved in the design of the study, and revised the manuscript. YL prepared the figure. JZ, ZL, CC and XY provided article ideas, modified the tables and revised the manuscript. QL, SZ, WT, ZC, YW, LH, ZQ, KW and ZM performed literature research and collected relevant articles. 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 

GBD 2019 Diseases and Injuries Collaborators, . Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet. 396:1204–1222. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Xia C, Dong X, Li H, Cao M, Sun D, He S, Yang F, Yan X, Zhang S, Li N and Chen W: Cancer statistics in China and United States, 2022: Profiles, trends, and determinants. Chin Med J (Engl). 135:584–590. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Qiu H, Cao S and Xu R: Cancer incidence, mortality, and burden in China: A time-trend analysis and comparison with the United States and United Kingdom based on the global epidemiological data released in 2020. Cancer Commun (Lond). 41:1037–1048. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Miller KD, Nogueira L, Devasia T, Mariotto AB, Yabroff KR, Jemal A, Kramer J and Siegel RL: Cancer treatment and survivorship statistics, 2022. CA Cancer J Clin. 72:409–436. 2022. View Article : Google Scholar : PubMed/NCBI

6 

Ponting CP, Oliver PL and Reik W: Evolution and functions of long noncoding RNAs. Cell. 136:629–641. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Agirre E and Eyras E: Databases and resources for human small non-coding RNAs. Hum Genomics. 5:192–199. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Lee JT: Epigenetic regulation by long noncoding RNAs. Science. 338:1435–1439. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Takahashi A, Tsutsumi R, Kikuchi I, Obuse C, Saito Y, Seidi A, Karisch R, Fernandez M, Cho T, Ohnishi N, et al: SHP2 tyrosine phosphatase converts parafibromin/Cdc73 from a tumor suppressor to an oncogenic driver. Mol Cell. 43:45–56. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and cancer: A new paradigm. Cancer Res. 77:3965–3981. 2017. View Article : Google Scholar : PubMed/NCBI

11 

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

12 

Hua Q, Mi B, Xu F, Wen J, Zhao L, Liu J and Huang G: Hypoxia-induced lncRNA-AC020978 promotes proliferation and glycolytic metabolism of non-small cell lung cancer by regulating PKM2/HIF-1α axis. Theranostics. 10:4762–4778. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Chen J, Yu Y, Li H, Hu Q, Chen X, He Y, Xue C, Ren F, Ren Z, Li J, et al: Long non-coding RNA PVT1 promotes tumor progression by regulating the miR-143/HK2 axis in gallbladder cancer. Mol Cancer. 18:332019. View Article : Google Scholar : PubMed/NCBI

14 

Devaux Y, Zangrando J, Schroen B, Creemers EE, Pedrazzini T, Chang CP, Dorn GN, Thum T II and Heymans S; Cardiolinc network, : Long noncoding RNAs in cardiac development and ageing. Nat Rev Cardiol. 12:415–425. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Magistri M, Faghihi MA, St Laurent G III and Wahlestedt C: Regulation of chromatin structure by long noncoding RNAs: Focus on natural antisense transcripts. Trends Genet. 28:389–396. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Cui XY, Zhan JK and Liu YS: Roles and functions of antisense lncRNA in vascular aging. Ageing Res Rev. 72:1014802021. View Article : Google Scholar : PubMed/NCBI

17 

Bian Z, Zhang J, Li M, Feng Y, Wang X, Zhang J, Yao S, Jin G, Du J, Han W, et al: LncRNA-FEZF1-AS1 promotes tumor proliferation and metastasis in colorectal cancer by regulating PKM2 Signaling. Clin Cancer Res. 24:4808–4819. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Yang MH, Zhao L, Wang L, Ou-Yang W, Hu SS, Li WL, Ai ML, Wang YQ, Han Y, Li TT, et al: Nuclear lncRNA HOXD-AS1 suppresses colorectal carcinoma growth and metastasis via inhibiting HOXD3-induced integrin β3 transcriptional activating and MAPK/AKT signalling. Mol Cancer. 18:312019. View Article : Google Scholar : PubMed/NCBI

19 

Braga EA, Fridman MV, Moscovtsev AA, Filippova EA, Dmitriev AA and Kushlinskii NE: LncRNAs in ovarian cancer progression, metastasis, and main pathways: ceRNA and alternative mechanisms. Int J Mol Sci. 21:88552020. View Article : Google Scholar : PubMed/NCBI

20 

Pan H, Ding Y, Jiang Y, Wang X, Rao J, Zhang X, Yu H, Hou Q and Li T: LncRNA LIFR-AS1 promotes proliferation and invasion of gastric cancer cell via miR-29a-3p/COL1A2 axis. Cancer Cell Int. 21:72021. View Article : Google Scholar : PubMed/NCBI

21 

Si Z, Yu L, Jing H, Wu L and Wang X: Oncogenic lncRNA ZNF561-AS1 is essential for colorectal cancer proliferation and survival through regulation of miR-26a-3p/miR-128-5p-SRSF6 axis. J Exp Clin Canc Res. 40:782021. View Article : Google Scholar

