Startseite Medizin SET domain containing protein 8 (SET8) promotes tumour progression and indicates poor prognosis in patients with laryngeal squamous cell carcinoma
Artikel Open Access

SET domain containing protein 8 (SET8) promotes tumour progression and indicates poor prognosis in patients with laryngeal squamous cell carcinoma

  • Li-Li Lan , Sheng-Hui Liu ORCID logo , Zhi-Tao Fan , Xue-Xia Wang , Jing-Tian Wang , Ke-Xin Wang und Rui-Li Zhao EMAIL logo
Veröffentlicht/Copyright: 22. Februar 2023
Oncologie
Aus der Zeitschrift Oncologie Band 25 Heft 1

Abstract

Objectives

SET Domain Containing Protein 8 (SET8), a member of the SET domain containing methyltransferase family involved in several biological processes and SET8 expression levels, reportedly affects the outcomes of patients with breast cancer, renal cancer, prostate carcinoma, and oesophageal squamous cell carcinoma. However, there have been no relevant studies on the biofunction and use of SET8 expression in the prediction of laryngeal squamous cell carcinoma (LSCC) outcomes.

Methods

In our study, SET8 expression levels were detected using immunohistochemical staining, western blotting, and quantitative real-time RT-PCR (qRT-PCR) with semi-quantitative analysis for laryngeal cancer outcomes. Additionally, we assessed the influence of SET8 on the behaviour of laryngeal cancer cells in vitro, using cell counting kit-8, clone formation, wound healing, and Transwell invasion assays. We subsequently performed qRT-PCR and western blotting for an in-depth study of SET8.

Results

Our study showed marked upregulation of SET8 in tumour tissues and laryngeal cancer cell lines. High SET8 expression predicts poor prognosis in patients with LSCC, and its expression can be used as an independent predictor of LSCC outcome. Subsequent functional analyses indicated that SET8 knockdown exerted an inhibitory effect on proliferation, migration, and invasiveness in vitro.

Conclusions

SET8 may be associated with epithelial-mesenchymal transition. Our results demonstrate that higher SET8 expression is an unfavourable prognostic predictor and exerts tumour-promoting effects in LSCC.

Introduction

Laryngeal carcinoma is the second most common head and neck tumour, with approximately 12,410 new cases and 3,760 deaths reported by the National Centre for Health Statistics in 2018 [1]. Over 95% of laryngeal cancers are squamous cell carcinomas, with subtypes including supraglottic, glottic, and subglottic. Despite remarkable advances in conventional treatments, the overall prognosis of laryngeal squamous cell carcinoma (LSCC) remains unsatisfactory due to the high incidence of metastasis and postoperative recurrence [2]. Thus, there is an urgent need to find valuable molecular markers for screening, risk assessment, and prognosis judgment of LSCC in high-risk individuals.

Different histone lysine methylation-mediated methyltransferases are associated with both transcriptional activation and repression [3]. SET8 (also known as KMT5a or SETD8) belongs to the SET domain-containing methyltransferase family that is specific to monomethylates histone H4K20, which is involved in numerous of essential physiological processes, including gene transcription regulation, regulation of replication origin, chromosome formation, maintenance of genome integrity, and participation in the process and development of the cell cycle [4], [5], [6], [7].

SET8 modifies the outcomes of hepatocellular carcinoma, gastric cancer, and oesophageal squamous cell carcinoma (ESCC) [8], [9], [10]. Anatomically, laryngeal carcinoma and cervical oesophageal carcinoma are carcinomas of the head and neck, respectively. Histologically, laryngeal carcinomas and oesophageal malignancies are both squamous cell carcinomas. Therefore, our aim was to determine the impact of SET8 expression on progression and prognosis of LSCC proximate to the oesophagus, anatomically.

Materials and methods

Clinical sample acquisition and processing

A total of 72 LSCC tumour specimens and corresponding adjacent normal tissues were obtained from patients who underwent surgical procedure, but not chemotherapy, at the Fourth Hospital of Hebei Medical University between 2014 and 2015. We obtained ethics approval from the Ethics Committee of the Fourth Hospital of Hebei Medical University (Ethic Committee NO. 2021169) in accordance with the Declaration of Helsinki (as revised in 2013), and informed consent was obtained from all patients. Tissue samples were immediately collected using RNA Latter (ComWin, Beijing, China) for subsequent RNA extraction, and the remaining samples were fixed with formalin for paraffin sectioning. Detailed patient clinical data were obtained from the medical records. Tumour stages were evaluated accordingance to the American Joint Committee on Cancer 8th edition on cancer tumour, node, metastasis (TNM) classification for LSCC [11].

