Next Article in Journal
Prokaryotic Na+/H+ Exchangers—Transport Mechanism and Essential Residues
Next Article in Special Issue
Tumor-Intrinsic Nuclear β-Catenin Associates with an Immune Ignorance Phenotype and a Poorer Prognosis in Head and Neck Squamous Cell Carcinomas
Previous Article in Journal
Morphological and Functional Alterations Induced by Two Ecologically Relevant Concentrations of Lead on Danio rerio Gills
Previous Article in Special Issue
Novel Systemic Treatment Modalities Including Immunotherapy and Molecular Targeted Therapy for Recurrent and Metastatic Head and Neck Squamous Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genome-Wide Super-Enhancer-Based Analysis: Identification of Prognostic Genes in Oral Squamous Cell Carcinoma

1
Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
2
Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
3
Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
4
MSD K.K., Tokyo 102-8667, Japan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(16), 9154; https://doi.org/10.3390/ijms23169154
Submission received: 20 June 2022 / Revised: 29 July 2022 / Accepted: 12 August 2022 / Published: 15 August 2022
(This article belongs to the Special Issue Advances in Molecular Mechanism of Head and Neck Cancer)

Abstract

:
Advanced-stage oral squamous cell carcinoma (OSCC) patients are treated with combination therapies, such as surgery, radiation, chemotherapy, and immunotherapy. However, OSCC cells acquire resistance to these treatments, resulting in local recurrence and distant metastasis. The identification of genes involved in drug resistance is essential for improving the treatment of this disease. In this study, we applied chromatin immunoprecipitation sequencing (ChIP-Seq) to profile active enhancers. For that purpose, we used OSCC cell lines that had been exposed to cetuximab for a prolonged period. In total, 64 chromosomal loci were identified as active super-enhancers (SE) according to active enhancer marker histone H3 lysine 27 acetylation (H3K27ac) ChIP-Seq. In addition, a total of 131 genes were located in SE regions, and 34 genes were upregulated in OSCC tissues by TCGA-OSCC analysis. Moreover, high expression of four genes (C9orf89; p = 0.035, CENPA; p = 0.020, PISD; p = 0.0051, and TRAF2; p = 0.0075) closely predicted a poorer prognosis for OSCC patients according to log-rank tests. Increased expression of the four genes (mRNA Z-score ≥ 0) frequently co-occurred in TCGA-OSCC analyses. The high and low expression groups of the four genes showed significant differences in prognosis, suggesting that there are clear differences in the pathways based on the underlying gene expression profiles. These data indicate that potential stratified therapeutic strategies could be used to overcome resistance to drugs (including cetuximab) and further improve responses in drug-sensitive patients.

1. Introduction

Oral cancer is a malignant neoplasm that begins in various parts of the oral cavity, e.g., the tongue, buccal mucosa, and the floor of the mouth. Most of them are oral squamous cell carcinomas (OSCC) [1]. According to Global Cancer Statistics 2018, there are approximately 350,000 new cases of OSCC and 180,000 deaths from the disease per year worldwide [2]. Metastatic and advanced stage cases of oral cancer have a poor prognosis, i.e., the 5-year survival rate is less than 50% [3]. Surgical resection is the first line of treatment of patients with OSCC. However, combinations of chemo-radiation therapy, molecularly targeted therapy and immunotherapy are selected for unresectable or advanced-stage cases [3,4]. First-line treatment regimens for recurrent or metastatic HNSCC include the immune checkpoint inhibitor pembrolizumab [5] and the EXTREME regimen, which includes cetuximab [6]. Cetuximab is an important treatment option as a second-line (and later) option for recurrent or metastatic HNSCC.
In general, cancer cells respond well to initial treatments, but develop resistance during ongoing treatment. There are few effective treatments for cancer cells that have acquired resistance to anticancer drugs (cisplatin; CDDP, 5-fluorouracil; 5-FU, and paclitaxel) or molecular targeted therapy (cetuximab) [7,8]. Identifying molecular events in cancer cells that lead to treatment resistance is an important goal in cancer research and represents significant challenges.
Various analytical approaches (gene expression profiles, noncoding RNA profiles, and chromosomal alteration) have been explored to elucidate the molecular mechanisms of drug resistance [8,9]. Previous studies have revealed that the expression of anticancer drug excretion genes, DNA repair genes, antiapoptotic genes, and epithelial mesenchymal transition (EMT)-related genes may be activated in anticancer drug-resistant cancer cells [10,11,12,13].
Recent studies have revealed that epigenetic factors (e.g., including DNA methylation, histone modifications, non-coding RNAs) are pivotal players in the malignant transformation of cancer cells [14,15,16]. Epigenetic changes in gene expression are reversible and do not alter the DNA sequence but may change the way the DNA sequence is read and expressed.
The binding of transcription factors to an enhancer is an important first step in gene expression [17]. Enhancers are short (50–1500 base pair) DNA regions to which transcription factors bind to increase the likelihood of transcription of a particular gene [18]. Super-enhancers are formed to strongly express pivotal genes that determine the fate of cells [19,20]. A super-enhancer (SE) is a region of the mammalian genome that contains multiple enhancers that collectively bind transcription factors to facilitate gene transcription [19,21]. Elevated enhancer activities are involved in the resistance of cancer cells following treatment with anticancer drugs [22,23,24,25].
Recent cancer research is clarifying the mechanism by which higher-order chromatin structures (modulated by DNA methylation and histone modification) are involved in human oncogenesis and drug resistance [26,27,28]. H3K27ac (acetylation of the lysine residue at N-terminal position 27 of the histone H3 protein) is associated with higher activation of transcription and is therefore defined as an active enhancer mark [29,30].
Chromatin immunoprecipitation sequencing (ChIP-Seq) is a well-established technology that combines chromatin immunoprecipitation (ChIP) with next-generation sequencing. By using this method, it is possible to analyze histone modification and the binding sites of transcriptional regulators (DNA-binding proteins) on the genome in a genome-wide manner [31,32]. ChIP-Seq analysis targeting H3K27ac reveals that a super enhancer in the genome is active [33,34].
In previous studies, we established cetuximab-resistant OSCC cell lines and identified several genes involved in cetuximab resistance [35]. In this study, we attempted to identify super-enhancers involved in drug resistance using OSCC cell lines that had been treated with cetuximab for prolonged periods. A total of 64 genomic loci of H3K27ac-related super-enhancers were identified by comparing the parental cell lines with the cetuximab-treated cell lines. A total of 131 genes were identified in the super-enhancers, and The Cancer Genome Atlas (TCGA) revealed that 34 genes were highly expressed in OSCC clinical tissues. Importantly, high expression of four genes (C9orf89; p = 0.035, CENPA; p = 0.020, PISD; p = 0.0051, and TRAF2; p = 0.0075) closely predicted a poorer prognosis of OSCC patients. Here, we provide information on super-enhancers involved in drug resistance. Identification of candidate genes should accelerate our understanding of the molecular mechanisms of drug resistance.

2. Results

2.1. Genome-Wide Screening of SE in OSCC Cell Lines following Long-Term Treatment with Cetuximab Using H3K27ac ChIP-Seq Analysis

To investigate the dynamic epigenetic state of OSCC after long-term exposure to anticancer drugs, we identified SEs with H3K27ac peaks in control cells (HSC-3 and SAS) and cells that had been subjected to long-term cetuximab exposure (Cmab-LTE). A total of 995 and 1043 SE peaks were detected in Cmab-LTE HSC-3 and Cmab-LTE SAS cells (Figure 1).
Next, we compared the control cells and the Cmab-LTE cells. A total of 152 and 481 SE gain peaks were identified in Cmab-LTE HSC-3 and Cmab-LTE SAS compared with the respective parental cell lines, respectively (Figure 1 and Figure 2). Of these, 68 gain peaks were common to the two cell lines. Analysis of a human genome database revealed that a total of 131 genes corresponded to those SE regions (Figure 2). The detailed information of the genes in the SE regions is shown in Table 1.
These genes might be prognostic markers in patients with OSCC. Thus, clinicopathological analysis of these genes in OSCC patients was performed using the TCGA-OSCC database (below). The 34 genes for which expression was upregulated by TCGA-OSCC tissues are shown in bold in the Table 1, and the details of the genes were listed in Table S1.