22 

Shuai Y, Ma Z, Liu W, Yu T, Yan C, Jiang H, Tian S, Xu T and Shu Y: TEAD4 modulated LncRNA MNX1-AS1 contributes to gastric cancer progression partly through suppressing BTG2 and activating BCL2. Mol Cancer. 19:62020. View Article : Google Scholar : PubMed/NCBI

23 

Yao W, Du X, Zhang J, Wang Y, Wang M, Pan Z and Li Q: SMAD4-induced knockdown of the antisense long noncoding RNA BRE-AS contributes to granulosa cell apoptosis. Mol Ther Nucleic Acids. 25:251–263. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Jiang B, Yuan Y, Yi T and Dang W: The roles of antisense long noncoding RNAs in tumorigenesis and development through Cis-Regulation of neighbouring genes. Biomolecules. 13:6842023. View Article : Google Scholar : PubMed/NCBI

25 

Liu B, Xiang W and Liu J, Tang J, Wang J, Liu B, Long Z, Wang L, Yin G and Liu J: The regulatory role of antisense lncRNAs in cancer. Cancer Cell Int. 21:4592021. View Article : Google Scholar : PubMed/NCBI

26 

Li D, Shen L, Zhang X, Chen Z, Huang P, Huang C and Qin S: LncRNA ELF3-AS1 inhibits gastric cancer by forming a negative feedback loop with SNAI2 and regulates ELF3 mRNA stability via interacting with ILF2/ILF3 complex. J Exp Clin Cancer Res. 41:3322022. View Article : Google Scholar : PubMed/NCBI

27 

Bartl J, Zanini M, Bernardi F, Forget A, Blumel L, Talbot J, Picard D, Qin N, Cancila G, Gao Q, et al: The HHIP-AS1 lncRNA promotes tumorigenicity through stabilization of dynein complex 1 in human SHH-driven tumors. Nat Commun. 13:40612022. View Article : Google Scholar : PubMed/NCBI

28 

Jadaliha M, Gholamalamdari O, Tang W, Zhang Y, Petracovici A, Hao Q, Tariq A, Kim TG, Holton SE, Singh DK, et al: A natural antisense lncRNA controls breast cancer progression by promoting tumor suppressor gene mRNA stability. PLoS Genet. 14:e10078022018. View Article : Google Scholar : PubMed/NCBI

29 

Pan K and Xie Y: LncRNA FOXC2-AS1 enhances FOXC2 mRNA stability to promote colorectal cancer progression via activation of Ca(2+)-FAK signal pathway. Cell Death Dis. 11:4342020. View Article : Google Scholar : PubMed/NCBI

30 

Zhao Y, Liu Y, Lin L, Huang Q, He W, Zhang S, Dong S, Wen Z, Rao J, Liao W and Shi M: The lncRNA MACC1-AS1 promotes gastric cancer cell metabolic plasticity via AMPK/Lin28 mediated mRNA stability of MACC1. Mol Cancer. 17:692018. View Article : Google Scholar : PubMed/NCBI

31 

Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E and Chang HY: Long noncoding RNA as modular scaffold of histone modification complexes. Science. 329:689–693. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Zhu L, Zhu Y, Han S, Chen M, Song P, Dai D, Xu W, Jiang T, Feng L, Shin VY, et al: Impaired autophagic degradation of lncRNA ARHGAP5-AS1 promotes chemoresistance in gastric cancer. Cell Death Dis. 10:3832019. View Article : Google Scholar : PubMed/NCBI

33 

Visvanathan A, Patil V, Arora A, Hegde AS, Arivazhagan A, Santosh V and Somasundaram K: Essential role of METTL3-mediated m(6)A modification in glioma stem-like cells maintenance and radioresistance. Oncogene. 37:522–533. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Tran NT, Su H, Khodadadi Jamayran A, Lin S, Zhang L, Zhou D, Pawlik KM, Townes TM, Chen Y, Mulloy JC and Zhao X: The AS-RBM15 lncRNA enhances RBM15 protein translation during megakaryocyte differentiation. EMBO Rep. 17:887–900. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Zhang J and Manley JL: Misregulation of pre-mRNA alternative splicing in cancer. Cancer Discov. 3:1228–1237. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Oltean S and Bates DO: Hallmarks of alternative splicing in cancer. Oncogene. 33:5311–5318. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Yuan JH, Liu XN, Wang TT, Pan W, Tao QF, Zhou WP, Wang F and Sun SH: The MBNL3 splicing factor promotes hepatocellular carcinoma by increasing PXN expression through the alternative splicing of lncRNA-PXN-AS1. Nat Cell Biol. 19:820–832. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Zhou M, Guo X, Wang M and Qin R: The patterns of antisense long non-coding RNAs regulating corresponding sense genes in human cancers. J Cancer. 12:1499–1506. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Luo N, Zhang K, Li X and Hu Y: ZEB1 induced-upregulation of long noncoding RNA ZEB1-AS1 facilitates the progression of triple negative breast cancer by binding with ELAVL1 to maintain the stability of ZEB1 mRNA. J Cell Biochem. 121:4176–4187. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Wang A, Bao Y, Wu Z, Zhao T, Wang D, Shi J, Liu B, Sun S, Yang F, Wang L and Qu L: Long noncoding RNA EGFR-AS1 promotes cell growth and metastasis via affecting HuR mediated mRNA stability of EGFR in renal cancer. Cell Death Dis. 10:1542019. View Article : Google Scholar : PubMed/NCBI