Assessment of SET8 protein expression in LSCC tissues using immunohistochemical (IHC) staining

Paraffin sections, 5-μm thick, were sequentially incubated with primary monoclonal SET8 antibodies (Abcam, Cambridge, UK, dilution 1:100) overnight at 4 °C. A secondary polymerised horseradish peroxidase antibody was then added at 37 °C for 1 h, followed by further incubation with streptavidin-peroxidase. Immunohistochemically stained slides were evaluated simultaneously by two experienced pathologists with no knowledge of the clinicopathological information and outcome data. The formula H-score=(i + 1) π was used for semi-quantified assessment, and i was set as the staining intensity (negative, weak, moderate, and strong were recorded as 0, 1, 2, and 3, respectively). π represents the weighted intensity of staining, which varied between 0 and 100%. These two values were multiplied to produce the H-score. The high expression group was characterised by scores greater than 100%, and the low expression group by scores of 100% or less.

Evaluation of SET8 protein expression differences in LSCC cells and normal tissues using western blotting

LSCC cell lines (AMC-HN-8 and TU177) and five cases of normal tissue protein extracts were lysed using a radioimmunoprecipitation assay lysis buffer (Elabscience Biotechnology Co., Ltd., Wuhan, China) and subjected to ultrasonic shaking. Each sample was adjusted for equal amounts and masses and fractionated using 10% SDS-polyacrylamide gel electrophoresis. The transferred membranes were blocked with 5% milk for 1 h and probed with anti-SET8 antibodies (Abcam, Cambridge, UK, dilution 1:1,000). The bands were visualised using ECL (Sangon Biotech Co., Ltd., Shanghai, China), and an anti-β-actin antibody (Abcam, Cambridge, UK) was used as a control.

Cell culture and gene transfection

Human LSCC cell lines, TU177 and AMC-HN-8, were purchased from BNBIO (Beijing, China). TU177 cells were cultured in a RPMI-1,640 medium (GIBCO, NY, USA), whereas AMC-HN-8 cells were grown in Dulbecco’s Modified Eagle’s medium (DMEM; Invitrogen, CA, USA). Both were supplemented with 10% foetal bovine serum (FBS; Invitrogen, CA, USA). Knock down of SET8 was accomplished by small interference RNA (si-RNA). Based on our previous research, we chose to use SET8-siRNA (GeneCopoeia, Rockville, MD; the sequence was GAAUGAAGAUUGACCUCAUCG) which dramatically reduced the protein expression levels of SET8 [10]. Psi-H1-SET8siRNA and psi-H1 plasmids were transfected into human LSCC cell lines using Lipofectamine 2,000 (Thermo Fisher, Shanghai, China) according to the instructions. Cells transfected with the Psi-H1-SET8siRNA were designated as the SET8-siRNA group, negative control siRNA (unspecific RNA interference, scrambled sequence) was used as the control, and untransfected LSCC cells were used as the blank control.

Extraction of total RNA and qRT-PCR

Total RNA was extracted from LSCC cells and 20 cases of corresponding adjacent normal tissues according to manufacturer instructions and reversed transcribed to cDNA using the Transcriptor First Strand cDNA Synthesis Kit (Roche, Vaud, Switzerland). Synthetic cDNA was used as a template and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an internal reference for qRT-PCR amplification. SET8 primers (Shanghai Sangon Biotech Co., Ltd., Shanghai, China) sequences were listed as follows: forward: 5′- CGCAAACTTACGGATTTCT -3′; reverse: 5′- CGATGAGGTCAATCTTCATT - 3′. qRT-PCR was performed as follows: 95 °C for 10 min; 40 cycles at 95 °C for 15 s, 59 °C for 30 s, and 72 °C for 30 s. The relative expression of SET8 was quantified using 2−△△Ct. The experiments were assayed in triplicate.

