Open Access

Transcription factor E2F4 facilitates SUMOylation to promote HCC progression through interaction with LIN9

  • Authors:
    • Zhenwei Ma
    • Qilan Li
    • Wenjing Wang
    • Zhengdong Deng
  • View Affiliations

  • Published online on: September 2, 2024     https://doi.org/10.3892/ijo.2024.5686
  • Article Number: 98
  • Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

SUMOylation plays a crucial role in numerous cellular biological and pathophysiological processes associated with human disease; however, the mechanisms regulating the genes involved in SUMOylation remain unclear. In the present study, E2F transcription factor 4 (E2F4) was identified as an E2F member related to hepatocellular carcinoma (HCC) progression by public database analysis. It was found that E2F4 promoted the proliferation and invasiveness of HCC cells via SUMOylation using Soft agar and Transwell migration assays. Mechanistically, it was demonstrated that E2F4 upregulated the transcript and protein expression levels of baculoviral IAP repeat containing 5, cell division cycle associated 8 and DNA topoisomerase II α using western blotting. Furthermore, the interaction between E2F4 with lin‑9 DREAM multi‑vulva class B core complex component (LIN9) was explored by co‑immunoprecipitation, immunofluorescence co‑localization and bimolecular fluorescence complementation assays. Moreover, it was demonstrated that E2F4 promoted the progression of HCC cells via LIN9. Rescue experiments revealed that LIN9 facilitated the SUMOylation and proliferation of HCC cells, which was prevented by knocking down E2F4 expression. In conclusion, the findings of the present study indicated that E2F4 plays a major role in the proliferation of HCC cells and may be a potential therapeutic target in the future.

Introduction

Hepatocellular carcinoma (HCC) is the most prevalent type of liver cancer, accounting for 70-85% of all primary liver cancer cases worldwide. This severe disease, with high morbidity and mortality rates, is the third most common cause of cancer-related death worldwide (1). Despite the continuous development of therapeutic methods, the rate of recurrence and metastasis in HCC is also high (2,3). However, the precise mechanism underlying the onset and progression of HCC remains elusive. A number of studies have demonstrated the involvement of various cellular pathways, such as the nuclear factor κB pathway (4,5), ubiquitin-proteasome system (6) and autophagy pathway (7), in HCC pathogenesis. SUMOylation is an essential protein modification pathway and plays a pivotal role in the regulation of diverse biological processes, such as cellular localization, protein activity, protein stability or protein degradation (8). Upregulated expression of SUMOylated proteins in tumor tissues has been observed, and the activation of SUMOylation is closely linked to the development of tumors (9,10).

E2F transcription factor 4 (E2F4), a member of the E2F family, contributes to tumor progression (11-13). E2F4 influences the expression of toll-like receptor 8 and CD14, as well as the downstream activation of the signal transducer and activator of the transcription (STAT1) pathway (14). Studies have demonstrated that elevated levels of E2F4 are observed in the development of various types of cancer, including bladder cancer (15), Burkitt lymphoma (16), breast cancer (17), gastric cancer (18), cervical cancer (19), colorectal cancer (20) and acute myeloid leukemia (21). Consistently, there is a decrease in the G1-S phase transition and the proliferation of colon cancer cells with stable silencing of E2F4 (22). However, the precise regulatory role of E2F4 in HCC has yet to be elucidated, particularly in terms of regulating the SUMOylation pathway.

In the present study, the potential role of E2F4 in HCC was investigated through public database mining. To determine the changes in target gene expression, western blotting and reverse transcription-quantitative PCR (RT-qPCR) assays were used. Furthermore, co-immunoprecipitation (Co-IP), immunofluorescence co-localization and bimolecular fluorescence complementation (BiFC) assays were used to observe the interactions between E2F4 and a copartner. Subsequently, soft agar and Transwell migration assays were used to detect the effect of E2F4 on the proliferation and invasion in HCC cells.

Materials and methods

Bioinformatics

Public RNA-sequencing data were obtained from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/; accession no. GSE112221) (23), which included 10 samples. The data used in the present study were derived from 4 patients. 'N' represents cirrhotic tissue samples, while 'T' denotes HCC samples. Both the cirrhosis and HCC tissues originated from the same patients. A comprehensive analysis identified 105 common differentially expressed genes (DEGs) across these 4 patients. Enrichment analysis of these genes was conducted utilizing Metascape (https://www.metascape.org). Then, ChIP-X (https://www.maayanlab.net/X2K/; version 1.6) software prediction was used to obtain candidate TFs that regulate these target genes. Survival analysis and differential expression were analyzed using the GEPIA2 database (http://gepia2.cancer-pku.cn/#index), which included gene expression data from The Cancer Genome Atlas (https://portal.gdc.cancer.gov/; TCGA-LIHC) (24). Correlation analysis was carried out using the TIMER database (https://cistrome.shinyapps.io/timer/). The protein E2F4 co-partners were obtained using GENEMANIA (http://genemania.org), HINT (http://hint.yulab.org/) and STRING (https://string-db.org/) databases. E2F4 and DREAM multi-vulva class B core complex component (LIN9) protein docking studies were performed using the HDOCK Server (http://hdock.phys.hust.edu.cn/). A cut-off of P<0.05 was used to define a significant result.

Cell lines and culture

The human liver cancer cell lines, HepG2 (cat. no. CL-0103), HCC-LM3 (cat. no. CL-0278), Huh7 (cat. no. CL-0120), Huh6 (cat. no. CL-0119), and PLC (cat. no. CL-0415) were obtained from Procell Life Science & Technology Co., Ltd. MHCC 97H cells (cat. no. C6585) was purchased from Beyotime Institute of Biotechnology. The cell lines were cultured in 5% CO2 at 37°C, with DMEM containing 10% fetal bovine serum (Thermo Fisher Scientific, Inc.; cat. no. 12483020) and 100 U penicillin/streptomycin (Thermo Fisher Scientific, Inc.; cat. no. 15140122).

Cell transfection

E2F4 and LIN9 overexpression vectors were constructed using the CV186 lentiviral vector (Shanghai GeneChem Co., Ltd.). Short hairpin RNA (shRNA) against E2F4 or SUMO2/3 (Table SI) for knockdown were inserted into the GV298 lentiviral vector (Shanghai GeneChem Co., Ltd.; cat. no. GCD0316554). For transfection, the Huh7 (1×106 cells/well) and PLC (1×106 cells/well) cells were cultured in 6-well plates for 12 h at 37°C, followed by replacement with fresh medium. Subsequently, 2 μg E2F4-CV186 plasmid was transfected into Huh7 cells or the E2F4-GV298 vector was transfected into PLC cells using Lipofectamine 3000 (Thermo Fisher Scientific, Inc.; cat. no. L3000008) in a biological safety cabinet, based on the kit's instructions. Following transfection, the cell cultures were maintained at 37°C for an additional 48 h. Stable cell lines were established with 2 μg/ml puromycin selection for 2 weeks. Then, the poly-clonal stable cell lines were cultured in fresh medium with 1 μg/ml puromycin for maintenance.

RT-qPCR

Total RNA samples from Huh7 or PLC cells were extracted by the RNeasy Mini Kit (Qiagen China Co., Ltd.; cat. no. 74104). RT was conducted using the Transcriptor First Strand cDNA Synthesis Kit (Takara Biotechnology Co., Ltd.; cat. no. 6110A) according to the manufacturer's instructions. In the qPCR analysis, the SYBR Premix Ex Taq II (Takara Biotechnology Co., Ltd.; cat. no. RR820A) and primer sets (Table SII) were applied to determine the transcript levels via the 2−ΔΔCq method (25), using β-actin (ACTB) as the internal control. The thermocycling protocol was as follows: Holding stage at 95°C for 30 sec, cycling stage for 40 cycles at 95°C for 5 sec and 60°C for 30 sec, the melting curve stage at 95°C for 15 sec, 60°C for 1 min and 95°C for 15 sec.

Western blotting

Protein from the liver cancer cells was isolated by cell lysis buffer (Beyotime Institute of Biotechnology; cat. no. P0013). The quantification of protein concentrations was measured with the BCA Protein Assay Kit (Beyotime Institute of Biotechnology; cat. no. P0012). Proteins (30 μg) were subjected to separation on a 10% SDS-polyacrylamide gel, followed by transfer to PVDF membranes (MilliporeSigma; cat. no. IPVH00010). The membranes were then blocked with 5% skimmed milk (Beyotime Institute of Biotechnology; cat. no. P0216) in Tris-buffered saline containing 0.1% Tween-20 (TBST; Beyotime Institute of Biotechnology; cat. no. ST671) at room temperature for 1 h and washed three times with TBST. Then, the membranes were incubated at 4°C overnight with primary antibodies. After washing three times with TBST, the membranes were incubated with HRP-conjugated Goat anti-Rabbit IgG (ABclonal Biotech Co., Ltd.; cat. no. AS014, 1:5,000) or HRP-conjugated Goat anti-Mouse IgG (ABclonal Biotech Co., Ltd.; cat. no. AS003, 1:5,000) at room temperature for 1 h. Following incubation, the membranes were again washed three times with TBST. Then, the membranes were incubated with ECL reagent (MedChemExpress; cat. no. HY-K1005). Band visualization was performed using a ChemiDoc Imaging System (Bio-Rad Laboratories, Inc.) for 1 min. Densitometric analysis was conducted using ImageJ software (National Institutes of Health; version 1.53m). Western blotting was undertaken using antibodies specific for E2F4, LIN9, baculoviral IAP repeat containing 5 (BIRC5), cell division cycle associated 8 (CDCA8), DNA topoisomerase II α (TOP2A), small ubiquitin like modifier (SUMO)1, SUMO2/3, SUMO1/sentrin specific peptidase (SENP)1, SENP2, SENP3, SENP5, SENP6, SENP7, SENP8, LIN54, RBBP4, LIN52, LIN37 and ACTB (Table SIII).

Co-IP

The Co-IP assay was carried out as previously reported (26-29). Huh7 and PLC cells were lysed in 300 μl RIPA lysis buffer (Beyotime Institute of Biotechnology; cat. no. P0013D). Cell lysates were incubated with 10 μg of antibodies specific for E2F4 (Proteintech Group, Inc.; cat. no. 67812-1-Ig), LIN9 (Proteintech Group, Inc.; cat. no. 17882-1-AP), SUMO2/3 (Proteintech Group, Inc.; cat. no. 11251-1-AP), negative control rabbit IgG (Beyotime Institute of Biotechnology; cat. no. A7016) or negative control mouse IgG (Beyotime Institute of Biotechnology; cat. no. A7028) overnight at 4°C. Subsequently, the samples were incubated with 30 μl Pierce Protein A/G magnetic beads (MedChemExpress; cat. no. HY-K0202) at 4°C for another 3 h. Following centrifugation at 1,000 × g at 4°C for 10 min, the beads were washed with 1 ml lysis buffer three times and boiled for 10 min at 100°C. The proteins were subsequently isolated and identified using western blotting analysis.

Immunofluorescence co-localization assay

Huh7 and PLC cells (1×104 cells/well) were grown on coverslips in 24-well plates at 37°C for 24 h. Cells were fixed with 4% paraformaldehyde for 30 min at room temperature (20-25°C) and washed three times with PBS. The glass coverslips were incubated with 5% milk for 1 h at room temperature, then treated with antibodies specific for E2F4 (Proteintech Group, Inc.; cat. no. 67812-1-Ig; 1:100 dilution) and LIN9 (Proteintech Group, Inc.; cat. no. 17882-1-AP; 1:100 dilution) at 4°C overnight. Next, the coverslips were treated with Alexa Fluor 488 goat anti-mouse IgG (Abcam; cat. no. ab150113; 1:1,000 dilution) or Alexa Fluor 594 goat anti-rabbit IgG (Abcam; cat. no. ab150160; 1:1,000 dilution) at room temperature for 1 h in the dark. Cells were washed and stained with 4',6-diamidino-2-phenylindole (DAPI; 300 nmol/l) for 10 min at room temperature. The images were imaged under a confocal microscope. For EdU staining, Huh7 cells were cultured in a 12-well plate at a density of 1×105 cells/well. Following corresponding treatment, cell proliferation was detected with the EdU Cell Proliferation Kit (Beyotime Institute of Biotechnology; cat. no. C0081S) according to the manufacturer's instructions. Finally, cells were observed and captured under a fluorescence microscope.

BiFC assay

Human LIN9 cDNA (1,473 bp) or E2F4 cDNA (1,241 bp) were respectively subcloned into BiFC vectors pBiFC-VC155 (Addgene, Inc.; cat. no. 22011) and pBiFC-VN173 (Addgene, Inc.; cat. no. 22010). Co-transfection of the recombinant vectors (1 μg LIN9-VC155 and 1 μg E2F4-VN173) into Huh7 cells was performed using Lipofectamine 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C for 24 h. Next, the cells were fixed with 4% paraformaldehyde at room temperature for 30 min and incubated with DAPI for 5 min at room temperature. Fluorescence emission was observed under a confocal microscope, with excitation and emission wavelengths set at 488 and 500 nm, respectively.

Soft agar assay

Huh7 cells (5×103 per well) were mixed with a solution of 0.05% Noble agar. The mixture was then incubated on 6-well plates containing solidified 0.1% Noble agar for 3 weeks. After the incubation period, the cellular colonies that had formed were stained using 0.5% crystal violet dye and subsequently observed and manually counted using a light microscope.

Matrigel invasion assay

The invasiveness of Huh7 cells was assessed using a 12-well Transwell insert with 8.0-μm pores (Corning, Inc.; cat. no. 3422). The upper wells of the inserts were coated with 100 μl Matrigel (Corning, Inc.; cat. no. 354262; 1 mg/ml) at 37°C for 3 h. Serum-starved cells (1×105 per well) were seeded into the upper chamber of the Transwell insert with 200 μl serum-free DMEM, and 500 μl DMEM containing 10% FBS was added to the lower chamber. After cell culture for 24 h, the invaded cells were fixed with 4% paraformaldehyde at room temperature for 15 min, stained with 0.1% crystal violet for 30 min at room temperature, and subsequently quantified manually using a light microscope.

SUMOylation inhibitor

To assess the impact of SUMOylation on the proliferation and invasiveness of Huh7 cells, the SUMOylation inhibitor, TAK-981 (10 nM; MedChemExpress; cat. no. HY-111789), was added to the cell medium after E2F4-overexpression vector transfection. Following treatment, the cell cultures were maintained at 37°C for 24 h, The soft agar assay was then used to detect cell proliferation and the Matrigel invasion assay was used to detect invasiveness of Huh7 cells (according to the aforementioned protocols).

Statistical analysis

In the present study, the data are presented as the mean ± standard deviation. To compare differences between two datasets, a two-tailed unpaired Student's t-test was applied. The differences between multiple groups were calculated using one-way ANOVA followed by Dunnett's post hoc test. Spearman's correlation analysis was used to determine the expression correlation. Survival curves were constructed using the Kaplan-Meier method. High- and low-expression levels were separated by a suitable adjustable cut-off value. The log-rank test was utilized to assess differences in survival. All statistical analyses conducted in the present study were two-sided. P<0.05 was considered to indicate a statistically significant difference.

Results

E2F4 is a potential TF in regulating the progression of HCC

To investigate potential genes involved in the outcomes of patients with HCC, a comprehensive analysis was performed using HCC and cirrhosis tissue gene expression profiles available from the GEO. A total of 105 DEGs in HCC tissues compared with cirrhosis tissues were identified [fold change (FC) >2; Fig. 1A]. Furthermore, 76 genes were upregulated whereas 29 genes were downregulated in patients with HCC (Fig. 1A). To analyze the crucial pathways affected by potential regulators, a REACTOME pathway analysis for 105 DEGs was performed using Metascape (30). The results revealed that SUMOylation was a significantly enriched pathway including 5 target genes (aurora kinase A, BIRC5, chromobox homolog, CDCA8 and TOP2A; Fig. 1B). To investigate the crucial TFs that regulate the SUMOylation-associated genes, ChIP-X software was used to reveal that the top 5 TFs were E2F4, ETS proto-oncogene 1 (ETS1), interferon regulatory factor 1 (IRF1), MYCN proto-oncogene and lysine demethylase 6A (KDM6A). Notably, E2F4 ranked first among the identified TFs by the number of target genes, including BIRCA, CDCA8 and TOP2A, which were more enriched in HCC tissues (Fig. 1C and E). Meanwhile, comprehensive analysis using GEPIA2 (31) showed that upregulation of E2F4 (P=0.016), BIRC5 (P<0.001), CDCA8 (P<0.001) and TOP2A (P=0.01) were associated with a poorer overall survival (OS) rate in patients with HCC (Figs. 1D and S1). A positive correlation between E2F4 and BIRC5 (ρ=0.395; P<0.001), CDCA8 (ρ=0.58; P<0.001) or TOP2A (ρ=0.552; P<0.001; Fig. 1F) in HCC specimens was observed using TIMER (32). The results indicated that E2F4 may be a potential regulator in the progression of HCC.

E2F4 promotes the expression of SUMOylation-associated target genes

To understand the direct effects of E2F4 on BIRC5, CDCA8 and TOP2A transcription and expression in HCC cell lines, JASPAR (http://jaspar.genereg.net) was used to identify the E2F4 binding site in the promotors (Fig. 2A). Moreover, analysis of E2F4 chromatin immunoprecipitation-sequencing datasets (UCSC browser) also revealed that E2F4 bound at the promoters of BICR5, CDCA8 and TOP2A in HeLa cells (Fig. 2A). Western blotting revealed higher E2F4 expression levels in HepG2 and PLC cells and lower E2F4 expression levels in Huh7 cells (Fig. 2B). Since the present study focused on HCC, PLC and Huh7 were chosen as the HCC cell line models. To explore the regulation of SUMOylation-associated target genes by E2F4, western blotting was performed, which revealed that stable overexpression of E2F4 increased the protein levels of BIRC5, CDCA8 and TOP2A in Huh7 cells (Fig. 2C). However, knocking down E2F4 expression led to a decrease in the BIRC5, CDCA5, and TOP2A levels in PLC cells (Fig. 2C). These findings demonstrated that E2F4 regulated the expression of SUMOylation-associated genes in HCC cells.

E2F4 facilitates the proliferation of HCC cells via SUMOylation

The effects of E2F4 on SUMOylation in HCC cells was further determined by western blotting. In PLC and Huh7 cells, stable overexpression or knockdown of E2F4 enhanced and reduced the whole cell SUMOylation levels, respectively (Fig. 3A). The SUMOylation process involves numerous crucial proteins and specific proteases. Comprehensive analysis using the GEPIA2 database revealed higher expression levels of SUMO2/3 and SENP3 in HCC (T) tissues compared with the normal control (N) group (P<0.05, FC>1.5; Fig. S2A). Consistently, western blotting analysis following E2F4 overexpression in Huh7 cells revealed an increase in SUMO2/3 protein levels, but not the levels of SUMO1 or other SUMO-specific proteases (Fig. S2B). The results of a SUMOylation assay indicated that the SUMOylation levels of target genes were elevated in cells overexpressing E2F4 (Fig. S3A and B), which were decreased by stable knockdown of SUMO2/3 (Fig. S3B). In addition, the results of an EdU assay revealed that stable overexpression or knockdown of E2F4 increased and decreased the number of EdU+ cells compared with the controls, respectively (Fig. S4A). Subsequently, soft agar colony formation and Transwell Matrigel assays were performed to demonstrate that E2F4 significantly promoted the proliferation and invasion of HCC cells (Fig. S4B and C). To investigate the involvement of SUMOylation in E2F4-promoted proliferation and invasion, a SUMOylation inhibitor (TAK981) was applied to HCC cells stably overexpressing E2F4. Subsequent western blotting indicated that the SUMOylation levels in Huh7 cells were decreased by TAK-981 treatment (Fig. 3B). The results of EdU, soft agar colony formation and Transwell Matrigel assays showed that TAK-981 prevented the increase in proliferation and invasion levels of HCC cells stably overexpressing E2F4 (Fig. 3C-E). Similarly, the proliferation and invasion of HCC cells were inhibited by knocking down SUMO2/3 (Fig. S3C). Taken together, these results suggested that E2F4 elevated the proliferation and invasion of HCC cells by promoting SUMOylation.

LIN9 is an E2F4 potential protein partner

To determine potential protein partners that interact with E2F4, a E2F4 protein-protein interaction network was obtained using GENEMANIA (http://genemania.org), HINT (http://hint.yulab.org/) and STRING (https://string-db.org/) databases (Fig. 4A-C). Overlapping the results of the analyses derived from the aforementioned databases, LIN9, RB transcriptional corepressor like (RBL) 1, RBL2 and transcription factor Dp-1 (TFDP1) were identified as potential E2F4 binding partners (Fig. 4D). Subsequently, higher LIN9 levels were observed to be associated with a poorer OS rate in patients with HCC (P=0.0028), whereas the OS curves were not significantly different between the high and low RBL1 (P=0.17), RBL2 (P=0.80) and TFDP1 (P=0.22) (Fig. 4E) expression groups. Therefore, LIN9 was identified as a potential E2F4 co-partner in HCC cells. In addition, high expression of LIN9 was positively associated with MKI67 (Ki-67, a biomarker of proliferation) and MCAM (CD136, a potential biomarker of angiogenesis) in patients with HCC (Fig. 4F). According to the TCGA database, higher expression levels of E2F4, LIN9, BIRCA5, CDCA8 and TOP2A were observed in multiple cancer types (Fig. S5). Taken together, these results indicated that LIN9 likely interacts with E2F4 in HCC cells.

LIN9 physically interacts with E2F4 in HCC cells

To investigate the interaction between E2F4 with LIN9, protein docking studies were performed. The binding energy (-10.2 kcal/mol) revealed that E2F4 directly binds to LIN9 (Fig. 5A). Subsequently, a co-IP assay was used to demonstrated the endogenous interaction between E2F4 and LIN9 protein in PLC and Huh7 cells (Fig. 5B). Consistently, the results of an immunofluorescence staining assay demonstrated that E2F4 co-localized with LIN9 in the nucleus of PLC and Huh7 cells (Fig. 5C). To further determine the physical interaction between E2F4 and LIN9, a BiFC assay was performed to reveal that notable fluorescence occurred in Huh7 cells co-transfected with E2F4 and LIN9 plasmids (Fig. 5D). These findings suggested that E2F4 physically interacted with LIN9 protein in HCC cells.

LIN9 promotes the proliferation and invasion of HCC cells via E2F4

The cooperative roles of E2F4 and LIN9 in SUMOylation in HCC cells were further investigated. The results of a SUMOylation assay indicated that elevated SUMOylation of LIN9 was observed in cells stably overexpressing E2F4 (Fig. S3A and B). Unexpectedly, the interaction between E2F4 and other components (LIN54, LIN52, RBBP4 and LIN37) of the multi-vulva class B (MuvB) complex was prevented by overexpression of E2F4 (Fig. S6). The results suggested that E2F4 might inhibit DREAM complex stability following SUMOylation of LIN9. Western blotting and RT-qPCR revealed that stable overexpression of LIN9 increased the transcript and protein expression levels of BIRC5, CDCA8 and TOP2A, which was prevented by knocking down E2F4 (Fig. 6A and B). Consistently, there was an increase in the SUMOylation levels and proliferation of Huh7 cells stably overexpressing LIN9, which was rescued by knocking down E2F4 (Fig. 6C and D). The results of the soft agar colony formation and Matrigel invasion assays revealed that overexpression of LIN9 facilitated the anchor-independent proliferation and invasiveness of Huh7 cells, which was prevented by knocking down E2F4 (Fig. 6E and F). The aforementioned findings suggested that E2F4 coordinated with LIN9 to promote SUMOylation and the progression of HCC. Meanwhile, the identified SUMOylation-related proteins were significantly highly expressed in a variety of tumor types, further indicating that SUMOylation plays a critical role in the progression of cancer (Fig. S5).

Discussion

HCC ranks as the sixth most prevalent malignancy globally and emerges as the fourth primary cause of mortality in cancer-related deaths (1). The morbidity and mortality of HCC is contingent upon the disease stage at initial diagnosis. HCC fatalities are primarily caused by metastasis and post-operative recurrence (3), with an estimated recurrence rate of 60-70% (33). The dysregulation of gene expression, encompassing inactivation of tumor suppressor genes and activation of oncogenes, is widely recognized as a catalyst for tumor metastasis (24,34,35). Therefore, it is crucial to explore the mechanism of gene expression in patients with HCC. In the present study, the identified DEGs in HCC were significantly enriched in the SUMOylation pathway. Several studies have demonstrated that SUMOylation is closely associated with tumorigenesis, proliferation (36) and metastasis (37), and higher expression is observed in the majority of tumor cases (37,38). However, the effects of SUMOylation in HCC still remain largely unclear.

SUMOylation, a posttranslational modification (PTM) marked by the attachment of a small ubiquitin-like modifier peptide to a lysine residue, significantly influences cellular biological processes and the progression of cancer (39). Comparable to other types of PTMs, such as phosphorylation, ubiquitination and acetylation, SUMOylation is a highly dynamic and reversible mechanism that regulates the translation, subcellular localization, stability and protein-protein interactions of target proteins (40,41). Proteins such as STAT1, when modulated via both SUMOylation and de-SUMOylation, have been linked to the pathogenesis of cancer (42,43). A study has revealed that higher levels of SUMOylation can be observed in HCC tissues, and inhibitors of SUMOylation (such as TAK-981 and ML-792) suppress tumor progression (44). Notably, the results of the present study further supported these findings and revealed that TAK-981 prevented the proliferation and invasion of HCC cells. The higher transcript and protein levels of SUMOylation-related genes (BIRC5, CDCA8 and TOP2A) in HCC cells was also observed in the present study. Nevertheless, the specific function of STAT1 SUMOylation in HCC remains unclear, warranting future investigative efforts.

Some studies have highlighted the critical role of BIRC5 in oncogenesis, demonstrating that its overexpression not only suppresses apoptosis but also endows cells with tumorigenic capabilities (45,46). Specifically, the dysregulation of BIRC5-related genes has been tightly linked to the malignant advancement of HCC (47). Moreover, BIRC5 has been identified as a downstream target of microRNA (miR)-497-5p, which notably exhibits diminished transcription levels in HCC tissues compared with healthy tissues (48). Circular (circ)ANKRD52 facilitates the proliferation of HCC by sponging miR-497-5p and increasing BIRC5 expression (49). CDCA8 serves as an essential component of the chromosomal passenger complex, indispensable for accurate chromosomal distribution during mitosis (50). Furthermore, abnormal expression of CDCA8 leads to disruption of cellular homeostasis (51). Targeted suppression of CDCA8 inhibits the MEK/ERK pathway and impedes colony formation by regulating the CDK1/cyclin B1 signaling axis (52). TOP2A maintains mitotic chromosome structure and genome stability by resolving DNA topological strains and controlling genome dynamics (53). Abnormal levels of TOP2A are related to tumor progression (54), and bioinformatic analyses have identified TOP2A as a driving factor of HCC progression (55,56). Notably, TOP2A is upregulated and associated with an unfavorable prognosis in HCC (57), while miR-139-5p inhibits TOP2A expression, triggering cellular senescence and inhibiting the proliferation of HCC cells (58). Furthermore, the interaction between TOP2A and β-catenin facilitates the detachment of β-catenin from Yes1 associated transcriptional regulator, which enhances the excessive proliferation of HCC cells (59). Taken together, these findings underscore the notable influence of BIRC5, CDCA8 and TOP2A on the pathogenesis and progression of HCC, marking them as significant players and potential therapeutic targets. Therefore, it is critical to identify the key factors that regulate these target genes.

E2F4, a member of the E2F TF family, markedly influences tumor progression via the regulation of various gene expression and cell cycle signaling pathways, such apoptosis, differentiation and G0 to S phase transition (60-63). The E2F family can be generally divided into canonical activators (E2F1, E2F2 and E2F3a), canonical repressors (E2F3b, E2F4, E2F5 and E2F6) and atypical repressors (E2F7 and E2F8) (64). Notably, the mRNA expression levels of E2F have been associated with the progression stage and histopathological grades in HCC, and E2F4 has emerged as a key regulator of HCC (65,66). E2F4 interacts with the CDCA3 promoter region, leading to increased CDCA3 expression and facilitating both cell cycle progression and the proliferation of HCC cells (67). Additionally, E2F4 has been shown to directly bind to viral covalently closed circDNA, activating the hepatitis B virus (HBV) core promoter and upregulating transcription levels (68). Therefore, further exploration of the epigenetic regulatory roles of E2F4 in HCC is warranted in the future. Moreover, E2F4 is associated with immune infiltration in HCC (65). In the present study, higher expression levels of E2F4 protein compared with the controls were observed in patients with HCC. Kaplan-Meier analyses also suggested that E2F4 was associated with poorer survival in patients with HCC. Therefore, the E2F4 protein may serve as a promising prognostic indicator for HCC.

The findings of the present study also indicated that E2F4 directly interacted with LIN9 protein in the nucleus of HCC cells. In addition, elevated levels of LIN9 were associated with poorer outcomes in patients with HCC. LIN9 is a part of the MuvB complex, including LIN9, LIN37, LIN52, RB binding protein 4 and LIN54 (69,70). During the G0 and early G1 phases, the MuvB core complex engages with E2F4/5, DP1/2 and RB-like proteins, p130 or p107, to form the DREAM complex, thus inhibiting gene transcription. In the late phase of G1, the MuvB core is separated from the DREAM complex and then binds to B-MYB to form the MMB complex in the S phase, activating cell-cycle genes related to the S/G2/M phases (71-74). Deregulation of the DREAM complex has been implicated in multiple cancer types (such as myeloid leukemia, glioblastoma and prostate, breast, lung, esophagus, ovarian and pancreas cancer) and plays a critical role in cell cycle dynamics (75,76). It has been observed that aberrant expression of the DREAM complex occurs in HCC (77). Similarly, the findings of the present study suggested that higher expression of LIN9 was related to a worse OS. Notably, the results of the rescue studies revealed that overexpression of LIN9 reduced whole cell levels of SUMOylation and the proliferation and invasion of HCC cells following stable knockdown of E2F4 expression. An elevation in the SUMOylation of LIN9 was also noted in Huh7 cells overexpressing E2F4, which may potentially disrupt the stability of the DREAM complex. These findings suggested that the LIN9-E2F4 axis promotes progression in HCC via activation of the SUMOylation pathway. However, the intricacies of these mechanisms warrant further in-depth research.

In addition, other major TFs, such as E2F1 (78), ETS1 (79), IRF1 (80) and KDM6A (81), have been identified as prognostic biomarkers in HCC. E2F1, the classical E2F member, has been extensively studied. Notably, higher expression of E2F1 is significantly related to unfavorable prognosis in HCC (78). E2F1 can stimulate the proliferation of HCC cells by driving the expression of genes such as B-MYB (82), BRCA1 (83), DNA-binding protein A (84) and stathmin 1 (85), which enhance the development or progression of HCC. Additionally, mutations in the HBV core promoter lead to upregulated transcription of E2F1, resulting in HCC cell proliferation (86). Notably, nuclear E2F1 expression is positively correlated with the HCC tumor apoptotic index (87,88). E2F1 may disrupt the regulation of hepatitis B viral X protein on the p53 promoter and directly activate the p53 promoter via its specific binding sites, thereby blocking the HBV life cycle and HBV-associated HCC (89). As such, the intricate functions of E2F1 in HCC merit further investigation. ETS1, a member of the ETS family, is a crucial TF in regulating cell proliferation, invasion and metastasis. The interaction between murine double minute binding protein and ETS1 activates ETS1, consequently leading to the progression of HCC cells (90). Moreover, ETS1 also acts as a co-activator of pregnane X receptor to stimulate drug resistance in HCC (91,92). miR-338-3p inhibits progression of HCC through the downregulation of EST1 (93), while small molecule inhibitors targeting ETS1 have been shown to suppress proliferation or invasion of HCC cells (94). IRF1 is the main transcription regulator in the interferon-γ signaling pathway (95). IRF1 promotes autophagy in HCC cells, thus inhibiting their proliferation (96). Nevertheless, IRF1 increases the expression of programmed death-ligand 1, which may facilitate HCC cells in evading the antitumor immune response of the host (80). KDM6A plays different functions at different stages of HCC. On one hand, KDM6A promotes the progression of HCC through the upregulation of FGFR4 expression (97). On the other hand, KDM6A may hinder the proliferation of HCC cells through the negative regulation of the TGF-β/SMAD signaling pathway (98). Hence, the complexity of these TFs underscores the necessity to delve deeper into HCC development and new molecular therapeutic targets.

In summary, the present study demonstrated that E2F4 protein was highly expressed and related to poorer outcomes in patients with HCC. Stable expression of E2F4 promoted the whole cell SUMOylation of proteins by upregulating the transcription and protein expression levels of SUMOylation-associated genes including BIRC5, CDCA8 and TOP2A protein. Following treatment with the inhibitor of SUMOylation, TAK-981, the proliferation and invasion of HCC cells were downregulated. Notably, it was demonstrated that E2F4 directly bound to LIN9 protein, leading to the promotion of E2F4-mediated SUMOylation associated with HCC progression. Therefore, the results of the present study suggested that E2F4 and LIN9 play crucial roles in the progression of HCC and indicated that the E2F4-LIN9-SUMOylation axis may be a valuable potential therapeutic target in HCC.

Supplementary Data

Availability of data and materials

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

Authors' contributions

ZD and QL conceived and designed the present study. ZM, QL and WW performed most of the experiments, analyzed the data and wrote the manuscript. ZD and WW supervised the entire project. ZM, QL, WW and ZD confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

The present study was supported by The Health Commission of Hubei Province Scientific Research Project (grant no. WJ2021M107), Hubei Provincial Natural Science Foundation of China (grant no. 2023AFB160) and Scientific Research Project of Tongji Hospital (grant no. 2023B05).

References

1 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Facciorusso A: Drug-eluting beads transarterial chemoembolization for hepatocellular carcinoma: Current state of the art. World J Gastroenterol. 24:161–169. 2018. View Article : Google Scholar : PubMed/NCBI

3 

El-Serag HB, Marrero JA, Rudolph L and Reddy KR: Diagnosis and treatment of hepatocellular carcinoma. Gastroenterology. 134:1752–1763. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Shen RR, Zhou AY, Kim E, O'Connell JT, Hagerstrand D, Beroukhim R and Hahn WC: TRAF2 is an NF-κB-activating oncogene in epithelial cancers. Oncogene. 34:209–216. 2015. View Article : Google Scholar

5 

Sunami Y, Ringelhan M, Kokai E, Lu M, O'Connor T, Lorentzen A, Weber A, Rodewald AK, Müllhaupt B, Terracciano L, et al: Canonical NF-κB signaling in hepatocytes acts as a tumor-suppressor in hepatitis B virus surface antigen-driven hepatocellular carcinoma by controlling the unfolded protein response. Hepatology. 63:1592–1607. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Liang X, Yao J, Cui D, Zheng W, Liu Y, Lou G, Ye B, Shui L, Sun Y, Zhao Y and Zheng M: The TRAF2-p62 axis promotes proliferation and survival of liver cancer by activating mTORC1 pathway. Cell Death Differ. 30:1550–1562. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Tian Y, Kuo CF, Sir D, Wang L, Govindarajan S, Petrovic LM and Ou JH: Autophagy inhibits oxidative stress and tumor suppressors to exert its dual effect on hepatocarcinogenesis. Cell Death Differ. 22:1025–1034. 2015. View Article : Google Scholar :

8 

Chang HM and Yeh ETHH: SUMO: From bench to bedside. Physiol Rev. 100:1599–1619. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Seeler JS and Dejean A: SUMO and the robustness of cancer. Nat Rev Cancer. 17:184–197. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Eifler K and Vertegaal ACO: SUMOylation-mediated regulation of cell cycle progression and cancer. Trends Biochem Sci. 40:779–793. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Souza RF, Yin J, Smolinski KN, Zou TT, Wang S, Shi YQ, Rhyu MG, Cottrell J, Abraham JM, Biden K, et al: Frequent mutation of the E2F-4 cell cycle gene in primary human gastrointestinal tumors. Cancer Res. 57:2350–2353. 1997.PubMed/NCBI

12 

Wang D, Russell JL and Johnson DG: E2F4 and E2F1 have similar proliferative properties but different apoptotic and oncogenic properties in vivo. Mol Cell Biol. 20:3417–3424. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Schwemmle S and Pfeifer GP: Genomic structure and mutation screening of the E2F4 gene in human tumors. Int J Cancer. 86:672–677. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Zamani-Ahmadmahmudi M, Najafi A and Nassiri SM: Reconstruction of canine diffuse large B-cell lymphoma gene regulatory network: Detection of functional modules and hub genes. J Comp Pathol. 152:119–130. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Cheng C, Varn FS and Marsit CJ: E2F4 program is predictive of progression and intravesical immunotherapy efficacy in bladder cancer. Mol Cancer Res. 13:1316–1324. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Molina-Privado I, Jiménez-P R, Montes-Moreno S, Chiodo Y, Rodríguez-Martínez M, Sánchez-Verde L, Iglesias T, Piris MA and Campanero MR: E2F4 plays a key role in Burkitt lymphoma tumorigenesis. Leukemia. 26:2277–2285. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Rakha EA, Pinder SE, Paish EC, Robertson JF and Ellis IO: Expression of E2F-4 in invasive breast carcinomas is associated with poor prognosis. J Pathol. 203:754–761. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Xiao W, Wang J, Wang X, Cai S, Guo Y, Ye L, Li D, Hu A, Jin S, Yuan B, et al: Therapeutic targeting of the USP2-E2F4 axis inhibits autophagic machinery essential for zinc homeostasis in cancer progression. Autophagy. 18:2615–2635. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Gong J, Fan H, Deng J and Zhang Q: LncRNA HAND2-AS1 represses cervical cancer progression by interaction with transcription factor E2F4 at the promoter of C16orf74. J Cell Mol Med. 24:6015–6027. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Paquin MC, Leblanc C, Lemieux E, Bian B and Rivard N: Functional impact of colorectal cancer-associated mutations in the transcription factor E2F4. Int J Oncol. 43:2015–2022. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Feng Y, Li L, Du Y, Peng X and Chen F: E2F4 functions as a tumour suppressor in acute myeloid leukaemia via inhibition of the MAPK signalling pathway by binding to EZH2. J Cell Mol Med. 24:2157–2168. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Garneau H, Paquin MC, Carrier JC and Rivard N: E2F4 expression is required for cell cycle progression of normal intestinal crypt cells and colorectal cancer cells. J Cell Physiol. 221:350–358. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Hlady RA, Sathyanarayan A, Thompson JJ, Zhou D, Wu Q, Pham K, Lee JH, Liu C and Robertson KD: Integrating the epigenome to identify drivers of hepatocellular carcinoma. Hepatology. 69:639–652. 2019. View Article : Google Scholar

24 

Cancer Genome Atlas Research Network: Electronic address: simplewheeler@bcm.edu; Cancer Genome Atlas Research Network: Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell. 169:1327–1341.e23. 2017. View Article : Google Scholar

25 

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

26 

Jiang G, Zheng L, Pu J, Mei H, Zhao J, Huang K, Zeng F and Tong Q: Small RNAs targeting transcription start site induce heparanase silencing through interference with transcription initiation in human cancer cells. PLoS One. 7:e313792012. View Article : Google Scholar : PubMed/NCBI

27 

Fang E, Wang X, Wang J, Hu A, Song H, Yang F, Li D, Xiao W, Chen Y, Guo Y, et al: Therapeutic targeting of YY1/MZF1 axis by MZF1-uPEP inhibits aerobic glycolysis and neuroblastoma progression. Theranostics. 10:1555–1571. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Li H, Yang F, Hu A, Wang X, Fang E, Chen Y, Li D, Song H, Wang J, Guo Y, et al: Therapeutic targeting of circ-CUX1/EWSR1/MAZ axis inhibits glycolysis and neuroblastoma progression. EMBO Mol Med. 11:e108352019. View Article : Google Scholar : PubMed/NCBI

29 

Fang E, Wang X, Yang F, Hu A, Wang J, Li D, Song H, Hong M, Guo Y, Liu Y, et al: Therapeutic targeting of MZF1-AS1/PARP1/E2F1 axis inhibits proline synthesis and neuroblastoma progression. Adv Sci (Weinh). 6:19005812019. View Article : Google Scholar : PubMed/NCBI

30 

Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C and Chanda SK: Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 10:15232019. View Article : Google Scholar : PubMed/NCBI

31 

Tang Z, Kang B, Li C, Chen T and Zhang Z: GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 47(W1): W556–W560. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, Li B and Liu XS: TIMER: A web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 77:e108–e110. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Chen Q, Li F, Zhong C, Zou Y, Li Z, Gao Y, Zou Q, Xia Y, Wang K and Shen F: Inflammation score system using preoperative inflammatory markers to predict prognosis for hepatocellular carcinoma after hepatectomy: A cohort study. J Cancer. 11:4947–4956. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Jiang Y, Sun A, Zhao Y, Ying W, Sun H, Yang X, Xing B, Sun W, Ren L, Hu B, et al: Proteomics identifies new therapeutic targets of early-stage hepatocellular carcinoma. Nature. 567:257–261. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Clark DJ, Dhanasekaran SM, Petralia F, Pan J, Song X, Hu Y, da Veiga Leprevost F, Reva B, Lih TSM, Chang HY, et al: Integrated proteogenomic characterization of clear cell renal cell carcinoma. Cell. 180:2072020. View Article : Google Scholar : PubMed/NCBI

36 

Du Y, Hou G, Zhang H, Dou J, He J, Guo Y, Li L, Chen R, Wang Y, Deng R, et al: SUMOylation of the m6A-RNA methyltransferase METTL3 modulates its function. Nucleic Acids Res. 46:5195–5208. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Bogachek MV, Park JM, De Andrade JP, Lorenzen AW, Kulak MV, White JR, Gu VW, Wu VT and Weigel RJ: Inhibiting the SUMO pathway represses the cancer stem cell population in breast and colorectal carcinomas. Stem Cell Reports. 7:1140–1151. 2016. View Article : Google Scholar : PubMed/NCBI

38 

He X, Riceberg J, Soucy T, Koenig E, Minissale J, Gallery M, Bernard H, Yang X, Liao H, Rabino C, et al: Probing the roles of SUMOylation in cancer cell biology by using a selective SAE inhibitor. Nat Chem Biol. 13:1164–1171. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Hay RT: SUMO: A history of modification. Mol Cell. 18:1–12. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Gareau JR and Lima CD: The SUMO pathway: Emerging mechanisms that shape specificity, conjugation and recognition. Nat Rev Mol Cell Biol. 11:861–871. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Geiss-Friedlander R and Melchior F: Concepts in sumoylation: A decade on. Nat Rev Mol Cell Biol. 8:947–956. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Yu B, Swatkoski S, Holly A, Lee LC, Giroux V, Lee CS, Hsu D, Smith JL, Yuen G, Yue J, et al: Oncogenesis driven by the Ras/Raf pathway requires the SUMO E2 ligase Ubc9. Proc Natl Acad Sci USA. 112:E1724–E1733. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Zhang J, Tan GL, Jiang M, Wang TS, Liu GH, Xiong SS and Qing X: Effects of SENP1-induced deSUMOylation of STAT1 on proliferation and invasion in nasopharyngeal carcinoma. Cell Signal. 101:1105302023. View Article : Google Scholar

44 

Wang Z, Pan B, Su L, Yu H, Wu X, Yao Y, Zhang X, Qiu J and Tang N: SUMOylation inhibitors activate anti-tumor immunity by reshaping the immune microenvironment in a preclinical model of hepatocellular carcinoma. Cell Oncol (Dordr). 47:513–532. 2024. View Article : Google Scholar

45 

Wang B, Li X, Zhao G, Yan H, Dong P, Watari H, Sims M, Li W, Pfeffer LM, Guo Y and Yue J: miR-203 inhibits ovarian tumor metastasis by targeting BIRC5 and attenuating the TGFβ pathway. J Exp Clin Cancer Res. 37:2352018. View Article : Google Scholar

46 

Kelly RJ, Lopez-Chavez A, Citrin D, Janik JE and Morris JC: Impacting tumor cell-fate by targeting the inhibitor of apoptosis protein survivin. Mol Cancer. 10:352011. View Article : Google Scholar : PubMed/NCBI

47 

Xu R, Lin L, Zhang B, Wang J, Zhao F, Liu X and Li Y and Li Y: Identification of prognostic markers for hepatocellular carcinoma based on the epithelial-mesenchymal transition-related gene BIRC5. BMC Cancer. 21:6872021. View Article : Google Scholar : PubMed/NCBI

48 

Tian LL, Qian B, Jiang XH, Liu YS, Chen T, Jia CY, Zhou YL, Liu JB, Ma YS, Fu D and Ding ST: MicroRNA-497-5p is downregulated in hepatocellular carcinoma and associated with tumorigenesis and poor prognosis in patients. Int J Genomics. 2021:66703902021. View Article : Google Scholar : PubMed/NCBI

49 

Zhang M, Yan X, Wen P, Bai W and Zhang Q: CircANKRD52 promotes the tumorigenesis of hepatocellular carcinoma by sponging miR-497-5p and upregulating BIRC5 expression. Cell Transplant. 30:96368972110088742021. View Article : Google Scholar : PubMed/NCBI

50 

Zhang C, Zhao L, Leng L, Zhou Q, Zhang S, Gong F, Xie P and Lin G: CDCA8 regulates meiotic spindle assembly and chromosome segregation during human oocyte meiosis. Gene. 741:1444952020. View Article : Google Scholar : PubMed/NCBI

51 

Yamanaka Y, Heike T, Kumada T, Shibata M, Takaoka Y, Kitano A, Shiraishi K, Kato T, Nagato M, Okawa K, et al: Loss of Borealin/DasraB leads to defective cell proliferation, p53 accumulation and early embryonic lethality. Mech Dev. 125:441–450. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Cui Y and Jiang N: CDCA8 facilitates tumor proliferation and predicts a poor prognosis in hepatocellular carcinoma. Appl Biochem Biotechnol. 196:1481–1492. 2024. View Article : Google Scholar

53 

Nielsen CF, Zhang T, Barisic M, Kalitsis P and Hudson DF: Topoisomerase IIα is essential for maintenance of mitotic chromosome structure. Proc Natl Acad Sci USA. 117:12131–12142. 2020. View Article : Google Scholar

54 

Zhong W, Yang Y, Zhang A, Lin W, Liang G, Ling Y, Zhong J, Yong J, Liu Z, Tian Z, et al: Prognostic and predictive value of the combination of TOP2A and HER2 in node-negative tumors 2 cm or smaller (T1N0) breast cancer. Breast Cancer. 27:1147–1157. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Shen S, Kong J, Qiu Y, Yang X, Wang W and Yan L: Identification of core genes and outcomes in hepatocellular carcinoma by bioinformatics analysis. J Cell Biochem. 120:10069–10081. 2019. View Article : Google Scholar

56 

Gao X, Wang X and Zhang S: Bioinformatics identification of crucial genes and pathways associated with hepatocellular carcinoma. Biosci Rep. 38:BSR201814412018. View Article : Google Scholar : PubMed/NCBI

57 

Meng J, Wei Y, Deng Q, Li L and Li X: Study on the expression of TOP2A in hepatocellular carcinoma and its relationship with patient prognosis. Cancer Cell Int. 22:292022. View Article : Google Scholar : PubMed/NCBI

58 

Wang K, Jiang X, Jiang Y, Liu J, Du Y, Zhang Z, Li Y, Zhao X, Li J and Zhang R: EZH2-H3K27me3-mediated silencing of mir-139-5p inhibits cellular senescence in hepatocellular carcinoma by activating TOP2A. J Exp Clin Cancer Res. 42:3202023. View Article : Google Scholar : PubMed/NCBI

59 

Zhao HC, Chen CZ, Tian YZ, Song HQ, Wang XX, Li YJ, He JF and Zhao HL: CD168+ macrophages promote hepatocellular carcinoma tumor stemness and progression through TOP2A/β-catenin/YAP1 axis. iScience. 26:1068622023. View Article : Google Scholar

60 

Yang J, Song K, Krebs TL, Jackson MW and Danielpour D: Rb/E2F4 and Smad2/3 link survivin to TGF-beta-induced apoptosis and tumor progression. Oncogene. 27:5326–5338. 2008. View Article : Google Scholar : PubMed/NCBI

61 

Zwicker J, Lucibello FC, Wolfraim LA, Gross C, Truss M, Engeland K and Müller R: Cell cycle regulation of the cyclin A, cdc25C and cdc2 genes is based on a common mechanism of transcriptional repression. EMBO J. 14:4514–4522. 1995. View Article : Google Scholar : PubMed/NCBI

62 

Ikeda MA, Jakoi L and Nevins JR: A unique role for the Rb protein in controlling E2F accumulation during cell growth and differentiation. Proc Natl Acad Sci USA. 93:3215–3220. 1996. View Article : Google Scholar : PubMed/NCBI

63 

van der Sman J, Thomas NS and Lam EW: Modulation of E2F complexes during G0 to S phase transition in human primary B-lymphocytes. J Biol Chem. 274:12009–12016. 1999. View Article : Google Scholar : PubMed/NCBI

64 

Aksoy O, Chicas A, Zeng T, Zhao Z, McCurrach M, Wang X and Lowe SW: The atypical E2F family member E2F7 couples the p53 and RB pathways during cellular senescence. Genes Dev. 26:1546–1557. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Zheng Q, Fu Q, Xu J, Gu X, Zhou H and Zhi C: Transcription factor E2F4 is an indicator of poor prognosis and is related to immune infiltration in hepatocellular carcinoma. J Cancer. 12:1792–1803. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Huang YL, Ning G, Chen LB, Lian YF, Gu YR, Wang JL, Chen DM, Wei H and Huang YH: Promising diagnostic and prognostic value of E2Fs in human hepatocellular carcinoma. Cancer Manag Res. 11:1725–1740. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Liu J, Xia L, Wang S, Cai X, Wu X, Zou C, Shan B, Luo M and Wang D: E2F4 promotes the proliferation of hepatocellular carcinoma cells through upregulation of CDCA3. J Cancer. 12:5173–5180. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Wei J, Shi Y, Zou C, Zhang H, Peng H, Wang S, Xia L, Yang Y, Zhang X, Liu J, et al: Cellular Id1 inhibits hepatitis B virus transcription by interacting with the novel covalently closed circular DNA-binding protein E2F4. Int J Biol Sci. 18:65–81. 2022. View Article : Google Scholar : PubMed/NCBI

69 

Korenjak M and Brehm A: E2F-Rb complexes regulating transcription of genes important for differentiation and development. Curr Opin Genet Dev. 15:520–527. 2005. View Article : Google Scholar : PubMed/NCBI

70 

Lewis PW, Beall EL, Fleischer TC, Georlette D, Link AJ and Botchan MR: Identification of a Drosophila Myb-E2F2/RBF transcriptional repressor complex. Genes Dev. 18:2929–2940. 2004. View Article : Google Scholar : PubMed/NCBI

71 

Korenjak M, Taylor-Harding B, Binné UK, Satterlee JS, Stevaux O, Aasland R, White-Cooper H, Dyson N and Brehm A: Native E2F/RBF complexes contain Myb-interacting proteins and repress transcription of developmentally controlled E2F target genes. Cell. 119:181–193. 2004. View Article : Google Scholar : PubMed/NCBI

72 

Litovchick L, Sadasivam S, Florens L, Zhu X, Swanson SK, Velmurugan S, Chen R, Washburn MP, Liu XS and DeCaprio JA: Evolutionarily conserved multisubunit RBL2/p130 and E2F4 protein complex represses human cell cycle-dependent genes in quiescence. Mol Cell. 26:539–551. 2007. View Article : Google Scholar : PubMed/NCBI

73 

Sadasivam S, Duan S and DeCaprio JA: The MuvB complex sequentially recruits B-Myb and FoxM1 to promote mitotic gene expression. Genes Dev. 26:474–489. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Fischer M, Grossmann P, Padi M and DeCaprio JA: Integration of TP53, DREAM, MMB-FOXM1 and RB-E2F target gene analyses identifies cell cycle gene regulatory networks. Nucleic Acids Res. 44:6070–6086. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Sadasivam S and DeCaprio JA: The DREAM complex: Master coordinator of cell cycle-dependent gene expression. Nat Rev Cancer. 13:585–595. 2013. View Article : Google Scholar : PubMed/NCBI

76 

MacDonald J, Ramos-Valdes Y, Perampalam P, Litovchick L, DiMattia GE and Dick FA: A systematic analysis of negative growth control implicates the DREAM complex in cancer cell dormancy. Mol Cancer Res. 15:371–381. 2017. View Article : Google Scholar

77 

Wang L and Liu X: Comprehensive analysis of the expression and prognosis for the DREAM complex in human cancers. Front Genet. 13:8147252022. View Article : Google Scholar : PubMed/NCBI

78 

Tan Z, Chen M, Peng F, Yang P, Peng Z, Zhang Z, Li X, Zhu X, Zhang L, Zhao Y and Liu Y: E2F1 as a potential prognostic and therapeutic biomarker by affecting tumor development and immune microenvironment in hepatocellular carcinoma. Transl Cancer Res. 11:2713–2732. 2022. View Article : Google Scholar : PubMed/NCBI

79 

Ito Y, Miyoshi E, Takeda T, Sakon M, Noda K, Tsujimoto M, Monden M, Taniguchi N and Matsuura N: Expression and possible role of ets-1 in hepatocellular carcinoma. Am J Clin Pathol. 114:719–725. 2000. View Article : Google Scholar : PubMed/NCBI

80 

Yan Y, Zheng L, Du Q, Yan B and Geller DA: Interferon regulatory factor 1 (IRF-1) and IRF-2 regulate PD-L1 expression in hepatocellular carcinoma (HCC) cells. Cancer Immunol Immunother. 69:1891–1903. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Qu LH, Fang Q, Yin T, Yi HM, Mei GB, Hong ZZ, Qiu XB, Zhou R and Dong HF: Comprehensive analyses of prognostic biomarkers and immune infiltrates among histone lysine demethylases (KDMs) in hepatocellular carcinoma. Cancer Immunol Immunother. 71:2449–2467. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Nakajima T, Yasui K, Zen K, Inagaki Y, Fujii H, Minami M, Tanaka S, Taniwaki M, Itoh Y, Arii S, et al: Activation of B-Myb by E2F1 in hepatocellular carcinoma. Hepatol Res. 38:886–895. 2008. View Article : Google Scholar

83 

Chen Q, Wang L, Jiang M, Huang J, Jiang Z, Feng H and Ji Z: E2F1 interactive with BRCA1 pathway induces HCC two different small molecule metabolism or cell cycle regulation via mitochondrion or CD4+T to cytosol. J Cell Physiol. 233:1213–1221. 2018. View Article : Google Scholar

84 

Arakawa Y, Kajino K, Kano S, Tobita H, Hayashi J, Yasen M, Moriyama M, Arakawa Y and Hino O: Transcription of dbpA, a Y box binding protein, is positively regulated by E2F1: Implications in hepatocarcinogenesis. Biochem Biophys Res Commun. 322:297–302. 2004. View Article : Google Scholar : PubMed/NCBI

85 

Chen YL, Uen YH, Li CF, Horng KC, Chen LR, Wu WR, Tseng HY, Huang HY, Wu LC and Shiue YL: The E2F transcription factor 1 transactives stathmin 1 in hepatocellular carcinoma. Ann Surg Oncol. 20:4041–4054. 2013. View Article : Google Scholar

86 

Huang Y, Tai AW, Tong S and Lok AS: HBV core promoter mutations promote cellular proliferation through E2F1-mediated upregulation of S-phase kinase-associated protein 2 transcription. J Hepatol. 58:1068–1073. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Farra R, Grassi G, Tonon F, Abrami M, Grassi M, Pozzato G, Fiotti N, Forte G and Dapas B: The role of the transcription factor E2F1 in hepatocellular carcinoma. Curr Drug Deliv. 14:272–281. 2017.

88 

Sun HX, Xu Y, Yang XR, Wang WM, Bai H, Shi RY, Nayar SK, Devbhandari RP, He YZ, Zhu QF, et al: Hypoxia inducible factor 2 alpha inhibits hepatocellular carcinoma growth through the transcription factor dimerization partner 3/E2F transcription factor 1-dependent apoptotic pathway. Hepatology. 57:1088–1097. 2013. View Article : Google Scholar

89 

Choi M, Lee H and Rho HM: E2F1 activates the human p53 promoter and overcomes the repressive effect of hepatitis B viral X protein (Hbx) on the p53 promoter. IUBMB Life. 53:309–317. 2002. View Article : Google Scholar

90 

Wang H, Chu F, Zhijie L, Bi Q, Lixin L, Zhuang Y, Xiaofeng Z, Niu X, Zhang D, Xi H and Li BA: MTBP enhances the activation of transcription factor ETS-1 and promotes the proliferation of hepatocellular carcinoma cells. Front Oncol. 12:9850822022. View Article : Google Scholar : PubMed/NCBI

91 

Bhagyaraj E, Ahuja N, Kumar S, Tiwari D, Gupta S, Nanduri R and Gupta P: TGF-β induced chemoresistance in liver cancer is modulated by xenobiotic nuclear receptor PXR. Cell Cycle. 18:3589–3602. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Shao Z, Li Y, Dai W, Jia H, Zhang Y, Jiang Q, Chai Y, Li X, Sun H, Yang R, et al: ETS-1 induces Sorafenib-resistance in hepatocellular carcinoma cells via regulating transcription factor activity of PXR. Pharmacol Res. 135:188–200. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Li YH, Lv MF, Lu MS and Bi JP: Bone marrow mesenchymal stem cell-derived exosomal MiR-338-3p represses progression of hepatocellular carcinoma by targeting ETS1. J Biol Regul Homeost Agents. 35:617–627. 2021.PubMed/NCBI

94 

Jie Y, Liu G, E M, Li Y, Xu G, Guo J, Li Y, Rong G, Li Y and Gu A: Novel small molecule inhibitors of the transcription factor ETS-1 and their antitumor activity against hepatocellular carcinoma. Eur J Pharmacol. 906:1742142021. View Article : Google Scholar : PubMed/NCBI

95 

Tamura T, Yanai H, Savitsky D and Taniguchi T: The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol. 26:535–584. 2008. View Article : Google Scholar : PubMed/NCBI

96 

Li P, Du Q, Cao Z, Guo Z, Evankovich J, Yan W, Chang Y, Shao L, Stolz DB, Tsung A and Geller DA: Interferon-γ induces autophagy with growth inhibition and cell death in human hepatocellular carcinoma (HCC) cells through interferon-regulatory factor-1 (IRF-1). Cancer Lett. 314:213–222. 2012. View Article : Google Scholar

97 

Guo W, Li S, Qian Y, Li L, Wang F, Tong Y, Li Q, Zhu Z, Gao WQ and Liu Y: KDM6A promotes hepatocellular carcinoma progression and dictates lenvatinib efficacy by upregulating FGFR4 expression. Clin Transl Med. 13:e14522023. View Article : Google Scholar : PubMed/NCBI

98 

Li Y, Yang J, Zhang X, Liu H and Guo J: KDM6A suppresses hepatocellular carcinoma cell proliferation by negatively regulating the TGF-β/SMAD signaling pathway. Exp Ther Med. 20:2774–2782. 2020.PubMed/NCBI

Related Articles

Journal Cover

October-2024
Volume 65 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma Z, Li Q, Wang W and Deng Z: Transcription factor E2F4 facilitates SUMOylation to promote HCC progression through interaction with LIN9. Int J Oncol 65: 98, 2024.
APA
Ma, Z., Li, Q., Wang, W., & Deng, Z. (2024). Transcription factor E2F4 facilitates SUMOylation to promote HCC progression through interaction with LIN9. International Journal of Oncology, 65, 98. https://doi.org/10.3892/ijo.2024.5686
MLA
Ma, Z., Li, Q., Wang, W., Deng, Z."Transcription factor E2F4 facilitates SUMOylation to promote HCC progression through interaction with LIN9". International Journal of Oncology 65.4 (2024): 98.
Chicago
Ma, Z., Li, Q., Wang, W., Deng, Z."Transcription factor E2F4 facilitates SUMOylation to promote HCC progression through interaction with LIN9". International Journal of Oncology 65, no. 4 (2024): 98. https://doi.org/10.3892/ijo.2024.5686