41 

Gong YQ, Lu TL, Hou FT and Chen CW: Antisense long non-coding RNAs in gastric cancer. Clin Chim Acta. 534:128–137. 2022. View Article : Google Scholar : PubMed/NCBI

42 

Su J, Chen D, Ruan Y, Tian Y, Lv K, Zhou X, Ying D and Lu Y: LncRNA MBNL1-AS1 represses gastric cancer progression via the TGF-β pathway by modulating miR-424-5p/Smad7 axis. Bioengineered. 13:6978–6995. 2022. View Article : Google Scholar : PubMed/NCBI

43 

Zhuang L, Ding W, Ding W, Zhang Q, Xu X and Xi D: lncRNA ZNF667-AS1 (NR_036521.1) inhibits the progression of colorectal cancer via regulating ANK2/JAK2 expression. J Cell Physiol. 236:2178–2193. 2021. View Article : Google Scholar : PubMed/NCBI

44 

Sun M, Nie F, Wang Y, Zhang Z, Hou J, He D, Xie M, Xu L, De W, Wang Z and Wang J: LncRNA HOXA11-AS promotes proliferation and invasion of gastric cancer by scaffolding the chromatin modification factors PRC2, LSD1, and DNMT1. Cancer Res. 76:6299–6310. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Lin X, Zhuang S, Chen X, Du J, Zhong L, Ding J, Wang L, Yi J, Hu G, Tang G, et al: lncRNA ITGB8-AS1 functions as a ceRNA to promote colorectal cancer growth and migration through integrin-mediated focal adhesion signaling. Mol Ther. 30:688–702. 2022. View Article : Google Scholar : PubMed/NCBI

46 

Bedoui S, Herold MJ and Strasser A: Emerging connectivity of programmed cell death pathways and its physiological implications. Nat Rev Mol Cell Bio. 21:678–695. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Liu J, Hong M, Li Y, Chen D, Wu Y and Hu Y: Programmed cell death tunes tumor immunity. Front Immunol. 13:8473452022. View Article : Google Scholar : PubMed/NCBI

48 

Zheng C, Chu M, Chen Q, Chen C, Wang ZW and Chen X: The role of lncRNA OIP5-AS1 in cancer development and progression. Apoptosis. 27:311–321. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Wang Z and Jin J: LncRNA SLCO4A1-AS1 promotes colorectal cancer cell proliferation by enhancing autophagy via miR-508-3p/PARD3 axis. Aging (Albany NY). 11:4876–4889. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Xia X, Wang X, Cheng Z, Qin W, Lei L, Jiang J and Hu J: The role of pyroptosis in cancer: Pro-cancer or pro-‘host’? Cell Death Dis. 10:6502019. View Article : Google Scholar : PubMed/NCBI

51 

Huang G, Xiang Z, Wu H, He Q, Dou R, Lin Z, Yang C, Huang S, Song J, Di Z, et al: The lncRNA BDNF-AS/WDR5/FBXW7 axis mediates ferroptosis in gastric cancer peritoneal metastasis by regulating VDAC3 ubiquitination. Int J Biol Sci. 18:1415–1433. 2022. View Article : Google Scholar : PubMed/NCBI

52 

Bai Y and Li S: Long noncoding RNA OIP5-AS1 aggravates cell proliferation, migration in gastric cancer by epigenetically silencing NLRP6 expression via binding EZH2. J Cell Biochem. 121:353–362. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Wang LW, Li XB, Liu Z, Zhao LH, Wang Y and Yue L: Long non-coding RNA OIP5-AS1 promotes proliferation of gastric cancer cells by targeting miR-641. Eur Rev Med Pharmacol Sci. 23:10776–10784. 2019.PubMed/NCBI

54 

Tao Y, Wan X, Fan Q, Wang Y, Sun H, Ma L, Sun C and Wu Y: Long non-coding RNA OIP5-AS1 promotes the growth of gastric cancer through the miR-367-3p/HMGA2 axis. Dig Liver Dis. 52:773–779. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Liao Y, Zhang B, Zhang T, Zhang Y and Wang F: LncRNA GATA6-AS promotes cancer cell proliferation and inhibits apoptosis in glioma by downregulating lncRNA TUG1. Cancer Biother Radiopharm. 34:660–665. 2019.PubMed/NCBI

56 

Russell RC and Guan KL: The multifaceted role of autophagy in cancer. EMBO J. 41:e1100312022. View Article : Google Scholar : PubMed/NCBI

57 

Li X, He S and Ma B: Autophagy and autophagy-related proteins in cancer. Mol cancer. 19:122020. View Article : Google Scholar : PubMed/NCBI

58 

Li J, Zhan H, Ren Y, Feng M, Wang Q, Jiao Q, Wang Y, Liu X, Zhang S, Du L, et al: Sirtuin 4 activates autophagy and inhibits tumorigenesis by upregulating the p53 signaling pathway. Cell Death Differ. 30:313–326. 2023. View Article : Google Scholar : PubMed/NCBI