In vitro functional assay

Cell proliferation and viability assays

The protein in two transfected LSCC cells was extracted to detect the efficiency of transfection. The experimental steps were the same as in 2.3. The effect of SET8 on LSCC cell line proliferation was evaluated using cell counting kit-8 (CCK-8; Dojindo Lab, Kumamoto, Japan) and clone formation assays. Transfected cells were inoculated into 96-well plates with the density of 2 × 103 cells/well. 20 µL CCK-8 solution were added to the cultured cells and incubated in a CO2 incubator at 37 °C for 2 h. Optical density was examined at a wavelength of 490 nm with a multi-well plate reader. Each experiment was triplicated. Clone formation assay was performed as follows: transfected cells with a density of 3,000 cells per well were seeded into 6-well plates and cultured for 10 days. The plates were fixed in 4% paraformaldehyde for 20 min and stained with 1% crystal violet for 20 min. The clone formation rate was calculated under a microscope; more than 50 cell clones were considered as one colony formation.

Wound-healing assay

The effect of SET8 on LSCC cell migration was assessed using a wound-healing assay. The transfected cells were cultured in 6-well plates and incubated in a tissue culture incubator for 24 h. A straight linear wound was created on the cell layer using a sterile pipette tip when the cells reached 100% confluency. Cells were then incubated in a straight linear area at 37 °C for 24 h. Photographs were taken using a light microscope 0, 12, and 24 h after scratching. Quintuplicates were performed for each scratch assay using ImageJ software.

In vitro invasion assays

The effect of SET8 on LSCC cell line invasiveness was tested with Transwell assay (Corning Costar, NY, USA). Transwell chambers were pre-coated with Matrigel (Solarbio, Beijing, China) prior to the invasion experiment. The transfected cells were placed in the upper chambers filled with 100 µL of FBS-free culture medium at a density of 1 × 105 cells/well. The subjacent chambers were supplemented with a medium containing 10% FBS. The cells were then incubated in a tissue culture incubator for 48 h. The invasive cells in the upper compartments were fixed with paraformaldehyde after incubation and stained with crystal violet. Cells that migrated through the membrane were counted in five randomly selected microscopy fields.

qRT-PCR and western blotting assays were performed to evaluate the mRNA and protein expression levels of EMT markers

qRT-PCR and western blotting assays were used to assess the mRNA and protein expression levels of E-cadherin, N-cadherin, and vimentin in AMC-HN-8 and TU177 cells with SET8 knock down. The primer sequences were listed as follows: E-cadherin: forward: 5′-CGAGAGCTACACGTTCACGG-3′; reverse: 5′- GGCCTTTTGACTGTAATCACACC – 3′, annealing at 59 °C; N-cadherin: forward: 5′-CAACTTGCCAGAAAACTCCAGG-3′; reverse: 5′-ATGAAACCGGGCTATCTGCTC – 3′, annealing at 59 °C; N-cadherin: forward: 5′-TCCACACGCACCTACAGTCT-3′; reverse: 5′-CCGAGGACCGGGTCACATA – 3′, annealing at 57 °C. Relative expression levels were calculated using the 2−ΔΔCt method. The remaining experimental steps were performed as described above (2.3 and 2.5).

Statistical analysis

Data were analyzed via SPSS 22.0 software and GraphPad Prism 7.0. The Kaplan-Meier method was employed to calculate survival curves, which were compared using the log-rank test. Multivariate survival analysis was performed using a Cox regression model. Student’s t-test and the chi-square test were used for comparisons of different groups. All statistical tests were 2-sided, and p<0.05 was taken as a statistically significant.

Results

SET8 expression was upregulated in LSCC tissues and cell lines, and a higher SET8 protein expression predicted poor prognosis in patients with LSCC

To evaluate SET8 expression in LSCC, IHC, western blotting, and qRT-PCR were performed in LSCC tissues, matched adjacent normal tissues, and cell lines. SET8-positive staining was mainly located in the nucleus with a weaker co-expression in the cytomembrane and cytoplasm. As shown in Figure 1A and B, SET8 proteins were expressed in LSCC tissues and showed absent or very low expression in matched adjacent normal tissues. The results of further correlation analysis revealed that upregulated SET8 expression was correlated with TNM stages in LSCC tissues (p=0.021), with a tendency toward increased SET8 expression at higher tumour stages (Table 1). SET8 expression was not correlated with age (p=0.953), smoking (p=0.133), pathological differentiation (p=0.352), and lymph node metastasis (p=0.055). Western blotting (Figure 1C) analysis confirmed elevated SET8 protein expression in LSCC cell lines and low SET8 protein expression in normal tissues. Consistent with IHC and western blotting results, qRT-PCR analysis showed elevated SET8 level expression in LSCC cells (Figure 1D). The “Pool” was defined as the mean expression level of SET8mRNA in 20 adjacent normal tissues. These results indicate that abnormally high SET8 expression may be vital to tumour progression.

