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
Oncology Letters
Join Editorial Board Propose a Special Issue
Print ISSN: 1792-1074 Online ISSN: 1792-1082
Journal Cover
November-2025 Volume 30 Issue 5

Full Size Image

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
November-2025 Volume 30 Issue 5

Full Size Image

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

  • Supplementary Files
    • Supplementary_Data.pdf
Article Open Access

Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation

  • Authors:
    • Weihua Zhu
    • Jiahui Feng
    • Yuanting Zhang
    • Qianqian Zhou
    • Herong Huang
    • Yu Yang
  • View Affiliations / Copyright

    Affiliations: Department of Basic Medical Sciences, Clinical College of Anhui Medical University, Hefei, Anhui 230031, P.R. China
    Copyright: © Zhu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 531
    |
    Published online on: September 18, 2025
       https://doi.org/10.3892/ol.2025.15277
  • 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

Ovarian cancer (OC) is a prevalent gynecological malignancy requiring advancements in treatment. Resveratrol (RES), a natural polyphenolic compound, has attracted widespread attention due to its potent anticancer properties. Long non‑coding (lnc)RNAs serve crucial regulatory roles in OC pathogenesis. However, the mechanism underlying the RES‑mediated inhibition of OC cell proliferation through lncRNA regulation is not fully understood. The present study aimed to assess the role of lncRNAs in RES‑mediated inhibition of OC cell growth. The A2780 OC cell line was used as the experimental model to establish control and RES‑treated groups. The effect of RES on OC cell migration was also evaluated. Differentially expressed lncRNAs (DELs) were identified after RES treatment. lncRNA‑mRNA regulatory networks of tumor‑related pathways were constructed and functionally assessed using reverse transcription‑quantitative PCR, proliferation assays, and knockdown and overexpression of hub lncRNAs. The results demonstrated that RES significantly inhibited OC cell migration. Moreover, 721 DELs were identified after RES treatment and were predominantly enriched in the p53 signaling pathway and cell cycle regulation associated with tumorigenesis. Key lncRNAs, including lnc‑small nucleolar RNA host gene 15 (SNHG15)‑203, demonstrated significant downregulation following RES treatment. Networks of tumor‑associated mRNAs and their target lncRNAs also revealed an association with lnc‑SNHG15‑203. Furthermore, functional validation demonstrated that lnc‑SNHG15‑203 suppression markedly reduced the expression of the downstream targets H2A clustered histone (H2AC)7 and H2AC20, inhibiting cell proliferation. These results were consistent with the antiproliferative effects observed in RES‑treated cells. Overall, the present study systematically characterized the lncRNA expression profiles associated with the RES‑mediated inhibition of cell proliferation, establishing lnc‑SNHG15‑203 as a critical mediator of the anti‑OC activity of RES. The findings could help to formulate strategies for improving OC treatment.

Introduction

Ovarian cancer (OC), one of the most lethal malignancies of the female reproductive system, has a morbidity rate of 3.7% and a mortality rate of 4.7%, threatening the lives and health of patients (1). As the ovary is located in the deep pelvis, early lesions often show nonspecific symptoms, such as abdominal distension, abdominal pain and indigestion, which can be easily neglected or misdiagnosed as other diseases. Most patients are already in an advanced stage when diagnosed with OC, and the 5-year survival rate is <45%, which seriously affects patient quality of life (2). At present, the diagnosis of OC mainly relies on comprehensive methods, such as imaging, serum tumor marker detection and histopathology; however, these methods have limitations regarding sensitivity and specificity for early diagnosis (3). Therefore, exploring new diagnostic and therapeutic targets is of great importance for improving the early diagnosis rate and therapeutic effects of OC (4).

Long non-coding (lnc)RNA are a class of non-coding RNA molecules with a length of >200 nucleotides that do not encode proteins but serve an important biological function in tumorigenesis and development. They can interact with DNA and transcription factors to regulate the transcription initiation, elongation and termination of genes (5). They also serve as molecular scaffolds, recruit relevant RNA-binding proteins, form ribonucleoprotein complexes and participate in mRNA splicing, translocation and stability regulation (6). lncRNAs typically act as competitive endogenous RNAs and bind to microRNAs (miRNAs/miRs), interfering with the normal regulation of their target mRNAs by miRNAs, thus affecting the stability and translation efficiency of mRNAs (7). Several studies have reported that lncRNAs are associated with cancer development and generally exhibit abnormal expression patterns (8–10). For example, in hepatocellular carcinoma, aberrant expression of lncRNAs, such as zinc finger E-box binding homeobox 1 (ZEB1)-antisense RNA 1 (AS1), hepatocellular carcinoma upregulated lncRNA and metastasis associated lung adenocarcinoma transcript 1 (MALAT1), directly promotes tumor growth and metastasis, including abnormal cell proliferation, invasion and metastasis (11,12). The lncRNA integrin (ITG)B8-AS1 regulates colorectal cancer cell proliferation by sequestering let-7c-5p/let-7d-5p and miR-33b-5p, which in turn affects the expression of ITG family members, such as ITGB3 and ITGA3, thereby modulating focal adhesion signaling (13). In OC, HOX transcript antisense RNA upregulation in OC cells was reported to enhance cell growth and migration by interacting with miR-222-3p (14). MALAT1, a key cancer-associated lncRNA, is overexpressed in OC and regulates cell proliferation and DNA synthesis via the miR-506-iASPP axis (15). lncRNA toll like receptor 8 (TLR8)-AS1 was reported to enhance OC chemoresistance and metastasis by increasing the stability of TLR8 mRNA (16). Conversely, overexpression of maternally expressed 3 in OC was reported to be beneficial for patient prognosis and modulate the miR-219a-5p/EGFR axis (17). Homeobox D-AS1 can enhance the invasion and epithelial-mesenchymal transition process of OC cells by targeting miR-133a-3p and activating the Wnt/β-catenin signaling pathway, making tumor cells more metastatic and malignant (4).

Resveratrol (RES), a polyphenolic phytonutrient with several biological properties, has shown promising anticancer potential in preclinical studies, exerting cytotoxic effects and inducing apoptosis in tumor cells (18). RES can inhibit cell proliferation by targeting key signaling pathways that regulate cyclins or induce apoptosis by activating apoptosis-related proteins. The anticancer effects of RES have been reported in several cancer types, which are associated with the regulation of lncRNAs (19). Research has revealed that RES exerts its antitumor effects by downregulating the expression of tumor-promoting lncRNAs, including MALAT1 and small nucleolar RNA host gene (SNHG)16 (20). Our previous study further demonstrated that RES markedly inhibited the proliferation of A2780 cells whilst regulating both circular RNA and miRNA expression. Several important candidate non-coding RNAs were also screened (21). However, the specific molecular mechanisms of RES in the lncRNA regulatory network affecting cell proliferation and apoptosis have not yet been fully elucidated.