59 

Feng X, Zhang H, Meng L, Song H, Zhou Q, Qu C, Zhao P, Li Q, Zou C, Liu X and Zhang Z: Hypoxia-induced acetylation of PAK1 enhances autophagy and promotes brain tumorigenesis via phosphorylating ATG5. Autophagy. 17:723–742. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Elias EE, Lyons B and Muruve DA: Gasdermins and pyroptosis in the kidney. Nat Rev Nephrol. 19:337–350. 2023. View Article : Google Scholar : PubMed/NCBI

61 

Wei X, Xie F, Zhou X, Wu Y, Yan H, Liu T, Huang J, Wang F, Zhou F and Zhang L: Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 19:971–992. 2022. View Article : Google Scholar : PubMed/NCBI

62 

Tang R, Xu J, Zhang B, Liu J, Liang C, Hua J, Meng Q, Yu X and Shi S: Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J Hematol Oncol. 13:1102020. View Article : Google Scholar : PubMed/NCBI

63 

Yang P, Yang W, Wei Z, Li Y, Yang Y and Wang J: Novel targets for gastric cancer: The tumor microenvironment (TME), N6-methyladenosine (m6A), pyroptosis, autophagy, ferroptosis and cuproptosis. Biomed Pharmacother. 163:1148832023. View Article : Google Scholar : PubMed/NCBI

64 

Wang Z, Cao L, Zhou S, Lyu J, Gao Y and Yang R: Construction and validation of a novel pyroptosis-related Four-lncRNA prognostic signature related to gastric cancer and immune infiltration. Front Immunol. 13:8547852022. View Article : Google Scholar : PubMed/NCBI

65 

Ren N, Jiang T, Wang C, Xie S, Xing Y, Piao D, Zhang T and Zhu Y: LncRNA ADAMTS9-AS2 inhibits gastric cancer (GC) development and sensitizes chemoresistant GC cells to cisplatin by regulating miR-223-3p/NLRP3 axis. Aging (Albany NY). 12:11025–11041. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Wu Y, Zhang S, Gong X, Tam S, Xiao D, Liu S and Tao Y: The epigenetic regulators and metabolic changes in ferroptosis-associated cancer progression. Mol Cancer. 19:392020. View Article : Google Scholar : PubMed/NCBI

68 

Katada S, Imhof A and Sassone-Corsi P: Connecting threads: Epigenetics and metabolism. Cell. 148:24–28. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Ganapathy-Kanniappan S and Geschwind JF: Tumor glycolysis as a target for cancer therapy: Progress and prospects. Mol Cancer. 12:1522013. View Article : Google Scholar : PubMed/NCBI

70 

Vaupel P, Schmidberger H and Mayer A: The Warburg effect: Essential part of metabolic reprogramming and central contributor to cancer progression. Int J Radiat Biol. 95:912–919. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Koppenol WH, Bounds PL and Dang CV: Otto Warburg's contributions to current concepts of cancer metabolism. Nat Rev Cancer. 11:325–337. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Liberti MV and Locasale JW: The warburg effect: How does it benefit cancer cells? Trends Biochem Sci. 41:211–218. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Li C, Wang P, Du J, Chen J, Liu W and Ye K: LncRNA RAD51-AS1/miR-29b/c-3p/NDRG2 crosstalk repressed proliferation, invasion and glycolysis of colorectal cancer. IUBMB life. 73:286–298. 2021. View Article : Google Scholar : PubMed/NCBI

74 

Cheng X, Li J and Guo D: SCAP/SREBPs are central players in lipid metabolism and novel metabolic targets in cancer therapy. Curr Top Med Chem. 18:484–493. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Li C, Zhang L, Qiu Z, Deng W and Wang W: Key molecules of fatty acid metabolism in gastric cancer. Biomolecules. 12:7062022. View Article : Google Scholar : PubMed/NCBI

76 

Gong J, Lin Y, Zhang H, Liu C, Cheng Z, Yang X, Zhang J, Xiao Y, Sang N, Qian X, et al: Reprogramming of lipid metabolism in cancer-associated fibroblasts potentiates migration of colorectal cancer cells. Cell Death Dis. 11:2672020. View Article : Google Scholar : PubMed/NCBI

77 

Guo D, Reinitz F, Youssef M, Hong C, Nathanson D, Akhavan D, Kuga D, Amzajerdi AN, Soto H, Zhu S, et al: An LXR agonist promotes glioblastoma cell death through inhibition of an EGFR/AKT/SREBP-1/LDLR-dependent pathway. Cancer Discov. 1:442–456. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Zhang J, Song F, Zhao X, Jiang H, Wu X, Wang B, Zhou M, Tian M, Shi B, Wang H, et al: EGFR modulates monounsaturated fatty acid synthesis through phosphorylation of SCD1 in lung cancer. Mol Cancer. 16:1272017. View Article : Google Scholar : PubMed/NCBI

79 

Tang Y, Tang R, Tang M, Huang P, Liao Z, Zhou J, Zhou L, Su M, Chen P, Jiang J, et al: LncRNA DNAJC3-AS1 Regulates fatty acid synthase via the EGFR pathway to promote the progression of colorectal cancer. Front Oncol. 10:6045342020. View Article : Google Scholar : PubMed/NCBI

80 

Currie E, Schulze A, Zechner R, Walther TC and Farese RJ: Cellular fatty acid metabolism and cancer. Cell Metab. 18:153–161. 2013. View Article : Google Scholar : PubMed/NCBI