Figure 1: 
SET8 protein expression in LSCC. (A) SET8 protein in LSCC using IHC (SP Left × 200; Right × 400). a: High expression in LSCC tissues; b: Low expression in adjacent normal tissues. (B) SET8 expression analysis in LSCC tissues and matched adjacent normal tissues (p<0.01; n=72). (C) The protein expression levels of SET8 were elevated in LSCC cells compared with normal tissue samples. (D) qRT-PCR analysis showing that the expression level of SET8 was significantly upregulated in LSCC cells compared with the “Pool” (p<0.01). (E) Kaplan-Meier curves for overall survival stratified by high and low expression of SET8 in patients with LSCC. Log-rank analysis shows that patients with over-expression of SET8 had significantly poorer survival than those with low expression (p=0.019).
Figure 1:

SET8 protein expression in LSCC. (A) SET8 protein in LSCC using IHC (SP Left × 200; Right × 400). a: High expression in LSCC tissues; b: Low expression in adjacent normal tissues. (B) SET8 expression analysis in LSCC tissues and matched adjacent normal tissues (p<0.01; n=72). (C) The protein expression levels of SET8 were elevated in LSCC cells compared with normal tissue samples. (D) qRT-PCR analysis showing that the expression level of SET8 was significantly upregulated in LSCC cells compared with the “Pool” (p<0.01). (E) Kaplan-Meier curves for overall survival stratified by high and low expression of SET8 in patients with LSCC. Log-rank analysis shows that patients with over-expression of SET8 had significantly poorer survival than those with low expression (p=0.019).

Table 1:

Expression of SET8 in LSCC and its relationship with clinicopathological features.

Characteristics n Expression level of SET8
High (%) χ2 p-Value
Age, years
 <60 32 23 (71.9) 0.003 0.953
 ≥60 40 29 (72.5)

Smoking

 No 20 17 (85) 2.254 0.133
 Yes 52 35 (67.3)

TNM stage

 Ⅰ + Ⅱ 42 26 (61.9) 5.349 0.021
 Ⅲ + Ⅳ 30 26 (86.7)

Lymph node metastasis

 N0 58 39 (67.2) 3.689 0.055
 N1/2/3 14 13 (92.9)

Pathological differentiation

 Well/moderate 48 33 (68.8) 0.865 0.352
 Poor 24 19 (79.2)

A total of 72 patients were enrolled for survival analysis based on their SET8 expression status. All patients were followed up every 6 months for longer than 60 months; five (6.9%) were lost to follow-up and 67 patients were enrolled in the survival analysis. Survival were analyzed with the Kaplan-Meier method and the log-rank test. Univariate analysis revealed that SET8 expression and pathological differentiation were associated with overall survival in patients with LSCC (Table 2). As shown in Figure 1E, patients with elevated SET8 expression had a significantly shorter overall survival time than those with low expression in the log-rank test analysis. The Cox proportional hazards model revealed that SET8 expression was an independent prognostic factor for survival in patients with LSCC (relative risk=3.713; 95% confidence interval [CI]=1.099–12.669; p=0.035). TNM stage was also identified as a powerful independent predictor for LSCC clinical outcomes (relative risk=0.372; 95% CI=0.161–0.858; p=0.020). These results confirmed that high protein expression of SET8 was an unfavourable prognostic factor for LSCC outcomes (Table 3).

Table 2:

Univariate analysis of SET8 expression and clinical characteristics associated with overall survival in LSCC patients.

Characteristics n 5-year survival rate, % p-Value
SET8 expression
 High expression 50 25 (50.0) 0.019
 Low expression 17 14 (82.4)

Age, years

 <60 30 18 (60.0) 0.619
 ≥60 37 21 (56.8)

Smoking

 No 17 12 (70.6) 0.321
 Yes 50 27 (54.0)

TNM stage

 Ⅰ + Ⅱ 41 24 (58.5) 0.922
 Ⅲ + Ⅳ 26 15 (57.7)

Lymph node metastasis

 N0 55 33 (60.0) 0.630
 N1/2/3 12 6 (50.0)

Pathological differentiation

 Well/moderate 52 33 (63.5) 0.049
 Poor 15 6 (40.0)
Table 3:

Multivariate analysis of prognostic factors associated with post operational survival in patients with LSCC with Cox proportional hazards model.