2.2. Clinical Significance of C9orf89, CENPA, PISD, and TRAF2, in OSCC Patients Determined by TCGA-OSCC Analysis

TCGA-OSCC database analysis showed that a total of 34 genes were upregulated in OSCC tissues compared to normal tissues (Figure 2, Table S1). Among these upregulated genes, expression of four genes (C9orf89, CENPA, PISD, and TRAF2) significantly predicted 5-year overall survival rates in OSCC patients (Figure 3).
Specific H3K27ac signals in Cmab-LTE cell lines (HSC-3 and SAS) are shown in Figure 4. Chromosomal regions located in C9orf89, CENPA, PISD, and TRAF2 genes are shown in Table 1. Further analysis of these four genes was performed.

2.3. Expression Levels of C9orf89, CENPA, PISD, and TRAF2 in OSCC Cells after Long-Term Exposure to Cetuximab

We verified that gene expression levels were induced by cetuximab treatment. The expression levels of four genes (C9orf89, CENPA, PISD, and TRAF2) were significantly upregulated after exposure to cetuximab compared to parent cells (Figure 5).

2.4. Alteration of mRNA Expression of C9orf89, CENPA, PISD, and TRAF2 in OSCC Patients Determined by TCGA-OSCC Analysis

The high expression status of four genes in the TCGA-OSCC cohort using cBioportal is illustrated in Oncoprint. In 321 OSCC clinical samples, we noted high mRNA expression for C9orf89 in 50%, CENPA in 40%, PISD in 33%, and TRAF2 in 22%, and (Z-score ≥ 0). These genes showed significantly higher mRNA expression with increasing DNA copy number (Figure 6A, Supplemental Figure S1). The expression of all four genes was often increased simultaneously (mRNA Z-score ≥ 0, Figure 6B). These data suggest that the four genes (which do not share the same locus) are regulated by SEs specific to cells that had been exposed to cetuximab for a prolonged period.
Patients with OSCC who had increased expression in at least one of the four genes showed an unfavorable survival outcome and were characterized by aberrant cell cycle gene signatures (Figure 6C,D). On the other hand, the OSCC patients without any increase of the four genes were associated with Focal Adhesion-PI3K-Akt-mTOR-signaling pathway, Ras Signaling, and Chemokine signaling pathway and had a more favorable prognosis (Figure 6D, Supplemental Figure S2).
This analysis of clinical specimens of TCGA-OSCC suggested that the four candidate genes were co-altered by the formation of a super-enhancer and these changes negatively affected the prognosis.

2.5. Immunostaining of C9orf89, CENPA, PISD, and TRAF2 in OSCC Clinical Tissues

Immunohistochemical staining of C9orf89, CENPA, PISD, and TRAF2 was analyzed with the Protein Atlas database (Figure 7, Tables S2 and S3).
C9orf89: Normal tissues displayed occasional nuclear positivity for HPA010921 whereas HPA038297 showed negative staining. In cancer tissues, both antibodies displayed moderate to strong immunoreactivity in the cytoplasm.
CENPA: Weak to strong nuclear positivity was observed in basal cells and parabasal cells in normal tissues. In addition to nuclear positivity, weak cytoplasmic immunoreactivity was observed in malignant cells.
PISD: HPA031090 showed strong cytoplasmic immunoreactivity in both normal and cancer tissues. On the other hand, HPA031091 displayed weak positive staining in normal tissue.
TRAF2: Most normal cells showed negative to weak cytoplasmic positivity with both antibodies, whereas cancer tissues showed strong cytoplasmic positivity.
The moderate to high expression of each gene was confirmed on the cancer tissues, however, normal epithelial tissues were stained in case of a few antibodies.

3. Discussion

OSCC is a highly malignant cancer, and the 5-year survival of OSCC has remained below 50% [3,36]. Discovery of drug susceptibility markers and molecules involved in drug resistance is essential for improving the prognosis of patients with OSCC. A vast number of studies have shown that dysregulated epigenetic control of cancer cells is closely involved in malignant transformation, metastasis, and drug resistance [14,15,16].
The super-enhancer concept is critically important in cancer research, and the identification of cancer cell-specific super-enhancers has been vigorously pursued [22,33,34,37]. A recent study using H3K27ac ChiP-seq analysis of HSC4 and BHY cells showed that 41 genes were regulated by super-enhancers [38]. Among these genes, high expression of AHCY, KCMF1, MANBAL, and TFDP1 predicted poor prognosis of the patients with OSCC [38]. It is evident that genome-wide super-enhancer analyses provide novel information regarding OSCC/HNSCC molecular pathogenesis.
Targeted molecular therapies that specifically block the oncogenic signaling pathways characteristic of cancer cells have improved patient prognosis [39]. Overexpression of EGFR and activation of EGFR-mediated oncogenic pathways are frequently observed in OSCC patients [40,41]. Therefore, anti-EGFR antibodies, such as cetuximab, are used for the treatment of this disease [4,6,42,43]. During treatment with cetuximab, cancer cells acquire genetic alterations such as gain-of-function mutations in EGFR and KRAS, resulting in treatment resistance [44,45]. In order to explore the molecular mechanism involved in cetuximab resistance, we established cetuximab-resistant OSCC cell lines and performed genome-wide gene expression analysis [35]. In this study, we analyzed super-enhancers involved in cetuximab-resistance.
A total of 64 chromosomal loci were identified as active super-enhancers (SE) by active enhancer marker histone H3 lysine 27 acetylation (H3K27ac) ChIP-Seq. Ultimately, we identified four genes (C9orf89, CENPA, PISD, and TRAF2) as super-enhancer-mediated prognostic markers of OSCC patients. In addition, these genes might well be involved in drug resistance in OSCC cells. Clarifying the role of these genes using various drug-resistant cell lines is an important task. Furthermore, it is necessary to confirm whether the expression of these genes fluctuates in clinical specimens before and after drug treatment.
TRAF2 is a member of the TNF-receptor-associated factor family. It acts as a mediator of TNF-induced signaling [46,47,48]. Overexpression of TRAF2 enhances the malignant phenotype of gastric cancer cells [47]. In nasopharyngeal carcinoma cells, overexpression of TRAF2 promotes cancer cell proliferation and anchorage-independent growth [48]. Importantly, its overexpression was associated with resistance to irradiation [48].
CENPA determines the location of the centromere on chromosomes in mitosis. CENPA protein is a histone H3 variant that replaces one or both of the standard H3 histones in the nucleosome histone complex within the centromere [49]. Expression levels of CENPA are associated with patients’ responses to chemo- and radiotherapy and they predict poorer survival rates of cancer patients [50,51].
Aberrant expression of NRMT (N-terminal regulator of chromatin condensation 1 methyltransferase) has been reported in various cancers [52,53]. NRMT controls the expression of CENPA through a promoter region of CENPA [54]. Furthermore, CENPA induces the transcription of Myc and elevates the expression of Bcl2 in retinoblastoma cells [53]. Importantly, aberrant expression of the NRTM/CENPA/Bcl2 axis developed in cisplatin (CDDP) resistance of retinoblastoma cells [53].
Interestingly, the group characterized by high expression of any of the four genes (the high expression group) was enriched for pathway terms associated with cell cycle upregulation in GSEA analysis compared to the group without such expression (low expression group). In this “high expression” patient population, combination therapies targeting the cell cycle, such as CDK4/6 inhibitors, are expected to provide responses not achieved with cetuximab alone. In fact, combination therapy with the CDK4/6 inhibitor palbociclib and cetuximab was used in a phase II trial, and the response rate in the cetuximab-resistant group was 19% (5 of 32 patients had PR) [55].
Pathways that were enriched in the “low-expression” group included the Focal Adhesion-PI3K-Akt-mTOR-signaling pathway, Ras Signaling and the chemokine signaling pathway. These results suggest approaches that co-inhibit RAS-MAPK-ERK signaling and PI3K-AKT-mTOR signaling, or that combine cetuximab with immune checkpoint inhibition. Such approaches have begun to be investigated in preclinical and clinical trials, and co-inhibition of RAS-MAPK-ERK and PI3K-AKT-mTOR signaling has demonstrated antitumor effects in the HNSCC PDX model [56] and the PIK3CA+ OSCC preclinical model [57]. Clinical efficacy will be confirmed in the KURRENT study (NCT04997902).
A combination of immune checkpoint inhibition with cetuximab has shown results suggesting efficacy as measured by CXCL10 expression at the ex vivo assay level [58]. A combination of pembrolizumab and cetuximab in a recent phase II trial demonstrated promising responses and a manageable safety profile [59].
We believe that the four genes we identified in this analysis will help elucidate the mechanisms underlying drug-resistance, including cetuximab-resistance in OSCC cells. In addition, it may be possible to construct a drug susceptibility/diagnostic system for OSCC patients to enable stratification into appropriate treatment regimens based on the expression of those genes as an index. Finally, we acknowledge a limitation of the current approach. That is, the TCGA-OSCC cohort we analyzed in this study does not provide direct evidence based on gene expression profiling of cetuximab-resistant patients and is therefore speculative. However, we anticipate validating these results in ongoing clinical trials.

