Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
J Physiol Pharmacol ; 75(2): 173-183, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38736264

ABSTRACT

Quercetin is widely distributed in plants as a flavonol compound with multiple biological activities. It has been found that quercetin can regulate bone homeostasis through multiple pathways and targets. This study investigated the role and specific molecular mechanisms of quercetin in regulating osteoblast viability, proliferation, migration and osteogenic differentiation. A mouse model of traumatic fracture was established and then 100 mg/kg quercetin corn oil suspension was gavaged at the same time every day for 28 days. miR-6089 and E2F transcription factor 2 (E2F2) expression levels in mice were measured. Fracture healing in mice was observed. MC3T3-E1 cells were transfected with plasmids targeting miR-6089 and E2F2, and cell viability, proliferation, migration, apoptosis, and osteogenic differentiation were determined. The targeting relationship between miR-6089 and E2F2 was verified. In vivo experiments showed that quercetin significantly increased osteocalcin (OCN) expression (P<0.05) and promoted fracture healing in traumatic fracture (TF) mice. miR-6089 expression was down-regulated (P<0.05) and E2F2 expression was up-regulated (P<0.05) in TF mice. Quercetin promoted miR-6089 expression and inhibited E2F2 expression (both P<0.05). In vitro results showed that quercetin promoted miR-6089 expression and inhibited E2F2 expression in a dose-dependent manner (both P<0.05). Quercetin dose-dependently promoted MC3T3-E1 cell viability, proliferation, migration, and osteogenic differentiation, and inhibited MC3T3-E1 cell apoptosis (all P<0.05). Up-regulating miR-6089 further promoted MC3T3-E1 cell viability, proliferation, migration and osteogenic differentiation, and inhibited MC3T3-E1 cell apoptosis (all P<0.05). miR-6089 targeted and regulated E2F2 expression. Up-regulating E2F2 attenuated the promoting effect of up-regulated miR-6089 on MC3T3-E1 cell viability, proliferation, migration, osteogenic differentiation, and inhibition of apoptosis (all P<0.05). We conclude that quercetin enhances osteoblast viability, proliferation, migration, and osteogenic differentiation by modulating the miR-6089/E2F2 axis, thereby promoting fracture healing.


Subject(s)
E2F2 Transcription Factor , Fracture Healing , MicroRNAs , Osteoblasts , Osteogenesis , Quercetin , Animals , Male , Mice , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , E2F2 Transcription Factor/metabolism , E2F2 Transcription Factor/genetics , Fracture Healing/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Quercetin/pharmacology
2.
Front Immunol ; 15: 1397303, 2024.
Article in English | MEDLINE | ID: mdl-38807594

ABSTRACT

The E2F family of transcription factors plays a crucial role in the regulation of cell cycle progression and cell proliferation. Accumulative evidence indicates that aberrant expression or activation of E2F2 is a common phenomenon in malignances. E2F2 has emerged as a key player in the development and progression of various types of tumors. A wealth of research has substantiated that E2F2 could contribute to the enhancement of tumor cell proliferation, angiogenesis, and invasiveness. Moreover, E2F2 exerts its influence on a myriad of cellular processes by engaging with a spectrum of auxiliary factors and downstream targets, including apoptosis and DNA repair. The dysregulation of E2F2 in the context of carcinogenesis may be attributable to a multitude of mechanisms, which encompass modifications in upstream regulatory elements or epigenetic alterations. This review explores the function of E2F2 in cancer progression and both established and emerging therapeutic strategies aiming at targeting this oncogenic pathway, while also providing a strong basis for further research on the biological function and clinical applications of E2F2.


Subject(s)
Disease Progression , E2F2 Transcription Factor , Neoplasms , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , E2F2 Transcription Factor/metabolism , E2F2 Transcription Factor/genetics , Animals , Gene Expression Regulation, Neoplastic , Molecular Targeted Therapy , Cell Proliferation
3.
J Mol Endocrinol ; 73(1)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38564418

ABSTRACT

The estrogen receptor-α (ER) drives 75% of breast cancers. On activation, the ER recruits and assembles a 1-2 MDa transcriptionally active complex. These complexes can modulate tumour growth, and understanding the roles of individual proteins within these complexes can help identify new therapeutic targets. Here, we present the discovery of ER and ZMIZ1 within the same multi-protein assembly by quantitative proteomics, and validated by proximity ligation assay. We characterise ZMIZ1 function by demonstrating a significant decrease in the proliferation of ER-positive cancer cell lines. To establish a role for the ER-ZMIZ1 interaction, we measured the transcriptional changes in the estrogen response post-ZMIZ1 knockdown using an RNA-seq time-course over 24 h. Gene set enrichment analysis of the ZMIZ1-knockdown data identified a specific delay in the response of estradiol-induced cell cycle genes. Integration of ENCODE data with our RNA-seq results identified that ER and ZMIZ1 both bind the promoter of E2F2. We therefore propose that ER and ZMIZ1 interact to enable the efficient estrogenic response at subset of cell cycle genes via a novel ZMIZ1-ER-E2F2 signalling axis. Finally, we show that high ZMIZ1 expression is predictive of worse patient outcome, ER and ZMIZ1 are co-expressed in breast cancer patients in TCGA and METABRIC, and the proteins are co-localised within the nuclei of tumour cell in patient biopsies. In conclusion, we establish that ZMIZ1 is a regulator of the estrogenic cell cycle response and provide evidence of the biological importance of the ER-ZMIZ1 interaction in ER-positive patient tumours, supporting potential clinical relevance.