Factors Relative risk 95% confidence interval p-Value
Age 0.964 0.426–2.182 0.929
Smoking 0.670 0.253–1.777 0.421
TNM stage 0.372 0.161–0.858 0.020
Expression of SET8 3.731 1.099–12.669 0.035
Lymph node metastasis 0.748 0.270–2.074 0.577
Pathological differentiation 0.507 0.210–1.223 0.131

Downregulation of SET8 suppressed LSCC cell proliferation in vitro

Psi-H1-SET8siRNA knockdown was verified using qRT-PCR and western blotting assays. qRT-PCR and western blotting results revealed that SET8 expression was markedly lower in Psi-H1-SET8siRNA transfected cells (Figure 2A, D). CCK-8 and clone formation assays indicated that SET8 knockdown markedly suppressed the proliferation capabilities of LSCC cell lines compared to those of the blank control cells and control-siRNA cell groups (Figure 2E, H).

Figure 2: 
Downregulation of SET8 expression suppresses proliferation of LSCC cells in vitro. (A–B) SET8 mRNA expression is markedly lower in SET8-siRNA transfected cells than in the blank control and control-siRNA groups. (C–D) Western blotting showing SET8 protein expression is markedly lower in SET8-siRNA transfected cells than in the blank control and control-siRNA groups. (E–H) CCK-8 and clone formation assays showing that down-regulation of SET8 expression inhibits LSCC cell proliferation ability. *p<0.05, **p<0.01.
Figure 2:

Downregulation of SET8 expression suppresses proliferation of LSCC cells in vitro. (A–B) SET8 mRNA expression is markedly lower in SET8-siRNA transfected cells than in the blank control and control-siRNA groups. (C–D) Western blotting showing SET8 protein expression is markedly lower in SET8-siRNA transfected cells than in the blank control and control-siRNA groups. (E–H) CCK-8 and clone formation assays showing that down-regulation of SET8 expression inhibits LSCC cell proliferation ability. *p<0.05, **p<0.01.

Downregulation of SET8 inhibited migration and invasion of LSCC cells in vitro

Wound-healing assays were used to assess migration abilities, and their results showed that the migration ratio was significantly impeded by SET8-siRNA in both SET8-siRNA transfected cell lines compared with that in the blank control and control-siRNA groups (Figure 3A, D). The in vitro invasion assay revealed that SET8 knockdown significantly suppressed LSCC cell invasiveness (Figure 3E and F). Overall, SET8 knockdown significantly suppressed the malignant biological features of LSCC cells.

Figure 3: 
Downregulation of SET8 inhibited migration and invasion of LSCC cells in vitro. (A) Wound-healing assay showing that the downregulation of SET8 expression restrains the migration ability of TU177; (B) Quantification of results from A. (C) Wound-healing assay showing that SET8 knockdown inhibits migration ability of AMC-HN-8; (D) Quantification of results from C. (E) Transwell assay demonstrating that downregulation of SET8 expression inhibits invasion ability of TU177; (F) Transwell assay showing that downregulation of SET8 expression inhibits invasion ability of AMC-HN-8; *p<0.05, **p<0.01.
Figure 3:

Downregulation of SET8 inhibited migration and invasion of LSCC cells in vitro. (A) Wound-healing assay showing that the downregulation of SET8 expression restrains the migration ability of TU177; (B) Quantification of results from A. (C) Wound-healing assay showing that SET8 knockdown inhibits migration ability of AMC-HN-8; (D) Quantification of results from C. (E) Transwell assay demonstrating that downregulation of SET8 expression inhibits invasion ability of TU177; (F) Transwell assay showing that downregulation of SET8 expression inhibits invasion ability of AMC-HN-8; *p<0.05, **p<0.01.

SET8 knockdown suppressed the EMT process

To explore the mechanism of SET8 in LSCC, we assessed whether SET8 knockdown affected EMT. SET8 knockdown upregulated the mRNA and protein expression level of E-cadherin and downregulated N-cadherin and vimentin in AMC-HN-8 and TU177, as detected using qRT-PCR and western blotting methods (Figure 4A, D, p<0.05). These findings demonstrated that higher SET8 exerts tumour-promoting effects via regulating EMT in LSCC.

Figure 4: 
SET8 knockdown suppresses EMT progress in LSCC cells. (A–B) Relative mRNA and protein expression levels of E-cadherin, N-cadherin, and vimentin in AMC-HN-8 cell transfected with control-siRNA or SET8-siRNA vectors. (C–D) Relative mRNA and protein expression levels of E-cadherin, N-cadherin, and vimentin in TU177 cell transfected with control-siRNA or SET8-siRNA vectors. Data are expressed as the mean ± SD; *p<0.05, **p<0.01.
Figure 4:

SET8 knockdown suppresses EMT progress in LSCC cells. (A–B) Relative mRNA and protein expression levels of E-cadherin, N-cadherin, and vimentin in AMC-HN-8 cell transfected with control-siRNA or SET8-siRNA vectors. (C–D) Relative mRNA and protein expression levels of E-cadherin, N-cadherin, and vimentin in TU177 cell transfected with control-siRNA or SET8-siRNA vectors. Data are expressed as the mean ± SD; *p<0.05, **p<0.01.

Discussion

The pivotal roles of histone post-translational modifications, especially histone lysine methylation, have attracted the attention of several investigators in the last decade [12]. Histone lysine methylation has been implicated in diverse biological processes, such as DNA damage repair, transcription regulation, pathogenesis, progression, and metastasis in most tumours [13, 14]. H4K20 methyltransferase SET8 is a crucial enzyme for the epigenetic modification of histone lysine methylation. Considerable evidence implicates that SET8 is upregulated in various tumours, such as hepatocellular carcinoma, ovarian cancer, and lung, breast, and oesophageal cancers [8, 10, 15], [16], [17]. Based on the previous finding that SET8 modified ESCC outcomes, we aimed to uncover the roles of SET8 in LSCC due to its histological similarity and anatomical proximity to ESCC. Our studies are the first to demonstrate the tumour-promoting effects of SET8 in LSCC. Elevated SET8 expression was found in LSCC tissues and cell lines and predicted poor prognosis in patients with LSCC. This was similar to previous studies data, which have indicated that patients with higher SET8 expression suffered poor outcomes in multiple malignancies [9, 10, 18].

Many recent studies have revealed the vital role of SET8 in tumour progression by participating in tumour cell growth and metastasis. SET8 methylates non-histones of the tumour suppressor p53 at the 382-lysine site to modulate cell cycle arrest and apoptosis and modify cancer development [17]. Studies have demonstrated that SET8 can prevent p53 stabilisation and accelerate p53 degradation by mediating the methylation of Numb, thereby mediating the apoptosis of cancer cells [19]. In addition to methylating p53, SET8 contributes breast cancer cells metastasis by inducing the methylation of Twist target genes in breast tumours [20]. A recent study reached a similar conclusion that patients with breast cancer with high levels of SET8 were more likely to exhibit a higher rate of metastasis [21]. To further reveal the effect of SET8 on LSCC progression, cell proliferation, migration, and invasion assays were used. We knocked down SET8 with siRNA and found that inhibition SET8 suppressed LSCC cell proliferation, migration, and invasion in vitro. Based on our findings, SET8 may contribute to the occurrence and development of LSCC as a tumour gene. Our results indicate the essential role of SET8 in the pathogenesis of LSCC and provides it as an emerging effective target for LSCC therapy.

EMT is a complicated and frequent process in cancers characterised by the reduction of epithelial characteristics and acquisition of mesenchymal polarity phenotype. EMT has been extensively studied due to its involvement in tumour evasion, apoptosis, adhesion, and metastasis. In our study, E-cadherin expression was significantly increased, whereas N-cadherin and vimentin were notably decreased in mRNA and protein levels accompanied by SET8 knockdown, indicating that SET8 was involved in LSCC progression through modulating the EMT process. These data further indicated the oncogenic role of SET8 in LSCC. These mechanisms may contribute to the SET8-related modification of tumours. Methytransferase was considered a vital therapeutic target in the epigenetic field. Drugs targeting epigenetic enzymes have been partially used clinically and accompanied with advances in targeted drug research. Those findings are likely to enrich our knowledge of targeted cancer therapy.