4. Materials and Methods

4.1. Parental Cell Lines and Cetuximab Long-Term Exposure Cell Lines

OSCC-derived cell lines (HSC-3 and SAS) were purchased from and authenticated by the Human Science Research Resources Bank (Osaka, Japan) or the RIKEN Bio Resource Center (Ibaraki, Japan) and cultured as previously described [60]. Parental cell lines were subjected to prolonged exposure to cetuximab as described previously [35] and used as cetuximab long-term exposure cell lines (Cmab-LTE HSC-3 and Cmab-LTE SAS). Genomic alterations (EGFR mutation status (exon 18 [G719X], exon 19 [E746_A750 deletion], exon 20 [V769_V774 insertions], exon 20 [T790M], and exon 21 [L858R]) and KRAS (codon 12/13) genes) in these cell lines were previously assessed and were not detected in all cell lines [35].

4.2. H3K27ac CHIP Sequencing

For H3K27ac ChIP-seq and super-enhancers analysis, cells were fixed with 1% formaldehyde for 15 min and quenched with 0.125 M glycine, and the frozen cell pellet containing 1 × 107 cells was sent to Active Motif Inc. (Carlsbad, CA, USA) according to the manufacturers’ instructions. ChIP-seq analysis was performed by Active Motif Inc. as reported [61,62,63].
Briefly, chromatin was isolated after treatment with Chromatin Prep Lysis Buffer (Active Motif) containing non-ionic detergent and protease inhibitors, followed by disruption with a Dounce homogenizer. Genomic DNA was sheared to an average length of 300–500 bp using an EpiShear probe sonicator (Active Motif, cat# 53051) and a cooled sonication platform (Active Motif, cat# 53080). The segments of interest were immunoprecipitated by 4 μL of specific antibody against H3K27Ac (Active Motif, cat# 39133, Lot 16119013). The protein and DNA complexes were washed, eluted from the Agarose beads and were treated with SDS buffer, RNase, and proteinase K. Crosslinks were reversed and ChIP DNA was purified by phenolchloroform extraction and ethanol precipitation.
Sequencing libraries were prepared and sequenced on Illumina’s NextSeq 500 (75 nt reads, single-end). The reads were aligned to the human genome (hg38) by BWA (default settings) and non-duplicated mapped reads (mapping quality > 25) were used for further analysis. Alignments were extended in silico at their 3′-ends to a length of 200 bp and assigned to 32-nt bins along the genome. The histograms were stored in bigWig files and peak locations were determined using the MACS algorithm (v2.1.0) with a cutoff p-value = 1 × 10−7. ENCODE blacklist, known as false ChIP-Seq peaks, were removed. Signal maps and peak locations were used as input data to an Active Motifs proprietary analysis program.
Our data resulting from CHIP Sequencing analysis were deposited in the GEO data-base (accession number: GSE205455).

4.3. Super Enhancer Analysis

Super Enhancer regions were determined using BED Tools software [64] and standard UNIX commands. The first step is identical to ROSE and stitches together MACS2 peak regions that are less than 12.5 kb apart (stitching parameter = 12.5 kb). Next, the number of tags (aligned reads in the normalized, BAM-derived BED files) in each of the stitched regions was determined, and the tag numbers were then used to rank the regions. The top 5% were designated Super Enhancers and those were annotated with genes and promoters.

4.4. RNA Extraction and Quantitative Reverse-Transcription PCR (qRT-PCR)

Total RNA was isolated using TRIzol reagent and the PureLink™ RNA Mini Kit (Invitrogen/Thermo Fisher Scientific (Waltham, MA, USA)). Reverse transcription was achieved with the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Waltham, MA, USA). qRT-PCR was performed with SYBR Green using the StepOnePlus™ Real-Time PCR System (Applied Biosystems). GAPDH was used as the normalized control. Primer sequences used in this report are shown in Table S4.

4.5. Analysis of Expression and Clinical Significance of Candidate Gene Expression in OSCC by TCGA Database Analysis

Analysis of expression and clinical significance of the candidate genes was performed by using data from cBioPortal (http://cbioportal.org), accessed on 10 April 2020 [65]. TCGA-OSCC data were defined as TCGA-HNSC data (Firehose Legacy) in which the primary site was the tongue, oral cavity, the floor of the mouth, buccal mucosa, alveolar ridge, hard palate, or lip. We ran queries on four genes (C9orf89, CENPA, PISD, and TRAF2) to specify changes in mRNA expression (Z score ≥ 0) and analyzed mutual exclusivity and overall survival. The ranked gene list for gene set enrichment analysis (GSEA) was obtained from the comparison of mRNA expression levels between altered and non-altered groups and uploaded into WEB-based GEne SeT AnaLysis Toolkit, “WebGestalt” (http://www.webgestalt.org), accessed on 10 April 2022 [66]. We applied “Wikipathway cancer” dataset for GSEA (https://www.wikipathways.org/index.php/WikiPathways), accessed on 10 April 2022 [67].

4.6. Immunostaining Analysis by Protein Atlas Database

To confirm the protein expression levels of target genes, images of immunohistochemical staining were downloaded from The Human Protein Atlas database (https://www.proteinatlas.org), accessed on 24 March 2022 [68,69]. The Human Protein Atlas is a Swedish-based program started in 2003 with the purpose of mapping all human proteins. All the data exhibited in this program is open access for exploration of the human proteome.
The links to the information of each gene, clinical features of the HNSCC patients and the antibody information is summarized in Tables S2 and S3.

4.7. Statistical Analysis

JMP Pro 15 (SAS Institute Inc., Cary, NC, USA) was used for statistical analyses. Comparisons between the two groups were assessed by Welch’s t-test. One-way analysis of variance (ANOVA) was used for comparisons between multiple groups. Overall survival analysis were analyzed by log-rank test. A p-value < 0.05 was considered statistically significant. Quantitative data are presented as the means and standard errors.

5. Conclusions

Elucidation of the mechanism underlying cells’ resistance to anti-cancer drugs is critical for improving the prognosis of OSCC patients. In this study, H3K27ac ChIP-Seq was applied to investigate cetuximab treatment-induced genomic changes. A total of 64 SE peaks were detected in OSCC cells following long-term exposure to cetuximab. A total of 131 genes were involved in the SE region, of which four genes (C9orf89, CENPA, PISD, and TRAF2) affected the prognosis of patients with OSCC. Analysis of these genes will contribute to improved understanding of drug resistance in OSCC patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23169154/s1.

Author Contributions

Conceptualization, T.S., K.U. and N.S.; methodology, T.S., A.K. (Atsushi Kasamatsu) and K.U.; validation, S.A., N.T., N.N. and C.M.; formal analysis, T.S., S.A., C.M. and N.N.; investigation, T.S., S.A. and N.T.; data curation, T.S., S.A., N.T., N.N., C.M. and A.K. (Ayaka Koma); writing—original draft preparation, T.S., S.A., N.T., N.N., C.M. and N.S.; writing—review and editing, A.K. (Ayaka Koma), A.K. (Atsushi Kasamatsu), T.H. and K.U.; visualization, T.S., S.A., N.T., N.N. and C.M.; supervision, A.K. (Atsushi Kasamatsu); project administration, K.U. and N.S.; funding acquisition, N.K., A.K. (Atsushi Kasamatsu), T.H., K.U. and N.S.; All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by JSPS KAKENHI under grant numbers 20H03883, 21K09577 and 22K09679.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Our data resulting from CHIP Sequencing analysis were deposited in the GEO database (accession number: GSE205455).