Subject(s)
Breast Neoplasms , E2F2 Transcription Factor , Estrogen Receptor alpha , Gene Expression Regulation, Neoplastic , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Female , Cell Line, Tumor , E2F2 Transcription Factor/metabolism , E2F2 Transcription Factor/genetics , Cell Proliferation/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Protein Binding , Promoter Regions, Genetic/genetics , Signal Transduction , Cell Cycle/genetics , Prognosis
4.
BMC Cancer ; 24(1): 214, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360622

ABSTRACT

BACKGROUND: Laryngeal squamous cell carcinoma (LSCC) is one of the most common types of cancer in the upper respiratory tract. It is well-known that it has a high mortality rate and poor prognosis in advanced stages. There are well-known risk factors for LSCC, though new specific and prognostic blood-based markers for LSCC development and prognosis are essential. The current study aimed to evaluate the impact of four different single nucleotide polymorphisms (SNPs), E2F1 (rs3213183 and rs3213180) and E2F2 (rs2075993 and rs3820028), on LSCC development, morphological features, and patient 5-year survival rate. METHODS: A total of 200 LSCC patients and 200 controls were included in this study; both groups were matched by age and sex. In the present study, we analyzed four single nucleotide polymorphisms (SNPs) in the genes E2F1 (rs3213183 and rs3213180) and E2F2 (rs2075993 and rs3820028) and evaluated their associations with the risk of LSCC development, its clinical and morphological manifestation, and patients 5-year survival rate. Genotyping was carried out using RT-PCR. RESULTS: None of the analyzed SNPs showed a direct association with LSCC development. E2F2 rs2075993 G allele carriers (OR = 4.589, 95% CI 1.050-20.051, p = 0.043) and rs3820028 A allele carriers (OR = 4.750, 95% CI 1.088-20.736, p = 0.038) had a statistically significantly higher risk for poor differentiated or undifferentiated LSCC than non-carriers. E2F1 rs3213180 GC heterozygotes were found to have a 3.7-fold increased risk for lymph node involvement (OR = 3.710, 95% CI 1.452-9.479, p = 0.006). There was no statistically significant association between investigated SNPs and patient 5-year survival rate. CONCLUSIONS: The present study indicates that E2F2 rs2075993 and rs3820028 impact LSCC differentiation, whereas E2F1 rs3213180 - on lymph node involvement.


Subject(s)
E2F1 Transcription Factor , E2F2 Transcription Factor , Laryngeal Neoplasms , Squamous Cell Carcinoma of Head and Neck , Humans , E2F1 Transcription Factor/genetics , E2F2 Transcription Factor/genetics , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Polymorphism, Single Nucleotide , Prognosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology
5.
Br J Haematol ; 202(4): 840-855, 2023 08.
Article in English | MEDLINE | ID: mdl-37365680

ABSTRACT

Multiple myeloma (MM) is the second most common haematological malignancy. Despite the development of new drugs and treatments in recent years, the therapeutic outcomes of patients are not satisfactory. It is necessary to further investigate the molecular mechanism underlying MM progression. Herein, we found that high E2F2 expression was correlated with poor overall survival and advanced clinical stages in MM patients. Gain- and loss-of-function studies showed that E2F2 inhibited cell adhesion and consequently activated cell epithelial-to-mesenchymal transition (EMT) and migration. Further experiments revealed that E2F2 interacted with the PECAM1 promoter to suppress its transcriptional activity. The E2F2-knockdown-mediated promotion of cell adhesion was significantly reversed by the repression of PECAM1 expression. Finally, we observed that silencing E2F2 significantly inhibited viability and tumour progression in MM cell models and xenograft mouse models respectively. This study demonstrates that E2F2 plays a vital role as a tumour accelerator by inhibiting PECAM1-dependent cell adhesion and accelerating MM cell proliferation. Therefore, E2F2 may serve as a potential independent prognostic marker and therapeutic target for MM.


