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

YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner

  • Authors:
    • Li Ren
    • Junjun Zhang
    • Jinjin Yang
    • Li Ji
    • Jingjing Guo
  • View Affiliations / Copyright

    Affiliations: Department of Gynecology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China, Department of Gynecology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
    Copyright: © Ren et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 498
    |
    Published online on: August 27, 2025
       https://doi.org/10.3892/ol.2025.15244
  • 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

Chronic infection with high‑risk human papillomavirus (HPV) types increases the risk of developing cervical cancer (CC). Notably, these HPV types are implicated in ~70% of all CC cases. YTH N6‑methyladenosine RNA‑binding protein C2 (YTHDC2) is an N6‑methyladenosine reader associated with several cancers, although its specific function in CC remains poorly understood. The present study aimed to elucidate new functions of YTHDC2 in HPV‑positive CC cell proliferation and ferroptosis, mediated by pathways dependent on protein synthesis. A comparative analysis revealed reduced levels of YTHDC2 in HPV‑positive CC samples and cell lines compared with those in HPV‑negative samples. YTHDC2 overexpression diminished cell viability and proliferation, whereas silencing YTHDC2 enhanced these processes, according to Cell Counting Kit‑8 and colony formation assays. Cells overexpressing YTHDC2 exhibited characteristics of ferroptosis, such as apoptosis, reactive oxygen species production and imbalances in several ferroptosis indicators, including solute carrier family 7 member 11 (SLC7A11). A bioluminescent assay using a firefly luciferase construct fused with the SLC7A11 5'‑untranslated region (UTR) demonstrated a reduction in luminescence in cells exhibiting enhanced YTHDC2 expression. This indicated that YTHDC2 catalyzes translation initiation via deconvolution of the 5'‑UTR of SLC7A11 mRNA. Alterations to the 5'‑UTR led to increased luciferase activity. Furthermore, increased SLC7A11 levels mitigated the effects of YTHDC2 overexpression on proliferation and ferroptosis in CC cells. In conclusion, the results of the present study suggest that low expression of the RNA helicase YTHDC2 contributes to CC proliferation and inhibition of ferroptosis by promoting SLC7A11 translation in HPV‑positive CC cells.

Introduction

Cervical neoplasia is the second most frequently diagnosed malignancy in women worldwide and remains the foremost cancer type in several low-income countries, imposing a notable socio-economic burden (1). The International Agency for Research on Cancer estimates that ~300,000 annual fatalities are attributed to cervical cancer (CC) in China (2,3). Human papillomavirus (HPV)-18 is an important strain responsible for initiating precancerous conditions that frequently progress to cervical malignancies, accounting for >90% of such cases globally. The occurrence of HPV-18 infections among women is escalating worldwide; however, most of these infections do not develop into cancer (4,5). Consequently, it has become essential to explore supplementary factors beyond mere infection that contribute to the onset of carcinogenesis and CC progression.

YTH domain-containing protein 2 (YTHDC2) forms part of the YTH protein unit and is instrumental in orchestrating an array of mRNA metabolic functions, encompassing splicing, export, dismantling and translation (6–8). Members of the YTH protein group selectively interact with mRNAs marked by N6-methyladenosine (m6A), a reversible and substantial post-transcriptional alteration regulated by the activity of methyltransferases and demethylases (9). In addition to its YTH domain, YTHDC2 has an RNA helicase domain, unique within this protein family that is essential for efficiently translating specific mRNAs mediated by YTHDC2 (10).

During tumorigenesis, YTHDC2 is pivotal for synthesizing proteins that result in malignant traits. Moreover, it suppresses colorectal cancer progression via its regulatory effects on hypoxia-inducible factor 1α (HIF-1α) synthesis (11). YTHDC2 expression is also increased in radiation-sensitive nasopharyngeal carcinoma, promoting radioresistance by activating the insulin-like growth factor 1/AKT/S6 signaling cascade (12). Furthermore, YTHDC2 expression is markedly increased in cancers such as prostate cancer and glioblastoma (13–16), but is diminished in pulmonary malignancies, as well as head and neck squamous cell carcinomas (17,18). However, the precise biological roles of YTHDC2 in CC remain poorly defined, particularly in HPV-positive cases.

The present study aimed to assess the functional effects of YTHDC2 on cervical carcinogenesis. The findings aimed to demonstrate the association between YTHDC2 expression and HPV prevalence across cervical neoplastic tissues and in vitro cell models. The effect of YTHDC2 expression on cellular proliferation and facilitated ferroptosis in CC was also studied.

Materials and methods

Human tissue samples

The present study was approved by the Ethics Committee of the First Affiliated Hospital of Henan University of Science and Technology (Luoyang, China; approval no. 2023-0147). Cervical tissue specimens were collected between July 2021 and September 2023 from 25 women (age, 35.5–60.1 years) histologically diagnosed with stage IA CC featuring lymphovascular space involvement, or classified as IA2, as detailed in our previous research (19,20). The selection criteria for patients with cervical cancer were as follows: i) Pathologically confirmed patients with cervical cancer; and (2) the patients had no history of other cancers. No patients had preoperative chemotherapy, radiotherapy, or other treatment history or other inflammatory diseases. Patient conditions were staged according to the criteria of the International Federation of Gynecology and Obstetrics (21). Additionally, samples were collected from women with high-risk (HR)-HPV-negative typical CC (n=25) and HR-HPV-positive cervical malignancies (n=25) who had undergone HR-HPV screening and ThinPrep® cytology (Hologic, Inc.) at the First Affiliated Hospital of Henan University of Science and Technology. Written informed consent was obtained from every participant involved.

Immunohistochemistry (IHC)

IHC was performed on cervical cancer tissue sections prepared as formalin-fixed paraffin-embedded samples. The tissues were fixed in 10% neutral-buffered formalin at room temperature for 24 h, embedded in paraffin wax, and sectioned at a 4-µm thickness. Fresh frozen tissues were snap-frozen in liquid nitrogen-cooled isopentane at approximately −150°C and sectioned at 7 µm. The sections were permeabilized with 0.2% Triton X-100 (Sigma-Aldrich; Merck KGaA) for 10 min at room temperature, followed by blocking with 5% normal goat serum (cat. no. 31873; Thermo Fisher Scientific, Inc.) in PBS for 1 h at room temperature. Primary antibody incubation was performed using anti-YTHDC2 (cat. no. ab220160; Abcam) at a 1:100 dilution overnight at 4°C. After washing, sections were incubated with HRP-conjugated goat anti-rabbit IgG secondary antibody (cat. no. 7074; Cell Signaling Technology) at a 1:500 dilution for 1 h at room temperature. Signal detection was performed using 3,3′-diaminobenzidine chromogen (cat. no. K3468; Dako; Agilent Technologies, Inc.). Slides were counterstained with hematoxylin and examined using a bright-field microscope (Olympus BX53; Olympus Corporation).

Cell culture

Cell lines (purchased from ATCC), including HFF-1 (HPV-negative human epithelial foreskin fibroblasts), C33A and DoTc2 4510 (both from HPV-negative cervical carcinomas), SiHa and CaSKi (from HPV16-positive cervical squamous carcinomas), SW756 (from an HPV18-positive squamous carcinoma), HeLa (from an HPV18-positive cervical epithelial adenocarcinoma) and H8 (from HPV-positive, immortalized cervical cells), were maintained in RPMI-1640 medium (cat. no. 11875093; Gibco; Thermo Fisher Scientific, Inc.) with 10% fetal bovine serum (cat. no. A5256701; HyClone; Cytiva). All cell lines were incubated at 37°C in a 5% CO2 atmosphere with humidity.

Transfection procedures

SiHa and CaSKi cells (5×106/well) were transfected with 1.5 µg YTHDC2-overexpression vectors (pCMV-YTHDC2) and/or 1.5 µg control vectors (pCMV-empty), and with 2 µg YTHDC2-specific small interfering (si)RNAs (#1, 5′-ATATAAGAGATGTGACGAGGG-3′; #2, 5′-CTTTAGTCGAAGTTCTGACTA-3′; and #3, 5′-GGAAGCTAAATCGAGCCTT-3′), and control siRNAs (5′-CACAGGGUAAGGAACUCGUCUCUCA-3′) using Lipofectamine™ 2000 (cat. no. 11668027; Invitrogen; Thermo Fisher Scientific, Inc.) at room temperature for 36 h according to the manufacturer's protocols. The vector and siRNA were constructed and purchased from Genscript Biotech Corporation. The effect of overexpression or knockdown was evaluated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), as described below, at 48 h post-transfection Subsequently, siRNA #1 was randomly selected for further experiments as the knockdown efficacies of all three siRNAs were similar (Fig. S1). SiHa and CaSKi cells were co-transfected with pCMV1-YTHDC2 along with either pcDNA3.1-empty or pcDNA3.1-SLC7A11 for 48 h.

Additionally, SiHa and CaSKi cells were transfected with 2 µg SLC7A11-overexpression vector pcDNA3-SLC7A11 or 2 µg SLC7A11-specific siRNA (5′-CTGGAGTTATGCAGCTAAT-3′), or co-transfected with pCMV1-YTHDC2 along with either pcDNA3.1-empty or pcDNA3.1-SLC7A11, using Lipofectamine 2000 (Thermo Fisher Scientific, Inc.) at room temperature for 36 h according to the manufacturer's instructions, to upregulate or downregulate SLC7A11 expression, respectively. Concentrations of 10 nM for vectors and 20 nM for siRNAs were used for the transfections. Subsequent experiments were performed 48 h after transfection.

Cell viability assay

Cell viability was assessed using Cell Counting Kit-8 (CCK-8) reagent (cat. no. CK04; Dojindo Laboratories, Inc.) at 48 h following transfection, according to the manufacturer's guidelines. A total of 10 µl CCK-8 solution was added to each designated well (5×106/well). Subsequently, the plate was incubated for 2 h and absorbance readings were then taken at 450 nm using the Tecan Infinite M200 microplate (Tecan Group, Inc.).

Colony formation assay

SiHa and CaSKi cells were cultured in a series of 12-well plates, with each well containing 3,000 cells/ml. These cells were cultured for 7–9 days at 37°C, after which they were fixed using 10% neutral-buffered formalin for ≥4 h at room temperature, stained using crystal violet for 30 min at room temperature (cat. no. C0121; Beyotime Institute of Biotechnology) and visually assessed using an advanced optical system, and counted manually (Olympus CX23; Olympus Corporation). A colony was typically defined as: i) A group containing at least 50 cells; ii) a group clearly separated from neighboring colonies; and iii) a group of cells that must be stained.

Flow cytometry

Following transfection, the cell specimens were incubated in 6-well plates for 48 h and then trypsinized and preserved in 75% ethanol at −20°C overnight. Following stabilization, these specimens were treated with 0.5 µg/ml RNase A (Thermo Fisher Scientific, Inc.) and 100 µg/ml propidium iodide (PI) for 30 min. Subsequently, apoptotic indices were evaluated using a dual staining Annexin V-FITC/PI apoptosis kit (Abcam) with analytical procedures performed using a BD FACScan flow cytometer (BD Biosciences) equipped with BD CellQuest™ software (version 5.1; BD Biosciences). Specimens unreactive to FITC/PI were utilized as negative controls in these assays.

Reactive oxygen species (ROS) measurements

A total of 5×104 SiHa and CaSKi cells were added to the wells of a 96-well white plate optimized for luminescence studies. Following cell adhesion, cells were cultured for 24 h. After thoroughly rinsing with PBS (cat. no. 10010023; Thermo Fisher Scientific, Inc.), a solution of 20 µM carboxy-H2-DCFDA (cat. no. C400; Invitrogen; Thermo Fisher Scientific, Inc.) was added to the cellular milieu and incubated at 37°C for 1 h to facilitate ROS-associated fluorescence development. Fluorescence detection was performed using the 1420 Multi-label Counter (PerkinElmer, Inc.). Furthermore, cells were resuspended in 1 ml PBS and incubated for 1 h under identical temperature conditions after applying a second dose of 20 µM carboxy-H2-DCFDA. The cells were rinsed with PBS to remove extraneous dye, and the fluorescence intensity was quantified using a microplate reader at 485 and 535 nm excitation and emission wavelengths, respectively, to determine the ROS levels within the samples accurately.

RT-qPCR

RNA isolation from SiHa and CaSKi cell or tissue samples (10 mg) was performed using TRIzol™ Reagent (cat. no. 15596018CN; Invitrogen; Thermo Fisher Scientific, Inc.), with the quantitative assessment of yields performed using a NanoDrop™ 2000 Spectrophotometer (Thermo Fisher Scientific, Inc.) at an optical density of 260 nm. Subsequent reverse transcription to generate cDNA was performed using the SuperScript™ IV First-Strand Synthesis System (cat. no. 18091050; Invitrogen; Thermo Fisher Scientific, Inc.) with oligo(dT) 20 primers. The thermal conditions for reverse transcription PCR were as follows: 37°C for 2 min, and 55°C for 5 min. For the quantitative analysis, the prepared cDNA was subjected to amplification using SYBR™ Select Master Mix (cat. no. 4472918; Invitrogen; Thermo Fisher Scientific, Inc.), adhering to the protocols specified by the supplier. The internal standard GAPDH mRNA was used to calibrate the reaction, which was initiated with a 10 min denaturation step at 95°C, followed by 40 cycles of 15 sec denaturation at the same temperature, and a 40 sec extension phase at 60°C. The abundance of the target mRNA was quantitatively evaluated using the comparative 2−ΔΔCq method (22). Each experimental sequence was performed in triplicate to ensure the reproducibility and reliability of the results. The sequences for primers used in this experiment were as follows: YTHDC2-forward (F), 5′-CCAGGCCGAGCAGCGTCTCC-3′; YTHDC2-reverse (R), 5′-ACAGTTAATCAGTATGGGAGCC-3′; GAPDH-F, 5′-AACAGCGACACCCACTCCTC-3′; GAPDH-R, 5′-CATACCAGGAAATGAGCTTGACAA-3′; SLC7A11-F, 5′-TCCTGCTTTGGCTCCATGAACG-3′; and SLC7A11-R, 5′-AGAGGAGTGTGCTTGCGGACAT-3′. The thermocycling conditions for qPCR were as follows: 95°C for 2 min, 95°C for 10 sec, 55°C for 30 sec and 72°C for 30 sec, for 40 cycles.

Western blotting (WB)

SiHa and CaSKi cells were disrupted in a lysis buffer comprising HEPES (0.02 M), NaCl (0.15 M), EDTA (1 mM, pH 7.4), glycerol (10%), Triton X-100 (1%), a comprehensive protease inhibitor mixture and Na3VO4 (5 mM). The resulting lysate was incubated at 4°C for 1 h, followed by centrifugation at 12,000 × g at 4°C for 15 min. After separation via 5–12% sodium dodecyl sulphate-polyacrylamide gel electrophoresis, proteins (10 µg/lane; determined by BCA method) were transferred onto polyvinylidene difluoride membranes. The membranes were then blocked for 2 h at 4°C in PBS supplemented with 5% skim milk powder (cat. no. LP0033B; Invitrogen; Thermo Fisher Scientific, Inc.) and 0.5% Tween 20. Subsequently, proteins were probed with appropriate primary antibodies, including anti-YTHDC2 (1:1,000; cat. no. ab220160; Abcam), anti-actin (1:5,000; cat. no. ab8227; Abcam), anti-SLC7A11 (1:2,000; cat. no. ab37185; Abcam), anti-p53 (1:2,000; cat. no. ab32389; Abcam), anti-ACSL4 (1:1,000; cat. no. ab155282; Abcam) and anti-GPX4 (1:1,000; cat. no. ab41787; Abcam) antibodies for 1 h at room temperature, and detected using horseradish peroxidase-conjugated secondary antibodies derived from rabbits or mice, including goat anti-mouse HRP antibody (1:10,000; cat. no. ab6708; Abcam) and goat anti-rabbit antibody (1:10,000; cat. no. ab6721; Abcam) for another 1 h at room temperature. Finally, protein expression was quantified using Image-Pro Plus software (version 6.0; Media Cybernetics, Inc.).

Dual-luciferase reporter assay

Fragments of SLC7A11 wild-type (−WT) or SLC7A11 mutant (−Mut) (where m6A was substituted with C) were inserted into the pRP (Exp)-Puro-EF1A-Luciferase (mSLC7A11_3′UTR_1862-4861bp: SNP) (VectorBuilder) plasmid to create pcDNA3-SLC7A11-WT and pcDNA3-SLC7A11-Mut plasmids as aforementioned. At 72 h post-transfection using Lipofectamine 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions, luciferase activity was measured using the Dual-Luciferase Reporter Assay System (Promega Corporation). Relative firefly luciferase (Fluc)/Renilla luciferase (Rluc) activity was calculated by normalizing the activity of firefly luciferase to that of Renilla luciferase. Each experiment was conducted in triplicate for each group.

Statistical analysis

Data were statistically analyzed using SPSS version 18.0 software (IBM Corp.). Data are expressed as the mean ± standard deviation. Comparisons between two groups were assessed using an unpaired t-test, whereas comparisons between multiple groups were analyzed using one-way analysis of variance with Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

YTHDC2 levels are increased in HPV-positive CC specimens and cell lines

CC specimens were first classified as HPV- positive or -negative. YTHDC2 mRNA levels in HPV-positive CC specimens were significantly lower than those in their HPV-negative counterparts, according to RT-qPCR, WB and immunohistochemistry results (Fig. 1A-C). To further evaluate these observations based on clinical specimens, YTHDC2 expression in HPV-negative (HFF-1, C33A and DoTc2 4510) and HPV-positive CC cell lines (SiHa, CaSKi, SW756, HeLa and H8) were assessed. At the cellular level, the significant downregulation of YTHDC2 mRNA and protein expression was detected in HPV-positive CC cells compared with levels in HPV-negative CC cells (Fig. 1D and E). These data indicate that HPV-positive CC specimens and cell lines consistently have lower YTHDC2 levels.

Analysis of YTHDC2 expression in
clinical and cell samples of HPV-positive cervical cancer. (A)
RT-qPCR results demonstrating YTHDC2 expression in tissue samples
from HPV-positive (n=25) and HPV-negative (n=25) cervical cancer.
(B) Western blot analysis of the expression of YTHDC2 in tissue
samples from HPV-positive (n=3) and HPV-negative (n=3) cervical
cancer from the aforementioned 25 HPV-positive tissues and 25
HPV-negative CC samples. (C) Immunohistochemistry assessment of the
expression of YTHDC2 in the tissue samples from HPV-positive and
HPV-negative cervical cancer. Magnification, ×40. Analysis of
YTHDC2 mRNA and protein levels in cervical cancer cell lines
(CaSKi, HFF-1, SiHa, SW756, HeLa, DoTc2 4510, C33A and H8) using
(D) RT-qPCR and (E) western blotting, respectively. *P<0.05;
**P<0.01. YTHDC2, YTH N6-methyladenosine RNA-binding protein C2;
HPV, human papillomavirus; RT-qPCR, reverse
transcription-quantitative PCR.

Figure 1.

Analysis of YTHDC2 expression in clinical and cell samples of HPV-positive cervical cancer. (A) RT-qPCR results demonstrating YTHDC2 expression in tissue samples from HPV-positive (n=25) and HPV-negative (n=25) cervical cancer. (B) Western blot analysis of the expression of YTHDC2 in tissue samples from HPV-positive (n=3) and HPV-negative (n=3) cervical cancer from the aforementioned 25 HPV-positive tissues and 25 HPV-negative CC samples. (C) Immunohistochemistry assessment of the expression of YTHDC2 in the tissue samples from HPV-positive and HPV-negative cervical cancer. Magnification, ×40. Analysis of YTHDC2 mRNA and protein levels in cervical cancer cell lines (CaSKi, HFF-1, SiHa, SW756, HeLa, DoTc2 4510, C33A and H8) using (D) RT-qPCR and (E) western blotting, respectively. *P<0.05; **P<0.01. YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; HPV, human papillomavirus; RT-qPCR, reverse transcription-quantitative PCR.

YTHDC2 silencing and overexpression in HPV-positive CC cells (SiHa and CaSKi)

YTHDC2 expression was modulated in SiHa and CaSKi cells through overexpression and silencing techniques to further evaluate the function of YTHDC2 in HPV-positive CC cells. After transfection with the overexpression vector, both RT-qPCR and WB analyses revealed a significant increase in YTHDC2 mRNA and protein levels compared with those in the control group. Conversely, silencing YTHDC2 expression via siRNA transfection significantly diminished its mRNA and protein levels in both SiHa and CaSKi cells, compared with those in the control group (Fig. 2).

Modulation of YTHDC2 expression in
SiHa and CaSKi cells. SiHa and CaSKi cells were either untreated or
transfected with pCMV1-empty, pCMV1-YTHDC2, control siRNA or YTHDC2
siRNA for 48 h. Reverse transcription-quantitative PCR of (A) SiHA
and (B) CaSKi cells, and western blotting analyses of (C) SiHa and
(D) CaSKi cells, demonstrating the changes in YTHDC2 expression
levels. **P<0.01. YTHDC2, YTH N6-methyladenosine RNA-binding
protein C2; si, small interfering; OE, overexpression; NTC,
non-transfected control.

Figure 2.

Modulation of YTHDC2 expression in SiHa and CaSKi cells. SiHa and CaSKi cells were either untreated or transfected with pCMV1-empty, pCMV1-YTHDC2, control siRNA or YTHDC2 siRNA for 48 h. Reverse transcription-quantitative PCR of (A) SiHA and (B) CaSKi cells, and western blotting analyses of (C) SiHa and (D) CaSKi cells, demonstrating the changes in YTHDC2 expression levels. **P<0.01. YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; si, small interfering; OE, overexpression; NTC, non-transfected control.

Upregulated YTHDC2 expression impedes cellular proliferation and induces ferroptosis in SiHa and CaSKi cells

YTHDC2 overexpression led to a significant decrease in cell viability in both cell lines at 48 h post-treatment, compared with that in the control group. Meanwhile, YTHDC2 silencing significantly increased cell viability, compared with that in the control group, according to CCK-8 assays (Fig. 3A and B). The results of the colony formation assays indicated that the cell proliferation rate was significantly decreased after YTHDC2 overexpression and significantly elevated after YTHDC2 knockdown, compared with that in the controls groups (Fig. 3C and D). These data suggest that YTHDC2 overexpression attenuates HPV-positive cell proliferation.

Effect of YTHDC2 on cell viability
and proliferation in SiHa and CaSKi cells. SiHa and CaSKi cells
were subjected to either no treatment or were transfected with
pCMV1-empty, pCMV1-YTHDC2, control siRNA or YTHDC2 siRNA for 48 h.
Post-transfection cell viability of (A) SiHA and (B) CaSKi cells
was assessed at 48 h using CCK-8 assays. Colony formation assays
were performed to evaluate the proliferation rates of (C) SiHa and
(D) CaSKi cells. *P<0.05; **P<0.01. YTHDC2, YTH
N6-methyladenosine RNA-binding protein C2; si, small interfering;
OE, overexpression; NTC, non-transfected control.

Figure 3.

Effect of YTHDC2 on cell viability and proliferation in SiHa and CaSKi cells. SiHa and CaSKi cells were subjected to either no treatment or were transfected with pCMV1-empty, pCMV1-YTHDC2, control siRNA or YTHDC2 siRNA for 48 h. Post-transfection cell viability of (A) SiHA and (B) CaSKi cells was assessed at 48 h using CCK-8 assays. Colony formation assays were performed to evaluate the proliferation rates of (C) SiHa and (D) CaSKi cells. *P<0.05; **P<0.01. YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; si, small interfering; OE, overexpression; NTC, non-transfected control.

Moreover, flow cytometry was used to assess the role of YTHDC2 in cell apoptosis, especially ferroptosis in SiHa and CaSKi cells. The numbers of apoptotic cells for both cell lines overexpressing YTHDC2 were significantly elevated, whereas cells with YTHDC2 knockdown exhibited a significantly decreased cell apoptosis rate, compared with that of controls (Fig. 4A and B). Ferroptosis is a newly identified type of apoptosis, and excessive ROS production is a predominant marker of this process (23). Therefore, the present study assessed the ROS levels in cell lines after overexpressing or silencing YTHDC2. Compared with that of controls, increased ROS levels were observed in cells overexpressing YTHDC2, whereas they were significantly reduced in cells with silenced YTHDC2 expression (Fig. 4C and D). Furthermore, the protein expression of the ferroptosis markers SLC7A11, p53, acyl-CoA synthetase long chain family member 4 (ACSL4) and glutathione peroxidase 4 (GPX4) in cells with varying YTHDC2 expression levels were assessed. WB analyses demonstrated that GPX4 and ACSL4 levels were negatively associated with the expression of YTHDC2 in SiHa and CaSKi cells, whereas SLC7A11 and p53 were positively associated with YTHDC2 levels in these cells (Fig. 4E and F). These findings indicate that YTHDC2 promotes ferroptosis in SiHa and CaSKi cells.

Effect of YTHDC2 on ferroptosis in
SiHa and CaSKi cells. SiHa and CaSKi cells were either untreated or
transfected with pCMV1-empty, pCMV1-YTHDC2, control siRNA or YTHDC2
siRNA for 48 h. Flow cytometry was performed to assess the
apoptosis rates in (A) SiHA and (B) CaSKi cells. ROS levels were
assessed after a 10 min exposure to the ROS-sensitive fluorescent
dye H2DCF-DA (5 µM) in (C) SiHa and (D) CaSKi cells. Western
blotting was performed to assess the levels of proteins involved in
ferroptosis, including p53, ACSL4, GPX4 and SLC7A11, in (E) SiHa
and (F) CaSKi cells. *P<0.05; **P<0.01. YTHDC2, YTH
N6-methyladenosine RNA-binding protein C2; si, small interfering;
OE, overexpression; NC, negative control; ROS, reactive oxygen
species; ACSL4, acyl-CoA synthetase long chain family member 4;
GPX4, glutathione peroxidase 4; SLC7A11, solute carrier family 7
member 11; NTC, non-transfected control.

Figure 4.

Effect of YTHDC2 on ferroptosis in SiHa and CaSKi cells. SiHa and CaSKi cells were either untreated or transfected with pCMV1-empty, pCMV1-YTHDC2, control siRNA or YTHDC2 siRNA for 48 h. Flow cytometry was performed to assess the apoptosis rates in (A) SiHA and (B) CaSKi cells. ROS levels were assessed after a 10 min exposure to the ROS-sensitive fluorescent dye H2DCF-DA (5 µM) in (C) SiHa and (D) CaSKi cells. Western blotting was performed to assess the levels of proteins involved in ferroptosis, including p53, ACSL4, GPX4 and SLC7A11, in (E) SiHa and (F) CaSKi cells. *P<0.05; **P<0.01. YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; si, small interfering; OE, overexpression; NC, negative control; ROS, reactive oxygen species; ACSL4, acyl-CoA synthetase long chain family member 4; GPX4, glutathione peroxidase 4; SLC7A11, solute carrier family 7 member 11; NTC, non-transfected control.

SLC7A11 expression is regulated by YTHDC2 through m6A methylation

Based on previous publications (24,25), we hypothesized that SLC7A11 could be modulated by YTHDC2 (an RNA m6A reader) in an m6A-dependent manner. Therefore, the present study aimed to assess this hypothesis. The results revealed that, compared with controls, SLC7A11 mRNA expression was significantly downregulated after YTHDC2 overexpression and significantly upregulated after YTHDC2 silencing in both cell lines, according to RT-qPCR analysis (Fig. 5A and B). Subsequently, luciferase reporter assays were performed to assess SLC7A11 mRNA 5′-UTR methylation and gene expression. SiHa and CaSKi cells demonstrating either upregulated or downregulated YTHDC2 expression were transfected with reporter plasmids that included the full-length SLC7A11 5′-UTR immediately upstream of the luciferase gene and incubated for 36 h. Compared with in controls, luciferase activity was significantly diminished in cells with elevated YTHDC2 levels, whereas cells with suppressed YTHDC2 expression exhibited a significant increase in luciferase activity (Fig. 5C and D). Subsequently, the G residue within the SLC7A11 5′-UTR consensus sequence was mutated (5′-GGCUGC-3′ to 5′-AACUAC- 3′ in mRNA), and wild-type and mutant reporter-driven luciferase activities were compared using luciferase assays. The A residue mutation reduced the luciferase activity by ~70% in SiHa and CaSKi cells (Fig. 5E and F). Collectively, these data suggest that YTHDC2 contributes to SLC7A11 mRNA 5′-UTR m6A methylation, thereby inhibiting SLC7A11 expression and protein translation.

Role of YTHDC2 in N6-methyladenosine
methylation and suppression of SLC7A11 mRNA expression. CaSKi cells
were either untreated or transfected with pCMV1-empty,
pCMV1-YTHDC2, control siRNA or YTHDC2 siRNA for 48 h. Reverse
transcription-quantitative PCR of (A) SiHA and (B) CaSKi cells
cells revealed the changes in SLC7A11 expression levels. Luciferase
assays were performed on (C) SiHa and (D) CaSKi cells transfected
with either YTHDC2 overexpression or silencing vectors alongside a
reporter plasmid, including the SLC7A11 5′-UTR associated with
luciferase sequences. Luciferase activity ratios corresponding with
the SLC7A11 5′-UTR were normalized to controls. Additional
luciferase assays involved (E) SiHa and (F) CaSKi cells transfected
with either WT or mutant SLC7A11 5′-UTR luciferase reporters.
*P<0.05; **P<0.01; ***P<0.001. YTHDC2, YTH
N6-methyladenosine RNA-binding protein C2; SLC7A11, solute carrier
family 7 member 11; UTR, untranslated region; si, small
interfering; OE, overexpression; NC, negative control; WT,
wild-type; NTC, non-transfected control.

Figure 5.

Role of YTHDC2 in N6-methyladenosine methylation and suppression of SLC7A11 mRNA expression. CaSKi cells were either untreated or transfected with pCMV1-empty, pCMV1-YTHDC2, control siRNA or YTHDC2 siRNA for 48 h. Reverse transcription-quantitative PCR of (A) SiHA and (B) CaSKi cells cells revealed the changes in SLC7A11 expression levels. Luciferase assays were performed on (C) SiHa and (D) CaSKi cells transfected with either YTHDC2 overexpression or silencing vectors alongside a reporter plasmid, including the SLC7A11 5′-UTR associated with luciferase sequences. Luciferase activity ratios corresponding with the SLC7A11 5′-UTR were normalized to controls. Additional luciferase assays involved (E) SiHa and (F) CaSKi cells transfected with either WT or mutant SLC7A11 5′-UTR luciferase reporters. *P<0.05; **P<0.01; ***P<0.001. YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; SLC7A11, solute carrier family 7 member 11; UTR, untranslated region; si, small interfering; OE, overexpression; NC, negative control; WT, wild-type; NTC, non-transfected control.

SLC7A11 overexpression counteracts YTHDC2-mediated SiHa and CaSKi cell proliferation and ferroptosis

Subsequently, the present study evaluated the function of SLC7A11 in YTHDC2-regulated cell proliferation and ferroptosis in SiHa and CaSKi cells by overexpressing it in these cell lines, both with and without YTHDC2 overexpression. Compared with the controls, SLC7A11 mRNA and protein levels were significantly upregulated in SiHa and CaSKi cells, independent of YTHDC2 overexpression (Fig. 6). Subsequently, proliferation and ferroptosis in SiHa and CaSKi cells was assessed following YTHDC2 overexpression and/or SLC7A11 overexpression. Compared with the controls, increased SLC7A11 expression was associated with a significant increase in cell viability and colony formation in SiHa and CaSKi cells (Fig. 7).

Augmentation of SLC7A11 expression in
YTHDC2-overexpressing SiHa and CaSKi cells. SiHa and CaSKi cells
were co-transfected with pCMV1-YTHDC2 along with either
pcDNA3.1-empty or pcDNA3.1-SLC7A11 for 48 h. Reverse
transcription-quantitative PCR of (A) SiHa and (B) CaSKi cells, and
western blot analysis of (C) SiHa and (D) CaSKi cells were
performed to assess SLC7A11 mRNA and protein levels, respectively,
demonstrating the effects of co-transfection on these cells.
**P<0.01. SLC7A11, solute carrier family 7 member 11; YTHDC2,
YTH N6-methyladenosine RNA-binding protein C2; OE, overexpression;
NC, negative control.

Figure 6.

Augmentation of SLC7A11 expression in YTHDC2-overexpressing SiHa and CaSKi cells. SiHa and CaSKi cells were co-transfected with pCMV1-YTHDC2 along with either pcDNA3.1-empty or pcDNA3.1-SLC7A11 for 48 h. Reverse transcription-quantitative PCR of (A) SiHa and (B) CaSKi cells, and western blot analysis of (C) SiHa and (D) CaSKi cells were performed to assess SLC7A11 mRNA and protein levels, respectively, demonstrating the effects of co-transfection on these cells. **P<0.01. SLC7A11, solute carrier family 7 member 11; YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; OE, overexpression; NC, negative control.

Mitigating effects of SLC7A11
overexpression on YTHDC2-induced changes in SiHa and CaSKi cell
viability. SiHa and CaSKi cells were subjected to co-transfection
with pCMV1-YTHDC2 and either pcDNA3.1-empty or pcDNA3.1-SLC7A11 for
48 h. (A) Cell viability was assessed at 48 h post-transfection
utilizing CCK-8 assays. (B) Colony formation assays were performed
to assess the proliferation rates of these cells. **P<0.01;
***P<0.001. SLC7A11, solute carrier family 7 member 11; YTHDC2,
YTH N6-methyladenosine RNA-binding protein C2; OE, overexpression;
NC, negative control.

Figure 7.

Mitigating effects of SLC7A11 overexpression on YTHDC2-induced changes in SiHa and CaSKi cell viability. SiHa and CaSKi cells were subjected to co-transfection with pCMV1-YTHDC2 and either pcDNA3.1-empty or pcDNA3.1-SLC7A11 for 48 h. (A) Cell viability was assessed at 48 h post-transfection utilizing CCK-8 assays. (B) Colony formation assays were performed to assess the proliferation rates of these cells. **P<0.01; ***P<0.001. SLC7A11, solute carrier family 7 member 11; YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; OE, overexpression; NC, negative control.

Finally, regarding ferroptosis, SLC7A11 overexpression was demonstrated to significantly reduce apoptosis in SiHa and CaSKi cells with silenced YTHDC2 expression, compared with controls (Fig. 8A and B). Moreover, the generation of ROS in cells overexpressing YTHDC2 with elevated SLC7A11 levels was significantly reduced compared with the control group (Fig. 8C and D). Meanwhile, SLC7A11 and p53 markedly promoted GPX expression, and ACSL levels were notably reduced by SLC7A11, according to WB analysis (Fig. 8E). These results suggest that SLC7A11 overexpression is associated with YTHDC2-mediated proliferation and ferroptosis in SiHa and CaSKi cells.

Effect of SLC7A11 on counteracting
YTHDC2-driven ferroptosis in SiHa and CaSKi cells. SiHa and CaSKi
cells were simultaneously transfected with pCMV1-YTHDC2 and either
pcDNA3.1-empty or pcDNA3.1-SLC7A11 for 48 h. Flow cytometry was
used to assess ferroptosis in (A) SiHa and (B) CaSKi cells.
Reactive oxygen species accumulation was assessed in (C) SiHa and
(D) CaSKi cells after treating the cells with the H2DCF-DA
fluorescent marker (5 µM) for 10 min. (E) GPX4, ACSL4, p53 and
SLC7A11 protein levels were analyzed using western blotting.
*P<0.05; **P<0.01. SLC7A11, solute carrier family 7 member
11; YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; ACSL4,
acyl-CoA synthetase long chain family member 4; GPX4, glutathione
peroxidase 4; OE, overexpression; NC, negative control.

Figure 8.

Effect of SLC7A11 on counteracting YTHDC2-driven ferroptosis in SiHa and CaSKi cells. SiHa and CaSKi cells were simultaneously transfected with pCMV1-YTHDC2 and either pcDNA3.1-empty or pcDNA3.1-SLC7A11 for 48 h. Flow cytometry was used to assess ferroptosis in (A) SiHa and (B) CaSKi cells. Reactive oxygen species accumulation was assessed in (C) SiHa and (D) CaSKi cells after treating the cells with the H2DCF-DA fluorescent marker (5 µM) for 10 min. (E) GPX4, ACSL4, p53 and SLC7A11 protein levels were analyzed using western blotting. *P<0.05; **P<0.01. SLC7A11, solute carrier family 7 member 11; YTHDC2, YTH N6-methyladenosine RNA-binding protein C2; ACSL4, acyl-CoA synthetase long chain family member 4; GPX4, glutathione peroxidase 4; OE, overexpression; NC, negative control.

Discussion

CC associated with HPV infection is a malignancy that is largely preventable through immunization strategies, with vaccines currently available and subject to ongoing refinement (26). Findings from the present study indicate the suppression of YTHDC2 expression in CC samples and cell cultures positive for HPV. The enhanced expression of YTHDC2 in HPV-positive CC cell lines (SiHa and CaSKi) curtailed cellular proliferation and increased ferroptosis in these cells, whereas YTHDC2 knockdown reversed these effects on proliferation and ferroptosis. Furthermore, SLC7A11 (a notable ferroptosis marker) was conversely controlled by YTHDC2 in an m6A modification-dependent manner. SLC7A11 overexpression in YTHDC2-overxpressing cells mitigated the effect of YTHDC2 on cell proliferation and the progression of ferroptosis. However, as the present analysis was limited to two cell lines, additional empirical studies using animal models are imperative to corroborate these findings.

Extensive research has demonstrated the role of m6A methylation in several pathologies, including acute myeloid leukemia and type 2 diabetes (27–30). Subsequent investigations have delved deeper into the functionality of m6A-associated proteins in CC. For example, methyltransferase-like 3 enhances the methylation-dependent stability of hexokinase 2 in CC cells, facilitating the Warburg effect and cellular proliferation (31). Furthermore, elevated levels of YTH N6-methyladenosine RNA binding protein F1 (YTHDF1), a protein from the YTH protein family, are associated with a worse clinical prognosis in CC. The attenuation of YTHDF1 expression markedly curtails the proliferation, migration and invasion of CC cells, whilst increasing apoptosis rates (32). In vivo experiments using nude mice have demonstrated the role of YTHDF1 in accelerating CC cell oncogenesis (32). Moreover, YTHDC2 was suggested to augment metastasis in colon cancer by promoting HIF-1α translation (11). Conversely, another study reported reduced YTHDC2 expression in lung cancer based on tissue and cellular models. Functionality assays reported that YTHDC2 overexpression can inhibit the proliferation and migration of lung cancer cells, indicating that it is a vital prognostic factor (18). Therefore, these results highlight controversy regarding the function of YTHDC2 in multiple types of cancers. Furthermore, YTHDC2 is implicated in breast cancer progression by modulating transcription factors responsible for cellular stemness, indicating its viability as a target for therapeutic interventions (33). In the present study, YTHDC2 overexpression alleviated HPV-positive CC cell proliferation and cell viability, as demonstrated by the CCK-8 and colony formation assay results. Additionally, the manifestations of ferroptosis, including apoptosis, ROS generation and biomarker (SLC7A11, ACSL4, GPX4 and p53) dysregulation, was induced after YTHDC2 overexpression in SiHa and CaSKi cells. These observations indicate that YTHDC2 exerts a notable tumor-suppressing effect on HPV-positive CC, with multifaceted roles across several cancer types.

Pertaining to the dynamics between HPV and YTHDC2, the findings of the present study indicate that m6A modifications may serve a pivotal role in the advancement of HPV-positive CC. Enhancing the expression levels of YTHDC2 within CC raises questions about the potential regulatory effects of HPV on YTHDC2 or other m6A modifying enzymes. Further evaluating the connections between HPV and m6A regulatory mechanisms with a particular focus on YTHDC2 could provide critical insights into the molecular processes driving the progression of CC induced by HPV.