Acknowledgments

The results shown here are in part based upon data generated by the TCGA Research Network: https://www.cancer.gov/tcga (accessed on 10 April 2020).

Conflicts of Interest

N.N. is an employee of MSD K.K., a subsidiary of Merck & Co., Inc., and reports personal fees from MSD K.K. outside of this study.

References

  1. Dhanuthai, K.; Rojanawatsirivej, S.; Thosaporn, W.; Kintarak, S.; Subarnbhesaj, A.; Darling, M.; Kryshtalskyj, E.; Chiang, C.P.; Shin, H.I.; Choi, S.Y.; et al. Oral cancer: A multicenter study. Med. Oral Patol. Oral Cir. Bucal 2018, 23, e23–e29. [Google Scholar] [CrossRef]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  3. Chow, L.Q.M. Head and Neck Cancer. N. Engl. J. Med. 2020, 382, 60–72. [Google Scholar] [CrossRef]
  4. Caudell, J.J.; Gillison, M.L.; Maghami, E.; Spencer, S.; Pfister, D.G.; Adkins, D.; Birkeland, A.C.; Brizel, D.M.; Busse, P.M.; Cmelak, A.J.; et al. NCCN Guideslines® Insights: Head and Neck Cancers, Version 1.2022. J. Natl. Compr. Cancer Netw. 2022, 20, 224–234. [Google Scholar] [CrossRef] [PubMed]
  5. Burtness, B.; Harrington, K.J.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Basté, N.; Neupane, P.; Bratland, Å.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
  6. Vermorken, J.B.; Mesia, R.; Rivera, F.; Remenar, E.; Kawecki, A.; Rottey, S.; Erfan, J.; Zabolotnyy, D.; Kienzer, H.R.; Cupissol, D.; et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N. Engl. J. Med. 2008, 359, 1116–1127. [Google Scholar] [CrossRef]
  7. Picon, H.; Guddati, A.K. Mechanisms of resistance in head and neck cancer. Am. J. Cancer Res. 2020, 10, 2742–2751. [Google Scholar] [PubMed]
  8. Zaryouh, H.; De Pauw, I.; Baysal, H.; Pauwels, P.; Peeters, M.; Vermorken, J.B.; Lardon, F.; Wouters, A. The Role of Akt in Acquired Cetuximab Resistant Head and Neck Squamous Cell Carcinoma: An In Vitro Study on a Novel Combination Strategy. Front. Oncol. 2021, 11, 697967. [Google Scholar] [CrossRef] [PubMed]
  9. Gomes, I.N.F.; da Silva-Oliveira, R.J.; da Silva, L.S.; Martinho, O.; Evangelista, A.F.; van Helvoort Lengert, A.; Leal, L.F.; Silva, V.A.O.; Dos Santos, S.P.; Nascimento, F.C.; et al. Comprehensive Molecular Landscape of Cetuximab Resistance in Head and Neck Cancer Cell Lines. Cells 2022, 11, 154. [Google Scholar] [CrossRef] [PubMed]
  10. Bar-Zeev, M.; Livney, Y.D.; Assaraf, Y.G. Targeted nanomedicine for cancer therapeutics: Towards precision medicine overcoming drug resistance. Drug Resist. Updates 2017, 31, 15–30. [Google Scholar] [CrossRef] [PubMed]
  11. Cleary, J.M.; Aguirre, A.J.; Shapiro, G.I.; D’Andrea, A.D. Biomarker-Guided Development of DNA Repair Inhibitors. Mol. Cell 2020, 78, 1070–1085. [Google Scholar] [CrossRef]
  12. Carneiro, B.A.; El-Deiry, W.S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 2020, 17, 395–417. [Google Scholar] [CrossRef]
  13. Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 2010, 29, 4741–4751. [Google Scholar] [CrossRef] [PubMed]
  14. Klutstein, M.; Nejman, D.; Greenfield, R.; Cedar, H. DNA Methylation in Cancer and Aging. Cancer Res. 2016, 76, 3446–3450. [Google Scholar] [CrossRef]
  15. Zhao, Z.; Shilatifard, A. Epigenetic modifications of histones in cancer. Genome Biol. 2019, 20, 245. [Google Scholar] [CrossRef] [PubMed]
  16. Anfossi, S.; Babayan, A.; Pantel, K.; Calin, G.A. Clinical utility of circulating non-coding RNAs-an update. Nat. Rev. Clin. Oncol. 2018, 15, 541–563. [Google Scholar] [CrossRef] [PubMed]
  17. Spitz, F.; Furlong, E.E. Transcription factors: From enhancer binding to developmental control. Nat. Rev. Genet. 2012, 13, 613–626. [Google Scholar] [CrossRef]
  18. Blackwood, E.M.; Kadonaga, J.T. Going the distance: A current view of enhancer action. Science 1998, 281, 60–63. [Google Scholar] [CrossRef] [PubMed]
  19. Hnisz, D.; Abraham, B.J.; Lee, T.I.; Lau, A.; Saint-André, V.; Sigova, A.A.; Hoke, H.A.; Young, R.A. Super-enhancers in the control of cell identity and disease. Cell 2013, 155, 934–947. [Google Scholar] [CrossRef] [PubMed]
  20. Fox, S.; Myers, J.A.; Davidson, C.; Getman, M.; Kingsley, P.D.; Frankiewicz, N.; Bulger, M. Hyperacetylated chromatin domains mark cell type-specific genes and suggest distinct modes of enhancer function. Nat. Commun. 2020, 11, 4544. [Google Scholar] [CrossRef]
  21. Whyte, W.A.; Orlando, D.A.; Hnisz, D.; Abraham, B.J.; Lin, C.Y.; Kagey, M.H.; Rahl, P.B.; Lee, T.I.; Young, R.A. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 2013, 153, 307–319. [Google Scholar] [CrossRef]
  22. Li, G.H.; Qu, Q.; Qi, T.T.; Teng, X.Q.; Zhu, H.H.; Wang, J.J.; Lu, Q.; Qu, J. Super-enhancers: A new frontier for epigenetic modifiers in cancer chemoresistance. J. Exp. Clin. Cancer Res. 2021, 40, 174. [Google Scholar] [CrossRef]
  23. Shang, S.; Yang, J.; Jazaeri, A.A.; Duval, A.J.; Tufan, T.; Lopes Fischer, N.; Benamar, M.; Guessous, F.; Lee, I.; Campbell, R.M.; et al. Chemotherapy-Induced Distal Enhancers Drive Transcriptional Programs to Maintain the Chemoresistant State in Ovarian Cancer. Cancer Res. 2019, 79, 4599–4611. [Google Scholar] [CrossRef] [PubMed]
  24. Bao, J.; Li, M.; Liang, S.; Yang, Y.; Wu, J.; Zou, Q.; Fang, S.; Chen, S.; Guo, L. Integrated high-throughput analysis identifies super enhancers associated with chemoresistance in SCLC. BMC Med. Genom. 2019, 12, 67. [Google Scholar] [CrossRef] [PubMed]
  25. Lytle, N.K.; Ferguson, L.P.; Rajbhandari, N.; Gilroy, K.; Fox, R.G.; Deshpande, A.; Schürch, C.M.; Hamilton, M.; Robertson, N.; Lin, W.; et al. A Multiscale Map of the Stem Cell State in Pancreatic Adenocarcinoma. Cell 2019, 177, 572–586.e22. [Google Scholar] [CrossRef] [PubMed]
  26. Gomez, S.; Tabernacki, T.; Kobyra, J.; Roberts, P.; Chiappinelli, K.B. Combining epigenetic and immune therapy to overcome cancer resistance. Semin. Cancer Biol. 2020, 65, 99–113. [Google Scholar] [CrossRef] [PubMed]
  27. Sharma, S.V.; Lee, D.Y.; Li, B.; Quinlan, M.P.; Takahashi, F.; Maheswaran, S.; McDermott, U.; Azizian, N.; Zou, L.; Fischbach, M.A.; et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 2010, 141, 69–80. [Google Scholar] [CrossRef] [PubMed]
  28. Toh, T.B.; Lim, J.J.; Chow, E.K. Epigenetics in cancer stem cells. Mol. Cancer 2017, 16, 29. [Google Scholar] [CrossRef] [PubMed]
  29. Creyghton, M.P.; Cheng, A.W.; Welstead, G.G.; Kooistra, T.; Carey, B.W.; Steine, E.J.; Hanna, J.; Lodato, M.A.; Frampton, G.M.; Sharp, P.A.; et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 2010, 107, 21931–21936. [Google Scholar] [CrossRef]
  30. Villar, D.; Berthelot, C.; Aldridge, S.; Rayner, T.F.; Lukk, M.; Pignatelli, M.; Park, T.J.; Deaville, R.; Erichsen, J.T.; Jasinska, A.J.; et al. Enhancer evolution across 20 mammalian species. Cell 2015, 160, 554–566. [Google Scholar] [CrossRef]