Subject(s)
Multiple Myeloma , Humans , Animals , Mice , Multiple Myeloma/genetics , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Gene Expression Regulation , Cell Proliferation , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism
6.
Int J Mol Sci ; 24(7)2023 Mar 28.
Article in English | MEDLINE | ID: mdl-37047293

ABSTRACT

It is known that E2F2 (E2F transcription factor 2) plays an important role as controller in the cell cycle. This study aimed to analyse the expression of the E2F2 gene and E2F2 protein and demonstrate E2F2 target microRNAs (miRNAs) candidates (miR-125b-5p, miR-155-3p, and miR-214-5p) in oral squamous cell carcinoma tumour and margin samples. The study group consisted 50 patients. The E2F2 gene and miRNAs expression levels were assessed by qPCR, while the E2F2 protein was assessed by ELISA. When analysing the effect of miRNAs expression on E2F2 gene expression and E2F2 protein level, we observed no statistically significant correlations. miR-125b-5p was downregulated, while miR-155-3p, and miR-214-5p were upregulated in tumour samples compared to margin. We observed a difference between the miR-125b-5p expression level in smokers and non-smokers in margin samples. Furthermore, HPV-positive individuals had a significantly higher miR-125b-5p and miR-214-5p expression level compared to HPV-negative patients in tumour samples. The study result showed that the E2F2 gene is not the target for analysed miRNAs in OSCC. Moreover, miR-155-3p and miR-125b-5p could play roles in the pathogenesis of OSCC. A differential expression of the analysed miRNAs was observed in response to tobacco smoke and HPV status.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , MicroRNAs , Mouth Neoplasms , Papillomavirus Infections , Humans , Carcinoma, Squamous Cell/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Papillomavirus Infections/genetics , Mouth Neoplasms/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Head and Neck Neoplasms/genetics , Gene Expression Regulation, Neoplastic
7.
Cells ; 12(3)2023 02 02.
Article in English | MEDLINE | ID: mdl-36766839

ABSTRACT

Human embryonic stem cells (hESCs) hold the potential to solve the problem of the shortage of functional hepatocytes in clinical applications and drug development. However, a large number of usable hepatocytes derived from hESCs cannot be effectively obtained due to the limited proliferation capacity. In this study, we found that enhancement of liver transcription factor C/EBPß during hepatic differentiation could not only significantly promote the expression of hepatic genes, such as albumin, alpha fetoprotein, and alpha-1 antitrypsin, but also dramatically reinforce proliferation-related phenotypes, including increasing the expression of proliferative genes, such as CDC25C, CDC45L, and PCNA, and the activation of cell cycle and DNA replication pathways. In addition, the analysis of CUT&Tag sequencing further revealed that C/EBPß is directly bound to the promoter region of proliferating genes to promote cell proliferation; this interaction between C/EBPß and DNA sequences of the promoters was verified by luciferase assay. On the contrary, the knockdown of C/EBPß could significantly inhibit the expression of the aforementioned proliferative genes. RNA transcriptome analysis and GSEA enrichment indicated that the E2F family was enriched, and the expression of E2F2 was changed with the overexpression or knockdown of C/EBPß. Moreover, the results of CUT&Tag sequencing showed that C/EBPß also directly bound the promoter of E2F2, regulating E2F2 expression. Interestingly, Co-IP analysis exhibited a direct binding between C/EBPß and E2F2 proteins, and this interaction between these two proteins was also verified in the LO2 cell line, a hepatic progenitor cell line. Thus, our results demonstrated that C/EBPß first initiated E2F2 expression and then coupled with E2F2 to regulate the expression of proliferative genes in hepatocytes during the differentiation of hESCs. Therefore, our findings open a new avenue to provide an in vitro efficient approach to generate proliferative hepatocytes to potentially meet the demands for use in cell-based therapeutics as well as for pharmaceutical and toxicological studies.


Subject(s)
Human Embryonic Stem Cells , Humans , Human Embryonic Stem Cells/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Hepatocytes/metabolism , Cell Proliferation/genetics , Promoter Regions, Genetic/genetics , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism
8.
Genes (Basel) ; 13(9)2022 08 24.
Article in English | MEDLINE | ID: mdl-36140689

ABSTRACT

Endothelial cell senescence has a vital implication for vascular dysfunction, leading to age-related cardiovascular disease, especially hypertension and atherosclerosis. E2F transcription factor 2 (E2F2) plays a critical role in cell proliferation, differentiation, and DNA damage response. Up to date, no study has ever connected E2F2 to vascular endothelial cell senescence. Here, we demonstrate that E2F2 is involved in endothelial cellular senescence. We found that E2F2 expression is decreased during the replicative senescence of human umbilical vein endothelial cells (HUVECs) and the aortas of aged mice. The knockdown of E2F2 in young HUVECs induces premature senescence characterized by an increase in senescence-associated ß-galactosidase (SA-ß-gal) activity, a reduction in phosphorylated endothelial nitric oxide synthase (p-eNOS) and sirtuin 1 (SIRT1), and the upregulation of senescence-associated secretory phenotype (SASP) IL-6 and IL-8. The lack of E2F2 promoted cell cycle arrest, DNA damage, and cell proliferation inhibition. Conversely, E2F2 overexpression reversed the senescence phenotype and enhanced the cellular function in the senescent cells. Furthermore, E2F2 deficiency downregulated downstream target genes including CNNA2, CDK1, and FOXM1, and overexpression restored the expression of these genes. Our findings demonstrate that E2F2 plays an indispensable role in endothelial cell senescence.