The results of the present study also demonstrated that SLC7A11 is conversely controlled by YTHDC2 in an m6A modification-dependent manner. Previous studies of the occurrence of ferroptosis in CC have not specifically targeted HPV-positive CC (34,35). Ferroptosis is distinguished by a non-apoptotic, programmed cell death process initiated by the deactivation of GPX4 and SLC7A11, followed by iron-dependent lipid peroxidation (36). Glutathione (GSH) is a crucial GPX4-reducing agent that is pivotal for maintaining cellular redox homeostasis and shielding cells from oxidative damage by curtailing ROS accumulation. Inhibition of the SLC7A11 system markedly reduces cystine levels within the cell, curtails GSH metabolism and consequently triggers ferroptosis (37). In the analysis in the present study, a negative association between SLC7A11 expression and YTHDC2 was identified in HPV-positive CC cells. Therefore, reduced SLC7A11 expression inhibited cell proliferation and induced ferroptosis in cells, which is consistent with previous studies (38). Increasing SLC7A11 levels partially negates the effects of YTHDC2 overexpression on proliferation and ferroptosis in SiHa and CaSKi cells. Consequently, the present study elucidated a novel pathway wherein YTHDC2 diminishes SLC7A11 expression, thereby amplifying ferroptosis.

In conclusion, the present study identified and assessed the interaction between YTHDC2 and the 5′-UTR of SLC7A11, based on results from luciferase assays and phenotypic changes. The present study also demonstrated that SLC7A11-associated ferroptosis is regulated by YTHDC2. However, future studies should focus on further experiments and an in-depth analysis of the underlying molecular interaction and signal pathway. This lack of an in-depth analysis is considered a limitation of the present study. Another limitation is that sample size used in the patient sample analysis was too low to reach a solid conclusion. In summary, the present research demonstrates that YTHDC2 facilitates cell proliferation and suppresses ferroptosis in HPV-positive CC cells. Additionally, SLC7A11 was identified as a direct target influenced by YTHDC2. The regulatory effect of YTHDC2 on SLC7A11 occurs through an m6A-mediated translational mechanism, which is critical for the pathology of HPV-positive CC. These insights suggest that targeting YTHDC2 could offer a promising therapeutic avenue for treating HPV-positive CC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

LR designed the study. JZ, JY and JG performed the research. LR and LJ analyzed the data. LR and JZ wrote the paper. All authors read and approved the final manuscript. LR and JZ confirm the authenticity of all the raw data.

Ethics approval and consent to participate

The present study was approved by the ethics committee of the First Affiliated Hospital of Henan University of Science and Technology (approval no. 2023-0147). Written informed consent was obtained from every participant involved.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kumar S, Malviya R and Meenakshi DU: Overview of Women's Health. Women's Health: A Comprehensive Guide to Common Health Issues in Women. Bentham Science Publishers; pp. 1–21. 2024

2 

Sun D, Li H, Cao M, He S, Lei L, Peng J and Chen W: Cancer burden in China: Trends, risk factors and prevention. Cancer Biol Med. 17:879–895. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Zhou L, Li Y, Wang H, Qin R, Han Z and Li R: Global cervical cancer elimination: Quantifying the status, progress, and gaps. BMC Med. 23:672025. View Article : Google Scholar : PubMed/NCBI

4 

Kombe Kombe AJ, Li B, Zahid A, Mengist HM, Bounda GA, Zhou Y and Jin T: Epidemiology and burden of human papillomavirus and related diseases, molecular pathogenesis, and vaccine evaluation. Front Public Health. 8:5520282021. View Article : Google Scholar : PubMed/NCBI

5 

Schiffman M, Castle PE, Jeronimo J, Rodriguez AC and Wacholder S: Human papillomavirus and cervical cancer. Lancet. 370:890–907. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Liao S, Sun H and Xu C: YTH domain: A family of N6-methyladenosine (m6A) readers. Genomics Proteomics Bioinformatics. 16:99–107. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Shi R, Ying S, Li Y, Zhu L, Wang X and Jin H: Linking the YTH domain to cancer: The importance of YTH family proteins in epigenetics. Cell Death Dis. 12:3462021. View Article : Google Scholar : PubMed/NCBI

8 

Ma C, Liao S and Zhu Z: Crystal structure of human YTHDC2 YTH domain. Biochem Biophys Res Commun. 518:678–684. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Zaccara S, Ries RJ and Jaffrey SR: Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol. 20:608–624. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Mao Y, Dong L, Liu XM, Guo J, Ma H, Shen B and Qian SB: m6A in mRNA coding regions promotes translation via the RNA helicase-containing YTHDC2. Nat Commun. 10:53322019. View Article : Google Scholar : PubMed/NCBI

11 

Tanabe A, Tanikawa K, Tsunetomi M, Takai K, Ikeda H, Konno J, Torigoe T, Maeda H, Kutomi G, Okita K, et al: RNA helicase YTHDC2 promotes cancer metastasis via the enhancement of the efficiency by which HIF-1α mRNA is translated. Cancer Lett. 376:34–42. 2016. View Article : Google Scholar : PubMed/NCBI

12 

He JJ, Li Z, Rong ZX, Gao J, Mu Y, Guan YD, Ren XX, Zi YY, Liu LY, Fan Q, et al: m6A reader YTHDC2 promotes radiotherapy resistance of nasopharyngeal carcinoma via activating IGF1R/AKT/S6 signaling axis. Front Oncol. 10:11662020. View Article : Google Scholar : PubMed/NCBI

13 

Wang W, Sun B, Xia Y, Sun S and He C: RNA N6-methyladenosine-related gene contribute to clinical prognostic impact on patients with liver cancer. Front Genet. 11:3062020. View Article : Google Scholar : PubMed/NCBI

14 

Liu J, Wang D, Zhou J, Wang L, Zhang N, Zhou L, Zeng J, Liu J and Yang M: N6-methyladenosine reader YTHDC2 and eraser FTO may determine hepatocellular carcinoma prognoses after transarterial chemoembolization. Arch Toxicol. 95:1621–1629. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Wang W, Li J, Lin F, Guo J and Zhao J: Identification of N 6-methyladenosine-related lncRNAs for patients with primary glioblastoma. Neurosurg Rev. 44:463–470. 2021. View Article : Google Scholar : PubMed/NCBI

16 

Wu Q, Xie X, Huang Y, Meng S, Li Y, Wang H and Hu Y: N6-methyladenosine RNA methylation regulators contribute to the progression of prostate cancer. J Cancer. 12:682–692. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Zhao X and Cui L: Development and validation of a m6A RNA methylation regulators-based signature for predicting the prognosis of head and neck squamous cell carcinoma. Am J Cancer Res. 9:2156–2169. 2019.PubMed/NCBI

18 

Sun S, Han Q, Liang M, Zhang Q, Zhang J and Cao J: Downregulation of m6A reader YTHDC2 promotes tumor progression and predicts poor prognosis in non-small cell lung cancer. Thorac Cancer. 11:3269–3279. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Benevolo M, Mottolese M, Marandino F, Vocaturo G, Sindico R, Piperno G, Mariani L, Sperduti I, Canalini P, Donnorso RP and Vocaturo A: Immunohistochemical expression of p16INK4a is predictive of HR-HPV infection in cervical low-grade lesions. Mod Pathol. 19:384–391. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Liu Y, Fan P, Yang Y, Xu C, Huang Y, Li D, Qing Q, Sun C and Zhou H: Human papillomavirus and human telomerase RNA component gene in cervical cancer progression. Sci Rep. 9:159262019. View Article : Google Scholar : PubMed/NCBI

21 

Wright JD, Matsuo K, Huang Y, Tergas AI, Hou JY, Khoury-Collado F, St Clair CM, Ananth CV, Neugut AI and Hershman DL: Prognostic performance of the 2018 international federation of gynecology and obstetrics cervical cancer staging guidelines. Obstet Gynecol. 134:49–57. 2019. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Yan HF, Zou T, Tuo QZ, Xu S, Li H, Belaidi AA and Lei P: Ferroptosis: Mechanisms and links with diseases. Sig Transduct Target Ther. 6:492021. View Article : Google Scholar : PubMed/NCBI

24 

Ma L, Chen T, Zhang X, Miao Y, Tian X, Yu K, Xu X, Niu Y, Guo S, Zhang C, et al: The m(6)A reader YTHDC2 inhibits lung adenocarcinoma tumorigenesis by suppressing SLC7A11-dependent antioxidant function. Redox Biol. 38:1018012021. View Article : Google Scholar : PubMed/NCBI

25 

Zhang K, Yang Z, Yang Z, Du L, Zhou Y, Fu S, Wang X, Li X, Liu D and He X: The m6A reader YTHDC2 promotes the pathophysiology of temporal lobe epilepsy by modulating SLC7A11-dependent glutamate dysregulation in astrocytes. Theranostics. 14:5551–5570. 2024. View Article : Google Scholar : PubMed/NCBI

26 

Chen SH, Song YY, Gan N, Wang PT, Yan K, Wang SF, Zu YE and Peng XW: Human papillomavirus infection and screening strategies. World J Clin Oncol. 16:1050552025. View Article : Google Scholar : PubMed/NCBI

27 

Su H, Wang G, Wu L, Ma X, Ying K and Zhang R: Transcriptome-wide map of m6A circRNAs identified in a rat model of hypoxia mediated pulmonary hypertension. BMC Genomics. 21:392020. View Article : Google Scholar : PubMed/NCBI

28 

Mo X-B, Zhang Y-H and Lei S-F: Genome-wide identification of N6-methyladenosine (m6A) SNPs associated with rheumatoid arthritis. Front Genet. 9:2992018. View Article : Google Scholar : PubMed/NCBI

29 

De Jesus DF, Zhang Z, Kahraman S, Brown NK, Chen M, Hu J, Gupta MK, He C and Kulkarni RN: m6A mRNA methylation regulates human β-cell biology in physiological states and in type 2 diabetes. Nat Metab. 1:765–774. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Paris J, Morgan M, Campos J, Spencer GJ, Shmakova A, Ivanova I, Mapperley C, Lawson H, Wotherspoon DA, Sepulveda C, et al: Targeting the RNA m6A reader YTHDF2 selectively compromises cancer stem cells in acute myeloid leukemia. Cell Stem Cell. 25:137–148.e6. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Wang Q, Guo X, Li L, Gao Z, Su X, Ji M and Liu J: N6-methyladenosine METTL3 promotes cervical cancer tumorigenesis and Warburg effect through YTHDF1/HK2 modification. Cell Death Dis. 11:9112020. View Article : Google Scholar : PubMed/NCBI

32 

Wang H, Luo Q, Kang J, Wei Q, Yang Y, Yang D, Liu X, Liu T and Yi P: YTHDF1 aggravates the progression of cervical cancer through m6A-mediated up-regulation of RANBP2. Front Oncol. 11:6503832021. View Article : Google Scholar : PubMed/NCBI

33 

Tanabe A, Nakayama T, Kashiyanagi J, Yamaga H, Hirohashi Y, Torigoe T, Satomi F, Shima H, Maeda H, Kutomi G, et al: YTHDC2 promotes malignant phenotypes of breast cancer cells. J Oncol. 2022:91889202022. View Article : Google Scholar : PubMed/NCBI

34 

Qi X, Zhou J, Wang X, Shen Y, Cao Y, Jiang L, Shen M, Zhang H, Wang T, Wei P, et al: HPV E6/E7-induced acetylation of a peptide encoded by a long non-coding RNA inhibits ferroptosis to promote the malignancy of cervical cancer. Adv Sci (Weinh). 12:e24140182025. View Article : Google Scholar : PubMed/NCBI

35 

Wei E: Heterogeneity analysis of low-risk HPV infection and high-risk HPV infection, HPV-positive and HPV-negative cancers. Dissertation; LMU München: pp. 1–88. 2023

36 

Song X, Zhu S, Chen P, Hou W, Wen Q, Liu J, Xie Y, Liu J, Klionsky DJ, Kroemer G, et al: AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc-activity. Curr Biol. 28:2388–2399.e5. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Chen H, Cao L, Han K, Zhang H, Cui J, Ma X, Zhao S, Zhao C, Yin S, Yin S, et al: Patulin disrupts SLC7A11-cystine-cysteine-GSH antioxidant system and promotes renal cell ferroptosis both in vitro and in vivo. Food Chem Toxicol. 166:1132552022. View Article : Google Scholar : PubMed/NCBI

38 

Angeli JPF, Shah R, Pratt DA and Conrad M: Ferroptosis inhibition: Mechanisms and opportunities. Trends Pharmacol Sci. 38:489–498. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Ren L, Zhang J, Yang J, Ji L and Guo J: YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner. Oncol Lett 30: 498, 2025.
APA
Ren, L., Zhang, J., Yang, J., Ji, L., & Guo, J. (2025). YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner. Oncology Letters, 30, 498. https://doi.org/10.3892/ol.2025.15244
MLA
Ren, L., Zhang, J., Yang, J., Ji, L., Guo, J."YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner". Oncology Letters 30.5 (2025): 498.
Chicago
Ren, L., Zhang, J., Yang, J., Ji, L., Guo, J."YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner". Oncology Letters 30, no. 5 (2025): 498. https://doi.org/10.3892/ol.2025.15244
Copy and paste a formatted citation
x
Spandidos Publications style
Ren L, Zhang J, Yang J, Ji L and Guo J: YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner. Oncol Lett 30: 498, 2025.
APA
Ren, L., Zhang, J., Yang, J., Ji, L., & Guo, J. (2025). YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner. Oncology Letters, 30, 498. https://doi.org/10.3892/ol.2025.15244
MLA
Ren, L., Zhang, J., Yang, J., Ji, L., Guo, J."YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner". Oncology Letters 30.5 (2025): 498.
Chicago
Ren, L., Zhang, J., Yang, J., Ji, L., Guo, J."YTHDC2 inhibits HPV‑positive cervical cancer growth by suppressing SLC7A11 in a ferroptosis‑dependent manner". Oncology Letters 30, no. 5 (2025): 498. https://doi.org/10.3892/ol.2025.15244
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