  31. Park, P.J. ChIP-seq: Advantages and challenges of a maturing technology. Nat. Rev. Genet. 2009, 10, 669–680. [Google Scholar] [CrossRef] [PubMed]
  32. Muhammad, I.I.; Kong, S.L.; Akmar Abdullah, S.N.; Munusamy, U. RNA-seq and ChIP-seq as Complementary Approaches for Comprehension of Plant Transcriptional Regulatory Mechanism. Int. J. Mol. Sci. 2019, 21, 167. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, C.Y.; Yu, G.T.; Gao, C.; Chen, J.; Li, Q.L.; Zhang, L.; Wu, M.; Sun, Z.J.; Li, L.Y. Genome-Wide Enhancer Analysis Reveals the Role of AP-1 Transcription Factor in Head and Neck Squamous Cell Carcinoma. Front. Mol. Biosci. 2021, 8, 701531. [Google Scholar] [CrossRef]
  34. Li, Q.L.; Lin, X.; Yu, Y.L.; Chen, L.; Hu, Q.X.; Chen, M.; Cao, N.; Zhao, C.; Wang, C.Y.; Huang, C.W.; et al. Genome-wide profiling in colorectal cancer identifies PHF19 and TBC1D16 as oncogenic super enhancers. Nat. Commun. 2021, 12, 6407. [Google Scholar] [CrossRef] [PubMed]
  35. Uzawa, K.; Amelio, A.L.; Kasamatsu, A.; Saito, T.; Kita, A.; Fukamachi, M.; Sawai, Y.; Toeda, Y.; Eizuka, K.; Hayashi, F.; et al. Resveratrol Targets Urokinase-Type Plasminogen Activator Receptor Expression to Overcome Cetuximab-Resistance in Oral Squamous Cell Carcinoma. Sci. Rep. 2019, 9, 12179. [Google Scholar] [CrossRef] [PubMed]
  36. Silverman, S., Jr. Demographics and occurrence of oral and pharyngeal cancers. The outcomes, the trends, the challenge. J. Am. Dent. Assoc. 2001, 132, 7s–11s. [Google Scholar] [CrossRef]
  37. Zhang, X.; Wang, Y.; Chiang, H.C.; Hsieh, Y.P.; Lu, C.; Park, B.H.; Jatoi, I.; Jin, V.X.; Hu, Y.; Li, R. BRCA1 mutations attenuate super-enhancer function and chromatin looping in haploinsufficient human breast epithelial cells. Breast Cancer Res. 2019, 21, 51. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, L.; Li, H.; Qiu, Y.; Liu, Y.; Liu, X.; Wang, W. Screening and cellular validation of prognostic genes regulated by super enhancers in oral squamous cell carcinoma. Bioengineered 2021, 12, 10073–10088. [Google Scholar] [CrossRef]
  39. Yip, H.Y.K.; Papa, A. Signaling Pathways in Cancer: Therapeutic Targets, Combinatorial Treatments, and New Developments. Cells 2021, 10, 659. [Google Scholar] [CrossRef]
  40. Rubin Grandis, J.; Melhem, M.F.; Barnes, E.L.; Tweardy, D.J. Quantitative immunohistochemical analysis of transforming growth factor-alpha and epidermal growth factor receptor in patients with squamous cell carcinoma of the head and neck. Cancer 1996, 78, 1284–1292. [Google Scholar] [CrossRef]
  41. Rubin Grandis, J.; Melhem, M.F.; Gooding, W.E.; Day, R.; Holst, V.A.; Wagener, M.M.; Drenning, S.D.; Tweardy, D.J. Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J. Natl. Cancer Inst. 1998, 90, 824–832. [Google Scholar] [CrossRef] [PubMed]
  42. National Comprehensive Cancer Network. Head and Neck Cancers (Version 1.2022). Available online: https://www.nccn.org/professionals/physician_gls/pdf/head-and-neck.pdf (accessed on 29 May 2022).
  43. Bonner, J.A.; Harari, P.M.; Giralt, J.; Cohen, R.B.; Jones, C.U.; Sur, R.K.; Raben, D.; Baselga, J.; Spencer, S.A.; Zhu, J.; et al. Radiotherapy plus cetuximab for locoregionally advanced head and neck cancer: 5-year survival data from a phase 3 randomised trial, and relation between cetuximab-induced rash and survival. Lancet Oncol. 2010, 11, 21–28. [Google Scholar] [CrossRef]
  44. Boeckx, C.; Weyn, C.; Vanden Bempt, I.; Deschoolmeester, V.; Wouters, A.; Specenier, P.; Van Laer, C.; Van den Weyngaert, D.; Kockx, M.; Vermorken, J.B.; et al. Mutation analysis of genes in the EGFR pathway in Head and Neck cancer patients: Implications for anti-EGFR treatment response. BMC Res. Notes 2014, 7, 337. [Google Scholar] [CrossRef] [PubMed]
  45. Janjigian, Y.Y.; Smit, E.F.; Groen, H.J.; Horn, L.; Gettinger, S.; Camidge, D.R.; Riely, G.J.; Wang, B.; Fu, Y.; Chand, V.K.; et al. Dual inhibition of EGFR with afatinib and cetuximab in kinase inhibitor-resistant EGFR-mutant lung cancer with and without T790M mutations. Cancer Discov. 2014, 4, 1036–1045. [Google Scholar] [CrossRef] [PubMed]
  46. Zhu, S.; Jin, J.; Gokhale, S.; Lu, A.M.; Shan, H.; Feng, J.; Xie, P. Genetic Alterations of TRAF Proteins in Human Cancers. Front. Immunol. 2018, 9, 2111. [Google Scholar] [CrossRef]
  47. Dai, H.; Chen, H.; Xu, J.; Zhou, J.; Shan, Z.; Yang, H.; Zhou, X.; Guo, F. The ubiquitin ligase CHIP modulates cellular behaviors of gastric cancer cells by regulating TRAF2. Cancer Cell Int. 2019, 19, 132. [Google Scholar] [CrossRef] [PubMed]
  48. Zhu, H.; Ding, W.; Wu, J.; Ma, R.; Pan, Z.; Mao, X. TRAF2 Knockdown in Nasopharyngeal Carcinoma Induced Cell Cycle Arrest and Enhanced the Sensitivity to Radiotherapy. BioMed Res. Int. 2020, 2020, 1641340. [Google Scholar] [CrossRef]
  49. Mahlke, M.A.; Nechemia-Arbely, Y. Guarding the Genome: CENP-A-Chromatin in Health and Cancer. Genes 2020, 11, 810. [Google Scholar] [CrossRef]
  50. Verrelle, P.; Meseure, D.; Berger, F.; Forest, A.; Leclère, R.; Nicolas, A.; Fortas, E.; Sastre-Garau, X.; Lae, M.; Boudjemaa, S.; et al. CENP-A Subnuclear Localization Pattern as Marker Predicting Curability by Chemoradiation Therapy for Locally Advanced Head and Neck Cancer Patients. Cancers 2021, 13, 3928. [Google Scholar] [CrossRef]
  51. Zhang, S.; Xie, Y.; Tian, T.; Yang, Q.; Zhou, Y.; Qiu, J.; Xu, L.; Wen, N.; Lv, Q.; Du, Z. High expression levels of centromere protein A plus upregulation of the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling pathway affect chemotherapy response and prognosis in patients with breast cancer. Oncol. Lett. 2021, 21, 410. [Google Scholar] [CrossRef]
  52. Shields, K.M.; Tooley, J.G.; Petkowski, J.J.; Wilkey, D.W.; Garbett, N.C.; Merchant, M.L.; Cheng, A.; Schaner Tooley, C.E. Select human cancer mutants of NRMT1 alter its catalytic activity and decrease N-terminal trimethylation. Protein Sci. 2017, 26, 1639–1652. [Google Scholar] [CrossRef]
  53. Li, Z.; Zhang, L.; Liu, D.; Yang, Z.; Xuan, D.; Zhang, Y. Knockdown of NRMT enhances sensitivity of retinoblastoma cells to cisplatin through upregulation of the CENPA/Myc/Bcl2 axis. Cell Death Discov. 2022, 8, 14. [Google Scholar] [CrossRef] [PubMed]
  54. Sathyan, K.M.; Fachinetti, D.; Foltz, D.R. α-amino trimethylation of CENP-A by NRMT is required for full recruitment of the centromere. Nat. Commun. 2017, 8, 14678. [Google Scholar] [CrossRef]