In conclusion, our results indicated that SET8 may modify the prognosis of patients with LSCC through its vital roles in proliferation, migration, and invasion. We also stressed the therapeutic potential of targeting SET8 for LSCC treatment. However, the findings require further research and laboratory-based functional studies for validation.


Corresponding author: Professor Rui-Li Zhao, Otolaryngology Head and Neck Surgery Department, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, Hebei 050011, P. R. China, E-mail:
Li-Li Lan and Sheng-Hui Liu contributed equally to this work.

Funding source: Medical Scientific Research of Hebei Health Commission

Award Identifier / Grant number: 20230907

  1. Research funding: This work was supported by the Medical Scientific Research of Hebei Health Commission (No. 20230907).

  2. Author contributions: The authors confirm contribution to the paper as follows: study conception and design: R.L. Zhao; administrative support: L.L. Lan, Z.T. Fan, X.X. Wang; provision of study materials or patients: L.L. Lan, S.H. Liu, J.T. Wang; collection and assembly of data: K.X. Wang; data analysis and interpretation: All authors; manuscript writing: All authors; final approval of manuscript: All authors. All authors reviewed the results and approved the final version of the manuscript.

  3. Competing interests: The authors declare that they have no conflicts of interest to report regarding the present study.

  4. Informed consent: Informed consent was obtained from all individuals included in this study.

  5. Ethical approval: This study was approved by the Ethics Committee of the Fourth Hospital of Hebei Medical University. (Shijiazhuang, China. Ethics approval number: 2021169). Written informed consent was obtained from all patients prior to the study.

References

1. Bray, F, Ferlay, J, Soerjomataram, I, Siegel, RL, Torre, LA, 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. https://doi.org/10.3322/caac.21492.Suche in Google Scholar PubMed

2. Ferlay, J, Soerjomataram, I, Dikshit, R, Eser, S, Mathers, C, Rebelo, M, et al.. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136:E359–86. https://doi.org/10.1002/ijc.29210.Suche in Google Scholar PubMed

3. Jorgensen, S, Schotta, G, Sorensen, CS. Histone H4 lysine 20 methylation: key player in epigenetic regulation of genomic integrity. Nucleic Acids Res 2013;41:2797–806. https://doi.org/10.1093/nar/gkt012.Suche in Google Scholar PubMed PubMed Central

4. Beck, DB, Oda, H, Shen, SS, Reinberg, D. PR-Set7 and H4K20me1: at the crossroads of genome integrity, cell cycle, chromosome condensation, and transcription. Genes Dev 2012;26:325–37. https://doi.org/10.1101/gad.177444.111.Suche in Google Scholar PubMed PubMed Central

5. Tardat, M, Brustel, J, Kirsh, O, Lefevbre, C, Callanan, M, Sardet, C, et al.. The histone H4 Lys 20 methyltransferase PR-Set7 regulates replication origins in mammalian cells. Nat Cell Biol 2010;12:1086–93. https://doi.org/10.1038/ncb2113.Suche in Google Scholar PubMed

6. Houston, SI, McManus, KJ, Adams, MM, Sims, JK, Carpenter, PB, Hendzel, MJ, et al.. Catalytic function of the PR-Set7 histone H4 lysine 20 monomethyltransferase is essential for mitotic entry and genomic stability. J Biol Chem 2008;283:19478–88. https://doi.org/10.1074/jbc.m710579200.Suche in Google Scholar

7. Abbas, T, Shibata, E, Park, J, Jha, S, Karnani, N, Dutta, A. CRL4(Cdt2) regulates cell proliferation and histone gene expression by targeting PR-Set7/Set8 for degradation. Mol Cell 2010;40:9–21. https://doi.org/10.1016/j.molcel.2010.09.014.Suche in Google Scholar PubMed PubMed Central

8. Chen, XY, Ding, XW, Wu, QC, Qi, J, Zhu, MM, Miao, CH. Monomethyltransferase SET8 facilitates hepatocellular carcinoma growth by enhancing aerobic glycolysis. Cell Death Dis 2019;10:312. https://doi.org/10.1038/s41419-019-1541-1.Suche in Google Scholar PubMed PubMed Central

9. Shi, XL, Guo, ZJ, Wang, XL, Liu, XL, Shi, GF. SET8 expression is associated with overall survival in gastric cancer. Genet Mol Res 2015;14:15609–15. https://doi.org/10.4238/2015.december.1.12.Suche in Google Scholar

