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
February-2026 Volume 31 Issue 2

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
February-2026 Volume 31 Issue 2

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
Article Open Access

Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells

  • Authors:
    • Chia-Wei Kao
    • Yen-Ping Yeh
    • Ting-Wen Chen
    • Min-Ying Lin
    • Jyh-Der Leu
    • Yi-Jang Lee
  • View Affiliations / Copyright

    Affiliations: Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan, R.O.C., Industrial Development Graduate Program of College of Engineering Bioscience, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan, R.O.C., Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan, R.O.C., Division of Radiation Oncology, Taipei City Hospital Renai Branch, Taipei 106, Taiwan, R.O.C.
    Copyright: © Kao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 56
    |
    Published online on: November 27, 2025
       https://doi.org/10.3892/ol.2025.15409
  • 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

Hypopharyngeal cancer (HPC) is a highly aggressive cancer with a poor prognosis due to frequent metastasis and resistance to therapy. However, genes associated with the malignancy of advanced HPC remain to be elucidated. To investigate molecular changes associated with HPC progression, RNA sequencing was performed to compare transcriptomic profiles between parental FaDu HPC cells and advanced FaDu cells (FaDuex) derived from late‑stage xenograft tumors. The present analysis revealed 86 markedly upregulated genes and 166 markedly downregulated genes in FaDuex cells compared with FaDu cells. Gene set enrichment analysis revealed enrichment of pathways associated with epithelial‑mesenchymal transition and cell proliferation among upregulated genes, whereas downregulated genes were associated with metastasis inhibition, angiogenesis suppression and immune response regulation. Notably, several interferon (IFN)‑stimulated genes were suppressed in advanced cells, indicating impaired IFN signaling and reduced antitumor immune responses. The most notable changes in the expression levels of genes associated with these tumor characteristics were then validated using quantitative PCR. These findings suggest that genes associated with tumor malignancy and immunosuppressive pathways potentially contribute to HPC progression and highlight potential molecular targets for diagnostic and therapeutic intervention in the future.

Introduction

Hypopharyngeal cancer (HPC) is a highly aggressive subtype of head and neck squamous cell carcinoma (HNSCC) developing from the epithelial lining of the hypopharynx. In the United States, HPC accounts for 3–5% of all head and neck malignancies and is often diagnosed at an advanced stage due to its asymptomatic early progression and the anatomical complexity of the hypopharynx (1,2). Risk factors, including tobacco use, excessive alcohol consumption and human papillomavirus infection, contribute markedly to its pathogenesis (3,4). HPC is clinically classified according to the TNM staging system; early-stage disease (stage I and II) is often localized, while in its advanced stages (stage III and IV), regional lymph node involvement and distant metastasis are observed, leading to poor prognosis (5). Due to the aggressive nature of late-stage HPC, treatment strategies involve a combination of surgery, radiotherapy and chemotherapy; however, five-year survival rates have been reported to be 15–45%, due to high recurrence and resistance to therapy (6).

The progression of HPC, at the molecular level, is driven by a complex network of signaling pathways that regulate tumor growth, angiogenesis and metastasis. A hallmark of HPC is aberrant activation of the EGFR pathway, promoting uncontrolled cell proliferation through downstream effectors such as Ras-Raf-MEK-extracellular signal-regulated kinase and phosphoinositide 3 kinase-mammalian target of rapamycin, which promote tumor cell survival, proliferation and resistance to apoptosis (7). The epithelial-mesenchymal transition (EMT) process, mediated by transforming growth factor-β, Wnt/β-catenin and Notch signaling, is pivotal in enhancing tumor cell motility and invasiveness (8). Therefore, angiogenesis is a key factor supporting metastasis, driven by hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor for neovascularization, ensuring an adequate supply of oxygen and nutrients to proliferating tumor cells (9). Increased angiogenic activity in advanced HPC is associated with increased metastatic potential and worse clinical outcomes (10). Recent studies have highlighted the role of HIF-1α in promoting metabolic reprogramming and angiogenesis, contributing to tumor progression and resistance to therapy (11,12).

Suppression of interferon (IFN) signaling has been identified as a key mechanism of immune evasion in HPC, reducing the susceptibility of the tumor to immune surveillance. IFN-stimulated genes (ISGs) are required for the enhancement of immune recognition and antiviral responses, while they have been reported to be frequently downregulated, impairing antitumor immunity (13,14). The loss of IFN signaling not only reduces immune-mediated tumor clearance but also promotes resistance to immunotherapy.

The present study aimed to identify the differentially expressed genes (DEGs) between parental FaDu HPC cells and advanced FaDu (FaDuex) cells, which were isolated from a late-stage xenograft tumor, were analyzed using the RNA-sequencing (RNA-seq) technique.

Materials and methods

Cell lines

The human HPC FaDu cells (cat no. HTB-43TM; American Type Culture Collection) and FaDuex cells were cultured in RPMI-1640 medium (Thermo Fisher Scientific, Inc.) containing 10% FBS (Thermo Fisher Scientific, Inc.), 1% penicillin/streptomycin and 1% L-glutamine (both MilliporeSigma; Merck KGaA). The FaDu cells were originally derived from a punch biopsy of a hypopharyngeal tumor obtained from a 56-year-old White male patient (15). FaDuex cells were previously established and considered as late-stage or advanced HPC cells, isolated from FaDu cells-derived xenograft tumors that reached 2,000 mm3 at 40 days (10). In brief, when the tumor size reached 600 mm3 exhibiting a necrotic region, the tumor was excised and cultured in RPMI-1640 medium supplemented with high-dose antibiotics, allowing viable cells to migrate from the tumor mass. These cells have been demonstrated to be more tumorigenic, exhibiting an increased rate of proliferation, angiogenesis and invasive capacity (10). The biological characteristics of FaDuex cells closely reflect the clinical features of human HPC diagnosed at a late stage (16).

RNA extraction and RNA-seq analysis

RNA was extracted from cells using the GENEZol™ TriRNA Pure Kit (cat. no. GZX100; Geneaid Biotech), according to the manufacturer's instructions. Up to 5×106 cells were harvested by centrifugation at 300 × g for 5 min at room temperature and the culture medium was completely removed. The cell pellet was thoroughly mixed in 700 µl of GENEzol™ reagent by pipetting and incubated for 5 min at room temperature. Ethanol was added in a 1:1 ratio to the lysate and the mixture was processed through a spin column-based protocol. RNA was eluted using RNase-free water and stored at −80°C until further use. For each condition (FaDu and FaDuex), there were two biological replicates and RNA was independently extracted from separate cultures, and reproducibility was ascertained. RNA library preparation and next-generation sequencing were performed by Biotools Co., Ltd. using their RNA-Seq (Q)20M package (service code: B-IRQTNT-E20P). Libraries were constructed using poly(A) mRNA enrichment and sequenced with 150 bp paired-end reads on the Illumina platform. All samples were processed and sequenced in a single batch.

The raw sequencing reads were processed for quality control using Trimmomatic (version 0.38) by Biotools Co., Ltd. and assessed using FastQC and MultiQC (17,18). Clean reads were aligned using HISAT2 to the human reference genome (GRCh38) and read counts for each gene were quantified using ‘featureCounts’. DEGs were analyzed by comparing the transcripts per million (TPM) values of genes between FaDuex and FaDu samples using ‘DESeq2’ to calculate fold-changes and statistical significance of the outcomes (19). Gene set enrichment analysis (GSEA) in ‘clusterProfiler’ (version 4.6.2, Bioconductor release 3.16) (20,21) was performed to identify significant pathways in the Hallmark and C2 gene sets from the Human Molecular Signatures Database (MSigDB; h.all.v2023.2.Hs and c2.all.v2023.2.Hs) (22–24), as well as other pathways of interest, including 23 downstream NFE2 like BZIP transcription factor 2 gene sets [Transcription_Factor_PPIs (25), Rummagene_transcription_factors (26), TRRUST_Transcription_Factors (27) and TRANSFAC_and_JASPAR_PWMs (28)] (26,28,29) and 20 cancer stem cell-related gene sets (30) collected from Enrichr (25,31,32). Only pathways with an adjusted P<0.05, as determined by Benjamini-Hochberg correction, were considered.

Complementary DNA (cDNA) synthesis and quantitative polymerase chain reaction (qPCR)

The cDNA was synthesized using the ToolsQuant II Fast RT kit (cat. no. KRT-BA-06; TOOLS Biotech) following the manufacturer's instructions. Briefly, after quantifying total RNA using a NanoDrop ND-1000 Spectrophotometer (Thermo Fisher Scientific, Inc.), 300 ng of RNA was used as the template for reverse transcription. Genomic DNA contamination was removed by incubating with gDNA Eraser at 42°C for 3 min, followed by reverse transcription at 42°C for 15 min. The resulting cDNA was diluted with 80 µl nuclease-free water and utilized directly for subsequent qPCR analysis employing the StepOnePlus™ Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). Primers used for qPCR are summarized in Table I. Each reaction was carried out in a final volume of 20 µl, comprising 6 µl nuclease-free water, 2 µl primer mix (6 µM forward primer and 6 µM reverse primer), 2 µl cDNA template and 10 µl, EnTurbo™ SYBR Green PCR SuperMix (cat. no. EQ013; High ROX Premixed; ELK Biotechnology). The thermal cycling program was as follows: Initial denaturation at 95°C for 20 sec, followed by 40 cycles of denaturation at 95°C for 3 sec and annealing/extension at 60°C for 30 sec, during which fluorescence signals were recorded. At the end of the amplification reaction, the melt curve was analyzed to confirm product specificity. The expression of each target gene, along with the internal control gene (S26), was analyzed in four technical replicates. Each datum represented a mean of four repeats and was compared using unpaired t-tests. Relative expression levels were estimated based on amplification curves (33).

Table I.

Sequences of primers for each gene analyzed by quantitative PCR.

Table I.

Sequences of primers for each gene analyzed by quantitative PCR.

GenePrimer sequence (5′-3′)
KRT13F: GATGCTGAGGAATGGTTCCACG
R: AGCTCCGTGATCTCTGTCTTGC
IFI44LF: TGCACTGAGGCAGATGCTGCG
R: TCATTGCGGCACACCAGTACAG
IFI6F: TGATGAGCTGGTCTGCGATCCT
R: GTAGCCCATCAGGGCACCAATA
STC1F: GCAGGAAGAGTGCTACAGCAAG
R: CATTCCAGCAGGCTTCGGACAA
MUC16F: GATGTCAAGCCAGGCAGCACAA
R: GAGAGTGGTAGACATTTCTGGGC
S26F: CCGTGCCTCCAAGATGACAAAG
R: ACTCAGCTCCTTACATGGGCTT

[i] KRT13, keratin 13; IFI44L, interferon-induced protein 44-like; IFI6, interferon α-inducible protein 6; STC1, stanniocalcin-1; MUC16, mucin-16; F, forward; R, reverse.

Results and Discussion

Analysis of DEGs between parental and advanced HPC cells isolated from late-stage xenograft tumors

The present study first analyzed the DEGs between FaDu and FaDuex cells (advanced FaDu cells) to identify transcriptional differences associated with their phenotypic variation. The heatmap in Fig. 1A presents distinct clustering patterns; genes upregulated in FaDuex cells are displayed in red and the downregulated genes are presented in green, indicating significant transcriptional reprogramming (Fig. 1A). The differences are further illustrated in the volcano plot (Fig. 1B). A total of 86 genes were significantly upregulated, while 166 genes were significantly downregulated in FaDuex cells relative to their expression in FaDu cells (adjusted P<0.05). These findings suggest that the expression levels of genes in FaDuex cells undergo significant changes, potentially explaining their distinct biological properties.

Comparison of DEGs between FaDuex and
FaDu cells. (A) Heatmap of 252 DEGs between FaDuex and FaDu cells,
with log2 fold-change ≥1.5 or ≤-1.5 and adjusted
P<0.05; (B) the volcano plot displays upregulated and
downregulated genes, with the x-axis representing the
log2 fold-change and the y-axis representing the
-log10 adjusted P-value with log2 fold-change
≥1.5 or ≤-1.5 and -log10(adjusted P-value)
>-log10(0.05). DEGs, differentially expressed genes.
TPM, transcripts per million; FaDuex, advanced FaDu.

Figure 1.

Comparison of DEGs between FaDuex and FaDu cells. (A) Heatmap of 252 DEGs between FaDuex and FaDu cells, with log2 fold-change ≥1.5 or ≤-1.5 and adjusted P<0.05; (B) the volcano plot displays upregulated and downregulated genes, with the x-axis representing the log2 fold-change and the y-axis representing the -log10 adjusted P-value with log2 fold-change ≥1.5 or ≤-1.5 and -log10(adjusted P-value) >-log10(0.05). DEGs, differentially expressed genes. TPM, transcripts per million; FaDuex, advanced FaDu.

Comparison of specific signaling pathways activated and suppressed in advanced HPC cells

GSEA was performed next using the Hallmark and C2 gene sets to investigate the biological processes associated with the DEGs between FaDuex and FaDu cells. Significantly enriched pathways were subsequently categorized into activated and suppressed groups. In the Hallmark gene set, only suppressed enriched pathways were significant and the most suppressed pathways included those involved in ‘IFNα response’, downregulated ‘KRAS signaling’, ‘IFNγ response’, ‘myogenesis’ and ‘heme metabolism’ (Fig. 2A). In the C2 gene set with significant changes, four enriched pathways (upregulated ‘Wotton RUNX targets’, ‘AMIT EGF response’ in MCF10A and HeLa cells and ‘Pedersen metastasis by ERBB2 isoform’) were activated, while 10 pathways were suppressed, including those involved in ‘BROWNE IFN response’ and ‘Pedersen IFN-α response’ (Fig. 2B). Since the IFN pathways are key for antiviral defense and tumor immune surveillance (34,35), their suppression in FaDuex cells possibly indicates an immune evasion phenomenon, potentially facilitating tumor progression and resistance to immune-mediated clearance. These findings highlighted a significant suppression of IFN signaling in FaDuex cells, potentially contributing to their altered immune landscape and tumorigenic potential.

Gene set enrichment analysis between
FaDuex and FaDu cells, using the human molecular signatures
database. The image displays (A) top 10 significantly suppressed
signaling pathways in FaDuex cells, identified using the hallmark
gene set. (B) top signaling pathways activated and suppressed in
FaDuex cells using the C2 gene set. The number of genes involved is
represented by the size of each dot and the color indicates the
adjusted P-value. FaDuex, advanced FaDu.

Figure 2.

Gene set enrichment analysis between FaDuex and FaDu cells, using the human molecular signatures database. The image displays (A) top 10 significantly suppressed signaling pathways in FaDuex cells, identified using the hallmark gene set. (B) top signaling pathways activated and suppressed in FaDuex cells using the C2 gene set. The number of genes involved is represented by the size of each dot and the color indicates the adjusted P-value. FaDuex, advanced FaDu.

Gene regulations of advanced HPC cells involved in different cancerous associated signaling pathways

The changes in the expression levels of genes associated with several pathways involved in metastasis, angiogenesis, EGF signaling and EMT in FaDuex compared with FaDu cells were further examined. Assessment of differential expression of genes revealed that several metastasis-related genes were altered, with keratin 13 (KRT13), RIBC2 and serpin family B member 13 being the most significantly downregulated metastasis-inhibiting genes, and stanniocalcin-1 (STC1), high mobility group nucleosome binding domain 5 and down syndrome cell adhesion molecule being the most significantly upregulated metastasis-promoting genes, suggesting an increase in metastatic potential in FaDuex cells (Fig. 3A). Regarding the angiogenesis pathway, only interferon α-inducible protein 6 (IFI6), insulin-like growth factor binding protein 7 and intercellular adhesion molecule 1 (ICAM1) were significantly downregulated angiogenesis-inhibiting genes in FaDuex cells, indicating a possible promotion of neovascularization (Fig. 3B). Furthermore, a significant upregulation of STC1, doublecortin-like kinase 1, transglutaminase 2, cytochrome P450 family 27 subfamily B member 1 and forkhead box D1 genes was noted, indicating the activation of the EGF-related pathway, which may contribute to enhanced proliferative and survival signaling in FaDuex cells (Fig. 3C). Furthermore, the genes involved in the inhibition of the EMT pathway, including KRT13, interleukin 1 receptor antagonist, aquaporin-3, complement component 1s, tripartite motif-containing protein 29 and transforming growth factor-β receptor 3, were downregulated in FaDuex cells, suggesting a shift toward EMT phenotypes (Fig. 3D). These findings indicated that FaDuex cells exhibit enhanced metastatic, angiogenic, proliferative and EMT-related potential, reflecting a more aggressive phenotype.

Heatmaps of Z-score normalized TPM
values and bar plots of log2 fold-change of DEGs. The
DEGs were associated with (A) metastasis, (B) angiogenesis, (C)
proliferation and survival and (D) epithelial-mesenchymal
transition-related genes. The color gradient indicates the Z-score
of TPM. Bars were colored black for adjusted P≥0.05 and
log2 fold-changes between −1.5 and 1.5. For bar plots
with adjusted P<0.05, log2 fold-changes <-1.5 are
marked in green, while those >1.5 are marked in red, indicating
significant fold-changes. TPM, transcripts per million; DEGs,
differentially expressed genes; FaDuex, advanced FaDu.

Figure 3.

Heatmaps of Z-score normalized TPM values and bar plots of log2 fold-change of DEGs. The DEGs were associated with (A) metastasis, (B) angiogenesis, (C) proliferation and survival and (D) epithelial-mesenchymal transition-related genes. The color gradient indicates the Z-score of TPM. Bars were colored black for adjusted P≥0.05 and log2 fold-changes between −1.5 and 1.5. For bar plots with adjusted P<0.05, log2 fold-changes <-1.5 are marked in green, while those >1.5 are marked in red, indicating significant fold-changes. TPM, transcripts per million; DEGs, differentially expressed genes; FaDuex, advanced FaDu.

Downregulation of interferon signaling-associated genes in advanced HPC cells

The present study analyzed the expression of IFN-responsive genes using the C2 and Hallmark gene sets to evaluate the differences in IFN signaling between FaDu and FaDuex cells. Both analyses revealed a broad suppression of ISGs in FaDuex cells, indicating a significant suppression of IFN signaling. In the C2 gene set (Fig. 4A), key ISGs such as interferon-induced protein 44-like (IFI44L), IFI6 and tripartite motif-containing protein 22, all of which are key mediators of the IFN response and antiviral defense mechanisms, were markedly downregulated. The Hallmark gene set (Fig. 4B) further confirmed the suppression of IFN signaling, demonstrating a decrease in the expression of genes such as IFI44L, caspase-1 and ICAM1, which are involved in immune activation and inflammatory responses. In the Hallmark gene set, mucin-16 (MUC16) was the most significantly downregulated; however, its expression was only stimulated by the combined action of tumor necrosis factor-α and IFN-γ (36). The downregulation of these ISGs suggested that FaDuex cells may have reduced sensitivity to IFN-mediated immune responses, potentially altering their interaction with the immune microenvironment and affecting their ability to respond to external immune challenges.

Heatmaps of Z-score-normalized TPM
values and bar plots of log2 fold-changes for
differentially expressed genes associated with the
interferon-responsive signaling pathway. (A) Analysis of
interferon-stimulated genes using the C2 gene set and (B) the
hallmark gene set. The color gradient indicates the Z-score of TPM.
Bars are colored black for adjusted P-values ≥0.05 and
log2 fold-changes between −1.5 and 1.5. For bar plots
with adjusted P-values <0.05, log2 fold-changes
<-1.5 are marked in green, while those >1.5 are marked in
red, indicating significant fold-changes. TPM, transcripts per
million; FaDuex, advanced FaDu.

Figure 4.

Heatmaps of Z-score-normalized TPM values and bar plots of log2 fold-changes for differentially expressed genes associated with the interferon-responsive signaling pathway. (A) Analysis of interferon-stimulated genes using the C2 gene set and (B) the hallmark gene set. The color gradient indicates the Z-score of TPM. Bars are colored black for adjusted P-values ≥0.05 and log2 fold-changes between −1.5 and 1.5. For bar plots with adjusted P-values <0.05, log2 fold-changes <-1.5 are marked in green, while those >1.5 are marked in red, indicating significant fold-changes. TPM, transcripts per million; FaDuex, advanced FaDu.

Validation of gene expression using qPCR

The results of DEGs analysis were validated by performing qPCR by selecting genes, in FaDu cells and FaDuex cells, whose expression levels exhibited the most significant changes. According to the DEGs analysis, KRT13 and IFI6 were most significantly downregulated in metastasis/EMT and angiogenesis, respectively, whereas STC1 was the most significantly upregulated in proliferative and survival-related gene of FaDuex cells. In addition, IFI44L and MUC16 were most downregulated in the IFN-responsive genes of FaDuex cells according to the C2 and Hallmark gene sets. The qPCR analysis further demonstrated the downregulation of KRT13, IFI44L, IFI6 and MUC16 transcripts (Fig. 5A, B, D and E) and STC1 was upregulated in FaDuex cells (Fig. 5C), consistent with DEG analysis. These results suggested that RNA-seq determined gene expression is useful for the prediction of cancer-related signaling pathways.

Quantitative PCR analysis of genes
expressed in FaDu cells and FaDuex cells. (A) KRT13; (B)
IF16; (C) STC1; (D) IFI44L and (E)
MUC16. Data are presented as mean ± SD from four independent
experiments. **P<0.01 and ****P<0.0001. KRT13, keratin 13;
IFI44L, interferon-induced protein 44-like; IFI6, interferon
α-inducible protein 6; STC1, stanniocalcin-1; MUC16, mucin-16;
FaDuex, advanced FaDu.

Figure 5.

Quantitative PCR analysis of genes expressed in FaDu cells and FaDuex cells. (A) KRT13; (B) IF16; (C) STC1; (D) IFI44L and (E) MUC16. Data are presented as mean ± SD from four independent experiments. **P<0.01 and ****P<0.0001. KRT13, keratin 13; IFI44L, interferon-induced protein 44-like; IFI6, interferon α-inducible protein 6; STC1, stanniocalcin-1; MUC16, mucin-16; FaDuex, advanced FaDu.

The limitations of the present study include a small sample size; in addition, the results were analyzed from human HPC cell lines rather than clinical samples. As mentioned, HPC is a rare subtype of HNSCC (6). In Taiwan, the age-standardized incidence rate for HPC was increased from 1980 and reached 6.46 per 100,000 in 2019 (37). A pilot study may be initiated in the future to determine notable changes of gene expression using a widely used HPC cell line and its derivatives from xenograft tumor. FaDu cells are isolated from the primary tumor of a White male patient with HPC in 1968 (15). Recently, another HPC cell type (CZH1) isolated from a Chinese patient with HPC has been reported, exhibiting a greater capacity for invasion and increased radiosensitivity compared with FaDu cells (38). The present study would be key in exploring the gene expression differences of patients with HPC from different ethnicities. FaDuex cells are isolated from xenograft tumors that have reached 2,000 mm3 and the present study defined them as late-stage tumors because of increased metastatic, tumorigenic, angiogenic and chemoradiotherapy-resistant capacity (10). However, it is difficult to explain the TNM stage of clinical HPC through late-stage FaDuex cells, as they exhibit similar characteristics as advanced human HPC diagnosed in clinics. In addition, the translational limitation may still exist when using FaDuex cells to represent late-stage HPC in clinical settings. Another limitation of the present study was the use of parental FaDu cells for comparison with tumor-derived FaDuex cells rather than the early stage of FaDu tumors (for example, tumor size at 100 mm3). It may be key to compare potent DEGs in FaDu-derived xenograft tumors at the early (small size) stage and the late (large size) stage in the future.

To conclude, the present study demonstrated that FaDuex cells, the arbitrarily defined late-stage HPC cells isolated from xenograft tumors, exhibited several genes - including KRT13, IFI6, STC1, MUC16 and IFI44L - that were differentially expressed and were associated with malignant characteristics, including increased metastasis, angiogenesis, proliferation and decreased inflammatory responses. The present study also observed that RNA-seq data-based DEGs analysis could be validated using qPCR. Although the use of human cell lines may encounter several of the aforementioned limitations, the variation in samples would be low and easy to interpret. The present study results suggest that several genes are associated with the advanced malignancy of HPC cells, which makes this information potentially key for diagnostic and therapeutic considerations in the future.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant of Taipei City Hospital, RenAi Branch (grant no. TPCH-112-12) and that of National Science and Technology Council (grant no. NSTC 114-2314-B-A49-065-MY3).

Availability of data and materials

The data generated in the present study may be found in the Sequence Read Archive (SRA) of National Library of Medicine under accession no. PRJNA1282043 or at the following URL: https://www.ncbi.nlm.nih.gov/sra/?term=PRJNA1282043.

Authors' contributions

CWK conducted the cell culture experiments, RNA preparation, RNA-sequencing arrangement and data analysis. YPY conducted the differential gene expression and gene set enrichment analyses. TWC, JDL and YJL confirmed the authenticity of all the raw data. TWC interpreted the RNA sequencing data, and resolved questions related to the accuracy of this work. MYL isolated and identified different types of FaDu cells. JDL and YJL contributed to the conception and the design of the present study. YJL drafted the manuscript. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Use of artificial intelligence tools

During the preparation of this work, AI tools were used to improve the readability and language of the manuscript or to generate images, and subsequently, the authors revised and edited the content produced by the AI tools as necessary, taking full responsibility for the ultimate content of the present manuscript.

References

1 

Petersen JF, Timmermans AJ, van Dijk BAC, Overbeek LIH, Smit LA, Hilgers FJM, Stuiver MM and van den Brekel MWM: Trends in treatment, incidence and survival of hypopharynx cancer: A 20-year population-based study in the Netherlands. Eur Arch Otorhinolaryngol. 275:181–189. 2018. View Article : Google Scholar

2 

Cordunianu AV, Ganea G, Cordunianu MA, Cochior D, Moldovan CA and Adam R: Hypopharyngeal cancer trends in a high-incidence region: A retrospective tertiary single center study. World J Clin Cases. 11:5666–5677. 2023. View Article : Google Scholar

3 

Sanders O and Pathak S: Hypopharyngeal Cancer. StatPearls. StatPearls Publishing; Treasure Island, FL: 2025

4 

Patel EJ, Oliver JR, Jacobson AS, Li Z, Hu KS, Tam M, Vaezi A, Morris LGT and Givi B: Human papillomavirus in patients with hypopharyngeal squamous cell carcinoma. Otolaryngol Head Neck Surg. 166:109–117. 2022. View Article : Google Scholar

5 

Rami-Porta R, Nishimura KK, Giroux DJ, Detterbeck F, Cardillo G, Edwards JG, Fong KM, Giuliani M, Huang J, Kernstine KH Sr, et al: The International association for the study of lung cancer lung cancer staging project: Proposals for revision of the TNM stage groups in the forthcoming (Ninth) Edition of the TNM classification for lung cancer. J Thorac Oncol. 19:1007–1027. 2024. View Article : Google Scholar

6 

Bozec A, Benezery K, Chamorey E, Ettaiche M, Vandersteen C, Dassonville O, Poissonnet G, Riss JC, Hannoun-Lévi JM, Chand ME, et al: Nutritional status and feeding-tube placement in patients with locally advanced hypopharyngeal cancer included in an induction chemotherapy-based larynx preservation program. Eur Arch Otorhinolaryngol. 273:2681–2687. 2016. View Article : Google Scholar

7 

Malecki K, Glinski B, Mucha-Malecka A, Rys J, Kruczak A, Roszkowski K, Urbańska-Gąsiorowska M and Hetnał M: Prognostic and predictive significance of p53, EGFr, Ki-67 in larynx preservation treatment. Rep Pract Oncol Radiother. 15:87–92. 2010. View Article : Google Scholar

8 

Zhang J, Tian XJ and Xing J: Signal transduction pathways of EMT Induced by TGF-beta, SHH, and WNT and their crosstalks. J Clin Med. 5:412016. View Article : Google Scholar

9 

Ahluwalia A and Tarnawski AS: Critical role of hypoxia sensor-HIF-1α in VEGF gene activation. Implications for angiogenesis and tissue injury healing. Curr Med Chem. 19:90–97. 2012. View Article : Google Scholar

10 

Lin MY, Wang CY, Chan YH, Su SP, Chiang HK, Yang MH and Lee YJ: The emergence of tumor-initiating cells in an advanced hypopharyngeal tumor model exhibits enhanced angiogenesis and nuclear factor erythroid 2-related factor 2-associated antioxidant effects. Antioxid Redox Signal. 41:505–521. 2024. View Article : Google Scholar

11 

Taneja N, Chauhan A, Kulshreshtha R and Singh S: HIF-1 mediated metabolic reprogramming in cancer: Mechanisms and therapeutic implications. Life Sci. 352:1228902024. View Article : Google Scholar

12 

Zhong X, Wang Y, He X, He X, Hu Z, Huang H, Chen J, Chen K, Wei P, Zhao S, et al: HIF1A-AS2 promotes the metabolic reprogramming and progression of colorectal cancer via miR-141-3p/FOXC1 axis. Cell Death Dis. 15:6452024. View Article : Google Scholar

13 

Meyer SP, Bauer R, Brune B and Schmid T: The role of type I interferon signaling in myeloid anti-tumor immunity. Front Immunol. 16:15474662025. View Article : Google Scholar

14 

Critchley-Thorne RJ, Yan N, Nacu S, Weber J, Holmes SP and Lee PP: Down-regulation of the interferon signaling pathway in T lymphocytes from patients with metastatic melanoma. PLoS Med. 4:e1762007. View Article : Google Scholar

15 

Rangan SR: A new human cell line (FaDu) from a hypopharyngeal carcinoma. Cancer. 29:117–121. 1972. View Article : Google Scholar

16 

Keski-Santti H, Luukkaa M, Carpen T, Jouppila-Matto A, Lehtio K, Maenpaa H, Vuolukka K, Vahlberg T and Mäkitie A: Hypopharyngeal carcinoma in Finland from 2005 to 2014: Outcome remains poor after major changes in treatment. Eur Arch Otorhinolaryngol. 280:1361–1367. 2023. View Article : Google Scholar

17 

Ewels P, Magnusson M, Lundin S and Kaller M: MultiQC: Summarize analysis results for multiple tools and samples in a single report. Bioinformatics. 32:3047–3048. 2016. View Article : Google Scholar

18 

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

19 

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

20 

Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, et al: clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation (Camb). 2:1001412021.

21 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar

22 

Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP and Tamayo P: The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 1:417–425. 2015. View Article : Google Scholar

23 

Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P and Mesirov JP: Molecular signatures database (MSigDB) 3.0. Bioinformatics. 27:1739–1740. 2011. View Article : Google Scholar

24 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar

25 

Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, et al: Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44:W90–W97. 2016. View Article : Google Scholar

26 

Clarke DJB, Marino GB, Deng EZ, Xie Z, Evangelista JE and Ma'ayan A: Rummagene: Massive mining of gene sets from supporting materials of biomedical research publications. Commun Biol. 7:4822024. View Article : Google Scholar

27 

Han H, Cho JW, Lee S, Yun A, Kim H, Bae D, Yang S, Kim CY, Lee M, Kim E, et al: TRRUST v2: An expanded reference database of human and mouse transcriptional regulatory interactions. Nucleic Acids Res. 46:D380–D386. 2018. View Article : Google Scholar