81 

Carracedo A, Cantley LC and Pandolfi PP: Cancer metabolism: Fatty acid oxidation in the limelight. Nat Rev Cancer. 13:227–232. 2013. View Article : Google Scholar : PubMed/NCBI

82 

He W, Liang B, Wang C, Li S, Zhao Y, Huang Q, Liu Z, Yao Z, Wu Q, Liao W, et al: MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer. Oncogene. 38:4637–4654. 2019. View Article : Google Scholar : PubMed/NCBI

83 

Greten FR and Grivennikov SI: Inflammation and cancer: Triggers, mechanisms, and consequences. Immunity. 51:27–41. 2019. View Article : Google Scholar : PubMed/NCBI

84 

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

85 

Nie C, Zhai J, Wang Q, Zhu X, Xiang G, Liu C, Liu T, Wang W, Wang Y, Zhao Y, et al: Comprehensive analysis of an individualized Immune-Related lncRNA pair signature in gastric cancer. Front Cell Dev Biol. 10:8056232022. View Article : Google Scholar : PubMed/NCBI

86 

Cui X, Morales RT, Qian W, Wang H, Gagner JP, Dolgalev I, Placantonakis D, Zagzag D, Cimmino L, Snuderl M, et al: Hacking macrophage-associated immunosuppression for regulating glioblastoma angiogenesis. Biomaterials. 161:164–178. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Wang H, Tian T and Zhang J: Tumor-Associated Macrophages (TAMs) in Colorectal Cancer (CRC): From mechanism to therapy and prognosis. Int J Mol Sci. 22:84702021. View Article : Google Scholar : PubMed/NCBI

88 

Lazăr DC, Avram MF, Romoșan I, Cornianu M, Tăban S and Goldiș A: Prognostic significance of tumor immune microenvironment and immunotherapy: Novel insights and future perspectives in gastric cancer. World J Gastroenterol. 24:3583–3616. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Denaro N, Merlano MC and Lo Nigro C: Long noncoding RNAs as regulators of cancer immunity. Mol Oncol. 13:61–73. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Xu M, Xu X, Pan B, Chen X, Lin K, Zeng K, Liu X, Xu T, Sun L, Qin J, et al: LncRNA SATB2-AS1 inhibits tumor metastasis and affects the tumor immune cell microenvironment in colorectal cancer by regulating SATB2. Mol Cancer. 18:1352019. View Article : Google Scholar : PubMed/NCBI

91 

Sanchez-Tillo E, Liu Y, de Barrios O, Siles L, Fanlo L, Cuatrecasas M, Darling DS, Dean DC, Castells A and Postigo A: EMT-activating transcription factors in cancer: Beyond EMT and tumor invasiveness. Cell Mol Life Sci. 69:3429–3456. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Nieto MA, Huang RY, Jackson RA and Thiery JP: EMT: 2016. Cell. 166:21–45. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Dongre A and Weinberg RA: New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Bio. 20:69–84. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Song Y, Lin M, Liu Y, Wang ZW and Zhu X: Emerging role of F-box proteins in the regulation of epithelial-mesenchymal transition and stem cells in human cancers. Stem Cell Res Ther. 10:1242019. View Article : Google Scholar : PubMed/NCBI

95 

Brabletz S, Schuhwerk H, Brabletz T and Stemmler MP: Dynamic EMT: A multi-tool for tumor progression. EMBO J. 40:e1086472021. View Article : Google Scholar : PubMed/NCBI

96 

Lin J, Shi Z, Yu Z and He Z: LncRNA HIF1A-AS2 positively affects the progression and EMT formation of colorectal cancer through regulating miR-129-5p and DNMT3A. Biomed Pharmacother. 98:433–439. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Yu J, Han Z, Sun Z, Wang Y, Zheng M and Song C: LncRNA SLCO4A1-AS1 facilitates growth and metastasis of colorectal cancer through β-catenin-dependent Wnt pathway. J Exp Clin Canc Res. 37:2222018. View Article : Google Scholar

98 

Zhou L, Jiang J, Huang Z, Jin P, Peng L, Luo M, Zhang Z, Chen Y, Xie N, Gao W, et al: Hypoxia-induced lncRNA STEAP3-AS1 activates Wnt/β-catenin signaling to promote colorectal cancer progression by preventing m(6)A-mediated degradation of STEAP3 mRNA. Mol Cancer. 21:1682022. View Article : Google Scholar : PubMed/NCBI

99 

Ali MM, Akhade VS, Kosalai ST, Subhash S, Statello L, Meryet-Figuiere M, Abrahamsson J, Mondal T and Kanduri C: PAN-cancer analysis of S-phase enriched lncRNAs identifies oncogenic drivers and biomarkers. Nat Commun. 9:8832018. View Article : Google Scholar : PubMed/NCBI

100 

Mo S, Zhang L, Dai W, Han L, Wang R, Xiang W, Wang Z, Li Q, Yu J, Yuan J, et al: Antisense lncRNA LDLRAD4-AS1 promotes metastasis by decreasing the expression of LDLRAD4 and predicts a poor prognosis in colorectal cancer. Cell Death Dis. 11:1552020. View Article : Google Scholar : PubMed/NCBI