10. Wang, CJ, Wu, JH, Zhao, Y, Guo, ZJ. miR-502 mediated histone methyltransferase SET8 expression is associated with outcome of esophageal squamous cell carcinoma. Sci Rep 2016;6:32921. https://doi.org/10.1038/srep32921.Suche in Google Scholar PubMed PubMed Central

11. Lydiatt, WM, Patel, SG, O’Sullivan, B, Brandwein, MS, Ridge, JA, Migliacci, JC, et al.. Head and neck cancers-major changes in the American Joint Committee on cancer eighth edition cancer staging manual. CA A Cancer J Clin 2017;67:122–37. https://doi.org/10.3322/caac.21389.Suche in Google Scholar PubMed

12. Black, JC, Whetstine, JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell 2012;48:491–507. https://doi.org/10.1016/j.molcel.2012.11.006.Suche in Google Scholar PubMed PubMed Central

13. Milite, C, Feoli, A, Viviano, M, Rescigno, D, Cianciulli, A, Balzano, AL, et al.. The emerging role of lysine methyltransferase SETD8 in human diseases. Clin Epigenet 2016;8:102. https://doi.org/10.1186/s13148-016-0268-4.Suche in Google Scholar PubMed PubMed Central

14. Qin, Y, Ouyang, H, Liu, J, Xie, Y. Proteome identification of proteins interacting with histone methyltransferase SET8. Acta Biochim Biophys Sin 2013;45:303–8. https://doi.org/10.1093/abbs/gmt011.Suche in Google Scholar PubMed

15. Miku, W, Asako, K, Kenbun, S, Ryuji, H, Syuzo, K, Masaaki, K, et al.. Epigenetic modifier SETD8 as a therapeutic target for high-grade serous ovarian cancer. Biomolecules 2020;10:1686. https://doi.org/10.3390/biom10121686.Suche in Google Scholar PubMed PubMed Central

16. Piao, LH, Feng, Y, Che, N, Li, MX, Li, XG, Jin, Y, et al.. SETD8 is a prognostic biomarker that contributes to stem-like cell properties in non-small cell lung cancer. Pathol Res Pract 2020;216:153258. https://doi.org/10.1016/j.prp.2020.153258.Suche in Google Scholar PubMed

17. Liu, B, Zhang, XN, Song, FJ, Zheng, H, Zhao, YR, Li, HX, et al.. MiR-502/SET8 regulatory circuit in pathobiology of breast cancer. Cancer Lett 2016;376:259–67. https://doi.org/10.1016/j.canlet.2016.04.008.Suche in Google Scholar PubMed

18. Lin, ZF, Jia, HL, Hong, L, Zheng, YH, Shao, WQ, Ren, XD, et al.. Prognostic impact of SET domain-containing protein 8 and protein arginine methyltransferase 5 in patients with hepatocellular carcinoma following curative resection. Oncol Lett 2018;16:3665–73. https://doi.org/10.3892/ol.2018.9083.Suche in Google Scholar PubMed PubMed Central

19. Dhami, GK, Liu, H, Galka, M, Voss, C, Wei, R, Muranko, K, et al.. Dynamic methylation of Numb by Set8 regulates its binding to p53 and apoptosis. Mol Cell 2013;50:565–76. https://doi.org/10.1016/j.molcel.2013.04.028.Suche in Google Scholar PubMed

20. Yang, F, Sun, LY, Li, Q, Han, X, Lei, LD, Zhang, H, et al.. SET8 promotes epithelial-mesenchymal transition and confers TWIST dual transcriptional activities. EMBO J 2012;31:110–23. https://doi.org/10.1038/emboj.2011.364.Suche in Google Scholar PubMed PubMed Central

21. Yu, N, Huangyang, PW, Yang, XH, Han, X, Yan, RR, Jia, HT, et al.. microRNA-7 suppresses the invasive potential of breast cancer cells and sensitizes cells to DNA damages by targeting histone methyltransferase SET8. J Biol Chem 2013;288:19633–42. https://doi.org/10.1074/jbc.m113.475657.Suche in Google Scholar

Received: 2022-09-14
Accepted: 2023-02-06
Published Online: 2023-02-22

© 2023 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Heruntergeladen am 26.12.2025 von https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2023-0019/html
Button zum nach oben scrollen