The present study aimed to assess the key biological processes and signaling pathways regulated by RES in OC cells using A2780 cell line as a model. Changes in lncRNA expression profiles after RES treatment were analyzed using high-throughput sequencing technology, screening five highly responsive RES hub genes, and constructing lncRNA-mRNA interaction networks. The findings of the present study could deepen the understanding of the molecular mechanism of RES against OC, provide potential biomarkers for the early diagnosis of OC, and lay a theoretical foundation for the development of novel targeted therapeutic strategies based on lncRNA regulation.

Materials and methods

Cell culture and RES treatment

The human A2780 OC cell line [cat. no. iCell-h004, Mirror Qidian (Shanghai) Cell Technology Co., Ltd.] was cultivated in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.), supplemented with 10% FBS and 1% antibiotic-antimycotic solution (all Gibco; Thermo Fisher Scientific, Inc.) at 37°C and 5% CO2 atmospheric conditions. Cells were incubated with 75 µM RES (Beijing Solarbio; Beijing, China) at 37°C for 24 h, where indicated, a dose previously reported to approximate an IC50 dose (21). The cells were confirmed to be free of Mycoplasma contamination and subjected to short tandem repeat identification.

RNA sequencing and expression of lncRNAs

A2780 cells were treated with or without 75 µM RES for 24 h, and TRIzol™ (Invitrogen™; Thermo Fisher Scientific, Inc.) were used to isolate total RNA from cells. RNA quality was quantified using RNase-free agarose gel electrophoresis using an Agilent 2100 Bioanalyzer (Agilent Technologies, Inc.). After eliminating ribosomal RNAs from total RNA, the samples were diluted in fragmentation buffer before reverse transcription of the mRNA/non-coding RNA transcripts with random primers using the VAHTS Universal V6 RNA-seq Library Prep Kit for Illumina (Vazyme Biotech Co., Ltd., Nanjing, China, Cat no. NR604). First-strand cDNA synthesis was performed at 25°C for 10 min, 42°C for 15 min, and 70°C for 15 min; second-strand cDNA synthesis was then performed at 16°C for 30 min, 65°C for 15 min. The cDNA fragments were purified using the QiaQuick PCR extraction kit (cat no. 28104, Qiagen, Inc.) and subsequently ligated to an Illumina sequencing adapter (Illumina, Inc.), to construct cDNA libraries. After digestion, PCR products were amplified and the library concentration was measured using the DNA 1000 assay kit (Agilent Technologies, Inc.), with quantification and pooling performed using the ABI StepOnePlus Real-Time PCR System (Thermo Fisher Scientific, Inc.). The libraries, with a loading concentration of 3 ng/µl, were then sequenced by Guangzhou Gene Denovo Biotechnology Co., Ltd. using the HiSeq 2500 Sequencing System (Illumina, Inc.) with a paired-end 150 bp sequencing strategy. Raw sequence reads were screened using fastp (version 0.18.0) to eliminate low-quality bases and adapter sequences, and high-quality clean reads were obtained (22). Once the reference genome index was constructed, clean reads from RNA sequencing were mapped to the reference genome using HISAT2 (version 2.1.0) (23). Transcript reconstruction was performed using StringTie (version 1.3.4) in combination with HISAT2 to identify the splice variants of known and novel genes (24–26). The protein-coding ability of the novel transcripts was evaluated using CNCI (version 2), CPC (version 0.9-r2) and FEELNC (version v0.2) with default parameters (27–29). The non-protein-coding ability results were intersected as lncRNAs, which were then divided into five types according to their comparison with protein-coding genes: Bidirectional, intergenic, antisense, intronic and sense-overlapping lncRNAs. For lncRNAs and mRNAs, the differential expression of their transcripts was assessed separately using DESeq2 (version 1.20.0) between groups and edgeR (version 3.20.9) between samples (30,31). Differential expression of genes/transcripts was determined using |fold change|≥2 and false discovery rate of <0.05.

Bioinformatics analysis

RNAplex (version 0.2) and the Vienna RNA package were used to predict the complementary relationships between antisense lncRNAs and mRNA (32). Optimal base pairing was predicted using minimal free-energy prediction based on thermodynamics. lncRNAs can function to cis-regulate the genes surrounding an identical allele (33). Upstream lncRNAs that interact with promoters or additional cis-elements can modulate gene expression transcriptionally or post-transcriptionally (33). Typically, lncRNAs in the 3′untranslated region or downstream can show additional regulatory effects. Therefore, lncRNAs annotated as ‘unknown region’ were reannotated as cis-regulators at <10 kb up/downstream of one specific gene. lncRNAs can also trans-regulate long-distance genes (33). Pearson's correlation coefficients were calculated between lncRNA and protein-coding genes, with the pairs satisfying |correlation|>0.999, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed. Moreover, the functions of the lncRNA parental genes were evaluated using GO and KEGG analyses. GO annotation was performed using GO resources (http://www.geneontology.org/).

lncRNA-mRNA association analysis

Based on the results of KEGG enrichment analysis, combined with published evidence supporting the anticancer mechanism pathways, including the p53 signaling pathway, oocyte meiosis and cell cycle pathways, differentially expressed lncRNAs (DELs) and mRNAs and in these pathways were screened for co-expression analysis (34–36). The lncRNA-mRNA co-expression network was visualized using Cytoscape software (version 3.9.1, cytoscape.org/).

Validation of DELs and mRNAs using RT-quantitative (qPCR)

RT-qPCR was performed to assess DELs and mRNA expression. Total RNA was extracted using the Ultrapure RNA kit (DNase I, Cat no. CW0597S, Jiangsu Cowin Biotech Co., Ltd). cDNA was synthesized using Easyscript one-step gDNA removal and cDNA synthesis supermix (Cat no. AE-311, TransGen Biotech Co., Ltd.) with incubation at 42°C for 1 h. Specific primers were designed for RT-qPCR assays in a 20 µl reaction (Table SI). Reactions were performed using a Light-Cycler96 instrument (Roche Diagnostics) with the TransStart green qPCR supermix containing SYBR GREEN (Cat no. AQ131-01, TransGen Biotech Co., Ltd.). The thermocycling conditions were as follows: pre-denaturation at 94°C for 30 sec, followed by 40 cycles with denaturation at 94°C for 5 sec and annealing at 61°C for 35 sec. Post-amplification, melting curve analysis was performed at 95°C for 10 sec, 65°C for 60 sec, and 97°C for 1 sec. Relative RNA expression subsequently calculated as 2−ΔΔCq values based on GAPDH as the housekeeping gene (37). Each experiment was performed in triplicate to ensure reproducibility.

Cell transfection

pcDNA3.1-SNHG15-203 overexpression plasmids (Nanjing GenScript Co., Ltd.) and small interfering (si)RNAs targeting SNHG15-203 (Table SII, Shanghai GenePharma Co., Ltd.) were constructed and transfected into A2780 cells. Specifically, 200 nM siRNA duplexes or the overexpression plasmid, and 2.5 µl Lipofectamine™ 2000 (Thermo Fisher Scientific, Inc.) were diluted in 100 µl Opti-MEM, mixed, and incubated at 25°C for 20 min before being added to each well of a 12-well plate containing target cells. An additional 800 µl Opti-MEM was introduced, followed by gentle shaking. Transfection was performed at 25°C. The transfection mixture was replaced with fresh medium without antibiotics after 6–8 h, and the cells were further cultured at 37°C for 48 h before harvesting. RT-qPCR was performed to validate the overexpression and knockdown efficiency, as aforementioned.