Subject(s)
Cellular Senescence , E2F2 Transcription Factor , Nitric Oxide Synthase Type III , Sirtuin 1 , Animals , Cells, Cultured , Cellular Senescence/genetics , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-6 , Interleukin-8 , Mice , Nitric Oxide Synthase Type III/metabolism , Sirtuin 1/genetics , beta-Galactosidase
9.
Med Oncol ; 39(9): 124, 2022 Jun 18.
Article in English | MEDLINE | ID: mdl-35716217

ABSTRACT

Both pro-oncogenic and anti-oncogenic effects of E2F2 have been revealed in different malignancies. However, the precise role of E2F2 in pancreatic cancer, in particular in relation to therapeutic intervention with gemcitabine, remains unclear. In this study, the effect of E2F2 on the proliferation and cell cycle modulation of pancreatic cancer cells, and whether E2F2 plays a role in the treatment of pancreatic cancer cells by gemcitabine, were investigated. The expression of E2F2 in pancreatic cancer was assessed by various methods including bioinformatics prediction, Western blotting, and real-time PCR. The effect of E2F2 on the proliferation and cell cycling of pancreatic cancer cells was analyzed by tissue culture and flow cytometry. In addition, the effect of E2F2 on the intervention of pancreatic cancer by gemcitabine was investigated using both in vitro and in vivo approaches. The expression of E2F2 was found to be significantly increased in pancreatic cancer tissues and cell lines. The pathogenic capacity of E2F2 lied in the fact that this transcription factor promoted the transformation of pancreatic cancer cell cycle from G1-phase to S-phase, thus enhancing the proliferation of pancreatic cancer cells. Furthermore, the expression of E2F2 was increased in pancreatic cancer cells in the presence of gemcitabine, and the augmented expression of E2F2 upregulated the gemcitabine resistance-related gene RRM2 and its downstream signaling molecule deoxycytidine kinase (DCK). The resistance of pancreatic cancer cells to gemcitabine was confirmed using both in vitro and in vivo models. In this study, E2F2 has been demonstrated for the first time to play a pro-oncogenic role in pancreatic cancer by promoting the transition of the cell cycle from G1-phase to S-phase and, therefore, enhancing the proliferation of pancreatic cancer cells. E2F2 has also been demonstrated to enhance the chemotherapy resistance of pancreatic cancer cells to gemcitabine by upregulating the expression of RRM2 and DCK that is downstream of RRM2.


Subject(s)
Deoxycytidine , Drug Resistance, Neoplasm , E2F2 Transcription Factor , Pancreatic Neoplasms , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Ribonucleoside Diphosphate Reductase/biosynthesis , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Up-Regulation/drug effects , Gemcitabine
10.
Cell Death Dis ; 13(1): 77, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075115

ABSTRACT

Long noncoding RNAs (lncRNAs) participate in biological processes in multiple types of tumors. However, the regulatory patterns of lncRNAs in prostate cancer remain largely unclear. Here, we evaluated the expression and roles of the lncRNA DLEU2 in prostate cancer. Our results showed that DLEU2 was upregulated in advanced prostate cancer tissues. Patients with prostate cancer displaying high expression of DLEU2 had a poor prognosis. Moreover, we demonstrated that overexpression of DLEU2 facilitated the proliferation, migration, and invasion of prostate cancer in vitro. Mechanistically, DLEU2 promoted serum and glucocorticoid-induced protein kinase 1 (SGK1) expression by acting as an miR-582-5p sponge, and the transcription of DLEU2 was activated by the dysregulation of E2F transcription factor 2 (E2F2) expression in prostate cancer. Furthermore, knockdown of DLEU2 attenuated prostate cancer tumorigenesis in vivo. Notably, these findings suggested that E2F2-activated DLEU2 may function as a competing endogenous RNA to facilitate prostate cancer progression by targeting the miR-582-5p/SGK1 axis.


Subject(s)
E2F2 Transcription Factor , Prostatic Neoplasms , RNA, Long Noncoding , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , E2F2 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Humans , Immediate-Early Proteins/metabolism , Male , MicroRNAs/genetics , Prostate/metabolism , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Long Noncoding/genetics
11.
Anticancer Drugs ; 33(1): e218-e227, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34387590

ABSTRACT

The aim of this study was to investigate the effect of circCUL2 on the proliferation, invasion and migration of retinoblastoma cells by regulating the miR-214-5p/E2F2 axis. qRT-PCR and western blot were performed to detect the expressions of circCUL2, miR-214-5p and E2F2 in tumor tissues and adjacent normal tissues from retinoblastoma patients, and in normal human retinal epithelial cells ARPE-19 and human retinoblastoma cells Y79 and SO-Rb50. qRT-PCR and western blot were performed for the detection of RNA levels of circCUL2 and miR-214-5p and the mRNA and protein levels of E2F2, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay for cell proliferation ability, Transwell assay for cell invasion ability, and scratch assay for cell migration ability. Luciferase dual reporter assay was used to detect the targeting relationship between circCUL2 and miR-214-5p, and between miR-214-5p and E2F2. CircCUL2 and E2F2 were lowly expressed, while miR-214-5p was highly expressed in retinoblastoma tumor tissues and cells. Transfection with pcDNA3.1-CUL2 or miR-214-5p inhibitor inhibited the proliferation, invasion and migration of Y79 and SO-Rb50 cells compared with the negative control; while transfection with sh-CUL2 or miR-214-5p mimics promoted the proliferation, invasion and migration of Y79 and SO-Rb50 cells. CircCUL2 negatively regulated miR-214-5p, while miR-214-5p negatively regulated E2F2. Overexpression of miR-214-5p or silencing of E2F2 in SO-Rb50 cells partially reversed the inhibitory effect of circCUL2 on the proliferation, invasion and migration of retinoblastoma cells. CircCUL2 inhibited the proliferation, invasion and migration of retinoblastoma cells by regulating the miR-214-5p/E2F2 axis.


Subject(s)
Cullin Proteins/genetics , E2F2 Transcription Factor/genetics , MicroRNAs/genetics , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic , Humans , RNA, Circular/genetics , RNA, Messenger/genetics , Retinoblastoma/pathology
12.
J Clin Lab Anal ; 35(12): e24031, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34716619

ABSTRACT

BACKGROUND: In this study, we aimed to screen methylation signatures associated with the prognosis of patients with clear cell renal cell carcinoma (ccRCC). METHODS: Gene expression and methylation profiles of ccRCC patients were downloaded from publicly available databases, and differentially expressed genes (DEGs)-differentially methylated genes (DMGs) were obtained. Subsequently, gene set enrichment and transcription factor (TF) regulatory network analyses were performed. In addition, a prognostic model was constructed and the relationship between disease progression and immunity was analyzed. RESULTS: A total of 23 common DEGs-DMGs were analyzed, among which 14 DEGs-DMGs were obtained with a cutoff value of PCC < 0 and p < 0.05. The enrichment analysis showed that the 14 DEGs-DMGs were enriched in three GO terms and three KEGG pathways. In addition, a total of six TFs were shown to be associated with the 14 DEGs-DMGs, including RP58, SOX9, NF-κB65, ATF6, OCT, and IK2. A prognostic model using five optimized DEGs-DMGs which efficiently predicted survival was constructed and validated using the GSE105288 dataset. Additionally, four types of immune cells (NK cells, macrophages, neutrophils, and cancer-associated fibroblasts), as well as ESTIMATE, immune, and stromal scores were found to be significantly correlated with ccRCC progression (normal, primary, and metastasis) in addition to the five optimized DEGs-DMGs. CONCLUSION: A five-gene methylation signature with the predictive ability for ccRCC prognosis was investigated in this study, consisting of CCNB2, CDKN1C, CTSH, E2F2, and ERMP1. In addition, potential targets for methylation-mediated immunotherapy were highlighted.


Subject(s)
Biomarkers, Tumor , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , DNA Methylation , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cathepsin H/genetics , Cyclin B2/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , E2F2 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Peptide Hydrolases/genetics , Prognosis , Transcription Factors/genetics
13.
Aging (Albany NY) ; 13(16): 20552-20568, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34433131

ABSTRACT

BACKGROUND: Nova Circular RNA (circRNA) of non-coding RNA has gradually become an important regulatory factor, and it has made people attach great concern over the occurrence and development of many diseases, particularly carcinomas. circ_RPPH1 is a newly discovered circRNA. Gene Expression Omnibus (GEO) analysis showed that there are high contents of circ_RPPH1 in breast cancer (BC), but the mechanism of circRNA in BC remains unclear. METHODS: Real-time quantitative PCR (qRT-PCR) was applied to test the role of circ_RPPH1 in BC patients, and functional experiments were applied to test the role of circ_RPPH1 on BC tumor. Fluorescence in situ hybridization, double luciferase reporter gene analysis, RNA pull-down and RNA immunoprecipitation experiments were performed to explore the correlation of circ_RPPH1 with miR-146b-3p/E2F2 in BC. RESULTS: circ_RPPH1 was evidently enhanced in BC, and its content was related to the clinical stage and pathological grade. circ_RPPH1 can accelerate the proliferation, migration and invasion, and promote tumorigenesis and metastasis. Mechanism exploration indicated that circ_RPPH1 acted as ceRNA (competing endogenous RNA) of miR-146b-3p, so as to reduce the inhibitory role of miR-146b-3p on its target E2F2. CONCLUSION: Circ_RPPH1/miR-146b-3p/E2F2 axis can promote the progression of BC, and it might be a latent therapeutic target for clinical BC.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Carcinoma/genetics , E2F2 Transcription Factor/genetics , MicroRNAs/genetics , RNA, Circular/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma/physiopathology , Cell Line, Tumor , Cell Proliferation , E2F2 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , MicroRNAs/metabolism , Neoplasm Metastasis , RNA, Circular/metabolism
14.
Cell Death Dis ; 12(7): 689, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244473