  55. Adkins, D.; Ley, J.; Neupane, P.; Worden, F.; Sacco, A.G.; Palka, K.; Grilley-Olson, J.E.; Maggiore, R.; Salama, N.N.; Trinkaus, K.; et al. Palbociclib and cetuximab in platinum-resistant and in cetuximab-resistant human papillomavirus-unrelated head and neck cancer: A multicentre, multigroup, phase 2 trial. Lancet Oncol. 2019, 20, 1295–1305. [Google Scholar] [CrossRef]
  56. Smith, A.; Chan, S.; McCloskey, A.; Vora, H.; Burrows, F.; Malik, S. Antitumor Activity of Tipifarnib and PI3K Pathway Inhibitor in HRAS-associated HNSCC. Int. J. Radiat. Oncol. Biol. Phys. 2022, 112, e48. [Google Scholar] [CrossRef]
  57. Yamaguchi, K.; Iglesias-Bartolomé, R.; Wang, Z.; Callejas-Valera, J.L.; Amornphimoltham, P.; Molinolo, A.A.; Cohen, E.E.; Califano, J.A.; Lippman, S.M.; Luo, J.; et al. A synthetic-lethality RNAi screen reveals an ERK-mTOR co-targeting pro-apoptotic switch in PIK3CA+oral cancers. Oncotarget 2016, 7, 10696–10709. [Google Scholar] [CrossRef]
  58. Berszin, M.; Michaelides, I.; Siemert, J.; Röhl, L.; Wellhausen, J.; Wald, T.; Bohr, C.; Künzel, J.; Gradistanac, T.; Dietz, A.; et al. Cytokine Profiles of Head and Neck Squamous Cell Carcinoma Undergoing Dual Immunotherapy With Cetuximab and Pembrolizumab Identify Interferon Gamma-Induced Protein 10 as Novel Biomarker. Front. Oncol. 2022, 12, 795277. [Google Scholar] [CrossRef]
  59. Sacco, A.G.; Chen, R.; Worden, F.P.; Wong, D.J.L.; Adkins, D.; Swiecicki, P.; Chai-Ho, W.; Oppelt, P.; Ghosh, D.; Bykowski, J.; et al. Pembrolizumab plus cetuximab in patients with recurrent or metastatic head and neck squamous cell carcinoma: An open-label, multi-arm, non-randomised, multicentre, phase 2 trial. Lancet Oncol. 2021, 22, 883–892. [Google Scholar] [CrossRef]
  60. Saito, T.; Kasamatsu, A.; Ogawara, K.; Miyamoto, I.; Saito, K.; Iyoda, M.; Suzuki, T.; Endo-Sakamoto, Y.; Shiiba, M.; Tanzawa, H.; et al. Semaphorin7A Promotion of Tumoral Growth and Metastasis in Human Oral Cancer by Regulation of G1 Cell Cycle and Matrix Metalloproteases: Possible Contribution to Tumoral Angiogenesis. PLoS ONE 2015, 10, e0137923. [Google Scholar] [CrossRef]
  61. Ndoja, A.; Reja, R.; Lee, S.H.; Webster, J.D.; Ngu, H.; Rose, C.M.; Kirkpatrick, D.S.; Modrusan, Z.; Chen, Y.J.; Dugger, D.L.; et al. Ubiquitin Ligase COP1 Suppresses Neuroinflammation by Degrading c/EBPβ in Microglia. Cell 2020, 182, 1156–1169.e12. [Google Scholar] [CrossRef]
  62. Wang, Q.; Ozer, H.G.; Wang, B.; Zhang, M.; Urabe, G.; Huang, Y.; Kent, K.C.; Guo, L.W. A hierarchical and collaborative BRD4/CEBPD partnership governs vascular smooth muscle cell inflammation. Mol. Ther. Methods Clin. Dev. 2021, 21, 54–66. [Google Scholar] [CrossRef] [PubMed]
  63. Bhattarai, S.; Akella, A.; Gandhi, A.; Dharap, A. Modulation of Brain Pathology by Enhancer RNAs in Cerebral Ischemia. Mol. Neurobiol. 2021, 58, 1482–1490. [Google Scholar] [CrossRef] [PubMed]
  64. Quinlan, A.R.; Hall, I.M. BEDTools: A flexible suite of utilities for comparing genomic features. Bioinformatics 2010, 26, 841–842. [Google Scholar] [CrossRef]
  65. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, B.; Kirov, S.; Snoddy, J. WebGestalt: An integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res. 2005, 33, W741–W748. [Google Scholar] [CrossRef] [PubMed]
  67. Martens, M.; Ammar, A.; Riutta, A.; Waagmeester, A.; Slenter, D.N.; Hanspers, K.; Ryan, A.M.; Digles, D.; Lopes, E.N.; Ehrhart, F.; et al. WikiPathways: Connecting communities. Nucleic Acids Res. 2021, 49, D613–D621. [Google Scholar] [CrossRef]
  68. Uhlen, M.; Zhang, C.; Lee, S.; Sjöstedt, E.; Fagerberg, L.; Bidkhori, G.; Benfeitas, R.; Arif, M.; Liu, Z.; Edfors, F.; et al. A pathology atlas of the human cancer transcriptome. Science 2017, 357, eaan2507. [Google Scholar] [CrossRef]
  69. Pontén, F.; Jirström, K.; Uhlen, M. The Human Protein Atlas—A tool for pathology. J. Pathol. 2008, 216, 387–393. [Google Scholar] [CrossRef]
Figure 1. Detection of SE peaks in OSCC cell lines after long-term exposure to cetuximab (Cmab-LTE). Identification of SEs in parental cell lines (HSC-3 and SAS) and Cmab-LTE cell lines (HSC-3 Cmab-LTE and SAS Cmab-LTE). The proximal genes of the top 5 super-enhancers in parental and Cmab-LTE cell lines are marked. Identification of specific gained and lost SE peaks in Cmab-LTE cell lines.
Figure 1. Detection of SE peaks in OSCC cell lines after long-term exposure to cetuximab (Cmab-LTE). Identification of SEs in parental cell lines (HSC-3 and SAS) and Cmab-LTE cell lines (HSC-3 Cmab-LTE and SAS Cmab-LTE). The proximal genes of the top 5 super-enhancers in parental and Cmab-LTE cell lines are marked. Identification of specific gained and lost SE peaks in Cmab-LTE cell lines.
Ijms 23 09154 g001
Figure 2. Flowchart of methods used for the identification of prognostic genes in OSCC patients. Venn diagram shows the overlapped gain in SE peak numbers between HSC-3 Cmab-LTE and SAS Cmab-LTE cell lines. A total of 68 SE loci and 131 corresponding genes in SE regions are identified. TCGA-OSCC database analysis shows that (C9orf89, CENPA, PISD, and TRAF2) are closely involved in OSCC molecular pathogenesis.
Figure 2. Flowchart of methods used for the identification of prognostic genes in OSCC patients. Venn diagram shows the overlapped gain in SE peak numbers between HSC-3 Cmab-LTE and SAS Cmab-LTE cell lines. A total of 68 SE loci and 131 corresponding genes in SE regions are identified. TCGA-OSCC database analysis shows that (C9orf89, CENPA, PISD, and TRAF2) are closely involved in OSCC molecular pathogenesis.
Ijms 23 09154 g002
Figure 3. Clinical significance of 4 genes (C9orf89, CENPA, PISD, and TRAF2) in OSCC clinical specimens determined by TCGA-OSCC analysis. (A) Expression levels of 4 target genes (C9orf89, CENPA, PISD, and TRAF2) in OSCC clinical specimens from TCGA-OSCC. All genes were found to be upregulated in OSCC tissues (n = 314) compared with normal tissues (n = 30). (B) Clinical significance of four target genes (C9orf89, CENPA, PISD and TRAF2) according to TCGA-OSCC data analysis. Kaplan–Meier curves of the 5-year overall survival rates according to the expression of each gene. Patients were divided into 2 groups according to the median gene expression level: high and low expression groups. The red and blue lines represent the high and low expression groups, respectively. High expression levels of all 4 genes significantly predicted a poorer prognosis in patients with OSCC. Nominal p-value was calculated by log-rank test.