Cell Counting Kit-8 (CCK-8) assay

Following transfection, cell proliferation was assessed using a CCK-8 kit (Biosharp Life Sciences). Briefly, the CCK-8 reagent was added to the treated cells cultured in a 96-well plate. After incubation for 48 h, the optical density of each well was measured at 450 nm using a microplate reader (NanJing DeTie Laboratory Equipment Co., Ltd.). Each experiment was performed in triplicate.

Transwell assay

Cell migration was assessed using Transwell chambers (Corning, Inc.) with a pore size of 8 µm. A total of 2×105 cells in 1 ml serum-free medium was added to the upper Transwell chamber, and 600 µl complete medium containing 10% FBS was added to the lower chamber. After 24 h incubation at 37°C, cells were fixed in paraformaldehyde at room temperature for 15 min and stained with crystal violet at room temperature for 10 min. The number of migrating cells in random areas was counted under an inverted microscope (IX51; Olympus Corporation).

Statistical analysis

GraphPad Prism statistical software (version 8.0; Dotmatics) was used for experimental data processing. For comparisons between two groups, an unpaired two-tailed Student's t-test was used if the data were normally distributed and had equal variances; otherwise, the Mann-Whitney U test was applied. For comparisons among multiple groups, one-way analysis of variance was used for parametric data, followed by Tukey's HSD test for multiple comparisons; otherwise, the Kruskal-Wallis H-test was used, followed by Dunn's test for post hoc pairwise comparisons. Pearson's correlation coefficient was used for correlation analysis. Data are expressed as mean ± standard error of the mean of ≥3 independent experimental repeats. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of RES on the migration of OC cells

The effects of RES treatment on the migration of the OC cell line, A2780, were assessed. Transwell assays revealed that cells in the control group had a strong migration ability. The number of cells passing through the membrane was high, and numerous cells were observed on the lower surface of the membrane in the field of view (Fig. 1). By contrast, the migration ability of cells treated with RES for 24 h was notably altered, with the number of cells crossing the membrane was greatly reduced. The difference was significant compared with that in the control group (P<0.01). These results indicate that RES significantly inhibited the migration of A2780 OC cells.

Migration of resveratrol-treated
ovarian cancer cells assessed using Transwell assay (magnification,
×100; scale bar, 1 mm). **P<0.01.

Figure 1.

Migration of resveratrol-treated ovarian cancer cells assessed using Transwell assay (magnification, ×100; scale bar, 1 mm). **P<0.01.

RES treatment alters the lncRNA expression profile of OC cells

To evaluate the potential mechanism underlying the inhibitory effect of RES on the proliferation of OC cells, lncRNA-sequencing analysis was performed on the OC cell line A2780 in the control and RES-treated groups. Novel lncRNAs were first identified and categorized into the following five groups based on their positions in the genome: 32,988 sense lncRNAs, 10,687 antisense lncRNAs, 7,316 intronic lncRNAs, 4,552 bidirectional lncRNAs and 35,927 intergenic lncRNAs (Fig. 2A). Pearson's correlation analysis demonstrated that lncRNA expression among biological replicates within groups showed a high degree of similarity, with significant differences between groups (Fig. 2B). Further analysis revealed 721 DELs, of which 461 were upregulated and 260 were downregulated (Fig. 2C). Hierarchical clustering analysis demonstrated significant differences in the lncRNA expression patterns between the two groups (Fig. 2D). Notably, lncRNAs H19-203 and vacuole membrane protein 1 (VMP1)-217 were highly expressed in the RES-treated group, whereas lncRNAs enolase 1 (ENO1)-210, SNHG15-203 and SNHG1-266 were markedly downregulated (Table SIII). Meanwhile, 24 SNHG family lncRNAs were identified, including lncRNA SNHG15-203.

lncRNA expression profiles of
RES-treated ovarian cancer cells. (A) Statistical analysis of
different types of lncRNA transcripts according to their genomic
locations. (B) Pearson's correlation analysis. (C) Volcano plot
depicting the DELs in the RES-treated and the control groups. (D)
Hierarchical clustering heatmap demonstrating the expression
profiles of DELs between RES-treated and control groups. Red, high
expression levels, the bluer one indicates lower expression levels.
lncRNA, long non-coding RNA; RES, resveratrol; DELs, differentially
expressed lncRNAs; C, control group; T, RES-treated group; FDR,
false discovery rate; FC, fold change.

Figure 2.

lncRNA expression profiles of RES-treated ovarian cancer cells. (A) Statistical analysis of different types of lncRNA transcripts according to their genomic locations. (B) Pearson's correlation analysis. (C) Volcano plot depicting the DELs in the RES-treated and the control groups. (D) Hierarchical clustering heatmap demonstrating the expression profiles of DELs between RES-treated and control groups. Red, high expression levels, the bluer one indicates lower expression levels. lncRNA, long non-coding RNA; RES, resveratrol; DELs, differentially expressed lncRNAs; C, control group; T, RES-treated group; FDR, false discovery rate; FC, fold change.

Feature distribution comparison of lncRNAs and mRNAs

Previously, mRNA sequencing in the RES-treated OC cell line A2780 revealed significant changes in mRNA expression in RES-treated cells and clarified its key role in cell proliferation and immunomodulation. Based on this, the potential roles of lncRNAs and their synergistic regulation of mRNAs were further explored. Compared with mRNAs, both annotated and novel lncRNAs exhibited fewer exons, shorter lengths and lower coding potentials (Fig. 3A-C). Analysis of total lncRNA expression data demonstrated markedly lower levels than those of mRNA, and the abundances of lncRNAs and mRNAs were notably increased by RES treatment (Fig. 3D). Differences in transcript abundance were also observed between DELs and mRNAs (Fig. 3E). In addition, the coding potential scores of differentially expressed mRNAs were higher than those of the lncRNAs, and those of novel lncRNAs were significantly lower than those of the annotated lncRNAs (Fig. 3F).

Feature distribution comparison of
lncRNAs and mRNAs. (A) Exon number. (B) Transcript length. (C) ORF
length. (D) Expression levels of mRNA and lncRNA transcripts in
control and resveratrol-treated groups. (E) Abundance of
differential mRNA and lncRNA transcripts. (F) Encoding potential
scores of mRNA and lncRNA transcripts. ***P<0.001. lncRNA, long
non-coding RNA; ORF, open reading frame; C, control group; T,
resveratrol-treated group; TPM, transcripts per million.

Figure 3.

Feature distribution comparison of lncRNAs and mRNAs. (A) Exon number. (B) Transcript length. (C) ORF length. (D) Expression levels of mRNA and lncRNA transcripts in control and resveratrol-treated groups. (E) Abundance of differential mRNA and lncRNA transcripts. (F) Encoding potential scores of mRNA and lncRNA transcripts. ***P<0.001. lncRNA, long non-coding RNA; ORF, open reading frame; C, control group; T, resveratrol-treated group; TPM, transcripts per million.

Target gene prediction and functional enrichment analysis of lncRNAs

To further assess the potential function of lncRNAs in the inhibition of OC cell proliferation by RES, the complementary binding relationship between lncRNAs and mRNAs was analyzed and the potential function of lncRNAs was inferred based on the functions of known mRNAs. The DELs were first predicted for cis and trans roles, and 5,503 and 1,780,009 lncRNA-mRNA target gene pairs were obtained, respectively. Subsequently, GO functional enrichment analysis of the DELs was performed. The results revealed that the target genes associated with these differentially expressed cis-acting lncRNAs were significantly enriched in several key biological processes, such as response to viruses, regulation of signal transduction by p53 class mediators, DNA metabolic processes and response to oxidative stress (Fig. 4A). KEGG enrichment analysis further indicated that these cis-acting lncRNAs were mainly involved in carcinogenesis, Notch, Hippo, cell cycle and other classical tumor-related signaling pathways (Fig. 4B). The Reactome pathways involved also covered cell cycle progression and antiviral activity, which corroborated the GO and KEGG analysis results (Fig. 4C). the GO function of the differentially expressed trans-acting lncRNAs were analyzed, which revealed that they were mainly enriched in signal transduction and response to stress and stimuli (Fig. 4D). The results of KEGG enrichment analysis demonstrated that the trans-acting lncRNAs were mainly involved in p53, Forkhead Box (Fox)O signaling pathway, cellular senescence, extracellular matrix-receptor interaction and cancer-related signaling pathways, whilst the Reactome pathways included deacetylation of histones, Signaling by Rho GTPases and phosphorylation (Fig. 4E and F).

Functional analysis of differentially
expressed lncRNAs. (A) GO, (B) KEGG pathway and (C) Reactome
enrichment analysis of differentially expressed cis-acting
lncRNAs. (D) GO, (E) KEGG pathway and (F) Reactome enrichment
analysis of differentially expressed trans-acting lncRNAs.
lncRNA, long non-coding RNA; GO, Gene Ontology; KEGG, Kyoto
Encyclopedia of Genes and Genomes.

Figure 4.

Functional analysis of differentially expressed lncRNAs. (A) GO, (B) KEGG pathway and (C) Reactome enrichment analysis of differentially expressed cis-acting lncRNAs. (D) GO, (E) KEGG pathway and (F) Reactome enrichment analysis of differentially expressed trans-acting lncRNAs. lncRNA, long non-coding RNA; GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes.

Furthermore, certain genes were revealed to be involved in multiple important biological functions (Fig. 4A-F). For example, cyclin dependent kinase inhibitor 1A (CDKN1A) and sequestosome (SQSTM) were enriched in cellular entries, anatomical entities, binding, cellular processes and biological regulation, and were associated with cellular senescence and cell cycle signaling pathways. Reactome analysis revealed certain specific pathways, such as estrogen-responsive MYC gene expression, lysine demethylase 6B demethylation of H3K27me3 on the p16-INK4A promoter, CDK1 phosphorylation of PHD finger protein 8, binding of serum amyloid P to DNA and chromatin, and binding of methyl-CpG binding domain protein 2 to methylcytosine in chromatin. These specific pathways contain several members of the histone H2A family, such as H2AC7, H2AC20 and H2AC18. Therefore, we hypothesize that the regulatory network centered on CDKN1A, SQSTM1 and histones may serve a crucial role in the inhibition of A2780 cell proliferation by RES.

lncRNA-mRNA networks

A total of five mRNAs associated with the p53 signaling pathway, oocyte meiosis, cellular senescence, cell cycle and their associated lncRNAs were selected and lncRNA-mRNA regulatory networks were constructed using Cytoscape. The results revealed that CDKN1A and SQSTM1 formed a large regulatory network, whereas the other genes, H2AC7, H2AC20 and H2AC18, formed a small network. Moreover, there was no overlap in the lncRNAs involved in the two networks (Fig. 5 and Table SIV). In the large network, CDKN1A and SQSTM1 demonstrated extensive regulatory associations, with shared regulatory links with 26 lncRNAs, including lnc-SQSTM1-206 and MSTRG.5444.1. It also formed unique regulatory relationships with MSTRG.22456.2 and MSTRG.3858.2. In the small network, H2AC7 and H2AC20 were targeted by five lncRNAs, including SNHG15-203 and cysteinyl-tRNA synthetase 1–212. A specific linkage between H2AC18, H2AC2 and MSTRG.7008.104 allowed the integration of the three histone genes into the same network.

Long non-coding RNA-mRNA networks.
CDKN1A, SQSTM1, H2AC7, H2AC20 and H2AC18 are hub
genes in the networks. CDKN1A, cyclin dependent kinase inhibitor
1A; SQSTM1, sequestosome 1; H2AC, H2A clustered histone.

Figure 5.

Long non-coding RNA-mRNA networks. CDKN1A, SQSTM1, H2AC7, H2AC20 and H2AC18 are hub genes in the networks. CDKN1A, cyclin dependent kinase inhibitor 1A; SQSTM1, sequestosome 1; H2AC, H2A clustered histone.

Validation of expression levels of lncRNAs and genes

To verify the accuracy of the data, five lncRNAs (ENO1-210, SNHG1-266, SNHG15-203, VMP1-217 and H19-203) were randomly selected and their expression levels were assessed using RT-qPCR. The expression levels of ENO1-210, SNHG1-266 and SNHG15-203 were significantly downregulated, and the expression levels of VMP1-217 and H19-203 were significantly upregulated in the RES-treated group compared with in the control group (Fig. 6A). These results are consistent with the RNA sequencing results (Table SIII).

RT-qPCR validation of long non-coding
RNA and mRNA expression in 75 µM resveratrol-treated ovarian cancer
cells. (A) RT-qPCR and RNA-seq quantification results of lncRNA.
Orange bars indicate RT-qPCR results, and blue lines indicate
RNA-seq results. (B) RT-qPCR results of mRNA expression.
*P<0.05; **P<0.01; ***P<0.001. RT-qPCR, reverse
transcription-quantitative PCR; RNA-seq, RNA-sequencing; TPM,
transcripts per million; ENO1, enolase 1; SNHG, small nucleolar RNA
host gene; VMP1, vacuole membrane protein 1; CDKN1A, cyclin
dependent kinase inhibitor 1A; SQSTM1, sequestosome 1; H2AC, H2A
clustered histone.

Figure 6.