101 

Gonzalez DM and Medici D: Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal. 7:re82014. View Article : Google Scholar : PubMed/NCBI

102 

Slattery ML, Herrick J, Curtin K, Samowitz W, Wolff RK, Caan BJ, Duggan D, Potter JD and Peters U: Increased risk of colon cancer associated with a genetic polymorphism of SMAD7. Cancer Res. 70:1479–1485. 2010. View Article : Google Scholar : PubMed/NCBI

103 

Rakic JM, Maillard C, Jost M, Bajou K, Masson V, Devy L, Lambert V, Foidart JM and Noel A: Role of plasminogen activator-plasmin system in tumor angiogenesis. Cell Mol Life Sci. 60:463–473. 2003. View Article : Google Scholar : PubMed/NCBI

104 

Bajou K, Maillard C, Jost M, Lijnen RH, Gils A, Declerck P, Carmeliet P, Foidart JM and Noel A: Host-derived plasminogen activator inhibitor-1 (PAI-1) concentration is critical for in vivo tumoral angiogenesis and growth. Oncogene. 23:6986–6990. 2004. View Article : Google Scholar : PubMed/NCBI

105 

Teng F, Zhang JX, Chen Y, Shen XD, Su C, Guo YJ, Wang PH, Shi CC, Lei M, Cao YO and Liu SQ: LncRNA NKX2-1-AS1 promotes tumor progression and angiogenesis via upregulation of SERPINE1 expression and activation of the VEGFR-2 signaling pathway in gastric cancer. Mol Oncol. 15:1234–1255. 2021. View Article : Google Scholar : PubMed/NCBI

106 

Duan S, Huang W, Liu X, Liu X, Chen N, Xu Q, Hu Y, Song W and Zhou J: IMPDH2 promotes colorectal cancer progression through activation of the PI3K/AKT/mTOR and PI3K/AKT/FOXO1 signaling pathways. J Exp Clin Canc Res. 37:3042018. View Article : Google Scholar

107 

Peng Y, Wang Y, Zhou C, Mei W and Zeng C: PI3K/Akt/mTOR pathway and its role in cancer therapeutics: Are we making headway? Front Oncol. 12:8191282022. View Article : Google Scholar : PubMed/NCBI

108 

Liu HT, Ma RR, Lv BB, Zhang H, Shi DB, Guo XY, Zhang GH and Gao P: LncRNA-HNF1A-AS1 functions as a competing endogenous RNA to activate PI3K/AKT signalling pathway by sponging miR-30b-3p in gastric cancer. Br J Cancer. 122:1825–1836. 2020. View Article : Google Scholar : PubMed/NCBI

109 

Guo C, Zhou S, Yi W, Yang P, Li O, Liu J and Peng C: Long non-coding RNA muskelin 1 antisense RNA (MKLN1-AS) is a potential diagnostic and prognostic biomarker and therapeutic target for hepatocellular carcinoma. Exp Mol Pathol. 120:1046382021. View Article : Google Scholar : PubMed/NCBI

110 

El-Ashmawy NE, Al-Ashmawy GM and Hamouda SM: Long non-coding RNA FAM83H-AS1 as an emerging marker for diagnosis, prognosis and therapeutic targeting of cancer. Cell Biochem Funct. 39:350–356. 2021. View Article : Google Scholar : PubMed/NCBI

111 

Da J, Liu P, Wang R and Bu L: Upregulation of the long non-coding RNA FAM83H-AS1 in gastric cancer and its clinical significance. Pathol Res Pract. 215:1526162019. View Article : Google Scholar : PubMed/NCBI

112 

Chen Q, Zhou L, Ma D, Hou J, Lin Y, Wu J and Tao M: LncRNA GAS6-AS1 facilitates tumorigenesis and metastasis of colorectal cancer by regulating TRIM14 through miR-370-3p/miR-1296-5p and FUS. J Transl Med. 20:3562022. View Article : Google Scholar : PubMed/NCBI

113 

Lu GH, Zhao HM, Liu ZY, Cao Q, Shao RD and Sun G: LncRNA SAMD12-AS1 promotes the progression of gastric cancer via DNMT1/p53 Axis. Arch Med Res. 52:683–691. 2021. View Article : Google Scholar : PubMed/NCBI

114 

Macleod K: Tumor suppressor genes. Curr Opin Genet Dev. 10:81–93. 2000. View Article : Google Scholar : PubMed/NCBI

115 

Sherr CJ: Principles of tumor suppression. Cell. 116:235–246. 2004. View Article : Google Scholar : PubMed/NCBI

116 

Sun W and Yang J: Functional mechanisms for human tumor suppressors. J Cancer. 1:136–140. 2010. View Article : Google Scholar : PubMed/NCBI

117 

Liu HT, Liu S, Liu L, Ma RR and Gao P: EGR1-Mediated Transcription of lncRNA-HNF1A-AS1 promotes cell-cycle progression in gastric cancer. Cancer Res. 78:5877–5890. 2018. View Article : Google Scholar : PubMed/NCBI

118 

Zhang J, Cui K, Huang L, Yang F, Sun S, Bian Z, Wang X, Li C, Yin Y, Huang S, et al: SLCO4A1-AS1 promotes colorectal tumourigenesis by regulating Cdk2/c-Myc signalling. J Biomed Sci. 29:42022. View Article : Google Scholar : PubMed/NCBI

119 

Ni X, Ding Y, Yuan H, Shao J, Yan Y, Guo R, Luan W and Xu M: Long non-coding RNA ZEB1-AS1 promotes colon adenocarcinoma malignant progression via miR-455-3p/PAK2 axis. Cell Prolif. 53:e127232020. View Article : Google Scholar : PubMed/NCBI

120 

Wu X, Cui F, Chen Y, Zhu Y and Liu F: Long Non-Coding RNA LOXL1-AS1 enhances colorectal cancer proliferation, migration and invasion through miR-708-5p/CD44-EGFR Axis. Onco Targets Ther. 13:7615–7627. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Flecken T, Schmidt N, Hild S, Gostick E, Drognitz O, Zeiser R, Schemmer P, Bruns H, Eiermann T, Price DA, et al: Immunodominance and functional alterations of tumor-associated antigen-specific CD8+ T-cell responses in hepatocellular carcinoma. Hepatology. 59:1415–1426. 2014. View Article : Google Scholar : PubMed/NCBI

122 

Chow A, Perica K, Klebanoff CA and Wolchok JD: Clinical implications of T cell exhaustion for cancer immunotherapy. Nat Rev Clin Oncol. 19:775–790. 2022. View Article : Google Scholar : PubMed/NCBI

123 

Pardoll DM: The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 12:252–264. 2012. View Article : Google Scholar : PubMed/NCBI

124 

He X and Xu C: Immune checkpoint signaling and cancer immunotherapy. Cell Res. 30:660–669. 2020. View Article : Google Scholar : PubMed/NCBI

125 

Pu Y and Ji Q: Tumor-Associated macrophages regulate PD-1/PD-L1 immunosuppression. Front Immunol. 13:8745892022. View Article : Google Scholar : PubMed/NCBI

126 

Topalian SL, Drake CG and Pardoll DM: Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr opin immunol. 24:207–212. 2012. View Article : Google Scholar : PubMed/NCBI

127 

Fan F, Chen K, Lu X, Li A, Liu C and Wu B: Dual targeting of PD-L1 and PD-L2 by PCED1B-AS1 via sponging hsa-miR-194-5p induces immunosuppression in hepatocellular carcinoma. Hepatol Int. 15:444–458. 2021. View Article : Google Scholar : PubMed/NCBI

128 

Han Y, Liu D and Li L: PD-1/PD-L1 pathway: Current researches in cancer. Am J Cancer Res. 10:727–742. 2020.PubMed/NCBI

129 

Zhou Z, Liu Q, Zhang G, Mohammed D, Amadou S, Tan G and Zhang X: HOXA11-AS1 Promotes PD-L1-Mediated immune escape and metastasis of hypopharyngeal carcinoma by facilitating PTBP1 and FOSL1 Association. Cancers (Basel). 14:36942022. View Article : Google Scholar : PubMed/NCBI

130 

Chen YG, Satpathy AT and Chang HY: Gene regulation in the immune system by long noncoding RNAs. Nat immunol. 18:962–972. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Wu Y, Song Y, Wang R and Wang T: Molecular mechanisms of tumor resistance to radiotherapy. Mol Cancer. 22:962023. View Article : Google Scholar : PubMed/NCBI

132 

Hou J, Zhang G, Wang X, Wang Y and Wang K: Functions and mechanisms of lncRNA MALAT1 in cancer chemotherapy resistance. Biomark Res. 11:232023. View Article : Google Scholar : PubMed/NCBI

133 

Chen QN, Wei CC, Wang ZX and Sun M: Long non-coding RNAs in anti-cancer drug resistance. Oncotarget. 8:1925–1936. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Ni J, Bucci J, Malouf D, Knox M, Graham P and Li Y: Exosomes in cancer radioresistance. Front Oncol. 9:8692019. View Article : Google Scholar : PubMed/NCBI

135 

Zhou M, Dong J, Huang J, Ye W, Zheng Z, Huang K, Pan Y, Cen J, Liang Y, Shu G, et al: Chitosan-Gelatin-EGCG Nanoparticle-Meditated LncRNA TMEM44-AS1 Silencing to Activate the P53 signaling pathway for the synergistic reversal of 5-FU resistance in gastric cancer. Adv Sci (Weinh). 9:e21050772022. View Article : Google Scholar : PubMed/NCBI

136 

Qu L, Chen Y, Zhang F and He L: The lncRNA DLGAP1-AS1/miR-149-5p/TGFB2 axis contributes to colorectal cancer progression and 5-FU resistance by regulating smad2 pathway. Mol Ther Oncolytics. 20:607–624. 2021. View Article : Google Scholar : PubMed/NCBI

137 

Gui Z, Zhao Z, Sun Q, Shao G, Huang J, Zhao W and Kuang Y: LncRNA FEZF1-AS1 promotes multi-drug resistance of gastric cancer cells via upregulating ATG5. Front Cell Dev Biol. 9:7491292021. View Article : Google Scholar : PubMed/NCBI

138 

Zou Y, Yao S, Chen X, Liu D, Wang J, Yuan X, Rao J, Xiong H, Yu S, Yuan X, et al: LncRNA OIP5-AS1 regulates radioresistance by targeting DYRK1A through miR-369-3p in colorectal cancer cells. Eur J Cell Biol. 97:369–378. 2018. View Article : Google Scholar : PubMed/NCBI

139 

Song L, Zhou Z, Gan Y, Li P, Xu Y, Zhang Z, Luo F, Xu J, Zhou Q and Dai F: Long noncoding RNA OIP5-AS1 causes cisplatin resistance in osteosarcoma through inducing the LPAATβ/PI3K/AKT/mTOR signaling pathway by sponging the miR-340-5p. J Cell Biochem. 120:9656–9666. 2019. View Article : Google Scholar : PubMed/NCBI

140 

Wu Q, Ma J, Wei J, Meng W, Wang Y and Shi M: FOXD1-AS1 regulates FOXD1 translation and promotes gastric cancer progression and chemoresistance by activating the PI3K/AKT/mTOR pathway. Mol Oncol. 15:299–316. 2021. View Article : Google Scholar : PubMed/NCBI

141 

Qi FF, Yang Y, Zhang H and Chen H: Long non-coding RNAs: Key regulators in oxaliplatin resistance of colorectal cancer. Biomed Pharmacother. 128:1103292020. View Article : Google Scholar : PubMed/NCBI

142 

Shi CJ, Xue ZH, Zeng WQ, Deng LQ, Pang FX, Zhang FW, Fu WM and Zhang JF: LncRNA-NEF suppressed oxaliplatin resistance and epithelial-mesenchymal transition in colorectal cancer through epigenetically inactivating MEK/ERK signaling. Cancer Gene Ther. 30:855–865. 2023. View Article : Google Scholar : PubMed/NCBI

143 

Li Y, Gan Y, Liu J, Li J, Zhou Z, Tian R, Sun R, Liu J, Xiao Q, Li Y, et al: Downregulation of MEIS1 mediated by ELFN1-AS1/EZH2/DNMT3a axis promotes tumorigenesis and oxaliplatin resistance in colorectal cancer. Signal Transduct Target. 7:872022. View Article : Google Scholar : PubMed/NCBI

144 

Liang J, Tian XF and Yang W: Effects of long non-coding RNA Opa-interacting protein 5 antisense RNA 1 on colon cancer cell resistance to oxaliplatin and its regulation of microRNA-137. World J Gastroenterol. 26:1474–1489. 2020. View Article : Google Scholar : PubMed/NCBI

145 

Hui B, Lu C, Wang J, Xu Y, Yang Y, Ji H, Li X, Xu L, Wang J, Tang W, et al: Engineered exosomes for co-delivery of PGM5-AS1 and oxaliplatin to reverse drug resistance in colon cancer. J Cell Physiol. 237:911–933. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Shao Y, Dong Y, Zhou J, Lu Z, Chen C, Yuan X, He L, Tang W, Chen Z, Wang Y, Wang Y, et al: Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review). Int J Oncol 64: 22, 2024.
APA
Shao, Y., Dong, Y., Zhou, J., Lu, Z., Chen, C., Yuan, X. ... Li, Y. (2024). Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review). International Journal of Oncology, 64, 22. https://doi.org/10.3892/ijo.2024.5610
MLA
Shao, Y., Dong, Y., Zhou, J., Lu, Z., Chen, C., Yuan, X., He, L., Tang, W., Chen, Z., Wang, Y., Li, Q., Zhan, S., Qiu, Z., Wang, K., Ma, J., Chen, Y., Li, Y."Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review)". International Journal of Oncology 64.3 (2024): 22.
Chicago
Shao, Y., Dong, Y., Zhou, J., Lu, Z., Chen, C., Yuan, X., He, L., Tang, W., Chen, Z., Wang, Y., Li, Q., Zhan, S., Qiu, Z., Wang, K., Ma, J., Chen, Y., Li, Y."Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review)". International Journal of Oncology 64, no. 3 (2024): 22. https://doi.org/10.3892/ijo.2024.5610
Copy and paste a formatted citation
x
Spandidos Publications style
Shao Y, Dong Y, Zhou J, Lu Z, Chen C, Yuan X, He L, Tang W, Chen Z, Wang Y, Wang Y, et al: Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review). Int J Oncol 64: 22, 2024.
APA
Shao, Y., Dong, Y., Zhou, J., Lu, Z., Chen, C., Yuan, X. ... Li, Y. (2024). Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review). International Journal of Oncology, 64, 22. https://doi.org/10.3892/ijo.2024.5610
MLA
Shao, Y., Dong, Y., Zhou, J., Lu, Z., Chen, C., Yuan, X., He, L., Tang, W., Chen, Z., Wang, Y., Li, Q., Zhan, S., Qiu, Z., Wang, K., Ma, J., Chen, Y., Li, Y."Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review)". International Journal of Oncology 64.3 (2024): 22.
Chicago
Shao, Y., Dong, Y., Zhou, J., Lu, Z., Chen, C., Yuan, X., He, L., Tang, W., Chen, Z., Wang, Y., Li, Q., Zhan, S., Qiu, Z., Wang, K., Ma, J., Chen, Y., Li, Y."Advances in the study of antisense long‑stranded non‑coding RNAs in tumors (Review)". International Journal of Oncology 64, no. 3 (2024): 22. https://doi.org/10.3892/ijo.2024.5610
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team