Figure 3. Clinical significance of 4 genes (C9orf89, CENPA, PISD, and TRAF2) in OSCC clinical specimens determined by TCGA-OSCC analysis. (A) Expression levels of 4 target genes (C9orf89, CENPA, PISD, and TRAF2) in OSCC clinical specimens from TCGA-OSCC. All genes were found to be upregulated in OSCC tissues (n = 314) compared with normal tissues (n = 30). (B) Clinical significance of four target genes (C9orf89, CENPA, PISD and TRAF2) according to TCGA-OSCC data analysis. Kaplan–Meier curves of the 5-year overall survival rates according to the expression of each gene. Patients were divided into 2 groups according to the median gene expression level: high and low expression groups. The red and blue lines represent the high and low expression groups, respectively. High expression levels of all 4 genes significantly predicted a poorer prognosis in patients with OSCC. Nominal p-value was calculated by log-rank test.
Ijms 23 09154 g003
Figure 4. The genome browser view show the H3K27ac signals in OSCC cell lines. (AD) Specific H3K27ac signals in Cmab-LTE cell lines are shown in 4 chromosomal regions located around the C9orf89, CENPA, PISD, and TRAF2 genes. SEs characterized by H3K27ac following cetuximab exposure in HSC-3 and SAS lines based on the UCSC genome browser. MACS-peak regions/intervals are represented in orange bars and are stitched together to generate SEs if their distance is <12.5 kb. The top 5% stitched regions are designated SEs and shown by red bars. (A) C9orf89 (chr9:93,096,217–93,113,283), (B) CENPA (chr2:26,786,014–26,794,589), (C) PISD (chr22:31,618,491–31,662,564), and (D) TRAF2 (chr9:136,881,933–136,926,621) loci are indicated in green bars. The blue peaks in the top track are the bigWIG data.
Figure 4. The genome browser view show the H3K27ac signals in OSCC cell lines. (AD) Specific H3K27ac signals in Cmab-LTE cell lines are shown in 4 chromosomal regions located around the C9orf89, CENPA, PISD, and TRAF2 genes. SEs characterized by H3K27ac following cetuximab exposure in HSC-3 and SAS lines based on the UCSC genome browser. MACS-peak regions/intervals are represented in orange bars and are stitched together to generate SEs if their distance is <12.5 kb. The top 5% stitched regions are designated SEs and shown by red bars. (A) C9orf89 (chr9:93,096,217–93,113,283), (B) CENPA (chr2:26,786,014–26,794,589), (C) PISD (chr22:31,618,491–31,662,564), and (D) TRAF2 (chr9:136,881,933–136,926,621) loci are indicated in green bars. The blue peaks in the top track are the bigWIG data.
Ijms 23 09154 g004
Figure 5. Expression levels of 4 genes (C9orf89, CENPA, PISD, and TRAF2) in OSCC cell lines after prolonged cetuximab exposure. The expression levels of 4 genes (C9orf89, CENPA, PISD, and TRAF2) were increased by the cetuximab treatment compared with the parental cells. Gene expression was measured by SYBR Green Real-time PCR methods. GAPDH was used as an internal control.
Figure 5. Expression levels of 4 genes (C9orf89, CENPA, PISD, and TRAF2) in OSCC cell lines after prolonged cetuximab exposure. The expression levels of 4 genes (C9orf89, CENPA, PISD, and TRAF2) were increased by the cetuximab treatment compared with the parental cells. Gene expression was measured by SYBR Green Real-time PCR methods. GAPDH was used as an internal control.
Ijms 23 09154 g005
Figure 6. Alterations of mRNA expression levels of 4 genes (C9orf89, CENPA, PISD and TRAF2) in OSCC clinical specimens based on TCGA-OSCC analyses. (A) Oncoprint of TCGA-OSCC on cBioPortal filtered by the mRNA expression (Z score ≥ 0) from the query for 4 genes. (B) Mutual exclusivity of 4 genes. q-value was derived from the Benjamini–Hochberg FDR correction procedure. Odds ratio shows how strongly the presence or absence of alterations in one are associated with the presence or absence of alterations in another in the selected samples. (C) Kaplan–Meier curves of overall survivals between altered and non-altered group (MST: median survival time). (D) Bar chart and enrichment plots of GSEA of alteration of 4 genes (FDR: false discovery rate).
Figure 6. Alterations of mRNA expression levels of 4 genes (C9orf89, CENPA, PISD and TRAF2) in OSCC clinical specimens based on TCGA-OSCC analyses. (A) Oncoprint of TCGA-OSCC on cBioPortal filtered by the mRNA expression (Z score ≥ 0) from the query for 4 genes. (B) Mutual exclusivity of 4 genes. q-value was derived from the Benjamini–Hochberg FDR correction procedure. Odds ratio shows how strongly the presence or absence of alterations in one are associated with the presence or absence of alterations in another in the selected samples. (C) Kaplan–Meier curves of overall survivals between altered and non-altered group (MST: median survival time). (D) Bar chart and enrichment plots of GSEA of alteration of 4 genes (FDR: false discovery rate).
Ijms 23 09154 g006
Figure 7. Protein expression of C9orf89, CENPA, PISD, and TRAF2 in OSCC clinical specimens according to the Protein Atlas database. Protein expression of C9orf89, CENPA, PISD and TRAF2 in OSCC clinical specimens is based on the Protein Atlas database. C9orf89: Both antibodies showed negative staining in normal tissues. In cancer tissues, HPA010921 showed weak immunoreactivity in the cytoplasm whereas HPA038297 showed strong immunoreactivity. CENPA: CAB008371 displayed strong nucleic positivity in both normal and cancer tissues, whereas weak cytoplasmic positivity was shown in both cancer tissues. PISD: Both antibodies strongly stained cancer cytoplasm whereas HPA031090 stained normal epithelium as well. TRAF2: Both antibodies stained cancer cytoplasm strongly whereas HPA0099972 weakly stained basal cells in normal epithelium.
Figure 7. Protein expression of C9orf89, CENPA, PISD, and TRAF2 in OSCC clinical specimens according to the Protein Atlas database. Protein expression of C9orf89, CENPA, PISD and TRAF2 in OSCC clinical specimens is based on the Protein Atlas database. C9orf89: Both antibodies showed negative staining in normal tissues. In cancer tissues, HPA010921 showed weak immunoreactivity in the cytoplasm whereas HPA038297 showed strong immunoreactivity. CENPA: CAB008371 displayed strong nucleic positivity in both normal and cancer tissues, whereas weak cytoplasmic positivity was shown in both cancer tissues. PISD: Both antibodies strongly stained cancer cytoplasm whereas HPA031090 stained normal epithelium as well. TRAF2: Both antibodies stained cancer cytoplasm strongly whereas HPA0099972 weakly stained basal cells in normal epithelium.
Ijms 23 09154 g007
Table 1. The detailed information of the genes in the super-enhancer regions.
Table 1. The detailed information of the genes in the super-enhancer regions.
No.Merged
Region
ChromosomeStartEndLengthGene ListPosition
1313,453,3523,483,69730,345PRDM16, ARHGEF16downstream, in gene
235131,689,43831,715,64026,202COL16A1, LOC101929444,
BAI(ADGRB2)
in gene, downstream,
downstream