RT-qPCR validation of long non-coding RNA and mRNA expression in 75 µM resveratrol-treated ovarian cancer cells. (A) RT-qPCR and RNA-seq quantification results of lncRNA. Orange bars indicate RT-qPCR results, and blue lines indicate RNA-seq results. (B) RT-qPCR results of mRNA expression. *P<0.05; **P<0.01; ***P<0.001. RT-qPCR, reverse transcription-quantitative PCR; RNA-seq, RNA-sequencing; TPM, transcripts per million; ENO1, enolase 1; SNHG, small nucleolar RNA host gene; VMP1, vacuole membrane protein 1; CDKN1A, cyclin dependent kinase inhibitor 1A; SQSTM1, sequestosome 1; H2AC, H2A clustered histone.

The expression levels of key mRNAs associated with the lncRNAs were further evaluated. A total of five key genes, CDKN1A, SQSTM1, H2AC7, H2AC20 and H2AC18, were evaluated for changes in expression levels, confirming alterations in the mRNA levels of each gene after RES treatment. Among these, CDKN1A and SQSTM1 were significantly upregulated, whereas H2AC7, H2AC20 and H2AC18 were significantly downregulated, compared with in the control group (Fig. 6B).

lnc-SNHG15-203 regulates OC cell proliferation and target gene expression

To assess the predicted regulatory network, lnc-SNHG15-203 was selected for an in-depth study, as it was predicted to regulate both H2AC7 and H2AC20. First, four siRNAs and plasmids were designed to knock-down and overexpress lnc-SNHG15-203 in A2780 cells, respectively. The knockdown efficiencies of different siRNAs varied, with the knockdown efficiency of siRNA-2 at >70%, whereas that of siRNA-1 was ~30% (Fig. 7A). By contrast, the overexpression group demonstrated significant upregulation, with the expression level increasing by ~12-fold compared with that in the control group (Fig. 7B). For further validation, siRNA-1 and siRNA-2 were also used. The CCK-8 cell proliferation assay results demonstrated that both siRNA-1 and siRNA-2 significantly inhibited the proliferation of A2780 cells compared with the siRNA negative control, and lnc-SNHG15-203 overexpression notably promoted cell proliferation compared with the empty vector control group (Fig. 7C). Compared with the negative control group, the expression levels of its potential target genes, H2AC7 and H2AC20, were notably downregulated after lnc-SNHG15-203 knock-down (Fig. 7D and E). Conversely, compared with the empty vector control group, lnc-SNHG15-203 overexpression significantly upregulated H2AC7 and H2AC20, with their expression levels increased by ~2- and 3-fold, respectively. These results suggest that knockdown of lnc-SNHG15-203, a type of lncRNA that is downregulated by RES, could inhibit the expression of its target genes and cell proliferation, whereas its overexpression exerts the opposite effects.

Effect of lnc-SNHG15-203 on
proliferation and target gene expression in ovarian cancer cells.
(A) Expression efficiency of siRNAs targeting
lnc-SNHG15-203. (B) Overexpression efficiency of
lnc-SNHG15-203. (C) Proliferation efficiency of A2780 cells
treated with siRNA and overexpression plasmid for
lnc-SNHG15-203, determined using Cell-Counting Kit-8 assays.
Relative expression levels of (D) H2AC7 and (E)
H2AC20 mRNA, determined using reverse
transcription-quantitative PCR. **P<0.01; ***P<0.001. lnc,
long non-coding; SNHG, small nucleolar RNA host gene; si, small
interfering; H2AC, H2A clustered histone; NC, negative control; OE,
overexpression.

Figure 7.

Effect of lnc-SNHG15-203 on proliferation and target gene expression in ovarian cancer cells. (A) Expression efficiency of siRNAs targeting lnc-SNHG15-203. (B) Overexpression efficiency of lnc-SNHG15-203. (C) Proliferation efficiency of A2780 cells treated with siRNA and overexpression plasmid for lnc-SNHG15-203, determined using Cell-Counting Kit-8 assays. Relative expression levels of (D) H2AC7 and (E) H2AC20 mRNA, determined using reverse transcription-quantitative PCR. **P<0.01; ***P<0.001. lnc, long non-coding; SNHG, small nucleolar RNA host gene; si, small interfering; H2AC, H2A clustered histone; NC, negative control; OE, overexpression.

Discussion

OC is the leading cause of mortality in women with gynecological cancers, with marked heterogeneity. Currently, surgery combined with chemotherapy is the mainstream treatment for OC. Although initial responses to therapy are often encouraging, there is a high recurrence rate, leading to a poor prognosis. Therefore, there is an urgent need to investigate the progression mechanisms of OC and develop novel therapeutic strategies. The natural product RES has been reported to have complex anticancer activities (38). Therefore, the present study analyzed the changes in lncRNA expression profiles after RES treatment in A2780 cells, a prototypical model of OC, to reveal the molecular mechanism by which RES inhibits the proliferation of OC cells by regulating the lncRNA-mRNA network. The analysis identified 1,038 novel lncRNA transcripts and 721 DELs. RES-regulated DELs were demonstrated to be mainly involved in the p53, FoxO, cellular senescence and cell cycle signaling pathways, which are biological processes associated with tumorigenesis and development (34–36). Reactome enrichment identified special pathways containing members of the histone H2A family of genes. These genes serve important regulatory roles in gene expression, DNA repair and structural stability of chromatin (39,40).

By combining the functional pathways and effect of RES on the mRNA expression profile from a previous study (21), the present study constructed a lncRNA-mRNA regulatory network. This yielded two discrete networks: A larger network formed by the upregulated CDKN1A and SQSTM1 genes, and a smaller network involving the downregulated H2AC7, H2AC20 and H2AC18 genes. In particular, key lncRNAs in the CDKN1A-SQSTM1 regulatory network serve important regulatory roles in several cancers. For instance, detected as part of the CDKN1A-SQSTM1 network, lnc-MAP3K13-7:1 suppresses OC proliferation in the context of polycystic ovary syndrome by inducing hypomethylation of the CDKN1A promoter by downregulating DNA methyltransferase 1 (41). lnc-cancer susceptibility 15 was reported to modulate GC cell proliferation, migration and epithelial-mesenchymal transition via ZEB1 and CDKN1A (42). Moreover, the lncRNA RUN and SH3 domain containing 1-AS1 was reported to enhance breast cancer cell growth through the epigenetic silencing of CDKN1A, Kruppel-like factor transcription factor 2 and Misato homolog 2 pseudogene (MSTO2P) upregulation in colorectal cancer, which promotes growth via enhancer of zeste homolog 2-mediated epigenetic silencing of CDKN1A (43,44). MSTO2P inhibition inhibited the growth and migration of HT29 and SW480 colorectal cancer cells, leading to cell cycle arrest and apoptosis. MNX1-AS1 is also markedly upregulated in GC compared with its matched counterpart tissues, affecting overall patient survival. Furthermore, it is functionally implicated in GC cell invasion and migration. MNX1-AS1 overexpression has also been reported to downregulate CDKN1A expression (45). Additionally, prostate cancer-associated transcript 1 was reported to promote the growth, invasion and migration of MGC-803 and AGS cells via CDKN1A (46). High expression of CDKN1A is considered an important prognostic factor for improving the overall survival of OC, and SQSTM1 has a notable impact on the multidrug resistance of OC (47,48). Nevertheless, this network involving multiple molecular interactions and regulation, requires further in-depth studies to elucidate the specific mechanisms underlying OC progression.

Although the roles of CDKN1A and SQSTM1 in tumors have been widely reported, therapeutic strategies may be limited due to drug resistance (49). Therefore, the present study focused on H2AC7/H2AC20. Multiple studies have reported that abnormal histone modifications serve a key role in the occurrence and development of OC, and that H2A family members can participate in malignant tumor phenotypes by regulating chromatin structure (50–52). The associations between the H2AC7-H2AC20-H2AC18 network and cancer have not been well-defined, and their roles in OC have not been described. However, these genes have also been studied in other contexts. For example, H2AC7 was identified as one of the 14 placental candidate genes in a screen for causal genes associated with spontaneous abortion (53). H2AC20 was suggested as a potential DNA methylation candidate gene during infection with Mycobacterium avium paratuberculosis, whereas H2AC18 was reported to be one of the most altered genes in the peripheral white blood cells of cows following embryo transfer (54,55). Members of the histone H2A family may influence the occurrence and progression of cancer by affecting the chromatin structure and regulating gene expression. lncRNAs participate in the regulation of biological processes by regulating target genes (56). The potential associations among H2AC7, H2AC20, H2AC18 and lncRNAs in cancer warrant further exploration to reveal their complex regulatory mechanisms in cancer initiation and progression. Moreover, lncRNA SNHGs are host genes for small nucleolar RNA. The SNHG family includes 22 members, most of which exert inhibitory or promoting functions in the initiation and progression of cancer, and their expression is abnormal in multiple cancers (57). As a lncRNA, SNHG is involved in tumorigenesis through several molecular regulatory mechanisms. The present study identified 24 SNHG lncRNAs, which may affect the expression of cell proliferation-associated genes through different mechanisms of action, serve a role in the treatment of OC and provide a possible direction for the development of novel therapeutic strategies based on the SNHG family lncRNAs. The network built in the present study also revealed lnc-SNHG15-203 in the core. Furthermore, given the lack of cancer research, the specific effects of lnc-SNHG15-203 on A2780 cells in vitro were assessed. In the network map, lnc-SNHG15-203 was evaluated, which is predicted to regulate H2AC7 and H2AC20. Similar to RES treatment, it was demonstrated that silencing lnc-SNHG15-203 significantly reduced the expression of H2AC7 and H2AC20 in A2780 cells. Moreover, this treatment inhibited cell proliferation, indicating that lncRNA-SNHG15-203 may normally promote cell proliferation by upregulating H2AC7 and H2AC20, and that RES acts to counter this signaling axis. The results infer that the SNHG family lncRNAs may inhibit the development of OC by influencing processes such as cell proliferation and may become potential targets.

Furthermore, the RNA-sequencing (RNA-seq) results suggest that DELs may be important in the inhibition of A2780 cell proliferation by RES by regulating its target genes, and participating in cell proliferation, senescence and apoptosis-related signaling pathways. By analyzing the expression profiles of lncRNAs, specific lncRNAs that were upregulated or downregulated in OC were identified. These lncRNAs, including SNHG15-203, could serve as potential biomarkers. These findings demonstrate the utility of lncRNA-mRNA regulatory networks in revealing RES targets and their underlying regulatory mechanisms, and emphasize the importance of considering lncRNAs and related genes in the treatment process, suggesting that the data in the present study and the construction of additional networks are important for OC treatment.

However, the present study has certain limitations. First, the in vitro experiments were performed using the A2780 OC cell line only, which may not fully reflect the heterogeneity of the different subtypes. Second, the lack of in vivo validation prevents confirmation of whether the regulatory effects observed in vitro are valid in vivo. Additionally, due to the lack of immunoprecipitation-level antibodies against the target proteins and database prediction of no binding sites, the present study was unable to confirm the specific mechanism of interaction between lnc-SNHG15-203 and H2AC7/H2AC20. Future research should be expanded to include multiple cell models and in vivo experiments, and alternative approaches to elucidate the molecular interaction mechanisms to enhance the generalizability of the conclusions.

In summary, the present study comprehensively assessed the role of RES in inhibiting OC cell proliferation by regulating lncRNAs using RNA-seq. The expression profiles of lncRNAs involved in RES-mediated inhibition of OC cell proliferation were systematically identified. These DELs were associated with several important signaling pathways. Subsequently, the lncRNA-mRNA regulatory networks were constructed. In addition, the results demonstrated that lnc-SNHG15-203 regulates A2780 cell proliferation and H2AC7 and H2AC20 expression. These findings provide a new perspective for the treatment of OC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present research was funded by the Scientific Research Project of Colleges and Universities in Anhui Province (grant no. 2024AH050832), Natural Science Research Project of Clinical College of Anhui Medical University (grant no. 2024XJKY001) and Research Platform Project of Clinical College of Anhui Medical University: Collaborative Innovation Platform for Tumor Translational Medicine (grant no. 2025PT005).

Availability of data and materials

The original RNA-sequencing data generated and in the present study may be found in the BioProject database under accession number PRJNA1119553 or at the following URL: https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA1119553. All other data generated in the present study may be requested from the corresponding author.

Authors' contributions

WZ conceived and designed the study, developed the methodology and wrote the manuscript. JF and YZ performed formal analysis. JF and QZ conducted the investigation. HH and YY interpreted the results. JF edited the manuscript. WZ and JF confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Webb PM and Jordan SJ: Global epidemiology of epithelial ovarian cancer. Nat Rev Clin Oncol. 21:389–400. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Wang A, Wang Y, Du C, Yang H, Wang Z, Jin C and Hamblin MR: Pyroptosis and the tumor immune microenvironment: A new battlefield in ovarian cancer treatment. Biochim Biophys Acta Rev Cancer. 1879:1890582024. View Article : Google Scholar : PubMed/NCBI

3 

Sideris M, Menon U and Manchanda R: Screening and prevention of ovarian cancer. Med J Aust. 220:264–274. 2024. View Article : Google Scholar : PubMed/NCBI

4 

Zhang Y, Dun Y, Zhou S and Huang XH: LncRNA HOXD-AS1 promotes epithelial ovarian cancer cells proliferation and invasion by targeting miR-133a-3p and activating Wnt/beta-catenin signaling pathway. Biomed Pharmacother. 96:1216–1221. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Liu J, Zhang Q, Yang D, Xie F and Wang Z: The role of long non-coding RNAs in angiogenesis and anti-angiogenic therapy resistance in cancer. Mol Ther Nucleic Acids. 28:397–407. 2022. View Article : Google Scholar : PubMed/NCBI

6 

Zhu YS and Zhu J: Molecular and cellular functions of long non-coding RNAs in prostate and breast cancer. Adv Clin Chem. 106:91–179. 2022. View Article : Google Scholar : PubMed/NCBI

7 

Zhou Z, Leng C, Wang Z, Long L, Lv Y, Gao Z, Wang Y, Wang S and Li P: The potential regulatory role of the lncRNA-miRNA-mRNA axis in teleost fish. Front Immunol. 14:10653572023. View Article : Google Scholar : PubMed/NCBI

8 

Zhao Y, Yuan D, Zhu D, Xu T, Huang A, Jiang L, Liu C, Qian H and Bu X: LncRNA-MSC-AS1 inhibits the ovarian cancer progression by targeting miR-425-5p. J Ovarian Res. 14:1092021. View Article : Google Scholar : PubMed/NCBI

9 

Fang W and Xia Y: LncRNA HLA-F-AS1 attenuates the ovarian cancer development by targeting miR-21-3p/PEG3 axis. Anticancer Drugs. 33:671–681. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Dong Q, Qiu H, Piao C, Li Z and Cui X: LncRNA SNHG4 promotes prostate cancer cell survival and resistance to enzalutamide through a let-7a/RREB1 positive feedback loop and a ceRNA network. J Exp Clin Cancer Res. 42:2092023. View Article : Google Scholar : PubMed/NCBI

11 

Tan YT, Lin JF, Li T, Li JJ, Xu RH and Ju HQ: LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun (Lond). 41:109–120. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Zhou D, Wang Y, Hu H, Liu H, Deng J, Li L and Zheng C: lncRNA MALAT1 promotes HCC metastasis through the peripheral vascular infiltration via miRNA-613: A primary study using contrast ultrasound. World J Surg Oncol. 20:2032022. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Fan L, Lei H, Lin Y, Zhou Z, Li J, Wu A, Shu G, Roger S and Yin G: Hotair promotes the migration and proliferation in ovarian cancer by miR-222-3p/CDK19 axis. Cell Mol Life Sci. 79:2542022. View Article : Google Scholar : PubMed/NCBI

15 

Lei R, Xue M, Zhang L and Lin Z: Long noncoding RNA MALAT1-regulated microRNA 506 modulates ovarian cancer growth by targeting iASPP. Onco Targets Ther. 10:35–46. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Xu Q, Lin YB, Li L and Liu J: LncRNA TLR8-AS1 promotes metastasis and chemoresistance of ovarian cancer through enhancing TLR8 mRNA stability. Biochem Biophys Res Commun. 526:857–864. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Wang L, Yu M and Zhao S: lncRNA MEG3 modified epithelial-mesenchymal transition of ovarian cancer cells by sponging miR-219a-5p and regulating EGFR. J Cell Biochem. 120:17709–17722. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Ren B, Kwah MX, Liu C, Ma Z, Shanmugam MK, Ding L, Xiang X, Ho PC, Wang L, Ong PS and Goh BC: Resveratrol for cancer therapy: Challenges and future perspectives. Cancer Lett. 515:63–72. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Rauf A, Imran M, Butt MS, Nadeem M, Peters DG and Mubarak MS: Resveratrol as an anti-cancer agent: A review. Crit Rev Food Sci Nutr. 58:1428–1447. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Asemi R, Rajabpoor Nikoo N, Asemi Z, Shafabakhsh R, Hajijafari M, Sharifi M, Homayoonfal M, Davoodvandi A and Hakamifard A: Modulation of long non-coding RNAs by resveratrol as a potential therapeutic approach in cancer: A comprehensive review. Pathol Res Pract. 246:1545072023. View Article : Google Scholar : PubMed/NCBI

21 

Zhu W, Zhang Y, Zhou Q, Zhen C, Huang H and Liu X: Identification and Comprehensive analysis of circRNA-miRNA-mRNA regulatory networks in A2780 cells treated with resveratrol. Genes (Basel). 15:9652024. View Article : Google Scholar : PubMed/NCBI

22 

Chen S, Zhou Y, Chen Y and Gu J: Fastp: An ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 34:i884–i890. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Kim D, Langmead B and Salzberg SL: HISAT: A fast spliced aligner with low memory requirements. Nat Methods. 12:357–360. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, Salzberg SL, Wold BJ and Pachter L: Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 28:511–515. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT and Salzberg SL: StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 33:290–295. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Pertea M, Kim D, Pertea GM, Leek JT and Salzberg SL: Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat Protoc. 11:1650–1667. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Sun L, Luo H, Bu D, Zhao G, Yu K, Zhang C, Liu Y, Chen R and Zhao Y: Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts. Nucleic Acids Res. 41:e1662013. View Article : Google Scholar : PubMed/NCBI

28 

Kong L, Zhang Y, Ye ZQ, Liu XQ, Zhao SQ, Wei L and Gao G: CPC: Assess the protein-coding potential of transcripts using sequence features and support vector machine. Nucleic Acids Res. 35:W345–W349. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Wucher V, Legeai F, Hedan B, Rizk G, Lagoutte L, Leeb T, Jagannathan V, Cadieu E, David A, Lohi H, et al: FEELnc: A tool for long non-coding RNA annotation and its application to the dog transcriptome. Nucleic Acids Res. 45:e572017.PubMed/NCBI

30 

Love MI, Huber W and Anders S: Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15:5502014. View Article : Google Scholar : PubMed/NCBI

31 

Robinson MD, McCarthy DJ and Smyth GK: edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Tafer H and Hofacker IL: RNAplex: A fast tool for RNA-RNA interaction search. Bioinformatics. 24:2657–2663. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Wilusz JE, Sunwoo H and Spector DL: Long noncoding RNAs: Functional surprises from the RNA world. Genes Dev. 23:1494–1504. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Nadile M, Retsidou MI, Gioti K, Beloukas A and Tsiani E: Resveratrol against Cervical Cancer: Evidence from in vitro and in vivo studies. Nutrients. 14:52732022. View Article : Google Scholar : PubMed/NCBI

35 

Liu X, Li P, Yan K, Du Y, Peng K, Li M, Cui K, Zhang H, Yang X, Lu S and Liang X: Resveratrol ameliorates the defects of meiotic maturation in lipopolysaccharide exposed porcine oocytes. Reprod Toxicol. 115:85–93. 2023. View Article : Google Scholar : PubMed/NCBI

36 

Wu H, Chen L, Zhu F, Han X, Sun L and Chen K: The cytotoxicity effect of resveratrol: Cell cycle arrest and induced apoptosis of breast cancer 4T1 cells. Toxins (Basel). 11:7312019. View Article : Google Scholar : PubMed/NCBI

37 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Xu XL, Deng SL, Lian ZX and Yu K: Resveratrol targets a variety of oncogenic and oncosuppressive signaling for ovarian cancer prevention and treatment. Antioxidants (Basel). 10:17182021. View Article : Google Scholar : PubMed/NCBI

39 

Poyton MF, Feng XA, Ranjan A, Lei Q, Wang F, Zarb JS, Louder RK, Park G, Jo MH, Ye J, et al: Coordinated DNA and histone dynamics drive accurate histone H2A.Z exchange. Sci Adv. 8:eabj55092022. View Article : Google Scholar : PubMed/NCBI

40 

Oberdoerffer P and Miller KM: Histone H2A variants: Diversifying chromatin to ensure genome integrity. Semin Cell Dev Biol. 135:59–72. 2023. View Article : Google Scholar : PubMed/NCBI