ABSTRACT

Renal cell carcinoma is the second malignant tumors in the urinary system with high mortality and morbidity. Increasing evidence suggests that long non-coding RNAs (lncRNAs) play critical roles in tumor development and progression. In the current study, based on the publicly available data obtained from GEO and TCGA database, we identified five prognosis-related lncRNAs with the ability to predict the prognosis of patients with renal cell carcinoma. Among them, the uncharacterized and upregulated lncRNA RCAT1 (renal cancer-associated transcript 1) was identified as the key lncRNA. Our data further revealed that the expression of lncRNA RCAT1 was significantly upregulated in renal cell carcinoma tissues and cells. Gain-of-function and loss-of-function studies showed that lncRNA RCAT1 promoted cell proliferation, migration, and invasion in vitro and in vivo. Furthermore, we verified that lncRNA RCAT1 could abundantly sponge miR-214-5p, which served as a tumor suppressor in renal cell carcinoma. Significantly, miR-214-5p overexpression could attenuate the promotion of cell proliferation and metastasis induced by lncRNA RCAT1. Moreover, we found that E2F2 was a direct target of miR-214-5p, and lncRNA RCAT1 could protect E2F2 from miR-214-5p-mediated degradation. Taken together, our findings suggested that lncRNA RCAT1 could enhance the malignant phenotype of renal cell carcinoma cells by modulating miR-214-5p/E2F2 axis, and lncRNA RCAT1 might be a novel prognostic biomarker and a potential therapeutic target for renal cell carcinoma.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Movement , E2F2 Transcription Factor/metabolism , Kidney Neoplasms/metabolism , Lung Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/secondary , Cell Line, Tumor , Cell Proliferation , Disease Progression , E2F2 Transcription Factor/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , RNA, Long Noncoding/genetics , Signal Transduction , Tumor Burden
15.
Aging (Albany NY) ; 13(10): 13626-13643, 2021 04 21.
Article in English | MEDLINE | ID: mdl-34091441

ABSTRACT

BACKGROUND: E2F2 is a member of the E2F transcription factor family and has important but not fully understood biological functions in cancers. The biological role of E2F2 in gastric cancer (GC) also remains unclear. METHODS: We examined the expression levels of E2F2 in GC using publicly available datasets such as TIMER, Oncomine, GEPIA, UALCAN, etc., and in our patient cohort, using quantitative real-time PCR, western blotting, and immunohistochemistry. We further investigated the effects of E2F2 on phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling, autophagy, and the migration and invasion of GC cells by the wound healing assay, Transwell assay and transmission electron microscopy. RESULTS: E2F2 was highly expressed in both GC tissues and cells compared with normal gastric tissues/cells. High E2F2 expression was associated with poor overall survival (OS). In addition, the expression of E2F2 in GC was strongly correlated with a variety of immune markers. E2F2 overexpression promoted the migration and invasiveness of GC cells in vitro through inhibition of PI3K/Akt/mTOR-mediated autophagy. CONCLUSION: High E2F2 expression was associated with the characteristics of invasive tumors and poor prognosis. E2F2 also had potential modulatory effects on tumor immunity. We discovered a novel function of E2F2 in the regulation of PI3K/Akt/mTOR-mediated autophagy and the downstream processes of cell migration and invasion.


Subject(s)
Autophagy , E2F2 Transcription Factor/antagonists & inhibitors , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Aged , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , DNA Methylation/genetics , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Male , Multivariate Analysis , Neoplasm Invasiveness , Prognosis , Programmed Cell Death 1 Receptor/metabolism , Protein Interaction Maps/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/genetics
16.
Biomed Res Int ; 2021: 8098130, 2021.
Article in English | MEDLINE | ID: mdl-33860054

ABSTRACT

Oral squamous cell carcinoma (OSCC) and oropharyngeal squamous cell carcinoma (OPSCC) are subgroups of head and neck squamous cell carcinoma. E2F Transcription Factor 2 (E2F2) could contribute to cancer development, because it plays a critical role in many cellular processes, including the cell cycle, proliferation, differentiation, DNA damage response, and cell death. In the current study, we assessed the associations of five E2F2 polymorphisms (rs6667575, rs3218121, rs3218211, rs3218148, and rs3218203) with OSCC and OPSCC and influence on the TNM staging and grading. This is the first such survey to concern the European population. The study included 94 primary tumour samples following surgical resection from patients, whereas the control group consisted of 99 healthy individuals. We tried a matching of cases and controls for age and sample size. DNA samples were genotyped by employing the 5' nuclease assay for allelic discrimination. Our results suggested that the most significant difference between the control group and the cancer group was the A/G heterozygote for rs3218121. Samples containing this genotype were mostly found in the control group. In our samples, rs6667575, rs3218121, rs3218211, and rs3218148 polymorphisms may affect the course of OSCC and OPSCC, while rs3218203 was not associated with OSCC and OPSCC. However, further studies are warranted to confirm our findings.


Subject(s)
E2F2 Transcription Factor/genetics , Genetic Predisposition to Disease , Oropharyngeal Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Adolescent , Adult , Aged , Case-Control Studies , Cohort Studies , Female , Genetic Association Studies , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Risk Factors , Young Adult
17.
Cell Signal ; 84: 110014, 2021 08.
Article in English | MEDLINE | ID: mdl-33894314

ABSTRACT

Ovarian cancer (OC) is a gynecological malignancy with a poor prognosis and low survival rate. E2F2 is a transcription activator that plays an indispensable role in cell proliferation and cell cycle progression. The preliminary analysis indicated that the E2F2 gene could produce three circular RNAs (circRNAs). This study aimed to investigate whether these circRNAs would be involved in OC tumorigenesis. The results showed that one of the circRNAs (termed circE2F2) was significantly upregulated in OC tissues and cell lines, and high circE2F2 expression was associated with poor survival in OC patients. The knockdown of circE2F2 in OC cells suppressed cell proliferation, migration, invasion, and cellular glucose metabolism. In circE2F2-deficient cells, the half-life of the E2F2 mRNA was significantly shorter than that in the control group, indicating that sufficient circE2F2 expression could strengthen the stability of the E2F2 mRNA. Further analysis revealed that circE2F2 could bind to RNA-binding protein Hu antigen R (HuR). Moreover, circE2F2 enhanced the stability of the E2F2 mRNA via binding to the HuR protein. Also, E2F2 overexpression significantly enhanced the mobility, invasiveness, and glucose metabolism of OC cells with insufficient circE2F2 expression, suggesting that circE2F2 induced OC cell growth and metastasis by upregulating E2F2. In conclusion, circE2F2 promoted OC cell proliferation, metastasis, and glucose metabolism by stabilizing the E2F2 mRNA via binding to the HuR protein. These findings suggest a novel regulatory mechanism for the oncogenic effects of circE2F2, E2F2, and HuR on ovarian carcinogenesis.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Cell Line, Tumor , Cell Proliferation/genetics , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Ovarian Neoplasms/pathology , RNA, Circular/genetics
18.
Life Sci ; 276: 119412, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33774025

ABSTRACT

AIMS: The effects of PFKFB4 on glycolysis during the cancer progression has been investigated, while its role in glioma remains unclear. The present study evaluated the molecular mechanism of PFKFB4 in glycolysis of glioma progression. MATERIALS AND METHODS: The pan-cancer platform SangerBox was inquired to investigate the E2F2 expression in tumors. The E2F2 expression was studied by qRT-PCR and immunohistochemistry in collected glioma and normal brain tissues and by qRT-PCR and western blot in glioma cells. The relationship between the E2F2 expression in glioma tissues and patients' prognosis was analyzed. The cell malignant phenotype, glycolysis, growth and metastasis were examined by CCK-8, EdU, colony formation, flow cytometry, wound healing, Transwell assays, ELISA kits, and tumorigenesis and metastasis assays. Downstream targets of E2F2 were searched in hTFtarget, followed by pathway enrichment analysis. The expression of these targets and their correlation with E2F2 expression in gliomas were investigated through the GEPIA website. After ChIP and luciferase assays, the effect of the target on glioma was investigated. KEY FINDINGS: E2F2 was overexpressed in glioma patients and predicted poor prognoses. E2F2 promoted cell proliferation, colony formation, DNA synthesis, migration, invasion and glycolysis, and inhibited apoptosis. Meanwhile, inhibition of E2F2 suppressed the growth and metastasis of gliomas. E2F2 elevated the PFKFB4 expression transcriptionally by binding to its promoter and activated PI3K/AKT pathway. The promotion of glioma metastasis and glycolysis by E2F2 was mitigated by PFKFB4 knockdown. SIGNIFICANCE: E2F2-mediated transcriptional enhancement of PFKFB4 expression regulated the phosphorylation of PI3K/AKT to promote glioma malignancy progression.


Subject(s)
Biomarkers, Tumor/metabolism , E2F2 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Glioma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphofructokinase-2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , E2F2 Transcription Factor/genetics , Female , Glioma/genetics , Glioma/metabolism , Glycolysis , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Phosphofructokinase-2/genetics , Phosphorylation , Prognosis , Proto-Oncogene Proteins c-akt/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Cancer Res ; 81(11): 2874-2887, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33771899

ABSTRACT

Lipid metabolism rearrangements in nonalcoholic fatty liver disease (NAFLD) contribute to disease progression. NAFLD has emerged as a major risk for hepatocellular carcinoma (HCC), where metabolic reprogramming is a hallmark. Identification of metabolic drivers might reveal therapeutic targets to improve HCC treatment. Here, we investigated the contribution of transcription factors E2F1 and E2F2 to NAFLD-related HCC and their involvement in metabolic rewiring during disease progression. In mice receiving a high-fat diet (HFD) and diethylnitrosamine (DEN) administration, E2f1 and E2f2 expressions were increased in NAFLD-related HCC. In human NAFLD, E2F1 and E2F2 levels were also increased and positively correlated. E2f1 -/- and E2f2 -/- mice were resistant to DEN-HFD-induced hepatocarcinogenesis and associated lipid accumulation. Administration of DEN-HFD in E2f1 -/- and E2f2 -/- mice enhanced fatty acid oxidation (FAO) and increased expression of Cpt2, an enzyme essential for FAO, whose downregulation is linked to NAFLD-related hepatocarcinogenesis. These results were recapitulated following E2f2 knockdown in liver, and overexpression of E2f2 elicited opposing effects. E2F2 binding to the Cpt2 promoter was enhanced in DEN-HFD-administered mouse livers compared with controls, implying a direct role for E2F2 in transcriptional repression. In human HCC, E2F1 and E2F2 expressions inversely correlated with CPT2 expression. Collectively, these results indicate that activation of the E2F1-E2F2-CPT2 axis provides a lipid-rich environment required for hepatocarcinogenesis. SIGNIFICANCE: These findings identify E2F1 and E2F2 transcription factors as metabolic drivers of hepatocellular carcinoma, where deletion of just one is sufficient to prevent disease. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/11/2874/F1.large.jpg.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/metabolism , Lipids/analysis , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/complications , Animals , Carcinogens , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Diet, High-Fat/adverse effects , E2F1 Transcription Factor/genetics , E2F2 Transcription Factor/genetics , Gene Expression Regulation , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Promoter Regions, Genetic
20.
J Cell Mol Med ; 25(2): 1178-1189, 2021 01.
Article in English | MEDLINE | ID: mdl-33342041

ABSTRACT

LBX2-AS1 is a long non-coding RNA that facilitates the development of gastrointestinal cancers and lung cancer, but its participation in ovarian cancer development remained uninvestigated. Clinical data retrieved from TCGA ovarian cancer database and the clinography of 60 ovarian cancer patients who received anti-cancer treatment in our facility were analysed. The overall cell growth, colony formation, migration, invasion, apoptosis and tumour formation on nude mice of ovarian cancer cells were evaluated before and after lentiviral-based LBX2-AS1 knockdown. ENCORI platform was used to explore LBX2-AS1-interacting microRNAs and target genes of the candidate microRNAs. Luciferase reporter gene assay and RNA pulldown assay were used to verify the putative miRNA-RNA interactions. Ovarian cancer tissue specimens showed significant higher LBX2-AS1 expression levels that non-cancerous counterparts. High expression level of LBX2-AS1 was significantly associated with reduced overall survival of patients. LBX2-AS1 knockdown significantly down-regulated the cell growth, colony formation, migration, invasion and tumour formation capacity of ovarian cancer cells and increased their apoptosis in vitro. LBX2-AS1 interacts with and thus inhibits the function of miR-455-5p and miR-491-5p, both of which restrained the expression of E2F2 gene in ovarian cancer cells via mRNA targeting. Transfection of miRNA inhibitors of these two miRNAs or forced expression of E2F2 counteracted the effect of LBX2-AS1 knockdown on ovarian cancer cells. LBX2-AS1 was a novel cancer-promoting lncRNA in ovarian cancer. This lncRNA increased the cell growth, survival, migration, invasion and tumour formation of ovarian cancer cells by inhibiting miR-455-5p and miR-491-5p, thus liberating the expression of E2F2 cancer-promoting gene.


Subject(s)
Disease Progression , E2F2 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Long Noncoding/metabolism , 3' Untranslated Regions/genetics , Base Sequence , Cell Line, Tumor , E2F2 Transcription Factor/metabolism , Female , Humans , MicroRNAs/genetics , Models, Biological , RNA, Long Noncoding/genetics , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...