28 

Rauluseviciute I, Riudavets-Puig R, Blanc-Mathieu R, Castro-Mondragon JA, Ferenc K, Kumar V, Lemma RB, Lucas J, Chèneby J, Baranasic D, et al: JASPAR 2024: 20th anniversary of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 52:D174–D182. 2024. View Article : Google Scholar

29 

Han H, Cho JW, Lee S, Yun A, Kim H, Bae D, et al: TRRUST v2: an expanded reference database of human and mouse transcriptional regulatory interactions. Nucleic Acids Res. 46:D380–D386. 2018. View Article : Google Scholar

30 

Hu C, Li T, Xu Y, Zhang X, Li F, Bai J, Chen J, Jiang W, Yang K, Ou Q, et al: CellMarker 2.0: An updated database of manually curated cell markers in human/mouse and web tools based on scRNA-seq data. Nucleic Acids Res. 51:D870–D876. 2023. View Article : Google Scholar

31 

Xie Z, Bailey A, Kuleshov MV, Clarke DJB, Evangelista JE, Jenkins SL, Lachmann A, Wojciechowicz ML, Kropiwnicki E, Jagodnik KM, et al: Gene set knowledge discovery with enrichr. Curr Protoc. 1:e902021. View Article : Google Scholar

32 

Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, Clark NR and Ma'ayan A: Enrichr: Interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 14:1282013. View Article : Google Scholar

33 

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

34 

Bonjardim CA, Ferreira PC and Kroon EG: Interferons: Signaling, antiviral and viral evasion. Immunol Lett. 122:1–11. 2009. View Article : Google Scholar

35 

Kaplan DH, Shankaran V, Dighe AS, Stockert E, Aguet M, Old LJ and Schreiber RD: Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc Natl Acad Sci USA. 95:7556–7561. 1998. View Article : Google Scholar

36 

Morgado M, Sutton MN, Simmons M, Warren CR, Lu Z, Constantinou PE, Liu J, Francis LL, Conlan RS, Bast RC Jr and Carson DD: Tumor necrosis factor-α and interferon-ү stimulate MUC16 (CA125) expression in breast, endometrial and ovarian cancers through NFĸB. Oncotarget. 7:14871–14884. 2016. View Article : Google Scholar

37 

Tsai YS, Chen YC, Chen TI, Lee YK, Chiang CJ, You SL, Hsu WL and Liao LJ: Incidence trends of oral cavity, oropharyngeal, hypopharyngeal and laryngeal cancers among males in Taiwan, 1980–2019: A population-based cancer registry study. BMC Cancer. 23:2132023. View Article : Google Scholar

38 

Ma J, Zhu X, Heng Y, Ding X, Tao L and Lu L: Establishment and characterization of a novel hypopharyngeal squamous cell carcinoma cell line CZH1 with genetic abnormalities. Hum Cell. 37:546–559. 2024. View Article : Google Scholar

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Kao C, Yeh Y, Chen T, Lin M, Leu J and Lee Y: Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells. Oncol Lett 31: 56, 2026.
APA
Kao, C., Yeh, Y., Chen, T., Lin, M., Leu, J., & Lee, Y. (2026). Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells. Oncology Letters, 31, 56. https://doi.org/10.3892/ol.2025.15409
MLA
Kao, C., Yeh, Y., Chen, T., Lin, M., Leu, J., Lee, Y."Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells". Oncology Letters 31.2 (2026): 56.
Chicago
Kao, C., Yeh, Y., Chen, T., Lin, M., Leu, J., Lee, Y."Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells". Oncology Letters 31, no. 2 (2026): 56. https://doi.org/10.3892/ol.2025.15409
Copy and paste a formatted citation
x
Spandidos Publications style
Kao C, Yeh Y, Chen T, Lin M, Leu J and Lee Y: Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells. Oncol Lett 31: 56, 2026.
APA
Kao, C., Yeh, Y., Chen, T., Lin, M., Leu, J., & Lee, Y. (2026). Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells. Oncology Letters, 31, 56. https://doi.org/10.3892/ol.2025.15409
MLA
Kao, C., Yeh, Y., Chen, T., Lin, M., Leu, J., Lee, Y."Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells". Oncology Letters 31.2 (2026): 56.
Chicago
Kao, C., Yeh, Y., Chen, T., Lin, M., Leu, J., Lee, Y."Differential gene expression analysis reveals signaling pathways of proliferation, angiogenesis and metastasis is associated with interferon suppression in advanced hypopharyngeal cancer cells". Oncology Letters 31, no. 2 (2026): 56. https://doi.org/10.3892/ol.2025.15409
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