41 

Geng X, Zhao J, Huang J, Li S, Chu W, Wang WS, Chen ZJ and Du Y: lnc-MAP3K13-7:1 inhibits ovarian GC proliferation in PCOS via DNMT1 Downregulation-mediated CDKN1A promoter hypomethylation. Mol Ther. 29:1279–1293. 2021. View Article : Google Scholar : PubMed/NCBI

42 

Wu Q, Xiang S, Ma J, Hui P, Wang T, Meng W, Shi M and Wang Y: Long non-coding RNA CASC15 regulates gastric cancer cell proliferation, migration and epithelial mesenchymal transition by targeting CDKN1A and ZEB1. Mol Oncol. 12:799–813. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Hu CC, Liang YW, Hu JL, Liu LF, Liang JW and Wang R: LncRNA RUSC1-AS1 promotes the proliferation of breast cancer cells by epigenetic silence of KLF2 and CDKN1A. Eur Rev Med Pharmacol Sci. 23:6602–6611. 2019.PubMed/NCBI

44 

Guo M and Zhang X: LncRNA MSTO2P promotes colorectal cancer progression through epigenetically silencing CDKN1A mediated by EZH2. World J Surg Oncol. 20:952022. View Article : Google Scholar : PubMed/NCBI

45 

Ma JX, Yang YL, He XY, Pan XM, Wang Z and Qian YW: Long noncoding RNA MNX1-AS1 overexpression promotes the invasion and metastasis of gastric cancer through repressing CDKN1A. Eur Rev Med Pharmacol Sci. 23:4756–4762. 2019.PubMed/NCBI

46 

Bi M, Yu H, Huang B and Tang C: Long non-coding RNA PCAT-1 over-expression promotes proliferation and metastasis in gastric cancer cells through regulating CDKN1A. Gene. 626:337–343. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Xu C, Chen C, Xu Y, Li Z, Chen H and Wang G: Prognostic significance of CDK1 in Ovarian and Cervical Cancers. J Cancer. 16:1656–1667. 2025. View Article : Google Scholar : PubMed/NCBI

48 

Pan Y, Zhou J, Zhang W, Yan L, Lu M, Dai Y, Zhou H, Zhang S and Yang J: The Sonic Hedgehog signaling pathway regulates autophagy and migration in ovarian cancer. Cancer Med. 10:4510–4521. 2021. View Article : Google Scholar : PubMed/NCBI

49 

Manousakis E, Miralles CM, Esquerda MG and Wright RHG: CDKN1A/p21 in breast cancer: Part of the problem, or part of the solution? Int J Mol Sci. 24:174882023. View Article : Google Scholar : PubMed/NCBI

50 

Zhao S, Bellone S, Lopez S, Thakral D, Schwab C, English DP, Black J, Cocco E, Choi J, Zammataro L, et al: Mutational landscape of uterine and ovarian carcinosarcomas implicates histone genes in epithelial-mesenchymal transition. Proc Natl Acad Sci USA. 113:12238–12243. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Saravi S, Katsuta E, Jeyaneethi J, Amin HA, Kaspar M, Takabe K, Pados G, Drenos F, Hall M and Karteris E: H2A histone family member X (H2AX) is upregulated in ovarian cancer and demonstrates utility as a prognostic biomarker in terms of overall survival. J Clin Med. 9:28442020. View Article : Google Scholar : PubMed/NCBI

52 

Yuan Y, Cao W, Zhou H, Qian H and Wang H: H2A.Z acetylation by lincZNF337-AS1 via KAT5 implicated in the transcriptional misregulation in cancer signaling pathway in hepatocellular carcinoma. Cell Death Dis. 12:6092021. View Article : Google Scholar : PubMed/NCBI

53 

Liu A, Zhou L, Huang Y and Peng D: Analysis of copy number variants detected by sequencing in spontaneous abortion. Mol Cytogenet. 17:132024. View Article : Google Scholar : PubMed/NCBI

54 

Ibeagha-Awemu EM, Bissonnette N, Bhattarai S, Wang M, Dudemaine PL, McKay S and Zhao X: Whole genome methylation analysis reveals role of DNA methylation in Cow's ileal and ileal lymph node responses to mycobacterium avium subsp. paratuberculosis infection. Front Genet. 12:7974902021. View Article : Google Scholar : PubMed/NCBI

55 

De Los Santos JA, Andrade JPN, Cangiano LR, Iriarte A, Penagaricano F and Parrish JJ: Transcriptomic analysis reveals gene expression changes in peripheral white blood cells of cows after embryo transfer: Implications for pregnancy tolerance. Reprod Domest Anim. 58:946–954. 2023. View Article : Google Scholar : PubMed/NCBI

56 

Lei K, Qu L, Liu F, Hao N, Chen J, Liu J and Lin A: Long non-coding RNAs regulate fatty acid and cholesterol metabolism. Genome Instability Dis. 3:70–82. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Braga EA, Filippova EA, Uroshlev LA, Lukina SS, Pronina IV, Kazubskaya TP, Kushlinskiy DN, Loginov VI, Fridman MV, Burdennyy AM and Kushlinskii NE: LncRNA genes of the SNHG family: Co-methylation and common functions in ovarian cancer. Biochemistry (Mosc). 89:2051–2068. 2024. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Zhu W, Feng J, Zhang Y, Zhou Q, Huang H and Yang Y: Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation. Oncol Lett 30: 531, 2025.
APA
Zhu, W., Feng, J., Zhang, Y., Zhou, Q., Huang, H., & Yang, Y. (2025). Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation. Oncology Letters, 30, 531. https://doi.org/10.3892/ol.2025.15277
MLA
Zhu, W., Feng, J., Zhang, Y., Zhou, Q., Huang, H., Yang, Y."Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation". Oncology Letters 30.5 (2025): 531.
Chicago
Zhu, W., Feng, J., Zhang, Y., Zhou, Q., Huang, H., Yang, Y."Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation". Oncology Letters 30, no. 5 (2025): 531. https://doi.org/10.3892/ol.2025.15277
Copy and paste a formatted citation
x
Spandidos Publications style
Zhu W, Feng J, Zhang Y, Zhou Q, Huang H and Yang Y: Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation. Oncol Lett 30: 531, 2025.
APA
Zhu, W., Feng, J., Zhang, Y., Zhou, Q., Huang, H., & Yang, Y. (2025). Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation. Oncology Letters, 30, 531. https://doi.org/10.3892/ol.2025.15277
MLA
Zhu, W., Feng, J., Zhang, Y., Zhou, Q., Huang, H., Yang, Y."Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation". Oncology Letters 30.5 (2025): 531.
Chicago
Zhu, W., Feng, J., Zhang, Y., Zhou, Q., Huang, H., Yang, Y."Role of long non‑coding RNA‑mRNA interactions in resveratrol‑mediated inhibition of ovarian cancer cell proliferation". Oncology Letters 30, no. 5 (2025): 531. https://doi.org/10.3892/ol.2025.15277
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