352146,170,38246,203,75733,375PIK3R3, LOC105378695, TSPAN1, POMGNT1, LURAP1upstream, upstream, in gene,
downstream, upstream
4891151,536,650151,553,90817,258CGN, TUFT1, MIR554downstream, in gene, upstream
51211180,501,359180,533,62232,263ACBD6upstream
61311200,885,778200,903,11917,341GPR25, C1orf106, MROH3Pdownstream, in gene, upstream
71351202,566,931202,600,28333,352PPP1R12B, SYT2, LOC105371686, LOC105371685in gene, downstream, upstream,
upstream
81661240,758,392240,811,76153,369LOC100506929, RGS7, LOC105373229upstream, in gene, upstream
92081072,243,61172,278,76935,158ANAPC16downstream
102121075,207,30075,264,01456,714VDAC2, COMTD1downstream, in gene
1124810132,395,732132,422,98627,254LRRC27, PWWP2B, LOC105378568, C10orf91downstream, in gene, downstream,
upstream
12254118,806,9998,841,14734,148ST5, LOC102724784, RNA5SP330in gene, downstream, upstream
132591112,788,05412,842,69654,642TEAD1in gene
142841163,559,61863,584,34824,730RARRES3, HRASLS2, PLA2G16, LOC105369335downstream, upstream, downstream,
upstream
152891165,371,02765,392,67521,648TIGD3, SLC25A45downstream, in gene
1631511114,280,212114,309,04728,835NNMTupstream
173901247,811,91447,836,61424,700LOC105369749upstream
184291279,545,48579,567,43821,953PAWRdownstream
1946012122,696,516122,727,56331,047HCAR2, HCAR3, HCAR1upstream, downstream, downstream
204711333,118,13133,129,38311,252STARD13in gene
214821379,480,69079,494,90614,216NDFIP2-AS1, NDFIP2upstream, in gene
225021422,588,16222,622,51734,355DAD1, ABHD4upstream, in gene
235851573,973,34173,997,59324,252STOML1, PMLin gene, upstream
246331668,731,54068,802,32670,786CDH1downstream
256411681,559,34381,602,11342,770MIR6504in gene, upstream
266721717,900,20717,972,35972,152TOM1L2, LRRC48, ATPAF2in gene, upstream, downstream
276741719,706,61319,729,96223,349SLC47A2, ALDH3A1in gene, downstream
286991742,658,91542,683,17624,261HMGB3P27, TUBG2, PLEKHH3,
CCR10, CNTNAP1, EZH1, MIR6780A
downstream, downstream, in gene,
downstream, upstream, downstream,
downstream
297461782,096,03082,108,20812,178FASNupstream
307641857,770,62057,846,98276,362ATP8B1, LOC1 + G3305376870, RSL24D1P11upstream, downstream,
upstream
31775192,523,7622,555,59331,831LOC101929097, GNG7upstream, in gene
32776194,367,9444,403,86735,923MPND, SH3GL1, CHAF1Adownstream, in gene, upstream
33778196,719,4226,747,51228,090C3, GPR108, MIR6791, TRIP10,
SH2D3A
upstream, in gene, downstream,
upstream, downstream
347901918,361,76918,388,01726,248PGPEP1, GDF15, MIR3189,
LRRC25
downstream, upstream, upstream, downstream
357971938,251,59538,320,09068,495PPP1R14A, SPINT2, YIF1B,
C19orf33, KCNK6
upstream, in gene, downstream,
upstream, upstream
368111943,104,46943,131,73527,266PSG5, PSG2in gene, upstream
378171946,191,36946,232,92241,553IGFL2, LOC105372424, LOC645553, LOC105372423, LOC105372422, IGFL1downstream, upstream, in gene,
downstream, downstream, upstream
38840226,755,01926,773,10418,085C2orf18(SLC35F6), CENPAupstream, upstream
39849236,476,17436,505,31529,141CRIMin gene
40905285,237,32885,298,81061,482TCF7L1, LOC102724579, LOC105374839in gene, downstream, downstream
419952010,653,95210,675,73321,781JAG1, MIR6870, LOC105372526in gene, upstream, upstream
429982019,903,48519,958,41854,933RIN2in gene
4310812138,898,23638,926,38428,148LOC400867in gene
4410842141,751,74741,787,73335,986RIPK4, MIR6814, LOC102724800,
PRDM15
upstream, upstream, in gene,
downstream
4511012224,950,10424,997,09846,994TMEM211, KIAA1671upstream, in gene
4611112231,629,89931,663,55933,660SFI1, PISD, MIR7109,
PRR14L
downstream, in gene, upstream,
downstream
4711212237,887,25037,908,09520,845EIF3L, MICALL1downstream, upstream
4811272240,482,36840,542,41160,043MKL1, LOC101927257, LOC105373037in gene, upstream, upstream
4911352246,731,87346,775,78843,915CERK, LOC105373077, TBC1D22Aupstream, upstream, upstream
501155337,934,27637,947,92313,647CTDSPL, MIR26A1in gene, upstream
5111893123,583,773123,653,83170,058HACD2, MYLK-AS1upstream, in gene
5212073153,130,215153,165,19534,980RAP2Bupstream
5312233183,253,290183,297,85244,562MCF2L2, B3GNT5, RNA5SP151in gene, downstream, upstream
5412373197,482,394197,521,06738,673LOC105374308, LOC105374309, BDH1upstream, downstream, downstream
551299557,681,78657,700,89119,105LOC101928505downstream
561392633,731,25033,789,20357,953C6orf125(UQCC2), IP6K3, LEMD2, LOC105375024, MLNupstream, upstream, downstream,
upstream, downstream
571473727,080,27627,115,99735,721HOXA1, HOTAIRM1, HOXA2, LOC105375205upstream, in gene, downstream,
upstream
581475728,034,60528,067,08632,481JAZF1, LOC105375208in gene, in gene
591496747,633,32047,694,06560,745LINC01447, C7orf65downstream, downstream
601555822,561,11622,605,49744,381PPP3CC, SORBS3, LOC105379320, PDLIM2, C8orf58, CCAR2, BIN3downstream, downstream, upstream,
in gene, upstream, upstream,
downstream
6116348140,722,495140,734,72712,232MIR151Adownstream
6216408142,777,613142,796,02518,412LYNX1, LY6Dupstream, upstream
631655922,079,70622,119,69339,987CDKN2B-AS1in gene
641672993,093,99093,149,53955,549SUSD3, LOC101927993, C9orf89, NINJ1, LOC105376150downstream, upstream, downstream, in gene, upstream
6516779106,860,435106,921,63961,204LOC105376204, ZNF462upstream, in gene
6617039129,314,856129,336,83121,975C9orf106, LINC01503downstream, upstream
6717089136,533,550136,579,45745,907NOTCH1, MIR4673, LOC1053763204, MIR4674, LINC01573upstream, upstream, downstream,
upstream, downstream
6817099136,881,365136,905,10823,743MAMDC4, EDF1, LOC105376326,
TRAF2, MIR4479
downstream, upstream, upstream,
in gene, downstream
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Saito, T.; Asai, S.; Tanaka, N.; Nohata, N.; Minemura, C.; Koma, A.; Kikkawa, N.; Kasamatsu, A.; Hanazawa, T.; Uzawa, K.; et al. Genome-Wide Super-Enhancer-Based Analysis: Identification of Prognostic Genes in Oral Squamous Cell Carcinoma. Int. J. Mol. Sci. 2022, 23, 9154. https://doi.org/10.3390/ijms23169154

AMA Style

Saito T, Asai S, Tanaka N, Nohata N, Minemura C, Koma A, Kikkawa N, Kasamatsu A, Hanazawa T, Uzawa K, et al. Genome-Wide Super-Enhancer-Based Analysis: Identification of Prognostic Genes in Oral Squamous Cell Carcinoma. International Journal of Molecular Sciences. 2022; 23(16):9154. https://doi.org/10.3390/ijms23169154

Chicago/Turabian Style

Saito, Tomoaki, Shunichi Asai, Nozomi Tanaka, Nijiro Nohata, Chikashi Minemura, Ayaka Koma, Naoko Kikkawa, Atsushi Kasamatsu, Toyoyuki Hanazawa, Katsuhiro Uzawa, and et al. 2022. "Genome-Wide Super-Enhancer-Based Analysis: Identification of Prognostic Genes in Oral Squamous Cell Carcinoma" International Journal of Molecular Sciences 23, no. 16: 9154. https://doi.org/10.3390/ijms23169154

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop